You are on page 1of 702

Cytochrome P450

Structure, Mechanism, and Biochemistry


Cytochrome P450
Structure, Mechanism, and
Biochemistry
Third edition

Edited by

Paul R. Ortiz de Montellano


Department of Pharmaceutical Chemistry
University of California, San Francisco, CA

KluwerAcadennic/Plenum Publishers
New York, Boston, Dordrecht, London, Moscow
Library of Congress Cataloging-in-Publication Data

Cytochrome P450 : structure, mechanism, and biochemistry / edited by Paul R. Ortiz de


Montellano.— 3rd ed.
p. cm.
Includes bibliographical references and index.
ISBN 0-306-48324-6
1. Cytochrome P-450. 2. Metalloenzymes. I. Ortiz de Montellano, Paul R.
QP671.C83C98 2004
572'.7—dc22
2004043512

ISBN 0-306-48324-6
©2005 Kluwer Academic/Plenum Publishers, New York
233 Spring Street, New York, N. Y. 10013
http://www.wkap.nl/
10 9 8 7 6 5 4 3 2 1
A C.I.P. record for this book is available from the Library of Congress
All rights reserved
No part of this book may be reproduced, stored in a retrieval system, or transmitted in any form or by any
means, electronic, mechanical, photocopying, microfilming, recording, or otherwise,without written
permission from the Publisher, with the exception of any material supplied specifically for the purpose of
being entered and executed on a computer system, for exclusive use by the purchaser of the work.
Permissions for books published in Europe: permissions@wkap.nl
Permissions for books published in the United States of America: permissions@wkap.com
Printed in the United States of America
Preface

In Dantean terms, three is a magic number, and this is the third edition of this book. Two decades ago the
first edition appeared at a time when the first crystal structure of a P450 enzyme had just been determined,
the multiphcity of P450 isoforms was just beginning to be reahzed, and determination of the human
genome was not even a dream. The first edition surveyed a field that was young and awkward but full of
promise. Ten years later, when the second edition appeared, enormous progress had been made in all
aspects of P450 chemistry and biology. The structures of several bacterial P450 enzymes were then avail-
able, a systematic nomenclature system had brought order to the chaos engendered by the rapidly grow-
ing number of isoforms, and the mechanisms involved in regulation of P450 activity were beginning to
yield their secrets. The subsequent 10 years have brought the field to the maturity reflected in this third
edition of the book. The dream of obtaining crystal structures of the mammalian P450 enzymes has
become a reality, the human genome has defined the number of P450 enzymes in homo sapiens, and P450
enzymes have taken center stage in the pharmaceutical industry because of their critical roles in the suc-
cess or failure of new therapeutic agents. The field continues to be exciting and vigorous, but it is now
the excitement of maturity and fulfillment. The increasing sophistication of both the questions that can
be asked and the experimental tools available for their investigation has led to a deeper, more satisfying,
understanding of the P450 system and, in some cases, to a reexamination of earlier conclusions.
In order to accommodate the new areas of P450 biology that have come into their own in the past
decade it has been necessary to eliminate the chapters on the peroxidases, nitric oxide synthases, and
related proteins that in earlier editions placed the P450 system in its hemoprotein context. The first sec-
tion of the book, which collects the work on the structure and mechanism of the P450 enzymes, includes
chapters on the model systems used to elucidate P450 chemistry (Chapter 1) and on recent develop-
ments in computational chemistry that rationalize the sometimes conflicting mechanistic observations
(Chapter 2). These chapters are followed by an up-to-date discussion of bacterial and mammalian crys-
tal structures (Chapter 3), a current review of the electron transfer partners (Chapter 4), a detailed dis-
cussion of the mechanism for activation of molecular oxygen (Chapter 5), and a general review of
substrate oxidation mechanisms (Chapter 6). This broad introduction to P450 structure and mechanism
closes with an updated review of the inhibition of P450 enzymes (Chapter 7). The second section of the
book, which focuses on the biology of manmialian P450 enzymes, includes a discussion of our current
understanding of the induction of P450 enzymes (Chapter 8), a review of the hormonal influences on
P450 expression and activity (Chapter 9), a very extensive summary of the biology of all the human
cytochrome P450 enzymes (Chapter 10), and a chapter on the oxidation of arachidonic acid and
eicosanoids (Chapter 11). The final section of the book is comprised of a chapter on non-mammalian—
primarily plant—P450 enzymes (Chapter 12) and a review of bacterial P450 enzymes and their biotech-
nological potential (Chapter 13). The book, as before, closes with an appendix containing practical
experimental information for individuals doing research in the P450 field.
It is my hope that this third edition will turn out to be as useful as the prior editions. Perhaps, with
luck, it may even prove to be sufficiently special to justify the folkloric expectations of a third effort.
I gratefully dedicate this third edition to my brother Bernard, who brought me into chemistry; to my
wife Kirby, who for the past 30 years has gracefully sacrificed Saturday companionship to the demands
of the laboratory; to Almira Correia, who has made P450 a congenial experience at UCSF for almost as
long; and to my daughters, Lara and Maya, who decided to pass on P450 but continue to amaze me.
Paul R. Ortiz de Montellano San Francisco
Contents

Contributors

I . Models and Mechanisms of Cytochrome P450 Action


John T. Groves
1. Introduction 1
2. Oxygen Activation by Heme-Thiolate Proteins 1
3. Mechanism of Hydroxylation by Cytochrome P450 3
4. Mechanisms and Molecular Trajectories for Hydroxylation by Cytochrome P450 7
5. On the Mechanism of Nitric Oxide Synthase 16
6. Synthetic Oxometalloporphyrins as Models for Cytochrome P450 17
7. Manganese Porphyrins in Catalytic Oxidations 19
8. Metalloporphyrins as Detectors and Decomposition Catalysts of Peroxynitrite 23
9. Synthetic Metalloporphyrins as Stereoselective Catalysts 25
10. Ruthenium Porphyrins in Oxidative Catalysis 26
II. Conclusion 34
Acknowledgments 34
References 34

2. Computational Approaches to Cytochrome P450 Function


Sason Shaik and Samuel P. De Visser
1. Introduction 45
2. Methods 45
3. The Catalytic Cycle of P450 48
3.1. The Resting State (1) 51
3.2. The Pentacoordinate Ferric-Porphyrin (2) and Ferrous-Porphyrin (3) Complexes 52
3.3. The Gating of the Catalytic Cycle 54
3.4. The Ferrous-Dioxygen (4) and Ferric-Dioxygen (5) Complexes 54
3.5. The Protonation Mechanism of Ferric-Dioxygen (5) to Cpd 0 (6) 56
3.6. Cpd 0: The Ferric Peroxide Complex (6) 57
3.7. Protonation of Cpd 0 and Formation of Cpd I (7) 57
3.8. The "Push Effect" on the 0 - 0 Cleavage Process 58
3.9. Cpd I (7) 59
3.10. What Makes the Catalytic Cycle Tick? A Summary 63
4. MM and MM/MD Studies of P450 Reactivity Aspects 63
4.1. Studies of Substrate Entrance, Binding, and Product Exit 63
4.2. MM and MM/MD Studies of Regioselectivity 65
5. QM Studies of P450 Reactivity Patterns 66
5.1. Reactivity of Cpd I: General Considerations of the Origins of
Two-State Reactivity (TSR) of Cpd I 66
5.2. A Primer to P450 Reactivity: Counting of Electrons 66
5.3. Alkane Hydroxylation 68
viii Contents

5.4. The Rebound Process: More Features than Meet the Eye 72
5.5. Alkene Epoxidation 73
5.6. Hydroxylation of Arenes 75
5.7. Sulfoxidation of Alkyl Sulfides 76
5.8. Can Ferric Peroxide (6) be a Second Oxidant? 77
5.9. Competitive Hydroxylation and Epoxidation in Propene 77
5.10. An Overview of Reactivity Features of Cpd I 79
6. Prospective 80
Acknowledgment 80
References 80

3. Structures of Cytochrome P450 Enyzmes


Thomas L Poulos and Eric F. Johnson
1. Introduction 87
2. Overall Architecture 87
3. P450s from Thermophiles 91
4. Membrane P450s 92
5. Electron Transfer Complexes 95
6. Substrate Complexes 99
7. Conformational Adaptations to Substrates and Inhibitors 100
8. Conformational Dynamics for Substrate Access 102
Acknowledgments Ill
References HI

4. Electron Transfer Partners of Cytochrome P450


Mark J.I. Paine, Nigei S. Scrutton, Andrew W. Munro, Aldo Gutierrez,
Gordon O.K. Roberts, and C. Roland Wolf
1. Introduction 115
2. NADPH-Cytochrome P450 Reductase and the Diflavin Reductase Family 116
2.1. Background 116
2.2. The Diflavin Reductase Family 117
2.3. CPR Genes 118
2.4. Probing the Physiological Role of CPR 119
2.5. Structure of CPR 120
2.5.1. The FMN-Binding Domain 120
2.5.2. FAD/NADPH-Binding Domain 122
2.6. The Electron Transfer Mechanism 124
2.6.1. Trp676 and FAD Reduction 126
2.6.2. Binding of Two Coenzyme Molecules 127
2.6.3. Internal Electron Transfer 127
2.6.4. Interaction with and Electron Transfer to P450 128
2.7. Cytochrome P450 BM3 131
2.7.1. Electron Transfer Properties of BM3 Reductase 132
2.8. Artificial CPR-P450 Fusion Constructs 133
3. Electron Transfer to P450s from Cytochrome b^ 133
4. Iron-Sulfur Electron Donors: Adrenodoxin, Putidaredoxin, and their Reductases 134
4.1. General 134
4.2. Interactions with P450 135
5. Novel Redox Systems 138
Contents ix

Acknowledgments 138
References 138

5, Activation of Molecular Oxygen by Cytochrome P450


Thomas M. Makris, Ilia Denisov, lime Schlichting, and Stephen G. Sligar
1. Introduction to Oxygen Activation 149
2. General Features of Dioxygen Activation in Heme Enzymes 151
2.1. The Oxidase/Oxygenase Pathway in Cytochrome P450 152
3. Enzymatic Cycle of Cytochrome P450 155
3.1. The Ferrous-Dioxygen Complex 156
3.2. Reduction of Oxy-Ferrous P450 and Formation of Peroxo-Ferric
Complexes: Properties, Stability, and Spectroscopy 157
3.3. The Second Branchpoint of P450 Catalysis: Uncoupling with Hydrogen
Peroxide Production or Dioxygen Bond Scission 160
4. Structural Input into the Mechanisms of P450-Catalyzed Dioxygen Activation 161
4.1. A "Conserved" Alcohol Side Chain in the Active Site of P450 162
4.2. The "Conserved" Acid Functionality 164
4.3. Crystallographic Studies of P450 Reaction Intermediates 165
4.4. Mechanism-Based Specificity of Proton Transfer 169
4.5. Summary 170
Acknowledgments 170
References 170

6. Substrate Oxidation by Cytochrome P450 Enzymes


Paul R. Ortiz de Montellano and James J. De Voss
1. Introduction 183
2. Activation of Molecular Oxygen 184
3. Hydrocarbon Hydroxylation 186
4. Heteroatom Oxidation and Dealkylation 193
5. Olefin and Acetylene Oxidation 198
6. Oxidation of Aromatic Rings 202
7. Dehydrogenation Reactions 208
8. Carbon-Carbon Bond Cleavage Reactions , 211
8.1. Cleavage between Oxygenated Carbons 211
8.2. Cleavage Alpha to Oxygenated Carbon 217
8.3. Cleavage Alpha to Carbon Bearing a Nitrogen Atom 228
9. Conclusions 229
Acknowledgments 230
References 230

7. Inhibition of Cytochrome P450 Enzymes


Maria Almira Correia and Paul R. Ortiz de Montellano
1. Introduction 247
2. Reversible Inhibitors 247
2.1. Coordination to Ferric Heme 248
2.2. Coordination to Ferrous Heme 248
2.3. Heme Coordination and Lipophilic Binding 248
X Contents

3. Catalysis-Dependent Inhibition 250


3.1. Covalent Binding to the Protein 250
3.1.1. Sulfur and Halogenated Compounds 250
3.1.2. Olefins and Acetylenes 255
3.1.3. Other P450 Protein Modifying Inactivators 259
3.2. Quasi-Irreversible Coordination to the Prosthetic Heme 263
3.2.1. Methylenedioxy Compounds 263
3.2.2. Amines 265
3.2.3. 1,1-Disubstituted and Acyl Hydrazines 266
3.3. Covalent Binding to the Prosthetic Heme 267
3.3.1. Terminal Olefins 267
3.3.2. Acetylenes 269
3.3.3. Dihydropyridines and Dihydroquinolines 272
3.3.4. Alkyl- and Arylhydrazines and Hydrazones 273
3.3.5. Other N-N Functions 275
3.3.6. Other Functionalities 278
3.4. Modification of the P450 Protein by Heme Fragments 280
3.5. Other Modes of P450 Heme Degradation and Protein Denaturation 282
4. P450 Enzyme Specificity 285
5. Inhibitors of Biosynthetic Enzymes 285
5.1. P450^^^ 286
5.2. Aromatase 286
5.3. Lanosterol 14-Demethylation 290
5.4. Other Biosynthetic Sterol Hydroxylases 292
5.5. Fatty Acid and Leukotriene Monooxygenases 292
6. Summary 294
Acknowledgment 295
References 295

8. Induction of Cytochrome P450 Enzymes


Susanne N. Williams, Elizabeth Dunham, and Christopher A. Bradfield
1. Introduction 323
1.1. Cytochrome P450 Enzymes and the Adaptive Response 323
1.2. Overview of Nuclear Receptors 323
2. The Pregnane X Receptor 324
2.1. Introduction 324
2.2. The PXR 325
2.3. PXR Ligands and Species Differences 325
2.4. Activation of Transcription 325
2.5. Mouse Models 326
2.6. Future Research 327
3. The Constitutive Androstane Receptor 328
3.1. Introduction 328
3.2. The Nuclear Receptor CAR 328
3.3. Mediators of CAR Activity 328
3.4. Activation of Transcription 330
3.5. Mouse Models 330
3.6. Future Directions 331
4. The Peroxisome Proliferator Activated Receptor a 331
Contents xi

4.1. Introduction 331


4.2. PPAR Isoforms 332
4.3. PPARa Ligands 332
4.4. Activation of Transcription 332
4.5. Species Differences 334
4.6. Mouse Models 334
4.7. Future Directions 334
5. The Aryl Hydrocarbon Receptor 335
5.1. Introduction 335
5.2. TheAHR 335
5.3. AHR Ligands 336
5.4. Activation of Transcription 337
5.5. Mouse Models 338
5.6. Future Directions 338
5.7. Conclusions 339
Acknowledgments 339
References 339

9. Hormonal Regulation of Liver Cytochrome P450 Enzymes


David J. Waxman and Thomas K.H. Chang
1. Introduction 347
2. Steroid Hormones as Substrates for Sex-Dependent Liver P450s 348
3. Developmental Regulation of Sex-Dependent Rat Liver P450s 348
4. Hormonal Control of Liver P450 Expression 350
4.1. Regulation by Gonadal Hormones 350
4.1.1. Testosterone 350
4.1.1.1. Distinct Effects of Neonatal Androgen and Adult Androgen 350
4.1.1.2. Testosterone Suppression of Female Enzymes 350
4.1.1.3. Mechanisms of Testosterone Regulation 351
4.1.2. Estrogen 351
4.2. Regulation by Growth Hormone 351
4.2.1. Sex-Dependent GH Secretory Profiles 351
4.2.2. Transcriptional Effects of GH on CYP Genes . 354
4.2.3. Cellular Mechanisms of GH Signaling 354
4.2.3.1. Significance of GH Pulse Frequency 355
4.2.3.2. Role of GH Receptor (GHR) 355
4.2.4. Role of STAT5b in Sex-Dependent CYP Expression 356
4.2.4.1. GH Signaling Pathways Involving STAT Transcription Factors . . . . . . 356
4.2.4.2. STAT5b Gene Knockout Mouse Model 359
4.2.4.3. Interaction of GH-Responsive CYP Promoters with
GH-Activated STAT5b 360
4.2.4.4. Interactions between STAT5b and Liver Transcription
Factors Regulating Sex-Specific CYPs 361
4.2.4.5. Downregulation of Hepatic STAT5b Signaling , 361
4.3. Regulation by Thyroid Hormone 362
4.3.1. Cytochromes P450 362
4.3.2. NADPH-Cytochrome P450 Reductase 362
5. Alteration of Liver P450 Expression by Hormonal Perturbation 362
5.1. Modulation by Drugs 362
xii Contents

5.2. Modulation by Polycyclic Aromatic Hydrocarbons 363


5.3. Modulation by Pathophysiological State 363
5.3.1. Diabetes 363
5.3.2. Liver Cirrhosis 364
5.4. Modulation by Ethanol and Dietary Factors 364
5.5. Impact on Drug Metabolism and Procarcinogen Activation 365
6. Conclusion 365
Acknowledgment 366
References 366

10. Human Cytochrome P450 Enzymes


F. Peter Guengerich
1. Background and History of Development of the Field 377
2. General Issues of VariabiUty and Polymorphism 383
3. Approaches to Defining Catalytic Specificity of Human P450s 388
3.1. Inhibitors 389
3.2. Correlations 389
3.3. Antibody Inhibition 390
3.4. Demonstration of Reaction with Recombinant P450 392
4. Relevance of P450s in In Vivo Drug Metabolism 392
5. Relevance of P450s in Toxicology and Cancer Risk 395
6. Individual Human P450 Enzymes 396
6.1. P450 lAl 396
6.1.1. Sites of Expression and Abundance 396
6.1.2. Regulation and Polymorphism 397
6.1.3. Substrates and Reactions 397
6.1.4. Knowledge about Active Site 397
6.1.5. Inhibitors 398
6.1.6. Clinical Issues 398
6.2. P450 1A2 398
6.2.1. Sites of Expression and Abundance 398
6.2.2. Regulation and Polymorphism 398
6.2.3. Substrates and Reactions 399
6.2.4. Knowledge about Active Site 399
6.2.5. Inhibitors 399
6.2.6. Clinical Issues 400
6.3. P450 IBl 400
6.3.1. Sites of Expression and Abundance 400
6.3.2. Regulation and Polymorphism 400
6.3.3. Substrates and Reactions 400
6.3.4. Knowledge of Active Site 402
6.3.5. Inhibitors 402
6.3.6. Clinical Issues 402
6.4. P450 2A6 402
6.4.1. Sites of Expression and Abundance 402
6.4.2. Regulation and Polymorphism 402
6.4.3. Substrates and Reactions 403
6.4.4. Knowledge about Active Site 403
6.4.5. Inhibitors 404
6.4.6. Clinical Issues 404
Contents xiii

6.5. P450 2A7 404


6.6. P450 2A13 404
6.6.1. Sites of Expression and Abundance 404
6.6.2. Regulation and Polymorphism 405
6.6.3. Substrates and Reactions 405
6.6.4. Knowledge about Active Site 405
6.6.5. Inhibitors 405
6.6.6. Clinical Issues 405
6.7. P450 2B6 405
6.7.1. Sites of Expression and Abundance 405
6.7.2. Regulation and Polymorphism 405
6.7.3. Substrates and Reactions 406
6.7.4. Knowledge about Active Site 406
6.7.5. Inhibitors 406
6.7.6. Clinical Issues 406
6.8. P450 2C8 407
6.8.1. Sites of Expression and Abundance 407
6.8.2. Regulation and Polymorphism 407
6.8.3. Substrates and Reactions 407
6.8.4. Knowledge about Active Site 407
6.8.5. Inhibitors 408
6.8.6. Clinical Issues 408
6.9. P450 2C9 408
6.9.1. Sites of Expression and Abundance 408
6.9.2. Regulation and Polymorphism 408
6.9.3. Substrates and Reactions 409
6.9.4. Knowledge about Active Site 409
6.9.5. Inhibitors 410
6.9.6. Clinical Issues 410
6.10. P450 2C18 411
6.10.1. Sites of Expression and Abundance 411
6.10.2. Regulation and Polymorphism 411
6.10.3. Substrates and Reactions 411
6.10.4. JCnowledge about Active Site 411
6.10.5. Inhibitors 411
6.10.6. Clinical Issues 412
6.11. P450 2C19 412
6.11.1. Sites of Expression and Abundance 412
6.11.2. Regulation and Polymorphism 412
6.11.3. Substrates and Reactions 412
6.11.4. Knowledge about Active Site 413
6.11.5. Inhibitors 413
6.11.6. Clinical Issues 413
6.12. P450 2D6 413
6.12.1. Sites of Expression and Abundance 413
6.12.2. Regulation and Polymorphism 413
6.12.3. Substrates and Reactions 414
6.12.4. Knowledge about Active Site 416
6.12.5. Inhibitors 417
6.12.6. Clinical Issues 418
6.13. P450 2E1 418
xiv Contents

6.13.1. Sites of Expression and Abundance 418


6.13.2. Regulation and Polymorphism 419
6.13.3. Substrates and Reactions 420
6.13.4. Knowledge about Active Site 420
6.13.5. Inhibitors 421
6.13.6. Clinical Issues 421
6.14. P450 2F1 422
6.15. P450 2J2 422
6.16. P450 2R1 423
6.17. P450 2S1 423
6.18. P450 2U1 423
6.19. P450 2W1 423
6.20. P450 3A4 423
6.20.1. Sites of Expression and Abundance 424
6.20.2. Regulation and Polymorphism 424
6.20.3. Substrates and Reactions 425
6.20.4. Knowledge about Active Site 426
6.20.5. Inhibitors 430
6.20.6. Clinical Issues 430
6.21. P450 3A5 431
6.21.1. Sites of Expression and Abundance 431
6.21.2. Regulation and Polymorphism 431
6.21.3. Substrates and Reactions 432
6.21.4. Knowledge about Active Site 432
6.21.5. Inhibitors 432
6.21.6. Clinical Issues 432
6.22. P450 3A7 432
6.22.1. Sites of Expression and Abundance 432
6.22.2. Regulation and Polymorphism 433
6.22.3. Substrates and Reactions 433
6.22.4. Knowledge about Active Site 433
6.22.5. Inhibitors 433
6.22.6. Clinical Issues 434
6.23. P450 3A43 434
6.24. P450 4A11 434
6.24.1. Sites of Expression and Abundance 434
6.24.2. Regulation and Polymorphism 434
6.24.3. Substrates and Reactions 434
6.24.4. Knowledge about Active Site 434
6.24.5. Inhibitors 435
6.24.6. Clinical Relevance 435
6.25. P450 4A22 435
6.26. P450 4B1 435
6.26.1. Sites of Expression and Abundance 435
6.26.2. Regulation and Polymorphism 435
6.26.3. Substrates and Reactions 435
6.26.4. Knowledge about Active Site 436
6.26.5. Inhibitors 436
6.26.6. Clinical Issues 436
6.27. P450 4F2 436
6.28. P450 4F3 436
Contents xv

6.29. P450 4F8 437


6.30. P450 4F11 437
6.31. P450 4F12 437
6.32. P450 4F22 437
6.33. P450 4V2 437
6.34. P450 4X1 437
6.35. P450 4Z1 437
6.36. P450 5A1 437
6.36.1. Sites of Expression and Abundance 437
6.36.2. Regulation and Polymorphism 438
6.36.3. Substrates and Reactions 438
6.36.4. Knowledge about Active Site 439
6.36.5. Inhibitors 439
6.36.6. Clinical Issues 439
6.37. P450 7A1 439
6.37.1. Sites of Expression 439
6.37.2. Regulation and Polymorphism 439
6.37.3. Substrates and Reactions 440
6.37.4. Knowledge about Active Site 441
6.37.5. Inhibitors 441
6.37.6. Clinical Issues 441
6.38. P450 7B1 441
6.39. P450 8A1 441
6.39.1. Sites of Expression and Abundance 442
6.39.2. Regulation and Polymorphism 442
6.39.3. Substrates and Reactions 442
6.39.4. Knowledge about Active Site 442
6.39.5. Inhibitors 442
6.39.6. Clinical Issues 443
6.40. P450 8B1 443
6.41. P450 U A l 443
6.41.1. Sites of Expression 443
6.41.2. Regulation and Polymorphism 445
6.41.3. Substrates and Reaction 445
6.41.4. Knowledge about Active Site 445
6.41.5. Inhibitors 445
6.41.6. Clinical Issues 446
6.42. P450 l l B l 446
6.42.1. Sites of Expression 446
6.42.2. Regulation and Polymorphism 446
6.42.3. Substrates and Reactions 446
6.42.4. Knowledge about Active Site 447
6.42.5. Inhibitors 447
6.42.6. Clinical Issues 447
6.43. P450 11B2 447
6.43.1. Sites of Expression 447
6.43.2. Regulation and Polymorphism 447
6.43.3. Substrates and Reactions 448
6.43.4. Knowledge about Active Site 448
6.43.5. Inhibitors 448
6.43.6. Clinical Issues 448
i Contents

6.44. P450 17A1 448


6.44.1. Sites of Expression 448
6.44.2. Regulation and Polymorphism 449
6.44.3. Substrates and Reactions 449
6.44.4. Knowledge about Active Site 450
6.44.5. Inhibitors 450
6.44.6. Clinical Issues 450
6.45. P450 19A1 450
6.45.1. Sites of Expression 451
6.45.2. Regulation and Polymorphism 451
6.45.3. Substrates and Reactions 452
6.45.4. Knowledge about Active Site 452
6.45.5. Inhibitors 452
6.45.6. Clinical Issues 452
6.46. P450 20A1 452
6.47. P450 21A2 453
6.47.1. Sites of Expression 453
6.47.2. Regulation and Polymorphism 453
6.47.3. Substrates and Reactions 453
6.47.4. Knowledge about Active Site 453
6.47.5. Inhibitors 453
6.47.6. Clinical Issues 453
6.48. P450 24A1 454
6.48.1. Sites of Expression and Abundance 454
6.48.2. Regulation and Polymorphism 454
6.48.3. Substrates and Reactions 455
6.48.4. Knowledge about Active Site 455
6.48.5. Inhibitors 455
6.48.6. Clinical Issues 455
6.49. P450 26A1 455
6.50. P450 26B1 456
6.51. P450 26C1 456
6.52. P450 27A1 456
6.52.1. Sites of Expression and Abundance 456
6.52.2. Regulation and Induction 456
6.52.3. Substrates and Reactions 458
6.52.4. Knowledge about Active Site 458
6.52.5. Inhibitors 458
6.52.6. Chnical Issues 458
6.53. P450 27B1 459
6.53.1. Sites of Expression and Abundance 459
6.53.2. Regulation and Polymorphism 459
6.53.3. Substrates and Reactions 460
6.53.4. Knowledge about Active Site 460
6.53.5. Inhibitors 460
6.53.6. Clinical Issues 460
6.54. P450 27C1 460
6.55. P450 39A1 460
6.56. P450 46A1 461
6.57. P450 51A1 461
6.57.1. Sites of Expression and Abundance 461
Contents xvii

6.57.2. Regulation and Polymorphism 461


6.57.3. Substrates and Reactions 462
6.57.4. Knowledge about Active Site 462
6.57.5. Inhibitors 462
6.57.6. Clinical Issues 462
7. Concluding Remarks 462
Acknowledgments 463
References 463

11. Cytochrome P450 and the Metabolism and Bioactivation of Arachidonic


Acid and Eicosanoids
Jorge H. Capdevila, Vijaykumar R. Holla, and John R. Faick
1. Introduction 531
2. Metabolism of Eicosanoids 532
2.1. NADPH-Independent Reactions 532
2.2. NADPH-Dependent Reactions 533
2.2.1. (o/w-l Oxidation of Prostanoids 533
2.2.2. a)/o)-l Oxidation of Leukotrienes and Other Eicosanoids 534
3. Metabolism of Arachidonic Acid: The Arachidonic Acid Monooxygenase 535
3.1. bis-Allylic Oxidation (Lipoxygenase-Like Reactions) 536
3.2. Hydroxylation at Cjg-C2o (^^^"1 Hydroxylase Reactions) 536
3.2.1. Introduction 536
3.2.2. Enzymology, Isoform Specificity 537
3.3. Olefin Epoxidation (Epoxygenase Reactions) 539
3.3.1. Introduction 539
3.3.2. Enzymology, Isoform Specificity 539
3.3.3. P450 Arachidonic Acid Epoxygenase: A Member of the Arachidonic
Acid Metabolic Cascade 541
3.4. Functional Roles of the P450 Arachidonic Acid Monooxygenase 542
3.4.1. Vascular Reactivity; Ion Channel Regulation 542
3.4.2. Blood Pressure Control and Hypertension 543
4. Conclusion 545
Acknowledgments 545
References 545

12. Cytochrome P450s in Plants


Kirsten Annette Nielsen and Birger Lindberg Moller
1. Introduction 553
1.1. Natural Products 553
1.2. Chemical Warfare 553
1.3. Chemical Communication 553
1.4. Medicinal Agents 554
2. The P450 Superfamily in Plants 554
2.1. Nomenclature 554
3. Tools Available to Identify Biological Functions 555
3.1. Phylogenetic Relationships 555
3.2. Mutant Collections in A. thaliana 556
xviii Contents

3.3. Reverse Genetics 556


3.4. Heterologous Expression in Microorganisms 556
3.5. Isolation of Enzymes 557
3.6. Homology-Based Cloning 557
4. Non-A-Type P450s Mediating Steroid Biosynthesis 557
4.1. CYP90S 558
4.2. CYP85S 560
5. A-Type P450s Mediating Plant Protection 560
5.1. Broad Defense: Cyanogenic Glucosides 560
5.1.1. Biosynthesis 561
5.1.2. Substrate Channeling and Metabolon Formation 563
5.1.3. Substrate Specificities 564
5.2. Functional Uniformity within the CYP79 Family 564
5.3. Functional Diversity among CYP71S 566
5.3.1. CYP71A and CYP71B Subfamilies 566
5.3.2. CYP71C Subfamily: Grass-Specific Defense Compounds 566
5.3.3. CYP71D, -F, and -R Subfamilies 568
5.4. SpeciaUzed Defense—Isoflavonoids in Legumes 569
6. P450 Mediated Production of Alkaloids with Medicinal Importance 571
7. Future Prospects: Crosstalk and Metabolic Engineering 573
References 575

13. The Diversity and Importance of Microbial Cytochromes P450


Steven L Kelly, Diane E. Kelly, Colin J. Jackson, Andrew G.S. Warrilow,
and David C. Lamb
1. Introduction to Microbial CYPs 585
2. Classes of Microbial CYPs 587
3. Considering the Origins and Relatedness of Microbial CYPs 589
3.1. CYP51 and Evolution of the Superfamily 590
3.2. Bacterial CYP51 592
4. Archetypal Bacterial CYPs 594
5. Biodiversity of Bacterial CYPs and the Actinomycetes 596
5.1. Mycobacterial CYPs 596
5.2. Biodiversity in Streptomycetes 598
5.3. CYP Biodiversity in Archaebacteria 601
6. Fungal CYPs 601
7. Azole Antifungals and the Evolution of New Resistant Genes 603
7.1. The Fungal CYP51 System 603
7.2. Azole Activity and Resistance in Fungi 605
8. Conclusions 610
Acknowledgments 610
References 610

Appendix: Human and Rat Liver Cytochromes P450: Functional


Markers, Diagnostic Inhibitor Probes, and Parameters Frequently
Used in P450 Studies 619
Maria Almira Correia

Index 659
Contributors

Christopher A. Bradfield, McArdle Laboratory for Cancer Research, University of Wisconsin,


Madison, WI

Jorge H. Capdevila, Departments of Medicine and Biochemistry, Vanderbilt University Medical


School, Nashville, TN

Thomas K.H. Chang, The University of British Columbia, Vancouver, BC, Canada

Maria Almira Correia, Department of Cellular and Molecular Pharmacology, Department of


Pharmaceutical Chemistry, and Department of Biopharmaceutical Sciences and the Liver Center,
University of California, San Francisco, CA

Paul R. Ortiz de Montellano, Department of Cellular and Molecular Pharmacology and Department
of Pharmaceutical Chemistry, University of California, San Francisco, CA

Samuel P. De Visser, Department of Organic Chemistry and the Lise-Meitner-Minerva Center for
Computational Quantum Chemistry, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel

Ilia Denisov, Max Planck Institut fiir Molekulare Physiologie, Abt. Biophysikalische Chemie,
Germany

James J. De Voss, Department of Chemistry, University of Queensland, Brisbane, QLD Australia

Elizabeth Dunham, McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI

John R. Falck, Department of Biochemistry, Southwestern Medical Center, Dallas, TX

John T. Groves, Department of Chemistry, Princeton University, Princeton, NJ

F. Peter Guengerich, Department of Biochemistry and Center in Molecular Toxicology, Vanderbilt


University School of Medicine, 638 Robinson Research Building, Nashville, TN

Aldo Gutierrez, Biological NMR Centre and Department of Biochemistry, University of Leicester,
Leicester, UK

Vijaykumar R. Holla, Department of Medicine, Vanderbilt University Medical School, Nashville, TN

Colin J. Jackson, Wolfson Laboratory of P450 Biodiversity, Institute of Biological Sciences,


University of Wales Aberystwyth, Aberystwyth, Wales, UK

Eric F. Johnson, Department of Molecular and Experimental Medicine, The Scripps Research
Institute, La JoUa, CA

Diane E. Kelly, Wolfson Laboratory of P450 Biodiversity, Institute of Biological Sciences, University
of Wales Aberystwyth, Aberystwyth, Wales, UK
XX List of Contributors

Steven L. Kelly, Wolfson Laboratory of P450 Biodiversity, Institute of Biological Sciences, University
of Wales Aberystwyth, Aberystwyth, Wales, UK

David C. Lamb, Wolfson Laboratory of P450 Biodiversity, Institute of Biological Sciences, University
of Wales Aberystwyth, Aberystwyth, Wales, UK

Thomas M. Makris, Center for Biophysics, University of Illinois, Urbana, IL

Birger Lindberg M0ller, Plant Biochemistry Laboratory, Royal Veterinary and Agricultural
University, 40, Thorvaldsensvej, DK-1871 Frederiksberg C, Copenhagen, Denmark

Andrew W. Munro, Department of Biochemistry and Department of Chemistry, University of


Leicester, Leicester, UK

Kirsten Annette Nielsen, Plant Biochemistry Laboratory, Royal Veterinary and Agricultural
University, 40, Thorvaldsensvej, DK-1871 Frederiksberg C, Copenhagen, Denmark

Mark J.I. Paine, Biomedical Research Centre, University of Dundee, Ninewells Hospital and Medical
School, Dundee, UK

Thomas L. Poulos, Department of Molecular Biology and Biochemistry and the Program in
Macromolecular Structure, University of California, Irvine, Irvine, CA

Gordon C.K. Roberts, Biological NMR Centre and Department of Biochemistry, University of
Leicester, Leicester, UK

lime Schlichting, Max Planck Institut fiir Molekulare Physiologic, Abt. Biophysikalische Chemie,
Germany

Nigel S. Scrutton, Department of Biochemistry and Department of Chemistry, University of Leicester,


Leicester, UK

Sason Shaik, Department of Organic Chemistry and the Lise-Meitner-Minerva Center for
Computational Quantum Chemistry, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel

Stephen G. Sligar, Departments of Biochemistry, Chemistry, the School of Medicine, and the Center
for Biophysics, University of Illinois, Urbana, IL

David J. Waxman, Division of Cell and Molecular Biology, Department of Biology, Boston
University, Boston, MA

Susanne N. Williams, McArdle Laboratory for Cancer Research, University of Wisconsin, Madison,
WI

C. Roland Wolf, Biomedical Research Centre, University of Dundee, Ninewells Hospital and Medical
School, Dundee, UK
1
Models and Mechanisms of
Cytochrome P450 Action
John T. Groves

1. Introduction from the hydrocarbon squalene were also shown to


derive their oxygen fiinctionality from molecular
oxygen^. Here, a single oxygen atom derived from
The reactions catalyzed by the cytochrome
molecular oxygen while the other was transformed
P450 family of enzymes have challenged and
to water. Later, the prostaglandins were shown to
intrigued chemists for more than three decades.
derive from the incorporation of two molecules of
Alkane hydroxylation and olefin epoxidation,
oxygen to form, initially, an alkyl hydroperoxide-
particularly, have attracted a sustained worldwide
endoperoxide. Thus, what appeared at first to be
effort, the allure deriving both from a desire to
an obscure process of bacteria and fiingi became
understand the details of biological oxygen activa-
recognized as a major theme of aerobic metabo-
tion and transfer and, as well as the sense that the
lism in higher plants and animals. The subsequent
development of new, selective catalysts, based on
search for "active oxygen species" and efforts to
these principles could be of considerable eco-
elucidate and understand the molecular mecha-
nomic value. The focus of this chapter is on the
nisms of oxygen activation and transfer have
advances in our understanding of the mechanisms
been richly rewarding. Novel and unusual iron
of the remarkable oxygenation reactions mediated
redox chemistry, particularly those of high-
by oxometalloporphyrins in both enzymatic and in
valent metal-oxo and metal-peroxo species, has
small molecule model systems. Particular empha-
appeared as our understanding of enzymatic
sis is on the period since the publication of second
oxidation strategies has developed.
edition of this monograph in 1995.
The activation and transfer of molecular
oxygen into its substrate by an iron-containing
enzyme was first demonstrated by Hayaishi in the 2. Oxygen Activation by
1950s^. It was shown, in some of the first mecha- Heme-Thiolate Proteins
nistically informative oxygen isotopic measure-
ments, that both the inserted oxygen atoms in The heme-containing metalloenzymes C5^o-
the conversion of catechol to c/5-muconic acid chrome P450^, chloroperoxidase (CPO)"*' ^, nitric
derived from O2 and not water. These findings oxide synthase (NOS)^, and their relatives catalyze
challenged the then firmly held view that oxygen a host of crucial biological oxidation reactions.
in biological molecules was derived exclusively Highly specific P450s are involved in the selective
from water via hydration processes. The bios)^- oxygenations of steroid and prostaglandin biosyn-
thesis of cholesterol and its precursor, lanosterol. thesis. Myeloperoxidase, which is a CPO, is an

John T. Groves Department of Chemistry, Princeton University, Princeton, NJ.


Cytochrome P450: Structure, Mechanism, and Biochemistry, 3e, edited by Paul R, Ortiz de Montellano
Kluwer Academic / Plenum Publishers, New York, 2005.
John T. Groves

integral part of the immune response, and NOS is biological substrates. The other oxygen atom is
the source of the highly regulated signal trans- transformed into H2O. All three proteins are pro-
ducer, nitric oxide (NO). Certain fungal CPOs and posed to initiate their chemistry through the oxi-
bacterial P450s have been genetically engineered dation of a resting iron(III) state (1) to a reactive
for large-scale biotransformations^"^^. The active oxoiron(IV) porphyrin cation radical intermediate
sites of these three protein families, known in (2) (Figure 1.1). A depiction of the CPO active
detail from a number of X-ray crystal structures'^' site derived from the crystal structure of this
^^~^^, are remarkably similar. All three have an protein from Caldariomyces fumago is shown in
iron protoporphyrin IX center coordinated to a Figure 1.2. The structure, biochemistry, molecular
cysteine thiolate. All of them are oxidoreductases biology, and the chemistry of cytochrome P450
that activate molecular oxygen (O2), in the cases and related model systems have been extensively
of P450 and NOS, or hydrogen peroxide in the reviewed ^^-^^.
case of CPO, at the iron center and incorporate Our understanding of the mechanism of action
one of the oxygen atoms into a wide variety of of these heme proteins comes from the direct

Figure 1.1. Iron(III) protoporphyrin IX with a cysteinate as the axial Hgand (1), which is typical of cytochrome
P450, chloroperoxidase (CPO), and nitric oxide synthase (NOS) enzymes. The active oxygen species of these
proteins and related heme enzymes is an oxoiron(IV) porphyrin cation radical (2), often called compound I.

Figure 1.2. Crystal structure of the active site of chloroperoxidase (CPO) (EC 1.11.1.10) from C. fumago. Protein
framework is shown as ribbons. The heme is buried in a hydrophobic binding pocket containing the iron-coordinating
cysteinate ligand. Adapted from the X-ray atomic coordinates of CPO^.
Models and Mechanisms of Cytochrome P450 Action

observation of intermediates in the catalytic cycle


through a variety of spectroscopic techniques, the
use of diagnostic substrates with mechanistically
revealing rearrangements during oxidation, and R-OH S-Cys
the parallel development of the chemistry of 3 \
R-HO
synthetic metalloporphyrins. The principal fea- R-H
tures of the consensus mechanism of cytochrome
P450^^ are as outlined in Scheme 1.1:
^S7
* O r / S-Cys
R_H 92 ^ 4
(1) binding of substrate to the enzyme,
sometimes accompanied by a spin-
state change of the iron, to afford an S-Cys S-Cys
enzyme-substrate adduct 3; 6 5
(2) reduction of the ferric cytochrome
P450 by an associated reductase with Scheme 1.1. Consensus catalytic cycle for oxygen
activation and transfer by cytochrome P450.
an NADPH-derived electron to the fer-
rous cytochrome P450 4;
(3) binding of molecular oxygen to the fer-
rous heme to produce a ferrous the oxygen donor to form the Fe=0 intermediate
cytochrome P450-dioxygen complex 7 and the subsequent oxygen transfer to form
5, similar to the situation in oxymyo- the substrate complex 8 that has been termed
globin; oxygen reboun(f^. Such an iron-oxo species
(4) a second one-electron reduction and (compound I) has been observed for the CPO of
protonation to arrive at the Fe(III)- C. fumago^^ but the active species of cytochrome
hydroperoxy complex 6; P450 has remained elusive. Very recently, it has
(5) protonation and heterolytic cleavage of been shown that an intermediate with the spectral
the O-O bond in 6 with concurrent pro- properties similar to those of CPO compound I
duction of a water molecule to form a and the model iron porphyrin systems is formed
reactive iron-oxo intermediate 7; upon the oxidation of Cypll9, a thermostable
(6) and, finally, oxygen-atom transfer from cytochrome P450, with a peroxyacid, analogous
this iron-oxo complex 7 to the bound to the model systems^^. Consistent with the
substrate to form the oxygenated prod- high reactivity expected for P450 compound I,
uct complex 8. Product dissociation this intermediate decayed with a rate constant
completes the cycle. of 29 s~^ at4°C. Interestingly, similar experiments
with P450^^j^, the camphor-oxidizing enzyme
There were a number of important realizations from Pseudomonas putida, resulted in an
in the course of elucidating this mechanism. That iron(IV)-protein tyrosine radical species, presum-
hydrogen peroxide, alkyl hydroperoxides, peroxy- ably via a one-electron oxidation of Tyr96 which
acids, periodate, and iodosylbenzene were also is only 9.4 Afi-omthe iron center^^.
functional with cytochrome P450 suggested that
the chemistry of "oxygen activation" was the
two-electron reduction of molecular oxygen to
hydrogen peroxide and that, in analogy to the per- 3. Mechanism of Hydroxylation
oxidases, the active oxygen species was a ferryl by Cytochrome P450
(or oxene) complex Fe==0, formally iron(V).
It was shown that a synthetic oxoiron(IV) por- There has been much discussion in the field
phyrin cation radical species could be formed at about the oxygen transfer process 6 ^ 7 ^ 8.
low temperature by the oxidation of an iron(III) The oxygen rebound mechanism in Scheme 1.1 is
precursor with peroxyacids (9 -^ 10)^^. Inter- consistent with the stereochemical, regiochemical,
mediate 10 did have the requisite reactivity to and allylic scrambling results observed in the
transfer an oxygen atom to hydrocarbon sub- oxidation of norbomane, camphor, and cyclohex-
strates. It is this oxygen-atom transfer from ene by cytochrome P450. The hydroxylation of
John T. Groves

a saturated methylene (CH2) in norbomane was inconsistent with an insertion process and indicate
accompanied by a significant amount of epimer- that the C-H bond is essentially half-broken in a
ization at the carbon center^^. Thus, the hydroxy- linear [ O H C ] transition state thus providing
lation of exo-exo-^xo-exo-tetradeuterionorbomane strong evidence for a nonconcerted mechanism.
by P450 2B1 and the hydroxylation of camphor by Significantly, model iron porphyrin systems dis-
P450^gj^^^ gave ^jco-alcohol with retention of the played the same behavior for both the norbor-
^xo-deuterium label (Scheme 1.2). The hydroxyla- nane^^ and cyclohexene^^ substrates. Thus, one
tion of selectively deuterated cyclohexene pro- concludes that the epimerization and allylic scram-
ceeded with substantial allylic scrambling^^. The bling processes are intrinsic properties of the
intrinsic isotope effects for the oxygen insertion oxygen transfer event from an oxoiron complex.
into a C-H bond are very large, in the range Another revealing probe of the nature of
of 10-13.5. These large isotope effects are P450-mediated hydroxylation is a study of the

P(H)
'D(H)

P D P OH

P450

HO'
D D D D D D

0=Fe
H
Z\

+"
HO—Fe Fe—OH

Z\ 9 y\^

OH OH
Z\ H
^ ^

Scheme 1.2. Epimerization and allylic scrambling observed for cytochrome P450 catalyzed hydroxylation.
Models and Mechanisms of Cytochrome P450 Action

Scheme 1.3. Hydroperoxide isomerase activity of cytochrome P450 is intramolecular.

hydroperoxide isomerase activity of these difference is likely to be due to the requisite


enzymes. 1-Hydroperoxyhexane has been shown prepositioning of the hydroperoxide as a ligand of
to afford 1,2-dihydroxyhexane upon exposure to Fe(III). During turnover via oxygen reduction,
P450 2 B P ^ This is an unusual reaction since the however, the positioning of the substrate will be
oxidizing equivalents of the hydroperoxide have dictated by substrate-active-site interactions. An
been used in this case to hydroxylate the neigh- important conclusion from these studies is that
boring methylene group. A mixed, double oxygen product selectivity can be affected significantly by
label experiment established that the rearrange- substrate mobility. Accordingly, changes in prod-
ment was intramolecular (Scheme 1.3). Thus, the uct selectivity, which have been used to suggest
terminal hydroperoxide oxygen was incorporated alternative oxidants, need to be interpreted with
into the adjacent C-H bond. This reaction path- caution.
way is pertinent to the discussion about the nature The hydroperoxy iron(III) complex 6 has also
of reactive P450 intermediates since the same been suggested to effect substrate oxygenations
ferryl species (compound I) can be accessed by based on observed changes in product ratios
this "peroxide shunt" pathway. Analysis of the and loss of hydrogen peroxide (uncoupling)
diols derived from chirally labeled 2-deuterio- upon P450 active-site mutations^^~^^. An impor-
1-hydroperoxyhexane showed that there was a loss tant recent advance has been the development of
of stereochemistry at the hydroxylated carbon cryospectroscopic studies by Hofftnan et al. that
center. Accordingly, the results support a mecha- have allowed the stepwise interrogation of inter-
nism involving initial peroxide heterolysis, hydro- mediates depicted in Scheme l.P^. Thus, the
gen abstraction at the adjacent methylene, and injection of an electron into complex 5 via
radical recombination to afford the product diol. 7-radiation, followed by thermal annealing of
The result is revealing since O-O bond homolysis the sample has produced EPR and ENDOR evi-
to form a hexyloxy radical should lead instead dence for the formation of, first, a hydrogen
to 7-hydrogen abstraction and products derived bonded iron-peroxo species and then the
therefrom. iron-hydroperoxo complex 6. While no ferryl
Kupfer et al. have used this P450-hydroperoxide intermediate 7 was observed, the product alcohol
isomerase reaction to explore substrate mobility was found to be formed with its oxygen atom
at the enzyme active site during the hydroxylation coordinated to the iron center and with the
event^^. Isomerase substrates were found to remain substrate-derived proton attached to the product
in proximity to the P450 oxoferryl intermediate alcohol as depicted in structure 8 (Scheme 1.1).
and were rapidly captured by the oxidant with This arrangement has important mechanistic
high efficiency. Monooxygenase substrates, by implications since, if a ferryl species 7 were the
contrast, apparently bind to ferric P450 in multiple immediate precursor of the product complex 8,
orientations and undergo more extensive sub- then coordination of the product hydroxyl oxygen
strate reorientation prior to oxidative attack. This would be a necessary consequence. By contrast, if
John T. Groves


ko/^W" k^/V°-H ko.V°-H
I I } H

nucleophilic addition "^^^^ homolytic cleavage desaturation

Scheme 1.4. Proposed mechanism for the deformylation typical of P450 aromatase activity.

the hydroperoxo species 6 were the source of to the reactions of enzymes such as cyclohexanone
the electrophihc oxygen, then water would be monooxygenase that proceed through a flavin
coordinated to iron rather than the product alco- 4a-hydroperoxide^^. Here, only electron-deficient
hol. A product complex such as 8 could also be the olefins react to afford epoxides even though the
source of cationic rearrangement products that are flavin hydroperoxide is 2 X 10^ times more reac-
sometimes observed during P450 oxygenations. tive than a simple alkyl hydroperoxide"^. The reader
Significant recent advances in computational is referred to an insightful review by Watanabe for
approaches to the study of biological catalysis, a thorough discussion of the various modes of reac-
and the applications of these techniques to the tivity of peroxoFe(III) porphyrins (Scheme 1.4)"^^.
cytochrome P450 mechanism have also been illu- The hydroxylation of a C-H bond does seem to
minating. Thus, Shaik et alP and Yoshizawa require the full formation of a reactive ferryl
et al?^, have presented the results of a density intermediate as in 11. This applies both for the
functional theory (DFT) analysis of the reactivity reductive activation of dioxygen and for the very
of hydroperoxyiron(III) complexes such as 6. revealing cases of alkyl hydroperoxide isomeriza-
Both groups conclude that a hydroperoxyiron(III) tion catalyzed by P450 discussed above^^' ^^. For
porphyrin, Fe(III)-OOH, would be an implausible P450s in which the proton relay system has been
primary oxidant. The protonation and heterolytic disrupted by active-site mutations, one would
O-O bond cleavage of 6 to afford a ferryl species expect that particularly reactive substrates could
analogous to 7 was found to proceed with almost interact with the proximal oxygen earlier in this
no energetic barrier, in accord with earlier experi- reaction profile as shown in 12. While similar
mental results for the oxidation of an iron(III) atomic trajectories and electronic charge redistrib-
porphyrin 9 to an oxoiron(IV) porphyrin cation utions are followed in each case, the former (11) is
radical species 10 with a peroxyacid^^. Further, analogous to the S^^l reaction in organic chem-
the oxygen transfer from 7 to ethylene to form an istry, generating a discrete ferryl intermediate,
epoxide proceeded with only a low barrier. It was while the latter (12) is Sj^2-like, requiring assis-
concluded that the DFT calculations exclude a tance from the electron-rich substrate. Indeed, in a
hydroperoxyiron(III) intermediate such as 6 as recent report by Sligar and Dawson, mutation of
a reactive, electrophilic oxidant. Several modes of the conserved active-site threonine-252 to alanine
oxygen transfer from the hydroperoxide interme- in P450^^j^ was shown to disable camphor hydrox-
diate encountered exceedingly high barriers for ylation while maintaining some reactivity for
reaction. The lowest energy of these was an inter- more reactive olefinic substrates'^^. Similarly, two
action of the substrate ethylene with ih& proximal, reactive intermediates, as suggested by Jones"^^ for
iron-bound oxygen of the Fe(III)-OOH ensemble. the reactions of a thioether substrate, and also for
Nucleophilic reactions of a hydroperoxyiron(III) model porphyrin systems described by Nam"*^,
intermediate 6, as have been suggested by could reasonably derive from a mechanistic spec-
Akhtar'*^, Robinson'^^ and Vaz and Coon"^^, for the trum of this type. An important precedent for this
deformylation reactions characteristic of the P450 behavior is seen in the reactions of peroxyacids
aromatase, do seem to be suggested by the signifi- with model Fe(III) porphyrins. Thus, Watanabe
cant basicity of the distal, hydroxylic oxygen and Morishima have shown that the iron-
found in the calculations for the Fe(III)-OOH coordinated peroxyacid 9 reacted with olefins
group. This mode of reactivity is highly analogous at the iron-coordinated oxygen atom in nonpolar
Models and Mechanisms of Cytochrome P450 Action

Thr-OH Ala-H
.O-H O-H
H

12

solvents to give epoxides but would not react with imagination of chemists more than the hydro-
saturated hydrocarbons'^^' ^^. By contrast, the same xylation of saturated carbon centers. Metal-oxo
oxoiron(IV) porphyrin cation radical, 10, was reagents such as chromates and permanganate can
formed with a variety of peroxyacids in more perform reactions of this type but are notoriously
polar media. This effect is also seen in model nonselective and must be used under forcing
compounds with a thiolate ligand to iron^^. The conditions. The selective hydroxylation of hydro-
protein-derived hydrogen bonds to the axial thio- carbons remains one of the grand challenges
late ligand to iron in P450^^^ have been shown to for the chemical catalysis community. How can
affect the O-O bond cleavage^ ^ a protein create an iron intermediate reactive
enough to hydroxylate even as inert a substrate as
cyclohexane and not oxidize the relatively fragile
4. Mechanisms and Molecular protein superstructure? What is the electronic
Trajectories for Hydroxylatlon structure of that intermediate and what are the
by Cytochrome P450 molecular pathways for oxygen insertion into a
C-H bond? Without clear answers to these ques-
Among all the varied reactions mediated tions, the chemical catalysis performed by these
by cytochrome P450, none has captured the metalloenzymes will remain an enigma and our
8 John T. Groves

attempts to draw conclusions will be without in support of this view^^. A nonconcerted pathway
physical meaning. Without knowledge of the for C-H bond cleavage is strongly supported
mechanism, we learn nothing of predictive value by the observations of a variety of molecular
that could be applied to other systems such as rearrangements that are known to accompany
the rational design of enzyme inhibitors or the P450-mediated hydroxylation as discussed above.
development of enzymatically inspired catalysts. Initially it was clear that the kinds of rearrange-
Presented in Scheme 1.5 is the range of ments observed were consistent with the forma-
mechanisms that have been considered as likely tion of a caged substrate radical at the heme active
candidates for the cytochrome P450-catalyzed site. The intermediate radical could be trapped in
hydroxylation of hydrocarbons and those of model a subsequent step. Both P450 enzymes and model
iron, manganese, and ruthenium porphyrins. systems showed a nonstereospecificity for the
A linear, homolytic transition state, as in interme- hydrogen removal step from norbomane or cam-
diate H, best fits the available data, such as the phor substrates. Such a process was counter-
very large hydrogen isotope effects. Indeed, exten- indicative of a cationic pathway to explain the
sive similarities to the hydrogen abstraction observed rearrangements. The results rule out
observed by cytochrome P450 and a ^butoxy rad- freely diffusing radicals, but a short-lived substrate
ical have been presented by Dinnocenzo and Jones radical would explain the observed results.

O H-R

!f7
S-Cys

/
H; ,R H
O o
47 S-Cys J
47 S-Cys

H^ /R H.^.R ,H R
Caged O
radical y^l

S-Cys
MmmmmmmmJr

Cys-S
237S-Cys
8 R
\ electron
/ transfer

47 S-Cys
Ion pair
Scheme 1.5. Pathways for oxygen-atom transfer from the active ferryl species 7 of heme-thiolate enzymes such
as cytochrome P450 to form the product alcohol coordinated to the ferric, resting form of the protein (8).
Models and Mechanisms of Cytochrome P450 Action 9

It was shown by Ortiz de Montellano et al. that porphyrin shown in Figure 1.3 has been found to
bicyclo[2.1.0]pentane was oxidized by rat hver hydroxylate ethylbenzene with a 70% ee. Stereo-
microsomes to a 7:1 mixture of e«^o-2-hydroxy- selective deuteration of the substrate revealed
bicyclo[2.1.0]pentane and 3-cyclopenten-1 -ol, that the pro-i? hydrogen of ethyl benzene was
consistent with a radical ring-opening reaction^^. hydroxylated with nearly complete retention of
Apphcations of the "radical-clock" method by configuration at carbon while the pro-*^ hydrogen
Ingold^"^ and by Newcomb^^ began to measure the underwent significant racemization (Figure 1.3).
lifetime of the suspected radical cage intermedi- Interestingly, the partition ratio, retention/inversion,
ate. The rate constant for the rearrangement of was nearly the same for the two enantiomers of
bicyclo[2.1.0]pent-2-yl radical to 3-cyclopenten- ethylbenzene-<3?^, suggesting similar mobility of the
1-yl radical was determined to be 2.4 X 10^ s~^ at radical intermediate at the active site.
room temperature by using laser flash photolysis Evidence for a similar type of host-guest com-
techniques'^. Thus, a rate constant of ^QJ^ = 1.7 X plementarity effect has been presented recently
10^^M~^s~^ was estimated for the rebound by Wiist for the hydroxylation of limonene by
process. Radical clocks with very fast rearrange- the limonene-6-hydroxylase, P450 CYP71D18'l
ment times were shown to produce less rearrange- The regiochemistry and facial stereochemistry
ment than slower clocks in the P450-mediated of the limonene hydroxylation was found to be
hydroxylations, however. The results led Newcomb determined by the absolute configuration of the
to question whether a radical pathway existed since substrate. Thus, (—)-(45)-limonene gave {—)-trans-
the apparent lifetimes revealed by these probes carveol as the only product, whereas (+)-
were in the range of 100 fs, too short to represent (4i?)-limonene afforded mostly (+)-c/5-carveol
a bona fide intermediate'^. Several suggestions in a mixture of products. Specifically deuterated
have been considered to resolve this dilemma and limonene enantiomers revealed that (4i?)-limonene
the question is still an area of active experiment has sufficient freedom of motion within the active
and debate. As shown in Scheme 1.5, the transi- site of CYP71D18 to allow formation of either the
tion state for hydrogen abstraction will position trans-3- or c/5-6-hydroxylated product. However,
the active oxygen only a few tenths of an the kinetic isotope effects resulting from deu-
AxigsixoxR farther from the hydroxylated carbon terium abstraction were significantly smaller than
atom than the transition state for the ultimate expected for an allylic hydroxylation. Signifi-
C-O bond formation. Thus, the extent of radical cantly, the oxygenation of (4i?)-limonene gave
rearrangement might be expected to depend criti- trans-carvQol with considerable allylic rearrange-
cally on the tightness of the radical cage and the ment and stereochemical scrambling, while the
ensemble of steric and electronic forces experi- formation of (+)-cw-carveol proceeded with high
enced by the incipient radical within the cage. stereospecificity for C6 hydrogen abstraction and
Even the molecular makeup of the active site will little rearrangement. These results are analogous
depend on how the substrate fills the site, leaving to the ethylbenzene hydroxylation by the chiral
room for movement of amino acid side chains in iron porphyrin described above, in that epimeriza-
the vicinity of the substrate or allowing additional tion and allylic rearrangement apparently depend
water molecules into the active-site area. The upon the fit and mobility of the substrate at the
extent of rearrangement detected by a particular active site. Another informative probe of substrate
probe may simply reflect a facile molecular tra- mobility at the active site using kinetic isotope
jectory from the hydrogen abstraction transition effect has been presented by Jones and Trager'^.
state to the hydroxylation transition state in this For a reaction that involves a paramagnetic
variable environment. For substrates with a very iron-oxo intermediate and proceeds to produce
strong C-H bond and a small steric size, both paramagnetic radical intermediates, it is likely
effects would push the reaction coordinate toward that spin-orbit coupling effects and the spin states
a tighter radical cage. of reacting intermediates may offer another sig-
Indeed, it has been shown that the effective nificant consideration^^. Schwarz first suggested
lifetime of a radical intermediate can even be that the unusually slow reaction of FeO^ with
affected by the stereochemistry of the hydrogen hydrogen in the gas phase was due to spin-
abstraction event^^. The chiral, binaphthyl conservation effects that were imposed on these
10 John T. Groves

9 9 p p
H07""'H H7""'0H D'|"""0H H0:/""'D
Me Me Me Me
SD„

Me
kpH ksD
92% 8%
P-Fe-0. O-Fe-P
D/
Me H'
Me
87% 5% 6% / \ 2%

9 p 9 p
Me Me Me Me
R-Dre ^Dre ^Hsi ^Hsi

Figure 1.3. Catalyic asymmetric hydroxylation by a chiral, binaphthyl porphyrin. The stereochemical outcome of
a hydroxylation depends upon the steric fit of the substrate.
Models and Mechanisms of Cytochrome P450 Action 11

intermolecular encounters^ ^. Detailed DFT calcu- intermediate to the product via formation of a
lations on this simplest iron-oxo electrophile carbon-oxygen bond. By contrast, the low-spin
showed that there was a spin-state crossover dur- trajectory could proceed to products without
ing the H-H bond cleavage step to form a species encountering this barrier. This two-state hypothe-
H-Fe-OH^, and another spin crossover leading to sis could provide a way out of the mechanistic
the product Fe(OH2)^. Thus, the lowest energy dilemma presented by the radical clock results
pathway for the reaction involved crossing from since the apparent timing of the clocks would
an initial high-spin, sextet state for the oxidant depend upon the relative importance of the high-
FeO^ to a low-spin, quartet state near the transi- and low-spin pathways that would likely vary
tion state for H-H bond cleavage. While such from substrate to substrate.
effects are common for first-row elements as, for Evidence for short-lived substrate radicals
example, with singlet and triplet carbenes, "spin has been presented recently for the oxidation of
forbiddenness" has usually been discounted for the mechanistically diagnostic probe molecule
reactions involving transition metals. However, norcarane by cytochrome P450^^. Among the
the successful application of DFT calculations to products found with P450 BM3, was 1.3% of the
explain the unusual behavior of FeO^ suggests radical rearrangement product hydroxymethyl-
that these effects may be significant in the area of cyclohexene while the cation rearrangement
oxidative catalysis. product 3-cycloheptenol was not observed with
Shaik has applied these considerations to that isozyme (Figure 1.4). An alternate interpre-
examine interactions of a prototype substrate, tation of similar data, involving unusual behavior
methane, with a ferryl intermediate similar to 7 to of the probe molecule at the active site, has
probe this chemistry of P450^^. The results are also been presented^"^. In all known cases of reac-
very revealing. The ferryl intermediate was shown tions involving a radical intermediate, this nor-
to have two nearly isoenergetic electron configura- carane probe produces a product derived from
tions, doublet and quartet, depending upon whether the 3-cyclohexenylmethyl radical, as the major
the unpaired electron in the porphyrin cation rearrangement product. The rate constant for the
radical is ferromagnetically or antiferromagneti- radical rearrangement of the 2-norcaranyl radical
cally coupled to the triplet ferryl center. Indeed, has been found to be 2 X 10^ s~^ By contrast,
both situations are known in enzymatic compoimds for reactions proceeding through discrete carboca-
I and model systems. The calculations indicate tions, rearrangement leads instead to 3-cyclohep-
that the transition state for C-H bond cleavage tenol as the major rearrangement product.
does look like the extended arrangement H in The extent of observed rearrangement with a
Scheme 1.5. Here, however, the molecular trajec- panel of P450 enzymes leads to a radical lifetime
tories for the high-spin and low-spin reaction in the picosecond to nanosecond regime, certainly
coordinates diverge. For the high-spin pathway, long enough to be considered an intermediate
there was a discemable intermediate caged radical (Figure 1.5). A consistent timing was found for
state with the carbon center interacting weakly several similar probes that were all small, aliphatic
with the iron-hydroxide. A significant energy hydrocarbons. Smaller amounts of cation-derived
barrier was found for collapse of this high-spin products were also observed and were attributed

OH OH

P450 bm3

02
R~1.3%
Figure 1.4. Norcarane as a molecular probe of radical intermediates during C-H hydroxylation by cytochrome
P450 BM3.
12 John T. Groves

Radical Clock Timing for Cytochrome P450


2.5X10'

2 X10' P Radical lifetime = 64 ps

^ 1.5X10' h
o

IXIO' \- k = 1.55x10
rebound

R = 0.997

5X10'
k = k (rear/unrear)
rear rebound

-J I I L_

0.00 0.02 0.04 0.06 0.08 0.10 0.12 0.14


Product Ratio (rear/unrear)
Figure 1.5. Plot of radical rearrangement rate constant vs observed product ratios for P450-mediated
hydroxylation of bicyclo[2.1.0]pentane, norcarane, and spiro[2,5]octane.

to a competing electron-transfer oxidation of the and Yoshizawa^^. While two spin states of the
incipient radical, a well-precedented process. By reactive intermediate were also found in this work,
contrast, the hydroxylation of norcarane with a it was the high-spin quartet state of the oxoferryl
ruthenium porphyrin catalyst that proceeds through that was lower in energy. Also significant in these
a reactive oxoruthenium(V) porphyrin intermedi- calculations, were the findings that there was an
ate, afforded no detectable rearrangement. interaction between the incipient substrate radical
DFT calculations on the ruthenium-mediated upon hydrogen abstraction and the Fe-OH center
hydroxylation show that the low-spin reaction at the P450 active site and that there was a
trajectory is preferred throughout, in accord with 3.3 kcalmol"^ activation energy for the highly
general expectations for the behavior of second- exothermic radical rebound to form the product
row transition metals^^' ^^. The ruthenium analog alcohol (Figure 1.6(a)). Such an interaction would
was found to be more electrophilic than its be expected to retard radical rearrangement rates,
iron complex, having lower hydrogen abstraction providing another possible avenue for the mis-
barriers. Thus, the data for the iron and ruthenium timing of the clocks. The reaction profile for the
porphyrin systems is in accord with the predic- oxygen rebound pathway of c)^ochrome P450
tions of theory that a radical rebound process computed by Shaik is presented in Figure 1.6(b)
is viable for iron which has an accessible high- for comparison. Computations on the details of
spin state but not for ruthenium which is always C-H bond cleavage in camphor by a P450 model
low-spin. described by Friesner have revealed an unusually
The hydroxylation of camphor by an oxoferryl low energy barrier for this process^^. The primary
porphyrin has also been described by Kamachi contribution to stabilization of the transition state
Models and Mechanisms of Cytochrome P450 Action 13

2.329(2.489)0^
/ Final complex

r (Fe-N)avg = 2.018 (2.020)


-43.1

Hydroxycamphor complex
r (Fe-N)avg = 2.020 (2.020)

(b)

Alk
xH
O
11
•Fe-
I
SH ^P -I-
SH

C-H Activation Reorientation Rebound


Figure 1.6. (a) Energy level diagram and reaction coordinate computed by Yoshizawa et al for the hydroxylation
of camphor by a ferry 1-porphyrin cation radical (7). Adapted from ref. [67]. (b) Reaction profile for oxygen rebound
computed by Shaik et al. Adapted from refs [60], [62] and unpublished material from S. Shaik.
14 John T. Groves

was attributed to the interaction of positively metal ion centers such as Li^ (ref [72]). Thus,
charged residues in the active-site cavity with for a stepwise reaction via the caged radical inter-
carboxylate groups on the heme periphery. Addi- mediate in Scheme 1.5, a spectrum of apparent
tional experiments on oxoferryl species of known lifetimes, perhaps dependent on such effects as
electronic configuration would seem to be neces- weak dipolar interactions and even vibrational
sary to address these questions. state, might be observed for rebound through tran-
Other, more exotic factors such as nonstochas- sition state R to intermediate 8. Consideration of
tic behavior^^ and tunneling effects^^, could also the energy landscape for C-H hydroxylation
be involved in causing the mistiming of events (Figure 1.7) suggests that the C-H bond cleavage
during C-H bond hydroxylation. Indeed, a and concomitant FeO-H bond formation will
carbene ring-expansion reaction was very recently occur on a high-energy plateau, since the scissile
found to have a large quantum-tunneling effect C-H bond should be similar in energy to the form-
that significantly affected the observed rate^^ ing FeO-H bond. Accordingly, the intrinsic
High-level calculations indicated that a thermal, exothermicity of the hydroxylation reaction will be
over-the-barrier process, and quantum tunneling expressed in the C-OH bond-forming step, hi such
of carbon were still competitive even at room a scenario, it becomes more clear as to how small
temperature. Applied to C-H hydroxylation by a changes in bond energies and weak interactions
reactive oxidant, this situation could give the of the reaction ensemble along the reaction coor-
appearance of multiple oxidants and non- dinate could have a significant effect on the out-
Arrhenius behavior. Further, computations have come, for example, positional or stereochemical
suggested that the speed of radical clocks can be scrambling, by shifting the position of the transi-
made to run fast via interactions with even simple tion states along the reaction coordinates.

H-R

M -R

Figure 1.7. Energy landscape for aliphatic hydroxylation by cytochrome P450.


Models and Mechanisms of Cytochronne P450 Action 15

The nonheme diiron hydroxylases, such as has been characterized as a bis-|UL-oxoiron(IV)


methane monooxygenase (MMO)^^ and AlkB, the intermediate. Both AlkB^^ and MMO^'^' ^^ have
(o-hydroxylase from Pputida, have also yielded to been interrogated recently with the diagnostic
similar structural, spectroscopic, and mechanistic probe norcarane and both have shown the radical
probes. Interestingly, there are striking similari- rearrangement product, hydroxymethylcyclo-
ties between the consensus mechanism for the hexene. With MMO, it was possible to show that it
heme and nonheme iron proteins (Figure 1.8). For was the reactive intermediate Q that was interact-
MMO, the resting enzyme has both iron centers in ing with the substrate probe. For the histidine-rich
the ferric state. Reduction and binding of oxygen hydroxylase, AlkB, the results were particularly
again produces a peroxo intermediate which is striking since 15% of the product was indicative
oxidized to a reactive species, compound Q, that of the radical rearrangement pathway. Similar

cell membrane

GIU

^ O N

His /
Asp

kr = 2 x l 0 « s - ]

/ \ /K N ^ O N
N W O N

Figure 1.8. Competing radical rearrangement and electron transfer during norcarane hydroxylation by the
histidine-rich hydroxylase XylM.
16 John T. Groves

results have been obtained recently for the related to citrulline and NO (Scheme 1.6). The initial
histidine-rich, diiron hydroxylase XylM^^. A sig- A/-hydroxylation of L-Arg to NHA by O2 is similar
nificant aspect of this work was that it was per- to the C-hydroxylations of P450 described above.
formed on whole cells and clones into which the The second step of the NOS reaction is unusual
AlkB and XylM genes had been introduced. Thus, because it is a three-electron, aerobic oxidation of
mechanistically informative biochemistry can be NHA to NO and citrulline^^' ^2.
obtained from this type of biological screen. Our current understanding of these processes
is constrained by the fact that the consensus mech-
anism (Scheme 1.6) contains several unknown
5. On the Mechanism of intermediates and unprecedented processes in
Nitric Oxide Synthase the second step. There have been a number of
significant recent advances in the mechanistic
Nitric oxide (NO) is produced by the heme- enzymology of NOS and the structures of the
containing metalloenzyme NOS (EC 1.14.13.39). enzyme-substrate complexes. However, while
Several NOS isoforms are homodimers with each these results have provided confirmation of the
monomer containing binding sites for NADPH, basic tenets for the A/-hydroxylation of arginine in
FMN, FAD, calmodulin, tetrahydrobiopterin the first part of the consensus mechanism, the
(H4B), and a heme groups. Similar proteins are results raise important questions regarding the
found in animals, plants, and bacteria indicating oxidation of NHA and the release of NO. Thus,
that this is a widely distributed and highly Poulos has shown that the X-ray structure of
conserved process in nature. The H4B cofactor is NOS with NO bound to the heme iron center as a
especially important, serving structural, allosteric, structural surrogate for O2, places the NO oxygen
and redox functions^^"^^. The X-ray crystal struc- within hydrogen-bonding distance to the co-N-H^^.
tures of substrate-bound NOS show that both the This juxtaposition provides support for the notion
substrate and H4B are bound at the heme site with that the arginine proton assists the heterolysis
a substantial network of hydrogen bonds^^"^^. of the FeO-0 bond during oxygen activation to
NOS catalyzes the two-step, five-electron oxida- afford the ferry 1 intermediate in a P450-like
tion of L-arginine via A^-hydroxyarginine (NHA) process. However, the same structure would have

PPIX-Fe"^-X PPIX-Fe"-02 ^ a

OH H ^ C OH H
1+ I
HzN^NH; H-N^N-H ^
NADPH NADP+ I 1/2 NADPH 1/2 NADP* T |
.NH V 4

O2 H2O
/NH
H-
> i .N-H O Glu.

HJ'N^^COO- H3N " C O O -


H3N ^ C O O -
NHA electron
L-arginine
transfer N-i
PPIX—Fe'"—OH '^
P P I X — F,in_
e"*~0-0
H2NA>N-6~

PPIX-Fe*"-0-OH N - H
O GIu.

H3N COO- NO H3N ^ C O O - H3N ^COO


citrulline p

Scheme 1.6.
Models and Mechanisms of Cytochrome P450 Action 17

difficulty accommodating both the hydroxyl 6. Synthetic


group of NHA where it would have to be, and the
Oxometalloporphyrins as
O2 of the next cycle. Significantly, very recent
EPR/ENDOR results by Hoffman et alP have Models for Cytochrome P450
indicated that the incipient hydroxyl of NHA is
formed bound to the heme iron in a manner simi- Studies using synthetic metalloporphyrins
lar to C-H hydroxylation by P450. This unsus- (Figure 1.10) as models for cytochrome P450
pected arrangement is consistent with an oxygen have afforded important insights into the nature
rebound scenario but inconsistent with the X-ray of the enzymatic processes^^' ^^. Indeed, each of
structure of NHA bound to the active site of NOS the intermediates shown in Scheme 1.1 has
obtained by Tainer et al}^, which shows the been independently identified by model studies
A^-hydroxy group to be displayed away from the using synthetic analogs, especially me^o-tetraaryl
heme iron. Thus, it appears that NHA, as biosyn- porphyrins'^'''.
thesized from arginine, may be formed in a non- The first report of a simple iron porphyrin sys-
equilibrium configuration with respect to the NOS tem that effected stereospecific olefin epoxidation
heme active site. In this light, the observation by and alkane hydroxylation was reported in 1979
Silverman^"^ that oxime ethers of the type (Scheme 1.7). This system introduced the use of
NHA-OR are active substrates for NOS and that iodosylbenzene as an oxygen-transfer agent to
NO is produced from these species is very mimic the chemistry of C5^ochrome P450'^.
informative. The NHA-NOS crystal structure It was later discovered that the reactive inter-
suggests that NHA-OR derivatives can be accom- mediates in the iron porphyrin model systems
modated at the active site in the configuration were high-valent oxoiron porphyrin complexes.
shown in Figure 1.9^^. Thus, the 0 - H of NHA A green oxoiron(IV) porphyrin cation radical
may not be mechanistically significant because species (13) has been well characterized by vari-
the mechanism of the A^-hydroxylation of L-Arg ous spectroscopic techniques, including visible
to afford NHA is still available to the oxime spectroscopy, NMR, EPR, M5ssbauer, and
ethers. EXAFS (Figure 1.11)90-98. It has recently been
shown by Nam and Que that the oxygen-atom
transfer from certain iodosylarenes is reversible
with some iron porphyrins. For the case of 1,2-
difluoro-4-iodobenzene both an oxoferryl species
and an iodosyl-ferric species were observed to be
in equilibrium^^.
A family of oxoiron(IV) porphyrin cation rad-
ical species (13) with different axial ligands has
been reported, each of which displayed a charac-
teristic ^H-NMR for the pyrrole protons. Addition
of methanol replaced each of these ligands with
a solvent molecule (8 = —22.8)^^0. A report of
the imidazole and /7-nitrophenolate complexes of
13 has also appeared, affording closer model com-
plexes for the compounds I of peroxidase and
catalase, respectively^0^. The oxoiron(IV) por-
phyrin cation radical complexes 13 were shown to
be highly reactive as oxygen-atom transfer agents
toward olefins and hydrocarbons, that is, reacting
with olefins to afford epoxides and with alkanes to
give alcohols^o^-^O"*. Notably, 13-4-Me-Im was
also reactive toward norbomene, giving a 67%
yield of the corresponding epoxide^^o. Significant
Figure 1.9. Structure suggested for the active site of
variations have been observed in the reactivity and
RO-NHA-bound murine iNOS.
18 John T. Groves

Figure 1.10. Typical synthetic tetraaryl porphyrins.

lO
o

lO
HOv-^H

Scheme 1.7. Olefin epoxidation and alkane hydroxylation catalyzed by an iron porphyrin, Fe'"(TPP)Cl.

X = C1 8 = -9.01

0-Bz 5=-15.6

OMe 5 =-16.2

4-Me-Im 5 = -13.7

Figure 1.11. The family of oxoiron(IV) porphyrin cation radical species, Fe(IV)(0)(TMP)^XX) and the
characteristic proton NMR resonances of the pyrrole protons.
Models and Mechanisms of Cytochrome P450 Action 19

selectivity of the complex 13 as a function of the applicability for the epoxidation of unfimctional-
axial ligand^^^"^^^. By contrast, the reactivity and ized olefins. First described by Kochi^^^, this sys-
stereospecificity of the corresponding oxoiron(IV) tem has been particularly effective for the
porphyrin complexes were low^^' ^^^' ^^^. asymmetric epoxidation of prochiral olefins with
Nam has described studies using observed readily available complexes such as 11^^"^. Evi-
changes in product ratios and ^^0-labeling to dence for an oxomanganese(V) salen complex ^^^
suggest that both oxoferryl complexes such as and an oxoiron(IV) salen complex [0=Fe(IV)
13 and the Fe(III)-0-X precursors are reactive (salen)]'"*^(ref [126]) have been presented. The
oxidants^^' ^^^. The nature of the anionic ligand area has been thoroughly reviewed^^^' ^^^. The
was shown to affect both product selectivities and reader is also referred to the growing literature on
the efficiency of ^^O exchange. It is difficult, high-valent metallocorroles^^^~^^^.
however, to discern the cause of the changes The intermediacy of reactive oxomanganese(V)
observed, since the two-oxidant scenario proposed porphyrin complexes has long been implicated in
by Nam and the anionic ligand effect on the reac- olefin epoxidation and alkane hydroxylation
tivity of the oxoferryl complex itself described because of the distinct reactivity patterns and
by Gross, both would seem to explain the results. H2^^0-exchange behavior^ ^^~^^^, as compared to
The two most-well-characterized intermediates, that of the relatively stable oxomanganese(IV)
Fe(IV)(0)(por)+-(X), ("compound I") and porphyrin 14 intermediates which have been iso-
(por)Fe(IV)==0 ("compound 11") are known to lated and well characterized^ ^^' ^^^. The oxoman-
react with olefins to afford epoxides with different ganese(IV) porphyrin complex transferred oxygen
stereoselectivities. The former is known to pro- to olefins with little stereoselectivity, while a tran-
duce a high cisltrans ratio of epoxide from sient oxomanganese(V) complex underwent oxy-
cis olefins, while the latter gives mostly trans gen transfer to olefins with predominant retention
epoxide via a stepwise process. The effect of axial of configuration (Scheme 1.8)^^^' ^^^ Further, the
ligands would then be on the lifetime of [(por)Fe oxomanganese(IV) species exchanged the oxo lig-
(IV) =0]^* (high cisltrans epoxide ratio), which and with water slowly while the positively charged
easily decays to (por)Fe(IV)=0 (low cisltrans oxomanganese(V) complex readily exchanged the
epoxide ratio). Thus, while an iron(III)(por)- 0X0 ligand with added ^^O water^^'^.
peroxyacid complex has been demonstrated to be Oxometalloporphyrin studies in aqueous
reactive toward organic substrates such as olefins, media have allowed the study of reactive interme-
as discussed above, there is no unambiguous diates and metal-oxo-aqua interchange. The
evidence as yet from the model studies that a small peptide-porphyrin fragment, microperoxi-
hydroperoxoiron(III) porphyrin species, HOO- dase 8, has afforded evidence of reactive
Fe(III)(por), is a reactive, electrophilic oxidant. metal-oxo intermediates upon reaction with oxi-
dants such as hydrogen peroxide^"^^' ^^^. Important
insights into this oxo-hydroxo tautomerism were
7. Manganese Porphyrins in first reported by Meunier^^' i37-i39, 144 j ^ ^^g
Catalytic Oxidations shown in these studies that metal-oxo species are
able to transfer an oxygen atom originating from
Manganese porphyrins have been shown to either the oxygen source or from water. Because
have unusually high reactivity toward olefin epox- the intermolecular exchange of metal-oxo with
idation and alkane hydroxylation^^^"^^^. However, water is slow, an intramolecular exchange of
the physical characteristics of the putative oxo- labeled oxygen atoms occurred, which is reminis-
manganese(V) porphyrin species remained partic- cent of a carboxylic acid. This mechanism
ularly elusive^^' ^^^ because of its high reactivity involves a rapid, prototropic equilibrium, probably
and transient nature. Stable oxomanganese(V) via a fra«5-dioxoMn(V) intermediate^"*^, that
complexes are few, the only examples involving interconverts an 0x0 group on one face of the
the use of tetraanionic ligands to stabilize the metalloporphyrin with an aqua or hydroxo group
high-valent manganese center^ ^^~^^^. on the other face. This type of rearrangement
Structurally related to the porphyrins, was revealed by performing a catalytic oxygena-
manganese salen catalysts have shown wide tion catalyzed by a manganese porphyrin in
20 John T. Groves

pyridine

cis : trans = 0.5

pyridine
+ lO
Oxidant:
cis : trans = 9.82

NaOCl cis : trans = 32.0


Scheme 1.8. Reactivity and stereoselectivity of oxomanganese(IV) and oxomanganese(V) (generated by Mn(III)
and oxidants in situ) in olefin epoxidation.

^^0-labeled water but with a ^^O oxidant such a [Mn"^(TMPyP)] with a variety of oxidants, m-
Oxone or a peroxyacid. The oxo-hydroxy tau- CPBA, HSO~, and CIO", has been shown to
tomerism would allow as much as 50% water- produce the same, short-lived intermediate. An
derived ^^O oxygen transfer to a substrate, while oxoMn(V) porphyrin structure was assigned to this
the other 50% of the oxygen would derive from intermediate. The rate of formation of oxoMn(V)
the peroxide. from Mn"^(TMPyP) followed second-order kinetics,

?n' OH 0H2~1
I V ' O-X
OH2
I „,
—Mn^ . —Mn^— _
—Mn^— ^ —Mn"I—
O H
The first direct detection of an oxoman- first-order in Mn(III) porphyrin, and first-order in
ganese(V) porphyrin intermediate under ambient oxidant. The rate constants have the following
catalytic conditions was achieved by using rapid- order: m-CPBA (2.7 X 10^ M"^ s"*) > HSO5-
mixing stopped-flow techniques'*^' ^'*^. A direct (6.9 X lO^M-i s"^) ~C10- (6.3 X 10^ M"^ s^^).
assessment of its reactivity in both one-electron Once formed, the intermediate oxoMn(V) species
and oxygen-transfer processes was made possible was rapidly converted to oxoMn(IV) by one-elec-
by these observations. The reaction of tetra- tron reduction with a first-order rate constant of
A^-methyl-4-pyridylporphyrinatomanganese(III) 5.7 s"^ The oxoMn(IV) species was relatively
Models and Mechanisms of Cytochrome P450 Action 21

stable under the reaction conditions and was these conditions. With w-CPBA as the oxidant in
shown by EPR spectroscopy to have a high-spin the presence of H2^^0, the product epoxide was
quartet ground state. The one-electron reduction shown to contain 35% '^O, consistent with an
of oxoMn(V) to oxoMn(IV) was greatly 0-exchange-labile oxoMn(V) intermediate. Nitrite
accelerated by nitrite ion (A:=1.5X ion inhibited the epoxidation reaction competi-
10^M~^ s~^). However, the reaction between tively by one-electron reduction of the oxoMn(V)
nitrite and oxoMn(IV) is much slower. The intermediate to the unreactive oxoMn(IV). In
oxoMn(V) intermediate was shown to be highly this way, it was possible to show that the oxo-
reactive toward olefins, affording epoxide products. aqua exchange rate was about 10^ s~^ for the
In the presence of carbamazepine, a water-soluble coordinated water. Significantly, bulk water
olefin, this oxygen transfer was extraordinarily exchange was slower than oxo-transfer to the
rapid with a second-order rate constant of 6.5 X olefin under these conditions. This simple
10^M"^s~^). By contrast, oxoMn(IV) was not observation explains a body of often confusing
capable of effecting the same reaction under data and claims in the literature in this field.

NO2 NOi
OH

OH2

^3

O
OH OH
Mn" Mn"
i H2 ^«0H,

Scheme 1.9. (a) Rate constants: k^ = 2.7 X lO^M-^s 1; k^ = 1.5 X 10'7 M-i s-i; k^ = 6.5 X 10^ M-^ s"^;
k^ = 1.4 X 10^ M-^ s"^; ^^O exchange - l O ^ s - i (b) ^^O incorporation: 35% ^^O labeling from the solvent (95%
Hj^^O) into the epoxide.
22 John T. Groves

Peroxyacids will not exchange the peroxo oxygen toward electron transfer, hydrogen atom abstrac-
with water at all and neither does the pro- tion, and epoxidation were all several orders
duct epoxide. Thus, in those cases for which of magnitude slower than the corresponding
^^0-incorporation is observed into the product 4-iV-methylpyridyl species (Scheme 1.10). This
epoxides or alcohols, an oxo-metal species in unusual effect, which did not appear in the corre-
strongly implicated as the oxygen donor. sponding reactions of the Mn(IV) and Mn(III)
However, failure to see ^^O-exchange is not a states, was ascribed to the low spin, d^ electronic
definitive result since the exchange rate may configuration of oxoMn(V). Thus, a spin-state
simply be too slow to compete with oxygen-atom crossover is required during reduction with the
transfer. These transformations and the ^^O- promotion of an electron from d to d^^ .
exchange process are summarized in Scheme 1.9. Oxomanganese(V)-5,10,15,20-tetrakis(A^-
It has been shown that hydrogen peroxide is also methyl-2-pyridyl)-porphyrin (15) was found to
an effective oxidant of water-soluble manganese transfer its oxo ligand efficiently to the bromide
porphyrins, affording a reactive oxoMn(V) inter- ion. Furthermore, this oxo transfer is rapid and
mediate as well^"*^. With water-soluble iron por- reversible^^^. The forward reaction mimics the
phyrins, hydrogen peroxide was able to epoxidize halide oxidation reaction catalyzed by haloperoxi-
olefins ^'*^. Significantly, the efficiency of epoxi- dases, while the reverse reaction is the catalyst
dation dropped drastically above pH 5, suggesting activation step in substrate oxygenation by man-
that an acid-catalyzed heterolytic 0 - 0 bond ganese porphyrins. This well-behaved equilibrium
cleavage is part of the oxygen-transfer process. has allowed the assignment of a free energy
Surprisingly, the 2-A^-methyl pyridyl isomer change for this reaction and is the first clear deter-
oxoMn(V)(TM-2-PyP) was found to be unusually mination of the thermodynamics of an oxo trans-
stable allowing its characterization by ^H-NMR"^^. fer reaction for a metal catalyst of this type. As
Reaction rate constants for oxoMn(V)(TM-2-PyP) can be seen in the Nernst plot in Figure 1.12, the

electron transfer Second Order Rate Constants (M l .s- l ^)


o
NO2 1.5x10' 2.0 X10"

hydrogen atom abstraction

3.5 xlO* 8.8 x 10^

atom transfer
R—N 6.5 xlO^ 10^
Mn^

S c h e m e 1.10. Reaction rates for OxoMn(V)TM-4-PyP and OxoMnT(Y)M-2-PyP.


Models and Mechanisms of Cytochrome P450 Action 23

Br

BrO

1.6
- cio-/cr
-A— BrO~/Br
Mn(V)/Mn(lll)
-m— H2O2/H2O
1,4

1.2

0.8

0.6
10 12 14
PH

Figure 1.12. Reversible oxygen transfer from hypobromite provided an energy calibration for oxomanganese(V)
porphyrins.

oxoMn(V) species is able to oxygenate bromide for a transient biological oxidant, perox5niitrite
ion below pH 8 and chloride ion below pH 5, the (ONOO~). ONOO~, a strong one- and two-
increase in oxidizing power being due to the electron oxidant, has been implicated in a
protonation of the oxoMn(V) intermediate. As can number of pathological conditions^^^. Structurally,
be seen, this species is tantalizingly close to the ONOO~ is the conjugate base of peroxynitrous
oxidation potential of water/hydrogen peroxide at acid and, like other similar peroxides, it rapidly
low pH. oxidizes manganese porphyrins^^^' ^^^' ^^^ Thus, its
reactions with manganese porphyrins to generate
oxomanganese intermediates allowed the sensitive
detection of this transient molecule under cell-like
8. Metalloporphyrins as conditions^^^' ^^^. Using manganese porphyrins as
Detectors and Decomposition ONOO~ detectors, it has been shown that 0 N 0 0 ~
Catalysts of Peroxynitrite can quickly diffuse across biological membranes
and react with its biological targets ^^^' ^^^.
The rapid reaction rates of peroxides with met- The fast oxidation of manganese(III) por-
alloporphyrins and the detection of observable phyrins to oxomanganese(IV) species by ONOO~
oxometalloporphyrin species, especially in the satisfied the prerequisite for the porphyrins to
reactions of manganese porphyrins with various be efficient ONOO~ decomposition catalysts ^^^.
oxidants (m-CPBA, NaOCl, KHS05)^^^ have also Indeed, manganese porphyrins, redox-coupled
inspired the use of these porphyrins as detectors with biological antioxidants (such as ascorbate,
24 John T. Groves

glutathione, Trolox®), rapidly reduce ONOO" and that water-soluble iron porphyrins have the ability
thus prevent the oxidation and nitration of biolog- to decompose ONOO" by isomerizing it to nitrate
ical substrates by this toxic oxidant^^^' ^^^. and these compounds, moreover, have shown sig-
Amphiphilic analogs of the water-soluble metal- nificant biological responses in animals ^^^. The
loporphyrins have been developed that are mechanism of action of the iron porphyrins
suitable for liposomal delivery^^^. Further, these is significantly different than the manganese
amphiphilic metalloporphyrins in sterically stabi- case. Significantly, both Fe(III) and oxoFe(IV)
lized liposomes are highly active in decomposing states are catalytically active toward ONOO~
0 N 0 0 ~ (ref [154]), and thus are potential thera- (Scheme 1.11)^^^'^^^. Compounds of this sort have
peutic agents for the treatment of ONOO~-related been shown to be highly potent in animal models
diseases. Iron(III) porphyrins are also rapidly oxi- of inflammation related pathologies such as
dized by ONOO", and Stem et al. have reported diabetes and colitis*^^.

ONpO" NO3
8

ONOO

ONOO" NO3

HpO

N(Oo :=;=^N204 ^ • NO33 + NO2


N O i +2H

ONOO" • NOi

Scheme 1.11. Mechanism of peroxynitrite decomposition by FeTMPS.


Models and Mechanisms of Cytochrome P450 Action 25

9. Synthetic Metalloporphyrins occurred only at carbon 6 to give the 6a-hydroxy-


as Stereoselective Catalysts steroid, even though there are many sites in this
molecule with similar intrinsic reactivity. This
high selectivity was found to depend critically
Understanding the mechanism of cytochrome
on the precise arrangement of the aromatic
P450 through the study of the synthetic model
groups of the substrate. Moreover, the manganese
systems offers an opportunity to develop practical
porphyrin-cyclodextrin construct was able to
regioselective and stereoselective catalysts. Such
release the product sterol and hydroxylate at least
catalytic systems have been extensively surveyed
four steroid molecules. Catalytic turnover with
in numerous reviews^^' ^^' ^^' i59-i62 Sp^ce does
such high positional selectivity is highly unusual
not permit a discussion of all the elegant catalysts
for this kind of model system ^^^.
developed, but a few systems are shown here as
examples. Collman et al. reported a series of "picket
basket" porphyrins (Figure 1.14) which show
By analogy to the natural enzymes which
extremely high shape selectivity (> 1,000 for
utilize a protein scaffold to effect substrate recog-
c/5-2-octene vs c/^-cyclooctene) and relatively
nition and stereoselectivity, special steric features
high enantioselectivity in olefin epoxidation
have been introduced into the synthetic porphyrin
{ee around lQ-%5%f^^^ ^^\ Chiral, binaphthyl
catalysts to achieve regio- and enantioselective
straps between adjacent o-aminophenyl groups
oxidation. The success of such systems relies on
have afforded very good enantiomeric selectivities
the steric interactions between the substrates and
for styrene epoxidations^^^.
porphyrin catalysts, which position the substrates
specifically toward the reactive metal-oxo center. The first use of a chiral porphj^in to carry
Breslow et al}^^' ^^^ have recently reported a out asymmetric epoxidation was reported in
remarkably selective, catal3^ic steroid hydroxyla- 1983, giving 50% ee with /?-chlorostyrene^^^.
tion using an artificial cytochrome P450 enzyme. The ee was improved to —70% for epoxidation
The synthetic strategy to induce selectivity in the of cw-p-methylstyrene with the use of a very
model system was the attachment of four cyclo- robust chiral vaulted binaphthyl porphyrin 16
dextrin appendages to a synthetic manganese por- (Figure 1.15)^^^' ^^^. More significantly, this cata-
phyrin used in place of the heme center of the lyst afforded the first case of catalytic asynmietric
natural enzyme. These donut shaped heptamylose hydroxylation by a model system, giving a ~70%
sugars have a hydrophobic central cavity, which is ee for hydroxylation of ethylbenzene and related
known to bind aromatic molecules. The substrate hydrocarbons ^^^' ^^^.
steroid was modified with such an aromatic group Further developments of this system have led
at either end of the molecule. The host-guest to studies of the binaphthyl-peptide-strapped por-
complex obtained from these designed partners, phyrin 11^^^. For styrene epoxidation this chiral
Figure 1.13, displays a limited region of the sub- catalyst afforded an ee greater than 90% in the ini-
strate steroid in the vicinity of the catal5^ic man- tial stages of the reaction, with the (i?)-styrene
ganese center. What is most significant about this oxide predominating. NMR T^ relaxation studies
artificial enzyme is the fact that hydroxylation with the copper(II) derivative of the same ligand

Figure 1.13. Steroid-manganese porphyrin host-guest complex.


26 John T. Groves

R = -(CH2)n- n = 2, 4, 6, 8, 10

-CH CH,

-CH2CH2O
OCH2CH2-
(CH3)3C C(CH3)3

Figure 1.14. Chiral "picnic basket" porphyrins.

HsCO^^OCH been described by Naruta^^^' ^^^. In this "twin


coronet" structural motif, four pendant hydroxy-
naphthyl groups provide a sterically encumbered,
hydrophobic substrate-binding cavity (Figure 1.17).
Upon treatment with m-chloroperoxybenzoic acid
an oxoiron(IV)-naphthyloxy radical species was
formed and detected by EPR. This radical interme-
diate reacted with a 1,4-diene and oxygen in
a single-turnover reaction to afford 5-trans-l-cis-
undeca-5,7-diene-4-ol in 156% yield. The forma-
tion of the aryloxy radical was shown to be
essential to this conversion in close analogy to the
enzymatic process.

10. Ruthenium Porphyrins in


Figure 1.15. A chiral vaulted binaphthyl metallo-
porphyrin. Oxidative Catalysis

The controlled oxygenation of alkanes, alkenes,


showed that it was the protons of the major and aromatic hydrocarbons is one of the most
enantiomer of the product epoxide that were more important technologies for the conversion of crude
efficiently relaxed in an inversion-recovery exper- oil and natural gas to valuable commodity chemi-
iment with Cu-17. Comparative T, data are shown cals^ ^'^. Biomimetic studies of metalloporphyrins
in Figure 1.16. Accordingly, the major product have led to important advances in practical cataly-
must fit better into the chiral cavity of this cata- sis, especially with ruthenium porphyrins. Reaction
lyst. Thus, one can conclude that the enantio- of m-CPBA, periodate, or iodosylbenzene with
selectivity observed can be attributed to a simple Ru(II)(TMP)(CO) produced Ru(VI)(TMP)(0)2^7^.
lock-and-key process in which the transition state Remarkably Ru(VI)(TMP)(0)2 was found to cat-
for epoxidation must look rather like the coordi- alyze the aerobic epoxidation of olefins under mild
nated epoxide. conditions ^^^. Thus, for a number of olefins includ-
An interesting functional active-site model ing cyclooctene, norbomene, cis-, and trans-^-
system for prostaglandin H synthase has recently methyl styrene 16-45 equivalents of epoxide were
Models and Mechanisms of Cytochrome P450 Action 27

(S)-styrene oxide (/?)-styrene oxide


0.012-
0.01- 1/T l p , s -
0.008-
0.006-
0.004-
0.002H
a iM itiiiiii

-0.002-
Hb

Li-Fe(lll)CI
^-

initial ee of f/?j-styrene oxide > 90% V N H ° HN H


0=C 'Me K/,.^.C^O
Me*^
\ /
Figure 1.16. NMR T. relaxation data compared to stereoselectivity.

-OH HO- -O* HO-


mCPBA
(^ Fe ^ (^ Fe

—OH HO- —OH HO-

Figure 1.17. Twin coronet model prostaglandin H synthase.

produced per equivalent of the ruthenium X-ray data obtained for a closely related
porphyrin complex over a 24-hr period at room Ru(VI)(TDCPP)(0)2, definitively established a
temperature under 1 atm of dioxygen. structural precedent for the /ra«5-dioxoRu(VI) por-
Dioxygen activation by Ru(II) porphyrins is phyrin complexes^^^. The structure revealed a nearly
likely to involve the formation of a jui-peroxo- planar arrangement of the porphyrin macrocycle
ruthenium(III) dimer followed by homolysis of the and two relatively long Ru=0 bonds of 1.729 A
0 - 0 bond to give two equivalent of oxoruthe- (Figure 1.18). Marchon and coworkers have pub-
nium(IV) species. Nakamoto et alP^^ ^^^ detected lished an X-ray structure of /ra«5-dihydroxoruthe-
such a iLjL-peroxoruthenium(III) intermediate by nium(rV) complex of H2TDCPP^^^, also obtained
resonance Raman spectroscopy (Vg(0=Ru) at from the reaction of Ru(II)(TDCPP)(CO) with
552 cm~i) in the toluene solution of Ru(II)(TPP) m-CPBA. The IR spectrum of the ^an^-dihydroxo-
saturated with O2 at — 80°C. Upon raising the ruthenium(IV) complex was reported to be differ-
temperature of the solution a Vg(0=Ru=0) band ent from that of the /ra«5-dioxoruthenium(VI)
corresponding to Ru(VI)(TMP)(0)2 appeared at complex. Thus, Ru(IV)(TDCPP)(0H)2 showed
811 cm-^ a Ru-O stretch at 760 cm"^ in the IR spectrum
28 John T. Groves

C1I4)

C120)

Figure 1.18. Molecular structure of Ru(VI)(TDCPP)(0)2

while the R u = 0 band of Ru(VI)(TDCPP)(0)2 Ru(VI)(TPP)(0)2/02 system, the electron-rich


appeared at 828 cm~^ The length of the R u - 0 styrenes gave more /?ara-substituted phenyl-
bonds in the X-ray structure of Ru''^(TDCPP) acetaldehyde byproducts than the electron-poor
(OH)2 was 1.790 A which is somewhat longer but styrenes. For example, 19.5 equiv of the aldehyde
not significantly different from the length of the and 3.8 equiv of the epoxide was produced in
R u = 0 bonds in Ru(VI)(TDCPP)(0)2^^^. the oxygenation of methoxystyrene while the
Nonlinear, upward U-shaped Hammett plots epoxidation of chlorostyrene produced less than
were obtained for the stoichiometric oxidation of 1 equiv of the aldehyde and 37 equiv of the
substituted styrenes with Ru(VI)(OEP)(0)2 and chlorostyrene oxide based on catalyst. In order
Ru(VI)(TPP)(0)2. The shape of the Hammett cor- to explain these data, a mechanism has been pro-
relations indicated a change in the structure of the posed (Scheme 1.12) in which the rate-limiting-
transition state of epoxidation with these com- step of the initial partial charge transfer is
plexes when switching from electron-donating to separated from the product-forming step. Phenyl-
electron-withdrawing substituents^^^ However, acetaldehydes are believed to be formed by a
the Hammett treatment of data for a series of com- 1,2-hydride shift of the cationic intermediate. This
petitive aerobic oxidations of /7ara-substituted intermediate would be more favored by electron-
styrenes catalyzed by Ru(VI)(TMP)(0)2 gave a donating /?<2ra-substituents in agreement with the
good linear correlation with a p^ value of -0.93 distribution of products.
against a^ set of parameters ^^^. Only para- Murahashi and coworkers have studied the
nitrostyrene deviated from the linear trend. This aerobic oxidation of alkanes catalyzed by poly-
p^ value was typical of the results obtained for fluorinated metalloporphyrins including Ru^^
iron and manganese-porphyrin-mediated epoxida- (TPFPP)(CO) in the presence of acetaldehyde
tions with iodosyl arenes and hypochlorite, (Scheme 1.13)18'*'^^^
suggesting a similar, relatively small charge sepa- The catalytic oxygenation of cyclohexane and
ration in the transition states of these reactions^^' adamantane at 70°C under 1 atm dioxygen gave
166,182,183 Significantly, in epoxidations with the c. 200 turnovers in 24 hr. The ruthenium porph5Tin
Models and Mechanisms of Cytochrome P450 Action 29

^TT X

RDS

X O

Ruiv )TMP

(X=C1,H,CH3) +
X

TMP
^
o

(X = 0CH3)

X O

Ru^^ TMP

1,2-H-shift

I
^
Ru^v TMP

O
Scheme 1.12. Proposed mechanism of oxygen-atom transfer from Ru(VI)(TMP)(0)2 to substituted styrenes.

I Ru"(TPFPP)(CO) I
R2 Cy H ^^ R2 C~
I CH3CHO, 1 atm O2 I
R3 70°C R3

Ru(TPFPP)

S c h e m e 1.13. Aerobic oxidation catalyzed by Ru(II)(TPFPP)CO, see references 184 and 185.
30 John T. Groves

demonstrated high stability toward oxidative oxygenations and therefore an unlikely intermedi-
degradation with turnover numbers as high as ate in the catalytic cycle. The authors noted that
14,000, achieved in the oxygenation of cyclo- the distribution of products in a cyclohexene oxy-
hexane when a low catalyst concentration was genation with Ru"(TPFPPCl8)(CO) was identical
used. The normalized 372° C-H bond preference to the ratio of allylic oxidation vs epoxidation
in oxidation of adamantane with this system was products obtained in the aerobic oxidation with
about 20. Competitive experiments with cyclo- Fe"i(TPFPPBr8)Cli^l For the iron porphyrin
hexane and cyclohexane-(i^^ resulted in an isotope catalyst these reactions have been established to
effect of 13. The mechanism of catalysis was not follow a radical autoxidation mechanism^ ^^' ^^^.
clear. It is known that the reaction of aldehydes Nitrous oxide is produced as a byproduct in
with dioxygen in the presence of metal complexes multimillion lb/year quantities in nylon manu-
produces peroxyacids^^^. The authors suggested facture worldwide. Currently, there is a great
an oxometal species which is formed by reaction interest toward the utilization of N2O due to the
of peroxyacid with ruthenium porphyrin as the environmentally hazardous nature of this gas
active intermediate although no good evidence in with respect to the greenhouse effect and ozone
favor of such species was presented. The possibil- layer depletion. In addition to their ability to
ity of a radical autoxidation mechanism cannot be utilize dioxygen for catalytic hydrocarbon oxida-
excluded. tions, ruthenium porphyrins have been shown to
Labinger and coworkers^^^ have investigated activate nitrous oxide ^^^ which is an extremely
the catalysis of aerobic olefin oxidation by a inert molecule'^' and a poor ligand.'^^^ Groves
highly electron-deficient perhalogenated ruthe- and Roman have found that N2O reacted with
nium porphyrin complex Ru"(TPFPPClg)(CO). Ru"(TMP)(THF)2 in toluene to produce Ru^^
The oxidation of cyclohexene under 1 atm dioxy- (TMP)(0)2^^^. The /ra«5-dioxoRu(VI) complex
gen at room temperature proceeded mostly in the can in turn epoxidize a suitable substrate such as
allylic positions affording 58% 2-cyclohexen-l-ol fra«5-p-methyl styrene. This system was subse-
and 27% 2-cyclohexen-l-one along with only quently shown to be catalytic under appropriate
15% of cyclohexene oxide. Three hundred conditions ^^^''.
turnovers of oxidation were achieved in 24 hr and Hirobe and coworkers were the first to use
90% of the catalyst was recovered at the end of the an unusual class of oxidants, 2,6-disubstituted
reaction. Oxygenation of styrene gave benzalde- pyridine TV-oxides, in conjunction with ruthenium
hyde (3 turnovers in 24 hr) as the only product. By porphyrins for oxidation of olefins (Scheme 1.14),
contrast, the oxidation of cyclooctene produced alcohols, and sulfides^^^~^^^.
cyclooctene oxide as the major product (42 The epoxidation of styrene with 2,6-
turnovers in 24 hr). With iodosylbenzene as oxi- dichloropyridine N-oxidQ was efficiently catalyzed
dant Ru"(TPFPPClg)(CO) demonstrated signifi- by of Ru(VI)(TMP)(0)2 or Ru(II)(TMP)(CO) to
cantly higher selectivity for epoxidation with 81%) afford styrene oxide with high selectivity and
styrene oxide and 42% cyclohexene oxide pro- nearly quantitative yields based on substrate
duced, based on the total amounts of oxidation used'^^. Moderate yields of the epoxide (26%)
products. The /ra«5-dioxoruthenium(VI) complex were obtained with Ru(II)(TPP)(CO). Turnover
Ru^i(TPFPPCl8)(0)2 prepared by oxidation of numbers up to 16,500 were achieved in epoxidation
Ru"(TPFPPCl8)(CO) with m-CPBA was found of norbomene in the presence of catalytic amoxmts
to be a less efficient catalyst for the aerobic of Ru(VI)(TMP)(0)2. The catalytic reactions were

Ri R2 Ru(Por) ^\ /^2 ^

R4 Ri' N R2' Ar, benzene R30 R4 Ri' ^ R

S c h e m e 1.14. RuPor = Ru(II)(TMP)(CO), Ru(II)(TPP)(CO) or Ru(VI)(TMP)(0)2


Models and Mechanisms of Cytochrome P450 Action 31

characterized by high chemoselectivity and stereo- catalyzed by Ru(II)(TPP)(CO), Ru(II)(TMP)


specificity. For example, when a 1:1 mixture of (CO), and Ru(II)(TDCPP)(CO) in the presence of
cis- and trans-sXiVoQnQ was reacted with 2,6-luti-jjQj.200 Xertiary C-H positions of steroidal sub-
dine iV-oxide in the presence of Ru(VI)(TMP)(0)2, strates were hydroxylated with complete retention
cw-stilbene oxide was produced in 87% yield, of configuration of the asymmetric centers. The
while only 1% of ^raw^'-stilbene was epoxidized^^^. regioselectivity of oxidation significantly depended
Further, the cis disubstituted double bond of on the steric bulk of the ortho substituents of the
trans, cis, trans-1,5,9-cyclododecatriene was pre- aromatic rings at the meso position of the porphyrin
dominantly oxidized under these conditions. catalysts and on the electronic properties of the
C/5-stilbene was stereospecifically epoxidized porphyrins.
with 2,4,6-trimethylpyridine. Only ruthenium por- A very promising system for hydrocarbon oxy-
phyrins displayed catalytic activity with respect to genation with 2,6-dichloropyridine TV-oxide in the
the oxygen transfer from pyridine A^-oxides. Other presence of electron-deficient, polyhalogenated
metalloporphyrins, such as Mn, Fe, Co, Mo, and ruthenium porphyrins has been reported^^^' ^^^.
Rh porphyrin complexes, were found to be inactive The oxidations with this system were character-
with these oxidants^^^' ^^^. ized by high rates and selectivity even under
While alkanes and aliphatic alcohols were mild conditions and, most significantly, could be
inert with respect to the RuPor/A^-oxide system, carried out in a nonacidic reaction media.
the catalytic activity of ruthenium porphyrins was Carbonyl (5,10,15,20-tetrapentafluorophenylpor-
dramatically enhanced in the presence of protic phyrinato) ruthenium(II)--Ru(II)(TPFPP)(CO)
acids such as HBr^^^' ^^^' ^^^. Hydroxylation of showed unusually high activity with 2,6-
adamantane with 2,6-dichloropyridine A/-oxide in dichloropyridine A/-oxide as the oxygen donor
the presence of Ru(II)(TMP)(CO) and HBr (Scheme 1.15).
afforded over 100,000 turnovers of the tertiary Adamantane and c/5-decalin were hydroxylated
C-H oxidation products, 1-adamantanol and 1,3- with high selectivity, complete stereo-retention,
adamantanediol, with a turnover frequency of extraordinarily high rates (up to 800 turnovers
5.6 turnovers s~^ Methylcyclohexane was selec- min~^), and high efficiency with up to 15,000
tively hydroxylated to give 1-methylcyclohexanol turnovers. Similar conversions were obtained when
in 94% yield. C/5-9-decalol was the major pro- Ru(VI)(TPFPP)(0)2 and Ru(VI)(TPFPBr8P)(0)2
duct in oxidation of c/^-decalin with no stereoiso- were used as catalysts. Oxygenation of less reac-
mer ^ra«5'-9-decalol detected. Oxygenation of a tive substrates such as benzene and cyclohexane
relatively inactive alkane such as cyclooctanol proceeded with lower but still significant turnover
afforded c. 300 turnovers of cyclooctanone (77% numbers (100-3,000). Tertiary vs secondary selec-
yield based on substrate) ^^^. tivity in adamantane oxidation was above 210.
Nagano et al. have studied the oxidation No rearrangement products were detected in
of steroids with 2,6-dichloropyridine iV-oxide c/5-decalin hydroxylation.

CO
RF )TPFPP
^ R—OH + II
Cl^N'^Cl
\
CO O
RT )TPFPP ^

Scheme 1.15. Fast catalytic hydroxylation with a fluorophenylmthenium porphyrin.


32 John T. Groves

The kinetics of product evolution in a typical and even higher maximum turnover rate of
reaction of adamantane hydroxylation showed 4.9 turnovers min~^ in adamantane hydroxylation
an initial induction period followed by a fast, vs 4.0 turnovers min~i for Ru(VI)(TPFPP)(0)2
apparently zero-order phase with the maximum under the same conditions indicates that an active
rate and highest efficiencies (Figure 1.19). catalyst species other than Ru(VI)(TPFPP)(0)2 is
Deviation from linear behavior took place only involved in the fast catalytic hydroxylation.
after 90% of the oxygen donor and 80% of the Therefore, a tj^ical fra«5-dioxoRu(VI)-
substrate had been consumed. When Ru(VI) oxoRu(IV) catalytic cycle can be ruled out as the
(TPFPP)(0)2, prepared by reaction of Ru(II) primary reaction pathway in case of rapid catalytic
(TPFPP)(CO) with 3-chloroperoxybenzoic acid oxygenation. The apparent zero-order kinetics
was used as the catalyst, no induction time was observed are consistent with a steady-state cat-
detected and zero-order kinetics were observed as alytic regime accessible from different initial
well. The well-defined and characteristic UV-vis states of ruthenium metalloporphyrin. Indeed,
spectra of metalloporphyrins provide an invalu- common oxidants, other than aromatic A^-oxides,
able tool for the mechanistic studies. Thus, moni- such as iodosylbenzene, magnesium monoperoxy-
toring the state of the metalloporphyrin catalysts phthalate, Oxone®, and tetrabutylammonium
during the course of both model reactions by periodate produced the fran5-dioxoRu(VI) species
UV-vis spectroscopy revealed that the initial form from Ru(II)(TPFPP)(CO) under reaction condi-
of the catalyst remained the predominant one tions but were ineffective for the rapid catalysis.
throughout the oxidation, that is, in the Ru(II) A two-electron oxidation of Ru(II)(TPFPP)
(TPFPP)(CO)-catalyzed reaction c. 80% of the (CO) would produce oxoRu(IV) porphyrin and
porphyrin catalyst existed as Ru(II) (TPFPP)(CO) eventually dioxoRu(VI). What is the alterna-
and in Ru(VI)(TPFPP)(0)2-catalyzed reaction tive pathway for Ru"(TPFPP)(CO) activation?
more than 90% of the catalyst was still in the It is known that ruthenium(II) ir-cation radicals
form of Ru(VI)(TPFPP)(0)2 despite the high are formed from the corresponding carbonyl
turnover numbers reached (—400 turnovers). The compounds by chemical or electrochemical one-
fact that Ru(II)(TPFPP)(CO) demonstrates similar electron oxidation^^^. Such species have been

400

300 H
)TPFPP

TO O

200H

100 H

200

Figure 1.19. Adamantane hydroxylation catalyzed by Ru(II)(TPFPP)(CO) and Ru(VI)(TPFPP)(0)2,


[adamantane] = [pyCl2N0] = 0.02 M, [catalyst] = 50 JJLM, CH2CI2, 40°C. TO (Turnovers) = moles of
product/moles of catalyst.
Models and Mechanisms of Cytochrome P450 Action 33

shown to undergo intramolecular electron transfer an attractive foundation for the design of the new
upon axial ligation and removal of CO to give chiral epoxidation catalysts due to their high
ruthenium(III) porphyrins^^"^. An emerald green intrinsic stability with respect to oxidative degra-
solution of Ru(II)(TPFPP)(CO)+* radical cation dation. Thus, extremely large turnover numbers in
with a strong EPR signal (g = 2.00) was quantita- the oxygenation of hydrocarbons, approaching
tively obtained when Ru(II)(TPFPP)(CO) was oxi- 10^-10^, have been achieved with achiral metallo-
dized with ferric perchlorate in methylene porphyrin catalysts^^^' ^^^.
chloride. An EPR signal typical of a ruthenium(III) Gross et al. reported the first use of a chiral
species (gn = 2.55, g^ = 2.05) was detected after ruthenium porphyrin Ru"(Lj)(CO) as a catalyst
the addition of 2,6-lutidine A^-oxide to the solution for st5Tene epoxidation^^^. Chiral ruthenium por-
of the radical cation. phyrin systems have also been reported by Che
Metastable Ru(III) and oxoRu(V) species have et al?^^' ^^^. The utilization of another chiral ruthe-
been proposed as key intermediates in the cat- nium porphyrin, Ru"(L2)(C0) as a catalyst for
alytic cycle of "rapid oxygenation" which can be enantioselective epoxidation of olefins with 2,6-
viewed as a part of the general scheme of diverse dichloropyridine TV-oxide has been described by
oxidative chemistry of ruthenium porphyrins Berkessel and Frauenkron^^^. The highest enan-
(Scheme 1.16). The aerobic oxygenation pathway, tiomeric excesses of the oxiranes were obtained in
involving the even oxidation states, is shown in the epoxidation of tetrahydronaphthalene and
the left half of Scheme 1.16. The new fast catalytic styrene, 77% and 70% ee, respectively, with high
process on the right half of the figure depicts the yields (up to 88%). Terminal aliphatic olefins
chemical interconnectivity between the fast and and ^ra«5-disubstituted olefins, represented by
slow catalytic regimes. 1-octene and tmns-stilhQne, were sluggish sub-
Enantioselective epoxidation of unfiinctional- strates and gave low ee's. The epoxidation of
ized olefins is an important area of asymmetric tetrahydronaphthalene with iodosylbenzene cat-
synthesis^^^' ^^^' ^^^. Metalloporphyrins represent alyzed by Ru(II)(L2)(CO) produced only 52% ee

S - substrate
OD - oxygen donor

Scheme 1.16. Mechanisms of ruthenium porphyrin oxidation catalysis.


34 John T. Groves

of the epoxide. Interestingly, the results were sim- inspirations, and insights. Their names are
ilar to those published by Gross et al. ^^^ in that the indicated in the referenced papers. Thanks go
enantioselectivity of epoxidation with Ru(II)(L2) also to the many colleagues around the world
(CO) was different when iodosylbenzene or pyri- who have brought such energy and excitement to
dine TV-oxides were used as primary oxidants but the P450 and metalloporphyrin fields for nearly
did not depend on the nature of the pyridine A^- three decades. Research in our laboratories
oxide. Under the comparable reaction conditions was supported by the National Institutes of
—72% ee of tetrahydronaphthalene oxide was Health (NIGMS) and the National Science
reported for the catalytic oxidation of tetrahydron- Foundation. I also thank the National Science
aphthalene with 2,6-dichloropyridine A^-oxide, Foundation and the Department of Energy for
2,6-dibromopyridine A/-oxide, and 7V-methylmor- their support of the Environmental Molecular
pholine A^-oxide. Science Institute (CEBIC) at Princeton University.

11. Conclusion References

Cytochrome P450 has been called the Rosetta 1. Hayaishi, O., M. Katagiri, and S. Rothberg (1955).
Stone of iron proteins. Perhaps nowhere else in the Mechanism of the pyrochatecase reaction. J. Am.
biological sciences has the rich interplay between Chem.Soc. 17, 5450-5451.
structural, spectroscopic, mechanistic, computa- 2. Tchen, T.T. and K. Block (1956). On the mechanism
tional, and chemical modeling techniques led to of cyclization of squalene. J. Am. Chem. Soc. 78,
1516-1517.
such a detailed level of understanding of such an
3. Ortiz de Montellano, P.R. and J.J. De Voss (2002).
important system. The central paradigm of biolog- Oxidizing species in the mechanism of cytochrome
ical oxygen activation is now recognized to P450. Nat. Prod. Rep. 19, 477^93.
involve the formation a ferry 1, or oxoiron interme- 4. Sundaramoorty, M., J. Terner, and T.L. Poulos
diate. Oxoiron(IV) porphyrin cation radicals have (1995). The crystal structure of chloroperoxidase:
been observed in peroxidase, cytochrome oxidase, A heme peroxidase-cytochrome P450 functional
CPO, cytochrome P450, and in a variety of model hybrid. Structure 3, 1367-1377.
systems. Model system studies, especially those 5. Manoj, K.M. and L.P Hager (2001). Utilization of
of iron, manganese, and ruthenium porphyrins peroxide and its relevance in oxygen insertion reac-
and related ligands, have led to important tions catalyzed by chloroperoxidase. Biochim.
Biophy. Acta 1547, 408^17.
advances in catalysis and in catalytic asymmetric
6. (a) Groves, J.T. and C.C.-Y. Wang (2000). Nitric
oxygenation. Advances in computational studies oxide synthase: Models and mechanisms. Curr
of such complex, open-shell systems have begun Opin. Chem. Biol. 4, 687-695; (b) Mansuy, D. and
to provide a rigorous physical underpinning for J.L. Boucher (2002). Oxidation of N-hydroxyguani-
the body of complex and sometimes confusing dines by cytochromes P450 and NO-synthases and
experimental results. In this chapter, I have tried to formation of nitric oxide, Drug Metab. Rev. 34(3),
weave together all of these aspects to provide for 593-606.
the reader a unified picture of the current under- 7. Santhanam, L. and J.S. Dordick (2002).
standing in the field of cytochrome P450 research. Chloroperoxidase-catalyzed epoxidation of styrene
More detailed presentations are to be found in the in aqueous and nonaqueous media. Biocatal.
chapters that follow. Biotransform. 20, 265-274.
8. Rantwijk, F. and R.A. Sheldon (2000). Selective
oxygen transfer catalysed by peroxidases: Synthetic
and mechanistic aspects. Curr Opin. Biotech. 11,
Acknowledgments 554-564.
9. Van Beilen, J.B. and Z. Li (2002). Enzyme technol-
ogy: An overview. Curr Opin. Biotech. 13, 338-344.
Special thanks are due to all the members 10. Glieder, A., E.T. Farinas, and F.H. Arnold (2002).
of my research group and my collaborators who Laboratory evolution of a soluble, self-sufficient,
participated in the projects described here from highly active alkane hydroxylase. Nat. Biotechnol.
our laboratory and contributed so many ideas, 20,1135-1139.
Models and Mechanisms of Cytochrome P450 Action 35

11. Crane, B.R., A.S. Arvai, S. Ghosh, E.D. Getzoff, 24. Groves, J.T, R.C. Haushalter, M. Nakamura,
D.J. Stuehr, and J.A. Tainer (2000). Structures T.E. Nemo, and B.J. Evans (1981). High-valent iron-
of the N-omega-hydroxy-L-arginine complex of porph3Tin complexes related to peroxidase and
inducible nitric oxide synthase oxygenase dimer cytochrome P-450. J ^m. Chem. Soc. 102,2884-2886.
with active and inactive pterins. Biochem. 39, 25. Groves, J.T. and G.A. McClusky (1976). Aliphatic
4608-4621. hydroxylation via oxygen rebound. Oxygen transfer
12. Raman, C.S., H.Y. Li, R Martasek, G. Southan, catalyzed by iron. J. Am. Chem. Soc. 98, 859.
B.S.S. Masters, and T.L. Poulos (2001). Crystal 26. Schunemann, V, C. Jung, J. Terner, A.X. Trautwein,
structure of nitric oxide synthase bound to nitro and R. Weiss (2002). Spectroscopic studies of per-
imdazole reveals a novel inactivation mechanism. oxyacetic acid reaction intermediates of cytochrome
Biochemistry 4{S, 13448-13455. P450cam and chloroperoxidase. J. Inorg. Biochem.
13. Li, H.Y., C.S. Raman, R Martasek, B.S.S. Masters, 91, 586-596.
and T.L. Poulos (2001). Crystallographic studies on 27. Kellner, D.G., S.C. Hung, K.E. Weiss, and
endothelial nitric oxide synthase complexed with S.G. Sligar (2002). Kinetic characterization of com-
nitric oxide and mechanism-based inhibitors. pound I formation in the thermostable cytochrome
Biochemistry 40, 5399-5406. P450 Cypl 19. J. Biol. Chem. 277, 9641-9644.
14. Fujii, H. (2002). Electronic structure and reactivity 28. Gelb, M.H., D.C. Heimbrook, R Malkonen, and
of high-valent oxo iron porphyrins. Coord. Chem. S.G. SHgar (1982). Stereochemistry and deuterium
Rev. 226, 51-60. isotope effects in camphor hydroxylation by the
15. Woggon, W.-D., H.-A. Wagenknecht, and C. Claude cytochrome P450cam monoxygenase system.
(2001). Synthetic active site analogues of heme- Biochemistry 21, 370-377.
thiolate proteins characterization and identification 29. Groves, J.T. and D.V Adhyam (1984). Hydroxy-
of intermediates of the catalytic cycles of cyto- lation by cytochrome P-450 and metalloporphyrin
chrome P450cam and chloroperoxidase. J. Inorg. models. Evidence for allylic rearrangement. J. Am.
Biochem. 83, 289-300. Chem. Soc. 106,2177-2181.
16. Ortiz de Montellano, RR. (ed.) (1995). Cytochrome 30. Traylor, TG. and R Xu (1988). Model reactions
P-450: Structure, Mechanism and Biochemistry, related to cytochrome P-450. Effects of alkene
2 edn. Plenum Press, New York. structure on the rates of epoxide formation. J. Am.
17. Guengerich, ER (2001). Common and uncommon Chem. Soc. 110, 1953-1958.
cytochrome P450 reactions related to metabolism 31. Fish, K.M., G.E. Avaria, and J.T. Groves (1988).
and chemical toxicity. Chem. Res. Toxicol. 14, Rearrangement of alkyl hydroperoxides mediated
611-650. by cytochrome P-450: Evidence for the oxygen
18. Meunier, B. and J. Bernadou (2002). Metal-oxo rebound mechanism. In J. Miners, D.J. Birkett,
species in P450 enzymes and biomimetic models. R. Dew, B.K. May and M.E. McManus (eds.).
Oxo-hydroxo tautomerism with water-soluble met- Microsomes and Drug Oxidations. Taylor and
alloporphyrins. Top. Catal. 21, 47-54. Francis, New York, pp. 176-183.
19. Groves, J.T. (2003). The bioinorganic chemistry of 32. Kupfer, R., S.Y. Liu, A.J. Allentoff, and
iron in oxygenases and supramolecular assemblies. J.A. Thompson (2001). Comparisons of hydroper-
Proc. Nat. Acad Sci. USA 100, 3569-3574. oxide isomerase and monooxygenase activities of
20. Shteinman, A.A. (2001). The role of metal-oxygen cytochrome P450 for conversions of allylic hydro-
intermediates in biological and chemical mono- peroxides and alcohols to epoxyalcohols and diols:
oxygenation of alkanes. Russ. Chem. Bull. 50, Probing substrate reorientation in the active site.
1795-1810. Biochemistry 40, 11490-11501.
21. Makris, T.M., R. Davydov, I.G. Denisov, 33. Vaz, A.D.N., D.R McGinnity, and M.J. Coon (1998).
B.M. Hoffman, and S.G. Sligar (2002). Mechanistic Epoxidation of olefins by cytochrome P-450:
enzymology of oxygen activation by the cyto- Evidence from site-specific mutagenesis for
chromes P450. DrugMetab. Rev. 34, 691-708. hydroperoxo-iron as an electrophilic oxidant. Proc.
22. Watanabe, Y. and H. Fujii (2000). Characterization Natl. Acad Sci. USA, 95, 3555-3560.
of high-valent oxo-metalloporphyrins. In B. Meunier 34. Newcomb, M., P.F. Hollenberg, and M.J. Coon
(ed.). Structure and Bonding, Vol 97, Springer- (2003). Multiple mechanisms and multiple oxi-
Verlag, Berlin, pp. 61-89. dants in P450-catalyzed hydroxylations. Arch.
23. McLain, J., J. Lee, and J.T. Groves (1999). Biochem. Biophys. 409, 72-79.
Biomimetic oxygenations related to cytochrome 35. Veeger, C. (2002). Does P450-type catalysis pro-
P450: Metal-oxo and metal-peroxo intermediates. ceed through a peroxo-iron intermediate? A review
In B. Meunier (ed.), Biomimetic Oxidations. ICP of studies with microperoxidase. J. Inorg. Biochem.
Publishers, pp. 91-170. 9, 35-45.
36 John T. Groves

36. Davydov, R., T.M. Markis, V Kofman, D.E. Werst, cytochrome P-450 BM3 mediated sulfoxidation and
S.G. Sligar, and B.M. Hoffman (2001). N-dealkylation reactions. J. Am. Chem. Soc. 124,
Hydroxylation of camphor by reduced oxy- 9724-9725.
cytochrome P-450cam: Mechanistic imphcations of 48. Nam, W, S.W. Jin, M.H. Lim, J.Y Ryu, and C. Kim
Epr and Endor studies of catalytic intermediates in (2002). Anionic ligand effect on the nature of epox-
native and mutant enzymes. J. Am. Chem. Soc, 123, idizing intermediates in iron porphyrin complex-
1413-1415. catalyzed epoxidation reactions. Inorg. Chem. 41,
37. Ogliaro, E, S.P. de Visser, S. Cohen, P.K. Sharma, 3647-3652.
and S. Shaik (2002). Searching for the second oxi- 49. Machii, K., Y. Watanabe, and I. Morishima (1995).
dant in the catalytic cycle of cytochrome P450: A Acylperoxo-iron(III) porphyrin complexes: A new
theoretical investigation of the iron(III)- hydroper- entry of potent oxidants for the alkene epoxidation.
0X0 species and its epoxidation pathways. J. Am. J.Am. Chem. Soc. Ill, 6691-6697.
Chem. Soc. 124,2806-2817. 50. Suzuki, N., T Higuchi, and T. Nagano (2002).
38. Kamachi, T., Y. Shiota, T. Ohta, and K. Yoshizawa Multiple active intermediates in oxidation reaction
(2003). Does the hydroperoxo species of catalyzed by synthetic heme-thiolate complex rele-
cytochrome P450 participate in olefin epoxidation vant to cytochrome P450. J. Am. Chem. Soc. 124,
with the main oxidant, compound I? criticism from 9622-9628.
density functional theory calculations. Bull. Chem. 51. Yoshioka, S., T. Tosha, S. Takahashi, K. Ishimori,
Soc. Jpn. 6, 721-732. H. Hori, and I. Morishima (2002). Roles of the
39. Groves, J.T. and Y. Watanabe (1988). Reactive iron proximal hydrogen bonding network in cytochrome
porphyrin derivatives related to the catalytic cycles P450cam-catalyzed oxygenation. J. Am. Chem. Soc.
of cytochrome P450 and peroxidase. Studies of the 124, 14571-14579.
mechanism of oxygen activation. J. Am. Chem. Soc. 52. Manchester, J.I., J.P. Dinnocenzo, L.A. Higgins, and
110, 8443-8452. J.P. Jones (1997). A new mechanistic probe for
40. Akhtar, M., D. Corina, S. Miller, A.Z. Shyadehi, and cytochrome P450: An application of isotope effect
J.N. Wright (1994). Mechanism of the acyl-carbon profiles. J Am. Chem. Soc. 119, 5069-5070.
cleavage and related reactions catalyzed by multi- 53. Ortiz de Montellano, PR. and R.A. Stearns (1987).
functional P-450s: Studies on cytochrome P45017r. Bicyclopentane P450. J Am. Chem. Soc. 109,
Biochemistry 33, 4410^418. 3415-3420.
41. Cole, RA. and C.H. Robinson (1988). A peroxide 54. Atkinson, J.K. and K.U Ingold (1993). Cytochrome
model reaction for placental aromatase. J. Am. P450 hydroxylation of hydrocarbons: Variation in
Chem. Soc. 110, 1284-1285. the rate of oxygen rebound using cyclopropyl radi-
42. Vaz, A.D.N., E.S. Roberts, and M.J. Coon (1991). cal clocks including two new ultrafast probes.
Olefin formation in the oxidative deformylation of Biochemistry 32, 9209-9214.
aldehydes by cytochrome P-450. Mechanistic impli- 55. Newcomb, M. and PH. Toy (2000). Hypersensitive
cations for catalysis by oxygen-derived peroxide. radical probes and the mechanisms of cytochrome
J.Am. Chem. Soc. 113, 5886-5887. P450-catalyzed hydroxylation reactions. Ace. Chem.
43. Sheng, D., D.P Ballou, and V. Massey (2001). Res. 33, 449-455.
Mechanistic studies of cyclohexanone monooxyge- 56. Bowry, V. W and K.U. Ingold (1991). A radical clock
nase: Chemical properties of intermediates involved investigation of microsomal cytochrome P-450
in catalysis. Biochemistry 40, 11156-11167. hydroxylation of hydrocarbons. Rate of oxygen
44. Colonna, S., N. Gaggero, G. Carrea, G. Ottolina, rebound. J Am. Chem. Soc. 113, 5699-5707.
P. Pasta, and F. Zambianchi (2002). First asymmet- 57. Newcomb, M., R. Shen, S.-Y Choi, PT Toy,
ric epoxidation catalysed by cyclohexanone mono- RE Hollenberg, A.D.N. Vaz et al. (2000).
oxygenase. Tetrahedron Lett. 43, 1797-1799. Cytochrome P450-catalyzed hydroxylation of mech-
45. Watanabe, Y. (2001). Alternatives to the oxoferryl anistic probes that distinguish between radicals and
porphyrin cation radical as the proposed reactive cations. Evidence for cationic but not for radical
intermediate of cytochrome P450: Two-electron oxi- intermediates. J Am. Chem. Soc. 122, 2677-2686.
dized Fe(III) porphyrin derivatives. J. Biol. Inorg. 58. Wust, M. and R.B. Croteau (2002). Hydroxylation
Chem. 6, 846-856. of specifically deuterated limonene enantiomers by
46. Groves, J.T. and N. Jin (1999). Unusual kinetic sta- cytochrome P450 limonene-6-hydroxylase reveals
bility of a ground state singlet oxomanganese(V) the mechanism of multiple product formation.
porphyrin. Evidence for a spin state crossing effect. Biochemistry 4\, 1820-1827.
J. Am. Chem. Soc. 121, 2923-2924. 59. Audergon, C , K.R. Iyer, J.P. Jones, J.F. Darbyshire,
47. Volz, TJ., D.A. Rock, and J.P Jones (2002). and WF. Trager (1999). Experimental and theoreti-
Evidence for two different active oxygen species in cal study of the effect of active-site constrained
Models and Mechanisms of Cytochrome P450 Action 37

substrate motion on the magnitude of the observed for a singlet biradical in supercritical propane.
intra-molecular isotope effect for the P450 101 cat- J Am. Chem. Soc. 120, 1641-1642.
alyzed benzylic hydroxylation of isomeric xylenes 70. McMahon, R.J. (2003). Chemical reactions involv-
and 4,4'-dimethylbiphenyl. J. Am. Chem. Soc. 121, ing quantum tunneling. Science 299, 833-834.
41^7. 71. Zuev, PS., R.S. Sheridan, TV Albu, D.G. Truhlar,
60. (a) Shaik, S., S.P. de Visser, F. Ogliaro, H. Schwarz, D.A. Hrovat, and W.T. Borden (2003). Carbon tunnel-
and D. Schroder (2002). Two-state reactivity mecha- ing from a single quantum state. Science 299,867-870.
nisms of hydroxylation and epoxidation by cyto- 72. Horn, A.H.C. and T. Clark (2003). Does metal ion
chrome P-450 revealed by theory. Curr. Opin. Chem. complexation make radical clocks run fast? J. Am.
Biol. 6, 556-567; (b) Sharma, P.K., S.P de Visser, and Chem. Soc. 125, 2809-2816.
S. Shaik (2003). Can a single oxidant with two spin 73. Kopp, D.A. and S.J. Lippard (2002). Soluble
states masquerade as two different oxidants? A study methane monooxygenase: Activation of dioxygen
of the sulfoxidation mechanism by cytochrome P450. and methane. Curr. Opin. Chem. Biol. 568-576.
J.Am. Chem. Soc. 125, 8698-8699. 74. Austin, R.N., H.-K. Chang, G.J. Zylstra, and
61. Schroder, D., A. Fiedler, M.F. Ryan, and H. Schwarz J.T Groves (2000). The non-heme diiron alkane
(1994). Surprisingly low reactivity of base FeO^ in monooxygenase of Pseudomonas oleovorans
its spin-allowed, highly exothermic reaction with (AlkB) hydroxylates via a substrate radical interme-
molecular hydrogen to generate Fe+ and water. diate. J:^m. Chem. Soc. Ill, 11747-11748.
J. Phys. Chem. 98, 68-70. 75. Brazeau, B.J., R.N. Austin, C. Tarr, J.T. Groves, and
62. Schoneboom, J.C, H. Lin, N. Reuter, W. Thiel, J.D. Lipscomb (2001). Intermediate Q from soluble
S. Cohen, F Ogliaro et al (2002). The elusive methane monooxygenase hydroxylates the mechanis-
oxidant species of cytochrome P450 enzymes: tic substrate probe norcarane: Evidence for a stepwise
Characterization by combined quantum mechani- reaction, ^ ^ w . Chem. Soc. 123, 11831-11837.
cal/molecular mechanical (Qm/Mm) calculations. 76. Austin, R.N., K. Buzzi, E. Kim, G.B. Zylstra, and
J.Am. Chem. Soc. 124, 8142-8151. J.T. Groves (2003). Xylene monooxygenase, a
63. Auclaire, K., Z. Hu, D.M. Little, PR. Ortiz de membrane-spanning non-heme diiron enzyme that
Montellano, and J.T. Groves (2002). Revisiting the hydroxylates hydrocarbons via a substrate radical
mechanism of P-450 enzymes using the radical intermediate. J Biol. Inorg. Chem. 8, 733-739.
clocks norcarane and spiro[2,5]octane, 2002. J. Am. 77. Wei, C.C, Z.-Q. Wang, A.L. Meade, J.F McDonald,
Chem. Soc. 124, 6020-6027. and D.J. Stuehr (2002). Why do nitric oxide syn-
64. Newcomb, M., R.N. Shen, Y. Lu, M.J. Coon, thases use tetrahydrobiopterin? J. Inorg. Biochem.
PF Hollenberg, D.A. Kopp et al. (2002). Evaluation 91, 618-624.
of norcarane as a probe for radicals in cyto- 78. Wei, C.C, Z.Q. Wang, Q. Wang, A.L. Meade,
chome P450-and soluble methane monooxygenase- C. Hemann, R. Hille et al. (2001). Rapid kinetic
catalyzed hydroxylation reactions. J. Am. Chem. studies link tetrahydrobiopterin radical formation to
Soc. 124, 6879-6886. heme-dioxy reduction and arginine hydroxylation in
65. Ogliaro, F, S.P. de Visser, J.T. Groves, and S. Shaik inducible nitric-oxide synthase. J. Biol. Chem. 276,
(2001). Chameleon states: High-valent metal-oxo 315-319.
species of cytochrome P450 and its ruthenium ana- 79. Hurshman, A.R., C. Krebs, D.E. Edmondson,
log. Angew. Chem. Int. Ed. 40, 2874-2878. B.H. Huynh, and M.A. Marietta (1999). Formation
66. Sharma, PK., S.P de Visser, F Ogharo, and S. Shaik of a pterin radical in the reaction of the heme
(2003). Is the ruthenium analogue of compound I of domain of inducible nitric oxide synthase with
cytochrome P450 an efficient oxidant? A theoretical oxygen. Biochemistry 38, 15689-15696.
investigation of the methane hydroxylation reaction. 80. Hurshman, A.R. and M.A. Marietta (2002).
J Am. Chem. Soc. 125, 2291-2300. Reactions catalyzed by the heme domain of
67. Kamachi, T. and K. Yoshizawa (2003). A theoretical inducible nitric oxide synthase: Evidence for the
study on the mechanism of camphor hydroxylation involvement of tetrahydrobiopterin in electron
by compound I of cytochrome P450. J. Am. Chem. transfer. Biochemistry 41, 3439-3456.
Soc. 125,4652^661. 81. Rosen, G.M., P Tsai, and S. Pou (2002). Mechanism
68. Guallar, V, M.-H. Baik, S.J. Lippard, and of free-radical generation by nitric oxide synthase.
R.A. Friesner (2003). Peripheral heme substituents Chem. Rev. 102, 1191-1199.
control the hydrogen-atom abstraction chemistry in 82. Blasko, E., C.B. Glaser, J.J. Devlin, W Xia,
cytochromes P450. Proc. Natl. Acad. Set, USA 100, R.I. Feldman, M.A. Polokoff et al. (2002).
6998-7002. Mechanistic studies with potent and selective
69. Reyes, M.B. and B.K. Carpenter (1998). Evidence inducible nitric-oxide synthase dimerization
for interception of nonstatistical reactive trajectories inhibitors. J Biol. Chem. 271, 295-302.
38 John T. Groves

83. Davydov, R., A. Ledbetter-Rogers, R Martasek, structure of the chloroiron(III) derivative of the
M. Larukhin, M. Sono, J.H. Dawson et al (2002). meso-unsubstituted 2,7,12,17-tetramethyl-3,8,13,
Epr and endor characterization of intermediates in 18-tetramesitylporphyrin and weak ferromagnetic
the cryoreduced oxy-nitric oxide synthase heme exchange interactions in the a(lu) oxoiron(IV)
domain with bound L-arginine or N-G-hydroxyargi- porphyrin Pi radical cation complex. Chem. Eur J.
nmQ. Biochemistry 41, 10375-10381. 2,1159-1163.
84. Huang, H., J.M. Hah, and R.B. Silverman (2001). 96. Jayaraj, K., J. Terner, A. Gold, D.A. Roberts,
Mechanism of nitric oxide synthase. Evidence that R.N. Austin, D. Mandon et al. (1996). Influence of
direct hydrogen atom abstraction from the O-H meso substituents on electronic states of (oxofer-
bond of N-G-hydroxyarginine is not relevant to the ryl)porphyrin Pi-cation radicals. Inorg. Chem. 35,
mechanism. J. Am. Chem. Soc. 123, 2674-2676. 1632-1640.
85. Li, H., H. Shimizu, M. Flinspach, J. Jamal, W. Yang, 97. Jayaraj, K., A. Gold, R.N. Austin, D. Mandon,
M. Xian et al. (2002). The novel binding mode of R. Weiss, J. Terner et al. (1995). Compound-I and
N-alkyl-N'hydroxyguanidine to neuronal nitric compound-II analogs of a chlorine. J. Am. Chem.
oxide synthase provides mechanistic insights into Soc. 117,9079-9080.
no biosynthesis. Biochemistry 41, 13868-13875. 98. Muther, M., E. Bill, A.X. Trautwein, D. Mandon,
86. Groves, J.T. and Y.-Z. Han (1995). Models and R. Weiss, A. Gold et al. (1994). Spin coupling in
mechanisms of cytochrome P450 action. In distorted high-valent Fe(IV) porphyrin radical
RR.O.d. Montellano (ed.). Cytochrome P-450. complexes. Hyperflne Interact. 91, 803-808.
Structure, Mechanism and Biochemistry. Plenum 99. Nam, W, S.K. Choi, M.H. Lim, J.U. Rohde, 1. Kim,
Press, New York, pp. 3 ^ 8 . J. Kim et al. (2003). Reversible formation of
87. Davies, J.A., PL. Watson, A. Greenberg, and iodosylbenzene-iron porphyrin intermediates in
J.F. Liebman (1994). Selective Hydrocarbon the reaction of oxoiron(IV) porphyrin Pi-cation
Activation: Principle and Progress. VCH, New York. radicals and iodobenzene. Angew. Chem. Int. Ed.
88. Watanabe, Y (1999). High valent intermediates. 42, 109-111.
In K. Kadish, (ed.). The Porphyrin Encyclopedia. 100. Groves, J.T. and T.J. McMurry (1985). Synthetic
pp.97-117. analogs of oxidized heme proteins. Preparation
89. Groves, J.T., T.E. Nemo, and R.S. Myers (1979). and characterization of iron(lV) porphyrins. Rev.
Hydroxylation and epoxidation catalyzed by iron- Port. Chim. 27, 102-103.
porphine complexes. Oxygen transfer from iodosyl- 101. Fujii, H., T Yoshimura, and H. Kamada (1997).
benzene. J: ^m. Chem. Soc. 101, 1032-1033. Imidazole, and/?-nitrophenolate complexes of oxo-
90. Penner-Hahn, J.E., T.J. McMurry, M. Renner, iron(IV) porphyrin-cation radicals as models for
L. Latos-Grazynsky, K.S. Eble, I.M. Davis et al. compounds I of peroxidase and catalase. Inorg.
(1983). X-ray absorption spectroscopic studies of Chem. 36, 6\42-6\43.
high-valent iron porphyrins: Horseradish peroxidase 102. Groves, J.T, R.C. Haushalter, M. Nakamura,
(HRP) compounds I and II. J. Biol. Chem. 258, T.E. Nemo, and B.J. Evans (1981). High-valent
12761-12764. iron-porphyrin complexes related to peroxidase
91. Penner-Hahn, J.E., K.S. Eble, T.J. McMurry, and cytochrome P-450. J. Am. Chem. Soc. 102,
M. Renner, A.L. Balch, J.T. Groves et al. (1986). 2884-2886.
Structural characterization of horseradish peroxi- 103. Groves, J.T. and Y Watanabe (1988). Reactive iron
dase using EXAFS spectroscopy. Evidence for porphyrin derivatives related to the catalytic cycles
F e = 0 ligation in compounds I and II. J. Am. Chem. of cytochrome P450 and peroxidase. Studies of the
Soc. 108, 7819-7825. mechanism of oxygen activation. J. Am. Chem.
92. Groves, J.T, R. Quinn, T.J. McMurry, G. Lang, and Soc. 110,8443-8452.
B. Boso (1984). Iron(IV) porphyrins from iron(III) 104. Nam, W, YM. Goh, Y.J. Lee, M.H. Lim, and
porphyrin cation radicals. J. Chem. Soc. Chem. C. Kim (1999). Biomimetic alkane hydroxylations
Commun. 1455-1456. by an iron(III) porphyrin complex with H2O2 and
93. Boso, B., G. Lang, T McMurry, and J.T. Groves by a high-valent iron(IV) 0x0 porphyrin cation
(1983). Mossbauer-effect study of tight spin cou- radical complex. Inorg. Chem. 38, 3238.
pling in oxidized chloro-5,10,15,20-tetra(mesityl) 105. Gross, Z. and S.A. Nimri (1994). Pronounced axial
porphyrinatoiron(III). .^ Chem. Phys. 79, 1122-1126. ligand effect on the reactivity of oxoiron(IV) por-
94. Jayaraj, K., A. Gold, R.N. Austin, L.M. Ball, phyrin cation radicals. Inorg. Chem. 33, 1731-1732.
J. Terner, D. Mandon et al. (1997). Compound I and 106. Gross, Z., S. Nimri, CM. Barzilay, and
compound II analogues from porpholactones. Inorg. L. Simkhovich (1997). Reaction profile of the last
Chem. 36, 4555^566. step in cytochrome P-450 catalysis revealed by
95. Ayougou, K., D. Mandon, J. Fischer, R. Weiss, studies of model complexes. J. Biol. Inorg. Chem.
M. Muther, and V Schunemann (1996). Molecular 2, 492-506.
Models and Mechanisms of Cytochrome P450 Action 39

107. Gross, Z. and S. Ini (1997). Remarkable (IV) porphyrin complexes, J. Am. Chem. Soc. 110,
effects of metal, solvent and oxidant on 8628-8638.
metalloporphyrin-catalyzed enantioselective epox- 119. Collins, T.L and S.W. Gordon-Wyhe (1989).
idation of olefins. J. Org. Chem. 62, 5514-5521. A manganese(V)-oxo complex. J. Am. Chem. Soc.
108. Groves, J.T., Z. Gross, and M.K. Stern (1994). 111,4511^513.
Preparation and reactivity of oxoiron(IV) por- 120. Collins, T.L, R.D. Powell, C. Slebodnick, and
phyrins. Inorg. Chem. 33, 5065-5072. E.S. Uffelman (1990). A water-stable man-
109. Liu, M.H. and Y.O. Su (1998). Selective electro- ganese(V)-oxo complex: Definitive assignment of
catalysis of alkene oxidations in aqueous media. a Mn(V)=0 infrared vibration. J. Am. Chem. Soc.
Electrochemical and spectral characterization of 112,899-901.
oxo-ferryl porphyrin, oxo-ferryl porphyrin radical 121. MacDonnell, EM., N.L.R Fackler, C. Stem, and TV
cation and their reaction products with alkenes O'Halloran (1994). Air oxidation of a five-coordinate
at room temperature. J. Electroanal. Chem. 452, Mn(III) dimer to a high-valent oxomanganese(V)
113-125. complex, ^^m. Chem. Soc. 116, 7431-7432.
110. Nam, W., H.J. Han, S.Y. Oh, Y.J. Lee, M.H. Choi, 122. Miller, C.G., S.W. Gordon-Wylie, C.R Horwitz,
S.Y Han et al (2000). New insights into the mech- S.A. Strazisar, D.K. Periano, G.R. Clark et al.
anisms of O-O bond cleavage of hydrogen perox- (1998). A method for driving 0-atom transfer:
ide and tert-alkyl hydroperoxides by iron(III) Secondary ion binding to a tetraamide macrocyclic
porphyrin complexes. J. Am. Chem. Soc. 122, ligand. J ^ m . Chem. Soc. 120, 11540-11541.
8677-8684. 123. Srinivasan, K.M., P. Michaud, and LK. Kochi
111. Groves, J.T., W.J. Kruper, and R.C. Haushalter (1986). Epoxidation of olefins with cationic
(1980). Hydrocarbon oxidations with oxometallo- (salen)manganese(III) complexes. The modulation
porphinates. Isolation and reactions of a (porphi- of catalytic activity by substituents. J. Am. Chem.
nato)manganese(V) complex. J. Am. Chem. Soc. Soc. 108, 2309-2320.
102, 6377-6380. 124. Palucki, M.K, N.S. Finney, PL Pospisil,
112. Hill, C.L. and B.C. Schardt (1980). Alkane activa- M.L. Giiler, T. Ishida, and E.N. Jacobsen (1998).
tion and functionalization under mild conditions The mechanistic basis for electronic effects on enan-
by a homogeneous manganese(III)porphyrin- tioselectivity in the (salen)Mn(III)-catalyzed epoxi-
iodosylbenzene oxidizing system. J. Am. Chem. dation reaction. J. Am. Chem. Soc. 120, 948-954.
Soc. 102, 6374-6375. 125. Feichtinger, D. and D.A. Plattner (1997). Direct
113. Meunier, B., E. Guilmet, M.E. De Carvalho, and proof for 0=-Mn-V(salen) complexes. Angew.
R. Poilblanc (1984). Sodium hypochlorite: Chem. Int. Ed. 36, 1718-1719.
A convenient oxygen source for olefin epoxidation 126. Sivasubramanian, K.V, M. Ganesan, S. Rajagopal,
catalyzed by (porphyrinato)manganese complexes. and R. Ramaraj (2002). Iron(III)-salen complexes
J. Am. Chem. Soc. 106, 6668-6676. as enzyme models: Mechanistic study of
114. De Poorter, B. and B. Meunier (1985). oxo(salen)iron complexes oxygenation of organic
Metalloporphyrin-catalyzed epoxidation of termi- sulfides. ^ Or^. Chem. 67, 1506-1514.
nal olefins with hypochlorite salts or potassium 127. Jacobsen, E.N. (1995). Transition metal-catalyzed
hydrogen persulfate. Perkin Trans. II, J. Chem. Soc. oxidations: asymmetric epoxidation. In G. Wilkinson,
1735-1740. FG.A. Stone, E.W Abel, and L.S. Hegedus (eds),
115. Collman, J.P., J.L Brauman, and B. Meunier Comprehensive Organometallic Chemistry II, Vol.
(1984). Epoxidation of olefins by cytochrome 12. Pergamon: New York.
P-450 model compounds: Mechanism of oxygen 128. (a) Katsuki, T. (1995). Catalytic asymmetric oxida-
atom transfer. Proc. Natl. Acad. Sci. USA 81, tions using optically-active (salen)manganese(III)
3245-3248. complexes as catalysts. Coord. Chem. Rev. 140,
116. Collman, J.P., J.L Brauman, B. Meunier, 189; (b) Katsuki, T. (2002). Chiral metallosalen
T. Hayashi, T. Kodadek, and S.A. Raybuck (1985). complexes: Structures and catalyst tuning for
Epoxidation of olefins by cytochrome P-450 asymmetric epoxidation and cyclopropanation.
model compounds: Kinetics and stereochemistry Adv Synth. Catal. 344(2), 131-147.
of oxygen atom transfer and origin of shape selec- 129. Gross, Z., G. Golubkov, and L. Simkhovich (2000).
tivity. J. Am. Chem. Soc. 107, 2000-2005. Epoxidation catalysis by a manganese corrole and
117. Collman, IP., T. Kodadek, and J.L Brauman isolation of an oxomanganese(V) corrole. Angew.
(1986). Oxygenation of styrene by cytochrome Chem. Int. Ed 39, 4045^047.
P-450 model systems. A mechanistic study. J. Am. 130. Meier-Callahan, A.E., H.B. Gray, and Z. Gross
Chem. Soc. 108, 2588-2594. (2000). Stabilization of high-valent metals by
118. Groves, J.T. and M.K. Stern (1988). Synthesis, corroles: Oxo tris(pentafluorophenyl)corrolato
characterization and reactivity of oxomanganese chromium(V). Inorg. Chem. 39, 3605-3607.
40 John T. Groves

131. Meier-Callahan, A.E., A.J. Di Bilio, oxidation of substrates catalyzed by microperoxi-


L. Simkhovich, A. Mahammed, I. Goldberg, dase-8. J. Biol. Inorg Chem. 6, 770-777.
H.B. Gray et al. (2001). Chromium corroles in four 144. Meunier, B. and J. Bernadou (2000). Active iron-
oxidation states. Inorg. Chem. 40, 6788-6793. 0X0, and iron-peroxo species in cytochrome P450
132. Mahammed, A., H.B. Gray, A.E. Meier-Callahan, and peroxidases; oxo-hydroxo tautomerism with
and Z. Gross (2003). Aerobic oxidations catalyzed water-soluble porphyrins. In B. Meunier and
by chromium corroles. J. Am. Chem. Soc. 125, Waldemar Adam (ed.). Metal-Oxo and Metal-
1162-1163. Peroxo Species in Catalytic Oxidations, Vol. 97.
133. Groves, IT., Y. Watanabe, and T.J. McMurry Springer-Verlag, Berlin, pp. 1-35.
(1983). Oxygen activation by metalloporphyrins— 145. Jin, N., J.L. Bourassa, S.C. Tizio, and J.T. Groves
formation and decomposition of an acylperoxy- (2000). Rapid, reversible oxygen atom transfer
manganese(III) complex. J. Am. Chem. Soc. 105, between an oxomanganese(V) porphyrin and bro-
4489. mide. A haloperoxidase mimic with enzymatic
134. Groves, J.T. andM.K. Stem (1988). Synthesis, char- rates. Angew. Chem. Int. Edit. 39, 3849-3851.
acterization and reactivity of oxomanganese(IV) 146. Groves, J.T, J. Lee, and S.S. Maria (1997).
porphyrin complexes. ^^ ^m. Chem. Soc. 110, 8628. Detection and characterization of an oxo-
135. Groves, J.T. and Y. Watanabe (1986). Oxygen acti- manganese(V) porphyrin complex by rapid-mixing
vation by metalloporphyrins related to peroxidase stopped-flow spectrophotometry. J. Am. Chem.
and cytochrome-P-450—direct observation of the Soc. 119, 6269-6273.
O-O bond-cleavage step. J. Am. Chem. Soc. 108, 147. Nam, W, 1. Kim, M.H. Lim, H.J. Choi, J.S. Lee,
7834-7836. and H.G. Jang (2002). Isolation of an oxo-
136. Groves, J.T. and S.S. Maria (1995). Peroxynitrite- manganese(V) porphyrin intermediate in the
induced DNA strand scission mediated by reaction of a manganese(III) porphyrin complex
a manganese porphyrin. J. Am. Chem. Soc. 117, and H2O2 in aqueous solution. Chem. Eur J. 8,
9578-9579. 2067-2071.
137. Bernadou, J., A.-S. Fabiano, A. Robert, and 148. Nam, W, H.J. Lee, S.Y Oh, C. Kim, and H.G. Jang
B. Meunier (1994). "Redox Tautomerism" in high- (2000). First success of catalytic epoxidation of
valent metal-oxo-aquo complexes. Origin of the olefins by an electron-rich iron(III) porphyrin
oxygen atom in epoxidation reactions catalyzed complex and H2O2: Imidazole effect on the activa-
by water-soluble metalloporphyrins. J. Am. Chem. tion of H2O2 by iron porphyrin complexes in apro-
Soc. 116, 9375-9376. tic solvent. J. Inorg. Biochem. 80, 219-225.
138. Pitie, M., J. Bernadou, and B. Meunier (1995). 149. Beckman, J.S. and W.H. Koppenol (1996). Nitric
Oxidation at carbon-T of DNA deoxyriboses by oxide, superoxide and peroxynitrite: The good, the
the Mn-TMPyP/KHS05 system results from a h2id?in&X\\Q\xg\y. Am. J. Physiol. 271,C1424-C1437.
cytochrome P-450-type hydroxylation reaction. 150. Maria, S.S., J. Lee, and J.T. Groves (1997).
J.Am. Chem. Soc. Ill, 2935-2936. Peroxynitrite rapidly permeates phospholipid
139. Balahura, R.J., A. Sorokin, J. Bernadou, and membranes. Proc. Natl. Acad. Sci. USA 94,
B. Meunier (1997). Origin of the oxygen atom in 14243-14248.
C-H bond oxidations catalyzed by a water-soluble 151. Lee, J., J.A. Hunt, and J.T Groves (1997). Rapid
metalloporphyrin. Inorg. Chem. 36, 3488-3492. decomposition of peroxynitrite by manganese
140. Czernuszewicz, R.S., YO. Su, M.K. Stern, porphyrin-antioxidant redox couples. Bioorg. Med.
K.A. Macor, D. Kim, J.T. Groves, and T.G. Spiro Chem. Lett. 7,2913-2918.
(1988). Oxomanganese(IV) porphyrins identified 152. Groves, J.T. (1999). Peroxynitrite: Reactive, inva-
by resonance raman and infrared spectroscopy: sive and enigmatic. Curr Opin. Chem. Biol. 3,
Weak bonds and the stability of the half-filled T2g 226-235.
subshell. J. ^m. Chem. Soc. 110, 4158-^165. 153. Lee, J., J.A. Hunt, and J.T. Groves (1998). Manga-
141. Arasasingham, R.D., G.X. He, and T.C. Bruice nese porphyrins as redox-coupled peroxynitrite
(1993). Mechanism of manganese porphyrin- reductases. J. Am. Chem. Soc. 120, 6053-6061.
catalyzed oxidation of alkenes. Role of man- 154. Hunt, J.A., J. Lee, and J.T Groves (1997).
ganese(IV)-oxo species. J. Am. Chem. Soc. 115, Amphiphilic peroxynitrite decomposition catalysts
7985-7991. in liposomal assemblies. Chem. Biol. 4, 845-858.
142. Yeh, H.C., C.H. Yu, J.S. Wang, S.T Chen, YO. Su, 155. Stern, M.K., M.P Jensen, and K. Kramer (1996).
and W.Y Lin (2002). Stopped-flow kinetic study of Peroxynitrite decomposition catalysts. J. Am.
the peroxidase reactions of mangano-microperoxi- Chem. Soc. 118, 8735-8736.
dase-8. J. Biol. Inorg. Chem. 7, 113-119. 156. Lee, J., J.A. Hunt, and J.T Groves (1998).
143. Yeh, H.C., J.S. Wang, YO. Su, and W.Y Lin Mechanisms of iron porphyrin reactions with
(2001). Stopped-flow kinetic study of the h2o2 peroxynitrite. J ^m. Chem. Soc. 120, 7493-7501.
Models and Mechanisms of Cytochrome P450 Action 41

157. Shimanovich, R. and J.T. Groves (2001). catalyzed by vaulted binaphthyl metallopor-
Mechanisms of peroxynitrite decomposition phyrins. J. Org. Chem. 55, 3628-3634.
catalyzed by fetmps, a bioactive sulfonated iron 171. Groves, J.T. and K.V. Shalyaev (1998).
porphyrin. Arch. Biochem. Biophys. 387, 307-317. Paramagnetic ^H-NMR relaxation probes of stereo-
158. Szabo, C, J.G. Mabley, S.M. Moeller, selectivity in metalloporphyrin catalyzed olefin
R. Shimanovich, R Pacher, L. Virag et al. (2002). epoxidation. Chirality 10, 106-119.
Pathogenic role of peroxynitrite in the development 172. Nakayama, S., F. Tani, M. Matsu-ura, and
of diabetes and diabetic vascular complications: Y Naruta (2002). Cobalt "single-coronet" por-
Studies with FP15, a novel, potent peroxynitrite phyrin bearing hydroxyl groups in its 0-2 binding
decomposition catalyst. Mol. Med. 8, 571-580. site as a new model for myoglobin and hemoglo-
159. Collman, J.P, X. Zhang, VJ. Lee, E.S. Uffelman, bin: Observation of unusually low frequency of
and J.I. Brauman (1993). Regioselective and enan- V(O-O) in resonance raman spectrum. Chem. Lett.
tioselective epoxidation catalyzed by metallopor- A96-A91.
phyrins. Science 261, 1404-1411. 173. Matsui, E., Y Naruta, F. Tani, and Y Shimazaki
160. Rose, E., A. Lecas, M. Quelquejeu, A. Kossanyi, (2003). An active-site model of prostaglandin H
and B. Boitrel (1998). Synthesis of biomimetic synthase: An iron "twin-coronet" porphyrin with
heme precursors. Coord. Chem. Rev. 178-180, an aryloxyl radical overhang and its catalytic oxy-
1407-1431. genation of 1,4-diene. Angew. Chem. Int. Edit. 42,
161. Tani, E, M. Matsu-ura, S. Nakayama, and Y. Naruta 2744-2747.
(2002). Synthetic models for the active site of 174. Shilov, A.E. and G.B. Shul'pin (1997). Activation
cytochrome P450. Coord Chem. Rev. 226,219-226. of C-H bonds by metal complexes. Chem. Rev. 97,
162. Fokin, A.A. and PR. Schreiner (2002). Selective 2897-2932.
alkane transformations via radicals and radical 175. Groves, J.T. and R. Quinn (1984). Models of oxi-
cations: Insights into the activation step from dized heme proteins. Preparation and characteriza-
experiment and theory. Chem. Rev. 102, tion of a trans-dioxoruthenium(VI) porphyrin
1551-1593. complex. Inorg. Chem. 23, 3844-3846.
163. Breslow, R., Y. Huang, and X.J. Zhang (1997). 176. Groves, J.T. and R. Quinn (1985). Aerobic epoxi-
An artificial cytochrome P450 that hydroxylates dation of olefins with ruthenium porphyrin
unactivated carbons with regio- and stereoselectiv- catalysts. J. Am. Chem. Soc. 107, 5790-5792.
ity and useful catalytic turnovers. Proc. Nad. Acad. 177. Paeng, I.R. and K. Nakamoto (1990). Resonance
Sci. 94, 11156-11158. raman spectra of reaction intermediates in oxida-
164. (a) Breslow, R., X. Zhang, and Y Huang (1997). tion process of ruthenium(II) and iron(II) por-
Selective catalytic hydroxylation of a steroid by an phyrins. J ^m. Chem. Soc. 112, 3289.
artificial cytochrome P-450 enzyme. J. Am. Chem. 178. Pronievich, L.M., I.R. Paeng, W. Lewandowski,
Soc. 119, 4535-4536; (b) Breslow, R., Y. Huang, and K. Nakamoto (1990). Vibrational spectra of
and X.J. Zhang (1997). An artificial cytochrome dioxygen adducts and oxo complexes of ruthenium
P450 that hydroxylates unactivated carbons with tetraphenylporphyrine (RuTPP) J. Mol. Struct.
regio- and stereoselectivity and useful catalytic 219, 335-339.
turnovers, Proc. Nad. Acad. Sci. 94, 11156-11158. 179. Han, Y-Z. (1991). PhD Thesis, Department of
165. Groves, J.T. (1997). Artificial enzymes—^the Chemistry, Princeton University.
importance of being selective. Nature 389, 329. 180. Dubourdeaux, P., M. Tavares, A. Grand,
166. Collman, J.P, X. Zhang, R.T. Hembre, and R. Ramasseul, and J.-C. Marchon (1995). Prepara-
J.I. Brauman (1990). Shape-selective olefin tion and crystal structure of trans-dihydroxo-
epoxidation catalyzed by manganese picnic basket [tetrakis(2,6-dichlorophenyl)porphinato]rutheniu
porphyrins. J.Am. Chem. Soc. 112, 5356-5357. m(IV) 2toluene. Inorg. Chem. Acta 240, 657-660.
167. Collman, J.P, Z.W.A. Straumanis, M. Quelquejeu, 181. Ho, C , W.-H. Leung, and C.-M. Che (1991).
and E. Rose (1999). An efficient catalyst for asym- Kinetics of C-H bond and alkene oxidation by
metric epoxidation of terminal olefins. J. Am. trans-dioxoruthenium(VI) porphyrins. J. Chem.
Chem. Soc. 121,460^61. Soc. Dalton. Trans. 11, 2933-2939.
168. Groves, J.T. and R.S. Myers (1983). Catalytic 182. (a) Lindsay-Smith, J.R. and PR. Sleath (1982).
asymmetric epoxidation with chiral iron por- Model systems for cytochrome P450 dependent
phyrins, a ^m. Chem. Soc. 105, 5791-5796. monooxygenases 1. Oxidation of alkenes and
169. Groves, J.T. and P Viski (1989). Asymmetric aromatic compounds by tetraphenylporphinatoi-
hydroxylation by a chiral iron porphyrin. J. Am. ron(III), Trans. II. J. Chem. Soc. 1009; (b) Prado-
Chem. Soc. I l l , 8537-8538. Manso, C.M.C., E.A. Vidoto, RS. Vinhado,
170. Groves, J.T. and P Viski (1990). Asymmetric H.C. Sacco, K.J. Ciuffi, PR. Martins et al
hydroxylation, epoxidation and sulfoxidation (1999). Characterization and catalytic activity of
42 John T. Groves

iron(III) mono(4-N-methyl pyridyl)-tris 195. Ohtake, H., T. Higuchi, and M. Hirobe (1992).
(halophenyl) porphyrins in homogeneous and het- Highly efficient oxidation of alkanes and alkyl
erogeneous systems. J. Mol Catal A-Chem. 150, alcohols with heteroaromatic N-oxides catalyzed
251-266. by ruthenium porphyrins. J. Am. Chem. Soc. 114,
183. Bortolini, O. and B. Meunier (1984). Enhanced 10660-10662.
selectivity by an open-well effect in a metallopor- 196. Ohtake, H., T. Higuchi, and M. Hirobe (1995). The
phyrin-catalyzed oxygenation reaction. Perkin highly efficient oxidation of olefins, alcohols, sul-
Trans. II, J. Chem. Soc. 1967-1970. fides and alkanes with heteroaromatic N-oxides
184. Murahashi, S.-I., T. Naota, and N. Komiya (1995). catalyzed by ruthenium porphyrins. Heterocycles
Metalloporphyrin-catalyzed oxidations of alkanes 40, 867-903.
with molecular oxygen in the presence of acetalde- 197. Nakagawa, H., T. Higuchi, K. Kikuchi, Y Urano,
hyde. Tetrahedron Lett. 36, 8059-8062. andT. Nagano (1998). Selective deoxygenation of
185. Murahashi, S.-I. andN. Komiya (1998). New types heteroaromatic N-oxides with olefins catalyzed by
of catalytic oxidations in organic synthesis. Catal. ruthenium porphyrin. Chem. Pharm. Bull. 46,
Today 41, 339-349. 1656-1657.
186. Ohkatsu, Y. and T. Osa (1977). Liquid-phase oxi- 198. Ohtake, H., T. Higuchi, and M. Hirobe (1992). The
dation of aldehydes with metal tetra(paratolyl)por- selectivities and the mechanism of highly efficient
phyrins. Bull. Chem. Soc. Jpn. 50, 2945. epoxidation of olefins with 2,6-disubstituted pyri-
187. Birnbaum, E.R., J.A. Labinger, J.E. Bercaw, and dine N-oxides catalyzed by ruthenium porphyrin.
H.B. Gray (1998). Catalysis of aerobic olefic oxi- Tetrahedron Lett. 33, 2521-2524.
dation by a ruthenium perhaloporphyrin complex. 199. Higuchi, T. and M. Hirobe (1996). Four recent
Inorg. Chim. Acta 270, 433^39. studies in cytochrome P450 modelings: A stable
188. Birnbaum, E.R., M.W. GrinstafF, J.A. Labinger, iron porphyrin coordinated by a thiolate ligand;
J.E. Bercaw, and H.B. Gray (1995). On the mecha- a robust ruthenium porphyrin-pyridine N-oxide
nism of catalytic alkene oxidation by molecular derivatives system; polypeptide-bound iron por-
oxygen and halogenated iron porphyrins. J. Mol. phyrin; application to drug metabolism studies.
Catal. A: Chemical 104, L119-L122. J. Mol. Catal. A: Chem. 113, 403^22.
189. Grinstaff, M.W., M.G. Hill, J.A. Labinger, and 200. Shingaki, T, K. Miura, T. Higuchi, M. Hirobe, and
H.B. Gray (1994). Mechanism of catalytic oxy- T. Nagano (1997). Regio- and stereo-selective
genation of alkanes by halogenated iron por- oxidation of steroids using 2, 6-dichloropyridine
phyrins. Science 264, 1311. N-oxide catalyzed by ruthenium porphyrins.
190. Groves, J.T. and J.S. Roman (1995). Nitrous oxide J. Chem. Soc, Chem. Commun. 861-862.
activation by a ruthenium porphyrin. J. Am. Chem. 201. Groves, J.T., M. Bonchio, T. Carofiglio, and
Soc. 117,5594-5595. K. Shalyaev (1996). Rapid catalytic oxygenation
191. Banks, R.G.S., R.J. Henderson, and J.M. Pratt of hydrocarbons by ruthenium pentafluorophenyl-
(1968). Reactions of gases in solution, 3. Some porphyrin complexes: Evidence for the involve-
reactions of nitrous oxide with transition metal ment of a Ru(IIl) intermediate. J. Am. Chem. Soc.
complexes. J. Chem. Soc. (A) 2886-2889. 118,8961-8962.
192. (a) Bottomley, F, I.J. Lin, and M. Mukaida (1980). 202. Groves, J.T, K.V. Shalyaev, M. Bonchio, and
Reactions of dinitrogen oxide (nitrous oxide) with T Carofiglio (1997). Rapid catalytic oxygenation
dicyclopentadienyltitanium complexes including a of hydrocarbons with polyhalogenated ruthenium
reaction in which carbon monoxide is oxidized. J. porphyrin complexes. Stud. Surf. Sci. Catal. 110,
Am. Chem. Soc. 102, 5238-5242; (b) Yamada, T., 865-872.
K. Hashimoto, Y Kitaichi, K. Suzuki, and T. Ikeno 203. James, B.R. (1986). Transition metal catalyzed
(2001). Nitrous oxide oxidation of olefins cat- oxidations. In A.E. Shilov (ed.), Funda-mentals of
alyzed by ruthenium porphyrin complexes. Chem. Research in Homogeneous Catalysis. Gordon
Lett. 3, 268-269. Breach, New York, pp. 309-324.
193. Higuchi, T., H. Ohtake, and M. Hirobe (1989). 204. Barley, M.H., D. Dolphin, and B.R. James (1984).
Highly efficient epoxidation of olefins with pyri- Reversible intramolecular electron transfer within a
dine N-oxides catalyzed by ruthenium porphyrins. ruthenium(lll) porphyrin-ruthenium(Il) porphyrin
Tetrahedron Lett. 30, 6545-6548. radical system induced by changes in axial ligation.
194. Higuchi, T., H. Ohtake, and M. Hirobe (1991). J. Chem. Soc, Chem. Commun. 1499-1500.
Highly efficient oxygen transfer reactions from 205. Ojima, 1. (ed.) (1993). Catalytic Asymmetric
various heteroaromatic N-oxides to olefins, alco- Synthesis, VCH, New York.
hols and sulfides catalyzed by ruthenium por- 206. (a) Moro-oka, Y. and M. Akita (1998). Bio-inorganic
phyrins. Tetrahedron Lett. 32, 7435-7438. approach to hydrocarbon oxidation. Catalysis Today
Models and Mechanisms of Cytochrome P450 Action 43

41, 327-338; (b) Zhang, R., W.Y. Yu, H.Z. Sun, 208. Gross, Z., S. Ini, M. Kapon, and S. Cohen (1996).
W.S. Liu, and CM. Che (2002). Stereo- and First utilization of a homochiral ruthenium
enantioselective alkene epoxidations: A compara- porphyrin as enantioselective epoxidation catalyst.
tive study of D-4-and D-2-symmetric homochiral Tetrahedron Lett. 37, 7325-7328.
trans-dioxoruthenium(VI) porphyrins. Chem. Eur. 209. Lai, T.S., R. Zhang, K.K. Cheung, H.L. Kwong,
J. 8, 2495-2507; (c) Simonneaux, G., and P. Le and CM. Che. Stoichiometric enantioselective
Maux (2002). Optically active ruthenium por- alkene epoxidation with a chiral dioxoruthe-
phyrins: Chiral recognition and asymmetric cataly- nium(VI) D-4-porphyrinato complex. J. Chem.
sis. Coord. Chem. Rev. 228, 43-60; (d) Funyu, S., Soc., Dalton Trans. 21, 3559-3564.
T. Isobe, S. Takagi, D.A. Tryk, and H. Inoue 210. Lai, T.S., H.L. Kwong, R. Zhang, and CM. Che
(2003). Highly efficient and selective epoxidation (1998). Aerobic enantioselective alkene epoxida-
of alkenes by photochemical oxygenation sensi- tion by a chiral trans-dioxo(D-4-porphyrinato)
tized by a ruthenium(II) porphyrin with water as ruthenium(VI) complex. J. Chem. Soc. Chem.
both electron and oxygen donor. J. Am. Chem. Soc. Commun. 15, 1583-1584.
125, 5734-5740. 211. Berkessel, A. and M. Frauenkron (1997). Catalytic
207. Mansuy, D. (1993). Activation of alkanes, the bio- asymmetric epoxidation with a chiral ruthenium
mimetic approach. Coord. Chem. Rev. 125, 129-141. porphyrin and N-oxides. J. Chem. Soc, Perkin
Trans. 16(1), 2265-2266.
2
Computational Approaches to
Cytochrome P450 Function
Sason Shaik and Samuel P. De Visser

This chapter describes computational strategies empirical QM method to calculate electronic


for investigating the species in the catalytic cycle structure and derive the Mssbauer parameters for
of the enzyme cytochrome P450, and the mecha- the species. However, it was only after the devel-
nisms of its main processes: alkane hydroxylation, opment of density functional theoretic (DFT)
alkene epoxidation, arene hydroxylation, and sul- methods that QM theory came of age and offered
foxidation. The methods reviewed are molecular a tool that combines reasonable accuracy with
mechanical (MM)-based approaches (used e.g., to speed. More recently, even better tools became
study substrate docking), quantum mechanical available when DFT calculations were combined
(QM) and QM/MM calculations (used to study with MM approaches, leading to hybrid QM/MM
electronic structure and mechanism). methods that enable the study of active species in
their native protein environment. All these devel-
opments have had a considerable impact on the
1. Introduction field and an ever-growing surge of theoreti-
cal activity. It was therefore deemed timely to
The action of cytochrome P450 (P450) review the results and insights provided by these
enzymes has been for years a very active arena of methods. The chapter starts with a very brief sum-
research that led to important insights, and gener- mary of the theoretical methods, and follows with
ated lively debates over the nature of the various a description of the various species in the catalytic
species and their reactivity patterns ^ The active cycle and the main mechanisms by which the
species of the enzyme is based on an iron ligated reactive species of the enzyme transfers oxygen
to a protoporphyrin IX macrocycle and two addi- into organic compounds. As a matter of policy, the
tional axial ligands (Figure 2.1): one, called prox- main emphasis of the chapter is on QM-based
imal, is a thiolate from a cysteinate side chain of methods; while other methods are mentioned, they
the protein and the other, called distal, is a variable receive less coverage.
ligand that changes during the cycle. When the
distal ligand becomes an oxo group, the species is
called Compound I (Cpd I), which is the reactive
species of the enzyme and one of the most potent 2. Methods
oxidants known in nature.
One of the pioneering theoretical studies on The recent monograph of Cramer^ provides an
Cpd I was carried out by Loew et al?, using an excellent exposition of the theoretical methods,

Sason Shaik and Samuel P. De Visser • Department of Organic Chemistry and the Lise-Meitner-Minerva
Center for Computational Quantum Chemistry, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel.
Cytochrome P450: Structure, Mechanism, and Biochemistry, 3e, edited by Paul R. Ortiz de Montellano
Kluwer Academic / Plenum Publishers, New York, 2005.

45
46 Sason Shaik and Samuel P. De Visser

The Active Species of tbe Enzyme:

Figure 2.1. The active site of Cytochrome P450, showing Cpd I.

and the interested reader may consult this book for that the ab initio methods, which are the most time
further details than the meager summary given consuming, are generally the most accurate, while
here. There are three generic theoretical cate- MM is the fastest and also the least accurate.
gories: ab initio methods, semiempirical methods, The ab initio methods are split into two parts:
and MM methods. Both ab initio and semi- the wave mechanical approaches and the DFT
empirical methods derive from QM theory; they methods. In the wave mechanical approach, one
provide electronic structure information and can starts with the Schrdinger equation and mini-
be applied to the study of complete reaction path- mizes the energy by optimizing the orbitals in
ways. By contrast, MM is a classical method: it a wave function that represents the system at a
provides only three dimensional structures and certain level of accuracy; the latter is defined by
energies, and is limited to the study of related the degree of coverage of electronic correlation.
structures, for example, conformers. In ab initio The resulting wave fiinction defines a specific
methods, once one selects a basis set (a set of electronic state, characterized by certain orbital
functions) that describes the atomic orbitals of the occupation, total spin and energy, from which
atoms in the molecule, the functional (in DFT) all the properties of the molecule can be derived.
and everything else is calculated starting from The lowest ab initio level, called Hartree-Fock,
scratch; DFT is such an approach. Semiempirical describes the wave function by a single determi-
methods calculate part of the terms while others nant built from doubly occupied orbitals, and
are imported from "appropriate" experimental as such does not include electron correlation.
data. MM too, uses a mix of empirical and theo- This method fails completely for transition metal
retically calibrated data (e.g., force constants, containing molecules, which require multi-
atomic size parameters, strain energies, van der determinantal methods that incorporate static and
Waals parameters, etc.) to calculate the energy as dynamic electron correlation, such as the com-
a function of geometry, and the parameters are plete active space self consistent field (CASSCF)
usually system specific, for example, to proteins, method and its further augmentation by second-
hydrocarbons, etc. As a crude generalization, order perturbation theory, the CASPT2 method
there is an inverse relationship between the speed that also incorporates dynamic electron correla-
of the method and its reliability/accuracy, such tion, or the CCSD(T) method that corresponds to
Computational Approaches to Cytochrome P450 Function 47

coupled cluster with single and double excitations same information as ab initio methods with the
and perturbative triple excitations. At the time of exception of accuracy. The current method that
writing this chapter, CASSCF, CASPT2, and can be employed for transition metal compounds
CCSD(T) methods are still too time consuming is SAMl that is based on the older AMI method.
for P450 species, if one wishes to optimize the While the method seems to be very good for zinc
geometries of the species or to trace their potential compounds, it is still inaccurate for many of the
energy surfaces for a given reaction"^. transition metals. Another semiempirical method
DFT methods also use a wave function, but used for spectroscopic properties is INDO/S/CI
this wave function serves merely to obtain the that also includes configuration interaction (CI).
electron density of the molecule, and it is from These semiempirical methods are very fast and
the density that the energy and all molecular prop- can be used for very large systems, but their
erties are subsequently derived. Even though the accuracy remains questionable.
auxiliary wave function in DFT has a single deter- MM expresses the total energy of a molecular
minant form, the energy expression extracted system as a sum of bond energy terms, electro-
from it incorporates static as well as dynamic static terms, and van der Waals interactions.
electron correlation. Consequently, the DFT pro- The bond energy terms include bond stretching
cedure is faster than the ab initio procedures; its angular deformation, and torsional deviation
time consumption scales like Hartree-Fock theory, that are evaluated based on a force-field derived
but its accuracy is much better, and is sometimes from parameters calibrated for these bond types.
competitive even with CASPT2'^. As such, DFT The electrostatic interactions involve classical
can treat systems of up to c.lOO or more atoms and electrostatics; for proteins, usually, the partial
obtain results with decent accuracy for an entire charges are kept fixed during the calculations. The
potential energy surface of an enzymatic reaction. van der Waals interactions involve a Lennard-
Pure fiinctionals with gradient correction of Jones potential with 1/r^ and 1/r^^ terms for the
the density, for example, BP86, BLYP, BPW91, interaction energy. Since MM calculations are
etc., are considered to be suitable for enzymatic cheap, they can be used to run molecular dynam-
species. However, much better are hybrid density ics (MD) calculations to study multiple conforma-
fimctional methods, such as B3LYP or B3PW91, tions, as well as to map and sample an entire
which can reproduce experimental enthalpies of potential energy surface and to determine free
formation within 3 kcal mol~^ while the highly energies. The dynamics is calculated according to
elaborate wave mechanical methods reproduce the Newton's laws of classical mechanics. However,
same set of data within 1.6 kcal mol~^. Reaction due to the dimensionality of the problem, there are
barriers are more difficult to reproduce with many algorithms for running dynamics and for
experimental accuracy, and here too B3LYP is sampling the configuration space. These MM
considered to be better than the other DFT proce- calculations require computer resources and can
dures. As such, B3LYP has become the standard be applied to large systems, such as enzymes and
method for carrying out biological and P450 proteins. Thus, MM and MD studies are usefiil for
related research"*. To instill some uniformity in the studies of large systems, in which one, for
this chapter, many of the calculations were instance, explores the entrance and exit channels
repeated here by us, using B3LYP with a double of substrates/products into an enzyme^' ^ and the
zeta basis set, composed of LACVP for Fe and preferred location of substrate binding^' ^. By
6-3IG for all other atoms, hence LACVP(Fe)/ contrast, the electronic structures of all species
6-31G(H,C,N,0,S). This basis set gives qualita- of P450 involve unpaired electrons, different spin
tively reliable results for the assemblage of P450 states, and unusual mixed-valent states, which can
species. be studied only with QM methods.
Semiempirical QM methods are also based on The QM/MM method combines the advan-
wave mechanics. However, unlike the ab initio tages of QM and MM. In QM/MM calculations,
methods, here many of the terms are not calcu- the system is divided into two subsystems that
lated but taken from some empirical data, which are treated at different levels. The small sub-
are fiirther recalibrated to fit a set of properties. system involves the active species that is computed
In principle, semiempirical methods provide the with a QM method such as a semiempirical or DFT.
48 Sason Shaik and Samuel P. De Visser

The larger subsystem is the protein, which does (HS) fashion, and the complex itself becomes
not actively take part in the studied reaction, but a good electron acceptor. This triggers a one-
influences the active species by providing an elec- electron transfer from the reductase domain
trostatic field and a hydrogen bonding machinery, that reduces 2 to the HS ferrous (Fe") complex
as well as a sterically constrained matrix with van 3. Ferrous porphyrin is a good dioxygen binder,
der Waals interactions. The protein subsystem is and this leads to the binding of molecular oxygen
considered to be "classical" and is treated with to produce the LS ferrous-dioxygen complex,
MM. The interactions between the QM and MM 4. The latter species is again a good electron
subsystems are split into electrostatic and van der acceptor and this causes another electron transfer
Waals types. The electrostatic interactions are from the reductase to give rise to the twice-
incorporated into the QM calculations by embed- reduced ferric-dioxo species (5). The ferric-dioxo
ding the MM charges into the QM Hamiltonian; complex is now a good Lewis base, and therefore
in this manner, the QM subsystem undergoes undergoes protonation to yield the ferric peroxide
polarization in response to the electric field complex 6 that is also referred to as Cpd 0. Since
and hydrogen bonding machinery of the protein Cpd 0 is still a good Lewis base, it undergoes a
environment. The van der Waals interactions are second protonation and releases a water molecule
treated classically. QM/MM calculations that leading to the reactive species 7, which is a high-
involve DFT for the QM subsystem are still very valent iron-oxo complex also known as Cpd L
demanding and elaborate, and, at the time of Cpd I, in turn, transfers the distal oxygen atom to
writing this chapter, are limited to a few species the substrate, which is released and is replaced by
of P450. Moreover, the method is still limited to a water molecule to regenerate the resting state of
QM/MM geometry optimization, but cannot yet the enzyme (1). Alternative species, such as
be used for proper sampling of configuration the Fe-0H2-0~ intermediate^^ and Cpd II that
space, which is required in order to derive free results from the one-electron reduction of Cpd I
energy quantities. (ref [11]), have been suggested to participate in
the cycle. However, as of now there is no experi-
mental evidence for the existence of these
3. The Catalytic Cycle of P450 intermediates in the P450 cycle itself, albeit
Cpd II is well known for related heme enz>ines^^.
The enzyme cytochrome P450 operates by There are two additional key features in the
means of a catalytic cycle^ that is schematically cycle. First is the good electron donor ability
depicted in Figure 2.2, where the cysteinate prox- of the proximal thiolate ligand, which is thought
imal ligand is abbreviated as L, and the porphyrin to have a significant impact on the entire cycle,
macrocycle is symbolized by the two bold lines and especially on the generation of Cpd I. This
flanking the iron. The cycle follows a beautiful property of the thiolate is also called the "push
chemical logic that highlights the impact of chem- effect"'^. A second important feature that controls
istry on the field. The resting state of the enzyme the efficacy of the cycle is the hydrogen-bonding
is the ferric (Fe"^) complex (1) that possesses interaction in the proximal side with the thiolate
a water molecule as a distal ligand. With six coor- ligand in the cysteine loop. Thus, as shown by
dination, the d-block orbitals of the complex are Poulos^'* and subsequently by Schlichting et al}^,
split into three-below-two molecular orbitals in P450^^j^ the sulftir ligand is hydrogen bonded to
(MOs). Consequently, the five d-electrons of the three amidic hydrogens due to the side chains,
complex occupy the lower MOs leading to a low- Gln3^Q, Gly359, and Leu33g, while a fourth hydro-
spin (LS) species. The entrance of the substrate gen bond is donated by Gln3^Q to the carbonyl
(RH) into the protein pocket displaces the water group of the cysteine. These hydrogen-bonding
molecule and generates the five-coordinated ferric interactions are implicated in the stability of the
species (2). The result is that the iron pops out of enzyme and are thought to have an effect on the
the plane of the porphyrin, and thereby weakens generation of Cpd I from Cpd 0. Other interac-
the interaction of the d-orbitals with the ligands, tions in the distal side, for example, with Thr252
resulting in a narrow d-block. Consequently, the and Asp25i, are implicated in the proton relay
five d-electrons occupy the d-block in a high-spin mechanism that leads from 5 to 7.
Computational Approaches to Cytochrome P450 Function 49

U^C HoO
^2'
C02-(CH2)2

C02-(CH2)2

H3C' I ^7^^^CHCH2

ROH

RH RH
I Fe,iv_ ,m.
I I
L L
(Cpdl)

-H^O
r'
OH^
RH o' RH

H ' O 20
RH ^/ RH /

• Fe^,111. iFe^^
I * - ^
L e L
5

• ^ =Por

L = SCys-

Figure 2.2. The catalytic cycle of Cytochrome P450.

The next two sections describe the results of catalytic cycle of P450 with methyl mercaptide
theoretical calculations on the intermediates 1-7 (SCH3') as an axial ligand using the semiempiri-
in the catalytic cycle. Most of the literature in the cal SAMl method, which found good overlap
field up to the year 2000 can be found in a review between their results and experimental assign-
by Loew and Harris ^^ and Loew^^. Extensive ments. The five-coordinated Fe"^ complex 2 was
work of Glier and Clark ^^ modeled the complete found to exist in a quartet ground state, whereas
50 Sason Shaik and Samuel P. De Visser

the true one is a sextet. The semiempirically in 6, one of the TT* orbitals becomes a nonbonding
calculated Cpd I species is not well described, d^^ orbital, while the other remains a TT* MO and
having too short an FeO bond and hence a dififer- possesses antibonding interaction across the Fe-S
ent electronic structure compared with ab initio linkage. These alternative energy orderings of
methods and experimental characterization of the d-block orbitals, for complexes with different
this species as triradicaloid. As such, we do not ligand types, are shown in the insets in Figure 2.3.
describe details of the semiempirical results and A high-lying porphyrin orbital that acquires
defer the discussion until the methods become single occupancy in some of the species is a2^ that
suitable for treating iron compounds. is depicted in the center of Figure 2.3. In the pres-
Each of the intermediates 1-7 has several ence of the thiolate, the di^^ orbital mixes strongly
closely lying spin states due to the dense orbital with the ag-hybrid orbital on the sulftir. The por-
manifold of the iron-porphyrin system. As a typi- phyrin-proximal ligand mixture of the 2^2^ orbital
cal example of the important orbitals that figure in depends on the nature of the axial ligand. In con-
the catalytic cycle, we depict in Figure 2.3 d-block trast to thiolate that mixes strongly, an imidazole
iron and the highest lying porphyrin orbital for axial ligand mixes weakly with the porphyrin a2^
Cpd I (7). The five iron d-orbitals on the left orbital. Finally, an orbital that may play a role is
exhibit the well-known three-below-two splitting, the p^-type sulftir lone pair depicted in the far
much like the U and e sets in the purely octahe- right of the diagram and is labeled TTg.
dral symmetry. The thJee lower orbitals include Generally, QM modeling cannot describe the
the nonbonding 8(d^2_ 2) orbital, which is fol- complete enzyme, and one needs to truncate models
lowed by a pair of almost degenerate TT^^Q that mimic faithftilly the active species. Most of
(TT*^ and TT* ) orbitals made from the dxz and d^z these models truncate the side chains on the por-
orbitals of iron in antibonding relationship with phyrin and use porphine, while the proximal ligand
the ligand p^ and p orbitals. The upper two are a* is truncated either to thiolate (SH~), methyl mer-
orbitals that have strong antibonding relationship captide (SCH^), or to cysteinate anion (CysS~).
with the ligand orbitals; o-* is Fe-N antibonding, The experience is that SH~ gives results closer to
in the plane of the porphyrin ring, while a*2 reality than the methyl mercaptide (SCH3^) ligand as
is antibonding along the 0-Fe-S axis. Without revealed by QM/MM calculations^^. This choice is
a ligand bound to the distal position, as in 2 and 3, due to the hydrogen-bonding machinery that
or with a ligand that has only one p^ lone pair as stabilizes the thiolate and decreases its donor ability.

— o\yX 0*xy
a*72 j _ Gr*z2

TH • 4 - ^*yz s**^

Vr Vr
NtN V dxz 6(dx2.y2)
4f T
N^N
Cpd 0 (6)
6 (dx2_y2)

o Porphine Ligand
II
Fe 0*^2

Cpd I (7)
dxz 6(dx2.y2) ( ^
(5)

Figure 2.3. Orbital occupation of some critical species in the catalytic cycle.
Computational Approaches to Cytochrome P450 Function 51

To appreciate the role of thiolate in P450, it is when water is replaced by a ligand with a stronger
instructive to also look at related heme-containing field such as imidazole.
enzymes like HRP or catalase, where iron is coordi- This conclusion was contested by Green^^ who
nated to imidazole from a histidine side chain or to calculated the resting state of cytochrome P450
phenoxyl from a tyrosine side chain. using DFT and the B3LYP franctional. Green's
Apart from O2 and water as ligands, calcula- model system had a water ligated iron-porphine
tions have been reported on other small molecules and a methyl mercaptide proximal ligand. The
bound to iron-porphyrin complexes, among which sextet-doublet energy difference was calculated
are carbon monoxide^^ and NO^^' ^^. Calculations with a series of basis sets; the result was found
of metal-free porphyrin systems were reviewed to converge at the 6-311+G* basis set which
by Ghosh^^ and Ghosh and Taylor"^, while DFT predicted a doublet ground state. It was reasoned
calculations on oxo-iron porphyrin without axial that the sextet state that involves the electronic
ligand^"* and studies of oxo-manganese porphyrins configuration S^ TT*^I TT*^! a*2i a* ^ (Figure 2.3) is
with different axial ligands^^ were also reported. destabilized with basis set improvement, due
Another complex that was studied is the hydrogen to the increased Fe-S antibonding character of
peroxide ferric complex, Fe^"(H202), which in the the a*2 orbital. Thus, Green concluded that the
P450 cycle is thought to lead to decouplings^, doublet state is an intrinsic property of the resting
while in HRP it is thought to be an intermediate state of P450.
in the formation of Cpd P^' ^^. Green's study^"^, however, did not involve
geometry optimization. An early DFT study by
Filatov et al?^, used the pure ftinctional BP86, and
partial geometry optimization. The ground state
3.1. The Resting State (1) was found to be a doublet state that existed in two
conformations; in the "upright" conformation,
One of the most thoroughly studied intermedi- the water molecule points upward away from the
ates in the catalytic cycle of P450 is the resting porphyrin ring, while in the "tilted" conformation,
state (1). Experimentally, the ferric-water complex the water molecule forms hydrogen bonds to the
was characterized by electron spin echo envelope nitrogen atoms of the porphyrin ring. The water-
modulation (ESEEM) spectroscopy and found to porphyrin hydrogen bonds were found to stabilize
have a doublet ground state^^. Based on a previous the "tilted" complex by about 6.6 kcal mol~^
study^^ of the resting state of cytochrome c perox- Our present B3LYP/LACVP(Fe),6-31G(H,C,N,0,S)
idase (CCP), Harris and Loew^^' ^^ studied the calculations, shown in Figure 2.4^^, support this
state for P450 by an early phase of the QM/MM conclusion, but shows that after geometry opti-
method. They employed initial MM minimization mization, the energy differences are small, c.1.1
of the X-ray structure and subsequently used kcal mol ^ An additional frequency analysis of
INDO/ROHF/CI semiempirical methods with, both species showed that while the "tilted" con-
and without, inclusion of the electric field of the formation is a true minimum, the "upright" struc-
protein in the QM procedure. The electric field- ture is a saddle point since it has one imaginary
free calculations predicted that the ground state frequency, driving it back to the "tilted" form.
should be the sextet state, while the doublet state Clearly therefore, DFT calculations indicate that
was found to lie significantly higher (3.84 kcal the resting state has an intrinsic preference for a
mol~^). With the electrostatic field included, the "tilted" conformation due to the propensity of
doublet state descended below the sextet state the water ligand to undergo hydrogen bonding
by -1.59 kcal mol~^ The authors concluded that with a suitable acceptor. Within the protein
the LS ground state is due to the electric field of pocket, this interaction can be supplied by the pro-
the protein and is not an intrinsic feature of the tein side chains that will stabilize the "upright"
water complex. Some support for this conclusion conformation, which might become the ground
was provided by synthesis of a model ferric-water state or, at least be, in equilibrium with the "tilted"
complex with a thiophenoxide ligand^^, which conformer. The ESEEM results fit better the
exhibits a ground state with a sextet spin, whereas "upright" conformation. A QM/MM study using
the doublet state becomes the ground state only BP86 ftinctional for the QM subsystem was
52 Sason Shaik and Samuel P. De Visser

1, "upright" 1, "tilted"

0-^2^,2.533

= 2.018
0.099

Figure 2.4. Orientations of the water ligand in the resting state (1). Here and elsewhere the parameter A specifies
the deviation (in A) of Fe from the porphyrin plane, while r^^^ is an average distance of the four bonds.

performed by Scherlis et al?^ and Scherlis et al?^ mixes less strongly with the d-orbitals and the
using X-ray data without geometry optimization. corresponding a*2 orbital is sufficiently low to
The studies confirm the previous DFT findings stabilize the sextet ground state by virtue of its
that the doublet state is indeed the ground state. higher exchange stabilization.
However, the study started with the "upright" con-
former, so that the conformational question is still
open. Future QM/MM calculations with full 3.2. The Pentacoordinate
geometry optimization will be required to resolve Ferric-Porphyrin (2) and
this issue. Ferrous-Porphyrin (3)
The related resting state of peroxidase Complexes
enzymes, involving iron-porphine with water and
imidazole as axial ligands, was studied using Ogliaro et al^^ calculated the pentacoordi-
CASSCF(5,5) calculations^^ in which all the elec- nated ferric- and ferrous-porphyrin and quanti-
tronic configurations that can be generated by fied the "push effect" of the thiolate ligand, using
distributing the five d-electrons into the five B3LYP hybrid functional. Figure 2.5 provides
d-orbitals, were included in the active space. The optimized structures and energy separation of
lowest lying state was found to be the sextet spin some of the lowest lying states. At the UB3LYP/
state with 8' TT^I IT* i a*2^ o-* ^ configuration. LACV3P+* level of theory, the ground state
The quartet and doublet states were found to be of 2 was found to be the sextet state with
2.21 eV (51.0kcalmol-i) and 3.41 eV (78.6 kcal ci^2_ 2^ d^i TT* 1 a*2' a* ' configuration, whereas
mol~^) higher in energy than the sextet ground the quartet and doublet states were higher by 4.21
state. The doublet state is characterized by short and 4.23 kcal mol~', respectively. Experimentally,
Fe-0 and Fe-Nj^^.^^^^jg distances of 2.09 and the HS and LS forms of 2 are in equilibrium"^^' ^^
2.04 A, respectively. By contrast, the Fe-0 dis- which suggests that their energy separation is
tances in the sextet and quartet states are 2.32 and somewhat smaller than the value predicted by the
2.34 A, respectively. This is mainly the result of calculations, and is perhaps modulated by the
occupation of the a*2 orbital which is antibonding electric field or steric constraints in the protein
across the O-Fe-N- , axis. These results sug- pocket. Other complications in the protein pocket
are the presence of the substrate and the entropic
gested that the actual spin state would depend on
driving force due to the expulsion of a few
the protein environment that can control the spin
water molecules upon the formation of 2. These
state by imposing bond length restriction.
features will have to await an appropriate QM/
Comparison of the resting states of P450 vs
MM study.
those of peroxidase highlights the difference
between the thiolate and imidazole ligand. Reduction of ^2 fills the d^ orbital and
Thiolate mixes more strongly with the iron generates a quintet ground state, ^3 for the reduced
d-orbitals and hence the resulting a*2 orbital is pentacoordinated ferrous complex. Another quin-
raised in energy, making the sextet state less tet state with a doubly filled d^^ orbital lies only
favorable than the doublet. By contrast, imidazole 2.14 kcal mor^ higher, while the triplet d2_ 2^
Computational Approaches to Cytochrome P450 Function 53

2.285 (2.491) {2.<


2.484 (2.586) {2.

?i.N =2.005 (2.013) {2.091} y,.. =2.155 (2.031) {2.015}


^ = -0.195 (-0.237) {-0.432} ^ = -0.593 (-0.213) {-0.163}
22 (42) {%

AE : +4.2 (+4.2) {0.0} 0.0 (+9.4) {+14.1}

Figure 2.5. Optimized (UB3LYP/LACVP) geometries of 2 and 3.

(a)
(+1)
-Fe'L ^ -Fe'li- - -Fe'iL 2
I
4 I |51.4 HS
-49.3
(-1) ,, 1 ••
-152.3
G
103.0 I
HS

•Fe"- ^

OH2
(b) OH2
(+1) OH2 -Fe'^ 1
- F e ' l,1L11. -
I
T HS
I
-40.7
e I
(-1) OH2 -56.2
I \AEQM = 15.5
-141.8 -Fe'i- H ) OH2
I - F e ' L 1-
OH2 '^ 0
AEifield = 85.6 I
HS

Figure 2.6. Calculated (UB3LYP/LACV3P+*) push effect of the thiolate ligand (a) on the reduction energy
2 - ^ 3 , and (b) on the reduction of 1 ^ 1".

d^2 ^* 1 (y*i state lies 9.4 kcal mol~^ higher. filling of the d^ 2 orbital, which does not mix
The singlet state, 4c2_ 2^ dj^ '^%h ^^^^ consider- with the thiolate orbital, this large "push effect"
ably above the quintet ground state by 14.1 kcal originates from the electrostatic interaction
mol~^ between the negatively charged thiolate and the
The process ^2 -^ ^3 was found to be exother- iron-porphyrin moiety. To ascertain the origins of
mic by 51.4 kcal mol~^ (Figure 2.6(a)), while in the "push effect," the thiolate was replaced by a
the absence of the thiolate ligand, the reduction point negative charge placed at precisely the same
energy would have been exothermic by 152.3 kcal position as the thiolate"^^. As shown in Figure 2.6(a),
mol~^. Thus, the thiolate makes the pentacoordi- the resulting reduction energy for the pseudo
nated complex a much poorer electron acceptor by complex (—49.3 kcal mol~^) was almost identical
the "push effect" that is a measure of its electron to the pentacoordinated complex. It was therefore
donation capability toward the iron-porphyrin. concluded that during the reduction ^2 -^ ^3, the
It was reasoned"^^ that since the reduction involves "push effect" is indeed a pure electrostatic field
54 Sason Shaik and Samuel P. De Visser

effect and not of quantum chemical origin. The 3.4. The Ferrous-Dioxygen (4)
effect of the protein electric field was estimated and Ferric-Dioxygen (5)
by embedding the bare species in a medium with Complexes
a dielectric constant of £ = 5.7. This was found to
reduce the "push effect" from 103 kcal mol~^ for Early CASSCF calculations of a ferric-
the bare system to 33 kcal mol~ ^ for the embedded dioxygen species (4) with ammonia as an axial
system"*^. Although significantly reduced, still the ligand were carried out by Yamamoto and
"push effect" is substantial; it makes the reduction Kashiwagi"^^, using a minimal basis set and no
of ^2 selective such that only the reductase can geometry optimization. The lowest singlet state
perform it and thereby trigger the initiation of the was found to possess a major weight of 64% of the
cycle. Pauling configuration that involves coupling of
Fe^^ with the neutral O2 moiety in its singlet situa-
tion. The state also has some Fe"^02^ character due
to the mixing of higher configurations.
3.3. The Gating of the The first DFT calculations on 4 and 5 were
Catalytic Cycle reported by Harris and Loew ^^ and Harris et al.^'^,
using the BPW91 and BLYP pure functionals with
To understand the reason why it is that only the a basis set of double-^ plus valence polarization
pentacoordinated complex, 2, and not the water quality (DZVP); the two functionals gave virtually
complex, 1, is reduced during the catalytic the same results, and in good agreement with
cycle, Ogliaro et al^^ studied the reduction of the experimental data. The most stable form of
ferric-water complex ^1, with and without a thio- ferrous-dioxygen (4) is an end-on complex in
late ligand. The results are shown in Figure 2.6(b), accord with the experimental predictions'^^ while
and project once more that without thiolate, the the symmetrically bridged isomer was found to
ferric complex is a much better electron acceptor, be much higher in energy, by c.28 kcalmol"^
by c. 101.1 kcal mol~^ It was further determined Reduction of 4 to 5 resulted in elongation of the
by the same technique as above, that most of this F e - 0 and Fe-S bonds, while leaving the 0 - 0
energy effect (AEfj^j^), c.85.6 kcal mol~', comes bond length intact, albeit the 0 - 0 bond order
from the electrostatic field effect, and a small decreased from 1.20 to 0.87 (ref [26]). In agree-
part, c.15.5 kcalmol"^ (^^QJ^J) is contributed ment with its silent ESR behavior, 4 was found to
by QM mixing with the p^ orbital of the thiolate have a singlet ground state, but the triplet state to
ligand, which raises the electron accepting orbital lie only 1.1 kcal mol~* higher. By comparison, 5
7T*^(ref [40]). was reported to have a doublet ground state with
Comparison of the reduction energies in spin densities distributed over both oxygen atoms,
Figures 2.6(a) vs (b) reveals that the reduction of in agreement with ESR data. Electronic spectra of
the pentacoordinated complex is more exothermic both the ferrous- and ferric-dioxygen species,
than that of the ferric-water complex by 10.7 kcal calculated with the semiempirical INDO/S/CI
mol~^. This difference safeguards the resting state method"^"^, exhibit, in agreement with experiment,
against reduction by the reductase, such that a sin- a split Soret band. The corresponding Mssbauer
gle water molecule can gate the catalytic cycle. parameters of 4 and 5 were calculated too, and
Note however that the ligand that actually controls those for 4 show a good fit to experimental data.
this gating is the thiolate; it makes all the species Figure 2.7 compares the structures of U and ^5
poorer electron acceptors, leading thereby to as derived by us, using the B3LYP functional
a selective reduction of the pentacoordinated with the LACVP(Fe)/6-31G(H,C,N,0,S) basis
species by the reductase. Without the thiolate set^^ vis-d-vis the BPW91/DZVP results of Harris
ligand, all the complexes are such good electron et al.^^ in brackets. The structures show a general
acceptors that most reducing agents would have fit, with the exception of 4 that appears more open
reduced all the species with no selectivity what- in B3LYP compared with BPW91. Transforma-
soever. Thus, the property of a gated cycle by a tion of the DFT orbitals to natural orbitals for
single ligand (water) is achieved due to the "push 4 revealed that its electronic structure is the
effect" of the thiolate^^. open-shell singlet 8^ d^/ir* 1 TT'^Q^ configuration.
Computational Approaches to Cytochrome P450 Function 55

£= 1 8 = 5.7

46.5 kcal mol"^

48.1 kcal mol •1

Figure 2.7. Top: optimized geometries of ^4 and ^5. Values out of parentheses are taken from De Visser and
Shaik^^, while in parentheses from Harris et al.^^. Bottom: the reduction energies U -^ ^5 (ref. [36]).

The TT*^ orbital has a strong mixing with the predominantly the '7T*(0-0)-type orbital of the
corresponding TT and IT* orbitals of the dioxygen dioxygen ligand (Figure 2.3), as found by Harris
moiety, and as such, an appropriate description of et al.^^. In reasonable agreement with Harris and
M is a resonating mixture of the ferrous and ferric Loew^^, the reduction U ^ ^ 5 was found to
forms, Fe^^02 and Fe^^^02. This conjugation be endothermic, by 46.5 kcal mol~^ However, in
requires that the dioxygen moiety, in the Fe^^02 the presence of an electric field modeled by
form, will be in its singlet state, so that the empty a dielectric constant of e = 5.7, the reduction
'TT*(0-0) orbital can mix with the doubly occu- energy becomes exothermic by 48.1 kcal mol~^
pied d (Fe) orbital; as such there is a very low The effect of the protein environment on U
lying triplet state, ^4, only 1.1 kcal mol~^ higher and/or ^5 was studied initially by IVID simulations
than U (ref. [44]). The electronic structure of M of P450^^^ and P450^ryp (refs [46], [47]) using
suggests its origin from the excited singlet state of MM/MD calculations. The study showed that
3 and ^A state of O2. This is further supported by the ferrous-dioxygen species is stabilized by a
the fact that 4 has low-lying HS states, for example, hydrogen bond from Thr252 (ref. [46]). In the case
quintet and heptuplet states which originate from of the species ^5 of P450 p, Harris and Loew^^
the coupling of ^3 with the triplet and singlet states found that the distal oxygen of the twice-reduced
of O2. In view of the fact that both moieties of 4 species ^5 is linked through a hydrogen-bonding
have HS ground states, an interesting feature of the network involving neighboring amino acids, such
O2 binding must be the spin crossover process that as Ala24j and Ser24^, and several water molecules.
is yet to be properly elucidated by theory. This stable hydrogen-bonding network was impli-
A natural orbital transformation analysis cated as the root of the double protonation that
reveals that the odd electron in ^5 populates eventually converts 5 to Cpd I.
56 Sason Shaik and Samuel P. De Visser

3.5. The Protonation Mechanism doubly negative ^5 species and the incipient anion
of Ferric-Dioxygen (5) to of the protonating species. Indeed, the study
Cpd 0 (6) showed incisively that hydronium ions are not
needed to initiate the protonation of the twice-
BPW91 DFT calculations^^ showed that ^5 is reduced species, -^5, and ruled out any putative
an extremely strong base with a proton affinity protonation of the ferrous-dioxygen complex, U,
of 422 kcalmol~\ and as such it may undergo by the W519-W564-Ser246 array. This result is
protonation by a water molecule. This study was consistent with the large kinetic solvent isotope
pursued by Guallar et al.^^ who carried out effect^^ that was observed for the reduction of U
a QM(DFT)/MM investigation of the protonation to ^5, which indicated that the reduction of M and
mechanism, leading from 5 to Cpd 0 (6) in protonation of ^5 to Cpd 0, ^6, are nearly com-
P450g p. This was followed by a full quantum mensurate events. The MD study"^^ further showed
dynamics simulation of the proton transfer that the initial protonation by W519 is completed
through a one-dimensional profile. The study of within 500 fs and is the rate-determining step that
the QM subsystem used B3LYP with a mixed triggers a sequential protonation from W564.
basis set, and focused on three different protona- In a subsequent paper, Harris^' extended the
tion mechanisms, by: (a) a single water molecule DFT study (B3LYP/ LACVP**(Fe)-6-31G*(H,C,
(W519), (b) an array of two water molecules N,0,S)) of the protonation process and analyzed
(W519 and W564), and (c) an array of W519, its features by calculating proton affinities and
W564, and an ethanol that mimics the Ser24^ transition states (TS) for protonations by various
amino acid. The latter model is depicted in candidate acids. His studies showed, inter alia, that
Figure 2.8(a), and seems to be highly conserved in the hydronium ion can indiscriminately protonate
many P450 isozymes that exhibit sequestered both ^4 and ^5, and is therefore ruled out as the
array of water molecules hydrogen bonded to a source of protons. By contrast, serine, threonine,
polar amino acid residue near the protein surface. or their clusters with two water molecules (e.g.,
The computed energy profile"^^ changed grad- W519, W564 in P450g^yp) can protonate ^5, but are
ually from an endothermic one (+20 kcal mol~') only capable of donating a hydrogen bond to U;
(a) to an exothermic one (—10.7 kcal mol"^) for the protonation of the HS species "^5 encounters
(c) with a concomitant decrease of the barrier a high barrier despite its identical proton affinity to
to 1.8 kcalmol"^ The dramatic effect caused ^5; this was ascribed to the reduced negative
by the Grotthuss-like mechanism is very likely charge on its distal oxygen. Using an extended
due to the diminishing repulsion between the array of two water molecules and alanine, the

(a)

SER246 I ,
2.03
^ Water564
^1.79
^1.85
252
Water5i9 ^^

Figure 2.8. Protonation models of ^5 -> ^6 taken from (a) Guallar et al^^, (b) Kamachi and Yoshizawa"*^.
Computational Approaches to Cytochrome P450 Function 57

barrier for proton transfer to -^5 was found B3LYP studies of ferric peroxide, 6, and its
to decrease to 1.3 kcal mol~ ^ and the product of the proximally protonated isomer, 6-iso, shown
hydrogen-bonded array showed characteristics of in Figure 2.9. In both studies, 6 was found to be
low-energy hydrogen bonds that may contribute to the stable isomer by c.18.4 (23.3) kcal mol~^ and
the process facility. In this manner, the hydrogen- to possess a doublet ground state, labeled ^11 ^
bonding network in the pocket creates a gentle as (Fe"^), a symbol that denotes its singly occupied
well as spin-state selective protonation process. TT* orbital^^. An interesting feature of the ferric
An alternative and more potent protonation peroxide is the internal hydrogen bond between
mechanism was very recently studied by Kamachi the hydroxo proton and the nitrogen of the
and Yoshizawa"^^, who calculated a model fash- porphyrin ring^^. Application of an electric field
ioned after Vidakovic et al.^^, with two water mol- simulated by a dielectric constant of £ = 5.7
ecules sequestered between the acidic end, CO2H, fiirther shortened the Fe-S bond and the OH—N
of Asp25i and the side hydroxyl group of Thr252. hydrogen bond^^. Whether this hydrogen bond
This study involves both doublet and quartet states will or will not survive in the protein pocket is an
of 5, and the model is shown in Figure 2.8(b) interesting question. But already it is clear that the
alongside the model of Guallar et al^^. The proto- porphyrin is an internal base that may participate
nation is initiated by proton transfer from the in deprotonation/ protonation events.
CO2H group of ASP251 to the adjacent water that
fixrther transfers a proton to the water molecule
adjacent to the distal oxygen of "^'^5, which 3.7. Protonation of Cpd 0 and
completes the transfer and generates '*'^6 sponta- Formation of Cpd I (7)
neously. The Kamachi-Yoshizawa process was DFT calculations show that ferric peroxide,
found to be much more exothermic, —61.5 Cpd 0 (6), is a fairly strong base with a high proton
(—50.1) kcalmol~\ than the model of Guallar
affinity; PA = 334 kcal mol" ^ at BPW91 and with
et al.^^, as would be expected from the stronger
CH3S~ proximal ligand^^ or 330.1 kcal mol"^ at
acid that relays the initial proton. Although
B3LYP and with HS" as the proximal ligand"^^' ^l
barriers were also reported, one cannot avoid
Both studies found that the protonated species
the conclusion that there are multiple protonation
yields Cpd I (7) spontaneously without a barrier. In
pathways that must be taken into account via QM/
any event, the study of Davydov et al? shows that
MM with proper sampling.
mutation that replaces Thr252 does not prevent the
formation of Cpd 0, but prevents, or at least slows
3.6. Cpd 0: The Ferric Peroxide down, its subsequent protonation to yield Cpd I.
Complex (6) This suggests that the protonation mechanisms
that lead to 5 and 6 are different. The protonation
Harris and Loew^^ used BPW91 calculations mechanism is thought to occur from a hydronium
and subsequently Ogliaro et alP performed ion sequestered in P450^^^ by Asp25i ^^^ ^^252

1.517 1.494
(1.46) (1.41)

^6-\so

AE = 0.0 (0.0) 23.3 (18.4)

Figure 2.9. Optimized geometries of Cpd 0 (^6) and its isomer ^6-\so. Values out of parentheses are from Ogliaro
et alP, values in parentheses are from Harris and Loew^^.
58 Sason Shaik and Samuel P. De Visser

^SP25lT^ ASP251
•; 1.370(1.370)

I
o ^ ' 1 . 5 6 6 (1.560)
2.743 f^ THR252
(2.938), THR252 1.559(1.542);' •^•^^
•=0^^1.610
1.836(1.843)^ (1.572)
1.531 (1.543)X"'\
• L949 1.674(1.675)1
2.039(1.

2.884 (2.,

Cpd0[46(26)] Cpdlfiei)]

Figure 2.10. Hydrogen bonding set-up for protonation of Cpd 0 to Cpd I, from Kamachi and Yoshizawa^^.

(ref. [52]). A recent study of Kamachi and a hydrogen peroxide complex. The latter complex
Yoshizawa"^^ used a similar model to the one pro- is able to release H2O2 and return to the resting
posed by Vidakovic et al.^^ with two water mole- state by coordination with a water molecule,
cules sequestered between the carboxylate end, thereby leading to decoupling. Energetically, the
CO2, of Asp25, and the hydroxyl side chain of formation of the hydrogen peroxide complex from
Thr252, as shown in Figure 2.10. The protonation 6 is only 6 kcal mol~' less exothermic than that of
starts with a proton transfer from Thr252 to the dis- Cpd I from 6 (ref. [26]), such that the decoupling
tal oxygen of 6, followed by a departure of reaction is a serious competitor with the produc-
a water molecule that is trapped through hydrogen tive process that leads to Cpd I. Not much is yet
bonding to the Thr252 anion and the array of the known about this competition. Clearly, a future
two waters that are linked by hydrogen bonding to theoretical study is required to explore alterna-
the Asp25j ^^^^^' This mechanism predicts that the tive protonation pathways that can reveal the intri-
protonation of 6 and formation of 7 is exothermic cate behavior of the wild-type and mutant enzymes.
by 13.1 and 5.5 kcalmol"' for the quartet and
doublet states, respectively The entire protonation
process from ^'^S to ^"^7 by this Asp25,-Thr252
3.8. The "Push Effect" on the
machinery was calculated to be exothermic
by 74.6 (55.6) kcalmol"^ (ref. [49]). However,
0 - 0 Cleavage Process
the barrier was not calculated for this process. The thiolate ligand was implicated as a crucial
Alternatively, the protonation mechanism may be factor in the 0 - 0 bond cleavage process through
nascent from a hydrogen-bonding array similar to its "push effect" that leads to Cpd I (ref. [54]).
the one discussed by Guallar et al^^ and Harris^ ^ Ogliaro et al.^^ have addressed this issue by com-
This, to the best of our knowledge, has not been paring the proton affinity of 6 to a reference com-
addressed by Harris^ ^ plex without a thiolate ligand. The thiolate ligand
In principle, protonation of Cpd 0, 6, can occur was found to increase the proton affinity of ferric
on either the proximal or the distal oxygen of peroxide by 81 kcal mol~^ At the same time, the
the complex. Distal protonation results in the protonated reference complex devoid of thiolate
departure of a water molecule and generation of loses water spontaneously as well. Thus, the
Cpd I. Alternatively, proximal protonation gives "push" effect of the thiolate does not concern
Computational Approaches to Cytochrome P450 Function 59

the mechanism of the O-O cleavage as such, if anion devoid of its internal hydrogen bonding^ ^
there could exist a strong enough acid to protonate predicted that the third singly occupied orbital is
the ferric peroxide devoid of a thiolate ligand. The the p^ lone pair orbital on sulfur leading to "^'^Ilg
"push" effect is expressed on the thermodynamics (TT*^! TT* 1 iTg) states (Figure 2.3) with spin
of the protonation, and by raising the proton density almost exclusively on the sulfur while the
affinity by 81 kcal mol~^ the thiolate enables the porphyrin is closed shell. Other studies^^' ^^ that
protonation of 6 by moderate acids such as those used HS~ or cysteinate with its internal hydrogen-
that exist in the protein pocket. The roots of this bonding interactions, found that the third singly
"push" effect were analyzed and were found to occupied orbital is 2i2^ strongly mixed with the
consist of a combination of a field effect and an sulfur a-hybrid (see Figure 2.3); this occupancy
orbital effect"^^. The field effect originates in the leads to ^'^A.2^ states, with spin density distributed
electrostatic interaction of the negatively charged over the porphyrin and sulfur. It was found^''' ^^,
thiolate with the ferric peroxide moiety, while that with HS~ or cysteinate as ligands, the ^'^H^
the orbital effect results from the mixing of the electronic states are more than 5 kcal mol~^
a-hybrid of the thiolate with the a2y orbital of higher in energy than the ^'^A2y state, whereas
the porphyrin (see Figure 2.3), which raises the with mercaptide all the four states were condensed
energy of this orbital. to within 1 kcal mol~^ This difference is not only
Another aspect of the "push" effect that was academic but also has clear physical manifes-
addressed by Ogliaro et al.^^ is the putative reduc- tations. Had Cpd I been a ^Hg ground state, it
tion of 6 by electron transfer. It was found that would have been red, as the Cpd II species with
the reduction of 6 to 6 ~ is highly endothermic by closed shell porphyrin, while if the ground state
43.3 kcalmol"^, whereas in the absence of thio- had been ^'^^2\x ^ ^ ' *^^ compound would have
late such a reduction would be exothermic by been green.
35.1 kcalmol"^ This very large change in the These considerations and other findings,
reduction energy is mostly due to the field effect which showed that the nature of the state is highly
of the negatively charged thiolate. This can be dependent on the Fe-S bond length^^, prompted a
compared to the high proton affinity of 6, which is DFT study of the effect of the NH—S hydrogen
very high due to the same "push" effect. Thus, the bonding and the protein electric field on the
thiolate ligand endows ferric peroxide with selec- nature of Cpd I, using simple modeling of these
tivity to undergo protonation rather than accepting effects^^' ^^. Figure 2.11 depicts a typical result,
an additional electron. by comparing the key bond lengths and spin
densities (p) for the bare molecule, the molecule
in an electric field characterized by a dielectric
3.9. Cpd I (7)
constant, 8 = 5.7, and when the bare molecule
Initial DFT studies of Cpd I were done for the is coordinated to two ammonia molecules by
bare molecule, that is, in vacuum or gas phase con- NH — S hydrogen bonds. As can be seen, under
ditions, and led to controversial results regarding all conditions, the FeO moiety has two spins,
the electronic structure of the ground state. These while the third spin is distributed over the sulfixr
results showed sensitivity to the thiolate used to and porphyrin ligands in proportions that are
model the cysteinate ligand, as well as to the func- highly dependent on the conditions. In the bare
tional used to calculate the species. Nevertheless, molecule, the third electron resides more on
all the calculations agreed that the species is a tri- the sulfur than on the porphyrin, for example,
radicaloid with three singly occupied orbitals. Two p(Por) = 44% in the "^A^^ state. With just two
of these are the ir*^ and ir* orbitals, depicted NH — S hydrogen bonds, the unpaired electron
above in Figure 2.3, which appear in all calcula- shifts mostly to the porphyrin, and so is the situa-
tions including the CASSCF study of Cpd 11^^. tion in a polarizing electric field (mimicked by a
However, the various studies differ significantly in dielectric constant, 8=5.7). Another interesting
the description of the third orbital, depending feature of Cpd I is that whereas most bond lengths
on the manner in which the study models the thio- do not change significantly with the application
late proximal ligand^^' ^6, 35, 56-61 jj^g studies, of hydrogen bonding and polarity, the Fe-S
especially those using mercaptide^^ or cysteinate linkage gets shorter by almost 0.1 A and its bond
60 Sason Shaik and Samuel P. De Visser

dissociation energy increases significantly. These Cpd I irrespective of whether the proximal ligand
results led to the conclusion that Cpd I is a was HS~, CH3S~ or a more extensive chunk of the
chameleon species that can change its character cysteine loop. Thus, in accord with experimental
and electronic state in response to the environment results on the analogous Cpd I species of the
to which it must accommodate^^' ^^. enzyme chloroperoxidase^^, Cpd I of P450cam is
Recent QM(DFT)/MM calculations of Cpd I a doublet state and it corresponds to the "green
of P450^^^ (ref [19]) used B3LYP, a variety of species."
basis sets, three different thiolate ligand models, The QM/MM calculations also confirmed
and four different snapshots selected from the MD the chameleonic nature of Cpd I, as can be
trajectory after equilibration had been established gleaned from Figure 2.11. Thus, in the gas phase
(200 ps simulation). These calculations retrieved situation, the Fe-S bond was long and the third
the important NH—S hydrogen bonds, donated to spin was located mostly on the sulftir, whereas in
the sulfiir by Leu33g, 0^339, and Gln3^Q, and the protein environment, the Fe-S underwent
assigned Cpd I in a definitive manner as the shortening and the spin transferred to the por-
doublet ^A2y state with a very closely lying ^A^^ phyrin; in both respects, the gas phase situation
state; the same QM/MM description applies to the with HS~ was closer to the QM/MM results^^ than

(a)
^A2U('A2U) ^A2U('A2U) '*A2u('A2u)
E=1 £ = 5.7
1.651(1.648) 1.661(1.660)

2.581 (2.600) 2.496 (2.503)

p(FeO) 2.02 (2.09) 2.03 (2.12) 2.03(2.11)


p(Por) 0.44 (-0.50) 0.75 (-0.89) 0.65 (-0.74)
p(SH) 0.54 (-0.59) 0.22 (-0.23) 0.29 (-0.33)

(b) ^A2U('A2U)
p(FeO) 2.05(2.17)
p(Por) 0.67 (-0.82) 1.626(1.625)
p(S) 0.28 (-0.35)
Heme

Cys357 X ® Gly359

Gln360 side chain

Figure 2.11. Hydrogen bonding (NH—S) and polarity effects (e is the dielectric constant) on geometrical
parameters and group spin densities (p) of Cpd I. (a) Model calculations^^, (b) QM/MM calculations^^.
Computational Approaches to Cytochrome P450 Function 61

the more extensive proximal ligand model, for radical. In the gas phase and at infinite Fe-S
example, mercaptide or cysteinate anion. A very distance, |a> is much lower than |b>. However, at
interesting feature of the QM/MM study^^ is the the equilibrium distance in the gas phase, |b> gets
variation in sulflir/porphyrin spin densities and stabilized by electrostatic interactions and closely
Fe-S bond length, imparted by the NH — 0 = C approaches |a>, but is still above |a>. The mixing
hydrogen bond, donated to the carbonyl group of of the resonance structures will lead to a state that
the cysteine ligand by the side-chain Gln^^^. As is |a>-like with a preponderant S» character. This
this hydrogen bond was allowed to intensify and character in the gas phase for the molecule will
change gradually toward its X-ray position, so did depend strongly on the donor capability of the thi-
the porphyrin spin density increase, the sulfur's olate ligand; it will be larger for a model like
spin density decrease, and the Fe-S bond length CH3S~ and smaller for a model like HS~ that is a
gets shorter. Thus, fine-tuning the hydrogen- relatively poorer donor. In a polarizing electric
bonding interaction around the thiolate ligand fine- field and in the presence of NH — S hydrogen
tunes the electronic structure of Cpd I and its Fe-S bonds, the ion-pair structure |b> gets stabilized
bond length; Cpd I is indeed a chameleon species and descends below |a>; the mixed state is now
that will be different for different P450 isozymes. |b>-like and has a S:~Por'^ character. It is appar-
A simple valence bond (VB) model was used ent that this model predicts that as the hydrogen-
to account for this chameleon nature of Cpd I, bonding situation and strength of the polarizing
as shown in Figure 2.12^^' ^^' ^^. The electronic field increase so will the Por'^ character. It is also
structure of Cpd I can be constructed from two apparent from Figure 2.12(b) that by changing
resonance structures, |a> and |b>; |a> describes from S*-like to S:~-like, the sulfrir changes its
a thiolyl radical and a closed-shell iron-oxo por- Fe-S bonding from a weak one-electron bond to
phyrin, while |b> is an ion pair composed of a strong two-electron bond. Thus, the VB model
a thiolate anion and an iron-oxo porphyrin cation shows that Cpd I is a mixed-valent state and as

(a)
O

lb> gas-phase protein


0 — b
se a^.
/ — b
AE^lOOkcalmor' ^^^^—' '^'^—'-'
O la> + Xlb> lb> + X\a>
II S- -like For®* -like
la>
0
S
/
(b) gas-phase protem
O o
II

^ 4f... / 0
S
••+•••• se -.J^
one-electron bond two-electron bond
Figure 2.12. Modeling the influence of hydrogen bonding and polarity effects on the electronic structure of Cpd
I: (a) Valence bond mixing of the contributing structures, (b) The Fe-S bond orbital, its occupancy and the type of
the Fe-S bond.
62 Sason Shaik and Samuel P. De Visser

such will change its electronic structure and Fe-S density is spread over the porphyrin and cysteinate
bond length depending on the hydrogen-bonding groups. Kuramochi et al}^ found an energy gap of
machinery and the electric field of the protein 0.15 eV (3.46 kcalmol"^) between the ground
pocket that accommodates it; it will behave as a state ^A^^ and the excited "^A^^ states of a HRP
chemical chameleon. model Cpd I with imidazole as the axial ligand.
Many other Cpd I species for different Subsequently, Deeth''^ studied the same species
enzymes and model systems were studied, and it is and showed that the most stable isomer involves
worthwhile to mention some of these even if they saddling of the porphyrin that stabilizes the mole-
are not P450 species. Ohta et al.^^ performed cule by 2.5 kcal mol~^ No saddling was observed
DFT-B3LYP calculations with a methoxide axial for thiolate ligands with porphine or octamethyl
ligand and found a LS ground state (^A2y) that porphyrin^^. However, with m^^o-tetramethyl-
contained 64-69% unpaired spin density on the porphyrin, a significant saddling was observed^ ^
axial ligand and 22-33% on the porphyrin ring. even when the proximal ligand was thiolate.
It is most likely that being a good electron The saddling of the me^o-tetra-substituted-
donor, methoxide will endow its Cpd I with porphyrinated Cpd I species was interpreted as the
a chameleonic behavior, which has not yet been means to relieve the steric repulsion between the
studied. DFT calculations with the iron substi- meso substituent and the hydrogen substituents on
tuted by manganese^^ or by ruthenium^^ showed the a and p positions^ ^
that these systems have different ground states The influence of the neighboring amino acids
than Cpd I of P450, and therefore will show dif- on the stability of Cpd I for HRP was studied by
ferences in reactivity as shown subsequently by Wirstam et alJ^ using DFT calculations. Their
Sharma et al.^^. These differences do not arise model used oxo-iron porphyrin and an imidazole
from changes in the nature of the orbitals, shown ligand replacing HiSjy5, a formate anion replacing
above in Figure 2.3, but rather from the relative Asp235 and an indole group instead of Trp^^p all
energy of the orbitals being modulated by the these three amino acids are located on the proxi-
transition metal. In particular, in the ruthenium mal side of the porphyrin. In their optimized,
substituted Cpd I species with HS~ as a proximal geometry, the indole moiety (of Trpj^,) is proto-
ligand, the ground state involves Ru^ with a nated and the formate group (of ASP235) ^^ nega-
single unpaired electron in the ir*^ orbital labeled tively charged forming hydrogen bonds with
2n^^(Ru^)^^ This state was found to be 4.58 kcal both the imidazole and the indole groups. It was
mol~' lower lying than the ^J^^^ state. In contrast, found that in the HS state (S = 3/2), two spins
in the case of Cpd I with iron, the ^n_^,(Fe'^) state are located on the FeO unit, while the third one is
was found to be 22 kcal mol~' higher in energy shared between the porphyrin and indole groups
than the ^K^^ ground state. However, in the case 0.48 and 0.47, respectively. Once again, it is
of Cpd I (Ru) too, the '^''^P^2u ^^^^^^ exhibit a apparent that the porphyrin cation radical is eager
chameleonic behavior, and become the ground to share its hole with other good donors. Perhaps
states when medium polarity effect is taken into the chameleon behavior is general for Cpd I
account. Thus, the Cpd I(Ru) species offers a won- species, even when the proximal ligand itself can-
derful opportunity to tune the nature and identity not participate in electron donation to the "hole,"
of the ground state and possibly also the reactivity other, better donor moieties will take its role.
patterns, by changing the proximal ligand, by Green^^ calculated the low-lying electronic
substituting the porphyrin, and by changing the states of Cpd I for a catalase model, with
polarity of the medium^^' ^^. phenolate as the proximal ligand. He found that
Replacing the cysteinate axial ligand with the ground state had the LS TT*^' TT* 1 n^
either imidazole^^ or phenolate^^ models the configuration, with TT^ being a lone-pair orbital on
related enzymes HRP and catalase, respectively. the phenoxy ligand. Two spins were located on the
In contrast to cysteinate, an imidazole ligand FeO moiety and the third almost exclusively on
hardly interacts with the porphyrin a2^ orbital. As the phenoxy ligand. A hydrogen bond donating to
a result, the spin densities of the singly occupied the oxygen of the phenolate ligand, or the place-
dL^^ orbital in Cpd I(HRP) are primarily located on ment of cationic species that mimic the presence
the porphyrin ring, while in Cpd I(P450), the spin of a charge-relay system in the protein, caused
Computational Approaches to Cytochrome P450 Function 63

a significant change and shifted half of the involves the acidic CO2H proton of ASP251 that is
spin density from the phenoxy Hgand to the transferred via an array of two waters. Protonation
porphyrin^^. These results are similar to the ones of ferric peroxide species, 6, appears, however,
obtained for Cpd I of P450i9' ^^^ ^^ and indicate to proceed by a gentler machinery^^ with exother-
that the chameleon concept may well be a general micity of c. 13.1 and 5.5 kcal mol~^ (Figure 2.10),
paradigm for heme enzymes. respectively, for the quartet and the doublet states.
These multiple protonation pathways suggest very
strongly that both the doublet and the quartet
3.10. What Makes the Catalytic states of Cpd I may be formed separately, and
Cycle Tick? A Summary those mutations may affect the production of the
two states in a different manner.
Two factors, mentioned above, emerge from
the calculations to strongly influence the catal3^ic
cycle: one is the "push" effect of the thiolate and
the other is the hydrogen-bonding machinery that
4. MM and MM/MD Studies of
is involved in protonation mechanisms of 5 and 6 P450 Reactivity Aspects
as well as in stabilization of Cpd L The "push"
effect is associated with the strong electron donor This section reviews MM/MD theoretical
property of the thiolate ligand. The calculations of work, which addresses the entrance of the sub-
Ogliaro et al.^^ showed that the "push" effect strate to the pocket, its binding, and the exit of
is responsible for gating the cycle by a single the substrate. Much theoretical work that rely on
molecule of water; for the O-O cleavage; for the quantitative structure activity relations, QSAR, is
preference of the twice-reduced species 5 and the not reviewed, but can be found in the authoritative
ferric peroxide Cpd 0 species, 6, to undergo pro- treatment of Lewis^^.
tonation rather than reduction; and for the propen-
sity of Cpd I to participate in hydrogen abstraction
or bond-making processes over electron transfer 4.1. Studies of Substrate
process. Without the thiolate ligand or with one Entrance, Binding, and
that is a much lesser electron donor than thiolate,
Product Exit
the resting state, as well as 5 and 6, would have
been prone to reduction, the O-O bond cleavage P450 enzymes usually have an active-
process would have been highly endothermic, and site pocket that is equipped with a substrate
Cpd I would have been an extremely powerful binding and 0 = 0 cleavage machineries^"*' ^^.
electron acceptor. Thus, the thiolate creates selec- Figure 2.13 shows the QM/MM calculated cam-
tivity toward reduction and thereby contributes to phor within the pocket ofVASQ^^^, in the presence
a stable cycle with a tightly gated reducibility and of Cpd l'^. In accord with experiment, the calcu-
basicity of the various species. lations reveal that two amino acids participate in
The calculations of Guallar et al.^^ and of the substrate binding; Tyr^^ holds camphor by an
Harris^ ^ demonstrate that the hydrogen-bonding OH — 0 = C hydrogen bond and Val295 interacts
machinery provides the means for a gentle protona- with the bridge methyl groups of camphor
tion that can protonate the twice-reduced species, 5, and thereby sequesters the substrate. Other P450
without touching its precursor ferrous-dioxygen isozymes have their own specific machineries and
complex, 4. This gentle machinery awaits, there- still others have larger and less selective pockets.
fore, patiently the second electron transfer and, Substrate access to the pocket, binding, and prod-
hence, ultimately enables the generation of Cpd I. uct exit have been probed by a variety of experi-
The study of Kamachi and Yoshizawa"^^ offers an mental techniques, and have been theoretically
alternative protonation mechanism that is more studied by means of docking and MM/MD
potent and exothermic than the one advocated by simulation techniques of various types.
Harris^ \ and which applies to both the doublet Docking calculations followed by MM/MD
and the quartet states of 5. This mechanism is and Monte Carlo simulations on the catalytic
based on a proposal of Vidakovic et alP and metabolism of the insecticide carboftiran by
64 Sason Shaik and Samuel P. De Visser

0=0
Cleavage

Figure 2.13. The active site of P450 showing functions of various residues.

P450^^j^ were performed by Keserii et alP^ and to reproduce their set of experimental data about
Keserii et alP^\ the tight binding of carbofuran by the enzyme function. A successful strategy was
hydrogen bonding to Tyr^^ and its steric confine- found to require, inter alia, active site plasticity
ment by Val247 and Val295 were found to fit the and considerable movement of the substrate,
preferred regioselectivity and stereospecificity of which thereby enable substrate positioning and
hydroxylation at the C3 atom of carbofuran. catal)^ic function. Other studies revealed that
Cavalli and Recanatini^^ used docking simula- some of the substrate motions are more con-
tions to study the selectivity of P450 inhibitors strained than others and depend on the topography
that carry imidazole groups, and found that the of the pocket and the substrate. Thus, the experi-
inhibitors bind to the heme via the lone pair of mental results of Atkins and Sligar^^ show
nitrogen in the imidazole group. This theoretical that during camphor hydroxylation by P450^^j^ the
approach, and similar ones not reviewed here, hydrogen is initially abstracted from both exo and
tacitly assumes that the preferred position of the endo C-H bonds, but the process only produces
substrate vis-d-vis the active species can foretell the exo alcohol product. This result indicates
the regio- and stereospecificity of the monooxy- that after the hydrogen abstraction, the alcohol
genation process, as well as efficacy patterns of formation step is sufficiently fast to reveal the
inhibitors. restriction of the camphor by its binding in the
Other approaches, which combined experi- pocket. Similarly, MD simulations and experi-
mental and theoretical studies, revealed some mental studies^^ of the kinetic isotope effect
generalities on binding modes and motions within (KIE), for hydroxylation of xylenes and 4,4'-
the pocket. NMR results^^ of perdeuterated dimethylbiphenyl, having equivalent, but isotopi-
adamantane in P450^^^ indicate that the substrate cally distinct, methyl and deuterated methyl
is conformational^ mobile on the timescale of the groups indicated that the mode that switches the
enzymatic turnover. This mobility was further position of the two methyl groups is restricted in
revealed by docking studies^ ^ of various substrates proportion to the distance between the groups.
and inhibitors of P450^^j^. It was found that the This restriction masks the intramolecular KIE
general strategy of rigid docking, which seeks value for 4,4'-dimethylbiphenyl for which the dis-
substrates that fill the active site completely, fails tance is 11.05 A, and the KIE drops from c.7-10
Computational Approaches to Cytochrome P450 Function 65

to c.l.l. In summary, the above studies show that 4.2. MM and MM/MD Studies of
the outcome of a given experiment depends on Regioselectivity
the inteq)lay of the rate constants for substrate
binding, de-binding, monooxygenation, and tum- An MD simulation technique was applied by
bling. Being "free" or "restricted" has a relative Audergon et alP to analyze the regio- and stereo-
meaning depending on the timescale of the selectivities of hydroxylation of the {\R)- and
various processes. (l»S')-norcamphor by P450^^j^. X-ray structures of
Still other approaches use MM/MD simulation the substrate-enzyme complex for the two enan-
to study the dynamics of various processes associ- tiomers of camphor showed that {\R) is oriented
ated with the water content of the protein pocket with its C5 atom pointing toward the heme iron,
and the entrance and exit of substrates therefrom. whereas (1*S) exhibits significant disorder. Despite
The hydration of the protein cavity was studied by the different substrate-binding conformations of
Helms and Wade^"^ using MD simulations of the the two enantiomers, both are known to give
substrate-free P450^^j^ followed by thermody- exclusively exo-C5-hydroxylation. To resolve this
namic integration. It was found that although apparent inconsistency between the two sets of
the cavity could hold, in principle, 10 water experimental results. Das et al?^ performed MD
molecules, the thermodynamically most favorable simulation on the substrate binding of these two
water content in the pocket was 6 molecules; most camphor enantiomers to P450^^^. The results^^ for
of them occupied the site of the sixth axial ligand both enantiomers revealed the strong orienting
(distal ligand) and one was coordinated to Tyr^^. effect of the hydrogen bond to Tyr^^. However,
The water molecules in the pocket were spread while the {\R) enantiomer gave one stable struc-
over a larger volume than in bulk water, and were ture, the {IS) enantiomer had greater mobility
theiefore more mobile than bulk water molecules. in the active-site pocket. In addition, the {\R)
MD simulations of the substrate entrance and exit enantiomer was found to orient with its C5 atom
channels of P450^^^, VA5%y^_^, and P450^^yp were pointing toward the heme iron, whereas this was
studied by Ldemann et al?' ^ and Winn et alP. not the case for the (1»S) enantiomer. These differ-
In all cases, the major access channels were found ences accounted for the X-ray structural findings.
to coincide with the ones predicted from crystal- To address the apparent inconsistency between the
lographic data based on thermal fluctuation fac- experimental X-ray and reactivity data, the simu-
tors (B factors) near the F/G loop and adjacent lation was repeated with a water molecule as the
helices. All these mechanisms involve backbone sixth ligand. The presence of the water molecule
motions and rotations that are specifically tailored was found to reorient the {\S) enantiomer with
to the physico-chemical properties of the sub- a C5 contact to the heme. This result was inter-
strate. In P450^^^, the channel is typified by small preted by the authors as a resolution of the
backbone displacements (1.8-2.4 A) and aromatic inconsistency, based on the contention that regio-
side-chain rotations of PhCg^, ^^Qx9V andTyr29. In selectivity ultimately depends on the orientation
P450gj^ 3, the positively charged Arg^^ located in of the substrate vis-a-vis the ferryl oxygen of
the entrance of the channel makes a salt-link that Cpd I. The same technique was employed for
guides the negatively charged substrate via its the monooxygenation of styrene by P450^^
carboxylate group, while in P450 p, the Arg^g^ (ref. [87]) where a good fit was obtained between
residue rotates and, by making intraprotein hydro- product distribution and the docked conformation.
gen bonds, gates the channel opening. Will such MD simulation studies by Harris and Loew^^
entrance/exit studies eventually account for the were used to rationalize the regiospecificity of
specificity of the P450 isozymes, as hoped by the hydroxylation of camphor and a variety of other
MM modeling community? It is a question that substrates. A series of trajectory calculations were
merits a proof of principle. Should this turn out to performed for the enzyme-substrate interactions,
be the right approach to the problem, then all the and these results were coupled with relative
chemical details of P450 activation would be stability of the organic radical intermediates
immaterial to its action. As shown below, this is to predict the product distributions. In a recent
certainly not the case. paper, Park and Harris^^ employed an integrated
66 Sason Shaik and Samuel P. De Visser

modeling approach that involves comparative been implicated as a second oxidant that functions
modeling (sequence and SCR alignment), a de alongside Cpd I, or in its absence, for example, in
novo loop construction and MD equilibration, to mutant enzymes where the 0 - 0 cleavage machin-
reconstruct a model of P4502gi of sufficient accu- ery has been impaired^^' ^^. Recent results on the
racy to ascertain the geometric determinants of mutant enzyme of P450^^^ (ref [92]) show that
diverse substrate metabolism via configurational the mutant P450^^^ (T252A), where the threonine
sampling techniques. Energy-based docking was that is responsible for the efficient protonation
shown to be an adequate predictor of binding machinery is mutated to an alanine, does not
modes correlated with experimentally deduced hydroxylate camphor, but does epoxidize cam-
metabolites. An MD-configurational sampling phene, albeit much less efficient than in the wild-
based on the low-energy docked configurations type enzyme. It was postulated that, in the absence
was found to be a more accurate predictor of geo- of Cpd I, Cpd 0 was the likely oxidant but that
metric factors. In this manner, it was possible to it is a much less efficient oxidizing species than
screen many substrates and locate the lowest Cpd I. The next few sections outline the results of
energy enzyme-substrate complexes. Assessment QM and QM/MM calculations on some of the
of the relative hydroxylation efficiency of three major reactions of P450: alkane hydroxylation,
prototypical substrates at their various functional alkene epoxidation, benzene hydroxylation, and
groups was carried out by combination of MD sulfoxidation. These reactions were studied using
sampling, of the docked configurations, and an Cpd I as the electrophilic oxidant. Two reactions,
energy criterion of the relative DFT-calculated ethene epoxidation and sulfoxidation, were stud-
energies of the radical intermediates produced in ied with both Cpd I and Cpd 0.
the reaction by hydrogen abstraction (see mecha-
nisms below). The relative energies of the radicals
were found to be good predictors of the relative 5.1. Reactivity of Cpd I: General
barriers of the C-H abstraction step. In several Considerations of the Origins
instances, these workers found that while equiva- of Two-State Reactivity
lent geometric exposure of metabolical sites (TSR) of Cpd I
occurred, an accurate prediction of the metabolite
pattern could be made only by means of electronic As seen already, Cpd I is a triradicaloid with
and energetic factors deduced from DFT. singly occupied TT*^, TT* and 2i2^ orbitals and,
In summary, investigations of P450 mechanis- hence, has a virtually degenerate pair of ground
tic problems by reliance on the modes of substrate states (^'^^2\)' ^^ such., it is expected that at
entrance and binding as the determinants of all the least these electronic states will participate in the
subsequent chemistry, while being a tempting and reactions, and will lead thereby to two-state reac-
an economical approach to the problem, are, in tivity (TSR)^^~^^. In TSR, each state may produce
our view, not well founded. Importantly, such its specific set of products with different rate-
approaches miss the crucial factors concerning the constants, regio- and stereoselectivities and lead
electronic structure determinants of the processes, thereby to apparently controversial information
as discussed in the rest of the review. The recent when viewed through the perspective of single-
results of Park and Harris^^ support this conclu- state reactivity (SSR). It is our contention that
sion and highlight the crucial nature of the funda- TSR resolves much of the controversy that has
mental mechanistic investigations by means of typified the P450 field of reaction mechanism
QM calculation, as reviewed in the remainder of in recent years^^, and opens new horizons for
this chapter. reactivity studies.

5. QM Studies of P450 5-2. A Primer to P450 Reactivity:


Reactivity Patterns Counting of Electrons
As a prelude to the reactivity discussion, it is
Cpd I is considered to be the primary reactive worthwhile to appreciate an important generaliza-
species of P450 enzymes. However, Cpd 0 (6) has tion, namely that the synchronous oxene insertion
Computational Approaches to Cytochrome P450 Function 67

by Cpd I is a forbidden reaction^^' ^^, and that we active orbitals, the a^^ orbital that figures in
expect, therefore, to deal with essentially nonsyn- hydroxylation and TT^^ that is important for epoxi-
chronous processes even if some of the mecha- dation. Thus, initially Cpd I involves Fe^^ and
nisms may turn out to be effectively concerted. porphyrin radical cation (Por^*), that is, the effec-
To assist us in keeping track of the electron tive oxidation state is Fe^. As Cpd I and the
count during this formal "two-electron oxidation" substrate make one bond (O-H or O-C), a single
process, we present in Figure 2.14, an oxidation electron shifts from the appropriate orbital of
state-orbital occupancy diagram that follows the the substrate (a^^ or TT^^) to either the 3i2u orbital
electronic reorganization and accounts for the for- of the porphyrin (1), or to the ir*^ orbital of the
mal oxidation states of Cpd I and the substrate at FeO moiety (2), leaving a singly occupied orbital
various phases of the process. The substrate is labeled as (f)^ on the substrate. Now, the effective
chosen to be one that can undergo hydroxylation oxidation state of the heme species is reduced to
or epoxidation and is symbolized by its two main Fe^^ (either Fe^^Por^* or Fe^^Por) accounting for

Cpd I (Fe^^Por+') (CH2=CHR / CH3-H)

0*2:2 ^ ^ ^

^i- ^cc (<^C-H)


v-j— 4""*''^ Jill.
6 ^ : ^ ^ "

-j—^C

e.g. o" ~ '


-H-H-41..:
"^^ '^*^^

- k - l 6-H-
4'2Fe"ipor+'

^ ' xy •
0*^2 I

± t -H- a2u

2Fe"¥or

Figure 2.14. Oxidation-states and orbital occupancy diagrams in various stages of alkene epoxidation and alkane
hydroxylation. Occupations within parentheses show the alternative spin arrangement, of the odd electron, in the
corresponding low-spin state.
68 Sason Shaik and Samuel P. De Visser

the first oxidation equivalent. In the second phase, from bicyclo[2.1.0]pentane had a finite, albeit
the substrate forms a second bond with the oxo short, lifetime. Everything looked fine for the
group of the heme, and a second electron is rebound mechanism until Newcomb et al.^^^ ^^^
shifted from the singly occupied substrate orbital used their ultrafast radical clocks to determine
(|)^ to the heme to populate either the a*2 orbital, radical lifetimes. Figure 2.15(b) depicts a typical
which results in a quartet state of the product probe substrate used by Newcomb and its
complex, or to the ir*^ orbital of Fe^^Por or the ^2^ rearrangement pattern; the corresponding lifetime
orbital of Fe"^Por"^*; the latter two options give the is determined from the inverse of the rate constant
doublet state of the product complex. The last step {k^ of radical rearrangement and the ratio [R/U]
accounts for the second oxidation equivalent, and of rearranged to unrearranged alcohol products.
the effective oxidation state of the heme is ftirther Using this method and determining only those
reduced to Fe"^. [R/U] quantities that do not involve carbocation
rearrangement, the resulting lifetimes quantified
by Newcomb were in the order of 80-200 fs
5.3. Alkane Hydroxylation (ref [100]). Since these lifetimes are too short to
The mechanism of alkane hydroxylation is correspond to a real intermediate, Newcomb
called the "rebound" mechanism. It transpires concluded that radicals are not present during
via an initial hydrogen abstraction, followed by the reaction and questioned the validity of the
rebound of the alkyl radical onto the oxygen of rebound mechanism.
the iron-hydroxo species, Figure 2.15(a)^^. This This problem was taken on by the Jerusalem
mechanism accounts for two key observations: group who used DFT (B3LYP) computations to
(a) a small but detectable amount of stereochemi- model the mechanism of methane hydroxylation^^'
cal scrambling, and (b) a large KIE due to replace- 101-103^ ally lie hydroxylation of propene*^"^' ^^^, and
ment of the hydrogen by deuterium in the C-H recently also of camphor hydroxylation by QM
bond undergoing hydroxylation. The first meas- and QM/MM calculations^^. In all these cases, we
urement of radical lifetime by Ortiz de Montellano could not locate a TS for a concerted oxene inser-
and Steams^^ indicated that the radical derived tion because the process possesses barriers that

(a) R-H
OH ^OH

IV. © • • Fe'^ .Fe"l.


I
L

(b)

Figure 2.15. (a) The rebound mechanism, (b) Derivation of the apparent lifetime (T ) of a putative radical
intermediate from the ratio of rearranged (/?) to unrearranged {U) alcohol products produced from P450
hydroxylation of a substrate probe.
Computational Approaches to Cytochrome P450 Function 69

are too high and transition structures that are A typical reaction mechanism is shown in
not real TSs; for example, they are second-order Figure 2.17, where one can see the doubling of
saddle points. The lowest energy mechanism was the profile due to the HS and LS states. The reac-
found to involve a hydrogen-abstraction like TS tion pathway involves three phases: (a) a C-H
(TS^), as exemplified in Figure 2.16 for camphor abstraction phase that leads to an alkyl radical
hydroxylation^^' ^^^. However, the calculations coordinated to the iron-hydroxo complex by a
reveal, as conjectured above, that the mechanism weak OH—C hydrogen bond, labeled as ^-^Cj.
involves TSR nascent from the degenerate ground (b) an alkyl (or OH) rotation phase whereby the
state of Cpd I that was modeled by the simplest alkyl group achieves a favorable orientation for
system (with a porphine macrocycle and HS~ as a rebound, and (c) a rebound phase that leads
proximal ligand). Subsequent studies of ethane to C - 0 bond making and the ferric-alcohol
and camphor hydroxylation by the Yoshizawa complexes, "^'^R The two profiles remain close in
group"^^' 10^-111 and of methane hydroxylation by energy throughout the first two phases and then
Hata et al}^'^, used porphine macrocycle and bifurcate. Whereas the HS state exhibits a signifi-
CH3S~ as a proximal ligand, and arrived at basi- cant barrier and a genuine TS for rebound, in the
cally the same conclusion, that the mechanism is LS state, once the right orientation of the alkyl
typified by TSR. group is achieved, the LS rebound proceeds in a
virtually barrier-free fashion to the alcohol. As
such, alkane hydroxylation proceeds by TSR, in
which the HS mechanism is truly stepwise with
a finite lifetime for the radical intermediate,
whereas the LS mechanism is effectively con-
certed with an ultrashort lifetime for the radical
intermediate. A recent study of camphor hydroxy-
lation"^^ identifies a rebound TS for the LS
process; this TS has a barrier of 0.7 kcal mol~^ but
is merely the rotational barrier of the camphor to
the rebound position.
By referring to Figures 2.15(b) and 2.17, it is
possible to rationalize the clock data of Newcomb
in a simple manner. The apparent lifetimes are
determined from the rate constant of free radical
rearrangement and the ratio [R/U] of rearranged
Figure 2.16. Camphor hydroxylation high-spin TS. to unrearranged alcohol product, assuming a

TSabs

Hydrogen abstraction Alkyl rotation Alkyl rebound

Figure 2.17. A two-state reactivity potential energy surface for alkane (R-H) hydroxylation by Cpd I.
70 Sason Shaik and Samuel P. De Visser

single-state reactivity (SSR). However, in TSR,


HO
the rearranged product, R, is formed only on the
HS surface, while the unrearranged product, U, is A- ,XHRR' ,
A^r
A,,s^^'CRR'
Xr
formed on the LS and possibly also on the HS sur- 0
face. Therefore, the ratio, [R/U], is associated with
m
Ar R, R' \U/R]
the relative yields of the HS vis-d-vis the LS reac- P-CF3 H, H 4
tions and not with the radical lifetime as such^^' ^^. Donor p-H H,H 4.3
Ability P-CF3 H,CH3 12
A simple TSR scheme leads to the following p-H H, CH3 25
expression for the ratio of the real to the apparent p-H CH3, CH3 >100
lifetimes:
Figure 2.18. Experimentally determined ratios of
unrearranged to rearranged product of various probes.
The probes are arranged from top to bottom in order of
V^L(TSR)/T, {[U/R](l+F)}
>1, increasing donor ability.
{[U/R]-F}
F= [LS/HS], (1)

where the quantity F is the relative yield of the LS


behavior of the LS and HS states"^^. However,
to HS reactions. It is clear that the real lifetime of
these calculations reverse the ordering of the bond
the radicals on the HS surface is longer than the
activation ^'^TS^ species from "LS-below-HS" as
apparent lifetime in proportion to the value of F,
in Figure 2.17, to "HS-below-LS." In addition, the
the LS vis-d-vis HS yield. Since in the calculations
LS pathway involves Fe"\ whereas by contrast,
the LS bond activation barrier is lower than the cor-
the HS pathway is Fe'^ type. We think that this
responding HS barrier, the F is larger than unity^^;
inversion of the HS-LS ordering originates in the
in the case of allylic hydroxylation, it reaches val-
powerful electron donor property of the CH3S~
ues of the order of 10 when the effect of the protein
ligand, such that its gas phase calculations do
electric field and hydrogen bonding are taken into
not represent as well as the HS~ ligand the actual
account^ ^^' ^^^. As such, the apparent radical life-
situation of the cysteinate within the protein
times will be unrealistically short compared with
pocket'^. Thus, much the same as in the case of
the real lifetimes. Furthermore, analysis of theCpd I where the use of CH3S~ as a proximal
rebound process itself^^' ^^' ^^^ showed that the
ligand in a gas phase calculation leads to a wrong
quantity [U/R] should increase significantly as assignment of the ground state (as a ^Hg state),
the alkane and its derived radical become betterthis ligand also misrepresents the LS-HS energy
electron donors. Therefore, in such a series, the
difference of the bond activation TS^ species. In
[U/R] quantity gradually increases, such that atfact, QM/MM calculations of camphor hydroxyla-
some critical donor ability of the radical when the
tion by P450^^^ (ref [76]) lead to a TS-situation
HS rebound barrier altogether vanishes, the [U/R]
of LS below HS, in agreement with the model
quantity converges to infinity, and the apparentstudies of Ogliaro et al.^^- '^^ and De Visser
lifetime becomes strictly meaningless. Such a trend
et al}^^' '^^ using HS~ as a proximal ligand.
has been observed in the series of probe substrates Figure 2.19 displays bond activation ^"^TS^
(^ra«^-alkylarylcyclopropanes) used by Newcomb structures for methane, ethane, propene, and cam-
et alJ^^ where the substrate, which is the best donor
phor hydroxylation. It is apparent that irrespective
and which leads to the best donor radical, exhibits
of the alkane and proximal ligand model, all the
virtually no rearrangement. This Newcomb series TSs exhibit an almost linear O—H—C triad of
is shown in Figure 2.18, which arranges the sub-atoms. These species closely resemble the genuine
strates in the order of increasing donor ability and
hydrogen abstraction TSs by alkoxyl radicals, one
displays the corresponding [U/R] quantity that of which is also displayed in the figure. The com-
exceeds 100 for the best donor situation. puted KIE {T = 300 K) values of the P450 TSs
Calculations of Yoshizawa et al} usmg range between 5.1 and 10.5 for the various sub-
CH3S as a proximal ligand retrieved the TSR strates and models"^^' ^^^' ^^^' ^^^ and are in good
scenario, and recently also the rebound barrier agreement with experimental values^^' ^^^.
Computational Approaches to Cytochrome P450 Function 71

1.500(1.510)
1.093 (i.r-^-^^ " 1.337(1.244)-^^, -
1.257 (L318)-H-^ 172.0
1.744 (1 797^T (170.6)
1.787(1.800)"

2.488 (2.476)

^ S H (^TSH) ^ S H (^TSH)
RH = CH4 RH = C3H6
4TSH
RH = Camphor
1.359 (1.404)-^ ^^^ ^fMf^Q\
1.183 (1.143)-^^^^-^ ^^^^-^^

^SH(2TSH)
RH = C2H6
T S H (CH30* PhCH3)

Figure 2.19. Hydrogen abstraction transition states (TSs) for some model reactions. The structures for methane
and allylic hydroxylation are taken from Ogliaro et al.^^' ^^^ and De Visser et al}^^, the structure for H-abstraction
from toluene from Ogliaro et al}^^, while for ethane hydroxylation from Yoshizawa et al}^^. Key geometric
parameters outside parentheses correspond to the high-spin TS, while those within to the corresponding low-spin
structure.

The hydrogen abstraction barriers in alkane Table 2.1. Hydrogen Abstraction Barriers
hydroxylation shown in Table 2.1 exhibit a high
sensitivity to the donor property of the alkane Substrate AEf A(E+ZPE)t AHi AGt
and the C-H bond energy. They range from 26.7,
26.5 (HS, LS) kcalmol"^ for methane down to CH/
13.5 kcalmol"^ for allylic hydroxylation. This HS 26.69 22.76 22.55 31.74
trend was analyzed and shown to conform to LS 26.54 22.31 21.68 32.35
hydrogen abstraction and oxidative character of the
bond activation step, as outlined above in the oxi- HS 19.4
dation state-orbital population diagram in Figure LS 16.2
2 1462, 105 Another feature in Table 2.1 is the CH2=CH-CH3^
reduction of the hydrogen abstraction barrier when HS 13.53 10.63 10.92 21.21
zero point energy (ZPE) is included. This arises LS 13.52 10.83 11.28 21.35
due to the loss of the ZPE for the C-H bond that is Camphor^
cleaved in the bond activation ^"^TS^ species. HS 18.8
Another feature in Table 2.1 is the very significant LS 17.3
entropic contribution to the free-energy barrier. Camphor^
Much of this arises due to the loss of translational HS 18.0
LS 20.2
and rotational degrees of freedom and structural
stiffening in the ^'^TS^ species. Substrate binding
Notes:
within the enzyme is, to a large extent, entropi- ''Ogliaro et al.^^.
cally driven, because it causes expulsion of the ^Yoshizawa e? a/.'".
water molecules from the binding pocket. Since ^De Visser e/a/.'^^
'^Cohen et alP^.
the hydroxylation begins with the bound substrate. ^Kamachi and Yoshizawa'^^.
72 Sason Shaik and Samuel P. De Visser

at least a good part of this entropic effect, due to of the corresponding HS and LS Por+Te"^OH/R*
restriction in the ^"^TS^ species, will not contribute
and PorFe^^OH/R* species. Indeed, gas phase cal-
to the free-energy barrier. Thus, the protein culations give as ground states either electromer,
machinery that utilizes mobile water molecules as for example found recently by Kamachi and
absorbs much of the entropic cost of establishing Yoshizawa^^ for camphor hydroxylation, where
a TS. Therefore, the gas phase quantities that are the LS electromer was of the Por+Te"^OH/R*
more informative of the situation in the protein are variety, while the HS electromer was of the
not AG^ but A(E+ZPE)^ and AH^. However, a PorFe^^OH/R* variety. All the five states can
thorough discussion of this feature is impossible in turn participate in rebound, and as was
at the time of the writing of this manuscript and repeatedly found^^' ^^^ that the HS intermediates
will have to await QM/MM calculations with real rebound with a significant barrier (2<AEjg|^<
sampling and thermodynamic integration. 6 kcalmol"^), while the LS intermediates rebound
without a barrier past the orientation phase
(Figure 2.17) that occurs by combined rotation of
5.4. The Rebound Process: the alkyl and OH groups.
More Features than The origin of the rebound barrier on the HS
Meet the Eye surface was analyzed^^' ^^2, i is ^j^^ shown to result
from the need to shift an electron from the alkyl
The iron-hydroxo intermediate that is formed radical to the heme and populate the high-lying
during the bond activation step exists in two a*2 orbital; this orbital is antibonding in the Fe-S
close-lying electromers^^^ which differ in the and Fe-O linkages (see Figure 2.14). In line
oxidation state of the metal and porphyrin ligand. with the population of the (T*2 orbital, both Fe-S
By reference to the orbital diagram in Figure 2.14, and Fe-0 bonds are seen from Figure 2.20
these electromers differ in the orbital occupancy to undergo lengthening in the '^TS^.^j^ species
of the d-block and porphyrin di^^ orbitals; the (relative to the values in the iron-hydroxo radical
Por+«Fe"^OH electromer has close lying singlet clusters, shown below the TSs). This bond length-
and triplet TT^^ ^* i ^^^^ configurations, while ening is the origin of the HS rebound barrier.
the PorFe^^OH state has a triplet TT^I TT*J 2i^^ By contrast, in the LS process, the electron,
configuration. These two electromeric situations shifted from the alkyl radical, fills only low-lying
are close in energy, and small changes such as orbitals, either the ir*^ orbital of iron or the
substituents on the porphyrin ring or replacement porphyrin 2i2n orbital depending on electromeric
of the axial ligand can reverse their ordering'^'. identity (Fe'" or Fe^^). The rate of the rebound
Coupling with the alkyl radical leads to five states process depends much on the electron donor

4TS,eb;R = C3H5
r p e o ^ 1-816 rFeO= 1.804
rpes = 2.480 rFeS = 2.356

Figure 2.20. Key geometric parameters for high-spin rebound transition states in methane hydroxylation^^ and
allylic hydroxylation^^^. The quantities below the structures correspond to the geometric parameters of the
corresponding iron-hydroxo/alkyl radical clusters ("^Cj in Figure 2.17).
Computational Approaches to Cytochrome P450 Function 73

capability of the radical and its C-0 bond strength more than QM or even QM/MM calculation can
(see above), but also on the acceptor properties of resolve at present. Here we have to look forward for
the iron-hydroxo species, its electromeric state, a combination of QM/MM with MD calculations.
and height of the (J*2 orbital; these properties
depend on the polarity and acidity of the protein 5.5. Alkene Epoxidation
pocket and its steric constraints on the Fe-S
bonding. The role of the a*2 orbital was recently The DFT (B3LYP/LACVP) computed mecha-
highlighted in the study of methane hydroxylation nistic scheme for ethene epoxidation by the
by the ruthenium analog of Cpd I (ref [67]), simplest model Cpd I species is summarized in
where a very high aji orbital led to a very high Figure 2.21^^' i^^, 116 jj^g fjj.g|^ g^^p involves bond
rebound barrier of c.11.9 kcalmol"^ An addi- activation and leads to the iron-alkoxy radical
tional factor revealed by the calculations'^^' ^^5,113 intermediate that appears in both Fe"^ and Fe^^
is that the rebound process may occur either by electromers of the HS and LS varieties. In a
OH rotation around the Fe-O bond, as in the case subsequent phase, these intermediates undergo
of the methane monooxygenase enzyme^ ^'*, or by ring-closure to form the epoxide complex. The
rotation of the alkyl group around the same bond, alternative synchronous concerted oxygen inser-
or still by some combination of the two modes. In tion was also tested^'' but ruled out as a viable
allylic hydroxylation, the prominent mode was mechanism. Precisely the same features were
found to be the rotation of the allyl group about obtained for propene epoxidation^^^, with the
the Fe-OH bond^^^, while in camphor hydroxy- exception that the bond activation barriers are
lation, both OH and alkyl rotations take part in cA kcalmol"^ lower than those, shown in
the rebound"*^. It is very clear that the topography Figure 2.21, for ethene epoxidation.
of the protein pocket and the mode of substrate The lowest energy TSs for bond activation
binding will play major roles in the rebound for ethene and propene activation are shown in
process, by selectively constraining/preferring Figure 2.22. Since the bond activation involves an
some of the rebound modes over others. The electron shift from the alkene to the heme (consult
results of Atkins and Sligar^^ that during camphor Figure 2.14), propene, which is a better electron
hydroxylation both exo and endo C-H bonds are donor than ethene, has lower barriers, 10.0 and
activated, but the only product is an exo-alcohol, 10.6 kcal mol~^ vis-a-vis 13.9 and 14.9 kcal mol~^
is indicative of the manifestations of such selec- (HS, LS). In accord, the TSs of propene are seen
tive constraints. All in all, the rebound process is to be earlier than those of ethene, with less C=C
more intricate than meets the eye, and certainly activation, etc. An interesting feature of the two

'^TS2-III
10.4

trs2-iv

ring closure C-O bond formation C-O bond formation ring closure

Figure 2.21. Two-state reactivity potential energy surface for ethene epoxidation^ ^^.
74 Sason Shaik and Samuel P. De Visser

TSs is their propensity for an upright orientation Past the bond activation phase, in Figure 2.21,
of the alkene moiety vis-a-vis the plane of the por- the radicals undergo ring closure. As in the
phyrin. This gas phase conformation avoids the hydroxylation, the LS radical complexes undergo
steric repulsion with the porphyrin. However, in ring-closure in a virtually barrierless fashion,
the protein pocket, the upright conformation may whereas on the HS surface, the radicals encounter
encounter repulsion from the side-chain amino significant barriers. These barriers for the HS Fe^^
acids, and a compromise may be achieved in the electromer are smaller than those for rebound in
parallel conformation, which is generated, in alkane hydroxylation. However, the ring-closure
Figure 2.22, from the gas phase TS by a simple barriers are large for the HS Fe^" electromers. This
rotation that achieves a dihedral FeOCC angle implies that the radical intermediate complexes,
of 90°. This structure is related to the one pro- and especially those for the Fe"^ electromer, will
posed by Groves et al}^^ to account for the have a significant lifetime only on the HS surface,
preferred reactivity of cis compared with trans where they may give rise to rearranged products or
isomers, due to enhanced steric repulsion of the lead to side reactions. For instance, rotations
substituents in the latter isomer with the por- around the C-C or the C - 0 bonds were found to
phyrin ring (see the corresponding distances in cost less than 1.5 kcal mol~'. Thus, C-C rotation
Figure 2.22). on the HS surface will result in the production

^ S (^TS) "hrs (^TS)

f 12.72

2.413(2.501)

AE^=: 13.9(14.9)
do
A E T = 10.0(10.6) parallel "TS"

Figure 2.22. Computed structures of the C-0 bond activation transition states, and barriers, for ethene^^^ and
propene epoxidation^^^. The putative "parallel" TS for ethene epoxidation is generated by rotating the computed one
around the 0 - C bond.

Relative
Energy 20
(kcal mol" )

1.5 1.6 1.7 1.8 1.9 2.0 2.1 2.2 2.3 2.4 2.5 2.6 2.7 2.8 2.9 3.0
r^.__^ (Angstrom)

Figure 2.23. Geometry scan for the formation of the suicidal complex by crossover from the high-spin Fe^"
intermediate ("^2-111') to the state generated by promoting an electron from the methylene radical group to the a*
orbital of iron (see Figure 2.3).
Computational Approaches to Cytochrome P450 Function 75

of both c/5-epoxide and trans-epoxidts from cis- formation of the aldehyde side product, which is
or tmns-alkQnQs, whereas the LS surface will formed in a related mechanism to the suicidal
essentially retain the original isomeric identity of complex^ ^^. The rebound barriers as well as those
the alkene. for the side product formation are subject to
Similarly, rotation around the C - 0 is facile polarity and NH — S hydrogen bonding effects ^^^.
and will bring the radical center to a position for
heme alkylation, where the alkylation barriers are 5.6. Hydroxylation of Arenes
small enough to compete with rebound. The DFT
calculations^ ^^ show that the HS radical complex One of the long-standing controversies con-
of the Fe^^^ electromer can cross over to a state that cerns the hydroxylation mechanism of arenes,
is initially higher lying, but which is adiabati- which apart from phenol can produce also ketone
cally connected with the suicidal complex in and arene oxide as side products. A universal
Figure 2.23. This crossing point is <10 kcal feature of arene-hydroxylation is the so-called
mol~^ above the Fe"^ radical intermediate, so that NIH-shift, which accounts for the fact that the
this process may be able to compete with the ring original hydrogen atom in the activated C-H bond
closure of the Fe"^ electromer to epoxide (with a is retained in the reaction products^' ^^. A recent
barrier of 7.2 kcal mol"^). The state, leading to the DFT investigation^ ^^ addressed the mechanism of
suicidal complex, is obtained by shifting an elec- benzene hydroxylation. A rebound mechanism,
tronfi-omthe CH2 moiety of the radical complex, analogous to alkane hydroxylation, and an elec-
which thereby becomes a carbocationic center, to tron transfer mechanism were ruled out due to
the a* orbital of the heme, which thereby their high-energy costs, while the lowest energy
becomes anionic with loose Fe-N bonds. As such, mechanisms were found to involve ir-attack as
the state of the suicidal complex is an internal summarized in Figure 2.24. In accord with deduc-
ion pair that undergoes cation (C+) anion (N~) tions from experimental data^^^' ^^^ the computa-
combination, which is likely to be facilitated tions show that the bond activation proceeds via
by the polar environment of the protein pocket. two mechanisms; one leads to a radical a-complex
We have preliminary results for the mechanism of C(J-C') and the other to a cationic a-complex

^ ^ B O
Figure 2.24. Potential energy surface for the competing oxidation mechanisms of benzene by Cpd I (ref [119]).
76 Sason Shaik and Samuel P. De Visser

(^a-C+). Both the radical and electrophihc mech- 5.7. Sulfoxidation of Alkyl
anisms involve the LS states, while the HS states Sulfides
give rise to TSs that are too high to compete with
the LS states. Polarity and NH—S hydrogen Another common reaction of P450 is the sul-
bonding prefer the electrophihc pathway com- foxidation of sulfides^ Figure 2.25 summarizes
pared with the radical one. Thus, the calculations the results of a recent DFT study of the sulfoxida-
predict that the major pathway for arene hydroxy- tion reaction of dimethyl sulfide with a Cpd I
lation will be the electrophihc pathway. model ^^^. The reaction is seen to proceed in a
The so-formed intermediates, in Figure 2.24, concerted manner via LS and HS pathways.
subsequently bifurcate either to the ferric benzene However, in contrast to the hydroxylation of
oxide (^BO) complex by ring closure, or to the benzene^ ^^, which occurs by a dominant LS
N-protonated porphyrin intermediate (^PP) by potential energy surface, sulfoxidation exhibits a
proton transfer from the ipso carbon to one of dominant HS pathway, which becomes even
the nitrogens of the porphyrin. In turn, the latter more so by inclusion of the effect of medium
intermediate reshuttles the proton either to the oxo polarity (using a dielectric constant, 8 = 5.7).
group to give phenol (^P) or to the ortho carbon to Unlike alkane hydroxylation and alkene epoxida-
give the ketone (^K). Since both TS^-shuttle tion where the TSs are related in their electronic
species lie well below TS^^^^^, the excess energy structures and differ in having ferro-, namely anti-
will give rise to both phenol and ketone. This ferromagnetic coupling of the three unpaired elec-
proton-shuttle mechanism accounts for the NIH- trons, in sulfoxidation, the LS and HS TSs are
shift, since the original hydrogen in the activated very different in their geometries and electronic
C-H bond ends up in the products. It follows structures because the two oxidation equivalents
therefore that, in addition to phenol production by (Figure 2.14) must be condensed into a single
nonenzymatic protonation of benzene oxide under step. Thus, sulfoxidation behaves as HS and LS
physiological condition, there should exist an displacement reactions where the sulfur attacks
enzymatic pathway that leads directly to phenol the 0X0 moiety and displaces the heme, which in
and ketone production without the intermediacy turn rebinds to the sulfoxide via a long Fe-0
of benzene oxide. bond. Different heme-oxo orbitals figure in the

/2.296
19.2(19.3) 'Ts o^am^i^^^m
12.282
16.8(16.0)/
^TS

0.0(-0.1)--J
0.0 (0.0) —

-24.7 (-27.4)
,.''4.164(4.160) 1.525(1.544)^ -32.0 (-33.1)
1.629(1.628) 2.508(2.1

2.559 (2.572) 2.458 (2.255)


43(^3)

Figure 2.25. Potential energy surface for the sulfoxidation of dimethyl sulfide by Cpd I (ref [122]). Barriers in
parentheses incorporate the effect of a dielectric constant, e == 5.7.
Computational Approaches to Cytochrome P450 Function 77

interaction with the lone pair of the attacking sul- simultaneous protonation (using a cluster of H30^
fur; in the LS process, these are the TT*(FeO) and and H2O) and oxygen insertion processes were
^2^ orbitals that get filled up during the displace- attempted too, and led to barriers which are at
ment, whereas in the HS process, the a*2 and least 10 kcal mol~^ higher than those by Cpd I^^^.
a2y orbitals accept each one electron through the These results clearly show that by itself, Cpd 0
interaction with the sulfur. It seems reasonable to cannot possibly compete with Cpd I because the
expect that the sulfoxidation results apply to other negative charge makes it a good base and a good
heteroatom oxidation processes. Our calculations, nucleophile, but not an electrophile^^. Its activa-
done for a single substrate, do not rule out an elec- tion by a proton source improves the situation*^^,
tron transfer mechanism for substrates that are but even then it appears that the Cpd 0 is a much
more powerful donors. However, as a rule, the cys- poorer oxidant than Cpd I.
teine ligand makes Cpd I of P450 a relatively poor
electron acceptor to participate in pure electron
transfer processes with diffusive products'*^. 5.9. Competitive Hydroxylation
and Epoxidation in Propene
5.8. Can Ferric Peroxide (6) be a The competition between C-H hydroxylation
Second Oxidant? and C=C epoxidation for a given substrate was
addressed^^'*' ^^^ using propene as a model. The
This question was addressed by the Jerusalem reaction profiles for in-vacuum conditions are
group using the marker reaction of Cpd 0: alkene shown in Figure 2.27, which exhibit the already
epoxidation^^; ethene served as the alkene. Four known features of TSR with effectively concerted
different mechanisms were computed, correspon- LS pathways and stepwise HS mechanisms. First,
ding to concerted and stepwise epoxidations by
the barriers are seen to be extremely small and
both distal and proximal oxygen groups of Cpd 0.
The barriers for the four mechanisms relative to they get even smaller when NH—S hydrogen
those obtained by Cpd I that are displayed in bonding is included (e.g., the barrier for the LS
Figure 2.26, show that Cpd 0 is by far inferior hydroxylation becomes c.8.96 kcal mol"^ only).
to Cpd I. With these small barriers, we may anticipate that
Similar results were obtained for sulfoxida- the substrate binding will control the regioselec-
tion^^^, where Cpd 0 led to barriers in excess of tivity, so that whichever moiety is bound to Cpd I
40 kcal mol~^ more than 20 kcal mol~^ higher will be the one to react, and will do so too fast to
than the barriers calculated for sulfoxidation by allow the observation of the competing reaction.
Cpd I. Hydrogen bonding (to an H2O molecule) or Indeed, propene undergoes exclusive epoxidation

AE'^ = J3.9 (HS);


14.9 (LS) kcal mol^ 0
TSR, nonsynchronous CH2—CH2

.Fe^; + CH2=CH2

AE^ = 21.1 kcal mol^ 0


TSR, synchronous / \
CH2—CH2
\E^ = 37-44kcalmol-^ 0
I
/ \
OH SSR, nonsynchronous CH2—CH2

, FU + CH2=CH2
I -OH*
SH AE^ = 45-53kcalmol-^ 0
SSR, synchronous CH2—CH2

Figure 2.26. Epoxidation barriers of ethene by Cpd I and Cpd 0 (refs [53,115]).
78 Sason Shaik and Samuel P. De Visser

E + ZPE

TSl
/4.
103
TSl'i
f
,^TS4 1+C3H6\ / \,
1.0 ^^ W.O
^1+C3H6 - 2 ^ ^ •
-2.6^-

I /'Hn
C3H6 ^ „ . "'-^^i?'^
-45.5- .C3H5
HO
siCH3
C3H5 Fe SH
HO
SH
-Fe- -Fe—
j„ - Hydroxy!ation - • Epoxidation -
L
Figure 2.27. Potential energy landscape for the hydroxylation and epoxidation pathways of propene in the gas
phase •°^. Relative energies include zero point energies (ZPE).

tlL^Sl
+i.2

I Epoxidation
I Hydroxylation! + /.0
i r s 3 +0.2.
trs3 o.oi go 0.0 2.TSl
^TSl -OJ- -0.6
-7.4 -7.4
I Epoxidation |
I Hydroxylation]

-3.0 2.TS3

,R
Q" O'

-L
SH
2.660 A \-HNH2
r„ ^•-*t:„?
HNH2
£=1.0,E-hZPE £=1.0, E + ZPE £ = 5.7, E -h ZPE £ = 5.7, E + ZPE

Figure 2.28. Relative energy of the transition states (TSs) for hydroxylation and epoxidation in the gas phase and
under the influence of NH — S hydrogen bonding and polarity; the latter is mimicked by a dielectric constant
e = 5.7 (adapted from ref. [104, 105] with permission).
Computational Approaches to Cytochrome P450 Function 79

with P450L^2 fr^f- [12^])' ^"t this is not the case polarity of the pocket and its hydrogen-bonding
for other simple alkenes, for example, cyclohex- machinery. The gas phase (in vacuum) calcula-
ene^^"^. As such, our results with propene should tions show that, while alkane hydroxylation and
be regarded as a model study of factors affecting alkene epoxidation feature TSR, by contrast,
regioselectivity and stereoselectivity rather than benzene hydroxylation exhibits a dominant LS
a specific study of a given substrate. In this reactivity and in sulfoxidation it is the HS state
respect, the calculations show that the bond acti- that dominates reactivity. As a rule of thumb, we
vation phase is the rate-limiting step for both may say that whenever the substrate deformation
processes. is small or identical for the two states, they
Figure 2.28 displays the four-bond activation will have similar electronic structures and be
TSs under different conditions. In the gas phase, energetically close, as for the reactant species
the four species are condensed within 0.6 kcal Cpd I. This appears to be the case in epoxidation
mol~ ^ with a slight preference for the epoxidation and hydroxylation, which exhibit clear TSR. The
species. This in turn means that the gas phase dominance of the LS state in benzene hydroxyla-
reaction will exhibit (a) a low regioselectivity of tion was shown^^^ to originate in the large defor-
C=C over C-H and (b) low stereospecificity due mation energy of benzene, due to the partial loss
to HS/LS scrambling. The addition of just two of the resonance energy, which is c.lO kcal mol~^
NH — S hydrogen bonds is sufficient to render the more severe for the HS TSs. In heteroatom oxida-
LS hydroxylation TS the lowest one by a wide tion, however, the electronic structures during the
margin. Adding the effect of a polar environment HS and LS processes are not the same since
(mimicked by a dielectric constant of e = 5.7) the two oxidation equivalents must be condensed
creates a clear preference for hydroxylation over into a single step, and for sulfoxidation, the HS
epoxidation, by 3.0 kcal mol~^ In addition, for state appears to be the lower one of the two.
each process, now the LS pathway has a lower We may therefore anticipate that arene hydro-
barrier than the HS pathway. These effects corre- xylation and heteroatom oxidation will feature
spond to a regioselectivity reversal by almost three a SSR with a substrate-dependent spin-state
orders of magnitude in favor of hydroxylation. In selection.
addition, since these effects favor the LS path- A feature encountered during benzene hydrox-
ways, they also induce improvement in the stere- ylation is the appearance of both radical and
oselectivity of both hydroxylation and epoxidation cationic mechanisms. This feature is associated
by almost three orders of magnitude. It follows with the stability of the cationic species and will
therefore that the factors that mimic the polarity of appear in alkane hydroxylation and alkene epoxi-
the protein pocket and its hydrogen-bonding dation whenever the corresponding radical center
machinery have a major impact on the selectivity has a sufficiently low ionization energy to transfer
patterns of Cpd I. In fact, since Cpd I is a an electron to the heme. In such an event, the reac-
chameleon species, it also acts as a chameleon tivity will be dominated by the LS state. The result
oxidant that tunes its reactivity patterns in of Newcomb and Toy^^ which indicates the
response to the polarity and hydrogen-bonding presence of carbocations may well belong to this
machinery of the protein pocket. Thus, along with category. Such results are in progress.
substrate binding, the hydrogen-bonding machin- The polarity of the pocket and hydrogen-
ery and polarity of the protein serve as means by bonding machinery were found to increase the
which the enzyme tunes its selectivity. One won- dominance of the LS reactivity in arene hydroxy-
ders whether this factor would not play a role in lation and the HS reactivity in sulfoxidation.
the great versatility of the superfamily of P450 Similarly, these factors will have a strong impact
enzymes. on the appearance of cationic intermediates dur-
ing hydroxylation and epoxidation. More intrigu-
ing is the result that these properties of the protein
5.10. An Overview of Reactivity pocket have a major impact on the regioselectivity
of C-H hydroxylation versus C=C epoxidation as
Features of Cpd I
well as on the stereospecificity of both processes.
cpd I is a chameleon and two-state oxidant Time will show whether these are generalities or
and as such exhibits TSR that can be tuned by the isolated findings.
80 Sason Shaik and Samuel P. De Visser

Theory also shows that the porphyrin ring fault of the theoretical calculations, this is likely to
is not a spectator hgand but plays quite a few be discovered as soon as calculations become
roles during the enzymatic reaction. It acts as an faster and allow the use of more sophisticated
electron sink by accepting the excess electrons methods (e.g., CCSD(T), CASPT2). If however,
generated during the oxidation, and as a proton these are reasonable estimates of the barrier, we
sponge by reshuttling protons needed for substrate will be facing a conceptual dilemma to explain the
rearrangement. These are in addition to its adverse potency of the enzyme despite the large barriers.
role in enabling heme alkylation. In our calcula- While a few such possible scenarios come to mind
tions, we find that heme alkylation is more facile immediately, we prefer to leave these as open
with ligands that are not good electron donors as questions that advanced theory will have to deal
the thiolate is. Thus, the thiolate ligand protects with in the future.
the porphyrin against heme alkylation, and at the
same time, renders its nitrogen more basic and,
hence, improves its catalytic performance as a Acknowledgment
proton shuttle.
Sason Shaik's research was supported by
ISF and GIF grants.
6. Prospective

As shown in this chapter, theoretical treatments References


of P450 problems have come of age, and they
enable the study of many features related to the 1. Ortiz de Montellano, P.R. (ed.), (1995). Cytochrome
three-dimensional structures, the electronic struc- P-450: Structures, Mechanism and Biochemistry,
tures, substrate binding, reactivity, and dynamics 2nd edn. Plenum Press, New York.
of the various physical processes in the cycle. This 2. Loew, G.H., C.J. Kert, L.M. Hjelmeland, and
ability will only increase with time, as the various R.F. Kirchner (1977). Active site models of horse-
techniques will be brought to bear simultaneously radish peroxidase compound 1 and a cytochrome
on a given problem. Techniques similar to combi- P450 analogue: Electronic structure and electric
natorial synthesis will have to be adopted in order field gradients. J. Am. Chem. Soc. 99, 3534-3536.
to screen fast many alternative possibilities. 3. Cramer, C.J. (2002). Essentials of Computa-
tional Chemistry: Theories and Models. Wiley,
At present, while calculations have elucidated Chichester.
some important features of the cycle, still others 4. Ghosh, A. and PR. Taylor (2003). High-level ab
remain obscure; especially features which con- initio calculations on the energies of low-lying spin
cern the protein machinery and the dynamics states of biologically relevant transition metal com-
of the various reactions, the role of the water plexes: A first progress report. Curr Opin. Chem.
molecules that are present in the pocket and Biol. 7, 113-124.
leave it upon entrance of the substrate, and the dia- 5. Ldemann, S.K., V. Lounnas, and R.C. Wade (2000).
logue between the oxygenase and the reductase How do substrates enter and products exit the
domains, as well as the thermodynamic aspects of buried active site of cytochrome P450cam? 1.
Random expulsion molecular dynamics investiga-
the entire cycle which must be determined by
tion of ligand access channels and mechanisms.
accounting for the changes in the reductase.
JM0/.5/0/. 303, 797-811.
Other outstanding issues that remain unat- 6. Ldemann, S.K., V Lounnas, and R.C. Wade (2000).
tended at present are the following: How do substrates enter and products exit the buried
active site of cytochrome P450cam? 2. Steered
(1) The role of spin crossover in the
molecular dynamics and adiabatic mapping of
various events in the cycle, including the TSR dur-
substrate pathways. J. Mol. Biol. 303, 813-830.
ing oxygenation, which will have to be elucidated. 7. Hermans, J. and L. Wang (1997). Inclusion of loss of
(2) All calculations of C - H hydroxyla- translational and rotational freedom in theoretical
tion, with the exception of allylic hydroxylation, estimates of free energies of binding. Application to
show a significant barrier of 18-24 kcalmol"^. a complex of benzene and mutant T4 lysozyme.
Such barriers seem to be overestimated. If this is a J.Am. Chem. Soc. 119, 2707-2714.
Computational Approaches to Cytochrome P450 Function 81

8. Kairys, V and M.K. Gilson (2002). Enhanced in metalloporphyrin-nitrosyl complexes. J. Am.


docking with the mining minima optimizer: Chem. Soc. 122, 8101-8102.
Acceleration and side-chain flexibihty. J. Comput. 22. Wondimagegn, T. and A. Ghosh (2001). A quantum
Chem. 23, 1656-1670. chemical survey of metalloporphyrin-nitrosyl
9. Davydov, R., T.M. Makris, V. Kofman, D.E. Werst, linkage isomers: Insights into the observation of
S.G. Sligar, and B.M. Hoffman (2001). Hydroxy- multiple FeNO conformations in a recent crystallo-
lation of camphor by reduced oxy-cytochrome graphic determination of nitrophorin 4. J. Am.
P450cam: Mechanistic implications of EPR and Chem. Soc. 123, 5680-5683.
ENDOR studies of catalytic intermediates in 23. Ghosh, A. (1998). First-principles quantum chemical
native and mutant enzymes. J. Am. Chem. Soc. 123, studies of porphyrins, ^cc. Chem. Res. 31, 189-198.
1403-1415. 24. Ghosh, A., J. Almlfi and L. Que Jr. (1994).
10. Hata, M., T. Hoshino, and M. Tsuda (2000). Density functional theoretical study of oxo(por-
An ultimate species in the substrate oxidation phyrinato) iron(IV) complexes, models of peroxi-
process by cytochrome P-450. Chem. Comm. dase Compounds I and II. J. Phys. Chem. 98,
2037-3038. 5576-5579.
11. Du, R, F.U. Axe, G.H. Loew, S. Canuto, and M.C. 25. Ghosh, A. and E. Gonzalez (2000). Theoretical
Zerner (1991). Theoretical study on the electronic studies on high-valent manganese porphyrins:
spectra of model Compound II complexes of Toward a deeper understanding of the energetics,
peroxidases. J^ ^m. Chem. Soc. 113, 8614-8621. electron distributions, and structural features of the
12. Kuramochi, H., L. Noodleman, and D.A. Case reactive intermediates of enzymatic and synthetic
(1997). Density functional study on the electronic manganese-catalyzed oxidative processes. Isr. J.
structures of model peroxidase compounds I and II. Chem. 40, 1-8.
J.Am. Chem. Soc. 119, 11442-11451. 26. Harris, D.L. and G.H. Loew (1998). Theoretical
13. Dawson, J.H., R.H. Holm, J.R. Trudell, G. Barth, investigation of the proton assisted pathway to for-
R.E. Linder, E. Bunnenberg et al. (1976). Oxidized mation of cytochrome P450 compound I. J. Am.
cytochrome P-450. Magnetic circular dichroism Chem. Soc. 120, 8941-8948.
evidence for thiolate ligation in the substrate-bound 27. Filizola, M. and G.H. Loew (2000). Role of protein
form. Implications for the catalytic mechanism. environment in horseradish peroxidase compound I
J. Am. Chem. Soc. 98, 3707-3709. formation: Molecular dynamics simulations of
14. Poulos, T.L. (1996). The role of the proximal ligand horseradish peroxidase-HOOH complex. J. Am.
in heme enzymes. J. Biol. Inorg. Chem. 1, 356-359. Chem. Soc. Ill, 18-25.
15. SchHchting, L, J. Berendzen, K. Chu, A.M. Stock, 28. Loew, G.H. and M. Dupuis (1996). Structure of a
S.A. Maves, D.E. Benson et al. (2000). The model transient peroxide intermediate of peroxi-
catalytic pathway of cytochrome P450cam at dases by ab initio methods. J. Am. Chem. Soc. 118,
atomic resolution. Science 287, 1615-1622. 10584-10587.
16. Loew, G.H. and D.L. Harris (2000). Role of the 29. Thomann, H., M. Bernardo, D. Goldfarb, RM.H.
heme active site and protein environment in struc- Kroneck, and V Ullrich (1995). Evidence for water
ture, spectra, and function of the cytochrome binding to the Fe-center in cytochrome P450cam
P450s. Chem. Rev. 100, 407-419. obtained by ^^O electron spin echo envelope
17. Loew, G.H. (2000). Structure, spectra, and function modulation spectroscopy. J. Am. Chem. Soc. Ill,
of heme sites. Int. J. Quantum Chem. 11, 54-70. 8243-8251.
18. Gfler, A.H. and T Clark (2001). SAMl semiem- 30. Collins, J.R., P Du, and G.H. Loew (1992).
pirical calculations on the mechanism of cyto- Molecular dynamics simulations of the resting and
chrome P450 metabolism. Theochem. 541,263-281. hydrogen peroxide-bound states of cytochrome c
19. Schiieboom, J.C, H. Lin, N. Reuter, W. Thiel, S. peroxidase. Biochemistry 31, 11166-11174.
Cohen, F. Ogliaro et al. (2002). The elusive oxidant 31. Harris, D.L. and G.H. Loew (1993). Mechanistic
species of cytochrome P450 enzymes: Char- origin of the correlation between spin state and
acterization by combined quantum mechanical/ spectra of model cytochrome P450 ferric heme pro-
molecular mechanical (QM/MM) calculations. teins. J:^m. Chem. Soc. 115, 5799-5802.
J.Am. Chem. Soc. 124, 8142-8151. 32. Harris, D.L. and G.H. Loew (1993). Determinants
20. Miyahara, T, Y. Tokita, and H. Nakatsuji (2001). of the spin state of the resting state of cytochrome
SAC/SAC-CI study of the ground, excited, and P450cam. J ^ m . Chem. Soc. 115, 8775-8779.
ionized states of cytochromes P450CO. J. Phys. 33. Aissaoui, H., R. Bachmann, A. Schweiger, and
Chem. B 105, 7341-7352. W.-D. Woggon (1998). On the origin of the
21. Ghosh, A. and T Wondimagegn (2000). A theoreti- low-spin character of cytochrome P450cam in the
cal study of axial tilting and equatorial asymmetry resting state-investigations of enzyme models with
82 Sason Shaik and Samuel P. De Visser

pulse EPR and ENDOR spectroscopy. Angew. catalytically active, ferryl species of P450s by
Chem. Int. Ed. 37, 2998-3002. molecular dynamics studies of P450eryF. J. Am.
34. Green, M.T. (1998). Role of the axial ligand in Chem. Soc. 118, 6377-6387.
determining the spin state of resting cytochrome 48. Guallar, V, D.L. Harris, VS. Batista, and
P450. J. Am. Chem. Soc. 120, 10772-10773. W.H. Miller (2002). Proton-transfer dynamics in
35. Filatov, M., N. Harris, and S. Shaik (1999). A the activation of cytochrome P450eryF. J. Am.
theoretical study of electronic factors affecting Chem. Soc. 124, 1430-1437.
hydroxylation by model ferryl complexes of 49. Kamachi, T. and K. Yoshizawa (2003). A theoretical
cytochrome P-450 and horseradish peroxidase. study on the mechanism of camphor hydroxylation
J. Chem. Soc. Perkin Trans. 2, 399^10. by compound I of cytochrome P450. J. Am. Chem.
36. De Visser, S.R, D. Kumar, S. Cohen, P.K. Sharma, Soc. 125,4652^661.
and S. Shaik (2004, in preparation). A theoretical 50. Aikens, J. and S.G. Sligar (1994). Kinetic solvent
investigation of the proton-relay mechanisms for isotope effects during oxygen activation by
the formation of Cpd 0 and Cpd I in the catalytic cytochrome P-450cam. J. Am. Chem. Soc. 116,
cycle of cytochrome P450. 1143-1144.
37. Scherlis, D.A., C.B. Cymeryng, and D.A. Estrin 51. Harris, D.L. (2002). Oxidation and electronic state
(2000). Nitric oxide binding to ferric cytochrome dependence of proton transfer in the enzymatic
P450: A computational study. Inorg. Chem. 39, cycle of cytochrome P450eryF. J. Inorg. Biochem.
2352-2359. 91, 568-585.
38. ScherUs, D.A., M.A. Mart; P OrdeJD, and D.A. 52. Vidakovic, M., S.G. Sligar, H. Li, and T.L. Poulos
Estrin (2002). Environment effects on chemical (1998). Understanding the role of the essential
reactivity of heme proteins. Int. J. Quantum Chem. Asp251 in cytochrome P450cam using site-directed
90, 1505-1514. mutagenesis, crystallography, and kinetic solvent
39. Loew, G.H. and M. Dupuis (1997). Characterization isotope effect. Biochemistry 37, 9211-9219.
of a resting state model of peroxidases by ab initio 53. Ogliaro, R, S.P De Visser, S. Cohen, PK Sharma,
methods: Optimized geometries, electronic struc- and S. Shaik (2002). Searching for the second
tures, and relative energies of the sextet, quartet, oxidant in the catalytic cycle of cytochrome P450:
and doublet spin states. J. Am. Chem. Soc. 119, A theoretical investigation of the iron(lII)-hydro-
9848-9851. peroxo species and its epoxidation pathways. J. Am.
40. Ogliaro, F, S.P De Visser, and S. Shaik (2002). The Chem. Soc. 124,2806-2817.
"push" effect of the thiolate ligand in cytochrome 54. Sono, M., M.P Roach, E.D. Coulter, and
P450: A theoretical gauging. J. Inorg. Biochem. 91, J.H. Dawson (1996). Heme-containing oxygenases.
554-567. Chem. Rev. 96,2841-2887.
41. Shgar, S.G. (1976). Coupling of spin, substrate, and 55. Yamamoto, S., J. Teraoka, and H. Kashiwagi
redox equilibria in cytochrome P450. Biochemistry (1988). Ab initio RHF and CASSCF studies on
15,5399-5406. Fe-O bond in high-valent iron-oxo porphyrins.
42. Auclair, K., P. MoQne-Loccoz, and PR. Ortiz de J. Chem. Phys. 88,303-312.
Montellano (2001). Role of the proximal heme thi- 56. Harris, D.L., G.H. Loew, and L. Waskell (2001).
olate ligand in cytochrome P450cam. J. Am. Chem. Calculation of the electronic structure and spectra
Soc. 123, 4877-4885. of model cytochrome P450 compound I. J. Inorg.
43. Yamamoto, S. and H. Kashiwagi (1989). CASSCF Biochem. 83,309-318.
study on the Fe-02 bond in a dioxygen heme com- 57. Ogliaro, F, S. Cohen, M. Filatov, N. Harris, and
plex. Chem. Phys. Lett. 161, 85-89. S. Shaik (2000). The high-valent compound of
44. Harris, D.L., G.H. Loew, and L. Waskell (1998). cytochrome P450: The nature of the Fe-S bond and
Structure and spectra of ferrous dioxygen and the role of the thiolate ligand as an internal electron
reduced ferrous dioxygen model cytochrome P450. donor. Angew. Chem. Int. Ed. 39, 3851-3855.
J. Am. Chem. Soc. 120, 4308^318. 58. Ogliaro, F, S.P De Visser, S. Cohen, J. Kaneti, and
45. Chottard, G., M. Schappacher, L. Ricard, and R. S. Shaik (2001). The experimentally elusive
Weiss (1984). Resonance Raman spectra of iron(Il) oxidant of cytochrome P450: A theoretical "trap-
cytochrome P450 model complexes: Influence of ping" defining more closely the "real" species.
the thiolate ligand. Inorg. Chem. 23, 4557^561. Chembiochem. 2, 848-851.
46. Harris, D.L. and G.H. Loew (1994). A role for 59. Antony, J., M. Grodzicki, and A.X. Trautwein
Thr252 in cytochrome P450cam oxygen activation. (1997). Local density functional study of oxo-
J. Am. Chem. Soc. 116, 11671-11674. iron(IV) porphyrin complexes and their one-
47. Harris, D.L. and G.H. Loew (1996). Investigation electron oxidized derivatives. Axial ligand effects.
of the proton-assisted pathway to formation of the J. Phys. Chem. A 101, 2692-2701.
Computational Approaches to Cytochrome P450 Function 83

60. Green, M.T. (1999). Evidence for sulfur-based 73. Lewis, D.FV (2001). Guide to Cytochromes P450.
radicals in thiolate compound I intermediates. Taylor and Francis, New York.
J.Am. Chem. Soc. 121, 7939-7940. 74. Poulos, TL., B.C. Finzel, and A.J. Howard (1986).
61. Ohta, T., K. Matsuura, K. Yoshizawa, and I. Crystal structure of substrate-free Pseudomonas
Morishima (2000). The electronic and vibrational putida Cytochrome P450. Biochemistry 25,
structures of iron-oxo porphyrin with a methoxide 5314-5322.
or cysteinate axial ligand. J. Inorg. Biochem. 82, 75. Mueller, E.J., RJ. Loida, and S.G. Sligar (1995).
141-152. Twenty-five years of P450cam research. In PR.
62. Ogliaro, R, N. Harris, S. Cohen, M. Filatov, S.R De Ortiz de Montellano (ed.), Cytochrome P-450:
Visser, and S. Shaik (2000). A model "rebound" Structures, Mechanism and Biochemistry, 2nd edn.
mechanism of hydroxylation by cytochrome P450: pp. 83-124. Plenum Press, New York.
Stepwise and effectively concerted pathways, and 76. Schoeboom, J.C, S. Cohen, H. Lin, S. Shaik, and
their reactivity patterns. J. Am. Chem. Soc. 122, W Thiel (2004). Quantum mechanical/molecular
8977-8989. mechanical investigation of the mechanism of C-H
63. Ogliaro, E, S. Cohen, S.P. De Visser, and S. Shaik hydroxylation of camphor by cytochrome P450^gj^:
(2000). Medium polarization and hydrogen bond- Theory supports a two-state rebound mechanism.
ing effects on compound I of cytochrome P450: J Am. Chem. Soc. 126, 4017-4034.
What kind of a radical is it really? J. Am. Chem. 77. KeseriiG.M., I. Kolossv^, and B. Bertfe (1997).
Soc. 122, 12892-12893. Cytochrome P-450 catalyzed insecticide metabo-
64. Rutter, R., L.R Hager, H. Dhonau, M. Hendrich, lism. Prediction of regio- and stereoselectivity in
M. Valentine, and P. Debrunner (1984). Chloro- the primer metabolism of carbofuran: A theoretical
peroxidase compound I: Electron paramagnetic study, ^ ^ m . Chem. Soc. 119, 5126-5131.
resonance and Mssbauer studies. Biochemistry 23, 78. KeserjiG.M., I. Kolossvfy, and I. Szfeely (1999).
6809-6816. Inhibitors of cj^ochrome P450 catalyzed insecticide
65. De Visser, S.P, F. Ogliaro, Z. Gross, and S. Shaik metabolism: A rational approach. Int. J. Quantum
(2001). What is the difference between the man- Chem. 73, 123-135.
ganese porphyrin and corrole analogues of 79. Cavalli, A. and M. Recanatini (2002). Looking for
cytochrome P450's compound I? Chem. Eur. J. selectivity among cytochrome P450s inhibitors.
7, 4954^960. J Med Chem. 45,251-254.
66. Ogliaro, K, S.R De Visser, J.T. Groves, and S. Shaik 80. Lee, H., PR. Ortiz de Montellano, and
(2001). Chameleon states: High-valent metal-oxo A.E. McDermott (1999). Deuterium magic angle
species of cytochrome P450 and its ruthenium spinning studies of substrates bound to cytochrome
analogue. Angew. Chem. Int. Ed. 40, 2874-2878. F450. Biochemistry 3S, 10808-10813.
67. Sharma, RK., S.R De Visser, F. Ogliaro, and S. 81. De Voss, J.J., O. Sibbesen, Z. Zhang, and PR. Ortiz
Shaik (2003). Is the ruthenium analog of compound de Montellano (1997). Substrate docking algo-
I of cytochrome P450 an efficient oxidant? A theo- rithms and prediction of the substrate specificity of
retical investigation of the methane hydroxylation cytochrome P450cam and its L244A mutant. J. Am.
reaction. J ^m. Chem. Soc. 125, 2291-2300. Chem. Soc. 119, 5489-5498.
68. Green, M.T. (2000). Imidazole-ligated compound I 82. Atkins, WM. and S.G. SHgar (1987). MetaboHc
intermediates: The effects of hydrogen bonding. switching in cytochrome P450cam: Deuterium
J.Am. Chem. Soc. Ill, 9495-9499. isotope effects on regiospecificity and the
69. Green, M.T. (2001). The structure and spin cou- monooxygenase/oxidase ratio. J. Am. Chem. Soc.
pling of catalase compound I: A study of noncova- 109, 3754-3760.
lent effects. J Am. Chem. Soc. 123, 9218-9219. 83. Audergon, C , K.R. Iyer, J.R Jones, J.F, DarByshire,
70. Deeth, R.J. (1999). Saddle distortions of ferryl- and WE Trager (1999). Experimental and theoreti-
porphyrin models for peroxidase compound I: cal study of the effect of active-site constrained
A density functional study. J.Am. Chem. Soc. Ill, substrate motion on the magnitude of the observed
6074-6075. intramolecular isotope effect for the P450 101 cat-
71. Ogliaro, F. and S. Shaik (2003). Substituent effects alyzed benzylic hydroxylation of isomeric xylenes
on structure and properties of compound I species. and 4,4'-dimethylbiphenyl. J. Am. Chem. Soc. Ill,
Unpublished results. 4\-41.
72. Wirstam, M., M.R.A. Blomberg, and RE.M. 84. Helms, V and R.C. Wade (1998). Hydration energy
Siegbahn (1999). Reaction mechanism of com- landscape of the active site cavity in cytochrome
pound I formation in heme peroxidases: A density P450cam. Proteins 32, 381-396.
functional theory study. J. Am. Chem. Soc. Ill, 85. Winn, P.J., S.K. Ldemann, R. Gauges, V Lounnas,
10178-10185. and R.C. Wade (2002). Comparison of the
84 Sason Shaik and Samuel P. De Visser

dynamics of substrate access channels in three cytochrome P450 does not proceed in a concerted
cytochrome P450s reveals different opening mecha- synchronous manner. Chem. Comm. 2322-2323.
nisms and a novel functional role for a buried argi- 98. Groves, J.T and G.A. McClusky (1976). Aliphatic
nine. Proc. Natl Acad. Sci. USA. 99, 5361-5366. hydroxylation via oxygen rebound. Oxygen trans-
86. Das, B., V Helms, V Lounnas, and R.C. Wade fer catalyzed by iron. J. Am. Chem. Soc. 98,
(2000). Multicopy molecular dynamics simulations 859-861.
suggest how to reconcile crystallographic and prod- 99. Ortiz de Montellano, PR. and R.A. Stearns (1987).
uct formation data for camphor enantiomers bound Timing of the radical recombination step in
to cytochrome P-450cam. J. Inorg. Biochem. 81, cytochrome P450 catalysis with ring-strained
121-131. probes. J : ^ W . Chem. Soc. 109, 3415-3420.
87. Fruetel, J.A., J.R. Collins, D.L. Camper, G.H. 100. Newcomb, M., R. Shen, S.-Y. Choi, PH. Toy, PF
Loew, and P.R. Ortiz de Montellano (1992). HoUenberg, A.D.N. Vaz et al. (2000). Cytochrome
Calculated and experimental absolute stereochem- P450-catalyzed hydroxylation of mechanistic
istry of the styrene and p-methylstyrene epoxides probes that distinguish between radicals and
formed by cytochrome P450cam. J. Am. Chem. Soc. cations. Evidence for cationic but not for radical
114, 6987-6993. intermediates. J. Am. Chem. Soc. 122, 2677-2686.
88. Harris, D.L. and G.H. Loew (1995). Prediction of 101. Filatov, M., N. Harris, and S. Shaik (1999). On the
regiospecific hydroxylation of camphor analogs "rebound" mechanism of alkane hydroxylation by
by cytochrome P450cam. J. Am. Chem. Soc. 117, cytochrome P450: Electronic structure of the inter-
2738-2746. mediate and the electron transfer character in the
89. Park, J.-Y. and D.L. Harris (2003). Construction rebound step. Angew. Chem. Int. Ed. 38, 3510-3512.
and assessment of models of CYP2E1: Predictions 102. Harris, N., S. Cohen, M. Filatov, F Ogliaro, and S.
of metabolism from docking, molecular dynamics Shaik (2000). Two-state reactivity in the rebound
and density functional theoretical calculations. step of alkane hydroxylation by cytochrome P-450:
J. Med. Chem. 46, 1645-1660. Origins of free radicals with finite lifetimes.
90. Vaz, A.D.N., D.F. McGinnity, and M.J. Coon Angew. Chem. Int. Ed. 39, 2003-2007.
(1998). Epoxidation of olefins by cytochrome 103. Ogharo, F, M. Filatov, and S. Shaik (2000).
P450: Evidence from site-specific mutagenesis for Alkane hydroxylation by cytochrome P450: Is
hydroperoxo-iron as an electrophilic oxidant. Proc. kinetic isotope effect a reliable probe of transition
Natl. Acad Sci. USA. 95, 3555-3560. state structure? Eur J. Inorg. Chem. 2455-2458.
91. Newcomb, M. and PH. Toy (2000). Hypersensitive 104. De Visser, S.P, F Ogliaro, PK. Sharma, and S.
radical probes and the mechanisms of cyto- Shaik (2002). Hydrogen bonding modulates the
chrome P450-catalyzed hydroxylation reactions. selectivity of enzymatic oxidation by P450:
Ace. Chem. Res. 33, 449^55. Chameleon oxidant behavior by Compound I.
92. Jin, S., T.M. Markis, T.A. Bryson, S.G. Sligar, and Angew. Chem. Int. Ed 41, 1947-1951.
J.H. Dawson (2003). Epoxidation of olefins by 105. De Visser, S.P, F Ogliaro, PK. Sharma, and
hydroperoxo-ferric cytochrome P450. J. Am. Chem. S. Shaik (2002). What factors affect the regiose-
Soc. 125, 3406-3407. lectivity of oxidation by cytochrome P450? A DFT
93. Shaik, S., M. Filatov, D. Schrder, and H. Schwarz study of allylic hydroxylation and double bond
(1998). Electronic structure makes a difference: epoxidation in a model reaction. J. Am. Chem. Soc.
Cytochrome P450 mediated hydroxylations of 124,11809-11826.
hydrocarbons as a two-state reactivity paradigm. 106. Cohen, S. and S. Shaik (2004). Quntum mechani-
Chem. Eur. J. 4, 193-199. cal/molecular mechanical study of the regioselec-
94. Schrder, D., S. Shaik, and H. Schwarz (2000). Two- tivity of camphor and cyclohexene oxidations by
state reactivity as a new concept in organometalHc cytochrome P450cam. In preparation; part of the
chemistry/ice. Chem. Res. 33, 139-145. Ph. D. thesis of Mrs. S. Cohen.
95. Shaik, S., S.P De Visser, F Ogliaro, H. Schwarz, 107. Yoshizawa, K. (2002). Theoretical study on kinetic
and D. Schrder (2002). Two-state reactivity mech- isotope effects in the C-H bond activation of
anisms of hydroxylation and epoxidation by alkanes by iron-oxo complexes. Coord. Chem. Rev.
cytochrome P-450 revealed by theory. Curr. Opin. 226,251-259.
Chem. Biol. 6, 556-567. 108. Yoshizawa, K., T. Ohta, M. Eda, and T. Yamabe
96. Sevin, A. and M. Fontecave (1986). Oxygen trans- (2000). Two-step concerted mechanism for the
fer from iron oxo porphyrins to ethylene. A semi- hydrocarbon hydroxylation by cytochrome P450.
empirical MO/VB approach. J. Am. Chem. Soc. Bull. Chem. Soc. Jpn. 73, 401^07.
108, 3266-3272. 109. Yoshizawa, K., Y. Shiota, and Y Kagawa (2000).
97. De Visser, S.P, F Ogliaro, and S. Shaik (2001). Energetics for the oxygen rebound mechanism
Stereospecific oxidation by Compound I of of alkane hydroxylation by the iron-oxo species of
Computational Approaches to Cytochrome P450 Function 85

cytochrome P450. Bull. Chem. Soc. Jpn. 73, 118. De Visser, S.P, D. Kumar, and S. Shaik (2004).
2669-2673. How do aldehyde side products occur during
110. Yoshizawa, K., Y. Kagawa, and Y. Shiota (2000). alkene epoxidation by cytochrome P450? Theory
Kinetic isotope effects in a C-H bond dissociation reveals a state-specific multi-state scenario where
by the iron-oxo species of cytochrome P450. the high-spin component leads to all side products.
J. Phys. Chem. B 104, 12365-12370. J. Inorg. Biochem. in press.
111. Yoshizawa, K., T. Kamachi, and Y. Shiota (2001). A 119. De Visser, S.P and S. Shaik (2003). A proton-shut-
theoretical study of the dynamic behavior of alkane tle mechanism mediated by the porphyrin in ben-
hydroxylation by a compound I model of cyto- zene hydroxylation by cytochrome P450 enzymes.
chrome P450. ^^m. Chem. Soc. 123, 9806-9816. J.Am. Chem. Soc. 125, 7413-7424.
112. Hata, M., Y. Hirano, T. Hoshino, and M. Tsuda 120. Korzekwa, K.R., D.C. Swinney, and W.F Trager
(2001). Monooxygenation mechanism by cyto- (1989). Isotopically labeled chlorobenzenes as
chrome P-450. ^ ^ m . Chem. Soc. 123, 6410-6416. probes for the mechanism of cytochrome P450
113. Shaik, S., S. Cohen, S.P de Visser, PK. Sharma, catalyzed aromatic hydroxylation. Biochemistry
D. Kumar, S. Kozuchs et al. (2004). The 28, 9019-9027.
"rebound controversy": An overview and theoreti- 121. Rietjens, I.M.C.M., A.E.M.E Soffers, C. Veeger,
cal modeling of the rebound step in C-H hydroxy- and J. Vervoort (1993). Regioselectivity of
laion by cytochrome P450. Eur. J. Inorg. Chem. cytochrome P-450 catalyzed hydroxylation of flu-
207-226. orobenzenes predicted by calculated frontier
114. Guallar, V, B.F. Gherman, W.H. Miller, S.J. orbital substrate characteristics. Biochemistry 32,
Lippard, and R.A. Friesner (2002). Dynamics of 4801-4812.
alkane hydroxylation at the non-heme diiron center 122. Sharma, PK., S.P de Visser, and S. Shaik (2003).
in methane monooxygenase. J. Am. Chem. Soc. Can a single oxidant with two-spin states masquer-
124, 3377-3384. ade as two different oxidants? A study of the sul-
115. De Visser, S.P, F. Ogliaro, N. Harris, and S. Shaik foxidation mechanism by cytochrome P450. J. Am.
(2001). Multi-state epoxidation of ethene by Chem. Soc. 125, 8698-8699.
cytochrome P450: A quantum chemical study. 123. Groves, J.T., G.E. Avaria-Neisser, K.M. Fish, M.
J. Am. Chem. Soc. 123, 3037-3047. Imachi, and R.L. Kuczkowski (1986). Hydrogen-
116. De Visser, S.P, R Ogliaro, and S. Shaik (2001). deuterium exchange during propylene epoxidation
How does ethene inactivate cytochrome P450 en by cytochrome P450. J. Am. Chem. Soc. 108,
route to its epoxidation? A density functional 3837-3838.
study. Angew. Chem. Int. Ed. 40, 2871-2874. 124. Groves, J.T. and D.V Subramanian (1984).
117. Groves, J.T., K.-H. Ahn, and R. Quinn (1988). Hydroxylation by cytochrome P450 and
Cis-trans isomerization of epoxides catalyzed by metalloporphyrin models. Evidence for
ruthenium(II) porphyrins. J. Am. Chem. Soc. 110, allylic rearrangement. J. Am. Chem. Soc. 106,
4217^220. 2177-2181.
3
Structures of Cytochrome P450
Enzymes
Thomas L. Poulos and Eric R Johnson

1. Introduction a snapshot in time on where we now stand in P450


crystallography.
Much of what we know about the molecular level
structure-function relationships in P450s is based on
studies with the camphor monooxygenase system 2. Overall Architecture
from Pseudomonasputida. Given that P450cam was
the first P450 to be purified in sufficient quantities There are now a sufficient number of struc-
for structure-function relationships, it is not too sur- tures to safely state that the overall P450 fold is
prising that P450cam was the first P450 crystal quite conservative. Perhaps more surprising is that
structure to be solved. The high-resolution structure the P450 fold is unique and despite the many new
of P450cam was published in 1987^ and remained structures that have been solved since P450cam,
the paradigm for P450 structure-function studies no non-P450 structure has yet been found to share
until the P450BM3 structure was solved in 19931 the P450 fold. Thus, the P450 fold appears to be
Since then the rate at which new P450 structures are uniquely adapted for the heme-thiolate chemistry
being solved has increased dramatically and at pres- required for oxygen activation, the binding of
ent there are structures for 20 unique P450s on redox partners, and the stereochemical require-
deposit in the Protein Data Bank with others waiting ments of substrate recognition.
in the wings. Some of the new advances that have The structures of several P450s are shown in
been made since the last edition of this book are the Figure 3.1, while Figure 3.2 highlights some of the
structures of other redox components of P450 key secondary structural elements. Although the
monooxygenase systems, of the first electron trans- overall fold is maintained, the precise positioning
fer complex, of new substrate complexes, of the first of various structural elements differs substantially.
P450s from thermophilic organisms, and of the first In general, the closer to the heme, the more
membrane-bound P450. hi the present chapter, we conserved the structure, especially helices I and L,
summarize some of these more important recent which directly contact the heme. As expected,
findings with full recognition that the P450 struc- those regions controlling substrate specificity dif-
tural field is moving much more quickly than in the fer the most, especially the B' helix. For example,
time frame of previous editions of this book. As in P450eryF, the B' helix is oriented about 90°
a result, the current chapter must be considered from the orientation observed in P450cam. The

Thomas L. Poulos • Department of Molecular Biology and Biochemistry and the Program in Macromolecular
Structure, University of California, Irvine, Irvine, CA. Eric F. Johnson • Department of Molecular and
Experimental Medicine, The Scripps Research Institute, La JoUa, CA.

Cytochrome P450: Structure, Mechanism, and Biochemistry, 3e, edited by Paul R. Ortiz de Montellano
Kluwer Academic / Plenum Publishers, New York, 2005.

87
88 Thomas L. Poulos and Eric F. Johnson

CYP102(P450BM3)
CYP101(P450cam)

CYP119
Figure 3.1. A representative example of known P450 structures illustrating the common three-dimensional fold.
structures of Cytochrome P450 Enzymes 89

Figure 3.2. The structure of P450cam with key hehcal


segments labeled.

effect is a substantial change in local environment Figure 3.3. The Cys ligand "loop" in P450cam. The
dashed lines indicate key hydrogen bonding interactions
that is required for substrate selectivity.
that aid in stabilizing the Cys ligand.
Not too surprisingly, the most conserved ele-
ments of the P450 structure center on the heme-
thiolate oxygen activation chemistry. The most it is necessary to decrease rather than increase the
noteworthy is the p-bulge segment housing the Cys redox potential^^. As a result, the His ligand H-
ligand (Figure 3.3), just prior to the L helix. This bonds with a buried Asp residue which imparts
rigid architecture is required to both protect the Cys greater imidazolate character to the His thus lower-
ligand and hold it in place in order to accept H- ing the heme iron redox potential^ ^~^^.
bonds from peptide NH groups. This arrangement is The other highly conserved region involved in
not only found in all P450s but in two closely related O2 activation is the portion of helix I near the heme
proteins, nitric oxide synthase (NOS) and Fe (Figure 3.4). Thr252 is involved in a local helical
chloroperoxidase (CPO). Both NOS and CPO are distortion in P450cam such that the Thr side chain
heme-thiolate enzymes and like P450s catalyze OH donates an H-bond to a peptide carbonyl oxy-
monooxygenation reactions. Exactly as in P450, the gen that would normally be involved in an a-helical
Cys ligand in CPO accepts H-bonds from peptide H-bond. This Thr is not strictly conserved. For
NH groups^. NOS is similar except that one of the example, P450eryF contains Ala instead of Thr
H-bonds is provided by the indole ring N atom of a (Figure 3.4). Even so, P450eryF also exhibits a sim-
conserved Trp residue^^. Such an H-bonding ilar distortion in the I helix. In addition, a water mol-
arrangement is not unique to heme thiolate proteins ecule in P450eryF takes the place of the Thr side
but is a characteristic feature of proteins containing chain OH thus maintaining a very similar H-bond-
Cys-Fe ligation and was first observed in the ferre- ing pattern. This arrangement is thought to be quite
doxins^. These H-bonds aid in regulating the heme important in the proper delivery of protons to the
iron redox potential^' ^. Without such H-bonds, the iron-linked oxygen required for cleavage of the
redox potential would be too low for reduction by O-O bond thus generating the active Fe-0 hydrox-
redox partners. Thus, it appears that the protein must ylating species. The growing consensus is that
provide a suitable electrostatic environment around ordered solvent at the active site serves as the direct
the Cys ligand in order to maintain the redox poten- proton donor to the iron-linked dioxygen^^' ^^. The
tial in a physiologically accessible range. The same most revealing crystal structures that support this
is true for a close cousin to P450, the peroxidases. view are the P450cam-oxy complex^^ and resting
Here His serves as the axial ligand, but in this case. state ferric P450eryF^^. In the P450cam-oxy
90 Thomas L. Poulos and Eric F. Johnson

P450cam P450eryF

Figure 3.4. A comparison of the I helix region in P450cam and P450eryF. The large gray spheres indicate water
molecules that complete the hydrogen bonding network in P450eryF.

6-position
P450eryF
hydroxy I ated

2.8 A •wat....2;8A ^ ^
*: ^ , -OH- o.
/3.8A /'^ ^y^
-Fe- Ser246 ^ Glii360

6«dexoerythronolide B
or 6DEB

P450cam

&3 ^. 3.lA .O-


3.5A ...-wat., 2.^9A
••••OH
5-position y<^
hydroxylated
|
I V
Thr252
Fe-
Figure 3.5. A comparison of the solvent-mediated hydrogen bonding network in P450eryF and the oxy-complex
of P450cam^^. In both cases, a critically positioned water molecule very likely serves as the proton donor to bound
dioxygen.
structures of Cytochrome P450 Enzymes 91

complex, two new waters are found in the active P450s also are found in thermophiles. The first to
site, one of which is shown in Figure 3.5. This new be discovered was CYP119 from the acidother-
water is close to dioxygen and may participate in mophilic archaeon Sulfolobus solfataricus^^.
relaying protons to dioxygen. P450eryF also has a Subsequent cloning and expression showed that
water similarly positioned (Figure 3.5). P450eryF, CYPl 19 melts near 90 °C compared to P450cam
however, uses a substrate-assisted mechanism since which melts near 50°C^^ The crystal structure of
a substrate OH anchors the key water in place via H- CYPl 19 was first solved by Yano et alP followed
bonding. While the details of the proton shuttle by an independent structure determination by Park
machinery may differ from one P450 to the next, the et alP. The most notable difference between
surrounding protein groups and, in at least one case, CYPl 19 and other P450s is that CYPl 19 is much
the substrate, generally position solvent in the active smaller, consisting of only 368 residues. The
site for proton delivery to dioxygen resulting in difference in size is primarily due to a shorter
cleavage of the 0 - 0 bond. N-terminal segment and shorter surface loops. A
unique clustering of aromatic residues running
down one side of the molecule (Figure 3.6) is
3. P450s from Thermophiles thought to be the structural basis for thermal sta-
bility. Mutating one of these residues, Phe24, to
Given that P450s are found in such a wide Ser lowers the melting temperature about 10°C^^.
variety of organisms, it is not surprising that A more extensive mutagenesis analysis of the

Arg287

Figure 3.6. The structure of CYPl 19 showing the aromatic cluster that is thought to be responsible for thermal
stability. The cluster also contains an Arg residue (Arg287) which is sandwiched between two tyrosine residues and
hence, contributes to the Tr-stacking interactions of the aromatic cluster.
92 Thomas L. Poulos and Eric F. Johnson

P450BM3 CYP175A1

Figure 3.7. A comparison between P450BM3 and the thermal stable P450, CYP175A1, showing the salt bridge
networks in both proteins.

aromatic cluster shows that the melting tempera- endoplasmic reticulum. However, several mam-
ture can be lowered lO-lS^C^"^ further implicat- malian P450s that participate in the synthesis of
ing aromatic clustering as a key factor in thermal sterols, steroids, and bile acids are located on the
stability. matrix side of the mitochondrial inner membrane.
The second thermophilic P450 to be discovered A longer N-terminal polypeptide chain of roughly
was CYP175A1 from Thermus thermophilus^^ 30-50 amino acids precedes the catalytic domain
which melts at 88°C. Like CYP119, CYP175A1 in eukaryotic P450s and mediates membrane tar-
is also shorter than most other P450s and contains geting. In the case of mitochondrial P450s, the tar-
378 residues. CYP175A1 most closely resem- geting sequences are cleaved during import of the
bles P450BM3 but with shorter surface loops. protein into mitochondria^^. In contrast, the leader
CYP175A1 lacks the aromatic clustering observed sequence of microsomal P450s is retained and is
in CYPl 19 and hence, the structural basis for ther- co-translationally inserted into the endoplasmic
mal stability lies elsewhere. Yano et alP carried reticulum^^. The insertion process stops at the end
out a detailed comparison of salt bridge networks of a hydrophobic stretch of roughly 20 amino acid
in various P450s and found that CYP175A1 con- residues, and the catalytic domain of microsomal
tains a larger fraction of salt bridges that contain P450s resides on the cytoplasmic side of the endo-
three or more residues. As shown in Figure 3.7, plasmic reticulum. A short region that generally
CYP175A1 has 26 residues involved in 8 salt contains positively charged residues links the cat-
bridge networks while the closest homologue to alytic domain to a conserved proline rich motif at
CYP175A1, P450BM3, has 14 residues participat- the N-terminus of the structurally conserved P450
ing in 4 salt bridge networks. fold. The N-terminal transmembrane domain
extends across the membrane and is unlikely to be
closely associated with the catalytic domain
4. Membrane P450s (Figure 3.8). This N-terminal domain is not
required for fiinction as illustrated by the
In contrast to prokaryotic P450s, eukaryotic expression and successful reconstitution of several
P450s are generally membrane-bound proteins. P450 monooxygenases in which this region was
Most eukaryotic P450s are incorporated into the deleted28-32.
structures of Cytochrome P450 Enzymes 93

P450 2C5

Phospholipid Bilayer

Figure 3.8. Hypothetical model for the membrane binding of microsomal P450s. The cartoon depicts the
experimentally determined fold of the modified CYP2C5 catalytic domain, PDB: 1N6B, attached to a hypothetical
model for the N-terminal transmembrane helix. The latter is flanked by a modeled array of phospholipid molecules
depicted as CPK atoms. The heme and bound substrate, DMZ, of CYP2C5 are also rendered as CPK atoms.

Another major difference relative to prokaryo- interact more extensively than their microsomal
tic P450s is the insertion of a longer polypeptide counterparts with the hydrophobic core of the
chain between helices F and G in eukaryotic lipid bilayer because detergents are required to
P450s. This region exhibits two short helices, release mitochondrial P450s from the membrane.
F' and G', in the structures of CYP2C5 (Figure In contrast, the catalytic domain of microsomal
3.9) and CYP2B4, a single, longer F' helix in P450 CYP2C5 is released by high salt buffers when it is
2C8, and a more relaxed coiled conformation in expressed as a truncated construct that does not
CYP2C9. This portion of the structure is in close contain the N-terminal transmembrane helix^^' ^^.
juxtaposition to the region between the proline- The model shown in Figure 3.8 is consistent
rich motif at the N-terminus of the catalytic with additional observations. Antibody epitope
domain and helix A, and together they form a mapping studies indicate that extensive portions of
hydrophobic surface near the N-terminus of the the surfaces of drug metabolizing P450s are acces-
protein^^. Epitope mapping studies of the related sible to the antibodies, reviewed in ref [35]. As
enzyme CYP2B4^'^ suggest that this portion of the shown in Figure 3.10, most of the epitopes reside
structure is in close proximity to and possibly along the edges of the protein with the exception
buried in the membrane as illustrated in Figure 3.8. of the tip of the protein near the N-terminus of the
Similar interactions may underlie the binding of catalytic domain. The tilt of the heme relative
mitochondrial P450s to the matrix surface of to the membrane has been estimated for CYP17
the inner membrane. As mitochondrial P450s lack and CYP21 based on the rate of decay of the
the N-terminal transmembrane helices found in absorption anisotropy following photodissociation
microsomal P450s, the catalj^ic domain must of carbon monoxide complexes of each protein.
94 Thomas L. Poulos and Eric F. Johnson

Helix G*

Helix F'

Meander

Figure 3.9. A side view showing the overall fold of modified CYP2C5. The largest insertions of additional
polypeptide chain relative to prokaryotic P450s occur for the N-terminal region depicted in Figure 3.8, the meander
region, and between helices F and G. The latter exhibits two short helical regions labeled F' and G'.

Figure 3.10. CPK rendering of the (A) proximal and (B) distal surfaces of modified CYP2C5. Antibody epitopes
recognized when the protein is bound to microsomal membranes are dark gray, as reviewed in ref [40]. Several
conserved amino acid side chains that have been implicated in P450 reductase interactions with CYP2B4'^^ are
medium gray. The orientation of the protein is similar to that depicted in Figure 3.8 with the N-terminus of the
catalytic domain positioned toward the bottom of the figure.
structures of Cytochrome P450 Enzymes 95

The results of these studies suggest that the angle insertion is unclear, but it resides in close proxim-
for the orientation of the heme relative to the ity to the probable site of P450 reductase binding
membrane surface is either 50° or 70° (See ref [36]). as discussed in the next section.
The latter value is consistent with the orientation
seen in Figure 3.8 and leads to significant solvent
exposure for the surfaces of the catalytic domain.
Atomic force microscopy experiments estimate 5. Electron Transfer Complexes
that the height of P450 2B4 above a model phos-
pholipid membrane is roughly 35-45 nm^^. This An important advance in understanding P450
would require a portion of the protein to be buried monooxygenase electron transfer systems was the
in the membrane, and this is likely to be the solution of the P450 reductase structure"^^ With
hydrophobic region near the N-terminus of the the addition of the first mammalian P450 struc-
catalytic domain. Studies of the association of ture, CYP2C5, the way is now open for under-
P450 2B4 with Langmuir-Blodget, phospholipid standing how these two microsomal proteins
monolayers indicate that the protein displaces an interact and transfer electrons. Nevertheless, the
area that is larger than a single transmembrane Bacillus megaterium fatty acid monooxygenase
helix^^. This result would be consistent with the system, P450BM3, has so far served as the best
penetration of the hydrophobic tip of the protein model system for understanding reductase-P450
into the lipid layer. A lower angle of the heme plane interactions. Although P450BM3 is a bacterial
would juxtapose a larger surface of the helix F-G enzyme, P450BM3 is more closely related in
region with the membrane surface, which is hydrated sequence, structure, activity, and redox partner to
and largely formed by the zwitterionic phospholipid microsomal P450s than to other bacterial P450s.
head groups. A steeper angle would bury the The unique feature of P450BM3 is that the
hydrophobic tip deeper into the membrane. The diflavin P450 reductase is linked to the C-terminal
propensity of the helix F to helix G region to form end of the heme domain thus giving a catalytically
hehcal structures rich in aromatic residues would self-sufficient enzyme.
enable this region to penetrate more deeply into the A significant advance was made when
membrane because the hydrogen bonding in the Sevrioukova et al.^^ succeeded in crystallizing a
helices would diminish the energetic cost of burying P450BM3 construct that contains just the heme
the peptide bonds in a hydrophobic environment^^. and FMN domains. The structure (Figure 3.11)
Membrane interactions with the catalytic shows that the FMN domain docks on the proximal
domain of microsomal P450s could promote surface of the P450, which was expected based on
the transfer of hydrophobic substrates from the complementary electrostatic surfaces and mutage-
membrane to the P450 and could also orient nesis studies. Nevertheless, the heme-FMN con-
the protein to facilitate interactions with P450 struct used to solve this structure was missing the
reductase as the two proteins diffuse along the sur- FAD domain and the linker connecting the heme
face of the endoplasmic reticulum. Mutagenesis and FMN domains had been proteolyzed during
experiments'^^ suggest that a group of relatively crystallization, thus raising the possibility that the
conserved, positively charged amino acids on the structure is an artifact of crystallization. In addi-
proximal surface of CYP2B4 interact with the tion, the structure of the P450BM3 complex is not
reductase (Figure 3.10). Significant insertions of compatible with the P450 reductase structure. As
additional amino acid residues relative to most shown schematically in Figure 3.12, the FMN and
prokaryotic P450 structures are also present on the FAD in P450 reductase form a direct nonbonded
proximal surface of the protein, and these are also contact. In order for the FMN domain to dock onto
in the structure of P450BM3. The additional the proximal surface of P450, there must be a large
residues form the J' helix extend a portion of the structural change which enables the FAD and
polypeptide chain termed the meander region FMN domains to move apart (Figure 3.12). Indeed,
that precedes the beta-turn containing the cysteine there are good indications that the linker connect-
that provides the axial ligation for the heme. The ing the FAD and FMN domains is quite flexible.
corresponding region of CYP2C5 is illustrated One of the more important studies related to the
in Figure 3.9. The functional importance of this question of domain flexibility is the structure of
96 Thomas L. Poulos and Eric F. Johnson

chemical modification studies. Sevrioukova


et al.^^ changed key residues on the heme domain
that lie at the interface between the heme and
FMN domains. Replacing Leul04 of P450BM3
with a Cys (Figure 3.11) at the interface should
not alter binding or electron transfer because
replacing Leu with a smaller side chain should not
cause any steric problems in forming the proper
heme complex. However, covalent modification of the
mutant Cys 104 side chain with a large fluo-
rophore should interfere with electron transfer.
For these studies laser flash photolysis was used
wherein a laser flash photoreduced a potent reduc-
tant, deazariboflavin, which in turn reduces the
FMN in the complex. The reduced FMN semi-
quinone then reduces the P450 heme. As pre-
dicted, mutation of Leu 104 to Cys had no effect,
while chemical modification of Cys 104 dramati-
cally decreased the FMN-to-heme electron trans-
fer rate, thus implicating Leu 104 as an important
residue in forming the proper electron transfer
complex.
Figure 3.11. Structure of complex formed between A second prediction from the P450BM3 elec-
the heme and FMN domains in P450BM3. The FMN tron transfer complex structure that can be tested
domain docks into the concave depression on the heme is the electron transfer path. The heme-FMN
domain proximal surface. domain interface is shown in Figure 3.13. A par-
ticularly noteworthy feature at the interface is the
participation of water molecules. Waters form
the homologous reductase domain in sulfite reduc- H-bonding bridges between the two domains
tase. This enzyme resembles P450BM3 in having a while there are only two direct H-bonds between
heme domain attached to a FMN/FAD reductase side chains. The closest point of contact between
domain. The FAD domain of sulfite reductase is the two domains places the FMN about 4 A from
very similar to the FAD domain of P450 reductase the peptide backbone of Gln387. The peptide
with an rms deviation of between 1.3 and 1.5 A"^^. chain from Gln387 to the heme ligand, Cys400,
However, the FMN domain is not visible in elec- could constitute an electron transfer path. To test
tron density maps in four different crystal forms this hypothesis, Gln387 was converted to Cys and
indicating that the FMN-FAD linker domain is modified with (4-bromomethyl-4'-methylbipyri-
quite flexible allowing the FMN domain to adopt dine)[bis(bipyridine)]ruthenium(II)'^^. The cova-
multiple orientations relative to the FAD domain. lently attached Ru(II) is photoreduced, and the
As it must, the large void in the crystal lattice can rate of reduction of the heme Fe(III) to Fe(II) by
accommodate the FMN module, but packing con- the photogenerated Ru(I) was followed. The same
siderations show that the possible interactions experiment was carried out with Ru(II) attached to
between the FMN and FAD modules are quite Cys60. Both Cys60 and Cys387 are about the
different than in P450 reductase"^^. This raises the same distance from the heme but electron transfer
possibility that the P450 reductase structure may from Cys60 to Ru(II) must make "through-space"
have captured the FMN domain in one possible jumps, while there is a continuous covalent con-
orientation while the P450BM3 electron transfer nection between Cys386-Ru(II) and the heme lig-
complex captured the orientation of the FMN and, Cys400. In the case of Cys387-Ru(II), the
domain required for proper docking to P450. heme iron was reduced at a rate of 4.6 X 10^ s~^
The P450BM3 electron transfer complex while Cys60-Ru(II) did not reduce the heme iron.
structure also is consistent with mutagenesis and These results indicate that if the crystal structure
structures of Cytochrome P450 Enzymes 97

reductase

P450
Figure 3.12. The crystal structure of P450 reductase^^ Note that the FMN and FAD are in direct contact. The
schematic diagram illustrates that it may be necessary for the FMN and FAD domains to separate to enable the FMN
domain to dock onto the P450 prior to the FMN-to-heme electron transfer reaction.

of P450BM3 electron transfer complex is shown in Figure 3.14 together with the crystal
functionally relevant, then the electron transfer structure of the adrenodoxin-adrenodoxin reduc-
reaction can readily proceed along the direct point tase complex^^. The Pdx and adrenodoxin (Adx)"^^
of contact between the FMN and heme domain. structures clearly are very similar. However, Pdr
Another recent advance in P450 electron trans- and adrenodoxin reductase (Adr)^^ exhibit large
fer is the solution of both the putidaredoxin differences. The FAD site is less exposed in Pdr
(Pdx)"^^ and putidaredoxin reductase (Pdr)"^^ crys- owing to an additional p-pair situated near the
tal structures. Thus, the structure of all compo- cofactor. In addition, the C-terminal region of Pdr
nents of the P450cam monooxygenase system (Figure 3.14) is positioned very differently than in
now are known. The Pdx and Pdr structures are Adr. Note that this segment in Pdr blocks part of
98 Thomas L. Poulos and Eric F. Johnson

W574,
N573

Figure 3.13. A close-up view of the interface formed between the heme and FMN domains in the P450BM3
electron transfer complex. The heme domain is more darkly shaded. There are only two direct H-bonds between the
two domains: Hisl00(heme)-Glu494(FMN) and Asnl01(heme)-Arg498(FMN). The remainder of the electrostatic
interactions are formed by bridging water molecules. Note that the FMN directly contacts the heme domain near
Gln387. The section of polypeptide leading from Gln387 to the Cys400 ligand and heme could provide a selective
electron transfer conduit.

the binding site for Adx in the Adx-Adr complex. known, a good deal has been learned about the
Therefore, if Pdx and Pdr form a complex similar primary forces involved in forming the com-
to the Adr-Adx complex, then the C-terminal plexes. Isothermal titration calorimetry shows
domain in Pdr must move. This seems unlikely that the Pdr-Pdx complex is entropically driven
since the C-terminal region that would be required suggesting that nonpolar interactions dominate'^.
to move is composed of p-sheets that tie this In contrast, electrostatic forces dominate the
region to the main body of the protein. Therefore, Pdx-P450cam complex. These equilibrium ther-
Pdx forms a complex with Pdr that must be quite modynamic studies agree with kinetic results
different than the Adx-Adr complex. Also shown obtained using laser flash photolysis methods to
in Figure 3.14 is the glutathione reductase struc- measure the P450-Pdx and Pdx-Pdr electron
ture^', which more closely resembles Pdr than transfer rates'"^.
does Adr. Note that both Pdr and glutathione Despite the lack of structural data on the com-
reductase have the common domain similarly plexes, Pochapsky et al. have developed a hypo-
positioned which limits exposure of the FAD. This thetical model of the P450cam-Pdx complex
similarity is especially interesting since Pdr using the crystal structure of P450cam and NMR
exhibits an NAD(H)-dependent dithiol/disulfide structure of Pdx''. The Pochapsky model was
oxidoreductase activity similar to glutathione developed well before the crystal structure of the
reductase activities'^. Whether or not this unex- P450BM3 complex was solved and was based on
pected Pdr activity is physiologically relevant data derived from NMR, mutagenesis, and com-
remains to be seen. putational studies. Quite interestingly, both the
Although the structures of the various electron P450cam-Pdx and P450BM3 complexes utilize
transfer complexes in the P450cam system are not the same proximal surface of P450 for the docking
structures of Cytochrome P450 Enzymes 99

Glutathione reductase

2Fe-2S

Figure 3.14. Structures of the adrenodoxin (Adx)-andrenodoxin reductase (Adr) complex, putidaredoxin (Pdx),
and putidaredoxin reductase (Pdr). For comparisons, the structure of glutathione reductase also is shown.

of redox partners (Figures 3.11 and 3.15). This as anticancer agents. P450BSP from Bacillus sub-
makes sense since the proximal region is the tilis is a novel enzyme that has the P450 fold but
closest approach of heme to the surface and thus catalyzes the peroxide-dependent hydroxylation of
affords the closest approach of redox active fatty acids^^. As shown in Figure 3.17, an OH
cofactors. group is attached to the polar carboxyl end of the
substrate which means the carboxyl head group
must be positioned deep in the active site.
6. Substrate Complexes P450BSP has adapted to accommodate the car-
boxyl group by having an Arg residue on the I helix
At present, the structure of five prokaryotic that electrostatically stabilizes the carboxyl head
P450-substrate complexes are known (Figures group (Figure 3.17). In the proposed peroxide acti-
3.16 and 3.17). P450epoK, the newest member of vation mechanism, the fatty acid carboxyl group
this group^^, catalyzes the 12,13-epoxidation of serves an acid-base catal3^ic function^^ similar to
epothilones (Figure 3.18) in Streptomyces coeli- the distal histidine in peroxidases. P450BSp pro-
color^^. Like Taxol, epothilones block microtubule vides a fascinating new example on the adaptability
depolymerization thus arresting the cell cycle in the of P450s to the requirements of different metabolic
G2-M phase. As a result, epothilones show promise pathways.
100 Thomas L. Poulos and Eric F. Johnson

F, G, B' helices, and the F/G loop (Figure 3.19).


The B' helix is rotated 90° in P450epoK com-
pared to P450cam. This reorientation opens the
substrate-binding pocket thus making room for
the thiazole ring of the substrate. The F and G
helices do not superimpose well and the F/G loop
adopts a substantially different conformation.
heme

7. Conformational Adaptations
to Substrates and Inhibitors

An unexpected insight into P450 dynamics


came from the S. solfataricus CYPl 19 structure^^,
the first known thermal stable P450^^. The
structure was solved in two crystal forms: one
P450cam with imidazole and one with phenylimidazole
coordinated to the heme iron. Compared to the
Figure 3.15. The P450cam-Pdx complex based on
phenylimidazole complex, in the imidazole com-
NMR and modeling studies^^. Pdx docks onto the
proximal surface of P450cam which is similar to the plex, the C-terminal end of the F helix unwinds
complex formed in P450BM3 (Figure 3.11). which increases the length of the F/G loop thus
allowing the loop to dip down into the active and
interact with imidazole ligand (Figure 3.20). In
The size and shape of the various substrates effect, the protein shapes itself around the ligand.
shown in Figure 3.16 are sufficiently diverse that Conformational adaptations also contribute to
the structural basis for what controls substrate the capacity of mammalian drug and xenobiotic
specificity can, at least in part, be understood. metabolizing enzymes to recognize structurally
As expected, all substrates are situated such that diverse substrates. Rabbit microsomal CYP2C5,
the atom to be hydroxylated is within 4-5 A of a drug and steroid metabolizing enzyme, has been
the heme iron. Thus, regio- and stereo-selective co-crystallized with substrates of different sizes,
hydroxylation by the hypothetical Fe(IV)-0 flexibility, and polarity. Figure 3.21 depicts the
species is achieved by specific protein-substrate binding of the anti-inflammatory drug diclofenac
interactions that hold the substrate in the correct to CYP2C5 that catalyzes the 4'-hydroxylation of
position. The exception is P450BM3. The struc- the dichlorophenyl ring of diclofenac. Diclofenac
ture of the P450BM3 heme domain with palmi- is positioned in CYP2C5 so that the flat surface of
toleic acid^^ and N-pamitoylglycine^^ show that the dichlorophenyl ring faces the heme Fe^^. The
the fatty acid substrate is ===7-8 A from the iron 4'-hydroxylation of diclofenac is likely to proceed
which is too far for hydroxylation. However, NMR through the formation of an epoxide intermediate
results indicate that the substrate moves to be formed by the addition of the iron oxo intermedi-
within 3 A of the iron upon reduction from Fe(III) ate to the pi-electron system. The epoxide inter-
to Fe(II)^^ Precisely how reduction is linked to mediate would rearrange to form the 4'-hydroxy
such a large repositioning of the substrate remains product. The 3' and 4' carbons are 4.4 and 4.7 A
unknown. from the heme Fe, respectively^^.
P450cam and P450epoK represents the two Multiple sites of oxidation are frequently seen
extremes of substrate size and shape. Hence, a for substrates of drug metabolizing enzymes. This
comparison between these two structures provides is likely to reflect either the capacity of the active
some insights on which regions of the structure site to bind the substrate in more than one orienta-
change most in response to the requirements of tion/location or motion of substrate within the
substrate specificity. The two regions that differ active site cavity. The former case is evident for
the most between P450epoK and P450cam are the the structure of CYP2C5 complex with DMZ,
structures of Cytochrome P450 Enzymes 101

P450cam P450BM3

Figure 3.16. Active site structures of various P450-substrate complexes. The arrow indicates the carbon atom that
is hydroxylated.

4-methyl-7V-methyl-A^-(2-phenyl-2i/-pyrazol-3- primary site of hydroxylation (>98%) is the ben-


yl)benzenesulfonamide (see ref. [63], Wester et al. zyllic methyl group. One conformation of DMZ
2003b). DMZ is a substrate for CYP2C5 as well places the benzylic methyl group 4.4 A from the
as for each of the four human CYP2C enzymes^"^. heme iron, and the reaction is likely to occur effi-
Electron density maps and in silico docking stud- ciently by a hydrogen abstraction mechanism. The
ies indicate that the substrate binds in two alter- second conformation places the other end of the
nate conformations, Figure 3.22. For DMZ, the molecule closest to the heme. However, DMZ is
102 Thomas L. Poulos and Eric F. Johnson

Figure 3.17. The substrate complex in P450BSp^^. This P450 utiUzes peroxide to oxidize the atom indicated by
the arrow. The I helix utilizes an arginine to help stabilize the carboxyl group of the substrate.

not located optimally for hydroxylation in this the enzyme for diclofenac and the regioselective
conformation as it is >5.9 A from the heme iron, hydroxylation of the dichlorophenyl group.
and the phenolic product accounts for < 1% of the Human CYP2C9 exhibits similar regioselectivity
total products. The two conformations of DMZ for the oxidation of diclofenac and displays a high
overlap but occupy distinct portions of the active catalytic efficiency for the reaction. In contrast, the
site cavity, which is larger than the substrate. other human CYP2C enzymes generally exhibit a
CYP2C5 exhibits adaptive changes when sub- much lower catalytic efficiency and a more relaxed
strates bind. The largest, adaptive changes involve regioselectivity in that they hydroxylate both rings
alterations in the conformations of the helix B-C of the substrate.
region, in the locations of helices F and G, and the
conformation of the region between the F and G
helices. A comparison of the diclofenac and DMZ 8. Conformational Dynamics
complexes illustrates differences that occur when for Substrate Access
bound substrates that are different in size, shape,
and polarity (Figure 3.23). The change in the posi- Substrate binding may involve rather large con-
tion of the F helix reflects differences in the space formational changes. Once the P450cam structure
occupied by the two substrates near the heme Fe. became available, an immediate puzzle was how
Changes in the conformation of the B' helix reflect camphor gains access to the active site since the
adaptations that maximize interactions with each substrate is buried and there is no obvious opening.
substrate and, in the case of diclofenac, that The substrate-free and -bound structures showed
accommodate residual hydration in the distal por- no differences although substrate-free P450cam
tion of the active site that is not occupied by the exhibited much higher thermal motion in the B', F,
substrate (Figure 3.23). These active site water and G helices suggesting that the these regions
molecules are ordered in a pattern that permits must move to allow substrate to enter the active
extensive intermolecular hydrogen bonding site^^. That this region is quite flexible was demon-
between water molecules, the protein and the sub- strated by binding of a ferrocene label to Cys85^^.
strate. This hydrogen bonding network is anchored The positioning of the ferrocene into the active site
by the polar carboxyl group of diclofenac and results in unfolding of the B' helix (Figure 3.24)
polar side chains in the distal portion of the active and exposure of the active site. Although these
site. These interactions contribute to the affinity of changes may not mimic what happens when
structures of Cytochrome P450 Enzymes 103

P450cam
OH
Camphor

P450BM3

Fatty acid

P450eryF

6-deoxyerythronolide B

OH

O OH O

Epothilone B
Figure 3.18. Reactions catalyzed by various P450s.

substrate binds and product leaves, this result does reporter group at the enzyme surface by attaching
support the view that the B' helix region can open it to a substrate analog that binds in the active site
to allow substrate to enter. Additional support for using a linker of the approximate length and size of
this view is provided by the structures of P450cam the substrate access channel^^' ^^. As shown in
complexed with tether compounds. The tether Figure 3.25, the tether molecule occupies a contin-
compounds are designed to position a fluorescent uous channel from the active site to the surface of
104 Thomas L. Poulos and Eric F. Johnson

F/G helix
B' heli

Figure 3.19. Differences between elements of structure that shape the substrate-binding pocket in P450cam
(light shading) and P450epoK (dark shading). Note the differences in the position of the F/G loop and B' helix.

G Helix

F/G Loop
phenylimidazole

Figure 3.20. Ligand-induced conformational changes in CYP119. Compared to the phenylimidazole complex
(dark shading), the C-terminal end of the F-helix in the imidazole complex unfolds which lengthens the F/G loop thus
allowing the F/G loop to dip into the active site and interact with the iron-linked imidazole. Since phenylimidazole is
larger than imidazole, the F/G loop cannot remain positioned in the active site complex. Therefore, the F/G helical
region and loop "shapes" itself around the ligand bound in the active site.

P450cam that is created by displacement of helix B' bound in the active site. To obtain the substrate-
and the F-G loop. free and -bound structures, DTT had to be back-
The early work with P450cam, however, pre- soaked out or camphor soaked in. The relatively
sented some experimental limitations. The initial tight crystal lattice of P450cam very likely pre-
diffraction quality crystals of P450cam had DTT vents the substrate-free structure from adopting
structures of Cytochrome P450 Enzymes 105

Figure 3.21. Diclofenac bound to CYP2C5, PDB: 1NR6. Hydroxylation of diclofenac at the 4' carbon is likely to
proceed through an intermediary epoxidation of the 3'-4' carbon bond of the substrate. The 3' and 4' carbons are
positioned at 4.4 and 4.7 A, respectively, from the heme Fe. Polar interactions of the substrate carboxylate lead to a
high degree of regioselectivity for oxidation of diclofenac by CYP2C5.

the open structure, although it remains unclear the surface of the I helix. This motion closes off
whether or not an open conformation is stable or the entry channel indicating that substrates enter
is only a transient conformer. near the F/G loop region which is similar to
The first clear indication that conformational P450cam.
changes are important in substrate binding was the Structures of several P450s exhibit open struc-
structure of palmitoleic acid bound to P450BM3^^ tures in the absence of substrates. P450nor is a
which was followed by a higher resolution struc- novel, soluble eukaryotic P450 that reduces nitric
ture^^. A solvent accessible surface diagram oxide. The enzyme is directly reduced by NADH
(Figure 3.26) illustrates how the substrate access which binds directly to an open cleft between the
channel is open in the substrate-free structure and F-G loop and the N-terminal (B-sheet system of
closed in the substrate-bound structure. Quite the enzyme^ ^ Two recently determined structures
interestingly, the experimentally observed confor- for prokaryotic P450s that are thought to be
mational change was correctly predicted based on involved in the oxidation of relatively large antibi-
computational methods^^' ^^ before the substrate- otic compounds exhibit more open structures. One
bound crystal structure was solved. The main is OxyB''^, a P450 that is thought to be involved in
motion involves the F and G helices sliding over the synthesis of vancomycin by Amycolatopsis
106 Thomas L. Poulos and Eric F. Johnson

<1°/c

Figure 3.22. Binding of DMZ to CYP2C5, PDB: 1N6B. DMZ binds to the enzyme in two distinct locations
depicted by the DMZ molecules with light and dark gray bonds. One orientation (light gray bonds) places the
benzyllic methyl group of DMZ 4.4 A from the heme iron, and oxidation of the benzyllic methyl group accounts for
>98% of the observed products. The alternate orientation (dark gray bonds) places the phenyl ring at >6 A from the
heme Fe. Oxidation of the phenyl ring accounts for 1% of the products. A solvent accessible surface for the substrate-
binding cavity is depicted by the light gray mesh.

orientalis. Helices F and G are rotated out of the slight tightening of the active site but there is
active site in this structure. It is thought that the no indication of the sorts of large motions
natural substrate of the enzyme is a relatively observed with P450BM3. Caution must be
large, glycosylated heptapeptide. A similar posi- exercised here because the energetics of adopting
tioning of helices F and G is also seen in the struc- the open conformation must be balanced with
ture of CYP154C1 from S. coelicolor A3(2) the energetics of crystallization. With P450BM3,
(Figure 3.27) that oxidizes macrolide substrates^^. the open conformation was trapped due in
In contrast to the structure seen for P450BM3, the part to crystal contacts. It may be that P450epoK
region between helices B and C separates from the simply prefers to crystallize in the "closed" form
G helix to form a cleft that opens along helix I. with or without substrate bound. The assumption
P450epoK presents a different picture. Here here, of course, is that P450s must undergo
the substrate-free and -bound structures were open/close motions to allow substrate access even
separately crystallized^^, yet there is very little if we only observe the closed form in crystal
difference in structure. Substrate binding causes a structures.
structures of Cytochrome P450 Enzymes 107

Helix B'

Val 106

Helix I

Figure 3.23. Conformational adaptations for substrate binding to CYP2C5. Among other changes, the position of
hehx B' adapts to the presence of diclofenac and DMZ that differ in size and polarity. The DMZ complex is rendered
in light gray, and the diclofenac complex is depicted in dark gray. Ca traces are shown for the helix B to C region
and a portion of the helix I with the heme rendered as CPK atoms. The different positions of two active site side
chains. Leu 103 and Val 106, are rendered as ball and stick figures, as are the two substrates.

While it would appear that the F/G loop channel and closure of the other might provide a
region may provide the point of substrate entry means for substrate to enter via one route but
and conformational dynamics for many P450s, product depart via the other.
the CYP51 structure^"^ indicates that other access Eukaryotic P450s generally exhibit longer
channels may be used. While CYP51 exhibits the polypeptide chains between helices F and G than
normal P450 fold, there is an unprecedented break are seen in prokaryotic P450s, and this region
in the I helix (Figure 3.28). This break creates interacts more extensively with the N-terminal
a new opening that runs roughly parallel to the P-sheet region, potentially limiting the opening of
heme as opposed to the F/G loop entry point the protein in this direction. In addition, this region
which is perpendicular to the heme. Podust et al?^ is likely to interact with the membrane. In contrast,
suggest that if the F/G loop region were to the B' helix region is not closely associated with
open, then the new opening must close. This offers the rest of the protein, suggesting that the flexibil-
the possibility that a concerted opening of one ity of the helix B to helix C region is likely to shift
108 Thomas L. Poulos and Eric F. Johnson

B' helix unfolded


B' helix

camphor

Figure 3.24. A comparison of the structures of B' helix region in P450cam (dark shading) and the ferrocene
attached to Cys85^^. To accommodate the bulky ferrocene, the B' helix must unfold (light shading) and move fiirther
away from the main body of the protein.

the opening to the active site to a direction along substrate channel near the F/G loop is the same as
the helix I between the helix B' to helix C loop and that derived from the crystal structure but also pre-
helix G. This is supported by the observation that dicts other possible routes of entry in P450cam,
progesterone can be soaked into CYP2C5 in crys- P450BM3, and P450eryF In addition, novel routes
tals where the N-terminal p-sheet region and of ligand exit were found. These pathways may be
helices F and G are highly constrained by crystal favored in other P450s that exhibit different pack-
packing. The solvent channel is closed by a single ing interactions between the flexible components
H-bonding interaction between K241 of helix G to of the distal surface of the enzyme. These studies
the backbone carbonyl of VI06 in helix B' as well have also provided insights into the energeti-
as a relatively weak van der Waals contact of VI06 cally accessible motions available to the various
with H230 of helix G. Crystallization of CYP2B4 P450s.
in an open conformation^^ supports this view; an Overall, the current picture is that while the
open cleft is observed in this structure between the P450 fold is conservative, it is quite flexible and
helix B' to C loop, helix I and helix F to helix G can undergo rather large changes in response to
regions. the requirements of substrate specificity. The F/G
A theoretical analysis of substrate-binding helical region and B' helix are subject to the great-
routes has helped to clarify the picture^^. The est structural variation as well as flexibility. This
computational approach predicts that the main is understandable considering the importance the
structures of Cytochrome P450 Enzymes 109

Fluorescent Reporter Tether

Figure 3.25. The structure of P450cam complexed with a tether compound adamantane-1-carboxyhc acid-
5-dimethylaminonaphthalene-l-sulfonylamino-octyl-amide rendered as CPK atoms, PDB: ILWL. The heme is
rendered as a ball and stick figure. The tether compound occupies an open channel between helices F and G, helix
B' and the p-sheet domain with the fluorescein moiety residing on the surface and the adamantane moiety positioned
in the substrate-binding site.

substrate-free substrate-bound, access channel closed

access channel open

Figure 3.26. Solvent accessible surface diagrams of P450BM3 in the substrate-free and -bound forms.

F/G region and the B' helix play in substrate entry hand show quite clearly that homology modeling
and binding. Toward the goal of understanding has a major challenge because the most difficult
selectivity, homology modeling has become an regions to predict are precisely those regions that
increasingly popular tool in P450 research. There are functionally most important. In many cases,
are simply too many interesting P450s to expect the low degree of amino acid identity renders
the various crystal structures to be solved in a threading of these sequences onto experimentally
timely fashion. However, the structures we have in determined structures ambiguous. This problem
110 Thomas L. Poulos and Eric F. Johnson

Helix G

Helix F

Helix B

Figure 3.27. The structure of CYP154C1, PDB: IGWl. A view along the N-terminal end of helix I is shown
depicting the rotation of helices F and G up and away from the substrate-binding cavity. An open cleft runs through
the molecule above the heme. The helix B to C segment resides on the opposite of the cleft in association with the
P-sheet 1. The heme is rendered as CPK atoms.

I helix CYP51

I helix P450cam

P450cam CYP51
Figure 3.28. The top diagram shows the I helix in CYP51 (dark shading) and P450cam (light shading). The CPK
diagrams are viewed along the plane of the heme and illustrate how the break in the I helix leaves the heme pocket
openinCYPSl.
structures of Cytochrome P450 Enzymes 111

has been largely overcome for the relatively large 7. Adman, E., K.D. Watenpaugh, and L.H. Jensen
number of drug metabolizing enzymes in family 2 (1975). NH-S hydrogen bonds in Peptococcus aero-
because alignments with known structures such genes ferredoxin, Clostridium pasteurianum rubre-
as those of CYP2C and CYP2B families are doxin, and Chromatium high potential iron protein.
Proc. Natl. Acad. Sci. USA 12, 4854^858.
relatively straightforward. However, the experi-
8. Ueyama, N., T. Terakawa, M. Nakata, and
mentally determined structures of the family 2
A. Nakamura (1983). Positive shift of redox
enzymes exhibit significant differences in the con- potential of [Fe2S4(Z-cys-Gly-Ala-OMe)4]2-
formations of the flexible portions of the catalytic in dichloromethane. J. Am. Chem. Soc. 105,
sites that underlie their functional diversity. Thus, a 7098-7102.
critical need to determine structures of specific 9. Ueyama, N., N. Nishikawa, Y. Yamada, T. Okamura,
drug metabolizing enzymes remains in order to and A. Nakamura (1996). Cytochrome P-450 model
accurately model substrate enzyme interactions. (porphinato)(thiolatio)iron(III) complexes with and
double NH-S hydrogen bonds. J. Am. Chem. Soc.
118, 1286-1287.
10. Poulos, T.L. and B.C. Finzel (1984). Heme enzyme
Acknowledgments structure and function. In M.T. Meam (ed.). Peptide
and Protein Reviews, Vol. 4. Marcel Dekker, New
York, pp. 115-171.
TLP would like to thank members of the UCI 11. Chang, C.K. andT.G. Traylor (1973). Proximal base
P450 group, Huiying Li, Shingo Nagano, and influence on the binding of oxygen and
Irina Sevrioukova, and NIH grant GM33688. carbon monoxide to heme. J. Am. Chem. Soc. 95,
EFJ would like to thank his colleagues at TSRI, 8477-8479.
Guillaume Schoch, Mike Wester, Jason Yano, and 12. Doef, M.M., D.A. Sweigart, and P O'Brien (1983).
C. David Stout, as well as NIH Grant GM31001. Hydrogen bonding from coordinated imidazole
in ferric porphyrin complexes. Effect on the
Fe(III)/Fe(n) reduction potential. Inorg. Chem. 22,
851-852.
13. Nappa, M., J.S. Valentine, and PA. Snyder (1977).
References Imidazolate complexes of ferric porphyrins. J. Am.
Chem. Soc. 99, 5799-5800.
1. Poulos, T.L., B.C. Finzel, and A.J. Howard (1987). 14. Valentine, J.S., R.P Sheridan, L.C. Allen, and
High-resolution crystal structure of cytochrome PC. Kahn (1979). Coupling between oxidation state
P450cam. J. Mol Biol. 195, 687-700. and hydrogen bond conformation in heme proteins.
2. Ravichandran, K.G., S.S. Boddupalli, Proc. Nat. Acad Sci. USA 16, 1009-1013.
C.A. Hasermann, J.A. Peterson, and J. Deisenhofer 15. Banci, L., LI. Bertin, E.A. Pease, M. Tien, and
(1993). Crystal structure of hemoprotein domain of P. Turano (1992). ^H NMR investigation of man-
P450BM-3, a prototype for microsomal P450's. ganese peroxidase from Phanerochaete chrysospo-
5c/e«ce 261, 731-736. rium. A comparison with other peroxidases.
3. Sundaramoorthy, M., J. Temer, and T.L. Poulos Biochemistry 31, 10009-10017.
(1995). The crystal structure of chloroperoxidase: A 16. Vidakovic, M., S.G SHgar, H. Li, and T.L. Poulos
heme peroxidase-cytochrome P450 functional (1998). Understanding the role of the essential
hybrid. Structure 3, 1367-1377. Asp251 in cytochrome p450cam using site-directed
4. Crane, B.R., A.S. Arvai, D.K. Ghosh, C. Wu, mutagenesis, crystallography, and kinetic solvent
E.D. Getzoff, D.J. Stuehr et al (1998). Structure of isotope effect. Biochemistry 37, 9211-9219.
nitric oxide synthase oxygenase dimer with pterin 17. Benson, D.E., K.S. Suslick, and S.G. Sligar
and substrate. Science 119, 2121-2126. (1997). Reduced oxy intermediate observed in
5. Fischmann, T.O., A. Hruza, X.D. Niu, J.D. Fossetta, D251N cytochrome P450cam. Biochemistry 36,
C.A. Lunn, E. Dolphin et al (1999). Structural char- 5104-5107.
acterization of nitric oxide synthase isoforms reveals 18. Schlichting, L, J. Berendzen, K. Chu, A.M. Stock,
striking active-site conservation. Nat. Struct. Biol. 6, S.A. Maves, D.E. Benson etal. (2000). The catalytic
233-242. pathway of cytochrome p450cam at atomic resolu-
6. Raman, C.S., H. Li, R Martasek, V Krai, tion. Science 287, 1615-1622.
B.S. Masters, and T.L. Poulos (1998). Crystal struc- 19. Cupp-Vickery, J.R., O. Han, C.R. Hutchinson,
ture of constitutive endothelial nitric oxide synthase: and T.L. Poulos (1996). Substrate-assisted cataly-sis
A paradigm for pterin function involving a novel in cytochrome P450eryF. Nat. Struc. Biol. 3,
metal center. Cell 95, 939-950. 632-637.
112 Thomas L. Poulos and Eric F. Johnson

20. Wright, R.L., K. Harris, B. Solow, R.H. White, and 32. Pernecky, S.J., J.R. Larson, R.M. Philpot, and
P.J. Kennelly (1996). Cloning of a potential M.J. Coon (1993). Expression of truncated forms of
cytochrome P450 from the archaeon Sulfolobus liver microsomal P450 cytochromes 2B4 and 2E1 in
solfataricus. FEES Lett. 384, 235-239. Escherichia coli: Influence of NH2-terminal region
21. McLean, M.A., S.A. Maves, K.E. Weiss, S. Krepich, on localization in cytosol and membranes. Proc.
and S.G. Sligar (1998). Characterization of a Natl. Acad Sci. USA 90, 2651-2655.
cytochrome P450 from the acidothermophilic 33. Williams, PA., J. Cosme, V Sridhar, E.R Johnson,
archaea Sulfolobus solfataricus. Biochem. Biophys. and D.E. McRee (2000). The crystallographic struc-
Res. Comm. 252, 166-172. ture of a mammalian microsomal cytochrome P450
22. Yano, J.K., L.S. Koo, D.J. Schuller, H. Li, PR. Ortiz monooxygenase: Structural adaptations for mem-
de Montellano, and T.L. Poulos (2000). Crystal brane binding and functional diversity. Mol. Cell 5,
structure of a thermophilic cytochrome P450 from 121-132.
the archaeon Sulfolobus solfataricus. J. Biol. Chem. 34. De Lemos-Chiarandini, C, A.B. Frey, D.D. Sabatini,
275,31086-31092. and G. Kreibich (1987). Determination of the
23. Park, S.Y., K. Yamane, S. Adachi, Y. Shiro, membrane topology of the phenobarbital-inducible
S.A. Maves, K.E. Weiss et al. (2002). Thermophilic rat liver cytochrome P-450 isoenzyme PB-4
Cytochrome P450 (CYP119) from Sulfolobus using site-specific antibodies. J. Cell Biol. 104,
solfataricus: High resolution structural origin of its 209-219.
thermostability and functional properties. J. Inorg. 35. Von Wachenfeldt, C. and E.R Johnson (1995).
Biochem. 91,491-501. Structures of eukaryotic cytochrome P450 enzymes.
24. Puchkaev, A.V., L.S. Koo, and PR. Ortiz de In PR. Ortiz de Montellano (ed.), Cytochrome
Montellano (2003). Aromatic stacking as a determi- P450: Structure, Mechanism, and Biochemistry.
nant of the thermal stability of CYP119 from Plenum Press, New York.
Sulfolobus solfataricus. Arch. Biochem. Biophys. 36. Ohta, Y, S. Kawato, H. Tagashira, S. Takemori, and
409, 52-58. S. Kominami (1992). Dynamic structures of adreno-
25. Yano, J.K., F. Blasco, H. Li, R.D. Schmid, A. Henne, cortical cytochrome P-450 in proteoliposomes and
and T.L. Poulos (2003). Preliminary characteriza- microsomes: Protein rotation study. Biochemistry
tion and crystal structure of a thermostable 31, 12680-12687.
cytochrome P450 from Thermus thermophilus. 37. Bayburt, TH. and S.G. Sligar (2002). Single-
J. Biol. Chem. 278, 608-616. molecule height measurements on microsomal
26. Omura, T. and A. Ito (1991). Biosynthesis and intra- cytochrome P450 in nanometer-scale phospholipid
cellular sorting of mitochondrial forms of bilayer disks. Proc. Natl. Acad. Sci. USA 99,
cytochrome P450. Meth. Enzymol. 206, 75-81. 6725-6730.
27. Sakaguchi, M. and T. Omura (1993). Topology and 38. Shank-Retzlaff, M.L., G.M. Raner, M.J. Coon, and
biogenesis of microsomal cytochrome P-450s. In K. S.G. Sligar (1998). Membrane topology of
Ruckpaul and H. Rein (eds.). Medicinal cytochrome P450 2B4 in langmuir-blodgett mono-
Implications in Cytochrome P-450 Catalyzed layers. Arch. Biochem. Biophys. 359, 82-88.
Biotransformations. Akademie Verlag, Berlin. 39. White, S.H., A.S. Ladokhin, S. Jayasinghe, and
28. Von Wachenfeldt, C , T.H. Richardson, J. Cosme, K. Hristova (2001). How membranes shape protein
and E.F. Johnson (1997). Microsomal P450 2C3 is structure. J. Biol. Chem. 276, 32395-32398.
expressed as a soluble dimer in Escherichia coli fol- 40. Bridges, A., L. Gruenke, YT Chang, l.A. Vakser,
lowing modifications of its N-terminus. Arch. G. Loew, and L. Waskell (1998). Identification of
Biochem. Biophys. 339, 107-114. the binding site on cytochrome P450 2B4 for
29. Cosme, J. and E.E Johnson (2000). Engineering cytochrome b5 and cytochrome P450 reductase.
microsomal cytochrome P450 2C5 to be a soluble, J. Biol. Chem. 273, 17036-17049.
monomeric enzyme. Mutations that alter aggrega- 41. Wang, M., D.L. Roberts, R. Paschke, TM. Shea,
tion, phospholipid dependence of catalysis, and B.S. Masters, and J.J. Kim (1997). Three-
membrane binding. J. Biol. Chem. 28, 2545-2553. dimensional structure of NADPH-cytochrome P450
30. Li, YC. and J.Y.L. Chiang (1991). The expression of reductase: Prototype for FMN- and FAD-
a catalytically active cholesterol 7alpha-hydroxylase containing enzymes. Proc. Natl. Acad. Sci. USA 94,
cytochrome P450 in Escherichia coli. J. Biol. Chem. 8411-8416.
266, 19186-19191. 42. Sevrioukova, I.E., H. Li, H. Zhang, J.A. Peterson,
31. Sagara, Y, H.J. Barnes, and M.R. Waterman (1993). and T.L. Poulos (1999). Structure of a cytochrome
Expression in Escherichia coli of functional P450-redox partner electron-transfer complex.
cytochrome P450cl7 lacking its hydrophobic Proc. Natl. Acad Sci. USA 96, 1863-1868.
amino-terminal signal anchor. Arch. Biochem. 43. Gruez, A., D. Pignol, M. Zeghouf, J. Coves,
Biophys. 304, 272-278. M. Fontecave, J.-L. Ferrer et al. (2000). Four crystal
structures of Cytochrome P450 Enzymes 113

structures of the 60kDa falvoprotein monomer of solution structure of oxidized putidaredoxin, a 2-Fe,
the sulfite reductase indicate a disordered flavo- 2-S ferredoxin from Pseudomonas. Biochemistry
doxin-Hke module. J. Mol. Biol. 299, 199-212. 33, 6424-6432.
44. Sevrioukova, I.F., J.T. Hazzard, G. Tollin, and 56. Nagano, S., H. Li, H. Shimizu, C. Nishida,
T.L. Poulos (1999). The FMN to heme electron H. Ogura, PR. Ortiz de Montellano et al. (2003).
transfer in cytochrome P450BM-3. Effect of Crystal structures of epothilone-D bound,
chemical modification of cysteines engineered at epothilone-B bound, and substrate-free forms of
the FMN-heme domain interaction site. J. Biol. cytochrome P450epoK. J. Biol. Chem. 278,
Chem. 274, 36097-36106. 44886-^4893.
45. Sevrioukova, I.E., C.E. Immoos, T.L. Poulos, and 57. Tang, L., S. Shah, L. Chung, J. Carney, L. Kaz,
P. Farmer (2000). Electron transfer in the ruthenated C. Khosla et al. (2000). Cloning and heterologous
heme domain of cytochrome P450BM-3. Isr. J. expression of the epothilone gene cluster. Science
Chem. 40, 47-53. 287, 640-642.
46. Sevrioukova, I.E., C. Gracia, H. Li, B. Bhaskar, 58. Lee, D.-S., A. Yamada, H. Sugimoto, L Matsunaga,
T.L. Poulos (2003). Crystal structure of putidare- H. Ogura, K. Ichihara et al. (2003). Substrate recog-
doxin, the [2Fe-2S] component of the P450cam nition and molecular mechanism of fatty acid
monooxygenase system from Pseudomonas putida. hydroxylation by cytochrome P450 from Bacillus
J. Molec. Biol 333, 377-392. subtilis. J. Biol. Chem. 278, 9761-9767.
47. Sevrioukova, LE, H. Li, T.L. Poulos (2004). Crystal 59. Li, H. and T.L. Poulos (1997). The structure of the
structure of putidaredoxin reductase from cytochrome p450BM-3 haem domain complexed
pseudomonas putida, the final structural component with the fatty acid substrate, palmitoleic acid. Nat.
of the P450cam monooxygenase system. J. Mol. Struct. Biol. 4, 140-146.
Biol. 236, 889-902. 60. Haines, D.C., D.R. Tomchick, M. Machius, and
48. Muller, J.J., A. Lapko, G. Bourenkov, K. Ruckpaul, J.A. Peterson (2001). Pivotal role of water in the
and U. Heinemann (2000). Adrenodoxin reductase- mechanism of P450BM-3. Biochemistry 40,
adrenodoxin complex structure suggests electron 13456-13465.
transfer path in steroid biosynthesis. J. Biol. Chem. 61. Modi, S., M.J. Sutchffe, W.U. Primrose, L.Y. Lian,
116, 2786-2789. and G.C. Roberts (1996). The catalytic mechanism
49. Mlier, A., J.J. MUer, Y.A. Muller, H. Uhlmann, of cytochrome P450 BM3 involves a 6 A movement
R. Bernhardt, and U. Heinemann (1998). New of the bound substrate on reduction. Nat. Struc.
aspects of electron transfer revealed by the crystal Biol. 3, 414-417.
structure of a truncated bovine adrenodoxin, Adx(4- 62. Wester, M.R., E.E Johnson, C. Marques-Soares,
108). Structure 6, 269-280. S. Dijols, PM. Dansette, D. Mansuy (2003). The
50. Ziegler, G.A. and G.E. Schulz (2000). Crystal struc- structure of mammalian cytochrome P450 2C5
tures of adrenodoxin reductase in complex with complexed with diclofenac at 2.1 A
NADP(+) and NADPH suggesting a mechanism resolution: Evidence for an induced fit model of
for the electron transfer of an enzyme family. substrate binding. Biochemistry 42, 9335-9345.
Biochemistry'^9, 10986-10995. 63. Wester, M.R., E.E Johnson, C. Marques-Soares,
51. Mittl, RR.E. and G.E. Schulz (1994). The structure PM. Dansette, D. Mansuy, and CD. Stout (2003,
of glutathione reductase from Escherichia coli at submitted). The structure of a substrate complex of
1.86A resolution: Comparison with the enzyme mammalian cytochrome P450 2C5 at 2.3 A resolu-
from human erythrocytes. Protein Sci. 3, 799-809. tion: Evidence for multiple substrate binding
52. Sevrioukova, LE and T.L. Poulos (2002). modes. Biochemistry 42, 6370-6379.
Putidaredoxin reductase, a new function for an old 64. Marques-Soares, C , S. Dijols, A. Macherey,
protein. J. Biol. Chem. 277, 25831-25839. M.R. Wester, E.E Johnson, P.M. Dansette et al.
53. Aoki, M., K. Ishimori, and L Morishima (1998). (2003, submitted). Sulfaphenazole derivatives as
Roles of negatively charged surface residues of puti- tools for comparing cytochrome P450 2C5 and
daredoxin in interactions with redox partners in human cytochrome P450 2Cs: Identification of a
P450cam monooxygenase system. Biochim. new high affinity substrate common to those
Biophys.Acta 1386, 157-167. enzymes. Biochemistry 42, 6363-6369.
54. Sevrioukova, LE, XT. Hazzard, G. Tollin, and 65. Poulos, T.L., B.C. Finzel, and A.J. Howard
T.L. Poulos (2001). Laser flash induced electron (1986). Crystal structure of substrate-free Pseudo-
transfer in P450cam monooxygenase: Putidaredoxin monas putida cytochrome P450. Biochemistry 25,
reductase-putidaredoxin interaction. Biochemistry 5314-5322.
40, 10592-10600. 66. DiGleria, K., D.R Nickerson, H.A.O. Hill,
55. Pochapsky, T.C., X.M. Ye, G. Ratnaswamy, and L.L. Wong, and V Fulop (1998). Covalent attach-
T.A. Lyons (1994). An NMR-derived model for the ment of an electroactive sulfhydryl reagent in the
114 Thomas L. Poulos and Eric F. Johnson

active site of cytochrome P450cam as revealed by 73. Podust, L., Y. Kim, M. Arase, B. Neely, B. Beck,
the crystal structure on the modified protein. J. Am. H. Bach et al (2003). The 1.92 A structure of
Chem. Soc. 120, 46-52. Streptomyces coelicolor A3(2) Cypl54Cl: A new
67. Dmochowski, I.J., B.R. Crane, J.J. Wilker, monooxygenase that fiinctionalizes macrolide ring
J.R. Winkler, and H.B. Gray (1999). Optical detection systems. J. Biol. Chem. 278, 12214-12221.
of cytochrome P450 by sensitizer-linked substrates. 74. Podust, L.M., T.L. Poulos, and M.R. Waterman
Proc. Natl. Acad. Sci. USA 9, 12987-12990. (2001). Crystal structure of cytochrome P450
68. Dunn, A.R., A.M. Hays, D.B. Goodin, CD. Stout, R. 14alpha-sterol demethylase (CYP51) from
Chiu, J.R. Winkler et al. (2002). Fluorescent probes Mycobacterium tuberculosis in complex with
for Cytochrome P450 structural characterization azole inhibitors. Proc. Natl. Acad. Sci. USA 98,
and inhibitor screening. J. Am. Chem. Soc. 124, 3068-3073.
10254-10255. 75. Scott, E.E., Y.A. He, M.R. Wester, M.A. White, C.C.
69. Li, H. and T.L. Poulos (1995). Modeling protein sub- Chin, J.R. Halpert, E.F Johnson, and D. Stout
strate interactions in the heme domain of cytochrome (2003). An open confirmation of mammalian
P450BM-3. Acta Crystallogr. D 51, 21-32. cytochrome P450 2B4 at 1.6A resolution. Proc.
70. Paulsen, M.D. and R.L. Ornstein (1995). Dramatic Natl. Acad. Sci. USA 100, 13196-13201.
differences in the motions of the mouth of open and 76. Winn, P.J., S.K. Ldemann, R. Gauges, V. Lounnas,
closed cytochrome P450BM-3 by molecular and R.C. Wade (2002). Comparison of the
dynamics simulations. Proteins 21, 237-243. dynamics of substrate access channels in three
71. Park, S.Y., H. Shimizu, S. Adachi, A. Nakagawa, cytochrome P450s reveals different opening
I. Tanaka, K. Nakahara et al. (1997), Crystal structure mechanisms and a novel functional role for a
of nitric oxide reductase from denitrifying fungus buried arginine. Proc. Natl. Acad. Sci. USA 99,
Fusarium oxysporum. Nat. Struc. Biol. 4, 827-832. 5361-5366.
72. Zerbe, K., O. Pylypenko, F. Vitali, W Zhang, 77. Pochapsky, T, T.A. Lyons, S. Kazanis, T. Arakaki,
S. Rouset, M. Heck etal. (2002). Crystal structure of and G. Ratnaswamy (1996). A structure-based
OxyB, a cytochrome P450 implicated in an oxidative model for cytochrome P450cam-putidaredoxin
phenol coupling reaction during vancomycin biosyn- interactions. Biochimie 78, 723-733.
thesis. J. Biol. Chem. Ill, 47476-47485.
4
Electron Transfer Partners of
Cytochrome P450
Mark J.l. Paine, Nigel S. Scrutton, Andrew W. Munro, Aldo Gutierrez,
Gordon C.K. Roberts, and C. Roland Wolf

1. Introduction Although P450 redox partners are usually


expressed independently, "self-sufficient" P450
monooxygenase systems have also evolved through
Cytochromes P450 contain a heme center
the fusion of P450 and CPR genes. These fusion
where the activation of molecular oxygen occurs,
molecules are found in bacteria and fungi, the best-
resulting in the insertion of a single atom of
known example being P450 BM3, a fatty acid
oxygen into an organic substrate with the con-
(0-2 hydroxylase from Bacillus megaterium, which
comitant reduction of the other atom to water. The
comprises a soluble P450 with a fiised carboxyl-
monooxygenation reaction requires a coupled and
terminal CPR module (recently reviewed by
stepwise supply of electrons, which are derived
Munro^). BM3 has the highest catalytic activity
from NAD(P)H and supplied via a redox partner.
known for a P450 monooxygenase^ and was for
P450s are generally divided into two major classes
many years the only naturally occurring ftised sys-
(Class I and Class II) according to the different
tem known until the identification of a eukaryotic
types of electron transfer systems they use. P450s
membrane-bound equivalent fatty acid hydroxy-
in the Class I family include bacterial and mito-
lase, CYP505A1, from the phytopathogenic fungus
chondrial P450s, which use a two-component
Fusarium oxysporurrP. A number of novel P450 sys-
shuttle system consisting of an iron-sulfur protein
tems are starting to emerge from the large numbers
(ferredoxin) and ferredoxin reductase (Figure 4.1).
of genome sequencing projects now underway^.
The Class II enzymes are the microsomal P450s,
which receive electrons from a single mem- In this chapter, we review the most recent
brane-bound enzyme, NADPH cytochrome P450 advances being made in understanding the func-
reductase (CPR), which contains FAD and FMN tion of P450 redox partners and the electron trans-
cofactors (Figure 4.1). Cytochrome b^ may also fer process. Special attention is paid to CPR,
couple with some members of the Class II P450s which occupies a particularly important position
family, notably CYP3A4, to enhance the rate of because of its central involvement in human drug
catalysis ^ metabolism.

Mark J.l. Paine and C. Roland Wolf Biomedical Research Centre, University of Dundee, Ninewells
Hospital and Medical School, Dundee, UK.
Nigel S. Scrutton Andrew W. Munro, Gordon C.K. Roberts and Aldo Gutierrez Department of
Biochemistry, University of Leicester, Leicester, UK.
Cytochrome P450: Structure, Mechanism, and Biochemistry, 3e, edited by Paul R. Ortiz de Montellano
Kluwer Academic / Plenum Publishers, New York, 2005.

115
116 Mark J.I. Paine et al.

Class I: Iron-sulfur partners

Adx/AdR Pdx/PdR
NADPH NADP NAD(P)H NAD(P)*

i FADp^\mQ)

Mitochondrial membrane

Class II: Diflavin reductase partners


CPR CPR fusion

NADPH NADP NADPH NADP


4 4

Novel Systems
Fe2S2 fusion
NAD*

NADH

Figure 4.1. Electron transfer partners of cytochrome P450. In Class I systems, electrons are shuttled from
NAD(P)H through an FAD-containing ferredoxin reductase and an iron-sulfur containing ferredoxin to P450; in
prokaryotes these are typified by putidoredoxin reductase (PdR) and putidoredoxin (Pdx), and in eukaryotes by
mitochondrial membrane associated adrenodoxin reductase (AdR) and adrenodoxin (Adx). Class II systems are driven
by electrons delivered from NADPH through diflavin (FMN- and FAD-containing) reductases. In eukaryotes these are
bound to the endoplasmic reticulum, while fused systems such as P450 BM3 exist in bacteria and fungi. Novel systems
now include P450RhF, which contains an FMN-containing reductase fused with a ferredoxin-like center and a P450.

2. NADPH-Cytochrome P450 apoptosis and cellular homeostasis^' ^, it is inter-


Reductase and the Diflavin esting that CPR was first isolated from yeast as an
Reductase Family FMN containing NADPH-dependent cytochrome
c reductase^. A mammalian equivalent was later
isolated from pig liver and reported to contain an
2.1. Background
FAD cofactor^. By 1962, the enzyme was shown
In view of the key role that cytochrome c to be localized at the endoplasmic reticulum^, and
has recently been found to play in regulating the flavin cofactors to be involved in the reduction
Electron Transfer Partners of Cytochrome P450 117

of cytochrome c^^. The true physiological redox A useful feature of flavins is that their absorption
partner was eventually discovered as P450 through spectra are altered by changes in their reduction
the reconstitution of laurate co-hydroxylase activ- state. Thus, the reduction state can be examined
ity from a detergent solubilized preparation by measuring changes in the visible absorbance
of cytochrome P450, NADPH cytochrome c range (Figure 4.2). This unique property stimu-
reductase^ ^' ^^, and a heat stable component, which lated research into the redox properties'^' '^, of the
was later identified as the phospholipid phos- enzyme and the complex processes of hydride/
phatidylcholine^^. electron transfer from NADPH, across the flavins
The initial difficulties in identifying a physio- and on to P450, discussed later in this chapter.
logical role for CPR were due to the purification The cDNA and corresponding primary amino
methods used, which incorporated trypsin or lipase acid sequences of several CPRs including rat'^,
treatment^' ^. These resulted in the cleavage of rabbit^^, and human^' were obtained by the
the amino-terminal membrane anchoring domain, mid-1980s, and the development of Escherichia
which constitutes the first 60 or so amino acid coli expression systems paved the way for detailed
residues and is responsible for interactions with the molecular characterization of the polypeptide
phospholipid bilayer and P450^'^. Thus, while through site-directed mutagenesis. The three-
proteolytically cleaved CPR is fully functional and dimensional structure of rat CPR was determined
capable of reducing c3^ochrome c and a range of by X-ray crystallography in 1997 by Kim and
artificial electron accepting compounds, it is unable coworkers^^, providing the structural prototype for
to reconstitute with P450s. The intact form of the dual flavin oxidoreductases.
reductase was eventually purified from liver micro-
somes using detergent solubilization procedures
2.2. The Diflavin Reductase
and found to have a molecular weight of 76-80 kDa
Family
and to support P450-dependent reactions^^' ^^.
In the early 1970s, the enzyme was shown to CPR is the prototype for a small family of
contain one molecule each of FMN and FAD^^' ^^. diflavin reductases which are believed to have

300 400 500 600 700

Wavelength (nm)
Figure 4.2. Absorbance spectra of oxidized and reduced human CPR. The absorption maxima for the oxidized
enzyme are located at 380 nm and 454 nm. The direction of the arrow shows the absorption changes that occur upon
reducton of the flavins, in this case using a 2-fold and 20-fold excess of NADPH.
118 Mark J.I. Paine et al.

ferredoxin reductase |

flavodoxin I FMN

CPR IMS

BM3 iPSgf^:

NOS[ ':^1^^''-
Figure 4.3. Schematic outline of the diflavin reductase family. Members contain an N-terminal FMN-binding
flavodoxin-like domain and a C-terminal FAD/NADPH-binding ferredoxin reductase-like domain, which contains an
additional linker region. Shown are: CPR, which has an amino-terminal membrane anchor region (Anc); NRl (Novel
reductase 1); MSR (methionine synthase reductase), which contains an additional interdomain sequence; P450 BM3,
which is fused to a P450 domain; and NOS (nitric oxide synthases), which has linked to a heme-containing
oxygenase domain that is structurally distinct from the P450s.

evolved as a result of a gene fusion event between electrons to the P450 monooxygenase complex,
ancestral FMN- and FAD-containing flavopro- albeit with greatly reduced efficiency^^. A num-
teins^^. Indeed, CPR is one of only four mam- ber of other diflavin reductases have been dis-
malian enzymes that contain both FMN- and sected into functional units including: rat CPR^^,
FAD-binding domains, the other three being BM3^'' ^^, NRl^^, and methionine synthase reduc-
methionine synthase reductase^"*, NRl^^, and the tase^"^. The ability to separate CPR and other
nitric oxide synthases^^ (Figure 4.3). Bacterial diflavin reductases into their component parts has
members of the family include the B. megaterium greatly facilitated structural and functional studies
cytochrome P450 BM3^^ and the reductase subunit of these enzymes.
of sulfite reductase^^. While these are all struc-
turally related, there exist some key differences.
For instance, methionine synthase reductase and
2.3. CPR Genes
NRl both contain the same domain organization as
P450 reductase but lack the membrane anchoring Apart from plants, which contain multiple
sequence, and are thus located in the cytosol. CPR genes^^'^^, most organisms contain a single
Methionine synthase also has a much larger inter- CPR gene. At the time of writing, a relatively
domain linker than CPR, although the functional modest total of 20 CPR genes have been identi-
significance of this is unclear. In nitric oxide syn- fied, which will doubtlessly increase as a result of
thase, the reductase region is fused with a heme the many genome sequencing projects currently
domain, and has acquired an extra calmodulin- underway. New sequences can be monitored using
binding domain to regulate electron transfer. the website www.icgeb.trieste.it, which provides
Experimental evidence for the gene fusion sequence updates on P450s and related enzymes.
concept comes from work of Smith et al?"^, who The rat gene was cloned in 1990^^ and comprises
dissected the FMN and FAD/NADH-binding 16 exons, with the first exon being non-coding.
domains of human CPR and expressed them as Amongst the coding exons there is a general
discrete functional units in E. colfl^. The individ- correspondence with the structural domains, with
ual domains not only folded correctly but also exons 2 and 3 encoding the membrane anchor
could be reconstituted to form an active complex region, exons 4-7 the FMN-binding domain, and
capable of reducing cytochrome c and donating exons 8-16 the FAD/NADPH-binding region.
Electron Transfer Partners of Cytochrome P450 119

A similar intronic arrangement is found for the 8-9 days and display a number of defects includ-
mouse gene located on chromosome 6^^' ^^. The ing neural tube, cardiac, eye, and limb abnormali-
human gene is located on chromosome 7^^ ties, and generalized defects in cell adhesion"^^.
Although the regulation of expression of Kasper's group has examined the effects of
individual P450s in response to exogenous and CPR gene disruption by removing the natural
endogenous factors has been studied extensively, translation initiation site and deleting the
the factors controlling the expression of the membrane-binding domain of CPR"^^. However,
CPR gene are less well understood. Reductase some homozygous null embryos (CPR~^~) were
expression may be induced by a number of found to produce a truncated 66 kDa c5^oplasmic
cytochrome P450 inducers such as phenobarbi- form of CPR, presumably through initiation at an
tal'^^ but the regulation of reductase gene expres- alternative start site. That these still produced the
sion is independent of that of cytochrome P450'^^. spectrum of embryonic defects leading to mid-
The upstream regulatory region of the rat gestational lethality provides strong evidence that
CPR gene has a number of interesting features"^^. the microsomal location of CPR is essential for the
Unlike many drug-metabolizing enzymes, the physiologically important functions of the P450s.
promoter does not contain either a TATA or The problem of the lethality of a CPR knock-
CCAAT box, it is GC rich, contains multiple out has been resolved by applying the regulatable
Spl consensus sequences, and utilizes two major Cre/loxP system^^ to generate mice in which
transcription start sites. CPR can be deleted post-natally in the liver^^. Such
The CPR gene is regulated by thyroid hor- "conditional knockout" mice, in which all the
mone, which stimulates CPR expression via the hepatic P450s are essentially inactivated, provide
thyroid response element (TRE) in the upstream the means of evaluating P450 function in normal
promoter'*^' ^^. Most recently, peroxisome prolifer-homeostasis, drug pharmacology, and chemical
ators, which regulate a battery of rodent P450 toxicity in vivo. Hepatic CPR-null mice exhibit
genes, have also been shown to regulate CPR many intriguing phenotypes^^. Due to an inability
levels"^^, possibly through a putative DNA bind- to produce bile acids, they no longer break down
ing site for PPARa that exists in the mouse cholesterol, and whereas hepatic lipid levels were
CPR promoter between - 5 6 8 and - 5 5 6 bp^^ significantly increased, circulating levels of cho-
Interestingly, following exposure to peroxisome lesterol and triglycerides are severely reduced.
proliferators CPR gene transcript levels appear There are also profound changes in the in vivo
to increase, while levels of CPR protein are metabolism of pentobarbital and acetaminophen,
decreased"^^. Thus, opposing transcriptional and demonstrating the predominant role of the hepatic
translational or post-translational mechanisms P450s in the pharmacology and toxicology of
appear to play a role in the regulation of CPR by these compounds, and illustrating the power of
peroxisome proliferators"^^. transgenic models in understanding P450 function.
One of the remarkable aspects of the hepatic
CPR deletion in mice was the fact that they live
2.4. Probing the Physiological and reproduce normally. This means that in adult
mice at least, the hepatic P450 system is not essen-
Role of CPR
tial for life, and indicates most strongly a funda-
The reductase gene has been knocked out in mental role in providing protection against toxic
mice^^' ^^; this leads to embryonic lethality, thus environmental agents. The data from these trans-
demonstrating that CPR is essential for normal genic models^^' ^^ also demonstrate that potential
development in higher organisms, presumably alternative electron transfer pathways for P450s,
because it plays a key role in the biosynthesis of such as the cytochrome b^/b^ reductase systems
signaling factors such as retinoic acid, sterols, that are discussed later, play only a minor role, if
prostaglandins, and steroids that are dependent any, in vivo.
on P450 catalysis. The precise role of CPR in The earliest CPR gene deletions were of
embryogenesis is still unclear and the effects of course carried out with yeast, and the data offer
gene deletion on the developing embryos are com- some interesting contrasts with mammalian
plex. Embryos lacking CPR do not survive beyond deletions"^^' '^^. The deletion of the CPR gene in
120 Mark J.I. Paine et al.

Saccharomyces cerevisae is not lethal but instead human heme oxygenase-1 (hHO-1) establish that
produces a viable mutant with an increased sensi- ionic interactions contribute to the binding of these
tivity (>200-fold) to ketoconazole, an azole proteins^^. Like P450, positively charged hHO-1
inhibitor of the ftingal P450 CYP51, a lanosterol surface residues contribute to the binding of this
14a-demethylase involved in the essential ergo- molecule to CPR, most likely via the negatively
sterol biosynthesis pathway"*^. Although this charged residues in the FMN domain of CPR.
hypersensitivity is suggestive of inefficient sterol
biosynthesis, it has been observed that the 2-5. Structure of CPR
cpr~ strains still accumulate significant amounts
of ergosterol, around 25% of those in the wild- The crystal structure of rat CPR lacking the
type parent"^^. This indicates that, unlike mice, an N-terminal membrane-binding sequence^^ shows
alternative electron donor is effective in delivering that the catalytic region comprises three distinct
two electrons for P450 activity^^. In contrast to domains: an FMN-binding domain structurally
Shen's observations with cpr-deleted mice"^^, a similar to flavodoxins; an NADPH/FAD-binding
cytosolic truncated version of native CPR with the domain similar to ferredoxin-NADP^ reductase;
N-terminal membrane anchor removed effectively and a "linker" domain, present as an insert in the
reversed the phenotypes associated with cpr sequence of the NADPH/FAD-binding domain
deletion in yeast^^. Thus, there appear to be (Figure 4.4). The "linker" sequence is the major
fundamental differences in the mechanisms of P450 structural feature that distinguishes these mole-
interactions between yeast and mammalian CPRs. cules from the single-domain ferredoxin reduc-
While the major role of CPR is associated with tases, and is likely to play a structural role in
cytochrome P450 and the phase I metabolism of positioning the FMN- and FAD-binding domains
xenobiotic compounds, the enzyme also has the correctly for direct electron transfer. This is sup-
ability to reduce other electron acceptors such as ported by mutagenesis studies in which compari-
cytochrome c^, cytochrome b^ (ref [50]), and heme son of wild-type and mutant structures shows
oxygenase^ ^ In addition, it has recently been impli- significant differences in the relative position of
cated, along with cytochrome Z?^, in the bioreduc- the two flavin domains of rat CPR^^. Furthermore,
tive activation of methionine synthase^^, thus there is increasing evidence from structural stud-
pointing to a possible role in the regulation of ies with other dual flavin reductases that the inter-
methionine synthesis. CPR has also been shown to play between NADPH, flavin cofactors, and heme
be involved in the regulation of oxidative response is directed by conformational changes. This is
genes such as hypoxia-inducible factor 1 (ref [53]). exemplified by nNOS, where structural relaxation
P450 reductase plays a role in the bioactivation appears to be an important part of the calmodulin-
of therapeutic prodrugs through its ability to dependent activation mechanism^ *.
reduce a range of one-electron acceptors such
as the quinone drugs doxorubicin^^ and mito-
mycin c^^ and aromatic TV-oxides such as the novel 2.5.1. The FMN-Binding Domain
benzotriazine, tirapazamine^^. Evidence is also
emerging that other members of the CPR family The FMN-binding domain interacts with
including nitric oxide synthases^^ and NRl^^ may cytochrome P450 to transfer electrons during
play a similarly important role in the metabolism catalysis. In addition to rat CPR, detailed struc-
of chemotherapeutic drugs. tural studies of the isolated human FMN domain
Overall, the biological relationships of CPR have been carried out using NMR^^, and X-ray
with proteins other than P450 are not especially crystallography^^. The isoalloxazine ring of FMN
well established. However, attention is starting is sandwiched between two aromatic side chains,
to shift toward understanding the role of CPR in Tyrl40 and TyrlTS^^' 62,63 jyj.173 ij^s parallel to
other redox pathways. In particular, data is accu- the 5/-side of the FMN ring, while the second
mulating on CPR interactions with cytochrome b^ aromatic, Tyrl40, located on the re-side, lies at
(ref [58]) and heme oxygenase^^. Consistent with an angle of ~40° to the isoalloxazine ring
the findings for cytochrome P450 enzymes, (Figure 4.5). A similar arrangement is found in
molecular investigations on the binding of CPR to the FMN-binding domain of P450 BM3 and in
Electron Transfer Partners of Cytochrome P450 121

most flavodoxins, apart from Clostridium flavo- isoalloxazine ring on the ^/-side at the pyrimidine
doxin where the r^-side aromatic side chain end, although not in contact with it (Figure 4.5).
residue is replaced by methionine^"^. Removal of This residue is highly conserved in P450 reductase
the aromatic side chains of Tyrl40 and/or Tyr 178 and related enzymes, indicating a possible role in
from rat liver CPR by site-directed mutagenesis FMN binding. Mutations that remove the aromatic
results in decreased affinity for FMN^^. side chain result in a 50-fold reduction in affinity
In addition to these two key residues, a third for FMN, and NMR spectra show significant
aromatic side chain, PhelSl, lies close to the FMN changes in amide chemical shifts of residues in

_ 'linker'

Figure 4.4. Schematic representation of rat CPR domains (protein data base [pdb] code lAMO). In the crystal,
the two flavins, FMN and FAD, are 4 A apart.

FMN Y178

Y140
F181

Figure 4.5. Close up of the FMN isoalloxazine ring showing the aromatic residues, Tyrl40 (Y140), Tyrl78
(Y178), and PhelSl (F181) associated with FMN binding in human CPR (pdb code IBIC).
122 Mark J.I. Paine et aL

the p4-a6 loop. This loop includes PhelSl, the chain of the tryptophan stacks against the isoallox-
key FMN-binding residue Tyrl78, and three azine ring of FAD, and structural rearrangements
other residues that form hydrogen bonds to the are thought to be essential to enable access of the
isoalloxazine ring, as well as residues in the adja- nicotinamide ring to the isoalloxazine ring of FAD
cent p5-a7 loop^^. These are consistent with local for hydride transfer (Figure 4.6). The proposed
changes in structure on substitution of PhelSl, mechanism for binding involves the tryptophan
and with the idea that the packing of this con- residue flipping away from FAD to be replaced by
served residue plays a significant role maintaining the nicotinamide region of NADPtf^.
the orientation of residues which interact with the There has been extensive site-directed mutage-
isoalloxazine ring and hence in the formation of nesis of amino acid residues in the vicinity of the
the FMN-binding site. NADPH-binding region to examine their role in
cofactor binding and hydride transfer (Figure 4.7).
Removal of the aromatic side chain from Trp676
2.5.2. FAD/NADPH-Binding Domain shows that it plays an essential role in the
Like FNR, FAD is bound in an extended con- discrimination between NADPH and NADH as
formation in rat CPR^^. Key residues that serve to cofactors for CPR^^' ^^, notwithstanding the fact
regulate flavin binding and reactivity are Arg454, that it does not interact with the 2'-phosphate
Tyr456, Cys472, Gly488, Thr491, and Trp67722. which is the sole difference between the two
Site-directed mutagenesis of these residues has cofactors. Investigation of the steady-state kinet-
been carried out to examine their role in FAD ics shows that substitution of Trp676 with Ala
binding and catalysis^^. Trp677 is stacked against decreases the K^ for NADH from 48 to 0.3 mM,
the re-face of FAD, while Tyr456 lies at its 5/-face. and increases the k^^^ to ~4,000 min~^ while the
Deleting Trp677 reduces catalytic activity 50-fold ^^NADPH ^Qps tQ 0.2 |ULM and k^^^ to lOmin^^
but has little effect on FAD content. However, The Trp676Ala mutant therefore binds both
mutations to Tyr456, which also hydrogen bonds coenzyme molecules more tightly, the change in
to the ribityl 4'-hydroxyl, decreases affinity for cofactor specificity (expressed as a k^JK^ ratio)
FAD by around 8,000-fold. Side chains from being predominantly a reflection of changes in
Thr491 and Arg454 stabilize the pyrophosphate k^^^. A similar change in cofactor specificity also
group of FAD. Substitution of Thr491 has occurs with pea FNR with the analogous mutation
a relatively modest effect on FAD binding (~ 100- Tyr308Ser^4' ^^ Both in pea FNR^^ and in CPR^^
fold decrease) compared with mutation of Arg454, substitution of the C-terminal aromatic residue
which decreases affinity 25,000-fold. allows the nicotinamide ring of the cofactor to
NMR studies show that the pro-R-hydrogen bind close to the isoalloxazine ring, the thermody-
attached to the C-4 atom of NADPH is transferred namically unfavorable flipping no longer being
directly to FAD as a hydride ion^^. However, an required. Hence, removal of this physical rate-lim-
understanding of NADPH binding and the factors iting step produces enzymes that have lower
involved in hydride transfer have been hampered by apparent K^ values for nicotinamide cofactors.
the spatial disorder in this region of the molecule Further kinetic evidence for a critical role for this
in the crystal structure. While the electron density Trp residue in regulating NADPH binding and
of the adenosine moiety of the coenzyme is well hydride transfer is detailed in Section 2.6.1.
defined in the structure, that from the nicotinamide Several conserved amino acids including ser-
ring is not, and is consistent with at least two dif- ine 596, arginine 597, and lysine 602 have been
ferent positions for this ring, neither of which is in proposed to be involved in binding the adenosine-
contact with the isoalloxazine ring of FAD^^. ribose moiety of NADPH (Figure 4.7), and
Similar observations have been made in enzymes specific charge-charge interactions between these
of the FNR family which are structurally similar to and the 2'-phosphate of NADPH are thought
the FAD/NADPH-binding domain of CPR^^-^i. An to be responsible for discrimination against
aromatic amino acid residue is located at the car- NADtf^' ^^-^l Working on the hypothesis that
boxyl-terminus in all members of the FNR family mutations to these residues might reduce the
In rat and human CPR, this position is occupied by affinity of the enzyme for NADP(H) and thus pre-
Trp677 and Trp676, respectively. The planar side vent inhibition by NADP^, Elmore and Porter^^
Electron Transfer Partners of Cytochrome P450 123

298

Figure 4.6. Arrangement of FMN, FAD, and NADP+ in the rat CPR crystal structure (pdb code lAMO). The
Trp677 aromatic ring has to move away from the FAD isoalloxazine ring to allow access of nicotinamide ring to FAD
for hydride transfer.

-oJ.,,-^J Nicotinamide

Adenosine-ribose

y
RS^ R597
S596
R634

Figure 4.7. Schematic diagram of residues associated with NADPH binding in human CPR (taken from Dohr^^).

have coupled mutations to residues in the 2'-phos- W677A, while their catalytic efficiency with
phate-binding site with the W677 substitution. NADH was increased at least 500-fold.
The mutant combinations R597MAV677A and Three residues Ser457, Asp675, and Cys630,
R597M/K602WAV677 A showed a significant 4- lie in close proximity to the FAD isoalloxazine
fold reduction in inhibition by NADP^ relative to ring in CPR, and have been closely examined
124 Mark J.I. Paine et al.

in relation to hydride transfer. Non-conservative these enzymes have distinct spectral characteris-
substitutions of Ser457, Asp675, and Cys630 pro- tics. However, the fact that the oxidised, semi-
duce large decreases in cytochrome c reductase quinone and reduced states of both FAD and FMN
activity^^' ^^' ^^' ^^. These parallel changes in the have virtually indistinguishable spectra means that
rate of flavin reduction are associated with large kinetic and equilibrium studies of the diflavin
NADPD isotope effects^ ^ thus pointing to reductases is complicated. This is alleviated, how-
impaired hydride transfer from NADPH. Based on ever, by using the genetic approaches discussed
spectral analysis of flavin reduction of mutants previously to dissect the enzymes into their com-
at residues Ser457, Asp675, and Cys630 and ponent domains^^' ^^' ^^^ ^^. This strategy has been
analysis of the kinetics and pH dependence of used successfiilly with human CPR, human
cytochrome c reduction, it was proposed that methionine synthase reductase, and human oxi-
Cys630 acts a proton donor/acceptor to FAD, and doreductase NRl. Molecular dissection of these
Ser457 and Asp675 interact to stabilize both the diflavin enzymes removes ambiguity in spectral
transition state and the FAD semiquinone^^. This assignment during redox titration, potentiometry,
is supported by crystallographic analysis of and kinetic analysis^^' ^'^' ^^-^^. With CPR, the do-
CPR mutants, which shows that both Ser457 and mains have also formed a basis for more detailed
Asp675 are directly involved in interaction with study of the kinetic and thermodynamic proper-
the nicotinamide group of NADP(H) and likely to ties of the frill-length enzymes^^' ^^' ^^. The use of
orient the C-4 atom of NADP(H) into an optimal FMN-depleted frill-length enzyme has also facili-
position for hydride transfer^^. The catalytic triad tated assignment of reduction potentials and indi-
of Ser457, Asp675, and Cys630 is highly con- vidual kinetic phases in other mammalian CPRs^^.
served in all diflavin oxidoreductases apart from Each isolated flavin-binding domain of human
NRl, which has a nonconservative Ala549 corre- CPR (and of related family members) is soluble,
sponding to Cys630^^ and a greatly decreased binds flavin, and is redox active^^' ^^. The mid-
(~ 100-fold) rate of reduction relative to CPR. point reduction potentials of the flavin couples are
similar in the isolated domains and ftill-length
CPR^^, indicating that the isolated domains are
2.6. The Electron Transfer
good mimics of domain properties in ftiU-length
Mechanism CPR. The blue semiquinone on the FMN is both
CPR catalyzes the transfer of electrons along kinetically and thermodynamically stabilized, and
the pathway NADPH-> FAD-> FMN-> P450, the reduction potential of the oxidized-semi-
and there are two striking features of its involve- quinone redox couple is much more positive than
ment in the monooxygenase system. First, the those of the other redox couples in CPR (Figure
flavin redox potentials are such that electron trans- 4.8). This accounts for the "blue/green" appear-
fer from NADPH to FAD is moderately unfavor- ance of purified CPR prior to chemical treatment
able (the physiological -direction of electron with exogenous oxidants. The midpoint reduction
transfer in the structurally related ferredoxin potentials measured for human CPR^^ are in broad
reductase is in the direction of NADPH forma- agreement with earlier studies performed with
tion). Second, NADPH is an obligate two-electron rabbit CPR^^. The reductive half-reaction of the
donor, but these two electrons must be delivered to isolated FAD domain has been studied by
P450s individually at the appropriate points in the stopped-flow methods^^. Hydride transfer is
catalytic cycle. CPR orchestrates the electron sup- reversible, and reduction of NADP^ by FADH2 is
ply from NADPH to the P450 cytochromes by sta- more rapid (8 s ') than FAD reduction by
bilizing the one-electron reduced form of the flavin NADPH (3 s~^), consistent with the relative val-
cofactors FAD and FMN. This stabilization gives ues of the midpoint reduction potentials for the
rise to the blue semiquinone species (FADH7 2-electron couples for FAD/FADH2 and NADPH/
FMNH*), which is observed in both kinetic and NADP^. The more rapid reduction of NADP^
equilibrium studies of CPR^^' ^^-84 cPR and the probably reflects the physiological role of the
aforementioned related structural isoforms of ancestral FNR-like protein, which evolved to
nitric oxide synthase^^, methionine synthase reduc- catalyze NADP^ reduction in photosynthetic
tase^"^, and protein NRl^^ can accept a maximum electron transfer. The isolated FAD domain trans-
of four electrons. The different redox states of fers electrons to the isolated FMN domain, but the
Electron Transfer Partners of Cytochrome P450 125

n^m^li jMf^'^s&^ki^-
-100 i

mm mm

-200' mmmtm
FMN sq/hq

FMN sq/hq iFlffNIl^tiNi


fMUQ^'Di^liil
FMN sq/hq
FMN sq/hq
'W0^mm fmmm
-300 i FMN sq/hq FAD sq/hq FAD sq/hq •PM^mm
FMIi^i^
NAE^PP
FAD sq/hq FAD sq/hq
FAD sq/hq
-400 i

CPR NR1 MSR NOS BM3


Figure 4.8. Midpoint reduction potentials of the various flavin couples in CPR and related diflavin enzymes.
Values are taken from Munro^^ (CPR), Finn^^ (NRl), Wolthers^^ (MSR), Noble^i (NOS), and Daff«9 (BM3).

observed rate is slow and shows a second-order Mycobacterium tuberculosis^^. A number of the
dependence (second-order rate constant 9.5 X 10"^ early kinetic phases are obscured in studies with
M"^ s~^) on domain concentration, reflecting full-length CPR^^, thus emphasizing the impor-
a bimolecular reaction. This indicates the impor- tance of conducting detailed studies of electron
tance of covalently tethering the two flavin- transfer mechanism in single domains of CPR.
binding domains of CPR to enable rapid The relatively high midpoint reduction poten-
interdomain electron transfer. tial of the FMN oxidized-semiquinone couple
Reduction of the isolated FAD domain by (—66mV) provides the driving force for inter-
NADPH occurs in a series of discrete kinetic domain electron transfer in full-length CPR^^.
steps including at least two different charge- This enables electron flow from NADPH to the
transfer complexes involving NADPH and FAD CPR FMN. Electron transfer then occurs from the
(Figure 4.9). Partial flavin reduction occurs in the FMN to the cytochrome P450 enzymes (or other
initial phase (—200 s~^) and this displays a kinetic redox acceptors) and is a key aspect of the elec-
isotope effect with ^-side NADPD. FAD is then tron transfer mechanism in virtually all diflavin
further reduced in the slower phase (~20s~^). reductase enzymes^^' ^^' ^^. Kinetic studies show
NADP+ release is thought to gate complete reduc- that the transient accumulation of the blue dis-
tion of the flavin in the slower phase, consistent emiquinoid species of CPR occurs at a rate identi-
with a reversible scheme for flavin reduction in cal to hydride transfer from NADPH to FAD^^.
the isolated FAD domain and full-length CPR This indicates that interdomain electron transfer is
(Figure 4.9). Similarly, it is proposed that the rate relatively fast. The blue disemiquinoid species
of flavin reduction is regulated by NADP^ release subsequently decays following a second hydride
in rat neuronal nitric oxide synthase^ ^' ^^ and the transfer from NADPH (Figure 4.9), as the flavins
adrenodoxin reductase homolog FprA from are reduced to their hydroquinone states.
126 Mark J.I. Paine et al.

k k k k
E + NADPH ., "* ^ E-NADPHCT1 ^ ^ N E-NADPHCT2 ^ ^ \ EHgNADP^ ^ ^ EH2 + NADP*
'^-l '^-2 ^^-3 '^-l
(I)
k^\k.
k
4'^
k
.i«
E-NADPHCT2 ^ i ^ EH2NADP . ^ i EH2 + NADP^
(NADPH) ^-^ (NADPH) ^-8 (NADPH)

FAD FAD FADHo FADH FAD


E + NADPH ' E NADPH. E NADPH** E NADP* E NADP*
FMN FMN FMN FMN FMNH2

i
FAD
-50% -50%

I -NADP*
E NADPH .^ [Refer to Scheme I]
FMN (NADPH) +NADPH

FADH2
FAD
E NADPH
' FMNHo
FMNHo

[Refer to Scheme I]., E


1
FAD
NADPH
FMNH (NADPH) (II)

Figure 4.9. Kinetic schemes for the isolated FAD domain (upper scheme) and full-length CPR (lower scheme)
derived from stopped-flow studies (taken from Gutierrez^^). The reversible nature of the reaction is indicated,
as is the presence of the second nicotinamide coenzyme-binding site (indicated in parenthesis for the FAD domain).
For clarity, only enzyme intermediates bound with a single coenzyme are shown in the lower scheme for CPR,
but reference to the upper scheme indicates that a second molecule of NADPH has the capacity to bind to the
enzyme.

2.6.1. Trp676 and FAD Reduction Trp676 by alanine substantially compromises the
rate of hydride transfer in CPR, revealing a key
As discussed above, the C-terminal aromatic role for the side chain of Trp676 in enzyme reduc-
residue (Trp676 in human CPR) is positioned over tion. In Trp676His CPR the rate of FAD reduction
the re-face of the FAD isoalloxazine ring in such a is modestly affected, but importantly the enzyme
way that it would sterically prevent hydride trans- is reduced only to the two-electron level in rapid-
fer from NADPH to FAD in the absence of side mixing experiments. Reduction beyond the two-
chain movement (Figure 4.6). In human CPR, electron level is prevented owing to the slow
rapid changes in tryptophan fluorescence emis- release of NADP^, indicating a role for Trp676 in
sion accompany hydride transfer in the isolated the release of oxidized coenzyme from the FAD-
FAD-binding domain and in CPR^^. The complex binding domain. A double-mixing stopped-flow
fluorescence transients observed in CPR as two method in which Trp676His CPR is reduced ini-
hydride equivalents that are transferred to the tially with stoichiometric NADPH, and following
enzyme are largely absent in a W676H mutant a suitable delay (100 ms) mixed with excess
CPR, suggesting that Trp676 is the origin of the NADPH, effects reduction to the four-electron
fluorescence signal in wild-type enzyme^^ and leveF^. In the delay period, NADP^ can escape,
that the observed transients may reflect the postu- albeit at a relatively slow rate, allowing binding of
lated movements of this side chain. Substitution of the second NADPH to the catalytic site.
Electron Transfer Partners of Cytochrome P450 127

2.6.2. Binding of Two Coenzyme data indicate that the coenzyme plays a role in the
IVIolecules control of CPR catalytic activity as well as acting
as a substrate. Models that assume a single
Rapid-mixing studies of the kinetics of coenzyme-binding site for steady-state turnover
hydride transfer in both the isolated FAD-binding to explain competitive inhibition by coenz>ine
domain and in CPR have led to a kinetic model fragments might be inappropriate. The effect of
(Figure 4.9) that invokes the existence of two the second coenzyme-binding site on steady-state
kinetically distinct binding sites for NADPH^^. turn-over and inhibition might lead to revised, and
Only one site is catalytic; binding to the second by necessity more complex, steady-state kinetic
site attenuates FAD reduction, probably by inter- models.
fering with NADP^ release from the catalytic site.
In wild-type CPR, NADP+ release from the cat-
alytic site is sufficiently rapid that binding to the
2.6.3. Internal Electron Transfer
second noncatalytic site does not prevent reduc-
tion to the four-electron level in direct-mixing Although a thermodynamic and kinetic frame-
stopped-flow experiments with excess NADPH. work for electron transfer in CPR has emerged
Following the identification of two kinetically from rapid-mixing kinetic and potentiometry
distinct coenzyme binding sites in CPR and the studies^^' ^^, alternative approaches have been
unusual mechanism for electron transfer to FAD, required to access the rate of internal electron
similar dual binding-site models have been pro- transfer between the flavins. Tollin and coworkers
posed for nitric oxide synthase^^ and the adreno- have used flash photolysis studies employing pho-
doxin reductase homolog FprA isolated from M toexcited 5-deazariboflavin to investigate the
tuberculosis^^. kinetics of inter-flavin electron transfer in rabbit
The presence of two coenzyme-binding sites is CPR^^. CPR is reduced rapidly by photoexcited
unexpected since they cannot be inferred solely 5-deazariboflavin (6.8 X lO^M-^s"^) and this
from the crystal structure of CPR^^. Kinetic stud- phase is followed by a slower (70 s~^) partial loss
ies with wild type and W676H CPR at different of blue semiquinone signal that reports on inter-
concentrations of NADPH have, however, pro- flavin electron transfer from FAD to FMN^^.
vided further support for the existence of two Pre-reduction of rabbit CPR prior to flash photol-
sites^^' ^^. The rate of flavin reduction in the iso- ysis yields a rate constant for internal electron
lated FAD domain and CPR increases as NADPH transfer of 15 s~ ^. This slower rate for pre-reduced
is decreased from molar excess to stoichiometric enzyme (FAD^^/FMN ) is consistent with the
concentrations. At stoichiometric concentration, smaller driving force for electron transfer from
the second noncatalytic site is predominantly FAD^ to FMN following electron donation by
vacant and the partial inhibition on the rate of 5-deazariboflavin. Studies with human CPR have
flavin reduction from the catalytic site is therefore used temperature-jump relaxation kinetic methods
relieved (Figure 4.9). Occupation of the noncat- to follow inter-flavin electron transfer^^' ^^. As
alytic site occurs at NADPH concentrations in with flash photolysis, relaxation kinetic methods
excess of the enzyme concentration, and impairs bypass preceding steps that might limit the rate of
NADP^ release from the catalytic site. This in inter-flavin electron transfer in rapid-mixing
turn partially inhibits flavin reduction, the rate of approaches (e.g., the preceding FAD reduction
which is gated by NADP^ release. Preincubation step). Application of relaxation kinetic methods to
of the enzyme with a stoichiometric amount of CPR has allowed direct measurement of the rate
adenosine 2',5'-diphosphate does not lead to of inter-flavin electron transfer in enzyme reduced
inhibition of the flavin reduction rate^^. We infer at the two- and three-electron level, and also
that the binding of adenosine 2',5'-diphosphate opened up studies of conformational gating and
prevents NADPH from binding to the noncatalytic ligand effects on this reaction^^. A key require-
site. This observation also suggests that it is the ment of the temperature-jump approach is to poise
nicotinamide-ribose-phosphate portion of NADPH the equilibrium of a reaction, such that perturba-
bound at the second site that hinders NADP^ tion of the equilibrium by rapid heating, effects
release from the catalytic site. Clearly, these new an absorption change that can be assigned to an
128 Mark J.I. Paine ef a/.

individual reaction step (i.e., internal electron the kinetics (1/T = 20 ± 0.2 s"^) of electron
transfer from FAD to FMN in the case of CPR). transfer in the nonphysiological direction
This is especially difficult with CPR where slow (FAD,^/FMN,^ ^ FAD^^/FMN^^).
thermodynamic relaxation would lead to an equi- In the W676H mutant CPR, this rate is substan-
librium mixture of several different redox forms tially increased (1/T = 263 ± 3 s~^), but the rate
of the enzyme. The experimental conditions were of electron transfer in the physiological direction
selected carefiilly to minimize any uncertainty (FAD^qFMN^q ^ FAD^^FMNj^q) is decreased by
arising from the identity of the enzyme species in only a factor of ~ 2 (1/T = 27 ± 1 s~^). This there-
the final equilibrium distribution. Potentiometry fore points to another important role for Trp676 in
studies indicate that in two-electron-reduced controlling electron transfer in CPR in favoring the
enzyme the electrons are distributed as an equilib- transfer of electrons in the physiological direction
rium between the two enzyme species FAD^^/ (NADPH -^ FAD -^ FMN -^ heme).
FMN^^ and FAD^/FMN. , and that - 5 0 % of each Although the initial equilibrium is the same,
sq ox nq'
inter-flavin electron transfer rates for CPR
enzyme species is populated^^. A rapid tempera-
reduced by NADH (1/T = 18 ± 0.7 s"') are - 3 -
ture increase shifts this equilibrium toward the
fold less than those for CPR reduced by NADPH
FAD^^/FMNH2 form, and the kinetics of this
(1/T = 55 ± 0.5 s~^). This cannot be attributed to
process report on the rate of inter-flavin electron
thermodynamic differences since the reduction
transfer^^. Inter-flavin electron transfer in human
potentials of the reducing coenzymes are essen-
CPR is relatively slow. For CPR reduced at the
tially identical. This suggests a role for coenzyme
two-electron level with NADPH, two kinetic
binding in modulating inter-flavin electron trans-
phases are seen in temperature-jump experiments.
fgj.95, 97 jjjjg jg supported by the observation
Fluorescence and absorbance studies with
that the rate of inter-flavin electron transfer in
dithionite-reduced CPR indicate that the fast
dithionite-reduced CPR (1/T = 11 ± 0.5 s"^ is
phase (1/T = 2,200 ± 300 s~^) is not associated
clearly less than that for CPR reduced with
with electron transfer, but is attributed to local
NADPH (1/T = 55 ±0.5s-^)^^ Furthermore,
conformational change in the vicinity of the FAD
addition of 2',5'-ADP to dithionite-reduced
when CPR is reduced with NADPH. The slow
enzyme effects a ~ 3-fold increase in the rate of
phase (1/T = 5 5 ± 2 S ~ ^ ) reports an internal elec-
inter-flavin electron transfer (1/T = 35 ± 0.2 s~^),
tron transfer, FAD^^-FMN^^ ^ FAD^^-FMN^^q.
and this is accompanied by an increase in the
An intrinsic electron transfer rate of —10^^ s"^ is
amplitude of the absorption signal change^^. Thus,
predicted based on the separation distance of the
the binding of the adenosine moiety of NADPH,
two flavins in rat CPR^^' ^^. The modest transfer
and in particular the 2'-phosphate group, is a
rates observed in the temperature-jump studies
major factor in enhancing the rate of inter-flavin
indicate that electron transfer is gated. Possible
electron transfer. Potentiometry measurements
origins of the gating reaction have been consid-
have shown that the binding of 2',5'-ADP does not
ered. For example, the reaction involves the
perturb the midpoint reduction potentials of the
deprotonation of the FAD blue semiquinone, but
four flavin couples. We propose that ligand binding
solvent isotope effects do not accompany the reac-
in the coenzyme-binding pocket effects a confor-
tion, ruling out a rate-limiting deprotonation reac-
mational change involving domain movement, the
tion. Electron transfer is compromised in the
rate of which controls inter-flavin electron transfer.
presence of glycerol (75% w/v glycerol), suggest-
ing conformational gating^^ and a conformational
search for domain orientations that maximize
electronic coupling between the flavins is consis- 2.6.4. Interaction with and Electron
tent with the atomic structure of CPR (and related Transfer to P450
diflavin enzymes), which are consistent with a
high degree of interdomain mobility^^' ^^' ^^ The The microsomal P450 monooxygenase
equilibrium distribution of enzyme species is complex is localized to the endoplasmic reticulum
poised differently by reduction of CPR at the membrane. Like P450, CPR contains an N-
three-electron level^^. Relaxation experiments terminal hydrophobic region that spans the lipid
with three-electron reduced CPR give access to membrane and anchors it to the surface. In yeast,
Electron Transfer Partners of Cytochrome P450 129

the soluble form of the enzyme can support P450 opposite ends, leading to a strong electric dipole
activity"^^, but soluble mammalian CPRs that have moment (677 Debye)^^, which may influence the
had the hydrophobic anchoring peptide removed orientation of the reductase at the membrane sur-
are incapable of coupling with cytochrome P450. face. The patch of positively charged residues
The anchoring of these enzymes to the membrane exposed at the surface of the human FMN-binding
surface thus appears to be the principal factor domain (K72, K74, K75, R78, R97, KlOO, H103,
required for correct spatial orientation of the and R108) in particular could form an additional
redox centers for effective electron transfer. membrane-binding site.
Studying the architecture and topology of Protein-protein interactions are essential to
membrane proteins on the phospholipid bilayer is enable electron transfer from the reduced FMN
experimentally extremely challenging. However, of the flavoprotein to the substrate-bound ferric
new developments are being applied to analyze form of the P450. Since P450 is present in a
the topography of the monooxygenase complex on 10-25-fold molar excess over CPR in the liver
a phospholipid bilayer. Sligar and coworkers have microsome^^'^' ^^^, rapid association and dissocia-
used 10-nm scale phospholipid bilayer disk struc- tion of P450: CPR complexes is important for the
tures to orient CPR and cytochrome P450 2B4 on system to work effectively. A number of lines of
a surface for visualization by AFM^^' ^^^. The evidence point to electrostatic interactions being
height of CYP2B4 protruding above the surface is the driving force behind the binding of P450 with
estimated at 3.5 nm, which is consistent with a CPR. For instance, it has been demonstrated that
hydrophobic tip of the molecule being partially specific positively charged lysine and arginine
inserted into the membrane. The exact spatial rela- residues on the rat P450, CYPlAl, are involved
tionship with CPR is not yet clear, but it is thought in forming an electron transfer complex with
that CPR lies in an orientation such that both the CPR^^6. It has also been reported that CYP2B1
FMN and FAD/NADPH domains lie close to the and CYP2B4 interact with CPR through comple-
membrane surface, which would allow close mentary charge interactions^^^' ^^^. In the case of
communication between the FMN and the P450 CYP2B4, site-directed mutagenesis has identified
heme22' ioo_ a series of lysine and arginine residues on the
Transient monooxygenase complexes are proximal surface near the heme ligand that inter-
formed on the membrane surface as a result of act with CPR^^^. Neutralization of carboxylate
collisions between P450s and CPR as each dif- groups on CPR by chemical modification inhibits
fuses laterally within the membrane of the endo- both cytochrome c reductase activity and P450-
plasmic reticulum. The role of the membrane in dependent monooxygenation"^^' ^^^. Chemical cross-
mediating these interactions is poorly understood. linking studies indicated that a cluster of acidic
However, early work has shown that the phospho- amino acids on CPR were involved in the interac-
lipid component of the membrane can affect tion with rat CYPlAl i^^.
intermolecular interactions of the monooxygenase The FMN domain may be assumed to provide a
complex^^^' ^^^ and influence substrate binding^^' ^^^ major portion of the docking surface for P450s,
and may therefore be important in maintaining although as discussed above some reorientation
efficient electron transfer from CPR to P450. of this domain from its position in the crystal struc-
The structure and sequence of rat and human ture may be required. Electrostatic potential meas-
CPR^^' ^^ show some interesting features that urement of the surface of the human CPR-
might influence the orientation of the protein at FMN-binding domain shows three distinct clusters
the membrane surface. One is a cluster of basic of acidic residues that could form ion-pair inter-
residues R'^^KKK, which lies at the membrane actions with the electron transfer partners^^' ^^^
anchor: FMN domain junction. Their functional Cluster 1 contains Asp207,Asp208,Asp209; cluster
significance is unclear, but it can be speculated 2, Glu213, Glu214, Asp215; and cluster 3, Glul42,
that the basic Arg and Lys side groups might Asp 144, Asp 147. The first two clusters correspond
interact with anionic phospholipid head groups, to the region of CPR that was cross-linked to a
possibly to restrict the movement of CPR on the lysine residue in cytochrome c^^^, and have been
membrane surface. The FMN-binding domain investigated by site-directed mutagenesis in rat^^^
has clusters of positive and negative charges at and human^^ CPR. The results of these experiments
130 Mark J.I. Paine ef a/.

show that while cluster 1 mutations do not afifect electron transfer reactions, this is not always
C3^ochrome c binding, removing the charge from reflected in the ionic-strength dependence of
Asp208 alters the binding of cytochrome P450. In P450 reduction in vitro with reconstituted mono-
contrast, cluster 2 mutations affected the binding of oxygenase systems. For instance, it has been
cytochrome c, but not P450. Mutations of residues shown that in the presence of high concentrations
in cluster 3 produce a modest decrease in P450 of salt, which should neutralize electrostatic inter-
activity with no effect on cytochrome c reductase actions, the reduction rate of P450 by CPR may
activity^^. Thus, discrimination is apparent between increase^ ^'^~^^^. It is possible that the observed
residues involved with the interaction of CPR with effects of ionic strength reflect the combined
cytochrome c and with P450. result of perturbations of CPR-P450 interactions
The finding that mutation of residues in clus- and of domain-domain interactions within CPR.
ters 1 and 3 on either side of the FMN-binding site Most recently Davydov et al}^'' have compared
affect P450 activity, suggests that c3^ochromes the role of electrostatic interactions in the associa-
P450 bind at the tip of the FMN domain in such a tion of rabbit CPR with CYP2B4 and the heme
way as to cover the FMN cofactor^^. Examination domain of P450 BM3, which is normally fused
of the crystal structure of rat CPR indicates that with a reductase domain' '^. They found an increase
structural rearrangements during catalysis of the in K^ (10-90 nM) for the CPR-CYP2B4 complex
kind discussed above would be required to bring with increased ionic strength (50-500 JJLM), con-
the redox centers of CPR and P450 into an sistent with the involvement of charge pairing.
appropriate configuration for efficient electron Interestingly a reverse relationship was observed
transfer^^ (Figure 4.10). A similar structural with BM3-heme and BM3-CPR domains, which
rearrangement has also been suggested for the showed low affinity for each other. It is postulated
FMN and FAD domains of BM3' ^l that this may reflect the fact that P450 BM3 is a
While it is generally considered that charge- tethered complex, where there is no strong require-
pair interactions are important in docking and ment for charge pairing.

"Open" conformation:
appropriate for electron
transfer to P450

Conformation in crystal
appropriate for inter-
flavin electron transfer

Figure 4.10. Schematic model for the domain movement required for opening CPR for efficient electron transfer
to P450.
Electron Transfer Partners of Cytochrome P450 131

2.7. Cytochrome P450 BM3 The overproduction of P450 BM3 facilitated by


addition of phenobarbital to B. megaterium
Since its discovery in the 1980s, flavocy- growth medium simplified purification of the
tochrome P450 BM3 (CYP102A1) has undergone enzyme, and cloning of the gene allowed its
intensive biochemical study and has established expression and purification from E. coli^^^. It
itself as a model P450 system to rival P450cam. quickly became apparent from analysis of both
The key aspect of the structure of P450 BM3 that gene and protein that P450 BM3 was an unusual
has led to this rise to prominence is the fact that it fusion protein comprised of a fatty acid-binding
was the first prokaryotic P450 enzyme recognized P450 (homologous to the eukaryotic CYP4 fatty
to interact with a eukaryotic-like P450 reductase acid hydroxylase family) fused to a CPR^^"^.
redox partner (i.e., a FAD- and FMN-containing Analysis of the catalytic properties of the purified
NADPH-cytochrome P450 reductase) rather enzyme showed it to have a very high catalytic
than with an iron-sulfur ferredoxin and FAD- center activity relative to its eukaryotic fatty acid
containing ferredoxin reductase, long considered hydroxylase relatives—with turnover numbers of
to represent the "typical" bacterial P450 reductase several thousand per minute with various long-
system^^l Thus P450 BM3, a soluble P450, off- chain fatty acids^^' ^^^. This was rationalized as
ered great promise as a tool for understanding being due to the efficiency of the electron transfer
redox partner interactions and electron transfer system covalently associated with the P450, and
in an experimentally more tractable system than opened the door to mechanistic and kinetic studies
was the case for the membrane-bound eukaryotic on the enzyme^^' ^^^' ^^^. In the BM3 system,
P450s and P450 reductase^. An even more attrac- NADPH reduces the FAD flavin transiently to the
tive feature of P450 BM3 was that it was a natural hydroquinone form. Electron transfer to the heme
fusion protein—^with the reductase linked to the is mediated by the FMN cofactor, which shuttles
P450 in a single 119 kDa pol5^eptide. Structural, electrons one at a time between the FAD and
spectroscopic, and enzymological studies of P450 heme^.
BM3 in the two decades since its discovery have Genetic dissection provided further proof for
been extremely informative as regards P450 struc- the multidomain nature of the enzyme-producing
ture and mechanism, establishing P450 BM3 first sub-genes encoding stable P450 (first —472
as a vitally important "model" system for the amino acids of P450 BM3) and CPR domain^^' ^^\
P450 superfamily^. and then constructs encoding the individual
P450 BM3 was discovered as a result of FAD/NADPH-binding and FMN-binding domains
studies by Armand Fulco's group on fatty acid of the CPR^^^. Domain dissection also enabled the
hydroxylase systems from the soil bacterium determination of the atomic structures of the heme
B. megaterium^^^. As implied by the name, BM3 (P450) domain of the enzyme and also the struc-
was the third enzyme with fatty acid hydroxylase ture of a nonstoichiometric complex of the heme
activity identified in the bacterium. P450 BM3 and FMN domains of the enzyme, arising from
became a major focus of Fulco's studies following proteolysis of a construct containing the heme
the finding that the gene was strongly induced by domain and the FMN-binding domain^^^.
addition of phenobarbital to the growth medium^^, The atomic structure of the heme domain has
suggesting that the gene was under transcriptional been solved in substrate-bound and substrate-free
control akin to the types of regulation seen previ- forms, showing that a large structural change in
ously for mammalian drug-metabolizing P450 the protein accompanies the binding of substrate
isoforms^^^. Subsequent studies have shown the (palmitoleic acid). Binding of fatty acid substrates
importance of a repressor protein (BM3R1) in the to the heme domain causes a shift in the heme iron
control of CYP102A1 expression, with barbitu- spin-state equilibrium toward the high-spin form,
rate drugs and a range of other hydrophobic mol- which is accompanied by a large increase in
ecules being able to displace the repressor to reduction potential for the ferric/ferrous transition
promote production of P450 BM312I' ^22. patty of the heme iron^^' ^^' ^^^. Binding of fatty acid
acids are likely to perform the same role in vivo, thus facilitates electron transfer to the heme iron
and have also been shown to facilitate expression and triggers catalysis. In the absence of bound
of a homolog of P450 BM3 in Bacillus subtilis. substrate there is negligible electron transfer
132 Mark J.I. Paine et al.

between the FMN and heme cofactors. When state as catalysis progresses (a neutral semi-
P450 BM3 binds its most efficient substrates, that quinone in the case of the FAD and a red, anionic
is those fatty acids, such as arachidonic acid, that semiquinone in the case of the FMN), suggesting
induce the greatest degrees of spin-state transition that the CPR domain of the enzyme cycles favor-
and promote the fastest turnover^ ^^' ^^^, FMN-to- ably between 0-2-1-0 electron-reduced states
heme electron transfer is rapid (c. 250 s^^ with during catalysis'^^. The situation appears similar
lauric acid) and catalysis occurs^^. The extent of for the housefly CPR, but contrasts with that
the increase in reduction potential accompanying reported for many eukaryotic CPRs, where the
saturation with fatty acid is approximately 130 mV, cycle appears to be 1-3-2-1 '^^. However, the pre-
a change which elevates the heme reduction cise reason for the apparent inactivation of BM3
potential above that of the ultimate reductant in in an "over-reduced" form remains obscure. Since
the system (NADPH: E^ = -320 mV cf -427 electron transfer to exogenous electron acceptors
for BM3 heme in absence of substrate and - 2 8 9 via the flavins remains largely unaflected'^^, it
in presence of arachidonate) and within range of appears to be the case that the catalytic step per-
that of the FMN cofactor (E^ = -203 mV for its turbed is that of first or second electron transfer to
oxidized/hydroquinone couple)^^' ^^K the heme iron. This may indicate that the reduc-
tion state and/or coenzyme-binding affects the
conformational status of the reductase domain,
in particular the movement of the FMN-binding
2.7.1. Electron Transfer Properties of
domain relative to the FAD-binding domain
BM3 Reductase
discussed above.
Interestingly, the BM3 reductase is the only Stopped-flow absorption spectroscopy has
member of the diflavin reductase class of enzymes provided information on some of the internal elec-
which does not stabilize a neutral, blue semi- tron transfer processes in P450 BM3^^. In particu-
quinone on its FMN. Formation of a blue semi- lar, the rate of electron transfer between NADPH
quinone during reductive titration is a feature and the FAD (in itself a composite of rates for
typical of flavodoxins, and retained in other mem- NADPH association, orientation and the actual
bers of the diflavin reductase family (e.g., nitric hydride transfer event) is very fast (500 s~' at
oxide synthase, methionine synthase reductase, 5°C) relative to those measured for mammalian
and other eukaryotic CPRs). The fact that the CPRs, nitric oxide synthase, methionine synthase
semiquinone is destabilized with respect to the reductase, and novel reductase 1 (NRl)^^' ^'^' ^^.
hydroquinone in BM3's CPR and FMN domains The rate of internal (inter-flavin) electron transfer
has been rationalized on the basis of the structure remains to be determined, but should be greater
of the FMN domain. The presence of basic than 250 s~' (the turnover rate with arachidonate).
residues (particularly Lys572 and Lys580) may The rapid electron transfer from NADPH to
stabilize the anionic hydroquinone form and flavin is diminished considerably for the NADH-
destabilize the semiquinone^'^. The redox state of dependent reaction, indicating the importance of
the flavins is of great importance to the catalytic the 2' phosphate group of NADPH in both recog-
properties of BM3, since early studies on the nition and binding of the pyridine nucleotide, and
enzyme showed that there was strong inhibition of in the actual electron transfer event. Comparison
fatty acid hydroxylase activity following preincu- of the thermodynamic properties of the BM3
bation of the enzyme with NADPH^^. It appears reductase with those of the other members of the
that an unfavorable three-electron (and possibly diflavin reductase family (Figure 4.8) indicates
four-electron) reduced form of the enzyme accu- that increased driving force does not underlie the
mulates within a few minutes of incubation of the enhanced rate of NADPH-to-FAD electron trans-
enzyme with NADPH, and that electron transfer fer observed for this isoform^^. For instance, in
in this species is diminished and results in a cat- human CPR there is a more favorable thermody-
alytic rate of fatty acid hydroxylation less than namic balance for electron transfer between
20% of the maximal rate. Analysis of the redox NADPH (E^' = -320 mV) and the FAD cofactor
state of the flavins during turnover suggests that (E^ = - 3 2 8 mV for the two-electron reduction to
both may exist predominantly in a semiquinone the hydroquinone form) and an apparent rate of
Electron Transfer Partners of Cytochrome P450 133

electron transfer of ~ 3 s~^ at 25°C with excess (ref [142]), and human CYP3A4 (ref [143]).
NADPH^^' ^l For BM3, the midpoint reduction Most recently, a fusion comprising entirely human
genes was constructed between CYP2D6 and
potential for the FAD cofactor is virtually identi-
human CPR^44
cal to that for human CPR (-332 mV), and yet the
rate of electron transfer is apparently —200-300- As yet none of these artificial systems
fold that of human CPR under similar conditions. approach P450 BM3 in terms of its high catalytic
Obviously, structural differences in BM3 reduc- activity. Indeed, most artificial mammalian
tase underlie its capacity to drive electron transfer fusions achieve no greater turnover levels than
so efficiently with respect to other diflavin reduc- their physiological counterparts in which the
tases. However, these differences await clarifica- reductase and P450 form noncovalent complexes.
tion through structural elucidation of the BM3 This may indicate that the exceptional catalytic
reductase and/or its FAD/NADPH domain. In rate exhibited by P450 BM3 arises from the
view of the proximity of the redox couples of details of its mechanism rather than simply
NADPH and the FAD flavin in BM3 (and other from the fact that the reductase and P450 are
diflavin reductases), reversibility of the electron covalently linked. Nevertheless, such catalytically
transfer process (i.e., hydride transfer from FAD self-sufficient P450-CPR fusion enzymes are
hydroquinone to NAD(P)^) might be expected. extremely useful for probing the biochemical
This has been shown to occur for both mammalian mechanisms involved in catalysis and electron
CPR and for BM3 reductase^^' ^^' 1^4 transfer between P450 and CPR. They also have
important practical applications in the develop-
Many aspects of the electron transfer reactions
ment of efficient P450-based biocatalysts and
in P450 BM3 are now reasonably well understood,
high throughput screening systems.
and the first mutagenesis experiments on the
enzyme ruled out the involvement of a tryptophan
residue adjacent to the heme in the flavin-to-heme
electron transport process^^^. However, even after 3. Electron Transfer to P450s
two decades of study of this enzyme, several fun-
from Cytochrome b^
damental issues remain unresolved. Key among
these are the reasons why the electron transfer
reactions in the reductase domain of the enzyme Cytochrome b^ was originally discovered in
(and between FMN and heme) are so efficient by silkworm larvae ^"^^ and has since been most exten-
comparison with the eukaryotic P450 and CPR sively studied in mammals^' ^^^. Like cytochromes
systems. Thus, further detailed rapid kinetic and P450 and CPR, cytochrome b^ is an integral mem-
structural studies are critical to gain a complete brane protein located on the C5^osolic side of the
understanding of this efficient electron transfer endoplasmic reticulum where it is principally
system. involved in lipid biosynthesis. It functions along
with cytochrome b^ reductase as the electron
donor to microsomal desaturases that synthe-
size unsaturated fatty acids, plasmalogens, and
2.8. Artificial CPR-P450 Fusion
sterols^'^^. Indications of the involvement of
Constructs cytochrome b^ in P450 reactions date from early
Many attempts have been made to increase studies that showed that NADH stimulates
the catalytic efficiency of mammalian P450s to the NADPH-supported metabolism of drugs in micro-
level of BM3 through artificial fusions with somes^"^^' ^^^. Since then, evidence has accumu-
CPR. The first such artificial fusion between rat lated that c)^ochrome b^ has a significant role in a
P450 CYPlAl and rat CPR was constructed by number of P450 systems^^^, most significantly
Murakami et alP^. Since then, several fusions CYP3A4 (ref. [151]), which is involved in the
with rat CPR have been constructed including majority of human drug oxidations.
bovine CYP17a (ref [137]), human CYP3A4 Cytochrome b^ is a small polypeptide
(ref [138]), human CYPlAl (ref [139]), CYP4A1 (~17kDa) containing 134 residues, which are
(ref [140]), and rat CYP2C11 (ref [141]). In divided into an amino-terminal hydrophilic heme
addition, yeast CPR has been fused to rat CYPlAl domain and a carboxyl-terminal hydrophobic
134 Mark J.I. Paine et al.

membrane-binding region^^^' ^^^. While the mem- P450 has provided a means of studying the actions
brane-bound form of Z?5 participates in elongation of the individual components in a more physio-
and desaturation of fatty acids^^' ^^^^ ^^^, cholesterol logical context. Using yeast systems, Perret and
biosynthesis^^^, and drug metabohsm, soluble forms Pompon^^^ have shown that the redox properties
of Z?3 and Z>3 reductase are also found in er5^hrocytes, of Z?5 were strictly required to enhance CYP3A4
where they catalyze methemoglobin reduction^^^. activity, with results of rapid kinetic analysis of Z73
Mutations to either the b^ gene or the b^ reductase reduction following NADPH addition suggesting
gene can cause congenital methemoglobinemia^^^. that Z>3 was reduced by the 3A4 ferrous-dioxygen
Soluble Z?5 has also been purified as a cytosolic complex and reoxidized by subsequent P450 oxy-
component in the NAD(P)H-reductive activation genated intermediates ^^^. In E. coli, cytochrome
pathway for porcine methionine synthase^^. b^ significantly enhances the testosterone and
Like CPR, it is generally accepted that nifedipine activity of CYP3A4 and results in
charge-pair interactions drive the formation of the stabilization of the P450 during substrate
P450:Z?5 complexes^^^~^^^. This is supported by turnover'^^. This is consistent with the proposal
site-directed mutagenesis of rabbit CYP2B4, that b^ interaction reduces uncoupling and super-
which has identified several positively charged oxide anion release'^^' '^^.
residues on the P450 surface (R122, R126, R133, With the increasing sophistication in the
K139, and K433) as being involved in binding b^ genetic tools now available it is becoming possible
(ref [58]). However, the precise mechanism by to investigate the complex interrelationships of
which b^ supports P450 catalysis is not yet clear. the monooxygenase system in the correct context
As the redox potentials of cytochrome b^ of the whole organism. For instance, the absence
and ferric P450 are approximately +25 mV and of any detectable P450 activity in mice where
-300 mV^^^ respectively, a role in the first elec- CPR has been knocked out of the liver provides
tron transfer step of the P450 catalytic cycle is compelling evidence that the alternative cyto-
unlikely, suggesting that b^ may only be involved chrome b^/b^ reductase electron transfer pathway
in the donation of the second electron''^^' •'*^. Since does not play any significant role in vivo^^. We
electrons may be supplied to cytochrome b^ by now await the construction ofb^ knockout mice to
either NADH cytochrome b^ reductase or CPR^^, provide further clues as to the precise role of b^
the pathway of electron movement is difficult to in the physiological function of P450s in higher
establish. Furthermore, studies that show that vertebrate organisms.
apocytochrome b^ can stimulate the catalytic
activity of CYP3A4^^4' ^^^ 2C9, 4A7, and 17^^^
question a formal role in electron transfer. Thus,
over the years several different mechanisms have 4. Iron-Sulfur Electron Donors:
been suggested by which b^ supports P450 cataly- Adrenodoxin, Putidaredoxin,
sis. These include: electron transfer from NADPH and their Reductases
through CPR to cytochrome b^ and thence to
P450166,167. electron transfer from NADH through
b^ reductase to cytochrome b^ and thence to 4.1. General
P450'^^; electron shuttling between P450 and b^ The Fe2S2-type ferredoxins can be arranged
resulting in a tighter coupling between electron into three distantly related classes based on amino
flow and product formation^^^; reduction in the acid sequence homologies: bacterial-, plant-, and
uncoupled generation of superoxide or hydrogen vertebrate-type*^"*. Extensive information on the
peroxide^''^; and an allosteric effect not dependent function and mechanisms of this system has been
on electron transfer^ ^'*' ^^^. gained through work on the bacterial P450cam
Much of the early data on cytochrome b^ system in Pseudomonas putida, which catalyzes
interactions with P450 has come from in vitro the conversion of (i-camphor to 5-exo-hydroxy-
analysis using reconstituted systems. However, the camphor^^^. In P. putida, the iron-sulfur protein
expression in E. coli^^^, insect cells^^^ or yeast^^^ is putidaredoxin (Pdx), a 106 amino acid resi-
of a complete mammalian P450 monooxygenase due ferredoxin. For catalysis, two reducing equiv-
complex comprising cytochrome b^, CPR, and alents are sequentially transferred from NADH
Electron Transfer Partners of Cytochrome P450 135

to P450cam via an FAD-containing enzyme, of electron transferases that includes bacterial-


putidaredoxin reductase (Pdr) and Pd. The puti- type putidaredoxin reductase (PdR)*^^, animal-type
daredoxin: P450cam binary complex forms with adrenodoxin reductase (AdR)*^^ *^^, and plant-
a K^ that is dependent on the oxidation states of type ferredoxin-NADP^ reductases'^. An AdR-
the proteins and on whether substrate is bound to type reductase system (FprA) has recently been
the P450^^^. In the first electron transfer, reduced characterized in the pathogen M. tuberculosis^^
Pd interacts with ferric (i-camphor-bound and the structure reveals extensive similarity with
P450cam and reduces it to the ferrous form. In the the mammalian enzyme*^^. In humans, AdR is
second electron transfer, reduced Pd forms a com- most abundant in the steroid-producing cells of
plex with ferrous dioxygen and (i-camphor-bound the adrenal cortex, the ovary, and the testis*^'.
P450cam to transfer the second electron and
reductively cleave bound dioxygen, producing
hydroxycamphor and water*^^. While most struc-
4.2. Interactions with P450
tural and biophysical studies of P450/ferredoxin
interactions have focused on the Fe2S2 putidare- The structures and models of structures of
doxin from P. putida (and its cognate P450cam), several bacterial, animal, and plant ferredoxins are
other types of bacterial ferredoxins are recognized available (e.g., putidaredoxin^^^ and the Fe4S4
to interact productively with P450s. For instance, ferredoxin from Bacillus thermoproteolyticus^^^).
the Fe3S4 ferredoxin SoyB is a redox partner In particular, details of the electron transfer mecha-
for the Streptomyces griseus P450 Soy (SoyC)*^^ nisms have developed through kinetic and muta-
and the Fe^S^ ferredoxin from B. subtilis (fer) has tional analysis of the Pdx-P450cam system^'^.
been shown to undergo redox reactions with P450 These indicate that Pdx interacts with proximal sur-
Biol from the same bacterium*^^. face of P450cam through electrostatic interactions
In animals, ferredoxins are primarily involved involving the acidic Pdx residues Asp38 and Asp34
in electron transfer interactions involving mito- and the basic P450cam residue Arg 112^^^' ^^^
chondrial P450s. This fact is in agreement with Additionally, Argl 12 appears to form the electron
the theory of the prokaryotic origin of this transfer route in the P450cam-Pdx complex with
organelle. Much work has been done with adreno- Cys39 and Asp38 of Pdx^^^' ^^^. A role for the
doxin (Adx) and the transfer of electrons from C-terminal amino acid of Pdx (W106) in electron
NADPH-dependent adrenodoxin reductase (AdR) transfer to P450cam was suggested on the basis of
to CYPUAl, which is involved in cholesterol diminished enzymatic activity in systems with
side chain cleavage, and to CYPUB, involved in Pdx W106 mutants, but later studies of various
the formation of Cortisol and aldosterone*^"*. W106 mutants showed that this residue primarily
Bovine adrenodoxin mRNA is translated as a 186 affects the K^ of Pdx for the P450i9^ Studies of
amino acid precursor molecule, which undergoes the interactions between cytochrome b^ and
proteolytic processing upon import into the mito- P450cam show that b^ and Pdx compete for the
chondria to remove the N-terminal 58 residues *^^. same binding site on the P450, suggesting a
The mature Adx comprises 128 amino acid common mode of charge-charge pairing that is
residues and has a molecular weight of 14.4 kDa. the major force driving interactions at the same
In plants, [2Fe-2S] ferredoxins are involved in surface on the P450^^'^.
conjunction with ferredoxin-reductase in photo- However, isothermal calorimetry experiments
synthesis reactions and the transfer of electrons show that the energetics of the P450cam/Pdx
from photosystem I to NADP^*^*. However, evi- association are most similar to those of binding
dence for involvement of ferredoxins in electron reactions of antibody to antigen complexes ^^^,
transfer to plant P450s awaits a more detailed char- suggesting that van der Waals and hydrogen-bond-
acterization of the multiple P450 systems present ing interactions may predominate in the formation
in the biotechnologically important plant species. of P450cam-Pdx complexes. This study also sug-
Ferredoxins receive their electrons from gested that binding between Pdr and Pdx might
ferredoxin reductase, which represents the first be dominated by hydrophobic interactions. Not-
component of the electron transfer system. These withstanding these data, a variety of mutagenesis
enzymes contain FAD and belong to a large group studies along with the fact that increased ionic
136 Mark J.I. Paine et al.

strength markedly increases the K^ of Pdx for the facilitating heterolysis of the O-O bond^^^. It is
P450, highlight the important influence of electro- thus proposed that in addition to its redox
statics on the docking process^^^' ^^^. function, Pdx invokes structural changes that
Catalytic interactions between Pdx and facilitate the oxygen activation reaction^^^ Only
P450cam are considered to follow a ping-pong Pdx is capable of reducing the oxy form of
mechanism^^^. The flavin (FAD) of Pdr is reduced P450cam, in agreement with this hypothesis^^^.
directly to its hydroquinone by interaction with The rate-limiting steps in the entire catalytic
the obligatory two-electron donor NADH, but cycle of P450cam are the electron transfers
negligible formation of semiquinone is observed between Pdx and the P450. The precise rate-
during the successive single-electron transfer limiting step appears to change from the first
reactions between Pdr and Pdx in steady-state electron transfer to the second electron transfer as
turnover of P450cam. This indicates destabiliza- the concentration of Pdx in the system is reduced
tion of the flavin semiquinone with respect to the toward levels considered typical of the situation
hydroquinone and oxidized forms ^^^. Rapid (laser in v/vo^^^.
flash photolysis) kinetic studies indicate transient With respect to the mammalian adrenodoxin/
formation of a blue semiquinone on the Pdr FAD, adrenodoxin reductase system, crystal structures
which disproportionates rapidly ^^^. of truncated and full-length oxidized bovine
Upon complex formation, Pdx induces confor- Adx^^^' ^^"^ have been determined, along with
mational changes in P450cam'^^ which convert NMR structures of both oxidized and reduced
the high-spin state of ferric P450cam to low-spin full-length forms of Adx^^^. Adrenodoxin, con-
state and stretch the heme-axial ligand^^^. tains a large hydrophobic core region that houses
NMR studies of the Pdx-P450cam complex also a single Fe2S2 cluster, and an interaction domain
show that Pdx binding perturbs several regions that contains acidic residues responsible for the
involving the substrate access channel in recognition of the redox partners, CYPllAl
P450cam^^^ and induces a tilt in the heme plane, and AdR206' 207 (pigure 4.11). The single Fe2S2
leading to the movement of J-camphor and the cluster is ligated by four cysteinyl thiolate ligands,
axial Cys to the heme-iron by 0.1-0.5 A and Cys46, Cys52, Cys55, Cys92, replacement of any

Interaction
Domain
Core
Domain

Figure 4.11. Ribbon representation of bovine Adx (pdb codelAYF). The Fe2S2 cluster is shown in black ligated
to cysteines 46, 52, 55, and 92. The Fe and S atoms are represented as small and large spheres respectively
Electron Transfer Partners of Cytochrome P450 137

of which, produces a nonfunctional apoprotein^^^. groove between a9 and pi2 at the surface (Figure
Sites involved in redox partner binding are also 4.12), which can accommodate the C-terminal
present in the core domain centered around an chain end of adrenodoxin^^^. Both Adx and AdR
acidic patch at position Asp59^^''. contain highly asymmetric surface charge distri-
Two different models have been described to butions, which indicates that interactions between
explain the electron delivery system mediated by these molecules and P450 are mediated by elec-
Adx: (a) an organized cluster model comprising trostatic interactions ^^'^. The structural data are
AdR: Adx: P450 ternary/quaternary complexes^^^ largely confirmatory of the earlier data from
and (b) a shuttle system where Adx sequentially mutagenesis and modification studies, showing
transports one electron at a time from AdR to the importance of charge pairing in the binary
P45Q210 Qf these^ t^g shuttle hypothesis is most complex between AdR and Ad^^^. Most recently,
favorable since the formation of a ternary com- the structure of the AdR/Ad complex was solved
plex formed by AdR, Adx, and P450 is unlikely at 2.3 A resolution^^^. Interactions sites were con-
from a structural perspective due to overlapping firmed as involving predominantly acidic residues
mteraction regions^ The sequential model of Adx (D76, 79, 72, and 39) with basic side
is also that favored for the well-characterized chains of Adr (R211, 240, 244, and K27).
P450cam system. Following the resolution of In the P450cam system, our understanding of
the bovine AdR crystal structure it has become the nature of the interactions between the Pdr and
possible to fully investigate the structural mecha- Pdx components is not fully developed. However,
nisms driving the protein complex formation^ ^^. Cys73 of Pdx has been reported as important in
The distinguishing feature of AdR is a large the docking interaction between Pdx and Pdr^^^.

AdR

Adx

,.. ^^::,

Figure 4.12. Surface representations of Adx and AdR (pdb codes lAYF and ICJC respectively). The molecules
are oriented according to their proposed docking positions^^"^j which involves charge-pair interactions between acidic
residues at the C-terminal end of Adx and basic residues around the docking groove of AdR.
138 Mark J.I. Paine ef a/.

Mutations to the adjacent Pdx surface residue ongoing genome sequencing projects reveal ever
Glu72 also affect electron transfer reactions in more diverse means by which this intriguing
the P450cam redox system^^^. The generation of class of enzymes can operate and obtain reducing
catalytically efficient putidaredoxin reductase- power.
putidaredoxin-P450cam triple fusion protein
demonstrates that a much more simple single
component camphor hydroxylase system can be Acknowledgments
produced, with obvious biotechnological implica-
tions. The addition of short peptide linkers The authors gratefully acknowledge the sup-
between the respective polypeptides was compati- port of the UK Medical Research Council, Cancer
ble with strong expression of fusion proteins in Research UK, the Biotechnology and Biological
an E. coli system and efficient electron transfer Sciences Research Council, the Wellcome Trust,
reactions between the redox centers^^'^. Although the Lister Institute of Preventative Medicine, and
much structure/function work remains to be done the European Union, EPSRC (Engineering and
on the Pdr component of the P450cam system, a Physical Sciences Research Council) and the
novel finding regarding its activity was made Royal Society.
recently with the demonstration that Pdr has
NAD(H)-dependent dithiol/disulfide oxidoreduc-
tase activity^^2. However, much structural and References
kinetic work remains to be done to provide a clear
description of the nature of interactions between 1. Schenkman, J.B. and I. Jansson (1999). Interactions
the Pdr and Pdx components of the P450cam sys- between cytochrome P450 and cytochrome b5.
tem, and how these relate to those in the analogous DrugMetab. Rev. 31, 351-364.
Adr/Adx mitochondrial system. 2. Munro, A.W., D.G. Leys, K.J. McLean, K.R.
Marshall, TW.B. Ost, S. Daff et al. (2002). P450
BM3: The very model of a modern flavocy-
tochrome. Trends Biochem. Sci. 21, 250-257.
5. Novel Redox Systems
3. Nakayama, N., A. Takemae, and H. Shoun (1996).
Cytochrome P450foxy, a catalytically self-sufficient
The identification of the P450 BM3 system led fatty acid hydroxylase of the fungus Fusarium
to the realization that alternative types of P450 oxysporum. J. Biochem. (Tokyo) 119, 435^40.
redox systems (beyond those typified by mam- 4. De Mot, R. and A.H. Parret (2002). A novel class of
malian hepatic class and P450cam) exist in nature. self-sufficient cytochrome P450 monooxygenases
In recent years, a variety of alternative modes of in prokaryotes. Trends Microbiol. 10, 502-508.
P450 reduction have been reported—including 5. Kluck, R.M., E. Bossy-Wetzel, D.R. Green, and
P450s that require no electron transfer at all (i.e., D.D. Newmeyer (1997). The release of cytochrome
c from mitochondria: A primary site for Bcl-2
catalyze molecular rearrangements without oxy-
regulation of apoptosis. Science 275, 1132-1136.
gen chemistry) such as the dehydratase allene
6. Kluck, R.M., S.J. Martin, B.M. Hoffman, J.S. Zhou,
oxide synthase^^^ and a P450 that appears to inter- D.R. Green, and D.D. Newmeyer (1997). Cyto-
act directly with hydrogen peroxide to facilitate chrome c activation of CPP32-like proteolysis plays
fatty acid oxidation in B. subtilis^^^. Various types a critical role in a Xenopus cell-free apoptosis
of ferredoxins have been shown to be competent system. EMBO J. 16, 4639-^649.
redox partners for selected P450 enzymes^'^ 7. Haas, E., B.L. Horecker, and T.T. Hogness (1940).
and fiavodoxins may also be physiologically The enzymatic reduction of cytochrome c\ Cyto-
relevant mediators of electron transfer to P450s chrome c reductase. J. Biol. Chem. 136,141-714.
in bacterial (and possibly plant) systems^^^' ^^^. 8. Horecker, B.L. (1950), Triphosphopyridine
More recent studies suggest more exotic types nucleotide-cytochrome c reductase in liver. J. Biol.
Chem. 183, 593-605.
of P450 fusion enzymes may exist—including
9. Williams, C.H., Jr. and H. Kamin (1962). Micro-
fusions of P450s to ferredoxin and phthalate
somal triphosphopyridine nucleotide—cytochrome
dioxygenase reductase-like domains^^^' •^^K It reductase of the liver. J. Biol. Chem. 237, 587-595.
is highly likely that further insights into bio- 10. Phillips, A.H. and R.G. Langdon (1962).
diversity of P450 redox systems will be gained as Hepatic triphosphopyridine nucleotide cytochrome
Electron Transfer Partners of Cytochrome P450 139

c reductase: Isolation characterisation and kinetic vaccinia virus-mediated expression and localization
studies. J. Biol. Chem. 237, 2652-2660. of the CYPOR gene to chromosome 7. Mol.
11. Lu, A.Y. and M.J. Coon (1968). Role of hemopro- Pharmacol. 36, 83-88.
tein P-450 in fatty acid omega-hydroxylation in a 22. Wang, M., D.L. Roberts, R. Paschke, T.M. Shea,
soluble enzyme system from liver microsomes. B.S. Masters, and J.J. Kim (1997). Three-dimensional
J. Biol. Chem. 243, 1331-1332. structure of NADPH-cytochrome P450 reductase:
12. Lu, A.Y., K.W. Junk, and M.J. Coon (1969). Prototype for FMN- and FAD-containing enzymes.
Resolution of the cytochrome P-450-containing Proc. Natl. Acad. Sci. USA 94, 8411-8416.
omega-hydroxylation system of liver microsomes 23. Porter, T.D. and C.B. Kasper (1986). NADPH-
into three components. J. Biol. Chem. 244, cytochrome P-450 oxidoreductase: Flavin mononu-
3714-3721. cleotide and flavin adenine dinucleotide domains
13. Strobel, H.W., A.Y. Lu, J. Heidema, and M.J. Coon evolved from different flavoproteins. Biochemistry
(1970). Phosphatidylcholine requirement in the 25,1682-1687.
enzymatic reduction of hemoprotein P-450 and in 24. Leclerc, D., A. Wilson, R. Dumas, C. Gafuik,
fatty acid, hydrocarbon, and drug hydroxylation. D. Song, D. Watkins et al. (1998). Cloning and
J. Biol. Chem. 245, 4851^854. mapping of a cDNA for methionine synthase reduc-
14. Black, S.D., J.S. French, C.H. Williams, Jr., and tase, a flavoprotein defective in patients with
M.J. Coon (1979). Role of a hydrophobic poly- homocystinuria. Proc. Natl. Acad. Sci. USA 95,
peptide in the N-terminal region of NADPH- 3059-3064.
cytochrome P-450 reductase in complex formation 25. Paine, M.J., A.P Garner, D. Powell, J. Sibbald,
with P-450LM. Biochem. Biophys. Res. Commun. M. Sales, N. Pratt et al. (2000). Cloning and charac-
91, 1528-1535. terization of a novel human dual flavin reductase.
15. Yasukochi, Y and B.S. Masters (1976). Some J. Biol. Chem. 275, 1471-1478.
properties of a detergent-solubilized NADPH- 26. McMillan, K., D.S. Bredt, D.J. Hirsch, S.H. Snyder,
cytochrome c (cytochrome P-450) reductase J.E. Clark, and B.S. Masters (1992). Cloned,
purified by biospecific affinity chromatography. expressed rat cerebellar nitric oxide synthase con-
J. Biol. Chem. 251, 5337-5344. tains stoichiometric amounts of heme, which binds
16. lyanagi, T. and H.S. Mason (1973). Some proper- carbon monoxide. Proc. Natl. Acad. Sci. USA 89,
ties of hepatic reduced nicotinamide adenine 11141-11145.
dinucleotide phosphate-cytochrome c reductase. 27. Narhi, L.O. and A.J. Fulco (1986). Characterization
Biochemistry 12, 2297-2308. of a catalytically self-sufficient 119,000-dalton
17. lyanagi, T., N. Makino, and H.S. Mason (1974). cytochrome P-450 monooxygenase induced by
Redox properties of the reduced nicotinamide barbiturates in Bacillus megaterium. J. Biol. Chem.
adenine dinucleotide phosphate-cytochrome P-450 261, 7160-7169.
and reduced nicotinamide adenine dinucleotide- 28. Ostrowski, J., M.J. Barber, D.C. Rueger, B.E.
cytochrome b5 reductases. Biochemistry 13, Miller, L.M. Siegel, and N.M. Kredich (1989).
1701-1710. Characterization of the flavoprotein moieties
18. Backes, W.L. (1993). NADPH-cytochrome P450 of NADPH-sulfite reductase from Salmonella
reductase: Function. In J.B. Schenkman and typhimurium and Escherichia coli. Physicochemical
H. Greim (eds) Handbook of Experimental and catalytic properties, amino acid sequence
Pharmacology, \o\. 105. Springer-Verlag, Berlin. deduced from DNA sequence of cysJ, and compari-
19. Porter, T.D. and C.B. Kasper (1985). Coding son with NADPH-cytochrome P-450 reductase.
nucleotide sequence of rat NADPH-cytochrome P- J. Biol. Chem. 264, 15796-15808.
450 oxidoreductase cDNA and identification of 29. Smith, G.C.M., D.G. Tew, and C.R. Wolf (1994).
flavin-binding domains. Proc. Natl. Acad. Sci. USA Dissection of NADPH-cytochrome P450 oxidore-
82, 973-977. ductase into distinct functional domains. Proc. Natl.
20. Katagiri, M., H. Murakami, Y Yabusaki, Acad Sci. USA 91, 8710-8714.
T. Sugiyama, M. Okamoto, T. Yamano et al. (1986). 30. Hodgson, A.V and H.W Strobel (1996). Character-
Molecular cloning and sequence analysis of full- ization of the FAD binding domain of cytochrome
length cDNA for rabbit liver NADPH-cytochrome P450 reductase. Arch. Biochem. Biophys. 325,
P-450 reductase mRNA. J. Biochem. (Tokyo) 100, 99-106.
945-954. 31. Sevrioukova, I., G. Truan, and J.A. Peterson (1996).
21. Yamano, S., T. Aoyama, O.W. McBride, The flavoprotein domain of P450BM-3: Expression,
J.P. Hardwick, H.V Gelboin, and F.J. Gonzalez purification, and properties of the flavin adenine
(1989). Human NADPH-P450 oxidoreductase: dinucleotide- and flavin mononucleotide-binding
Complementary DNA cloning, sequence and subdomains. Biochemistry 35, 7528-7535.
140 Mark J.I. Paine et al.

32. Munro, A.W. (1993). Purification schemes for the NADPH cytochrome P-450 oxidoreductase gene.
constituent domains of cytochrome P450 BM3 in Arch. Biochem. Biophys. 330, 271-280.
E. coli. Biochem. Soc. Trans. 21, 316S. 44. O'Leary, K.A., H.C. Li, PA. Ram, P McQuiddy,
33. Finn, R.D., J. Basran, O. Roitel, C.R. Wolf, A.W. D.J. Waxman, and C.B. Kasper (1997). Thyroid reg-
Munro, M.J. Paine et al. (2003). Determination of ulation of NADPH:cytochrome P450 oxidoreduc-
the redox potentials and electron transfer properties tase: Identification of a thyroid-responsive element
of the FAD- and FMN-binding domains of the in the 5'-flank of the oxidoreductase gene. Mol.
human oxidoreductase NRl. Eur. J. Biochem. 270, Pharmacol. 52, 46-53.
1164-1175. 45. Ram, PA. and D.J. Waxman (1992). Thyroid
34. Wolthers, K.R., J. Basran, A.W. Munro, and N.S. hormone stimulation of NADPH P450 reductase
Scrutton (2003). Molecular dissection of human expression in liver and extrahepatic tissues.
methionine synthase reductase: Determination of Regulation by multiple mechanisms. J. Biol. Chem.
the flavin redox potentials in full-length enzyme and 267,3294-3301.
isolated flavin-binding domains. Biochemistry 42, 46. Fan, L.Q., J. Coley, R.T Miller, R.C. Cattley, and J.C.
3911-3920. Corton (2003). Opposing mechanisms of NADPH-
35. Benveniste, I., A. Lesot, M.P. Hasenfratz, G. Kochs, cytochrome P450 oxidoreductase regulation by
and F. Durst (1991). Multiple forms of NADPH- peroxisome proliferators. Biochem. Pharmacol. 65,
cytochrome P450 reductase in higher plants. 949-959.
Biochem. Biophys. Res. Commun. 177, 105-112. 47. Le, Y. and B. Sauer (2001). Conditional gene knock-
36. Koopmann, E. and K. Hahlbrock (1997). out using Cre recombinase. Mol. Biotechnol. 17,
Differentially regulated NADPH:cytochrome P450 269-275.
oxidoreductases in parsley. Proc. Natl. Acad. Sci. 48. Sutter, T.R. and J.C. Loper (1989). Disruption of the
USA 94, 14954-14959. Saccharomyces cerevisiae gene for NADPH-
37. Urban, P., C. Mignotte, M. Kazmaier, F. Delorme, cytochrome P450 reductase causes increased
and D. Pompon (1997). Cloning, yeast expression, sensitivity to ketoconazole. Biochem. Biophys. Res.
and characterization of the coupling of two distantly Commun. 160, 1257-1266.
related Arabidopsis thaliana NADPH-cytochrome 49. Venkateswarlu, K., DC. Lamb, D.E. Kelly, N.J.
P450 reductases with P450 CYP73A5. J. Biol. Manning, and S.L. Kelly (1998). The N-terminal
Chem. 212, 19176-19186. membrane domain of yeast NADPH-cytochrome
38. Porter, T.D., T.W Beck, and C.B. Kasper (1990). P450 (CYP) oxidoreductase is not required for
NADPH-cytochrome P-450 oxidoreductase gene catalytic activity in sterol biosynthesis or in recon-
organization correlates with structural domains of stitution of CYP activity J. Biol. Chem. 273,
the protein. Biochemistry 29, 9814-9818. 4492-4496.
39. Henderson, C.J., DM. Otto, D Carrie, 50. Enoch, H.G. and P Strittmatter (1979). Cytochrome
M.A. Magnuson, A.W. McLaren, I. Rosewell et al. b5 reduction by NADPH-cytochrome P-450 reduc-
(2003). Inactivation of the hepatic cytochrome P450 tase. J 5/o/. Chem. 254, 8976-8981.
system by conditional deletion of hepatic cytochrome 51. Schacter, B.A., E.B. Nelson, H.S. Marver, and B.S.
P450 reductase. J. Biol. Chem. 278, 13480-13486. Masters (1972). Immunochemical evidence for an
40. Shen, A.L., K.A. O'Leary, and C.B. Kasper (2002). association of heme oxygenase with the microsomal
Association of multiple developmental defects and electron transport system. J. Biol. Chem. 247,
embryonic lethality with loss of microsomal 3601-3607.
NADPH-cytochrome P450 oxidoreductase. J. Biol. 52. Chen, Z. and R. Banerjee (1998). Purification of
Chem. 277,6536-6541. soluble cytochrome b5 as a component of the reduc-
41. Gonzalez, FJ. and C.B. Kasper (1982). Cloning of tive activation of porcine methionine synthase.
DNA complementary to rat liver NADPH- J. Biol. Chem. 273, 26248-26255.
cytochrome c (P-450) oxidoreductase and 53. Osada, M., S. Imaoka, T. Sugimoto, T Hiroi, and
cytochrome P-450b mRNAs. Evidence that pheno- Y Funae (2002). NADPH-cytochrome P-450 reduc-
barbital augments transcription of specific genes. tase in the plasma membrane modulates the activa-
J. Biol. Chem. 257, 5962-5968. tion of hypoxia-inducible factor 1. J. Biol. Chem.
42. Shen, A.L. and C.B. Casper (1993). Protein and 277, 23367-23373.
gene regulation of NADPH-cytochrome P450 oxi- 54. Bachur, N.R., S.L. Gordon, M.V. Gee, and H. Kon
doreductase. In J.B. Shenkman and H. Grein (eds). (1979). NADPH cytochrome P-450 reductase acti-
Handbook ofExperimental Pharamacology, Vol. 105. vation of quinone anticancer agents to free radicals.
Springer-Verlag, Berlin. Proc. Natl. Acad. Sci. USA 76, 954-957.
43. O'Leary, K.A., P McQuiddy, and C.B. Kasper 55. Keyes, S.R., P.M. Fracasso, D.C. Heimbrook,
(1996). Transcriptional regulation of the TATA-less S. Rockwell, S.G. Sligar, and A.C. Sartorelh (1984).
Electron Transfer Partners of Cytochrome P450 141

Role of NADPH:cytochrome c reductase and DT- of the conserved phenylalanine 181 of NADPH-
diaphorase in the biotransformation of mitomycin cytochrome p450 oxidoreductase in FMN binding
CI. Cancer Res. 44, 5638-5643. and catalytic activity. Biochemistry 40,13439-13447.
56. Walton, M.I., C.R. Wolf, and R Workman (1992). 67. Shen, A.L. and C.B. Kasper (2000). Differential
The role of cytochrome P450 and cytochrome P450 contributions of NADPH-cytochrome P450 oxido-
reductase in the reductive bioactivation of the novel reductase FAD binding site residues to flavin bind-
benzotriazine di-N-oxide hypoxic cytotoxin 3-amino- ing and catalysis. J. Biol. Chem. 275, 41087^1091.
l,2,4-benzotriazine-l,4-dioxide (SR 4233, WIN 68. Sem, D.S. and C.B. Kasper (1992). Geometric rela-
59075) by mouse liver. Biochem. Pharmacol. 44, tionship between the nicotinamide and isoalloxazine
251-259. rings in NADPH-cytochrome P-450 oxidoreduc-
57. Vasquez-Vivar, J., P. Martasek, N. Hogg, tase: Implications for the classification of evolu-
B.S. Masters, K.A. Pritchard, Jr., and tionarily and functionally related flavoproteins.
B. Kalyanaraman (1997). Endothelial nitric oxide Biochemistry 31, 3391-3398.
synthase-dependent superoxide generation from 69. Serre, L., F.M. Vellieux, M. Medina, C. Gomez-
adriamycin. Biochemistry 36, 11293-11297. Moreno, J.C. Fontecilla-Camps, and M. Frey,
58. Bridges, A., L. Gruenke, Y.T. Chang, I.A. Vakser, (1996). X-ray structure of the ferredoxin:NADP+
G. Loew, and L. Waskell (1998). Identification of reductase from the cyanobacterium Anabaena PCC
the binding site on cytochrome P450 2B4 for 7119 at 1.8 A resolution, and crystallographic stud-
cytochrome b(5) and cytochrome P450 reductase. ies of NADP+ binding at 2.25 A resolution. J. Mol.
J. Biol Chem. li:^, 17036-17049. Biol. 263, 20-39.
59. Wang, J. and PR. Ortiz De Montellano (2003). The 70. Karplus, PA., M.J. Daniels, and J.R. Herriott
binding sites on human heme oxygenase-1 for (1991). Atomic structure of ferredoxin-NADP+
cytochrome P450 reductase and biliverdin reduc- reductase: Protype for a structurally novel flavoen-
tase. J. Biol. Chem. 6, 6. zyme family. Science 251, 60-66.
60. Hubbard, PA., A.L. Shen, R. Paschke, C.B. Kasper, 71. Gruez, A., D. Pignol, M. Zeghouf, 1 Coves,
and J.J. Kim (2001). NADPH-cytochrome P450 oxi- M. Fontecave, J.L. Ferrer et al. (2000). Four crystal
doreductase. Structural basis for hydride and elec- structures of the 60 kDa flavoprotein monomer of
tron transfer. J. Biol. Chem. 276, 29163-29170. the sulfite reductase indicate a disordered flavo-
61. Craig, D.H., S.K. Chapman, and S. Daff (2002). doxin-like module. J. Mol Biol 299, 199-212.
Calmodulin activates electron transfer through neu- 72. Dohr, O., M.J. Paine, T. Friedberg, G.C. Roberts,
ronal nitric-oxide synthase reductase domain by and C.R. Wolf (2001). Engineering of a functional
releasing an NADPH-dependent conformational human NADH-dependent cytochrome P450 system.
lock. J. Biol. Chem. Ill, 33987-33994. Proc. Natl Acad. ScL USA 98, 81-86.
62. Barsukov, I., S. Modi, L.Y. Lian, K.H. Sze, M.J. 73. Gutierrez, A., O. Doehr, M. Paine, C.R. Wolf,
Paine, C.R. Wolfe/ al (1997). IH, 15N and 13C N.S. Scrutton, and G.C.K. Roberts (2000). Trp-676
NMR resonance assignment, secondary structure facilitates nicotinamide coenzyme exchange in the
and global fold of the FMN-binding domain of reductive half-reaction of human cytochrome P450
human cytochrome P450 reductase. J. Biomol. NMR reductase: Properties of the soluble W676H and
10, 63-75. W676A mutant reductases. Biochemistry 39,
63. Zhao, Q., S. Modi, G. Smith, M. Paine, PD. 15990-15999.
McDonagh, C.R. Wolfe/ al. (1999). Crystal struc- 74. Piubelli, L., A. Aliverti, A.K. Arakaki, N. Carrillo,
ture of the FMN-binding domain of human E.A. Ceccarelli, PA. Karplus et al (2000).
cytochrome P450 reductase at 1.93 A resolution. Competition between C-terminal tyrosine and
Protein Sci. 8, 298-306. nicotinamide modulates pyridine nucleotide affinity
64. Ludwig, M.L., K.A. Pattridge, A.L. Metzger, M.M. and specificity in plant ferredoxin-NADP(+)
Dixon, M. Eren, Y. Feng et al. (1997). Control of reductase. J 5/o/. Chem. 275, 10472-10476.
oxidation-reduction potentials in flavodoxin from 75. Deng, Z., A. Aliverti, G. Zanetti, A.K. Arakaki,
Clostridium beijerinckii: The role of conformation J. Ottado, E.G. Orellano et al (1999). A productive
changes. Biochemistry 36, 1259-1280. NADP+ binding mode of ferredoxin-NADP+
65. Shen, A.L., T.D. Porter, T.E. Wilson, and C.B. reductase revealed by protein engineering and crys-
Kasper (1989). Structural analysis of the FMN bind- tallographic studies. Nat. Struct. Biol 6, 847-853.
ing domain of NADPH-cytochrome P-450 oxidore- 76. Sem, D.S. and C.B. Kasper (1993). Interaction
ductase by site-directed mutagenesis. J. Biol. Chem. with arginine 597 of NADPH-cytochrome P-450
264, 7584-7589. oxidoreductase is a primary source of the uniform
66. Paine, M.J.I., S. Ayivor, A. Munro, P. Tsan, binding energy used to discriminate between NADPH
L.Y Lian, G.C.K. Roberts et al. (2001). Role and NADH. Biochemistry 32, 11548-11558.
142 Mark J.I. Paine ef al.

11. Sem, D.S. and C.B. Kasper (1993). Enzyme-substrate flavocytochrome P-450 BM3 and its component
binding interactions of NADPH-cytochrome P-450 domains. Eur J. Biochem. 239, 403^09.
oxidoreductase characterized with pH and aher- 91. Noble, M.A., A.W. Munro, S.L. Rivers, L.
nate substrate/inhibitor studies. Biochemistry 32, Robledo, S.N. Daff, L.J. Yellowlees et al. (1999).
11539-11547. Potentiometric analysis of the flavin cofactors of
78. Elmore, C.L. and T.D. Porter (2002). Modification neuronal nitric oxide synthase. Biochemistry 38,
of the nucleotide cofactor-binding site of cytochrome 16413-16418.
P-450 reductase to enhance turnover with NADH 92. Knight, K. and N.S. Scrutton (2002). Stopped-
in vivo. J. Biol. Chem. 277, 48960-48964. flow kinetic studies of electron transfer in the reduc-
79. Shen, A.L. and C.B. Kasper (1996). Role of Ser457 tase domain of neuronal nitric oxide synthase:
of NADPH-cytochrome P450 oxidoreductase in Reevaluation of the kinetic mechanism reveals new
catalysis and control of FAD oxidation-reduction enzyme intermediates and variation with cyto-
potential. Biochemistry 35, 9451-9459. chrome P450 reductase. Biochem. J. 367, 19-30.
80. Shen, A.L., D.S. Sem, and C.B. Kasper (1999). 93. McLean, K.J., N.S. Scrutton, and A.W. Munro
Mechanistic studies on the reductive half-reaction (2003). Kinetic, spectroscopic and thermodynamic
of NADPH-cytochrome P450 oxidoreductase. characterization of the Mycobacterium tuberculo-
J. Biol. Chem. 274, 5391-5398. sis adrenodoxin reductase homologue FprA.
81. Shen, A.L., D.S. Sem, and C.B. Kasper (1999). Biochem. J. :^12,?>\1-?>21.
Mechanistic studies on the reductive half-reaction 94. Noble, M.A., C.S. Miles, S.K. Chapman,
of NADPH-cytochrome P450 oxidoreductase. D.A. Lysek, A.C. Mackay, G.A. Reid et al. (1999).
J. Biol. Chem. 274, 5391-5398. Roles of key active site residues in flavocy-
82. Vermilion, J.L. and M.J. Coon (1978). Identification tochrome P450 BM3. Biochem. J. 339, 371-379.
of the high and low potential flavins of liver micro- 95. Gutierrez, A., A.W. Munro, A. Grunau, C.R. Wolf,
somal NADPH-C3^ochrome P-450 reductase. N.S. Scrutton, and G.C.K. Roberts (2003).
J. Biol. Chem. 253, 8812-8819. Interflavin electron transfer in human cytochrome
83. Vermilion, J.L. and M.J. Coon (1978). Purified liver P450 reductase is enhanced by coenzyme binding.
microsomal NADPH-cytochrome P-450 reductase. Eur. J. Biochem. 210, 2612-2621.
Spectral characterization of oxidation-reduction 96. Bhattacharyya, A.K., J.J. Lipka, L. Waskell, and G.
states. J. Biol. Chem. 253, 2694-2704. Tollin (1991). Laser flash photolysis studies of the
84. Yasukochi, Y., J.A. Peterson, and B.S. Masters reduction kinetics of NADPH.cytochrome P-450
(1979). NADPH-cytochrome c (P-450) reductase. reductase. Biochemistry 30, 759-765.
Spectrophotometric and stopped flow kinetic 97. Gutierrez, A., M. Paine, C.R. Wolf, N.S. Scrutton,
studies on the formation of reduced flavoprotein and G.C.K. Roberts (2002). Relaxation kinetics
intermediates. ^ 5/o/. Chem. 254, 7097-7104. of cytochrome P450 reductase: Internal electron
85. Griffith, O.W. and D.J. Stuehr (1995). Ann. Rev. transfer is limited by conformational change and
Physiol. 57, 707-736. regulated by coenzyme binding. Biochemistry 41,
86. Miles, J.S., A.W. Munro, B.N. Rospendowski, 4626-4637.
W.E. Smith, J. McKnight, and A.J. Thomson (1992). 98. Page, C.C, C.C. Moser, X. Chen, and PL. Dutton
Domains of the catalytically self-sufficient (1999). Natural engineering principles of elec-
cytochrome P-450 BM-3. Genetic construction, tron tunnelling in biological oxidation-reduction.
overexpression, purification and spectroscopic char- Nature 402, 47-52.
acterization. Biochem. J. 288, 503-509. 99. Bayburt, T.H., J.W Carlson, and S.G. Sligar
87. Gutierrez, A., L.Y. Lian, C.R. Wolf, N.S. Scrutton, (1998). Reconstitution and imaging of a membrane
and G.C.K. Roberts (2001). Stopped-flow kinetic protein in a nanometer-size phospholipid bilayer.
studies of flavin reduction in human cytochrome J. Struct. Biol. 123, 3 7 ^ 4 .
P450 reductase and its component domains. 100. Bayburt, TH. and S.G. Sligar (2002). Single-
Biochemistry ^{^, 1964-1975. molecule height measurements on microsomal
88. Munro, A.W., M.A. Noble, L. Robledo, S.N. Daff, cytochrome P450 in nanometer-scale phospholipid
and S.K. Chapman (2001). Determination of the bilayer disks. Proc. Natl. Acad. Sci. USA 99,
redox properties of human NADPH-cytochrome 6725-6730.
P450 reductase. Biochemistry 40, 1956-1963. 101. Ingelman-Sundberg, M., T Haaparanta, and
89. Daff, S.N., S.K. Chapman, K.L. Turner, R.A. Holt, J. Rydstrom (1981). Membrane charge as effector
S. Govindaraj, T.L. Poulos et al. (1997). Redox con- of cytochrome P-450LM2 catalyzed reactions
trol of the catalytic cycle of flavocytochrome P450 in reconstituted liposomes. Biochemistry 20,
BM3. Biochemistry 36, 13816-13823. 4100^106.
90. Munro, A.W., S. Daff, J.R. Coggins, J.G. Lindsay, and 102. Ingelman-Sundberg, M., J. Blanck, G. Smettan, and
S.K. Chapman (1996). Probing electron transfer in K. Ruckpaul (1983). Reduction of cyto-chrome
Electron Transfer Partners of Cytochrome P450 143

P-450 LM2 by NADPH in reconstituted phospho- between NADPH-cytochrome P450 reductase and
lipid vesicles is dependent on membrane charge. cytochrome P450 enzymes. J. Biol. Chem. 269,
Eur. J. Biochem. 134, 157-162. 15724-15731.
103. Coon, M.J., A.P. Autor, and H.W. Strobel (1971). 115. Voznesensky, A.I. and J.B. Schenkman (1992). The
Role of phospholipid in electron transfer in a cytochrome P450 2B4-NADPH cytochrome P450
reconstituted liver microsomal enzyme system reductase electron transfer complex is not formed by
containing cytochrome P-450. Chem. Biol. charge-pairing. J. Biol. Chem. 267, 14669-14676.
Interact. 3, 248-250. 116. Schenkman, J.B., A.I. Voznesensky, and I. Jansson
104. Estabrook, R.W., A.G. Hildebrandt, J. Baron, (1994). Influence of ionic strength on the P450
K.J. Netter, and K. Leibman (1971). A new spec- monooxygenase reaction and role of cytochrome
tral intermediate associated with cytochrome P450 b5 in the process. Arch. Biochem. Biophys. 314,
function in liver microsomes. Biochem. Biophys. 234-241.
Res. Commun. 42, 132-139. 117. Davydov, D.R., A.A. Kariakin, N.A. Petushkova,
105. Peterson, J.A., R.E. Ebel, D.H. O'Keeffe, and J.A. Peterson (2000). Association of cyto-
T. Matsubara, and R.W. Estabrook (1976). chromes P450 with their reductases: Opposite sign
Temperature dependence of cytochrome P-450 of the electrostatic interactions in P450BM-3 as
reduction. A model for NADPH-cytochrome compared with the microsomal 2B4 system.
P-450 reductase cytochrome P-450 interaction. Biochemistry 39, 6489-6497.
J. Biol. Chem. 251, 4010^016. 118. Munro, A.W and J.G. Lindsay (1996). Bacterial
106. Shimizu, T., T. Tateishi, M. Hatano, and cytochromes P-450. Mol. Microbiol. 20,1115-1125.
Y. Fujiikuriyama (1991). Probing the role of 119. Matson, R.S., R.S. Hare, and A.J. Fulco (1977).
lysines and arginines in the catalytic function of Characteristics of a cytochrome P-450-dependent
cytochrome-P450d by site-directed mutagenesis— fatty acid omega-2 hydroxylase from Bacillus
interaction with NADPH-cytochrome-P450 reduc- megaterium. Biochim. • Biophys. Acta. 487,
tase. J. Biol. Chem. 266, 3372-3375. 487^94.
107. Shen, S. and H.W. Strobel (1993). Role of lysine 120. Sueyoshi, T. and M. Negishi (2001). Phenobarbital
and arginine residues of cytochrome P450 in the response elements of cytochrome P450 genes and
interaction between cytochrome P4502B1 and nuclear receptors. Ann. Rev. Pharmacol. Toxicol.
NADPH-cytochrome P450 reductase. Arch. 41, 123-143.
Biochem. Biophys. 304, 257-265. 121. Shaw, G.C. and A.J. Fulco (1993). Inhibition by
108. Bridges, A., L. Gruenke, Y.T. Chang, LA. Vakser, barbiturates of the binding of Bm3Rl repressor
G. Loew, and L. Waskell (1998). Identification of to its operator site on the barbiturate-inducible
the binding site on cytochrome P450 2B4 for cytochrome P450BM-3 gene of Bacillus mega-
cytochrome b{5) and cj^ochrome P450 reductase. terium. J.Biol. Chem. 268, 2997-3004.
J. Biol. Chem. 213, 17036-17049. 122. Palmer, C.N., E. Axen, V Hughes, and C.R. Wolf
109. Strobel, H.W., S.G. Nadler, and D.R. Nelson (1989). (1998). The repressor protein, Bm3Rl, mediates
Cytochrome P-450: Cytochrome P450 reductase an adaptive response to toxic fatty acids in Bacillus
interactions. DrugMetab. Rev. 20, 519-533. megaterium. J. Biol. Chem. 273, 18109-18116.
110. Nadler, S.G. and H.W. Strobel (1991). Iden- 123. Fulco, A.J. (1991). P450BM-3 and other inducible
tification and characterization of an NADPH- bacterial P450 cytochromes: Biochemistry and
cytochrome P450 reductase derived peptide regulation. Ann. Rev. Pharmacol. Toxicol. 31,
involved in binding to cytochrome P450. Arch. 177-203.
Biochem. Biophys. 290, 277-284. 124. Narhi, L.O. and A.J. Fulco (1987). Identification
111. Shen, S. and H.W. Strobel (1994). Probing the puta- and characterization of two functional domains
tive cytochrome P450- and cytochrome c-binding in cytochrome P-450BM-3, a catalytically self-
sites on NADPH-cytochrome P450 reductase sufficient monooxygenase induced by barbiturates
by anti-peptide antibodies. Biochemistry 33, in Bacillus megaterium. J. Biol. Chem. 262,
8807-8812. 6683-6690.
112. Nisimoto, Y. (1986). Localization of cytochrome c- 125. Boddupalli, S.S., B.C. Pramanik, C.A. Slaughter,
binding domain on NADPH-cytochrome P-450 R.W. Estabrook, and J.A. Peterson (1992). Fatty
reductase. J. Biol. Chem. 261, 14232-14239. acid monooxygenation by P450BM-3: Product
113. Sevrioukova, I.F., H. Li, H. Zhang, J.A. Peterson, identification and proposed mechanisms for the
andT.L. Poulos (1999). Structure of a cytochrome sequential hydroxylation reactions. Arch. Biochem.
P450-redox partner electron-transfer complex. Biophys. 292, 20-28.
Proc. Natl. Acad. Sci. USA 96, 1863-1868. 126. Peterson, J.A. and S.S. Boddupalli (1992).
114. Voznesensky, A.I. and J.B. Schenkman (1994). P450BM-3: Reduction by NADPH and sodium
Quantitative analyses of electrostatic interactions dithionite. Arch. Biochem. Biophys. 294, 654-661.
144 Mark J.I. Paine et ai.

127. Li, H.Y., K. Darwish, and XL. Poulos (1991). recombinant fiision protein containing NADPH-
Characterization of recombinant Bacillus mega- P450 reductase. Proc. Natl. Acad. Sci. USA 90,
terium cytochrome P-450 BM-3 and its two func- 11748-11752.
tional domains. J. Biol. Chem. 266, 11909-11914. 139. Chun, Y.J., T. Shimada, and FP Guengerich
128. Govindaraj, S. andT.L. Poulos (1997). The domain (1996). Construction of a human cytochrome
architecture of cytochrome P450BM-3. J. Biol. P450 lAl: Rat NADPH-cytochrome P450 reduc-
Chem. 212,1915-7921. tase fusion protein cDNA and expression in
129. Noble, M.A., C.S. Miles, S.K. Chapman, D.A. Escherichia coli, purification, and catalytic prop-
Lysek, A.C. MacKay, G.A. Reid et al (1999). erties of the enzyme in bacterial cells and after
Roles of key active-site residues in flavocy- purification. Arch. Biochem. Biophys. 330, 48-58.
tochrome P450 BM3. Biochem. J. 339, 371-379. 140. Shet, M., C.W. Fisher, PL. Holmans, and R.W.
130. Shirane, N., Z. Sui, J.A. Peterson, and PR. Ortiz Estabrook (1996). The omega-hydroxlyation of lau-
de Montellano (1993). Cytochrome P450BM-3 ric acid: Oxidation of 12-hydroxylauric acid to
(CYP102): Regiospecificity of oxidation of omega- dodecanedioic acid by a purified recombinant fusion
unsaturated fatty acids and mechanism-based protein containing P450 4A1 and NADPH-P450
inactivation. Biochemistry 32, 13732-13741. reductase. Arch. Biochem. Biophys. 330, 199-208.
131. Ost, T.W., C.S. Miles, A.W. Munro, J. Murdoch, 141. Helvig, C. and J.H. Capdevila (2000). Biochemical
G.A. Reid, and S.K. Chapman (2001). Phenyl- characterization of rat P450 2C11 fused to rat or
alanine 393 exerts thermodynamic control over the bacterial NADPH-P450 reductase domains.
heme of flavocytochrome P450 BM3. Biochemistry Biochemistry 39, 5196-5205.
40,13421-13429. 142. Sakaki, T, S. Kominami, S. Takemori, H. Ohkawa,
132. Murataliev, M.B. and R. Feyereisen (2000). M. Akiyoshi-Shibata, and Y. Yabusaki (1994).
Functional interactions in cytochrome P450BM3. Kinetic studies on a genetically engineered fused
Evidence that NADP(H) binding controls redox enzyme between rat cytochrome P4501A1 and
potentials of the flavin cofactors. Biochemistry 39, yeast NADPH-P450 reductase. Biochemistry 33,
12699-12707. 4933-4939.
133. Murataliev, M.B. and R. Feyereisen (1999). 143. Hayashi, K., T. Sakaki, S. Kominami, K. Inouye,
Mechanism of cytochrome P450 reductase from and Y. Yabusaki (2000). Coexpression of geneti-
the house fly: Evidence for an FMN semiquinone cally engineered fused enzyme between yeast
as electron donor. FEBS Lett. 453, 201-204. NADPH-P450 reductase and human cytochrome
134. Murataliev, M.B. and R. Feyereisen (2000). P450 3A4 and human cytochrome b5 in yeast.
Interaction of NADP(H) with oxidized and reduced Arch. Biochem. Biophys. 381, 164-170.
P450 reductase during catalysis. Studies with 144. Deeni, YY, M.J. Paine, A.D. Ayrton, S.E. Clarke,
nucleotide analogues. Biochemistry 39, 5066-5074. R. Chenery, and C.R. Wolf (2001). Expression,
135. Munro, A.W., K. Malarkey, J. McKnight, A.J. purification, and biochemical characterization
Thomson, S.M. Kelly, N.C. Price et al. (1994). The of a human cytochrome P450 CYP2D6-NADPH
role of tryptophan 97 of cytochrome P450 BM3 cytochrome P450 reductase fusion protein. Arch.
from Bacillus megaterium in catalytic function. Biochem. Biophys. 396, 16-24.
Evidence against the 'covalent switching' hypothesis 145. Sanborn, R.C. and CM. William (1950). The
of P-450 electron transfer. Biochem. J. 303,423-428. cytochrome system in the Cecropia silkworm, with
136. Murakami, H., Y. Yabusaki, T. Sakaki, M. Shibata, special reference to the properties of a new com-
and H. Ohkawa (1987). A genetically engineered ponent. J. Gen. Physiol. 33, 579-588.
P450 monooxygenase: Construction of the func- 146. Porter, T.D. (2002). The roles of cytochrome b5 in
tional fused enzyme between rat cytochrome cytochrome P450 reactions. J. Biochem. Mol.
P450c and NADPH-cytochrome P450 reductase. Toxicol. 16,311-316.
DMA 6, 189-197. 147. Vergeres, G. and L. Waskell (1995). Cytochrome
137. Fisher, C.W., M.S. Shet, D.L. Caudle, C.A. Martin- b5, its functions, structure and membrane topol-
Wixtrom, and R.W. Estabrook (1992). High-level ogy. Biochimie 11, 604-620.
expression in Escherichia coli of enzymatically 148. Hildebrandt, A. and R.W. Estabrook (1971).
active fusion proteins containing the domains of Evidence for the participation of cytochrome b5
mammalian cytochromes P450 and NADPH-P450 in hepatic microsomal mixed-function oxidation
reductase flavoprotein. Proc. Natl. Acad. Sci. USA reactions. Arch. Biochem. Biophys. 143, 66-79.
89, 10817-10821. 149. Correia, M.A. and G.J. Mannering (1973).
138. Shet, M.S., C.W. Fisher, PL. Holmans, and Reduced diphosphopyridine nucleotide synergism
R.W. Estabrook (1993). Human cytochrome of the reduced triphosphopyridine nucleotide-
P450 3A4: Enzymatic properties of a purified dependent mixed-function oxidase system of
Electron Transfer Partners of Cytochrome P450 145

hepatic microsomes. II. Role of the type I for P450 residues Lys-122 and Arg-125. Biochem.
drug-binding site of cytochrome P-450. Mol. Biophys. Res. Commun. 201, 1090-1095.
Pharmacol. 9, 470-485. 162. Omata, Y, R.C. Robinson, H.V Gelboin, M.R.
150. Yamazaki, H., M. Nakamura, T. Komatsu, Pincus, and F.K. Friedman (1994). Specificity of
K. Ohyama, N. Hatanaka, S. Asahi et al. (2002). the cytochrome P-450 interaction with cytochrome
Roles of NADPH-P450 reductase and apo- and b5. FEBSLett. 346, 241-245.
holo-cytochrome b5 on xenobiotic oxidations cat- 163. Guengerich, F.P. (1983). Oxidation-reduction
alyzed by 12 recombinant human cytochrome properties of rat liver cytochromes P-450 and
P450s expressed in membranes of Escherichia NADPH-cytochrome p-450 reductase related to
coli. Protein Expr Purif. 24, 329-337. catalysis in reconstituted systems. Biochemistry
151. Voice, M.W., Y. Zhang, C.R. Wolf, B. Burchell, and 22,2811-2820.
T. Friedberg (1999). Effects of human cytochrome 164. Yamazaki, H., W.W. Johnson, Y.-F. Ueng, T
b5 on CYP3A4 activity and stability in vivo. Arch. Shimada, and F.P. Guengerich (1996). Lack of
Biochem. Biophys. 366, 116-124. electron transfer from cytochrome b5 in stimula-
152. Hlavica, P and D.F. Lewis (2001). Allosteric phe- tion of catalytic activities of cytochrome P450
nomena in cytochrome P450-catalyzed monooxy- 3A4. Characterization of a reconstituted cyto-
genations. Eur J. BioChem. 268, 4817-^832. chrome P450 3A4/NADPH-cytochrome P450
153. Mitoma, J. and A. Ito (1992). The carboxy-termi- reductase system and studies with apo-cytochrome
nal 10 amino acid residues of cytochrome b5 are b5. J. Biol. Chem. 271, 27438-27444.
necessary for its targeting to the endoplasmic retic- 165. Yamazaki, H., T. Shimada, M.V Martin, and F.P
ulum. EMBOJ. 11, 4197-4203. Guengerich (2001). Stimulation of cytochrome
154. Keyes, S.R. and D.L. Cinti (1980). Biochemical P450 reactions by apocytochrome b5. Evidence
properties of cytochrome b5-dependent microso- against transfer of heme from cytochrome P450
mal fatty acid elongation and identification of 3A4 to apo-cytochrome b5 or heme oxygenase.
products. ^ 5/o/. Chem. 255, 11357-11364. J. Biol. Chem. 276, 30885-30891.
155. Oshino, N., Y. Imai, and R. Sato (1971). A function 166. Yamazaki, H., M. Nakano, Y Imai, Y.F. Ueng,
of cytochrome b5 in fatty acid desaturation F.P. Guengerich, and T. Shimada (1996). Roles of
by rat liver microsomes. J. Biochem. (Tokyo) 69, cytochrome b5 in the oxidation of testosterone and
155-167. nifedipine by recombinant cytochrome P450 3A4
156. Reddy, VV, D. Kupfer, and E. Caspi (1977). and by human liver microsomes. Arch. Biochem.
Mechanism of C-5 double bond introduction in the Biophys. 325, 174-182.
biosynthesis of cholesterol by rat liver micro- 167. Bell, L.C. and EP Guengerich (1997). Oxidation
somes. J. Biol. Chem. 252, 2797-2801. kinetics of ethanol by human cytochrome P450
157. Hegesh, E., J. Hegesh, and A. Kaftory (1986). 2E1. Rate-limiting product release accounts for
Congenital methemoglobinemia with a deficiency effects of isotopic hydrogen substitution and
of cytochrome b5. N. Engl. J. Med. 314, 757-761. cytochrome b5 on steady-state kinetics. J. Biol.
158. Manabe, J., R. Arya, H. Sumimoto, T. Yubisui, A.J. Chem. 272,29643-29651.
Bellingham, D.M. Layton et al. (1996). Two novel 168. Schenkman, J.B., A.I. Voznesensky, and I. Jansson
mutations in the reduced nicotinamide adenine (1994). Influence of ionic strength on the P450
dinucleotide (NADH)-cytochrome b5 reductase monooxygenase reaction and role of cytochrome
gene of a patient with generalized type, hereditary b5 in the process. Arch. Biochem. Biophys. 314,
methemoglobinemia. Blood SS, 3208-3215. 234-241.
159. Tamburini, P.P., S. MacFarquhar, and J.B. 169. Perret, A. and D. Pompon (1998). Electron shuttle
Schenkman (1986). Evidence of binary complex between membrane-bound cytochrome P450 3A4
formations between cytochrome P-450, cyto- and b5 rules uncoupling mechanisms. Biochemistry
chrome b5, and NADPH-cytochrome P-450 reduc- 37, 11412-11424.
tase of hepatic microsomes. Biochem. Biophys. 170. Gruenke, L.D., K. Konopka, M. Cadieu, and
Res. Commun. 134, 519-526. L. Waskell (1995). The stoichiometry of the
160. Stayton, PS. and S.G. Sligar (1990). The cytochrome P-450-catalyzed metabolism of
cytochrome P-450cam binding surface as defined methoxyflurane and benzphetamine in the presence
by site-directed mutagenesis and electrostatic and absence of cytochrome b5. J. Biol. Chem. 270,
modeling. Biochemistry 29, 7381-7386. 24707-24718.
161. Omata, Y, H. Sakamoto, R.C. Robinson, M.R. 171. Wang, M.H., C.J. Patten, G.Y Yang, S.R.
Pincus, and F.K. Friedman (1994). Interaction Paranawithana, Y Tan, and C.S. Yang (1996).
between cytochrome P450 2B1 and cytochrome Expression and coupling of human cytochrome
b5: Inhibition by synthetic peptides indicates a role P450 2E1 and NADPH-cytochrome P450
146 Mark J.I. Paine et al.

oxidoreductase in dual expression and co-infection 184. Hanukoglu, I., C.T. Privalle, and C.R. Jefcoate
systems with baculovims in insect cells. Arch. (1981). Mechanisms of ionic activation of adrenal
Biochem. Biophys. 334, 380-388. mitochondrial cytochromes P-450scc and P-45011
172. Cooper, M.T. andT.D. Porter (2001). Cytochrome beta. J. Biol. Chem. 256, 4329^335.
b(5) coexpression increases the CYP2E1-depend- 185. Ziegler, G.A., C. Vonrhein, I. Hanukoglu, and G.E.
ent mutagenicity of dialkylnitrosamines in methyl- Schulz (1999). The structure of adrenodoxin
transferase-deficient strains of Salmonella reductase of mitochondrial P450 systems: Electron
typhimurium. Mutat. Res. 484, 61-68. transfer for steroid biosynthesis. J. Mol. Biol. 289,
173. Jansson, I. and J.B. Schenkman (1987). Influence 981-990.
of cytochrome b5 on the stoichiometry of the dif- 186. Bossi, R.T., A. Aliverti, D. Raimondi, F Fischer,
ferent oxidative reactions catalyzed by liver micro- G. Zanetti, D. Ferrari et al. (2002). A covalent
somal cytochrome P-450. Drug Metab. Dispos. 15, modification of NADP^ revealed by the atomic
344-348. resolution structure of FprA, a Mycobacterium
174. Grinberg, A.V, F. Hannemann, B. Schiffler, J. tuberculosis oxidoreductase, Biochemistry 41,
Muller, U. Heinemann, and R. Bernhardt (2000). 8807-8818.
Adrenodoxin: Structure, stability, and electron 187. Hanukoglu, I. (1992). Steroidogenic enzymes:
transfer properties. Proteins 40, 590-612. Structure, function, and role in regulation of
175. Poulos, T.L., B.C. Finzel, and A.J. Howard (1987). steroid hormone biosynthesis. J. Steroid. Biochem.
High-resolution crystal structure of cytochrome Mol. Biol. 43, 779-804.
P450cam. J. Mol Biol. 195, 687-700. 188. Pochapsky, T.C., N.U. Jain, M. Kuti, T.A. Lyons,
176. Mathews, FS., A.G. Mauk, and G.R. Moore and J. Heymont (1999). A refined model for the
(2000). In C. Kleanthous (ed.), Protein-Protein solution structure of oxidized putidaredoxin.
Recognition. Oxford University Press, Oxford, Biochemistry 38, 4681-4690.
pp. 60-101. 189. Fukuyama, K., T. Okada, Y. Kakuta, and Y.
177. Shimada, H., S. Nagano, H. Hori, and Y. Ishimura Takahashi (2002). Atomic resolution structures of
(2001). Putidaredoxin-cytochrome P450cam inter- oxidized [4Fe-4S] ferredoxin from Bacillus
action. J. Inorg. Biochem. 83, 255-260. thermoproteolyticus in two crystal forms:
178. Trower, M.K., R. Lenstra, C. Omer, S.E. Buchholz, Systematic distortion of [4Fe-4S] cluster in the
and FS. Sariaslani (1992). Cloning, nucleotide protein. J. Mol. Biol. 315, 1155-1166.
sequence determination and expression of the 190. Holden, M., M. Mayhew, D. Bunk, A. Roitberg,
genes encoding cytochrome P-450soy (soyC) and and V Vilker (1997). Probing the interactions of
ferredoxinsoy (soyB) from Streptomyces griseus. putidaredoxin with redox partners in camphor
Mol. Microbiol. 6, 2125-2134. P450 5-monooxygenase by mutagenesis of surface
179. Green, A.J., A.W. Munro, M.R. Cheesman, residues. J 5/o/. Chem. 272, 21720-21725.
G.A. Reid, C. von Wachenfeldt, and S.K. Chapman 191. Unno, M., J.F Christian, T. Sjodin, D.E. Benson,
(2003). Expression, purification and characterisa- I.D. Macdonald, S.G. SMgdiV etal. (2002). Complex
tion of a Bacillus subtilis ferredoxin: A potential formation of cytochrome P450cam with
electron transfer donor to cytochrome P450 Biol. Putidaredoxin. Evidence for protein-specific
J. Inorg. Biochem. 93, 92-99. interactions involving the proximal thiolate ligand.
180. Matocha, M.F and M.R. Waterman (1984). J. Biol. Chem. Ill, 2547-2553.
Discriminatory processing of the precursor forms 192. Roitberg, A., H.M. Holden, M.P Mayhew, I.V
of cytochrome P-450scc and adrenodoxin by Kurnikov, D.N. Beraten, and V.L. Vilker (1998).
adrenocortical and heart mitochondria. J. Biol. Binding and electron transfer between putidare-
Chem. 259, 8672-8678. doxin and cytochrome P450cam. J. Am. Chem.
181. Matsubara, H. and K. Saeki (1992). Structural and Soc. 120, 8927-8932.
functional diversity of ferredoxins and related pro- 193. Davies, M.D. and S.G. Sligar (1992). Genetic vari-
teins. Adv. Inorg Chem. 38, 223-280. ants in the putidaredoxin-cytochrome P-450cam
182. Sevrioukova, I.F and T.L. Poulos (2002). electron-transfer complex: Identification of the
Putidaredoxin reductase, a new function for an old residue responsible for redox-state-dependent
protein. J. Biol. Chem. Ill, 25831-25839. conformers. Biochemistry 31, 11383-11389.
183. Omura, T., E. Sanders, R.W. Estabrook, D.Y. 194. Stayton, PS. and S.G. Sligar (1990). The
Cooper, and O. Rosenthal (1966). Isolation from cytochrome P-450cam binding surface as defined
adrenal cortex of a nonheme iron protein and a by site-directed mutagenesis and electrostatic
flavoprotein functional as a reduced triphosphopy- modeling. Biochemistry 29, 7381-7386.
ridine nucleotide-cytochrome P-450 reductase. 195. Aoki, M., K. Ishimori, H. Fukada, K. Takahashi,
Arch. Biochem. Biophys. Ill, 660-673. and I. Morishima (1998). Isothermal titration
Electron Transfer Partners of Cytochrome P450 147

calorimetric studies on the associations of puti- cytochrome P450(scc) (CYPllAl). J. Inorg.


daredoxin to NADH-putidaredoxin reductase and Biochem. 91, 644-654.
P450cam. Biochim. Biophys. Acta. 1384, 180-188. 208. Uhlmann, H., V Beckert, D. Schwarz, and
196. Mueller, E.J., P.J. Loida, and S.G. Sligar (1995). In R. Bernhardt (1992). Expression of bovine adren-
P.R. Ortiz de Montellano (ed.), Cytochrome P450; odoxin in E. coli and site-directed mutagenesis of
Structure, Mechanism and Biochemistry (2nd edn.). /2 Fe-2S/ cluster ligands. Biochem. Biophys. Res.
Plenum Press, New York, pp. 83-124. Commun. 188, 1131-1138.
197. Hintz, M.J. and J.A. Peterson (1981). The kinetics 209. Hara, T, C. Koba, M. Takeshima, and Y. Sagara
of reduction of cytochrome P-450cam by reduced (2000). Evidence for the cluster model of mito-
putidaredoxin. J. Biol. Chem. 256, 6721-6728. chondrial steroid hydroxylase system derived from
198. Roome, PW., J.C. Philley, and J.A. Peterson dissociation constants of the complex between
(1983). Purification and properties of putidare- adrenodoxin reductase and adrenodoxin. Biochem.
doxin reductase. J. Biol. Chem. 49, 10592-10600. Biophys. Res. Commun. 276, 210-215.
199. Sevrioukova, I.F., J.T. Hazzard, G. Tollin, and T.L. 210. Lambeth, J.D., D.W Seybert, J.R. Lancaster, Jr.,
Poulos (2001). Laser flash induced electron trans- J.C. Salerno, and H. Kamin (1982). Steroidogenic
fer in P450cam monooxygenase putidaredoxin electron transport in adrenal cortex mitochondria.
reductase-putidredoxin interaction. Biochemistry Mol. Cell Biochem. 45, 13-31.
49, 10592-10600. 211. Muller, J.J., A. Lapko, K. Ruckpaul, and
200. Pochapsky, S.S., T.C. Pochapsky, and J.W. Wei U. Heinemann (2003). Modeling of electrostatic
(2003). A model for effector activity in a highly recognition processes in the mammalian mito-
specific biological electron transfer complex: The chondrial steroid hydroxylase system. Biophys.
cytochrome P450(cam)-putidaredoxin couple. Chem. 100, 281-292.
Biochemistry 42, 5649-5656. 212. Muller, J.J., A. Lapko, G. Bourenkov, K. Ruckpaul,
201. Tosha, T, S. Yoshioka, S. Takahashi, K. Ishimori, and U. Heinemann (2001). Adrenodoxin reduc-
H. Shimada, and I. Morishima (2003). NMR study tase-adrenodoxin complex structure suggests elec-
on the structural changes of cytochrome P450cam tron transfer path in steroid biosynthesis. J. Biol.
upon the complex formation with putidaredoxin: Chem. 276, 2786-2789.
Functional significance of the putidaredoxin- 213. Aoki, M., K. Ishimori, and I. Morishima (1998).
induced structural changes. J. Biol. Chem. 2, 2. Roles of negatively charged surface residues of
202. Brewer, C.B. and J.A. Peterson (1988) Single putidaredoxin in interactions with redox partners
turnover kinetics of the reaction between oxycy- in p450cam monooxygenase system. Biochim.
tochrome P450cam and reduced putidaredoxin. J. Biophys. Acta. 1386, 157-167.
Biol. Chem. 263, 791-798. 214. Sibbesen, O., J.J. De Voss, and PR. Ortiz de
203. Muller, A., J.J. Muller, Y.A. Muller, H. Uhlmann, Montellano (1996). Putidaredoxin reductase-
R. Bernhardt, and U. Heinemann (1998). New putidaredoxin-cytochrome P450cam triple fusion
aspects of electron transfer revealed by the crystal protein. Construction of a self-suff'icient Escheri-
structure of a truncated bovine adrenodoxin, chia coli catalytic system. J. Biol. Chem. 211,
Adx(4-108). Structure 6, 269-280. 22462-22469.
204. Pikuleva, I.A., K. Tesh, M.R. Waterman, and Y. 215. Song, W C , CD. Funk, and A.R Brash (1993).
Kim (2000). The tertiary structure of fiill-length Molecular cloning of an allene oxide synthase: A
bovine adrenodoxin suggests functional dimers. cytochrome P450 specialized for the metabolism
Arch. Biochem. Biophys. 373, 44-55. of fatty acid hydroperoxides. Proc. Natl. Acad. Sci.
205. Beilke, D., R. Weiss, F. Lohr, P Pristovsek, E t/&4 90, 8519-8523.
Hannemann, R. Bernhardt et al. (2002). A new 216. Lee, D.S., A. Yamada, H. Sugimoto, I. Matsunaga,
electron transport mechanism in mitochondrial H. Ogura, K. Ichihara et al. (2003). Substrate
steroid hydroxylase systems based on structural recognition and molecular mechanism of fatty
changes upon the reduction of adrenodoxin. acid hydroxylation by cytochrome P450 from
Biochemistry 41, 7969-7978. Bacillus subtilis. Crystallographic, spectroscopic,
206. Vickery, L.E. (1997). Molecular recognition and and mutational studies. J. Biol. Chem. 278,
electron transfer in mitochondrial steroid hydroxy- 9761-9767.
lase systems. Steroids 62, 124-127. 217. Green, A.J., A.W Munro, M.R Cheesman, G.A.
207. ZoUner, A., E Hannemann, M. Lisurek, and Reid, C. von Wachenfeldt, and S.K. Chapman
R. Bernhardt (2002). Deletions in the loop sur- (2003). Expression, purification and characterisa-
rounding the iron-sulfiir cluster of adrenodoxin tion of a Bacillus subtilis ferredoxin: A potential
severely affect the interactions with its native electron transfer donor to cytochrome P450 Biol.
redox partners adrenodoxin reductase and J. Inorg. Biochem. 93, 92-99.
148 Mark J.I. Paine et al.

218. Hawkes, D.B., G.W. Adams, A.L. Burlingame, 220. Jackson, C.J., D.C. Lamb, T.H. Marczylo, A.G.
P.R. Ortiz de Montellano, and J.J. De Voss (2002). Warrilow, N.J. Manning, D.J. Lowe et al. (2002).
Cytochrome P450(cin) (CYP176A), isolation, A novel sterol 14alpha-demethylase/ferredoxin
expression, and characterization. J. Biol. Chem. fusion protein (MCCYP51FX) from Methylo-
277, 27725-27732. coccus capsulatus represents a new class of the
219. Mclver, L., C. Leadbeater, D.J. Campopiano, R.L. cytochrome P450 superfamily. J. Biol. Chem. 277,
Baxter, S.N. Daff, S.K. Chapman et al. (1998). 46959^6965.
Characterisation of flavodoxin NADP^ oxido- 221. Roberts, G.A., G. Grogan, A. Greter, S.L. Flitsch,
reductase and flavodoxin; key components of and N.J. Turner (2002). Identification of a new
electron transfer in Escherichia coli. Eur. J. class of cytochrome P450 from a Rhodococcus sp.
Biochem. 257, 577-585. J. Bacteriol. 184, 3898-3908.
5
Activation of Molecular Oxygen by
Cytochrome P450
Thomas M. Makris, Ilia Denisov, lime Schlichting, and
Stephen G. Sligar

Life depends on the kinetic barriers to oxygen reactions


RJ.R Williams, in the preface to the Oxygen Chemistry by
Donald T. Sawyer^

1. Introduction to Oxygen the importance of oxygen as a cosubstrate, and the


Activation thoughts of overall common mechanisms with the
heme monooxygenases and other systems that
effect reductive metabolism of atmospheric dioxy-
The cytochromes P450 have occupied a central gen^' ^. Thus, from the very beginning, the phrase
focal point in biochemistry, pharmacology, toxicol- "active oxygen" was used to label a holy grail of
ogy, and biophysics for more than three decades. cytochrome P450 research: the documentation
This rich history is faithfully conveyed in accom- of the intermediate states of oxygen and heme
panying articles appearing in this volume as well as iron that give rise to the diversity of metabolic
those of the first and second editions of Cytochrome chemistries observed.
P450 edited by Ortiz de Montellano^' ^. In addition,
The chemistry of "oxygen activation" cat-
there have been innumerable review articles and
alyzed by a variety of metal centers has a long and
book chapters devoted to the subject^' ^. Interest in
rich history, and as many as eight variations of
the cytochromes P450 perhaps began due to the
structurally different metal-oxygen complexes
importance of these catalysts in human physiology
were discussed as early as 1965^. This publication
and pharmacology. The interest of chemists and
of Cytochrome P450, Third Edition is particularly
biophysicists peaked due to the many unique spec-
timely. During the past 3-5 years, a great deal of
tral features of these heme enzymes. One of the
progress has been realized in the direct observa-
most intriguing aspects of P450 systems is the
tion of P450 intermediate states and the evolution
range of difficult chemistry efficiently catalyzed.
of a detailed understanding of the structural

Thomas M. Makris • Center for Biophysics, University of Illinois, Urbana, IL.


Ilia Denisov • Department of Biochemistry, University of Illinois, Urbana, IL.
lime Schlichting • Max Planck Institute for Medical Research, Department of Molecular Mechanisms, Germany.
Stephen G. Sligar • Departments of Biochemistry, Chemistry, the School of Medicine, and the Center for
Biophysics, University of Illinois, Urbana, IL.
Cytochrome P450: Structure, Mechanism, and Biochemistry, 3e, edited by Paul R. Ortiz de Montellano
Kluwer Academic / Plenum Publishers, New York, 2005.

149
150 Thomas M. Makris ef al.

features of the protein, which control the popula- P450 monooxygenase catalysis that was available
tion and stepwise movement through the reduction at the time of the previous edition of this volume.
and protonation states of heme and oxygen. The In discussing the reaction cycle of the
veil surrounding "oxygen activation" has been cytochrome P450s, it is useful to incorporate
lifted through the systematic application of a vari- similar states which appear to be more or less
ety of biochemical and biophysical tools, such as stable in other heme-oxygen systems. The sequen-
mutagenesis, high-resolution cryocrystallography, tial steps of "oxygen activation" are those that
multiparameter spectroscopic studies of interme- follow the formation of the ferrous reduced iron-
diate states isolated using cryogenic or fast kinetic porphyrin state that is generated by electron
techniques, and many rigorous quantum chemical transfer from the physiological redox donor. These
and molecular dynamics computational studies. steps are formally:
Equally important has been the explosion of
genomic and proteomic breakthroughs. No longer 1. Dioxygen binding to the ferrous heme
are we dealing with a single microbial P450 and a center to form an oxygenated complex which can
few membrane bound mammalian counterparts. be written as either Fe^^-OO or Fe^^-00~.
Rather, with over 3,000 P450 genes known, we 2. One-electron reduction of this complex
can find examples from unique environmental to form the Fe^^-00^~, ferric-peroxo complex.
niches where the enzymatic reaction cycle yields 3. Protonation of the distal oxygen of
newly observable features. Thus today, the current this peroxo anion to generate the Fe^^-OOH~,
view of oxygen activation mechanisms catalyzed ferric-hydroperoxo complex.
by metal centers in heme enzymes ensures a much 4. A second protonation of the Fe^^-OOH~
better opportunity to develop recurring catalytic complex at the distal oxygen atom to form a
paradigms than was possible at the time of the transient oxo-water adduct which immediately
Cytochrome P450, Second Edition. undergoes heterolytic scission of the 0 - 0 bond.
A broader view of heme-oxygen catalysis with This process generates a single oxygen atom con-
the understanding of how proximal ligand, protein taining iron-porphyrin species, which is formally
structure, and subtle control of electron/proton two redox equivalents above the ferric heme start-
stoichiometry contribute to very different reactiv- ing state. This intermediate has been variously
ities is now within reach^. Common mechanistic termed the "iron-oxo," "ferryl-oxo porphyrin
hypotheses for oxygen activation in metalloen- TT-cation radical," or "Compound I," the latter after
zymes are currently emerging. Recent progress in the pioneering discoveries made by Theorell,
mechanistic studies on other heme enzymes, Keilin, George, and Chance while studying the
which use different forms of so-called "active peroxidases and catalases'^.
oxygen intermediates," such as peroxidases'^' •',
heme oxygenases (HOs)'^' '^, catalases'"^, nitric The intermediate states so defined have coun-
oxide synthases (NOS)'^, and peroxygenases'^' '^, terpoints in nonheme oxygen activation and have
have provided an impressive degree of unifica- been also studied and reviewed in numerous arti-
tion. A fundamental question that has always cles over past decades. We note that a search of the
plagued the interpretation of experimental data term "oxygen activation" in SciFinder Chemical
has been the functional differentiation of these Abstracts Database returns almost 29,000 refer-
enzymes, which efficiently catalyze a range of ences published after 1995! Thus, we will not
diverse chemistries yet utilize similar highly reac- attempt to link nonheme iron chemistry, nor
tive heme-oxygen complexes. The comparison of oxygen activation in multicenter metalloenzymes
similar reactive intermediates in different enzymes which was recently reviewed by Siegbahn'^, with
has helped to distinguish between the essential fea- that of the P450 systems in this chapter. Rather,
tures of each of the enzymes. This in turn has pro- we focus on those features which directly address
vided important clues into the role of protein the important aspects of the oxygen activation
structure in the heme-oxygen catalytic events. The cascade in P450. The structure and properties of
recent progress in actually observing, through relevant porphyrin complexes, including models
kinetic isolation and cryogenic techniques, these of cytochrome P450 and other oxygen-activation
fundamental intermediate states in the cytochrome enzymes, are extensively reviewed in the recent
P450s brings us to a much deeper understanding of edition of the Porphyrin Handbook^^'^^.
Activation of IVIolecular Oxygen by Cytochrome P450 151

2. General Features of Dioxygen overall process of O-O bond cleavage^^. The rela-
Activation in Heme Enzymes tively easy oxidation of Fe-protoporphyrin IX,
which serves as an additional buffer supply of
electrons to the dioxygen ligand, is a common
As discussed in the first section of this chapter,
feature in the mechanisms of related heme enzyme
the P450 cytochromes, NOS, HOs, and cyto-
families, such as peroxidases, heme catalases,
chrome oxidases are all heme enzymes which are
cytochromes P450, NOS, and peroxygenases,
able to "activate" molecular oxygen. Related
which catalyze 0 - 0 bond cleavage of a peroxide
heme enzymes, such as the peroxidases and cata-
ligated to the heme iron^^' ^K Donation of one elec-
lases, work with partially reduced dioxygen
tron from the porphyrin to the peroxide ligand ini-
species. It has always been intriguing that each
tiates the effective heterol3^ic scission of the O-O
of these enzymes uses the same cofactor, heme
bond and results in formation of a cation radical
or Fe-protoporphyrin IX, but performs a distinct
on the porphyrin ring, a definitive feature of a
physiological function.
"Compound I" state^^
In heme enzymes that are able to activate
Oxygen activation in all of these systems
molecular dioxygen, the porphyrin and proximal
begins with the binding of dioxygen as an axial
ligand provide the heme iron with five coor-
ligand to the Fe^+ heme iron, or with binding
dination sites. Thus, the side-on coordination of
of H2O2 to the Fe^^ heme. Thereby hydroperoxo-
peroxide, which is common for nonheme metal-
ferric heme complex is formed through different
loenzymes and oxygen activation mechanisms, is
pathways as a common intermediate state, shown
not usually observed in heme enzymes and is also
in Figure 5.1. Thus, a current focus of research
rare in model metalloporphyrins^^' ^^~^^.
is this common "peroxo," both in the pathways
A common role of the porphyrin ring in oxygen for its formation as well as in the control of
activation is the donation of an electron during the

P450 Monoxygenase ©
Oxidase ©
©
Compound ft
Heme Oxygenase ® -
O
Peroxygenase ©
Peroxfdase/Catalase © -MD / Fe^v/
N f-~n
Etectrophiiic QI
Nucleophiiic Nucleophlfic
Oxidations Oxidations
N-AH2

ferrous dioxygen peroxo-mmon ferric hydroperoxo Compound i


02^J o
N N
\^ N4J N Radical
/ Fei"/L. ^' »X Fell'/—^ Oxidations
N f-N \ N h-N
H,0 tl

© © ®
2e . 2HU
H9O,
rvH:>o

/ Fe^ V ^
N f-N

©
Figure 5.1. Common reactive intermediates in bioinorganic catalysis. The catalytic pathways of various heme
enzymes are shown in the inset.
152 Thomas M. Makris et al.

proton delivery events, which dictate the inherent Thus, the relative spatial orientation of catalytic
reactivity of the resulting "active oxygen" inter- residues and active-site water molecules may
mediates. In the classic P450 reactivities of a facilitate or suppress various branches of the
"Compound I like" intermediate, it is also this microscopic pathways that contribute to the possi-
critical acid-base chemistry which initiates dioxy- ble chemical reactions involving these activated
gen bond scission to form the higher valent oxygen compounds. An encompassing scheme
iron-oxo state. that graphically illustrates the commonality of
In addition, there are other redox active these various processes is depicted in Figure 5.1.
pathways operant in the cytochromes P450 Many laboratories have focused on the role of
(Figure 5.1). First is an "oxidase" pathway using protein structure in defining the pathways and
atmospheric dioxygen and two additional redox control points outlined in Figure 5.1. Recent
equivalents in addition to the two required for results, which are discussed in this chapter, have
heterolytic 0 - 0 bond cleavage. Additionally there greatly improved the understanding of these
is a "peroxidase/peroxygenase" pathway wherein events. For example, a hydrogen-bond network
hydrogen peroxide serves as an oxygen donor. has been implicated as the critical component of
A detailed analysis of these pathways with respect the enzymatic mechanism which defines both cat-
to oxygen activation in the cytochromes P450 alytic activity and the effective branching points
was provided in earlier editions of this volume^' ^. between productive and nonproductive pathways
Obviously, these two mechanisms differ by the for the camphor hydroxylation catalyzed by
overall redox state of oxygen vs peroxide and the cytochrome P450 CYPIOP^-^^ Modification of
need in two additional reduction steps (one- this network, either through mutagenesis of the
electron reductions by the physiological redox CYPlOl enzyme, or by the use of substrate
partner) in the oxidase pathway. Perhaps most analogs, points to the subtle active-site features of
important, however, is the difference in the proto- the protein, which can in some cases have dire
nation states that necessitate specific delivery of consequences on the productive monooxygenase
two protons to a bound peroxo intermediate when pathway with resultant abortive processes that
beginning with dioxygen as opposed to a 1-2 short circuit the classic P450 mechanistic "wheel"
proton shift to form the transient Fe-0-0H2 pre- (Figure 5.2). These additional pathways, since
cursor when starting with hydrogen peroxide and they bleed redox equivalents and dioxygen from
ferric heme. the pure monooxygenase stoichiometry, have been
termed "uncoupling" events. While the mecha-
2.1. The Oxidase/Oxygenase nisms of uncoupling may have either a thermody-
namic and/or kinetic origin, it is most certainly
Pathway in Cytochrome
governed by the role of a protein-provided "proton
P450 relay" system. These pathways of proton delivery
The pivotal role of different protonation path- can involve not only backbone and side-chain
ways to bound oxygen derivatives in bioinorganic moieties from the protein structure, but also local-
chemistry has been shown in various mechanistic ized water molecules, which are appropriately
studies ^^' ^^' ^^. The same concept has also been stabilized in the active center of the enzyme.
considered for the case of catalysis involving The realization of how Nature uses specialized
H2O2, in which the correlation between the ability water alludes to analogous discussion of proton
to efficiently deliver the proton to the metal- delivery in diverse enzymes such as cytochrome
bound hydroperoxide anion, and the efficiency of oxidase"^^' ^^, and introduces these concepts into
catalytic decomposition of hydrogen peroxide has the vernacular of P450 specialists. Again, geno-
been noted^'*. In heme-enzyme catalysis, however, mics and proteomics have helped identify critical
the ability to supply the necessary one or two structural features, for instance the famous "acid-
protons for the generation of the proper active alcohol pair," Asp251-Thr252 in the cytochrome
intermediate from the iron-bound peroxide/ P450 CYPIOI. Only most recently, however, has
hydroperoxide anion state is an essential and structural information shed critical light on how
crucial aspect of the detailed structural "fine- these residues work in concert to deliver protons
tuning" provided by the enzyme active center. at the right place and time.
Activation of Molecular Oxygen by Cytochrome P450 153

Figure 5.2. The P450 catalytic cycle.

An oxidase mechanism of oxygen activation generate the hydroperoxo-ferric intermediate"*^' ^^.


supported by NADH has also been suggested as The formation of this intermediate from the ferrous-
an alternative activity in the peroxidases, but was oxy complex, and its subsequent transformations
later disproved"^^. Both the cytochromes P450 and were recently investigated in detail using the
peroxidases are thought to share a high-valent method of cryogenic radiol3^ic reduction"*^' ^^' ^^.
"ferryl-oxo" porphyrin cation radical intermedi- The same iron-hydroperoxo intermediate is
ate^^' ^^. In P450 catalysis, atmospheric dioxygen assumed to be a common state in the formation of
binds to the reduced heme iron of the enzyme, "Compound F' in peroxidases and a variety of
and this ferrous-oxygen, or ferric-superoxide other systems such as the HOs^^' ^^' ^^~^^. Contrary
complex accepts one more electron from its to P450 and HO, in peroxidases the natural for-
protein redox partner to form the peroxo-ferric mation of this active intermediate involves the
intermediate. This in turn is quickly protonated to hydrogen peroxide starting material. In contrast to
154 Thomas M. Makris et al.

dioxygen, H2O2 brings the two reducing equi- delivery of two protons in addition to the two
valents and two protons necessary for the redox equivalents which are consumed per one O2
"Compound I" generation to the ferric heme in molecule. While the reduction of the heme iron is
one step. The key step in the enzymatic cycle of provided by the protein redox partner, it is an
the peroxidases, therefore, involves the abstrac- essential part of the P450 enzyme to catalyze the
tion of a proton from the proximal (closest to thetransfer of two protons to the distal oxygen atom
iron center) oxygen atom as the H2O2 molecule is of the bound peroxide anion through a sophisti-
ligated and the delivery of a proton which is cated proton relay mechanism. As noted in the
introduction, this pathway was shown to include
necessary to initiate O-O bond scission^"^' ^^. The
formation of the "0x0" complex and water is protein-bound water molecules which are stabi-
essentially a barrierless process once protonationlized at the enzyme active site through specific
occurs^^. A feature of the peroxidase mechanism hydrogen-bond interactions^^' ^^^ ^^. Through
mutagenesis of the residues involved in this
is the fact that it proceeds with no other external
source of electrons and protons, with an imidazolenetwork, it was shown that the overall enzyme
side chain from a histidine residue in the distal kinetics and the observed efficiency of coupling
pocket thought to serve as the species responsibleredox equivalents into product was very sensitive
for this proton shift. The distal histidine thus to the hydrogen-bonding properties of these
sites^^. Similarly strong effects, including drastic
serves, first, as a proton acceptor from the proxi-
mal oxygen, and then, as a donor of the same pro- changes in the formed product ratios, were found
ton to the distal oxygen at the second step of thein other enzymes of the P450 superfamily^^~^^.
reaction. Kinetics of the productive and nonproductive
Defining the parameters involved in the bind- turnover by human cytochromes P450 with
ing and proton-mediated transformation of the various substrates were extensively studied by
peroxide-coordinated ferriheme complex is criti- Guengerich and coauthors^^^^. Their results
cal in defining the properties of these states in directly show the existence of multiple steps, each
both the peroxidase and related oxygenase mech- potentially rate limiting depending on the specific
anisms. The pK^ of the Fe-coordinated HOO(H) reaction being catalyzed.
was estimated as 3.6^.0 for horseradish per- A linked event is the resultant chemical
oxidase (HRP) and cytochrome c peroxidase processing that occurs after proton delivery.
(CcP)^^' ^^. Thus, the iron-coordinated peroxide is
Following second proton transfer, the 0 - 0 bond
in an anionic form at neutral pH. The polarity of order is significantly diminished, so that heteroly-
the peroxidase distal pocket vs the dioxygen sis is ensured, with release of water and generation
carrier, myoglobin, or even the rather hydrophobicof the higher valent "Compound I" state. Without
nature of the P450 cytochromes, stabilizes the efficient delivery of this second proton, one has to
resulting charge separation upon deprotonation. consider the possibility of the radical process of
The resulting hydroperoxo-ferriheme complex 0 - 0 bond homolysis. Homolytic and heterolytic
is nonetheless unstable, as the peroxide ligand scission of 0 - 0 bond in alkylperoxides and in
weakly ligates the heme iron^^, and the dissocia- hydrogen peroxide catalyzed by various heme
tion rate constant for the peroxide anion as the enzymes have been extensively studied^'~'^^. In the
sixth ligand in Fe-microperoxidase-8 (Fe-MP8) cytochromes P450, the ratio of homolytic/
and Mn-MP8 is in the neighborhood of 10-20 s"' heterolytic cleavage of the O-O bond has been
(ref [60]). Similar estimates can be made from addressed through numerous investigations'^^' ^^'
other kinetic studies^^' ^^ which measured the K ^^~^^. The vast majority of these studies utilized
m
the exogenous oxidant driven pathways, with a
and k^^^ values for the formation of "Compound T' variety of substituted peroxides and peroxo acids.
in reaction of HRP with hydrogen peroxide. The Significantly, the results were found to depend on
reported millimolar K^ values and the observed the ability of the leaving group to stabilize a radi-
k^^^ on the order of 10^M~^s~' suggest that cal; the more stable this species, the greater the
the dissociation rate of HOO(H) may be as high percentage of homolytic 0 - 0 bond scission^^.
as lO^s-i. The product distribution with a cumene hydroper-
In contrast, the cytochromes P450 face the oxide driven reaction was used as a measure for
challenge of "activating" dioxygen, with the
Activation of Molecular Oxygen by Cytochrome P450 155

the relative ability of particular isozyme to form pication hydroxylating intermediate in cytochrome
a "Compound I" intermediate''^' '^^^ ^^. Attempts P450 and peroxidases^^. Despite their common
were also made to correlate the "push" electron- use of intermediate states, the oxygenases and
donating effect^ ^ from the proximal thiolate with peroxidases have distinct functions, begirming
the spectroscopic markers of the porphyrin ring their active cycle with different heme ligand com-
and with the formation of "Compound I" (i.e., plexes^^"^^. The variability of different pathways
the heterolysis of O-O bond) and product^^. The of oxygen activation provides one with a tool
formation of hydroxyl radicals via homolytic to alter the performance of the enzyme through
cleavage of the O-O bond in the reconstituted different mutations, or even to directly design new
P450 CYP2B4 system resulted in covalent types of activity into well-known systems^^' ^^^.
modification of the protein and degradation of However, only in the last decade have the proton
the heme^^. delivery mechanisms in P450 catalysis been
The general acid catalysis of heterolytic O-O directly revealed^^' 39-42, 62, 92-94, ioi-io4_ j^ese
bond scission via protonation from solvent water important details of P450 catalysis will be dis-
was suggested by many authors^^. In the absence cussed in detail in the remainder of this chapter.
of additional catalysis of heterolytic scission
through the second protonation of the distal oxy-
gen atom, heterolysis/homolysis of O-O bond is
governed by the general thermodynamic stability
3. Enzymatic Cycle of
of the reactant and product, as predicted by Lee Cytochrome P450
and Bruice^^ for porphyrin models, and recently
analyzed by Que and Solomon^^' ^^' ^^"^^. If the It is humbling to note that an outline of the
iron-peroxide complex is predominantly in a high- common catalytic cycle for the cytochromes P450
spin (HS) state, as it is very often in nonheme was proposed as early as 1968"^' ^^^. The critical
metal enzymes with weak or moderate ligand features of an atmospheric dioxygen binding event
field, homolysis of the 0 - 0 bond is favorable, as interspersed between two single-electron transfer
the HS state of the iron in both the reactant and events remain as a generally accepted outline of
product forms is conserved, with the process the P450 catalytic "wheel" ^3, io6 (Figure 5.2).
involving release of a hydroxyl radical. The sequential two-electron reduction of cyto-
For heterolytic O-O scission following a chrome P450 and the existence of multiple inter-
second protonation, in which the departing mediates of substrate binding and reduction
oxygen atom leaves as water, electron density is was documented in bacterial P450 CYPlOl (refs
withdrawn from the porphyrin moiety, and a [107]-[109]) and in microsomal systems^^^' ^^^
porphyrin pi-cation radical is formed. The het- Substrate binding to a resting state of the low-spin
erolysis/homolysis ratio and overall product dis- (LS) ferric enzyme [1] perturbs the water, coordi-
tributions are thus coupled in the native enzyme nated as the sixth ligand of the heme iron if there
systems. Various parameters such as bulk and is a close fit to the active-site pocket, and alters
local pH, ligation state of the metal, structure, and the spin state to favor the HS substrate-bound
redox properties of porphyrin and peroxide complex [2]. In P450 CYPlOl, the HS Fe^^ form
species certainly play important roles in control- of the protein has a more positive redox potential,
ling the spin state, O-O bond order, and proton is a more efficient electron acceptor from its
delivery events. redox partner, and accompanies a correspondingly
Perhaps the most important is the enzyme's tighter association of the substrate to the reduced
ability to control the delivery of protons to the form of the protein [3]^^^. In other systems, it
developing negative charge of the iron-dioxygen was observed that this spin shift is absent or
complex as electrons are introduced into the incomplete for some combinations of P450
center. The protonation/deprotonation events at isozyme and substrate^ ^^. Oxygen binding leads to
the bound superoxo, peroxo, and hydroperoxo an oxy-P450 complex [4], which is the last quasi-
anions were theoretically analyzed and claimed to stable and observable intermediate in the reaction
account for the definitive reaction sequence in the cycle. Subsequent sequential steps are reduc-
formation of the active oxo-ferryl porphyrin tion of the Fe02 complex, formation of the
156 Thomas M. Makris et al.

peroxo-ferric intermediate [5a], its protonation 3.1. The Ferrous-Dioxygen


yielding a hydroperoxo-ferric intermediate [5b], Complex
a second protonation at the distal oxygen atom
and subsequent heterolysis of the 0 - 0 bond to Oxygen binding to reduced P450 yields the
form the iron-oxo "Compound I" [6] and water, species [4], Fe^+-00 (ferrous-dioxygen) or
and finally the oxygenation of the substrate to Pe3+_oo~ (ferric-superoxide) complex. The
form a product complex [7] and its subsequent chemical structure of oxy-P450 is similar to anal-
release. These steps have, over the years, been ogous complexes in oxygen-carrier-heme proteins
addressed by many methods, but direct observa- (hemoglobin and myoglobin) and other heme
tion of key intermediates was difficult due to enzymes (HRP, HO, etc.). This ferrous Fe^^-OO
the high inherent reactivity of these states and complex is EPR silent, but shows Mossbauer
their lack of accumulation in kinetic studies. For quadruple splitting of the heme iron center consis-
instance, [5a], [5b], and [6] had not been unam- tent with a ferric state ^^'^, similar to that observed
biguously observed until recently, and alternative for "Compound IE" in HRP^^^ and for oxy-Hb^i^.
bidentate structures of the "reduced oxyP450" had The low frequency of the O-O stretch band,
been proposed^^' ^^. observed at 1140 cm"' in the resonance Raman
A beautiful feature of the overall reaction spectra of P450 CYPlOl^'^ is also typical for a
pathway for the cytochrome P450s is the rich superoxide complex. These features have often
chemistry afforded by the various steps in the prompted discussion about the correct electronic
reductive metabolism of the heme-oxygen center. assignments of these complexes and more sophis-
These include nucleophilic, electrophilic, and ticated models, such as a three-centered four-
hydrogen-abstracting species. In addition, there electron ozone-like bond that was envisioned
are at least three branchpoints where multiple almost 30 years ago' •^.
side reactions are possible and realized in vivo The properties of "oxy-P450" were first
(Figure 5.2). Abortive reactions include: (a) autox- characterized in P450 CYPlOl and other
idation of the oxy-ferrous enzyme [4] with con- isozymes using optical absorption''^"'^'', reso-
comitant production of a superoxide anion and nance Raman"^' '^^' '^^, and Mossbauer""^ spec-
return of the enzyme to its resting state [2], (b) a troscopy. The electronic structure of P450 and
"peroxide shunt," where the coordinated peroxide chloroperoxidase (CPO) complexes with O2, CO,
[5b] dissociates from the iron and forms hydrogen and CN~, together with their magnetic circular
peroxide, thus completing the unproductive dichroism (MCD) spectra were also analyzed^'' '^^.
(in terms of substrate turnover) two-electron Theoretical explanation of the specific split Soret
reduction of oxygen, and (c) an "oxidase uncou- band for the complexes of the ferrous P450
pling pathway," whereby the "ferryl-oxo" and CPO with diatomic ligands was also pro-
intermediate [6] is reduced to water by two vided in the mid-1970s'^^' '^^ for the case of car-
additional electrons in lieu of hydrogen abstrac- bon monoxide, and later experimentally extended
tion and subsequent substrate oxygenation. This to other electron-rich ligands with a strong back-
results in an effective four-electron reduction of donation (dioxygen, cyanide, thiolate)^'' '^'^.
the dioxygen molecule with a net formation The absorption spectra and autoxidation
of two molecules of water, thus anointing the properties of oxygen complexes for several
P450s with a "cytochrome oxidase" activity. cytochromes P4505^' 121, 122, no, 131^ ^j^^ related
The oxidase activity in P450, in comparison to enzymes, such as NOS'^^ and CPO'^"^, have also
those enzymes involved exclusively in bioenerget- been reported. While all of these states are not
ics, proceeds at a drastically reduced rate and particularly stable, and are quickly autoxidized,
without a productive proton-pumping cycle. Thus the autoxidation rates for P450 and other thiolate-
the physiological processing of dioxygen and ligated enzymes (CPO and NOS) are significantly
substrates via reductive metabolism is a rich and higher than for the oxygen carriers Mb and Hb.
varied set of events. The control of the specific This can be aided by a general acid catalysis in
reactivities is determined by proton delivery in the the heme monooxygenases, through facile proto-
enzyme. nation of the coordinated oxygen'^"*. As will be
Activation of Molecular Oxygen by Cytochrome P450 157

discussed in later sections, active proton delivery heme proteins^^^, including cytochrome P450''^^.
to the bound oxygen or peroxide ligand is a salient Particularly noteworthy is the signature LS EPR
feature of the P450 mechanism of oxygen activa- spectra with narrow g span (^ = 2.25-2.31,
tion, and similar mechanisms could be responsible 2.16-2.21, 1.93-1.96) and the red shift of Soret
for faster autoxidation of the ferrous-oxygen band (as compared to the spectrum of oxy-ferrous
intermediate in these enzymes. precursor) of the Fe^^-OOH" complexes in heme
The recent use of cryocrystallographic tech- systems. The direct reactions of superoxide anion
niques has enabled the direct visualization of with free Fe^^-porphyrins in the absence of strong
intermediate states in the P450 cycle, and thus proximal ligand usually afford the HS Fe^^-00^~
the modulation of active-site moieties within the complex with a side-on bound peroxide and iron
catalytic timeframe. With regards to dioxygen displaced out of the porphyrin plane toward the
ligation, however, the X-ray structure of Fe^^-OO bound ligand^^' ^^' ^'^^. In order to realize such a
complex of P450 CYPlOl determined recently'^^ structure in a heme protein, it would be necessary
demonstrates that the structure of the heme to break the proximal ligand bond to the iron and
ferrous-oxy complex of P450 appears very similar force a ir-bonded configuration, acting against the
to analogous complexes of other heme enzymes ^^^' steric hindrance of the heme prosthetic group. It
^^^. Oxygen is found to be coordinated in the bent turns out that the presence of the strong proximal
"end-on" mode (with a F e - 0 - 0 angle of 136°). ligand, which favors the LS state in hexacoordi-
This structure provides a starting point for the nated Fe^^-OOH~ complexes, is an important
formation of the end-on bound peroxo-ferric restriction on the chemistry of oxygen activation
complex, which is the first of three unstable and in the heme enzymes. Structure and properties of
highly reactive intermediates in the P450 catalytic model Fe^^-OOH~ complexes in heme enzymes
cycle. have been extensively studied theoretically^^' ^^' ^^'
93,96,97,145,146 ^hesc investigations, together with
analogous studies on heme^"^^ and nonheme
3.2. Reduction of Oxy-Ferrous enzymes ^^ have played a definitive role in estab-
P450 and Formation of lishing the currently accepted view of the impor-
Peroxo-Ferric Complexes: tance of the second protonation step of the distal
oxygen atom of the peroxide ligand for the effi-
Properties, Stability, and cient heterolytic cleavage of the O-O bond and
Spectroscopy "Compound I" formation.
The stability of iron-peroxo complexes is Early attempts by Estabrook and Peterson to
marginal in heme systems in the presence of a create and stabilize the reduced oxy-ferrous, or
strong proximal ligand (His, Cys, or Tyr), and the "peroxo" complex in cytochrome P450 were first
aqueous solution at a near-neutral pH. The numer- made more than 30 years ago"^' ^^^ Subsequent
ous attempts to isolate such complexes obtained in efforts included steady-state and stopped-flow
reactions of hydrogen peroxide with P450 at studies of reconstituted systems^"^^"^^^, replacing
ambient conditions have failed because of the dioxygen by superoxide^^^' ^^^ or peroxides as
inherent low stability and fast conversion to oxygen donors, or the use of alternative chemical,
"ferryl-oxo" species with O-O bond scis- photochemical ^^^' ^^^, and pulse radiolytic^^^' ^^^
sion^^^. There have been successful isolations of methods for fast and efficient reduction of the
the ferriheme-peroxide complex in myoglobin, preformed oxy-ferrous P450. However, only in
however^ ^^. recent years have reproducible, stable, and high-
Chemical models of the Fe^^-OOH~ and yield preparations of Fe^^-OOH~ complexes
Fe^^-OOR~ complexes have been prepared by been obtained with cytochrome P450 and other
reacting metalloporph3^ins with peroxides at heme systems. This was achieved using the
low temperatures (either 200-230 K in solutions methods of radiolytic reduction of oxy-ferrous
or freeze-quenched at 120 K and below) and stud- precursors in frozen solutions at 77 K"^^' ^^' 1^7-159
ied by EPR, NMR, and optical spectroscopic Irradiation with high-energy photons from a ^^Co
methods^^^"^^^. Similar results were obtained with gamma-source or using ^^P enriched phosphate as
158 Thomas M. Makris et al.

an internal source of high-energy electrons"^^' ^^^ cytochrome P450, but at 420 nm in HRP and HO,
generates radiolytic electrons, which reduce a pre- Figure 5.3.
formed oxy-ferrous complex which is stabilized The radiolytic reduction of [4] at 77 K yielded,
against autoxidation by low temperature. Similar in most cases, an already protonated hydroperoxo-
approaches have long been used by physicists and ferric complex [5b]. In several proteins, however,
chemists in matrix isolation chemistry of highly such as oxy-Mb, oxy-HRP, and the D251N variant
reactive intermediates^^^~^^^. In the solid state, at of cytochrome P450 CYPlOl, it was possible to
the temperatures below the glass transition where observe the unprotonated species [5a]. Interest-
translational and rotational diffusion is minimized, ingly, the irradiation of oxy-P450 at 4 K in liquid
the peroxo complex is stable due to the impedi- helium yielded the unprotonated form [53]^*^
ment of trap migration and proton transfer events. suggesting that an activation process of proton
Historically, in biological systems, cryogenic transfer occurs between liquid helium and ligand
radiolysis was first used as a tool for studies of the nitrogen temperatures. Most exciting is the direct
nonequilibrium intermediates in heme proteins observation of the protein-catalyzed proton trans-
in the early 1970s^^^. For example, radiolysis of fer event, [5a] to form [5b], as the temperature is
several ferric proteins with different ligands in raised. A second protonation and catalytic conver-
frozen aqueous-organic solutions at 77 K was sion of the substrate to a product complex then
shown to produce the corresponding ferrous ensues"^ ^. Alternatively, a separate uncoupling
species, which then could be annealed at elevated channel can be opened with peroxide release and
temperatures, and the conformational and chemi- direct transition to the resting state of the enzyme,
cal relaxation processes monitored by EPR and demonstrating the subtle control of proton deliv-
optical spectroscopy methods ^^'^^^^. The first ery provided by the protein matrix. The lack of
reports on cryoradiolysis of oxy-ferrous com- "Compound I" formation on the oxidase pathway
plexes in heme proteins and formation of the in HRP'^' is explained by the inability of this
Fe^^-OO(H)^"^"^ "peroxo" complex in hemoglo- enzyme to deliver this second proton to bound
bin, myoglobin, and HRP'^^' ^^^'^^ established the dioxygen, despite the facile formation of the
characteristic EPR spectrum of Fe3+-00(H)2-(-) "Compound I" from hydrogen peroxide.
complexes with a signature narrow span of Thus, during the last decade, the method of
g values (2.3-2.25, 2.2-2.14, and 1.94-1.97). cryoradiolytic reduction has emerged as a new
Optical absorption'^^' '^'^' '^^, Mossbauer^'^, and tool to investigate critical intermediates of redox
EPR analysis have been used to characterize the systems. X-ray-induced radiation chemistry is
peroxo intermediates in heme enzymes including also increasingly recognized both as a potential
cytochrome P450 CYPlOl'^^' '^^ source of misinterpretations due to measurement-
Recently, this approach was further expanded induced changes of the sample, and as a new,
to understand the detailed electronic structure and important tool in X-ray crystallography, where
stability of peroxo-ferric intermediates in heme'^^' the irradiation of protein crystals may be used to
41, 49, 50, 53, 157, 159, 179-181 ^ ^ d nOUhcmC SystcmS^^' deliberately alter the redox state of metals, flavins,
182-188 ^g ^ result, the direct spectral identifica- disulfides, and other cofactors'*^' i89-i9i jj^g
tion of the intermediates [5a] and [5b] in several chemical details of the radiolytic process in frozen
heme proteins has been clearly achieved. solutions and protein crystals may, however, be
Importantly, the EPR spectra were found to be quite different. In the former, there is usually a high
sensitive to the protonation state of the peroxide concentration of glycerol or ethylene glycol present
ligand, but less responsive to the nature of the as a cosolvent to improve the optical glass quality
^ra«5-proximal ligand (His or Cys), Table 5.1. of the sample at low temperature. These cosolvents
UV spectroscopy shows a weak response to are necessary as effective quenchers of hydroxyl
the protonation state of the peroxide with the radicals generated by the radiolysis of water. On
Soret and Q bands shifting by only a few nano- the other hand, protein crystals can sometimes
meters with iron-peroxo protonation, but high contain a much lower concentration of organic
sensitivity to the identity of the proximal ligand. cosolvents, thus potentially altering the processes
For example, the Soret band of the [5b] inter- operating during low temperature radiolysis and
mediate appears at 440 nm in the thiolate-ligated thermal annealing. For example, it was shown^^^
Activation of Molecular Oxygen by Cytochrome P450 159

Table 5.1. Electron Paramagnetic R e s o n a n c e ( E P R ) Parameters o f M o n o n u c l e a r Peroxo-Ferric


C o m p l e x e s from Select Synthetic M o d e l s and E n z y m e s

Model Ligands 02/e donor g-value Assignment References

Synthetic models
Bztpen(OOH)2+ N4 H,0, 2.22,2.18, 1.97 FeOOH (Til) [277]
Bztpen (O^f^ N4 H2O2, base 7.6, 5.8, 4.5 FeOO (TI2) [277]
Fe(SMe2N4tren) N4S H2O2 2.14, 1.97 FeOOH (Til) [278]
Fe(rtpen) N5 H2O2, acid 2.20,2.16, 1.96 FeOOH (TIJ) [279]
Fe(rtpen) N5 H2O2, base 7.6, 5.74 FeOO (TI2) [279]
Fe(rtpen) N5 Methanol 2.32,2.14, 1.93 FeOCH3 (Til) [279]
trispicMeen N5 up. 2.19.2.12, 1.95 FeOOH (Til) [280], [281]
trispicMeen N5 H2O2, acid 7.4, 5.7, 4.5 FeOO (Ti2) [280], [281]
TPEN N5 H2O2 2.22,2.15, 1.97 FeOOH (Til) [281]
BLM N5 H2O2 2.26,2.17, 1.94 FeOOH (Til) [281]
trispicen N5 H2O2 2.19,2.14, 1.97 FeOOH (Til) [281]
FePMA N4 H2O2 2.27,2.18, 1.93 FeOOH (Til) [282]
TMC N4 KO2 8.5, 4.23 FeOO (TI2) [283]
OEC N4 KO2 8.8, 4.24 FeOO (TI2) [283]
MemMemxyl N4 H2O2 or KO2 2.23,2.16, 1.93 FeOOH (Til) [284]
2.25,2.10, 1.97
FeTPP N4O BuOOH 2.31,2.16, 1.96 FeOOR (Til) [285]
FeEDTA N2O3 H2O2 4.2,4.15,4.14 FeOO (TI2) [286]
FeOEP N4 Me4N02 9.5, 4.2 FeOO (TI2) [287]

Enzymatic intermediates
Horseradish peroxidase N„ 02,7 2.32,2.18, 1.90 FeOOH (Til) [181]
Histidine 2.27,2.18, 1.90 FeOO (Til)
Nitric oxide synthase N4 02,7 2.26,2.16, 1.95 FeOO (Til) [179]
Cysteine
P450 N4 02,7 2.25,2.16, 1.96 FeOO (Til) [40], [41], [158]
Cysteine 2.30,2.17, 1.96 FeOOH (Til)
P450 N4 BuOOH 2.29,2.24, 1.96 FeOOR [144]
Cysteine
Superoxide reductase HiS4 H2O2 4.30,4.15 FeOO (TI2) [288]
Cysteine
Heme oxygenase N4 02,7 2.25,2.17, 1.91 FeOO (Til) [54]
Histidine 2.37,2.18, 1.92 FeOOH (Til)
Hemoglobin N4 02,7 2.25,2.15, 1.97 FeOO [171], [177]
Histidine 2.31,2.18, 1.94 FeOOH (Til)
Myoglobin N4 02,7 FeOO (Til) [177], [178]
Histidine FeOOH (Til)
Myoglobin N4 H2O2 2.29,2.16, 1.91 FeOOH (Til) [138]
(H64N,V) Histidine

that the yield of cryoradiolytically reduced ribonu- redox centers, as occurs during radiolysis at room
cleotide reductase increases 100-fold when the temperature^^^. The presence of the broad absorp-
glycerol concentration is raised from 0% to 50%. tion band in the visible and near infrared, charac-
An insufficient concentration of the quencher of teristic for the trapped electrons in irradiated
OH' radicals in the protein crystal will result in frozen samples, is an indication of the successful
oxidative, instead of reductive, modification of entrapment of hydroxyl radicals which would
160 Thomas M. Makris et al.

O
C
03

O
(0
<

600 700
Wavelength, nm
Figure 5.3. Absorption spectra of ferrous-oxy (1) and hydroperoxo-ferric (2) CYPlOl at 80 K in 70% glycerol/
phosphate buffer.

Otherwise recombine with radiolytically produced several different transformations, including dis-
electrons and thus change the overall radiolysis sociation from the metal center as a hydro-
products observed ^^^. peroxide anion. The dissociation rate for the
EPR spectroscopy has been a powerful tool hydroperoxo-ferric intermediate was estimated
to investigate radiol)^ic events in nonheme com- for microperoxidase as 10-20 s~' (ref [60]), and
plexes ^^^' ^^^. Very recently, the signature EPR the half-life of these complexes at ambient condi-
spectrum for the Fe^^-OOH~ intermediate in tions is likely to be much less than a second. The
activated bleomycin was calculated^^' '^'^, and the dissociation of the peroxide ligand, termed the
analysis of electronic structure of this complex "peroxide shunt," is shown in Figure 5.2 as a
provides a detailed insight into the chemical sta- conversion of intermediate [5b] to [2]. It is actu-
bility of Fe^^-OOH~ moiety. Neese also noted ally a reversible process since a "peroxygenase"
that the side-on coordinated iron-peroxide is not reaction wherein [2] is converted to [5b] with the
activated for O-O bond cleavage ^^^. addition of exogenous peroxide is also possible.
Homolytic cleavage of the O-O bond generating
a hydroxyl radical and forming "Compound II,"
3.3. The Second Branchpoint of a ferryl-oxo porphyrin complex, or heterolytic
P450 Catalysis: Uncoupling scission of the 0 - 0 bond to produce water and
with Hydrogen Peroxide "Compound I" are also competing processes.
Production or Dioxygen The rate constants for the latter two are difficult
to measure.
Bond Scission
In peroxidases, both protons are essential for
The formation of the hydroperoxo-ferric heme heterolytic scission of the 0 - 0 bond and subse-
intermediate is a common step in the different quent formation of "Compound I." Hence, in
pathways, in oxygenases and peroxidases. In order for a similar mechanism in P450 to occur,
heme enzymes, this intermediate can undergo the enzyme must deliver two protons to provide
Activation of Molecular Oxygen by Cytochrome P450 161

the same reaction stoichiometry. If the second 4. Structural Input into


proton is not available, the hydroperoxo-ferric the Mechanisms of
complex can still undergo O-O bond scission, but
must compete with dissociation of the complex to P450-Catalyzed Dioxygen
form hydrogen peroxide. The weakness of perox- Activation
ide ligand affinity provides a straightforward
explanation for the possibility of uncoupling at While the purely chemical characterization of
this stage. The low binding affinity for the perox- P450 reaction intermediates has largely focused
ide to the heme presumes the shift of the equilib- on redox-linked changes in heme-iron systems, a
rium [5b] < > [2] (Figure 5.2) to the right at low complete understanding of intermediate evolution
peroxide concentrations. In the case of protona- and reactivity must involve the role of the protein
tion of the distal oxygen, the O-O bond is cleaved scaffold in dioxygen scission. In the case of P450
with little or no apparent barrier to the reaction^"^^. monooxygenation chemistry, this goal has been
The resultant "Compound I" [6] then reacts formulated into ascertaining the role of the distal
with the substrate or undergoes an "oxidase pocket in the control of concerted proton transfer
shunt" process to be discussed in a subsequent events to a reduced oxy-ferrous intermediate,
section. resulting ultimately in heterolytic cleavage to pro-
Unfortunately, the only method of observing duce the high-valent oxo-ferryl, or "Compound I"
the intermediate [6] to date in P450 enzymes has intermediate. While numerous processes in the
been through the reaction of the ferric enzyme P450 catal5^ic scheme have been linked to both
with exogenous oxidants such as with m- protons and the ionization state of particular
CPBA^^^^^^. Direct evidence for the quasi-stable residues, we focus specifically on those proton
existence of a "Compound I" state, through meas- transfer events that result in the transformation of
urement of both formation and breakdown kinet- the reduced ferrous-dioxygen complex, to yield
ics, in the cytochromes P450 has been provided the hydroperoxo intermediate. It is critical not
only recently^^^. This work utilized hyperther- only for any structural model to explain a frilly
mostable CYP119 and low concentrations of per- ftanctional "wild-type" P450 reaction chemistry,
oxyacid to prevent secondary reactions. The but also to give a structural basis for the compila-
attempts to detect "Compound I" in annealing tion of kinetic and spectroscopic parameters that
experiments with cryogenically generated peroxo- result from alteration of the active site by mutage-
and hydroperoxo-ferric complexes in P450, nesis or by utilization of a variety of alternate sub-
wherein atmospheric dioxygen was sequentially strate structures. This then translates into
reduced, have not been successfiil. Possible rea- knowledge of the inherent reactivity of peroxo-
sons include the very high rate of substrate oxy- ferric complexes on the pathway to "Compound I"
genation by this active intermediate, and the formation, the control of branchpoints and their
subtleties of competing proton delivery pathways. specific chemistries providing oxidant in nucle-
Recently, Friesner et al. (personal communication) ophilic, electrophilic, and radical oxidations. The
estimated the barrier height of the reaction [6] -^ entire P450 reactivity landscape cannot be unified
[7] in a P450 model to be only a few kcal mol~^ into a strict and inflexible structural model, as
Thus, the catal)^ic step of hydrogen abstraction some aspects of proton delivery and dioxygen
and oxygen rebound^^^ may be too rapid to allow activation are "finely tuned" according to the
intermediate buildup. It is worth noting that [6] structural requirements of a particular P450
does not accumulate even during the annealing of isozyme active site and are moderated by particu-
cryogenically irradiated oxy-samples at low tem- lar substrate recognition events. Nonetheless,
peratures 180-200 K. However, the use of solvent many common structural motifs of the enz3mie
exchange and ENDOR spectroscopy^^ of the prod- distal pocket have been evidenced which link
uct complexes was able to provide important indi- the P450 superfamily both mechanistically and
rect evidence for the operation of a "Compound I" structurally.
like species. The attempts to detect these interme- In order to resolve structural components
diates in experiments with substrate-free P450 potentially involved in proton donation to a
have also proved frustrating. reduced ferrous-dioxygen or peroxo-anion species.
162 Thomas M. Makris et al.

at least three potential mechanisms of proton acid-alcohol side-chain pair is observed in a


delivery should be addressed. The first involves large majority of P450 genes. These are typically
ionization in which the relay of protons involves a threonine, or in some cases serine, and either
change in the charge state of the participating aspartate or glutamate^^^, 209 j ^ P450 CYPlOl
amino acid side chain or solvent moieties. The these residues are Asp251 and Thr252. Given their
assignment of participating side chains involved in placement in proximity to a heme-dioxygen bind-
proton transfer can be inferred by their local pK^ ing site, a plethora of mutagenesis experiments
values. In a second type of process, the two partic- provided mechanistic suggestions as to their role
ipating moieties undergo simultaneous protonation in stabilizing distal pocket hydrogen-bonding
and deprotonation in a Grotthuss mechanism^^^, networks and contributing to proton delivery to a
and all moieties participating in hydrogen-bonding peroxo-anion complex. For example, upon muta-
interactions can be potential proton donors in the tion of the conserved threonine in CYPlOl to a
enzyme active site. Finally, the involvement of the hydrophobic residue, one observes normal kinetic
peptide bond itself in proton-transfer reactions has parameters of pyridine nucleotide oxidation and
been implicated in the proton-transfer mechanism dioxygen consumption, yet the enzyme is fully
of CcO^^^ and model peptides^^^. uncoupled, resulting almost entirely in hydrogen
The identification of potential proton-transfer peroxide production rather than the oxidation
conduits in P450 catalysis was aided by the first product hydroxycamphor^'^' ^' ^ This phenotype is
P450 crystallographic structure of CYPlOl from certainly not exclusive to the case of CYPlOl, as
Pseudomonas putida^^^^ ^^^. While this structure similar mutations across the space of P450 phy-
played an indispensable role in visualizing the logeny have shown similar effects^^^' ^^^. The roles
structural features that years of P450 spectro- assigned for the "conserved" threonine have
scopic studies had suggested, such as confirma- included a direct proton source for the peroxo-
tion of the ligands of the HS substrate bound anion species, or a secondary effect such as stabi-
ferriheme species^ ^"^^ ^^^, the positive identifica- lizing critical hydrogen bonding and water
tion of proton donors in the distal pocket was elu- placement which in turn serves in proton delivery
sive. In fact, the P450 CYPlOl distal pocket was and dioxygen bond scission. Several mutagene-
shown to be mostly hydrophobic in nature, in sis experiments were designed to discriminate
sharp contrast to the polar active sites of other between these two possibilities. Ishimura and
heme enzymes, such as CcP^^^ and catalase^^'^. Of colleagues, for instance, demonstrated that the
particular interest was the absence of charged uncoupling reaction resulting in peroxide forma-
residues in the immediate vicinity of the heme- tion could be avoided if the Thr residue was
oxygen binding site, such as the well-character- altered to a side chain which could participate in a
ized acid-base pairs (His-Arg and His-Asp) in hydrogen-bonding interaction'^^' ^^^. This is par-
the peroxidase and catalase structures, which were ticularly well evidenced in the cases of Asn252
thought to mediate dioxygen scission reactions. and Ser252 mutations in P450 CYPlOl, where
Readily apparent from these early structures of both substitutions retain more than half of
CYPlOl however, was the interruption of normal the enzyme's activity with regards to pyridine
hydrogen-bonding pattern in the distal I-helix, and nucleotide oxidation and hydroxylation turnover.
hydrogen bonding of the Thr252 side chain with Furthermore, through the utilization of unnatural
the carbonyl oxygen of Gly248. The resultant amino acid mutagenesis^ •^, this avenue could be
"kink" in the I-helix has proved to be a common explored without the stringency of naturally
structural element of most P450 isozymes, and occurring side chains. In an important experiment,
was postulated to create a pocket in which dioxy- the hydroxyl side chain on the conserved alcohol
gen could easily bind^^^. side chain (Thr252 in CYPlOl) was replaced
with a methoxy groups'^' ^'^, although the modi-
fied protein was not structurally characterized.
4.1. A "Conserved" Alcohol Side Interestingly, the mutant enzyme still retained
Chain in the Active Site relatively normal kinetic parameters and nearly
of P450 full coupling of reducing equivalents into product.
Upon examination of aligned sequences of Some caution is needed, however, since a common
P450 isozymes, it has become clear that an activity of the C3^ochromes P450 is oxidative
Activation of Molecular Oxygen by Cytochrome P450 163

demethylation^^^' ^^^ and a single turnover could pocket, have since been shown to be important
result in restoration of a functional hydroxyl at the in understanding peroxo-ferric stabilization and
enzyme active site. Nonetheless, this result has reactivity.
often been interpreted to imply that the Thr252 The implication of "extra" water as a potential
side chain itself is not directly involved in donat- source of uncoupling has also been included as a
ing protons to a reduced oxygen intermediate but parameter in the redesign of the substrate-binding
rather provides its ftinctionality by providing pocket to oxidize hydrocarbon substrates of
hydrogen-bonding interactions that stabilize varying sizes. In the reaction of CYPlOl with
active-site waters that are the immediate source of ethylbenzene, for example, the effective coupling
catalytic protons ^^^. to monooxygenase activity is highly sensitive
High-resolution X-ray crystallographic studies to the sterics of the substrate-binding pocket^'^' ^^^.
of the Thr252Ala mutant have also proved essen- Although a redesign in the distal pocket to
tial in dissecting its role in catalysis and peroxide accommodate different substrates certainly dimin-
evolution^^. Despite mutating this residue to one ished unwarranted uncoupling pathways, both at
unable to form a hydrogen bond to the carbonyl the peroxide and oxidase branchpoints, a strict
oxygen of Gly248, the I-helix "kink" is still mapping of potential solvation pathways leading
observed in the mutant enzyme. The observed new to peroxide uncoupling was difficult. Active-site
solvent molecules in this mutant structure^^, in mutagenesis also served as a starting point for the
particular Wat720, have been inferred as a source redesign of the active sites in other P450 isozymes
of peroxide uncoupling in the mutant. This could to oxidize a number of substrates of varying polar-
arise either by decreased stability of the peroxo ity and size^^^~^^^. While the precise structural
ligand in protic solvents or due to the delivery of details behind the uncoupling pathways in these
protons to the proximal oxygen, both of which "redesigned" enzymes is not yet realized, an
would result in the release of hydrogen peroxide^^. important independence of substrate-binding
The inclusion of extra waters as a potential source parameters and efficient coupling to monooxy-
of uncoupling has likewise been structurally genation is clear.
inferred with a series of camphor analogs bound Even though the presence of an active-site alco-
to CYPlOl (ref [36]) and in the substrate-free hol ftinctionality is highly conserved throughout
enzyme^^^. The use of alternate substrates for the the landscape of P450 isozymes, one important
enzyme displayed a varying degree of hydroxyla- exception has ftirther established the importance of
tion regiospecificity and uncoupling, resulting an active-site hydrogen-bonding network in dioxy-
either in the production of peroxide or water gen catalysis. CYP107 (P450eryF), which catal-
through an "oxidase" channeP^^"^^^. The former yzes the hydroxylation of 6-deoxyerythronolide B
can often be explained through an increased sub- in the erythromycin biosynthetic pathway, has an
strate mobility owing to perturbed hydrogen- alanine (Ala245) instead of threonine at the con-
bonding interactions with Tyr96^^^' ^^^, but a served position^^^' ^^^. Despite the absence of the
consistent water-mediated mechanism was diffi- I-helix threonine, a kink or cleft is still retained in
cult to understand for the latter. Nonetheless, the structure and the side chains of Glu360 and
the localization and superposition of active-site Ser246 and a series of ordered water molecules
waters has emerged as being of critical impor- contribute a distal pocket hydrogen-bonding net-
tance in evaluating the proton-aided dioxygen ^Qj.]^232,233 Interestingly, the 5-hydroxyl group of
scission and uncoupling chemistries. The crystal- the macrolide substrate provides a key H-bonding
lographic structure of Thr252Ala also demon- interaction to a water molecule (Wat564). This
strated that, both the mutated residue and Asp251 water is in a nearly identical position as the threo-
residue adopt different conformations^^. For nine hydroxyl moiety in P450 CYPlOl, thus sug-
example, in the Ala252 mutant, the Co atom gesting a similar proton delivery network via
moves 1.4 A away from the dioxygen-binding "substrate-assisted catalysis"^^"*. Alteration of the
pocket. In addition, the Asp251 backbone car- substrate C5 and C9 side chains, which normally
bonyl has flipped (120°) toward the distal donates a hydrogen bond to crystallographically
helix. The participation of solvent in mediating observable active-site water (Wat563), results in
P450 catalysis, and the conformational adapta- dramatic effects on catalytic efficiency. Through
tion of the acid-alcohol pair in the distal reintroduction of the active-site hydroxyl in the
164 Thomas M. Makris et al.

Ala245Ser and Ala245Thr mutant enzymes, it Thr252Ala/Asp251Asn, in which the stoichiomet-


appears as though the loss of catalytic activity^^^ ric ratio of the two pathways is equal at room
in the mutant forms correlates with the loss temperature ^^^. As primary proton transfer is also
or repositioning of Wat519 (ref [234]). None- visible at low temperature in this mutant con-
theless, these mutants are still able to metabo- struct, prior to decay to the resting state, one is
lize the substrate testosterone at different sites^^^, able to definitely assess that the protonated peroxo
implying that a very intricate hydrogen- intermediate serves as the branching intermediate
bonding interplay exists between substrate and of these two pathways.
enzyme.
Molecular dynamics simulations performed by 4.2. The "Conserved" Acid
Harris and Loew^^ provide additional evidence
Functionality
regarding the role of active-site hydrogen bonding
in CYP107. By subjecting the peroxo-anion As in the case of the conserved hydroxyl
species in the substrate-bound active site of the moiety in the P450 active site, a conserved acid
enzyme to molecular dynamics simulation, two- residue has similarly been implicated as the criti-
proton-donating partners, the C5 hydroxy 1 of cal proton transfer vehicle in the P450-catalyzed
the bound substrate, and a bound water Wat519 oxygen activation. Upon mutagenesis of Asp251
are seen to form-up hydrogen bonds on the distal in P450 CYPlOl to asparagine, a phenotype quite
oxygen atom of the ligand within 25 ps from distinct from that of the alcohol mutagenesis is
initiation of the simulation^^. Furthermore, con- observed^^' ^^K Instead of the normally high, but
nectivity of this hydrogen-bonding network to decoupled, rate of pyridine nucleotide consump-
other exchangeable groups, namely to Wat564, tion seen in the Thr252Ala mutant, the enzymatic
Ser246, and Glu360 suggests a specific proton turnover activity of the Asp251Asn mutant
relay pathway in the enzyme. is decreased by more than one order of magni-
In order to properly assess the molecular basis tude^^' ^^. With close examination of the separate
of peroxide forming chemistries in the Thr252X catalytic parameters of the P450 reaction wheel
mutants, one must first adequately assess the (Figure 5.2), this decreased activity was specifi-
operant chemical mechanism or mechanisms that cally localized to second electron transfer and
give rise to the observed uncoupling. As described linked protonation events following formation of
earlier, the use of cryogenic irradiation has proven the ferrous-oxy complex^^' •^^. Unlike the con-
to be an invaluable tool in dissecting the particular served hydroxyl moiety in the immediate vicinity
intermediate state involved in both P450 mono- of the heme iron, a number of mutations at this
oxygenation and peroxide evolving chemistries. site have revealed that the observed phenotypical
Using low temperature radiolytic reduction of the variance is poorly explained by a discrete physic-
Tlir252Ala mutant ferrous-oxy complex, the ochemical property of the amino acid side chain
enzyme directly forms the protonated peroxo that is inserted. For example, both the Ala251 and
anion"^^' ^^ as is readily identified by the character- Gly251 substitutions behave similarly to that of
istic shift in g-tensor upon protonation of the the isosteric charge reversal Asp251Asn^^^. While
distal oxygen*''^' '^^. This result is congruent these phenotypical consequences of Asp251
in terms of the unmodified rates of pyridine mutation are similar throughout many P450
nucleotide oxidation seen in room temperature sequences, such as CYP107, the kinetic efficiency
studies. First, despite mutation, the Thr252Ala of the mutant enzymes often depends on the iden-
mutant of CYPlOl, like the wild-type enzyme, is tity of the substrate being metabolized'^^' ^36-238
able to protonate the distal oxygen atom of the Some indication of active-site acidic ftinction-
peroxo-anion species. This in turn, in the course of ality in P450 dioxygen scission can be revealed by
temperature annealing, decays to the ferriheme the comparative evaluation of kinetic parameters
resting state with no intervening intermediates. in protonated and deuterated waters. The use
In order to ftirther assign the hydroperoxo-ferric of kinetic solvent isotope effects (KSIEs) has been
species as the branchpoint of peroxide forming valuable in the determination of potential proton-
and monooxygenation studies, analogous experi- linked dioxygen activating enzymes such as
ments were performed with the double mutant. P45038, 39^ methane monooxygenase (MMO)^^^,
Activation of Molecular Oxygen by Cytochrome P450 165

and the cytochrome oxidases^'*^. It is important As in the case of the Ala252 mutation, the
to prove that the observed isotopic effects are annealing profile of cryogenically reduced oxy-
specifically linked to proton transfer events in the ferrous Asn251 mutant in P450 CYPlOl corrobo-
dioxygen cleavage reaction. In the case of cyto- rates the kinetic data just discussed. Unlike the
chrome P450 CYPlOl, detailed kinetic studies wild-type and Ala252 mutant enzymes, which
demonstrated that only the second electron trans- form the hydroperoxo-ferric complex upon irradi-
fer rate, and the linked concomitant catal3^ic steps ation at 77 K, EPR g-tensors and ^H ENDOR
were sensitive to solvent isotope content^^. This in couplings reveal that the primary reduced com-
turn has allowed the comparable gauging of iso- plex of Asn251 is the unprotonated peroxo-anion
topic sensitivity of the dioxygen cleavage reaction species"^^'"^^ Upon annealing to the glass transi-
simply through the rate ratio measurements of tion temperature (190 K), protonation of the
product formation. Through the comparative peroxo anion occurs, confirming that the pheno-
analysis of KSIEs and proton inventories of the typic effects seen in this mutant are localized
wild-type and the Asp251Asn mutant CYPlOl at primary proton delivery. Moreover, the
enzymes, a startling difference in these parame- temperature-dependent annealing profile likewise
ters is observed^^. In the wild-type enz3ane, by implies an active-site conformational change in
fitting the proton inventory to a range of fraction- the mutant protein is required to enable efficient
ation factors, it was shown that at least two proton transfer. This reorientation of the bifur-
protons are in flight during the observed transi- cated salt-bridge during CYPlOl catalysis has
tion state process. This is consistent with the additionally been implicated by the dependence
previously discussed schemes involving distal of turnover rate on hydrostatic^'*^ and osmotic^'*^
pocket waters and the hydrogen-bonding Thr252. pressures, as well as detailed photoacoustic
Furthermore, a 4-fold increase of the KSIE in the calorimetry measurements^"^^' -^^^ of both wild-
Asp251Asn mutant, as well as linear dependences type and mutant cytochromes. These results
of NADH oxidation rate on bulk proton concen- implicate a distinct and localized re-solvation of
tration, intimates that the proton delivery conduit active-site residues during substrate turnover.
in this mutant is drastically altered, perhaps medi- However, localization of these dependences to
ated by a chain of water molecules^^. protonation of the peroxo anion is complicated as
In structural terms, crystallographic analysis of it is mixed with analogous reorientations and
the Asp251Asn mutant has provided additional conformational changes occurring during other
evidence for its involvement in proton delivery^^. catalytic steps such as substrate binding. Never-
In the wild-type ferric structure of CYPlOl, theless, the implication of a rotameric flexibility
the Asp251 side chain itself points away from the of the Asp251 side chain, stabilized by discrimi-
distal pocket, and instead interacts in a bifur- nating electrostatic interactions, presents an
cated salt-bridge involving Argl86 and Lysl78. important theme in the development of a dynamic
Although other structural features of the distal model of P450 dioxygen scission.
pocket remain intact, mutagenesis to Asn251
results in the simultaneous rotation of the Asn251
and Lysl78 side chains to the protein surface, 4.3. Crystallographic Studies
resulting in loss of their intermolecular contacts. of P450 Reaction
This in turn is compensated by a new interaction
Intermediates
of the Asn251 side chain with Asp 182. These mul-
tiple motions speak to a highly plastic active site As with spectroscopic characterization of the
with a resulting open access of the heme iron from peroxo-ferric intermediates in CYPlOl, one of
the protein surface. While it is difficult to translate the most revealing studies of P450 dioxygen activa-
the observed KSIE and pH dependence data of the tion has been the recent structural resolution of
Asn251 single mutant to a rigorous structural P450 intermediates using cryocrystallography^^.
model, in part due to difficulty in visualizing This work not only provided a molecular picture of
active-site mobile waters, it nonetheless remains P450 intermediate states, but also a road-map for
consistent with a proton delivery mechanism the participation of the distal pocket in the effective
modulated by additional solvent molecules. delivery of protons to a ferrous-dioxygen species.
166 Thomas M. Makris et al.

Analogous to the spectroscopic work outlined in precursor. The accompaniment of dioxygen


previous sections, these studies rely on cryogenic ligated in an end-on (T^J coordination geometry) to
radiol)^ic reduction in the form of exposure to the heme-iron correlates to a number of structural
long-wavelength X-rays in order to isolate normally nuances. First, and perhaps most obvious, is the
reactive heme-oxygen intermediates. However, in presence of two new water molecules, WAT901
contrast to the spectroscopic analysis of low- and WAT902, in the active site of the enzyme. One
temperature reduced samples, the X-ray crystallo- of these, WAT901, appears both within hydrogen-
graphic analysis of the X-ray reduced sample is bonding distance of the dioxygen ligand and the
complicated by the fact that the X-rays not only Thr252 hydroxyl side chain (Figure 5.4). This
serve as a pulse but also as probe. Therefore, water molecule is unique to the ferrous-dioxygen
sample radiolysis cannot be totally controlled or structure, but may in fact represent the stabilized
eliminated. Diffusion of dioxygen into dithionite- water implicated in proton delivery in the threonine
reduced crystals of CYPlOl at low temperature, mutagenesis and solvent isotope-effect studies out-
and using short wavelength X-rays to mini- lined above. In addition, the backbone amide of
mize radiolysis, revealed the heretofore structurally Thr252 occupies a "flipped" orientation allowing
uncharacterized P450 ferrous-dioxygen complex. not only additional stabilization of this bound water
These structural studies nicely complement the molecule through new hydrogen-bonding interac-
low-temperature spectroscopic characterizations. tions with the peptide backbone, but also a new
Structural resolution of oxy-ferrous P450 inter-action of the Asp251 backbone carbonyl with
reveals important differences from the ferrous the amide and amino groups of Asn255.

Figure 5.4. Overlaid crystal structures of the ferrous (light) and oxy-ferrous (grey) wild-type CYPIOI.
Activation of IVIolecular Oxygen by Cytochrome P450 167

The observation of this new hydrogen bond mutants show spectral changes upon D2O
network in the crystals of the ferrous-dioxygen exchange in both linear and bent conformations.
complex provides a satisfying structural explana- As the bent conformer better represents the
tion of the observed changes in coupling and ferrous-oxy complex as observed in structural
turnover upon mutation of Asp251Asn and studies, one would expect that it is exclusively
Thr252Ala. In the case of the latter, the threonine subject to isotopic exchange in the wild-type
hydroxyl, in addition to the backbone amide, enzyme. Meanwhile, as mutation would cause
donates key stabilization interactions with both drastic changes in hydrogen-bond patterning in
bound dioxygen and WAT901. One might expect the distal pocket, it is reasonable to expect iso-
that the removal of these interactions in the topically induced vibrational changes of both con-
Thr252Ala mutant would have effects at multi- formers in the mutants. Crystallography of the
ple branchpoints of the P450 catalytic cycle. cyanide complex of wild-type CYPlOl, in which
Elimination of this H-bonding interaction would the bent conformer is observed, illustrates the
result in a relative destabilization of the oxy- interaction of the cyanide ligand with both the
ferrous complex, resulting in an increased rate of threonine hydroxyl and WAT901 (ref [246]).
autoxidation in the Thr252Ala mutant^^^ In addi- By irradiating oxy-ferrous P450 crystals with
tion, the newly observed water molecule in the longer X-ray wavelengths, catalytic processing in
ferric Thr252Ala structure, WAT720, is localized the active site can be initiated and structurally
about 1 A further from the dioxygen moiety than resolved. Radiolytic reduction of the oxy-ferrous
in the wild-type oxy-ferrous counterpart. While crystal of CYPlOl results in distinct changes
efficient primary-proton transfer is observed in localized in the distal pocket. These are per-
this mutant in radiolytic studies, the crucial proton haps best illustrated in overall electron density
transfer step, resulting in water formation and difference maps"^^. Most importantly, following
heterolytic cleavage to form "Compound I," is introduction of an electron to the preformed oxy-
perturbed by the modification of these inter- complex is the appearance of a negative difference
actions. Interestingly, the flip of the Asp251 density localized at the distal oxygen atom of
carbonyl is also seen in the Thr252Ala ferric the iron-bound dioxygen. This suggests that oxy-
structure, although the rotation is enhanced even gen-oxygen bond cleavage has occurred leaving
more (120° vs 90°). behind a single oxygen atom ligated to the heme
The structure of the CYPlOl wild-type iron. The resulting Fe-O bond distance appears
ferrous-dioxygen complex also provides insight shortened to about 1.67 A, consistent with an iron-
into the diminished efficacy of primary proton 0x0 "Fe=0" type structure in analogy with that
delivery in the Asp251Asn mutant, as observed in observed in CcP (1.66 A)^^^, H R P (1.70 Ay^\ and
both room temperature kinetics and EPR annealing catalase (1.76 A) (Protein Databank entry IMQF).
profiles. The "flip" of the Asp251 backbone is syn- In these cases, the iron is seen to move to a
ergistically coupled with the rotation of the Thr252 position slightly above the heme plane. Frustrat-
backbone. In the ferric structure of Asp251Asn, ingly, despite numerous attempts, the resulting
the mutated Asn251 side chain forms new interac- "ferryl-oxo" structure does not have a fully occu-
tions with Asp 182 and Asn255, with the latter pied oxygen atom and could not be precisely
effectively blocking the interaction with the 251 refined without constraints. Nonetheless, the
carbonyl in the "flipped" conformation typical for resulting picture is extremely exciting as it shows
the oxy complex. Examination of the oxy-ferrous that the twice-reduced iron-oxygen complex is
structure of Asp251Asn corroborates this finding, catalytically competent. Indeed, warming the
as neither of the catalytically critical solvent mole- crystal to a point above the glass transition tem-
cules is observed (Schlichting et ai, unpublished). perature reveals a structure with clear positive
The differential solvation of the active site in electron density corresponding to a 5-exo alcohol
the Asn251 and Ala252 mutants is also observed hydroxycamphor product. In the product complex,
in resonance Raman spectra of the ferric cyanide the protein displays the resting state conformation
adducts^"^^. Specifically, while the wild-type with respect to water and protein structure. No
enzyme shows exclusive sensitivity to isotopic "extra" water molecules appear in the active site
exchange in the bent conformer, those of the and the Thr251-Asp252 backbone conformation
168 Thomas M. Makris ef a/.

has moved back to that found in the ferric and complex, prior to its structural resolution described
ferrous camphor-bound complexes. above, suggested a possible role for Thr252 (ref
Further differences between studies performed [92]). During the course of the simulation, the
on frozen solutions and crystals can be attributa- Thr252 hydroxyl, instead of interacting with the
ble to crystal contacts that may have a crucial Gly248 backbone as modeled initially, prefer-
impact on the dynamics of the system, thereby entially participates in a hydrogen-bonding inter-
changing relative rates of subsequent reaction action with the distal oxygen atom. While this
steps. For example, the lack of accumulation of interaction persists throughout the simulation,
[6] in cryoradiolytically reduced frozen solu- no significant movement of the Asp251 residue
tions'*^' ^^ but apparently not in crystals'*^ may be was observed. A more direct role for the con-
related to a crystal contact involving the potas- served acid residue in protonation events has
sium binding site and the carbonyl oxygen atom of very recently been made by Hummer and col-
Tyr96 in the monoclinic crystal form of CYPlOl leagues^"^^. Through the examination of allowable
(Figure 5.5). The hydroxyl group of Tyr96 forms a side chain and water fluctuations, utilizing the
hydrogen bond with the keto group of camphor, ferrous-dioxygen state as a precursor, an interest-
thereby controlling its position, mobility, and ing model for the active-site modulation of proton
thus the distance between the C5 atom of camphor networks emerges. The authors note that the
and the reactive oxygen species. movement of the Asp251 side chain toward heme-
Given the dynamic nature of both intermediates bound dioxygen is almost unrestricted and that the
and the isomerization of amino acids of side chains correlated movement of this residue and WAT901
occurring during catalysis, it is not surprising that provides effective proton connectivity between
a number of computational studies have provided these two moieties. The addition of one new water
subtly different structural mechanisms for the molecule links this local network to the surface of
acid-alcohol pair in P450 catalysis. Molecular the protein and bulk solvent. While such a
d3niamics simulations of the ferrous-dioxygen dramatic rotation of the Asp251 side chain was

molecule 2

molecule 1

Figure 5.5. A crystal contact at the potassium binding site may influence the dynamics of active-site moieties,
particularly Tyr96, and thus indirectly, camphor.
Activation of Molecular Oxygen by Cytochrome P450 169

not observed in static crystal structures of resting are present is unclear. Similar differences in the
or dioxygen bound form, such localized confor- relative positioning or flexibility of the threonine
mational changes can obviously significantly have likewise been inferred in the examination of
modulate proton delivery^^^ lysine mutants in various P450s^^^' ^^^. The con-
nectivity of the threonine hydroxyl in P450nor
with the I-helix main chain is retained, however,
4.4. Mechanism-Based through a solvent molecule, WAT46, to the pep-
Specificity of Proton tide carbonyl of the i-4 residue Ala239.
Meanwhile, the threonine is involved in another
Transfer
water channel leading to the distal face, and
Conformational changes in the distal pocket finally bulk solvent. Such a change in polarity
reorientation can result in drastic effects on the of the distal pocket between P450nor and CYPlOl
concerted delivery of protons to heme-bound is transmitted even into the observed coordina-
dioxygen. Coupled with these movements is the tion geometry of bound NO and isocyanide
mechanism-based specificity of proton transfer as complexes^^^' ^^^.
determined with the identity of the substrate to Mutagenesis of the Thr243 residue in P450nor,
the specific P450. The site directed transforma- like the Thr252, results in a drastic reduction
tion of various heme enzymes to accommodate of activity, in this case the NADH dependent
novel reactivities, both in substrate recognition reduction of NO to N2O (ref [265]). Nonetheless,
and through mechanistic inferences of proton structural analysis of several mutants at this
delivery, has led to a successful redesign of pro- position show very minor perturbations of a plau-
tein peroxo-iron chemistries. These have been sible proton delivery network^^^. Thus, while the
documented in histidine- and thiolate-ligated per- appearance of new water molecules in mutant
oxidases^^^"^^^. As a feat in reengineering, the forms is certainly correlated with a change in a
directed alteration of reactivity is perhaps best evi- possible proton delivery network, it is difficult to
denced in the redesign of the dioxygen carrier eliminate the possibility that the residue could be
myoglobin, normally defunct with regards to het- functioning by facilitating electron transfer from
eroatom oxygen transfer reactions, to evolve suc- NADH, as the enzyme directly catalyzes electron
cessful monooxygenation chemistries utilizing transfer from pyridine dinucleotide to the active
both hydrogen peroxide^^^' 252-254 ^^^ dioxygen^^^ site. This potential alteration in the role of a con-
as the source of oxidant. Within the family of served threonine in proton delivery is likewise
P450 enzymes, two examples of altered reactivity, supported by the absence of a conserved acid
that of nitric oxide reductase and peroxygenase residue, Ala at position 242, in P450nor, and may
activities, illuminate mechanisms of active-site reflect an alteration of the distal pocket network to
complementarity to a specific ligand. accommodate its unique mechanism. Based on
The role of proton transfer in P450-catalyzed its closer proximity to the NO binding site and
nitric oxide reduction has been implicated in a similar abolishment of activity upon mutation,
number of computationaP^^ and experimental another hydroxyl, Ser286, may instead fulfill
studies^^^, even though a complete mechanistic the role of the conserved P450 hydroxyl^^^' ^^'^.
assignment of particular nitrogen ligated states Unlike the Thr243 mutants, structural analysis of
remains elusive. Structural analysis of the various Ser286 mutants shows drastic changes
P450nor active site from Fusarium oxysporum by in hydrogen-bonding networks through the sys-
Shiro and colleagues reveal that, unlike CYPlOl, tematic destabilization of water networks^^^' ^^^.
the active site is comprised of a number of The effects of these water networks involv-
hydrophilic residues^^^"^^^. Although the "con- ing Ser286 appear critical, as even the minor
served" threonine is localized in the distal pocket Ser286Thr mutation results in a rotamer con-
(Thr243), the orientation of its side-chain figuration, which destabilizes the water and
hydroxyl is subtly different, with the alcohol side results in a loss of activity. Thus, the active-site
chain pointing toward the dioxygen-binding site. proton delivery networks of these heme-oxygen
Whether this reflects a more natural rotamer con- enzymes could be very plastic with the precise
figuration not seen in the other P450s structurally dynamical picture of hydrogen ion delivery not yet
examined, or if significantly different mechanisms revealed.
170 Thomas M. Makris et ai.

While P450s are typically poor in the utiliza- the ferric-peroxo anion and ferric-hydroperoxo
tion of hydrogen peroxide as a dioxygen/elec- intermediates. In the exogenous oxidant driven
tron/proton surrogate in monooxygenation pathway, an archaeal P450 allowed facile obser-
chemistry, a number of P450s have been shown to vation of the formation and breakdown of
utilize the peroxygenase pathway in the hydroxy- the "Compound I" ferryl-oxo state. Yet much
lation of fatty acids^^^"^^^ Evaluation of these remains. Stabilization and characterization of the
enzymes is important in order to assess the "Compound I" state in the dioxygen reaction has
isozyme dependence of various P450s in the uti- not yet been achieved. With the ability to separate,
lization of the peroxide shunt pathway in catalytic through time and temperature, the population of
processes. In order to accommodate this funda- multiple "active oxygen" intermediates in P450
mental difference in active-site chemistry with catalysis, it remains to precisely define the reac-
these P450s, as in this case both protons and elec- tivity profiles of each state and thereby realize
trons are delivered to the ferriheme moiety, the a mapping of observed in vivo metabolic profiles
enzyme is devoid of the conserved acid-alcohol to specific states in the reaction cycle. An over-
pair. For example, in P450 CYP152A1 these posi- riding revelation has been the subtle way in which
tions in the active site are occupied by Arg242 and Nature controls the reactivity of atmospheric
Pro243 respectively. However, mutagenesis has dioxygen, electrons, and transition metal through
succinctly demonstrated their critical role in both delicate hydrogen-bonding interactions. Thus,
fatty acid binding and in the hydrogen peroxide in a Periclesian control of mechanism, the cyto-
mediated hydroxylation reaction^^^. Likewise, chromes P450 utilize a variety of proton pathways
both the presence and positioning of a substrate to finely tune this versatile catalyst for critical
carboxylate is similarly crucial presumably in physiological processes.
an electrostatic interaction with this conserved
arginine in peroxygenase^^^' ^'^^. Recent crystallo-
graphic structure determination of this P450 Acknowledgments
by Shiro and coworkers have suggested a very
elegant mechanism of substrate mediated peroxy- This work was supported by National Institutes
genase catalysis^^^. The fatty acid carboxylate is of Health grants R37 GM31756 and GM33775
found in a similar orientation as the Glul83
(SGS) and the German Research Foundation (IS)
residue in chloroperoxidase^^^ and thereby seems
grant BIO2-CT942060 (IS).
to serve as the catalyst for O-O bond scission
of hydrogen peroxide^^'. While the conserved
histidine found in chloroperoxidase is absent in
the peroxygenase distal pocket, the role of modu-
References
lating the basicity of the substrate carboxylate
may be served by Arg242 (ref [275]). 1. Sawyer, DT. (1991). Oxygen Chemistry. Oxford
University Press, Oxford, p. 223.
2. Ortiz De Montellano, P.R. (1986). Cytochrome P450:
Structure, Mechanism, and Biochemistry. Plenum
4.5. Summary Press, New York, p. 556.
3. Ortiz De Montellano, PR. (1995). CytochromeP450:
The rich mechanistic enzymology of the Structure, Mechanism, and Biochemistry, 2nd edn.
cytochrome P450s has occupied chemists, bio- Plenum Press, New York, p. 652.
chemists, pharmacologists, and toxicologists for 4. Griffin, B.W., J.A. Peterson, and R.W. Estabrook
over three decades. Are we near to a detailed (1979). Cytochrome P450: Biophysical proper-
molecular understanding? We have attempted to ties and catalytic function. In D. Dolphin (ed.).
Porphyrins, Vol. 7. Academic Press, New York,
convey in this chapter of Cytochrome P450 the
pp. 333-375.
recent discoveries that fill many of the lacunas in
5. Makris, TM., I.G. Denisov, and S.G. Sligar (2003).
our understanding of P450-catalyzed substrate Haem-oxygen reactive intermediates: Catalysis by
oxidations. We now have a precise three- the two-step. Biochem. Soc. Trans. 31, 516-519.
dimensional structure of the ferrous-oxygenated 6. Mason, H.S. (1957). Mechanisms of oxygen metabo-
state, and ample spectroscopic characterization of \ism. Adv. Enzymol. Relat. Subj. Biochem. 19, 79-233.
Activation of IVIolecuiar Oxygen by Cytochrome P450 171

7. Saito, Y., O. Hayaishi, and S. Rothberg (1957). Porphyrin Handbook, Vol. 4. Academic Press,
Oxygenases. IV. Enzymic formation of 3-hydroxy- NewYork, pp. 17^0.
L-kynurenine from L-kynurenine. J. Biol. Chem. 22. Poulos, TL. (2000). Peroxidase and cytochrome
229, 921-933. P450 structures. In K.M. Kadish, K.M. Smith, and
8. Winfield, M.E. (1965). Mechanisms of oxygen R. Guilard (eds). Porphyrin Handbook, Academic
uptake: The autoxidation of myoglobin and of Press, NewYork, pp. 189-218.
reduced cyanocobaltates and their significance to 23. Scheldt, W.R. (2000). Systematics of the stereo-
oxidase reactions. In T.E. King, H.S. Mason, and chemistry of porphyrins and metalloporphyrins.
M. Morrison (eds), Oxidases and Related Redox In K.M.S. Kadish, K.M. Smith, and R. Guilard
Systems, Vol. 1. Wiley, New York, pp. 115-130. (eds). Porphyrin Handbook, Vol. 3. Academic Press,
9. Que, L. and Y. Watanabe (2001). Bioinorganic NewYork, pp. 49-112.
chemistry—oxygenase pathways: Oxo, peroxo, and 24. Watanabe, Y. (2000). High-valent intermediates.
superoxo. Science 292, 651-653. In K.M.S. Kadish, K.M. Smith, and R. Guilard
10. Miner, A.N.P., E.L. Raven, R.N.F. Thorneley, (ed.). Porphyrin Handbook, Vol. 4. Academic Press,
E Garcia-Canovas, and J.N. Rodrigues-Lopez NewYork, pp. 97-117.
(2002). Mechanisms of compound I formation in 25. Weiss, R., A. Gold, A.X. Trautwein, and J. Terner
heme peroxidases. J. Inorg. Biochem. 91, 27-34. (2000). High-valent iron and manganese complexes
11. Erman, J.E. and L.B. Vitello (2002). Yeast cyto- of porphyrins and related macrocycles. In
chrome c peroxidase: Mechanistic studies via protein K.M.S. Kadish, K.M. Smith, and R. Guilard (eds).
engineering. Biochim. Biophys. Acta 1597, 193-220. Porphyrin Handbook, Vol. 4. Academic Press,
12. Ortiz De Montellano, RR. and A. Wilks (2001). NewYork, pp. 65-96.
Heme oxygenase structure and mechanism. Adv. 26. Selke, M., M.E Sisemore, R.Y.N. Ho, D.L. Wertz,
Inorg. Chem. 51, 359-407. and J.S. Valentine (1997). Dioxygen activation by
13. Fujii, H., X.H. Zhang, T. Tomita, M. Ikeda-Saito, iron complexes. The search for reactive intermedi-
and T. Yoshida (2001). A role for highly conserved ates. J. Mol. Catal. A 111, 71-82.
carboxylate, aspartate-140, in oxygen activation and 27. Valentine, A.M., P. Tavares, A.S. Pereira,
heme degradation by heme oxygenase-1. J. Am. R. Davydov, C. Krebs, B.M. Hoffman et al. (1998).
Chem. Soc. 123, 6475-6484. Characterization of a mixed-valent Fe(III)Fe(IV)
14. Nicholls, R, I. Fita, and RC. Loewen (2001). form of intermediate Q in the reaction cycle of
Enzymology and structure of catalases, Adv. Inorg. soluble methane monooxygenase, an analog of
Chem. 51, 51-106. intermediate X in ribonucleotide reductase R2
15. Groves, J.T. and C.C.-Y Wang (2000). Nitric oxide assembly. J ^m. Chem. Soc. 120, 2190-2191.
synthase: Models and mechanism. Curr. Opin. 28. Valentine, J.S., D.L. Wertz, T.J. Lyons, L.-L. Liou,
Chem. Biol. 4, 687-695. J.J. Goto, and E.B. Gralla (1998). The dark side of
16. Noordermeer, M.A., G.A. Veldink, and dioxygen biochemistry. Curr Opin. Chem. Biol. 2,
J.F. Vliegenthart (2001). Fatty acid hydroperoxide 253-262.
lyase: A plant cytochrome p450 enzyme involved in 29. Wertz, D.L. and J.S. Valentine (2000). Nucleo-
wound healing and pest resistance. ChemBioChem philicity of iron-peroxo porphyrin complexes.
2, 494-504. Struct. Bond (Berlin) 97, 37-60.
17. Van der Donk, W.A., A.L. Tsai, and R.J. Kulmacz 30. Stillman, M. (2000). Formation and electronic
(2002). The cyclooxygenase reaction mechanism. properties of ring-oxidized and ring-reduced radical
Biochemistry 41, 15451-15458. species of the phthalocyanines and porphyrins.
18. Dunford, H.B. and J.S. Stillman (1976). On the J. Porph. Phtalocya. 4, 374-376.
function and mechanism of the action of peroxi- 31. Dolphin, D.E and R.H. Felton (1974). The biochem-
dases. Coord Chem. Rev 19, 187-251. ical significance of porphyrin IT cation radicals.
19. Siegbahn, RE. (2003). Mechanisms of metallo- Accounts Chem. Res. 1, 26-32.
enzymes studied by quantum chemical methods. 32. Chen, K., M. Costas, and L. Que (2002). Spin state
Q. Rev Biophys. 36, 91-145. tuning of non-heme iron-catalyzed hydrocarbon
20. Kincaid, J.R. (2000). Resonance Raman spectra oxidations: Participation of Fe(III)-OOH and
of heme proteins and model compounds. In Fe(V)=0 intermediates. J. Chem. Soc. Dalton
K.M. Kadish, K.M. Smith, and R. Guilard (eds). Trans., 672-679.
Porphyrin Handbook, Vol. 7. Academic Press, 33. Chen, K., M. Costas, J. Kim, A.K. Tipton, and
NewYork, pp. 225-291. L. Que, Jr. (2002). Olefin cw-dihydroxylation
21. Groves, J.T., K. Shalyaev, and J. Lee (2000). versus epoxidation by non-heme iron catalysts: Two
Oxometalloporphyrins in oxidative catalysis. faces of an Fe(III)-OOH coin. J. Am. Chem. Soc.
In K.M. Kadish, K.M. Smith, and R. Guilard (eds). 124, 3026-3035.
172 Thomas M. Makris et al.

34. Salem, I.A., M. El-Maazawi, and A.B. Zaki (2000). 47. Groves, J.T. and Y. Han (1995). Models and mecha-
Kinetics and mechanisms of decomposition reac- nisms of cytochrome P450 action. In PR. Ortiz de
tion of hydrogen peroxide in presence of metal Montellano (ed.). Cytochrome P450. Structure,
complexes. Int. J. Chem. Kinet. 32, 643-666. Mechanism, and Biochemistry, 2nd edn. Plenum
35. Raag, R., S.A. Martinis, S.G. Shgar, andT.L. Poulos Press, New York, pp. 3-48.
(1991). Crystal structure of the cytochrome 48. Watanabe, Y and J.T. Groves (1992). Molecular
P-450cam active site mutant Thr252Ala. Biochemistry mechanism of oxygen activation by P-450. In D.S.
30, 11420-11429. Sigman (ed.). The Enzymes, Vol. 20. Academic
36. Raag, R. andT.L. Poulos (1991). Crystal structures Press, San Diego, CA, pp. 405-452.
of cytochrome P-450cam complexed with 49. Denisov, I.G., T.M. Makris, and S.G. Sligar
camphane, thiocamphor, and adamantane: Factors (2001). Cryotrapped reaction intermediates of cyto-
controlling P-450 substrate hydroxylation. chrome P450 studied by radiolytic reduction with
Biochemistry 30, 2674-2684. phosphorus-32. J: Biol. Chem. 216, 11648-11652.
37. Loida, PJ. and S.G. Sligar (1993). Engineering 50. Denisov, I.G., S.C. Hung, K.E. Weiss, M.A. Mclean,
cytochrome P-450cam to increase the stereo- Y. Shiro, S.Y Park et al. (2001). Characterization of
specificity and coupling of aliphatic hydroxylation. the oxygenated intermediate of the thermophilic
Protein Eng. 6, 207-212. cytochrome P450 CYP119. J. Inorg. Biochem. 87,
38. Aikens, J. and S.G. Sligar (1994). Kinetic solvent 215-226.
isotope effects during oxygen activation by 51. Ortiz De Montellano, PR. (1998). Heme oxy-
cytochrome P-450cam. J. Am. Chem. Soc. 116, genase mechanism: Evidence for an electrophilic,
1143-1144. ferric peroxide species. Accounts Chem. Res. 31,
39. Vidakovic, M., S.G. Sligar, H. Li, and T.L. Poulos 543-549.
(1998). Understanding the role of the essential 52. Yoshida, T. and C.T. Migita (2000). Mechanism of
Asp251 in cytochrome p450cam using site-directed heme degradation by heme oxygenase. J. Inorg.
mutagenesis, crystallography, and kinetic solvent Biochem. 82,33-41.
isotope effect. Biochemistry 37, 9211-9219. 53. Davydov, R., V Kofman, H. Fujii, T. Yoshida,
40. Davydov, R., I.D.G. Macdonald, T.M. Makris, M. Ikeda-Saito, and B.M. Hoffman (2002). Cataly-
S.G. Sligar, and B.M. Hoffman (1999). EPR and tic mechanism of heme oxygenase through EPR
ENDOR of catalytic intermediates in cryoreduced and ENDOR of cryoreduced oxy-heme oxygenase
native and mutant oxy-cytochromes P450cam: and its Asp 140 mutants. J. Am. Chem. Soc. 124,
Mutation-induced changes in the proton delivery 1798-1808.
system. J. Am. Chem. Soc. 121, 10654-10655. 54. Jones, P and H.B. Dunford (1977). On the mecha-
41. Davydov, R., T.M. Makris, V. Kofman, D.E. Werst, nism of compound I formation from peroxidases
S.G. Sligar, and B.M. Hoffman (2001). Hydroxyla- and catalases. J. Theor Biol. 69, 457-470.
tion of camphor by reduced oxy-cytochrome 55. Poulos, TL. and J. Kraut (1980). The stereo-
P450cam: Mechanistic implications of EPR and chemistry of peroxidase catalysis. J.Biol. Chem.
ENDOR studies of catalytic intermediates in 255,8199-8205.
native and mutant enzymes. J. Am. Chem. Soc. 123, 56. Loew, G.H. and D.L. Harris (2000). Role of the
1403-1415. heme active site and protein environment in struc-
42. Schlichting, I., J. Berendzen, K. Chu, A.M. Stock, ture, spectra, and function of the cytochrome P450s.
S.A. Maves, D.E. Benson et al. (2000). The catalytic Chem. Rev. 100,407-419.
pathway of cytochrome p450cam at atomic resolu- 57. Erman, J.E., L.B. Vitello, M.A. Miller, A. Shaw,
tion. Science 287, 1615-1622. K.A. Brown, and J. Kraut (1993). Histidine 52
43. Gennis, R.B. (1998). How does cytochrome oxidase is a critical residue for rapid formation of cyto-
pump protons? Proc. Natl. Acad. Sci. USA 95, chrome c peroxidase compound I. Biochemistry 32,
12747-12749. 9798-9806.
44. Wikstrom, M. and M.I. Verkhovsky (2002). Proton 58. Rodrigues-Lopez, J.N., D.J. Lowe, J. Hernandez-
translocation by cytochrome c oxidase in different Ruiz, A.N.P Hiner, F. Garcia-Canovas, and
phases of the catalytic cycle. Biochim. Biophys. Acta R.N.F. Thorneley (2001). Mechanism of reaction of
1555, 128-132. hydrogen peroxide with horseradish peroxidase:
45. Dunford, H.B. (1999). Heme Peroxidases. Wiley, Identification of intermediates in the catalytic cycle.
New York, p. 507. J.Am. Chem. Soc. 123, 11838-11847.
46. Groves, J.T and Y. Watanabe (1988). Reactive iron 59. Mirza, S.A., B. Bocquet, C. Robyr, S. Thomi, and
porphyrin derivatives related to the catalytic cycles A.F. Williams (1996). Reactivity of the coordinated
of cytochrome P-450 and peroxidase. Studies of the hydroperoxo ligand. Inorg. Chem. 35, 1332-1337.
mechanism of oxygen activation. J. Am. Chem. Soc. 60. Primus, J.-L., S. Grunenwald, P.-L. Hagedoorn,
110, 8443-8452. A.-M. Albrecht-Gary, D. Mandon, and C. Veeger
Activation of Molecular Oxygen by Cytochrome P450 173

(2002). The nature of the intermediates in the reac- heterolytic versus homolytic O-O bond cleavage.
tions of Fe(III)- and Mn(III)-microperoxidase-8 Arch. Biochem. Biophys. 317, 471-478.
with H2O2: A rapid kinetics study. J. Am. Chem. Soc. 73. Barr, D.R, M.V. Martin, ER Guengerich, and
124, 1214-1221. R.R Mason (1996). Reaction of cytochrome P450
61. Dunford, H.B. (1991). Horseradish peroxidase: with cumene hydroperoxide: ESR spin-trapping evi-
Structure and kinetic properties. In J. Everse, dence for the homolytic scission of the peroxide
K.E. Everse, and M.B. Grisham (eds). Peroxidases 0 - 0 bond by ferric cytochrome P450 1A2. Chem.
in Chemistry and Biology, Vol. 2. CRC Press, Boca Res. Toxicol. 9,318-325.
Raton, FL, pp. 2-24. 74. Adams, RA. and J. Louw (1995). Dioxygen bond
62. Gerber, N.C. and S.G. Sligar (1994). A role for scission and heme degradation in hemoproteins: A
Asp-251 in cytochrome P-450cam oxygen activa- kinetic study of chemical model systems using fer-
tion. J. Biol. Chem. 269, 4260^266. rimyoglobin and hemepeptide: Non-hemepeptide
63. Ortiz De Montellano, RR. and J.J. De Voss (2002). complexes as catalysts for 'peroxidasic' reduction of
Oxidizing species in the mechanism of cytochrome hydrogen peroxide. J. Chem. Soc. Perkin Trans. 2,
P450. Nat. Prod Rep. 19, 477-493. 1683-1690.
64. Vaz, A.D.N., S.J. Pernecky, G.M. Raner, and 75. Shimizu, T, Y Murakami, and M. Hatano (1994).
M.J. Coon (1996). Peroxo-iron and oxenoid-iron Glu318 and Thr319 mutations of cytochrome P450
species as alternative oxygenating agents in 1A2 remarkably enhance homolytic O-O cleavage
cytochrome P450-catalyzed reactions: Switching of alkyl hydroperoxides. An optical absorption
by threonine-302 to alanine mutagenesis of spectral study J. Biol. Chem. 269, 13296-13304.
cytochrome P450 2B4. Proc. Natl. Acad. Sci. USA 76. White, R.E., S.G. Sligar, and M.J. Coon (1980).
93, 4644-4648. Evidence for a homolytic mechanism of peroxide
65. Vaz, A.D.N, and M.J. Coon (1994). On the mecha- oxygen-oxygen bond cleavage during substrate
nism of action of cytochrome P450: Evaluation of hydroxylation by cytochrome P-450. J. Biol. Chem.
hydrogen abstraction in oxygen-dependent alcohol 255, 11108-11111.
oxidation. Biochemistry 33, 6442-6449. 77. Kuo, C.-L., G.M. Raner, A.D.N. Vaz, and M.J. Coon
66. Vaz, A.D.N., D.K McGinnity, H.M. Peng, G.M. (1999). Discrete species of activated oxygen yield
Raner, C.L. Kuo, K.R Vatsis et al. (1997). different cytochrome P450 heme adducts from alde-
Mechanism of oxidations involving the cytochrome hydes. Biochemistry'i^, 10511-10518.
P450 peroxo-iron complex as probed by site- 78. Barr, D.R and R.R Mason (1995). Mechanism
specific mutagenesis. FASEB J. 11, P32. of radical production from the reaction of cyto-
67. Vaz, A.D.N., D.K McGinnity, P Zarehbin-Irani, chrome c with organic hydroperoxides. An ESR
M.J. Coon, J.P Salaun, D.R. Koop et al (1997). spin trapping investigation. J. Biol. Chem. 270,
Epoxidation of olefins by cytochrome P450: 12709-12716.
Possible role for hydroperoxo-iron as the direct 79. Lee, W.A. and T.C. Bruice (1985). Homolytic and
oxidant. FASEB J. 11, P207. heterolytic oxygen-oxygen bond scissions accom-
68. Yun, C.-H., G.R Miller, and RR Guengerich (2000). panying oxygen transfer to iron(III) porphyrins by
Rate-determining steps in phenacetin oxidations by percarboxylic acids and hydroperoxides. A mecha-
human cytochrome P450 1A2 and selected mutants. nistic criterion for peroxidase and cytochrome
Biochemistry 39, 11319-11329. ?A50.J.Am. Chem. Soc. 107, 513-514.
69. Yun, C.-H., H.S. Lee, H.-Y Lee, S.-K. Yim, 80. Yoshioka, S., S. Takahashi, K. Ishimori, and
K.-H. Kim, E. Kim et al. (2003). Roles of human I. Morishima (2000). Roles of the axial push effect
liver cytochrome P450 3A4 and 1A2 enzymes in the in cytochrome P450cam studied with the site-
oxidation of myristicin. Toxicol. Lett. 137, 143-150. directed mutagenesis at the heme proximal site.
70. Guengerich, ER (2002). Rate-limiting steps in J. Inorg. Biochem. 81, 141-151.
cytochrome P450 catalysis. Biol. Chem. 383, 81. Dawson, J.H. and M. Sono (1987). Cytochrome
1553-1564. P-450 and chloroperoxidase—^thiolate-ligated
71. Allentoff, A.J., J.L. Bolton, A. Wilks, heme enzymes—spectroscopic determination
J.A. Thompson, and RR. Ortiz de Montellano of their active-site structures and mechanistic
(1992). Heterolytic versus homolytic peroxide implications of thiolate ligation. Chem. Rev. 87,
bond-cleavage by sperm whale myoglobin and 1255-1276.
myoglobin mutants. J. Am. Chem. Soc. 114, 82. Karuzina, I.I., VG. Zgoda, G.R Kuznetsova, N.E
9744-9749. Samenkova, and A.I. Archakov (1999). Heme and
72. Correia, M.A., K. Yao, A.J. Allentoff, apoprotein modification of cytochrome P450 2B4
S.A. Wrighton, and J.A. Thompson (1995). Inter- during its oxidative inactivation in monooxygenase
actions of peroxyquinols with cytochromes P450 reconstituted system. Free Radic. Biol. Med. 26,
2B1, 3A1, and 3A5: Influence of the apoprotein on 620-632.
174 Thomas M. Makris ef al.

83. Lehnert, N., R.Y.N. Ho, L. Que, Jr., and mechanism for activation of peroxidases:
E.L Solomon (2001). Spectroscopic properties and Cooperative catalytic contributions from the ion
electronic structure of low-spin Fe(III)-alkylperoxo and microsolvent water. J. Phys. Chem. A 102,
complexes: Homolytic cleavage of the O-O bond. 10380-10384.
J.Am. Chem. Soc. 123, 8271-8290. 96. Loew, G.H. and M. Dupuis (1996). Structure of a
84. Lehnert, N., R.Y.N. Ho, L. Que, Jr., and model transient peroxide intermediate of peroxi-
E.I. Solomon (2001). Electronic structure of high- dases by an Ab Initio methods. J. Am. Chem. Soc.
spin iron(III)-alkylperoxo complexes and its rela- 118, 10584-10587.
tion to low-spin analogues: Reaction coordinate 97. Loew, G. and M. Dupuis (1997). Characterization
of 0 - 0 bond homolysis. J. Am. Chem. Soc. 123, of a resting state model of peroxidases by Ab Initio
12802-12816. methods: Optimized geometries, electronic struc-
85. Lehnert, N., F. Neese, R.YN. Ho, L. Que, Jr., and tures, and relative energies of the sextet, quartet,
E.I. Solomon (2002). Electronic structure and reac- and doublet spin states. J. Am. Chem. Soc. 119,
tivity of low-spin Fe(III)-hydroperoxo complexes: 9848-9851.
Comparison to activated bleomycin. J. Am. Chem. 98. Blomberg, M.R.A., RE.M. Siegbahn, G.T. Babcock,
Soc. 124, 10810-10822. and M. Wikstroem (2000). O-O bond split-
86. Lehnert, N., K. Fujisawa, and E.I. Solomon (2003). ting mechanism in cytochrome oxidase. J. Inorg.
Electronic structure and reactivity of high-spin iron- Biochem. 80, 261-269.
alkyl- and -pterinperoxo complexes. Inorg. Chem. 99. Lu, Y, S.M. Berry, and T D Pfister (2001).
42,469-481. Engineering novel metalloproteins: Design of
87. Chen, K., and L. Que, Jr. (2001). Stereospecific metal-binding sites into native protein scaffolds.
alkane hydroxylation by non-heme iron catalysts: Chem. Rev. 101, 3047-3080.
Mechanistic evidence for an Fe(V)=0 active 100. Watanabe, Y (2002). Construction of heme
species. J. Am. Chem. Soc. 123, 6327-6337. enzymes: Four approaches. Curr Opin. Chem.
88. Bassan, A., M.R.A. Blomberg, RE.M. Siegbahn, Biol. 6,208-216.
and L. Que, Jr. (2002). A density functional study 101. Gerber, N.C. and S.G. Sligar (1992). Catalytic
of O-O bond cleavage for a biomimetic non-heme mechanism of cytochrome P-450: Evidence
iron complex demonstrating an Fe(V)-intermediate. for a distal charge relay. J. Am. Chem. Soc. 114,
J.Am. Chem. Soc. 124, 11056-11063. 8742-8743.
89. Harris, D.L. and G.H. Loew (1996). Identification 102. Shimada, H., Y Watanabe, M. Imai, R. Makino,
of putative peroxide intermediates of peroxidases by H. Koga, T Horiuchi et al. (1991). The role of thre-
electronic structure and spectra calculations. J. Am. onine 252 in the oxygen activation by cytochrome
Chem. Soc. 118, 10588-10594. P-450cam: Mechanistic studies by site-directed
90. Loew, G.H., J.R. Collins, and FU Axe (1989). mutagenesis. In L.I. Simandi (ed.), Dioxygen
Theoretical investigation of a proposed [His 175- Activation and Homogeneous Catalytic Oxida-
Asp235-Trpl91] proton transfer system in cyto- tion. Elsevier Science Publishers, Amsterdam,
chrome c peroxidase. Int. J. Quantum. Chem. 16, pp.3136-319.
199-209. 103. Shimada, H., R. Makino, M. Unno, T Horiuchi, and
91. Collins, J.R., R Du, and G.H. Loew (1992). Y Ishimura (1994). Proton and electron transfer
Molecular dynamics simulations of the resting and mechanism in dioxygen activation by cytochrome
hydrogen peroxide-bound states of cytochrome P450cam. In M.C. Lechner and J. Libbey (eds).
c peroxidase. Biochemistry 31, 11166-11174. Cytochrome P450. 8th International Conference.
92. Harris, D.L. and G.H. Loew (1994). A role for Eurotext, Paris, pp. 299-306.
Thr-252 in cytochrome-P450cam oxygen activation. 104. Shimada, H., S.G. Sligar, H. Yeom, H. and
J Am. Chem. Soc. 116, 11671-11674. Y Ishimura (1997). Heme monooxygenases. A
93. Harris, D.L. and G.H. Loew (1996). Investigation chemical mechanism for cytochrome P450 oxygen
of the proton-assisted pathway to formation of the activation. Catalysis by Metal Complexes, 19,
catalytically active, ferry 1 species of P450s by 195-221.
molecular dynamics studies of P450eryF. J. Am. 105. Gunsalus, I.C, TC. Pederson, and S.G. Sligar
Chem. Soc. 118, 6377-6387. (1975). Oxygenase-catalyzed biological hydroxy-
94. Harris, D.L. and G.H. Loew (1998). Theoretical lations. Ann. Rev. Biochem. 44, 377^07.
investigation of the proton assisted pathway to 106. Guengerich, F.P. (1991). Reactions and signifi-
formation of cytochrome P450 compound I. J. Am. cance of cytochrome P-450 enzymes. J. Biol.
Chem. Soc. 120, 8941-8948. Chem. 266, 10019-10022.
95. Woon, D.E. and G.H. Loew (1998). An Ab 107. Hedegaard, J. and I.C. Gunsalus (1965). Mixed
Initio model system investigation of the proposed function oxidation. IV An induced methylene
Activation of Molecular Oxygen by Cytochrome P450 175

hydroxylase in camphor oxidation. J. Biol. Chem. 120. Eisenstein, L., P Debey, and P Douzou (1977).
240, 4038^043. P450cam: Oxygenated complexes stabilized at low
108. Conrad, H.E., K. Lieb, and l.C. Gunsalus (1965). temperature. Biochem. Biophys. Res. Commun. 11,
Mixed function oxidation. III. An electron trans- 1377-1383.
port complex in camphor ketolactonization. J. Biol. 121. Bonfils, C , P Debey, and P Maurel (1979).
Chem. 240, 4029^037. Highly-purified microsomal P-450: The oxyferro
109. Katagiri, M., B.N. Ganguli and l.C. Gunsalus intermediate stabilized at low temperature.
(1968). A soluble cytochrome P-450 functional in Biochem. Biophys. Res. Commun. 88, 1301-1307.
methylene hydroxylation. J. Biol. Chem. 243, 122. Lambeir, A.M., C.A. Appleby, and H.B. Dunford
3543-3546. (1985). The formation and decay of the oxyferrous
110. Estabrook, R.W., A.G. Hildebrandt, J. Baron, forms of the cytochromes P-450 isolated from
K.J. Netter, and K. Leibman (1971). New spectral Rhizobium japonicum. Rapid spectral scan and
intermediate associated with cytochrome P-450 stopped flow studies. Biochim. Biophys. Acta 828,
function in liver microsomes. Biochem. Biophys. 144-150.
Res. Commun. 42, 132-139. 123. Lipscomb, J.D., S.G. Sligar, M.J. Namtvedt,
111. Estabrook, R.W., J. Baron, and A. Hildebrandt and I.e. Gunsalus (1976). Autooxidation and
(1971). New spectral species associated with hydroxylation reactions of oxygenated cytochrome
cytochrome P-450 in liver microsomes. Chem. P-450cam. ^ 5/0/. Chem. 251, 1116-1124.
Biol. Interact. 3, 260-261. 124. Sligar, S.G., J.D. Lipscomb, PG. Debrunner, and
112. SHgar, S.G. (1976). Coupling of spin, substrate, and I.e. Gunsalus (1974). Superoxide anion production
redox equilibria in cytochrome P450. Biochemistry by the autoxidation of cytochrome P450cam.
15, 5399-5406. Biochem. Biophys. Res. Commun. 61, 290-296.
113. Guengerich, F.R and W.W. Johnson (1997). 125. Hu, S.Z., A.J. Schneider, and J.R. Kincaid (1991).
Kinetics of ferric cytochrome P450 reduction by Resonance Raman studies of oxycytochrome
NADPH-cytochrome P450 reductase: Rapid reduc- P450cam—effect of substrate structure on v(O-O)
tion in the absence of substrate and variations and v{VQ-0^).J.Am. Chem. Soc. 113, 4815^822.
among cytochrome P450 systems. Biochemistry 126. Macdonald, I.D.G., S.G. SHgar, J.R Christian,
36, 14741-14750. M. Unno, and P.M. Champion (1999). Identification
114. Sharrock, M., PG. Debrunner, C. Schulz, of the Fe-O-O bonding mode in oxycytochrome
J.D. Lipscomb, V Marshall, and l.C. Gunsalus P450cam by resonance Raman spectroscopy. J. Am.
(1976). Cytochrome P450cam and its complexes. Chem. Soc. 121, 376-380.
Moessbauer parameters of the heme iron. Biochim. 127. Dawson, J.H. (1996). Heme-containing oxyge-
Biophys. Acta 420, 8-26. nases. Chem. Rev. 96, 2841-2888.
115. Schulz, C.E., R. Rutter, J.T. Sage, PG. Debrunner, 128. Hanson, L.K., W.A. Eaton, S.G. Sligar,
and L.P. Hager (1984). Moessbauer and electron I.e. Gunsalus, M. Gouterman, and C.R. Connell
paramagnetic resonance studies of horseradish (1976). Origin of the anomalous Soret spectra of
peroxidase and its catalytic intermediates. carboxycytochrome P-450. J. Am. Chem. Soc. 98,
Biochemistry 23, 4743^754. 2672-2674.
116. Prusakov, VE., R.A. Stukan, and R.M. Davydov 129. Hanson, L.K., S.G. Sligar, and l.C. Gunsalus (1977).
(1984). Moessbauer study of the relaxation of Electronic structure of cytochrome P450. Croat.
nonequilibrium hemoglobin states. Biofizika 29, Chem. Acta 49, 237-250.
359-364. 130. Sevrioukova, I.F. and J.A. Peterson (1995).
117. Bangcharoenpaurpong, O., A.K. Rizos, P.M. Reaction of carbon monoxide and molecular
Champion, D. Jollie, and S.G. Sligar (1986). oxygen with P450terp (CYP108) and P450
Resonance Raman detection of bound dioxygen BM-3 (CYP102). Arch. Biochem. Biophys. 317,
in cytochrome P-450cam. J. Biol. Chem. 261, 397-^04.
8089-8092. 131. Bee, N., P. Anzenbacher, E. Anzenbacherova,
118. Goddard, W.A., lii, and B.D. Olafson (1975). A.C.F Gorren, A.W. Munro, and R. Lange (1999).
Ozone model for bonding of an O2 to heme Spectral properties of the oxyferrous complex of
in oxyhemoglobin. Proc. Natl. Acad. Sci. USA 72, the heme domain of cytochrome P450 BM-3
2335-2339. (CYP102). Biochem. Biophys. Res. Commun. 266,
119. Peterson, J.A., Y. Ishimura, and B.W. Griffin 187-189.
(1972). Pseudomonas putida cytochrome P-450. 132. Bee, N., A.C. Gorren, C. Voelker, B. Mayer, and
Characterization of an oxygenated form of the R. Lange (1998). Reaction of neuronal nitric-oxide
hemoprotein. Arch. Biochem. Biophys. 149, synthase with oxygen at low temperature. Evi-
197-208. dence for reductive activation of the oxy-ferrous
176 Thomas M. Makris et al.

complex by tetrahydrobiopterin. J. Biol. Chem. 145. Filizola, M. and G.H. Loew (2000). Probing the
273, 13502-13508. role of protein environment in compound I forma-
133. Sono, M., K.S. Eble, J.H. Dawson, and L.P. Hager tion of chloroperoxidase {C?0).J.Am. Chem. Soc.
(1985). Preparation and properties of ferrous Ill, 3599-3605.
chloroperoxidase complexes with dioxygen, nitric- 146. Loew, G. (2000). Structure, spectra, and function
oxide, and an alkyl isocyanide—spectroscopic of heme sites. Int. J. Quantun. Chem. 11, 54-70.
dissimilarities between the oxygenated forms of 147. Wirstam, M., M.R.A. Blomberg, and RE.M.
chloroperoxidase and cytochrome-P-450. J. Biol. Siegbahn (1999). Reaction mechanism of com-
Chem. 260, 5530-5535. pound I formation in heme peroxidases: A density
134. Shikama, K. (1998). The molecular mechanism functional theory study. J. Am. Chem. Soc. Ill,
of autoxidation for myoglobin and hemoglobin: 10178-10185.
A venerable puzzle. Chem. Rev. 98, 1357-1374. 148. Guengerich, F.P, D.R Ballou, and M.J. Coon
135. Vojtechovsky, J., K. Chu, J. Berendzen, (1976). Spectral intermediates in the reaction
R.M. Sweet, and I. Schlichting (1999). Crystal of oxygen with purified liver microsomal cyto-
structures of myoglobin-ligand complexes at near- chrome P-450. Biochem. Biophys. Res. Commun.
atomic resolution. Biophys. J. 11, 2153-2174. 70,951-956.
136. Miller, M.A., A. Shaw, and J. Kraut (1994). 2.2 A 149. Benson, D.E., K.S. Suslick, and S.G. Sligar
structure of oxy-peroxidase as a model for the (1997). Reduced oxy intermediate observed in
transient enzyme: peroxide complex. Nat. Struct. D251N cytochrome P450(cam). Biochemistry 36,
Biol. 1,524-531. 5104-5107.
137. Schunemann, V, C. Jung, A.X. Trautwein, 150. Yeom, H.Y. and S.G. Sligar (1997). Oxygen activa-
D. Mandon, and R. Weiss (2000). Intermediates in tion by cytochrome P450(BM-3): Effects of mutat-
the reaction of substrate-free cytochrome P450cam ing an active site acidic residue. Arch. Biochem.
with peroxy acetic acid. FEBS Lett. 479, 149-154. Biophys. 331, 209-2\6.
138. Brittain, T., A.R. Baker, C.S. Butler, R.H. Little, 151. Shimizu, N., K. Kobayashi, and K. Hayashi (1984).
D.J. Lowe, C. Greenwood et al. (1997). Reaction of The reaction of superoxide radical with catalase.
variant sperm-whale myoglobins with hydrogen Mechanism of the inhibition of catalase by super-
peroxide: The effects of mutating a histidine oxide radical. J. Biol. Chem., 259, 4414-4418.
residue in the haem distal pocket. Biochem. J. 326, 152. Shimizu, N., K. Kobayashi, and K. Hayashi (1989).
109-115. Kinetics of the reaction of superoxide anion with
139. Tajima, K., K. Ishizu, H. Sakurai, and N.H. Ohya ferric horseradish peroxidase. Biochim. Biophys.
(1986). A possible model of hemoprotein-peroxide Acta 995, 133-137.
complexes formed in an iron-tetraphenylporphyrin 153. Debey, P., E.J. Land, R. Santus, and A.J. Swallow
system. Biochem. Biophys. Res. Commun. 135, (1979). Electron transfer from pyridinyl radicals,
972-978. hydrated electrons, CO2 and O2 to bacterial
140. Tajima, K., J. Jinno, K. Ishizu, H, Sakurai, and cytochrome P450. Biochem. Biophys. Res.
H. Ohya-Nishiguchi (1989). Direct evidence of Commun. 86, 953-960.
heme-tert-butyl peroxide adduct formation demon- 154. Bonfils, C , J.L. Saldana, P Debey, R Maurel,
strated by simultaneous ESR and optical measure- C. Balny, and P Douzou (1979). Fast photo-
ments. Inorg. Chem. 28, 709-715. chemical reactions of cytochrome P450 at subzero
141. Balch, A.L., R.L, Hart, L. Latos-Grazynski, and temperatures. Biochimie, 61, 681-687.
T.G. Traylor (1990). Nuclear magnetic resonance 155. Kobayashi, K. and K. Hayashi (1981). One-
studies of the formation of tertiary alkyl complexes electron reduction in oxyform of hemoproteins.
of iron(III) porphyrins and their reactions with J. Biol. Chem. 256, 12350-12354.
dioxygen. ^ y^m. Chem. Soc. 112, 7382-7388. 156. Kobayashi, K., M. Amano, Y. Kanbara, and
142. Balch, A.L. (1992). The reactivity of spectro- K. Hayashi (1987). One-electron reduction of the
scopically detected peroxy complexes of iron oxyform of 2,4-diacetyldeuterocytochrome
porphyrins. Inorg. Chim. Acta 198-200, 297-307. P450^^^. J. Biol. Chem. 161, 5445-5447.
143. Jinno, J., M. Shigematsu, K. Tajima, H. Sakurai, 157. Davydov, R.M., T Yoshida, M. Ikeda-Saito, and
H. Ohya-Nishiguchi, and K. Ishizu (1991). Coor- B.M. Hoffman (1999). Hydroperoxy-heme oxyge-
dination structure and chemical reactivity of nase generated by cryoreduction catalyzes the
hemoprotein-butyl peroxide complex. Biochem. formation of a-meso-hydroxyheme as detected
Biophys. Res. Commun. 176, 675-681. by EPR and ENDOR. J. Am. Chem. Soc. Ill,
144. Tajima, K., T Edo, K. Ishizu, S. Imaoka, Y. Funae, 10656-10657.
S. Oka et al. (1993). Cytochrome P-450-butyl 158. Davydov, R., R. Kappl, J. Huettermann, and
peroxide complex detected by ESR. Biochem. J. A. Peterson (1991). EPR-spectroscopy of reduced
Biophys. Res. Commun. 191, 157-164. oxyferrous-P450cam. F£'55Le//. 295, 113-115.
Activation of Molecular Oxygen by Cytochrome P450 177

159. Denisov, I.G., T.M. Makris, and S.G. Sligar (2002). spin resonance spectroscopy. Biochim. Biophys.
Cryoradiolysis for the study of P450 reaction Acta 535, 241-247.
intermediates. Meth. Enzymol. 357, 103-115. 171. Symons, M.C.R. and R.L. Petersen (1978).
160. Turner, J.J. and G.C. Pimentel (1963). Krypton The relative electron affinities of the a and p
fluoride: Preparation by the matrix isolation chains of oxyhaemoglobin as a function of pH
technique. Science 140, 974-975. and added inositol hexaphosphate. An electron
161. Ryan, E.A., Y.N. Lwin, J.J. Jackson, and spin resonance study. Biochim. Biophys. Acta 537,
D.L. Emerson (1975). Production and annealing 70-76.
studies of lead irradiated with protons or deuterons. 172. Symons, M.C.R. and R.L. Petersen (1978).
Radial Eff. 24, 89-94. Electron capture by oxyhaemoglobin: An e.s.r.
162. Khriachtchev, L., M. Pettersson, S. Jolkkonen, study. Proc. Roy Soc. B 201, 285-300.
S. Pehkonen, and M. Rasanen (2000). Photo- 173. Gasyna, Z. (1979). Intermediate spin-states in
chemistry of hydrogen peroxide in Kr and Xe one-electron reduction of oxygen-hemoprotein
matrixes. J. Chem. Phys., 112, 2187-2194. complexes at low temperature. FEBS Lett. 106,
163. Blyumenfeld, L.A., R.M. Davydov, S.P Kuprin, 213-218.
and S.V Stepanov (1977). Chemical characteristics 174. Davydov, R.M. (1980). Optical and ESR spectro-
of non-equilibrium conformational states of metal- scopic studies of electron adducts of oxymyoglobin
loproteins. Biofizika 22, 977-994. and oxyhemoglobin. Biofizika 25, 203-207.
164. Blumenfeld, L.A., R.M. Davydov, S.M. Magonov, 175. Davydov, R.M. and A.N. Ledenev (1981).
and R.O. Vilu (1974). Studies on the conforma- Activation mechanism of molecular oxygen by
tional changes of metalloproteins induced by cytochrome P-450. Biofizika 26 (1096).
electrons in water-ethylene glycol solutions 176. Davydov, R.M. and S.V Khangulov (1983). ESR
at low temperatures. Haemoglobin. FEES Lett. 49, spectroscopy of unstable intermediates of the
246-248. reduction of oxygen complexes of cytochrome
165. Davydov, R.M., S.N. Magonov, A.M. Arutyunyan, P450 and other hemoproteins. Stud. Biophys. 95,
and Y.A. Sharonov (1978). Absorption and 97-106.
magnetic circular dichroism spectra of heme- 177. Kappl, R., M. Hoehn-Berlage, J. Huettermann,
containing proteins in nonequilibrium states. IV. N. Bartlett, and M.C.R. Symons (1985). Electron
Cytochrome c and its derivatives. Molek. Biol. spin and electron nuclear double resonance of the
(Eng. Transl) 12, 1037-1042. [Fe02]~ [ferrite] center from irradiated oxyhemo-
166. Davydov, R.M., S. Greshner, S.N. Magonov, and oxymyoglobin. Biochim. Biophys. Acta 827,
K. Ruckpaul, and L.A. Blyumenfel'd (1978). 327-343.
Absorption spectra of cytochrome P-450 nonequi- 178. Leibl, W., W. Nitschke, and J. Huttermann (1986).
librium states formed during the low-temperature Spin-density distribution in the [Fe02]-complex.
reduction of protein. Dokl. Acad. Nauk SSSR 241, Electron spin resonance of myoglobin single
707-709. crystals. Biochim. Biophys. Acta 870, 20-30.
167. Magonov, S.N., R.M. Davydov, L.A. Blyumenfel'd, 179. Davydov, R., A. Ledbetter-Rogers, P. Martasek,
R. Vilu, A.M. Arutyunyan, and Y.A. Sharonov M. Larukhin, M. Sono, J.H. Dawson et al. (2002).
(1978). Absorption and magnetic circular dichroism EPR and ENDOR characterization of intermedi-
spectra of nonequilibrium states of heme-containing ates in the cryoreduced oxy-nitric oxide synthase
proteins. II. Myoglobin and its complexes. Molek. heme domain with bound L-arginine or N-7-
Biol. (Eng. Transl.) 12, 1182-1190. hydroxyarginine. Biochemistry 41, 10375-10381.
168. Magonov, S.N., R.M. Davydov, L.A. Blyumenfel'd, 180. Denisov, I.G., M. Ikeda-Saito, T. Yoshida, and
A.M. Arutyunyan, and Y.A. Sharonov (1978). S.G. Sligar (2002). Cryogenic absorption spectra
Absorption and magnetic circular dichroism spec- of hydroperoxo-ferric heme oxygenase, the active
tra of nonequilibrium states of heme-containing intermediate of enzymatic heme oxygenation.
proteins. III. Peroxidase complexes. Molek. Biol. FEBS Lett. 532,203-206.
(Eng Transl.) 12, 1191-1197. 181. Denisov, I.G., T.M. Makris, and S.G. Sligar (2002).
169. Greschner, S., R.M. Davydov, G.R. Jaenig, Formation and decay of hydroperoxo-ferric heme
K. Ruckpaul, and L.A. Blumenfel'd (1979). complex in horseradish peroxidase studied by
Spectral properties of nonequilibrium states in cryoradiolysis. J. Biol. Chem. 217, 42706^2710.
cytochrome P-450 formed by reduction at sub- 182. Davydov, R., S. Kuprin, A. Graeslund, and
zero temperatures. Acta Biol. Med. Germ. 38, A. Ehrenberg (1994). Electron paramagnetic reso-
443^48. nance study of the mixed-valent diiron center in
170. Symons, M.C.R. and R.L. Petersen (1978). Escherichia coli ribonucleotide reductase produced
Electron capture at the iron-oxygen centre in single by reduction of radical-free protein R2 at 77 K.
crystals of oxymyoglobin studied by electron J.Am. Chem. Soc. 116, 11120-11128.
178 Thomas M. Makris et al.

183. Davydov, R.M., A. Davydov, R. Ingemarson, (2002). Models of the low-spin iron(III) hydroper-
L. Thelander, A. Ehrenberg, and A. Graeslund oxide intermediate of heme oxygenase: Magnetic
(1997). EPR study of the mixed-valent diiron sites resonance evidence for thermodynamic stabiliza-
in mouse and herpes simplex virus ribonucleotide tion of the d(xy) electronic state at ambient tem-
reductases. Effect of the tyrosyl radical on struc- peratures. J. Am. Chem. Soc. 124, 6077-6089.
ture and reactivity of the diferric center. 194. Neese, F., J.M. Zaleski, K.L. Zaleski, and
Biochemistry 36, 9093-9100. E.I. Solomon (2000). Electronic structure of acti-
184. Dikanov, S.A., R.M. Davydov, L. Xun, and vated bleomycin: Oxygen intermediates in heme
M.K. Bowman (1996). CW and pulsed EPR char- versus non-heme iron. J. Am. Chem. Soc. 122,
acterization of the reduction of the Rieske-type 11703-11724.
iron-sulfur cluster in 2,4,5-trichlorophenoxy- 195. Neese, F. (2003). Quantum chemical calculations
acetate monooxygenase. J. Magn. Reson. B 112, of spectroscopic properties of metalloproteins and
289-294. model compounds: EPR and Mossbauer proper-
185. Lecloux, D.D., R. Davydov, and S.J. Lippard (1998). ties. Curr Opin. Chem. Biol. 7, 125-135.
Mixed-valence CuI-CuII and heterodimetallic Cul- 196. Kellner, D.G., S.-C. Hung, K.E. Weiss, and
MII bis(carboxylate-bridged) complexes: Structural, S.G. Sligar (2002). Kinetic characterization of
electrochemical, and spectroscopic investigations. compound I formation in the thermostable
Inorg. Chem. 37, 6814-6826. cytochrome P450 CYP119. J. Biol. Chem. 277,
186. Telser, J., R. Davydov, C.-H. Kim, M.W.W. Adams, 9641-9644.
and B.M. Hoffman (1999). Investigation of the 197. Egawa, T., H. Shimada, and Y. Ishimura (1994).
unusual electronic structure of Pyrococcus furiosus Evidence for compound I formation in the reaction
4Fe ferredoxin by EPR spectroscopy of protein of cytochrome-P45Ocam with m-chloroperbenzoic
reduced at ambient and cryogenic temperatures. acid. Biochem. Biophys. Res. Commun. 201,
Inorg Chem. 38, 3550-3553. 1464-1469.
187. Mizoguchi, T.J., R.M. Davydov, and S.J. Lippard 198. Egawa, T., DA. Proshlyakov, H. Miki, R. Makino,
(1999). Structural and spectroscopic compari- T. Ogura, T. Kitagawa et al. (2001). Effects of
sons between (JJL-OXO)- and (jx-hydroxo)bis a thiolate axial ligand on the TT -TT* electronic states
(ix-carboxylato)diiron(Ill) complexes that contain of oxoferryl porphyrins: A study of the optical
all-oxygen donor ligands. Inorg. Chem. 38, and resonance Raman spectra of compounds I and
4098^103. II of chloroperoxidase. J. Biol. Inorg. Chem. 6,
188. Krebs, C , R. Davydov, J. Baldwin, B.M. Hoffman, 46-54.
J.M. Bollinger, Jr., and B.H. Huynh (2000). 199. Groves, J.T. and G.A. McClusky (1976). Aliphatic
Moessbauer and EPR characterization of the hydroxylation via oxygen rebound. Oxygen
S — 9/2 mixed-valence Fe(II)Fe(III) cluster in the transfer catalyzed by iron. J. Am. Chem. Soc. 98,
cryoreduced R2 subunit of Escherichia coli 859-861.
ribonucleotide reductase. J. Am. Chem. Soc. 122, 200. Agmon, N. (1995). The Grotthuss mechanism.
5327-5336. Chem. Phys. Lett. 244, 456-462.
189. Sjogren, T. and J. Hajdu (2001). Structure of 201. Yoshikawa, S., K. Shinzawa-Itoh, R. Nakashima,
the bound dioxygen species in the cytochrome oxi- R. Yaono, E. Yamashita, N. Inoue et al. (1998).
dase reaction of cytochrome cdl nitrite reductase. Redox-coupled crystal structural changes in
J. Biol. Chem. 276, 13072-13076. bovine heart cytochrome c oxidase. Science 280,
190. Hajdu, J., R. Neutze, T. Sjogren, K. Edman, 1723-1729.
A. Szoke, and R.C. Wilmouth (2000). Analyzing 202. Perrin, C.L., T.J. Dwyer, J. Rebek, Jr., and R.J. Duff
protein functions in four dimensions. Nat. Struct. (1990). Exchange of amide protons. Effect of
Biol. 7, 1006-1012. intramolecular hydrogen bonding. J. Am. Chem.
191. Berglund, G.I., G.H. Carlsson, A.T. Smith, Soc. 112,3122-3125.
H. Szoke, A. Henriksen, and J. Hajdu (2002). The 203. Poulos, TL., B.C. Finzel, I.C. Gunsalus,
catalytic pathway of horseradish peroxidase at high G.C. Wagner, and J. Kraut (1985). The 2.6 A
resolution. Nature 417, 463^68. crystal structure of Pseudomonas putida cyto-
192. Maleknia, S.D., C.Y. Ralston, M.D. Brenowitz, chrome P450. J. Biol. Chem. 260, 6122-6130.
K.M. Downard, and M.R. Chance (2001). 204. Poulos, TL., B.C. Finzel, and A.J. Howard (1987).
Determination of macromolecular folding and High resolution crystal structure of cytochrome
structure by synchrotron X-ray radiolysis tech- P450cam. J. Mol. Biol. 195, 687-700.
niques. Anal. Biochem. 289, 103-115. 205. Tsai, R., C.A. Yu, I.G. Gunsalus, J. Peisach,
193. Rivera, M., G.A. Caignan, A.V Astashkin, W Blumberg, W.H. Orme-Johnson et al. (1970).
A.M. Raitsimring, T. Shokhireva, and F.A. Walker Spin state changes in cytochrome P450cam on
Activation of Molecular Oxygen by Cytochrome P450 179

binding of specific substrates. Proc. Natl. Acad. 217. Kimata, Y, H.Shimada, T.-A. Hirose, and
Sci. USA 66, 1157-1163. Y Ishimura (1998). Substitutions of artificial
206. Poulos, T.L., S.T. Freer, R.A. Alden, S.L. Edwards, amino acids O-methyl-Thr, 0-methyl-Asp, S-
U. Skogland, K. Takio et al. (1980). The crystal methyl-Cys, and 3-amino-Ala for Thr-252 of
structure of cytochrome c peroxidase. J. Biol. cytochrome P-450cam: Probing the importance of
Chem. 255, 575-580. the hydroxyl group of Thr-252 for oxygen activa-
207. Reid, T.J., III, M.R.N. Murthy, A. Sicignano, tion. Keio Univ. Symp. Life Sci. Med. 1, 161-165.
N. Tanaka, W.D.L. Musick, and M.G. Rossmann 218. Scheller, F, R. Renneberg, P Mohr, G.R. Janig, and
(1981). Structure and heme environment of beef K. Ruckpaul (1976). Peroxidatic activity of liver
liver catalase at 2.5 A resolution. Proc. Natl. Acad. microsomal cytochrome P-450. FEBS Lett. 71,
Sci. USA IH, 4167^771. 309-312.
208. Nebert, D.W., D.R. Nelson, M. Adesnik, 219. Nebert, D.W., J.R. Robinson, and H. Kon (1973).
M.J. Coon, R.W. Estabrook, EJ. Gonzalez et al. Genetically mediated differences in monoxygenase
(1989). The P450 superfamily—updated listing of activities and spin state of cytochrome P450 iron
all genes and recommended nomenclature for the from rabbit, rat, and mouse liver. J. Biol. Chem.
chromosomal loci. DNA J. Mol. Cell. Biol. 8, 1-13. 248, 7637-7647.
209. Nebert, D.W., D.R. Nelson, M.J. Coon, 220. Poulos, TL., B.C. Finzel, and A.J. Howard (1986).
R.W. Estabrook, R. Feyereisen, Y. Fujiikuriyama Crystal structure of substrate-free Pseudomonas
et al. (1991). The P450 superfamily—^update on putida cytochrome P-450. Biochemistry 25,
new sequences, gene-mapping, and recommended 5314-5322.
nomenclature. DNA Cell Biol. 10, 1-14. 221. Atkins, W.M., and S.G. Sligar (1988). Deuterium
210. Imai, M., H. Shimada, Y. Watanabe, isotope effects in norcamphor metabolism by
Y Matsushima-Hibiya, R. Makino, H. Koga et al. cytochrome P450cam—kinetic evidence for the
(1989). Uncoupling of the cytochrome P450cam 2-electron reduction of a high-valent iron oxo
monooxygenase reaction by a single mutation, intermediate. Biochemistry 27, 1610-1616.
threonine-252 to alanine or valine: Possible role of 222. Atkins, W.M. and S.G. Sligar (1988). The roles
the hydroxy amino acid in oxygen activation. Proc. of active site hydrogen bonding in cytochrome
Natl. Acad. Sci. USA 86, 7823-7827. P450cam as revealed by site-directed mutagenesis.
211. Martinis, S.A., W.M. Atkins, PS. Stayton, and J. Biol. Chem. 263, 18842-18849.
S.G. Sligar (1989). A conserved residue of 223. Sligar, S.G. and W. Atkins (1988). Metabolic
cytochrome P450 is involved in heme-oxygen switching and reduced oxygen intermediates dur-
stability and activation. J. Am. Chem. Soc. I l l , ing cytochrome P450cam catalysis. FASEB J. 2,
9252-9253. A1738-A1738.
212. Yeom, H., S.G. Sligar, H.Y Li, TL. Poulos, and 224. Atkins, W.M. and S.G. Sligar (1989). Molecular
A.J. Fulco (1995). The role of Thr268 in oxygen recognition in cytochrome P450—alteration of
activation of cytochrome P450(Bm-3). Biochemistry regioselective alkane hydroxylation via protein
34,14733-14740. engineering, c/^m. Chem. Soc. I l l , 2715-2717.
213. Imai, Y and M. Nakamura (1988). The importance 225. Paulsen, M.D. and R.L. Ornstein (1992). Pre-
of threonine-301 from cytochromes P-450 (laurate dicting the product specificity and coupling of
((o-l)-hydroxylase and testosterone 16a-hydroxy- cytochrome P450cam. J. Comput. Aided Mol. Des.
lase) in substrate binding as demonstrated by site- 6, 449-460.
directed mutagenesis. FEBSLett. 234, 313-315. 226. Loida, PJ. and S.G. Sligar (1993). Molecular
214. Ishimura, Y, H. Shimada, and M. Suematsu recognition in cytochrome P450—^mechanism for
(1998). Oxygen Homeostasis and its Dynamics. the control of uncoupling reactions. Biochemistry
International Symposium held at Minato-ku, 32, 11530-11538.
Tokyo, on December 8-13 1996 [In: Keio Univ. 227. Bell, S.G., X. Chen, R.J. Sowden, E Xu,
Symp. Life Sci. Med., 1998; 1]. 619 pp. J.N. Williams, L.-L. Wong et al (2003). Molecular
215. Cornish, VW, D. Mendel, and PG. Schultz (1995). recognition in (+)-a-pinene oxidation by cyto-
Probing protein structure and function with an chrome P450cam. ^ ^m. Chem. Soc. 125, 705-714.
expanded genetic code. Angew. Chem. Int. Ed. 34, 228. Harford-Cross, C.E, A.B. Carmichael, EK. Allan,
621-633. PA. England, D.A. Rouch, and L.-L. Wong (2000).
216. Kimata, Y, H. Shimada, T. Hirose, and Protein engineering of cytochrome P450cam
Y. Ishimura (1995). Role of Thr-252 in cytochrome (CYPlOl) for the oxidation of polycyclic aromatic
P450(Cam)—a study with unnatural amino-acid \vydioc?ivhons. Protein Eng. 13, 121-128.
mutagenesis. Biochem. Biophys. Res. Commun. 229. Jones, N.E., PA. England, DA. Rouch, and
208, 96-102. L.L. Wong (1996). Engineering the selectivity
180 Thomas M. Makris et al.

of aliphatic C-H bond oxidation catalysed water in the catalytic cycle of cytochrome P450
by cytochrome P450cam. Chem. Commun. camphor. FEBS Lett. 312, 252-254.
2413-2414. 242. Deprez, E., N.C. Gerber, C. DiPrimo, P Douzou,
230. Andersen, J.F. and C.R. Hutchinson (1992). S.G. Sligar, and G.H.B. Hoa (1994). Electrostatic
Characterization of Saccharopolyspora erythraea control of the substrate access channel in
cytochrome P-450 genes and enzymes, including cytochrome P-450(Cam). Biochemistry 33,
6-deoxyerythronolide B hydroxylase. J. Bacterial. 14464-14468.
174, 725-735. 243. DiPrimo, C , G. Hui Bon Hoa, E. Deprez,
231. Donadio, S. and C.R. Hutchinson (1991). Cloning R Douzou, and S.G. Sligar (1993). Conformational
and characterization of the Saccharopolyspora dynamics of cytochrome P-450cam as monitored
erythraea fdxA gene encoding ferredoxin. Gene by photoacoustic calorimetry. Biochemistry 32,
100,231-235. 3671-3676.
232. Cupp-Vickery, J.R., H. Li, and T.L. Poulos 244. DiPrimo, C , E. Deprez, S.G. Sligar, and
(1994). Preliminary crystallographic analysis of an G. Hui Bon Hoa (1997). Origin of the photo-
enzyme involved in erythromycin biosynthesis: acoustic signal in cytochrome P-450cam: Role of
Cytochrome P450eryF. Proteins 20, 197-201. the Argl86-Asp251-Lysl78 bifurcated salt bridge.
233. Cupp-Vickery, J.R. and T.L. Poulos (1995). Biochemistry 36, 112-118.
Structure of cytochrome P450eryF involved in 245. Deng, T.J., I.D.G. Macdonald, M.C. Simianu,
erythromycin biosynthesis. Nat. Struct. Biol. 2, M. Sykora, J.R. Kincaid, and S.G. Sligar (2001).
144-153. Hydrogen-bonding interactions in the active sites
234. Cupp-Vickery, J.R., O. Han, C.R. Hutchinson, of cytochrome P450cam and its site-directed
and T.L. Poulos (1996). Substrate-assisted catalysis mutants. J. Am. Chem. Soc. 123, 269-278.
in cytochrome P450eryF. Nat. Struct. Biol. 3, 246. Fedorov, R., D.K. Ghosh, and 1. Schlichting (2003).
632-637. Crystal structures of cyanide complexes of
235. Xiang, H., R.A. Tschirret-Guth, and PR. Ortiz P450cam and the oxygenase domain of inducible
De Montellano (2000). An A245T mutation nitric oxide synthase—structural models of the
conveys on cytochrome P450eryF the ability to short-lived oxygen complexes. Arch. Biochem.
oxidize alternative substrates. J. Biol. Chem. 275, Biophys. 409, 25-3\.
35999-36006. 247. Fulop, v., R.P Phizackerley, S.M. Soltis,
236. Ellis, S.W., G.P Hayhurst, G. Smith, T Lightfoot, I.J. Clifton, S. Wakatsuki, J. Erman et al. (1994).
M.M.S. Wong, A.P Simula et al. (1995). Evidence Laue diffraction study on the structure of cyto-
that aspartic acid 301 is a critical substrate-contact chrome c peroxidase compound I. Structure 2,
residue in the active site of cytochrome P450 2D6. 201-208.
J. Biol. Chem. 270, 29055-29058. 248. Taraphder, S. and G. Hummer (2003). Protein side-
237. Hiroya, K., M. Ishigooka, T Shimizu, and chain motion and hydration in proton-transfer
M. Hatano (1992). Role of Glu318 and Thr319 pathways. Results for cytochrome P450cam. J. Am.
in the catalytic function of cytochrome P450d Chem. Soc. 125,3931-3940.
(P4501A2): Effects of mutations on the methanol 249. Ozaki S., I. Hara, T Matsui, and Y. Watanabe
hydroxylation. FASEB J. 6, 749-751. (2001). Molecular engineering of myoglobin:
238. Zhou, D., K.R. Korzekwa, T.M. Poulos, and The improvement of oxidation activity by replac-
S. Chen (1992). A site-directed mutagenesis study ing Phe-43 with tryptophan. Biochemistry 40,
of human placental aromatase. J. Biol. Chem. 267, 1044-1052.
762-768. 250. Ozaki, S. and PR. Ortiz De Montellano (1995).
239. Lee, S.-K. and J.D. Lipscomb (1999). Oxygen Molecular engineering of horseradish peroxidase-
activation catalyzed by methane monooxygenase thioether sulfoxidation and styrene epoxidation by
hydroxylase component: Proton delivery during Phe-41 leucine and threonine mutants. J. Am.
the O-O bond cleavage steps. Biochemistry 38, Chem. Soc. 117,7056-7064.
4423-4432. 251. Yi, X., A. Conesa, PJ. Punt, and L.P Hager (2003).
240. Schwartz, B., J.E. Dove, and J.P Klinman Examining the role of glutamic acid 183 in
(2000). Kinetic analysis of oxygen utilization chloroperoxidase catalysis. J. Biol. Chem. 278,
during cofactor biogenesis in a copper-containing 13855-13859.
amine oxidase from yeast. Biochemistry 39, 252. Kato, S., H.J. Yang, T Ueno, S. Ozaki,
3699-3707. G.N. Phillips, Jr., S. Fukuzumi et al. (2002).
241. DiPrimo, C , S.G. Sligar, G.H.B. Hoa, and Asymmetric sulfoxidation and amine binding by
P. Douzou (1992). A critical role of protein-bound H64D/V68A and H64D/V68S Mb: Mechanistic
Activation of Molecular Oxygen by Cytochrome P450 181

insight into the chiral discrimination step. J. Am. characterization of n-butyl-isocyanide complexes of
Chem. Soc. 124, 8506-8507. cyto-chromes P450nor and P450cam. Biochemistry
253. Herold, S., T. Matsui, and Y. Watanabe (2001). 40,2669-2677.
Peroxynitrite isomerization catalyzed by His64 myo- 265. Okamoto, N., Y Imai, H. Shoun, and Y Shiro
globin mutants. J. Am. Chem. Soc. 123, 4085-4086. (1998). Site-directed mutagenesis of the conserved
254. Adachi, S., S. Nagano, Y. Watanabe, K. Ishimori, threonine (Thr243) of the distal helix of fungal
and I. Morishima (1991). Alteration of human cytochrome P450nor. Biochemistry 37, 8839-8847.
myoglobin proximal histidine to cysteine or tyro- 266. Obayashi, E., H. Shimizu, S.Y. Park, H. Shoun, and
sine by site-directed mutagenesis: Characterization Y Shiro (2000). Mutation effects of a conserved
and their catalytic activities. Biochem. Biophys. threonine (Thr243) of cytochrome P450nor on its
Res. Commun. 180, 138-144. structure and fiinction. J. Inorg. Biochem. 82,
255. Matsuo, T, T. Hayashi, and Y. Hisaeda (2002). 103-111.
Reductive activation of dioxygen by a myoglobin 267. Shimizu, H., E. Obayashi, Y Gomi, H. Arakawa,
reconstituted with a flavohemin. J.Am. Chem. Soc. S.-Y. Park, H. Nakamura et al. (2000). Proton
124, 11234-11235. delivery in NO reduction by fungal nitric-oxide
256. Harris, D.L. (2002). Cytochrome P450nor: A nitric reductase. J Biol. Chem. 275, 4816-4826.
oxide reductase-structure, spectra, and mechanism. 268. Matsunaga, I., E. Kusunose, I. Yano, and
Int. J. Quantum Chem. 88, 183-200. K. Ichihara (1994). Separation and partial charac-
257. Obayashi, E., S. Takahashi, and Y. Shiro (1998). terization of soluble fatty acid a-hydroxylase from
Electronic structure of reaction intermediate of Sphingomonas paucimobilis. Biochem. Biophys.
cytochrome P450nor in its nitric oxide reduction. Res. Commun. 201, 1554-1560.
J.Am. Chem. Soc. 120, 12964-12965. 269. Matsunaga, I., M. Yamada, E. Kusunose,
258. Park, S.-Y, H. Shimizu, S.-I. Adachi, Y Shiro, Y Nishiuchi, I. Yano, and K. Ichihara (1996).
T. lizuka, A. Nakagawa et al. (1997). Crystalli- Direct involvement of hydrogen peroxide in bacte-
zation, preliminary diffraction and electron para- rial a-hydroxylation of fatty acid. FEBS Lett. 386,
magnetic resonance studies of a single crystal of 252-254.
cytochrome P450nor. FEBS Lett. 412, 346-350. 270. Matsunaga, I., N. Yokotani, O. Gotoh, E. Kusunose,
259. Shimizu, H., S.Y Park, Y Shiro, and S. Adachi M. Yamada, and K. Ichihara (1997). Molecular
(2002). X-ray structure of nitric oxide reductase cloning and expression of fatty acid ot-hydroxylase
(cytochrome P450nor) at atomic resolution. Acta from Sphingomonas paucimobilis. J. Biol. Chem.
Crystallogr. D. Biol. Crystallogr. 58, 81-89. 272, 23592-23596.
260. Shimizu, H., S. Park, D. Lee, H. Shoun, and Y Shiro 271. Matsunaga, I., M. Yamada, E. Kusunose, T. Miki,
(2000). Crystal structures of cytochrome P450nor and K. Ichihara (1998). Further characteriza-
and its mutants (Ser286~>Val, Thr) in the ferric tion of hydrogen peroxide-dependent fatty acid
resting state at cryogenic temperature: A compara- a-hydroxylase from Sphingomonas paucimobilis.
tive analysis with monooxygenase cytochrome J Biochem. (Tokyo) 124, 105-110.
P450s. J Inorg. Biochem. 81, 191-205. 272. Matsunaga, I., A. Ueda, T. Sumimoto, K. Ichihara,
261. Imai, Y, T. Fukuda, M. Komori, and M. Nakamura M. Ayata, and H. Ogura (2001). Site-directed
(1994). Comparison of heme environment at the mutagenesis of the putative distal helix of peroxy-
putative distal region of P-450s utilizing their genase cytochrome P450. Arch. Biochem. Biophys.
external and internal nitrogenous ligand bound 394, 45-53.
forms. Biochim. Biophys. Acta 1207, 49-57. 273. Matsunaga, I., T. Sumimoto, M. Ayata, and
262. Imai, Y and M. Nakamura (1991). Nitrogenous H. Ogura (2002). Functional modulation of a per-
ligation at the sixth coordination position of the oxygenase cytochrome P450: Novel insight into
Thr-301 to Lys-mutated P450IIC2 heme iron. the mechanisms of peroxygenase and peroxidase
J Biochem. (Tokyo) 110, 884-888. enzymes. FEBS Lett. 528, 90-94.
263. Obayashi, E., K. Tsukamoto, S. Adachi, 274. Imai, Y, I. Matsunaga, E. Kusunose, and K. Ichihara
S. Takahashi, M. Nomura, T. lizuka et al. (1997). (2000). Unique heme environment at the puta-
Unique binding of nitric oxide to ferric nitric oxide tive distal region of hydrogen peroxide-dependent
reductase from Fusarium oxysporum elucidated fatty acid a-hydroxylase from Sphingomonas
with infrared, resonance Raman, and X-ray paucimobilis (peroxygenase P450(SPa). J. Biochem.
absorption spectroscopies. J. Am. Chem. Soc. 119, (Tokyo) 128, 189-194.
7807-7816. 275. Lee, D.S., A. Yamada, H. Sugimoto, I. Matsunaga,
264. Lee, D.-S., S.-Y. Park, K. Yamane, E. Obayashi, H. Ogura, K. Ichihara et al. (2003). Substrate
H. Hori, and Y Shiro (2001). Structural recognition and molecular mechanism of fatty
182 Thomas M. Makris et al.

acid hydroxylation by cytochrome P450 from 0 - 0 stretching modes. Eur J. Inorg. Chem.
Bacillus subtilis: Crystallographic, spectroscopic 1627-1633.
and mutational studies. J. Biol. Chem. 278, 282. Nguyen, C , R.J. Guajardo, and PK. Mascharak
9761-9767. (1996). [FeIII(PMA)]2+: A mononuclear non-
276. Sundaramoorthy, M., J. Terner, and T.L. Poulos heme-iron complex that catalyzes alkane oxidation.
(1995). The crystal structure of chloroperoxidase: Inorg Chem. 35, 6273-6281.
A heme peroxidase-cytochrome P450 functional 283. Ozawa, S., Y. Watanabe, and I. Morishima (1994).
hybrid. Structure 3, 1367-1377. Spectroscopic characterization of peroxo-iron(III)
277. Horner, O., C. Jeandey, J.-L. Oddou, P. Bonville, chlorin complexes. The first model for a reaction
C.J. Mckenzie, and J.-M. Latour (2002). Hydrogen- intermediate of cytochrome d. Inorg. Chem. 33,
peroxo-[(bztpen)Fe(OOH)]2+ and its deprotona- 306-313.
tion product peroxo-[(bztpen)Fe(02)] + , studied by 284. Sauer-Masarwa, A., N. Herron, CM. Fendrick, and
EPR and Mossbauer spectroscopy—implications D.H. Busch (1993). Kinetics and intermediates in
for the electronic structures of peroxo model the autoxidation of synthetic, non-porphyrin
complexes. Eur. J. Inorg. Chem. 3278-3283. iron(II) dioxygen carriers. Inorg. Chem. 32,
278. Shearer, J., R.C. Scarrow, and J.A. Kovacs (2002). 1086-1094.
Synthetic models for the cysteinate-ligated non- 285. Tajima, K., K. Mikami, K. Tada, S. Oka, K. Ishizu,
heme iron enzyme superoxide reductase: Obser- and H. Ohya-Nishiguchi (1992). Autoreduction of
vation and structural characterization by XAS of an a six-coordinate iron(III) tetraphenylporphyrin-
Fe(III)-OOH intermediate. J. Am. Chem. Soc. 117, peroxide complex by heterolytic iron-oxygen bond
11709-11717. cleavage. Inorg. Chim. Acta 194, 57-65.
279. Hazell, A., C.J. Mckenzie, L.P Nielsen, 286. Fujii, S., H. Ohya-Nishiguchi, and N. Hirota
S. Schindler, and M. Weitzer (2002). Mononuclear (1990). EPR evidence of intermediate peroxo com-
non-heme iron(III) peroxide complexes: Syntheses, plexes formed in a superoxide dismutase (SOD)
characterisation, mass spectrometric and kinetic model system. Inorg. Chim. Acta 175, 27-30.
studies. J Chem. Soc, Dalton Trans. 310-317. 287. Burstyn, J.N., J.A. Roe, A.R. Miksztal,
280. Simaan, A.J., K Banse, J.-J. Girerd, K. Wieghardt, B.A. Shaevitz, G. Lang, and J.S. Valentine
and E. Bill (2001). The electronic structure of non- (1988). Magnetic and spectroscopic characteriza-
heme iron(lII)-hydroperoxo and iron(IIl)-peroxo tion of an iron porphyrin peroxide complex.
model complexes studied by Moessbauer and elec- Peroxoferrioctaethylporphyrin(l-). J. Am. Chem.
tron paramagnetic resonance spectroscopies. Inorg. Soc. 110, 1382-1388.
Chem. 40, 6538-6540. 288. Clay, M., D., FE. Jenney, Jr., PL. Hagedoom,
281. Simaan, A.J., S. Dopner, F. Banse, S. Bourcier, G.N. George, M.W.W. Adams, and M.K, Johnson
G. Bouchoux, A. Boussac et al. (2000). (2002). Spectroscopic studies of Pyrococcus
Felll-hydroperoxo and peroxo complexes with furiosus superoxide reductase: Implications for
aminopyridyl ligands and the resonance Raman active-site structures and the catalytic mechanism.
spectroscopic identification of the Fe-0 and J. Am. Chem. Soc. 124, 788-805.
6
Substrate Oxidation by Cytochrome
P450 Enzymes
Paul R. Ortiz de Montellano and James J. De Voss

1. Introduction P450^^^ (CYPlOl)io, P450B^.3 (CYP102)11' ' \


P450^^^p (CYP108)i3, P450^^yp (CYP107Al)l^
The cytochromes P450 are catalytic hemo- P450^^^ (CYP55Al)i^ Sulfolobus solfataricus
proteins in which the heme iron atom is coordi- CYPl 19^^ Streptomyces coelicolor CYP154Cli\
nated to a proximal cysteine thiolate. This thiolate Mycobacterium tuberculosis CYP52^^, Sorangium
ligand is responsible for the characteristic Soret cellulosum P450epoK^^, and the mammalian
absorption maximum of the Fe"-CO complex at CYP2C5 (see Chapter 3>f^. Although the thiolate
—450 nm and is critical for P450 catalysis^. Early ligand is always present, the distal water ligand
site-specific mutagenesis studies with CYP1A2 appears to be absent in some mammalian
and P450^^j^ suggested that replacement of the enzymes, either because the water does not bind in
cysteine thiolate by a histidine ligand gave inac- those structures or because it is displaced by an
tive protein^' ^. Detailed studies of the P450^^^ endogenous ligand^ ^
Cys357His mutant have recently confirmed that The c)^ochrome P450 catalytic cycle is initiated
this mutant enzyme has an almost undetectable by the binding of a substrate, usually with con-
catalytic activity^' ^. The low camphor-oxidizing comitant displacement of the distal water ligand.
activity is paralleled by a low rate of reduction of The ferric heme is then reduced to the ferrous state
the iron, an elevated autooxidation rate, and an using electrons provided by suitable electron donor
observable peroxidase activity"*. The thiolate lig- proteins (see Chapter 4). In cytochrome P450^^
and is thus clearly critical for P450^^j^ function, and many other P450 enzymes, substrate binding
although the relative contributions of the elec- is widely believed to be a prerequisite for the trans-
tronic vs structural perturbations of the mutation fer of the first electron to the iron, but in some
to the low catal)^ic activity remain unclear. These enzymes electron transfer can occur without the
results agree with the results of experiments with prior binding of a substrate^ ^. Reduction of the iron
thiolate ligated metalloporphyrin model systems^' ^ is followed by binding of oxygen to give the fer-
and of computational analyses of the role of the rous dioxy complex. Transfer of a second electron
thiolate (see Chapter 2)^' ^. to this complex produces the ferric peroxy anion
The heme iron ligand on the distal side is a (PorFe"^-00~, where Por = porphyrin) or, after
water molecule in all the available crystal struc- protonation, the ferric hydroperoxo complex
tures of substrate-free P450 enzymes, including (Por"^-OOH) (Figure 6.1). Heterolytic cleavage of

Paul R. Ortiz d e Montellano • Department of Pharmaceutical Chemistry, University of California, San


Francisco, CA. J a m e s J. De Voss • Department of Chemistry, University of Queensland, Brisbane, QLD
Australia.

Cytochrome P450: Structure, Mechanism, and Biochemistry, 3e, edited by Paul R. Ortiz de Montellano
Kluwer Academic / Plenum Publishers, New York, 2005.

183
184 Paul R. Ortiz de Montellano and James J. De Voss

|Fe"-02|[sH]--^02

yj+ f 1 H '
|Fe=o]|sH]-^--— |Fe"i-02H||sH]-*— [Fe"'-02-l[sH]
—rl20
• 2e-
t t
H2O H2O2

Figure 6.1. The general catalytic cycle of cytochrome P450 enzymes. The [Fe"^] stands for the resting ferric state
of P450, and SH for a substrate molecule. The shunt pathway utilizing H2O2 is shown as are three sites for the
uncoupling of the enzyme to give, respectively, O^, H2O2, or H2O.

the dioxygen bond in this peroxo intermediate (b) a molecule of H2O2 dissociates from the ferric
extrudes a molecule of water and forms the hydroperoxide complex, or (c) two electrons are
putative ferryl oxidizing species (Figure 6.1). used to reduce the ferryl species to a molecule of
Hydrogen bonding of the distal ferric hydroperoxo water before it can be used in a reaction with the
oxygen, directly or via a water molecule, to a substrate (Figure 6.1). The parameters that govern
highly conserved threonine facilitates this het- uncoupling at each of the three stages are unclear,
erolytic cleavage (see Chapter 5)^^~^^. The ferryl but factors that contribute to uncoupling appear to
species is thought to be responsible for most P450- be the degree of uncontrolled water access to the
catalyzed oxidations, although the ferric peroxo active site, the extent to which the substrate can
anion and the ferric hydroperoxo complex have reside at unproductive distances from the ferryl
been invoked as oxidizing species (see below). It is species, and the presence or absence of sufficiently
usually, but not always, possible to circumvent the reactive sites on the substrate molecule^^' ^'^-'^^, The
requirement for activation of molecular oxygen in catalytic efficiency of a P450 enzyme can be seri-
a so-called "shunt" pathway by employing H2O2 or ously impaired by uncoupling, as evidenced by the
some other peroxide as a co-substrate (Figure 6.1). contrast between nearly quantitative coupling in the
However, the oxidizing species thus obtained is oxidation of camphor by P450^^j^ and a process that
apparently not identical to that obtained by normal is more than 95% uncoupled when the same
oxygen activation. Thus, peroxides cannot replace enzyme oxidizes styrene^^. A higher degree of
molecular oxygen activation in some reactions, intrinsic uncoupling is often observed in mam-
they often give product distributions that differ sig- malian P450 enzymes, some of which can be sup-
nificantly from those obtained by molecular oxy- pressed by interaction of the P450 enzyme with
gen activation^^"^^, and they cause a more rapid reduced cytochrome b^^^' ^^.
degradation of the prosthetic heme group^'.
The P450 oxidation stoichiometry requires one
molecule of oxygen and two electrons from
NAD(P)H to add one oxygen atom to a substrate. If 2. Activation of
the ratio of reduced pyridine nucleotide (or oxygen) iVIolecufar Oxygen
consumed to product formed is greater than one,
the enzyme is said to be uncoupled. Uncoupling Cryogenic X-ray crystallographic, EPR,
occurs when (a) the ferrous dioxy complex reverts ENDOR, and spectroscopic studies have convinc-
to the ferric state by dissociation of superoxide. ingly identified several intermediates in the P450
Substrate Oxidation by Cytochrome P450 Enzymes 185

catalytic cycle (see Chapter S)^^"^^. These include three substrates plus styrene"*^. To rationalize this
the ferric, ferrous, ferrous dioxo, and ferric observation, the authors argued that hydroxylation is
hydroperoxo complexes of P450^^j^. Crystallo- mediated exclusively by the ferryl whereas epoxida-
graphic evidence has also been reported for the tion can be mediated by both the ferryl and ferric
ferry 1 species^^, but this intermediate has not been hydroperoxide intermediates. Thus, impairing for-
detected by other sensitive cryogenic approaches mation of the ferryl species by removing the cat-
and its attribution to the ferryl species remains alytic threonine would decrease hydroxylation but
open to question. In low-temperature EPR, have little effect upon epoxidation. However, in con-
ENDOR, and spectroscopic studies, the ferric trast to the results with the CYP2E1 T303A mutant,
hydroperoxide intermediate disappears as the the corresponding T302A mutant of CYP2B4
hydroxylated camphor product appears without exhibited both decreased hydroxylation and epoxi-
the observation of any intermediate species^^' ^^. dation rates. This discrepancy does not necessarily
All the intermediates in oxygen activation by contradict the hypothesis, as it could reflect differ-
P450 have thus been observed except for the crit- ential changes in the active sites of the two proteins
ical ferryl species, which remains elusive and in addition to elimination of the hydrogen bond that
undefined. facilitates ferryl formation, hi a more recent study in
As already mentioned, the activation of molec- which Thr252, the catalytic threonine of P450^^^,
ular oxygen can often be circumvented if perox- was mutated to an alanine, it was found that cam-
ides are used as activated oxygen donors. Efforts phor hydroxylation was suppressed, but the epoxi-
to identify the reactive oxygen species in these dation of an olefinic camphor analogue could still
peroxide-supported reactions have been pursued be observed'*^. However, the epoxidation reaction
for many years'^ ^^^. The species that has been spec- occurred at a much slower rate (<20%) despite the
troscopically detected in these reactions has the expectation that the steady-state level of the ferric
spectroscopic signature of a ferryl intermediate^^, hydroperoxide should be elevated. This finding is
but evidence is lacking that this intermediate is the consistent with the prediction by computational
same as that produced by the activation of molecu- studies that the ferric hydroperoxo complex should
lar oxygen. To the contrary, the reactions with per- be a very poor olefin-oxidizing agent^^. These
oxides have been shown to produce EPR signals results argue that in the wild-type proteins, the fer-
tentatively attributed to tyrosine radicals^ ^' '^^' ^^, but ric hydroperoxide makes no more than a small con-
no such radicals have been observed under normal tribution to epoxidation, and none to hydroxylation.
turnover conditions. Furthermore, as noted earlier, In a second study, the iV-oxidation of amines by
the peroxide-mediated reactions do not always CYP2B4 and its T302A mutant supported by either
faithfully reproduce the normal reactions. NADPH-cytochrome P450 reductase or H2O2 was
Two additional intermediates, the ferric peroxy investigated^ ^ In contrast to what would be
anion and ferric hydroperoxo complex, have been expected if the ferric hydroperoxide were a primary
proposed to substitute for the ferryl as the actual catalytic species, the rates of iV-demethylation and
oxidizing species in at least some P450 reactions. A/-oxidation of 7V,N-dimethylaniline were both
The role of the ferric peroxy anion in some reac- decreased in the mutant. However, as these activities
tions is supported by good evidence and is dis- were also decreased when H2O2 or phenyliodoso-
cussed in the section on carbon-carbon bond benzene was used in a shunt reaction, little can be
cleavage reactions (see Section 8), but the pro- said from these results relative to the role of the fer-
posed role of the ferric hydroperoxide in elec- ryl vs ferric hydroperoxide species in these reac-
trophilic double bond and heteroatom oxidations tions. The oxidation of/7ara-substituted phenols via
is discussed here. an /p5o-substitution mechanism using the CYP2E1
The current interest in the ferric hydroperoxo T303A and CYP2B4 T302A mutants has also given
complex as a P450-oxidizing species derives largely contradictory results (Figure 62f^. The T303A
fi*om the work by Vaz et aL, who observed that CYP2E1 mutation increased the rates of ipso-
mutation of the conserved threonine (Thr303) in substitution with \Qpara substituents ranging fi-om
CYP2E1 to an alanine decreased the allylic hydrox- a chloride to a ^^r^butyl group but did not increase
ylation of cyclohexene, cis-2-butQnQ, and trans-2- or decrease the rate of reaction of/7(3!ra-fluorophenol,
butene, but increased the epoxidation of the same by far the most active of the investigated substrates
186 Paul R. Ortiz de Montellano and James J. De Voss

techniques, and two because the ferryl produced


with peroxides may not be identical to the reactive
species formed by oxygen activation. Although it
has not been reliably detected as a normal interme-
diate, the ferryl species is nevertheless almost cer-
tainly responsible for the majority of the chemistry
X^ OH
supported by P450 enzymes. The circumstantial
and contradictory evidence so far available does
not provide strong support for significant involve-
ment of the ferric hydroperoxide species in normal
P450-catalyzed reactions. Although mutation of
the conserved threonine appears in some instances
to cause the apparent intervention of a second dif-
^OH ferentiable oxidizing species, the evidence does
not actually indicate the nature of this second
Figure 6.2. Hypothetical i/75o-substitution mechanism species. Computational comparison of the ferric
involving the ferric hydroperoxo complex and ferryl hydroperoxo and ferryl reactivities suggests that
species as the potential oxidizing species^^.
the ferric hydroperoxo complex is a poor oxidizing
agent unlikely to contribute significantly to P450
for this reaction. Furthermore, although the increase catalysis (see Chapter 2)^^' ^^.
in the reaction rate correlated well with the sub-
stituent electronegativity for wild-type CYP2E1, the
reaction with the mutant gave a biphasic correlation 3. Hydrocarbon Hydroxylation
that included a region in which the reactivity was
shown to decrease with increasing electron with- Proposals on the mechanism of hydrocarbon
drawal^^. CYP2B4 exhibited only a low activity in hydroxylation have become increasingly complex
this reaction and this activity was not greatly and sophisticated over the past decade. The most
changed when the conserved threonine was mutated widely accepted mechanism involving hydrogen
to an alanine. It has finally also been proposed that atom abstraction by the ferryl oxygen followed by
the ferric hydroperoxo complex may play a role in rebound recombination of the resulting carbon rad-
the hydroxylation of saturated hydrocarbons^^"^^. ical with the iron-bound oxygen, first clearly stated
As discussed in Section 3, these studies indicate that in 1978^^, has more recently been challenged, pri-
a second (or altered) oxidant contributes to the oxi- marily on the basis of work with radical clock
dation in threonine mutant enzymes, and suggest probes. As already discussed, the high-valent oxi-
that a minor fraction of the reaction products are dizing species responsible for most, if not all,
formed via nonobligate cationic intermediates, but cytochrome P450 substrate oxidations is likely to
do not specifically implicate the ferric hydroperoxo be the iron(IV)oxo porphyrin radical cation
species in hydrocarbon hydroxylation reactions. (Por^Te'^=0). Recent calculations support this
It may be relevant to the observation of two oxidiz- formulation^^' ^\ although they suggest that the
ing species that hydrogen bonding to the thiolate radical density may reside to a greater or lesser
ligand, which is very sensitive to structure, has been extent on the thiolate iron ligand or other protein
found in calculations to govern the distribution of residues (see Chapter 2). In the conventional oxygen
unpaired electron density between the porphyrin rebound mechanism, the Por^Te^^^O species
and protein^^. abstracts a hydrogen atom from a carbon of the
In sum, all the reaction intermediates with the substrate, producing a PorFe^^-OH species and a
exception of the ferryl species have been clearly carbon radical. The Fe^^-OH species, which can
detected and identified in the catalytic cycle of at also be viewed as a complex of Fe^^^ with a
least one P450 enzyme. The two instances in which hydroxyl radical, then undergoes a recombination
it has been proposed that the ferryl species step in which the hydroxyl radical equivalent and
was detected have shortcomings, one because the carbon radical combine to produce the hydroxy-
finding is not reproduced with other detection lated product. The discrete radical intermediate
Substrate Oxidation by Cytochrome P450 Enzymes 187

proposed in this mechanism readily explains the site of the radical intermediate. The ring strain
repeated experimental observation of high intrinsic inherent in a cyclopropyl carbinyl (or related)
isotope effects (often >10)^^' ^^' ^^, partial scram- radical leads to a rapid and essentially irreversible
bling of substrate stereochemistry^^' ^^^ ^^, and inci- rearrangement to the corresponding homoallylic
dence of ally lie rearrangements in P450-catalyzed radical. In the case of P450 hydroxylation, the
hydroxylations^"^' ^^. Scrambling of stereochem- hydroxyl group can be delivered either to the cyclo-
istry has been seen in many situations and includes propyl carbinyl or rearranged homoallylic radical,
the early observation that P450^^j^, removes either and the ratio of the two resulting products is deter-
a S-exo or 5-endo hydrogen fi*om camphor but mined by the relative magnitudes of the rate con-
transfers the oxygen exclusively to the 5-exo posi- stants for radical quenching (k^. Figure 6.5) and
tion to yield the 5-^x<9-hydroxy product^^. Allylie rearrangement (A^., Figure 6.5). As the intrinsic
rearrangements, indicative of a delocalized inter- rearrangement rate can be independently measured
mediate, have been observed with 3,4,5,6-tetra- in nonbiological experiments, one can calculate
chlorocyclohexene and other cyclohexenes^^' ^^, both the lifetime of the radical and the rate of radi-
linoleic acid^^, and a variety of other compounds cal recombination from the ratio of unrearranged to
(Figure 6.3). In the same vein, the recently rearranged products. The first experiments with
observed cleavage of a carbon-carbon bond in the substrates containing simple, unsubstituted cyclo-
P450-catalyzed oxidation of marmesin is most propyl rings {k^= 1.3 X 10^ s~^) only gave unre-
plausibly rationalized by a mechanism involving a arranged products^^'^^. However, bicyclo[2.1.0]
carbon radical intermediate (Figure 6.4)^^. pentane, which gives a radical that rearranges much
Since 1987^^, major efforts have been made to faster (k^ = 2.4 X 10^ s"^) due to the additional
use radical clocks to estimate the rate of the oxygen strain in the system'''^, was converted by
rebound step and the lifetime of the radical inter- cytochrome P450 into a mixture of rearranged and
mediate in the hydroxylation reaction. In radical unrearranged products from which a rebound rate
clocks, a strained—^usually cyclopropyl—^ring is of 1.4 X IQio s-i could be calculated^^, 74
directly bonded to the carbon that is the proposed Radical clocks of increasing sophistication were
subsequently employed to define better the radical
intermediate in hydrocarbon hydroxylation^"^^^.
Addition of substituents to the cyclopropyl ring
increases the rate of the ring-opening reaction. For
example, the rearrangement rate for a 2-aryl substi-
tuted cyclopropyl carbinyl radical in solution is
OH approximately 1,000-times faster than that of the
parent compound without the 2-aryl group^^.
D D Experiments with these faster radical clocks should
have increased the proportion of rearranged prod-
ucts and thus led to a more accurate value for the
lifetime of the radical. Contrary to expectation, the
D D D D
measured rate of the radical recombination step
Figure 6.3. Allylic rearrangements observed in the appeared to increase in parallel with the rate of the
hydroxylation by cytochrome P450 of two substituted probe rearrangement, resulting in a lower rather
cyclohexenes. than higher proportion of the rearranged

[Fe=0]- [Fe-OH]

OH O o"^o OH O O^^O
Me2CO

Figure 6.4. Mechanism proposed for the unusual P450-catalyzed carbon-carbon bond fragmentation observed
during the biosynthesis of psoralen^^.
188 Paul R. Ortiz de Montellano and James J. De Voss

[Fe=0]^ ^CH3 CH.


CH3
Cgfis C6H5C6H5
[Fe—OH]^ kt [Fe—OH]' 1 2

OH

Figure 6.5. The principle of radical clock probes of


the cytochrome P450 mechanism based on the methyl-
cyclopropyl radical rearrangement.

product^^' ^^. When the very small amount of Figure 6.6. Examples of radical clock probes of the
cytochrome P450 mechanism, including the radical
rearranged product was used to calculate the radi-
rearrangements typical of norcarane and bicyclo[2.1.0]
cal rebound rate and radical lifetime, values were pentane.
obtained that challenged the existence of a discrete
radical intermediate. Thus, the 2-phenyl and 2,2-
diphenyl substituted probes 1 and 2 (Figure 6.6) the hydroxylation reaction may be more complex
used by Atkinson and Ingold yielded a radical than predicted by a simple radical rebound mecha-
rebound rate of 2-7 X lO^^ s" values that nism. Norcarane and bicyclo[2.1.0]pentane differ
approach the limiting rate constant of approxi- from most of the radical clocks examined to date in
mately 6 X lO'^s"^ at 37°C imposed by transition that the radical is located on a secondary rather than
state theory. At these rates, the existence of a dis- primary carbon. This led to the proposal that the
crete radical intermediate is reduced to a question extent of rearrangement might depend on both
of semantics. The observation of similar high rates the initial hydrogen abstraction transition state and
with other radical clocks, the demonstration that the subsequent radical recombination transition
even large substrates undergo significant motion state, and that the shift from one to the other might
within P450 active sites, and the observation of be easier for methyl probes because of the tighter
intramolecular isotope effects with some of the transition state due to the higher C-H bond strength
probes, suggests that the rearrangement is not being and smaller size of the methyl group^^. However,
suppressed by interaction of the probe with the pro- the finding that four (2-phenyl)cyclopropylalkyl
tein structure^^. Indeed, analysis of the products probes, in which the radical also resides on a sec-
formed from the structurally rigid probe 3 gave the ondary (or tertiary) carbon, give very high recom-
unbelievably high rebound rate of 1.4 X 10' ^ s ~' ^^. bination rates is at odds with this explanation^^. An
These results argued that a radical intermediate was attractive solution for this dilemma is provided by
not mandatory in hydrocarbon hydroxylation. the two-state reactivity theory, which postulates a
However, recent experimental work has again com- competition between two parallel reaction path-
plicated the radical clockresults^^' ^^. The oxidation ways (see Chapter 2). One of these pathways is
of norcarane (4) by four P450 enzymes gave a rad- equivalent to a concerted oxygen insertion and the
ical rebound rate o f ~ 1 0 ^ ^ s ^ a rate very close to other akin to a conventional radical recombination
that of 1.4 X 10'^ s~^ from the original experi- mechanism, and the dominant pathway in any
ments with bicyclo[2.1.0] pentane (Figure 6.6)^^' ^'^. given situation is substrate and environment
Similar experiments with spiro[2,4]octane, a dependent. A substrate-dependent reaction mecha-
related structure that rearranges much more slowly nism readily rationalizes the differences in the
than norcarane, did not, as expected, detectably results obtained with the different radical clock
yield rearranged products^^. probes. Reevaluation of the radical clock results
The discrepancies among the radical clocks, and additional experimental and theoretical work
which give credible radical lifetimes in the case are required to satisfactorily reconcile the contra-
of simple cyclopropylmethyl and bicyclic dictory results provided by the probes.
probes, but impossibly short lifetimes with the The conclusion from some of the radical clock
phenylcyclopropylcarbinyl probes, suggests that data that P450 hydroxylation might not proceed
Substrate Oxidation by Cytochrome P450 Enzymes 189

via a radical intermediate led Newcomb to A mechanism that reconciles the differences in
propose that oxygen might be inserted into the the radical clock results has been postulated by
C-H via a concerted, nonradical mechanism^^' ^^. Shaik on the basis of theoretical calculations. This
According to this proposal, the hydroxylation mechanistic hypothesis, which elegantly combines
traverses a bifurcated transition state that allows an essentially concerted insertion and a noncon-
some of the probes to leak into a radical certed radical reaction in a single pathway^*' ^^~^^,
rearrangement manifold, thus explaining the is at once complex, intriguing, and satisfying. As
observation of rearranged products. A shortcom- discussed in detail in Chapter 2, the oxidizing
ing of this rationale is that it must explain a large species is an F e ^ ^ = 0 porphyrin radical cation
diversity of reaction outcomes, including radical that is present as two approximately equienergetic
clock rearrangements, allylic transpositions, and electromers, one in a doublet spin state and the
stereochemical scrambling, by postulating a range other in a quartet spin state^^' ^^. Intuitively, this
of electronically and structurally different bifur- small difference can be viewed as arising from the
cated transition states. Furthermore, even more combination of two electrons with unpaired spins in
complex structural rearrangements have been the d orbitals of the iron and a third unpaired elec-
reported that are most consistent with the inter- tron in the a2y orbital of the porphyrin (Figure 6.8).
vention of a radical intermediate. For example, The quartet and singlet species abstract a hydrogen
dieldrin 5 is converted into the intramolecularly atom from the hydrocarbon through nearly identi-
bridged ketone 6 and Dolphin has demonstrated cal transition states, which readily explains the
that this product is directly produced when 5 is measured isotope effects and the similarities in the
oxidized by a highly halogenated iron porphyrin reactivities of the P450 enzymes and the ^BuO•
(Figure 6.7)^^. The alcohol 7 is also produced radical^^' ^^. The transition states lead to complexes
metabolically in vivo and in vitro, but no chemical in which the alkyl radical is weakly coordinated to
or biochemical conditions have yet been found the iron-bound hydroxyl. These complexes, which
that will convert it to the ketone. The most rea- can be in a doublet or quartet spin state and are
sonable explanation for the formation of the again of nearly the same energy, derive from
ketone product involves hydrogen abstraction to the corresponding doublet and quartet states of the
give a carbon radical, cyclization of this radical ferryl species. The species in the doublet spin
driven by the formation of a chlorine stabilized state can collapse to the hydroxylated product in
radical, recombination with the iron-bound a virtually barrierless (i.e., essentially concerted),
hydroxy radical to give the alcohol, and elimina- nonsynchronous reaction with no true intermediate.
tion of HCl to give the ketone (Figure 6.7). The porphyrin cation acts to accept an electron and

H H CI H
[Fe=0]^

[Fe-OH]^+

Cl.^\ H^OH

-HC^ HO W l

Figure 6.7. Unusual reactions observed in the oxidation of dieldrin.


190 Paul R. Ortiz de Montellano and James J. De Voss

TSReb_,

^Agu + RH
% u + RH

R-H R*
O

L
•4"'- L
L
III
II
dz^(a*) d/ia*) d2'^(CT*)
4 dz2(a'

V .H
djt (7t*FeO)

1 T a2u
'A2U(2A2U) '^Por+*FeOH(^Por+*FeOH) R*

Figure 6.8. The ferryl radical cation-substrate complex (I) has two unpaired electrons in the iron d^ orbitals and
one in the 3.2^ porphyrin orbital. This may give rise to either a quartet C^A2u) state if all spins are unpaired or, if the
spin of the electron in the ^2^^ orbital is inverted, the complex is in a doublet (^A2y) state. (Another possible pair of
doublet/quartet configurations has a filled 32^ orbital and a single electron in p'n-[S] orbital.) In the first step, a
hydrogen atom is abstracted from the substrate and an electron is transferred to either the iron, reducing it to the Fe™
state (II), or to the porphyrin, quenching the radical cation, the former being suggested by calculations. Each of these
"intermediates" (II) can be in a quartet or doublet state, depending on the electron pairing between the carbon radical
R" and the electrons in the iron porphyrin system. Low-spin II collapses to a low-spin product (III) in a rebound
reaction that is virtually barrierless while high-spin II traverses a significant barrier to arrive at high-spin III. Thus,
only high-spin II is believed to behave as a true intermediate (radical). The energy diagram corresponding to the
transformation is shown above. This scheme is based on the work of Shaik and coworkers.

the thiolate ligand increases its interaction with the Our discussion of the P450 mechanism to this
iron atom and both of these processes facilitate the point has been based on the assumption that the
effectively concerted nature of the transformation. reactive intermediate is a ferryl species. However,
In contrast, the quartet species must traverse a sig- the possibility has been raised that the ferric
nificant energy barrier to form the product, thus hydroperoxide rather than the ferryl species might
allowing the formation of true intermediate radicals be the active agent in carbon hydroxylation^^' ^^' ^^.
and the consequent reactions (rearrangements) that A direct role for the Fe^"-OOH intermediate in
this implies. The calculated barrier to product for- substrate oxidation was postulated to explain the
mation in this pathway appears to arise from the fact formation of ring opened products in the oxidation
that in the recombination reaction, to maintain the of trans-1 -methyl-2-(4-trifluoromethyl)phenylcy-
quartet spin state, one electron must reside in a rel- clopropane (8) by CYP2B1 (Figure 6.9)^1 Radical
atively high energy (a*) d^ iron orbital and this and cationic pathways could both explain the for-
leads to a loss of bonding across the S-Fe-0 axis. In mation of these ring-opened products, but the
general terms, the above mechanism is a specific authors favored the cationic pathway and proposed
example of the two-state reactivity paradigm that the cationic products arose from an oxidation
recently proposed as an important feature in under- mediated directly by the Fe"^-OOH intermediate.
standing organometallic reactivity (see Chapter 2)^^. Subsequent studies focused on oxidation of the
Substrate Oxidation by Cytochrome P450 Enzymes 191

CH3 CH2OH
HO
OH CHsOH

11 12 14 15
8 R = CF3
9R = H

CH3 CH2 OH
A * A - CfiHs CgHt
CeHs OCH3 OCH3 OCH3
13 GeHs OCH3

6H2
A rrocHa--^
CeHs OCH3 16 17

Figure 6.9. Probes of the P450 mechanism, showing the two rearrangement pathways for one of the probes
intended to distinguish between radical and cation intermediates.

radical clock probes 8 and 9 by the CYP2B4 wild-type and mutant enzymes in the Shaik two-state
T302A mutant, in which the threonine mutant reactivity model (see Chapter 2), or the ferryl
thought to facilitate the formation of the ferryl species may be perturbed by differential hydrogen
species by hydrogen bonding to the Fe"^-OOH bonding or other environmental factors, giving rise
complex was replaced by an alanine. CYP2B4 and to distinguishable ferryl reactivities. It is important
its T302A mutant oxidized 9 to products obtained to note, of course, that evidence for a second oxi-
by reaction with the methyl group (including unre- dizing species in the absence of the threonine that is
arranged 10 and rearranged 11) and the phenyl required for normal oxygen activation does not nec-
ring. Although the same ratio of ring opened to essarily mean that the second oxidizing species is
ring closed products was obtained with the wild- relevant to catalysis in the presence of the threonine.
type and mutant enzymes, the ratio of phenyl to In fact, cryogenic ENDOR studies with P450^^
methyl oxidation products was higher with the provide direct evidence that the ferric hydroperox-
T302A mutant. This shift in product profile was ide is not involved in camphor hydroxylation^^. In
interpreted as evidence for the involvement in the this work, the ferric hydroperoxide complex of
mutant enzyme of a second oxiding species that P450^^ was prepared at 77 K by radiolytic reduc-
preferentially oxidized the phenyl group. tion of the camphor-bound ferrous dioxygen com-
Substitution of the phenyl group in 8 with an elec- plex. EPR and ENDOR experiments then
tron-withdrawing CF3 group, which disfavors demonstrated that the hydroperoxide complex was
electrophilic attack on the aromatic ring, resulted smoothly and quantitatively converted on warming
in a change in the ratio of ring opened to ring to ~200K to a complex of the enzyme with
closed products. This change was also interpreted the normal 5-^xo-hydroxycamphor product. The
as resulting from an alteration in the nature of the hydroxyl of the 5-6xo-hydroxycamphor was coordi-
oxiding species, specifically, as evidence nated to the iron atom in the product formed imme-
for involvement of the Fe^"-OOH intermediate in diately upon warming, as expected for insertion of
methyl group hydroxylation. However, different a ferryl oxygen into a C-H bond. If the ferric
explanations are possible for the nature of the two hydroperoxide had been the oxidizing species, the
oxidizing species. For example, differences may initially formed hydroxycamphor product would
exist in the proportions of the low spin (LS) incorporate the distal oxygen of the peroxide and
and high spin (HS) ferryl species produced by the the proximal oxygen would have remained bound
192 Paul R. Ortiz de Montellano and James J. De Voss

to the iron. Coordination of the product hydroxyl to the rearranged products 12 and 13 would be
the iron would therefore require displacement of the expected. The cationic intermediate thus diverges
water ligand, an unlikely exchange reaction at 200 K. from the normal hydroxylation at some branch point
Furthermore, ENDOR studies demonstrated that the in the reaction trajectory.
C-5 hydrogen abstracted from the camphor was Two mechanisms have been considered for the
bound to the hydroxyl oxygen of the product, in formation of cationic intermediates. In the first of
accord with a ferryl insertion mechanism but not these, electron transfer from the radical intermediate
with oxidation by the ferric hydroperoxide. in the conventional oxygen rebound mechanism
The formation of products from 12 and 13 sug- occurs more rapidly than oxygen transfer and pro-
gestive of a cationic intermediate is relevant to argu- duces a cationic intermediate. Direct electron trans-
ments for participation of the Fe^"-OOH species in fer from a substrate to the P450-oxidizing species is
C-H oxidation, as insertion of the terminal proposed to occur in the oxidation of electron-rich
hydroperoxide "HO^" into a C-H bond would give nitrogen atoms (see Section 4), and a precedent
a protonated alcohol that readily explains the obser- exists for such electron transfer even in the case of
vation of (carbo)cationic rearrangements. Minor hydrocarbon hydroxylation. Thus, the product
amounts of such rearrangement products are formed in the P450-catalyzed oxidation of quadri-
obtained in the oxidation of methylcubane 12 and the cyclane 18, a hydrocarbon with a low oxidation
methylcyclopropane 13 (Figure 6.9)^^. Formation of potential, is most consistent with electron transfer to
small amounts of homocubyl alcohol 14 along with give a radical cation that is subsequently trapped by
alcohol 15 upon oxidation of the methyl group of 12 the iron-bound oxygen (Figure 6.10)^^. An alterna-
provides evidence for cationic intermediates as the tive explanation for the formation of cation
cation but not radical derived from the methyl group rearrangement products is that the oxidation is
of 12 rearranges to 14. However, one shortcoming of mediated by the P450 Fe"^-OOH complex. As
12 as a mechanistic probe, as pointed out by the already noted, insertion of the hydroperoxide oxy-
authors, is that the cationic rearrangement product is gen into a C-H bond would produce the protonated
readily identified but the products of the radical alcohol that could either undergo deprotonation to
pathway are too unstable to detect. This is not a give the alcohol or ionization to give the carboca-
shortcoming in the case of 13. Ring opening of the tion. In this mechanism, the carbocation is not an
cyclopropylcarbinyl radical derived from this probe intermediate in the hydroxylation reaction but rather
to the benzylic radical gives rise to structurally a result of decomposition of the initial product. The
related products, while the corresponding cation is observation that the proportion of cationic products
cleaved in the opposite direction to give first oxo- formed in the oxidation of 12 and 13 increases when
nium ion 16 and then aldehyde 17 (Figure 6.9). the catalytic threonine is mutated to an alanine can
These radical and carbocationic ring-opening reac- be taken as evidence for this mechanism^^, but can
tions occur with very high regioselectivity: in the also be rationalized by environmental perturbation
case of the cation reaction, the indicated ring open- of a ferryl oxidizing species. In any case, the signif-
ing is favored by > 1000:1 relative to the direction of icance of results with the mutants to catalysis by the
ring opening observed with the radical. The traces of native enzyme is questionable. A further important
aldehyde 17 observed in the oxidation of 13 by a observation is that the extent of cationic rearrange-
P450 enzyme thus provide credible evidence for at ment does not parallel the stability of the cation, as
least a minor pathway involving a cationic interme- shown by studies of a series of l-aryl-2-alkyl-cyclo-
diate. A corollary, however, is that cations cannot be propane probes with either a phenyl or/?am-trifluoro-
mandatory intermediates in normal hydrocarbon methylphenyl aryl group and an ethyl, propyl,
hydroxylation, otherwise, much higher amounts of or isopropyl alkyl group^^. Although ring opened

[ F e—m3+
=0] [Fe=0]^
Fe-O.
OHC
18

Figure 6.10. The cytochrome P450-catalyzed oxidation of quadricyclane involving an initial electron
abstraction step.
Substrate Oxidation by Cytochrome P450 Enzymes 193

products were observed with these probes, substitu- 5-dealkylation, and oxidative dehalogenation are
tion with cation-stabihzing groups yielded less rather all examples of such processes. However, although
than more rearrangement products, contrary to in appearance the outcome may be initially similar,
expectation if a protonated alcohol were the initial that is, carbon hydroxylation, the mechanisms of
oxidation product^^. This result is not consistent with hydroxylation of a simple C-H bond and a C-H
initial formation of a protonated alcohol product, and bond adjacent to a heteroatom need not be the
therefore does not support the involvement of an same. Whereas carbon hydroxylation involves
Fe"^-OOH species in hydrocarbon oxidation. In con- hydrogen abstraction from the carbon to give a
trast, the two-state radical recombination model (see transi^it carbon radical, hydroxylation adjacent to
Chapter 2) predicts that better electron donors such a heteroatom may proceed via initial electron
as a methine hydrogen will favor oxidation through abstraction from the heteroatom to give the radical
the LS manifold which proceeds without the forma- cation, deprotonation of the adjacent carbon, and
tion of a true radical intermediate, precluding ftirther recombination of the resulting carbon radical with
oxidation of the carbon to a cation. the iron-bound hydroxyl radical (Figure 6.11).
In sum, the evidence in hand is most consistent Such a mechanism is particularly feasible in the
with Shaik's two-state reactivity model for hydro- case of atoms such as nitrogen that are electron
carbon hydroxylation. Both the LS and HS elec- rich and easily oxidized. Of course, if the oxygen
tromers of the ferryl species first abstract a rebound reaction is faster than deprotonation of the
hydrogen atom. The LS species then follows a bar- adjacent carbon, the oxidation will simply result in
rierless, effectively concerted, pathway to give the oxidation of the heteroatom, as in conversion of a
unrearranged product, while the presence of a sig- trialkylamine to a trialkylamine A/-oxide or a
nificant energy barrier in the pathway for the HS dialkyl sulfide to a sulfoxide. The reaction out-
species leads to the formation of a true radical come, and the mechanism of the reaction (i.e.,
intermediate. Radical formation readily explains electron abstraction vs C-H oxygen insertion)
reaction characteristics such as high kinetic deu- would thus be expected to depend on the ease of
terium isotope effects, stereochemical scrambling, oxidation of the heteroatom and the relative ener-
and structural rearrangement, while the existence gies of the various reaction pathways.
of two parallel pathways allows the reactivity
pattern to vary both with the substrate and the
enzyme. Carbocationic species are not obligate
hydroxylation intermediates, as a much higher
extent of cationic rearrangements would be
expected if they were. Small amounts of cationic [Fe=0H]3+
products, however, apparently can be formed by a
pathway that diverges from that responsible for
normal hydroxylation, possibly by a mechanism
that involves electron transfer from a radical inter-
mediate to the active oxidizing species.
L^H^

4. Heteroatom Oxidation and


Dealkylation
[Fe=0]^

Heteroatom oxidation can be viewed as part of


the hydrocarbon hydroxylation continuum if the
reaction outcome is the introduction of a hydroxyl
group onto the carbon attached to the heteroatom,
an outcome that is usually followed by elimination Figure 6.11. The reaction manifold for the oxidation
of the heteroatom with concomitant formation of a of an amine or related nitrogen compound by
carbonyl group. 0-dealkylation, A/-dealkylation, cytochrome P450.
194 Paul R. Ortiz de Montellano and James J. De Voss

In view of the high electronegativity of oxygen 4-dihydropyridines is transferred to a nitrogen of


and, therefore, the high energy required to remove the prosthetic heme group almost certainly requires
one of its electrons, it is not surprising that the initial oxidation of the dihydropyridine nitrogen to
oxidation of a dialkyl ether occurs by direct reac- a radical cation (see Chapter 7)^^ This heme alky-
tion of the oxidizing species with the C-H bond, lation reaction occurs upon oxidation of the dihy-
although the distribution of electron density in the dropyridine within the P450 active site. However,
hydroxylation transition state is likely to be per- in incubations with liver microsomes, the dihy-
turbed by the vicinal oxygen atom. There is no dropyridine can also be oxidized by trace metals
evidence for electron abstraction from the oxygen in the solution. This adventitious oxidation
to give an oxygen radical cation, and also none for releases the 4-alkyl group as a spin-trappable free
transfer of the ferryl oxygen to an ether or alkoxy radical that obscures whatever radical release, if
oxygen to give a 1,1-disubstituted zwitterionic any, occurs in the enzyme-catalyzed reaction^^' ^^.
peroxo species. Thus, the 0-dealkylation of ethers No nitrogen radicals have been observed by EPR
with at least one C-H bond next to the oxygen is in P450 systems that are free of medium-depend-
most appropriately treated as an extension of car- ent peroxidative reactions except perhaps for the
bon hydroxylation. In the absence of a vicinal reported observation of nitrogen radicals in the
C-H bond, ether functions are resistant to P450- CYP2B1-catalyzed oxidation of para-substituted
catalyzed oxidation. dimethylanilines supported by iodosobenzene
The oxidation of nitrogen compounds gives rather than NADPH-cytochrome P450 reduc-
more diverse products than that of oxygen com- tase^^. It is possible that radical formation is
pounds, and the attendant mechanisms are more detected in this system due to a faster rate of sub-
varied and controversial. As a result of the lower strate oxidation with phenyliodosobenzene than
electronegativity of nitrogen relative to oxygen, P450 reductase, but the possibility also exists that
oxidation of a nitrogen center can result in the radicals stem from an abnormal process sup-
hydroxylation of the adjacent carbon (and thus N- ported by the artificial oxidizing agent.
dealkylation), oxidation of the nitrogen electron Differences in the deuterium isotope effects for
pair to an A^-oxide, or insertion of an oxygen into the oxidation of carbons adjacent to nitrogen vs
an N-H bond. The key mechanistic question in the oxygen suggest that the two reactions are medi-
P450-catalyzed oxidation of a nitrogen function is ated by different mechanisms. The intramolecular
whether it proceeds via initial electron abstraction isotope effect for 0-deethylation of deuterium
to give the nitrogen radical cation, followed by substituted 7-ethoxycoumarin is ~13 and for O-
either collapse to give the A^-oxide or deprotona- demethylation of 4-nitroanisole with a trideuterio
tion of the vicinal carbon to give a carbon radical methyl group is lO^'*' ^^. In contrast, an isotope
that combines with the iron-bound oxygen to give effect of 2-3 is obtained for the A^-dealkylation
the alcohol. Oxidation of an N-H bond to a of iV-methyl-A^-trideuteriomethylaniline^^' ^^. The
hydroxylamine by a mechanism analogous to that intrinsic isotope effects for 0-dealkylation thus
for carbon hydroxylation is also possible. To the approach those for normal carbon hydroxylation
extent that the nitrogen radical cation mechanism reactions, but the isotope effects for A^-dealkyla-
is operative, A^-oxide formation and A^-dealkyla- tion are much lower.
tion represent the divergent partitioning of a com- The intramolecular isotope effects observed
mon intermediate (Figure 6.11). The alternative is for A^-demethylation of para-substituted N-
that nitrogen oxidation and carbon hydroxylation methyl-A^-trideuteriomethylanilines increased from
are independent reactions, one involving reaction k^/k^ = 2.0 to 3.3 in traversing the range from the
of the ferryl oxygen with the nitrogen electron most electron withdrawing (NO2) to the most
pair, and the other a more or less classical hydroxy- electron-donating (CH3O) substituent^^. Similar
lation of the vicinal carbon. values were obtained in an earlier study^^. The
The ability of P450 enzymes to oxidize nitro- iV-dealkylation rates are also increased by elec-
gen atoms to radical cations via an initial electron tron-donating substituents^^, in accord with the
abstraction is supported by a number of experi- finding that the rates of oxidation of 12 para-sub-
mental results. The finding that the 4-alkyl group stituted A/;A^-dimethylanilines can be fit to the
of 3,5-(Z)/5)carbethoxy-2,6-dimethyl-4-alkyl-1, equation log ^^ax "" ^•^^'^ -1.02a -0.023MR
Substrate Oxidation by Cytochrome P450 Enzymes 195

+ 1.72 (r = 0.953)^^ where TT is the partition is provided by the fact that A^-demethylation is
coefficient, a, the Hammett electronic factor, and usually favored over A^-deethylation. For example,
MR, the molecular refractivity. A strong enhance- A^-demethylation is favored over A^-deethylation
ment of the reaction by electron-donating sub- by a factor of 16:1 in the CYP2B1-catalyzed oxi-
stituents is indicated by the negative sign and dation of A/-methyl-A^-ethylaniline^^. Electronic
magnitude of the cofactor of the electronic param- factors would favor deethylation if a direct
eter. These results are consistent with a mecha- hydroxylation of the carbon adjacent to the nitro-
nism involving a nitrogen radical cation. gen were involved because the incipient radical
The oxidizing species of cytochrome P450 is would be better stabilized by hyperconjugation. In
thought to have some resemblance to the well- contrast, demethylation should be favored if the
characterized ferryl species of horseradish peroxide reaction involves deprotonation of an initially
(HRP). It is therefore relevant that the rates of reduc- formed nitrogen radical cation because a methyl is
tion of the HRP Compound I by /?ara-substituted more acidic than an ethyl methylene. However,
AyV-dimethylanilines and AyV-di(trideuteri- these arguments are not unambiguous because the
omethyl)anilines correlate with the oxidation poten- electronic differences in the two reactions may be
tials of the anilines^^' ^^, and that no kinetic isotope masked by the differences in the steric effects of a
effects are observed in these reactions^^. Earlier methyl and an ethyl group. Thus, steric effects
studies measuring the rates of product formation possibly account for the observation that N-
rather than the reduction of the ferryl species led to demethylation of A^-methyl-iV-alkyl-4-chloroben-
the conclusion that dimethylaniline A^-demethyla- zamides is favored over A^-deethylation by a factor
tion by HRP is subject to large isotope eflFects^"^' ^^^. of 2.2, as this reaction (see below) is thought to
However, in the case of HRP, product formation involve direct oxygen insertion into the C-H
involves a disproportionation reaction subsequent to bond^^^ Despite this caveat, the observation of
radical cation formation that is subject to a large iso- intramolecular isotope effects <2.0 in the N-
tope effect. The more recent results of Goto et alP dealkylation of A/;A^-dimethylaniline by CYP2B1,
are most consistent with a single electron transfer chloroperoxidase, and metalloporphyrin models,
(SET) mechanism for the A/-dealkylation mediated but of isotope effects > 8 in the corresponding
by HRP. A similar dependence of the 7V-dealkylation reactions catalyzed by hemoglobin, HRP, and
reaction on the substrate oxidation potential was prostaglandin H synthase, provides independent
observed in the reactions mediated by evidence that the iron-bound oxygen removes a
TMP+«Fe^^=0 (TMP = 5,10,15,20-tetramesityl- proton from the carbon adjacent to the nitrogen rad-
porphyrin dianion), but with this P450 model sys- ical cation in the first but not the second set of pro-
tem, kinetic isotope effects of 3.9 (P-CH3O) to 6.2 teins (Figure 6.12)^^^. The pK^ values of the protons
(P-NO2) and intramolecular isotope effect of next to the trimethylamine and dimethylaniline
1.3-5.9 for the corresponding A^-methyl-A/-trideu-
teriomethylanilines were observed^^. The authors
argue that isotope effects are observed in this
instance due to a competition between back electron ^CH2CH3 ^p^^op. CH2CH3
transfer from the partially reduced TMPFe^^=0 R-N ^ R-V
species to the nitrogen radical cation and oxygen CH3 CH.
transfer to the nitrogen radical cation. The implica-
tion of a SET mechanism in both the HRP and P450
model reactions agrees with earlier findings on the [Fe=Of-
rates of oxidation of dimethylaniline by HRP,
CYP2B1, and two metalloporphyrin systems. CH2CH3
CH2CH3
Unfortunately, similar spectroscopic rate studies R-N
cannot be carried out with P450 itself because the R-N
CH2OH
analogous "Compound F' form of cytochrome P450 CH2
[Fe—OHf
is not sufficiently stable.
Support for a nitrogen radical cation mecha- Figure 6.12. The nitrogen radical cation pathway
nism in P450-catalyzed A/-dealkylation reactions proposed for a P450-catalyzed A^-dealkylation reaction.
196 Paul R. Ortiz de Montellano and James J. De Voss

nitrogen radical cations have been estimated to be, observed if the entropy factor is similar for a series
respectively, ~15 and 9^^^, 103 of substrates and thus cancels out, but caution must
It has been proposed that the isotope effects for be exercised in interpreting such correlations.
the P450-catalyzed oxidations of hydrocarbons Cyclopropylamines can, in principle, be used to
and alkylamines are similar to those observed in probe the mechanism of nitrogen oxidation because
the reactions of the same substrates with the tert- a cyclopropyl substituent on a nitrogen radical
butoxyl radical^^' ^^^. The finding that the meas- cation can undergo ring-opening reactions analo-
ured kinetic isotope effects for the hydrogen gous to those of a cyclopropyl attached to a carbon
abstraction from benzylic methyl groups fall on radical. The inactivation of P450 enzymes by cyclo-
the same line as those for the A^-demethylation of propyl amines was postulated in early studies to
4-substituted dimethylanilines has, therefore, been involve formation of the nitrogen radical cation, ring
advanced as evidence that iV-dealkylations occur cleavage to give an iminium carbon radical species,
via a hydrogen abstraction (HAT) rather than SET and alkylation of the heme group ^^^' ^^^. The obser-
mechanism, in contrast to the evidence for a SET vation of a correlation between the one-electron oxi-
mechanism provided by the already discussed iso- dation potentials and the rates of P450 inactivation
tope effect and rate data. Recent studies of the by a series of nitrogen-, oxygen-, and halide-
rates and isotope effects in the reactions of deuter- substituted cyclopropanes offers circumstantial sup-
ated 1 -methyl-4-phenyl-1,2,3,6-tetrahydropyridines port for an electron abstraction mechanism^ ^^.
with the ^^r^butoxyl radical suggest, however, Hanzlik has recently examined the oxidation of
that the rer^butoxyl radical may not fully mimic cyclopropylamine probes by HRP, an enzyme that
the enzymatic oxidizing species, as the tert- demethylates A^,A^-dimethyl- and A^-methyl,A/-iso-
butoxyl radical did not discriminate between C-H propylaniline in the presence of H2O2 and 02^^^.
bonds that differed in bond strength by as much as A^-cyclopropyl,A^-methylaniline (19) was oxidized
10 kcal /mol~^ ^^^. A correlation of reaction rates to both A^-methylaniline and a cyclized product
with bond dissociation energies for a range of that derives from a radical-based ring opening of
alkylamine C-H bonds indicated that entropy fac- the cyclopropyl group (Figure 6.13). However,
tors make a significant contribution to the rate con- cyclopropanone was obtained as the product in the
stant. The poor correlation between the absolute P450-catalyzed oxidation of N-methyl, iV-(l-alkyl-
rates and C-H bond strengths is caused by differ- cyclopropyl)aniline''^. No trace of radical cation
ences in the entropy required to align the C-H products such as those obtained when the same sub-
bond to be broken with the electron pair on the strate was oxidized by HRP were detected. The
adjacent nitrogen. A correlation between isotope results suggest that direct oxygen insertion into the
effects and bond strength may nevertheless be cyclopropyl group occurs faster than oxidation of

Me Me Me
/ HRP / " ^ +•/
V_/ VVA
<
19

Ph^""^^CHO

Figure 6.13. Probes designed to investigate whether a nitrogen radical cation is involved in P450-catalyzed
iV-dealkylation reactions.
Substrate Oxidation by Cytochrome P450 Enzymes 197

the nitrogen, and thus is more consistent with a HAT and supported by H202^^^. However, these criteria
than a SET mechanism. do not differentiate between the ferric hydroper-
4-Phenyl-rra«5'-l-(2-phenylcyclopropyl)-l,2, oxide and ferryl species, as one is the precursor of
3,6-tetrahydropyridine (20) is oxidized by rat hver the other. In the absence of more direct evidence,
microsomes to cinnamaldehyde and A/-dealky- it is not possible to determine whether A^-oxide
lated tetrahydropyridine in addition to conven- and hydroxylamine formation proceeds by a
tional metabolites (Figure 6.13). The first two mechanism other than reaction with the ferryl
metabolites have been postulated to be formed via species to give a transient nitrogen radical cation
the nitrogen radical cation, cyclopropyl ring open- intermediate.
ing, electron abstraction, proton elimination to The conversion of thioethers to sulfoxides or
form the double bond, and hydrolysis of the iS-dealkylated products, as noted for the oxidation
iminium link to release the aldehyde^^^ of amines, could involve the formation of a tran-
The oxidation potential for an amide nitrogen sient sulfur radical cation or direct oxygen trans-
is higher than that of an amine due to the electron- fer to either the sulfur or the adjacent carbon. If
withdrawing effect of the carbonyl group. The any function can be oxidized by direct reaction
P450-catalyzed 7V-dealkylation of amides with a with the P450 ferric hydroperoxide species, it
deuterated and undeuterated A^-methyl substituent, would appear to be a thioether sulfur. The ratio
RCON(CH3)(CD3), are subject to intramolecular of *^-dealkylation to sulfoxidation products was
isotope effects of 4-7^^^' ^^^. The corresponding reported in early work to correlate well with the
isotope effect for the iV-dealkylation catalyzed acidity of the protons adjacent to the sulfur^^^.
by a model porphyrin was 5.6^^^, a much higher Furthermore, electron-donating groups modestly
value than that observed for the electrochemical accelerate the rate of formation of sulfoxides from
reaction that proceeds via a nitrogen radical cation substituted thioanisoles (Hammett p^ = —0.16),
intermediate^^^. These results suggest that amide and of the sulfoxides to the corresponding
A^-dealkylation occurs by direct carbon hydroxyla- sulfones (Hammett p+ = -0.2)^2^' ^^^ in an
tion as a result of the higher oxidation potential of intramolecular competition experiment, it has
the amide nitrogen. been found that the thioether of thianthrene-5-
P450-catalyzed oxygen transfer to amines to oxide is oxidized in preference to the symmetry-
give the N-oxidQ or 7V-hydroxyl product is gener- related thioether sulfoxide function, confirming
ally considered to involve nitrogen radical cation the expected higher reactivity of the sulfide than
formation followed by recombination with the sulfoxide ^^^. Unfortunately, although these results
ferryl oxygen (Figure 6.11)^^^' ^^^. As these reac- indicate that sulfoxidation occurs most readily at
tions are less amenable to direct investigation with electron-rich sulfur atoms, the magnitudes of the
mechanistic probes, the postulate of a radical effects are such that they cannot be used to unam-
cation mechanism rests largely on the evidence for biguously differentiate between radical cation and
such intermediates in A^-dealkylation reactions. oxygen transfer mechanisms for sulfur oxidation.
However, the reported absence of a systematic Bacciochi et al. have shown that a radical
relationship between the electronic properties of cation localized on the trimethoxy-substituted
substituents and the rates of oxidation of anilines phenyl ring is generated when a thioether, with a 2-
and dimethylanilines to hydroxylamines and (3,4,5-trimethoxyphenyl)ethyl on one side and a
A^-oxides, respectively, provides no support for phenyl group on the other, is chemically oxidized.
such a mechanism^^^' ^^^. One as yet unproven They have then shown that liver microsomes
explanation for the absence of a correlation is that exclusively oxidize a thioether with a 3,4,5-
the stability of the 7V-oxide-iron complex makes trimethoxyphenyl group on one of the sulfur-
dissociation of the A^-oxide partially rate limiting^ ^^. bearing carbons to a sulfoxide rather than to the
Hlavica has also proposed that iV-oxide formation products expected from formation of the trimetho-
is mediated by the P450-ferric hydroperoxide xyphenyl radical cation^^"^. In view of the finding
rather than ferryl species based, in part, on the that chemical oxidation yields the trimetho-
observation that the oxidation of A^,A/-dimethylani- xyphenyl radical cation, they conclude that the sul-
line to the corresponding A^-oxide mediated by foxide is formed by direct oxygen transfer from the
CYP2B4 is both inhibited by superoxide dismutase P450 to the sulfur because oxygen rebound to the
198 Paul R. Ortiz de Montellano and James J. De Voss

sulfur should be slow if the unpaired electron is not the olefin stereochemistry, as illustrated by results
localized on the sulfur. In contrast, HRP gives both on the epoxidation of c/5-stilbene^^^, oleic acid^^^,
sulfoxidation and radical cation cleavage products, and ^ra«5-[l-^H]-l-octene^^^ supports the view
but only the sulfoxide is formed with chloroperox- that the reaction occurs by a concerted mecha-
idase^^^. However, these studies all assume that the nism. However, retention of stereochemistry does
ferryl oxidation is equivalent to a chemical oxida- not preclude a nonconcerted mechanism, a point
tion in favoring the trimethoxyphenyl ring over the clearly made by the fact that the stereochemistry
sulfur. It is possible that the P450-oxidizing is retained in most carbon hydroxylation reactions
species oxidizes the sulfur to the radical cation and even though they are mediated by a stepwise rad-
recombines with it faster than the electron can be ical mechanism. Early isotope effect studies also
transferred to the trimethoxyphenyl ring. A similar provided evidence for a nonconcerted, or at least
caveat tempers the conclusions that can be drawn asynchronous, reaction mechanism^^^. Thus, sub-
from the finding that phenyl cyclopropyl sulfide is stitution of a deuterium on the internal but not ter-
oxidized by Mortierella isabellina to the sulfoxide minal carbon of the exocyclic double bond of
without opening of the cyclopropyl ring^^^. p-methyl- and /?-phenylstyrene led to the observa-
In sum, the course of heteroatom oxidation tion of an inverse secondary isotope effect k^k^^ =
appears to be sensitive to the oxidation potential of 0.93 in the epoxidation reaction. Similar isotope
the heteroatom, the acidity of hydrogens on the effects would be expected at both carbons if the two
adjacent carbon, and steric factors. The bulk of the carbon-oxygen bonds were formed simultaneously.
evidence suggests that oxidation of the nitrogen in However, differential secondary isotope effects can
amines generally involves electron abstraction fol- be observed if one carbon-oxygen bond is formed
lowed primarily by A/-dealkylation if a labile proton to a significantly greater extent than the other in an
is present, or nitrogen oxidation if it is not. As the asynchronous epoxidation transition state. This is
nitrogen oxidation potential increases, there is a clearly shown by the finding that olefin epoxidation
shift toward direct insertion into the C-H bond, as by m^fa-chloroperbenzoic acid, a well-established
is thought to occur in the A/-dealkylation of amides. concerted reaction, also gives differential second-
0-dealkylation reactions are mediated by direct ary isotope effects, in this instance, the isotope
insertion of the oxygen into the vicinal C-H bond, effect being seen on the terminal but not internal
as electron abstraction from the oxygen is too diffi- carbon^ ^^
cult due to the high oxygen oxidation potential. Acetylenes, like olefins, have oxidizable TT-
Transfer of the P450 ferryl oxygen to an oxygen bonds, although it is harder to oxidize an
atom to give a peroxide is not known, presumably acetylenic than an olefinic ir-bond because the
for the same reason. The mechanisms of sulfur oxi- triple bond is shorter and stronger. Nevertheless,
dation remain more uncertain, but the limited evi- cytochrome P450 readily oxidizes terminal
dence suggests that sulfur dealkylation may occur acetylenic bonds to give ketene products in which
via direct insertion into the vicinal C-H bond, as the terminal hydrogen of the acetylene has
found for 0-dealkylation, in a reaction that diverges migrated quantitatively to the internal acetylenic
from that responsible for sulfoxidation. carbon (Figure 6.14)'^^' '^^. The oxirene that would
result from "epoxidation" of the triple bond has not
been detected and may not form, as oxirenes are
highly unstable moieties. If formed, they would be
5. Olefin and Acetylene expected to rearrange to the observed products.
Oxidation The finding of a substantial kinetic isotope effect
on formation of the arylacetic acid metabolites
No critical experimental advances have been when the terminal hydrogen of the arylacetylene is
made in the past decade toward a fuller under- replaced by a deuterium indicates that the hydro-
standing of the mechanisms of P450-catalyzed gen migration occurs in the rate-determining step
epoxidation reactions, although new insights into of the catalytic process^^^' ^^^. This finding, and the
the process are emerging from computational observation that the same products are formed
studies. The finding that P450-catalyzed olefin with similar isotope effects in the oxidation of aryl
epoxidations invariably proceed with retention of acetylenes by m-chloroperbenzoic acid^^^' ^^^,
Substrate Oxidation by Cytochrome P450 Enzymes 199

Figure 6.14. Schematic mechanism for the oxidation of a terminal acetylene by cytochrome P450, showing that
addition at the terminal end of the triple bond leads to a ketene product, whereas addition to the internal carbon
results in alkylation of a nitrogen of the heme group.

O )
N-Il--:^N
N

Figure 6.15. Alkylation of a nitrogen of the heme during the P450-catalyzed oxidation of a terminal double bond.

suggest that the hydrogen migrates to the vicinal addition of the porphyrin nitrogen to the terminal
carbon as the oxygen is transferred to the terminal carbon of the epoxide, control experiments have
carbon in a concerted reaction process. The clearly established that epoxides do not alkylate the
demonstration that 2-biphenylpropionic acid is a heme group^^^. Furthermore, the stereochemistry of
minor product in the CYPlAl- and CYP1A2- the A^-alkylated products is opposite to that expected
catalyzed oxidation of 4-(l-propynyl)biphenyl from backside attack of the nitrogen on the epox-
indicates that it is also possible to oxidize a triple ide^^^. The heme alkylation must, therefore, occur
bond with the migration of an alkyl group rather prior to formation of the epoxide metabolite. Given
than a hydrogen^^^. the requirement for catalytic turnover of the
Two types of evidence, again based on relatively enzyme^^^ and the fact that the resulting 7V-alkyl
early experiments, provide serious support for the group has incorporated an atom from molecular
availability of a nonconcerted olefin epoxidation oxygen^^^' ^^^, it is clear that the heme is alkylated
pathway. Thus, the observation that terminal olefins, by a reactive intermediate generated during the
including ethylene gas, are not only oxidized to the olefin epoxidation reaction (Figure 6.15). The reac-
corresponding epoxides but simultaneously alkylate tive species that alkylates the heme must be a pre-
one of the prosthetic heme nitrogen atoms is incom- cursor of the epoxide product, or the result of a
patible with a concerted epoxidation mechanism parallel but divergent olefin oxidation pathway. The
(Figure 6.15) (see Chapter 7)^^^. Although the struc- partitioning of the common intermediate, or of the
ture of the adducts is that which would result from flow of oxidation equivalents into two parallel, but
200 Paul R. Ortiz de Montellano and James J. De Voss

distinct, pathways is defined by the partition ratio a few olefins. As a case in point, the oxidation
between epoxidation and heme alkylation. This par- of trichloroethylene yields both trichloroethylene
tition ratio ranges from values as low as 1-2 (i.e., oxide and trichloroacetaldehyde. As control experi-
nearly every oxidation leads to heme alkylation) to ments indicated that trichloroethylene oxide did not
values as high as several hundred (i.e., heme alkyla- rearrange to trichloroacetaldehyde under the incu-
tion competes poorly with epoxide formation). bation conditions, the aldehyde apparently arose by
Prosthetic heme alkylation is also observed an oxidation pathway distinct from that which gen-
during the oxidation of terminal acetylenes by erated the epoxide (Figure 6.16)^"^^' ^^^. Similar
cytochrome P450 (Figure 6.14). As found in inac- results were obtained for the oxidation of 1,1-
tivation by olefins, the terminal carbon of the dichloroethylene to the epoxide vs monochloro- and
acetylene is bound to a nitrogen of the P450 heme dichloroacetic acids—again the epoxide did not
group and an atom derived from molecular oxy- appear in control experiments to rearrange to the
gen to the internal carbon^^^' ^^^. The oxygen is acids under physiological conditions ^'^^. Direct for-
present as a carbonyl group due to tautomerization mation of a carbonyl product during the oxidation
of the enol that would be formed by simple addi- of an olefin has been observed in a few other
tion of a hydroxyl group to the internal acetylenic situations, notably in the formation of 1-phenyl-1-
carbon. In the case of acetylene oxidation, a clear butanone and l-phenyl-2-butanone as minor prod-
distinction is possible between the reaction path- ucts of the oxidation of ^aw^-l-phenylbutene^"^^,
way that produces the ketene metabolites and that and of 2-phenylacetaldehyde in the oxidation of
which yields the A^-2-ketoalkyl adducts because styrene^'*^. Under physiological conditions, these
metabolite formation involves delivery of the oxy- carbonyl products do not appear to be formed by
gen to the terminal carbon, but 7V-alkylation deliv- rearrangement of the epoxides. The carbonyl prod-
ery to the internal carbon. In the case of acetylenes, ucts are consistent with the formation of a carboca-
enzyme inactivation can also occur by a different tion intermediate, possibly through leakagefromthe
mechanism subsequent to metabolite formation normal epoxidation pathway into an alternative
because the initial ketene product can acylate nucle- pathway within the overall reaction manifold. This
ophilic protein residues before it is hydrolyzed to a could occur, for example, if a competition exists
stable carboxylic acid (see Chapter 7)'"^^' *'*^. The between closing the second epoxide bond and
partition ratios for metabolite formation vs heme electron transfer from a radical-like carbon interme-
alkylation are usually smaller for acetylene than for diate to the iron to give the cation. However, the
olefin oxidation. search for products indicative of radical intermedi-
The second type of evidence that strongly argues ates in olefin epoxidation reactions has so far been
for the availability of a nonconcerted epoxidation fruitless. Thus, no cyclopropane ring-opened prod-
pathway is provided by the finding that carbonyl ucts were observed in the oxidation of trans-\-
products are directly formed during the oxidation of phenyl-2-vinylcyclopropane ^ ^^.

ci
CCI3CH0 +
Cl
o

H o
Cl
Cl

Cl
Cl

o
Cl
jFe-
N N^- N

Figure 6.16. The oxidation of some halogenated olefins has been shown to yield the corresponding epoxides and
rearrangement products that appear to arise via a reaction path that does not include the epoxide as an intermediate.
Substrate Oxidation by Cytochrome P450 Enzymes 201

The seemingly contradictory evidence for con- PorFei^-0-CH2CH2, and a barrier of 7.2 kcal
current concerted and nonconcerted epoxidation mol"^ for the quartet intermediate with the
pathways can be satisfyingly rationahzed by the Por+Te"^-0-CH2CH2 structure^^l These energy
two-state reactivity paradigm advocated by Shaik barriers are sufficiently high to allow alternative
and colleagues (see Chapter 2). These investigators reactions to compete with epoxide ring closure and
have shown by density functional theoretical calcu- provide a ready explanation for the experimental
lations that the P450 ferryl porphyrin radical cation observation that the heme undergoes nitrogen alky-
can exist in doublet and quartet spin states that are lation in the epoxidation of some olefins^^^. The
quite close in energy. The calculations suggest that partitioning between epoxidation and heme alkyla-
ethylene epoxidation by both the doublet and quar- tion is largely determined in this model by the pro-
tet oxidizing species involves addition of the oxy- portion of the doublet and quartet transition-state
gen to one carbon of the olefin, leaving the other complexes. The critical feature of the two-state
carbon of the olefin with an unpaired electron epoxidation mechanism is the presence of a doublet
(Figure 6.17)^'^^. This radical complex can again and a quartet transition state of sufficiently close
exist in both doublet and quartet states, although energies that the oxidation reaction can proceed via
the system is more complex in that each of the two either of the two transition states. This computa-
states has two equilibrating electromers that differ tional model rationalizes the experimental data
in whether the electron contributed by the olefin is available on olefin oxidation in a satisfying and
used to neutralize the porphyrin radical cation or to comprehensive manner, although the model is
reduce the iron to the Fe"^ state (see Figure 6.8 and difficult to test experimentally.
Chapter 2). The critical difference between the two The discussion of epoxidation has been
states is that there is no barrier to closure of the framed in terms of a ferryl catalytic species. It
doublet state to the epoxide, so that epoxidation has been proposed that the ferric hydroperoxo
occurs via an almost concerted trajectory with intermediate may contribute to olefin epoxida-
retention of the olefin stereochemistry even though tion"^^, but as discussed in Section 2, the support
a true, concerted mechanism is not predicted by the for this postulate is contradictory. Although it
calculations^'^^. In contrast, a barrier of 2.3 kcal appears that the ferric hydroperoxo intermediate
mol~^ is found for closure to the epoxide of the can oxidize double bonds at a low rate, the data
quartet intermediate with the configuration strongly suggests that the ferric hydroperoxide

//,,

^ , O
>Y7<
I barrierless o
— Fe- I
I
RS RS
doublet
state ,.,\\\
O H2C=CH2
II
-Fe-
I
^\7^O
RS quartet I
state _ F e
I
RS
o
I
-Fe-
I
RS

Figure 6.17. Explanation of the dual pathways of olefin oxidation resulting in epoxide formation and heme
alkylation in terms of the two-state hypothesis of P450 catalysis extensively described in Chapter 2.
202 Paul R. Ortiz de Montellano and James J. De Voss

makes little contribution to oxidations catalyzed a ketone intermediate. Tautomerization of this


by the wild-type proteins. ketone yields a phenol product. This sequence of
Although the stereochemical evidence sug- steps is the so-called "NIH-shift" (Figure 6.18)'^^.
gests that olefin oxidation occurs by a concerted Key evidence for this mechanism is provided by
mechanism, it is clear from the observation of the finding that the hydrogen atom (H*) at the
heme alkylation and rearranged products that non- position that is oxidized migrates to the adjacent
concerted oxidation pathways are also operative. carbon, where it is partially retained in the final
The oxidation of terminal acetylenes to ketenes by phenol product. Partial retention of the migrating
addition of the oxygen to the unsubstituted carbon hydrogen reflects nonstereospecific elimination of
and to species that alkylate the heme group by one of the two hydrogens in the tautomerization
addition to the internal carbon also suggests that step. This mechanism is widely applicable, but
multiple oxidation pathways are possible. The ferryl oxygen transfer to aromatic rings can also
puzzle that is posed by these mechanistic proceed via a transient intermediate that under-
dichotomies may find a solution in the recently goes the NIH shift or eliminates a substituent on
formulated hypothesis of two-state reactivity, in the tetrahedral carbon before the epoxide is actu-
which two energetically similar transition states ally formed (vide infra). The migrating atom in the
are obtained, one of which is in a doublet spin NIH shift is usually a hydrogen, but other moi-
state and reacts essentially as if the reaction were eties, notably a halide or an alkyl group, can also
concerted, and the second of which is in a quartet shift to the adjacent carbon as the result of the
spin state and can give rise to products character- hydroxylation event'^^' '^^.
istic of nonconcerted reactions (see Chapter 2). The rate of the hydroxylation reaction is not
very sensitive to deuterium substitution because
the deuterium-sensitive tautomerization step
occurs after the rate-limiting enzymatic oxidation.
6. Oxidation of Aromatic Rings The observation of a small inverse secondary iso-
tope effect (0.83-0.94) for ring hydroxylation of
The oxidation of an aromatic ring by ortho- and p^ra-xylene is consistent with rate-
cytochrome P450 invariably involves oxidation of limiting addition of the ferryl oxygen to the ir-
one of the ir-bonds rather than direct insertion of bond, as the transition state for the addition
the oxygen into one of the aromatic ring C-H reaction requires partial rehybridization of the car-
bonds. Thus, benzene oxide has been specifically bon from the sp^- to the sp'^-state'^^. The observa-
identified as a product of the oxidation of benzene tion of an inverse isotope effect does not support a
by liver microsomes'^^ However, benzene oxide mechanism in which the rate-determining step is
and the similarly unstable epoxides expected from oxidation of the aromatic ring to a ir-cation radi-
the oxidation of other aromatic rings readily cal, as the secondary isotope effect for such a
undergo heterolytic cleavage of one of the epoxide process should be negligible. The oxidation of
C-0 bonds. This bond cleavage is followed by cyclopropylbenzene to 1-phenylcyclopropanol
migration of a hydride from the carbon retaining and cyclopropylphenols without detectable open-
the oxygen to the adjacent carbocation to give ing of the cyclopropyl ring also argues against the

[Fe=or

H*(H)

Figure 6.18. The NIH shift involving initial formation of an epoxide metabolite in the oxidation of the aromatic
ring by cytochrome P450. The starred hydrogen shows that the hydrogen undergoes a 1,2-shift and then is partially
lost in the final tautomerization step.
Substrate Oxidation by Cytochrome P450 Enzymes 203

involvement of a radical cation intermediate in the or by an jip^o-substitution mechanism that obvi-


oxidation of small, unactivated aromatic rings ^^^. ates the epoxide intermediate (vide infra).
In some instances, particularly in hydroxyla- The cytochrome P450-catalyzed oxidation of
tions meta to a halide substituent, the hydrogen on pentafluorochlorobenzene to tetrafluorochlorophe-
the hydroxylated carbon is quantitatively lost (i.e., nol has been proposed to involve ferryl oxygen
there is no NIH shift), and a small deuterium addition to the fluorine-substituted carbon para to
kinetic isotope effect is observed^^^' ^^^. These the chloride, followed by electron donation from
hydroxylations could result from direct oxygen the chloride to eliminate the fluorine as a fluoride
insertion into the C-H bond, as in a true "hydrox- ion (Figure 6.19). The resulting positively charged
ylation" mechanism, but they are more likely to chloronium intermediate can then be reduced to
result from oxidation of the aromatic ring without the phenol, possibly by cytochrome P450 reduc-
the formation of a discrete epoxide intermediate. tase, or can undergo hydrolysis to the tetrafluoro-
Isotope effect studies with deuterated benzenes quinone^^^. This mechanism is supported by
bearing a variety of substituents have shed some ^^F-NMR evidence for the release of fluoride ion
light on this process^^^' ^^^. A small, normal iso- and by molecular orbital calculations. A correla-
tope effect is observed for me^a-hydroxylation tion of molecular orbital calculations with the
when deuterium is located meta- to the halogen in regiochemistry of the oxidation of 1-fluoroben-
chlorobenzene {k^kj^ = 1.1-1.3), but a small, zene, 1,2-difluorobenzene, 1,3-difluorobenzene,
inverse isotope effect (k^/k^ = —0.95) is observed 1,2,3-trifluorobenzene, and 1,2,4-trifluorobenzene
for ortho' and /7«ra-hydroxylation when the deu- suggests that the reaction is initiated by direct
terium is at those positions^^^. Simultaneous for- attack of the electrophilic ferryl oxygen on the aro-
mation of the two epoxide bonds in a concerted matic TT-system rather than by an initial electron
process should be subject to a small, normal iso- abstraction^ ^^ More refined local density approxi-
tope effect when either of the two oxidized carbons mation calculations for the oxidation of benzene
bears a deuterium atom, although asynchronous and mowo-fluorobenzene suggest that epoxidation
formation of the two bonds could give rise to dif- is disfavored vs a direct NIH shift from a tetrahe-
ferent isotope effects at the two sites. In the limiting dral oxygen-addition intermediate, and that
situation in which one carbon-oxygen bond is hydroxylation/?ara to the fluorine is favored^^^. In
completely formed first, formation of this bond addition to the electronic effect of the halide, a
could be followed either by closure to the epoxide steric interference is observed in the ability of the

Figure 6.19. Oxidation of a polyhalogenated ring via an ipso mechanism that does not involve formation of an
epoxide metabolite but rather an addition-elimination reaction that directly yields a quinone product.
204 Paul R. Ortiz de Montellano and James J. De Voss

enzyme to hydroxylate ortho to the halide when it is Furthermore, the methyl in 4-methylphenol is not
a bromine or iodine but not fluorine or chlorine ^^^. a viable leaving group and this compound is sim-
The oxidation by rat liver microsomes of phe- ply oxidized to 4-hydroxy-4-methy 1-2,5-cyclo-
nols bearing a para-0V\^02, -NO2, -CN, hexadiene-1-one^^^. It is to be noted that the
-CH2OH, -COCH3, -COPh, -CO2H, -F, -CI, or -Br regiochemical results do not rule out epoxide for-
substituent eliminates the /?ara-substituent and mation, as electron donation from the phenolic
forms the hydroquinone (Figure 6.20)^^'^' ^^^. hydroxyl group would regiospecifically open the
Studies with ^^62 show that an atom of molecular epoxide to the same products, but epoxide forma-
oxygen is incorporated into one of the two tion appears an unlikely explanation for the col-
quinone carbonyl groups. These results, and the lective results.
finding that the /7-nitrophenoxy group is not elim- Evidence for aniline oxidation without the for-
inated when the phenol hydroxyl is replaced by a mation of an epoxide intermediate is provided by
methyl ether, suggest that the phenoxy radical the demonstration that an atom of molecular oxy-
generated by one-electron oxidation of the phenol gen is incorporated into the quinone oxygen when
undergoes /p^o-recombination with the ferryl />-ethoxyacetanilide (phenacetin) is oxidized to iV-
oxygen to give a tetrahedral intermediate. Direct acetyl-/7-benzoquinoneimine^^^'^^^. This finding
elimination of the /7flra-substituent then gives the requires that the reaction proceeds with cleavage of
quinone. However, it has also been reported that the bond between the oxygen and the aryl carbon
4-iodoanisole is oxidized to 4-methoxyphenol rather than via a conventional 0-dealkylation
without the incorporation of label from H2^^0 or mechanism. The most probable mechanism for this
^np^^^. This finding argues that the phenol reaction is P450-mediated hydrogen abstraction
hydroxyl is not absolutely required for the reac- from the nitrogen to give a radical that undergoes
tion, so the addition can occur via //750-addition z/75(9-recombination with the ferryl oxygen at the
without prior formation of the phenoxy radical. In carbon bearing the ethoxy moiety (Figure 6.21)^^^.
accord with an rp^o-mechanism, the substituent is The concurrent formation of 2-hydroxyphenacetin
eliminated from 4-halophenols as a halide anion, is consistent with this mechanism because high
a /?ara-CH20H group as formaldehyde, and unpaired electron density would be present in
a PhCO-substituent as benzoic acid^^^'^^. the aniline radical on both the ortho- and para-
carbons''^^. The formation of quinoneimines
accompanied by the elimination of fluoride anion
in the oxidation of 4-fluoroanilines can be
explained by a similar mechanism'^'.
ArO' The oxidation of phenols via HAT from the
hydroxyl group (or sequential electron transfer
and deprotonation) is supported by data on the
oxidation of estradiol and estrone. In accord with
a key role for the phenolic hydroxyl group, the
predominant orf/zo-hydroxylation of estradiol
does not occur when the phenolic hydroxyl is
replaced by a methyl ether^^^. Early experiments
ArO'
established that 2-hydroxylation of estradiol
occurs without a detectable NIH shift^^^. More
recent work has shown that, whereas estrone is
converted to both 2- and 4-hydroxyestrone by
CYP3A4, conjugation of an additional aromatic
ring, as in equilenin and 2-naphthol, leads exclu-
sively to 4-hydroxylation of estrone and 1-hydrox-
Figure 6.20. Two possible pathways for the direct ylation of 2-naphthol. In both these reactions, the
P450-catalyzed oxidation of a /?-aryloxy phenol to the site that is exclusively hydroxylated is that
quinone, one involving initial formation of a phenoxy expected to carry the greatest share of the
radical and the other of an epoxide. unpaired electron density if the initial step is
Substrate Oxidation by Cytoclirome P450 Enzymes 205

NHCOMe •NCOMe NHCOMe NHCOMe

[Fe=0*]^-'

OEt OEt

NHCOMe NHCOMe NHCOMe


0*H

^* H O O epL^
OEt ^^^

Figure 6.21. Possible mechanisms for the oxidation of phenacetin.

HO

HO

higher energy

Figure 6.22. The additional aromatic ring in equilenin leads to a change in regiochemistry of hydroxylation of the
phenol ring relative to that observed in the oxidation of estrone, possibly due to a change in the localization of the
unpaired electron density from the 2- to the 4-position.

formation of the phenoxy radical, followed by plant cytochrome P450 enzymes, including the
recombination with the iron-bound hydroxy! radi- enzyme from Berberis stolonifera that catalyzes
cal (Figure 6.22)i^4 the biosynthesis of dibenzylisoquinoline alkaloids
The cytochrome P450-catalyzed formation of (Figure 6.23)^^^' ^^^, and the enzymes that convert
phenol radicals is clearly required for the cross- reticuline to salutaridine^^^' ^^^, and autumnaline
linking of phenol rings catalyzed by a variety of to isoandrocymbine in colchicine biosynthesis^^^.
206 Paul R. Ortiz de Montellano and James J. De Voss

Another interesting example is provided by the


therapeutically important antibiotic vancomycin

-tx.
(Figure 6.24), which consists of a crosslinked hep-
tapeptide backbone glycosylated with a disaccha-
ride residue. The phenolic coupling that occurs
OH
between the aromatic side chains of the heptapep-
MeO.
tide core is believed to be mediated by a P450
enzyme. The vancomycin biosynthetic cluster
encodes three highly related P450 enzymes^^^ that
are suggested by gene knockout studies to be
involved in the coupling of residue 4 with resides
MeO. 2 and 6 via C-0 bonds and of residues 5 and 7 via
a C-C bond'^'. One of these enzymes (P450Q^yB)
has been cloned and overexpressed in Escherichia
coli and an X-ray crystal structure obtained. It has
NMe
a relatively open active site, consistent with a large
substrate, but whether the substrate is the free
MeO'
heptapeptide or one bound to a peptidyl carrier
domain is unclear^ ^^. Balhimycin, chloroere-
momycin*^^' ^^^, and complestatin^^"^ are antibiotics
Figure 6.23. The formation of phenoxy radicals is structurally related to vancomycin in which analo-
indicated by the couphng products formed in the P450- gous C-C and C - 0 bond formation is believed
catalyzed biosynthesis of some alkaloids. to be P450 mediated. These reactions, like the

©
1 .NH2CH3

0OOC

OH

NHo

Figure 6.24. The biosynthesis of vancomycin involves a phenoxy radical crosslinking step that is catalyzed by a
cytochrome P450 enzyme.
Substrate Oxidation by Cytochrome P450 Enzymes 207

MeO. products were 9-hydroxymethylanthracene, 10-


methyl-lO-hydroxy-9-anthrone, and anthraqui-
none^^^. The 9-hydroxymethyl product results from
a straightforward carbon hydroxylation, but more
complex reactions are required to rationalize the
other two products. The incorporation of label from
MeO.
both H2^^0 and ^^02 , but not H2^^02, into the ring-
oxidized products can best be rationalized by the
MeO OMe formation of a radical cation species that combines
with water and/or molecular oxygen to give the
Figure 6.25. The oxidation of a highly oxidizable
aromatic probe substituted with muhiple electron-
observed products. However, with cytochrome
donating substituents yields a detectable radical cation P450, ^^O-label was incorporated only from ^^62
product. and not H2^^0. The absence of label from water in
the products from the P450 system, in view of its
incorporation with HRP, is inconsistent with diffu-
sion of a radical cation out of the enzyme active
peroxidative crosslinking of tyrosine residues, site. Thus, if a radical cation is formed, it occurs as
require the coupling of two phenoxy radicals pre- a highly transient intermediate that is immediately
sumably generated by proton-coupled electron trapped by the ferryl oxygen to give the observed
transfer from each phenol to the ferryl species. products. The results are reminiscent of the report
Concurrent formation of the two phenoxy radicals by Ohe, Mashino, and Hirobe that (a) a hydrox-
within the confines of the P450 active site should ymethyl group is eliminated as formaldehyde when
suffice to generate the observed crosslinked the ferryl oxygen adds in an ipso-msumQr to the
products. substituted carbon in a 4-substituted phenol, lead-
The clearest available evidence that aromatic ing to formation of the 1,4-quinone, and (b) when
rings can be oxidized to radical cations by the substituent is a methyl, the reaction results in
cytochrome P450, at least when the ring is substi- addition of the hydroxyl group to the substituted
tuted with multiple electron-donating groups, is ring carbon with concomitant oxidation of the phe-
provided by the reported oxidation of 1,2,4,5- nol group to a keto frmction (Figure 6.20)^^"^' ^^^. As
tetramethoxybenzene to a radical cation by reported by Rizk and Hanzlik, a methoxy group
CYP1A2 (Figure 6.25)^^^. The radical cation was also makes possible this kind of reaction^^^. Thus,
detected by absorption spectroscopy and spin-trap- mechanisms based on ip^o-addition of the ferryl
ping EPR. The stable metabolites formed in the oxygen to the aromatic ring are likely to account for
reaction were 2,5-dimethoxy-l,4-benzoquinone the products formed from 9-methylanthracene. A
and 4,5-dimethoxy-l,2-benzoquinone, the products scheme based on that proposed by Anzenbacher et
expected from hydrolysis of the radical cation. al. (Figure 6.26)^^^, or a variant of it, readily
A negligible deuterium isotope effect was observed explains the observed results without requiring a
on formation of the radical cation or products radical cation intermediate. The results do not,
derived from it even though a large isotope effect however, preclude the existence of nondiffusible
was seen for simple 0-dealkylation. The evidence radical cations as transient intermediates.
for other aromatic radical cations is less conclusive. Cavalieri and coworkers, following earlier
In an interesting set of experiments, the products investigators^^^, have championed the hypothesis
formed in the incubation of 9-methylanthracene that the covalent binding of polycyclic aromatic
and related compounds with the following systems hydrocarbons to DNA is due to radical cations
were determined: (a) CYP2B1 in the presence formed from them by the action of cytochrome
of either NADPH-cytochrome P450 reductase P450 and/or peroxidase enzymes^^^' ^^^. They have
or PhIO, (b) HRP in the presence of H2O2 or reported that polycyclic aromatic hydrocarbons
EtOOH, and (c) a model system consisting of iron with ionization potentials below 7.35 eV can be
tetraphenylporphine and PhlO^^^. Apart from oxidized to radical cations by peroxidases^^^' ^^^
the uninformative 1,2- and 3,4-diols that are formed Furthermore, the formation of a benzo[a]pyrene-
exclusively with the P450 system, the relevant DNA adduct consistent with oxidation of the
208 Paul R. Ortiz de Montellano and James J. De Voss

CH. H+ CH3

OH O
Figure 6.26. Reactions proposed to explain the oxidation of 9-methylanthracene by cytochrome P450.'^^

hydrocarbon to a radical cation by rat liver micro- to one carbon of the aromatic ring, producing a
somes and rat skin^^^' ^^^, and the presence of tetrahedral intermediate that decays by extrusion
cytochrome P450 in the nuclear membrane ^^"^j sup- of a substituent at that carbon or by electron trans-
port the proposal that cytochrome P450 enzymes fer, resulting in two-electron oxidation of the ring
may also oxidize polycyclic aromatic hydrocarbons system and/or addition of a second nucleophile
to radical cations^^^. The oxidation of 6-fluo- from the solution. This type of reaction is particu-
robenzo[a]pyrene by liver microsomes to 6-hydrox- larly favored with aromatic rings such as phenols
ybenzo[a]pyrene has been interpreted as evidence and anilines that bear electron-donating sub-
for c)^ochrome P450-catalyzed radical cation for- stituents. The evidence for the formation of radi-
mation, although the reaction could arise, as postu- cal cations by direct electron abstraction from
lated for polyhalogenated benzenes *^^, by direct polycyclic aromatic hydrocarbons remains ambigu-
addition of the activated oxygen to the aromatic ous, although the formation of such intermedi-
system ^^^. Addition of the ferry1 oxygen in such a ates is favored by multiple electron-donating
mechanism would be expected to occur at the substituents.
6-position as that is the position most sensitive to
electrophilic attack. The evidence for the formation
of a diffusible radical cation in the normal (as 7. Dehydrogenation Reactions
opposed to peroxide-dependent) cytochrome P450-
catalyzed oxidation of polycyclic aromatic hydro- Cytochrome P450 enzymes catalyze dehydro-
carbons remains inconclusive. genation as well as oxygenation reactions, includ-
In sum, considerable evidence is now available ing the oxidation of saturated to unsaturated
for the oxidation of aromatic rings not only via the hydrocarbons, alcohols to carbonyl compounds,
conventional epoxidation pathway, but also by and amines to imines or other unsaturated prod-
mechanisms that do not involve formation of an ucts. The most extensively investigated of these
epoxide as an intermediate. The non-epoxide reactions in terms of mechanism is the desatura-
mechanisms involve addition of the ferryl oxygen tion of valproic acid to 2-«-propyl-4-pentenoic
Substrate Oxidation by Cytochrome P450 Enzymes 209

been obtained when the oxidation is mediated


CO2H by either CYP2B1 or CYP4Bl202. These results
indicate that removal of a hydrogen from C-4
is rate limiting for both 4-hydroxylation and
desaturation, whereas loss of a hydrogen from
C-5 is not. These results agree well with a mecha-
nism in which removal of a C-4 hydrogen is
followed by either oxygen rebound to give the
4-hydroxy product or transfer of a hydrogen from
the terminal methyl to the ferryl oxygen to give
CO2H
the olefin product. The hydrogen could be trans-
ferred to the ferryl oxygen together with an elec-
tron or could be transferred as a proton following
transfer of the electron to give a cationic interme-
diate (Figure 6.27). Analogous mechanisms can
be postulated for the finding that CYP3A1 also
oxidizes valproic acid to the A^'^-unsaturated iso-
Figure 6.27. Two mechanistic alternatives for the
mer, except that in this case deuterium isotope
dehydrogenation of valproic acid catalyzed by P450
enzymes. experiments suggest that the olefin is obtained
equally well by initial oxidation of C-3 (k^/kj^ =
acid (Figure 621)^^^^^^. Formation of the A^'^- 2.00) or C-4 (V^j3 = 2.36)201. Interestingly, the
unsaturated product from valproic acid is cat- observation of an isotope effect at only one of
alyzed by rat, rabbit, mouse, monkey, and human the two carbons in an aerobic desaturation process
liver microsomes and by purified CYP2B1, is also found for the nonheme iron-dependent
CYP2C9, CYP2A6, CYP3A1, and CYP4B1, but fatty acid desaturases, for which a related mecha-
not by CYP3A4 or CYP4Ali96, 197, 200-203 j ^ ^ nism involving a nonheme iron center has been
A^'"^ isomer is also formed, in some instances in postulated^o^.
greater amounts than the A"^'^ isomer^^^ The 3- and The ratio of 4-hydroxy to A'^'^-desaturated
4-hydroxyvalproic acids are also formed, but these metabolites depends on the P450 isoform and is
hydroxylated products are not precursors of the larger for CYP2B1 (37:1) than for CYP4B1
unsaturated compounds ^^^. Oxidation of the two ^2 1)202 Yj^g proportion of the olefin is much
enantiomers of stereospecifically [3-^^C]-labeled higher when the substrate is the A^'^-unsaturated
valproic acid by cultured hepatocytes shows that valproic acid, a result that is particularly consis-
the /7ro-(i?)-side chain is preferentially desatu- tent with a mechanism in which the electron is
rated^^^. The A^'^-unsaturated analogue of valproic transferred to the ferryl oxygen before the hydro-
acid, 2-«-propyl-2(£r)-pentenoic acid, is also gen^^^. The structural determinants that control
desaturated to give the A^'^, A'^'^-diene, and an whether hydroxylation or desaturation occurs are
asymmetric but related molecule, 2-ethylhexanoic unknown, but if the Shaik formalism applies (see
acid, is oxidized to both 2-ethyl-l,6-hexanedioic Chapter 2), it is probable that the desaturation
acid and 2-ethyl-5-hexenoic acid^^^' '^^^. reaction involves the quartet rather than doublet
The intramolecular isotope effects for the oxi- hydroxylation transition state.
dation of valproic acid with two deuterium atoms The isopropyl group of ezlopitant, which bears
on the C-4 carbon of one of the two propyl side a 2-methoxy-5-isopropylbenzylamino group, is
chains by rabbit liver microsomes reveal that oxidized by both CYP3A4 and CYP2D6 to the
4-hydroxylation {k^k^ = 5.05) and A'^'^-desatura- tertiary alcohol and the desaturated 1-methylvinyl
tion (k^/kj^ = 5.58) are sensitive to isotopic moiety^o^. The alcohol was specifically shown not
substitution^ ^^. In contrast, when the methyl group to be a precursor of the unsaturated product, and a
of one of the side chains is trideuterated, only small primary isotope effect was observed when
minor intramolecular isotope effects are observed deuterium was placed at the benzylic but not
for 4-hydroxylation (k^/k^ = 1 09) or A'^'^-desatu- methyl carbons of the isopropyl group. Although
ration (k^/kj^ = 1.62). Comparable results have not studied in detail, concurrent hydroxylation and
210 Paul R. Ortiz de Montellano and James J. De Voss

desaturation of an isopropyl group was also the deuterium is at C-7^^^. The formation of
observed in the metabolism of a- and p-thujone 17p-hydroxy-4,6-androstadiene-3-one in this reac-
even though the isopropyl group is not bound to tion presumably occurs via the mechanism dis-
an aromatic or conjugating function^^^. These cussed above, although the finding that oxidation of
observations are well accommodated by the mech- C-6, and not C-7, leads to desaturation again sug-
anistic alternatives proposed for desaturation of gests that electron transfer from the free-radical
valproic acid. intermediate to the iron to give the allylically stabi-
The desaturation of sterols has also been lized cation may contribute to the emergence of the
observed. Quantitatively, the most important of desaturation pathway.
these is the P450-mediated A^^-desaturation in the The past decade has shown that hydrocarbon
ergosterol biosynthetic pathway of Saccharomyces desaturation is not uncommon but, except in cases
cerevisiae^^^. The enzyme (CYP61) has been puri- such as the biosynthesis of ergosterol, it generally
fied and shown to specifically catalyze the accounts for a minor proportion of the metabolic
A^^-desaturation without forming hydroxylated products. The earliest reported example of P450-
sterol products^^^' ^^^. Related A^^-desaturases are mediated hydrocarbon desaturation appears to be
found in other organisms, including mammals^^ ^' ^^^. the conversion of lindane (1,2,3,4,5,6-hexachloro-
Sterol desaturation also occurs at other positions. cyclohexane) to 1,2,3,4,5,6-hexachlorocyclohex-
Thus, the CYP2A1-catalyzed oxidation of testos- ene^^^, but the known hydrocarbon desaturation
terone yields the 7-hydroxylated, 6-hydroxylated, reactions now include the A^'^-desaturation of
and A^'^-desaturated sterols in a 38:1:1 ratio androstenedione and deoxycorticosterone by adre-
(Figure 6.28)^'^' '^^^. As might be expected, a pri- nal mitochondria^'^, the oxidation of dihydronaph-
mary intermolecular isotope effect is only observed thalene to naphthalene and 7,8-dihydrobenzo[a]
for 6-hydroxylation and A^''^-desaturation when the pyrene to benzo[a]pyrene^'^' ^'^, the conversion of
deuterium is at the allylic C-6 position, although an warfarin to dehydrowarfarin^^^, the desaturation of
isotope effect is observed for 7-hydroxylation when lovostatin and simvastatin to the 6-ejco-methylene

OH

Figure 6.28. Parallel hydroxylation and dehydrogenation of testosterone.


Substrate Oxidation by Cytochrome P450 Enzymes 211

derivatives^^ ^"^^^, and the formation of 11-dode- aromatization of 4-alkyl- and 4-aryl-l,4-dihydropy-
cenoic acid from lauric acid^^'*. ridines^^^' ^^^, the oxidation of 3-methylindole to
The direct desaturation of carbon adjacent to the highly reactive 3-methyleneindolenine^^^, and
heteroatoms, notably oxygen and nitrogen, has possibly the conversion of the A^-ethyl to an A^-vinyl
also been observed. These reactions include moiety in the metabolism of tracazolate^^"^.
the desaturation of a tetrahydrofiiran ring in the
biosynthesis of aflatoxin and sterigmatocystin by
a specific P450 enzyme^^^, the P450-catalyzed 8. Carbon-Carbon Bond
desaturation of flavanones to flavones^^^, and the Cleavage Reactions
conversion of ethylcarbamate to vinyl carba-
mate^^^. In some instances, the desaturation may
The power of P450 enzymes to catalyze oxida-
involve the carbon and the heteroatom instead of
tive transformations is perhaps nowhere better
two carbon atoms. The CYP2B1-catalyzed oxida-
illustrated than in their ability to catalyze the
tion of testosterone to androstenedione, which
cleavage of unactivated C-C bonds. Somewhat
involves oxidation of the 17-hydroxy to a 17-keto
ironically, these reactions generally form part of
function, is possibly such a reaction, because only
biosynthetic pathways and allow organisms to
5-8% of the keto group oxygen derives from O2
build complex molecules via striking metabolic
with testosterone, but 84% with epitesto-
transformations. However, C-C bond cleavages
sterone^^^. A similar observation has been made
have also been reported for some degradative/
for the P450-catalyzed oxidation of 6-hydroxy- to
xenobiotic metabolizing enzymes. Additionally,
6-keto-progesterone by CYP2C1322^. Thus, either
many of these C-C bond cleavage reactions require
one of the two hydroxyls in a conventional gem-
a sequence of oxidations that are all carried out
diol intermediate is eliminated with high stereose-
by the one enzyme. These P450 enzymes thus
lectivity, or HAT is followed by loss of an electron
form a mechanistically fascinating group as they
without actual formation of the gem-dioh
are not only capable of standard oxygen activation
and hydroxylation chemistry, but also react
RR'CHOH + [FeO]+3 -^ RR'C • (OH) through different mechanisms to eventually cleave
+ [FeOH]+3 -^ R R ' C = 0 + [Fe]+3 + H2O a C-C bond. The examples below are arranged by
the frinctional group(s) that is (are) adjacent to the
The dehydrogenation of a carbon next to a C-C bond cleaved, although this group(s) may be
nitrogen has been unambiguously demonstrated. introduced by the P450 during the course of oxi-
Acetaminophen (4-hydroxyacetanilide) is oxidized dation of the original substrate. Excluded from
to the iminoquinone intermediate by a mechanism this discussion are reactive compounds specifi-
explicitly shown not to involve hydroxylation of the cally designed to undergo cleavage of C-C bonds
nitrogen (Figure 6.29)^^^. Other examples are the as mechanistic probes, for example, cyclopropyl-
methyl containing compounds (Section 3) and the
cleavage of C-C bonds as part of the oxidation of
an aromatic ring (Section 6).

8.1. Cleavage between


Oxygenated Carbons
Diols. One of the best known examples of a
OH P450-catalyzed C-C bond cleavage is carried out by
P450^^^ (CYPllA) which converts cholesterol to
Figure 6.29. Dehydrogenation of acetaminophen to
pregnenolone and 4-methylpentanal (Figure 6.30).
the iminoquinone occurs by a mechanism that does not
include the hydroxyamide structure as an actual The mechanism by which this P450 effects scission
intermediate, even though the hydroxylamide product is of the C20-C22 bond of cholesterol has been exten-
found with related compounds that do not have the para- sively studied. The enzyme is trifunctional, catalyz-
hydroxyl group. ing three sequential reactions that each consumes
212 Paul R. Ortiz de Montellano and James J. De Voss

HO H

HO'
Pregnenolone

Figure 6.30. The intermediates in the P450g^,^ catalyzed conversion of cholesterol to pregnenolone and
4-methylpentanal.

one molecule of dioxygen and one molecule of decomposes to release one carbonyl fragment and
NADPH. The first two regio- and stereospecific a carbon radical. This radical is then intercepted
hydroxylation reactions lead to, respectively, 22(R)- by the Fe(IV)OH species to yield the second prod-
hydroxy and 20(R), 22(R) dihydroxycholesterol. uct (Figure 6.31, path B). An alternative mecha-
These reactions are unremarkable P450-catalyzed nism suggests that one of the hydroxyls of the diol
oxidations, proceeding with retention of configura- intermediate intercepts an activated oxygen
tion as expected^^^' ^^^. The third oxidative transfor- species to produce a peroxy complex. Loss of a
mation leads to cleavage of the C-C bond between proton from the adjacent alcohol initiates a het-
the two oxygenated carbons and is of considerable erolytic fragmentation reaction that leads directly
mechanistic interest. The overall transformation to the two carbonyl products (Figure 6.31, path A).
is quite efficient as the intermediate hydroxylated Formation of a hydroperoxide may be seen as
cholesterol derivatives are bound up to 300 times precedented by the exchange of oxygen between
more tightly than the parent substrate^^^ and the hydrogen peroxide and water via a putative ferryl
ferrous-dioxygen complex is more stable in each species in model systems^"^^. The chemistry of
successive tumover^^^' ^^^. such a hydroperoxide would also explain nicely
Mechanisms for cleavage of the intermediate the intriguing early observation that (20»S)-20-
diol that involve further oxidation at C-22 are (/7-tolyl)-5-pregnen-3p-ol is cleaved to preg-
excluded by the fact that the 22(S) hydrogen is nenolone and presumably phenol by P450g^^
retained in the 4-methylpentanal produced as a (Figure 6.32)^"^'. This remarkable transformation
result of C-C bond cleavage^^^. The most likely would be analogous to the well-known formation of
mechanism is thus one in which one of the acetone and phenol from cumene hydroperoxide
hydroxyl moieties is activated in some fashion, under acid catalysis.
followed by decomposition with C-C bond cleav- Recently another biosynthetic enzyme, P450g.^j
age. The nature of this activation has led to a num- (CYP107H1) has been shown to cleave an aliphatic
ber of mechanistic proposals. First, a hydrogen chain via a diol intermediate. First found as a gene
may be abstracted from one of the alcohols by the of unknown function in the biotin biosynthetic
ferryl species to form an alkoxy radical, which operon of Bacillus subtilis, P450QJQJ was implicated
Substrate Oxidation by Cytochrome P450 Enzymes 213

[Fe]

A + A

HO OH [Fe—OH]3+
[Fe=0]3 -H2O
H3C^R'

HO O;)
OH o
H3CA A H
R^CH3V\
Figure 6.31. Possible mechanisms for the final step in the cholesterol side-chain cleavage reaction, where R •
the sterol nucleus and R' = CH2CH2CH(CH3)2.

Figure 6.32. A possible mechanism for the P450g^ catalyzed conversion of (205)-20-(/7-tolyl)-5-pregnen-3(3-ol
to pregnenolone and phenol (R = sterol nucleus).

through analysis of mutants in the formation of a indicated that a range of hydroxy fatty acids was
biological equivalent of pimelic acid (heptane- produced but no co-oxidation was observed, indi-
dioic acid)^"^^. Cloning and overexpression in E. cating that production of a long-chain diacid as a
coli resulted in isolation of a complex between the pimeloate precursor was not the biological func-
P450 and acylated acyl carrier protein (ACP) as tion of this P450^'^^. Subsequently, a series of
well as the P450 alone^^^. It was shown that the potential intermediates in the C-C bond cleavage
acyl moiety of the complex could be cleaved to reaction were synthesized and incubated with the
produce a pimeloyl ACP utilizing a novel flavo- Qnzymo^^^- It was shown that pimelic acid produc-
doxin as the redox partner. P450g-Qj was also tion increased when the substrate was changed
shown to act on free fatty acids to produce pimelic from the Cj4 fatty acid to the 7-hydroxy derivative
acid as well as a range of hydroxylated fatty with the threo'l,'^-6io\ as the best substrate
acids^"^^' ^^. Careful analysis of these latter products (Figure 6.33). Other derivatives such as the 7- or
214 Paul R. Ortiz de Montellano and James J. De Voss

8-0X0, 8-hydroxy or erythro-1,% diol gave no One example of P450 mediated C-C cleavage
increase in pimelic acid formation. It was also via a presumed diol intermediate during xenobiotic
found that, as with P450g^^,, the postulated oxy- metabolism has been reported. Olanexidine, an
genated intermediates bound much more tightly to antimicrobial agent, is metabolized mainly to a
the enzyme than the parent substrate^"^^. These range of chain shortened carboxylic acids in both
results are clearly in keeping with a C-C bond rats and dogs, as well as to various other oxy-
cleavage mechanism in which the P450 operates genated metabolites (Figure 6.34)^^^' ^^^. Dog liver
on one face of the extended conformation of the microsome studies indicated that vicinal diol
fatty acid chain to produce the threo-diol, which is metabolites were further transformed to the C-C
then cleaved to produce two aldehyde fragments. bond cleavage products and specific inhibitor stud-
The pimeloyl semialdehyde initially formed is ies implicated enzymes of the CYP2D family in all
somewhat unstable to aerial oxidation and both it of the oxidative transformations^'*^. Interestingly,
and pimelic acid are seen in the cleavage of the in contrast to P450^^^ and P450QJ^J, no diastereose-
threo'7,S'dio\. Interestingly, only a small enantio- lectivity was observed in the further oxidation of
selectivity was seen for the 7-(«S) alcohol and the the diols investigated (Figure 6.34)^"*^. This may be
derived threo diol. This perhaps reflects the fact due to the position of the diol near the terminus
that the true substrate is an ACP-bound acyl of the aliphatic chain such that there is little differ-
group, making P450QJ^J one of a growing number ence in the energy of binding or oxidation of the
of P450s found to act on carrier protein bound conformations accessible to the erythro and threo
substrates^"^^. isomers. An alternative explanation is that C-C

.(CH2)5COOH (CH2)5COOH
H3C(H2C)5" H3C(H2C)5
HO H

HO H
HO. .(CH2)5COOH (CH2)5COOH (CH2)5COOH
H3C(H2C)5
O HO H
Pimelic Acid

Figure 6.33. The intermediates in the C-C bond cleavage reaction catalyzed by P450gj^, that produces pimelic
acid from tetradecanoic acid.

NH NH NH NH
(CH2)7CH3 CI J<^ JK^ ^(CH2)nCOOH

CI n = 3, 4, 5
Olanexidine

NH NH Y
CI .(CH2)4
"N'
H
X
CI X = H, Y = OH, Z = H
X = H, Y = H, Z = OH
X = OH, Y = H, Z = H
X = H, Y = OH, Z =OH (both diastereomers)
X = H, Y = OH, Z = O or Y = O, Z = OH

Figure 6.34. Oxygenated metabolites produced by P450-mediated oxidation of olanexidine.


Substrate Oxidation by Cytochrome P450 Enzymes 215

jy-
X
CH3COOH

CH3COOH

Pregnenolone A^'^ X = H, pOH


Progesterone A"*^' X = O
n
/vjJL..OH
_^ 1^ \ + CH3COOH
V
OCOCH3
_ - ^ .
17-0-Acetyltestosterone

Figure 6.35. Various oxidation products reported to be formedfrompregnenolone/progesterone via the action of
CYP17A.

bond cleavage does not proceed directly from the (Figure 6.35)-^^^. The proposed mechanism for
diol but rather an a-hydroxy ketone which is also an the dominant reaction involves an unremarkable
observed metabolite (cf. CYP17A below). However, hydroxylation at CI7 of the steroid nucleus
the different effects of specific P450 inhibitors on (Figure 6.36)^^^. This is then followed by an attack
a-hydroxy ketone formation and C-C bond cleav- of the ferric peroxo moiety on the carbonyl to yield
age argue against a precursor product relation- a species that fragments to an alkoxy radical and a
ship^"^^. Final delineation of the pathway awaits one-electron oxidized ferryl species. The alkoxy
studies with purified isoforms but does suggest that radical subsequently decomposes to produce acetic
C-C bond cleavage may be a significant metabolic acid and a carbon radical that recombines with the
pathway for compounds with aliphatic chains. ferryl species to yield a gem-dio\, which dehydrates
Keto Alcohols. CYP17A is a remarkable, mul- to the C17 carbonyl of the product. This mechanism
tifunctional P450 that is primarily responsible is in accord with a wealth of labeling studies and can
for the 17a-hydroxylation of pregnenolone (or prog- be modified simply to explain the origin of the other
esterone) and the subsequent lysis of the C17-C20 observed products.
bond to produce dehydroepiandrosterone (or andro- Studies with ^^62 have demonstrated the incor-
stendione)^^^' ^^^ In addition, it catalyzes the poration of one atom of ^^O into the acetic acid
cleavage of this same C17-C20 bond in mechanisti- fragment produced upon C17-C20 cleavage of
cally distinct ways to yield a number of minor prod- pregnenolone in all of the observed pathways^^^'
ucts. These less common pathways lead to the 256,257 Additionally, ^^O incorporation from ^^03
formation of 17a-hydroxyandrost-5-en-3p-ol (note is seen at the C17 position of the steroidal prod-
inversion of stereochemistry at CI7) and the corre- ucts from pregnenolone bearing an oxygen atom
sponding A ^^'^ ^-olefin from pregnenolone^^^"^^^ as at this position^^^. The minor products from C17-
well as 17-0-acetyltestosterone from progesterone C20 cleavage are proposed to arise from attack
216 Paul R. Ortiz de Montellano and James J. De Voss

•O—Fe(IlI)

CH3COOH

Figure 6.36. Proposed mechanism for the C17-C20 lyase reaction catalyzed by CYP17A. The key steps involve
addition of a P450 ferric peroxide species to the C20 carbonyl and subsequent free radical fragmentation of the
peroxyhemiacetal.

,..xOH

iO' •O—Fe(III)
(J 'O—Fe(III) /
+ CH3COOH

':b
V
17-(9-Acety Itestosterone

Figure 6.37. Proposed mechanistic manifold to account for the formation of other products in the CYP17A
catalyzed oxidation of pregnenolone/progesterone. Ionic decomposition (pathway b) of the peroxyhemiacetal
competes with free-radical fragmentation (pathway a) to yield the observed mixture of minor products.

of the ferric peroxo species on the carbonyl prior This latter product may arise via direct hydrogen
to any C17 hydroxylation (Figure 6.37). The abstraction or via a single-electron oxidation of the
resultant adduct then fragments to an alkoxy radi- radical to the cation followed by elimination of a
cal which loses acetic acid to produce a C17 radi- proton^^^. Results of CYP17A catalyzed oxidation
cal in a fashion analogous to that proposed for the of substrates bearing deuterium labels at the CI6,
major pathway. This C17 radical then partitions C17 and the methyl group a to the ketone are in
between direct oxygen rebound to the 17a- agreement with this mechanism^^^. The 17-0-
hydroxy product and elimination to the A ^^'^ ^-olefin. acetyl-testosterone is probably best explained as
Substrate Oxidation by Cytochrome P450 Enzymes 217

the result of a Baeyer-Villiger like decomposition bond a to an aldehyde is discussed further in


of the peroxo adduct derived initially from an Section 8.3.
attack on the carbonyl (Figure 6.37)^^^. This indi-
cates that the peroxo adduct may decompose by
an ionic mechanism as well as by the radical 8.2. Cleavage Alpha to
pathway proposed to explain the other observed Oxygenated Carbon
products.
The role of the ferric peroxo moiety in the Ketones. The CYP17A-mediated cleavage of
mechanism has been supported by mutagenesis the C17-C20 bond of pregnenolone (or proges-
studies in which Thr306 has been replaced by an terone) without prior CI7 hydroxylation provides
alanine^^^. This threonine is believed to be the the only clearly documented example of cleavage of
active site residue that directs the delivery of pro- a C-C bond a to a ketone (Figure 6.37, Section 8.1).
tons required to cleave the O-O bond and form The manifold of products formed, however, nicely
the ferryl species. As expected, its loss results in indicates the variety of mechanistic pathways that
an approximately 20-fold decrease in the ferryl might be envisioned. A peroxo adduct fi*om the
dependent C17 hydroxylation activity but a much carbonyl and the ferric peroxide intermediate
smaller decrease in C17-C20 lyase activity medi- forms and subsequently decomposes by one of two
ated by the ferric peroxo moiety^^^. Experiments pathways. A radical mechanism leads to an alkoxy
involving analysis of the solvent deuterium iso- radical that eventually gives C-C bond cleavage to
tope effect as a function of pH have suggested that form an alcohol or olefinic product. An ionic
the protonation of the ferric peroxide intermediate mechanism (Baeyer-Villiger) leads to an ester
governs whether the reaction proceeds via a ferryl product in which insertion of oxygen has occurred
dependent (17a hydroxylation) or a peroxy adduct with retention of configuration. It will be of inter-
(C17-C20 lyase) pathway259. est to determine whether such pathways might pro-
The aldehyde corresponding to pregnenolone vide the dominant activity of some P450s.
has also been used as a mechanistic probe with Aldehydes. Cleavage of a C-C bond a to an
CYP17A as it is reported to undergo exclusive aldehyde has already been discussed in the context
cleavage of the C17-C20 bond to produce formic of the CYP17A-catalyzed oxidation of an alde-
acid and the 17a alcohol or A^^'^^-olefin^^^. These hyde analogue of pregnenolone (Section 8.1).
reactions are believed to proceed via pathways However, such reactions are believed to play
analogous to those proposed for the formation of a central role in the activities of several other P450s
minor cleavage products of CYP17A catalyzed including the important steroid biosynthetic
oxidation of pregnenolone. The more electrophilic enzymes aromatase (CYP19) and 14a-demethy-
carbonyl of the aldehyde favors the direct bond lase (CYP51). It is worth emphasizing that P450-
scission pathways by more effectively trapping catalyzed aldehyde oxidation does not necessarily
the ferric peroxide moiety. The aldehyde is not result in C-C bond cleavage and that in fact often
reported to be oxidized to the corresponding oxidation to the corresponding carboxylic acid
acid^^^. This suggests that an ionic cleavage of the occurs^^^ The factors that govern partitioning
proposed peroxy intermediate (Baeyer-Villiger between these different modes of oxidation are
pathway) does not occur to any great extent as unknown at present^^^.
hydrogen migration, which would lead to acid for- Aromatase (CYP19), like P450g^^, plays an
mation, is known to be favored for this type of essential role in the biosynthesis of steroid hor-
reaction. The experiments with this aldehyde do, mones. It catalyzes the aromatization of the C^^
however, provide evidence for the bifurcation of a androgen, androstendione to the C^g estrogen
single pathway leading to the two minor products estrone (Figure 6.38), as well as similar aromati-
of pregnenolone oxidation (Figure 6.37). Deute- zations of testosterone and 16a-hyroxyandrosten-
ration of the CI6a position led to an apparent dione^^^' ^^^. These conversions involve three
enrichment in deuteration of the 17a-hydroxy sequential oxidations at the angular C19 methyl
product, suggesting an isotope-induced partition- group that result in its eventual loss and aromati-
ing away from the A ^^'^ ^-olefin that requires zation of the A-ring of the substrate. Each oxida-
cleavage of the C-D bond^^o. Cleavage of the C-C tion requires a molecule of NADPH and of
218 Paul R. Ortiz de Montellano and James J. De Voss

HO, HO OH
Ha Hp

o A ^ ,V o ^ ^ ^ ,
Androstendione

O^ H
V

/
Estrogen

Figure 6.38. Intermediates in the catalytic turnover of aromatase (CYP19).

oxygen^^'*. The first two steps appear to be unex- Schiff base formed from the 3-keto moiety^^"^.
ceptional P450-catalyzed hydroxylation steps. The Several of these intermediates are known to be
initial reaction produces the CI9 primary alcohol converted spontaneously^^ ^ or by aromatase^^^ into
and proceeds, as expected, with retention of estrone but none of them are currently accepted as
configuration^^^' ^^^, while the second oxidation lying upon the major pathway for aromatization.
abstracts the \9-pro-R hydrogen to yield a gem- This is primarily due to the '^O labeling studies
diol intermediate^^^' ^^^. This latter compound is indicating that the third oxygen atom is incorpo-
believed to dehydrate to yield the more stable, rated into formate^^^' ^^^. The difficulties in estab-
observed C19 aldehyde. There is an observable lishing the mechanism are illustrated nicely with
tritium isotope effect on the first hydroxylation the postulated 2p-hydroxy intermediate. This was
step^^^, but not on the subsequent one with [19-^H] synthesized and shown to aromatize rapidly in the
androst-4-ene-3,17-dione or analogues^^^' ^^^ absence of enzyme^^' and it was also detected in
This is understandable as the first step can dis- enzymic incubations at low pH (which slows the
criminate between the hydrogen and tritium atoms aromatization reaction)^^^. However, the facts that
on a given methyl group. An isotope effect on the the 2p-hydroxyl was not incorporated into the
second step, however, which stereospecifically released formate^^^, and the stereochemistry of
removes the pro-R hydrogen, would require the loss of hydrogen from C-2 appears to be substrate
kind of /«/er-molecular effect commonly sup- dependent ruled this compound out as an obliga-
pressed in P450 reactions. It is the mechanism of tory intermediate^^^' ^^^. The currently accepted
the third oxidative transformation that involves mechanism^^^' 288-290 explains all experimental
C-C bond cleavage and aromatization that has observations and is supported by model stud-
attracted the most attention. In this reaction, the ip jgg291-293 ^j^^ analogy with the mechanisms of
and 2(3 hydrogens are lost^^^~^^^ into water and the other P450s such as CYP17A and CYP2B4 {vide
CI9 carbon as formate which contains an oxygen infra) (Figure 6.39). Thus, the ferric peroxide inter-
atom from the first and third oxidation steps^^^' ^^^. mediate is believed to add to the electrophilic alde-
A large number of different mechanisms have been hyde carbonyl to yield a peroxyhemiacetal. This
proposed to account for this transformation involv- can fragment to give an alkoxy radical that loses
ing the intermediacy of a steroid containing a C19 formic acid to produce a CIO radical. Loss of the
formyl group and, variously, a 4,5-epoxide^^^, ip hydrogen and enolization of the carbonyl is
a ip-hydroxyl276, a 2p-hydroxyl ^su 282^ ^^ ^ ^ 9 required to produce the aromatized A ring. Recent
peroxide ^^^' ^^^ as well as a possible enzymic model studies by Valentine and coworkers provide
Substrate Oxidation by Cytochrome P450 Enzymes 219

H , O-O-Fe(III) HO O-O-Fe(III)
V^ c O-Fe(III)

+ HCOOH

Figure 6.39. The currently accepted mechanism for the final step in the aromatase catalyzed reaction. The timing
of enolization of the carbonyl with respect to the addition of the ferric peroxide to the aldehyde and to C-C and 0 - 0
bond fission is still uncertain.

« ^

+ Fe(III)(TMP)02" + Fe(III)(TMP)OH + HCOOH

F3CSO3' F3CSO3

TMP = Tetramesitylporphyrin

Figure 6.40. Conversion of enolized analogue of the natural aromatase substrate to the corresponding aromatized
compound is catalyzed by a model peroxo ferric porphyrin complex.

strong support for the involvement of the ferric per- group from a variety of steroidal nuclei with con-
oxo in the mechanism^^^. They demonstrated that a comitant introduction of a carbon-carbon double
model peroxo ferric porphyrin complex will quan- bond (Figure 6.41). The archetypal reaction is the
titatively convert an enolized model of androsten- loss of the C14 angular methyl group (C32) from
dione to the corresponding aromatic compound and lanosterol with formation of a C14-C15 double
formate (Figure 6.40). Reaction of the ferric peroxo bond during cholesterol biosynthesis^^^' ^^^. Once
model with androstendione itself results in the again, this conversion is believed to involve three
chemically reasonable epoxidation of the electron sequential oxidation steps and proceed initially via
deficient C4-C5 double bond. This follows sugges- an alcohol that is subsequently converted into an
tions in the literature that enolization of the C3 car- aldehyde^^^"^^^. These steps parallel the first two
bonyl occurs prior to C19-C10 bond cleavage and steps catalyzed by aromatase and are believed to be
additionally activates the i p hydrogen toward unexceptional hydroxylation reactions^^^"^^^. The
loss^^^' ^^^. However, whether the chemoselectivity stereochemical course of the second hydroxylation is
required (C-C bond cleavage vs epoxidation) is unknown, but studies with mechanism-based inhi-
achieved enzymatically via prior enolization or by bitors have shown that steroidal 32-5-vinylalcohols
selective positioning of the substrate within the are transformed to covalent inhibitors, presumably
active site remains to be established. with a C32 carbonyl via a C32 gem-diol while the
Enzymes of the CYP51 family (sterol 14a-deme- 32-jR isomers are not oxidized^ ^^ These results do
thylases) catalyze the removal of the 14a-methyl demonstrate stereospecificity in the oxidation of
220 Paul R. Ortiz de Montellano and James J. De Voss

Figure 6.41. The stable intermediates in the 14a-demethylation of lanosterol.

+ HCOOH

Figure 6.42. A possible mechanism for the final step in the 14a-demethylation of lanosterol that employs the
isolated Baeyer-Villiger rearrangement product. Radical decomposition of the peroxyhemiacetal intermediate may
also lead to the observed demethylated product.

a C32 alcohol but the deduction that theproS hydro- hydrogen in the formation of the A'"^' ^^ double
gen is removed is more questionable. The 32-»S'-vinyl bond^^^' 313,314 gy analogy with the aromatase
alcohols that are oxidized have a hydrogen that is reaction, the oxidation is proposed to be initiated
stereochemically equivalent to the proR hydrogen of by addition of the ferric peroxo intermediate to the
the hydroxymethyl intermediate in the demethyla- electrophilic aldehyde to produce a peroxyhemi-
tion reaction. This would suggest that it is this proR acetal (Figure 6.42). However, the decomposition
hydrogen that is abstracted in the second hydroxyla- of this intermediate is proposed to occur via an
tion step catalyzed by CYP51. ionic Baeyer-Villiger-like mechanism rather than
The facts on the oxidation of the C32 aldehyde a radical one. The primary reason for this is the
are remarkably similar to those seen for the aro- isolation and spectroscopic identification of the
matase catalyzed reaction. The formate moiety 14a-formyloxy compound required as an interme-
that is expelled contains one dioxygen-derived diate in the ionic mechanism^^^. It was also
oxygen atom and incorporates one oxygen and one demonstrated that the 14a-formyloxy intermediate
hydrogen atom from the aldehyde precursor^^^. could be converted into the final product by the
There is also a stereospecific loss of the syn 15a enzyme^ ^^. The mechanism of this elimination has
Substrate Oxidation by Cytochrome P450 Enzymes 221

not been studied in detail but loss of the tertiary of catalyzing the conversion of some aldehydes
allylic formate would be expected to be a facile into the one-carbon diminished alkene and for-
reaction, subject to standard acid-base catalysis. mate^^^' ^^^. It appears that there is a structural
However, the proposed mechanism invokes an requirement for a or p branching of the aldehyde
ionic decomposition of a peroxyhemiacetal while for the reaction, with alkene formation occurring
those suggested for CYP17 and CYP19 invoke with compounds such as isobutyraldehyde and
radical pathways. The possibility remains there- 2-methylbutyraldehyde but not with the straight-
fore that, as is believed to be the case with CYP17, chain propionaldehyde or valeraldehyde^^^.
the Baeyer-Villiger product is formed as a result Although most work has been carried out with
of a minor pathway while the major route pro- CYP2B4, other isoforms such as CYP1A2, 2E1,
ceeds via simultaneous elimination of C32 and the 2C3, and 3A6 are all reported to catalyze this type
15a hydrogen. A unified view of the mechanism of transformation^^^. The extent to which this
of the three P450 families would suggest that this occurs relative to oxidation of the aldehyde to the
occurs via radical decomposition of the peroxy- corresponding carboxylic acid appears small with
hemiacetal (cf. Figure 6.39). It is perhaps also a ratio of 50:1 favoring acid formation in the
possible that the isolated formyloxy compound CYP2B4 catalyzed oxidation of 2-phenylpropi-
arises from formate trapping of a C14 cation, the onaldehyde^^^. However, the reaction has proved
major fate of which would be to undergo elimina- very valuable as a model for understanding the
tion with loss of the CI5a hydrogen. The cation mechanism of the C-C bond cleavage reactions of
would derive from a single-electron oxidation of a CYP17,19, and 51 and has played a significant role
C14 radical, the intermediate in the radical decom- in the formulation of their mechanisms above. It
position of the peroxyhemiacetal. The availability was shown that the deformylation reaction was
of cloned and overexpressed CYP51 from ani- supported by P450 reductase/NADPH or H2O2 but
mals^^^' ^^\ plants^i^frmgi^^^,and bacteria^^^-^^i not cumene hydroperoxide or iodosyl benzene^^"*.
with differing substrate specificity^^^ and also of a This concurs with the hypothesis that it is the ferric
crystal structure of one of the bacterial enzymes ^^ peroxo species that adds to the aldehyde rather than
should facilitate complete elucidation of the the ferry1 species. Fragmentation of the hydroper-
mechanism of this interesting family of P450s. oxyhemiacetal then occurs to produce formate and
Some xenobiotic metabolizing enzymes, par- the alkene (Figure 6.43). Significantly, formation of
ticularly CYP2B4, are also reported to be capable the one-carbon reduced alcohol has also been

O.^ / H , O-O-Fe(III) a*0-Fe(III)


O-O-Fe(III)

+ HCOOH

[Fe=0]3

[Fe-OH]3 .OH

Fe(III)

Figure 6.43. The oxidation of cyclohexanecarboxaldehyde by CYP2B4 is believed to proceed via the ferryl
species to yield the carboxylic acid and via the ferric peroxo species to yield the deformylated products, cyclohexene
and cyclohexanol.
222 Paul R. Ortiz de Montellano and James J. De Voss

reported, although only m passing !^^^ This product the peroxyhemiacetal intermediate. An intermedi-
is analogous to the 17a-hydroxy C^^ products ate was detected in this work that was spectro-
reported from CYP17A oxidation of pregnenolone scopically consistent with an isoporphyrin which
and its analogues. It should be noted that while would be formed upon addition of a carbon radi-
deformylation is thought to involve the ferric per- cal to the heme cofactor^^^. Finally, it is of note
0X0 species, oxidation to the acid is believed to pro- that the ferryl catalyzed oxidation of aldehydes to
ceed via the ferryl species^^^. acids can also cause enzyme inactivation by heme
The relevance of the CYP2B4 catalyzed adduct formation, but in this case as predicted for
deformylation reaction as a model for CYP51 is an H abstraction mechanism, an acylated heme is
clearly demonstrated by the aromatization of the formed^^^.
androstendione analogue 3-oxodecalin-4-ene-10- Cytochrome P450s can also interact with alde-
carboxaldehyde to the corresponding tetrahydron- hydes in a different way to generate the corre-
aphthalene (cf Figure 6.40) with concomitant sponding hydrocarbon and C02,^^^. Hydrocarbons
formate production^^^' ^^^. Deuterium isotope are abundant components of cuticular lipids in
studies showed that the formyl hydrogen was most insects and can also play a role in their
retained in the formate, that the 1 p hydrogen was chemical communication. It has been demon-
specifically lost, and that loss of the C2 hydrogen strated that in microsomes derived from the house
was not stereoselective. These results faithfully fly, Musca domestica, hydrocarbons are formed
reproduce the characteristics of the aromatase cat- from the corresponding aldehyde with concomi-
alyzed reaction. tant generation of CO2 and with all the character-
Recently, support for the role of ferric peroxo istics expected of a P450-mediated reaction:
species in CYP2B4 catalyzed deformylation, and
by analogy for the mechanisms of CYP17, -19,
CH3(CH2)8CH=CH(CH2)i2CD2CDO + NADPH
and -51, has come from mutagenesis studies^^^. Vaz
and Coon reported the effect of replacing Thr302, + O2 + H^ • CH3(CH2)8CH=CH(CH2)i2CD3
^2 + H2C
the residue thought to facilitate O-O bond cleavage
in CYP2B4, with alanine. It was expected that this
would favor the peroxo pathway and decrease the There is a requirement for NADPH and oxygen
availability of the ferryl species. In line with these and the reaction is inhibited by both CO and an
expectations, normal P450-catalyzed reactions, antibody to house fly P450 reductase^^^. Labeling
including aldehyde to carboxylic acid oxidation, studies showed that deuterium atoms at the C-1,
were suppressed but deformylation to the alkene C-2, and C-3 positions were all retained^^^ In
and alcohol products was significantly enhanced^^^. addition, active oxygen donors such as hydrogen
Evidence for the radical nature of the decomposition peroxide, cumene hydroperoxide, and iodosylben-
of the peroxyhemiacetal has come from examination zene all support hydrocarbon production to some
of the mechanism-based inactivation of P450s that extent. The ability of the latter species to support
occurs concurrently with aldehyde oxidation^^^' ^^^. oxidation clearly indicates that the ferric peroxide
For saturated aldehydes, it was shown that inactiva- species is not the active oxidant in this case. On
tion of CYP2B4 paralleled their ability to undergo a the basis of these results, an unusual mechanism
deformylation reaction, suggesting that both of has been proposed^^' and a slightly more conven-
these processes flowed from a common intermedi- tional version is presented here (Figure 6.44). The
ate, the peroxyhemiacetal (Figure 6.43)^^^. It was first step is the oxidation of the aldehyde to a
shown that inactivation of the P450 was due to addi- dioxirane or its resonance form, a carbonyl oxide.
tion of the carbon radical, formed in a homolytic Dioxiranes are known to decompose with release
process, to the -y-meso position of the prosthetic of CO2 and formation of two radicals that can
heme^^^. Interestingly, although P450Q]^3 is reported recombine as shown to form a hydrocarbon^^^.
to oxidize a variety of aldehydes without detectable Presumably, this recombination would be favored
deformylation^^ \ it was demonstrated that a mutant by retention of the fragments within the active
is deactivated by aldehydes when the co-oxidant is site. Complete elucidation of the reaction mecha-
^2^2^^^' This presumably again occurs through an nism awaits identification and purification of the
alkyl radical formed by homolytic decomposition of P450 but recent studies have shown this to be a
Substrate Oxidation by Cytochrome P450 Enzymes 223

^Q/

A ^ •e=0]^^
"X'^—W ^ R-^A RH + CO2

Figure 6.44. Possible mechanism for the P450-catalyzed conversion of an aldehyde into the corresponding
hydrocarbon and CO2 seen in the biosynthesis of insect-derived hydrocarbons.

[Fe=0]3-t

HO.

R = H (+)-marmesin
R = OH prandiol

B
[Fe=0]3+ [Fe-OH]3+
P^('II> HP t
Ionic
a OH
b(^0.
O
-H2O

Radical
b
Fe(III)

o*

A,
Figure 6.45. Mechanistic possibilities for the P450-cataIyzed conversion of (+)-marmesin into psoralen and
acetone. Prandiol is known not to be an intermediate in this process.

widespread reaction in insects for the formation of (Figure 6.45). The stereochemistry of the elim-
hydrocarbons^^^. ination was exclusively syn and a small isotope
Alcohols. Boland and coworkers demon- effect {k^lkj^ = 4) was observed when the abstracted
strated conclusively that a P450 can catalyze the hydrogen was replaced with deuterium. The mech-
direct fragmentation of an alcohol into an olefin anism proposed (Figure 6.45, pathway A) consists
and a carbonyl-containing fragment^^. They studied of P-hydrogen atom abstraction, decomposition
the conversion of marmesin to psoralen in micro- of the radical intermediate to produce the olefin
somes derived from cell cultures of the plsnatAmmi and an isopropoxyl radical, the latter of which is
majus (Figure 6.45)^^. Deuterium labeled precur- intercepted by the Fe(IV)OH species^^. A possible
sors allowed them to demonstrate that marmesin mechanistic alternative (Figure 6.45, pathway B)
was converted into an equimolar mixture of acetone invokes intermediates analogous to those proposed
and psoralen, excluding the possibility of other oxy- for the diol cleavage reactions above. In these, it
genated intermediates such as the known prandiol would be the alcohol moiety that is initially
224 Paul R. Ortiz de Montellano and James J. De Voss

attacked and initiates fragmentation to the observed Aflatoxins are mycotoxins produced by strains
products. in the fungal genus Aspergillus and are notable for
This type of C-C cleavage reaction, however, the complexity of their biogenesis. Genetic evi-
appears to be a general and important biosynthetic dence suggested that a single P450 was responsible
one in plants and a number of other analogous for the transformation of 0-methylsterigmatocystin
oxidative C-C bond cleavage reactions seen in to aflatoxin Bj (Figure 6.47)^^1 A P450 from
bacteria and plants have now been postulated to be Aspergillus parasiticus was subsequently cloned,
P450 catalyzed^^"^' ^^^. In particular, secologanin heterologously expressed in yeast, and was demon-
synthase from Catharanthus roseus (CYP72A1) is strated to be capable of catalyzing this remarkable
believed to catalyze the C-C bond cleavage that conversion in vivo^^^. The first formed 11-hydroxy-
transforms loganin into secologanin, the final 0-methylsterigmatocystin (Figure 6.47) was also
common non-nitrogenous precursor of many plant synthesized and shown to be converted to aflatoxin
indole alkaloids (Figure 6.46)^^^' ^^^. In this case, Bp These experiments interlocked with a wealth of
the reaction involves cleavage of a carbocyclic previous results from in vivo isotope labeling stud-
ring rather than fragmentation of the substrate. ies and led to the mechanistic hypothesis shown^^^.
This activity was demonstrated in vitro with After C-C bond cleavage and formation of the pro-
CYP72A1 heterologously expressed in E. coli as a posed hydrolytically unstable lactone, the ensuing
fusion with its homologous P450 reductase^^^. decarboxylation, dehydration, rearrangement, and
A reaction that involves cleavage of the C-C bond 0-demethylation reactions are presumed to proceed
a to a phenol occurs in aflatoxin biosynthesis^^^. spontaneously. Two plausible mechanisms for the

HO

COCH3
Secologanin

Figure 6.46. Loganin is converted into secologanin via a P450-catalyzed C-C bond cleavage reaction analogous
to that seen in psoralen biosynthesis.

CH3O

CH3O CH3O
Aflatoxin B
R = H O-Methylsterigmatocystin
R = OH 11-hydroxy O-Methylsterigmatocystin

Figure 6.47. A single cytochrome P450 is responsible for the conversion of O-methylsterigmatocystin to aflatoxin
Bj via 11-hydroxy O-methylsterigmatocystin.
Substrate Oxidation by Cytochrome P450 Enzymes 225

> / Fe(III)
..^^^^^^^^
^ CH3O
CH3O

[Fe=0]^+
OH
'

/ 0
[^ V X
CHsO^ r 0

H2O—Lactone Hydrolysis;
Spontaneous -CO2, -CH3OH, -H2O
and rearrangement
Aflatoxin B1

Figure 6.48. Mechanistic proposals for the P450-catalyzed C-C bond cleavage during the biosynthesis of
aflatoxin Bj. One possibility involves a Baeyer-Villiger-like reaction of the ferric peroxo species with the keto
tautomer of the phenolic substrate while the other proceeds via the epoxide intermediate typical of ferryl catalyzed
aromatic oxidations.

COOH
COOH
^«r-kaurenoic acid
R = CHO GAi2-aldehyde
R = COOH GA12

Figure 6.49. CYP88A catalyzes the three oxidative transformations required to convert ew^kaurenoic acid into GAj2.

C-C bond cleavage process have been proposed Several similarly remarkable multifunctional
(Figure 6.48)^^^. The first involves a Baeyer- P450s have been implicated in their biosynthesis in
Villiger-like oxidation of the keto tautomer of the both plants and fungi339, 34o CYP88A from
phenol and the second a rearrangement of the epox- Arabidopsis thaliana and barley has been shown
ide intermediate in aromatic oxidation. Delineation to catalyze the three oxidative steps required to
of the mechanism will require experimentation convert ^«^kaurenoic acid to GAj2 (Figure 6.49)^'*^
with purified enzyme, mutants, and substrate The experiments involved the expression of
analogues. CYP88A in yeast strains containing A. thaliana
The gibberellins (GAs) are important plant hor- P450 reductase and monitoring in vivo oxidation of
mones with remarkably complex structures. potential substrates. The key step in the proposed
226 Paul R. Ortiz de Montellano and James J. De Voss

rr""—
OH
I

4 OR
-H+

\ ^ CHO
Figure 6.50. Mechanism of a pinacol rearrangement of a diol.

[Fe=0]^-

-CO,

Figure 6.51. Likely mechanisms for the P450-catalyzed conversion of valerate into isobutene and CO2. A pathway
involving direct hydride abstraction has also been proposed but appears less probable.

reaction involves cleavage of a C-C bond a to an characterization of the catalytic capabilities of this
alcohol in an oxidative ring contraction to yield an enzyme will be of great interest.
aldehyde (GAj2-aldehyde, Figure 6.49)^'^'. The Acids. Two isolated examples of P450-
mechanism has not been investigated but the catalyzed oxidative decarboxylation have appeared
process follows the pathway predicted for an a- in the literature. The first concerns the formation of
hydroxy carbocation, such as the intermediate isobutene from isovalerate by the yeast Rhodotorula
proposed for a pinacol rearrangement of a diol minuta^^^. A P450 and a homologous reductase
(Figure 6.50). Such a cation could arise from a were purified and a reconstituted system that pro-
diol formed by CYP88A catalyzed C6 hydroxyla- duced isobutene from isovalerate was con-
tion under the influence of the Lewis acidic heme structed^"^^. A large isotope effect upon isobutene
iron or directly via a SET process from the hydrox- formation was found when the p-hydrogen was sub-
ylation radical intermediate. Subsequently, a P450 stituted with deuterium {k^lk^ = 14), clearly impli-
from the fungus Gibberella fujikuroi was also cating cleavage of this bond in the rate-determining
shown to catalyze the same pinacol-like transfor- step. It was also found that p-branching appeared to
mation, once again by expression and in vivo mon- be necessary for alkene formation. A mechanism
itoring of putative substrate transformation^^^. In involving direct hydride abstraction and decarboxy-
this case, a 6,7-diol was also isolated but was not lation of the resultant cation was proposed^"^^.
further transformed via ring contraction, suggest- However, more conventional pathways are also
ing that such a compound is not an intermediate in possible in which either (a) hydrogen atom abstrac-
this pathway. This single fungal P450 was also pro- tion to give a carbon radical is followed by electron
posed to be capable of catalyzing at least seven transfer to generate the corresponding carbocation,
other biosynthetically significant oxidative trans- or (b) the tertiary alcohol is formed but ionizes to
formations as well as the three assigned to the carbocation under the influence of the Lewis
CYP88A, explaining the various GA metabolites acidic heme iron (Figure 6.51). One caveat with this
found in G. fujikuroi. One of these other transfor- system is that the P450 was subsequently shown to
mations is a proposed oxidative cleavage of the hydroxylate benzoate to 4-hydroxybenzoate as part
vicinal 6,7-diol. Clearly, the results of in vitro of phenylalanine catabolism and this latter reaction
Substrate Oxidation by Cytochrome P450 Enzymes 227

appears to be its primary metabolicfimction^'^'^'^'^^. shorter alcohoP"^^. The acid must have an a-
It is also unclear whether other nonvolatile products carbon bearing either a phenyl group or three
of isovalerate oxidation are formed in the incuba- substituents. This transformation was originally
tions which were monitored by headspace gas observed in iron-porphyrin model systems but
chromatography^"^^. Thus, the exact nature of this was subsequently reproduced in vivo in rats and in
decarboxylation reaction remains to be established. rat liver microsomes for two therapeutic car-
Hirobe has reported the P450-catalyzed decar- boxylic acids (Figure 6.52)^'*^. Once again, the
boxylation of carboxylic acids to a one-carbon mechanism has not been investigated but the

[ F e. ^=n0i]3 +
R—COOH •O—Fe(III) _^ R* 'O—Fe(III) + CO2
o
[Fe=0]^+

ROH
R. ^
N|^ ^O—Fe(III)
O

C6H5CO Ketoprofen Indomethacin ^ /7-CIC6H4


O

Figure 6.52. Possible pathways for the oxidative decarboxylation of some therapeutic carboxylic acids catalyzed
by both P450s and some iron-porphyrin model systems.

Isoflavone Synthase

O. ,X Ij HO.

[Fe—OH]3

Figure 6.53. Isoflavone synthase (CYP93C) catalyzes the formation of isoflavone from 25-flavone via an
oxidative aryl migration. A possible mechanism involving an anchimerically stabilized radical is shown.
228 Paul R. Ortiz de Montellano and James J. De Voss

proposed decomposition of a carboxyl radical is containing a tetramethylpiperidine moiety as they


attractive, especially as this process is known to be have also observed similar metabolism in other
quite sensitive to a-substitution. The radical might mammals^^^. The mechanism of the reaction has
be produced either directly from the carboxylate not been investigated in detail but clearly appears
by the ferryl species or by decomposition of a to be a transformation of a secondary amine,
peroxyacid initially formed by reaction of the acid formed via A^-dealkylation if necessary, given the
and an ironoxo species (Figure 6.52). This latter structures of the pyrrolidines produced. The inter-
mechanism would then be analogous to the mediacy of hydroxylamines or the corresponding
reported CYP2B4 catalyzed conversion of 2- nitroxyl radical in this reaction has been sug-
phenylperacetic acid to CO2 and benzyl alcohol gested. One possibility (Figure 6.54) is that the
by homolysis of the O-O bond^"^^. heme iron may promote ionization of a hydroxyl-
Ethers. Isoflavone synthase (CYP93C) cat- amine to an incipient nitrogen cation that
alyzes the formation of isoflavone from 25'- rearranges, a pathway comparable to the P450-
flavone via an unusual oxidative aryl migration catalyzed dehydration of oximes to nitriles^^^.
(Figure 6.53>f^^-^^^. (This C-C bond cleavage Alternatively, it may simply be a rearrangement
occurs a to an ether and is classified as such here, of the intermediate nitrogen cation radical formed
but it is unclear whether this is a mechanistically during amine oxidation. This can no longer be
significant feature.) Little is known about the stabilized by a-hydrogen elimination and the
reaction, but it is postulated to proceed via 3(3 steric congestion of the surrounding methyl
HAT to give a carbon radical anchimerically sta- groups may slow the oxygen rebound, allowing
bilized by the adjacent phenoP^^' ^^'. Oxygen rearrangement (Figure 6.54). The piperidine to
rebound can then occur at the C2 position to give pyrrolidine rearrangement has precedent in the
the unstable 2-hydroxyisoflavone that dehydrates chemistry of 7V-fluoroamines that undergo the
to isoflavone (Figure 6.53). Support for this mech- same ring contraction in the presence of a Lewis
anism is provided by the isolation of the 3(3- acid^^"^. This latter reaction, however, presumably
hydroxyisoflavone as a side product of the involves the equivalent of a nitrogen cation,
reaction^^^ The availability of heterologously rather than a cation radical species favoring the
overexpressed wild-type protein and site-directed former pathway. More detailed investigations are
mutants should facilitate investigation of this required to determine the mechanism of this
unusual transformation^^'. interesting transformation.
Finally, another of the remarkable multifunc-
tional P450s involved in GA formation in fungi
8.3. Cleavage Alpha to Carbon has recently been demonstrated to catalyze
Bearing a Nitrogen Atom the demethylation of an angular carbon along
the biosynthetic pathway (Figure 6.55)^^^. The
Amines. Recently, an example of C-C bond apparently concomitant formation of the lactone
cleavage a to an amine has been reported with demethylation suggests that a different
(Figure 6.54)^^^. Interestingly, this is also a pathway is followed from that seen in aromatase
rearrangement reaction and one of the few exam- and 14a-demethylase. It is tempting to speculate
ples of C-C bond scission catalyzed by non- that this represents a biosynthetically novel
biosynthetic enzymes. It was found that a variety oxidative decarbonylation or decarboxylation
of tetramethylpiperidine containing compounds reaction in which an alcohol or the correspond-
were transformed into the corresponding ing cation is the initial product. This could then
dimethylpyrrolidine derivatives (Figure 6.54). By be intercepted by the adjacent carboxylate to
incubations with recombinant human liver P450s form the observed lactone (Figure 6.55). Clearly,
and immuno-inhibition studies, this reaction was however, the cytochrome P450s are capable of
shown to be catalyzed by a variety of P450s, with catalyzing C-C cleavage by a variety of mecha-
CYP3A4 the major isoform responsible for this nisms and much work remains to understand all
transformation. The authors suggest that this is a the possible permutations of these interesting
general metabolic pathway for compounds reactions.
Substrate Oxidation by Cytochrome P450 Enzymes 229

Fe(II)

Figure 6.54. A variety of tetramethylpiperidine compounds are converted into the corresponding
dimethylpyrrolidine derivatives by a number of xenobiotic metabolizing P450s, particularly CYP3A4. Two mechanistic
possibilities for this process are shown. (R = H, R' = /7-nitrophenyl-NH- or R = CH3, R' = (CgH3)2HCO-)

COOH

Figure 6.55. Gibberellin 20-oxidase from the fungus Gibberellafujikuroi is a multifunctional P450 that catalyzes
the angular demethylation of GAj2 to produce the lactone GA^.

9. Conclusions frequently encountered. Experimentally, the past


few years have seen major progress in characteriz-
Cytochrome P450 mechanisms continue to ing the intermediates that are formed as molecular
surprise and delight, although the field is growing oxygen is activated to the final oxidizing species.
to maturity and the completely unexpected is less All the intermediates, with the exception of the
230 Paul R. Ortiz de Montellano and James J. De Voss

critical ferryl species, have now been directly 2. Shimizu, T., K. Hirano, M. Takahashi, M. Hatano,
observed by various spectroscopic and crystallo- and Y. Fujii-Kuriyama (1988). Site-directed muta-
graphic methods. The ferric peroxo anion has genesis of cytochrome P-450d: Axial ligand and
been found to act as the oxidizing agent with a heme incorporation. Biochemistry 27, 4138-4141.
3. Unger, B. (1988). Ph.D. Thesis. University of
growing range of highly electrophilic substrates.
Illinois Urbana, Champaign, IL.
In contrast, the proposed role for the ferric
4. Auclair, K., P. Moenne-Loccoz, and PR. Ortiz de
hydroperoxo complex as an electrophilic oxidiz- Montellano (2001). Roles of the proximal heme thi-
ing agent remains a matter of debate, as the evi- olate ligand in cytochrome P450cam. J. Am. Chem.
dence advanced in support of the proposal is Soc. 123, 4877-^885.
circumstantial and contradictory. Although the 5. Yoshioka, S., S. Takahashi, H. Hori, K. Ishimori,
ferryl species remains elusive, it is increasingly and I. Morishima (2001). Proximal cysteine residue
clear that it plays the predominant role as the oxi- is essential for the enzymatic activities of cyto-
dizing agent in the P450 catalytic cycle. A second chrome P450^,^. Eur. J. Biochem. 268, 252-259.
cam '
area that has recently received considerable atten- 6. Tani, E, M. Matsu-ura, S. Nakayama, M. Ichimura,
tion is the mechanism of hydrocarbon hydroxyla- N. Nakamura, and Y Naruta (2001). Synthesis and
tion, the key question being whether the radical characterization of alkanethiolate-coordinated iron
rebound mechanism that has held sway for three porphyrins and their dioxygen adducts as models
decades is in fact valid. The contradictory results for the active center of cytochrome P450: Direct
obtained with radical and cation probes, which evidence for hydrogen bonding to bound dioxygen.
have provided most of the new evidence, must be J. Am. Chem. Soc. 123, 1133-1142.
resolved by fiirther experimentation in order for 7. Woggon, W.-D., H.-A. Wagennecht, and C. Claude
this question to be settled. The development of a (2001). Synthetic active site analogues of heme-
thiolate proteins. Characterization and identifica-
two-state model for the catalytic action of P450
tion of intermediates of the catalytic cycles of
enzymes may be one of the most important recent
cytochrome P450cam and chloroperoxidase. J.
advances in the field, as it provides a ready expla- Inorg. Biochem. 83, 289-300.
nation for a variety of otherwise contradictory 8. Ogliaro, R, S. Cohen, M. Filatov, N. Harris, and
data, some of which argues for concerted and S. Shaik (2000). The high-valent compound of
some for nonconcerted oxidation mechanisms. No cytochrome PA50: The nature of the Fe-S bond and
doubt, the next few years will uncover novel the role of the thiolate ligand as an internal electron
aspects of P450 function and will lead to deeper donor. Angew. Chem. Int. Ed. 39, 3851-3855.
and more sophisticated understanding of the cat- 9. Green, M.T (1999). Evidence for sulfur-based rad-
alytic mechanisms of the amazing family of P450 icals in thiolate compound I intermediates. J. Am.
Chem. Soc. 121, 7939-7940.
enzymes.
10. Poulos, TL., B.C. Finzel, and A.J. Howard (1987).
High-resolution crystal structure of cytochrome
P450cam. J. Mol. Biol. 192, 687-700.
11. Ravichandran, K.G., S.S. Boddupalli, CA.
Acknowledgments Hasemann, J.A. Peterson, and J. Deisenhofer
(1993). Crystal structure of hemoprotein domain of
The work from the authors' laboratories and P450BM-3, a prototype for microsomal P450's.
the preparation of this review were supported by Science 261, 731-736.
National Institutes of Health Grant GM25515 12. Li, H. and T.L. Poulos (1997). The structure of the
(PROM) and Australian Research Council Grant cytochrome p450BM-3 haem domain complexed
DP0210635 (JJDV). with the fatty acid substrate, palmitoleic acid. Nat.
Struct. Biol. 4, 140-146.
13. Hasemann, C.A., K.G. Ravichandran, J.A.
Peterson, and J. Deisenhofer (1994). Crystal struc-
References ture and refinement of cytochrome P450terp at
2.3 A resolution. J. Mol. Biol. 236, 1169-1185.
14. Cupp-Vickery, J. and TL. Poulos (1995). Structure
1. Dawson, J.H. and M. Sono (1987). Cytochrome
of cytochrome P450eryF involved in erythromycin
P-450 and chloroperoxidase: Thiolate-ligated heme
biosynthesis. Nat. Struct. Biol. 2, 144-153.
enzymes. Spectroscopic determination of their
15. Shimizu, H., S.-Y Park, Y Gomi, H. Arakawa,
active-site structures and mechanistic implications
H. Nakamura, S.-I. Adachi et al. (2000). Proton
of thiolate ligation. Chem. Rev. 87, 1255-1276.
delivery in NO reduction by fungal nitric-oxide
Substrate Oxidation by Cytochrome P450 Enzymes 231

reductase. Cryogenic crystallography, spectroscopy, cytochrome P450: Comparison of hydrogen perox-


and kinetics of ferric-NO complexes of wild-type and ide and NADPH-catalyzed N-demethylation VQ2iC-
mutant enzymes. J. Biol. Chem. 275, 4816-4826. tions. Xenobiotica 14, 87-104.
16. Yano, J.K., L.S. Koo, D.J. Schuller, H. Li, P.R. Ortiz 27. Renneberg, R., J. Capdevila, N. Chacos, R.W.
de Montellano, and T.L. Poulos (2000). Crystal Estabrook, and R.A. Prough (1981). Hydrogen
structure of a thermophilic cytochrome P450 from peroxide-supported oxidation of benzo[a]pyrene
the archaeon Sulfolobus solfataricus. J. Biol. Chem. by rat liver microsomal fractions. Biochem.
275,31086-31092. Pharmacol. 30, 843-848.
17. Podust, L.M., Y. Kim, M. Arase, B.A. Neely, 28. Fasco, M.J., L.J. Piper, and L.S. Kaminsky (1979).
B.J. Beck, H. Bach et al. (2003). The 1.92-A struc- Cumene hydroperoxide-supported microsomal
ture of Streptomyces coelicolor A3(2) CYPl 54C1. A hydroxylations of warfarin—^A probe of cytochrome
new monooxygenase that functionalizes macrolide P450 multiplicity and specificity. Biochem. Pharma-
ring systems, J. Biol. Chem. 278, 12214-12221. col. 28, 97-103.
18. Podust, L.M., T.L. Poulos, and M.R. Waterman 29. Kelly, WG. and A.H. Stolee (1978). Stabilization of
(2001). Crystal structure of cytochrome P450 placental aromatase by dithiothreitol in the pres-
14alpha-sterol demethylase (CYP51) from ence of oxidizing agents. Steroids 31, 533-539.
Mycobacterium tuberculosis in complex with azole 30. Kupfer, R., S.Y Liu, A.J. Allentoff, and J.A.
inhibitors. Proc. Natl. Acad. Sci. USA 98, 3068-3073. Thompson (2001). Comparisons of hydroperoxide
19. Nagano, S., H. Li, H. Shimizu, C. Nishida, H. Ogura, isomerase and monooxygenase activities of
PR. Ortiz de Montellano et al. (2003). Crystal struc- cytochrome P450 for conversions of allylic
ture of epothilone-D bound, epothilone-B bound, hydroperoxides and alcohols to epoxyalcohols and
and susbstrate-free forms of cytochrome P450epoK. diols: Probing substrate reorientation in the active
J. Biol. Chem. 278, 44886-44893. site. Biochemistry 40, 11490-11501.
20. Williams, PA., J. Cosme, V Sridhar, E.F. Johnson, 31. He, K., L.M. Bornheim, A.M. Falick, D. Maltby,
and D.E. McRee (2000). Mammalian microsomal H. Yin, and M.A. Correia (1998). Identification
cytochrome P450 monooxygenase: Structural of the heme-modified peptides from cumene
adaptations for membrane binding and fimctional hydroperoxide-inactivated cytochrome P450 3A4.
diversity. Mol. Cell. 5, 121-131. Biochemistry 31, 17448-17457.
21. Guengerich, F.P and WW. Johnson (1997). 32. Mueller, E.J., PJ. Loida, and S.G. Sligar (1995).
Kinetics of ferric cytochrome P450 reduction by Twenty-five years of P450cam research. In P.R.
NADPH-cytochrome P450 reductase: Rapid reduc- Ortiz de Montellano (ed.). Cytochrome P450:
tion in the absence of substrate and variations Structure, Mechanism, and Biochemistry, 2nd edn.
among cytochrome P450 systems. Biochemistry 36, Plenum, New York, pp. 83-124.
14741-14750. 33. Loida, PJ. and S.G. SUgar (1993). Engineering
22. Imai, M., H. Shimada, Y. Watanabe, Y. cytochrome P-450^,^j^ to increase the stereospeci-
Matsushima-Hibiya, R. Makino, H. Koga et al. ficity and coupling of aliphatic hydroxylation.
(1989). Uncoupling of the cytochrome P-450cam Protein Eng. 6, 207-212.
monooxygenase reaction by a single mutation, 34. Atkins, WM. and S.G. Sligar (1987). MetaboHc
threonine-252 to alanine or valine: Possible role of switching in cyctochrome ^-^^0^^^: Deuterium iso-
the hydroxy amino acid in oxygen activation. Proc. tope effects on regiospecificity and the monooxy-
Natl. Acad. Sci. USA 86, 7823-7827. genase/oxidase ratio. J. Am. Chem. Soc. 109,
23. Martinis, S.A., WM. Atkins, PS. Stayton, and 3754-3760.
S.G. SHgar (1989). A conserved residue of cyto- 35. Fruetel, J.A., J.R. Collins, D.L. Camper, G.H.
chrome P-450 is involved in heme-oxygen stability Loew, and PR. Ortiz de Montellano (1992).
and activation, o^^m. Chem. Soc. I l l , 9252-9253. Calculated and experimental absolute stereochem-
24. Kimata, Y, H. Shimada, T. Hirose, and Y Ishimura istry of the styrene and beta-methylstyrene epox-
(1995). Role of Thr-252 in cytochrome P450cam: A ides formed by cytochrome P 450cam. J. Am.
study with unnatural amino acid mutagenesis. Cherri. Soc. 114, 6987-6993.
Biochem. Biophys. Res. Commun. 208, 96-102. 36. Perret, A. and D. Pompon (1998). Electron shuttle
25. Ortiz de Montellano, PR. (1995). Oxygen activa- between membrane-bound cytochrome P450 3A4
tion and reactivity. In P.R. Ortiz de Montellano and b^ rules uncoupling mechanism. Biochemistry
(ed.). Cytochrome P450: Structure, Mechanism, 37, 11412-11424.
and Biochemistry, 2nd edn. Plenum, New York, 37. Reed, J.R. and PR Hollenberg (2003). Comparison
pp. 245-304. of substrate metabolism by cytochromes P450 2B1,
26. Estabrook, R.W, C. Martin-Wixtrom, Y Saeki, 2B4, and 2B6: Relationship of heme spin state,
R. Renneberg, A. Hildebrandt, and J. Werringloer catalysis, and the effects of cytochrome b5. J. Inorg.
(1984). The peroxidatic function of liver microsomal Biochem. 93, 152-160.
232 Paul R. Ortiz de Montellano and James J. De Voss

38. Schlichting, I., J. Berendzen, K. Chu, A.M. Stock, theoretical investigation of the iron(III)-hydroper-
S.A. Maves, D.E. Benson et al (2000). The cat- 0X0 species and its epoxidation pathways. J. Am.
alytic pathway of cytochrome P450^^j^ at atomic Chem. Soc. 124, 2806-2817.
resolution. Science 1^1, 1615-1622. 51. Guengerich, F.R, A.D.N. Vaz, G.N. Raner, S.J.
39. Davydov, R., T.M. Makris, V Kofman, D.E. Werst, Pernecky, and M.J. Coon (1997). Evidence for a
S.G. SHgar, and B.M. Hoffman (2001). Hydroxy- role of a perferryl-oxygen complex, FeO^^, in the
lation of camphor by reduced oxy-cytochrome N-oxygenation of amines by cytochrome P450
P450cam: Mechanistic implications of EPR and enzymes. Mol. Pharmacol. 51, 147-151.
ENDOR studies of catalytic intermediates in native 52. Vatsis, K.P and M.J. Coon (2002). Ipso-substitution
and mutant enzymes. J. Am. Chem. Soc. 123, by cytochrome P450 with conversion of p-hydroxy-
1403-1415. benzene derivatives to hydroquinone: Evidence for
40. Denisov, I.G., T.M. Makris, and S.G. Sligar (2001). hydroperoxo-iron as the active oxygen species.
Cryotrapped reaction intermediates of cytochrome Arch. Biochem. Biophys. 397, 119-129.
P450 studied by radiolytic reduction with phospho- 53. Toy, PH., B. Dhanabalasingam, M. Newcomb,
rus-32. J. Biol. Chem. 216, 11648-11652. I.H. Hanna, and PF HoUenberg (1997). A substituted
41. Rahimtula, A.D., PJ. O'Brien, E.G. Hrycay, hypersensitive radical probe for enzyme-catalyzed
J.A. Peterson, and R.W. Estabrook (1974). Possible hydroxylations: Synthesis of racemic and enan-
higher valence states of cytochrome P-450 during tiomerically enriched forms and application in a
oxidative reactions. Biochem. Biophys. Res. cytochrome P450-catalyzed oxidation. J. Org.
Commun. 60, 695-702. Chem. 62, 9\\4-9l22.
42. Blake, R.C. and M.J. Coon (1981). On the mecha- 54. Toy, PH., M. Newcomb, and PF. HoUenberg
nism of action of cytochrome P-450. Role of (1998). Hypersensitive mechanistic probe studies
peroxy spectral intermediates in substrate hydroxy- of cytochrome P450-catalyzed hydroxylation reac-
lation. J. Biol. Chem. 256, 5755-5763. tions. Implications for the cationic pathway. J. Am.
43. Wagner, G.C., M.M. Palcic, and H.B. Dunford Chem. Soc. 120, 7719-7729.
(1983). Absorption spectra of cytochrome 55. Toy, PH., M. Newcomb, M.J. Coon, and A.D.N. Vaz
P450CAM in the reaction with peroxy acids. FEBS (1998). Two distinct electrophilic oxidants effect
Lett. 156, 244-248. hydroxylation in cytochrome P-450-catalyzed reac-
44. Egawa, T, H. Shimada, and Y. Ishimura (1994). tions. J. Am. Chem. Soc. 120, 9718-9719.
Evidence for compound I formation in the reaction of 56. Schoneboom, J.C, H. Lin, N. Renter, W Thiel,
cytochrome P450cam with m-chloroperbenzoic acid. S. Cohen, F Ogliaro et al. (2002). The elusive
Biochem. Biophys. Res. Commun. 201, 1464-1469. oxidant species of cytochrome P450 enzymes:
45. Schiinemann, V, C. Jung, A.X. Trautwein, Characterization by combined quantum mechani-
D. Mandon, and R. Weiss (2000). Intermediates in cal/molecular mechanical (QM/MM) calculations.
the reaction of substrate-free cytochrome P450cam J.Am. Chem. Soc. 124, 8142-8151.
with peroxy acetic acid. FEBS Lett. 179, 149-154. 57. Ogliaro, F, S.P de Visser, S. Cohen, PK. Sharma,
46. Schiinemann, V, C. Jung, J. Terner, A.X. Trautwein, and S. Shaik (2002). Searching for the second oxi-
and R. Weiss (2002). Spectroscopic studies of dant in the catalytic cycle of cytochrome P450: A
peroxyacetic acid reaction intermediates of theoretical investigation of the iron(III)-hydroper-
cytochrome P450^^j^ and chloroperoxidase. J. 0X0 species and its epoxidation pathways. J. Am.
Inorg. Biochem. 91, 586-596. Chem. Soc. 124,2806-2817.
47. Kellner, D.G., S.-C. Nung, K.E. Weiss, and S.G. 58. Kamachi, T, Y. Shiota, T. Ohta, and K. Yoshizawa
Sligar (2002). Kinetic characterization of com- (2003). Does the hydroperoxo species of
pound I formation in the thermostable cytochrome cytochrome P450 participate in olefin epoxidation
P450 CYPl 19. J. Biol. Chem. 271, 9641-9644. with the main oxidant, Compound 1: Criticism from
48. Vaz, A.D.N., D.F. McGinnity, and M.J. Coon density functional theory calculations. Bull. Chem.
(1998). Epoxidation of olefins by cytochrome Soc. Jpn. 16, 721-132.
P450: Evidence from site-specific mutagenesis for 59. Groves, J.T, G.A. McClusky, R.E. White, and M.J.
hydroperoxo-iron as an electrophilic oxidant. Proc. Coon (1978). Aliphatic hydroxylation by highly
Natl. Acad. Sci. USA 95, 3555-3560. purified liver microsomal cytochrome P-450.
49. Jin, S., T.M. Makris, T.A. Bryson, S.G. Sligar, and Evidence for a carbon radical intermediate.
J.H. Dawson (2003). Epoxidation of olefins by Biochem. Biophys. Res. Commun. 81, 154-160.
hydroperoxo-ferric cytochrome P450. J. Am. Chem. 60. Ogliaro, F, S.P. de Visser, S. Cohen, J. Kaneti, and
Soc. 125, 3406-3407. S. Shaik (2001). The experimentally elusive oxi-
50. Ogliaro, K, S.P de Visser, S. Cohen, RK. Sharma, dant of cytochrome P450: A theoretical "trapping"
and S. Shaik (2002). Searching for the second oxi- defining more closely the "real" species.
dant in the catalytic cycle of cytochrome P450: A ChemBiochem. 11, 848-851.
Substrate Oxidation by Cytochrome P450 Enzymes 233

61. Ogliaro, R, N. Harris, S. Cohen, M. Filatov, S.P. de 73. Houghton, J.D., S.E. Beddows, K.E. Suckling,
Visser, and S. Shaik (2000). A model "rebound" L. Brown, and C.J. Suckling (1986). 5a,6a-
mechanism of hydroxylation by cytochrome P450: Methanocholestan-3p-ol as a probe of the mecha-
Stepwise and effectively concerted pathways and nism of action of cholesterol 7a-hydroxylase.
their reactivity patterns. J. Am. Chem. Soc. 122, Tetrahedron Lett. 11, 4655-4658.
8977-8989. 74. Bowry, VW and K.U. Ingold (1991). A radical
62. Foster, A.B. (1985). Deuterium isotope effects in clock investigation of microsomal cytochrome
the metabolism of drugs and xenobiotics: P-450 hydroxylation of hydrocarbons. Rate of
Implications for drug design. Adv. Drug Res. 14, oxygen rebound. J. Am. Chem. Soc. 113,
2^0. 5699-5707.
63. Hjelmeland, L.M., L. Aronow, and J.R. Trudell 75. Newcomb, M. and PH. Toy (2000). Hypersensitive
(1977). Intramolecular determination of primary radical probes and the mechanisms of cytochrome
kinetic isotope effects in hydroxylations catalyzed P450-catalyzed hydroxylation reactions. Ace. Chem.
by cytochrome P-450. Biochem. Biophys. Res. Res. 33, 449-455.
Commun. 76, 541-549. 76. Atkinson, J.K. and K.U. Ingold (1993).
64. White, R.E., J.P. Miller, L.V Favreau, and A. Cytochrome P450 hydroxylation of hydrocarbons:
Bhattacharyaa (1986). Stereochemical dynamics of Variation in the rate of oxygen rebound using
aliphatic hydroxylation by cytochrome P-450. J. cyclopropyl radical clocks including two new ultra-
Am. Chem. Soc. 108, 6024-6031. fast probes. Biochemistry 32, 9209-9214.
65. Gelb, M.H., D.C. Heimbrook, P Malkonen, and 77. Atkinson, J.K., PF. Hollenberg, K.U. Ingold,
S.G. Sligar (1982). Stereochemistry and deuterium C.C. Johnson, M.-H. Le Tadic, M. Newcomb et al.
isotope effects in camphor hydroxylation by the (1994). Cytochrome P450-catalyzed hydroxylation
cytochrome P450 monoxygenase system. of hydrocarbons: Kinetic deuterium isotope effects
Biochemistry 21, 370-377. for the hydroxylation of an ultrafast radical clock.
66. Groves, J.T. and D.V Subramanian (1984). Biochemistry 33, 10630-10637.
Hydroxylation by cytochrome P-450 and metallo- 78. Newcomb, M., M.-H. Le Tadic, D.A. Putt, and
porphyrin models. Evidence for allylic rearrange- P.F. Hollenberg (1995). An incredibly fast apparent
ment. J: .4m. Chem. Soc. 106, 2177-2181. oxygen rebound rate constant for hydrocarbon
67. Oliw, E.H., I.D. Brodowsky, L. Hornsten, and hydroxylation by cytochrome P-450 enzymes. J.
M. Hamberg (1993). Bis-allylic hydroxylation of Am. Chem. Soc. Ill, 3312-3313.
polyunsaturated fatty acids by hepatic monooxyge- 79. Newcomb, M., M.-H. Le Tadic-Biadatti, D.L.
nases and its relation to the enzymatic and nonen- Chestney, E.S. Roberts, and PF. Hollenberg (1995).
zymatic formation of conjugated hydroxy fatty A nonsynchronous concerted mechanism for
acids. Arch. Biochem. Biophys. 300, 434^39. cytochrome P450 catalyzed hydroxylation. J. Am.
68. Tanaka, K., N. Kurihara, and M. Nakajima (1979). Chem. Soc. Ill, 12085-12091.
Oxidative metabolism of tetrachlorocyclohexenes, 80. Auclair, K., Z. Hu, D.M. Little, PR. Ortiz de
pentachlorocyclohexenes, and hexachlorocyclo- Montellano, and J.T. Groves (2002). Revisiting the
hexenes with microsomes from rat liver and mechanism of P450 enzymes using the radical
house fly abdomen. Pestic. Biochem. Physiol. 10, clocks norcarane and spiro[2,5]bicyclooctane. J.
79-95. Am. Chem Soc. 124, 6020-6027.
69. Stanjek, V, M. Miksch, P. Lueer, U. Matern, and 81. Newcomb, M., R. Shen, Y. Lu, M.J. Coon, RE
W. Boland (1999). Biosynthesis of psoralen: Hollenberg, D.A. Kopp et al. (2002). Evaluation of
Mechanism of a cytochrome P450 catalyzed oxida- norcarane as a probe for radicals in cytochrome
tive bond cleavage. Angew. Chem. Int. Ed. 38, P450- and soluble methane monooxygenase-
400-402. catalyzed hydroxylation reactions. J. Am. Chem.
70. Ortiz de Montellano, PR. and R.A. Stearns (1987). Soc. 124, 6879-6886.
Timing of the radical recombination step in 82. Hino, F and D. Dolphin (1999). The biomimetic
cytochrome P-450 catalysis with ring-strained oxidation of dieldrin using polyhalogenated metal-
probes, ^ ^ m . Chem. Soc. 109, 3415-3420. loporphyrins. J. Chem. Soc. Chem. Commun.
71. White, R.E., J.T. Groves, and G.A. McClusky 629-630.
(1979). Electronic and steric factors in regio- 83. Shaik, S., M. Filatov, D. Schroder, and H. Schwarz
selective hydroxylation catalyzed by purified (1998). Electronic structure makes a difference:
cytochrome P-450. Acta Biol. Med. Ger. 38, Cytochrome P450 mediated hydroxylations of
475-482. hydrocarbons as a two-state reactivity paradigm.
72. Sligar. S.G., M.H. Gelb, and D.C. Heimbrook Chem. Eur. 4, 193-199.
(1984). Bio-organic chemistry and cytochrome 84. Harris, N., S. Cohen, M. Filatov, and R Ogliaro
P-450-dependent catalysis. Xenobiotica 14, 63-86. (2000). Two-state reactivity in the rebound step of
234 Paul R. Ortiz de Montellano and James J. De Voss

alkane hydroxylation by cytochrome P-450: Origins catalyzed oxidation reactions. The oxidative
of free radicals with finite Hfetimes. Angew. Chem. 0-dealkylation of 7-ethoxycoumarin. J. Biol.
Int. Ed. 39, 2003-2007. Chem. 259, 3000-3004.
85. Schroder, D., S. Shaik, and H. Schwarz (2000). 96. Baciocchi, E., O. Lanzalunga, A. Lapi, and
Two-state reactivity as a new concept in L. Manduchi (1998). Kinetic deuterium isotope
organometalHc chemistry. Ace. Chem. Res. 33, effect profiles and substituent effects in the oxida-
139-145. tive N-demethylation of N,N-dimethylanilines
86. Ogliaro, R, S.P. de Visser, J.T. Groves, and S. Shaik catalyzed by tetrakis(pentafluorophenyl)porphyrin
(2001). Chameleon states: High-valent metal-oxo iron(III) chloride. J. Am. Chem. Soc. 120,
species of cytochrome P450 and its ruthenium 5783-5787.
analogue. Angew. Chem. Int. Ed. 40, 2874-2878. 97. Galliani, G., M. Nali, B. Rindone, S. Tollari,
87. Manchester, J.I., J.P. Dinnocenzo, L. Higgins, and M. Rocchetti, and M. Salmona (1986). The rate of
J.P. Jones (1997). A new mechanistic probe for N-demethylation of N,N-dimethylanilines and
cytochrome P450: An application of isotope effect N-methylanilines by rat-liver microsomes is related
profiles. J. ^m. Chem. Soc. 119, 5069-5070. to their first ionization potential, their lipophilicity
88. Choi, S.-Y., RE. Eaton, PE HoUenberg, K.E. Liiu, and to a steric bulk factor. Xenobiotica 16,
S.J. Lippard, M. Newcomb et al. (1996). 511-517.
Regiochemical variations in reactions of methyl- 98. Macdonald, T.L., W.G. Gutheim, R.B. Martin, and
cubane with ^er/-butoxyl radical, cytochrome P-450 F.P. Guengerich (1989). Oxidation of substituted
enzymes and a methane monooxygenase system. J. N,N-dimethylanilines by cytochrome P-450:
Am. Chem. Soc. 118, 6547-6555. Estimation of the effective oxidation-reduction
89. Newcomb, M., R. Shen, S.-Y. Choi, PH. Toy, potential of cytochrome P-450. Biochemistry 28,
PR HoUenberg, A.D.N. Vaz et al. (2000). 2071-2077.
Cytochrome P450-catalyzed hydroxylation of mech- 99. Goto, Y, Y Watanabe, S. Fukuzumi, J.P Jones,
anistic probes that distinguish between radicals and and J.P Dinnocenzo (1998). Mechanisms of
cations. Evidence for cationic but not for radical N-demethylations catalyzed by high-valent species
intermediates, o^ ^m. Chem. Soc. 122, 2677-2686. of heme enzymes: Novel use of isotope effects and
90. Stearns, R.A. and PR. Ortiz de Montellano (1985). direct observation of intermediates. J. Am. Chem.
Cytochrome P-450 catalyzed oxidation of quadri- Soc. 120, 10762-10763.
cyclane. Evidence for a radical cation intermediate. 100. Miwa, G.T, J.S. Walsh, G.L. Kedderis, and PR
J.Am. Chem. Soc. 107, 4081^082. HoUenberg (1983). The use of intramolecular iso-
91. Ortiz de Montellano, PR., H.S. Beilan, and tope effects to distinguish between deprotonation
IC.L. Kunze (1981). N-Alkylprotoporphyrin IX for- and hydrogen atom abstraction mechanisms in
mation in 3,5-dicarbethoxy-l,4-dihydrocollidine- cytochrome P-450- and peroxidase-catalyzed
treated rats. Transfer of the alkyl group from the N-demethylation reactions. J. Biol. Chem. 258,
substrate to the porphyrin. J. Biol. Chem. 256, 14445-14449.
6708-6713. 101. Hall, L.R. and R.P Hanzlik (1991). N-dealkyla-
92. Augusto, O., H.S. Beilan, and PR. Ortiz de tion of tertiary amides by cytochrome P-450.
Montellano (1982). The catalytic mechanism of Xenobiotica 2\, 1127-1138.
cytochrome P-450. Spin-trapping evidence for one- 102. Okazaki, O. and F.P. Guengerich (1993). Evidence
electron substrate oxidation. J. Biol. Chem. 257, for specific base catalysis in N-dealkylation reac-
11288-11295. tions catalyzed by cytochrome P450 and
93. Kennedy, C.H. and R.P Mason (1990). A reexami- chloroperoxidase. Differences in rates of deproto-
nation of the cytochrome P-450-catalyzed free rad- nation of aminium radicals as an explanation for
ical production from a dihydropyridine. Evidence high kinetic hydrogen isotope effects observed
of trace transition metal catalysis. J. Biol. Chem. with peroxidases. J. Biol. Chem. 268, 1546-1552.
265, 11425-11428. 103. Nelsen, S.F and J.T. Ippoliti (1986). The deproto-
94. Guengerich, F.P, C.-H. Yun, and T.L. Macdonald nation of trialkylamine cation radicals by amines.
(1996). Evidence for a 1-electron oxidation mecha- J. Am. Chem. Soc. 108, 4879-^881.
nism in N-dealkylation of N,N-dialkylanilines by 104. Karki, S.B., J.P Dinnocenzo, J.P Jones, and K.R.
cytochrome P450 2B1. Kinetic hydrogen isotope Korzekwa (1995). Mechanism of oxidative amine
effects, linear free energy relationships, comparisons dealkylation of substituted N,N-dimethylanilines
with horseradish peroxidase and studies with oxygen by cytochrome P-450: Application of isotope
surrogates. J. Biol. Chem. 271, 27321-27329. effect profiles, ^^m. Chem. Soc. Ill, 3657-3664.
95. Miwa, G.T, J.S. Walsh, and A.Y.H. Lu (1984). 105. Tanko, J.M., R. Friedline, N.K. Suleman, and N.
Kinetic isotope effects on cytochrome P-450- Castagnoli (2001). tert-BuioxyX as a model for
Substrate Oxidation by Cytochrome P450 Enzymes 235

radicals in biological systems: Caveat emptor. J. 118. Burstyn, J.N., M. Iskandar, J.F Brady, J.M.
Am. Chem. Soc. 123, 5808-5809. Fukuto, and A.K. Cho (1991). Comparative stud-
106. Macdonald, T.L., K. Zirvi, L.T. Burka, P. Peyman, ies of N-hydroxylation and N-demethylation by
and P.P. Guengerich (1982). Mechanism of microsomal cytochrome P-450. Chem. Res. Toxicol.
cytochrome P-450 inhibition by cyclopropy- 4, 70-76.
lamines. a ^ w . Chem. Soc. 104, 2050-2052. 119. Hlavica, P and U. Kunzel-Mulas (1993).
107. Hanzlik, R.P and R.H. Tullman (1982). Suicidal Metabolic N-oxide formation by rabbit-liver
inactivation of cytochrome P-450 by cyclopropyl- microsomal cytochrome P-4502B4: Involvement
amines. Evidence for cation-radical intermediates. of superoxide in the NADPH-dependent N-
J. Am. Chem. Soc. 104, 2048-2050. oxygenation of N,N-dimethylaniline. Biochim.
108. Guengerich, F.P, R.J. Willard, J.P Shea, Biophys. Acta 1158, 83-90.
L.E. Richards, and T.L. Macdonald (1984). 120. Watanabe, Y., T. Numata, T. lyanagi, and S. Oae
Mechanism-based inactivation of cytochrome (1981). Enzymatic oxidation of alkyl sulfides by
P-450 by heteroatom-substituted cyclopropanes cytochrome P450 and hydroxyl radical. Bull.
and formation of ring-opened products. J. Am. Chem. Soc. Jpn. 54, 1163-1170.
Chem. Soc. 106, 6446-6447. 121. Watanabe, Y, T. lyanagi, and S. Oae (1980). Kinetic
109. Shaffer, C.L., M.D. Morton, and R.P Hanzlik study on enzymatic S-oxygenation promoted by a
(2001). A^-Dealkylation of an A^-cyclopropylamine reconstituted system with purified cytochrome
by horseradish peroxidase. Fate of the cyclopropyl P450. Tetrahedron Lett. 21, 3685-3688.
group. J. Am. Chem. Soc. 123, 8502-8508. 122. Watanabe, Y, T. lyanagi, and S. Oae (1982). One
110. Shaffer, C.L., S. Harriman, Y.M. Koen, and electron transfer mechanism in the enzymatic
R.P. Hanzlik (2002). Formation of cyclopropanone oxygenation of sulfoxide to sulfone promoted by a
during cytochrome P450-catalyzed N-dealkylation reconstituted system with purified cytochrome
of a cyclopropylamine. J. Am. Chem. Soc. 124, P450. Tetrahedron Lett. 23, 533-536.
8268-8274. 123. Alvarez, J.C. and PR. Ortiz de Montellano (1992).
111. Kuttab, S., J. Shang, and N. CastagnoH (2001). Thianthrene 5-oxide as a probe of the elec-
Rat liver microsomal enzyme catalyzed oxida- trophilicity of hemoprotein oxidizing species.
tion of 4-phenyl-trans-l-(2-phenylcyclopropyl)- Biochemistry 2^1, 8315-8322.
1,2,3,6-tetrahydropyridine. Bioorg. Med. Chem. 9, 124. Bacciochi, E., O. Lanzalunga, and B. Pirozzi
1685-1689. (1997). Oxidations of benzyl and phenethyl
112. Hall, L.R. and R.P Hanzlik (1989). Kinetic deu- phenyl sulfides. Implications for the mechanism
terium isotope effects on the N-demethylation of of the microsomal and biomimetic oxidation of
tertiary amides by cytochrome P-450. J. Biol. sulfides. Tetrahedron 53, 12287-12298.
Chem. 264, 12349-12355. 125. Baciocchi, E., O. Lanzalunga, S. Malandrucco, M.
113. Constantino, L., E. Rosa, and J. Iley (1992). The lolel, and S. Steenken (1996). Oxidation of sul-
microsomal demethylation of N,N-dimethylben- fides by peroxidases. Involvement of radical
zamides. Substituent and kinetic deuterium isotope cations and the rate of the oxygen rebound step. J.
effects. Biochem. Pharmacol. 44, 651-658. Am. Chem. Soc. 118, 8973-8974.
114. Iley, J., L. Constantino, F. Norberto, and E. Rosa 126. Holland, H.L., M.J. Chernishenko, M. Conn,
(1990). Oxidation of the methyl groups of A. Munoz, T.S. Manoharan, and M.A. Zawadski
N,N-dimethylbenzamides by a cytochrome P450 (1990). Enzymic hydroxylation and sulfoxidation
mono-oxygenase model system. Tetrahedron Lett. of cyclopropyl compounds by fungal biotransfor-
31,4921-4922. mation. Can. J. Chem. 68, 696-700.
115. Hall, L.R., R.T. Iwamoto, and R.P Hanzlik 127. Watabe, T. and K. Akamatsu (1974). Microsomal
(1989). Electrochemical models for cytochrome epoxidation of c/^-stilbene: Decrease in epoxidase
P-450. N-Demethylation of tertiary amides by activity related to lipid peroxidation. Biochem.
anodic oxidation. J. Org. Chem. 54, 2446-2451. Pharmacol. 23, 1079-1085.
116. Hlavica, P. and M. Lehnerer (1995). Some aspects of 128. Watabe, T., Y Ueno, and J. Imazumi (1971).
the role of cytochrome P-450 isozymes in the N- Conversion of oleic acid into //zreo-dihydroxy-
oxidative transformation of secondary and tertiary stearic acid by rat liver microsomes. Biochem.
amine compounds. J. Biochem. Toxicol. 10,275-285. Pharmacol. 20, 912-913.
117. Seto, Y. and F.P Guengerich (1993). Partitioning 129. Ortiz de Montellano, PR., B.L.K. Mangold,
between N-dealkylation and N-oxygenation in the C. Wheeler, K.L. Kunze, and N.O. Reich (1983).
oxidation of N,N-dialkylarylamines catalyzed by Stereochemistry of cytochrome P-450-catalyzed
cytochrome P450 2B1. J. Biol. Chem. 268, epoxidation and prosthetic heme alkylation. J.
9986-9997. Biol. Chem. 258, 4208-4213.
236 Paul R. Ortiz de Monteliano and James J. De Voss

130. Hanzlik, R.R and G.O. Shearer (1978). Secondary of rat hepatic lauric acid omega-hydroxylase.
deuterium isotope effects on olefin epoxidation Protein versus prosthetic heme alkylation in the
by cytochrome P450. Biochem. Pharmacol. 27, omega-hydroxylation of acetylenic fatty acids. J.
1441-1444. Biol. Chem. 263, 18640-18649.
131. Hanzlik, R.R and G.O. Shearer (1975). Transition 143. Henschler, D., WR. Hoos, H. Fetz, E. Dallmeier,
state structure for peracid epoxidation: Secondary and M. Metzler (1979). Reactions of trichloroeth-
deuterium isotope effects. J. Am. Chem. Soc. 97, ylene epoxide in aqueous systems. Biochem.
5231-5233. Pharmacol. 28, 543-548.
132. Ortiz de Monteliano, RR. and K.L. Kunze (1981). 144. Miller, R.E. and RR Guengerich (1982).
Shift of the acetylenic hydrogen during chemical Oxidation of trichloroethylene by liver microso-
and enzymatic oxidation of the biphenylacetylene mal cytochrome P-450: Evidence for chlorine
triple bond. Arch. Biochem. Biophys. 209, migration in a transition state not involving
710-712. trichloroethylene oxide. Biochemistry 21,
133. McMahon, R.E., J.C. Turner, G.W. Whitaker, and 1090-1097.
H.R. Sullivan (1981). Deuterium isotope effect in 145. Liebler, D.C. and RP Guengerich (1983). Olefin
the biotransformation of 4-ethynylbiphenyls to oxidation by cytochrome P-450: Evidence for
4-biphenylacetic acids by rat hepatic microsomes. group migration in catalytic intermediates formed
Biochem. Biophys. Res. Commun. 99, 662-667. with vinylidene chloride and ^ra«5-1-phenyl-1-
134. Ortiz de Monteliano, RR. and E.A. Komives butene. Biochemistry 22, 5482-5489.
(1985). Branchpoint for heme alkylation and 146. Mansuy, D., J. Leclaire, M. Fontecave, and M.
metabolite formation in the oxidation of aryl Momenteau (1984). Oxidation of monosubstituted
acetylenes by cytochrome P450. J. Biol. Chem. olefins by cytochromes P450 and heme models:
260, 3330-3336. Evidence for the formation of aldehydes in addi-
135. Foroozesh, M., G. Primrose, Z. Guo, L.C. Bell, tion to epoxides and allylic alcohols. Biochem.
W.L. Alworth, and RR Guengerich (1997). Aryl Biophys. Res. Commun. 119, 319-325.
acetylenes as mechanism-based inhibitors of 147. Miller, VP, J.A. Fruetel, and PR. Ortiz de
cytochrome P450-dependent monooxygenase Monteliano (1992). Cytochrome ?450cam-C2Li-
enzymes. Chem. Res. Toxicol. 10, 91-102. alyzed oxidation of a hypersensitive radical probe.
136. Ortiz de Monteliano, PR. and M.A. Correia Arch. Biochem. Biophys. 298, 697-702.
(1995). Inhibition of cytochrome P450 enzymes. 148. de Visser, S.P, F Ogliaro, N. Harris, and S. Shaik
In PR. Ortiz de Monteliano (ed.). Cytochrome (2001). Multi-state epoxidation of ethene by
P450: Structure, Mechanism and Biochemistry, cytochrome P450: A quantum chemical study. J.
2nd edn. Plenum, New York, pp. 305-364. Am. Chem. Soc. 123, 3037-3047.
137. Ortiz de Monteliano, PR. and B.A. Mico (1980). 149. de Visser, S.P, F Ogliaro, and S. Shaik (2001).
Destruction of cytochrome P-450 by ethylene and Stereospecific oxidation by compound I of
other olefins. Mo/. Pharmacol. 18, 128-135. cytochrome P450 does not proceed in a concerted
138. Kunze, K.L., B.L.K. Mangold, C. Wheeler, synchronous manner. J. Chem. Soc. Chem.
H.S. Beilan, and PR. Ortiz de Monteliano (1983). Commun. 2322-2323.
The cytochrome P-450 active site. Regiospecificity 150. de Visser, S.P, F Ogliaro, and S. Shaik (2001).
of the prosthetic heme alkylation by olefins and How does ethene inactivate cytochrome P450 en
acetylenes. J. Biol. Chem. 258, 4202-4207. route to its epoxidation? A density functional
139. Ortiz de Monteliano, PR., R.A. Stearns, and study Angew. Chem. Int. Ed. 40, 2871-2874.
K.C. Langry (1984). The allylisopropylacetamide 151. Lovern, M.R., M.J. Turner, M. Meyer, G.L.
and novonal prosthetic heme adducts. Mol. Kedderis, WE. Bechtold, and P.M. Schlosser
Pharmacol. 25,310-317. (1997). Identification of benzene oxide as a product
140. Ortiz de Monteliano, PR. and K.L. Kunze (1981). of benzene metabolism by mouse, rat, and human
Cytochrome P-450 inactivation: Structure of liver microsomes. Carcinogenesis 18, 1695-1700.
the prosthetic heme adduct with propyne. 152. Jerina, DM. and J.W. Daly (1974). Arene oxides:
Biochemistry 20, 1266-121 \. A new aspect of drug metabolism. Science 185,
141. Gan, L.-S., A.L. Acebo, and W.L. Alworth (1984). 573-582.
1-Ethynylpyrene, a suicide inhibitor of cytochrome 153. Koerts, J., A.E.M.E Soffers, J. Vervoort, A. De
P-450 dependent benzo[a]pyrene hydroxylase Jager, and I.M.C.M. Rietjens (1998). Occurrence
activity in liver microsomes. Biochemistry 23, of the NIH shift upon the cytochrome P450-cat-
3827-3836. alyzed in vivo and in vitro aromatic ring hydroxy-
142. CaJacob, C.A., W Chan, E. Shephard, and lation of fluorobenzenes. Chem. Res. Toxicol. 11,
PR. Ortiz de Monteliano (1988). The catalytic site 503-512.
Substrate Oxidation by Cytochrome P450 Enzymes 237

154. Hanzlik, R.R and K.-HJ. Ling (1993). Active site models and cytochrome P450. Arch. Biochem.
dynamics of xylene hydroxylation by cytochrome Biophys. 310, 402^09.
P-450 as revealed by kinetic deuterium isotope 165. Ohe, T, T. Mashino, and M. Hirobe (1997).
effects. J ^m. Chem. Soc. 115, 9363-9370. Substituent elimination from /7-substituted phenols
155. Riley, R and R.R Hanzlik (1994). Electron trans- by cytochrome P450. ^''^o-substitution by the oxy-
fer in P450 mechanisms. Microsomal metabolism gen atom of the active species. DrugMetab. Dispos.
of cyclopropylbenzene and /7-cyclopropylanisole. 25,116-122.
Xenobiotica 24, 1-16. 166. Rizk, RN. and R.R Hanzlik (1995). Oxidative and
156. Tomaszewski, J.E., D.M. Jerina, and J.W. Daly non-oxidative metabolism of 4-iodoanisole by rat
(1975). Deuterium isotope effects during forma- liver microsomes. Xenobiotica 25, 143-150.
tion of phenols by hepatic monooxygenases: 167. Hinson, J.A., S.D. Nelson, and J.R. Mitchell
Evidence for an alternative to the arene oxide (1977). Studies on the microsomal formation of
pathway. Biochemistry 14, 2024-2030. arylating metabolites of acetaminophen and
157. Preston, B.D., J.A. Miller, and E.G. Miller (1983). phenacetin. Mol. Pharmacol. 13, 625-633.
Non-arene oxide aromatic ring hydroxylation of 168. Hinson, J.A., S.D. Nelson, and J.R. Gillette (1979).
2,2',5,5'-tetrachlorobiphenyl as the major metabolic Metabolism of [p-^^0]-phenacetin: The mechanism
pathway catalyzed by phenobarbital-induced rat of activation of phenacetin to reactive metabolites
liver microsomes. J. Biol Chem. 258, 8304-8311. in hamsters. Mol. Pharmacol. 15, 419^27.
158. Hanzlik, R.R, K. Hogberg, and CM. Judson 169. Koymans, L., J.H.V Lenthe, G.M.D. Den Kelder,
(1984). Microsomal hydroxylation of specifically and N.P.E. Vermeulen (1990). Mechanisms of acti-
deuterated monosubstituted benzenes: Evidence vation of phenacetin to reactive metabolites by
for direct aromatic hydroxylation. Biochemistry cytochrome P-450: A theoretical study involving
23,3048-3055. radical intermediates. Mol. Pharmacol. 37,452-^60.
159. Korzekwa, K.R., D.C. Swinney, and W.R Trager 170. Veronese, M.E., S. McLean, C.A. D'Souze, and
(1989). Isotopically labeled chlorobenzenes as N.W. Davies (1985). Formation of reactive
probes for the mechanism of cytochrome P-450 metabolites of phenacetin in humans and rats.
catalyzed aromatic hydroxylation. Biochemistry Xenobiotica 15, 929-940.
28,9019-9027. 171. Rietjens, I.M.C.M., B. Tyrakowska, C. Veeger, and
160. Rietjens, I.M.C. and J. Vervoort (1992). A new J. Vervoort (1990). Reaction pathways for bio-
hypothesis for the mechanism for cytochrome dehalogenation of fluorinated anilines. Eur J.
P-450 dependent aerobic conversion of hexahalo- Biochem. 194, 945-954.
genated benzenes to pentahalogenated phenols. 172. Stresser, D.M. and D. Kupfer (1997). Catalytic
Chem. Res. Toxicol. 5, 10-19. characteristics of CYP3A4: Requirement for a
161. Rietjens, I.M.C.M., A.E.M.R Soffers, C. Veeger, phenolic function in or^/^o-hydroxylation of
and J. Vervoort (1993). Regioselectivity of estradiol and mono-O-demethylated methoxy-
cytochrome P-450 catalyzed hydroxylation of flu- chlor. Biochemistry 36, 2203-2210.
orobenzenes predicted by calculated frontier 173. Jellinck, PH., E.R Hahn, and J. Fishman (1986).
orbital substrate characteristics. Biochemistry 32, Absence of reactive intermediates in the forma-
4801^812. tion of catechol estrogens by rat liver microsomes.
162. Zakharieva, O., M. Grodzicki, A.X. Trautwein, J. Biol. Chem. 261, 7729-7732.
C. Veeger, and I.M.C.M. Rietjens (1996). Molecular 174. Sarabia, S.R, B.T. Zhu, T. Kurosawa, M. Tohma,
orbital study of the hydroxylation of benzene and and J.G. Liehr (1997). Mechanism of cytochrome
monofluorobenzene catalysed by iron-oxo por- P450-catalyzed aromatic hydroxylation of estro-
phyrin TT-cation radical complexes. J. Biol. Inorg. gens. Chem. Res. Toxicol. 10,161-111.
Chem. 1, 192-204. 175. Stadler, R. and M.H. Zenk (1993). The purifica-
163. Koerts, X, M.M.C. Velraeds, A.E.M.R Sofferse, tion and characterization of a unique cytochrome
J. Vervoort, and I.M.C.M. Rietjens (1997). P-450 enzyme from Berberis solonifera plant cell
Influence of substituents in fluorobenzene deriva- cultures. J. Biol. Chem. 268, 823-831.
tives on the cytochrome P450-catalyzed hydroxy- 176. Kraus, REX. andT.M. Kutchan (1995). Molecular
lation at the adjacent ortho aromatic carbon cloning and heterologous expression of a cDNA
center. Chem. Res. Toxicol. 10, 279-288. encoding berbamunine synthase, a C-O phenol-
164. Ohe, T., T. Mashino, and M. Hirobe (1994). Novel coupling cytochrome P450 from the higher plant
metabolic pathway of arylethers by cytochrome Berberis stolonifera. Proc. Natl. Acad. Sci. USA.
P450: Cleavage of the oxygen-aromatic ring bond 92, 2071-2075.
accompanying i/75o-substitution by the oxygen 177. Amann, T. and M.H. Zenk (1991). Formation of
atom of the active species in cytochrome P450 the morphine precursor salutaridine is catalyzed
238 Paul R. Ortiz de Monteliano and James J. De Voss

by a cytochrome P-450 enzyme in mammalian 189. CavaUeri, E. and E. Rogan (1995). Central role of
liver. Tetrahedron Lett. 32, 3675-3678. radical cations in metabolic activation of poly-
178. Gerardy, R. and M.H. Zenk (1993). Formation of cyclic aromatic hydrocarbons. Xenobiotica 25,
salutaridine from (i?)-reticuline by a membrane- 677-688.
bound cytochrome P450 enzymes from Papaver 190. Cavalieri, E., E. Rogan, R.W Roth, R.K. Saugier,
somniferum. Phytochemistry 32, 79-86. and A. Hakam (1983). The relationship between
179. Nasreen, A., M. Rueffer, and M.H. Zenk (1996). ionization potential and horseradish peroxidase/
Cytochrome P-450-dependent formation of isoan- hydrogen peroxide-catalyzed binding of aromatic
drocymbine from autumnaline in colchicine hydrocarbons to DNA. Chem. Biol. Interact. 47,
biosynthesis. Tetrahedron Lett. 37, 8161-8164. 87-109.
180. Zerbe, K., O. Pylypenko, F. Vitali, W. Zhang, 191. Devanesan, P., E. Rogan, and E. Cavalieri (1987).
S. Rouset, M. Heck et al (2002). Crystal structure of The relationship between ionization potential and
OxyB, a cytochrome P450 implicated in an oxida- prostaglandin H synthase-catalyzed binding of
tive phenol coupling reaction during vancomycin aromatic hydrocarbons to DNA. Chem. Biol.
biosynthesis. J. Biol. Chem. 27, 47476-47485. Interact. 61, 89-95.
181. Bischoff, D., S. Pelzer, B. Bister, G.J. Nicholson, 192. Cavalieri, E.L., E.G. Rogan, PD. Devanesan,
S. Stockert, M. Schirle et al. (2001). The biosyn- P. Cremonesi, R.L. Cerny, M.L. Gross et al.
thesis of vancomycin-type glycopeptide antibi- (1990). Binding of benzo[a]pyrene to DNA by
otics—The order of the cyclization steps. Angew. cytochrome P450-catalyzed one-electron oxida-
Chem. Int. Ed. 40, 4688-4691. tion in rat liver microsomes and nuclei.
182. van Wageningen, A.M., P.N. Kirkpatrick, D.H. Biochemistry 29, 4820-4827.
Williams, B.R. Harris, J.K. Kershaw, N.J. Lennard 193. Rogan, E.G., PD. Devanesan, N.VS.
et al. (1998). Sequencing and analysis of genes RamaKrishna, S. Higginbotham, N.S. Padmavathi,
involved in the biosynthesis of a vancomycin K. Chapman et al. (1993). Identification and quan-
group antibiotic. Chem. Biol. 5, 155-162. titation of benzo[a]pyrene-DNA adducts formed in
183. Pelzer, S., R. Sussmuth, D. Heckmann, mouse skin. Chem. Res. Toxicol. 6, 356-363.
J. Recktenwald, P Huber, G. Jung et al. (1999). 194. Khandwala, A.S. and C.B. Kasper (1973).
Identification and analysis of the balhimycin Preferential induction of aryl hydroxylase activity
biosynthetic gene cluster and its use for manipu- in rat liver nuclear envelope by 3-methylcholan-
lating glycopeptide biosynthesis in Amycolatopsis threne. Biochem. Biophys. Res. Commun. 54,
mediterranei DSM5908. Antimicrob. Agents 1241-1246.
Chemother. 43, 1565-1573. 195. Cavalieri, E.L., E.G. Rogan, P Cremonesi, and
184. Chiu, H.-T, B.K. Hubbard, A.N. Shah, J. Eide, PD. Devanesan (1988). Radical cations as pre-
R.A. Fredenburg, C.T Walsh et al. (2001). cursors in the metabolic formation of quinones
Molecular cloning and sequence analysis of the from benzo[a]pyrene and 6-fluorobenzo[a]pyrene.
complestatin biosynthetic gene cluster. Proc. Natl. Fluoro substitution as a probe for one-electron
Acad Sci. USA. 98, 8548-8553. oxidation in aromatic substrates. Biochem.
185. Sato, H. and FP Guengerich (2000). Oxidation of Pharmacol. 31, 2\13-2\S2.
1,2,4,5-tetramethoxybenzene to a cation radical 196. Rettie, A.E., A.W Rettenmeier, W.N. Howald, and
by cytochrome P450. J. Am. Chem. Soc. 122, T.A. Baillie (1987). Cytochrome P-450-catalyzed
8099-8100. formation of A'^-VPA, a toxic metabolite of val-
186. Anzenbacher, P, T Niwa, L.M. Tolbert, S.R. proic acid. Science 235, 890-893.
Sirimanne, and F.P Guengerich (1996). Oxidation 197. Rettie, A.E., M. Boberg, A.W. Rettenmeier, and
of 9-alkylanthracenes by cytochrome P450 2B1, T.A. Baillie (1988). Cytochrome P-450-catalyzed
horseradish peroxidase, and iron tetraphenylpor- desaturation of valproic acid in vitro. Species dif-
phine/iodosylbenzene systems: Anaerobic and aer- ferences, induction effects, and mechanistic stud-
obic mechanisms. Biochemistry 35, 2512-2520. ies. ^ 5/o/. Chem. 263, 13733-13738.
187. Ts'o, PO., Caspary, W.J. and R.J. Lorentzen 198. Porubeck, D.J., H. Barnes, G.P Meier, L.J.
(1977). The involvement of free radicals in chem- Theodore, and T.A. Baillie (1989). Enantiotopic
ical carcinogenesis. In WA. Pryor (ed.). Free differentiation during the biotransformation of
Radicals in Biology vol. III. Academic Press, New valproic acid to the hepatotoxic olefin 2-«-propyl-
York, pp. 251-303. 4-pentenoic acid. Chem. Res. Toxicol. 2, 3 5 ^ 0 .
188. Cavalieri, E.L. and E.G. Rogan (1992). The 199. Kassahun, K. and TA. Baillie (1993).
approach to understanding aromatic hydrocarbon Cytochrome P-450-mediated dehydrogenation of
carcinogenesis. The central role of radical cations in 2-n-propyl-2(£)-pentenoic acid, a pharmacologi-
metabolic activation. Pharmacol. Ther. 55,183-199. cally-active metabolite of valproic acid, in rat liver
Substrate Oxidation by Cytochrome P450 Enzymes 239

microsomal preparations. Drug Metab. Dispos. 211. Lamb, D.C, S. Maspahy, D.E. Kelly, N.J.
21, 242-248. Manning, A. Geber, J.E. Bennett et al (1999).
200. Sadeque, A.J.M., M.B. Fisher, K.R. Korzekwa, F.J. Purification, reconstitution, and inhibition of
Gonzalez, and A.E. Rettie (1997). Human cytochrome P-450 sterol A^^-desaturase from the
CYP2C9 and CYP2A6 mediate formation of the pathogenic fungus Candida glabrata. Antimicrob.
hepatotoxin 4-ene-valproic acid. J. Pharmacol Agents Chemother. 43, 1725-1728.
Exp. Ther. 283, 698-703. 212. Rodrigues, C.M.P, B.T. Kren, C.J. Steer, and
201. Fisher, M.B., S.J. Thompson, V Ribeiro, M.C. K.D.R. Setchell (1996). Formation of D22-bile
Lechner, and A.E. Rettie (1998). P450-catalyzed in- acids in rats is not gender specific and occurs in
chain desaturation of valproic acid: Isoform selec- the peroxisome. J. Lipid Res. 37, 540-550.
tivity and mechanism of formation of A^-valproic 213. Nagata, K., D.J. Liberato, J.R. Gillette, and H.A.
acid generated by baculovirus-expressed CYP3A1. Sasame (1986). An unusual metabolite of testos-
Arch. Biochem. Biophys. 356, 63-70. terone: 17p-hydroxy-4,6-androstadiene-3-one. Drug
202. Rettie, A.E., PR. Sheffels, K.R. Korzekwa, FJ. Metab. Dispos. 14, 559-565.
Gonzalez, R.M. Philpot, and T.A. Baillie (1995). 214. Aoyama, T., K. Korzekwa, K. Nagata, J. Gillette,
CYP4 isozyme specificity and the relationship H.V Gelboin, and FJ. Gonzalez (1989). cDNA-
between co-hydroxylation and terminal desatura- directed expression of rat testosterone 7a-hydrox-
tion of valproic acid. Biochemistry 34, 7889-7895. ylase using the modified vaccinia virus,
203. Rettenmeier, A.W., W.P Gordon, K.S. Prickett, R.H. T7-RNA-polymerase system and evidence for
Levy, J.S. Lockard, K.E. Thummel et al. (1986). 6n-hydroxylation and A^-testosterone formation.
Metabolic fate of valproic acid in the rhesus mon- Eur J. Biochem. 181, 331-336.
key. Formation of a toxic metabolite, 2-n-propyl-4- 215. Korzekwa, K.R., W.K Trager, K. Nagata, A.
pentenoic acid. Drug Metab. Dispos. 14, 443^53. Parkinson, and J.R. Gillette (1990). Isotope effect
204. Pennanen, S., A. Kojo, M. Pasanen, J. Liesivuori, studies on the mechanism of the cytochrome P-
R.O. Juvonen, and H. Kumulainen (1996). CYP 450IIA1-catalyzed formation of A^-testosterone
enzymes catalyze the formation of a terminal from testosterone. Drug Metab. Dispos. 18,
olefin from 2-ethylhexanoic acid in rat and human 974-979.
liver. Hum. Exp. Toxicol. 15, 435-442. 216. Chadwick, R.W., L.T. Chuang, and K. Williams
205. Behrouzian, B. and PH. Buist (2002). Fatty acid (1975). Dehydrogenation: A previously unre-
desaturation: Variations on an oxidative theme. ported pathway of lindane metabolism in mam-
Curr. Opin. Chem. Biol 6, 577-582. mals. Pestic. Biochem. Physiol 5, 575-586.
206. Obach, R.S. (2001). Mechanism of cytochrome 217. Mochizuki, H., K. Suhara, and M. Katagiri (1992).
P4503A4- and 2D6-catalyzed dehydrogenation of Steroid 6(3-hydroxylase and 6-desaturase reactions
ezlopitant as probed with isotope effects using catalyzed by adrenocortical mitochondrial P-450.
five deuterated analogs. Drug Metab. Dispos. 29, J. Steroid Biochem. Mol Biol 42, 95-101.
1599-1607. 218. Boyd, D.R., N.D. Sharma, R. Agarwal, R.A.S.
207. Hold, K.M., N.S. Sirisoma, and IE. Casida (2001). McMordie, J.G.M. Bessems, B. van Ommen et al
Detoxification of a- and P-thujones (the active (1993). Biotransformation of 1,2-dihydronaphtha-
ingredients of absinthe): Site specificity and species lene and 1,2-dihydroanthracene by rat liver
differences in cytochrome P450 oxidation in vitro microsomes and purified cytochromes P-450.
and in vivo. Chem. Res. Toxicol 14, 589-595. Formation of arene hydrates of naphthalene and
208. Hata, S., T. Nishino, M. Komori, and H. Katsuki anthracene. Chem. Res. Toxicol 6, 808-812.
(1981). Involvement of cytochrome P-450 in A^^- 219. Adams, J.D., H. Yagi, W. Levin, and D.M. Jerina
desaturation in ergosterol biosynthesis in yeast. (1995). Stereo-selectivity and regio-selectivity in
Biochem. Biophys. Res. Commun. 103, 272-277. the metabolism of 7,8-dihydrobenzo[a]pyrene by
209. Kelly, S.L., D.C. Lamb, B.C. Baldwin, A.J. cytochrome P450, epoxide hydrolase and hepatic
Corran, and D.E. Kelly (1997). Characterization microsomes from 3-methylcholanthrene-treated
of Saccharomyces cerevisiae CYP61, sterol A22- rats. Chem. Biol Interact. 95, 57-77.
desaturated, and inhibition by azole antifungal 220. Kaminsky, L.S., M.J. Fasco, and F.P Guengerich
agents. J. Biol Chem. 272, 9986-9988. (1980). Comparison of different forms of purified
210. Skaggs, B.A., J.F Alexander, C.A. Pierson, K.S. cytochrome P-450 from rat liver by immunologi-
Schweitzer, K.T. Chun, C. Koegel et al (1996). cal inhibition of regio- and stereoselective metab-
Cloning and characterization of the Saccharomyces olism of warfarin. J. Biol Chem. 255, 85-91.
cerevisiae C-22 sterol desaturase gene, encoding a 221. Vyas, K.R, PH. Kari, S.R. Prakash, and D.E.
second cytochrome P-450 involved in ergosterol Duggan (1990). Biotransformation of lovastatin.
biosynthesis. Gene 169, 105-109. II. In vitro metabolism by rat and mouse liver
240 Paul R. Ortiz de Montellano and James J. De Voss

microsomes and involvement of cytochrome 232. Guengerich, FR, W.R. Brian, M. Iwasaki, M.-A.
P-450 in dehydrogenation of lovastatin. Drug Sari, C. Baarnhielm, and P. Berntsson (1991).
Metab. Dispos. 18, 218-222. Oxidation of dihydropyridine calcium channel
222. Wang, R.W., P.H. Kari, A.Y.H. Lu, RE. Thomas, F.P. blockers and analogues by human liver
Guengerich, and K.P. Vyas (1991). Biotrans- cytochrome P-450 IIIA4. J. Med. Chem. 34,
formation of lovostatin. IV Identification of 1838-1844.
cytochrome P450 3A proteins as the major enzymes 233. Skiles, G.L. and G.S. Yost (1996). Mechanistic
responsible for the oxidative metabolism of lovo- studies on the cytochrome P450-catalyzed dehy-
statin in rat and human liver microsomes. Arch. drogenation of 3-methylindole. Chem. Res.
Biochem. Biophys. 290, 355-361. Toxicol. 9, 291-297.
223. Vickers, S. and C.A. Duncan (1991). Studies on 234. Zuleski, FR., K.M. Kirkland, M.D. Melgar and
the metabolic inversion of the 6' chiral center of J.O. Malbica (1985). Tracazolate metabolites in rat
simvastatin. Biochem. Biophys. Res. Commun. tissue. Drug Metab. Dispos. 13, 129-147.
181, 1508-1515. 235. Byon, C.-Y and M. Gut (1980). Steric considera-
224. Guan, X., M.B. Fisher, D.H. Lang, Y.-M. Zhen, tions regarding the biodegradation of cholesterol to
D.R. Koop, and A.E. Rettie (1998). Cytochrome pregnenolone. Exclusion of (22iS)-22-hydroxycho-
P450-dependent desaturation of lauric acid: lesterol and 22-ketocholesterol as intermediates.
Isoform selectivity and mechanism of formation Biochem. Biophys. Res. Commun. 94, 549-552.
of 11-dodecenoic acid. Chem. Biol. Interact. 110, 236. Burstein, S., B.S. Middleditch, and M. Gut (1975).
103-131. Mass spectrometric study of the enzymic conver-
225. Kelkar, H.S., T.W. Skloss, J.F. Haw, N.R Keller, sion of cholesterol to (22i?)-22-hydroxycholes-
and T.H. Adams (1997). Aspergillus nidulans stcL terol, (20/?,22/?)-20,22-dihydroxycholesterol, and
encodes a putative cytochrome P-450 monooxy- pregnenolone, and of (22R)-22- hydroxycholes-
genase required for bisfuran desaturation during terol to the glycol and pregnenolone in bovine
aflatoxin and sterigmatocystin biosynthesis. J. adrenocortical preparations. Mode of oxygen
Biol. Chem. 272, 1589-1594. incorporation. J. Biol. Chem. 250, 9028-9037.
226. Akashi, T., M. Fukuchi-Mizutani, T. Aoki, Y. 237. Lambeth, J.D, S.E. Kitchen, A.A. Farooqui, R.
Ueyama, K. Yonekura-Sakakibara, Y. Tanaka et al. Tuckey, and H. Kamin (1982). Cytochrome
(1999). Molecular cloning and biochemical char- P-450scc-substrate interactions. Studies of bind-
acterization of a novel cytochrome P450, flavone ing and catalytic activity using hydroxycholes-
synthase II, that catalyzes direct conversion of terols. J. Biol. Chem. 257, 1876-1884.
flavanones to flavones. Plant Cell Physiol. 40, 238. Tuckey, R.C. and H. Kamin (1983). Kinetics of
1182-1186. oxygen and carbon monoxide binding to adrenal
227. Guengerich, FR and D.H. Kim (1991). Enzymatic cytochrome P-450scc. Effect of cholesterol, inter-
formation of ethyl carbamate to vinyl carbamate and mediates, and phosphatidylcholine vesicles.
its role as an intermediate in the formation of 1,N^- J. Biol. Chem. 258, 4232-4237.
ethenoadenosine. Chem. Res. Toxicol. 4, 413^21. 239. Tuckey, R.C. and H. Kamin (1982). The oxyferro
228. Wood, A.W, DC. Swinney, RE. Thomas, D.E. complex of adrenal cytochrome P-450scc. Effect
Ryan, RF Hall, W Levin et al. (1988). of cholesterol and intermediates on its stability
Mechanism of androstenedione formation from and optical characteristics. J. Biol. Chem. 257,
testosterone and epitestosterone catalyzed by puri- 9309-9314.
fied cytochrome P-450b. J. Biol. Chem. 263, 240. Primus, J.-L., K. Teunis, D Mandon, C. Veeger,
17322-17332. and I.M.C.M. Rietjens (2000). A Mechanism for
229. Swinney, D C , D.E. Ryan, RE. Thomas, and W oxygen exchange between ligated oxometallopor-
Levin (1988). Evidence for concerted kinetic oxi- phinates and bulk water. Biochem. Biophys. Res.
dation of progesterone by purified rat hepatic Commun. Ill, 551-556.
cytochrome P-450g. Biochemistry 27, 5461-5470. 241. Hochberg, R.B., RD. McDonald, M. Feldman, and
230. Nelson, S.D, A.J. Forte, and D.C. Dhalin (1980). S. Lieberman (1974). Biosynthetic conversion of
Lack of evidence for N-hydroxyacetaminophen as cholesterol into pregnenolone. Side chain cleavage
a reactive metabolite of acetaminophen in vitro. of some 20-/7-tolyl analogs of cholesterol and 20a-
Biochem. Pharmacol. 29, 1617-1620. hydroxycholesterol. J:5/O/. Chem. 249, 1274-1285.
231. Lee, J.S., N.E. Jacobsen, and PR. Ortiz de 242. Bower, S., J.B. Perkins, R.R. Yocum, C.L. Howitt,
Montellano (1988). 4-Alkyl radical extrusion in P. Rahaim, and J. Pero (1996). Cloning, sequenc-
the cytochrome P-450-catalyzed oxidation of ing, and characterization of the Bacillus subtilis
4-alkyl-l,4-dihydropyridines. Biochemistry 27, biotin biosynthetic operon. J. Bacteriol. 178,
7703-7710. 4122-4130.
Substrate Oxidation by Cytoclirome P450 Enzymes 241

243. Stok, J.E. and J.J. De Voss (2000). Expression, 255. Mak, A.Y and D.C. Swinney (1992). 17-0-
purification, and characterization of Biol: A car- Acetyltestosterone formation from progesterone
bon-carbon bond cleaving cytochrome P450 in microsomes from pig testes: Evidence for the
involved in biotin biosynthesis in Bacillus subtilis. Baeyer-Villiger rearrangement in androgen for-
Arch. Biochem. Biophys. 384, 351-360. mation catalyzed by CYP17. J. Am. Chem. Soc.
244. Green, A.J., S.L. Rivers, M. Cheesman, G.A. Reid, 114,8309-8310.
L.G. Quaroni, I.D.G. Macdonald et al. (2001). 256. Akhtar, M., D. Corina, S. Miller, A.Z. Shyadehi,
Expression, purification and characterization of and J.N. Wright (1994). Mechanism of the acyl-
cytochrome P450 Biol: A novel P450 involved in carbon cleavage and related reactions catalyzed by
biotin synthesis in Bacillus subtilis. J. Biol. Inorg. multifunctional P-450s: Studies on cytochrome P-
Chem. 6, 523-533. 45017a. Biochemistry 33, 4410-4418.
245. Cryle, M.J., N.J. Matovic, and J.J. De Voss (2003). 257. Akhtar, M., D.L. Corina, S.L. Miller, A.Z.
Products of cytochrome P450BioI (CYP107H1)- Shyadehi, and J.N. Wright (1994). Incorporation
catalysed oxidation of fatty acids. Org. Lett. 5, of label from 18O2 into acetate during side-chain
3341-3344. cleavage catalyzed by cytochrome P-45017a
246. Cryle, M.J. and J.J. De Voss (2004). Carbon car- (17a-hydroxylase-17,20-lyase). J. Chem. Soc,
bon bond cleavage by cytochrome P450BioI Perkin Trans. I, 263-267.
(CYP107H1). Chem. Commun. 86-87. 258. Lee-Robichaud, P, M.E. Akhtar, and M. Akhtar
247. Cryle, M.J., J.E. Stok, and J.J. De Voss (2003). (1998). An analysis of the role of active site protic
Reactions catalyzed by bacterial cytochromes residues of cytochrome P-450s: Mechanistic and
P450. Aust. J. Chem. 56, 749-762. mutational studies on 17a-hydroxylase-17,20-
248. Umehara, K., S. Kudo, Y. Hirao, S. Morita, T. lyase (P-45017a also CYP17). Biochem. J. 330,
Ohtani, M. Uchida et al. (2000). In vitro charac- 967-974.
terization of the oxidative cleavage of the octyl 259. Swinney, D.C. and A.Y Mak (1994). Androgen
side chain of olanexidine, a novel antimicrobial formation by cytochrome P450 CYP17. Solvent
agent, in dog liver microsomes. Drug Metab. isotope effect and pL studies suggest a role for
Dispos. 28, 1417-1424. protons in the regulation of oxene versus peroxide
249. Umehara, K., S. Kudo, Y Hirao, S. Morita, M. chemistry. Biochemistry 33, 2185-2190.
Uchida, M. Odomi et al. (2000). Oxidative cleavage 260. Lee-Robichaud, P, A.Z. Shyadehi, J.N. Wright, M.
of the octyl side chain of l-(3,4-dichlorobenzyl)- Akhtar, and M. Akhtar (1995). Mechanistic kin-
5-octylbiguanide (OPB-2045) in rat and dog liver ship between hydroxylation and desaturation reac-
preparations. Drug Metab. Dispos. 28, 887-894. tions: Acyl-carbon bond cleavage promoted by pig
250. Barnes, H.J., M.R Arlotto, and M.R. Waterman and human CYP17 (P-45017a; 17a-hydroxylase-
(1991). Expression and enzymatic activity of 17,20-lyase). Biochemistry 34, 14104-14113.
recombinant cytochrome P450 17 a-hydroxylase 261. Davis, S.C, Z. Sui, J.A. Peterson, and PR. Ortiz
in Escherichia coli. Proc. Natl. Acad. Sci. USA 88, de Montellano (1996). Oxidation of w-oxo fatty
5597-5601. acids by cytochrome P450BM-3 (CYP102). Arch.
251. Zuber, M.X., E.R. Simpson, and M.R. Waterman Biochem. Biophys. 328, 3 5 ^ 2 .
(1986). Expression of bovine 17 a-hydroxylase 262. Thompson, E.A., Jr. and PK. Siiteri (1974). The
cytochrome P-450 cDNA in nonsteroidogenic involvement of human placental microsomal
(COS 1) cells. Science 234, 1258-1261. cytochrome P-450 in aromatization. J. Biol.
252. Nakajin, S., M. Takahashi, M. Shinoda, and RE Hall Chem. 249, 5373-5378.
(1985). Cytochrome b5 promotes the synthesis of 263. Kellis, XT, Jr. and L.E. Vickery (1987).
A^^-C19 steroids by homogeneous cytochrome Purification and characterization of human pla-
P-450 C21 side-chain cleavage from pig testis. cental aromatase cytochrome P-450. J. Biol.
Biochem. Biophys. Res. Commun. 132, 708-713. Chem. 262,4413^420.
253. Shimizu, K. (1978). Formation of 5-[17P-2H] 264. Thompson, E.A., Jr. and PK. Siiteri (1974).
androstene-3p,17a-diol from 3p-hydroxy-5- Utilization of oxygen and reduced nicotinamide
[17,21,21,21-'^H]pregnen-20-one by the microsomal adenine dinucleotide phosphate by human placen-
fraction of boar testis. J. Biol. Chem. 253, tal microsomes during aromatization of andro-
4237-4241. stenedione. J. Biol. Chem. 249, 5364-5372.
254. Corina, D.L., S.L. Miller, J.N. Wright, and M. 265. Caspi, E., T. Arunachalam, and PA. Nelson (1983).
Akhtar (1991). The mechanism of cytochrome P- Biosynthesis of estrogens: The steric mode of the
450 dependent carbon-carbon bond cleavage: initial C-19 hydroxylation of androgens by human
Studies on 17a-hydroxylase-17,20-lyase. Chem. placental aromatase. J. Am. Chem. Soc. 105,
Commun. 782-783. 6987-6989.
242 Paul R. Ortiz de Montellano and James J. De Voss

266. Caspi, E., T. Arunachalam, and P.A. Nelson demethylation in estrogen biosynthesis. Biochem.
(1986). Biosynthesis of estrogens: Aromatization J. 201, 569-580.
of (19i^)-, (195)-, and (\9RS)-[19-^HMM]-^^- 279. Caspi, E., J. Wicha, T. Arunachalam, R Nelson,
hydroxyandrost-5-en-17-ones by human placental and G. Spiteller (1984). Estrogen biosynthesis:
aromatase. J: ^m. Chem. Soc. 108, 1847-1852. Concerning the obligatory intermediacy of 2^-
267. Osawa, Y., K. Shibata, D. Rohrer, C. Weeks, and hydroxy-10(3-formyl androst-4-ene-3,17-dione. J.
W.L. Duax (1975). Reassignment of the absolute Am. Chem. Soc. 106, 7282-7283.
configuration of 19-substituted 19-hydroxys- 280. Morand, R, D.G. Williamson, D.S. Layne,
teroids and stereomechanism of estrogen biosyn- L. Lompa-Krzymien, and J. Salvador (1975).
thesis. J. Am. Chem. Soc. 97, 4400-4402. Conversion of an androgen epoxide into 17^-
268. Arigoni, D., R. Battaglia, M. Akhtar, and T. Smith estradiol by human placental microsomes.
(1975). Stereospecificity of oxidation at C-19 in Biochemistry 14, 635-638.
estrogen biosynthesis. Chem. Comm. 185-186. 281. Hosoda, H. and J. Fishman (1974). Unusually facile
269. Miyairi, S. and J. Fishman (1983). Novel method aromatization of 2b-hydroxy-19-oxo-4-androstene-
of evaluating biological 19-hydroxylation and 3,17-dione to estrone. Implications in estrogen
aromatization of androgens. Biochem. Biophys. biosynthesis. J. Am. Chem. Soc. 96, 7325-7329.
Res. Commun. Ill, 392-398. 282. Goto, J. and J. Fishman (1977). Participation of a
270. Miyairi, S. and J. Fishman (1985). Radiometric nonenzymic transformation in the biosynthesis of
analysis of oxidative reactions in aromatization by estrogens from androgens. Science 195, 80-81.
placental microsomes. Presence of differential 283. Covey, D.R and WF Hood (1982). A new hypoth-
isotope effects. J. Biol. Chem. 260, 320-325. esis based on suicide substrate inhibitor studies
271. Numazawa, M., K. Midzuhashi, and M. Nagaoka for the mechanism of action of aromatase. Cancer
(1994). Metabolic aspects of the Ip-proton and Res. 42, 3327-3333.
the 19-methyl group of androst-4-ene-3,6,17-tri- 284. Beusen, D.D. and D.F Covey (1984). Study of the
one during aromatization by placental micro- role of Schiff base formation in the aromati-
somes and inactivation of aromatase. Biochem. zation of 3-[i^O]testosterone and 3,17-di-[^^0]
Pharmacol. 41,1X1-126. androstenedione by human placental aromatase. J.
212. Brodie, H.J., K.J. Kripalani, and G. Possanza Steroid Biochem. 20, 931-934.
(1969). Mechanism of estrogen biosynthesis. VI. 285. Numazawa, M., A. Yoshimura, M. Tachibana,
The stereochemistry of hydrogen elimination at C-2 M. Shelangouski, and M, Ishikawa (2002). Time-
during aromatization. J. Am. Chem. Soc. 91, dependent aromatase inactivation by 4p,5p-epox-
1241-1242. ides of the natural substrate androstenedione and
273. Fishman, J. and H. Guzik, (1969). Stereochemistry its 19-oxygenated analogs. Steroids 67, 185-193.
of estrogen biosynthesis. J. Am. Chem. Soc. 91, 286. Cole, PA. and C.H. Robinson (1990). Conversion
2805-2806. of 19-oxo[2b-2H]androgens into estrogens by
274. Fishman, J., H. Guzik, and D. Dixon (1969). human placental aromatase. An unexpected stere-
Stereochemistry of estrogen biosynthesis. ochemical outcome. Biochem. J. 268, 553-561.
Biochemistry 8, 4304-4309. 287. Swinney, D.C., D.M. Watson, and O.Y So (1993).
275. Fishman, J. and M.S. Raju (1981). Mechanism of Accumulation of intermediates and isotopically
estrogen biosynthesis. Stereochemistry of C-1 sensitive enolization distinguish between aro-
hydrogen elimination in the aromatization of 2(3- matase (cytochrome P450 CYP19) from rat ovary
hydroxy-19-oxoandrostenedione. J. Biol. Chem. and human placenta. Arch. Biochem. Biophys.
256, 4472-4477. 305, 61-67.
276. Townsley, J.D. and H.J. Brodie (1968). Mechanism 288. Akhtar, M., D. Corina, J. Pratt, and T. Smith
of estrogen biosynthesis. III. Stereochemistry of (1976). Studies on the removal of C-19 in estrogen
aromatization of C19 and CI 8 steroids. biosynthesis using oxygen-18. Chem. Commun.
Biochemistry 7, 33-40. 854-856.
277. Osawa, Y., N. Yoshida, M. Fronckowiak, and 289. Stevenson, D.E., J.N. Wright, and M. Akhtar
J. Kitawaki (1987). Immunoaffinity purification (1988). Mechanistic consideration of P-450
of aromatase cytochrome P-450 from human dependent enzymic reactions: Studies on oestriol
placental microsomes, metabolic switching from biosynthesis. J. Chem. Soc, Perkin Trans. I
aromatization at 1(3 and 2(3-monohydroxylation, 2043-2052.
and recognition of aromatase isozymes. Steroids 290. Akhtar, M., VC.O. Njar, and J.N. Wright (1993).
50, 11-28. Mechanistic studies on aromatase and related
278. Akhtar, M., M.R. Calder, D.L. Corina, and J.N. carbon-carbon bond cleaving P-450 enzymes. J.
Wright (1982). Mechanistic studies on C-19 Steroid. Biochem. Mol. Biol. 44, 375-387.
Substrate Oxidation by Cytochrome P450 Enzymes 243

291. Wertz, D.L., M.F. Sisemore, M. Selke, J. Driscoll, cytochrome to whole process of lanosterol 14a-
and J.S. Valentine (1998). Mimicking cytochrome demethylation. J 5/o/. Chem. 262, 1239-1243.
P-450 2B4 and aromatase: Aromatization of a 304. Aoyama, Y, Y Yoshida, Y Sonoda, and Y Sato
substrate analog by a peroxo Fe(lll) porphyrin (1989). Deformylation of 32-oxo-24,25-dihy-
complex. J. Am. Chem. Soc. 120, 5331-5332. drolanosterol by the purified cytochrome
292. Goto, Y, S. Wada, I. Morishima, and Y. Watanabe P-45014DM (lanosterol 14a-demethylase) from
(1998). Reactivity of peroxoiron(III) porphyrin yeast evidence confirming the intermediate step
complexes: Models for deformylation reactions cat- of lanosterol 14a-demethylation. J. Biol. Chem.
alyzed by cytochrome P-450. J. Inorg. Biochem. 69, 264, 18502-18505.
241-247. 305. Trzaskos, X, S. Kawata, and LL. Gaylor (1986).
293. Cole, P.A. and C.H. Robinson (1988). Peroxide Microsomal enzymes of cholesterol biosynthesis.
model reaction for placental aromatase. J. Am. Purification of lanosterol 14a-methyl demethy-
Chem. Soc. 110, 1284-1285. lase cytochrome P-450 from hepatic microsomes.
294. Graham-Lorence, S., B. Amameh, R.E. White, J. Biol. Chem. 261, 14651-14657.
J.A. Peterson, and E.R. Simpson (1995). A three- 306. Sono, H., Y Sonoda, and Y Sato (1991).
dimensional model of aromatase cytochrome Purification and characterization of cytochrome
P450. Protein. Sci. 4, 1065-1080. P-45014DM (lanosterol 14a-demethylase) from
295. Alexander, K., M. Akhtar, R.B. Boar, J.F. McGhie, pig liver microsomes. Biochim. Biophys. Acta 1078,
and D.H.R. Barton (1972). Removal of the 388-394.
32-carbon atom as formic acid in cholesterol bio- 307. Sonoda, Y, M. Endo, K. Ishida, Y Sato, N.
synthesis. Chem. Commun. 383-385. Fukusen, and M. Fukuhara (1993). Purification of
296. Mitropoulos, K.A., G.E Gibbons, and B.E.A. a human cytochrome P-450 isoenzyme catalyzing
Reeves (1976). Lanosterol 14a-demethylase. lanosterol 14a-demethylation. Biochim. Biophys.
Similarity of the enzyme system from yeast and Acta 1170, 92-97.
rat liver. Steroids 27, 821-829. 308. Alexander, K.TW, M. Akhtar, R.B. Boar, I F
297. Canonica, L., A. Fiecchi, K.M. GalU, A.A. Scala, McGhie, and D.H.R. Barton (1971). Pathway for
G. Galh, E. Grossi-Paoletti et al. (1968). Evidence the removal of C-32 in cholesterol biosynthesis. J.
for the biological conversion of A^'^^ sterol dienes Chem. Soc, Chem. Commun. 1479-1481.
into cholesterol. J. Am. Chem. Soc. 90, 6532-6534. 309. Akhtar, M., C.W. Freeman, D.C. Wilton, R.B.
298. Gibbons, G.E, L.J. Goad, and T.W. Goodwin Boar, and D.B. Copsey (1977). The pathway for the
(1968). Stereochemistry of hydrogen elimination removal of the 15a-methyl group of lanosterol.
from C-15 during cholesterol biosynthesis. J. The role of lanost-8-ene-3p,32-diol in cholesterol
Chem. Soc. Chem. Commun. 1458-1460. biosynthesis. Bioorg. Chem. 6, 473^81.
299. Watkinson, LA., D.C. Wilton, K.A. Munday, and 310. Akhtar, M., K. Alexander, R.B. Boar, J.F McGhie,
M. Akhtar (1971). Formation and reduction of the and D.H.R. Barton (1978). Chemical and enzymic
14,15-double bond in cholesterol biosynthesis. studies on the characterization of intermediates
Biochem. J. 121, 131-137. during the removal of the 14a-methyl group in cho-
300. Shafiee, A., LM. Trzaskos, YK. Paik, and lesterol biosynthesis. The use of 32-functionalized
J.L. Gaylor (1986). Oxidative demethylation of lano- lanostane derivatives. Biochem. J. 169, 449^63.
sterol in cholesterol biosynthesis: Accumulation of 311. Trzaskos, LM., R.T Fischer, S.S. Ko, R.L.
sterol intermediates. J. Lipid Res. 27, 1-10. Magolda, S. Stam, P Johnson et al. (1995).
301. Trzaskos, IM., R.T. Fischer, and M.F. Favata Substrate-based inhibitors of lanosterol 14a-
(1986). Mechanistic studies of lanosterol C-32 methyl demethylase: II. Time-dependent enzyme
demethylation. Conditions which promote oxys- inactivation by selected oxylanosterol analogs.
terol intermediate accumulation during the Biochemistry 34, 9677-9681.
demethylation process. J. Biol. Chem. 261, 312. Shyadehi, A.Z., D.C. Lamb, S.L. Kelly, D.E. Kelly,
16936-16937. W.-H. Schunck, J.N. Wright et al. (1996). The
302. Saucier, S.E., A.A. Kandutsch, S. Phirwa, and mechanism of the acyl-carbon bond cleavage
T.A. Spencer (1987). Accumulation of regulatory reaction catalyzed by recombinant sterol 14a-
oxysterols, 32-oxolanosterol and 32-hydroxy- demethylase of Candida albicans (other names
lanosterol in mevalonate-treated cell cultures. J. are: lanosterol 14a-demethylase, P-45014DM,
Biol. Chem. 262, 14056-14062. and CYP51). J. Biol. Chem. 271, 12445-12450.
303. Aoyama, Y, Y Yoshida, Y Sonoda, and Y Sato 313. Ramm, PL and E. Caspi (1969). Stereochemistry
(1987). Metabolism of 32-hydroxy-24,25-dihy- of tritium at carbon atoms 1, 7, and 15 in choles-
drolanosterol by purified cytochrome P 45014DM terol derived from mevalonic-(3i^,2i?)-2-^H acid.
from yeast. Evidence for contribution of the J. Biol. Chem. 244, 6064-6073.
244 Paul R. Ortiz de Montellano and James J. De Voss

314. Akhtar, M., A.D. Rahimtula, LA. Watkinson, D.C. implications for catalysis by oxygen-derived per-
Wilton, and K.A. Munday (1969). Status of C-6, oxide. J .4m. Chem. Soc. 113, 5886-5887.
C-7, C-15, and C-16 hydrogen atoms in choles- 325. Raner, G.M., E.W Chiang, A.D.N. Vaz, and M.J.
terol biosynthesis. Eur. J. Biochem. 9, 107-111. Coon (1997). Mechanism-based inactivation of
315. R.T. Fischer, J.M. Trzaskos, R.L. Magolda, S.S. cytochrome P450 2B4 by aldehydes: Relationship
Ko, C.S. Brosz, and B. Larsen (1991). Lanosterol to aldehyde deformylation via a peroxyhemiacetal
14a-methyl demethylase. Isolation and character- intermediate. Biochemistry 36, 4895^902.
ization of the third metabolically generated oxida- 326. Vaz, A.D.N., K.J. Kessell, and M.J. Coon (1994).
tive demethylation intermediate. J. Biol. Chem. Aromatization of a bicyclic steroid analog, 3-oxode-
266, 6124-6132. calin-4-ene-lO-carboxaldehyde, by liver microso-
316. Lamb, D.C, D.E. Kelly, M.R. Waterman, M. mal cytochrome P450 2B4. Biochemistry 33,
Stromstedt, D. Rozman, and S.L. Kelly (1999). 13651-13661.
Characteristics of the heterologously expressed 327. Vaz, A.D., S.J. Pernecky, G.M. Raner, and M.J.
human lanosterol 14a-demethylase (other names: Coon (1996). Peroxo-iron and oxenoid-iron
P45014DM, CYP51, P45051) and inhibition of species as alternative oxygenating agents in
the purified human and Candida albicans CYP51 cytochrome P450-catalyzed reactions: Switching
with azole antifungal agents. Yeast 15, 755-763. by threonine-302 to alanine mutagenesis of
317. Sloane, D.L., O.-Y. So, R. Leung, L.E. Scarafia, cytochrome P450 2B4. Proc. Natl. Acad. Sci. USA
N. Saldou, K. Jarnagin et al. (1995). Cloning and 93, 4644^648.
functional expression of the cDNA encoding rat 328. Kuo, C.L., G.M. Raner, A.D. Vaz, and M.J. Coon
lanosterol 14a-demethylase. Gene 161, 243-248. (1999). Discrete species of activated oxygen yield
318. Cabello-Hurtado, E, M. Taton, N. Forthofifer, R. different cytochrome P450 heme adducts from
Kahn, S. Bak, A. Rahier et al. (1999). Optimized aldehydes. Biochemistry 3S, 10511-10518.
expression and catalytic properties of a wheat 329. Raner, G.M., J.A. Hatchell, M.U Dixon, T.L. Joy,
obtusifoliol 14a-demethylase (CYP51) expressed in A.E. Haddy, and E.R. Johnston (2002). Regio-
yeast. Complementation of erg 11D yeast mutants by selective peroxo-dependent heme alkylation in
plant CYP51. Eur. J. Biochem. 262, 435^H6. P450BM3-F87G by aromatic aldehydes: Effects
319. Bellamine, A., A.T. Mangla, WD. Nes, and M.R. of alkylation on catalysis. Biochemistry 41,
Waterman (1999). Characterization and catalytic 9601-9610.
properties of the sterol 14a-demethylase from 330. Reed, J.R., D. Vanderwel, S. Choi, G. Pomonis,
Mycobacterium tuberculosis. Proc. Natl. Acad. R.C Reitz, and G.J. Blomquist (1994). Unusual
Sci. USA 96, 8937-8942. mechanism of hydrocarbon formation in the
320. Lamb, D.C, K. Fowler, T. Kieser, N. Manning, housefly: Cytochrome P450 converts aldehyde to
L.M. Podust, M.R. Waterman et al. (2002). Sterol the sex pheromone component (Z)-9-tricosene and
14a-demethylase activity in Streptomyces coeli- CO2. Proc. Natl. Acad. Sci. USA 91, 10000-10004.
colorA3(2) is associated with an unusual member 331. Reed, J.R., DR. Quilici, G.J. Blomquist, and
of the CYP51 gene family. Biochem. J. 364, R.C. Reitz (1995). Proposed mechanism for the
555-562. cytochrome P450-catalyzed conversion of aldehy-
321. Jackson, C.J., D.C. Lamb, T.H. Marczylo, A.G.S. des to hydrocarbons in the house fly, Musca
Warrilow, N.J. Manning, D.J. Lowe et al. (2002). A domestica. Biochemistry 34, 16221-16227.
novel sterol 14a-Demethylase/Ferredoxin fusion 332. Adam, W, R. Curci, M.E. Gonzalez Nunez, and
protein (MCCYP51FX) from Methylococcus capsu- R. Mello (1991). Thermally and photochemically
latus represents a new class of the cytochrome P450 initiated radical chain decomposition of ketone-
superfamily J. Biol. Chem. 277, 46959^6965. free methyl(trifluoromethyl)dioxirane. J. Am.
322. Lamb, D.C, D.E. Kelly, and S.L. Kelly (1998). Chem. Soc. 113,7654-7658.
Molecular diversity of sterol 14a-demethylase 333. Mpuru, S., J.R. Reed, R.C. Reitz, and G.J.
substrates in plants, fungi and humans. FEBSLett. Blomquist (1996). Mechanism of hydrocarbon
425, 263-265. biosynthesis from aldehyde in selected insect
323. Roberts, E.S., A.D.N. Vaz, and M.J. Coon (1991). species: Requirement for O2 and NADPH and car-
Catalysis by cytochrome P-450 of an oxidative bonyl group released as CO2. Insect Biochem.
reaction in xenobiotic aldehyde metabolism: Mol. Biol. 26, 203-208.
Deformylation with olefin formation. Proc. Natl. 334. Spiteller, D., A. Jux, J. Piel, and W Boland (2002).
Acad. Sci. USA 88, 8963-8966. Feeding of [5,5-^H2]-l-desoxy-D-xylulose and
324. Vaz, A.D.N., E.A. Roberts, and M.J. Coon (1991). [4,4,6,6,6-^H5]-mevalolactone to a geosmin-
Olefin formation in the oxidative deformylation of producing Streptomyces sp. and Fossombronia
aldehydes by cytochrome P-450. Mechanistic pusilla. Phytochemistry 61, 827-834.
Substrate Oxidation by Cytochrome P450 Enzymes 245

335. Yamamoto, H., N. Katano, A. Ooi, and K. Inoue 346. Komuro, M., T. Higuchi, and M. Hirobe (1995).
(2000). Secologanin synthase which catalyzes the Application of chemical cytochrome P-450 model
oxidative cleavage of loganin into secologanin is a systems to studies on drug metabolism-VIII.
cytochrome P450. Phytochemistry 53, 7-12. Novel metabolism of carboxylic acids via oxida-
336. Irmler, S., G. Schroder, B. St-Pierre, N.P. Crouch, tive decarboxylation. Bioorg. Med. Chem. 3,
M. Hotze, J. Schmidt et al. (2000). Indole alkaloid 55-65.
biosynthesis in Cathamnthus mseus: New enzyme 347. White, R.E., S.G. Sligar, and M.J. Coon (1980).
activities and identification of cytochrome P450 Evidence for a homolytic mechanism of peroxide
CYP72A1 as secologanin synthase. Plant J. 24, oxygen-oxygen bond cleavage during substrate
797-804. hydroxylation by cytochrome P-450. J. Biol.
337. Udwary, D.W., L.K. Casillas, and C.A. Townsend Chem. 255, 11108-11111.
(2002). Synthesis of 11-hydroxyl 0-methylsterig- 348. Hashim, M.F., T Hakamatsuka, Y Ebizuka, and
matocystin and the role of a cytochrome P-450 in U. Sankawa (1990). Reaction mechanism of oxida-
the final step of aflatoxin biosynthesis. J. Am. tive rearrangement of flavanone in isoflavone
Chem. Soc. 124, 5294-5303. biosynthesis. FEBS Lett. 271, 219-222.
338. Prieto, R. and C.P Woloshuk (1997). ordl, an 349. Akashi, T, T Aoki, and S.-I. Ayabe (1999).
oxidoreductase gene responsible for conversion Cloning and functional expression of a
of 0-methylsterigmatocystin to aflatoxin in cytochrome P450 cDNA encoding 2-hydroxy-
Aspergillus flavus. Appl. Environ. Microbiol. 63, isoflavanone synthase involved in biosynthesis
1661-1666. of the isoflavonoid skeleton in licorice. Plant
339. Coolbaugh, R.C. (1997). Cytochrome P450- Physiol. 121, 821-828.
dependent steps in gibberellin biosynthesis. Proc. 350. Hakamatsuka, T, M.F. Hashim, Y Ebizuka, and
Plant Growth Regulator Soc. Am. 24, 10-14. U. Sankawa (1991). P-450-dependent oxidative
340. Helliwell, C.A., W.J. Peacock, and E.S. Dennis rearrangement in isoflavone biosynthesis:
(2002). Isolation and functional characterization Reconstitution of P-450 and NADPH:P-450
of cytochrome P450s in gibberellin biosynthesis reductase. Tetrahedron 47, 5969-5978.
pathway. Meth. Enzymol. 357, 381-388. 351. Sawada, Y, K. Kinoshita, T Akashi, T Aoki, and
341. Helliwell, C.A., PM. Chandler, A. Poole, E.S. S.-I. Ayabe (2002). Key amino acid residues
Dennis, and W.J. Peacock (2001). The CYP88A required for aryl migration catalysed by the
cytochrome P450, e«^kaurenoic acid oxidase, cat- cytochrome P450 2-hydroxyisoflavanone syn-
alyzes three steps of the gibberellin biosynthesis thase. Ha«^/. 31, 555-564.
pathway. Proc. Natl. Acad. Sci. USA 98,2065-2070. 352. Yin, W, G.A. Doss, R.A. Steams, A.G.
342. Rojas, M.C., P Hedden, P Gaskin, and B. Chaudhary, C.E. Hop, R.B.Franklin et al. (2003).
Tudzynki (2001). The P450-1 gene of Gibberella A novel P450-catalyzed transformation of the
fujikuroi encodes a multifunctional enzyme in 2,2,6,6-tetramethyl piperidine moiety to a 2,2-
gibberellin biosynthesis. Proc. Natl. Acad. Sci. dimethyl pyrrolidine in human liver microsomes:
USA 98, 5838-5843. Characterization by high resolution quadrupole-
343. Fukuda, H., T. Fujii, E. Sukita, M. Tazaki, S. time-of-flight mass spectrometry and ^H-NMR.
Nagahama, and T Ogawa (1994). Reconstitution DrugMetab. Dispos. 31, 215-223.
of the isobutene-forming reaction catalyzed by 353. Boucher, J.-L., M. Delaforge, and D. Mansuy
cytochrome P450 and P450 reductase from (1994). Dehydration of alkyl- and arylaldoximes
Rhodotorula minuta: Decarboxylation with the as a new cytochrome P450-catalyzed reaction:
formation of isobutene. Biochem. Biophys. Res. Mechanism and stereochemical characteristics.
Commun. 201, 516-522. Biochemistry 33, 7811-7818.
344. Shimaya, C. and T. Fujii (2000). Cytochrome 354. Gupta, O.D., R.L. Kirchmeier, and J.M. Shreeve
P450rm of Rhodotorula functions in the P-ketoad- (1990). Reactions of trifluoroamine oxide: A route
ipate pathway for dissimilation of L-phenylalanine. to acyclic and cyclic fluoroamines and N-
J. Biosci. Bioeng. 90, 465^67. nitrosoamines. J ^m. Chem. Soc. 112, 2383-2386.
345. Fukuda, H., K. Nakamura, E. Sukita, T. Ogawa, 355. Tudzynski, B., M.C. Rojas, P Gaskin, and P
and T Fujii (1996). Cytochrome P450rm fi-om Hedden (2002). The gibberellin 20-oxidase of
Rhodotorula minuta catalyzes 4-hydroxylation of Gibberella fujikuroi is a multifunctional monooxy-
benzoate. J. Biochem. 119, 314-318. genase. J. Biol. Chem. 277, 21246-21253.
7
Inhibition of Cytochrome P450 Enzymes
Maria Almira Correia and Paul R. Ortiz de Monteflano

1. Introduction of P450 inhibitors are available in various


reviews"^^^. This chapter focuses on the mecha-
Three steps in the catalytic cycle of nisms of inactivation; thus, most of the chapter is
cytochrome P450 (P450, CYP; see Chapters 5 and devoted to the discussion of agents that require
6) are particularly vulnerable to inhibition: (a) the P450 catalysis to fiilfill their inhibitory potential.
binding of substrates, (b) the binding of molecular The mechanisms of reversible competitive and
oxygen subsequent to the first electron transfer, noncompetitive inhibitors, despite their practical
and (c) the catalytic step in which the substrate is importance, are relatively straightforward and are
actually oxidized. Only inhibitors that act at one of discussed more briefly.
these three steps will be considered in this chapter.
Inhibitors that act at other steps in the catalytic
cycle, such as agents that interfere with the 2. Reversible Inhibitors
electron supply to the hemoprotein by accepting
electrons directly from P450 reductase^"^, are not Reversible inhibitors compete with substrates
discussed here. for occupancy of the active site and include agents
P450 inhibitors can be divided into three that (a) bind to hydrophobic regions of the active
mechanistically distinct classes: Agents that site, (b) coordinate to the heme iron atom, or
(a) bind reversibly, (b) form quasi-irreversible (c) enter into specific hydrogen bonding or ionic
complexes with the heme iron atom, and (c) bind interactions with active-site residues"*"^^. The first
irreversibly to the protein or the heme moiety, or mechanism, in which the inhibitor simply competes
accelerate the degradation and/or oxidative frag- for binding to lipophilic domains of the active site,
mentation of the prosthetic heme. Agents that is often responsible for the inhibition observed
interfere in the catalytic cycle prior to the actual when two substrates compete for oxidation by a sin-
oxidative event are largely reversible competitive gle P450 isoform. A clear example of such an inter-
or noncompetitive inhibitors. Those that act dur- action is provided by the mutual in vitro and in vivo
ing or subsequent to the oxygen transfer step are inhibition of benzene and toluene metabolism^^.
generally irreversible or quasi-irreversible inhibitors This form of inhibition, which is optimal when the
and often fall into the category of mechanism- inhibitory compound is bound tightly but is a poor
based (or suicide) inactivators. Extensive lists substrate, is usually not highly effective but can

Maria Almira Correia • Department of Cellular and Molecular Pharmacology, Department of Pharmaceutical
Chemistry, Department of Biopharmaceutical Sciences and the Liver Center, University of California,
San Francisco, CA. Paul R. Ortiz de Montellano • Department of Pharmaceutical Chemistry, and
Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA.

Cytochrome P450: Structure, Mechanism, and Biochemistry, 3e, edited by Paul R. Ortiz de Montellano
Kluwer Academic / Plenum Publishers, New York, 2005.

247
248 M.A. Correia and P.R. Ortiz de Montellano

cause physiologically relevant metabolic changes 2.2. Coordination to


and clinically significant interactions^"*. Ferrous Heme
In its simplest form, inhibition through coordi-
2.1. Coordination to nation to the heme iron is exemplified by carbon
Ferric Heme monoxide, a neutral ligand that binds exclusively
The binding of a strong sixth ligand to the pen- to the ferrous (reduced) form of P450. The
tacoordinated heme iron atom of a P450 enzyme, binding of carbon monoxide involves donation of
or displacement of a weak ligand in a P450 hexa- electrons from the carbon to the iron through a
coordinated state by a strong ligand, causes a shift a-bond as well as back-donation of electrons from
of the iron from the high- to the low-spin state. the occupied ferrous iron d-orbitals to the empty
This shift is characterized by a "type H" binding antibonding ir-orbitals of the ligand^^. P450
spectrum with a Soret maximum at 425-435 nm enzymes (P450, CYP) are so named because their
and a trough at 390-405 nm^^~^^. The change in carbon monoxide complexes have spectroscopic
the redox potential of the enzyme associated with absorption maxima at approximately 450 nm^^.
this spin state change makes its reduction by P450 Early studies with model ferroporphyrins, which
reductase more difficult (see Chapter 5)^^' ^^. This indicated that only those with a thiolate ligand
change in reduction potential, as much as physical trans to the carbon monoxide yielded the 450-nm
occupation of the sixth coordination site, is absorption, provided key evidence for the pres-
responsible for the inhibition associated with the ence of a thiolate fifth ligand in P450.^' Inhibition
binding of strong iron ligands. by carbon monoxide is a signature of P450-
Cyanide and other ionic ligands bind preferen- catalyzed processes, although the sensitivities of
tially, albeit weakly, to the ferric state of a P450 different P450 isoforms to carbon monoxide dif-
enzyme^^' ^^. The three positive charges of the iron fer^^ and a few P450-catalyzed reactions are
are matched in the ferric hemoprotein by the three resistant to inhibition by carbon monoxide^^"^^. In
negative charges of its permanent ligands (i.e., the particular, the sensitivity of aromatase^^' ^^ and
two porphyrin nitrogens and the thiolate ion), but P450g(.^^^^ to inhibition by carbon monoxide
in the reduced state there is a charge imbalance of decreases drastically as the enzymes traverse the
three ligand negative charges but only two ferrous conformational and ligand states inherent in their
iron positive charges. The cyanide ion, a nega- multistep catalytic sequences. Among the human
tively charged species, binds more readily to the liver P450 isoforms, the susceptibility of different
neutral (ferric) than the negative (ferrous) families to carbon monoxide inhibition appears to
enzyme. Indeed, cyanide binds more weakly to decrease in the order 2D > 2 C > 3A^^.
ferric P450 than to ferric myoglobin because the
P450 thiolate ligand places a higher electron den-
sity on the iron than does the imidazole ligand of 2.3. Heme Coordination and
myoglobin^^. The chelation of ionic ligands is dis- Lipophilic Binding
favored, in addition, by the lipophilic nature of the
P450 active site^^. Agents that simultaneously bind to lipophilic
P450 enzymes are inhibited by nitric oxide regions of the active site and to the heme iron atom
(NO), a molecule of great interest because of its (Figure 7.1) are inherently more effective P450
role in diverse physiological and pathological inhibitors than agents that only exploit one of these
processes. Inhibition initially involves reversible binding interactions. The effectiveness of these
coordination of the nitrogen to the iron but a sub- agents as P450 inhibitors is governed both by their
sequent time-dependent, irreversible inactivation hydrophobic character and the strength of the bond
of the enzyme by an undefined mechanism has between their heteroatomic lone pair and the heme
been reported^"^^^. Inhibition by endogenous NO iron. Agents such as alcohols, ethers, ketones, lac-
of P450 enzymes involved in endogenous sub- tones, and other structures in which an oxygen
strate metabolism, including eicosanoid formation atom of the ligand coordinates to the iron, or which
and sterol metabolism, may have physiological act by stabilizing the coordination of the distal
consequences^^' •^^. water ligand, are relatively poorly bound and are
Inhibition of Cytochrome P450 Enzymes 249

MeCON

O^ O

N . ^
N-X^
rCI, - ^ ^ ^ ^ ,CI
Ketoconazole

Metyrapone

Cimetidine
Figure 7.1. Schematic illustration of the binding of an inhibitor to a P450 active site by both coordinating to the
heme-iron atom and interacting with the surrounding protein residues. The structures of three agents that inhibit P450
by binding in this manner are shown.

weak inhibitors^'^' ^^^^. The Soret band of such affinity of the ligand nitrogen electron pair for the
complexes is found at approximately 415 nm^^' ^^. heme iron, (b) the degree to which the intrinsic
In contrast, agents that interact strongly with both affinity of the ligand for the iron is moderated by
the protein and the heme iron atom are often highly steric interactions with substituents on the
effective reversible inhibitors'*"^^. As already noted, inhibitor^^' ^^, (c) the lipophilicity of the nonligat-
the binding of such inhibitors yields a "type IF' ing portion of the inhibitor^^' ^^, and, naturally,
difference spectnmi with a Soret maximum at (d) the congruence between the geometry of
430 nm^^' ^^' ^^^ ^^. The structures of these powerful the inhibitor and that of the active site. The
inhibitors usually incorporate nitrogen-containing synergism that results from binding simultane-
aliphatic or aromatic functions. ously through lipophilic interactions and coordi-
P)nridine, imidazole, and triazole derivatives nation with the heme iron is illustrated by the
have proven particularly useful in P450 inhibitors^. fact that imidazole and benzene individually
Metyrapone (Figure 7.1), one of the first P450 are weak inhibitors, but when coupled together as
inhibitors to be widely employed, first gained in phenylimidazole they produce a powerful
prominence as an inhibitor of 11 p-hydroxylase, inhibitor^^. Optimization of these structural fea-
the enzyme that catalyzes the final step in Cortisol tures has made possible the therapeutic exploita-
biosynthesis"*^. This activity led to its use in the tion of azole compounds such as ketoconazole
diagnosis and treatment of hypercortisolism (Figure 7.1) as antifungal and cancer chemothera-
(Cushing's syndrome) and other hormonal disor- peutic agents^. On the other hand, similar features
ders^^. The determinants of the inhibitory potency in cimetidine are responsible for the drug-drug
of metjo-apone and other nitrogen heterocycles are interactions that result from the inhibition of
valid for most reversible inhibitors: (a) the intrinsic the metabolism of co-administered drugs. This
250 M.A. Correia and P.R. Ortiz de Montellano

inadvertent side effect instigated the search for modify the protein. However, this mechanistic
non-imidazole containing H2-antagonists devoid classification is not rigid, as in the course of its
of this undesirable feature"^^' ^^ Improvement of the P450, metabolism, a compound may simultane-
potency and particularly specificity of metyrapone, ously partition into two or more of these inhibitory
aminoglutethimide, and other classical inhibitors trajectories.
by structural modification continues to be of
therapeutic interest. This consideration has led to
the development and clinical testing of, for exam- 3.1. Covalent Binding to
ple, pyridylaminoglutethimide, CGS16949A {4- the Protein
(5,6,7,8-tetra-hydroimidazo[l,5a]pyridin-5-yl)
Agents that are oxidatively activated and inacti-
benzonitrile}, CGS18320B bis-(/?-cyanophenyl)
vate the enzyme by covalently binding to it include
imidazo-1-yl-methane hemisuccinate and R-76713
(a) diverse sulfur compounds (e.g., carbon disul-
[6-(4-chlorophenyl)lH (1,2,4 triazol-l-yl)-methyl]-
fide^^"^^, parathion^^' ^^, diethyldithiocarbamate^^,
1-methyl-IH-benzotriazole as aromatase inhibitors
isothiocyanates^^, thioureas^ ^ thiophenes^^, tie-
(see Section 5.2)^. Likewise, efforts to improve
nilic acid^^' ^^, and mercaptosteroids^'^"^^ (b) halo-
the properties of ketoconazole have fostered the
genated structures such as chloramphenicoF^'^^,
design and use in antifungal therapy of sterol
A^-monosubstituted dichloroacetamides^^, and N-
14a-demethylase inhibitors such as miconazole,
(2-/?-nitrophenethyl)dichloroacetamide^^, (c) alkyl
fluconazole, saperconazole, and terconazole
and aryl olefins and acetylenes^^"^"^ such as
(see Section 5.3)^.
10-undecynoic acid''^' ^^, 10-dodecynoic acid^"^,
l-ethynylpyrene^^' ^^, 17p-ethynylprogesterone^^'
^^, 17a-ethynylestradiol^^"^^ gestodene^^, 1- and
3. Catalysis-Dependent 2-ethynylnaphthalene^^' ^^' ^^^ ^\ 7-ethynyl-
coumarin (l-ECf^, mifepristone^^' ^^, and seco-
Inhibition
barbital^^, (d) furanocoumarins such as
8-methoxypsoralen (8-MOP, methoxsalen)^^"^^^,
Multiple classes of compounds are now known 6',7'-dihydroxybergamottin (6',7'-DHB)iO7-i09^
that undergo P450-catalyzed activation to reactive and the flirano-pyridine, L-754,394"^~^^^ and
intermediates that irreversibly or quasi-irreversibly (e) compounds such as carbamazepine (CBZ) and
inactivate the enzyme responsible for their activa- tamoxifen that are hydroxylated to catechol
tion. This irreversible inactivation by a catalytically metabolites ••''-" 6. 2-Phenylphenanthridinone,
generated species is superimposed on the which inhibits CYPlAl, is a member of a distinct
reversible inhibition associated with competitive class of mechanism-based inactivating agents that
binding of the parent agent to the ferric enzyme. is thought to bind covalently to the protein^ ^^. The
Mechanism-based^^ ^^ (catalysis-dependent) inac- details of the mechanisms by which a few of these
tivators can be highly enzyme-specific because compounds inactivate P450 remain unclear, but
(a) the compound must first bind reversibly to the much is now known about the mechanisms of acti-
enzyme and must satisfy all the constraints vation of many of these inhibitors and, in some
imposed on normal substrates of the enzyme, (b) it instances, about the sites on the P450 enzymes
must be acceptable as a substrate and thus undergo which they covalently modify.
catalytic activation and, finally, (c) the resulting
reactive intermediate must irreversibly alter the
enzyme and permanently remove it from the 3.1.1. Sulfur and Halogenated
catalytic pool. Four general classes of mechanism-
based irreversible P450 inactivators are known:
Compounds
(a) agents that bind covalently to the protein, Incubation of liver microsomes with
(b) agents that quasi-irreversibly bind to the [^^S]parathion results in radiolabeling of the pro-
prosthetic heme iron atom, (c) agents that alkylate tein but no radiolabeling occurs when the
or arylate the porphyrin framework of the heme, parathion ethyl groups are labeled with ^"^C^^' ^^.
and (d) agents that degrade the prosthetic heme Ninety percent of the ^^S-label, bound covalently
to products that can, in some cases, themselves to the microsomal proteins is immunoprecipitated
Inhibition of Cytochrome P450 Enzymes 251

EtO- 1-0^^ N02' EtO—P—O—/ \ NO2


OEt OEt

P450 Reactive
inactivation sulfur species

EtO- Lo^l NO2


OEt
Figure 7.2. The activation of parathion to species that destroy cytochrome P450 is proposed to involve oxidation
of the thiophosphonate to reactive sulfiir species that bind to protein residues. The reactive species can be envisioned
to be a sulfur atom with six electrons or HSOH.

by anti-P450 antibodies. Approximately 75% of are also inactivated, whereas CYP2E1 is not
the P450 prosthetic heme is degraded to unknown inactivated^ ^^~^^^ While P450 heme destruction
products in incubations with parathion, but unequivocally occurs in vitro in incubations of
~ 4 nmol of radiolabeled sulfur are bound cova- parathion with NADPH-supplemented rat and
lently to the protein for each nanomole of heme human liver microsomes or purified recombinant
chromophore that is lost. The bulk (50%-75%) of P450 enzymes, the in vivo relevance of these find-
the radiolabeled sulfur is removed from the protein ings remains to be established^ ^^. It appears that
by treatment with cyanide or dithiothreitol, which the concentrations required for in vitro P450
suggests that most of the sulfur label is present in destruction are considerably higher than the con-
the form of hydrodisulfides (RSSH), but the centrations that cause death through inhibition of
enzyme is not reactivated by these treatments. The acetylcholinesterase^^^' ^^^.
binding of multiple equivalents of labeled sulfur Tienilic acid (Figure 7.3), a substituted thio-
indicates that catalytic sulfur activation persists phene, is oxidized by CYP2C9 and -2C10 to a
despite covalent attachment of the sulfur to the product that irreversibly inactivates these enzymes
protein until the heme itself is damaged^^ or is [the values for half-maximal inactivation
released from the protein as a consequence of pro- (^j) = 4.3 |xM, the maximal rate constant for
tein damage. The oxidative mechanism in Figure 7.2 inactivation (^.j^^^^) = 0.22min~^ and partition
is suggested by (a) the covalent binding of sulfur, ratio = 11.6]^^' ^^. Tienilic acid is oxidized by
(b) the oxidation of sulfur compounds to these P450 enzymes to 5-hydroxytienilic acid and
S-oxides, and (c) the formation of metabolites in a product that is covalently bound to the P450 pro-
which the sulfur is replaced by an oxygen. More tein. Covalent labeling of the protein is partially
recent studies reveal that parathion competitively prevented by glutathione (GSH) but GSH does not
inhibits certain rat liver P450 enzymes (i.e., protect the enzyme from inactivation or loss of the
CYP2B1 and -2C6) at low concentrations, but at heme chromophore. In the presence of GSH,
higher concentrations inactivates CYP2A1, -2A2, approximately 0.9 equivalents of label are cova-
-2C11, -3A2, and -3A4 (but not CYP2B1 or -2C6) lently bound to the protein before catalytic activity
in vitro but not in v/vo^^^~^^^ Analogous in vitro is suppressed. The results are explained by the for-
studies with human liver microsomes reveal that mation of a thiophene sulfoxide that reacts with
CYP3A4 is the principal isoform that is inacti- water to give the isolated metabolite or with a pro-
vated, CYP2C9 and -1A2 are minor forms that tein nucleophile to inactivate the enzyme ^^^. The
252 M.A. Correia and P.R. Ortiz de Montellano

O^ ^COoH

Protein-X-
C^ "CO2H

Ticlopidine Clopidogrel CO2CH3

Figure 7.3. The mechanism proposed for P450 bioactivation of the thiophene in tienihc acid to a reactive species
that binds covalently to the protein (the protein nucleophile is denoted by Protein-XH). The structures of two other
thiophene-containing drugs, ticlopidine and clopidogrel, that might be similarly activated are also shown.

CYP2C9 site that is modified by tienilic acid has CYP2C19 inactivation suggests that it occurs
not been identified, but HPLC/electrospray mass within the active site of the enzyme. The inactiva-
spectrometric (ESI-MS) analysis of the tienilic tion follows classical mechanism-based inactiva-
acid-modified and native proteins reveals the pres- tion criteria with the following kinetic parameters:
ence of mono- and diadducted CYP2C9 species './2n,ax = 3.4 min, ^,„^„ = 3.2 X lO"' s"', K, =
with molecular masses of 55,923 ±1.1 and 56,273 97 JULM, k-^^^JK^ = 37 L mol ^ s ^ and partition
± 4.4 Da, respectively, w^hich corresponds to mass ratio = 26'^^. In vitro inhibition studies with fixnc-
shifts of 344.4 ± 1.1 and 694 ± 4.2 Da^^s tionally competent yeast-expressed recombinant
Diadduct formation is abolished by the inclusion human liver CYP1A2, -2C8, -2C9, -2C18, -2C19,
of GSH (10 mM), suggesting that the diadduct -2D6, and -3A4 yielded IC50 (JULM) values of 75 ±
may result from modification of a second residue 15, 100 ± 20, >200, 100 ± 15, 10 ± 5, 10 ± 3,
outside the active site. The mass shift in the and 50 ± 10, respectively ^^'*. These findings estab-
CYP2C9 monoadduct is consistent with the bind- lish that ticlopidine eff'iciently inhibits CYP2C19
ing of a monohydroxylated tienilic acid obtained and -2D6. Furthermore, within the human liver
either by thiophene ring oxidation or formation of CYP2C subfamily, ticlopidine is a relatively selec-
a sulfoxide that resists dehydration ^^^. tive mechanism-based inactivator of CYP2C19^^'^,
Ticlopidine (Figure 7.3), another substituted thio- although it is not quite as potent as tienilic acid is in
phene that is clinically utilized as an anti-platelet CYP2C9 inactivation. These findings rationalize
aggregation agent, is reportedly the first selective the clinical observation that ticlopidine inhibits
mechanism-based inactivator of CYP2C19^2'^. the clearance of phenytoin, another CYP2C19
This inactivation apparently occurs during S- substrate *^^~^^^. However, recent studies with
oxidation of the thiophene moiety. The failure of an extensive battery of recombinant human
GSH (5 mM) to inhibit ticlopidine-mediated P450 enzymes in Supersomes^^ indicate that
Inhibition of Cytochrome P450 Enzymes 253

O- -v^ - v / -SCOCH3 SOH


Spironolactone

Figure 7.4. The activation of spironolactone to products that inactivate sterol 17a-hydroxylase by covalent
attachment to the protein, and hepatic P450 3 A by degradation of the heme group, occurs during oxidation of the free
thiol group unmasked by the action of a thiolesterase.

CYP2B6 is even more effectively inactivated than sulfhydryl radical, whereas protein modification
CYP2C19 not only by ticlopidine, but also by involves reaction of the sulfenic acid metabolite
clopidogrel, a related thienopyridine antiplatelet with amino acid side chains (Figure 7.4).
aggregating agent^^^. These observations suggest Three other sulfur-containing drugs are note-
that clinically relevant drug-nirug interactions worthy in that they are also bioactivated by human
with these two agents may occur with substrates liver CYP3A4 and result in inactivation of the
of not only CYP2C19 but also CYP2B6 and -2D6. enzyme. The first is the oral antidiabetic drug
The aldosterone antagonist spironolactone troglitazone (Figure 7.5)^^^, which was recently
(Figure 7.4), which is used as a diuretic and anti- withdrawn from the US market due to its associa-
hypertensive^^^, inactivates P450 enzymes in both tion with clinically severe hepatotoxicity^^^' ^^^. As
the liver and steroidogenic tissues^'*"^^ including revealed by structural analyses of its GSH-adducts,
the adrenal steroid ITa-hydroxylase^"^' ^^' ^^' ^^ and this 2,4-thiazolidinedione is activated by two dis-
members of the hepatic CYP3A and CYP2C sub- tinct metabolic routes, one involving oxidation of
families^^' ^^. Spironolactone-mediated hepatic the substituted chromane ring system to a reactive
P450 inactivation requires hydrolysis of the o-quinone methide, and the other involving a
thioester function to give the free thiol that^^ is novel oxidative cleavage of the thiazolidinedione
subsequently oxidized to a species that reacts with ring to highly electrophilic a-ketoisocyanate and
the protein and/or the heme^^. Hepatic P450 sulfenic acid intermediates^^^
inactivation involves fragmentation of the pros- The second drug is the selective estrogen recep-
thetic heme to products that bind covalently to the tor modulating drug, raloxifene (Figure 7.5)^^"^,
protein (see Section 3.4)^^' ^^. In contrast, inacti- which is used for the treatment of postmenopausal
vation of the adrenal P450 appears to result from osteoporosis. The in vitro bioactivation of ralox-
covalent binding of the thiosteroid itself to the ifene by human liver microsomal CYP3A4 results
protein^"^' ^^' ^^' ^^. A role for thiol oxidation in in mechanism-based inactivation of this enzyme.
enzyme inactivation is supported by the fact that The inactivation is attenuated by the CYP3A4-
rat hepatic microsomes enriched in CYP3A selective inhibitor ketoconazole (10 |ULM), is
enzymes oxidize the thiol group (-SH) to the quenched minimally (-15%) by GSH (5 mM),
sulfinic (-SO2H), and sulfonic (-SO3H) acids^^ and is characterized by K^, ^inacf ^^^ partition
and give rise to a disulfide adduct with GSH^^ ratio values of 9.9 JULM, 0.16 min~^ and 1.8,
However, formation of the disulfide adduct with respectively^^"^. Indeed, raloxifene, albeit a
GSH is catalyzed, at least in part, by a flavin slower CYP3A4 inactivator, is more potent than
monooxygenase^^ Two reactive intermediates that gestodene, which exhibits K^, A:-^^^^ and partition
can arise by thiol oxidation are the sulfhydryl ratio values of 46 ixM, 0.39 min~^ and 9, respec-
radical (-S') and the sulfenic acid (-SOH), either tively^^. CYP2D6 is also capable of bioactivating
or both of which could be involved in P450 inac- raloxifene at half the rate of CYP3A4, but the
tivation. One possibility is that fragmentation of CYP2D6 contribution to human liver microsomal
the heme results from its reaction with the raloxifene activation is minimal as judged by
254 M.A. Correia and P.R. Ortiz de Montellano

NH
Troglitazone

HO

Raloxifene

HO
Ritonavir

Figure 7.5. Structures of sulfur-containing compounds that inactivate P450 enzymes by mechanisms that have not
been elucidated but may involve oxidation of a sulfur atom.

the failure of quinidine to significantly inhibit to act through irreversible protein modifica-
the process at concentrations known to inhibit tion^^"^^. Not only does the binding of ['"^C]
CYP2D6^^^. The mechanism of this inactivation chloramphenicol to the apoprotein correlate with
has not yet been explored, but could involve the loss of CYP2B1-dependent ethoxycoumarin
oxidative activation of the phenolic or thiophene 0-deethylase activity, but proteolytic digestion of
functions. the inactivated P450 has been shown to yield a
Ritonavir (Figure 7.5), an inhibitor of the single ['''C]-modified amino acid^^"^^. Lysine and
HIV-1 protease, is another example of a potent, the chloramphenicol fragment in Figure 7.6 were
sulfur-based inactivator of CYP3A4/CYP3A5, released by hydrolysis of the modified amino acid.
although it is also a CYP2D6 inhibitor^^^'^^. These results indicate that chloramphenicol is
Its inhibitory potency depends on the presence converted to an oxamyl chloride intermediate
of both the 5-thiazolyl and 2-(l-methylethyl) that either acylates a critical lysine, probably at
thiazolyl groups. It has been proposed that it is the protein surface, or is hydrolyzed to the oxamic
oxidized by CYP3A to a chemically reactive frag- acid. Acylation of the lysine apparently inhibits
ment containing the 2-(l-methylethyl)thiazolyl electron transfer from P450 reductase to the
group that causes enzyme inactivation'^^. CYP2B1 heme because the inactivated enzyme
Other noteworthy sulfur-containing mecha- still catalyzes ethoxycoumarin O-deethylation
nism-based inactivators include thiocyanates such in the presence of cumene hydroperoxide
as phenethyl isothiocyanate'"^^"'^^, benzylisothio- or iodosobenzene'^^. Furthermore, the unimpaired
cyanate^ ^er^butylisothiocyanate' ^^; diallyl ethoxycoumarin 0-deethylation supported by
sulfide, and diallylsulfone derivatives'"^^ ''^^, 1,2- activated oxygen donors suggests that chloram-
dithiole-3-thione'^^, oltipraz, and its deriva- phenicol is not covalently bound within the sub-
tives'''^, sulforaphane'^^, and thiosteroids'''^. strate binding-site.
Chloramphenicol was among the first chlori- The P450 isoform selectivity of inactivation
nated mechanism-based P450 inactivators shown by chloramphenicol and several of its analogs
Inhibition of Cytochrome P450 Enzymes 255

OH H
N^^CCl20H ,N.,^.COCl
T — • T
N02^ "^ HO o o
Chloramphenicol [Acylated protein] -<- H20
Proteolysis

OH H C) H
.N CO2H

NO2
T
o
H
CHC12

o (2-/7-Bromophenethyl)dichloroacetaniide
Br
Figure 7.6. The inactivation of P450 by chloramphenicol involves oxidation of the dichloromethyl group to an
acyl chloride that reacts with an amino group of the protein. A potent inactivating agent related to chloramphenicol
is (2-p-bromophenethyl)dichloroacetamide.

has been reported^^^. Using four regiospecific also been probed for their selectivity of P450 inac-
androstenedione hydroxylases as functional mark- tivation using regiospecific and/or stereoselective
ers, chloramphenicol was found to inactivate rat progesterone or androstenedione hydroxylases as
liver microsomal CYP2B1 > -3A > -2C11 > functional probes^^^' ^^^. The findings reveal that
-2Ali^o. The P450 selectivity of the chloram- 21,21-dichloropregnenolone and 21,21-dichloro-
phenicol analogs as mechanism-based inactivators progesterone are comparably effective in the inac-
was determined by at the least three structural tivation of rat liver microsomal CYP3 A enzymes,
features: (a) the number of halogen atoms, (b) the with t . values of «^0.1 min~ ^ for both substrate
inact
presence of ap^fra-nitro group on the phenyl ring, probes, while 21,21-dichloroprogesterone even
and (c) substitutions on the ethyl side chain. Thus more efficiently inactivated CYP2C6 (A:-^^^^^ =
the analog A/-(2-phenethyl)dichloroacetamide —0.2 min"^)^^^ 21,21-Dichloropregnenolone was
reversibly inhibited but did not inactivate CYP3A also a very efficient mechanism-based inactivator
enzymes, in contrast to A^-(2-p-nitrophenethyl)- of rabbit liver CYP2C5 but not of rabbit adrenal
and 7V-(l,2-diphenethyl)dichloroacetamide, both (-YP21152 On the other hand, CYP21 was rapidly
of which rapidly did so^^^. The incorporation inactivated by 21,21 -dichloroprogesterone ^ ^^.
of a para-nitro or -bromo substituent on the Thus, replacement of a methyl group that is nor-
phenyl ring, or a second phenyl at the 1- or 2- mally oxidized by a P450 enzyme with a
position of the phenethyl side chain, yielded dichloromethyl moiety may be a valuable strategy
compounds that selectively inactivated CYP2B1 in the design of specific P450 inhibitors.
over CYP2C11, -2C6, or -2Al7^. Thus, N-(2-p-
bromophenethyl)-dichloroacetamide (Figure 7.6)
3.1.2. Olefins and Acetylenes
and A^-(2-/?-nitrophenethyl)dichloroacetamide were
the two most effective and selective CYP2B1 inac- As discussed in detail below (Section 3.2), com-
tivators both in vitro and in vivo^^. pounds containing an olefinic bond, ranging from
21 -Chloropregnenolone, 21,21 -dichloropreg- compounds as simple as ethylene to more complex
nenolone, and 21,21-dichloroprogesterone have structures such as allylisopropylacetamide (ALA),
256 M.A. Correia and P.R. Ortiz de Montellano

allylisopropylcarbamide (sedormid), aprobarbi- narrowed down the region of secobarbital modifi-


tal, allobarbital, and secobarbital, can inactivate cation to residues G299"^304^^^' Although the pre-
P450 enzymes by A^-alkylating the porphyrin group cise residue that is modified remains to be
of the prosthetic herne^^^"^^^. However, studies with identified, these findings are consistent with mod-
secobarbital show that it completely inactivates ification of the protein within the active site. In
CYP2B1 but only causes partial loss of the heme view of this, it is not surprising that specific muta-
chromophore^^' ^^^' ^^^. Isolation of the A/-alkylated tions in the CYP2B1 putative substrate-recognition
porphyrins (see Section 3.3.1) and of the modified sites (SRS) 2, 4, 5, and 6, but not SRS-1, attenuate
CYP2B1 protein reveal that the compound par- secobarbital-mediated inactivation. The SRS-
titions between heme A/-alkylation, CYP2B1 pro- mutations T302S and V363L markedly reduced
tein modification, and formation of an epoxide CYP2B1 heme-modification and a V367A
metabolite in a ratio of 0.8:0.2:59, respectively^''' (SRS-5) mutation most markedly impaired protein
157,158 Yhe in situ presence of the heme adduct in modification^ ^^.
the CYP2B1 active site is spectrally confirmed by Terminal acetylenes, like terminal olefins,
its typical —445 nm absorption maximum, ^^^ alkylate the P450 prosthetic heme (see Section
a feature characteristic of iron-complexed N- 3.3.2), but compounds such as 10-undecynoic
modified porphyrins ^^^. The A/-modified por- acid, 1-ethynylpyrene, 17p-ethynylprogesterone,
phyrins have been isolated both as the parent 17a-ethynylestradiol (EE), and 9- and 2-ethynyl-
adducts and as the corresponding dimethyl esters naphthalene have been shown to inactivate P450s
and identified by mass spectrometry (MH^ primarily by binding covalently to the protein with
816.9 and 845.8 Da, respectively) as adducts of only partial loss of the heme group^^"^^' ^^' ^^
protoporphyrin IX and hydroxysecobarbital Thus, near stoichiometric binding of 2-ethynyl-
(Figure l.iy^l The CYP2B1 peptide modified by naphthalene and 1-ethynylpyrene to CYPlAl and
the drug has also been isolated and shown to be -1A2, of EE to CYP3A4, and of 10-undecynoic
comprised of residues 277-323, residues that by acid to rat liver CYP4A1 (w-hydroxylase) has
sequence analogy to P450^^^ correspond to the been observed^^"^^' ^'^'^K The isolation of the
distal I helix^^^' i60-i63 similar structural analysis terminal acid metabolites from the incubations
after ftirther digestion of this CYP2B1 peptide has of 10-undecynoic acid (Figure 7.8) and

CYP2B1

CYP2B1 protein

Figure 7.7. The oxidation of secobarbital by CYP2B1 results in both alkylation of one of the residues between
Gly299 and Ser304 and A/-alkylation of the heme group as well as the generation of an epoxide.
Inhibition of Cytochrome P450 Enzymes 257

RCH2CO2H

IH2O
O
Protein-XH
R-
H*
> -
O
RCH X X-Protein

"CO2H

2-Ethynylnaphthalene 10-Undecynoic acid

Figure 7.8. The oxidation of terminal acetylenes, and even some internal acetylenes, results in the formation of
ketene intermediates that react with water to give the carboxylic acids (see Chapter 6). It appears that the ketenes
also react with active-site residues, inactivating the P450 enzyme that forms them. The hydrogen that undergoes a
1,2-migration during the oxidation reaction is indicated by a star. The structures of 2-ethynylnaphthalene and 10-
undecynoic acid, both of which inactivate P450 enzymes, at least in part by this mechanism, are shown.

1-ethynylpyrene strongly supports a mechanism site as a determinant of the inactivation mecha-


in which oxygen transfer from the P450 heme to nism. Both of these terminal aryl acetylenes yield
the terminal carbon of the triple bond triggers ketene metabolites but only that from 2-ethynyl-
migration of the terminal hydrogen to the adjacent naphthalene acylates the CYP2B1 protein. Protein
carbon (Figure 7.8). The reactive ketene generated acylation by 2-ethynylnaphthalene demonstrates
by this hydrogen shift mechanism, which is sup- that the failure of phenylacetylene to acylate the
ported by deuterium labeling experiments, either CYP2B1 protein is not due to the absence of
acylates the protein or is hydrolyzed to the car- appropriate nucleophilic residues. Furthermore,
boxylic acid metabolite^^. The corresponding the formation of phenylacetic acid in the oxidation
ketene metabolites have been similarly invoked in of phenylacetylene confirms that the ketene is
the acylation of bovine adrenal CYP21 by 17p- formed. The differential inactivation of CYP2B1
ethynylprogesterone^^' ^^ and of CYP1A2, -2B1, by 2-ethynylnaphthalene and phenylacetylene
and -2B4 by 2-ethynylnaphthalene (Figure 7.8)^^' suggests, in fact, that: (a) 2-ethynylnaphthalene,
^2' 93 In accord with this postulate, CYP2B1 oxi- unlike phenylacetylene, is bound in a manner that
dizes 2-ethynylnapthalene to 2-(2-napthyl)acetic prevents delivery of the ferryl oxygen to the
acid, presumably via the same ketene intermediate internal carbon and (b) heme alkylation by pheny-
as is postulated to modify the protein. The mecha- lacetylene is sufficiently efficient relative to pro-
nism-based inactivation of CYP2B1 by 2-ethynyl- tein acylation by the phenylketene metabolite that
naphthalene is characterized by a Xj = 0.08 JULM, the enzyme is inactivated before protein acylation
^inact ~ ^'^^ min~^, and a partition ratio of approx- becomes significant. These differences presum-
imately 4-5 moles of acid formed per inactivation ably stem from differential binding affinities
event^^. and/or orientations of the two agents within the
In contrast, CYP2B1-catalyzed addition of the CYP2B1 active site.
activated oxygen to the internal rather than termi- Peptide mapping and amino acid sequence
nal triple bond carbon of phenylacetylene results analysis of radiolabeled peptides indicate that
in heme iV-alkylation rather than protein acylation 2-ethynylnaphthalene binds to amino acid residues
(see below)^^' ^^^. Predominant inactivation of 67-78 and 175-184, respectively, of rat and rab-
CYP2B1 by phenylacetylene via heme alkyla- bit CYPlA2^o. However, the residue that
tion^^"^ and by 2-ethynylnaphthalene via protein is actually modified and the precise nature of
acylation^^' ^^' ^^ sheds some light on the influence the covalent link to the inhibitor have escaped
exerted by the fit of the inhibitor within the active definition due to the instability of the adducts.
258 M.A. Correia and P.R. Ortiz de Montellano

Alignment of the modified peptides with the native and 7-EC-inactivated CYP2B1 yielded
sequence of P450^^^ (CYPlOl) suggests that the masses of 55,899 ± 1 Da and 56,084 ± 3 Da,
CYP1A2 peptides correspond to hehces A and respectively, for the two proteins^"^. This corre-
D of P450^^^ (see Chapter 3). Thus, the rat sponds to a mass difference of 185 Da (0.005%
CYP1A2 peptide may include residues from the variability in mass assignment), and indicates that
substrate-binding regions^^^"^^^. The 2-ethynyl- the entire 7-EC molecule together with an oxygen
naphthalene-modified peptides from CYP2B1 and atom was covalently bound to CYP2B1 in a 1:1
CYP2B4 have also been characterized and pro- stoichiometry. The precedents set by other ary-
posed to be adducts of 2-naphthylacetic acid with lacetylenes suggest that the protein adduct
a peptide residue^^' ^^. Digestion of CYP2B1 after involves addition of an active-site nucleophilic
its oxidation of radiolabeled 2-ethynylnapthalene residue to the activated acetylene. Two mecha-
yields a radiolabeled peptide (ISLLSLFFAGT- nisms have been proposed for the acylation: One
ETSSTTLRYGFLLM) that includes residues that is mediated by a ketene metabolite and a sec-
290-314 of the protein. An analogous peptide ond involving attack on a putative oxirene inter-
(E273-M3J4) is obtained with CYP2B4. The two mediate^"*. In view of the fact that oxirenes are
modified peptides correspond in sequence to the only hypothetical species due their practically bar-
highly conserved distal I helix of P450^^^ rierless conversion to ketenes, the protein modifi-
(CYPlOl) that contacts both the substrate and the cation is almost certainly mediated by the ketene.
heme group^^^~^^^. The specific residue modified 17a-Ethynylestradiol (EE) (Figure 7.9) has
in each peptide has not been identified but several long been known to inactivate rat and human liver
nucleophilic residues, including serine, threonine, P450 enzymes in a mechanism-based fashion^^"^^.
and tyrosine, are present. If the protein nucleophile Its inactivation was shown to result from P450-
is the hydroxyl group of such a residue, the result- dependent metabolic activation of its acetylenic
ing adduct would be an ester. Indeed, recent stud- moiety to a species that alkylates a heme pyrrole
ies with a CYP2B4 T302A mutant reveal that the nitrogen. More recently, EE has been shown to also
^inact ^^ 2-eth3aiylnaphthalene is decreased from modify the CYP3A4 protein in a reconstituted
0.20 ± 0.05 min-^ for the wild type to 0.05 ± enzyme system with ^^^^^^^ = 0.04 min" ^ K^ =
0.01 min"' in the mutant, suggesting that T302 is 18 |JLM, ^1/2= 16 min, and a partition ratio of
at least one acylated residue ^^^. ~50^^. Loss of activity was paralleled
7-Ethynylcoumarin (7-EC) (Figure 7.9)^^ by some loss of CO-dependent chromophore.
is a rationally designed mechanism-based inacti- A net stoichiometry of 1.3 nmol of EE-metabolite
vator with the ethynyl moiety at the 7-position bound/nmol CYP3A4 was observed with [^H]EE
of coumarin where, by analogy to 7-ethoxy- as the substrate. HPLC analysis yielded several
coumarin and 7-ethoxy-4-(trifluoromethyl) NADPH-dependent [^HJEE-labeled fractions,
coumarin, it should be readily oxidized by CYP2B the predominant one of which eluted at 31 min.
enzymes•^^' ^^^. In comparative assays, 7-EC ESIMS analysis in the negative ion mode of this
(100 JULM) only marginally (15%-20%) inactivated peak fraction yielded a mass (M-H)~ of 479 Da.
human liver microsomal CYP2A6, but markedly Although the chemical structure of this EE-related
(>90%) inactivated purified CYP2B191 This species has not been definitively characterized, the
inactivation left the heme and its thiolate ligand authors believe it to be an EE-adducted mono-
unscathed, as the electronic absorption of the pyrrolic hemeftagment^^.EE thus appears to modify
reduced-CO complex of the inactivated enzyme both the heme and protein moieties of CYP3A4,
was little affected. The inactivation by 7-EC was whereas only the heme moieties of CYP2B1 and
unaffected by the presence of nucleophiles such as -2B6 are susceptible to EE-modification.
GSH and NaCN, of the iron-chelator deferoxam- The monoamine oxidase (MAO) inhibitor
ine, or of superoxide dismutase or catalase and deprenyl (Figure 7.9) is a propargylamine whose
conformed to all the other established criteria for terminal acetylene reacts irreversibly with the
a mechanism-based inactivation. It exhibited a MAO flavin moiety^^^. Deprenyl also inacti-
K^ of 25 ± 2 |xM, a k-^^^^ at 30°C of 0.39 ± 0.01 vates CYP2B1 relatively selectively with K^ =
min~', a partition ratio of 25, and a half-life (ty^) 1.05 fjiM, A:.j^^^j = 0.23 min~^ and partition
of 1.8 min. ESI-LC/MS of the dialysed intact ratio = 2^^^. However, although there is a loss of
Inhibition of Cytochrome P450 Enzymes 259

O^^O ^

CO2H
M e ^ ^ - ^ O

7-Ethynylcoumarin Deprenyl 9'-Propargyl abscisic acid

Me2N

HO
17a-Ethynylestradiol Mifepristone

Figure 7.9. The structures of acetylenes that have been shown to inactivate P450 enzymes. In all cases except
perhaps that of mifepristone the reactive intermediate appears to arise by oxidation of the triple bond.

spectrally detectable P450 chromophore, no modification^ ^^' ^^^. Licubations of radiolabeled


significant change is observed in the 405 nm PCP with NADPH-supplemented liver microsomal
heme absorbance. The complete loss of CYP2B1 preparations from phenobarbital-pretreated rab-
function, however, suggests protein modification bits demonstrated a time- and concentration-
following activation of the terminal acetylene. dependent loss of ketamine and benzphetamine
Analogs of deprenyl such as (i?)-A^-(2-heptyl)-A^- A^-demethylase activity as well as irreversible
methyl-propargylamine and (i?)-A^-(2-heptyl)- binding of the radiolabeled amine to microsomal
propargylamine have been tested for their P450 protein^^^^^^. Inhibition of both of these events by
isoform inactivation selectivity^^^. Of these, (R)- cyanide (NaCN), but not GSH, led to the proposal
A/-(2-heptyl)-propargylamine was the more potent, that a-oxidation yielded a PCP iminium species that
with K^ = 0.5 |JLM and k-^^^^ = 0.36 min-^ i^^. was responsible for the inactivation. This iminium
A specific inhibitor of the P450 enzyme that derivative could directly bind to proteins, but appar-
degrades the phytohormone abscisic acid has been ently could also undergo further NADPH-depend-
constructed by replacing the methyl group of ent metabolism by P450, resulting in inactivation of
abscisic acid that is normally oxidized with those enzymes ^^^^'^^. This second oxidation was
acetylenic moieties^^^ The most active of the proposed to convert the PCP iminium ion to an allyl
inactivating agents is 9'-propargyl abscisic acid alcohol via a reactive, electrophilic 2,3-dihydropyri-
(Figure 7.9), which in vivo exhibits a greater inhi- dinium intermediate [l-(l-phenylcyclohexyl)-2,3-
bition of Arabidopsis thaliana seed germination dihydro-4-pyridone] that reacted irreversibly with
than abscisic acid itself. the protein^ ^^. In vitro studies with five rabbit liver
P450 isoforms confirmed that the inactivation was
indeed mechanism-based, highly selective for
CYP2B4, and involved parallel loss of the CYP2B4
3.1.3. Other P450 Protein
heme chromophore and function^^^. However, par-
Modifying Inactivators
allel studies of PCP and its iminium metabolite
The recreationally abused psychotomimetic revealed that the latter was less selective than the
amine phencyclidine [l-(l-phenylcyclohexyl) parent compound and inactivated P450 3b (a consti-
piperidine; PCP] (Figure 7.10) is another mecha- tutive rabbit liver isoform) as well^^^. More recent
nism-based inactivator that inactivates CYP2B1, in vivo studies have shown that CYP2D is also
-2B4, and -2B6 relatively selectively via protein markedly (>74%) inactivated by PCP in uninduced
260 M.A. Correia and P.R. Ortiz de Montellano

Phencyclidine Cannabidiol

Figure 7.10. Phencyclidine is oxidized to an iminium species whose role as an intermediate in the inactivation of
P450 enzymes is unclear. The structure of cannabidiol (CBD), which also inactivates P450 enzymes by an undefined
mechanism, is also shown.

rats^^^. Comparative mviYro studies of PC? analogs coumarin 0-deethylase, respectively) yielded
containing a five [PCPY; l-(l-phenylcyclohexyl) K^ values of 3.8 and 10 JULM, k.^^^^^ values of
pyrrolidine]- or six [PCHMI; l-(l-phenylcyclohexyl) 0.12 min~' and 0.01 min"', respectively, and
hexamethyleneimine]-membered heterocyclic ring partition ratios of ~45 for both processes. More
indicated that the rates and t^i^ of inactivation and importantly, contrary to previous reports, not only
heme loss were comparable for PCPY and PCP, was CN (up to 1 mM) incapable of inhibiting
both of which were 10-fold higher than those of either inactivation event, but little inactivation was
PCHMI even though the latter was metabolized to a observed when the synthetic amino enone [1-
greater extent^^'. No mechanism-based inactivation (l-phenylcyclohexyl)-2,3-dihydro-4-pyridone],
was observed with either phenylcyclohexylamine or the putative reactive electrophilic 2,3-dihydropyri-
the diethylamino analog of PCP, suggesting that the dinium intermediate, was used as the inactivating
substituted nitrogen is necessary and must be incor- substrate'^2, 173, 182 gaudies with pHJPCP
porated into a heterocyclic ring. The observation of revealed a molar stoichiometry of PCP binding to
prosthetic heme loss and its sensitivity to cyanide CYP2B1 and CYP2B6 protein of 4:1 and 5.5 : 1 ,
ion reinforced the notion that the iminium ion is respectively. GSH (10 mM) did not decrease the
involved in the PCP-mediated inactivation^^'. rate of PCP-mediated inactivation but reduced
HPLC-analyses of heme extracts from PB- the binding of [^H]PCP to CYP2B6 protein to the
pretreated rabbit liver microsomes, incubated with expected 1:1 molar stoichiometry. Consistent
or without PCP, identified PCP-modified heme with these findings, LC-MS analyses of the
fractions proposed to be A/-modified porphyrins, PCP-P450 adduct in the ESI-LC/MS mode
but the nature of these porphyrins was not estab- yielded a mass difference of 244 ± 5 Da between
lished'^^. It is therefore unclear whether heme PCP-inactivated CYP2B1 and the wild-type
modification or degradation was responsible for protein, confirming the formation of a 1:1
the loss of P450 activity or simply coincidental PCP-CYP2B1 adduct. Similar studies of PCP-
with it. However, a substantial discrepancy in the inactivated CYP2B4 gave a mass difference of 261
partition ratio obtained for PCP-mediated func- ± 2 Da, consistent with the binding of one mole-
tional inactivation and that for heme loss sug- cule of monohydroxylated PCP to the CYP2B4
gested that this enzyme inactivation might also protein. Similar analyses of PCP-CYP2B6 adducts
entail protein modification. were less conclusive due to inconsistent results'^^.
Protein modification might indeed be prima- Collectively, the recent findings argue for protein
rily responsible for the PCP-mediated inactivation modification at the CYP2B active site as the major
of CYP2B1 and -2B6, as no loss of heme chro- mode of inactivation. Although a reactive elec-
mophore in its ferric or CO-bound form was trophilic inactivating species is involved, the PCP-
detected'^2, 173 studies with CYP2B1- and iminium ion may not be its precursor.
CYP2B6-selective probes (7-ethoxycoumarin Modification of the P450 protein is also
0-deethylase and 7-ethoxy-4-(trifluoromethyl) observed with caimabidiol (CBD) (Figure 7.10), a
Inhibition of Cytoclirome P450 Enzymes 261

major constituent of marijuana. CBD inactivates GSH in these incubations did not prevent P450
mouse P450 isoforms 2C and 3A via a mechan- inactivation but suppressed nonspecific covalent
ism that involves stoichiometric covalent binding binding. The residual covalent binding of 8-MOP
of the inhibitor and loss of its CBD oxidase activ- to microsomal protein was nearly stoichiometric
j^i83, 184 Limited structure-activity studies show with the loss of microsomal P450i°^' ^^^. This
that a free phenolic group in the resorcinol moiety finding, together with the observation of a type I
of CBD is essential for inactivation^^^. Mass spec- spectrum in incubations of rat liver microsomes
trometric analyses of the concomitantly generated with 8-MOP in the presence but not absence of
CBD metabolite trapped as a GSH-adduct led NADPH^^^, implicated an active-site directed
to identification of CBD-hydroxyquinone as the inactivation mechanism. Furthermore, because
inactivating species ^^"^j a finding consistent with cysteine markedly quenches the covalent binding
the electrophilic reactivity and P450 inactivat- of the label from [^HJ-methoxy- but not [4-^'^C]-
ing potential of this compound^ ^^. HPLC-peptide ring-labeled methoxypsoralen, the reactive inter-
mapping, microEdman degradation, and mass mediate is likely to reflect oxidation of the frjran
spectrometric analyses of the CBD-modified pep- j.jjjgi02 Yjjjg inference is supported by the finding
tides obtained by proteolytic digestion of CBD- that trioxsalen (trimethylpsoralen), in which the
inactivated CYP3A11 identified the peptide region furan ring bears a methyl substituent, does not
spanning residues A344-K379 and G426-K434 as the destroy P450. Methyl-substituted fiirans, however,
loci of CBD modification^^^. These regions corre- can also inactivate P450 enzymes, as illustrated by
spond to the CYP3A11 active site SRS-5 in the the inactivation of human liver enzymes observed
K-region and the heme-binding Cys443-region/ in incubations with (i?)-(+)-menthofuran^^^.
helix L domain^^^^^^, fiilfilling an essential crite- 8-MOP is a potent inactivator of human liver
rion of a mechanism-based inactivation process^^'*. CYP2A6 and rat liver CYP2B1, but also inactivates
Interestingly, both peptides contain Cys residues CYP2B2, -lA, -3A, and -2C11^^' i^^, 188 Qf all the
that could react with the CBD-hydroxyquinone. friranocoumarins tested, 8-MOP was the most
Analogous studies with CYP3A4 have identi- potent inactivator of CYP2B1 with a K^, k^nacv ^^^
fied similar protein adducts, albeit at a lower partition ratio of 2.9 (ULM, 0.34 min~^ and 1.3,
level than those detected with the mouse ortho- respectivelyi^l SDS-PAGE and/or HPLC analy-
log (L.M. Bomheim, personal communication). ses of the components from incubations of puri-
Although A^- and A^-tetrahydrocannabinol (THC), fied CYP2B1 with [i4C-]8-MOP indicated that
the other major and minor psychoactive compo- the radiolabel bound to the protein rather than to
nents of marijuana, respectively, are efficiently the heme with a stoichiometry of 0.7 : 1 . This [^"^C-
metabolized by CYP3A and -2C, they are not ]8-M0P-binding was unaffected by GSH or
inactivating agents. In contrast, unsaturated A^- methoxylamine (MOA), suggesting that covalent
THC-enyl and -ynyl analogs that are also present binding occurred within the active site where it
in the complex marijuana extracts have been was inaccessible to external nucleophiles.
found to be selective mechanism-based inactivat- LC-ESIMS analyses of the [i4C-]8-MOP-modi-
ing agents in vitro but not in vivo^^^. fied CYP2B1 revealed a mass shift of 237.9 ± 9.6
Other classes of compounds that inactivate Da over that of the native enzyme. Analogous
human and rat liver microsomal P450 enzymes studies of 5-MOP- and psoralen-modified
via protein modification are known. One such CYP2B1 gave mass shifts of 240 ± 6.2, and 204
class is represented by the furanocoumarins, ±11.8 Da, respectively^ ^^. These results indicate
natural constituents of foods such as celery, that a single psoralen molecule is covalently
parsley, figs, parsnips, and grapefruit juice^^^. bound to the protein and are consistent with
Among these compounds, the photoactive lin- CYP2B1-catalyzed oxidation of the 8-MOP ftiran
ear furanocoumarin, 8-MOP (methoxsalen) is a ring to the 8-MOP ftiranoepoxide (MW, 232.2 Da)
particularly potent P450 inactivator. Evidence that followed by reaction of the protein with the epoxide
8-MOP was activated by P450 enzymes to an or a cationic species derived from it.
electrophilic product that bound covalently to the Another notable fiiranocoumarin, 6',7'-DHB,
protein^^~^^^ was obtained from incubations with that is derived from the processing of grapefixiit
rodent liver microsomes. Inclusion of cysteine or juice and is one of its common constituents.
262 M.A. Correia and P.R. Ortiz de Montellano

is now a well-recognized mechanism-based inacti- are tamoxifen and CBZ. Tamoxifen (Z-
vator of CYP3A4io^-^09, i89 inactivation of intes- {1 -[4-(2-dimethyl-aminoethoxy)phenyl]-1,2-
tinal CYP3A4 after oral grapefruit juice intake has diphenyl-1-butene}) (Figure 7.11), a nonsteroidal
been clinically associated with increased bioavail- antiestrogen used in hormone-dependent breast
ability of several CYP3A4 drug substrates. cancer chemotherapy, inactivates CYP2B6 (but
Numerous reports exist of clinically relevant not CYPIBI and CYP3A4) with K^ = 0.9 JULM,
adverse drug-drug interactions between grapefruit ^inact =" ^'^^ min~\ and ty2 = 34 min. CYP2D6
juice and CYP3A4 substrates, in some instances and CYP2C9 were also partially (25%) inacti-
resulting in withdrawal of the drug in question^^^. vated''"^''^' '^'. The ultimate inactivating species
Studies with 6',7'-DHB indicate that it is activated remains to be identified, but the sequential metab-
to a reactive species that irreversibly modifies the olism of tamoxifen to 4-hydroxytamoxifen, and
CYP3A4 protein with K^ = 59 |LJLM and ^-^^^^ = then to 3,4-dihydroxytamoxifen-o-quinone, a
0.16 min~^ 107-109^ resulting in accelerated hepatic known reactive and carcinogenic species'^', sug-
degradation of the protein (see below; Malhotra and gested it might be responsible for the inactiva-
Watkins, personal communication). tion"^. This proposal has been challenged by
The ftiranopyridine L-754,394, 7V-[2(i^)- studies showing that incubation with 3-hydroxy or
hydroxy-l(5')-indanyl]-5-[2(5')-((l,l- 4-hydroxytamoxifen reversibly inhibits CYP3A4
dimethylethyl)amino)carbonyl]-4-[(furo[2,3-^] but does not inactivate it"^. A mechanism-based
pyridin-5-yl)methylpiperazin-1 -yl]-4(»S)-hydroxy- inactivation by tamoxifen and its A^-desmethylta-
2(i?)-(phenylmethyl)pentenamide, is also a potent moxifen metabolite (instead of 3- or 4-hydroxyta-
mechanism-based inactivator of human liver moxifen) involving the formation of a metabolic
microsomal CYP3A4 but is less selective, as it intermediate (MI) complex was proposed as an
also inactivates human CYP2D6^io-*i2,190 j ^ ^ ^^^ alternative explanation"^.
^inact' ^^^ partition ratio for human liver microso- CBZ (Figure 7.11), a widely used anticonvul-
mal CYP3A4 are reportedly 7.5 JULM, 1.62 min~^ sant, is known to be metabolized to several reac-
and 1.35, respectively^^'. Structural characteriza- tive species, including the arene oxide, 9-acridine
tion of the concomitantly generated L-754,394 carboxaldehyde, and an iminoquinone metabo-
metabolites revealed that the mechanism of inacti- jj|.gi92-i96 Clinically, CBZ is often associated with
vation probably entails CYP3A4-dependent oxi- idiosyncratic hypersensitivity syndromes'^^"'^^.
dation of the furan ring to the corresponding CBZ-treated patients often exhibit anti-CYP3A4
epoxide and/or 7-ketoenal that binds to the protein autoantibodies, suggesting that covalent binding
within the active site. Accordingly, neither the cor- of CBZ to CYP3A4 leads to the immunoproteoso-
responding dihydrofuran derivative nor the analog mal formation of antigenic CBZ-modified pep-
that lacks the furan ring is active as a CYP3A4 tides'^^' 199 CBZ is indeed an excellent CYP3A4
irreversible inhibitor. Indeed, Tricine-SDS-PAGE, substrate that is metabolized predominantly to the
HPLC-peptide mapping, and MALDI-TOF-MS CBZ 10,11-epoxide, a reaction that has been used
analyses of L-754,394-bound CYP3A4 showed as a CYP3A4 functional marker^^^. The incuba-
that the inhibitor bound to the I-helix peptide tion of [^HJCBZ with CYP3A4 results in irre-
^257~^3i7^^^- ^^^ chemical instability of this versible binding of [^H]CBZ to the protein with a
adduct under the acidic conditions required for stoichiometry of 1.58 ± 0.15 pmol pH]CBZ
these analyses precluded identification of the bound/pmol CYP3A4. In the presence of GSH (4
active-site residue actually modified. In view of mM) this stoichiometry is reduced to 1.09.
the markedly higher stability of the adducts with However, no concentration (0-1 mM) nor time-
GSH, A^-acetylcysteine (NAC), and MOA, and of dependent CYP3A4 inactivation was detected in
0-linked heterodimers of the hydroxylated/parent these incubations^^'. In the absence of other sub-
furano- compounds, the instability led the authors strates, CBZ, if anything, protected from
to propose an ester linkage between activated L- NADPH-dependent oxidative uncoupling^^'.
754,394 and CYP3A4. The authors proposed Given these findings, one can speculate that the
E307 as the most plausible target''^. serum anti-CYP3A4 autoantibodies detected in
Two other drugs of note that have been reported CBZ-dependent hypersensitivity might arise in
to inactivate P450 by protein modification patients whose CBZ-CYP3A4 adducts remain
Inhibition of Cytochrome P450 Enzymes 263

[Fe=0]^

OMe
8-Methoxypsoralen
.0^
^NMe2

Ph CONH2
Tamoxifen Carbamazepine

Figure 7.11. 8-Methoxypsoralen (8-MOP) is oxidized to an epoxide, and either the epoxide or a ring-opened
product derived from it is responsible for inactivation of P450. The structures of two drugs, tamoxifen and
carbamazepine (CBZ), that inactivate P450 by unknown mechanisms are also shown.

stable, possibly because of low intrahepatic GSH 3.2. Quasi-Irreversible


levels, and are therefore available for antigenic Coordination to the
processing ^ ^^'^^^
Prosthetic Heme
Collectively, the results convincingly establish
that the reactive species formed by P450 enzymes This section focuses on inhibitors that are
can alkylate, acylate, or otherwise modify the transformed by P450 enzymes into metabolic
protein skeleton, resulting in the loss of catalytic intermediate (MI) products that coordinate so
activity. It is likely, furthermore, that protein tightly to the heme iron atom that they can be dis-
modification has gone undetected in some placed only under unique experimental condi-
instances where inactivation has been attributed to tions. The two major classes of these inhibitors are
heme modification. Conversely, the data on compounds with a dioxymethylene function and
parathion and CBZ suggests that heme destruction nitrogen compounds, usually amines that are con-
is required for enzyme inactivation in some verted in situ to nitroso metabolites. A related
instances where protein modification clearly mechanism is also partially responsible for the
occurs. Furthermore, the fact that CYP2B1 is inhibition of P450 by 1,1-disubstituted hydrazines
inactivated by secobarbital by both heme alkyla- and acyl hydrazines. The anaerobic reductive
tion and protein modification, by phenylacetylene coordination of halocarbons to the heme iron atom
predominantly by heme alkylation, and by 2- is discussed in Section 3.4 because the reaction is
ethynylnaphthalene predominantly by protein linked to destruction of the heme.
acylation, underscores the critical role of active-
site-substrate interactions in dictating the mode
of inactivation. Active-site-inhibitor interactions 3.2.1. Methylenedioxy Compounds
presumably also explain why A/-phenyl or iV-octyl
P450 enzymes oxidize aryl and alkyl methyl-
2,2-dichloroacetamides predominantly inactivate
enedioxy compounds, some of which are used as
CYP2B1 via protein acylation whereas the
insecticide synergists^^^' ^^^, to species that coordi-
corresponding N-hexy\, A^-butyl, or iV-methyl
nate tightly to their heme iron atom^^^. The time,
dichloroacetamides do so via heme destruction^^' ^^.
NADPH, oxygen, and concentration dependence
Similar factors presumably also explain the dif-
of the reaction, as well as the finding that NADPH
ferential modes of inactivation observed with ter-
and oxygen can be replaced by cumene hydroper-
minal acetylenes (e.g., A^-alkylation of CYP2B1 by
oxide, confirm that the inhibitory species is
phenylacetylene vs protein acylation of CYP4A1
unmasked by the catalytic action of the P450
by 10-undecynoic acid)^^' ^^.
264 M.A. Correia and P.R. Ortiz de Montellano

enzymes^^^' ^^^' ^^^. The resulting ferrous complex stability: alkyl chains of 1-3 carbons yield unsta-
is characterized by a difference absorption spec- ble complexes whereas those with longer alkyl
trum with maxima at 427 and 455 nm, whereas groups are stable^ ^^' ^^^. As in the case of reversible
the ferric complex exhibits a single absorption inhibitors (Section 2.3), the ferrous complex is sta-
maximum at 437 nm^^^' ^^^. The ferrous peaks at bilized by concurrent binding interactions of the
427 and 455 nm are due to distinct complexes, ligand with the lipophilic active site^^^. The ferrous
although their structural interrelationship is to ferric transition weakens the complex, indicat-
obscure^^"^. The ferrous complex can be isolated ing that the activated species, like carbon monox-
intact from animals treated with isosafrole, demon- ide, only strongly coordinates to the ferrous iron.
strating its stability, but the less stable ferric com- The above results are consistent with the
plex can be disrupted by incubation with lipophilic catalysis-dependent generation of a carbene-iron
compounds with concomitant regeneration of complex (Figure 7.12). The synthesis and charac-
the catalytically active enzyme^^^' ^^^. The ferrous terization of model carbene complexes provides
complex is unaffected by incubation with supporting evidence for a carbene complex^^'^' ^^^.
lipophilic compounds but can be disrupted by The structural analogy between a carbene and
irradiation at 400-500 nm^^^' ^^K Structure activity carbon monoxide provides a ready explanation for
studies of 4-alkoxy-l,2-methylenedioxybenzene the unusual 455-nm absorption maximum of the
reveal that the size and lipophilicity of the alkoxy complexes. As already noted, a different complex
group is an important determinant of the complex is responsible for the absorption maximum at

HoO

Figure 7.12. The quasi-irreversible inactivation of P450 enzymes by methylenedioxy compounds involves
oxidation of the methylene bridge to a species that forms a tight, but reversible, complex with the heme iron atom.
The coordinating species is probably a carbene, as shown. Paroxetine is an example of a drug that inhibits P450 by
this mechanism.
Inhibition of Cytochrome P450 Enzymes 265

427 nm, perhaps a carbene complex in which precede formation of the bridge-hydroxylated
the trans hgand, as in P420, is not a thiolate^^^. metabolite if the radical formed in the hydroxyla-
A carbene complex also provides a ready rationale tion reaction is oxidized by the ferryl species
for the incorporation of oxygen from the medium before the oxygen rebound occurs (Figure 7.12,
into the carbon monoxide metabolite formed from path b). Finally, the same radical intermediate
the dioxymethylene bridge carbon (see below), could bind to the iron of the [Fe-OH]^+ catalytic
and the observation that carbon monoxide forma- intermediate^ ^'^. Deprotonation and intramole-
tion is enhanced by electron-withdrawing sub- cular transfer of the oxygen from the iron to
stituents^^^. Water addition to the iron-coordinated the carbon would give the bridge-hydroxylated
carbene produces an iron-coordinated anion that metabolite that could then decompose to the
should readily decompose into the observed cate- carbene complex as in the first mechanism.
chol and carbon monoxide metabolites. A differ- Whatever the precise mechanism, in vitro
ent but undefined mechanism is required to experiments with purified CYP2D6 indicate that
explain the incorporation of an atom of molecular the formation of MI complexes with a telltale
oxygen into a fraction of the carbon monoxide^ ^'^. spectroscopic signature at 456 nm may be respon-
The link between the dioxymethylene function sible for the clinical reports of potent CYP2D6
and P450 inhibition, the requirement for catalytic inhibition by paroxetine (Figure 7.12), a serotonin
activation of the inhibitor, and the fact that the reuptake inhibitor^^^"^^^. The formation of a
dioxymethylene group is oxidized implicate this carbene complex is supported by the fact that
function in the inhibitory events. An inhibitory paroxetine is metabolized by CYP2D6 via
role has been postulated for free radical^ ^^, demethylenation of the methylenedioxy group to
carbocation^^^, and carbanion^^^ intermediates, a catechol and formic acid^^^' ^^^. Kj and A:.^^^^
but formation of the carbene from the bridge- values of 6.6 ± 2.7 |JLM and 0.25 ± 0.09 min"^
hydroxylated metabolite or from its radical respectively, have been calculated for the paroxe-
precursor is most consistent with the results tine-mediated inhibition of human liver micro-
(Figure 7.12). Substituents other than an alkoxy somal CYP2D6-dependent dextromethorphan
group on the dioxymethylene group suppress 0-demethylation^^ ^.
complex formation^^^' ^^'^' ^^°. The retention of Additional methylenedioxyphenyl compounds
activity with an alkoxy substituent is understand- have been synthesized and their human isoform
able because O-dealkylation of the substituent selectivity as mechanism-based inactivators evalu-
provides an independent route to the bridge- ated^^ ^. Their inactivating potential depends on
hydroxylated precursor of the carbene^ ^^ The oxi- the side-chain structure, with bulky side chains
dation of aryldioxymethylenes to catechols, such as 1,4-benzothiazine inactivating some P450
carbon monoxide, carbon dioxide, and formic acid enzymes but not others^^^.
is consistent with hydroxylation of the methylene-
dioxy bridge^^^' ^^^' 221-223^ ^g jg ^^ observation
that deuterium substitution on the dioxymethylene
3.2.2. Amines
carbon decreases the rate of formation of carbon Alkyl and aromatic amines, including the
monoxide {k^k^^ = 1.7-2.0). The observation MAO inhibitor clorgyline^^"^, and a number of
of a similar isotope effect on the insecticide clinically usefiil amine antibiotics such as trolean-
synergizing in vivo activity of these compounds domycin (TAO) (Figure 7.13) and erythromycin,
clearly links the formation of carbon monoxide belong to a second large class of agents that form
with complex formation and P450 inhibition^^^. quasi-irreversible (MI) P450 complexes^' 235-240
Three mechanisms can be envisioned for These amines are oxidized to intermediates that
oxidation of the dioxymethylene bridge to the coordinate tightly to the ferrous heme and give
iron-coordinated carbene. In one mechanism, eli- rise to a spectrum with an absorbance maximum
mination of a molecule of water after hydroxyla- at 445-455 nm^^^. Complex formation requires a
tion of the dioxymethylene bridge yields an acidic primary amine but secondary and tertiary amines,
oxonium ion that upon deprotonation gives the as in the case of TAO, can give P450 complexes if
carbene (Figure 7.12, path a). In a second mecha- they are first iV-dealkylated to the primary amines.
nism, formation of the oxonium species could The complexes from aromatic amines differ from
266 M.A. Correia and P.R. Ortiz de Montellano

R-N(CH3)2- R-NH2
t
O AcO NMe2
Me?

R-N=0- R-NHOH
"'0*^0'"'''Me

Troleandomycin

N-N —N

OAc

Figure 7.13. The spectroscopically detectable metabolic intermediate (MI) heme complexes formed when some
primary amines are oxidized by P450 enzymes involve oxidation of the nitrogen to a nitroso species that coordinates
to the iron. The primary amine function can be unmasked by A^-demethylation reactions, as is the case in the
inhibition of CYP3A enzymes by troleandomycin (TAO) (AcO- in the structure represents CH3CO2). The arrow
shows the nitrogen that is involved in the reaction in TAO.

those from alkyl amines in that they are unstable concentration of the protein in the cell increases,
to reduction by dithionite^^^. The normal compet- an example of "induction" through protein stabili-
itive inhibition associated with the binding of zation. It remains unclear whether the protein
amines does not, of course, depend on catalytic levels are elevated because of a substrate-induced
oxidation of the inhibitor, but catalytic activation conformational stabilization or because the for-
is required for formation of the tight, quasi- mation of a heme complex suppresses normal
irreversible complexes^^"*' ^^^' ^^^. It is likely that damage to the protein associated with the reactive
the primary amines are first hydroxylated because O2 species produced through uncoupled turnover
the same complexes are obtained with the corre- of the enzyme. This latter possibility is the most
sponding hydroxylamines^"^^, but the coordination likely, given that inhibition of P450 reductase^"^^'
requires further oxidation and thus involves a •^^^ or conditional deletion of the reductase^"*^,
function beyond the hydroxylamine^^^' •^^^. In fact, which suppresses catalytic turnover, also results in
the moiety that chelates to the iron appears to be enzyme stabilization.
the nitroso function obtained by two-electron oxi-
dation of the hydroxylamine (Figure 7.13)^^^"^'^^ 3.2.3. 1,1-Disubstituted and
As hydroxylamines readily autooxidize, the final
Acyl Hydrazines
oxidative step may not always require catalytic
participation of the enzyme^^^. The coordination P450 enzymes oxidize 1,1-disubstituted, but
of a nitroso function is consistent with the obser- not monosubstituted, hydrazines (see Section
vation that apparently identical complexes are 3.3.4) to products that coordinate tightly to the
obtained by reduction of nitro compounds^"^^. The heme iron atom. The complexes, which are
crystal structure of a complex between a nitroso characterized by a ferric absorption maximum
compound and a model iron porphyrin shows, as at ~438 nm and a ferrous maximum at 449 nm,
expected, that the nitrogen rather than the oxygen are formed in a time-, NADPH-, and oxygen-
of the nitroso group is bound to the iron^^"^. dependent manner^^^. The oxidation of isoniazid
It is noteworthy that the in vivo complexation and other acyl hydrazines by liver microsomes
of TAO to the heme of CYP3A enzymes stabilizes yields a transient complex with a similar absorp-
them and prolongs their half-lives in hepato- tion maximum at 449 nm^^^' ^^^. However, the
cytes^ ^. A consequence of this is that the isoniazid complex dissociates on addition of
Inhibition of Cytochrome P450 Enzymes 267

R [Fe=0]^ R
N-NH2 N-NH
R

[Fe-OH] +3

R H
N-N: N-N
-H2O / \
R R OH

Figure 7.14. The nitrene-iron structure proposed for the complexes formed during the metaboHsm of
1,1-dialkylhydrazines and possible mechanisms for formation of the nitrene.

ferricyanide and thus is only stable in the ferrous to be generated that is either reversible or too
state^^^. Model studies indicate that 1,1-dialkylhy- unstable to be isolated. Unfortunately, the quanti-
drazines are oxidized to disubstituted nitrenes that tative correlation of heme adduct formation
form end-on complexes with the iron of metallo- with enzyme inactivation is technically difficult.
porphyrins. The nitrene complexes formed in Without such data, it is difficult to exclude the
the reactions of l-amino-2,2,6,6-tetramethyl- possibility that the enzyme is also inactivated by
piperidine and several iron tetraarylporphyrins mechanisms such as protein modification even
have been characterized by NMR, Mossbauer, and when heme alkylation is conclusively demonstrated.
X-ray methods^^^' ^^^. The P450 complexes gener-
ated during the metabolism of 1,1-disubstituted
hydrazines, and possibly acyl hydrazines, are
therefore likely to be aminonitrene-iron com-
3.3.1. Terminal Olefins
plexes (Figure 7.14). Oxidation of the dialkylhy- The P450-catalyzed epoxidation of terminal
drazines to aminonitrenes is easily rationalized by olefins is often associated with iV-alkylation of its
initial hydroxylation of the hydrazine or, more prosthetic heme and inactivation of the enzyme
probably, by stepwise electron removal from the (see Figure 7.7)^^^' 1^^' ^^^. Early studies with
hydrazine (Figure 7.14). 2-isopropyl-4-pentenamide (AIA) and 5-allyl-
substituted barbiturates such as secobarbital^^^'
3.3. Covalent Binding to the ^^^, established that the oxidative metabolism of
Prosthetic Heme homoallylic amides results in: (a) comparable loss
of P450 and heme content, (b) the accumulation
P450 is often irreversibly inactivated via cova- of "green pigments" identified as abnormal
lent attachment of the catalytically activated porphyrins, and (c) derangement of the heme
inhibitor, or a fragment of it, to the heme group. biosynthetic pathway.
A heme alkylation mechanism has been unam- The only structural requirement for prosthetic
biguously demonstrated, in many instances, by heme alkylation by olefins is a monosubstituted
evidence of equivalent activity and heme loss and double bond. Accordingly, ethylene, but not
the isolation and structural characterization of the ethane, is able to destroy the P450 heme while
modified hemes. It must be noted that in the 3-hexene, cyclohexene, and 2-methyl-l-heptene
absence of explicit evidence for heme adduct for- are inactive^^^' ^^^. Even monosubstituted olefins
mation, an equimolar loss of enzyme content and fail to inactivate the enzyme if they are not sub-
heme does not unambiguously establish that heme strates for the enzyme, if the double bond is not
alkylation is responsible for enzyme inactivation the site of catalytic oxidation, or if the double
because alternative mechanisms exist for the bond is a part a of a conjugated system^^^. Thus,
catalysis-dependent destruction of the heme (see the oxidation of styrene by a model iron porphyrin
Section 3.4). It is also possible for a heme adduct showed that heme alkylation only occured once in
268 M.A. Correia and P.R. Ortiz de Montellano

ten thousand turnovers^^^, in contrast to the ratio porphyrin nitrogen to the epoxide metabolite of
of less than 300 turnovers per alkylation even the olefin, but this possibility is precluded by the
that is commonly observed with unconjugated ter- following findings: (a) the enzyme is refractory to
minal olefins^. These observations suggest that inactivation by the epoxides of olefins that destroy
alkylation of the heme by olefins is compromised the enzyme ^^^' ^^^' ^^^, (b) c/^-addition of the nitro-
by steric constraints and/or by the presence of gen and oxygen to the double bond is inconsistent
substituents that can delocalize charge or electron with the trans sterochemistry expected for the
density from the double bond. addition of a nucleophile to an epoxide^^^, (c) the
Spectroscopic methods have unambiguously nitrogen reacts with the terminal rather than inter-
established the structures of A^-alkylated porphyrins nal carbon of vinyl ethers although the internal
isolated from the livers of rats treated with diverse (oxygen-substituted) carbon is more reactive in
olefins, including ethylene, propene, octene, the corresponding epoxides^^^, and (d) the pyrrole
fluroxene, 2,2-diethyl-4-pentenamide, 2-isopropyl- nitrogens are weak nucleophiles and do not react
4-pentenamide, and vinyl fluoride'^' 257-26O with epoxides even under harsh chemical condi-
Analogous products are probably formed in the tions. These considerations and the requirement
inactivation of P450 by other olefins, such as in the for enzyme turnover indicate that catalytic oxygen
inactivation of CYP2E1 by the garlic components transfer to the double bond initiates enzyme inac-
diallyl sulfide and diallylsulfone26i-264^ but the tivation but it does not result from reaction with
resulting adducts have not been isolated. The the epoxide metabolite.
terminal carbon of the double bond is bound to a Ethylene, propene, and octene, all linear
porphyrin nitrogen and the internal carbon of olefins, only detectably alkylate pyrrole ring D
the olefin bears a hydroxyl group in the structures of the prosthetic heme of the phenobarbital-
of all the olefin adducts determined so far inducible rat liver P450 enzymes (Figure 7.15),
(Figures 7.7 and 7.15'^^*' ^58^ jj^g oxygen in the but heme alkylation by the more "globular"
ethylene and 2-isopropyl-4-pentenamide adducts, olefins 2-isopropyl-4-pentenamide and 2,2-
which has been shown by '^O studies to derive diethyl-4-pentenamide is less regiospecific^^^' ^^^.
from molecular oxygen, is presumed to be the cat- The regiochemistry and stereochemistry of heme
alytically activated oxygen^^^' ^^^. The structure of alkylation by ^ra«5-[l-^H]-l-octene has estab-
the adduct is consistent with addition of the lished that the olefin stereochemistry is preserved

P450

P450

HO^ H
Figure 7.15. The oxidation of ^ra«5-l-[l-^H]octene by rat liver microsomes yields both the epoxide metabolite
and the indicated A^-alkyl heme adduct, the structure of which has been unambiguously established. The heme
substituents are: Me=CH3, V=CH=CH2, P=CH2CH2C02H. The peripheral pyrrole carbons and the meso carbons
of the porphyrin are labeled.
Inhibition of Cytoclirome P450 Enzymes 269

during heme alkylation. Furthermore, heme alky- transient carbon radical that alkylates the heme,
lation only occurs when the oxygen is delivered to closes to the epoxide, or transfers the unpaired
the re face of the double bond even though stere- electron to the heme to give a cation that alkylates
ochemical analysis of the epoxide metabolite the heme. The partitioning between metabolite
shows that the oxygen is delivered almost equally formation and heme alkylation may be deter-
to both faces of the 7r-bond^^^. P450 heme alkyla- mined, in part, by the regiochemistry (i.e., inner or
tion is thus a highly regio- and stereospecific outer carbon) of oxygen addition to the Tr-bond.
process. The P450-catalyzed oxidation of olefins can also
Chemical models have successfully repro- be explained by initial addition of the oxoiron
duced autocatalytic heme alkylation and have con- complex to the ir-bond to give one of the two pos-
firmed and extended the mechanistic information sible metallacyclobutane intermediates. The ratio
provided by enzymatic studies^^^"^^^. Thus, iron of epoxide formation to heme alkylation might
porphyrins can oxidize terminal olefins to species then reflect the relative proportion of the metalla-
that alkylate the porphyrin nitrogens and give the cyclobutane with the oxygen bound to the internal
same types of adducts as are obtained biologi- carbon vs that with the oxygen bound to the ter-
^^jjy262-265 jj^ ^^Q models, as in the biological minal carbon. However, the heme alkylation
process, the oxygen is added to the olefin from details, the parameters that govern partitioning
the same side as the porphyrin nitrogen, the nitro- between epoxidation and heme alkylation, and the
gen is not alkylated by epoxide or aldehyde relationship between the mechanism of heme
metabolites, and the reaction is subject to steric alkylation vs epoxide formation remain to be clar-
interference by substituents on the olefin^^^"^^"^. ified. The recent formulation by Shaik of a two-
However, in the model systems, heme alkylation state oxidation mechanism, in which spin state
can occur with disubstituted olefins, and binding pairing of electrons in the transition state deter-
of the porph3^in nitrogen to the internal carbon mines whether oxygen transfer follows a
and the hydroxyl group to the terminal carbon virtually concerted pathway or occurs stepwise,
of monosubstituted olefins occurs to a limited provides a highly attractive rationale for the
extent^^^"^^^. As a case in point, spectroscopic observation of both heme alkylation and epoxide
studies suggest that an A^-alkylated porphyrin is formation pathways in the turnover of a single
transiently formed in the oxidation of norbornene substrate^^^. The two-state hypothesis is treated in
by an iron porphyrin^^^' ^^^. The finding that for- detail in Chapter 2 and in less detail in Chapter 6.
mation of the adduct appears to be reversible led
to the suggestion that this reversibility may mask
the biological formation of secondary iV-alkyl
adducts266,269,270 Reversible 7V-alkyl adduct for-
3.3.2. Acetylenes
mation has not been generally detected with P450, P450-catalyzed oxidation of terminal acetylenes
although evidence for the reversible A/-alkylation to substituted acetic acids (Chapter 6) is more
of the heme of a CYP2E1 T303A mutant by tert- prone to result in heme alkylation than the oxida-
butyl acetylene has recently been reported^^^ On tion of terminal olefins. The structure-activity
the other hand, it has been known for some time relationships for the acetylene reaction are similar
that the catalytic oxidation of terminal olefins to those for terminal olefins, except that there are
by chloroperoxidase and H2O2 results in reversible fewer instances in which the reaction does not
iV-alkylation of its heme group, with up to 80% result in enzyme inactivation. For example, P450 is
recovery of the enzyme activity over several hours inactivated by phenylacetylene but not detectably
at 25°C2'72. by styrene^^"^, and P450 is inactivated by internal
Several mechanisms can be envisaged for acetylenes, albeit without heme adduct formation,
heme alkylation that are consistent with the exper- but not by internal olefins ^^^' ^^'^. Catalytic oxida-
imental data, none of which involves a concerted tion of the acetylenic function is required for
transfer of the oxygen to the ir-bond. Subsequent enzyme inactivation and terminal acetylenes give
to possible formation of a charge transfer complex heme adducts analogous to those obtained with
between the ferryl species and the olefin ir-bond, terminal olefins^^^' ^^^. The salient difference in
addition of the oxygen to the ir-bond could give a the adducts obtained with acetylenes and olefins
270 M.A. Correia and P.R. Ortiz de Montellano

R
y=c=o-
H

H2O

RCH2CO2H

Figure 7.16. The P450-catalyzed oxidation of a terminal acetylene partitions between formation of the ketene and
heme alkylation. Which of these events occur is determined by the carbon to which the activated oxygen is added:
addition to the internal carbon (a) results in heme alkylation, and to the terminal carbon (b) yields the ketene. In the
absence of the iron, the enol adduct tautomerizes to the ketone.

is that addition of the hydroxyl group and the metabolism of terminally deuterated phenylacety-
porphyrin nitrogen across the triple bond produces lene, indicates that the commitment to either
an enol that eventually tautomerizes to a ketone. metabolite formation or heme alkylation occurs as
A second difference is that in the adducts of pheno- the oxygen transfer step is initiated (Figure
barbital-inducible rat liver P450 QnzymQS the alky- 7.16)^^"*. The factors that determine to which triple
lation occurs almost exclusively on the nitrogen of bond carbon the oxygen is transferred, and there-
pyrrole ring A whereas linear olefins primarily fore whether heme or protein modification occurs,
alkylate the nitrogen of pyrrole ring D. A topo- are unclear. However, the observation that 1-
graphical rationale has been provided to explain ethynylpyrene and phenylacetylene inactivate
this difference in alkylation regiochemistry. CYP1A2 by, respectively, protein and heme mod-
The mechanisms proposed for P450 inactiva- ification demonstrates that the reaction regio-
tion by terminal olefins can be applied to inacti- chemistry is governed by substrate-enzyme
vation by terminal acetylenes if one keeps in mind interactions and not simply by intrinsic differ-
that all the reaction intermediates bear an addi- ences between P450 enzymes^^. Indeed, such spe-
tional double bond. Triple bond oxidation is fur- cific substrate/inactivator-active sit fit may
thermore unique in that the carbon to which the account for the relatively selective inactivation of
oxygen is initially bound is revealed by the prod- CYP3A4 by gestodene (K^ = 46 |xM, k-^^^^ = 0.4
ucts, whereas this information is lost in olefin min~', and partition ratio of ~9)^^.
epoxidation because the oxygen is bound to both To gain insight into these reactivity determi-
carbons in the epoxide metabolite. Thus, in all the nants, a series of aryl and arylalkyl acetylenes
heme adducts with acetylenes that have been suf- varying in the size and shape of the aromatic
ficiently well characterized, the oxygen is bound ring system, the placement of the carbon-carbon
to the internal carbon, whereas all the metabolites triple bond, the length of the alkyl side chains,
are derived from the ketene obtained by addition and/or the presence of a terminal hydrogen or
of the oxygen to the terminal carbon. As already methyl group have been examined as inhibitors/
mentioned, the ketene or a closely related species inactivators of human and rat liver CYPlAl,
can also inactivate the enzyme by reacting with -1A2, and -2Bl/2B2^9-83,275 j h e findings reveal
the protein^^' ^^ This difference in oxygen addi- that all these features can influence the potency,
tion regiochemistry, in conjunction with the obser- type, and selectivity of P450 inhibition.
vation of a large isotope effect on metabolite Accordingly, the arylacetylenic compounds with
formation but not heme alkylation in the the larger pyrcnQ, phenanthrene, or biphenyl rings
Inhibition of Cytochrome P450 Enzymes 271

appear to inactivate the CYPl A isoforms, whereas CYP2B1, which do not oxidize 4PBi to 2-biphenyl-
2-ethynylnaphthalene, 4-phenyl-l-butyne, 1- ylpropionic acid, are refractory to inactivation.
phenyl-1-propyne, and 5-phenyl-l-pentyne are There are other documented examples of
selective CYP2B1 inactivators^^-^^' ^^^ On the mechanism-based P450 inactivation by methyl-
other hand, the 9-ethynyl- and 9-propynylphenan- substituted (i.e., internal) acetylenes^"*' ^^. A clini-
threne isomers reversibly inhibit the CYPl A iso- cally relevant internal acetylene that potently
forms, but are among the most effective and selectively inactivates human liver CYP3A4
mechanism-based inactivators of CYP2B1/2B22^^ is the antiprogestin drug mifepristone [RU486;
The length of the alkyl side chain was an (lip,17P)-ll-[4-(dimethylamino)-phenyl]-17-
important reactivity determinant among the hydroxy-17-( 1 -propynyl)-estra-4,9-dien-3-one]
arylalkyl acetylenes^^^. Thus, while phenyl- (Figure 7.9)^^' ^^^, a drug used for medical abor-
acetylene is a reversible CYP2B1/2B2 inhibitor, tion in the first trimester of pregnancy^^^. K^ and
analogues with three or four methylene groups ^inact values of 4.7 JULM and 0.089 min~^ place
(5 -phenyl-1 -pentyne and 6-phenyl-1 -hexyne, mifepristone among the most potent CYP3A4
respectively) are among the most potent prototype inactivators^^. Although the activities of CYPIA, -
CYP2B1/2B2 inactivators. Replacement of the 2B, and -2D6 enzymes were also inhibited
terminal hydrogen with a methyl, giving disub- in vitro, this inhibition, unlike that of CYP3A4
stituted acetylenes, results in reduced CYP2B and -3A2, was reversed when mifepristone
and increased CYPl A inactivation. Thus, 2-(l- was removed by dialysis. Inactivation with
propynyl)phenanthrene, 4-ethynylbiphenyl, and [^H]mifepristone showed that the drug binds cova-
4-(l-propynyl)biphenyl are very effective inacti- lently to the CYP3A4 protein with a stoichiome-
vators of both rat liver CYPlAl and -1A2, try of 1.02 ± 0.15 mol per mol of protein^^. In
whereas l-(l-propynyl)pyrene, 2-ethynylphenan- vitro studies with CYP3A4 and -3A5, the other
threne, 3-ethynylphenanthrene, 3-(l-propynyl) major adult human liver CYP3A isoform, indicate
phenanthrene, 2-(l-propynyl)naphthalene, and that the latter, although capable of metabolizing
6-phenyl-2-hexyne are effective inactivators of the drug, is not subject to mifepristone-mediated
CYPlAl but not CYP\A2^^\ Furthermore, inactivation^^. Mifepristone may be a usefiil probe
replacement of the terminal acetylenic hydrogen with which to distinguish these two CYP3A iso-
with a methyl enhanced the mechanism-based inac- forms. The acetylenic moiety of mifepristone is
tivation of both CYPlAl and -1A2, or converted a thought to also be activated to a ketene, although
reversible inhibitor into an effective inactivator, as the expected propionic acid metabolites have
exemplified by 1-ethynylpyrene and l-(l-propy- not been detected with either enzyme. However,
nyl)pyrene, 2-ethynylphenanthrene and 2-propy- LC-MS of mifepristone metabolites revealed that
nylphenanthrene, 3-ethynylphenanthrene and although both enzymes generate the NJ\f'-
3-propynylphenanthrene, and 6-phenyl-1-hexyne didemethylated and A^-monodemethylated prod-
and 6-phenyl-2-hexyne^^^. Indeed, 2-pro- ucts, only CYP3A4 hydroxylates the terminal
pynylphenanthrene and 4-propynylbiphenyl (4PBi) methyl group. Thus, the susceptibility of CYP3A4
are among the more selective inhibitors of rat but not CYP3A5 to inactivation may be due to the
liver CYPIA and human liver CYP1A2 ability of the first but not the second to oxidize the
enz3mies. acetylenic moiety of mifepristone^^. Although not
In contrast, 4PBi fails to inactivate human liver considered in the publications, it is very possible
microsomal CYP2E1, -2C9/10, -3A4 or -2C19. that the inactivation observed with mifepristone
The identification of 2-biphenylylpropionic acid does not reflect oxidation of the triple bond at all
from the CYPlAl- and -lA2-catalyzed metabo- but rather oxidation of the terminal methyl to an
lism of 4PBi links this mechanism-based inactiva- aldehyde, giving an a,p-unsaturated aldehyde that
tion with that of terminal acetylenes, as it involves adds to the protein as a Michael acceptor.
a 1,2-shift of the terminal methyl to give a ketene Not surprisingly, the acetylenic fimction has
intermediate^^^. The importance of the 1,2 methyl been exploited in the design and synthesis of P450
shift and the resulting ketene in P450 inactivation isoform-selective or -specific irreversible
by internal acetylenes such as 4PBi is underscored inhibitors, including inhibitors of P450g^^, aro-
by the finding that P450 enzymes such as matase, prostaglandin w-hydroxylase-^^^, and the
272 M.A. Correia and P.R. Ortiz de Montellano

P450 enzymes that oxidize saturated fatty acids, mechanism of analogues that alkylate the heme
arachidonic acid, and leukotriene B^ (Section 5.5). nitrogen is understood better. The adducts consist
It may also play a role in the alterations of oxidative
of protoporphyrin IX with the 4-alkyl group of the
metabolism observed in individuals treated with parent substrate covalently attached to one of its
ethynyl sterols such as gestodene and 17a-EE^^~^^. nitrogen atoms (Figure 7.17)^^^' ^^^' 2^^' ^89
Strategies to convert selective P450 substrates Different nitrogens are alkylated in different P450
to suicide inactivators by the incorporation of a enzymes-^^^' ^^^, and the dihydropyridines cause
suitable activatable function at the position oxi- isoform-selective inactivation^^^' ^^^. The catalytic
dized are not always successful. For instance, the turnover of 4-alkyl-1,4-dihydropyridines thus
introduction of an acetylenic moiety into the leads to transfer of the 4-alkyl group to a nitrogen
chemical template A/-(3,5-dichloro-4-pyridyl)- of the heme. The following observations further
3-(cyclopentyloxy)-4-methoxybenzamide elucidate the nature of the enzyme inactivation:
(DCMB), a CYP2B6 functional marker, to yield (a) the dihydropyridines are oxidized to the
A^-(3,5-dichloro-4-pyridyl)-4-methoxy-3-(prop-2- pyridines with partial loss of 4-alkyl but not 4-aryl
ynyloxy)benzamide gave a mechanism-based groups^^^' 2^^, (b) the A/-methyl or A^-ethyl
agent that, based on a correlation of activity and derivatives of 4-alkyldihydropyridines still inacti-
ferrous-CO chromophore loss, probably inacti- vate P450 enzymes but inactivation may follow
vated CYP2B6 via heme modification^^^. How- A^-dealkylation because with those substrates
ever, this inactivation was not very selective as A^-dealkylation is faster than dihydropyridine
other human liver CYP2C isoforms were also aromatization^^^, (c) no primary isotope effect is
inactivated, indicating that the catalytic selectivityobserved on enzyme inactivation when the hydro-
for CYP2B6 resides in the 0-alkyl chain of the gen at position 4 is replaced by deuterium^^^,
parent DCMB molecule. (d) the heme adducts that are formed are chiral
and therefore are generated within the active
site^^^, and (e) free radicals have been detected
3.3.3. Dihydropyridines and with a spin trap in incubations of a 4-ethyldihy-
dropyridine with hepatic microsomes^^^. However,
Dihydroquinolines
studies with deferoxamine-washed microsomes
The administration of 3,5-bis(carbethoxy)- suggest metal-catalyzed oxidation of the dihy-
2,4,6-trimethyl-1,4-dihydropyridine (DDC)280-284 dropyridine accounts for most if not all of the spin-
perturbs heme biosynthesis and causes a loss of trapped radicaF^^. In any case, as neither GSH nor
hepatic P450 content, both of which have been the radical trap prevent enzyme inactivation, the
traced to iV-methylation of the P450 prosthetic radicals detected in the medium appear not to be
heme^^^~2^^. Substitution of the dihydropyridine at involved in heme alkylation. Conversely, if radicals
position 4 with a primary, unconjugated moiety are formed within the active site, they are not read-
(methyl, ethyl, propyl, sec-buty\, nonyl), but not an ily detected in the medium. In view of these
aryl (phenyl), secondary (isopropyl), or conjugated results, it is highly likely that electron abstraction
(benzyl) group results in A^-alkylation of the from the dihydropyridine produces a radical cation
heme^^^' 291-294 4-Aryl-substituted dihydropy- that aromatizes either by extruding the 4-alkyl
ridines do not inactivate the enzyme, whereas group as a radical or by directly transferring the
those bearing secondary or conjugated substituents alkyl group to the heme. Although the 4-alkyl
inactivate the QYoyme but do not yield detectable group may be directly trapped by the porphyrin
A^-alkyl heme adducts^^^"^^"^. Dihydropyridines nitrogen, model studies suggest that it first adds to
with simple 4-alkyl groups 7V-alkylate the heme of the iron to form an alkyl-iron complex from which
certain P450 isoforms, but inactivation of others it migrates to the porphyrin nitrogen. The absence
occurs by a mechanism that appears to involve of detectable heme adducts in the P450 inactiva-
heme degradation to fragments that irreversibly tion by the 4-isopropyl and 4-benzyl analogs is
modify the protein (Section 3.4). consistent with such a mechanism, not only
The mechanisms of enzyme inactivation and because the iron-nitrogen shifl is sensitive to steric
heme destruction by analogs that do not give iden- efifects^^^, but also because the more oxidizable
tifiable heme adducts remain unclear, but the secondary or benzylic moieties may be converted
Inhibition of Cytochrome P450 Enzymes 273

H R H R H(R)
^t^2C^jyC/C02Et Et02C^>C/C02Et Et02C^^X^C02Et

CUf
1 1 ^N "CH3 cnf
X I —^ XT
N "CH3 N CH3
H H

CH3 (R)

Figure 7.17. Oxidation of 4-alkyl-l,4-dihydropyridines and 2,2-dialkyl-l,2-dihydroquinolines is proposed to


yield cation radical intermediates that aromatize by either losing a second electron and a proton or an alkyl radical
that adds to a nitrogen of the prosthetic heme group.

to the corresponding cations by electron loss to the accompanied by the generation of a heme adduct
iron in preference to undergoing the oxidative identified as A/-(2-phenylethyl)protoporphyrin IX
iron-nitrogen shift. (Figure 7.18)'^^. The generation of products
The precedent set by the P450-catalyzed oxi- in a microsomal system that implicate the 2-
dation of dihydropyridines to radical cations that phenylethyl radical as a central MI suggests a role
aromatize by radical extrusion has led to the for the 2-phenylethyl radical in this enzyme inac-
observation of comparable processes in related tivation'^'. Spin-trapping experiments confirm
structures. Thus, 2,2-dialkyl-l,2-dihydroquino- that the 2-phenylethyl radical is generated in the
lines are also oxidized by P450 enzymes to incubations but the bulk of the radical that is spin
species that iV-alkylate the heme and inactivate trapped is formed by transition metal, rather than
the enzymes. In these reactions, the 2-alkyl P450-catalyzed reactions'^^' ^^^. It appears from
substituent of the dihydroquinoline is bound to these results that phenelzine is converted to the 2-
a nitrogen of protoporphyrin IX, presumably by phenylethyl radical within the P450 active site
a mechanism similar to that proposed for the where it is captured by the heme to give the
4-alkyldihydropyridines (Figure 7.17)^^^. iV-(2-phenylethyl) adduct (Figure 7.18). The 2-
phenylethyl radical could react directly with the
porphyrin nitrogens, but by analogy to the reactions
of hemoproteins with arylhydrazines (see below) it
3.3.4. Alkyl- and Arylhydrazines and
is likely that the alkyl radical is trapped by reaction
Hydrazones
with the heme iron to give an unstable alkyl-
The P450-destructive mechanism of iron complex that subsequently rearranges to the
phenelzine, an alkylhydrazine, is relatively well isolated A/-alkyl heme adduct.
defined. It causes an approximately equimolar A complex with an absorbance maximum
loss of enzyme and heme when incubated with at 480 nm is generated in the reaction of P450
hepatic microsomes'^^ and these losses are with phenylhydrazines and A^-phenylhydrazones.
274 M.A. Correia and P.R. Ortiz de Montellano

K3Fe(CN)6
or A

PhN=NH

N- =;N PhCH2CH2N=:NH
,Fe-
N- -N

PhCH2CH2.

Figure 7.18. The oxidation of phenelzine (PhCH2CH2NHNH2) and phenylhydrazine (PhNHNH2) by P450
produces carbon radical products that bind to the prosthetic heme group. In the case of phenylhydrazine, the phenyl
radical binds first to the iron atom to give a complex that subsequently rearranges under oxidative conditions to the
yV-phenyl adduct. It is not known if phenelzine initially forms a similar but less stable carbon-iron intermediate.

Formation of this complex inactivates the enzyme migration is sensitive to steric effects because the
and precedes irreversible destruction of its aryl moiety in aryl-iron complexes obtained from
prosthetic heme^^^"^^^. The reactions of myoglo- ortho-suhsXitaiQd phenylhydrazines does not
bin, hemoglobin, and catalase with phenylhy- undergo the oxidative shift^^^. Arylhydrazines,
drazine yield a similar complex, in these cases including phenyl-, 2-naphthyl-, and /7-biphenyl-
with an absorbance maximum at —430 nm due hydrazine, are known to be P450 substrates^^^.
to the difference between the proximal thiolate Migration of the phenyl group from the iron to
and histidine ligation^^^~^^^. As revealed by X-ray the porphyrin nitrogens can be induced to occur
crystallography, the myoglobin and CYPlOl within the undenatured active site by the addition
(P450^^^) structures are a-aryl-iron complexes of ferricyanide to the intact P450 com-
with the phenyl group bound end-on to the iron plexes^'^~^^^. The distribution of the four iV-aryl
(Figure 7.18)^^^' ^'^. Extraction of the heme com- protoporphyrin IX regioisomers produced by
plex from CYPlOl under oxidative conditions, this method reflects the active-site topology
yields a roughly equal mixture of the four possible and varies widely from enzyme to enzyme.
A^-phenylprotoporphyrin IX regioisomers, as The migration of aryl groups within the active
found with the complexes from myoglobin, hemo- sites of P450 enzymes provides a tool for the
globin, and catalase^^^' ^^^. The intact phenyl-iron determination of their active-site topology
heme complex, which has been characterized by because the regioselectivity of the migration is
absorption and NMR spectroscopy, is obtained if controlled by the degree to which the active site
the prosthetic group is extracted under anaerobic is sterically unencumbered above each of the
conditions^^^' ^^^. Exposure of the anaerobically four pyrrole ring nitrogens^ ^^~^^^. The in situ
extracted phenyl-iron heme complex to oxygen or shift of the aryl moiety from the iron to the por-
other oxidizing agents under acidic conditions phyrin nitrogens occurs readily with most, but
results in migration of the phenyl from the iron to not all, of the P450 iron-aryl complexes, but
the porphyrin nitrogens (Figure 7.18)^^^. This does not occur with the complexes of proteins
Inhibition of Cytochrome P450 Enzymes 275

such as myoglobin that have an imidazole as the two nitrogens, generating an N,N-bhdgQd species
fifth iron ligand. that autooxidizes to the isolated bridged por-
phyrin, or may first bind to the iron and a nitrogen
The likelihood that alkyl radicals, like their aryl
counterparts, bind to the iron before shifting to theof the heme and subsequently rearrange to the
nitrogen is supported by the observation that the A^,A^-bridged species (Figure 7.19). Introduction of
type II complexes formed between alkyldiazenes small substituents on the phenyl ring or on the
and P450 in the absence of oxygen are converted, exocyclic nitrogen of ABT, or replacement of the
in the presence of limited amounts of oxygen, phenyl fi'amework with alternative aryl moieties,
to complexes with an absorption maximum at does not impair destructive activity^^^"^^^. It is not
~480 nm characteristic of iron-carbon a-bonded known whether the oxidation of ABT to benzyne
complexes^^^' ^^^. Furthermore, alkyl diazene-iron proceeds via hydroxylation of the exocyclic nitro-
tetraphenylporphyrin complexes can be prepared gen or electron abstraction to give a radical or rad-
under anaerobic conditions^^"^. However, the ical cation (Figure 7.20), but a notable similarity
alkyl-iron complexes are much less stable and less exists between the activation mechanism proposed
well characterized than the aryl-iron complexes for ABT and other 1,1-disubstituted hydrazines
and their involvement in heme A^-alkylation (Figure 7.14).
reactions remains to be demonstrated. ABT inactivates a wide variety of P450
In addition, some aryl hydrazines, notably dihy- enzymes without detectable toxic eflfects^^^"^^^.
dralazine, also inactivate P450 via protein modifi- Thus, ABT administration to guinea-pigs inacti-
cation in a mechanism-based process^^^. Thus, vates both adrenal steroidogenic- and xenobiotic-
dihydralazine has been shown to inactivate rat liver metabolizing P450 isoforms^^^' ^^^. The inacti-
microsomal CYP1A2, -2C11 and -3A, but not vation of steroidogenic enzymes is apparently
CYP2B1 or -lAl^^^, and also inactivates human indirect and due to the generation of an extra-
liver CYP1A2 and -3A4, but not CYP2C9326 jy^^^ adrenal ABT metabolite(s), as these guinea-pig
inactivation appears to involve irreversible binding adrenal P450 isoforms (unlike their hepatic and
of a reactive metabolite to the P450 protein in a adrenal xenobiotic metabolizing counterparts) are
process that is not affected by co-incubation with not susceptible to direct ABT-mediated inactiva-
GSH (5 mM)^^^. The generation of irreversible tion in reconstituted in vitro systems^^^' ^^^. P450
dihydralazine-protein adducts in vivo and their isoform and tissue selectivity is conveyed by plac-
subsequent immunoproteasomal processing into ing substituents on the exocyclic nitrogen of the
antigenic P450 peptides could possibly account for aminotriazoleftinction^^^'^^^' 339-341 Furthermore,
the detection of CYPlA2-reactive anti-liver micro- it is noteworthy that certain ABT analogs [A^-ben-
somal (anti-LM) autoantibodies, in the sera of zyl-, 7V-(a-methylbenzyl)-, or iV-(a-ethylbenzyl)-
patients with dihydralazine-induced immunoaller- 1-ABT] also inactivate phenobarbital-inducible
gic hepatitis^^^' ^^^. hepatic P450 enzymes (other than the CYP2B4/
CYP2B1 orthologs) via the formation of MI com-
plexes rather than by heme modification^^^
3.3.5. Other N-N Functions
Overall, ABT is a highly effective agent for the in
The P450 heme is iV-alkylated or 7V-arylated vivo inactivation of a variety of P450 enzymes in
by reactive intermediates formed when it oxidizes plants343,344^ insects345, and animals334-342,346-348
1-aminoaryltriazoles, 2,3-bis(carbethoxy)-2,3- Cyclobutadiene, which can be envisioned as a
diazabicyclo[2.2.0]hex-5-ene, and the sydnones. rectangular structure with a singlet electronic state
The oxidation of 1-aminobenzotriazole (ABT) or a square structure with a triplet electronic
by chemical reagents yields benzyne, an exceed- state, is formed upon chemical oxidation of 2,3-
ingly reactive species, and two molecules of nitro- diazabicyclo[2.2.0]hex-5-ene34^. Bis(carbethoxy)-
gen^^^. The finding that benzyne, or its equivalent, 2,3-diazabicyclo-[2.2.0]hex-5-ene (DDBCH), a
is bound across two of the nitrogens of the pros- precursor of the above compound, is a mecha-
thetic heme isolated fi-om inactivated P450 nism-based irreversible inhibitor of P450 that
enzymes suggests that the enzyme-catalyzed oxi- exploits the basic reactivity of the parent bicyclic
dation of ABT follows the same reaction trajec- system^^^. The bis(carbethoxy) derivative was
^Qj^330,331 jY^Q benzyne may add directly to the employed for the enzymatic studies because the
276 M.A. Correia and P.R. Ortiz de Montellano

/ \

/ \
H2N—N^ ^ N
N

N-jl N -"-° N N
Fe^III
-2N, ^: ^-
N N

Figure 7.19. Two alternative mechanisms for addition of the benzyne released from 1-aminobenzotriazole (ABT)
to the heme. The heme porphyrin framework is represented by a square of nitrogens, each of which represents one
of the four nitrogens of the porphyrin.

-^m+3 N
N [Fe=0]^ [Fe-OH]1+3 N
N • N
N N
I
NH2 HN NHOH

[Fe-OH]-'^

N
N
k H2O
]

"7^ N
I
2N2 :N:

Figure 7.20. Mechanistic alternatives for the P450-catalyzed generation of benzyne from 1-aminobenzotriazole.

parent bicylic hydrazine autooxidizes too readily internal olefins and acetylenes to detectably alky-
to be biologically useful. The heme of the P450 late the prosthetic heme suggests, in fact, that sec-
enzyme is converted into the 7V-2-cyclobutenyl ondary carbons are generally too sterically
derivative during the inactivation reaction encumbered to react with the heme moiety.
(Figure 7.21). The secondary, allylic, carbon- Although the precise nature of the reactive species
nitrogen bond in this adduct makes it much less remains undefined, the generation of the 2-
stable than other adducts, which bear the primary, cyclobutenyl adduct implies the involvement of
unactivated, 7V-alkyl groups. The failure of cyclobutadiene itself, or of a closely related
Inhibition of Cytochrome P450 Enzymes 277

^COoEt
u II
C02Et

XOoEt
I
C02Et C02Et

Figure 7.21. Possible mechanisms for the oxidative generation of cyclobutadienoid species that alkylate the
prosthetic heme group of P450. The heme of P450 is abbreviated as indicated in Figure 7.19.

species, in heme alkylation. The structure of the both A^-(2-phenylethyl)- and 7V-(2-phenylethenyl)
adduct is readily rationalized if cyclobutadiene adds protoporphyrin IX^^^. These results are most
to a porphyrin nitrogen to give a transient, probably consistent with oxidation of the sydnone to the
anionic intermediate, that is neutralized by a proton (2-phenylethyl)diazonium cation that reacts with
from the medium. The transient intermediate could the heme in two different ways. In one mecha-
be stabilized by formation of a carbon-iron bond nism, deprotonation of the diazonium intermedi-
with the carbon that is eventually protonated. ate, as discussed above, results in a carbene-like
Electron abstraction from DDBCH can lead to the addition. In the absence of a p-leaving group,
observed adduct by pathways that depend on the resulting carbanion intermediate is oxidized to
whether the cyclobutadiene is generated as a neu- a cation that is deprotonated to introduce the dou-
tral, cationic, or anionic species (Figure 7.21). ble bond into the N-a\ky\ group^^^. In the second
The accumulation of a fluorescent hepatic mechanism, reduction of the diazo intermediate
pigment in dogs and rats administered a sydnone prior to deprotonation yields a phenyldiazenyl rad-
derivatives^ ^ led to the discovery that the sydnone ical that is trapped by a porph3Tin nitrogen atom
is catal)^ically activated by P450 enzymes to a in the same manner as the 2-phenylethyl radical
species that alkylates the prosthetic heme^^^. The produced by the oxidation of phenylethyldiazene
heme adduct isolated from rats treated with the (Figure 7.18)^^^. The validity of this mechanism is
sydnone has been identified as A/-vinylprotopor- strengthened by the fact that the 2-phenylethyl
phyrin IX (Figure 7.22). This finding suggests that adduct obtained from the 1,1-dideuterated sub-
the sydnone is first activated by hydroxylation of strate retains both deuteriums and therefore
the electron-rich zwitterionic carbon, followed by arises by a mechanism that does not involve
ring opening and elimination of the carboxylic deprotonation of the diazo intermediate^^^.
fragment to give the diazo species (Figure 7.22). The in vivo administration of diethylni-
A similar mechanism explains the oxidation of trosamine (Et2N-N=0) to mice reportedly gener-
sydnones by simple chemical reagents^^^. The dia- ates an alkylated porphyrin that was tentatively
zoalkane then reacts with the heme, possibly via identified by mass spectrometry as iV-(2-hydroxy-
an initial carbene complex, to give a nitrogen-iron ethyl)protoporphyrin IX^^^. In vitro studies with
bridged intermediate. The formation of just such a rabbit liver microsomes and purified P450
bridged nitrogen-iron complex has been observed enzymes have independently shown that P450 oxi-
in model porphyrin systems^^"^' ^^^. The negative dizes diethylnitrosamine to ethylene^^^. Although
charge on the carbon in the bridged intermediate not actually demonstrated, the proposed N-(2-
finally eliminates the thiophenyl moiety and gen- hydroxyethyl) porphyrin adduct is likely to be
erates the A^-vinyl adduct^^^. The mechanism is derived from P450 enzymes inactivated by the
fiirther clarified by the fact that P450 inactivation ethylene metabolically generated from diethylni-
by 3-(2-phenylethyl)-4-methylsydnone produces trosamine (see Section 3.3.1).
278 M.A. Correia and P.R. Ortiz de Montellano

Me. QH R.
RCHov. + RCHo
~N. 1
[Fe=0]3-' C>

N- —N R = CH2SPh N-/-^N

N N N^^ N

R = CHsPh R

N- -N
Fe
N- -N

Figure 7.22. Mechanism proposed for the oxidation of sydnones to reactive intermediates that add to
the prosthetic heme group of P450. The structures of the A^-alkylporphyrins isolated from rats treated with the
3-(2-phenykhioethyl)- and 3-(2-phenylethyl) sydnones are shown.

hepatic porphyrias, has been reported to cause the


3.3.6. Other Functionalities
catalysis-dependent destruction of P450 and the
The herbicide l-[4-(3-acetyl-2,4,6-trimethyl- formation of a green pigment^^^. In order to char-
phenyl)-2,6-cyclohexanedionyl]-0-ethyl propi- acterize the pigment, the A^-alkyl group was trans-
onaldehyde oxime (ATMP) (Figure 7.23) causes ferred to an amine in a copper-mediated reaction
hepatic protoporphyria in the mouse, albeit not and the alkyl amine was analyzed by mass
in other species, and this derangement of the spectrometry. The results suggested that N-
porphyrin biosynthetic pathway appears to be methylprotoporphyrin IX was a minor product and
linked to P450 inactivation via a heme modifica- a porphyrin with most of the griseofulvin struc-
tion mechanism^^^. A pigment tentatively identi- ture bound to the nitrogen of pyrrole ring C of the
fied as A/-methylprotoporphyrin IX by HPLC heme was the major product^^^"^^^. The NMR
analysis has been isolated from mice treated spectrum of the latter adduct confirms that it is
with ATMP, and its formation has been shown an iV-alkylated porphyrin with the griseofulvin
to be decreased by pretreatment with the P450 structure attached to the nitrogen of either pyrrole
inhibitors SKF 525A and piperonyl butoxide, con- ring C or D^^"^. The process appears to be species-
sistent with a P450-dependent process^^^. specific because, at most, only very low levels of
Replacement of the ethyl on the oxime carbon pigment have been observed with rats or chicken
with a propyl group suppresses the porphyrino- embryos^^^"^^^. It is difficult to postulate a mech-
genic activity of the analog, presumably by pre- anism for the formation of TV-methyl heme from
venting heme alkylation. Nothing further is griseofulvin, particularly as an identical pigment
known about the mechanism by which ATMP is reportedly present in lower amounts in the
mediates the P450-dependent formation of an livers of control mice^^^' ^^^. The presence of an
A^-alkyl (possibly iV-methyl) protoporphyrin IX endogenous A/-alkylporphyrin in mice would
adduct. clearly be of high interest, but more definitive evi-
Treatment of mice with griseofulvin dence on its origin is required before the signifi-
(Figure 7.23), an agent long known to cause cance of these results can be evaluated. It is to be
Inhibition of Cytochrome P450 Enzymes 279

CH3O o OCH3

CH3
CH3O
CI CH3

ATMP Griseofulvin

Figure 7.23. The structures of two compounds that cause P450 inactivation. In the case of griseofulvin, the
inactivation appears to involve heme A^-alkylation, but the detailed mechanism of inactivation by ATMP is not known.

CH
I I >^CH3 CH2OH

[Fe=0]^

CH3 ^ ''^'^\^^ N
I |[ y-CH2-X-Protein

Figure 7.24. The oxidation of furafylline is proposed to yield a chemically reactive intermediate that binds to
a protein residue (denoted by Protein-X).

noted that a mechanism is also not obvious for molecules oxidized per enzyme molecule inacti-
attachment of the griseofulvin structure to the por- vated^^^. Oxidation of the C-8 methyl in the inac-
phyrin nitrogen via a mechanism-based process. tivation process is suggested by the fact that
Furafylline (Figure 7.24), a potent selective deuterium substitution on the C-8 methyl gives an
inhibitor of human CYP1A2, causes time- and isotope effect of —2.0 on k^^^^^ but not on K^, and
NADPH-dependent inactivation of this enz- the observation that inactivation activity is sup-
y^e368,369 CYPIAI, -2A6, -2B6, -2C9, -2C19, pressed when the methyl is removed^^^.
-2D6, -3A4, and -2E1 are not similarly inactivated, Recent mechanistic studies confirm that the
although the evidence suggests that other enzymes oxidation of the 8-methyl of furafylline yields the
can be inhibited^^^. Clinical studies confirm that corresponding 8'-carbinols and/or results in cova-
furafylline can almost completely suppress in vivo lent binding of the agent to the CYP1A2 protein
human CYP1A2 function^^^' ^'^K This loss of func- (Figure 7.24)^^^. This result suggests that the oxi-
tion is paralleled by a similar loss of the heme dation of furafylline converts it to a two-electron
chromophore, with K^ = 23 fjiM and A:.^^^^ = 0.87 oxidized electrophilic intermediate such as
min~^ and a partition ratio of 3-6 substrate the exocyclic 8-methyleneimidazolenine or
280 M.A. Correia and P.R. Ortiz de Montellano

imidazomethide, and that this intermediate is the inactivation is not associated with covalent
trapped by a nucleophilic side chain within the binding of the radiolabeled agent to the protein,
CYP1A2 active site^^^. Consistent with this, nei- the formation of P420, or loss of the heme^^^.
ther GSH nor cyanide significantly impair either Similarly, the inactivation of CYP3A4 by delavir-
covalent binding or inactivation. Although the dine (1 - [3 - [(1 -methylethy l)amino] -2-pyridinyl] -4-
identity of the alkylated active-site residue [[5-[(methylsulfonyl)amino]-lH-indol-2-yl]
remains undetermined, studies of furafylline carbonyl]-piperazine), a potent inhibitor of HIV-1
docked in a homology model of the CYP1A2 reverse transcriptase, also reportedly adheres to
active site suggest that the interactions of the furan the criteria for a mechanism-based inactivation,
moiety with the protein indeed favor oxidation of but the chemical details of the process remain to
the 8-methyl group^^^"^'^'^. Ancillary evidence that be elucidated^^^.
the 8-methyl and not the furan moiety of
furafylline is critical for CYP1A2 inactivation is
provided by comparable findings {h^^ 0.89 3.4. Modification of the P450
min~\ partition ratio = 7.6, equivalent covalent
Protein by Heme Fragments
binding to CYP1A2) with cyclohexylline, in
which the furan is replaced by a cyclohexyP^^. During the catalytic oxidation of some
Supportive evidence for the proposal that the imi- substrates, certain P450 enzymes (i.e. CYP3A,
dazole A^^-hydrogen is lost during CYPl A2 inacti- CYP2E) undergo a mechanism-based inactiva-
vation is provided by the inactivity of the tion process in which fragments of the heme are
corresponding A^^-methylated analogs as CYPl A2 irreversibly bound to the protein. Examples of
inactivators^^^. such inactivators include CCl^^^^"^^^, spironolac-
Analogous P450-catalyzed dehydrogenations tone (see Figure 7.4)^^' ^^, 3,5-dicarbethoxy-2,6-
have been invoked in the mechanism-based inacti- dimethyl-4-ethyl-1,4-dihydropyridine (DDEP),
vation of select P450 isoforms by the pulmonary and its 4-isopropyl and 4-isobutyl analogs (see
toxin 3-methylindole^^^, the anticonvulsant val- also Figure 7.17)^^^"^^^. The features that predis-
proic acid (see Chapter 6)^^^, and the leukotriene pose an enzyme to cross-linking of heme frag-
inhibitor zafirlukast (Figure 7.25)^^^. ments to the protein remain unclear. Studies with
The mechanism of P450 inactivation is unclear the above substrates suggest that the generation of
for some classes of agents. As an example, free radical products is important, but per se is not
CYP2E1 is inactivated by 3-amino-l,2,4-triazole sufficient because not all the P450 enzymes that
in a time- and NADPH-dependent manner but produce radicals undergo such inactivation. Thus,

ai
Zafirlukast

Figure 7.25. Structures of three agents that cause irreversible inactivation of cytochrome P450.
Inhibition of Cytochrome P450 Enzymes 281

during the one-electron oxidation of DDEP, attachment of the heme via one of its vinyl groups
CYP2C6 and -2C11 undergo heme A^-ethylation, to His93^^'*' ^^^. In both of these processes, as well
whereas the CYP3A enzymes predominantly as during the hemoglobin-mediated reduction of
incur cross-linking of heme fragments to the pro- CBrCl3^^^ the cross-linked heme retains its Soret
tein^^^' ^^^. Furthermore, studies with DDEP and absorption maximum (at —405 nm) and is thus
its analogs with a secondary carbon attached to bound to the protein without substantial structural
the 4-position (4-isopropyl and 4-isobutyl) reveal disruption of its chromophore. Furthermore, 7-
that the reaction outcome is largely dictated by the meso alkylated heme adducts without appreciable
P450 active-site structure rather than by the inabil- heme-protein cross-linking are observed during
ity of the inactivator to iV-alkylate the heme^^^. the myoglobin-mediated oxidative metabolism of
Thus, DDEP, which can form iV-ethyl porphyrins, alkylhydrazines^^^. In contrast, the cross-linking
and the 4-isopropyl and 4-isobutyl analogs that of heme fragments to protein observed in the P450
cannot, exhibit comparable extents of heme reactions with H2O2, cumene hydroperoxide,
destruction and heme fragment cross-linking. DDEP, and spironolactone involves complete loss
Cross-linking of heme fragments to the protein is of the heme chromophore and therefore major
thus not merely the result of a defective or ineffi- structural disruption of the heme skeleton^^' ^^^'
cient heme A/-alkylation process. The fact that 386-391 jjjjg heme degradation also occurs if the
spironolactone inactivates hepatic CYP3A cumene hydroperoxide-mediated inactivation is
enzymes via heme fragment cross-linking^^' ^^ but carried out under anaerobic conditions, albeit at
inactivates adrenal P450 enzymes by direct pro- a considerably slower rate, implying a role for
tein modification^^ frirther demonstrates that the molecular O2 in this process^^^' ^^^.
binding of heme fragments to the protein is iso- Attempts to elucidate this process have focused
form-specific. Conceivably, the propensity of the on cumene hydroperoxide-inactivated [^"^CJ-heme-
CYP3A enzymes to undergo heme fragment labeled CYP3A23, -3A4, and -261^89, 390
cross-linking is related to their unusually large Proteolytic digestion with lysyl endopeptidase-C
and, given their ability to accommodate large sub- of the [^"^CJ-heme-modified P450enzymes, cou-
strates such as cyclosporin, macrolide antibiotics, pled with HPLC-mapping of the [^"^CJ-heme-
and FK506, as well as small substrates such as modified peptides, Tricine-SDS-PAGE, elec-
DDEP, highly flexible active sites. Their active troblotting, microEdman degradation/amino
sites may therefore exhibit an unusual degree of acid sequencing, and electrospray ionization mass
substrate mobility and/or water content (Chapter spectrometry (ESIMS), have located the specific
10). Regardless of the mechanism, it is clear that sites modified by the heme fragments within the
CYP3A active sites are particularly susceptible to active sites of the P450 enzymes^^^' ^^^. Speci-
inactivation by heme fragment cross-linking. A fically, the labeled peptide in CYP3A23 encom-
related process is mediated by peroxides such as passes residues 287-330, and in CYP2B1 residues
H2O2 and cumene hydroperoxide that partially 434^66. Sequence alignment of CYP3A23
degrade the prosthetic heme to soluble monopyr- and -2B1 with bacterial CYPlOl, -102,-107 and
role and dipyrrole fragments^^^' 386-388 ^^ these -108 reveal that the [^'^CJ-heme-fragment-
reactions, the bulk of the heme is fragmented to modified peptide in CYP3A23 corresponds to the
products that irreversibly bind to the pro- bacterial I-helix^^^"^^3 j ^ i s domain contains the
tein382-39i. conserved Thr, which in the crystal structure of
Myoglobin and hemoglobin have been CYPlOl is known to interact both with the sub-
employed as models in efforts to elucidate this strate and the heme-bound O2 and to be part of the
unusual heme degradation process, but it now active site (Chapter 3)^^^^^^. On the other hand,
appears that these hemoproteins are not good the labeled CYP2B1 region corresponds to the
models for the P450 reaction392-395 j^ie Rfi^' bacterial L-helix that provides the conserved
mediated oxidation of myoglobin results in the Cys thiolate ligand, and thus is also within the
covalent binding of its heme via either the a or active site. However, until recently, the structure
^-meso carbon or one of the vinyl groups to of the attached heme-derived fragments remained
Tyrl03^^^. In contrast, the reduction of CCI4 uncharacterized, largely because of their highly
or CBrClg by myoglobin results in covalent labile nature under the experimental conditions
282 M.A. Correia and P.R. Ortiz de Montellano

required for isolation and structural analysis of heme-derived species appear to irreversibly mod-
the modified P450 peptides. ify the CYP3A proteins.
Optimization of the methodology combined The chemical nature of the heme fragment-
with the larger peptide amounts available through protein adduct remains to be elucidated. In princi-
the use of recombinant [^"^CJ-heme-labeled ple, it could entail a Schiff-base between a protein
CYP3A4, enabled the structural characterization NH2-group and the formyl group of a hydroxy-
not only of the CYP3A4 peptide targets, but dipyrrolic fragment. Alternatively, it could involve
also of the protein modifying heme-fragments^^^. attack by a suitable nucleophilic protein moiety on
The combined structural analyses identified three the 2-formylated dipyrrole. The possibility of a
major heme-modified CYP3A4 peptides com- protein adduct with the heme vinyl also exists,
prised of residues 354-371, 372-386, and even though no vinyl-modified dipyrrolic species
429-450. Sequence alignments and homology have been detected in model heme degradation
modeling of CYP3A4 reveal that the 354-371 and systems.
429-450 peptides correspond to the K-region Both in vivo and in vitro, cross-linking of heme
and helix L/Cys region respectively, of P450. fragments to the CYP2E1 and -3 A proteins targets
Several residues in these peptides are within 5 A them for proteasomal degradation by the 20S
of the heme and thus within striking distance. or ubiquitin-dependent 26S species^^^' ^01-405 ^ ^ ^
Differential LC-ESMS analyses of the native and basal levels of microsomal heme fragment-cross-
heme-modified peptide fragments provided linked P450 proteins detected after [^'^C]labeling
molecular masses of «302, 314, and 197 for the of the P450 heme moiety in vivo indicate that
heme-modifying species, corresponding to the P450 heme-modification probably occurs physio-
deformylated and formylated A-D/B-C ring logically, possibly as a result of futile oxidative
dipyrroles and the monopyrrole 2-formyl hema- cycling of the enzymes. Furthermore, the extent of
tinic acid^^^. The precise amino acid residues this cross-linking is increased considerably after
modified in these peptides remain to be identified. CYP3A is induced by dexamethasone and pheno-
Nevertheless, these findings suggest that the per- barbitaP^^'"^^^. Since this endogenous post-transla-
oxidative inactivation of CYP3A4 (and presum- tional modification targets the P450 proteins for
ably other P450 enzymes) involves rupture of its proteolytic degradation, it could serve as a deter-
tetrapyrrolic skeleton along its a-7 and/or (3-8 minant of their normal physiological turnover. Not
axes to yield reactive heme fragments that modify surprisingly, suppression of P450 futile oxidative
residues in their immediate proximity. cycling by blocking the P450 heme iron with
It is instructive that in this process, HCOOH TAO or isosafrole, or interrupting the electron
(rather than CO) is the major product, and that flow through chemical or genetic impairment of
together with minor amounts of CO and CO2, P450 reductase, results in protein stabilization
it stoichiometrically accounts for the oxidative and consequent "induction" of CYP1A2, -2E1,
loss of two heme me^o-carbons^^^' ^^^' 397-399 j ^ and -3A246-249^
contrast, peroxidative heme degradation in model
systems yields considerably larger quantities of
soluble dipyrrolic species [hydroxylated and nonhy- 3.5- Other Modes of P450
droxylated propentdyopents and HCOOH] as major Heme Degradation and
products^^^' 397^00 Approximately 15^(^20% of
Protein Denaturation
the prosthetic heme loss after NADPH-induced
oxidative uncoupling can be traced to soluble The inactivation of CYP2B4 by aldehydes such
mono- and dipyrrolic products in incubations of as citral (an a,p unsaturated terpenoid aldehyde),
purified CYP2BP^^. However, this fraction drops and other aromatic aldehydes (cinnamaldehyde,
to —2.5% (comprised largely of hematinic acid, benzaldehyde, and 3-phenylpropionaldehyde) is
with traces of methylvinylmaleimide and propent- accompanied by bleaching of the heme chro-
dyopents) in CYP3A-enriched rat liver microso- mophore that is not prevented by catalase, super-
mal incubations with DDEP or cumene oxide dismutase, epoxide hydrolase, GSH, or
hydroperoxide^^^. Accordingly, in liver microso- ascorbic acid"^^^' '^^^. The corresponding k^^^^^ val-
mal or purified P450 incubations, the bulk of the ues revealed that saturated aldehydes are generally
Inhibition of Cytochrome P450 Enzymes 283

more inhibitory than their afi unsaturated coun- heme plus a phenylethyl group (Figure 7.26). The
terparts, and primary aldehydes are more potent adduct was proposed to involve reaction of the
inactivators than the structurally related secondary hydroperoxy catalytic intermediate with the alde-
and tertiary aldehydes^^^. Studies with wild-type hyde to give a peroxyhemiacetal that fragmented
CYP2B4 and its T302A mutant, including meas- to yield an alkyl radical^^^. In contrast, with
urements of the deuterium isotope effects, rates of w-chloroperbenzoic acid, 3-phenylpropionalde-
inactivation, and rates of product formation sug- hyde yielded a phenylpropionyl-modified heme
gest that aldehyde-mediated CYP2B4 inactivation adduct purportedly generated from the reaction of
involves deformylation of the aldehyde. In the the heme with the corresponding carbonyl radical
inactivation of CYP2B4 by 3-phenylpropionalde- (Figure 7.27). In this reaction, homolytic oxygen-
hyde, a heme adduct is formed with a molecular oxygen bond cleavage of m-chloroperbenzoic acid
weight equal to that of native heme plus 104 mass itself also generated a chlorobenzoyloxy-heme
units, in agreement with loss of the carbonyl adduct^io.
group from the original aldehyde. P450 inactiva- Similar studies with fra«5'-4-hydroxy-2-none-
tion by aldehydes has been proposed to involve nal (HNE, a cytotoxic byproduct of biological
homolytic cleavage of a peroxyhemiacetal inter- membrane lipid peroxidation), indicate that it is
mediate to yield formic acid and an alkyl radical also metabolically activated by CYP2B1 and -2B4
that adds to the heme moiety'*^^. The heme adduct to a reactive species that binds irreversibly to their
obtained in similar reactions of the F87G mutant prosthetic heme'^^^ Unlike the mechanism-based
of CYP108 (P450BM3) has been fully character- inactivation by aromatic aldehydes, structural
ized by NMR and has been shown to involve analyses of the corresponding heme adduct (MW
addition of the decarbonylated substrate radi- 770) revealed that the reaction proceeds without
cal specifically to the y-meso position of the deformylation and involves an acyl carbon radical
heme group (Figure 7.26)"^^^. The resulting heme- that partitions between addition to the heme and
modified enzyme could be reduced in the pres- formation of the carboxylic acid'^^^ Together these
ence of NADPH and lauric acid but was not able findings suggest that the P450-mediated meta-
to actually oxidize the lauric acid. bolic activation of aldehydes is a versatile process
Interestingly, incubation of CYP2B4 with arti- wherein the enzyme may be inactivated via mech-
ficial oxidants and aldehydes yielded different anistically diverse heme modifications.
heme adducts. 3-Phenylpropionaldehyde yielded It is to be noted that P450 enzymes are
an adduct with a mass equal to that of native sometimes inactivated by mechanisms that
involve destruction of the prosthetic heme without
the detectable formation of heme adducts. In some
instances, these reactions result in binding of
heme fragments to the protein (Section 3.4),
but in most instances the incidence of heme-
protein cross-linking has not been investigated.
The destructive mechanisms of most perox-
ides386-388^ halocarbons (CCl4)380' 38i^ internal
acetylenes (3-hexyne)2^'^, allenes (1,1-dimethylal-
lene)"^^^, cyclopropylamines (7V-methyl-A^-benzyl-
cyclopropylamine)^^^' ^^^, benzothiadiazoles such
as 5,6-dichloro-l,2,3-benzothiadiazole^^^, and
methyl thieno[3.2-d][ 1,2,3]-thidiazole-6-carboxy-
late"^^^, phenolic compounds such as the anti-
inflammatory drug diclofenac"^^^' '^^^,
rhapontigenin"^^^, and resveratrol"^^^, the HIV-1
reverse transcriptase inhibitor delavirdine^^^' ^^^,
and the D4 dopamine receptor antagonist
Figure 7.26. Structure of the heme adduct isolated
from P450 inactivated by 3-phenylpropionaldehyde. SCH66712422 (Figure 7.28) remain poorly character-
ized. Hypothetical mechanisms can be formulated
284 M.A. Correia and P.R. Ortiz de Montellano

N- 7N ,N- 7N
Fe y
N ^ -HC02H ^^
OH

CH2.

Different
^- heme
adducts

Figure 7.27. The inactivation of P450 enzymes by aldehydes appears to involve free radical intermediates, one of
which retains the carbonyl group and one which does not. These radicals add to the heme group.

N
N OMe
-S' I II CO2H HO
OH
1,2,3-Benzothiadiazole Diclofenac Rhapontigenin

NHSO2CH3

Delavirdine SCH66712

Figure 7.28. Structures of other classes of P450 mechanism-based inactivating agents.


Inhibition of Cytochrome P450 Enzymes 285

for these reactions but experimental evidence to Although it is likely that other agents cause P450
support the mechanisms is not available. denaturation, it is unlikely that the process is phys-
The P450 destruction mediated by halocarbons iologically relevant because of the high drug
was once believed to stem from the secondary concentrations that are required.
action of the lipid peroxides that are concomi-
tantly formed, but it is now evident that sub-
stances like CCI4 can destroy the heme group 4. P450 Enzyme Specificity
directly^^^' ^^^' 423^26 J^IQ associated cross-linking
of heme fragments to the protein suggests that a The isoform-specific inhibition of P450
radical species (CCI3 or CCI3O2) may be respon- enzymes is a promising avenue for the develop-
sible for heme destruction^^ ^ On the other hand, ment of therapeutic, insecticidal, and herbicidal
the catalytic reduction of halocarbons, including agents, as well as for investigation of the struc-
CCI4, produces semistable complexes with tures, mechanisms, and biological roles of
Soret maxima in the 450-500 nm range'^^^"^^^ individual P450 enzymes. The biosynthetic P450
Studies with model iron porphyrins, including the enzymes have been the primary focus of efforts
detailed characterization of a dichlorocarbene- to develop isoform-specific P450 inhibitors
metalloporphyrin complex"^^^, suggest that the because (a) they are better targets for specific
long-wavelength Soret bands are due to ferrous inhibitors because of their high substrate speci-
halocarbene-heme complexes. This hypothesis is ficity and (b) there is high practical utility for such
supported by the finding that carbon monoxide is inhibitors. In contrast, the broad, overlapping,
formed in the reductive metabolism of CCI4 by specificities of xenobiotic metabolizing P450
P450. This reaction is likely to occur by a mecha- isoforms makes the design of isoform-specific
nism similar to that for the generation of carbon rather than -selective inhibitors more dififi-
monoxide from methylenedioxyphenyl complexes cult'^'^^' ^^. Selective inhibitors of P450 enzymes,
(Section 3.2.1)"^^^. In fact, porphyrin dichlorocar- as illustrated by the amphetamines^^^' ^^^,
bene-iron complexes do react with water to give TA0236,237,244-246^ Secobarbital^^' ^^\ gestodene^^^
carbon monoxide and with primary amines to give ftirafylline^^^' ^^2, 1-ethynylpyrene^^' ^^, and
isonitriles"^^^' ^^^. Studies with halothane suggest 2,3',4,5'-tetramethoxystilbene'*'^^ are fairly com-
that it is also possible to form complexes in which mon (see Appendix). Caution is required in
the halocarbon is a-bonded to ferric heme iron evaluating claims of inhibitor selectivity or speci-
atom435-^37 ficity, as they are limited by the range of P450
The links between formation of an iron-alkyl enzymes actually examined. Only in the case where
complex and irreversible destruction of the heme an inhibitor has been tested with all the known
moiety have not been forged, but model studies P450 isoforms in an organism can it be truly said to
with diaryl- and carbethoxy-substituted carbene be specific, at least in that organism. The claim for
complexes suggest that the halogenated carbenes specificity of inhibitors tested against only two or
may shift to form a bond with a nitrogen of the three isoforms is necessarily limited.
porphyrin'^^^^'^^ The resulting A/-haloalkyl adduct
are likely to undergo water-dependent hydrolysis
and might therefore not be detected by the methods 5. Inhibitors of Biosynthetic
used to isolate other iV-alkyl porphyrins. However, Enzymes
the formation of alternative reactive species that
attack the protein or the heme cannot be ruled out. Several comprehensive review articles have
High (1-5 mM) concentrations of indo- discussed the potential clinical relevance and
methacin and other nonsteroidal anti-inflamma- applications of inhibitors of biosynthetic P450
tory agents reportedly denature P450 enzymes enzymes^' 446-448 jj^^ following discussion will
because of their surfactant properties"^"^^. The therefore be limited to an illustration of the
loss of P450 content seen when indomethacin is strategies employed in the design and develop-
added to liver microsomes is paralleled by essen- ment of the currently available and/or prospective
tially stoichiometric appearance of a P420 peak. inhibitors and their mechanistic diversity.
286 M.A. Correia and P.R. Ortiz de Montellano

5.1. P450,,, is critical for inhibition since (225)-22-aminocho-


lesterol, binds to P450g^^ —1,000 times more
A single P450 enzyme (P450^^^, CYPl 1 A) cat- weakly (^i = 13 |LJLM) even though the amino
alyzes the three oxidative steps required to cleave function is located on the correct carbon.
the side chain of cholesterol. A rational approach Various mechanism-based, irreversible inhi-
to the development of P450g^^ inhibitors was bitors of P450g^^, such as analogs of pregnenediol
based on the incorporation of amino"^"^^^^^ and with an acetylenic group grafted into their side
thiol^^^ functions on the cholesterol side chain chain, have been developed (Figure 7.30)^^^^^^^.
at positions that favor their coordination to the Although this enzyme inactivation results in
prosthetic heme iron (Figure 7.29), yielding destruction of the heme chromophore, no alkylated
potent reversible inhibitors (K^ = 25-700 nM). heme was detected. Replacement of the side-chain
Thus, replacement of the first hydroxyl group carbons beyond C-23 in 20-hydroxycholesterol by
catalytically inserted into the cholesterol side a trimethylsilyl group also results in a P450g^^
chain with an amine function yields (22R)-22- mechanism-based inactivator (Figure 7.30)^^^.
aminocholesterol, one of the most potent P450g^^ Model studies have demonstrated that 1-substi-
inhibitors"^^^. The stereochemistry of this insertion tuted 3-trimethylsilyl-l-propanol is oxidized by
chemical reagents to ethylene, the trimethylsilyl
radical, and an aldehyde"^^^:

RCH(OH)CH2CH2Si(CH3)3 -> RCHO


+ CH2=CH2 + (CH3)3Si-
If the chemical model is relevant, P450g^^ may be
inactivated by reaction of the enzyme with the
trimethylsilyl radical or, less likely, from oxidation
of the ethylene produced in the initial catalytic
turnover. An interesting variant of a mechanism-
based inhibitor is provided by (20»S)-22-«or-22-
thiacholesterol, in which the sulfur that replaces
the carbon at position 22 is oxidized by P450g^^ to
a sulfoxide that is a potent but not an irreversible
inhibitor of the enzyme^^^' ^59,46o

Figure 7.29. Reversible inhibitors of biosynthetic


enzymes are usually constructed by incorporating a 5.2. Aromatase
nitrogen or other coordinating atom into a hpophiHc
structure with high affinity for the protein active site Aromatase, through a three-step catalytic trans-
(see Figure 7,1). An inhibitor of P450^^^ was thus formation, controls the conversion of androgens to
obtained by placing an amino group at the position estrogens. Competitive and mechanism-based
normally occupied by the first hydroxyl group added to inhibitors of aromatase have been clinically
the cholesterol side chain by P450 exploited in the treatment of estrogen-dependent

SiMe^

HO

Figure 7.30. Two mechanism-based inhibitors of cytochrome P450


Inhibition of Cytochrome P450 Enzymes 287

mammary tumors^' ^61^66 ^j^^ benign prostatic tempting to speculate that aminoglutethimide
hyperplasia"^^^' ^^^, and have some promise in the binds within the active sites of P450 and
control of coronary heart disease"^^^. Indeed, some sec

of the more promising newer agents are in cUnical P450^j.Q^ in approximately the same orientation as
^j^^jg463^66 Aminoglutethimide, an inhibitor of aro- the normal sterol substrate, and that the location
matase, has been used to treat hormone-dependent of the nitrogen in the two inhibitors dictates their
metastatic breast carcinoma, but its poor specificity differential enzyme selectivity.
and the incidence of side effects, primarily Potent inhibitors have also been developed that
use an imidazole or related functions to coordinate
from inhibition of P450g^^, has compromised its
to the aromatase iron atom. The most promising of
utility^^^^^^. Replacement of the aminophenyl
these for the treatment of breast cancer are fadro-
group in aminoglutethimide (Figure 7.31) by a
zole, {4-(5,6,7,8-tetrahydroimidazo[l ,5a]-pyridin-
p)nidine moiety affords [pyridoaminoglutethimide
5-yl)benzonitrile monochloride} (CGS16949;
(3-ethyl-3-(4-pyridyl) piperidine-2,6-dione)], an
Figure 7.31), and its congener [bis-(p-
agent that inhibits aromatase but not P450g^^'^^^'
cyanophenyl)-imidazo-1 -yl-methane hemisucci-
'*^^. The enhanced specificity for aromatase may
nate]^^^^^^. Both of these agents selectively inhibit
reflect a differential positioning of the pyridyl
aromatase rather than of P450g^,^, P45021, or
nitrogen within the two active sites that enables it
P45011B475,478 Fadrozole does inhibit aldosterone
to coordinate with the heme of aromatase but not
production (18-hydroxylase activity) in rats, but
of P450g^^. Interestingly, an aminoglutethimide
this is much less pronounced with its congener^^^.
analog with a nitrogen at the opposite end is a
Phase I clinical trials of fadrozole indicated that it
more potent P450g^^ inhibitor than glutethimide
is a potent inhibitor of estrogen biosynthesis in
but has little or no activity against aromatase^^^.
postmenopausal women with advanced breast can-
These findings suggest that the 19-methyl and
^gj474-478 jj^ addition to being well tolerated by
carbon 22 of the sterol side chain are separated by
patients, even at the maximally effective dose, it did
a distance roughly equal to the length of the not significantly alter Cortisol, androstenedione,
aminoglutethimide structure. It is therefore testosterone or aldosterone levels"^^"^^^.

CH3N, .,N
H2N N
S O
Aminoglutethimide R-76713
X = CH2SCH3,N3, / \, / \

N ^
N
: % : N^y

Me Me
CN
Fadrozole Anastrozole
Figure 7.31. Structures of some competitive inhibitors of aromatase. In each of these compounds, a heteroatom
is placed so that it can coordinate to the heme iron atom.
288 M.A. Correia and P.R. Ortiz de Montellano

However, fadrozole may now be surpassed by letro- the adjuvant treatment of postmenopausal women
zole (CGS20267 or Femara; Figure 7.31), an with hormone receptor-positive early breast
advanced nonsteroidal aromatase inhibitor, which cancer^^^' ^^^.
appears to be more potent and effective than fadro- Powerful competitive steroid inhibitors of
zole in the treatment of postmenopausal women aromatase have been synthesized by replacing the
with advanced breast cancer^^^' '*^^. C-19 methyl with sulfur- or nitrogen-containing
6 [(4-Chlorophenyl)( 1H-1,2,4-triazol-1 -yl) functions that can coordinate to the heme iron
methyl]-1 -methyl-1 H-benzotriazole, (R76713; (e.g.. Figure 7.31)^^^^^^ The 19-methyl substit-
Figure 7.31), is a relatively selective and very uents include the following: CH^SCR^'^^^^^,
potent inhibitor of human placental aro- CH3SCH2CH/93^ HSCH2^9i, 494^ RSSCH2 (Org-
matase"^^ ^~^^^. Its (+) enantiomer has a lower IC5Q 30958, R = ethyl is best)^^^, SH^^i, NH2, and
value than the ( - ) enantiomer when assayed NH2CH2^^^ Oxygen (oxiranyl), sulfur (thiiranyl),
against human placental aromatase and exhibits and nitrogen (aziridinyl) three-membered rings
no appreciable inhibition of other steroidogenic have also been used to replace the 19-methyl group
enzymes or liver microsomal P450 enzymes at (Figure 7.32)^^^-^^^ the best of the resulting
concentrations up to 1,000-times the aromatase steroids exhibiting K^ values in the 1 nM range. The
Tp 482,483 epoxide, episulfide, and aziridine functions inhibit
Several other nonsteroidal compounds have the enzyme by stereoselective coordination of the
been developed as novel and selective aromatase heteroatom to the iron atom but, despite their reactiv-
inhibitors, including 4-(4'-aminobenzyl)-2-oxazo- ity, apparently do not inactivate the enzyme.
lidinones"^^"^, 7-(alpha-azolylbenzyl)-1 H-indoles However, the steroids in wiiich the 19-methyl group
and indolines of which l-ethyl-7-[(imidazol-l-yl) is replaced by a sulfliydryl or HSCH2 group
(4-chlorophenyl)methyl]-lH-indole 12c exhibited (Figure 7.32) are irreversible mechanism-based
the most promising potency*^^, 4-imidazolylfla- inhibitors rather than simple competitive
vans"^^^, and anastrozole (Arimidex; Figure 7.31)"^^^. inhibitors'*^*.
Of these anastrozole has recently been approved The strategies developed for the inactivation of
in the United States and several other countries for hepatic P450 enzymes have also been exploited

CHF2
SH
CH2SH
CH2SSCH2CH3 (prodrug)
CH2C=CH
CH=C=CH2

OH
OCOCH2CH3
CH2SH

Exemestane

CH2

Figure 7.32. Structures of a selection of mechanism-based inactivators of aromatase.


Inhibition of Cytochrome P450 Enzymes 289

in the design and synthesis of mechanism-based human placental aromatase activity as potently as
aromatase inactivators. Substitution of the nor- 4-OHA and FCE-24304 (6-methylene-androstan-
mally hydroxylated methyl group (C-19) with a l,4,-diene-3,l7-dione) but, unlike both these
propargylic or allenic moiety (Figure 7.32) con- compounds, it has little intrinsic androgenic
verts the sterol into an irreversible aromatase inhi- activity and does not affect 5a-reductase or
bitor^^^"^^^. The details of aromatase inactivation P450 516-518^
sec
by these acetylenic and allenic agents remain
Conjugation of the 4-hydroxyandrostene
unclear, but it is likely that they are activated
nucleus as in 1,4,6 androstatriene-3,17-dione
to intermediates that react with either the heme
(ATD), conveys aromatase inhibitory and marked
or the protein (see Sections 3.1 and 3.3.2).
tumor regression activities (—80%)^^^' ^^^. On the
Replacement of the C-19 methyl with a difluo-
other hand, the introduction of a C^-methyl into
romethyl also yields a mechanism-based inactiva-
l,4-androstadiene-3,l7-dione as in Atamestane
tor that must be activated by C-19 hydroxylation
(1 -methylandrosta-1,4-diene-3,17-dione, SH-489),
(Figure 7.32)^^^^^^ as tritium release from the tri-
apparently enhances its affinity (K^ ~ 2 nM vs K^
tium-labeled difluoromethyl derivative is required
of 29 nM for 4-OHA) for the human placental aro-
for enzyme inactivation^^^. It is likely that the
matase while slowing its inactivation of the
difluoromethylalcohol thus produced decomposes
enzyme, thereby reducing the production of estro-
to the acyl fluoride that irreversibly binds to a pro-
genic products^ ^^' ^^^. The compound along with
tein nucleophile.
its 1,2 methylene-substituted congeners has been
The 19-substituted analog of androst-4-ene-3, evaluated in Phase I clinical trials for possible
17 dione steroid inhibitors, Org-30958 [19- therapy of estrogen-dependent conditions such
(ethyldithio)androst-4-ene-3,17-dione], has been as breast cancer and benign prostatic hypertrophy.
assessed in Phase I clinical trials for estrogen- Additional steroidal agents explored for their
dependent breast cancer chemotherapy^^'*. The aromatase suicide inactivation include androst-
ethyldithio substitution apparently renders the 5-ene-7,l 7-dione and its 19-hydroxy derivative^^^.
steroid more stable extracellularly than the free Turnover-dependent irreversible inactivation of
thiol Org-30365 (19-mercapto-androst-4-ene-3, the enzyme via protein modification is also
17-dione), resulting in vivo in animal models in an achieved by introducing a 6-keto group into the
8-fold greater aromatase inhibitory activity than steroid skeleton (Figure 7.33)^^^"^^^. Monitoring
either 4-OHA or SH-489. Its in vivo potency the ^H: ^^^C ratio in studies with the C-19 double-
requires intracellular reduction of the disulfide to labeled inhibitor indicates that the C-19 methyl,
release the 19-mercapto analog Org-30365, a one of the C-19 hydrogens and, from a separate
more potent mechanism-based human placental double label experiment, the Ip-hydrogen, are
aromatase inactivator"^^^ than 4-OHA or retained in the covalently bound species^^^. These
SH489494.
findings do not define the underlying inactivation
Clii^ically effective mechanism-based aro- mechanism but appear to exclude the involvement
matase inactivators can also be obtained by intro- of C-19 demethylation and aromatization,
ducing substituents at the 4- or 6-positions of the although normal aromatization is possible because
sterol skeleton. 4-Acetoxy- and 4-hydroxy- 6-oxoestrone and 6-oxoestradiol are concurrently
4-androstene-3,17-dione (4-OHA) (Figure 7.32) formed.
irreversibly inactivate placental aromatase by Exemestane, 6-methylene-androsta-1,4-diene-
catalysis-dependent mechanisms involving the 19- 3,17-dione (Figure 7.32), is an aromatase inhibitor
methyl^^^' ^^'*. A possible mechanism for inhibition with an IC^Q for inhibition of human placental
of aromatase by the 4-substituted analogs, as illus- aromatase comparable to that of 4-OHA^^^' ^^^.
trated by 4-OHA, is shown in Figure 7.33. 4-OHA The K^ (nM) and 1^2 (min) values for the inactiva-
is used for the treatment of estrogen-dependent tion processes were 26 ± 1.4 and 29.0 ± 7.5, and
breast cancer^^^' ^^^. Of a series of A^'^, A"^'^, 13.9 ± 0.7 and 2.1 ± 0.2, for Exemestane and
and A^'^ analogs evaluated as prospective aro- 4-OHA, respectively. In spite of its relatively
matase inhibitors in preclinical trials, FC 24928 slow inactivation of aromatase, Exemestane is a
(4-aminoandrostan-l,4,6-triene-3,l7-dione) is the more potent agent in experimental animals^ ^^, and
most promising candidate because it inactivates also much more effectively causes regression of
290 M.A. CorreJa and P.R. Ortiz de Montellano

[Fe]

HO-V

4-Hydroxyandrostenedione
(4-HOA)

Figure 7.33. A possible mechanism for the inactivation of aromatase by 4-OHA.

chemically induced mammary tumors in rat aromatization reaction, have been found to be
than MDL18962 or SH-4895i^ Because of its potent competitive inhibitors of the human placen-
oral effectiveness and potent irreversible aro- tal aromatase^^^. In contrast, related halohydrin
matase inhibition, Exemestane has been recently analogs are potent mechanism-based aromatase
approved and introduced into the global market inhibitors^"^^.
under the name Aromasin^^^^^ •. Finally, 4p,5p-epoxyandrostenedione as well
MDL 18,962 10-(2-propynyl)estr-4-ene-3,17- as its 19-hydroxy and 19-oxo derivatives have
dione is one of the most potent of the mechanism- been examined as aromatase inhibitors. The epox-
based inactivators of aromatase, with K^ values of ides were found to be weak competitive inhibitors,
the order of 3-4 nM, and ty2 of 9 and 6 min, whereas the 19-hydroxy and 19-oxo derivatives
respectively, for the human and baboon placental were largely ineffective^^^.
aromatases^^^' ^^"^^ ^^^^ ^^^' ^^^ pj^ase I clinical trials
and preclinical findings indicate that MDL 18,962
5.3. Lanosterol 14-Demethylation
is indeed effective in lowering estrogen levels^^^.
The time-dependent inactivation of aromatase The 14-demethylation of lanosterol is a
by 10-hydroperoxy-4-estrene-3,17-dione, reportedly key step in the biosynthesis of cholesterol. The
a mechanism-based inactivator of aromatase^^'*' ^^^, preferential inhibition of the P450 enzyme that
is inhibited by NADPH or an alternative substrate catalyzes this reaction by a number of substituted
and is partially reversed by dithiothreitol. Other imidazoles, pyridines, pyrimidines, and other
high affinity 10-substituted analogs, such as the lipophilic heterocycles has been exploited in the
mechanism-based inhibitor 10-P-mercaptoestr- construction of clinically important antifungal
4-ene-3,l7-dione, and the competitive inhibitors agents^' The antifungal action following the
(19i^)-10-oxiranyl and lO-thiiranyl-estr-4-ene- inhibition of 14-demethylase is thought to result
3,17-diones and 10-p,-MeSCH2-estr-4-ene-3,17- from the accumulation of 14-methyl sterols in the
dione, have been reported^^i, 492,494,536,537 Q_IQ^ membranes of susceptible ftmgi that cause deleteri-
C-2 hydroxyethyl bridged steroids synthesized as ous changes in their membrane permeability^"^^"^^^.
stable carbon analogs of the 2p-hydroxylated 19- Miconazole (Figure 7.34), fluconazole
oxoandrostenedione, a putative intermediate in the (Figure 7.34), ketoconazole (Figure 7.1), and
Inhibition of Cytochrome P450 Enzymes 291

CI
/ = / O-

COCH3 N ^ CI
CH(0H)CH3 Miconazole

N-N

HQ ^ Fluconazole

Figure 7.34. Competitive inhibitors of lanosterol 14-demethylase. The group R on the sterols is either
C(CH3)CH2CH2CH2C(CH3)2 or a close variant. Fluconazole and miconazole are clinically employed as antifungal
agents.

related azole structures inhibit the fungal 14- activities as well as respectable in vivo efficacies
demethylase activity at extremely low (nM) con- in a murine model of candidiasis^^'^. Introduction
centrations^"*^' ^^'^^ but only inhibit the of an oxime functionality (=NOH) at position 15
14-demethylase activity of the mammalian host at of the sterol group, also results in an inhibitor with
higher (up to 100 |JLM) concentrations^"^^' ^^^. Low a very similar geometry of the nitrogen as that in
doses of ketoconazole, however, appear to inhibit the 15-azahomosterols^^^. Similarly, 7-oxo-24,25-
not only the C^^ 20 ty^se, but also human hepatic dihydrolanosterols, which uncouple the catalytic
CYP3A4, and thus this agent is not very clinically turnover of CYP51 by competitively blocking
useful^^^' ^^^ As expected, the substituted imida- transfer of a second electron to the oxyfer-
zoles and other nitrogen-based heterocyclic rocomplex, have also been considered as prospec-
antifungal agents coordinate with the P450 heme tive inhibitors^^^. Furthermore, 15-, 32-, and
iron to yield typical type II binding spectra^^^. 15,21-oxylanosterol analogs have been examined
Potent isoprenoid-containing imidazole as mechanism-based inactivators of lanosterol
antifungal agents, such as AFK-108 (l-[2- demethylase. Of these (32*S)-vinyllanost-8-en-
(2,4-dichlorophenyl)-2-((2£)-3,7-dimethylocta- 3p,32-diol is a potent time-dependent inactivator
2,6-dienyloxy) ethyl]-IH-imidazole), have also (^inac/^i = 0.36 min-^ |JLM-^), while the (32i?)-
been developed whose geranyl moiety specifically vinyllanost-8-en-3p,32-diol functions solely as a
interacts with the sterol side-chain recognition competitive demethylase inhibitor^^^. Replace-
region within the 14a-demethylase active site. ment of the 15-hydrogen normally lost during
Unfortunately, this compound and its famesyl catalysis by a fluorine yields 15a-fluorolanost-
derivative inhibited not only the rat liver counter- 7-en-3p-ol which is oxidized by the enzyme to the
part but also other hepatic drug metabolizing 14-aldehyde but proceeds no further due to the
P450 enzymes, placing their practical utility in fluorine substitution^^^. The compound is there-
question^^^. fore a "prodrug" inhibitor that requires metabolic
Structural considerations have also prompted the activation of its latent form rather than a true
development of 14a-methyl-15-aza-D-homosterols mechanism-based inactivating agent.
(Figure 7.34), which exhibit relatively high in Steroid analogs with amino and imidazole
vitro fungistatic potencies and high fungicidal functions attached to the 14-methyl carbon have
292 M.A. Correia and P.R. Ortiz de Montellano

been developed as inhibitors of lanosterol 14a- androst-5-en-3p-ol^^^. This compound reportedly


demethylation (Figure 7.34)^^^. Steroids with a does not inhibit P450g^^ or the sterol 21-hydroxy-
14-(l-hydroxyethyl), 14-(l-oxoethyl), or 14-car- lase. In contrast, sterols with a 17-difluoromethyl
boxyl group are competitive inhibitors of the group selectively inactivate the C-21 hydroxylase,
enzyme (Figure 7.34)^^^"^^^. Lanosterol analogs whereas sterols with a 17-dichloromethyl, -vinyl,
in which the 14a-methyl group has been replaced or -ethynyl function inactivate both the CI7- and
by a vinyl, ethynyl, allyl, propargyl, 1-hydroxy- C21-hydroxylases^^. More recently, 20-fluoro-
propargyl, 1-ketopropargyl, difluoromethyl, 17(20)-pregnenolone derivatives were designed as
epoxide, or episulfide moiety have been synthe- enol mimics of pregnenolone. All of the targeted,
sized as potential mechanism-based inhibitors of novel fluoroolefins were found to be potent
lanosterol 14a-demethylase^^^~^^^. Although the inhibitors of C-17(20) lyase579.
ethynyl sterols are clearly mechanism-based inac- Similar strategies have been used to develop
tivators of the enzyme^^^, most of these com- inhibitors that target the C-18 hydroxylation
pounds act as simple competitive inhibitors (e.g., involved in the biosynthesis of aldosterone^^^"^^^.
the epoxide and episulfide analogs). Finally, Thus, aldosterone analogs with C-18 iodomethyl,
because of their cholesterol lowering potential, chloromethyl, allyl, propargyl, vinyl, and methyl-
14a-demethylase inhibitors have also been thiomethyl functionalities have been synthesized
considered as hypolipidemic agents^^^. and some have been found to irreversibly inactivate
the enzyme. An active site-directed 18-acetylenic
deoxycorticosterone [21 -hydroxy-13 (-2-)propy-
5.4. Other Biosynthetic nyl)-18-nor-preg-4-ene-3,20-dione, MDL19,347]
is a promising inactivator of the rat and rhesus
Sterol Hydroxylases
monkey adrenal corticosterone 18-hydroxylase
A current strategy for the development of new (K^ ^ 38 nM; f,/2, 4.6 min) that also effectively
drugs for the treatment of prostatic cancer is the reduces plasma aldosterone levels in vivo^^^. This
development of inhibitors of CYP17-dependent compound appears to be a relatively selective
androgen biosynthesis. Thus, as in the case of inhibitor of aldosterone biosynthesis, as it fails to
other steroidogenic enzymes, an intense search inhibit the 11 p-hydroxylation of corticosterone and
has been undertaken for reversible and irreversible j)Q^586 Rationally developed inhibitors such
CYP17 inhibitors^^^. The resulting agents include as these may be useful in the management of con-
substituted imidazoles, pyridines, pyrimidines, ditions such as hypertension, hypokalemia, and
and other lipophilic heterocycles as the enabling edema that are associated with hyperaldosteronism.
moiety^ Of these, abiraterone (17-(3-
pyridyl)androsta-5,16-dien-3p-ol), a potent inhi- 5.5. Fatty Acid and Leukotriene
bitor (IC5Q, 4 nM) of the hydroxylase activity of Monooxygenases
human cytochrome CYP17, has been employed
clinically^^^ Its potency is apparently due to acti- Microsomal P450 enzymes of the CYP2C and
vation of the 16,17-double bond that is required 4A subfamilies in tissues such as the liver, lung,
for irreversible binding of these pyridyl steroids to kidney, peripheral vasculature, and polymor-
CYPn^^^ Analogs such as [17-(5- phonuclear leukocyte oxidize a variety of fatty
pyrimidyl)androsta-5,16-diene-3p-ol] and its 3- acids, including arachidonic acid and its deriva-
acetyl derivative, which are even more potent tives, to physiologically active metabolites.
CYP17 inhibitors in rats than abiraterone, are par- Some of these metabolites are well recognized
ticularly promising^^^. With testicular microsomal as biologically important regulators of renal,
CYP17, these compounds apparently exhibit the pulmonary, and cardiac function and vascular
dual characteristics of a type II binding spectrum tone^^^"^^^. Two general classes of such vaso-
and noncompetitive inhibition^^^ active metabolites are known to be produced in
Another mechanism-based inactivator of vascular and extravascular tissues: EETs gener-
both the 17-hydroxylase and C-17/C-20 lyase ated by CYP2C epoxygenases, and HETEs
activities of CYP17 is 17p-(cyclopropylamino)- products hydroxylated at the o)- and w-l positions
Inhibition of Cytochrome P450 Enzymes 293

by CYP4A^^^-^^^. Furthermore, with the excep- more selective, A/-methylsulfonyl-12,12-dibro-


tion of 20-HETE, all the other arachidonic acid mododec-11-enamide (DDMS; Figure 7.35) and
products occur as stereo- and regioisomers that its acid analog, 12,12-dibromo-dodec-ll-enoic
vary considerably in their biological activities and (DBDD)602-^oi DDMS has been found to be an
potencies^92, 593 jj^^s^ 55.^ 8,9-, 11,12-, and effective acute and chronic blocker of 20-HETE
14,15-EETs are potent vasodilators, specially in formation in vivo in rats^^^"^^"^. More recently an
various capillary beds, that act through activation even more potent and selective inhibitor of 20-
of K+-channels in vascular smooth muscle cells. HETE formation, HET0016 (A^-hydroxy-A^'-(4-
In contrast, 12(i^)- and 20-HETEs are potent vaso- butyl-2-methylphenyl)formamidine) (Figure 7.35)
constrictors. 20-HETE is a particularly potent has been discovered through combinatorial chem-
cerebral and renal microvessel vasoconstrictor, as istry, with an IC3Q of 35 ± 4 nM^^^. Because of its
well as a mediator of other important physiologi- high selectivity, it holds considerable promise as
cal processes (reviewed in Chapter 11). The a probe of 20-HETE dependent function.
altered production of EETs and 20-HETE in vari- Several mechanism-based irreversible inhi-
ous genetic and experimental models of patholog- bitors of P450-dependent arachidonic acid metab-
ical diseases has led to their consideration as olism have also been developed. For instance, in
plausible causative factors. Thus, not surprisingly, 11-dodecynoic acid, the terminal acetylenic analog
the P450 enzymes responsible for their biosynthe- of lauric acid, and 10-undecynoic acid have been
sis have been singled out as key targets not only shown to inactivate hepatic CYP4A lauric acid
for defining the pathological roles of these (^-hydroxylases while minimally altering the
metabolites, but also as possible sites for pharma- spectrophotometrically detectable hepatic P450
cological intervention in the treatment of these content or function^^' ^°^' ^^^.
conditions^^^"^^^. In this context, both the previ- The high susceptibility of these acetylenic
ously existing and new agents have been exploited fatty acid inactivators to p-oxidation, however,
as reversible or irreversible inhibitors of these compromises their in vivo utility and prompted the
enzymes. development of 10-undec5niyl sulfate (10-SUYS),
Accordingly, two reversible inhibitors of the acetylenic analog of 10-undecynoic acid in
CYP4A enzymes have been assessed as probes of which the carboxyl group is replaced by a sulfate
20-HETE involvement in various physiological (Figure 7.35)^^^. The sulfate is not susceptible to
processes with varying degrees of success: the not P-oxidation and thus prolongs the biological
so selective antifungal agent miconazole (Section lifetime, reduces the toxicity, and enables the
2.3)^^^"^^^ and the rationally designed and thus use of sulfate agent as an in vivo mechanism-based

CONHSO2CH3 Me

x-^ .OH
N N
DDMS HET0016 H

.CO2H

17-ODYA

.OSOgH .CONHSO2CH3

10-SUYS MSPPOH

Figure 7.35. Reversible and mechanism-based inhibitors of enzymes involved in the metabolism of fatty acids and
related endogenous substrates.
294 M.A. Correia and P.R. Ortiz de Montellano

CYP4A inactivator^^^. Administration of a iV-methylsulfonyl-6-(2-propargyloxyphenyl)hexa-


single dose of 10-SUYS, a potent and selective namide (MSPPOH; Figure 7.35)^^^' ^25-627
mechanism-based CYP4A inactivator, acutely Finally, since fatty acid hydroxylations at the
reduced the mean arterial blood pressure as well 0), (0-1, (0-2, (0-3, and (o-4 positions also occur in
as the urinary 20-HETE excretion in sponta- plants and bacteria^^^' ^^^, acetylenic fatty acids
neously hypertensive rats, consistent with the have also been employed to examine whether
inactivation of renal 20-HETE formation^ ^^ these hydroxylases are mechanistically similar
These findings thus suggest that 20-HETE could to their mammalian counterparts and/or to iden-
play an important role in blood pressure regulation tity the role of specific plant and bacterial P450
in hypertensive states and that the inhibition of enzymes^^^' ^^^. Thus, midchain and terminal
its synthesis in these conditions may be of thera- acetylenes such as 10-dodecynoic acid have been
peutic benefit^ ^^ used as probes of plant lauric acid (o—hydroxy-
CYP4A and CYP2C isoforms are also lases^^'6^^. Similarly, 17-ODYA has been shown to
inactivated by the acetylenic fatty acid analog, inactivate P450BJ^.3 (CYP108), an enzyme that
17-octadecynoic acid (17-ODYA; Figure 7.35), hydroxylates fatty acids at positions other than the
in a mechanism-based process^ ^^~^^'*. 17-ODYA terminal ((o) carbon, through a heme alkylation
has been particularly useful in vivo to probe the mechanism^^o.
involvement of these metabolites in physiological
and/or pathological processes^^v, 598, 603, 605-609,
612-618 ji^g ygg jj^g helped elucidate the specific
role of 20-HETE in the myogenic activation and 6. Summary
h)^oxic dilation of skeletal muscle resistance
arteries as well as the vasodilatory effects of NO Our knowledge of the structure and function
in the renal microcirculation^^^' ^^^' ^^'^' ^^l of P450 enzymes has greatly expanded over the
The acetylenic fatty acids 15-hexadecynoic past three decades, as has our appreciation of
(15-HDYA) and 17-ODYA have also been the chemical functionalities that they accept as
explored as modulators of leukotriene B^ (LTB^), substrates and of the moieties that can inhibit or
an important and clinically relevant inflamma- terminate their catalytic action. As a result, multi-
tory mediator, and its physiologically active ple routes are now available for the inhibition or
(o-hydroxylated metabolite^i^'^22. Both 15-HDYA inactivation of P450 enzymes that target either the
and 17-ODYA inactivated the polymorphonuclear protein and/or the heme. This knowledge has been
leukocytic LTB^ w-hydroxylase in whole cells exploited in the design and construction of both
and cell lysates^^^. In contrast, the shorter-chain reversible and irreversible inhibitors of increasing
acid, 10-undecynoic acid was much less effective, potency, specificity, and potential practical utility.
while the saturated analogs of 15-HDYA and One area of continuing growth is that of reversible
17-ODYA were inactive. 15-HDYA and 17- inhibitors that, by simultaneously conforming to
ODYA also inactivate pulmonary prostaglandin the predominantly lipophilic contours of the P450
(o-hydroxylases^^^. active site and providing a nitrogen that coordi-
The mechanism-based inactivator 1-ABT nates to the heme iron atom, have achieved
(Section 3.3.5), which is not very selective and enhanced potency and specificity. These agents
inactivates multiple P450 enzymes, including include the clinically relevant azole antifungal and
those responsible for the synthesis of both EETs cancer chemotherapeutic drugs (Section 5).
and 20-HETE, has also been used, alone or in con- The high promise of irreversible P450
junction with other inhibitors, to assess the role of inactivating agents has not yet culminated in
these metabolites in skeletal muscle angiogenesis agents of large-scale practical utility. Progress
induced by electrical stimulation as well as in in the field has continued to provide instruction on
the renal and vasoconstrictor actions of angio- approaches for the design of mechanism-based
tensin IP'^^' 602, 608, 624^ inhibitors that inactivate a P450 enzyme with high
Mechanism-based inhibitors of EET formation specificity and negligible release of reactive
have also been used as probes of the physiological metabolites into the medium, whether the inacti-
roles of these metabolites. One such agent is vation involves modification of the protein or the
Inhibition of Cytoclirome P450 Enzymes 295

heme. High specificity is provided by the require- References


ment not only for binding within the P450
active site but also for the more rigorous test of 1. Rahimtula, A.D. and P.J. O'Brien (1977). The peroxi-
compatibility with catalytic turnover by the target dase nature of cytochrome P450. In V Ullrich,
enzyme. Potency results from the irreversible 1. Roots, A. Hildebrandt, R.W. Estabrook, and A.H.
nature of the inhibition, which means that rela- Conney (eds.), Microsomes and Drug Oxidations,
tively high concentrations of the agent need not be Pergamon Press, Elmsford, NY, pp. 210-217.
maintained throughout the period of inhibition. 2. Rodrigues, A.D., D. Fernandez, M.A. Nosarzewski,
However, the involvement of reactive intermedi- W.M. Pierce, and R.A. Prough (1991). Inhibition of
ates such as radicals and ketenes in the inactiva- hepatic microsomal cytochrome P-450 dependent
tion requires that the inactivation process be monooxygenation activity by the antioxidant 3-
?er^butyl-4-hydroxyanisole. Chem. Res. Toxicol. 4,
highly efficient and occur without leakage of reac-
281-289.
tive functions into the medium. As our increased
3. Kharasch, E.D., N.K. Wendel, and R.F. Novak
understanding of mechanism-based inhibition (1987). Anthracenedione antineoplastic agent
makes this feasible, this remains an attractive and effects on drug metabolism in vitro and in vivo:
highly promising approach to novel therapeutic Relationship between structure and mechanism of
agents and other commercial applications. inhibition. Fundam. Appl. Toxicol. 9, 18-25.
On the negative side, the inhibition of P450 4. Testa, B. and P Jenner (1981). Inhibitors of
enzymes has increasingly been recognized as cytochrome P-450s and their mechanism of action.
a key locus of clinically relevant drug-drug inter- Drug. Metab. Rev 12, 1-117.
5. Correia, M.A., and PR. Ortiz de Montellano (1993).
actions and other pharmacological effects. One
Inhibitors of cytochrome P450 and possibilities for
needs only to consider the consequences of the
their therapeutic application. In K. Ruckpaul (ed.),
lethal inhibition of CYP3A4 in patients treated Frontiers in Biotransformation. Akademie-Verlag,
with agents like terfenadine to appreciate the gen- Berlin, pp. 74-146.
eral concern in the pharmaceutical industry with 6. Murray, M. and G.F. Reidy (1990). Selectivity in the
the subject of P450 inhibition. Our growing inhibition of mammalian cytochromes P-450 by
understanding of the structural and chemical basis chemical agents. Pharmacol. Rev. 42, 85-101.
of P450 inhibition should greatly improve our 7. Ortiz de Montellano, PR. (1988). Suicide substrates
ability to rapidly elucidate their nature when and for drug metabolizing enzymes: Mechanism and
if they do occur and, of course, to avoid their biological consequences. In G.G. Gibson (ed.),
occurrence in the later stages of drug design. Progress in Drug Metabolism. Taylor and Francis,
NewYork, pp. 99-148.
Finally, on a more heuristic level, the past few 8. Vanden Bossche, H. (1992). Inhibitors of P450-
years have led to the development of an increasing dependent steroid biosynthesis: From research to
repertoire of isoform-selective inhibitors that can medical treatment. J. Steroid Biochem. Mol. Biol.
be deployed in basic studies of the physiological 43, 1003-1021.
function of individual P450 enzymes. One area 9. Rendic, S. and RJ. Di Carlo (1997). Human
to which such inhibitors have contributed is to cytochrome P450 enzymes: A status report summa-
elucidation of the specific roles of the products rizing their reactions, substrates, inducers, and
of P450-catalyzed arachidonic acid oxidation. inhibitors. Drug Metab. Rev. 29, 413-580.
A toolbox of highly isoform-specific inhibitors 10. Lewis, D.R (2003). Human cytochromes P450 asso-
covering the full range of human and rodent P450 ciated with the Phase 1 metabolism of drugs and
other xenobiotics: A compilation of substrates and
enzymes, each with pharmacokinetic properties
inhibitors of the CYPl, CYP2 and CYP3 famiUes.
and a low toxicological profile that makes it suit- Curr Med Chem. 10, 1955-1972.
able for in vivo use, would be a panacea but at this 11. Kent, U.M., M.I. Juschyshyn, and PR Hollenberg
time is nothing more than a possibility. (2001). Mechanism-based inactivators as probes of
cytochrome P450 structure and function. Curr
Drug Metab. 2, 215-243.
Acknowledgment 12. Brueggemeier, R.W. (2002). Aromatase inhibitors in
breast cancer therapy. Expert Rev. Anticancer Ther
The preparation of this review was assisted by 2, 181-191.
13. Sato, A. and T Nakajima (1979). Dose-dependent
grants from the National Institutes of Health.
metabolic interaction between benzene and toluene
296 M.A. Correia and P.R. Ortiz de Montellano

in vivo and in vitro. Toxicol. Appl. Pharmacol. 48, 26. Kim, Y.-M., H.A. Bergonia, C. Miiller, B.R. Pitt,
249-256. WD. Watkins, and J.R. Lancaster, Jr. (1995). Loss
14. Watkins, P.B. (1990). Role of cytochromes P450 in and degradation of enzyme-bound heme induced by
drug metabolism and hepatotoxicity. Semin. Liver cellular nitric oxide synthesis. J. Biol. Chem. 110,
Dis. 10, 235-250. 5710-5713.
15. Jefcoate, C.R. (1978). Measurement of substrate 27. Alonso-Galicia, M., H. Drummond, K. Reddy,
and inhibitor binding to microsomal cytochrome J. Falck, and R. Roman (1997). Inhibition of 20-
P-450 by optical-difference spectroscopy. Meth. HETE production contributes to the vascular
Enzymol. 52, 258-279. responses to nitric oxide. Hypertension 29, 320-325.
16. Kumaki, K., M. Sato, H. Kon, and D.W. Nebert 28. Drewett, J.G., R.L. Adams-Hays, B.Y. Ho, and D.J.
(1978). Correlation of type I, type II, and reverse Hegge (2002). Nitric oxide potently inhibits the
type I difference spectra with absolute changes in rate-limiting enzymatic step in steroidogenesis.
spin state of hepatic microsomal cytochrome P-450 Mol. Cell. Endocrinol. 194, 39-50.
iron from five mammalian species. J. Biol. Chem. 29. Hanson, L.K., W.A. Eaton, S.G. Sligar,
253, 1048-1058. I.e. Gunsalus, M. Gouterman, and C.R. Connell
17. Schenkman, J.B., S.G. Sligar, and D.L. Cinti (1981). (1976). Origin of the anomalous Soret spectra of
Substrate interactions with cytochrome P-450. carboxycytochrome P450. J. Am. Chem. Soc. 98,
Pharmacol. Then 12, 43-71. 2672-2674.
18. Sligar, S.G., D.L. Cinti, G.G. Gibson, and J.B. 30. Omura, T. and R. Sato (1964). The carbon
Schenkman (1979). Spin state control of the hepatic monoxide-binding pigment of liver microsomes. 1.
cytochrome P-450 redox potential. Biochem. Biophys. Evidence for its hemoprotein nature. J. Biol. Chem.
Res. Commun. 90, 925-932. 239, 2370-2378.
19. Guengerich, F. P. (1983). Oxidation-reduction prop- 31. Collman, J.P and T.N. Sorrell (1975). A model for
erties of rat liver cytochromes P450 and NADPH- the carbonyl adduct of ferrous cytochrome P-450.
cytochrome P-450 reductase related to catalysis in J. Am. Chem. Soc. 97, 4133-4134.
reconstituted systems. Biochemistry 11, 2811-2820. 32. Leeman, T., P Bonnabry, and P Dayer (1994).
20. Kitada, M., K. Chiba, T. Kamataki. and H. Kitagawa Selective inhibition of major drug metabolizing
(1977). Inhibition by cyanide of drug oxidations in cytochrome P450 isozymes in human liver
rat liver microsomes. Jpn. J. Pharmacol. 11, microsomes by carbon monoxide. Life Sci. 54,
601-608. 951-956.
21. Ho, B. and N. Castagnoli (1980). Trapping of 33. Canick, J.A. and K.J. Ryan (1976). Cytochrome
metabolically generated electrophilic species with P-450 and the aromatization of 16-alpha-hydroxy-
cyanide ion: Metabolism of 1-benzylpyrrolidine. testosterone and androstenedione by human placen-
J. Med. Chem. 23, 133-139. tal microsomes. Mol. Cell. Endocrinol. 6, 105-115.
22. Sono, M. and J.H. Dawson (1982). Formation of low 34. Gibbons, G.F, C.R. Pullinger, and K.A.
spin complexes of ferric cytochrome P-450-CAM Mitropoulos (1979). Studies on the mechanism of
with anionic ligands: Spin state and ligand affinity lanosterol 14-alpha-demethylation: A requirement
comparison to myoglobin. J. Biol. Chem. 151, for two distinct types of mixed-function-oxidase
5496-5502. systems. Biochem. J. 183, 309-315.
23. Backes, W.L., M. Hogaboom, and W.J. Canady 35. Hansson, R. and K. Wikvall (1982). Hydroxylations
(1982). The true hydrophobicity of microsomal in biosynthesis of bile acids: Cytochrome P-450
cytochrome P-450 in the rat: Size dependence of the LM4 and 12a-hydroxylation of 5p-cholestane-
free energy of binding of a series of hydrocarbon 3a,7a-diol. Eur J. Biochem. 125, 423-429.
substrates from the aqueous phase to the enzyme 36. Meigs, R.A. and K.J. Ryan (1971). Enzymatic
and to the membrane as derived from spectral bind- aromatization of steroids. I. Effects of oxygen and
ing data. J. Biol. Chem. 257, 4063^070. carbon monoxide on the intermediate steps of estro-
24. Wink, D.A., Y. Osawa, J.E Darbyshe, C.R. Jones, gen biosynthesis. J. Biol. Chem. 246, 83-87.
S.C. Eshenaur, and R.W. Nims (1993). Inhibition of 37. Zachariah, PK. and M.R. Juchau (1975).
cytochromes P450 by nitric oxide and a nitric oxide- Interactions of steroids with human placental
releasing agent. Arch. Biochem. Biophys. 300, cytochrome P-450 in the presence of carbon mon-
115-123. oxide. Life Sci. 16, 1689-1692.
25. Khatsenko, O.G., S.S. Gross, A.B. Rifkind, and 38. Tuckey, R.C. and H. Kamin (1983). Kinetics of
J.R. Vane (1993). Nitric oxide is a mediator of the O2 and CO binding to adrenal cytochrome P-
decrease in cytochrome P450-dependent metabo- 450scc: Effect of cholesterol, intermediates, and
lism caused by immunostimulants. Proc. Natl. Acad. phosphatidylcholine vesicles. J. Biol. Chem. 258,
Sci. t/5'^90, 11147-11151. 4232-4237.
Inhibition of Cytochrome P450 Enzymes 297

39. Cohen, G.M. and G.J. Mannering (1972). Involve- 53. Silverman, R.B. (1988). Mechanism-Based Enzyme
ment of a hydrophobic site in the inhibition of the Inactivation: Chemistry and Enzymology. CRC Press,
microsomal para-hydroxylation of aniline by alco- Boca Raton, FL.
hols. Mol Pharmacol 8, 383-397. 54. Dalvi, R.R. (1987). Cytochrome P-450-dependent
40. Gerber, M.C., G.A Tejwani, N. Gerber, and J.R. covalent binding of carbon disulfide to rat liver
Bianchine (1985). Drug interactions with cimeti- microsomal protein in vitro and its prevention by
dine: An update. Pharmacol. Then 11, 353-370. reduced glutathione, ^rc/i. Toxicol. 61, 155-157.
41. Testa, B. (1981). Structural and electronic factors 55. De Matteis, RA. and A.A. Seawright (1973).
influencing the inhibition of aniline hydroxylation Oxidative metabolism of carbon disulphide by the
by alcohols and their binding to cytochrome P-450. rat: Effect of treatments which modify the liver tox-
Chem. Biol. Interact. 34, 287-300. icity of carbon disulphide. Chem. Biol. Interact. 7,
42. Wattenberg, L.W., L.K.T. Lam, and A.V Fladmoe, 375-388.
(1979). Inhibition of chemical carcinogen-induced 56. Bond, E.J. and FA. De Matteis (1969).
neoplasia by coumarins and alpha-angelicalactone. Biochemical changes in rat liver after administra-
Cancer Res. 39, 1651-1654. tion of carbon disulphide, with particular reference
43. Remmer, H., J. Schenkman, R.W. Estabrook, to microsomal changes. Biochem. Pharmacol. 18,
H. Sasame, J. Gillette, S. Narasimhulu et al. (1966). 2531-2549.
Drug interaction with hepatic microsomal cyto- 57. Halpert, J., D. Hammond, and R.A. Neal (1980).
chrome. Mol. Pharmacol. 2, 187-190. Inactivation of purified rat liver cytochrome P-450
44. Jefcoate, C.R., XL. Gaylor, and R.L, Callabrese during the metabolism of parathion (diethyl/7-nitro-
(1969). Ligand interactions with cytochrome phenyl phosphorothionate). J. Biol. Chem. 255,
P-450. 1. Binding of primary amines. Biochemistry 8, 1080-1089.
3455-3463. 58. Neal, R.A., T Kamataki, M. Lin, K.A. Ptashne,
45. Schenkman, J.B., H Remmer, and R.W. Estabrook R. Dalvi, and R.Y. Poore (1977). Studies of the
(1967). Spectral studies of drug interaction with formation of reactive intermediates of parathion. In
hepatic microsomal cytochrome P-450. Mol. D.J. Jollow, J.J. Koesis, R. Snyder, and H. Vaino
Pharmacol. 3, 113-123. (eds.), Biological Reactive Intermediates. Plenum
46. Dominguez, O.V and L.T. Samuels (1963). Press, New York, pp. 320-332.
Mechanism of inhibition of adrenal steroid 11-beta- 59. Miller, G.E., M.A. Zemaitis, and FE. Greene
hydroxylase by methopyrapone (metopirone). (1983). Mechanisms of diethyldithiocarbamate-
Endocrinology 73, 304-309. induced loss of cytochrome P-450 from rat liver.
47. Temple, T.E. and G.W. Liddle (1970). Inhibitors of Biochem. Pharmacol. 32, 2433-2442.
adrenal steroid biosynthesis. Ann. Rev. Pharmacol. 10, 60. El-hawari, A.M. and G.L. Plaa (1979). Impairment of
199-218. hepatic mixed-function oxidase activity by alpha- and
48. Rogerson, T.D., C.F. Wilkinson, and K. Hetarski beta-naphthylisothiocyanate: Relationship to hepato-
(1977). Steric factors in the inhibitory interaction of toxicity. Toxicol. Appl. Pharmacol. 48, 445^58.
imidazoles with microsomal enzymes. Biochem. 61. Lee, PW, T Arnau, and R.A. Neal (1980).
Pharmacol. 26, 1039-1042. Metabolism of alpha-naphthylthiourea by rat liver
49. Wilkinson, C.R, K Hetarski, G.R Cantwell, and F.J. and rat lung microsomes. Toxicol. Appl. Pharmacol.
DiCarlo (1974). Structure-activity relationships in 53, 164-173.
the effects of 1-alkylimidazoles on microsomal oxi- 62. Lopez-Garcia, M.P, P.M. Dansette, and D. Mansuy
dation in vitro and in vivo. Biochem. Pharmacol. 23, (1993). Thiophene derivatives as new mechanism-
2377-2386. based inhibitors of cytochromes P450: Inactivation
50. Duquette, P.H., R.R. Erickson, and J.L. Holtzman of yeast-expressed human liver P450 2C9 by tienilic
(1983). Role of substrate lipophilicity on the N- acid. Biochemistry 33, 166-175.
demethylation and type I binding of 3-O-alkylmor- 63. Lopez-Garcia, M.P, P.M. Dansette, P. Valadon, C.
phine analogues. J. Med. Chem. 26, 1343-1348. Amar, PH. Beaune, F.P Guengerich et al. (1993).
51. Smith, S.R. and M.J. Kendall (1988). Ranitidine Human liver P450s expressed in yeast as tools for
versus cimetidine. A comparison of their potential reactive metabolite formation studies: Oxidative
to cause clinically important drug interactions. Clin. activation of tienilic acid by P450 2C9 and P450
Pharmacokinet. 15, 44-56. 2C10. Eur J. Biochem. 213, 223-232.
52. Ator, M.A. and Ortiz PR. de Montellano (1990). 64. Menard, R.H., T.M Guenthner, A.M. Taburet,
Mechanism-based (suicide) enzyme inactivation. In H. Kon, L.R. Pohl, J.R. Gillette et al. (1979).
D.S. Sigman and P.D. Boyer (eds.). The Enzymes: Specificity of the in vitro destruction of adrenal and
Mechanisms of Catalysis, Vol. 19, 3rd edn.. hepatic microsomal steroid hydroxylases by thio-
Academic Press, New York, pp. IXA-l'il. sterols. Mol. Pharmacol. 16, 997-1010.
298 M.A. Correia and P.R. Ortiz de Montellano

65. Kossor, D.C., S Kominami, S. Takemori, and H.D. 77. Halpert, J., J.-Y. Jaw, C. Balfour, and L.S. Kaminsky
Colby (1991). Role of the steroid 17a-hydroxylase (1990). Selective inactivation by chlorofluoroac-
in spironolactone-mediated destruction of adrenal etamides of the major phenobarbital-inducible
cytochrome P-450. Mol. Pharmacol. 40, 321-325. form(s) of rat liver cytochrome P-450. Drug Metab.
66. Decker, C , K. Sugiyama, M. Underwood, and M.A. Dispos. 18, 168-174.
Correia (1986). Inactivation of rat hepatic 78. CaJacob, C.A., W. Chan, E. Shephard, and PR.
cytochrome P-450 by spironolactone. Biochem. Ortiz de Montellano (1988). The catalytic site of rat
Biophys. Res. Commun. 136, 1162-1169. hepatic lauric acid ca-hydroxylase. Protein vs pros-
67. Decker, C.J., M.S. Rashed, T.A. Baillie, D. Maltby, thetic heme alkylation in the w-hydroxylation of
and M.A. Correia (1989). Oxidative metabolism of acetylenic fatty acids. J. Biol. Chem. 263,
spironolactone: Evidence for the involvement of 18640-18649.
electrophilic thiosteroid species in drug-mediated 79. Hammons, G.J., W.L. Alworth, N.E. Hopkins,
destruction of rat hepatic cytochrome P450. FP Guengerich, and FF Kadlubar (1989). 2-Ethyny-
Biochemistry 2H, 5128-5136. Inaphthalene as a mechanism-based inactivator
68. Menard, R.H., T.M. Guenthner, H. Kon, and J.R. of the cytochrome P-450-catalyzed N-oxidation of
Gillette (1979). Studies on the destruction of adre- 2-naphthylamine. Chem. Res. Toxicol. 2, 367-374.
nal and testicular cytochrome P-450 by spironolac- 80. Yun, C.-H., M.V Martin, N.E. Hopkins, W.L. Alworth,
tone: Requirement for the 7-alpha-thio group and G.J. Hammons, and FP Guengerich (1992). Modi-
evidence for the loss of the heme and apoproteins of fication of cytochrome P4501A2 enzymes by the
cytochrome P-450. J. Biol. Chem. 254, 1726-1733. mechanism-based inactivator 2-ethynylnaphthalene.
69. Sherry, J.H., J.P O'Donnell, L. Flowers, L.B. Biochemistry 31, 10556-10563.
Lacagnin, and H.D. Colby (1986). Metabolism of 81. Gan, L.-S.L., A.L. Acebo, and W.L. Alworth (1984).
spironolactone by adrenocortical and hepatic micro- 1-Ethynylpyrene, a suicide inhibitor of cytochrome
somes: Relationship to cytochrome P-450 destruc- P-450 dependent benzo(a)pyrene hydroxylase
tion. J. Pharmacol. Exp. Ther 236, 675-680. activity in liver microsomes. Biochemistry 23,
70. Colby, H.D., J.R O'Donnell, N. Lynn, D.C. Kossor, 3827-3836.
PB. Johnson, and M. Levitt (1991). Relationship 82. Roberts, E.S., N.E. Hopkins, W.L. Alworth, and PF
between covalent binding to microsomal protein and Hollenberg (1993). Mechanism-based inactivation
the destruction of adrenal cytochrome P-450 by of cytochrome P450 2B1 by 2-ethynylnaphthalene:
spironolactone. Toxicology 61, 143-154. Identification of an active-site peptide. Chem. Res.
71. Decker, C.J., J.R. Cashman, FC. Sugiyama, D. Toxicol. 6, 470-479.
Maltby, and M.A. Correia (1991). Formation of glu- 83. Chan, W.K., Z Sui, and PR. Ortiz de Montellano
tathionyl-spironolactone disulfide by rat liver (1993). Determinants of protein modification ver-
cytochromes P450 or hog liver flavin-containing sus heme alkylation: Inactivation of cytochrome
monooxygenases: A functional probe of two- P450 lAl by 1-ethynylpyrene and phenylacetylene.
electron oxidations of the thiosteroid? Chem. Res. Chem. Res. Toxicol. 6, 38-^5.
Toxicol. 4, 669-677. 84. Helvig, C , C. Alayrac, C. Mioskowski, D. Koop,
72. Halpert, J. and R.A. Neal (1980). Inactivation of D. Poullain, F Durst et al. (1997). Suicide inactiva-
purified rat liver cytochrome P-450 by chloram- tion of cytochrome P450 by midchain and terminal
phenicol. Mol. Pharmacol. 17, 427^34. acetylenes. A mechanistic study of inactivation of a
73. Halpert, J. (1982). Further studies of the suicide plant lauric acid omega-hydroxylase. J. Biol. Chem.
inactivation of purified rat liver cytochrome P-450 272,414-^21.
by chloramphenicol. Mol. Pharmacol. 21, 166-172. 85. Halpert, J., J.-Y. Jaw, and C. Balfour (1989). Specific
74. Halpert, J. (1981). Covalent modification of lysine inactivation by 17p-substituted steroids of rabbit
during the suicide inactivation of rat liver and rat liver cytochromes P-450 responsible for
cytochrome P-450 by chloramphenicol. Biochem. progesterone 21 -hydroxylation. Mol. Pharmacol. 34,
Pharmacol. 30, 875-881. 148-156.
75. Halpert, J., B. Naslund, and I. Betner (1983). 86. Stevens, J.C, J-Y. Jaw, C.-T. Peng, and J. Halpert
Suicide inactivation of rat liver cytochrome P-450 (1991). Mechanism-based inactivation of bovine adre-
by chloramphenicol in vivo and in vitro. Mol. nal cytochromes P450 C-21 and P450 17a by 17^-
Pharmacol. 23, 445^52. substituted steroids. Biochemistry 30, 3649-3658.
76. Halpert, J., C. Balfour, N.E. Miller, and 87. Guengerich, FP (1988). Oxidation of 17 alpha-
L.S. Kaminsky (1986). Dichloromethyl compounds ethynylestradiol by human liver cytochrome P-450.
as mechanism-based inactivators of rat liver cyto- Mol. Pharmacol. 33, 500-508.
chromes P450 in vitro. Mol. Pharmacol. 30, 88. Guengerich, FP (1990). Metabolism of 17 alpha-
19-24. ethynylestradiol in humans. Life Sc. 47, 1981-1988.
Inhibition of Cytochrome P450 Enzymes 299

89. Guengerich, F.P. (1990). Inhibition of oral contra- 100. Tinel, M., J. Belghiti, V Descatoire, G. Amouyal,
ceptive steroid-metabolizing enzymes by steroids P. Letteron, J. Geneve (1987). Inactivation of
and drugs. Am. J. Obstet. Gynecol. 163 (Pt 2), human liver cytochrome P-450 by the drug
2159-2163. methoxsalen and other psoralen derivatives.
90. Lin, H.L., U.M. Kent, and P.F. HoUenberg, (2002). Biochem. Pharmacol. 36, 951-955.
Mechanism-based inactivation of cytochrome P450 101. Labbe, G., V Descatoire, P. Beaune, P. Letteron,
3A4 by 17 alpha-ethynylestradiol: Evidence for D. Larrey, and D. Pessayre (1989). Suicide inacti-
heme destruction and covalent binding to protein. vation of cytochrome P450 by methoxsalen.
J. Pharmacol. Exp. Then 301, 160-167. Evidence for the covalent binding of a reactive
91. Kent, U.M., D.E. Mills, R.V Rajnarayanan, intermediate to the protein moiety. J. Pharmacol.
W.L. Alworth, and PR HoUenberg, (2002). Effect of Exp. Ther 250, 1034-1042.
17-a-ethynylestradiol on activities of cytochrome 102. Mays, D.C., J.B. Hilliard, D.D. Wong, M.A.
P450 2B (P450 2B) enzymes: Characterization of Chambers, S.S. Park, H.V Gelboin et al. (1990).
inactivation of P450s 2B1 and 2B6 and identifica- Bioactivation of 8-methoxypsoralen and irre-
tion of metabolites. J. Pharmacol. Exp. Ther 300, versible inactivation of cytochrome P450 in mouse
549-558. liver microsomes: Modification by monoclonal
92. Guengerich, F.P. (1990). Mechanism-based inacti- antibodies, inhibition of drug metabolism and dis-
vation of human liver microsomal cytochrome tribution of covalent adducts. J. Pharmacol. Exp.
P-450 IIIA4 by gestodene. Chem. Res. Toxicol. 3, Ther 254,720-731.
363-371. 103. Khojasteh-Bakht, S.C, L.L Koenigs, R.M. Peter,
93. Roberts, E.S., N.E. Hopkins, E.J. Zalulec, D.A. WE Trager, and S.D. Nelson (1998). (R)-(+)-
Gage, W.L. Alworth, and PF. HoUenberg (1994). Menthofuran is a potent, mechanism-based
Identification of active-site peptides from Un- inactivator of human liver cytochrome P450 2A6.
labeled 2-ethynylnaphthalene-inactivated P450 2B1 Drug Metab. Dispos. 26, 701-704.
and 2B4 using amino acid sequencing 104. Cai, Y., D. Bennett, R.V Nair, O. Ceska,
and mass spectrometry. Biochemistry 33, M.J. Ashwood-Smith, and J. DiGiovanni (1993).
3766-3771. Inhibition and inactivation of murine hepatic
94. Regal, K.A., M.L. Schrag, L.C. Wienkers, U.M. ethoxy- and pentoxyresorufin 0-dealkylase by nat-
Kent, and P.F. HoUenberg (2000). Mechanism-based urally occurring coumarins. Chem. Res. Toxicol. 6,
inactivation of cytochrome P450 2B1 by 872-879.
7-ethynylcoumarin: Verification of apo-P450 add- 105. Cai, Y., W Baer-Dubowska, M.J. Ashwood-Smith,
uction by electrospray ion trap mass spectrometry. O. Ceska, S. Tachibana, and J. DiGiovanni (1996).
Chem. Res. Toxicol. 13, 262-270. Mechanism-based inactivation of hepatic ethoxy-
95. He, K., T.E Woolf, and PR HoUenberg (1999). resorufin O-dealkylation activity by naturally
Mechanism-based inactivation of cytochrome occurring coumarins. Chem. Res. Toxicol. 9,
P-450-3A4 by mifepristone (RU486). J. Pharmacol. 729-736.
Exp. Ther 288, 791-797. 106. Koenigs, L.L., and WE Trager (1998).
96. Khan, K.K., Y.Q. He, M.A. Correia, and J.R. Mechanism-based inactivation of P450 2A6 by
Halpert (2002). Differential oxidation of mifepris- fiiranocoumarins. Biochemistry 37, 10047-10061.
tone by cytochromes P450 3A4 and 3A5: Selective 107. Schmiedlin-Ren, P, D.J. Edwards, M.E.
inactivation of P450 3A4. Drug Metab. Dispos. 30, Fitzsimmons, K. He, K.S. Lown, P.M. Woster
985-990. et al. (1997). Mechanisms of enhanced oral
97. Lunetta, J.M., K. Sugiyama, and M.A. Correia availability of CYP3A4 substrates by grapefruit
(1989). Secobarbital-mediated inactivation of rat constituents. Decreased enterocyte CYP3A4
liver cytochrome P-450b: A mechanistic reap- concentration and mechanism-based inactivation
praisal. Mol. Pharmacol. 35, 10-17. by fiiranocoumarins. Drug Metab. Dispos. 25,
98. Letteron, P., V Descatoire, D. Larrey, M. Tinel, 1228-1233.
J. Geneve, and D. Pessayre (1986). Inactivation and 108. Lown, K.S., D.G. Bailey, R.J. Fontana,
induction of cytochrome P-450 by various psoralen S.K. Janardan, C.H. Adair, L.A. Fortlage et al.
derivatives in rats. J. Pharmacol. Exp. Ther 238, (1997). Grapefruit juice increases felodipine oral
685-692. availability in humans by decreasing intestinal
99. Fouin-Fortunet, H., M. Tinel, V Descatoire, CYP3A protein expression. J. Clin. Invest. 99,
P Letteron, D. Larrey, J. Geneve et al. (1986). 2545-2553.
Inactivation of cytochrome P450 by the drug 109. He, K., K.R. Iyer, R.N. Hayes, M.W Sinz,
methoxsalen. J. Pharmacol. Exp. Ther 236, T.E Woolf, and PE HoUenberg (1998). Inactivation
237-247. of cytochrome P450 3A4 by bergamottin, a
300 M.A. Correia and P.R. Ortiz de Montellano

component of grapefruit juice, Chem. Res. Toxicol. microsomal oxidation in vitro. Chem. Res. Toxicol.
11, 252-259. 7, 792-799.
110. Chiba, M., J.A. Nishime, and J.H. Lin (1995). 122. Chambers, J.E. and H.W. Chambers (1990). Time
Potent and selective inactivation of human liver course of inhibition of acetylcholinesterase and
microsomal cytochrome P-450 isoforms by L-754, aliesterases following parathion and paraoxon
394, an investigational human immune deficiency exposures in rats. Toxicol. Appl. Pharmacol. 103,
virus protease inhibitor. J. Pharmacol. Exp. Then 420^29.
215, 1527-1534. 123. Koenigs, L.L., R.M. Peter, A.P Hunter, R.L.
111. Sahali-Sahly, Y., S.K. Balani, J.H. Lin, and T.A. Haining, A.E. Rettie, T. Friedberg et al. (1999).
Baillie (1996). In vitro studies on the metabolic Electrospray ionization mass spectrometric analy-
activation of the furanopyridine L-754,394, a sis of intact cytochrome P450: Identification of tie-
highly potent and selective mechanism-based nilic acid adducts to P450 2C9. Biochemistry 38,
inhibitor of cytochrome P450 3A4. Chem. Res. 2312-2319.
Toxicol. 9, 1007-1012. 124. Ha-Duong, N.T., S. Dijols, A.C. Macherey J.A.
112. Lightning, L.K., J.P Jones, T. Friedberg, M.P Goldstein, PM. Dansette, and D. Mansuy (2001).
Pritchard, M. Shou, T.H. Rushmore et al. Ticlopidine as a selective mechanism-based
(2000). Mechanism-based inactivation of inhibitor of human cytochrome P450 2C19.
cytochrome P450 3A4 by L-754,394. Biochemistry Biochemistry 4iS, 12112-12122.
39, 4276-4287. 125. Donahue, S.R., D.A. Flockhart, D.R. Abernethy,
113. Masubuchi, Y., T. Nakano, A. Ose, and T. Horie and J.W. Ko (1997). Ticlopidine inhibition of
(2001). Differential selectivity in carbamazepine- phenytoin metabolism mediated by potent inhibi-
induced inactivation of cytochrome P450 enzymes tion of CYP2C19. Clin. Pharmacol. Then 62,
in rat and human liver. Arch. Toxicol. 75, 538-543. 572-577.
114. Mani, C , R. Pearce, A. Parkinson, and D. Kupfer 126. Klaassen, S.L. (1998). Ticlopidine-induced pheny-
(1994). Involvement of cytochrome P4503A in toin toxicity ^««. Pharmacother 32, 1295-1298.
catalysis of tamoxifen activation and covalent 127. Lopez-Ariztegui, N., M. Ochoa, M.J. Sanchez-
binding to rat and human liver microsomes. Migallon, C. Nevado, and M. Martin (1998). Acute
Carcinogenesis 15, 2715-2720. phenytoin poisoning secondary to an interaction
115. Sridar, C , U.M. Kent, L.M. Notley E.M. Gillam, with ticlopidine. Rev. Neurol. 26, 1017-1018.
and PF. Hollenberg (2002). Effect of tamoxifen on 128. Donahue, S., D.A. Flockhart, and D.R. Abernethy
the enzymatic activity of human cytochrome (1999). Ticlopidine inhibits phenytoin clearance.
CYP2B6, J. Pharmacol. Exp. Then 301, 945-952. Clin. Pharmacol. Then 66, 563-568.
116. Zhao, X.J., D.R. Jones, YH. Wang, S.W. Grimm, 129. Richter, T, K. Klein, TE. Murdter, M. Eichelbaum,
and S.D. Hall (2002). Reversible and irreversible M. Schwab, and U.M. Zanger (2003). Clopidogrel
inhibition of CYP3A enzymes by tamoxifen and and ticlopidine are specific mechanism-based
metabolites. Xenobiotica 32, 863-878. inhibitors of human cytochrome P450 2B6.
117. Liu, H., M. Santostefano, and S. Safe (1994). 2- Proceedings of the 13th International Conference
Phenylphenanthridinone and related compounds: on Cytochromes P450, Prague, Czech Republic,
Aryl hydrocarbon receptor agonists and suicide p. S119.
inactivators of P4501A1. Arch. Biochem. Biophys. 130. Saunders, F J. and R.L. Alberti (1978). Aldactone:
313,206-214. Spironolactone: A Comprehensive Review. Searle,
118. Butler, A.M. and M. Murray (1993). Inhibition and New York.
inactivation of constitutive cytochromes P450 in rat 131. Kassahun, K., P.G. Pearson, W. Tang, 1. Mcintosh,
liver by parathion, Mol. Pharmacol. 43, 902-908. K. Leung, C. Elmore et al. (2001). Studies on the
119. Murray M. and A.M. Butler (1995). Identification metabolism of troglitazone to reactive intermedi-
of a reversible component in the in vitro inhibition ates in vitro and in vivo. Evidence for novel
of rat hepatic cytochrome P450 2B1 by parathion. biotransformation pathways involving quinone
J. Pharmacol. Exp. Then 272, 639-644. methide formation and thiazolidinedione ring
120. Butler, A.M. and M. Murray (1997). Biotrans- scission. Chem. Res. Toxicol. 14, 62-70.
formation of parathion in human liver: 132. Gitlin, N., N.L. Julie, C.L. Spurr, K.M. Lim, and
Participation of CYP3A4 and its inactivation dur- H.M. Juarbe (1998). Two cases of severe clinical
ing microsomal parathion oxidation. J. Pharmacol. and histologic hepatotoxicity associated with
Exp. Then 280, 966-973. troglitazone. Ann. Intern. Med. 129, 36-38.
121. Murray M. and A.M. Butler (1994). Hepatic bio- 133. Neuschwander-Tetri, B.A., W.L. Isley J.C. Oki,
transformation of parathion: Role of cytochrome S. Ramrakhiani, S.G. Quiason, N.J. Phillips et al.
P450 in NADPH- and NADH-mediated (1998). Troglitazone-induced hepatic failure
Inhibition of Cytochrome P450 Enzymes 301

leading to liver transplantation. A case report. Ann. 144. Kent, U.M., E.S. Roberts, J. Chun, K. Hodge, J.
Intern. Med. 129,38-41. Juncaj, and RE Hollenberg (1998). Inactivation of
134. Chen, Q., J.S. Ngui, G.A. Doss, R.W. Wang, cytochrome P450 2E1 by ?^r^butylisothiocyanate.
X. Cai, F.P. DiNinno et al. (2002). Cytochrome Chem. Res. Toxicol. 11, 1154-1161.
P450 3A4-mediated bioactivation of raloxifene: 145. Chen, L., M. Lee, J.Y Hong, W. Huang, E. Wang,
Irreversible enzyme inhibition and thiol adduct and C.S. Yang (1994). Relationship between
formation. Chem. Res. Toxicol. 15, 907-914. cytochrome P450 2E1 and acetone catabolism in
135. Kempf, D.J., K.C. Marsh, J.F. Denissen, rats as studied with diallyl sulfide as an inhibitor.
E. McDonald, S. Vasavanonda, C.A. Flentge et al. Biochem. Pharmacol. 48, 2199-2205.
(1995), ABT-538 is a potent inhibitor of human 146. Lin, M.C., E.J. Wang, C. Patten, M.J. Lee, E Xiao,
immunodeficiency virus protease and has high K.R. Reuhl et al. (1996). Protective effect of dial-
oral bioavailability in humans. Proc. Natl. Acad. lyl sulfone against acetaminophen-induced
Sci. USA 92, 2484-2488. hepatotoxicity in mice. J. Biochem. Toxicol. 11,
136. Kempf, D.J., K.C. Marsh, G. Kumar, 11-20.
A.D. Rodrigues, J.F. Denissen, E. McDonald 147. Premdas, RD., R.J. Bowers, and RG. Forkert
et al. (1997). Pharmacokinetic enhancement of (2000). Inactivation of hepatic CYP2E1 by an
inhibitors of the human immunodeficiency virus epoxide of diallyl sulfone. J. Pharmacol. Exp.
protease by coadministration with ritonavir. Ther 292,, 1112-1120.
Antimicrob. Agents Chemother. 41, 654-660. 148. Langouet, S., L.L. Furge, N. Kerriguy, K.
137. Koudriakova, T., E. Latsimirskaia, I. Utkin, Nakamura, A. Guillouzo, and ER Guengerich
E. Gangl, R Vouros, E. Storozhuk et al. (1998). (2000). Inhibition of human cytochrome P450
Metabolism of the human immunodeficiency virus enzymes by l,2-dithiole-3-thione, oltipraz and its
protease inhibitors indinavir and ritonavir by derivatives, and sulforaphane. Chem. Res. Toxicol.
human intestinal microsomes and expressed 13,245-252.
cytochrome P4503A4/3A5: Mechanism-based 149. Underwood, M.C., J.R. Cashman, and M.A.
inactivation of cytochrome P4503A by ritonavir. Correia (1992). Specifically designed thiosteroids
Drug Metab. Dispos. 26, 552-561. as active site-directed probes for functional dissec-
138. Smith, T.J., Z. Guo, C. Li, S.M. Ning, RE. Thomas, tion of cytochrome P-450 3A isozymes. Chem.
and C.S. Yang (1993). Mechanisms of inhibition of Res. Tox. 5, 42-53.
4-(methylnitrosamino)-1 -(3-pyridyl)-1 -butanone 150. Stevens, J.C. and J. Halpert (1988). Selective inac-
bioactivation in mouse by dietary phenethyl isoth- tivation of four rat liver microsomal androstene-
iocyanate. Cancer Res. 53, 3276-3282. dione hydroxylases by chloramphenicol analogs.
139. Moreno, R.L., T. Goosen, U.M. Kent, FL. Chung, Mol. Pharmacol. 33, 103-110.
and RE Hollenberg (2001). Differential effects of 151. Halpert, J., J.Y. Jaw, L. Cornfield, C. Balfour, and
naturally occurring isothiocyanates on the activi- E.A. Mash (1989). Selective inactivation of rat
ties of cytochrome P450 2E1 and the mutant P450 liver cytochromes P-450 by 21-chlorinated
2E1 T303A. Arch. Biochem. Biophys. 391, 99-110. steroids. Drug Metab. Dispos. 17, 26-31.
140. Nakajima, M., R. Yoshida, N. Shimada, H. 152. Halpert, J., J.Y. Jaw, C. Balfour, E.A. Mash, and
Yamazaki, and T. Yokoi (2001). Inhibition and E.E Johnson (1988). Selective inactivation by 21-
inactivation of human cytochrome P450 isoforms chlorinated steroids of rabbit liver and adrenal
by phenethyl isothiocyanate. Drug Metab. Dispos. microsomal cytochromes P-450 involved in prog-
29, 1110-1113. esterone hydroxylation. Arch. Biochem. Biophys.
141. Goosen, T.C., U.M. Kent, L. Brand, and RE 264,462-471.
Hollenberg (2000). Inactivation of cytochrome 153. Ortiz de Montellano, PR. (1985). Alkenes and
P450 2B1 by benzyl isothiocyanate, a chemopre- alkynes. In M. Anders (ed.), Bioactivation of
ventative agent from cruciferous vegetables. Foreign Compounds. Academic Press, New York,
Chem. Res. Toxicol. 13, 1349-1359. pp. 121-155.
142. Goosen, T.C., D.E. Mills, and RE Hollenberg 154. De Matteis, E (1978). Loss of liver cytochrome
(2001). Effects of benzyl isothiocyanate on rat and P-450 caused by chemicals. In E De Matteis and
human cytochromes P450: Identification of WN. Aldridge (eds.). Heme and Hemoproteins,
metabolites formed by P450 2B\. J. Pharmacol. Handbook of Experimental Pharmacology, Vol. 44.
Exp Ther 296, 198-206. Springer-Verlag, Berlin, pp. 95-127.
143. Moreno, R.L., U.M. Kent, K. Hodge, and RE 155. Ortiz de Montellano, RR. and M.A. Correia
Hollenberg (1999). Inactivation of cytochrome (1983). Suicidal destruction of cytochrome P-450
P450 2E1 by benzyl isothiocyanate. Chem. Res. during oxidative drug metabolism. Ann. Rev.
Toxicol. 12, 582-587. Pharmacol. Toxicol. 23, 481-503.
302 M.A. Correia and P.R. Ortiz de Montellano

156. Ortiz de Montellano, P.R. and B.A. Mico (1980). 168. Yu, PH., B.A. Davis, and A.A. Boulton (1993).
Destruction of cytochrome P-450 by ethylene and Effect of structural modification of alkyl N-
other olefins. Mol. Pharmacol. 18, 128-135. propargylamines on the selective inhibition of
157. He, K., A.M. Fahck, B. Chen, F. Nilsson, and M.A. monoamine oxidase B activity. Biochem.
Correia (1996). Identification of the heme adduct Pharmacol. 46, 753-757.
and an active site peptide modified during mecha- 169. Sharma, U, E.S. Roberts, and PF Hollenberg
nism-based inactivation of rat liver cytochrome (1996). Inactivation of cytochrome P4502B1 by
P450 2B1 by secobarbital. Chem. Res. Toxicol. 9, the monoamine oxidase inhibitors R-i-)-
614-622. deprenyl and clorgyline. Drug Metab. Dispos. 24,
158. He, K., Y.A. He, G. Szklarz, J.R. Halpert, and M.A. 669-675.
Correia (1996). Secobarbital-mediated inactivation 170. Dyck, L.E. and B.A. Davis (2001). Inhibition of rat
of cytochrome P450 2B1 and its active site liver microsomal CYP1A2 and CYP2B1 activity
mutants: Partitioning between heme and protein by N-(2-heptyl)-N-methyl-propargylamine and
alkylation and epoxidation. J. Biol. Chem. Ill, by N-(2-heptyl)-propargylamine. Drug Metab.
25864-25872. Dispos. 29, 1156-1161.
159. Lukton, D., J.E. Mackie, J.S. Lee, G.S. Marks, and 171. Cutler, A.J., PA. Rose, T.M. Squires, M.K.
PR. Ortiz de Montellano (1988). 2,2-Dialkyl-l,2- Loewen, A.C. Shaw, J.W. Quail et al. (2000).
dihydroquinolines: Cytochrome P-450 catalyzed Inhibitors of abscisic acid 8'-hydroxylase.
N-alkylporphyrin formation, ferrochelatase Biochemistry 39, 13614-13624.
inhibition, and induction of 5-aminolevulinic acid 172. Crowley, J.R. and RE Hollenberg (1995).
synthase activity. Chem. Res. Toxicol. 1, 208-215. Mechanism-based inactivation of rat liver
160. Poulos, T.L., J.R. Cupp-Vickery, and H. Li (1995). cytochrome P4502B1 by phencyclidine and its
In P.R. Ortiz de Montellano (ed.), Cytochrome oxidative product, the iminium ion. Drug Metab.
P450: Structure, Mechanism and Biochemistry. Dispos. 23, 786-793.
Plenum Press, New York, pp. 125-150. 173. Jushchyshyn, M.I., U.M. Kent, and P.F Hollenberg
161. Gotoh, O. (1992). Substrate recognition sites in (2003). The mechanism-based inactivation of
cytochrome P450 family 2 (CYP2) proteins inferred human cytochrome P450 2B6 by phencyclidine.
from comparative analyses of amino acid and coding Drug Metab. Dispos. 31, 46-52.
nucleotide sequences. J. Biol. Chem. 267, 83-90. 174. Ward, DP, A.J. Trevor, A. Kalir, J.D. Adams, TA.
162. Nelson, D.R. and H.W. Strobel (1988). On the Baillie, and N. Castagnoli (1982). Metabolism of
membrane topology of vertebrate cytochrome phencyclidine. The role of iminium ion formation
P-450 proteins. J. Biol. Chem. 263, 6038-6050. in covalent binding to rabbit microsomal protein.
163. von Wachenfeldt, C. and E.F. Johnson (1995). Drug Metab. Dispos. 10, 690-695.
Structures of eukaryotic cytochrome P450 enzymes. 175. Hoag, M.K., A.J. Trevor, Y. Asscher, J. Weissman,
In P.R. Ortiz de Montellano (ed.). Cytochrome and N. Castagnoli (1984). Metabolism-dependent
P450: Structure, Mechanism and Biochemistry. inactivation of liver microsomal enzymes by phen-
Plenum Press, New York, pp. 183-223. cyclidine. Drug Metab. Dispos. 12, 371-375.
164. Ortiz de Montellano, P.R. and E.A. Komives 176. Hoag, M.K.P, A.J. Trevor, A. Kalir, and N.
(1985). Branchpoint for heme alkylation and Castagnoli (1987). NADPH-dependent metabo-
metabolite formation in the oxidation of aryl lism, covalent binding to macromolecules, and
acetylenes. J. Biol. Chem. 260, 3330-3336. inactivation of cytochrome(s) P450. Drug Metab.
165. Roberts, E.S., S.J. Pernecky, W.L. Alworth, and Dispos. 15,485^90.
PF Hollenberg (1996). A role for threonine 302 in 177. Owens, S.M., M. Gunnell, E.M. Laurenzana, and
the mechanism-based inactivation of P450 2B4 by J.L. Valentine (1993). Dose- and time-dependent
2-ethynylnaphthalene. Arch. Biochem. Biophys. changes in phencyclidine metabolite covalent
331, 170-176. binding in rats and the possible role of CYP2D1.
166. Ullrich, V and P Weber (1972). The 0-dealkyla- J. Pharmacol. Exp. Ther 265, 1261-1266.
tion of 7-ethoxycoumarin by liver microsomes. A 178. Hoag, M.K., M. Schmidt-Peetz, P Lampen, A.
direct fluorometric test. Hoppe-Seyler s Z. Physiol. Trevor, and N. Castagnoli (1988). Metabolic stud-
Chem. 353, 1171-1177. ies on phencyclidine: Characterization of a phen-
167. Buters, J.T., CD. Schiller, and R.C. Chou (1993). cyclidine iminium ion metabolite. Chem. Res.
A highly sensitive tool for the assay of cytochrome Toxicol. 1, 128-131.
P450 enzyme activity in rat, dog and man. Direct 179. Osawa, Y and M.J. Coon (1989). Selective mecha-
fluorescence monitoring of the deethylation nism-based inactivation of the major phenobarbi-
of 7-ethoxy-4-trifluoromethylcoumarin. Biochem. tal-inducible P-450 cytochrome from rabbit liver
Pharmacol. 46, 1577-1584. by phencyclidine and its oxidation product, the
Inhibition of Cytochrome P450 Enzymes 303

iminium compound. Drug Metab. Dispos. 17, of tamoxifen: 3,4-dihydroxytamoxifen-o-quinone.


7-13. Chem. Res. Toxicol. 13, 53-62.
180. Hiratsuka, A., T.Y. Chu, E.W. Distefano, L.Y. Lin, 192. Pirmohamed, M., N.R. Kitteringham, T.M.
D.A. Schmitz, and A.K. Cho (1995). Inactivation Guenthner, A.M. Breckenridge, and B.K. Park
of constitutive hepatic cytochromes P450 by phen- (1992). An investigation of the formation of cyto-
cyclidine in the rat. Drug Metab. Dispos. 23, toxic, protein-reactive and stable metabolites from
201-206. carbamazepine in vitro. Biochem. Pharmacol. 43,
181. Brady, J.F., J. Dokko, E.W. Di Stefano, and 1675-1682.
A.K. Cho (1987). Mechanism-based inhibition of 193. Madden, S., J.L. Maggs, and B.K. Park (1996).
cytochrome P-450 by heterocyclic analogues of Bioactivation of carbamazepine in the rat in vivo.
phencyclidine. Drug Metab. Dispos. 15, 648-652. Evidence for the formation of reactive arene
182. Sharma, U, E.S. Roberts, U.M. Kent, S.M. Owens, oxide(s). Drug Metab. Dispos. 24, 469-479.
and RE Hollenberg (1997). Metabolic inactiv- 194. Furst, S.M., R Sukhai, R.A. McClelland, and J.R
ation of cytochrome P4502B1 by phencyclidine: Uetrecht (1995). Covalent binding of carba-
Immunochemical and radiochemical analyses of mazepine oxidative metabolites to neutrophils.
the protective effects of glutathione. Drug Metab. Drug Metab. Dispos. 23, 590-594.
Dispos. 25, 243-250. 195. Ju, C. and J.R Uetrecht (1999). Detection of 2-
183. Bornheim, L.M., E.T. Everhart, J. Li, and M.A. hydroxyiminostilbene in the urine of patients tak-
Correia (1993). Characterization of cannabidiol- ing carbamazepine and its oxidation to a reactive
mediated cytochrome P450 inactivation. Biochem. iminoquinone intermediate. J. Pharmacol. Exp.
Pharmacol. 4S, 1323-1331. Ther 288,51-56.
184. Bornheim, L.M. and M.R Grillo (1998). 196. Wolkenstein, P., C. Tan, S. Lecoeur, J. Wechsler,
Characterization of cytochrome P450 3 A inactiva- N. Garcia-Martin, D. Charue et al. (1998).
tion by cannabidiol: Possible involvement of Covalent binding of carbamazepine reactive
cannabidiol-hydroxyquinone as a P450 inactivator. metabolites to P450 isoforms present in the skin.
Chem. Res. Toxicol. 11, 1209-1216. Chem. Biol. Interact. 113, 39-50.
185. Watanabe, K., N. Usami, I. Yamamoto, and H. 197. Pirmohamed, M., A. Graham, R Roberts, D. Smith,
Yoshimura (1991). Inhibitory effect of cannabidiol D. Chadwick, A.M. Breckenridge et al (1991).
hydroxy-quinone, an oxidative product of Carbamazepine-hypersensitivity: Assessment of
cannabidiol, on the hepatic microsomal drug- clinical and in vitro chemical cross-reactivity
metabolizing enzymes of mice. J. Pharmaco- with phenytoin and oxcarbazepine. Br J. Clin.
biodyn. 14,421^27. Pharmacol. 32, 741-749.
186. Bornheim, L.M. (2000). Effects of unsaturated side- 198. Riley, R.J., G. Smith, C.R. Wolf, VA. Cook, and
chain analogs of tetrahydrocannabinol on J.S. Leeder (1993). Human anti-endoplasmic
cytochromes P450. Biochem. Pharmacol. 60, reticulum autoantibodies produced in aromatic
955-961. anticonvulsant hypersensitivity reactions recog-
187. Pathak, M.A., R Daniels, and T.B. Fitzpatrick nise rodent CYP3A proteins and a similarly regu-
(1962). The presently known distribution of furo- lated human P450 enzyme(s). Biochem. Biophys.
coumarins (psoralens) in plants. J. Invest. Res. Commun. 191, 32-40.
Dermatol. 38, 225-239. 199. Leeder, J.S., A. Gaedigk, X. Lu, and VA. Cook
188. Koenigs, L.L., R.M. Peter, S.J. Thompson, A.E. (1996). Epitope mapping studies with human anti-
Rettie, and W.R Trager (1997). Mechanism-based cytochrome P450 3A antibodies. Mol. Pharmacol.
inactivation of human liver cytochrome P450 2A6 49, 234-243.
by 8-methoxypsoralen. Drug Metab. Dispos. 25, 200. Ueng, YE, T. Kuwabara, Y.J. Chun, and ER
1407-1415. Guengerich (1997). Cooperativity in oxidations
189. Fuhr, U. (1998). Drug interactions with grapefruit catalyzed by cytochrome P450 3A4. Biochemistry
juice. Extent, probable mechanism and clinical rel- 36,370-381.
evance. Drug Saf. 18, 251-272. 201. Kang, R, S.L. Leeder, and M.A. Correia (2003).
190. Lin, J.H., M. Chiba, I.W. Chen, K.J. Vastag, J.A. CYP3A4-mediated bioactivation of carbamazepine.
Nishime, B.D. Dorsey et al. (1995). Time- and \n Proceedings, 12th North American ISSXMeeting.
dose-dependent pharmacokinetics of L-754,394, Providence, Rhode Island, p. 107.
an HIV protease inhibitor, in rats, dogs and mon- 202. Casida, J.E. (1970). Mixed-function oxidase
keys. J. Pharmacol. Exp. Then 274, 264-269. involvement in the biochemistry of insecticide
191. Zhang, E, RW. Fan, X. Liu, L. Shen, R.B. van synergists. J. Agric. Food Chem. 18, 753-772.
Breeman, and J.L. Bolton (2000). Synthesis and 203. Hodgson, E. and R.M. Philpot (1974). Interaction
reactivity of a potential carcinogenic metabolite of methylenedioxyphenyl (1,3-benzodioxole)
304 M.A. Correia and P.R. Ortiz de Montellano

compounds with enzymes and their effects on 215. Mansuy, D., J.P Battioni, J.C. Chottard, and V
mammals. Drug Metab. Rev. 3, 231-301. Ullrich (1979). Preparation of a porphyrin-iron-
204. Wilkinson, C.F., M. Murray, and C.B. Marcus carbene model for the cytochrome P-450 com-
(1984). Interactions of methylenedioxyphenyl com- plexes obtained upon metabolic oxidation of the
pounds with cytochrome P-450 and effects on insecticide synergists of the 1,3-benzodioxole
microsomal oxidation. In E. Hodgson, J.R. Bend, series. J : ^ W . Chem. Soc. 101, 3971-3973.
and R.M. Philpot (eds.). Reviews in Biochemical 216. Dahl, A.R. and E. Hodgson (1979). The interaction
Toxicology, Vol. 6. Elsevier, Amsterdam, pp. 27-63. of aliphatic analogs of methylenedioxyphenyl
205. Kulkarni, A.P., and E. Hodgson (1978). Cumene compounds with cytochromes P-450 and P-420.
hydroperoxide-generated spectral interactions of Chem. Biol. Interact. 27, 163-175.
piperonyl butoxide and other synergists with 217. Anders, M.W, J.M. Sunram, and C.F Wilkinson
microsomes from mammals and insects. Pest. (1984). Mechanism of the metabolism of 1,3-
Biochem. Physiol. 9, 75-83. benzodioxoles to carbon monoxide. Biochem.
206. Elcombe, C.R., J.W. Bridges, R.H. Nimmo-Smith, Pharmacol. 33, 577-580.
and J. Werringloer (1975). Cumene hydroperox- 218. Hansch, C. (1968). The use of homolytic, steric,
ide-mediated formation of inhibited complexes of and hydrophobic constants in a structure-activity
methylenedioxyphenyl compounds with cyto- study of 1,3-benzodioxole synergists. J. Med.
chrome P-450. Biochem. Soc. Trans. 3, 967-970. Chem. 11, 920-924.
207. Franklin, M.R. (1971). The enzymic formation of 219. Hennessy, D.J. (1965). Hydride-transferring abihty
a methylene dioxyphenyl derivative exhibiting an of methylene dioxybenzenes as a basis of synergis-
isocyanide-like spectrum with reduced cytochrome tic activity. J. Agric Food Chem. 13, 218-231.
P-450 in hepatic microsomes. Xenobiotica 1, 220. Cook, J.C. and E. Hodgson (1983). Induction of
581-591. cytochrome P-450 by methylenedioxyphenyl
208. Elcombe, C.R., J.W. Bridges, T.J.B. Gray, R.H. compounds: Importance of the methylene carbon.
Nimmo-Smith, and K.J. Netter (1975). Studies on Toxicol. Appl. Pharmacol. 68, 131-139.
the interaction of safrole with rat hepatic micro- 221. Casida, J.E., J.L Engel, E.G. Essac, EX.
somes. Biochem. Pharmacol. 24, 1427-1433. Kamienski, and S. Kuwatsuka (1966). Methylene
209. Dickins, M., C.R. Elcombe, S.J. Moloney, K.J. '^C-dioxyphenyl compounds: Metabolism in rela-
Netter, and J.W. Bridges (1979). Further studies on tion to their synergistic action. Science 153,
the dissociation of the isosafrole metabolite- 1130-1133.
cytochrome P-450 complex. Biochem. Pharmacol. 222. Kamienski, EX. and J.E. Casida (1970).
28,231-238. Importance of methylenation in the metabolism
210. Ullrich, V and K.H. Schnabel (1973). Formation in vivo and in vitro of methylenedioxyphenyl
and binding of carbanions by cytochrome P-450 of synergists and related compounds in mammals.
liver microsomes. Drug Metab. Dispos. 1, 176-183. Biochem. Pharmacol. 19, 91-112.
211. Ullrich, V. (1977). Mechanism of microsomal 223. Yu, L.-S., C.F Wilkinson, and M.W. Anders
monooxygenases and drug toxicity. In D.J. Jollow, (1980). Generation of carbon monoxide during the
J. Kocsis, R. Snyder, and H. Vaino (eds.), microsomal metabolism of methylenedioxyphenyl
Biological Reactive Intermediates, Plenum Press, compounds. Biochem. Pharmacol. 29, 1113-1122.
New York, pp. 65-82. 224. Metcalf, R.L., C.W. Fukuto, S. Fahmy S. El-Azis,
212. Murray, M., K. Hetnarski, and C.F Wilkinson and E.R. Metcalf (1966). Mode of action of
(1985). Selective inhibitory interactions of carbamate synergists. J. Agric Food. Chem. 14,
alkoxymethylenedioxybenzenes towards mono- 555-562.
oxygenase activity in rat-hepatic microsomes. 225. Greenblatt, D.J., L.L. von Moltke, J.S. Harmatz,
Xenobiotica 15, 369-379. and R.I. Shader (1999). Human cytochromes and
213. Murray, M., C.F. Wilkinson, C. Marcus, and some newer antidepressants: Kinetics, metabolism,
C.E. Dube (1983). Structure-activity relationships and drug interactions. J. Clin. Psychopharmacol.
in the interactions of alkoxymethylenedioxyben- 19(Suppl 1), 23S-35S.
zene derivatives with rat hepatic microsomal 226. Belpaire, FM., P Wijnant, A. Temmerman, B.B.
mixed-function oxidases in vivo. Mol. Pharmacol. Rasmussen, and K. Brosen (1998). The oxidative
24, 129-136. metabolism of metoprolol in human liver micro-
214. Mansuy, D. (1981). Use of model systems in bio- somes: Inhibition by the selective serotonin
chemical toxicology: Heme models. In E. reuptake inhibitors. Eur J. Clin. Pharmacol. 54,
Hodgson, J.R. Bend, and R.M. Philpot (eds.). 261-264.
Reviews in Biochemical Toxicology, Vol. 3. 227. Otton, S.V, S.E. Ball, S.W Cheung, T Inaba, R.L.
Elsevier, Amsterdam, pp. 283-320. Rudolph, and E.M. Sellers (1996). Venlafaxine
Inhibition of Cytochrome P450 Enzymes 305

oxidation in vitro is catalysed by CYP2D6. Br. J. arylhydroxylamines or reduction of nitroarenes


Clin. Pharmacol. 41, 149-156. in situ. Eur J. Biochem. 86, 573-579.
228. Bloomer, J.C, F.R. Woods, R.E. Haddock, M.S. 239. Jonsson, J. and B. Lindeke (1976). On the forma-
Lennard, and G.T. Tucker (1992). The role of tion of cytochrome P-450 product complexes dur-
cytochrome P4502D6 in the metabolism of parox- ing the metabolism of phenylalkylamines. Acta.
etine by human liver microsomes. Br. J. Clin. Pharm.Suec. 13,313-320.
Pharmacol. 33,521-523. 240. Franklin, M.R. (1974). The formation of a 455 nm
229. Sindrup, S.H., K. Brosen, and L.F. Gram (1992). complex during cytochrome P-450-dependent
Pharmacokinetics of the selective serotonin reup- N-hydroxylamphetamine metabolism. Mol.
take inhibitor paroxetine: Nonlinearity and relation Pharmacol. 10, 975-985.
to the sparteine oxidation polymorphism. Clin. 241. Mansuy, D. (1978). Coordination chemistry of
Pharmacol. Ther 51, 288-295. cytochromes P-450 and iron-porphyrins:
230. Sindrup, S.H., K. Brosen, L.F. Gram, J. Hallas, Relevance to pharmacology and toxicology.
E. Skjelbo, A. Allen et al. (1992). The relationship Biochimie. 60, 969-977.
between paroxetine and the sparteine oxida- 242. Lindeke, B., E. Anderson, G. Lundkvist, H.
tion polymorphism. Clin. Pharmacol. Ther. 51, Jonsson, and S.O. Eriksson (1975). Autoxidation
278-287. of N-hydroxyamphetamine and N-hydroxyphen-
231. Bertelsen, K.M., K. Venkatakrishnan, L.L. Von termine: The formation of 2-nitroso-l-phenyl-
Moltke, R.S. Obach, and D.J. Greenblatt (2003). propanes and l-phenyl-2-propanone oxime. Acta
Apparent mechanism-based inhibition of human Pharm. Suec. 12, 183-198.
CYP2D6 in vitro by paroxetine: Comparison with 243. Mansuy, D., R Gans, J.C. Chottard. and J.R Bartoli
fluoxetine and quinidine. Drug Metab. Dispos. 31, (1977). Nitrosoalkanes as Fe(II) ligands in the
289-293. 455-nm-absorbing cytochrome P-450 complexes
232. Haddock, R.E., A.M. Johnson, RE Langley, D.R. formed from nitroalkanes in reducing conditions.
Nelson, J.A. Pope, D.R. Thomas et al. (1989). Eur J. Biochem. 76, 607-615.
Metabolic pathway of paroxetine in animals and 244. Pessayre, D., M. Konstantinova-Mitcheva, V
man and the comparative pharmacological proper- Descatoire, B. Cobert, J.C. Wandscheer, R. Level
ties of its metabolites. Acta. Psychiatr. Scand. 80, et al. (1981). Hypoactivity of cytochrome P-450
24-26. after triacetyloleandomycin administration.
233. Nakajima, M., M. Suzuki, R. Yamaji, H. Biochem. Pharmacol. 30, 559-564.
Takashina, N. Shimada, H. Yamazaki et al. (1999). 245. Wrighton, S.A., R Maurel, E.G. Schuetz, RB.
Isoform selective inhibition and inactivation of Watkins, B. Young, and RS. Guzelian (1985).
human cytochrome P450s by methylenedioxy- Identification of the cytochrome P-450 induced by
phenyl compounds. Xenobiotica 29, 1191-1202. macrolide antibiotics in rat liver as the glucocorti-
234. Sharma, U, E.S. Roberts, and RE Hollenberg coid responsive cytochrome P-450p. Biochemistry
(1996). Formation of a metabolic intermediate 24, 2171-2178.
complex of cytochrome P4502B1 by clorgyline. 246. Watkins, RB., S.A. Wrighton, E.G. Schuetz,
Drug Metab. Dispos. 24, 1247-1253. R Maurel, and RS. Guzelian (1986). Macrolide
235. Franklin, M.R. (1977). Inhibition of mixed- antibiotics inhibit the degradation of the glucocor-
function oxidations by substrates forming reduced ticoid-responsive cytochrome P-450p in rat hepa-
cytochrome P-450 metabolic-intermediate com- tocytes in vivo and in primary monolayer culture.
plexes. Pharmacol. Then A. 2, 227-245. J. Biol. Chem. 261, 6264-6271.
236. Larrey, D, M. Tinel, and D. Pessayre (1983). 247. Zhukov, A. and M. Ingelman-Sundberg (1999).
Formation of inactive cytochrome P450 Fe(II)- Relationship between cytochrome P450 catalytic
metabolite complexes with several erythromycin cycling and stability: Fast degradation of ethanol-
derivatives but not with josamycin and midecamycin inducible cytochrome P450 2E1 (CYP2E1) in
in rats. Biochem. Pharmacol. 32, 1487-1493. hepatoma cells is abolished by inactivation of its
237. Delaforge, M., M. Jaquen, and D. Mansuy (1983). electron donor NADPH-cytochrome P450 reduc-
Dual effects of macrolide antibiotics on rat liver tase. Biochem. J. 34, 453-458.
cytochrome P-450. Induction and formation of 248. Goasduff, T. and A.I. Cederbaum (1999). NADPH-
metabolite-complexes: A structure-activity rela- dependent microsomal electron transfer increases
tionship. Biochem. Pharmacol. 32, 2309-2318. degradation of CYP2E1 by the proteasome com-
238. Mansuy, D., R Beaune, T. Cresteil, C. Bacot, J.C. plex: Role of reactive oxygen species. Arch.
Chottard, and P. Gans (1978). Formation of com- Biochem. Biophys. 370, 258-270.
plexes between microsomal cytochrome P-450- 249. Henderson, C.J., D.M. Otto, D. Carrie, M.A.
Fe(II) and nitrosoarenes obtained by oxidation of Magnuson, A.W McLaren, I. Rosewell et al.
306 M.A. Correia and P.R. Ortiz de Montellano

(2003). Inactivation of the hepatic cytochrome 261. Brady, J.R, H. Ishizaki, J.M. Fukuto, M.C. Lin,
P450 system by conditional deletion of hepatic A. Fadel, J.M. Gapac et al. (1991). Inhibition of
cytochrome P450 reductase. J. Biol. Chem. 278, cytochrome P-450 2E1 by diallyl sulfide and its
13480-13486. metabolites. Chem. Res. Toxicol. 4, 642-647.
250. Hines, R.N. and R.A. Prough (1980). The charac- 262. Collman, J.P, P.D. Hampton, and J.I. Brauman
terization of an inhibitory complex formed with (1986). Stereochemical and mechanistic studies
cytochrome P-450 and a metabolite of 1,1-disub- of the "suicide" event in biomimetic P-450
stituted hydrazines. J. Pharmacol. Then 214, olefin epoxidation. J. Amer Chem. Soc. 108,
80-86. 7861-7862.
251. Muakkasah, S.F., W.R. Bidlack, and W.C.T. Yang 263. Collman, J.P, P.D. Hampton, and J.I. Brauman
(1981). Mechanism of the inhibitory action of iso- (1990). Suicide inactivation of cytochrome P-450
niazid on microsomal drug metabolism. Biochem. model compounds by terminal olefins. Part I: A
Pharmacol. 30, 1651-1658. mechanistic study of heme N-alkylation and epox-
252. Moloney, S.J., B.J. Snider, and R.A. Prough idation. J. Am. Chem. Soc. 112, 2977-2986.
(1984). The interactions of hydrazine derivatives 264. Collman, J.P, P.D. Hampton, and J.I. Brauman
with rat-hepatic cytochrome P-450. Xenobiotica (1990). Suicide inactivation of cytochrome P-450
14, 803-814. compounds by terminal olefins. Part II: Steric and
253. Muakkassah, S.F., W.R. Bidlack, and W.C.T Yang electronic effects in heme N-alkylation and epoxi-
(1982). Reversal of the effects of isoniazid on dation. J ^w. Chem. Soc. 112, 2986-2998.
hepatic cytochrome P-450 by potassium ferri- 265. Mansuy, D., L. Devocelle, I. Artaud, and J.P.
cyanide. Biochem. Pharmacol. 31, 249-251. Battioni (1985). Alkene oxidations by iodosylben-
254. Mahy, J.R, P Battioni, D. Mansuy, J. Fisher, zene catalyzed by iron-porphyrins: Fate of the cat-
R. Weiss, J. Mispelter et al. (1984). Iron alyst and formation of N-alkyl-porphyrin green
porphyrin-nitrene complexes: Preparation from pigments from monosubstituted alkenes as in
1,1-dialkylhydrazines: Electronic structure from cytochrome P-450. Nouv. J. Chim. 9, 711-716.
NMR, Mossbauer, and magnetic susceptibility 266. Artaud, I., L. Devocelle, J.-P Battioni, J.-P Girauh,
studies and crystal structure of the [tetrakis(/?- and D. Mansuy (1987). Suicidal inactivation of
chlorophenyl) porphyrinato-(2,2,6,6-tetramethyl- iron porphyrin catalysts during alk-1-ene oxida-
1-piperidyl) nitrene]iron complex. J. Am. Chem. tion: Isolation of a new type of N-alkylporphyrin.
Soc. 106, 1699-1706. J. Am. Chem. Soc. 109, 3782-3783.
255. Mansuy, D., P Battioni, and J.P Mahy (1982). 267. Traylor, TG., T Nakano, A.R. Mikztal, and B.E.
Isolation of an iron-nitrene complex from the Dunlap (1987). Transient formation of N-alkyl-
dioxygen and iron porphyrin dependent oxidation hemins during hemin-catalyzed epoxidation of
of a hydrazine, ^ ^ w . Chem. Soc. 104, 4487^489. norbornene. Evidence concerning the mechanisms
256. CoUman, J.P, PD. Hampton, and J.I. Brauman ofepoxidation. ^.4A72. Chem. Soc. 109, 3625-3632.
(1986). Stereochemical and mechanistic studies of 268. Traylor, TG., and A.R. Mikztal (1989). Alkene
the "suicide" event in biomimetic P-450 olefin epoxidations catalyzed by iron(III), manganese(III),
epoxidation. J. Am. Chem. Soc. 108, 7861-7862. and chromium(III) porphyrins. Effects of metal and
257. Ortiz de Montellano, PR., R.A. Stearns, and K.C. porphyrin substituents on selectivity and regio-
Langry (1984). The allylisopropylacetamide and chemistry of epoxidation. J. Am. Chem. Soc. I l l ,
novonal prosthetic heme adducts. Mol. Pharmacol. 7443-7448.
25,310-317. 269. Nakano, T, T.G. Traylor, and D. Dolphin (1990).
258. Ortiz de Montellano, PR., B.L.K. Mangold, The formation of N-alkylporphyrins during epoxi-
C. Wheeler, K.L. Kunze, and N.O. Reich (1983). dation of ethylene catalyzed by iron(III) meso-
Stereochemistry of cytochrome P-450-catalyzed tetrakis(2,6-dichlorophenyl)porphyrin. Can J Chem.
epoxidation and prosthetic heme alkylation. 10, 1859-1866.
J. Biol. Chem. 258, 4208-4213. 270. Tian, Z.Q., J.L. Richards, and TG. Traylor (1995).
259. Ortiz de Montellano, PR., K.L. Kunze, H.S. Beilan, Formation of both primary and secondary N-alkyl-
and C. Wheeler (1982). Destruction of cytochrome hemins during hemin-catalyzed epoxidation of ter-
P-450 by vinyl fluoride, fluroxene, and acetylene: minal alkenes. J. Am. Chem. Soc. 117, 21-29.
Evidence for a radical cation intermediate in olefin 271. Blobaum, A.L., U.M. Kent, and PF Hollenberg
oxidation. Biochemistry 21, 1331-1339. (2003). Novel reversible adduction of the P450
260. Kunze, K.L., B.L.K. Mangold, C. Wheeler, H.S. heme: Inactivation of cytochrome P450 2E1
Beilan, and PR. Ortiz de Montellano (1983). The T303A by tert-butyl acetylene. In Proceedings of
cytochrome P-450 active site. J. Biol. Chem. 258, the 13th International Conference on Cytochromes
4202^207. P450. Prague, Czech Republic, p. SI73.
Inhibition of Cytochrome P450 Enzymes 307

272. Dexter, A.F. and L.P. Hager (1995). Transient heme 284. Cole, S.P and G.S. Marks (1984). Ferrochelatase
N-alkylation of chloroperoxidase by terminal and N-alkylated porphyrins. Mol. Cell Biochem.
alkenes and alkynes. J. Am. Chem. Soc. 117, 64, 127-137.
817-818. 285. Ortiz de Montellano, PR., H.S. Beilan, and K.L.
273. Shaik, S., S.P. de Visser, F. Ogliaro, H. Schwarz, Kunze (1981). N-Alkylprotoporphyrin IX forma-
and D. Schroder (2002). Two-state reactivity tion in 3,5-dicarbethoxy-l,4-dihydrocollidine-
mechanisms of hydroxylation and epoxidation by treated rats. Transfer of the alkyl group from the sub-
cytochrome P450 revealed by theory. Curr. Opin. strate to the porphyrin. J. Biol. Chem. 256,
Chem. Biol. 6, 556-567. 6708-6713.
274. Ortiz de Montellano, P.R., and K.L. Kunze (1980). 286. Augusto, O., H.S. Beilan, and PR. Ortiz de
Self-catalyzed inactivation of hepatic cytochrome Montellano (1982). The catalytic mechanism of
P-450 by ethynyl substrates. J. Biol. Chem. 255, cytochrome P-450: Spin-trapping evidence for
5578-5585. one-electron substrate oxidation. J. Biol. Chem.
275. Foroozesh, M., G. Primrose, Z. Guo, L.C. Bell, 257, 11288-11295.
W.L. Alworth, and F.R Guengerich (1997). Aryl 287. De Matteis, E, C. Hollands, A.H. Gibbs, N. de Sa,
acetylenes as mechanism-based inhibitors of and M. Rizzardini (1982). Inactivation of cyto-
cytochrome P450-dependent monooxygenase chrome P-450 and production of N-alkylated
enzymes. Chem. Res. Toxicol. 10, 91-102. porphyrins caused in isolated hepatocytes by sub-
276. Reilly, RE., R.J. Gomi, and S.R. Mason (1999). stituted dihydropyridines: Structural requirements
Mechanism-based inhibition of rat liver microsomal for loss of haem and alkylation of the pyrrole
diazepam C3-hydroxylase by mifepristone associ- nitrogen atom. FEBS Lett. 145, 87-92.
ated with loss of spectrally detectable cytochrome 288. Tephly, T.R., B.L. Coffman, G. Ingall, M.S. Abou
P450. Chem. Biol. Interact. 118, 39-49. Zeit-Har, H.M. Goff, H.D. Tabba et al. (1981).
277. Spitz, I.M. and C.W. Bardin (1993). Mifepristone Identification of N-methylprotoporphyrin IX in
(RU 486)—a modulator of progestin and glucocor- livers of untreated mice and mice treated with 3,5-
ticoid action. N. Engl. J. Med. 329, 404-412. diethoxycarbonyl-l,4-dihydrocollidine: Source of
278. Burger, A., J.E. Clark, M. Nishimoto, A.S. the methyl group. Arch. Biochem. Biophys. Ill,
Muerhoflf, B.S. Masters, and PR. Ortiz de 120-126.
Montellano (1993). Mechanism-based inhibitors 289. De Matteis, E, A.H. Gibbs, PB. Farmer, and J.H.
of prostaglandin omega-hydroxylase: (R)- and (S)- Lamb (1981). Liver production of N-alkylated por-
12-hydroxy-16-heptadecynoic acid and 2,2- phyrins caused by treatment with substituted dihy-
dimethyl-12-hydroxy-16-heptadecynoic acid. J. dropyridines. FEBS Lett. 129, 328-331.
Med. Chem. 36, 1418-1424. 290. De Matteis, E, A.H. Gibbs, and C. Hollands
279. Fan, PW., C. Gu, S.A. Marsh, and J.C. Stevens (1983). N-Alkylation of the haem moiety of
(2003). Mechanism-based inactivation of cytochrome P-450 caused by substituted dihy-
cytochrome P450 2B6 by a novel terminal acety- dropyridines. Preferential attack of different pyr-
lene inhibitor. Drug Metab. Dispos. 31, 28-36. role nitrogen atoms after induction of various
280. De Matteis, E, G. Abbritti, and A.H. Gibbs (1973). cytochrome P-450 isoenzymes. Biochem. J. 211,
Decreased liver activity of porphyrin-metal 455-461.
chelatase in hepatic porphyria caused by 3,5- 291. McCluskey, S.A., G.S. Marks, E.P Sutherland,
diethoxycarbonyl-l,4-dihydrocollidine: Studies in N. Jacobsen, and PR. Ortiz de Montellano (1986).
rats and mice. Biochem. J. 134, 717-727. Ferrochelatase-inhibitory activity and N-alkylpro-
281. De Matteis, E and A. Gibbs (1972). Stimulation of toporphyrin formation with analogues of 3,5-
liver 5-aminolaevulinate synthetase by drugs diethoxycarbonyl-1,4-dihydro-2,4,6-trimethylpyri-
and its relevance to drug-induced accumulation dine (DDC) containing extended 4-alkyl groups:
of cytochrome P-450. Biochem. J. 126, Implications for the active site of ferrochelatase.
1149-1160. Mol. Pharmacol. 30, 352-357.
282. Gayarthri, A.K. and G. Padmanaban (1974). 292. McCluskey, S.A., D.S. Riddick, J.E. Mackie, R.A.
Biochemical effects of 3,5-diethoxycarbonyl-l,4- Kimmett, R.A. Whitney, and G.S. Marks (1992).
dihydrocollidine in mouse liver. Biochem. Inactivation of cytochrome P450 and inhibition of
Pharmacol. 23, 2713-2725. ferrochelatase by analogues of 3,5-diethoxycar-
283. Tephly, T.R., A.H. Gibbs, G. Ingall, and E De bonyl-1,4-dihydro-2,4,6-trimethylpyridine with
Matteis (1980). Studies on the mechanism of 4-nonyl and 4-dodecyl substituents. Can. J.
experimental porphyria and ferrochelatase inhibi- Physiol. Pharmacol. 70, 1069-1074.
tion produced by 3,5-diethoxycarbonyl-l,4-dihy- 293. Lee, J.S., N.E. Jacobsen, and PR. Ortiz de
drocollidine. Int. J. Biochem. 12, 993-998. Montellano (1988). 4-Alkyl radical extrusion in the
308 M.A. Correia and P.R. Ortiz de Montellano

cytochrome P-450-catalyzed oxidation of 4-alkyl- in the microsomal metabolism of alkyl hydrazines


1,4-dihydropyridines. Biochemistry 11,1103-1110. to carbon-centered free radicals. J. Biol. Chem.
294. Bocker, R.H. and RP. Guengerich (1986). Oxidation 266, 21422-21427.
of 4-aryl- and 4-alkyl-substituted 2,6-dimethyl- 305. Jonen, H.G., J. Werringloer, R.A. Prough, and
3,5-bis(alkoxycarbonyl)-1,4-dihydropyridines by R.W. Estabrook (1982). The reaction of phenyl-
human Uver microsomes and immunochemical evi- hydrazine with microsomal cytochrome P-450:
dence for the involvement of a form of cytochrome Catalysis of heme modification. J. Biol. Chem.
?-450. J. Med Chem. 29, 1596-1603.. 257,4404-4411.
295. Tephly, T.R., K.A. Black, M.D. Green, B.L. 306. Mansuy, D., P Battioni, J.E Bartoli, and J.P Mahy
Coffrnan, G.A. Dannan, and F.P. Guengerich (1985). Suicidal inactivation of microsomal
(1986). Effect of the suicide substrate 3,5-diethoxy- cytochrome P-450 by hydrazones. Biochem.
carbonyl-2,6-dimethyl-4-ethyl-1,4-dihydropyridine Pharmacol. 34, 431-432.
on the metabolism of xenobiotics and on 307. Delaforge, M., P Battioni, J.P Mahy, and D.
cytochrome P-450 apoproteins. Mol. Pharmacol. Mansuy (1986). In vivo formation of a-methyl and
29, 81-87. a-phenyl-ferric complexes of hemoglobin and
296. Riddick, D.S., S.S. Park, H.V Gelboin, and G.S. liver cytochrome P-450 upon treatment of rats with
Marks (1990). Effects of 4-alkyl analogues of 3,5- methyl and phenylhydrazine. Chem. Biol. Interact.
diethoxycarbonyl-1,4-dihydro-2,4,6-trimethyl- 60, 101-114.
pyridine on hepatic cytochrome P-450 heme, 308. Raag, R., B.S. Swanson, TL. Poulos, and PR. Ortiz
apoproteins, and catalytic activities following in de Montellano (1990). Formation, crystal structure,
vivo administration to rats. Mol. Pharmacol. 37, and rearrangement of a cytochrome P450^^^ iron-
130-136. phenyl complex. Biochemistry, 29, 8119-8126.
297. McCluskey, S.A., R.A. Whitney, and G.S. Marks 309. Ortiz de Montellano, PR. and K.L. Kunze (1981).
(1989). Evidence for the stereoselective inhibition Formation of N-phenylheme in the hemolytic
of chick embryo hepatic ferrochelatase by N-alky- reaction of phenylhydrazine with hemoglobin. J.
lated porphyrins. Mol. Pharmacol. 36, 608-614. Am. Chem. Soc. 103, 581-586.
298. Kennedy, C.H. and R.P Mason (1990). A reexam- 310. Saito, S. and H.A. Itano (1981). Beta-meso-
ination of the cytochrome P-450-catalyzed free phenylbiliverdin IX-alpha and N-phenylproto-
radical production from dihydropyridine: Evidence porphyrin IX, products of the reaction of
of trace transition metal catalysis. J. Biol. Chem. phenylhydrazine with oxyhemoproteins. Proc.
265, 11425-11428. Natl. Acad. Sci. USA 78, 5508-5512.
299. Ortiz de Montellano, PR. and D.E. Kerr (1985). 311. Augusto, O., K.L. Kunze, and PR. Ortiz de
Inactivation of myoglobin by or^/zo-substituted Montellano (1982). N-Phenylprotoporphyrin IX
aryl hydrazines: Formation of prosthetic heme formation in the hemoglobin-phenylhydrazine reac-
aryl-iron but not N-aryl adducts. Biochemistry 24, tion: Evidence for a protein-stabilized iron-phenyl
1147-1152. intermediate. J. Biol. Chem. 257, 6231-6241.
300. Lukton, D., J.E. Mackie, J.S. Lee, G.S. Marks, and 312. Kunze, K.L. and PR. Ortiz de Montellano (1983).
PR. Ortiz de Montellano (1988). 2,2-Dialkyl-l,2- Formation of a sigma-bonded aryl-iron complex in
dihydroquinolines: Cytochrome P-450 catalyzed the reaction of arylhydrazines with hemoglobin
N-alkylporphyrin formation, ferrochelatase inhibi- and myoglobin. J. Am. Chem. Soc. 105,
tion, and induction of 5-aminolevulinic acid syn- 1380-1381.
thase activity. Chem. Res. Toxicol. 1, 208-215. 313. Ortiz de Montellano, PR. and D.E. Kerr (1983).
301. Muakkassah, W.F., and W.C.T. Yang (1981). Inactivation of catalase by phenylhydrazine:
Mechanism of the inhibitory action of phenelzine Formation of a stable aryl-iron heme complex.
on microsomal drug metabolism. J. Pharmacol. J. Biol. Chem. 258, 10558-10563.
Exp. Ther 219, 147-155. 314. Ringe, D., G.A. Petsko, D.E. Kerr, and PR. Ortiz
302. Ortiz de Montellano, P.R. O. Augusto, F. Viola, and de Montellano (1984). Reaction of myoglobin
K.L. Kunze (1983). Carbon radicals in the metab- with phenylhydrazine: A molecular doorstop.
olism of alkyl hydrazines. J. Biol. Chem. 258, Biochemistry 23, 2—4.
8623-8629. 315. Swanson, B.A. and PR. Ortiz de Montellano
303. Ortiz de Montellano, PR. and M.D. Watanabe (1991). Structure determination and absolute stereo-
(1987). Free radical pathways in the in vitro hepatic chemistry of the four N-phenylprotoporphyrin IX
metabolism of phenelzine. Mol. Pharmacol. 31, regioisomers. J ^w. Chem. Soc. 113, 8146-8153.
213-219. 316. Tuck, S.F., S. Graham-Lorence, J.A. Peterson, and
304. Rumyantseva, G.V, C.H. Kennedy, and R.P. Mason PR. Ortiz de Montellano (1993). Active sites of the
(1991). Trace transition metal-catalyzed reactions cytochrome P450^^^ (CYPlOl) F87W and F87A
Inhibition of Cytochrome P450 Enzymes 309

mutants. Evidence for significant structural reor- 327. Bourdi, M., J.C. Gautier, J. Mircheva, D. Larrey, A.
ganization without alteration of catalytic regio- Guillouzo, C. Andre et al (1992). Anti-liver micro-
specificity. J. Biol Chem. 268, 269-275. somes autoantibodies and dihydralazine-induced
317. Swanson, B.A., D.R. Dutton, C.S. Yang, and P.R. hepatitis: Specificity of autoantibodies and inductive
Ortiz de Montellano (1991). The active sites of capacity of the drug. Mol Pharmacol 42, 280-285.
cytochromes P450 lAl, IIBl, IIB2, and IIEl. 328. Belloc, C., A. Gauffre, C. Andre, and PH. Beaune
Topological analysis by in situ rearrangement of (1997). Epitope mapping of human CYP1A2 in
phenyl-iron complexes J. Biol Chem. 266, dihydralazine-induced autoimmune hepatitis.
19258-19264. Pharmacogenetics 7, 181-186.
318. Swanson, B.A., J.R. Halpert, L.M. Bomheim, and 329. Campbell, C.D. and C.W. Rees (1969). Reactive
RR. Ortiz de Montellano (1992). Topological analy- intermediates. Part III. Oxidation of 1-aminobenzo-
sis of the active sites of cytochromes P450IIB4 (rab- triazole with oxidants other than lead tetra-acetate.
bit), P450IIB10 (mouse) and P450IIB11 (dog) by in J. Chem. Soc. Chem. Commun. 752-756.
situ rearrangement of phenyl-iron complexes. Arch. 330. Ortiz de Montellano, P.R. and J.M. Mathews
Biochem. Biophys. 292, 4 2 ^ 6 . (1981). Autocatalytic alkylation of the
319. Tuck, S.F., J.A. Peterson, and PR. Ortiz de cytochrome P-450 prosthetic haem group by
Montellano (1992). Active site topologies of bacte- 1-aminobenzotriazole: Isolation of an N,N-bridged
rial cytochromes P450 101 (P450^^J, P450 108 benzyne-protoporphyrin IX adduct. Biochem. J.
(P450^^^p, and P450 102 (P4503j^_3): In situ 195, 761-764.
rearrangement of their phenyl-iron complexes. 331. Ortiz de Montellano, PR., J.M. Mathews, and
J. Biol Chem. 267, 5614-5620. K.C. Langry (1984). Autocatalytic inactivation
320. Tuck, S.E, Y. Aoyama, Y. Yoshida, and PR. Ortiz de of cytochrome P-450 and chloroperoxidase by
Montellano (1992). Active site topology of 1-amino- benzotriazole and other aryne precursors.
Saccharomyces cerevisiae lanosterol 14a-demethy- Tetrahedron 40, 511-519.
lase (CYP51) and its A310D mutant (cytochrome 332. Mathews, J.M. and J.R. Bend (1986). N-
P450sQi). / 5/o/. Chem. 267, 13175-13179. Alkylaminobenzotriazoles as isozyme-selective
321. Tuck, S.E, and PR. Ortiz de Montellano (1992). suicide inhibitors of rabbit pulmonary microsomal
Topological mapping of the active sites of cytochrome P-450. Mol Pharmacol 30, 25-32.
cytochromes P4502B1 and P4502B2 by in situ 333. Mathews, J.M. and J.R. Bend (1993). N-Aralkyl
rearrangement of their aryl-iron complexes. derivatives of 1-aminobenzotriazole as potent
Biochemistry 31, 6911-6916. isozyme-selective mechanism-based inhibitors of
322. Tuck, S.E, K. Hiroya, T. Shimizu, M. Hatano, and rabbit pulmonary cytochrome P450 in vivo. J.
P.R. Ortiz de Montellano (1993). The cytochrome Pharmacol Exp. Then 265, 281-285.
P450 1A2 (CYP1A2) active site: Topology and 334. Ortiz de Montellano, PR. and A.K. Costa (1985).
perturbations caused by Glu-318 and Thr-319 Dissociation of cytochrome P450 inactivation and
mutations. Biochemistry 32, 2548-2553. induction. Arch. Biochem. Biophys. 251, 514-524.
323. Battioni, P., J.P Mahy, M. Delaforge, and D. Mansuy 335. Mico, B.A., D.A. Eederowicz, M.G. Ripple, and
(1983). Reaction of monosubstituted hydrazines W. Kerns (1988). In vivo inhibition of oxidative drug
and diazenes with rat-liver cytochrome P-450: metabolism by, and acute toxicity of, 1-aminobenzo-
Formation of ferrous-diazene and ferric sigma-alkyl triazole (ABT). Biochem. Pharmacol 37,
complexes. Eur J. Biochem. 134, 241-248. 2515-2519.
324. Battioni, P, J.-P Mahy, G. Gillet, and D. Mansuy 336. Mugford, C.A., M. Mortillo, B.A. Mico, and
(1983). Iron porphyrin dependent oxidation of J.B. Tarloff (1992). 1-Aminobenzotriazole-induced
methyl- and phenylhydrazine: Isolation of iron(II)- destruction of hepatic and renal cytochromes P450
diazene and sigma-alkyliron (III) (or aryliron(III)) in male Sprague-Dawley rats. Fundam. Appl
complexes. Relevance to the reactions of hemopro- Toxicol 19, 4 3 ^ 9 .
teins with hydrazines. J. Am. Chem. Soc. 105, 337. Xu, D., J.M. Voigt, B.A. Mico, S. Kominami,
1399-1401. S. Takemori, and H.D. Colby (1994). Inhibition of
325. Masubuchi, Y andT. Horie (1998). Dihydralazine- adrenal cytochromes P450 by 1-aminobenzotria-
induced inactivation of cytochrome P450 enzymes zole in vitro. Selectivity for xenobiotic metabo-
in rat liver microsomes. Drug Metab. Dispos. 26, lism. Biochem. Pharmacol 48, 1421-1426.
338-342. 338. Colby, H.D., B. Abbott, M. Cachovic, K.M.
326. Masubuchi, Y and T. Horie (1999). Mechanism- Debolt, and B.A. Mico (1995). Inactivation of
based inactivation of cytochrome P450s 1A2 and adrenal cytochromes P450 by 1-aminobenzotria-
3A4 by dihydralazine in human liver microsomes. zole. Divergence of in vivo and in vitro actions.
Chem. Res. Toxicol 12, 1028-1032. Biochem. Pharmacol 49, 1057-1062.
310 M.A. Correia and P.R. Ortiz de Montellano

339. Woodcroft, K.J., E.W. Szczepan, L.C. Knickle, and 350. Steams, R.A. and PR. Ortiz de Montellano (1985).
J.R. Bend (1990). Three N-aralkylated derivatives Inactivation of cytochrome P450 by a catalytically
of 1-aminobenzotriazole as potent isozyme- generated cyclobutadiene species. J. Am. Chem.
selective mechanism-based inhibitors of guinea Soc. 107, 234-240.
pig pulmonary cytochrome P450 in vitro. Drug 351. Stejskal, R., M. Itabashi, J. Stanek, and Z. Hruban
Metab. Dispos. 18, 1031-1037. (1975). Experimental porphyria induced by 3-[2-
340. Sinai, C.J. and J.R. Bend (1996). Kinetics and (2,4,6-trimethylphenyl)-thioethyl]-4-methyl-
selectivity of mechanism-based inhibition of sydnone. Virchows Arch. 18, 83-100.
guinea pig hepatic and pulmonary cytochrome 352. Ortiz de Montellano, PR. and L.A. Grab (1986).
P450 by N-benzyl-1-aminobenzotriazole and N- Inactivation of cytochrome P-450 during catalytic
alpha-methylbenzyl-1 -aminobenzotriazole. Drug oxidation of a 3-[(arylthio)ethyl]sydnone: N-vinyl
Metab. Dispos. 24, 996-1001. heme formation via insertion into the Fe-N bond.
341. Sinai, C.J., M. Hirst, CD. Webb, and J.R. Bend J.Am. Chem. Soc. 108, 5584-5589.
(1998). Enantioselective, mechanism-based inacti- 353. White, E.H. and N. Egger (1984). Reaction of syd-
vation of guinea pig hepatic cytochrome P450 by nones with ozone as a method of deamination: On
N-(alpha-methylbenzyl)-1 -aminobenzotriazole. the mechanism of inhibition of monoamine oxidase
Drug Metab. Dispos. 26, 681-688. by sydnonQS. J. Am. Chem. Soc. 106, 3701-3703.
342. Sinai, C.J. and J.R. Bend (1995). Isozyme-selective 354. Chevrier, B., R. Weiss, M.C. Lange, J.C. Chottard,
metabolic intermediate complex formation of and D. Mansuy (1981). An iron(III)-porphyrin
guinea pig hepatic cytochrome P450 by N-aralky- complex with a vinylidene group inserted into an
lated derivatives of 1-aminobenzotriazole. Chem. iron-nitrogen bond: Relevance of the structure of
Res. Toxicol. 8,82-91. the active oxygen complex of catalase. J. Am.
343. Moreland, D.E., ET. Corbin, and J.E. McFarland Chem. Soc. 103,2899-2901.
(1993). Effects of safeners on the oxidation of 355. Latos-Grazynski, L., R.J. Cheng, G.N. La Mar, and
multiple substrates by grain sorghum microsomes. A.L. Balch (1981). Reversible migration of an
Pest. Biochem. Physiol. 45, 43-53. axial carbene ligand into an iron-nitrogen bond of
344. Cabanne, E, D. Huby, P Gaillardon, R. Scalla, and a porphyrin: Implications for high oxidation states
E Durst (1987). Effect of the cytochrome P-450 of heme enzymes and heme catabolism. J. Am.
inactivator 1-aminobenzotriazole on the metabo- Chem. Soc. 103,4271-4273.
lism of chlortoluron and isoproturon in wheat. 356. Grab, L.A., B.A. Swanson, and PR. Ortiz de
Pest. Biochem. Biophys. 28, 371-380. Montellano (1988). Cytochrome P-450 inactiva-
345. Feyereisen, R., K.C. Langry, and PR. Ortiz de tion by 3-alkylsydnones: Mechanistic implications
Montellano (1984). Self-catalyzed destruction of of N-alkyl and N-alkenyl heme adduct formation.
insect cytochrome P-450. Insect Biochem. 14,19-26. Biochemistry 27, 4805^814.
346.. Capello, S., L. Henderson, E DeGrazia, D. Liberato, 357. White, I.N.H., A.G. Smith, and PB. Farmer (1983).
W. Garland, and C. Town (1990). The effect of the Formation of N-alkylated protoporphyrin IX in the
cytochrome P-450 suicide inactivator, 1-aminoben- livers of mice after diethylnitrosamine treatment.
zotriazole, on the in vivo metabolism and pharma- Biochem. J. 212, 599-608.
cologic activity of flurazepam. Drug Metab. Dispos. 358. Ding, X. and M.J. Coon (1988). Cytochrome
18,190-196. P-450-dependent formation of ethylene from N-
347. Kaikaus, R.M., W.K. Chan, N. Lysenko, R. Ray, nitrosoethylamines. Drug Metab. Dispos. 16,
PR. Ortiz de Montellano, and N.M. Bass (1993). 265-269.
Induction of peroxisomal fatty acid p-oxidation 359. Frater, Y., A. Brady, E.A. Lock, and F De Matteis
and liver fatty acid-binding protein by peroxisome (1993). Formation of N-methyl protoporphyrin in
proliferators: Mediation via the cytochrome chemically-induced protoporphyria. Studies with a
P450IVA1 a)-hydroxylase pathway. J. Biol. Chem. novel porphyrogenic agent. Arch. Toxicol. 67,
268, 9593-9603. 179-185.
348. Su, P, K.M. Kaushal, and D.L. Kroetz (1998). 360. De Matteis, F and A.H. Gibbs (1980). Drug-
Inhibition of renal arachidonic acid omega- induced conversion of liver haem into modified
hydroxylase activity with ABT reduces blood porphyrins. Biochem. J. 187, 285-288.
pressure in the SHR. Am. J. Physiol. 275, 361. Holley, A. E., Y. Frater, A.H. Gibbs, F De Matteis,
R426-R438. J.H. Lamb, PB. Farmer et al. (1991). Isolation of
349. Whitman, D.W. and B.K. Carpenter (1980). two N-monosubstituted protoporphyrins, bearing
Experimental evidence for nonsquare cyclobutadi- either the whole drug or a methyl group on the
ene as a chemically significant intermediate in pyrrole nitrogen atom, from liver of mice given
solution. J. Am. Chem. Soc. 102, 4272-4274. griseofulvin. Biochem. J. 274, 843-848.
Inhibition of Cytochrome P450 Enzymes 311

362. Gibbs, A.H., S. Naylor, J.H. Lamb, Y. Prater, 374. Lozano, J.J., E. Lopez-de-Brinas, N.B. Centeno,
F. De Matteis (1990). Copper-induced dealkylation R. Guigo, and F Sanz (1997). Three-dimensional
studies of biologically N-alkylated porphyrins by modelling of human cytochrome P450 1A2 and its
fast atom bombardment mass spectrometry. Anal. interaction with caffeine and MelQ. J. Comput.
Chim. Acta 241, 233-239. Aided Mol. Des. 11, 395-408.
363. De Matteis, F. and G.S. Marks (1996). Cytochrome 375. Nichols, W.K., D.N. Larson, and G.S. Yost (1990).
P450 and its interactions with the heme biosynthetic Bioactivation of 3-methylindole by isolated
pathway. Can. J. Physiol. Pharmacol. 74, 1-8. rabbit lung cells. Toxicol. Appl. Pharmacol. 105,
364. Bellingham, R.M.A., A.H. Gibbs, F De Matteis, 264-270.
L.-Y. Lian, and G.C.K. Roberts (1995). 376. Rettie, A.E., A.W. Rettenmeier, W.N. Howald, and
Determination of the structure of an N-substituted T.A. Baillie (1987). Cytochrome P-450-catalyzed
protoporphyrin isolated from the livers of griseo- formation of delta 4-VPA, a toxic metabolite of
fulvin-fed mice. Biochem. J. 307, 505-512. valproic acid. Science 235, 890-893.
365. Holley, A., L.J. King, A.H. Gibbs, and F De 377. Skordos, K.W, S.J. Smeal, C.A. Reilly, D.L.
Matteis (1990). Strain and sex differences in the Lanza, and G.S. Yost (2003). Selective dehydro-
response of mice to drugs that induce protopor- genated intermediates are mechanism-based inac-
phyria: Role of porphyrin biosynthesis and tivators of CYP3A4, CYP2E1, and CYP2F1. In
removal. J. Biochem. Toxicol. 5, 175-182. Proceedings of the J 3th International Conference
366. De Matteis, F, A.H. Gibbs, S.R. Martin, and on Cytochromes P450. Prague, Czech Republic,
R.L.B. Milek (1991). Labeling in vivo and chiral- p. S127.
ity of griseofulvin-derived N-alkylated protopor- 378. Koop, D.R. (1990). Inhibition of ethanol-inducible
phyrins. Biochem. J. 280, 813-816. cytochrome P450IIE1 by 3-amino-l,2,4-triazole.
367. Kobus, S.M., S.G. Wong, and G.S. Marks (2001). Chem. Res. Toxicol. 3, 377-383.
Isolation of regioisomers of N-alkylprotopor- 379. Voorman, R.L., S.M. Maio, N.A. Payne, Z. Zhao,
phyrin IX from chick embryo liver after treatment K.A. Koeplinger, and X. Wang (1998).
with porphyrinogenic xenobiotics. Can. J. Physiol. Microsomal metabolism of delavirdine: Evidence
Pharmacol. 79,814-821. for mechanism-based inactivation of human
368. Kunze, K.L. and W.F Trager (1993). Isoform- cytochrome P450 3A4. J. Pharmacol. Exp. Ther.
selective mechanism-based inhibition of human 287, 381-388.
cytochrome P450 1A2 by fiirafylline. Chem. Res. 380. Guzelian, PS. and R.W Swisher (1979).
Toxicol. 6, 649-656. Degradation of cytochrome P-450 haem by carbon
369. Clarke, S.E., A.D. Ayrton, and R.J. Chenery tetrachloride and 2-allyl-2-isopropylacetamide in
(1994). Characterization of the inhibition of rat liver in vivo and in vitro: Involvement of
P4501A2 by ftirafylline. Xenobiotica 24, 517-526. non-carbon monoxide-forming mechanisms.
370. Tarrus, E., J. Cami, D.J. Roberts, R.G. Spickett, Biochem. J. 184,481^89.
E. Celdran, and J. Segura (1987). Accumulation of 381. Davies, H.S., S.G. Britt, and L.R. Pohl (1986).
caffeine in healthy volunteers treated with Carbon tetrachloride and 2-isopropyl-4-penten-
ftirafylline. Br J. Clin. Pharmacol. 23, 9-18. amide-induced inactivation of cytochrome P-450
371. Boobis, A.R., A.M. Lynch, S. Murray, R.R. De la leads to heme-derived protein adducts. Arch.
Torre, A. Solans, M. Farre, J. Segura et al. (1994). Biochem. Biophys. 244, 387-352.
CYPlA2-catalyzed conversion of dietary hetero- 382. Osawa, Y. and L.R. Pohl (1989). Covalent bonding
cyclic amines to their proximate carcinogens is of the prosthetic heme to protein: A potential mech-
their major route of metabolism in humans. anism for the suicide inactivation or activation of
Cancer Res. 54, 89-94. hemoproteins. Chem. Res. Toxicol. 2, 131-141.
372. Racha, J.K., A.E. Rettie, and K.L. Kunze (1998). 383. Correia, M.A., C. Decker, K. Sugiyama, P.
Mechanism-based inactivation of human cyto- Caldera, L. Bomheim, S.A. Wrighton et al. (1987).
chrome P450 1A2 by ftirafylline: Detection of a Degradation of rat hepatic cytochrome P-450
1:1 adduct to protein and evidence for the forma- heme by 3,5-dicarbethoxy-2,6-dimethyl-4-ethyl-
tion of a novel imidazomethide intermediate. 1,4-dihydropyridine to irreversibly bound
Biochemistry 31, 7407-7419. protein adducts. Arch. Biochem. Biophys. 258,
373. Lewis, D.F and B.G. Lake (1996). Molecular mod- 436-451.
elling of CYPl A subfamily members based on an 384. Sugiyama, K., K. Yao, A.E. Rettie, and M.A.
alignment with CYPl 02: Rationalization of Correia (1989). Inactivation of rat hepatic
CYPl A substrate specificity in terms of active cytochrome P-450 isozymes by 3,5-dicarbethoxy-
site amino acid residues. Xenobiotica 26, 2,6-dimethyl-4-ethyl-1,4-dihydropyridine. Chem.
723-753. Res. Toxicol. 2, 400-410.
312 M.A. Correia and P.R. Ortiz de Montellano

385. Riddick, D.S. and G.S. Marks (1990). Irreversible prosthetic heme of human hemoglobin by BrCC^.
binding of heme to microsomal protein during Cross-linking of heme to cysteine residue 93.
inactivation of cytochrome P450 by alkyl ana- J. Biol. Chem. 267, 8739-8743.
logues of 3,5-diethoxycarbonyl-l,4-dihydro-2,4,6- 397. Docherty, J.C, G.D. Firneisz, and B.A. Schacter
trimethylpyridine. Biochem. Pharmacol. 40, (1984). Methene bridge carbon atom elimination
1915-1921. in oxidative heme degradation catalyzed by heme
386. Guengerich, F.P. (1978). Destruction of heme and oxygenase and NADPH-cytochrome P-450 reduc-
hemoproteins mediated by liver microsomal tase. Arch. Biochem. Biophys. 235, 657-664.
reduced nicotinamide adenine dinucleotide phos- 398. Yoshinaga, T., S. Sassa, and A. Kappas (1982). A
phate-cytochrome P-450 reductase. Biochemistry comparative study of heme degradation by NADPH-
17, 3633-3639. cytochrome C reductase alone and by the complete
387. Guengerich, P. (1986). Covalent binding to apopro- heme oxygenase system. Distinctive aspects of heme
tein is a major fate of heme in a variety of reactions degradation by NADPH-cytochrome c reductase.
in which cytochrome P-450 is destroyed. Biochem. J. Biol. Chem. 257, 7794-7802.
Biophys. Res. Commun. 138,193-198. 399. Cantoni, L., A.H. Gibbs, and R De Matteis (1981).
388. Schaefer, W.H., T.M. Harris, and F.P Guengerich Loss of haem and haemoproteins during the gener-
(1985). Characterization of the enzymatic and non- ation of superoxide anion and hydrogen peroxide:
enzymatic peroxidative degradation of iron por- A pathway not involving production of carbon
phyrins and cytochrome P-450 heme. Biochemistry monoxide. Int. J. Biochem. 13, 823-830.
24, 3254-3263. 400. Bonnett, R. and J.C.M. Stewart (1975).
389. Yao, K., A.M. Falick, N. Patel, and M.A. Correia Photooxidation of bilirubin in hydroxylic solvents.
(1993). Cumene hydroperoxide-mediated inactiva- J. Chem. Soc, Perkin Trans. 1, 224-229.
tion of cytochrome P450 2B1: Identification of an 401. Tierney, D.J., A.L. Haas, and D.R. Koop (1992).
active site heme-modified peptide. J. Biol. Chem. Degradation of cytochrome P450 2E1: Selective
268, 59-65. loss after labilization of the enzyme. Arch.
390. He, K., L.M. Bornheim, A.M. FaHck, D. Maltby Biochem. Biophys. 293, 9-16.
H. Yin, and M.A. Correia (1998). Identification of 402. Correia, M.A., K. Yao, S.A. Wrighton, D.J.
the heme-modified peptides from cumene hydro- Waxman, and A. Rettie (1992). Differential
peroxide-inactivated cytochrome P450 3A4. apoprotein loss of rat liver cytochromes P450
Biochemistry 31, 17448-17457. after their inactivation by 3,5-dicarbethoxy-2,6-
391. Karuzina, 1.1. and A.I. Archakov (1994). The oxida- dimethyl-4-ethyl-l,4-dihydropyridine: A case for
tive inactivation of cytochrome P450 in monooxy- distinct proteolytic mechanisms? Arch. Biochem.
genase reactions. Free Rad. Biol. Med. 16, 73-97. Biophys. 294, 493-503.
392. Catalano, C.E., Y.S. Choe, and PR. Ortiz de 403. Correia, M.A., S.H. Davoll, S.A. Wrighton, and
Montellano (1989). Reactions of the protein radical RE. Thomas (1992). Degradation of rat liver
in peroxide-treated myoglobin: Formation of a heme- cytochromes P-450 3A after their inactivation by
protein cross-link. ^ 5/o/. Chem. 264, 10534-10541. 3,5-dicarbethoxy-2,6-dimethyl-4-ethyl-1,4-dihy-
393. Choe, Y.S. and PR. Ortiz de Montellano (1991). dropyridine: Characterization of the proteolytic
Differential additions to the myoglobin pros- system. Arch. Biochem. Biophys. 297, 228-238.
thetic heme group. Oxidative 7-meso substitu- 404. Korsmeyer, K.K., S. Davoll, M.E. Figueiredo-
tion by alkylhydrazines. J. Biol. Chem. 266, Pereira, and M.A. Correia (1999). Proteolytic
8523-8530. degradation of heme-modified hepatic
394. Osawa, Y, B.M. Martin, PR. Griffin, J.R. cytochromes P450: A role for phosphorylation,
Yates III, J. Shabanowitz, D.F Hunt et al. (1990). Ubiquitination and the 26S Proteasome? Arch.
Metabolism-based covalent bonding of the heme Biochem. Biophys. 365, 3 1 ^ 4 .
prosthetic group to its apoprotein during the reduc- 405. Wang, H., M.E. Figueiredo-Pereira, and M.A.
tive debromination of BrCC13 by myoglobin. Correia (1999). CYP 3A degradation in isolated
J. Biol. Chem. 265, 10340-10346. rat liver hepatocytes: 26S proteasome inhibitors as
395. Osawa, Y, R.J. Highet, A. Bax, and L.R. Pohl probes. Arch. Biochem. Biophys. 365, 45-53.
(1991). Characterization by NMR of the heme- 406. Correia, M.A., K. Sugiyama, and K. Yao (1989).
myoglobin adduct formed during the reductive Degradation of rat hepatic cytochrome P450p.
metabolism of BrCC^. Covalent bonding of the DrugMetab. Rev. 20, 615-628.
proximal histidine to the ring 1 vinyl group. J. Biol. 407. Raner, G.M., A.D. Vaz, and M.J. Coon (1996).
Chem. 266,3208-3214. Metabolism of all-trans, 9-cis, and 13-cis isomers
396. Kindt, J.T., A. Woods, B.M. Martin, R.J. Cotter, of retinal by purified isozymes of microsomal
and Y Osawa (1992). Covalent alteration of the cytochrome P450 and mechanism-based inhibition
Inhibition of Cytochrome P450 Enzymes 313

of retinoid oxidation by citral. Mol. Pharmacol. mechanism-based inactivation of CYP1A2. J.


49,515-522. Pharmacol. Exp. Ther 299, 874-882.
408. Raner, G.M., E.W. Chiang, A.D. Vaz, and M.J. 421. Voorman, R.L., N.A. Payne, L.C. Wienkers, M.J.
Coon (1997). Mechanism-based inactivation of Hauer, and PE. Sanders (2001). Interaction of
cytochrome P450 2B4 by aldehydes: Relationship delavirdine with human liver microsomal cyto-
to aldehyde deformylation via a peroxyhemiacetal chrome P450: Inhibition of CYP2C9, CYP2C19,
intermediate. Biochemistry 36, 4895-4902. and CYP2D6. Drug Metab. Dispos. 29, 4 1 ^ 7 .
409. Raner, G.M., J.A. Hatchell, M.U. Dixon, T.L. Joy, 422. Palamanda, J.R., C.N. Casciano, L.A. Norton, R.P
A.E. Haddy, and E.R. Johnson, (2002). Regio- Clement, RV Favreau, C.C. Lin et al. (2001).
selective peroxo-dependent heme alkylation in Mechanism-based inactivation of CYP2D6 by 5-
P450BM3-F87G by aromatic aldehydes: Effects fluoro-2-[4-[(2-phenyl-l//-imidazol-5-yl)methyl]-
of alkylation on catalysis. Biochemistry 41, l-piperazinyl]pyrimidine. Drug Metab. Dispos. 29,
9601-9610. 863-867.
410. Kuo, C.L., G.M. Raner, A.D. Vaz, and M.J. Coon 423. Nerland, D.E., M.M. Iba, and G.J. Mannering
(1999). Discrete species of activated oxygen yield (1981). Use of linoleic acid hydroperoxide in the
different cytochrome P450 heme adducts from determination of absolute spectra of membrane-
aldehydes. Biochemistry 38, 10511-10518. bound cytochrome P450. Mol. Pharmacol. 19,
411. Kuo, C.L., A.D. Vaz, and M.J. Coon (1997). 162-167.
Metabolic activation of trans-4-hydroxy-2-none- 424. Poli, G., K. Cheeseman, T.R Slater, and M.U.
nal, a toxic product of membrane lipid peroxida- Danzani (1981). The role of lipid peroxidation
tion and inhibitor of P450 cytochromes. J. Biol. in CCl4-induced damage to liver microsomal
Chem. 272,22611-22616. enzymes: Comparative studies in vitro using
412. Ortiz de Montellano, PR. and K.L. Kunze (1980). microsomes and isolated liver cells. Chem. Biol.
Inactivation of hepatic cytochrome P-450 by Interact. 2^1, 13-24.
allenic substrates. Biochem. Biophys. Res. 425. De Groot, H. and W. Haas (1981). Self-catalyzed
Commun. 94, 443-449. 02-independent inactivation of NADPH- or
413. Hanzlik, R.P, V Kishore, and R. Tullman (1979). dithionite-reduced microsomal cytochrome P-450
Cyclopropylamines as suicidesubstrates for cyto- by carbon tetrachloride. Biochem. Pharmacol. 30,
chromes P-450. J. Med. Chem. 22, 759-761. 2343-2347.
414. Macdonald, T.L., K. Zirvi, L.T. Burka, P. Peyman, 426. Fernandez, G., M.C. Villaruel, E.G.D. de Toranzo,
and P.P. Guengerich (1982). Mechanism of and J.A. Castro (1982). Covalent binding of carbon
cytochrome P-450 inhibition by cyclopropy- to the heme moiety of cytochrome P-450 and its
lamines. J. Am. Chem. Soc. 104, 2050-2052. degradation products. Res. Commun. Chem.
415. Ortiz de Montellano, PR. and J.M. Mathews Pathol. Pharmacol. 35, 283-290.
(1981). Inactivation of hepatic cytochrome P-450 427. De Groot, H., U. Harnisch, and T. Noll (1982).
by a 1,2,3-benzothiadiazole insecticide synergist. Suicidal inactivation of microsomal cytochrome
Biochem. Pharmacol. 30, 1138-1141. P-450 by halothane under hypoxic conditions.
416. Babu, B.R. and A.D.N. Vaz (1997). 1,2,3- Biochem. Biophys. Res. Commun. 107, 885-891.
Thiadiazole: A novel heterocyclic heme ligand for 428. Reiner, O. and H. Uehleke (1971). Bindung von
the design of cytochrome P450 inhibitors. Tetrachlorkohlenstoff an reduziertes mikrosomales
Biochemistry 36, 7209-7216. Cytochrome P-450 und an Ham. Hoppe-Seylers Z.
417. Masubuchi, Y., A. Ose, and T. Horie (2001). Physiol. Chem. 352, 1048-1052.
Diclofenac-induced inactivation of CYP3A4 and 429. Cox, PJ., L.J. King, and D.V Parke (1976). The
its stimulation by quinidine. Drug Metab. Dispos. binding of trichlorofluoromethane and other
30, 1143-1148. haloalkanes to cytochrome P-450 under aerobic
418. Shen, S., S.J. Hargus, B.M. Martin, and L.R. Pohl and anaerobic conditions. Xenobiotica 6, 363-375.
(1997). Cytochrome P4502C11 is a target of 430. Roland, W.C, D. Mansuy, W. Nastainczyk, G.
diclofenac covalent binding in rats. Chem. Res. Deutschmann, and V Ullrich (1977). The
Toxicol. 10, AIQ^IZ. reduction of polyhalogenated methanes by liver
419. Chun, Y.J., S.Y Ryu, T.C. Jeong, and M.Y Kim microsomal cytochrome P450. Mol. Pharmacol.
(2001). Mechanism-based inhibition of human 13, 698-705.
cytochrome P450 lAl by rhapontigenin. Drug. 431. Mansuy, D. and M. Fontecave (1983). Reduction of
Metab. Dispos. 29, 389-393. benzyl halides by liver microsomes: Formation of
420. Chang, T.K.H., J. Chen, and W.B.K. Lee (2001). 478 nm-absorbing sigma-alkyl-ferric cytochrome
Differential inhibition and inactivation of human P-450 complexes. Biochem. Pharmacol. 32,
CYPl enzymes by trans-resveratrol: Evidence for 1871-1879.
314 M.A. Correia and P.R. Ortiz de Montellano

432. Mansuy, D., M. Lange, J.C. Chottard, IF. Bartoli, 443. Guengerich, FR, G.A. Dannan, T.S. Wright, M.V
B. Chevrier, and R. Weiss (1978). Dichlorocarbene Martin, and L.S. Kaminsky (1982). Purification
complexes of iron(II)-porphyrins—Crystal and and characterization of liver microsomal cyto-
molecular structure of Fe(TPP)(CCl2)(H20). chromes P-450: Electrophoretic, spectral, cat-
Angew. Chem. Int. Ed. Engl. 17, 781-782. alytic, and immunochemical properties and
433. Ahr, H.J., L.J. King, W. Nastainczyk, and V Ullrich inducibility of eight isozymes isolated from rats
(1980). The mechanism of chloroform and carbon treated with phenobarbital or beta-naphthoflavone.
monoxide formation from carbon tetrachloride Biochemistry 21, 6019-6030.
by microsomal cytochrome P-450. Biochem. 444. Halpert, J.R. (1995). Structural basis of selective
Pharmacol. 29, 2855-2861. cytochrome P450 inhibition. Annu. Rev. Pharmacol.
434. Mansuy, D., M. Lange, J.C. Chottard, and J.F. Toxicol. 35, 29-53.
Bartoli (1978). Reaction du complexe carbenique 445. Kim, S., H. Ko, J.E. Park, S. Jung, S.K. Lee, and
Fe(II)(tetraphenylporphyrine)(CCl2) avec les Y. Chun (2002). Design, synthesis, and discovery
amines primaires: Formation d'isonitriles. of novel ^ra«5-stilbene analogues as potent and
Tetrahedron Lett. 33, 3027-3030. selective human cytochrome P450 IBl inhibitors.
435. Mansuy, D. and J.-R Battioni (1982). Isolation of J.Med Chem. 45, 160-164.
sigma-alkyl-iron(III) or carbene-iron(II) com- 446. Covey, D.F (1988). Aromatase inhibitors: Specific
plexes from reduction of polyhalogenated com- inhibitors of oestrogen biosynthesis. In Berg and
pounds by iron(II)-porphyrins: The particular case Plempel (eds). Sterol Biosynthesis Inhibitors. Ellis
of halothane CF3CHClBr. J. Chem. Soc. Chem. Horwood Ltd., Cambridge, pp. 534-571.
Commun. 638-639. 447. Henderson, D., U.-F. Habenicht, Y. Nishino,
436. Ruf, H.H., H. Ahr, W. Nastainczyk, V. Ullrich, D. U. Kerb, and M.F El Etreby (1986). Aromatase
Mansuy, J.-R Battioni (1984). Formation of a inhibitors and benign prostatic hyperplasia.
ferric carbanion complex from halothane and J. Steroid Biochem. 25, 867-876.
cytochrome P-450: Electron spin resonance, elec- 448. Van Wauwe, J.P and PA.J. Janssen (1989). Is there
tronic spectra and model complexes. Biochemistry a case for P-450 inhibitors in cancer treatment.
23,5300-5306. J. Med Chem. 32, 2231-2239.
437. Castro, C.E., R.S. Wade, and N.O. Belser (1985). 449. Kellis, J.T, J.J. Sheets, and L.E. Vickery (1984).
Biodehalogenation: Reactions of cytochrome P- Amino-steroids as inhibitors and probes of the
450 with polyhalomethanes. Biochemistry 24, active site of cytochrome P-450^^^,. Effects on the
204-210. enzyme from different sources. J. Steroid Biochem.
438. Callot, H. J. and E. Schefifer (1980). Model for the in 20,671-676.
vitro transformation of cytochrome P-450 into 450. Sheets, J.J. and L.E. Vickery (1983). Active site-
"green pigments." Tetrahedron Lett. 21, 1335-1338. directed inhibitors of cytochrome P-450g^^:
439. Lange, M. and D. Mansuy (1981). N-Substituted Structural and mechanistic implications of a side
porphyrin formation from carbene iron-porphyrin chain-substituted series of amino-steroids. J. Biol.
complexes: A possible pathway for cytochrome Chem. 258, 11446-11452.
P-450 heme destruction. Tetrahedron Lett. 22, 451. Sheets, J.J. and L.E. Vickery (1982). Proximity of
2561-2564. the substrate binding site and the heme-iron cat-
440. Chevrier, B., R. Weiss, M. Lange, J.C. Chotard, and alytic site in cytochrome P-450g^^. Proc. Natl.
D. Mansuy (1981). An iron(III)-porphyrin com- Acad Sci. USA 79, 5773-5777.
plex with a vinylidene group inserted into an 452. Nagahisa, A., T Foo, M. Gut, and WH. Orme-
iron-nitrogen bond: Relevance to the structure of Johnson (1985). Competitive inhibition of
the active oxygen complex of catalase. J. Am. cytochrome P-450scc by (IIR)- and (22^-22-
Chem. Soc. 103,2899-2901. aminocholesterol: Side chain stereochemical
441. Olmstead, M.M., R.-J. Cheng, and A.L. Balch requirements for C-22 amine coordination to the
(1982). X-ray crystallographic characterization of active-site heme. J. Biol. Chem. 260, 846-851.
an iron porphyrin with a vinylidene carbene 453. Vickery, L.E. and J. Singh (1988). 22-Thio-23,24-
inserted into an iron-nitrogen bond. Inorg. Chem. bisnor-5-cholen-3p-ol: An active site-directed
21,4143^148. inhibitor of cytochrome P450g^^. J. Steroid Biochem.
442. Falzon, M., A. Nielsch, and M.D. Burke (1986). 29, 539-543.
Denaturation of cytochrome P-450 by indo- 454. Nagahisa, A., R.W Spencer, and WH. Orme-
methacin and other non-steroidal anti-inflamma- Johnson (1983). Acetylenic mechanism-based
tory drugs: Evidence for a surfactant mechanism inhibitors of cholesterol side chain cleavage by
and a selective effect of a p-chlorophenyl moiety. cytochrome P-450^^^. J. Biol. Chem. 258,
Biochem. Pharmacol. 35, 4019-4024. 6721-6723.
Inhibition of Cytochrome P450 Enzymes 315

455. Olakanmi, O. and D.W. Seybert (1990). Modified 470. Santen, R.J., T.J. Worgul, E. Samojlik, A.
acetylenic steroids as potent mechanism-based Interrante, A.E. Boucher, A. Lipton et al. (1981). A
inhibitors of cytochrome P-450^^^. J. Steroid randomized trial comparing surgical adrenalec-
Biochem. 36, 273-280. tomy with aminoglutethimide plus hydrocortisone
456. Krueger, R.J., A. Nagahisa, M. Gut, S.R. Wilson, in women with advanced breast cancer. Engl.
and W.H. Orme-Johnson (1985). Effect of P-450^^^ ^MeJ. 305, 545-551.
inhibitors on corticosterone production by rat 471. Harris, A.L., T.J. Powles, I.E. Smith, R.C.
adrenal cells. J. Biol. Chem. 260, 852-859. Coombes, H.T Ford, J.C. Gazet et al. (1983).
457. Nagahisa, A., W.H. Orme-Johnson, and S.R. Aminoglutethimide for the treatment of advanced
Wilson (1984). Silicon mediated suicide inhibi- postmenopausal breast cancer. Eur J. Cancer Clin.
tion: An efficient mechanism-based inhibitor of Oncol. 19, 11-17.
cytochrome P-450g^^ oxidation of cholesterol. J. 472. Foster, A.B., M. Jarman, C.S. Leung, M.G.
Am. Chem. Soc. 106, 1166-1167. Rowlands, G.N. Taylor, R.G. Plevey et al. (1985).
458. Trahanovsky, WS. and A.L. Himstedt (1974). Analogues of aminoglutethimide: Selective inhibi-
Oxidation of organic compounds with cerium(lV). tion of aromatase. J. Med. Chem. 28, 200-204.
XX. Abnormally rapid rate of oxidative clea- 473. Foster, A.B., M. Jarman, C.-S. Leung, M.G.
vage of (beta-trimethylsilylethyl)-phenylmethanol. Rowlands, and G.N. Taylor (1983). Analogues of
J. Am. Chem. Soc. 96, 7974-7976. aminoglutethimide: Selective inhibition of choles-
459. Vickery, L.E. and J. Singh (1988). 22-Thio-23,24- terol side-chain cleavage. J. Med. Chem. 26,
bisnor-5-cholen-3p-ol: An active site-directed 50-54.
inhibitor of cytochrome P450g^,^.. J. Steroid Biochem. 474. Bhatnagar, A.S., A. Hausler, K. Schieweck, L.J.
29, 539-543. Browne, R. Bowman, and R.E. Steele (1990).
460. Miao, E., C. Zuo, A. Nagahisa, B.J. Taylor, Novel aromatase inhibitors. J. Steroid Biochem.
S. Joardar, C. Byon et al. (1990). Cytochrome Mol. Biol. 37, 363-367.
P450^^^ mediated oxidation of (205)-22-nor-22- 475. Lipton, A., H.A. Harvey, L.M. Demers, J.R.
thiacholesterol: Characterization of mechanism- Hanagan, M.T Mulagha, G.M. Kochak et al.
based inhibition. Biochemistry 29, 2199-2204. (1990). A phase I trial of CGS 16949A: A new
461. Brodie, A.M., H.M. Dowsett, and R.C. Coombes aromatase inhibitor. Cancer 65, 1279-1285.
(1988). Aromatase inhibitors as new endocrine ther- 476. Santen, R.J., L.M. Demers, H. Adlercreutz, H.
apy for breast cancer. Cancer Treat. Res. 39, 51-65. Harvey, S. Santner, S. Sanders et al. (1989).
462. Brodie, A.M.H., PK. Banks, S.E. Inkster, M. Inhibition of aromatase with CGS 16949 A in post-
Dowsett, and R.C. Coombes (1990). Aromatase menopausal women. J. Clin. Endocrinol. Metab.
inhibitors and hormone-dependent cancers. J. 68, 99-106.
Steroid Biochem. Mol. Biol. 37, 327-333. 477. Stein, R.C, M. Dowsett, J. Davenport, A. Hedley,
463. Johnston, J.O. (1998). Aromatase inhibitors. Crit. H.T. Ford, J.-C. Gazet et al. (1990). Preliminary
Rev. Biochem. Mol. Biol. 33, 375^05. study of the treatment of advanced breast cancer
464. Brodie, A., Q. Lu, and B. Long (1999). Aromatase in postmenopausal women with the aromatase
and its inhibitors. J. Steroid Biochem. Mol. Biol. inhibitor CGS 16949A. Cancer Res. 50,
69, 205-210. 1381-1384.
465. Seralini, G. and S. Moslemi (2001). Aromatase 478. Demers, L.M., J.C. Melby, T. E. Wilson, A. Lipton,
inhibitors: Past, present and future. Mol. Cell. H.A. Harvey, and R.J. Santen (1990). The effects of
Endocrinol. 178, 117-131. CGS 16949A, an aromatase inhibitor on adrenal
466. Recanatini, M., A. Cavalli, and P Valenti (2002). mineralocorticoidbiosynthesis../ Clin. Endocrinol.
Nonsteroidal aromatase inhibitors: Recent advances. Metab. 10, 1162-1166.
Med Res. Rev. 22, 282-304. 479. Tominaga, T, I. Adachi, Y. Sasaki, T. Tabei, T
467. Henderson, D., U.-F. Habenicht, Y. Nishino, and Ikeda, Y. Takatsuka et al. (2003). Double-blind ran-
M. E El Etreby (1987). Estrogens and benign pro- domised trial comparing the non-steroidal aro-
static hyperplasia: The basis for aromatase inhibitor matase inhibitors letrozole and fadrozole in
therapy. Steroids 50, 219-233. postmenopausal women with advanced breast can-
468. Schweikert, H.-U and U.W Tunn (1987). Effects of cer. Ann. Oncol. 14, 62-70.
the aromatase inhibitor testolactone on human 480. Goss, RE. and R.E. Smith (2002). Letrozole for
benign prostatic hyperplasia. Steroids 50, 191-199. the management of breast cancer. Expert Rev.
469. Phillips, G.B., WP CasteUi, R.D. Abbott, and PM. Anticancer Ther 2, 249-260.
McNamara (1983). Association of hyperestrogene- 481. Wouters, W, R. De Coster, R.W Tuman, C.R.
mia and coronary heart disease in men in the Bowden, J. Bruynseels, H. Vanderpas et al. (1989).
Framingham cohort. Am. J. Med. 74, 863-869. Aromatase inhibition by R 76713: Experimental
316 M.A. Correia and P.R. Ortiz de Monteilano

and clinical pharmacology. J. Steroid Biochem. 34, tight-binding aromatase inhibitor potentially use-
427-430. ful for the treatment of breast cancer. J. Steroid
482. Wouters, W., R. De Coster, J. Van Dun, M.D.W.G. Biochem. Mol. Biol. 41,113-111.
Krekels, A. Dillen, A. Raeymaekers et ah (1990). 494. Geelen, J.A.A., G.H. Deckers, J.TH. Van Der
Comparative effects of the aromatase inhibitor Wardt, H.J.J. Loozen, L.J.W Tax, and H.J.
R76713 and of its enantiomers R83839 and Kloosterboer (1991). Selection of 19-(ethyldithio)-
R83842 on steroid biosynthesis in vitro and in vivo. androst-4-ene-3,17-dione (ORG 30958): A potent
J. Steroid Biochem. Mol. Biol. 37, 1049-1054. aromatase inhibitor in vivo. J. Steroid Biochem.
483. Vanden Bossche, H., G. Willemsens, I. Roels, Mol. Biol. 3H, 181-188.
D. Bellens, H. Moereels, M.-C. Coene et al. (1990). 495. Lovett, J.A., M.V Darby, and R.E. Counsell
R 76713 and enantiomers: Selective, nonsteroidal (1984). Synthesis and evaluation of 19-aza- and
inhibitors of the cytochrome P450-dependent 19-aminoandrostenedione analogues as potential
oestrogen synthesis. Biochem. Pharmacol. 40, aromatase inhibitors. J. Med. Chem. 27, 734-740.
1707-1718. 496. Johnston, J.O., C.L. Wright, and B.W. Metcalf
484. Ahmed, S., S. Adat, A. Murrells, C.R Owen, and Y. (1984). Time-dependent inhibition of aromatase in
Amanuel (2002). Design, synthesis, and evaluation trophoblastic tumor cells in tissue culture. J.
of 4-(4')-aminobenzyl)-2-oxazolidinones as novel Steroid Biochem. 20, 1221-1226.
inhibitors of the cytochrome P-450 enzyme aro- 497. Shih, M.-J., M.H. Carrell, H.L. Carrell, C.L.
matase. Bioorg. Chem. 30, 315-331. Wright, J.O. Johnston, and C.H. Robinson (1987).
485. Marchand, P., M. Le Borgne, M. Palzer, G. Le Baut, Stereoselective inhibition of aromatase by novel
and R.W. Hartmann (2003). Preparation and phar- epoxysteroids. J. Chem. Soc, Chem. Commun.
macological profile of 7-(alpha-Azolylbenzyl)-1H- 213-214.
indoles and indolines as new aromatase inhibitors. 498. Childers, WE. and C.H. Robinson (1987). Novel
Bioorg. Med Chem. Lett. 13, 1553-1555. lOp-thiiranyl steroids as aromatase inhibitors. J.
486. Pouget, C , C. Fagnere, J.P Basly, G. Habrioux, Chem. Soc, Chem. Commun. 320-321.
and A.J. Chulia (2003). Design, synthesis and 499. Childers, W.E., J.V Silverton, J.T KeUis, L.E.
evaluation of 4-imidazolylflavans as new leads for Vickery and C.H. Robinson (1991). Inhibition of
aromatase inhibition. Bioorg. Med. Chem. Lett. 12, human placental aromatase by novel homologated
2859-2861. 19-oxiranyl and 19-thiiranyl steroids. J. Med.
487. Buzdar, A.U (2002). Anastrozole (Arimidex) in clin- Chem. 34, 1344-1349.
ical practice versus the old 'gold standard', tamox- 500. Kellis, J.T, WE. Childers, C.H. Robinson, and L.E.
ifen. Expert. Rev. Anticancer Ther 2, 623-629. Vickery (1987). Inhibition of aromatase cytochrome
488. Wellington, K. and D.M. Faulds (2002). P-450 by 10-oxirane and 10-thiirane substituted
Anastrozole: In early breast cancer. Drugs 62, androgens. Implications for the structure of the
2483-2490. active site. J. Biol. Chem. 262, 4421^426.
489. Miller, W.R., M. Stuart, T. Sahmoud, and J.M. 501. Njar, VC.O., E. Safi, J.V Silverton, and C.H.
Dixon (2002). Anastrozole ('Arimidex') blocks Robinson (1993). Novel lOp-aziridinyl steroids:
oestrogen synthesis both peripherally and within Inhibitors of aromatase. J. Chem. Soc. Perkin
the breast in postmenopausal women with large Trans. 710, 1161-1168.
operable breast cancer. Br J. Cancer 87, 502. Metcalf, B.W, C.L. Wright, J.P Burkhan, and J.O.
950-955. Johnston (1981). Substrate-induced inactivation of
490. Flynn, G.A., J.O. Johnston, C.L. Wright, and B.W aromatase by allenic and acetylenic steroids. J. Am.
Metcalf (1981). The time-dependent inactivation Chem. Soc. 103, 3221-3222.
of aromatase by 17-p-hydroxy-lO-methylthioestra- 503. Johnston, J.O. (1987). Biological characterization
l,4-dien-3-one. Biochem. Biophys. Res. Commun. of 10-(2-propynyl)estr-4-ene-3,17-dione (MDL
103,913-918. 18,962), an enzyme-activated inhibitor of aro-
491. Bednarski, PJ. and S.D. Nelson (1989). Interactions matase. Steroids 50, 105-120.
of thiol-containing androgens with human placental 504. Covey D.G., WF Hood, and VD Parikh (1981). lOp-
aromatase. J. Med. Chem. 32, 203-213. Propynyl-substituted steroids: Mechanism-based
492. Wright, J.N., G. Slatcher, and M. Akhtar (1991). enz3mie-activated irreversible inhibitors of estrogen
'Slow-binding' sixth-ligand inhibitors of cyto- biosynthesis, a 5/o/. Chem. 256, 1076-1079.
chrome P-450 aromatase. Studies with 19-thio- 505. Brandt, M.E., D. Puett, D.F Covey and S.J. Zimniski.
methyl- and 19-azido-androstenedione. Biochem. J. Characterization of pregnant mare's serum
273, 533-539. gonadotropin-stimulated rat ovarian aromatase and
493. Delaisi, C , B. Coucet, C. Hartmann, B. Trie, J.F its inhibition by 10-propargyl-estr-4-ene-3,17-dione.
Gourvest, and D. Lesuisse (1992). RU54115, a J. Steroid Biochem. 34, 317-324.
Inhibition of Cytochrome P450 Enzymes 317

506. Marcotte, P.A. and C.H. Robinson (1982). Synthesis 519. Marsh, D.A., E.J. Brodie, W. Garrett, C.-H. Tsai-
and evaluation of 10-beta-substituted Morris, and A.M. Brodie (1985). Aromatase
4-estrene-3,17-diones as inhibitors of human pla- inhibitors. Synthesis and biological activity of
cental microsomal aromatase. Steroids 39, 325-344. androstenedione derivatives. J. Med. Chem. 28,
507. Numazawa, M., A. Mutsumi, N. Asano, and Y. Ito 788-795.
(1993). A time-dependent inactivation of aro- 520. Brodie, A.M.H., H.J. Brodie, W.M. Garrett, J.R.
matase by 19-substituted androst-4-ene-3,6,17- Hendrickson, D.H. Marsh, and C.-H. Tsai-Morris
diones. Steroids 58, 4 0 ^ 6 . (1982). Effect of an aromatase inhibitor, 1,4,6-
508. Marcotte, P.A. and C.H. Robinson (1982). Design androstatriene-3,17-dione, on 7,12-dimethyl-[«]-
of mechanism-based inactivators of human placen- anthracene-induced mammary tumors in the rat
tal aromatase. Cancer Res. 42, 3322-3325. and its mechanism of action in vivo. Biochem.
509. Marcotte, PA. and C.H. Robinson (1982). Pharmacol. 31, 2017-2023.
Inhibition and inactivation of estrogen synthetase 521. Henderson, D., G. Norbisrath, and U. Kerb (1986).
(aromatase) by fluorinated substrate analogues. 1-Methyl-l,4-androstadiene-3,17-dione (SH 489):
Biochemistry 21, 2773-2778. Characterization of an irreversible inhibitor of
510. Numazawa, M., A. Mutsumi, K. Hoshi, M. Oshibe, estrogen biosynthesis. J. Steroid Biochem. 24,
E. Ishikawa, and H. Kigawa (1991). Synthesis and 303-306.
biochemical studies of 16- and 19-substituted 522. Numazawa, M., A. Mutsumi, K. Hoshi, and Y.
androst-4-enes as aromatase inhibitors. J. Med. Tanaka (1992). Androst-5-ene-7,17-dione: A novel
Chem. 34, 2496-2504. class of suicide substrate of aromatase. Biochem.
511. Mann, J. and B. Pietrzak (1987). Preparation of Biophys. Res. Commun. 186, 32-39.
aromatase inhibitors. Synthesis of 19,19-difluoro-4- 523. Covey, D.E and W.F Hood (1981). Enzyme-gener-
hydroxyandrost-4-ene-3,7-dione and related com- ated intermediates derived from 4-androstene-
pounds. J. Chem. Soc. Perkin Trans. 7385-388. 3,6,17-trione and l,4,6-androstatriene-3,17-dione
512. Furth, PS. and C.H. Robinson (1989). Tritium cause a time-dependent decrease in human placen-
release from [19-^H]19,19-difluoroandrost-4-ene- tal aromatase activity. Endocrinology 108,
3,17-dione during inactivation of aromatase. 1597-1599.
Biochemistry 2H, 1254-1259. 524. Numazawa, M., M. Tsuji, and A. Mutsumi (1987).
513. Covey, D.F. and W.F. Hood (1982). Aromatase Studies on aromatase inhibition with 4-androstene-
enzyme catalysis is involved in the potent inhibi- 3,6,17-trione: Its 3p-reduction and time-dependent
tion of estrogen biosynthesis caused by 4- irreversible binding to aromatase with human
acetoxy- and 4-hydroxy-4-androstene-3,17-dione. placental microsomes. J. Steroid Biochem. 28,
Mol. Pharmacol. 21, 173-180. 337-344.
514. Brodie, A.M.H., W.M. Garrett, J.R. Hendrickson, 525. Numazawa, M., K. Midzuhashi, and M. Nagaoka
C.-H. Tsai-Morris, PA. Marcotte, and C.H. (1994). Metabolic aspects of the l^-proton and the
Robinson (1981). Inactivation of aromatase in 19-methyl group of androst-4-ene-3,6,17-trione
vitro by 4-hydroxy-4-androstene-3,17-dione and 4- during aromatization by placental microsomes and
acetoxy-4-androstene-3,17-dione and sustained inactivation of aromatase. Biochem. Pharmacol.
effects in vivo. Steroids 38, 693-702. 47, 717-726.
515. Brodie, A.M.H. (1994). Aromatase inhibitors in 526. Di Salle, E., G. Ornati, D. Giudici, M. Lassus,
the treatment of breast cancer. J. Steroid. Biochem. T.R. Evans and R.C. Coombes (1992). Exemestane
Mol. Biol. 49, 2SI-2S1. (FCE 24304), a new steroidal aromatase inhibitor.
516. Di Salle, E., D. Giudici, G. Briatico, and G. Ornati J. Steroid Biochem. Mol. Biol. 43, 137-143.
(1990). Novel irreversible aromatase inhibitors. 527. Geisler, J., N. King, G. Anker, G. Ornati, E. Di Salle,
Ann. N. Y.Acad Sci. 595, 357-367. PE. Lonning et al. (1998). In vivo inhibition of
517. Di Salle, E., D. Giudici, G. Ornati, G. Briatico, R. aromatization by exemestane, a novel irreversible
D'Alessio, V Villa et al. (1990). 4-Amino- aromatase inhibitor, in postmenopausal breast can-
androstenedione derivatives: A novel class of irre- cer patients. Clin. Cancer Res. 4, 2089-2093.
versible aromatase inhibitors. Comparison with 528. Clemett, D. and H.M. Lamb (2000). Exemestane:
FCE 24304 and 4-hydroxyandrostenedione. J. A review of its use in postmenopausal women with
Steroid Biochem. Mol. Biol. 37, 369-374. advanced breast cancer. Drugs 59, 1279-1296.
518. Di Salle, E., G. Briatico, D. Giudici, G. Ornati, and 529. Brueggemeier, R.W (2002). Overview of the phar-
T. Zaccheo (1989). Aromatase inhibition and macology of the aromatase inactivator exemestane.
experimental antitumor activity of FCE 24304, Breast Cancer Res. Treat. 74, 177-185.
MDL 18962 and SH 489. J. Steroid Biochem. 34, 530. Dixon, J.M. (2002). Exemestane: A potent irre-
431-434. versible aromatase inactivator and a promising
318 M.A. Correia and P.R. Ortiz de Montellano

advance in breast cancer treatment. Expert Rev. and lanosterol on artificial membranes. Proc. Natl.
Anticancer Then 2, 267-275. Acad. Sci. USA 74, 4924-4926.
531. Higa, G.M. (2002). Exemestane: Treatment of 545. Freter, C.E., R.C. Laderson, and D.E Sibert (1979).
breast cancer with selective inactivation of aro- Membrane phospholipid alterations in response to
matase. Am. J. Health Syst. Pharm. 59, 2194-2201. sterol depletion of LM cells. J. Biol. Chem. 254,
532. Longcope, C , A. Femino, and J.O. Johnston 6909-6916.
(1988). Inhibition of peripheral aromatization in 546. Vanden Bossche, H., W Lauwers, G. Willemsens,
baboons by an enzyme-activated aromatase P Marichal, E Cornelissen and W Cools (1984).
inhibitor (MDL 18,962). Endocrinology 111, Molecular basis for the antimycotic and antibacter-
2007-2011. ial activity of N-substituted imidazoles and tria-
533. Johnston, J.O. (1990). Studies with the steroidal zoles: The inhibition of isoprenoid biosynthesis.
aromatase inhibitor, 19-acetylenic androstene- Pestic.Sci. 15, 188-198.
dione (MDL 18,962). J. Cancer Res. Clin. Oncol. 547. Heeres, J., M. De Brabander, and H. Vanden
116, 880. Bossche (1982). Ketoconazole: Chemistry and basis
534. Covey, D.E., W.E Hood, D.D. Bensen, and H.L. for selectivity. In P. Periti and G.G. Grossi (eds.).
Carrell (1984). Hydroperoxides as inactivators of Current Chemotherapy and Immunotherapy, Vol. 2.
aromatase: 10-Beta-hydroperoxy-4-estrene-3,17- American Society of Microbiology, Washington,
dione, crystal structure and inactivation character- D C , pp. 1007-1009.
istics. Biochemistry 23, 5398-5406. 548. Willemsens, G., W Cools, and H. Vanden Bossche
535. Covey, D.E, W.E Hood, and PC. McMullan (1980). Effects of miconazole and ketoconazole on
(1986). Studies of the inactivation of human pla- sterol synthesis in a subcellular fraction of yeast
cental aromatase by 17a-ethynyl-substituted 10(3- and mammalian cells. In H. Van den Bossche (ed.).
hydroperoxy and related 19-nor steroids. Biochem. The Host-Invader Interplay Elsevier/North Holland,
Pharmacol. 35, 1671-1674. Amsterdam, pp. 691-694.
536. Bednarski, P.J., D.J. Porubek, and S.D. Nelson 549. Murray, M., A.J. Ryan, and PJ. Little (1982).
(1985). Thiol-containing androgens as suicide sub- Inhibition of rat hepatic microsomal aminopyrine
strates of aromatase. J. Med. Chem. 28, 775-779. N-demethylase activity by benzimidazole deriva-
537. Wright, J.N., van PT. Leersum, S.G. Chamberlin tives: Quantitative structure-activity relationships.
and M. Akhtar (1989). Inhibition of aromatase by J. Med Chem. 25, 887-892.
steroids substituted at C-19 with halogen, sulphur, 550. Santen, R.J., H. Vanden Bossche, J. Symoens,
and nitrogen. J. Chem. Soc. Perkin Trans. I J. Brugmans, and R. DeCoster (1983). Site of
1647-1655. action of low dose ketoconazole or androgen
538. Burkhart, J.P, N.P Peet, C.L. Wright, and J.O. biosynthesis in men. J. Clin. Endocrinol. Metab.
Johnston (1991). Novel time-dependent inhibitors 57, 732-736.
of human placental aromatase. J. Med. Chem. 34, 551. Albengres, E., H. Le Louet, and J.P Tillement
1748-1750. (1998). Systemic antifungal agents. Drug interac-
539. Numazawa, M., A. Yoshimura, M. Tachibana, M. tions of clinical significance. Drug Saf. 18, 83-97.
Shelangouski, and M. Ishikawa (2002). Time- 552. Gahder, P, E.I. Mercer, B.C. Baldwin, and TE.
dependent aromatase inactivation by 4 beta, 5 beta- Wiggins (1983). A comparison of the potency of
epoxides of the natural substrate androstenedione some fungicides as inhibitors of sterol 14-
and its 19-oxygenated analogs. Steroids 67, demethylation. Pest. Biochem. Physiol.
185-193. 19, 1-10.
540. Vanden Bossche, H., G. Willemsens, W. Cools, 553. Ito, T, Y. Aoyama, K. Ishida, M. Kudoh, K. Hori,
P. Marichal and W. Lauwers (1983). Hypothesis S. Tsuchiya etal. (1994). Selectivity of isoprenoid-
on the molecular basis of the antifungal activity of containing imidazole antifungal compounds for
N-substituted imidazoles and triazoles. Biochem. sterol 14-demethylase P450 (P450(14)DM) and 7-
Soc. Trans. 11,665-667. ethoxycoumarin 0-deethylase P450 of rat liver
541. Mercer, E.I. (1991). Sterol biosynthesis inhibitors: microsomes. Biochem. Pharmacol. 48, 1577-1582.
Their current status and modes of action. Lipids 554. Dolle, R.E., H.S. Allaudeen, and L.I. Kruse
26, 584-597. (1990). Design and synthesis of 14a-methyl-15-
542. Berg, M. and M. Plempel (eds.) (1988). Sterol aza-D-homosterols as novel antimycotics. J. Med.
Biosynthesis Inhibitors. Horwood, Ellis. Chem. 33, 877-880.
543. Nes, WR. (1974). Role of sterols in membranes. 555. Frye, L.L., K.P Cusack, D.A. Leonard, and J.A.
Lipids 9, 596-6X2. Anderson (1994). Oxolanosterol oximes: Dual-
544. Yeagle, PL., R.B. Martin, A.K. Lala, H.K. Lin, and action inhibitors of cholesterol biosynthesis. J.
K. Block (1977). Differential effects of cholesterol Lipid Res. 35, 1333-1344.
inhibition of Cytochrome P450 Enzymes 319

556. Aoyama, Y., Y. Yoshida, Y Sonoda, and Y Sato modeling of human CYP17 inhibitors. Potential
(1987). 7-Oxo-24,25-dihydrolanosterol: A novel agents for prostate cancer therapy. J. Med. Chem.
lanosterol 14a-demethylase {P-450 14DM) inhibitor 46,2345-2351.
which blocks electron transfer to the oxyferro inter- 568. Angelastro, M.R., M.E. Laughlin, G.L. Schatzman,
mediate. Biochim. Biophys. Acta. 922, 270-277. P Bey, and TR. Blohm (1989). 17p-
557. Trzaskos, J.M., R.T. Fischer, S.S. Ko, R.L. (Cyclopropylamino)-androst-5-en-3p-ol, a selec-
Magolda, S. Stam, P. Johnson et al. (1995). tive mechanism-based inhibitor of cytochrome
Substrate-based inhibitors of lanosterol 14 alpha- P45017a(steroid 17a-hydroxylase/C 17-20 lyase).
methyl demethylase: II. Time-dependent enzyme Biochem. Biophys. Res. Commun. 162, 1571-1577.
inactivation by selected oxylanosterol analogs. 569. Njar, V C , M. Hector, and R.W Hartmann (1996).
Biochemistry 34, 9677-9681 20-amino and 20,21-aziridinyl pregnene steroids:
558. Trzaskos, J.M., R.L. Magolda, M.F. Favata, R.T. Development of potent inhibitors of 17 alpha-
Fischer, RR. Johnson, H.W. Chen et al. (1993). hydroxylase/C17,20-lyase (P450 17). Bioorg. Med.
Modulation of 3-hydroxy-3-methylglutaryl- Chem. 4, 1447-1453.
CoA reductase by 15a-fluorolanost-7-en-3p-ol. A 570. Ling, Y Z., I S . Li, Y Liu, K. Kato, G.T Klus, and
mechanism-based inhibitor of cholesterol biosyn- A. Brodie (1997). 17-Imidazolyl, pyrazolyl, and
thesis. J. Biol. Chem. 268, 22591-22599. isoxazolyl androstene derivatives. Novel steroidal
559. Cooper, A.B., J.J. Wright, A.K. Ganguly, J. Desai, inhibitors of human cytochrome C17,20-lyase
D. Loenberg, R. Parmegiani et al. (1989). (P450(17) alpha). J. Med Chem. 40, 3297-3304.
Synthesis of 14-a-aminomethyl substituted lanos- 571. Jarman, M., S.E. Barrie, and J.M. Llera (1998).
terol derivatives; inhibitors of fungal ergosterol The 16,17-double bond is needed for irreversible
biosynthesis. J. Chem. Soc, Chem. Commun. inhibition of human cytochrome P45017alpha by
898-900. abiraterone (17-(3 -pyridyl)androsta-5, 16-dien-
560. Frye, L.L., K.P Cusack, and D.A. Leonard (1993). 3beta-ol) and related steroidal inhibitors. J. Med.
32-Methyl-32-oxylanosterols: Dual-action inhibitors C/zem. 41, 5375-5381.
of cholesterol biosynthesis. J. Med. Chem. 36, 572. Njar, V C , K. Kato, LP Nnane, D.N. Grigoryev, B.J.
410-416. Long, and A.M. Brodie (1998). Novel 17-azolyl
561. Frye, L.L. and C.H. Robinson (1988). Novel steroids, potent inhibitors of human cytochrome 17
inhibitors of lanosterol 14a-methyl demethylase, a alpha-hydroxylase-C17,20-lyase (P450(17) alpha):
critical enzyme in cholesterol biosynthesis. J. Potential agents for the treatment of prostate cancer.
Chem. Soc. Chem. Commun. 129-131. J.Med Chem. 41, 902-9X2.
562. Mayer, R.J., J.L. Adams, M.J. Bossard, and T.A. 573. Nnane, LP, VC. Njar, Y Liu, Q. Lu, and A.M. Brodie
Berkhout (1991). Effects of a novel lanosterol 14a- (1999). Effects of novel 17-azolyl compounds on
demethylase inhibitor on the regulation of androgen synthesis in vitro and in vivo. J. Steroid
3-hydroxy-3-methylglutaryl-coenzyme A reductase Biochem. Mol. Biol. 71, 145-152.
in Hep G2 cells. J. Biol. Chem. 266, 20070-20078. 574. Njar, VC. and A.M. Brodie (1999). Inhibitors of
563. Frye, L.L. and C.H. Robinson (1990). Synthesis of 17alpha-hydroxylase/17,20-lyase (CYP17): Potential
potential mechanism-based inactivators of lanosterol agents for the treatment of prostate cancer. Curr.
14a-demethylase. J Org. Chem. 55, 1579-1584. Pharm.Des.5, 163-180.
564. Tuck, S.E, C.H. Robinson, and J.V Silverton 575. Zhuang, Y, B.G. Wachall, and R.W. Hartmann
(1991). Assessment of the active-site requirements (2000). Novel imidazolyl and triazolyl substituted
of lanosterol 14a-demethylase: Evaluation of novel biphenyl compounds: Synthesis and evaluation as
substrate analogues as competitive inhibitors. J. nonsteroidal inhibitors of human 17alpha-hydroxy-
Org. Chem. 56, 1260-1266. lase-C17, 20-lyase (P450 17). Bioorg. Med. Chem.
565. Bossard, M.J., T.A. Tomaszek, T. Gallagher, B.W. 8, 1245-1252.
Metcalf, and J.L. Adams (1991). Steroidal 576. Long, B.J., D.N. Grigoryev, LP Nnane, Y Liu,
acetylenes: Mechanism-based inactivators of lanos- YZ. Ling, and A.M. Brodie (2000). Antiandrogenic
terol 14a-demethylase. 5/oorg. Chem. 19,418^32. effects of novel androgen synthesis inhibitors on
566. Swinney, D.C., O.Y So, D.M. Watson, RW. Berry, hormone-dependent prostate cancer. Cancer Res.
A.S. Webb, D.J. Kertesz et al (1994). Selective 60, 6630-6640.
inhibition of mammalian lanosterol 14 alpha- 577. Haidar, S., PB. Ehmer, and R.W Hartmann (2001).
demethylase by RS-21607 in vitro and in vivo. Novel steroidal pyrimidyl inhibitors of P450 17 (17
Biochemistry 33, 4702^713. alpha-hydroxylase/C17-20-lyase). Arch. Pharm.
567. Clement, O.O., CM. Freeman, R.W Hartmann, (Weinheim) 334, 373-374.
YD. Handratta, T.S. Vasaitis, A.M.H. Brodie et al. 578. Burkhart, J.P, PM. Weintraub, CA. Gates, R.L
(2003). Three dimensional pharmacophore Resvick, R.J. Vaz, D. Friedrich et al. (2002).
320 M.A. Correia and P.R. Ortiz de Montellano

Novel steroidal vinyl fluorides as inhibitors of 590. McGiff, J.C, C.P Quilley, and M.A. Carroll
steroid CI7(20) lyase. Bioorg. Med. Chem. 10, (1993). The contribution of cytochrome P450-
929-934. dependent arachidonate metabolites to integrated
579. Clement, O.O., CM. Freeman, R.W. Hartmann, VD. renal function. Steroids 58, 573-579.
Handratta, T.S. Vasaitis, A.M. Brodie et al (2003). 591. Harder, D.R., WB. Campbell, and R.J. Roman
Three dimensional pharmacophore modeling of (1995). Role of cytochrome P-450 enzymes and
human CYP17 inhibitors. Potential agents for metabolites of arachidonic acid in the control of
prostate cancer therapy. J. Med. Chem. 46, vascular tone. J. Vase. Res. 32, 79-92.
2345-2351. 592. Roman, R.J. (2002). P-450 metabolites of arachi-
580. Haidar, S., P.B. Ehmer, S. Barassin, C. Batzl- donic acid in the control of cardiovascular func-
Hartmann, and R.W. Hartmann (2003). Effects of tion. Physiol Rev. 82, 131-185.
novel 17alpha-hydroxylase/C17,20-lyase (P450 593. Hoagland, K.M., K.G. Maier, C. Moreno, M. Yu,
17, CYP 17) inhibitors on androgen biosynthesis and R.J. Roman (2001). Cytochrome P450 metabo-
in vitro and in vivo. J. Steroid Biochem. Mol. Biol. lites of arachidonic acid: Novel regulators of renal
84, 555-562. function. Nephrol. Dial. Transplant. 16, 2283-2285.
581. Vinh, T.K., S.W. Yee, A.J. Kirby, PJ. Nicholls, and 594. Maier, K.G. and R.J. Roman (2001). Cytochrome
C. Simons (2001). l-[(Benzofuran-2-yl)phenyl- P450 metabolites of arachidonic acid in the control
methyljtriazoles as steroidogenic inhibitors: Syn- of renal function. Curr Opin. Nephrol. Hypertens.
thesis and in vitro inhibition of human placental 10, 81-87.
CYP 19 aromatase. Anticancer Drug Des. 16, 595. Roman, R.J., K.G. Maier, C.W Sun, D.R. Harder,
217-225. and M. Alonso-Galicia (2000). Renal and cardio-
582. Berg, A.M., A.B. Kickman, E. Miao, A. Cochran, vascular actions of 20-hydroxyeicosatetraenoic
S.R. Wilson, and WH. Orme-Johnson (1990). acid and epoxyeicosatrienoic acids. Clin. Exp.
Effects of inhibitors of cytochrome P-45017a on Pharmacol. Physiol. 11, 855-865.
steroid production in mouse Leydig cells and 596. Roman, R.J., M. Alonso-Galicia, and T.W. Wilson
mouse and pig testes microsomes. Biochemistry (1997). Renal P450 metabolites of arachidonic
29,2193-2201. acid and the development of hypertension in Dahl
583. Viger, A., S. Coustal, S. Perard, B. Chappe, and A. salt-sensitive rats. Am. J. Hypertens. 10, 63S-67S.
Marquet (1988). Synthesis and activity of new 597. Zou, A.P, YH. Ma, Z.H. Sui, PR. Ortiz de
inhibitors of aldosterone biosynthesis. J. Steroid Montellano, J.E. Clark, B.S. Masters et al. (1994).
Biochem. 30, 469-472. Effects of 17-octadecynoic acid, a suicide-
584. Viger, A., S. Coustal, S. Perard, A. Piflfeteau, and substrate inhibitor of cytochrome P450 fatty acid
A. Marquet (1989). 18-Substituted progesterone co-hydroxylase, on renal function in rats. J.
derivatives as inhibitors of aldosterone biosynthe- Pharmacol. Exp. Ther 268, 474^81.
sis. J. Steroid Biochem. 33, 119-124. 598. Imig, J.D., A.P Zou, PR. Ortiz de Montellano,
585. Gomez-Sanchez, C.E., S. Chiou, and N. Yamakita Z. Sui, and R.J. Roman (1994). Cytochrome P-450
(1993). 18-Ethynyl-deoxycorticosterone inhibition inhibitors alter afferent arteriolar responses to eleva-
of steroid production is different in freshly isolated tions in pressure. Am. J. Physiol. 266, HI 879-H1885.
compared to cultured calf zona glomerulosa cells. 599. Alkayed, N.J., E.K. Birks, A.G. Hudetz,
J. Steroid Biochem. Mol. Biol 46, 805-810. R.J. Roman, L. Henderson, and D.R. Harder
586. Johnston, J.O., C.L. Wright, R.A. Bohnke, and PR. (1996). Inhibition of brain P-450 arachidonic acid
Kastner (1991). Inhibition of aldosterone biosyn- epoxygenase decreases baseline cerebral blood
thesis in primates by 18-acetylenic deoxycorticos- flow. Am. J. Physiol. Ill, H1541-H1546.
terone. Endocrinology 128 (Suppl. Abstract 24). 600. Messer-Letienne, I., N. Bernard, R.J. Roman,
587. Kupfer, D. (1982). Endogenous substrates of J. Sassard, and D. Benzoni (1999). Cytochrome
monooxygenases: Fatty acids and prostaglandins. P-450 arachidonate metabolite inhibition improves
In J.B. Schenkman and D. Kupfer (eds). Hepatic renal function in Lyon hypertensive rats. Am. J.
Cytochrome P450 Monooxygenase System. Hypertens. 12, 398-404.
Pergamon Press, Elmsford, NY, pp. 157-190. 601. Wang, M.H., E. Brand-Schieber, B.A. Zand,
588. Kupfer, D. (1980). Endogenous substrates of X. Nguyen, J.R. Falck, N. Balu et al. (1998).
monooxygenases: Fatty acids and prostaglandins. Cytochrome P450-derived arachidonic acid metab-
Pharmacol. Ther 11, 469-496. olism in the rat kidney: Characterization of
589. Hirt, D.L. and H.R. Jacobson (1991). Functional selective inhibitors. J. Pharmacol. Exp. Ther 284,
effects of cytochrome P450 arachidonate 966-973.
metabolites in the kidney. Semin. Nephrol. 11, 602. Alonso-Galicia, M., K.G. Maier, A.S. Greene,
148-155. A.W. Cowley, Jr., and R.J. Roman (2002). Role of
Inhibition of Cytochrome P450 Enzymes 321

20-hydroxyeicosatetraenoic acid in the renal and 614. Zou, A.P, J.T. Fleming, J.R. Falck, E.R. Jacobs,
vasoconstrictor actions of angiotensin II. Am. J. D. Gebremedhin, D.R. Harder et al. (1996). 20-
Physiol. Regul. Integr. Comp. Physiol. 283, HETE is an endogenous inhibitor of the large-
R60-R68. conductance Ca(2+)-activated K+ channel in
603. Alonso-Galicia, M., C.W. Sun, J.R. Falck, renal arterioles. Am. J. Physiol 270, R228-R237.
D.R. Harder, and R.J. Roman (1998). Contribution 615. Stec, D.E., D.L. Mattson, and R.J. Roman (1997).
of 20-HETE to the vasodilator actions of nitric Inhibition of renal outer medullary 20-HETE pro-
oxide in renal arteries. J. Physiol. 275, F370-F378. duction produces hypertension in Lewis rats.
604. Quigley, R., M. Baum, K.M. Reddy, J.C. Griener, Hypertension. 29, 315-319.
and J.R. Falck (2000). Effects of 20-HETE and 616. Evans, R.G., K.H. Day, R.J. Roman, K.H. Hopp,
19(S)-HETE on rabbit proximal straight tubule and W.P Anderson (1998). Effects of intrarenal
volume transport. Am. J. Physiol. Renal. Physiol. infusion of 17-octadecynoic acid on renal antihy-
21S, F949-953. pertensive m echanisms in anesthetized rabbits.
605. Frisbee, J.C, R.J. Roman, U.M. Krishna, Am. J. Hypertens. 11, 803-812.
J.R. Falck, and J.H. Lombard (2001). Relative con- 617. Sun, C.W., M. Alonso-Galicia, M.R. Taheri,
tributions of cyclooxygenase- and cytochrome J.R. Falck, D.R. Harder, and R.J. Roman (1998).
P450 omega-hydroxylase-dependent pathways to Nitric oxide-20-hydroxyeicosatetraenoic acid
hypoxic dilation of skeletal muscle resistance arter- interaction in the regulation of K+ channel activ-
ies. J. Vase. Res. 38, 305-314. ity and vascular tone in renal arterioles. Circ. Res.
606. Frisbee, J.C, R.J. Roman, J.R. Falck, 83, 1069-1079.
U.M. Krishna, and J.H. Lombard (2001). 20-HETE 618. Messer-Letienne, I., N. Bernard, R.J. Roman,
contributes to myogenic activation of skeletal mus- J. Sassard, and D. Benzoni (1999). 20-
cle resistance arteries in Brown Norway and Hydroxyeicosatetraenoic acid and renal function in
Sprague-Dawley rats. Microcireulation 8, 45-55. Lyon hypertensive rats. Eur J. Pharmacol 378,
607. Kunert, M.R, R.J. Roman, J.R. Falck, and J.H. 291-297.
Lombard (2001). Differential effect of cytochrome P- 619. Shak, S. I. and Goldstein (1984). Omega-oxidation
450 omega-hydroxylase inhibition on 02-induced is the major pathway for the catabolism of
constriction of arterioles in SHR with early and estab- leukotriene B4 in human polymorphonuclear
lished hypertension. Microcireulation 8, 435-443. leukocytes. J: 5/o/. Chem. 259, 10181-10187.
608. Miyata, N., K. Taniguchi, T. Seki, T. Ishimoto, 620. Kikuta, Y, E. Kusunose, K. Endo, S. Yamamoto,
M. Sato-Watanabe, Y. Yasuda et al (2001). K. Sogawa, Y Fujii-Kuriyama et al (1993). A
HET0016, a potent and selective inhibitor of 20- novel form of cytochrome P-450 family 4 in
HETE synthesizing enzyme. Br J. Pharmacol. human polymorphonuclear leukocytes. cDNA
133, 325-329. cloning and expression of leukotriene B4 co-
609. Ortiz de Montellano, RR. and N.O. Reich (1984). hydroxylase. J. Biol Chem. 268, 9376-9380.
Specific inactivation of hepatic fatty acid hydrox- 621. Clancy, R.M., CA. Dahinden, and T.E. Hugh
ylases by acetylenic fatty acids. J. Biol. Chem. 259, (1984). Oxidation of leukotrienes at the co-end:
4136^141. Demonstration of a receptor for the 20-hydroxy
610. CaJacob, CA. and RR. Ortiz de Montellano (1986). derivative of leukotriene B4 on human neutrophils
Mechanism-based in vivo inactivation of lauric acid and implications for the analysis of leukotriene
hydroxylases. Biochemistry 25, 4705^711. receptors. Proc. Natl Acad. Sci. USA 81,
611. Xu, R, W.O. Straub, W. Pak, R Su, K.G. Maier, M. 5729-5733.
Yu et al. (2002). Antihypertensive effect of mech- 622. Shak, S., N.O. Reich, I.M. Goldstein, and PR.
anism-based inhibition of renal arachidonic acid Ortiz de Montellano (1985). Leukotriene B4
omega-hydroxylase activity. Am. J. Physiol. Regul. co-hydroxylase in human polymorphonuclear
Integr Comp. Physiol 283, R710-720. leukocytes: Suicidal inactivation by acetylenic
612. Zou, A.P, J.D. Imig, M. Kaldunski, PR. Ortiz de fatty acids. J. Biol Chem. 260, 13023-13028.
Montellano, Z. Sui, and R.J Roman (1994). 623. Williams, D.E., A.S. Muerhoff, N.O. Reich,
Inhibition of renal vascular 20-HETE production CA. CaJacob, PR. Ortiz de Montellano, and
impairs autoregulation of renal blood flow. Am. J. B.S.S. Masters (1989). Prostaglandin and fatty
Physiol 266, F275-282. acid o) and (w-l) oxidation in rabbit lung.
613. Zou, A.P, J.D. Imig, PR. Ortiz de Montellano, Acetylenic fatty acid mechanism based inactiva-
Z. Sui, J.R. Falck, and R.J. Roman (1994). Effect of tors as specific inhibitors. J. Biol Chem. 264,
P-450 omega-hydroxylase metabolites of arachi- 749-756.
donic acid on tubuloglomerular feedback. Am. J. 624. Amaral, S.L., K.G. Maier, D.N. Schippers,
Physiol 266,-^934-^941. R.J. Roman, and A.S. Greene (2003). CYP4A
322 M.A. Correia and P.R. Ortiz de Montellano

metabolites of arachidonic acid and VEGF are 628. Fulco, A.J. (1991). P450gj^_3 and other inducible
mediators of skeletal muscle angiogenesis. Am. J. bacterial P450 cytochromes: Biochemistry and reg-
Physiol. Heart Circ. Physiol. 284, H1528-H1535. ulation. y4««. Rev. Pharmacol. Toxicol. 31, 177-203.
625. Wang, M.H., B.A. Zand, A. Nasjletti, and 629. Salaun, J.R, D. Reichhart, A. Simon, F Durst,
M. Laniado-Schwartzman (2002). Renal 20- N.O. Reich, and RR. Ortiz de Montellano (1984).
hydroxyeicosatetraenoic acid synthesis during Autocatalytic inactivation of plant cytochrome
pregnancy. Am. J. Physiol. Regul. Integr. Comp. P-450 enzymes: Selective inactivation of the lauric
Physiol. 282, R383-R389. acid in-chain hydroxylase from Helianthus
626. Nguyen, X., M.H. Wang, K.M. Reddy, J.R. Falck, tuberosus L. by unsaturated substrate analogs.
and M.L. Schwartzman (1999). Kinetic profile of Arch. Biochem. Biophys. 232, 1-7.
the rat CYP4A isoforms: Arachidonic acid metab- 630. Shirane, N., Z. Sui, J.A. Peterson, and PR. Ortiz
olism and isoform-specific inhibitors. Am. J. de Montellano (1993). Cytochrome P450QJ^_3
Physiol. 276, R1691-R1700. (CYP102): Regiospecificity of oxidation of
627. Brand-Schieber, E., J.F. Falck, and (o-unsaturated fatty acids and mechanism-based
M. Schwartzman (2000). Selective inhibition of inactivation. Biochemistry 32, 13732-13741.
arachidonic acid epoxidation in vivo. J. Physiol.
Pharmacol. 51, 655-672.
8
Induction of Cytochrome P450 Enzymes
Susanne N. Williams, Elizabeth Dunham, and
Christopher A. Bradfield

1. Introduction 1.2. Overview of Nuclear


Receptors
1.1. Cytochrome P450 Enzymes
and the Adaptive Response The adaptive response to xenobiotics is orches-
trated in the cell by a subset of receptors that act
Organisms are constantly exposed to an ever- primarily in the nucleus. For this chapter, we will
changing spectrum of foreign chemicals or xeno- employ a liberal definition of nuclear receptor
biotics. In response to this challenge, adaptive (NR) that includes all signaling molecules that
mechanisms have evolved in higher eukaryotes ftinction as ligand-binding transcription factors
that allow them to detect an insulting agent and that bind to specific DNA enhancer sequences and
accordingly increase its metabolism and clear- upregulate the transcription of CYP genes. Two
ance. Commonly, the front line in this metabolic classes of NRs will be discussed in this chapter.
defense is the cytochrome P450 monooxygenases Members of the nuclear hormone receptor (NHR)
(CYPs). These enzymes catalyze the first step in superfamily to be reviewed include the constitu-
the metabolism of lipophilic xenobiotics to more tive androstane receptor (CAR), the pregnane
water-soluble compounds that can be readily X-receptor (PXR), and the peroxisome prolifera-
excreted. A common feature of the CYPs is that tor activated receptors (PPARs). A single member
exposure to a xenobiotic substrate often results in of the PAS superfamily, the aryl hydrocarbon
increased expression of the CYP enzyme(s) capa- receptor (AHR), will also be discussed.
ble of its metabolism. This adaptive response, The NHRs have a modular structure character-
known as induction, is a tightly regulated process ized by an N-terminal ligand-independent AF-1
that is controlled primarily at the level of tran- transactivation domain (TAD), a highly conserved
scription. Regulating the expression of CYPs in a DNA binding domain (DBD) containing two zinc
manner that is sensitive to xenobiotic exposure finger motifs, and a ligand binding domain (LBD)
allows the cell to increase the levels of the neces- that contains a ligand-dependent AF-2 TAD in its
sary CYP enzymes only as needed to facilitate C-terminal portion^ The CAR and the PXR play
elimination of a toxicant. major roles in the induction of the CYP2 and CYP3

Susanne N. Williams, Elizabeth Dunham, and Christopher A. Bradfield McArdle Laboratory for
Cancer Research, University of Wisconsin, Madison, WI.
Cytochrome P450: Structure, Mechanism, and Biochemistry, 3e, edited by Paul R. Ortiz de Montellano
Kluwer Academic / Plenum Publishers, New York, 2005.

323
324 Susanne N. Williams et al.

genes, while PPARa mediates the upregulation of Our goal for this chapter is to provide an
the CYP4 family^. The PXR, CAR, and PPARa overview of the discoveries that have occurred
are similar in that they fonn heterodimers with over the last 10 years in the area of NR-mediated
the retinoid X receptor (RXR) to bind response ele- induction of CYP enzymes by xenobiotics. Our
ments that contain two copies of the core sequence current understanding of the mechanism of signal
AG(G/T)TCA arranged as everted repeats (ER) or transduction of each receptor will be presented,
direct repeats (DR) separated by varying numbers and we will highlight some areas where fiirther
of nucleotide spacers^. These response elements are investigation is needed. Along the way, we will
commonly designated by the type of repeat, fol- also touch on emerging physiological roles of
lowed by the nucleotide spacer number; for exam- some NRs. While we hope to provide a basic
ple, a direct repeat separated by four nucleotides working knowledge of NR-mediated signal trans-
is termed "DR4." The binding specificity and the duction, the breadth of the topic prevents us from
transactivation potential of different NHR/RXR discussing in depth many of the more detailed
heterodimers can be determined by the spacer aspects of NR signaling pathways. For those desir-
number, the nucleotide sequence in the half-sites, ing more information on specific topics, readers
and often by sequences 5' of the half-sites'. are referred to relevant reviews.
The nuclear receptor known as the aryl hydro-
carbon receptor (AHR) is a member of the PAS
superfamily of proteins and regulates the CYPl 2. The Pregnane X Receptor
genes^' ^. The PAS domain was named for the
first three proteins in which it was recognized: 2.1. Introduction
PER, ^RNT, and SIM^. Most members of the
PAS superfamily, including the AHR, have an The CYP3A enzymes are the most abundant
N-terminal basic helix-loop-helix (bHLH) domain cytochrome P450s in human liver and are respon-
adjacent to the PAS domain and a carboxy termi- sible for the metabolism of endogenous steroids
nal domain that influences transcription^. The and numerous xenobiotics^^. The main isoform in
AHR binds to specific response elements upstream humans, CYP3A4, is estimated to be responsible
of the CYPl genes containing the core sequence for the metabolism of more than 50% of the cur-
TNGCGTG to induce gene expression. rently used drugs and is considered central in
A common characteristic of NRs is that they many clinically important drug interactions ^^ Due
interact with coactivators and corepressors. In to the importance of the CYP3A enzymes, the
general, in the absence of ligand or when bound mechanisms of CYP3A induction are of special
by antagonist, NRs exist in a complex with core- interest and have been an area of intense research.
pressors, such as SMRT or NcoR, which inhibit A series of discoveries over many years
transcriptional activity through recruitment of have led to our current understanding of CYP3A
histone deacetylases or other mechanisms^' ^. induction. Early studies demonstrated that the
Activation of the NR by agonist binding or phos- administration of certain steroids to rats caused
phorylation causes a conformational change in the greatly enhanced transcription of CYP3A genes
receptor that results in the dissociation of core- in the liver and small intestine. Induction was
pressors and the recruitment of coactivators, seen after treatment with the potent glucocorticoid
such as SRC-1 and CBP, which interact with NRs dexamethasone and, paradoxically, also with the
through conserved LXXLL motifs^"^. Coactiva- synthetic antiglucocorticoid pregnenolone 16a-
tors can increase the rate of gene transcription carbonitrile (PCN)*2-'4 ^he response of CYP3A
through chromatin remodeling or by interacting to glucocorticoids was distinct from a classical
with components of the basic transcriptional glucocorticoid receptor (GR)-dependent response
machinery to increase the number of ftinctional with respect to both the time course of induction
basal promoter complexes^' ^. The final composi- and the dose of dexamethasone required, as well
tion and activity of the recruited multiprotein as the rank order of the potency of various
transcriptional complex is dependent on both steroids^^' ^^. Analysis of the promoter region of
promoter and enhancer sequences, as well as on the CYP3A23 gene revealed conserved enhancer
the specific ligand bound to the NR. elements similar to those recognized by NRs;
Induction of Cytochrome P450 Enzymes 325

however, further experimentation showed that the the human PXR (hPXR) LBD, both with and with-
elements were not bound by the GR^^' ^^. It out agonist, have been useful in understanding the
was postulated that the induction of CYP3A23 receptor's ability to accommodate ligands of vari-
involved a novel NR acting by a mechanism ous structures and sizes^"^. The crystal structure of
distinct from that of the classical GR pathway the hPXR LBD in the absence of ligand revealed
a hydrophobic ligand binding pocket that is larger
than that of most NHRs; furthermore, a unique
2.2. The PXR flexible loop found adjacent to the ligand-binding
cavity likely contributes to the ability of the PXR
The long-standing paradox of CYP3A induc-
to bind both small and large ligands^"^. When the
tion by both GR agonists and antagonists was
LBD was cocrystallized with a hPXR ligand,
explained after a novel orphan NR was cloned and
SRI2813, it was discovered that the ligand could
characterized^^. Initial experiments to identify lig-
dock into the ligand binding pocket in three dif-
ands of the orphan receptor demonstrated that it
ferent orientations, each with a distinct pattern of
could be activated by many compounds, including
hydrogen bonding and van der Waals contacts^"^.
dexamethasone, 6,16a-dimethyl pregnenolone,
Thus, unlike many NHRs, the PXR can bind a
and PCN. The unusual pharmacology of the
variety of hydrophobic ligands in multiple binding
orphan receptor, specifically its activation by glu-
orientations.
cocorticoids (dexamethasone) and antiglucocorti-
coids (RU486 and PCN), strongly suggested that The ligand-binding specificity of the PXR is
it was the unknown mediator of CYP3A induction markedly different among species^^. For example,
observed in earlier studies. Further experimenta- PCN is a strong activator of rat and mouse PXR,
tion proved this to be the case and the receptor was but has little effect on rabbit or human PXR.
named the pregnane X receptor (PXR) because Conversely, rifampicin, phenobarbital, and
of its strong activation by natural and synthetic SR12813 activate both rabbit and human PXR, but
pregnanes. have little effect on rodent PXR. These species dif-
ferences are due to differences in the amino acid
After the identification of PXR in the mouse,
sequence of the LBD of the receptor. Using the
orthologues of the receptor were identified in
crystal structure of SR12813 bound to hPXR, four
many other species including human, rabbit, and
polar residues in the LBD that interacted with
j.^^20-22 jY^Q human orthologue was named steroid
SRI 2813 were identified that were different from
and xenobiotic receptor (SXR) and also pregnane
the corresponding amino acids in the mouse PXR
activated receptor (PAR)^^' ^^ For simplicity, we
(mPXR)^"^. When the residues in the mPXR were
will use the name PXR to refer to all orthologues.
mutated to the amino acids found in the human
A comparison of PXR amino acid sequences
receptor, the mutant mPXR was no longer respon-
among different mammalian species shows that
sive to the rodent-specific inducer PCN but rather to
while the DBD is highly conserved (>90% iden-
the human-specific agonist SR12813^'^. The
tity), the LBD displays much more variability
dependence of specific ligand binding on PXR
(-80% identity)23. In all species, PXR is highly
amino acid sequence has also been demonstrated in
expressed in the liver and to a lesser extent in the
vivo. A PXR-null mouse that has been "humanized"
small intestine^^' ^^. The PXR expression profile
by integrating an albumin-SXR (hPXR) transgene is
matches that observed for induction of CYP3A
responsive to the PXR ligands rifampicin and PB,
enzymes and provides further evidence for the
but is no longer responsive to PCN^^.
idea that PXR is the master regulator of CYP3A
expression.

2.4. Activation of Transcription


2.3. PXR Ligands and Species
Differences Analyses of PXR target gene promoters
revealed that the receptor can upregulate tran-
The PXR is a very promiscuous, low affinity scription by binding as a heterodimer with RXR to
receptor that is activated by a wide array of struc- several different motifs, including DR3, DR4, and
turally diverse compounds^^. Crystal structures of ER6 elements (Figure 8.1)^^. The human CYP3A4
326 Susanne N. Williams et al.

c
Substrate
CYP3A

Substrate-OH

Cytosol,

Figure 8.1. A model of the transcriptional regulation of CYP3A expression by PXR. The PXR binds as a
heterodimer with RXR to response elements in the promoter of CYP3A and other target genes. Binding of ligand to
the PXR results in increased CYP3A enzyme activity, which in turn increases the hydroxylation of substrates such
as steroids, bile acids, and drugs.

gene contains a proximal ER6 response element and its induction by dexamethasone^^. Similarly,
and a distal xenobiotic response element module binding of HNF-4a to a specific cis-acting ele-
(referred to as XREM) consisting of both an ment in the CYP3A4 gene promoter was found to
imperfect DR3 and an ER6 element^^. Although be necessary for transactivation of gene expression
PXR-mediated transactivation can be conferred by PXR or CAR^l Moreover, HNF-4a-null mice
by the proximal ER6 element alone, maximal express neither PXR nor CAR, indicating that
induction of the CYP3A4 gene requires both the expression of these receptors is regulated by HNF-
ER6 and XREM motifs^^. The PXR can also 4a^^. Given the complexity of emerging cross talk
activate the transcription of a number of CYP2B pathways among receptors, it is likely that other
genes, which are classically thought of as target NRs may be implicated in CYP3A regulation in
genes for the nuclear receptor, CAR^^~^^. the ftiture.
Interestingly, the PXR has been demonstrated to
upregulate CYP2B expression by binding to the
same DR4 elements upstream of the CYP2B gene
2.5. Mouse Models
to which CAR binds^^"^^. The reciprocal is also
true in that CAR can bind to response elements in An important advancement in the PXR field
the CYP3A genes to induce gene expression^^' ^^' came with the generation of a PXR-nuU mouse
^^ These findings and others have made it increas- model^^' ^^. Data obtained using these mice have
ingly clear that CAR and PXR serve broad and confirmed that the PXR plays a major role in reg-
often overlapping fiinctions^^' ^^. ulating CYP3A gene expression and in xenobiotic
In addition to PXR and CAR, other NRs are metabolism. Mice that lack PXR do not induce
also involved in the regulation of CYP3A CYP3A in response to PCN or other PXR-specific
expression. For example, activation of the GR by ligands and exhibit altered metabolism of xenobi-
dexamethasone increases the expression of both otics that are CYP3A substrates^^' '^^. The exact
PXR and CAR through glucocorticoid response role of the PXR in maintaining the constitutive
elements (GREs) in their promoters, and this can expression of CYP3 A remains unclear as the two
increase the expression of CYP3A^^^^. The tran- independently derived PXR-null models display
scription factor hepatocyte nuclear factor-4 (HNF- either unchanged or increased levels of basal
4) seems to play an important role in CYP3A C Y P 3 A 2 5 ' 4 0 ^
expression as well. It has been shown that binding Mouse models have also been used to demon-
of HNF-4 to the promoter of CYP3A23 is neces- strate a role for PXR in regulating the levels of
sary to maintain both its constitutive expression toxic bile acids. It had been previously established
Induction of Cytochrome P450 Enzymes 327

Cholesterol

Figure 8.2. An overview of the PXR's involvement in regulation of bile acid metabolism. Bile acids, such as LCA
can bind and activate the PXR to regulate hepatic gene expression. The PXR negatively regulates the expression of
CYP7A, which catalyzes the rate-limiting step in the conversion of cholesterol to bile acids. Conversely, the PXR
upregulates the expression of CYP3A and OATP2, which are involved in the metabolism and transport of bile acids,
respectively. This coordinate regulation of genes results in the increased clearance of toxic bile acids from the
hepatocyte.

that treatment of rats with PCN decreased the demonstrate that a regulatory loop exists by which
expression of CYP7A1, the enzyme that catalyzes elevated concentrations of bile acids activate the
the rate limiting step in the synthesis of bile acids PXR to block new bile acid biosynthesis and to
from cholesterol'*^ To examine whether the PXR induce the metabolism and excretion of existing
played a role in the repression of CYP7A1, a PXR- bile acids (Figure 8.2).
null mouse model was utilized"*^. It was demon-
strated that the PXR mediated not only the
repression of CYP7A1 by PCN, but also its basal
2.6. Future Research
expression. Additionally, the organic anion trans-
porter polypeptide 2 (0ATP2), a bile acid trans- The finding that CAR binds to many of the
porter, was found to be induced by PCN in same response elements as the PXR and that these
wild-type animals but not in PXR-nuU mice. When two receptors share many ligands and target genes
bile acids were examined for their ability to acti- has made it clear that the net effect of a xenobiotic
vate the PXR, it was found that a secondary bile on CYP3A gene expression will often depend on
acid, lithocholic acid (LCA), was an efficacious more than one receptor pathway. Identifying all of
activator of both mouse and human PXR"^^. In a the NRs involved in the regulation of CYP3A will
parallel study utilizing the humanized PXR mouse be necessary in the future. In addition, the pro-
model, bile acids such as LCA were identified as moters of many suspected PXR target genes,
PXR ligands that could induce CYP3A expression including CYP7A and 0ATP2, have not yet been
and it was shown that CYP3A catalyzed the characterized. Since the expression of CYP3A is
hydroxylation and detoxification of bile acids^^. coordinately regulated by PXR, CAR, and other
Administration of LCA to mice results in severe NRs, it seems likely that other PXR target genes
hepatotoxicity. Both studies demonstrated that are regulated in a similar fashion. The analyses of
PXR-null mice were resistant to LCA toxicity and regulatory regions in novel genes may provide
furthermore, that sustained activation of the PXR additional insight as to how the PXR interacts
protected against LCA-induced hepatotoxicity in with other NRs at response elements to regulate
wild-type mice"*^' ^^. Collectively, these findings gene expression. In the fiiture, continued analyses
328 Susanne N. Williams et al.

of the cross talk that occurs among the PXR and enhancer module (PBREM)^^' ^^. Sequence analy-
other NRs will be an exciting area of research sis revealed that the PBREM contained two DR4
that will eventually provide the details necessary motifs, commonly referred to as NR-binding sites
to understand how the PXR works with other one and two (NRl and NR2), which flanked a
receptors to form a master regulatory circuit that nuclear factor 1 (NFl)-binding site^^'^^.
controls CYP3A expression.

3.2. The Nuclear Receptor CAR


3. The Constitutive Androstane
A search for the receptors capable of binding
Receptor to PBREM ensued in the hopes of identifying the
elusive "PB receptor." Findings from two labora-
3.1. Introduction tories were incorporated to eventually identify the
In early studies it was observed that treatment NR that could bind PBREM in response to PB. In
of rats with phenobarbital (PB) caused a marked one experimental approach, proteins that could
proliferation of the liver and endoplasmic reticu- bind the NRl sequence of the PBREM were iso-
lum, an increase in DNA synthesis, and increased lated from PB-treated mouse liver nuclear extracts
activities of drug- and steroid hormone- using DNA affinity chromatography^^. When the
metabolizing enzymes^^' ^^. PB is now considered proteins were analyzed using electromobility shift
the prototype for a large group of structurally assays with the NRl element and various NR anti-
diverse, lipophilic chemicals that induce a similar bodies, it was found that the NRl-nuclear protein
spectrum of effects. PB and PB-type chemicals complex contained RXRa. A search of the litera-
induce the expression of numerous cytochrome ture revealed that a separate laboratory had previ-
P450 genes, including genes in the CYPIA, ously identified a liver-enriched orphan NR that
CYP2B, CYP2C, and CYP3A subfamilies^^' ^^ could function as a heterodimer with RXRa to
Of these, the CYP2B subfamily is most effectively bind a retinoic acid receptor element (RARE),
induced and will be discussed here as a paradigm. which contains a DR5^'^' ^^. The orphan receptor
had originally been identified as a "constitutively
The coordinate induction of hepatic enzymes
active receptor," or CAR, based on findings that
by PB has long been recognized to require direct
the receptor could activate transcription from a
activation of transcription'^^. While evidence was
RARE without the addition of exogenous lig-
suggestive of a receptor-mediated process, studies
and^^. Based on these earlier findings, the uniden-
aimed at identifying a PB-binding receptor were
tified NR binding to NRl of the PBREM was
hindered for years by lack of an appropriate model
postulated to be CAR. Further experimentation
system. A significant advance in understanding
using primary mouse hepatocytes and whole ani-
PB-induced gene expression came with the char-
mals proved this to be the case and, furthermore,
acterization of a regulatory element in the CYP2B
suggested that CAR could mediate the induction
genes. Using transgenic mice containing rat
of CYP2B by PB^^' 52,53,56 Contrary to findings
CYP2B2 promoter constructs of different lengths,
in early studies that used transfected cell lines, it
it was determined that PB-responsiveness was due
was later demonstrated in primary hepatocytes
to regulatory regions at least ~ 1 kb upstream of
and in vivo that CAR is sequestered in the cytosol
the CYP2B2 core promoter region"^^. Experiments
in untreated cells and that its nuclear translocation
in primary cultures of rat hepatocytes identified
is dependent on treatment with PB or PB-type
a 163 bp fragment —2.3 kb upstream of the
chemicals^^' ^^.
CYP2B2 gene that conferred PB-responsive activ-
ity and this enhancer was termed the PB response
element (PBRE)"^^. The responsiveness to PB con-
ferred by the PBRE was eventually refined to a 3.3. Mediators of CAR Activity
core 50 bp element that contained three distinct Although PB treatment induces the nuclear
DNA-binding motifs^^. Later, a similar 51 bp translocation and transcriptional activity of CAR,
enhancer was characterized in the mouse results from ligand-binding assays have indicated
CYP2B10 gene and was termed the PB responsive that neither PB nor known PB metabolites are
Induction of Cytochrome P450 Enzymes 329

®.

Figure 8.3. A model of CAR-mediated induction of CYP2B expression by PB. Inactive CAR normally resides in
the cytoplasm. PB acts on unknown cellular targets to induce the nuclear accumulation of CAR. In the nucleus,
activated CAR heterodimerizes with RXR to bind response elements in the promoter of CYP2B and other target
genes to induce gene expression. Phosphorylation events are thought to be important in regulating the CAR signaling
pathway. Inset: In the absence of PB and other CAR activators, inverse agonists can bind CAR and repress its
transcriptional activity.

bona fide CAR ligands. So how might PB func- androstanes are effective mouse CAR (mCAR)
tion to activate CAR? Because of CAR's apparent inverse agonists, they have little effect on human
constitutive activity, it has been postulated that CAR (hCAR). Also, it is important to note that
subcellular localization may be a major determi- supraphysiological concentrations of androstanes
nant of receptor activity. In this regard, treatment are required to repress mCAR-mediated gene
with okadaic acid, a phosphatase inhibitor, has expression; thus, androstanes are not likely the
been shown to block the PB-induced nuclear physiological ligand of CAR. Interestingly, phar-
translocation of CAR^^. These findings suggest macological concentrations of several endogenous
that the localization of CAR in the cell may steroids have also been demonstrated to activate
be regulated by phosphorylation events. Thus, PB (estrogens) or repress (androgens, progesterone)
may activate CAR-mediated gene transcription by mCAR activity while having little effect on hCAR
altering the phosphorylation status of the receptor activity^^. Thus far, the only identified steroidal
or related cellular targets, resulting in CAR's compound that exerts activity toward hCAR is the
translocation to the nuclear compartment progesterone metabolite 5p-pregnane-3,20-dione,
(Figure 8.3). Additional phosphorylation events which at pharmacological concentrations can
in the nucleus have also been postulated to be directly bind the receptor and increase its activity
important^^. above the constitutive leveP^. Collectively, these
Ligands that bind directly to CAR have findings raise the intriguing possibility that a yet
also been identified. Initially, in a search for CAR unidentified physiological steroid may function as
activators, it was discovered that the constitu- an endogenous CAR ligand to either activate or
tive activity of CAR could be repressed by repress activity.
androstanes, which are testosterone metabolites^^. Direct-binding xenobiotic ligands of CAR have
As a result, CAR is now known as the "constitu- also been identified and many of these show
tive androstane receptor." Androstanes bind CAR marked species specificity as well. For example,
directly to repress its transcriptional activity and the planar hydrocarbon l,4-"bis[2-(3,5-dichloropy-
have been termed "inverse agonists"^^' ^^. While ridyloxy)]benzene binds directly to mCAR and is
330 Susanne N. Williams et a/.

the strongest agonist identified to date, but Aside from the PXR, other NRs are also
it apparently lacks activity toward hCAR^^. important in CYP2B expression. As mentioned
Conversely, an agonist selective for hCAR, 6-(4- earlier, HNF-4a is critical for CAR expression,
chlorophenyl)imidazo[2,l-b][l,3]thiazole-5- as HNF-4a-null mice express neither PXR nor
carbaldehyde 0-(3,4-dichlorobenzyl)oxime, has CAR^^. Both the GR and HNF4-a can bind to ele-
recently been identified^ ^. Moreover, the antifun- ments in the CAR promoter to regulate the level of
gal agent clotrimazole is a potent inverse agonist CAR expression, which in turn can influence the
of hCAR while it has little or no activity toward expression of CYP2B and likely other CAR target
mCAR^^. Other CAR xenobiotic activators that genes^^' ^^. The study of interactions of NRs with
have been reported include PCBs, chlorinated the CAR pathway is a relatively new area of inves-
pesticides such as DDT, and methoxychlor^*' ^^. tigation and roles for other NRs in CAR-mediated
CYP expression are likely to be identified in the
future.
3.4. Activation of Transcription
The fact that both PB and direct-binding lig- 3.5. IVIouse Models
ands can regulate CAR suggests that there are
multiple mechanisms for the regulation of CAR The generation of mice null at the CAR locus
activity. While important differences likely exist has recently been reported^"^. Mice lacking CAR are
in the cellular targets affected by receptor agonists resistant to many of the toxic effects of PB, includ-
compared to PB, agonists of CAR are similar to ing hepatomegaly and increased DNA synthesis,
PB in that they induce the nuclear translocation confirming that CAR mediates these toxic pheno-
and binding of CAR to DNA. A model of how PB types^"^. In addition, studies using this model have
may induce CYP2B expression through the CAR confirmed that CAR is essential in mice for the
pathway is shown in Figure 8.3. PB interacts with induction of the CYP2B genes by PB^l The CAR-
unknown cellular targets to likely alter the phos- null model has been invaluable in identifying novel
phorylation status of CAR and induce its translo- PB-inducible genes that are regulated by CAR. The
cation to the nucleus. The receptor may undergo analysis of over 8,500 genes using DNA microarray
further modifications before binding as a het- technology was recently performed to examine PB-
erodimer with RXR to PBREM to induce CYP2B induced hepatic gene expression in CAR-null mice
expression. The PBREM is highly conserved in compared to wild-type mice^^. Findings from this
rat, mouse, and human CYP2B genes. The NRl study demonstrate that CAR mediates the PB-
site seems to serve as the major CAR-binding site inducible expression of numerous hepatic genes,
and is critical for CAR transactivation of CYP2B both negatively and positively. After PB treatment,
genes^^. Once bound to PBREM, the final effect the expression of more than 70 genes was found to
of CAR regulators on gene expression seems to be be dependent on CAR, while 60 genes were regu-
determined by the ability of CAR to recruit coac- lated in a CAR-independent manner. About half
tivators to the transcriptional complex. In this of the CAR-dependent genes encoded xenobiotic
regard, it has been demonstrated that CAR can metabolizing enzymes (XMEs), highlighting the
interact with a number of coregulators, including importance of this receptor in protecting organisms
SMRT, SRC-1, and GRIP-l^^^ 62,63 against xenobiotic exposure. Interestingly, some
In addition to the DR4 elements in PBREM, CAR-dependent genes were downregulated in
CAR can bind to a variety of DNA motifs includ- response to PB and were found to encode proteins
ing DR3 elements, DR5 motifs (e.g., those found that play roles in basic liver function, fatty acid
in RARE), and ER6 motifs^^' ^'^' ^^. These response metabolism, and signal transduction. These find-
elements are the same as those recognized by PXR ings provide evidence for the idea that CAR is
and, not surprisingly, CAR and the PXR share not only important in regulating the expression
many overlapping target genes"^^. Indeed, it has of XMES, but also that it plays an important
been demonstrated that CAR transactivates the physiological role as well.
CYP3A genes by binding to the same response Using a combination of both PXR- and CAR-
element that serves as the PXR-binding site^^' ^^' ^^ null mice, the ability of CAR and PXR to share
Induction of Cytochrome P450 Enzymes 331

response elements and induce the same target dependence of CAR signaling on phosphorylation
genes has been demonstrated in vivo. For exam- may represent a model of activation that could
ple, treatment of PXR-null mice with PB results in be applicable to the other xenobiotic receptors. It
the induction of CYP3A and this has been shown is not known whether physiologically relevant
to occur through CAR binding to the CYP3A pro- endogenous agonists and inverse agonists exist. If
moters^''^^. Similarly, treatment of CAR-null mice identified, these endogenous CAR ligands will
with the mPXR ligand dexamethasone results in offer clues as to what role CAR plays in normal
CYP2B induction through the binding of PXR physiology. Compared to the PXR, CAR seems to
to PBREM"^^. Through studies such as these, the bind to a more limited spectrum of steroidal com-
relative contribution of each of these receptors on pounds and xenobiotics. Solving the crystal struc-
CYP gene expression has begun to be explored. ture of CAR'S LBD will allow for the investigation
A "humanized" mouse model that expresses of how ligand specificity is determined between
hCAR rather than mCAR in the liver has recently these two receptors and may provide informa-
been engineered^^. Given the fact that the effect of tion useful in the evaluation of their separate but
xenobiotics on CAR activity differs significantly overlapping roles in regulating gene expression.
among species, this model should prove useful in Finally, the most challenging area of research for
evaluating the relevance of toxic responses. For the future will be in understanding how CAR, the
example, once the toxicity of an agent is deter- PXR, and other NRs interact to regulate CYP gene
mined to be dependent on mCAR using the CAR- expression.
null mouse model, the humanized mice can be
used to evaluate whether the toxic response can
also be mediated by hCAR. Recent studies
employing this approach have implicated CAR in 4. The Peroxisome Proliferator
the hepatotoxicity of acetaminophen in humans^^. Activated Receptor a
It was found that acetaminophen at high doses
activates hCAR and induces the expression of 4.1. Introduction
CYP1A2 and CYP3A, which are the enzymes that
catalyze the rate-limiting step in the formation of Peroxisome proliferators (PPs) are a group of
toxic acetaminophen metabolites. These findings structurally dissimilar chemicals that cause a sim-
have identified CAR as a possible therapeutic tar- ilar spectrum of effects including a proliferation
get in cases of acetaminophen overdose^^. In addi- of peroxisomes in the hepatocyte, liver hyperpla-
tion, a separate study using humanized mice and sia, and an increase in the expression of numerous
CAR-null mice demonstrated that CAR plays a enzymes involved in fatty acid oxidation^^. The
role in protecting the body from elevated bilirubin enzymes upregulated by PPs include a large num-
levels by inducing the expression of enzymes ber of enzymes involved in the p-oxidation of
involved in bilirubin clearance^^. fatty acids and the CYP4A enzymes, which are
important in the co-oxidation of many medium and
long-chain fatty acids^^' ^^. In the body, fatty acids
are oxidized to produce energy when other sub-
3.6. Future Directions strates are not available, such as during times of
The identification of PBREM and CAR, have fasting or starvation^^.
led to major advances in understanding how PB The ability of PPs to cause the rapid, coordinate
and PB-like chemicals regulate gene expression transcriptional upregulation of gene expression in
However, many unanswered questions remain. a tissue-specific manner suggested that PPs acted
Exactly how PB interacts with the CAR signaling through a NR-mediated mechanism^ ^ This proved
pathway to induce gene expression is still unclear. to be the case when a screen for novel NHRs iden-
Moreover, it is not known if CAR agonists mediate tified a mouse cDNA encoding an orphan receptor
gene expression by mechanisms similar to or that could be activated by known PPs, such as the
distinct from those of PB. In addition, fiirther drug clofibrate^^. The receptor was named the per-
investigation into how phosphorylation is involved oxisome proliferator activated receptor, or PPAR^^.
in regulating the CAR pathway is important. The In later studies, the rat homologue of the PPAR
332 Susanne N. Williams et a/.

was identified and it was found that the receptor phthalate, trichloroacetic acid, and the pesticide
could not only be activated by PPs, but also by j)jj8o Interestingly, these xenobiotics induce the
endogenous fatty acids^^. expression of CYP4A even though this enzyme
does not seem to play a role in their metabolism.
Many of the endogenous fatty acids that are
4.2. PPAR Isoforms metabolized by CYP4A are also PPARa ligands.
The PPAR that was originally cloned from These include an array of saturated and unsatu-
mouse is now known as the alpha isoform, or rated very long-chain fatty acids, such as linoleic
PPARa. This designation arose after the identifi- acid, palmitic acid, and arachidonic acid^^' ^^
cation of two additional distinct PPAR isoforms, Moreover, findings using acyl-CoA oxidase
termed PPARp (also referred to as 8) and PPAR7 (AOX)-null mice suggest that the acyl-CoA deriv-
The three PPAR isoforms are encoded by three atives of very long-chain fatty acids are most
separate genes and have been identified in many likely endogenous PPARa ligands. In mice with a
species including human, rat, and rabbit^^. The disrupted AOX gene, acyl-CoA derivatives accu-
three PPAR isoforms play distinct roles and mulate to high levels and the animals display a
display tissue specific expression pattems^"^. The phenotype similar to that seen after treatment of
PPARa is highly expressed in the liver and kid- rodents with synthetic PPs^^. Some eicosanoids
ney and plays a major role in regulating the catab- and eicosanoid metabolites that are important
olism of fatty acids. Not surprisingly, the CYP4A mediators of inflammation, including leukotriene
enzymes are coexpressed with PPARa in these B4 and prostaglandins, are PPARa ligands^^' ^^.
tissues^^. The PPAR7 gene actually gives rise to These arachidonic acid derivatives can be metab-
two gene products, PPAR7I and PPAR72, through olized by CYP4A to compounds that are inactive
differential promoter usage. The PPAR72 isoform in terms of mediating the inflammatory
is highly expressed in adipose tissue and mediates response^^. In light of the role that PPARa plays in
adipogenesis and lipid storage; however, PPAR7I, the induction of CYP4A, it is not surprising that
which is expressed more broadly and at lower lev- mice lacking PPARa have been demonstrated to
els, can also induce adipogenesis^^. The PPARp display a prolonged inflammatory response^^.
is ubiquitously expressed and while the exact
physiological function of this isoform is still
unclear, recent findings have suggested that this 4.4. Activation of Transcription
isoform modulates the activity of both PPARa
and PPAR7^l Since PPARp and PPAR7 do not The experimental drug Wy 14643, an acetic
seem to regulate the expression of CYP4A or any acid derivative of clofibrate, is a potent PPARa
other P450 enzyme, these isoforms will not be agonist and was instrumental in elucidating the
discussed to any great extent. signaling pathway of PPARa. The PPARa binds
as a heterodimer with RXR to DNA motifs
termed peroxisome proliferator response elements
(PPREs) (Figure 8.4)^^ The core PPRE sequence
4.3. PPARa Ligands
was initially identified as an imperfect DRl motif
While the quantitative effects of agonist bind- by analyzing the promoter of the AOX gene^^.
ing on the activity of PPARa seem to be species- Unlike the PXR and CAR, PPARa can form het-
specific, the spectrum of ligands that can activate erodimers with either ligand-free or 9-cis retinoic
the PPARa across species is similar. Clofibrate, acid-bound RXR, and ligand binding to either
originally recognized for its ability to increase both RXR or PPARa can activate gene expression
the number and size of peroxisomes when admin- through PPREs^^' ^^. Other NRs can also bind to
istered to rats, is considered the prototype for a PPREs and competition for binding has been
class of drugs called fibrates, which are all potent observed among the three PPAR isoforms as
PPARa ligands^^' ^^. The fibrate drugs are widely well as HNF-1, thyroid receptor, and RXR/RXR
used today as lipid lowering agents in humans. dimers. Depending on the NR complex bound to
Other synthetic ligands of the PPARa include the PPRE, the transcription of a target gene can be
the industrial plastisizer mono (2-ethylhexyl) either activated or repressed. Studies have shown
Induction of Cytochrome P450 Enzymes 333

Fatty Acid Ligands


9-cis retinoic acid Fibrate Drugs

a aa V
Nucleus

G)-Oxidation Enzymes
(CYP4A)
P-Oxidation Enzymes
FA IVIetabolism Genes

PPRE

Figure 8.4. A model of the transcriptional regulation of gene expression by PPARa. The PPARa binds as a
heterodimer with RXR to PPREs upstream of target genes. Ligand binding to either the RXR or PPAR activates the
transcriptional complex resulting in the induction of numerous genes involved in fatty acid oxidation and
metabolism, such as CYP4A.

that the specificity of a PPRE for NR binding is recently been resolved^^' ^'^. It was found that ago-
determined not only by the sequence of the DRl nists cause the recruitment of coactivators by
element, but also by the sequence immediately 5' interacting with the ligand-dependent activation
ofthePPRE^O'^i. helix (AF-2) to maintain it in an active conforma-
As with other NRs, transcriptional regulation tion. In this active conformation, the AF-2 helix
by the liganded PPAR involves the interaction of can bind tightly to LXXLL motifs in coactivators
many cofactors that form distinctive multiprotein and stabilize coactivator binding into a hydropho-
complexes. In addition to several common coreg- bic cleft that is formed in the receptor^^. In the
ulators that have been shown to interact with the unliganded state, the PPARa is preferentially
PPAR (e.g., SRC-1, CBP, SMRT), coactivators bound to corepressors. The crystal structures
that seem to be PPAR-specific have also been reveal that corepressor motifs bind to a hydropho-
identified including PPAR binding protein (PBP) bic groove in the receptor and prevent the AF-2
and PPAR interacting protein (PRJP). Interest- helix from interacting with coactivators. The bind-
ingly, a transcriptionally active PPARa-interacting ing of antagonists fiirther stabilizes the inactive
cofactor (PRIC) complex from rat liver nuclear conformation of the receptor by altering the posi-
extracts was recently isolated and was found to tion of a residue in the AF-2 helix that is critical to
contain over 25 different proteins, including PBP, agonist binding. These studies have demonstrated
PRIP, CBP, and a novel coactivator PRJCISS^^. how ligands can promote basic structural changes
The composition of this PRIC complex may in the PPARa to mediate its interaction with
provide an insight into the basis for differences in coregulators, and hence, its transcriptional activ-
tissue and species sensitivity to PPs. ity. Results of these studies have also allowed
The crystal structures of the PPARa LBD for the realization that NRs can distinguish
bound to an agonist with a coactivator motif from coactivators from corepressors by the length of
SRC-1 and, alternatively, bound to an antagonist their conserved interaction motifs. Importantly,
with a corepressor motif from SMRT have these findings have also resulted in a model of
334 Susanne N. Williams ef al.

receptor repression that can likely be applied to While exhibiting no detectable gross phenotype in
many NRs^'^. the fed state, experiments using null mice have
demonstrated that the PPARa plays an important
role in the hepatic response to fasting. Unlike
4.5. Species Differences wild-type mice, fasted PPARa null animals do
not upregulate the expression of fatty acid oxida-
Quantitatively, the response of humans and
tion enzymes, including CYP4A, and they exhi-
rodents to PPs has been found to differ dramati-
bit hypoglycemia, hypoketonemia, and elevated
cally. Humans are indeed responsive to PPs in
plasma levels of free fatty acids^^^. These findings
regard to their ability to reduce serum lipids in vivo,
and others have demonstrated that PPARa plays
a response known to be mediated by PPARa in
a central role in maintaining lipid homeostasis.
rodents^^. However, chronic exposure of rats and
The PPARa-null mice do not display the typi-
mice to PPs causes a dramatic peroxisome prolifer-
cal toxic responses after exposure to PPs. Studies
ation response in the liver and eventually leads to
have shown that treatment of null mice with PPs
liver tumors. These PP-induced toxicities have not
does not induce liver hyperplasia, peroxisome
been observed in humans even though the fibrate
proliferation, or hepatocarcinogenesis, confirm-
drugs have long been used at high doses in humans
ing that PPARa is the mediator of these PP-
to lower triglyceride and cholesterol levels^^' ^^.
induced toxic responses^^' ^^^ Moreover, these
Several mechanisms have been postulated to play a
findings suggest that the PPARp and PPAR7 iso-
role in the seemingly refractory nature of humans
forms do not play a critical role in these PP-
to PP toxicities. Different expression levels of
induced toxicities. The induction of CYP4A and
PPARa and the existence of a splice variant of
many other fatty acid oxidation enzymes in
PPARa in humans that may negatively regulate
response to PPs is also absent in mice lacking
PPARa have been suggested to play a role^^' ^^.
PPARa confirming that it mediates the induction
Recently, the human PPARa transgene was intro-
of these enzymes. Interestingly, while PPARa-null
duced by an adenoviral approach into PPARa-nuU
mice have lost the CYP4A induction response,
mice and was found to be as effective as the mouse
basal levels of CYP4A are not affected, indicating
PPARa in transcriptionally activating PPARa tar-
that other NRs control the constitutive expression
get genes under in vivo conditions'^. The findings
ofCYP4Aio2.
of this study demonstrate that the human PPARa is
fully competent to induce PP-induced pleiotropic Studies using rodents have also demonstrated
responses in the context of mouse liver''. Thus, that a normal AOX gene is necessary for proper
other factors in the human liver environment are physiological regulation of the PPARa^^. The
likely important in PPARa function and in deter- AOX gene encodes an enzyme critical in the
mining the PP response in humans. Competition P-oxidation of certain very long-chain fatty acid
between PPARa and other NRs for binding to RXR acyl-CoA metabolites^^. Targeted disruption of
or coactivators has been postulated to play a role in the AOX gene in mice results in sustained PPARa
species differences^^. Moreover, differences in the activation, leading to profound peroxisome prolif-
sequence of PPREs and surrounding sequences in eration and increased levels of PPARa target
target genes exist between humans and rodents, and it genes, such as the CYP4A genes^^. These findings
is not known exactly how this affects PPARa trans- suggest that acyl-CoA metabolites, and possibly
activation potential in vivo. The analysis of changes in other unmetabolized oxidase substrates, are
global gene expression in wild-type and null animals endogenous ligands of the PPARa and that AOX
in response to PPs has been performed using DNA is critical in metabolizing these ligands in vivo.
microarrays and this approach may eventually allow
for a better understanding of how the PPARa medi- 4.7. Future Directions
ates the toxic response to PPs in rodents^^^.
Over the last 10 years, great strides have been
made in understanding the biology of the PPARa.
4.6. Mouse Models The synthesis of specific and potent PPARa
agonists have made it possible to examine the
A PPARa-null mouse model has been gener- mechanism of signal transduction of the PPARa.
ated and the animals are viable and fertile^^^. Furthermore, the resolution of the crystal structures
Induction of Cytochrome P450 Enzymes 335

of PPARa bound to ligands and coregulators has than DBA mice to the PAH-induced upregula-
resulted in a model of how agonists and antagonists tion of AHH activity^^^. Using classical genetic
alter the conformation of NRs to mediate coregula- approaches, the locus responsible for the AHH
tor binding. In the future, a more complete under- inducibility phenotype was shown to segregate in a
standing at the molecular level is needed as to how simple autosomal dominant fashion. This locus was
PPAR/coregulator complexes interact with other termed the ''Ah" locus because of its ability to
proteins to modulate gene expression in a species- mediate responsiveness to aryX /hydrocarbons ^^^' ^^^.
and tissue-specific fashion. The allele found in the more responsive C57BL/6
The generation of a PPARa-null mouse has strain was designated as Ah^ while the allele that
been critical in establishing a major role for conferred decreased responsiveness in DBA mice
PPARa in lipid homeostasis and has confirmed was termed ^Z^'^^^^.
the role of PPARa in PP-induced toxicity in The second line of evidence came from phar-
rodents. However, many questions remain con- macological studies using an extremely potent
cerning differences between mice and humans inducer of AHH activity, 2,3,7,8-tetrachloro-
in regard to the PPARa pathway. The basis for dibenzo-/7-dioxin (TCDD or "dioxin")^^^. Using
species differences in the response to PPs is radiolabeled TCDD, a receptor in mouse liver
unclear and it is not known what role differences cytosol was identified that bound this ligand with
in the expression of CYP4A and other PPARa tar- high affinity and in a saturable and reversible
get genes play in mediating this response. In the manner^i^' ^^^ The proof that this TCDD-binding
future, generation of a "humanized" PPARa site was in fact the AHR was 3-fold. First, it was
mouse, such as those available for the NRs, PXR found that receptor isolated from mice harboring
and CAR, will be useful for long-term studies to the responsive Ahf^ allele bound ligands with
investigate species differences and to allow for higher affinity than did receptor isolated from
the more accurate extrapolation of findings to mice harboring the less responsive Ah^ allele^^^'
human risk assessment when evaluating PP- 106, 112, 113 Second, competitive binding studies
induced toxicities. with various dioxin congeners revealed that bind-
ing affinities correlated with their potency as
inducers of AHH activity^ ^'^^^^. The last line of
evidence was biochemical in nature. In the absence
5. The Aryl Hydrocarbon of ligand, the receptor was found in the cytosolic
Receptor fraction of cell extracts; however, the binding
site/receptor was found in the nuclear fraction after
exposure to ligand^ ^^. Thus, genetic, biochemical,
5.1. Introduction and pharmacological evidence demonstrated that
Nearly 50 years ago, it was noted that rats the Ah locus encoded the AHR and this protein
exposed to 3-methylcholantlirene (3-MC) dis- was the mediator of AHH induction.
played a marked increase in metabolic capacity
toward that substrate and other polycyclic aro-
matic hydrocarbons (PAHs)^^^. This enhanced 5.2. The AHR
metabolic activity was referred to as "aryl hydro-
carbon hydroxylase" (AHH) based on the ability It was many years before the AHR was cloned
of these enzymes to efficiently hydroxylate aro- and characterized. Attempts to purify the receptor
matic hydrocarbons ^^'^. It is now known that AHH were initially hampered by its low cellular
activity is the collective activities of the CYPlAl, concentration and relative instability. The devel-
CYP1A2, and CYPIBI enzymes. opment of a photoaffinity ligand, 2-azido-3-
Over the next 30 years, two lines of evidence [^^^I]iodo-7,8-dibromodibenzo-p-dioxin, was the
led to the identification of the AHR, the protein essential step that allowed the eventual purifica-
that functioned as the PAH sensor and regulated tion of the AHR^^^' ^^^. Once the receptor was
AHH activity. The first indications that such a purified, a partial amino acid sequence was
receptor existed came from genetic studies of obtained and this lead to the cloning of the AHR
inbred mouse strains. Early studies demonstrated cDNA from mouse liver^^^' ^^^ The deduced
that C57BL/6 mice were much more responsive amino acid sequence revealed that the AHR was
336 Susanne N. Williams et al.

as a member of the PAS superfamily of pro- mesenchymal cells^^^' ^^^. This indicates that fac-
teins ^^°' ^^^ The AHR was found to be most simi- tors other than AHR expression level are involved
lar in amino acid sequence to the AHR nuclear in the tissue specific expression of these CYPl
translocator (ARNT). Interestingly, ARNT had genes.
been cloned only a year before in a screen to iden-
tify gene products that were important in AHR
signaling in a mouse hepatoma cell line^^^. One 5.3. AHR Ligands
mutant cell line that was deficient in signaling
expressed normal amounts of AHR, but the cells Putative orthologues of the AHR have been
did not upregulate AHH activity after agonist identified in numerous higher eukaryotes, includ-
exposure. A human genomic DNA fragment that ing nematodes, insects, fish, birds, and mammals.
rescued the mutant phenotype was found to con- Striking differences in molecular weight of the
tain the ARNT gene product. Further experimen- AHR are observed among various species, and
tation demonstrated that the corresponding ARNT even in different strains of laboratory mice. This
protein was required to direct the activated AHR difference is mostly due to differences in the
to specific regulatory elements upstream of target length of the C-terminus and results from different
genes such as CYPlAl^^^. The structural similar- stop codon usage. Despite differences in receptor
ities of the AHR and ARNT were recognized and size, the vertebrate AHR signaling pathway is
it was postulated that the proteins might be dimer- highly conserved across species and the induction
ization partners. This proved to be the case, mak- of CYPl gene expression is observed in all
ing AHR and ARNT the first PAS protein species•^^' ^^'^. Importantly, significant species and
heterodimer to be shown to have physiological strain differences have been observed in ligand-
relevance^^^' ^^^. binding affinities. It seems that changes in
The overall structural organization of the AHR specific amino acid residues in the LBD may be
is typical of most members of the PAS superfam- responsible for these differences. For example, the
ily of proteins. The N-terminus of the AHR Ah alleles found in C57BL/6 and DBA mice
contains a bHLH domain that is important in exhibit a 10-fold difference in ligand binding and
dimerization and subsequent positioning of the this arises, at least in part, from an alanine to
basic regions of the proteins such that they can valine substitution at amino acid 375^^^' ^^^.
bind to specific DNA enhancer motifs^^^~^^^. As Moreover, the human AHR has the same mutation
in most PAS proteins, the bHLH region is found at the corresponding amino acid and is similar to
immediately N-terminal to the PAS domain. The the Ah^ allele in that it binds the ligand with 10-
250-300 amino acids comprising the PAS domain fold less affinity compared with the Ah^ allele^^'^.
contain two highly degenerate repeats, termed "A" Since the crystal structure of the AHR has not
and "B" repeats^. The PAS domain of the AHR been solved, the identification of amino acids
harbors the LBD, a dimerization surface for bind- important in ligand binding has relied upon the
ing to ARNT, and an interaction surface for chap- examination of ligand-binding affinities of AHRs
erones such as Hsp90 and ARA9 (also called AIP, with different amino acid mutations.
or XAP2)6' *28.129 j^Q ^egJQj^ ^f^y^^ AHR jj^p^^. The most extensively studied agonists are the
tant in ligand binding and chaperone binding over- halogenated aromatic hydrocarbons such as
laps the PAS B repeat^25, no J^IQ C-terminus of TCDD, polychlorinated biphenyls, and polychlori-
the AHR encodes a hypervariable TAD*^^. nated dibenzofiirans as well as PAHs such as
The AHR is expressed in many cell types and benzo[a]pyrene and 3-MC^. One of the highest
tissues with high levels of expression found in affinity ligands of the AHR and the most potent
placenta, lung, thymus, and liver. The expression inducer of CYPl Al expression is TCDD. As the
profile of the AHR is in good agreement with result of this ligand-receptor interaction, expo-
the expression of PAH-target genes. However, the sure to TCDD produces a wide variety of toxic
expression of CYPl genes is fairly tissue specific, effects that are species- and tissue-specific^. The
with CYP1A2 primarily found in the liver, response to TCDD is due to the fact that TCDD
CYPlAl highly expressed in epithelial cells has a remarkably high affinity for the AHR (on the
throughout the body, and CYPIBI found in order of 10~^^M, K^) and that this ligand is
Induction of Cytochrome P450 Enzymes 337

resistant to metabolism. The toxic endpoints are 5.4. Activation of Transcription


dependent on the AHR and are thought to arise
from long-term alterations in AHR-mediated While it has long been recognized that the
gene expression, but it is still unclear if TCDD expression of CYPl genes is regulated at the level
toxicity involves the transcriptional upregulation of transcription, it took many years to develop our
of CYPIA genes. The discussion of TCDD here current understanding of how the AHR mediates
will focus primarily on its use as a prototype ago- upregulation of gene transcription in response
nist of the AHR and the mechanism by which it to xenobiotics. An overview of the mechanism
acts as an inducer of the CYPl genes. of AHR-mediated gene expression is shown in
Apart from xenobiotics, it is assumed the AHR Figure 8.5. In the absence of ligand, the AHR is
recognizes some endogenous ligand. While some found in a cytosolic complex with two molecules
endogenous compounds, such as heme degradation of Hsp90, an immunophilin-like chaperone pro-
products, have been shown to bind and activate the tein known as ARA9 and the chaperone p23^^^~^'^^.
AHR, no compound has been convincingly demon- The Hsp90 chaperone is a necessary component of
strated to be the bona fide "endogenous AHR the AHR pathway and seems to anchor the recep-
ligand"^^^. Naturally occurring AHR ligands have tor in the cytosol as well as hold the protein in a
been found in teas, fruits, vegetables, and herbal high affinity ligand-binding conformation^^^"^"^^.
supplements and include polyphenolic compounds The ARA9 protein has been shown to increase the
such as flavonoids, indoles, and various carotinoids. amount of properly folded AHR in the cytoplasm,
The continued identification and analysis of these while the chaperone p23 has been suggested to play
naturally occurring ligands may provide insight that a role in regulating ligand responsiveness and
could lead to the identification of an endogenous receptor translocation^'^^' ^^^.
AHR ligand in the friture, or to the identity The signal transduction pathway of the AHR is
of the environmental stresses that have led to the well characterized and analogous to that of many
evolutionary conservation of the receptor system. NHRs, described above. Ligand binding to the

Cytosol

ARi

Dioxin

Figure 8.5. A model of AHR signal transduction. The AHR normally resides in the cytoplasm with the chaperones
Hsp90, ARA9, and p23. Upon ligand binding, the AHR translocates to the nucleus where it exchanges its chaperones
for ARNT. The AHR/ARNT heterodimer binds to dioxin response elements (DREs) to activate the transcription of
downstream target genes, including the AHRR and XMEs, such as CYPIA. The ligand-activated AHR is exported
from the nucleus and degraded through a proteosome pathway, or may undergo recycling within the cytoplasm. The
AHRR protein, a negative regulator, can compete with the AHR for dimerization with ARNT resulting in inhibition
of AHR-mediated gene expression.
338 Susanne N. Williams ef a/.

cytosolic AHR induces a conformational change The ARNT protein serves as a dimerization
and nuclear translocation of the receptor. In the partner not only for the AHR, but also for other
nucleus, the AHR sheds some of its associated PAS proteins, such as the hypoxia inducible factors
chaperones and binds to its partner ARNT^"^^"^^^. (HIFla, HIF2a, HIF3a). When in a complex with
The resulting AHR/ARNT heterodimers bind to ARNT, these various heterodimers mediate the
specific enhancers in DNA to alter DNA confor- upregulation of various genes important in dealing
mation and increase the transcription of target with cellular hypoxia^^'^. It has been postulated that
genes^^^ The enhancers, made up of the consen- competition among PAS proteins for the limited
sus sequence 5'-TNGCGTG-3', were first charac- pool of ARNT could be an important mechanism of
terized in the mouse CYPlAl gene and have been transcriptional regulation. Some studies have found
called "dioxin responsive elements" (DREs), that activation of the HIFla pathway can interfere
"xenobiotic responsive elements" (XREs), or with AHR-mediated induction of CYPlAl ^^^i^l
"AH-responsive elements" (AHREs)^^^"'^^. For However, others have reported that simultaneous
the remainder of this chapter, we shall refer to the activation of both the HIFla and the AHR path-
enhancers as DREs. In addition to nucleotides in ways caused no changes in the expression level of
the core DRE, sequences outside of the DRE can any AHR or HIFla target genes, suggesting ARNT
modulate the binding affinity of AHR/ARNT to is not a limiting factor^ ^^. These conflicting results
DNA and appear to be important determinants of may be due to differences in cell type and/or exper-
AHR-mediated gene expression^^^"^^^. imental conditions. Although cross talk between
Functional DREs have been identified upstream the AHR and HIFla pathways seems to occur
of numerous AHR-inducible genes, many of which under certain conditions in vitro, it remains to be
encode XMEs. These genes are collectively referred proven that competition for ARNT occurs in vivo
to as the Ah gene battery and include CYPlAl, and what, if any, effect this has on CYPl gene
CYPlA2, CYPIBI, NQOl (NADPH: quinone oxi- expression or TCDD toxicity.
doreductase), ALDH3A1 (an aldehyde dehydroge-
nase), UGT1A6 (a UDP glucuronosyl transferase), 5.5. Mouse Models
and GSTYa (a glutathione ^S-transferase)^^^. The
coordinate upregulation of these enzymes results Targeted disruption of the Ah locus in mice
in the enhanced metabolism of most inducers to has been achieved by a number of laboratories.
hydrophilic compounds that can be more easily As expected, AHR-null mice fail to upregulate
excreted from the body. Thus, the AHR plays an CYPlAl, CYP1A2, and other members of the
integral role in mediating the adaptive response to Ah battery in response to AHR agonists^^^' ^^^.
EAHs and related environmental chemicals. Furthermore, AHR-null mice are resistant to
Another interesting aspect of the AHR pathway TCDD- and PAH-induced toxicity, confirming
is that prolonged agonist exposure results in the that the AHR is the mediator of dioxin toxicity^^^'
attenuation of signaling through the AHR. One •^^. The AHR-null mouse models have also pro-
mechanism by which this occurs is mediated vided evidence for a physiological role for this
through the AHR repressor protein (AHRR)'^^' ^^^. receptor. These mice have defects in vascular
The AHRR is structurally similar to the AHR, development, display decreased fertility, and have
except it lacks the PAS B-domain and its C- overall decreased body weight compared to wild-
terminus functions as a transcriptional repressor. type mice. Thus, in addition to mediating TCDD
Because of these features, the AHRR can dimerize toxicity and the adaptive response to PAHs and
with ARNT in a manner that is independent of other chemicals, the AHR clearly plays an impor-
agonist. This heterodimer can bind to DREs to tant role in development. Such an observation
repress target gene transcription^^^' ^^'. The supports the hypothesis that the AHR has an
expression of the AHRR gene is controlled by a unknown endogenous ligand.
DRE and its transcription is upregulated upon
exposure of the cell to AHR ligands. Another way
5.6. Future Directions
the cell attenuates agonist-induced AHR signaling
is by targeting ligand-bound AHR for degradation Significant advances have been made in many
through the ubiquitin/proteosome pathway^^^' ^^^. areas of AHR biology, especially in understanding
Induction of Cytochrome P450 Enzymes 339

the AHR signal transduction mechanism in respond to a wide range of chemicals. The chal-
response to xenobiotics. However, the lack of a lenge for the future will be to understand how the
three-dimensional structure for the receptor has NRs participate in a complex network to regulate
hindered the investigation of mechanisms underly- CYP gene expression and to mediate the physio-
ing species-specific responses to certain ligands, logical response to xenobiotics.
such as TCDD. Also, it is not understood how lig-
and binding to the AHR alters its conformation to
induce nuclear translocation. Determination of the
Acknowledgments
crystal structure of the AHR will greatly facilitate
the investigation of these and other aspects of AHR
We thank Scott Auerbach, Curt Omiecinski,
research. In spite of the questions remaining, the
Anna Shen, and Janardan Reddy for their critical
AHR signaling pathway has been a useful model to
review of this chapter.
provide a broad understanding of the biological
roles of PAS proteins. Yet, how the R\S domain
mediates protein-protein interactions is still not
fully understood. More in depth examination of the References
interactions between AHR and ARNT in the future
should prove helpful in the identification and char- 1. Mangelsdorf, D.J., C. Thummel, M. Beato,
acterization of R\S domain function. Finally, P. Herrlich, G. Schutz, K. Umesono et al (1995). The
although we know the AHR plays an important nuclear receptor superfamily: The second decade.
physiological role in development, the mechanism Cell 83, 835-839.
by which the AHR mediates these processes is not 2. Waxman, D.J. (1999). P450 gene induction by struc-
clear. For example, we do not know if AHR signal- turally diverse xenochemicals: Central role of nuclear
ing during development is similar or different from receptors CAR, PXR, and PPAR. Arch. Biochem.
the pathway by which AHR regulates xenobiotic Biophys. 369, 11-23.
metabolism. The ultimate identification of an 3. Poland, A. and J.C. Knutson (1982). 2,3,7,8-
endogenous AHR ligand will shed light on the tetrachlorodibenzo-p-dioxin and related halogenated
aromatic hydrocarbons: Examination of the mecha-
physiological role of the AHR.
nism of toxicity. Annu. Rev. Pharmacol. Toxicol. 22,
517-554.
4. Whitlock, J.R, Jr., S.T Okino, L. Dong, H.R Ko,
6. Conclusions R. Clarke-Katzenberg, Q. Ma et al. (1996).
Cytochromes P450 5: Induction of cytochrome
Over the last decade, great strides have been P4501A1: A model for analyzing mammalian gene
made in understanding the roles that the nuclear transcription. FASEB J. 10, 809-818.
receptors PXR, CAR, PEARa, and AHR play in the 5. Nambu, J.R., J.O. Lewis, K.A. Wharton, Jr., and
induction of CYP genes. The ability of xenobiotics S.T. Crews (1991). The Drosophila single-minded
gene encodes a helix-loop-helix protein that acts
to bind and activate NRs to induce the expression of
as a master regulator of CNS midline development.
the CYP enzymes involved in their metabolism Cell 61, 1157-1167.
provides a mechanism by which an organism can 6. Gu, Y.-Z., J. Hogenesch, and C. Bradfield (2000).
mount an adaptive response to its changing chemi- The PAS superfamily: Sensors of environmental
cal environment. The identification of endogenous and developmental signals. Annu. Rev. Pharmacol.
ligands for some NRs indicates that these receptors Tox/co/. 40,519-561.
play important roles in regulating CYP levels dur- 7. Glass, C.K. and M.G. Rosenfeld (2000). The coregu-
ing physiological processes as well. It has become lator exchange in transcriptional functions of nuclear
clear that the expression of many CYP genes is receptors. Genes Dev. 14, 121-141.
dependent on more than one NR. Recent studies 8. McKenna, N.J. and B.W. O'Malley (2002).
have demonstrated that NRs oflen share xenobiotic Combinatorial control of gene expression by nuclear
receptors and coregulators. Cell 108, 465^74.
ligands, response elements, and target CYP genes.
9. Heery, D.M., E. Kalkhoven, S. Hoare, and
The existence of multiple xenobiotic receptors with M.G. Parker (1997). A signature motif in transcrip-
broad and sometimes overlapping functions likely tional co-activators mediates binding to nuclear
increases the ability of an organism to detect and receptors. Nature 387, 733-736.
340 Susanne N. Williams ef a/.

10. Goodwin, B., M.R. Redinbo, and S.A. Kliewer 21. Blumberg, B., W Sabbagh, Jr., H. Juguilon,
(2002). Regulation of cyp3a gene transcription by J. Bolado, Jr., C. M van Meter, E.S. Ong et al.
the pregnane x receptor. Annu. Rev. Pharmacol. (1998). SXR, a novel steroid and xenobiotic-sensing
Toxicol. 42, 1-23. nuclear receptor. Genes Dev. 12, 3195-3205.
11. Michalets, E.L. (1998). Update: Clinically signifi- 22. Jones, S.A., L.B. Moore, J.L. Shenk, G.B. Wisely,
cant cytochrome P-450 drug interactions. Pharma- G.A. Hamilton, D.D. McKee et al. (2000). The
cotherapy IS, 84-112. pregnane X receptor: A promiscuous xenobiotic
12. Lu, A.Y., A. Somogyi, S. West, R. Kuntzman, and receptor that has diverged during evolution. Mol.
A.H. Conney (1972). Pregnenolone-16-carbonitrile: Endocrinol. 14, 27-39.
A new type of inducer of drug-metabolizing 23. Moore, L.B., J.M. Maglich, D.D. McKee, B. Wisely,
enzymes. Arch. Biochem. Biophys. 152, A51-A62. T.M. Willson, S.A. Kliewer et al. (2002). Pregnane
13. Elshourbagy, N.A. and RS. Guzelian (1980). X receptor (PXR), constitutive androstane receptor
Separation, purification, and characterization of a (CAR), and benzoate X receptor (BXR) define three
novel form of hepatic cytochrome P-450 from rats pharmacologically distinct classes of nuclear recep-
treated with pregnenolone-16 alpha-carbonitrile. tors. Mol. Endocrinol. 16, 977-986.
J. Biol. Chem. 255, 1279-1285. 24. Watkins, R.E., G.B. Wisely, L.B. Moore,
14. Hardwick, J.P, F.J. Gonzalez, and C.B. Kasper J.L. Collins, M.H. Lambert, S.P Williams et al
(1983). Cloning of DNA complementary to (2001). The human nuclear xenobiotic receptor
cytochrome P-450 induced by pregnenolone-16 PXR: Structural determinants of directed promiscu-
alpha-carbonitrile. Characterization of its mRNA, ity. Science 292, 2329-2333.
gene, and induction response. J. Biol. Chem. 258, 25. Xie, W, J.L. Barwick, M. Downes, B. Blumberg,
10182-10186. CM. Simon, M.C. Nelson et al. (2000). Humanized
15. Schuetz, E.G., S.A. Wrighton, J.L. Barwick, and xenobiotic response in mice expressing nuclear
P.S. Guzelian (1984). Induction of cytochrome receptor SXR. Nature 406, 435^39.
P-450 by glucocorticoids in rat liver. I. Evidence 26. Goodwin, B., E. Hodgson, and C. Liddle (1999).
that glucocorticoids and pregnenolone 16 alpha- The orphan human pregnane X receptor medi-
carbonitrile regulate de novo synthesis of a common ates the transcriptional activation of CYP3A4 by
form of cytochrome P-450 in cultures of adult rat rifampicin through a distal enhancer module. Mol.
hepatocytes and in the liver in vivo. J. Biol. Chem. Pharmacol. 56, 1329-1339.
259, 1999-2006. 27. Xie, W, J.L. Barwick, CM. Simon, A.M. Pierce,
16. Schuetz, E.G. and PS. GuzeHan (1984). Induction S. Safe, B. Blumberg et al. (2000). Reciprocal
of cytochrome P-450 by glucocorticoids in rat liver. activation of xenobiotic response genes by nuclear
II. Evidence that glucocorticoids regulate induction receptors SXR/PXR and CAR. Genes Dev 14,
of cytochrome P-450 by a nonclassical receptor 3014-3023.
mechanism. J. Biol. Chem. 259, 2007-2012. 28. Smirlis, D, R. Muangmoonchai, M. Edwards,
17. Quattrochi, L.C., A.S. Mills, J.L. Barwick, I.R. Phillips, and E.A. Shephard (2001). Orphan
C.B. Yockey, and PS. Guzelian (1995). A novel receptor promiscuity in the induction of cyto-
cis-acting element in a liver cytochrome P450 3A chromes P450 by xenobiotics J. Biol. Chem. 276,
gene confers synergistic induction by glucocorti- 12822-12826.
coids plus antiglucocorticoids. J. Biol. Chem. 270, 29. Goodwin, B., L.B. Moore, CM. Stoltz,
28917-28923. D.D. McKee, and S.A. Kliewer (2001). Regulation
18. Huss, J.M., S.I. Wang, A. Astrom, P McQuiddy, and of the human CYP2B6 gene by the nuclear
C.B. Kasper (1996). Dexamethasone responsiveness pregnane X receptor. Mol. Pharmacol. 60,427^31.
of a major glucocorticoid-inducible CYP3A gene is 30. Wei, P, J. Zhang, D.H. Dowhan, Y Han, and
mediated by elements unrelated to a glucocorticoid D.D. Moore (2002). Specific and overlapping func-
receptor binding motif Proc. Natl. Acad. Sci. USA tions of the nuclear hormone receptors CAR and
93, 4666^670. PXR in xenobiotic response. Pharmacogenomics
19. Kliewer, S.A., J.T. Moore, L. Wade, J.L. Staudinger, J. 2, 117-126.
M.A. Watson, S.A. Jones et al. (1998). An orphan 31. Sueyoshi, T, T. Kawamoto, I Zelko, P. Honkakoski,
nuclear receptor activated by pregnanes defines a and M. Negishi (1999). The repressed nuclear
novel steroid signaling pathway. Cell 92, 73-82. receptor CAR responds to phenobarbital in activat-
20. Bertilsson, G., J. Heidrich, K. Svensson, M. Asman, ing the human CYP2B6 gene. J. Biol. Chem. 274,
L. Jendeberg, M. Sydow-Backman et al. (1998). 6043-6046.
Identification of a human nuclear receptor defines a 32. Moore, L.B., D.J. Parks, S.A. Jones, R.K. Bledsoe,
new signaling pathway for CYP3A induction. Proc. T.G. Consler, J.B. Stimmel et al. (2000). Orphan
Natl. Acad. Sci. USA 95, 12208-12213. nuclear receptors constitutive androstane receptor
Induction of Cytochrome P450 Enzymes 341

and pregnane X receptor share xenobiotic and 44. Conney, A.H. (1967). Pharmacological implications
steroid ligands. ^ 5/0/. Chem. 215, 15122-15127. of microsomal enzyme induction. Pharmacol. Rev.
33. Pascussi, J.M., S. Gerbal-Chaloin, L. Drocourt, 19,317-366.
P. Maurel, and M.J. Vilarem (2003). The expression 45. Sueyoshi, T. and M. Negishi (2001). Phenobarbital
of CYP2B6, CYP2C9 and CYP3A4 genes: A tangle response elements of cytochrome P450 genes and
of networks of nuclear and steroid receptors. nuclear receptors. Annu. Rev. Pharmacol. Toxicol.
Biochim. Biophys. Acta. 1619, 243-253. 41, 123-143.
34. Huss, J.M. and C.B. Kasper (2000). Two-stage 46. Maglich, J.M., C M Stoltz, B. Goodwin,
glucocorticoid induction of CYP3A23 through both D. Hawkins-Brown, J.T. Moore, and S.A. Kliewer
the glucocorticoid and pregnane X receptors. Mol. (2002). Nuclear pregnane x receptor and constitu-
Pharmacol 58, 48-57. tive androstane receptor regulate overlapping but
35. Pascussi, J.M., L. Drocourt, J.M. Fabre, P. Maurel, distinct sets of genes involved in xenobiotic
and M.J. Vilarem (2000). Dexamethasone induces detoxification. Mol. Pharmacol. 62, 638-646.
pregnane X receptor and retinoid X receptor-alpha 47. Adesnik, M., S. Bar-Nun, F. Maschio, M. Zunich,
expression in human hepatocytes: Synergistic A. Lippman, and E. Bard (1981). Mechanism of
increase of CYP3A4 induction by pregnane X induction of cytochrome P-450 by phenobarbital.
receptor activators. Mol. Pharmacol. 58, 361-372. J. Biol. Chem. 256, 10340-10345.
36. Pascussi, J.M., M. Busson-Le Coniat, P. Maurel, and 48. Ramsden, R., K.M. Sommer, and C.J. Omiecinski
M.J. Vilarem (2003). Transcriptional analysis of (1993). Phenobarbital induction and tissue-specific
the orphan nuclear receptor constitutive androstane expression of the rat CYP2B2 gene in transgenic
receptor (NR1I3) gene promoter: Identification mice. J. Biol. Chem. 268, 21722-21726.
of a distal glucocorticoid response element, Mol. 49. Trottier, E., A. Belzil, C Stoltz, and A. Anderson
Endocrinol. 17, 42-55. (1995). Localization of a phenobarbital-responsive
37. Huss, J.M. and C.B. Kasper (1998). Nuclear recep- element (PBRE) in the 5'-flanking region of the rat
tor involvement in the regulation of rat cytochrome CYP2B2 gene. Gene 158, 263-268.
P450 3A23 expression. J. Biol. Chem. 113, 50. Stoltz, C , M.H. Vachon, E. Trottier, S. Dubois,
16155-16162. Y. Paquet, and A. Anderson (1998). The CYP2B2
38. Tirona, R.G., W. Lee, B.F. Leake, L.B. Lan, phenobarbital response unit contains an accessory
C.B. Cline, V Lamba et al. (2003). The orphan factor element and a putative glucocorticoid
nuclear receptor HNF4alpha determines PXR- and response element essential for conferring maximal
CAR-mediated xenobiotic induction of CYP3A4. phenobarbital responsiveness. J. Biol. Chem. 273,
Nat. Med. 9, 220-224. 8528-8536.
39. Li, J., G. Ning, and S.A. Duncan (2000). Mammalian 51. Honkakoski, P and M. Negishi (1997). Characteri-
hepatocyte differentiation requires the transcription zation of a phenobarbital-responsive enhancer mod-
factor HNF-4alpha. Genes Dev. 14, 464-474. ule in mouse P450 Cyp2bl0 gene. J. Biol. Chem.
40. Staudinger, J.L., B. Goodwin, S.A. Jones, 272, 14943-14949.
D. Hawkins-Brown, K.I. MacKenzie, A. LaTour 52. Honkakoski, P., R. Moore, K.A. Washburn, and
et al. (2001). The nuclear receptor PXR is a litho- M. Negishi (1998). Activation by diverse xenochemi-
cholic acid sensor that protects against liver toxicity. cals of the 51-base pair phenobarbital-responsive
Proc. Natl. Acad Sci. USA 98, 3369-3374. enhancer module in the CYP2B10 gene. Mol.
41. Li, Y C , D.R Wang, and J.Y. Chiang (1990). Pharmacol. 53,591-601.
Regulation of cholesterol 7 alpha-hydroxylase in the 53. Honkakoski, P., I. Zelko, T. Sueyoshi, and
liver. Cloning, sequencing, and regulation of choles- M. Negishi (1998). The nuclear orphan receptor
terol 7 alpha-hydroxylase mRNA. J. Biol. Chem. CAR-retinoid X receptor heterodimer activates the
265, 12012-12019. phenobarbital-responsive enhancer module of the
42. Xie, W., A. Radominska-Pandya, Y Shi, CYP2B gene. Mol. Cell. Biol. 18, 5652-5658.
CM. Simon, M.C. Nelson, E.S. Ong et al. (2001). 54. Baes, M., T. Guhck, H.S. Choi, M.G. Martinoli,
An essential role for nuclear receptors SXR/PXR in D. Simha, and D.D. Moore (1994). A new orphan
detoxification of cholestatic bile acids. Proc. Natl. member of the nuclear hormone receptor superfam-
Acad Sci. USA 98, 3375-3380. ily that interacts with a subset of retinoic acid
43. Orrenius, S., J.L. Ericsson, and L. Emster (1965). response elements. Mol. Cell. Biol. 14, 1544-1551.
Phenobarbital-induced synthesis of the microsomal 55. Choi, H.S., M. Chung, I. TzameU, D. Simha,
drug-metabolizing enzyme system and its relation- YK. Lee, W Seol et al. (1997). Differential
ship to the proliferation of endoplasmic membranes. transactivation by two isoforms of the orphan
A morphological and biochemical study. J. Cell. nuclear hormone receptor CAR. J. Biol. Chem.
Biol. 25, 627-639. 272,23565-23571.
342 Susanne N. Williams et al.

56. Kawamoto, T., T. Sueyoshi, I. Zelko, R. Moore, receptor (CAR). Proc. Natl. Acad. Sci. USA 100,
K. Washburn, and M. Negishi (1999). Phenobarbital- 4156^161.
responsive nuclear translocation of the receptor CAR 68. Reddy, J.K. and T. Hashimoto (2001). Peroxisomal
in induction of the CYP2B gene. Mol Cell. Biol. 19, beta-oxidation and peroxisome proliferator-
6318-6322. activated receptor alpha: An adaptive metabolic
57. Zelko, I., T. Sueyoshi, T. Kawamoto, R. Moore, and system. Annu. Rev. Nutr 21, 193-230.
M. Negishi (2001). The peptide near the C terminus 69. Johnson, E.E, M.H. Hsu, U. Savas, and K.J. Griffin
regulates receptor CAR nuclear translocation (2002). Regulation of P450 4A expression by per-
induced by xenochemicals in mouse liver. Mol. Cell. oxisome proliferator activated receptors. Toxicology
Biol. 21, 2838-2846. 181-182, 203-206.
58. Kawamoto, T., S. Kakizaki, K. Yoshinari, and 70. Owen, O.E., G.A. Reichard Jr., M.S. Patel, and
M. Negishi (2000). Estrogen activation of the G. Boden (1979). Energy metabolism in feasting
nuclear orphan receptor CAR (constitutive active and fasting. Adv. Exp. Med. Biol. I l l , 169-188.
receptor) in induction of the mouse Cyp2bl0 gene. 71. Reddy, J.K. andN.D. Lalwai (1983). Carcinogenesis
Mol. Endocrinol. 14, 1897-1905. by hepatic peroxisome proliferators: Evaluation
59. Forman, B.M., I. Tzameli, H.S. Choi, J. Chen, of the risk of hypolipidemic drugs and industrial
D. Simha, W. Seol et al. (1998). Androstane metabo- plasticizers to humans. Crit. Rev. Toxicol. 12, 1-58.
lites bind to and deactivate the nuclear receptor 72. Issemann, 1. and S. Green (1990). Activation of a
CAR-beta. Nature 395, 612-615. member of the steroid hormone receptor superfamily
60. Tzameli, I., R Pissios, E.G. Schuetz, and by peroxisome proliferators. Nature 347, 645-650.
D.D. Moore (2000). The xenobiotic compound 1,4- 73. GottHcher, M., E. Widmark, Q. Li, and
bis[2-(3,5-dichloropyridyloxy)]benzene is an ago- J.-A. Gustafsson (1992). Fatty acids activate a
nist ligand for the nuclear receptor CAR. Mol. Cell. chimera of the clofibric acid-activated receptor and
Biol. 20, 295\-295S. the glucocorticoid receptor. Proc. Natl. Acad. Sci.
61. Maglich, J.M., D.J. Parks, L.B. Moore, J.L. Collins, USA 89, 4653-^657.
B. Goodwin, A.N. Billin et al. (2003). Identification 74. Kliewer, S.A., B.M. Forman, B. Blumberg,
of a novel human constitutive androstane receptor E.S. Ong, U. Borgmeyer, D.J. Mangelsdorf et al.
(CAR) agonist and its use in the identification (1994). Differential expression and activation of a
of CAR target genes. J. Biol. Chem. 11%, family of murine peroxisome proliferator-activated
17277-17283. receptors. Proc. Natl. Acad Sci. USA 91, 7355-7359.
62. Dussault, I., M. Lin, K. HolHster, M Fan, J. Termini, 75. Capdevila, J.H., R.C. Harris, and J.R. Falck (2002).
M.A. Sherman et al. (2002). A structural model of Microsomal cytochrome P450 and eicosanoid
the constitutive androstane receptor defines novel metabolism. Cell. Mol. Life Sci. 59, 780-789.
interactions that mediate ligand-independent activ- 76. Willson, T.M., M.H. Lambert, and S.A. Kliewer
ity. Mol. Cell. Biol. 22, 5270-5280. (2001). Peroxisome proliferator-activated receptor
63. Min, G., J.K. Kemper, and B. Kemper (2002). gamma and metabolic disease. Annu. Rev. Biochem.
Glucocorticoid receptor interacting protein-1 70,341-367.
(GRIPI) mediates ligand-independent nuclear 77. Shi, Y, M. Hon, and R.M. Evans (2002). The per-
translocation and activation of constitutive andro- oxisome proliferator-activated receptor delta, an
stane receptor (CAR) in vivo. J. Biol. Chem. Ill, integrator of transcriptional repression and nuclear
26356-26363. receptor signaling. Proc. Natl. Acad. Sci. USA 99,
64. Wei, P, Zhang, J. Egan-Hafley, M. Liang, S. and 2613-2618.
D.D. Moore (2000). The nuclear receptor CAR 78. Hess, R., W Staubli, and W Riess (1965). Nature
mediates specific xenobiotic induction of drug of the hepatomegalic effect produced by ethyl-
metabolism. Nature 407, 920-923. chlorophenoxy-isobutyrate in the rat. Nature 208,
65. Ueda, A., H.K. Hamadeh, H.K Webb, Y. Yamamoto, 856-858.
T. Sueyoshi, C.A. Afshari et al. (2002). Diverse 79. Thorp, J.M. and WS Waring (1962). Modification
roles of the nuclear orphan receptor CAR in regu- and distribution of lipids by ethylchlorophenoxy-
lating hepatic genes in response to Phenobarbital. isobutyrate. Nature 194, 948-949.
Mol. Pharmacol. 61, 1-6. 80. Corton, J.C, S.P Anderson, and A. Stauber (2000).
66. Zhang, J., W Huang, S.S. Chua, P Wei, and Central role of peroxisome proliferator-activated
D.D. Moore (2002). Modulation of acetaminophen- receptors in the actions of peroxisome proliferators.
induced hepatotoxicity by the xenobiotic receptor Annu. Rev. Pharmacol. Toxicol. 40, 491-518.
CAR. Science 298, All-AIA. 81. Issemann, I. and S. Green (1990). Activation of
67. Huang, W, J. Zhang, S.S. Chua, M. Qatanani, a member of the steroid hormone receptor super-
Y Han, R. Granata et al. (2003). Induction of family by peroxisome proliferators. Nature 347,
bilirubin clearance by the constitutive androstane 645-650.
Induction of Cytochrome P450 Enzymes 343

82. Fan, C.Y., J. Pan, N. Usuda, A.V. Yeldandi, proliferator-activated receptor alpha-interacting
M.S. Rao, and J.K. Reddy (1998). Steatohepatitis, cofactor complex in rat liver and characterization
spontaneous peroxisome proliferation and liver of PRIC285 as a coactivator. Proc. Natl. Acad. Sci.
tumors in mice lacking peroxisomal fatty acyl-CoA USA 99, 11836-11841.
oxidase. Implications for peroxisome proliferator- 93. Xu, H.E., M.H. Lambert, VG. Montana,
activated receptor alpha natural ligand metabolism. K.D. Plunket, L.B. Moore, J.L. Collins et al.
J. Biol. Chem. 273, 15639-15645. (2001). Structural determinants of ligand binding
83. Kliewer, S.A., S.S. Sundseth, S.A. Jones, selectivity between the peroxisome proliferator-
RJ. Brown, G.B. Wisely, C.S. Koble et al (1997). activated receptors. Proc. Natl. Acad. Sci. USA 98,
Fatty acids and eicosanoids regulate gene expres- 13919-13924.
sion through direct interactions with peroxisome 94. Xu, H.E., TN. Stanley, VG. Montana,
proliferator-activated receptors alpha and gamma. M.H. Lambert, B.G. Shearer, J.E. Cobb et al.
Proc. Natl. Acad. Sci. USA 94, 4318-4323. (2002). Structural basis for antagonist-mediated
84. Forman, B.M., J. Chen, and R.M. Evans recruitment of nuclear co-repressors by
(1997). Hypolipidemic drugs, polyunsaturated fatty PPARalpha. Nature 415, 813-817.
acids, and eicosanoids are ligands for peroxisome 95. Peters, J.M., N. Hennuyer, B. Staels, J.C. Fruchart,
proliferator-activated receptors alpha and delta, C. Fievet, F.J. Gonzalez et al. (1997). Alterations in
Proc. Natl. Acad. Sci. USA 94, 4312-4317. lipoprotein metabolism in peroxisome proliferator-
85. Devchand, RR., H. Keller, J.M. Peters, M. Vazquez, activated receptor alpha-deficient mice. J. Biol.
RJ. Gonzalez, and W. Wahli (1996). The C/iem. 272, 27307-27312.
PPARalpha-leukotriene B4 pathway to inflamma- 96. Reddy, J.K., D.L. Azarnoff; and C.E. Hignite
tion control. Nature 384, 3 9 ^ 3 . (1980). Hypolipidaemic hepatic peroxisome
86. Marcus, S.L., K.S. Miyata, B. Zhang, S. Subramani, proliferators form a novel class of chemical
R.A. Rachubinski, and J.R Capone (1993). Diverse carcinogens. Nature 283, 397-398.
peroxisome proliferator-activated receptors bind to 97. Palmer, C.N., M.H. Hsu, K.J. Griffin, J.L Raucy,
the peroxisome proliferator-responsive elements of and E.F. Johnson (1998). Peroxisome proliferator
the rat hydratase/dehydrogenase and fatty acyl-CoA activated receptor-alpha expression in human liver.
oxidase genes but differentially induce expression. Mol. Pharmacol. 53, 14-22.
Proc. Natl. Acad Sci. USA 90, 5723-5727. 98. Gervois, R, I.R Torra, G. Chinetti, T. Grotzinger,
87. Osumi, T., N. Ishii, S. Miyazawa, andT. Hashimoto G. Dubois, J.C. Fruchart et al. (1999). A truncated
(1987). Isolation and structural characterization of human peroxisome proliferator-activated receptor
the rat acyl-CoA oxidase gene. J. Biol. Chem. 262, alpha splice variant with dominant negative activ-
8138-8143. ity. Mol. Endocrinol. 13, 1535-1549.
88. Issemann, I., R.A. Prince, J.D. Tugwood, and 99. Yu, S., W.Q. Cao, R Kashireddy, K. Meyer, Y. Jia,
S. Green (1993). The peroxisome proliferator- D.E. Hughes et al. (2001). Human peroxisome
activated receptor: retinoid X receptor heterodimer proliferator-activated receptor alpha (PPARalpha)
is activated by fatty acids and fibrate hypolipi- supports the induction of peroxisome proliferation
daemic drugs. J. Mol. Endocrinol. 11, 3 7 ^ 7 . in PPARalpha-deficient mouse liver. J. Biol. Chem.
89. Kliewer, S.A., K. Umesono, D.J. Noonan, 276,42485-42491.
R.A. Heyman, and R.M. Evans (1992). Conver- 100. Cherkaoui-Malki, M., K. Meyer, W.Q. Cao,
gence of 9-cis retinoic acid and peroxisome prolif- N. Latruffe, A.V Yeldandi, M.S. Rao et al (2001).
erator signalling pathways through heterodimer Identification of novel peroxisome proliferator-
formation of their receptors. Nature 358,11\-11A. activated receptor alpha (PPARalpha) target genes
90. Palmer, C.N., M.H. Hsu, H.J. Griffin, and in mouse liver using cDNA microarray analysis.
E.F. Johnson (1995). Novel sequence determinants GeneExpr 9,291-304.
in peroxisome proliferator signaling. J. Biol. Chem. 101. Lee, S.S., T. Pineau, J. Drago, E.J. Lee,
270, 16114-16121. J.W. Owens, D.L. Kroetz et al (1995). Targeted
91. Juge-Aubry, C , A. Pemin, T. Favez, A.G. Burger, disruption of the alpha isoform of the peroxisome
W. Wahli, C.A. Meier et al. (1997). DNA binding proliferator-activated receptor gene in mice
properties of peroxisome proliferator-activated results in abolishment of the pleiotropic effects of
receptor subt3^es on various natural peroxisome peroxisome proliferators. Mol Cell. Biol 15,
proliferator response elements. Importance of 3012-3022.
the 5'-flanking region. J. Biol. Chem. Ill, 102. Leone, T C , C.J. Weinheimer, and D.R Kelly
25252-25259. (1999). A critical role for the peroxisome pro-
92. Surapureddi, S., S. Yu, H. Bu, T. Hashimoto, liferator-activated receptor alpha (PPARalpha)
A.V Yeldandi, R Kashireddy et al. (2002). Identi- in the cellular fasting response: The PPARalpha-
fication of a transcriptionally active peroxisome null mouse as a model of fatty acid oxidation
344 Susanne N. Williams et al.

disorders. Proc. Natl. Acad. Sci. USA 96, 115. Poland, A. and E. Glover (1979). An estimate of the
7473-7478. maximum in vivo covalent binding of 2,3,7,8-tetra-
103. Conney, A.H., E.C. Miller, and J.A. Miller (1956). chlorodibenzo-/7-dioxin to rat liver protein, riboso-
The metabolism of methylated aminoazo dyes. mal RNA, and DNA. Cancer Res. 39, 3341-3344.
V Evidence for induction of enzyme synthesis 116. Poland, A., WF. Greenlee, and A.S. Kende (1979).
in the rat by 3-methylcholanthrene. Cancer Res. Studies on the mechanism of action of the chlori-
16, 450-459. nated dibenzo-p-dioxins and related compounds.
104. Conney, A.H., J.R. Gillette, J.K. Inscoe, Ann. N.Y.Acad. Sci. 320, 214-230.
E.R. Trams, and H.S. Posner (1959). Induced syn- 117. Tukey, R.H., R.R. Hannah, M. Negishi,
thesis of liver microsomal enzymes which metabo- D.W Nebert, and H.J. Eisen (1982). The Ah locus:
lize foreign compounds. Science 130, 1478-1479. Correlation of intranuclear appearance of inducer-
105. Nebert, D.W. and H.V Gelboin (1969). The in vivo receptor complex with induction of cytochrome
and in vitro induction of aryl hydrocarbon hydrox- Pj-450 mRNA. Cell 31, 275-284.
ylase in mammalian cells of different species, 118. Poland, A., E. Glover, EH. Ebetino, and
tissues, strains, and development and hormonal A.S. Kende (1986). Photoafifinity labeling of the
states. Arch. Biochem. Biophys. 134, 76-89. Ah receptor. J. Biol. Chem. 261, 6352-6365.
106. Gielen, J.E., KM. Goujon, and D.W. Nebert (1972). 119. Bradfield, C.A., E. Glover, and A. Poland (1991).
Genetic regulation of aryl hydrocarbon hydroxy- Purification and N-terminal amino acid sequence
lase induction. J. Biol. Chem. 247, 1125-1137. of the Ah receptor from the C57BL/6J mouse.
107. Thomas, P.E., R.E. Kouri, and J.J. Hutton (1972). Mol. Pharmacol. 39, 13-19.
The genetics of aryl hydrocarbon hydroxylase 120. Ema, M., K. Sogawa, N. Watanabe, Y. Chujoh,
induction in mice: A single gene difference N. Matsushita, O. Gotoh et al. (1992). cDNA
between C57BL/6J and DBA/2J. Biochem. cloning and structure of mouse putative Ah recep-
Genetics 6, 157-168. tor. Biochem. Biophys. Res. Comm. 184, 246-253.
108. Green, M.C. (1973). Nomenclature of genetically 121. Burbach, K.M., A. Poland, and C.A. Bradfield
determined biochemical variants in mice, Biochem. (1992). Cloning of the Ah receptor cDNA reveals
Genet. 9, 369-374. a distinctive ligand-activated transcription factor.
109. Poland, A. and E. Glover (1974). Comparison Proc. Natl. Acad Sci. USA 89, 8185-8189.
of 2,3,7,8-tetrachlorodibenzo-p-dioxin, a potent 122. Hoffman, E.C, H. Reyes, F.F Chu, F Sander,
inducer of aryl hydrocarbon hydroxylase, with L.H. Conley B.A. Brooks et al. (1991). Cloning of
3-methylcholanthrene. Mol. Pharmacol. 10, a factor required for activity of the Ah (dioxin)
349-359. receptor. Science 252, 954-958.
110. Poland, A. and E. Glover (1973). Chlorinated 123. Reyes, H., S. Reisz-Porszasz, and O. Hankinson
dibenzo-p-dioxins: Potent inducers of delta- (1992). Identification of the Ah receptor nuclear
aminolevulinic acid synthetase and aryl hydrocar- translocator protein (Arnt) as a component of the
bon hydroxylase. II. A study of the structure-activity DNA binding form of the Ah receptor. Science
relationship. Mol. Pharmacol. 9,136-741. 256, 1193-1195.
111. Poland, A., E. Glover, and A.S. Kende (1976). 124. Dolwick, K.M., J.V Schmidt, L.A. Carver,
Stereospecific, high affinity binding of 2,3,7,8- H.I. Swanson, and C.A. Bradfield (1993). Cloning
tetrachlorodibenzo-/7-dioxin by hepatic cytosol. and expression of a human Ah receptor cDNA.
J. Biol. Chem. 251, 4936-4946. Mol. Pharmacol. 44, 911-917.
112. Nebert, D.W., EM. Goujon, and J.E. Gielen (1972). 125. Dolwick, K.M., H.I. Swanson, and C.A. Bradfield
Aryl hydrocarbon hydroxylase induction by poly- (1993). In vitro analysis of Ah receptor domains
cyclic hydrocarbons: Simple autosomal dominant involved in ligand-activated DNA recognition.
trait in the mouse. Nat. New Biol. 236, 107. Proc. Natl. Acad Sci. USA 90, 8566-8570.
113. Poland, A.P., E. Glover, J.R. Robinson, and 126. Murre, C , G. Bain, M.A. van Dijk, I. Engel,
D.W. Nebert (1974). Genetic expression of aryl B.A. Furnari, M.E. Massari et al. (1994). Structure
hydrocarbon hydroxylase activity. Induction of and function of helix-loop-helix proteins. Biochim.
monooxygenase activities and cytochrome PI-450 Biophys. Acta 1218, 129-135.
formation by 2,3,7,8-tetrachlorodibenzo-p-dioxin in 127. Fukunaga, B.N., and O. Hankinson (1996).
mice genetically "nonresponsive" to other aromatic Identification of a novel domain in the aryl
hydrocarbons. J. Biol. Chem. 249, 5599-5606. hydrocarbon receptor required for DNA binding.
114. Poland, A. and E. Glover (1977). Chlori- J. Biol. Chem. 271, 3743-3749.
nated biphenyl induction of aryl hydrocarbon 128. Perdew, G.H. (1988). Association of the Ah recep-
hydroxylase activity: A study of the structure- tor with the 90-kDa heat shock protein. J. Biol.
activity relationship. Mol. Pharmacol. 13, 924-938. Chem. 263, 13802-13805.
Induction of Cytochrome P450 Enzymes 345

129. Carver, L.A. and C.A. Bradfield (1997). Ligand (Aryl hydrocarbon) receptor. J. Biol. Chem. 274,
dependent interaction of the Ah receptor with a 13519-13524.
novel immunophilin homolog in vivo. J. Biol. 142. Antonsson, C , M.L. Whitelaw, J. McGuire,
Chem. 272, 11452-11456. J.A. Gustafsson, and L. Poellinger (1995). Distinct
130. Fukunaga, B.N., M.R. Probst, S. Reisz-Porszasz, roles of the molecular chaperone hsp90 in
and O. Hankinson (1995). Identification of func- modulating dioxin receptor function via the basic
tional domains of the aryl hydrocarbon receptor helix-loop-helix and PAS domains, Mol. Cell. Biol.
J. Biol. Chem. 270, 29270-29278. 15, 756-765.
131. Tuteja, N., F.J. Gonzalez, and D.W. Nebert (1985). 143. Carver, L.A., V Jackiw, and C.A. Bradfield (1994).
Developmental and tissue-specific differential The 90-kDa heat shock protein is essential for
regulation of the mouse dioxin-inducible PI-450 Ah receptor signaling in a yeast expression system.
and P3-450 genes. Dev Biol. 112, 177-184. J. Biol. Chem. 269, 30109-30112.
132. Murray, G.I., W.T. Melvin, W.F. Greenlee, and 144. Pongratz, I., G.G. Mason, and L. Poellinger
M.D. Burke (2001). Regulation, function, and tissue- (1992). Dual roles of the 90-kDa heat shock
specific expression of cytochrome P450 CYPIBI. protein hsp90 in modulating functional activities
Annu. Rev. Pharmacol. Toxicol. 41, 297-316. of the dioxin receptor. J. Biol. Chem. 267,
133. Poland, A. and E. Glover (1987). Variation in the 13728-13734.
molecular mass of the Ah receptor among verte- 145. Kazlauskas, A., S. Sundstrom, L. Poellinger, and
brate species and strains of rats. Biochem. Biophys. I. Pongratz (2001). The hsp90 chaperone complex
Res. Commun. 146, 1439-1449. regulates intracellular localization of the dioxin
134. Bank, PA., E.F Yao, C.L. Phelps, PA. Harper, and receptor. Mol. Cell. Biol. 21, 2594-2607.
M.S. Denison (1992). Species-specific binding of 146. LaPres, J.J., E. Glover, E.E. Dunham,
transformed Ah receptor to a dioxin responsive tran- M.K. Bunger, and C.A. Bradfield (2000). ARA9
scriptional enhancer Eur. J. Pharmacol. 228, 85-94. modifies agonist signaling through an increase iji
135. Poland, A., D. Palen, and E. Glover (1994). cytosolic aryl hydrocarbon receptor. J. Biol. Chem.
Analysis of the four alleles of the murine aryl 275, 6153-6159.
hydrocarbon receptor. Mol. Pharmacol. 46, 147. Gasiewicz, T.A., C.J. Elferink, and E.C. Henry
915-921. (1991). Characterization of multiple forms
136. Ema, M., N. Ohe, M. Suzuki, J. Mimura, of the Ah receptor: Recognition of a dioxin-
K. Sogawa, I. Ikawa et al. (1994). Dioxin binding responsive enhancer involves heteromer forma-
activities of polymorphic forms of mouse and tion. Biochemistry 30, 2909-2916.
human aryl hydrocarbon receptors. J. Biol. Chem. 148. Wilhelmsson, A., S. Cuthill, M. Denis,
269, 27337-27343. A.C. Wikstrom, J.A. Gustafsson, and L. Poellinger
137. Denison, M.S., A. Pandini, S.R. Nagy, (1990). The specific DNA binding activity of the
E.P Baldwin, and L. Bonati (2002). Ligand bind- dioxin receptor is modulated by the 90 kd heat
ing and activation of the Ah receptor. Chem. Biol. shock protein. EMBOJ. 9, 69-76.
Interact. 141, 3-24. 149. Probst, M.R., S. Reisz-Porszasz, R.V Agbunag,
138. Carver, L.A. and C.A. Bradfield (1997). Ligand- M.S. Ong, and O. Hankinson (1993). Role of the
dependent interaction of the aryl hydrocarbon aryl hydrocarbon receptor nuclear translocator
receptor with a novel immunophilin homolog in protein in aryl hydrocarbon (dioxin) receptor
vivo. J. Biol. Chem. 272, 11452-11456. action. Mol. Pharmacol. 44, 511-518.
139. Ma, Q. and J.R Whitlock, Jr. (1997). A novel cyto- 150. Elferink, C.J. and J.R Whitlock, Jr. (1994). Dioxin-
plasmic protein that interacts with the Ah receptor, dependent, DNA sequence-specific binding of a
contains tetratricopeptide repeat motifs, and multiprotein complex containing the Ah receptor.
augments the transcriptional response to 2,3,7,8- Receptor 4, 157-173.
tetrachlorodibenzo-p-dioxin. J. Biol. Chem. 272, 151. Whitlock, J.R, Jr. (1999). Induction of cytochrome
8878-8884. P4501A1. Annu. Rev Pharmacol. Toxicol. 39,
140. Meyer, B.K., M.G. Pray-Grant, J.R Vanden Heuvel, 103-125.
and G.H. Perdew (1998). Hepatitis B virus X- 152. Jones, RB., L.K. Durrin, D.R. Galeazzi, and
associated protein 2 is a subunit of the unliganded J.R Whitlock, Jr. (1986). Control of cytochrome
aryl hydrocarbon receptor core complex and PI-450 gene expression: Analysis of a dioxin-
exhibits transcriptional enhancer activity. Mol. responsive enhancer system. Proc. Natl. Acad. Sci.
Cell. Biol. 18, 978-988. USA 83, 2802-2806.
141. Kazlauskas, A., L. Poellinger, and I. Pongratz 153. Fujisawa-Sehara, A., K. Sogawa, M. Yamane, and
(1999). Evidence that the co-chaperone p23 Y. Fujii-Kuriyama (1987). Characterization of
regulates ligand responsiveness of the dioxin xenobiotic responsive elements upstream from
346 Susanne N. Williams et al.

the drug-metabolizing cytochrome P-450c gene: 164. Huang, L.E. and H.F Bunn (2003). Hypoxia-
A similarity to glucocorticoid regulatory elements. inducible factor and its biomedical relevance.
Nucleic Acids Res. 15,4179^ 191. J. Biol. Chem. 278, 19575-19578.
154. Durrin, L.K. and J.P. Whitlock, Jr. (1987). In situ 165. Chan, W.K., G. Yao, YZ. Gu, and C.A. Bradfield
protein-DNA interactions at a dioxin-responsive (1999). Cross-talk between the aryl hydrocarbon
enhancer associated with the cytochrome PI-450 receptor and hypoxia inducible factor signaling
gene. Mol. Cell. Biol. 7, 3008-3011. pathways. Demonstration of competition and
155. Denison, M.S., J.M. Fisher, and J.R Whitlock, compensation. J 5/o/. Chem. IIA, 12115-12123.
Jr. (1988). The DNA recognition site for the 166. Gradin, K., J. McGuire, R.H. Wenger,
dioxin-Ah receptor complex. Nucleotide sequence I. Kvietikova, M.L. fhitelaw, R. Toftgard et al.
and functional analysis. J. Biol. Chem. 263, (1996). Functional interference between hypoxia
17221-17224. and dioxin signal transduction pathways: Competi-
156. Yao, E.F. and M.S. Denison (1992). DNA tion for recruitment of the Arnt transcription
sequences determinants for binding of transformed factor. Mol. Cell. Biol. 16, 5221-5231.
Ah receptor to a dioxin-responsive enhancer. 167. Nie, M., A.L. Blankenship, and J.R Giesy (2001).
Biochemistry 31, 5060-5067. Interactions between aryl hydrocarbon receptor
157. Swanson, H.L, W.K. Chan, and C.A. Bradfield (AhR) and hypoxia signaling pathways. Environ.
(1995). DNA binding specificities and pairing Toxicol. Pharmacol. 10, 17-27.
rules of the Ah receptor. ARNT, and SIM proteins. 168. Pollenz, R.S., N.A. Davarinos, and T.P Shearer
J. Biol. Chem. ll^i, 26292-26302. (1999). Analysis of aryl hydrocarbon receptor-
158. Nebert, D.W., A. Puga, and V Vasiliou (1993). mediated signaling during physiological hypoxia
Role of the Ah receptor and the dioxin-inducible reveals lack of competition for the aryl hydro-
[Ah] gene battery in toxicity, cancer, and signal carbon nuclear translocator transcription factor.
transduction. Ann. N. Y. Acad. Sci. 685, 624-640. Mol. Pharmacol. 56, 1127-1137.
159. Mimura, J., M. Ema, K. Sogawa, and Y. Fujii- 169. Fernandez-Salguero, P, T. Pineau, DM. Hilbert,
Kuriyama (1999). Identification of a novel mecha- T. McPhail, S.S. Lee, S. Kimura et al. (1995).
nism of regulation of Ah (dioxin) receptor Immune system impairment and hepatic fibrosis in
function. Genes Dev. 13, 20-25. mice lacking the dioxin-binding Ah receptor.
160. Karchner, S.I., D.G. Franks, W.H. Powell, and Science 268, 722-726.
M.E. Hahn (2002). Regulatory interactions among 170. Schmidt, J.V, G.H. Su, J.K. Reddy, M.C. Simon,
three members of the vertebrate aryl hydrocarbon and C.A. Bradfield (1996). Characterization of
receptor family: AHR repressor, AHRl, and a murine Ahr null allele: Involvement of the
AHR2. J. Biol. Chem. 277, 6949-6959. Ah receptor in hepatic growth and development.
161. Baba, T., J. Mimura, K. Gradin, A. Kuroiwa, Proc. Natl. Acad Sci. USA 93, 6731-6736.
T. Watanabe, Y Matsuda et al. (2001). Structure 171. Hirose, K., M. Morita, M. Ema, J. Mimura,
and expression of the Ah receptor repressor gene. H. Hamada, H. Fujii et al. (1996). cDNA cloning
J. Biol. Chem. 276, 33101-33110. and tissue-specific expression of a novel basic
162. Davarinos, N.A. and R.S. PoUenz (1999). Aryl helix-loop-helix/Pas factor (Amt2) with close
hydrocarbon receptor imported into the nucleus sequence similarity to the aryl hydrocarbon recep-
following ligand binding is rapidly degraded via tor nuclear translocator (Arnt), Mol. Cell. Biol. 16,
the cytosplasmic proteasome following nuclear 1706-1713.
export. J. Biol. Chem. 274, 28708-28715. 172. Fernandez-Salguero, PM., DM. Hilbert,
163. Roberts, B.J. and M.L. Whitelaw (1999). S. Rudikoflf, J.M. Ward, and FJ. Gonzalez (1996).
Degradation of the basic helix-loop-helix/Per- Aryl-hydrocarbon receptor-deficient mice are
ARNT-Sim homology domain dioxin receptor via resistant to 2,3,7,8-tetrachlorodibenzo-p-dioxin-
the ubiquitin/proteasome pathway. J. Biol. Chem. induced toxicity. Toxicol. Appl. Pharmacol. 140,
274,36351-36356. 173-179.
9
Hormonal Regulation of Liver
Cytochrome P450 Enzymes
David J. Waxman and Thomas K.H. Chang

1. Introduction 57 ftinctional human cytochrome P450 genes,


grouped into 17 distinct gene families^. Many of
Sex differences in hepatic drug metabolism these P450 enzymes catalyze the biosynthesis of
have been known for more than 30 years, based on physiologically important endogenous substances,
the early studies of Kato, Conney, Gillette, and such as steroid hormones, whereas others are pri-
others^~^. In rats and certain other species, the rate marily involved in metabolism of environmental
of drug metabolism is often several-fold higher chemicals and other xenobiotics, notably drugs.
in males as compared to females as revealed by From the perspective of foreign compound meta-
in vivo pharmacokinetics and as demonstrated bolism, the most important cytochrome P450
in vitro by assaying prototypic phase I c5^ochrome ("CYP") genes are those in the CYPl, CYP2, and
P450 drug substrates, such as ethylmorphine, CYP3 families. These three families encompass
benzo[a]pyrene, or hexobarbital using isolated — 15-20 different cytochrome P450 enzymes, and
liver microsome preparations (Figure 9.1). This collectively carry out essentially all of the phase I
sex-dependence of P450 metabolism is most strik- cytochrome P450 metabolic reactions in mam-
ing in the rat, where sex differences in metabolic malian liver^^. As individual liver P450 enzymes
rates can be as high as 5-fold with some drug sub- were purified and characterized, and subsequently,
strates. This finding was initially unexpected, when their genes were cloned from multiple
because measurements of total liver P450 content species, it became apparent that in certain species,
indicated that the overall P450 levels in rat liver such as the rat and mouse, a subset of these drug-
tissue are only —20% higher in males compared metabolizing liver P450s is expressed in a sex-
to females (Figure 9.1). Research carried out dur- dependent fashion and subject to endocrine
ing the 1980s resolved this discrepancy with the control^ ^ Human liver P450 enzyme levels and
finding that there are multiple liver-expressed their associated drug metabolism activities may
P450 enzymes^' ^, each of which is encoded by a also be determined, in part, by age, sex, and hor-
separate gene, and only some of which are mone status^^~^^. Studies of the underlying mech-
expressed in a sex-dependent manner. anisms governing the endocrine regulation of rat
Based on the recently published human liver P450 enzymes may thus be of general impor-
genome sequence, we now know that there are tance for our understanding of the hormonal

David J. Waxman • Division of Cell and Molecular Biology, Department of Biology, Boston University, Boston,
MA. Thomas K.H. Chang • Faculty of Pharmaceutical Sciences, The University of British Columbia,
Vancouver, BC, Canada.
Cytochrome P450: Structure, Mechanism, and Biochemistry, 3e, edited by Paul R. Ortiz de Montellano
Kluwer Academic / Plenum Publishers, New York, 2005.

347
348 David J. Waxman and Thomas K.H. Chang

12 2. Steroid Hormones as
^ ^ H MALE
o EZZ2 FEMALE Substrates for Sex-Dependent
Liver P450s
.^ fl 8
The precise physiological function of the
1 endocrine-regulated rodent liver P450s is unclear;
1 however, steroid hormones are metabolized by
liver P450 enzymes with a higher degree of
N ^

0
EM
i li 1. L
1
BP HB P450(10X)
M
regiospecificity and stereoselectivity than many
foreign compound substrates^ ^ suggesting that
these endogenous lipophiles serve as physiologi-
cal P450 substrates. The physiological require-
Figure 9.1. Sex-differences in rat hepatic microsomal ments with respect to steroid hormone
drug metabolism. Data shown are based on enzyme hydroxylation differ between the sexes, and not
assays in rat liver microsomes using the three indicated surprisingly, several steroid hydroxylase liver
xenobiotic substrates: ethylmorphine (EM)"*, benzo[a] P450s are expressed in a sex-dependent manner^ ^' ^^.
pyrene (BP)^, and hexobarbital (HB)^. Ethylmorphine Rat P450 enzymes CYP2C11 and CYP2C12 are
A^-demethylase and benzo[(3]pyrene hydroxylase prototypic examples of sex-specific liver P450
activities are expressed as nmol product formed per enzymes, and they have been a major focus of
minute per mg microsomal protein, whereas hexo- studies of the underlying endocrine factors, as
barbital hydroxylase activity is expressed as nmol
well as the cellular and molecular regulatory
product formed per 30 min per g liver. Also shown is
hepatic microsomal total cytochrome P450 content, mechanisms that govern sex-specific liver gene
which is expressed as nmol per mg microsomal pro- expression. CYP2C11 is the major male-specific
tein (values multiplied by 10)^. The data are shown as androgen 16a- and 2a-hydroxylase in adult rat
mean ± SD for 4 or 5 rats, except for ethylmorphine liver, and is induced at puberty in males but not
A^-demethylase and total P450 which are based on a pool females •^' ^^ under the influence of neonatal
of 6 livers. androgenic imprinting (programming)^ ^ By
contrast, the steroid sulfate 15p-hydroxylase
CYP2C12 is expressed in a female-specific
manner in adult rats^'' ^^.
regulation of liver-expressed genes in both rodent
models and in man. Endocrine-regulated steroid
hydroxylase P450s contribute in an important way
to foreign compound metabolism in the liver, and
3. Developmental Regulation of
studies of their hormonal control may shed light
on the underlying basis for the influence of hor- Sex-Dependent Rat Liver
mone status on a broad range of P450-catalyzed P450S
drug metabolism and carcinogen activation
reactions. Multiple rat liver cytochrome P450 enzymes
This chapter reviews studies leading to the are expressed in a sex-dependent manner, sub-
identification of the key endocrine regulatory ject to complex developmental regulation and
factors and the underlying mechanisms through endocrine control (Table 9.1). CYP2C11, the
which these factors operate to control the expres- major male-specific androgen 2a- and 16a-
sion of liver cytochrome P450 enzymes. Primary hydroxylase of adult liver, is not expressed in
emphasis is given to studies on the hormonally immature rats but is induced dramatically at
regulated P450s expressed in rat liver, the best- puberty (beginning at 4-5 weeks of age) in male
studied model system. Also discussed are environ- but not female rat liver^^' ^^. A similar develop-
mental and pathophysiological factors that can mental profile characterizes three other male-
perturb hormonal status and the impact of these specific rat liver cytochromes P450, CYP2A2
factors on the expression of sex-dependent (refs [27], [36]), CYP2C13 (refs [28], [37]), and
cytochromes P450. CYP3A18 (refs [25], [38]). By contrast, another
Hormonal Regulation of Liver Cytochrome P450 Enzymes 349

Table 9 . 1 . Hormonal Regulation of Sex-Dependent Rat Liver P450 Enzymes

Hormonal regulation^

Testosterone
hydroxylase Androgenic
CYP enzyme^ activities^ imprinting'^ Thyroid hormone^

/. Male-specific
2A2 15a ++ +/-
2C11 2a, 16a ++ +/-
2C13 6^, 15a ++ ND
3A2 6P,2(3 ++ --
3A18 6(3,2(3, 15P, 16a ND ND
4A2 (see footnote g) ND -
//. Female-specific
2C12 15P^ -- 4-/-

///. Female-predominanf
2A1 7a ND -
2C7 16a ND ++
3A9 6P ND ND
5a-reductase — ++

''P450 gene designations are based on the systematic nomenclature of ref. [23]. Table is modified from ref [24].
^The major sites of testosterone hydroxylation catalyzed by the individual P450 proteins are shown. Testosterone
metabolites specific to the P450's activity in rat liver microsomal incubations are underlined. Based on refs [8], [11],
[21], [25], [26] and references therein.
cu_|_ _)_» in(jjcates a positive effect on adult enzyme expression, while " " indicates a suppressive effect. "—" indi-
cates a lesser degree of suppression, while " + / - " indicates no major effect. ND—^not determined in a definitive
manner.
'^For further details see refs [27]-[29].
^Based on refs [30]-[34].
^Purified CYP2C13 exhibits high testosterone hydroxylase activity in a reconstituted enzyme system, but this
enzyme makes only marginal contributions to liver microsomal testosterone hydroxylation^^.
^CYP4A2 catalyzes fatty acid co-hydroxylation, but does not catalyze testosterone hydroxylation.
^15P-hydroxylation of steroid sulfates. CYP2C12 also catalyzes weak testosterone 15a- and la-hydroxylase
activities.
'Liver expression of these enzymes is readily detectable in both male and female rats, but at a 3-10-fold higher level
in females as compared to males.

adult male-specific liver P450, the steroid 6|3- and steroid 5a-reductase, which is not a
hydroxylase CYP3A2, is expressed in prepubertal cytochrome P450 enzyme but plays an important
rat liver at similar levels in both sexes, but is selec- role in steroid metaboHsm in adult female rats^^' ^^.
tively suppressed at puberty in females^ ^' ^^"^^ The Finally, CYP2A1 is a female-predominant steroid
steroid sulfate ISP-hydroxylase CYP2C12 is 7a-hydroxylase that is expressed in both sexes
expressed at a moderate level in both male and shortly afler birth, but is suppressed at puberty to a
female rats at 3 ^ weeks of age. Beginning at greater extent in male than in female rat liver^^' ^^' ^^
puberty, however, CYP2C12 levels are further (Table 9.1). Each of these sex-dependent P450
increased in females while they are fully sup- enzymes is primarily expressed in liver, although
pressed in males^^' '^^. Several other female-pre- low-level extrahepatic expression may occur in
dominant liver enzymes have also been shown to some cases"^^^^. Studies of liver P450 expression
increase at puberty in adult female rats. These during senescence have revealed a general loss of
include: CYP2C7 (refs [37], [42]), which catalyzes gender-dependent enzyme expression that reflects
retinoic acid 4-hydroxylation'^^; CYP3A9 (ref a decrease in male P450 levels and an increase in
[44]), which catalyzes steroid 6p-hydroxylation^^; expression of female-predominant P450 enzymes
350 David J. Waxman and Thomas K.H. Chang

in aging male rats^^' ^^. This appears to be related exposure is thus sufficient to "imprint" or irre-
to the age-dependent reduction in the secretion of versibly program the male rat to express these
growth hormone (GH) releasing factor and the P450 enzymes later on in adult life. These effects
changes in plasma profile of GH^^, which is a of neonatal androgen on male-specific P450
major regulator in the sex-dependent expression of enzymes are very similar to the androgenic
liver CYP enzymes (see Section 4.2). imprinting effects observed in earlier studies of
liver microsomal steroid hydroxylase activities'^'
54, 55^ several of which can be associated with
specific liver P450 forms^^
4. Hormonal Control of Liver However, neonatal testosterone given to birth-
P450 Expression castrated male rats only partially restores
CYP2C11 (refs [21], [53]) or CYP2C13 (ref [28])
4.1. Regulation by Gonadal to normal adult male levels, indicating that neona-
Hormones tal androgen alone is insufficient for full adult
expression of these male-specific P450s. Con-
Initial studies on the endocrine regulation of the sistent with this observation, the combination
liver P450 enzymes demonstrated that the gonadal of neonatal androgen treatment with adult andro-
steroids play an important role in regulating gen exposure results in complete restoration of
enzyme expression, but only indirectly. Gonadal normal adult male expression of the male-specific
steroids do not act directly on the liver to confer P450s (ref [28]). Moreover, testosterone treat-
the sex-dependent pattern of hepatic steroid and ment of adult male rats that were castrated either
drug metabolism and P450 expression, but rather, neonatally or prepubertally, can substantially
act indirectly via the hypothalamus, which regu- increase the expression of CYP2C11 (refs [29], [53],
lates the pituitary gland and its secretion of the [56], [57]) and CYP2C13 (ref [28]). However, in
polypeptide hormone, GH (see Section 4.2). contrast to the irreversible imprinting effects of
neonatal androgen treatment, the effects of adult
androgen exposure are likely to be reversible, as
4.1.1. Testosterone evidenced by the partial loss of CYP2C11 in male
4.1.1.1. Distinct effects of neonatal rats castrated at adulthood^^' ^' and by the reversal
androgen and adult androgen. Gonadal hor- of this loss by the synthetic androgen methyl-
mones play an essential role in determining the trienolone^^. Similarly, the continued presence of
expression of the major sex-specific rat liver P450 testosterone at adulthood is also required to main-
forms at adulthood. In the case of testosterone, tain normal adult expression of CYP3A2, since
there are two distinct postnatal developmental castration at 90 days of age reduces hepatic
periods of hormone production, neonatal and post- CYP3A2 mRNA levels by >80%, but this can be
pubertal, and each period makes a distinct con- restored by subsequent administration of testos-
tribution to the expression of the sex-dependent terone to the adult rat^^. Thus, while neonatal
liver P450s at adulthood. Castration of male rats at testosterone imprints the rat for expression of the
birth eliminates both periods of androgen produc- male-specific P450 enzymes beginning at
tion, and this in turn abolishes the normal adult puberty, a time when the demand for P450-
expression of each of the male-specific P450s: dependent liver steroid metabolism is increased,
CYP2A2 (ref [27]), CYP2C11 (refs [20], [21], [29], the additional presence of androgen during the
[37], [53]), CYP2C13 (ref [28]), and CYP3A2 pubertal and postpubertal periods is required to
(refs [21], [29], [53]). CYP2C13 mRNA levels^^ maintain full enzyme expression during adult
are also abolished in birth-castrated rats, indicat- life^O'^i.
ing that enzyme expression is regulated at a pre- 4.1.1.2. Testosterone suppression of
translational step. Treatment of birth-castrated female enzymes. In contrast to the positive reg-
male rats with testosterone during the neonatal ulation by testosterone of the male-specific
period leads to a partial restoration of the expres- enzymes, testosterone suppresses expression of
sion of these male-specific P450 forms at adult- the female-specific CYP2C12 as well as the
hood^^' ^^' ^^. A brief period of neonatal androgen female-predominant enzymes CYP2A1 and
Hormonal Regulation of Liver Cytochrome P450 Enzymes 351

steroid 5a-reductase. Hepatic CYP2C12 content absence of CYP2C11 in adult female rats is not
is reduced in intact, adult female rats exposed due to a negative effect of estrogen. Thus, ovari-
chronically to testosterone^^ or to the synthetic ectomy alone does not lead to CYP2C11 expression
androgen methyltrienolone^^. Similarly, treatment in female rats^^' ^^' ^^. In male rats, the suppression
of neonatally or prepubertally ovariectomized rats of CYP2C11 by estradiol may be irreversible, as
with testosterone, either neonatally or pubertally, demonstrated by the major loss of this P450 in
results in a major decrease in microsomal steroid adult male rats exposed to estradiol during the
5a-reductase activity^^' ^^. Birth castration of male neonatal period or at puberty. However, this effect
rats increases the adult levels of hepatic CYP2A1, is not a consequence of a direct action of estradiol
but testosterone administration to these animals on the liver, since estradiol does not impact on
re-masculinizes (i.e., decreases) the levels of hepatic CYP2C11 levels in hypophysectomized
this P450 (ref [62]). Androgens thus exert a sup- rats^^. Rather, the effects of estradiol on liver P450
pressive effect on liver CYP2A1 expression. expression involve the hypothalamo-pituitary
Studies of the effect of testosterone on the expres- ^^jg64,72^ ^j^^ j^Qg^ likely result from an estrogen-
sion of the female-predominant CYP2C7 are dependent increase in the interpeak baseline levels
inconclusive^^' ^^. of plasma GH^"^' ^^. This effect of estradiol may be
4.1.1.3. Mechanisms of testosterone sufficient to alter the sex-specific effects of the
regulation. The mechanisms by which neonatal GH secretory pattern since, as discussed in greater
testosterone imprints the expression of liver P450 detail below, recognition of a "masculine" GH
enzymes during adulthood are only partially pulse by hepatocytes requires an obligatory recov-
understood. Testosterone's primary effects on liver ery period during which there is no plasma GH
P450 profiles are mediated by the hypothalamic- and hence no stimulation of hepatocyte GH recep-
pituitary axis^"^ and its control of the sex-depend- tors (GHRs)^^. In addition, estrogen may antago-
ent pattern of pituitary GH secretion^^' ^^. nize the induction of CYP2C11 by testosterone as
Consistent with this conclusion, testosterone has suggested by the absence of androgen imprinting
only minor effects on liver enzyme profiles in of this P450 in intact female rats treated with
hypophysectomized rats in most^^ but not all^^' ^^ neonatal or pubertal testosterone^^' ^^. Indeed, the
instances. As discussed in Section 4.2, pituitary stimulatory effect of testosterone on pulsatile GH
GH secretory patterns play a key role in regulating secretion can be blocked by the presence of intact
the expression of the sex-dependent P450 forms. ovaries in female rats^^. Interestingly, prepubertal
treatment of intact (i.e., non-ovariectomized)
female rats with tamoxifen enhances the induction
of CYP2C11 and CYP3A2 protein expression by
4.1.2. Estrogen pubertal and postpubertal androgen^^. The precise
neuroendocrine mechanisms responsible for the
Whereas testosterone has a major positive reg-
antagonistic effects of estrogen on androgen
ulatory influence on the male-specific P450
imprinting remain to be elucidated.
forms, estrogen plays a somewhat lesser role in
the expression of the female-specific and the
female-predominant liver P450 enzymes. Ovari- 4.2. Regulation by Growth
ectomy at birth reduces, but does not abolish,
Hormone
expression of hepatic CYP2C7, CYP2C12, and
steroid 5a-reductase in adult female rats^^' ^^' ^^ and 4.2.1. Sex-Dependent GH Secretory
normal adult enzyme levels can be restored by Profiles
estrogen replacement. Ovariectomy during adult-
hood^^ or neonatal administration of an estrogen In many species, the pituitary gland secretes
receptor antagonist, tamoxifen^ \ reduces hepatic GH into plasma in a highly regulated temporal
CYP3A9 expression in adult female rats. The fashion that differs between males and females.
reduced expression of CYP3A9 in ovariectomized This sex-dependent secretion of GH is most strik-
rats can be restored by estrogen^^. By contrast, ing in rodents^"*"^^, but key features are conserved
estradiol suppresses hepatic CYP2C11 in both in humans^^"^^. In the rat, GH is secreted by the
intact and castrated male rats^^' ^^. However, the pituitary gland in adult males in an intermittent, or
352 David J. Waxman and Thomas K.H. Chang

pulsatile, maimer that is characterized by high (Figure 9.2(A)). By contrast, in the adult female
peaks of hormone in plasma (150-200 ng/ml) rat, GH is secreted more frequently (multiple pitu-
each 3.5-4 hr followed by a period of very low or itary secretory events per hour) and in a manner
undetectable circulating GH ( < 1-2 ng/ml) such that the plasma GH pulses overlap and the

Females

I I I I" I I I I I I I I I' I I I' I I I I I I I I I I I I I I I I I I

9:00 11:00 13:00 15:00 17:00 9:00 11:00 13:00 15:00 17:00

Figure 9.2. Sex-dependence of plasma GH profiles in adult rats. Shown are plasma GH profiles measured during
the course of an 8 hr day in unrestrained and unstressed male (panel A) and female rats (panel B). Data shown are
fromref [73].

A GH Pulse Induction oiCYPICll: B Continuous GH Feminization


Pulse Frequency Dependence M+ M+
M F hGH rGH M
Male Hx + GH
M Hx 6P 2P F
4A2

2C11-
mRNA
fir1 2 3 4 5 6 7 8
W^M
^/;'
I
W 3A2
2C11

F Hx
Female6Hx
P + GH
2P M
2C12
2C11_
mRNA

1 2 3 4 5
Mrtl 6 7 8 9 1 2 3 4 5 6 7 8 9 10 11
Tubulin

Figure 9.3. Role of GH secretory profiles in the expression of sex-dependent rat hepatic GYP mRNAs. Shown are
Northern blots probed with oligonucleotide probes specific to each of the indicated GYP RNAs. Panel A shows
the male (M) specificity of GYP2G11, its absence in hypophysectomized rats (Hx) and its induced expression in
either male or female (F) hypophysectomized rats given either 2 or 6 pulses (P) of GH/day for 7 days. Data based
on ref [73]. Panel B shows the effects of continuous rat (r) or human (h) GH infusion in male rats (lanes 6-10) on
the expression of CYPs 4A2, 2G11, and 3A2 (all male-specific; lanes 1, 2, 11 vs lanes 3-5), as well as the induction
of GYP2G12. Tubulin RNA is shown as a loading control. Data based on ref [32].
Hormonal Regulation of Liver Cytochrome P450 Enzymes 353

hormone is continually present in circulation at expression of the male-specific liver enzyme


significant levels (~15^0ng/ml) at nearly all CYP2C11 (Figure 9.3(A)) and its associated testos-
times^'' (Figure 9.2 (B)). Hypophysectomy and terone 2a-hydroxylase activity^^' ^^' ^^' ^^. The stim-
GH replacement experiments carried out in sev- ulatory effects of intermittent GH stimulation on
eral laboratories have demonstrated that these sex- this "class I" male P450 enzyme can be distin-
dependent plasma GH profiles are, in turn, guished from the effects of GH pulses on a larger,
responsible for establishing and for maintaining second group ("class II") male-specific liver P450s
the sex-dependent patterns of liver P450 gene (CYP2A2, CYP2C13, CYP3A2, CYP3A18, and
expression in rats^^' ^^' ^^' ^^' ^^ and mice^"^' ^^ (for CYP4A2). In contrast to the class ICYP2C11, class
earlier reviews, see refs [24], [86]). Clinical stud- II male P450s are not obligatorily dependent on GH
ies in humans also demonstrate a role for GH^^~^^ pulses, as judged by their high level of expression in
and its sex-dependent plasma secretory patterns^^ the absence of GH, as shown in hypophysectomized
in regulating P450-dependent drug metabolism. rats of both sexes27'28,38,4i,97,98,100,101. Nevertheless,
Studies in the rat model reveal three distinct liver expression of the class II enzymes CYP2A2
responses of liver P450s to plasma GH profiles and CYP3A2 is induced when intermittent GH
(Figure 9.3). pulses are given to adult male rats that are depleted
of circulating GH by neonatal monosodium gluta-
(1) Continuous plasma GH, a characteristic mate (MSG) treatment^^^.
of adult female rats, stimulates expression of female (3) Continuous GH exposure exerts major
specific enzymes, such as CYP2C12 and steroid negative regulatory effects on liver P450 enzyme
5a-reductase^^' ^^, and female-dominant liver expression, as revealed by the marked suppression
enzymes, such as CYP2A1, CYP2C7, and CYP3A9 of each of the class I and class II male-specific rat
(refs [31], [62], [93], [94]). Hepatic levels of P450s following continuous GH treatment of
CYP2C12 and steroid 5a-reductase are unde- intact male rats (Figure 9.3(B)). In some cases this
tectable in hypophysectomized female rats but can effect can be achieved at fairly low GH levels, cor-
be restored to near-normal female level by continu- responding to only 3-12% of the physiological
ous GH replacement^^' ^^~^^. This restoration can be GH levels present in intact female rats^^. The high
achieved with as little as 12-25% of the physiologi- level expression of class II P450 mRNAs seen in
cal levels of GH^^. Higher levels of GH are required the absence of GH pulses, that is, in hypophysec-
to induce expression of CYP2C12 and steroid 5a- tomized male rats, is also suppressed by continu-
reductase in hypophysectomized male rats^^. ous GH, indicating that continuous GH actively
(2) Intermittent plasma GH pulses, which suppresses P450 gene expression, and does not
are characteristic of adult male rats, induce simply abolish the stimulatory pulsatile plasma

Table 9.2. Response of Sex-Specific Rat CYPs to GH

Intact rats Hypophysectomized rats MSG-treated rats

M M M M
+ + + +
GYP F M GH_, F M GH,„, GH,,„, M GHi„,

CYP2C1F - ++ - - + 4- - - ++
(Male class I)
CYP2A2^ _ ++ _ ++ ++ ++ +/- _ + 4-
(Male class II)
CYPC12^ ++
(Female-specific)

F, female; M, male; GH^^j^^, continuous GH; GHj^^j, intermittent (pulsatile) GH.


"+ + " indicates a positive effect, "—" indicates a suppressive effect, or absence of expression, and "+/—" indicates no major effect.
''Data are based on refs [20], [73], [97], [98], [100], [101], [104]-[110].
^Data are based on refs [27], [97], [98], [100], [101], [104], [109]-[111].
'^Data are based on refs [95], [97], [98], [104], [107], [109]-[111].
354 David J. Waxman and Thomas K.H. Chang

GH pattern. GH suppression is also a key deter- mRNAs^^^' ^^^, whose male-specific expression is
minant of the lower responsiveness of female rats primarily a consequence of the suppressive effects
to phenobarbital induction of CYP2B1 (refs of continuous GH exposure in adult female rats^^.
[102], [103]), and probably also the lower respon- Thus, transcription initiation is the key step at
siveness of the females to the induction of CYP4A which the three distinct effects of GH outlined in
enzymes by peroxisome proliferators such as Section 4.2.1 are operative: stimulation of 2C11
clofibrate^^. expression by pulsatile GH, suppression of both
class I and class II male-specific P450s by contin-
The response of the class II male P450 genes
uous GH, and stimulation of CYP2C12 expres-
to hypophysectomy of female rats, which de-
sion by continuous GH^^^.
represses, that is, increases liver enzyme levels to
Consistent with the finding that GH regulates
near-normal intact male liver levels, demonstrates
sex-dependent liver CYPs by transcriptional
that the class II male liver P450s are subject to neg-
mechanisms, the 5'-flanking DNA segments of
ative pituitary regulation in female rat liver, where
the CYP2C11 (ref [115]) and CYP2C12 genes^^^
their expression is strongly repressed by the near
both contain specific DNA sequences that interact
continuous pattern of plasma GH exposure. These
in a sex-dependent and GH-regulated manner with
patterns of hormonal regulation are summarized in
DNA-binding proteins (putative transcription fac-
Table 9.2, which presents the responses of repre-
tors) that are differentially expressed in male vs
sentative sex-specific liver CYPs to continuous and
female rat liver nuclei^^^' ^^^. These DNA
intermittent GH treatment applied to intact, hypo-
sequences are hypothesized to include GH res-
physectomized and neonatal MSG-treated rats.
ponse elements that contribute to the sex-specific
transcription of the CYP2C11 and CYP2C12
4.2.2. Transcriptional Effects of GH on genes. Two negative regulatory elements
CYP Genes ("silencer elements") were also identified in the
CYP2C11 promoter; however, their significance
GH regulates liver P450 steady-state mRNA with respect to GH regulation and sex-specific
levels (e.g.. Figure 9.3) in parallel with P450 P450 expression is as yet unclear^ ^^. Functional
protein and P450 enzyme activity levels, all but studies of the sex-specific CYP promoters
ruling out major regulation by translational and and their interactions with liver-enriched and
post-translational mechanisms, such as GH regu- GH-responsive transcription factors are discussed
lation of P450 protein turnover. Induction of below.
CYP2C12 mRNA by continuous GH requires
ongoing protein synthesis^ *^, suggesting either an
indirect induction mechanism or a requirement for 4.2.3. Cellular Mechanisms of
one or more protein components that may have a
GH Signaling
short half-life. Analysis of liver nuclear RNA
pools demonstrates that unprocessed, nuclear The cellular mechanisms whereby pituitary GH
CYP2C11 and CYP2C12 RNAs respond to circu- secretory profiles differentially regulate expres-
lating GH profiles in a manner that is indistin- sion of the sex-dependent liver P450s are only
guishable from the corresponding mature, partially understood. GH can act directly on the
cytoplasmic mRNAs^^^. Consequently, transport hepatocyte to regulate liver P450 expression, as
of CYP2C11 and CYP2C12 mRNA to the cyto- demonstrated by the responsiveness of primary rat
plasm, and cytoplasmic P450 mRNA stability are hepatocyte cultures to continuous GH-stimulated
unlikely to be important GH-regulated control expression of CYP2C12 mRNA; however, these
points for sex-specific P450 expression. More- effects do not involve IGF-I, a mediator of several
over, nuclear run-on transcription analyses have of GH's physiological effects on extrahepatic
established that GH regulates the sex-specific tissues'^^' ''^. Discrimination by the hepatocyte
expression of the CYP2C11 and CYP2C12 between male and female plasma GH profiles is
genes at the level of transcript initiation^^^' ^^^. likely to occur at the cell surface, where a higher
Transcription is also the major step for regulation level of GHRs (see below) is found in female as
of the male class II CYP2A2 and CYP2C13 compared to male rats'^^. This sex difference in
Hormonal Regulation of Liver Cytochrome P450 Enzymes 355

cell surface GHR abundance may, at least in part, studies), which implies the need for an obligatory
be due to differential effects of intermittent vs recovery period to effectively stimulate CYP2C11
continuous GH stimulation^^^ and could play a expression. This condition is not met in the case of
role in the activation of distinct intracellular sig- hepatocytes exposed to GH continuously (female
naling pathways by chronic (female) as compared hormone profile). This recovery period may serve
to intermittent (male) GH stimulation. to reset the cellular signaling apparatus, for exam-
4.2.3.1. Significance of GH pulse ple, by replenishing GHRs at the cell surface (see
fi^equency. Studies have been carried out to deter- below).
mine which of the three descriptive features of a 4.2.3.2. RoleofGH receptor (GHR). The
GH pulse—^namely, GH pulse duration, GH pulse effects of GH on hepatocytes and other responsive
height, and GH pulse frequency—is required for cells are transduced by GHR, a 620 amino acid
proper recognition of a GH pulse as "masculine." cell surface transmembrane protein^^^ belonging
Direct measurement of the actual plasma GH to the cytokine receptor superfamily^^"^. GHR is
profiles achieved when GH is administered to comprised of a 246 amino acid extracellular
hypophysectomized rats by twice daily s.c. GH domain that binds GH, a single transmembrane
injection (i.e., the intermittent GH replacement pro- segment, and a 350 amino acid intracellular
tocol most commonly used to stimulate CYP2C11 domain that participates in the intracellular signal-
expression) has revealed broad peaks of circulating ing events stimulated by GH^^^' ^^^. X-ray crystal-
GH, which last as long as 5-6 hr^^. These sustained lographic and other studies establish that a single
GH "pulses" are effective in stimulating expression molecule of GH binds in a stepwise manner to
of the male-specific CYP2C11, provided that they two GHR molecules to yield receptor dimers:
are not administered in close succession. It is thus +GHR
apparent that physiological GH pulse duration (<2 GH + GHR -> GH-GHR -> GH-(GHR)2 ^^^^ ^^7
hr) is not required to elicit a male CYP response. (Figure 9.4).
Studies carried out in GH-deficient rat models The four helix bundle protein GH is proposed
(either dwarf rats or rats depleted of adult circulat- to initially bind to a single receptor molecule by
ing GH by neonatal MSG treatment) demonstrate contacts that involve amino acids comprising
that GH pulse height is also not a critical factor for GH's Site 1, followed by binding of a second
stimulation of CYP2C11 expression^^^' ^^^. This molecule of GHR, which interacts with Site 2
finding can be understood in terms of the K^ of on the GH molecule to give a heterotrimeric
the GH-GHR complex, which at 10"^^ M (~2 GH-(GHR)2 complex. GH-induced receptor
ng/ml)^^^, is only 1% of the peak plasma hormone dimerization is reversible, and the equilibrium
level in adult male rats. In contrast, GH pulse fre- may be shifted in favor of monomer formation
quency is a critical determinant for GH stimulation in the continued presence of excess GH:
CTH
of a male pattern of liver P450 expression, as GH-(GHR)2^2GH-GHR. Other, more recent
shown in hypophysectomized rats given physiolog- studies suggest that GH may bind to, and thereby
ical replacement doses of GH for 7 days by inter- activate a preformed receptor dimer at the cell sur-
mittent intravenous injections at frequencies of 2,4, face ^^^' ^^^. Independent of whether the receptor
6, or 7 times per day^^. Analysis of liver CYP2C11 dimer is preformed, however, it is clear that the
levels in these rats revealed a normal male pattern conformational changes that accompany forma-
of liver CYP2C11 gene expression in response to 6 tion of the active GH-(GHR)2 complex are neces-
GH pulses per day (which approximates the normal sary, and probably sufficient, for stimulation of
male plasma GH pulse frequency), as well as in GH-induced intracellular signaling events ^^^
response to GH pulses given at lower frequencies, 2 (Figure 9.4). Although GHR dimerization is thus
or 4 times per day (e.g., Figure 9.3). However, required for most GH responses, some GH
hypophysectomized rats are not masculinized by responses might not require receptor dimeriza-
7 daily GH pulses, indicating that the hepatocyte tion^^ ^ and indeed, might be mediated by
does not recognize the pulse as "masculine" if GH monomeric GH-GHR complexes. Conceivably,
pulsation becomes too frequent. Hepatocytes thus the distinct patterns of liver P450 gene expression
require a minimum GH off time (—2.5 hr in the induced by continuous plasma GH (female GH
hypophysectomized rat model used in these pattern; CYP2C12 expression) as compared to
356 David J. Waxman and Thomas K.H. Chang

GHR
Intermediate

i
Physiological [GH]
i
High [GH]

Signaling via
Protein Tyrosine
Phosphorylation

Figure 9.4. Activation of GHR by GH-induced sequential dimerization mechanism. GHR is shown localized in
the plasma membrane, and sites 1 and 2 of the GH molecule (see text) are as indicated. The active receptor dimer
dominates at physiological GH concentrations. Model based on data presented in ref [128].

pulsatile GH (male GH pattern; CYP2C11 inhibitor IVIG132. Interestingly, although cellular


expression) might, in part, arise from distinct GH ubiquitination activity is required for receptor
signaling pathways perhaps stimulated by endocytosis, GHR itself does not need to undergo
monomeric (GH-GHR) as compared to dimeric ubiquitination, as shown using a mutant GHR
(GH-(GHR)2) hormone-receptor complexes. GH devoid of its cytoplasmic lysine residue targets for
mutants and analogs that bind GHR without ubiquitination'^''' '^^. Thus, the ubiquitin-protea-
effecting fiinctional receptor dimerization'^^~^^^ some system is a major regulator of intracellular
could be used to test this hypothesis. GHR trafficking.
In intact male rat liver, GHR internalizes to an
intracellular compartment coincident with its
stimulation by plasma GH pulses, and then reap- 4.2.4. Role of STAT5b in Sex-Dependent
pears at the cell surface at the time of the next hor- CYP Expression
mone pulse'^^' '^^. Other studies suggest that GHR
undergoes endocytosis constitutively, that is, in a 4.2.4.1. GH signaling pathways involving
ligand-independent manner. GHR internalization STAT transcription factors. How does GH
involves coated vesicles and ultimately takes the impart sex-dependent transcriptional regulation to
receptor to lysosomes for degradation. GHR endo- liver P450 genes? To answer this question, we may
C5^osis and degradation both require (a) an intact consider the following hypotheses: (a) that cell sur-
ubiquitin conjugation system, which targets a spe- face GHRs can discriminate between the two tem-
cific 10 amino acid-long cytoplasmic GHR tail porally distinct patterns of plasma GH stimulation,
sequence, and (b) 26S proteasome activity, as evi- and (b) the receptors can then transduce this infor-
denced by the inhibitory effects of the proteasome mation to the nucleus, where both pulse and
Hormonal Regulation of Liver Cytochrome P450 Enzymes 357

continuous GH-stimulated transcriptional events GH stimulation^^^. This led to the discovery that
occur. Presumably, GH-activated GHR accom- an intracellular signaling protein and transcription
plishes this by activating two distinct pathways of factor, termed STATSb, is present in its nuclear,
intracellular signaling, one in response to GH pulses tyrosine phosphorylated form at a substantially
and the other in response to continuous GH stimu- higher level in male rat liver than in female rat
lation (Figure 9.5). Studies of GH-induced signal liver^^^. STATs are latent cytoplasmic transcrip-
transduction pathways^^^^^^ have highlighted the tion factors that are activated by tyrosine phos-
importance of the GH-bound receptor dimer in acti- phorylation induced by a variety of cytokines and
vating JAK2, a GHR-associated tyrosine kinase that growth factors, and were first discovered as signal
initiates several downstream pathways of intracellu- mediators that carry transcription signals into the
lar protein tyrosine phosphorylation (Figure 9.4). nucleus in the interferon signaling pathway^"^^.
Other studies have shown that a distinct pattern of In hypophysectomized rat liver, where there is
nuclear protein tyrosine phosphorylation is no endogenous GH signaling, there is little or no
induced in rat liver when the effects of the male tyrosine phosphorylated STATSb protein in the
pulsatile plasma GH hormone profile are com- nucleus; essentially all of the STAT5b protein is
pared to those of the female pattern of continuous found in the C3^osolic fraction, where it resides in a

3.5 hr

hr

I I
Male-specific CYP Female-specific CYP
Steroid OHases Steroid OHases
(CYP2C11) (CYP2C12)
Figure 9.5. Distinct intracellular signaling pathways induced by GH are proposed to be activated by plasma GH
pulses, leading to male-specific CYP expression (left), and by continuous GH stimulation, leading to female-specific
CYP expression (right).
358 David J. Waxman and Thomas K.H. Chang

latent, inactive (non-tyrosine phosphorylated) male plasma GH pulse. It thus undergoes repeated
form. However, when a hypophysectomized rat is cycles of translocation from the cytoplasm into the
injected with a single pulse of GH, STATSb protein nucleus, and then back out to the cytoplasm^^^' ^'^^.
appears in the nucleus in its active, tyrosine phos- For example, if the liver is excised from a rat killed
phorylated state within 10-15 min^^^' ^"^^ This tyro- at the peak of a plasma GH pulse, then STATSb is
sine phosphorylation reaction occurs on STATSb tyrosine phosphorylated and localized to the
tyrosine residue 699, enabling two STATSb mole- nucleus, whereas if the liver is excised from a rat
cules to dimerize via mutual interactions between killed at a time point between successive plasma
the phosphotyrosine residue on one STATSb mole- GH pulses, STATSb is inactive and cytoplasmic.
cule and the SH2 domain (a protein module that Moreover, in female rats there is generally a much
recognizes and binds specifically to phosphotyro- lower level of active, nuclear STATSb protein
sine residues) on a second STATSb molecule. The (~S-10% that of the peak male liver level) ^'^^. This
STATSb-STATSb dimer that is thus formed quickly close temporal linkage between plasma GH pattern
enters the nucleus, where it binds with high affinity and the state of liver STATSb activation has been
to DNA sites upstream of genes that are transcrip- confirmed in intact male rats killed at times shown
tionally activated in response to the initial GH to be specifically associated with spontaneous
stimulus (Figure 9.6). peaks or troughs of the plasma GH rhythm^"^"^. The
STATSb is not present in the nucleus at all key difference between male and female rat liver is
times in male rat liver. Rather, STATSb is repeat- that STATSb is repeatedly, and efficiently, acti-
edly activated in concert with the onset of each vated by plasma GH pulses in the case of the

GH

Figure 9.6. Role of complex formed by GH, GH receptor, and the tyrosine kinase JAK2 in activation of STATSb
by tyrosine phosphorylation. JAK2 tyrosine phosphorylates itself and multiple tyrosines on the cytoplasmic tail of
GHR. Several of these sites serve to recruit STATSb to the receptor-kinase complex. STATSb is then tyrosine
phosphorylated, whereupon it dimerizes, translocates to the nucleus, and binds to DNA regulatory elements upstream
of target genes.
Hormonal Regulation of Liver Cytochrome P450 Enzymes 359

males, but is inefficiently activated by the more expression is strongly supported by studies carried
continuous GH profile of the females. The rela- out in mice that are deficient in STAT5b (STATSb
tively weak STATSb activation pathway in female knockout mouse model) ^"^^ (see ref [147] for a
rat liver appears to reflect a partial desensitization review). Disruption of the STAT5b gene results in
of this signaling pathway in response to the chronic two striking phenotypes (Figure 9.8). These
presence of GH in plasma (Figure 9.7)^'*^. phenotypes are seen in STATSb-deficient males
4.2.4.2. STATSb gene knockout mouse but not in corresponding females. First, there is a
model. The proposal that STATSb is a critical global loss of GH-regulated, male-specific liver
factor in mediating GH-regulated liver P450 gene gene expression, including male-specific P450

Activation of STATSb by Plasma GH Pulses


Proposed Role in Male-specific Liver Gene Transcriptionl

time

Plasma
Membrane

Cytosol

Nucleus
rm:

Transcriptional Activation:
Male-specific LiverCVP Genes

Figure 9.7. Proposed cycle for STATSb activation and deactivation in response to sexually dimorphic plasma GH
profiles. GH pulses, but not continuous GH, induce robust STAT5b tyrosine phosphorylation (pTyr). Nuclear
STATSb is dephosphorylated by a tyrosine phosphatase and then returned to the cytoplasm, where it may undergo
a subsequent round of activation/deactivation.
360 David J. Waxman and Thomas K.H. Chang

Loss of Male-Characteristic
Body Weight Gain
Impaired
Pulsatile Suppression of Male-
GH Signaling Specific Gene Expression
Direct in Liver
Loss of Stimulation of Female-Specific
STATSb Liver Gene Expression
Indirect

II Impaired
Feedback-Inhibition Pertubation of
of Pituitary GH Release Sexually Dimorphic
Plasma GH Pattern

Figure 9.8. Impact of STAT5b loss in knockout (KO) mice. Shown in the box at right are the major effects of
STAT5b deficiency. These effects all are a direct result of the loss of GH-induced liver STAT5b signaling (model I),
rather than an indirect response to impaired feedback-inhibition of pituitary GH release (model 11), as demonstrated
by the lack of responsiveness of hypophysectomized STATSb-deficient male mice to GH pulses^^^.

gene expression. Thus, in the absence of STATSb, Hypophysectomy and GH pulse replacement stud-
the liver does not express the male-specific P450s. ies establish that both major phenotypes of
Moreover, the expression of female-specific, GH- STATSb-deficient mice are a direct response to
regulated liver P450s is increased to near-normal the loss of STATSb-dependent GH signaling in the
female levels in STAT5-deficient male mice. Hver, as opposed to indirect effects of the loss of
Thus, the overall pattern of sexually dimorphic STATSb on the overall pattern of GH secretion by
liver gene expression is critically dependent on the the pituitary gland'^^ (Figure 9.8).
presence of STATSb. The elevated expression in 4.2.4.3. Interaction of GH-responsive
STATSb-deficient males of female-specific P4S0s CYP promoters with GH-activated STATSb.
indicates that STATSb can serve as a negative reg- The strong, repeated pulses of nuclear GH-
ulator of female-specific liver P4S0s, in addition activated STATSb that occur in adult male rats
to its positive regulatory effects on male-specific have been proposed to induce binding of STATSb
P4S0 genes. directly to STATS response elements found in pro-
A second phenot3^e exhibited by STATSb- moters of STATS target genes, which may include
deficient mice is that the male pubertal growth sex-dependent P4S0 genes, and thereby medi-
spurt is absent^"^^. This growth deficiency does ate GH pulse-stimulated gene transcription^ ^^.
not emerge until the beginning of puberty, and Consistent with this hypothesis, STATS response
is not seen in STATSb-deficient females. A male- elements matching the consensus sequence
enhanced pubertal growth spurt is characteristic TTC-NNN-GAA have been found upstream of
of all mammals, including humans. In the case of several male-specific rat liver P4S0 genes, includ-
rodents, the pubertal growth spurt is augmented ing CYPs 2C11, 2A2, and 4A2 (ref. [1S2]). GH-
by the strong growth stimulatory effects of the stimulated CYP promoter-luciferase reporter
male pattern of pulsatile GH secretion, which activity has been demonstrated using the corre-
explains why this growth response is enhanced in sponding isolated STATS response elements,
the males. The two major phenotypes that charac- although the magnitude of the GH- and STATSb-
terize STATSb knockout mice are also seen in dependent gene induction is small, generally only
STATSa/STATSb double knockout mice^"^^ but are ~2-3-fold'^^' '^^. Moreover, although pulsatile
not seen in mice where the disruption is limited to STATSb signaling is first seen in young male rats
the STATS a gene^'*^' ^^^, whose protein-coding at ~S weeks of age, when liver CYP2C11 expres-
sequence is ~90% identical to that of STATSb^^^ sion is first detected, precocious activation of
Hormonal Regulation of Liver Cytochrome P450 Enzymes 361

STAT5b, achieved in 3-week-old male rats given of the CYP2C12 promoter have shown that
pulsatile GH injections, does not lead to preco- STAT5b can substantially block the induction
cious CYP2C11 gene induction^'^^. These and of promoter activity by HNF3P and HNF6
other findings suggest that STAT5b may in part (ref. [152]). This inhibitory action of male GH
act in an indirect manner, by regulating the tran- pulse-activated STAT5b on HNF3P- and HNF6-
scriptional activity of liver-enriched transcription stimulated CYP2C12 transcription is consistent
factors (HNFs, hepatocyte nuclear factors) that with the de-repression (i.e., upregulation) of cer-
cooperate with STAT5b to control the expression tain female-specific, GH-regulated mouse P450
of sexually-dimorphic liver P450 genes^^^' i^^-^^e genes seen in male STAT5b-deficient mice^"^^' ^^^.
4.2.4.4. Interactions between STAT5b and The synergistic interaction between HNF6 and
liver transcription factors regulating sex- HNF3P in stimulating CYP2C12 promoter activ-
specific CYPs. Liver-enriched transcription ity illustrates how small differences in the levels
factors are key determinants of the liver-specific of these factors in liver cells in vivo (cf. the 2-3-
expression of many hepatic P450s (ref. [157]), fold higher expression of HNF6 in females) might
including sex-dependent P450s. Functional lead to much larger differences in CYP2C12 gene
analysis of sex-specific liver CYP proximal pro- expression, particularly when coupled to the
moters has led to the identification of the liver inhibitory action of GH pulse-activated STAT5b
transcription factors HNFla and HNF3p as in males. Other studies point to additional com-
strong ^ra«5-activators of CYP2C11 (ref. [152]). plexities, based on the potential of STAT5b to
By contrast, HNFSp and HNF6 serve as strong upregulate CYP2C12 gene transcription via an
^ra«^-activators of CYP2C12 (refs [155], [158]), upstream pair of STAT5 response elements^^^, and
with synergistic interactions between these two liver the potential role of an inhibitory, COOH-terminal
transcription factors leading to an overall ~300-fold truncated STAT5 form^^^
activation of CYP2C12 promoter activity^ ^^. Other 4.2.4.5. Downregulation of hepatic
studies indicate a role for C/EBPa^^^' ^^^ and STAT5b signaling. Other issues relating to GH
HNF4^56 in the regulation of CYP2C12 trans- and the STAT5b signaling pathway that are of cur-
cription. Interestingly, two of the factors regulating rent research interest include how the cycle of
CYP2C12 expression, HNF6 and HNF3P, require STAT5b activation is turned off at the conclusion
GH for maximal expression in liver, with HNF6 of each GH pulse, and how STAT5b is subse-
being expressed at 2-3-fold higher levels in female quently returned to the cytoplasm in an inactive
compared to male rat liver^^^. This sex difference in form, where it apparently waits for ~2-2.5 hr until
HNF6 levels is too small to account for the it can be re-activated by the next pulse of GH
>50-fold higher level of CYP2C12 in female com- (Figure 9.7)^^^. Another important question is why
pared to male liver. Similarly, the STAT5b-respon- robust activation of the STAT5b signaling path-
siveness of male CYP promoter-derived regulatory way is not achieved in female liver. The answer to
elements^^^' ^^^ is too weak to account for the these questions may, in part, involve an intriguing
high degree of male-specificity (> 100-fold) that family of inhibitory proteins, referred to as SOCS
characterizes male-specific CYP expression. and CIS proteins, which turn off signals to various
Accordingly, additional factors and GH regulatory hormones and cytokines, including GH^^^' ^^^. In
mechanisms are likely to be required for the sex- the case of GH signaling, SOCS proteins bind to
specific pattern of gene expression seen in vivo. the GHR-JAK2 tyrosine kinase complex,
One such mechanism may involve an as yet enabling them to inhibit GH signaling by a com-
uncharacterized "regulator of sex-limitation" plex series of interrelated mechanisms ^^^. One of
("Rsl"), which has been defined genetically and these inhibitory factors, CIS, may be induced to a
appears to repress certain GH-regulated, sex- higher level by the continuous (female) GH pat-
dependent liver CYP genes in female mouse liver tern than by the pulsatile (male) GH pattern. This
in a manner that is independent of GH action^^^. inhibitor of STAT5b signaling has been implicated
Another possible mechanism for GH-regulated in the downregulation of GH-induced STAT5b
sex-specific CYP expression involves interactions signaling in liver cells exposed to the female GH
between STAT5b and HNFs. For example, studies pattem^^^
362 David J. Waxman and Thomas K.H. Chang

4.3. Regulation by Thyroid animals^'^^. The induction of rat hepatic P450


Hormone reductase by thyroid hormone in rats occurs by
transcriptional ^^^ and post-transcriptional mecha-
4.3.1. Cytochromes P450 nisms ^^^ and appears to involve enhanced protein
stability in hyperthyroid rat liver* ^'^. P450 reductase
Although GH is the major regulator of specific
levels are also modulated by thyroid hormone sta-
liver P450s, thyroid hormone also plays a critical
tus in several extrahepatic tissues*^^ Conceivably,
role. The major thyroid hormones, T3 and T4, pos-
interindividual differences in P450 reductase levels
itively regulate some^^' ^^ but not alP^ of the
may occur in response to physiological or patho-
female-predominant liver P450 enzymes, while
physiological differences in circulating th)Toid hor-
they negatively regulate several of the male-
mone levels and could be an important contributory
specific enzymes^^'^"^ (Table 9.1). These effects
factor to individual differences in P450 reduc-
of thyroid hormone are operative at the level of
tase/c54ochrome P450-catalyzed carcinogen metab-
mRNA expression, and are independent of the
olism and carcinogen activation reactions.
indirect effects that thyroid hormone has on liver
P450 levels as a consequence of its effects on liver
GHRs^^^ and its stimulation of GH gene tran-
scription and GH secretion by the pituitary^^^. 5. Alteration of Liver P450
Molecular studies of these effects of thyroid hor- Expression by Hormonal
mone have not been carried out. Perturbation

As discussed earlier in this chapter, many,


although not all, liver P450s are under hormone
4.3.2. NADPH-Cytochrome P450 regulatory controls. An individual's circulating
Reductase hormonal profile can, however, be altered under
Thyroid hormone is also required for full certain situations, including drug therapy, exposure
expression of NADPH-cytochrome P450 reduc- to chemicals found in the environment, and disease
tase, a flavoenzyme that catalyzes electron transfer states such as diabetes and liver cirrhosis. The
to all microsomal P450s. P450 reductase is an resultant changes in circulating hormone levels or
obligatory, and oflen rate-limiting electron-transfer alterations in hormone secretory dynamics could,
protein that participates in all microsomal P450- therefore, influence the expression of specific liver
catalyzed drug oxidation and steroid hydroxylase P450s. The following sections describe some of
reactions *^^' '^^. This thyroid hormone dependence the factors that are known to cause hormonal per-
of P450 reductase enzyme expression is evidenced turbation and discuss the impact of these changes
by the major decrease (>80% reduction) in liver on liver P450 expression and on P450-dependent
microsomal P450 reductase activity and P450 drug and xenobiotic metabolism and toxicity.
reductase mRNA levels that occurs following
hypophysectomy'^^ or in response to methimazole-
5.1. Modulation by Drugs
induced hypothyroidism^'''. It is further supported
by the reversal of this activity loss when thyroxine The anticancer drugs cisplatin'^^' ''^^, cyclo-
(T4), but not GH or other pituitary-dependent fac- phosphamide''^^' '^^, and ifosfamide'^^ alter the
tors, is given at a physiological replacement profile of P450 enzyme expression in liver and
dose'^^' ^^K Restoration of liver P450 reductase perhaps other tissues, at least in part due to the hor-
activity in vivo by T4 replacement also effects a monal perturbations that these cytotoxic agents
substantial increase in liver microsomal P450 induce. Treatment of adult male rats with a single
steroid hydroxylase activities. A similar effect can dose of cisplatin depletes serum androgen, and this
be achieved when liver microsomes isolated from effect persists for up to 28 days after drug adminis-
hypophysectomized rats are supplemented with tration'^^. Serum androgen depletion by cisplatin is
exogenous, purified P450 reductase, which prefer- associated with a feminization of hepatic liver
entially stimulates steroid hydroxylation catalyzed enzyme expression. Thus, cisplatin-treated male
by microsomes prepared from thyroid-deficient rats have elevated levels of the female-predominant
Hormonal Regulation of Liver Cytochrome P450 Enzymes 363

CYP2A1, CYP2C7, and steroid 5a-reductase, but role in the suppression of CYP2C11 and CYP3A2
have reduced levels of the male-specific CYP2A2, by cyclosporine, a drug that does not alter the
CYP2C11, and CYP3A2 (refs [175], [176]). The plasma GH peak amplitude, number, or dura-
effects of cisplatin on androgen levels may result tion^^l Phenobarbital^^' s^^' ^^o, dexamethasoneS9\
from the drug's action on the testes^^^' ^^^; however, and S-fluorouracil^^^ also reduce hepatic CYP2C11
effects on the hypothalamus are also suggested to expression, but the mechanism(s) by which these
contribute, both to the observed depletion of circu- effects occur have not been elucidated.
lating testosterone and the resultant alteration in
liver P450 expression^ ^^. Cisplatin treatment of
adult female rats severely decreases circulating 5.2. Modulation by Polycyclic
estradiol levels and significantly reduces the Aromatic Hydrocarbons
expression of the estrogen-dependent CYP2A21,
CYP2C7, and CYP2C12 (ref [176]). Exposure of adult male rats to polycyclic
Serum testosterone is also depleted in adult aromatic hydrocarbons, including 3-methylcholan-
male rats treated with cyclophosphamide^^^' ^^^^^^^ threne (3MC)i^2, i90, 193^ 2,3,7,8-tetrachlorodi-
or ifosfamide^^^ and this depletion is associated benzo-p-dioxin^^"^, anthracene, benz(a)anthracene,
with feminization of liver enzyme profiles^^^' ^^^ in dibenz(a,c)anthracene, dibenz(a,/i)anthracene, and
a manner similar to that produced by cisplatin. 7,12-dimethylbenz(a)anthracene^^^, leads to decre-
While endogenous androgen secretion can be ases in hepatic CYP2C11 protein and activity levels.
stimulated in cyclophosphamide-treated rats by In the case of 3MC, this suppression reflects a
the luteinizing hormone analog chorionic decrease in the rate of CYP2C11 transcription^^^.
gonadotropin, the resultant increase in serum The hormonal mechanisms by which polycyclic aro-
testosterone does not reverse the loss of hepatic matic hydrocarbons modulate CYP2C11 expression
CYP2C11 expression^^^. This observation is anal- are not known, however, 3MC^^^ and 2,3,7,8-tetra-
ogous to the earlier finding that the suppression of chlorodibenzo-/7-dioxin^^^ have been reported to
CYP2C11 by 3,4,5,3',4',5'-hexachlorobiphenyli82 decrease serum testosterone levels. 3MC may inter-
fere with the stimulation of CYP2C11 expression by
is not causally related to the associated depletion
GH^^^, but in a manner that does not involve
of serum testosterone ^^^. Consequently, modu-
STAT5bi99. Interestingly, the extent of CYP2C11
lation of liver enzyme expression by cyclo-
suppression by polycyclic aromatic hydrocarbons
phosphamide may involve action at the
is correlated with Ah receptor-binding affinity and
hypothalamic-pituitary axis, which establishes the
Ah receptor transformation potency^ ^^. However, it
sex-dependent plasma GH profile that in turn dic-
is not clear whether the Ah receptor plays a role in
tates the expression of CYP2C11 and other sex-
the transcriptional suppression of CYP2C11 by poly-
dependent liver P450 enzymes, as discussed earlier
cyclic aromatic hydrocarbons^^^.
in this chapter. CYP2C11 can also be suppressed
by other mechanisms, as demonstrated by the
finding that CYP2C11 levels are suppressed by
the anticancer drug l-(2-chloroethyl)-3-cyclo-
5.3. Modulation by
hexyl-1-nitrosourea (CCNU; lomustine) with- Pathophysiological State
out affecting circulating testosterone levels ^^'^. 5.3.1. Diabetes
Conceivably, CCNU may act directly on the hypo-
thalamic-pituitary axis to alter key signaling ele- Uncontrolled insulin-dependent diabetes is
ments in the ultradian rhythm of circulating GH. accompanied not only by defective carbohydrate
Other drugs that suppress hepatic CYP2C11 metabolism, which results in hyperglycemia,
and CYP3A2 levels include chloramphenicols^^ hyperlipidemia, and hyperketonemia, but it is also
and cyclosporine^^^' ^^^. The effects of chlo- associated with hormonal perturbation, including
ramphenicol are strain-specific, occurring in a reduction in circulating testosterone^^ ^~^^^,
Sprague-Dawley rats but not in Fischer 344 rats. th3a'oid hormone, and plasma Gtf^'*' ^^^. As
Moreover, this suppression is accompanied by a described earlier in this chapter, these hormones
modest reduction in plasma levels of thyroxine but regulate many liver P450 enzymes, either directly
not testosterone s^^. GH does not appear to play a or indirectly. Accordingly, the diabetic state is
364 David J. Waxman and Thomas K.H. Chang

associated with profound changes in the levels of diet to induce cirrhosis have enhanced serum
various hepatic P450 enzymes. Whereas diabetes estradiol concentrations^^^ and reduced testicular
leads to induction of several rat liver P450 forms, weight^^^ and serum testosterone levels^^^. In
including CYP2A1 (refs [205], [206]), CYP2B1 association with the perturbation in hormonal
(refs [207], [208]), CYP2C7 (ref [206]), CYP2E1 status is a major decline in hepatic CYP2C11 con-
(refs [209]-[212]), CYP4A2 (ref. [206]), and tent^^^, and this decline is not accompanied by
CYP4A3 (ref [206]), it suppresses CYP2A2 (ref induction of steroid 5a-reductase activity^^^. The
[205]), CYP2C11 (refs [205], [207], [208], [212]), suppression of hepatic CYP2C11 is also evident in
and CYP2C13 (ref. [205]). Changes in the levels other models of liver cirrhosis, including bile duct
of some of these liver P450s (e.g., CYP2C11 and ligation^^^' ^^^ and carbon tetrachloride-induced
CYP2E1) have been shown at the mRNA leveP^^, cirrhosis^^^' ^^^. Whether the alteration in serum
208,213,214 ^jj^ ^j.g reversed by insulin replacement. steroid hormone levels is directly or indirectly
The profile of GH secretion in the diabetic male responsible for the apparent demasculinization of
rat is altered so as to resemble the pattern found in liver P450 remains to be determined.
the normal female rat^^^. The induction of CYP2A1
and CYP2C7 in diabetic male rats can therefore be
explained, at least in part, as a response to the more
continuous pattern of GH secretion, which stimu-
5.4. Modulation by Ethanol and
lates expression of these P450 forms^^' 3U 63,215 jj^ Dietary Factors
contrast, this pattern of GH secretion reduces Adult male rats administered ethanol by a total
CYP2A2 and CYP2C13 levels because continuous enteral nutrition system have reduced hepatic
GH administration suppresses these two CYP2C11 and CYP3A2 levels, whereas their
P450s2^'28' 116 CYP2C11 expression is obligatorily CYP2A1 activity is unaltered^^^. The same ethanol
dependent on the intermittent male pattern of treatment alters the dynamics of plasma GH secre-
plasma GH secretion^^. Therefore, the more contin- tion by decreasing the GH pulse amplitude and
uous secretion of GH in diabetic male rats^^'* would increasing the GH pulse frequency. The increased
be expected to suppress this P450. In the case of frequency of GH pulses thus explains the reduced
CYP2B1, GH pulse height is the suppressive expression of CYP2C11 after chronic ethanol
signal ^^^ and accordingly, the reduction in GH peak intake because hepatocytes require a minimum "off
concentration in diabetic male rats^^^ leads to time" in order to express the male-pattern of GH
increases in CYP2B1 levels^^^' 208 A GH-inde- secretion that stimulates CYP2CI1 expression^^. In
pendent mechanism is likely to contribute to some another study, chronic intragastric infusion of
of the other effects of diabetes on liver P450 ethanol-containing diets resulted in suppression of
expression. GH, independent of its plasma profile, CYP3A2 while substantially increasing the expres-
is suppressive toward hepatic CYP2E1 (ref [62]), sion of CYP3A9 in adult male rats^^^
but the levels of this P450 are substantially elevated
Dietary vitamin-A deficiency reduces hepatic
in both diabetic male and female rats^^^' 216,217
CYP2C11 (refs [227]-[229]) and CYP4A2
CYP2E1 induction in diabetes has been attributed
levels^^^, while inducing steroid 5a-reductase
to increased plasma concentrations of ketone bod-
activity in adult male rats^"^^. These effects, which
jgg2io, 218 Recently, a role of hypoinsulinemia and
are accompanied by a reduction in plasma testos-
hyperglucagonemia was proposed for the suppres-
terone levels, can be restored by the inclusion of all-
sion of CYP2C11 in diabetes, based on the finding
trans-TQiinoic acid in the diet^^^ or by exogenous
that treatment of cultured rat hepatocytes with
administration of androgen^^^' ^^K Interestingly,
glucagon decreases CYP2C11 expression in a
twice daily subcutaneous administration of GH,
cAMP-dependent manner and this decrease can be
which is known to induce CYP2C11 expression
reversed by insulin administration^ ^^.
in hypophysectomized male rats^^, is not effective
in restoring the levels of CYP2C11 or CYP4A2 in
male rats fed a vitamin A-deficient diet^^^.
5.3.2. Liver Cirrhosis
Dietary trace minerals can also alter liver
Gonadal abnormalities occur in liver cirrhosis. expression of sex-dependent P450 enzymes.
Adult male rats fed a chronic choline-deficient Prepubertal male rats fed a zinc-deficient diet
Hormonal Regulation of Liver Cytochrome P450 Enzymes 365

during the pubertal period have depleted serum of GH secretory patterns and the resultant changes
androgen levels and a feminized pattern of hepatic in the expression of multiple liver P450 enzymes.
mRNA expression, as evidenced by a reduction in Accordingly, diabetes is associated with a decrease
CYP2C11, CYP3A2, and CYP3A18 and by an in P450-mediated in vitro hepatic metabolism of
elevation in CYP2C12 and CYP3A9 (ref [232]). imipramine^^^' ^^^, lidocaine^^^, codeine, and chlor-
However, the precise neuroendocrine mechanisms promazine^^^. In addition, alteration of liver P450
responsible for the feminization of hepatic P450 expression in diabetes is postulated to be responsi-
enzyme expression by dietary zinc deficiency ble for the enhanced in vitro metabolic activation of
remain to be elucidated. certain chemical carcinogens, including Try-P-
Finally, caloric restriction and food deprivation 1 (3-amino-1,4-dimethyl,5H-pyrido(4,5-b)indole)
have been shown to modulate hepatic expression and Try-P-2(3-amino-1 -methyl-5H-pyrido(4,3-b)
of sex-dependent liver steroid-metabolizing indole)^'*^ These examples demonstrate the poten-
enzymes, including CYP2C11 (ref [233]) which is tial for alterations in liver P450 expression that
suppressed, and CYP3A9 (ref [234]) and steroid potentially lead to reduced drug metabolism and
5a-reductase^^^, which are induced. These are sit- enhanced procarcinogen activation. Further investi-
uations in which glucagon levels are high and gations will be necessary to determine the extent
insulin levels are low, analogous to the diabetic to which these events occur in humans and to eval-
state (see Section 5.3.1). uate their true pharmacological and toxicological
significance.

5.5. Impact on Drug Metabolism


and Procarcinogen 6. Conclusion
Activation
Sex-dependent liver P450 enzymes are subject
As discussed in Section 5.1, the anticancer to complex patterns and multiple mechanisms of
drug cisplatin provides an example of a foreign hormonal control. Plasma GH pulsatility, acting
chemical that depletes serum testosterone and via transcriptional mechanisms, is a key regulator
consequently feminizes the expression of liver of liver-expressed, sex-dependent P450 enzymes
P450s in adult male rats. This type of alteration in and their associated steroid hydroxylation and
the profile of liver P450 enzymes could have xenobiotic metabolism activities. This results in
important pharmacological consequences, as sug- the sex differences in metabolism, pharmacoki-
gested by the finding that cisplatin suppression of netics, and toxicity of various xenobiotics that are
CYP2C11 decreases liver P450-catalyzed activa- seen in rodents and other species, including
tion of anticancer prodrugs, such as cyclophos- humans. The pulsatile plasma GH profile charac-
phamide^^^' ^^^' ^^^ and ifosfamide^^^ when teristic of adult male rats repeatedly stimulates
assayed in isolated microsomal systems. Liver tyrosine phosphorylation that directly activates
P450 activation of these latter two drugs is the transcription factor STAT5b. This triggers
required for their anticancer drug activity^^^, and translocation of STAT5b into the nucleus, where it
CYP2C11 contributes significantly to this meta- is proposed to modulate transcription of sex-
bolic pathway in male rat liver^^^' ^^^. Clinical dependent liver P450 genes through both direct
studies indicate that cisplatin may exert effects and indirect mechanisms. Continuous or near-
on circulating hormone levels in human cancer continuous exposure to GH, as occurs in the
patients that are similar, though not identical, to female rodent liver, can downregulate liver
those seen in rats^^^. If these hormone perturba- STAT5b signaling. As a consequence, active
tions in turn alter P450 enzyme levels in human nuclear STAT5b protein is generally found at a
liver, this could have an impact on drug-drug low level in adult female rat liver. Thyroid hor-
interactions in patients given cisplatin in combina- mones are also important endocrine regulators of
tion with anticancer P450 prodrugs such as liver metabolic function that can act directly, by
cyclophosphamide. influencing the expression of individual P450
As discussed above, drug metabolism in diabetic enzymes, as well as indirectly, via their effects on
rats can be altered as a consequence of perturbations NADPH-P450 reductase gene expression.
David J. Waxman and Thomas K.H. Chang

Gonadal hormones also play a role, but their 8. Ryan, D.E. and W. Levin (1990). Purification and
effects on liver P450 activity are largely indirect characterization of hepatic microsomal cytochrome
and are mediated by the hypothalamic-pituitary P-450. Pharmacol. Ther. 45, 153-239.
axis and its control of plasma GH levels. Sex dif- 9. Nelson, D.R. (2003). Comparison of P450s from
human and fugu: 420 million years of vertebrate
ferences in GH secretory dynamics also occur in
P450 evolution. Arch. Biochem. Biophys. 409,18-24.
humans and can be modulated by gonadal hor-
10. Nelson, D.R., L. Koymans, T. Kamataki,
mones. This sexually dimorphic GH secretion J.J. Stegeman, R. Feyereisen, D.J. Waxman et al.
may contribute to the apparent sex differences in (1996). P450 superfamily: Update on new
the levels of certain human P450s and in human sequences, gene mapping, accession numbers and
P450-catalyzed drug metabolism and pharmaco- nomenclature. Pharmacogenetics 6, 1-42.
kinetics. Finally, diverse factors, including many 11. Waxman, D.J. (1988). Interactions of hepatic
drugs and other xenochemicals, pathophysiologi- cytochromes P-450 with steroid hormones.
cal state, and dietary factors can affect hormone Regioselectivity and stereospecificity of steroid
secretion and the hepatic expression of sex- metabolism and hormonal regulation of rat P-450
dependent P450s, which in turn may impact drug enzyme expression. Biochem. Pharmacol. 37,
71-84.
metabolism and procarcinogen activation.
12. Beierle, I., B. Meibohm, and H. Derendorf (1999).
Gender differences in pharmacokinetics and phar-
macodynamics. Int. J. Clin. Pharmacol. Ther. 37,
529-547.
Acknowledgment 13. Schwartz, J.B. (2003). The influence of sex on phar-
macokinetics. Clin. Pharmacokinet. 42, 107-121.
14. Tanaka, E. (1999). Gender-related differences in
Studies carried out in the laboratory of DJW pharmacokinetics and their clinical significance.
were supported in part by the National Institutes J. Clin. Pharm. Ther. 24, 339-346.
of Health (Grant DK33765). 15. Harris, R.Z., L.Z. Benet, and J.B. Schwartz (1995).
Gender effects in pharmacokinetics and pharmaco-
dynamics. Drugs 50, 222-239.
16. Meibohm, B., I. Beierle, and H. Derendorf (2002).
How important are gender differences in pharmaco-
References kinetics? Clin. Pharmacokinet. 41, 329-342.
17. Hunt, CM., WR. Westerkam, and G.M. Stave
1. Kato, R. (1974). Sex-related differences in drug (1992). Effect of age and gender on the activity of
metabolism. Drug Metab. Rev. 3, 1-32. human hepatic CYP3A. Biochem. Pharmacol. 44,
2. Kato, R. (1977). Drug metabolism under pathological 275-283.
and abnormal physiological states in animals and 18. Zaphiropoulos, PG,, A. Mode, G. Norstedt, and
man. Xenobiotica 7, 25-92. J. A. Gustafsson (1989). Regulation of sexual differ-
3. Skett, P. (1987). Hormonal regulation and sex differ- entiation in drug and steroid metabolism. Trends
ences of xenobiotic metabolism. Prog. Drug. Metab. Pharmacol. Sci. 10, 149-153.
10, 85-139. 19. Waxman, D.J. (1984). Rat hepatic cytochrome P-450
4. Davies, D.S., PL. Gigon, and J.R. Gillette (1969). isoenzyme 2c. Identification as a male-specific,
Species and sex differences in electron transport sys- developmentally induced steroid 16 alpha-hydroxy-
tems in liver microsomes and their relationship to lase and comparison to a female-specific cytochrome
ethylmorphine demethylation. Life Sci. 8, 85-91. P-450 isoenzyme. J. Biol. Chem. 259, 15481-15490.
5. Gurtoo, H.L. and N.B. Parker (1976). Organ speci- 20. Morgan, E.T., C. MacGeoch, and J.-A. Gustafsson
ficity of the sex dependent regulation of aryl hydro- (1985). Hormonal and developmental regulation
carbon hydroxylase (AHH) in rat. Biochem. Biophys. of expression of the hepatic microsomal steroid
Res. Commun. 11, 216-222. 16alpha-hydroxylase cytochrome P-450 apoprotein
6. Kato, R. and J.R. Gillette (1965). Sex differences in in the rat. J. Biol. Chem. 260, 11895-11898.
the effects of abnormal physiological states on the 21. Waxman, D.J., G.A. Dannan, and F.P Guengerich
metabolism of drugs by rat liver microsomes. J. (1985). Regulation of rat hepatic cytochrome P-450:
Pharmacol. Exp. Ther. 150, 285-291. Age-dependent expression, hormonal imprinting,
7. Guengerich, F.P. (1987). Enzymology of rat liver and xenobiotic inducibility of sex-specific isoen-
cytochromes P450. In F.P. Guengerich (ed.), zymes. Biochemistry 24, 4409-4417.
Mammalian Cytochromes P450, Vol. 1. CRC Press, 22. MacGeoch, C , E.T. Morgan, and J.A. Gustafsson
Boca Raton, FL, pp. 1-54. (1985). Hypothalamo-pituitary regulation of
Hormonal Regulation of Liver Cytochrome P450 Enzymes 367

cytochrome P-450(15) beta apoprotein levels in rat Male specificity of liver and kidney CYP4A2 mRNA
liver. Endocrinology 111, 2085-2092. and tissue-specific regulation by growth hormone and
23. Nelson, D.R., T. Kamataki, D.J. Waxman, testosterone. J. Biol. Chem. 267, 3915-3921.
F.P. Guengerich, R.W. Estabrook, R. Feyereisen 33. Waxman, D.J., PA. Ram, G. Notani, G.A. LeBlanc,
et al. (1993). The P450 superfamily: Update on new J.A. Alberta, J.J. Morrissey et al. (1990). Pituitary
sequences, gene mapping, accession numbers, early regulation of the male-specific steroid 6 beta-
trivial names of enzymes, and nomenclature. DMA hydroxylase P-450 2a (gene product IIIA2) in adult
Cell Biol 12, 1-51. rat liver. Suppressive influence of growth hormone
24. Waxman, D.J. (1992). Regulation of liver-specific and thyroxine acting at a pretranslational level. Mol.
steroid metabolizing cytochromes P450: Cholesterol Endocrinol. 4, 447^54.
Talpha-hydroxylase, bile acid 6beta-hydroxylase, 34. Ram, PA. and D.J. Waxman (1991). Hepatic P450
and growth hormone-responsive steroid hormone expression in hypothyroid rats: Differential respon-
hydroxylases. J. Steriod Biochem. Mol Biol. 43, siveness of male-specific P450 forms 2a (IIIA2), 2c
1055-1072. (IICl 1), and RLM2 (IIA2) to thyroid hormone. Mol.
25. Nagata, K., N. Murayama, M. Miyata, M. Shimada, Endocrinol. 5, 13-20.
A. Urahashi, Y. Yamazoe et al. (1996). Isolation and 35. McClellan, G.P, D.J. Waxman, M. Caveness, and
characterization of a new rat P450 (CYP3A18) J.A. Goldstein (1987). Phenotypic differences in
cDNA encoding P450(6)beta-2 catalyzing testos- expression of cytochrome P-450g but not its mRNA
terone 6 beta- and 16 alpha-hydroxylations. in outbred male Sprague-Dawley rats. Arch.
Pharmacogenetics 6, 103-111. Biochem. Biophys. 253, 13-25.
26. Wang, H., K.L. Napoh, and H.W. Strobel (2000). 36. Thummel, K.E., L.V Favreau, J.E. Mole, and
Cytochrome P450 3A9 catalyzes the metabolism of J.B. Schenkman (1988). Further characterization of
progesterone and other steroid hormones. Mol. Cell. RLM2 and comparison with a related form of
Biochem. 213, 127-135. cytochrome P450, RLM2b. Arch. Biochem. Biophys.
27. Waxman, D.J., G.A. LeBlanc, J.J. Morrissey, 266,319-333.
J. Staunton, and D.P. Lapenson (1988). Adult male- 37. Bandiera, S., D.E. Ryan, W. Levin, and PE. Thomas
specific and neonatally programmed rat hepatic (1986). Age- and sex-related expression of
P-450 forms RLM2 and 2a are not dependent on cytochromes P450f and P450g in rat liver. Arch.
pulsatile plasma growth hormone for expression. Biochem. Biophys. 248, 658-676.
J. Biol. Chem. 263, 11396-11406. 38. Kawai, M., S.M. Bandiera, T.K.H. Chang, and
28. McClellan, G.P, P Linko, H.N. Yeowell, and G.D. Bellward (2000). Growth hormone regulation
J.A. Goldstein (1989). Hormonal regulation of and developmental expression of rat hepatic
male-specific rat hepatic cytochrome P-450g (P- CYP3A18, CYP3A9, and CYP3A2. Biochem.
450IIC13) by androgens and the pituitary. J. Biol. Pharmacol. 59, 1277-1287.
Chem. 264, 18960-18965. 39. Gonzalez, EJ., B.J. Song, and J.P Hardwick (1986).
29. Dannan, G.A., F.P. Guengerich, and D.J. Waxman Pregnenolone 16 alpha-carbonitrile-inducible P-450
(1986). Hormonal regulation of rat liver microsomal gene family: Gene conversion and differential regu-
enzymes. Role of gonadal steroids in programming, lation. Mol. Cell Biol 6, 2969-2976.
maintenance, and suppression of delta 4-steroid 40. Sonderfan, A.J., M.P Arlotto, D.R. Dutton,
5 alpha-reductase, flavin-containing monooxyge- S.K. McMillen, and A. Parkinson (1987).
nase, and sex-specific cytochromes P-450. J. Biol. Regulation of testosterone hydroxylation by rat liver
Chem. 261, 10728-10735. microsomal cytochrome P-450. Arch. Biochem.
30. Yamazoe, Y, X. Ling, N. Murayama, D. Gong, Biophys. 255, 21^1.
K. Nagata, and R. Kato (1990). Modulation of 41. Yamazoe, Y, N. Murayama, M. Shimada,
hepatic level of microsomal testosterone 7 alpha- K. Yamauchi, K. Nagata, S. Imaoka et al (1988).
hydroxylase, P-450a (P450IIA), by thyroid hormone A sex-specific form of cytochrome P-450 catalyzing
and growth hormone in rat liver. J. Biochem. (Tokyo) propoxycoumarin 0-depropylation and its identity
108, 599-603. with testosterone 6 beta-hydroxylase in untreated rat
31. Ram, PA. and D.J. Waxman (1990). livers: Reconstitution of the activity with microso-
Pretranslational control by thyroid hormone of rat mal lipids. J. Biochem. (Tokyo) 104, 785-790.
liver steroid 5 alpha-reductase and comparison to 42. Gonzalez, F.J., S. Kimura, B.J. Song, J. Pastewka,
the thyroid dependence of two growth hormone- H.V Gelboin, and J.P Hardwick (1986). Sequence
regulated CYP2C mRNAs. J. Biol. Chem. 265, of two related P-450 mRNAs transcriptionally
19223-19229. increased during rat development. An R.dre.l
32. Sundseth, S.S. and D.J. Waxman (1992). Sex- sequence occupies the complete 3' untranslated
dependent expression and clofibrate inducibility of region of a liver mRNA, J. Biol. Chem. 261,
cytochrome P450 4A fatty acid omega-hydroxylases. 10667-10672.
368 David J. Waxman and Thomas K.H. Chang

43. Leo, M.A., S. lida, and C.S. Lieber (1984). Retinoic hydroxylation and reduction of steroids. J. Biol.
acid metabolism by a system reconstituted with Chem. 248, 4987-4997.
cytochrome P-450. Arch. Biochem. Biophys. 234, 55. Gustafsson, J.A., A. Mode, G. Norstedt, and R Skett
305-312. (1983). Sex steroid induced changes in hepatic
44. Mahnke, A., D. Strotkamp, P.H. Roos, W.G. Hanstein, enzymes. Annu. Rev. Physiol. 45, 51-60.
G.G. Chabot, and R Nef (1997). Expression and 56. Bandiera, S. and C. Dworschak (1992). Effects of
inducibility of cytochrome P450 3A9 (CYP3A9) and testosterone and estrogen on hepatic levels of
other members of the CYP3 A subfamily in rat liver. cytochromes P450 2C7 and P450 2C11 in the rat.
Arch. Biochem. Biophys. 337, 62-68. Arch. Biochem. Biophys. 296, 286-295.
45. Colby, H.D. (1980). Regulation of hepatic drug and 57. Cadario, B.J., G.D. Bellward, S. Bandiera,
steroid metabolism by androgens and estrogens. T.K.H. Chang, W.W.W. Ko, E. Lemieux et at. (1992).
Adv. Sex Norm. Res. 4, 27-71. Imprinting of hepatic microsomal cytochrome P-450
46. Arlotto, M.R and A. Parkinson (1989). enzyme activities and cytochrome P-450IIC11 by
Identification of cytochrome P450a (P450I1A1) as peripubertal administration of testosterone in female
the principal testosterone 7 alpha-hydroxylase in rat rats. Mol. Pharmacol. 41, 981-988.
liver microsomes and its regulation by thyroid hor- 58. Janeczko, R., D.J. Waxman, G.A. LeBlanc,
mones. Arch. Biochem. Biophys. 270, 4 5 8 ^ 7 1 . A. Morville, and M. Adesnik (1990). Hormonal
47. de Waziers, I., RH. Cugnenc, C.S. Yang, J.R Leroux, regulation of levels of the messenger RNA encoding
and RH. Beaune (1990). Cytochrome P-450 isoen- hepatic P450 2c (IICU), a constitutive male-spe-
zymes, epoxide hydrolase and glutathione trans- cific form of cytochrome P450. Mol. Endocrinol. 4,
ferases in rat and human hepatic and extrahepatic 295-303.
tissues. J. Pharmacol. Exp. Ther. 253, 387-394. 59. Ribeiro, V. and M.C. Lechner (1992). Cloning and
48. Friedberg, T., P. Siegert, M.A. Grassow, characterization of a novel CYP3A1 allelic variant:
B. Bartlomowicz, and R Oesch (1990). Studies of Analysis of CYP3A1 and CYP3A2 sex-hormone-
the expression of the cytochrome P4501A, P4501IB, dependent expression reveals that the CYP3A2 gene
and P4501IC gene family in extrahepatic and hepatic is regulated by testosterone. Arch. Biochem.
tissues. Environ. Health Perspect. 88, 67-70. Biophys. 293, 147-152.
49. Chen, G.F, M.J. Ronis, RE. Thomas, D.J. Fhnt, and 60. Chang, T.K.H. and G.D. Bellward (1996).
T.M. Badger (1997). Hormonal regulation of micro- Peripubertal androgen imprinting of rat hepatic
somal cytochrome P450 2C11 in rat liver and kid- cytochrome P450 2C11 and steroid 5 alpha-
ney J. Pharmacol. Exp. Ther. 283, 1486-1494. reductase: Pretranslational regulation and impact on
50. Fujita, S., R. Morimoto, M. Chiba, K. Kitani and microsomal drug activation. J. Pharmacol. Exp.
T. Suzuki (1989). Evaluation of the involvement of Ther.UH, 1383-1391.
a male specific cytochrome P-450 isozyme in 61. Anderson, M.D., S.M. Bandiera, T.K.H. Chang, and
senescence-associated decline of hepatic drug G.D. Bellward (1998). Effect of androgen adminis-
metaboHsm in male rats. Biochem. Pharmacol. 38, tration during puberty on hepatic CYP2C11,
3925-3931. CYP3A, and CYP2A1 expression in adult female
51. Robinson, R.C., K. Nagata, H.V Gelboin, J. Rifkind, rats. Drug Metab. Dispos. 26, 1031-1038.
F.J. Gonzalez, and FK. Friedman (1990). Develop- 62. Waxman, D.J., J.J. Morrissey, and G.A. LeBlanc
mental regulation of hepatic testosterone hydroxy- (1989). Female-predominant rat hepatic P-450 forms
lases: Simultaneous activation and repression of j (IIEI) and 3 (IIAl) are under hormonal regulatory
constitutively expressed cytochromes P450 in senes- controls distinct from those of the sex-specific P-450
cent rats. Arch. Biochem. Biophys. 277, 42-46. forms. Endocrinology 124, 2954-2966.
52. Dhir, R.N., W. Dworakowski, and B.H. Shapiro 63. Sasamura, H., K. Nagata, Y Yamazoe, M. Shimada,
(2002). Middle-age alterations in the sexually T. Saruta and R. Kato (1990). Effect of growth hor-
dimorphic plasma growth hormone profiles: mone on rat hepatic cytochrome P-450f mRNA:
Involvement of growth hormone-releasing factor A new mode of regulation. Mol. Cell Endocrinol.
and effects on cytochrome p450 expression. Drug 68, 53-60.
Metab. Dispos. 30, 141-147. 64. Mode, A. and G. Norstedt (1982). Effects of gonadal
53. Shimada, M., N. Murayama, Y. Yamazoe, steroid hormones on the hypothalamo-pituitary-liver
T. Kamataki, and R. Kato (1987). Further studies on axis in the control of sex differences in hepatic steroid
the persistence of neonatal androgen imprinting on metabolism in the rat. J. Endocrinol. 95, 181-187.
sex-specific cytochrome P-450, testosterone and 65. Jansson, J.O. and L.A. Frohman (1987). Differential
drug oxidations. Jpn. J. Pharmacol. 45, 467^78. effects of neonatal and adult androgen exposure on
54. Einarsson, K., J.A. Gustafsson, and A. Stenberg the growth hormone secretory pattern in male rats.
(1973). Neonatal imprinting of liver microsomal Endocrinology 120, 1551-1557.
Hormonal Regulation of Liver Cytochrome P450 Enzymes 369

66. Jansson, J.O. and L.A. Frohman (1987). Inhibitory diabetes mellitus. J. Clin. Endocrinol. Metab. 69,
effect of the ovaries on neonatal androgen imprint- 239-245.
ing of growth hormone secretion in female rats. 79. Winer, L.M., M.A. Shaw, and G. Baumann (1990).
Endocrinology 121, 1417-1423. Basal plasma growth hormone levels in man: New
67. Jansson, J.-O., S. Ekberg, and O. Isaksson (1985). evidence for rhythmicity of growth hormone secre-
Sexual dimorphism in the control of growth tion. J. Clin. Endocrinol. Metab. 70, 1678-1686.
hormone secretion. Endocr. Rev. 6, 128-150. 80. Pincus, S.M., E.F. Gevers, I.C. Robinson, G. van
68. Kamataki, T., M. Shimada, K. Maeda, and R. Kato den Berg, F. Roelfsema, M.L. Hartman et al. (1996).
(1985). Pituitary regulation of sex-specific forms of Females secrete growth hormone with more process
cytochrome P-450 in liver microsomes of rats. irregularity than males in both humans and rats. Am.
Biochem. Biophys. Res. Commun. 130, 1247-1253. J. Physiol. 270, E107-E215.
69. LeBlanc, G.A. and D.J. Waxman (1990). Regulation 81. Veldhuis, J.D. (1996). Gender differences in secre-
and ligand-binding specificities of two sex-specific tory activity of the human somatotropic (growth
bile acid-binding proteins of rat liver cytosol. hormone) axis. Eur J. Endocrinol. 134, 287-295.
J. Biol. Chem. 265, 5654-5661. 82. Ho, K.Y., WS. Evans, R.M. Blizzard, J.D. Veldhuis,
70. Wang, H. and H.W. Strobel (1997). Regulation of G.R. Merriam, E. Samojlik et al. (1987). Effects of
CYP3A9 gene expression by estrogen and catalytic sex and age on the 24-hour profile of growth
studies using cytochrome P450 3A9 expressed in hormone secretion in man: Importance of endoge-
Escherichia coli. Arch. Biochem. Biophys. 344, nous estradiol concentrations. J. Clin. Endocrinol.
365-372. Metab. 64, 5\-5H.
71. Kawai, M., S.M. Bandiera, T.K.H. Chang, 83. Kato, R., Y Yamazoe, M. Shimada, N. Murayama,
P.M. Poulet, P.M. Vancutsem, and G.D. Bellward and T. Kamataki (1986). Effect of growth hormone
(1999). Modulation of hepatic CYP2A1, CYP2C11, and ectopic transplantation of pituitary gland on
and CYP3A9 expression in adult rats by neonatal sex-specific forms of cytochrome P450 and testo-
administration of tamoxifen. Drug Metab. Dispos. sterone and drug oxidations in rat liver. J. Biochem.
27,1392-1398. 100, 895-902.
72. Carlsson, L., E. Eriksson, H. Seeman, and 84. Noshiro, M. and M. Negishi (1986).
J.O. Jansson (1987). Oestradiol increases baseline Pretranslational regulation of sex-dependent testo-
growth hormone levels in the male rat: Possible sterone hydroxylases by growth hormone in mouse
direct action on the pituitary. Acta Physiol. Scand. liver. J. Biol. Chem. 261, 15923-15927.
129, 393-399. 85. Sueyoshi, T., N. Yokomori, K.S. Korach, and
73. Waxman, D.J., N.A. Pampori, RA. Ram, M. Negishi (1999). Developmental action of estrogen
A.K. Agrawal, and B.H. Shapiro (1991). Interpulse receptor-alpha feminizes the growth hormone-Stat5b
interval in circulating growth hormone patterns reg- pathway and expression of Cyp2a4 and Cyp2d9
ulates sexually dimorphic expression of hepatic genes in mouse liver. Mol. Pharmacol. 56, 473^77.
cytochrome P450. Proc. Natl. Acad. Sci. USA 88, 86. Shapiro, B.H., A.K. Agrawal, and N.A. Pampori
6868-6872. (1995). Gender differences in drug metabolism reg-
74. Eden, S. (1979). Age- and sex-related differences in ulated by growth hormone. Int. J. Biochem. Cell
episodic growth hormone secretion in the rat. Biol. 27, 9-20.
Endocrinology 105, 555-560. 87. Cheung, N.W, C. Liddle, S. Coverdale, J.C. Lou, and
75. Tannenbaum, G.S., J.B. Martin, and E. Colle (1976). S.C. Boyages (1996). Growth hormone treatment
Ultradian growth hormone rhythm in the rat: Effects increases cytochrome P450-mediated antipyrine
of feeding, hyperglycemia, and insulin-induced clearance in man. J. Clin. Endocrinol. Metab. 81,
hypoglycemia. Endocrinology 99, 720-727. 1999-2001.
76. Tannenbaum, G.S. and J.B. Martin (1976) Evidence 88. Berglund, E., G. Johannsson, O. Beck, B.A.
for an endogenous ultradian rhythm governing Bengtsson, and A. Rane (2002). Growth hormone
growth hormone secretion in the rat. Endocrinology replacement therapy induces codeine clearance.
98, 562-570. Eur J. Clin. Invest. 32, 507-512.
77. MacLeod, J.N., N.A. Pampori, and B.H. Shapiro 89. Jurgens, G., K.H. Lange, L.O. Reuther,
(1991). Sex differences in the ultradian pattern of B.B. Rasmussen, K. Brosen, and H.R. Christensen
plasma growth hormone concentrations in mice. (2002). Effect of growth hormone on hepatic
J. Endocrinol. 131, 395-399. cytochrome P450 activity in healthy elderly men.
78. Asplin, CM., A.C. Faria, E.G. Carlsen, Clin. Pharmacol. Ther. 71, 162-168.
VA. Vaccaro, R.E. Barr, A. Iranmanesh et al. (1989). 90. Levitsky, L.L., D.A. Schoeller, G.H. Lambert, and
Alterations in the pulsatile mode of growth hormone D.V Edidin (1989). Effect of growth hormone ther-
release in men and women with insulin-dependent apy in growth hormone-deficient children on
370 David J. Waxman and Thomas K.H. Chang

cytochrome P-450-dependent 3-N-demethylation growth hormone profiles of the rat. Mol. Cell
of caffeine as measured by the caffeine ^^C02 Endocrinol, li:^, 167-181.
breath test. Dev. Pharmacol. Ther. 12, 90-95. 102. Shapiro, B.H., N.A. Pampori, D.P. Lapenson, and
91. Redmond, G.P., J.J. Bell, P.S. Nichola, and Waxman D.J. (1994). Growth hormone-dependent
J.M. Perel (1980). Effect of growth hormone on and -independent sexually dimorphic regulation of
human drug metabolism: time course and substrate phenobarbital-induced hepatic cytochromes P450
specificity. Pediatr. Pharmacol. 1, 63-70. 2B1 and 2B2. Arch. Biochem. Biophys. 312,
92. Jaffe, C.A., D.K. Turgeon, K. Lown, R. Demott- 234-239.
Friberg, and P.B. Watkins (2002). Growth hormone 103. Yamazoe, Y, M. Shimada, N. Murayama, and
secretion pattern is an independent regulator of R. Kato (1987). Suppression of levels of
growth hormone actions in humans. Am. J. phenobarbital-inducible rat liver cytochrome
Physiol. Endocrinol. Metab. 283, E1008-E1015. P-450 by pituitary hormone. J. Biol. Chem. 262,
93. Rasmussen, E., B. Ask, N. Finnstrom, A. Skottner- 7423-7428.
Lundin, and A. Rane (1998). Insulin-like growth 104. Shapiro, B.H., J.N. MacLeod, N.A. Pampori,
factor 1 (lGF-1) effects on sex-specific cytochrome J.J. Morrissey, D.P. Lapenson, and D.J. Waxman
P450 enzymes in normal and hypophysectomised (1989). Signalling elements in the ultradian
male rats. Biochem. Pharmacol. 56, 459^66. rhythm of circulating growth hormone regulating
94. Robertson, G.R., G.C. Farrell, and C. Liddle expression of sex-dependent forms of hepatic
(1998). Sexually dimorphic expression of rat cytochrome P450. Endocrinology 125,
CYP3A9 and CYP3A18 genes is regulated by 2935-2944.
growth hormone. Biochem. Biophys. Res. 105. Yamazoe, Y, M. Shimada, N. Murayama,
Commun. 242, 57-60. K. Yamauchi, and R. Kato (1988). Alteration of
95. MacGeoch, C, E.T. Morgan, B. Cordell, and hepatic drug metabolizing activities and contents of
J.A. Gustafsson (1987). Growth hormone regulates cytochrome P-450 isozymes by neonatal
expression of rat liver cytochrome P-450 15 beta at monosodium glutamate treatment. Biochem.
a pretranslational level. Biochem. Biophys. Res. Pharmacol. 2^1, 1687-1691.
Commun. 143, 782-788. 106. Shapiro, B.H., N.A. Pampori, PA. Ram, and
96. Pampori, N.A. and B.H. Shapiro (1994). D.J. Waxman (1993). Irreversible suppression of
Subnormal concentrations in the feminine profile growth hormone-dependent cytochrome P450 2C11
of circulating growth hormone enhance expression in adult rats neonatally treated with monosodium
of female-specific CYP2C12. Biochem. glutamate. J. Pharmacol. Exp. Ther. 265, 979-984.
Pharmacol. 47, 1999-2004. 107. Pampori, N.A. and B.H. Shapiro (1994). Over-
97. Pampori, N.A. and B.H. Shapiro (1996). expression of CYP2C11, the major male-specific
Feminization of hepatic cytochrome P450s by form of hepatic cytochrome P450, in the presence
nominal levels of growth hormone in the feminine of nominal pulses of circulating growth hormone
plasma profile. Mol. Pharmacol. 50, 1148-1156. in adult male rats neonatally exposed to low levels
98. Pampori, N.A. and B.H. Shapiro (1999). Gender of monosodium glutamate. J. Pharmacol. Exp.
differences in the responsiveness of the sex- Ther. Ill, 1067-1073.
dependent isoforms of hepatic P450 to the feminine 108. Pampori, N.A. and B.H. Shapiro (2000). Nominal
plasma growth hormone profile. Endocrinology growth hormone pulses in otherwise normal mas-
140, 1245-1254. culine plasma profiles induce intron retention of
99. Chang, T.K.H., M.M.Y. Chan, S.L. Holsmer, overexpressed hepatic CYP2C11 with associated
S.M. Bandiera, and G.D. Bellward (1996). Impact nuclear splicing deficiency. Endocrinology 141,
of tamoxifen on peripubertal androgen imprinting 4100-4106.
of rat hepatic cytochrome P450 2C11, cytochrome 109. Agrawal, A.K. and B.H. Shapiro (2000).
P450 3A2, and steroid 5 alpha-reductase. Biochem. Differential expression of gender-dependent
Pharmacol. 51,357-368. hepatic isoforms of cytochrome P-450 by pulse
100. Waxman, D.J., PA. Ram, N.A. Pampori, and signals in the circulating masculine episodic
B.H. Shapiro (1995). Growth hormone regulation growth hormone profile of the rat. J. Pharmacol.
of male-specific rat liver P450s 2A2 and 3A2: Exp. Ther. 292, 228-237.
Induction by intermittent growth hormone pulses 110. Pampori, N.A., A.K. Agrawal, and B.H. Shapiro
in male but not female rats rendered growth (2001). Infusion of gender-dependent plasma
hormone deficient by neonatal monosodium gluta- growth hormone profiles into intact rats: Effects of
mate. Mol. Pharmacol. 48, 790-797. subcutaneous, intraperitoneal, and intravenous
101. Agrawal, A.K. and B.H. Shapiro (2001). Intrinsic routes of rat and human growth hormone on
signals in the sexually dimorphic circulating endogenous circulating growth hormone profiles
Hormonal Regulation of Liver Cytochrome P450 Enzymes 371

and expression of sexually dimorphic hepatic C YP continuity of growth hormone (GH) administration
isoforms. Drug Metab. Dispos. 29, 8-16. in the regulation of hepatic GH-receptors, and cir-
111. Waxman, D.J., J.J. Morrissey, J.N. MacLeod, and culating GH-binding protein and insulin-like
B.H. Shapiro (1990). Depletion of serum growth growth factor-I. Endocrinology 131, 423^29.
hormone in adult female rats by neonatal 122. Legraverend, C , A. Mode, T. Wells, 1. Robinson,
monosodium glutamate treatment without loss of and J.A. Gustafsson (1992). Hepatic steroid
female-specific hepatic enzymes P450 2d (TIC 12) hydroxylating enzymes are controlled by the
and steroid 5 alpha-reductase. Endocrinology 126, sexually dimorphic pattern of growth hormone
712-720. secretion in normal and dwarf rats. E4SEB J. 6,
112. Toilet, P., B. Enberg, and A. Mode (1990). Growth 711-718.
hormone (GH) regulation of cytochrome P- 123. Leung, D.W, S.A. Spencer, G. Cachianes,
450IIC12, insulin-like growth factor-I (IGF-I), and R.G. Hammonds, C. Collins, W.J. Henzel et al.
GH receptor messenger RNA expression in (1987). Growth hormone receptor and serum bind-
primary rat hepatocytes: A hormonal interplay ing protein: Purification, cloning and expression.
with insulin, IGF-I, and thyroid hormone. Mol. Nature 330, 531-543.
Endocrinol. 4, 1934-1942. 124. Kelly, PA., S. Ali, M. Rozakis, L. Goujon,
113. Sundseth, S.S., J.A. Alberta, and D.J. Waxman M. Nagano, I. Pellegrini et al. (1993). The growth
(1992). Sex-specific, growth hormone-regulated hormone/prolactin receptor family. Recent Prog.
transcription of the cytochrome P450 2C11 and Horm. Res. 48, 123-164.
2C12 genes. J. Biol Chem. 267, 3907-3914. 125. Colosi, P, K. Wong, S.R. Leong, and W.I. Wood
114. Legraverend, C , A. Mode, S. Westin, A. Strom, (1993). Mutational analysis of the intracellular
H. Eguchi, PG. Zaphiropoulos et al. (1992). domain of the human growth hormone receptor.
Transcriptional regulation of rat P-450 2C gene J. Biol. Chem. 268, 12617-12623.
subfamily members by the sexually dimorphic 126. de Vos, A.M., M. Ultsch, and A.A. Kossiakoff
pattern of growth hormone secretion. Mol. (1992). Human growth hormone and extracellular
Endocrinol. 6, 259-266. domain of its receptor: Crystal structure of the
115. Morishima, N., H. Yoshioka, Y. Higashi, K. Sogawa, complex. Science 255, 306-312.
and Y. Fujii-Kuriyama (1987). Gene structure of 127. Cunningham, B.C., M. Ultsch, A.M. de Vos,
cytochrome P450 (M-1) specifically expressed in M.G. Mulkerrin, K.R. Clauser, and J.A. Wells
male rat liver. Biochemistry. 26, 8279-8285. (1991). Dimerization of the extracellular domain
116. Zaphiropoulos, P.G., A. Strom, J.A. Robertson, and of the human growth hormone receptor by a single
J.A. Gustafsson (1990). Structural and regulatory hormone molecule. Science 254, 821-825.
analysis of the male-specific rat liver cytochrome 128. Fuh, G., B.C. Cunningham, R. Fukunaga,
P-450 g: Repression by continuous growth hor- S. Nagata, D.V Goeddel, and J.A. Wells (1992).
mone administration. Mol. Endocrinol. 4, 53-58. Rational design of potent antagonists to the human
117. Waxman, D.J., S. Zhao, and H.K. Choi (1996). Inter- growth hormone receptor. Science 256,
action of a novel sex-dependent, growth hormone- 1677-1680.
regulated liver nuclear factor with CYP2C12 129. Ross, R.J., K.C. Leung, M. Maamra, W Bennett,
promoter. J. Biol. Chem. 211, 29978-29987. N. Doyle, M.J. Waters et al. (2001). Binding and
118. Strom, A., H. Eguchi, A. Mode, P Toilet, PE. functional studies with the growth hormone recep-
Stromstedt, and J. Gustafson (1994). Characteri- tor antagonist, B2036-PEG (pegvisomant), reveal
zation of the proximal promoter and two silencer effects of pegylation and evidence that it binds to a
elements in the CYP2C gene expressed in rat liver. receptor dimer. J. Clin. Endocrinol. Metab. 86,
DNA Cell Biol. 13, 805-819. 1716-1723.
119. Guzelian, PS., D. Li, E.G. Schuetz, P Thomas, 130. Gent, J., P van Kerkhof, M. Roza, G. Bu, and
W. Levin, A. Mode et al. (1988). Sex change in G.J. Strous (2002). Ligand-independent growth
cytochrome P-450 phenotype by growth hormone hormone receptor dimerization occurs in the endo-
treatment of adult rat hepatocytes maintained in a plasmic reticulum and is required for ubiquitin
culture system on matrigel. Proc. Natl. Acad. Sci. system-dependent endocytosis. Proc. Natl. Acad.
USA 85, 9783-9787. Sci. USA. 99, 9858-9863.
120. Baxter, R.C. and Z. Zaltsman (1984). Induction of 131. Staten, N.R., J.C. Byatt, and G.G. Krivi (1993).
hepatic receptors for growth hormone (GH) and Ligand-specific dimerization of the extracellular
prolactin by GH infusion is sex independent. domain of the bovine growth hormone receptor.
Endocrinology 115, 2009-2014. J. Biol. Chem. 268, 18467-18473.
121. Bick, T, Z. Hochberg, T Amit, O.G. Isaksson, and 132. Bick, T, M.B.H. Youdim, and Z. Hochberg (1989).
J.O. Jansson (1992). Roles of pulsatility and Adaptation of liver membrane somatogenic and
372 David J. Waxman and Thomas K.H. Chang

lactogenic growth hormone (GH) binding to the 145. Gebert, C.A., S.H. Park, and D.J. Waxman (1999).
spontaneous pulsation of GH secretion in the male Down-regulation of liver JAK2-STAT5b signaling
rat. Endocrinology 125, 1711-1717. by the female plasma pattern of continuous growth
133. Bick, T., M.B.H. Youdim, and Z. Hochberg (1989). hormone stimulation. Mol. Endocrinol. 13,
The dynamics of somatogenic and lactogenic 213-227.
growth hormone binding: Internalization to Golgi 146. Udy, G.B., R.P Towers, R.G. Snell, R.J. Wilkins,
fractions in the male rat. Endocrinology 125, S.H. Park, PA. Ram et al. (1997). Requirement of
1718-1722. STAT5b for sexual dimorphism of body growth
134. Strous, G.J. and J. Gent (2002). Dimerization, rates and liver gene expression. Proc. Nad. Acad.
ubiquitylation and endocytosis go together in Sci. USA 94, 7239-7244.
growth hormone receptor fiinction. FEBS Lett. 147. Davey, H.W, R.J. Wilkins, and D.J. Waxman
529, 102-109. (1999). STAT5 signaling in sexually dimorphic
135. Strous, G.J. and P. van Kerkhof (2002). The gene expression and growth patterns. Am. J. Hum.
ubiquitin-proteasome pathway and the regulation Genet. 65, 959-965.
of growth hormone receptor availability. Mol. Cell. 148. Davey, H.W, S.H. Park, DR. Grattan,
Endocrinol. 197, 143-51. M.J. McLachlan, and D.J. Waxman (1999).
136. Carter-Su, C , L. Rui, and J. Herrington (2000). STAT5b-deficient mice are growth hormone pulse-
Role of the tyrosine kinase JAK2 in signal trans- resistant. Role of STAT5b in sex-specific liver P450
duction by growth hormone. Pediatr Nephrol. 14, expression. J. Biol. Chem. 274, 35331-35336.
550-557. 149. Teglund, S., C. McKay, E. Schuetz, J.M. van
137. Waxman, D.J. and S.J. Frank (2000). Growth Deursen, D. Stravopodis, D. Wang et al. (1998).
Hormone Action: Signaling via a JAK/STAT- Stat5a and Stat5b proteins have essential and
coupled Receptor. Humana Press, Totowa, pp. nonessential, or redundant roles in cytokine
55-83. responses. C^//93, 841-850.
138. Finidori, J. (2000). Regulators of growth hormone 150. Park, S.H., X. Liu, L. Hennighausen, H.W. Davey,
signaling. Vitam. Norm. 59, 71-97. and D.J. Waxman (1999). Distinctive roles of
139. Waxman, D.J., PA. Ram, S.H. Park, and H.K. Choi STAT5a and STAT5b in sexual dimorphism of
(1995). Intermittent plasma growth hormone trig- hepatic P450 gene expression. Impact of STAT5a
gers tyrosine phosphorylation and nuclear trans- gene disruption. J. Biol. Chem. 11 A, 7421-7430.
location of a liver-expressed, Stat 5-related DNA 151. Grimley, PM., F Dong, and H. Rui (1999). Stat5a
binding protein. Proposed role as an intracellular and Stat5b: Fraternal twins of signal transduction
regulator of male-specific liver gene transcription. and transcriptional activation. Cytokine Growth
J. Biol. Chem. 270, 13262-13270. Factor Rev. 10, 131-57.
140. Darnell, J.E., Jr. (1997). STATs and gene regula- 152. Park, S.H. and DJ. Waxman (2001). Inhibitory
tion. Science 277, 1630-1635. cross-talk between STAT5b and liver nuclear fac-
141. Ram, PA., S.H. Park, H.K. Choi, and D.J. Waxman tor HNF3beta: Impact on the regulation of growth
(1996). Growth hormone activation of Stat 1, Stat 3, hormone pulse-stimulated, male-specific liver
and Stat 5 in rat liver. Differential kinetics of hor- cytochrome P-450 gene expression. J. Biol. Chem.
mone desensitization and growth hormone stimu- 276,43031-43039.
lation of both tyrosine phosphorylation and 153. Subramanian, A., J. Wang, and G. Gil (1998). STAT
serine/threonine phosphorylation. J. Biol. Chem. 5 and NF-Y are involved in expression and growth
211, 5929-5940. hormone-mediated sexually dimorphic regulation
142. Choi, H.K. and D.J. Waxman (2000). Plasma of cytochrome P450 3A10/lithocholic acid 6beta-
growth hormone pulse activation of hepatic JAK- hydroxylase. Nucleic Acids Res. 26, 2173-2178.
STAT5 signaling: Developmental regulation and 154. Lahuna, O., M. Rastegar, D. Maiter, J.P Thissen,
role in male-specific liver gene expression. FP Lemaigre, and G.G. Rousseau (2000).
Endocrinology 141, 3245-3255. Involvement of STAT5 (signal transducer and acti-
143. Choi, H.K. and D.J. Waxman (1999). Growth vator of transcription 5) and HNF-4 (hepatocyte
hormone, but not prolactin, maintains, low-level nuclear factor 4) in the transcriptional control
activation of STAT5a and STAT5b in female rat of the hnf6 gene by growth hormone. Mol.
liver. Endocrinology 140, 5126-5135. Endocrinol. 14, 285-294.
144. Tannenbaum, G.S., H.K. Choi, W Gurd, and 155. Delesque-Touchard, N., S.H. Park, and D.J.
D.J. Waxman (2001). Temporal relationship Waxman (2000). Synergistic action of hepatocyte
between the sexually dimorphic spontaneous GH nuclear factors 3 and 6 on CYP2C12 gene expres-
secretory profiles and hepatic STAT5 activity. sion and suppression by growth hormone-activated
Endocrinology 142, 4599^606. STAT5b. Proposed model for female specific
Hormonal Regulation of Liver Cytochrome P450 Enzymes 373

expression of CYP2C12 in adult rat liver. J. Biol. 167. Samuels, H.H., B.M. Forman, Z.D. Horowitz, and
Chem. 215, 34113-34^2. Z.S. Ye (1988). Regulation of gene expression by
156. Sasaki, Y., Y. Takahashi, K. Nakayama, and thyroid hormone. J. Clin. Invest. 81, 957-967.
T. Kamataki (1999). Cooperative regulation of 168. Kaminsky, L.S. and E.P Guengerich (1985).
CYP2C12 gene expression by STAT5 and liver- Cytochrome P450 isozyme/isozyme functional
specific factors in female rats. J. Biol. Chem. 274, interactions and NADPH-cytochrome P450 reduc-
37117-37124. tase concentrations as factors in microsomal metab-
157. Akiyama, I.E. and F.J. Gonzalez (2003). olism of warfarin. Eur J. Biochem. 149, 479^89.
Regulation of P450 genes by liver-enriched tran- 169. Miwa, G.T, S.B. West, and A.Y.H. Lu (1978).
scription factors and nuclear receptors. Biochim. Studies on the rate-limiting enzyme component in
Biophys. Acta. 1619, 223-234. the microsomal monooxygenase system. Incorpo-
158. Lahuna, O., L. Fernandez, H. Karlsson, D. Maiter, ration of purified NADPH cytochrome c-reductase
F.P. Lemaigre, G.G. Rousseau et al. (1997). and cytochrome P450 into rat liver microsomes.
Expression of hepatocyte nuclear factor 6 in rat JBC 253, 1921-1929.
liver is sex-dependent and regulated by growth 170. Waxman, D.J., J.J. Morrissey, and G.A. LeBlanc
hormone. Proc. Natl. Acad. Sci. USA 94, (1989). Hypophysectomy differentially alters P-450
12309-12313. protein levels and enzyme activities in rat liver:
159. Buggs, C., N. Nasrin, A. Mode, P Toilet, Pituitary control of hepatic NADPH cytochrome
H.F Zhao, J. A. Gustafsson et al. (1998). IRE-ABP P-450 reductase. Mol. Pharmacol. 35, 519-525.
(insulin response element-A binding protein), an 171. Ram, PA. and D.J. Waxman (1992). Thyroid hor-
SRY-like protein, inhibits C/EBPalpha (CCAAT/ mone stimulation of NADPH P450 reductase
enhancer-binding protein alpha)-stimulated expression in liver and extrahepatic tissues.
expression of the sex-specific cytochrome P450 Regulation by multiple mechanisms. J. Biol.
2C12 gene. Mol. Endocrinol. 12, 1294-1309. Chem. 261, 3294-3301.
160. TuUis, K.M., C.J. Krebs, J.Y. Leung, and 172. Li, H.C., D. Liu, and D.J. Waxman (2001).
D.M. Robins (2000). The regulator of sex-limitation Transcriptional induction of hepatic NADPH:
gene, rsl, enforces male-specific liver gene expres- Cytochrome P450 oxidoreductase by thyroid hor-
sion by negative regulation. Endocrinology 144, mone. Mol. Pharmacol. 59, 987-995.
1854-1860. 173. Liu, D. and D.J. Waxman (2002). Post-transcriptional
161. Helander, H., J.A. Gustafsson, and A. Mode regulation of hepatic NADPH-cytochrome P450
(2002). Possible involvement of truncated signal reductase by thyroid hormone: Independent effects
transducer and activator of transcription-5 in the on poly(A) tail length and mRNA stability. Mol.
GH pattern-dependent regulation of CYP2C12 Pharmacol. 61, 1089-1096.
gene expression in rat liver. Mol. Endocrinol. 16, 174. Apletalina, E.V, H.C. Li, and D.J. Waxman (2003).
1598-1611. Evaluation of thyroid hormone effects on liver P450
162. Gebert, C.A., S.H. Park, and D.J. Waxman (1999). reductase translation. Arch. Biochem. Biophys. 409,
Termination of growth hormone pulse-induced 172-179.
STAT5b signaling. Mol. Endocrinol. 13, 38-56. 175. LeBlanc, G.A. and D.J. Waxman (1988).
163. Ram, PA. and D.J. Waxman (1999). SOCS/CIS Feminization of rat hepatic P-450 expression by
protein inhibition of growth hormone-stimulated cisplatin. Evidence for perturbations in the hor-
STAT5 signaling by multiple mechanisms. J. Biol. monal regulation of steroid-metabolizing enzymes.
C/^em. 274, 35553-35561. J. Biol. Chem. 263, 15732-15739.
164. Adams, T.E., J.A. Hansen, R. Starr, N.A. Nicola, 176. LeBlanc, G.A., S.S. Sundseth, G.F. Weber, and
D.J. Hilton, and N. Billestrup (1998). Growth hor- D.J. Waxman (1992). Platinum anticancer drugs
mone preferentially induces the rapid, transient modulate P-450 mRNA levels and differentially
expression of SOCS-3, a novel inhibitor of alter hepatic drug and steroid hormone metabolism
cytokine receptor signaling. J. Biol. Chem. 273, in male and female rats. Cancer Res. 52, 540-547.
1285-1287. 177. LeBlanc, G.A. and D.J. Waxman (1990). Mech-
165. Ram, PA. and D.J. Waxman (2000). Role of the anisms of cyclophosphamide action on hepatic
cytokine-inducible SH2 protein CIS in desensitiza- P-450 expression. Cancer Res. 50, 5720-5726.
tion of STAT5b signaling by continuous growth 178 Chang, TK.H. and D.J. Waxman (1993).
hormone. J. Biol. Chem. 275, 39487-39496. Cyclophosphamide modulates rat hepatic
166. Hochberg, Z., T Bick, and Z. Hard (1990). cytochrome P450 2C11 and steroid 5alpha-
Alterations of human growth hormone binding reductase activity and messenger RNA levels
by rat liver membranes during hypo- and hyper- through the combined action of acrolein and phos-
thyroidism. Endocrinology 126, 325-329. phoramide mustard. Cancer Res. 53, 2490-2497.
374 David J. Waxman and Thomas K.H. Chang

179. Maines, M.D. and R.D. Mayer (1985). Inhibition 190. Shimada, M., N. Murayama, K. Yamauchi,
of testicular cytochrome P-450-dependent steroid Y Yamazoe, and R. Kato (1989). Suppression in
biosynthesis by cis-platinum. J. Biol. Chem. 260, the expression of a male-specific cytochrome
6063-6068. P450, P450-male: Difference in the effect of chem-
180. Maines, M.D., P.M. Sluss, and M. Iscan (1990). ical inducers on P450-male mRNA and protein in
cis-Platinum-mediated decrease in serum testo- rat livers. Arch. Biochem. Biophys. 270, 578-587.
sterone is associated with depression of luteinizing 191. Levin, W, PE. Thomas, D.E. Ryan, and
hormone receptors and cytochrome P-450scc in rat A.W Wood (1987). Isozyme specificity of testos-
testis Endocrinology 126, 2398-2406. terone 7 alpha-hydroxylation in rat hepatic micro-
181. McClure, M.T. and I. Stupans, (1995). Hormonal somes: Is cytochrome P-450a the sole catalyst?
perturbation as a possible mechanism for the alter- Arch. Biochem. Biophys. 258, 630-635.
ation of cytochrome P450 by cyclophosphamide. 192. Afsar, A., C. Lee, and D.S. Riddick (1996).
Biochem. Pharmacol. 49, 1827-1836. Modulation of the expression on constitutive rat
182. Yeowell, H.N., D.J. Waxman, A. Wadhera, and hepatic cytochrome P450 isozymes by 5-
J. A. Goldstein (1987). Suppression of the constitu- fluorouracil. Can. J. Physiol. Pharmacol. 74,
tive, male-specific rat hepatic cytochrome P-450 150-156.
2c and its mRNA by 3,4,5,3',4',5'-hexachloro- 193. Jones, E.J. and D.S. Riddick (1996). Regulation
biphenyl and 3-methylcholanthrene. Mol. of constitutive rat hepatic cytochromes P450 by
Pharmacol. 32, 340-347. 3-methylcholanthrene. Xenobiotica 26, 995-1012.
183. Yeowell, H.N., D.J. Waxman, G.A. LeBlanc, 194. Gustafsson, J.A. and M. Ingelman-Sundberg
P. Linko, and J.A. Goldstein (1989). Suppression of (1979). Changes in steroid hormone metabolism in
male-specific cytochrome P450 2c and its mRNA rat liver microsomes following administration
by 3,4,5,3',4',5'-hexachlorobiphenyl in rat liver is of 2,3,7,8-tetrachlorodibenzo-p-dioxine (TCDD).
not causally related to changes in serum testos- Biochem. Pharmacol. 28, 497-9.
terone. Arch. Biochem. Biophys. 271, 508-514. 195. Safa, B., C. Lee, and D.S. Riddick (1997). Role of
184. Chang, T.K.H., H. Chen, and D.J. Waxman (1994). the aromatic hydrocarbon receptor in the suppres-
1 -(2-chloroethyl)-3-cyclohexyl-1 -nitrosourea sion of cytochrome P-450 2C11 by polycyclic aro-
(CCNU) modulates rat liver microsomal matic hydrocarbons. Toxicol. Lett. 90, 163-175.
cyclophosphamide and ifosphamide activation by 196. Lee, C. and D.S. Riddick (2000). Transcriptional
suppressing cytochrome P450 2C11 messenger suppression of cytochrome P450 2C11 gene
RNA levels. Drug Metab. Dispos. 22, 673-679. expression by 3-methylcholanthrene. Biochem.
185. Kraner, J.C, E.T. Morgan, and J.R. Halpert (1994). Pharmacol. S9, 1417-1423.
Selective suppression of rat hepatic cytochrome 197. Moore, R.W, C.L. Potter, H.M. Theobald,
P450 2C11 by chloramphenicol. J. Pharmacol J. A. Robinson, and R.E. Peterson (1985)
Exp. Ther. 210, 1367-1372. Androgenic deficiency in male rats treated with
186. Isogai, M., N. Shimada, T. Kamataki, S. Imaoka, 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol. Appl.
and Y. Funae (1993). Changes in the amounts of Pharmacol. 19, 99-\\\.
cytochromes P450 in rat hepatic microsomes 198. Timsit, YE. and D.S. Riddick (2000). Interference
produced by cyclosporin A. Xenobiotica. 23, with growth hormone stimulation of hepatic
799-807. cytochrome P4502C11 expression in hypophy-
187. Brunner, L.J., WM. Bennett, and DR. Koop sectomized male rats by 3-methylcholanthrene.
(1996). Selective suppression of rat hepatic micro- Toxicol. Appl. Pharmacol. 163, 105-14.
somal activity during chronic cyclosporine nephro- 199. Timsit, YE. and D.S. Riddick (2002). Stimulation
toxicity J. Pharmacol. Exp. Ther. 211, 1710-1718. of hepatic signal transducer and activator of tran-
188. Lu, S.K., S.M. Callahan, and L.J. Brunner (2003). scription 5b by GH is not altered by 3-methyl-
Suppression of hepatic CYP3A1/2 and CYP2C11 cholanthrene. Endocrinology 143, 3284-3294.
by cyclosporine is not mediated by akering growth 200. Riddick, D.S., C. Lee, A. Bhathena, and
hormone levels. J. Pharmacol. Exp. Ther. 305, YE. Timsit (2003). The 2001 Veylien Henderson
331-337. Award of the Society of Toxicology of Canada.
189. Guengerich, F.P, G.A. Dannan, S.T. Wright, Positive and negative transcriptional regulation of
M.V Martin, and L.S. Kaminsky (1982). Purifi- cytochromes P450 by polycyclic aromatic hydro-
cation and characterization of liver microsomal carbons. Can. J. Physiol. Pharmacol. 81, 59-77.
cytochromes P-450: Electrophoretic, spectral, cat- 201. Murray, F.T., J. Orth, G. Gunsalus, J. Weisz, J.B. Li,
alytic, and immunochemical properties and L.S. Jefferson et al. (1981). The pituitary-testicular
inducibility of eight isozymes isolated from rats axis in the streptozotocin diabetic male rat:
treated with phenobarbital or beta-naphthoflavone. Evidence for gonadotroph, Sertoli cell and Leydig
Biochemistry 21, 6019-6030. cell dysfunction. Int. J. Androl. 4, 265-80.
Hormonal Regulation of Liver Cytochrome P450 Enzymes 375

202. Warren, B.L., R. Pak, M. Finlayson, L. 214. Yamazoe, Y, N. Murayama, M. Shimada,


Gontovnick, G. Sunahara, and G.D. Bellward S. Imaoka, Y Funae, and R. Kato (1989).
(1983). Differential effects of diabetes on microso- Suppression of hepatic levels of an ethanol-
mal metabolism of various substrates. Comparison inducible P-450DM/J by growth hormone:
of streptozotocin and spontaneously diabetic Relationship between the increased level of
Wistar rats. Biochem. Pharmacol. 32, 327-335. P-450DM/J and depletion of growth hormone in
203. Skett, P., R.A. Cochrane, and L.A. Joels (1984). diabetes. Mol. Pharmacol. 36, 716-722.
The role of androgens in the effect of diabetes mel- 215. Westin, S., A. Strom, J.A. Gustafsson, and
litus on hepatic drug metabolism in the male rat. P.G. Zaphiropoulos (1990). Growth hormone regu-
Acta Endocrinol. 107, 506-512. lation of the cytochrome P-450IIC subfamily in the
204. Tannenbaum, G.S. (1981). Growth hormone secre- rat: Inductive, repressive, and transcriptional
tory dynamics in streptozotocin diabetes: Evidence effects on P-450f (nC7) and P-450PB1 (IIC6)
of a role for endogenous circulating somatostatin. gene expression. Mol. Pharmacol. 38, 192-197.
Endocrinology 108, 76-82. 216. Donahue, B.S., L.A. Skottner, and E.T. Morgan
205. Thummel, K.E. and J.B. Schenkman (1990). (1991). Growth hormone-dependent and
Effects of testosterone and growth hormone treat- -independent regulation of cytochrome P-450
ment on hepatic microsomal P450 expression in isozyme expression in streptozotocin-diabetic rats.
the diabetic rat. Mol. Pharmacol. 37, 119-129. Endocrinology 128, 2065-2076.
206. Shimojo, N., T. Ishizaki, S. Imaoka, Y. Funae, 217. Barnett, C.R., S. Rudd, PR. Flatt, and C. loannides
S. Fujii, and K. Okuda (1993). Changes in amounts (1993). Sex differences in the diabetes-induced
of cytochrome P450 isozymes and levels of catalytic modulation of rat hepatic cytochrome P450
activities in hepatic and renal microsomes of rats proteins. Biochem. Pharmacol. 45, 313-319.
with streptozocin-induced diabetes. Biochem. 218. Barnett, C.R., L. Petrides, J. Wilson, PR. Flatt, and
Pharmacol 46, 621-627. C. loannides (1992), Induction of rat hepatic
207. Yamazoe, Y., N. Murayama, M. Shimada, mixed-function oxidases by acetone and other
K. Yamauchi, and R. Kato (1989). Cytochrome physiological ketones: Their role in diabetes-
P450 in livers of diabetic rats: Regulation by induced changes in cytochrome P450 proteins.
growth hormone and insulin. Arch. Biochem. Xenobiotica 11, 1441-1450.
Biophys. 268, 567-575. 219. Iber, H., T. Li-Masters, Q. Chen, S. Yu, and
208. Donahue, B.S. and E.T. Morgan (1990). Effects of E.T. Morgan (2001). Regulation of hepatic
vanadate on hepatic cytochrome P-450 expression cytochrome P450 2C11 via cAMP: Implications
in streptozotocin-diabetic rats. Drug. Metab. for down-regulation in diabetes, fasting, and
Dispos. 18,519-526. inflammation. J. Pharmacol. Exp. Ther. 297,
209. Favreau, L.V, D.M. Malchoff, J.E. Mole, and 174-180.
J.B. Schenkman (1987). Responses to insulin by 220. Murray, M., E. Cantrill, I. Mehta, and G.C. Farrell
two forms of rat hepatic microsomal cytochrome (1992). Impaired expression of microsomal
P-450 that undergo major (RLM6) and minor cytochrome P450 2C11 in choline-deficient rat
(RLM5b) elevations in diabetes. J. Biol. Chem. liver during the development of cirrhosis. J.
161, 14319-14326. Pharmacol. Exp. Ther. 261, 373-380.
210. Bellward, G.D., T. Chang, B. Rodrigues, J.H. 221. Murray, M., L. Zaluzny, and G.C. Farrell (1986).
McNeill, S. Maines, D.E. Ryan etal. (1988). Hepatic Drug metabolism in cirrhosis. Selective changes in
cytochrome P-450J induction in the spontaneously cytochrome P-450 isozymes in the choline-defi-
diabetic BB rat. Mol. Pharmacol. 33, 140-143. cient rat model. Biochem. Pharmacol. 35,
211. Dong, Z.G., J.Y Hong, Q.A. Ma, D.C. Li, 1817-1824.
J. Bullock, F.J. Gonzalez et al. (1988). Mechanism 222. Murray, M., L. Zaluzny, and G.C. Farrell (1987).
of induction of cytochrome P-450ac (P-450J) in Impaired androgen 16 alpha-hydroxylation in
chemically induced and spontaneously diabetic hepatic microsomes from carbon tetrachloride-
rats. Arch. Biochem. Biophys. 263, 29-35. cirrhotic male rats. Gastroenterology 93, 141-147.
212. Ma, Q., G.A. Dannan, F.P. Guengerich, and 223. Chen, J., M. Murray, C. Liddle, X.M. Jiang, and
C.S. Yang (1989). Similarities and differences in G.C. Farrell (1995). Downregulation of male-
the regulation of hepatic cytochrome P-450 specific cytochrome P450s 2C11 and 3A2 in bile
enzymes by diabetes and fasting in male rats. duct-ligated male rats: Importance to reduced
Biochem. Pharmacol. 38, 3179-3184. hepatic content of cytochrome P450 in cholestasis.
213. Song, B.J., T. Matsunaga, J.P Hardwick, S.S. Park, Hepatology 11, 580-587.
R.L. Veech, C.S. Yang et al. (1987). Stabilization 224. Bastien, M.C., F Leblond, V Pichette, and
of cytochrome P450j messenger ribonucleic acid J.P Villeneuve (2000). Differential alteration of
in the diabetic rat. Mol. Endocrinol. 1, 542-547. cytochrome P450 isoenzymes in two experimental
376 David J. Waxman and Thomas K.H. Chang

models of cirrhosis. Can. J. Physiol. Pharmacol. 233. Manjgaladze, M., S. Chen, L.T. Frame, J.E.
78, 912-919. Seng, PH. Duffy, R.J. Feuers et al. (1993).
225. Badger, T.M., M.J.J. Ronis, C.K. Lumpkin, Effects of caloric restriction on rodent drug and
C.R. Valentine, M. Shahare, D. Irby et al. (1993). carcinogen metabolizing enzymes: Implications
Effects of chronic ethanol on growth hormone for mutagenesis and cancer. Mutat. Res. 295,
secretion and hepatic cytochrome P450 isozymes 201-222.
of the rat. J. Pharmacol. Exp. Ther. 264, 438^47. 234. Cheesman, M.J. and PE. Reilly (1998).
226. Rowlands, J.C, H. Wang, R. Hakkak, M.J. Ronis, Differential inducibility of specific mRNA corre-
H.W. Strobel, and T.M. Badger (2000). Chronic sponding to five CYP3A isoforms in female rat
intragastric infusion of ethanol-containing diets liver by RU486 and food deprivation: Comparison
induces CYP3A9 while decreasing CYP3A2 in with protein abundance and enzymic activities.
male rats. J. Pharmacol. Exp. Ther. 295, 747-752. Biochem. Pharmacol. 56, 473^81.
227. Martini, R. and M. Murray (1994). Suppression of 235. Clarke, L. and D.J. Waxman (1989). Oxidative
the constitutive microsomal cytochrome P450 metabolism of cyclophosphamide: Identification
2C11 in male rat liver during dietary vitamin A of the hepatic monooxygenase catalysts of drug
deficiency. Biochem. Pharmacol. 48, 1305-1309. activation. Cancer Res. 49, 2344-2350.
228. Martini, R., A.M. Butler, X.M. Jiang, and M. Murray 236. Weber, G.F and D.J.Waxman (1993). Activation of
(1995). Pretranslational down regulation of the anti-cancer drug ifosphamide by rat liver
cytochrome P450 2C11 in vitamin A-deficient male microsomal P450 enzymes. Biochem. Pharmacol.
rat liver: Prevention by dietary inclusion of retinoic 45, 1685-1694.
acid. J. Pharmacol. Exp. Ther. 273, 427-^34. 237. Sladek, N.E. (1988). Metabolism of oxazaphos-
229. Murray, M., R.M. Sefton, K.D. Croft, and phorines. Pharmacol. Ther. 37, 301-355.
A.M. Butler (2001). Differential regulation of 238. LeBlanc, G.A., PW Kantoff, S.F Ng, E. Frei 3rd,
endobiotic-oxidizing cytochromes P450 in vitamin and D.J. Waxman (1992). Hormonal perturbations
A-deficient male rat liver Br J. Pharmacol. 134, in patients with testicular cancer treated with
1487-1497. cisplatin. Cancer 69, 2306-2310.
230. Murray, M. and A.M. Butler (1999). 239. Rouer, E., A. Lemoine, T. Cresteil, P. Rouet, and
Pretranslational up-regulation of the hepatic J.P Leroux (1987). Effects of genetically or chem-
microsomal delta4-3-oxosteroid 5alpha-oxidore- ically induced diabetes on imipramine metabolism.
ductase in male rat liver by all-trans-retinoic acid. Respective involvement of flavin monooxygenase
Biochem. Pharmacol. 58, 355-362. and cytochrome P450-dependent monooxyge-
231. Murray, M., A.M. Butler, and C. Agus (1996). nases. Drug Metab. Dispos. 15, 524-528.
Restoration of cytochrome P450 2C11 in vitamin 240. Dixon, R.L., L.G. Hart, and J.R. Fouts (1961). The
A-deficient rat liver by exogenous androgen. metabolism of drugs by liver microsomes from
FASEBJ. 10, 1058-1063. alloxan-diabetic rats. J. Pharmacol. Exp. Ther.
232. Xu, Z., M. Kawai, S.M. Bandiera, andT.K.H. Chang 133,7-11.
(2001). Influence of dietary zinc deficiency during 241. loannides, C, S.L. Bass, A.D. Ayrton, J. Trinick,
development on hepatic CYP2C11, CYP2C12, R. Walker, and PR. Flatt (1988). Streptozotocin-
CYP3A2, CYP3A9, and CYP3A18 expression in induced diabetes modulates the metabolic activa-
postpubertal male rats. Biochem. Pharmacol. 62, tion of chemical carcinogens. Chem. Biol. Interact.
1283-1291. 68, 189-202.
10
Human Cytochrome P450 Enzymes
R Peter Guengerich

1. Background and History of human P450s lAl (ref [6]), 1A2 (ref [7]), 2A6
(ref [8]), 2C9 (ref [9]), 2D6 (refs [7], [10], [11]),
Development of the Field and 3A4 (ref [12]), were isolated in this general
manner. Another general approach that was used
Much of P450 research has always been done was purification from tissue on the basis of
with the view of appUcation to humans, even immunochemical cross-reactivity with animal
when done with experimental animals and P450si3-i5.
microorganisms. Research with the human P450s With the development of recombinant DNA
has been done in several stages, and clinical phar- technology, cDNAs for many of the human P450s
macology has utilized the knowledge at each point were cloned in the 1980s^^ In the late 1980s,
in its development. methods came into use for the heterologous
Aside from in vivo experiments with drugs, expression of P450s, first in mammalian and yeast
human P450 work in the late 1960s and 1970s was systems, and then in baculovirus and (by the mid-
done with tissue samples, primarily biopsies. Some 1990s) in bacterial systems^^^^. In the late 1980s,
data on metabolic patterns and rates were col- the nomenclature system developed by Nebert^^
lected^. In the late 1970s, several groups began to was applied and allowed individual human and
purify P450s from human liver microsomes. The other P450s to be discussed on the basis of their
first purified proteins, selected because of their sequences. (For a guide to some of the earlier
abundance and ease of purification, were probably nomenclature, see ref [22].)
what are recognized now as P450 3A and 2C sub-
By the early 1990s, much of the interest in
family proteins^^. Efforts were shifted to purifying
P450 research had shifted to the human P450
individual P450s on the basis of catalytic activities
enzymes because of the availability of systems for
with the evidence that in some cases a single P450
handling these. In particular, studies in the areas
could be identified in this way; for example, the
of drug metabolism and chemical toxicology/car-
enzyme now known as P450 2D6 was found to be
cinogenesis were facilitated by the knowledge that
under monogenic control^. The approach is techni-
a relatively small number of the P450s account for
cally demanding because of the need to do separa-
a large fraction of the metabolism of the drugs and
tions in the presence of detergents and then remove
other chemicals of interest. In the pharmaceutical
them from individual chromatography fractions
industry, the roles of the major hepatic P450s are
prior to analysis of catal3^ic activity. Nevertheless,
extensively studied in developing predictions

F. Peter Guengerich • Department of Biochemistry and Center in Molecular Toxicology, Vanderbilt University
School of Medicine, 638 Robinson Research Building, Nashville, TN.
Cytochrome P450: Structure, Mechanism, and Biochemistry, 3e, edited by Paul R. Ortiz de Montellano
Kluwer Academic / Plenum Publishers, New York, 2005.

377
378 F. Peter Guengerich

about bioavailability, drug-drug interactions, and utmem.edu/CytochromeP450.html). Much of the


toxicity. Since the publication of the chapter on progress since 1995 has involved extrahepatic
human P450s in the last edition of this book^^, P450s, many with roles in the processing of
apparently all of the remaining human P450 genes "endobiotic" chemicals, for example, sterols and
have been identified, and the number (57) appears vitamins.
to be complete because of more general knowl- The list of the 57 human P450s is presented in
edge about the human genome (html://dmelson. Table 10.1, along with available knowledge about

Table 10.1. Human P450s

Subcellular
P450 Tissue sites localization'^ Typical reaction^

lAl Lung, several ER Benzo[a]pyrene 3-hydroxylation


extrahepatic sites,
peripheral blood cells
1A2 Liver ER Caffeine TV^-demethylation

IBl Many extrahepatic ER 17(3-Estradiol 4-hydroxylation


sites, including lung
and kidney
2A6 Liver, lung, and ER Coumarin 7-hydroxylation
several extrahepatic sites
2A7 ER
2A13 Nasal tissue ER Activation of
4-(methylnitrosamino)-1 -
(3-pyridyl)-1 -butanone
(NNK)
2B6 Liver, lung ER (iS)-Mephenytoin A^-demethylation
2C8 Liver ER Taxol 6a-hydroxylation
2C9 Liver ER Tobutamine methyl hydroxylation
2C18 Liver ER
2C19 Liver ER (5)-Mephenytoin 4'-hydroxylation
2D6 Liver ER^ Debrisoquine 4-hydroxylation
2E1 Liver, lung, other ER Chlorzoxazone
tissues 6-hydroxylation
2F1 Lung ER 3-Methylindole activation
2J2 Lung ER Arachidonic acid oxidations
2R1
2S1 Lung ER
2U1
2W1
3A4 Liver, small intestine ER Testosterone 6p-hydroxylation
3A5 Liver, lung ER Testosterone 6 p-hydroxylation
3A7 Fetal liver ER Testosterone 6 p-hydroxylation
3A43 (mRNA detected in gonads) (ER)

4A11 Liver ER Fatty acid w-hydroxylation


4A22 ER
4B1 Lung ER Laurie acid w-hydroxylation
4F2 Liver ER Leukotriene B^ w-hydroxylation
4F3 Neutrophils ER Leukotriene B^ w-hydroxylation
4F8 Seminal vesicles ER Prostaglandin a)-2 hydroxylation
Human P450s 379

Table 10.1. Continued

Subcellular
P450 Tissue sites localization" Typical reaction*

4F11 Liver ER
4F12 Liver ER Arachidonic acid ci)-,a)-2-hydroxylation
4F22
4V2
4X1
4Z1
5A1 Platelets ER Thromboxane A^ synthase reaction
7A1 Liver ER Cholesterol 7a-hydroxylation
7B1 Brain ER Dehydroepiandrosterone
7a-hydroxylation
8A1 Aorta, others ER Prostacyclin synthase reaction
8B1 Liver ER 7a-hydroxyprogesterone
12-hydroxylation (?)
llAl Adrenals, other Mit Cholesterol side-chain
steroidogenic tissues cleavage
llBl Adrenals Mit 11-Deoxycortisol 11-hydroxylation
11B2 Adrenals Mit Corticosterone 18-hydroxylation
17A1 Steroidogenic tissues ER Steroid 17a-hydroxylation
19A1 Steroidogenic tissues, ER Androgen aromatization
adipose, brain
20A1
21A2 Steroidogenic tissues ER 17-Hydroxyprogesterone
21-hydroxylation
24A1 Kidney Mit 25-Hydroxyvitamin D3 24-hydroxylation
26A1 Several ER Retinoic acid 4-hydroxylation
26B1 Brain ER Retinoic acid 4-hydroxylation
26C1 (ER?)
27A1 Liver Mit Sterol 27-hydroxylation
27B1 Kidney Mit Vitamin D3 1-hydroxylation
27C1
39A1 Liver (?) ER 24-Hydroxycholesterol 7-hydroxylase
46A1 Brain ER Cholesterol 24-hydroxylation (?)
51A1 Liver, testes ER Lanosterol 14a-demethylation

^ER = endoplasmic reticulum (microsomal), Mit = mitochondria.


^If known.
•^Mainly ER, some detected in mitochondria.

sites of tissue expression, subcellular localization, appear to be expressed primarily in the endoplas-
and a typical reaction. In the 1995 edition^^, the mic reticulum and only 6 are located exclusively
list included 31 human P450s, and 2 of these have in mitochondria. (However, work in animal mod-
been dropped as apparent cloning artifacts (2C10, els by Avadhani has clearly demonstrated the
2C17), leaving 29. Thus, the list was only half import of what have been generally considered
complete at that time. It should be emphasized microsomal P450s into mitochondria^"^' ^^. The
that we still have little knowledge about the roles basis of this transport appears to be signals in the
of some of these P450s beyond the genomic infor- N-terminal region^^. Limited information is avail-
mation. Of the 57, most that have been examined able about this phenomenon with human P450s;
380 F. Peter Guengerich

recent collaborative experiments with Avadhani's vary considerably (Figures 10.1 and 10.2).
group indicate that many human liver samples Deficiencies in the expression or catalytic activities
contain immunochemically detectable and catalyt- of most of the P450s involved in steroid metabohsm
ically active P450 2D6 in mitochondrial as well lead to serious diseases (Table 10.3) (included in
as microsomal fractions pST. Avadhani and this hst^^ are P450s IBl [true function unknown]
RR Guengerich, unpublished results].) In many and 24A1 [vitamin D hydroxylation]). In one case,
cases, no direct information is available because a high level of P450 activity (P450 19) can be a
protein studies have not been done, although most problem (estrogen formation in estrogen-dependent
of these "orphan" P450s are predicted to reside tumors) and this P450 is a target for therapeutic
primarily in the endoplasmic reticulum. attenuation^^. The 15 identified xenobiotic-metabo-
Of the P450s with known catalytic activities, 14 lizing P450 are mainly in the families 1-3, and lev-
are clearly involved in steroidogenesis, 4 are els of these vary considerably in humans (Figures
involved in what appear to be important aspects of 10.1 and 10.2). Studies with transgenic (knockout)
metabolism of vitamins (vitamins A and D), 5 are mice do not indicate critical function associated
involved in eicosanoid metabolism, 4 appear to have with the apparent orthologs in the absence of xeno-
fatty acids as their substrates, and 15 catalyze trans- biotic challenge^ ^ Analysis of the lists of drugs in
formation of xenobiotic chemicals (Table 10.2). which individual human P450s are involved^^ indi-
Some of these categories should be considered ten- cates that —75% of the drugs can be oxidized by
tative. This classification accounts for only 42 of the 3 P450s (3A4, 2D6, 2C9), and a set of 6-7 P450s
57 P450s. Many of the xenobiotic-metabolizing will account for 90-95% of all drug metabolism
P450s can also catalyze oxidation of steroids and (Figure 10.3)^^. Similar numbers of P450s are
fatty acids, but these functions do not appear to be involved in the metabolism of chemical carcino-
critical to homeostasis (e.g., testosterone 6p-hydro- gens, although the pattern shifts from that of Figure
xylation by P450 3A4, lauric acid 11-hydroxylation 10.3, with P450s 2C19 and 2D6 being replaced by
by P450 2E1, possibly 17p-estradiol 4-hydroxyla- P450S lAl, IBl, 2A6, and 2E1 (Table 10.4). The
tion by P450 IBl). Most of the steroid-oxidizing relative contributions of these xenobiotic-metaboliz-
enzymes are critical, and the levels of these P450s ing P450s are, to some extent, a function of the rel-
are relatively invariable among individuals, in con- ative abundance (Table 10.5, Figure 10.4), although
trast to the xenobiotic-metabolizing P450s, which there are some important exceptions. Further, the

Table 10.2. Classification of Human P450s based on Major Substrate Class""

Sterols Xenobiotics Fatty acids Eicosanoids Vitamins Unknown

IBl lAl 2J2 4F2 24 2A7


7A1 1A2 4A11 4F3 26A1 2R1
7B1 2A6 4B1 4F8 26B1 2S1
8B1 2A13 4F12 5A1 27B1 2U1
llAl 2B6 8A1 2W1
llBl 2C8 3A43
11B2 2C9 4A22
17 2C18 4F11
19 2C19 4F22
21A2 2D6 4V2
27 A1 2E1 4X1
39 2F1 4Z1
46 3A4 20
51 3A5 26C1
3A7 27C1

''As pointed out in the text, this classification is somewhat arbitrary, for example, P450s IBl and 27A1
could be grouped in two different categories.
Human P450s 381

uoi)e|AxojpAg-,|. |0|ejn^ng

•§ eei
0) ^
^^^^^^H ozv
zzi
g
^^^^^1
^^^^Hjm ezi-
931.
uojiepjxo 3U|djpa^!N I^
CO ^
o>
T—
1
'
8U CO -g
O
CM
^HH^^^HHIH^ 9 U
^^^^^^^^^^^^^H 9U
oLO ^^^"^^^jj^^^^^S* zu
Q. IHHl 01-1-
lull 601.
801-
^^^^^^^9 lOi

^^^^^^^5
^•[
001.
86
68
O O
o
CO
CM
d
d
uojieiAxojpAii-,^ ujO)Au3Ljda|/\|-(s)

uo|)e|AxojpAi|-9 auozexozjomo

ri ^
_o o
"Si
o
> ^
r^
-S CO

X!
o
uo!)eiAi|)adp-o uj^mosajAxoiii^-z 1— s
a
3
>.2 (1>
o> '^
o
382 F. Peter Guengerich

^ 2
1 •• j,„ — , , , , ,
Is
w O
< <| < < < <| <i <
-D
<A
t/) PH

< <
CM CO
o • • • 1 • • •• • • o
in
1
IT) • • • • ^ •••
GL
• • 1•••• •• Q.

D
o
D
1 l_ -1- 1 ..._., 1 . , , 1 1 ..,.

in o o o o
d <0 'sf CM

r 1 1 ' 1 ' 1

< < < \< << < <C 1 << <

< < < <S<< < Q ^ < < B


CM
<< «€iei^< <W<R1

CM m a m iDDa aa • • u. .2 «
o U> B OH

D a • DO D • DDDXCDn Q. •• •
o 3
0. "oS
D ^ >
D Z

1 • 1 - J 1 J 1
1
CM 00 ^ O
>
O
as
c .a

r—T '— 1 — 1 1 1 j '> c c3


T3 (U -^
II op 13
fi -s
"D ^o C/3 O
<< < <$i«i <cmf <m <>5 d N
CM s >
JJ
o • • m • • •• • •• u.
to TS

P ~^-^ t/5

D
T3 ^ S
_ 1 - I .......1 • J
>
O ^ -S
s T3
(U
43 2
^
^
S f^
o
u.|
-D S
s cd (U
0)
CX 00
UL
o
1/3
^1
2
2
o O
c
o V / "O

u
p^ c3 cd
o o o fa
CO CM 1- o
U O b
09l7d |B»01 o/o
uia^ojd leuiosojojui Bui/os^d puiu CM 5 ^
O
T1 iS
£
3
SF2

il 1^
Human P450s 383

Table 10.3. Diseases Associated with Mutations in CYP Genes^^

Gene Disorder

CYPIBI Primary congenital glaucoma (buphthalmos)


CYP4A, 4B^ Defects in salt metabolism, water balance leading to arterial hypertension
CYP5A1, 8A1 Defects leading to clotting and inflammatory disorders, coronary artery disease, and
pulmonary hypertension
CYP7A1 Hypercholesterolemia, resistance to statin drugs
CYP7B1 Severe hyperoxysterolemia and neonatal liver disease
CYPUAl Lipoid adrenal hyperplasia; occasional congenital adrenal hyperplasia (CAH)
CYPllBl Occasional CAH
CYP11B2 Corticosterone methyloxidase deficiency type I, or type II; occasional CAH
CYPllBl, 11B2 Chimeric enzymes causing glucocorticoid-remediable aldosteronism; occasional CAH
CYPUAl Mineralocorticoid excess syndromes, glucocorticoid, and sex hormone deficiencies;
association with increased risk of prostate cancer and benign prostatic hypertrophy;
occasional CAH
CYP19A1 Loss of function: virilization of females, hypervirilization of males, occasional CAH; gain
of function: gynecomastia in young males
CYP21A2 >90%ofallCAH
CYP24A1^ Hypervitaminosis D
CYP27A1 Cerebrotendinous xanthomatosis
CYP27B1 Vitamin D-dependent rickets type I

''Strong evidence of disease in animal models but not yet in clinical studies.

2. General Issues of Variability


and Polymorphism

Variability in patterns of drug metabolism has


been recognized for some time, even before the
discovery of P450s. For instance, the phenomenon
of pharmacogenetic variation had been identified
1A1 by the 1950s^^' ^^ and the early work of Remmer"^^
showed the influence of barbiturates upon drug
metabolism. Further, a number of congenital
defects in steroid metabolism were known and
some could be attributed to alterations in specific
hydroxylations'*^ Much of the subsequent work
on inducibility has been done in experimental ani-
mal models"^^ and later, cell culture.
2C19 In the 1960s and 1970s, a number of accounts
Figure 10.3. Estimated contributions of individual appeared describing variations in rates of metabo-
human P450s to the metabolism of all drugs, based upon lism of drugs in human liver biopsy samples^ The
literature available (adapted from Evans and Relling)^^. first characterization of a monogenic variability in
a human drug-metabolizing P450 was the work of
Smith with debrisoquine^, which was paralleled
patterns reported for human liver (Table 10.5, by the work of Dengler and Eichelbaum on
Figures 10.1, 10.2, and 10.4) do not necessarily sparteine"*^. This polymorphism was first described
apply in other tissues, some of which may be targets in the context of extensive metabolizers (EMs) and
for carcinogens and other toxicants^^. poor metabolizers (PMs) (Figure 10.5). These
384 F. Peter Guengerich

u
o

.-o-I t i l l
.5 .S 2 >. ^
.2 b <u J^

bo^ m
X ^ g ^ o 6 ^ i^ 2
o s^
(U OH

^^
Xi
r'^

H
o
O)
w-i

a
(D
<L>
C/3 T—1

w
^—^ rs|
^
ly,
<==>
u^
a
S
W) ^ CIH
o
.s
u

II 11
t 5> 2 t £
( D O , - — <D ON

4 -^ Z 4 -^

e / S
1
^ 4, ^
00^

6 16 O
3
T3 PU en
1c
rT
1 < c

•S N 6
•c
OH £
a
g 'B t 0)
c
<u
o
o
^ .s
o e s OH
o c
'Jo
it
1
O 6 6 o o o
c c c c 43
c
c N
S rs» "s 6 'o 'B •g "? < ^
•g •g t5
<1 < c
< 1 < 4 < <
<N 1
(N

o o /•

CL|
S

T3
c
cd o
-e
cd
S
PH
o 2^ O
C ^ N

>. 43O 6 1 ^•£a.


rQ
^
13 c
N
'>>
C
'o < O. i n
PQ o.
Human P450s 385

^II
P +1 ^J"

+1 +1
o

:3

u +1 ^ +1 2

u +1 ^

w
0^
OQ +1 :^ +1 p +1 ^
00 o ^ O

'B
+1 ::: +1 ^ +1 ^
o ^^

d
•^ n
12 +1 o +1 o +1 o

•I ^

C «^

U
II
386 F. Peter Guengerich

Substrates
Nifedipine
Mephenytoin Tolbutamide Midazolam Caffeine Debrisoquine
Omeprazole Phenytoin Erytfiromycin Theophyline Sparteine
Coumarin Warfarin Cyclosporin Tacrine Chlorzoxazone

Inhibitors Fluconazole Disulfiram Quinidine


Methoxsalen Sulfaphenazole

Inducers Barbiturates Barbiturates Barbiturates Omeprazole


Rifampicin Rifampicin Rifampicin Tobacco smoke Ethanol
Dexamethasone Isoniazid
Carbamazepine

Figure 10.4. A summary of major human P450s involved in drug metabolism, including major substrates,
inhibitors, and inducers (adapted from Breimer)"^^' ^^. The sizes of the circles indicate the approximate mean
percentages of the total hepatic P450 attributed to each P450 (See also Figure 10.1 and Table 10.5). The overlap of
the circles is to make the point that overlap of catalytic action is often observed, although the overlap does not
necessarily refer to the indicated substrates (or inhibitors).

25

20

^ 15
c
0)

¥ 10 h

5 h

-20 -15 - 1 0 - 5 0 5 10 15 20

log-io Metabolic ratio

Figure 10.5. Frequency distribution histogram of (in vivo) debrisoquine 4-hydroxylation in a Caucasian
population'*'*. The metabolic ratio is the ratio of debrisoquine/4-hydroxydebrisoquine in the urine of individuals
who were administered debrisoquine (10 mg free base) 8 hr previously. The groups are designated PM (poor
metabolizers, solid bars) and EM (extensive metabolizers, gray bars). The group labeled "Ultra" is from retrospective
research"^^ and probably represents gene duplication.
Human P450s 387

polymorphisms were first studied at the level of Several allelic variants clearly lead to the PM
phenotype, that is, pharmacokinetics and in some phenotype, for a variety of reasons. A relatively rare
cases unusual responses to drugs due to reduced case is a gene deletion (*5)^^. The most common
metabolism"*^. The area of pharmacogenetics (now (Caucasian) PM phenotype is an SNP that leads to
also known as—or expanded to—"pharmaco- aberrant RNA splicing (i.e., in splice site) and no
genomics") was facilitated by the identification of mRNA or protein. Other alleles involve partial
the P450 enzymes involved in the drug metabolism deletions, frameshifts, and coding for protein with
phenotypes, and particularly by the development of either intrinsically low catalytic activity or instabil-
molecular biology, which allows the precise charac- ity (reduced halflife). These general patterns have
terization of genetic differences between individu- been seen in other P450s (and other genes). In addi-
als. The majority of the allelic differences are single tion to the EM and PM phenotypes, there is also an
nucleotide polymorphisms (SNPs), or single base "ultrarapid metabolizer" phenotype, due to gene
changes. As anticipated from previous knowledge of duplication. A Swedish family has been identified
pharmacoethnicity, many of these SNPs and poly- with 13 gene copies, leading to 13 times more
morphisms show racial linkage. (A polymorphism enzyme"^^. The level of hepatic P450 2D6 and a
is generally defined as a 1% frequency of an allelic parameter of in vivo debrisoquine metabolism (the
variant in a population; below this frequency, the urinary metabolic ratio = urinary debrisoquine/
terms "rare genetic trait" or "rare allele" are applied 4-hydroxydebrisoquine) vary ~10^ fold among
or, in the case of a very detrimental allele, a mutant people (Figure 10.5). With P450 2D6, and several
or "inborn error of metabolism.") other P450s, the alleles describing the high and low
The debrisoquine polymorphism is now under- levels of metabolism have been described, but the
stood in terms of P450 2D6 and has been a proto- kinetic parameters for many of the alleles have not
type for research in this area. The characterization of been determined by heterologous expression and
the gene^^ led to a basic understanding of the PM measurement of catalytic activity. This is still the
phenotype. The incidence of the PM phenotype is general case with most of the human P450s. P450
about 7% in most Northern European populations, 2D6 is regulated by a hepatic nuclear factor (HNF)
with different phenotypic incidence (and SNPs) in element^^, but is not considered to be inducible by
other racial groups'*"^' '^^~^^. More than 70 allelic xenobiotics. With many other P450s, there is regu-
variants are now known, and 98% of the PMs in lation and variability due to noncoding region
Northern European populations can be accounted SNPs, levels of inducers consumed, and inter-
for by four variant alleles^^' ^^ A nomenclature sys- actions between P450s and transporters, such as
tem has been set up for P450 alleles (using the P-glycoprotein^"^' ^^, may influence the phenotype.
suffixes *1, *2, *3,...) and is maintained by Although the level of P450 2D6 may have a
Oscarson at http://www.imm.ki.se/Cypalleles/. dramatic effect on the metabolism of certain drugs

Table 10.6. Some Major Inducers of Human P450 Enzymes

Class of inducers Some sources Example P450s induced"

Ah ligands Tobacco, broiled Polychlorinated lAl, 1A2


meat, accidental biphenyls
exposures
Barbiturates and similar Drugs, some Diphenylhydantoin 2C, 3A4
compounds polyhalogenated
biphenyls, DDT
PXR ligands Some steroids and Rifampicin 3A4
antibiotics, other
drugs
P450 2E1 inducers Ethanol, isomiazid Ethanol 2E1

^Based on in vivo responses.


388 F. Peter Guengerich

E
(movement
to nucleus?)

BR JR' J , P450 gene


P450gene \~ X^ ^
DNA DNA ^ — ^ ^ — ^
t •
RNA pol
(increased access
to promoter, start site)

Increased transcription

Figure 10.6. Generalized model for regulation of P450 genes by induction. L = ligand, R = receptor,
R' = partner protein for heterodimer of R, Coactiv = coactivator, RNA pol = RNA polymerase.

(Figure 10.5), no other biological changes have seen in humans, they tend to be relatively soon
been reported in PMs. This appears to be the after birth (e.g., P450 3A4, 3A7, see refs [58],
general case for many of the hepatic P450s prima- [59]), and changes in expression seen in the
rily involved in the metabolism of xenobiotics, elderly have not been very dramatic^^"^^.
and few observable physiological effects have
been reported in transgenic mice in which these
genes have been deleted^ ^. As pointed out earlier,
however, deficiencies in some of the steroid-
3. Approaches to Defining
hydroxylating P450s can be very debilitating or Catalytic Specificity of
lethal"* ^ In general, the variation in the levels Human P450s
of these "more critical" P450s is limited in most
of the population, compared to the xenobiotic- Knowledge of the roles of individual P450s in
metabolizing P450s in which an order of magni- specific reactions is critical in the application of
tude variation is not unusual^^. P450 biochemistry to practical issues in drug
The influence of inducers on the expression of metabolism. Originally some of the P450s were
each P450 will be mentioned later (Table 10.6, purified on the basis of their catalytic activities
Figure 10.6). It should be pointed out that several toward certain specific drugs^' ^' "' ^^^ i^y^ gy^j^ jj^
of the P450s can be downregulated by cytokines, those cases, there are the issues of the extent of
and the result has practical significance in the contribution of that form and the involvement of
impairment of drug metabolism in individuals that P450 in other reactions, particularly with new
with colds or flu, or who have received vaccina- drugs. Identification of the individual P450s
tions^^. Another general point to make is that, in contributing to the metabolism of a new drug can-
contrast to some animal models (see Chapter 8)^^, didate is routinely done in the pharmaceutical
human P450 expression shows little if any gender industry. This information is usually required by
differences. When developmental differences are the Food and Drug Administration at the time of
Human P450s 389

application. Identifying P450s involved in oxida- a titration approach (concentration dependence)


tion is important in predicting drug-drug interac- has merit^^.
tions and the extent of variation in bioavailability. Another general issue is the selection of a protein
In general, it is desirable to develop drugs for concentration. Microsomal proteins can bind drugs
which several P450s have a contribution to metab- in a nonselective manner and effectively lower
olism. Drug candidates that are metabolized the free concentration of substrate or inhibitor^^' ^^,
exclusively by a highly polymorphic P450 (e.g., which can influence the interpretation of results.
2D6, 2C19) are usually dropped from further Another point is that the concentration of the P450 of
development. interest should be less than that of the drug and the
A combination of methods involving the use of inhibitor, in order for the basic assumptions about
human tissues and recombinant human P450s is steady-state kinetics to apply (and for the reaction to
usually used to identify P450s involved in a particu- remain linear during the incubation time, although
lar reaction, using an approach outlined earlier^^' ^^. some of the inhibitors are mechanism-based and the
A combination of the following methods is usually loss of activity will be time dependent, requiring pre-
done, not necessarily in a particular order. Lu^^ has incubation). A corollary of these latter points, which
recently reviewed these approaches. also apply to the other approaches that follow, is that
having a very sensitive assay method is very desir-
able. Thus, methods such as HPLC/fluorescence and
particularly HPLC/ mass spectrometry have gained
3.1. Inhibitors
popularity.
The reaction is demonstrated in NADPH- Finally, the choice of an organic solvent is an
fortified human liver microsomes (if the reaction issue. Ideally the substrate should be dissolved in
of interest is restricted to another tissue, then this H2O or very little organic solvent, but this may not
tissue would be used instead). The effects of selec- be possible with many drugs. Several examina-
tive inhibitors on the reaction are examined. A list tions of the effects of individual solvents on
of some of the inhibitors that have been used is human P450s have been published^^' ^^.
presented elsewhere in this volume by Correia In principle, the extent of inhibition of a reac-
(Chapter 7)64,65 tion by a P450-selective inhibitor indicates the
The choice of concentration parameters is fraction of that reaction attributable to that P450.
important in this and some other approaches. For instance, if a 1 |xM concentration of quinidine
Ideally the effect of the substrate concentration on (a P450 2D6 inhibitor) inhibits 50% of a reaction,
the rate of catalytic activity should be determined in then 50% of that reaction may be attributed to
the absence of inhibitor to determine F _ and K P450 2D6. If one desires a more global view than
parameters. If this information is available, the inhi- within a single liver sample, then a pooled set of
bition experiments are best done with a concentra- microsomes (e.g., from 10 samples, balanced on
tion of substrate at or below the K. in order to the basis of liver weight or protein) may be used for
m'
the inhibition assays. However, if one desires to
observe the effect of the inhibitor on the ratio examine the differences among individuals in terms
V^^JK^, which is the parameter usually most rele- of the contribution of a P450, then doing several
vant to human drug metabolism. If the F ^^ and K experiments with individual liver samples is the
o max m
information is not available, an alternative is to approach to use.
select a substrate concentration near that expected
for the in vivo plasma concentration (C ^ ^ or less).
With regard to inhibitor concentration, ideally 3.2. Correlations
a range of concentrations would be used. However,
if a single concentration of the diagnostic inhibitor Another approach with a set of human tissue
is used, it must be selected on the basis of previous microsomal samples is to measure the new reac-
literature because nonselective effects are often tion of interest in each and attempt correlation
observed. For instance, a-naphthoflavone (otNF) with rates of marker activities (for individual
(5,6-benzoflavone) can inhibit P450s other than P450s). Lists are also published in this volume in
1A2 at high concentrations^^ and azoles inhibit Chapter 7 by Correia^^ and elsewhere^ ^.
many P450s at higher concentrations^"^' ^^. Use of
390 F. Peter Guengerich

Correlation can be done by plotting the spe- the antibody raised against the P450. (c) The anti-
cific activity for the new reaction vs the marker body should be shown not to inhibit a reaction
reaction (Figure 10.7). In principle, the correlation known to be attributable to other P450s. Immuno-
coefficient r^ estimates the fraction of the vari- globulin G fractions are generally preferred in that
ance attributable to the relationship between the they produce less nonspecific inhibition than crude
two activities, that is, the fraction of the activity preparations such as sera. Polyclonal antibodies can
catalyzed by the particular enzyme (assuming that vary in their specificity and titer from one animal to
all of the marker activity is catalyzed by this another and from one bleed to another, so constant
enzyme). In some cases, excellent correlations properties cannot necessarily be assumed. In prin-
have been reported^^' ^^. An alternative method of ciple, monoclonal antibodies and antibodies eluted
analysis is the Spearman rank plot, which has from phage display libraries should not vary,
some deficiencies but avoids the overweighting of although this has not always been the case with
unusually high or low values^"^. monoclonals.
Although the approach works well when high In general, antibodies are often selective for
correlation coefficients are generated, the method individual P450 subfamilies, for example, lA vs
is less usefiil when several P450s contribute to a IB vs 2A vs 2B vs 2C, etc., but cross-reaction
reaction, that is, r^ < 0.4. The results should, in all among families can be detected, and in some
cases, be considered in the context of results cases the (P450) sites of cross-reactivity have
obtained with other approaches. been identified^^. Achieving selectivity among
individual P450 subfamily members (e.g., P450
3A4 vs 3A5 vs 3A7) is more difficult. With poly-
clonal antibodies, this can be achieved by cross-
3.3. Antibody Inhibition
adsorption^^; with monoclonals and phage display
The points raised in the Section 3.1, Inhibitors, libraries, this can be done by selection. The point
apply to antibodies as well. Antibodies are used to should be made that any selectivity demonstrated
inhibit activities in human liver (or other tissue) among classes of animal P450s (e.g., rat P450
microsomes and are of several general types: families) cannot be assumed to carry over to
(a) polyclonal antibodies raised against purified human P450s.
animal P450s, (b) polyclonal antibodies raised Antipeptide antibodies have become popular
against purified human P450s, (c) monoclonal in recent years and have two major advantages:
antibodies raised against purified human P450s, (a) peptides can be synthesized and readily puri-
(d) polyclonal antibodies raised against peptide fied by HPLC, avoiding the need to express and
fragments of P450s, and (e) antibody phage dis- rigorously purify P450 proteins (although demon-
play library antibodies selected for recognition of stration of purity by HPLC, capillary electro-
individual P450s. phoresis, and mass spectrometry is still in order),
At this time, almost all antibodies raised against and (b) peptides can be selected for use as anti-
intact P450s have been generated using recombi- gens by sequence comparisons, favoring specific
nant P450s (or against peptides), in contrast to regions.
early work in the field with P450s isolated from Phage display antibody libraries are relatively
liver. Another point to make is that not all antibod- new and have been used in a few P450 applica-
ies inhibit catalytic activity. Further, specificity in tions to date (D.S. Keeney personal communica-
one immunochemical assay (e.g., electrophoretic/ tion). These have a number of advantages,
immunoblotting) does not necessarily implicate including potential selectivity due to the large
specificity in another (immunoinhibition). number of potential antibodies in libraries, the
Three points should be made in designing ability to avoid animal protocols, the immediate
immunoinhibition experiments, (a) The concen- availability of libraries (as opposed to waiting on
tration of antibody should be varied and increased animals to develop antibodies), the consistency of
to the point where the extent of inhibition is con- reproduction of the proteins propagated in bacter-
stant, (b) A nonimmune antibody should be used ial systems, and the ability to include a second
as a control, using the same concentrations as with "epitope tag" for recovery, etc.
Human P450s 391

to

uo!)e|AxojpAq-,|. |0|ejn^ng c
•• T"' o
Q.
« ] "5
£
o
(0
o o
o
r i • **r\« • • ] 1
CD
E
o

[_
O
-• 1

00 CD -^ CJ
E
Q.

0)
B B
uoj)e|AxojpAM-z uueuinoo co
o
oin
Q.

oIf) uo!)e|AL|)8ep-o uueuinooAxoL|)3-z o a


'St

0.
^ C3

13 o
o (Z)
^o
W)
c
o (L)
'rrt
?^ <CU3
^
t:
o
U W)
C
uoi)e|Ai|)aep-o uijnjosajAxomg-z 1^
cr>

o ^
TS
T- C

3
o> (U
U . TS
392 F. Peter Guengerich

3.4. Demonstration of Reaction (and the number is even higher if one moves the
with Recombinant P450 esterase and epoxide hydrolase reactions to the
Phase II group because they are not involved in
In early work in this field, this point would redox reactions). Constructing a figure of this type
have been the demonstration of the reaction of can be somewhat misleading in that the contribu-
interest with an enzyme purified from tissue. tion of each P450 is more difficult to evaluate
Today P450 proteins are generally produced in in vivo than in vitro (for a more original tabulation,
recombinant systems and seldom purified from see ref [32]). The large contributions of P450s
tissue sources. In routine practice in the pharma- 3A(4) and 2C9 are driven to a large extent by the
ceutical industry, new reactions are examined with high levels of expression of these two enzymes in
a battery of the major recombinant human (liver) human liver (and small intestine) and to their broad
P450s, many of which are available from com- substrate specificity. The charts do not necessarily
mercial sources. Systems used for expression reflect all drugs currently in development. A cur-
include bacteria, yeast, baculovirus (-infected rent tendency has been the development of larger
insect cells), and mammalian cells. The P450s molecules as drug candidates, in order to achieve
need not be purified for these comparisons but target specificity and affinity, and a general axiom
must have suitable provision for NADPH-P450 is that these are more readily accommodated by
reductase in a crude system (and cytochrome b^ P450s 3A(4) and 2C9. In recent years, pharmaceu-
[b^] in certain cases). tical companies have tried to avoid developing
Usually activity results obtained with several drug candidates that are substrates (or inhibitors)
of the major P450s are compared to each other for the highly polymorphic P450s 2D6 and 2C19.
and to those obtained with tissue microsomes, in With all of these caveats in hand, the allocation of
order to put the work in context. Ideally assays are the chart in Figure 10.3 is probably a good estimate
done at several substrate concentrations and the and may not change considerably in the near
parameters k^^^ (^max) ^^^ ^cat/^m ^^^ obtained. future. However, a point to be made here is that the
These values should be normalized on the basis of metabolism of many drugs is a function not only of
P450 concentration, in that any values based on P450s but also of other enzymes and, as recog-
milligram protein for the expression system can- nized more in recent years, transporters that alter
not be used for comparisons with tissue micro- the concentrations of drugs within cells. A discus-
somes. In principle, the k^^^ (total P450 basis) sion of drug transporters is outside the scope of
should be at least as high for the recombinant this chapter, and the reader is referred elsewhere^ ^
reaction as for the tissue microsomes. A more The subjects of P450 regulation and polymor-
realistic way to make a comparison is to immuno- phism (or mutation in some cases) have already
quantify the amount of the particular P450 in the been mentioned, and will be treated again, with
tissue microsomes and then use this value in cor- individual P450s. At this point, some general
recting the microsomal k^^^ for comparison with practical considerations will be discussed. If one
the recombinant system. The matter of scaling considers the total concentration of P450 in liver
these parameters to generate predicted microso- samples from different healthy individuals (on a
mal (or in vivo) rates from in vitro experiments milligram protein basis), most individuals fall
with recombinant enzymes is not trivial, but a within a range of ~3-fold'. However, when indi-
number of efforts have been made^'^~^^. vidual "drug-metabolizing" P450s (e.g., families
1, 2, 3) are considered, the variation is consider-
able, with 5-10-fold being common and 40-fold
not unusual, for example, P450 1A2 (ref [73]).
4. Relevance of P450s in In Vivo With P450 1A2, a similar variability (40-fold) is
Drug IVIetabolism seen in in vivo caffeine pharmacokinetics^^. With
highly polymorphic enzymes, the variability in the
P450s are the major enzymes involved in same in vivo pharmacokinetic parameters can be
human drug metabolism. In looking at the fraction as much as lO'^-fold (Figure 10.5).
of the number of drugs processed by "Phase I" Two examples of studies of the variability
enzymes (Figure 10.3), P450s account for >80% among individuals are presented in Figure 10.5
Human P450s 393

(Caucasians) and Figure 10.2 (Caucasian and Extensive Metabolizer (EM, normal)
Japanese). Gender has not been shown to have a
major influence on levels of expression of the
major xenobiotic-metabolizing P450s, and inter-
gender pharmacokinetic differences are probably
due to other influences on bioavailability or vol-
Plasma
ume of distribution^^. Racial differences exist due level of
to allelic variations, which may influence either drug
levels of expression or the inherent catal3^ic activ-
ity of the P450s [ref [49]). Some apparent racial
differences are seen here (Figure 10.2) and have
also been reported in in vivo studies (e.g., 3A4
(ref [83]), 2E1 (ref [84])). Controlling diets is an
issue in many in vivo studies of this type, and
in vitro studies can also be affected. In general, the
differences in activities of a given P450 between Time (arrows show repeated doses)
races are much less than within a race (e.g..
Figure 10.2). Finally, the point made above should Figure 10.8. Significance of low metabolism of a
be noted that the levels of the P450s involved in drug by P450s (or other enzymes). A "typical" pattern is
seen in the upper panel (EM), where the plasma level of
steroid metabolism (e.g., families 11, 17, 19, 21)
the drug is maintained in a certain range when a
vary considerably less than do the xenobiotic- particular repetitive dose is prescribed. Unusually slow
metabolizing P450s (families 1, 2, 3), probably metabolism (lower panel, PM) results in an elevated
due to their well-defined roles in regulation of plasma level of the drug. C ^^ = maximum plasma
physiological processes. concentration; AUC = area under the curve.
Many chemicals are capable of inducing
P450s, as clearly demonstrated in animals and population of interest. The plasma concentration
with cell culture systems^^. In vivo induction rises to a peak (C ^^J following the first dose and
experiments with humans are not as readily done then decreases to a lower level prior to the next
as with animals, but ample evidence for P450 dose. With subsequent doses, the plasma concen-
induction is available, going back to the barbitu- tration remains within this region and yields the
rate observations of Remmer in the 1950s^^. A list desired pharmacological effect. Without prior
of some established P450 inducers is presented in knowledge about a problem with this drug, the
Table 10.6. This list is rather conservative in that PM (lower panel of Figure 10.8) would be admin-
only information is included from studies in istered the same doses. Very limited metabolism
which in vivo evidence has been obtained. Many would occur between doses, and the plasma con-
of the studies have involved pharmacokinetics, but centration of the drug (and presumably the con-
some "moderately invasive" studies have involved centration of the drug in the target tissue) will rise
direct measurement of proteins, mRNA, or enzyme to an unexpectedly high level, with an attendant
activities in peripheral blood cells or small intes- increase in the area under the curve (AUC). The
tinal biopsies; liver biopsy data is rare. Table 10.6 simplest effect would be an exaggerated (and
could probably be expanded considerably if all probably undesirable) pharmacological response.
information from in vitro studies were included, One can also imagine a situation in which metab-
for example, P450s IBl and 2S1 are probably olism is more rapid than expected in the typical
inducible by Ah ligands^^' ^^. The major problem in patient, for example, due to gene amplification or
demonstrating human P450 induction in vivo is the enzyme induction. In this case, C ^^^ and AUC
lack of diagnostic pharmacokinetic parameters for would be smaller than in the case of the EM
many ofP450s. (Figure 10.8, upper panel), and decreased drug
The clinical influence of differences in P450 efficacy would be expected.
activity can be rationalized using the scheme of Some practical situations follow. With regard
Figure 10.8. In this model example, the drug doses to polymorphisms, several are known that can
have been developed with the EMs as the general render some drugs dangerous due to toxicity
394 F. Peter Guengerich

(e.g., perhexiline, leading to peripheral neuropa- is not available, it is possible that blocking the
thy due to lack of metabolism by P450 2D6 oxidation of one drug by a P450 could cause it to
(ref. [88]) or can alter the recommended dose (e.g., accumulate and behave as an inhibitor toward
warfarin/P450 2C9 (refs [89-91]) and omepra- another. A potential example would be decreasing
zole/P450 2C19 (refs [92], [93]). Drug interac- the P450 3A4-catalyzed oxidation of quinidine
tions are a serious problem, and pharmacokinetic and having the accumulated drug inhibit P450
interactions have several molecular bases. One 2D6 (ref [106]). P450 induction could result not
is enzyme induction, which usually results in only in decreased oral availability but also in the
decreased bioavailability. The decreased bioavail- enhanced bioactivation of chemicals. This is a
ability of a drug can be the result of induction by general concern with potential carcinogens, as
that same drug or by another drug. A classic discussed in the next section of this chapter, and
example is the decreased bioavailability of the one of the reasons why regulatory agencies have
oral contraceptive 17a-ethynylestradiol following concerns about P450 lA inducers.
treatment of individuals with rifampicin, barbitu- The phenomenon of P450 stimulation has been
rates, or St. John's wort and consequent P450 3A4 studied in some detail in vitro^^^. By stimulation
induction^^' ^^' ^^. Another aspect of drug-drug we mean the enhancement of P450 catalytic activ-
interactions involves P450 inhibition. The inhibi- ity by the direct addition of another compound,
tion can be of a competitive nature, that is, two outside of a cellular environment in which gene
substrates competing for a limiting amount of regulation is involved. Some aspects of P450 stim-
a P450 or a bona fide inhibitor (no enzymatic trans- ulation will be treated under the topic of P450
formation) competing with substrates. An example 3A4 (Section 6.20.4), with which much of the
here is the antihistamine terfenadine, the metabo- work has been done. An open question is whether
lism of which is inhibited by the P450 3A4 such behavior occurs in humans. At least four
inhibitors, erythromycin and ketoconazole. Another pieces of evidence suggest that such behavior is
major type of P450 inhibition is "mechanism- possible: (a) cooperativity has been reported in
based" (or "suicide") inactivation, in which oxida- hepatocyte cultures'^^, (b) an early experiment
tion of a substrate destroys the P450 (refs [64], with neonatal mice (individual P450s unknown)
[96]). An example here is the inactivation of P450 by Conney's group indicated the immediate
3A4 by bergamottin and other flavones found in enhancement of an activity by flavones'^^; (c) the
grapefruit juice^^ ' ^^. work of Slattery and Nelson with rats showed an
In the above cases, the effects have been dis- interaction between caffeine and acetaminophen
cussed only in terms of altered bioavailability; that that implies such behavior''^; and (d) quinidine
is, with increased clearance of 17a-ethynylestradiol, enhanced the in vivo oxidation of diclofenac in
unexpected menstruation and pregnancies have monkeys, in a manner consistent with in vitro
resulted^^' '^*' ^^^. Some of the drug interaction human work'''. If stimulation does occur in vivo,
problems can be more complex, even when the it is a phenomenon that has been very difficult to
analysis is restricted to pharmacokinetic aspects. predict (even in vitro), and in the case of P450
For instance, in the example mentioned above, ter- 3A4 substrates, the situation would probably
fenadine can be considered a prodrug'^^; in most be further complicated by issues involving P-
individuals, the P450 oxidation (followed by fur- glycoprotein behavior (and P-glycoprotein also
ther oxidation) yields fexofenadine, the circulat- shows cooperativity of its own^'^).
ing form of the drug. Low levels of P450 3A4 In the process of drug development, there are
activity (due to inhibition or other reasons) cause three guiding principles to dealing with P450
the accumulation of the parent (prodrug) terfena- metabolism, aside from details of each specific
dine to toxic levels that can cause arrhythmia^^^' ^^'^. case: (a) use in vitro screening to delete com-
Another possibility is that blocking a primary pounds that will have poor bioavailability (i.e.,
route of metabolism of a drug may favor second- rapid in vitro oxidation); (b) use in vitro screens to
ary pathways that lead to toxicity, for example, avoid obvious problems of toxicity, induction, and
blocking phenacetin 0-deethylation (P450 1A2) inhibition; and (c) seek drug candidates in which
can lead to deacetylation, A^-oxygenation, and the metabolism is the result of several different
methemoglobinemia^^^. Although a good example enzymes and not dependent upon a single one,
Human P450s 395

particularly a highly polymorphic P450 (or difficulties in the earlier phases of the work^^^.
another highly polymorphic enzyme). Many of the early problems have been circumvented
with the ability to measure mRNA expression and
the access to DNA sequences. While evidence for
correlation of P450 lAl mRNA expression with
5. Relevance of P450s in lung cancer incidence has been obtained^^^, an unre-
Toxicology and Cancer Risk solved issue is the nature of any genetic variability.
In contrast to the situation seen in mouse models ^^^,
Historically much of the attention given to P450s the allelic variations in the human Ah receptor
has come from the interest in cancer, going back to (which has apparently considerably lower affinity
some of the first demonstrations of redox reactions for many of the ligands of interest than the mouse
in the metabolism of chemical carcinogens^ ^^ and receptor^^^) do not appear to account for inter-
the inducibility of P450s by carcinogens^^. The individual levels of inducibility of P450 1 Al (refs
interest in P450s was also extended to chemical tox- [127], [128]). Kawajiri's laboratory has presented
icities other than cancer with the demonstration of epidemiological evidence for association of lung
bioactivation of compounds such as the drug aceta- cancer incidence with an Mspl polymorphism of
minophen^ ^"^ and the insecticide parathion^^^' ^^^. P450 1 Al (ref [129]). However, these results, from
Many studies have been done with P450 animal studies done with Japanese, have not been repro-
models, particularly using P450 inducers and ducible in Caucasians (refs [130-132]). Further, the
inhibitors and genetically modified mice, either nat- heterologously expressed human P450 lAl allelic
urally occurring or transgenic. These studies provide variant (V462I) showed only a relatively small
strong evidence that alterations in the activities of change in oxidation of the prototype polycyclic aro-
P450s can modify the sensitivity of mice to various matic hydrocarbon carcinogen benzo[a]pyrene
chemicals. For instance, the Ah locus (which con- diolepoxide^^^' ^^'*. A recent explanation to the
trols P450s lAl, 1A2, and IBl as well as some quandary comes from the work of Kamataki's
Phase II enzymes) can modify the sensitivity in Ah group, who have shown that P450 IBl, not P450
receptor-deficient mice, depending upon the chem- lAl, is the major P450 responsible for the aryl
ical and the organ site^^^. Effects of specific P450 hydrocarbon hydroxylation activity in lymphocytes
knockouts have been reported in transgenic mice as and that it is P450 IBl expression that shows the
well, for example, prevention of acetaminophen tox- classic trimodality, not P450 lAl, (ref [135]).
icity by deleting P450 2E1 (ref [118]) and of Today the field is such that the search for roles
7, 12-dimethylbenz[«]anthracene-induced lym- of a particular P450 in human disease follows a
phomas by deletmg P450 IBl (ref [119]). route similar to that just discussed for P450 lAl,
Despite the strong evidence for effects of vari- that is, the identification of SNPs is a basis for epi-
ability of P450 on chemical toxicity and cancer risk demiological associations with various maladies.
in animals and the knowledge that human P450 lev- This approach is commonly applied to the possible
els vary considerably (Figures 10.1, 10.2, 10.5, and roles of P450s in cancers at various organ sites. The
10.7), demonstrating relationships with human general concept is also utilized for other diseases
disease has been difficult. In the 1960s, the demon- and is the major basis for the Environmental
stration of the inducibility of aryl hydrocarbon Genome Project of the National Institute of
hydroxylase (thought to be what is now known as Environmental Health Sciences (which includes
P450 lAl) by Nebert and Gelboin^^o j ^ ^ ^^ ^^^^ many other gene candidates in addition to
investigations with human samples, particularly P450s)^^^. The positive aspects of this strategy are
peripheral blood cells. The work of Shaw and that we have an extensive knowledge base of allelic
Kellerman^^^' ^^^ suggested that the inducibility of variations of P450s (e.g., http://www.imm.ki.se/
aryl hydrocarbon hydroxylase (now recognized as CYPalleles/), sophisticated and very sensitive bio-
P450 1 Al and IBl under these conditions) is corre- logical tools, and the potential to noninvasively
lated with susceptibility of smokers to lung cancer. analyze large populations, at least in the case of
In the early work, this apparently genetic variability some diseases and P450s. On the negative side,
was trimodal. Subsequently, this phenomenon has the ability to rapidly screen for associations
proven difficult to study, in part due to technical without serious thought about chemical exposure
396 F. Peter Guengerich

levels has lead to many studies with little or only In the process of drug development, the induc-
marginal biological plausibility. Many association tion of P450 1 and P450 2B enzymes (in animals
studies have been difficult to repeat. An example or in human cell or reporter assays) has often been
in point is the reported association of attenuated considered an issue for potential toxicity'^^' ^^'.
lung cancer risk (of smokers) with the P450 2D6 The concern about induction is that the rodents
PM phenotype. Although the initial reports were may be likely to develop liver or other tumors in
quite exciting ^^^, subsequent studies yielded vari- cancer bioassays with these compounds, and any
able results, and meta analysis has not supported association between these inductions and human
an association^^^; moreover, no real experimental cancer is not established; for example, epileptics
support for a biological association was ever with long-term exposure to barbiturates and
found^^^. A recent review by Vineis*^^ concludes hydantoins have not been found to have more can-
that the risks of cancer due to genetics are consid- cer'^^. Likewise, the induction of P450 4A is an
erably less than those associated with smoking or indicator of peroxisomal proliferation, a phenom-
other environmental factors. enon associated with rodent liver tumors but prob-
What associations of P450 have been ade- ably not human'^^. Thus, induction of rodent
quately demonstrated? The list below is short and P450s has been shown to be a means of identify-
not intended to necessarily be totally inclusive, but ing types of potential rodent toxicity^ ^'^, some of
emphasizes some of the more positive associa- which may be relevant to humans, but should not
tions found to date. (The absence of several of the be used as evidence for adverse roles of these
steroid-oxidizing P450s is known to be debilitat- agents in humans.
ing [Table 10.3], but these are not treated here; see
the sections on individual P450s and ref [29].)
The possible association between P450 lAl and
lung cancer has already been discussed above; a 6. Individual Human P450
confounding factor may be expression of P450 Enzymes
IBl. Truncation of P450 IBl is associated with
glaucoma, for unknown reasons^^^; this defect has Each of the 57 human P450s will be covered
not been seen in the P450 IBl-knockout mice^'' ^ •^. here. Clearly much more information is available
Allelic variants in P450 IBl do not appear to have about some than others. Points to be covered with
major effects in the oxidation of carcinogens'"^^; each, when possible, include sites of expression
some differences in cancer risk have been reported and relative abundance, polymorphism and
in the epidemiology literature''^^' ''^^. P450 1A2 inducibility, substrates and reactions, knowledge
activity has been reported to be associated with of important residues and active site characteris-
colon cancer incidence, when the factors of N- tics, inhibitors, and clinical issues.
acetyltransferase and well-done meat intake are
considered''^^; an association has plausibility in
the activation of heterocyclic amines by P450 1A2 6.1. P450 1A1
(ref [146]). One of the strongest associations
6.1.1. Sites of Expression and
reported to date involves that of P450 2A6 with
lung cancer; the association is driven by the data
Abundance
obtained with individuals with the gene dele- The gene has seven exons, and the cDNA
tion'^^. A relationship is plausible due to the region is -^70% identical to that of the closest
demonstrated ability of P450 2A6 to activate relative, P450 1A2. P450 lAl is expressed in fetal
A^-nitrosamines (Table 10.4), and possibly via the liver but not at appreciable levels in adult
decreased smoke intake of null-type individuals ljygj.155-157 P45Q lAl can be induced in primary
due to impaired metabolism of nicotine'"^^ (see human hepatocyte cultures^^^. The dominance of
Section 6.4.6). Although many epidemiological P450 1A2 over 1 Al in vivo may be due to prefer-
studies have been done with SNPs of P450 2E1, ential induction of P450 1A2 > lAl at low doses
any putative changes in P450 2E1 phenotype have of inducers (a phenomenon established in rats^^^)
not been validated with in vivo assays and must be or the presence of factors in liver that are not
considered suspect^'*^. preserved in hepatocyte cultures.
Human P450s 397

P450 lAl is expressed in human lung and has to initiate transcription (Figure 10.6, with R = Ah
been partially purified^. A recent estimate of a receptor), R^ = ARNT, and L = TCDD or other
median level of P450 in human lung is 6.5 pmol/mg inducers. A number of details regarding this
microsomal protein (n = 7) and 16 pmol/mg micro- scheme remain to be elucidated, such as roles of
somal protein for smokers (n = 18)^^^. The varia- coactivators, whether an endogenous ligand exists
tion in levels of P450 lAl is very high and if so what it is, etc. The list of inducers
(> 100-fold)^' ^^^, as suggested from earlier work in reported from in vitro studies includes TCDD and
which only benzo[a]pyrene hydroxylation was used is quite long. The list of compounds for which
as an indicator^^^ in vivo evidence of induction is more limited, but
P450 lAl is also expressed in placenta^^^ and it is generally accepted that it includes cigarette
peripheral blood cells (lymphocytes, monocytes)^^^, smoke, heterocyclic amines, polychlorinated
and these tissues have been used in many studies. biphenyls^^^, and some drugs (e.g., omeprazole^^^).
Expression (at least at the mRNA level) has been
reported in a number of other extrahepatic tissues
including pancreas, thymus, prostate, small intes-
tine, colon, uterus, and mammary gland^^"^. 6.1.3. Substrates and Reactions
This enzyme was first explored in the context
of an aryl hydrocarbon hydroxylase, using fluo-
6.1.2. Regulation and Polymorphism rescence assays that measured primarily the
Polymorphism in the inducibility of benzo[fl] 3-hydroxylation of benzo[fl]pyrene^^^. (It should
P3^ene hydroxylation activity has attracted consid- be noted that the fluorescence assay also detects
erable interest following the reports of Shaw and other fluorescent products, for example, 9-hydroxy-
Kellerman^^^' ^^^ that the induction in lymphocytes benzo[a]pyrene, and that other P450s also cat-
of smokers can be associated with susceptibility alyze the 3-hydroxylation reaction, for example,
to lung cancer. The link to lung cancer has been P450 2C9 in human liver^^^.) Another classic
studied extensively but few general conclusions model reaction used for P450 lAl is 7-ethoxyre-
can be reached. Smoking clearly induces levels of sorufin O-deethylation^^^, i73 Human P450 lAl
lungP450 lAl (refs [124], [160], [165]). Some epi- oxidizes benzo[a]p5n-ene to a variety of prod-
demiological investigations link the *2A (Mspl) ucts^^^' ^'^^. Many other polycyclic hydrocarbons
and *2B (I462V) polymorphisms to lung cancer are substrates for P450 1 Al and have been studied
incidence in Japanese^^^, but this association extensively^^^' ^'^'^. Some heterocyclic and aro-
has not been reproducible in other studies with matic amines can also be activated by P450 lAl
Caucasians^^^' ^^^ These two alleles are in linkage (ref. [178]). P450 lAl does not appear to play a
disequilibrium^ ^^. Two studies with recombinant major role in the metabolism of many drugs, pos-
human P450 lAl have not shown a major differ- sibly because of its locations of expression.
ence in any catal3^ic activities due to the substitu-
tion at codon 462 (refs [133], [134]). Although
there is a general consensus that phenotypic
variation in the inducibility of P450 lAl is 6.1.4. Knowledge about Active Site
observed, extensive searches have not associated Relatively little is known about the active site of
the inducibility with any known polymorphisms in P450 lAl. Early work on pharmacophore models
the P450 lAl, Ah receptor, or arylhydrocarbon for rat P450 1 Al was done by Jerina's group^^^. The
nuclear translocator (ARNT) genes ^^^' ^^^. early modeling of substrates and inhibitors sug-
The induction of P450 lAl has been studied gested that P450 lAl ligands were relatively
extensively and is discussed elsewhere in this planar. Some homology modeling has been done by
book^^^. Briefly, the Ah receptor resides in the Lewis^^^, although little has been done with site-
cytosol and, when activated by binding of an appro- directed mutagenesis on the roles of individual
priate agonist, loses the accessory protein Hsp90 amino acids. The lack of effect of interchanging
and dimerizes with the ARNT protein, moving to Val and Leu at position 462 has already been
the nucleus and interacting with an XRE element mentioned^^^' ^^^.
398 F. Peter Guengerich

6.1.5. Inhibitors 6.2.2. Regulation and Polymorphism


Despite the long interest in this enzyme, the The variability and inducibility of P450 1A2
hst of inhibitors is relatively short, and many have been recognized for some time, indirectly,
inhibitors show overlap with P450s 1A2 and IBl going back to studies on phenacetin metabolism
(ref. [181]). For instance, aNF is often used as by Conney and his associates ^^'^. The characteriza-
an inhibitor but is more effective against P450 tion of P450 1A2 CT450p/') as the low K^
1A2 (refs [181], [182]). Another inhibitor is ellip- phenacetin 0-deethylase^ led to some interpreta-
ticine^"^. 1 -(1' -Prop)aiyl)pyrene and 2-( 1 -propynyl) tion of the earlier results. P450 1A2 was shown to
phenanthrene were found to be selective P450 be the caffeine iV^-demethylase^^, and the 40-fold
1 Al inhibitors when compared with human P450s variation in levels of liver P450 1A2 is reflected in
l A 2 a n d l B l (ref [181]). the 40-fold variation in some in vivo parameters of
caffeine metabolism^^, some of Vesells's earlier
work on the metabolism of antipyrine in twins
6.1.6. Clinical Issues suggests a role for genetic polymorphism in P450
Due to a rather limited role of P450 lAl in 1A2 activity^^^, and a more recent twin study
drug metabolism, there are no real pharmacoki- confirms the strong genetic component of caf-
netic issues. The issue with P450 1 Al is induction feine demethylation^^^. However, elucidating
and a possible role in chemical carcinogenesis. details of any functional polymorphism has been
Work with animal models shows that P450 lAl difficult.
inducers can be co-carcinogens^^' ^^^. Thus, regu- At least 13 allelic variants of P450 1A2 are
latory agencies tend to look unfavorably at induc- now known^^. Of these, at least five have changes
tion of P450 lAl by potential drugs in animal in the coding sequences that cause amino acid
models. However, the point should be made that changes. Recent work in this laboratory with the
there is presently little experimental or epidemio- expressed coding region variants indicates that
logical evidence to support this hypothesis, and most do not differ more than 2-fold in their kinetic
Ah inducers can afford protection from cancer in parameters for several assays (phenacetin O-
some animal models^^^. deethylation and A^-hydroxylation of heterocyclic
amines), although one of the variants (R431W)
did not express holoprotein in Escherichia
coli^^^^. Some (*1C and *1F) have been proposed
6.2. P4501A2 to modify levels of expression^^. However, the
basis for a polymorphism is not clear. One view is
6.2.1. Sites of Expression and
that the variability is not genetic, based upon the
Abundance
lack of modality breaks in analysis of some in vivo
As mentioned earlier, human P450s lAl and parameters of caffeine metabolism^^^.
1A2 both have seven exons and 70% sequence One complication with genetic polymorphism,
identity in their coding regions. Both these genes as with P450 lAl (vide supra), is the inducibility.
show similar patterns of regulation by the Ah Because of the availability of markers of hepatic
receptor system, but P450 1A2 is essentially P450 1A2 function (phenacetin is no longer used
expressed only in the liver^^'^, probably due to the but caffeine and theophylline are), demonstrating
involvement of HNF in its regulation (vide infra). in vivo changes in P450 is relatively easy to do and
Several lines of evidence indicate that the level of the effects are consistently seen, at least quantita-
expression is substantial (Figures 10.1 and 10.2, tively. The mechanism of induction appears to be
Table 10.5), - 1 0 - 1 5 % of the total P450, on the similar to that of P450 lAl (Figure 10.6), with
average, with levels varying ~40-fold among expression restricted to the liver because of the
individuals (Figure 10.4). need for HNF (ref [188]). An interesting observa-
Occasional reports cite mRNA expression in tion made recently in mice is that the inducer
some extrahepatic tissues, for example, colon^^^. 3-methylcholanthrene causes a persistent induc-
Extensive searches have not found expression in tion (of P450 lAl) in liver, lasting beyond the
human lung^^"^. time suggested by pharmacokinetic expectations ^^^.
Human P450s 399

One interpretation is that a P450 lA2-generated other P450s, e.g., 3A4 (ref [203])). Induction of
metabolite is involved. Further details and any P450 1A2 and 2-hydroxylation has been proposed
relevance to humans remain to be established. as a means of preventing oxidation of 17(3-
With animal P450 1A2, one mechanism of induc- estradiol to the potentially more reactive 4- and
tion involves protein stabilization, for example, 16a-hydroxy products^^"^' ^^^.
by isosafrole-derived products *^^. Whether or not
this mechanism is relevant in humans is unknown.
Reported inducers include cigarette smoking, 6.2.4. Knowledge about Active Site
charbroiled food (presumably via polycyclic
hydrocarbons and heterocyclic amines), crucifer- Considerable site-directed mutagenesis work
ous vegetables, vigorous exercise^^\ and the drug has been done with rat P450 1A2 (refs [206-208])
omeprazole (actually a metabolite) ^^^. but relatively little with human P450 1A2. Some
pharmacophore^^^ and homology^^^' •^^^ modeling
work has been reported.
6.2.3. Substrates and Reactions An approach was developed in this laboratory
for doing random mutagenesis of P450 1A2, uti-
P450 1A2 has been expressed in a number of
lizing changes in the rates of formation of muta-
systems and is used in analyses of catalytic selec-
genic hydroxylamines^^^ Changing Glu226 (to He
tivity. Of the P450s, this has one of the highest
or Gin) had the effect of increasing rates of
levels of expression in bacterial systems^^^' ^^^.
phenacetin 0-deethylation 8-fold (effect on ^cat)"^^^-
The list of drug substrates is long^^, and only a
Also of interest is a 100-fold decrease of activity
few of the more well-known reactions are listed in
seen in mutation of the neighboring Phe225 (to He
Table 10.7.
or Tyr)^^^. The mutation of Asp320 (to Ala) also
Many carcinogens are substrates, particularly
decreased activity^^^. Kinetic deuterium isotope
aromatic and heterocyclic amines (Table 10.4).
effect studies suggest little change in rate-limiting
Other carcinogens shown to be substrates include
steps of the 0-dealkylation reaction over a wide
polycyclic hydrocarbons, nitropolycyclic hydro-
range of activities with these mutants, and the
carbons, and some A^-nitrosamines^^^.
mutations do not affect ground-state substrate
The only major endogenous substrates are 17p- binding or rates of P450 reduction^^^. The effects
estradiol and estrone (2-hydroxylation). The phys- of these substitutions have not been interpreted in
iological relevance of this reaction is unknown, terms of specific roles.
particularly because of the wide variation in levels
The issue of cooperativity will be discussed
of P450 1A2 (this reaction is also catalyzed by
later under P450 3A4. Cooperativity has not been
reported for human P450 1A2 but behavior of the
rabbit ortholog has been interpreted in the context
Table 10.7. Some Drug Substrates for Human of multiple, overlapping binding sites^^"^.
P450 1A2^

Drug^ References
6.2.5. Inhibitors
Acetaminophen (3') 195 Several human P450 1A2 inhibitors are known
Antipyrine (4,3-methyl) 196 from clinical work, including fiirafylline (mecha-
Bufuralol(l,4) 197 nism based)^^^ and fuvoxamine. aNF is a
Caffeine (3) 73 commercially available and strong inhibitor of
Clozapine 49
human P450 1A2 (K- ~ 6 nM)^^^ for in vitro work.
Olanzapine 49
Ondansetron (7,8) 198
A number of polycyclic acetylenes are potent
Phenacetin 7 inhibitors of P450 1A2 (ref [181]). With rat
Tacrine 199, 200 P450 1A2, 2,3,7,8-tetrachloro[/?]dibenzodioxin
Theophylline (1,3,8) 201 and some polyhalogenated biphenyls are strong
inhibitors, but these studies have not been extended
"See also Rendic^^. to human P450 1A2 (ref [216]).
400 F. Peter Guengerich

6.2.6. Clinical Issues the levels are of an order of magnitude lower than
for P450 lAl (ref [160]). These low values may
Some drug interactions have been reported. An explain the lack of immunostaining in (nontumor)
older example is that of low activity toward tissues reported by Murray et al?^^ Specific val-
phenacetin favoring a potentially toxic secondary ues for levels of expression in tissues other than
pathway, deacetylation followed by quinoneimine lung have not been published. Recently Chang
formation and methemoglobinemia^^^. Furafylline
et alP^ found traces of P450 IBl mRNA in
was a drug candidate but was never developed
human liver using real-time PCR, but the protein
because of its strong P450 1A2 inhibition and
was undetectable within the limit of sensitivity.
interference with caffeine metabolism^^^. High
levels of P450 1A2 activity have also been associ-
ated with ineffectiveness of theophylline therapy
(forasthma)2i8,2i9
6.3.2. Regulation and Polymorphism
The other concern about P450 1A2 is the same Levels of P450 IBl in human lung vary by at
discussed earlier for P450 1 Al, the co-carcinogenic least one order of magnitude ^^^. An interesting
effect. In this regard, there is some epidemiological observation is that a termination variant of P450
evidence that high P450 1A2 activity (measured as IBl is strongly associated with glaucoma^"^^' ^^^.
in vivo caffeine metabolism) is associated with A similar phenotype has not been seen in P450
enhanced risk of colon cancer, although the effect IBl-knockout mice^^^. Other polymorphisms of
was not seen in the absence of high A^-acetyltrans- (human) P450 IBl are known and are predomi-
ferase activity and high consumption of charbroiled nant in a set of haplotypes involving four varia-
meat^"^^. tions Arg/Gly 48, Ala/Ser 119, Val/Leu 432, and
Asn/Ser 453. Assays involving the metabolism of
17p-estradiol and polycyclic hydrocarbons by
6.3. P450 1B1 recombinant P450 IBl variants show some varia-
tions but have not been particularly dramatic
6.3.1. Sites of Expression and (reviewed by Shimada et al.^"^^).
Abundance In vitro experiments show the inducibility of
P450 IBl in patterns expected for an^/?-responsive
P450 IBl was originally discovered in ker-
gene, which is the way in which the gene was
atinocyte cultures in a search for new dioxin-
found^^. Unlike P450 lAl and particularly 1A2
inducible genes^^ and in work on adrenals in
(vide supra), there is limited direct evidence for
animal models^^^. In contrast to P450 lAl and
inducibility of P450 IBl in vivo because of the low,
1A2 (seven exons), the P450 IBl gene has only
extrahepatic expression and the lack of a diagnostic
three exons and is located on chromosome 2
probe drug. Although the expression of P450 IBl is
instead of 15 (ref [221]). Although most of the
driven by the Ah system, additional factors must be
detailed studies of tissue-specific expression have
involved because of the known tissue and cell line
been done at the mRNA level and not protein,
selectivity of expression. For instance, major differ-
strong responses are seen in fetal kidney, heart,
ences are seen between HepG2, MCF-7, and ACHN
and brain, in that order^^^. In adults (human), there
cells (of liver, breast, and kidney tumor origins,
is little detectable expression in liver, but there is
respectively)^^ ^ With the information available
more detectable expression in kidney, spleen, thy-
today, one would expect the gene to be induced (in
mus, prostate, lung, ovary, small intestine, colon,
extrahepatic tissues) by the compounds that induce
uterus, and mammary gland^^^. Many of these tis-
P450s lAl and 1A2.
sues are of particular interest because of the
tumors that develop there. Immunochemical stain-
ing of P450 IBl has been reported in a variety of
malignant tumors^^^. 6.3.3. Substrates and Reactions
The level of expression (of the protein) in Human P450 IBl, like P450 lAl, has never
human lung has been estimated to be at the level been purified from tissue and all of our informa-
of ~ 1 pmol/mg microsomal protein in nonsmokers tion has come from protein expressed in heterolo-
and 2-4 pmol/mg microsomal protein in smokers. gous systems. 7-Ethoxyresorufin O-deethylation
Human P450s 401

can be used as a model reaction^^^. The catalytic from 7,12-dimethylbenz[a]anthracene is of partic-


activity of P450 IBl is intermediate between ular importance^ ^^.
P450s lAl and 1A2 (ref. [181]). Some other One of the interesting findings with human
model reactions can be used as welP^^. P450 IBl is that this enzyme is an efficient cata-
Much of the interest in P450 IBl has been lyst of np-estradiol hydroxylation and that the
because of its ability to activate a broad spectrum pattern is for 4- > 2-hydroxylation^^^' ^^^' ^^^. This
of chemical carcinogens, including polycyclic pattern is the opposite of that seen for P450s 1A2
hydrocarbons and their oxygenated derivatives, and 3A4 (2- > 4-hydroxylation)^^^' ^^^ and is of
heterocyclic amines, aromatic amines, and significance because 4-hydroxyestradiol is chemi-
nitropolycyclic hydrocarbons^^^ (Table 10.8). cally more reactive with oxygen and more likely
Of particular interest is the observation that to oxidize (to 0-quinone) and bind DNA^^"^. Thus,
human P450 IBl is at least as active as P450 lAl 4-hydroxyestrogens are considered to be candi-
in the conversion of the classic carcinogen dates for causing estrogen-dependent tumors^^^.
benzo[«]pyrene to the 7,8-dihydrodiol, the first The available information indicates that mouse
step in the formation of the diol epoxide^^^. P450 IBl does not catalyze estrogen hydroxyl-
In general, it would appear from the available ation^^ ^' ^^^, providing a potentially important
information that the rodent P450 IBl cnzyrnQS difference with the human enzyme. This apparent
have similar catalytic specificity as human P450 lack of conservation of selectivity has relevance in
toward carcinogens, from the available informa- the use of mouse (and rat) models in some of the
tion^^ ^ If this is a valid view, then the observation biology, for example, the human glaucoma
that P450 IBl-knockout mice do not form tumors mentioned earlier^'*^' ^^^.

Table 10.8. Carcinogens Activated by Human P450 IBl

Substrate References Substrate References

Polycyclic aromatic hydrocarbons IQ 178


Benzo[<a!]pyrene 181 Trp-Pl ' 178
Benzo[fl]pyrene-4,5-diol 178 Trp-P2 178
(+) Benzo[fl]pyrene-7,8-diol 178 PhIP 178
( - ) Benzo[«]pyrene-7,8-diol 178
Aromatic amines
Dibenzo[fl, /]pyrene 226
2-Aminoanthracene 178
Dibenzo[fl, /jpyrene-11,12-diol 178
2 - Aminofluorene 178
Benz[a]anthracene 181
4-Aminobiphenyl 178
Benz[a]anthracene-1,2-diol 178
3-Methoxy-4-aminoazobenzene 178
Benz[Gf]anthracene-c/5-5,6-diol 178
O-Aminoazotoluene 178
7,12-Dimethylbenz [a] anthracene 178
6-Aminochrysene 178
7,12-Dimethylbenz[fl]anthracene-3,4-diol 178
Benzo[c]phenanthrene-3,4-diol 178 Nitropolycyclic hydrocarbons
Fluoranthene-2,3-diol 178 1-Nitropyrene 227
Benzo[6]fluoranthene-9,10-diol 178 2-Nitropyrene 178
Chrysene-1,2-diol 178 6-Nitrochrysene 178
5 -Methylchry sene 226 2-Nitrofluoranthene 227
5-Methylchrysene-1,2-diol 178 3 -Nitrofluoranthene 227
5,6-Dimethylchrysene-1,2-diol 178 6-Nitrobenzo [ajpyrene 227
Benzo[g] chrysene-11,12-diol 178 1,8-Dinitropyrene 227
6-Aminochrysene-1,2-diol 178 1-Aminopyrene 227
Heterocyclic amines Estrogens
MelQ 178 np-Estradiol 228
MelQx 178 Estrone 229
402 F. Peter Guengerich

6.3.4. Knowledge of Active Site light of the number of carcinogens that P450 IBl
activates (Table 10.8). The issue of oxidation of
Very little knowledge about the active site is estrogens to reactive products is one worth con-
available. The general pattern of catalytic speci- sidering, in light of the experimental evidence
ficity, with similarity to P450 lAl and 1A2, supporting a link with cancer in estrogen-
would argue for some similarity. The effects of the dependent tumors. Another matter that has not
allelic variants are probably not strong enough to been addressed is the possible metabolism of the
be of much use in understanding the effects of various estrogens in postmenopausal hormone
those residues ^'^^. Some homology modeling has treatments (e.g., Premarin® by P450 IBl, e.g., see
been done^^^. refs 234 and 241 regarding DNA adducts formed
by some of these estrogens).
6.3.5. Inhibitors
aNF is a strong inhibitor, as in the case of 6.4. P450 2A6
P450 1A2 (ref [181]). Some acetylenes devel-
oped by Alworth's group have been found to selec-
6.4.1. Sites of Expression and
tively inhibit P450 IBl (at least relative to P450s Abundance
lAl and 1A2), including 2-ethynylpyrene^^^ A P450 2A6 (formerly termed IIA3 and 2A322)
potential drawback to these compounds is that was purified from human liver microsomes^ and a
they are rapidly oxidized by P450 IBl. cDNA was isolated from a human liver library^"^^.
Resveratrol is a polyphenol found in red The protein is expressed at medium to low levels
grapes and has been of interest in the context of its in liver (Table 10.5, Figure 10.2). In one study, the
potential to inhibit cancer^^^. This compound is a fraction of total human liver P450 attributed to
noncompetitive inhibitor of P450 IBl, with a K^ P450 2A6 ranged from <0.2% to 13% among
value of 23 |LJLM in model systems^^^ (with selec- individual samples, with a mean of —4%^^. P450
tivity toward P450 lAl). Recently, Potter et alP^ 2A6 was not found in placenta (frill term)^"^^.
reported that P450 IBl oxidizes resveratrol to the P450 2A6 is also expressed in other tissues,
known anticancer agent piceatannol, a tyrosine particularly in the nasopharyngeal region.
kinase inhibitor. Further studies showed that the Expression has been detected in nasal mucosa,
natural product rhapontigenin is a low K^ inhibitor trachea, lung^"*"^, and esophageal mucosa^"*^. These
of P450 lAl (ref [240]). A series of methoxy- sites of expression are of interest regarding certain
substituted trans stilbene compounds of the cancers. In liver cancers, overexpression of P450
resveratrol/rhapontigenin family were prepared 2A6 protein was associated with chronic inflam-
and tested: of these, 2,4,3',5'-tetramethoxystil- mation and cirrhosis^^^.
bene was found to be a strong and selective com-
petitive inhibitor of P450 IBl (K. = 3 nM) and
resisted demethylation^"^^. 6.4.2. Regulation and Polymorphism
The regulation of P450 2A6 expression has
been studied in primary cultures of human hepato-
6.3.6. Clinical Issues
cytes. Expression (mRNA, protein) is inducible by
No issues regarding drug interactions have rifampicin^"^^ and phenobarbitaF"*^ and, to a lesser
been raised. As with the P450 lA subfamily extent, clofibrate, cobalt, griseofiilvin, and pyra-
enzymes, an issue is that induction of P450 IBl zole^"*^. The nuclear receptor HNF-4 is involved in
might increase the activation of procarcinogens. the expression of cultured hepatocytes^'*^.
This issue may be real, although presently there is Many polymorphisms (> 11) are known for the
no epidemiological evidence to support such a CYP2A6 gene^^. These include a splice variant
relationship. Although the coding region polymor- (* 12) in which CYP2A 7 exons are included and the
phisms have only indicated a limited potential protein has lost catalytic activity^^^' ^^^ Another
for contribution to cancer (vide supra), the recent SNP polymorphism (*2), recognized earlier, is the
evidence for trimodal induction^^^ is certainly of L160H change which yields very low catalytic
interest (see Section 5, vide supra), particularly in activity^^^. Also of interest is a gene deletion (*4).
Human P450s 403

The incidence of these polymorphisms is racially (SM-12502)277, 278 and tegafur^^^, 28o^ ^j^ich is
linked^^. P450 2A6 is involved in nicotine oxida- converted to 5-fluorouracil. Halothane is reduc-
tion, and in 1998, Tyndale and her associates tively converted to a free radical by P450 2A6,
reported that individuals with low P450 2A6 activ- which can yield at least two products and initiate
ity smoke less and might have lower cancer risk^"^^. lipid peroxidation^^ ^
This proposal seems reasonable but the findings Some of the catalytic selectivity of P450 2A6
have been questioned. General agreement exists overlaps with that of P450 2E1 {vide infra). One
that defective P450 2A6 genes cause reduced nico- area in which the overlap has been noted is in the
tine metabolism (the presumed basis for reduced oxidation of nitrosamines. P450 2A6 preferen-
smoking)^^^"^^^. Several reports conclude that tially catalyzes the oxidation (and activation) of
deficient P450 2A6 reduces smoking^^^"^^^ and A^-nitro- sodiethylamine, in contrast to P450 2E1
also lung cancer^'^^' ^^^' ^^^ in smokers. The latter which preferentially oxidizes A^-nitrosodimethy-
hypothesis has biological plausibility because lamine^^^' ^^^. P450 2A6 is also involved in the
many carcinogens from tobacco are activated by oxidation of many tobacco-specific nitrosamines,
P450 2A6 (Table 10.4 and vide infra). However, including 4-(methylnitrosamino)-1 -(3 -pyridyl)-
other studies have not revealed any relationship 1-butanone (NNK)283-286 P45Q 2A6 appears
between CYP2A6 genotype and smoking; cancer is to be the major human P450 involved in the
also controversiaP^^"^^^. Some of the discrepan- activation of iV-nitroso-benzylmethylamine^^^,
cies may be raciaP^^ but even this is unclear^^^. iV-nitrosodipropylamine, A^-nitrosobutylamine,
Some problems are attributed to technical short- A^-nitrosophenyl- methylamine, and iV-nitrosonor-
comings in genotype analyses^^^ and a definite nicotine^^^. Fujita and Kamataki^^^ studied the
relationship is still lacking^^^ in Caucasians, but is bacterial mutagenicity of a number of tobacco-
more likely in Asians^^^, where the incidence of specific iV-nitrosamines and concluded that P450
gene deletion is higher. 2A6 is the major human enzyme involved in the
activation of all examined.
6.4.3. Substrates and Reactions P450 2A6 is also involved in the metabolism
of nicotine {vide supra). P450 2A6 is the main cat-
The most characteristic and specific reaction alyst in the oxidation of nicotine to cotinine^^^"^^^.
of P450 2A6 is coumarin 7-hydroxylation^' ^^^. P450 2A6 is also involved in the 3'-hydroxylation
Coumarin 7-hydroxylation has also been used as of cotinine^^^. In addition, P450 2A6 catalyzes 2'-
an in vivo diagnostic assay^^^"^^^. hydroxylation of nicotine, yielding a precursor of
One issue with P450 2A6 is whether b^ is a lung carcinogen^^"^.
required for optimal catalytic activity. Soucek-^^^ P450 2A6 can also A^-demethylate hexa-
demonstrated that a 1:1 ratio of b^ to P450 was methylphosphoramide^^^.
optimal in coumarin 7-hydroxylation catalyzed by Several forms of human P450 catalyze the
the purified recombinant enzyme. The effect of Z?^ 3-hydroxylation of indole^^^, and the product
on catalytic selectivity has not been evaluated in dimerizes to indigo. P450 2A6 was the most active
all reports on P450 2A6. human P450 identified for this activity and could
Coumarin 7-hydroxylation can be used in vivo also catalyze several oxidations of indole^^^.
with humans as a phenotypic assay. An alternative Mutants of P450 2A6 generated from a random-
procedure is to administer caffeine to individuals ized library were shown to catalyze the oxidation
and determine the conversion of 1,7-dimethyl- of several substituted indoles to generate variously
xanthine to 1,7-dimethyluric acid, a reaction cat- colored indigos and indirubins^^^.
alyzed by P450 2A6 (ref [274]).
Some industrial chemicals are substrates for
oxidation by P450 2A6, including alkoxyethers
6.4.4. Knowledge about Active Site
(used as fuel additives, e.g., tert-h\xty\ methyl
ether)^^^ and the vinyl monomer 1,3-butadiene, Relatively little site-directed mutagenesis work
a cancer suspect^^^. has been done on P450 2A6 (although the rodent
Some drugs are also substrates, including (+) P450 2A enzymes were an early target for the
c/5-3,5-dimethyl-2-(3-pyridyl)thiazolidin-4-one approach)^^^. Some information has been gleaned
404 F. Peter Guengerich

from naturally occurring SNPs. In vivo studies are in Asians^^^ but remain controversial in
consistent with a shift from coumarin 7-hydroxyla- Caucasians266,268,269,3i5,3i6^
tion to 3-hydroxylation associated with the L160H Some drugs are P450 substrates, although the
allele^^^, although this phenomenon has not been relative contribution of P450 2A6 is still so small
verified in vitro. The substitution R128Q yields a (Figure 10.3) that P450 2A6 reactions are gener-
protein with half the content of heme, but the ally not included in screens.
inability to bind carbon monoxide (to ferrous iron) P450 2A6 expression has been reported to be
(plus a loss of 98% of the coumarin 7-hydroxyla- induced during infection by (carcinogenic) liver
tion activity)^^^. flukes^ ^^ and downregulated during infection by
Lewis has published several homology models hepatitis A virus^^^.
of P450 2A6 (refs [300-302]) and also attempted
to rationalize the pattern of nicotine oxidation
using molecular orbital calculations^^^. 6-5. P450 2A7
The situation involving the CYP2A7 gene is
6.4.5. Inhibitors complex, and sometimes this has even been
erroneously referred to as a pseudogene^^. Two
Several selective inhibitors of P450 2A6 are
pseudogenes {CYP2A7PTX md CYP2A7PCX) are
known. Diethyldithiocarbamate appears to be a
known. The P450 2A7 mRNA transcript is pro-
mechanism-based inactivator, although the inactiva-
duced in human liver, at roughly the same level as
tion has not been extensively characterized^^^.
that for P450 2A6 (ref [250], [319]). Gonzalez's
Diethyldithiocarbamate and its oxidized form,
laboratory had isolated cDNA clones now recog-
disulfiram, also inhibit P450 2E1 (ref [304]).
nized as 2A6, the 2D6 variant LI60H, and 2A7, and
In vivo single-dose treatment of people with disulfi-
expressed all three in HepG2 cells^"*^. Of the three,
ram inhibits P450 2E1 but not P450 2A6 (ref [305]).
only the "wild type" P450 2A6 incorporated heme.
The vegetable watercress, a source of phenethyl
Others have also expressed P450 2A7 in heterolo-
isothiocyanate, did not inhibit P450 2A6 in vivo^^^.
gous systems but not reported any evidence of a cat-
A number of chemicals have been tested as
alytically active P450 2A7 holoprotein^^^. Whether
inhibitors of P450 2A6 in human liver micro-
or not a fiinctional P450 2A7 is transcribed from
somes^^'^. Of these, the most selective and potent
the mRNA in human tissues is still unclear, and
inhibitors appear to be 8-methoxypsoralen, tranyl-
nothing can be said about catalytic activity.
cypromine, and tryptamine, with K^ values
~ 1 JULM^^^"^^^. The inhibition by the natural prod- Gene conversion events between the CYP2A6
uct 8-methoxypsoralen (in many foods) is mecha- and CYP2A7 genes have been reported, yielding
nism based^^^. 8-Methoxypsoralen (methoxysalen) chimeric proteins in humans^^^' ^^^' ^^^. These pro-
inhibits P450 2A6 in vivo^^^ and has also been teins have some of the coumarin 7-hydroxylation
reported to decrease nicotine metabolism in smok- conferred by the 2A6 component^^^
ers^ ^^ Both of the inhibitors 8- and 5-methoxypso-
ralen were covalently bound to P450 2A6 during
incubation with NADPH^^^. Menthofiiran, another 6.6. P450 2A13
natural product, is also a mechanism-based inacti-
6.6.1. Sites of Expression and
vator of P450 2A6 (ref [313]).
Abundance
Isoniazid has been reported to be a weak
mechanism-based inactivator of P450 2A6 The CYP2A13 gene has been recognized for
(ref [314]). some time^^^ The gene is expressed in human
liver^^^' ^^^ and several other extrahepatic tissues,
including nasal mucosa, lung, trachea, brain, mam-
6.4.6. Clinical Issues mary gland, prostate, testis, and uterus^^^. The
As indicated in Section 6.4.2, the major highest level seems to be in nasal mucosa^^^, which
issue regarding P450 2A6 polymorphisms is is of interest in the context of tobacco-related can-
the effects on lung and esophageal cancers and cers because of some of the catalytic activities
smoking habits, which have good epidemiology toward nitrosamine substrates {vide infra).
Human P450s 405

6.6.2. Regulation and Polymorphism Much of the early work with P450s in experi-
mental animals was focused on the phenobarbital-
Little is known about the regulation and inducible enzymes now recognized to be in the 2B
inducibility of the CYP2A13 gene. Several variant subfamily^^^' ^^^ and a general expectation was
alleles have been identified, including one in the that similar P450s would be prominent in human
coding region (R257C) with somewhat less activ-
liver (and further suggested by immunochemical
ity toward NNK^23
studies^ and early cloning work^^^). However, the
major P450 in human liver (and small intestine)
proved to be P450 3A4 (Figures 10.2 and 10.3).
6.6.3. Substrates and Reactions The mean level of P450 2B6 in human liver has
Recombinant P450 2A13 has much lower been somewhat controversial. One of the prob-
coumarin 7-hydroxylation activity than does P450 lems has been antibody specificity. Antibodies
2A6, but coumarin is also converted to the 3,4- raised against rat P450 2B1 have not been very
epoxide^^"^. P450 2A13 also catalyzes several reac- specific^^^; unfortunately many papers in this area
tions at rates as high or higher than P450 2A6, show only limited sections of gels or actually
including 2,6-dichlorobenzonitrile activation, iV-nitro- show major cross-reactive material. The results
sodiethylamine 7V-deethylation, hexamethylphos- tend to fall into two groups. One set reports levels
phoramide A/-demethylation, 7V,7V-dimethylaniline very low to 80 pmol P450 2B6 per milligram pro-
A/-demethylation, 2'-methoxyacetophenone O-deme- ^gjj^330-332 Another set of reports range from near
thylation, A^-nitrosomethylphenylamine A/-demethy- zero levels to 28 pmol P450 2B6/mg microsomal
lation, and the activation of NNK^^^. The latter protein^^^' ^33-336 However, the mean values differ
reaction is of particular interest with regard to considerably in both the former and latter groups.
tobacco-related cancer because of the localization of While some of the discrepancy may be attributa-
expression of this P450 in nasal mucosa. ble to the differences in liver samples, the main
difference is probably with the antibodies used
and cross-reactivity with other proteins, as well as
6.6.4. Knowledge about Active Site error inherent in other aspects of immunochemi-
cal methods. Our own work is in line with the
No information is presently available beyond lower set of estimates of expression levels (mean
an effect of the R257C variant allele^^s ~ 1 % of total P450, with values rarely exceeding
5% even in samples from individuals administered
inducers)^^^. This level is an order of magnitude
6.6.5. Inhibitors less than for P450 3A4 (Figures 10.2 and 10.4).
No inhibitors have been reported.

6.6.6. Clinical Issues 6.7.2. Regulation and Polymorphism


Until recently, the mechanisms of induction by
P450 2A13 probably does not make a major
barbiturates had been rather vague in humans and
contribution to the metabolism of drugs. The
experimental animals. Studies with HepG2 cells
major interest in P450 2A13 involves a possible
(derived from hepatocytes) show the role of the
role in chemical carcinogenesis^^^.
constitutive androstane receptor (CAR), a member
of the steroid receptor superfamily, and its interac-
tion with the phenobarbital-responsive enhancer
6.7. P450 2B6 module (PBREM) in the region between —1733
and —1683 bp in the 5' flanking region^^^. Other
6.7.1. Sites of Expression and
work with HepG2 cells has implicated the liver-
Abundance
selective transcription factor C/EBPa^^^. Kliewer's
P450 2B6 is expressed primarily in liver, and group^^^ also demonstrated the involvement of
the protein has been partially purified^^^. The pro- another previously orphan receptor, pregnane X
tein has also been detected in human lung^^^. receptor (PXR), in binding to PBREM in primary
406 F. Peter Guengerich

human hepatocytes to induce P450 2B6. PXR is and propofol hydroxylation^^^. The iV-demethylation
active only when hgand-activated but CAR appar- of (iS)-mephenytoin has been used as a marker of
ently acts without an added ligand; both CAR and P450 2B6 in vitro (microsomes)^^^' ^^^' ^^°.
PXR heterodimerize with (liganded) RXR^^^. However, a valid in vivo probe for P450 2B6 is
"Cross-talk" also exists at the PBREM site with the still lacking354,360,361
vitamin D receptor as well as CAR and PXR^"*^' ^^^. As with animal P450 2B enzymes, P450 2B6
The levels of CAR and PXR mRNA in individual can also oxidize some environmental pollutants^^^.
human livers are correlated with the level of P450 Nonhyperbolic kinetics have been reported for
2B6 mRNA^^l The regulation of P450 2B6 has some P450 2B6-catalyzed reactions but these have
considerable similarity to that of P450 3A4 (vide not been extensively characterized^^^.
infra), with some differences. Several recent find-
ings provide some further insight into the mecha-
nism, although several questions persist. CAR 6.7.4. Knowledge about Active Site
does have ligand-activated effects and 6-(4-
Several homology models of P450 2B6 have
chlorophenyl)imidazo[2,l-Z?][l,3]thiazole-5-
been published^^^' ^^'^, including one using molec-
carbaldehyde 0-(3,4-dichlorobenzyl)oxime has
ular dynamics^^^.
been identified as an agonist^^"^. A novel distal
Relatively little site-directed mutagenesis has
enhancer regulated by PXR and CAR has been
been done with P450 2B6. Halpert's laboratory
identified in the CYP2B6 gene^^^.
modified 10 residues and measured some activities,
Alternative splicing in the CYP2B6 gene was although most of the changes were <2-fold^^^.
already identified in 1990^^^, with use of a cryptic Recently, Halpert's laboratory has solved a crys-
exon within introns 3 and a splice site acceptor tal structure of a derivative of the related rabbit
within exon 4. Extensive polymorphism (mainly P450 2B4 (ref [366a]), which should be of rele-
SNPs) has been identified in the CYP2B6 vance in understanding P450 2B6.
gene^^' ^^^' ^^^. Several of the mutants appear to
yield attenuated levels of protein and catalytic
activity^'^^. Some evidence for enhanced catalytic 6.7.5. Inhibitors
activity of a P450 2B6 SNP variant (N172H) has
been reported (~ 2-fold) and attributed to the Lists of the reported inhibitors of P450 2B6
homotropic activation seen in 7-ethoxycoumarin have been compiled by Rendic^^. Orphenadine had
O-deethylation^"*^. been utilized in some work with microsomes but
does not appear to be particularly selective^^^' ^^^.
More recently 2-isopropenyl-2-methyladamantane
and 3-isopropenyl-3-methyldiamantane have been
6.7.3. Substrates and Reactions reported as selective inhibitors of P450 2B6 (ref
Many reactions have now been demonstrated [368]). Triethylenethiophosphoramide has also
to be catalyzed by recombinant P450 2B6, mirror- been reported to be a selective inhibitor of P450
ing the early research in the P450 field with rat 2B6 (ref [369]).
P450 2B1 and rabbit P450 2B4 (refs [350-353]). The oral contraceptive 17a-ethynylestradiol
However, this information must be considered in is a mechanism-based inactivator of P450 2B6
the context of the amount of P450 2B6 present in and modifies the (apo)protein^^^, but the in vivo
liver and intestine, particularly in comparison with relevance of the inhibition has not been
P450 3A4 (vide supra). One estimate has been established.
made that P450 2B6 is involved in —3% of drug
metabolism reactions (Figure 10.3).
6.7.6. Clinical Issues
Lists of P450 2B6 substrates have been pub-
lished elsewhere, for example, refs 32 and 354, No real clinical issues have been identified yet,
and will not be reiterated here. One of the drugs to primarily because of the difficulty in identifying
which P450 2B6 apparently makes a significant reactions catalyzed in vivo due to the lack of spe-
contribution is cyclophosphamide^^^' ^^^. Some cific inhibitors and the overlapping regulatory
other reactions attributed to P450 2B6 involve mechanisms with other enzymes. Some pharma-
anesthetics, e.g., ketamine A^-demethylation^^^ ceutical companies have begun to include P450
Human P450s 407

2B6 in their in vitro screens for individual P450s appears to be inducible by barbiturates^^^ but a
with potential roles, however. PBREM was not found in the 5' untranslated
The phenomenon of barbiturate-like enzyme region of the gene^^ ^.
induction is still an issue in drug development, Several polymorphisms have been reported and
however. The point is not only drug interactions, studied^^^' ^^^' ^^^. Two coding region polymor-
but particularly the prospect of tumor promotion phisms involve the amino acid substitutions I264M
in rodent cancer bioassays, which is probably and K399R, with the latter appearing in a haplo-
unrelated to the P450 induction ^^^. type with R139K (ref [382]). Polymorphisms
upstream of the coding region are also known^^^.
The metabolism of taxol (paclitaxel) is decreased
6.8. P450 2C8 with the *3 allele (K399R/R139K haplotype), but
The P450s in the 2C subfamily have been of the extent of the decrease has been variable in dif-
interest for some time. In retrospect, some of the ferent studies, ranging from 90%^^^ to 25%^^^, 384
first human P450 preparations purified were prob- The *1C polymorphism appears to be associated
ably P450 2C9 (refs [3], [4]). A major impetus for with some attenuation of the mean level of
research in this field was the observed genetic expression^^^.
polymorphism in (5)-mephenytoin 4'-hydroxyla-
tion^^^' ^^^, which led to efforts at purification.
Purified proteins had some catalytic activity
toward mephenytoin^, but subsequent in vivo phar- 6.8.3. Substrates and Reactions
macokinetic^^^ and heterologous expression exper- P450 2C8 does not appear to have the general
iments^''^ demonstrated a distinction between significance of P450 2C9 (or 2C19) in drug metab-
tolbutamide and (*S)-mephenytoin hydroxylation. olism. An important substrate is taxol (pacli-
Genomic analysis indicated the complexity of the taxel)(6a-hydroxylation)^^' ^^^. Another substrate
CYP2C gene subfamily^^^. Subsequently the sub- for P450 2C8 is SilUrans retinoic acid^^^ P450
family was characterized in terms of four P450s: 2C8 also contributes to the oxidation of troglita-
2C8,2C9,2C18, and2C19 (ref [376]). P450 2C19 zone^^^ and verapamil, rosiglitazone, cerivastatin,
is the polymorphic (*S)-mephenytoin 4'-hydroxy- amiodarone, dapsone, and amodiaquine (reviewed
lase^^^' ^^^; P450 2C9 is involved in a considerable in refs [32, 382]).
number of drug oxidations (Figure 10.3). Two pre- In general, P450 2C8 has relatively low catalytic
vious entries, 2C10 and 2C17, are considered activity toward the known substrates of P450s
allelic variants cf other genes or other artifacts and 2C9 and 2C19. However, Mansuy's laboratory has
have been deleted (Table 10.1)^^^. recently synthesized a sulfaphenazole derivative
toward which all of the human P450 2C subfamily
P450s have activity^^^.
6.8.1. Sites of Expression and
Abundance
P450 2C8 was first purified from human
liver^; the enzyme is known to be expressed in 6.8.4. Knowledge about Active Site
liver and kidney^ ^^. The available data indicate Some of the knowledge about the catal5^ic
that the level of expression of P450 2C8 is rela- selectivity of various P450 2C enzymes can be
tively low in liver but may be one of the more sub- interpreted in terms of the active site. The rabbit
stantial P450s in the kidney. Other sites of P450 P450 2C5 structure provides some possible insight
2C8 (mRNA) include adrenal gland, brain, uterus, in this area. Recently, the groups of Johnson
manmiary gland, ovary, and duodenum^^^ and Mansuy^^^ have obtained a three-dimensional
structure with a common P450 2C subfamily
substrate^^^ bound. Two binding modes were
6.8.2. Regulation and Polymorpiiism
observed, one of which corresponds to the
The level of P450 2C8 expression in human observed oxidation^^^. Very recently Johnson's
liver varies at least 20-fold^^^. Rifampicin induces group has obtained a crystal structure of a slightly
P450 2C8 in hepatocyte culture^"^^. The enzyme modified P450 2C8 (ref [389a]).
408 F. Peter Guengerich

6.8.5. Inhibitors allelic variant of P450 2C9, although since no


evidence for the Asp417 mutation has been found
In contrast to P450 2C9, sulfaphenazole is not in population studies, no allele designation has
a strong inhibitor of P450 2C8. Mansuy's group been made; the original assignment had been based
has synthesized some sulfaphenazole-based selec-
on the now unexplained distinct 3' noncoding
tive inhibitors of individual P450 2C enzymes,
sequence^^^. Most of the literature dealing with
including P450 2C8 (refs [390], [391]). The early
P450 2C10 can be interpreted as 2C9.
work on paclitaxel metabolism suggests that high
concentrations of the natural flavonoids narin-
genin, quercitin, and kaempferol and the synthetic 6.9.1. Sites of Expression and
(xNF inhibit^^, but little in vivo inhibition would
Abundance
be expected.
P450 2C9 is primarily a hepatic P450. The
level of expression is probably the highest, on
6.8.6. Clinical Issues the average, except for P450 3A4 (Figures 10.1
Induction and inhibition of P450 2C8 are not and 10.4; Table 10.5)^1 All P450 2C enzymes are
particular issues at this point. Although P450 2C8 absent in fetal liver, including P450 2C9 (ref
may play a prominent role in the hepatic and renal [393]), and levels rise quickly in the first month
oxidation of arachidonic acid and retinoic acid, no after birth^^^. Pharmacokinetic experiments with
disease etiology has been implicated at this point. accepted P450 2C9 substrates indicate that the
The most serious issue is probably any impact on level of hepatic P450 2C9 does not change with
the disposition of the cancer chemotherapeutic age, at least to 68 years^^^.
agent paclitaxel. Polymorphisms may have some P450 2C9 is also expressed in the small
effect on in vivo 6a-hydroxylation^^^' ^^^, although intestine^^^.
any influence may be modulated in part by the
contribution of P450 3A4 to other reactions^^.
6.9.2. Regulation and Polymorphism

6.9. P450 2C9 Early work with human hepatocytes showed


induction of P450 2C9 by barbiturates and
In retrospect, many of the observations regard- rifampicin"^^^, consistent with earlier in vivo work
ing in vivo metabolism of barbiturates^^' ^^^ are on the induction of barbiturate metabolism^^^.
some of the first reports on what is known as P450 Subsequent studies have shown that P450 2C9 is
2C9. P450 2C9 is one of the major enzymes the only P450 2C subfamily enzyme expressed at
involved in drug metabolism (Figure 10.3). a significant level in untreated hepatocytes and
Retrospectively some of the first purified human that expression is induced by rifampicin, dexam-
liver P450s can now be recognized as P450 2C9 ethasone, and phenobarbitaP^^' ^^K The induction
(refs [3], [4]). The protein purified with some involves a glucocorticoid receptor, CAR, and
mephenytoin 4'-hydroxylation activity (MP-1) is PXR, with CAR and PXR apparently competing
also P450 2C9 (ref [9]), and the cDNA corre- at the same site'*^^.
sponds"^^^. Proteins were also purified from liver on Recently, evidence for action of CAR at an
the basis of their oxidation of tolbutamide^^^ and additional site has been presented^^^. It should be
hexobarbitaP^"^' ^^\ The human P450 2C subfamily emphasized that the action of CAR is somewhat
is complex^'^^ and characterization of individual different than other receptors from the steroid
members was not achieved without heterologous receptor superfamily, in that it may be enhanced in
expression and careful analysis of catalytic activi- the absence of a bound ligand and some of the
tjg5374,396 ^ transcript designated as P450 2C10 control is at the level of nuclear translocation'*^'*.
from this laboratory had only two apparent coding Other factors involved are HNF-4 (ref [405]) and
region changes (Cys358 and Asp417) from the C/EBPa (ref [338]), accounting at least in part for
CYP2C9*1 allele, one of which (reported initially hepatic localization.
as Cys358) was subsequently shown to result from The genetic polymorphism of P450 2C9
a sequencing error^^^. This is now thought to be an has been studied extensively and has clinical
Human P450s 409

significance, although P450 2C9 probably does P450 2C9 is the major catalyst of oxidation, and
not have a critical function in normal physiology. polymorphisms affect the in vivo pharmacokinetic
Tolbutamide metabolism had been reported to dis- parameters'^ ^'^^^.
play polymorphism^^^, which was an impetus to Several aspects of P450 2C9 reactions are of
purify the protein catalyzing the hydroxylation^^^. concern regarding interpretation of results, at least
A 6-base deletion in the coding region lowered in in vivo research. One issue is the effect of sol-
catalytic activity in a recombinant enzyme"^^^. vents on catalytic activity'^^. A concentration of
A number of P450 2C9 SNPs have been identi- 1% (v/v) CH3CN markedly inhibited the catalytic
fied"^^^ and their racial linkage has been explored"^^^. activity of P450 2C9 (ref [423]). Another issue is
P450 2C9 polymorphism has been reviewed the enhancement of most reactions by b^ (ref.
recently^^^' ^^^ and the reader is referred to these [424]). Further work also showed that apo-Z)^
reviews and to the website http://www.imm.ki.se/ (devoid of heme) was as effective as ^5 (ref
Cypalleles/ for more details. Of some interest, in [425]), arguing against a need for electron trans-
addition to the *2 and *3 alleles with generally fer. Other work showed that even other P450s
lower catalytic activity, is the *5 allele (of higher fre- could enhance the rates of some P450 2C9 reac-
quency in Africans) with lower catalytic activit)^^^. tions, even though those P450s did not catalyze
Some of the SNPs occur in the 5'-flanking region the reactions themselves"^^"^. These results are rem-
and attenuate the expression of P450 2C9 (ref iniscent of some of the interactions of rabbit
[413]). Also of interest is an unusual phenomenon in P450s 1A2 and 2B4 reported by Backes'^^ and are
which the CYP2C18 exon 1-like locus is fused with still unexplained.
combinations of exons and introns from CYP2C9 to Other work with P450 2C9 has provided evi-
yield chimeric RNA transcripts^^^. Finally, linkage dence for cooperativity in some reactions, although
between CYP2C8 and CYP2C9 genetic polymor- the area has not been as developed as for P450 3A4
phisms has been reported"^^^. {vide infra). Dapsone and some analogs enhance the
binding and 4-hydroxylation of diclofenac"^^^' '^^.
However, the activity of P450 2C9 toward dapsone is
6.9.3. Substrates and Reactions
unaffected by diclofenac, in a situation similar to that
P450 2C9 is one of the major P450s involved of P450 3A4, aflatoxin B^, and aNF'29 jy^^ jj^^er-
in drug metabolism (Figures 10.3 and 10.4). Some pretation that P450 2C9 uses two binding sites in
aspects of substrate specificity have been these interactions is probably valid"^^^, although (as
reviewed by Miners and Birkett^^^. A more exten- with P450 3A4) the mechanism remains to be eluci-
sive recent compendium of substrates has been dated (including the exact nature of the binding).
developed by Rendic^^.
One of the early substrates examined was pheny-
toin, which undergoes 4-hydroxylation^. P450s
6.9.4. Knowledge about Active Site
2C19 and 2C18 (R. Kinobe and E.M.J. Gillam,
personal communication) can also catalyze this The point should be made before detailed con-
reaction but P450 2C9 is the major catalyst"^^^. siderations of site-directed mutagenesis, etc., that
Recently, Mansuy's group has used the P450 changes in particular residues of P450 2C9 yield
2C9 inhibitor sulfaphenazole to build a substrate markedly different effects depending on the sub-
common to all four P450 2C subfamily enzymes^^^. strate and reaction under consideration. For
Some compounds normally in body are instance, the polymorphism *3 (I359L), which
oxidized by P450 2C9, including linoleic acid appears to be very conservative, changed catalytic
(epoxidation)"^^^ and vitamin A (all-^ra«5-retinoic efficiencies of different reactions by factors of
acid, 4-hydroxylation)'*^^, although the physiolog- 3-27-fold {in vitrof^^. Although the *2 and *3
ical significance is unknown. polymorphisms cause considerable changes with
Several reactions have been used as in vivo some substrates, diclofenac metabolism is not
probes, including tolbutamide, warfarin, flurbi- altered^^^ consistent with the in vitro findings.
profen, and losartan^^^. With the above caveats, roles of a number of
One substrate of recent interest is celocoxib, amino acids have been examined with several reac-
a cyclooxygenase (C0X)-2 inhibitor (Celebrex®). tions, although extrapolation to more reactions
410 F. Peter Guengerich

requires caution. Arg97 and Arg98 affected group has also announced a P450 2C9 crystal
activity toward diclofenac in a yeast recombinant structure"^"^^.
system"^^^; in contrast, mutation of Arg97 ablated
hemoprotein expression in a bacterial system
(E.MJ. Gillam, personal communication). Muta- 6.9.5. Inhibitors
tion of Lys72 failed to affect affinity for ibuprofen Sulfaphenazole has been recognized as a
or diclofenac (E.M.J. Gillam, personal communi- highly selective competitive inhibitor of P450 2C9
cation). Asp293 has been shown to have a rela- for some time"^"^^ and has relatively poor affin-
tively general structural role, possibly by bonding ity for other P450 2C subfamily enzymes^^^.
to a partner amino acid or amide^^^. Studies with Mansuy's group has examined some other similar
coumarins suggested two sites, one for Il-stacking compounds as ligands and inhibitors^^^' ^^^.
of aromatic rings and an ionic binding site for
Other inhibitors have been reported, although
organic anions^^"^; many P450 2C9 ligands have an
some have relatively poor affinity"^"^^' ^'^^, including
anionic charge^^^' '^^^.
several warfarin analogs^^^. For a more extensive
P450 2C9 was converted into an enzyme with compilation of inhibitors, see Rendic^^.
(5)-mephenytoin 4'-hydroxylation activity (i.e., Tienilic acid is a mechanism-based inactivator
P450 2C19-like) with a relatively small number of of P450 2C9 (ref [451]). The mechanism involves
changes (I99H, S220P, P221T, S286N, V292A, iS-oxygenation, and the unstable product reacts
F295L). Comparisons with the crystal structure of with P450 2C9 (ref [452]). Subsequently, autoim-
rabbit P450 2C5 suggests that most of these mune antibodies develop in some patients that rec-
residues are unlikely to directly contact the sub- ognize unmodified P450 2C9 (ref [451]). Exactly
strate but probably influence packing of substrate- how (or if) this process is related to the hepatitis
binding sites and substrate-access channels^^^. seen in some individuals who used tienilic acid is
Conversely, P450 2C19 could be transformed to still unclear'*^\ but the phenomenon has raised
an enzyme with warfarin hydroxylation activity concerns about whether such processes might be
similar to that of P450 2C9 (and also sul- associated with other drugs that covalently modify
faphenazole binding) with the changes N286S, proteins and could lead to idiosyncratic drug reac-
I289N, and E241K (ref [438]). Other work iden- tion in patients, one of the major concerns today for
tified roles of residues 292, 295, and 399 plus safety assessment in drug development. Structure-
residues 231-288 (substrate-binding sequence activity relationships have been reported on
[SRS] 3) as important in P450 2C9 activities^^^. thiophenes other than tienilic acid"^^^.
Mansuy's laboratory identified residues 476, 365,
and 114 as being important in diclofenac and sul-
faphenazole binding and in inactivation by tienilic
acid^"^^. Phell4 is proposed to be involved in 6.9.6. Clinical Issues
Il-stacking'^'^^ perhaps serving the role proposed The major issue regarding P450 2C9 is its
in the coumarin studies mentioned earlier^^"^. It role in drug development because of the sizeable
might be speculated that Phel20 in P450 2D6 fraction of drugs oxidized by this enzyme
could serve a similar role in that enzyme (vide (Figure 10.3)^^. Although the polymorphism is not
infmY^K as dramatic as with P450 2C19 or P450 2D6 (vide
Several models of P450 2C9 have been infra), it can be an issue in drug interactions and
published^^^'^^^^*^^. Some of these take experimen- safety.
tal binding studies into consideration in their formu- A general issue with P450 2C9, because of its
lation while others are only based on homology. Of relatively high abundance (Figures 10.1 and 10.4),
interest is the recent work with rabbit P450 2C5 is its role in reducing bioavailability. However,
using P450 2C9 ligands, showing multiple substrate- estimating in vivo pharmacokinetic properties
binding modes^^^. from in vitro data is still not trivial. Houston has
A crystal structure of P450 2C9 with bound reviewed the issue with P450 2C9 recently^^"*.
warfarin has been published recently^"^^^. Goldstein'*^^ has reviewed the clinical relevance
Obviously no information is available regarding of genetic polymorphisms in the P450 2C subfam-
ligand interactions either. Very recently Johnson's ily. One of the most relevant involves warfarin.
Human P450s 411

which has a relatively low therapeutic index"^^^. and protein"^^^ levels. However, expression in lung
(i^)-Warfarin is oxidized by P450 1A2 (6- and and skin appears to be significant^"^"^' ^'^^^ ^'^^.
8-hydroxy) and P450 3A4 (10-hydroxy), and (S)-
warfarin is oxidized primarily by P450 2C9
(7-hydroxy)^^^' ^^^. The metabolism of (*S)-warfarin 6.10.2. Regulation and Polymorpiiism
is competitively inhibited by (i?)-warfarin, but the The variability in levels of expression of P450
converse is not the case"^^^. The hydroxylation of 2C18 in human liver is difficult to assess because
(5)-warfarin by P450 2C9 (ref [459]) is an issue of the very low levels (<2.5 pmol/mg microsomal
because of reduced catalytic efficiency by the *2 protein)"^^-^. The extent of variability in other
and *3 variants^^' ^^^' ^^^ The differences are mani- tissues is not known.
fested in altered toxicity of warfarin (hemorrhaging) Rae et al?"^^ reported that P450 2C18 was not
at a given dose and in an altered optimal dose of inducible by rifampicin in human hepatocytes, in
warfarin^^"^^' ^^^^ ^^^. The issue extends to the analog contrast to P450s 2C8 and 2C9.
acenocoumarol"^^"^. The principles of physiologically Polymorphisms in the CYP2C18 gene have
based pharmacokinetic modeling have been applied been reported"^^^, but the effects on expression and
to the variation of warfarin risk in individuals with catalytic activities are not well characterized. One
different genotypes/phenotypes"^^^; this effort may possible polymorphism has an exon 5 deletion'^^^
serve as a paradigm for other efforts to convert in
vitro data on P450 variability into estimates of risk.
Tolbutamide hydroxylation is another example 6.10.3. Substrates and Reactions
of a manifestation of in vitro knowledge about
P450 2C18 has low catalytic activity in tolbu-
P450 2C9 in clinical pharmacolog/^^^^^ In one
tamide methyl hydroxylation"^^ ^ Limited activity
sense, this is rather logical because the in vitro
toward drugs has been shown, and P450 2C18
work with tolbutamide^^^ was developed from in
probably does not make much contribution in gen-
vivo findings"*^^.
eral drug disposition, in part because of low expres-
In other clinically relevant research involving
sion levels. P450 2C18 is active in phenytoin
P450 2C9, the genotype has been reported to pre-
metabolism, having an enzyme efficiency {k^JK^
dict the blood pressure response to the drug irbe-
for 4-hydroxylation comparable to P450 2C9, and
sartan^^^ a relative to the P450 2C9 substrate (and
being more active in the bioactivation to a reactive
the prodrug losartan)^^^' 47i Although P450 2C9
intermediate (R. Kinobe and E.M.J. Gillam,
is involved in the metabolism of diclofenac, no
personal communication).
relationship of the genotype with the cases of
Minoletti et al.^^^ studied a series of derivatives
diclofenac-induced hepatitis was observed"^^^.
of tienilic acid and characterized an aroylthiophene,
The final issue about P450 2C9 is possible rel-
3-[2,3-dichloro-4-(2-thenoyl)phenoxy]propan-1 -ol,
evance to cancer risk. Some carcinogens are sub-
as a selective substrate for 5-hydroxylation by P450
strates (e.g., benzo[a]pyrene^^^) although many of
2C18 (k. = 125 min-i, K^ = 9 |ULM).
the reactions are probably detoxications. CYP2C9
SNPs have been analyzed in relation to colorectal
cancer. An association was found in one study"^^^, 6.10.4. Knowledge about Active Site
but not a subsequent one"^^"^. In another study, no
association of CYP2C9 SNPs was found with lung Information about the active site of P450 2C18
cancer^^^. is relatively limited beyond the substrates cited
above"^^^, the interaction of other P450 2C proteins
with general 2C substrates^^^ and inhibitors^^^, and
6.10- P450 2C18 inferences from the rabbit P450 2C5 structures^^^.
At least one homology model has been published"*^^.
6.10.1. Sites of Expression and
Abundance
6.10.5. iniiibitors
Of the four human P450 2C subfamily mem-
bers, the level of hepatic expression appears to be P450 2C18 is not appreciably inhibited by sul-
lowest for P450 2C18, at both the mRNA376, 476 faphenazole. Mansuy's group has published on
412 F. Peter Guengerich

some synthetic inhibitors (sulfaphenazole deriva- 6.11.2. Regulation and Polymorphism


tives) that can be used in vitro^^^- ^^^
In vivo work had shown that the enzyme was
inducible by rifampicin^^^. Thus, this P450 dif-
fered from P450 2D6 in that it was both polymor-
6.10.6. Clinical Issues phic and inducible. Analysis of the regulatory
The limited expression and repertoire of cat- system has not been extensive, but studies with
alytic activity of P450 2C18 preclude considera- human hepatocytes have demonstrated induction
tion of clinical issues at this point in time. of P450 2C19 mRNA by rifampicin, dexametha-
sone, and phenobarbital'^^^
The polymorphism is now relatively well
understood. The incidence of the PM phenotype in
6-11. P450 2C19 Caucasians is generally 3-5% but the incidence in
Asians is —20%"^^. On some Pacific islands, the
Interest in P450 2C19 developed from the dis-
incidence is as high as 75%"^^^' ^^^. The major
covery of the pol3miorphic metabolism of the
defect in Caucasians and Japanese was first iden-
iS-isomer of mephenytoin, the first major poly-
tified in an exon 5 mutation that leads to an aber-
morphism to be studied following P450 2D6
rant splice site and yields a truncated protein^^^.
(refs [371], [372]). Initial work led to the purifi-
Other polymorphisms are collected at the website
cation of an enzyme with some (*S)-mephenytoin
http://www.imm.ki.se/CYPalleles/. These are rather
4'-hydroxylation activity^. Exactly how this and
diverse and include a mutation of the initiation
other gene products from the complex P450 2C
codon"^^^ and altered enzymatic properties"^^^.
family^^^' ^^^ were involved was unclear"^^^' ^^^.
Although there were some indications that the
hexobarbital 3'-hydroxylase (P450 2C9) was the
6.11.3. Substrates and Reactions
enzyme of investigation^^^' ^^^, expression of P450
2C9 cDNA^^^ in yeast yielded a protein with (»S)-Mephenytoin 4'-hydroxylation is the classic
activity towards tolbutamide but not (iS)-mepheny- reaction attributed to P450 2C19. Early studies on
tQijj374,396 P450 2C18 had also been suggested to the basis of the polymorphism of tolbutamide
be the enzyme^^^. hydroxylation suggested that the same enzyme
Wrighton^^'^ compared (»S)-mephenytoin 4'- might be responsible for both activities^^^, but in
hydroxylation activity in different liver samples vivo work^^^ and heterologous expression studies^^'*
with a protein gel band recognized by anti-rat distinguished the two activities. Nevertheless,
P450 2B1 and correlated this with P450 2C19, recombinant P450 2C19 has now been shown to
a sequence which had been reported earlier. have some tolbutamide hydroxylation activity^^^.
Subsequently, Goldstein et al?^^ expressed several Extensive lists of reports of P450 2C19 reactions
P450 2C subfamily cDNAs in yeast and identified have been published by Rendic^^ and only a few will
P450 2C19 as having the highest activity be mentioned. The scope of P450 2C19 in drug
metabolism is relatively restricted (Figure 10.3).
One drug of particular interest is the ulcer drug
omeprazole (and related compounds), because indi-
6.11.1. Sites of Expression and
viduals with low enzyme activity show a better
Abundance response to treatment for ulcers^^' ^^. Some of the
Apparently significant expression only occurs early variations seen in warfarin metabolism^^^ can
in the liver. As with all other P450s examined to be explained by the finding that P450 2C19
date, there appears to be no gender difiference"^^^. catalyzes the 8-hydroxylation of (/?)-warfarin'^^'^.
P450 2C19 is a relatively minor P450 in its abun- 18-Methoxycoronaridine is 0-demethylated by
dance, probably accounting for < 5 % of total P450 P450 2C1949^ P450 2C19 is responsible for the 5-
even in EM liver samples (Figure 10.4). and 5'-hydroxylation of thalidomide, an older drug
P450 2C19 and (5)-mephenytoin 4'-hydroxy- notorious for teratogenic effects that has been
lation activity were not detected in fetal liver "rediscovered'"^^^. Whether the polymorphism was
samples^^^. related to the birth defects is unclear.
Human P450s 413

P450 2C19 also oxidizes steroids, including Most pharmaceutical companies and regula-
progesterone 21-hydroxylation and testosterone tory agencies discourage development of a P450
IT-oxidation"^^^. Finally, the organphosphate 2C19 substrate because of potential problems for
insecticide diazinon is activated in human liver by PM individuals. However, several studies indicate
P450 2C19 (ref. [498]). that PM patients may have more effective therapy
(for ulcers) with omeprazole and related com-
pOUnds489, 500-503^
6.11.4. Knowledge about Active Site As with many polymorphisms, epidemiology
As with other P450 2C subfamily enzymes, studies have been done to explore risks to diseases
P450 2C19 activities are usually stimulated by b^ in the absence of information about etiology, sub-
(ref. [425]). In this case, stimulation is not strates, etc. Some of the reports include sugges-
dependent on heme in the b^ so electron transfer tion of more hepatocellular cancer in PMs^^"^ and
cannot be involved"^^^. lack of association of leukemia with polymor-
Homology models of P450 2C19 have been phism^^^. Other possible relationships have been
published302,444 explored but evidence for any associations is
Goldstein's group did chimeric analysis and limited at this time"^^^.
then site-directed mutagenesis on P450 2C9 to
convert it to a protein with P450 2C19-character- 6.12. P450 2D6
istic omeprazole hydroxylation activity^^^. Only
three changes were needed to achieve the activity P450 2D6 is one of the main enzymes involved
of wild-type P450 2C19:199H, S200P, and P221T. in drug metabolism (Figure 10.3). It was the first
However, at least three different mutations were "xenobiotic-metabolizing" P450 recognized to be
needed to convert P450 2C9 to an enzyme with under monogenic regulation^.
(»S)-mephenytoin 4'-hydroxylation activity, even
to a catal3^ic efficiency one third of wild-t3q)e
P450 2C19 (ref [437]). In an opposite experi- 6.12.1. Sites of Expression and
ment, P450 2C19 was converted to a P450 2C9- Abundance
like warfarin hydroxylase with high sensitivity to
P450 2D6 is expressed mainly in liver and was
sulfaphenazole"^^^. Residues 286 and 289 appear
first purified from liver microsomes^' ^^. In the
to be important. However, these residues may
average person, P450 2D6 accounts for —5% of
exert an indirect influence by adjusting the active
total P450 (with wide variation)^^. However, this
site or substrate-access channels"^^^.
enzyme is involved in the oxidation of ~ 2 5 % of
all drugs oxidized by P450s (Figure 10.3).
6.11.5. Inhibitors Developmental studies show little P450 2D6 in
fetal liver and a rapid increase in protein shortly
Relatively little has been published concerning after birth, yielding a peak accumulation in
P450 2C19 inhibitors, although screening may be newborns and decline in adulthood^^^.
done in some pharmaceutical companies. Recently, P450 2D6 is also expressed at low levels in
Mansuy's group has developed some P450- lung (bronchial mucosa and lung parenchyma)^^^.
selective inhibitors for the 2C subfamily enzymes, Another site of P450 2D6 expression is brain,
including P450 2C19 (refs [390], [391]). with localization in large principal neurons^^^.
Higher levels of brain expression have been
reported in alcoholics^^^.
6.11.6. Clinical Issues
The issue is the polymorphism, particularly for
6.12.2. Regulation and Polymorphism
drugs marketed in Asian populations. At least
eight alleles have been associated with the PM AH information available indicates that P450
phenotype"^^^. Desta et al.^^^ have reviewed some 2D6 is not inducible. Some factors are known to
of the drugs for which the 2C19 phenotype is a be involved in constitutive expression, including
problem. C/EBPa338 and HNF-4a53.
414 F. Peter Guengerich

The wide variability in the activity of P450 2D6 result from less stable proteins^ *^, although low
is attributed to genetic variabihty (Figure 10.5). activity P450 2D6 variant proteins have also been
Reduced ability to metabolize the drug debrisoquine reported^*^'^*^. Some of the allelic differences are
was first noted (personally) by Smith in a drug trial. present as haplotypes^*^.
Subsequent work led to the report of polymorphic In addition to the "poor" and "intermediate"
hydroxylation of debrisoquine^, including a pheno- metabolizer phenotypes, an "ultrarapid" metabo-
typic hypotensive response^^^. Racial differences lizer phenotype was identified in early work
were first noted with Africans'^'^. The phenomenon (Figure 10.5). Ingelman-Sundberg's group identi-
of polymorphic debrisoquine hydroxylation^*^ was fied the basis for this as a gene duplication, with up
also reported for sparteine oxidation"^^' ^*^. to 13 copies present in some individuals'*^. The
Purification of the P450 2D6 enzyme^' *^' *' was fol- main form of this phenomenon is a haplotype
lowed by Gonzalez's cloning of the gene'^'^ and iden- resulting from gene duplication"*^' ^^^. The amplifi-
tification of some of the genetic defects as mRNA cation appears to result from unequal segregation
splicing variants^*^. and extrachromosomal replication of the acentric
Today more than 70 alleles of P450 are DNA^^*. As many as 7% of Caucasians show some
known and have been classified with a nomen- of this effect, and the incidence is even higher in
clature system^ ^ Systems for genotyping have some Ethiopian and Middle Eastern populations^^^.
become relatively powerful^^"^ and the "intermedi-
ate metabolizer" phenotype has been character-
6.12.3. Substrates and Reactions
ized^*^. The most significant decreases in activity
for P450 2D6 alleles, aside from mRNA splicing Since the original work with debrisoquine^,
problems and gene deletion^^, are considered to many substrates and reactions have been reported

Figure 10.9. A pharmacophore model for the active site of P450 2D6 (ref [527]). Inhibitors are overlaid to keep
the nitrogen atoms (marked with arrow) in a fixed position.
Human P450s 415

for P450 2D6. In some cases, the role of P450 usually Asp301 in most studies. (Recent work
2D6 is very dominant in vivo and the clinical man- shows a role for Glu216, however, vide infra.)
ifestations of genetic polymorphism are important The use of these models requires some caveats.
and even deadly^^^' ^^^. An extensive list of P450 Although the pK^ of the substrate has been proposed
2D6 substrates has been published recently by to have a dominant influence^^^, work in this labo-
Rendic^^. ratory has shown that the intrinsic pK^ of a substrate
P450 2D6 catalyzes many of the basic kinds can be altered in the active site of P450 2D6 (ref
of oxidative reactions of P450s, for example, [531]). Another issue is that some compounds with
aliphatic and aromatic hydroxylations, heteroatom a single amine nitrogen undergo A/-dealkylation, for
dealkylations, etc^^"^. In early work in this labora- example, deprenyl^^^, which cannot be rationalized
tory^^^, the observation was made that most of the with an amine-oxidation site interatomic distance of
substrates contained a basic nitrogen atom situ- 5-7 A. Some substrates devoid of basic nitrogen
ated ~ 5 A away from the site of oxidation, possi- (and any nitrogen) have been reported, including
bly due to a specific anionic charge in P450 2D6. steroids^^^' ^^^. Spirosulfonamide and several
Subsequently, more detailed pharmacophore mod- analogs are devoid of basic nitrogen and have been
els have been developed^^^"^^^ (Figure 10.9). All shown to be good substrates and ligands for P450
of these are based on the premise that a basic 2D6 (ref [535]) (Figure 10.10).
nitrogen atom in the molecule interacts (coulom- A large fraction of the population is devoid of
bic bond) with an acidic amino acid in P450 2D6, active P450 2D6 but appears to fimction well. This

Bufuralol

1 R = so2NH2 Ka^^\iM 6
/Cd 6.2 ^iM
(spirosulfonamide) Kd 4.0 ^iM

2 R = SO2CH3 Kd 32 |iM
(spirometiiyisulfone)

3 R = CONH2

4 R = CSNH2 /Cd1.7|iM

5 R = C02H (Kd >75 |IM)


Testosterone Progesterone
KdlS^iM /Cd1.5|LiM
CH3
H3C-

a
Ritonavir K^i1.2\ihA

Figure 10.10. Analogs of spirosulfonamide and other P450 2D6 ligands. K^ values were estimated by spectral
titrations^^^.
416 F. Peter Guengerich

information may be interpreted to mean that P450 A list of P450 2D6 residues postulated to form
2D6 has no "physiological" substrate. Nevertheless, the active site includes at least Asp 100, Trp316,
some reactions may be catalyzed by P450 2D6 and Pro371 (ref [539]), Prol03, Ilel06, ThrlOT,
yield physiological responses that yield less than LeullO, Proll4, Serll6, Alal22, Asp301, Ser304,
obvious changes. For instance, overexpression of Ala305, Thr309, Val370, Gly373, Val374, and
human P450 2D6 in transgenic mice produces Phe483 (refs [542], [551]), Phel20, Glu216 (refs
a somewhat lethargic phenotype (F.J. Gonzalez, [441], [541], [543], [544], [550], [552], [553]), and
personal communication). Tryptamine has been G l n i n , Leul21, Leu213, Phe219, and Phe481
proposed as a physiological substrate in one study^^^ (ref [543]). Only six of these residues have been
but discounted in another^^^. Proposed physio- examined experimentally to date. The effects of
logical reactions catalyzed by P450 2D6 are Asp301 have already been mentioned, with caveats
the 0-demethylations of 5-methoxytryptamine, about general changes in the protein"^"^^' ^^^.
5-methoxy-i\yV-dimethyltryptamine, and pinoline Changing Val374 to Met also has an eflfect^'^^' ^^7.
(6-methoxy-l,2,3,4-tetrahydro-p-carboline)^^^' ^^l Mutation at Asp 100 or Ser304 has been reported to
Whether significant catalytic is seen at the low con- have little effect, if any^'^^' ^^\ Mutation of Phe483
centrations that occur in vivo and what the effect is to He produced some alteration of the pattern of
remains to be established. testosterone oxidation by P450 2D6 (ref [551]). A
change in Phe481 yielded a 10-fold lower catalytic
efficiency (k^JK^) toward some substrates but not
6.12.4. Knowledge about Active Site others^^^. The effects of Glu216 have already been
The active site of P450 2D6 has been the sub- mentioned"*"^^' ^^^ and seem to be restricted largely
ject of considerable interest, probably because of to the basic amines'^'*'. Recent models of the P450
the relevance to issues in the pharmaceutical 2D6 active site (Figure 10.11) suggest that both
industry. Some residues have been identified as Asp301 and Glu216 are within bonding distance of
being important, and many homology and phar- amine substrates'^'*'' ^'*^. Another suggestion from
macophore models have been published^^^"^^^' the more recent models'*"*'' ^^^ is that one role of
539-545 (Figure 10.9). Asp301 is to use amide hydrogen bonds to estab-
The original clone reported by Gonzalez"^^ had lish the juxtaposition of Phel20, which may be
Met at position 374 but this now appears to be an involved in hydrophobic bonds with substrates.
artifact and the correct residue is Val^"*^' ^^^. This Site-directed mutagenesis experiments with this
residue appears to be in the active site and affects residue are currently in progress (F.P Guengerich
activity. and E.M.J. Gillam, unpublished results).
In 1995, Ellis et al.^"^^ found that mutation of The work cited above brings up the point that
Asp301 to neutral residues reduced catalytic activ- certain mutations may alter activity toward some
ity toward several substrates and concluded that substrates but not others (e.g., Phe481 (ref [555]),
this acidic residue was involved in docking amine Glu216 (ref [441])). Similar behavior is seen with
substrates through coulombic interaction. Subse- some of the natural allelic variants of P450 2D6 as
quently, all models published until recently have well556.
been based on this view. A caveat about the reduc- Modi et al.^^^ reported differences in product
tion in the catalytic activity of the Asp301 mutants profiles of P450 2D6 reactions supported with
is that heme incorporation is diminished (and artificial oxygene surrogates and NADPH-P450
is completely abolished when basic residues are reductase, and interpreted these as evidence for an
substituted)^^^. Further, as indicated earlier, some allosteric influence of the reductase. Subsequent
P450 2D6 substrates (e.g., spirosulfonamide) are experiments in this laboratory did not support this
devoid of basic nitrogen but the hydroxylations conclusion and are in accord with some differ-
are still attenuated by mutation at Asp301 ences in the chemical mechanisms for the oxygen
(ref [535]). Subsequent work in this laboratory surrogates^^^.
showed that the oxidations of basic amine sub- Detailed experiments have been done on the
strates (and their binding) are dependent upon 0-demethylation of 3- and 4-methoxyphenethy-
Glu216 (Asp216 is also efiFective)'^^^ a result lamine by P450 2D6 (ref [559]). Analysis of kinetic
independently reported by Wolf's group^^^. deuterium isotope effects, kinetic simulation, and
Human P450s 417

Figure 10.11. Model of some residues in the P450 2D6 active site'^'^^ Views of the substrate-binding cavity in the
P450 2D6 homology model are shown with only the relevant residues, plus the heme, I-helix backbone, and other
areas of peptide backbone shown. Hydrogens are not shown except for the amide hydrogens of residues 119 and 120
hypothesized to hydrogen bond to the carboxylate oxygen atoms of Asp301. Side view of the active site from the
perspective of the I-helix: I-helix residues have been cut away excepting the side chains of residues 309, 301, and
313 shown at the front of the view.

other experiments yield evidence that both late steps Rendic^^. Inhibition of P450 2D6 is an undesirable
in O2 activation and C-H bond breaking contribute issue in drug development, and most pharmaceuti-
to k^^^. The exact meaning of X^ is still not defined cal companies have screening programs in place.
with this and most P450 reactions. Some of the The most established inhibitor of P450 2D6 is
P450 2D6 allelic variants show no changes in k^^^ quinidine^^^ The K^ is ~50 nM and inhibition is
for certain reactions but do show K^ differences^^^; competitive. Interestingly, quinidine is not a sub-
these are probably more complex than simple strate for P450 2D6 (refs [106], [559]).
"affinity" for the substrate. Mechanism-based inactivation of P450 2D6 is
known, for example, 5-fluoro-2-[4-[(2-phenyl-
li/-imidazoyl-5-yl)methyl]-1 -piperazinyl]pyrimi-
6.12.5. Inhibitors
dine (SCH66712)^^^. In the case of this compound,
Many inhibitors of P450 2D6 have been covalent binding to protein was detected but the
reported; for a compilation of the literature, see position of attachment has not been identified.
418 F. Peter Guengerich

6.12.6. Clinical Issues Another disease in which P450 2D6 has been
proposed to play a role, on the basis of epide-
The clinical issues regarding P450 2D6 are miology, is Parkinson's disease^^^. Contradictory
considerable due to the large variation in the findings have been reported^^^' ^^^. Although a
genetics in the population (Figures 10.2 and 10.5), hypothesis has been raised that induction of P450
and the contribution of P450 2D6 in the total 2D6 by smoking might explain some discrepan-
scheme of drug metabolism (Figure 10.3). cies^^ ^ this proposal lacks biological plausibility in
Individuals seem to be rather tolerant of the wide light of the known refractory response of P450 2D6
variability in expression with many marketed to induction.
drugs, probably because of generally wide thera- A final issue is that of autoantigens. Auto-
peutic windows selected for in the basic process of antigens (LKMl) that recognize P450 2D6 have
drug development. However, P450 2D6 PMs can been known for some time^^^' ^^^. These antibod-
be at considerable risk when they encounter cer- ies are associated with some cases of hepatitis.
tain drugs, as first observed by Smith^' ^^^. The The exact mechanism of how they arise is still
problem is seen with drugs having a relatively unclear, as is the relationship with hepatitis. The
narrow therapeutic index, for example, debriso- antibodies may arise by molecular mimicry^^"^
quine^, phenformin^^^, captopril^^"^. The effects of or they may result from P450 2D6 translocation
P450 2D6 deficiency are seen not only in short- to the outer plasma membrane^^^' ^^^. These
term treatments but also in long-term therapy^^^. "LKMl" antibodies may serve as diagnostic tools
The issue of ineffectiveness of drugs that are very for particular types of hepatitis^^^' ^^^, but causal
rapidly metabolized by "ultrarapid" metabolizers relationships have never been demonstrated.
is an issue (Figure 10.5). Modeling of the vari-
ability is still an issue^^^ and may be a function of
particular drugs. The issue of whether genotyp-
ing/phenotyping is economical has been consid-
ered, particularly in the case of neuroactive and 6-13. P450 2E1
antipsychotic drugs^^^' ^^^. The overlap between The mixed-function oxidation of ethanol was
P450 2D6 substrates and neuroactive drugs is also reported nearly 40 years ago^^^. The view that
an issue in drug development, largely due to the ethanol could be a P450 substrate was not readily
overlap of these two groups of compounds^^^. accepted because of the hydrophilic nature of the
Another issue with P450 2D6 is the relevance molecule, but Lieber's group characterized the
of the polymorphism to cancer risks. In 1984, enzyme in rat liver^^^' ^^K Collaborative work with
Idle^^^ reported an association of lower risk of Levin led to the isolation of the P450 ("j"), which
lung cancer (in smokers) with the P450 2D6 PM was also found to be inducible by isoniazid^^^.
phenotype. These epidemiology results were Human P450 2E1 was purified by Wrighton
reproduced in some studies^^^, but not others'^^. et al.^^^, and Gonzalez's group characterized the
Attempts were made to resolve the discrepancies human gene^^'*.
on the basis of levels of smoking^^^ Although
some expression of P450 2D6 is detectable in
lung^^^, no clear role for P450 2D6 in carcinogen
6.13.1. Sites of Expression and
activation could be established, even with crude
Abundance
tobacco smoke fractions^^^. The issue of whether
lung cancer is associated with P450 2D6 was not The greatest concentration is in the liver, and
resolved by changing analyses from phenotyping P450 2E1 is a moderately abundant P450
to genotyping. The generally accepted epidemio- (Figure 10.4). The inter-individual variation is an
logical conclusion today is that P450 2D6 is not order of magnitude (Figure 10.2)^^' ^^^. A racial
related to lung cancer^ ^^' ^72-575 difference exists, with Japanese samples having
Other epidemiology studies have suggested rela- mean expression levels less than Caucasians
tionships of P450 2D6 with other cancers^^^' ^^^, but (Figure 10.2)^1
these findings have not been scrutinized as much as P450 2E1 is reported not to be present in fetal
the lung cancer hypothesis. liver but appears within a few hours after birth,
Human P450s 419

regardless of the gestational age^^^. The activity increased mRNA translation efficiency, and
increases during the first year of childhood, and decreased protein degradation^^^.
transcriptional regulation due to hypermethylation HNF-1 is reported to regulate CYP2E1 gene
has been proposed. transcription^^^. Obesity and diabetes are known to
P450 2E1 is expressed in many extrahepatic sites modulate P450 2E1 in rat models. In rat hepatocyte
including lung^^^, esophagus, small intestine^^^, cell culture, insulin attenuated mRNA levels and
brain^^^' ^^^, nasal mucosa^^^, and pancreas^^^ (some glucagon or dibutyryl cyclic AMP elevated mRNA,
of the evidence is extrapolated from rat work but not with the latter effect downregulated by a protein
necessarily extended to humans). kinase A inhibitor^ ^^. mRNA levels are also selec-
P450 2E1 is found mainly in the endoplasmic tively attenuated in mice or cell culture (relative to
reticulum. With heterologous expression in bacte- other P450s) by interleukin-6^^\ interleukin-4^^^, or
ria, (rabbit) P450 2E1 is membrane bound and interleukin-ip or tumor necrosis factor (TNF)a^^^.
catalytically active even when amino acids 3-29 Multiple mechanisms have been invoked, including
are deleted^ ^' ^^^. The same bacterial localization kinase pathways, control of HNF-1 a fimction, and
was seen with human P450 2E1 from which 21 regulation of other transcription factors.
N-terminal residues were deleted^^^. However, Evidence for control at the level of mRNA sta-
P450 2E1 can show some unusual localization in bility and enhanced translation efficiency has
mammalian systems. Ingelman-Sundberg's group been presented by Novak^^"^' ^^^. The 3'-region of
deleted residues 2-29 of rat P450 2E1 and demon- the gene appears to be important in stability. The
strated the presence of a shattered fragment in the relevance of this rat model to human P450 2E1 is
mitochondria of a mouse hepatoma cell line^^"^. still unknown.
Avadhani's group found P450 2E1 intact in rat Another mechanism, generally well accepted
liver mitochondria and reported that it could cou- although not completely understood, involves pro-
ple these with adrenodoxin and adrenodoxin tein stabilization by substrate. Rat studies {in vivo)
reductase there with frill catalytic activity^^^. showed that - 1 / 2 of P450 2E1 was lost in 1 hr,
Subsequent work demonstrated a cryptic mito- and a ubiquitin-linked pathway was invoked^^^.
chondrial targeting signal at positions 21-31 that Similar findings were also reported for human
was activated by cyclic AMP-dependent phospho- P450 2E1 in HepG2 cells^^l An attempt has been
rylation of Serl29 (ref. [26]). Neve et al^^^ found made to estimate the halflife of P450 2E1 in
that the charge of the iV-terminus of (rat) P450 humans in vivo using chlorzoxazone pharmacoki-
2E1 was such that a part is directed to either the netics and a P450 2E1 inhibitor^^^. The halflife
lumen of the endoplasmic reticulum or the outside was estimated at 50 ± 19 hr, but this approach
of the plasma membrane. The relevance of these may not be sensitive enough to detect a short-lived
localizations to human tissues is still unknown P450 2E1 pool. The relevance of substrate stabi-
but likely. lization of P450 2E1 to in vivo parameters has
been addressed by Thunmael and Slattery^^^.
P450 2E1 is polymorphic and, because of the
6.13.2. Regulation and nature of many of the substrates, many efforts have
been made to determine the relevance of SNPs and
Polymorphism
other polymorphisms to disease and risk of injury.
Early work in experimental animals was For a current update on CYP2E1 polymorphisms,
focused on the induction of P450 2E1 in rat see http://www.imm.ki.se/Cypalleles/. A polymor-
liver^^^. Subsequently, many other chemicals, phism in the 5' flanking region was suggested to
including isoniazid and some solvents, were shown be related to the binding of a transcription factor
to induce P450 2E1 (ref [607]). It is also of and related to alcohol intake^^' ^^^. A number of
interest to note that some of the common poly- other polymorphisms have been identified^^' ^^^'
cyclic hydrocarbons and other inducers of P450 1 ^^^. However, the evidence to date indicates that
family enzymes attenuated the level of P450 2E1 these polymorphisms do not seem to have much
(ref [607]). The regulation of P450 2E1 has come significance in terms of their effects on in vitro
to be recognized to be relatively complex, involv- or in vivo activity of P450 2E1 (refs [84], [621],
ing transcriptional activation, mRNA stabilization, [623-625]).
420 F. Peter Guengerich

6.13.3. Substrates and Reactions molecular weight cancer suspects including not
only nitrosamines but also benzene, styrene, CCI4,
P450 2E1 was originally characterized as an CHCI3, CH2CI2, CH3CI, CH3CCI3, 1,2-dichloro-
ethanol-oxidizing enzyme. P450 2E1 can oxidize propane, ethylene dichloride, ethylene dibromide,
some compounds that are present in the body, vinyl chloride, vinyl bromide, acrylonitrile, vinyl
including acetone and possibly other ketones carbamate, ethyl carbamate, and trichloroethyl-
involved in certain physiological syndromes gj^g304 jj^Q oxidations by P450 2E1 all have rele-
(fasting, diabetes)^^^. Transgenic P450 2E1-knock- vance to the activation and detoxication of these
out mice appear to be relatively normal, although compounds and their risk assessments^"*' ^^^.
the blood acetone levels become much higher Another substrate is the gasoline additive methyl
(than in wild-type mice) after fasting^^^. tert-buty\ ether^^l A role of P450 2E1 has been
The role that P450 2E1 plays in ethanol metab- shown in the activation of some of these chemicals
olism has been debated for many years^^^. What in knockout mice^"*"*' ^'*^.
seems to be the general consensus is that alcohol Another substrate for human P450 2E1 is lau-
dehydrogenase is the main enzyme involved in ric acid, which undergoes 1 l-hydroxylation^'*^' ^'*^.
ethanol oxidation. P450 2E1 may make a contri- The physiological relevance of this reaction
bution at very high ethanol concentrations or in is unknown. Indole is oxidized by P450 2E1
individuals with low levels of alcohol dehydroge- (3-hydroxylation, generating indigo) as well as
nase activity. P450 2E1-knockout mice have blood by other P450s, particularly P450 2A6 and 2C19
ethanol levels not significantly different from wild- (refs [296], [648]). The relevance of this reaction to
type animals after administration of ethanol^^^. the urinary excretion of indigoids^"*^ is still unclear.
Acetaldehyde, the product of ethanol oxidation, is
Relatively few drugs are oxidized by P450 2E1
also oxidized to acetic acid by rat and human P450
(Figure 10.3). Chlorzoxazone is one^^"*. Halogenated
2E1 (ref [630-632]).
anesthetics are often metabolized by P450 2E1,
The oxidation of 4-nitrophenol to 4-nitrocate- including halothane^^^ and isoflurane^^^
chol has been used as an in vitro marker of human For more lists of substrates, see Rendic^^.
P450 2E1 (ref [633]). Chlorzoxazone 6-hydroxyla-
tion was demonstrated to be a relatively specific
reaction catalyzed by human P450 2E1; other
6.13.4. Knowledge about Active Site
enzymes (e.g., P450 lAl) can catalyze the reaction
but with poor catalytic eff'iciency^^'*' ^^^. Chlorzo- One of the issues in P450 2E1 reactions is
xazone is a relatively innocuous muscle relaxant and the need for b^, first demonstrated with the rat
the assay can be used in vivo to estimate hepatic enzyme^s^ and also the human enzyme^"*^' ^^^; the
P450 2E1 fimction noninvasively^'^' ^^^. involvement also exists in microsomes^^^. b^ also
One group of substrates of interest is A^- augments P450 2E1 activity in bacterial expres-
nitrosamines, which are carcinogens at many sites sion systems"*^^' ^^^. In contrast to several of the
and can be formed by chemical reactions within P450s, apo-Z?3 (minus heme) does not ftinction,
the body (e.g., stomach acid)^^^. Early research on arguing for a "classic" role of electron donation in
the activation of A^-nitrosodimethylamine (N,N- enhancement of catalysis"^^^' ^^^.
dimethylnitrosamine) indicated biphasic kinetics A number of homology models of human P450
of the activating iV-demethylation reaction and the 2E1 have been published, based upon bacterial
possible contribution of multiple P450s and possi- P450s and rabbit P450 2C5 (refs [302], [656],
bly other enzymes^^^' ^^^. The enzyme involved in [657]). One of the diff'iculties in dealing with mod-
the "low KJ' reaction was shown to be P450 2E1 els for the low molecular weight substrates is that
m
many of these compounds have very little in the
in rat and human liver^^^' ^^^. An in vivo role of
way of features to bond to, other than hydrophobic
P450 2E1 has been confirmed in rats^"^'. However,
residues or halogens. Utilizing these models for
P450 2A6 has a significant share of the role of
both very small substrates (e.g., ethanol, CH3CI)
activation of some more complex nitrosamines,
and larger, more conventional ones (e.g., chlorzox-
even A^-nitrosodiethylamine-^^^' ^^^.
azone, lauric acid) is an issue, unless only parts of
P450 2E1 has been shown to be a major P450
the larger substrates are inserted. One problem is
involved in the oxidation of a number of low
Human P450s 421

that inherent binding affinities are generally isotope effect is expressed in the K^ parameter,
unknown and spin-state changes have not been which includes the C-H bond-breaking step, k^^^ is
very useful with P450 2E1 (refs [652], [658]). governed largely by an enzyme physical step after
Mathematical models have also been devel- oxidation of substrate. In this system, the K^ term
oped for rates of oxidation by P450 2E1 contains k^^^ as a variable^^^' ^^^.
(refs [659], [660]). In essence, these are based on A final point involves a report of the kinetics
chemical reactivity at individual substrate atom of CO binding to human P450 2E1 following flash
sites. In both of the cited examples^^^' ^^^, the photolysis^^^. The kinetics appeared to be
models were used for relatively small sets of monophasic and the rate was decreased in the
related compounds and may have some utility. An presence of (400 mM) ethanol. One interpretation
inherent problem in more extended sets is the dif- of the results is that binding of the substrate makes
ficulty in interpretation of the parameters k^^^ and P450 2E1 more rigid^^l
K^. Thus, the rate-limiting step may not be related
to hydrogen abstraction or a similar chemical step 6.13.5. Inhibitors
involving the substrate {vide infra).
As mentioned earlier, many low molecular
Keefer et al.^^^ reported a kinetic deuterium
weight solvents are substrates for P450 2E1.
isotope effect on the carcinogenicity of iV-nitroso-
These are also inhibitors of P450 2E1 (refs [69],
dimethylamine in rat liver. Subsequent work with
[70]). Such inhibition is a problem in that histori-
rat and liver microsomes indicated that the effect
cally many insoluble P450 substrates have been
of the deuterium substitution was expressed in the
added to enzymes using final solvent concentra-
parameter K^ but not k^^^ (^max)^^^' ^^^ ^^^^ ^^^"
tions of 1% (v/v), which is often —100 mM. Thus,
tope effects on K^ were seen when deuterated
care is needed in analyses. It is possible to dilute
iV-nitrosodimethylamine was used as a competitive
many of the P450 2E1 low molecular weight sub-
inhibitor of other P450 2E1 reactions^^^. These
strates directly in water to add them to incuba-
results were of interest in that deuterium substitu-
tions, for example, methylene chloride has a
tion would not be expected to modify the affinity
solubility of - 1 0 0 mM in H^^^^.
of a substrate for an enzyme. Studies with deuter-
ated and tritiated ethanol in this laboratory also Some of the alcohol and aldehyde dehydroge-
indicated an isotope effect on the oxidation of both nase inhibitors are also inhibitors of P450 2E1,
ethanol and acetaldehyde by recombinant human making interpretations of in vivo ethanol metabo-
P450 2E1, manifested mainly in Kj^^^ ^^^ J^IQ lism studies difficult. 4-Methylpyrazole is an
results are understood in the context of a reaction excellent inhibitor^^^' ^^^ and probably one of the
sequence where burst kinetics are observed, that is, best choices for in vitro experiments at this time.
the first reaction cycle is much faster (—400 min" ^) 3-Amino-l,2,4-triazole^^'^ and diethyldithiocarba-
than the subsequent ones, which control k^^^. Pulse- mate^^"^ are mechanism-based inactivators. The
chase experiments suggest that little of the latter is of interest in that the oxidized form, disul-
acetaldehyde (or its hydrated form CH3CH(OH)2) firam (Antabuse®), is an aldehyde dehydrogenase
dissociates, due to kinetic phenomena. Neither inhibitor used in patients in alcohol aversion ther-
ethanol, acetaldehyde, nor acetic acid has much apy. Many of the early animal and human studies
affinity for P450 2E1. The rate-limiting step on interactions of ethanol and disulfiram with
occurs after product formation (for both ethanol various chemicals can now be rationalized in the
and acetaldehyde oxidations), but is not product context of P450 2E1 (refs [668], [669]).
release per se. This view of the reaction sequence A number of compounds of natural origin have
may apply to some P450 2E1 reactions but not also been examined as P450 2E1 inhibitors, many
others. Recent work in this laboratory with both of which are derived from vegetables such as
P450s 2E1 and 2A6 has shown a kinetic isotope onions, garlic, and cruciferous vegetables^^^' ^^^
effect primarily on X^, for the iV-dealkylation of
iV-nitrosodimethylamine but not A'-nitrosodiethy- 6.13.6. Clinical Issues
lamine^^^, and the kinetic mechanisms remain to The major clinical issues involve the role of
be further elaborated. An interesting point of the P450 2E1 in the oxidation of certain drugs, alco-
deuterated ethanol work is that the intermolecular holism, oxidative stress, and risk from cancer.
422 F. Peter Guengerich

As pointed out earlier, the most generally results appear to be very mixed. An early report
accepted noninvasive human assay involves suggested a link of lung cancer with a polymor-
6-hydroxylation of the muscle relaxant chlorzoxa- phism^^"^, but since then the results have been mixed
zone^^^' ^^^. Studies with humans show little effect for cancers of the lung^^^"^^^, oral cavity^^^' ^^^, and
of diabetes^^^' ^''^, but an effect of body weight/ stomach^^^. In most of these cases, it should be
obesity^^^' ^^^. As mentioned before, genotype has emphasized that there is little information about
shown little impact on the in vivo parameters to exposure and the only relevant etiology is probably
dateH673 tobacco-derived nitrosamines. In a study of workers
Another issue is drug metabolism and toxicity. exposed to vinyl chloride (a P450 2E1 substrate^^"^),
Acetaminophen overdose remains a major cause some association was found between P450 2E1
of liver failure in the United States. Several P450s polymorphisms and p53 mutations^^"^. However, it
are involved in the oxidation to the reactive imi- should be emphasized again that the relevance of
noquinone^^^. Studies with P450 2E1 knockout CYP2E1 polymorphisms to known P450 2E1 reac-
mice indicate that P450 2E1 is a major determi- tions is unclear, particularly in vivo^^^, and it is diffi-
nant of acetaminophen toxicity, because the toxic- cult to define roles of these genetic polymorphisms
ity was considerably attenuated in null animals^^^. in cancer risk; overall P450 2E1 expression due to
P450 2El-null mice have the same blood environmental influences may have a role but is
ethanol levels as wild-type animals after ethanol more difficult to establish.
dosing^^^ suggesting that P450 2E1 activity is not
a major factor in ethanol metabolism, at least in 6.14. P450 2F1
mice. The situation regarding a role for P450 2E1
in alcohol-induced liver injury in other models is This is primarily a lung P450. In 1990,
unclear, with some reports suggesting a link^^"^' ^^^ Nhamburo et al.^"^^ cloned the cDNA from a
and others not^^^' ^^^. Autoantibodies against P450 human lung library. The level of expression
2E1 have been reported in alcoholics^^^ and attrib- appears to be relatively low, as judged by the
uted to hydroxyethyl radicals^^^ (which may arise mRNA abundance. The apparent orthologs 2F2
from lipid peroxidation processes rather than as and 2F3 have been studied in mouse and goat
intermediates in P450-catalyzed oxidation, vide lung, respectively.
supra). P450 2E1 is also a major autoantigen asso- P450 2F1 has been expressed in heterologous
ciated with halothane hepatitis, a rather idiosyn- systems. Catalytic activity was observed for
cratic response^^^. As with other autoimmunities 7-ethoxy- and -propoxycoumarin O-dealkylation
involving P450s, causal associations remain to be and 7-pentoxyresorufin 0-depentylation. The
demonstrated"^^^. enzyme showed modest activation of the lung toxin
Another issue is the contribution of P450 2E1 and (potential drug candidate) 4-ipomeanol^^^.
to oxidative stress. Ingleman-Sundberg reported However, the ability of P450 2F1 to activate the
that P450 2E1 contributed - 2 0 % of the NADPH- potential lung toxicants 3-methylindole, naphtha-
dependent lipid peroxidation in rat liver micro- lene^^^, and styrene^^^ is more impressive (the acti-
somes (and 45% in microsomes prepared from vation of 3-methylindole appears to involve initial
rats treated with acetone to induce P450 2E1)^^^. desaturation^^^).
Transfection of human P450 2E1 into a rat hepatic The basis for the selective expression of P450
stellate cell culture system yielded elevated pro- 2F1 in lung is unknown. Recently, Carr etal.^^^ iso-
duction of reactive species^^^ Cederbaum^^^ has lated the CYP2F1 gene. Using luciferase-based con-
reviewed studies on the relationship of oxidative structs, they identified a specific promoter element
stress to P450 in liver cell models. The exact rele- that binds a protein in the -152 to —182 5' region.
vance to liver injury and alcohol-induced disease This protein is termed a lung specific factor (LSF).
requires more investigation.
Many studies have been reported on the rela- 6.15. P450 2J2
tionship of CYP2E1 polymorphisms to risk of
diseases. Benzene poisoning in Chinese workers The P450 2J2 cDNA was first isolated from a
showed some changes in risk with one genotype but human liver library but was found to be most
only in smokers^^^. With regard to cancers, the highly expressed in heart^^^. Expression (mRNA)
Human P450s 423

has also been found in kidney and muscle^^^, No other information is presently available
lung^^^ and the gastrointestinal tract^^^. about P450 2S1.
Zeldin's group has done most of the work on this
P450, including the initial cloning and analysis of
tissue expression. Incubation of a recombinant P450 6.18. P450 2U1
2 J2 (plus reductase and NADPH) with arachidonic
acid yielded all four epoxides, that is, epoxy- As with some of the other human P450 genes,
eicosatetraenoic acids (EETs)^^^. These EETs were the only information presently available is the
found in heart tissue, and the stereochemistry of the identification of the CYP2U1 gene in the human
,705
recombinant P450 2J2 products was found to match genome
that of the compounds isolated from tissue. A num-
ber of physiological functions have been postulated
for the EETs, reviewed elsewhere^^^. 6.19. P450 2W1
The extent of human variability of expression No information is available at this time except
of P450 2J2 has not been reported. However, for the existence of the CYP2W1 gene in the
Zeldin's group has sequenced CYP2J2 genes and human genome^^^.
found a number of SNPs^^^. One was in the pro-
moter region, eight were exonic regions, five were
in introns, and four were in the 3'-untranslated 6.20. P450 3A4
region. Only four of the SNPs resulted in amino
acid changes. These allelic variants were expressed P450 3A4 is the most abundant P450 in the
in a baculovirus system; all had activity toward body (e.g.. Figures 10.2 and 10.4) and has a dom-
arachidonic and linoleic acids within a 2-fold level inant role in drug metabolism (Figure 10.3). Some
of wild-type P450 2J2, with the N404Y variant of the earliest preparations of human P450
showing only 10% catalytic activity (all assays (refs [3], [4]) were retrospectively found to be
only done at a 100 jxM substrate concentration), P450 3A4. Two approaches led to an extensive
although some qualitative changes in products characterization. Watkins et alP isolated a P450
were seen with the I192N substitution. The physi- from human liver using the criterion of immuno-
ological relevance of these substitutions is presently chemical cross-reactivity with what is now recog-
unknown. nized as a rat 3A subfamily P450; this laboratory
isolated an enzyme from human livers that cat-
alyzed the oxidation of the hypotensive dihy-
6.16. P450 2R1 dropyridine drug nifedipine^^. cDNA cloning
yielded sequences corresponding to CYP3A3
The only information available is the presence (ref. [707]) and CYP3A4 (ref. [708]). (The former
of the CYP2R1 gene in the human genome^^^. differed from CYP3A4 at 14 sites and could be
considered a rare allele, although it has not been
reported again^^^"^^^ and originally came from the
6.17. P450 2S1 same single-liver cDNA library as the CYP3A4
This gene was identified by searching data- clone; CYP3A3 has been dropped from the
bases by Rylander et alJ^^ mRNA and protein nomenclature and earlier references to this should
blotting work indicate highest expression in tra- probably be considered to indicate P450 3A4.)
chea, lung (and fetal lung), stomach, small intes- Subsequently, studies with microsomes, anti-
tine, and spleen. Expression was also relatively bodies, and purified P450 3A4 quickly indicated
abundant (mRNA level) in colon, appendix, liver, that nifedipine was not the only substrate; other
kidney, thymus, substantia nigra, peripheral substrates included other dihydropyridines^^^,
leukocytes, and placenta. Absolute levels of abun- steroids^^' ^^^, quinidine^^^, the oral contraceptive
dance are unknown. 17a-ethynylestradioF^, and the carcinogen afla-
Rivera et alP demonstrated that both mouse toxin Bj (ref. [714]). With more studies and the
and human P450 2S1 mRNA transcripts are application of recombinant systems, the repertoire
inducible by TCDD in cell culture. of substrates expanded rapidly^^^.
424 F. Peter Guengerich

6.20.1. Sites of Expression and extent because of difficulty in finding appropri-


Abundance ately responsive cells to utilize the CYP3A4 gene
and vector constructs derived from it. Guzelian's
P450 3A4 is the most abundant P450 in human laboratory reported that the source of liver cells
liver and in the small intestine. The average frac- was a greater issue than the CYP3A regulatory
tion of the total P450 in liver accounted for by region in comparing interspecies differences in
P450 3A4 is -25-30%28 (Figures 10.2 and 10.4); CYP3A gene regulation^^^, and this result can now
in the small intestine, the fraction attributed to be rationalized in the context of new knowledge
P450 3A4 is even higher. A study with the selec- about receptors (vide infra).
tive inhibitor gestodene, which destroys P450 Although most CYP3A subfamily genes are
3A4, indicates that P450 3A4 can constitute 60% inducible by dexamethasone, the classic glucocor-
of the total hepatic P450 (ref [716]). ticoid receptor was shown not to be involved in rat
P450 3A4 is also expressed in some extra- liver^^^. In early 1998, Maurel reported that the
hepatic tissues, including lung^"^"^' '''^, stomach, macrolide antibiotic rifampicin acted as a non-
colon^^"^, and adrenal (weak)^^l P450 3A4 does not steroid ligand and agonist of the human glucocor-
appear to be expressed in kidney, prostate, testis, or ticoid receptor, providing a possible mechanism
thymus, but other 3A subfamily P450s are^^^. The for regulation and a difference with the rodent sys-
literature is mixed on whether expression occurs in tems^^^. The interpretation of these conclusions
peripheral blood lymphocytes or not^'^' ^'^. was questioned by Ray et alP^.
A gender difference in P450 3A4 expression Shortly thereafter, Kliewer's group character-
does not appear to occur^^ and apparent pharma- ized the human homolog of a mouse receptor
cokinetic differences may be attributable to P- (PXR) that bound steroids and interacted with
glycoprotein, not P450 3A4 (ref. [55]). In fetal CYP3A subfamily genes in the manner expected
liver, P450 3A7 is the most abundant form and for a major regulatory influence^^^' ^^^ (some liter-
P450 3A4 expression is very low^^' ^^^. P450 ature also refers to the human PXR as "SXR").
3A4 expression increases rapidly after birth and This member of the steroid receptor family
reaches 50% of adult levels between 6 and "orphan" group interacted with barbiturates,
12 months of age^^^. Although many general steroids (including dexamethasone), statin drugs,
regulatory concerns have been expressed about macrolide antibiotics, and some organochlorine
additional safety margins for children with drugs pesticides^''^' -^^^
and other chemicals, the evidence in this case Knowledge of the PXR and its cognate binding
indicates that P450 3A4 activity levels in infants site has led to the development of PXR receptor
are slightly higher than in adults^^^. and reporter assays to screen for P450 3A4 induc-
P450 3A4 is expressed in some tumors, tion with new drug candidates^^"^"^^^. The discov-
although the literature is mixed as to reports of ery of the PXR receptor suggested that alleles of
levels lower and higher than the surrounding this receptor might be responsible for the variable
tisSUe^21-723 _
inducibility in different individuals. However, the
SNPs found to date have not been found to control
P450 3A4 induction^^^. The regulation of CYP3A4
6.20.2. Regulation and Polymorphism expression is more complicated than simple load-
The CYP3A4 gene is at chromosome 7q22.1 ing of activated PXR (e.g.. Figure 10.6), as sug-
(ref [724]). Although 3A subfamily enzymes gested by Kliewer's early work showing the roles
were long known to be inducible in animals'^^^ and of coactivators'^^''^^^. However, the glucocorticoid-
considerable literature existed on the in vivo mediated induction of P450 3A4 is mediated by
induction of many activities by barbiturates and elements in addition to the now-canonical PXR
macrolide antibiotics (e.g., rifampicin)"^^, early gjl^g738,739 Some compounds (e.g., ketoconazole)
demonstrations of inducibility were indirect but suppress CYP3A4 gene expression, apparently via
some progress was made^^, A general correlation binding to the PXR and interaction with "co-
between enzymes and mRNA levels could be repressors" (NCoR, SMRT)^^^. CAR (see Section
shown in human livers^^^' '^^^. Defining the 6.7.2) appears to interact with the CYP3A4 gene at
mechanism of regulation was difificult^^^, to some the PXR site and induce^'^^ Further, there is
Human P450s 425

evidence that la,25-dihydroxyvitamin D3 (see as lovastatin (Mevacor®) and other statins^^^, the
Section 6.53) also controls the transcription of prostate hypertrophy inhibitor finasteride
P450 3A4 (ref [742]). This effect is mediated (Proscar®/Propecia®)^^^, the immune suppressant
through the vitamin D receptor^"^^, which has sim- cyclosporin^^^' ^^^, protease inhibitors such as
ilarity to PXR and CAR in the steroid receptor indinavir^^^, and sildenafil (Viagra®)^^"^.
superfamily. Kinases have been shown to modu- In the course of these reactions, P450 3A4 cat-
late the induction of P450 3A4 via the vitamin D alyzes examples of some atypical reactions^^"^
receptor in Caco-2 cells^^^. including desaturation'^^^, oxidative carboxylic
Other factors also contribute to P450 3A4 reg- acid ester cleavage^^^, and oxidation of a nitrile to
ulation. Among these are C/EPPa and DBP^"^"^ and an amide''^^. An unexpected reaction encountered
PINF-4a (ref [745]). Interleukin-6 has been in this laboratory was the oxidation of alkylphenyl
reported to downregulate P450 3 A4 through trans- ether non-ionic detergents, which have been com-
lational induction of the repressive C/EBPp-LIP monly used in enzyme purifications^^^ and also
protein^"^^. Thus, the transcriptional regulation of have some medical and industrial applications^^^.
P450 3A4 expression centers on PXR but involves Methylene hydroxylations yield hemiacetals,
many other aspects. which break down to shorten the chains''^^.
Another aspect of P450 3A4 regulation involves One of the classic (and fastest) reactions cat-
degradation. Troleandomycin, erthyromycin, and alyzed by P450 3A4 is testosterone 6p-hydroxyla-
some related amine macrolide antibiotics form tion^^. However, the physiological significance of
"metabolite complexes" (C-nitroso: iron, R-N= this and other (P450 3A4-catalyzed) steroid
0:Fe) and inactive protein accumulates'^^^' ^^^. hydroxylations^ ^^ is unclear. The significance of
These studies have relevance to in vivo P450 3A4 P450 3A4 in physiology may be questioned,
inhibition by these drugs. given its variability (Figure 10.1 and Table 10.5).
P450 3 A4 appears to be degraded by a ubiquitin- However, some contributions are possible and may
linked pathway^^^. Correia's group also reported be suggested in recent work. Cholesterol is oxidized
that protein kinase C modified P450 3A4 at Thr264 by P450 3A4 to 4p-hydroxycholesterol, a major cir-
and Ser420; the relevance of these phosphorylations culating oxysteroF^^' ^^^. P450 3A4 also catalyzes
to ubiquitin-linked degradation is yet unknown^"^^. the 25-hydroxylation of 5p-cholestane-3a,7a,12a-
The issue of polymorphism is considered in trio^^^' '^^^ The product is a potent PXR agonist,
the context of attempts to explain the population and this system might function as an autoregulatory
variability in P450 3A4 activity, which does not pathway (i.e., excess triol activates PXR and P450
show true modality in its distribution'^^^. A num- 3A4, which reduces the level of trioF'^^).
ber of SNPs and other polymorphisms have been P450 3A4 also functions in the metabolism of
identified, but they have not shown much relation- cancer chemotherapeutic drugs. In addition, atten-
ship to catalytic activities yet'^^^"^^^. tion has been given to activations of drugs and
chemical carcinogens. P450 3A4 activates the
estrogen receptor antagonist tamoxifen to produce
6.20.3. Substrates and Reactions DNA adducts^^^. Another example of carcinogen
Analysis of the catalytic activity of P450 3A4 activation involves aflatoxin B^ which undergoes
and other 3A subfamily enzymes is not always both a detoxicating 3a-hydroxylation and forma-
easy to assess because of nuances about the effects tion of the highly mutagenic 8, 9-6xo-epoxide^^'^'
of the membranes and other proteins, as discussed 774,775 Some other carcinogen substrates of P450
in Section 6.20.4. Wrighton has examined P450s 3A4 are listed in Table 10.4.
3A4, 3A5, and 3A7 under identical conditions and One of the issues with P450 3A4 is which reac-
concluded that P450 3A4 is generally more tion provides the most appropriate index of activ-
catalytically active than 3A4 or 3A7 toward all ity, both in vitro and in vivo. Historically
substrates examined^^^. nifedipine oxidation and testosterone 6p-hydroxy-
P450 3A4 contributes to the metabolism of lation were among the first activities identified^^
—50% of the drugs on the market or under devel- and are still used in vitro^^. Midazolam 1 '-hydrox-
opment (Figure 10.3). For an extensive list, see ylation has also been used^^ in part because of its
Rendic^^. Many of these are important drugs such acceptance for in vivo assays.
426 F. Peter Guengerich

Some higher throughput fluorescence assays 6.20.4. Knowledge about Active Site
have also been developed and gained commercial
appear'^^' ^'^^. One issue regarding these and also Because of the importance of this enzyme in
several other P450 3A4 reactions is that they show drug metabolism (Figure 10.3), many efforts have
variable effects of added chemicals, that is, one dealt with developing a better understanding of its
compound may inhibit a certain P450 3 A4 reaction function and there is hope that intelligent predic-
but stimulate another. Chauret et alP^ reported a tions can be made regarding activities toward new
fluorescence reaction that behaves in a very similar drug candidates. However, as already alluded to,
way to testosterone 6p-hydroxylation. Houston has P450 3 A4 has some unusual properties and a num-
examined the behavior of P450 3A4 probe sub- ber of important issues have not been resolved.
strates in vitro and grouped them into two cate- In the early purifications of P450 3A4 (ref [12]),
gories. Although all of these reactions are catalyzed reconstitution conditions were difficult to optimize
by P450 3A4, they are categorized into two groups and showed some unexplained variations, which
by their behavior in the presence of other com- was also the case with recombinant enzyme^ ^^' ^^^.
pounds, as mentioned above^^^. One group includes A major factor was the composition of the
testosterone, cyclosporin, and erythromycin. The lipid/detergent environment^^^' ^^^. Another issue
second includes midazolam, triazolam, dextro- was the NADPH-P450 reductase. P450 3A4
methorphan, and diazepam. Terfenadine fits in either expressed in yeast showed poor coupling with
group and nifedipine seemed to have properties yeast NADPH-P450 reductase^'^ although P450
unique from both groups^^^. 2C9 had coupled welP^"^. Pompon developed a
yeast system in which yeast NADPH-P450 reduc-
The ambivalence about the variability of probe
tase and Z?3 were eliminated and replaced by the
drugs is even worse for in vivo human experi-
human counterparts, enabling P450 3A4 func-
ments than in vitro, as one might expect. A num-
tion^^^. Studies with purified P450 3A4 and
ber of reactions have been used including
NADPH-P450 reductase have shown that P450
nifedipine oxidation^^^, erythromycin iV-demethy-
3A4 reduction is generally slow in the absence of
lation^^^ lidocaine oxidation^^^, dapsone A^-
substrate and greatly enhanced by substrate^^^.
hydroxylation^^^, midazolam 1'-hydroxylation^^'*,
and quinine 3'-hydroxylation^^^. In most cases, The role ofb^ in P450 3A4 reaction is a some-
the test drug is administered orally for conven- what complex subject. Some reactions of purified
ience, except for some uses of erythromycin and P450 3A4 are stimulated by b^ but others are
midazolam (i.v). The ratio of urinary 63-hydroxy- j^Ql^799,800 With reactions that are influenced by
cortisol to Cortisol has also been used to assess b^ (e.g., nifedipine oxidation and testosterone
P450 3A4 function^^^. Many of the assays reflect 6p-hydroxylation), a role for b^ could be demon-
the activity of P450 3A4 in the small intestine, strated in human liver microsomes using antibod-
particularly with the drugs administered orally. ies^^'; stimulation by b^ can also be demonstrated
The erythromycin breath test (exhaled CO2 pro- with P450 3A4 heterologously expressed in
duced from the HCHO released in the reaction) is bacterial membranes"^^^' ^^^, although the presence
generally used to estimate hepatic P450 3A4 and of Z?3 does not seem as critical for function as with
has been used as an aid in selecting cyclosporin the purified systems^^^' ^^^. Other considerations
doses for liver transplant patients^^'^. The lack of suggest that the amount of b^ in heterologous
correlation of these indicators is still a problem in expression systems should not be an issue in the
the practical analysis of drug interactions^^^"'^^^. development of "relative activity factors" for
Some of the discrepancies are probably inherent in extrapolations from hepatic systems^^^. The
the nature of P450 3A4 itself (i.e., see in vitro mechanism of stimulation of P450 3A4 activities
assays, vide supra). Other issues involve the lack by Z?3 is still not clear. Pompon used a membrane
of coordinate regulation of hepatic and intestinal system and interpreted the results in the context of
P450 3A4 (ref [791]) and the activity of P- a "classical"^^^' ^^^ transfer of an electron to the
glycoprotein^^^, which shows some overlap in Fe02^^ complex^^^. However, the activities of
regulation patterns with P450 3A4 (ref [793]) and purified P450 3A4 were stimulated by apo-Z75
influence the availability of substrates to P450 (devoid of heme) as effectively as by b^
3A4 in both small intestine and liver. (ref [808]), and similar effects have now been
Human P450s 427

reported for several other P450s (refs [425], [809], appear to accumulate in P450 3A4 reactions^^^,
[810]). Although an alternate mechanism involv- these are not well characterized yet. One approach
ing rapid transfer of P450 3A4 heme to apo-Z)^ has to analysis of rate-limiting components of P450
been proposed^ ^^ evidence ruling out this mecha- (and other enzyme) reactions is the use of kinetic
nism has been presented^^^. deuterium isotope effects^^^' ^•^^. Interpretation of
A number of models of the P450 3 A4 protein kinetic isotope effects in enzymatic reactions is a
have been presented^^^' 8i3-8i5^ j^^g^ of which are complex subject, but a simple interpretation of a
based on homology modeling. At the time of this significant intermolecular noncompetitive hydro-
proof (April 2004), the Astex company has pub- gen isotope effect is that C-H bond-breaking is at
licly announced a crystal structure for P450 3A4, least partially rate limiting^^^. Despite the interest
presumably in the absence of ligands, but the in P450 3A4, relatively few kinetic deuterium iso-
information is proprietary and no details have tope effect studies have been reported. Obach^^^
been released. reported an isotope effect of only 1.3 on the
A number of site-directed mutagenesis studies hydroxylation of the drug ezlopitant, although
on the possible roles of individual residues have details regarding k^^^ and K^ are lacking and ftir-
been published. FhQ304^^^ and Ala305 (ref [817]), ther interpretation of this result is diff'icult. Work in
in the putative I-helix, are proposed to control this laboratory with 6-(i2-testosterone has some-
access to the catalytic center. Phe304 was also what surprisingly shown a low competitive isotope
implicated in the partitioning of aflatoxin B^ oxi- effect (^Fand ^(V/K) - 3 , J.A. Krauser and
dation (between 3a-hydroxylation and S,9-exo- F.P Guengerich, unpublished results). Testosterone
epoxidation)^^^. A role for Asn206 was also 6p-hydroxylation is one of the fastest reactions
proposed in the work with aflatoxin B j (ref [818]). catalyzed by P450 3A4 and one might expect less
Leu211 is also postulated to control the size of the masking of an isotope effect in a system in which
active site^^^. other steps are known to be occurring eff'iciently.
Another issue about considerations of predict- In some of the previous sections, compounds
ing sites and rates of P450 3A4 reactions deals have been mentioned that stimulate the catal3^ic
with models based on chemical reactivity. The activities of P450s after direct addition to the
concept has been proposed that P450 3A4 has a enzyme, as opposed to regulation of genes in cells
relatively open active site and that reactions are or animals. This process is referred to as "stimula-
influenced largely by the chemical lability of C-H tion" (as opposed to induction). The phenomenon
bonds^^^, and some commercial software systems has been recognized for some time with P450s
have utilized such concepts. This approach to P450 (refs [107], [109], [827]). Conney's group demon-
catalysis may have some utility, and substrate lin- strated the activation of benzo[a]pyrene 3-hydroxy-
ear free energy relationships have been exploited lation and aflatoxin B^ activation by aNF in human
in our own research with rat P450 2B1 (ref [821]). liver microsomes^^^' ^^^. Johnson also demonstrated
However, there are some concerns about exactly the enhancement of human liver microsomal 17(3-
how appropriate the predictions are beyond spe- estradiol 2-hydroxylation by aNF^^^. Subsequent
cific sets of substrates. Knowledge of rate-limiting work in this laboratory provided evidence that P450
steps in P450 3A4 reactions is still rather rudimen- 3A4 was the human liver P450 most stimulated by
tary, in part because of some of the various com- aNFi46, 714 P45Q 3A4 is the P450 about which
plications discussed here. The rate of transfer of most of the discussion about cooperative behavior
the first electron from NADPH-P450 reductase is has been given, although some reports of coopera-
slow in the absence of substrate, but quite rapid in tivity have appeared regarding P450s 1A2, 2B6,
the presence of substrate and an equimolar con- and 2C9 (Sections 6.7.4 and 6.9.4). In addition,
centration of NADPH-P450 reductase"^^^. In liver P450 2C19 may show stimulation by some
microsomes, the testosterone-stimulated rate of compounds (R. Kinobe, B.D. Hammock, and
P450 reduction appears to be faster than the over- E.M.J. Gillam, personal communication).
all rate of 6p-hydroxylation^^^, but conclusions are In considering cooperativity, two types will be
complicated by the inability to observe only the described, using a convention we"^^^' ^^^ have
P450 3A4 component of the reduction. Although adapted from Kuby^^"^. "Homotropic" cooperativ-
some apparent (FeO) intermediate complexes do ity refers to nonhyperbolic phenomena (either
428 F. Peter Guengerich

steady-state reaction kinetics or binding) seen more substrates), and many of the data can be fit
with the addition of a single compound to an to a model with this much freedom (basically
enzyme. The most typical type is a sigmoidal, or Michaelis-Menten expression with two values
"S-shaped" curve, which when analyzed by a Hill each for k^^^ and K^, or insert of proportionality
plot (v = V* S'^/iS + S^^]) yields a value for « > 1 factors before the parameters)^^^"^"*^.
(S^^ is an approximation of the usual K^, or "pos- Halpert's laboratory has changed a number of
itive cooperativity." Examples of "negative coop- the amino acids in P450 3A4, based mainly upon
erativity" are less common but documented (n < homology modeling, and found that several can
1) as in a case with rabbit P450 1A2 (ref. [214]). alter the homotropic and the heterotropic cooper-
The other phenomenon is "heterotropic coopera- ativity. These residues include Ala305 (ref [844]),
tivity," described above as "stimulation," where Leu211, Asp214 (ref [845]), Serll9, Ala370,
two compounds are added to an enzyme and one Ile301 (ref [846]), and possibly some others as
enhances the catalytic action of the enzyme on well^"^^. A general conclusion from much of this
the other. In some cases, both homotropic and work is that two or possibly three ligands co-
heterotropic cooperativity can be operative^^^. occupy the binding site and alter each other's
Homotropic cooperativity was seen in the oxi- juxtaposition to generate some of the observed
dation of aflatoxin Bj by P450 3A4 (ref [832]). effects. One problem with much of the work in
The previously reported stimulation of P450 3A4 this field is that actual binding phenomena are
activities by aNF^^"* was not seen for some reac- not necessarily investigated. However, binding has
tions, and the A^-oxygenation of 4, 4'-methylene- been analyzed in some of the work^^^' ^^^ and
Z?/5(2-chloroaniline) was inhibited^^^. Subsequently, shown to exhibit homotropic and heterotropic
studies with aflatoxin B^ oxidation showed that cooperativity. Further, combinations of binding
3a-hydroxylation was inhibited and 8,9-(exo)- and inhibition results obtained with several lig-
epoxidation was stimulated by aNF^^^' '^^^. ands in this laboratory were consistent with a
Aflatoxin B^ (or an analog) did not modify the scheme in which three ligand subdomains exist in
5,6-epoxidation of aNF, however. Interestingly, the overall binding site of P450 3A4 (ref [831]),
the positive cooperativity seen in Hill plots for the in agreement with the current hypotheses of
oxidation of aflatoxin B^ (for both reactions) was Halpert^"*^. More evidence consistent with such a
eliminated in the presence of aNF"*^^. The values model is available from a fluorescence study by
for n in the Hill plots (2.1-2.3) are probably the Atkins and Weinkers^"^^, in which pyrene-pyrene
highest for any P450 apparent cooperativity stacking spectra were observed. This work pro-
reported to date. Most are much lower. One tech- vides some of the stronger evidence to support the
nical issue of particular note is that those reactions "multiple-substrate site" model.
that proceed too far at low substrate concentra- A crystal structure of bacterial P450 107A1
tions will show apparently low rates (due to sub- has been solved with two ligand molecules pres-
strate depletion or product inhibition) and ent^"^^. The binding titration shows homotropic
artificial sigmoidicity can be created. cooperativity^"^^ and also some heterotropic coop-
Many seemingly unusual P450 3A4 reactions erativity^^^. Because the redox partner of P450
and patterns have been reported (vide supra). 107A1 is not known, obtaining reasonable cat-
P450 3A4-catalyzed testosterone 6p-hydroxyla- alytic activity is difficult and the relevance to
tion and erythromycin A^-demethylation are not P450 3A4 is still not definite^^^.
very competitive^^^. Hydroxylation of meloxicam Another aspect and possibly another solution
is stimulated by another substrate, quinidine^^^. to the issue comes from work by Friedman using
Lu and his group showed that inhibition patterns flash photolysis kinetics (of CO rebinding after
for several known P450 3A4 reactions were photodissociation from ferrous P450 3A4). The
substrate dependent^^^. Similar discrepancies kinetics were multiphasic and were selectively
were reported by Weinkers' laboratory^^^. The altered by the presence of different substrates^^^
(mechanism-based?) inactivation of P450 3A4 by Heterotropic effects were observed with benzo[<2]-
diclofenac was stimulated by the substrate quini- pyrene and aNF^^^. The interpretation of the
^jj^g838 Qj^g interpretation of some of the results results is that different substrates differentially
is that a single active site accommodates two (or modulate these kinetics by (a) changing the P450
Human P450s 429

conformation to alter the rate, and/or (b) steric already mentioned that different protein conforma-
effects (of ligands) that reduce rates^^^. Both tions exist throughout the catalytic cycle, can differ
effects are possible, although the enhancement of in affinities and substrate orientation, and may not
rates in some cases^^' argues against the general- be in rapid equilibria. Many steady-state kinetic
ity of the latter explanation and in favor of multi- schemes have been proposed but, in considering the
ple conformations for P450 3A4 bound to various possible origins^^"*' ^^^ can never be considered
ligands. The concept advanced is that some lig- unique and do not provide mechanistic answers.
ands act as allosteric factors to "switch" P450 3A4 The availability of a series of three-dimensional
conformations^^"^. Some possibly relevant work X-ray structures of P450 3A4 with various ligands
has been done by Anzenbacherova et al.^^^, who would provide insight into the conformational
did pressure studies on P450 3A4 and found that rigidity of P450 3A4 and the number of modes of
the compressibility of P450 3A4 was less than that binding. Another possible set of experiments
of bacterial P450 102; the compressibility was involves restraining conformational changes
modified by the ligand troleandomycin (TAO). through engineering (e.g., with reversible disulfide
The concept of preexisting multiple conformers bonds) and examination of the effects. Another
of P450 3A4 is an explanation for the flash concern, already expressed here, is that most of the
photolysis work^^^"^^"^ and has support in newer studies in this field have avoided measuring bind-
nonclassical approaches to general protein ing, with some exceptions'^ ^' ^^^^ ^^^. Some attempts
chemistry^^^^^^. This view differs from the more have been made to directly quantify P450 3A4-
general static "lock-and-key" view of enzyme/ ligand interactions (e.g., dialysis and equivalent
substrate complexes and the induced-fit theory in methods), but the technical problems associated
which enzymes are "shaped" by their substrates. with equilibrium binding (e.g., insolubility and
The basic concept is protein dynamics present an nonspecific components) are not trivial. At this
ensemble of structures of an enzyme in solution time a priori prediction of cooperativity is not
and different ligands bind to individual states really possible, except perhaps in extension of
depending upon their complementation^^^^^^. chemical classes already covered. The lack of abil-
Another consideration in this discussion, some- ity to predict P450 3A4 cooperative ligand interac-
what related, is that there is good evidence that tions indicates a deficiency in being able to predict
P450 conformations change during the course of all ligand interactions.
the catal3^ic cycle^^^, and evidence has already Is the cooperativity of P450 3A4 relevant to
been presented that different forms of P450 3A4 any practical issues? Atkins'^^ has discussed the
can differ in their binding of a ligand (e.g., ferric general implications of the issue to toxicology,
and ferrous)^^^ although conclusions are speculative because the
Where does all of the work in this area to date function of P450 3A4 can be good, bad, or not
leave us? A recent review by Atkins et al.^^^ sum- really selected for anyway. Some evidence for an
marizes much of the work in more detail and pres- interaction between caffeine and acetaminophen
ents a cogent analysis. Summarizing and expanding in rat models is suggestive of a heterotropic inter-
on this, there are several major possibilities to action^ ^^' '^^. Dextromethorphan studies in pri-
explain the observed cooperativity of P450 (and the mary hepatocytes also show cooperativity^^'.
other P450s showing this behavior), which are not Cooperativity in human hepatocyte systems has
necessarily exclusive: (a) a "classic" allosteric also been reported for oxidation of midazolam and
model with binding of effectors at a site that then warfarin'^"^. Cooperativity is a possible mecha-
regulates the conformation of substrate binding, nism for a drug interaction between felbamate and
(b) a relatively rigid P450 with a large active site carbamazepine'^^. One of the strongest cases
that can accommodate 2-3 ligands, with the results involves an in vivo study on the enhancement of
depending on the chemical interactions of the two diclofenac in monkeys by quinidine^^^' ^^^, which
ligands with each other and with P450 residues; apparently cannot be attributed to induction. In
and (c) a series of preexisting conformations of summary, there is some evidence for in vivo P450
P450 3A4 that selectively interact with individual 3A4 cooperativity, but at this time, the issue is
ligands^^^^^^. A general concept of induced fit is generally considered to be less of a problem than
related to the third possibility, as in the phenomena enzyme induction or inhibition.
430 F. Peter Guengerich

6.20.5. Inhibitors gestodene^^^. Because of the very low doses of these


contraceptives that are used today, the effects might
Inhibition of P450 3A4 is a major issue in the be expected to be small^^"^ although some in vivo
pharmaceutical industry because of a number of effects have been reported^^^' ^^^.
important drug-drug interactions. One example Finally, some chemicals and also oxidants have
of a problem leading to recall of a drug is that of been shown to cause the covalent crosslinking of
terfenadineio^'104,866
heme to apo-P450 (ref. [877]). Correia's group has
Erythromycin and ketoconazole are two of the characterized the products of the destruction of
most established inhibitors of P450 3A4, based on P450 3A4 with cumene hydroperoxide; the infor-
clinical experience. Ketoconazole, used at ~ 1 JULM, mation is consistent with a dipyrrolic fragment of
is probably the best established P450 3A4 inhibitor heme bound to a fragment of the protein^^^.
for in vitro use^^. Another P450 inhibitor is TAO^^^
which also has clinical implications. TAO has been
used as a diagnostic in vitro inhibitor of P450 3A4,
6.20.6. Clinical Issues
although its mode of action (activation to a nitroso
that complexes P450 iron) requires time for the The major issues involving P450 3A4 in drug
inhibition to occur. development and clinical use are related to the role
A compendium of P450 3A4 inhibitors has been of the enzyme in drug disposition, particularly
compiled by Rendic^^. Only a few other specific bioavailability and drug-drug interactions due to
examples of P450 inhibitors will be mentioned here. induction or inhibition^^^' ^^^. High enzyme activ-
One issue is the inhibition of P450 3A4 by ity toward a drug will reduce bioavailability, and
grapefruit juice, first reported by Bailey^^^. The variations in levels of P450 3A4 can cause clinical
effect was rather specific for grapefruit and a few problems when the therapeutic window is narrow.
other citrus fruits (not orange), and warning labels For instance, low cyclosporin levels will not pre-
now include this contraindication for many vent organ rejection during transplant but high lev-
(jj^gg869 Naringenin has some effect^'^^, but the els cause renal toxicity, so adjustment of the dose
most active principles appear to be the ftirano- can be very useful^^^ Terfenadine has a relatively
coumarins bergamottin and 6',7'-dihydroxyberg- wide window for use but a few serious problems
amottin, which behave as mechanism-based were encountered ^^'^' ^^^. Renwick has considered
inactivators to destroy intestinal P450 3A4 (refs population models of P450 3A4 variability and
[99], [100]). The magnitude of the effect of the inter- concluded that there is more inter-individual
action varies with drugs, with some of the statins, variability from the oral route than i.v, which is
buspirone, terfenadine, astemizole, and amiodarone not surprising in light of the previous discussion of
reported to show the greatest interactions^^^. the intestinal contribution to drug metabolism.
Many of the HIV protease inhibitors are also A "default factor" for adults of 3.2-fold is pre-
potent inhibitors of P450 3A4 as well as substrates sented, but a factor of 12(-fold) was calculated to
in some cases^^'. Because of the variety of drugs be needed to cover 99% of the neonates as well^^^.
that AIDS patients use, the potential for interac- The effbct of disease on P450 3A4 has
tions is considerable. been considered. P450 3A4 expression appears to
The effects of some herbal medicines on P450 be decreased as a result of liver cirrhosis or
3A4 have already been mentioned. In addition to cancer^^^' '^2'' ^^^. P450 3A4 levels were also
P450 3A4 induction (e.g., St. John's wort), some decreased in celiac disease and reversed by a
of these materials also contain inhibitors. For change in diet^^^.
instance, kava-kava extracts produce kavapyrones The interactions of herbal medicines with P450
that inhibit P450 3A4 (ref. [872]). 3 A4 have already been mentioned and are one of the
Oral contraceptives contain acetylenes and worst problems with these mixtures^^^. One of the
can be mechanistic inactivators of P450 3A4. most studied issues is St. John's wort, which induces
Inactivation has been demonstrated for 17a- P450 3A4 as an agonist of the PXR receptor^^^' ^^^.
ethjoiylestradiol, the major estrogenic component The induction of P450 3A4 by St. John's wort has
of oral contraceptives^^' ^^^, and several of been responsible for the loss of the effectiveness of
the progestogenic components, particularly oral contraceptives^^' ^^^. The resulting pregnancies
Human P450s 431

are the result of contraceptive failure due to more expressed^^^. Gonzalez found a liver sample appar-
rapid elimination of 17a-ethynylestradiol, a phe- ently expressing only P450 3A5 and not 3A4, and
nomenon previously reported for P450 3A4 induc- used this to clone the cDNA^^^.
tion by rifampicin and barbiturates^^' ^4, loi P450 3 A5 expression has been reported in liver,
P450 3A4 is also of some interest regarding small intestine, kidney, lung prostate, adrenal
cancer, regarding exogenous carcinogens, drugs gland, and pituitary^ ^^' 902-904 gome researchers
used to treat cancer, and metabolism of steroids or have reported expression of P450 3A5 in periph-
other compounds that may affect cancer risk or eral blood cells (and not P450 3A4)^^^ but others
response to chemotherapy. Some chemical carcino- have not^^^.
gens activated by P450 3A4 are shown in Table 10.4. P450 3A5 is expressed in fetal liver, in contrast
The activation and detoxication of aflatoxin B^ to P450 3 A4, but in a polymorphic manner^^^. The
have already been discussed in the context of overall expression of P450 3A5 (mRNA) as a part
3a-hydroxylation (to aflatoxin Q^) and formation of all P450 3 A subfamily transcripts has been esti-
of the highly reaction exO'S,9-QpoxidQ^^^' '^^^. mated at 2%^^^. However, only about 25% of
However, aflatoxin B^ is a hepatocarcinogen and Caucasians express P450 3A5, and when it is pres-
must reach the liver to cause damage. In a rat ent, the level is usually less than that of P450 3A4.
model, induction of rat P450 led to an increase in However, a few individuals have been identified in
small intestinal DNA adducts, suggesting that acti- which P450 3A5 is the predominant P450 3A sub-
vation of aflatoxin B^ at this site constitutes a family enzyme. The variability in expression levels
detoxication process, in that these cells are rapidly has been linked to a polymorphism (vide infra).
sloughed and do not progress to tumors^^^.
CYP3A4 genotypes have been reported to be
6.21.2. Regulation and Polymorphism
related to leukemias caused by prior treatment
with epipodophyllotoxin^^^ P450 3A4 expres- The regulation of CYP3A5 gene seems to be
sion, measured at the mRNA level, has shown an similar to that of CYP3A4, although P450 3A5
inverse correlation with response of breast cancer does not seem as inducible. The fetal/adult selec-
patients to docetaxel, presumably due to changes tivity of P450 3A4/3A7 is not seen with P450 3A5
in bioavailability^^^. However, no relationships (ref. [906]).
were found for any CYP3A4 genotypes in therapy- Maurel^^^ reported genomic clones and found
related myeloid malignancies^^^. One of the more a CATA box (not TATA) in the promoter. The
controversial issues involves whether CYP3A4 responses to glucocorticoids are probably
genotypes are linked with prostate cancer, with explained by the PXR system^^^. A general con-
reports for and against an association^^"^^^^. The clusion has been reached that P450s 3A4 and 3A5
point should be made that strong evidence for a are co-regulated in the liver and intestine, in terms
change in an accepted P450 3A4 phenotype has of transcriptional control^^^, although other
not been made in many of these cases. factors may alter the expression^^^
The polymorphic variation of P450 3A5 has
6.21. P450 3A5 been studied and several variants have now been
identified (http://www.imm.ki.se/Cypalleles/)^^^.
P450 3A5 has 85% sequence identity with Alternate splicing is a very common phenomenon
P450 3A4 and, although generally accepted to with CYP3A5, with ~50% of liver samples show-
have less importance than P450 3A4, is of interest ing this (E.G. Schuetz, personal communication).
because of its polymorphic and racial distribution Most Caucasians with low P450 3A5 protein
and possible relevance to clinical issues with P450 expression have the *3 allele with an inserted
3A subfamily reactions. intron^^^' ^^^. Interesting, the representation of the
*1 allele is much higher in Africans and they
express active P450 3A5. Other alleles are known,
6.21.1. Sites of Expression and
including changes in the 5'-regulatory region
Abundance where transcription factors bind^^^
P450 3A5 ("Hlp3") was first purified from The in vivo consequences of 3A5 polymor-
human adult liver and found to be polymorphically phism are not clear. For instance, Huang found no
432 F. Peter Guengerich

significant effect of the *3 polymorphism on mentioned previously, Huang^^^ found no signifi-


midazolam pharmacokinetics^^^. cant effect of the *3 allele on midazolam pharma-
cokinetics in Chinese individuals. However, it is
possible that the extrahepatic expression^ ^^ may
6.21.3. Substrates and Reactions influence the course of particular drugs and other
Since the discovery of P450 3A5, the catalytic chemicals.
selectivity has been known to be similar to that of
P450 3A4 (ref [900]), and subsequent compar-
isons with P450 3A4 confirmed this view^^^. 6.22. P450 3A7
However, a general problem with P450 3A sub- Early work in the field of human P450
family enzymes is that they are sensitive to the research was done by Kamataki and his associates
membrane environment and many reactions of with fetal samples which led to the purification of
P450 3A5 (but not all) are stimulated by b^ (refs a P450 termed HFLa, now known as P450 3A7
[425], [800]). In a few cases, the selectivity of P450 (refs [915], [916]). Early research established that
3A5 for different oxidation sites appears to differ this was a major P450 in fetal liver (not in adult
from that of P450 3A4, for example, aflatoxin Bj liver), and that the enzyme could catalyze several
3a-hydroxylation vs 8,9-epoxidation^^^' ^^^. reactions^ ^^.
Recently, as noted above, Wrighton reported
an extensive comparison of many reactions by
recombinant P450s 3A4, 3A5, 3A7 under identi- 6.22.1. Sites of Expression and
cal reconstitution conditions and concluded that Abundance
P450 3 A5 had equal or reduced activity compared
to P450 3A4 in all cases^^l Early work established that P450 3A7 is the
major P450 present in fetal liver^^^ and is also
present in other fetal tissues including kidney,
6.21.4. Knowledge about Active Site adrenal, and lung^'^. Further work by Kamataki's
group showed the existence of some immuno-
Because of the similarity of reactions of P450s
chemically detectable P450 3A7 in gynecologic
3A4 and 3A5, homology models for P450 3A4 are
tumors and in human placenta, but interestingly
probably about as valid for P450 3A5. The avail-
not in cynomologous monkey placenta^^^.
ability of the Astex P450 3A4 three-dimensional
Guzelian's group also reported P450 3A7 protein
structure should improve the understanding of
in human placenta and endometrium, with eleva-
P450 3A5 as well. Relatively little site-directed
tion in the latter site during pregnancy or during
mutagenesis of P450 3A5 has been done, but one
the secretory phase of the menstrual cycle^^^.
study of note is the effort by Correia and Halpert
Subsequently, Sarkar et al?'^^ reported 10-fold
to utilize the differences in reactions with afla-
greater expression of P450 3A7 in endometrium
toxin B, (refs [774], [800]) to probe the effects of
in the proliferative rather than the secretory phase.
changing residues in the putative active site^^^.
Hakkola et alP^^ reported some expression of
P450 3 A7 mRNA in some first trimester placentas
6.21.5. Inhibitors but not in fiill-term placenta^"*^.
With regard to development in fetal tissues,
In general, the P450 3A4 inhibitors also inhibit Juchau's group found expression of P450 3A7 in
P450 3A5. For instance, ketoconazole and flu- early fetal tissue (50-60 days)^^^. Schuetz et al.^^^
conazole inhibited both P450s 3A4 and 3A5 (ref found P450 3A7 mRNA in all fetal liver samples
[914]). The mechanism-based inactivator gesto- analyzed and also reported its presence in one half
dene^^^ also inhibits P450 3A5 (ref [913]). of adult liver samples. However, the issue may
be the level of expression because Kamataki's
group^^ had reported the fetal > adult selectivity.
6.21.6. Clinical Issues
De Wildt et al™ also found fetal specificity and
At this point, the significance of the wide only very low levels of P450 3A7 in adults. P450
variability in P450 3A5 is difficult to assess. As 3A7 expression was high during embryonic and
Human P450s 433

fetal life, and decreased rapidly during the first Some interesting variants of CYP3A7 genes
week of life. Similar findings were reported by have been reported. An mRNA species was found
Hakkola et al?^^ Also, the variability of P450 3A7 that contains exons 2 and 13 of a nearby CYP3A
expression was 5-fold in fetal tissue (and 77-fold pseudogene spliced at the 3 end^^^. The CYP3A7*
in mRNA). In another report^24^ P45Q 3^7 ^^so IC allele is unusual in the sense that a part of the
disappeared rapidly after infancy. CYP3A4 promoter replaces the corresponding
region of CYP3A 7 (ER6 motif) and thus confers
high levels of expression to CYP3A7*1C
(ref [932]).
6.22.2. Regulation and Polymorphism
As in the case of P450 3A4, relatively little
solid evidence is available regarding the ftinc- 6.22.3. Substrates and Reactions
tional relevance of coding region SNPs. However,
the regulation of this gene is complex, as one Early studies with P450 3A7 purified from
might expect after considering the temporal pat- fetal liver established that testosterone 6p-hydrox-
terns of expression during development that were ylation is catalyzed by this enzyme^^^. Another
discussed earlier. early study indicated 16a-hydroxylation of dehy-
Kamataki's group published the cDNA^^^ and droepiandrosterone (DHEA) 3-sulfate^^'*. These
genomic^^^ sequences, which are similar to those activities were later verified with the use of
of P450 3A4. However, more identity (-90%) is recombinant P450 3A7 (ref. [935]).
seen in the coding region than elsewhere^^^' ^^^. In general, P450 3A7 has catalytic activities
Recent work by Koch et aV^^ re-established that rather similar to P450 3A4 and 3A5 (refs [936],
P450 3A7 only accounted for <2% of all P450 937]). Activation of aflatoxin B^ (refs [938-940])
expression in adult human liver; a bimodality of and heterocyclic amines^^^ has been observed in
P450 3A7 expression was seen, however. P450 various recombinant and transgenic systems,
3 A4 and 3 A7 constructs were expressed in various including transgenic mice^^^ Retinoic acid
cell lines by Ourlin et aV^^, who showed differen- 4-hydroxylation by P450 3A7 has also been
tial responses to C/EBPa and DBP As in the case reported^"^^. Wrighton's laboratory has done an
with P450 3A4, P450 3A7 was inducible by extensive comparison of catalytic activities and
rifampicin in cell culture^^^. P450 3A7 has a ftmc- concluded that rates for P450 3A7 are generally
tional PXR element^^s^ ^^ ^^^^ P450 3^4 {vide considerably lower for P450 3A7 than for P450
3A4 or 3A5 under similar conditions^^^.
supra), explaining the rifampicin response. Thus,
one would expect fetal P450 3A7 induction by the
usual P450 3A4 inducers.
Bertilsson et al?^'^ have also reported a distal 6.22.4. Knowledge about Active Site
xenobiotic response enhancer module (XREM) in Much less has been done with P450 3A7 than
the CYP3A7 gene. An 3A7^B element in CYP3A7 with P450s 3A4 and 3A5. Because the catalytic
is inactive in CYP3A4 (ref [930]), and this ele- selectivity of P450 3A7 is similar to P450s 3A4
ment has recently been shown to respond to p53 and 3A5, those models are probably about as
(T. Kamataki, personal communication). CYP3A7 applicable. One point of interest is the work of
expression is regulated by Spl, Sp3, HNF-3P, and Kamataki's group showing that the substitution
upstream stimulatory factor (USF) 1. Far upstream T485P improved holoprotein expression in
(-11 kb) there are HNF-1 and HNF-4 and USFl
elements, which differ from the CYP3A4 gene.
Exactly how these and other sequence differences
are involved in the rapid onset of P450 3A4, and
6.22.5. Inhibitors
decrease in P450 3A7 shortly after birth^^ is still not
totally clear. Recent work in Kamataki's group sug- Inhibitors have not been studied extensively,
gests that after birth, CEB/Pa recruits an uncharac- but presumably all inhibitors of P450 3A4 are eff-
terized protein factor to squelch the 3A7^B site ective with P450 3A7, for example, ketoconazole,
(T. Kamataki, personal communication). troleandomycin, etc.
434 F. Peter Guengerich

6.22.6. Clinical Issues keratinocytes^^^. In cell cultures, P450 4A11


expression has been observed in primary cultures
The general point has already been made that of human kidney proximal tubular cells^^^ and
P450 3A7 is the major human fetal P450 and, HepG2 cells956.
therefore, makes a major contribution to drug A gene originally assigned as CYP4A11 by
metabolism in the fetus. Thus, many, if not most, Kawashima's group^^^ has now been assigned
of the considerations regarding drug interactions as that corresponding to CYP4A22 and replaced
etc. with P450 3A4 should be considered with by the CYP4A11 gene corresponding to the P450
respect to the fetus during pregnancy. 4A11 CDNA952_
Another potentially important aspect is a
report that P450 3A7 expression increases in
hepatocellular carcinoma^"^^, possibly as a part of
6.24.2. Regulation and Polymorphism
dedifferentiation.
The levels of hepatic P450 4A11 vary
~ 10-fold among humans^^^' ^^^. To date no
6.23. P450 3A43 polymorphisms have been reported (April 2004;
In 2001, three groups reported the characteri- http://www.imm.ki.se/CYPalleles/). The regula-
zation of a fourth member of the 3A subfamily, tion of P450 4A11 expression is not well under-
P450 3A43 (refs [945-947]). The sequence iden- stood but has relevance in consideration of the
tity with the other 3A subfamily proteins is peroxisome proliferation system and any rele-
71-76%. Expression could be detected in liver, vance to cancer. Induction of P450 4A11 by per-
kidney, pancreas, and prostate. Rifampicin was oxisome proliferators has not been seen in
reported to induce P450 3A43 in human liver^"^^. primary human hepatocytes cultures, although
The level of expression in liver was generally P450 4A11 expression is readily detected^^^. In
agreed to be very low ( - 0 . 1 % of P450 3A4). confluent HepG2 cell cultures, P450 4A11 is
Heterologous expression was achieved in bac- induced (independently) by peroxisome prolifera-
teria^"^^ but not any of several eukaryotic sys- tors (e.g., Wy 14643) and dexamethasone^^^. The
tems^"^^. The recombinant protein had only very relevance of these results to the induction in vivo
low testosterone 6p-hydroxylation activity^"^^. and the observed variability in expression is
No information is available about polymor- unknown.
phisms, although the transcripts appear very prone
to splicing^'*^. There is a general agreement that
P450 3A43 makes little contribution to drug 6.24.3. Substrates and Reactions
metabolism, but specialized roles in extrahepatic
P450 4A11 is the major lauric acid w-hydrox-
tissues may be possible.
ylase^^^' ^^•' ^^'^^ ^^^' ^^^. The enzyme also catalyzes
(0- and some co-l hydroxylation of myristic and
6.24. P450 4A11 palmitic acids^^^' ^^^. Some papers have reported
arachidonic acid hydroxylation^^^' ^^'' ^^^, but
6.24.1. Sites of Expression and most studies have not associated this activity with
Abundance P450 4A11 at any appreciable level^^^, 957, 960
Oliw's group has reported some hydroxylation
P450 4A11 cDNAs were first cloned from of prostaglandin H2 and analogs by P450 4A11
human kidney cDNA libraries using rodent P450 (ref [963]).
4A probes^'^^-^^^. P450 4A11 is now known to be
the major lauric acid w-hydroxylase in human
liver and kidney^^^' ^^^, a fact of some historical
6.24.4. Knowledge about Active Site
interest in the sense that this was the activity first
utilized in the separation and reconstitution of Some information is available about the active
(rabbit) P450 (ref [953]). The exact level of site from the catalytic selectivity among fatty acid
expression in these tissues is unknown; P450 substrates^^^. Some homology models have been
4A11 expression has also been reported in human presented^o^'964.
Human P450s 435

The interesting observation was made that the 4A22 in human liver^^^. P450 4A22 expression
LI3IF mutant catalyzes only w-l hydroxylation could not be observed in HepG2 cells or PPARa-
and not oo-hydroxylation of lauric acid^^^. Residue overexpressing cells^^^.
131 also controlled access to substituted imida-
zole inhibitors. Interestingly, some of the results
on binding of imidazoles provide evidence that the
ferric enzyme undergoes a conformational change 6.26. P450 4B1
that depends on both reduction of the iron and the
6.26.1. Sites of Expression and
presence of both NADPH-P450 reductase and
Abundance
NADPH959.
Another interesting observation is that P450 P450 4B1 was cloned by Nhamburo et alP^^
4A enzymes, including P450 4A11, show at least from a human lung cDNA library. P450 4B1 expres-
partial covalent heme attachment^^^. Covalent sion has also been reported (in addition to lung)
heme binding involves a conserved I-helix glu- in kidney, bladder^^^, breast^^^, and prostate^^^.
tamic acid (apparently unique in the P450 4A sub- Expression has also been reported in bladder and
family) and covalent heme binding occurs via an breast tumors^^^. Definitive information about the
ester bond to the heme 5-methyl group, mediated level of expression of P450 4B1 is not available.
by an autocatal3^ic process^^^. The extent of effect
of this modification on catalytic activity is diffi-
cult to define, although with animal P450 4A 6.26.2. Regulation and Polymorphism
enzymes, there appears to be some effect.
The extent of variability of P450 4B1 expres-
sion is considerable, at least in bladder where the
6.24.5. Inhibitors variation is two orders of magnitude^^^. Several
SNPs have been reported, including one resulting
Substituted imidazoles have been used as in a premature truncation^^^.
inhibitors in vitro^^^. Presumably some acetylenic No evidence for the inducibility of human 4B1
fatty acids might be inhibitors but no studies have has been presented.
been reported.

6.26.3. Substrates and Reactions


6.24.6. Clinical Relevance
Direct information about the catalytic speci-
The significance of P450 4A11 is not very
ficity of human P450 4B1 has been difficult to
clear. Apparently individuals can vary in their
obtain because of problems in heterologous expres-
expression levels by an order of magnitude^^^.
sion. Following the initial cDNA cloning, expres-
Further, the w-hydroxylation of medium chain
sion in a baculovirus system was unsuccessful and
fatty acids occurs, but its relevance is generally
only yielded inactive cytochrome P420 (ref. [971]).
considered not to be as important as in the case of
The substitution S427P allowed for expression, and
long chain fatty acids.
o)-hydroxylation of lauric acid could be demon-
strated. However, information about the native
human enzyme has not been available (the S427P
6.25. P450 4A22
mutant does not occur naturally^^^).
Relatively little is known about P450 4A22. Imaoka et alP^^ found that functional P450
The originally reported CYP4A11 gene^^^ was 4B1 could be successfully expressed as a fusion
subsequently shown to be CYP4A22 (ref. [952]), protein with NADPH-P450 reductase. They were
but the cDNA and protein have not been reported. also successful in developing transgenic mice in
The similarity of the two genes is 95%. which fiinctional P450 was expressed in liver. The
Johnson's laboratory^^^ has reported that P450 authors postulate that expression in the presence
4A22 is expressed at lower levels than P450 4A11 of auxiliary proteins (NADPH-P450 reductase,
in human liver, as well as kidney^^^. There was no b^) may stabilize P450 4B1 (ref [972]). With
correlation of expression levels of P450 4A11 and these systems, it was possible to demonstrate that
436 F. Peter Guengerich

P450 4B1 catalyzes the iV-hydroxylation of expression was distinct from P450 4F3, which is
2-aminofluorene and w-hydroxylation of lauric acid, restricted to polymorphonuclear leukocytes. P450
as expected from studies with rabbit P450 4B1. 4F2 is found not only in liver but in several extra-
If other results from work with animal P450 4B1 hepatic tissues, however^ •^^, including kidney (S2
enzymes also carry over to the human QnzymQS, one and S3 segments of proximal tubules, in cortex
might expect the reaction 4-ipomeanol activation and outer medulla). The extent of variation of
(epoxidation?), 3-methoxy-4-aminoazobenzene P450 4F2 in human liver was -S-fold^^^.
7V-hydroxylation, 2-aminoanthracene A^-hydroxyla- P450 4F2 catalyzes co-hydroxylation of several
tion, valproic acid hydroxylation (and desatura- lipids, including leukotriene B^ (refs [979], [980]),
tion?), and dehydrogenation of 3-methylindole^^^. arachidonic acid^^^, 6-^ra«5-leukotriene B^, lipoxin
A^, 8-hydroxyeicosatetraenoic acid, 12-hydroxy-
eicosatetraenoic acid, and 12-hydroxystearic
6.26.4. Knowledge about Active Site acid^^^ The physiological relevance of some of
Because of the paucity of information about these reactions is of interest but the effects of vari-
catalytic selectivity (vide supra), little can be said ability of P450 4F2 have not been demonstrated.
about the active site of human P450 4B1. Some Part of the interest lies in the fact that leukotriene
kinetic hydrogen isotope effect work with rabbit B4 is a potent proinflammatory agent^^^' ^^^.
P450 4B1 suggests that the active site is more
restricted than that of P450 2B1 (ref [973]).
Another interesting observation with rabbit P450 6,28. P450 4F3
4B1 is the covalent linking of the heme to the pro-
tein^^^, a phenomenon observed with several of In 1993, Kikuta et al!^^^ cloned a P450 now
the P450 4 family proteins^^^' ^^^ Whether this known as P450 4F3 from a human leukocyte
binding is seen in human P450 4B1 is unknown. cDNA library. The protein was expressed in a
yeast vector system and was shown to catalyze
leukotriene B^ a)-hydroxylation. The K^ (0.7 JULM)
6.26.5. Inhibitors was much lower than that reported for P450 4F2
for this reaction (although the k^^^ and k^JK^
No inhibitors of human P450 4B1 have been
values have not been carefully compared)^^^.
reported.
The gene was cloned in 1998^^^. Interestingly,
the CYP4F3 gene has been shown to use tissue-
6.26.6. Clinical Issues specific splicing and alternate promoters. A 4F3A
form contains exon 4 (but not 3) and is expressed
There are two issues with P450 4B1. First, the in neutrophils; a 4F3B form contains exon 3 (but
enzyme has been shown to activate carcinogens, not 4) and is expressed in fetal and adult liver and
for example, 2-aminofluorene, and could be a risk kidney, trachea, and gastrointestinal tract^^"^' ^^^.
factor in bladder cancer^^^. The level of P450 4B1 The K^ of the 4F3B (liver) form was 26-fold
in tumorous tissue was not higher than in the sur- higher than for the 4F3A (neutrophil) form^^^,
rounding tissue, levels of bladder P450 4B1 were although the significance of this report is quali-
higher in tumor patients than in controls^^^. fied by the absence of k^^^ or k^JK^ parameters.
The other aspect is the use of P450 4B1 as a Further studies by Soberman's group have shown
means of drug delivery. Rabbit P450 4B1 has been that the substitution of exon 3 changes the cat-
utilized as an experimental transgenic activation sys- alytic selectivity from leukotriene B^ to arachi-
tem in the activation of 4-ipomeanol and 2-amino- donic acid (w-hydroxylation in both cases)^^^.
anthracene, to date only in cell culture models^"^^. Again, the usefulness of the observation is limited
by the lack of kinetic parameters. The relevance of
the preferential localization^^^ and altered cat-
6.27. P450 4F2
alytic selectivity are presently unknown but there
The Kusunose laboratory reported the cloning is potential clinical relevance in light of the known
of a human liver cDNA corresponding to the physiological action of both leukotriene B4 and
leukotriene B^ o)-hydroxylase^^^. The site of 20-hydroxyeicosatetraenoic acid.
Human P450s 437

6.29. P450 4F8 20 (ref [991]), hydroxylation of the antihistamine


ebastine^^^, and w-oxidation of leukotriene B^
Bylund et al?^^ used degenerate PCR primers (refs [990], [991]), and w-hydroxylation of some
and isolated a cDNA from human seminal vesi- prostaglandins and prostaglandin analogs^^^.
cles, denoted P450 4F8. This P450 was shown to No further information is yet available about
be a 19-hydroxylase with prostaglandin endoper- the relevance of this enzyme in physiological or
oxides^^^' ^^^ Recombinant P450 4F8 catalyzed the clinical situations.
a)-2 hydroxylation of arachidonic acid and three
stable prostaglandin H2 analogs but prostaglandins
D2, Ej, E2, and F2^ and leukotriene B^ were poor 6.32. P450 4F22
substrates^^^. (19^)-Hydroxy prostaglandins E^
No information is available except the exis-
and E2 are the main prostaglandins of human sem-
tence of the CYP4F22 gene in the human
inal fluid. Bylund et al.^^'^ propose that a)-2
genome^^^.
hydroxylation of prostaglandins H^ and H2 by
P450 4F8 occurs in seminal vesicles, and that iso-
merization to (19/?)-hydroxy prostaglandin E is the 6.33. P450 4V2
result of action of prostaglandin E synthase.
Further investigations by Bylund and Oliw^^^ No further information is available except for
have demonstrated the expression of P450 4F8 the existence of the human CYP4V2 gene^^^.
protein in human epidermis, hair follicles, sweat
glands, corneal epithelium, proximal renal
tubules, and epithelial linings of the gut and uri-
6.34. P450 4X1
nary tract. P450 4F8 was shown to be upregulated Relatively little is known beyond the existence
(mRNA and protein) in the epidermis in psoria- of the human CYP4X1 gene except for one recent
sis^^^. The exact physiological role of P450 4F8 is paper on rat P450 4X1 (ref [993]). mRNA expres-
unclear, although 19-hydroxy prostaglandins do sion was highly selective in brain (cortex, hip-
have several biological activities^^^. pocampus, cerebellum, brainstem). The rat protein
was expressed in yeast but has not been examined
for catalytic activity.
6.30. P450 4F11
P450 4F11 is another member of the CYP4A 6.35. P450 4Z1
gene cluster found on chromosome 19^^^.
Expression has been demonstrated primarily in The only information available is the existence
liver, with some expression also in kidney, heart, of the CYP4Z1 gene in the human genome^^^.
and skeletal muscle. No other information is
presently available, although it might be expected
to be capable of leukotriene hydroxylation based 6.36. P450 5A1
upon its similarity to other P450 4F enzymes. P450 5A1 is the classification of thromboxane
synthase, which converts prostaglandin R^ ^^
thromboxane (Figure 10.12). Thromboxane
6.31. P450 4F12 causes vasoconstriction and platelet aggregation,
P450 4F12 was originally cloned from human which are of considerable interest.
liver^^^ and small intestine^^^ cDNA libraries.
Expression has been demonstrated in liver, kidney, 6.36.1. Sites of Expression and
colon, small intestine, and heart^^^' ^^^ Actual lev-
Abundance
els of abundance are unknown, although this
would appear to be a minor P450. P450 5A1 is expressed in platelets and also
Two groups have expressed P450 4A12 in erythroleukemia cells^^^. The enzyme is also
yeast. Catalytic activities include the hydroxylation found in human monocytes^^^, leukocytes^^^, and
of arachidonic acid at carbons 18 (ref [990]) and kidney interstitial dendritic reticulum cells
438 F. Peter Guengerich

P450 5A1
a
O,,

P450 8A1

Fe'V-Q

'<x: homolytic
0 - 0 cleavage
Fe'v-d

Fe'^a
rearrangement
to C» radical
R2
electron -Fe"
transfer

carbocatlon
formation
+

o.O
stabilization
to product HO"

PGI2

Figure 10.12. Rearrangement of prostaglandin H2 to prostacyclin (PGI2) by P450 8A1 and thromboxane (TXA2)
by P450 5A1 (ref. [994]).

surrounding the tubules^^^. Some expression is factor NF-E2 is critical for both alteration of the
also seen in lung and liver^^^. nucleosomal structure and activation of the P450
5A1 promoter^ooo
Chevalier e^ a/.'^^' identified 11 polymorphic
6.36.2. Regulation and Polymorphism variants in the CYP5A1 gene, including 8 mis-
As one might expect from its function, P450 sense changes in the coding region. The effects of
5A1 is a highly regulated system. Dexamethasone these changes have not been reported yet.
induces P450 5A1 in human monocytes^^^.
Phorbol esters also induce P450 5A1 (e.g., \2-0- 6.36.3. Substrates and Reactions
tetradecanoylphorbol-13-acetate) in human ery-
throleukemia cells^^^. Patients with systemic The thromboxane synthase reaction has been
sclerosis showed 6-fold enhanced levels of leuko- known for many years but was associated with a
cyte P450 5A1 (ref. [997]). P450 by Ullrich and his associates, first in spectral
Promoter analysis indicates a 39-bp core pro- studies ^^^^ and then by purification^^^^. With
moter, containing TATA and initiator elements that the purified enzyme or one expressed in
control transcription. Binding of the transcription a baculovirus system^^^^, prostaglandin H2 was
Human P450s 439

converted to thromboxane A2 and 12-hydroxyhep- search for inhibitors as drug candidates has
tatrienoic acid (HHT) plus malondialdehyde, in continued^^^^.
equimolar amounts ^^^^. Prostaglandin G2 was
transformed to malondialdehyde and the corre-
sponding 15- and 12-hydroperoxy products. 6.36.6. Clinical Issues
Prostaglandin Hj was enzymatically transfor- As indicated earlier, platelet aggregation due
med into 12(Z)-hydroxy-8, 10-heptadecadienoic to the produced thromboxanes is important but
acid, and prostaglandin H3 yielded thromboxane overproduction can yield plugs, so control of
B3 and 12(L)-hydroxy-5,8,10,14-heptadecate- homeostasis is desirable.
traenoic acid^^^^ (Figure 10.12).
These are all rearrangement reactions, not
involving input of O2 or electrons from pyridine 6.37. P450 7A1
nucleotides. The reaction of the "oxygen-surro-
P450 7A1 catalyzes cholesterol 7a-hydroxyla-
gate" iodosylbenzene with a P450 5Al-containing
tion, the rate-limiting step in bile acid synthesis.
preparation and the stable prostaglandin H2 analog
Much has been done in several animal models,
15(*S)-hydroxy-l la,9a-epoxymethano-5(Z), 13(E)-
including purification of the enzyme from rabbit
prostadienoic acid (U46619) yielded three oxida-
and rat liver^^^^' ^^^^. The enzyme was partially
tion products (that could also be formed in a
purified from human liver^^^^ and the cDNA was
similar system using rat liver microsomes)^^^^. cloned by several groups in 19901016-1018
These and other studies led Hecker and Ullrich^^^''
to propose a mechanism involving homolytic
cleavage of the prostaglandin endoperoxide (with 6.37.1. Sites of Expression
the Fe^^ bonded to one oxygen and the other oxy-
gen bearing a radical), transfer of the radical to a Apparently the only site of P450 7A1 expres-
carbon, further electron transfer to generate Fe"^ sion is the liver. The CYP7A1 gene is on chromo-
plus a carbocation, and collapse of the bis-ionic some 8 q l l - q l 2 and contains recognition
structure to yield thromboxane A2 (ref [994]). sequences for a number of liver-specific transcrip-
Fragmentation competes with the electron transfer tion factorsioi9-io2i
step to also yield malondialdehyde and hepta- The level of the enzyme in liver appears to be
trienoic acid^^^. similar to some of the low-to-moderately abun-
dant xenobiotic-metabolizing enzymes in liver.

6.36.4. Knowledge about Active Site


6.37.2. Regulation and Polymorphism
Relatively little is known about the active site
The regulation of the CYP7A1 gene is very
of P450 5A1 beyond th^ information about the
complex, as might be expected from the important
reactions presented above. As indicated, the pro-
physiological role this enzyme plays.
tein does not bind NADPH-P450 reductase.
P450 7A1 activity has long been known to be
Presumably, the active site is rather specific,
upregulated by dietary cholesterol in most animal
although iodosylbenzene could be utilized as an
models^^^^, although there are some excep-
oxygen surrogate.
tions ^^^^. Feeding rats the competitive inhibitor
7-oxocholesterol led to reduced bile acid synthesis
6.36.5. Inhibitors (due to inhibition) and a compensatory increase
in P450 7A1 synthesis^^^s chiangi024 identified
Thromboxane synthase inhibitors have been a a bile acid-responsive element in the CYP7A1
matter of interest for many years because of their promoter.
potential use in preventing plugs of platelets, and Studies with P450 7A1-knockout mice show
efforts at development preceded the characteriza- that this reaction (cholesterol 7a-hydroxylation) is
tion of the enzyme as a P450 (refs [1008-1010]). essential for proper absorption of dietary lipids
Many of these inhibitors have a basic nitrogen and fat-soluble vitamins in newborn mice, but not
atom that binds to the P450 5A1 heme^^i^. The for maintenance of cholesterol and lipid levels^^^^.
440 F. Peter Guengerich

The mice exhibit a complex phenotype with increased (rat) CYP7A1 expression in a reporter
abnormal lipid excretion, skin pathologies, and assay^^"^"*.
behavioral irregularities. The cholesterol levels In addition to the mouse CYP7A1 knockouts,
were not altered. Interestingly, vitamin D3 and E work has been done with overexpression in
levels were low to undetectable. j^j^gi045, 1046 j^Q j^j^,g ^ j ^ jjQt exhibit altered
A new era in the regulation of P450 7A1 began cholesterol levels ^^'^^. The lack of an LXR element
with reports of the involvement of some of the in a region (—56 to -49) of the human promoter
orphan steroid receptors. The proximal promoter may dictate some of the differences seen in mouse
region interacts with LXRa. The oxysterols 24(S)- and human models. With regard to humans, one
hydroxycholesterol and 24(»S)-epoxycholesterol study of biopsy samples from gallstone patients
activate LXRa (and LXRP)^^26 Further, mice led to the conclusion that there was no correlation
devoid of LXRa fail to induce CYP7A1 transcrip- between levels of total bile acids and P450 7A1
tJQj^i027 j^Q other proteins, FXR and CPF, are activity^^'^''. A correlation was seen with levels of
also involved^^^^"^^^^. Chenodeoxycholate, a bile chenodeoxycholic acid.
acid derived from cholesterol, interacts with A long-standing observation from rodent stud-
FXR to suppress CYP7A1 transcription^^^^ ies is the apparent circadian rhythm of P450 7A1
However, the action of FXR has been reported to (ref [1048]). This phenomenon has been sug-
be indirect^^^^ PXR binds lithocholic acid and gested to be indicative of a short halflife of the
downregulates CYP7A1 (ref [1032]). Thus, cho- enzyme^^^^' *^^^. The phenomenon has also been
lesterol metabolites control their synthesis in the reported in nonhuman primates^^^^. The circadian
liver through feedback suppression of CYP7A1 rhythm can be demonstrated at the level of actual
(ref [1028]). Hylemon^^^^ has concluded that the P450 7A1 in rats^^^^. The molecular mechanism
dominant factor is LXRa. CPF binds to the of the rhythm is still not clear. One aspect is the
promoter (as a monomer) and leads to CYP7A1 instability of P450 7A1 in microsomes {in vitro),
transcription^ °^^. with a /,/2 of ~ l - 2 h r in humans and rats^^^^.
Other studies have addressed the role of Alternatively, the mRNA has a short /,/2 and the
PPARa in P450 7A1 downregulation'^"^^. circadian rhythm can be seen at the mRNA
However, differences exist between humans and jgyg|i054 Another unresolved aspect of P450 7A1
mice gene responses have been observed, with the research is the issue of phosphorylation, postu-
mouse gene showing an enhanced response to lig- lated early in the field^^^^. In vitro experiments
ands because of an additional binding site'^^^ (fur- with microsomes show some effects of various
ther, humans have much less PPARa than treatments^^^^' '^^^. More recent work with micro-
rodents^o^^). Chiang^^^^ analyzed the PPARa somes and recombinant proteins also shows
response and provided evidence that the downreg- effects'^^^, although the in vivo significance is yet
ulation by PPARa-agonist complex is due to com- unclear.
petition with HNF-4 for the DR-1 sequence. Polymorphisms in the coding and noncoding
The regulation of P450 7A1 by other factors has regions of the CYP7A1 gene are known'^^^. Some
been considered. Downregulation by TNFa has have been associated with clinical changes'^^^, but
been interpreted in the context of MEKKl, an others have not'^^'.
upstream nitrogen-activated protein kinase, affect-
ing HNF-4 (ref [1038]). The same mechanism may
6.37.3. Substrates and Reactions
be involved in the repression by endotoxin and
interleukin-1 (ref [1039]). A novel CYP7A1 site The classic reaction of P450 7A1 is choles-
appears to be involved in the repression of CYP7A1 terol 7a-hydroxylation^^, and esterified choles-
by thyroid hormone (T3)^^'^^. Studies with rats indi- terol is not a substrate ^^^^. However, recent
cate differences in the regulation of P450 7A1 and experiments have established that the enzyme
P450 27A1, a sterol 27-hydroxylase^^4i Human also catalyzes the 7a-hydroxylation of
CYP7A1 expression is also repressed by insulin and 24-hydroxycholesterol, with preference for the
phorbol esters *^^^. Estrogen (100 juig/kg/week) (»S)-isomer^^^^. 7a-Hydroxylation (with recombi-
increased hepatic cholesterol 7a-hydroxylation 2.7- nant human P450 7A1) was observed with 20(5)-
fold in ovariectomized baboons ^^^^. Retinoic acid hydroxycholesterol, 25-hydroxycholesterol, and
Human P450s 441

27-hydroxycholesterol^^^^. The relevance of interpretation of results of family and experimen-


the activity toward 25(»S)-hydroxycholesterol is tal studies with P450 7A1.
unknown compared to P450 39 (ref. [1065]).

6.37.4. Knowledge about Active Site 6.38- P450 7B1


Relatively little has been done with site- Almost all of the work with P450 7B1 is from
directed mutagenesis or modeling. As indicated rodent models and application to the human
(vide supra), the enzyme only catalyzes 7a- CYP7BI gene system is by inference. A P450 7B1
hydroxylation but is not very sensitive to side- transcript was first characterized in a rat (brain)
chain hydroxyIs. hippocampus cDNA library^^^^. A heterologously
The region 214-227 has been postulated to expressed protein was shown to catalyze the 7a-
interact with the membrane and to serve as a hydroxylation of the steroids DHEA and preg-
substrate-access channel ^^^^. Mutations in this nenolone ^^^^. Expression has also been reported in
region yielded some changes in kinetic parameters liver and kidney^^^^' ^^'^^. Disruption of the mouse
toward cholesterol. CYP7B1 gene yielded animals that were viable
and apparently normal, but ex vivo 7a-hydroxyla-
tion of DHEA and 25-hydroxycholesterol was
6.37.5. Inhibitors blocked in brain, spleen, thymus, heart, lung,
Limited information about inhibitors is avail- prostate, uterus, and mammary gland^^^^.
able. As indicated earlier, 7-oxocholesterol is a (n) Although extrapolation to humans has not
(competitive) inhibitor ^^^^. been reported, P450 7B1 is considered to be a
neurosteroid hydroxylase and have a potentially
important role^^^^' ^^^^. Functional polymorphisms
6.37.6. Clinical Issues in the human CYP7B1 gene have not been
reported, but have been postulated to lead to
P450 7A1 has been a topic of considerable inter-
severe hypercholesterolemia and neonatal liver
est in the areas of hepatology and gastroenterology.
disease^^.
The hypersecretion of cholesterol in obesity
does not appear to be due to reduced 7a-hydroxy-
lation^^^^. Coffee terpenes (e.g., cafestol) inhibit
P450 7A1 and also raise cholesterol levels^^^^, 6.39. P450 8A1
although it is not clear that the two phenomena are
linked. The complex regulation of P450 7A1 Prostacyclin (prostaglandin I2) has strong
makes interpretation of some experiments diffi- vasodilation and anti-aggregation effects on
cult. Overexpression of P450 7A1 in HepG2 platelets, and the imbalance of prostacyclin and
cells increased bile acid synthesis but led to thromboxane A2 (product of P450 5A1) is a factor
decreased hydroxymethylglutarate (HMG) CoA in several diseases, for example, myocardial
reductase activity (rate-limiting step in cholesterol infarction, stroke, atherosclerosis^^^^' ^^^^. The
biosynthesis) ^^^^. reaction yielding prostacyclin from prostaglandin
Alterations in P450 7A1 were not seen in H2 is another "internal" oxygen transfer, without
hypo- or h5q)erthyroidism^^^^. the input of O2 and electrons from NADPH
A 10-week old child with a stop-codon muta- (Figure 10.12), and the involvement of a P450 was
tion and lacking P450 7A1 presented with severe not immediately obvious. Ullrich hypothesized
cholestasis, cirrhosis, and liver synthetic fail- P450 involvement on the basis of spectral interac-
^j.gi060 ^ frameshift leading to (homozygous) tion studies^o^l DeWitt and Smith^^^^ used a
lack of P450 7A1 was associated with high low- monoclonal antibody to purify catalytically active
density lipoprotein (LDL) cholesterol, but not prostacyclin synthase from bovine aorta and
total cholesterol^^^^. Heterozygotes were also demonstrated a P450 Fe^^'CO spectrum.
hyperlipidemic. However, Beigneux et al}^^'^ have Subsequently, P450 8A1 was cloned from bovine
discussed some of the caveats associated with endothelial cells^^^^.
442 F. Peter Guengerich

6.39.1. Sites of Expression and the corresponding prostacyclins ^^^^. Spectral bind-
Abundance ing studies with 9,11-epoxymethano prostaglandins
F2 and F2^ lead to the view that the binding juxta-
Human P450 8A1 was cloned from aorta position is the key determinant in distinguishing the
endothelial cells by Tanabe's group ^^^^. The courses of catalysis by P450s 5A1 and 8A1 (ref
mRNA is widely expressed in human tissues, [1007]). A mechanism consistent with available
including ovary, heart, skeletal muscle, lung, data has been proposed (Figure 10.12)^^"*' ^^^^.
prostate^^''^, and umbilical vein^^^^ More recent
work has shown some localization in the brain,
including neurons^^^^' ^^^^. Another site of expres- 6.39.4. Knowledge about Active Site
sion is fallopian tubes, with expression in luminal
epithelia, tubal smooth muscle, vascular endothe- Mutagenesis of Cys441 (heme binding Cys) or
lial cells, and vascular smooth muscle cells^^^"^. Glu347 or Arg350 (EXXR motif) abolished cat-
alytic activity, suggesting that the placement of
these residues is correct'^^^. Other site-directed
6.39.2. Regulation and Polymorphism mutagenesis studies suggest roles of Ile67, Val76,
Leu384, Pro355, Glu360, and Asp364, which have
P450 8A1 is constitutively expressed in human been suggested in models'°^^. However, the level
endothelial cells^^^^. The human CYP8A1 gene of residual activity was 5-10% and only a single
(chromosome 20) has 10 exons^^^^"^^^^ and has substrate concentration was used; another caveat
consensus sequences for Spl, AP-2, an interferon-7 is that the expression work was done in COS cells
response element, GATA NF^B, a CACCC box, and the level of expression of holoprotein was not
glucocorticoid receptor, and a shear stress respon- measured.
sive element (GAGACC)!^^^ Whether or not all Other work has been on membrane topology,
of these are functional and how they interact to and antibody studies indicate that P450 8A1 is
maintain constitutive expression is not well under- mainly exposed on the cytoplasmic site of the
stood yet. endoplasmic reticulum with a single transmem-
Polymorphisms have been of interest because brane anchor^ ^^^' •^^^. The (unstable) substrate,
of disease relevance. The coding sequence con- prostaglandin H2, is produced in the lumen and
tains at least five alleles^^^^. In the 5'-region, these apparently passes through the membrane to reach
are polymorphisms involving a variable number of P450 8A1. Antibodies raised to the peptides of the
tandem repeats (VNTR) that affect transcription, putative substrate channel (66-75 and 95-116)
as demonstrated in reporter systems in vitro^^^^. At interact only after membrane solubilization,
least nine of these allelic variants are known'^^^. implying that the substrate-access channel is very
An association between this VNTR polymorphism near the membrane'^^^.
and cerebral infarction has been reported •^^^.
A SNP in exon 8 has been reported to be linked
to myocardial infarction, although no amino acid 6.39.5. Inhibitors
change occurs'^^^ However, the VNTR polymor-
phism does not appear to be related to essential Relatively little interest has been shown in the
hypertension'^^^, nor does the 5'-flanking region development of drugs that inhibit P450 8A1
SNP T192G (ref [1093]). However, a novel splic- because inhibition is generally considered to be
ing variation leading to skipping of exon 9 has deleterious.
inhibit^^oo
Phenylbutazone has been reported to
been linked to hypertension'^^'^.
P450 8A1 is slowly inactivated during the
normal reaction itself, apparently by one of the
reactive intermediates in the catalytic cycle
6.39.3. Substrates and Reactions
(Figure lO.ny^'K A )^,,^,,,^,^„ of 0.06 s'^ was
P450 8A1 has a very limited catalytic speci- reported^^^^
ficity, functioning only as the prostacyclin synthase Peroxynitrite is a powerful inhibitor of P450
(Figure 10.12). Prostaglandins G2, H2, 13(5)- 8A1, with a reported K. of 50 nM^^^^
hydroxy H2, 15-keto H2, and H3 are isomerized to Peroxynitrite is formed by the chemical reaction
Human P450s 443

of NO* and O2' (ref. [1103]). The mechanism is Regulation of the gene is of interest, in that
believed to involve tyrosine nitration^ ^^"^j and P450 8B1 catalyzes the synthesis of cholic
recently Tyr430 has been implicated as the site of acid and controls the ratio of cholic acid to
nitration^ *^^. chenodeoxycholic acid in the bile^^^^. HNF-4a
activates human CYP8B1 expression in HepG2
cells^^^^. Bile acids and farnesoid X receptor
6.39.6. Clinical Issues (FXR) downregulate HNFa expression. Inflam-
As mentioned earlier, prostacyclin is a power- mation in liver cells causes increased synthesis of
ful vasodilator and inhibits platelet adhesion and a J-antitrypsin, a serum protease inhibitor, and in a
undesired cell growth. Although this view may be derived peptide (C-36). C-36 appears to interact
overly simplistic, prostacyclins are a counterbal- with the a J-fetoprotein transcription factor (FTF)
ance to thromboxanes in a "yin-yang" relation- site in the human CYP8B1 promoter, inducing a
ship. Thus, the action of P450 8A1 balances that conformational change to lower DNA binding
ofP450 5Al. ability, and suppressing the transcription of the
Decreased expression of P450 8A1 has been CYP8B1 (and CYP7A1) genes^^^^' ^^^^. HNFa
reported in severe pulmonary hypertension^ ^^^. could overcome the inhibitory effects of FTF and
With regard to general cardiovascular disease, a bile acids^i^^ Thus, regulation of P450 8B1 is
study of Japanese subjects associated the VNTR involved in bile acid feedback inhibition.
polymorphism with hypertension (odds ratio
1.9)^^^^. Individuals with 3 - ^ repeats had less
promoter activity and higher risk. In experimen-
tal studies, the overexpression of P450 8A1 in 6.41. P450 11A1
transgenic mice protected against the develop-
P450 11 Al is the enzyme involved in the initi-
ment of hypoxic pulmonary hypertension^ ^^^. In
ation of steroid synthesis (Figures 10.13 and
another study, the expression of human P450
10.14). It catalyzes the conversion of cholesterol
8A1 in the carotid arteries of rats after arterial
to pregnenolone by side-chain cleavage and has
balloon injury (using a virus) led to increased
been referred to in the older literature as P450^^^
synthesis of prostacyclin and to reduced neointi-
or cholesterol desmolase. The enzyme was puri-
mal formation^ ^^^.
fied from bovine adrenal cortex mitochondria^ ^^^.
P450 8A1 also has relevance in cancer treat- The human gene was cloned by Omura and Fujii-
ment. Transfection of colon adenocarcinoma cells Kuriyama in 1987^^^^ and includes nine exons. Of
with P450 8A1 led to slower growth and reduced historical significance is the fact that this P450
vascular development following inoculation into only contains a single cysteine and further estab-
syngeneic mice^^^^. lishes the position of the heme thiolate peptide
Finally, antibodies in the sera of some patients in P450s, extending the work on the location
with hypersensitivity reactions to phenytoin and from the crystal structure of bacterial P450 101
carbamazepine recognize rat P450 3A1 but not (ref [1118]).
human P450 3A (ref [1111]). The antisera also
recognize peptides derived from P450s 8A1 and
5A1, although relationships of etiology and
causality are unclear. 6.41.1. Sites of Expression
P450 11 Al is found primarily in steroidogenic
6.40. P450 8B1 tissues, that is, adrenal cortex and gonads, includ-
ing ovary (corpus luteum^^^^' ^^^^ and theca
P450 8B1 is a sterol 12a-hydroxylase interna cells^^^^ and others^^^^). Of interest are
expressed in the liver. The human CYP8B1 gene recent reports of P450 l l A l in brain^^^^-^^^^ and
was characterized on the basis of the rabbit and pancreas^ ^^^.
mouse orthologs^^^^. Of interest is the finding that P450 l l A l is one of the few P450s localized
this gene is devoid of introns, unique for this gene in the mitochondria (Table 10.1 and Figure 10.14).
among the P450 family^ ^^^. Studies with the bovine enzyme demonstrated that
444 F. Peter Guengerich

P450 21A2 ) (P450 11B1


Progesterone ^ Deoxycorticosterone • Corticosterone

(M5017A1^ ,P45017A1^
I > 4 5 0 21A2;
17-OH Pregnenolone 17-OH Progesterone "—^ 11-Deoxycortisol
18-OH Corticosterone

Cortisol Aldosterone

Testosterone

P45019AO

Estradiol

Figure 10.13. A view of the metabolic pathway of steroidogenesis and the major P450s involved^^.

P45011A1 P45017A1 P45017A1


Cholesterol • Pregnenolone 17-OH Dehydroepi-
Pregnenolone androsterone
3li-0H 3/3-OH 3/3-OH
Steroid D.H. Steroid D.H. Steroid D.H.
P45017A1 P450 17A1
Progesterone • 17-OH T >- Androstenedione
I Pregesterone
P450 21A2 P450 21A2 P45019A1

P450 P450
11B2 11B1 Estrone
18-OH Cortico^^^ Deoxy- 11-Deoxy-
Corticosterone sterone"^ corticosterone cortisol
P450
11B2
P450
11B1
Aldosterone Cortisol

Mitochondrion Endoplasmic reticulum

Figure 10.14. Overview of steroidogenic pathway and cellular compartmentalization^^.

P450 l l A l , synthesized on ribosomes in the efficient mitochondrial import^ ^^^. Miller's group
cytosol, is imported into mitochondria without constructed vectors that could be used to direct
processing of the amino terminal extension pep- P450 l l A l to the endoplasmic reticulum and
tide^'^^. The protein moves to the mitochondrial found that the enzyme was inactive' '^^. The mem-
inner membrane and is then cleaved to yield the brane environment was concluded to be more
mature form^^^^. Alteration of the basic amino important in modulating catalytic function than
acid residues of the A^-terminus resulted in less the electron transfer partners.
Human P450s 445

6.41.2. Regulation and Polymorphism intermediate^ ^^^. Oxidative cleavage of the


diol to pregnenolone and 4-methylpentanal
The regulation of P450 1 lAl is relatively com-
(isocaproic aldehyde) completes the overall
plex, as might be expected for the initial step in
reaction. The mechanism of the last step is not
steroid formation^ ^^^. Moreover, the system must
completely clear, but some proposals have been
be able to respond to signals in many different tis-
presented^ ^'*^~^^^^.
sues. Much of our understanding of the regulation
The rate of electron transfer from adrenodoxin
of P450 1 lAl expression is based on studies with
is important and appears to be the rate-limiting
CYPllAl genes of experimental animals and rein-
step for the enzyme in human placenta^ ^'^^. The
vestigated with human CYPllAl.
redox potential of adrenodoxin can be varied by
P450 l l A l has long been known to be regu-
site-directed mutagenesis, but had little effect on
lated by ACTH and cyclic AMR In the bovine
rates of electron transfer, consistent with the view
CYPllAl gene two Spl-binding sites mediate
that other factors such as protein-protein inter-
cyclic AMP transcription through the protein
actions are more important than the intrinsic ther-
kinase A signaling pathway, utilizing the rather
modynamics ^^ 47 y^YiQii P450s 1 lAl and 1 IBl are
ubiquitous transcription factor Spl (ref [1131]).
expressed together in cells, they can compete for
The steroidogenic factor 1 (SFl) activates
reducing equivalents from adrenodoxin^ ^'^^;
CYPllAl transcription through interaction with
exactly how important the competition is in tis-
protein factors upstream^ ^^^. An upstream CREB-
sues is unclear. Another report indicates interac-
binding region and an AP-1 site are also involved
tion of P450 l l A l with and enhancement by b^
in the cyclic AMP response. Sp3 can also be
(ref [1149]) although the relevance is unclear
involved^ ^^^. The TATA box drives cell type-
because of the compartmental separation of P450
specific cyclic AMP-dependent transcription^ ^^^.
11 Al (mitochondria) and b^ (endoplasmic reticu-
SF-1 also interacts with Spl (refs [1134-1136]).
lum) (Figure 10.14).
Thus, the regulation of the human CYPllAl gene
involves all the above factors plus an AdE ele-
j^gjj^ii22 More recently, the expression of the
human gene has been shown to involve the zinc 6.41.4. Knowledge about
finger protein TreP-132, interacting with both Active Site
CBP/p300 (ref [1137]) and SF-1 (ref [1138]).
Also, salt-inducible kinase (SIK) represses cyclic Knowledge about this enzyme is still relatively
AMP-dependent protein kinase-mediated activa- limited. Studies with bovine P450 l l A l indicated
tion through the CREB basic leucine zipper the significance of Lys377 and Lys381 in adreno-
domain^ ^^^. doxin binding^ ^^^. As indicated earlier, a mutation
at Arg353 was found to attenuate the function
Other recent work with human placenta show
of P450 l l A l in a patient^^^i site-directed
that activating protein-2 (AP-2) assumes the role
mutagenesis of human P450 l l A l (in E. coli)
of SF-1 by binding to an overlapping promoter
indicated that Ile462 had some effect on kinetic
elementii40.
parameters^ ^^^
Mutations are also found in CYPllAl and can
cause congenital adrenal insufficiency. Arg353
was found to be critical in a study with an afflicted
patient^ ^41 6.41.5. Inhibitors
A number of inhibitors of P450 l l A l have
been reported, although some were studied only
with the bovine enzyme ^^^^' ^^^^, including some
6.41.3. Substrates and Reaction
acetylenic mechanism-based inactivators^^^"*.
P450 l l A l appears to be quite specific in With regard to the human enzyme, there is some
using cholesterol as a substrate. The reaction potential for use of inhibitors in treatment of pro-
proceeds in a three-step sequence, with genera- static cancer, and prodrug forms of amino-
tion of (22i?)-20a, 22-dihydroxycholesterol as an glutethimide have been examined^ ^^^.
446 F. Peter Guengerich

Anti-convulsants have been reported to inhibit the early research in this field was done with
P450 1 lAl, but the interaction is not strong^ ^^^. bovine adrenal glands because of the need for
large amounts of material, and the bovine
P450 IIB protein has the function that P450
6.41.6. Clinical Issues l l B l (11-hydroxylation) and the P450 11B2
(11-hydroxylation, 18-hydroxylation, and oxida-
Two major issues are of interest. Because of tion of the 18-alcohol to an aldehyde) have in
the nature of P450 1 lAl in initiating steroidogen- most other species, including humans^^^^. The
esis, deficient P450 11 Al can lead to (congenital) two human genes (CYPllBl, CYP11B2) were
adrenal hyperplasia^^. Rabbit and mouse models characterized and clearly shown in both to be
show the effects^i^^' ^^^l C7P77^7-null mice die essential^ 16^-^ ^^^
shortly after birth, but can be rescued by steroid P450 1 IBl expression has also been deleted in
injection* ^^^. ACTH levels become very high due human fetal adrenal gland, particularly in the
to lack of feedback regulation by glucocorticoids. "fetal zone" (as opposed to neocortex)^^^^.
Male null mice are feminized with female external
genitalia and underdeveloped male accessory sex
organs. These manifestations resemble various 6.42.2. Regulation and Polymorphism
human steroid deficiency syndromes. Much of the background on regulation of P450
Another issue is autoantibodies to P450 11 Al l l B l came from studies with the bovine gene,
(and also P450 17A1) in patients with autoim- which responds to ACTH and has six c/^-acting
mune polyglandular syndrome types I and II, and regulatory elements^^^^. The protein (Ad4BP) that
Addison's disease**^^' **^°. As with other P450s binds to one of these (Ad4) is a member of the
recognized by autoantibodies, causal relationships steroid hormone receptor superfamily^^^^. Other
between immunity and disease are unclear. studies by Omura^^^^ indicated the cooperative
nature of these elements in transcription. Work
with rat CYPllBl showed that ACTH stimulates
transcription by changing composition in AP-1
6.42. P450 11B1 factors (Fos, Jun)*'^^
P450s l l B l and 11B2 differ in only 32 The human gene also has a cyclic AMP
residues. P450 1 IBl catalyzes the 11 p-hydroxyla- response element (CRE)"^^. The Adl element
tion of deoxycortisol to yield Cortisol, which is the bound CRE-binding protein, activating transcrip-
main glucocorticoid in the body. Deficiencies in tion factor-1 (ATF-1), and ATF-2. Steroidogenic
the enzyme are known, causing congenital adrenal factor-1 (SF-1) interacted at the Ad4 site
hyperplasia^^' **^*. (-2427-234) and was required for transcrip-
^JQj^ii72, 1173^ which contrasts with the lack of
response of C7P7752.
Many mutations are known because of the
6.42.1. Sites of Expression
relationship of the gene with congenital adrenal
P450 1 IBl is expressed in the adrenal cortex, hyperplasia''^^ These include a 5-base duplica-
specifically the zonafasciculate/reticularis^^^K In tion"'^'^ and clusters of mutations in exons 6-8
rats, some expression has been detected in brain (ref [1175]). The high similarity and proximity of
but the relevance is not clear. the CYPllBl and CYP11B2 genes appear to lead
P450 l l B l is synthesized in the cytosol and to mutant generated by unequal crossover and
directed to the mitochondria with a 24-residue A^- inactive chimeric product "^^""^^. Splice donor
terminal targeting sequence (where this is lost after site mutations are also known"^^.
entry). As with the other four (exclusively) mito-
chondrial P450s (Table 10.1 and Figure 10.14),
P450 l l B l receives electrons fi-om adrenodoxin
6.42.3. Substrates and Reactions
instead of NADPH-P450 reductase.
The characterization of the CYPllB gene has As indicated previously, the only substrate for
developed considerably in recent years. Much of P450 l l B l is deoxycortisol, which undergoes
Human P450s 447

11 P-hydroxylation to yield Cortisol (Figures 10.13 excess 11^^' ^^^^. Overproduction of glucocorti-
and 10.14). coids, which could have any of several causes
including overactive P450 l l B l , is associated
with Cushing's syndrome^i^i.
6.42.4. Knowledge about Active Site
One of the concerns about studies on the func-
tion of particular residues in site-directed mutagen- 6.43. P450 11B2
esis is that expressions in some cellular systems
lead to competition between P450s 1 lAl and 1 IBl P450 11B2 is highly related to P450 l l B l
for (adrenodoxin) reducing equivalents in cellular {vide supra) and has a somewhat similar function.
systems^ ^'^^. Another issue is that human P450s P450 11B2 catalyzes the 11 (B-hydroxylation of
l l B l and 11B2 have been difficult to express in 11-deoxycorticosterone followed by 18-hydroxy-
bacteria, so that most experiments have relied on lation and 2-electron oxidation of the 18-alcohol
to an aldehyde (Figures 10.13 and 10.14).
mammalian cells {Schizosaccharomyces pombe has
Changes in the gene can lead to corticosterone
provided some success^ ^^^). Nevertheless, much
methyloxidase deficiency and hyperaldostero-
information about function has been obtained from
nism29' 1165, 1192, 1193 i^ tjjg Qi^g^ literature, this
patients' samples ^ ^ ^ I
The close similarity of P450s l l B l and 11B2 P450 is sometimes termed as "P450 , ,^."
(and their reactions) has also facilitated studies.
Making the changes S288G and V320A yielded
an enzyme with both P450 1 IBl and 11B2 activ- 6.43.1. Sites of Expression
ities^^^^. Changes at positions 147 (refs [1182],
[1183]), and 301/355 (ref [1184]) have also had P450 11B2 is expressed in the adrenal cortex
the same effect. (zona glomerulosa) and is involved in the synthe-
sis of aldosterone (the lip-hydroxy, 19-aldehyde
Homology models of P450 l l B l have also
product). It is a mitochondrial P450, as are the
been published^^^^' ^^^^^ ^^^^, although the effects
other 11 family P450s. The cDNA was first cloned
of all of the mutants known to alter function have
from an adrenal tumor of a patient suffering from
not been systematically rationalized.
primary aldosteronism^ ^^'*. Another early study
showed higher levels of P450 11B2 in aldos-
6.42.5. Inhibitors terone-secreting tumors^ ^^^.
There is some evidence for the synthesis
Compared with some of the other steroido- of aldosterone outside of the adrenals, and Li
genic P450s, there is some reason to develop P450 et al}^^^ reported expression of P450 11B2 in
1 IBl inhibitors. High levels of Cortisol are associ- hepatic stellate cells of liver; the activation of
ated with Cushing's syndrome^ ^^^ Cellular these cells is a key event in liver fibrogenesis.
expression systems have been set up to assay for
inhibitors, using measurements of concentrations
o f s t e r o i d s ! 187, 1188

18-Vinylprogesterone and 18-ethynylproges-


6.43.2. Regulation and
terone have been reported to be mechanism-based Polymorphism
inactivators of bovine P450 IIB, but apparently Some of the research on regulation overlaps
have not been tested with human P450 1 IBl (ref that presented for CYPllBl {vide supra). A
[1189]). CRE/Adl element and ATF-1 (and ATF-2?) play a
role with both CYPllBl and CYP11B2 (ref.
[1197]). However, SF-1 does not appear to regulate
6.42.6. Clinical Issues
CYP11B2, in contrast with CYPllBl (ref [1173]).
As indicated previously, the main issue with Many aspects of regulation remain to be further
P450 l l B l is the impaired synthesis of Corti- investigated, including the mechanisms of the
sol and congenital adrenal h3q)erplasia, character- observed Ca^^ and cyclic AMP signalingn^^ and
ized by hypertension and signs of androgen the effects of kinase inhibitors^ ^^^' ^^^^.
448 F. Peter Guengerich

As in the case of CYPllBl, many mutations (ref [1208]), although that paper reported that
have now been defined from clinical studies. The the enzyme used catalyzed l i p - , 18-, and 19-
"crossovers" between P450s 1 IBl and 11B2 yield hvdroxvlatinn
hydroxylation
inactive P450 11B2, as well as P450 l l B l (refs
[1178], [1179], [1201], [1202]). Other mutations
in CYP11B2 were associated with corticosterone 6.43.6. Clinical Issues
methyloxidase I and II deficiency^^^^' ^^^^' ^^^^.
Polymorphisms in CYP11B2 have also been The issues of congenital adrenal hyperplasia
linked to idiopathic hyperaldosteronism, a condi- and Types I and II corticosterone methyloxidase
tion characterized by autonomous production of deficiency in individuals with attenuated P450
aldosterone and arterial hypertension^^^'^. A poly- 11B2 activity have already been mentioned. The
morphism in the promoter region of CYP11B2 other issue also mentioned is elevated aldosterone.
(—344 TK) has been associated with predisposi- Several studies have reported an association
tion to essential hypertension^^^^. between polymorphisms and essential hyperten-
sion, although the measurements of aldosterone
excretion are still lacking in some studies ^^^^.
Other studies show association of the -344C allele
6.43.3. Substrates and Reactions
with increased left ventricular size^^^^"^^^^. The
P450 11B2 catalyzes the three-step conversion hypertension association has been seen in several
of 11-deoxycorticosterone to aldosterone, with 11 (3- studies^^^^' *2'^' ^2^^' ^2^^, but not in a Japanese
hydroxylation, 18-hydroxylation, and 2-electron study^^^^.
oxidation of the 18-carbinol (Figures 10.13 and
10.14). No other substrates are known. Information
about the processivity of the human enzyme 6.44. P450 17A1
(i.e., extent of release of intermediate products) is
not available at this time. 17-Hydroxylation and the 17, 20-lyase reac-
tion C'desmolase") are two important reactions in
steroid biosynthesis (Figures 10.13 and 10.14).
Cloning of a cDNA which, when expressed,
6.43.4. Knowledge about Active Site
yielded both activities that established the role of
Some studies with the closely related P450 1 IBl what is now known as human P450 17A1 (previ-
have already been mentioned. Bemhardt's labora- ously termed P450,7^, etc.)'2»^ The gene^^n
tory found that changes only at positions 320 and showed similarity to CYP21AL The demonstra-
335 conferred some 18-hydroxylation activity to tion of both catalytic activities in a single protein
P450 l l B l (ref [1184]). Homology modeling has established work previously done with purified
also been done''^^' ^^^^. In other site-directed muta- hog protein^^'^. The two activities have long been
genesis work, residues that differed among species known to be regulated by b^ (refs. [1219], [1220]),
were changed and residues 112, 147, and 152 and aspects of this duality of ftinction still remain
were found to have effects'^^^. Modeling suggested unclear.
an indirect effect of residue 147 and that residue
112 might be in the substrate-access channel.
6.44.1. Sites of Expression
Human P450 17A is expressed in steroidogenic
6.43.5. Inhibitors
tissues, including adrenals and gonads. The enzyme
Elevated aldosterone levels can be detrimental has also been reported in fetal kidney, thymus, and
and some interest exists in targeting P450 11B2. A spleen^^^^ The enzyme has also been found in
yeast system has been developed that can be used human (adult) heart^^^^ and adipose tissue^^^^.
for screening for inhibitors ^^^^. P450 17A1 is a microsomal enzyme. A proline
The literature contains one older report of the rich region in the A/-terminus has been found to be
use of an acetylenic P450 19 inhibitor to inhibit important for efficient folding, but not for subse-
the activity of what may have been P450 l l B l quent maintenance of the folded structure ^^^^.
Human P450s 449

6.44.2. Regulation and Polymorphism mechanism of the lyase reaction is not com-
pletely established, but mechanisms have been
As with the other steroidogenic P450s, the reg- proposed using analogs ^244 Lieberman^245 j^^g
ulation of the CYP17A1 gene is relatively com- proposed alternative reactions, although the
plex. Induction of P450 17A1 has long been favored pathway involves what would be a very
known to be cyclic AMP-mediated and the induc- unstable diradical. No other substrates are known
tion is suppressed by testosterone (mouse presently, other than pregnenolone and proges-
model)*^^^, and a cyclic AMP response region was terone and possibly closely related analogs.
mapped in porcine Leydig cells ^^^^. Very recently, Soucy et al}^^^ have provided
With the human CYP17AI gene, the homeo- evidence that human P450 17A1 also converts
domain protein Pbxl was shown to interact with pregnenolone into 5,16-androstadien-3p-ol, a
protein kinase A in the cyclic AMP-dependent "16-ene synthase" reaction (without intermediate
regulation (at -2507-241)^227 YurthQT analysis formation of an alcohol).
showed interaction at a cyclic AMP-related site A key to research on the protein was the devel-
(-80/-40) by SF-1 (ref [1228]). Further, inter- opment of a robust E. coli expression system by
actions were shown for Spl and Sp3 Waterman's group in 1991^^. Further work on the
(-227/-184) and NF-IC ( - 1 0 7 / - 8 5 and differential effects of b^ on individual catalytic
-1787-152)1229 sp_j (yide supra) also interacts activities has been reported^247 j ^ ^ J.^^JQ ^f ^^ ^^
with p54"*, NonO, and protein-associated splic-
P450 is high in testis and this phenomenon might
ing factori23o j^Q ACTH/cyclic AMP response is
regulate the two activities of P450 17A1. Miller's
dependent upon phosphatase activity, as well as group has proposed that phosphorylation of Ser
kinase activity^^si, 1232 jj^g cyclic AMP-depend- and Thr residues in P450 17A1 may alternatively
ent protein kinase enhances transcription via influence the two activities^248^ although any
MKP-1 activation, involving phosphorylation of experimental evidence in support of this hypothe-
SF-1 (ref [1233]). sis is still very limited^248,1249
Polymorphisms of CYP17A1 are known, but A second h^ gene has been identified recently,
most of the attention has been given to mutations and this protein also has the same stimulatory
that result in serious defects in patients ^^34 Most effect on lyase activity^250 Auchus et al.^^^ also
of the mutations have been SNPs in the coding demonstrated that the same stimulatory effect of
region 1234-1236^ ^^^ others include a 2-bp deletion b^ could be obtained with apo-Z?^, arguing against
yielding a frameshift and premature stop the requirement for electron transfer. P450 17A
codon^^^^^ a 4-bp duplication changing the C-ter- enzymes from other species vary in their ability to
minal 28 amino acids^^^^^ and a 5'-splice site catalyze the 17,20-lyase reaction, and compar-
mutationi239 Some of the patients presenting with isons of the rat and human enzymes also led to
symptoms yielded P450 17A1 that, upon heterol- the conclusion that selective enhancement of the
ogous expression, retained 17-hydroxylation but
lyase reaction was not due to changes in electron
not 17,20-lyase activity^^^o. 1241 Mutations of the transfer ^251
latter type led Auchusi242 to propose a model in
The concertedness of the P450 17A1 lyase
which neutralization of positive charges in the
reaction has been examined, and both the studies
redox partner binding surface of P450 17A may
both reached the conclusion that much of the 17a-
block the lyase activity but not 17-hydroxylation.
hydrox3^regnenolone dissociates ^252, 1253 jj^ ^j^^
of the studies^252^ ^^^ authors concluded that the
off-rate was an important factor in determining
the balance between 17-hydroxypregnenolone and
6.44.3. Substrates and Reactions
DHEA. Exactly how b^ would control this rate,
The generally accepted reactions of P450 which was modeled to be rather slow (2.6-
17A are the 17a-hydroxylation of pregnenolone 29min~^), is unclear, unless the effect is on the
to 17-hydroxypregnenolone and of progesterone protein conformation. However, a classic burst
to 17-hydroxyprogesterone. 17-Hydroxypregne- kinetic experiment was not done in the cited work
nolone is also oxidized to DHEA in the lyase and the h)q)othesis remains to be addressed in
reaction (Figures 10.13 and 10.14)^241,1243 J^IQ more detail.
450 F. Peter Guengerich

6.44.4. Knowledge about Active Site 6.44.6. Clinical Issues


Much of the information about the signifi- P450 17A1 has a central role in human
cance of active site residues comes from the steroid metabolism because of its role in regulat-
analysis of mutations in patients presenting with ing steroid flux (Figures 10.13 and 10.14).
diseases (see Section 6.44.2.). The changes Perturbations lead to problems in adrenarche,
H373L (ref [1254]) and P409R (ref [1255]) led to aging, and polycistric ovary syndrome ^^'^^' ^^^^.
a loss of heme incorporation. Mutation at Thr306, Some of the more serious mutations have been
possibly involved in protonation of Fe-00~ or mentioned already; another is a case of pseudo-
O-O cleavage, impaired 17a-hydroxylation more hermaphroditism due to lack of lyase activity^^^^.
than the lyase reaction^^^^. However, the change Some of the other possible disease conditions
R346A selectively abolished lyase activity^^^^, as or risks are being studied in relationship to less
did F417C (ref [1258]). Mutations at Lys83, serious polymorphisms. In most of these cases,
Arg347, Arg358, and Arg449 produced proteins the relationships are more difficult to establish in
that were refractory to b^ stimulation and attenu- the serious diseases. A possible link of CYP17A1
ated in lyase activityi259-i26i o f these, only R347H polymorphism has been made with rheumatoid
and R358Q have been found in patients'^^^. Some arthritis^ ^^^. Little influence of polymorphism was
mutants found in patients do cause the loss of seen on age of menarche^^^^. However, a link was
both 17-hydroxylation and the lyase reaction, made between a particular polymorphism and the
howeveri263, i264^ prediction to use hormone replacement therapy
Some animal P450 17A1 enzymes have (i.e., postmenopausal estrogen therapy)^^^^. No
different ratios of 17-hydroxylation/lyase activity association was found with polycistronic ovarian
and efforts have been made to use these properties syndrome in a study with an SNP at the regulatory
to define more elements controlling the latter Spl sitei29o.
steps, although the results have been limited to Much attention has been given to the possibil-
datel265, 1266^ ity of a link between CYP17A1 allelic SNPs and
Several homology models of human P450 breast cancer risk^^^^ The epidemiology results
17A1 have been published and some of the muta- are mixed at best^^^^"^^^^ and a conclusion in
genesis results can be interpreted^ ^^^' 1267-1271 favor of a relationship cannot be made at this
time'296-1298^
Some positive epidemiology has been pre-
sented for a relationship with prostate cancer^^^^,
6.44.5. Inhibitors although probably not a strong one'^^^. Some pos-
Inhibitors of P450 17A1 have been studied for itive results have also been reported for endome-
some time. Interestingly, ketoconazole inhibits trial cancer and CYP17AI alleles ^ 3^'.
lyase activity but not 17-hydroxylation activ- As with some other P450s, circulating anti-
bodies to P450 17A are seen in some autoimmune
ity^ 7a-Thiospirolactone is a mechanism-
diseases, for example, autoimmune polyglandular
based inhibitor of (guinea pig) P450 17A1 (ref
syndrome and Addison's disease^ ^^^' ^^^^, but no
[1273]).
causal relationship has been demonstrated.
A number of steroidal inhibitors have been
studied, primarily with the goal of treating
cancers ^^^^~'^^^. The enantiomer of progesterone
(e«f-progesterone) is reported to be a competitive
inhibitor of P450 17A {K. = 0.2 |ULM)1279
6.45. P45019A1
Nonsteroidal inhibitors have also been P450 19A1 is the classic "aromatase," often
Studiedl280,1281_ known by that name in endocrinology. This
Molecular modeling (Section 6.44.4) has also enzyme oxidizes the androgens androstandione
been applied to searches for inhibitors ^^^^' ^^^^. and testosterone to the estrogens estrone and
Other approaches utilize P450 17A expressed in 17p-estradiol, respectively (Figure 10.15). This
E. coli to screen for P450 17A inhibition in process is very important in normal physiology
medium-to-high throughput systems^^^^' ^^^'^. and also a target for inhibition in some tumors.
Human P450s 451

Figure 10.15. Aromatization reactions catalyzed by P450 19. The three distinct steps are shown, with the possible
substrates: Testosterone, Rj: -OH, R2: H; androstenedione, Rj.- ^ O , R2: -OH; 16-hydroxytestosterone,
R,—R~ -OH.

6.45.1. Sites of Expression In adipose tissue, the promoter from exon 1.4 is
utilized^^^^. The same exon is utilized in bone and
Estrogens have a number of functions and not skin^^^^, and in leiomyoma tissue derived from
only feminization. Sites of (human) expression myometrium^^^^. This system is regulated with
include the ovaries, testes, placenta, fetal (but not Spl, a glucocorticoid regulatory element, STAT3,
adult) liver, adipose tissue, chondrocytes and and possibly PPAR7^^^^' ^^^^. Pre-adipocytes also
osteoblasts of bone, vasculature smooth muscle, involve regulation with liver receptor homolog-1
and several sites in brain, including parts of the (LRH-l)i309.
hypothalamus, limbic system, and cerebral cor- In placenta exon I.l, an 89 kb upstream ele-
I^g^i303 ^g discussed later, regulatory mechanisms ment is utilized^^^^. This is a strong promoter and
differ considerably in these tissues. P450 19A1 is involves C/EBP-p^^^^. A strong positive enhancer
also expressed in some tumors, particularly those element between —42 and - 5 0 1 is present^^^^.
derived from these tissues. The possibility exists that vitamin D receptor/
The actions of androgens and estrogens in the RXRa heterodimers and PPAR7 may have
gonadal tissues are fairly well understood, but less eflFectsi303.
is known in the brain. Androgens and androgen- Regulation in bone uses exon 1.6 (ref. [1303]).
derived estrogens regulate complementary and The study of regulation in bone is less extensive
interacting genes in many neural networks^^^"^. than in other sites, and 1,25-dihydroxycholecalcif-
erol, interleukins, TNFa, and TGF-p^ have had
stimulatory activity.
6.45.2. Regulation and Polymorphism
Regulation in brain uses exon I.f and has also
The regulation of the CYP19A1 gene is quite not been as extensively studied^^^^. P450 19A1
complex, primarily because of the use of four does seem to be upregulated by androgens.
tissue-selective promoters^^^^' ^^^^. Either the LI, Regulation in fetal liver involves exon 1.4, as
1.4, I.f, or 1.6 sequence is read as exon I and with adipose tissue ^^^^. The same pattern appears
spliced into the mRNA, depending upon the tis- to apply in skin fibroblasts and intestine.
sue. However, exon I does not code for the protein, In cancer cells, alternate regulatory pathways
so the P450 19A1 enzyme is always the same. are utilized^ ^^^.
In preovulatory follicles and corpora lutea of A number of polymorphisms have been
human ovary, the 5'-untranslated region of P450 reported in the CYP19A1 gene. These have been
19A1 transcripts is encoded by exon Ila (ref studied in relationship to breast but without
[1306]). The major operatives here are CRE and convincing relationships {vide infra)\ also, there
SF-1 elementsi303_ was no relationship with breast density^^^^
452 F. Peter Guengerich

The number of cases of serious P450 19A1 Another report suggests generation of
deficiency is apparently only about ten, including inhibitory Michael agents from prostaglandin J2,
two adult males^^^^. but detailed characterization has not been
done^326
Other nonsteroidal inhibitors of P450 19A1 are
6.45.3. Substrates and Reactions also under consideration^^^^.
The reaction involves three steps and has been Breast cancer is the major target area for P450
the subject of considerable mechanistic interest 19A1 inhibition, but other cancers are also under
(Figure 10.15). Androstanedione is converted to investigation^ ^^^.
estrone, testosterone to 17p-estradiol, and 16- The point has been made by Simpson et alP^^
hydroxy DHEA to estriol. The first two steps are that a future goal of P450 19A1 inhibition should
relatively straightforward, for example, RCH3 -> be tissue selectivity. The diverse role of P450
R C H 2 0 H ^ C H 0 (at CI9). The third step was 19A1 in different tissues might indicate that gen-
difficult to rationalize with "classic" FeO^^ chem- eralized inhibition of estrogen synthesis may be
istry, and there has been general acceptance of a less than desirable. Targeted inhibition of P450
FeOO~-based mechanism originally developed by 19A1 could, in principle, be achieved by (a) selec-
Robinson^^^^ and Akhtar^^^^, and further devel- tive targeting of inhibitors of P450 19A1 catalysis
oped in models by Coon and Vaz^^^"^. to tumors/individual organs or (b) targeted down-
regulation of P450 19A1 synthesis in selected
The possibility of utilization of DHEA as a
areas.
substrate for estrone synthesis has been proposed
but not addressed directly^^^^.
6.45.6. Clinical Issues
6.45.4. Knowledge about Active Site Several clinical issues have already been
alluded to. The first issue is congenital aromatase
One of the historic problems in studying
deficiency. Serious cases in adults appear to be
structure-function relationships in P450 19A1
relatively rare'^^^' ^^^^ and have been treated with
has been the availability of expression systems.
estrogen replacement therapy'^^^. However, some
Recently two E. coli systems have been
children are considered to have attenuated P450
developed^^^^' ^3^^.
19A1 activity'3^'.
Site-directed mutagenesis has been done using
Studies with P450 19A1 knockout mice show
mammalian and insect cell-based systems, and
expected reproductive and sexual phenotypes and
models have been in existence for some time'^^^.
also adipose and bone phenotypes'^^^' '^^^, as well
More recent modeling work'^^^ has been done, with
as a sociosexual behavior phenotype'^^^.
an emphasis on docking of inhibitors in SRS-1.
The issue of using P450 19A1 inhibitors in the
treatment of a variety of estrogen-dependent tumors
6.45.5. Inhibitors has already been presented. In addition, there is
consideration of the use of inhibitors for breast
P450 19A1 inhibitors have long been of interest cancer prevention in high-risk individuals'^^'^.
for treatment of estrogen-dependent cancers in a A number of studies have been made on the
variety of tissues^^' ^^^^. Today, the process has relationship of CYP19A1 polymorphisms with
reached the stage of "3rd-generation" inhibitors^^^^ breast cancer, but the evidence has not shown a
moving beyond early drugs such as amino- change in risk'^''' '^^^. No strong association was
glutethimide^^^^. The newer inhibitors are more seen for endometriosis either'^^^.
effective in lowering the body limit of estrogens ^^^^.
One example of a newer drug is exemestane, a
mechanism-based inactivator currently in Phase III
6.46- P450 20A1
clinical trials and being compared with the estrogen
receptor antagonist tamoxifen^^2^~^^^^. The inactiva- At the time of writing this chapter, the only
tion appears to involve generation of a Michael available information was the existence of the
acceptor in the active site^^^^. CYP20 gene in the human genome^^^. The gene
Human P450s 453

appeared to be vertebrate specific and had been hyperplasia, and many mutations are now
speculated to be involved in development. known to be associated with the disease. Many are
the result of recombination with the related
pseudogene^^"^^' ^^^^. Some are in the coding
6.47. P450 21A2 ' and the 5'-flanking region^^'*^. The
region^
P450 21A2 is the enzyme involved in the 21- incidence of carriers of congenital adrenal hyper-
hydroxylation of progesterone and 17-hydroxy- plasia is 1-2% in the population, and many delete-
progesterone, yielding deoxycorticosterone and rious mutations have now been identified^^^^"^^^•^.
11-deoxycortisol from the two substrates, respec-
tively (Figures 10.13 and 10.14). The 21-hydroxy-
6.47.3. Substrates and Reactions
lation reaction is an important step in the synthesis
of glucocorticoids and mineralcorticoids, and The only known substrates are progesterone
deficiencies lead to "salt-wasting syndrome," if and 17-hydroxyprogesterone, which are hydroxy-
not treated, and to congenital adrenal hyperplasia lated only at the 21-position (Figures 10.13 and
in the worst cases. 10.14).

6.47.1. Sites of Expression 6.47.4. Knowledge about Active Site


The major site of expression is the adrenal cor- Homology models have been reported^^^^' ^^^^.
tex. This reaction has been known for some time, The amount of site-directed mutagenesis has been
and many of the early biochemical studies were limited, but the disease has yielded many locations
done with bovine adrenals because of the need for for loss of function because the severity of the dis-
large amounts of tissue ^^^^. ease is (inversely) correlated to the residual 21-
Low amounts of P450 21A2 have been hydroxylation activity. Many of the mutants could
reported in human lymphocytes^^^^ and brain^^^^. be rationalized in the context of a homology
Any specific function in these tissues is unknown model^^^^, although some associated with disease
at this time. are more subtle (e.g., E380D).

6.47.2. Regulation and Polymorphism 6.47.5. Inhibitors


The regulation of P450 21A2 has some similar- Relatively little has been published about
ity to that of P450 17A1, in that both are regulated inhibitors. Detrimental effects of spironolactone
by ACTH. The cyclic AMP responsive sequence in have been attributed to inhibition of 21-hydroxy-
the 5'-flanking region^^"^^ uses adrenal-specific lation^^^^, although further details with this P450
protein factor and an Ad4-like sequence ^^"^^ are lacking. Recently, Auchus^^^^ reported that the
One issue in the regulation of the CYP21A2 gene is enantiomeric form of progesterone (ew^proges-
the neighboring homologous but nonfunctional terone) is a competitive inhibitor of P450 21A2
CYP21A1 pseudogene, which can compete for (although not as effective as with P450 17).
transcription factors and other regulatory pro-
teins ^^^^. In other work, protein kinases A and C
6.47.6. Clinical Issues
and Ca^^ were found to regulate CYP21A2 gene
expression in a human cortical cell line^^^^. As mentioned earlier, the incidence of defects
Another interesting aspect of the regulation of is relatively frequent and the ability to form Corti-
the CYP21A2 gene is that it is located very close sol is a problem. At least 56 different mutations
to the major histocompatibility locus, 2.3 kb have been identified^ ^^^ Patients who cannot syn-
downstream from the C4 gene. Transcriptional thesize sufficient aldosterone may lose sodium
regulatory elements for the CYP21A2 gene lie balance and can develop a fatal "salt-wasting"
within intron 35 kb of the C4 gene^^"^"^. syndrome. Treatment involves administration of
Steroid 21-hydroxylase deficiency is the mineralocorticoids and glucocorticoids. Females
most common cause of congenital adrenal with severe, classic P450 21A2 deficiency are
454 F. Peter Guengerich

exposed to excess androgens prenatally and bom 6.48.2. Regulation and Polymorphism
with virilized external genitalia, but prenatal diag-
nosis permits prenatal treatment of affected The regulation of the CYP24A1 gene appears
females^^^^. Experimental research is being done to be complex, although some phenomena
on gene therapy to transfer active CYP21A2 observed in animal models have not been exam-
genes; work done on mice suggests feasibility^^^^. ined in as much detail in humans. The activity has
long been known to be inducible by vitamin D,
perhaps to relieve the cells of an overload, and
a vitamin D receptor element has been found
6.48. P450 24A1 in the 5'-region of the CYP24A1 gene^^^^' ^^^^
Parathyroid hormone and cyclic AMP both
The next three P450s (24A1, 27A1, 27B1) are enhance induction by the vitamin D receptor^^^^.
involved in vitamin D metabolism (Figure 10.16). In human keratinocytes, P450 24A1 mRNA
All three are mitochrondrial and receive electrons was also elevated by la,25-dihydroxyvitamin D3
from the iron sulfur protein adrenodoxin (via the (ref [1367]). Studies with rat systems indicate that
flavoprotein adrenodoxin reductase) (Table 10.1). this response is also mediated by vitamin D
response elements and that two of these (VDRE-1,
VDRE-2) operate synergistically^^^^. A functional
6.48.1. Sites of Expression and Ras-dependent Ets-binding site is located down-
stream from the proximal VDRE site and was
Abundance
critical; the model indicates transcriptional cooper-
The 24-hydroxylation of 25-hydroxyvitamin ation between Ras-activated Ets proteins and the
D3 has long been known to occur in the kidney vitamin D receptor-RXR complex in mediating
mitochondrial membrane ^^^^ Following the la,25-dihydrox)rvitamin D action on the P450
purification of a rat P450 with this activity^^^2, 24A1 promoter'^^^. The YYl transcription factor
cDNA clones for chicken^^^^ and human^^^"^ has been reported to repress la,25-dihydroxy-
homologs were obtained. vitamin D3-induced transcription in cell cul-
The enzyme is expressed in both proximal and ^gi375 jYiQ isoflavone genistein was reported to
distal kidney tubules^^^^, but has also been found block the transcription of the CYP24A1 gene in
in human nonsmall cell lung carcinomas ^^^^. This human prostatic cancer cells and this block could
would appear to be a relatively low abundance be relieved with the histone deacetylase inhibitor
P450. Expression has also been reported in human trichostatin A*^''^. Finally, the earlier results with
keratinocytes^^^^' ^^^^, colon carcinoma cells^^^^, Ets proteins {vide supra) have been expanded
and prostatic cancer cells'^^^. to show distinct roles of the MAP kinases

,P45027A1 J ( P450 24A1


Vitamin D3 - ^ 25-OH D3 ^^ ^> 24,25-(0H)2 D3
(chloecalciferol)

P450 24A1^
1a,25-(OH)2D3 ^'^^ ^ 1 a,24,25-(OH)3 D3

P450 3A4 regulation - • Vitamin D receptor • Bone Ca^"^ mobilization,


intestinal Ca^"*" absorption

Figure 10.16. Overview of P450s involved in key steps of vitamin D activation^^.


Human P450s 455

HO^'

25(OH)

Figure 10.17. C-23 hydroxylation pathway for 25-hydroxyvitamin D3 (25(OH)) oxidation catalyzed by P450
24A1 (ref. [1378]).

ERK1/ERK2 and ERK5 (ref. [1376]). Induction 6.48.5. Inhibitors


of P450 24A1 by la,25-dihydroxyvitamin D3
involves Ets-1 phosphorylation at Thr38, but Inhibition of P450 24A1 is of some interest in
la,25-dihydrox3rvitamin D3 stimulation of ERKl/ the context of raising levels of active vitamin D
ERK2 required RXRa phosphorylation on Ser260 metabolites. Schuster^^^^' ^^^^' ^^^^ has identified
(ref. [1376]). some inhibitors that differ in their selectivity
between P450 24A1 and P450 27B1 and have
Polymorphisms in P450 24A1 have apparently
not been reported. sub-jxM IC5Q values.

6.48.6. Clinical Issues


6.48.3. Substrates and Reactions
The scheme presented in Figure 10.16 depicts
Both 25-hydroxyvitamin D3 and la,25-dihy- P450 24A1 as an enzyme involved in deactivating
droxyvitamin D3 are substrates for 24-hydroxyla- the activated form of vitamin D. The possibility
tion (Figure 10.16), with the latter being the has been considered that defects in P450 24A1
preferred substrate^^^^. However, human P450 might lead to hypervitaminosis TP^. An overactive
24A1 can also catalyze other side-chain reactions P450 24A1 could lead to vitamin D deficiency.
(Figure 10.17). Studies with side-chain-fluori- Henry^^^^ has reviewed the role of P450 24A1 and
nated vitamin D analogs also provide evidence for made comparisons to other "multistep" P450
some flexibility of this side chain in allowing enzymes. The possibility is raised that P450 24
P450 24A1 to oxidize^^^o, i38i ^^t P450 24A1 dif- could serve to generate products with their own
fers from the human ortholog in taking la,25- biological activities, with P450 24 thus being
dihydroxyvitamin D3 on to calcitroic acid instead involved in an anabolic pathway. Recently, trans-
of the products shown in Figure 10.17^^^^"^^^"^. genic rats overexpressing (rat) P450 24A1 were
found to have low plasma levels of 24,25-dihy-
droxy vitamin 03^^^^, which was unexpected.
6.48.4. Knowledge about Active Site Further, the transgenic rats developed albuminuria
Limited site-directed mutagenesis has been and hyperlipidemia shortly after weaning, and
done with P450 24A1. Expression studies using later developed atherosclerotic lesions in the
E. coli indicate species conservation in the aorta. These results raise the possibility that P450
residues 245-253 (F-helix), and the mutants 24A1 is involved in ftmctions other than vitamin
M246F and F249T showed changes in the ratio of D metabolism^^^^.
24-hydroxylation to other pathways^^^'^.
Studies by Jones' group^^^^ with analogs indi- 6.49. P450 26A1
cate that the site of hydroxylation by P450 24A1
depends on the distance from the ring to the site Retinoic acid is a vitamin A derivative that
(23 or 24), and not by the distance beyond this. plays an important role in gene regulation
Schuster's group has developed pharmacophore and development. The metabolism of retinoic acid
models^^^^. has been a matter of interest for some time, and
456 F. Peter Guengerich

the oxidation of retinoic acid by some hepatic 6.51. P450 26C1


P450s has been demonstrated^^^' ^^2, i39o
However, the relevance of these transformations to The only information presently available about
retinoic acid homeostasis in target tissues is not the CYP26C1 gene is its existence in the human
clear. White et alP^^ probed a panel of mRNAs genome^^^. Any suggestion of a role in retinoid
from mammalian cell lines with a cDNA from metabolism is only speculatory at this time.
a zebrafish P450 shown to be involved in
retinoic acid-inducible retinoic acid oxidation and
characterized P450 26A1 (ref [1391]). The
6.52. P450 27A1
heterologously expressed enzyme converted all- This is a mitochondrial enzyme that was
tmns-rQtinoic acid to the 4-hydroxy-, and 4-oxo-, characterized on the basis of two rather divergent
and 18-hydroxy products. The turnover numbers catal)^ic activities, the 25-hydroxylation of vita-
are unknown because the amount of P450 was min D3 (Figure 10.16) and the oxidation of cho-
not quantified, but the enzyme is clearly able to lesterol at the C27 position (Figure 10.18). Thus,
catalyze the oxidation of sub-iuiM additions of all- the enzyme bridges between hormone (vitamin D)
trans-YQtinoic acid^^^^. Apparently other retinoic and oxysterol pathways, and the clinical relevance
acid isomers are not substrates. of P450 27A1 is considerable.
The enzyme is expressed in cell lines derived
from several different tissues (kidney, lung, liver,
breast)^^^^. P450 26A1 has also been shown to be 6.52.1. Sites of Expression and
expressed in human fetal liver and brain tis- Abundance
sues^^^^' ^^^^, with a pattern differing from P450
26B1. The enzyme is localized in liver mitochondria.
P450 26A1 (and 26B1, vide infra) may func- Confusion existed in the early literature because
tion to protect fetal brain and possibly other tis- some animal species have liver microsomal vita-
sues from excess retinoic acid, which is known to min D3 25-hydroxylases (e.g., hog liver and kid-
be teratogenic, by metabolism. The interplay ney P450 2D25, (refs [1398], [1399])), but not
between P450s 26A1 and 26B1, and the signifi- humans''^^^. The rat and human liver mitochrondr-
cance of changes in their levels to normal tissue ial P450 27A1 recombinant enzymes were clearly
fimction is not yet known. shown to catalyze both vitamin D3 25-hydroxyla-
tion and the 27-hydroxylation of the side chains of
cholesterol and several derivatives^'^^^' ^'^^^.
Expression, at least at the mRNA level,
6.50. P450 26B1 has also been reported in leukocytes ^"^^^j skin
The CYP26B1 gene has 44% sequence identity fibroblasts''^^'^, kidney^"^^^ (and fetal liver and
with CYP26A1; it was found by searches of kidney'"^^^), and the arterial wall''^^^
genomic databases^^^^. The gene expression pat-
tern for P450 26B1 is different from that of P450
26A1 in mice^^^^ and also in human fetal
6.52.2. Regulation and Induction
brain^^^^ The localization of P450 26B1 in adult In a "normal" human population, the variation
human brain differs from that of P450 26A1, with in the steady-state P450 27A1 mRNA level was
a higher level of P450 26B1 mRNA in the cere- reported to be ~25-fold, compared with 60-fold
bellum ^^^^. Exactly how critical this enzyme is in for P450 7A1 in the same study'^"^^ However, at
brain function is unknown^^^^. The catalytic speci- least two polymorphisms ( > 1 % incidence, no dra-
ficity is very similar to that of P450 26A1, acting matic effect) and 42 mutations (rare alleles, usually
only on dd\-tmns retinoic acid and catalyzing the debilitating) are known^'^^'*' ^^^'^. Truncation muta-
formation of 4-hydroxy-, 4-oxo-, and 18-hydroxy tions are known^"^^^, as well as splice variants^'^^^.
(trans) retinoic acid^^^^. Defects in the CYP27A1 gene are associated
As with P450 26A1, frirther studies are needed with a condition known as cerebrotendinous
to define the presence of any defects in the gene xanthomatosis (CTX), a rare, autosomal recessive
and what the consequences might be. disorder characterized by accumulation of
Human P450s 457

J ] /-OH J ] CO2H
^
^ ^ ^ A ^ J ^ ^ P450 27A1^ ^ x l I V P450 27A1^
^.~/L> '
HO'^^---'^
Cholesterol
HO' -CU CXJ
27-OH Cholesterol 3^0H-5-cholestenoic acid

P450 7A1
\ \ ^

rXM rX^" Y-^CO.H


^ - x A ^ J ^

Ho-V.''^"'OH
P450 27A1^ ^ . I l

HO^^ - - ^ • • • O H
V P450 27A1^
Vr^
H O ^- - ^ • • • O H
7a-0H cholesterol 7a,27-(OH)2 cholesterol 3/3,7a-(OH)2-5-cholestenoic acid

]f
' ^ ^ r . ^-

rAH M V /OCP ^
r-vX V°^"
cxc
^ Y S ^ " ^ P^50 27A1^ ^.v^Jv^^Ly P450 27A1^

0^=^V.A.^".OH o<^^.AJ'-^OH 0^
7a-OH-4 cholestene-3-one 7a;27-(OH)2-4'Cholestene-3-one 7a-OH-3-oxo-4-cholestenolc acid

CO2H

O^ " ^ ^ "OH HO '•^^ ^-^ "OH


7c^12a-(OH)2-4-cholestene-3-one Chenodeoxycholic acid

CO2H

5j3-Cholestane-3a,7a,12a-triol 5j3-Cholestane-3o^7a^12(z,27-tetrol ZoL, la, 12c»-(OH)3 coprostanoic acid

P450 3A4
f
OH^

CO2H

5j3-Cholestane-3 0^70^12a^25-tetrol

Figure 10.18. Bile acid synthesis from cholesterol^^^^. The steps shown with dashed arrows are tentative.

cholestanol and cholesterol in many tissues. The signs, myelopathy, and peripheral neuropathy^^'
clinical manifestations include tendon xanthoma, ^^^^.
premature cataracts, juvenile atherosclerosis, and a Several aspects of regulation of the CYP27A1
progressive neurological syndrome involving men- gene have been studied. In rats, the enzyme can be
tal retardation, cerebellar atoxia, pyramidal tract induced by gonadotropin^"^^^. A hamster model
458 F. Peter Guengerich

showed downregulation of the gene in cholestatic (human) P450 2D6 abolished the activity toward
liver^^^^, although human P450 27A1 (used in vitamin 03^"^^^.
HepG2 cells) was not subject to negative feedback
regulation^'^^^
6.52.5. Inhibitors
Apparently little specific work has been done
6.52.3. Substrates and Reactions
on inhibition of this enzyme. Inhibition of this
Expanding on previous discussion, P450 27A1 enzyme by a drug would probably be undesirable.
catalyzes the 25-hydroxylation of vitamin D3
(Figure 10.16), la-hydroxyvitamin D3, vitamin
D2, and la-hydrox3rvitamin D2, and also the 27- 6.52.6. Clinical Issues
hydroxylation of cholesterol and several deriva-
Low serum 25-hydroxyvitamin D3 concentra-
tives (Figure lO.lS)^^!^' ^^^i. The cholesterol
tions have been reported in a variety of other med-
alcohols are further oxidized by the enzyme to
ical conditions and are considered to be potential
aldehydes and then carboxylic acids''^^^. The
problems^'^^^. Although CTX is linked with defec-
available information suggests release of the inter-
tive P450 27A1 (ref [29]), there are a number of
mediates in the pathway^^^^. The regioselectivity
enigmas about the etiology. A heterozygote
of the enzyme is considered to be a function of the
showed frontal lobe dementia and abnormal cho-
distance of the hydroxylation site to the end of the
lesterol metabolism^'^^^. Compound heterozygous
side chain^^^^.
mutations have also been reported to cause a vari-
More detailed analysis of the vitamin D3 reac- ation of CTX ^^^l
tion has been with E. coli recombinant P450
Bjorkhem has recently reviewed the issue of
27A1, with evidence for the following products
whether oxysterols (e.g., hydroxycholesterol) con-
(from vitamin D3): 25-hydroxy, 26-hydroxy, 27-
trol cholesterol homeostasis ^'*^^. Studies with
hydroxy, 24i^-25-dihydroxy, la,25-dihydroxy, 25,
rodents and cultured cells have not been very clear
26-dihydroxy, 25-,27-dihydroxy, 27-oxo, and an
to date. For instance, disruption of the mouse
unidentified dehydrogenated product^'^^^' ^^^^.
CYP27A1 gene yielded reduced bile acid synthe-
sis but apparently caused no change in levels of
cholesterol or la,25-dihydroxyvitamin 03^"*^^
6.52.4. Knowledge about Active Site
P450 27A1 is constitutively expressed in the nor-
Some information about the roles of amino mal artery wall and is substantially upregulated in
acids can be inferred from the knowledge of alle- atherosclerosis, and the possibility has been raised
les involved in CTX; many of these proteins were that P450 27A1 may be a protective mechanism
unstable when attempts were made at heterologous for removing cholesterol ^'*^^. Further, immune
expression^'*^^. Other work by Pikuleva et al}^^^ complexes and IFN-7 decreased P450 27A1
with the putative F and G helices has shown dif- expression in human aortic endothelial cells,
ferences due to substitution of Phe207, Ile211, peripheral blood mononuclear cells, monocytes-
Phe215, Trp235, and Tyr238. Interestingly, the derived macrophages, and a human monoc3^oid
121 IK and F215K mutations affected the regiose- cell line, suggesting downregulation of P450
lectivity and enabled the enzyme to catalyze C-C 27A1 to maintain cholesterol homeostasis in the
bond cleavage. Further work with mutants in this arterial wall^"^^^.
region led to weaker association of P450 27A1 In Cyp27Al~'~ mice, a dramatic increase in the
with the membrane, and some of the nonconserva- level of P450 3A enzymes is seen; some sterols
tive changes yielded impaired catalytic activity ^^^^. accumulate and induce via the mouse PXR sys-
Human P450 27A1 can be contrasted with ^gj^i423 P45Q 3^4 j^^g gQj^g side-chain hydroxyla-
porcine P450 2D25, which also catalyzes vitamin tion activities toward cholesterol-derived
D3 25-hydroxylation. The only human P450 2D sub- sterols^^^ However, elevated P450 3A4 activities
family enzyme which does not have activity toward were not increased in CTX^^^ indicating a differ-
vitamin D is P450 2D6. Further, changing a set of ence in the murine and human systems. Recently,
residues of porcine 2D25 to their counterparts in Escher et al}^'^^ have reported that cholesterol
Human P450s 459

efflux in CHOP cells is enhanced by (heterolo- vitamin D-related activities, including skin (basal
gous) expression of human P450 27A1, and the keratinocytes, hair follicles), lymph nodes (granu-
authors suggest this as part of a protective system lomata), colon (epithelial cells and parasympa-
against atherosclerosis. The basis is probably the thetic ganglia), pancreas (islets), adrenal medulla,
ability of 27-hydroxycholesterol to act as an brain (cerebellum and cerebral cortex) ^^^'^, pla-
endogenous ligand for the liver X receptor in cho- centa (decidual and trophoblastic cells)^^^'^~^^^^,
lesterol-loaded cells ^'^^^. cervix^"^^^, and parathyroid glands^"^^^. Thus, P450
In considering the general question of whether 27B1 may be an intracrine modulator of vitamin D
oxysterols (e.g., 27-hydroxycholesterol) control function in peripheral tissues^'^^'*. The expression
cholesterol homeostasis, the hypothesis is still of P450 27B1 was elevated in parathyroid adeno-
open and the rodent data are not totally clear here. mas, but attenuated in carcinomas, relative to nor-
Bjorkhem^^'*^ has made the point that humans mal parathyroid tissue ^^^^.
lacking P450 27A1 have normal circulating levels
of cholesterol.
6.53.2. Regulation and Polymorphism
Although the CYP27B1 gene is only 5 kb in
6.53- P450 27B1 size, the regulation is quite complex. The pro-
moter is in the -85/+22 region and requires
As discussed earlier, vitamin D is an important a functional CCATT element. Three consensus
hormone. A critical step in activation is la- AP-1 sites are upstream^"^^^. Enzyme activity has
hydroxylation^"^^^ (Figure 10.16). Early work long been known to be stimulated by low phos-
established the P450 nature of the enzyme, local- phorus diets (in animal models)^"^"^^j and more
ized in kidney mitochondria^'*^^. Subsequent work recently, this phenomenon has been linked to a
demonstrated that the la- and 24-hydroxylation growth hormone mechanism^^^^' ^'*'^^; its relevance
activities could be attributed to different in humans is not known.
enzymes^'*^^' ^^'^^. Some early work had suggested
Expression is also regulated by calcium,
that the la- and 25-hydroxylation activities were
parathyroid hormone, and by the product la,25-
associated with the same enzyme^^^^, but later
dihydroxyvitamin 03^^"*^' ^^^. Regulatory regions
work showed that these activities were due to
involving the responses to parathyroid hormone,
P450 27B1 and 27A1, respectively
calcitonin, and la,25-hydroxyvitamin D3 are
located in the region —0.4 to —0.5 kb^"^"^^.
Forskolin also regulates gene expression, and an
6.53.1. Sites of Expression and
Spl site is involved in aspects of regulation^"^"^^' ^^'^'^.
Abundance
Complexity is seen in different models.
The cloning of the human cDNA for what is Parathyroid hormone-related protein and Ca^^
now known as P450 27B1 established the kidney have conflicting actions in a nude rat model of
mitochondrial P450 (27B1) as the vitamin D3 la- humoral hypercalcemia of malignancy^"^"^^. In dif-
hydroxylase^'*^^ The gene has nine exons and ferentiated Caco cells, there is upregulation of
spans only 5 kb^"^^^. P450 27B1 expression by la,25-dihydrox5rvita-
P450 27B1 is expressed in many parts of the min D3 and epidermal growth factor, but down-
human kidney, including the distal convoluted regulation in less differentiated Caco cell lines^^^^.
tubule, the cortical and medullary part of the col- Another aspect of regulation of P450 27B1 is
lecting ducts, and the papillary epithelia^"^^^. genetic. P450 27B1 results in Type I vitamin D-
Lower expression was observed along the thick dependent rickets^'^'*^. The genetics have been
ascending limb of the loop of Henle and established in more than 30 patients involving at
Bowman's capsule. Some weaker expression was least 20 mutations^'^^^' ^^^^. At least 13 missense
observed in glomeruli or vascular structures. In mutations have been observed, none of which
normal humans, the distal nephron is the predom- encode an active protein. Some of the mutants are
inant site of expression^^^^. splicing defects^"^^^. Some mutations in CYP27B1
P450 27B1 is also expressed in many are also involved in what is termed pseudovitamin
extrarenal sites (human) where it is involved in D-deficiency rickets^"^^^' ^^^^. Beyond these
460 F. Peter Guengerich

debilitating mutations, little information is avail- responsible for vitamin D-dependent rickets Type
able about actual polymorphisms. I^"^^^. At least 30 different mutations are known in
patients^'^'^^' ^'^^^. Even the "mild" phenotype of
Type I rickets is due to deficiency in P450 27B1
6.53.3. Substrates and Reactions (ref [1461]).
P450 27B1 can catalyze the la-hydroxylation CYP27B1 knockout mice have been character-
of both 25-hydroxy and 24(i?),25-dihydroxyvita- ized and show a typical rickets phenotype ^'^^^.
min D3^^^^' ^^^^ (Figure 10.16). The intrinsic activ- Another mouse model in which the gene has
ity (^ca/^m) ^^^ ^^^ recombinant human enzyme is been ablated showed skeletal, reproductive, and
better for 24(i?),25-hydroxy vitamin D3, but this immune dysfunction^^^^. Rickets was also observed
does not mean that this is the favored substrate in in a conditional knockout model^"^^^.
the cell, because of the balance of vitamin D Patients with severe renal insufficiency show
metabolites regulated by P450s 24A1 and 27A1 attenuated la-hydroxylation activity^^^^.
(ref [29]). Apparently the 25-hydroxy group is an Another aspect of P450 27B1 research
obligatory requirement^^^^' ^^^^. involves cancer. Increased activity was reported in
parathyroid tumors ^'^^^. Some splice variants of
the CYP27B1 gene (coding for truncated proteins)
6.53.4. Knowledge about Active Site were amplified in human (brain) gliomas ^'^^^.
Reports have also appeared on the relationship of
Some information is available from the natural
P450 27B1 expression to various biological
mutants of P450 27B1, even if the basis for loss of
processes in human nonsmall cell lung carcino-
activity is not obvious. Inouye's group^'^^'^ has pro-
mas'^^^, colon tumors''^^^"^'^^^, and prostate can-
vided evidence that ArglOT, Glyl25, and Pro497
^gj.gi47i, 1472^ generally with decreased expression
are not simply involved in binding substrate but
in tumors.
required for proper folding. It was also suggested
Finally, la,25-dihydroxyvitamin D3 is used to
that Arg389 and Arg453 are involved in heme
treat psoriasis, and patients can develop resist-
binding and that Asp 164 stabilizes the bundle of
ance. An experimental model for therapy involves
the four helices D, E, I, and J. Thr321 is suggested
enhancement of the endogenous production of
to be involved in O2 activation^"^^"^. The natural
la,25-dihydroxyvitamin D3 by gene therapy^'^^^.
mutants L343F and E189G show partial activity
and the individuals bearing these have only mar-
ginal impairment ^'*^^.
6.54. P450 27C1
As with some of the other P450s, the only
6.53.5. Inhibitors knowledge currently available is the existence of
Little has been done because impairment of the CYP27C1 gene in the human genome^^^.
this enzyme is a clinical problem. Some thiavita- Suggestions that this enzyme is involved in vita-
min D analogs have been evaluated in animal min D metabolism are still only speculatory.
models^"^^^.

6.55. P450 39A1


6.53.6. Clinical Issues
An expression-cloning approach was utilized
The significance of the enzyme is due to the to isolate a cDNA from {Cyp7bl~'~) mice that
pleiotropic actions of the active form of vitamin could, when expressed, catalyze the 7a-hydroxyla-
D, la,25-dihydroxyvitamin D3, which include tion of 24-hydroxycholesterol^'^^'^. P450 39 has a
regulation of calcium homeostasis, control of microsomal location with a preference for the sub-
bone cell differentiation, and modification of strate 24-hydroxycholesterol and is expressed in
immune responses^^^^. The la-hydroxylation liver. Presumably these characteristics of mouse
reaction is rate limiting and hormonally con- P450 39 apply to the human ortholog but no fur-
trolled. The expression of the gene is usually ther information is yet available. Potential rele-
tightly regulated (vide supra), but gene defects are vance of this enzyme is in the inactivation of
Human P450s 461

24-hydroxycholesterol, a ligand for the LXR for many years and then demonstrated the reaction
nuclear hormone receptor (see Section 6.56 on in rat liver microsomes in 1994^^^^ Subsequently
P450 46, vide infra), the reaction was also demonstrated in rat brain
microsomes ^^^^.

6.56- P450 46A1


6.57.1. Sites of Expression and
An expression cloning approach was utilized Abundance
to clone cDNAs encoding both murine and human
cholesterol 24-hydroxylase, P450 46A1 (ref. Waterman's group identified the human
[1475]). The mouse and human sequences are CYP51A1 gene and two pseudogenes^"^^^. mRNA
95% identical. Expression is predominantly in blot analysis showed the highest levels in testis,
brain (neurons in several regions). The enzyme is ovary, adrenal, prostate, liver, kidney, and lung. In
in the endoplasmic reticulum; alternate substrates mouse testis, P450 51A1 was localized in both
have not been explored but may be unlikely. round and elongated spermatids ^'^^^. The enzyme
is also found in (rodent) oocytes^"^^^.
The significance of P450 46A1 rests in the
fact that the brain is the most cholesterol-rich
tissue in the body^"^^^. LXRs, members of the 6.57.2. Regulation and Polymorphism
nuclear hormone receptor family, are activated
by 24-hydroxycholesterol. LXRp and P450 46 Polymorphisms in the human CYP51A1 gene
have overlapping expression patterns in brain^'*'^^. have not been reported nor have debilitating
The system may be counter-balanced by P450 39, mutants been defined. However, Kelley et al}^^^
which catalyzes the 7a-hydroxylation of reported a patient with Antley-Bixler syndrome
25-hydroxycholesterol (Section 6.55). and ambiguous genitalia with lanosterol accumu-
lation and an apparent defect in P450 51A1.
Recently, there has been considerable interest
With regard to regulation of the human gene,
in the relationship between P450 46A1 and
primer extension studies indicated predominant
Alzheimer's Disease. P450 46A1 had a marked
transcription initiation sites in liver, lung, and kid-
difference in distribution in the brains of normal
ney, and placenta 250 and 249 bp upstream from
and Alzheimer's patients, with less staining of
the translation start site and a second major site at
neuronal cells but more of glial cells in diseased
-lOObp, with the absence of TATA and CAAT
patients^'^^^. Also, elevated levels of 25(5)-hydroxy-
patterns and a GC-rich sequence in the promoter
cholesterol were found in cerebral spinal fluid in
region^"^^^. Multiple (rat) testis-specific transcripts
the early stages of dementia^^^^. Associations^"^^^
arise from differential polyadenylation site
and lacks of associations ^"^^^ between CYP46
usage ^'*^^.
SNPs and Alzheimer's Disease have been
reported. One of the issues in the research with In human adrenocortical H295R cells (in cul-
P450 46A1 is the apparently very low rate of ture), cholesterol deprivation led to a 2.6-3.8-fold
cholesterol 24-hydroxylation (although an exact induction of P450 51A1 mRNA, which was sup-
rate can be deduced from the published informa- pressed by the addition of 25-hydroxycholes-
tion). Recently, Pikuleva's laboratory has found terol^'^^^. In the liver and other somatic tissues,
that 24-hydroxycholesterol is a much better sub- CYP51 is regulated by a sterol/sterol-regulatory
strate for recombinant P450 46A1 than is choles- element binding protein (SREBP)-dependent
terol, being oxidized to as yet uncharacterized pathway^"^^^. In testis, cAMP/cAMP-responsive
producti480a element modulator (CREM)^-dependent regula-
tion predominates. Spl fiinctions to maximize the
sterol regulatory pathway of P450 51 (ref. [1490]).
6.57. P450 51A1 Insulin is an essential factor in "basal" expres-
sion of P450 51 in rat liver, with possible involve-
Lanosterol is an important intermediate in ment of SREBP-lc involvement^'^^^. In a porcine
cholesterol synthesis, and 14a-demethylation vascular endothelial cell model (and in arterial
has been established as a step in the pathway. wall), LDLs downregulated P450 51 through an
Yoshida's laboratory had studied the yeast enzyme SREBP-2 mechanismi492.
462 F. Peter Guengerich

6.57.3. Substrates and Reactions 6.57.6. Clinical Issues


Stimulation of human P450 51 activity by b^ in Most of the work discussed here is from exper-
a reconstituted system has been reported by imental studies on the possible role of P450 51 in
Kelly's laboratory^'*^^. reproduction, and the translation of phenomena
The normal mammalian substrate for P450 51 fi-om animal models to humans is still somewhat
is 24,25-dihydrolanosteroli^^^, with the 14a- speculatory. However, the very high level of P450
demethylation process proceeding in what are 51 expression in postmeiotic haploid spermatids is
assumed to be three consecutive steps, as with striking. The action of P450 51 is proposed to lead
some other P450s, for example, l l A l , 17A1, to the production of signaling steroids in haploid
19A1. Interestingly, both human and yeast germ cells ^^^^. Meiosis-activating substances
{Candida albicans) P450 51 showed relatively lit- (MAS) are produced by 14-reduction of products of
tle selectivity among a closely related group of the action of P450 51 on lanosterol^^^^. Follicular
analogs^^^'*. It is also interesting to note that even fluid MAS (FF-MAS) is formed from lanosterol in
though this P450 has a relatively defined role in rat spermatids *^^^ Yoshida's group has reported
a physiological process, the kinetic parameters gonadotropin-dependent expression of P450 51 in
are relatively poor among P450s {k^JK^ = 300 rat ovaries and the production of MAS ^^^^.
The reaction and possible physiological signif-
icance of the system in reproduction have been
reviewed recently by Rozman^^^^. Leydig cells
6.57.4. Knowledge about Active Site and acrosomes of spermatids have the highest
A crystal structure of human P450 51 is not P450 51 levels, and primary mouse oocytes and
yet available but high resolution structure of the granulose cells also contain P450 51. The MAS
soluble Mycobacterium tuberculosis P450 51 is may have a role in fertilization •^^^.
(ref [1495]). Two notable features are a bent I
helix and an open conformation of the BC loop.
The bacterial structure has been utilized in con- 7. Concluding Remarks
sideration of mammalian models, and the SRS
predictions do not seem to apply well^"^^^. Further, Some information has been presented about the
the mutation hotspots for known azole-resistant known P450s. Because the genome is nearly com-
C. albicans P450 51 mutants tend to be outside the pleted, no more human P450s are likely to be added
predicted active site and suggest the contribution unless our views of marker sequences change.
of long-range effects on ligand binding''^^^' ^'^^^. Although much of the interest in human P450 is
Studies on Arg448 indicate that, despite directed toward drug metabolism, the majority of
sequence conservation, aspects of the role of this P450s appear to be involved in the metabolism of
and other residues are different for human and endobiotics (Table 10.2). In one sense, the fact that
yeast P450 51 (ref [1498]). about 5 or 6 of the P450s are so dominant in xeno-
biotic metabolism (Figure 10.3) is not surprising
when we recognize that only about 15 of the 57 use
6.57.5. Inhibitors
xenobiotics as substrates (Table 10.1) and consider
Most of the interest in inhibition has been with the dominant levels of expression of a few P450s
fungal P450 51, as a target for antimycotic drugs. that have rather broad selectivity (Figure 10.4).
The goal is to select candidate drugs inhibitory to Aside from the current problems already men-
fungal P450 51, but not human P450 51. tioned, including practical issues, what are some of
Some work on the interaction of azoles with the future challenges and in what areas should
human P450 51 has been published^'^^^. Although work be done? Some of the problems are very
human P450 51 has been suggested as a target for basic, such as the goals of deriving more experi-
cholesterol-lowering drugs, apparently little has mental three-dimensional structures, answering
been done and potential toxicity due to the questions about catalytic mechanisms, and under-
steroidogenic and potential germ cell side effects standing the complexities of regulation of these
{vide infra) could be an issue. genes.
Human P450s 463

NCH3

H3C0. H3CO.

NCH3 NCH3

Neopinone

Figure 10.19. Steps in mammalian morphine synthesis^'

There is considerable opportunity to improve there is an opportunity for the introduction of


screening and predictability for use with new novel approaches.
chemical entities in the pharmaceutical industry. Note added in proof. Recently Pai et alP^^
Another open issue is whether large-scale SNP have demonstrated an unusual phenomenon with
analysis can be practical in drug development. the pseudogene, CYP2D7. A frame shifted allelic
Another area is understanding the relevance of variant yields expression of an enzyme specifi-
variations in P450s (both SNPs and expression cally in the brain, not liver, and this enzyme is
levels) to diseases, not only drug therapies. For more efficient than P450 2D6 in converting
instance, one can ask if some of the "minor" sterol codeine to morphine (Figure 10.19).
products produced by P450 3A4 have any impact
on cardiovascular disease. How important are
some of the P450s that oxidize arachidonic acid to Acknowledgments
various products (in humans)? Does a P450 2D6
deficit have any relevance to long-term health? Research in the author's laboratory has been
Finally, there are two more major problems supported in part by USPHS grants ROl CA90426
in understanding what human P450s do. One and P30 ES00267. Thanks are extended to the
problem is the oxidation reactions, largely involv- many individuals who provided unpublished
ing endobiotics, for which no P450s have been results, to Drs. E.M.J. Gillam, F.F. Kadlubar, and
defined. One example is the synthesis of endoge- M.R. Waterman for comments, and to present and
nous morphine in the body, which appears to past members of the author's laboratory for their
require multiple oxidations and is subject to short- involvement in some of the original research. This
term regulation^^^"^' ^^^^ (Figure 10.19). Although chapter is dedicated to Dr. Tsutomu Shimada, my
P450 2D6 can oxidize codeine to morphine^^^^, long-term collaborator, on the occasion of his
this enzyme does not contribute to the endogenous retirement from the Osaka Prefectural Institute of
formation of morphine ^^^^. What other oxidative Public Health (March 2003), and to my postdoc-
reactions remain to be discovered and character- toral mentor. Prof M.J. Coon, who introduced me
ized? In the past year, the oxidative dealkylation to these enzymes 30 years ago.
in DNA repair was demonstrated to involve an
ct-ketoglutarate-dependent dioxygenase^^^^' ^^^^.
Perhaps similar roles for P450s may be found. References
The other issue is identifying functions for the
remaining human P450s, of which there are at 1. Distlerath, L.M. and F.F. Guengerich (1987).
least 15 (Table 10.2). Doing this on a step-by-step Enzymology of human liver cytochromes P-450.
basis as in the past will not be very efficient, and In F.P. Guengerich (ed.) Mammalian Cytochromes
464 F. Peter Guengerich

P-450, Vol. 1. CRC Press, Boca Raton, FL, Characterization of rat and human liver microsomal
pp. 133-198. cytochrome P-450 forms involved in nifedipine oxi-
2. Beaune, R, R Dansette, J.R Flinois, S. Columelli, dation, a prototype for genetic polymorphism in
D. Mansuy, and J.R Leroux (1979). Partial purifica- oxidative drug metabolism. J. Biol. Chem. 261,
tion of human liver cytochrome P-450. Biochem. 5051-5060.
Biophys. Res. Commun. 88, 826-832. 13. Watkins, RB., S.A. Wrighton, R Maurel,
3. Wang, R, PS. Mason, and F.R Guengerich (1980). E.G. Schuetz, G. Mendez-Picon, G.A. Parker et al.
Purification of human liver cytochrome P-450 and (1985). Identification of an inducible form of
comparison to the enzyme isolated from rat liver. cytochrome P-450 in human liver. Proc. Natl. Acad.
Arch. Biochem. Biophys. 199, 206-219. Sci. 0X4 82,6310-6314.
4. Wang, PR, P. Beaune, L.S. Kaminsky, G.A. Dannan, 14. Wrighton, S.A., C. Campanile, RE. Thomas,
FR Kadlubar, D. Larrey et al. (1983). Purification S.L. Maines, RB. Watkins, G. Parker et al. (1986).
and characterization of six cytochrome P-450 Identification of a human liver cytochrome P-450
isozymes from human liver microsomes. homologous to the major isosafrole-inducible
Biochemistry 11, 5375-5383. cytochrome P-450 in the rat. Mol. Pharmacol. 29,
5. Mahgoub, A., J.R. Idle, L.G. Dring, R. Lancaster, 405^10.
and R.L. Smith (1977). Polymorphic hydroxylation 15. Wrighton, S.A., RE. Thomas, R Willis, S.L. Maines,
of debrisoquine in man. Lancet ii, 584-586. PB. Watkins, W Levin et al. (1987). Purification of
6. Shimada, T., C.-H. Yun, H. Yamazaki, J.-C. Gautier, a human liver cytochrome P-450 immunochemi-
RH. Beaune, and FR Guengerich (1992). cally related to several cytochromes P-450 purified
Characterization of human lung microsomal from untreated rats. J. Clin. Invest. 80, 1017-1022.
cytochrome P-450 lAl and its role in the oxidation 16. Gonzalez, F.J. (1989). The molecular biology of
of chemical carcinogens. Mol. Pharmacol. 41, cytochrome P450s. Pharmacol. Rev. 40, 243-288.
856-864. 17. Larson, J.R., M.J. Coon, and T.D. Porter (1991).
7. Distlerath, L.M., PE.B. Reilly, M.V. Martin, Alcohol-inducible cytochrome P-450IIE1 lacking
G.G. Davis, G.R. Wilkinson, and FP Guengerich the hydrophobic NH2-terminal segment retains cat-
(1985). Purification and characterization of the alytic activity and is membrane-bound when
human liver cytochromes P-450 involved in debriso- expressed in Escherichia coli. J. Biol. Chem. 266,
quine 4-hydroxylation and phenacetin O-deethylation, 7321-7324.
two prototypes for genetic polymorphism in oxidative 18. Li, Y.C. and J.Y.L. Chiang (1991). The expression of
drug metabolism. J. Biol. Chem. 260, 9057-9067. a catalytically active cholesterol 7a-hydroxylase
8. Yun, C.-H., T. Shimada, and FR Guengerich (1991). cytochrome P-450 in Escherichia coli. J. Biol.
Purification and characterization of human liver Chem. 266, 19186-19191.
microsomal cytochrome P-450 2A6. Mol. 19. Barnes, H.J., M.P Arlotto, and M.R. Waterman
Pharmacol. 40, 679-685. (1991). Expression and enzymatic activity of
9. Shimada, T., K.S. Misono, and FP. Guengerich recombinant cytochrome P450 17a-hydroxylase in
(1986). Human liver microsomal cytochrome P-450 Escherichia coli. Proc. Natl. Acad. Sci. USA 88,
mephenytoin 4-hydroxylase, a prototype of genetic 5597-5601.
polymorphism in oxidative drug metabolism. 20. Guengerich, FP, E.M.J. Gillam, and T. Shimada
Purification and characterization of two similar (1996). New applications of bacterial systems to
forms involved in the reaction. J. Biol. Chem. 261, problems in toxicology. Crit. Rev. Toxicol. 26,
909-921. 551-583.
10. Gut, J., T. Catin, R Dayer, T. Kronbach, U. Zanger, 21. Nebert, D.W, M. Adesnik, M.J. Coon,
and U.A. Meyer (1986). Debrisoquine/sparteine- R.W Estabrook, F.J. Gonzalez, FP. Guengerich
type polymorphism of drug oxidation: Purification et al. (1987). The P450 gene superfamily:
and characterization of two functionally different Recommended nomenclature. DNA 6, 1-11.
human liver cytochrome P-450 isozymes involved 22. Nelson, DR., T Kamataki, D.J. Waxman,
in impaired hydroxylation of the prototype substrate FP. Guengerich, R.W. Estabrook, R. Feyereisen
h\xfyxx2\o\. J. Biol. Chem. 261, 11734-11743. et al. (1993). The P450 superfamily: Update on new
11. Birgersson, C , E.T. Morgan, H. Jornvall, and C. von sequences, gene mapping, accession numbers, early
Bahr (1986). Purification of a desmethylimipramine trivial names of enzymes, and nomenclature. DNA
and debrisoquine hydroxylating cytochrome P-450 Cell Biol. 12, 1-51.
from human liver. Biochem. Pharmacol. 35, 23. Guengerich, FR (1995). Human cytochrome P450
3165-3166. enzymes. In PR. Ortiz de Montellano (ed.).
12. Guengerich, RR, M.V Martin, RH. Beaune, Cytochrome P450. Plenum Press, New York,
R Kremers, T. Wolff, and D.J. Waxman (1986). pp. 473-535.
Human P450s 465

24. Niranjan, B.G., N.M. Wilson, C.R. Jefcoate, and Factors in the Clinical Evaluation of Medicines.
N.G. Avadhani (1984). Hepatic mitochondrial Kluwer Academic Publishers, Dordrecht/Boston/
cytochrome P-450 system: Distinctive features of London, pp. 13-26.
cytochrome P-450 involved in the activation of afla- 37. Guengerich, RR and D.C. Liebler (1985). Enzy-
toxin Bj and benzo(a)pyrene. J. Biol. Chem. 259, matic activation of chemicals to toxic metabolites.
12495-12501. Crit. Rev Toxicol. 14, 259-307.
25. Addya, S., H.K. Anandatheerthavarada, G. Biswas, 38. Motulsky, A.G. (1957). Drug reactions, enzymes
S.V Bhagv^at, J. Mullick, and N.G. Avadhani and biochemical genetics. J. Am. Med. Assoc. 165,
(1997). Targeting of NH2-terminal-processed 835-837.
microsomal protein to mitochondria: A novel path- 39. Kalow, W (1962). Pharmacogenetics. W B. Saunders,
way for the biogensis of hepatic mitochondrial Philadelphia, PA.
P450j^T-2- -^ Cell Biol 139, 589-599. 40. Remmer, H. (1957). The acceleration of evipan oxi-
26. Robin, M.A., H.K. Anandatheerthavarada, dation and the demethylation of methylaminopyrine
G. Biswas, N.B. Sepuri, D.M. Gordon, D. Pain et al. by barbiturates. Naunyn-Schmiedebergs Arch. Exp.
(2002). Bimodal targeting of microsomal CYP2E1 Pathol. Pharmakol. 231, 296-307.
to mitochondria through activation of an N-terminal 41. Keeney, D.S. and M.R. Waterman (1993). Regu-
chimeric signal by cAMP-mediated phosphoryla- lation of steroid hydroxylase gene expression:
tion. J. Biol. Chem. 277, 40583-40593. Importance to physiology and disease. Pharmacol.
27. Tateishi, T., Y. Krivoruk, Y.-F. Ueng, A.J.J. Wood, Then 58,301-317.
F.R Guengerich, and M. Wood (1996). Identifi- 42. Conney, A.H., E.G. Miller, and J.A. Miller (1956).
cation of human liver cytochrome P450 3A4 as The metabolism of methylated aminoazo dyes.
the enzyme responsible for fentanyl and sulfentanyl V Evidence for induction of enzyme synthesis in
^-&Q^\ky\dX\on. Anesth. Analg. 82, 167-172. the rat by 3-methylcholanthrene. Cancer Res. 16,
28. Shimada, T., H. Yamazaki, 4. Mimura, Y. Inui, and 450-459.
P.P. Guengerich (1994). Interindividual variations in 43. Eichelbaum, M., N. Spannbrucker, B. Steincke, and
human liver cytochrome P450 enzymes involved in H.J. Dengler (1979). Defective N-oxidation of
the oxidation of drugs, carcinogens, and toxic chem- sparteine in man: A new pharmacogenetic defect.
icals: Studies with liver microsomes of 30 Japanese Eur. J. Clin. Pharmacol. 16, 183-187.
and 30 Caucasians. J. Pharmacol. Exp. Then 270, 44. Woolhouse, N.M., B. Andoh, A. Mahgoub, T.P Sloan,
414^23. J.R. Idle, and R.L. Smith (1979). Debrisoquin
29. Nebert, D.W. and D.W. Russell (2002). Clinical hydroxylation polymorphism among Ghanaians and
importance of the cytochromes P450. Lancet 360, Caucasians. Clin. Pharmacol. Ther 26, 584-591.
1155-1162. 45. Johansson, I., E. Lundqvist, L. Bertilsson,
30. Brodie, A.M.H. (1985). Aromatase inhibition and its M.L. Dahl, R Sjoqvist, and M. Ingelman Sundberg
pharmacologic implications. Biochem. Pharmacol. (1993). Inherited amplification of an active gene in
34,3213-3219. the cytochrome P450 CYP2D locus as a cause of
31. Gonzalez, RJ. and S. Kimura (2003). Study of P450 ultrarapid metabolism of debrisoquine. Proc. Natl.
function using gene knockout and transgenic mice. Acad Sci. USA 90, 11825-11829.
Arch. Biochem. Biophys. 409, 153-158. 46. Smith, R.L., J.R. Idle, A.A. Mahgoub, T.R Sloan,
32. Rendic, S. (2002). Summary of information on and R. Lancaster (1978). Genetically determined
human CYP enzymes: Human P450 metabolism defects of oxidation at carbon centres of drugs.
data. DrugMetab. Rev. 34, 83^48. Lancet i, 943-944.
33. Evans, WE. and M.V Relling (1999). 47. Gonzalez, F.J., R.C. Skoda, S. Kimura, M. Umeno,
Pharmacogenomics: Translating function genomics U.M. Zanger, D.W. Nebert et al. (1988). Characteri-
into rational therapeutics. Science 286, 487^91. zation of the common genetic defect in humans
34. Guengerich, RR andT. Shimada (1991). Oxidation deficient in debrisoquine metabolism. Nature 331,
of toxic and carcinogenic chemicals by human 442^46.
cytochrome P-450 enzymes. Chem. Res. Toxicol. 4, 48. Nakamura, K., R Goto, W.A. Ray, C.B. McAllister,
391^07. E. Jacqz, G.R. Wilkinson et al. (1985). Interethnic
35. Breimer, D.D. and J.H. Schellens (1990). A "cocktail" differences in genetic polymorphism of debrisoquin
strategy to assess in vivo oxidative drug metabolism and mephenytoin hydroxylation between Japanese
in humans. Trends Pharmacol. Sci. 11, 223-225. and Caucasian populations. Clin. Pharmacol. Then
36. Breimer, D.D. (1994). Genetic polymorphisms in 38, 402-408.
drug metabolism; clinical implications and conse- 49. Daly, A.K. (2003). Pharmacogenetics of the major
quences in ADME studies. In S. Walker, C. Lumley, polymorphic metabolizing enzymes. Fundam. Clin.
and N. McAuslane (eds), The Relevance of Ethnic Pharmacol. \1,21-A\.
466 F. Peter Guengerich

50. Nagata, K. and Y. Yamazoe (2002). Genetic isoform identification. Drug Metab. Dispos. 31,
polymorphism of human cytochrome P450 involved 345-350.
in drug metabolism. Drug Metab. Pharmacokinet. 64. Correia, M.A. and PR. Ortiz de Montellano (2004,
17, 167-189. in press). Inhibition and degradation of cytochrome
51. Daly, A.K., J. Brockmoller, F. Broly, M. Eichelbaum, P450 enzymes. In P. R. Ortiz de Montellano (ed.).
W.E. Evans, F.J. Gonzalez et al. (1996). Nomen- Cytochrome P450: Structure, Mechanism, and
clature for human CYP2D6 alleles. Pharmacogenetics Biochemistry. Plenum Press, New York (Chap. 7 of
6, 193-201. this monograph).
52. Gaedigk, A., M. Blum, R. Gaedigk, M. Eichelbaum, 65. Correia, M.A. (2004, in press). Isoform functional
and U.A. Meyer (1991). Deletion of the entire markers, isoform substrate specificities, and
cytochrome P450 CYP2D6 gene as a cause of fluorescent substrate assays. In PR. Ortiz de
impaired drug metabolism in poor metabolizers of Montellano (ed.). Cytochrome P450: Structure,
the debrisoquine/sparteine polymorphism. Am. J. Mechanism, and Biochemistry. Plenum Press,
Hum. Genet. 48, 943-950. New York (Appendix of this monograph).
53. Corchero, J., C.P. Granvil, T.E. Akiyama, 66. Harris, J.W., A. Rahman, B.-R. Kim,
G.P. Hayhurst, S. Pimprale, L. Feigenbaum et al. FP Guengerich, and J.M. Collins (1994).
(2001). The CYP2D6 humanized mouse: Effect of Metabolism of taxol by human hepatic microsomes
the human CYP2D6 transgene and HNF4a on the and liver slices: Participation of cytochrome P450
disposition of debrisoquine in the mouse. Mol. 3A4 and of an unknown P450 enzyme. Cancer Res.
Pharmacol. 60, 1260-1267. 54, 4026-4035.
54. Schuetz, E.G., D.R. Umbenhauer, K. Yasuda, 67. Tran, T.H., L.L. von Moltke, K. Venkatakrishnan,
C. Brimer, L. Nguyen, M.V Relling et al. (2000). B.W. Granda, M.A. Gibbs, R.S. Obach et al. (2002).
Altered expression of hepatic cytochromes P-450 in Microsomal protein concentration modifies the
mice deficient in one or more mdr^ genes. Mol. apparent inhibitory potency of CYP3A inhibitors.
Pharmacol. 57, 188-197. Drug Metab. Dispos. 30, 1441-1445.
55. Cummins, C.L., C.Y Wu, and L.Z. Benet (2002). 68. Austin, R.P, P Barton, S.L. Cockroft,
Sex-related differences in the clearance of M.C Wenlock, and R.J. Riley (2002). The influence
cytochrome P450 3A4 substrates may be caused by of nonspecific microsomal binding on apparent
P-glycoprotein. Clin. Pharmacol. Ther 72, 474-489. intrinsic clearance, and its prediction from physico-
56. Renton, K.W. and L.C. Knickle (1990). Regulation chemical properties. Drug Metab. Dispos. 30,
of hepatic cytochrome P-450 during infectious dis- 1497-1503.
ease. Can. J. Physiol. Pharmacol. 68, 177-lSl. 69. Yoo, J.S.H., R.J. Cheung, CJ. Patten, D. Wade,
57. Kato, R. and Y. Yamazoe (1992). Sex-specific and CS. Yang (1987). Nature of 7V-nitrosodi-
cytochrome P450 as a cause of sex- and species- methylamine demethylase and its inhibitors. Cancer
related differences in drug toxicity. Toxicol. Lett. Res. 47, 3378-3383.
64/65, 661-667. 70. Chauret, N., A. Gauthier, and D.A. Nicoll-Grififith
58. Komori, M., K. Nishio, M. Kitada, K. Shiramatsu, (1998). Effect of common organic solvents on in
K. Muroya, M. Soma et al. (1990). Fetus-specific vitro cytochrome P450-mediated metabolic activi-
expression of a form of cytochrome P-450 in human ties in human liver microsomes. Drug Metab.
livers. Biochemistry 29, 4430^433. Dispos. 26, 1-4.
59. Lacroix, D., M. Sonnier, A. Moncion, G. Cheron, 71. Yuan, R., S. Madani, X.X. Wei, K. Reynolds, and
and T. Cresteil (1997). Expression of CYP3A in the S.M. Huang (2002). Evaluation of cytochrome P450
human liver: Evidence that the shift between probe substrates commonly used by the pharmaceu-
CYP3A7 and CYP3A4 occurs immediately after tical industry to study in vitro drug interactions.
birth. Eur J. Biochem. 247, 625-634. Drug Metab. Dispos. 30, 1311-1319.
60. Loi, CM. and R.E. Vestal (1988). Drug metaboHsm 72. Guengerich, FP (1988). Oxidation of 17a-
in the elderly. Pharmacol. Ther 36, 131-149. ethynylestradiol by human liver cytochrome P-450.
61. Durnas, C., CM. Loi, and B.J. Cusack (1990). Mol. Pharmacol. 33, 500-508.
Hepatic drug metabolism and aging. Clin. 73. Butler, M.A., M. Iwasaki, FP. Guengerich, and
Pharmacokinet. 19, 359-389. FF Kadlubar (1989). Human cytochrome P-450p^
62. George, J., K. Byth, and G.C Farrell (1995). Age (P-450j^2)' the phenacetin 0-deethylase, is prima-
but not gender selectively affects expression of indi- rily responsible for the hepatic 3-demethylation
vidual cytochrome P450 proteins in human liver. of caffeine and N-oxidation of carcinogenic ary-
Biochem. Pharmacol. 50, 727-730. lamines. Proc. Natl. Acad Sci. USA 86, 7696-7700.
63. Lu, A.Y.H., R.W. Wang, and J.H. Lin (2003). 74. Disderath, L.M. and FP Guengerich (1984).
Cytochrome P450 in vitro reaction phenotyping: Characterization of a human liver cytochrome P-450
A re-evaluation of approaches used for P450 involved in the oxidation of debrisoquine and other
Human P450s 467

drugs using antibodies raised to the analogous rat 85. Conney, A.H. (1982). Induction of microsomal
enzyme. Proc. Natl. Acad. Sci. USA 81, 7348-7352. enzymes by foreign chemicals and carcinogenesis
75. Soucek, P., M.V Martin, Y.-F. Ueng, and by polycyclic aromatic hydrocarbons: G. H, A.
F.P. Guengerich (1995). Identification of a common Clowes Memorial Lecture. Cancer Res. 42,
epitope near the conserved heme-binding region 4875-4917.
with polyclonal antibodies raised against 86. Sutter, T.R., K. Guzman, K.M. Dold, and
cytochrome P450 family 2 proteins. Biochemistry W.E Greenlee (1991). Targets for dioxin: Genes for
34, 16013-16021. plasminogen activator inhibitor-2 and interleukin-
76. Thomas, P.E., D. Koreniowski, D. Ryan, and \^. Science 254, 415^\S.
W. Levin (1979). Preparation of monospecific 87. Rivera, S.P, S.T. Saarikoski, and O. Hankinson
antibodies against two forms of rat liver cyto- (2002). Identification of a novel dioxin-inducible
chrome P-450 and quantitation of these antigens cytochrome P450. Mol. Pharmacol. 61, 255-259.
in microsomes. Arch. Biochem. Biophys. 192, 88. Shah, R.R., N.S. Gates, J.R. Idle, R.L. Smith, and
524-532. J.D.E Lockhart (1982). Impaired oxidation of
77. Venkatakrishnan, K., L.L. von Moltke, M.H. Court, debrisoquine in patients with perhexiline neuropa-
J.S. Harmatz, C.L. Crespi, and D.J. Greenblatt thy. Br Med. J. 284, 295-299.
(2000). Comparison between cytochrome P450 89. Steward, D.J., R.L. Haining, K.R. Henne, G. Davis,
(CYP) content and relative activity approaches to T.H. Rushmore, W.E Trager et al. (1997). Genetic
scaling from cDNA-expressed CYPs to human liver association between sensitivity to warfarin and
microsomes: Ratios of accessory proteins as sources expression of CYP2C9*3. Pharmacogenetics 1,
of discrepancies between the approaches. Drug 361-367.
Metab. Dispos. 28, 1493-1504. 90. Aithal, G.P, C.P Day, PJ. Kesteven, and A.K. Daly
78. Venkatakrishnan, K., L.L. von Moltke, and (1999). Association of polymorphisms in the
D.J. Greenblatt (2001). Application of the relative cytochrome P450 CYP2C9 with warfarin dose
activity factor approach in scaling from heterolo- requirement and risk of bleeding complications.
gously expressed cytochromes P450 to human liver Lancet 3,5^, 1X1-1\9.
microsomes: Studies on amitriptyline as a model 91. Daly, A.K., C.P Day, and G.P Aithal (2002).
substrate. J. Pharmacol. Exp. Then 291, 326-337. CYP2C9 polymorphism and warfarin dose require-
79. Venkatakrishnan, K., L.L. von Moltke, and ments. Brit. J. Clin. Pharmacol. 53, 408-409.
D.J. Greenblatt (2001). Human drug metabolism 92. Chiba, K., K. Kobayashi, K. Manabe, M. Tani,
and the cytochromes P450: Application and rele- T. Kamataki, and T. Ishizaki (1993). Oxidative
vance of in vitro models. J. Clin. Pharmacol. 41, metabolism of omeprazole in human liver micro-
1149-1179. somes: cosegregation with 5'-mephenytoin 4'-
80. Soars, M.G., H.V Gelboin, K.W. Krausz, and hydroxylation. J. Pharmacol. Exp. Ther 266,
R.J. Riley (2003). A comparison of relative abun- 52-59.
dance, activity factor and inhibitory monoclonal 93. Karam, W.G., J.A. Goldstein, J.M. Lasker, and
antibody approaches in the characterization of human B.I. Ghanayem (1996). Human CYP2C19 is a major
CYP enzymology. Br J. Clin. Pharmacol. 55, omeprazole 5-hydroxylase, as demonstrated with
175-181. recombinant cytochrome P450 enzymes. Drug
81. Schwab, M., M. Eichelbaum, and M.F. Fromm Metab. Dispos. 24, 1081-1087.
(2003). Genetic polymorphisms of the human mdr^ 94. Bolt, H.M., M. Bolt, and H. Kappus (1977).
drug transporter. Annu. Rev. Pharmacol. Toxicol. 43, Interaction of rifampicin treatment with pharmaco-
285-307. kinetics and metabolism of ethinyloestradiol in
82. Butler, M.A., N.R Lang, J.E Young, N.E. Caporaso, man. Acta Endocrinol. 85, 189-197.
P Vineis, R.B. Hayes et al. (1992). Determination of 95. Schwarz, U.I., B. Buschel, and W Kirch (2003).
CYP1A2 and NAT2 phenotypes in human popula- Unwanted pregnancy on self-medication with
tions by analysis of caffeine urinary metabolites. St. John's wort despite hormonal contraception.
Pharmacogenetics 2, 116-127. Br J. Clin. Pharmacol. 55, 112-113.
83. Ahsan, C.H., A.G. Renwick, B. Macklin, 96. Guengerich, EP (1999). Inhibition of drug
VE Challenor, D.G. Waller, and C.E George (1991). metabolizing enzymes: Molecular and biochemical
Ethnic differences in the pharmacokinetics of oral aspects. In T. E Woolf (ed.), Handbook of
nifedipine. Br J. Clin. Pharmacol. 31, 399^03. Drug Metabolism. Marcel Dekker, New York,
84. Kim, R.B., H. Yamazaki, M. Mimura, T. Shimada, pp. 203-227.
EP Guengerich, K. Chiba et al. (1996). 97. Bailey, D.G., J.D. Spence, B. Edgar, CD. Bayliff,
Chlorzoxazone 6-hydroxylation in Japanese and and J.M.O. Arnold (1990). Ethanol enhances the
Caucasians. In vitro and in vivo differences. J. hemodynamic effects of felodipine. Clin. Invest.
Pharmacol. Exp. Ther 279, 4-11. Med 12, 357-362.
468 F. Peter Guengerich

98. Edgar, B., D.G. Bailey, R. Bergstrand, G. Johnsson, P450 3A4-mediated interaction of diclofenac and
and L. Lurje (1990). Formulation dependent inter- quinidine. Drug Metab. Dispos. 28, 1043-1050.
action between felodipine and grapefruit juice. 112. Shapiro, A.B., K. Fox, R Lam, and V Ling (1999).
Clin. Pharmacol. Ther. 47, 181. Stimulation of P-glycoprotein-mediated drug
99. He, K., R. Iyer, R.N. Hayes, M.W. Sinz, T.F. Woolf, transport by prazosin and progesterone. Eur J.
and RF. Hollenberg (1998). Inactivation of Biochem. 259, 841-850.
cytochrome P450 3A4 by bergamottin, a compo- 113. Mueller, G.C. and J.A. Miller (1948). The metabo-
nent of grapefruit juice. Chem. Res. Toxicol. 11, lism of 4-dimethylaminoazobenzene by rat liver
252-259. homogenates. J. Biol. Chem. 176, 535-544.
100. Schmiedlin-Ren, R, D.J. Edwards, 114. Jollow, D.J., J.R. Mitchell, W.Z. Potter,
M.E. Fitzsimmons, K. He, K.S. Lown, RM. Woster D.C. Davis, J.R. Gillette, and B.B. Brodie (1973).
et al. (1997). Mechanisms of enhanced oral Acetaminophen-induced hepatic necrosis. II. Role
availability of CYP3A4 substrates by grapefruit of covalent binding in vivo. J. Pharmacol. Exp.
constituents. Drug Metab. Dispos. 25, 1228-1233. Ther 187, 195-202.
101. Reimers, D. and A. Jezek (1971). Rifampicin und 115. Neal, R.A. and J. Halpert (1982). Toxicology of
andere antituberkulotika bei gleichzeitiger oraler thiono-sulfur compounds. Annu. Rev. Pharmacol.
kontrzeption. Przx. Pneumolonology 25, 255-262. Toxicol. 22,321-339.
102. Nocke-Finck, L. H. Brewer, and D. Reimers 116. Wing, K.D., A.H. Glickman, and J.E. Casida
(1973). Wirkung van rifampicin auf den menstra- (1983). Oxidative bioactivation of S-alkyl phospho-
tionszylkus und die ostrogenausscheidung bei ein- rothiolate pesticides: Stereospecificity of profeno-
nahme oraler kontrazeptive. Deutsch. Med. fos insecticide activation. Science 219, 63-65.
Wochenschrifften 9H, 1521-1523. 117. Nebert, D.W. (1989). The Ah locus: Genetic differ-
103. Kivisto, K.T., RJ. Neuvonen, and U. Klotz (1994). ences in toxicity, cancer, mutation, and birth
Inhibition of terfenadine metabolism: Pharma- defects. Crit. Rev Toxicol. 20, 153-174.
cokinetic and pharmacodynamic consequences. 118. Lee, S.S.T., J.TM. Buters, T. Pineau, R Fernandez-
Clin. Pharmacokinet. 21, 1-5. Salguero, and F.J. Gonzalez (1996). Role of
104. Thompson, D. and G. Oster (1996). Use of terfe- CYP2E1 in the hepatotoxicity of acetaminophen.
nadine and contraindicated drugs. J. Am. Med. J. Biol. Chem. 271, 12063-12067.
Assoc, lis, 1339-1341. 119. Buters, J.T.M., S. Sakai, T Richter, T. Pineau, D.L.
105. Fischbach, T. and W. Lenk (1985). Additional routes Alexander, 0. Savas et al. (1999). Cytochrome
in the metabolism of phenacetin. Xenobiotica 15, P450 CYPIBI determines susceptibility to 7,12-
149-164. dimethylbenz[fl]anthracene-induced lymphomas.
106. Guengerich, F.R, D. Miiller-Enoch, and LA. Blair Proc. Natl. Acad. Sci. USA 96, 1977-1982.
(1986). Oxidation of quinidine by human liver 120. Nebert, D.W. and H.V. Gelboin (1968).
cytochrome P-450. Mol. Pharmacol. 30, 287-295. Substrate-inducible microsomal arylhydroxylase
107. Halpert, J.R. and FR Guengerich (1997). Enzyme in mammalian cell culture: Assay and proper-
inhibition and stimulation. In F P. Guengerich ties of induced enzyme. J. Biol. Chem. 243,
(ed.). Biotransformation, Vol. 3, Comprehen- 6242-6249.
sive Toxicology. Elsevier Science Ltd., Oxford, 121. Kellerman, G., M. Luyten-Kellerman, and
pp. 21-35. C.R. Shaw (1973). Genetic variation of aryl hydro-
108. Witherow, L.E. and J.B. Houston (1999). Sig- carbon hydroxylase in human lymphocytes. Am. J.
moidal kinetics of CYP3A substrates: An approach Hum. Genet. 25,327-331.
for scaling dextromethorphan metabolism in 122. Kellerman, G., C.R. Shaw, and M. Luyten-
hepatic microsomes and isolated hepatocytes to Kellerman (1973). Aryl hydrocarbon hydroxylase
predict in vivo clearance in rat. J. Pharmacol. Exp. inducibility and bronchogenic carcinoma. A'; Engl.
Then 290, 58-65. J. Med 298, 934-937.
109. Lasker, J.M., M.-T. Huang, and A.H. Conney 123. Paigen, B., E. Ward, A. Reilly, L. Houten,
(1982). In vivo activation of zoxazolamine metab- H.L. Gurtoo, J. Minowada et al (1981). Seasonal
olism by flavone. Science 216, 1419-1421. variation of aryl hydrocarbon hydroxylase activity
110. Lee, C.A., J.H. Lillibridge, S.D. Nelson, and in human lymphocytes. Cancer Res. 41,2757-2761.
J.T. Slattery (1996). Effects of caffeine and theo- 124. Kouri, R.E., C.E. McKinney, D.J. Slomiany,
phylline on acetaminophen pharmacokinetics: D.R. Snodgrass, N.P. Wray, and T.L. McLemore
P450 inhibition and activation. J. Pharmacol. Exp. (1982). Positive correlation between high aryl
Ther 211,2^7-291. hydrocarbon hydroxylase activity and primary
111. Ngui, J.S., W. Tang, R.A. Stearns, M.G. Shou, lung cancer as analyzed in cryopre served lympho-
R.R. Miller, Y. Zhang et al. (2000). Cytochrome cytes. Cancer Res. 42, 5030-5037.
Human P450s 469

125. Chang, C , D.R. Smith, VS. Prasad, C.L. Sidman, hydrocarbon hydroxylase represents CYPIBI, and
D.W. Nebert, and A. Puga (1993). Ten nucleotide not CYPlAl, in human freshly isolated white cells:
differences, five of which cause amino acid Trimodal distribution of Japaneses population
changes, are associated with the Ah receptor locus according to induction of CYPIBI mRNA by envi-
polymorphism of C57BL/6 and DBA/2 mice. ronmental dioxins. Cancer Epidemiol. Biomarkers
Pharmacogenetics 3, 312-321. Prev. 12,219-222.
126. Dolwick, K.M., J.V Schmidt, L.A. Carver, 136. Guengerich, F.P. (1998). The environmental genome
H.I. Swanson, and C.A. Bradfield (1993). Cloning project: Functional analysis of polymorphisms.
and expression of a human Ah receptor cDNA. Environ. Health Perspect. 106, 365-368.
Mol. Pharmacol 44, 911-917. 137. Ayesh, R., J.R. Idle, J.C. Ritchie, M.J. Crothers,
127. Fujii-Kuriyama, Y., M. Ema, J. Mimura, and M.R. Hetzel (1984). Metabolic oxidation phe-
N. Matsushita, and K. Sogawa (1995). Polymorphic notypes as markers for susceptibility to lung can-
forms of the Ah receptor and induction of the cer. A^a^re 312, 169-170.
CYPlAl gene. Pharmacogenetics 5, S149-S153. 138. d'Errico, A., E. Taioli, X. Chen, and P. Vineis
128. Kawajiri, K., J. Watanabe, H. Eguchi, K. Nakachi, (1996). Genetic metabolic polymorphisms and
C. Kiyohara, and S. Hayashi (1995). Polymorphisms the risk of cancer: A review of the literature.
of human Ah receptor gene are not involved in lung BioMarkers 1, 149-173.
cancer. Pharmacogenetics 5, 151-158. 139. Shimada, T and FR Guengerich (1991). Activa-
129. Hayashi, S., J. Watanabe, K. Nakachi, and tion of amino-a-carboline, 2-amino-l-methyl-
K. Kawajiri (1991). Genetic linkage of lung 6-phenylimidazo[4,5-^]pyridine, and a copper
cancer-associated Msp\ polymorphisms with phthalo-cyanine cellulose extract of cigarette
amino acid replacement in the heme binding smoke condensate by cytochrome P-450 enzymes
region of the human cytochrome P450IA1 gene. in rat and human liver microsomes. Cancer Res.
J. Biochem. (Tokyo) 110, 407-^11. 51,5284-5291.
130. Tefre, T., D. Ryberg, A. Haugen, D.W. Nebert, 140. Vineis, P. (2002). The relationship between poly-
V Skaug, A. Brogger et al (1991). Human CYPlAl morphisms of xenobiotic metabolizing enzymes and
(cytochrome Pj450) gene: Lack of association susceptibility to cancer. Toxicology 181, 457^62.
between the Msp I restriction fragment length poly- 141. Stoilov, I., A.N. Akarsu, I. Alozie, A. Child,
morphism and incidence of lung cancer in a M. Barsoum-Homsy, M.E. TuracH et al. (1998).
Norwegian population. Pharmacogenetics 1, Sequence analysis and homology modeling suggest
20-25. that primary congenital glaucoma on 2p21 results
131. Hirvonen, A., K. Husgafvel-Pursiainen, from mutations disrupting either the hinge region
A. Karjalainen, S. Anttila, and H. Vainio (1992). or the conserved core structures of cytochrome
Point-mutational MspX and Ile-Val polymorphisms P4501B1. ylm. J. Hum. Genet. 62, 573-584.
closely linked in the CYPlAl gene: Lack of asso- 142. Shimada, T, J. Watanabe, F.P. Guengerich,
ciation with susceptibility to lung cancer in a K. Inoue, and E.M.J. Gillam (2001). Specificity
Finnish study population. Cancer Epidemiol. of 17p-oestradiol and benzo[a]pyrene oxidation
Biomarkers Prev. 1, 485^89. by polymorphic human cytochrome P450 IBl
132. Wedlund, PJ., S. Kimura, EL Gonzalez, and variants substituted at residues 48, 119, and 432.
D.W. Nebert (1994). I462V mutation in the human Xenobiotica 31, 163-176.
CYPlAl gene: Lack of correlation with either the 143. Watanabe, J., T. Shimada, E.M.J. Gillam, T. Ikuta,
Msp I 1.9kb (M2) allele or CYPlAl inducibility K. Suemasu, Y Higashi et al. (2000). Association
in a three-generation family of East Mediterranean of CYPIBI genetic polymorphism with incidence
descent. Pharmacogenetics 4, 21-26. to breast and lung cancer. Pharmacogenetics 10,
133. Zhang, Z.-Y, M.J. Fasco, L. Huang, F.P. Guengerich, 25-33.
and L.S. Kaminsky (1996). Characterization of puri- 144. Zheng, W, D.W. Xie, F Jin, J.R. Cheng, Q. Dai,
fied recombinant human CYP 1 Al-Ile462 and NdX^^^\ WQ. Wen et al. (2000). Genetic polymorphism of
Assessment of a role for the rare allele in carcino- cytochrome P450-1B1 and risk of breast cancer.
genesis. Cancer Res. 56, 3926-3933. Cancer Epidemiol. Biomarkers Prev. 9, 147-150.
134. Persson, I., I. Johansson, and M. Ingelman-Sundberg 145. Lang, N.R, M.A. Butler, J. Massengill, M. Lawson,
(1997). In vitro kinetics of two human CYPlAl R.C. Stotts, M. Maurer-Jensen etal. (1994). Rapid
variant enzymes suggested to be associated with metabolic phenotypes for acetyltransferase and
interindividual differences in cancer susceptibility. cytochrome P4501A2 and putative exposure to
Biochem. Biophys. Res. Commun. 231, 227-230. food-borne heterocyclic amines increase the risk
135. Toide, K., H. Yamazaki, R. Nagashima, K. Itoh, for colorectal cancer or polyps. Cancer Epidemiol.
S. Iwano, Y Takahashi et al. (2003). Aryl Biomarkers Prev. 3, 675-682.
470 F. Peter Guengerich

146. Shimada, T., M. Iwasaki, M.V Martin, and benzo[^]fluoranthene in human hepatocytes:
F.P. Guengerich (1989). Human liver microsomal CYPlAl or CYP1A2?. Arch. Biochem. Biophys.
cytochrome P-450 enzymes involved in the bioac- 389, 130-134.
tivation of procarcinogens detected by umu gene 159. Fagan, J.B., J.V Pastewka, S.R. Chalberg,
response in Salmonella typhimurium TA1535/ E. Gozukara, F.P. Guengerich, and H.V Gelboin
pSK1002. Cancer Res. 49, 3218-3228. (1986). Noncoordinate regulation of the mRNAs
147. Ariyoshi, N., M. Miyamoto, Y. Umetsu, encoding cytochromes P-450g and P-
H. Kunitoh, H. Dosaka-Akita, Y. Sawamura et al. 450,, , Arch. Biochem. Biophys. 244,261-272.
(2002). Genetic polymorphism of CYP2A6 gene 160. Kim, J.H., M.E. Sherman, EC. Curreiro, FP
and tobacco-induced lung cancer risk in male Guengerich, PT. Strickland, and T.R. Sutter (2004,
smokers. Cancer Epidemiol. Biomarkers Prev. 11, in press). Expression of cytochromes P450 lAl and
890-894. IBl in human lung from smokers, non-smokers,
148. Pianezza, M.L., E.M. Sellers, and R.F. Tyndale and ex-smokers. Toxicol. Appl. Pharmacol.
(1998). Nicotine metabolism defect reduces smok- 161. Prough, R.A., Z. Sipal, and S.W. Jakobsson (1977).
ing. Nature 393, 750. Metabolism of benzo(a)pyrene by human lung
149. Kim, R.B., D. O'Shea, and G.R. Wilkinson (1994). microsomal fractions. Life Sci. 21, 1629-1636.
Relationship in healthy subjects between CYP2E1 162. Fujino, T, K. Gottlieb, D.K. Manchester, S.S. Park,
genetic polymorphism and the 6-hydroxylation of D. West, H.L. Gurtoo et al. (1984). Monoclonal anti-
chlorzoxazone: A putative measure of CYP2E1 body phenotyping of interindividual differences in
activity. Pharmacogenetics 4, 162-165. cytochrome P-450-dependent reactions of single and
150. loannides, C. and D.V Parke (1993). Induction of twin human placenta. Cancer Res. 44, 3916-3923.
cytochrome P4501 as an indicator of potential 163. Robie-Suh, K., R. Robinson, H.V Gelboin, and
chemical carcinogenesis. Drug Metab. Rev. 25, F.P. Guengerich (1980). Aryl hydrocarbon
485-501. hydroxylase is inhibited by antibody to rat liver
151. Rice, J.M., B.A. Diwan, J.M. Ward, R.W. Nims, and cytochrome P-450. Science 208, 1031-1033.
R.A. Lubet (1992). Phenobarbital and related com- 164. Shimada, T, H. Yamazaki, M. Mimura,
pounds: Approaches to interspecies extrapolation. N. Wakamiya, F.P. Guengerich, and Y Inui (1996).
In Relevance ofAnimal Studies to the Evaluation of Characterization of microsomal cytochrome P450
Human Cancer Risk. Wiley-Liss, Inc., pp. 231-249. enzymes involved in the oxidation of xenobiotic
152. Olsen, J.H., J.D. Boice Jr., J.PA. Jensen and chemicals in human fetal livers and adult lungs.
J.F. Fraumeni Jr. (1989). Cancer among epileptic Drug Metab. Dispos. 24, 515-522.
patients exposed to anticonvulsant drugs. J. Natl. 165. McLemore, T.L., S. Adelberg, M.C. Liu,
Cancer Inst. 81,803-808. N.A. McMahon, S.J. Yu, W.C. Hubbard et al
153. Kluwe, WM. (1994). The relevance of hepatic per- (1990). Expression of CYPlAl gene in patients
oxisome proliferation in rats to assessment of with lung cancer: Evidence for cigarette smoke-
human carcinogenic risk for pharmaceuticals. induced gene expression in normal lung tissue and
Regul. Toxicol. Pharmacol. 20, 170-186. for altered gene regulation in primary pulmonary
154. Thomas, R.S., D.R. Rank, S.G. Penn, carcinomas. .( A^a//. Cancer Inst. 82, 1333-1339.
G.M. Zastrow, K.R. Hayes, K. Pande et al. (2001). 166. Anttila, S., X.D. Lei, E. Elovaara, A. Karjalainen,
Identification of toxicologically predictive gene W.M. Sun, H. Vainio et al. (2000). An uncommon
sets using cDNA microarrays. Mol. Pharmacol. phenotype of poor inducibility of CYPlAl in
60, 1189-1194. human lung is not ascribable to polymorphisms in
155. Omiecinski, C.J., C.A. Redlich, and P Costa the AHR, ARNT, or CYPlAl genes. Pharmaco-
(1990). Induction and developmental expression of genetics 10, 741-751.
cytochrome P450IA1 messenger RNA in rat and 167. Anttila, S., P. Tuominen, A. Hirvonen, M. Nurminen,
human tissues: Detection by the polymerase chain A. Karjalainen, O. Hmkmsonetal. (2001). CYPlAl
reaction. Cancer Res. 50, 4315^321. levels in lung tissue of tobacco smokers and poly-
156. Kitada, M., M. Taneda, K. Itahashi, andT. Kamataki morphisms of CYPlAl and aromatic hydrocarbon
(1991). Four forms of cytochrome P-450 in human receptor. Pharmacogenetics 11, 501-509.
fetal liver: Purification and their capacity to activate 168. Bradfield, C.A. (2004, in press). Induction of P450
promutagens. Jpn. J. Cancer Res. 82, 426-432. enzymes: Receptors. In P. R. Ortiz De Montellano
157. Kitada, M. and T. Kamataki (1994). Cytochrome (ed.), Cytochrome P450: Structure, Mechanism,
P450 in human fetal liver: Significance and fetal- and Biochemistry, Plenum, New York (Chap. 8 of
specific expression. Drug Metab. Rev. 26, 305-323. this monograph).
158. Liu, N., Q.Y Zhang, D. Vakharia, D. Dunbar, and 169. Lucier, G.W, K.G. Nelson, R.B. Everson,
L.S. Kaminsky (2001). Induction of CYPIA by T.K. Wong, R.M. Philpot, T. Tiernan et al (1987).
Human P450s 471

Placental markers of human exposure to polychlo- 181. Shimada, T, H. Yamazaki, M. Foroozesch,


rinated biphenyls and polychlorinated dibenzofu- N.E. Hopkins, W.L. Alworth, and F.R Guengerich
rans. Environ. Health Perspect. 76, 79-87. (1998). Selectivity of polycyclic inhibitors for
170. Diaz, D., I. Fabre, M. Daujat, B. Saintaubert, human cytochromes P450 lAl, 1A2, and IBl.
P. Bories, H. Michel et al. (1990). Omeprazole is Chem. Res. Toxicol. 11, 1048-1056.
an aryl hydrocarbon-like inducer of human hepatic 182. McManus, M.E., W.M. Burgess, M.E. Veronese,
cytochrome-P450. Gastroenterology 99, 12>1-1A1. A. Huggett, L.C. Quattrochi, and R.H. Tukey
171. Yun, C.-H., T. Shimada, and F.R Guengerich (1990). Metabolism of 2-acetylaminofluorene and
(1992). Roles of human liver cytochrome P-4502C benzo[(2]pyrene and activation of food-derived het-
and 3A enzymes in the 3-hydroxylation of erocyclic amine mutagens by human cytochromes
benzo[«]pyrene. Cancer Res. 52, 1868-1874. P-450. Cancer Res. 50, 3367-3376.
172. Prough, R.A., M.D. Burke, and R.T. Mayer (1978). 183. Mercurio, M.G., S.J. Shiff, R.A. Galbraith, and
Direct fluorometric methods for measuring mixed- S. Sassa (1995). Expression of cytochrome P450
function oxidase activity. Meth. Enzymol. 52, mRNAs in the colon and the rectum in normal
372-377. human subjects. Biochem. Biophys. Res. Commun.
173. Guo, Z., E.M.J. Gillam, S. Ohmori, R.H. Tukey, 210, 350-355.
and P.P. Guengerich (1994). Expression of modi- 184. Pantuck, E.J., K.-C. Hsiao, A. Maggio,
fied human cytochrome P450 lAl in Escherichia K. Nakamura, R. Kuntzman, and A.H. Conney
coli: Effects of 5' substitution, stabilization, purifi- (1974). Effect of cigarette smoking on phenacetin
cation, spectral characterization, and catalytic metabolism. Clin. Pharmacol. Then 15, 9-17.
properties. Arch. Biochem. Biophys. 312, 436-446. 185. Vesell, E.S. and J.G. Page (1968). Genetic control of
174. Shou, M., K.R. Korzekwa, C.L. Crespi, drug levels in man: Antipyrine. Science 161, 72-73.
F.J. Gonzalez, and H.V Gelboin (1994). The role of 186. Rasmussen, B.B., T.H. Brix, K.O. Kyvik, and
12 cDNA-expressed human, rodent, and rabbit K. Brosen (2002). The interindividual differences
cytochromes P450 in the metabolism of in the 3-demethylation of caffeine alias CYP1A2
benzo[a]pyrene and benzo[fl]pyrene trans-l,S- is determined by both genetic and environmental
dihydrodiol. M?/. Carcinogen. 10, 159-168. factors. Pharmacogenetics 12, 473-478.
175. Bauer, E., Z. Guo, Y.-E Ueng, L.C. Bell, and 186a.Zhou, H., RD. Josephy, D. Kim, and ER
F.R Guengerich (1995). Oxidation of benzo[a]- Guengerich (2004). Functional characterization of
pyrene by recombinant human cytochrome P450 four allelic variants of human cytochrome P450
enzymes. Chem. Res. Toxicol. 8, 136-142. 1A2. Arch. Bioch. Biophys. 422, 23-30.
176. Shou, M., K.W. Krausz, F.J. Gonzalez, and 187. Kalow, W. and B.K. Tang (1993). The use of
H.V Gelboin (1996). Metabolic activation of caffeine for enzyme assays: A critical appraisal.
the potent carcinogen dibenzo[flf,/?]anthracene by Clin. Pharmacol. Ther 53, 503-514.
cDNA-expressed human cytochromes P450. Arch. 188. Quattrochi, L.C, T. Vu, and R.H. Tukey (1994).
Biochem. Biophys. 328, 201-207. The human CYP1A2 gene and induction by
177. Shou, M., K.W. Krausz, EJ. Gonzalez, and 3-methylcholanthrene: A region of DNA that sup-
H.V Gelboin (1996). Metabolic activation of the ports Ah-receptor binding and promoter-specific
potent carcinogen dibenzo[fl,/]pyrene by human induction. J. Biol. Chem. 269, 6949-6954.
recombinant cytochromes P450, lung and liver 189. Kondraganti, S.R., W.W. Jiang, and B. Moorthy
microsomes. Carcinogenesis 17, 2429-2433. (2002). Differential regulation of expression of
178. Shimada, T, C.L. Hayes, H. Yamazaki, S. Amin, hepatic and pulmonary cytochrome P4501A
S.S. Hecht, ER Guengerich et al. (1996). Activation enzymes by 3-methylcholanthrene in mice lacking
of chemically diverse procarcinogens by human the CYP1A2 gene. J. Pharmacol. Exp. Ther 303,
cytochrome P450 IBl. Cancer Res. 56,2979-2984. 945-951.
179. Balani, S.K., H.J.C. Yeh, D.E. Ryan, RE. Thomas, 190. Fisher, G.J., H. Fukushima, and J.L. Gaylor
W. Levin, and D.M. Jerina (1985). Absolute con- (1981). Isolation, purification, and properties of a
figuration of the 5,6-oxide formed from 7,12- unique form of cytochrome P-450 in microsomes
dimethylbenz[a]anthracene by cytochrome P450c. of isosafrole-treated rats. J. Biol. Chem. 256,
Biochem. Biophys. Res. Commun. 130, 610-616. 4388-4394.
180. Lewis, D.EV, B.G. Lake, S.G. George, M. Dickins, 191. Vistisen, K., H.E. Poulsen, and S. Loft (1992).
RJ. Eddershaw, M.H. Tarbit et al. (1999). Foreign compound metabolism capacity in man
Molecular modeling of CYP family enzymes measured from metabolites of dietary caffeine.
CYPlAl, CYP1A2, CYP1A6, and CYPIBI based Carcinogenesis 13, 1561-1568.
on sequence homology with CYP 102. Toxicology 192. Rost, K.L., H. Brosicke, J. Brockmoller,
139, 53-79. M. Scheffler, H. Helg, and L Roots (1992).
472 F. Peter Guengerich

Increase of cytochrome P450IA2 activity by involved in theophylline 8-hydroxylation.


omeprazole: Evidence by the ^^C-[A^-3-methyl]- Biochem. Pharmacol. 50, 205-211.
caffeine breath test in poor and extensive metabo- 202. Yamazaki, H., Y. Oda, Y Funae, S. Imaoka, Y Inui,
Hzers of *S-mephenytoin. Clin. Pharmacol. Then F.P. Guengerich et al. (1992). Participation of rat
52, 170-180. liver cytochrome P450 2E1 in the activation of
193. Fisher, C.W., D.L. Caudle, C. Martin-Wixtrom, A^-nitrosodimethylamine and A^-diethylnitrosamine
L.C. Quattrochi, R.H. Tukey, M.R. Waterman et al to products genotoxic in Salmonella typhimurium
(1992). High-level expression of functional NM2009. Carcinogenesis 13, 979-985.
cytochrome P450 1A2 in Escherichia coli, 203. Yamazaki, H., RM. Shaw, ER Guengerich, and
FASEB J. 6,759-164. T Shimada (1998). Roles of cytochromes P450
194. Sandhu, P., Z. Guo, T. Baba, M.V. Martin, 1A2 and 3A4 in the oxidation of estradiol and
R.H. Tukey, and F.P. Guengerich (1994). Expres- estrone in human liver microsomes. Chem. Res.
sion of modified human cytochrome P450 1A2 in Toxicol. 11,659-665.
Escherichia coli: Stabilization, purification, spec- 204. Michnovicz, J.J., R.J. Hershcopr, H. Naganuma,
tral characterization, and catalytic activities of the H.L. Bradlow, and J. Fishman (1986). Increased
enzyme. ^rcA. Biochem. Biophys. 309, 168-177. 2-hydroxylation of estradiol as a possible mecha-
195. Patten, C.J., RE. Thomas, R.L. Guy, M. Lee, nism for the anti-estrogenic effect of cigarette
RJ. Gonzalez, ER Guengerich et al. (1993). smoking. New Engl. J. Med. 315, 1305-1309.
Cytochrome P450 enzymes involved in acetamin- 205. Bradlow, H.L., R.J. Hershcopf, C.R Martucci, and
ophen activation by rat and human liver micro- J. Fishman (1985). Estradiol 16a-hydroxylation in
somes and their kinetics. Chem. Res. Toxicol. 6, the mouse correlates with mammary tumor inci-
511-518. dence and presence of murine mammary tumor
196. Engel, G., U. Hofmann, H. Heidemann, J. Cosme, virus: A possible model for the hormonal etiology
and M. Eichelbaum (1996). Antipyrine as a probe of breast cancer in humans. Proc. Natl. Acad. Sci.
for human oxidative drug metabolism: Identifica- USA 82, 6295-6299.
tion of the cytochrome P450 enzymes catalyzing 206. Hiroya, K., M. Ishigooka, T. Shimizu, and
4-hydroxyantipyrine, 3-hydroxymethylantipyrine, M. Hatano (1992). Role of Glu318 and Thr319
and norantipyrine formation. Clin. Pharmacol. in the catalytic function of cytochrome P450j
Then 59, 613-623. (P4501A2): Effects of mutations on the methanol
197. Yamazaki, H., Z. Guo, M. Persmark, M. Mimura, hydroxylation. FASEB J. 6, 749-751.
EJ. Gonzalez, C. Sugahara et al. (1994). Bufuralol 207. Ishigooka, M., T. Shimizu, K. Hiroya, and
hydroxylation by cytochrome P450 2D6 and 1A2 M. Hatano (1992). Role of Glu318 at the putative
enzymes in human liver microsomes. Mol. distal site in the catalytic function of cytochrome
Pharmacol. 46, 568-577. P450j. Biochemistry 3\, 1528-1531.
198. Berthou, E, V Carriere, D. Ratanasavanh, 208. Shimizu, T, A.J.M. Sadeque, G.N. Sadeque,
T Goasdufif, E Morel, J.C. Gautier et al. (1993). M. Hatano, and Y Fujii-Kuriyama (1991). Ligand
On the specificity of chlorzoxazone as drug binding studies of engineered cytochrome P-450j
probe of cytochrome P4502E1. In Abstracts, wild type, peroximal mutants, and distal mutants.
5th European ISSXMeeting, Vol. 3. p. 116, 26-29 Biochemistry 30, 1490-1496.
September, Tours, France. 209. Fuhr, U, G. Strobl, E Manaut, E.M. Anders,
199. Woolf, TE, W.E Pool, M. Kukan, S. Bezek, E Sorgel, E. Lopez-de-Brinas et al. (1993).
K. Kunze, and W.E Trager (1993). Characterization Quinolone antibacterial agents: Relationship
of tacrine metabolism and bioactivation using between structure and in vitro inhibition of the
heterologous expression systems and inhibition human cytochrome P450 isoform CYP1A2.
studies: Evidence for CYP1A2 involvement. Mol. Pharmacol. 43, 191-199.
In Abstracts, 5th North American ISSX Meeting, 210. Lozano, J.J., E. L6pez-de-Briiias, N.B. Centeno,
Vol. 4, p. 139, 17-21 October, Tucson, AZ. E Sanz, and R. Guigo (1997). Three-dimensional
200. Benoit, G.G., C.E Naud, M.A. Simard, and modelling of human cytochrome P450 1A2 and
A.L. Astier (1997). Noninterference of cytochrome its interaction with caffeine and MelQ. J. Comput.
P4501A2 in the cytotoxicity of tacrine using genet- Aided Mol. Des. 11, 395^08.
ically engineered V79 Chinese hamster cells for 211. Parikh, A., P.D. Josephy, and F.P. Guengerich
stable expression of the human or rat isoform and (1999). Selection and characterization of human
two human hepatocyte cell lines. Biochem. cytochrome P450 1A2 mutants with altered cat-
Pharmacol. 53, 423-427. alytic properties. Biochemistry 38, 5283-5289.
201. Zhang, Z.Y. and L.S. Kaminsky (1995). 212. Yun, C.-H., G.R Miller, and EP Guengerich
Characterization of human cytochromes P450 (2000). Rate-determining steps in phenacetin
Human P450s 473

oxidation by human cytochrome P450 1A2 and mutations in cytochrome P4501B1 (CYPIBI) as
selected mutants. Biochemistry 39, 11319-11329. the principal cause of primary congenital glaucoma
213. Yun, C.-H., G.P. Miller, and F.P. Guengerich (2001). (Buphthalmos) in families linked to the GLC3A
Oxidations of /7-alkoxyacylanilides by human locus on chromosome 2p21. Hum. Mol. Genet. 6,
cytochrome P450 1A2: Structure-activity relation- 641-647.
ships and simulation of rate constants of individual 225. Shimada, T, E.M.J. Gillam, T.R. Sutter,
steps in catalysis. Biochemistry 40, 4521-4530. PT. Strickland, F.P. Guengerich, and H. Yamazaki
214. Miller, G.P and F.P Guengerich (2001). Binding (1997). Roles of recombinant human cytochrome
and oxidation of alkyl 4-nitrophenyl ethers by P450 IBl in the oxidation of xenobiotic chemicals.
rabbit cytochrome P450 1A2: Evidence for two Drug Metab. Dispos. 25, 617-622.
binding sites. Biochemistry 40, 7262-7272. 226. Shimada, T, Y Oda, E.M.J. Gillam,
215. Racha, J.K., A.E. Rettie, and K.L. Kunze (1998). F.P. Guengerich, and K. Inoue (2001). Metabolic
Mechanism-based inactivation of human cyto- activation of polycyclic aromatic hydrocarbons
chrome P450 1A2 by fiirafylline: Detection of a and their dihydrodiol derivatives and other pro-
1:1 adduct to protein and evidence for the forma- carcinogens by cytochrome P450 lAl and IBl
tion of a novel imidazomethide intermediate. allelic variants and other human cytochrome P450
Biochemistry 37, 7407-7419. enzymes in Salmonella typhimurium NM2009.
216. Voorman, R. and S.D. Aust (1987). Specific bind- Drug Metab. Dispos. 29, 1176-1182.
ing of polyhalogenated aromatic hydrocarbon 227. Yamazaki, H., N. Hatanaka, R. Kizu, K. Hayakawa,
inducers of cytochrome P-450j to the cytochrome N. Shimada, FP Guengerich et al. (2000).
and inhibition of its estradiol 2-hydroxylase activ- Bioactivation of diesel exhaust particle extracts
ity. Toxicol. Appl. Pharmacol 90, 69-78. and their major nitrated polycyclic aromatic hydro-
217. Sesardic, D., A. Boobis, B. Murray, S. Murray, carbon components, 1-nitropyrene and dinitropy-
J. Segura, R. De La Torre et al (1990). FurafyUine is renes, by human cytochrome P450s lAl, 1A2, and
a potent and selective inhibitor of cytochrome P450 IBl. Chem. Res. Toxicol. 411, 129-138.
1A2 in man. Br. J. Clin. Pharmacol. 29, 651-663. 228. Hayes, C.L., D.C. Spink, B.C. Spink, J.Q. Cao,
218. Kappas, A., A.P Alvares, K.E. Anderson, N.J. Walker, and TR. Sutter (1996). 17p-Estradiol
E.J. Pantuck, C.B. Pantuck, R. Chang et al. (1978). hydroxylation catalyzed by human cytochrome P450
Effect of charcoal-broiled beef on antipyrine and IBl. Proc. Natl. Acad Sci. USA 93, 9776-9781.
theophylline metabolism. Clin. Pharmacol. Ther 229. Shimada, T, J. Watanabe, K. Kawajiri, T.R. Sutter,
23, 445-450. F.P Guengerich, E.M.J. Gillam et al. (1999). Cata-
219. Feldman, C.H., VE. Hutchinson, C.E. Pippenger, lytic properties of polymorphic human cytochrome
T.A. Blemenfeld, B.R. Feldman, and W.J. Davis P450 IBl variants. Carcinogenesis 20, 1607-1614.
(1980). Effect of dietary protein and carbohydrate 230. Shimada, T, E.M.J. Gillam, Y. Oda, F. Tsumura,
on theophylline metabolism in children. Pediatrics T.R. Sutter, RP Guengerich et al. (1999). Meta-
66, 956-962. bolism of benzo[fl]pyrene to ^ra«5-7,8-dihydroxy-
220. Otto, S., C. Marcus, C. Pidgeon, and C. Jefcoate 7,8-dihydrobenzo[fl]pyrene by recombinant human
(1991). A novel adrenocorticotropin-inducible cytochrome P450 IBl and purified liver epoxide
cytochrome P450 from rat adrenal microsomes hydrolase. Chem. Res. Toxicol. 12, 623-629.
catalyzes polycyclic aromatic hydrocarbon metab- 231. Savas, U., C.P Carstens, and C.R. Jefcoate (1997).
olism. Endocrinology 129, 970-982. Recombinant mouse CYPIBI expressed in
221. Murray, B.P and M.A. Correia (2001). Ubiquitin- Escherichia coli exhibits selective binding by poly-
dependent 26S proteasomal pathway: A role cyclic hydrocarbons and metabolism which paral-
in the degradation of native human liver CYP3 A4 lels C3H10T1/2 cell microsomes, but differs from
expressed in Saccharomyces cerevisiael Arch. human recombinant CYPIBI. Arch. Biochem.
Biochem. Biophys. 393, 106-116. Biophys. 347, 181-192.
222. Murray, G.I., M.C. Taylor, M.C. McFadyen, 232. Hanna, I.H., S. Dawling, N. Roodi, F.P. Guengerich,
J.A. McKay, W.F Greenlee, M.D. Burke et al. and R Pari (2000). Cytochrome P450 IBl (CYPIBI)
(1997). Tumor-specific expression of cytochrome pharmacogenetics: Association of polymorphisms
P450 CYPIBI. Cancer Res. 57, 3026-3031. with functional differences in estrogen hydroxyla-
223. Chang, TK.H., J. Chen, V Pillay, J.-Y. Ho, and tion activity. Cancer Res. 60, 3440-3444.
S.M. Bandiera (2003). Real-time polymerase chain 233. Shou, M., K.R. Korzekwa, E.N. Brooks,
reaction analysis of CYPIBI gene expression in K.W Krausz, F.J. Gonzalez, and H.V Gelboin
human liver. Toxicol. Sci. 71, 11-19. (1997). Role of human hepatic cytochrome P450
224. Stoilov, I., A.N. Akarsu, and M. Sarfarazi 1A2 and 3A4 in the metabolic activation of
(1997). Identification of three different truncating estrone. Carcinogenesis 18, 207-214.
474 F. Peter Guengerich

234. Bolton, XL., E. Pisha, F. Zhang, and S. Qiu (1998). expression and relevance to nitrosamine metabo-
Role of quinoids in estrogen carcinogenesis. lism. Carcinogenesis 23, 611-616.
Chem. Res. Toxicol. 11, 1113-1127. 246. Raunio, H., R. Juvonen, M. Pasanen, O. Pelkonen,
235. Liehr, J.G., M.J. Ricci, C.R. Jefcoate, R Paakko, and Y Soini (1998). Cytochrome
E.V Hannigan, J.A. Hokanson, and B.T. Zhu P4502A6 (CYP2A6) expression in human hepato-
(1995). 4-Hydroxylation of estradiol by human cellular carcinoma. Hepatology 27, 427-432.
uterine myometrium and myoma microsomes: 247. Rae, J.M., M.D. Johnson, M.E. Lippman, and
Implications for the mechanism of uterine tumori- D.A. Flockhart (2001). Rifampin is a selective,
genesis. Proc. Natl. Acad. Sci. USA 92, 9220-9224. pleiotropic inducer of drug metabolism genes in
236. Lewis, D.F.V, M. Dickins, RJ. Eddershaw, human hepatocytes: Studies with cDNA and
M.H. Tarbit, and RS. Goldfarb (1999). Cyto- oligonucleotide expression arrays. J. Pharmacol.
chrome P450 substrate specificities, substrate Exp. Ther 299, 849-857.
structural templates and enzyme active site geome- 248. Donato, M.T., R Viitala, C. Rodriguez-Antona,
tries. Drug Metabol Drug Interact. 15, 1^9. A. Lindfors, J.V Castell, H. Raunio et al. (2000).
237. Jang, M., L. Cai, G.O. Udeani, K.V Slowing, C.R CYP2A5/CYP2A6 expression in mouse and
Thomas, C.W.W. Beecher et al (1997). Cancer human hepatocytes treated with various in vivo
chemopreventive activity of resveratrol, a natural inducers. Drug Metab. Dispos. 28, 1321-1326.
product derived from grapes. Science 275,218-220. 249. Jover, R., R. Bort, M.J. Gomez-Lechon, and
238. Chun, Y.-J., M.-R Kim, and RR Guengerich (1999). J.V. Castell (2001). Cytochrome P450 regulation
Resveratrol is a selective human cytochrome P450 by hepatocyte nuclear factor 4 in human hepato-
lAl inhibitor. Biochem. Biophys. Res. Commun. cytes: A study using adenovirus-mediated anti-
262, 20-24. sense targeting. Hepatology 33, 668-675.
239. Potter, G.A., L.H. Patterson, E. Wanogho, 250. Ding, S., B.C. Lake, T. Friedberg, and C.R. Wolf
RJ. Perry, PC. Butler, T. Ijaz et al. (2002). The (1995). Expression and alternative sphcing of the
cancer preventative agent resveratrol is converted to cytochrome P-450 CYP2A7. Biochem. J. 306,
the anticancer agent piceatannol by the cytochrome 161-166.
P450 enzyme CYPIBI. Br J. Cancer 86, 774-778. 251. Oscarson, M., R.A. McLellan, V Asp, M. Ledesma,
240. Chun, Y.-J., S. Kim, D. Kim, S.-K. Lee, and M.L. Ruiz, B. Sinues etal. (2002). Characterization
RR Guengerich (2001). A new selective and potent of a novel CYP2A7/CYP2A6 hybrid allele
inhibitor of human cytochrome P450 IBl and its {CYP2A6*]2) that causes reduced CYP2A6 activ-
application to antimutagenesis. Cancer Res. 61, ity Hum. Mutat. 20, 275-283.
8164-8170. 252. Hadidi, H., K. Zahlsen, J.R. Idle, and S. Cholerton
241. Shen, L., S. Qiu, R.B. van Breemen, R Zhang, (1997). A single amino acid substitution
Y. Chen, and J.L. Bolton (1997). Reaction of the (Leul60His) in cytochrome P450 CYP2A6 causes
premarin metabolite 4-hydroxyequilenin semi- switching from 7-hydroxylation to 3-hydroxylation
quinone radical with 2'-deoxyguanosine: Formation of coumarin. Food Chem. Toxicol. 35, 903-907.
of unusual cyclic adducts. J. Am. Chem. Soc. 119, 253. Nakajima, M., J.-T. Kwon, N. Tanaka, T. Zenta,
11126-11127. Y. Yamamoto, H. Yamamoto et al, (2001).
242. Yamano, S., J. Tatsuno, and RJ. Gonzalez (1990). Relationship between interindividual differences in
The CYP2A3 gene product catalyzes coumarin nicotine metabolism and CYP2A6 genetic polymor-
7-hydroxylation in human liver microsomes. phism in humans. Clin. Pharmacol. Ther 69, 72-78.
Biochemistry 19, 1322-1329. 254. Tyndale, R.R and E.M. Sellers (2001). Variable
243. Hakkola, J., M. Pasanen, J. Hukkanen, O. Pelkonen, CYP2A6-mediated nicotine metabolism alters
J. Maenpaa, R.J. Edwards et al. (1996). Expression smoking behavior and risk. Drug Metab. Dispos.
of xenobiotic-metabolizing cytochrome P450 forms 29, 548-552.
in human full-term placenta. Biochem. Pharmacol. 255. Yoshida, R., M. Nakajima, Y Watanabe, J.T. Kwon,
51,403-411. and T. Yokoi (2002). Genetic polymorphisms in
244. Ding, X. and L.S. Kaminsky (2003). Human extra- human CYP2A6 gene causing impaired nicotine
hepatic cytochromes P450: Function in xenobiotic metabolism. Br J. Clin. Pharmacol 54, 511-517.
metabolism and tissue-selective chemical toxicity 256. Gu, D.R, L.J. Hinks, N.E. Morton, and I.N. Day
in the respiratory and gastrointestinal tracts. Annu. (2000). The use of long PCR to confirm three
Rev. Pharmacol. Toxicol. 43, 149-173. common alleles at the CYP2A6 locus and the rela-
245. Godoy W., R.M. Albano, E.G. Moraes, PR. Pinho, tionship between genotype and smoking habit.
R.A. Nunes, E.H. Saito et al. (2002). CYP2A6/2A7 Ann. Hum. Genet. 64, 383-390.
and CYP2E1 expression in human oesophageal 257. Rao, Y, E. Hoffmann, M. Zia, L. Bodin,
mucosa: Regional and inter-individual variation in M. Zeman, E.M. Sellers et al. (2000). Duplications
Human P450s 475

and defects in the CYP2A6 gene: Identification, chromatographic-fluorescence detection method


genotyping, and in vivo effects on smoking. Mol. with a spectrofluorometric method for the determi-
Pharmacol. 58, 747-755. nation of 7-hydroxycoumarin in human urine.
258. Xu, C, S. Goodz, E.M. Sellers, and R.F. Tyndale J. Chromatogr 575, 325-330.
(2002). CYP2A6 genetic variation and potential con- 272. Rautio, A., H. Kraul, A. Kojo, E. Salmela, and
sequences. ^Jv. DrugDeliv. Rev. 54, 1245-1256. O. Pelkonen (1992). Interindividual variability of
259. Tyndale, R.F. and E.M. Sellers (2002). Genetic coumarin 7-hydroxylation in healthy volunteers.
variation in CYP2A6-mediated nicotine metabo- Pharmacogenetics 2, 227-233.
lism alters smoking behavior. Ther. Drug Monit. 273. Soucek, P. (1999). Expression of cytochrome P450
24, 163-171. 2A6 in Escherichia coli: Purification, spectral and
260. Kamataki, T., K. Nunoya, Y. Sakai, H. Kushida, catalytic characterization, and preparation of poly-
and K. Fujita (1999). Genetic polymorphism of clonal antibodies. Arch. Biochem. Biophys. 370,
CYP2A6 in relation to cancer. Mutat. Res. 428, 190-200.
125-130. 274. Nowell, S., C. Sweeney, G. Hammons,
261. Miyamoto, M., Y Umetsu, H. Dosaka-Akita, FF Kadlubar, and N.P Lang (2002). CYP2A6
Y Sawamura, J. Yokota, H. Kunitoh et al. (1999). activity determined by caffeine phenotyping:
CYP2A6 gene deletion reduces susceptibility to Association with colorectal cancer risk. Cancer
lung cancer. Biochem. Biophys. Res. Commun. Epidemiol. Biomarkers Prev. 11, 377-383.
261, 658-660. 275. Le Gal, A., Y Dreano, PG. Gervasi, and F Berthou
262. London, S.J., J.R. Idle, A.K. Daly, and (2001). Human cytochrome P450 2A6 is the major
G.A. Coetzee (1999). Genetic variation of CYP2A6, enzyme involved in the metabolism of three
smoking, and risk of cancer. Lancet 353, 898-899. alkoxyethers used as oxyfuels. Toxicol. Lett. 124,
263. Schulz, T.G., P Ruhnau, and E. Hallier (2001). 47-58.
Lack of correlation between CYP2A6 genotype 276. Duescher, R.J. and A.A. Elfarra (1994). Human
and smoking habits. Adv. Exp. Med. Biol. 500, liver microsomes are efficient catalysts of 1,3-
213-215. butadiene oxidation: Evidence for major roles by
264. Raunio, H., A. Rautio, H. Gullsten, and O. cytochrome P450 2A6 and 2E1. Arch. Biochem.
Pelkonen (2001). Polymorphisms of CYP2A6 and Biophys. 311, 342-349.
its practical consequences. Br J. Clin. Pharmacol. 277. Nunoya, K., T. Yokoi, K. Kimura, K. Inoue,
52,357-363. T. Kodama, M. Funayama et al. (1998). A new
265. Tricker, A.R. (2003). Nicotine metabolism, human deleted allele in the human cytochrome P450 2A6
drug metabolism polymorphisms, and smoking (CYP2A6) gene found in individuals showing poor
behaviour. Toxicology 183, 151-173. metabolic capacity to coumarin and (+)-cis-3,5-
266. Loriot, M.A., S. Rebuissou, M. Oscarson, S. Cenee, dimethyl-2-(3-pyridyl)thiazolidin-4-one hydrochlo-
M. Miyamoto, N. Ariyoshi et al. (2001). Genetic ride (SM-12502). Pharmacogenetics 8, 239-249.
polymorphisms of cytochrome P450 2A6 in a case- 278. Nunoya, K., T. Yokoi, K. Kimura, T. Kainuma,
control study on lung cancer in a French popula- K. Satoh, M. Kinoshita et al. (1999). A new
tion. Pharmacogenetics 11, 3 9 ^ 4 . CYP2A6 gene deletion responsible for the in vivo
267. Zhang, X., K. Amemo, S. Ameno, K. Iw^ahashi, polymorphic metabolism of (+)-c^5-3,5-dimethyl-
H. Kinoshita, T. Kubota et al. (2001). Lack of 2-(3-pyridyl)thiazolidin-4-one hydrochloride in
association between smoking and CYP2A6 gene humans. J. Pharmacol. Exp. Ther 289, 437^42.
polymorphisms in a Japanese population. Nihon 279. Komatsu, T., H. Yamazaki, N. Shimada,
Arukoru Yakuhutsu Igakkai Zasshi 36, 486-490. M. Nakajima, and T. Yokoi (2000). Roles of
268. Oscarson, M. (2001). Genetic polymorphisms cytochromes P450 1A2, 2A6, and 2C8 in 5-
in the cytochrome P450 2A6 (CYP2A6) gene: fluorouracil formation from tegafiir, an anticancer
Implications for interindividual differences in nico- prodrug, in human liver microsomes. Drug Metab.
tine metabolism. Drug Metab. Dispos. 29, 91-95. Dispos. 28, 1457-1463.
269. Nakajima, M., Y Kuroiwa, and T. Yokoi (2002). 280. Ikeda, K., K. Yoshisue, E. Matsushima, S.
Interindividual differences in nicotine metabolism Nagayama, K. Kobayashi, C.A. Tyson et al. (2000).
and genetic polymorphisms of human CYP2A6. Bioactivation of tegafur to 5-fluorouracil is catalyzed
Drug Metab. Rev. 34, 865-877. by cytochrome P-450 2A6 in human liver micro-
270. Daly, A.K., S. Cholerton, W. Gregory, and J.R. Idle somes in vitro, Clin. Cancer Res. 6, 4409-4415.
(1993). Metabolic polymorphisms. Pharmacol. 281. Minoda, Y and E.D. Kharasch (2001). Halothane-
Ther 57, 129-160. dependent lipid peroxidation in human liver micro-
271. Cholerton, S., M.E. Idle, A. Vas, FJ. Gonzalez, and somes is catalyzed by cytochrome P4502A6
J.R. Idle (1992). Comparison of a novel thin-layer (CYP2A6). Anesthesiology 95, 509-514.
476 F. Peter Guengerich

282. Crespi, C.L., B.W. Penman, J.A. Leakey, 291. Messina, E.S., R.F Tyndale, and E.M. Sellers
M.P. Arlotto, A. Stark, A. Parkinson et al. (1990). (1997). A major role for CYP2A6 in nicotine
Human cytochrome P450IIA3:cDNA sequence, C-oxidation by human liver microsomes.
role of the enzyme in the metabolic activation of J. Pharmacol. Exp. Then 282, 1608-1614.
promutagens, comparison to nitrosamine activa- 292. Yamazaki, H., K. Inoue, M. Hashimoto, and
tion by human cytochrome P450IIE1. Carcino- T. Shimada (1999). Roles of CYP2A6 and
genesis 11, 1293-1300. CYP2B6 in nicotine C-oxidation by human liver
283. Yamazaki, H., Y. Inui, C.-H. Yun, M. Mimura, microsomes. Arch. Toxicol. 73, 65-70.
F.P Guengerich, and T. Shimada (1992). 293. Nakajima, M., T. Yamamoto, K. Nunoya, T. Yokoi,
Cytochrome P450 2E1 and 2A6 enzymes as major K. Nagashima, K. Inoue et al. (1996). Characteri-
catalysts for metabolic activation of A^-nitrosodi- zation of CYP2A6 involved in 3'-hydroxylation of
alkylamines and tobacco-related nitrosamines in cotinine in human liver microsomes. J. Pharmacol.
human liver microsomes. Carcinogenesis 13, Exp. Ther 111, 1010-1015.
1789-1794. 294. Hecht, S.S., J.B. Hochalter, PW Villalta, and
284. Smith, T.J., Z. Guo, F.J. Gonzalez, F.P Guengerich, S.E. Murphy (2000). 2'-Hydroxylation of nico-
G.D. Stoner, and C.S. Yang (1992). Metabolism of tine by cytochrome P450 2A6 and human
4-(methylnitrosamino)-1 -(3-pyridyl)-1 -butanone liver microsomes: Formation of a lung carcinogen
(NNK) in human lung and liver microsomes and precursor. Proc. Natl. Acad. Sci. USA 97,
cytochromes P-450 expressed in hepatoma cells. 12493-12497.
Cancer Res. 52, 1757-1763. 295. Su, T., Z.P Bao, Q.Y. Zhang, T.J. Smith, J.Y Hong,
285. Crespi, C.L., B.W. Penman, H.V. Gelboin, and and X.X. Ding (2000). Human cytochrome P450
F.J. Gonzalez (1991). A tobacco smoke-derived CYP2A13: Predominant expression in the res-
nitrosamine, 4-(methylnitrosamino)-1 -(3-pyridyl)- piratory tract and its high efficiency metabolic
1-butanone, is activated by multiple human activation of a tobacco-specific carcinogen, 4-
cytochrome P450s including the polymorphic (methylnitrosamino)-l-(3-pyridyl)-1-butanone.
human cytochrome P4502D6. Carcinogenesis 12, Cancer Res. 60, 5074-5079.
1197-1201. 296. Gillam, E.M.J., L.M. Notley, H. Cai, J.J. DeVoss,
286. Tiano, H.F, R.L. Wang, M. Hosokawa, C. Crespi, and F.P. Guengerich (2000). Oxidation of indole by
K.R. Tindall, and R. Langenbach (1994). Human cytochrome P450 enzymes. Biochemistry 39,
CYP2A6 activation of 4-(methylnitrosamino)-l- 13817-13824.
(3-pyridyl)-l-butanone (NNK): Mutational speci- 297. Nakamura, K., l.H. Hanna, H. Cai, Y Nishimura,
ficity in the gpt gene of AS52 cells. Carcinogenesis K.M. Williams, and FP Guengerich (2001).
15,2859-2866. Coumarin substrates for cytochrome P450 2D6
287. von Weymarn, L.B., N.D. Felicia, X. Ding, and fluorescence assays. Anal. Biochem. 292,280-286.
S.E. Murphy (1999). iV-Nitrosobenzylmethylamine 298. Lindberg, R.L.P and M. Negishi (1989). Alteration
hydroxylation and coumarin 7-hydroxylation: of mouse cytochrome P450^p,^ substrate specificity
Catalysis by rat esophageal microsomes and by mutation of a single amino-acid residue. Nature
cytochrome P450 2A3 and 2A6 enzymes. Chem. 339, 632-634.
Res. Toxicol. 12, 1254-1261. 299. Kitagawa, K., N. Kunugita, M. Kitagawa, and
288. Kushida, H., K. Fujita, A. Suzuki, M. Yamada, T. Kawamoto (2001). CYP2A6''6, a novel polymor-
T. Endo, T. Nohmi et al. (2000). Metabolic activation phism in cytochrome P450 2A6, has a single amino
of A^-alkylnitrosamines in genetically engineered acid substitution (R128Q) that inactivates enzy-
Salmonella typhimurium expressing CYP2E1 or matic activity^ Biol. Chem. 216, 17830-17835.
CYP2A6 together with human NADPH-cytochrome 300. Lewis, D.FV, M. Dickins, B.G. Lake,
P450 reductase. Carcinogenesis 2\, 1227-1232. PJ. Eddershaw, M.H. Tarbit, and PS. Goldfarb
289. Fujita, K. and T. Kamataki (2001). Predicting the (1999). Molecular modeling of the human cyto-
mutagenicity of tobacco-related A^-nitrosamines chrome P450 isoform CYP2A6 and investigations
in humans using 11 strains of Salmonella typhi- of CYP2A substrate selectivity. Toxicology 133,
murium YG7108, each coexpressing a form of 1-33.
human cytochrome P450 along with NADPH- 301. Lewis, D.F. (2002). Molecular modeling of human
cytochrome P450 reductase. Environ. Mol. cytochrome P450-substrate interactions. Drug
Mutagen. 38, 339-346. Metab. Rev. 34, 55-67.
290. Nakajima, M., T. Yamamoto, K. Nunoya, T. Yokoi, 302. Lewis, D.F (2002). Homology modelling of
K. Nagashima, K. Inoue et al. (1996). Role of human CYP2 family enzymes based on the
human cytochrome P4502A6 in C-oxidation of CYP2C5 crystal structure. Xenobiotica 32,
mcoXiwQ. Drug Metab. Dispos. 24, 1212-1217. 305-323.
Human P450s 477

303. Lewis, D.F. and J.W. Gorrod (2002). Molecular cytochrome P450 enzymes in drug abuse and
orbital calculations and nicotine metabolism: A dependence. Pharmacogenomics 3, 185-199.
rationale for experimentally observed metabolite 316. Tan, W, G.E Chen, D.Y. Xing, C.Y Song,
ratios. Drug Metabol. Drug Interact. 19, 29-39. EE Kadlubar, and D.X. Lin (2001). Frequency of
304. Guengerich, F.P., D.-H. Kim, and M. Iwasaki CYP2A6 gene deletion and its relation to risk of
(1991). Role of human cytochrome P-450 IIEl in lung and esophageal cancer in the Chinese popula-
the oxidation of many low molecular weight cancer tion. Int. J. Cancer 95, 96-101.
suspects. Chem. Res. Toxicol. 4, 168-179. 317. Satarug, S., M.A. Lang, P. Yongvanit, P.
305. Kharasch, E.D., D.C. Hankins, P.J. Baxter, and Sithithaworn, E. Mairiang, P. Mairiang et al.
K.E. Thummel (1998). Single-dose disulfiram (1996). Induction of cytochrome P450 2A6 expres-
does not inhibit CYP2A6 activity. Clin. Pharmacol. sion in humans by the carcinogenic parasite infec-
Then 64, 39-45. tion, opisthorchiasis viverrini. Cancer Epidemiol.
306. Murphy, S.E., L.M. Johnson, L.M. Losey, Biomarkers Prev. 5, 795-800.
S.G. Carmella, and S.S. Hecht (2001). Consumption 318. Pasanen, M., Z. Rannala, A. Tooming,
of watercress fails to alter coumarin metabolism in E.A. Sotaniemi, O. Pelkonen, and A. Rautio (1997).
humans. Drug Metab. Dispos. 29, 786-788. Hepatitis A impairs the function of human hepatic
307. Draper, A.J., A. Madan, and A. Parkinson (1997). CYP2A6 in vivo. Toxicology 123, 177-184.
Inhibition of coumarin 7-hydroxylase activity in 319. Koskela, S., J. Hakkola, J. Hukkanen, O. Pelkonen,
human liver microsomes. Arch. Biochem. Biophys. M. Sorri, A. Saranen et al. (1999). Expression
341,47-61. of CYP2A genes in human liver and extrahepatic
308. Zhang, W.J., T. Kilicarslan, R.E Tyndale, and tissues. Biochem. Pharmacol. 57, 1407-1413.
E.M. Sellers (2001). Evaluation of methoxsalen, 320. Oscarson, M., R.A. McLellan, H. GuUsten,
tranylcypromine, and tryptamine as specific and J.A.G. Agundez, J. Benitez, A. Rautio et al.
selective CYP2A6 inhibitors in vitro. Drug Metab. (1999). Identification and characterisation of
Dispos. 26, 897-902. novel polymorphisms in the CYP2A locus:
309. Taavitsainen, P., R. Juvonen, and O. Pelkonen Implications for nicotine metabolism. FEBS Lett.
(2001). In vitro inhibition of cytochrome P450 460, 321-327.
enzymes in human liver microsomes by a potent 321. Fernandez-Salguero, P, S.M.G. Hoffman, S.
CYP2A6 inhibitor. ^ra«^-2-phenylcyclopropy- Cholerton, H. Mohrenweiser, H. Raunio, A. Rautio
lamine (tranylcypromine), and its nonamine ana- et al. (1995). A genetic polymorphism in coumarin
log, cyclopropylbenzene. Drug Metab. Dispos. 29, 7-hydroxylation. Sequence of the human CYP2A
217-222. genes and identification of variant CYP2A 6 aWeles.
310. Koenigs, L.L., R.M. Peter, S.J. Thompson, Am. J. Hum. Genet. 57, 651-660.
A.E. Rettie, and W.F. Trager (1997). Mechanism- 322. Koskela, S., J. Hakkola, J. Hukkanen, O. Pelkonen,
based inactivation of human liver cytochrome M. Sorri, A. Saranen et al. (1999). Expression of
P450 2A6 by 8-methoxypsoralen. Drug Metab. CYP2A genes in human liver and extrahepatic
Dispos. 25, 1407-1415. tissues. Biochem. Pharmacol. 57, 1407-1413.
311. Sellers, E.M., H.L. Kaplan, and R.E Tyndale (2000). 323. Zhang, X., T. Su, Q.Y Zhang, J. Gu, M. Caggana,
Inhibition of cytochrome P450 2A6 increases nico- H. Li et al. (2002). Genetic polymorphisms of the
tine's oral bioavailability and decreases smoking. human CYP2AI3 gene: Identification of single-
Clin. Pharmacol. Then 68, 35-43. nucleotide polymorphisms and functional charac-
312. Koenigs, L.L. and W.F. Trager (1998). Mechanism- terization of an Arg257Cys variant. J. Pharmacol.
based inactivation of P450 2A6 by fiira- Exp. Ther 302, 416^23.
nocoumarins. Biochemistry 37, 10047-10061. 324. Von Weymarn, L.B. and S.E. Murphy (2003).
313. Khojasteh-Bakht, S.C, L.L. Koenigs, R.M. Peter, CYP2A13-catalysed coumarin metabolism: Com-
W.E Trager, and S.D. Nelson (1998). (i?)-(+)- parison with CYP2A5 and CYP2A6. Xenobiotica
Menthofuran is a potent, mechanism-based inacti- 33,73-81.
vator of human liver cytochrome P450 2A6. Drug 325. Mimura, M., T. Baba, Y Yamazaki, S. Ohmori,
Metab. Dispos. 26, 701-704. Y Inui, EJ. Gonzalez et al (1993). Characteri-
314. Wen, X., J.S. Wang, RJ. Neuvonen, and zation of cytochrome P450 2B6 in human liver
J.T Backman (2002). Isoniazid is a mechanism- microsomes. Drug Metab. Dispos. 21, 1048-1056.
based inhibitor of cytochrome P450 1A2, 2A6, 326. Hukkanen, J., A. Pelkonen, J. Hakkola, and
2C19 and 3A4 isoforms in human liver micro- H. Raunio (2002). Expression and regulation of
somes. Eur J. Clin. Pharmacol. 57, 799-804. xenobiotic-metabolizing cytochrome P450 (CYP)
315. Howard, L.A., E.M. Sellers, and R.E Tyndale enzymes in human lung. Crit. Rev. Toxicol. 32,
(2002). The role of pharmacogenetically-variable 391-411.
478 F. Peter Guengerich

327. van der Hoeven, T.A., D.A. Haugen, and MJ. Coon 338. Jover, R., R. Bort, M.J. Gomezlechon, and
(1974). Cytochrome P-450 purified to apparent J.V Castell (1998). Re-expression of C/EBPa
homogeneity from phenobarbital-induced rabbit Hver induces CYP2B6, CYP2C9 and CYP2D6 genes in
microsomes: Catalytic activity and other properties. HepG2 cells. FEBS Lett. 431, 227-230.
Biochem. Biophys. Res. Commun. 60, 569-575. 339. Goodwin, B., L.B. Moore, CM. Stoltz, D.D.
328. Imai, Y. and R. Sato (1974). A gel-electrophoretically McKee, and S.A. Kliewer (2001). Regulation of
homogeneous preparation of cytochrome P-450 from the human CYP2B6 gene by the nuclear pregnane
liver microsomes of phenobarbital-pretreated rabbits. X receptor. Mol. Pharmacol. 60, 427-431.
Biochem. Biophys. Res. Commun. 60, 8-14. 340. Willson, TM. and S.A. KHewer (2002). PXR,
329. Phillips, I.R., E.A. Shephard, A. Ashworth, and CAR and drug metabolism. Nat. Rev. Drug Discov.
B.R. Rabin (1985). Isolation and sequence of a 1,259-266.
human cytochrome P-450 cDNA clone. Proc. Natl. 341. Makinen, J., C. Frank, J. Jyrkkarinne, J. Gynther,
Acad. Sci. USA 82, 983-987. C. Carlberg, and P Honkakoski (2002).
330. Stresser, D.M. and D. Kupfer (1999). Mono- Modulation of mouse and human phenobarbital-
specific antipeptide antibody to cytochrome P-450 responsive enhancer module by nuclear receptors.
2B6. DrugMetah. Dispos. 11, 517-525. Mol. Pharmacol. 62, 366-378.
331. Code, E.L., C.L. Crespi, B.W. Penman, 342. Drocourt, L., J.C. Ourlin, J.M. Pascussi, P. Maurel,
F.J. Gonzalez, T.K.H. Chang, and D.J. Waxman and M.J. Vilarem (2002). Expression of CYP3A4,
(1997). Human cytochrome P4502B6. Interindivi- CYP2B6, and CYP2C9 is regulated by the vitamin
dual hepatic expression, substrate specificity, and D receptor pathway in primary human hepatocytes.
role in procarcinogen activation. Drug Metab. J. Biol. Chem. Ill, 25125-25132.
Dispos. 25, 985-993. 343. Chang, TK., S.M. Bandiera, and J. Chen (2003).
332. Ekins, S., M. VandenBranden, B.J. Ring, Constitutive androstane receptor and pregnane X
J.S. Gillespie, T.J. Yang, H.V Gelboin et al. (1998). receptor gene expression in human liver: Inter-
Further characterization of the expression in liver individual variability and correlation with CYP2B6
and catalytic activity of CYP2B6. J. Pharmacol. mRNA levels. Drug Metab. Dispos. 31, 7-10.
Exp. Ther 286, 1253-1259. 344. Maghch, J.M., D.J. Parks, L.B. Moore, J.L. Collins,
333. Imaoka, S., T. Yamada, T. Hiroi, K. Hayashi, B. Goodwin, A.N. Billin et al. (2003).
T. Sakaki, Y Yabusaki etal. (1996). Multiple forms Identification of a novel human CAR agonist and
of human P450 expressed in Saccharomyces its use in the identification of CAR target genes.
cerevisiae: Systematic characterization and com- J. Biol. Chem. 11%, 17277-17283.
parison with those of the rat. Biochem. Pharmacol. 345. Wang, H., S. Faucette, T. Sueyoshi, R. Moore,
51, 1041-1050. S. Ferguson, M. Negishi et al. (2003). A novel
334. Roy, P, L.J. Yu, C.L. Crespi, and D.J. Waxman distal enhancer module regulated by PXR/CAR
(1999). Development of a substrate-activity based is essential for the maximal induction of CYP2B6
approach to identify the major human liver P-450 gene expression, o^ 5/o/. Chem. 11%, 14146-14152.
catalysts of cyclophosphamide and ifosfamide 346. Miles, J.S., A.W McLaren, F.J. Gonzalez, and
activation based on cDNA-expressed activities and C.R. Wolf (1990). Alternative splicing in the
liver microsomal P-450 profiles. Drug Metab. human cytochrome P450IIB6 gene: Use of a cryp-
Dispos. 27, 655-666. tic exon within intron 3 and splice acceptor site
335. Gervot, L., B. Rochat, J.C. Gautier, within exon 4. Nucleic Acids Res. 18, 189.
F. Bohnenstengel, H. Kroemer, V de Berardinis 347. Zanger, U.M., J. Fischer, K. Klein, andT Lang (2002).
et al. (1999). Human CYP2B6: Expression, Detection of single nucleotide polymorphisms in
inducibility and catalytic activities. Pharmaco- CYP2B6 gene. Meth. Enzymol. 357, 45-53.
genetics 9, 295-306. 348. Lang, T, K. Klein, J. Fischer, A.K. Nussler,
336. Hanna, I.H., J.R. Reed, F.P. Guengerich, and P Neuhaus, U. Hofmann et al. (2001). Extensive
PF. Hollenberg (2000). Expression of human genetic polymorphism in the human CYP2B6 gene
cytochrome P450 2B6 in Escherichia coli: with impact on expression and fiinction in human
Characterization of catalytic activity and expres- liver. Pharmacogenetics 11, 399^15.
sion levels in human liver. Arch. Biochem. 349. Ariyoshi, N., M. Miyazaki, K. Toide, Y Sawamura,
Biophys. 316, 206-216. and T. Kamataki (2001). A single nucleotide
337. Sueyoshi, T., T. Kawamoto, 1. Zelko, polymorphism of CYP2B6 found in Japanese
P Honkakoski, and M. Neigishi (1999). The enhances catalytic activity by autoactivation.
repressed nuclear receptor CAR responds to Biochem. Biophys. Res. Commun. 281, 1256-1260.
phenobarbital in activating the human CYP2B6 350. Haugen, D.A. and M.J. Coon (1976). Properties
gene. J. Biol. Chem. 274, 6043-6046. of electrophoretically homogenous phenobarbital-
Human P450s 479

inducible and p-naphthoflavone-inducible forms 363. Lewis, D.FV, B.G. Lake, M. Dickins,
of liver microsomal cytochrome P-450. J. Biol. RJ. Eddershaw, M.H. Tarbit, and PS. Goldfarb
Chem. 251, 7929-7939. (1999). Molecular modelling of CYP2B6, the
351. Coon, M.J. (1981). The 1980 Bernard B. Brodie human CYP2B isoform, by homology with the sub-
Award lecture. Drug metabolism by cytochrome strate-bound CYP102 crystal structure: Evalua-
P-450: Progress and perspectives. Drug Metab. tion of CYP2B6 substrate characteristics, the
Dispos. 9, \~^. cytochrome b^ binding site and comparisons with
352. Guengerich, P.P. (1977). Separation and purifica- CYP2B1 and CYP2B4. Xenobiotica 29, 361-393.
tion of multiple forms of microsomal cytochrome 364. Lewis, D.F, S. Modi, and M. Dickins (2001).
P-450. Activities of different forms of cytochrome Quantitative structure-activity relationships
P-450 towards several compounds of environ- (QSARs) within substrates of human cytochromes
mental interest. J. Biol. Chem. 252, 3970-3979. P450 involved in drug metabolism. Drug Metabol.
353. Lu, A.Y.H. and S.B. West (1978). Reconstituted Drug Interact. 18, 221-242.
mammalian mixed-function oxidases: Requirements, 365. Bathelt, C , R.D. Schmid, and J. Pleiss (2002).
specificities and other properties. Pharmacol. Then Regioselectivity of CYP2B6: Homology model-
2, 337-338. ing, molecular dynamics simulation, docking.
354. Ekins, S. and S.A. Wrighton (1999). The role of J. Mol. Model. (Online) 8, 327-335.
CYP2B6 in human xenobiotic metabolism. Drug 366. Domanski, T.L., K.M. Schultz, F. Roussel,
Metab. Rev. 31, 719-754. J.C. Stevens, and J.R. Halpert (1999). Structure-
355. Chang, TK.H., G.F. Weber, C.L. Crespi, and function analysis of human cytochrome P-450 2B6
D.J. Waxman (1993). Differential activation of using a novel substrate, site-directed mutagenesis,
cyclophosphamide and ifosphamide by cyto- and molecular modeling. J. Pharmacol. Exp. Then
chromes P-450 2B and 3A in human liver micro- 290, 1141-1147.
somes. Cancer Res. 53, 5629-5637. 366a.Scott, E.E., YA. He, M.A. Wester, C.C. Chin, J.R.
356. Xie, H.J., U. Yasar, S. Lundgren, L. Griskevicius, Halpert, E.R Johnson, and CD. Stout (2003). An
Y Terelius, M. Hassan et al. (2003). Role of poly- open conformation of mammalian cytochrome
morphic human CYP2B6 in cyclophosphamide P450 2B4 at 1.6-A resolution. Proc. Natl. Acad.
bioactivation. Pharmacogenomics J. 3, 53-61. Sci. U.S.A. 100, 13121-13122.
357. Yanagihara, Y, S. Kariya, M. Ohtani, K. Uchino, 367. Guo, Z., S. Raeissi, R.B. White, and J.C. Stevens
T. Aoyama, Y Yamamura et al. (2001). Involve- (1997). Orphenadrine and methimazole inhibit
ment of CYP2B6 in A^-demethylation of ketamine multiple cytochrome P450 enzymes in human
in human liver microsomes. Drug Metab. Dispos. liver microsomes. Drug Metab. Dispos. 25,
29, 887-890. 390-393.
358. Court, M.H., S.X. Duan, L.M. Hesse, 368. Stiborova, M., L. Borek-Dohalska, P Hodek,
K. Venkatakrishnan, and D.J. Greenblatt (2001). J. Mraz, and E. Frei (2002). New selective
Cytochrome P-450 2B6 is responsible for inhibitors of cytochromes P450 2B and their
interindividual variability of propofol hydroxyla- application to antimutagenesis of tamoxifen. Arch.
tion by human liver microsomes. Anesthesiology Biochem. Biophys. 403, 4 1 ^ 9 .
94, 110-119. 369. Rae; J.M., N.V Soukhova, D.A. Flockhart, and
359. Heyn, H., R.B. White, and J.C. Stevens (1996). Z. Desta (2002). Triethylenethiophosphoramide
Catalytic role of cytochrome P4502B6 in the is a specific inhibitor of cytochrome P4502B6:
A^-demethylation of iS-mephenytoin. Drug Metab. Implications for cyclophosphamide metabolism.
Dispos. 24, 948-954. Drug Metab. Dispos. 30, 525-530.
360. Ko, J.W, Z. Desta, and D.A. Flockhart (1998). 370. Kent, U.M., D.E. Mills, R.V Rajnarayanan,
Human A^-demethylation of (5')-mephenytoin by W.L. Alworth, and PR HoUenberg (2002). Effect of
cytochrome P450s 2C9 and 2B6. Drug Metab. 17a-ethynylestradiol on activities of cytochrome
Dispos. 26,115-11^. P450 2B (P450 2B) enzymes: Characterization of
361. Ekins, S., M. VandenBranden, B.J. Ring, and inactivation of P450s 2B1 and 2B6 and identifica-
S.A. Wrighton (1997). Examination of purported tion of metabolites. J. Pharmacol. Exp. Then 300,
probes of human CYP2B6. Pharmacogenetics 7, 549-558.
165-179. 371. Kiipfer, A. and R. Preisig (1984). Pharmacogenetics
362. Ariyoshi, N., K. Oguri, N. Koga, H. Yoshimura, of mephenytoin: A new drug hydroxylation poly-
andY Funae (1995). MetaboUsm of highly persist- morphism in man. Eur J. Clin. Pharmacol. 26,
ent PCB congener, 2, 4,5, 2',4',5'-hexachloro- 753-759.
biphenyl, by human CYP2B6. Biochem. Biophys. 372. Wedlund, P I , W.S. Aslanian, C.B. McAllister,
Res. Commun. Ill, 455-460. G.R. Wilkinson, and R.A. Branch (1984).
480 F. Peter Guengerich

Mephenytoin hydroxylation deficiency in 383. Gerbal-Chaloin, S., J.M. Pascussi, L. Pichard-


Caucasians: Frequency of a new oxidative drug Garcia, M. Daujat, F. Waechter, J.M. Fabre et al.
metabolism polymorphism. Clin. Pharmacol. Ther. (2001). Induction of CYP2C genes in human hepa-
36, 773-780. tocytes in primary culture. Drug Metab. Dispos.
373. Knodell, R.G., S.D. Hall, G.R. Wilkinson, and 29,242-251.
F.P. Guengerich (1987). Hepatic metabolism 384. Soyama, A., Y. Saito, N. Hanioka, N. Murayama,
of tolbutamide: Characterization of the form of O. Nakajima, N. Katori et al. (2001). Non-
cytochrome P-450 involved in methyl hydro- synonymous single nucleotide alterations found in
xylation and relationship to in vivo disposition. the CYP2C8 gene result in reduced in vitro pacli-
J. Pharmacol. Exp. Ther 241, 1112-1119. taxel metabolism. Biol. Pharm. Bull. 24,1427-1430.
374. Brian, W.R., RK. Srivastava, D.R. Umbenhauer, 385. Rahman, A., K.R. Korzekwa, J. Grogan, F.J.
R.S. Lloyd, and F.R Guengerich (1989). Expression Gonzalez, and J.W. Harris (1994). Selective bio-
of a human liver cytochrome P-450 protein with transformation of taxol to 6a-hydroxytaxol by
tolbutamide hydroxylase activity in Saccharomyces human cytochrome P450 2C8. Cancer Res. 54,
cerevisiae. Biochemistry 28, 4993^999. 5543-5546.
375. Ged, C, D.R. Umbenhauer, T.M. Bellew, 386. Leo, M.A., J.M. Lasker, J.L. Raucy, C.I. Kim,
R.W. Bork, PK. Srivastava, N. Shinriki et al. M. Black, and C.S. Lieber (1989). Metabolism
(1988). Characterization of cDNAs, mRNAs, and of retinol and retinoic acid by human liver
proteins related to human liver microsomal cytochrome P450IIC8. Arch. Biochem. Biophys.
cytochrome P-450 (iS')-mephenytoin 4'-hydroxy- 269,305-312.
lase. Biochemistry 27, 6929-6940. 387. Yamazaki, H., A. Shibata, M. Suzuki, M. Nakajima,
376. Romkes, M., M.B. Faletto, J.A. Blaisdell, N. Shimada, FP Guengerich et al (1999). Oxi-
J.L. Raucy, and J.A. Goldstein (1991). Cloning and dation of troglitazone to a quinone-type metabolite
expression of complementary DNAs for multiple catalyzed by cytochrome P450 2C8 and 3A4 in
members of the human cytochrome P450I1C human liver microsomes. Drug Metab. Dispos. 27,
subfamily. Biochemistry 30, 3247-3255. 1260-1266.
377. Wrighton, S.A., J.C. Stevens, G.W. Becker, and 388. Marques-Soares, C , S. Dijols, A.-C. Macherey,
M. VandenBranden (1993). Isolation and charac- M.R. Wester, E.F. Johnson, PM. Dansette et al.
terization of human liver cytochrome P450 2C19: (2003). Sulfaphenazole derivatives as tools for
Correlation between 2C19 and 5-mephenytoin comparing cytochrome P450 2C5 and human
4'-hydroxylation. Arch. Biochem. Biophys. 306, cytochrome P450 2Cs: Identification of a new high
240-245. affinity substrate common to those CYP 2C
378. Goldstein, J.A., M.B. Faletto, M. Romkes-Sparks, enzymes. Biochemistry 42, 6363-6369.
T. Sullivan, S. Kitareewan, J.L. Raucy et al. 389. Wester, M.R., E.F. Johnson, C. Marques-Soares,
(1994). Evidence that CYP2C19 is the major P Dansette, D. Mansuy, and CD. Stout (2003). The
(»S)-mephenytoin 4'-hydroxylase in humans. structure of a substrate complex of mammalian
Biochemistry ^:^, 1743-1752. cytochrome P450 2C5 at 2.3 A resolution:
379. Nelson, D.R., L. Koymans, T. Kamataki, Evidence for multiple substrate binding modes.
J.J. Stegeman, R. Feyereisen, D.J. Waxman Biochemistry 42, 9335-9345.
et al. (1996). P450 superfamily: Update on new 389a.Schoch, G.A., J.K. Yano, M.R. Wester, K.J. Griffin,
sequences, gene mapping, accession numbers, and CD. Stout, and E.F Johnson (2004). Structure
nomenclature. Pharmacogenetics 6, 1-42. of human microsomal cytochrome P450 2C8.
380. Dai, D., D.C. Zeldin, J.A. Blaisdell, B. Chanas, Evidence for a peripheral fatty acid binding site. J.
S.J. Coulter, B.I. Ghanayem et al. (2001). Biol. Chem. 279, 9497-9503.
Polymorphisms in human CYP2C8 decrease 390. Ha-Duong, N.T., S. Dijols, C Marques-Soares,
metabolism of the anticancer drug paclitaxel and C Minoletti, PM. Dansette, and D. Mansuy (2001).
arachidonic acid. Pharmacogenetics 11, 597-607. Synthesis of sulfaphenazole derivatives and their
381. Klose, T.S., J.A. Blaisdell, and J.A. Goldstein use as inhibitors and tools for comparing the active
(1999). Gene structure of CYP2C8 and extra- sites of human liver cytochromes P450 of the 2C
hepatic distribution of the human CYP2Cs. subfamily J. Med. Chem. 44, 3622-3631.
J. Biochem. Mol. Toxicol. 13, 289-295. 391. Ha-Duong, N.T., C Marques-Soares, S. Dijols,
382. Bahadur, N., J.B. Leathart, E. Mutch, D. Steimel- M.A. Sari, PM. Dansette, and D. Mansuy (2001).
Crespi, S.A. Dunn, R. Gilissen et al. (2002). Interaction of new sulfaphenazole derivatives with
CYP2C8 polymorphisms in Caucasians and human liver cytochrome P4502Cs: Structural
their relationship with paclitaxel 6a-hydroxylase determinants required for selective recognition by
activity in human liver microsomes. Biochem. CYP2C9 and for inhibition of human CYP2Cs.
Pharmacol. 64, 1579-1589. Arch. Biochem. Biophys. 394, 189-200.
Human P450s 481

392. Zilly, W., D.D. Breimer, and E. Richter (1975). CYP2C9 by the constitutive androstane receptor:
Induction of drug metabolism in man after Discovery of a new distal binding site. Mol.
rifampicin treatment measured by increased hexo- Pharmacol. 62, 737-746.
barbital and tolbutamide clearance. Eur. J. Clin. 404. Pascussi, J.M., S. Gerbal-Chaloin, L. Drocourt,
Pharmacol. 9,219-227. P. Maurel, and M.J. Vilarem (2003). The expression
393. Umbenhauer, D.R., M.V. Martin, R.S. Lloyd, and of CYP2B6, CYP2C9 and CYP3A4 genes: A tangle
F.P. Guengerich (1987). Cloning and sequence of networks of nuclear and steroid receptors.
determination of a complementary DNA related Biochim. Biophys. Acta 1619, 243-253.
to human liver microsomal cytochrome P-450 405. Ibeanu, G.C. and J.A. Goldstein (1995). Transcrip-
iS-mephenytoin 4-hydroxylase. Biochemistry 26, tional regulation of human CYP2C genes:
1094-1099. Functional comparison of CYP2C9 and CYP2C18
394. Knodell, R.G., R.K. Dubey, G.R. Wilkinson, and promoter regions. Biochemistry 34, 8028-8036.
F.R Guengerich (1988). Oxidative metabolism 406. Scott, J. and PL. Poffenbarger (1978). Pharmaco-
of hexobarbital in human liver: Relationship to genetics of tolbutamide metabolism in humans.
polymorphic 5-mephenytoin 4-hydroxylation. Diabetes 2^, A\-5\.
J. Pharmacol. Exp. Then 245, 845-849. 407. Ohgiya, S., M. Komori, H. Ohi, K. Shiramatsu,
395. Yasumori, T., N. Murayama, Y. Yamazoe, A. Abe, N. Shinriki, and T. Kamataki (1992). Six-base
Y. Nogi, T. Fukasawa et al. (1989). Expression deletion occurring in messages of human cyto-
of a human P-450IIC gene in yeast cells using chrome P-450 in the CYP2C subfamily results in
galactose-inducible expression system. Mol reduction of tolbutamide hydroxylase activity.
Pharmacol. 35, 443-449. Biochem. Int. 27, 1073-1081.
396. Srivastava, RK., C.-H. Yun, RH. Beaune, C. Ged, 408. Goldstein, J.A. and S.M.F Demorais (1994).
and RR Guengerich (1991). Separation of Biochemistry and molecular biology of the human
human liver tolbutamine hydroxylase and (S)- CYP2C subfamily. Pharmacogenetics 4, 285-299.
mephenytoin 4'-hydroxylase cytochrome P-450 409. Inoue, K., H. Yamazaki, K. Imiya, S. Akasaka,
enzymes. Mol. Pharmacol. 40, 69-79. KR Guengerich, andT. Shimada (1997). Relation-
397. Treluyer, J.M., G. Gueret, G. Cheron, M. Sonnier, ship between CYP2C9 and CYP2C19 genotypes
and T. Cresteil (1997). Developmental expres- and tolbutamide methyl hydroxylation and
sion of CYP2C and CYP2C-dependent activities in iS'-mephenytoin 4'-hydroxylation in livers of
the human liver: in-vivo/in-vitro correlation and Japanese and Caucasian populations. Pharma-
inducibility. Pharmacogenetics 7, 441-452. cogenetics 7, 103-113.
398. Brenner, S.S., C. Herrlinger, K. Dilger, 410. Lee, C.R., J.A. Goldstein, and J.A. Pieper (2002).
T.E. Murdter, U. Hoftnann, C. Marx et al (2003). Cytochrome P450 2C9 polymorphisms: A compre-
Influence of age and cytochrome P450 2C9 geno- hensive review of the in-vitro and human data.
type on the steady-state disposition of diclofenac Pharmacogenetics 12, 251-263.
and celecoxib. Clin. Pharmacokinet. 42, 283-292. 411. Xie, H.G., H.C. Prasad, R.B. Kim, and CM. Stein
399. Obach, R.S., Q.Y. Zhang, D. Dunbar, and (2002). CYP2C9 allelic variants: Ethnic distribu-
L.S. Kaminsky (2001). Metabolic characterization tion and functional significance. Adv. Drug Deliv.
of the major human small intestinal cytochrome Rev. 54, 1257-1270.
P450s. DrugMetab. Dispos. 29, 347-352. 412. Dickmann, L.J., A.E. Rettie, M.B. Kneller,
400. Morel, R, RH. Beaune, D. Ratanasavanh, R.B. Kim, A.J. Wood, CM. Stein et al (2001).
J.-P. Flinois, C.-S. Yang, F.P. Guengerich et al. Identification and functional characterization of a
(1990). Expression of cytochrome P-450 enzymes new CYP2C9 variant (CYP2C9*5) expressed among
in cultured human hepatocytes. Eur J. Biochem. African Americans. Mol Pharmacol 60, 382-387.
191,AZl-A^A. 413. Shintani, M., I. leiri, K. Inoue, K. Mamiya,
401. Raucy, XL., L. Mueller, K. Duan, S.W. Allen, H. Ninomiya, N. Tashiro et al (2001). Genetic
S. Strom, and J.M. Lasker (2002). Expression and polymorphisms and functional characterization of
induction of CYP2C P450 enzymes in primary the 5'-flanking region of the human CYP2C9
cultures of human hepatocytes. J. Pharmacol. Exp. gene: In vitro and in vivo studies. Clin. Pharmacol.
Then 302, 475-482. Then 70, 175-182.
402. Gerbal-Chaloin, S., M. Daujat, J.M. Pascussi, 414. Warner, S.C, C Finta, and RG. Zaphiropoulos
L. Pichard-Garcia, M.J. Vilarem, and P. Maurel (2001). Intergenic transcripts containing a novel
(2002). Transcriptional regulation of CYP2C9 gene. human C5^ochrome P450 2C exon 1 spliced to
Role of glucocorticoid receptor and constitutive sequences from the CYP2C9 gene. Mol Biol Evol
androstane receptor. J. Biol. Chem. 211, 209-217. 18, 1841-1848.
403. Ferguson, S.S., E.L. LeCluyse, M. Negishi, and 415. Yasar, U, S. Lundgren, E. Eliasson, A. Bennet,
J.A. Goldstein (2002). Regulation of human B. Wiman, U. de Faire et al (2002). Linkage
482 F. Peter Guengerich

between the CYP2C8 and CYP2C9 genetic 426. Backes, W.L., C.J. Batie, and G.E Cawley (1998).
polymorphisms. Biochem. Biophys. Res. Commun. Interactions among P450 enzymes when combined
299, 25-28. in reconstituted systems: Formation of a 2B4-1A2
416. Miners, J.O. and D.J. Birkett (1998). Cytochrome complex with a high affinity for NADPH cytochrome
P4502C9: An enzyme of major importance in P450 reductase. Biochemistry 37, 12852-12859.
human drug metabolism. Br. J. Clin. Pharmacol. 427. Hutzler, J.M., D. Kolwankar, M.A. Hummel, and
45, 525-538. TS. Tracy (2002). Activation of CYP2C9-
417. Giancarlo, G.M., K. Venkatakrishnan, B.W. Granda, mediated metabolism by a series of dapsone
L.L. von Moltke, and D.J. Greenblatt (2001). analogs: Kinetics and structural requirements.
Relative contributions of CYP2C9 and 2C19 to Drug Metab. Dispos. 30, 1194-1200.
phenytoin 4-hydroxylation in vitro: Inhibition by 428. Hutzler, J.M., L.C. Wienkers, J.L. Wahlstrom,
sulfaphenazole, omeprazole, and ticlopidine. Eur J. T.J. Carlson, and TS. Tracy (2003). Activation of
Clin. Pharmacol. 57, 31-36. cytochrome P450 2C9-mediated metabolism:
418. Draper, A.J. and B.D. Hammock (2000). Mechanistic evidence in support of kinetic obser-
Identification of CYP2C9 as a human liver micro- vations. Arch. Biochem. Biophys. 410, 16-24.
somal linoleic acid epoxygenase. Arch. Biochem. 429. Ueng, Y-E, T Kuwabara, Y.-J. Chun, and
Biophys. 376, 199-205. EP Guengerich (1997). Cooperativity in oxidations
419. McSorley, L.C. and A.K. Daly (2000). Identification catalyzed by cytochrome P450 3A4. Biochemistry
of human cytochrome P450 isoforms that contribute 36,370-381.
to all-/ra«^-retinoic acid 4-hydroxylation. Biochem. 430. Takanashi, K., H. Tainaka, K. Kobayashi,
Pharmacol. 60, 517-526. T Yasumori, M. Hosakawa, and K. Chiba (2000).
420. Lee, C.R., J.A. Pieper, R.F. Frye, A.L. CYP2C9 Ile^^^ and Leu^^^ variants: Enzyme kinetic
Hinderliter, J.A. Blaisdell, and J.A. Goldstein study with seven substrates. Pharmacogenetics 10,
(2003). Tolbutamide, flurbiprofen, and losartan as 95-104.
probes of CYP2C9 activity in humans. J. Clin. 431. Yasar, U, E. Eliasson, C. Forslund-Bergengren,
Pharmacol. 43,84-91. G. Tybring, M. Gadd, E Sjoqvist et al. (2001). The
421. Sandberg, M., U. Yasar, P Stromberg, J.O. Hoog, and role of CYP2C9 genotype in the metabolism of
E. Eliasson (2002). Oxidation of celecoxib by poly- diclofenac in vivo and in vitro. Eur J. Clin.
morphic cytochrome P450 2C9 and alcohol dehy- Pharmacol. 57, 729-735.
drogenase. Br J. Clin. Pharmacol. 54, 423-429. 432. Ridderstrom, M., C. Masimirembwa, S. Trump-
422. Tang, C , M. Shou, T.H. Rushmore, Q. Mei, Kallmeyer, M. Ahlefelt, C. Otter, andTB. Andersson
P Sandhu, E.J. WooXf etal (2001). In-vitro metab- (2000). Arginines 97 and 108 in CYP2C9 are impor-
olism of celecoxib, a cyclooxygenase-2 inhibitor, tant determinants of the catalyticfiinction.Biochem.
by allelic variant forms of human liver microsomal Biophys. Res. Commun. 270, 983-987.
cytochrome P450 2C9: Correlation with CYP2C9 433. Flanagan, J.U., L.A. McLaughlin, M.J. Paine,
genotype and in-vivo pharmacokinetics. M.J. Sutclifife, G.C. Roberts, and C.R. Wolf
Pharmacogenetics 11, 223-235. (2003). Role of conserved Asp^^^ of cytochrome
423. Tang, C.Y, M.G. Shou, and A.D. Rodrigues P450 2C9 in substrate recognition and catalytic
(2000). Substrate-dependent effect of acetonitrile activity Biochem. J. 370, 921-926.
on human liver microsomal cytochrome P4502C9 434. He, M., K.R. Korzekwa, J.P Jones, A.E. Rettie, and
(CYP2C9) activity Drug Metab. Dispos. 28, WF. Trager (1999). Structural forms of phenpro-
567-572. coumon and warfarin that are metabolized at the
424. Yamazaki, H., E.M.J. Gillam, M.-S. Dong, active site of CYP2C9, Arch. Biochem. Biophys.
WW. Johnson, EP Guengerich, and T Shimada 372, 16-28.
(1997). Reconstitution of recombinant human 435. Mancy, A., S. Dijols, S. Poli, EP. Guengerich, and
cytochrome P450 2C9 and comparison with D. Mansuy (1997). Interaction of sulfaphenazole
cytochrome P450 and other forms: Effects of derivatives with human liver cytochromes P450 2C:
cytochrome P450:P450 and cytochrome ?450:b^ Molecular origin of the specific inhibitory effects
interactions. Arch. Biochem. Biophys. 342,329-337. of sulfaphenazole on CYP 2C9 and consequences
425. Yamazaki, H., T. Komatsu, K. Ohyama, M. for the sub-strate binding topology. Biochemistry
Nakamura, S. Asahi, N. Shimada et al. (2002). 35, 16205-16212.
Roles of NADPH-P450 reductase and apo- and 436. Mancy A., P Broto, S. Dijols, PM. Dansette, and
holo-cytochrome b^ on xenobiotic oxidations cat- D. Mansuy (1995). The substrate binding site of
alyzed by 12 recombinant human cytochrome human liver cytochrome P450 2C9: An approach
P450s expressed in membranes of Escherichia using designed tienilic acid derivatives and molec-
coli. Protein Expr Purif. 24, 329-337. ular modeling. Biochemistry 34, 10365-10375.
Human P450s 483

437. Tsao, C.C., M.R. Wester, B. Ghanayem, 447. Veronese, M.E., J.O. Miners, D. Randies,
S.J. Coulter, B. Chanas, E.F. Johnson et al. (2001). D. Gregov, and D.J. Birkett (1990). Validation of
Identification of human CYP2C19 residues that the tolbutamide metabolic ratio for population
confer .S'-mephenytoin 4'-hydroxylation activity to screening with use of sulfaphenazole to produce
CYP2C9. Biochemistry 40, 1937-1944. model phenotypic poor metabolizers. Clin.
438. Jung, E, K.J. Griffin, T.H. Richardson, M. Yang, Pharmacol. Ther 47, 403^11.
and E.F. Johnson (1998). Identification of amino 448. Wen, X., J.S. Wang, J.T. Backman, J. Laitila, and
acid substitutions that confer a high affinity for P.J. Neuvonen (2002). Trimethoprim and sul-
sulphaphenazole binding and a high catalytic effi- famethoxazole are selective inhibitors of CYP2C8
ciency for warfarin metabolism to P450 2C19. and CYP2C9, respectively. Drug Metab. Dispos.
Biochemistry:^!, 16270-16279. 30,631-635.
439. Niwa, T., A. Kageyama, K. Kishimoto, 449. Wen, X., J.S. Wang, K.T Kivisto, PJ. Neuvonen,
Y. Yabusaki, R Ishibashi, and M. Katagiri (2002). and J.T. Backman (2001). In vitro evaluation of
Amino acid residues affecting the activities of valproic acid as an inhibitor of human cytochrome
human cytochrome P450 2C9 and 2C19. Drug P450 isoforms: Preferential inhibition of
Metab. Dispos. 30, 931-936. cytochrome P450 2C9 (CYP2C9). Br J. Clin.
440. Melet, A., N. Assrir, P. Jean, M. Pilar Lopez- Pharmacol. 52, 547-553.
Garcia, C. Marques-Soares, M. Jaouen etal. (2003). 450. Zhang, Z.Y., J. Kerr, R.S. Wexler, H.Y. Li,
Substrate selectivity of human cytochrome P450 A.J. Robinson, PR Harlow et al. (1997). Warfarin
2C9: Importance of residues 476, 365, and 114 in analog inhibition of human CYP2C9-catalyzed S-
recognition of diclofenac and sulfaphenazole and in warfarin 7-hydroxylation. Thromb. Res. 88,
mechanism-based inactivation by tienilic acid. Arch. 389-398.
Biochem. Biophys. 409, 80-91. 451. Beaune, P, PM. Dansette, D. Mansuy, L. Kiffel,
441. Guengerich, EP, I.H. Hanna, M.V Martin, and M. Finck, C Amar et al. (1987). Human anti-
E.M.J. Gillam (2003). Role of glutamic acid 216 endoplasmic reticulum autoantibodies appearing
in cytochrome P450 2D6 substrate binding and in a drug-induced hepatitis are directed against a
catalysis. Biochemistry 42, 1245-1253. human liver cytochrome P-450 that hydroxylates
442. Lewis, D.EV, M. Dickins, R.J. Weaver, the drug. Proc. Natl. Acad Sci. USA 84, 551-555.
PJ. Eddershaw, PS. Goldfarb, and M.H. Tarbit 452. Dansette, PM., D.C Thang, H. El Amri, and
(1998). Molecular modelling of human CYP2C D. Mansuy (1992). Evidence for thiophene-S-
subfamily enzymes CYP2C9 and CYP2C19: oxide as a primary reactive metabolite of thio-
Rationalization of substrate specificity and site- phene in vivo: Formation of a dihydrothiophene
directed mutagensis experiments in the CYP2C sulfoxide mercapturic acid. Biochem. Biophys.
subfamily. Xenobiotica 28, 235-268. Res. Commun. 186, 1624-1630.
443. Afzelius, L., I. Zamora, M. Ridderstrom, 453. Beaune, P., D. Pessayre, P. Dansette, D. Mansuy,
T.B. Andersson, A. Karlen, and CM. Masimirembwa and M. Manns (1994). Autoantibodies against
(2001). Competitive CYP2C9 inhibitors: Enzyme C3^ochromes P450: Role in human diseases. Adv.
inhibition studies, protein homology modeling, and Pharmacol. 30, 199-245.
three-dimensional quantitative structure-activity 454. Carlile, D.J., N. Hakooz, M.K. BayUss, and
relationship analysis. Mol. Pharmacol. 59, 909-919. J.B. Houston (1999). Microsomal prediction of
444. Ridderstr5m, M., I. Zamora, O. Fjellstrom, and in vivo clearance of CYP2C9 substrates in humans.
T.B. Andersson (2001). Analysis of selective regions Br J. Clin. Pharmacol. 47, 625-635.
in the active sites of human cytochromes P450,2C8, 455. Goldstein, J.A. (2001). Clinical relevance of
2C9, 2C18, and 2C19 homology models using genetic polymorphisms in the human CYP2C
GRID/CPCA. J. Med. Chem. 44, 4072^081. subfamily. Br J. Clin. Pharmacol. 52, 349-355.
445. de Groot, M.J., A.A. Alex, and B.C. Jones (2002). 456. Kaminsky, L.S. and Z.Y. Zhang (1997). Human
Development of a combined protein and pharma- P450 metabolism of warfarin. Pharmacol. Ther
cophore model for cytochrome P450 2C9. J. Med. 73, 67-74.
Chem. 45, 1983-1993. 457. Yamazaki, H. and T Shimada (1997). Human
445a.Williams, PA., J. Cosme, A. Ward, H.C. Angove, liver cytochrome P450 enzymes involved in the
D. Matak Vinkovic, and H. Jhoti (2003). Crystal 7-hydroxylation of ^- and »S-warfarin enantiomers.
structure of human cytochrome P450 2C9 with Biochem. Pharmacol. 54, 1195-1203.
bound warfarin. Nature 424, 464^68. 458. Kunze, K.L., A.C Eddy, M. Gibaldi, and
446. Wester, M.R., CD. Stout, G. Schoch, J.K. Yano, W.F. Trager (1991). Metabolic enantiomeric inter-
and E.F. Johnson (2003). Crystallization of human actions: The inhibition of human (S)-warfarin-7-
P450 2C9. FASEB J. 17, A609. hydroxylase by (R)-warfarin. Chirality 3, 24-29.
484 F. Peter Guengerich

459. Rettie, A.E., K.R. Korzekwa, K.L. Kunze, Investigation vs Atenolol (SILVHIA) trial.
R.F. Lawrence, A.C. Eddy, T. Aoyama et al. (1992). J. Hypertens. 20, 2089-2093.
Hydroxylation of warfarin by human cDNA- 470. Yun, C.-H., H.S. Lee, H. Lee, J.K. Rho,
expressed cytochrome P-450: A role for P-4502C9 H.G. Jeong, and F.P Guengerich (1995). Oxidation of
in the etiology of (5)-warfarin-drug interactions. the angiotensin II receptor antagonist losartan (DuP
Chem. Res. Toxicol. 5, 54-59. 753) in human liver microsomes: Role of cytochrome
460. Rettie, A.E., L.C. Wienkers, F.J. Gonzalez, P450 3A(4) in formation of the active metabolite
W.F. Trager, and K.R. Korzekwa (1994). Impaired EXP 3174. DrugMetab. Dispos. 23, 285-289.
(iS)-warfarin metabolism catalysed by the R144C 471. Stearns, R.A., PK. Chakravarty, R. Chen, and
allelic variant of CYP2C9. Pharmacogenetics 4, S.H.L. Chiu (1995). Biotransformation of losartan
39^2. to its active carboxylic acid metabolite in human
461. Yamazaki, H., K. Inoue, and T. Shimada (1998). liver microsomes: Role of cytochrome P4502C
Roles of two allelic variants (Argl44Cys and and 3A subfamily members. Drug Metab. Dispos.
Ile359Leu) of cytochrome P4502C9 in the oxida- 23,207-215.
tion of tolbutamide and warfarin by human liver 472. Aithal, G.P, C.P Day, J.B. Leathart, and A.K. Daly
microsomes. Xenobiotica 28, 103-115. (2000). Relationship of polymorphism in CYP2C9
462. Scordo, M.G., V Pengo, E. Spina, M.L. Dahl, to genetic susceptibility to diclofenac-induced
M. Gusella, and R. Padrini (2002). Influence of hepatitis. Pharmacogenetics 10, 511-518.
CYP2C9 and CYP2C19 genetic polymorphisms 473. Martinez, C , E. Garcia-Martin, J.M. Ladero,
on warfarin maintenance dose and metabolic J. Sastre, F. Garcia-Gamito, M. Diaz-Rubio et al.
clearance. Clin. Pharmacol. Ther 72, 702-710. (2001). Association of CYP2C9 genotypes leading
463. Higashi, M.K., D.L. Veenstra, L.M. Kondo, to high enzyme activity and colorectal cancer risk.
A.K. Wittkowsky, S.L. Srinouanprachanh, Carcinogenesis 12, 1323-1326.
F.M. Farin et al. (2002). Association between 474. Yasar, U, E. Eliasson, and M.L. Dahl (2002).
CYP2C9 genetic variants and anticoagulation- Association of CYP2C9 genotypes leading to
related outcomes during warfarin therapy. J. Am. high enzyme activity and colorectal cancer risk.
Med. Assoc. 287, 1690-1698. Carcinogenesis 23, 665; author reply 667-668.
464. Tassies, D., C. Freire, J. Pijoan, S. Maragall, 475. Garcia-Martin, E., C. Martinez, J.M. Ladero,
J. Monteagudo, A. Ordinas et al. (2002). Pharma- F.J. Gamito, A. Rodriguez-Lescure, and
cogenetics of acenocoumarol: Cytochrome P450 J.A. Agundez (2002). Influence of cytochrome
CYP2C9 polymorphisms influence dose require- P450 CYP2C9 genotypes in lung cancer risk.
ments and stability of anticoagulation. Haemato- Cancer Lett. 180,41^6.
logicaSl, 1185-1191. 476. Minoletti, C , S. Dijols, PM. Dansette, and
465. Gentry, PR., C.E. Hack, L. Haber, A. Maier, and D. Mansuy (1999). Comparison of the substrate
H.J. Clewell, III (2002). An approach for the quan- specificities of human liver cytochrome P450s 2C9
titative consideration of genetic polymorphism and 2C18: Apphcation to the design of a specific
data in chemical risk assessment: Examples with substrate of CYP 2C18. Biochemistry 38,
warfarin and parathion. Toxicol. Sci. 70, 120-139. 7828-7836.
466. Lee, C.R., J.A. Pieper, A.L. Hinderliter, 477. Richardson, T.H., K.J. Griffin, F Jung, J.L. Raucy,
J.A. Blaisdell, and J.A. Goldstein (2002). Evaluation and E.F. Johnson (1997). Targeted antipeptide anti-
of cytochrome P4502C9 metabolic activity with bodies to cytochrome P450 2C18 based on epitope
tolbutamide in CYP2C91 heterozygotes. Clin. mapping of an inhibitory monoclonal antibody to
Pharmacol. Ther 72, 562-571. P450 2C5. Arch. Biochem. Biophys. 338, 157-164.
467. Shon, J.H., Y.R. Yoon, K.A. Kim, YC. Lim, 478. Zaphiropoulos, PG. (1999). RNA molecules con-
K.J. Lee, J.Y Park et al (2002). Effects of CYP2C19 taining exons originating from different members
and CYP2C9 genetic polymorphisms on the disposi- of the cytochrome P450 2C gene subfamily
tion of and blood glucose lowering response to tolbu- (CYP2C) in human epidermis and liver. Nucleic
tamide in humans. Pharmacogenetics 12, 111-119. Acids Res. 27, 2585-2590.
468. Miners, J. (2002). CYP2C9 polymorphism: Impact 479. Mace, K., E.D. Bowman, P Vautravers, PG. Shields,
on tolbutamide pharmacokinetics and response. C.C. Harris, and A.M. Pfeifer (1998). Characteri-
Pharmacogenetics 12, 91-92. sation of xenobiotic-metabolising enzyme expres-
469. Hallberg, R, J. Karlsson, L. Kurland, L. Lind, sion in human bronchial mucosa and peripheral lung
T. Kahan, K. Malmqvist et al (2002). The tissues. Eur J. Cancer 34, 914-920.
CYP2C9 genotype predicts the blood pressure 480. Mizugaki, M., M. Hiratsuka, Y Agatsuma,
response to irbesartan: Results from the Y Matsubara, K. Fujii, S. Kure et al (2000). Rapid
Swedish Irbesartan Left Ventricular Hypertrophy detection of CYP2C18 genotypes by real-time
Human P450s 485

fluorescence polymerase chain reaction. J. Pharm. tolbutamide hydroxylation by human liver micro-
Pharmacol. 52, 199-205. somes. DrugMetab. Dispos. 28, 354-359.
481. Zhu-Ge, J., Y.N. Yu, YL. Qian, and X. Li (2002). 493. Kaminsky, L.S., D.A. Dunbar, PR Wang,
Establishment of a transgenic cell line stably P. Beaune, D. Larrey, F.P, Guengerich et al. (1984).
expressing human cytochrome P450 2C18 and Human hepatic cytochrome P-450 composition as
identification of a CYP2C18 clone with exon 5 probed by in vitro microsomal metabolism of war-
missing. World J. Gastroenterol. 8, 888-892. farin. Drug Metab. Dispos. 12, 470-477.
482. Payne, VA., Y.T. Chang, and G.H. Loew (1999). 494. Wienkers, L.C., C.J. Wurden, E. Storch,
Homology modeling and substrate binding study K.L. Kunze, A.E. Rettie, and W F Trager (1996).
of human CYP2C18 and CYP2C19 enzymes. Formation of (^)-8-hydroxywarfarin in human
Proteins 37,204-211. liver microsomes: A new metabolic marker for the
483. Meier, U.T. and U.A. Meyer (1987). Genetic (5)-mephenytoin hydroxylase, P4502C19. Drug
polymorphism of human cytochrome P-450 (S)- Metab. Dispos. 24, 610-614.
mephenytoin 4-hydroxylase. Studies with human 495. Zhang, W, Y Ramamoorthy, R.F Tyndale,
autoantibodies suggest a functionally altered S.D. Glick, I.M. Maisonneuve, M.E. Kuehne et al.
cytochrome P-450 isozyme as cause of the genetic (2002). Metabolism of 18-methoxycoronaridine,
deficiency. Biochemistry 26, 8466-8474. an ibogaine analog, to 18-hydroxycoronaridine by
484. Wilkinson, G.R., F.P Guengerich, and R.A. Branch genetically variable CYP2C19. Drug Metab.
(1989). Genetic polymorphism of 5-mephenytoin Dispos. 30, 663-669.
hydroxylation. Pharmacol. Then 43, 53-76. 496. Ando, Y, E. Fuse, and WD. Figg (2002).
485. Yasumori, T., N. Murayama, Y Yamazoe, and Thalidomide metabolism by the CYP2C subfam-
R. Kato (1990). Polymorphism in hydroxylation of ily. Clin. Cancer Res. 8, 1964-1973.
mephenytoin and hexobarbital stereoisomers in 497. Yamazaki, H. andT Shimada (1997). Progesterone
relation to hepatic P-450 human-2. Clin. and testosterone hydroxylation by cytochromes
Pharmacol. Ther 47, 313-322. P450 2C19, 2C9, and 3A4 in human liver micro-
486. Kim, M.J., J.S. Bertino, Jr., A. Gaedigk, Y Zhang, somes. Arch. Biochem. Biophys. 346, 161-169.
E.M. Sellers, and A.N. Nafziger (2002). Effect of 498. Kappers, W.A., R.J. Edwards, S. Murray, and
sex and menstrual cycle phase on cytochrome A.R. Boobis (2001). Diazinon is activated by
P450 2C19 activity with omeprazole used as a bio- CYP2C19 in human liver. Toxicol. Appl.
marker. Clin. Pharmacol. Ther 12, 192-199. Pharmacol. Ill, 68-76.
487. Zhou, H.H., L.B. Anthony, A.J. Wood, and 499. Ibeanu, G.C., B.I. Ghanayem, R Linko, L. Li,
G.R. Wilkinson (1990). Induction of polymorphic E.G. Pedersen, and J.A. Goldstein (1996).
4'-hydroxylation of S-mephenytoin by rifampicin. Identification of residues 99, 220, and 221 of
Br I Clin. Pharmacol. 30, 471^75. human cytochrome P450 2C19 as key determi-
488. Blaisdell, J., H. Mohrenweiser, J. Jackson, nants of omeprazole hydroxylase activity. J. Biol.
S. Ferguson, S. Coulter, B. Chanas et al. (2002). Chem. 271, 12496-12501.
Identification and functional characterization of 500. Furuta, T, N. Shirai, M. Takashima, F Xiao,
new potentially defective alleles of human H. Hanai, K. Nakagawa et al. (2001). Effects of
CYP2C19. Pharmacogenetics 12, 703-711. genotypic differences in CYP2C19 status on cure
489. Desta, Z., X. Zhao, J.G. Shin, and D.A. Flockhart rates for Helicobacter pylori infection by dual
(2002). Clinical significance of the cytochrome therapy with rabeprazole plus amoxicillin.
P450 2C19 genetic polymorphism. Clin. Pharma- Pharmacogenetics 11, 341-348.
cokinet. 41, 913-958. 501. Furuta, T, N. Shirai, F Watanabe, S. Honda,
490. de Morals, S.M.F., G.R. Wilkinson, J. Blaisdell, K. Takeuchi, T. lida et al. (2002). Effect of cyto-
K. Nakamura, U.A. Meyer, and J.A. Goldstein chrome P4502C19 genotypic differences on cure
(1994). The major genetic defect responsible for rates for gastroesophageal reflux disease by lanso-
the polymorphism of ^S-mephenytoin metabolism prazole. Clin. Pharmacol. Ther 72, 453-460.
in humans. J. Biol. Chem. 269, 15419-15422. 502. Kita, T., T Sakaeda, N. Aoyama, T. Sakai,
491. Ferguson, R.J., S.M. De Morals, S. Benhamou, Y Kawahara, M. Kasuga et al. (2002). Optimal
C. Bouchardy, J. Blaisdell, G. Ibeanu et al. (1998). dose of omeprazole for CYP2C19 extensive
A new genetic defect in human CYP2C19: metabolizers in anti-Helicobacter pylori therapy:
Mutation of the initiation codon is responsible for Pharmacokinetic considerations. Biol. Pharm.
poor metabolism of S-mephenytoin. J. Pharmacol. Bull. 25, 923-927.
Exp. Ther 2H4, 356-361. 503. Kita, T, T Sakaeda, T. Baba, N. Aoyama,
492. Wester, M.R., J.M. Lasker, E.F. Johnson, and M. Kakumoto, Y Kurimoto et al. (2003). Different
J.L. Raucy (2000). CYP2C19 participates in contribution of CYP2C19 in the in vitro metabolism
486 F. Peter Guengerich

of three proton pump inhibitors. Biol. Pharm. Bull Elucidation of the genetic basis of the common
26, 386-390. 'intermediate metabolizer' phenotype for drug oxi-
504. Chau, T.K., S. Marakami, B. Kawai, K. Nasu, dation by CYP2D6. Pharmacogenetics 10,
T. Kubota, and A. Ohnishi (2000). Genotype analy- 577-581.
sis of the CYP2C19 gene in HCV-seropositive 516. Zanger, U.M., J. Fischer, S. Raimundo, T. Stuven,
patients with cirrhosis and hepatocellular carci- B.O. Evert, M. Schwab et al. (2001). Comprehen-
noma. Life Sci. 67, 1719-1724. sive analysis of the genetic factors determining
505. Roddam, P.L., S. Rollinson, E. Kane, E. Roman, expression and function of hepatic CYP2D6.
A. Moorman, R. Cartwright et al. (2000). Poor Pharmacogenetics 11, 573-585.
metabolizers at the cytochrome P450 2D6 and 517. Tyndale, R., T Aoyama, F Broly, T. Matsunaga,
2C19 loci are at increased risk of developing adult T. Inaba, W. Kalow et al. (1991). Identification of
acute leukaemia. Pharmacogenetics 10, 605-615. a new variant CYP2D6 allele lacking the codon
506. Treluyer, J.M., E. Jacqz-Aigrain, F. Alvarez, and encoding Lys-281: Possible association with the
T. Cresteil (1991). Expression of CYP2D6 in devel- poor metabolizer phenotype. Pharmacogenetics 1,
oping human liver. Eur. J. Biochem. 202, 583-588. 26-32.
507. Lo Guidice, J.M., D. Marez, N. Sabbagh, 518. Yu, A., B.M. Kneller, A.E. Rettie, and
M. LegrandAndreoletti, C. Spire, E. Alcaide et al. R.L. Haining (2002). Expression, purification,
(1997). Evidence for CYP2D6 expression in biochemical characterization, and comparative
human lung. Biochem. Biophys. Res. Commun. function of human cytochrome P450 2D6.1,
241, 79-85. 2D6.2, 2D6.10, and 2D6.17 allelic isoforms.
508. Siegle, I., R Fritz, K. Eckhardt, U.M. Zanger, and J. Pharmacol. Exp. Ther 303, 1291-1300.
M. Eichelbaum (2001). Cellular localization and 519. Fletcher, B., D.B. Goldstein, A.L. Bradman,
regional distribution of CYP2D6 mRNA and pro- M.E. Weale, N. Bradman, and M.G. Thomas
tein expression in human brain. Pharmacogenetics (2003). High-throughput analysis of informative
11,237-245. CYP2D6 compound haplotypes. Genomics 81,
509. Miksys, S., Y. Rao, E. Hoffmann, D.C. Mash, and 166-174.
R.F. Tyndale (2002). Regional and cellular expres- 520. Dahl, M.L., I. Johansson, L. Bertilsson,
sion of CYP2D6 in human brain: Higher levels in M. Ingelman-Sundberg, and F Sjoqvist (1995).
alcoholics. J. Neurochem. 82, 1376-1387. Ultrarapid hydroxylation of debrisoquine in a
510. Idle, J.R., A. Mahgoub, R. Lancaster, and Swedish population. Analysis of the molecular
R.L. Smith (1978). Hypotensive response to genetic basis. J. Pharmacol. Exp. Ther 174,
debrisoquine and hydroxylation phenotype. Life 516-520.
Sci. 11, 979-984. 521. Lundqvist, E., I. Johansson, and M. Ingelman-
511. Alvan, G., C. von Bahr, P. Seideman, and Sundberg (1999). Genetic mechanisms for dupli-
F Sjoqvist (1982). High plasma concentrations of cation and multiduplication of the human CYP2D6
p-receptor blocking drugs and deficient debriso- gene and methods for detection of duplicated
quine hydroxylation. Lancet i, 333. CYP2D6 genes. Gene 226, 327-338.
512. Evans, D.A.P, D. Harmer, D.Y Downham, 522. Lovlie, R., A.K. Daly, A. Molven, J.R. Idle, and
E.J. Whibley, J.R. Idle, J. Ritchie et al. (1983). The V.M. Steen (1996). Ultrarapid metabolizers of
genetic control of sparteine and debrisoquine debrisoquine: Characterization and PCR-based
metabolism in man with new methods of analysing detection of alleles with duplication of the
bimodal distributions. J. Med. Genet. 20, 321-329. CYP2D6 gene. FEBS Lett. 392, 30-34.
513. Skoda, R.C., F.J. Gonzalez, A. Demierre, and 523. Kohnke, M.D., E.U. Griese, D. Stosser, I. Gaertner,
U.A. Meyer (1988). Two mutant alleles of the and G. Barth (2002). Cytochrome P450 2D6 defi-
human cytochrome P-450dbl gene (P450C2D1) ciency and its clinical relevance in a patient treated
associated with genetically deficient metabolism with risperidone. Pharmacopsychiatry 35, 116-118.
of debrisoquine and other drugs. Proc. Natl. Acad. 524. Guengerich, FP. (2001). Common and uncommon
Sci. USA 85, 5240-5243. cytochrome P450 reactions related to metabolism
514. Gaedigk, A., R.R. Gotschall, N.S. Forbes, and chemical toxicity. Chem. Res. Toxicol. 14,
S.D. Simon, G.L. Keams, and J.S. Leeder (1999). 611-650.
Optimization of cytochrome P4502D6 (CYP2D6) 525. Wolff, T, L.M. Distlerath, M.T Worthington,
phenotype assignment using a genotyping algorithm J.D. Groopman, G.J. Hammons, FF Kadlubar et al.
based on allele frequency data. Pharmacogenetics (1985). Substrate specificity of human liver
9, 669-682. cytochrome P-450 debrisoquine 4-hydroxylase
515. Raimundo, S., J. Fischer, M. Eichelbaum, probed using immunochemical inhibition and
E.U. Griese, M. Schwab, and U.M. Zanger (2000). chemical modeling. Cancer Res. 45, 2116-2122.
Human P450s 487

526. Islam, S.A., C.R. Wolf, M.S. Lennard, and substrates reveals that CYP2D6 is a 5-methoxyin-
M.J.E. Sternberg (1991). A three-dimensional dolethylamine O-demethylase. Pharmacogenetics
molecular template for substrates of human 13,307-319.
cytochrome P450 involved in debrisoquine 4- 538. Yu, A.M., J.R. Idle, E.G. Byrd, K.W Krausz,
hydroxylation. Carcinogenesis 12, 2211-2219. A. Kupfer, and F.J. Gonzalez (2003). Regeneration
527. Strobl, G.R., S. von Kruedener, J. Stockigt, of serotonin from 5-methoxytryptamine by poly-
F.P. Guengerich, andT. Wolff (1993). Development morphic human CYP2D6. Pharmacogenetics 13,
of a pharmacophore for inhibition of human liver 173-181.
cytochrome P-450 2D6: Molecular modeling and 539. Koymans, L., N.P.E. Vermeulen, A. Baarslag, and
inhibition studies. J. Med. Chem. 36, 1136-1145. G. Donne-Op den Kelder (1993). A preliminary
528. Koymans, L., N.P.E. Vermeulen, S.A.B.E. van 3D model for cytochrome P450 2D6 constructed
Acker, J.M. te Koppele, J.J.P. Heykants, by homology model building. J. Comput.Aided
K. Lavrijsen et al. (1992). A predictive model for Mol.Des. 7,281-289.
substrates of cytochrome P450-debrisoquine 540. de Groot, M.J., N.RE. Vermeulen, J.D. Kramer,
(2D6). Chem. Res. Toxicol. 5, 211-219. F.A.A. van Acker, and G.M. Donne-Op den Kelder
529. de Groot, M.J., G.J. Bijloo, B.J. Martens, (1996). A three-dimensional protein model for
F.A.A. van Acker, and N.RE. Vermeulen (1997). A human cytochrome P450 2D6 based on the crystal
refined substrate model for human cytochrome structure of P450 101, P450 102, and P450 108.
P450 2D6. Chem. Res. Toxicol. 10, 4 1 ^ 8 . Chem. Res. Toxicol. 9, 1079-1091.
530. Upthagrove, A.L. and W.L. Nelson (2001). 541. Lewis, D.F.V, RJ. Eddershaw, RS. Goldfarb, and
Importance of amine pKa and distribution coeffi- M.H. Tarbit (1997). Molecular modeling of
cient in the metabolism of fluorinated propranolol cytochrome P4502D6 (CYP2D6) based on an
derivatives. Preparation, identification of metabo- alignment with CYP102: Structural studies on spe-
lite regioisomers, and metabolism by CYP2D6. cific CYP2D6 substrate metabolism. Xenobiotica
DrugMetah. Dispos. 29, 1377-1388. 27,319-340.
531. Miller, G.R, I.H. Hanna, Y. Nishimura, and 542. Modi, S., M.J. Paine, M.J. Sutcliffe, L.Y Lian,
F.P. Guengerich (2001). Oxidation of phenylethy- WU. Primrose, C.R. Wolfe/ al. Roberts (1996). A
lamine derivatives by cytochrome P450 2D6: model for human cytochrome P450 2D6 based on
The issue of substrate protonation in binding and homology modeling and NMR studies of substrate
catalysis. Biochemistry 40, 14215-14223. binding. Biochemistry 35, 4540-^550.
532. Grace, J.M., M.T. Kinter, and T.L. Macdonald 543. de Groot, M.J., M.J. Ackland, VA. Home,
(1994). Atypical metabolism of deprenyl and A.A. Alex, and B.C. Jones (1999). A novel
its enantiomer, (*S)-(+)-7V,a-dimethyl-A^-propyny- approach to predicting P450 mediated drug metab-
Iphenethylamine, by cytochrome P450 2D6. Chem. olism. CYP2D6 catalyzed A^dealkylation reac-
Res. Toxicol. 7, 286-290. tions and qualitative metabolite predictions using a
533. Niwa, T., Y. Yabusaki, K. Honma, N. Matsuo, combined protein and pharmacophore model for
K. Tatsuta, F. Ishibashi et al. (1998). Contribution CYP2D6. J. Med. Chem. 42, 4062-4070.
of human hepatic cytochrome P450 isoforms to 544. Kirton, S.B., C.A. Kemp, N.R Tomkinson,
regioselective hydroxylation of steroid hormones. S. St.-Gallay, and M.J. Sutcliffe (2002). Impact
Xenobiotica 28, 539-547. of incorporating the 2C5 crystal structure into
534. Hiroi, T, T. Chow, S. Imaoka, and Y Funae (2000). comparative models of cytochrome P450 2D6.
Catalytic specificity of each CYP2D isoform in rat Pro/ems 49, 216-231.
and human. Abstracts, 13th Int. Sympos. Microsomes 545. Venhorst, J., A.M. ter Laak, J.N. Commandeur,
and Drug Oxidations, 10-14 July, Stresa: p. 113. Y Funae, T. Hiroi, and N.R Vermeulen (2003).
535. Guengerich, EP, G.R Miller, I.H. Hanna, Homology modeling of rat and human cytochrome
M.V Martin, S. Leger, C. Black et al (2002). P450 2D (CYP2D) isoforms and computational
Diversity in the oxidation of substrates by rationalization of experimental ligand-binding
cytochrome P450 2D6. Lack of an obligatory role specificities. J. Med. Chem. 46, 74-86.
of aspartate 301-substrate electrostatic bonding. 546. Crespi, C.L., D.T. Steimel, B.W. Penman,
Biochemistry 41, 11025-11034. K.R. Korzekwa, P. Femandez-Salguero, J.T.M.
536. Martinez, C , J.A. Agundez, G. Gervasini, Buters et al. (1995). Comparison of substrate
R. Martin, and J. Benitez (1997). Tryptamine: A metabolism by wild type CYP2D6 protein and
possible endogenous substrate for CYP2D6. a variant containing methionine, not valine, at
Pharmacogenetics 7, 85-93. position 374. Pharmacogenetics 5, 234-243.
537. Yu, A.M., J.R. Idle, T. Herraiz, A. Kiipfer, and 547. Ellis, S.W, K. Rowland, M.J. Ackland, E. Rekka,
F.J. Gonzalez (2003). Screening for endogenous A.R Simula, M.S. Lennard et al. (1996). Influence
488 F. Peter Guengerich

of amino acid residue 374 of cytochrome P-450 NADPH-cytochrome P450 reductase. Biochemistry
2D6 (CYP2D6) on the regio- and enantio-selective 36, 4461-4470.
metaboUsm of metoprolol. Biochem. J. 316, 558. Hanna, LH., J.A. Krauser, H. Cai, M.-S. Kim, and
647-654. F.P. Guengerich (2001). Diversity in mechanisms
548. ElUs, S.W., G.P. Hayhurst, G. Smith, T. Lightfoot, of substrate oxidation by cytochrome P450 2D6.
M.M.S. Wong, A.R Simula et al (1995). Evidence Lack of an allosteric role of NADPH-cytochrome
that aspartic acid 301 is a critical substrate-contact P450 reductase in catalytic regioselectivity. J. Biol.
residue in the active site of cytochrome P450 2D6. Chem. 276,39553-39561.
J. Biol. Chem. 270, 29055-29058. 559. Guengerich, RP, G.P Miller, LH. Hanna, H. Sato,
549. Hanna, I.H., M.-S. Kim, and F.R Guengerich and M.V Martin (2002). Oxidation of
(2001). Heterologous expression of cytochrome methoxyphenethylamines by cytochrome P450
P450 2D6 mutants, electron transfer, and catalysis 2D6. Analysis of rate-limiting steps. J. Biol. Chem.
of bufiiralol hydroxylation. The role of aspartate 277,33711-33719.
301 in structural integrity. Arch. Biochem. Biophys. 560. Fukuda, T, Y Nishida, S. Imaoka, T Hiroi,
393,255-261. M. Naohara, Y Funae et al. (2000). The decreased
550. Paine, M.J., L.A. McLaughlin, J.U. Flanagan, in vivo clearance of CYP2D6 substrates by
C.A. Kemp, M.J. Sutcliffe, G.C. Roberts et al. CYP2D6* 10 might be caused not only by the low-
(2003). Residues glutamate 216 and aspartate 301 expression but also by low affinity of CYP2D6.
are key determinants of substrate specificity and Arch. Biochem. Biophys. 380, 303-308.
product regioselectivity in cytochrome P450 2D6. 561. Otton, S.V, T Inaba, and W Kalow (1984).
J. Biol. Chem. 278, 4021-4027. Competitive inhibition of sparteine oxidation in
551. Smith, G., S. Modi, 1. Pillai, L.-Y. Lian, human liver by p-adrenoceptor antagonists and
M.J. Sutcliffe, M.P Pritchard et al. (1998). other cardiovascular drugs. Life Sci. 34, 73-80.
Determinants of the substrate specificity of human 562. Palamanda, J.R., C.N. Casciano, L.A. Norton,
cytochrome P-450 CYP2D6: Design and construc- R.P Clement, L.V Favreau, C.C. Lin et al. (2001).
tion of a mutant with testosterone hydroxylase Mechanism-based inactivation of CYP2D6 by 5-
activity Biochem. J. 331, 783-792. fluoro-2-[4-[(2-phenyl-1 //-imidazol-5-yl)methyl]-
552. Wiseman, H. and D.F Lewis (1996). The metabo- 1 -piperazinyl]pyrimidine. Drug Metab. Dispos. 29,
lism of tamoxifen by human cytochromes P450 863-867.
is rationalized by molecular modelling of the 563. Gates, N.S., R.R. Shah, J.R. Idle, and R.L. Smith
enzyme-substrate interactions: Potential impor- (1983). Influence of oxidation polymorphism on
tance to its proposed anti-carcinogenic/carcino- phenformin kinetics and dynamics, Clin.
genic actions. Carcinogenesis 17, 1357-1360. Pharmacol. Then 34, 827-834.
553. Lightfoot, T, S.W Ellis, J. Mahling, M.J. 564. Gates, N.S., R.R. Shah, PL. Drury, J.R. Idle, and
Ackland, EE. Blaney, G.J. Bijloo et al. (2000). R.L. Smith (1982). Captopril-induced agranulocy-
Regioselectivity hydroxylation of debrisoquine by tosis associated with an impairment of debriso-
cytochrome P4502D6: Implications for active site quine hydroxylation. Br J. Pharmacol. 14, 60IP
modelling. Xenobiotica 30, 219-233. 565. Rau, T, R. Heide, K. Bergmann, H. Wuttke,
554. Ellis, S.W, G.P Hayhurst, T. Lightfoot, G. Smith, U. Werner, N. Feifel, and T. Eschenhagen (2002).
J. Harlow, K. Rowland-Yeo et al. (2000). Evidence Effect of the CYP2D6 genotype on metoprolol
that serine 304 is not a key ligand-binding residue metabolism persists during long-term treatment.
in the active site of cytochrome P450 2D6. Pharmacogenetics 12, 465^72.
Biochem. J. ^^5, 565-51 \. 566. Dome, J.L., K. Walton, W Slob, and A.G, Renwick
555. Hayhurst, G.P, J. Harlow, J. Chowdry, E. Gross, (2002), Human variability in polymorphic CYP2D6
E. Hilton, M.S. Lennard et al. (2001). Influence of metabolism: Is the kinetic default uncertainty factor
phenylalanine-481 substitutions on the catalytic adequate? Food Chem. Toxicol. 40, 1633-1656.
activity of cytochrome P450 2D6. Biochem. J. 355, 567. Chou, W.H., EX. Yan, J. de Leon, J, Bamhill,
373-379. T, Rogers, M, Cronin et al. (2000). Extension of a
556. Ramamoorthy, Y, R.F Tyndale, and E.M. Sellers pilot study: Impact from the cytochrome P450 2D6
(2001). Cytochrome P450 2D6.1 and cytochrome polymorphism on outcome and costs associated with
P450 2D6.10 differ in catalytic activity for multi- severe mental illness. J. Clin. Psychopharmacol. 20,
ple substrates. Pharmacogenetics 11, 477^87. 246-251.
557. Modi, S., D.E. Gilham, M.J. SutcHffe, L.-Y. Lian, 568. Dahl, M.L. (2002). Cytochrome P450 phenotyp-
WU. Primrose, C.R. Wolf e/a/. (1997). 1-Methyl- ing/genotyping in patients receiving antipsy-
4-phenyl-l,2,3,6-tetrahydropyridine as a substrate chotics: Useful aid to prescribing?. Clin.
of cytochrome P450 2D6: Allosteric effects of Pharmacokinet. 41, 453^70.
Human P450s 489

569. Scolnick, E.M. (2002). Discovery and develop- 582. Zanger, U.M., H.R Hauri, J. Loeper, J.C Homberg,
ment of antidepressants: A perspective from a and U.A. Meyer (1988). Antibodies against human
pharmaceutical discovery company. Biol. cytochrome P-450^^| in autoimmune hepatitis type
Psychiatry SI, 154-156. II. Proc. Natl. Acad. Sci. USA 85, 8256-2860.
570. Caporaso, N., R.B. Hayes, M. Dosemeci, R. Hoover, 583. Manns, M.P. (1991). Cytochrome P450 enzymes as
R. Ayesh, M. Hetzel et al. (1989). Lung cancer risk, human autoantigens. Immunol. Res. 10, 503-507.
occupational exposure, and the debrisoquine meta- 584. Manns, M.P, K.J. Griffin, K.F Sullivan, and
bolic phenotype. Cancer Res. 49, 3675-3679. E.F. Johnson (1991). LKM-1 autoantibodies recog-
571. Bouchardy, C , S. Benhamou, and R Dayer (1996). nize a short linear sequence in P450IID6, a
The effect of tobacco on lung cancer risk depends cytochrome P-450 monooxygenase. J. Clin. Invest.
on CYP2D6 activity Cancer Res. 56, 251-253. 88, 1370-1378.
572. Shaw, G.L., R.T. Falk, J.N. Frame, B. WeiflFenbach, 585. Loeper, J., V Descatoire, M. Maurice, P. Beaune,
J.V Nesbitt, H.I. Pass et al. (1998). Genetic poly- J. Belghiti, D. Houssin et al. (1993). Cytochromes
morphism of CYP2D6 and lung cancer risk. P-450 in human hepatocyte plasma membrane:
Cancer Epidemiol. Biomarkers Prev. 7, 215—219. Recognition by several autoantibodies.
573. Rostami-Hodjegan, A., M.S. Lennard, H.F. Woods, Gastroenterology 104, 203-216.
and G.T. Tucker (1998). Meta-analysis of studies 586. Vergani, D. (2000). LKM antibody: Getting in
of the CYP2D6 polymorphism in relation to lung some target practice. Gut 46, 449^50.
cancer and Parkinson's disease. Pharmacogenetics 587. Vitozzi, S., P. Lapierre, I. Djilali-Saiah, and
8, 227-238. F. Alvarez (2002). Autoantibody detection in type
574. Legrand-Andreoletti, M., I. Stucker, D. Marez, 2 autoimmune hepatitis using a chimera recombi-
R Galais, J. Cosme, N. Sabbagh et al. (1998). nant protein. J. Immunol. Meth. 262, 103-110.
Cytochrome P450 CYP2D6 gene polymorphism 588. Nolte, W, F Polzien, B. Sattler, G. Ramadori, and
and lung cancer susceptibility in Caucasians. H. Hartmann (1995). Recurrent episodes of acute
Pharmacogenetics 8, 7-14. hepatitis associated with LKM-1 (cytochrome
575. Christensen, P.M., PC. Gotzsche, and K. Brosen P450 2D6) antibodies in identical twin brothers.
(1997). The sparteine/debrisoquine (CYP2D6) J. Hepatol. 23, 734-739.
oxidation polymorphism and the risk of lung can- 589. Orme-Johnson, W.H. and D.M. Ziegler (1965).
cer: A meta-analysis. Eur J. Clin. Pharmacol. 51, Alcohol mixed function oxidase activity of
389-393. mammalian liver micoromes. Biochem. Biophys.
576. Fleming, CM., A. Kaisary, G.R. Wilkinson, Res. Commun. 21, 78—82.
R Smith, and R.A. Branch (1992). The ability to 590. Lieber, C S . and L.M. DeCarU (1970). Hepatic
4-hydroxylate debrisoquine is related to recurrence microsomal ethanol oxidizing system: In vitro
of bladder cancer. Pharmacogenetics 2, 128-134. chracteristics and adaptive properties in vivo.
577. Worrall, S.F, M. Corrigan, A. High, D. Starr, J. Biol. Chem. 245, 2505-2512.
C. Matthias, C.R. Wolfed al. (1998). Susceptibility 591. Teschke, R., Y Hasumura, and C S . Lieber (1974).
and outcome in oral cancer: Preliminary data Hepatic microsomal ethanol-oxidizing system:
showing an association with polymorphism in Solubilization, isolation and characterization.
cytochrome P450 CYP2D6. Pharmacogenetics 8, Arch. Biochem. Biophys. 163, 404-415.
433^*39. 592. Ryan, D.E., L. Ramanathan, S. lida, PE. Thomas,
578. Barbeau, A., M. Roy, S. Paris, T. Cloutier, M. Haniu, J.E. Shively et al (1985).
L. Plasse, and J. Poirier (1985). Ecogenetics of Characterization of a major form of rat hepatic
Parkinson's disease: 4-hydroxylation of debriso- microsomal cytochrome P-450 induced by isoni-
quine. Lancet ii, 1213-1215. azid. J. Biol. Chem. 260, 6385-6393.
579. Armstrong, M., A.K. Daly, S. Cholerton, 593. Wrighton, S.A., PE. Thomas, D.E. Ryan, and
D.N. Bateman, and J.R. Idle (1992). Mutant W. Levin (1987). Purification and characterization
debrisoquine hydroxylation genes in Parkinson's of ethanol-inducible human hepatic cytochrome
disease. Lfl^ce? 339, 1017-1018. P-450HLJ. Arch. Biochem. Biophys. 258, 292-297.
580. Harhangi, B.S., B.A. Oostra, R Heutink, CM. van 594. Umeno, M., O.W McBride, C.-S. Yang,
Duijn, A. Hofman, and M.M. Breteler (2001). H.V Gelboin, and FJ. Gonzalez (1988). Human
CYP2D6 polymorphism in Parkinson's disease: ethanol-inducible P450IIE1: Complete gene
The Rotterdam Study. Mov. Disord. 16, 290-293. sequence, promoter characterization, chromosome
581. Allam, M.F, A. Serrano del Castillo, and mapping, and cDNA-directed expression.
R. Fernandez-Crehuet Navajas (2002). Smoking Biochemistry 21, 9006-9013.
and Parkinson's disease: Explanatory hypothesis. 595. Guengerich, FP and CG. Turvy (1991).
Int. J. Neurosci. 112, 851-854. Comparison of levels of several human microsomal
490 F. Peter Guengerich

cytochrome P-450 enzymes and epoxide hydrolase P450 2E1 to the plasma membrane. J. Biol. Chem.
in normal and disease states using immunochemi- 275, 17130-17135.
cal analysis of surgical liver samples. J. Pharmacol. 607. Thomas, RE., S. Bandiera, S.L. Maines,
Exp. Then 256, 1189-1194. D.E. Ryan, and W. Levin (1987). Regulation of
596. Vieira, I., M. Sonnier, and T. Cresteil (1996). cytochrome P-450J, a high-affinity A^-nitrosodi-
Developmental expression of CYP2E1 in the methylamine demethylase, in rat hepatic micro-
human liver: Hypermethylation control of gene somes. Biochemistry 26, 2280-2289.
expression during the neonatal period. Eur. J. 608. Koop, D.R. and D.J. Tierney (1990). Multiple
Biochem. 238, 476-483. mechanisms in the regulation of ethanol-inducible
597. Ding, X. and L.S. Kaminsky (2003). Human extra- cytochrome P450IIE1. BioEssays 12, 429^35.
hepatic cytochromes P450: Function in xenobiotic 609. Ueno, T. and F.J. Gonzalez (1990). Transcriptional
metabolism and tissue-selective chemical toxicity control of the rat hepatic CYP2E1 gene. Mol. Cell
in the respiratory and gastrointestinal tracts. Annu. Biol. 10, 4495^505.
Rev. Pharmacol. Toxicol. 43, 149-173. 610. Woodcroft, K.J. and R.F Novak (1999). The role of
598. Warner, M. and J.A. Gustafsson (1994). Effect of phosphatidylinositol 3-kinase, Src kinase, and pro-
ethanol on cytochrome P450 in the rat brain. Proc. tein kinase A signaling pathways in insulin and
Natl. Acad. Sci. USA 91, 1019-1023. glucagon regulation of CYP2E1 expression.
599. Upadhya, S.C, RS. Tirumalai, M.R. Boyd, T. Mori, Biochem. Biophys. Res. Commun. 266, 304-307.
and V. Ravindranath (2000). Cytochrome P4502E 611. Siewert, E., R. Bort, R. Kluge, PC. Heinrich,
(CYP2E) in brain: Constitutive expression, induc- J. Castell, and R. Jover (2000). Hepatic
tion by ethanol and localization by fluorescence cytochrome P450 down-regulation during aseptic
in situ hybridization. Arch. Biochem. Biophys. 373, inflammation in the mouse is interleukin 6 depend-
23-34. ent. Hepatology 32, 49-55.
600. Kazakoff, K., P. Iversen, T. Lawson, J. Baron, 612. Lagadic-Gossmann, D , C. Lerche, M. Rissel,
F.P. Guengerich, and P. Pour (1994). Involvement F. Joannard, M. Galisteo, A. Guillouzo et al. (2000).
of cytochrome P450 2El-like isoform in the acti- The induction of the human hepatic CYP2E1 gene
vation of A^-nitrosobis(2-oxopropyl)amine in the by interleukin 4 is transcriptional and regulated by
rat nasal mucosa. Eur J. Cancer 30B, 179-185. protein kinase C. Cell Biol. Toxicol. 16, 221-233.
601. Norton, I.D., M.V Apte, RS. Haber, 613. Hakkola, J., Y Hu, and M. Ingelman-Sundberg (2003).
G.W. McCaughan, R.C. Pirola, and J.S. Wilson Mechanisms of down-regulation of CYP2E1 expres-
(1998). Cytochrome P4502E1 is present in rat pan- sion by inflammatory cytokines in rat hepatoma
creas and is induced by chronic ethanol adminis- cdls. J. Pharmacol. Exp. Then 304, 1048-1054.
tration. Gut 42, 426^30. 614. Kim, S.G. and R.F Novak (1990). Induction of rat
602. Larson, J.R., M.J. Coon, and T.D. Porter (1991). hepatic P450IIE1 (CYP 2E1) by pyridine:
Purification and properties of a shortened form Evidence for a role of protein synthesis in the
of cytochrome P-450 2E1: Deletion of the NH2- absence of transcriptional activation. Biochem.
terminal membrane-insertion signal peptide does Biophys. Res. Commun. 166, 1072-1079.
not alter the catalytic activities. Proc. Natl. Acad. 615. Kocarek, T.A., R.C. Zangar, and R.F Novak
Sci. USA ^^,9\4\-9U5. (2000). Post-transcriptional regulation of rat
603. Gillam, E.M.J., Z. Guo, and F.R Guengerich CYP2E1 expression: Role of CYP2E1 mRNA
(1994). Expression of modified human untranslated regions in control of translational effi-
cytochrome P450 2E1 in Escherichia coli, purifi- ciency and message stability. Arch. Biochem.
cation, and spectral and catalytic properties. Arch. Biophys. 376, 180-190.
Biochem. Biophys. 312, 59-66. 616. Roberts, B.J., B.J. Song, Y Soh, S.S. Park, and
604. Neve, E.RA. and M. Ingelman-Sundberg (1999). S.E. Shoaf (1995). Ethanol induces CYP2E1 by
A soluble NH2-terminally truncated catalytically protein stabilization: Role of ubiquitin conjugation
active form of rat cytochrome P450 2E1 targeted to in the rapid degradation of CYP2E1. J. Biol. Chem.
liver mitochondria. FEBS Lett. 460, 309-314. 270, 29632-29635.
605. Robin, M.A., H.K. Anandatheerthavarada, J.K. Fang, 617. Yang, M.X. and A.I. Cederbaum (1997).
M. Cudic, L. Otvos, and N.G. Avadhani (2001). Characterization of cytochrome P4502E1 turnover
Mitochondrial targeted cytochrome P450 2E1 (P450 in transfected HepG2 cells expressing human
MT5) contains an intact N-terminus and requires CY?2E\.Arch. Biochem. Biophys. 341, 25-33.
mitochondrial specific electron transfer proteins 618. Emery, M.G., C. Jubert, K.E. Thummel, and
for activity J. Biol. Chem. 276, 24680-24689. E.D. Kharasch (1999). Duration of cytochrome P-
606. Neve, E.R and M. Ingelman-Sundberg (2000). 450 2E1 (CYP2E1) inhibition and estimation of
Molecular basis for the transport of cytochrome functional CYP2E1 enzyme half-life after
Human P450s 491

single-dose disulfiram administration in humans. (CYP2E1). Biochem. Biophys. Res. Commun. 179,
J. Pharmacol. Exp. Ther. 291, 213-219. 689-694.
619. Chien, J.Y., K.E. Thummel, and J.T. Slattery 631. Kunitoh, S., S. Imaoka, T. Hiroi, Y. Yabusaki,
(1997). Pharmacokinetic consequences of induc- T. Monna, and Y. Funae (1997). Acetaldehyde as
tion of CYP2E1 by ligand stabilization. Drug well as ethanol is metabolized by human CYP2E1.
Metab. Dispos. 25, 1165-1175. J. Pharmacol. Exp. Ther 280, 527-532.
620. Watanabe, J., S. Hayashi, and K. Kawajiri (1994). 632. Bell-Parikh, L.C. and EP Guengerich (1999).
Different regulation and expression of the human Kinetics of cytochrome P450 2E1-catalyzed oxida-
CYP2E1 gene due to the Rsal polymorphism in tion of ethanol to acetic acid via acetaldehyde.
the 5'-flanking region. J. Biochem. (Tokyo) 116, J. Biol. Chem. 274, 23833-23840.
321-326. 633. Tassaneeyakul, W, M.E. Veronese, D.J. Birkett,
621. Fairbrother, K.S., J. Grove, I. de Waziers, F.J. Gonzalez, and J.O. Miners (1993). Validation
D.T. Steimel, C.P Day, C.L. Crespi et al. (1998). of 4-nitrophenol as an in vitro substrate probe
Detection and characterization of novel polymor- for human liver CYP2E1 using cDNA expression
phisms in the CYP2E1 gene. Pharmacogenetics 8, and microsomal kinetic techniques. Biochem.
543-552. Pharmacol. 46, 1975-1981.
622. Fritsche, E., G.S. Pittman, and D.A. Bell (2000). 634. Peter, R., R.G. Bocker, PH. Beaune, M. Iwasaki,
Localization, sequence analysis, and ethnic distri- EP Guengerich, and C.-S. Yang (1990). Hydro-
bution of a 96-bp insertion in the promoter of the xylation of chlorzoxazone as a specific probe for
human CYP2E1 gene. Mutat. Res. 432, 1-5. human liver cytochrome P-450 IIEl. Chem. Res.
623. Powell, H., N.R. Kitteringham, M. Pirmohamed, Toxicol. 3, 566—573.
D.A. Smith, and B.K. Park (1998). Expression of 635. Yamazaki, H., Z. Guo, and EP Guengerich (1995).
cytochrome P4502E1 in human liver: Assessment Selectivity of cytochrome P450 2E1 in chlorzoxazone
by mRNA, genotype and phenotype. Pharma- 6-hydroxylation. Drug Metab. Dispos. 23, 438^40.
cogenetics 8, 411-^21. 636. Preussmann, R. and B.W Stewart (1984). A^-Nitroso
624. Inoue, K., H. Yamazaki, and T. Shimada (2000). carcinogens. In C.E. Searle (ed.). Chemical
Characterization of liver microsomal 7-ethoxy- Carcinogens, Vol. 2. American Chemical Society,
coumarin O-deethylation and chlorzoxazone 6- Washington, D. C , pp. 643-828.
hydroxylation activities in Japanese and Caucasian 637. Argus, M.E, J.C. Arcos, K.M. Pastor, B.C. Wu,
subjects genotyped for CYP2E1 gene. Arch. and N. Venkatesan (1976). Dimethylnitrosamine-
Toxicol. 74, 372-378. demethylase: Absence of increased enzyme
625. O'Shea, D., S.N. Davis, R.B. Kim, and catabolism and multiplicity of effector sites in
G.R. Wilkinson (1994). Effect of fasting and obe- repression. Hemoprotein involvement. Chem. Biol.
sity in humans on the 6-hydroxylation of chlorox- Interact. 13, 127-140.
azone: A putative probe of CYP2E1 activity. Clin. 638. Lake, B.G., C.E. Heading, J.C. Phillips,
Pharmacol. Then 56, 359-367. S.D. Gangolh, and A.G. Lloyd (1974). Some studies
626. Koop, D.R. and J.R Casazza (1985). Identification on the metabolism in vitro of dimethylnitrosamine
of ethanol-inducible P-450 isozyme 3a as the ace- by rat liver. Biochem. Soc. Transact. 2, 610-612.
tone and acetol monooxygenase of rabbit micro- 639. Levin, W, PE. Thomas, N. Oldfield, and
somes. J. Biol. Chem. 260, 13607-13612. D.E. Ryan (1986). 7V-Demethylation of 7V-nitroso-
627. Bondoc, EY, Z. Bao, W.Y Hu, EJ. Gonzalez, dimethylamine catalyzed by purified rat hepatic
Y Wang, C.S. Yang et al. (1999). Acetone cataboUsm microsomal cytochrome P-450: Isozyme speci-
by cytochrome P450 2E1: Studies with CYP2E1- ficity and role of cytochrome b^. Arch. Biochem.
nuU mice. Biochem. Pharmacol. 105, 83-88. Biophys. 248, 158-165.
628. Kashiwagi, T., S. Ji, J.J. Lemasters, and 640. Wrighton, S.A., PE. Thomas, D.T. Molowa,
R.G. Thurman (1982). Rates of alcohol dehydro- M. Haniu, J.E. Shively, S.L. Maines et al. (1986).
genase-dependent ethanol metabolism in peripor- Characterization of ethanol-inducible human liver
tal and pericentral regions of the perfused rat liver. A^-nitrosodimethylamine demethylase. Biochemistry
Mol. Pharmacol. 21, 438^43. 25,6731-6735.
629. Kono, H., B.U. Bradford, M. Yin, K.K. Sulik, 641. Bastien, M.C. and J.R Villeneuve (1998).
D.R. Koop, J.M. Peters et al. (1999). CYP2E1 is Characterization of cytochrome P450 2E1 activity
not involved in early alcohol-induced liver injury. by the [^"^CJnitrosodimethylamine breath test.
Am. J. Physiol. Ill, G1259-G1267. Can. J. Physiol. Pharmacol. 76, 756-763.
630. Terelius, Y, C. Norsten-Hoog, T. Cronholm, and 642. Raucy, J.L., J.C. Kraner, and J.M. Lasker (1993).
M. Ingelman-Sundberg (1991). Acetaldehyde as a Bioactivation of halogenated hydrocarbons by
substrate for ethanol-inducible cytochrome P450 cytochrome P4502E1. Crit. Rev. Toxicol. 23, 1-20.
492 F. Peter Guengerich

643. Hong, J.Y., C.S. Yang, M. Lee, Y.Y. Wang, mutagenicity of dialkylnitrosamines in methyl-
W. Huang, Y. Tan et ah (1997). Role of cytochromes transferase-deficient strains of Salmonella
P450 in the metabolism of methyl tert-\miy\ ether typhimurium. Mutat. Res. 484, 61-68.
in human livers. Arch. Toxicol. 71, 266-269. 655. Schenkman, LB. and I. Jansson (2003). The many
644. Wang, H., B. Chanas, and B.I. Ghanayem (2002). roles of cytochrome by Pharmacol. Ther 97,
Cytochrome P450 2E1 (CYP2E1) is essential for 139-152.
acrylonitrile metabolism to cyanide: Comparative 656. Tan, Y, S.P White, S.R. Paranawithana, and
studies using CYP2El-null and wild-type mice. C.S. Yang (1997). A hypothetical model for the
DrugMetab. Dispos. 30, 911-917. active site of human cytochrome P4502E1.
645. Hoffler, U., H.A. El-Masri, and B.I. Ghanayem Xenobiotica 27, 287-299.
(2003). Cytochrome P450 2E1 (CYP2E1) is the prin- 657. Lewis, D.F, B.G. Lake, M.G. Bird, G.D. Loizou, M.
cipal enzyme responsible for urethane metabolism: Dickins, and PS. Goldfarb (2003). Homology mod-
Comparative studies using CYP2El-null and wild- elling of human CYP2E1 based on the CYP2C5
type mice. J. Pharmacol Exp. Ther. 305, 557-564. crystal structure: Investigation of enzyme-substrate
646. Clarke, S.E., S.J. Baldwin, J.C. Bloomer, and enzyme-inhibitor interactions. Toxicol. In Vitro
A.D. Ayrton, R.S. Sozio, and R.J. Chenery (1994). 17, 93-105.
Laurie acid as a model substrate for the simultaneous 658. Smith, S.V, A.P Koley, R. Dai, R.C. Robinson,
determination of cytochrome P450 2E1 and 4A in H. Leong, A. Markowitz et al. (2000).
hepatic microsomes. Chem. Res. Toxicol. 7, 836-842. Conformational modulation of human cytochrome
647. Castle, PL, J.L. Merdink, LR. Okita, P450 2E1 by ethanol and other substrates: A CO
S.A. Wrighton, and R.T. Okita (1995). Human flash photolysis study. Biochemistry 39, 5731-5737.
liver lauric acid hydroxylase activities. Drug 659. Yin, H., M.W. Anders, K.R. Korzekwa, L. Higgins,
Metab. Dispos. 23, 1037-1043. K.E. Thummel, E.D. Kharasch et al. (1995).
648. Gillam, E.M.L, A.M. Aguinaldo, L.M. Notley, Designing safer chemicals: Predicting the rates of
D. Kim, R.G. Mundkowski, A. Volkov et al. metabolism of halogenated alkanes. Proc. Natl.
(1999). Formation of indigo by recombinant mam- Acad. Sci. USA 92, 11076-11080.
malian cytochrome P450. Biochem. Biophys. Res. 660. Lewis, D.F, C. Sams, and G.D. Loizou (2003).
Commun. 265, 469-472. A quantitative structure-activity relationship
649. Adachi, J., Y. Mori, S. Matsui, H. Takigami, analysis on a series of alkyl benzenes metabolized
L Fujino, H. Kitagawa et al. (2001). Indirubin and by human cytochrome P450 2E1. J. Biochem. Mol.
indigo are potent aryl hydrocarbon receptor lig- Toxicol. 17, 47-52.
ands present in human urine. J. Biol. Chem. 276, 661. Keefer, L.K., W. Lijinsky, and H. Garcia (1973).
31475-31478. Deuterium isotope effect on the carcinogenicity of
650. Spracklin, D.K., D.C. Hankins, J.M. Fisher, dimethylnitrosamine in rat liver. J. Natl. Cancer
K.E. Thummel, and E.D. KJiarasch (1997). Inst. 51, 299-302.
Cytochrome P450 2E1 is the principal catalyst of 662. Wade, D., C.S. Yang, C.J. Metral, LM. Roman,
human oxidative halothane metabolism in vitro. J.A. Hrabie, C.W. Riggs et al. (1987). Deuterium
J. Pharmacol. Exp. Ther 281, 400-411. isotope effect on denitrosation and demethylation
651. Kharasch, E.D., D.C. Hankins, and K. Cox (1999). of A^-nitrosodimethylamine by rat liver micro-
Clinical isoflurane metabolism by cytochrome somes. Cancer Res. 47, 3373-3377.
P450 2E\. Anesthesiology 90, 766-771. 663. Yang, C.S., H. Ishizaki, M. Lee, D. Wade, and
652. Bell, L.C. and FP Guengerich (1997). Oxidation A. Fadel (1991). Deuterium isotope effect in the
kinetics of ethanol by human cytochrome P450 interaction of A^-nitrosodimethylamine, ethanol,
2E1. Rate-limiting product release accounts for and related compounds with cytochrome
effects of isotopic hydrogen substitution and P-450IIE1. Chem. Res. Toxicol. 4, 408-413.
cytochrome b^ on steady-state kinetics. J. Biol. 664. Calcutt, W and FP Guengerich (2003). Kinetic
C/^ew. 272, 29643-29651. isotope effects in dialkylnitrosamine dealkylations
653. Yamazaki, H., M. Nakano, E.M.J. Gillam, catalyzed by human cytochrome P450s 2A6 and
L.C. Bell, RP Guengerich, andT. Shimada (1996). lEX.FASEBJ. 17,A1325.
Requirements for cytochrome b^ in the oxidation 665. Reitz, R.H., A. Mendrala, and FP Guengerich
of 7-ethoxycoumarin, chlorzoxazone, aniline, and (1989). In vitro metabolism of methylene chloride
A^-nitrosodimethylamine by recombinant cyto- in human and animal tissues: Use in physiologi-
chrome P450 2E1 and by human liver microsomes. cally-based pharmacokinetic models. Toxicol.
Biochem. Pharmacol. 52, 301-309. Appl. Pharmacol. 97, 230-246.
654. Cooper, M.T. and T.D. Porter (2001). Cytochrome 666. Pernecky, S.I, TD. Porter, and M.L Coon (1990).
b^ coexpression increases the CYP2E1-dependent Expression of rabbit cytochrome P-450IIE2 in
Human P450s 493

yeast and stabilization of the enzyme by 4- major antigen in autoantibody formation among
methylpyrazole. Biochem. Biophys. Res. Commun. alcoholics. Gastroenterology 111, 206-216.
Ill, 1331-1337. 679. Bourdi, M., W Chen, R.M. Peter, J.L. Martin,
667. Koop, D.R. (1990). Inhibition of ethanol-inducible J.T.M. Buters, S.D. Nelson et al. (1996). Human
cytochrome P450IIE1 by 3-amino-l, 2,4-triazole. cytochrome P450 2E1 is a major autoantigen asso-
Chem. Res. Toxicol. 3, 377-383. ciated with halothane hepatitis. Chem. Res.
668. Hultmark, D., K. Sundh, L. Johansson, and Toxicol. 9, 1159-1166.
E. Arrhenius (1979). Ethanol inhibition of vinyl 680. Ekstrom, G. and M. Ingelman-Sundberg (1989).
chloride metabolism in isolated rat hepatocytes. Rat liver microsomal NADPH-supported oxidase
Chem. Biol. Interact. 25, 1-6. activity and lipid peroxidation dependent on
669. Wong, L.C.K., J.M. Winston, C.B. Hong, and ethanol-inducible cytochrome P-450 (P-450IIE1).
H. Plotnick (1982). Carcinogenicity and toxicity of Biochem. Pharmacol. 38, 1313-1319.
1,2-dibromoethane in the rat. Toxicol Appl. 681. Nieto, N., S.L. Friedman, P. Greenwel, and
Pharmacol. 63, 155-165. A.I. Cederbaum (1999). CYP2E1-mediated oxida-
670. Kwak, M.K., S.G. Kim, J.Y. Kwak, R.F. Novak, tive stress induces collagen type I expression in rat
and N.D. Kim (1994). Inhibition of cytochrome hepatic stellate cells. Hepatology 30, 987-996.
P4502E1 expression by organosulfur compounds 682. Cederbaum, A.L, D. Wu, M. Mari, and J. Bai
allylsulfide, allylmercaptan and allylmethylsulfide (2001). CYP2E1-dependent toxicity and oxidative
in rats. Biochem. Pharmacol. 47, 531-539. stress in HepG2 cells. Free Radic. Biol. Med. 31,
671. Nakajima, M., R. Yoshida, N. Shimada, 1539-1543.
H. Yamazaki, and T. Yokoi (2001). Inhibition and 683. Wan, J., L Shi, L. Hui, D. Wu, X. Jin, N. Zhao et al.
inactivation of human cytochrome P450 isoforms (2002). Association of genetic polymorphisms in
by phenethyl isothiocyanate. Drug Metab. Dispos. CYP2E1, MPO, NQOl, GSTMl, and GSTTl
29,1110-1113. genes with benzene poisoning. Environ. Health
672. Lucas, D., C. Farez, L.G. Bardou, J. Vaisse, Perspect. 110, 1213-1218.
J.R. Attali, and R Valensi (1998). Cytochrome 684. Uematsu, R, H. Kikuchi, M. Motomiya, T. Abe,
P450 2E1 activity in diabetic and obese patients I. Sagami, T. Ohmachi et al. (1991). Association
as assessed by chlorzoxazone hydroxylation. between restriction fragment length polymorphism
Fundam. Clin. Pharmacol. 12, 553-558. of the human cytochrome P450IIE1 gene and sus-
673. Le Marchand, L., G.R. Wilkinson, and L.R. Wilkens ceptibility to lung cancer. Jpn. J. Cancer Res. 82,
(1999). Genetic and dietary predictors of CYP2E1 254-256.
activity: A phenotyping study in Hawaii Japanese 685. Hirvonen, A., K. Husgafvel-Pursiainen, S. Anttila,
using chlorzoxazone. Cancer Epidemiol. Biomarkers A. Karjalainen, M. Sorsa, and H. Vainio (1992).
Prev. 8, 495-500. Metabolic cytochrome P450 genotypes and assess-
674. Morimoto, M., A.L. Hagbjork, A.A. Nanji, ment of individual susceptibility to lung cancer.
M. Ingelman-Sundberg, K.O. Lindros, P.C. Fu et al. Pharmacogenetics 2, 259-263.
(1993). Role of cytochrome P4502E1 in alcoholic 686. Ingelman-Sundberg, M., I. Johansson, I. Persson,
liver disease pathogenesis. Alcohol 10, 459^64. Q.Y Yue, M.L. Dahl, L. Bertilsson et al. (1992).
675. Morgan, K., S.W. French, andT.R. Morgan (2002). Genetic polymorphism of cytochromes P450:
Production of a cytochrome P450 2E1 transgenic Interethnic differences and relationship to inci-
mouse and initial evaluation of alcoholic liver dence of lung cancer. Pharmacogenetics 2,
damage. Hepatology 36, 122-134. 264-271.
676. Koop, D.R., B. Klopfenstein, Y. limuro, and 687. Persson, L, I. Johansson, H. Bergling, M.L. Dahl,
R.G. Thurman (1997). Gadolinium chloride blocks J., Seidegard, R. Rylander et al. (1993). Genetic
alcohol-dependent liver toxicity in rats treated polymorphism of cytochrome P4502E1 in a
chronically with intragastric alcohol despite the Swedish population: Relationship to incidence of
induction of CYP2E1. Mol. Pharmacol. 51, lung cancer. FEBS Lett. 319, 207-211.
944-950. 688. London, S.J., A.K. Daly, J. Cooper, C.L. Carpenter,
677. Lytton, S.D., A. Helander, Z.Q. Zhang-Gouillon, WC. Navidi, L. Ding et al. (1996). Lung cancer risk
K. Stokkeland, R. Bordone, S. Arico et al. (1999). in relation to the CYP2E1 Rsal genetic polymor-
Autoantibodies against cytochromes P-4502E1 phism among African-Americans and Caucasians
and P-4503A in alcoholics. Mol. Pharmacol. 55, in Los Angeles County. Pharmacogenetics 6,
223-233. 151-158.
678. Clot, P., E. Albano, E. Eliasson, M. Tabone, 689. Kato, S., PG. Shields, N.E. Caporaso,
S. Arico, Y. Israel et al. (1996). Cytochrome H. Sugimura, G.E. Trivers, M.A. Tucker et al
P4502E1 hydroxyethyl radical adducts as the (1994). Analysis of cytochrome P450 2E1 genetic
494 F. Peter Guengerich

polymorphisms in relation to human lung cancer. 700. Wu, S., C.R. Moomaw, K.B. Tomer, J.R. Falck, and
Cancer Epidemiol. Biomarkers Prev. 3, 515-518. D.C. Zeldin (1996). Molecular cloning and expres-
690. Itoga, S., F. Nomura, Y. Makino, T. Tomonaga, sion of CYP2J2, a human cytochrome P450
H. Shimada, T. Ochiai et al. (2002). Tandem repeat arachidonic acid epoxygenase highly expressed in
polymorphism of the CYP2E1 gene: An associa- heart. J. Biol. Chem. Ill, 3460-3468.
tion study with esophageal cancer and lung cancer 701. Zeldin, D.C, J. Foley, J. Ma, IE. Boyle,
Alcohol. Clin. Exp. Res. 26, 15S-19S. J.M.S. Pascual, C.R. Moomaw et al (1996). CYP2J
691. Bouchardy, C , A. Hirvonen, C. Coutelle, P.J. Ward, subfamily P450s in the lung: Expression, localiza-
P. Dayer, and S. Benhamou (2000). Role of alcohol tion, and potential functional significance. Mol
dehydrogenase 3 and cytochrome P-4502E1 geno- Pharmacol. SQ, 1111-1117.
types in susceptibility to cancers of the upper 702. Zeldin, D.C, J. Foley, S.M. Goldsworthy,
aerodigestive tract. Int. J. Cancer 87, 734-740. M.E. Cook, J.E. Boyle, J. Ma et al. (1997). CYP2J
692. Liu, S., J.Y. Park, S.P Schantz, J.C. Stern, and subfamily cytochrome P450s in the gastrointesti-
P Lazarus (2001). Elucidation of CYP2E1 5' reg- nal tract: Expression, localization, and potential
ulatory RsaVPstl allelic variants and their role in functional significance. Mol. Pharmacol. 51,
risk for oral cancer Oral Oncol. 37, 437^45. 931-943.
693. Kato, S., M. Onda, N. Matsukura, A. Tokunaga, 703. Zeldin, D.C. (2001). Epoxygenase pathways of
T. Tajiri, D.Y Kim et al. (1995). Cytochrome arachidonic acid metabolism. J. Biol. Chem. 276,
P4502E1 (CYP2E1) genetic polymorphism in a 36059-36062.
case-control study of gastric cancer and liver dis- 704. King, L.M., J. Ma, S. Srettabunjong, J. Graves, J.A.
ease. Pharmacogenetics 5, S141-S144. Bradbury, L. Li et al. (2002). Cloning of CYP2J2
694. Wong, R.H., C.L. Du, J.D. Wang, C.C. Chan, gene and identification of functional polymor-
J.C. Luo, and T.J. Cheng (2002). XRCCl and phisms. Mol. Pharmacol. 61, 840-852.
CYP2E1 polymorphisms as susceptibility factors 705. Nelson, D.R. (2003). Comparison of P450s from
of plasma mutant p53 protein and anti-p53 anti- human and fugu: 420 million years of vertebrate
body expression in vinyl chloride monomer- P450 evolution. Arch. Biochem. Biophys. 409,
exposed polyvinyl chloride workers. Cancer 18-24.
Epidemiol. Biomarkers Prev. 11, 475^82. 706. Rylander, T, E.P.A. Neve, M. Ingelman-Sundberg,
695. Nhamburo, PT, S. Kimura, O.W. McBride, and M. Oscarson (2001). Identification and tissue
C.A. Kozak, H.V Gelboin, and F.J. Gonzalez (1990). distribution of the novel human cytochrome P450
The human CYP2F gene subfamily: Identification 2S1 (CYP2S1). Biochem. Biophys. Res. Commun.
of a cDNA encoding a new cytochrome P450, 281,529-535.
cDNA-directed expression, and chromosome 707. Molowa, D.T, E.G. Schuetz, S.A. Wrighton,
mapping. Biochemistry 29, 5491-5499. PB. Watkins, P. Kremers, G. Mendez-Picon et al.
696. Czerwinski, M., TL. McLemore, R.M. Philpot, (1986). Complete cDNA sequence of a
PT. Nhamburo, K. Korzekwa, H.V Gelboin et al. cytochrome P-450 inducible by glucocorticoids in
(1991). Metabolic activation of 4-ipomeanol by human liver Proc. Natl. Acad. Sci. USA 83,
complementary DNA-expressed human cytochro- 5311-5315.
mes P-450: Evidence for species-specific metabo- 708. Beaune, PH., D.R. Umbenhauer, R.W. Bork,
lism. Cancer Res. 51, 4636-4638. R.S. Lloyd, and FP Guengerich (1986). Isolation
697. Lanza, D.L., E. Code, C.L. Crespi, F.J. Gonzalez, and sequence determination of a cDNA clone
and G.S. Yost (1999). Specific dehydrogenation of related to human cytochrome P-450 nifedipine oxi-
3-methylindole and epoxidation of naphthalene dase. Proc. Natl. Acad. Sci. USA 83, 8064-8068.
by recombinant human CYP2F1 expressed in 709. Bork, R.W, T Muto, PH. Beaune, PK. Srivastava,
lymphoblastoid cells. Drug Metab. Dispos. 27, R.S. Lloyd, and FP Guengerich (1989).
798-803. Characterization of mRNA species related to
698. Nakajima, T, E. Elovaara, F.J. Gonzalez, human liver cytochrome P-450 nifedipine oxidase
H.V Gelboin, H. Raunio, O. Pelkonen et al. and the regulation of catalytic activity. J. Biol.
(1994). Styrene metabolism by cDNA-expressed Chem. 264, 9\0~9\9.
human hepatic and pulmonary cytochromes P450. 710. Kolars, J., P Schmiedelin-Ren, W Dobbins,
Chem. Res. Toxicol. 7, 891-896. R. Merion, S. Wrighton, and P Watkins (1990).
699. Carr, B.A., J. Wan, R.N. Hines, and G.S. Yost Heterogeneity of P-450 III A expression in human
(2003). Characterization of the human lung gut epithelia. FASEB J. 4, A2242.
CYP2F1 gene and identification of a novel lung- 711. Kolars, J.C, P Schmiedlin-Ren, J.D. Schuetz,
specific binding motif. J. Biol. Chem. 278, C Fang, and PB. Watkins (1992). Identification of
15473-15483. rifampin-inducible P450IIIA4 (CYP3A4) in
Human P450s 495

human small bowel enterocytes. J. Clin. Invest. 90, 723. Hughes, S.J., M.A. Morse, CM. Weghorst,
1871-1878. H. Kim, RB. Watkins, RR Guengerich et al.
712. Bocker, R.H. and F.R Guengerich (1986). (1999). Cytochromes P450 are expressed in prolif-
Oxidation of 4-aryl- and 4-alkyl-substituted erating cells in Barrett's metaplasia. Neoplasia 1,
2,6-dimethyl-3,5-bis(alkoxycarbonyl)-l,4-dihy- 145-153.
dropyridines by human liver microsomes and 724. Inoue, K., J. Inazawa, H. Nakagawa, T. Shimada,
immunochemical evidence for the involvement H. Yamazaki, RR Guengerich et al. (1992).
of a form of C3^ochrome P-450. J. Med. Chem. 29, Assignment of the human cytochrome P450
1596-1603. nifedipine oxidase gene (CYP3A4) to band 7q22.1
713. Waxman, D.J., C. Attisano, F.R Guengerich, and by in situ fluorescence hybridization. Jpn. J. Hum.
D.R Lapenson (1988). Cytochrome P-450 steroid Genet. 37, 133-138.
hormone metabolism catalyzed by human liver 725. Lu, A.Y.H., A. Somogyi, S. West, R. Kuntzman,
microsomes. Arch. Biochem. Biophys. 263, and A.H. Conney (1972). Pregnenolone-16a-
424-436. carbonitrile: A new type of inducer of drug-metab-
714. Shimada, T. and RR Guengerich (1989). Evidence olizing enzymes. Arch. Biochem. Biophys. 152,
for C3^ochrome P-450^p, the nifedipine oxidase, 457-462.
being the principal enzyme involved in the bioacti- 726. Guengerich, RR (1999). Human cytochrome P-450
vation of aflatoxins in human liver. Proc. Natl. 3A4: Regulation and role in drug metabolism.
Acad. Sci. USA 86, A62-A65. Annu. Rev. Pharmacol. Toxicol. 39, 1-17.
715. Brian, W.R., M.-A. Sari, M. Iwasaki, T. Shimada, 727. Barwick, J.L., L.C. Quattrochi, A.S. Mills,
L.S. Kaminsky, and RR Guengerich (1990). C. Potenza, R.H. Tukey, and PS. Guzelian (1996).
Catalytic activities of human liver cytochrome Trans-species gene transfer for analysis of gluco-
P-450 IIIA4 expressed in Saccharomyces cere- corticoid-inducible transcriptional activation of
visiae. Biochemistry 29, 11280-11292. transiently expressed human CYP3A4 and rabbit
716. Guengerich, RR (1990). Mechanism-based inacti- CYP3A6 in primary cultures of adult rat and rabbit
vation of human liver cytochrome P-450 IIIA4 by hepatocytes. Mol. Pharmacol. 50, 10-16.
gestodene. Chem. Res. Toxicol. 3, 363-371. 728. Schuetz, E.G. and PS. Guzelian (1984). Induction
717. Kelley, J.D., D.L. Eaton, RR Guengerich, and of cytochrome P-450 by glucocorticoids in rat
R.A. Coulombe, Jr. (1997). Aflatoxin Bj activation liver: II. Evidence that glucocorticoids regulate
in human lung. Toxicol. Appl. Pharmacol. 144, induction of cytochrome P-450 by a nonclassical
88-95. receptor mechanism. J. Biol. Chem. 259,
718. Koch, I., R. Weil, R. Wolbold, J. Brockmoller, 2007-2012.
E. Hustert, O. Burk et al (2002). Interindividual 729. Calleja, C , J.M. Pascussi, J.C. Mani, R Maurel,
variability and tissue-specificity in the expression and M.J. Vilarem (1998). The antibiotic rifampicin
of cytochrome P450 3A mRNA. Drug Metab. is a nonsteroidal ligand and activator of the human
Dispos. 30, 1108-1114. glucocorticoid receptor. Nat. Med. 4, 92-96.
719. Nakamoto, T., I. Hase, S. Imaoka, T. Hiroi, Y. Oda, 730. Ray, D.W., A.M. Lovering, J.R. Davis, and
A. Asada et al. (2000). Quantitative RT-PCR for A. White (1998). Rifampicin: A glucocorticoid
CYP3A4 mRNA in human peripheral lympho- receptor ligand?. Nat. Med. 4, 1090-1091.
cytes: Induction of CYP3A4 in lymphocytes and 731. Kliewer, S.A., J.T. Moore, L. Wade,
in liver by rifampicin. Pharmacogenetics 10, J.L. Staudinger, M.A. Watson, S.A. Jones et al.
571-575. (1998). An orphan nuclear receptor activated by
720. de Wildt, S.N., G.L. Kearns, J.S. Leeder, and pregnanes defines a novel steroid signaling path-
J.N. van den Anker (1999). Cytochrome P450 way. Cell 92, 73-82.
3A. Ontogeny and drug disposition. Clin. 732. Lehmann, J.M., D.D. McKee, M.A. Watson,
Pharmacokinet. 37, 485-505. TM. Wilson, J.T Moore, and S.A. Kliewer (1998).
721. El Mouelhi, M., M.S. Didolkar, E.G. Elias, The human orphan nuclear receptor PXR is activated
RR Guengerich, and EC. Kauffman (1987). by compounds that regulate CYP3A4 gene expres-
Hepatic drug metabolizing enzymes in primary sion and cause drug interactions. J. Clin. Invest. 102,
and secondary tumors of human liver. Cancer Res. 1016-1023.
47, 460-466. 733. Coumoul, X., M. Diry, and R. Barouki (2002).
722. Fujitaka, K., T. Oguri, T. Isobe, Y Fujiwara, and PXR-dependent induction of human CYP3A4
N. Kohno (2001). Induction of cytochrome P450 gene expression by organochlorine pesticides.
3A4 by docetaxel in peripheral mononuclear cells Biochem. Pharmacol. 64, 1513-1519.
and its expression in lung cancer. Cancer 734. Jones, S.A., L.B. Moore, G.B. Wisely, and
Chemother. Pharmacol. 48, 4 2 ^ 6 . S.A. Kliewer (2002). Use of in vitro pregnane X
496 F. Peter Guengerich

receptor assays to assess CYP3A4 induction CAR-mediated xenobiotic induction of CYP3A4.


potential of drug candidates. Meth. Enzymol. 357, Nat. Med. 9, 220-224.
161-170. 746. Jover, R., R. Bort, M.J. Gomez-Lechon, and
735. Raucy, J., L. Warfe, M.F. Yueh, and S.W. Allen J.V Castell (2002). Down-regulation of human
(2002). A cell-based reporter gene assay for deter- CYP3A4 by the inflammatory signal interleukin-6:
mining induction of CYP3A4 in a high-volume Molecular mechanism and transcription factors
system. J. Pharmacol. Exp. Then 303, 412-423. 'mwo\\Q&. FASEB J. 16, 1799-1801.
736. Luo, G., M, Cunningham, S. Kim, T. Burn, J. Lin, 747. Danan, G., V Descatoire, and D. Pessayre (1981).
M. Sinz et al. (2002). CYP3A4 induction by drugs: Self-induction by erythromycin of its own trans-
Correlation between a pregnane X receptor reporter formation into a metabolite forming an inactive
gene assay and CYP3A4 expression in human complex with reduced cytochrome P-450.
hepatocytes. Drug Metab. Dispos. 30, 795-804. J. Pharmacol. Exp. Ther 218, 509-514.
737. Zhang, J., P Kuehl, E.D. Green, J.W. Touchman, 748. Bensoussan, C , M. Delaforge, and D. Mansuy
PB. Watkins, A. Daly et al. (2001). The human (1995). Particular ability of cytochromes P450 3 A
pregnane X receptor: Genomic structure and iden- to form inhibitory P450-iron-metabolite com-
tification and functional characterization of natural plexes upon metabolic oxidation of amino drugs.
alleHc variants. Pharmacogenetics 11, 555-572. Biochem. Pharmacol. 49, 591-602.
738. Goodwin, B., E. Hodgson, and C. Liddle (1999). 749. Wang, X.Y, K.F. Medzihradszky, D. Maltby, and
The orphan human pregnane X receptor mediates M.A. Correia (2001). Phosphorylation of native
the transcriptional activation of CYP5A4 by and heme-modified CYP3A4 by protein kinase C:
rifampicin through a distal enhancer module. Mol. A mass spectrometric characterization of the phos-
Pharmacol. 56, 1329-1339. phorylated peptides. Biochemistry 40,
739. El-Sankary, W., V Bombail, G.G. Gibson, and 11318-11326.
N. Plant (2002). Glucocorticoid-mediated induc- 750. Sy, S.K., A. Ciaccia, W. Li, E.A. Roberts, A. Okey,
tion of CYP3A4 is decreased by disruption of a W. Kalow et al. (2002). Modeling of human
protein: DNA interaction distinct from the preg- hepatic CYP3A4 enzyme kinetics, protein, and
nane X receptor response element. Drug Metab. mRNA indicates deviation from log-normal distri-
Dispos. 30, 1029-1034. bution in CYP3A4 gene expression. Eur J. Clin.
740. Takeshita, A., M. Taguchi, N. Koibuchi, and Pharmacol. 58, 357-365.
Y. Ozawa (2002). Putative role of the orphan 751. Ball, S.E., J. Scatina, J. Kao, G.M. Ferron, R.
nuclear receptor SXR (steroid and xenobiotic Fruncillo, P. Mayer et al. (1999). Population distri-
receptor) in the mechanism of CYP3A4 inhibition bution and effects on drug metaboHsm of a genetic
by xenobiotics. J. Biol. Chem. Ill, 32453-32458. variant in the 5' promoter region of CYP3A4. Clin.
741. Goodwin, B., E. Hodgson, D.J. D'Costa, Pharmacol. Ther 66, 288-294.
G.R. Robertson, and C. Liddle (2002). 752. Spurdle, A.B., B. Goodwin, E. Hodgson,
Transcriptional regulation of the human CYP3A4 J.L. Hopper, X. Chen, DM. Purdie et al. (2002). The
gene by the constitutive androstane receptor. Mol. CYP3A4*1B polymorphism has no functional sig-
Pharmacol. 62, 359-365. nificance and is not associated with risk of breast or
742. Thummel, K.E., C. Brimer, K. Yasuda, ovarian cancer. Pharmacogenetics 12, 355-366.
J. Thottassery, T. Senn, Y. Lin et al. (2001). 753. Eiselt, R., T.L. Domanski, A. Zibat, R. Mueller,
Transcriptional control of intestinal cytochrome E. Presecan-Siedel, E. Hustert et al. (2001).
P-4503A by la,25-dihydroxy vitamin D3. Mol. Identification and functional characterization of
Pharmacol. 60, 1399-1406. eight CYP3A4 protein variants. Pharmacogenetics
743. Hara, H., Y Yasunami, and T. Adachi (2002). 11, 447-458.
Alteration of cellular phosphorylation state affects 754. Garcia-Martin, E., C. Martinez, R.M. Pizarro,
vitamin D receptor-mediated CYP3A4 mRNA F.J. Garcia-Gamito, H. GuUsten, H. Raunio et al.
induction in Caco-2 cells. Biochem. Biophys. Res. (2002). CYP3A4 variant alleles in white individu-
Commun. 296, 182-188. als with low CYP3A4 enzyme activity. Clin.
744. Ourlin, J.C, Y. Jounaidi, P. Maurel, and Pharmacol. Ther 71, 196-204.
M.J. Vilarem (1997). Role of the liver-enriched 755. Lamba, J.K., YS. Lin, K. Thummel, A. Daly,
transcription factors C/EBPa and DBP in the PB. Watkins, S. Strom et al. (2002). Common allelic
expression of human CYP3A4 and CYP3A7. variants of cytochrome P4503A4 and their preva-
J. Hepatol. 26(Suppl 2), 54-62. lence in different populations. Pharmacogenetics
745. Tirona, R.G., W. Lee, B.F. Leake, L.B. Lan, 12, 121-132.
C.B. Cline, V Lamba et al. (2003). The orphan 756. Dai, D, J. Tang, R. Rose, E. Hodgson,
nuclear receptor HNF4a determines PXR- and R.J. Bienstock, H.W Mohrenweiser et al. (2001).
Human P450s 497

Identification of variants of CYP3A4 and charac- 766. Zhang, Z., Y. Li, R.A. Stearns, RR. Ortiz De
terization of their abihties to metaboHze testos- Montellano, T.A. Baillie, and W. Tang (2002).
terone and chlorpyrifos. J. Pharmacol. Exp. Ther. Cytochrome P450 3A4-mediated oxidative conver-
299,825-831. sion of a cyano to an amide group in the metabo-
757. Lamba, J.K., Y.S. Lin, E.G. Schuetz, and lism of pinacidil. Biochemistry 41, 2712-2718.
K.E. Thummel (2002). Genetic contribution to 767. Hosea, N.A. and FR Guengerich (1998).
variable human CYPSA-mediated metabolism. Oxidation of non-ionic detergents by cytochrome
Adv. DrugDeliv. Rev. 54, 1271-1294. P450 enzymes. Arch. Biochem. Biophys. 353,
758. Williams, J.A., B.J. Ring, VE. Cantrell, D.R. Jones, 365-373.
J. Eckstein, K. Ruterbories et al. (2002). 768. Bodin, K., L. Bretillon, Y Aden, L. Bertilsson,
Comparative metabolic capabilities of CYP3A4, U. Broome, C. Einarsson, and U. Diczfalusy
CYP3A5, and CYP3A7. Drug Metab. Dispos. 30, (2001). Antiepileptic drugs increase plasma levels
883-891. of 4(3-hydroxycholesterol in humans: Evidence for
759. Wang, R.W., RH. Kari, A.Y.H. Lu, RE. Thomas, involvement of cytochrome P450 3A4. J. Biol.
ER Guengerich, and K.R Vyas (1991). Chem. 276, 38685-38689.
Biotransformation of lovastatin. IV Identification 769. Bodin, K., U. Andersson, E. Rystedt, E. Ellis,
of cytochrome P-450 3A proteins as the major M. Norlin, I. Pikuleva et al. (2002). Metabolism of
enzymes responsible for the oxidative metabolism 4(3-hydroxycholesterol in humans. J. Biol. Chem.
of lovastatin in rat and human liver microsomes. 277,31534-31540.
Arch. Biochem. Biophys. 290, 355-361. 770. Furster, C. and K. Wikvall (1999). Identification of
760. Huskey, S.E.W, D.C. Dean, R.R. Miller, CYP3A4 as the major enzyme responsible for 25-
G.H. Rasmusson, and S.H.L. Chiu (1995). hydroxylation of 5p-cholestane-3a,7a,12a-triol in
Identification of human cytochrome P450 isozymes human liver microsomes. Biochim. Biophys. Acta
responsible for the in vitro oxidative metabolism of 1437, 46-52.
finasteride. Drug Metab. Dispos. 23, 1126-1135. 771. Honda, A., G. Salen, Y Matsuzaki, A.K. Batta,
761. Kronbach, T., V Fischer, and U.A. Meyer (1988). G. Xu, E. Leitersdorf et al. (2001). Side chain
Cyclosporine metabolism in human liver: hydroxylations in bile acid biosynthesis catalyzed
Identification of a cytochrome P-450III gene fam- by CYP3A are markedly up-regulated in Cyp27~^~
ily as the major cyclosporine-metabolizing enzyme mice but not in cerebrotendinous xanthomatosis.
explains interactions of cyclosporine with other J. Biol. Chem. 276, 34579-34585.
drugs. Clin. Pharmacol. Ther. 43, 630-635. 772. Dussault, I., H.D. Yoo, M. Lin, E. Wang, M. Fan,
762. Combalbert, J., I. Fabre, G. Fabre, I. Dalet, A.K. Batta et al. (2003). Identification of an
J. Derancoiirt, J.R Cano et al. (1989). Metabolism endogenous ligand that activates pregnane X
of cyclosporin A. IV Purification and identifica- receptor-mediated sterol clearance. Proc. Natl.
tion of the rifampicin-inducible human liver Acad. Sci. USA 100, 833-838.
cytochrome P-450 (cyclosporin A oxidase) as a 773. Boocock, D.J., K. Brown, A.H. Gibbs, E. Sanchez,
product of P450IIIA gene subfamily. Drug Metab. K.W. Turteltaub, and I.N. White (2002).
Dispos. 17, 197-207. Identification of human CYP forms involved in the
763. Koudriakova, T., E. latsimirskaia, I. Utkin, activatidn of tamoxifen and irreversible binding to
E. Gangl, R Vouros, E. Storozhuk et al. (1998). DNA. Carcinogenesis 23, 1897-1901.
Metabolism of the human immunodeficiency virus 774. Ueng, Y.-F, T. Shimada, H. Yamazaki, and
protease inhibitors indinavir and ritonavir by ER Guengerich (1995). Oxidation of aflatoxin B^
human intestinal microsomes and expressed by bacterial recombinant human cytochrome P450
cytochrome P4503A4/3A5: Mechanism-based enzymes. Chem. Res. Toxicol. 8, 218-225.
inactivation of cytochrome P4503A by ritonavir. 775. Iyer, R., B. Coles, K.D. Raney, R. Thier,
Drug Metab. Dispos. 26, 552-561. ER Guengerich, and T.M. Harris (1994). DNA
764. Warrington, J.S., R.I. Shader, L.L. von Moltke, and adduction by the potent carcinogen aflatoxin B^:
D.J. Greenblatt (2000). In vitro biotransformation Mechanistic studies. J. Am. Chem. Soc.116,
of sildenafil (Viagra): Identification of human 1603-1609.
cytochromes and potential drug interactions. Drug 776. Crespi, C.L., VR Miller, and B.W Penman (1997).
Metab. Dispos. 28, 392-397. Microtiter plate assays for inhibition of human,
765. Kudo, S., M.G. Okumura, and T. Ishizaki (1999). drug-metabolizing cytochromes P450. Anal.
Cytochrome P-450 isoforms involved in car- Biochem. 248, 188-190.
boxylic acid ester cleavage of Hantzsch pyridine 777. Stresser, D.M., S.D. Turner, A.R Blanchard,
ester of pranidipine. Drug Metab. Dispos. 27, VR Miller, and C.L. Crespi (2002). Cytochrome
303-308. P450 fluorometric substrates: Identification of
498 F. Peter Guengerich

isoform-selective probes for rat CYP2D2 and human 789. Krivoruk, Y, M.T. Kinirons, A.J.J. Wood, and
CYP3A4. DrugMetab. Dispos. 30, 845-852. M. Wood (1994). Metabohsm of cytochrome
778. Chauret, N., N. Tremblay, R.L. Lackman, P4503A substrates in vivo administered by the
J.-Y. Gauthier, J.M. Silva, J. Marois et al (1999). same route: Lack of correlation between alfentanil
Description of a 96-well plate assay to measure clearance and erythromycin breath test. Clin.
cytochrome P4503A inhibition in human liver Pharmacol. Ther 56, 608-614.
microsomes using a selective fluorescent probe. 790. Kivisto, K.T and H.K. Kroemer (1997). Use of
Anal. Biochem. 276, 215-226. probe drugs as predictors of drug metabolism in
779. Kenworthy, K.E., J.C. Bloomer, S.E. Clarke, and humans. J. Clin. Pharmacol. 37, 40S-48S.
J.B. Houston (1999). CYP3A4 drug interactions: 791. Thummel, K.E., D. O'Shea, M.K Paine, D.D. Shen,
Correlation of 10 in vitro probe substrates. Br. J. K.L. Kunze, J.D. Perkins et al. (1996). Oral first-
Clin. Pharmacol. 48, 716-727. pass elimination of midazolam involves both
780. Schellens, J.H.M., PA. Soons, and D.D. Breimer gastrointestinal and hepatic CYP3A-mediated
(1988). Lack of bimodality in nifedipine plasma metabolism. Clin. Pharmacol. Ther 59, 491-502.
kinetics in a large population of healthy subjects. 792. Paine, M.K, D.A. Wagner, K.A. Hoffmaster, and
Biochem. Pharmacol. 37, 2507-2510. PB. Watkins (2002). Cytochrome P450 3A4 and
781. Lown, K., J. Kolars, K. Turgeon, R. Merion, P-glycoprotein mediate the interaction between an
S.A. Wrighton, and PB. Watkins (1992). The oral erythromycin breath test and rifampin. Clin.
erythromycin breath test selectively measures Pharmacol. Ther 72, 524-535.
P450IIIA in patients with severe liver disease. 793. Schuetz, E.G., WT. Beck, and J.D. Schuetz (1996).
Clin. Pharmacol. Ther 51, 229-238. Modulators and substrates of P-glycoprotein and
782. Gremse, D.A., H.H. A-Kader, T.J. Schroeder, and cytochrome P4503A coordinately up-regulate
W.F. BaHstreri (1990). Assessment of lidocaine these proteins in human colon carcinoma cells.
metabolite formation as a quantitative hver func- Mol. Pharmacol. 49, 311-318.
tion test in children. Hepatology 12, 565-569. 794. Gillam, E.M.J., T Baba, B.-R. Kim, S. Ohmori,
783. May, D.G., J. Porter, G.R. Wilkinson, and and F.P Guengerich (1993). Expression of modi-
R.A. Branch (1994). Frequency distribution of fied human cytochrome P450 3A4 in Escherichia
dapsone yV-hydroxylase, a putative probe for P450 coli and purification and reconstitution of the
3A4 activity, in a white population. Clin. QX\zymQ. Arch. Biochem. Biophys. 305, 123-131.
Pharmacol. Ther 55, 492-500. 795. Imaoka, S., Y. Imai, T. Shimada, and Y Funae
784. Thummel, K.E., D.D. Shen, R.L. Carithers, Jr., (1992). Role of phospholipids in reconstituted
P Hartwell, TD. Podoll, W.F. Trager et al. (1993). cytochrome P450 3A forms and mechanism of
Prediction of in vivo midazolam clearance from their activation of catalytic activity. Biochemistry
hepatic CYP3A content and midazolam 1 '-hydrox- 31, 6063-6069.
ylation activity in liver transplant patients. In 796. Ingelman-Sundberg, M., A.-L. Hagbjork, Y.-F. Ueng,
Abstracts, 5th North American ISSX Meeting, Vol. H. Yamazaki, and F.P Guengerich (1996). High rates
4. p. 235, 17-21 October, Tucson, AZ. of substrate hydroxylation by human cytochrome
785. Wanwimolruk, S., M.F Paine, S.N. Pusek, and P450 3A4 in reconstituted membranous vesicles:
PB. Watkins (2002). Is quinine a suitable probe to Influence of membrane charge. Biochem. Biophys.
assess the hepatic drug-metabolizing enzyme Res. Commun. 221, 318-322.
CYP3A4? Br J. Clin. Pharmacol. 54, 643-651. 797. Peyronneau, M.A., J.P Renaud, G. Truan, P. Urban,
786. Ged, C , J.M. Rouillon, L. Pichard, J. Combalbert, D Pompon, and D. Mansuy (1992). Optimization
N. Bressot, P. Bories et al. (1989). The increase in of yeast-expressed human liver cytochrome-P450
urinary excretion of 6P-hydroxycortisol as a 3A4 catalytic activities by coexpressing NADPH-
marker of human hepatic cytochrome P450IIIA cytochrome P450 reductase and cytochrome b^.
induction. Br J. Clin. Pharmacol. 28, 373-387. Eur J. Biochem. 207, 109-116.
787. Watkins, PB., T.A. Hamilton, T.M. Annesley, 798. Guengerich, FP and WW Johnson (1997).
C.N. Ellis, J.C. Kolars, and J.J. Voorhees (1990). Kinetics of ferric cytochrome P450 reduction by
The erythromycin breath test as a predictor of NADPH-cytochrome P450 reductase: Rapid
cyclosporine blood levels. Clin. Pharmacol. Ther reduction in absence of substrate and variations
48, 120-129. among cytochrome P450 systems. Biochemistry
788. Kinirons, M.T., D. Oshea, T.E. Downing, 36, 14741-14750.
A.T. Fitzwilliam, L. Joellenbeck, J.D. Groopman et 799. Shet, M.S., C.W Fisher, PL. Holmans, and
al. (1993). Absence of correlations among 3 puta- R.W. Estabrook (1993). Human cytochrome P450
tive in vivo probes of human cytochrome P4503 A 3A4: Enzymatic properties of a purified recombi-
activity in young healthy men. Clin. Pharmacol. nant fusion protein containing NADPH-P450 reduc-
Ther 54,621-629. tase. Proc. Natl. Acad. Sci. USA 90, 11748-11752.
Human P450s 499

800. Gillam, E.M.J., Z. Guo, Y.-F. Ueng, H. Yamazaki, CYP4A4 metabolism of prostaglandin E-1 and
I. Cock, P.E.B. Reilly et aL (1995). Expression of arachidonic acid. Arch. Biochem. Biophys. 393,
cytochrome P450 3A5 in Escherichia coli: Effects 329-338.
of 5' modifications, purification, spectral charac- 811. Guryev, O.L., A.A. Gilep, S.A. Usanov, and
terization, reconstitution conditions, and catalytic R.W. Estabrook (2001). Interaction of apo-
activities. Arch. Biochem. Biophys. 317, 374-384. cytochrome b^ with cytochromes P4503A4 and
801. Yamazaki, H., M. Nakano, Y. Imai, Y.-F. Ueng, P45017A: Relevance of heme transfer reactions.
EP. Guengerich, and T. Shimada (1996). Roles of Biochemistry 40, 5018-5031.
cytochrome b^ in the oxidation of testosterone and 812. Yamazaki, H., T. Shimada, M.V Martin, and
nifedipine by recombinant cytochrome P450 3A4 F.P. Guengerich (2001). Stimulation of cytochrome
and by human liver microsomes. Arch. Biochem. P450 reactions by apo-cytochrome b^ Evidence
Biophys. 325, 174-182. against transfer of heme from cytochrome P450
802. Yamazaki, H., M. Nakajima, M. Nakamura, 3A4 to apo-cytochrome b^ or heme oxygenase.
S. Asahi, N. Shimada, E.M.J. Gillam et al. (1999). J. Biol. Chem. 276, 30885-30891.
Enhancement of cytochrome P-450 3A4 catalytic 813. Ekins, S., G. Bravi, J.H. Wikel, and S.A. Wrighton
activities by cytochrome b^ in bacterial mem- (1999). Three-dimensional-quantitative structure
branes. DrugMetab. Dispos. 27, 999-1004. activity relationship analysis of cytochrome P-450
803. Parikh, A., E.M.J. Gillam, and ER Guengerich 3A4 substrates. J. Pharmacol. Exp. Ther 291,
(1997). Drug metabolism by Escherichia coli 424-433.
expressing human cytochromes P450. Nat. 814. Ekins, S., G. Bravi, S. Binkley, J.S. Gillespie,
Biotechnol. 15, 784-788. B.J. Ring, J.H. Wikel et al. (1999). Three- and
804. Blake, J.A.R., M. Pritchard, S. Ding, G.C.M. Smith, four-dimensional quantitative structure activity
B. Burchell, C.R. Wolfe/ al (1996). Coexpression relationship analyses of cytochrome P-450 3A4
of a human P450 (CYP3A4) and P450 reductase inhibitors. J. Pharmacol. Exp. Ther 290,
generates a highly functional monooxygenase sys- 429-438.
tem in Escherichia coli, FEBS Lett. 397, 210-214. 815. Stevens, J.C, TL. Domanski, G.R. Harlow,
805. Nakajima, M., K. Tane, S. Nakamura, N. Shimada, R.B. White, E. Orton, and J.R. Halpert (1999). Use
H. Yamazaki, and T. Yokoi (2002). Evaluation of of the steroid derivative RPR 106541 in combina-
approach to predict the contribution of multiple tion with site-directed mutagenesis for enhanced
cytochrome P450s in drug metabolism using relative cytochrome P-450 3A4 structure/function analysis.
activity factor: Effects of the differences in expres- J. Pharmacol. Exp. Ther 290, 594-602.
sion levels of NADPH-cytochrome P450 reductase 816. Domanski, T.L., YA. He, G.R. Harlow, and
and cytochrome b^ in the expression system and the J.R. Halpert (2000). Dual role of human
differences in the marker activities. J. Pharm. Sci. 91, cytochrome P450 3A4 residue Phe-304 in sub-
952-963. strate specificity and cooperativity. J. Pharmacol.
806. Noshiro, M. and T. Omura (1978). Immuno- Exp. Ther 293,585-591.
chemical study on the electron pathway from 817. Fowler, S.M., R.J. Riley, M.R Pritchard,
NADH to cytochrome P-450 of liver microsomes. M.J. Sutchffe, T Friedberg, and C.R. Wolf (2000).
J. Biochem. (Tokyo) 83, 61-77. Amino acid 305 determines catalytic center acces-
807. Perret, A. and D. Pompon (1998). Electron shuttle sibility in CYP3A4. Biochemistry 39, 4406-4414.
between membrane-bound cytochrome P450 3A4 818. Xue, L., H.F Wang, Q. Wang, G.D. Szklarz,
and 63 rules uncoupling mechanisms. Biochemistry T.L. Domanski, J.R. Halpert et al. (2001).
37, 11412-11424. Influence of P450 3A4 SRS-2 residues on cooper-
808. Yamazaki, H., W.W. Johnson, Y.-F Ueng, ativity and/or regioselectivity of aflatoxin Bj oxi-
T. Shimada, and F.P. Guengerich (1996). Lack of dation. Chem. Res. Toxicol. 14, 483-491.
electron transfer from cytochrome b^ in stimulation 819. Fowler, S.M., J.M. Taylor, T. Friedberg, C.R. Wolf,
of catalytic activities of cytochrome P450 3A4: and R.J. Riley (2002). CYP3A4 active site volume
Characterization of a reconstituted cytochrome modification by mutagenesis of leucine 211. Drug
P450 3A4/NADPH-cytochrome P450 reductase Metab. Dispos. 30, 452^56.
system and studies with apo-cytochrome b^. 820. Smith, D.A. and B.C. Jones (1992). Speculations
J. Biol. Chem. 271, 27438-27444. on the substrate structure-activity relationship
809. Auchus, R.J., T.C. Lee, and W.L. Miller (1998). (SSAR) of cytochrome P450 enzymes. Biochem.
Cytochrome b^ augments the 17,20-lyase activity Pharmacol. 44, 2089-2098.
of human P450cl7 without direct electron transfer. 821. Macdonald, T.L., WG. Gutheim, R.B. Martin, and
J. Biol. Chem. 273, 3158-3165. F.P Guengerich (1989). Oxidation of substituted
810. Aitken, A.E., L.J. Roman, PA, Loughran, M. de la A'.A^-dimethylanilines by cytochrome P-450:
Garza, and B.S.S. Masters (2001). Expressed Estimation of the effective oxidation-reduction
500 F. Peter Guengerich

potential of cytochrome P-450. Biochemistry 28, 834. Wang, R.W., D.J. Newton, T.D. Scheri, and
2071-2077. A.Y.H. Lu (1997). Human cytochrome P450 3A4-
822. Yamazaki, H., Y.-F. Ueng, T. Shimada, and catalyzed testosterone 6p-hydroxylation and eryth-
F.P. Guengerich (1995). Roles of divalent metal romycin A^-demethylation. Drug Metab. Dispos.
ions in oxidations catalyzed by recombinant 25, 502-507.
cytochrome P450 3A4 and replacement of 835. Ludwig, E., J. Schmid, K. Beschke, and T. Ebner
NADPH-cytochrome P450 reductase with other (1999). Activation of human cytochrome P-450
flavoproteins, iron-sulfur proteins, and oxygen sur- 3A4-catalyzed meloxicam 5'-methylhydroxylation
rogates. Biochemistry 34, 8380-8389. by quinidine and hydroquinidine in vitro.
823. Northrop, D.B. (1975). Steady-state analysis of J. Pharmacol Exp. Ther 290, 1-8.
kinetic isotope effects in enzymic reactions. 836. Wang, R.W, D.J. Newton, N. Liu, W.M. Atkins,
Biochemistry 14, 2644-2651. and A.Y.H. Lu (2000). Human cytochrome P-450
824. Kuby, S.A. (1991). A Study of Enzymes, Vol I, 3A4: In vitro drug-drug interaction patterns are
Enzyme Catalysis, Kinetics, and Substrate substrate-dependent. Drug Metab. Dispos. 28,
Binding. CRC Press, Boca Raton, FL. 360-366.
825. Walsh, C. (1979). Enzymatic Reaction Mechanisms. 837. Schrag, M.L. and L.C. Wienkers (2001). Covalent
W. H. Freeman Co, San Francisco, CA. alteration of the CYP3A4 active site: Evidence for
826. Obach, R.S. (2001). Mechanism of cytochrome multiple substrate binding domains. Arch.
P4503A4- and 2D6-catalyzed dehydrogenation of Biochem. Biophys. 391, 49-55.
ezlopitant as probed with isotope effects using five 838. Masubuchi, Y, A. Ose, and T. Horie (2002).
deuterated analogs. Drug Metab. Dispos. 29, Diclofenac-induced inactivation of CYP3A4 and
1599-1607. its stimulation by quinidine. Drug Metab. Dispos.
827. Wiebel, RJ., J.C. Leutz, L. Diamond, and 30, 1143-1148.
H.V Gelboin (1971). Aryl hydrocarbon 839. Shou, M., J. Grogan, J.A. Mancewicz, K.W. Krausz,
(benzo[«]pyrene) hydroxylase in microsomes from FJ. Gonzalez, H.V Gelboin et al (1994). Activation
rat tissues: Differential inhibition and stimulation of CYP3A4: Evidence for the simultaneous binding
by benzoflavones and organic solvents. Arch. of two substrates in a cytochrome P450 active site.
Biochem. Biophys. 144, 78-86. Biochemistry 33, 6450-6455.
828. Kapitulnik, J., PJ. Poppers, M.K. Buening, 840. Korzekwa, K.R., N. Krishnamachary, M. Shou,
J.G. Fortner, and A.H. Conney (1977). Activation A. Ogai, R.A. Parise, A.E. Rettie et al. (1998).
of monooxygenases in human liver by 7,8-ben- Evaluation of atypical cytochrome P450 kinetics
zoflavone. Clin. Pharmacol Ther 11, 475^85. with two-substrate models: Evidence that multiple
829. Buening, M.K., J.G. Fortner, A. Kappas, and substrates can simultaneously bind to cytochrome
A.H. Conney (1978). 7,8-Benzoflavone stimulates P450 active sites. Biochemistry 37, 4137^147.
the metabolic activation of aflatoxin Bj to muta- 841. Shou, M., R. Dai, D. Cui, K.R. Korzekwa,
gens by human liver. Biochem. Biophys. Res. T.A. Baillie, and T.H. Rushmore (2001). A kinetic
Commun. 82, 348-355. model for the metabolic interaction of two sub-
830. Schwab, G.E., J.L. Raucy, and E.F. Johnson strates at the active site of cytochrome P450 3A4.
(1988). Modulation of rabbit and human hepatic J. Biol Chem. 276, 2256-2262.
cytochrome P-450-catalyzed steroid hydroxyla- 842. Kenworthy K.E., S.E. Clarke, J. Andrews, and
tions by a-naphthoflavone. Mol Pharmacol 33, J.B. Houston (2001). Multisite kinetic models for
493-499. CYP3A4: Simultaneous activation and inhibition
831. Hosea, N.A., G.P Miller, and FP Guengerich (2000). of diazepam and testosterone metabolism. Drug
Elucidation of distinct binding sites for cytochrome Metab. Dispos. 29, 1644-1651.
P450 3A4. Biochemistry 39, 5929-5939. 843. Galetin, A., S.E. Clarke, and J.B. Houston (2002).
832. Guengerich, RP, B.-R. Kim, E.M.J. Gillam, and Quinidine and haloperidol as modifiers of
T. Shimada (1994). Mechanisms of enhancement CYP3A4 activity: Multisite kinetic model
and inhibition of cytochrome P450 catalytic activ- approach. Drug Metab. Dispos. 30, 1512-1522.
ity In M. C. Lechner (ed.). Proceedings, 8th 844. Domanski, T.L., J. Liu, G.R. Harlow, and
International Conference on Cytochrome P450: J.R. Halpert (1998). Analysis of four residues
Biochemistry, Biophysics, and Molecular Biology, within substrate recognition site 4 of human
John Libbey Eurotext, Paris, pp. 97-101. cytochrome P450 3A4: Role in steroid hydroxy-
833. Yun, C.-H., T. Shimada, and FP Guengerich (1992). lase activity and a-naphthoflavone stimulation.
Contributions of human liver cytochrome P-450 Arch. Biochem. Biophys. 350, 223-232.
enzymes to the N-oxidation of 4, 4'-methylene- 845. Harlow, G.R. and J.R. Halpert (1998).
bis(2-chloroaniline). Carcinogenesis 13, 217-222. Analysis of human cytochrome P450 3A4
Human P450s 501

cooperativity: Construction and characterization of protein site: A matter of pre-existing populations.


a site-directed mutant that displays hyperboHc Protein Sci. 11, 184-197.
steroid hydroxylation kinetics. Proc. Natl. Acad. 857. Carlson, H.A. (2002). Protein flexibility and drug
Sci. t/&4 95,6636-6641. design: How to hit a moving target. Curr Opin.
846. He, Y.A., F. Roussel, and J.R. Halpert (2003). Chem. Biol. 6, 447^52.
Analysis of homotropic and heterotropic coopera- 858. Carlson, H.A. (2002). Protein flexibility is an
tivity of diazepam oxidation by CYP3A4 using important component of structure-based drug dis-
site-directed mutagenesis and kinetic modeling. covery. Curr Pharm. Des. 8, 1571-1578.
Arch. Biochem. Biophys. 409, 92-101. 859. Schlichting, I., J. Berendzen, K. Chu, A.M. Stock,
847. Domanski, T.L., Y.A. He, K.K. Khan, F. Roussel, S.A. Maves, D.E. Benson et al (2000). The cat-
Q. Wang, and J.R. Halpert (2001). Phenylalanine alytic pathway of cytochrome P450^^j^ at atomic
and tryptophan scanning mutagenesis of CYP3A4 resolution. Science 287, 1615-1622.
substrate recognition site residues and effect 860. Atkins, W.M., R.W. Wang, and A.Y.H. Lu (2001).
on substrate oxidation and cooperativity. Allosteric behavior in cytochrome P450-dependent
Biochemistry ^^, 10150-10160. in vitro drug-drug interactions: A prospective
848. Dabrowski, M.J., M.L. Schrag, L.C. Wienkers, and based on conformational dynamics. Chem. Res.
W.M. Atkins (2002). Pyrene-pyrene complexes at Toxicol. 14, 338-347.
the active site of cytochrome P450 3A4: Evidence 861. Segel, I.H. (1975). Enzyme Kinetics. Behavior and
for a multiple substrate binding site. J. Am. Chem. Analysis of Rapid Equilibrium and Steady-State
Soc. 124, 11866-11867. Enzyme Systems. John Wiley & Sons, New York.
849. Cupp-Vickery, J., R. Anderson, and Z. Hatziris 862. Atkins, W.M., WD. Lu, and D.L. Cook (2002). Is
(2000). Crystal structures of ligand complexes of there a toxicological advantage for non-hyperbolic
P450eryF exhibiting homotropic cooperativity. kinetics in cytochrome P450 catalysis? Functional
Proc. Natl. Acad. Sci. USA 97, 3050-3055. allostery from "distributive catalysis." J. Biol.
850. Khan, K.K., H. Liu, and J.R. Halpert (2003). Chem. Ill, 33258-33266.
Homotropic versus heterotorpic cooperativity of 863. Di Petrillo, K., S. Wood, V Kostrubsky, K. Chatfield,
cytochrome P450eryF: A substrate oxidation and J. Bement, S. Wrighton et al (2002). Effect of caf-
spectral titration study. Drug Metab. Dispos. 31, feine on acetaminophen hepatotoxicity in cultured
356-359. hepatocytes treated with ethanol and isopentanol.
851. Koley, A.P, J.T.M. Buters, R.C. Robinson, Toxicol. Appl. Pharmacol. 185, 91-97.
A. Markowitz, and RK. Friedman (1995). CO 864. Tang, W and R.A. Stearns (2001). Heterotropic
binding kinetics of human cytochrome P450 3A4: cooperativity of cytochrome P450 3A4 and poten-
Specific interaction of substrates with kinetically tial drug-drug interactions. Curr Drug Metab. 2,
distinguishable conformers. J. Biol. Chem. 270, 185-198.
5014-5018. 865. Egnell,A.C., J.B. Houston, andC.S. Boyer (2003). In
852. Koley, A.P, J.T.M. Buters, R.C. Robinson, vivo CYP3A4 heteroactivation is a possible mecha-
A. Markowitz, and F.K. Friedman (1997). nism for the drug interaction between felbamate and
Differential mechanisms of cytochrome P450 inhi- carbamazepine. J. Pharmacol. Exp. Ther. 305,
bition and activation by a-naphthoflavone. J. Biol. 1251-1262.
Chem. 272,3149-3152. 866. Yun, C.-H., R.A. Okerholm, and F.P Guengerich
853. Koley, A.P., R.C. Robinson, and F.K. Friedman (1993). Oxidation of the antihistaminic drug terfe-
(1996). Cytochrome P450 conformation and sub- nadine in human liver microsomes: Role of
strate interactions as probed by CO binding cytochrome P450 3A(4) in N-dealkylation and
kinetics. Biochimie 78, 706-713. C-hydroxylation. Drug Metab. Dispos. 21,
854. Koley, A.P, R.C. Robinson, A. Markowitz, and 403^09.
F.K. Friedman (1997). Drug-drug interactions: 867. Delaforge, M., M. Jaouen, and D. Mansuy (1983).
Effect of quinidine on nifedipine binding to human Dual effects of macrolide antibiotics on rat liver
cytochrome P450 3A4. Biochem. Pharmacol. 53, cytochrome P-450: Induction and formation of
455^60. metabolite-complexes: A structure-activity rela-
855. Anzenbacherova', E., N. Bee, P. Anzenbacher, tionship. Biochem. Pharmacol. 32, 2309-2318.
J. Hudecek, P Soucek, C. Jung et ah (2000). 868. Bailey, D.G., B. Edgar, J.D. Spence, C. Munzo, and
Flexibility and stability of the structure of J.M.O. Arnold (1990). Felodipine and nifedipine
cytochromes P450 3A4 and BM-3. Eur J. interactions with grapefruit juice. Clin. Pharmacol.
Biochem. 261, 2916-2920. Ther 47, 180.
856. Ma, B., M. Shatsky, H.J. Wolfson, and R. Nussinov 869. Greenblatt, D.J., K.C. Patki, L.L. von Moltke, and
(2002). Multiple diverse ligands binding at a single R.I. Shader (2001). Drug interactions with grapefruit
502 F. Peter Guengerich

juice: An update. J. Clin. Psychopharmacol. 21, cyclosporine pharmacokinetics in marrow transplant


357-359. patients. Blood 64, 1277-1279.
870. Guengerich, F.P. and D.-H. Kim (1990). In vitro 882. von Moltke, L.L. D.J. Greenblatt, S.X. Duan,
inhibition of dihydropyridine oxidation and afla- J.S. Harmatz, and R.I. Shader (1994). In vitro pre-
toxin Bj activation in human liver microsomes by diction of the terfenadine-ketoconazole pharmacoki-
naringenin and other flavonoids. Carcinogenesis netic interaction.^ Clin. Pharmacol. 34,1222-1227.
11, 2275-2279. 883. Dome, J.L., K. Walton, and A.G. Renwick (2003).
871. von Moltke, L.L., D.J. Greenblatt, J.M. Grassi, Human variability in CYP3A4 metabolism and
B.W. Granda, S.X. Duan, S.M. Fogelman et al CYP3A4-related uncertainty factors for risk
(1998). Protease inhibitors as inhibitors of human assessment. Food Chem. Toxicol. 41, 201-224.
cytochromes P450: High risk associated with 884. Yang, L.Q., S.J. Li, YE Cao, X.B. Man, W.E Yu,
ritonavir. J. Clin. Pharmacol. 38, 106-111. H.Y Wang et al. (2003). Different alterations of
872. Unger, M., U. Holzgrabe, W. Jacobsen, C. Cummins, cytochrome P450 3A4 isoform and its gene
and L.Z. Benet (2002). Inhibition of cytochrome expression in livers of patients with chronic liver
P450 3A4 by extracts and kavalactones of Piper diseases. World J. Gastroenterol. 9, 359-363.
methysticum (kava-kava). Planta Med. 68, 885. Lang, C.C, R.M. Brown, M.T. Kinirons,
1055-1058. M.-A. Deathridge, F.P. Guengerich, D. Kelleher
873. Lin, H.L., U.M. Kent, and PR Hollenberg (2002). et al. (1996). Decreased intestinal P450 3A4 in
Mechanism-based inactivation of cytochrome celiac sprue: Reversal following successful gluten
P450 3A4 by 17a-ethynylestradiol: Evidence for free diet. Clin. Pharmacol. Ther 59, 4 1 ^ 6 .
heme destruction and covalent binding to protein. 886. Zhou, S., Y Gao, W Jiang, M. Huang, A. Xu, and
J. Pharmacol. Exp. Then 301, 160-167. J.W Paxton (2003). Interactions of herbs with
874. Palovaara, S., K.T. Kivisto, P. Tapanainen, cytochrome P450. Drug Metab. Rev 35, 35-98.
P. Manninen, P.J. Neuvonen, and K. Laine (2000). 887. Moore, L.B., B. Goodwin, S.A. Jones, G.B. Wisely
Effect of an oral contraceptive preparation contain- C.J. Serabjit-Singh, T.M. Willson et al. (2000). St.
ing ethinylestradiol and gestodene on CYP3A4 John's wort induces hepatic drug metabolism
activity as measured by midazolam 1'-hydroxyla- through activation of the pregnane X receptor. Proc.
tion. Br J. Clin. Pharmacol. 50, 333-337. Natl. Acad Sci. USA 97, 7500-7502.
875. Kuhl, H., C. Jung-Hoffmann, and E Heidt (1988). 888. Watkins, R.E., J.M. Maglich, L.B. Moore,
Alterations in the serum levels of gestodene and G.B. Wisely, S.M. Noble, PR. Davis-Searles et al.
SHBG during 12 cycles of treatment with 30 |jLg (2003). 2.1 A crystal structure of human PXR in
ethinylestradiol and 75 |jLg gestodene. complex with the St. John's wort compound hyper-
Contraception 38, 477-486. forin. Biochemistry 42, 1430-1438.
876. Balogh, A., S. Gessinger, U. Svarovsky, M. Hippius, 889. Henderson, L., Q.Y Yue, C. Bergquist, B. Gerden,
U. Mellinger, G. Klinger et al. (1998). Can oral con- and P. Arlett (2002). St. John's wort (Hypericum
traceptive steroids influence the elimination of perforatum): Drug interactions and clinical out-
nifedipine and its primary pryidine metabolite in comes. Br J. Clin. Pharmacol. 54, 349-356.
humans?. Eur J. Clin. Pharmacol. 54, 729-734. 890. Kolars, J.C, P Benedict, P Schmiedlin-Ren, and
877. Guengerich, EP (1986). Covalent binding to PB. Watkins (1994). Aflatoxin B,-adduct forma-
apoprotein is a major fate of heme in a variety of tion in rat and human small bowel enterocytes.
reactions in which cytochrome P-450 is destroyed. Gastroenterology 106, 433-439.
Biochem. Biophys. Res. Commun. 138, 193-198. 891. Felix, C.A., A.H. Walker, B.J. Lange, T.M. Williams,
878. He, K., L.M. Bomheim, A.M. Ealick, D. Maltby N.J. Winick, N.-K.V Cheung et al. (1998).
H. Yin, and M.A. Correia (1998). Identification of Association of CYP3A4 genotype with treatment-
the heme-modified peptides from cumene related leukemia. Proc. Natl. Acad. Sci. USA 95,
hydroperoxide-inactivated cytochrome P450 3A4. 13176-13181.
Biochemistry 2>1, 17448-17457. 892. Miyoshi, Y, A. Ando, Y Takamura, T. Taguchi,
879. Wienkers, L.C. (2001). Problems associated with in Y Tamaki, and S. Noguchi (2002). Prediction of
vitro assessment of drug inhibition of CYP3A4 and response to docetaxel by CYP3A4 mRNA expression
other P-450 enzymes and its impact on drug dis- in breast cancer tissues. Int. J. Cancer 97, 129-132.
covery. J. Pharmacol. Toxicol. Methods 45, 79-84. 893. Blanco, J.G., M.J. Edick, M.L. Hancock,
880. Plant, N.J. and G.G. Gibson (2003). Evaluation of N.J. Winick, T Dervieux, M.D. Amylon etal. (2002).
the toxicological relevance of CYP3A4 induction. Genetic polymorphisms in CYP3A5, CYP3A4 and
Curr Opin. Drug Discov. Devel. 6, 50-56. NQOl in children who developed therapy-related
881. Yee, G.C., M.S. Kennedy R. Storb, and E.D. Thomas myeloid malignancies. Pharmacogenetics 12,
(1984). Effect of hepatic dysfunction on oral 605-611.
Human P450s 503

894. Rebbeck, T.R., J.M. Jaffe, A.H. Walker, A.J. Wein, 905. Janardan, S.K., K.S. Lown, P Schmiedlin-Ren,
and S.B. Malkowicz (1998). Modification of clini- K.E. Thummel, and RB. Watkins (1996). Selective
cal presentation of prostate tumors by a novel expression of CYP3A5 and not CYP3A4 in human
genetic variant in CYP3A4. J. Natl. Cancer Inst. blood. Pharmacogenetics 6, 379-385.
90, 1225-1228. 906. Hakkola, J., H. Raunio, R. Purkunen, S. Saarikoski,
895. Tayeb, M.T., C. Clark, L. Sharp, N.E. Haites, K. Vahakangas, O. Pelkonen et al. (2001).
RH. Rooney, G.I. Murray et al. (2002). CYP3A4 Cytochrome P450 3 A expression in the human fetal
promoter variant is associated with prostate cancer liver: Evidence that CYP3A5 is expressed in only a
risk in men with benign prostate hyperplasia. limited number of fetal livers. Biol. Neonate 80,
Oncol. Rep. 9, 653-655. 193-201.
896. Tayeb, M.T., C. Clark, N.E. Haites, L. Sharp, 907. Jouna'idi, Y, PS. Guzelian, and M.J. Vilarem
G.I. Murray, and H.L. McLeod (2003). CYP3A4 (1994). Sequence of the 5'-flanking region of
and VDR gene polymorphisms and the risk of CYP3A5: Comparative analysis with CYP3A4 and
prostate cancer in men with benign prostate hyper- CYP3A7. Biochem. Biophys. Res. Commun. 205,
plasia. Br. J. Cancer 88, 928-932. 1741-1747.
897. Ando, Y., T. Tateishi, Y Sekido, T. Yamamoto, 908. Schuetz, J.D., E.G. Schuetz, J.V Thottassery,
T. Satoh, Y Hasegawa et al (1999). Modification PS. Guzelian, S. Strom, and D. Sun (1996).
of clinical presentation of prostate tumors by a Identification of a novel dexamethasone respon-
novel genetic variant in CYP3A4. J. Natl. Cancer sive enhancer in the human CYP3A5 gene and its
Inst. 91, 1587-1588. activation in human and rat liver cells. Mol.
898. Kittles, R.A., W. Chen, R.K. Panguluri, Pharmacol. 49, 63-72.
C. Ahaghotu, A. Jackson, C.A. Adebamowo et al. 909. Lin, Y.S., A.L. Dowling, S.D. Quigley, KM. Farin,
(2002). CYP3A4-V and prostate cancer in African J. Zhang, J. Lamba et al. (2002). Co-regulation of
Americans: Causal or confounding association CYP3A4 and CYP3A5 and contribution to hepatic
because of population stratification?. Hum. Genet. and intestinal midazolam metabolism. Mol.
110, 553-560. Pharmacol. 62, 162-172.
899. Wojnowski, L., E. Hustert, K. Klein, M. Goldammer, 910. Hustert, E., M. Haberl, O. Burk, R. Wolbold,
M. Haberl, J. Kirchheiner et al. (2002). Re: modifi- YQ. He, K. Klein et al. (2001). The genetic deter-
cation of clinical presentation of prostate tumors by a minants of the CYP3A5 polymorphism.
novel genetic variant in CYP3A4. J. Natl. Cancer Pharmacogenetics 11, 773-779.
Inst. 94, 630-631. 911. Paulussen, A., K. Lavrijsen, H. Bohets,
900. Wrighton, S.A., B.J. Ring, RB. Watkins, and J. Hendrickx, P Verhasselt, W Luyten et al. (2000).
M. VandenBranden (1989). Identification of a Two linked mutations in transcriptional regulatory
polymorphically expressed member of the human elements of the CYP3A5 gene constitute the major
cytochrome P-450III family. Mol. Pharmacol. 86, genetic determinant of polymorphic activity in
97-105. humans. Pharmacogenetics 10, 415^24.
901. Aoyama, T., S. Yamano, D.J. Waxman, 912. Shih, PS. and J.D. Huang (2002). Pharmacokinetics
D.R Lapenson, U.A. Meyer, V Fischer et al. of midazolam and 1 '-hydroxymidazolam in Chinese
(1989). Cytochrome P-450 hPCN3, a novel with different CYP3A5 genotypes. Drug Metab.
cytochrome P-450 III A gene product that is differ- Dispos. 30, 1491-1496.
entially expressed in adult human liver. J. Biol. 913. Wrighton, S.A., W.R. Brian, M.A. Sari, M. Iwasaki,
Chem. 264, 10388-10395. EP Guengerich, J.L. Raucy et al. (1990). Studies on
902. Murray, G.L, S. Pritchard, W.T. Melvin, and the expression and metabolic capabilities of human
M.D. Burke (1995). Cytochrome P450 CYP3A5 in liver cytochrome P450IIIA5 (HLp3). Mol.
the human anterior pituitary gland. FEBS Lett. Pharmacol. 38, 207-213.
364, 79-82. 914. Gibbs, M.A., K.E. Thummel, D.D. Shen, and
903. Yamakoshi, Y, T. Kishimoto, K. Sugimura, and K.L. Kunze (1999). Inhibition of cytochrome P-
H. Kawashima (1999). Human prostate CYP3A5: 450 3A(CYP3A) in human intestinal and liver
Identification of a unique 5'-untranslated sequence microsomes: Comparison of K- values and impact
and characterization of purified recombinant protein. of CYP3A5 expression. Drug Metab. Dispos. 27,
Biochem. Biophys. Res. Commun. 260, 676-681. 180-187.
904. Hukkanen, J., T. Vaisanen, A. Lassila, R. Piipari, 915. Kamataki, T., M. Sugiura, Y Yamazoe, and R. Kato
S. Anttila, O. Pelkonen et al. (2003). Regulation of (1979). Purification and properties of cytochrome
CYP3A5 by glucocorticoids and cigarette smoke P-450 and NADPH-cytochrome c (P-450) reduc-
in human lung-derived cells. J. Pharmacol. Exp. tase from human liver microsomes. Biochem.
Then 304, 745-752. Pharmacol. 28, 1993-2000.
504 F. Peter Guengerich

916. Kitada, M., T. Kamataki, K. Itahashi, T. Rikihisa, element in the 5' flanking region. Biochim.
R. Kato, and Y. Kanakubo (1985). Purification and Biophys. Acta 1130, 133-138.
properties of cytochrome P-450 from homogenates 927. Greuet, J., L. Pichard, C. Bonfils, J. Domergue,
of human fetal livers. Arch. Biochem. Biophys. and P. Maurel (1996). The fetal specific gene
241, 275-280. CYP3A7 is inducible by rifampicin in adult human
917. Kitada, M., T. Kamataki, K. Itahashi, T. Rikihisa, hepatocytes in primary culture. Biochem. Biophys.
R. Kato, andY. Kanakubo (1985). Immunochemical Res. Commun. 225, 689-694.
examinations of C3^ochrome P-450 in various tissues 928. Pascussi, J.M., Y Jounaidi, L. Drocourt,
of human fetuses using antibodies to human fetal J. Domergue, C. Balabaud, P Maurel et al. (1999).
cytochrome P-450, P-450 HFLa. Biochem. Biophys. Evidence for the presence of a functional pregnane
Res. Commun. 131, 1154-1159. X receptor response element in the CYP3A7 pro-
918. Okajima, Y, N. Inaba, I. Fukazawa, Y Ota, moter gene. Biochem. Biophys. Res. Commun. 260,
Y Hirai, N. Sato etal. (1993). Immunohistochemical 377-381.
and immunoelectron microscopic study of 929. Bertilsson, G., A. Berkenstam, and P. Blomquist
cytochrome P-450 of human fetal livers (P- (2001). Functionally conserved xenobiotic respon-
450HFLa): Implications for an onco-feto-placental sive enhancer in cytochrome P450 3A7. Biochem.
enzyme. Asia Oceania J. Obstet. Gynaecol. 19, Biophys. Res. Commun. 280, 139-144.
329-341. 930. Saito, T., Y Takahashi, H. Hashimoto, and
919. Schuetz, J.D., S. Kauma, and PS. Guzelian (1993). T. Kamataki (2001). Novel transcriptional regula-
Identification of the fetal liver cytochrome tion of the human CYP3A7 gene by Spl and Sp3
CYP3A7 in human endometrium and placenta. through nuclear factor KB-like element. J. Biol.
J. Clin. Invest. 92, 1018-1024. C/zew. 276, 38010-38022.
920. Sarkar, M.A., V Vadlamuri, S. Ghosh, and 931. Finta, C. and PG. Zaphiropoulos (2000). The
D.D. Glover (2003). Expression and cychc vari- human cytochrome P450 3A locus. Gene evolution
ability of CYP3A4 and CYP3A7 isoforms in by capture of downstream exons. Gene 260, 13-23.
human endometrium and cervix during the men- 932. Burk, O., H. Tegude, I. Koch, E. Hustert,
strual cycle. Drug Metab. Dispos. 31, 1-6. R. Wolbold, H. Glaeser, K. et al. (2002). Molecular
921. Hakkola, J., H. Raunio, R. Purkunen, O. Pelkonen, mechanisms of polymorphic CYP3A7 expression
S. Saarikoski, T. Cresteil et al. (1996). Detection of in adult human liver and intestine. J. Biol. Chem.
cytochrome P450 gene expression in human pla- 277, 24280-24288.
centa in first trimester of pregnancy. Biochem. 933. Kitada, M., T. Kamataki, K. Itahashi, T. Rikihisa,
Pharmacol. 52, 379-383. and Y Kanakubo (1987). Significance of
922. Yang, H.Y.L., Q.P Lee, A.E. Rettie, and cytochrome P-450 (P-450 HFLa) of human fetal
M.R. Juchau (1994). Functional cytochrome livers in the steroid and drug oxidations. Biochem.
P4503A isoforms in human embryonic tissues: Pharmacol. 36, 453-^56.
Expression during organogenesis. Mol. 934. Kitada, M., T. Kamataki, K. Itahashi, T. Rikihisa,
Pharmacol. 46, 922-928. and Y Kanakubo (1987). P-450 HFLa, a form
923. Schuetz, J.D., D.L. Beach, and PS. Guzelian of cytochrome P-450 purified from human
(1994). Selective expression of cytochrome P450 fetal livers, is the 16a-hydroxylase of dehy-
CYP3A mRNAs in embryonic and adult human droepiandrosterone 3-sulfate. J. Biol. Chem. 262,
liver. Pharmacogenetics 4, 11-20. 13534-13537.
924. Tateishi, T., H. Nakura, M. Asoh, M. Watanabe, 935. Ohmori, S., N. Fujiki, H. Nakasa, H. Nakamura,
M. Tanaka, T. Kumai et al. (1997). A comparison I. Ishii, K. Itahashi et al. (1998). Steroid hydroxy-
of hepatic cytochrome P450 protein expression lation by human fetal CYP3A7 and human
between infancy and postinfancy. Life Sci. 61, NADPH-cytochrome P450 reductase coexpressed
2567-2574. in insect cells using baculovirus. Res. Commun.
925. Komori, M., K. Nishio, H. Ohi, M. Kitada, and Mol. Pathol. Pharmacol. 100, 15-28.
T. Kamataki (1989). Molecular cloning and 936. Gorski, J.C., S.D. Hall, D.R. Jones,
sequence analysis of cDNA containing entire cod- M. VandenBranden, and S.A. Wrighton (1994).
ing region for human fetal liver cytochrome P-450. Regioselective biotransformation of midazolam by
J. Biochem. (Tokyo) 106, 161-163. members of the human cytochrome P450 3A
926. Itoh, S., T. Yanagimoto, S. Tagawa, H. Hashimoto, (CYP3A) subfamily. Biochem. Pharmacol. 47,
R. Kitamura et al. (1992). Genomic organization 1643-1653.
of human fetal specific P-450IIIA7 (cytochrome 937. Gillam, E.M.J., R.M. Wunsch, Y.-R Ueng,
P-450HFLa)-related gene(s) and interaction of T. Shimada, PE.B. Reilly, T. Kamataki et al.
transcriptional regulatory factor with its DNA (1997). Expression of cytochrome P450 3A7 in
Human P450s 505

Escherichia coli: Effects of 5' modification and Characterization of a cDNA encoding a human
catalytic characterization of recombinant enzymes kidney, cytochrome P-450 4A fatty acid w-hydroxy-
expressed in bicistronic format with NADPH- lase and the cognate enzyme expressed in
cytochrome P450 reductase. Arch. Biochem. Escherichia coli. Biochim. Biophys. Acta 1172,
Biophys. 346, 81-90. 161-166.
938. Hashimoto, H., Y. Yanagawa, M. Sawada, S. Itoh, 949. Bell, D.R., N.J. Plant, C.G. Rider, L. Na, S. Brown,
T. Deguchi, andT. Kamataki (1995). Simultaneous I. Ateitalla et al. (1993). Species-specific induction
expression of human CYP3A7 and A^-acetyltrans- of cytochrome P-450 4A RNAs: PCR cloning
ferase in Chinese hamster CHL cells results in high of partial guinea-pig, human and mouse CYP4A
cytotoxicity for carcinogenic heterocyclic amines. cDNAs. Biochem. J. 294, 173-180.
Arch. Biochem. Biophys. 320, 323-329. 950. Imaoka, S., H. Ogawa, S. Kimura, and F.J. Gonzalez
939. Li, Y, T. Yokoi, M. Katsuki, J.S. Wang, (1993). Complete cDNA sequence and cDNA-
J.D. Groopman, and T. Kamataki (1997). In vivo directed expression of CYP4A11, a fatty acid co-
activation of aflatoxin Bj in C57BL/6N mice car- hydroxylase expressed in human kidney. DNA Cell
rying a human fetus-specific CYP3A7 gene. Biol. 12, 893-899.
Cancer Res. 57, 641-645. 951. Powell, PK., I. Wolf, and J.M. Lasker (1996).
940. Yamada, A., K. Fujita, T. Yokoi, S. Muto, A. Suzuki, Identification of CYP4A11 as the major lauric
Y. Gondo et al. (1998). In vivo detection of muta- acid w-hydroxylase in human liver microsomes.
tions induced by aflatoxin Bj using human Arch. Biochem. Biophys. 335, 219-226.
CYP3A7/HITEC hybrid mice. Biochem. Biophys. 952. Bellamine, A., Y Wang, M.R. Waterman, J.V Gainer,
Res. Commun. 250, 150-153. III, E.R Dawson, N.J. Brown et al. (2003).
941. Li, Y, T. Yokoi, R. Kitamura, M. Sasaki, M. Gunji, Characterization of the CYP4A11 gene, a second
M. Katsuki et al. (1996). Establishment of CYP4A gene in humans. Arch. Biochem. Biophys.
transgenic mice carrying human fetus-specific 409,221-227.
CYP3A7. Arch. Biochem. Biophys. 329, 235-240. 953. Lu, A.Y.H. and M.J. Coon (1968). Role of hemo-
942. Chen, H., A.G. Fantel, and M.R. Juchau (2000). protein P-450 in fatty acid co-hydroxylation in
Catalysis of the 4-hydroxylation of retinoic acids a soluble enzyme system from liver microsomes.
by CYP3A7 in human fetal hepatic tissues. Drug J.Biol. Chem. 243, 1331-1332.
Metab. Dispos. 28, 1051-1057. 954. Gonzalez, M.C., C. Marteau, J. Franchi, and
943. Inoue, E., Y Takahashi, Y Imai, and T. Kamataki D. Migliore-Samour (2001). Cytochrome P450
(2000). Development of bacterial expression sys- 4A11 expression in human keratinocytes: Effects
tem with high yield of CYP3A7, a human fetus- of ultraviolet irradiation. Br J. Dermatol. 145,
specific form of cytochrome P450. Biochem. 749-757.
Biophys. Res. Commun. 269, 623-627. 955. Cummings, B.S., J.M. Lasker, and L.H. Lash
944. Kondoh, N., T. Wakatsuki, A. Ryo, A. Hada, (2000). Expression of glutathione-dependent
T. Aihara, S. Horiuchi et al. (1999). Identification enzymes and cytochrome P450s in freshly isolated
and characterization of genes associated with and primary cultures of proximal tubular cells
human hepatocellular carcinogenesis. Cancer Res. from human kidney. J. Pharmacol. Exp. Then 293,
59, 4990-4996. 677-685.
945. Gellner, K., R. Eiselt, E. Hustert, H. Arnold, 956. Savas, U., M.H. Hsu, and E.E Johnson (2003).
I. Koch, M. Haberl et al. (2001). Genomic organi- Differential regulation of human CYP4A genes by
zation of the human CYP3 A locus: Identification of peroxisome proliferators and dexamethasone.
a new, inducible CYP3A gene. Pharmacogenetics Arch. Biochem. Biophys. 409, 212-220.
II, 111-121. 957. Kawashima, H., T. Naganuma, E. Kusunose,
946. Domanski, T.L., C. Finta, J.R. Halpert, and T. Kono, R. Yasumoto, K. Sugimura et al. (2000).
P.G. Zaphiropoulos (2001). cDNA cloning and ini- Human fatty acid co-hydroxylase. CYP4AJI:
tial characterization of CYP3A43, a novel human Determination of complete genomic sequence and
cytochrome P450. Mol. Pharmacol. 59, 386-392. characterization of purified recombinant protein.
947. Westlind, A., S. Malmebo, I. Johansson, C. Otter, Arch. Biochem. Biophys. 378, 333-339.
T.B. Andersson, M. Ingelman-Sundberg et al. 958. Cattley, R.C., J. DeLuca, C. Elcombe, P Fenner-
(2001). Cloning and tissue distribution of a novel Crisp, B.G. Lake, D.S. Marsman et al. (1998). Do
human cytochrome P450 of the CYP3A subfamily, peroxisome proliferating compounds pose a hepa-
CYP3A43. Biochem. Biophys. Res. Commun. 281, tocarcinogenic hazard to humans?. Regul. Toxicol.
1349-1355. Pharmacol. 27, 47-60.
948. Palmer, C.N., T.H. Richardson, K.L Griffin, 959. Dierks, E.A., Z. Zhang, E.R Johnson, and PR. Ortiz
M.H. Hsu, A.S. Muerhofif, J.E. Clark et al. (1993). de Montellano (1998). The catalytic site of
506 F. Peter Guengerich

cytochrome P4504A11 (CYP4A11) and its L131F P450 CYP4B1: Evidence for a non-functional
mutant. J. Biol. Chem. 273, 23055-23061. allelic variant. Pharmacogenetics 12, 367-374.
960. Hoch, U, Z. Zhang, D.L. Kroetz, and P.R. Ortiz de 971. Zheng, Y.-M., M.B. Fisher, N. Yokotani, Y Fujii-
Montellano (2000). Structural determination of the Kuriyama, and A.E. Rettie (1998). Identification
substrate specificities and regioselectivities of the of a meander region proline residue critical for
rat and human fatty acid w-hydroxylases. Arch. heme binding to cytochrome P450: Implications
Biochem. Biophys. 373, 63-71. for the catalytic function of human CYP4B1.
961. Powell, PK., I. Wolf, R. Jin, and J.M. Lasker Biochemistry'il, 12847-12851.
(1998). MetaboHsm of arachidonic acid to 20- 972. Imaoka, S., K. Hayashi, T. Hiroi, Y Yabusaki,
hydroxy-5,8,ll,14-eicosatetraenoic acid by P450 T. Kamataki, and Y Funae (2001). A transgenic
enzymes in human liver: Involvement of CYP4F2 mouse expressing human CYP4B1 in the liver.
and CYP4A11. J. Pharmacol. Exp. Then 285, Biochem. Biophys. Res. Commun. 284, 757-762.
1327-1336. 973. Henne, K.R., M.B. Fisher, K.R. Iyer, D.H. Lang,
962. Lasker, J.M., W.B. Chen, I. Wolf, B.R Bloswick, WF. Trager, and A.E. Rettie (2001). Active site
PD. Wilson, and PK. Powell (2000). Formation of characteristics of CYP4B1 probed with aromatic
20-hydroxyeicosatetraenoic acid, a vasoactive and ligands. Biochemistry 40, 8597-8605.
natriuretic eicosanoid, in human kidney. J. Biol. 974. Henne, K.R., K.L. Kunze, YM. Zheng, P Christmas,
Chem. 275,4118^126. R.J. Soberman, and A.E. Rettie (2001). Covalent
963. Oliw, E.H., K. Stark, and J. Bylund (2001). linkage of prosthetic heme to CYP4 family P450
Oxidation of prostaglandin W^ and prostaglandin enzymes. Biochemistry 40, 12925-12931.
H2 analogues by human cytochromes P450: 975. Hoch, U and PR. Ortiz de Montellano (2001).
Analysis of w-side chain hydroxy metaboHtes and Covalently linked heme in cytochrome P4504A
four steroisomers of 5-hydroxyprostaglandin Ij by fatty acid hydroxylases. J. Biol. Chem. 276,
mass spectrometry. Biochem. Pharmacol. 62, 11339-11346.
407-^15. 976. Frank, S., S. StefFens, U Fischer, A. Tlolko,
964. Chang, Y.T. and G.H. Loew (1999). Homology N.G. Rainov, and CM. Kramm (2002).
modeling and substrate binding study of human Differential cytotoxicity and bystander effect of
CYP4A11 enzyme. Proteins 34, 403^15. the rabbit cytochrome P450 4B1 enzyme gene by
965. LeBrun, L.A., U. Hoch, and P R. Ortiz de two different prodrugs: Implications for pharma-
Montellano (2002). Autocatalytic mechanism and cogene therapy. Cancer Gene Ther 9, 178-188.
consequences of covalent heme attachment in the 977. Kikuta, Y, E. Kusunose, T. Kondo, S. Yamamoto,
cytochrome P4504A family. J. Biol. Chem. Ill, H. Kinoshita, and M. Kusunose (1994). Cloning
12755-12761. and expression of a novel form of leukotriene B4
966. Nhamburo, PT., F.J. Gonzalez, O.W. McBride, omega-hydroxylase from human liver. FEBS Lett.
H.V Gelboin, and S. Kimura (1989). Identification 348,70-74.
of a new P450 expressed in human lung: Com- 978. Kikuta, Y, E. Kusunose, and M. Kusunose (2002).
plete cDNA sequence, cDNA-directed expression, Prostaglandin and leukotriene w-hydroxylases.
and chromosome mapping. Biochemistry 28, Prostaglandins Other Lipid Medial 68-69, 345-362.
8060-8066. 979. Jin, R., D.R. Koop, J.L. Raucy, and J.M. Lasker
967. Imaoka, S., Y Yoneda, T. Sugimoto, T. Hiroi, (1998). Role of human CYP4F2 in hepatic catabo-
K. Yamamoto, T. Nakatani et al. (2000). CYP4B1 is lism of the proinflammatory agent leukotriene B4.
a possible risk factor for bladder cancer in humans. Arch. Biochem. Biophys. 359, 89-98.
Biochem. Biophys. Res. Commun. Ill, 776-780. 980. Kikuta, Y, Y. Miyauchi, E. Kusunose, and
968. Iscan, M., T. Klaavuniemi, T. Coban, M. Kusunose (1999). Expression and molecular
N. Kapucuoglu, O. Pelkonen, and H. Raunio cloning of human liver leukotriene B4 w-hydroxy-
(2001). The expression of cytochrome P450 lase (CYP4F2) gene. DMA Cell Biol. 18, 723-730.
enzymes in human breast tumours and normal 981. Kikuta, Y, E. Kusunose, and M. Kusunose (2000).
breast tissue. Breast Cancer Res. Treat. 70, 47-54. Characterization of human liver leukotriene B^ w-
969. Finnstrom, N., C. Bjelfman, T.G. Soderstrom, hydroxylase P450 (CYP4F2). J. Biochem. (Tokyo)
G. Smith, L. Egevad, B.J. Norlen et al. (2001). 111, 1047-1052.
Detection of cytochrome P450 mRNA transcripts 982. Kikuta, Y, E. Kusunose, K. Endo, S. Yamamoto,
in prostate samples by RT-PCR. Eur J. Clin. Invest. K. Sogawa, Y Fujii-Kuriyama et al. (1993). A
31, 880-886. novel form of cytochrome P-450 family 4 in
970. Lo-Guidice, J.M., D. Allorge, C. Cauffiez, human polymorphonuclear leukocytes: cDNA
D. Chevalier, J.J. Lafitte, M. Lhermitte et al. (2002). cloning and expression of leukotriene B^ co-
Genetic polymorphism of the human cytochrome hydroxylase. J. Biol. Chem. 268, 9376-9380.
Human P450s 507

983. Kikuta, Y., M. Kato, Y. Yamashita, Y Miyauchi, platelet thromboxane A synthase. Biochem.
K. Tanaka, N. Kamada et al (1998). Human Biophys. Res. Commun. 178, 1479-1484.
leukotriene B^ w-hydroxylase (CYP4F3) gene: 996. Nusing, R. and V Ullrich (1992). Regulation of
Molecular cloning and chromosomal localization. cyclooxygenase and thromboxane synthase in
DNA Cell Biol. 17, 221-230. human monocytes. Eur J. Biochem. 206, 131-136.
984. Christmas, P., S.R. Ursino, J.W. Fox, and 997. Young, V, M. Ho, H. Vosper, J.J. Belch, and
R.J. Soberman (1999). Expression of the CYP4F3 C.N. Palmer (2002). Elevated expression of the
gene. J. Biol. Chem. 274, 21191-21199. genes encoding TNF-a and thromboxane syn-
985. Christmas, P., J.P Jones, C.J. Patten, D.A. Rock, thase in leucocytes from patients with systemic
Y Zheng, S.M. Cheng et al. (2001). Alternative sclerosis. Rheumatology 41, 869-875.
splicing determines the function of CYP4F3 by 998. Nusing, R., RM. Fehr, F. Gudat, E. Kemeny,
switching substrate specificity. J. Biol. Chem. 276, M.J. Mihatsch, and V Ullrich (1994). The local-
38166-38172. ization of thromboxane synthase in normal and
986. Bylund, J., N. Finnstrom, and E.H. Oliw (1999). pathological human kidney tissue using a mono-
Gene expression of a novel cytochrome P450 of clonal antibody Tu 300. Virchows Arch. 424,
the CYP4F subfamily in human seminal vesicles. 69-74.
Biochem. Biophys. Res. Commun. 261, 169-174. 999. Ihara, H., C. Yokoyama, A. Miyata, T. Kosaka,
987. Bylund, J., M. Hidestrand, M. Ingelman- R. Nusing, V Ullrich et al. (1992). Induction of
Sundberg, and E.H. Oliw (2000). Identification of thromboxane synthase and prostaglandin endoper-
CYP4F8 in human seminal vesicles as a prominent oxide synthase mRNAs in human erythroleukemia
19-hydroxylase of prostaglandin endoperoxides. cells by phorbol ester. FEBS Lett. 306, 161-164.
J. Biol. Chem. 275, 21844-21849. 1000. Yaekashiwa, M. and L.H. Wang (2002).
988. Stark, K., H. Torma, M. Cristea, and E.H. Oliw Transcriptional control of the human thrombox-
(2003). Expression of CYP4F8 (prostaglandin H ane synthase gene in vivo and in vitro. J. Biol.
19-hydroxylase) in human epithelia and prominent Chem. 277, 22497-22508.
induction in epidermis of psoriatic lesions. Arch. 1001. Chevaher, D., J.M. Lo-Guidice, E. Sergent,
Biochem. Biophys. 409, 188-196. D. Allorge, H. Debuysere, N. Ferrari et al. (2001).
989. Cui, X., D.R. Nelson, and H.W. Strobel (2000). Identification of genetic variants in the human
A novel human cytochrome P450 4F isoform thromboxane synthase gene (CYP5A1). Mutat.
(CYP4F11): cDNA cloning, expression, and Res. 432, 61-67.
genomic structural characterization. Genomics 68, 1002. Ullrich, V and M. Haurand (1983). Thromboxane
161-166. synthase as a cytochrome P450 enzyme. Adv.
990. Bylund, J., M. Bylund, and E.H. Oliw (2001). Prostaglandin Thromboxane Leukot. Res. 11,
cDNA cloning and expression of CYP4F12, 105-110.
a novel human cytochrome P450. Biochem. 1003. Haurand, M. and V Ullrich (1985). Isolation and
Biophys. Res. Commun. 280, 892-897. characterization of thromboxane synthase from
991. Hashizume, T, S. Imaoka, T. Hiroi, Y Terauchi, human platelets as a cytochrome P-450 enzyme.
T. Fujii, H. Miyazaki et al. (2001). cDNA cloning J. Biol. Chem. 260, 15059-15067.
and expression of a novel cytochrome P450 1004. Yokoyama, C , A. Miyata, K. Suzuki, Y Nishikawa,
(CYP4F12) from human small intestine. Biochem. T. Yoshimoto, S. Yamamoto et al. (1993).
Biophys. Res. Commun. 280, 1135-1141. Expression of human thromboxane synthase using
992. Hashizume, T., S. Imaoka, M. Mise, Y Terauchi, a baculovirus system. FEBS Lett. 318, 91-94.
T. Fujii, H. Miyazaki et al. (2002). Involvement 1005. Hecker, M., M. Haurand, V Ullrich, U. Diczfalusy,
of CYP2J2 and CYP4F12 in the metabolism and S. Hammarstrom (1987). Products, kinetics,
of ebastine in human intestinal microsomes. and substrate specificity of homogeneous
J. Pharmacol. Exp. Ther 300, 298-304. thromboxane synthase from human platelets:
993. Bylund, J., C. Zhang, and D.R. Harder (2002). Development of a novel enzyme assay. Arch.
Identification of a novel cytochrome P450, Biochem. Biophys. 254, 124-135.
CYP4X1, with unique localization specific to the 1006. Hecker, M., W.J. Baader, P Weber, and V Ullrich
brain. Biochem. Biophys. Res. Commun. 296, (1987). Thromboxane synthase catalyses hydrox-
677-684. ylation of prostaglandin H2 analogs in the
994. Ullrich, V (2003). Thoughts on thiolate tethering. presence of iodosylbenzene. Eur J. Biochem.
Tribute and thanks to a teacher. Arch. Biochem. 169, 563-569.
Biophys. 409,45-51. 1007. Hecker, M. and V Ullrich (1989). On the
995. Yokoyama, C , A. Miyata, H. Ihara, V Ullrich, and mechanism of prostacyclin and thromboxane A2
T. Tanabe (1991). Molecular cloning of human biosynthesis. J. Biol. Chem. 264, 141-150.
508 F. Peter Guengerich

1008. Alusy, UD. and S. Hammarstrom (1977). 1021. Wang, D.P and J.Y. Chiang (1994). Structure and
Inhibitors of thromboxane synthase in human nucleotide sequences of the human cholesterol
platelets. FEES Lett. 82, 107-110. 7a-hydroxylase gene {CYP7). Genomics 20,
1009. Gorman, R.R., G.L. Bundy, D.C. Peterson, 320-323.
F.F. Sun, O.V Miller, and F.A. Fitzpatrick (1977). 1022. Xu, G., G. Salen, S. Shefer, G.C. Ness,
Inhibition of human platelet thromboxane L.B. Nguyen, T.S. Parker et al. (1995).
synthetase by 9,ll-azoprosta-5,13-dienoic acid. Unexpected inhibition of cholesterol 7a-hydrox-
Proc. Natl Acad. Sci. USA 74, 4007-4011. ylase by cholesterol in New Zealand white
1010. Vane, J.R. (1978). Inhibitors of prostaglandin, and Watanabe heritable hyperlipidemic rabbits.
prostacyclin, and thromboxane synthesis. Adv. J. Clin. Invest. 95, 1497-1504.
Prostaglandin Thromboxane Res. 4, 2 7 ^ 4 . 1023. Breuer, O., E. Sudjana-Sugiaman, G. Eggertsen,
1011. Hecker, M., M. Haurand, V Ullrich, and S. Terao J.Y. Chiang, and I. Bjorkhem (1993). Cholesterol
(1986). Spectral studies on structure-activity 7a-hydroxylase is up-regulated by the competi-
relationships of thromboxane synthase inhibitors. tive inhibitor 7-oxocholesterol in rat liver. Eur J.
Eur J. Biochem. 157, 217-223. Biochem. 215, 705-710.
1012. Pace-Asciak, C.R., D. Reynaud, P Demin, 1024. Shibata, A., PF Ohneseit, YC. Tsai, C.H. Spruck,
R. Aslam, and A. Sun (2002). A new family of III, PW Nichols, H.S. Chiang et al. (1994).
thromboxane receptor antagonists with secondary Mutational spectrum in the p53 gene in bladder
thromboxane synthase inhibition. J. Pharmacol. tumors from the endemic area of black foot dis-
Exp. r/?er. 301, 618-624. ease in Taiwan. Carcinogenesis 15, 1085-1087.
1013. Miki, N., R. Miura, and Y. Miyake (1987). 1025. Schwarz, M., E.G. Lund, K.D. Setchell,
Purification and characterization of cholesterol H.J. Kayden, J.E. Zerwekh, I. Bjorkhem et al.
7a-hydroxylase cytochrome P-450 of untreated (1996). Disruption of cholesterol 7a-hydroxylase
rabbit liver microsomes. J. Biochem. (Tokyo) 101, gene in mice. II. Bile acid deficiency is overcome
1087-1094. by induction of oxysterol 7a-hydroxylase. J. Biol.
1014. Ogishima, T, S. Deguchi, and K. Okuda (1987). Chem. 271, 18024-18031.
Purification and characterization of cholesterol 1026. Lehmann, J.M., S.A. Kliewer, L.B. Moore,
7a-hydroxylase from rat liver microsomes. T.A. Smith-Oliver, B.B. Oliver, J.L. Su et al.
J. Biol. Chem. 262, 7646-7650. (1997). Activation of the nuclear receptor LXR
1015. Nguyen, L.B., S. Shefer, G. Salen, G. Ness, by oxysterols defines a new hormone response
R.D. Tanaka, V Packin et al. (1990). Purification pathway J Biol. Chem. 272, 3137-3140.
of cholesterol 7a-hydroxylase from human and 1027. Peet, D.J., S.D. Turley W. Ma, B.A. Janowski,
rat liver and production of inhibiting polyclonal J.M. Lobaccaro, R.E. Hammer et al. (1998).
antibodies. J. Biol. Chem. 265, 4541^546. Cholesterol and bile acid metabolism are
1016. Noshiro, M. and K. Okuda (1990). Molecular impaired in mice lacking the nuclear oxysterol
cloning and sequence analysis of cDNA encoding receptor LXRa. Cell 93, 693-704.
human cholesterol 7 a-hydroxylase. FEBS Lett. 1028. Russell, D.W. (1999). Nuclear orphan receptors
268, 137-140. control cholesterol catabolism. Cell 91, 539-542.
1017. Li, Y.C., D.P Wang, and J.Y.L. Chiang (1990). 1029. Makishima, M., A.Y Okamoto, J.J. Repa, H. Tu,
Regulation of cholesterol 7a-hydroxylase in the R.M. Learned, A. Luk et al. (1999).
liver: Cloning, sequencing, and regulation of cho- Identification of a nuclear receptor for bile acids.
lesterol 7a-hydroxylase mRNA. J. Biol. Chem. Science 2M, 1362-1365.
265, 12012-12019. 1030. Nitta, M., S. Ku, C. Brown, A.Y. Okamoto, and
1018. Jelinek, D.F., S. Andersson, C.A. Slaughter, and B. Shan (1999). CPF: An orphan nuclear receptor
D.W. Russell (1990). Cloning and regulation of that regulates liver-specific expression of the
cholesterol 7a-hydroxylase, the rate-limiting human cholesterol 7a-hydroxylase gene. Proc.
enzyme in bile acid biosynthesis. J. Biol. Chem. Natl. Acad Sci. USA 96, 6669-6665.
265,8190-8197. 1031. Chiang, J.Y, R. Kimmel, C. Weinberger, and
1019. Cohen, J.C, J.J. Cah, D.F Jelinek, M. Mehrabian, D. Stroup (2000). Farnesoid X receptor responds
R.S. Sparkes, A.J. Lusis et al. (1992). Cloning to bile acids and represses cholesterol 7a-hydrox-
of the human cholesterol 7a-hydroxylase gene ylase gene (CYP7AJ) transcription. J. Biol. Chem.
(CYP7) and localization to chromosome 8ql l-ql2. 215, 10918-10924.
Genomics 14, 153-161. 1032. Staudinger, J.L., B. Goodwin, S.A. Jones,
1020. Nishimoto, M., M. Noshiro, and K. Okuda (1993). D. Hawkins-Brown, K.I. MacKenzie, A. LaTour
Structure of the gene encoding human liver cho- et al. (2001). The nuclear receptor PXR is a litho-
lesterol 7a-hydroxylase. Biochim. Biophys. Acta cholic acid sensor that protects against liver toxi-
nil, 147-150. city Proc. Natl. Acad Sci. USA 98, 3369-3374.
Human P450s 509

1033. Gupta, S., W.M. Pandak, and P.B. Hylemon ovariectomized baboons. Biochim. Biophys. Acta
(2002). LXRa is the dominant regulator of 1084, 300-302.
CYP7A1 transcription. Biochem. Biophys. Res. 1044. Crestani, M., A. Sadeghpour, D. Stroup, G. Galli,
Commun. 293, 338-343. and J.Y. Chiang (1996). The opposing effects of
1034. Patel, D.D., B.L. Knight, A.K. Soutar, G.F. Gibbons, retinoic acid and phorbol esters converge to a
and D.P. Wade (2000). The effect of peroxisome- common response element in the promoter of the
proliferator-activated receptor-a on the activity of rat cholesterol 7a-hydroxylase gene (CYP7A).
the cholesterol 7a-hydroxylase gene. Biochem. Biochem. Biophys. Res. Commun. 225, 585-592.
J. 351, 747-753. 1045. Goodart, S.A., C. Huynh, W Chen, A.D. Cooper,
1035. Cheema, S.K. and L.B. Agellon (2000). The and B. Levy-Wilson (1999). Expression of the
murine and human cholesterol 7a-hydroxylase human cholesterol 7a-hydroxylase gene in trans-
gene promoters are differentially responsive to genic mice. Biochem. Biophys. Res. Commun.
regulation by fatty acids mediated via peroxisome 266, 454-459.
proliferator-activated receptor a. J. Biol. Chem. 1046. Chen, J.Y, B. Levy-Wilson, S. Goodart, and
215, 12530-12536. A.D. Cooper (2002). Mice expressing the human
1036. Palmer, C.N.A., M.H. Hsu, K.J. Grififin, J.L. Raucy, CYP7A1 gene in the mouse CYP7A1 knock-out
and E.F. Johnson (1998). Peroxisome proliferator background lack induction of CYP7A1 expres-
activated receptor-a expression in human liver. sion by cholesterol feeding and have increased
Mol. Pharmacol. 53, 14-22. hypercholesterolemia when fed a high fat diet.
1037. Marrapodi, M. and J.Y. Chiang (2000). J. Biol. Chem. 277, 42588^2595.
Peroxisome proliferator-activated receptor a 1047. Reihner, E., I. Bjorkhem, B. Angelin, S. Ewerth,
(PPARa) and agonist inhibit cholesterol and K. Einarsson (1989). Bile acid synthesis in
7a-hydroxylase gene (CYP7A1) transcription. humans: Regulation of hepatic microsomal cho-
J. Lipid Res. 41, 514-520. lesterol 7a-hydroxylase activity. Gastroenterology
1038. De Fabiani, E., N. Mitro, A.C. Anzulovich, 97, 1498-1505.
A. Pinelli, G. GalH, and M. Crestani (2001). The 1048. Mayer, D. (1976). The circadian rhythm of syn-
negative effects of bile acids and tumor necrosis thesis and catabolism of cholesterol. Arch.
factor-a on the transcription of cholesterol 7a- Toxicol. 36, 267-276.
hydroxylase gene {CYP7A1) converge to hepatic 1049. Gielen, J., J. Van Cantfort, B. Robaye, and
nuclear factor-4: A novel mechanism of feedback J. Renson (1975). Rat-liver cholesterol 7a-
regulation of bile acid synthesis mediated hydroxylase. 3. New results about its circadian
by nuclear receptors. J. Biol. Chem. 276, rhythm. Eur J. Biochem. 55, 4 1 ^ 8 .
30708-30716. 1050. Danielsson, H. and K. Wikvall (1981). Evidence
1039. Feingold, K.R., D.K. Spady, A.S. Pollock, for a specific cytochrome P-450 with short half-
A.H. Moser, and C. Grunfeld (1996). Endotoxin, life catalyzing 7a-hydroxylation of cholesterol.
TNF, and IL-1 decrease cholesterol 7a-hydroxy- Biochem. Biophys. Res. Commun. 103, 46-51.
lase mRNA levels and activity. J. Lipid Res. 37, 1051. Hulcher, EH., R.D. MargoUs, and D.J. Bowman
223-228. (1978). Circadian rhythm of cholesterol-7a-
1040. Drover, VA., N.C. Wong, and L.B. Agellon hydroxylase and Cortisol in the African green
(2002). A distinct thyroid hormone response ele- monkey (Cercopithecus aethiops). Biochim.
ment mediates repression of the human choles- Biophys. Acta 529, 409-418.
terol 7a-hydroxylase (CYP7A1) gene promoter. 1052. Chiang, J.Y, WE Miller, and G.M. Lin (1990).
Mol. Endocrinol. 16, 14-23. Regulation of cholesterol 7 alpha-hydroxylase in
1041. Bjorkhem, I., Z. Araya, M. Rudling, B. Angelin, the liver. Purification of cholesterol 7a-hydroxy-
C. Einarsson, and K. Wikvall (2002). Differences lase and the immunochemical evidence for the
in the regulation of the classical and the alterna- induction of cholesterol 7a-hydroxylase by
tive pathv^ay for bile acid synthesis in human cholestyramine and circadian rhythm. J. Biol.
liver. No coordinate regulation of CYP7A1 and Chem. 265, 3889-3897.
CYP27A1. J. Biol. Chem. 277, 26804-26807. 1053. Kinowaki, M., S. Tanaka, Y Maeda, S. Higashi,
1042. Wang, D., D. Stroup, M. Marrapodi, M. Crestani, K. Okuda, and T. Setoguchi (2002). Half-life of
G. Galh, and J.Y.L. Chiang (1996). Transcriptional cholesterol 7a-hydroxylase activity and enzyme
regulation of the human cholesterol 7a-hydroxy- mass differ in animals and humans when deter-
lase gene {CYP7A) in HepG2 cells. J. Lipid Res. mined by a monoclonal antibody against human
37,1831-1841. cholesterol 7a-hydroxylase. J. Steroid Biochem.
1043. Kushwaha, R.S. and K.M. Born (1991). Mol. Biol. 81, 377-380.
Effect of estrogen and progesterone on the 1054. Noshiro, M., M. Nishimoto, and
hepatic cholesterol 7a-hydroxylase activity in K. Okuda (1990). Rat liver cholesterol
510 R Peter Guengerich

7a-hydroxylase: Pretranslational regulation for cir- 1067. Stahlberg, D., M. Rudling, B. Angelin, I. Bjorkhem,
cadian rhythm. J. Biol. Chem. 265, 10036-10041. P Forsell, K. Nilsell et al. (1997). Hepatic choles-
1055. Sanghvi, A., E. Grassi, V Warty, W. Diven, terol metabolism in human obesity. Hepatology 25,
C. Wight, and R. Lester (1981). Reversible 1447-1450.
activation-inactivation of cholesterol 7a- 1068. Post, S.M., E.C. de Wit, and H.M. Princen
hydroxylase possibly due to phosphorylation- (1997). Cafestol, the cholesterol-raising factor
dephosphorylation. Biochem. Biophys. Res. in boiled coffee, suppresses bile acid synthesis
Commun. 103, 886-892. by downregulation of cholesterol 7a-hydroxy-
1056. Goodwin, CD., B.W Cooper, and S. Margolis lase and sterol 27-hydroxylase in rat hepato-
(1982). Rat liver cholesterol 7a-hydroxylase: cytes. Arterioscler. Thromb. Vase. Biol. 17,
Modulation of enzyme activity by changes in 3064-3070.
phosphorylation state. J. Biol. Chem. 257, 1069. Pandak, W.M., C. Schwarz, PB. Hylemon,
4469-^472. D. Mallonee, K. Valerie, D.M. Heuman et al.
1057. Holsztynska, E.J. and D.J. Waxman (1987). (2001). Effects of CYP7A1 overexpression on cho-
Cytochrome P-450 cholesterol 7a-hydroxylase: lesterol and bile acid homeostasis. Am. J. Physiol.
Inhibition of enzyme deactivation by structurally Gastrointest. Liver Physiol. 281, G878-G889.
diverse calmodulin antagonists and phosphatase 1070. Sauter, G., M. Weiss, and R. Hoermann (1997).
inhibitors. Arch. Biochem. Biophys. 256, 543-559. Cholesterol 7a-hydroxylase activity in hypothy-
1058. Nguyen, L.B., S. Shefer, G. Salen, J.Y. Chiang, roidism and hyperthyroidism in humans. Horm.
and M. Patel (1996). Cholesterol 7a-hydroxylase Metah. Res. 29, 176-179.
activities from human and rat liver are modulated 1071. Pullinger, C.R., C. Eng, G. Salen, S. Shefer,
in vitro posttranslationally by phosphorylation/ A.K. Batta, S.K. Erickson et al. (2002). Human
dephosphorylation. Hepatology 24, 1468-1474. cholesterol 7a-hydroxylase (CYP7A1) defi-
1059. Karam, WG. and J.Y.L. Chiang (1992). Polymor- ciency has a hypercholesterolemic phenotype.
phisms of human cholesterol 7a-hydroxylase. J. Clin. Invest. 110, 109-117.
Biochem. Biophys. Res. Commun. 185, 588-595. 1072. Beigneux, A., A.F. Hofmann, and S.G. Young
1060. Setchell, K.D., M. Schwarz, N.C. O'Connell, (2002). Human CYP7A1 deficiency: Progress and
E.G. Lund, D.L. Davis, R. Lathe et al. (1998). enigmas. J. Clin. Invest. 110, 29-31.
Identification of a new inborn error in bile acid 1073. Stapleton, G., M. Steel, M. Richardson, J.O.
synthesis: Mutation of the oxysterol 7a-hydroxy- Mason, K.A. Rose, R.G.M. Morris et al. (1995).
lase gene causes severe neonatal liver disease. A novel cytochrome P450 expressed primarily in
J. Clin. Invest. 102, 1690-1703. brain. J. Biol. Chem. 270, 29739-29745.
1061. Hegele, R.A., J. Wang, S.B. Harris, J.H. Brunt, 1074. Rose, K.A., G. Stapleton, K. Dott, M.P Kieny,
T.K. Young, A.J. Hanley et al. (2001). Variable R. Best, M. Schwarz et al. (1997). Cyp7b, a novel
association between genetic variation in the brain cytochrome P450, catalyzes the synthesis of
CYP7 gene promoter and plasma lipoproteins in neurosteroids 7a-hydroxy dehydroepiandros-
three Canadian populations. Atherosclerosis 154, terone and 7a-hydroxy pregnenolone. Proc. Natl.
579-587. Acad Sci. USA 94, 4925-4930.
1062. Balasubramaniam, S., K.A. Mitropoulos, and 1075. Rose, K., A. Allan, S. Gauldie, G. Stapleton,
N.B. Myant (1975). The substrate for cholesterol L. Dobbie, K. Dott et al. (2001). Neurosteroid
7a-hydroxylase. Biochim. Biophys. Acta 398, hydroxylase CYP7B: Vivid reporter activity in
172-177. dentate gyrus of gene-targeted mice and aboli-
1063. Norlin, M., A. Toll, I. Bjorkhem, and K. Wikvall tion of a widespread pathway of steroid and
(2000). 24-hydroxycholesterol is a substrate for oxysterol hydroxylation. J. Biol. Chem. 276,
hepatic cholesterol 7a-hydroxylase (CYP7A). 23937-23944.
J. Lipid Res. 41, 1629-1639. 1076. Bunting, S., S. Moncada, and J.R. Vane (1983).
1064. Norlin, M., U. Andersson, I. Bjorkhem, and The prostacyclin-thromboxane A2 balance:
K. Wikvall (2000). Oxysterol 7a-hydroxylase Pathophysiological and therapeutic implications.
activity by cholesterol 7a-hydroxylase (CYP7A). Br Med Bull. 39,271-216.
J. Biol. Chem. 275, 34046-34053. 1077. Miyata, A., S. Hara, C. Yokoyama, H. Inoue,
1065. Lathe, R. (2002). Steroid and sterol 7-hydroxyla- V Ullrich, andT. Tanabe (1994). Molecular cloning
tion: Ancient pathways. Steroids 67, 967-977. and expression of human prostacyclin synthase.
1066. Nakayama, K., A. Puchkaev, and LA. Pikuleva Biochem. Biophys. Res. Commun. 200, 1728-1734.
(2001). Membrane binding and substrate access 1078. Ullrich, V, L. Castle, and P Weber (1981).
merge in cytochrome P450 7A1, a key enzyme in Spectral evidence for the cytochrome P450 nature
degradation of cholesterol. J. Biol. Chem. 276, of prostacyclin synthetase. Biochem. Pharmacol.
31459-31465. 30,2033-2036.
Human P450s 511

1079. DeWitt, D.L. and W.L. Smith (1983). Purification prostacyclin synthase gene variant with cerebral
of prostacyclin synthase from bovine aorta by infarction. Am. J. Hypertens. 13, 1263-1267.
immunoafifinity chromatography. Evidence that 1091. Nakayama, T., M. Soma, S. Saito, J. Honye,
the enzyme is a hemoprotein. J. Biol. Chem. 258, J. Yajima, D. Rahmutula et al. (2002). Association
3285-3293. of a novel single nucleotide polymorphism of the
1080. Kara, S., A. Miyata, C. Yokoyama, H. Inoue, prostacyclin synthase gene with myocardial
R. Brugger, F. Lottspeich et al. (1994). Isolation infarction. Am. Heart J. 143, 797-801.
and molecular cloning of prostacyclin synthase 1092. Nakayama, T., M. Soma, Y Takahashi,
from bovine endothelial cells. J. Biol Chem. 269, D. Rehemudula, H. Tobe, M. Sato et al. (2001).
19897-19903. Polymorphism of the promoter region of prosta-
1081. Spisni, E., G. Bartolini, M. Orlandi, B. Belletti, cyclin synthase gene is not related to essential
S. Santi, and V Tomasi (1995). Prostacyclin (PGj2) hypertension. Am. J. Hypertens. 14, 4 0 9 ^ 1 1 .
synthase is a constitutively expressed enzyme in 1093. Nakayama, T, M. Soma, D. Rahmutula, H. Tobe,
human endothelial cells. Exp. Cell Res. 219, M. Sato, J. Uwabo et al. (2002). Association
507-513. study between a novel single nucleotide polymor-
1082. Mehl, M., H.J. Bidmon, H. Hilbig, K. Zilles, phism of the promoter region of the prostacyclin
R. Dringen, and V Ullrich (1999). Prostacyclin synthase gene and essential hypertension.
synthase is localized in rat, bovine and human Hypertens. Res. 25, 65-68.
neuronal brain cells. Neurosci. Lett. 271,187-190. 1094. Nakayama, T, M. Soma, Y Watanabe, B. Hasimu,
1083. Siegle, I., T. Klein, M.H. Zou, R Fritz, and M. Sato, N. Aoi et al. (2002). Splicing mutation
M. Komhofif (2000). Distribution and cellular of the prostacyclin synthase gene in a family
localization of prostacyclin synthase in human associated with hypertension. Biochem. Biophys.
brain. J. Histochem. Cytochem. 48, 631-641. Res. Commun. 297, 1135-1139.
1084. Huang, J.C, F Arbab, K.J. Tumbusch, J.S. Goldsby, 1095. Hatae, T, S. Hara, C. Yokoyama, T. Yabuki,
N. Matijevic-Aleksic, and K.K. Wu (2002). Human H. Inoue, V Ullrich et al. (1996). Site-directed
fallopian tubes express prostacyclin (PGI) synthase mutagenesis of human prostacyclin synthase:
and cyclooxygenases and synthesize abundant PGI. Alteration of Cys"^^^ of the Cys-pocket, and
J. Clin. Endocrinol. Metab. 87, 4361^368. Glu347 and Arg350 of the EXXR motif FEBSLett.
1085. Yokoyama, C., T. Yabuki, H. Inoue, Y. Tone, 389, 268-272.
S. Hara, T. Hatae et al. (1996), Human gene 1096. Shyue, S.K., K.H. Ruan, L.H. Wang, and K.K. Wu
encoding prostacyclin synthase (PTGIS): (1997). Prostacyclin synthase active sites.
Genomic organization, chromosomal localization, Identification by molecular modeling-guided site-
and promoter activity. Genomics 36, 296-304. directed mutagenesis. J. Biol. Chem. 272,
1086. Nakayama, T., M. Soma, Y. Izumi, and 3657-3662.
K. Kanmatsuse (1996). Organization of the human 1097. Lin, Y, K.K. Wu, and K.H. Ruan (1998).
prostacyclin synthase gene. Biochem. Biophys. Characterization of the secondary structure and
Res. Commun. Ill, 803-806. membrane interaction of the putative membrane
1087. Wang, L.H. and L. Chen (1996). Organization anchor domains of prostaglandin I2 synthase and
of the gene encoding human prostacyclin cytochrome P450 2C1. Arch. Biochem. Biophys.
synthase. Biochem. Biophys. Res. Commun. 226, 352, 78-84.
631-637. 1098. Lin, YZ., H. Deng, and K.H. Ruan (2000).
1088. ChevaUer, D., C. Cauffiez, C. Bernard, Topology of catalytic portion of prostaglandin I2
J.M. Lo-Guidice, D. AUorge, F. Fazio et al. synthase: Identification by molecular modeling-
(2001). Characterization of new mutations in the guided site-specific antibodies. Arch. Biochem.
coding sequence and 5'-untranslated region of Biophys. 379, 188-197.
the human prostacylcin synthase gene 1099. Deng, H., A. Huang, S.R So, YZ. Lin, and
(CYP8A1). ifi/m. Genet. 108, 148-155. K.H. Ruan (2002). Substrate access channel
1089. Chevalier, D., D. Allorge, J.M. Lo-Guidice, topology in membrane-bound prostacyclin syn-
C. Cauffiez, C. Lepetit, F. Migot-Nabias et al. thase. Biochem. J. 362, 545-551.
(2002). Sequence analysis, frequency and ethnic 1100. Reed, G.A., I.O. Griffin, and T.E. Eling (1985).
distribution of VNTR polymorphism in the 5'- Inactivation of prostaglandin H synthase and
untranslated region of the human prostacyclin prostacyclin synthase by phenylbutazone.
synthase gene (CYP8A1). Prostaglandins Other Requirement for peroxidative metabolism. Mol.
Lipid Mediat. 70, 31-37. Pharmacol. 21, 109-114.
1090. Nakayama, T, M. Soma, D. Rehemudula, 1101. Wade, M.L., N.F Voelkel, and FA. Fitzpatrick
Y Takahashi, H. Tobe, M. Satoh et al. (2000). (1995). "Suicide" inactivation of prostaglandin I2
Association of 5' upstream promoter region of synthase: Characterization of mechanism-based
512 F. Peter Guengerich

inactivation with isolated enzyme and endothelial human and mouse: Eukaryotic cytochrome P-450
cells. Arch. Biochem. Biophys. 321, 453-458. gene devoid of introns. Genomics 56, 184-196.
1102. Zou, M.H. and V Ullrich (1996). Peroxynitrite 1113. Zhang, M. and J.Y Chiang (2001).
formed by simultaneous generation of nitric Transcriptional regulation of the human sterol
oxide and superoxide selectively inhibits bovine 12a-hydroxylase gene (CYPSBl): Roles of
aortic prostacyclin synthase. FEBS Lett. 382, hepatocyte nuclear factor 4a in mediating
101-104. bile acid repression. J. Biol. Chem. 276,
1103. Crow, J.P. and J.S. Beckman (1995). Reactions 41690-41699.
between nitric oxide, superoxide, and peroxyni- 1114. Gerbod-Giannone, M.C., A. Del Castillo-
trite: Footprints of peroxynitrite in vivo. Adv. Olivares, S. Janciauskiene, G. Gil, and
Pharmacol. 34, 1 7 ^ 3 . P.B. Hylemon (2002). Suppression of cholesterol
1104. Zou, M., C. Martin, and V Ullrich (1997). 7a-hydroxylase transcription and bile acid syn-
Tyrosine nitration as a mechanism of selective thesis by an a j-antitrypsin peptide via interaction
inactivation of prostacyclin synthase by peroxyni- with a J-fetoprotein transcription factor. J. Biol.
trite. Biol. Chem. 378, 707-713. Chem. 211, 42973^2980.
1105. Schmidt, P., N. Youhnovski, A. Daiber, A. Balan, 1115. Yang, Y, M. Zhang, G. Eggertsen, and J.Y Chiang
M. Arsic, M. Bachschmid et al. (2003). Specific (2002). On the mechanism of bile acid inhibition
nitration at tyrosine-430 revealed by high resolu- of rat sterol 12a-hydroxylase gene (CYPSBl)
tion mass spectrometry as basis for redox regula- transcription: Roles of a-fetoprotein transcription
tion of bovine prostacyclin synthase. J. Biol. factor and hepatocyte nuclear factor 4a. Biochim.
Chem. 278, 12813-12819. Biophys. Acta 1583, 63-73.
1106. Tuder, R.M., CD. Cool, M.W. Geraci, J. Wang, 1116. Wang, H.P and T. Kimura (1976). Purification
S.H. Abman, L. Wright et al. (1999). Prostacyclin and characterization of adrenal cortex mitochon-
synthase expression is decreased in lungs from drial cytochrome P-450 specific for cholesterol
patients with severe pulmonary hypertension. side chain cleavage activity. J. Biol. Chem. 251,
Am. J. Respir. Crit. Care Med. 159, 1925-1932. 6068-6074.
1107. Iwai, N., T. Katsuya, K. Ishikawa, T. Mannami, 1117. Morohashi, K., K. Sogawa, T Omura, and
J. Ogata, J. Higaki et al. (1999). Human prostacy- Y Fujii-Kuriyama (1987). Gene structure of
clin synthase gene and hypertension: The Suita human cytochrome P-450(SCC), cholesterol
study Circulation 100, 2231-2236. desmolase. J. Biochem. (Tokyo) 101, 8879-8887.
1108. Geraci, M.W, B. Gao, D.C. Shepherd, M.D. Moore, 1118. Poulos, T.L., B.C. Finzel, l.C. Gunsalus,
J.Y. Westcott, K.A. Pagan et al. (1999). Pulmonary G.C. Wagner, and J. Kraut (1985). The 2.6-A crys-
prostacyclin synthase overexpression in transgenic tal structure of Pseudomonas putida cytochrome
mice protects against development of hypoxic P-450. J. Biol. Chem. 260, 16122-16130.
pulmonary hypertension. J. Clin. Invest. 103, 1119. Belfiore, C.J., D.E. Hawkins, M.C. Wiltbank, and
1509-1515. G.D. Niswender (1994). Regulation of
1109. Todaka, T, C. Yokoyama, H. Yanamoto, cytochrome P450g^^ synthesis and activity in the
N. Hashimoto, I. Nagata, T. Tsukahara et al. ovine corpus luteum. J. Steroid Biochem. Mol.
(1999). Gene transfer of human prostacyclin syn- Biol. 51,283-290.
thase prevents neointimal formation after carotid 1120. Matocha, M.F and M.R. Waterman (1986).
balloon injury in rats. Stroke 30, 419-426. Import and processing of P-450g^,^ and P-4501 Ip
1110. Pradono, P., R. Tazawa, M. Maemondo, precursors by corpus luteal mitochondria: A pro-
M. Tanaka, K. Usui, Y Saijo et al. (2002). Gene cessing pathway recognizing homologous and
transfer of thromboxane A2 synthase and heterologous precursors. Arch. Biochem. Biophys.
prostaglandin I2 synthase antithetically altered 250, 456^60.
tumor angiogenesis and tumor growth. Cancer 1121. Sasano, H., M. Okamoto, J.l. Mason,
Res 62, 63-66. E.R. Simpson, C.R. Mendelson, N. Sasano et al.
nil. Leeder, J.S., X. Lu, Y Timsit, and A. Gaedigk (1989). Immunolocalization of aromatase, 17a-
(1998). Non-monooxygenase cytochromes P450 hydroxylase and side-chain-cleavage cytochromes
as potential human auto antigens in anticonvulsant P-450 in the human ovary. J. Reprod. Fertil. 85,
hypersensitivity reactions. Pharmacogenetics 8, 163-169.
211-225. 1122. Chung, B.C., l.C. Guo, and S.J. Chou (1997).
1112. Gafvels, M., M. Olin, B.P Chowdhary, Transcriptional regulation of the CYPllAl and
T. Raudsepp, U Andersson, B. Persson et al. ferredoxin genes. Steroids 62, 3 7 ^ 2 .
(1999). Structure and chromosomal assignment 1123. Walther, B., J.R Ghersi-Egea, A. Minn, and
of the sterol 12a-hydroxylase gene (CYP8B1) in G. Siest (1987). Brain mitochondrial cytochrome
Human P450s 513

P-450g^^: Spectral and catalytic properties. Arch. 1136. Liu, Z. and E.R. Simpson (1999). Molecular
Biochem. Biophys. 254, 592-596. mechanism for cooperation between Spl and
1124. Warner, M. and J.A. Gustafsson (1995). steroidogenic factor-1 (SF-1) to regulate bovine
Cytochrome P450 in the brain: Neuroendocrine CYPl 1A gene expression. Mol. Cell Endocrinol.
functions. Front. Neuroendocrinol. 16, 224-236. 153,183-196.
1125. Beyenburg, S., B. Stoffel-Wagner, M. Watzka, 1137. Gizard, F, B. Lavallee, F DeWitte, and D.W Hum
I. Blumcke, J. Bauer, J. Schramm et al. (1999). (2001). A novel zinc finger protein TReP-132 inter-
Expression of cytochrome P450g^^ mRNA in the acts with CBP/p300 to regulate human CYPllAl
hippocampus of patients with temporal lobe gene expression. J. Biol. Chem. 276, 33881-33892.
epilepsy. Neuroreport 10, 3067-3070. 1138. Gizard, F, B. Lavallee, F DeWitte, E. Teissier,
1126. Watzka, M., F. Bidlingmaier, J. Schramm, B. Staels, and D.W. Hum (2002). The transcrip-
D. KlingmuUer, and B. StofiFel-Wagner (1999). tional regulating protein of 132 kDa (TReP-132)
Sex- and age-specific differences in human brain enhances P450scc gene transcription through
CYPllAl mRNA expression. J. Neuroendocrinol. interaction with steroidogenic factor-1 in human
II, 901-905. adrenal cells. J. Biol. Chem. Ill, 39144-39155.
1127. Morales, A., A. Cuellar, J. Ramirez, F. Vilchis, 1139. Doi, J., H. Takemori, X.Z. Lin, N. Horike,
and V Diaz-Sanchez (1999). Synthesis of steroids Y Katoh, and M. Okamoto (2002). Salt-inducible
in pancreas: Evidence of cytochrome P-450g^^ kinase represses cAMP-dependent protein
activity. Pancreas 19, 39-44. kinase-mediated activation of human cholesterol
1128. Ou, W, A. Ito, K. Morohashi, Y. Fujii-Kuriyama, side chain cleavage cytochrome P450 promoter
and T. Omura (1986). Processing-independent through the CREB basic leucine zipper domain.
in vitro translocation of cytochrome P-450g(^j^ J. Biol. Chem. Ill, 15629-15637.
precursor across mitochondrial membranes. 1140. Ben-Zimra, M., M. Koler, and J. Orly (2002).
J. Biochem. (Tokyo) 100, 1287-1296. Transcription of cholesterol side-chain cleavage
1129. Kumamoto, T., K. Morohashi, A. Ito, and cytochrome P450 in the placenta: Activating
T. Omura (1987). Site-directed mutagenesis of protein-2 assumes the role of steroidogenic
basic amino acid residues in the extension peptide factor-1 by binding to an overlapping promoter
of P-450g(^(-, precursor: Effects on the import of element. Mol. Endocrinol. 16, 1864-1880.
the precursor into mitochondria. J. Biochem. 1141. Katsumata, N., M. Ohtake, T. Hojo, E. Ogawa,
(Tokyo) 102, 833-838. T. Hara, N. Sato et al. (2002). Compound het-
1130. Black, S.M., J.A. Harikrishna, G.D. Szklarz, and erozygous mutations in the cholesterol side-chain
WL. Miller (1994). The mitochondrial environ- cleavage enzyme gene (CYPl 1 A) cause congeni-
ment is required for activity of the cholesterol tal adrenal insufficiency in humans. J. Clin.
side-chain cleavage enzyme, cytochrome P450g^^.. Endocrinol. Metab. 87, 3808-3813.
Proc. Natl. Acad. Sci. USA 91, 7247-7251. 1142. Tuckey, R.C. and K.J. Cameron (1993). Human
1131. Venepally, R and M.R. Waterman (1995). Two placental cholesterol side-chain cleavage:
Spl-binding site mediate cAMP-induced tran- Enzymatic synthesis of (22/?)-20a,22-dihydroxy-
scription of the bovine CYPl 1A gene through the cholesterol. Steroids 58, 230-233.
protein kinase A signaling pathway. J. Biol. 1143. Murray, R.I. and S.G. Sligar (1985). Oxidative
Chem. 270, 25402-25410. cleavage of 1-phenyl-1,2-ethanediol by 4-cyano-
1132. Ahlgren, R., G. Suske, M.R. Waterman, and A^,A^-dimethylaniline N-oxide and chloro
J. Lund (1999). Role of Spl in cAMP-dependent (5,10,15,20-tetraphenylporphinato) chromium
transcriptional regulation of the bovine CYPl 1A (III): A model for cholesterol side-chain cleavage
gene. J. Biol. Chem. 274, 19422-19428. by cytochrome P-450g^,^. J. Am. Chem. Soc. 107,
1133. Guo, I.e. and B.C. Chung (1999). Cell-type 2186-2187.
specificity of human CYPllAl TATA box. 1144. Okamoto, T., K. Sasaki, and S. Oka (1988).
J. Steroid Biochem. Mol. Biol. 69, 329-334. Biomimetic oxidation with molecular oxygen.
1134. Huang, Y, M. Hu, N. Hsu, C.L. Wang, and Selective carbon-carbon bond cleavage of 1,2-
B. Chung (2001). Action of hormone responsive diols by molecular oxygen and dihydropyridine in
sequence in 2.3 kb promoter of CYPllAl. Mol. the presence of iron-porphyrin catalysts. J. Am.
Cell Endocrinol. 175,205-210. Chem. Soc. 110, 1187-1196.
1135. Hu, M.C., N.C. Hsu, C.I. Pai, C.K. Wang, and 1145. Ortiz de Montellano, PR. (1995). Oxygen activa-
B. Chung (2001). Functions of the upstream and tion and reactivity. In RR. Ortiz de Montellano
proximal steroidogenic factor 1 (SF-l)-binding sites (ed.), Cytochrome P450: Structure, Mechanism,
in the CYPllAl promoter in basal transcription and and Biochemistry. Plenum Press, New York,
hormonal response. Mol. Endocrinol. 15, 812-818. pp. 245-303.
514 F. Peter Guengerich

1146. Tuckey, R.C., S.T. Woods, and M. Tajbakhsh 1158. Hu, M.C., N.C. Hsu, N.B. El Hadj, C.I. Pai,
(1997). Electron transfer to cytochrome P-450g^^ H.P Chu, C.K. Wang et al. (2002). Steroid
limits cholesterol-side-chain-cleavage activity in deficiency syndromes in mice with targeted disrup-
the human placenta. Eur. J. Biochem. 244, tion of Cyp] J al. Mol. Endocrinol. 16, 1943-1950.
835-839. 1159. Chen, S., J. Sawicka, C. Betterle, M. Powell,
1147. Beckert, V and R. Bernhardt (1997). Specific L. Prentice, M. Volpato et al. (1996).
aspects of electron transfer from adrenodoxin to Autoantibodies to steroidogenic enzymes in
cytochromes P450g^^ and P450jjg. J. Biol. Chem. autoimmune polyglandular syndrome, Addison's
272, 4883-4888. disease, and premature ovarian failure. J. Clin.
1148. Cao, P. and R. Bernhardt (1999). Interaction of Endocrinol. Metab. 81, 1871-1876.
CYPllBl (cytochrome P-450,,p) with 1160. Seissler, J., M. Schott, H. Steinbrenner,
CYPl 1 Al (cytochrome P-450^ J in COS-1 cells. P Peterson, and WA. Scherbaum (1999).
Eur. J. Biochem. 262, 720-726. Autoantibodies to adrenal cytochrome P450 anti-
1149. Usanov, S.A. and VL. Chashchin (1991). gens in isolated Addison's disease and autoim-
Interaction of cytochrome P-450^^^ with mune polyendocrine syndrome type II. Exp. Clin.
cytochrome b^. FEBS Lett. 278, 279-282. Endocrinol. Diabetes 107, 208-213.
1150. Wada, A. and M.R. Waterman (1992). 1161. Bureik, M., M. Lisurek, and R. Bernhardt (2002).
Identification by site-directed mutagenesis of two The human steroid hydroxylases CYPllBl and
lysine residues in cholesterol side chain cleavage CYP11B2. Biol. Chem. 383, 1537-1551.
cytochrome P450 that are essential for adreno- 1162. Watanuki, M., B.E. Tilley and PE Hall (1978).
doxin binding. J. Biol. Chem. 267, 22877-22882. Cytochrome P-450 for 11(3- and 18-hydroxylase
1151. Woods, S.T, J. Sadleir, T. Downs, activities of bovine adrenocortical mitochondria:
T. Triantopoulos, M.J. Headlam, and R.C. Tuckey One enzyme or two?. Biochemistry 17, 127-130.
(1998). Expression of catalytically active human 1163. Mornet, E., J. Dupont, A. Vitek, and PC. White
cytochrome P450g^^ in Escherichia coli and muta- (1989). Characterization of two genes encoding
genesis of isoleucine-462. Arch. Biochem. human steroid 11 beta-hydroxylase (P-450(ll)p).
Biophys. 353, 109-115. J. Biol. Chem. 264, 20961-20967.
1152. Vickery, L.E. and J.T Kellis (1983). Inhibition of 1164. Kawamoto, T, Y Mitsuuchi, K. Toda,
adrenocortical cytochrome P-450^^^ by (20/?)-20- K. Miyahara, Y Yokoyama, K. Nakao et al.
phenyl-5-pregnene-3(3,20-diol: Mechanism and (1990). Cloning of cDNA and genomic DNA for
implications for the structure of the active site. human cytochrome P-450,,^. FEBS Lett. 269,
J. Biol. Chem. 258, 3832-3836. 345-349.
1153. Vickery, L.E. and J. Singh (1988). 22- 1165. Zhang, G. and WL. Miller (1996). The human
Thio-23,24-bisnor-5-cholen-3 P-ol: An active genome contains only two CYPl IB (P450cll)
site-directed inhibitor of cytochrome P450j,^^. genes. ^ Clin. Endocrinol. Metab. 81, 3254-3256.
J. Steroid Biochem. 29, 539-543. 1166. Kawamoto, T, Y Mitsuuchi, K. Toda,
1154. Olakanmi, O. and D.W Seybert (1990). Modified Y Yokoyama, K. Miyahara, S. Miura et al.
acetylenic steroids as potent mechanism-based (1992). Role of steroid 11 (3-hydroxylase and
inhibitors of cytochrome P-450g^^. J. Steroid steroid 18-hydroxylase in the biosynthesis of glu-
Biochem. 36, 273-280. cocorticoids and mineralocorticoids in humans.
1155. Jarman, M., S.E. Barrie, C.S. Leung, and M.G. Proc. Natl. Acad Sci. USA 89, 1458-1462.
Rowlands (1988). Selective inhibition of choles- 1167. Freije, W.A., V Pezzi, A. Arici, B.R. Carr, and
terol side-chain cleavage by potential pro-drug WE. Rainey (1997). Expression of 11 beta-
forms of aminoglutethimide. Anticancer Drug hydroxylase (CYPllBl) and aldosterone syn-
Design 3, 185-190. thase (CYP11B2) in the human fetal adrenal.
1156. Ohnishi, T and Y. Ichikawa (1997). Direct J. Soc. Gynecol. Investig. 4, 305-309.
inhibitions of the activities of steroidogenic 1168. Morohashi, K., U.M. Zanger, S. Honda, M. Hara,
cytochrome P-450 mono-oxygenase systems by M.R. Waterman, andT Omura (1993). Activation
anticonvulsants. J. Steroid Biochem. Mol. Biol. of CYPl 1A and CYPl 1B gene promoters by the
60, 77-85. steroidogenic cell-specific transcription factor,
1157. Yang, X., K. Iwamoto, M. Wang, J. Artwohl, Ad4BR Mol. Endocrinol. 7, 1196-1204.
J.I. Mason, and S. Pang (1993). Inherited congen- 1169. Honda, S., K. Morohashi, M. Nomura, H. Takeya,
ital adrenal hyperplasia in the rabbit is caused M. Kitajima, and T Omura (1993). Ad4BP regu-
by a deletion in the gene encoding cytochrome lating steroidogenic P-450 gene is a member of
P450 cholesterol side-chain cleavage enzyme. steroid hormone receptor superfamily. J. Biol.
Endocrinology 132, 1977-1982. Chem. 268, 7494-7502.
Human P450s 515

1170. Hashimoto, T., K. Morohashi, K. Takayama, splice donor sites of CYPl IB 1 in congenital
5. Honda, T. Wada, H. Handa et al (1992). adrenal hyperplasia due to 11 p-hydroxylase
Cooperative transcription activation between deficiency. Endocrinol. Res. 26, 797-801.
Adl, a CRE-like element, and other elements in 1181. Mulatero, P., K.M. Curnow, B. Aupetit-Faisant,
the CYPl IB gene promoter. J. Biochem. (Tokyo) M. Foekling, C. Gomez-Sanchez, F. Veglio,
112, 573-575. X. Jeunemaitre et al. (1998). Recombinant
1171. Mukai, K., F. Mitani, R. Agake, and Y. Ishimura CYPl IB genes encode enzymes that can catalyze
(1998). Adrenocorticotropic hormone stimulates conversion of 11-deoxycortisol to Cortisol, 18-
CYPl IB 1 gene transcription through a mecha- hydroxycortisol, and 18-oxocortisol. J. Clin.
nism involving AP-1 factors. Eur. J. Biochem. Endocrinol. Metab. 83, 3996-4001.
256, 190-200. 1182. Fisher, A., E. Davies, R. Eraser, and J.M. Connell
1172. Wang, X.L., M. Bassett, Y Zhang, S. Yin, C. Clyne, (1998). Structure-function relationships of aldos-
PC. White et al. (2000). Transcriptional regu- terone synthase and 11 ^-hydroxylase enzymes:
lation of human 11 ^-hydroxylase QiCYPllBl). Implications for human hypertension. Clin. Exp.
Endocrinology 141, 3587-3594. Pharmacol. Physiol. Suppl. 25, S42-S46.
1173. Bassett, M.H., Y Zhang, C. Clyne, PC. White, 1183. Fisher, A., R. Eraser, J. McConnell, and E. Davies
and WE. Rainey (2002). Differential regulation (2000). Amino acid residue 147 of human aldos-
of aldosterone synthase and 11 (^-hydroxylase terone synthase and 11 (3-hydroxylase plays a key
transcription by steroidogenic factor-1. J. Mol. role in 11 ^-hydroxylation. J. Clin. Endocrinol.
Endocrinol. 28, 125-135. Metab. 85, 1261-1266.
1174. Skinner, C.A. and G. Rumsby (1994). Steroid 11 1184. Bottner, B., K. Denner, and R. Bernhardt (1998).
beta-hydroxylase deficiency caused by a five Conferring aldosterone synthesis to human
base pair duplication in the CYPl IB 1 gene. CYPl IB 1 by replacing key amino acid residues
Hum. Mol. Genet. 3, 377-378. with CYPllB2-specific ones. Eur. J. Biochem.
1175. Curnow, K.M., L. Slutsker, J. Vitek, T. Cole, 252, 458^66.
PW Speiser, M.I. New et al. (1993). Mutations in 1185. Lewis, D.E and P Lee-Robichaud (1998). Mole-
the CYPl IB 1 gene causing congenital adrenal cular modelling of steroidogenic cyto-chromes
hyperplasia and hypertension cluster in exons P450 from families CYPll, CYP17, CYP19 and
6, 7, and 8. Proc. Natl. Acad. Sci. USA 90, CYP21 based on the CYPl02 crystal structure.
4552-4556. J. Steroid Biochem. Mol. Biol. 66, 217-233.
1176. Lifton, R.P, R.G. Dluhy, M. Powers, G.M. Rich, 1186. Belkina, N.V, M. Lisurek, A.S. Ivanov, and
S. Cook, S. UHck et al. (1992). A chimaeric 11 (3- R. Bernhardt (2001). Modelling of three-
hydroxylase/aldosterone synthase gene causes dimensional structures of cytochromes P450
glucocorticoid-remediable aldosteronism and l l B l and 11B2. J. Inorg. Biochem. 87, 197-207.
human hypertension. Nature 355, 262-265. 1187. Denner, K., R. Vogel, W Schmalix, J. Doehmer,
1177. Pascoe, L., K.M. Curnow, L. Slutsker, and R. Bernhardt (1995). Cloning and stable
J.M. Connell, PW. Speiser, M.I. New et al. (1992). expression of the human mitochondrial cyto-
Glucocorticoid-suppressible hyperaldosteronism chrome P45011B1 cDNA in V79 Chinese ham-
results from hybrid genes created by unequal ster cells and their application for testing of
crossovers between CYPl IB 1 and CYP11B2, potential inhibitors. Pharmacogenetics 5, 89-96.
Proc. Natl. Acad Sci. USA 89, 8327-8331. 1188. Johnston, J.O., C.L. Wright, and G.W Holbert
1178. Hampf, M., N.T. Dao, N.T. Hoan, and (1995). Enzyme-activated inhibitors of steroidal
R. Bernhardt (2001). Unequal crossing-over hydroxylases. J. Steroid Biochem. Mol. Biol. 52,
between aldosterone synthase and 11 p-hydroxy- 17-34.
lase genes causes congenital adrenal hyperplasia. 1189. Delorme, C , A. Piffeteau, E Sobrio, and
J. Clin. Endocrinol. Metab. 86, 4445^452. A. Marquet (1997). Mechanism-based inactivation
1179. Portrat, S., P Mulatero, K.M. Curnow, of bovine cytochrome P450jjp by 18-unsaturated
J.L. Chaussain, Y Morel, and L. Pascoe (2001). progesterone derivatives. Eur. J. Biochem. 248,
Deletion hybrid genes, due to unequal crossing 252-260.
over between CYPl IB 1 (11(3-hydroxylase) and 1190. White, PC. (2001). Steroid 11 ^-hydroxylase
CYP11B2 (aldosterone synthase) cause steroid deficiency and related disorders. Endocrinol.
11 p-hydroxylase deficiency and congenital adre- Metab. Clin. North Am. 30, 61-79.
nal hyperplasia. J. Clin. Endocrinol. Metab. 86, 1191. Peter, M., J.M. Dubuis, and WG. Sippell (1999).
3197-3201. Disorders of the aldosterone synthase and steroid
1180. Chabre, O., S. Portrat-Doyen, J. Vivier, Y Morel, 11 ^-hydroxylase deficiencies. Horm. Res. 51,
and G. Defaye (2000). Two novel mutations in 211-222.
516 F. Peter Guengerich

1192. Pascoe, L., K.M. Curnow, L. Slutsker, A. Rosier, corticosterone methyl oxidase II deficiency. Am.
and P.C. White (1992). Mutations in the human J. Hum. Genet. 57, 1037-1043.
CYP11B2 (aldosterone synthase) gene causing 1204. Mulatero, P, D. Schiavone, F. Fallo, F Rabbia,
corticosterone methyloxidase II deficiency. Proc. C. Pilon, L. Chiandussi et al. (2000). CYPl 1B2
Natl. Acad. Sci. USA 89, 4996-5000. gene polymorphisms in idiopathic hyperaldos-
1193. Mitsuuchi, Y., T. Kawamoto, K. Miyahara, teronism. Hypertension 35, 694-698.
S. Ulick, D.H. Morton, Y. Naiki et al. (1993). 1205. Tsukada, K., T. Ishimitsu, M. Teranishi, M. Saitoh,
Congenitally defective aldosterone biosynthesis M. Yoshii, H. Inada et al. (2002). Positive associa-
in humans: Inactivation of the P-450C18 gene tion of CYPl 1B2 gene polymorphism with genetic
(CYPl 1B2) due to nucleotide deletion in CMO I predisposition to essential hypertension. J. Hum.
deficient patients. Biochem. Biophys. Res. Hypertens. 16, 789-793.
Commun. 190, 864-869. 1206. Bechtel, S., N. Belkina, and R. Bernhardt (2002).
1194. Kawamoto, T., Y Mitsuuchi, T. Ohnishi, The effect of amino-acid substitutions II12P, D147E
Y Ichikawa, Y Yokoyama, H. Sumimoto et al. and K152N in CYPl 1B2 on the catalytic activities
(1990). Cloning and expression of a cDNA for of the enzyme. Eur J. Biochem. 269, 1118-1127.
human cytochrome P-450^J^Q as related to 1207. Ehmer, PB., M. Bureik, R. Bernhardt, U. Muller,
primary aldosteronism. Biochem. Biophys. Res. and R.W Hartmann (2002). Development of a
Commun. 173,309-316. test system for inhibitors of human aldosterone
1195. Curnow, K.M., M.T. Tusie-Luna, L. Pascoe, synthase (CYP11B2): Screening in fission yeast
R. Natarajan, J.L. Gu, J.L. Nadler et al. (1991). and evaluation of selectivity in V79 cells.
The product of the CYPl 1B2 gene is required for J. Steroid Biochem. Mol. Biol. 81, 173-179.
aldosterone biosynthesis in the human adrenal 1208. GrifFing, G.T, M. Holbrook, J.C. Melby J. Alberta,
cortex. Mol. Endocrinol. 5, 1513-1522. and N.R. Orme-Johnson (1989). 19-Hydroxylase
1196. Li, X., Y Meng, X.S. Yang, PS. Wu, S.M. Li, and inhibition of adrenal mitochondrial P450
W.Y Lai (2000). CYP11B2 expression in HSCs 1 ip/18/19-hydroxylase by a suicide inhibitor. Am.
and its effect on hepatic fibrogenesis. World J. Med Sci. 298, 83-88.
J. Gastroenterol. 6, 885-887. 1209. Davies, E., CD. Holloway, M.C Ingram,
1197. Bassett, M.H., Y Zhang, PC. White, and G.C Inglis, E.C Friel, C Morrison et al. (1999).
WE. Rainey (2000). Regulation of human Aldosterone excretion rate and blood pressure in
CYPl 1B2 and CYPl IBl: Comparing the role of essential hypertension are related to polymorphic
the common CRE/Adl element. Endocrinol. Res. differences in the aldosterone synthase gene
26,941-951. CYPl 1B2. Hypertension 33, 703-707.
1198. Clyne, CD., PC. White, and WE. Rainey (1996). 1210. White, P C , A. Hautanen, and M. Kupari (1999).
Calcium regulates human CYP11B2 transcrip- Aldosterone synthase (CYP11B2) polymor-
tion. Endocrinol. Res. 22, 485-492. phisms and cardiovascular function. J. Steroid
1199. Bureik, M., A. Zeeh, and R. Bernhardt (2002). Biochem. Mol. Biol. 69, 409-412.
Modulation of steroid hydroxylase activity in sta- 1211. Kupari, M., A. Hautanen, L. Lankinen,
bly transfected V79MZhl 1 Bl and V79MZhl 1B2 P. Koskinen, J. Virolainen, H. Nikkila et al.
cells by PKC and PKD inhibitors. Endocrinol. (1998). Associations between human aldosterone
Res. 28,351-355. synthase (CYP11B2) gene polymorphisms and
1200. J.G. LeHoux, G. Dupuis, and A. Lefebvre (2000). left ventricular size, mass, and function.
Regulation of CYPl 1B2 gene expression by pro- Circulation 97, 569-575.
tein kinase C. Endocrinol. Res. 26, 1027-1031. 1212. Satoh, M., M. Nakamura, H. Saitoh, H. Satoh,
1201. Ise, T., A. Shimoda, H. Takakuwa, T. Kato, T Akatsu, J. Iwasaka et al. (2002). Aldosterone
Y Izumiya, K. Shimizu et al. (2001). A chimeric synthase (CYP11B2) expression and myocardial
CYP11B1/CYP11B2 gene in glucocorticoid- fibrosis in the failing human heart. Clin. Sci.
insuppressible familial hyperaldosteronism. Clin. (Zo«Jo«; 102,381-386.
Endocrinol. (Oxford) 55, 131-134. 1213. Russo, P, A. Siani, A. Venezia, R. lacone,
1202. Jackson, R.V, A. Laflferty, D.J. Torpy, and O. Russo, G. Barba et al. (2002). Interaction
C. Stratakis (2002). New genetic insights in between the C(-344)T polymorphism of CYPl 1B2
familial hyperaldosteronism. Ann. N. Y. Acad. Sci. and age in the regulation of blood pressure and
970, 77-88. plasma aldosterone levels: Cross-sectional and lon-
1203. Zhang, G., H. Rodriguez, C.E. Fardella, gitudinal findings of the Olivetti Prospective Heart
D.A. Harris, and WL. Miller (1995). Mutation Study J. Hypertens. 20, 1785-1792.
T318M in the CYP11B2 gene encoding 1214. Lim, PO., T.M. Macdonald, C Holloway,
P450cllAS (aldosterone synthase) causes E. Friel, N.H. Anderson, E. Dow et al. (2002).
Human P450s 517

Variation at the aldosterone synthase (CYPl 1B2) by an androgen receptor-mediated mechanism.


locus contributes to hypertension in subjects with J. Biol. Chem. 262, 11200-11206.
a raised aldosterone-to-renin ratio. J. Clin. 1226. Zhang, P, X.G. Han, S.H. Mellon, and PR Hall
Endocrinol. Metab. 87, 4398^402. (1996). Expression of the gene for cytochrome
1215. Tsujita, Y., N. Iwai, T. Katsuya, J. Higaki, P-450 17a-hydroxylase/C 17-20 lyase (CYPl7)
T. Ogihara, S. Tamaki et al (2001). Lack of in porcine Leydig cells: Identification of a DNA
association between genetic polymorphism of sequence that mediates cAMP response. Biochim.
CYP11B2 and hypertension in Japanese: The Biophys. Acta 1307, 73-82.
Suita Study. Hypertens. Res. 24, 105-109. 1227. Ogo, A., M.R. Waterman, J.M. McAllister, and
1216. Chung, B., J. Picado-Leonard, M. Haniu, N. Kagawa (1997). The homeodomain protein
M. Bienkowski, PR Hall, J.E. Shively et al. (1987). Pbxl is involved in cAMP-dependent transcrip-
Cytochrome P450cl7 (steroid 17a-hydroxylase/17, tion of human CYPl7. Arch. Biochem. Biophys.
20 lyase): Cloning of human adrenal and testis 348,226-231.
cDNAs indicates the same gene is expressed in both 1228. Bischof, L.J., N. Kagawa, and M.R. Waterman
tissues. Proc. Natl. Acad. Sci. USA 84, 407-411. (1998). The bovine CYPl7 promoter contains
1217. Picado-Leonard, J. and W.L. Miller (1987). Cloning a transcriptional regulatory element coopera-
and sequence of the human gene for P450cl7 tively bound by tale homeodomain proteins.
(steroid 17 a-hydroxylase/17, 20 lyase): Similarity Endocrinol. Res. 24, 489^95.
with the gene for P450c21. DNA 6, 439-448. 1229. Lin, C.J., J.W. Martens, and W.L. Miller (2001).
1218. Nakajin, S., J.E. Shively, PM. Yuan, and PR Hall NP-IC, Spl, and Sp3 are essential for transcrip-
(1981). Microsomal cytochrome P-450 from tion of the human gene for P450cl7 (steroid 17a-
neonatal pig testis: Two enzymatic activities hydroxylase/17, 20 lyase) in human adrenal
(17a-hydroxylase and C17,20-lyase) associated NCI-H295A cells. Mol. Endocrinol. 15,
with one protein. Biochemistry 20, 4037^042. 1277-1293.
1219. Katagiri, M., K. Suhara, M. Shiroo, and 1230. Sewer, M.B., VQ. Nguyen, C.J. Huang,
Y. Rujimura (1982). Role of cytochrome b^ in the PW Tucker, N. Kagawa, and M.R. Waterman
cytochrome P-450-mediated C21-steroid 17,20- (2002). Transcriptional activation of human
lyase reaction. Biochem. Biophys. Res. Commun. CYPl7 in H295R adrenocortical cells depends on
108, 379-384. complex formation among p5'^"'^'^/NonO, protein-
1220. Shinzawa, K., S. Kominami, and S. Takemori associated splicing factor, and SR-1, a complex
(1985). Studies on cytochrome P-450 (P-45017a, that also participates in repression of transcrip-
lyase) from guinea pig adrenal microsomes. tion. Endocrinology 143, 1280-1290.
Dual function of a single enzyme and effect of 1231. Sewer, M.B. and M.R. Waterman (2002).
cytochrome b^. Biochim. Biophys. Acta 833, Adrenocorticotropin/cyclic adenosine 3' ,5' -
151-160. monophosphate-mediated transcription of the
1221. Casey, M.L. and PC. MacDonald (1982). human CYPl7 gene in the adrenal cortex is
Demonstration of steroid 17a-hydroxylase activ- dependent on phosphatase activity. Endocrinology
ity in human fetal kidney, thymus, and spleen. 143, 1769-1777.
Steroids 40, 91-91. 1232. Sewer, M.B. and M.R. Waterman (2002). cAMP-
Mil. Kayes-Wandover, K.M. and PC. White (2000). dependent transcription of steroidogenic genes
Steroidogenic enzyme gene expression in the in the human adrenal cortex requires a dual-
human heart. J. Clin. Endocrinol. Metab. 85, specificity phosphatase in addition to protein
2519-2525. kinase A. J. Mol. Endocrinol. 29, 163-174.
1223. Puche, C , M. Jose, A. Cabero, and A. Meseguer 1233. Sewer, M.B. and M.R. Waterman (2003). cAMP-
(2002). Expression and enzymatic activity of the dependent protein kinase enhances CYPl7
P450cl7 gene in human adipose tissue. Eur transcription via MKP-1 activation in H295R
J. Endocrinol. 146, 223-229. human adrenocortical cells. J. Biol. Chem. 278,
1224. Kusano, K., M. Sakaguchi, N. Kagawa, 8106-8111.
M.R. Waterman, and T Omura (2001). 1234. Yanase, T. (1995). 17a-Hydroxylase/17,20-lyase
Microsomal P450s use specific proline-rich defects. J. Steroid Biochem. Mol. Biol. 53,
sequences for efficient folding, but not for main- 153-157.
tenance of the folded structure. J. Biochem. 1235. Rardella, C.E., D.W Hum, J. Homoki, and
(Tokyo) 129, 259-269. W.L. Miller (1994). Point mutation of Arg440 to
1225. Hales, D.B., L.L. Sha, and A.H. Payne (1987). His in cytochrome P450cl7 causes severe 17a-
Testosterone inhibits cAMP-induced de novo hydroxylase deficiency. J. Clin. Endocrinol.
synthesis of Leydig cell cytochrome P-450(17a) Metab. 79, 160-164.
518 F. Peter Guengerich

1236. Imai, T., H. Globerman, J.M. Gertner, N. Kagawa, 1247. Katagiri, M., N. Kagawa, and M.R. Waterman
and M.R. Waterman (1993). Expression and (1995). The role of cytochrome b^ in the biosyn-
purification of functional human 17a-hydroxy- thesis of androgens by human P450cl7. Arch.
lase/17,20-lyase (P450cl7) in Escherichia coli. Biochem. Biophys. 317, 343-347.
Use of this system for study of a novel form of 1248. Miller, W.L., R.J. Auchus, and D.H. Geller
combined 17a-hydroxylase/17,20-lyase defi- (1997). The regulation of 17, 20 lyase activity.
ciency. J. Biol. Chem. 268, 19681-19689. Steroids 62, 133-142.
1237. Monno, S., Y. Mizushima, N. Toyoda, T. Kashii, 1249. Biason-Lauber, A., B. Kempken, E. Werder,
and M. Kobayashi (1997). A new variant of the M.G. Forest, S. Einaudi, M.B. Ranke et al.
cytochrome P450cl7 (CYP17) gene mutation in (2000). 17a-hydroxylase/17,20-lyase deficiency
three patients with 17a-hydroxylase deficiency. as a model to study enzymatic activity regulation:
Ann. Hum. Genet. 61, 275-279. Role of phosphorylation. J. Clin. Endocrinol.
1238. Kagimoto, K., M.R. Waterman, M. Kagimoto, Metab. H5, 1226-1231.
P. Ferreira, E.R. Simpson, and J.S. Winter (1989). 1250. Soucy, P and V Luu-The (2002). Assessment of
Identification of a common molecular basis the ability of type 2 cytochrome ^5 to modulate
for combined 17a-hydroxylase/17,20-lyase defi- 17,20-lyase activity of human P450cl7. J. Steroid
ciency in two Mennonite families. Hum. Genet. Biochem. Mol. Biol. 80, 71-75.
82, 285-286. 1251. Brock, B.J. and M.R. Waterman (1999).
1239. Yamaguchi, H., M. Nakazato, M. Miyazato, Biochemical differences between rat and human
K. Kangawa, and S. Matsukura (1997). A 5'- cytochrome P450cl7 support the different
splice site mutation in the cytochrome P450 steroidogenic needs of these two species.
steroid 17a-hydroxylase gene in 17a-hydroxy- Biochemistry 3H, 1598-1606.
lase deficiency. J. Clin. Endocrinol. Metab. 82, 1252. Yamazaki, T, T. Ohno, T Sakaki, M. Akiyoshi-
1934-1938. Shibata, Y Yabusaki, T. Imai et al. (1998). Kinetic
1240. Geller, D.H., R.J. Auchus, B.B. Mendonga, and anaylsis of successive reactions catalyzed by
W.L. Miller (1997). The genetic and functional bovine cytochrome P450j7^, . Biochemistry
basis of isolated 17,20-lyase deficiency. Nat. 37, 2800-2806.
Genet. 17,201-205. 1253. Soucy, P and V Luu-The (2000). Conversion of
1241. Miller, W.L., D.H. Geller, and R.J. Auchus pregnenolone to DHEA by human 17a-hydroxy-
(1998). The molecular basis of isolated 17, 20 lase/17,20-lyase (P450c 17). Evidence that DHEA
lyase deficiency. Endocrinol. Res. 24, 817-825. is produced from the released intermediate, 17a-
1242. Auchus, R.J. and M.K. Gupta (2002). Towards hydroxypregnenolone. Eur J. Biochem. 267,
a unifying mechanism for CYP17 mutations 3243-3247.
that cause isolated 17,20-lyase deficiency. 1254. Monno, S., H. Ogawa, T. Date, M. Fujioka, W.L.
Endocrinol. Res. 28, 443-447. Miller, and M. Kobayashi (1993). Mutation of
1243. Kagawa, N. and M.R. Waterman (1995). histidine 373 to leucine in cytochrome P450cl7
Regulation of steroidogenic and related P450s. In causes 17a-hydroxylase deficiency. J. Biol.
PR. Ortiz de Montellano (ed.). Cytochrome Chem. 268,25811-25817.
P450-Structure, Mechanism, and Biochemistry, 1255. Lam, C.W, W Arlt, C.K. Chan, J.W. Honour,
2nd edn. Plenum Press, New York, pp. 419-442. C.J. Lin, S.F Tong et al. (2001). Mutation of
1244. P Lee-Robichaud, A.Z. Shyadehi, J.N. Wright, proline 409 to arginine in the meander region of
M.E. Akhtar, and M. Akhtar (1995). Mechanistic cytochrome p450cl7 causes severe 17a-hydroxy-
kinship between hydroxylation and desaturation lase deficiency Mol. Genet. Metab. 72, 254-259.
reactions: Acyl-carbon bond cleavage pro- 1256. Lee-Robichaud, P, M.E. Akhtar, and M. Akhtar
moted by pig and human CYP17 {?-450^^^, 17a- (1998). An analysis of the role of active site pro-
hydroxylase-17,20-lyase). Biochemistry 34, tic residues of cytochrome P-450s: Mechanistic
14104-14113. and mutational studies on 17a-hydroxylase-
1245. Lieberman, S. and PA. Warne (2001). 17- 17,20-lyase (P-45017a also CYP17). Biochem.
Hydroxylase: An evaluation of the present view J. 330, 967-974.
of its catalytic role in steroidogenesis. J. Steroid 1257. Kitamura, M., E. Buczko, and M.L. Dufau
Biochem. Mol. Biol. 78, 299-312. (1991). Dissociation of hydroxylase and lyase
1246. Soucy, P, L. Lacoste, and V Luu-The (2003). activities by site-directed mutagenesis of the rat
Assessment of porcine and human 16-ene- P450i7^. ^o^- Endocrinol. 5, 1373-1380.
synthase, a third activity of P450cl7, in the 1258. Biason-Lauber, A., E. Leiberman, and
formation of an androstenol precursor. Eur J. M. Zachmann (1997). A single amino acid sub-
Biochem. 210, 1349-1355. stitution in the putative redox partner-binding site
Human P450s 519

of P450cl7 as cause of isolated 17,20-lyase (17a-hydroxylase/17,20-lyase): Insights into


deficiency. J. Clin. Endocrinol Metab. 82, reaction mechanisms and effects of mutations.
3807-3812. Mol. Endocrinol. 13, 1169-1182.
1259. Lee-Robichaud, P., M.E. Akhtar, and M. Akhtar 1270. Ahmed, S. (1999). A novel molecular modelling
(1998). Control of androgen biosynthesis in the study of inhibitors of the 17a-hydroxylase com-
human through the interaction of Arg^^^ and ponent of the enzyme system 17a-hydroxy-
^j.g358 of CYP17 with cytochrome b^ Biochem. J. lase/17,20-lyase (P-450j7^). Bioorg. Med. Chem.
332, 293-296. 7, 1487-1496.
1260. LeeRobichaud, R, M.E. Akhtar, and M. Akhtar 1271. Schappach, A. and H.D. Holtje (2001). Molecular
(1999). Lysine mutagenesis identifies cationic modelling of 17a-hydroxylase-17,20-lyase.
charges of human CYP17 that interact with Pharmazie 56, 435^42.
cytochrome b^ to promote male sex-hormone 1272. Kan, PB., M.A. Hirst, and D. Feldman (1985).
biosynthesis. Biochem. J. 342, 309-312. Inhibition of steroidogenic cytochrome P-450
1261. Auchus, R.J., K. Worthy, D.H. Geller, and enzymes in rat testis by ketoconazole and related
W.L. Miller (2000). Probing structural and func- imidazole anti-fiingal drugs. J. Steroid Biochem.
tional domains of human P450cl7. Endocrinol. 23, 1023-1029.
Res. 26, 695-703. 1273. Kossor, D.C., S. Kominami, S. Takemori, and
1262. Gupta, M.K., D.H. Geller, and R.J. Auchus H.D. Colby (1992). Destruction of testicular
(2001). Pitfalls in characterizing P450cl7 muta- cytochrome P-450 by 7a-thiospironolactone is
tions associated with isolated 17,20-lyase catalyzed by the 17od-hydroxylase. J. Steroid
deficiency. J. Clin. Endocrinol. Metab. 86, Biochem. Mol. Biol. 42, 421^24.
4416-4423. 1274. Potter, G.A., S.E. Barrie, M. Jarman, and
1263. Di Cerbo, A., A. Biason-Lauber, M. Savino, M.G. Rowlands (1995). Novel steroidal inhi-
M.R. Piemontese, A. Di Giorgio, M. Perona et al. bitors of human cytochrome P45017a (17a-
(2002). Combined 17a-hydroxylase/17,20-lyase hydroxylase-Cj7 20"^^^^^)- Potential agents for the
deficiency caused by Phe93Cys mutation in the treatment of prostatic cancer. J. Med. Chem. 38,
CYP17 gene. J. Clin. Endocrinol. Metab. 87, 2463-2471.
898-905. 1275. Li, J.S., Y Li, C. Son, and A.M. Brodie (1996).
1264. Katsumata, N., M. Satoh, A. Mikami, S. Mikami, Synthesis and evaluation of pregnane derivatives
A. Nagashima-Miyokawa, N. Sato et al. (2001). as inhibitors of human testicular 17a-hydroxy-
New compound heterozygous mutation in the lase/Cj7 2o-lyase. J. Med. Chem. 39, 4335-4339.
CYP17 gene in a 46,XY girl with 17a-hydroxy- 1276. Njar, VC. and A.M. Brodie (1999). Inhibitors of
lase/17,20-lyase deficiency. Horm. Res. 55, 17a-hydroxylase/l 7,20-lyase (CYP17): Potential
141-146. agents for the treatment of prostate cancer. Curr.
1265. Brock, B.J. and M.R. Waterman (2000). The use Pharm.Des. 5, 163-180.
of random chimeragenesis to study structure/ 1277. Hartmann, R.W, M. Hector, B.G. Wachall,
function properties of rat and human P450cl7. A. Palusczak, M. Palzer, V Huch et al. (2000).
Arch. Biochem. Biophys. 373, 401^08. Synthesis and evaluation of 17-aliphatic hetero-
1266. Mathieu, A.P., R.J. Auchus, and J.G. LeHoux cycle-substituted steroidal inhibitors of 17a-
(2002). Comparison of the hamster and human hydroxylase/C17-20-lyase (P450 17). J. Med.
adrenal P450cl7 (17a-hydroxylase/17,20-lyase) Chem. 43, 4437-4445.
using site-directed mutagenesis and molecular 1278. Burkhart, J.P, PM. Weintraub, C.A. Gates,
modeling. J. Steroid Biochem. Mol. Biol. 80, R.J. Resvick, R.J. Vaz, D. Friedrich et al (2002).
99-107. Novel steroidal vinyl fluorides as inhibitors of
1267. Lin, D., L.H. Zhang, E. Chiao, and W.L. Miller steroid CI7(20) lyase. Bioorg. Med. Chem. 10,
(1994). Modeling and mutagenesis of the active 929-934.
site of human P450cl7. Mol. Endocrinol. 8, 1279. Auchus, R.J., A. Sampath Kumar, C. Andrew
392^02. Boswell, M.K. Gupta, K. Bruce, N.P Rath et al
1268. Burke, D.F., C.A. Laughton, and S. Neidle (2003). The enantiomer of progesterone {ent-
(1997). Homology modelling of the enzyme P450 progesterone) is a competitive inhibitor of human
17a-hydroxylase/17,20-lyase—a target for pro- cytochromes P450cl7 and P450c21. Arch.
state cancer chemotherapy—from the crystal Biochem. Biophys. 409, 134-144.
structure of P450gj^ 3. Anticancer Drug Des. 12, 1280. Owen, C.P, RJ. NichoUs, H.J. Smith, and
113-123. R. Whomsley (1999). Inhibition of aromatase
1269. Auchus, R.J. and W.L. Miller (1999). (P450^pjj^) by some l-(benzofuran-2-ylmethyl)
Molecular modeling of human P450cl7 imidazoles. J. Pharm. Pharmacol. 51, 427-433.
520 F. Peter Guengerich

1281. Recanatini, M., A. Bisi, A. Cavalli, F. Belluti, A polymorphism in the CYP 17 gene increases the
S. Gobbi, A. Rampa et al. (2001). A new class of risk of breast cancer. Cancer Res. 57, 1063-1065.
nonsteroidal aromatase inhibitors: Design and 1293. Feigelson, H.S., R. McKean-Cowdin,
synthesis of chromone and xanthone derivatives G.A. Coetzee, D.O. Stram, L.N. Kolonel, and
and inhibition of the P450 enzymes aromatase B.E. Henderson (2001). Building a multigenic
and 17a-hydroxylase/C17,20-lyase. J. Med. model of breast cancer susceptibility: CYP 17 and
Chem. 44, 672-680. HSD17B1 are two important candidates. Cancer
1282. Cavalli, A. and M. Recanatini (2002). Looking Res. 61, 785-789.
for selectivity among cytochrome P450 1294. Thompson, PA. and C. Ambrosone (2000).
inhibitors. J. Med. Chem. 45, 251-254. Molecular epidemiology of genetic polymor-
1283. Ehmer, RB., J. Jose, and R.W. Hartmann (2000). phisms in estrogen metabolizing enzymes in
Development of a simple and rapid assay for the human breast cancer. J. Natl. Cancer Inst.
evaluation of inhibitors of human 17cx-hydroxy- Monogr, 125-134.
lase-Cj7 2o"ly^se (P450cl7) by coexpression of 1295. Mitrunen, K., N. Jourenkova, V Kataja,
P450cl7' with NADPH-cytochrome-P450-reduc- M. Eskelinen, V.M. Kosma, S. Benhamou et al.
tase in Escherichia coli. J. Steroid Biochem. Mol. (2000). Steroid metabolism gene CYP17
Biol. 75, 57-63. polymorphism and the development of breast
1284. Grigoryev, D.N., K. Kato, VC. Njar, B.J. Long, cancer. Cancer Epidemiol. Biomarkers Prev. 9,
Y.Z. Ling, X. Wang et al. (1999). Cytochrome 1343-1348.
P450cl7-expressing Escherichia coli as a first- 1296. Ye, Z. and J.M. Parry (2002). The CYP17 M^pAl
step screening system for 17Q:-hydroxylase- polymorphism and breast cancer risk: A meta-
Cj7 2o"^y^^^ inhibitors. Anal. Biochem. 267, analysis. Mutagenesis 17, 119-126.
319-330. 1297. Feigelson, H.S., R. McKean-Cowdin, and
1285. Auchus, R.J. (2001). The genetics, pathophysiol- B.E. Henderson (2002). Concerning the CYP 17
ogy, and management of human deficiencies of MspAl polymorphism and breast cancer risk:
P450cl7. Endocrinol. Metab. Clin. North Am. 30, A meta-analysis. Mutagenesis 17, 445-446.
101-119, vii. 1298. Ambrosone, C.B., K.B. Moysich, H. Furberg,
1286. Kaufman, F.R., G. Costin, U. Goebelsmann, J.L. Freudenheim, E.D. Bowman, S. Ahmed et al.
F.Z. Stanczyk, and M. Zachmann (1983). Male (2003). CYP 17 genetic polymorphism, breast
pseudohermaphroditism due to 17,20-desmolase cancer, and breast cancer risk factors. Breast
deficiency. J. Clin. Endocrinol. Metab. 57, 32-36. Cancer Res. 5, R45-51.
1287. Huang, J., T. Ushiyama, K. Inoue, K. Mori, and 1299. Stanford, J.L., E.A. Noonan, L. Iwasaki, S. Kolb,
S. Hukuda (1999). Possible association of CYP17 R.B. Chadwick, Z. Feng et al. (2002). A poly-
gene polymorphisms with the onset of rheuma- morphism in the CYPl 7 gene and risk of prostate
toid arthritis. Clin. Exp. Rheumatol. 17, 721-724. cancer. Cancer Epidemiol. Biomarkers Prev. 11,
1288. Lai, J., D. Vesprini, W. Chu, H. Jernstrom, and 243-247.
S.A. Narod (2001). CYP gene polymorphisms 1300. Haiman, C.A., M.J. Stampfer, E. Giovannucci,
and early menarche. Mol. Genet. Metab. 74, J. Ma, N.E. Decalo, PW. Kantoff etal. (2001). The
449-457. relationship between a polymorphism in CYP 17
1289. Feigelson, H.S., R. McKean-Cowdin, M.C. Pike, with plasma hormone levels and prostate cancer.
G.A. Coetzee, L.N. Kolonel, A.M. Nomura Cancer Epidemiol. Biomarkers Prev. 10, 743-748.
et al. (1999). Cytochrome P450cl7alpha gene 1301. McKean-Cowdin, R., H.S. Feigelson, M.C. Pike,
(CYP 17) polymorphism predicts use of hormone G.A. Coetzee, L.N. Kolonel, and B.E. Henderson
replacement therapy. Cancer Res 59, 3908-3910. (2001). Risk of endometrial cancer and estrogen
1290. Marszalek, B., M. Lacinski, N. Babych, E. Capla, replacement therapy history by CYP 17 genotype.
J. Biemacka-Lukanty, A. Warenik-Szymankiewicz Cancer Res. 61, 848-849.
et al. (2001). Investigations on the genetic poly- 1302. de Carmo Silva, R., C.E. Kater, S.A. Dib,
morphism in the region of CYP 17 gene encoding S. Laureti, F. Forini, A. Cosentino et al. (2000).
5'-UTR in patients with polycystic ovarian syn- Autoantibodies against recombinant human
drome. Gynecol. Endocrinol. 15, 123-128. steroidogenic enzymes 21-hydroxylase, side-
1291. Kristensen, VN., E.H. Kure, B. Erikstein, chain cleavage and 17a-hydroxylase in Addison's
N. Harada, and A. Borresen-Dale (2001). Genetic disease and autoimmune polyendocrine syn-
susceptibility and environmental estrogen-like drome type 111. Eur J. Endocrinol. 142, 187-194.
compounds. Mutat. Res. 482, 77-82. 1303. Simpson, E.R., C. Clyne, G. Rubin, W.C. Boon,
1292. Feigelson, H.S., G.A. Coetzee, L.N. Kolonel, K. Robertson, K. Britt et al. (2002). Aromatase—
R.K. Ross, and B.E. Henderson (1997). a brief overview. Annu. Rev. Physiol. 64, 93-127.
Human P450s 521

1304. Roselli, C.E. and J.A. Resko (2001). Cytochrome 1315. Takayanagi, R., K. Goto, S. Suzuki, S. Tanaka,
P450 aromatase (CYP19) in the non-human pri- S. Shimoda, and H. Nawata (2002). Dehy-
mate brain: Distribution, regulation, and func- droepiandrosterone (DHEA) as a possible source
tional significance. J. Steroid Biochem. Mol. Biol. for estrogen formation in bone cells: Correlation
79, 247-253. between bone mineral density and serum
1305. Harada, N. (1992). A unique aromatase DHEA-sulfate concentration in postmenopausal
(P-450^j^Qj^) mRNA formed by alternative use women, and the presence of aromatase to
of tissue-specific exons 1 in human skin fibrob- be enhanced by 1,25-dihydroxyvitamin D3 in
lasts. Biochem. Biophys. Res. Commun. 189, human osteoblasts. Mech. Ageing Dev. 123,
1001-1007. 1107-1114.
1306. Hinshelwood, M.M. and C.R. Mendelson (2001). 1316. Zhang, R, D. Zhou, Y.C Kao, J. Ye, and S. Chen
Tissue-specific expression of the human CYP19 (2002). Expression and purification of a recombi-
(aromatase) gene in ovary and adipose tissue of nant form of human aromatase from Escherichia
transgenic mice. J. Steroid Biochem. Mol. Biol. coli. Biochem. Pharmacol. 64, 1317-1324.
79,193-201. 1317. Kagawa, N., Q. Cao, and K. Kusano (2003).
1307. Shozu, M., H. Sumitani, T. Segawa, H.J. Yang, Expression of human aromatase (CYP19) in
K. Murakami, T. Kasai et al. (2002). Over- Escherichia coli by N-terminal replacement and
expression of aromatase P450 in leiomyoma induction of cold stress response. Steroids 68,
tissue is driven primarily through promoter 1.4 205-209.
of the aromatase P450 gene (CYP19). J. Clin. 1318. Graham-Lorence, S., M.W. Khalil, M.C Lorence,
Endocrinol. Metab. 87, 2540-2548. C.R. Mendelson, and E.R. Simpson (1991).
1308. Rubin, G.L., J.H. Duong, CD. Clyne, C.J. Speed, Structure-function relationships of human aro-
Y. Murata, C. Gong et al. (2002). Ligands for the matase cytochrome P-450 using molecular mod-
peroxisomal proliferator-activated receptor eling and site-directed mutagenesis. J. Biol.
gamma and the retinoid X receptor inhibit aro- Chem. 266, 11939-11946.
matase cytochrome P450 (CYPJ9) expression 1319. Conley, A., S. Mapes, CJ. Corbin, D. Greger, and
mediated by promoter II in human breast adipose. S. Graham (2002). Structural determinants of
Endocrinology 143, 2863-2871. aromatase cytochrome P450 inhibition in sub-
1309. Clyne, CD., C.J. Speed, J. Zhou, and E.R. strate recognition site-1. Mol. Endocrinol. 16,
Simpson (2002). Liver receptor homologue-1 1456-1468.
(LRH-1) regulates expression of aromatase in 1320. Marcotte, PA. and CH. Robinson (1982). Design
preadipocytes. J. Biol. Chem. 277, 20591-20597. of mechanism-based inactivators of human pla-
1310. Kamat, A. and CR. Mendelson (2001). cental aromatase. Cancer Res. 42, 3322s-3326s.
Identification of the regulatory regions of the 1321. Brueggemeier, R.W. (2002). Aromatase inhibitors
human aromatase P450 {CYP19) gene involved in in breast cancer therapy. Expert Rev. Anticancer
placenta-specific expression. J. Steroid Biochem. Then 2, 181-191.
Mol. Biol. 79, 173-180. 1322. Lombardi, P. (2002). Exemestane, a new steroidal
1311. Haiman, CA., S.E. Hankinson, I. De Vivo, aromatase inhibitor of clinical relevance.
C Guillemette, N. Ishibe, D.J. Hunter et al. Biochim. Biophys. Acta 1587, 326-337.
(2003). Polymorphisms in steroid hormone path- 1323. Lonning, PE. (2002). The role of aromatase
way genes and mammographic density. Breast inactivators in the treatment of breast cancer. Int.
Cancer Res. Treat. 11, 27-36. J. Clin. Oncol 7, 265-270.
1312. Cole, PA. and CH. Robinson (1988). A peroxide 1324. Lonning, PE. (2002). Aromatase inhibitors and
model reaction for placental aromatase. J. Am. inactivators for breast cancer treatment. Eur
Chem. Soc. 110, 1284-1285. J. Cancer 38(Suppl 6), S47-S48.
1313. Akhtar, M., D. Corina, S. Miller, A.Z. Shyadehi, 1325. Jones, S.A. and S.E. Jones (2000). Exemestane:
and J.N. Wright (1994). Mechanism of the A novel aromatase inactivator for breast cancer.
acyl-carbon cleavage and related reactions Clin. Breast Cancer 1, 211-216.
catalyzed by multifunctional P-450s: Studies 1326. Winnett, G., D. van Hagen, and M. Schrey
on cytochrome P45017a. Biochemistry 33, (2003). Prostaglandin ]^ metabolites inhibit aro-
4410-4418. matase activity by redox-sensitive mechanisms:
1314. Roberts, E.S., A.D.N. Vaz, and M.J. Coon (1991). Potential implications for breast cancer therapy.
Catalysis by cytochrome P-450 of an oxidative Int. J. Cancer 103, 600-605.
reaction in xenobiotic aldehyde metabolism: 1327. Pouget, C , C Fagnere, J.P Basly, G. Habrioux,
Deformylation with olefin formation. Proc. Natl. and A.J. Chulia (2002). Design, synthesis and
Acad Set USA 88, 8963-8966. evaluation of 4-imidazolylflavans as new leads
522 F. Peter Guengerich

for aromatase inhibition. Bioorg. Med. Chem. 1339. Yu, L., D.G. Romero, C.E. Gomez-Sanchez, and
Lett. 12,2859-2861. E.R Gomez-Sanchez (2002). Steroidogenic
1328. Smith, M.R., D. Kaufman, D. George, W.K. Oh, enzyme gene expression in the human brain.
M. Kazanis, J. Manola et al. (2002). Selective Mol. Cell Endocrinol. 190, 9-17.
aromatase inhibition for patients with androgen- 1340. Zanger, U.M., N. Kagawa, J. Lund, and
independent prostate carcinoma. Cancer 95, M.R. Waterman (1992). Distinct biochemical
1864-1868. mechanisms for cAMP-dependent transcription
1329. Murata, Y., K.M. Robertson, M.E. Jones, and of CYP17 and CYP21. FASEB J. 6, 719-723.
E.R. Simpson (2002). Effect of estrogen defi- 1341. Watanabe, N., M. Kitazume, J. Fujisawa,
ciency in the male: The ArKO mouse model. Mol. M. Yoshida, and Y. Fujii-Kuriyama (1993). A
Cell Endocrinol. 193, 7-12. novel cAMP-dependent regulatory region includ-
1330. Herrmann, B.L., B. Sailer, O.E. Janssen, ing a sequence like the cAMP-responsive ele-
P. Gocke, A. Bockisch, H. Sperling et al. (2002). ment, far upstream of the human CYP21A2 gene.
Impact of estrogen replacement therapy in a male Eur J. Biochem. 214, 521-531.
with congenital aromatase deficiency caused by a 1342. Chang, S.E and B.C. Chung (1995). Difference in
novel mutation in the CYP19 gene. J. Clin. transcriptional activity of two homologous
Endocrinol. Metab. 87, 5476-5484. CYP21A genes. Mol. Endocrinol. 9, 1330-1336.
1331. Meinhardt, U. and RE. Mulhs (2002). The aro- 1343. Bird, I.M., J.I. Mason, and WE. Rainey (1998).
matase cytochrome P-450 and its clinical impact. Protein kinase A, protein kinase C, and Ca-^^-
Horm.Res. 57, 145-152. regulated expression of 21-hydroxylase cyto-
1332. Britt, K.L., A.E. Drummond, M. Dyson, chrome P450 in H295R human adrenocortical
N.G. Wreford, M.E. Jones, E.R. Simpson et al. cells. J. Clin. Endocrinol. Metab. 83, 1592-1597.
(2001). The ovarian phenotype of the aromatase 1344. Wijesuriya, S.D., G. Zhang, A. Dardis, and
knockout (ArKO) mouse. J. Steroid Biochem. WL. Miller (1999). Transcriptional regulatory
Mol. Biol. 79, 181-185. elements of the human gene for cytochrome
1333. Bakker, J., S. Honda, N. Harada, and J. Balthazart P450c21 (steroid 21-hydroxylase) lie within
(2002). The aromatase knock-out mouse provides intron 35 of the linked C4B gene. J. Biol. Chem.
new evidence that estradiol is required during 274,38097-38106.
development in the female for the expression of 1345. White, P C , M.T. Tusie-Luna, M.I. New, and
sociosexual behaviors in adulthood. J. Neurosci. PW. Speiser (1994). Mutations in steroid 21-
22,9104-9112. hydroxylase (CYP21). Hum. Mutat. 3, 373-378.
1334. Lonning, RE., L.E. Kragh, B. Erikstein, 1346. M.T. Tusie-Luna, PW. Speiser, M. Dumic,
A. Hagen, T Risberg, E. Schlichting et al. M.l. New, and PC. White (1991). A mutation
(2001). The potential for aromatase inhibition in (Pro-30 to Leu) in CYP21 represents a potential
breast cancer prevention. Clin. Cancer Res. 7, nonclassic steroid 21-hydroxylase deficiency
4423s-4428s; discussion 441 ls^412s. allele. Mol. Endocrinol. 5, 685-692.
1335. Suspitsin, E.N., M.Y. Grigoriev, A.V Togo, 1347. Amor, M., K.L. Parker, H. Globerman, M.I. New,
E.S. Kuligina, E.V. Belogubova, K.M. Pozharisski and PC. White (1988). Mutation in the CYP21B
et al. (2002). Distinct prevalence of the CYP19 gene (He-172—->Asn) causes steroid 21-
A3(TTTA)7 allele in premenopausal versus post- hydroxylase deficiency. Proc. Natl. Acad. Sci.
menopausal breast cancer patients, but not in con- USAH5, 1600-1604.
trol individuals. Eur J. Cancer 38, 1911-1916. 1348. Owerbach, D., L. Sherman, A.L. Ballard, and
1336. Kado, N., J. Kitawaki, H. Obayashi, H. Ishihara, R. Azziz (1992). Pro-453 to Ser mutation in CYP21
H. Koshiba, I. Kusuki et al. (2002). Association is associated with nonclassic steroid 21-hydroxy-
of the CYP17 gene and CYP19 gene polymor- lase deficiency. Mol. Endocrinol. 6, 1211-1215.
phisms with risk of endometriosis in Japanese 1349. Bobba, A., E. Marra, P Lattanzio, A. lolascon, and
women. Hum. Reprod. 17, 897-902. S. Giannattasio (2000). Characterization of the
1337. Bryan, G.T., A.M. Lewis, J.B. Harkins, CYP21 gene 5' flanking region in patients affected
S.F. Micheletti, and G.S. Boyd (1974). by 21-OH deficiency Hum. Mutat. 15, 481.
Cytochrome P450 and steroid 21-hydroxylation 1350. White, PC. and PW. Speiser (2000). Congenital
in microsomes from beef adrenal cortex. Steroids adrenal hyperplasia due to 21-hydroxylase defi-
23, 185-201. ciency. Endocrinol. Rev. 21, 245-291.
1338. Zhou, Z., VR. Agarwal, N. Dixit, P White, and 1351. Lee, H. (2001). CYP21 mutations and congenital
PW. Speiser (1997). Steroid 21-hydroxylase adrenal hyperplasia. Clin. Genet. 59, 293-301.
expression and activity in human lymphocytes. 1352. Krone, N., A. Braun, A.A. Roscher, D. Knorr, and
Mol. Cell Endocrinol. Ill, 11-18. H.P Schwarz (2000). Predicting phenotype in
Human P450s 523

steroid 21-hydroxylase deficiency? Comprehensive a vitamin D3 hydroxylase-associated protein.


genotyping in 155 unrelated, well defined patients J. Biol. Chem. 269, 176-182.
from southern Germany. J. Clin. Endocrinol. 1364. Chen, K.S., J.M. Prahl, and H.F. DeLuca (1993).
Metab. 85, 1059-1065. Isolation and expression of human 1,25-dihy-
1353. Levo, A. and J. Partanen (2001). Novel mutations droxyvitamin D^ 24-hydroxylase cDNA. Proc.
in the human CYP21 gene. Prenat. Diagn. 21, Natl. Acad Sci. USA 90, 4543-4547.
885-889. 1365. Yang, W., PA. Friedman, R. Kumar, J.L. Omdahl,
1354. Koyama, S., T. Toyoura, S. Saisho, B.K. May, M.L. SiuCaldera et al. (1999).
K. Shimozawa, and J. Yata (2002). Genetic analy- Expression of 25(OH)D3 24-hydroxylase in distal
sis of Japanese patients with 21-hydroxylase nephron: Coordinate regulation by 1, 25(OH)2D3
deficiency: Identification of a patient with a and cAMP or PTH. Am. J. Physiol. 276,
new mutation of a homozygous deletion of E793-E805.
adenine at codon 246 and patients without 1366. Jones, G., H. Ramshaw, A. Zhang, R. Cook,
demonstrable mutations within the structural V Byford, J. White et al. (1999). Expression and
gene for CYP21. J. Clin. Endocrinol. Metab. 87, activity of vitamin D-metabolizing cytochrome
2668-2673. P450s (CYPla and CYP24) in human nonsmall
1355. Lee, H.H., D.M. Niu, R.W. Lin, P Chan, and cell lung carcinomas. Endocrinology 140,
C.Y. Lin (2002). Structural analysis of the chimeric 3303-3310.
CYP21P/CYP21 gene in steroid 21-hydroxylase 1367. Chen, M.L., G. Heinrich, Y.I. Ohyama, K. Okuda,
deficiency. J. Hum. Genet. 47, 517-522. J.L. Omdahl, T.C. Chen et al. (1994). Expression
1356. Koppens, PR, T. Hoogenboezem, and of 25-hydroxyvitamin D3-24-hydroxylase mRNA
H.J. Degenhart (2002). Duplication of the in cultured human keratinocytes. Proc. Soc. Exp.
CYP21A2 gene complicates mutation analysis of Biol. Med. 207,57-61.
steroid 21-hydroxylase deficiency: Characteristics 1368. Schuster, I., H. Egger, N. Astecker, G. Herzig,
of three unusual haplotypes. Hum. Genet. I l l , M. Schussler, and G. Vorisek (2001). Selective
405-410. inhibitors of CYP24: Mechanistic tools to
1357. Dain, L.B., N.D. Buzzalino, A. Oneto, S. Belh, explore vitamin D metabolism in human ker-
M. Stivel, T. Pasqualini et al. (2002). Classical atinocytes. Steroids 66, 451-462.
and nonclassical 21-hydroxylase deficiency: 1369. Bareis, P, E. Kallay, M.G. Bischof, G. Bises,
A molecular study of Argentine patients. Clin. H. Hofer, C. Potzi et al. (2002). Clonal differ-
Endocrinol. (Oxford) 56, 239-245. ences in expression of 25-hydroxyvitamin D3-
1358. Mornet, E. and J.F. Gibrat (2000). A 3D model of 1 a-hydroxylase, of 25-hydroxyvitamin D3-24-
human P450c21: Study of the putative effects of hydroxylase, and of the vitamin D receptor in
steroid 21-hydroxylase gene mutations. Hum. human colon carcinoma cells: Effects of epider-
Genet. 106, 330-339. mal growth factor and la,25-dihydroxyvitamin
1359. Menard, R.H., EC. Bartter, and J.R. Gillette D3. Exp. Cell Res. 276, 320-327.
(1976). Spironolactone and cytochrome P-450: 1370. Farhan, H. and H.S. Cross (2002). Transcriptional
Impairment of steroid 21-hydroxylation in the inhibition of CYP24 by genistein. Ann. N. Y.
adrenal cortex. Arch. Biochem. Biophys. 173, Acad Sci. 973, 459-462.
395-402. 1371. Zierold, C , H.M. Darwish, and H.F DeLuca
1360. Tajima, T., T. Okada, X.M. Ma, W. Ramsey, (1994). Identification of a vitamin D-response
S. Bomstein, and G. Aguilera (1999). Restoration element in the rat calcidiol (25-hydroxyvitamin
of adrenal steroidogenesis by adenovirus-mediated D3) 24-hydroxylase gene. Proc. Natl. Acad. Sci.
transfer of human cytochrome P450 21-hydroxy- USA 91, 900-902.
lase into the adrenal gland of 21-hydroxylase-defi- 1372. Ohyama, Y, K. Ozono, M. Uchida, T. Shinki,
cientmice. GeneTher 6, 1898-1903. S. Kato, T. Suda et al. (1994). Identification
1361. Pedersen, J.I., H.H. Shobaki, I. Holmberg, of a vitamin D-responsive element in the 5'-
S. Bergseth, and I. Bjorkhem (1983). 25- flanking region of the rat 25-hydroxyvitamin
Hydroxyvitamin D3-24-hydroxylase in rat kidney D3 24-hydroxylase gene. J. Biol. Chem. 269,
mitochondria. J. Biol. Chem. 258, 742-746. 10545-10550.
1362. Ohyama, Y, S. Hayashi, and K. Okuda (1989). 1373. Kerry, D.M., PP Dwivedi, C.N. Hahn,
Purification of 25-hydroxyvitamin D3 24-hydrox- H.A. Morris, J.L. Omdahl, and B.K. May (1996).
ylase from rat kidney mitochondria. FEBS Lett. Transcriptional synergism between vitamin D-
255, 405^08. responsive elements in the rat 25-hydroxyvitamin
1363. Ettinger, R.A., R. Ismail, and H.F. DeLuca D3 24-hydroxylase (CYP24) promoter. J. Biol.
(1994). cDNA cloning and characterization of Chem. 271,29715-29721.
524 F. Peter Guengerich

1374. Dwivedi, P.P., J.L. Omdahl, I. Kola, D.K. Hume, 1385. Dilworth, KJ., I. Scott, A. Green, S. Strugnell,
and B.K. May (2000). Regulation of rat cyto- YD. Guo, E.A. Roberts et al (1995). Different
chrome P450C24 (CYP24) gene expression. Evi- mechanisms of hydroxylation site selection by
dence for functional cooperation of Ras-activated liver and kidney cytochrome P450 species
Ets transcription factors with the vitamin D (CYP27 and CYP24) involved in vitamin D
receptor in 1,25- dihydroxyvitamin D3-mediated metaboUsm. ^ 5zo/. Chem. 270, 16766-16774.
induction. J. Biol. Chem. 275, 47-55. 1386. Schuster, I., H. Egger, P. Nussbaumer, and
1375. Raval-Pandya, M., P. Dhawan, F. Barletta, and R.T. Kroemer (2003). Inhibitors of vitamin D
S. Christakos (2001). YYl represses vitamin D hydroxylases: Structure-activity relationships.
receptor-mediated 25-hydroxyvitamin D3 24- J. Cell. Biochem. 88, 372-380.
hydroxylase transcription: Relief of repression by 1387. Schuster, I., H. Egger, D. Bikle, G. Herzig,
CREB-binding protein. Mol Endocrinol. 15, G.S. Reddy, A. Stuetz et al. (2001). Selective
1035-1046. inhibition of vitamin D hydroxylases in human
1376. Dwivedi, PR, C.S. Hii, A. Ferrante, J. Tan, keratinocytes. Steroids 66, 409-422.
C.J. Der, J.L. Omdahl et al (2002). Role of MAP 1388. Henry, H.L. (2001). The 25(OH)D3/la,25(OH)2D3-
kinases in the 1,25-dihydroxyvitamin D^-induced 24R-hydroxylase: A catabolic or biosynthetic
transactivation of the rat cytochrome P450C24 enzyme? Steroids 66, 391-398.
(CYP24) promoter. Specific functions for 1389. Kasuga, H., N. Hosogane, K. Matsuoka,
ERK1/ERK2 and ERK5. J. Biol. Chem. Ill, I. Mori, Y Sakura, K. Shimakawa et al. (2002).
29643-29653. Characterization of transgenic rats constitutively
1377. Jones, G. and H.S. Tenenhouse (2002). expressing vitamin D-24-hydroxylase gene.
l,25(OH)2D, the preferred substrate for CYP24. Biochem. Biophys. Res. Commun. 297,1332-1338.
J. Bone Miner. Res. 17, 179-181. 1390. Martini, R. and M. Murray (1993). Participation
1378. Sakaki, T., N. Sawada, K. Komai, S. Shiozawa, of P450 3A enzymes in rat hepatic microsomal
S. Yamada, K. Yamamoto et al. (2000). Dual retinoic acid 4-hydroxylation. Arch. Biochem.
metabolic pathway of 25-hydroxyvitamin D3 cat- Biophys. 303, 57-66.
alyzed by human CYP24. Eur J. Biochem. 267, 1391. White, J.A., B. Beckett-Jones, YD. Guo,
6158-6165. F.J. Dilworth, J. Bonasoro, G. Jones et al.
1379. Beckman, M.J., P Tadikonda, E. Werner, J. Prahl, (1997). cDNA cloning of human retinoic acid-
S. Yamada, and H.F DeLuca (1996). Human 25- metabolizing enzyme (hP450RAI) identifies
hydroxyvitamin D3-24-hydroxylase, a muhicat- a novel family of cytochromes P450 (CYP26).
alytic enzyme. Biochemistry 35, 8465-8472. J. Biol. Chem. 272, 18539-18541.
1380. Miyamoto, Y, T. Shinki, K. Yamamoto, 1392. White, J.A., H. Ramshaw, M. Taimi, W. Stangle,
Y Ohyama, H. Iwasaki, R. Hosotani et al. (1997). A. Zhang, S. Everingham et al. (2000).
1 a,25-dihydroxyvitamin D3-24-hydroxylase Identification of the human cytochrome P450,
(CYP24) hydroxylates the carbon at the end of P450RA1-2, which is predominantly expressed in
the side chain (C-26) of the C-24-fluorinated ana- the adult cerebellum and is responsible for all-
log of la,25-dihydroxyvitamin D3. J. Biol. Chem. trans-XQimo\c acid metabolism. Proc. Natl. Acad.
212, 14115-14119. Sci. USA 97, 6403-6408.
1381. Hayashi, K., M. Akiyoshi-Shibata, T. Sakaki, and 1393. Trofimova-Griffin, M.E. and M.R. Juchau (1998).
Y Tabusaki (1998). Rat CYP24 catalyses 235"- Expression of cytochrome P450RAI (CYP26) in
hydroxylation of 26,26,26,27,27,27-hexafluoro- human fetal hepatic and cephalic tissues.
calcitriol in vitro. Xenobiotica 28, 457^63. Biochem. Biophys. Res. Commun. 252,487-491.
1382. Sakaki, T, N. Sawada, Y Nonaka, Y Ohyama, 1394. Nelson, D.R. (1999). A second CYP26 P450 in
and K. Inouye (1999). Metabolic studies using humans and zebrafish: CYP26B1. Arch. Biochem.
recombinant Escherichia coli cells producing rat Biophys. 371, 345-347.
mitochondrial CYP24. CYP24 can convert 1395. Abu-Abed, S., G. MacLean, V Fraulob,
la,25-dihydroxyvitamin D3 to calcitroic acid. P Chambon, M. Petkovich, and P Dolle (2002).
Eur J. Biochem. 262, 4 3 ^ 8 . Differential expression of the retinoic acid-
1383. Inouye, K. and T. Sakaki (2001). Enzymatic metabolizing enzymes CYP26A1 and CYP26B1
studies on the key enzymes of vitamin D metabo- during murine organogenesis. Mech. Dev. 110,
lism; la-hydroxylase (CYP27B1) and 24-hydrox- 173-177.
ylase (CYP24). Biotechnol. Annu. Rev. 7, 179-194. 1396. Trofimova-Griffin, M.E. and M.R. Juchau
1384. Omdahl, J.L., E.V Bobrovnikova, A. Annalora, (2002). Developmental expression of cytochrome
P Chen, and R. Serda (2003). Expression, CYP26B1 (P450RAI-2) in human cephalic
structure-function, and molecular modeling of tissues. Brain. Res. Develop. Brain Res. 136,
vitamin D P450s. J. Cell. Biochem. 88, 356-362. 175-178.
Human P450s 525

1397. Matsuzaki, Y., B. Bouscarel, T. Ikegami, 1409. Su, P, H. Rennert, R.M. Shayiq, R. Yamamoto,
A. Honda, M. Doy, S. Ceryak et al (2002). Y Zheng, S. Addya et al. (1990). A cDNA encod-
Selective inhibition of CYP27A1 and of chen- ing a rat mitochondrial cytochrome P450 catalyz-
odeoxycholic acid synthesis in cholestatic ham- ing both the 26-hydroxylation of cholesterol and
ster liver. Biochim. Biophys. Acta 1588, 139-148. 25-hydroxylation of vitamin D3: Gonadotropic
1398. Postlind, H., E. Axen, T. Bergman, and K. Wikvall regulation of the cognate mRNA in ovaries. DNA
(1997). Cloning, structure, and expression of a Cell Biol. 9,657-665.
cDNA encoding vitamin D3 25-hydroxylase. 1410. Wikvall, K. (2001). Cytochrome P450 enzymes
Biochem. Biophys. Res. Commun. 241, 491^97. in the bioactivation of vitamin D to its hormonal
1399. Hayashi, S., M. Noshiro, and K. Okuda (1984). form (review). Int. J. Mol. Med. 7, 201-209.
Purification of cytochrome P-450 catalyzing 25- 1411. Pikuleva, I.A., I. Bjorkhelm, and M.R. Waterman
hydroxylation of vitamin D3 from rat liver micro- (1997). Expression, purification, and enzymatic
somes. Biochem. Biophys. Res. Commun. 121, properties of recombinant human cytochrome
994-1000. P450c27 (CYP27). Arch. Biochem. Biophys. 343,
1400. Saarem, K. and J.I. Pedersen (1985). 25- 123-130.
Hydroxylation of la-hydroxyvitamin D3 in rat and 1412. Pikuleva, I.A., A. Babiker, M.R. Waterman, and
human liver. Biochim. Biophys. Acta 840, 117-126. I. Bjorkhem (1998). Activities of recombinant
1401. Akiyoshi-Shibata, M., E. Usui, T. Sakaki, human cytochrome P450c27 (CYP27) which pro-
Y. Yabusaki, M. Noshiro, K. Okuda et al. (1991). duce intermediates of alternative bile acid biosyn-
Expression of rat liver vitamin D3 2 5-hydroxylase thetic pathways, a 5/0/. Chem. 273, 18153-18160.
cDNA in Saccharomyces cerevisiae. FEBS Lett. 1413. Sawada, N., T. Sakaki, M. Ohta, and K. Inouye
280, 367-370. (2000). Metabolism of vitamin D3 by human
1402. Guo, YD., S. Strugnell, D.W. Back, and G. Jones CYP27A1. Biochem. Biophys. Res. Commun.
(1993). Transfected human liver cytochrome 213, 977-984.
P-450 hydroxylates vitamin D analogs at different 1414. Sawada, N., T. Sakaki, S. Kitanaka, S. Kato, and
side-chain positions. Proc. Natl. Acad. Sci. USA K. Inouye (2001). Structure-function analysis of
90, 8668-8672. CYP27B1 and CYP27A1—Studies on mutants
1403. Shiga, K., R. Fukuyama, S. Kimura, K. Nakajima, from patients with vitamin D-dependent rickets
and S. Fushiki (1999). Mutation of the sterol 27- type I (VDDR-I) and cerebrotendinous xan-
hydroxylase gene (CYP27) results in truncation thomatosis (CTX). Eur J. Biochem. 268,
of mRNA expressed in leucocytes in a Japanese 6607-6615.
family with cerebrotendinous xanthomatosis. 1415. Pikuleva, I.A., A. Puchkaev, and I. Bjorkhem
J. Neurol. Neurosurg. Psychiatr. 67, 675-677. (2001). Putative helix F contributes to regio-
1404. Garuti, R., M.A. Croce, R. Tiozzo, M.T. Dotti, selectivity of hydroxylation in mitochon-
A. Federico, S. Bertolini et al. (1997). Four novel drial cytochrome P450 27A1. Biochemistry 40,
mutations of sterol 27-hydroxylase gene in Italian 7621-7629.
patients with cerebrotendinous xanthomatosis. 1416. Murtazina, D., A.V Puchkaev, C.H. Schein,
J. Lipid Res. 38, 2322-2334. N. Oezguen, W Braun, A. Nanavati et al. (2002).
1405. Gascon-Barre, M., C. Demers, O. Ghrab, Membrane-protein interactions contribute to effi-
C. Theodoropoulos, R. Lapointe, G. Jones et al. cient 27-hydroxylation of cholesterol by mito-
(2001). Expression of CYP27A, a gene encoding chondrial cytochrome P450 llAl.J. Biol. Chem.
a vitamin D-25 hydroxylase in human liver and Ill, 37582-37589.
kidney. Clin. Endocrinol. (Oxford) 54, 107-115. 1417. Hosseinpour, F, M. Hidestrand, M. Ingelman-
1406. Shanahan, CM., K.L. Carpenter, and N.R. Gary Sundberg, and K. Wikvall (2001). The
(2001). A potential role for sterol 27-hydroxylase importance of residues in substrate recognition
in atherogenesis. Atherosclerosis 154, 269-276. site 3 for the catalytic function of CYP2D25
1407. Lee, M.J., YC. Huang, M.G. Sweeney, (vitamin D 25-hydroxylase). Biochem. Biophys.
N.W. Wood, M.M. Reilly, and RK. Yip (2002). Res. Commun. 288, 1059-1063.
Mutation of the sterol 27-hydroxylase gene 1418. Wills, M.R. and J. Savory (1984). Vitamin D
(CYP27A1) in a Taiwanese family with cerebro- metabolism and chronic liver disease. Ann. Clin.
tendinous xanthomatosis. J. Neurol. 249, Lab. Sci. 14, 189-197.
1311-1312. 1419. Sugama, S., A. Kimura, W Chen, S. Kubota,
1408. Chen, W., S. Kubota, H. Ujike, T. Ishihara, and Y Seyama, N. Taira et al. (2001). Frontal lobe
Y Seyama (1998). A novel Arg362Ser mutation dementia with abnormal cholesterol metabolism
in the sterol 27-hydroxylase gene (CYP27): and heterozygous mutation in sterol 27-hydroxy-
Its effects on pre-mRNA splicing and enzyme lase gene (CYP27). J. Inherit. Metab. Dis. lA,
activity. Biochemistry 37, 15050-15056. 379-392.
526 F. Peter Guengerich

1420. Bjorkhem, I. (2002). Do oxysterols control cho- 25-hydroxyvitamin D3 l a -hydroxylase. Biochem.


lesterol homeostasis?. J. Clin. Invest. 110, Biophys. Res. Commun. 239, 527-533.
725-730. 1432. Fu, G.K., A.A. Portale, and WL. Miller (1997).
1421. Rosen, H., A. Reshef, N. Maeda, A. Lippoldt, Complete structure of the human gene for the
S. Shpizen, L. Triger et ah (1998). Markedly vitamin D la-hydroxylase, P450cla. DNA Cell
reduced bile acid synthesis but maintained levels Biol. 16, 1499-1507.
of cholesterol and vitamin D metabolites in mice 1433. Zehnder, D., R. Bland, E.A. Walker,
with disrupted sterol 27-hydroxylase gene. A.R. Bradwell, A.J. Howie, M. Hewison et al.
J. Biol. Chem. 273, 14805-14812. (1999). Expression of 25-hydroxyvitamin D3-la-
1422. Reiss, A.B., N.W. Awadallah, S. Malhotra, hydroxylase in the human kidney. J. Am. Soc.
M.C. Montesinos, E.S. Chan, N.B. Javitt et al. Nephrol. 10, 2465-2473.
(2001). Immune complexes and IFN-7 decrease 1434. Zehnder, D., R. Bland, M.C. Williams,
cholesterol 27-hydroxylase in human arterial R.W McNinch, A.J. Howie, RM. Stewart et al.
endothelium and macrophages. J. Lipid Res. 42, (2001). Extrarenal expression of 25-hydroxyvita-
1913-1922. min D3-la-hydroxylase. J. Clin. Endocrinol.
1423. Goodwin, B., K.C. Gauthier, M. Umetani, Metab. 86, 888-894.
M.A. Watson, M.I. Lochansky, J.L. Collins et al. 1435. Zehnder, D., K.N. Evans, M.D. Kilby,
(2003). Identification of bile acid precursors as J.N. Bulmer, B.A. Innes, P.M. Stewart et al.
endogenous ligands for the nuclear xenobiotic (2002). The ontogeny of 25-hydroxyvitamin D3
pregnane X receptor. Proc. Natl. Acad. Sci. USA la-hydroxylase expression in human placenta
100, 223-228. and decidua. Am. J. Pathol. 161, 105-114.
1424. Escher, G., Z. Krozowski, K.D. Croft, and 1436. Diaz, L., C. Arranz, E. Avila, A. HalhaH,
D. Sviridov (2003). Expression of sterol 27- F Vilchis, and F Larrea (2002). Expression and
hydroxylase (CYP27A1) enhances cholesterol activity of 25-hydroxyvitamin D-la-hydroxylase
efflux. J. Biol. Chem. 278, 11015-11019. are restricted in cultures of human syncytiotro-
1425. Fu, X., J.G. Menke, Y. Chen, G. Zhou, phoblast cells from preeclamptic pregnancies.
K.L. MacNaul, S.D. Wright et al. (2001). 27- J. Clin. Endocrinol. Metab. 87, 3876-3882.
Hydroxycholesterol is an endogenous ligand for 1437. Friedrich, M., C. Villena-Heinsen, R. Axt-
liver X receptor in cholesterol-loaded cells. Fliedner, R. Meyberg, W Tilgen, W Schmidt
J. Biol. Chem. 276, 38378-38387. et al. (2002). Analysis of 25-hydroxyvitamin
1426. H.F. DeLuca (1977). Vitamin D as a prohormone. D3-1 a-hydroxylase in cervical tissue. Anticancer
Biochem. Pharmacol. 26, 563-566. Res. 21, 183-186.
1427. Paulson, S.K. and H.F DeLuca (1985). 1438. Segersten, U, P. Correa, M. Hewison, P. Hellman,
Subcellular location and properties of rat renal H. Dralle, T. Carling et al. (2002). 25-hydroxyvi-
25-hydroxyvitamin D3-la-hydroxylase. J. Biol. tamin D3-1 a-hydroxylase expression in normal
Chem. 260, 11488-11492. and pathological parathyroid glands. J. Clin.
1428. M. Burgos-Trinidad, R. Ismail, R.A. Ettinger, Endocrinol. Metab. 87, 2967-2972.
J.M. Prahl, and H.F DeLuca (1992). 1439. Brenza, H.L. and H.F DeLuca (2001). Analysis
Immunopurified 25-hydroxyvitamin D la- of basal regulatory elements in the 25-hydroxyvi-
hydroxylase and 1,25-dihydroxyvitamin D 24- tamin D3 la-hydroxylase gene promoter. Arch.
hydroxylase are closely related but distinct Biochem. Biophys. 388, 121-126.
enzymes. J. Biol. Chem. 267, 3498-3505. 1440. Baxter, L.A. and H.F DeLuca (1976).
1429. Arabian, A., J. Grover, M.G. Barre, and Stimulation of 25-hydroxyvitamin D3-la-hydro-
E.E. Delvin (1993). Rat kidney 25-hydroxyvita- xylase by phosphate depletion. J. Biol. Chem.
min D3 la- and 24-hydroxylases: Evidence for 251,3158-3161.
two distinct gene products. J. Steroid Biochem. 1441. Yoshida, T, N. Yoshida, T. Monkawa, M. Hayashi,
Mol. Biol. 45, 513-516. and T. Saruta (2001). Dietary phosphorus depri-
1430. Axen, E., H. Postlind, H. Sjoberg, and K. Wikvall vation induces 25-hydroxyvitamin D3 la-hydrox-
(1994). Liver mitochondrial cytochrome P450 ylase gene expression. Endocrinology 142,
CYP27 and recombinant expressed human 1720-1726.
CYP27 catalyze la-hydroxylation of 25-hydrox- 1442. Zhang, M.Y, X. Wang, J.T Wang,
yvitamin D3. Proc. Natl. Acad. Sci. USA 91, N.A. Compagnone, S.H. Mellon, J.L. Olson et al.
10014-10018. (2002). Dietary phosphorus transcriptionally reg-
1431. Monkawa, T., T. Yoshida, S. Wakino, T. Shinki, ulates 25-hydroxyvitamin D-la-hydroxylase
H. Anazawa, H.F.DeLuca et al. (1997). Molecular gene expression in the proximal renal tubule.
cloning of cDNA and genomic DNA for human Endocrinology 143, 587-595.
Human P450s 527

1443. Bland, R., E.A. Walker, S.V Hughes, P.M. Stewart, D3 la-hydroxylase gene in patients with
and M. Hewison (1999). Constitutive expression of pseudovitamin D-deficiency rickets. A^. Engl.
25-hydroxyvitamin D3-la-hydroxylase in a trans- J. Med 33H, 653-661.
formed human proximal tubule cell line: Evidence 1454. Smith, S.J., A.K. Rucka, J.L. Berry, M. Davies,
for direct regulation of vitamin D metabolism by S. Mylchreest, C.R. Paterson et al. (1999). Novel
calcium. Endocrinology 140, 2027-2034. mutations in the la-hydroxylase (P450cl) gene
1444. Takeyama, K., S. Kitanaka, T. Sato, M. Kobori, in three families with pseudovitamin D-
J. Yanagisawa, and S. Kato (1997). 25- deficiency rickets resulting in loss of functional
Hydroxyvitamin D3 la-hydroxylase and vitamin enzyme activity in blood-derived macrophages.
D synthesis. Science 111, 1827-1830. J. Bone Miner Res. 14, 730-739.
1445. Murayama, A., K. Takeyama, S. Kitanaka, 1455. Sawada, N., T. Sakaki, S. Kitanaka, K. Takeyama,
Y. Kodera, T. Hosoya, and S. Kato (1998). The S. Kato, and K. Inouye (1999). Enzymatic
promoter of the human 25-hydroxyvitamin D3 properties of human 25-hydroxyvitamin D3 la-
la-hydroxylase gene confers positive and nega- hydroxylase coexpression with adrenodoxin and
tive responsiveness to PTH, calcitonin, and NADPH-adrenodoxin reductase in Escherichia
la,25(OH)2D3. Biochem. Biophys. Res. Commun. coll Eur J. Biochem. 265, 950-956.
249, 11-16. 1456. Wang, X., M.Y Zhang, WL. Miller, and
1446. Kong, X.E, X.H. Zhu, Y.L. Pei, D.M. Jackson, A.A. Portale (2002). Novel gene mutations in
and M.F. Holick (1999). Molecular cloning, char- patients with la-hydroxylase deficiency that con-
acterization, and promoter analysis of the human fer partial enzyme activity in vitro. J. Clin.
25-hydroxyvitamin D3-la-hydroxylase gene. Endocrinol. Metab. 87, 2424-2430.
Proc. Natl. Acad. Sci. USA 96, 6988-6993. 1457. Muralidharan, K.R., M. Rowland-Goldsmith,
1447. Gao, X.H., PR Dwivedi, S. Choe, E Alba, A.S. Lee, G. Park, A.W. Norman, H.L. Henry
H.A. Morris, J.L. Omdahl et al. (2002). Basal and et al. (1997). Inhibitors of 25-hydroxyvitamin D3-
parathyroid hormone induced expression of the la-hydroxylase: Thiavitamin D analogs and bio-
human 25-hydroxyvitamin D la-hydroxylase logical evaluation. J. Steroid Biochem. 62,13-lS.
gene promoter in kidney AOK-B50 cells: Role of 1458. Hewison, M., D. Zehnder, R. Bland, and
Spl, Ets and CCAAT box protein binding sites. PM. Stewart (2000). la-Hydroxylase and the
Int. J. Biochem. Cell Biol. 34, 921-930. action of vitamin D. J. Mol. Endocrinol. 25,
1448. Michigami, T, H. Yamato, H. Suzuki, Y. Nagai- 141-148.
Itagaki, K. Sato, and K. Ozono (2001). Conflicting 1459. Kitanaka, S., K. Takeyama, A. Murayama,
actions of parathyroid hormone-related protein and and S. Kato (2001). The molecular basis of
serum calcium as regulators of 25-hydroxyvitamin vitamin D-dependent rickets type I. Endocrinol.
D3-la-hydroxylase expression in a nude rat J. 48, 427-432.
model of humoral hypercalcemia of malignancy. 1460. Portale, A.A. and W.L. Miller (2000). Human 25-
J. Endocrinol. Ill, 249-257. hydroxyvitamin D-la-hydroxylase: Cloning,
1449. Kato, S., T. Yoshizazawa, S. Kitanaka, mutations, and gene expression. Pediatr Nephrol.
A. Murayama, and K. Takeyama (2002). 14, 620-625.
Molecular genetics of vitamin D-dependent 1461. Kitanaka, S., A. Murayama, T. Sakaki, K. Inouye,
hereditary rickets. Horm. Res. 57, 73-78. Y. Seino, S. Fukumoto et al. (1999). No enzyme
1450. Wang, J.T., C.J. Lin, S.M. Burridge, O.K. Fu, activity of 25-hydroxyvitamin D3 la-hydroxylase
M. Labuda, A.A. Portale et al. (1998). Genetics gene product in pseudovitamin D deficiency rick-
of vitamin D la-hydroxylase deficiency in 17 ets, including that with mild clinical manifesta-
families. Am. J. Hum. Genet. 63, 1694-1702. tion. J. Clin. Endocrinol. Metab. 84, 4111^117.
1451. Portale, A.A. and W.L. Miller (2000). Human 25- 1462. Dardenne, O., J. Prud'homme, A. Arabian,
hydroxyvitamin D-la-hydroxylase: Cloning, EH. Glorieux, and R. St-Arnaud (2001). Targeted
mutations, and gene expression. Pediatr Nephrol. inactivation of the 25-hydroxyvitamin D3-la-
14, 620-625. hydroxylase gene (CYP27B1) creates an animal
1452. Porcu, L., A. Meloni, L. Casula, I. Asunis, model of pseudovitamin D-deficiency rickets.
M.G. Marini, A. Cao et al. (2002). A novel Endocrinology 142,3135-3141.
sphcing defect (IVS6+1G>T) in a patient 1463. Panda, D.K., D. Miao, M.L. Tremblay, J. Sirois,
with pseudovitamin D deficiency rickets. R. Earookhi, G.N. Hendy et al. (2001). Targeted
J. Endocrinol. Invest. 25, 557-560. ablation of the 25-hydroxyvitamin D la-hydrox-
1453. Kitanaka, S., K. Takeyama, A. Murayama, ylase enzyme: Evidence for skeletal, reproduc-
T Sato, K. Okumura, M. Nogami et al. (1998). tive, and immune dysfiinction. Proc. Natl. Acad.
Inactivating mutations in the 25-hydroxyvitamin Sci. USA 98, 7498-7503.
528 F. Peter Guengerich

1464. St-Arnaud, R., O. Dardenne, J. Prud'homme, oxysterol 7a-hydroxylase selective for 24-hydrox-
S.A. Hacking, and KH. Glorieux (2003). ycholesterol. ^ 5/0/. Chem. 275, 16543-16549.
Conventional and tissue-specific inactivation 1475. Lund, E.G., J.M. Guileyardo, and D.W. Russell
of the 25-hydroxyvitamin D-la-hydroxylase (1999). cDNA cloning of cholesterol 24-hydroxy-
(CYP27B1). J. Cell. Biochem. 88, 245-251. lase, a mediator of cholesterol homeostasis in the
1465. Satomura, K., Y. Seino, K. Yamaoka, Y Tanaka, brain. Proc. Natl. Acad Sci. USA 96, 7238-7243.
M. Ishida, H. Yabuuchi et al. (1988). Renal 25- 1476. Russell, D.W. (2000). Oxysterol biosynthetic
hydroxyvitamin D3-I-hydroxylase in patients enzymes. Biochim. Biophys.Acta 1529, 126-135.
with renal disease. Kidney Int. 34, 712-716. 1477. Bogdanovic, N., L. Bretillon, E.G. Lund,
1466. Correa, R, U. Segersten, P. Hellman, U Diczfalusy, L. Lannfelt, B. Winblad et al.
G. Akerstrom, and G. Westin (2002). Increased (2001). On the turnover of brain cholesterol in
25-hydroxyvitamin D3 la-hydroxylase and patients with Alzheimer's disease. Abnormal
reduced 25-hydroxyvitamin D3 24-hydroxylase induction of the cholesterol-catabolic enzyme
expression in parathyroid tumors—^new prospects CYP46 in glial cells. Neurosci. Lett. 314, 45-48.
for treatment of hyperparathyroidism with vitamin 1478. Papassotiropoulos, A., D. Lutjohann, M. Bagli,
D. J. Clin. Endocrinol. Metab. 87, 5826-5829. S. Locatelli, E Jessen, R. Buschfort et al. (2002).
1467. Maas, R.M., K. Reus, B. Diesel, W.I. Steudel, 24iS-hydroxycholesterol in cerebrospinal fluid is
W. Feiden, U. Fischer et al. (2001). Amplification elevated in early stages of dementia. J. Psychiatr
and expression of splice variants of the gene Res. 36, 27-32.
encoding the P450 cytochrome 25-hydroxyvita- 1479. Kolsch, H., D. Lutjohann, M. Ludwig, A. Schulte,
min D3 1 ,a-hydroxylase {CYP 27B1) in human U Ptok, F. Jessen et al. (2002). Polymorphism in
malignant glioma. Clin. Cancer Res. 7, 868-875. the cholesterol 245-hydroxylase gene is associ-
1468. Tangpricha, V, J.N. Flanagan, L.W. Whitlatch, ated with Alzheimer's disease. Mol. Psychiatry 7,
C.C. Tseng, T.C. Chen, PR. Hoh et al. (2001). 25- 899-902.
hydroxyvitamin D-la-hydroxylase in normal and 1480. Desai, P, S.T. DeKosky, and M.I. Kamboh
malignant colon tissue. Lancet 357, 1673-1674. (2002). Genetic variation in the cholesterol 24-
1469. Cross, H.S., P Bareis, H. Hofer, M.G. Bischof, hydroxylase (CYP46) gene and the risk of
E. Bajna, S. Kriwanek et al. (2001). 25- Alzheimer's disease. Neurosci. Lett. 328, 9-12.
Hydroxyvitamin D3-1 a-hydroxylase and vitamin 1480a.Mast, N., R. Norcross, U. Andersson, M. Shou, K.
D receptor gene expression in human colonic Nakayama, I. Bjorkhem, and LA. Pikuleva (2003).
mucosa is elevated during early cancerogenesis. Broad substrate specificity of human cytochrome
Steroids 66, 287-292. P450 46A1 which initiates cholesterol degradation
1470. Ogunkolade, B.W., B.J. Boucher, PD. Fairclough, in the brain. Biochemistry 42, 14284--14292.
G.A. Hitman, S. Dorudi, PJ. Jenkins etal. (2002). 1481. Aoyama, Y, Y Funae, M. Noshiro, T. Horiuchi,
Expression of 25-hydroxyvitamin D-1-a-hydrox- and Y Yoshida (1994). Occurrence of a P450
ylase mRNA in individuals with colorectal cdiX\- showing high homology to yeast lanosterol 14-
CQT. Lancet 359, 1831-1832. demethylase (P450,4p^) in rat liver. Biochem.
1471. Hsu, J.Y, D. Feldman, J.E. McNeal, and DM. Peehl Biophys. Res. Commun. 201, 1320-1326.
(2001). Reduced la-hydroxylase activity in human 1482. Aoyama, Y, T. Horiuchi, and Y Yoshida (1996).
prostate cancer cells correlates with decreased sus- Lanosterol 14-demethylase activity expressed in
ceptibility to 25-hydroxyvitamin Dj-induced rat brain microsomes. J. Biochem. (Tokyo) 120,
growth inhibition. Cancer Res. 61, 2852-2856. 982-986.
1472. Whitlatch, L.W, M.V Young, G.G. Schwartz, 1483. Rozman, D, M. Stromstedt, and M.R. Waterman
J.N. Flanagan, K.L. Burnstein et al. (2002). 25- (1996). The three human cytochrome P450 lanos-
Hydroxyvitamin D-la-hydroxylase activity is terol 14a-demethylase (CYP51) genes reside on
diminished in human prostate cancer cells and is chromosomes 3, 7, and 13: Structure of the two
enhanced by gene transfer. J. Steroid Biochem. retrotransposed pseudogenes, association with a
Mol. Biol. 81, 135-140. line-1 element, and evolution of the human CYP51
1473. Flanagan, J.N., L.W. Whitlatch, T.C. Chen, family. Arch. Biochem. Biophys. 333, 466-474.
X.H. Zhu, M.T Holick, X.R Kong et al. (2001). 1484. Cotman, M., D. Rozma, L. Banek, and D. Jezek
Enhancing la-hydroxylase activity with the (2001). Localisation of lanosterol 14a-demethylase
25-hydroxyvitamin D-la-hydroxylase gene in in round and elongated spermatids of the mouse
cultured human keratinocytes and mouse skin. testis: An immunoelectron microscopic and stereo-
J. Invest. Dermatol. 116, 910-914. logical study PJlugers Arch. 442, R167-R168.
1474. Li-Hawkins, J., E.G. Lund, A.D. Bronson, and 1485. Rozman, D (2000). Lanosterol 14a-demethylase
D.W Russell (2000). Expression cloning of an (CYP51)—a cholesterol biosynthetic enzyme
Human P450s 529

involved in production of meiosis activating 1496. Podust, L.M., J. Stojan, T.L. Poulos, and
sterols in oocytes and testis—a minireview. M.R. Waterman (2001). Substrate recognition
Pflugers Arch. 439, R56-R57. sites in 14 a-sterol demethylase from comparative
1486. Kelley, R.I, L.E. Kratz, R.L. Glaser, analysis of amino acid sequences and X-ray
M.L. Netzloff, L.M. Wolf, and E.W. Jabs (2002). structure of Mycobacterium tuberculosis CYP51.
Abnormal sterol metabolism in a patient with J. Inorg. Biochem. 87, 227-235.
Antley-Bixler syndrome and ambiguous geni- 1497. Marichal, P., L. Koymans, S. Willemsens,
talia. Am. J. Med. Genet. 110, 95-102. D. Bellens, R Verhasselt, W Luyten et al. (1999).
1487. Debeljak, N., M. Fink, and D. Rozman (2003). Contribution of mutations in the cytochrome
Many facets of mammalian lanosterol 14a- P450 14a-demethylase (Ergllp, CYP51p) to
demethylase from the evolutionarily conserved azole resistance in Candida albicans.
cytochrome P450 family CYP51. Arch. Biochem. Microbiology 145, 2701-2713.
Biophys. 409, 159-171. 1498. Lepesheva, G.I., L.M. Podust, A. Bellamine, and
1488. Stromstedt, M., D. Rozman, and M.R. Waterman M.R. Waterman (2001). Folding requirements are
(1996). The ubiquitously expressed human different between sterol 14a-demethylase
CYP51 cDNA encodes lanosterol 14a-demethy- (CYP51) from Mycobacterium tuberculosis and
lase, a cytochrome P450 whose expression is reg- human or fungal orthologs. J. Biol. Chem. 276,
ulated by oxysterols. Arch. Biochem. Biophys. 28413-28420.
329,73-81. 1499. Lamb, D.C., D.E. Kelly, M.R. Waterman,
1489. Rozman, D., M. Fink, G.M. Fimia, P Sassone- M. Stromstedt, D. Rozman, and S.L. Kelly
Corsi, and M.R. Waterman (1999). Cychc (1999). Characteristics of the heterologously
adenosine 3',5'-monophosphate(cAMP)/ cAMP- expressed human lanosterol 14a-demethylase
responsive element modulator (CREM)-depend- (other names: P45014DM, CYP51, P45051) and
ent regulation of cholesterogenic lanosterol inhibition of the purified human and Candida
14a-demethylase (CYP51) in spermatids. Mol. albicans CYP51 with azole antifungal agents.
Endocrinol. 13, 1951-1962. Yeast 15, 755-763.
1490. Haider, S.K., M. Fink, M.R. Waterman, and 1500. Rozman, D. and M.R. Waterman (1998).
D. Rozman (2002). A cAMP-responsive element Lanosterol 14a-demethylase (CYP51) and sper-
binding site is essential for sterol regulation of the matogenesis. DrugMetab. Dispos. 26, 1199-1201.
human lanosterol 14a-demethylase gene 1501. Majdic, G., M. Parvinen, A. Bellamine,
(CYP51). Mol. Endocrinol. 16, 1853-1863. H.J. Harwood, Jr., WW Ku, M.R. Waterman, and
1491. Yamashita, C , M. Kudo, H. Ishida, M. Noshiro, D. Rozman (2000). Lanosterol 14a-demethylase
Y. Aoyama, and Y Yoshida (2000). Insulin is the (CYP51), NADPH-cytochrome P450 reductase
essential factor maintaining the constitutive and squalene synthase in spermatogenesis: Late
expression of hepatic sterol 14-demethylase P450 spermatids of the rat express proteins needed to
(CYP51). J. Biochem. (Tokyo) 128, 93-99. synthesize follicular fluid meiosis activating
1492. Rodriguez, C , J. Martinez-Gonzalez, S. Sanchez- sterol. J. Endocrinol. 166, 463-474.
Gomez, and L. Badimon (2001). LDL downregu- 1502. Yamashita, C , Y Aoyama, M. Noshiro, and
lates CYP51 in porcine vascular endothelial cells Y Yoshida (2001). Gonadotropin-dependent
and in the arterial wall through a sterol regulatory expression of sterol 14-demethylase P450 (CYP51)
element binding protein-2-dependent mecha- in rat ovaries and its contribution to the production
nism. Circ. Res. 88, 268-274. of a meiosis-activating steroid. J. Biochem. (Tokyo)
1493. Lamb, D.C., N.N. Kaderbhai, K. Venkateswarlu, 130, 849-856.
D.E. Kelly, S.L. Kelly, and M.A. Kaderbhai (2001). 1503. Rozman, D., M. Cotman, and R. Frangez
Human sterol 14a-demethylase activity is enhanced (2002). Lanosterol 14a-demethylase and MAS
by the membrane-bound state of cj^ochrome ^^3. sterols in mammalian gametogenesis. Mol. Cell
Arch. Biochem. Biophys. 395, 78-84. Endocrinol. 187, 179-187.
1494. Lamb, D.C., D.E. Kelly, and S.L. Kelly (1998). 1504. Kodaira, H., C.A. Lisek, A. Arimura, I. Jardine,
Molecular diversity of sterol 14a-demethylase and S. Spector (1989). Identification of the con-
substrates in plants, fungi and humans. FEBS vulsant opiate thebaine in mammalian brain.
Lett. 425, 263-265. Proc. Natl. Acad. Sci. USA 86, 716-719.
1495. Podust, L.M., T.L. Poulos, and M.R. Waterman 1505. Kodaira, H. and S. Spector (1988).
(2001). Crystal structure of cytochrome P450 14a- Transformation of thebaine to oripavine, codeine,
sterol demethylase (CYP51) from Mycobacterium and morphine by rat liver, kidney, and brain
tuberculosis in complex with azole inhibitors. microsomes. Proc. Natl. Acad. Sci. USA 85,
Proc. Natl. Acad Sci. USA 98, 3068-3073. 1267-1271.
530 F. Peter Guengerich

1506. Dayer, P., J. Desmeules, T. Leemann, and demethylation by Escherichia coli AlkB
R. Striberni (1988). Bioactivation of the narcotic directly reverts DNA base damage. Nature 12,
drug codeine in human liver is mediated by 174-178.
the polymorphic monooxygenase catalyzing 1509. Welford, R.W., I. Schlemminger, L.A. McNeill,
debrisoquine 4-hydroxylation. Biochem. Biophys. K.S. Hewitson, and C.J. Schofield (2003). The
Res. Commun. 152,411^16. selectivity and inhibition of AlkB. J. Biol. Chem.
1507. Mikus, G., F. Bochner, M. Eichelbaum, P. Horak, 278, 10157-10161.
A.A. Somogyi, and S. Spector (1994). Endo- 1510. Pai, H.V, R.R Kommaddi, S.J. Chinta, T. Mori,
genous codeine and morphine in poor and exten- M.R. Boyd, and V Ravindranath (2004, in press).
sive metabolisers of the CYP2D6 (debrisoquine/ A frame shift mutation and alternate splicing
sparteine) polymorphism. J. Pharmacol. Exp. in human brain generates a functional form of
Then 268,546-551. the pseudogene, cytochrome P4502D7 that
1508. Trewick, S.C, T.K Henshaw, R.R Hausinger, demethylates codeine to morphine. J. Biol. Chem.
T. Lindahl, and B. Sedgwick (2002). Oxidative 39774.
11
Cytochrome P450 and the Metabolism
and Bioactivation of Arachidonic Acid
and Eicosanoids
Jorge H. Capdevila, Vijaykumar R. Holla, and John R. Faick

1. Introduction content stored in the AA metabolites, is limited


compared to that contained in complex informa-
The convergence of important advances in the tional molecules such as proteins or nucleic acids,
identification of several lipid-derived mediators as low energy cost, versatility, and rapid turnover,
inter- and intracellular signaling molecules, and in makes them efficient on/off molecular switches
the biochemistry of oxidative lipid metabolism, for rapid and efficient intra- or intercellular sig-
has focused interest in the functional roles of path- naling. Metabolism by prostaglandin H2 synthase
ways responsible for their formation, and the generates a cyclic endoperoxide, prostaglandin
physiological significance of their products. H2 (PGH2) that serves as the precursor for the
Among these, the studies of the enzymes of the formation of prostaglandins, prostacyclin, and
arachidonic acid (AA) cascade, consisting of thromboxanes^' ^. Metabolism by lipoxygenases
prostaglandin H2 synthase^'^, lipoxygenases^, and leads to the formation of several regioisomeric
cytochrome P450'^' ^, constitute a premier example hydroperoxides, the precursors of leukotrienes,
of the biological importance of these reactions and regioisomeric cis/trans conjugated hydroxye-
of their products (eicosanoids). Studies of the last icosatetraenoic acids (HETEs), lipoxins, and hep-
two decades, have implicated the enzymes of the oxilins^. Metabolism by microsomal cytochrome
AA cascade in the pathophysiology of diseases P450s (P450s) generates several hydroxy- and
such as hypertension, diabetes, and cancer, and epoxy-AA derivatives'^' ^. The reactions catalyzed
some of these enzymes serve as molecular targets by prostaglandin H2 synthase and lipoxygenases
for drugs of extensive use in clinical medicine, are mechanistically similar to those of the free-
including many nonsteroidal anti-inflanmiatory, radical-mediated autooxidation of polyunsaturated
antipyretic, and anti-asthmatic drugs^~^. The bio- fatty acids in that they are initiated by hydrogen
logical and signaling properties of eicosanoids are atom abstraction from a bis-allylic methylene
derived from the enzymatic, regio-, and stereo- carbon, followed by coupling of the resulting car-
selective oxygenation of AA, a rather simple bon radical to groimd state molecular oxygen. The
molecular template. While the informational kinetics, regiochemistry, and chirality of these

Jorge H. Capdevila • Departments of Medicine and Biochemistry, Vanderbilt University Medical School,
Nashville, TN. Vijaykumar R. Holla • Department of Medicine, Vanderbilt University Medical School,
Nashville, TN. John R. FaIck • Department of Biochemistry, Southwestern Medical Center, Dallas, TX.
Cytochrome P450: Structure, Mechanism, and Biochemistry, 3e, edited by Paul R. Ortiz de Montellano
Kluwer Academic / Plenum Publishers, New York, 2005.

531
532 Jorge H. Capdevila et al.

reactions are under strict enzymatic control. In pathophysiology of genetically controlled experi-
contradistinction to the c)^ochrome P450-catalyzed, mental hypertension^' ^. These earlier studies estab-
redox coupled, activation of molecular oxygen and lished P450 AA monooxygenation as a formal
delivery to ground state carbon, prostaglandin H2 metabolic pathway, P450 as an endogenous mem-
synthase and the lipoxygenases are typical dioxyge- ber of the AA metabolic cascade, and more impor-
nases that catalyze substrate carbon activation tantly, suggested functional roles for this enzyme in
instead of oxygen activation. the bioactivation of the fatty acid and thus, in cell
It is apparent from the literature that the P450 and organ physiology. Many of the biological activ-
gene superfamily of hemoproteins is, as a group, ities attributed to the P450-derived eicosanoids,
one of the most intensively studied enzyme sys- as well as the potential physiological importance of
tems and yet our knowledge of their endogenous these reactions, have been reviewed^"^^.
metabolic or physiological roles remains limited. Prior to the demonstration of AA metabolism
This is partly due to the complexity of mammalian by P450, several groups demonstrated the role of
P450 isoforms, and the wide structural diversity microsomal P450s in the (o/w-l hydroxylation of
of substrates known to be metabolized by these prostanoids^^"^^ and, more recently, leukotrienes^^.
proteins. This catalytic versatility pointed to, and Most of these reactions are considered to be
served as the basis for, many of the documented involved in eicosanoid catabolism and excretion,
roles for P450 in the metabolism of foreign chem- but their potential relevance in eicosanoid bioacti-
icals, and has contributed to establish its toxico- vation or inactivation, and/or in the control of
logical and pharmacological importance. In the organ/cell eicosanoid levels has only begun to be
last few years, there has been an increasing inter- explored. We will first discuss the role of P450
est in the understanding of the physiological sig- in the metabolism of eicosanoids, and then con-
nificance of the P450 enzyme system, and its centrate on the studies of its role in AA metabolism
role(s) in the metabolism of endogenous sub- and bioactivation.
strates. In this regard, the studies of the P450
branch of the AA cascade have provided a new
focus to these efforts, and far-reaching results 2. Metabolism of Eicosanoids
from several laboratories are generating new par-
adigms in fatty acid metabolism, as well as in cell
During the metabolism of eicosanoids,
and organ physiology^^^.
depending on the nature of the oxygenated sub-
The studies of the role of P450 in the metabo- strate, P450 catalyzes both NADPH-dependent
lism and bioactivation of AA were initiated in 1981 and -independent reactions. This differential
with the demonstration that liver and kidney requirement for NADPH-mediated changes in the
microsomal fractions, as well as purified P450 redox state of the heme-iron illustrates the marked
isoforms^ ^"^^ actively catalyzed the NADPH- differences in oxygen chemistries for these reac-
dependent, oxidative metabolism of AA to prod- tions, that is, the isomerization of AA peroxides,
ucts that were different from prostanoids and vs the more demanding activation and delivery of
leukotrienes"'^. The widely documented physio- a reactive form of atomic oxygen to ground state
logical importance of AA suggested that these carbon-hydrogen bonds.
observations were unique and likely to be func-
tionally significant, and led to the rapid structural
characterization of most P450-eicosanoids, their 2.1. NADPH-lndependent
chemical synthesis, and subsequent biological
Reactions
evaluation"^'^. Interest in these novel P450 reac-
tions was stimulated by: (a) the initial demonstra- P450 catalyzes the isomerization of a variety
tion that some of the products displayed potent of fatty acid hydroperoxides, including 15-hydro-
biological activities, including the inhibition of peroxyeicosatetraenoic acid (15-HPETE)^^' ^^,
Na^ reabsorption in the distal nephron^^, (b) the and of the prostaglandin H2 (PGH2) endoperox-
documentation of P450 participation in the in vivo ide^^. A distinctive feature of some P450 fatty acid
metabolism of endogenous AA pools^^, and (c) the peroxide isomerases is their inability to accept
proposal of a role for these enzymes in the electrons from NADPH and to activate molecular
Cytochrome P450 and the Metabolism of AA and Eicosanoids 533

oxygen^ ^"^^. Moreover, while all these enzymes terminal (C20 or co-carbon) or penultimate carbon
possess a heme-thiolate prosthetic group, their atoms (C J9 or (0-1 carbon). However, the epoxida-
overall homology to other members of the P450 tion of infused PGI2 by a perfused kidney prepa-
gene superfamily is limited and suggests an early ration'^, and the metabolism of 5,6- and
evolutionary functional specialization^^"^^. The 8,9-EET by prostaglandin H2 synthase, were
mechanism by which the hemoprotein cleaves the described several years ago'^' ^^. The former leads
peroxide oxygen-oxygen bond, that is, homolytic to a variety of 5,6-oxygenated prostanoids'^.
or heterolytic scission, plays a decisive role in Oxidation of the latter was stereodependent,
determining the catalytic outcome of these reac- that is, 8(5),9(i?)-EET formed ll(i?)-hydroxy-
tions and is highly dependent on the nature of 8(-S),9(i?)-epoxyeicosatrienoic acid exclusively,
the P450 isoform, the chemical properties of the whereas the 8(/?),9(*S)-enantiomer formed both
organic peroxide, and the nature of the oxygen Cjj and Cj5 hydroxylated metabolites^^. A
acceptor^^"^^' ^^. A homolytic pathway was proposed detailed study of the secondary metabolism of
for the formation of 11- and 13-hydroxy-14, 12(i?)-HETE and 14,15-EET by P450 has been
15-epoxyeicosatrienoic acids (EETs) from 15- reported^ ^' ^'. The efficient w/w-l oxidation of
HPETE by rat liver microsomes'^. Prostacyclin EETs by rat CYP4A isoforms to the correspon-
and thromboxane synthases are P450-like proteins ding regioisomeric epoxy-alcohols at rates com-
containing a heme-thiolate prosthetic group'^~'^. parable to those observed with lauric acid, a
The heterolytic cleavage of PGH2 and an oxygen prototype substrate for these enzymes, was pub-
atom transfer or oxenoid mechanism has been lished recently^^. One of these metabolites, the
proposed to account for the P450-catalyzed forma- o)-alcohol of 14,15-EET, was shown to bind and
tion of prostacyclin (PGI2) and thromboxane A2 activate the peroxisomal proliferator activated
(TXA2)'^. The participation of P450s in the bio- receptor alpha (PPAR^) type of nuclear receptor^ ^.
synthesis of these important mediators of endothe- It is important to note that, with the exception of
lial cell and platelet function was one of the first the (o/o)-! hydroxylation of prostanoids and
demonstrations of a role for this enzyme system in leukotrienes, none of the transformations described
vascular biology. However, its pharmacological and above has been shown to occur in vivo from
clinical implications remain to be fully explored. endogenous precursors.

2.2. NADPH-Dependent 2.2.1. ct)/(o-1 Oxidation of


Reactions Prostanoids
P450 plays an important role in the NADPH- Since the initial report of in vivo w-l hydroxy-
dependent metabolism of several bioactive oxy- lation of prostanoids in 1966^^' ^^, w- and o)-l
genated eicosanoids^^'^. These reactions are of hydroxylation has become a recognized route of
importance in that they: (a) increase eicosanoid prostanoid metabolism. Early studies of prostanoid
structural diversity and, hence, modify informa- o)/o)-l hydroxylation demonstrated these reactions
tional content, (b) may alter the pharmacological were NADPH-dependent, localized to the endo-
profile of the substrate, and (c) may participate plasmic reticulum^^' ^^, and catalyzed by microso-
in the regulation of steady state and/or stimulated mal P450 (ref [36]). Incubations with purified
levels of physiologically relevant molecules. While enzymes or recombinant P450s showed that most
these oxidations were generally viewed as catabolic, of these reactions were catalyzed by members of
that is, yielding an attenuated biological activity, the 4 gene family of P450s'^' ^^^^. In general, while
recent studies indicate that some w-oxidized pro- CYP4F isoforms are more active in the metabolism
stanoids show unique and potent biological proper- of eicosanoids than fatty acids, the opposite appears
^jgg6-io However, in most cases the sequence of to be true for most CYP4A isoforms'^' ^^.
steps leading to w/w-l oxidized prostanoids from Approximately 18 CYP4F isoforms have been
endogenous AA pools remains to be clarified. identified in rats (4F1, 4F4, 4F5, and 4F6)4^ mice
Typically, the P450-dependent metabolism of (4fl3, 4fl4, 4fl5, 4fl6, 4fl7, 4f37, 4 0 9 , and
eicosanoids results in the hydroxylation of their 4f40)45, and humans (4F2, 4F3, 4F8, 4F11, 4F12,
534 Jorge H. Capdevila ef a/.

Table 11.1. Metabolism of Fatty Acids and Prostanoids by


Cytochrome P450 4A Isoforms

4A isoform Species Enzymatic activities"

4A1 Rat (o-oxidation of laurate and arachidonate


4A2 Rat co/w-l oxidation of laurate and arachidonate
4A3 Rat (o/o)-! oxidation of laurate and arachidonate
4A8 Rat (o/w-l oxidation of laurate and arachidonate
4A4 Rabbit (rt-oxidation of palmitate, arachidonate, and of
prostaglandins A, E, D, and F2^
4A5 Rabbit co/(x)-l oxidation of laurate and palmitate,
some a)-oxidation of PGAj and arachidonate
4A6 Rabbit w-oxidation of laurate, palmitate, and
arachidonate. Low PGAj w-oxidation
4A7 Rabbit (o-oxidation of laurate, palmitate, arachidonate,
and PGAj, inactive toward PGE2
4al0 Mouse (O-oxidation of laurate
4al2 Mouse co/o)-] oxidation of laurate and arachidonate
4al4 Mouse (o/o)-1 oxidation of laurate
4A11 Human (o/to-1 oxidation of laurate and arachidonate
4a22 Human Unknown

''Compiled from references [4]-[10], [17]-[20], [36H44], [46]-[52].

and 4F22)'^^. On the other hand, approximately 11 to catalyze the o)-l hydroxylation of PGH2, the pre-
CYP4A isoforms have been cloned and/or isolated cursor of all prostanoids"^^. Based on its catalytic
and purified from rats (4A1,4A2,4A3, and 4A8)'^^, activity, and its high levels of expression in the
mouse (4a 10, 4a 12, and 4a 14)"^^, and rabbits (4A4, seminal vesicles it was proposed that CYP4F8 is
4A5, 4A6, and 4A7)'^^. In stark contrast with the involved in the formation of 19-hydroxy-PGE2,
known multiplicity of rodent CYP4A and of rodent present at high concentrations in human seminal
and human CYP4F isoforms, only two highly fluid34, 43 CYP4F12 is a regioselective AA a)-3
homologous CYP4A genes, CYP4A11 and hydroxylase^^ but, it is also active in the hydro-
CYP4A22, have been identified in humans"*^"^^. xylation of prostanoids and several prostanoid
Most CYP4A enzymes that have been character- analogs^^.
ized enzymatically, are either inactive toward
prostanoids or catalyze their (o- or o)/o)-1 hydroxy-
lation at rates that are generally substantially lower 2.2.2. a>/co-1 Oxidation of
than fatty acid hydroxylation (Table 11.1)^^' ^^~^'
Leukotrienes and Other
^^~^K A special case is that of rabbit lung CYP4A4,
Eicosanoids
an isoform induced during pregnancy^^' ^^' ^^' ^^
and active in the co-hydroxylation of several The (O-oxidation of leukotriene B4 (LTB^), an
prostanoids, including PGE2 (Table 11.1)^^' ^^. As important proinflammatory mediator^^' ^^, has
with AA, none of the CYP4A isoforms character- been documented in whole animals, isolated cells,
ized to date is selective for the a)-l carbon of and subcellular fractions^^' ^^^o, and shown to be
prostanoids (Table 11.1)^^' ^^^^' ^^~^^. A more spe- catalyzed by a unique P450 isoform, distinct from
cific role for CYP4F isoforms as predominantly those involved in fatty acid and prostanoid metab-
prostanoid and eicosanoid w/co-l hydroxylases has olism^^' ^^~^^. Soon after, the cDNA coding for
emerged during the last few years^^. The cDNA CYP4F3 was cloned and expressed, and recombi-
coding for CYP4F8 was cloned from human semi- nant CYP4F3 shown to catalyze the o)-oxidation
nal vesicles, and the recombinant protein was shown of LTB4 with a K^ of 0.71 luiM^i' ^2. A role for
Cytochrome P450 and the Metabolism of AA and Eicosanoids 535

CYP4F3 as an endogenous LTB^ hydroxylase is 3. Metabolism of Arachidonic


supported by its selective expression in human
Acid: The Arachidonic Acid
polymorphonuclear leukocytes, and its lack of
activity toward fatty acids such lauric, palmitic, IVIonooxygenase
and AAs^O' ^i, 62 CYP4F3 also supports the
(o-hydroxylation of lipoxygenase metabolites such As with the other enzymes of the AA
as lipoxins A and B, and of 5- and 12-HETE^2. A metabolic cascade, P450 metabolizes only free,
splice variant of CYP4F3, CYP4F3B, is expressed nonesterified forms of AA and thus, in vivo
in liver and kidney, and shows significant struc- metabolism requires the release of the fatty acid
tural and fiinctional similarities to CYP4F2^^. from selected glycerophospholipid pools. CYP
Human CYP4F2 and rat 4F1 are active LTB^ P450, prostaglandin H2 synthase, and lipoxyge-
(o-hydroxylases capable of HETE (o-hydroxyla- nases are capable of metabolizing polyunsaturated
tion^^' ^^' ^'^. CYP4F2 is expressed in human liver fatty acids other than AA, however, it is the unique
and kidney, and responsible for most of the nature of the AA containing phospholipids, and
hepatic hydroxylation of LTB^^^. Four members of the control of its release by hormonally sensitive
the rat 4F gene subfamily (CYPs 4F1, 4F4, 4F5, phospholipases that makes the oxidative metabo-
and 4F6) have been cloned"^^. Recombinant CYPs lism of AA distinctive, and fimctionally important.
4F1,4F4, and 4F5 catalyze the o)-hydroxylation of Under conditions favoring primary metabolism,
LTB4, and CYP4F1 also metabolizes lipoxins and the P450 AA monooxygenase oxidizes AA by one
HETEs64' 66 xhe co-oxidation of 12(5)-HETE by or more of the following of reactions: (a) bis-
polymorphonuclear leukocytes was demonstrated allylic oxidation (lipoxygenase-like reaction) to
in 1984 by Wong et al^^ and Marcus et al^^. generate any of six regioisomeric HETEs contain-
Moreover, the latter authors further showed that ing a cis,trans'Con]u%2iiQ6. dienol functionality
endogenous AA pools are converted to 12,20- (5-, 8-, 9-, 11-, 12-, and 15-HETEs) (Figure 11.1),
dihydroxyeicosatetraenoic acid by a co-incubated (b) Hydroxylations at or near the terminal sp^
mixture of human platelets and polymorpho- carbon (AA w/w-l hydroxylase) affording 16-, 17-,
nuclear leukocytes, thus providing one of the 18-, 19-, and 20-HETEs (16-, 17-, 18-, 19-, and
first examples of intercellular eicosanoid meta- 20-HETE) (o), (0-1, (0-2, o)-3, and (J()-4 alcohols)
bolism^^ Both 5- and 15-HETE are known to (Figure 11.1), and (c) Olefin epoxidation (AA
undergo o)-oxidation by P450^^' ^^. epoxygenase) frimishing four regioisomeric EETs

AA MONOOXYGENASE

AA Epoxygenase Lipoxygenase-like AA (DI(J>1 Hydroxylase

5,6-EET 5,6-DHET 5-, 8-HETE 16-HETE (co-4)


8,9-EET ^ 8,9-DHET 9-, 11-HETE 17-HETE (co-3)
11,12-EET 11,12-DHET 12-HETE 18-HETE (co-2)
14,15-EET 14,15-DHET 15-HETE 19-HETE (co-1)
20-HETE (CO)

Figure 11.1. The cytochrome P450 arachidonic acid monooxygenases.


536 Jorge H. Capdevila et al.

(5,6-, 8,9-, 11,12-, and 14,15-EETs) (Figure 11.1). unresolved issues continue to offer a major
The classification of P450-derived eicosanoids in challenge for this area of research.
Figure 11.1 continues to provide a rational and
useful framework for most of the studies of this
3.1. bis-Allylic Oxidation
branch of AA metabolic cascade"^' ^.
The chemistry of the P450-derived eicosanoids
(Lipoxygenase-Like
is highly dependent on the tissue source of Reactions)
enzymes, animal species, sex, age, hormonal sta- The products of these reactions are structurally
tus, diet, and exposure to xenobiotics"^' ^. For similar to those of plant and mammalian lipoxy-
example, EETs are the predominant products gen- genases, and yet there is no evidence that
erated by most liver microsomal fractions (>70% hydroperoxide intermediates are formed during
of total products), while most kidney microsomal the P450-catalyzed reactions^' ^' ^^' ^^. A mecha-
fractions generate mainly a mixture of 19- and nism for P450-dependent HETE formation
20-HETE (77% of total products)^. Studies with involving bis-allylic oxidation at either C7, CIO,
microsomal, purified, and/or recombinant forms or CI3, followed by acid-catalyzed rearrangement
of rat, rabbit, and human P450s'^'^ showed that the to the corresponding cis-trans dienols was pro-
hemoprotein controls, in an isoform-specific fash- posed, and the intermediate 7-, 10-, and 13-
ion, oxygen insertion into the fatty acid template HETEs isolated^^' ^^ sj^ce 12(i?)-HETE is the
at three levels: (a) type of reaction, that is, olefin
predominant enantiomer generated by a P450-
epoxidation, bis-allylic oxidation, or hydroxyla- catalyzed reaction^^, it was thought that all the
tions at the Cjg-C2Q sp^ carbons, (b) regioselectiv-mammalian 12(i?)-HETE was a product of the
ity of oxygen insertion, that is, epoxidation at P450 enzyme system^^ However, the cloning and
either of the four olefin bonds, allylic oxidation characterization of mammalian 12(i^)-lipoxyge-
initiated at any of the three bis-allylic methylene,nases has led to a reevaluation of the role of P450s
or hydroxylation at C^^-C2Q (ref [37]), and (c) the in 12(/?)-HETE biosynthesis^^' ^l The formation
enantiofacial selectivity of oxygenation leading to of 12(5)- and 12(i^)-HETE by P450-independent
chiral products. The broad structural, functional, and -dependent pathways in bovine cornea
regulatory, and catalytic redundancy displayed by epithelium has been reported^^' ^^. Importantly,
many P450 isoforms, as well as their often over- in vitro studies showed that 12(i?)-HETE is a pow-
lapping patterns of tissue expression, continues erful and enantioselective inhibitor of Na^/K+
to complicate the task of assigning catalytic roles ATPase^'*. Finally, the enzymatic formation of
to a P450 or to a group of P450 isoforms. This 12(i^)-hydroxy-5,8,14-eicosatrienoic acid (12(i?)-
is of current interest because many of the P450 HETrE), an ocular proinflammatory and vasodila-
eicosanoids are biologically active^"'^, and the tory substance in rabbits, has been described^^.
identification and characterization of P450 iso- Areas in need of clarification are: (a) the role of
forms involved in the in vivo metabolism of AA P450 in the biosynthesis of endogenous HETE
is needed for accurate molecular descriptions of and HETrE pools, (b) the identity and molecular
their mechanism of action, regulatory control, and properties of the P450 isoforms responsible
ultimately, physiological significance. For exam- for these reactions, and (c) the contributions of
ple, several CYP 2B, 2C, 2D, 2E, and 2J proteins P450 and 12-lipoxygenases to organ-specific
have been shown to catalyze the in vitro oxidation 12(ie)-HETE and 12-HETrE biosynthesis.
of AA to hydroxy- and/or epoxy-acids^'"^^; how-
ever, their participation in metabolism of the
endogenous fatty acid pools remains unclear^' ^^.
In a few cases, a role for individual 2 gene family 3.2. Hydroxylation at C^g-Cgo
isoforms in endogenous AA bioactivation has ((o/co-1 Hydroxylase
been suggested based on enzymatic and/or Reactions)
immunological evidence^^' ^^' ^^. Nevertheless,
the identities of the P450 isoforms responsible 3.2.1. Introduction
for organ-specific AA metabolism remains pre- The hydroxylation of saturated medium-chain
liminary (and many cases speculative) and these fatty acids at their ultimate and penultimate carbons
Cytochrome P450 and the Metabolism of AA and Eicosanoids 537

was one of the first enzymatic activities attributed epithelium, and anterior pituitaries^^^. However,
to microsomal P450s^^. In general, medium-chain it is in renal tissues that these reactions are best
saturated fatty acids (Cj2-Ci5) are far better sub- characterized, most prevalent, and have been
strates for the microsomal co/w-l hydroxylases assigned their most important functional roles"^"^^.
than AA^^' ^^' ^^' ^^, and reaction rates decrease as Extensive biological, enzymatic, and molecular
the substrate carbon-chain length increases from evidence shows that the CYP4A isoforms are the
C12 to CI8. For example, lauric acid, a fatty predominant, and functionally relevant, AA w/w-l
acid absent from most mammalian tissues, is hydroxylases in the mammalian kidney"^"^^' ^^^. The
metabolized by the microsomal w/w-l hydroxy- CYP4A gene subfamily encodes a group of struc-
lases or by purified CYP4A isoforms at rates turally and functionally conserved proteins that
significantly higher than AA^^' ^^' ^K Common are specialized for fatty acid oxidation and that
oxygen chemistries and reaction mechanisms for show little or no activity toward xenobiotics^^' ^^.
these reactions are suggested by the fact that, The expression of the CYP4A fatty acid hydroxy-
regardless of the carbon length of the fatty acid or lases is regulated by a variety of physiological
its degree of saturation, the a)/o)-l hydroxylases and pathophysiological effectors such as age, sex
deliver a reactive form of oxygen to ground state, hormones, dietary lipids, fasting, starvation, min-
sp^ carbons. However, the unequal chemical reac- eralocorticoids, insulin, diabetes, and hyperten-
tivities of the carbon atoms in the AA molecular sion"^"^^' 100-108 Moreover, the sexual dimorphic,
template impose additional steric requirements on androgen sensitive, expression of rat kidney CYPs
the P450 catalyst. Hydroxylation at the thermody- 4A2 and 4A8, and of mouse kidney Cyp4al2 have
namically less reactive C^^ through C2Q rather than been demonstrated^o^' ^^4, i09,110^
at the chemically comparable C2 through C^ indi- In rats and rabbits, the 4A gene subfamily is
cates a rigid and highly structured binding site for composed of four highly homologous genes"^^.
the AA molecule. This binding site must position Amino acid sequence analysis showed that the rat
the acceptor carbon atom(s) in optimal proximity 4A proteins could be divided into two groups that
to the heme-bound active oxygen, with complete share > 7 1 % overall homology (Figure 11.2)"^^.
segregation of the AA-reactive olefins and bis- CYPs 4A1 and 4A8 (76% sequence identity)
allylic methylene carbons. Studies with CYP102 constitute one group, and the other is composed
(P450BM3), a high turnover bacterial AA hydrox- of the highly homologous CYPs 4A2 and 4A3
ylase of known atomic structure^^' ^^, suggested a (98% sequence identity) (Figure 11.2)"^^. The high
rigid active-site binding geometry for AA, and level of nucleotide sequence identity shared by the
indicated that the regiochemistry of P450 oxygen CYPs 4A2 and 4A3 genes extends into their
insertion was determined by the fatty acid binding intronic areas, suggesting that they arose from
coordinates, and not by chemical properties of the a relatively recent gene duplication event"*^. The
acceptor carbon or the heme-bound active oxygen three characterized murine Cyp4a genes are
species^^' ^^. Thus, X-ray crystallography, molecu- localized in a ---200 kb segment of chromosome 4
lar modeling, site-specific mutational analysis,
as well as enzymatic studies indicate that the
"substrate access channel" in CYP BM-3, holds
the AA molecule in a rigid orientation that:
RAT P450 4A GENE SUB FAMILY
(a) precludes significant rotation and/or displace-
4A2 -, r4Al (4A10)
ment along the channel's longitudinal axis, and
(4A14) 96% 65%
(b) shields the heme-bound oxidant from non-
4A3^ L4A8 (4A12)
acceptor carbons^^' ^^.

60%
3.2.2. Enzymology, Isoform Specificity
CYP 4
AA o)/(o-l hydroxylation has been observed in
microsomal fractions from several organs, includ- Figure 11.2. Nucleotide sequence identity between
ing liver, kidney, brain, lung, intestine, olfactory rat and murine CYP4A isoforms.
538 Jorge H. Capdevila et al.

(ref. [110]). Cyps 4a 10 and 4a 12 are the murine kinetic and immunological evidence suggested
homologs of rat CYPs 4A10 and 4A8, respec- that both CYPs 4A11 and 4F2 may contribute to
tively (Figure 11.2)"^^. The presence of a single the biosynthesis of 20-HETE by human kidney
murine gene (CYP4al4) highly homologous to microsomes and nephron segments"*"^.
both rat CYPs 4A2 and 4A3 indicates that the The liver microsomal P450 hydroxylation of
4A2/4A3 gene duplication event occurred after AA at C^g, Cj-7, Cjg, and C^^, but not at C20,
the evolutionary separation of rat and mouse was induced after treatment of the animals with
(Figure 11.2). Southern analysis and the human a-naphthoflavone or dioxin^^^' ^^^. Reconstitution
genome database show that CYPs 4All and experiments using purified liver CYPs lAl and
4A22 are likely to be the only members of 1A2, the major liver P450 isoforms induced by
the CYP4A gene subfamily in humans^^^^' ^^l these chemicals, demonstrated that CYPs lAl and
These two genes share 96% nucleotide sequence 1A2 were more or less regioselective for oxidations
identity, contain 12 exons, and have similar at the AA Cj^-C,9 carbons (87% and 44% of total
intron/exon distributions'^^. The cDNA coding for products for CYPlAl and 1A2, respectively)^^^
CYP4A11 has been cloned, expressed, and char- Furthermore, while CYPlAl oxidized AA prefer-
acterized as an active renal fatty acid oo-hydroxy- entially at C,g, oxygenation by CYP1A2 occurred
lase"^"^' ^^' '*'^' ^^ On the other hand, the enzymatic predominantly at C,^ (ref [111]). It is of interest
activity of CYP4A22 is unknown, and its mRNA that despite a very limited sequence homology,
is expressed in kidney at levels that can be CYPs lA and 4A show distinct regioselectivities
detected only after RT-PCR amplification"^^. for the adjacent Cjg and C2Q carbons of AA.
Table 11.1 summarizes the published fatty acid Purified CYP2E1, an isoform induced in rat liver
metabolic properties for several purified and by diabetes, fasting, and alcohol, converts AA
recombinant CYP4A isoforms. All enzymatically stereoselectively to 19(5)- and 18(i?)-HETE as
characterized CYP4A proteins (either purified its major reaction products^^. Finally, members of
or recombinant proteins) catalyze saturated fatty the 2J gene subfamily are also active AA w-l
acid (o-oxidation and most also hydroxylate AA hydroxylases^^' ^^ and, recently, Cyp 2j9, an iso-
at either the C20, or the C,g and C20 carbon form expressed in mouse brain was cloned,
atomsi6-i9, 38-12, 44, 46, 47, 49-52 ^o date, nonc of expressed and shown to be a regioselective AA
them have been shown to be selective for fatty o)-l hydroxylase " \
acid (0-1 hydroxylation. Despite their high struc- The potent biological activities attributed to
tural homology, CYP4A2 metabolizes AA while the products of the AA (JO/CO- 1 hydroxylases have
CYP4A3 is either inactive^^, or reacts at very low stimulated an intense search for the physiological
rates49' ^K Both CYP 4A2 and 4A3 are, on the and/or pathophysiological roles of these reac-
other hand, active lauric acid w/co-l hydroxy- tions'^-'^. Among these, 20-HETE4-IO has been
lases^^' ^^ A microsomal form of recombinant rat characterized as: (a) a powerful vasoconstrictor of
CYP4A2 was shown to oxidize AA to 20-HETE the renal and cerebral microcirculations, (b) an
and 11,12-EET'^^. In contrast, two different inhibitor of vascular calcium-dependent K chan-
laboratories showed that purified recombi- nels, (c) a regulator of Na^/K^ ATPase activity,
nant CYP4A2 oxidizes AA to only 19- and 20- and (d) a modulator of Ca^^ and CI" fluxes.
HETE^^' ^^ All three murine Cyp4a proteins are Furthermore, analysis of the segmental distribu-
active lauric acid hydroxylases, but only Cyp4al2 tion of CYP 4A isoforms along the rat nephron is
catalyzes AA co/co-l hydroxylation, providing an consistent with many of the proposed renal
explanation for the low levels of 20-HETE syn- actions of 19- and 20-HETE"l A role for 20-
thase activity present in microsomes isolated from HETE as a powerful mitogen in cultured kidney
the kidneys of female 129SvJ mice'^^. An unre- epithelial cells, as well as in vasopressin, parathy-
solved issue is that of the relative roles played by roid hormone and norepinephrine signaling has
CYPs 4A11 and 4F2 in the biosynthesis of 20- been described^"^^. Of importance during analyses
HETE by human kidney As discussed, recombi- of the functional significance of the AA (o/w-l
nant CYP4F2 is an active LTB^ hydroxylase^^' ^\ hydroxylases is the recognition that, as discussed,
and it has been reported to be active toward many P450 fatty acid w/w-l hydroxylases also
AA co-hydroxylation^^' ^^. On the other hand. play important roles in the metabolism of
Cytochrome P450 and the Metabolism of AA and Eicosanoids 539

bioactive eicosanoids such as prostanoids and significance of EET hydration or GSH conjuga-
leukotrienes. tion remain mostly unexplored, although recently
The expression of several CYP4A isoforms is a role for cytosolic epoxide hydrolase in the regu-
under transcriptional control by the nuclear lation of antihypertensive EETs levels has been
PPAR^^^^ The coordinated, PPAR^-controlled, proposed^^^' ^^^. The catalysis of AA epoxidation,
induction of peroxisomal fatty acid p-oxidation or the presence of endogenous EET pools has
and microsomal co/w-l hydroxylation, has sug- been demonstrated using microsomal fractions
gested a role for CYP4A isoforms in hepatic lipol- or samples obtained from numerous tissues,
ysis and fatty acid homeostasis, and a role for including liver, kidney, lung, skin, pituitary, brain,
these isoforms in PPARa-signaling has been adrenal, endothelium, and ovaries"^"^^.
advanced based on gene knockout studies^^^. The
potential for an involvement of CYP4A isoforms
in fatty acid and lipid homeostasis is opening new 3.3.2. Enzymology, Isoform Specificity
opportunities for an understanding of the physio-
The isoform multiplicity of the AA epoxyge-
logical roles of these enzymes, vis-a-vis their
nase was first suggested by changes in the regio-
recognized functional roles as AA hydroxylases.
and stereoselectivity of the microsomal enzymes,
resulting from animal treatment with known P450
3.3. Olefin Epoxidation inducers^ ^ For example, animal treatment with
(Epoxygenase Reactions) phenobarbital inverted the overall enantiofacial
selectivity of the rat liver microsomal AA epoxy-
3.3.1. Introduction genases (Table 11.2), and caused marked increases
in the organ levels of endogenous of S(S),9(R)-
The demonstration of NADPH-dependent EET, an effect likely due to the induction of CYPs
metabolism of AA to 11,12- and 14,15-dihydroxy- 2B1 and 2B2, both of which are highly stereose-
eicosatrienoic acids (DHETs) by microsomal lective 8(*S),9(i?)-epoxygenases^^ These studies
incubates indicated a role for P450 in AA epoxi- showed that the regio- and stereochemical selectiv-
dation^l Soon after, 5,6-, 8,9-, 11,12-, and 14, ity of the microsomal AA epoxygenase was under
15-EET were isolated and shown to be products of regulatory control and could be altered, in vivo, by
the P450-dependent metabolism of AA^^^. In animal manipulation"^' ^' ^^ Subsequently, it was
mammals, the epoxidation of polyunsaturated demonstrated that arachidonate epoxidation was
fatty acids to nonallylic, cis-epoxidQs is unique to highly asymmetric and that P450s control, in an
the P450 enzyme system and, in contrast with
fatty acid co/w-l hydroxylation, is more or less
selective for AA^' ^. Thus, while the enzymatic or
Table 11.2. Effect of Phenobarbital Treatment
nonenzymatic reduction and/or isomerization of
on the Enantioselectivity of the Liver
polyunsaturated fatty acid hydroperoxides can
Microsomal Arachidonic Acid Epoxygenase
yield epoxides or epoxy-alcohol derivatives, these
products are structurally different from those Liver microsomes
generated by the P450 enzymes^' ^^. The EETs are EET enantiomer
bis-allylic epoxides and as such, remarkably (% distribution) Control Phenobarbital
resistant to attack by nucleophiles such as water
and glutathione (GSH). However, in most cells 8(5),9(i?)-EET 32 ± 2 78 ± 2
and organ tissues, the EETs are metabolically 8(i?),9(5)-EET 68 ± 2 22 ± 2
unstable and are rapidly esterified to glycero- U{S),l2{RyEET 19 ± 1 83 ± 2
phospholipids, degraded by fatty acid p-oxidation ll(RX12(S)-EET 81 ± 1 17 ± 2
pathways^^^' ^^^, conjugated to GSH^^^, and/or l4(S),l5{RyEET 67 ± 2 25 ± 3
hydrated and excreted^ ^^. Cytosolic epoxide hydro- 14(R19(S)-EET 33 ± 2 75 ± 3
lase and GSH-transferases catalyze the enzymatic
conversion of EETs to the corresponding vic- Microsomes were isolated from the livers of control- and pheno-
DHETs (Figure 11.ly^^, and GSH-conjugatesi^o, barbital-treated rats (10 days; 0.05% w/v phenobarbital in the
drinking water). Values are averages ± SE calculated seven
respectively. The biological role(s) and in vivo different experiments. See ref [71] for experimental details.
540 Jorge H. Capdevila et al.

isoform-specific fashion, the regio- and enantiose- Reconstitution experiments using purified
lectivities of the reaction^' ^' ^^' ^^"^'^' ^^. These prop- P450 isoforms and/or recombinant proteins show
erties of the AA epoxygenase are in contrast with that most AA epoxygenases belong to the CYP 2
those of prostaglandin H2 synthases, where the gene family^^^. The CYP2B and 2C subfamily
known isoforms of the enzyme oxidize AA to the isoforms identified so far as epoxygenases
same single product^' ^. include rat 2B1, 2B2, 2B12, 2C11, 2C23, and
Another distinctive feature of the AA epoxyge- 2C24; rabbit 2B4, 2C1, and 2C2; mouse 2b 19,
nase pathway is the ability of a single P450 isoform 2c37, 2c38, 2c39, and 2c40; and human 2C8,
to epoxidize, stereoselectively, multiple olefins of 2C9/2C10, 2C18, and 2C19 (refs [4H10],
the AA template. For example, purified recombi- [71]-[83], [123]). On the other hand, CYPs 2J2
nant rat kidney CYP2C23 generates 11,12-EET as and 2J4 have also been identified as organ-
its major reaction product (58% of total) but, it is specific epoxygenases and o)-l hydroxylases^^' ^^.
also an efficient AA 8,9-, and 14,15-epoxygenase^^. CYPs lAl, 1A2, and 2E1 are active AA Wco-l
Despite this limited regioselectivity, CYP2C23 is oxygenases, that also produce low and variable
highly stereoselective and forms the corresponding amounts of EETs (<20% of total products)^^'
8(i?),9(*S)-, n(R),l2(S)-, and 14(5), 15(i?)-EETs 80,85 ^ P45Q purified from the livers of dioxin-
enantiomers with optical purities of 94%, 89%, and treated chick embryos has structural features typ-
75%, respectively^'^. On the other hand, the other ical of proteins of the lA gene subfamily, but
two 2C AA epoxygenases expressed in rat kidney, metabolizes AA to EETs as the major reaction
CYPs 2C11 and 2C24, show moderate regioselec- products^^'*. Recent studies characterized 11,12-
tivity for the 11,12- and 14,15-olefins'^^, and epox- EET as an "endothelium-derived hyperpolarizing
idized the 8,9- and 14,15-olefins with opposing factor" (EDHF)'25 and CYP2C34, the porcine
enantiofacial selectivities^^^. Extensive studies with homolog of human CYPs 2C8 and 2C9, as a
a variety of organ purified and/or recombmant coronary artery EDHF synthase'^^. While mem-
epoxygenases, including several CYP 2B and 2C bers of the CYP2C gene subfamily share exten-
isoforms, showed that: (a) with the exception of rat sive sequence homology, this structural homology
CYP2B12, which generates 11,12-EET as nearly is often accompanied by significant catalytic het-
the only reaction product'^'^, most do not catalyze the erogeneity^' ^ For example, CYPs 2C8 and 2C9
selective epoxidation of a single AA olefin to the proteins are —90% homologous in their amino
exclusion of the other three"^' ^, and (b) most mam- acid sequences, yet recombinant CYPs 2C8 and
malian P450 isoforms preferentially epoxidize the 2C9 epoxidize AA with distinct regio- and stereo-
11,12- and 14,15-double bonds'^' ^. The role that sin- chemical selectivities^'.
gle amino acid residues play in the regio- and stere- Comparisons of the regio- and enantioselectiv-
ochemical selectivity of AA epoxidation was ity of the microsomal epoxygenases with that
revealed by replacements introduced into rat and of purified recombinant P450 isoforms, as well
bacterial P450s 2B1 and BM3, respectively^^' ^^. as antibody inhibition experiments indicate that
Recombinant CYP2B1 metabolizes AA to predom- CYPs 2C11 and 2C9, and 2C23 and 2C8 are the
inantly 11,12- and 14,15-EET^^. Replacement of major AA epoxygenases in rat and human liver
isoleucine 114 for alanine in CYP2B1, changed its and kidney, respectively^' ^^' ^'' '^^. Thus, for
regioselectivity toward the preferential epoxidation example, of the three major 2C epoxygenases
of the AA 5,6- and 8,9-olefins^^. On the other hand, expressed in the rat kidney, CYPs 2C11, 2C23,
a single active-site replacement, phenylalanine 78 and 2C24 (ref [123]), only CYP2C23 mimics the
for valine, changed P450 BM3 from a predomi- regio- and stereochemical selectivity of the micro-
nantly AA 18(7^)-hydroxylase into a regio- and somal enzymes^^' '^^. CYP2C23 was shown to be
enantioselective 14(R), 15(iS)-epoxygenase (14(R\ abundantly expressed in rat kidney, and anti-P450
15(5)-EET, >98% of total products)^^. These 2C23 antibodies were selective inhibitors of the
studies indicate that the outcome of the reactions renal microsomal epoxygenase^' ^^^. Furthermore,
catalyzed by many of these highly homologous with the exception of CYPs 2C23 and 2C11,
P450 isoforms is determined by a few amino acid none of the members of the CYP2 gene family
residues, strategically located within the confine- expressed in kidney, including 2A, 2B, 2C, 2E,
ments of what, in most other cases, is known to and 2J isoforms, can account for the degree of
be a rather promiscuous active-site cavity. regio- and stereoselectivity displayed by the rat
Cytochrome P450 and the Metabolism of AA and Eicosanoids 541

renal microsomal epoxygenase^^"^^^. Sequence Moreover, the analysis of the effects of known
comparisons show that the degree of homology P450 inducers on the levels and stereochemistries
between CYP2C23 and the remaining 2C rat of endogenous EETs confirmed the role of P450
proteins is limited, indicating its early evolution- in the in vivo epoxidation of AA^^. These experi-
ary divergence from the other CYP2C proteins. ments documented a new metabolic function for
The regulation of renal CYP2C23 levels by the P450 enzyme system in the oxidation and
dietary salt intake ^^^, and its hormonally control- bioactivation of endogenous fatty acids such as
led expression in the renal microcirculation^^^' ^^^ AA, and demonstrated that the tissue levels and
has suggested important roles for this enzyme in chemical properties of the endogenous EETs
kidney physiology The application of recombi- reflect the organ biosynthetic capacity, as well the
nant DNA methods and heterologous protein contribution of tissue-specific regio- and stereo-
expression should continue to facilitate unequivo- selective EET metabolism"^' ^' n^-iiQ jj^g presence
cal assignments of regio- and enantioselectivities of endogenous chiral EETs has been shown in rat
as well as epoxygenase activities to individual liver, lung, kidney, brain, plasma, and urine; in
P450 isoforms. As more of these recombinant iso- rabbit lung, kidney, and urine; and in human liver,
forms become available, they will be useful in kidney, lung, brain, plasma, and urine"^^^.
defining their: (a) contribution to the epoxidation A distinctive feature of endogenous EET pools
of endogenous AA pools, (b) tissue and/or organ- in rat liver and kidney is their presence as esters of
specific distribution, and (c) regulation by physio- several glycerophospholipids (—99% of the total
logically meaningful stimuli. liver EETs)^^^ with 55% of the total liver
Several powerful in vitro biological activities EETs in phosphatidylcholine, 32% in phos-
have been described for the products of the AA phatidyl-ethanolamine, and 12% in phosphatidyli-
epoxygenases. The EETs"^^^ have been described nositols^^^. Chiral analysis of the fatty acids
as: (a) mediators for the release of several peptide at sn-2 revealed an enantioselective preference
hormones, (b) inhibitors of Na^ reabsorption in the for S(S),9(Ry, 11(5), 12(i?)-, and 14{R), 15(5)-
distal nephron, (c) vasodilators in several microvas- epoxyeicosatrienoates in all three classes of
cular beds and activators of Ca++-dependent vas- phospholipids^ ^^. EET-phospholipid formation
cular K^ channels, mediators of Ca^^ influx in involves a multistep process, initiated by the
several isolated cell systems, powerful mitogens, P450 enantioselective epoxidation of AA, ATP-
and mediators of EGF and Angiotensin II signaling. dependent activation, and enantiomer-selective
lysophospholipid acylation^^^. This EET in vivo
esterification process is unique since most endo-
genously formed eicosanoids are either secreted,
3.3.3. P450 Arachidonic Acid excreted, or further oxidized. Furthermore, these
Epoxygenase: A Member of the studies also show, in contrast to other classes of
eicosanoids, the potential for the rapid, hormon-
Endogenous Arachidonic Acid
ally controlled, generation of preformed bioactive
IVIetabolic Cascade EETs via hydrolytic reactions, thus obviating
In view of their known catalytic versatility, the the need for AA oxidative metabolism. The asym-
in vitro catalysis of AA epoxidation by microso- metric nature of the esterified EETs established
mal P450s was not completely unexpected. It was the existence of novel oxidized glycerolipid pools
therefore apparent that the uniqueness and signif- and demonstrated their enzymatic synthesis from
icance of the P450 AA epoxygenase reaction was endogenous precursors and under normal physio-
going to be defined by whether or not the enzyme logical conditions^ ^^. Greater than 90% of the
system participated in the in vivo metabolism of circulating EETs in rat and human plasma were
the fatty acid. Since asymmetric synthesis is an also found esterified to the phospholipids pre-
accepted requirement for the biosynthetic origin sent in the VLDL, LDL, and HDL lipoprotein
of most eicosanoids, the demonstration of chiral fractions ^^^.
EET pools in several rat and human organs and The biosynthesis of endogenous phospholipids
plasma proved their enzymatic origin and estab- containing esterified EET moieties in several
lished the AA epoxygenase as a formal metabolic human, rat, and rabbit organs suggested a new
pathway and a member of the AA cascade^' ^' ^^. and potentially important functional role for
542 Jorge H. Capdevila et al.

o.
i
? AA-PLs
PLA.
-•AA
P450
P>-' > EETs — p -
ATP, CoA-SH
*• EET-PLs

mm^ , 00©© a

U
Ca^7
Figure 11.3. Regulation of membrane microenvironments by P450 epoxygenase metabolites.

P450 in membrane biology. A few years ago, 3.4. Functional Roles of the
based on studies of the capacity of synthetic P450 Arachidonic Acid
epoxyeicosatrienoyl-phosphochohnes to alter the Monooxygenase
Ca^^ permeability of synthetic liposomes, we
proposed that microsomal P450s could participate Recent reviews provide excellent and detailed
in the real-time control of cellular membrane descriptions of the biological roles attributed to
microenvironments and, hence their functional the products and enzymes of the AA monooxyge-
properties (Figure 11.3)^' ^^^. This proposal nase^ ^^. We address here what we considered to
envisioned as an initial step, the phospholipase- be the two most relevant issues, and for which a
catalyzed release of AA from membrane phospho- more or less generalized consensus exists regard-
lipids, followed by sequential P450-dependent ing their potential physiological importance.
epoxidation, EET activation to the corresponding
acyl-CoA thioesters, and enzymatic lysophospho- 3.4.1. Vascular Reactivity;
lipid acylation to generate the EET-containing
Ion channel regulation
phospholipid pools present in many mammalian
organs. The process could then be termi- Because of their physiological and pathophys-
nated by phospholipase-A2-mediated EET release iological implications, the vasoactive properties of
and enzymatic hydration to the corresponding EETs and 20-HETE are under extensive scrutiny.
DHETs. Under conditions favorable for EET The consensus is that EETs (5,6-and 11,12-EET
acylation we were unable to show the acylation in particular) and 20-HETE are powerful vasodila-
of lysolipids by DHETs ^'^. Many of the enzy- tors and vasoconstrictors of small diameter vascu-
matic steps described above have been character- lar beds, respectively^^^, and that these actions
ized in vitro using either purified proteins or are associated with their ability to inactivate
microsomal membranes^. Inasmuch as it is well (20-HETE) or activate (EETs) Ca++-activated
documented that oxidation of the fatty acid vascular smooth muscle K^ channels^"^^' ^^^' ^^^
moieties present in membrane-bound phospho- It has been proposed that an EET-mediated hyper-
lipids has profound effects on membrane struc- polarization event is required to complete the
tural properties, fluidity, and permeability, vasodilatory response of vascular smooth muscle
the formation and incorporation of EETs into cel- cells to hormones such as, for example,
lular lipids may provide the molecular basis bradykinin^' ^' ^^7, i28 jj^^ identification of 11,12-
underlying some of the EET biological properties, EET as an EDHF^' 1^5, i26^ ^nd of 20-HETE as an
many of which can be attributed to their ability to anti-EDHF molecule^' ^' ^^' ^^^ are supported by the
alter the physicochemical properties of cellular demonstration of hormonally controlled 20-HETE
membranes^"^^. and EET biosynthesis by isolated vascular smooth
Cytochrome P450 and the Metabolism of AA and Eicosanoids 543

muscle and endothelial cells, respectively^"^^' ^25, i3i 20-HETE was proposed based on: (a) P450
By facilitating the introduction of molecular inhibitor studies, and their effects on tubular Na^
and mechanistic approaches to the characteriza- transport^^, (b) differences between DS and
tion of the renal and vascular roles of the P450 DR rats in CYP4A expression and 20-HETE
eicosanoids, these studies are generating concep- synthase activity^^' ^^^, and (c) normalization of
tually a coherent and mechanistic understanding C r transport in DS rats by 20-HETE^O' ^^\
of their ion transport and vasoactive proper- The inhibition of distal nephron Na^ reabsorp-
ties'^"^ ^. The accumulating evidence for a role of tion by 5,6-EET'^' ^^, the induction of kidney
microsomal P450s in vascular biology is creating CYP2C23 and EET biosynthesis by excess dietary
new and important avenues for research, and salt^^' ^^^, and EET-induced dilation of micro-
generating new concepts and experimental circulatory beds'^^ suggested antihypertensive
approaches for the study of cardiovascular and functions for the EETs^' ^. In agreement
renal physiology. with this: (a) clotrimazole inhibition of the rat
kidney epoxygenases caused reductions in renal
EET biosynthesis ^^^, and the development of
3.4.2. Blood Pressure Control and clotrimazole-dependent, salt-sensitive hyperten-
sion^ ^^, (b) high salt diets failed to induce the
Hypertension
activity of the kidney AA epoxygenase in hyper-
An important contribution to the studies of tensive DS rats^^^, and (c) the EETs contribute to
the functional significance of the AA monooxy- the prostanoid- and NO-independent dilation pf
genase was the proposal of a role for renal P450s renal afferent arterioles'^^' ^^^' ^^^. In summary,
in the pathophysiology of experimental hyper- the pro- and antihypertensive roles attributed to
tension^' ^. In addition to its potential clinical the (o-hydroxylase and epoxygenase eicosanoids
relevance, animals models of genetically con- can be rationalized in terms of their biologi-
trolled hypertension afforded the opportunity to cal properties and their site of action^ ^. In the
associate functional phenotypes with alterations renal tubule, EETs and 20-HETE block Na+
in gene structure, function, and/or regulation. transport and function as antihypertensive mole-
From integrations of the renal responses to P450- cules'^^. In the renal vasculature, the EETs
eicosanoids, and correlations between their and 20-HETE have opposing activities as either
biosynthesis and the development of hyperten- powerful vasodilators (EETs) or a vasoconstrictor
sion, pro- and antihypertensive roles were iden- (20-HETE), and can act, therefore, as anti- or
tified for the AAo)/o)-l hydroxylases and prohypertensive mediators, respectively'^^.
epoxygenases^' ^. For example, the developmental Conclusive evidence for a role of P450s in
phase of hypertension, in the Spontaneously renal and cardiovascular physiology was provided
Hypertensive Rat model (SHR model), was by the demonstration that the disruption of the
shown to be accompanied by increases in murine Cyp4al4 gene caused a type of hyperten-
renal CYP4A2 expression and 20-HETE syn- sion that, like most human hypertension, was sex-
thase activity^' ^; and chemical ^^ or antisense ually dimorphic, and more severe in males^^^. As
nucleotide ^^^ inhibition of renal 20-HETE bio- shown in Figure 11.4, lack of a Cyp4al4 gene
synthesis or CYP4A expression, lowered the product(s) raises the mean arterial blood pressure
blood pressure of hypertensive SHR rats. of male Cyp4al4 ( - / - ) mice by 38 mm of Hg,
Importantly, 20-HETE is a powerful vasoconstric- respectively^^^. Hypertensive Cyp4al4 (—/—)
tor of the renal microcirculation^"^^, may mediate mice show increases in plasma androgen
the autoregulatory responses of renal afferent levels ^^^, and in renal 20-HETE synthase activity
arterioles'^^, is formed in situ by CYP4A iso- (Figure 11.4)^^^. Castration reduced kidney micro-
forms'^^' ^^'^, and its vasoactive properties are somal 20-HETE biosynthesis, and normalized the
consistent with its proposed prohypertensive blood pressure of hypertensive Cyp4al4 (—/—)
role'i^. Dahl Salt Sensitive (DS) rats fed high salt mice (Figure 11.4), while, on the other hand,
diets become hypertensive while comparable Dahl androgen administration raised systemic blood
Salt Resistant (DR) animals remain normotensive. pressures and microsomal 20-HETE biosynthesis,
An antihypertensive role for CYP4A2 and regardless of the animal's genotype^^^. Northern
544 Jorge H. Capdevila et al.

Arterial Pressure 20-HETE Synthase


(mm of Hg) (pmol/mg/min)

150 270

125 \- 180

100 90
m
7^

75 -• DiA B C D
Figure 11.4. Genetically controlled or experimentally-induced alterations in the levels of plasma androgens levels
are associated with changes in systemic blood pressure and renal 20-HETE synthase activity. The mean arterial blood
pressure of conscious, wild-type, and Cyp4al4 ( - / - ) adult male mouse, was determined and described^^^.
Androgens were administered by means of implanted 5a-dehydrotestosterone containing pellets (21-day release
pellets; 5 mg/day; Innovative Research of America, Sarasota, PL). After 10 days of treatment, groups of animals were
utilized for either pressure measurements, or the determination of kidney microsomal 20-HETE synthase activity. For
more details see ref. [100]. A, wild-type; B, Cyp4al4 ( - / - ) ; C, castrated Cyp4al4 ( - / - ) ; D, castrated Cyp4al4
(—/—) mice, treated for 10 days with 5-a-dehydrotestosterone. Values show are averages ± SE, calculated from
groups of at least 10 (Arterial Pressure) or 7 (20-HETE synthase) mice.

blot analyses showed that either disruption of the 57 mm of Hg, respectively, and caused parallel
Cyp4al4 gene, or androgen administration caused increases in 20-HETE biosynthesis and in the kid-
the upregulation of Cyp4al2, an active, androgen ney levels of CYP4A8 mRNAs^^?
sensitive, renal 20-HETE synthase'^^, and pro- The characterization of hypertensive Cyp4al4
vided a convenient explanation to paradoxical knockout mice showed that the pressure effects
effects of Cyp4al4 disruption in renal AA w- of this gene were apparently independent of
hydroxylase activity. Based on: (a) the known the intrinsic AA monooxygenase activity of its
hemodynamic effects of 20-HETE^'^ and (b) the encoded protein but, rather were associated with
increased renovascular resistance and impaired changes in the regulation of alternate AA w/co-l
afferent arteriole autoregulatory capacity of hydroxylases (Cyp4al2)^^^. Based on the known
Cyp4al4 ( - / - ) mice^^^, it was proposed that prohypertensive properties of 20-HETE^^^, and
androgen-mediated increases in the renal biosyn- the described gene-dependent, androgen-mediated,
thesis of vasoconstrictor 20-HETE were responsi- control of 20-HETE renal expression and activity,
ble for the hypertensive phenotype of Cyp4al4 we concluded that blood pressure regulation by
(—/—) mice^^^. The prohypertensive effects of kidney CYP4A proteins involves a combination of
androgens were confirmed by administering 5-a- transcriptional and hemodynamic mechanisms that
dehydrotestosterone to male or female Sprague- determine the levels and site of expression of CYP
Dawley rats. In these animals, chronic treatment 4A co-hydroxylases and, ultimately, the level and
with 5-a-dehydrotestosterone raised the systolic site(s) of 20-HETE biosynthesis. Support for this
blood pressure of male and female rats by 29 and conclusion was provided by the demonstration that
Cytochrome P450 and the Metabolism of AA and Eicosanoids 545

dissected rat renal microvessels, the target organ as the precursor for the biosynthesis of several
for most of the prohypertensive effects of 20- physiologically important lipid mediators. The
HETE, possess an androgen-regulated CYP4A8 functional relevance of this metabolic pathway is
20-HETE synthase^^^. suggested by the many important biological activ-
The mechanism(s) by which Cyp4al4 gene ities attributed to its products. These studies, as
product(s) control plasma androgen levels are yet well as the documented endogenous roles of
to be defined; however, ample precedent supports P450s in cholesterol, steroid, and vitamin metabo-
a role for the kidney androgen receptor in regu- lism are contributing to establish this enzyme
lating renal Cyp 4A and 2C expression^^^"^^"^. system as a major participant in the regulation of
Furthermore, in male and female rats, the androgen- cell, organ, and body physiology. Among these,
mediated increases in systemic blood pressure, the phenotypic analysis of mice carrying dis-
and in the levels of kidney CYP4A8 transcripts ^^^, rupted copies of the CYP4al4 gene unveiled new
are accompanied by a marked decrease in the and important roles for the P450 enzymes in car-
levels of renal CYP2C23 epoxygenase protein diovascular physiology and the control of systemic
and diminished microsomal EET biosynthesis^^^. blood pressures, and suggested the human
The androgen-mediated counter-regulation of homologs of the rodent CYP 2C and 4A AA
renal CYP4A w-hydroxylases and 2C epoxy- epoxygenases and ca-hydroxylases as candidate
genases^^^ suggests that its effects on blood genes for the study of their role in the pathophysi-
pressure results from coordinated, nephron site- ology of hypertension, and cardiovascular disease.
specific, increases in the biosynthesis of prohy-
pertensive 20-HETE, and decreases in
antihypertensive EET formation. Finally, in mice, Acknowledgments
activ^ation of PPARa upregulates the expression of
Cyps 4al0 and 4al4 (but not 4al2)i3^ and the
None of the studies carried out in the authors'
counter-regulation of rat Cyps 4A and 2C iso-
laboratory could have been possible without the
forms by PPARa ligands is published^ ^^.
continuous and generous grant support from
Nevertheless, the pressure effects of PPAR ligands
NIGMS and NIDDK.
in mice have yet to be fully defined, although, they
have been characterized as antihypertensive in
rats^^' 140,141 jjjg studies summarized suggest that
blood pressure regulation by renal P450s involves References
combinations of regulatory (transcriptional) and
functional (tubular and hemodynamic) compo- 1. Smith, W.L., D.L. DeWitt, and R.M. Garavito (2000).
nents, and that the organ balance of pro- and anti- Cyclooxygenases: Structural, cellular, and molecular
hypertensive mediators and thus, its functional biology. Ann. Rev. Biochem. 69, 145-182, and cited
status, is dictated by: (a) the nephron segment- references.
specific expression and regulation of the corre- 2. Prescott, S.M. and C. Yang (2002). Many actions of
sponding genes, (b) the enzymatic properties of cyclooxygenase-2 in cellular dynamics and in
Cancer. J. Cell. Biol. 190, 279-286.
the encoded proteins, and (c) the expression and
3. Brash, A.R. (1999). Lipoxygenases: Occurrence,
activities of ancillary enzymes responsible for function, catalysis, and acquisition substrate. J. Biol.
EET and/or HETE metabolism, disposition or Chem. 214, 23679-23680, and cited references.
activation. 4. Capdevila, J.H. and J.R. Falck (2001). The CYP P450
arachidonic acid monooxygenases: From cell signal-
ing to blood pressure regulation. Biochem. Biophys.
4. Conclusion Res. Comm. 285, 571-576, and cited references.
5. Capdevila, J.H. and J.R. Falck (2002). Biochemical
and molecular properties of the cytochrome P450
The studies of the P450 AA monooxygenase
arachidonic acid monooxygenases. Prostaglandins
have uncovered new and important roles for P450 other Lipid Mediat. 68-69, 325-344, and cited
in the metabolism of endogenous substrates, and references.
added P450 to the list of enzymes that participate 6. McGiff, J.C. and J. Quilley (1999). 20-HETE and
in the metabolism of AA, a fatty acid that serves the kidney: Resolution of old problems and new
546 Jorge H. Capdevila et al.

beginnings. Am. J. Physiol. 277, R607-R623, and kidney cDNAs encoding lauric acid-hydroxylases.
cited references. Biochemistry 29, 873-879.
7. McGiff, J.C. and J. Quilley (2001). 20-Hydroxy- 20. Kikuta, Y., E. Kusunose, and M. Kusunose (2002).
eicosatetraenoic acid and epoxyeicosatrienoic acids Prostaglandin and leukotriene w-hydroxylases.
and blood pressure. Curr. Opin. Nephrol. Hypertens. Prostaglandins other Lipid Mediat. 68-69,
10, 31-237, and cited references. 325-344, and cited references.
8. Imig, J.D. (2000). Eicosanoid regulation of renal 21. Rahimtula, A.D. and PJ. O'Brien (1974).
vasculature. Am. J. Physiol. 279, F965-F981, and Hydroperoxide catalyzed liver microsomal aromatic
cited references. hydroxylation reactions involving cytochrome P-450.
9. Campbell, W.B. and D.R. Harder (1999). Biochem. Biophys. Res. Commun. 60, 440-447.
Endothelium-derived hyperpolarizing factors and 22. Weiss, R.H, XL. Arnold, and R.W. Estabrook
vascular cytochrome P450 metabolites of arachi- (1987). Transformation of an arachidonic acid
donic acid in the regulation of tone. Circ. Res. 84, hydroperoxide into epoxyhydroxy and trihydroxy
484^88, and cited references. fatty acids by liver microsomal cytochrome P-450.
10. Roman, R.J. (2002). P450 metabolites of arachi- Arch. Biochem. Biophys. 252, 334-338.
donic acid in the control of cardiovascular function. 23. Hecker, M. and V Ullrich (1989). On the mecha-
Physiol. Rev. 82, 131-185, and cited references. nism of prostacyclin and thromboxane A2 biosyn-
11. Capdevila, J.H., N. Chacos, J. Werringloer, thesis. ^ Biol. Chem. 264, 141-150.
R.A. Prough, and R.W. Estabrook (1981). Liver 24. Hara, S,, A. Miyata, C. Yokoyama, R. Brugger,
microsomal cytochrome P-450 and the oxidative F Lottspeich, V Ullrich et al. (1995). Molecular
metabolism of AA. Proc. Natl. Acad. Sci. USA 78, cloning and expression of prostacyclin synthase
5362-5366. from endothelial cells. Adv. Prostaglandin
12. Morrison, A.R. and N. Pascoe (1981). Metabolism of Thromboxane Leukot. Res. 23, 121-123.
AA through NADPH-dependent oxygenase of renal 25. Yokoyama, C , A. Miyata, H. Ihara, V Ullrich, and
cortex. Proc. Natl. Acad. Sci. USA 78, 7375-7378. T. Tanabe (1991). Molecular cloning of the human
13. Oliw, E.H. and J.A. Gates (1981). Oxygenation of platelet thromboxane A synthase. Biochem.
AA by hepatic microsomes of the rabbit. Biophys. Res. Commun. 178, 1479-1484.
Mechanism of biosynthesis of two vicinal diols. 26. Ohashi, K., K.H. Ruan, R.J. Kulmacz, K.K. Wu, and
Biochim. Biophys. Acta 666, 327-340. L.H. Wang (1992). Primary structure of human
14. Jacobson, H.R., S. Corona, J.H. Capdevila, thromboxane synthase determined from the cDNA
N. Chacos, S. Manna, A. Womack et al. (1984). sequence. J. Biol. Chem. 261, 789-793
Effects of epoxyeicosatrienoic acids on ion trans- 27. White, R.E. and M.G. Coon (1980). Oxygen activa-
port in the rabbit cortical collecting tubule. In tion by cytochrome P-450. Ann. Rev. Biochem. 49,
P. Braquet, J.C. Frolich, S. Nicosia, and R. Garay 315-356, and references therein.
(eds), Prostaglandins and Membrane Ion Transport. 28. Wong, PY.K., K.U. Malik, B.M. Taylor,
Raven Press, New York, p. 311. WP Schneider, J.C. McGiff, and FE Sun (1985).
15. Karara, A., E. Dishman, 1. Blair, J.R. Falck, and Epoxidation of prostacyclin in the rabbit kidney.
J.H. Capdevila (1989). Endogenous epoxye- J. Biol. Chem. 260, 9150-9153.
icosatrienoic acids. Cytochrome P-450 controlled 29. Oliw, E.H. (1984). Metabohsm of 5(6)-epoxy-
stereoselectivity of the hepatic arachidonic acid eicosatrienoic acid by ram seminal vesicles forma-
epoxygenase. ^ ^/o/. Chem. 264, 19822-19827. tion of novel prostaglandin Ej metabolites. Biochim.
16. Kupfer, D. (1982). Endogenous substrates of Biophys. Acta. 793, 408^15.
monooxygenases: Fatty acids and prostaglandins. In 30. Zhang, J.Y, C. Prakash, K. Yamashita, and LA. Blair
J.B. Schenkman and D. Kupfer (eds), Hepatic (1992). Regiospecific and enantioselective meta-
Cytochrome P-450 Monooxygenase System. bolism of 8,9-epoxyeicosatrienoic acids by cyclooxy-
Pergamon Press, New York, pp. 157-187. genase. Biochem. Biophys. Res. Commun. 183,
17. Sharma, R.K., M.V Doig, D.FV Lewis, and 138-143.
G. Gibson (1989). Role of hepatic and renal 31. Jajoo, H.K., J.H. Capdevila, J.R. Falck, R.K. Bhatt,
cytochrome P450 IVAl in the metabolism of lipid and LA. Blair (1992). Metabolism of 12(R)-hydroxy-
substrates. Biochem. Pharmacol. 38, 3621-3629. eicosatetraenoic acid by rat liver microsomes.
18. Imaoka, S., S. Tanaka, and Y. Funae (1989). Biochim. Biophys. Acta 1123, 110-116.
(w/w-l) hydroxylation of lauric acid and arachi- 32. Capdevila, J., P. Mosset, P. Yadagiri, Sun Lumin, and
donic acid by rat renal cytochrome P-450. Biochem. J.R. Falck (1988). NADPH-Dependent microsomal
Int. 18,731-740. metabolism of 14,15-epoxyeicosatrienoic acid to
19. Johnson, E.E, D.L. Walker, and K.J. Griffin diepoxides and epoxyalcohols. Arch. Biochem.
(1990). Cloning and expression of three rabbit Biophys. 261, 122-132.
Cytochrome P450 and the Metabolism of AA and Eicosanoids 547

33. Cowart, L.A., S. Wei, M.-S. Hsu, E.F. Johnson, 44. Lasker, J.M., B.W. Chen, I. Wolf, B.P Blswck,
M.U Krishna, J.R. Falck et al. (2002). The CYP 4A RD. Wilson, and RK. Powell (2000). Formation of
isoforms hydroxylate epoxyeicosatrienoic acids to 20-hydroxyeicosatetraenoic acid, a vasoactive and
form high affinity peroxisome proliferator-activated natriuretic eicosanoid in human kidney. J. Biol.
receptor Hgands. J. Biol. Chem. Ill, 35105-35112. Chem. 264, 17845-17853.
34. Hamberg, M. and B. Samuelsson (1966). 45. An updated list of P450 genes and sequences is found
Prostaglandins in human seminal plasma. at: http://dmelson.utmem.edu/CytochromeP450.html
Prostaglandins and related factors. J. Biol Chem. 46. Imaoka, S., H. Ogawa, S. Kimura, and F.J. Gonzalez
241, 257-263. (1993). Complete cDNA sequence and cDNA-
35. Israelson, U, M. Hamberg, and B. Samuelsson directed expression of CYP4A11, a fatty acid
(1969). Biosynthesis of 19-hydroxy-prostaglandin omega-hydroxylase expressed in human kidney.
A,. Eur. J. Biochem. 11, 390-394. DNA Cell Biol. 12, 893-899.
36. Kupfer, D., I. Jansson, L.V Favreau, 47. Kawashima, H., T. Naganuma, E. Kusunose, T. Kono,
A.D. Theoharides, and J.B. Schenkman (1988). R. Yasumoto, K. Sugimura et al. (2000). Human fatty
Regioselective hydroxylation of prostaglandins by acid (o-hydroxylase, CYP 4A11: Determination of
constitutive forms of cytochrome P-450 from rat liver: complete genomic sequence and characterization of
Formation of a novel metabolite by a female-speci- purified recombinant protein. Arch. Biochem.
fic P-450. Arch. Biochem. Biophys. 262, 186-195. Biophys. 378, 333-339.
37. Matsubara, S., S. Yamamoto, K. Sogawa, 48. Bellamine, A., Y. Wang, M.R. Waterman, J.V Gainer,
N. Yokotani, Y. Fujii-Kuriyama, M. Haniu et al. E. Dawson, N.J. Brown et al. (2003). Characteri-
(1987). cDNA cloning and inducible expression zation of the CYP4A11 gene, a second CYP4A gene
during pregnancy of the MRNA for rabbit pul- in humans. Arch. Biochem. Biophys. 409, 221-227.
monary prostaglandin oa-hydroxylase (cytochrome 49. Wang, M.H., D.E. Stec, M. Balazy, V Mastyugin,
P-450p-2). J. Biol. Chem. 62, 13366-13371. C.S. Yang, R.J. Roman et al. (1997). Cloning,
38. Sharma, R.K., M.V Doig, D.F.V Lewis, and sequencing, and cDNA-directed expression of rat
G. Gibson (1989). Role of hepatic and renal renal CYP4A2: AA w-hydroxylation and 11,12-
cytochrome P450 IVAl in the metabolism of lipid epoxidation by CYP4A2 protein. Arch. Biochem.
substrates. Biochem. Pharmacol. 38, 3621-3629. Biophys. 336, 240-250.
39. Yokotani, N., R. Bernhardt, K. Sogawa, 50. Helvig, C , E. Dishman, and J.H. Capdevila (1998).
E. Kusunose, O. Gotoh, M. Kusunose et al. (1989). Molecular, enzymatic, and regulatory characteriza-
Two forms of w-hydroxylase toward prostaglandin tion of rat kidney cytochromes P450 4A2 and 4A3.
A and laurate. cDNA cloning and their expression. Biochemistry'M, 12546-12558.
J. Biol. Chem. 264, 21665-21669. 51. Hoch, U, Z. Zhang, D.L. Kroetz, and PR. Ortiz de
40. Kusonuse, E., A. Sawamura, H. Kawashima, Montellano (2000). Structural determination of the
I. Kubota, and M. Kusunose (1989). Isolation of a substrate specificities and regioselectivities of the
new form of cytochrome P450 with prostaglandin A rat and human fatty acid omega hydroxylases. Arch.
and fatty acid w-hydroxylase activities from rabbit Biochem. Biophys. 373, 63—71.
kidney cortex microsomes. J. Biochem. 106, 52. Nishimoto, M., J.E. Clark, and B.S.S. Masters
194-206. (1993). Cytochrome P450 4A4: Expression in
41. Kikuta, Y, E. Kusunose, T. Okumoto, I. Kubota, and Escherichia coli, purification, and characteri-
M. Kusunose (1990). Purification and partial char- zation of catalytic properties. Biochemistry 32,
acterization of two forms of cytochrome P450 with 8863-8870.
co-hydroxylase activity towards prostaglandin A and 53. Bylund, J., M. Bylund, and E.H. Oliw (2001).
fatty acids from rat liver microsomes. J. Biochem. cDNA cloning and expression of CYP4F12, a novel
107,280-286 human cytochrome P450. Biochem. Biophys. Res.
42. Roman, L.J., C.A.N. Palmer, J.E. Clark, Commun. 280, 892-897.
A.S. Muerhoff, K.J. Griffin, E.F. Johnson et al. 54. Yokomizo, T., T. Izumi, and T. Shimizu (2001).
(1993). Expression of rabbit cytochrome P4504A Leukotriene B4: Metabolism and signal transduc-
which catalyzes the w-hydroxylation of fatty acids, tion. Arch. Biochem. Biophys. 385, 231-241, and
and prostaglandins. Arch. Biochem. Biophys. 307, cited references.
57-67. 55. Haeggstrom, J.Z. and A. Wetterholm (2002).
43. Bylund, J., M. Hidestrand, M. Ingelman-Sundberg, Enzymes and receptors in the leukotriene cascade.
and E.H. Oliw (2000). Identification of CYP4F8 Cell. Mol. Life Sci. 59, 742-753, and cited references.
in human seminal vesicles as a prominent 19- 56. Powell, WS. (1984). Properties of leukotriene B4
hydroxylase of prostaglandin endoperoxides. 20-hydroxylase from polymorphonuclear leuko-
J. Biol. Chem. 275, 21844-21849. cytes. J. Biol. Chem. 259, 3082-3089.
548 Jorge H. Capdevila et al.

57. Shak, S. and I.M. Goldstein (1984). Oxidation is the icosanoid synthesized by neutrophils from
major pathway for the catabolism of leukotriene B4 125-hydroxyeicosatetraenoic acid produced by
in human polymorphonuclear leukocytes. J. Biol. thrombin- or collagen-stimulated platelets. Proc.
Chem. 259, 10181-10187. Natl. Acad. Sci. USA 81, 903-907.
58. Romano, M.C., R.D. Eckardt, RE. Bender, 69. Flaherty, J.T and J. Nishihira (1987). 5-Hydroxyei-
T.B. Leonard, K.M. Straub, and J.K Newton (1987). cosatetraenoate promotes Ca^^ and protein kinase C
Biochemical characterization of hepatic microsomal mobilization in neutrophils. Biochem. Biophys. Res.
leukotriene B^ hydroxylases. J. Biol Chem. 262, Commun. 148, 575-579.
1590-1595. 70. Okita, R.T., R.J. Soberman, J.M. Bergholte, B.S.S.
59. Soberman, R.J., R.T. Okita, B. Fitzsimmons, Masters, R. Hayes, and R.C. Murphy (1987).
J. Rokach, B. Spur, and K.F. Austen (1987). (o-Hydroxylation of 15-hydroxyeicosatetraenoic
Stereochemical requirements for substrate specificity acid by lung microsomes from pregnant rabbits.
of LTB4 20-hydroxylase. J. Biol. Chem. 262, 12421. Mol. Pharmacol. 32, 706-709.
60. Sumimoto, J., K. Takeshige, and S. Minakami 71. Capdevila, J.H., A. Karara, D.J. Waxman,
(1988). Characterization of human neutrophil M.V Martin, J.R. Falck, and FP Guengerich (1990).
leukotriene B4 omega-hydroxylase, a system Cytochrome P-450 enzyme-specific control of the
involving a unique cytochrome P-450 and NADPH- regio- and enantiofacial selectivity of the micro-
cytochrome P-450 reductase. Eur. J. Biochem. 172, somal AA epoxygenase. J. Biol. Chem. 265,
315-324. 10865-10871.
61. Kikuta, Y., E. Kusunose, K. Endo, S. Yamamoto, 72. Knickle, L.C. and J.R. Bend (1994). Bioactivation
K. Sogawa, Y. Fujii-Kuriyama et al. (1993). A novel of arachidonic acid by the cytochrome P450
form of cytochrome P450 family 4 in human poly- monooxygense of guinea pig lung: The orthologue
morphonuclear leukocytes. cDNA cloning and of cytochrome P450 2B4 is solely responsible for
expression of leukotriene B^ co-hydroxylase. J. Biol. the formation of epoxyeicosatrienoic acids. Mol.
Chem. 268, 9376-9380. Pharmacol. 45, 1273-1280.
62. Kikuta, Y, E. Kusunose, H. Sumimoto, Y Mizukami, 73. Laethem, R.M., J.R. Halpert, and D.R. Koop (1994).
K. Takeshige, T. Sakai et al. (1998). Purification and Epoxidation of arachidonic acid as an active site
characterization of recombinant human neutrophil probe of cytochrome P450 2B isoforms. Biochim.
leukotriene B^ w-hydroxylase (Cytochrome 4F3). Biophys. Acta 1206, 4 2 ^ 8 .
Arch. Biochem. Biophys. 355, 201-205. 74. Keeney, D.S., C. Skinner, S. Wei, T. Friedberg, and
63. Christmas, P, S.R. Ursino, J.W. Fox, and M.R. Waterman (1998). A keratinocyte-specific
R.J. Soberman (1999). Expression of the CYP4F3 epoxygenase, CYP2B12, metabolizes arachidonic
gene: Tissue-specific splicing and alternative promot- acid with unusual selectivity, producing a single
ers generate high and low Km forms of leukotriene major epoxyeicosatrienoic acid. J. Biol. Chem. 273,
B4 w-hydroxylase. J. Biol. Chem. 214, 21191-21199. 9279-9284.
64. Kikuta, Y, E. Kusunose, M. Ito, and M. Kusunose 75. Keeney, D.S., C. Skinner, J.B. Travers, J.H. Capdevila,
(1999). Purification and characterization of recom- J.B. Nanney L.E. King et al. (1998). Differentiating
binant rat hepatic 4F1. Arch. Biochem. Biophys. keratinocytes express a novel cytochrome P450
369, 193-196. enzyme, CYP2B19, having arachidonate monooxyge-
65. Jin, R., D.R. Koop, J.L. Raucy, and J.M. Lasker nase activity. J. Biol. Chem. 112,, 32071-32079.
(1998). Role of human CYP4F2 in hepatic catabo- 76. Capdevila, J.H., S. Wei, Y Yan, A. Karara, H.R.
lism of the proinflammatory agent, leukotriene B^. Jacobson, J.R. Falck et al. (1992). Cytochrome P-
Arch. Biochem. Biophys. 359, 89-98. 450 arachidonic acid epoxygenase. Regulatory con-
66. Kawashima, H., E. Kusunose, CM. Thompson, and trol of the renal epoxygenase by dietary salt loading.
H.W. Strobel (1997). Protein expression, characteri- J. Biol. Chem. 267, 21720-21726.
zation, and regulation of CYP4F4 and CYP4F5 77. Karara, A., K. Makita, H.R. Jacobson, J.R. Falck,
cloned from rat brain. Arch. Biochem. Biophys. 347, FP Guengerich, R.N. DuBois et al. (1993).
148-154. Molecular cloning, expression, and enzymatic char-
67. Wong, PYK., P Westlund, M. Hamberg, acterization of the rat kidney cytochrome P-450
E. Granstrom, P.W.H. Chao, and B. Samuelsson arachidonic acid epoxygenase. J. Biol. Chem. 268,
(1984). (o-Hydroxylation of 12-L-hydroxy-5,8,10,14- 13565-13570.
eicosatetraenoic acid in human polymorphonuclear 78. Imaoka, S., P.J. Zwedlung, H. Ogawa, S. Kimura,
leukocytes. J. Biol Chem. 259, 2683-2686. FJ. Gonzalez, and H.Y Kim (1993). Identification
68. Marcus, A.J., L.B. Safier, H.L. Ullman, of CYP2C23 expressed in rat kidney as an arachi-
N. Broekman, T. Islam, D. Oglesby et al. (1984). donic acid epoxygenase. J. Pharmacol. Exp. Then
lis, 20-Dihydroxyeicosatetraenoic acid: A new 267, 1012-1016.
Cytochrome P450 and the Metabolism of AA and Eicosanoids 549

79. Daikh, B.E., R.M. Laethem, and D. Koop (1994). hydroxyeicosatetraenoic acids (HETEs) formed
Stereoselective epoxidation of arachidonic acid by during catalytic oxygenation of AA by microsomal
cytochrome P-450s 2CAA and 2C2. J. Pharmacol. cytochrome P-450. Biochem. Biophys. Res.
Exp. Then 269, 1130-1135. Commun. 141, 1007-1011.
80. Rifkind, A.B., C. Lee, T.K. Chang, and D.J. Waxman 91. WooUard, PM. (1986). Stereochemical differen-
(1995). Arachidonic acid metabolisms by human ces between 12-hydroxy-5,8,10,14-eicosatetraenoic
cytochrome P450s 2C8, 2C9, 2E1, and 1A2: acid in platelets and psoriatic lesions. Biochem.
Regioselective oxygenation and evidence for Biophys. Res. Commun. 136, 169-176.
a role for CYP2C enzymes in arachidonic acid 92. Boeglin, W.E., R.B. Kim, and A.R. Brash (1998).
epoxygenation in human liver microsomes. Arch. A 12R-lipoxygenase in human skin: Mechanistic
Biochem. Biophys. 320, 380-389. evidence, molecular cloning, and expression. Proc.
81. Zeldin, D.C., R.N. DuBois, J.R. Falck, and Natl. Acad. Sci. USA 95, 6744-6749.
J.H. Capdevila (1995). Molecular cloning, expres- 93. Sun D., M. McDonnel, X.S. Chen, M.M. Lakkis,
sion and characterization of an endogenous human S.N. Isaacs, S.H. Elsea etal. (1998). Human 12(R)-
cytochrome P450 arachidonic acid epoxygenase lipoxygenase and the mouse ortholog. Molecular
isoform. Arch. Biochem. Biophys. 322, 76-86. cloning, expression, and gene chromosomal assign-
82. Luo, G., D.C. Zeldin, J.A. Blaisdell, E. Hodgson, ment. J. Biol. Chem. 273, 33540-33547.
and J.A. Goldstein (1998). Cloning, and expression 94. Schwartzman, M.L., M. Balazy, J. Masferrer,
of murine CYP2Cs and their ability to metabolize N.G. Abraham, J.C. McGiff, and R.C. Murphy
arachidonic acid. Arch. Biochem. Biophys. 357, (1987). 12(i?)-Hydroxyeicosatetraenoic acid: A
45-57. cytochrome P450-dependent arachidonate metabo-
83. Tsao, C.C, S.J. Coulter, A. Chien, G. Luo, lite that inhibits Na^,K+-ATPase in the cornea.
N.R Clayton, R. Maronpot et al (2001). Identifica- Proc. Natl. Acad. Sci. USA 84, 8125-8129.
tion and localization of five CYP2Cs in murine 95. Oliw, E.H. (1993). Biosynthesis of 12(S)-hydrox-
extrahepatic tissues and their metabolism of arachi- yeicosatetraenoic acid by bovine corneal epithe-
donic acid to regio- and stereoselective products. lium, ^c^a P/i^^w/. Scand. 147, 117-121.
J. Pharmacol. Exp. Ther 299, 3 9 ^ 7 . 96. Murphy, R.C, J.R. Falck, S. Lumin, P Yadagiri,
84. Thompson, CM., J.H. Capdevila, and H.W. Strobel J.A. ZirroUi, M. Balazy et al (1988). \2{Ry
(2000). Recombinant cytochrome P450 2D 18 Hydroxyeicosatrienoic acid: A vasodilator cyto-
metabolism of dopamine and arachidonic acid. chrome P-450-dependent arachidonate metabolite
J. Pharmacol. Exp. Ther 294, 1120-1130. from bovine corneal epithelium. J. Biol. Chem. 263,
85. Laethem, R.M., M. Balazy, J.R. Falck, C.L. Laethem, 17197-17202.
and D.R. Koop (1993). Formation of 19(S)-, 19(R)-, 97. Lu, A.Y., H.K. Junk, and M.J. Coon (1969).
and 18(R)-hydroxyeicosatetraenoic acids by alcohol- Resolution of the cytochrome P-450 containing w-
inducible cytochrome P450 2E1. J Biol. Chem. 268, hydroxylation system of liver microsomes into
12912-12918. three components. J. Biol. Chem. 244, 3714-3721.
86. Scarborough, RE., J. Ma, W. Qy, and D.C. Zeldin 98. Capdevila, J.H., S. Wei, C Helvig, J.R. Falck,
(1999). P450 subfamily CYP2J and their role in the Y. Belosludtsev, G. Truan et al. (1996). The highly
bioactivation of arachidonic acid in extrahepatic stereoselective oxidation of polyunsaturated fatty
tissues. Drug Metab. Rev. 31, 205-234, and cited acids by cytochrome P450BM-3. J. Biol. Chem.
references. Ill, 22663-22671
87. Zeldin, D.C. (2001). Epoxygenase pathways of 99. Graham-Lorence, S., G. Truan, J.A. Peterson,
arachidonic acid metabolism. J. Biol. Chem. 216, J.R. Falck, S. Wei, and C Helvig et al. (1997). An
36059-36062, and cited references. active site substitution, F87V, converts cytochrome
88. Oliw, E.H. (1993). bis-AWylic hydroxylation of P450 BM3 into a regio- and stereoselective
linoleic acid and AA by human hepatic monooxyge- (14)S,15(R)-AA epoxygenase. J. Biol. Chem. 272,
nases. Biochim. Biophys. Acta 1166, 258-263. 1127-1135.
89. Brash, A.R., W.E. Boeglin, J.H. Capdevila, S. Yeola, 100. Holla, VR., E Adas, J.D. Imig, X. Zhao, E. Price,
and LA. Blair (1995). 7-HETE, 10-HETE, and 13- N. Olsen et al. (2001). Alterations in the regulation
HETE are major products of NADPH-dependent of androgen-sensitive Cyp 4a monooxygenases
AA metabolism in rat liver microsomes: Analysis of cause hypertension. Proc. Natl. Acad. Sci. USA 98,
their stereochemistry, and the stereochemistry of 5211-5216.
their acid-catalyzed rearrangement. Arch. Biochem. 101. Henderson, C.J., A.R. Scott, C S . Yang, and
Biophys. 321, 485^92. CR. Wolf (1990). Testosterone-mediated regula-
90. Capdevila, J.H., P. Yadagiri, S. Manna, and tion of mouse renal cytochrome P-450 isoen-
J.R. Falck (1986). Absolute configuration of the zymes. Biochem. J. 266, 675-681.
550 Jorge H. Capdevila et al.

102. Lund, J., P.G. Zaphiropoulos, A. Mode, M. Warner, 114. Marji, IS., M.H. Wang, and M. Laniado-
and J.A. Gustafsson (1991). Hormonal regulation Schwartzman (2002). Cytochrome P-450 4A
of cytochrome P450 gene expression. Adv. in isoform expression and 20-HETE synthesis in
Pharmacol. 22, 325-354, and cited references. renal preglomerular arteries. Am. J. Physiol. 283,
103. Henderson, C.J. and C.R. Wolf (1991). Evi- F60-F67.
dence that the androgen receptor mediates sexual 115. Hashimoto, T., T. Fujita, N. Usuda, W Cook, C. Qi,
differentiation of mouse renal cytochrome P450 J.M. Peters et al. (1999). Peroxisomal and
expression. Biochem. J. 278, 499-503. mitochondrial fatty acid oxidation in mice nullizy-
104. Sundseth, S.S. and D.J. Waxman (1992). Sex- gous for both peroxisome proliferator-activated
dependent expression and clofibrate inducibility receptor and peroxisomal fatty acyl-CoA oxidase.
of cytochrome P450 AA fatty acid w-hydroxylases. Genotype correlate with fatty liver phenotype.
J. Biol. Chem. 267(6), 3915-3921. J. Biol. Chem. 274, 19228-19236.
105. Lee, S.S., T. Pineau, J. Frago, E.J. Lee, J.W. Owens, 116. Chacos, N., J.R Falck, C. Wixtrom, and
D.L. Kroetz et al. (1995). Targeted disruption J. Capdevila (1982). Novel epoxides formed during
of the a isoform of the peroxisome proliferator- the liver cytochrome P-450 oxidation of AA.
activated receptor gene in mice results in abolish- Biochem. Biophys. Res. Commun. 104, 916-922.
ment of the pleiotropic effects of peroxisomal 117. Karara, A., E. Dishman, J.R. Falck, and
proHferators. Mol. Cell. Biol. 15, 3012-3022. J.H. Capdevila (1991). Endogenous epoxyeicosa-
106. Waxman, D.J. andT.K.H. Chang (1995). Hormonal trienoyl-phospholipids. A novel class of cellular
regulation of liver cytochrome P450 enzymes. In glycerolipids containing epoxidized arachidonate
PR. Ortiz de Montellano (ed.). Cytochrome P450: moieties. ^ 5/o/. Chem. 266, 7561-7569.
Structure, Mechanism, and Biochemistry, 2nd edn. 118. Fang, X., T.L. Kaduce, M. VanRollins,
Plenum Press, New York, pp. 391^18. N.L. Weintraub, and A.A. Spector (2000).
107. Waxman, D.J. (1999). P450 gene induction by Conversion of epoxyeicosatrienoic acids (EETs) to
structurally diverse xenochemicals: Central role of chain-shortened epoxy fatty acids by human skin
nuclear receptors CAR, PXR, and PPAR. Arch. fibroblasts. J. Lipid Res. 41, 66-74.
Biochem. Biophys. 369, 11-23. 119. Zeldin, D C , J. Kobayashi, J.R. Falck, B.S. Winder,
108. Savas, U, M-H. Hsu, and E.R Johnson (2003). B.D. Hammock, J.R. Snapper et al. (1993).
Differential regulation of human CYP4A genes by Regio- and enantiofacial selectivity of epoxye-
peroxisome prohferators and dexamethasone. icosatrienoic acid hydration by cytosolic epoxide
Arch. Biochem. Biophys. 409, 212-220. hydrolase. J. Biol. Chem. 268, 6402-6407.
109. Stromsteadt, M., S. Hayashi, P.G. Zaphiropoulos, 120. Spearman, M.E., R.A. Prough, R.W Estabrook,
and J.A. Gustafsson (1990). Cloning and charac- J.R. Falck, S. Manna, K.C. Leibman et al. (1985).
terization of a novel member of the cytochrome Novel glutathione conjugates formed from epoxye-
P450 subfamily IVA in rat prostate. DNA Cell Biol. icosatrienoic acids (EETs). Arch. Biochem.
8, 569-577. Biophys. 242, 225-230.
110. Heng, Y.M., C.S. Kuo, PS. Jones, R. Savory, 121. Yu, Z., F Xu, L.M. Huse, C. Morisseau,
R.M. Schultz, S.R. Tomlinson et al. (1997). A novel A.J. Draper, J.W Newman et al. (2000). Soluble
murine P-450 gene, Cyp4al4, is part of a cluster of epoxide hydrolase regulates hydrolysis of vasoactive
Cyp4a and Cyp4b, but not of CYP4F, genes in epoxyeicosatrienoic acids. Circ. Res. 87, 992-998.
mouse and humans. Biochem. J. 325, 741-749. 122. Sinai, C.J., M. Miyata, M. Tohkin, K. Nagata,
111. Falck, J.R., S. Lumin, I.A. Blair, E. Dishman, J.R. Bend, and F.J. Gonazalez (2000). Targeted dis-
M.V Martin, D.J. Waxman et al. (1990). ruption of soluble epoxide hydrolase reveals a role
Cytochrome P-450-dependent oxidation of arachi- in blood pressure regulation. J. Biol. Chem. 275,
donic acid to 16-, 17-, and 18-hydroxyeicosate- 40504-40510.
traenoic acids. J. Biol. Chem. 265, 10244-10249. 123. Holla, V, K. Makita, P.G. Zaphiropoulos, and
112. Rifkind, A.B., A. Kanetoshi, J. Orlinick, J.H. Capdevila (1999). The kidney cytochrome
J.H. Capdevila, and C. Lee (1994). Purification P450 2C23 AA epoxygenase is upregulated during
and biochemical characterization of two major dietary salt loading. J. Clin. Invest. 104, 751-760
cytochrome P-450 isoforms induced by 2,3,7,8- 124. Rifkind, A.B., A. Kanetoshi, J. Orlinick,
tetrachlorodibenzo-/7-dioxin in chick embryo liver. J.H. Capdevila, and C. Lee (1994). Purification
J. Biol. Chem. 269, 3387-3396. and biochemical characterization of two major
113. Qu, W, J.A. Bradbury, C-C. Tsao, R. Maronpot, cytochrome P-450 isoforms induced by 2,3,7,8-
G.J. Harry, C.E. Parker et al. (2001). Cytochrome tetrachlorodibenzo-/7-dioxin in chick embryo liver.
P450 CYP2J9, a new mouse arachidonic acid co-1 J. Biol. Chem. 269, 3387-3396.
hydroxylase predominantly expressed in brain. 125. Campbell, W.B., D. Gebremedhin, PF Pratt, and
J. Biol. Chem. 276(27), 25467-25479. D.R. Harder (1996). Identification of epoxye-
Cytochrome P450 and the Metabolism of AA and Eicosanoids 551

icosatrienoic acids as endothelium-derived hyper- CYP4A1 antisense oligonucleotide reduces


polarizing factors. Cir. Res. 78, 415^23. mesenteric vascular reactivity and blood pressure
126. Fisslthaler, B., R. Popp, L. Kiss, M. Potente, in SHR. Am. J. Physiol. 280, 255-261.
D.R. Harder, I. Fleming et al. (1999). Cytochrome 134. Ma, Y.H., M.L. Schwartzman, and R.J. Roman
P450 2C is an EDHF synthase in coronary arteries. (1994). Altered renal P-450 metabolism of
Nature 410, 493-497. arachidonic acid in dahl salt-sensitive rats. Am. J.
127. Imig, J.D., J.R. Falck, S. Wei, and J.H. Capdevila Physiol. 267, R579-R589.
(2001). Epoxygenase metabolites contribute to 135. Zhou, A.P, H.A. Drummond, and R.J. Roman
nitric oxide-independent afferent arteriolar vasodi- (1996). Role of 20-HETE in elevating loop chlo-
lation in response to bradykinin. J. Vase. Res. 38, ride reabsorption in Dahl SS/Jr. rats. Hypertension
247-255. 27,631-635.
128. Imig, J.D., X. Zhao, J.R. Falck, S. Wei, and 136. Makita, K., K. Takahashi, A. Karara, H.R. Jacobson,
J.H. Capdevila (2001). Enhanced renal microvascular J.R. Falck, and J.H. Capdevila (1994). Experimental
reactivity to angiotensin II in hypertension is amelio- and/or genetically controlled alterations of the renal
rated by the sulfonimide analog of 11,12-epoxye- microsomal cytochrome P450 epoxygenase induce
icosatrienoic acid. J. Hypertension 19, 983-992. hypertension in rats fed a high salt diet. J. Clin.
129. Karara, A., S. Wei, S.D. Spady, L. Swift, Invest. 94, 2414-2420.
J.H. Capdevila, and J.R. Falck (1992). Arachidonic 137. Nakagawa, K., J.S. Marji, M.L. Schwartzman,
acid epoxygenase: Structural characterization and M.R. Waterman, and J.H. Capdevila (2003). The
quantification of epoxyeicosatrienoates in plasma. androgen-mediated induction of kidney arachidonate
Biochem. Biophys. Res. Commun. 182, 1320-1325. hydroxylases is associated with the development of
130. Capdevila, J.H., Y. Jin, A. Karara, and J.R. Falck hypertension. Am. J. Physiol. 284, R1055-R1062.
(1993). Cytochrome P450 epoxygenase depen- 138. Kroetz, D.L., P Yook, P Costet, P Bianchi, and
dent formation of novel endogenous epoxye- T. Pineau (1998). Peroxisome proliferator-
icosatrienoyl-phospholipids. In S. Nigam, activated receptor a controls the hepatic CYP4A
L.J. Marnett, K.V Honn, andT.L. Walden Jr. (eds), induction adaptative response to starvation and
Eicosanoids and Other Bioactive Lipids in Cancer, diabetes. J. Biol. Chem. 273, 31581-31589.
Inflamation and Radiation Injury. Kluwer 139. Corton, J.C, L-Q. Fan, S. Brown, S.P Anderson,
Academic Publishers, Boston, MA, pp. 11-15. C. Bocos, and R.C. Cattley et al. (1998).
131. Zou, A.P, J.T. Fleming, J.R. Falck, E.R. Jacobs, Down-regulation of cytochrome P450 2C family
D. Gebremedhin, and D.R. Harder et al. (1996). members and positive acute-phase response gene
20-HETE is an endogenous inhibitor of the large- expression by peroxisomal proliferator chemicals.
conductance Ca^^-activated K+ channel in renal Mol. Pharmacol. 54, 463^73.
arterioles. Am. J. Physiol. 270, R228-R237. 140. Shatara, R.K., D.W. Quest, and T.W Wilson
132. Su, P, K.M. Kaushal, and D.L. Kroetz (1998). (2000). Fenofibrate lowers blood pressure in two
Inhibition of renal arachidonic acid omega- genetic models of hypertension. Can. J. Physiol.
hydroxylase activity with ABT reduces blood Pharmacol. IS, 361-311.
pressure in the SHR. Am. J. Physiol. 215, 141. Roman, R.J., Y.H. Ma, B. Frohlich, and
R426-R438. B. Markham (1993). Clofibrate prevents the devel-
133. Wang, M.H., E Zhang, J. Marji, B.A. Zand, opment of hypertension in Dahl salt-sensitive rats.
A. Nasjletti, and M. Laniado-Schwartzman (2001). Hypertension 21, 985-988.
12
Cytochrome P450s in Plants
Kirsten Annette Nielsen and Birger Lindberg Meller

1. Introduction alkaloids. In addition, a number of plant P450s


have been shown to catalyze detoxification of
Plants are sessile organisms that cannot avoid harmfiil agents including herbicides'^.
exposure to adverse climatic conditions or attack
from herbivores and pests by escaping. To survive 1.2. Chemical Warfare
and protect themselves, they are dependent on the
The chemical warfare between plants and her-
ability to (a) redirect their overall metabolism to
bivores and pests is complex and takes place at
meet environmental constraints, (b) construct
many trophic levels. The plant Apium graveolens
physical barriers that are difficult to penetrate,
(celery) is known to combat Helicoverpa zea (com
(c) produce chemicals that make the plant toxic to
earworm) by producing allelochemicals including
pests and herbivores, and (d) communicate with
fiiranocoumarins in a P450-dependent series of
the environment, for example, to attract pollinators.
reactions^. The herbivore, however, is able to
Cytochrome P450 enzymes (P450s) play a key role
detoxify the friranocoumarins. The detoxification
in enabling plants to achieve these main goals.
pathway is induced by jasmonate, a wound-induced
plant signal compound. Jasmonate activates the
1.1. Natural Products transcription at least of four herbivore P450 genes^.
The continuous chemical warfare between plants
Plants are the best organic chemists in nature as on one side and herbivores and pests on the other
evidenced by their ability to synthesize all neces- may enforce plants to constantly evolve new natu-
sary carbon compounds with carbon dioxide as the ral products and insects to find means to detoxify
sole carbon source and by their ability to synthe- these. This requires recruitment of enzymes with
size a vast number of natural products. Currently, altered biological functions probably mediated by
structures for more than 100,000 different natural modifications and duplications of existing genes.
products isolated from plants are known^~^, and P450s are key players in securing recruitment of
with time this number will increase into millions. these new functions as exemplified by the recruit-
Natural products are classified as phytoanticipins, ment of an allele specific P450 in Drosophila to
phytoalexins, and/or attractants. In the last decade, acquire resistance to chemical insecticides^.
the majority of the biosynthetic pathways responsi-
ble for natural product synthesis have been shown
1.3. Chemical Communication
to include P450s as key enzymes. Such pathways
include the biosynthetic pathways for cyanogenic Plants are dependent on intense communi-
glucosides, glucosinolates, isoflavonoids, and cation with their surroundings via biochemical

Kirsten Annette Nielsen and Birger Lindberg IVIoller • Plant Biochemistry Laboratory, Royal Veterinary
and Agricultural University, 40, Thorvaldsensvej, DK-1871 Frederiksberg C, Copenhagen, Denmark.
Cytochrome P450: Structure, Mechanism, and Biochemistry, 3e, edited by Paul R. Ortiz de Montellano
Kluwer Academic / Plenum Publishers, New York, 2005.

553
554 Kirsten A. Nielsen and Birger L. Moller

signalling, for example, to mediate pollination and and isoflavonoids. Special emphasis is on the
seed spreading ^ In cases with insect-mediated plant model Arabidopsis thaliana for which the
pollination, plants synthesize insect attractants. A complete genome sequence is available'^ and
special group of secondary metabolites, glucosi- which is easily amenable to genetical modifica-
nolates, is produced within the taxonomic order tions'^ and provides an excellent model plant for
Capparales. Glucosinolates have dual roles acting metabolic engineering. Exploitation of P450s to
both as attractants for specialized insects and as reach increased production levels for desired
deterrents for generalist herbivores. P450s are key natural compounds and transfer of entire biosyn-
enzymes in the biosynthesis of glucosinolates^' ^. thetic pathways into other plant species will be
Nicotiana tabacum (tobacco) from the taxonomic discussed. The first part is a short presentation of
order Solanes produces cembranoid-type terpenes different tools used to achieve gene—^to function
as insect attractants. The cembranoid terpene gland relationship, the bottleneck in P450 functional
exudates contain a- and p-epimers of cembra- genomics
2,7,1 l-triene-4,6-diol and these attract different
pollinating insects. Unfortunately, they also
enhance oviposition of the unwanted insect Myzus 2. The P450 Superfamily
nicotiana (red aphid)^^.
in Plants
1.4. Medicinal Agents So far genomic and expressed sequence tag
Humans take advantage of plant natural prod- (EST) sequencing projects have revealed a total of
ucts as drugs or lead compounds in medicine, for 1,059 plant P450 sequences'^. Phylogenetic
example, as anesthetics and anticarcinogens. analyses based on translated raw DNA sequence
Alkaloid-containing plants have been used in data have spaced the P450s into 10 clans that
human medicine for thousands of years. One very include 59 families and an extensive number of
large and structurally diverse group of alkaloids subfamilies'^' ^^. In the A. thaliana genome alone,
are the tetrahydrobenzylisoquinoline alkaloids. a superfamily of 272 cytochrome P450 genes
Papaver somniferum (opium poppy) from the tax- including 26 pseudo genes were annotated and
onomic order Ranunculales produces more than named'^' ^'. These genes represent members of
100 such L-tyrosine derived alkaloids including 45 out of the 59 currently assigned plant P450
the potent anesthetic morphine"' '^. The biosyn- families^^' ^^. P450s constitute the largest and con-
thesis of tetrahydrobenzylisoquinoline alkaloids tinuously expanding superfamily in plants. From
involves P450s with unique catalytic properties'^. the Oryza sativa cvs japonica and indica (rice
cultivars)^^' '^^ genome sequence projects, as many
The nutraceuticals daidzein and genistein
as 458 predicted P450 genes were annotated by
belong to the isoflavonoids and are phytoestrogens
the end of September 2002^^.
preventing breast and prostate cancers'"*. The
dietary compounds are synthesized especially
in Leguminosae belonging to the Fabales order. 2.1. Nomenclature
Isoflavonoid biosynthesis also requires a unique
P450 enzyme that catalyzes aryl group migra- The large number of P450 enzymes found in
tion^^. Alkaloids and isoflavonoids play important the Plant Kingdom are named and categorized
roles in plant defense and their biosynthesis are based on protein sequence identity and phyloge-
tightly regulated and inducible processes^' \ netic relationships^^. P450s assigned to the same
The purpose of this review is to highlight family share more than 40% sequence identity at
recent key findings on plant P450s. The genome the amino acid level. Correspondingly, P450s
sequencing programs have identified the P450s as assigned to the same subfamily share more than
the largest superfamily in plants. The catalytic 55% sequence identity.
properties of most of these P450s remain elusive. In plants, the identity rule has some exceptions
We focus on P450s involved in the synthesis of due to gene duplications and shuffling as
natural products belonging to the groups of pointed out in Werck-Reichart et al (2002)^2. The
cyanogenic glucosides, glucosinolates, alkaloids. plant P450s are categorized into the following
Cytochrome P450s in Plants 555

families: CYP51, CYP71-99, CYP701-727, and CYP79F2, methionine TV-hydroxylases^^-^^;


CYP73620. The CYP51 family is unique because CYP83A1 and CYP83B1 enzymes converting
the sequence identity of the P450s belonging to indole-3-acetaldoxime, p-hydroxyphenylacetal-
this family is well-enough conserved across phyla doxime, and phenylacetaldoxime into the
to contain plant, fungal, bacterial as well as animal corresponding ^S-alky l-thiohydroximates"^^"^"^;
sequences^^. The precise structure of the sterols CYP84A1, ferulic acid hydroxylase^^; CYP85A1,
that serve as substrates for CYPSls varies among steroid C-6-hydroxylase^^; CYP86A1 and
different eucaryotes. This variability has been sug- CYP86A8, fatty acid G5-hydroxylases^^' 4^;
gested to represent adaptation to the availability of CYP88A3 and CYP88A4 enzymes converting ent-
different sterol precursors in different Kingdoms. kaurenoic acid to GA^/^; CYP90A1, steroid C-23
Plant P450s are membrane-bound proteins. hydroxylase^^; CYP90B1, steroid C-22 hydroxy-
They are classified into the A-type and the non-A- lase^^; CYP98A3, 3'-hydroxylase of phenolic
type P450s^^' ^^. It has been proposed that the esters^^; and CYP701A3, ^w^kaurene oxidase^^.
plant-specific A-type P450s originate fi*om a sin-
gle ancestral P450^^. A-type P450s share a simple
3.1. Phylogenetic Relationships
gene organization with a single phase 0 intron
with a highly conversed position^^. Typically, the The categorization of P450s from different
P450 genes are found to cluster with close rela- plant species into families and subfamilies based
tives on short stretches of all five^. thaliana chro- on sequence identity and phylogenetic relation-
mosomes indicating recent duplication events^^. ships as discussed above typically does not con-
P450s involved in the biosynthesis of plant natural comitantly lead to an assignment of biological
products belong to the A-type. In this review, we and/or enzymatic function. In some families, the
focus on A-type P450s belonging to the CYP71, P450s all appear to catalyze the same enzymatic
CYP79, CYP80, CYP83, and CYP93 families and reaction. In other families, members of the same
their respective involvement in the biosynthesis of family clearly catalyze very different enzymatic
cyanogenic glucosides, glucosinolates, alkaloids, reactions. These differences are illustrated with
and isoflavonoids. A short description of the the following examples. The CYP73 family is
biological function of non-A-type plant P450s is composed of one subfamily, CYP73A with 37
confined to members of the CYP85 and CYP90 members. CYP73As from Helianthus tuberus
families that are involved in the production of (artichoke)^"^, Phaseolus aureus (mung bean)^^,
polyhydroxylated steroidal molecules. Medicago sativa (alfalfa)^^, Petroselium crispum
(parsley)^^, Popupus tremuloides (querken
aspen)^^' ^^, A. thaliana^^, Triticum aestivum
(wheat)^^, Cicer arietinum (chickpea)^ ^ have all
3. Tools Available to Identify been demonstrated to be cinnamate 4-hydroxy-
Biological Functions lases. The rest of the members of the CYP73A
subfamily are therefore with great confidence
Only very few of the 246 predicted assigned as cinnamate 4-hydroxylases solely
P450 enzymes present in A. thaliana have had a based on their amino acid sequence identity.
biological function assigned. Functional assign- Members of the CYP74A subfamily have been
ments of the A. thaliana P450s are restricted to characterized as allene oxide synthases in
23 enzymes belonging to 14 of the 45 plant fami- A. thaliana^^' ^^, Linum usitatissium (flaxseed)^^,
lies represented in the A. thaliana genome. These Hordeum vulgare (barley)^^, and Lycopersicon
identified enzymatic activities are: CYP51, esculentum (tomato)^"^. However, members of the
obtusifoliol 14a-demethylase^^; C7P725i,brassi- closely related CYP74B subfamily possess fatty
nolide 26-hydroxylase^^; CYP73A5, cinnamate-4- acid hydroperoxide lyase activity as demonstrated
hydroxylase^^; CYP74A1, allene oxide synthase^^'^'^; in A. thaliana and L. esculentum (tomato)^^' ^^.
CYP74B2, hydroperoxide lyase^^; CYP75B1, Members of a single subfamily may also catalyze
flavonoid 3'-hydroxylase^^; CYP79A2, phenyl- different and consecutive steps in a biosynthetic
alanine iV-hydroxylase^^; CYP79B2 and CYP79B3, pathway as reported for members of CYP90A and
tryptophan A/-hydroxylases^^; CYP79F1 and CYP90B (see Section 4.1). The different steps of
556 Kirsten A. Nielsen and Birger L. Moller

entire biosynthetic pathways may be mediated by generated by either T-DNA insertion^^"^^ ethyl
P450s belonging to the same subfamily as exem- methanesulphonate (EMS) mutagenesis^^' ^'^, or
plified by the CYP71C subfamily (see Section ionizing radiation'''^. Special attention in screening
5.3.2). In contrast to the latter examples, members programmes has been paid toward phenotypic
of five different subfamilies of the CYP79 family mutants showing aberrant growth characteristics.
are all A/-hydroxylases (see Section 5.2). A final This way, non-A-type P450s was shown to affect,
example on the existence of numerous subfamilies for example, dwarfism^*^' ^^ (Section 4) and A-type
with widely different biological functions is the 18 P450s to affect excessive lateral root formation"^^
subfamilies CYP71A to CYP71R in the CYP71 (Section 7). Genetic analysis of phenotypes recog-
family^^. Enzymatic activities have solely been nized by a lack of blue-green autofluorescence
demonstrated for members of subfamilies caused by absence of sinapoyl malate identified
CYP71C, CYP71D, and CYP71E as described additional members of A-type P450s. Sinapoyl
in detail in Section 5.1.1. One member of a malate is a phenylpropanoid that serves as a
fourth subfamily, the CYP71A10 was shown to biochemical sunscreen^^ (Section 7).
possess enhanced detoxifying properties against Methodologies to provide gain-of-function
phenylurea-derived herbicides, an activity in mutants in existing knockout collections use
unlikely to be the major biological fiinction of the activation tagging in weak-mutant-allelic back-
enzyme^^. Of the 110 known members of the grounds^^. This facilitates identification of domi-
CYP71 family, 97 belong to the subfamilies nant suppressor genes, which will show enhanced
CYP71A to CYP71D. No catalytic function has expression after incorporation of multimeric,
been assigned to any of the 37 members of the positive c/5-acting elements close to suppressor
CYP71B subfamily genes. Using activation tagging, it was found that
The difficulties in assigning function to a P450 expression levels of a gene encoding a non-A-type
solely based on its amino acid sequence will be P450 proved to influence regulation of light
partly alleviated as more catalytic functions responsiveness and accumulation of steroid phyto-
become known and diagnostic sequence elements hormones^' (Section 4.1).
identified. The matter is particularly complicated
for the A-type P450s involved in natural product
synthesis. Plants are known to produce more than
100,000 different natural products with P450s 3.3. Reverse Genetics
involved in most pathways and sometimes being Reverse genetics provides a tool to identify the
multifunctional^^"^^. To illustrate the preponder- mutant genotype causing specific phenotypic
ance of A-type P450s, they account for 153 out of characteristics. Using T-DNA tagged phenotypic
the predicted 246 P450 genes in the A. thaliana mutants (Section 3.2), genomic DNA sequences
genome.^^ In general, an A-type P450 is thought flanking the T-DNA integration site are identified.
to possess high substrate specificity and its func- Subsequently, the wild-type allele is identified
tion to be limited to a single or a few parallel and cloned and inserted into the mutant to revert
biosynthetic pathway(s). its phenotype into wild type. Catalytic properties
The wide diversity in amino acid sequences of the P450 are thereafter studied by heterologous
found among the P450s is evident by the fact that expression of the plant cDNA in microorganisms.
in A. thaliana, P450s'^' ^o belong to 45 of the 59
plant P450 families.

3.4. Heterologous Expression in


3.2. Mutant Collections in Microorganisms
A. thaliana
Heterologous expression of individual cDNAs
The biological function of some A. thaliana in Escherichia coli followed by enzyme assays in
P450s have been elucidated in planta by taking the presence of putative substrates have been
advantage of the availability of knockout mutants used extensively for characterization of plant
in this model plant. Mutant collections have been P450s^^, for example, for the CYP79s involved in
Cytochrome P450s in Plants 557

cyanogenic glucoside and glucosinolate synthe- 3.6. Homology-Based Cloning


sis^' ^^. Recombinant P450 protein can be subjected
to classical protein characterization including CO The CYP79A1 cDNA sequence^^ has been
difference spectroscopy^^ and recording of substrate- used to design degenerate DNA oligonucleotide
binding spectra^^ and finally assayed for desired primer sequences for identification of homolo-
catalytic properties. The first plant P450 cDNA was gous genes in other cyanogenic crops like
isolated from ripening fruits of Persea americana Manihot esculenta (cassava) using polymerase
(avocado)^ ^ It was designated CYP71A1. Expres- chain reactions (PCR). Cassava was found to
sion of the cDNA in Saccharomyces cerevisia^^ express two P450 isoforms belonging to the
yielded high amounts of recombinant protein, but CYP79 family They showed 53% and 54%
the predicted catalytic property of CYP71A1 was amino acid sequence identity, respectively, to
not identified^^. CYP79A1^^. Because the sequence identity to the
Cinnamic acid 4-hydroxylase from Helianthus CYP79A1 is below 55%, the two cassava homo-
tuberosus (Jerusalem artichoke) was the first plant logues established a new subfamily and were
P450 to be fimctionally characterized^'*. CYP73A1 named CYP79D1 and CYP79D2. The two iso-
was designated as the first member of the CYP73 forms exhibit 85% sequence identity and the
family. This cDNA was isolated from an expres- recombinant proteins catalyze the same biochem-
sion library using antibodies raised against the iso- ical reaction (Sections 5.2). A similar PCR
lated P450 protein (Section 3.5). Cinnamic acid strategy served to identify additional CYP79
4-hydroxylase catalyzes an essential step in the homologues from Triglochin maritima (seaside
phenylpropanoid pathway and it is considered to arrowgrass)^^.
be ubiquitous in plants (see Section 3.1). Based on known cinnamate 4-hydroxylase
sequences from Jerusalem artichoke and mung
bean^"^' ^^, a homology search in an EST library
identified an EST clone with 84-86% sequence
3.5. Isolation of Enzymes identity, which was then used as a probe to isolate
Cinnamate 4-hydroxylases catalyze the the CYP73A5 from a genomic library^^.
hydroxylation of ^ra«5-cinnamic acid into trans- To identify and clone cDNAs encoding
/7-coumaric acid. The ability to monitor this inducible P450s involved in the biosynthesis of
enzyme activity in Jerusalem artichoke allowed tetrahydrobenzylisoquinoline alkaloids, a PCR
isolation of the P450 enzyme CYP73A1 using strategy based on the conserved sequence ele-
conventional chromatography and generation of ments in the haem-binding domain of A-type
specific antibodies^"^' ^^. P450s was applied^ ^' ^^. Based on mRNA isolated
A general isolation procedure based on dye from induced, tetrahydrobenzylisoquinoline alka-
affinity chromatography has been developed loid producing plant tissue, 17 different P450
and has been used to isolate CYP79A1 that con- sequences were found. The sequences were com-
verts L-tyrosine into /7-hydroxyphenylacetal- pared with existing sequence data and heterologous
doxime^^. This A^-hydroxylase catalyzes the expression assays based on predicted enzymatic
first committed step in the production of the activities that identified two alleles of (S)-
cyanogenic glucoside dhurrin in Sorghum bicolor. iV-methylcoclaurine 3'-hydroxylase^^ (Section 6).
Isolated CYP79A1 was catalytically active as
demonstrated by its ability to convert tyrosine
into /?-hydroxyphenylacetaldoxime when recon- 4. Non-A-Type P450s Mediating
stituted in artificial liposomes in the presence Steroid Biosynthesis
of NADPH-cytochrome P450 oxidoreductase,
NADPH, and molecular oxygen^^. Based on Like vertebrates and fungi, plants produce
partial amino acid sequencing, the corresponding polyhydroxylated steroidal hormones to regulate
cDNA sequence was cloned from expression and control tissue morphology. In plants these
libraries of sorghum seedlings and subsequently types of hormones are designated brassino-
used to produce recombinant protein^^' ^^ (see steroids^^"^^ and they are built on a campestanol
Section 5.1.2). carbon skeleton (Figure 12.1). The brassinosteroids
558 Kirsten A. Nielsen and Birger L. Moller

are nonessential phytohormones with impact on suggested^^^~^^^. Non-A-type plant P450s partici-
morphological characteristics, for example, leaf pate in the production of the plant sterols that are
shape and dwarfism^^' ^^ Biological functions of brassinosteroid precursors. A key enzyme is
brassinosteroids are controlled by specific recep- obtusifoliol 14a-demethylase. This belongs to the
tors and suppressors (see Figure 12.2). These CYP51 family and gene sequences encoding this
mediate signal transduction and control regulation ubiquitous plant enzyme that has been obtained
of target genes including those for brassinosteroid from S. bicolor (sorghum)^^^' ^^^ and T. aestivum
biosynthesis^^' ^^. Brassinosteroids may potentiate (wheat)^^^' ^^^. Recently, an orthologue of obtusi-
plant fitness and defense in response to pathogen foliol 14a-demethylase was identified in
attack, since brassinosteroids induce systemic A. thaliana based on its ability to complement a
defense responses in tobacco and rice^^. lanosterol 14a-demethylase mutant of yeast^^.
The biosynthetic pathway for brassinosteroids The expression level of another gene CYP72B1,
has not yet been fully elucidated. Models as also assigned as BASl belonging to the A-type
presented in figure 12.3 for two parallel path- family and encoding a brassinolide 26-hydroxy-
ways assigned as "the early C-6 oxidation" and lase, has been shown to regulate light perception
"the late C-6-oxidation" pathways have been and control accumulation of brassinosteroids^ ^
Two non-A-type P450 families with known enzy-
matic activities, CYP90 and CYP85, participating
in brassinosteroid biosynthesis are selected as rep-
resentatives for detailed description (Sections 4.1
and 4.2).

4.1. CYP90S
CYP90A1, also assigned as constitutive
photomorphogenesis and dwarfism (cpd) from
A. thaliana encodes an enzyme in steroid biosyn-
4 ^ 6
thesis that catalyzes hydroxylation (Figure 12.3) of
Figure 12.1. Polyhydroxylated steroids in plants cathasterone to testarone and of 6-deoxycatha-
indicating the carbon numbers of brassinosteroids as sterone to 6-deoxytestarone^^. CYP90A1 was the
reprinted with permission from Bishop and Yokota first plant P450 identified by reverse genetics
(2001)94. using a morphological screen for aberrant growth

Figure 12.2. A model of the regulatory machinery for brassinosteroid sensing and biosynthesis. Upon perception
of brassinosteroids, the receptor BRII signals via a phosphorylation cascade to regulate gene expression and cell
expansion. Reprinted with permission from Thummel and Chory (2002)^^.
Cytochrome P450s in Plants 559

(24fl)'24-IIMItyl-
CllOlMt«4<«lV2^-Oi

$-Qxoc«nipe«imMH
($-OxoCN)

Catliast&rone

r
(CT)

r
I
•I I
1 !
" H (6-D«»xo30T>

€8
t
Ha*

e-Peoxoodftliisterone $a4lyiiroxycast8Slei0ii«
(6-DeoxoCS) (6<OHC$)

Bfassinoltite

Figure 12.3. The proposed late and the early C-6 oxidation pathways for biosynthesis of brassinolides as outlined
and reprinted with permission from Nogushi et al. (2000)^^2.
560 Kirsten A. Nielsen and Birger L. Meller

characteristics among a collection of T-DNA to 3-dehydroteasterone, from 6-deoxotyphasterol


tagged mutants^^. CYP90A1 shared 24% amino to typhasterol, and from 6-deoxocastasterone to
acid sequence identity to the rat testosterone- castasterone'^' ^^^. The enzymatic activity of
16a-hydroxylase, CYP2B1^^^. Transcription of CYP85 enables crosstalk between "the early C-6
CYP90A1 is negatively controlled by brassino- oxidation" and "the late C-6-oxidation" pathways
steroids^^' ^^^, most likely as part of a regulatory for brassinosteroid formation and transforms the
mechanism to ensure optimal physiological levels two pathways into a metabolic grid (Figure 12.3).
of endogenous brassinosteroids during growth Transcriptional activity of the gene is negatively
(see Figure 12.2). The ability of CYP90A1 to regulated by brassinosteroids^^.
hydroxylate the steroid side chain of both cathas-
terone and 6-deoxycathasterone illustrates that
steps in "the early C-6 oxidation" and "the late C-
6-oxidation" pathways may be mediated by the 5. A-Type P450s Mediating
same enzyme. Homologues categorized into the Plant Protection
CYP90A family have subsequently been identi-
fied in Saccharum sp. (sugar cane) and in Vigna Plants need to defend and protect themselves
radiata (mung bean)-^^. CYP90A encoding genes against attack from herbivores and microorgan-
are expected to be ubiquitous in the Plant isms. Toward this goal, plants produce a vast array
Kingdom. of natural products some of which mediate broad
A second dwarfed phenotype, dwarf 4 (dwf4), resistance toward herbivores and pests and some
in A. thaliana was also characterized using reverse of which are highly specific. Accordingly, the
genetics^ ^ The mutation affects a 22-a-hydroxy- ability of plants to produce natural products
lase, assigned as CYP90B1, that hydroxylates the enhances plant fitness by efficiently counteracting
brassinosteroid side chain (Figure 12.3). The enzy- otherwise damaging biotic and abiotic stresses.
matic activity of CYP90B1 provides the substrate
of CYP90A1. CYP90B1 shares 40% amino acid
sequence identity with CYP90A1 and, like 5.1. Broad Defense: Cyanogenic
CYP90A1, functions in "the early C-6 oxidation" Glucosides
as well and in "the late C-6-oxidation" path-
ways'^. Although the A. thaliana CYP90B1 is cur- Cyanogenesis is the ability of plants to release
rently the only member of this subfamily, it is hydrogen cyanide upon tissue damage.
thought to be ubiquitous in plants. Two additional Cyanogenesis is an old trait widely distributed
subfamilies CYP90C and CYP90D have been in the Plant Kingdom^^' 110-112 ^nd currently
established each containing one gene from documented in more than 2,650 plant species^ ^^.
A. thaliana^^. The enzymatic activities of these Cyanogenesis is mediated by cleavage of cyano-
P450s remain to be elucidated. genic glucosides into the corresponding cyanohy-
drin and glucose by the action of p-glucosidase.
Subsequent cleavage of the cyanohydrin into
a ketone or aldehyde and hydrogen cyanide pro-
4.2. CYP85S
ceeds catalyzed by an a-hydroxy-nitrilase or non-
The CYP85 family is also involved in enzymatically. Cyanogenic glucosides belong to
brassinosteroid biosynthesis. cDNA sequences the class of natural products known as phyto-
encoding enzymes belonging to this non-A-type anticipins. They are also present in healthy plant
family has been obtained from A. thaliana and tissues anticipating and ready to combat pathogen
Solanum lycopersicon (potato). Members of attack. Cyanogenic glucosides are present in many
the CYP85 family share approximately 35% important crop plants like barley, sorghum, and
identity to those of CYP90^^. Recombinant ver- cassava^ ^^.
sions of the two CYP85 plant genes were The release of poisonous hydrogen cyanide
expressed in yeast and both enzymes were shown upon tissue disruption may render the presence
to catalyze multiple steps from 6-deoxoteasterone of cyanogenic glucosides in a crop plant, a nutri-
to teasterone, from 3-dehydro-6-deoxoteasterone tional problem. This is of special concern in
Cytochrome P450s in Plants 561

cassava where use of this crop as a staple food COOH

requires careful processing to remove the


cyanogenic glucosides or their degradation prod- L-tyrosine
ucts before consumption^ ^'^. In barley, major focus
02 + NADPH
has been on cyanide potential in malt (5-day-old
seedlings) and breeding programs have estab- ic NADP+
COOH
lished genotypes assigned as low, medium, and
high producers^ ^^. Despite domestication and con- HO
OH
trolled breeding, null-mutants have neither been Af-hydroxytyro8ine
identified in cassava nor in barley. It has been
I / 02 + NADPH
hypothesized that the ability of humans to remove 1 ^ NADP+
cyanide by food processing explains why humans
have continued to select and use cyanogenic crops
as important components in the diet. In the early " HO OH

phases of plant breeding, selection of cyanogenic MA^dihydroxytyrosine

crops may have afforded protection from herbi-


vore damage and may have helped to prevent theft
of the crop^^^.
Cyanogenic glucosides are derived from the
amino acids L-valine, L-isoleucine, L-leucine, {£)-/>-hydroxyphenyl-
acetaidoxime
L-phenylalanine, and L-t3a*osine and from the non-
protein amino acid cyclopentenyl glycine^^' ^^^.
Typically, a cyanogenic plant contains only one or
two different cyanogenic glucosides. The biosyn-
thetic pathway for cyanogenic glucosides has been
elucidated using dhurrin production in S. bicolor
xrx"
(2)-/>'hydroxyphenyl-
acetaldoxlme
as a model system. A general scheme for biosyn-
thesis of cyanogenic glucosides involving two
membrane-bound P450s and a soluble UDPG-
glucosyltransferase was established as described
below (see Figure 12.4). XT'p*hydroxyphenyi - P450OX
acetonttriie

1^02 + NADPH

5.1.1. Biosynthesis |^NADP+

Initial studies on the biosynthetic pathway for


the L-tyrosine-derived cyanogenic glucoside dhur-
rin demonstrated that the covalent bond linking
p-hydroxymandetonitrile
the a-and p-carbon atoms in L-tyrosine was
preserved throughout dhurrin synthesis^ ^^.
Subsequently, it was shown that the C-N bond in
the parent amino acid is preserved during the
biosynthetic process^ ^^. These studies were car-
CHO
ried out by administration of double-labeled tyro- 1] +HCN
sine to excised, biosynthetically active sorghum
seedlings. Based on these observations, a biosyn- />hydroxybenzaldehyde

thetic pathway including an aldoxime, a nitrile,


Figure 12.4. The biosynthetic pathway for
and an a-hydroxynitrile as intermediates was the cyanogenic glucoside dhurrin is catalyzed
proposed, although no such compounds were by two multifunctional cytochrome P450s,
detectable^ ^^' ^^^. A major breakthrough in the elu- CYP79A1, and CYP71E1 (P450ox) and by a
cidation of the dhurrin pathway was based on the glucosyltransferase, UGT85B1.
562 Kirsten A. Nielsen and Birger L. Moller

isolation of a biosynthetically active microsomal of an oxime to the corresponding nitrile, which


system. Upon administration of NADPH, molecu- subsequently is C-hydroxylated to the cyanohydrin
lar oxygen, and radiolabeled L-tyrosine to this (Figure 12.4)^^^. The nitrile intermediate in the
experimental system in the presence of a large CYP71E1 catalyzed reaction was demonstrated
surplus of unlabeled putative intermediates, it was using trapping experiments ^^^' ^^^. A single
possible to trap radiolabeled A^-hydroxytyrosine, oxygen molecule is consumed in the CYP71E1
(i^-/>-hydroxyphenylacetaldoxime, (Z)-/?-hydroxy- catalyzed reaction sequence^^^' ^^'^.
phenylacetaldoxime, and /7-hydroxymandelo- The last step in cyanogenic glucoside synthesis
nitriiei2i, 122 involves conversion of a cyanohydrin into the cor-
The enzymes responsible for cyanogenic responding cyanogenic glucoside. Using dye-
glucoside synthesis have been characterized using column affinity chromatography, a soluble
the sorghum microsomal system as biological UDP-glucose:/>-hydroxymandelonitrile-0-gluco-
starting material^^' ^^^. The conversion of the syltransferase, designated UGT85B1'^^, was iso-
parent amino acid L-tyrosine into the correspon- lated from etiolated sorghum seedlings and shown
ding (Z)-aldoxime is catalyzed by CYP79A1 to glucosylate the cyanohydrin function of/?-hydro-
(Figure 12.4). This multiftinctional P450 monooxy- xymandelonitrile to produce dhurrin (Figure 12.4).
genase constitutes the first identified member of Reconstitution of CYP79A1 and CYP71E1 into
the CYP79 family and catalyzes two consecutive artificial liposomes in the presence of UGT85B1
iV-hydroxylations, a decarboxylation and a dehy- resulted in the formation of dhurrin, that is, in
dration reaction. Isolated CYP79A1 was success- reconstitution of the entire pathway for dhurrin
fully reconstituted into artificial liposomes also production from its parent amino acid tyrosine ^^^
containing isolated NADPH cytochrome P450 (Figure 12.5).
oxidoreductase^^. Administration of radiolabeled cDNA sequences encoding CYP79A1,
tyrosine to this reconstituted enzyme system CYP71E1, and UGT85B1 have been isolated^^'
in the presence of putative intermediates as 125, 128 ^^^ functionally active proteins were
unlabeled compounds permitted identification of obtained by heterologous expression of each of
7V-hydroxytyrosine, 7V,A^-dihydrotyrosine, and (£")- the cDNA clones in E. coli. The entire pathway
/?-hydroxyphenylacetaldoxime as intermediates in for dhurrin synthesis has been transferred to
the conversion of tyrosine to the (Z)-aldoxime. A. thaliana^^"^, 2i plant species that in nature does
From stoichiometric analyses, it was shown not possess the ability to produce cyanogenic
that two molecules of oxygen are consumed in glucosides. Sequential introduction of each of the
this conversion. Enzyme assays carried out in three enzymes into A. thaliana demonstrated that
an^^02 atmosphere using either tyrosine or dhurrin is produced only after coordinated expres-
A^-hydroxytyrosine as substrates demonstrated sion of all three sorghum genes'^^. Importantly,
that the two oxygen atoms introduced in the expression of UGT85B1 proved obligatory
A^-hydroxylation steps are enzymatically distin- despite the availability in the A. thaliana genome
guishable as demonstrated by specific loss of the of 120 family 1 glycosyl transferase genes^^' ^^^.
oxygen atom introduced by the first A/-hydroxyla- In transgenic plants co-expressing CYP79A1 and
tion reaction in the subsequent conversion of CYP71EU'^', /7-hydroxymandelonitrile is the
A'^A^-dihydroxytyrosine into the (Z)-aldoxime^^' final product produced by the enzymes intro-
112, 124 yj^jg demonstrates that the intermediate duced. In such transgenic plants, /7-hydroxyman-
A(;A^-dihydroxytyrosine is bound to the active site delonitrile is metabolized by endogenous enzymes
of CYP79A1 in a manner that prevents free rota- into a large number of different products. This
tion around the C-N single bond. is in sharp contrast to the results obtained
The further conversion of the (Z)-aldoxime into when CYP79A1 and CYP71E1 are expressed
the cyanohydrin was demonstrated to also be together with UGT85B1, in which case only
mediated by a multifunctional P450 using the micro- dhurrin formation is observed^^^. The transgenic
somal system isolated from sorghum as the biolog- dhurrin-producing A. thaliana plants showed
ical starting material. This P450 was assigned improved resistance against the flea beetle
CYP71E1 as the first member of the CYP71E sub- Phyllotreta nemorum, which is a crucifer
family. CYP71E1 catalyzes an unusual dehydration specialist^ ^^.
Cytochrome P450s in Plants 563

Enzymes catalysing
dhiirrm biosynthesis Ohwrrin

Tyrosine

Figure 12.5. A model for metabolon formation of the three biosynthetic enzymes CYP79A1 (P450Tyr),
CYP71E1 (P450ox), and UGT85B1 (glucosyltransferase) at the cytosoUc surface of endoplasmic reticulum.
Modified after Nelson and Strobel (1988).

5.1.2. Substrate Channeling and The possible organization of the enzymes


Metabolon Formation catalyzing a specific biosynthetic pathway into
multi-enzyme complexes, also denoted metabolons,
Administration of radiolabeled tyrosine to eti- has for many years been a point of discussion in
olated sorghum seedlings resulted in a 49% incor- plant biology. The existence of metabolons in
poration into dhurrin, but surprisingly no radio plants becomes increasingly apparent^^^, for
labeled intermediates involved in this conversion example, in the biosynthesis of cyanogenic gluco-
were detectable^ ^^' ^^°. Biosynthetic studies using sides^^^, phenylpropanoid, and flavonoid path-
highly active microsomal enzyme preparations ways^^^"^^^. Metabolon formation may serve to
demonstrated efficient channeling of the interme- overcome kinetic constraints, for example, by
diates in the pathway and provided an explanation mediating a considerable local increase in sub-
as to why no intermediates accumulate^^^ (Section strate availability and concentration and secure
5.1.1). Likewise, biosynthetic studies with recom- that labile and/or toxic intermediates are swiftly
binant CYP79A1 and CYP71E1 reconstituted converted into more stable and less toxic con-
with NADPH cytochrome P450 oxidoreductase stituents. Evolution of a metabolon for dhurrin
(ATR2) in artificial liposomes demonstrated effi- synthesis would appear essential to ensure rapid
cient flux through the pathway with barely conversion of the toxic /?-hydroxymandelonitrile
detectable levels of intermediates accumulating. intermediate by UGT85B1 to prevent its dissocia-
Upon inclusion of cytosolic sorghum extracts or tion into hydrogen cyanide and aldehyde at the
heterologously expressed UGT85B1 in the assays, same time as gaining efficacy in dhurrin produc-
almost complete stereospecific glycosylation tion. To demonstrate metabolon formation and to
of /7-hydroxymandelonitrile into dhurrin was identify the subcellular compartment into which
observed^^^' ^^^. These different sets of data sug- the metabolon accumulates, expression plasmids
gest that the combined presence of CYP79A1, harboring DNA sequences encoding fusion proteins
CYP71E1, and UGT85B1 results in the formation between the biosynthetic enzymes and spectral
of an active metabolon (Figure 12.5). variants of green fluorescent protein (GFP)^^^' ^^^
564 Kirsten A. Nielsen and Birger L. Moller

were designed. Fusion proteins in which each of obtained from studies of microsomal preparations.
the three enzymes, CYP79A1, CYP71E1, and The broadest substrate specificity is observed
UGT85B1, were C-terminally linked to either with the cassava microsomal preparation that is
cyano fluorescent protein (CFP) or yellow fluo- able to metabolize oximes derived from L-valine,
rescent protein (YFP) were functionally active L-isoleucine, L-phenylalalnine, L-tyrosine as well
when heterologously expressed in E. coli or as from cyclopentenylglycine^^^. Sorghum micro-
A. thaliana. Dhurrin-producing A. thaliana plants somal preparations are able to metabolize oximes
were obtained by simultaneous expression of derived from L-tyrosine and L-phenylalanine^^^.
CYP79A1, CYP71E1-CFP, and UGT85B1-YFP, Barley contains five different L-leucine-derived
but not by simultaneous expression of CYP79A1- cyanoglucosides of which only one is cyanogenic.
YFP, CYP71E1-CFP, and UGT85B1. This indi- These are thought to be formed by the action of
cates prevention of proper interaction between a single P450 that is able to hydroxylate all indi-
CYP79A1 and CYP71E1 when both are fused to vidual carbon atoms of the nitrile intermediate
fluorescent protein in spite of a retained function- and to facilitate multiple hydroxylations as well as
ality of each separate P450 fusion. Examination of dehydrations (Figure 12.7)^"*^. So far, the only
the transgenic plants by confocal laser scanning P450 known to^'catalyze this set of reactions is
microscopy (CLSM) demonstrated that a CYP71E1 isolated from sorghum.
metabolon visualized by UGT85B1-YFP is indeed
formed afler coordinated expression of the three
5.2. Functional Uniformity within
biosynthetic genes. The metabolon located in dis-
tinct domains at the cytosolic surface of the endo-
the CYP79 Family
plasmic reticulum appressed against the plasma To date the CYP79 family consists of six
membrane at the periphery of biosynthetically subfamilies denoted CYP79A, -B, -C, -D, -E, and
active cells (Figure 12.6A, B, see color insert). -F^o. Currently, the CYP79A subfamily has eight
When UGT85B1-YFP was expressed alone, it members covering four plant species of which
showed an even cytosolic distribution (Figure sorghum, T. aestivum (wheat) and H. vulgare
12.6C, see color insert). (barley) belong to the Poacea^^. The fourth plant
species is Arabidopsis that does not contain
5.1.3. Substrate Specificities cyanogenic glucosides. Instead, Arabidopsis is
able to synthesize glucosinolates, a closely related
The type of cyanogenic glucoside present in a group of natural products^' ^^'^. The amino acid
given plant species is defined by the substrate sequence identity between CYP79A1 from
specificity of the enzyme catalyzing the first sorghum and CYP79A2 from Arabidopsis is
committed step in the pathway. This conclusion 53%, slightly below the 55%i^' ^o, 22, 26 criterion
was reached from investigations of the amino acid usually required to assign P450s to the same sub-
specificity of active microsomal systems from family. Whereas the precise catalytic properties of
sorghum that is specific to L-tyrosine, the precur- the CYP79C subfamily remain to be established,
sor of dhurrin^^, seaside arrowgrass showing all other members of the CYP79 family have been
specificity to L-tyrosine, the precursor of shown to catalyze the conversion of an amino
taxiphyllin^^^' ^^^, cassava, flax, and white clover, acid to the corresponding oxime. Subfamilies
which are all specific to L-valine and L-isoleucine, CYP79A, -D, and -Es are involved in cyanogenic
the precursors of linamarin and lotaustralin'^^"^^^, glucoside synthesis whereas the subfamilies
and barley with specificity to L-leucine, the CYP79A, -B, and -F are involved in glucosinolate
precursor of epiheterodendrin^"^^. These same synthesis^. Introduction of the sorghum CYP79A1
specificities are also observed in in vitro assays gene into A. thaliana by genetic engineering
using recombinant protein from sorghum, cassava, resulted in the production of large amounts of the
and seaside arrowgrass^^' ^i' ^23 tyrosine-derived glucosinolate p-hydroxyglucosi-
The enz3niies catalyzing the subsequent steps nolate^"^^. This illustrates that the oxime produced
in cyanogenic glucoside synthesis, that is, the con- by the "cyanogenic" CYP79A1 serves as an
version of oximes into cyanohydrins are not nearly efficient substrate for the endogenous A. thaliana
as substrate specific. Again this knowledge was downstream biosynthetic enzymes mediating
Figure 12.6. Confocal laser scanning microscopy of ^. thaliana roots. (A) YFP fluorescence monitored using a
color code gradient ranging from black over red to orange to illustrate increased fluorescence intensities. The arrow
indicates the confined fluorescence at the periphery of cells co-expressing CYP79, CYP71, UGT85B1-YFP.
(B) Transmitted light image to visualize the cell shape. Arrow as in (A). (C) YFP fluorescence in cells expressing
UGT85B1-YFP shows even cytosolic distribution and high accumulation in and around the nucleus (arrow).
Bar = 5 |jLm. According to Tattersall et ah, unpublished.
Cytochrome P450s in Plants 565

!ll

c .~ t^
o 4 2fc -o
(D
" ^ •"«
m CD o^
_B^^
^e •D
CO

sS ^.-i o
, . L-
LU .-ti
T— C '5
03 C/5

^ 2 O
1-
S5
o
^ o

I
566 Kirsten A. Nielsen and Birger L. Moller

ac/-nitro
compound

AUK'M + U , NAUKM H cysteine


R,,^^COOH

NH, " ^ Kl

< >
S-alkyl-
thiohydroximate

nitrile oxide

UDPG PAPS

^ M
N^ ^ N

thioliydroximic desulfo-
acid glucosinolate
glucosinolate

Figure 12.8. The biosynthetic pathway for glucosinolate production. Reprinted with permission from Wittstock
and Halkier (2002)9.

glucosinolate formation. Most likely, this involves max (soybean) catalyzes conversion of the phenyl-
formation of a metabolon as demonstrated in urea herbicides, fluometuron, linuron, chlor-
sorghum (Section 5.1.2). An Arabidopsis double toluron, and diuron into more polar compounds^^.
mutant knocked out in both CY79B2 and This is unlikely to be the in planta biological
CYP79B3 completely lack indole-derived glu- function of the enzyme and surely does not
cosinolates, but show subtle morphological explain the apparent evolutionary need for main-
mutant phenotype. The subsequent conversion of tenance of 17 isoforms in the A. thaliana
oximes to glucosinolates is catalyzed by members genome^ ^. The CYP71B family is very large and
of the CYP83 family (Section 7; Figure 12.8). composed of 36 members all annotated from the
A. thaliana genome. The subfamily was first
established based on a sequence with unknown
biological function from Thlaspi arvense (field
5.3. Functional Diversity penny-cress), which like A. thaliana belongs to
among CYP71s the Brassicacea^'^.
In contrast to the CYP79 family, the CYP71
family is functionally diverse and constitutes the
largest A-type plant P450 family with a total of 5.3.2. CYP71C Subfamily: Grass-
110 members divided into 18 subfamilies. Specific Defense Compounds
The CYP71C subfamily is comprised of a total
of 23 members with 11 from Zea mays (com), 11
5.3.1. CYP71AandCYP71B from Trititum aestivum (wheat), and a single
member from H. vulgare (barley), all belonging to
Subfamilies
Poacea. The CYP71C subfamily possesses some
The CYP71A subfamily contains 28 members very special enzymatic features related to the fact
including 17 annotations from the A. thaliana that together different members of this subfamily
genome. The first member of this subfamily was are able to mediate the synthesis of the grass-spe-
derived from avocado^ ^ No specific enzymatic cific phytoalexin 2,4-dihydroxy-l,4-benzoxazin-
activity has been demonstrated for the members of 3-one (DIBOA)i49. Each of the 23 members
the CYP71A subfamily. CYP71A10 from Glycine catalyzes one of four consecutive enzyme reactions
Cytochrome P450s in Plants 567

in the DIBOA pathway (Figure 12.9). Thus, co- a maize EST collection in combination with a
ordinated enzymatic activities of CYP71C1, reverse genetics approach that revealed C-7
CYP71C2, CYP71C3vl, and CYP71C4 from hydroxylation of DIBOA forming 2,4,7-trihy-
maize mediate the production of DIBOA that is fur- droxy-2H-1,4-benzoxazin-3(4H)-one (TRIBOA)
ther metabolized to yield the cyclic hydroxamic acid by a 2-oxoglutarate-dependent dioxygenase^^^
2,4-dihydroxy-7-methoxy-1,4-benzoxazin-3-one (Figure 12.10). The high sequence identity among
(DIMBOA)i50. CYP71C4, CYP71C2, CYP71C1, and CYP71C3
Biosynthetic experiments using maize seedling does not compromise substrate specificity as
and radiolabeled [3-^^C]-indole as precursor demonstrated by determining the catalytic activi-
demonstrated that CYP71C4, CYP71C2, ties of the recombinant proteins expressed in
CYP71C1, and CYP71C3 catalyze the consecu- yeast^^^.
tive conversions into [3-^^C]-indolin-2-one, From an evolutionary perspective, it is inter-
[3-^^C]-hydroxyindonin-2-one, 2-hydroxy-l, 4-ben- esting that the phylogenetically closely related
zoxazin-3-one (HBOA), and DIBOA, respectively genes Bx2 (encoding CYP71C4), Bx3 (encoding
(Figure 12.9). An additional hydroxylation at the CYP71C2), Bx4 (encoding CYP71C1), and Bx5
C-7 position followed by C-7 specific methylation (encoding CYP71C3) co-locate to the short arm
gave rise to the formation of DIMBOA. The C-7 on chromosome 4 in the maize genome and to
hydroxylating enzyme was obtained by screening chromosome 5 on wheat genomes^^^. A fifth gene.

[ndoie>3<giyoerot phosphate
BXl^ TSA

cy
Indole Indole

C^»71C0v1 . « ^ )
wa CYI»7lC9v2«.(A) TSB I
aflP7lC4...(B>
Tryptophan

}ndollii-2>one
BX3 CYP71CTV2 .«(/^
CYP71Ca.,.CB)

3-Hydro)cymdoHn-2-one

BX4
1 CVF71Ce.^CA)
CVI*71C1 ,«(B)

^N^O CYP71C7^ r ^ W ^ l A o V^N^O


CYI»71C2...P)
1,4-B^zoxazin^>one HBOA HMBOA
^^ I CYf»7IC8v2.„C^
CyP71C3...(IE9 ^

OH OH
DIBOA Dt^OA

Figure 12.9. The biosynthetic pathway for DIMBOA. Bxl-Bx5 are gene names encoding the corresponding
CYPTlCs as indicated in (A) wheat and (B) maize. Reprinted with permission from Nomura et al. (2002)^^^.
568 Kirsten A. Nielsen and Birger L. Moller

O. ^OH

OCT "XXY
OH OH
TRIBOA

I
DIBCA &J\§

I
OH
DIMBOA
Figure 12.10. The 2-oxoglutarate-dependent dioxygenase Bx6 catalyzes hydroxylation of DIMBOA to produce
TRIBOA. Reprinted with permission from Frey et al. (2003)^^^

Bx6, encoding the oxoglutarate-dependent dioxy- this working hypothesis, Zhou et al. (1999)^^^
genase clusters with the CYPTlCs at the short have published that a padS A. thaliana mutant
arm of chromosome 4. In maize, DIMBOA con- unable to accumulate camalexin is defective in a
fers resistance to herbivores like Ostrinia nubilalis putative P450 monooxygenase gene, atmotated as
(European com borer) and Rhophalosiphum CYP71B15'8'2o^
maydis (maize plant aphid) and to the fungal
pathogen Helminthosporium turcicum (Northern
com blight). The DIMBOA pathway may exem- 5.3.3. CYP71D,-F, and-R
plify an evolutionary recent recruitment of new
Subfamilies
biological activities of P450s. The substrate for
DIMBOA synthesis, indole or indole-3-glycerol CYP71D subfamily is also large and currently
phosphate is suggested to derive from a branch comprises a total of 22 members from 10 different
point in L-tryptophan synthesis. A sixth gene Bxl plant species. At present, the catalytic properties
encoding a tryptophan synthase homologue is situ- of five CYP71D enzymes have been determined
ated together with the cluster of DIMBOA genes on and the enzymes assigned to specific steps in
chromosome 4 in maize and was shown to be indole alkaloid, sequiterpenoid, cyclic terpenoid,
essential for DIMBOA production''^^. A homologue and flavonoid synthesis. Accordingly, enzymes
of this gene was activated by a herbivore elicitor, belonging to the CYP71D subfamily do not nec-
thus strengthening the suggestion of an introduc- essarily share similar functional characteristics.
tion of a branch point in L-tryptophan biosynthe- The first member to be functionally character-
sis for DIMBOA production in response to ized was CYP71D12 from Catharanthus rosea
herbivore attack^^^. Transcription of the maize (Madagascar periwinkle). CYP71D12 was identi-
genes encoding CYP71C1 (Bx4) and CYP71C3 fied as the tabersonine 16-hydoxylase enz5niie
(Bx5) are induced in response to the maize bacte- involved in the biosynthetic pathway for the two
rial pathogen Acidovorax avenae and in response medically important bisindole alkaloids vinblas-
to wounding^^^. No CYP71C homologues are tine and vincristine^^'^ (Figure 12.11). Microsomal
identified in the Arabidopsis genome. However, preparations from etiolated seedlings of Madagas-
the stmcture of DIMBOA is sufficiently close to car periwinkle were shown to be low in taberson-
the indole-derived phytoalexin camalexin that is ine 16-hydroxylase activity in comparison to light
produced by A. thaliana to allow speculations on grown seedling. Interestingly, the light regulation
a tight functional relationship between CYPTlCs was retained in suspension cultures of Madagascar
and Arabidopsis P450 candidates^^'*. In support of periwinkle. A cDNA clone encoding tabersonine
Cytochrome P450s in Plants 569

Tryptophan convert loganin into serologanin (Figure 12.11)^^^.


Garaoiol
Activation of the 0RCA3 gene is regulated by
i t
methyl jasmonate. This plant hormone is produced
in response to stress and wounding^^^ thereby
Tryptamine Secolc^anin enabling synthesis of the bisindole alkaloids as
response to herbivore attack.
Strictosidine
A second functionally identified member of the
CYP71D subfamily is CYP71D20i^^ This enzyme
Tabersonine Catharanthina from tobacco mediates production of the sesquiter-
pene capsidiol, an antimicrobial compound. The
t enzyme catalyzes hydroxylations of 5-epi-aristone
Vindoline
as well as of 1-deoxy-capsidiol to capsidiol^^^ The
functional and mechanistic features of CYP71D20
3,4~Anhydrovinbfa8tine were determined in a coupled assay using substrate
(Bisindole)
production by sesquiterpene synthases and a micro-
somal system ^^^. CYP71D20 was found to catalyze
unique stereo- and regiospecific hydroxylations
first at carbon atom-1 followed by rotation of
the molecule in the active site and a second hydro-
xylation at carbon atom-3 of the bicyclic sesquiter-
pene hydrocarbon skeleton. The CYP71D20 gene
is induced in response to fungal elicitors like
paraciticein^^^.
The third functionally characterized member
of the CYP71D subfamily is CYP71D9. This
enzyme has been identified in soybean as a
flavonoid 6-hydroxylase. It was demonstrated that
hydroxylation of carbon atom-6 of the A-ring pre-
Vioblastlria, R = CHg
Vincristine, R = CHO cedes 1,2-aryl migration to produce isoflavonoids
as described in Section 5.4^^"^.
Figure 12.11. The biosynthetic pathway for the Regiospecific hydroxylation of the monoter-
bisindole alkaloids vinblastine and vincristine. Reprinted pene (—)-4*S'-limonene at the C-3 or C-6-allylic
with permission from Schroeder et al. (1999)^^^. positions to yield (—)-menthol (peppermint) or
(—)-carvone (spearmint), respectively, is accom-
16-hydroxylase was isolated from a cDNA library plished by the last two functionally characterized
prepared from light-induced cells using degener- CYP71DS, the CYP71D13 and CYP79D18 found
ate oligodeoxynucleotide primers and verified by in commercial mint species {Mentha sp.)^^^"^^^.
heterologous expression in E. coli. The other
enzymes involved in the conversion of tabersonine 5.4. Specialized Defense—
to vindoline may also be light induced and this
Isoflavonoids in Legumes
may provide a route for their isolation and cloning
and for the production of vinblastine and vin- Plant isoflavonoids possess a wide range of
cristine by expression of the entire pathway from biological activities. They are efficient antimicro-
the precursors tryptamine and secologanin in cell bial agents, inducers of the nodulation genes of
cultures. A transcriptional regulator Octadecanoid- symbiotic Rhizobium bacteria and phytoestrogens
derivative responsive Catharanthus ^P2-domain that work through the human estrogen receptor
protein (0RCA3) activates the expression of causing alterations in serum lipids and bone
genes mediating L-tr5^tophan and tryptamine metabolism^' ^^^. Isoflavonoids are produced
production as well of several genes in the synthe- almost exclusively in the Leguminosae in the
sis of vindoline from tryptamine and secologanin^^^. order Fabales. Isoflavonoids are produced from L-
A cytochrome P450, CYP72A1, was shown to phenylalanine that condenses with 4-coumaroyl
570 Kirsten A. Nielsen and Birger L. Moller

^ tfa

^ ^

>. c^
Cytochrome P450s in Plants 571

CoA and three molecules of malonyl CoA to pro- and an isoflavone-O-methyltransferase, desig-
duce chalcone and subsequently the flavanones nated I0MT8 was suggested to guide 4'-0-methy-
naringenin and liquiritigenin (Figure 12.12). The lation and to prevent 7'-0-methylation in spite of
synthesis of isoflavonoids from these flavanones the fact that in vitro IOMT8 was found to catalyze
is mediated by a CYP93C that catalyzes the 7'-0-methylation^^^. However, this intricate reac-
migration of the B-ring to the 3-position followed tion mechanism has recently been challenged by
by hydroxylation at the 2-position. The CYP93Cs the cloning and functional characterization of
are therefore termed 2-hydroxy-isoflavone 2,7,4'-trihydroxyisoflavanone 4'-O-methyltrans-
synthases^^' ^^^. CYP93C genes have been ferases from G. echinata (licorice) and Lotus
cloned from G. max (soybean; CYP93Clv2)^^ Japonicus (Bird's foot trefoil) that exhibit high
Glycyrrhiza echinata (licorice; CYP93C2)^^^, and affinity for 4-0-methylation of daidzein^'^^.
several other legumes: Trifolium pratense (red
clover), Trifolium repens (white clover), mung
bean, M. sativa (alfalfa), Lens culinaris Medik.
(lentil), Pisum sativum L (snow pea), Vicia villosa
6. P450 Mediated Production
(hairy vetch), and Lupinus spp. Lupin^-^^ of Alkaloids with Medicinal
CYP93C enzymes catalyze the first committed Importance
step in the isoflavonoid pathway. Insertion of
CYP93Cs into A. thaliana by genetic engineering In previous parts of this review, natural products
enabled production of low levels of genistein in with interestmg medicinal uses have been men-
this non-leguminous plant^^^' ^^^' ^^^. Increased tioned like the bisindoles and isoflavonoids^"^' ^^'^. In
expression of CYP93Cv2 did not add to produc- this context, a number of other alkaloids are impor-
tion^^^. When CYP93Cv2 was expressed in the tant. The tetrahydrobenzylisoquinoline alkaloid
tt3, tt6 double-mutant^^^-i^^ that is blocked with berberine constitutes the first complex alkaloid for
respect to flavonol synthesis (see Figure 12.12), which the enzymes catalyzing the entire biosyn-
the genistein content was increased 3-fold^^^. thetic pathway from the primary precursor L-
Accordingly, competition for common substrates tyrosine have been identified. Biosynthesis of
is an important parameter to consider in optimiz- tetrahydrobenzylisoquinoline alkaloids involves a
ing the production of desired natural products ^^^. number of P450s with high substrate specificity and
The production of the two isoflavonoids catalysing stereo- and regiospecific oxidations^^^.
daidzein and genistein is highly induced by (»S')-Ar-methylcoclaurine 3'-hydroxylase assigned as
pathogen attack. Elicitation by crude polysaccha- CYPSOBl^i catalyzes the conversion of {S)-N-
ride preparations from yeast cell wall was used to methylcoclaurine to (/S')-3'-hydroxy-A/-methylco-
facilitate biosynthetic studies in alfalfa cell sus- claurine, which by methylation is transformed into
pension cultures ^^^. A signal pathway dependent (*S)-reticuline, which represents the branch point for
on endogenously generated nitric oxide is also formation of a vast number of different tetrahy-
responsible for the induction of daidzein and droisoquinoline alkaloids including the berberine-,
genistein synthesis ^^^. Nitric oxide is generated phenan-threne- and benzo[c]phenanthridine-type
from L-arginine by nitric oxide synthases (NOS). alkaloids (Figure 12.13).
Although NOS belongs to the class of heme-thio- The conversion of (*S)-reticuline into berberine
late proteins, the crystal structure of NOS clearly includes two remarkable P450 enzymes. The first is
demonstrates that they belong to a different class the unique berberine bridge QnzymQ (BBE) that
of heme proteins as the P450 superfamily^^^ catalyzes the introduction of a new C-C bond in its
enzymes. In alfalfa, improved protection against product, (*S)-scoulerinei3' ^^^ (Figure 12.13). The
fungal pathogens is achieved by 4'-0-methylation second enzyme has been designated as canadine
of daidzein into formononetin followed by a num- synthase and introduces a methylene dioxy-bridge^^^
ber of unidentified hydroxylation steps to yield The enzymatic mechanism for methy-lene dioxy-
the highly antifungal phytoalexin, medicarpin bridge formation^^^ is outlined in Figure 12.14.
(Figure 12.12)^^^. The 4'-0-methylation reaction Synthesis of the phenanthrine-type alkaloid morphine
has been studied in detail. Intricate physical inter- from (*S)-reticuline via (i?)-reticuline demands the
action between the CYP93C isoflavonoid synthase involvement of three NADPH-dependent reductases
572 Kirsten A. Nielsen and Birger L. Moller

XJi m

CYP8081

{A)«/tf-Me(hylcoctaurw!8

HO

mathyr«ocla«ri«e

I
OCM,

^*.

H^CO

(»)«R6liculkw

OCHs
' ' ^ ' "
^^;*''^(OCH5

Figure 12.13. The biosynthetic pathway for L-tyrosine derived tetrahydrobenzyUsoquinohne alkaloids. The core
production of (iS)-reticuline and the branch points for berberine, macarpine, and morphine production. Reprinted with
permission from Chou and Kutchan (1998)'^^.

and the P450 enzyme salutaridine synthase ^^^ methylene dioxy-bridge formations are key catalytic
that like the BBE introduces a new C-C bond in the features of the conversion^ ^^.
product (Figure 12.15). So far the corresponding The CYP80 family involved in the production
gene of only one of the reductases has been of ((S')-reticuline in the opium poppy is not at all
cloned^ ^. Synthesis of the antimicrobial represented in the Arabidopsis genome. This
benzo[c]phenanthridine-type alkaloid macarpine reflects the fact that the production of a specific
(Figure 12.13) from (*S)-reticuline involves the alkaloid typically is restricted to a particular plant
action of six P450s that have been studied in plant species or to a limited number of species within a
cell cultures^^^~^^'^. New C-C bond formation and family. Accordingly, it is normally not possible to
Cytochrome P450s in Plants 573

study such pathways in genetically well-character- encoding the entire pathway and hampered by
ized model plants. This greatly complicates eluci- technical problems in the co-expression of a
dation of the biosynthetic pathways involved in multitude of heterologous genes. ^^^
alkaloid formation. Furthermore, alkaloids may
accumulate very slowly over a period of months to
years and in a highly tissue-specific manner. The
establishment of cell cultures have helped to over-
7. Future Prospects: Crosstalk
come some of these experimental difficulties ^^^. and Metabolic Engineering
Availability of native alkaloid producing plants as
sources for isolation of important medicinal drugs The multigene family of plant P450s repre-
remain of high importance because controlled sents a very rich source for metabolic engineering.
production in, for example, transgenic A. thaliana The A-type P450s involved in the synthesis of low
is dependent on the availability of the genes molecular mass natural products is a key target
because many of these compounds are of high
value either as fine chemicals or as plant con-
stituents that provide desired agronomical traits
such as insect or fungal resistance. In all cases, the
Ring A Of D | Y V * ^ " * ^ ^ ^ ^I ^ ' Heme-iron of
of protobdrberlne ^OH cytochrome P-450 P450 enzymes catalyzing the first committed step
alkaloid in the different pathways leading to the production
of natural products appear to exert a very high
DOC '^OHj " • ' O r f S S '
degree of substrate specificity. The successful
transfer of the entire pathway for dhurrin forma-
tion from sorghum to A. thaliana^^^ demonstrates
that metabolon formation may be achieved also
after heterologous expression of a biosynthetic
pathway in a plant species that would not in nature
produce the same type of natural products.
l> Insertion of an incomplete pathway was shown to
favor crosstalk with other metabolic pathways
Figure 12.14. A proposed mechanism of methylene and the formation of side products ^^^. When
dioxy-bridge formation. Reprinted with permission from separately introduced into A. thaliana, sorghum
Chou and Kutchan (1998)^^0. CYP79A1 was able to establish highly efficient

Salutaridlne ^ ^
synthase
^ ^Q.
NADPH, O2

fP^"^

Demethytation
(P-480)

H3CO.
Demethylation

(P-450)

Neopinone

Figure 12.15. Biosynthesis of morphinefrom(i?)-reticuline. Reprinted with permissionfromChou and Kutchan


(1998)180.
574 Kirsten A. Nielsen and Birger L. Moller

interaction with downstream glucosinolate-pro- and knockout of these two enzyme activities result
ducing enzymes to create a new metabolon that in altered phenotypes and pleiotrophic effects.
resulted in the accumulation of large amounts of Increased formation of lateral roots was associated
/7-hydroxybenzylglucosinolate in A. thaliana^^^ with altered levels of indole acetic acid and pro-
and thereby changing the overall glucosinolate vided evidence that fluxes of intermediates
profile of ^. thaliana^^^. directed toward natural product formation may
The possibility to redirect L-tyrosine into the serve an important frinction to balance primary
glucosinolate or cyanogenic glucoside pathways metabolism"*^' ^'*' ^^^. Surprisingly, disturbance of
without loss of plant fitness^' ^^^ demonstrates the oxime metabolism affects phenylpropanoid metab-
existence of immanent routes for transport and olism and the monomer composition of lignin^^.
storage of new classes of natural products intro- The link between these different phenomena is not
duced into plants by genetic engineering, and an yet understood.
inherent ability to redirect and optimize the flux of In the synthesis of natural products, increased
intermediates to counteract inbalances in primary diversity is often achieved by a final set of modi-
and secondary metabolism"*^. The availability of a fications including hydroxylations, glucosyla-
metabolic grid with numerous metabolic cross- tions, methylations, and acylations. As a result, the
points to accommodate the synthesis of natural flavonol quercitin may be transformed into 300
products upon demand is well documented. To different glucosides^^^. Berries of Vitis vinifera
enable the production of physiologically active (grape wine) accumulate over 200 different
amounts of DIMBOA in grasses without depleting aglycones that each may be decorated differ-
the indole-3-glycerol phosphate pool for trypto- ently^^^' ^^^. Most likely, the synthesis of the basic
phan synthesis, gene duplication has provided two structures of natural products is facilitated by
modified genes each encoding enzymes that cat- metabolon formation. Dependent on cell type,
alyze the same reaction but are directed toward developmental stage and elicitation as a result of
different biochemical routes^^'*. In periwinkle, a abiotic or biotic stresses, additional enzyme activ-
transcription factor 0RCA3 upregulates the syn- ities may be bound to the basic metabolons to
thesis of L-tryptophan to provide efficient synthesis secure that desired specific modifications are
of the inducible bisindole alkaloids. Bisindole alka- obtained. The broad in vitro substrate specificity
loid synthesis is also dependent on the availability observed for 0-methyltransferases^'^^' *^^ and
of secologanin and the rate-limiting step in its syn- UDPG-glucosyltransferases*^^' ^^"^ may reflect that
thesis appears unaffected by 0RCA3. The opposite in vivo these will be associated to metabolons that
situation where L-tryptophan accumulates due to prevent general access to their active sites. In this
blockage of natural product synthesis is also possi- manner, the cell is able to maintain the potential
ble as observed in the double knockout mutant in to specifically decorate a large array of natural
Arabidopsis lacking the tryptophan metabolizing products without having to produce a separate
CYP79B2 and CYP79B3 enzymes'^l Such plants enzyme for each reaction. As an added benefit,
completely lack indole-derived glucosinolates but metabolon formation may prevent undesired reac-
only exhibit temperature-dependent phenotypic tions, for example, random glucolylation of plant
difference. So accumulation of free L-tryptophan hormones.
does not appear to severely compromise wild-type Based on the understanding of the basic prin-
growth characteristics, for example, by the forma- ciples for metabolon formation, in a foreseeable
tion of excess amounts of the tryptophan-derived future it may be possible to transfer the entire
indole acetic acid. pathways for synthesis of desired alkaloids into
The ability to accommodate altered levels of more convenient production plants from which
intermediates depends on the type of compounds these compounds can be isolated in high amounts.
involved. In A. thaliana, tryptophan-derived A main obstacle to reach these goals is knowl-
oximes are key intermediates in the formation of edge of the proper P450, UDPG-glucosyltrans-
the phytohormone indole acetic acid as well as in ferases, methyltransferases, and acyltransferases.
the synthesis of glucosinolates. CYP83A1 and Typically, these genes are not present in geneti-
CYP83B1 are the enzymes responsible for con- cally well-defined model plants like A. thaliana
verting oximes into glucosinolates. Overexpression and rice. They have to be traced often from exotic
Cytochrome P450s in Plants 575

plants for which no genome program and not even associated with insecticide resistance in
cDNA hbraries are available. System biology Drosophila. Science 297, 2253-2256.
technologies like metabolite profiling, pro- 8. Mikkelsen, M.D., B.L. Petersen, C.E. Olsen, and
teomics, and transcriptomics may help to identify B. Halkier (2002). Biosynthesis and metabolic
engineering of glucosinolates. Amino Acids 22,
the proper enzymes and genes by unraveling coin-
269-275.
cidences of enhanced expression, protein appear-
9. Wittstock, U. and B.A. Halkier (2002). Glucosinolate
ance, and accumulation of specific metabolites. A research in the Arabidopsis era. Trends Plant Sci. 7,
genomics approach to elucidate the biosynthesis 263-270.
of the triterpene saponin in Medicago truncatula 10. Wang, E., R. Wang, J. DeParisis, J.H. Loughrin,
based on data mining of EST resources^^^ S. Gan, and G.J. Wagner (2001). Suppression of a
and saponin metabolite profiles ^^^ resulted P450 hydroxylase gene in plant trichome glands
in the identification of three putative pathway enhances natural-product-based aphid resistance.
enzymes^^^. In such approaches, metabolon for- Nat. Biotechnol 19, 371-374.
mation may render it difficult to detect the true 11. PauH, H.H. and TM. Kutchan (1998). Molecular
intermediates of a pathway. Reconstitution of a cloning and functional heterologous expression
of two alleles encoding (5)-N-methylcoclaurine
biosynthetic pathway by heterologous expression
3'-hydroxylase (CYP80B1), a new methyl jas-
in a model plant is important to avoid wrong
monate-inducible cytochrome P-450-dependent
conclusions. In spite of the experimental limita- mono-oxygenase of benzylisoquinoline alkaloid
tions described above, progress on P450s and nat- biosynthesis. Plant J. 13, 793-801.
ural product synthesis moves quickly ahead 12. Unterlinner, B., R. Lenz, and T.M. Kutchan (1999).
thanks to hard work and the original approaches Molecular cloning and functional expression of
taken by many scientists involved in this codeinone reductase—^the penultimate enzyme in
research area. morphine biosynthesis in the opium poppy Papaver
somniferum. Plant J. 18, 465^75.
13. Huang, EC. and T.M. Kutchan (2000). Distribut-
ion of morphinan and benzo[c]phenanthridine alka-
References loid gene transcript accumulation in Papaver
somniferum. Phytochemistry 53, 555-564.
1. Croteau, R., T.M. Kutchan, and N.G. Lewis (2000). 14. Dixon, R.A. and D. Ferreira (2002). Genistein.
Natural products (secondary metabolites). In Phytochemistry 60, 205-211.
B.B. Buchanan, W. Gruissem and R.L. Jones, 15. Steele, C.L., M. Gijzen, D. Qutob, and R.A. Dixon
(eds), Biochemistry and Molecular Biology of (1999). Molecular characterization of the enzyme
Plants. American Society of Plant Physiologists, catalysing the aryl migration reaction of
Rockville, MD, pp. 1250-1318. isoflavonoid biosynthesis in soybean. Arch.
2. Dixon, R. A. (2001). Natural products and plant dis- Biochem. Biophys. 367, 146-150.
ease resistance. Nature 411, 843-847. 16. The Arabidopsis initiative (2000). Analysis of the
3. Kutchan, T.M. (2001). Ecological arsenal and genome sequence of the flowering plant
developmental dispatcher. The paradigm of second- Arabidopsis thaliana. Nature 408, 796-815.
ary metabolism. Plant Physiol 125, 58-60. 17. Clough, S.J. and A.R Bent (1998). Floral dip: A
4. Morant, M., S. Bak, B.L. MHer, and D. Werck- simplified method for Agrobacterium—^mediated
Reichart (2003). Plant cytochromes P450: Tools for transformation of Arabidopsis thaliana. Plant J. 16,
pharmacology, plant protection and phytoremedia- 735-743.
tion. Cum Opin. Biotech. 14, 1-12. 18. Schuler, M.A. and D. Werck-Reichhart (2003).
5. Volker, S., P Joern, and W. Boland (1999). Functional genomics of P450s. Annu. Rev. Plant
Biosynthesis of furanocoumarins: Mevalonate Biol. 54, 629-667.
independent prenylation of umbelliferone in Apium 19. Cytochrome P450 homepage: http://dmelson.
graveolens (Apiaceae) Phytochemistry 50, utmem. edu/LAtalk.html
1141-1145. 20. Cytochrome P450 homepage: http://dmelson.
6. Li, X., M.A. Schuler, and M.R. Berenbaum (2002). utmem. edu/biblioD.html
Jasmonate and salicylate induce expression of her- 21. The Arabidopsis P450, cytochrome bj, P450 reduc-
bivore cytochrome P450 genes. Nature 419, tase, and Glycosyltransferase Family 1 Site at
712-715. PlaCe: http://biobase.dk/P450/p450.shtml
7. Daborn, PJ., J.L. Yen, M.R. Bogwitz, G.L. Goff, 22. Werck-Reichart, D., S. Bak, and S. Paquette (2002).
E. Feil, S. Jeffers et al. (2002). A single P450 allele Cytochromes P450. In C.R. Somerville and
576 Kirsten A. Nielsen and Birger L. Moller

E.M. Meyerowitz (eds), The Arabidopsis book. Molecular characterization of an Arabidopsis gene
American Society of Plant Biologists, Rockville, encoding hydroperoxide lyase, a cj^ochrome P-450
MD (www.aspb.org/publications/arabidopsis). that is wound inducible. Plant Physiol. 117,
23. Goff, S.A., D. Ricke, T.H. Lan, G. Presting, 1393-1400.
R.L. Wang, M. Dunn et al (2002). A draft sequence 36. Schoenbohm, C, S. Martens, C. Eder,
of the rice genome {Oryza sativa L. ssp Japonica). G. Forkmann, and B. Weisshaar (2000).
Science 296, 92-100. Identification of \hQ Arabidopsis thaliana flavonoid
24. Yu, J., S. Hu, J. Wang, G.K.S. Wong, S. Li, 3'hydroxylase gene and functional expression of the
B. Liu et al. (2002). A draft sequence of the rice encoded P450 enzymes. Biol. Chem. 381, 749-753.
genome (Oryza sativa L. ssp indica). Science 296, 37. Wittstock, U. and B. Halkier (2000). Cytochrome
79-92. P450 CYP79A2 from Arabidopsis thaliana L.
25. Cytochrome P450 homepage: http://drnelson. catalyzes the conversion of L-phenylalanine to
utmem. edu/rice. color, sept 12 .html phenylacetaldoxime in the biosynthesis of benzyl-
26. Nelson, D.R., L. Koymans, and T. Kamataki (1996). glucosinolate. ^ 5/o/. Chem. 275, 14659-14666.
P450 superfamily: Update on new sequences, gene 38. Mikkelsen, M.D., C.H. Hansen, U. Wittstock, and
mapping, accession numbers and nomenclature. B.A. Halkier (2000). Cytochrome P450CYP79B2
Pharmacogenetics 6, 1^2, from Arabidopsis catalyzes the conversion of tryp-
27. Yoshida, Y, Y Aoyama, M. Noshiro, and O. Gotoh tophan to indole-3-acetaldoxime, a precursor of
(2000). Sterol 14-demethylase P450 (CYP51) pro- indole glucosinolates and indole-3-acetic-acid.
vides a breakthrough for the discussion on the evo- J. Biol. Chem. 275, 33712-33717.
lution of cytochrome P450 gene superfamily. 39. Hansen, C.H., U Wittstock, C.E. Olsen, A.J. Hick,
Biochem. Biophys. Res. Comm. 273, 799-804. J.A. Pickett, and B.A. Halkier (2001). Cytochrome
28. Durst, F. and DR. Nelson (1995). Diversity and P450 CYP79F1 from Arabidopsis catalyzes the
evolution of plant P450 and P450-reductases. Drug conversion of dihomomethionine and trihomome-
Metabol Drug Interact. 12, 189-206. thionine to the corresponding aldoximes in the
29. Paquette, S.M., S. Bak, and R. Feyereisen (2000). biosynthesis of aliphatic glucosinolates. J. Biol.
Intron-exon organization and phylogeny in a large Chem. 276, 11078-11085.
superfamily, the paralogous cytochrome P450 genes 40. Reintanz, B., M. Lehnen, M. Reichelt,
oiArabidopsis thaliana. DNA Cell Biol. 19,307-317. J. Gershenzon, M, Kowalczyk, G. Sandberg et al.
30. Kushiro, M., T. Nakano, K. Sato, K. Yamagishi, (2001). bus, a bushy Arabidopsis CYP79F1 knock-
T. Asami, A. Nakano et al. (2001). Obtusifoliol out mutant with abolished synthesis of short-chain
14a-demethylase (CYP51) antisense Arabidopsis aliphatic glucosinolates. Plant Cell 13, 351-367.
shows slow growth and long life. Biochem. 41. Chen, S., E. Glawischnig, K. Jegensen, P. Naur,
Biophys. Res. Comm. 285, 98-104. B. Jcgensen, C.E. Olsen et al. (2003). CYP79F1
31. Neff, M.M., S.M. Nguyen, E.J. Malancharuvil, CYP79F2 have distinct functions in the biosynthe-
S. Fujioka, T. Noguchi, H. Seto et al. (1999). BASl sis of aliphatic glucosinolates in Arabidopsis. Plant
a gene regulating brassinosteroid levels and light J. 33, 923-937.
responsiveness in Arabidopsis. Proc. Natl. Acad. 42. Barlier, I., M. Kowalczyk, A. Marchant, K. Ljung, R.
Sci. USA 96, 15316-15323. Bhalerao, M. Bennett et al. (2000). The SUR2 gene
32. Bell-Lelong, DA., J.C. Cusumano, K. Meyer, of Arabidopsis thaliana encodes the cytochrome
and C. Chappie (1997). Cinnamate-4-hydroxylase P450 CYP83B1, a modulator of auxin homeostasis.
expression in Arabidopsis. Regulation in response to Proc. Natl. Acad Sci. USA 97, 14819-14824.
development and the environment. Plant Physiol. 113, 43. Bak, S. and R. Feyereisen (2001). The involvement
729-738. of two P450 enzymes, CYP83B1 CYP83A1, in
33. Laudert, D., U. Pfannenschmidt, F. Lottspeich, homeostasis and glucosinolate biosynthesis. Plant
H. Hollander-Czytko, and E.W Weiler (1996). Physiol. 127, 108-118.
Cloning, molecular and functional characterization 44. Hansen, C.H., L. Du, R Naur, C.E. Olsen,
of Arabidopsis thaliana allene oxide synthase K.B. Axelsen, A.J. Hick et al. (2001). CYP83B1 is
(CYP74), the first enzyme of the octadecanoid the oxime metabolizing enzyme in the glucosino-
pathway to jasmonates. Plant Mol. Biol. 31, late pathway in Arabidopsis. J. Biol. Chem. 276,
323-335. 24790-24796.
34. Staswick, RE. (1999). Sequence of an alene oxide 45. Meyer, K., J. Cusumano, C. Somerville, and
synthase cDNA from Arabidopsis thaliana. Plant C. Chappie (1996). Ferulate-5-hydroxylase from
Physiol. 121,312. Arabidopsis thaliana defines a new family of
35. Bate, N.J., S. Sivasankar, C. Moxon, J.M.C. Riley, cytochrome P450-dependent monooxygenases.
J.E. Thompson, and S.J. Rothstein (1998). Proc. Natl. Acad Sci. USA 93, 6869-6874.
Cytochrome P450s in Plants 577

46. Shimada, Y., S. Fujioka, N. Miyaushi, M. Kushiro, 4-hydroxylase cytochrome P450 from alfalfa. Arch.
S. Takatsuto, T. Nomura et al (2001). Biochem. Biophys. 305, 509-515.
Brassinosteroid 6-oxidases from Arabidopsis and 57. Logemann, E., M. Parniske, and K. Hallbrock
tomato catalyze multiple C-6 oxidations in brassi- (1995). Modes of expression and common structu-
nosteroid biosynthesis. Plant Physiol 126, ral features of the complete phenylalanine ammonia-
770-779. lyase gene family in parsley. Proc. Natl. Acad. Sci.
47. Benveniste, I., N. Tijet, F. Ada, G. Phillipps, USA 92, 5905-5909.
J.P. Salaii, and F. Durst (1998). CYP86A1 from 58. Ge, L. and VL. Chiang (1996). A full length cDNA
Arabidopsis thaliana encodes a cytochrome P450- encoding trans-cinnamate 4-hydroxylase from
dependent fatty acid omega-hydroxylase. Biochem. developing xylem of Populus tremuloides. Plant
Biophys. Res. Commun. 243, 688-693. Physiol. 112,861.
48. Wellesen, K., F Durst, F. Pinot, I. Benveniste, 59. Ro, D.K., N. Mah, B.E. Ellis, and C.J. Douglas
K. Nettesheim, E. Wisman et al. (2001). Functional (2001). Functional characterization and subcellular
analysis of the LACERATA gene of Arabidopsis localization of poplar {Populus trichocarpa X
provides evidence for different roles of fatty acid Populus deltoids) cinnamate 4-hydroxylase. Plant
co-hydroxylation in development. Proc. Natl. Acad. Physiol. 126,317-329.
Sci. USA 98, 9694-9699. 60. Batard, Y, A. Hehn, S. Nedelkina M. Schalk,
49. Helliwell, C.A., PM. Chandler, A. Poole, K. Pallet, H. Schaller et al. (2000). Increasing
E.S. Dennis, and W.J. Peacock (2001). The CYP88A expression of P450 and P450-reductase proteins from
cytochrome P450, ent-kaurenoic acid oxidase, cat- monocots in heterologous systems. Arch. Biochem.
alyzes three steps of the gibberellin biosynthetic Biophys. 379, 161-169.
pathway. Proc. Natl. Acad Sci. USA 98, 2065-7080. 61. Overkamp, S. and W Barz (1999). Isolation of a full
50. Szekeres M., K. Nmeth, Z. Konz-K&nan, length cDNA encoding trans-cinnamate 4-hydroxy-
J. Mathur, A. Kauschmann, T. Altman et al. (1996). lase from chickpea. Plant Physiol. 120, 635.
Brassinosteroids rescue the deficiency of CYP90, a 62. Song, W.C, CD. Funk, and A.R. Brash (1993).
cytochrome P450, controlling cell elongation de- Molecular cloning of an allene oxide synthase: A
etiolation in Arabidopsis. Cell 85, 171-182. cytochrome P450 specialized for the metabolism of
51. Choe, S., B.P Dilkes, S. Fujioka, S. Takatsuto, fatty acid hydroperoxides. Proc. Natl. Acad. Sci.
A. Sakurai, K.A. Feldman (1998). The DWF4 gene USA 90, 8519-8523.
of Arabidopsis encodes a cytochrome P450 that 63. Maucher, H., B. Hause, I. Feussner, J. Ziegler, and
mediates multiple 22a-hydroxyaltion steps in C. Wasternack (2000). Allene oxide synthases of
brassinoid biosynthesis. Plant Cell 10, 231-243. barley (Hordeum vulgare cv. Salome): Tissues spe-
52. Schoch, G., S. Goepfert, M. Morant, A. Hehn, cific regulation in seedling development. Plant J.
D. Meyer, P Ullmann et al. (2001). CYP98A3 from 21, 199-213.
Arabidopsis thaliana is a 3'-hydroxylase of pheno- 64. Howe, G.A., G.L Lee, A. Itoh, L. Li, and
lic esters, a missing link in the phenylpropanoid A.E. DeRocher (2000). Cytochrome P450-depend-
pathway. J. Biol. Chem. 276, 36566-36574. ent metabolism of oxylipids in tomato. Cloning and
53. Helliwell, C.A., C.C. Sheldon, M.R. Olive, expression of allene oxide synthase and fatty acid
A.R. Walker, J.A.D. Zeevaart, W.J. Peacock et al. hydroperoxide lyase. Plant Physiol. 123, 711-724.
(1998). Cloning of tht Arabidopsis ent-kameriQ oxidase 65. Siminszky, B., ET. Corbin, E.R. Ward,
gene GA3. Proc. Natl. Acad. Sci. USA 95,9019-9024. T.J. Fleischmann, and R.E. Dewey (1999).
54. Teusch, H.G., M.R Hasenfratz, A. Lesot, C. Stoltz, Expression of a soybean cytochrome P450
J.M. Gamier, J.M. Jeltsch et al. (1993). Isolation monooxygenase cDNA in yeast and tobacco
sequence of a cDNA encoding the Jerusalem arti- enhances the metabolism of phenylurea herbicides.
choke cinnamate 4-hydroxylase, a major plant Proc. Natl. Acad Sci. USA 96, 1750-1755.
cytochrome P450 involved in the general phenyl- 66. Fiehn, O., J. Kopka, P. Dcmann, T. Altmann,
propanoid pathway. Proc. Natl. Acad. Sci. USA 90, R.N. Trethewey, and L. Willmitzer (2000).
4102-4106. Metabolite profiling for plant functional genomics.
55. Mizutani, M., E. Ward, J. DiMaio, D. Ohta, J. Ryals, Nat. Biotechnol. 18, 1157-1161.
R. Sato (1993). Molecular cloning and sequencing 67. Sumner, L.W, P Mendes, and R.A. Dixon (2003).
of a cDNA encoding mung bean cytochrome P450 Plant metabolomics: Large-scale phytochemistry in
(P450C4H) possessing cinnamate 4-hydroxylase the functional genomics era. Phytochemistry 62,
activity. Biochem. Biophys. Res. Commun. 190, 817-836.
875-880. 68. Schwab, W (2003). Metabolome diversity: Too few
56. Fahrendorflf, T. and R.A. Dixon (1993). Molecular genes, too many metabolites. Phytochemistry 62,
cloning of the elicitor-inducible cinnamic acid 837-849.
578 Kirsten A. Nielsen and Birger L. Moller

69. Koncz, C , K. Nmeth, G.P. Rdel, and J. Schell 82. Pompon, D., B. Louerat, A. Bronine and P. Urban
(1992). T-DNA insertional mutagenesis in (1996). Yeast expression of animal and plant P450
Arabidopsis. Plant Mol. Biol. 20, 963-976. in optimized redox environments. In E.F. Johnson
70. Feldmann, K. (1992). T-DNA insertion mutagenesis (ed.). Methods in Enzymology, vol. 272. Academic
in Arabidopsis: Seed infection/transformation. In Press, San Diego, CA, pp. 51-64.
C. Koncz, N.-H. Chua and J. Schell (eds), Methods 83. Bozak, K.R., D P Okeefe, and R. Christoffersen
in Arabidopsis Research. World Scientific, (1992). Expression of a ripening-related avocado
Singapore, pp. 274-289. (Persea-americana) cytochrome P450 in yeast.
71. Krysan, P.J., J.C. Young, and M.R. Sussman (1999). Plant Physiol. 100, 1976-1981.
T-DNA as an insertional mutagen in Arabidopsis. 84. Gabriac, B,, D. Werck-Reichart, H. Teutsch, and
Plant Cell 11, 2283-2290. F. Durst (1991), Purification and immunocharacter-
72. Koorneef, M., H.C. Dresselhuys, and K.S. Ramulu ization of a plant cytochrome-P450—the cinnamic
(1982). The genetic identification of translocation acid 4-hydroxylase, Arch. Biochem. Biophys. 288,
in Arabidopsis. Arab. Inf. Serv. 19, 93-99. 302-309.
73. Haughn, G.W., J. Smith, B. Mazur, and 85. Kochs, G., D. Werck-Reichhart, and H. Grisebach
C. Somerville (1988). Transformation with a (1992). Further characterization of cytochrome-
mutant Arabidopsis acetolactate synthase gene ren- P450 involved in phytoalexin synthesis in soybean:
ders tobacco resistance to sulfonylurea herbicides. cytochrome-P450 cinnamate 4-hydroxylase and
Mol. Gen. Genet. 211, 266-271. 3,9-dihydroxypterocarpan 6a-hydroxylase. Arch.
74. Shirley, B.W., S. Hanley, and H.M. Goodman Biochem. Biophys. 293, 187-194.
(1992). Effects of ionizing radiation on a plant 86. Sibbesen, O,, B. Koch, B. Halkier, and B.L. MHer
genome: Analysis of two Arabidopsis transparent (1994). Isolation of a heme-thiolate enzyme
testa mutations. Plant Cell 4, 333-347. cytochrome P450Tyr, which catalyzes the com-
75. Hemm, M.R., M.O. Ruegger, and C. Chappie mitted step in the biosynthesis of the cyanogenic
(2003), The Arabidopsis ref2 mutant is defective in glucoside dhurrin in Sorghum bicolor (L) Moench.
the gene encoding CYP83A1 and shows both Proc. Natl. Acad Sci. USA 91, 9740-9744.
phenylpropanoid and glucosinolate phenotypes. 87. Sibbesen, O., B. Koch, B.A. Halkier, and
Plant Cell 15, 179-194. B.L. MHer (1995). Cytochrome P450Tyr is a mul-
76. Weigel, D, J.H. Ahn, M.A. Blaquez, J.O. Borevitz, tifunctional heme-thiolate enzyme catalyzing the
S.K. Christensen, C. Fankhouser et al. (2000). conversion of L-tyrosine to p-hydroxyphenylacetal-
Activation tagging in Arabidopsis. Plant Physiol. doxime in the biosynthesis of the cyanogenic glu-
122,1003-1013. coside dhurrin in Sorghum bicolor (L) Moench.
77. Barnes, H.J. (1996). Maximizing expression of J. Biol. Chem. 11^, 3506-3511.
eucaryote cytochrome P450s in Escherichia coli. In 88. Koch, B., O. Sibbesen, B.A. Halkier, I. Svendsen,
Methods in Enzymology, vol. 272. Academic Press, and MHer B.L. (1995). The primary sequence of
San Diego, CA, pp. 3-14. cytochrome P450Tyr, the muhifunctional N-hydro-
78. Jones, PR., M.D. Andersen, J.S. Nielsen, PB. H^ xylase catalyzing conversion of L-tyrosine to
and B.L. MHer (2000). The biosynthesis, degrada- p-hydroxyphenylacltaldoxime in the biosynthesis
tion, transport and possible function of cyanogenic of the cyanogenic glucoside dhurrin in Sorghum
glucosides. In J.T. Romeo (ed.). Recent Advances in bicolor (L) Moench. Arch. Biochem. Biophys. 323,
Phytochemistry "Evolution of metabolic path- 177-186.
ways". Elsevier Science Ltd., pp. 191-247. 89. Halkier, B.A., H.L. Nielsen, B. Koch, and
79. Omura, T. and R. Sato (1964). The carbon monox- B.L. MHer (1995). Purification and characteriza-
ide-binding pigment of liver microsomes. II. tion of recombinant cytochrome P450tyr expressed
Solubilization, purification, and properties. J. Biol. at high levels in Escherichia coli. Arch. Biochem.
Chem. 239, 2379-2385. Biophys. 322, 369-377.
80. Jefcoate, C.R. (1978). Measurement of substrate 90. Andersen, M.D, PK. Busk, I. Svendsen, and
and inhibitor binding to microsomal cytochrome B.L. MHer (2000). Cytochromes P-450 from cas-
P-450 by optical-difference spectroscopy. In sava (Manihot esculenta Crantz) catalyzing the first
J.P. Klinman (ed.). Methods in Enzymology, vol. 52. steps in the biosynthesis of the cyanogenic gluco-
Academic Press, San Diego, CA, p. 258. sides linamarin and lotaustralin. Cloning, func-
81. Bozak, K.R., H. Yu, R. Sirevg, and tional expression in Pichia pastoris, and substrate
R.E. Christofifersen (1990). Sequence analysis of specificity of the isolated recombinant enzymes,
ripening-related cytochrome P450 cDNAs from J. Biol. Chem. 215, 1966-1975,
avocado fruit. Proc. Natl. Acad. Sci. USA 87, 91. Nielsen, J.S. and B.L. MHer (2000). Cloning and
3904-08. expression of cytochrome P450 enzymes catalysing
Cytochrome P450s in Plants 579

the conversion of tyrosine to p-hydrophenylacetal- ortologous to the sterol 14a-demethylase (CYP51)


doxime in the biosynthesis of cyanogenic gluco- from fungi and mammals. Plant J. 11, 191-201.
sides in Triglochin maritima. Plant Physiol. 122, 105. Cabello-Hurtado, F, A. Zimmerlin, A. Rahier,
1311-1321. M. Taton, R. DeRose, S. Nedelkina et al. (1997).
92. Udvardi, M.K., J.D. Metzger, V Krishnapillai, Cloning functional expression in yeast of a
W.J. Peacock, and E.S. Dennis (1994). Cloning and cDNA coding for an obtusifoliol 14a-demethylase
nucleotide sequence of a full length cDNA from (CYP51) in wheat. Biochem. Biophys. Res.
Thlaspi arvense that encodes a cytochrome P450. Commun. 230,381-385.
Plant Physiol. 104, 755-756. 106. Cabello-Hurtado, F, M. Taton, N. Forthoflfer,
93. Grove, M.D., G.F. Spencer, W.K. Rohwedder, R. Kahn, S. Bak, A. Rahier et al. (1999). Optimized
N.B. Mandava, J.E Worley, J.D. Warthen et al. expression and catalytic properties of a wheat
(1979). A unique plant growth promoting steroid obtusifoliol 14a-demethylase (CYP51) expressed
from Brassica napus pollen. Nature 281, 216-217. in yeast. Complementation of erg 11 Delta yeast
94. Bishop, G.J. and T. Yokota (2001). Plant steroid mutants by plant CYP51. Eur J. Biochem. 262,
hormones, brassinosteroids: Current highlights of 435-446.
molecular aspects on their synthesis/metabolism, 107. Fujii-Kuriyama, Y, Mizukami, Y Kawajiri,
transport, perception response. Plant Cell Physiol. K. Sogawa, M. Muramatsu (1982). Primary struc-
42, 114-120. ture of a cytochrome P450; Coding nucleotide
95. Bancos, S., T. Nomura, T. Sato, G. Molnar, sequence of phenobarbital-inducible cytochrome
G.J. Bishop, CM. Koncz et al. (2002). Regulation P450 cDNA from rat liver. Proc. Natl. Acad. Sci.
of transcript levels of the Arabidopsis cytochrome USA 79, 2793-97.
P450 genes involved in brassinosteroid biosynthe- 108. Mathur, J., G. Molnar, S. Fujioka, S. Takatsuto,
sis. Plant Physiol. 130, 504-513. A. Sakurai, T. Yokata et al. (1998). Transcription
96. Bajguz, A. and A. Tretyn (2003). The chemical of the Arabidopsis CPD gene, encoding a steroido-
characteristics distribution of brassinosteroids in genic cytochrome P450, is negatively controlled by
plants. Phytochemistry 62, 1027-1046. brassinosteroids. Plant J. 14, 593-602.
97. Thummel, C.S. and J. Chory (2002). Steroid in 109. Bishop, G.J., T Nomura, T Yokota, K. Harrison,
plants and insects—common themes, different T. Nogushi, S. Fujioka et al. (1999). Tomato dwarf
pathways. Genes Dev. 16, 3113-3129. enzymes catalyzes C-6 oxidation in brassino-
98. Li, J., J. Wen, K.A. Lease, J.T. Doke, F.E. Tax and steroid biosynthesis. Proc. Natl. Acad. Sci. USA
J.C. Walker (2002). BAKl, an Arabidopsis LRR 96, 1761-1766.
receptor-like protein kinase, interacts with BRIl 110. Conn, E.E. (1980). Cyanogenic compounds. Ann.
and modulates brassinosteroid signalling. Cell Rev. Plant Physiol. 31, 4 3 3 ^ 5 1 .
110,213-222. 111. Lechtenberg, M. and A. Nahrstedt (1995).
99. Nakashita, H., M. Yasuda, Nitta, T., T. Asami Cyanogenic glucosides. In Ikan (ed.), Naturally
S. Fujioka, Y Aral et al. (2003). Brassinosteroid Occurring Glucosides. John Wiley & sons Ltd.,
functions in a broad range of disease resistance in Chichester, UK, pp. 147-191.
tobacco rice. Plant J. 33, 887-898. 112. MBer, B.L. and D.S. Seigler (1999). Biosynthesis
100. Fujioka, S. and A. Sakurai (1997). Biosynthesis of cyanogenic glycosides, cyanolipids, and related
and metabolism of brassinosteroids. Physiol. compounds. In B.K. Singh (ed.). Plant Amino
Plant. 100,710-715. Acids. Marcel Dekker Inc., New York, pp. 563-609.
101. Fujioka, S. and A. Sakurai (1997). Bras- 113. Seigler, D. (1998). Cyanogenic glucosides and
sinosteroids. Nat. Prod. Rep. 14, 1-10. cyanolipids. In D. Seigler (ed.), Plant Secondary
102. Nogushi, T, S. Fujioka, S. Choe, S. Takatsuto, Metabolism. Klii/er academic Press, Norwell,
EE. Tax, S. Yoshida et al (2000). Biosynthetic MA, pp. 273-299.
pathways of brassinolide in Arabidopsis. Plant 114. Tylleskar, T., M. Banea, N. Bikangi, R.D. Cooke,
Physiol. 124, 201-209. N.H. Poulter, and H. Rosling (1992). Cassava
103. Kahn, R.A., S. Bak, C.E. Olsen, I. Svendsen, cyanogens and konzo, an upper motoneuron dis-
B.L. MHer (1996). Isolation and reconstitution of ease found in Africa. Lancet 339, 208-221.
the heme-thiolate protein obtusifoliol 14a-methyl- 115. Swantson, J.S., WTB. Thomas, W Powell,
ase from Sorghum bicolor (L). Moench. J. Biol. G.R. Young, RE. Lawrence, L. Ramsey et al.
Chem. 211, 32944-32950. (1999). Using molecular markers to determine bar-
104. Bak, S., R.A. Kahn, C.E. Olsen and B.A. Halkier. ley most suitable for malt whisky distilling. Mol.
(1997). Cloning and expression in Escherichia Breed. 5, 103-109.
coli of the obtusifoliol 14a-demethylase of 116. Jones, D.A. (1998). Why are so many plants
Sorghum bicolor (L.) Moench, a cytochrome P450 cyanogenic? Phytochemistry 47, 155-162.
580 Kirsten A. Nielsen and Birger L. Moller

117. Koukol, J., P. Miljanich E.E. Conn (1962). The cytochrome P450 enzymes CYP79A1 and
metabolism of aromatic compounds in higher CYP71E1 involved in the biosynthesis in Sorghum
plants. VI: Studies on the biosynthesis of dhurrin, bicolor. Arch. Biochem. Biophys. 363, 9-18.
the cyanogenic glucoside of Sorghum vulgare. 128. Jones, PR., B.L., MBer, and PB. Hp (1999).
J. Biol. Chem. 237, 3223-3228. The UDP-glucose: p-hydroxymandelonitrile-o-
118. Uribe, E.G. and E.E. Conn (1966). The metabo- glucosyltransferase that catalyzes the last step in
lism of aromatic compounds in higher plants. VII synthesis of the cyanogenic glucoside dhurrin
The origin of the nitrile nitrogen atom of dhurrin in Sorgum bicolor J. Biol. Chem 274, 35483-35491.
(B-D-glucopyranosyloxy-L.p-hydroxymandeloni- 129. Tattersall, D.B., S. Bak, PR. Jones, C.E. Olsen,
trile). J. Biol. Chem. 241, 92-94. J.K. Nielsen, and M.L. Hansen (2001). Resistance
119. Farnden, K.J.F., M.A. Rosen and D.R, Liljegren to an herbivore through engineered cyanogenic
(1973). Aldoximes and nitriles as intermediates in glucoside synthesis. Science 293, 1826-1828.
the biosynthesis of cyanogenic glucosides. 130. Paquette, S.M., B.L. MHer, and S. Bak (2003). On
Phytochemistry 12, 2673-2677. the origin of family 1 plant glycosyltransferases.
120. Conn, E.E. and G.W. Butler (1969). The bio- Phytochemistry 62, 399^13.
synthesis of cyanogenic glucosides and other 131. Bak, S., C.E. Olsen, B.A. Halkier, and B.L. MHer
simple nitrogen compounds. In B. Harborne and (2000). Transgenic tobacco and Arabidopsis plants
T. Swain (eds), Perspectives in Phytochemistry. expressing the two multifunctional sorghum
Academic Press, London and New York, pp. 47-74. cytochrome P450 enzymes, CYP79A1 and
121. McFarlane, I.J., E.M. Lees and E.E. Conn (1975). CYP71E1, are cyanogenic and accumulate
The in vitro biosynthesis of dhurrin, the metabolites derived from intermediates in dhurrin
cyanogenic glucoside of Sorghum bicolor J. Biol. h\osynt\iQS\s. Plant Physiol. 123, 1437-1448.
C/zem. 250,4708-4714. 132. Winkel-Shirley, B. (2001). Flavonoid biosynthesis:
122. MHer, B.L. and E.E. Conn (1980). The biosynthe- A colourful model for genetics, biochemistry, cell
sis of cyanogenic glucosides in higher plants. biology, and biotechnology. Plant Physiol. 126,
N-hydroxytyrosine as an intermediate in the 485^93.
biosynthesis of dhurrin by Sorghum bicolour 133. Rasmussen, S. and R.A. Dixon (1999). Transgene-
(Linn) Moench. J. Biol. Chem. 254, 8575-8583. mediated and elicitor-induced pertubation of
123. Kahn, R.A., S. Bak, I. Svendsen, B.A. Halkierand metabolic channelling at the entry point into
B.L. Moller (1997). Isolation and reconstitution of the phenylpropanoid pathway. Plant Cell 11,
cytochrome P450ox and in vitro reconstitution of 1537-1551.
the entire biosynthetic pathway of the cyanogenic 134. BurbuHs, I.E. and B. Winkel-Shirley (1999).
glucoside dhurrin from sorghum. Plant Physiol. Interactions among enzymes of the Arabidopsis
115, 1661-1670. flavonoid biosynthetic pathway. Proc. Natl. Acad.
124. Halkier, B.A., J. Lykkesfeldt and B.L. MHer Sci. USA 96, 12929-12934.
(1991). 2-Nitro-3(p-hydroxyphenyl)propionate and 135. Winkel-Shirley, B. (1999). Evidence for enzyme
aci-l-nitro-2-(p-hydroxyphenyl)ethane, two inter- complexes in the phenylpropanoid and flavonoid
mediates in the biosynthesis of the cyanogenic glu- paihv/ays. Plant Physiol. 107, 142-149.
coside dhurrin in Sorghum bicolor (L) Moench. 136. Chalfie, M., Y Tu, G. Euskirchen, WW. Ward, and
Proc. Natl. Acad. Sci. USA 88, 487^91. D.C. Prasher (1994). Green fluorescent protein as a
125. Bak, S., R.A. Kahn, H.L. Nielsen and B.L. MHer marker for gene-expression. Science 263, 802-805.
(1998). Cloning of three A-type cytochromes 137. Chalfie, I. and S. Kain (1998). Green fluorescent
P450, CYP71E1, CYP98, and CYP99 form protein. Properties, Applications and Protocols.
Sorghum bicolor (L) Moench by a PCR approach Wiley-Liss Inc; New York, p. 385.
and identification by expression in Escherichia 138. Hosel, W and A. Nahrstedt (1980). In vitro biosyn-
coli of CYP71E1 as a multifunctional cytochrome thesis of the cyanogenic glucoside taxiphyllin in
P450 in the biosynthesis of the cyanogenic gluco- Triglochin maritima. Arch. Biochem. Biophys. 203,
side dhurrin. Plant Mol. Biol. 36, 393-405. 753-757.
126. Halkier, B.A. and B.L. MMer (1990). The biosyn- 139. Nielsen, J. and B.L. Mtier (1999). Biosynthesis of
thesis of cyanogenic glucosides in higher plants. cyanogenic glucosides in Triglochin maritima and
Identification of three hydroxylation steps in the the involvement of cytochrome P450 enzymes.
biosynthesis of dhurrin in Sorghum bicolor (L) Arch. Biochem. Biophys. 368, 121-130.
Moench and the involvement of l-ac/-nitro-2- 140. Koch, B., VS. Nielsen, B.A. Halkier, C.E. Olsen,
(/7-hydroxyphenyl)ethane as an intermediate. J. and B.L. MBer (1992). The biosynthesis of cyano-
Biol. Chem. 265, 21114-21121. genic glucosides in seedlings of cassava {Manihot
127. Kahn, R.A., T Fahrendorf, B.A. Halkier, and esculenta Crantz.). Arch. Biochem. Biophys. 292,
B.L. Mfler (1999). Substrate specificity of the 141-150.
Cytochrome P450s in Plants 581

141. Lykkesfeldt, J., B.L. MHer (1994). Cyanogenic monooxygenases induced in response to safeners
glucosides in cassava, Manihot esculenta Crantz. and bacterial pathogens. Plant Physiol. 125,
Acta Chem. Scand. 48, 178-180. 1126-1138.
142. Lykkesfeldt, J. and B.L. MBer (1995). On the 156. Zhou, N., T.L. Tootle, and J. Glazebrook (1999).
absence of 2-(2'-cyclopentenyl)glycine-derived Arabidopsis PAD3, a gene required for camalexin
cyanogenic glycosides in cassava, Manihot esculenta biosynthesis encodes a putative cytochrome P450
Crantz. Acta Chem. Scand. 49, 540-542. monooxygenase. Plant Cell 11, 2419-2428.
143. Collinge, D. and M.A. Hughes (1982). In vitro 157. Schroeder, G., E. Unterbusch, M. Kaltenbach,
characterization of the Ac locus in white clover J. Schmidt, D. Strack, V De Luca et al. (1999).
(Trifolium repens L). Arch. Biochem. Biophys. 218, Light-induced cytochrome P450-dependent enzyme
38-45. in indole alkaloid biosynthesis: Tabersonine 6-
144. Collinge, D. and M.A. Hughes (1984). Evidence hydroxylase. FEBS Lett. 458, 97-102.
that linamarin and lotaustralin, the two cyanogenic 158. Fits, L. and J. Memelink (2000). 0RCA3, a
glucosides of Trifolium repens L., are synthesized jasmonate-responsive transcriptional regulator of
by a single set of microsomal enzymes controlled plant primary and secondary metabolism. Science
by the Ac/ac locus. Plant Sci. Lett. 34, 119-125. 289,295-297.
145. Hahlbrock, K. and E.E. Conn (1971). Evidence for 159. Irmler, S., G. Schrder, B. St-Pierre, N.P Crouch,
the formation of linamarin and lotaustralin in flax M. Hotze, J. Schmidt et al. (2000). Indole alkaloid
seedlings by the same glycosyltransferase. biosynthesis in Catharanthus roseus: New enzyme
Phytochemistry 10, 1019-1023. activities and identification of cytochrome P450
146. Nielsen, K.A., C.E. Olsen, K. Pontoppidan, and CYP72A1 as secologanin synthase. Plant J. 24,
B.L. MHer (2002). Leucine-derived cyano gluco- 797-804.
sides in barley. Plant Physiol. 129, 1066-1075. 160. Aerts, R.J., D. Gisi, E. De Carohs, V De Luca, and
147. Halkier, B.A. and L. Du (1997). The biosynthesis T.W. Bauman (1994). Methyl jasmonate vapor
of glucosinolates. Trends Plant Sci. 2, 425^31. increases the developmentally controlled synthesis
148. Bak, S., C.E. Olsen, B.L. Petersen, B.L. MBer, of alkaloids in Catharanthus and Cinchona
and B.A. Halkier (1999). Metabolic engineering of seedlings. Plant J. 5, 635-643.
p-hydroxybenzylglucosinolate in Arabidopsis by 161. Ralston, L., S.T. Kwon, M. Schoenbeck, J. Ralston,
expression of the cyanogenic CYP79A1 from D.J. Schenk, R.M. Coates et al. (2001). Cloning,
Sorghum bicolor. Plant J. 20, 663-671. heterologous expression, fiinctional characteriza-
149. Frey, M., P Chomet, E. Glaswischnig, C. Stettner, tion of 5-epi-aristolochene-l,3-dihydroxylase from
S. Grii, A. Winkmair et al. (1997). Analysis of a tobacco (Nicotiane tabacum). Arch. Biochem.
chemical plant defense mechanism in grasses. Biophys. 393, 222-235.
Science 111, 696-699. 162. Greenhagen, B.T., P. Griggs, S. Takahashi,
150. Gierl, A. and M. Frey (2001). Evolution of benz- L. Ralston, and J. Chappell (2003). Probing
oxazinone biosynthesis and indole production in sesquiterpene hydroxylase activities in a coupled
maize. Planta 213, 493-498. assay with terpene synthases. Arch. Biochem.
151. Frey, M., K. Huber, W.J. Park, D. Sicker, Biophys. 409, 385-394.
P Lindberg, R.B. Meeley et al (2003). A 2-oxoglu- 163. O'Donohue, M.J., H. Gousseau, J.C. Huet, D. Tepfer,
tarate-dependent dioxygenase is integrated in DIM- and J.C. PemoUet (1995). Chemical synthesis,
BOA-biosynthesis. Phytochemistry 62, 371-376. expression and mutagenesis of a gene encoding beta-
152. Glawischnig, E., S. Grii, M. Frey, A. Gierl (1999). cryptogein, an elicitin produced by Phytophtera-
Cytochrome P450 monooxygenases of DIBOA cryptogea. Plant Mol. Biol. 27, 577-586.
biosynthesis. Specificity and conservation among 164. Latunde-Dada, A.O., F Hurtado-Cabello,
grasses. Phytochemistry 50, 925-930. N. Czittish, L. Didierjean, C. Schopfer, N. Hertkorn
153. Nomura, T., A. Ishihara, H. Imaishi, T.R. Endo, et al. (2001). Flavonoid 6-hydroxylase from soy-
H. Ohkawa, and H. Iwamura (2002). Molecular bean (Glycine max L.) a novel plant P-450
characterization and chromosomal localization of monooxygenase. J Biol. Chem. 276, 1688-1695.
cytochrome P450 genes involved in the biosynthe- 165. Lupien, S., F Karp, M. Wildung, R. Croteau (1999).
sis of cyclic hydroxamic acids in hexaploid wheat. Regiospecific cytochrome P450 limonene hydroxy-
Mol. Gen. Genet. 261, 210-217. lases from mint (Mentha) species. cDNA isolation,
154. Frey, M., C. Stettner, PW. Pare, E.A. Schmelz, characterization, and functional expression of (-)-4S-
J.H. Tumlinson, and A. Gierl (2000). An herbivore limonene-3-hydroxylase and (-)-4S-limonene-6-
elicitor activates the gene for indole emission in hydroxylase. ^rc/i. Biochem. Biophys. 368,181-192.
maize. Proc. Natl. Acad Sci. USA 97,14801-14806. 166. Haudenschild, C , M. Schalk, F Karp and
155. Persans, M.W., J. Wang, and M.A. Schuler (2001). R. Croteau (2000). Functional expression of
Characterization of maize cytochrome P450 regiospecific cytochrome P450 limonene
582 Kirsten A. Nielsen and Birger L. Moller

hydroxylases from Mint {Mentha spp.) in methionine :2,4,7' -trihydroxyisoflavanone 4' -O-
Escherichia coli and Saccharomyces cerevisiae. methyltransferase, a critical enzyme of the legume
Arch. Biochem. Biophys. 379, 127-136. isoflavonoid phytoalexin pathway. Plant Cell
167. Wat, M., D.B. Little, M. Schalk, and R. Croteau Physiol. 44, 103-112.
(2001). Hydroxylation of limonene enantiomers 180. Chou, W.M. andT.M. Kutchan (1998). Enzymatic
and analogs by recombinant (-)-limonene 3- and oxidations in the biosynthesis of complex alka-
6-hydroxylases from mint (mentha) species: loids./^/aw/^ 15,289-300.
Evidence for catalysis within sterically constrained 181. Rueflfer, M. and M.H. Zenk (1994). Canadine syn-
active sites, ^rc/?. Biochem. Biophys. 387,125-136. thase from Thalictrum tuberosum cell cultures cat-
168. Fitzpatrick, L.A. (2003). Soy isoflavones: Hope or alyzes the formation of the methylenedioxy bridge in
hype? Maturitas 44(suppl), S21-S29. berberine synthesis. Phytochemistry 36, 1219-1223.
169. Liu, C.J., J.W. Blount, C.L. Steele, and R.A. Dixon 182. De-Eknamkul, W, T. Tanahashi, and M.H. Zenk
(2002). Bottlenecks for metabolic engineering of (1992). Enzymatic 10-hydroxylationand 10-O-methyl-
isoflavone glycoconjugates in Arabidopsis. Proc. ation of dihydrosanguinarine in dihydrochelirubine
Natl. Acad. Sci. USA 99, 14578-14583. formation by Eschscholtia. Phytochemistry 31,
170. Akashi, T., T. Aoki, and S. Ayabe (1999). Cloning 2713-2717.
and functional expression of a cytochrome P450 183. Kammerer, L., W. De Eknamkul, and
cDNA encoding 2-hydroxy-isoflavanone synthase M.H. Zenk (1994). Enzymatic 12-hydroxylation
involved in biosynthesis of the isoflavonoid skele- and 12-0-methylation of dihydrochelirubine in
ton in licorice. Plant Physiol. Ill, 821-828. dihydromacarpine formation by Thalictrum bul-
171. Jung, W., O. Yu, S.M.C. Lau, D.P. O'Keefe, garicum. Phytochemistry 36, 1409-1416.
J. Odell, G. Fader et al. (2000). Identification 184. Blechert, S., W. Brodschelm, S. HMer,
expression of isoflavone synthase, the key enzyme L. Kammerer, T.M. Kutchan, M.J. Mueller et al.
for biosynthesis of isoflavones in legumes. Nat. (1995). The octadecanoic pathway: Signal
Biotech. 18,208-212. molecules for the regulation of secondary
172. Yu, O., W. Jung, J. Shi, R.A. Croes, G.M. Fader, pathways. Proc. Natl. Acad. Sci. USA 92,
S. McGonigle et al. (2000). Production of the 4099^105.
isoflavones genistein daidzein in non-legume dicot 185. Kutchan, T. and J. Schrder (2002). Selected cell
and monocot tissues. Plant Physiol. 124, 718-793. cultures and induction methods for cloning and
173. Koornneef, M. (1990). Mutations affecting the assaying cytochromes P450 in alkaloid pathways.
testa color in Arabidopsis. Arab. Inf. Serv. 27, 1 ^ . Meth. Enzymol. 357, 370-381. (E.F Johnson and
174. Ohio State University Arabidopsis Biological M.R. Waterman, eds.)
Resource Center: www.biosci.ohio-state.edu/ 186. Grothe, T., R. Lenz and T. Kutchan (2001).
~plantbio/Facilities/abrc/abrchome.htm Molecular characterization of the salutaridionol 7-
175. Shirley, B.W., W.L. Kubasek, G. Storz, O-acetyltransferase involved in morphine biosyn-
E. Bruggemann, M. Koornneef, F.M. Ausubel et al. thesis in opium poppy Papaver somniferum.
(1995). Analysis of Arabidopsis mutants deficient J. Biol. Chem. 276, 30717-30723.
in flavonoid biosynthesis. Plant J. 8, 659-671. 187. Petersen, B.L., E. Andrason, S. Bak, N. Agerbirk,
176. Liu, C.J. and R.A. Dixon (2001). Elicitor-induced and B.A. Halkier (2001). Characterization of trans-
association of isoflavone O-methyltransferase with genic Arabidopsis thaliana with metabolically
endomembranes prevents the formation of 7-0- engineered high levels of p-hydroxybenzylglucosi-
methylation of daidzein during isoflavonoid phy- nolate. Planta lU, 612-618.
toalexins biosynthesis. Plant Cell 13, 2643-2658. 188. Zhao, Y, A.K. Hull, N.R. Gupta, K.A. Goss,
177. Mondolo, L.V, FQ. Cunha, M.R. Braga, I. Salgado J. Alonso, J.R. Ecker et al. (2002). Trp-dependent
(2002). Nitric oxide synthase-mediated phytoalexin auxin biosynthesis in Arabidopsis: involvement of
accumulating in soybean cotelydons in response to cytochrome P450s CYP79B2 CYP79B3. Gen.
the Diaporthe phaseolorum f sp. Meridionalis elic- Devel. 16,3100-3112.
itOT. Plant Physiol. 130, 1288-1297. 189. Bak, S., FE. Tax, K.A. Feldman, D.W. Galbraith,
178. Li, H., C.S. Raman, R Martasek, B.S.S. Masters, and R. Feyereisen (2001). CYP83B1, a cytochrome
and T. Poulos (2001). Crystallographic studies on P450 at the metabolic branchpoint in auxin and
endothelial nitric oxide synthase complexed with indole glucosinolate biosynthesis in Arabidopsis
nitric oxide and mechanism-based inhibitors. thaliana. Plant Cell 13, 101-111.
Biochemistry 40, 5399-5406. 190. Harboume, IB. and H. Baxter (1999). The Handbook
179. Akashi, T., Y. Sawada, N. Shimada, N. Sakurai, of Natural Flavonoids, vol. 1 Wiley, Chichester, UK.
T. Aoki, and S. Ayabe (2003). cDNA cloning and 191. Sefton, M.A., I.L. Francis, and PJ. Williams
biochemical characterization of S-adenosyl-L- (1993). The volative composition of chardonnay
Cytochrome P450s in Plants 583

juices. A study by flavour precursor analysis. Am. 195. Bell, C.A., R.A. Dixon, A.D. Farmer, R. Flores,
J. Enol. Vitil 44, 359-370. J. Inman, R.A. Gonzales et al. (2000). The med-
192. Sefton, M.A., I.L. Francis, and P.J. Willians (1994). icago genome initiative: A model legume database.
Free and bound volative secondary metabolites of Nucleic Acid Res. 29, 1-4.
Vitis vivifera grape cv Sauvignon blanc. J. Food 196. Huhman, D.V and L.W. Sumner (2002). Metabolic
Sci. 59, 142-147. profiling of saponins in Medicago sativa and
193. Frick, S. and T.M. Kutchan (1999). Molecular Medicago truncatula using HPLC coupled to
cloning and functional expression of 0-methyl- an electrospray ion-trap mass spectrometer.
transferases common to isoquinoline alkakoid Phytochemistry 59, 347-360.
and phenylpropanoid biosynthesis Plant J. 17, 197. Suzuki, H., L. Achinine, R. Xu, S.RT. Matsuda,
329-339. and R.A. Dixon (2002). A genomics approach to
194. Hansen, K.S., C. Kristensen, D.B. Tattersall, the early stages of triterpene saponin biosynthesis
P.R. Jones, C.E. Olsen, S. Bak et al (2003). The in Medicago truncatula. Plant J. 32, 1033-1048.
in vitro substrate regiospecificity of UGT85B1, the 198. Nelson, D.R. and H.W. Strobel (1988). On the
cyanohydrin glucosyltransferase from Sorghum membrane topology of vertebrate cytochrome
bicolor Phytochemistry. 64, 143-151. P-450 Proteins. J. Biol. Chem. 263, 6038-6050.
13
The Diversity and Importance of
Microbial Cytochromes P450
Steven L. Kelly, Diane E. Kelly, Colin J. Jackson,
Andrew G.S. Warriiow, and David C. Lamb

The cytochromes P450 (CYPs) of microbes are ranging from new therapeutics to biotransforma-
enormously diverse as revealed in discoveries from tions and bioremediation.
the era of molecular biology and as subsequently
found in genomic investigations. One percent of the
genes of a microbe can encode CYPs, but in stark
contrast most bacteria studied so far can survive 1. Introduction to Microbial
without CYPs. Microbial eukaryotes usually have CYPs
at least one CYP, due to the essential requirement of
most to synthesize sterol involving CYP51, sterol The discovery of cytochromes P450 in mam-
14a-demethylase. The roles of the vast majority of malian tissues rich with these proteins, such as liver
microbial CYPs remain to be elucidated, but many and the adrenal gland, resulted in intense scrutiny
already have important fundamental roles in nature, of their roles in xenobiotic metabolism and endoge-
and others are important for biotechnological pur- nousfiinctions^"^.Following their discovery came
poses. Some others have, of course, provided facile the realization that mammalian proteins required
models for understanding CYP structure and activ- electron donor systems for activity, either NADPH-
ity, such as CYPlOl (P450(3^j^) of Pseudomonas cytochrome P450 reductase (CPR) or adrenodoxin
putida and CYP102A1 (P450gj^_3) of Bacillus and adrenodoxin reductase in the endoplasmic
megaterium. The purpose of this chapter is to pro- reticulum or the mitochondria respectively^' ^.
vide an outline of the important biomedical and Protein biochemistry and molecular biology
environmental roles of the microbial CYPs, includ- revealed the multiplicity of CYP forms and mam-
ing many which were unsuspected when the respec- malian genomes exhibited CYP diversity.
tive microorganisms were originally studied. This The microbial CYP systems were studied at
includes involvement of CYP in some of the earli- the same time, with yeast CYP being reported by
est metabolic alterations in the production of peni- Lindenmeyer and Smith (1964) and bacterial CYP
cillin, some of the early biosynthetic steps allowing by Appleby (1967)^' ^. The systems were viewed
the production of corticosteroids, and the first as models and this was true especially for a CYP
application of therapeutic CYP inhibitors, the azole from P. putida called P450CAM (CYPlOl) that
antifungal agents. Current and future applications allowed this bacterium to grow on camphor as a
involving microbial CYPs are manifestly clear. carbon source^"^^. In pioneering work from the

Steven L. Kelly, Diane E. Kelly, Colin J. Jackson, Andrew G.S. Warriiow, and David
C. Lamb • Wolfson Laboratory of P450 Biodiversity, Swansea Clinical School, University of Wales Swansea,
Swansea, Wales, UK.
Cytochrome P450: Structure, Mechanism, and Biochemistry, 3e, edited by Paul R. Ortiz de Montellano
Kluwer Academic / Plenum Publishers, New York, 2005.

585
586 Steven L. Kelly et al.

Gunsalus laboratory, the P450(^^j^ system allowed undertake C22-desaturation^^. Ergosterol in yeast
biochemical and biophysical investigation of the has been used to produce vitamin D2, although
CYP catalytic cycle as well as of the genetics this has been uneconomical recently, but manipu-
of a bacterial catabolic plasmid. This typical CYP lation of this pathway has allowed production in
system was found to require a ferredoxin and whole-cells of hydrocortisone^^.
ferredoxin reductase for catalytic activity, unlike The modern era of biotechnology began with
the model for eukaryote CYPs, CYP 102A1 or the discovery of antibiotics and the steps taken to
P450gj^ 3, which was discovered in the Fulco improve yield. We now realize that streptomycetes
laboratory and consisted of a fusion polypeptide contain many CYPs for drug (secondary meta-
containing CYP and reductase domains'^. bolism) synthesis and this is discussed later in
Studies in yeast have revealed deep insights more detail, but Penicillium chrysogenum was the
into eukaryotic processes, and this is also true in first utilized in antibiotic production. In early
studies on CYPs, where the first microbial CYP work following the pioneering studies, phenyl-
cloned was found to undertake an ancestral role in acetate (precursor) feeding was found to elevate
the superfamily That is, CYP51 is needed for yields from fermentations and, of course, muta-
sterol biosynthesis and is found in plants, fungi, tion and screening strategies increased the titer.
protists, animals, and some bacteria^'^. Recently, the basis of genetic change in the
The nomenclature for CYPs is based on amino Wisconsin strains revealed that a CYP mutation
acid identity with 40% identity and above needed produced increased penicillin yield at the begin-
to place CYPs in the same family and more than ning of the genesis of improved fungal strains^^.
55% to place them in the same subfamily^^. These The gene concerned, PahA encodes CYP504,
rules can be relaxed, as is the case for CYP51s that a CYP also identified in Aspergillus nidulans that
can fall below 40% identity, if the CYPs under- allows growth on phenylacetate'^. A CYP504
take the same function. For microbial eukaryotes, mutant containing the substitution LI8IF resulted
the family numbers 51-69 and 501-699 are avail- in the reduced 2-hydroxylation of phenylacetate,
able and at the time of writing, numbers up to and this mutation channeled the carbon flux away
CYP553 are listed, but each genome reveals many from the side-pathway and through into increased
more and many are not yet assigned. Bacterial penicillin titer.
CYP family numbers are initiated at CYPlOl and Parallel with developments after World War II
a similar expanding scenario can be envisaged in antibiotic production, the therapeutic value of
with more and more genomes. corticosteroids was discovered. Interest during the
In this chapter, we will outline historical early 1940s was based on rumors of experiments
perspectives on the discovery and importance of in performance enhancement of pilots in the
CYPs in biotechnology before going on to Luftwaffe by corticosteroids^^. The chemical syn-
describe the diversity of functions and activities thesis route was inefficient and microbial hydrox-
associated with microbial CYPs. The coverage is ylations by fungi were some of the first successful
relatively extensive and is illustrative of the field, biotransformations for pharmaceutical produc-
but with so many CYPs now revealed it is impos- tion. 1 la-Hydroxylation of a steroid was achieved
sible to discuss each one individually. Obviously by Aspergillus ochraceous and Rhizopus niger,
many of the CYPs that remain orphan in function and the 11 p-hydroxylation achieved with other
today will emerge as being important in future fungi such as Cochliobolous lunatus, allowing the
studies. production of Cortisol. We now realize these con-
Microbial science is generally reported to versions were achieved by fungal CYPs, although
begin with the fermentation of yeast observed by the genes concerned are not yet known.
Pasteur, and although yeast CYP is not a A last example to note before moving onto
cytochrome involved in respiration, it does con- describing the diversity of microbial CYPs and
tribute to the osmotic robustness of the microor- their importance, is provided by the azole antifun-
ganism, including ethanol tolerance, through the gal compounds^ ^ First developed for agriculture,
synthesis of ergosterol. This product requires where they are known as DMI compounds
CYP51 to remove the C14-methyl group of the (demethylase inhibitors), these compounds have
precursor as well as a second CYP, CYP61, to become central to antifungal therapy in the clinic
The Diversity and Importance of IViicrobial Cytochromes P450 587

A. Mutation in CYP504 leads to overproduction of penicillin in Penicillium chrysogenum

CH2COOH CH2COOH
I HO I Fumarate
Benzylpenicillin -. _CYP504
+
Acetoacetate
O2 H2O

B. Commercial application of P450 catalysed lla-hydroxylation of progesterone in hydrocortisone


biosynthesis Q

-- Hydrocortisone

C. CYP51 and sterol biosynthesis, application to azole antifungal development

HCOOH

Figure 13.1. (A) The metabolism of phenylacetate by Penicillium chrysogenum by CYP504 is impeded
during early strain improvement by mutation and selection increasing flux through to penicillin production; (B) The
11-hydroxylation of steroid by some filamentous fungi were among the first commercial biotransformations in
the pharmaceutical industry allowing the production of corticosteroids, first lla-hydroxylation by Aspergillus sp.
and Rhizopus sp. and subsequently 11 P-hydroxylation by Curvularia sp.; (C) Sterol C14-demethylation in fungi is
the target of the antifungal azole compounds.

and new compounds continue to be evaluated. The Chapter 4). Over the last 20 years other novel
mode of action in relation to CYP51 and forms have been identified including the fusion
the repercussions of that for the fungus only protein CYP102A1 (P450gj^_3) identified in
became clear after their development, but they Bacillus megaterium by the Fulco laboratory^^' ^^.
were the first commercial CYP inhibitors. Some This CYP resembles the class II system with a
of the microbial CYP activities described here are flavoprotein reductase domain while CYP55
shown in Figure 13.1. (P450j^Qj.) is a stand-alone catalytic entity with an
NADPH-binding site and a third class^^. Over the
last years, several new types have been identified
2. Classes of Microbial CYPs which, in some cases, have been placed into
classes. One response to this diversity is to have
CYPs in bacteria are generally soluble proteins new classes for each new form and to not disrupt
requiring ferredoxin and ferredoxin reductase for the assignments made already, and we have previ-
the two electrons needed in the CYP catalytic ously adopted that approach^^. However, with the
cycle, while CYPs in eukaryotic microbes are typ- emergence of further forms of CYP fusion pro-
ically located in the endoplasmic reticulum with teins, and the anticipation that more will arise, it
an associated NADPH-CPR providing the neces- is a suggestion here that classes should reflect
sary reducing equivalents. As such, these are often novelty only in the method CYP reduction. In this
called class I and class II, respectively (see also way, CYP fUsion proteins involving ferredoxin
588 Steven L. Kelly et al.

and ferredoxin reductase would be subclasses of mononucleotide and NADH-binding domains)


class I, while CYP fusions involving only flavo- and a C-terminal ferredoxin center (2Fe2S) and
proteins would be subclasses of class II. the end of the polypeptide^^. This catalytically
Catalytically self-sufficient CYPs represent class self-sufficient enzyme will be of interest for bio-
III. Different subclasses can occur for the CYPs technological modifications and directed evolu-
involved, but class I and class II would give the tion studies, but as it contains a ferredoxin system,
immediate impression of the type of electron it could be placed within class lb. A CYP51 has
transfer system concerned. been observed in Methylococcus capsulatus fused
A number of new CYP fusion forms and a to a C-terminal ferredoxin domain and unless this
novel CYP operon have been cloned at the time of has a novel reductase partner, it can be considered
writing that represent new forms. The gene encod- a Class Ic form as it conforms to the ferredoxin/
ing CYP176A1 (P450^.j^) was found in an operon ferredoxin reductase model (Figure 13.2)^^.
with genes encoding a flavodoxin and flavodoxin A further CYP gene has also been cloned that
reductase and so could represent an ancestor of the represents a new form. It confers a capability to
class II system. These would be placed as a class metabolize the high explosive hexahydro-1,3,5-
lie after CYP102A1, class Ilb^^. The flavodoxin trinitro-l,3,5-triazine and has been identified in
and flavodoxin reductase could have become Rhodococcus rhodochrous, with a flavodoxin
fused in other class II systems. Also a novel CYP domain at its N-terminus, but appears also to need
was identified from a Rhodococcus sp. containing a ferredoxin reductase for activity, which may be
a reductase domain at the C-terminus similar to encoded adjacent to the CYP gene^^. As this gene
dioxygenase reductase protein (containing flavin contains a flavodoxin domain, this can be placed

CLASS I

(a)

(b) (b) ( ^ ^ N H .
HOOC

(c)
(c)

HOOC CLASS ni-P450nor—P450nor

Figure 13.2. A proposal for a simplified classification of CYPs with reference to either use of a ferredoxin or
alternative electron transport mechanism. Class la—^typical bacterial system supported by ferredoxin and ferredoxin
reductase, for example, CYPlOl; Class lb—Rhodococcus sp. CYP fusion protein containing a ferredoxin domain^^;
Class Ic—Methylococcus capsulatus CYPS 1-ferredoxin fusion^^. Class Ila—^typical eukaryotic CYP/NADPH-
cytochrome P450 reductase system; Class lib—a fusion protein of a CYP and flavoprotein reductase, for example,
P450gj^ 3; Class lie—P450^-j^ containing separate flavodoxin and flavodoxin reductase partners-^^. Class III—stand-
alone functional CYPs, for example, P450j^^j.^^.
The Diversity and Importance of IVIicrobial Cytochromes P450 589

with the Class II CYP systems if substantiated by secondary metabolites, and in detoxification, and
protein studies. Another CYP has been detected in it is the general view that these selective pressures
Ralston metallidurans and has a C-terminal fusion have produced much of the observed CYP diver-
to a phthalate family oxygenase reductase module, gjlyi4,30-32 However, some bacteria contain many
but appears to be similar to another fusion CYPs, including Mycobacterium smegmatis that
described above^^' ^^. contains approximately 39 CYP genes, that is,
about 1% of all genes in this microorganism,
together with additional CYP pseudogenes^^.
CYPs have a role in bacteria to enable growth
3. Considering the Origins and
on carbon sources in the environment, such as on
Relatedness of Microbial camphor by P putida containing CYPlOl, or to
CYPs undertake secondary metabolism as part of the
biochemical warfare between organisms. Many
Clues to the basic functions and the origins of CYPs are found in the pathways s)aithesizing
CYPs can be found among the microorganisms. important therapeutic compounds and this is dis-
Among eighteen archaebacterial genomes probed, cussed later in this chapter. The number of vital
five contained CYP genes. Similarly, about two endogenous steps that have evolved for bacterial
thirds of proteobacterial genomes (28 among 90 CYP function is still very small; for instance, Biol
genomes probed), such as Escherichia coli, con- in Bacillus subtilis is needed to produce biotin
tain no CYPs, indicating how nonessential CYPs by a pathway distinct from other bacteria^"*.
are to basic metabolism. This correlates with the Some CYPs, among the numerous forms uncov-
involvement of CYPs in mechanisms of deter- ered, must be anticipated to be involved in key
rence and attraction through the production of areas of endogenous metabolism especially in the

Table 13.1. The Numbers of CYPs in Various Sequenced


Microbial Genomes

Microorganism CYP complement (CYPome)

Prokaryotes (most have no CYPs)


Campylobacter jejun i
Bacillus halodurans
Methanosarcinia barkeri
Mycobacterium leprae
Halobacterium species NRCl
Sulfolobus tokodaii
Sinorhizobium meliloti 2
Agrobacterium tumafaciens 2
Pseudomonas aeruginosa 3
Deinococcus radiodurans 3
Bacillus subtilis 8
Mycobacterium bovis 18
Streptomyces coelicolor 18
Mycobacterium tuberculosis 20
Streptomyces avermitilis 33
Mycobacterium smegmatis 39
Eukaryotes (usually have CYPs)
Schizosaccharomyces pombe 2
Saccharomyces cerevisiae 3
Candida albicans 12
Neurospora crassa 38
Phanerochaete chrysosporium >100
590 Steven L. Kelly et al.

morphologically and developmentally complex lanosterol synthase). This is again a point of


organisms. This is an important area of investiga- divergence in metabolism, with many plants and
tion, as therapeutic CYP inhibitors need targets some protists synthesizing cycloartenol using the
that are implicated in viability or pathogenicity. related cycloartenol synthase rather than lanos-
No such general CYP target exists across the bac- terol synthase, prior to producing obtusifoliol for
teria. Table 13.1 shows some examples of the num- CYP51 metabolism^i.
bers of CYPs present in bacterial genomes. It is The original route to sterol biosynthesis is
clear that the actinomycetes can be especially rich unclear as some protists synthesize lanosterol and
in CYPs and that, unlike prokaryotes, eukaryotic many, like fungi, produce ergosterol as an end-
microorganisms usually contain at least a few CYPs, product"^^. Recently, Mycobacterium tuberculosis
including CYP51 that is used for making sterols. was revealed to possess a CYP51 with sterol
14-demethylase activity"^^, and this protein has
been crystallized"^^. The true function of this pro-
3.1. CYP51 and Evolution of the tein remains to be clarified^^, as the earlier detec-
tion of mycobacterial sterols was probably the
Superfamily
result of sequestration from medium. Other bacte-
Work on yeast resulted in the cloning ria have been shown to contain sterols at relatively
of the first microbial CYP, CYP51, encoding high concentration, as confirmed by purification
sterol 14-a-demethylase, in the Loper labora- and nuclear magnetic resonance (NMR) studies.
tory^^. Orthologues of this CYP were later The earliest of these is the methane utilizing
revealed to be present in animals^^' ^^, and proteobacterium M. capsulatus that synthesizes
plants^^. Many different CYPS Is have now been sterols via a lanosterol route"^^, but not as far as
identified and Figure 13.3 shows a phylogram of ergosterol, sitosterol, or cholesterol. This organ-
CYP51 sequences. Substrates differ slightly ism contains a CYP51 and as such was the first
between the different kingdoms^^, especially in proven bacterial sterol biosynthesis gene and
plants where the enzymes studied so far utilize the protein studied^^. The closest homologues to this
C4-methyl sterol obtusifoliol as the substrate and CYP51 are among the mycobacteria, and then
not the C4-dimethyl substrate (e.g., lanosterol) CYP 170 of Streptomyces coelicolor and plant and
used in fungi and animals. CYP51 is found protist CYP51 (Figure 13.3). More recently,
throughout eukaryotes, although some, for exam- another myxobacterium has been revealed to pro-
ple, nematodes and insects obtain sterol from their duce sterols and a cycloartenol synthase has been
diet. The primitive eukaryote Giardia lamblia also identified, although a CYPS 1 awaits identification
lacks CYP genes as judged by probing of its from this bacterium"^^. We have also identified a
genome using conserved heme-binding motifs for lanosterol synthase and squalene epoxidase homo-
CYP, but this remains unusual among eukaryote logues at a locus in M. capsulatus (unpublished
genomes that may require sterols for stabilizing observation).
membranes. In general, most eukaryote genomes It seems reasonable to assume that sterol bio-
searched to date have at least a few CYPs. The synthesis arose in gram-negative bacteria at least,
question arises: If sterol biosynthesis represents and possibly in gram-positive bacteria, although
an early CYP function, when did it arise? Previous the homologues here could have been the result of
thoughts on what early function CYPs might have horizontal transfer. The need for several genes that
evolved to undertake have centered on their poten- are unlinked in order to make bacterial sterols
tial role in detoxifying oxygen"^^. Within sterol seems to suggest that sequential horizontal trans-
biosynthesis, squalene epoxidase is a non-P450 fer or independent evolution is unlikely, and that a
monooxygenase preceding CYP51 and it is prob- sterol biosynthetic pathway may have evolved as a
able that CYP51 conferred on primitive microor- feature of prokaryotic ancestors of eukaryotes.
ganisms a more robust membrane by producing There is a cycloartenol-type (plant-like) pathway,
sterols. An alternative CYP ancestor could easily as well as a lanosterol-type (fungal/animal-like)
have preceded CYP51, of course. Following squa- pathway, in different bacteria that provides an
lene epoxidase, the next enzyme needed to make extra level of complexity to these considerations,
sterols is 2,3-oxidosqualene sterol cyclase (or as it might be expected that, if ancestral, a single
The Diversity and Importance of IVIicrobial Cytochromes P450 591

A. fumigatus 2
V. nastiicda
M. graminicola
p. digitatum
1 N. crassa
p. italicum
T. acuformis
A. fumigatus 1 LmT. yallundae
B. fuckeliana
C. glabrata / M. fructicola
S. cerevisiae
^^^^^^ B. graminis
C. dubliniensis

C. albicans ^ ' U. necator

S. pombe
C. tropicalis ——

P. anomola —***"
*• —P. chrysosporium

/. orientalis '^

U. maydis

S. scrofa

H. sapiens Cu. elegans

M. marinum —

M. avium T. brucei

M. smegmatis j
D. discoideum
M. tuberculosis
A. thaliana chrom4
0.1
M. capsulatus
S. bicolor T. aestivum A.. thaliana chroml
0. sativa

Figure 13.3. A phylogenetic tree containing sequences from bacterial, fungal, protist, plant, and animal CYPSls
constructed using clustal X and Treeview 6. The sequences are Aspergillus fumigatus CYP51 1 (AF222068),
Aspergillus fumigatus CYP51 2 (AF338660), Botryotinia fuckeliana (AF346594), Blumeria graminis (AF052515),
Candida albicans (AB006856), Candida dubliniensis (AY034867), Candida glabrata (S75389), Candida tropicalis
(M23673), Cunninghammela elegans (AF046863), Issatchenkia orientalis (S75391), Mycosphearella graminicola
(AF263470), Neurospora crassa (http://drnelson.utmem.edu), Penicillium italicum (Z49750), Phanerochaete
chrysosporium (http://drnelson.utmem.edu), Pichia anomola (AFO19903), Saccharomyces cerevisiae (Ml8109),
Schizosaccharomyces pombe (NC003424), Tapesia acuformis (AF208657), Tapesia yallundae (AF276662),
U. maydis (Z48164), Venturia nashicola (AJ314649), Dictyostelium discoideum (http://drnelson.utmem.edu),
Trypanosoma brucei (AF363026), Arabidopsis thaliana (chromosome 1 CYP51, AY05860), Arabidopsis thaliana
(chromosome 4 CYP51, NC003071), Oryza sativa (AB25047), Sorghum bicolor (U74319), Triticum aestivum
(AJ251798), Homo sapiens (U23942), Sus scrofa (AF198112), Methylococcus capsulatus (TIGR 414),
Mycobacterium avium (TIGR 1764/3294), Mycobacterium marinum (Sanger mar388e06), Mycobacterium
smegmatis (TIGR 1772/3269), diwd Mycobacterium tuberculosis (AL123456).

t5q)e of prokaryotic pathway would exist. Some genome duplication. Generally, it is accepted
clarification will emerge with more post-genomic that pathways can evolve sequentially. The pro-
studies. gressively more complex tailoring of sterols was
The further evolution of CYPs must in some selected for in the microbial era based on
cases have included gene duplication events, increased fitness of the organism, probably when
including involving CYP51 individually, besides exposed to physical parameters such as osmotic
592 Steven L. Kelly et al.

stress. In fungi a further CYP, CYP61, evolved to biosynthesis proteins, they are not all part of a
perform the C22-desaturation of sterols and this sterol biosynthetic apparatus.
may have evolved after a gene duplication of Another CYPSl-Wks gene was identified in
CYP5L The two gene families today have very lit- the emerging genome of S. coelicolor A3 (2) and
tle sequence identity (approx. 30%). The fungal the protein had sterol 14-demethylase activity, but
sterol, ergosterol, is also found in protists such as was not essential when the gene was knocked out
trypanosomes and algae such as Chlamydomonas and no sterols are made by this microbe^^. This
reinhardtii^^' ^^, and so a CYP61 may be encoun- gene has been assigned to a different family
tered elsewhere as proposed earlier^^. Stigmasterol (CYP170A1) and the protein shares 23 of the
production in plants requires a C22-desaturation conserved amino acids across CYPSls, whereas
and yet no CYP61 was observed in Arabidopsis the other CYPSls share approximately 40 amino
thaliana. The closest amino acid identity of acids.
a plant CYP to CYP61 was for CYP710, and One of the first important bacterial pathogens
no CYP61 has yet been seen in the green alga sequenced, M. tuberculosis, was found to contain
C. reinhardtii, which also makes ergosterol. It 20 CYPs, one of which was identified as a CYP51.
may be that other CYP families undertaking sterol The hypothesis of azole inhibition of this CYP51,
C22-desaturation will need to be reclassified and parallels to antifungal activity with inhibition
as CYP61 due to common function. Obviously, of fungal CYP51, stimulated the examination of
higher organisms and those that have homeostasis the sensitivity among mycobacteria to known anti-
exist in a different evolutionary scenario fi-om fungal drugs. It was shown in M smegmatis that
single-celled microbes. This presumably resulted potent activity of azole antifungals existed, except
in selection for other sterols such as cholesterol in for fluconazole, but particularly for the topical
animals and the array of phytosterols in plants. In agents^ ^ and this was also found in another
some cases, such as in nematodes and insects that study^^. The latter report and a separate assess-
obtain sterol from their diet, sterol bios3aithesis ment of Mycobacterium bovis BCG sensitivity^'*,
has been lost, while most bacteria have never had showed less sensitivity in this slow-growing bac-
this requirement. terium than in the fast-growing species, where the
minimum inhibitory and bactericidal concentra-
tions of miconazole were less than 2 |ULg/ml.
3.2. Bacterial CYP51 Our further unpublished work indicates that the
sensitivity of M. tuberculosis resembles that of
The identification of sterols in bacteria was M. bovis BCG, but other pathogenic species caus-
first achieved in 1971 for the gram-negative bac- ing skin infections, Mycobacterium chelonei and
terium M capsulatus, where the end-products Mycobacterium fortuitum, are sensitive and may
reported are various C4-methyl and desmethyl be treatable with current topical antifungals
products'*^. A CYP51 was identified in the (Table 13.2). To develop potent azole-type
genome of this bacterium with homology to other inhibitors as antimycobacterial agents will require
CYP5Is and represented a new form with a screening other compounds, and it may well be
(3Fe4S) ferredoxin domain fused at the C-termi- that CYP51 is not the target, although both M
nus of the CYP domain via an alanine rich tuberculosis and M smegmatis CYP51 bind
linker^''. This 62-kDa protein was expressed and azoles with high affinity^^' ^^. Indeed another
purified from E. coli and metabolized lanosterol M tuberculosis CYP, CYP 121, also shows high
(0.24 nmol/min/nmol protein) on addition of a affinity binding for azole compounds^^. Other
ferredoxin reductase and NADPH. Other homo- CYPs from M. tuberculosis, such as CYP 125,
logues of eukaryotic sterol biosynthesis, for exam- bind antifungal azoles poorly (unpublished obser-
ple, lanosterol synthase and squalene epoxidase, vation). Further work in this area is needed to
are evident in this genome, although no genes are establish the mode of action, but while M. smeg-
detected so far for the formation of end-product matis is sensitive to azole compounds it contains
of 4-methyl and 4-desmethyl sterols. With the no CYP121. It does, however, contain a strong
presence of many prokaryotic genomes it has homologue of the sole Mycobacterium leprae
become obvious that while some contain open- CYP, CYP164A1, with which the M smegmatis
reading frames with considerable identity to sterol CYP164A2 has 60% identity. No CYP family is
The Diversity and Importance of Microbial Cytochromes P450 593

I ^ I P! I I

s.s
•3 ^

' ' t (N VO
I ? IS I I
I*
<
o
^.S ^
o cs 5 iS
u
I
U
:5 oo ^ s
A o
5
^ ««
O t^

PQ S ^ .2

00
csj rsi in
V V ^ °°
to o 55
^ S C3
OB O
C« O 4>

g - §

•2 ^ _ - t> (U

Tt OO (N fJ
•t- (U .

^^ -
A A Q W3 (O
S </, .^
I ^^
05 C3 t?
.S t» (u

rsi csj »o vo >^ <N ( ^ m


S Zl, <N V ^
A ^
^ ^ ^ §
«t C O
A O <U C5
?£ ^ S

^ .D -S
^ O ?^
•so ^O /-o \D
^ Q
-•go

.2 § §5
CM
CO ^ '^ J
<N "^ -^
V V O ^^
A
§ §1
|2 O

lisiiiii S o §
1^^
594 Steven L. Kelly et al.

present in all mycobacteria to present a common operon/gene cluster. Also included among the
antimycobacterial target, although many are present genes is another CYP, CYP123. If the CYP51
in all so far known except M leprae (Table 13.3). performs a dififerent endogenous role fi-om other
The genome sequences of various mycobacte- CYPSls, this would require reclassification,
ria have been completed and it became apparent possibly as a CYP170 as with the CFP57-like
that CYP51 was part of a putative hexacistronic gene of S. coelicolor.
operon that was conserved across mycobacteria
except for M leprae, in which no gene or pseudo-
gene exists for CYP51 due to divergent evolution 4. Archetypal Bacterial CYPs
and massive gene decay^"^. One striking similarity
is the close homology between the CYP51 of Table 13.1 shows a list of some bacteria with
mycobacteria and that from M. capsulatus. This is the number of CYPs associated with their
also true for the ferredoxin gene that lies down- genomes to date. Most striking are the number and
stream of CYP51 in mycobacteria, but is fused to diversity seen among actinomycetes such as strep-
the CYP51 domain in M. capsulatus. Possibly the tomycetes and mycobacteria, although some actin-
CYP51 in mycobacteria was transferred by hori- omycetes, such as Corynebacterium diptheriae,
zontal transfer from an ancestor of M capsulatus, have no CYPs. Much of the CYP diversity is
where it has possibly been recruited to a new likely to be due to their role in secondary metabo-
function linked to the other genes in that putative lism, as is true in the filamentous gram-negative

Table 13.3. The CYPome of M tuberculosis Compared to Another Strain with Genomic
Information and the Presence or Absence of these Forms in M bovis, M. avium, M. smegmatis.

H37Rv
CYP number Gene CDC1441,210 M. bovis M. avium M. smegmatis M leprae

CYP51 Rv0754c + + + +
CYP121 Rv2276 + +
CYP123 Rv0766c + + + +
CYP124 Rv2266 + + + + ML1787
CYP125 Rv3545c + + + + ML2024
CYP126 Rv0778 + + + + ML2229
CYP128 Rv2268c + +
CYP130 Rvl256c + + + ML1102
CYP132 Rvl394c + +
CYP135A1 Rv0357c + +
CYP135B1 Rv0568 + + +
CYP136 Rv3059 + + + + ML1742
CYP 137 Rv3685c + +
CYP138 Rv0136 + + + + ML2684
CYP139 Rv1666c + + + ML 1237
CYP140 Rvl880c + + + + ML2033
CYP141 Rv3121 +
CYP142 Rv3518c + + + +
CYP143 Rv 1785c + + + ML1542
CYP144 Rvl777 + + + + ML 1185
CYP102 ML0447
CYP102 ML2159
CYP164 + + ML2088

Notes: Also shown is whether a pseudogene for the M. tuberculosis CYP exists in M. leprae. ML0447 and ML2159 (M. leprae ORF
identifier) are identical pseudogenes in M. leprae that resemble S. coelicolor A3 (2) CYP102B1 and ML 2088 is the only putative func-
tional CYP of M leprae where another member of this family is only found in M. smegmatis. The TIGR databases were utilized here.
The Diversity and Importance of IS/licrobial Cytociiromes P450 595

myxobacterium that produces the anticancer drug (camB). A fourth gene camD encodes a 5-exo
epothilone^^, and the numerous forms present in hydroxy camphor dehydrogenase and the operon
the rifamycin gene cluster of Amycolatopsis camDCAB is controlled by a camR repressor.
mediterranei^^. Some examples of bacterial CYP CYPlOl still provides an archetypal system for
reactions are shown in Figure 13.4. investigations of the CYP catalytic cycle and was
Many of the bacterial forms are described by the first CYP structure that revealed the triangular
David Nelson in his central website for CYPs prism shape of the proteins and the heme with its
(http://dmelson.utmem.edu/nelsonhomepage.html), cysteinyl thiolate ligand (see Chapter 3)^^' ^^.
but others are not yet assigned CYP numbers. The Other bacterial CYPs also undertake the break-
archetypal bacterial CYP is of course P450(^^j^, down of carbon sources for microbial growth. For
assigned as CYPlOl, obtained from the bacterium instance, CYP 108A1 (P450^g^p) metabolizes ter-
P. putida ATCC17453^"^^ This enzyme catalyzes pineol^^ andCYP176Al (P450^J can metabolize
the 5-exo hydroxylation of camphor, part of the cineol^^, while others can metabolize pollutants
breakdown of this carbon source for growth. such as thiocarbamate herbicides and atrazine, as
The CYPlOl operon contains the CYP (camC) illustrated by CYPl 16 from a Rhodococcus sp.^^.
together with class I electron donor partners, a The CYP 105 family of streptomycetes especially
putidaredoxin reductase (camA), and putidaredoxin is associated with a wide variety of xenobiotic

'OH

(+)-Camphor 5 -exo-Hydroxy camphor

OOOH
^^^^v^"°°" CYP,02 Z ^ ^ ^ ^ ^ T

Arachidonate 18 - hydroxy-Arachidonate

HOHnC^

OH OH
CYP 108

'CH3 •CH,
CH(

CYP176A

Cineole

^PU. ^ULJ J^^

o • o
Epothilone C

Figure 13.4. Examples of bacterial CYP reactions.


596 Steven L. Kelly et aL

metabolism, although roles in pathways of sec- no CYPs, but the actinomycetes have revealed
ondary metabolism are also emerging^ ^ many genomes containing numerous CYPs.
As with all CYP activities, the substrates are Mostly these are orphans with no known ftinction,
largely lipophilic, and this is true of the other bac- but included are new classes of CYPs from
terial archetypal CYP, CYP102A1 (P450BJ^.3), Rhodococcus described above, and from genome
which as a class II system utilizing an FAD and projects the diversity is surprising, with 20
FMN containing reductase domain, has provided a CYPs i n M tuberculosis^^, 18 in 5. coelicolor^-^'^^,
model for eukaryotic CYPs. This protein, contain- 33 in .S. avermitilis^^, and 39 in M smegmatis^^.
ing both CYP and reductase domains in a soluble The genomes of streptomycetes are larger than the
fusion protein that metabolizes various fatty acids, mycobacterial genomes with almost twice as
mostly at (n-l, was characterized in a series of many genes arranged on a linear chromosome.
publications from Fulco and colleagues during the Thus, S. coelicolor contains 18 CZPs among
1980s^^' ^^~^^. The rates of various reactions have approximately 7,825 open reading frames, that is,
been studied and this protein turns over substrate 0.2% of genes, while M. smegmatis has 39 out of
more efficiently than other CYPs utilizing sepa- approximately 3,800 genes (1% of genes). This lat-
rate redox proteins, with a rate of 17,000 min~^ ter proportion is similar to that observed in plants.
observed for arachidonate metabolism. Structural Figure 13.5 shows a phylogenetic tree of the
considerations for this protein are not the object of mycobacterial CYPomes of both M. tuberculosis
this chapter, but resolution of the structure of the and M. smegmatis.
CYP domain was a landmark in the field^^, and The genomes of the mycobacteria and strepto-
allowed numerous subsequent investigations using mycetes do not contain many CYP families in
site-directed mutagenesis to probe the structure as common. Both contain a CYP51-like CYP, and in
well as the reductase domain^^"^^. S. coelicolor and S. avermitilis this CYP is called
Besides being present in proteobacteria, CYPs CYP170A and lies adjacent to a sesquiterpene
are also present in archaebacteria and some of cyclase that may well be of relatedftinction^^'^^.
these, CYP 119 from Sulfobolus solfataricus and One CYP, CYP125, was originally observed in
CYP 175 from Thermus thermophilus, have had M. tuberculosis, and recently a CYP125 was also
their structure solved^^' ^^. While these are inter- found in S. avermitilis?"^ CYP 105s originally found
esting, the absence of known endogenous function in streptomycetes and associated with xenobiotic
precludes their discussion here. Thermophilic metabolism have also now been identified in M
CYPs have been studied and we await functions, smegmatis (Figure 13.5), as well as a CYP107^^~^^.
as well as CYP stucture/ftinction for CYPs The only other CYP family seen in both genera
with activity at low temperatures that may have is a CYP 102 identified in S. coelicolor and
industrial uses. S. avermitilis^^' '^^, and as a pseudogene seen in the
dramatic gene decay observed in M leprae
(Table 13.3).

5. Biodiversity of Bacterial CYPs


and the Actinomycetes
5.1. Mycobacterial CYPs
The actinomycete bacteria encompass a wide The diversity of mycobacterial CYPs has been
range of species, including Rhodococcus spp., mentioned and trees relating the different CYP
Corynebacterium spp., Mycobacterium spp., and families of M tuberculosis and S. coelicolor indi-
Streptomyces spp., and represent important organ- cate they are quite different, reflecting the many
isms for biotechnology in terms of enzymes, natural hundreds of millions years since divergence from
products, biotransformations, and bioremediation. a conamon ancestor. ^'*' ^^ A list of CYPs of
Many are saprophytic, soil-inhabiting, gram- M tuberculosis is shown in Table 13.2, where the
positive bacteria with a high G+C content, and presence of these families in other mycobacteria is
some are also life-threatening human pathogens. also shown. All M tuberculosis CYPs conform to
As mentioned earlier, many bacteria, including the expected conserved amino acids within the
within the actinomycetes C. diptheriae, possess sequence of a CYP, including a conserved T
The Diversity and Importance of Microbial Cytochromes P450 597

1 2
-CYP187A1 ^ s
- CYP186A1
CYP136B1
CYP136A2
o B
CYP136A1
^
I I pCYP51smeg
L.cYP51mtb
CYP132
CYP139
o o
CYP185A1
• ^ CYP185B1
CYP137
CYP138A2
CYP138A1
^3
CYP135A1
CYP135B2
^ CYP135B1 ^
CYP188A1
CYP143
CYP192A1 B o
CYP151A1
CYP107AA1
CYP107AB1P X

r^ CYP164A2
CYP140A2
CYP140A1
CYP105S1
CYP105T1
CYP121
1^ i !
CYP141
CYP191A1
CYP190A1
CYP123A2
CYP123A1
^ 3^+- _3i ^
CYP189A1
2 3 U
CYP189A2
CYP189A3
CYP189A4
CYP130A2
CYP130A1 -Ci --^ 00 ^
CYP128
CYP150A3 ^ 2 OH

CYP150A4
CYP150A2
CYP144A2
^ CYP144A1
CYP108B3
CYP108B1
CYP108B2 ::! I I
CYP124A2 z
CYP124A1
CYP124B1
CYP124C1
CYP125A5P
g U
CYP125A4
CYP125A3
CYP125A1
CYP126A2
CYP126A1
CYP142A2
CYP142A1

111
598 Steven L. Kelly et al.

within the I-hehx involved in oxygen-binding/ complement. Included in this CYPome are the
electron transport, an EXXR motif in the K-helix, first other members of the CYP 108 family similar
and a C-heme ligand in the C-terminal region. to P450TERP (CYP108A1)5^ This may well reflect
Compared to the published sequence of the utilization of similar carbon sources to terpineol
M tuberculosis genome, M bovis^ although very for growth. The study of M smegmatis CYPs
closely related to M tuberculosis, has only 18 in bioremediation will be an important area of
CYPs, with CYP130 being absent and CYP141 future research, as for the fungus Phanerochaete
being present as a pseudogene. The sequence of chrysosporium discussed later, for which 1% of
M. leprae showed massive gene decay as this the genes also encode CYPs. Common with the
organism moved toward parasitism, with only findings of many other genomic projects, the
approximately 1,800 genes retained vs 3,800 in function of the mycobacterial CYPs remains
M tuberculosis. Only 12 close homologues of the unknown; however, as these are elucidated by
M tuberculosis CYP complement were detected gene knockouts, transcriptomics, proteomics, and
in M smegmatis (Table 13.2), and this has the metabolomics, there will be benefits for medical
largest complement so far with 39 CYPs. When science and biotechnology.
completed, the genomes of the related species
Mycobacterium avium and M avium ssp. para-
tuberculosis will have a similar number of CYPs, 5.2. Biodiversity in
based on preliminary analysis of the data
deposited at TIGR.
Streptomycetes
CYP164A2, the M smegmatis homologue of Streptomycetes are organisms with a complex
M leprae ML2088 (CYP164A1), is 1,245 bp in life cycle, which involves the formation of a
length, encoding a predicted protein of 414 aa filamentous mycelium giving rise to aerial hyphae
with a molecular weight of 44.9 kDa. By compar- that produce spores. This, in part, explains
ison, CYP 164A1 is 1,305 bp long and encodes a the requirement for a larger genome in these
predicted protein of 434 aa with a molecular bacteria that are also important producers of
weight of 47.6 kDa. M. smegmatis CYP162A2 is bioactive molecules (secondary metabolites).
60% identical (249/415) and 75% similar These metabolites represent about two thirds of
(313/415) in a 415 aa overlap with CYP164A1 the microbially derived compounds that include
(BLASTP score e-130). Homology extends across antibacterial (erythromycin, tetracycline), antifun-
all regions of the proteins, with only two gaps. gal (amphotericin, nystatin), antiparasitic (aver-
In contrast, the closest M tuberculosis homologue mectin), immunosuppressor (FK506), anti
to CYP164A1, CYP140, shows only 38% iden- cancer (adriamycin), and herbicidal (bialaphos)
tity (145/379) and 515 similarity (196/397), with compounds. Structural diversity is observed
30 introduced gaps (BLASTP score 3e-58). within all of the compounds and CYPs participate
The leprosy genome also contains a separate in oxidative tailoring of many of these, and thus
pseudogene of M tuberculosis CYP 140 at locus play a key role in many of these pathways. The
ML2033, so these CYPs are likely to be function- smell of wet earth on a spring day, resulting from
ally distinct. geosmin, is also a product of actinomycetes/strep-
The analysis of the CYP families present in M tomycetes. Geosmin requires CYP for its biosyn-
smegmatis, reveal that, as expected, many new thesis and in S. avermitilis this is probably
families covering CYP186-192, have been identi- undertaken by CYP 180A 1^1 With all these
fied. The CYP family members in M. tuberculosis important biosynthetic pathways in which CYPs
CYP121, 128, 132,135A, 137, 139,141, and 143 are known to participate (Table 13.4, Figure 13.6),
are not found in M smegmatis. One CYP had there is of course interest in the cryptic pathways
already been identified before the M. smegmatis associated with the many orphan CYPs of strepto-
genome sequence, and this CYP was involved in mycete genomes for which function has yet to be
morpholine utilization (CYP15iy^. Interestingly, detected. Apart from natural product biosynthesis,
this soil microorganism has been associated with streptomycete CYPs have been identified as
useful bioremediation properties and this may in good biocatalysts with particular attention to
some instances be associated with the CYP xenobiotic metabolism by CYP105D1, identified
The Diversity and Importance of Microbial Cytochromes P450 599

in Streptomyces griseus^^, and CYP105A1 associated with operons with a conserved structure
from S. griseolus, which has been used to that have been called conservons^^. In these, the first
manipulate herbicide tolerance in plants^ ^ Indeed, open-reading frame encodes a sensor kinase, then
streptomycetes have been used as a source of two open-reading frames of unknown ftinction
drug metabolites by fermentation^^, and have followed by a gene encoding an ATP-binding
also been used for stereo- and regio-specific domain and finally, in some cases, encoding one or
biotransformations^^' ^^. two CYPs. In conservon 10, the downstream CYPs
Among the streptomycetes, two genomes are CYP157A1 and CYP154C1; in conservon 11,
have already been released into the public domain, the downstream CYP is CYP157B1; in conservon
for S. coelicolor A3(2) (ref [72]) and for 12, the downstream CYPs are CYP156A1 and
S. avermitilis^^. The S. coelicolor genome of CYP154A1; and in conservon 13, the downstream
7,825 open-reading frames contained 18 putative CYP is CYP157CL The frmctions of these CYPs is
CYPs that were cataloged and expressed in a sys- intriguing, and as CYP154C1 can metabolize
tematic study^^. This laboratory strain is the antibiotics, it could be related to a chemical defense
model for streptomycetes and produces a number and detoxification system^"^. Recently, a conservon
of secondary metabolites, although none are cur- from S. griseus was isolated containing a
rently commercially important. The CYP roles are CYP 157 homologue. Gene inactivation of the first
unclear, as the CYPS 1-like protein now called gene of the conservon resulted in precocious forma-
CYP170A1, is not involved in sterol biosynthesis tion of mycelium and secondary metabolism, sug-
and a gene deletion event was found not to be gesting this operon regulates the onset of
lethal^^. Surprisingly, six of the eighteen CYPs are differentiation^^.

Table 13.4. Streptomycete Cytochromes P450 Including CYP Assignments Where Available
(www.dmelson.utmem.edu/P450.family.list.html).

Bioactive molecule
Streptomyces sp. CYP identification produced Function

S, griseolus CYP105A1, 105B1, 105C1


S. carbophilus CYP105A3
S. griseus CYP105D1, 105D2, 107F1
S. scerotialus CYP105D3
S. lividans CYP105D4
S. lavendulae CYP105F1, 107N1,160A1 Complestatin Anti-HIV
S. noursei CYP105H1,161A1 Nystatin Antifungal
S. tendae CYP105K1,162A1 Nikkomycin Insecticidal
S. fradiae CYP105L1, 113B1,154B1 Tylosin Promotant
S. clavuligerus CYP105M1
S. thermotolerans CYP107C1
S. erythraea CYP107A1,107B1 Erythromycin Antibacterial
S. antibioticus CYP107D1 Oleandomycin Antibacterial
S. hygroscopius CYP107G1, 122A2,122A3 Rapamycin Antibacterial
S. venezuelae PikC (PicK) Pikromycin Antibacterial
S. maritimus CYP107R1
S. peucetius CYP129A2, 131A1,131A2 Daunorubicin Antitumor
S. spheroides CYP163A1 Novobiocin Antibacterial
S. avermitilis CYP171A1 Avermectin Antiparasitic
S. acidscabies TxtC Thaxtomin Phytotoxin
S. nodosus Orfl, Orf2 Amphotericin Antifungal

Notes: Many are involved in biosynthetic gene clusters of commercially important bioactive natural products and those implicated
in this biosynthesis are in bold. The list does not include those found in the genomes of Streptomyces coelicolor and Streptomyces
avermitilis.
600 Steven L. Kelly et al.

HO''
O N(CH3)2

Methymycin (Ri=OH, R2=H) Pikromycin(R3=OH)


Neomethycin (Rj =H, R2=OH) Narbomycin (R3 =H)
YC-17(Ri=H, R2=H)
OH

H3C"

Amphotericin

Figure 13.6. Some of the natural products used as drugs and produced by various streptomycetes.

Some of the functions of the S. coelicolor have previously detected known end-products.
CYPs are in secondary metabohsm operons, for The structural genomics of these CYPs is also in
example, CYP158A2 located downstream of a progress, with the first structure reported already
Type III polyketide synthase and CYP105N1 forCYP154Cl81
located downstream of a nonribosomal peptide The second streptomycete genome completed
synthase. These are currently subject to functional has revealed 33 CYPs comprising 0.4% of genes.
studies using the tools of gene disruption and This industrial microorganism is important for
metabolite/metabolome profiling, although not the production of avermectin, an anti-helminthic
all the secondary metabolite genes identified agent. We have cataloged the S. avermitilis
The Diversity and Importance of IN^icrobial Cytochromes P450 601

CYPome and of note is the discovery of seven new polyketide synthase, CYP154D1 is adjacent to an
families and two conservons containing CYPs^"^. indole dioxygenase (and maybe involved in xeno-
The CYP157s within the conservons, as in biotic breakdown), while CYP158A3 is adjacent to
S. coelicolor, deviate from the prior conservation a Type III polyketide synthase, as is CYP158A2 in
in CYPs of an EXXR motif within the K-hehx. S. coelicolor, which is likely to have a common
CYP157A2 and CYP157C2 exhibited a ^^^ENYSSf function.
and ^^^EQSLW motif, respectively. Together with The emergence of further streptomycete
the 5. coelicolor CYP156A1 motif of 272STVR, genomes will add to our understanding of CYP
the need for E and R is not essential, leaving evolution, and the numbers of CYPs in unknown
only the heme cysteinyl ligand as the amino acid pathways of secondary metabolism will reveal
essential in all CYPs. These motifs are the subject new natural products. Given the estimate that only
of current experimental examination. 1% of microorganisms are culturable, the depth
There were no clusters of S. avermitilis CYPs of the biocatal3^ic reservoir of CYPs becomes
in a single subfamily except for CYP105D6 and evident. The reason why the streptomycetes and
CYP105D7. These CYP105D forms seem associ- mycobacteria have many CYPs is not clear, as
ated with filipin bios)aithesis, so that the roles of other soil bacteria contain either small numbers of
CYPlOSs in xenobiotic rather than secondary CYPs (Table 13.1) or none.
metabolism may well need reconsideration.
CYP171A1 was involved in C8a oxidation in
avermectin biosynthesis, CYP107W1 in oligo- 5-3. CYP biodiversity in
mycin biosynthesis through oxidation at C12j Archaebacteria
and CYP180A1 is predicted to be involved in
geosmin biosynthesis^'*. The closest homologue of CYPs also are found among the archaea in
CYP180A1 in S. coelicolor is CYP107U1, but under a third of the genomes so far sequenced.
geosmin is made by S. coelicolor and so must be Functional information about them is totally
S5mthesized using a CYP from a different family. absent. Whether they arose later in evolution, after
When identified, it will require a modification of the appearance of oxygen, is also unclear. Their
the nomenclature to place CYPs with common ability to maintain integrity in extreme conditions
function in the same family, an illustration of of temperature, and so on, could be useful in
some of the problems relating homology and CYP biotechnology as is the ability to function at low
families to function. temperatures. The structures of two CYPs have
been obtained. First, the CYP119 structure from
Other CYPs identified in this genome include
S. solfataricus has been obtained and the heat sta-
CYP 102s, one of which encodes a fusion protein
bility of the protein has been ascribed to clusters
with a CYP and a reductase domain unlike the sin-
of aromatic residues^^. The second structure of
gle CYP102B1 found in S. coelicolor A3(2). The
CYP 175A1 from the thermophilic T. thermophilus
CYP 102s are also seen in bacillus species that are
HB27 has also recently been solved^^.
sporulating bacteria, but the endogenous function
remains unclear. The S. avermitilis genome also
contained a CYP 125^^. This had only previously
been seen in mycobacteria, but was not present in 6. Fungal CYPs
S. coelicolor and was the first CYP family
detected in two diverse actinomycete species. The yeast Saccharomyces cerevisiae has three
Eleven of the CYPs from S. avermitilis were CYP genes. CYP51 was identified in 1987^^ CYP57
located in known gene clusters involved in sec- was found in 1994 to be responsible for the syn-
ondary metabolism, including that of geosmin, thesis of dityrosine, which is needed for the yeast
avermectin, filipin, and pentalenolactone biosyn- spore wall^^, and CYP61 was identified in 1995 as a
thesis, and again, as in S. coelicolor, others appear sterol C22-desaturase in a proteomic study linking
to be involved in uncharacterized pathways of protein information to the emergence of this
secondary metabolism. CYP 178A1 is in a cluster genome^^. Fungal CYPs are generally class II asso-
with a nonribosomal peptide synthase, CYP107Y1 ciated with the endoplasmic reticulum and a single
and CYP 181Al are associated with a Type II reductase drives all the CYPs, as in humans.
602 Steven L. Kelly et al.

This limited diversity of CYPs was also seen in revealed approximately 123 heme-binding motifs,
the fission yeast Schizosaccharomyces pombe so that approximately 1% of the genes of this
that had CYP51 and CYP61 only, the minimal microorganism encode for CYPs. This organism
CYP requirement for ergosterol biosynthesis. can degrade recalcitrant pollutants and the CYP
The number of completed fungal genomes in the system has been implicated in this activity^^"^^
public domain is limited, but will expand in the The fungus is also commonly seen as a bracket
coming years with sequences for major human fungus that can break down wood. As plants
and plant pathogens. This will rectify the current utilize only a small number (four) of CYPs to
imbalance, given the economic, biomedical, synthesize lignin it seems unlikely that all the
biotechnological, and scientific reasons for CYPome of P. chrysosporium is involved in this
obtaining fungal genome data and the large scien- aspect of metabolism, so much remains to be
tific community that will use this information. A discovered about function of the orphan CYPs.
shotgun analysis of the genome of Candida albi- Only one NADPH-reductase for the CYPs
cans, a major human pathogen, has been com- was present and this has been expressed and char-
pleted, but is not yet published at the time of acterized^^'^^.
writing. Our analysis of this genome reveals Fungal comparative genomics is in an early
approximately 12 putative CYPs, including stage and, as with streptomycetes, much informa-
CYP51 and CYP61, but also a novel orphan form tion about secondary metabolism is anticipated.
CYP501 and many members of the CYP52 family. Fifteen further fiangi are to be sequenced,
This was surprising, as these alkane utilization including the pathogen Aspergillus fumigatus.
proteins are found in soil/environmental yeasts Also of interest will be the pathogenic basidio-
like Candida tropicalis and Candida maltosa. mycete Cryptococcus neoformans that, as with
Perhaps these CYPs are involved in utilizing the P chrysosporium, may have a large CYPome as it
host lipid in animals, or an otherwise unrecog- is also associated with life in hollow eucalyptus
nized environmental niche for C. albicans exists. trees and may therefore have evolved in a similar
Another interesting observation is the presence of niche. Information on these projects is available
a CYP56 homologue, that in S. cerevisiae is on the web at the Sanger Center and TIGR sites.
involved in dityrosine formation for the spore wall Purification of fungal CYPs from cell extracts
after yeast meiosis to produce tetrads^^. However, is a difficult task due to the usual low specific
dityrosine has been detected in the mitotic cell content, instability, and the presence of multiple
wall of C albicans^^. forms. The fungal steroid hydroxylase CYPs have
It was surprising that the Neurospora crassa been studied and a polycyclic aromatic hydrocar-
genome contained 38 CYPs in a 40 MB genome. bon hydroxylase^^' ^^. The emergence of genomes
The genome of this filamentous fungus contained and the ability to express the CYPs present in
many CYPs from existing families, but as E. coli or yeast has greatly facilitated their
usual for a eukaryotic species of a previously study, as will the application of transcription
unvisited biological type, it contained many profiling.
new families of orphan function. The families In an early study by British Petroleum, the use
observed were CYP5U 61A5, 53A4, 54, 55A6, of a Candida sp. producing single-cell protein
65B1, 65C1, 68D1, 505A2, and 507A1, besides from oil was envisaged. Although this became
CYP527A1 to CYP553A1 which represent economically unviable during the 1970s with the
new families. No doubt similar numbers of rise in oil prices, it became apparent that CYP was
CYPs will be identified in other fungi, but so far responsible for the initial oxidation and that the
P chrysoporium has many more for a fungal species CYPs responsible were from a new family,
(http://drnelson.utmem.edu/nelsonhomepage. CYP52^^^ 96 Other CYP52s have been found and
html). studied in many yeasts, including C. maltosa and
There is interest in using fungi in bioremedia- Yarrowia lipolytica^^' ^^. Typically, many CYP52s
tion and one of those that has been used commer- are present in these yeast. Eight were found in
cially is the white-rot fungus P. chrysosporium. C. maltosa, and knocking out four of these genes
This is a basidiomycete, higher fungus with about (also called ALK genes in the yeast nomenclature)
10,000 genes. Probing the unannotated genome prevented growth on n-alkane^^.
The Diversity and Importance of IVIicrobial Cytochromes P450 603

Other fungal CYP families identified include a opportunistic nature of these fungal microorgan-
benzoate/7fl!ra-hydroxylase from Aspergillus niger isms that prey on the old and young, but also
and a cycloheximide inducible CYP54 from increasingly on patients in intensive care as well
N. crassa^"^^ ^^^. CYP55 (P450^ J from Fusariumas with HIV, during cancer chemotherapy, and
oxysporum represented a new class of CYP, as it is after organ transplantation^ ^^. The infections
soluble, and carries out nitric oxide reduction are by a variety of fungal species that also vary
without the need for a CYP-reductase (CPR) or a geographically, as well as demographically, but
requirement for molecular oxygen. It was the first among the most important are C. albicans,
eukaryotic CYP to have its structure resolved and increasingly other Candida spp. (such as Candida
was a member of a new class of CYP^^. glabrata and Candida krusei), C neoformans,
CYP56 was found to be needed for dityrosine A. fumigatus, Histoplasma capsulatus, Pneumo-
production for the spore walls of S. cerevisiae^^, cystis carina, Coccidiodes immitis, Penicillium
while CYP5 7 was identified among a group of italicum, Fusarium (normally associated with dis-
six pea pathogenicity genes as a gene on a super- eases in plants), and even man's best friend,
numary chromosome of Nectria haematococca. It S. cerevisiae. Equally frequent are the skin and
plays a role in detoxifying the phytoalexin pisatin nail infections produced by dermatophytic fungi
produced by the plant host^^^ CYP61, as men- (Trichophyton rubrum, Epidermophyton spp.,
tioned above, is responsible for sterol C22-desatu- Microsporum spp.), which represent a significant
ration during ergosterol biosynthesis^^' 102-104 market for drugs, albeit not because of life-threat-
Interestingly, some rice planthoppers and anobiid ening conditions. However, of the hundreds of
beetles use symbiotic yeast-like symbionts as a thousands of fungal species, only about a hundred
sterol source ^^^. However, unlike in the beetles, are reported as pathogens^^^.
the planthopper symbiont has a defective CYP61
containing nonsense mutations and therefore
7.1. The Fungal CYP51 System
accumulates ergosta-5,7,24(28)-trienoL The selec-
tion of this change is interesting in terms of the During the 1960s and 1970s a series of agro-
benefits in the relationship. chemical fiingicides and clinical antimycotics
Some CYPs (CYP58, 59, 64) have been found became available that were found, in studies with
to play a role in aflatoxin and mycotoxin biosyn- the plant pathogen Ustilago maydis, to be inhibit-
thesislo^^^^ while CYP68 is in a gene cluster ing sterol CH-demethylation^^"^. This was also
involved in giberellin biosynthesis in Giberella observed for azole compounds when treating
fujikuroi^^^, and another CYP is in the biosynthetic C. albicans infections^^^. This step of sterol bio-
pathway of paxilline synthesis by Penicillium synthesis had been postulated to be a cytochrome
paxilli^^^. Fungal genome analysis will reveal P450-mediated activity^ ^^' ^^^. In pioneering work
many more CYPs involved in biosynthetic path- by Yoshida, Aoyama, and colleagues, CYP was
ways of known and unknown natural products. purified and characterized from S. cerevisiae and,
Further novel CYP forms can also be anticipated, in a series of studies, they looked at the demethy-
such as CYP505 from E oxysporum that metabo- lation event occurring via three sequential
lizes fatty acids and is a membrane-bound CYP monooxygenase reactions, and also at the enzy-
with a C-terminal CPR fusion^ ^ ^ matic and electron transport requirements of
the system that involved a typical eukaryotic
NADPH-CPRii^' 119. The reaction sequence was
tested using recombinant C albicans CYP51 pro-
7. Azole Antifungals and the tein and it was shown that the acyl-carbon bond
Evolution of New Resistant cleavage occurred by a mechanism similar to
Genes those proposed by Akhtar and colleagues for the
reactions involving CYP17 and CYPI9120,121. in
Antifungal treatments have become increas- other studies, CYP was purified from S. cerevisiae
ingly important as fungal infections have become and shown to metabolize benzo((3)pyrenei^^. This
one of the top five most frequently encountered in was presumably the form that was responsible for
the clinic. This increase is associated with the activating pro-carcinogens in yeast genotoxicity
604 Steven L. Kelly et al.

HO''
Eburicol Ergosterol

HO'
14-methylfecoster ol

C5,6-desaturase

HO''
14-methyl-3,6-diol

Figure 13.7. The sterols accumulating under azole treatment of C. albicans including the end-product, 14a-
methylergosta-8,24(28)-dien-3p,6a-diol, that does not support growth and requires sterol C5-desaturase
for biosynthesis. In most fungi, the substrate for CYP51 is more likely eburicol under normal conditions, and
not lanosterol, although both are metabolized.

assays^^^' ^'^^. The CYP purified was most likely the sterol accumulating under treatment from a
CYP61, which has a low-level activity in fungistatic end-product, 14a-methylergosta-
benzo((af)pyrene metabolism as well as an endoge- 8,24(28)-dien-3p,6a-diol, to a functional end-
nous activity in sterol C22-desaturation'*^' ^^^^ ^^^. product, 14a-methylfecosterol. This conversion
Genetic analysis of CYP51 by gene disruption, required sterol C5-desaturase and therefore
the first for a CYP in any organism, demonstrated revealed sterol C5-desaturase to be implicated in
the essential nature of the gene product^^. the mode of action of azoles through introduction
Chemical inhibition of CYP activity was also of a 6-OH group into 14-methyl sterol in a pre-
studied and genetic suppression of the effect of sumed attempted desaturation^^^. Subsequently,
azole inhibition (by ketoconazole and flucona- this was also found to suppress the genetic disrup-
zole) was found to be mediated by sterol tion of the CYP51 locus^^^' ^^l The discovery in
C5-desaturase mutants. These mutants changed the clinic of similar azole-resistant sterol
The Diversity and Importance of Microbial Cytochromes P450 605

C5-desaturase mutants of C albicans confirmed the 7.2. Azole Activity and


central role of this biotransformation in the mode of Resistance in Fungi
action in a clinical setting^^^' ^^^. Sterol profiles
of untreated and treated C albicans illustrate the In the early 1980s, it was first reported that
major sterols accumulating in wild-type and sterol resistance to the antifungal agent ketoconazole
C5-desaturase defective mutants (Figure 13.7). occurred in patients suffering chronic mucocuta-
It has also been found that the lethal effect of a neous candidiasis, but a general problem with
sterol C4-methyl oxidase gene knockout can be antifungal resistance had never been encountered
suppressed by a mutation in CYP51^^^. These clinically. Resistance, had however, been seen in
mutants accumulate lanosterol without further agriculture in the 1980s with the use of related
sterol metabolism and this indicates that lano- fungicides generically termed demethylase
sterol itself can support yeast growth despite pre- inhibitors (DMIs).
vious sterol-feeding studies to the contrary^ ^^. Azoles bind to the heme of fungal CYP as a
Consequently, retention of a C14 methyl group on sixth ligand, as evidenced by the generation of
a sterol does not in itself make the sterol nonfunc- Type II spectra using fungal microsomes, tj^ically
tional, as might be expected from the original with a maximum at approximately 430 nm and a
events resulting in the evolution of the sterol minimum at approximately 410 nm^"^^. This inter-
pathway. action involves the N-3 of an imidazole ring or
One additional point of interest here is the use the N-4 of a triazole ring as a ligand to the heme,
of azole antifungals and CYP51 in proof of prin- resulting in the formation of a low-spin, azole-
cipal experiments within functional genomics, as bound complex. It had been known from 1972 that
pioneered using the yeast genome. Transcriptome imidazoles could be CYP inhibitors ^'^'^. A similar
studies with S. cerevisiae following treatment with interaction was seen for the antifungal pyridyl
fluconazole revealed increased CYP51 expres- compound buthiobate on binding to purified
sion, showing the effect of the inhibitor and the S. cerevisiae CYPSl^"^^. The studies on buthiobate
usefulness of transcriptomics for determining showed saturation of the Type II spectra with
drug mode of action^^^' ^^^. a one-to-one ratio of the antifungal and CYP51,
For activity, eukaryotic CYPs located in the reflecting the high affinity of binding, although
endoplasmic reticulum require NADPH-CPR to buthiobate and other azole antifungals can be
provide the first and/or the second electron needed displaced by carbon monoxide.
for the catalytic cycle (see Chapter 4)^^^. Gene The orally administered antifungal drug keto-
disruption of yeast CPR was surprisingly not conazole was followed into clinical use by flu-
lethal^^^, although the genome subsequently conazole and itraconazole, and more recently
revealed no further CPR genes^^^, and cells still voriconazole, with further compounds still in
synthesized ergosterol^^^. The source of electrons clinical evaluation trials, including posaconazole
to support the requirement of CYPS land CYP61 and ravuconazole (Figure 13.8)^^^. In contrast,
for ergosterol biosynthesis has been studied and the range of agrochemicals that are in use is more
genetic evidence and reconstitution studies have diverse, and although newer compounds have
shown that cytochrome b5 and cytochrome b5- been developed with alternate modes of action,
reductase are responsible (Figure 13.7)^^^. This resistance problems will emerge, necessitating a
represents a difference from animals, where a requirement for azoles. Resistance has emerged as
CPR gene knockout in Caenorhabditis elegans a serious problem for agricultural and clinical use
and mouse is lethal ^"^^'^"^^ Further differences exist of azoles, but there is seemingly no potential
in the yeast CPR, for which soluble forms have causal link as with bacteria and concerns over the
been produced that can support CYP activity, as use of growth promotant antibiotics on the farm.
shown in reconstitution assays and in genetic The compounds have a selective effect on the
complementation studies using suitable yeast pathogen over the effect on the human/plant
strains^^^' ^^'^. Thus assumptions based on studies CYP51, although direct comparisons at the level
with mammalian systems may not be applicable of the enzyme suggest that the sensitivities are
across other Kingdoms or in other areas of CYP closer to each other than might be anticipated
biology. (>10-fold)i46' 147. Possibly CYP pools give a
606 Steven L. Kelly et aL

(1st electron)

P4503+ AH

P^502+-AH (2ncl electron)


O2

(1st electron)

P45(h'+ AH

^5 ^ bSFT* NADIi

P4502+ -AH ^-^"^^ electron)


02

Figure 13.8. The typical CYP catalytic cycle with the potential for the second electron to be provided via
cytochrome b5 rather than NADPH-cytochrome P450 reductase and the alternative observed in yeast where NADPH-
cytochrome P450 reductase is not essential.

protective effect against CYP51 inhibitors in for new azole drugs is being investigated. Some of
plants and humans. these are shown in Figure 13.9.
With the emergence of the AIDS epidemic Resistance to ketoconazole was studied in the
oropharengeal candidiasis became a serious prob- 1980s in isolates obtained from patients suffering
lem and is often the presenting symptom indicat- chronic mucocutaneous candidiasis. One of these,
ing HIV infection, while in Southeast Asia it is from the Darlington isolate(s), has been found to
often cryptococcosis. Aspergillosis is a serious contain defective sterol C5-desaturase and altered
health risk in organ-transplant patients and treat- CYP51 proteins, with the latter containing the
ment success is much less than for common bac- substitutions Y132H and MTIT^^^ Multiple
terial infections: here it appears that the latest mechanisms of resistance, between and within
azole drug voriconazole is superior to ampho- individual strains, is a common finding since the
tericin, the previous drug of choice for this infec- mid-1990s, when fluconazole resistance, prima-
tion^^^' ^^^. With prolonged and often prophylactic rily in HIV positive patients, became a clinical
use of azole drugs it is not surprising that resist- problem. First, some resistant strains of C albicans
ance has emerged, requiring increasing drug treat- from HIV patients were found to contain reduced
ment to control disease, especially for candidiasis. concentrations of fluconazole ^^^' ^^^, and this was
Molecular investigation of the mechanisms giving correlated with overexpression of transporters of
rise to resistance is ongoing the relevance of these the ABC superfamily, notably Cdrlp and Cdr2p
The Diversity and importance of iS^icrobiai Cytocliromes P450 607

CH3
,CH2-N J,
,CH-C2H5
0\ yo y—V ,—V ,—. -y—n

OH
/N I ,N
K' "N—CH,—C CH2—N 7
N

Fluconazole
^CH,~^^,/^N

Ketoconazole

Ravuconazole

L^ / Posaconazole

Figure 13.9. Some azole antifungals in current clinical use and under trial.

(Candida drug resistance proteins)^^^' ^^^. In addi- on different series of isolates from two patients
tion, other studies implicated a major facilitator during the emergence of resistance, increased
transporter of C albicans in drug resistance mRNA levels for CYP51 were observed in addi-
{MDR1\ as well as showing that CDRl and CDR2 tion to increased transcription of drug transporters
played a role in transporting the drug^^^' ^^^' ^^'*. (CDR and MDRl) and substitutions in the amino
Other C albicans resistant trains have shown acid sequence of CYP51 (R467K and G464S,
defective sterol C5-desaturase activities, including respectively) ^^^' ^^^. Overexpression of CYP51
some from AIDS patients and from patients suf- on plasmids causes only a small increase in
fering infections as a result of leukemia^^^' ^^^. resistance, so this is not definitively identified as
Changes in CYP51 expression have been a cause of resistance ^^^. In a different setting, a
detected in resistant strains, but the role in causing Penicillium digitanum resistant strain exhibited
resistance is unclear. For instance, in two studies five repeats of a 126 bp sequence in the CYP51
608 Steven L. Kelly et al.

promoter of a resistant strain, in comparison to Table 13.5. A List of Amino acid


one in a sensitive strain, and this may represent a Substitutions Observed in CYP51 Among
resistance mechanism that results in enhanced Clinical Fluconazole Resistant Candida albicans
CYP51 expression^^^. A. fumigatus has two
CYP51 genes, as in other aspergilH^^^, and this CYP51 substitution References
could also be the mechanism that makes it insen-
sitive to fluconazole, although itraconazole resist- F72L FsvrQetai, 1999
ance in A. fumigatus may be the result of F105L Loefflere/fl/., 1997
mechanisms similar to those described in more D116E FavTQetal., 1999
detail here for C albicans^^^. F126L FsLYTQetai, 1999
K128T Sanglarde^fl/., 1998
An isolate of S. cerevisiae, SGI, defective in G129A Sanglard etal, 1998
sterol C14-demethylation, contained the substitu- Y132H Sanglarde/fl/., 1998
tion G310H, had an inactive protein, and was K143E FsiYTQetal., 1999
azole resistant^ ^^. The resistance was due to A149 V Marichal etai, 1999
the second sterol C5-desaturase defect of the D153E Marichale^a/., 1999
strain that suppressed the effect of the block E165Y MahchsAetaL, 1999
in 14-demethylation. 14-Methyl-ergosta-24,(28)- T229A FawQetai, 1999
dien-3,6-diol normally accumulates when 14- E266Q Sanglarde/fl/., 1998
demethylation is blocked, but with a second defect E266D Locmcr etal., 1997
in 5-desaturation, the functional sterol 14- Fa\TQetal., 1999
S279Y Marichale^fl/., 1999
methylfecosterol accumulates instead^^^. Other
K287R Loefflere/a/., 1997
circumstantial evidence for mutant CYP51 result- G307S Pereae^fl/.,2001
ing in azole resistance was observed^^^, but it was S405Y Favre etal., 1999
not until 1997 that the first published evidence of S405F Sanglarde^fl/., 1998
changes in C. albicans CYP51 as a cause of resist- Fawre etal., 1999
ance in practical settings appeared. These were V437I Sanglard etal., 1998
reported for laboratory studies that had indicated FavTQetaL, 1999
that the change T315A in C albicans CYP51 G448E Loefflere/a/., 1997
could alter fluconazole resistance when expressed F449L FavrQetaL, 1999
in S. cerevisiae^^^. However, in a study of 18 G450E Loefflere/a/., 1997
resistant and 18 fluconazole resistant C albicans V452A Marichale/a/., 1999
G464S Loefflere/fl/., 1997
isolates, a number of amino acid substitutions
Sanglard etal., 1998
were observed in the resistant cohort suggestive Marichale/fl/., 1999
of a causal link (Table 13.5). These included the G465S Loemer etai, 1997
change G464S observed in many subsequent Marichale/a/., 1999
investigations'^^ One particular problem was the R467K White, 1997
absence of a parental isolate to compare resistant Sanglard etal., 1998
strains to, but a number of studies allowed 147 IT Kakeyae^«/.,2000
sequential sets to be investigated that were V488I Loemcr etal., 1997
obtained during the treatment of a patient and the
emergence of resistance. In one set of 17 isolates, Notes: Not all these changes are known to cause resistance and
only Y132H, G464S, and R467K have been subject to investiga-
a number of molecular changes were observed, tion at the level of protein.
including increased transcription of Cdrlp ABC
transporter that can confer cross-resistance to
other azoles, and of Mdrlp, a major facilitator
transporter, that confer fluconazole resistance'^'. biosynthesis in cell-free extracts and later in stud-
During the study, increased transcript levels of ies on the protein after expression in S. cerevisiae^^^.
CYP51 were also observed in later isolates. Another matched-set of C. albicans detected the
Furthermore, the substitution R467K in CYP51 appearance of G464S during the emergence of
produced an active protein with reduced affinity resistance^^'*. As noted by White ^^^ and Loeffler
for fluconazole, as demonstrated by in vitro sterol et al}^^ the presence of homozygosity in this
The Diversity and Importance of Microbial Cytochromes P450 609

diploid yeast implied two molecular events in be related to resistance and located in the I-helix.
the selection of resistant CYP51, an initial Another cluster of residues associated with
forward mutation followed by gene conversion of fluconazole resistance in C albicans CYP51 are
the second allele or (less likely on frequency located before the heme-binding region consisting
grounds) a second identical mutational event in of G448E, F449L, G450E, and V452A, while
the second allele. This might be expected to pro- G464S, G465S, R467K, and I471T are adjacent to
duce higher level resistance than the heterozygous the heme-cysteinyl ligand C470. While all these
condition. Further point mutations in CYP51 from changes, together with fiirther numerous unpub-
fluconazole-resistant C. albicans were reported lished alterations detected recently, need fiirther
and yeast expression used to assess the resistance characterization to define their effects on flucona-
phenotype associated with the altered proteins zole binding, only F126, G464, and R467 are con-
and the altered sensitivity^ ^^. In many cases, served across CYPSls. The numbers of mutants
more than one change from "wild-type" sequence found with G464S and R467K were a surprise, as
existed and this allowed additive resistance fluconazole binds above the heme as a sixth ligand
changes and neutral CYP51 polymorphisms to be with interaction of the N-1 substituent group
detected. It is possible, as in reverse transcriptase within the active site. These substitutions below
substitutions in drug-resistant HIV, that selection the plane of the heme are presumed to result in
could also involve changes that alter activity of a change in the plane or orientation of the heme
the resistant protein and assist fitness. and to have a knockon effect on the location
Since these studies, a number of others have of the fluconazole bound above the heme^^^' ^^^.
identified point mutations implicated in resist- Surprisingly, in plant pathogens only a change in
ance using either in vitro sterol biosynthesis on the equivalent residue to Y132 in C. albicans has
cell-free extracts of the C. albicans strains, or so far been detected in grape powdery mildew
more specifically, expression of the respective among 19 resistant isolates ^^^. This could make a
CYPSls and sensitivity testing using a hetero- simple diagnostic test for resistance feasible, as
logous host, S. cerevisiae. These studies have was also explored by Loeffler et al}^^ but the
revealed a hugely diverse series of changes diversity of changes found in C albicans CYP51
scattered across the protein and a list of these make this more complicated for clinical use.
is included in Table 13.4i48, i54, i63-i65 g^rly To complete an understanding of the structural
molecular modeling of the CYP51 protein based basis of resistance, biophysical studies will be
on the CYPlOl structure did not predict these required. A structure for a mammalian endoplasmic
residues as being important in azole interac- reticulum associated GYP has been achieved
tion^ ^^, although later models took into account through expression of a soluble derivative and its
a number of prokaryotic structures and existing crystallization^ ^^' ^^^. So far no reports of using
information on fluconazole resistance muta- E. coli and CYP51 for this purpose have occurred,
tions ^^^. Interestingly, the latter model predicted a but a soluble derivative that is active has been
kink in the C. albicans CYP51 I-helix, and in the produced by expression of CYP51 containing an
structure of M tuberculosis CYP51 a break in the engineered protease site beyond the N-terminal
helix was found"^^. membrane anchor. This may provide the route to
Of the mutations observed to-date, the helices resolving the structure if crystals can be obtained^^^.
B and B', F126L, K128T, G129A, Y132H, The nature of the diverse mutations giving rise
K143R, F145L, and K147R would be close to the to fluconazole resistance in C. albicans is remark-
access channel. The Y132H mutation has been able, but mutations arose mainly in AIDS patients
detected in a number of studies as well as in com- where resistance occurred in >10% during
bination with different substitutions^^^' ^^^' ^^^, and extended and prophylactic treatment^ ^ With
upon expression in yeast confers a 4-fold increase effective HIV treatment, this source of isolates has
in fluconazole resistance and cross-resistance to diminished as the immunocompetence of patients
itraconazole (2-fold) and ketoconazole (16- has improved. There will remain a problem, how-
fold)i52 E266D, R276H, D278E, and S279F are ever, with continuation of azole use and
located at the end of the G-helix and G307S is development, and many facets of the resistance
the first substitution observed in the clinic to mechanisms remain to be unraveled, including
610 Steven L. Kelly et a/.

precise roles and effects of the diverse mutations Council, Natural and Environmental Research
in CYP5L Council, The Wellcome Trust, and The Wolfson
Foundation.

8. Conclusions
References
Microbial CYP biodiversity is presenting an
unexpected challenge as the numbers of CYPs 1. Klingenberg, M. (1958). Pigments of rat liver
microsomes. Arch. Biochem. Biophys. 75, 376-386.
uncovered makes functional investigation through
2. Omura, T. and R. Sato (1962). A new cytochrome in
high-throughput genomics mandatory in addition to liver microsomes, o^ Biol. Chem. 237, 1375-1376.
the more traditional, but essential approaches. The 3. Estabrook, R.W., D.Y. Cooper, and O. Rosenthal
most facile organisms to study are the model organ- (1963). The light reversible carbon monoxide inhi-
isms with well-established genetic systems, but bition of the steroid C21-hydroxylase system of the
many of the most interesting in terms of their CYP adrenal cortex. Biochem. Z. 338, 741.
complements and environmental relevance, such as 4. Lu, A.YH., K.W. Junk, and M.J. Coon (1969).
P. chrysosporium, will be important to tackle even Resolution of the cytochrome P450 in fatty acid
if more recalcitrant. With many CYPs involved w-hydroxylation system into three components.
in metabolic pathways, an emphasis on changing J. Biol. Chem. 243, 1331-1332.
CYPs to alter end-products will emerge. One of the 5. Hanukoglu, I. (1992). Steroidogenic enzymes:
Structure, function and regulation of expression.
most ambitious metabolic engineering projects to
J. Steroid Biochem. Mol. Biol. 43, 779-804.
date involved the diversion of yeast ergosterol 6. Lindenmeyer, A. and L. Smith (1964).
biosynthesis toward corticosteroid production, Cytochromes and other pigments of bakers yeast
which involved deleting and adding CYPs to trans- grown aerobically and anaerobically. Biochem.
genic yeast^^. Coupled to the ability to improve Biophys. Acta. 93, 445-^61.
enzymes through processes of directed evolution, 7. Appleby, C.A. (1967). A soluble haemoprotein
much will be possible in pathway improvement and P450 from nitrogen-fixing rhizobium bacteroids.
invention as well as in biocatalysis^^^ Biochem. Biophys. Acta. 147, 399-^02.
Comparative genomics between close species 8. Katagiri, M., B.N. Ganguli, and I.C. Gunsalus
(1968). A soluble cytochrome P450 functional
and within strains will add to our information
in methylene hydroxylation. J. Biol. Chem. 243,
on the biological roles and evolution of CYPs. 3543-3546.
Contributions from currently unrepresented 9. Tyson, C.A., J.D. Lipscomb, and I.C. Gunsalus
organisms from the fungi, protista, and algae will (1972). The role of putidaredoxin and P450 cam in
be of great interest in understanding CYP evolu- methylene hydroxylation. J. Biol. Chem. 247,
tion. With 42 CYPs already detected in the slime- 5777-5784.
mould Dictyostelium discoidium, other protists 10. Rheinwald, J.G., A.M. Chakrabarty, and I.C.
are likely to have large numbers of CYPs. Where Gunsalus (1973). A transmissible plasmid control-
they are pathogenic, such as Leischmeinia and ling camphor oxidation in Pseudomonas putida.
Trypanosoma, there will be the added interest of Proc. Natl. Acad Sci. USA 70, 885-889.
developing drugs to target these diseases via CYP 11. Sligar, S.G., RG. Debrunner, J.D. Lipscomb, M.J.
Namtvedt, and I.C. Gunsalus (1974). A role of
inhibition, and azoles are already being used. The
the putidaredoxin COOH-terminus in P-450cam
huge wave of CYP information from these pro- (cytochrome m) hydroxylations. Proc. Natl. Acad.
jects will provide a major impetus to finding ways Sci. t/^^ 71, 3906-3910.
of establishing function and biotechnological uses 12. Koga, H., B. Rauchfuss, and I.C. Gunsalus
in a systematic manner. (1985). P450cam gene cloning and expression in
Pseudomonas putida and Escherichia coli.
Biochem. Biophys. Res. Commun. 130, 412-417.
13. Narhi, L.O. and A.J. Fulco (1982). Phenobarbital
Acknowledgments induction of a soluble cytochrome P450 depend-
ent fatty acid monooxygenase from Bacillus
Research in the Wolfson Laboratory of P450 megaterium. J. Biol. Chem. 257, 2147-2150.
Biodiversity is currently supported by the 14. Kelly, S.L., D.C. Lamb, M. Cannieux, D. Greetham,
Biotechnology and Biological Science Research C.J. Jackson, T. Marczylo et al. (2001). An old
The Diversity and importance of IS^icrobiai Cytochromes P450 611

activity in the P450 superfamily (CYP51) and a 26. Roberts, G.A., G. Grogan, A. Greter, S.L. Flitsch,
new story of drugs and resistance. Biochem. Soc. and N.J. Turner (2002). Identification of a new class
Trans. 29, 122-128. of cytochrome P450 from a Rhodococcus sp.
15. Nelson, D.R., L. Koymans, T. Kamataki, J.J. J. Bact. 184, 3898-3908.
Stegeman, R. Feyereisen, D.J. Waxman et al. 27. Jackson, C.l, D.C. Lamb, T. Marczylo, A.G.S.
(1996). P450 superfamily: Update on new Warrilow, N.J. Manning, D.J. Lowe et al. (2002). A
sequences, gene mapping, accession numbers and novel sterol 14a-demethylase/ferredoxin fusion
nomenclature. Pharmacogenetics 6, 1-42. protein (MCCYP51FX) from Methylococcus
16. Kelly, S.L., D.C. Lamb, and D.E. Kelly (1999). capsulatus represents a new class of the cyto-
Inhibitors of CYP51 as antifungal agents and chrome P450 superfamily. J. Biol. Chem. Ill,
resistance to azole antifungals. In (E. Arinc, J.B. 46959-^6965.
Schenkman, and E. Hodgson (eds). Molecular 28. Seth-Smith, H.M.B., S.J. Rosser, A. Basran,
and Applied Aspects of Oxidative Drug E.R. Travis, E.R. Dabbs, S. Nicklin et al. (2002).
Metabolising Enzymes. Plenum, New York, pp. Cloning, sequencing and characterisation of the
157-172. hexahydro-1,3,5-trinitro-1,3,5-triazine degradation
17. Szczebara, P.M., C. Chandelier, C. Villeret, gene cluster from Rhodococcus rhodochrous. Appl.
A. Masurel, S. Bourot, C. Duport et al. (2003). Env Microbiol. 68, 4764-^771.
Total biosynthesis of hydrocortisone from a simple 29. De Mot, R. and A.H. Parret (2002). A novel class
carbon source. Nat. Biotechnol 21, 143-149. of self-sufficient cytochrome P450 monooxy-
18. Rodriguez-Saiz, M., J.L. Barredo, M.A. Moreno, genase in prokaryotes. Trends Microbiol. 10,
J.M. Fernandez-Canon, M.A. Penalva, and B. Diez 502-508.
(2001). Reduced function of a phenylacetate- 30. Gonzalez, FJ. and D.W. Nebert (1990). Evolution
oxidising cytochrome P450 caused by strong of the P450 gene superfamily: Animal-plant war-
genetic improvement in early phytogeny of peni- fare, molecular drive and human genetic differ-
cillin-producing strains. J. Bact. 183, 5465-5471. ences in drug oxidation. Trends Genet. 6, 182-186.
19. Mingot, J.M., M.A. Penalva, and J.M. Fernandez- 31. Nelson, D.R. (1999). Cytochrome P450 and the
Canon (1999). Disruption of phacA, diW Aspergillus individuality of species. Arch. Biochem. Biophys.
nidulans gene encoding a novel cytochrome 369, 1-10.
P450 monooxygenase catalysing phenylacetate 32. Yoshida, Y, Y. Aoyama, M. Noshiro, and O. Gotoh
2-hydroxylation results in penicillin overproduc- (2000). Sterol 14-demethylase P450 (CYP51) pro-
tion. J. Biol. Chem. 214, 14545-14550. vides a breakthrough for the discussion of the
20. Smith, K.E., R Ahmed, and T. Antoniou (1993). evolution of cytochrome P450 gene superfamily.
Microbial transformation of steroids. Biochem. Biochem. Biophys. Res. Commun. 213, 799-804.
Soc. Trans. 21, 1077-1080. 33. Jackson, C.J., DC. Lamb, T.H. Marczylo,
21. Lupetti, A., R. Danesi, M. Campa, M. Del Tacca, J.E. Parker, N.J. Manning, D.E. Kelly et al. (2003).
and S. Kelly (2002). Molecular basis of resistance Conservation and cloning of CYP51, a sterol 14a-
to azole antifungals. Trends Mol. Med. 8, 76-81. demethylase from Mycobacterium smegmatis.
22. Wen, L.R and A.J. Fulco (1987). Cloning of the Biochem. Biophys. Res. Commun. 307, 558-563.
gene encoding a catalytically self-sufficient cyto- 34. Stoke, J.E. and J.J. De Voss (2000). Expression,
chrome P450 fatty-acid monooxygenase induced purification and characterisation of biol: A carbon-
by barbiturates in Bacillus megaterium and its carbon bond cleaving cytochrome P450 involved in
functional expression and regulation in heterolo- biotin biosynthesis in Bacillus subtilis. Arch.
gous (Escherichia coli) and homologous {Bacillus Biochem. Biophys. 384, 351-360.
megaterium) hosts. J. Biol. Chem. 262, 6676-6682. 35. Kalb, VF, C.W Woods, T.G. Turi, C.R. Dey,
23. Park, S.Y, H. Shimizu, S. Adachi, A. Nakagawa, T.R. Sutter, and J.C. Loper, (1987). Primary structure
I. Tanaka, K. Nakahara et al. (1997). Crystal structure of P450 lanosterol demethylase gene from
of nitric oxide reductase from denitrifying fungus Sacchaaromyces cerevisiae. DNA 6, 529-537.
Fusarium oxysporum. Nat. Struct. Biol. 4, 827-832. 36. Aoyama,Y, Y Funae, M. Noshiro, T. Horiuchi, and
24. Kelly, S.L., D.C. Lamb, C.J. Jackson, A.G.S. Y Yoshida (1994). Occurence of a P450 showing
Warrilow, and D.E. Kelly (2003). The biodiversity high homology to yeast lanosterol 14-demethylase
of microbial cytochromes P450. Adv. Microb. (P45014DM) in the rat liver. Biochem. Biophys.
Physiol. 47, 131-186. Res. Commun. 201, 1320-1326.
25. Hawkes, D.B., G.W. Adams, A.L. Burlingame, 37. Stromstedt, M., D. Rozman, and M.R. Waterman
PR.O. de Montellano, and J.J. De Voss (2002). (1996). The ubiquitously expressed human CYP51
Cytochrome P450(cin) (CYP176A1), isolation, encodes lanosterol 14-demethylase, a cytochrome
expression and characterization. J. Biol. Chem. 277, P450 whose expression is regulated by oxysterols.
27725-27732. Arch. Biochem. Biophys. 329, 73-81.
612 Steven L. Kelly et aL

38. Bak, S., R.A. Kahn, C.E. Olsen, and B. Halkier 50. Lamb, D.C, K. Fowler, T. Kieser, N.J. Manning,
(1997). Cloning and expression in E. coli of L.M. Podust, M.R. Waterman et al. (2002).
the obtusifoliol of Sorghum bicolor (L.) Moench, Sterol 14a-demethylase activity in Streptomyces
a cytochrome P450 orthologous to the sterol coelicolor A3(2) is associated with an unusual
14a-demethylases (CYP51) from mammals and member of the CYP51 gene family. Biochem. J.
fmigl Plant J. n, 191-201. 364, 555-562.
39. Lamb, D.C., D.E. Kelly, and S.L. Kelly (1998). 51. Jackson, C.J., D.C. Lamb, D.E. Kelly, and S.L.
Molecular diversity of sterol 14a-demethylase sub- Kelly (2000). Bactericidal and inhibitory effects of
strates in plants, fungi and humans. FEBSLett. 425, azole antifungal compounds on Mycobacterium
263-265. smegmatis. FEMS Lett. 192, 159-162.
40. Wickramashighe, R.H. and C.A. Villee (1975). 52. Guardiola-Diaz, H.M., L.A. Foster, D. Mushrush,
Early role during chemical evolution for cyto- and A.D.N. Vaz (2001). Azole-binding to a novel
chrome P450 in oxygen detoxification. Nature 256, cytochrome P450 from Mycobacterium tuberculosis:
509-510. Implications for treatment of tuberculosis.
41. Godzina, S.M., M.A. Lovato, M.M. Meyer, K.A. Biochem. Pharmacol. 61, 1463-1470.
Foster, W.K. Wilson, W. Gu et al (2000). Cloning 53. McLean, K.J., M.R. Cheeseman, S.L. Rivers,
and characterisation of the Dictyostelium dis- A. Richmond, D. Leys, S.K. Chapman etal. (2002).
coideum. Lipids 35, 249-255. Expression, purification and spectroscopic charac-
42. Joubert, B.M., ES. Buckner, and S.P.T. Matsuda terisation of the cytochrome P450 CYP121 from
(2001). Trypanosome and animal lanosterol syn- Mycobacterium tuberculosis. J. Inorg. Biochem. 91,
thase use different catalytic motifs. Org. Lett. 3, 527-541.
1957-1960. 54. Cole, S.T., K. Eiglmeier, J. Parkhill, K.D. James, N.R.
43. Bellamine, A., A.T. Mangla, W.D. Nes, and M.R. Thomson, PR. Wheeler et al. (2001). Massive gene
Waterman (1999). Characterisation and catalytic decay in leprosy bacillus. Nature 409, 1007-1011.
properties of the sterol 14a-demethylase from 55. Tang, L., S. Shah, L. Chung, J. Carney, L. Katz,
Mycobacterium tuberculosis. Proc. Natl. Acad. Sci. C. Khosla et al. (2000). Cloning and expression of
USA 96, 8937-8942. the epothilone gene cluster. Science 287, 640-642.
44. Podust, L.M., T.L. Poulos, and M.R. Waterman 56. August, PR., L. Tang, Y.J. Yoon, S. Ning, R. Muller,
(2001). Crystal structure cytochrome P450 14a- T-W Yu et al. (1998). Biosynthesis of the
sterol demethylase from Mycobacterium tuberculo- ansamycin antibiotic rifamycin: Deductions from
sis in complex with azole inhibitors. Proc. Natl. the molecular analysis of the rif biosynthetic
Acad Sci. USA 98, 3068-3073. cluster of Amycolatopsis mediteranei S699. Chem.
45. Bird, C.W, J.M. Lynch, RJ. Pirt, WW Reid, C.J.W Biol. 5, 69-79.
Brooks, and B.S. Middleditch (1971). Steroids and 57. Poulos, TL., B.C. Finzel, I.C. Gunsalus, G.C.
squalene in Methylococcus capsulatus grown on Wagner, and J. Kraut (1985). The 2.6A crystal
methane. Nature 230, A12i-A1A. structure of Pseudomonas putida cytochrome
46. Bode, H.B., B. Zeggel, B. Silakowski, S.C. Wenzel, ?450. J. Biol. Chem. 260, 16122-16130.
H. Reichenbach, and R. Muller (2003). Steroid 58. Poulos, T.L., B.C. Finzel, and A.J. Howard (1987).
biosynthesis in prokaryotes: Identification of High-resolution crystal structure of cytochrome
myxobacterial steroids and cloning of the first P450cam. J. Mol. Biol. 195, 687-700.
bacterial 2,3(S)-oxidosqualene cyclase from the 59. Peterson, J.A., J.Y Lu, J. Geisselsoder, S. Graham
myxobacterium Stigmatella aurantiaca. Mol. Lorence, C. Carmona, F. Witney et al. (1992).
Microbiol. 41, 47\^S\. Cytochrome P450 terp-isolation and purification of
47. Buckner, ES., A.J. Wilson, T.C. White, and WC. the protein and cloning and sequencing of its
Van Voorhis (1998). Induction of resistance to azole oipQxon. J. Biol. Chem. 267, 14193-14203.
drugs in Trypanosoma cruzi. Antimicrob. Agents 60. Nagy, I., F. Compemolle, K. Ghys, J. VanderLeyden,
Chemother. 42, 3245-3250. and R. Demot (1995). A single cytochrome P450
48. Salimova, E., A. Boschetti, W Eichenberger, and system is involved in degradation of the herbicides
L. Lutova (1999). Sterol mutants of Chlamy- EPTC (S-ethyl dipropylthiocarbamate) and atrazine
domonas reinhardtii: Characterisation of three by Rhodococcus sp. strain NI86/21. J. Bact. 61,
strains deficient in C24(28) reductase. Plant 2056-2060.
Physiol. Biochem. 37, 241-249. 61. O'Keefe, DP, J.M. Tepperman, C. Dean, K.J. Leto,
49. Kelly, S.L., D.C. Lamb, and D.E. Kelly (1997). D.L. Erbes, and J.T. O'Dell (1994). Plant expres-
Sterol 22-desaturase, cytochrome P450 61, pos- sion of a bacterial cytochrome P450 that catalyzes
sesses activity in xenobiotic metabolism FEBSLett. activation of a sulfonylurea pro-herbicide. Plant
412, 233-235. Physiol. 105, 473-482.
The Diversity and Importance of iVIicrobial Cytochromes P450 613

62. Ruettinger, R.T. and A.J. Fulco (1981). Epoxidation P450 complement of the avermectin-producer
of unsaturated fatty acids by a soluble cyto- Strepto-myces avermitilis and comparison to that of
chrome P-450-dependent system from Bacillus Streptomyces coelicolor A3(2). Biochem. Biophys.
megaterium. J. Biol. Chem. 256, 5728-5734. Res. Commun. 307, 610-619.
63. Narhi, L.O. and A.J. Fulco (1986). Characterization 75. Sariaslani, F.S. and J.R Rosazza (1983). Novel
of a catalytically self-sufficient 119,000-dalton biotransformations of 7-ethoxycoumarin by
cytochrome P-450 monooxygenase induced by Streptomyces griseus. Appl. Environ. Microbiol. 46,
barbiturates in Bacillus megaterium. J. Biol. Chem. 468-474.
261, 7160-7169. 76. Sariaslani, FS. and J.R Rosazza (1983). Microbial
64. Narhi, L.O. and A.J. Fulco (1987). Identification transformation of natural anti-tumor agents—
and characterization of two functional domains products of rotenone and dihydrorotenone transfor-
in cytochrome P-450BM-3, a catalytically self- mation. Appl. Environ. Microbiol. 49, 451-452.
sufficient monooxygenase induced by barbiturates in 77. Trower, M.K., R. Lenstra, C. Omer, S.E. Buchholz,
Bacillus megaterium. J. Biol. Chem. 262, 6683-6690. and F.S. Sariaslani (1992). Cloning, nucleotide
65. Ravichandran, K.G., S.S. BoddupalH, C.A. sequence determination and expression of the
Hasermann, J.A. Peterson, and J. Deisenhofer genes encoding cytochrome P450soy (SOYC) and
(1993). Crystal structure of hemoprotein domain of ferredoxinSOY (SOYC) from Steptomyces griseus.
P450BM-3, a prototype for microsomal P450's. Mol. Microbiol. 6, 2125-2134.
Science 16\,n\-12>6. 78. O'Keefe, D.R, S.M.C. Lau, and PA. Harder
66. Munro, A.W. (1994). The role of Tryptophan 97 of (1993). Efficiency and substrate-specificity of
cytochrome P450 BM3 from Bacillus megaterium Streptomyces griseolus cytochrome P450SU1 and
in catalytic function. Biochem. J. 303, 423^28. cytochrome P450SU2 monooxygenase reactions.
67. Yeom, H., S.G. Sligar, H.Y. Li, TL. Poulos, and A.J. Biochem. Soc. Trans. 21, 1073-1077.
Fulco (1995). The role of Thr268 in oxygen activa- 79. Poupin, P., V Ducrocq, S. Hallier-Soulier, and
tion in cytochrome P450 BM3. Biochemistry 34, N. Truffeut (1999). Cloning and characterisation of
14733-14740. the genes encoding a cytochrome P450 (PipA)
68. Klein, M.L. and A.J. Fulco (1993). Critical residues involved in piperidine and pyrrolidine utilisa-
involved in FMN binding and catalytic activity in tion and its regulatory protein (PipR) in Myco-
cytochrome P450BM3. J. Biol. Chem. 268, bacterium smegmatis mc(2)155. J. Bacteriol. 181,
7553-7561. 3419-3426.
69. Yano, J.K., L.S. Koo, D.J. SchuUer, H. Li, PR. Ortiz 80. Cannell, R.J.P, A.R. Knaggs, M.J. Dawson,
de Montellano, and T.L. Poulos (2000). Crystal G.R. Manchee, P.J. Eddershaw, L Waterhouse et al.
structure of a thermophilic cytochrome P450 from (1995). Microbial biotransformation of the
the archaeon Sulfolobus solfataricus. J. Biol. Chem. angiotensin II antagonist GRl 17289 by
275,31086-31092. Streptomyces rimosus to identify a mammalian
70. Yano, J.K., F. Blasco, H. Li, R.D. Schmid, metabolite. Drug Metab. Disp. 23, 724-729.
A. Henne, and T.L. Poulos (2003). Preliminary 81. Berrie, J.R., R.A.D. Williams, and K.E. Smith
characterization and crystal structure of a ther- (1999). Microbial transformations of steroids-XI.
mostable cytochrome P450 from Thermus ther- Progesterone transformation by Streptomyces roseo
mophilus. J. Biol. Chem. 278, 608-616. chromogenes-purification and characterisation of
71. Cole, S.T, R. Brosch, J. Parkhill, T Gamier, the 16 alpha-hydroxylase system. J. Steroid
C. Churcher, D. Harris et al. (1998). Deciphering Biochem. 71, 153-165.
the biology of Mycobacterium tuberculosis from the 82. Ito, S., T. Hata, I. Watanabe, T. Matsuoka, and N.
complete genome sequence. Nature 393, 537-554. Serizawa (1997). Crystal structure of cytochrome
72. Bentley, S.D., K.F Chater, A.M. Cerdeno-Tarraga, P450sca from Streptomyces carbophilus involved
G.L. Challis, N.R. Thomson, K.D. James et al. in production of pravastatin, a cholesterol lowering
(2002). Complete sequence of the model actino- drug.E^^^-^J. 11, 36.
myctete Streptomyces coelicolor A3(2). Nature 83. Ikeda, H., J. Ishikawa, A. Hanamoto, M. Shinose,
417,141-147. H. Kikuchi, T. Shiba et al (2003). Complete
73. Lamb, D.C., T. Skaug, H.-L. Song, C.J. Jackson, genome sequence and comparative analysis of the
L. Podust, M.R. Waterman et al (2002). The industrial microorganisms Streptomyces avermitilis
cytochrome P450 complement (CYPome) of Organism. Nat. Biotechnol 21, 526-531.
Streptomyces coelicolor A3(2). J. Biol. Chem. Ill, 84. Podust, L.M., Y Kim, M. Arase, B.A. Neely,
24000-24005. B.J. Beck, H. Bach et al (2003). The 1.92-
74. Lamb, D.C., H. Ikeda, D.R. Nelson, J. Ishikawa, angstrom structure of Streptomyces coelicolor
T. Skaug, Jackson C.J. et al. (2003). Cytochrome A3(2) CYP154C1—a new monooxygenase that
614 Steven L. Kelly ef a/.

functionalises macrolide ring systems. J. Biol. 96. Sanglard, D. and J.C. Loper (1989). Characterisa-
Chem.nS, 12214-12221. tion of the alkane-inducible cytochrome P450
85. Komatsu, M., Y. Kuwara, A. Hiroishi, K. Hosono, (P450ALK) gene from the yeast Candida
T. Beppu, and K. Ueda (2003). Cloning of the tropicalis—identification of a new P450-gene
conserved regulatory operon by its aerial family. Gene 76, 121-136.
mycelium-inducing activity in an amfR mutant of 97. Ohkuma, M., T. Zimmer, T. lida, W.H. Schunck,
Streptomyces griseus. Gene 306, 79-89. A. Ohta, and M. Takagi (1998). Isozyme function
86. Briza, P., M. Eckerstorfer, and M. Breitenbach of n-alkane-inducible cytochromes P450 in
(1994). The sporulation-specific enzymes encoded Candida maltosa revealed by sequential gene dis-
by the DITl and DIT2 genes catalyze a two-step ruption. J. Biol. Chem. 273, 3948-3953.
reaction leading to a soluble LL-dityrosine-contain- 98. lida, T., A. Ohta, and M. Takagi (1998). Cloning and
ing precursor of the yeast spore wall. Proc. Natl. characterisation of an n-alkane inducible cyto-
Acad. Sci. USA 91, 4524-4528. chrome P450 gene essential for n-decane assimila-
87. Kelly, S.L., D.C. Lamb, A.J. Corran, B.C. Baldwin, tion by Yarrowia lipolytica,. Yeast 14, 1387-1397.
L.W. Parks, and D.E. Kelly (1995). Purification and 99. VanGorcom, R.F, J.G. Boschloo, A.
reconstitution of activity of Saccharomyces cere- Kuijvenhoven, J. Lange, A.J. vanVark, C.J. Bos
visiae P450 61, a sterol 22-desaturase. FEBS Lett. et al. (1990). Isolation and molecular characterisa-
311, 217-220. tion of the benzoate para-hydroxylase gene (bphA)
88. Small, E.H., P. Briza, A. Panagos, and L. Berenfeld of Aspergillus niger: A member of a new gene
(1995). Candida albicans cell walls contain the family of the cytochrome P450 superfamily Mol.
fluorescent cross-linking amino acid dityrosine. Gen. Genet. 223, 192-197.
Inf. Immun. 63, 4078-4083. 100. Attar, R.M., E. Grotewold, G.E. Taccioli, G.O.
89. Masaphy, S., D. Levanon, Y. Henis, Aisemberg, H.N. Torres, and N.D. Judewicz (1989).
K. Venkateswariu, and S.L. Kelly (1996). Evidence A cycloheximide-inducible gene of Neurospora
for cytochrome P450 and P450-mediated crassa belongs to the cytochrome P450 superfam-
benzo(a)pyrene hydroxylation in the white-rot fun- ily Nucleic Acids Res. 17, 7535-7536.
gus Phanerochaete chrysosporium. FEMS Lett. 101. Han, Y.N., X.G. Liu, U. Benny, H.C. Kistler, and
135,51-55. H.D. VanEtten (2001). Genes determining patho-
90. Kullman, S.W. and F. Matsumura (1997). genicity to pea are clustered on a supernumerary
Identification of a novel cytochrome P-450 gene chromosome in the ftingal plant pathogen Nectria
from the white-rot fungus Phanerochaete haematococca. Plant J. 25, 305-314.
chrysosporium. Appl. Environ. Microbiol. 63, 102. Hata, S., T. Nishino, M. Komori, and H. Katsuki
2741-2746. (1981). Involvement of cytochrome P-450 in delta
91. Yadav, J.S. and J.C. Loper (2000). Cytochrome 22-desaturation in ergosterol biosynthesis of yeast.
P450 oxidoreductase gene and its differentially Biochem. Biophys. Res. Commun. 103, 272-277.
terminated cDNAs from the white-rot fungus 103. Skaggs, B.A., J.F. Alexander, C.A. Pierson,
Phanerochaete chrysosporium. Curr. Genet. 37, K.S. Schweitzer, K.T Chun, C. Koegel etal. (1996).
65-73. Cloning and characterization of the Saccharomyces
92. Warrilow, A.G.S., D.C. Lamb, D.E. Kelly, and cerevisiae C-22 sterol desaturase gene, encoding a
S.L. Kelly (2002). Phanerochaete chrysosporium second cytochrome P-450 involved in ergosterol
NADPH-cytochrome P450 reductase kinetic mech- biosynthesis. Gene 169, 105-109.
anism. Biochem. Biophys. Res. Commun. 299, 104. Kelly S.L., D.C. Lamb, B.C. Baldwin, A.J. Corran,
189-195. and D.E. Kelly (1997). Characterisation of
93. Breskvar, K., B. Cresnar, and T. Hudnik-Plevnik Saccharomyces cerevisiae CYP61, a sterol delta
(1987). Resolution and reconstitution of 22-desaturase, and inhibition by azole antifimgal
cytochrome P450 containing steroid hydroxylation agents. J. Biol. Chem. 272, 9986-9988.
system of Rhizopus nigricans. J. Steroid Biochem. 105. Noda, H. and Y Koizumi (2003). Sterol biosynthe-
14,395-399. sis by symbiotes: Cytochrome P450 sterol C5-
94. Masaphy, S., D.C. Lamb, and S.L. Kelly (1999). desaturase genes from yeastlike symbiotes of rice
Purification and characterization of a benzo(a)pyrene planthoppers and anobiid beetles. Insect Biochem.
hydroxylase from Pleurotus pulmonarius. Biochem. Mol. Biol. 33, 649-658.
Biophys. Res. Commun. 266, 326-329. 106. Hohn, T.M., A.E. Desjardins, and S.P McCormick
95. Lebeault, J.M., E.T. Lode, and M.J. Coon (1971). (1995). The TRI4 gene of Fusarium sporotri-
Fatty acid and hydrocarbon hydroxylation in yeast: choides encodes a cytochrome P450 monooxyge-
Role of cytochrome P450 in Candida tropicalis. nase involved in trichothene biosynthesis. Mol.
Biochem. Biophys. Res. Commun. 42, 413^19. Gen. Genet. 248, 95-102.
The Diversity and Importance of IVIicrobial Cytochiromes P450 615

107. Yu, JJ., RK. Chang, K.C. Ehrlich, B. Montalbano, 119. Aoyama, Y, Y Yoshida, Y Sonoda, and Y Sato
J.M. Dyer, D. Bhatnagar et al. (1998). (1989). Deformylation of 32-hydroxy-24,25-
Characterisation of the critical amino acids of dihydrolanosterol by purified cytochrome
an Aspergillus parasiticus cytochrome P450 P45014DM (lanosterol 14a-demethylase) from
monooxygenase encoded by ordA that is involved yeast: Evidence confirming the intermediate step
in the biosynthesis of aflatoxins B-1, G(l), B-2 and of lanosterol 14a-demethylation. J. Biol. Chem.
G(2). Appl Env. Microbiol 64, 4834-4841. 264, 18502-18505.
108. Yu, J., P.K. Chang, D. Bhatnagar, and 120. Akhtar, M., K. Alexander, R.B. Boar, J.F McGhie,
T.E. Cleveland (2000). Genes encoding cyto- and D.H.R. Barton (1978). Chemical and enzymic
chrome P450 and monooxygenase enzymes define studies on the characterisation of intermediates
one end of the aflatoxin pathway gene cluster in during the removal of the 14a methyl group
Aspergillus parasiticus. Appl. Micro. Biotech. 53, in cholesterol biosynthesis. Biochem. J. 169,
583-590. 449^63.
109. Tudzynski, B., P. Hedden, E. Carrera, and 121. Shyadehi, A.Z., D.C. Lamb, S.L. Kelly, D.E. Kelly,
R Gaskin (2001). The P450-4 gene of Gibberella W.H. Schunck, and J.N. Wright et al. (1996). The
fujikuroi encodes ent-kaurene oxidase in the gib- mechanism of the acyl-carbon bond cleavage
berellin biosynthesis pathway App. Environ. reaction catalyzed by recombinant sterol 14a-
Microbiol. 61,3514-3522. demethylase of Candida albicans (other
110. Young, C , L. McMillan, E. Telfer, and B. Scott names are: Lanosterol 14 alpha-demethylase,
(2001). Molecular cloning and genetic analysis of P-45014DM, and CYP51). J. Biol. Chem. Ill,
an indole-diterpene gene cluster from Penicillium 12445-12450.
paxilli. Mol. Microbiol. 39, 754-764. 122. King, D.J., M.R. Azari, and A. Wiseman (1984).
111. Kitazume, T., N. Takaya, N. Nakayama, and Studies on the properties of highly purified cyt.
H. Shoun (2000). Fusarium oxysporum fatty acid P448 and its dependent activity benzxo(a)pyrene
subterminal hydroxylase (CYP505) is a membrane- hydroxylase from S. cerevisiae. Xenobiotica 14,
bound counterpart of Bacillus megaterium 187-206.
P450BM3. J. Biol. Chem. 275, 39734-39740. 123. Callen, D.E and R.M. Philpot (1977). Cyto-
112. Lamb, D.C., D.E. Kelly, and S.L. Kelly (1999). chrome P450 and activation of promutagens in
Molecular aspects of azole antifungal action and Saccharomyces cerevisiae. Mutat. Res. 55,
resistance. Drug Resist. Updates 2, 90-402. 153-163.
113. Odds, F.C. (1996). Antifungal therapy. In 124. Kelly, D. and J.M. Parry (1983). Metabolic activa-
C.C. Kibbler, D.W.R. Mackenzie, and F.C. Odds tion of cytochrome P450/P448 in the yeast
(eds), Principles and Practise of Clinical Saccharomyces cerevisiae. Mutat. Res. 108,
Mycology, John Wiley & Sons, Chichester, pp. 1-6. 147-159.
114. Ragsdale, N.N. and H.D. Sisler (1972). Mode of 125. Kelly, S.L., D.C. Lamb, B.C. Baldwin, and D.E.
action of triarimol in Ustilago maydis. Biochem. Kelly (1993). Benzo[a]pyrene activity in yeast is
Biophys. Res. Commun. 46, 2048-2053. mediated by P450 other than sterol 14a-demethy-
115. Marriott, M. (1980). Inhibition of sterol biosynthe- lase. Biochem. Biophys. Res. Commun. 197,
sis in Candida albicans by imidazole-containing 428-432.
antifungals. J. Gen. Microbiol. Ill, 253-255. 126. Watson, RF, M.E. Rose, S.W Ellis, H. England,
116. Alexander, K., M. Akhtar, R.B. Boar, J.F. McGhie, and S.L. Kelly (1989). Defective sterol C5-6 desat-
and D.H.R. Barton (1972). The removal of the 32- uration and azole resistance: A new hypo-
carbon atom as formic acid in cholesterol biosyn- thesis for the mode of action of azole antifungals.
thesis. J. Chem. Soc. Chem. Commun. 383-385. Biochem. Biophys. Res. Commun. 164, 1170-1175.
117. Aoyama, Y, Y Yoshida, and R. Sato (1984). Yeast 127. Kelly, S.L., A. Amoldi, and D.E. Kelly (1993).
cytochrome P450 catalysing lanosterol 14a- Molecular genetic analysis of azole antifungal
demethylation. II. Lanosterol metabolism by puri- mode of action. Biochem. Soc. Trans. 21,
fied P45014DM and by intact microsomes. J. Biol. 1034-1038.
Chem. 259, 1661-1666. 128. Bard, M., N.D. Lees, T. Turi, D. Craft, L. Cofrin,
118. Aoyama, Y, Y Yoshida, Y Sonoda, and Y Sato R. Barbuch et al. (1993). Sterol synthesis and via-
(1987). Metabolism of 32-hydroxy-24,25- bility of ERG 11 (cytochrome P450 lanosterol
dihydrolanosterol by purified cytochrome demethylase) mutations in Saccharomyces cere-
P45014DM from yeast: Evidence for the contribu- visiae and Candida albicans. Lipids 28, 963-967.
tion of the cytochrome to the whole process of 129. Kelly, S.L., D.C. Lamb, D.E. Kelly, J. Loeffler, and
lanosterol 14a-demethylation. J. Biol. Chem. 262, H. Einsele (1996). Resistance to fluconazole in
1239-1243. Candida albicans from AIDS patients involving
616 Steven L. Kelly et al.

cross-resistance to amphotericin. Lancet 348, genomic analysis of cell division in C. elegans


1523-1524. using RNAi of genes on chromosome III. Nature
130. Nolte, F.S., T. Parkinson, D.J. Falconer, S. Dix, 408,331-336.
J. Williams, C. Gilmore et al. (1997). Isolation and 141. Shen, A.L., K.A. O'Leary, and C.B. Kasper
characterisation of fluconazole and amphotericin (2002). Association of multiple developmental
B-resistant Candida albicans from blood of two defects and embryonic lethality with loss of micro-
patients with leukaemia Antimicroh. Agents somal NADPH-cytochrome P450 oxidoreductase.
Chemother, 44, 196-199. J. Biol. Chem. Ill, 6536-6541.
131. Gachotte, D., C.A. Pierson, N.D. Lees, R. Barbuch, 142. Lamb, D C , D.E. Kelly K. Venkateswarlu, N.J.
C. Koegel, and M. Bard (1997). A yeast sterol aux- Manning, H.F.J. BUgh, W.H. Shunck et al. (1999).
otroph (erg25) is rescued by addition of azole anti- Generation of a complete, soluble and catalytically
fungals and reduced levels of heme. Proc. Natl. active sterol 14a-demethylase-reductase complex.
Acad Sci. USA, 94, 11173-11178. Biochemistry 38, 8733-8738.
132. Nes, D.W., G.G. Janssen, F.G. Cromley, 143. Yoshida, Y. (1988). Cytochrome P450 of fungi:
M. Kalinowska, and T. Akihisa (1993). The struc- Primary target for azole antifungal agents. In M.R.
tural requirements of sterols for membrane function McGinnis (ed.), Current topics in mycology,
in Saccharomyces cerevisiae. Arch. Biochem. Springer-Verlag, New York, p. 388.
Biophys. 300, 724-733. 144. Wilkinson, C.F, K. Hetnarski, and TO. Yellin
133. Bammert, G.F and J.M. Fostel (2000). Genome- (1972). Imidazole derivatives—a new class of
wide expression patterns in Saccharomyces cere- microsomal enzyme inhibitors. Biochem.
visiae: Comparison of drug treatments and genetic Pharmacol. 21, 3\Sl-3\92.
alterations affecting biosynthesis of ergosterol. 145. Aoyama, Y, Y Yoshida, S. Hata, T Nishino, and
Antimicrob. Agents Chemother 44, 1255-1263. H. Katsuki (1983). Buthiobate: A potent inhi-
134. De Backer, M.D., T. Ilyina, X. Ma, S. Vandoninck, bitor for yeast cytochrome P450 catalysing 14a-
W.H.M. Luyten, and H. Vanden Bossche (2001). demethylation of lanosterol. Biochem. Biophys.
Genomic profiling of the response of Candida Res. Commun. 115, 642-647.
albicans to itraconazole treatment using a DNA 146. Lamb, D C , DE. Kelly, M.R. Waterman, M.
microarray. Antimicrob. Agents Chemother. 45, Stromstedt, D. Rozman, and S.L. Kelly (1999).
1660-1670. Characteristics of the heterologously expressed
135. Black, S.D. and M.J. Coon (1982). Structural fea- human lanosterol 14a-demethylase (other names:
tures of hver microsomal NADPH-cytochrome P45014DM, CYP51, P45051) and inhibition of the
P450 reductase; hydrophobic domain, hydrophilic purified human and Candida albicans CYP51 with
domain and connecting region. J. Biol. Chem. 257, azole antifungal agents. Yeast 15, 755-763.
5929-5938. 147. Lamb, D C , M. Cannieux, A.G.S. Warrilow,
136. Sutter, T.C. and J.C. Loper (1989). Disruption of S. Bak, R.A. Kahn and N.J. Manning (2001). Plant
the Saccharomyces cerevisiae gene for NADPH- sterol 14 a-demethylase affinity for azole fungi-
cytochrome P450 reductase causes increased sen- cides. Biochem. Biophys. Res. Commun. 284,
sitivity to ketoconazole. Biochem. Biophys. Res. 845-849.
Commun. 160, 1257-1266. 148. Kakeya, H., Y Miyazaki, H. Miyazaki, K.
137. Goffeau, A., B.G. Barrell, H. Bussey, R.W. Davis, Nyswaner, B. Grimberg, and J.E. Bennett (2000).
B. Dujon, H. Feldman et al. (1996). Life with 6000 Genetic analysis of azole resistance in the
genes. Science 274, 563-567. Darlington strain of Candida albicans. Antimicrob.
138. Venkateswarlu, K., DC. Lamb, D.E. Kelly Agents Chemother 44, 2985-2990.
N.J. Manning, and S.L. Kelly (1998). The N-termi- 149. Venkateswarlu, K., DW Denning, N.J. Manning,
nal membrane domain of yeast NADPH- and S.L. Kelly (1995). Resistance to fluconazole
cytochrome P450 (CYP) oxidoreductase is not in Candida albicans from AIDS patients correlated
required for catalytic activity in sterol biosynthesis with reduced intracellular accumulation of the
or in reconstitution of CYP activity. J. Biol. Chem. drug. FEMS Microbiol. Lett. 131, 337-341.
273, 4492^496. 150. Sanglard, D, K. Kuchler, F. Ischer, J-L. Pagani,
139. Lamb, D.C., D.E. Kelly, N.J. Manning, M.A. M. Monod and J. Bille (1995). Mechanisms of
Kaderbhai, and S.L. Kelly (1999). Biodiversity of resistance to azole antifungal agents in Candida
the P450 catalytic cycle: Yeast cytochrome b^/ albicans from AIDS patients involve specific
NADH cytochrome b^ reductase complex effi- multi-drug transporters. Antimicrob. Agents
ciently drives the entire sterol 14-demethylation Chemother 39, 2378-2386.
(CYP51) reaction. FEBS Lett. 462, 283-288. 151. Sanglard, D., F. Ischer, M. Monod, and J. Bille
140. Gonczy, P., C. Eecheverri, K. Oegema, A. Coulson, (1997). Cloning of Candida albicans genes
S.J.M. Jones R.R. Copley et al. (2000). Functional conferring resistance to azole antifungal
The Diversity and importance of iVIicrobial Cytociiromes P450 617

agents: Characterisation of CDR2, a new mul- 161. Loeffler, J., S.L. Kelly, H. Hebart, U. Schumaher,
tidrug ABC transporter gene. Microbiology 143, C. LassFlorl, and E. Einsele (1997). Molecular
405-416. analysis of CYP51 from fluconazole-resistant
152. Sanglard, D., F. Ischer, L. Koymans, and J. Bille Candida albicans strains. FEMS Lett. 151,
(1998). Amino-acid substitutions in the 263-268.
cytochrome P450 lanosterol 14a-demethylase 162. Lamb, D C , D.E. Kelly, T C White, and S.L. Kelly
(CYP51A1) from azole resistant Candida albicans (2000). The R467K amino acid substitution in
clinical isolates contribute to resistance to azole Candida albicans sterol 14a-demethylase causes
antifungal agents. Antimicrob. Agents Chemother. drug resistance through reduced affinity.
42, 241-253. Antimicrob. Agents Chemother 44, 63-67.
153. White, T.C. (1997). Increased mRNA levels of 163. Favre, B., M. Didmon, and N.S. Ryder (1999).
ERG 16, CDR and MDRl correlate with increases Multiple amino acid substitutions in lanosterol
in azole resistance in Candida albicans isolates 14 alpha-demethylase contribute to azole resist-
from an HIV-infected patient. Antimicrob, Agents ance in Candida albicans. Microbiology 145,
Chemother 41, 1482-1487. 2715-2725.
154. Franz, R., M. Ruhnke, D.C. Lamb, S.L. Kelly, and 164. Marichal, P., L. Koymans, S. Willemsens, D.
J. Morschhauser (1998). Multiple molecular mech- Bellens, R Verhasselt, W. Luyten et al. (1999).
anisms contribute to a stepwise development of Contribution of mutations to the cytochrome P450
fluconazole resistance in clinical Candida albicans 14 alpha-demethylase (Erglip, CYP51) to azole
strains. Antimicrob. Agents Chemother. 42, resistance in Candida albicans. Microbiology 145,
3065-3072. 2701-2713.
155. Hamamoto, H., K. Hasegawa, R. Nakaune, 165. Perea, S., J.L. Lopez-Ribot, WR. Kirkpatrick,
Y.J. Lee, Y. Makizumi, K. Akutsu et al. (2000). K. McAtee, R.A. Santillan, M. Martinez,
Tandem repeat of a transcriptional enhancer D. Calabrese etal. (2001). Prevalence of molecular
upstream of the sterol 14alpha-demethylase gene mechanisms of resistance to azole antifungal
(CYP51) in Penicillium digitatum Appl. Environ. agents in Candida albicans strains displaying
Microbiol. 66, 3421-3426. high-level fluconazole resistance isolated from
156. Mellado, E., T.M. Diaz-Guerra, M. Cuenca- human immunodeficiency virus infected patients.
Estrella, and J.L. Rodriguez-Tudela (2001). Antimicrob. Agents Chemother 45, 2676-2684.
Identification of two different 14a sterol demethy- 166. Boscott, RE. and G.H. Grant (1994). Modeling
lase-related genes (cyp51A and cyp51B) in cytochrome P450 14-alpha demethylase {Candida
Aspergillus fumigatus and other aspergillus albicans) from P450CAM. J. Mol. Graph. 12,
species. J. Clin. Microbiol. 39, 2431-2438. 185-192.
157. Denning, D.W., K. Venkateswarlu, K.L. Oakley, M.J. 167. Ji, H.T, W.N. Zhang, YJ. Zhou, YL. Song, J. Lu,
Anderson, N.J. Manning, D.A. Stevens et al. (1997). and J. Zhu (2000). A three-dimensional model of
Itraconazole resistance in Aspergillus fumigatus. lanosterol 14 alpha-demethylase of Candida
Antimicrob. Agents Chemother 41, 1364-1368. albicans and its interaction with azole antifungals.
158. Ishida, N., Y Aoyama, R. Hatanaka, Y. Oyama, J. Med Chem. 43, 2495-2505.
S. Imajo, M. Ishiguro et al. (1988). A single amino 168. Delye, C , F Laigret, and M.F. Corio-Costet
acid substitution converts cytochrome P450j4pj^: (1997). A mutation in the 14a-demethylase gene
Complete primary structures deduced from cloned of Uncinula necator that correlates with resistance
DNAs. Biochem. Biophys. Res. Commun. 155, to a sterol biosynthesis inhibitor. Appl. Environ.
317-323. Microbiol. 63, 2966-2970.
159. Vanden Bossche, H., P. Marichal, J. Gorens, 169. Cosme, J. and E.F. Johnson (2000). Engineering
D. Bellens, H. Moereels, and P. Janssen microsomal cytochrome P450 2C5 to be a
(1990). Mutation in cytochrome P450 dependent soluble, monomeric enzyme. J. Biol. Chem. 275,
14a-demethylase results in decreased affinity 2545-2553.
for azole antifungals. Biochem. Soc. Trans. 18, 170. Williams, RA., J. Cosme, V Sridhar, E.R Johnson
56-59. and D.E. McCree (2000). Mammalian microsomal
160. Lamb, D.C, D.E. Kelly, W.H. Schunck, A.Z. C3^ochrome P450 monooxygenase: Structural
Shyadehi, M. Akhtar, D.J. Lowe et al (1997). The adaptations for membrane binding and functional
mutation T315A in Candida albicans sterol 14a- diversity. Mol Cell 5, 121-131.
demethylase causes reduced enzyme activity and 171. Joo, H., Z.L. Lin, and EH. Arnold (1999).
fluconazole resistance through reduced affinity. Laboratory evolution of peroxide-mediated cyto-
J. Biol. Chem. 212, 5682-5688. chrome P450 hydroxylation. Nature 399, 670-673.
Appendix

Human and Rat Liver Cytochromes


P450: Functional Markers, Diagnostic
Inhibitor Probes, and Parameters
Frequently Used in P450 Studies
Maria Almira Correia

The tables in this appendix summarize the rel- different P450 isoforms included in its evaluation
ative functional selectivities of substrates and as well as the range of substrate/inhibitor concen-
inhibitors for the major human and rat liver trations tested. Second, substrates and inhibitors
C3^ochrome P450 isoforms (P450s). These hepatic determined to be "relatively selective" for a human
isoforms are well recognized to catalytically par- liver isoform, may not necessarily be so for its rat
ticipate in the metabolism of chemically diverse liver ortholog, and vice versa. Third, the relative
endo- and xenobiotics including drugs, and in the metabolic contribution of a P450 isoform to the
case of human liver P450s to thus contribute to in vivo hepatic metabolism of a given drug is directly
clinically adverse drug-drug interactions. Conse- proportional to the relative hepatic microsomal
quently, these P450s are the targets of intense abundance of that isoform and its affinity for that
scrutiny in the pharmaceutical screening of exist- compound, irrespective of its in vitro high meta-
ing or novel chemical agents of potential clinical bolic profile assessed under "optimized" condi-
relevance for drug development. At a more basic tions. This issue arises because recent advances in
level, these tables provide information on estab- recombinant P450 technology have made unprece-
lished and/or potential diagnostic tools for the dented amounts of purified human liver enzymes
identification and/or characterization of the meta- readily available for comparative in vitro charac-
bolic role of each individual P450 in the disposition terization of drug metabolism, at relative P450
of an as yet uncharacterized xeno- or endobiotic. concentrations that may be irrelevant in vivo.
Three critical issues are however worth considera- Furthermore, this abundance albeit highly desir-
tion before use of these experimental probes: First, able, has nevertheless also sidelined the compara-
as discussed in Chapter 7 (Inhibition of Cytochrome tive characterization of rat liver orthologs of newly
P450 Enzymes), given the vast diversity of P450 discovered human P450 isoforms. Thus, in some
isoforms and their differential active-site affinities cases (i.e., CYPIBI), better characterized func-
for a given compound, the "relative" selectivity tional probes are available for the human liver
of a substrate or inhibitor probe for a given P450 enzyme than for its rat counterpart. In addition to
isoform is entirely defined by the number of the functional probe information, a table with

Maria Almira Correia • Departments of Cellular and Molecular Pharmacology, Pharmaceutical Chemistry, and
Biopharmaceutical Sciences and the Liver Center, University of California, San Francisco, CA.
Cytochrome P450: Structure, Mechanism, and Biochemistry, 3e, edited by Paul R. Ortiz de Montellano
Kluwer Academic / Plenum Publishers, New York, 2005.

619
620 M.A. Correia

some signature P450 spectral characteristics and Biology (Volume 107, Cytochrome P450
parameters commonly used in P450 studies has Protocols)^, Chapter 7 (Inhibition of Cytochromes
been included as a quick reference guide. P450), and Chapter 10 (Human Cytochrome P450
It is to be noted that the tables in this appendix Ervz^mQs). Last, but not the least, I wish to partic-
have been compiled with practical utility rather ularly acknowledge Prof S. Rendic for having pro-
than comprehensiveness as the overall objective. vided access to his regularly tipdated human P450
Much more comprehensive coverage is available in metabolism database through the Gentest website
several excellent books and reviews on human and during the preparation of this Appendix. Wherever
rat liver P450s that are gratefully acknowledged as feasible, the literature citations in this appendix
the sources of some of the information presented have favored those providing methodological
herein^"^^. In particular, the reader is referred to details, assay modifications, and/or controversial
the series Methods in Enzymology (Volumes 10, information, sometimes at the expense of accurate
52, 206, 272, and 357)3-^, Methods in Molecular chronological reporting of the literature.
Human and Rat Liver Cytochromes P450 621

2 ^g 45
.. &
I *o
1^
2^ o
i
<u
^
OH t/) O
0? •C is o
S
Co f2iS c^ QQ u

u
^ 1O ^4D o
^ >

c
o
^
"O
fl
^^ 1
•S e
1
OH
1
^
C3

sS &>§>•§»&
o
o
1 "s H i l l , & I11
o cS
PQPQ
en en 3
§2 "b
2^ ii
u
PM
S"'5b
a 03
s+s
5^
^ o
3
III O
to il
c>
^u ^
.^
s '5
PH
o cN CO H H U H GO H H O^PLHC/3 O sS
*wa
A
% ^
ci
1
fl
o
s
O
iz;
T3 CD
fi fl ^3
P A
EaM ^
, a
W
b
O
I.S
^
o OH

2'^
K)
-^
OH
^ fl
9
PA
I
O

£2
SH

III
o
>- TJ
'2 w
o
a
a0
Q
I
< 2 ^^a .§
a
J) o

r r
13

^2 5 ^ .S q:: « 5 ^
gag ^
<D 2 0) O
•§lfe
PLH

<s vo vo 00 o^ ^H
^^ vo
^
1-H < n
rl
U
fS
U
fs u
fs u
fs
Q M
n <s
622 M.A. Correia

i cS oo w
• [X]
w
Q Q
' I

2 c
ts <u
^3 ^3 O
V5

I
o
^-v
^
±
to .
-^
.2
W

o
^ .2 o
o^ ts ^

4^
3

I -«
fl
U
O

•s g

11 i
Si

o 1 "
8 Is
It
_o o ^_^
CO >
I
I II
2^
til
£^1 PQ
(N
PH

00 o m ^

I/)
CM
in
<
m
r--
<U
m
TH

^
TT Si < PQ
iiij SlIJlH ^1lIlJI
Human and Rat Liver Cytochromes P450 623

»^ >^ o
ed o O
o

-S. o .

lis
c
T3

1^^

S s= 5
OH
•2II
•-^ O T3

<:|.^
^ J*- ^
o £.§

;^'5 2 o2
a.
•S B

D ON
, . > ^ «3 ex
I
is p
^ OTS ^ O o ^

^a

3 . CM .
-5 ^
W N
•-d
72 g o
• o ' ""^
O ?'^
(N O
3 ^13^
^—
,^ «2 "flj "o -O c3 ^ . 1 ^
O lo •-^
gjfl S.2 c ^ o^

2LVON
.1 ^^ g ^
. .^-'
3;-5^2::
o c I
JIJCN ^ rr- a — " — ' 03'—"—i"-
;i o o
O cp Os^ § - ^ ^ .
"S P^ OO - ^ ^ t^
r r ^ ^ eg c^ .g .£2 ^ ci
- i - " ^ ( - (<M>"-'
- H j <OJ fli
^'^ qi
^'^ nj
^^ fli
^^
i5i5§^| .^ u m ^^ p^ P^Cci
624 M.A. Correia

Table A.2. Human Liver P450s: Chemical Structures of Diagnostic


Substrate and Inhibitor Probes

P450S Substrate^ Inhibitor^

CYP1A2

OH O
Galangin (3, 5,7-trihydroxyflavone)^ ^^

CH3CH2'

Ethoxyresorufin^o^^ Furafylline (MBI/S)3^ ^2

^H—COCH3

CYP2A6 NH.

o^^o .HCl

Coumarin'^ Tranylcypromine'^' '*^^^

x)^
(R)-(+)Menthofuran (MBI/S)"^
Human and Rat Liver Cytochromes P450 625

Table A.2. (continued)

P450s Substrate^ Inhibitor^

CYP2B6

O' XH3
(S)-Mephenytoin^^ 2-PMADA^'^52

Bupropion hydrochloride^' 3-PIVIDIA^

9-Ethynylphenanthrene (MBI/S)^^^

/CI ,x^^\^Sv

' J.
Ticlopidine (MBI/S)'
CYP2C8
^^^ JD OH
CH
\==/ NH

Taxol (paclitaxel) ^^^^


CYP2C9

^"•-v/fv^^ O CI
Tienilicacid(MBI/Sp,73
Tolbutamide ^
626 M.A. Correia

Table A.2. (continued)

P450s Substrate^ Inhibitor^

COOH '^-vjhQ"
Diclofenac ^^ ^"^ Sulfaphenazole^

CYP2C18

3-[2, 3-Dichloro-4-(2-thenoyl)phenoxy]-
propan-1-or^

CYP2C19

(S)-Mephenytoin7i^4,8o^i (+)-A/-3-Benzyl-nirvanol'^'

(+)-A/-3-Benzyl-phenobarbital^

CYP2D6

4 > NH
. II
N—C—NH2
Debrlsoqulne^^^^ Quinidlne^^9,9o
Human and Rat Liver Cytochromes P450 627

Table A.2. (continued)

P450S Substrate" Inhibitor^

\>—CH—CH2—NH—C—CHg
CH3

(+/-) Bufuralor SCH66712^^96

c„.o^J>-o
CH3O -^^^
Dextromethorphan Hydrobromide^"^ ^^ Paroxetine (l\/IBI/Qlf

CYP2E1 H

Chlorzoxazone 97-99
1>CH3
4-l\/lethylpyrazole^^9^^^

(CH3)2N—N=0 (CH3CH2)2N-C-SH
yV-Nitrosodimethylamine^^^^°^ Diethyldithiocarbamate^^ ^^^ ^^^

CYP3A4

CH3

CH3

X
Testosterone^o^^^^ Azamulin"^
628 M.A. Correia

Table A.2. (continued)

P450s Substrate^ Inhibitor^

OH
CH3^.^-^^OCH3
I >CH3
H3C

CH3 ^ H3C' ^O
^ OH I/CH3

OHCH3
\^^^0C0CH3
6CH3
Erythromycin 116,117 ^ ^ 3
Troleandomycin (MBI/QO^i^ ^^^

H3CO2C

Nifedipine' Gestodene (MBI/S)

Ketoconazole'A 1 8 , 4 8 , 115, 122


Human and Rat Liver Cytochromes P450 629

Table A.2. (continued)

P450s Substrate^ Inhibitor^

f<::^=^^^^N-0CH3

V-OCOCHa

H3C CH3
Midazolam'' Diltiazam (MBI/QI)^ ^^e

CYP3A5

% ^
Midazolam^ 128,129,131

OCH3
Aflatoxin BV
630 M.A. Correia

Table A.2. (continued)

P450S Substrate^ Inhibitor^

CYP3A7

HO3SO
DHEA-Sulfate'

CYP4A11 COOH

COOH

Laurie acid ^ 17-0DYA(MBI/S)>

^ ^ N ^ ^ N ' ° "

HET0016^^^^^2

~ \ - COOH
\
H 10-IDA^^ 141
Human and Rat Liver Cytochromes P450 631

Table A.2. (continued)

P450S Substrate^ Inhibitor^

CYP4F2 OH

COOH

10-UDYA(MBI/Sr

CYP4F12 OOH

Arachidonic acid^"^^

CYP7A1

Cholesterol^

CYP8B1

3-One-4-ene-7a-hydroxycholesterol
632 M.A. Correia

Table A.2. (continued)


Additional literature references are listed in Table A.2.
"Arrow(s) indicate(s) the substrate position(s) oxidized by that particular P450 isoform, enabling the assay of the
corresponding oxidized metabolite(s) as its relatively selective functional probe(s).
*The arrow indicates the inhibitor site that is metabolically activated by that P450 isoform resulting in mechanism-
based inactivation (MBI) of the enzyme that is either irreversible (suicide, S) or quasi-irreversible (QI).
^Inhibitor acts competitively by coordinating to the P450 heme-iron and/or ligation to the protein at the active site.
^2-PMADA, 2-isopropenyl-2-methyladamantane.
^3-PMDIA, 3-isopropenyl-3-methyldiadamantane.
^SCH66712, 5-fluoro-2-[4-[(2-phenyl-1 H-imidazol-5-yl)methyl]-1 -piperazinyl]pyrimidine.
^A metabolic intermediate complex (MIC) observed only with CYP3A4 but not CYPs 3A5 and 3A7.
^Given their —89% sequence similarity, CYP3A4 and CYP3A5 have similar functional and inhibitory profiles.
However, CYP3A5 may be distinguished from CYP3A4 by its higher metabolic ratio of midazolam 1 '-/4-hydroxyla-
tion, aflatoxin Bl 8,9-epoxidation to 3a-hydroxylation, and alprazolam 4-/1'-hydroxylation, as well as by its inabil-
ity to form a diltiazam-MIC. Mifepristone has also been found to distinguish between the two CYP3A isoforms.
'DHEA-sulfate, dehydroepiandrosterone sulfate.
^17-ODYA, 17-octadecynoic acid. (Ortiz de Montellano, personal communication)
^ETOOl 6, A^-Hydroxy-A'^'(4-butyl-2-methylphenyl)-formamidine.
^10-IDA, 10-imidazolyldecanoic acid.
'"LTB4, leukotriene B4.
"10-UDYA, 10-undecynoic acid. (Ortiz de Montellano, personal communication)

Table A.3. Rat Liver P450s: Chemical Structures of Diagnostic


Substrate and Inhibitor Probes

P450s Substrate^ Inhibitor^

<^YP1A1 NH-COCH3 H0\

Acetanilide^^^ Rhapontigenin(MBI/S)^^^

(R)-Warfarin^5^6^^i

7-Ethoxyresorufin^ 40,41
(O-Deethylation)
Human and Rat Liver Cytochromes P450 633

Table A.3. (continued)

P450S Substrate^ Inhibitor^

CYP1A2 Caffeine^' ^^ Galangin (3,5,7-trihydroxyflavoneyf-e, 38

(N'Demethylation)

Theophylline^ Furafylline (MBI/S)^

\
CH3O

in40,4i
7-Methoxyresorufin

Phenacetin"^ ^^2
(O'Deethylation)

CYPIBI CH3OH

CH3O

-OCH3

"OCH3
TMS (2, 4, 3', 5'-tetramethoxystilbene)i^4

7-Ethoxyresorufin^ ^' i54^i56


(O-Deethylation)

CYP2A1
9^3 PH

Testosterone^
634 M.A. Correia

Table A.3. (continued)

P450S Substrate^ Inhibitor^

CYP2A2

Testosterone'^^ '^^

CYP2B1 9^3 OH

Testosterone'* Secobarbital (MBI/S)'^^ '^^

BK\ y/"CH2CH2—NH—C—CHCIF

O ^
A/-2-P-BPCFA (MBI/S)'68

\
CH3CH2CH2Cn2CH2
^^2 \ y-CH2CH2—NH—C—CHCIF
O "X
7-Pentoxyresoruf in' A/-2-P-NPCFA (IVIBI/S)'68

9-Ethynylphenanthrene (l\/IBI/Sy ,169.

CYP2C6
2ICH3 CH2
C—H

Progesterone 171,172 Pregn-4,20-diene-3-one (l\/IBI/S)'


Human and Rat Liver Cytochromes P450 635

Table A.3. (continued)

P450S Substrate^ Inhibitor^

(S)-Warfarin^5^6^5i Sulfaphenazole"^

CYP2C7 Testosterone^ 1^2, i63


(16a-Hydroxylation)

Progesterone^' ^^^
(16a-Hydroxylation)

CYP2C11
9"3 PH
COOH

Testosterone^^^' ^^^ Diclofenac (l\1BI/S)^

CYP2C12 CH3
^"^C-0

Progesterone^^^

OSO3H

HOaSO-^

5a-androstane-3a,17pdiol, 3,17-disulfate^^^

CYP2C13 Testosterone"^ ^^°^^^


(6p'Hydroxylatlon)
636 M.A. Correia

Table A.3. (continued)

P450S Substrate^ Inhibitor^

CYP2D2 CH3
NHCH3
NHCO—CH^-N(CH2CH3)2

4-Allyloxymethamphetamine(MBI/S)^^^

Debrisoquine
(4-Hydroxylationy ^^^

(+/-) Bufuralol
(T-Hydroxylationy ^^9, iso

CYP2E1
^H=CH
CI
Chlorzoxazone"^ ^^^ ^^2 trans'^ ,2-dichloroethylene (MBl/S)^^^
(6-Hydroxylation)

/V-Nltrosodlmethylamine^ 100, i84, i89 DIallylsulfone (MBI/S)^ 186-189

(N-Demethylation)

CYP3A2/3A23 CH3

Testosterone^' i^^, i63,190 ep-Thiotestosterone (MBI/S)^-


Human and Rat Liver Cytochromes P450 637

Table A.3. (continued)

P450s Substrate^ Inhibitor^

Erythromycin^' ^^^ 2p-Thiotestosterone (MBI/S) 191


(N-Demethylation)
Nifedipines^ ^2 Troleandomycin {MB\/Q\y
(Oxidation)

CYP3A9
2rcH3

Progesterone^^^, 195
(6p, 21-Dihydroxylation)

CYP4A1 Laurie acid^ ^^^ 17-ODYA"'(MBI/S)


^(o -Hydroxylation)

CYP7A1 Cholesterol^ 199,200 10-UDYA"'(MBI/S),c, 197, 198

(7oL-Hydroxylation)

^The arrow(s) indicate(s) the substrate position(s) oxidized by that particular P450 isoform, enabUng the assay of the
corresponding oxidized metaboHte(s) as its relatively selective functional probe(s).
*The arrow indicates the inhibitor site that is metabolically activated by that P450 isoform resulting in mechanism-
based inactivation (MBI) of the enzyme that is either irreversible (suicide, S) or quasi-irreversible (QI).
'^The chemical structures is depicted in Table A.2. The reaction used as a functional probe is shown in parentheses.
^A polyclonal anti-rat CYPl Al antibody is available that recognizes CYPl A2 in immunoblotting, but that selectively
immunoinhibits CYPlAl but not CYP1A2 function.
^Inhibitor acts competitively by coordinating to the P450 heme-iron and/or protein binding at the active site.
^Steroid structure and site of 16a-hydroxylation shown under CYP2C11. Lower activity than that of CYP2C11.
^Phenethylisocyanate is also reported to selectively inhibit this enzyme^ ^9.
^Progesterone or androstenedione 6p-hydroxylase may also be used as functional probe^^^, ni
'17-ODYA, 17-octadecynoic acid. (Ortiz de Montellano, PR., unpublished observations).
^10-UDYA, 10-undecynoic acid, (Ortiz de Montellano, PR., unpublished observations).
638 M.A. Correia

Table A.4. Commonly Used P450 Substrates and Inhibitors


Trivial name Chemical structure Type of oxidation P450 catalyst(s)

A. Substrates
Acetaminophen!^^ 200,201 iV-Oxidation CYPs 2E1,3A,
1A2
NH—COCH3

OH

Aminopyrine^^!' ^^^ A^-Demethylation Multiple


CHa JS1/CH3
CH3

CHa

Androstenedione^^^, i63 C-Hydroxylation Multiple

Aniline! C-Hydroxylation Multiple

Benzphetamine^^! 203 /=^=\ CH3 / = \ iV-Demethylation Multiple

\j-'ytt^
Human and Rat Liver Cytochromes P450 639

Table A.4. (continued)

Trivial name Chemical structure Type of oxidation P450 catalyst(s)

Benzo(a)pyrene^^^' ^^"^ C-Hydroxylation CYPs lA, IB

7-Ethoxycoumarin i ^ i, 205 ^ O-Deethylation Multiple

CH3CH20.,^^;s>ss^O\^0

^^^^^^^

7-Ethoxy-4- O-Deethylation Multiple


trifluoromethyl-
26,53 CH3CH2'
coumarm

Ethylmorphine ^ iV-Demethylation Multiple


CH3

CH3CH2O O OH

/j-Chloro-A'-methylaniline iV-Demethylation CYPs lA


(PCNMA)206 HCH3

CI
640 M.A. Correia

Table A.4. (continued)

Trivial name Chemical structure lype of oxidation P450 catalyst(s)

/;-Nitroanisole^^^ O-Demethylation Multiple


CH3

NO2

Progesterone^ ^^ C-Hydroxylation Multiple


2rCH3
^ c=o

Testosteronei60-i63, i90 C-Hydroxylation Multiple


'3 OH

(i?/5)-Warfarin^ C-Hydroxylation Multiple

/ X
Human and Rat Liver Cytochromes P450 641

Table A.4. (continued)

Trivial name Chemical structure Modeof inactivation P450s inactivated

B. Inhibitors
Allylisopropylacetamide^^^ MBI/S Multiple
H2
/

1-ABTb 207-209 NH2 MBI/S Multiple


N
>
N

7,8-Benzoflavone^ CYPs lA, IB

Carbon monoxide^' ^ C=0 Multiple

Cimetidine^,210-213 H H C Multiple
N—TT-CHz-S-CHa-CHg-N-C-N-CHa MBI/QI CYPs 2C6 and
/ \ 2C11
NC=N

DDEP^ HV/CH2CH3 MBI/S Multiple


CH3CH202CN,^^^\S-^C02CH2CH3

H3U l^ CH3
H

Ellipticine^ CYPs lA, IB,


2A6
642 M.A. Correia

Table A.4. (continued)

Trivial name Chemical structure Mode of inactivation P450s inactivated

Fluvoxamine^^^'^^^ CYPs 1A2,


2C19,2C9

F3C-/ V9(CH2)40CH3
N—O—CH2CH2NH2

Ketoconazole^ Multiple^

I—COCH3

"6°
8-Metlioxypsoralen^ MBI/S Multiple^

Metyrapone^^^' ^^^ Multiple

N =

V_/S-EO
Piperonyl butoxide^^^' ^^"^ MBI/QI Multiple
Human and Rat Liver Cytochromes P450 643

Table A.4. (continued)

Trivial name Chemical structure Mode of inactivation P450s inactivated

SKF525A^225 .^^^^ MBI/S Multiple

CH3CH2CH2-C-C-0-CH2-CH2-N(C2H5)2HCI

"C, competitive inhibition through P450 Fe'''VFe"'"^-heme complexation; MBI, Mechanism-based inactivation that is
either quasi-irreversible (QI) via metabohc intermediate complexation or irreversible (suicidal, S).
* 1 - Aminobenzotriazole
^DDEP, 3,5-Dicarbethoxy-2,6-dimethyl-4-ethyl-1,4-dihydropyridine.
^Potent inhibitor of CYP3A4, but can inhibit other P450s at higher concentrations. Nonselective for rat liver P450s.
^Potent inhibitor of CYP2A6, but can inactivate other P450s at higher concentrations.
/SKF525A, Proadifen hydrochloride
644 M.A. Correia

11 <u.
i "« O ^W)
o .S ^ 'A
^ JD O
ffi
CU
^^ P Of) O
<D
^
o xn
^ . 2 (u eg ^ ^•
"^ • SP^
PH
111 <D "^
45 «
s ^ . So
^1
-1
£ " ^ T3 . ^
PH <L)
.^
e« g 73
O I i! "T ^ U 0)
O B
>^ ^ S I ^H T3 >—r
U <L> O c/5 U
^ I 5 <N
5l
<^ o ^'^ ^ <N O OS ^ o
00 l O ^ < X <^ <D »r^
s a> rj- OH ^ 5S ^
•^;?
s > ^ O »^
CQ L_J 4_>
s irt
p .

<N 0 0
^ 1 vo ^ 0>

0^ o ^ r>4 ) rvj '-H m ^^


o
'—1 o ^ ( ' ^ OS ( N OO Tf ^ <N
^^

O o oOS
E OS
OS

1 c/3 CO. d c/5


r- ^ -^ ' ^ T f »r> CN OS »^
^ CO r ^ <N u ^ OS u-^ "1- ^^'/^
"^ u-^ u-^ Tt lo m
1^^ 1 rsi o Ti- OS o <N o lo vo vo o^rsi
^ m ^ o Tf OS Tt r t T T CO CO » 0
s Tf u-i »r> Tt «ri CO u^ VO rj- l o Tf Tt

< t:^ 9^
<u
•\ <i^
a sS zU S
^ 5^ J>
O ,i>
< 'o O
.(0 t/5
U ^ - ^ T2
? S ? S ? 5 + < + <^Q
fs" .1 w
•«
«
V.
•««*

IN
s:
4>
Q
t> ;j
sA
k^ ^
CQ
PLH
«
>^
0^
Human and Rat Liver Cytochromes P450 645

•e*
o a>
ys >^

o,

x^
t3<: o -g o
o a
CO
U

&~ -SB 5SJ P(^ PilH

<N O '^t
CO fS ro

O o o o U^ '—1 O
ON ON OS OS r- Tt o
Tf to u^ ^
^ ^

lyj GO. d c/3 oQ. d


o O OS ^H 00 iTi
CO «o <N en vo *r> *o »o
I m »o t o
t o u-^ Ti" i n »o

§^
IS & S
+
^
0)
(D
J
CD CO

6
+
1 ;^ pL, < ;^

I
I
^
s i
I
I
S I I
646 M.A. Correia

— w
OH

a
(U Ok
&
o .c
4> (Z>
-w B
SS fi CS "oS 'B c«
is "B M o
43 CO
<u (L> CA (U (U
OH (X ,fi J3 "^o ex
s _^
(73
on 1/3

o CA B a. o
(N JP ^e3 "o K 13
S "£1
^
TS
X>
1
g
o Di^ PH
•g 1o
fe
O
X
OQ 'S
<
U
g
O

s
<

s
3

r- ro O u-^ Tf vo ^ yn o
»n ^^ m vo <N rN «o 00 <N
yn -rf in IT) 'Tt *r) *r) CO Tf

I
^ +
I
to I «
(U a> (u
5+ :
§ I
•I
i .5
s
2 I I
Human and Rat Liver Cytochromes P450 647

1. Estimation of Cytochrome 2. Sato, R., T. Omura, Y Imai, and Y. Fujii-Kuriyama


(eds) (1987). Cytochrome P450: New trends.
P420 in Cytochrome P450 Yamada Science Foundation, Japan.
Preparations 3. Estabrook, R.W. and M.E. Pullman (eds) (1967).
Methods in Enzymology Volume 10: Oxidation and
Using the spectral baseline correction mode, Phosphorylation. Academic Press, New York, NY.
determine the peak spectral absorption differ- 4. Fleischer, S. and L. Packer (eds) (1978). Methods in
ences (AA) between 420-490 nm (P420) and Enzymology, Volume 52: Biological Oxidations,
450-490 nm (P450) of CO-reduced-reduced Microsomal cytochrome P450, and other hemopro-
tein systems. Academic Press, New York, NY.
(Fe+2C0-Fe+2) samples226,227,233,245
5. Johnson, E.F. and M.R. Waterman (eds) (1991).
[P450]: AA45(^9o^/0.091=X nmol/ml (with Methods in Enzymology, Volume 206: Cytochrome
P450, Part A. Academic Press, New York, NY.
emM'^cm"^ = 91)
6. Johnson, E.R and M.R. Waterman (eds) (1996).
[P420]: AA42o^9o„^ ^^-^^^ = ^ nmol/ml (with Methods in Enzymology, Volume 272: Cytochrome
emM-^cm-i = 110) P450, Part B. Academic Press, New York, NY.
(~0.041)X nmol/ml = correction for negative 7. Johnson, E.R and M.R. Waterman (eds) (2002).
Methods in Enzymology, Volume 357: Cytochrome
contribution of P450 to P420 peak absorption at
P450, Part C. Academic Press, New York, NY.
-420-424 nm(emM-icm-i = - 4 1 )
8. Phillips, I.R. and E.A. Shephard (eds) (1998).
Actual [P420] in P450 preparations: Methods in Molecular Biology, Volume 107:
7-[(-0.041X)/0.110] = Z nmol/ml Cytochrome P450 Protocols.
9. Testa, B. and R Jenner (eds) (1976). Drug
Metabolism: Chemical and Biological Aspects.
2. Determination of Cytochrome Marcel Dekker.
P450 Reductase Activity 10. Ortiz de Montellano, PR. (ed.) (1986). Cytochrome
P450: Structure, Mechanism and Biochemistry.
Carried out with NADPH as the electron donor Plenum Press, New York, NY.
and cytochrome c as the artificial electron 11. Guengerich, F.P. (ed.) (1987). Mammalian
acceptor by the method of Philips & Langdon^"*^ Cytochromes P450, Vols 1 & 2, CRC Press, Boca
as detailed245-247 Raton, FL.
12. Schenkman, J.B. and D. Kupfer (eds) (1982).
Using the AA^^Q^Jmin and an EmM~^cm~^ Hepatic Cytochrome P450 Monooxygenase System.
= 21 determine [AA55Qjjjjj/min]/0.021 = X nmol Pergamon Press, Elmsford, NY.
cytochrome c reduced/min^"^^"^"^^. 13. Ortiz de Montellano, PR. (ed.) (1995). Cytochrome
Purified liver P450 reductase preparations P450: Structure, Mechanism and Biochemistry.
range from 40-70 ixmol c3^oclirome c reduced/ Plenum Press, New York, NY.
mg protein/min. Molar concentrations of purified 14. Waxman, D.J. (1986). Rat hepatic cytochrome
P450 reductase are estimated using either a P-450: Comparative study of multiple isozymic
molecular weight of 74kDa (based on cDNA forms. In PR. Ortiz de Montellano (ed.). Cytochrome
P-450. Structure, Mechanism, and Biochemistry.
sequence as well as flavin and protein content)
Plenum Press, New York, NY, pp. 525-538.
or an apparent molecular weight of 79.5 kDa
15. Gonzalez, F.J. (1989). The molecular biology of
(SDS-PAGE).
cytochrome P450s. Pharmacol. Rev. 40, 243-288.
16. Guengerich, F.P. (2002). Update information on
human P450s. Drug Metab. Rev. 34, 7-15.
Aclcnowledgments
17. Wrighton, S.A., M. VandenBranden, J.C. Stevens,
L.A. Shipley, and B.J. Ring (1993). In vitro methods
Supported by NIH grants DK26506 and for assessing human hepatic drug metabolism: Their
GM44037. use in drug development. Drug Metab. Rev. 25,
453^84.
18. Newton, D.J., R.W. Wang, and A.Y Lu (1995).
References Cytochrome P450 inhibitors. Evaluation of speci-
ficities in the in vitro metabolism of therapeutic
1. Sato, R. and T. Omura (eds) (1978). Cytochrome agents by human liver microsomes, Drug Metab.
P450. Academic Press, New York, NY. Dispos. 23, 154-158.
648 M.A. Correia

19. Nelson, D.R., L. Koymans, T. Kamataki, probes for human cytochromes P450 lAl and 1A2.
J.J. Stegeman, R. Feyereisen, D.J. Waxman et al. J. Pharmacol. Exp. Ther 265, 401-407.
(1996). P450 superfamily: Update on new 31. Sesardic, D, A. Boobis, B. Murray, S. Murray,
sequences, gene mapping, accession numbers and J. Segura, R. De La Torre et al. (1990). Furafylline is
nomenclature. Pharmacogenetics 6, \-A2. a potent and selective inhibitor of cytochrome P450
20. Guengerich, F.R (1995). Human cytochrome P450 1A2 in man. Br J. Clin. Pharmacol. 29, 651-663.
enzymes. In RR. Ortiz de Montellano (ed.), 32. Kunze, K.L. and W.F Trager (1993). Isoform-
Cytochrome P450: Structure, Mechanism and selective mechanism-based inhibition of human
Biochemistry. Plenum Press, New York, NY, cytochrome P450 1A2 by fiirafylline. Chem. Res.
pp. 473-574. Toxicol. 6, 649-656.
21. Guengerich, F.R (2004). Human cytochrome P450 33. Clarke, S.E., A.D. Ayrton, and R.J. Chenery (1994).
enzymes. In, PR. Ortiz de Montellano (ed.). Cyto- Characterization of the inhibition of P4501A2 by
chrome P450: Structure, Mechanism and Bio- furafylline. Xenobiotica 24, 517-526.
chemistry. Plenum Press, New York, NY, pp. 370-530. 34. Tjia, J.R, J. Colbert, and D.J. Back (1996).
22. Rendic, S. and RJ. Di Carlo (1997). Human Theophylline metabolism in human liver micro-
C5^ochrome P450 enzymes: A status report summa- somes: Inhibition studies. J. Pharmacol. Exp. Ther
rizing their reactions, substrates, inducers, and 276,912-917.
inhibitors. Drug Metab. Rev. 29, 413-580. 35. Butler, M.N., M. Iwasaki, F.R Guengerich, and
23. Rendic, S. (2002). Summary of information on F.K. Kadlubar (1989). Human cytochrome
human CYP enzymes: Human P450 metabolism P-450PIA (P450IA2), the phenacetin 0-deethylase,
data. Drug Metab. Rev. 34, 83-448. is primarily responsible for the hepatic 3-
24. Kobayashi, K., K. Urashima, N. Shimada, and demethylation of caffeine and N-oxidation of car-
K. Chiba (2003). Selectivities of human cytochrome cinogenic arylamines, Proc. Natl. Acad. Sci. USA,
P450 inhibitors toward rat P450 isoforms: Study 86, 7696-7700.
with cDNA-expressed systems of the rat. Drug 36. Yun, C.H., G.R Miller, and FP Guengerich (2000).
Metab. Dispos. 31, 833-836. Rate-determining steps in phenacetin oxidations by
25. Kobayashi, K., K. Urashima, N. Shimada, and human cytochrome P450 1A2 and selected
K. Chiba (2002). Substrate specificity for rat mutants. Biochemistry 39, 11319-11329.
cytochrome P450 (CYP) isoforms: Screening with 37. Yang, TJ., Y Sai, K.W. Krausz, FJ. Gonzalez, and
cDNA-expressed systems of the rat. Biochem. H.V Gelboin (1998). Inhibitory monoclonal
Pharmacol 63, 889-896. antibodies to human cytochrome P450 1A2:
26. Yuan, R., S. Madani, X.X. Wei, K. Reynolds, and Analysis of phenacetin 0-deethylation in human
S.M. Huang (2002). Evaluation of cytochrome P450 liver. Pharmacogenetics 8, 375-382.
probe substrates commonly used by the 38. Zhai, S., R. Dai, FK. Friedman, and R.E. Vestal
pharmaceutical industry to study in vitro drug inter- (1998). Comparative inhibition of human
actions. Drug Metab. Dispos. 30, 1311-1319. cytochrome P450 lAl and 1A2 by flavonoids.
27. Eagling, VA., J.R Tjia, and D.J. Back (1998). Drug Metab. Dispos. 26, 989-992.
Differential selectivity of cytochrome P450 inhi- 39. Lee, H., H. Yeom, YG. Kim, C.N. Yoon, C. Jin,
bitors against probe substrates in human and rat J.S. Choi et al. (1998). Structure-related inhibition
liver microsomes. Br J. Clin. Pharmacol. 45, of human hepatic caffeine N3-demethylation by
107-114. naturally occurring flavonoids. Biochem. Pharmacol.
28. Fuhr, U, J. Doehmer, N. Battula, C. Wolfel, 55, 1369-1375.
C. Kudla, Y. Keita et al. (1992). Biotransformation 40. Burke, M.D., S. Thompson, R.J. Weaver, C.R. Wolf,
of caffeine and theophylline in mammalian cell and R.T. Mayer (1994). Cytochrome P450 speci-
lines genetically engineered for expression of single ficities of alkoxyresorufin 0-dealkylation in human
cytochrome P450 isoforms. Biochem. Pharmacol. and rat liver. Biochem. Pharmacol. 48, 923-936.
43, 225-235. 41. Rodrigues, A.D. and R.A. Prough (1992). Induction
29. Tassaneeyakul, W., Z. Mohamed, D.J. Birkett, M.E. of cytochromes P450IA1 and P450IA2 and
McManus, M.E. Verones, R.H. Tukey et al. (1992). measurement of catalytic activities. Meth. Enzymol.
Caffeine as a probe for human cytochromes P450: 206,423^31.
Validation using cDNA expression, immuno- 43. Yun, C.H., T Shimada, and FR Guengerich (1991).
inhibition and microsomal kinetic and inhibitor Purification and characterization of human liver
techniques. Pharmacogenetics 2, 173-183. microsomal cytochrome P-450 2A6. Mol.
30. Tassaneeyakul, W., D.J. Birkett, M.E. Veronese, Pharmacol. 40, 679-685.
M.E. McManus, R.H. Tukey, L.C. Quattrochi et al. 44. Yamano, S., J. Tatsuno, and FJ. Gonzalez (1990).
(1993). Specificity of substrate and inhibitor The CYP2A3 gene product catalyzes coumarin
Human and Rat Liver Cytochromes P450 649

7-hydroxylation in human liver microsomes. marker of human cytochrome P450 2B6 catalytic
Biochemistry 19, 1322-1329. activity. Drug Metab. Dispos. 28, 1222-1230.
45. Sai, Y., T.J. Yang, K.W. Krausz, F.J. Gonzalez, and 57. Richter, T, K. Klein, T.E. Murdter, M. Eichelbaum,
H.V Gelboin (1999). An inhibitory monoclonal M. Schwab, and U.M. Zanger (2003). Clopidogrel
antibody to human cytochrome P450 2A6 defines and ticlopidine are specific mechanism-based
its role in the metabolism of coumarin, 7- inhibitors of human cytochrome P450 2B6. In
ethoxycoumarin and 4-nitroanisole in human liver. Proceedings of the 13th International Conference
Pharmacogenetics 9, 229-237. on Cytochromes P450. Prague, Czech Republic,
46. Zhang, W., T. Kilicarslan, R.F. Tyndale, and p. S119.
E.M. Sellers (2001). Evaluation of methoxsalen, 58. Dai, D., D.C. Zeldin, J.A. Blaisdell, B. Chanas,
tranylcypromine, and trj^tamine as specific and S.J. Coulter, B.I. Ghanayem et al. (2001). Poly-
selective CYP2A6 inhibitors in vitro. Drug Metab. morphisms in human CYP2C8 decrease metabo-
Dispos. 29, 897-902 lism of the anticancer drug paclitaxel and arachidonic
47. Taavitsainen, P., R. Juvonen, and O. Pelkonen acid, Pharmacogenetics 11, 597-607.
(2001). In vitro inhibition of cytochrome P450 59. Harris, J.W, A. Rahman, B.R. Kim, F.P
enzymes in human liver microsomes by a potent Guengerich, and J. Collins (1994). Metabolism of
CYP2A6 inhibitor, trans-2-phenylcyclopropylamine taxol by human hepatic microsomes and human
(Tranylcypromine), and its nonamine analog, liver slices: Participation of cytochrome P450 3A4
cyclopropylbenzene. Drug Metab. Dispos. 29, and an unknown P450 enzyme. Cancer Res. 54,
217-222. 4026-4035.
48. Draper, A.J., A. Madan, and A. Parkinson (1997). 60. Rahman, A., K.R. Korzekwa, J. Grogan, F.J.
Inhibition of coumarin 7-hydroxylase activity in Gonzalez, and J.W. Harris (1994). Selective bio-
human liver microsomes. Arch. Biochem. Biophys. transformation of taxol to 6 alpha-hydroxytaxol by
341,47-61. human cytochrome P450 2C8. Cancer Res. 54,
49. Khojasteh-Bakht, S.C, L.L. Koenigs, R.M. Peter, 5543-5546.
W.F. Trager, and S.D. Nelson (1998). (R)-(+)- 61. Wen, X., J.S. Wang, J.T. Backman, J. Laitila, and
Menthofuran is a potent, mechanism-based inacti- P.J. Neuvonen (2002). Trimethoprim and sul-
vator of human liver cytochrome P450 2A6. Drug famethoxazole are selective inhibitors of CYP2C8
Metab. Dispos. 26, 701-704. and CYP2C9, respectively. Drug Metab. Dispos.
50. Koenigs, L.L. and W.F. Trager (1998). Mechanism- 30,631-635.
based inactivation of P450 2A6 by furano- 62. Wang, J.S. and C.L. DeVane (2003). Involvement of
coumarins. Biochemistry 37, 10047-10061. CYP3A4, CYP2C8, and CYP2D6 in the metabolism
51. Heyn, H., R.B. White, and J.C. Stevens (1996). of (R)- and (S)-methadone in vitro. Drug Metab.
Catalytic role of cytochrome P450 2B6 in Dispos. 31, 742-747.
N-demethylation of ^'-mephenytion. Drug Metab. 63. Tang, W, R.A. Stearns, R.W Wang, S.H. Chiu, and
Dispos. 24, 948-954. T.A. Baillie (1999). Roles of human cytochrome
52. Stiborova, M., L. Borek-Dohalska, P. Hodek, P450s 2C9 and 3A4 in the metabolic activation of
J. Mraz, and E. Frei (2002). New selective inhibitors diclofenac. Chem. Res. Toxicol. 12, 192-199.
of cytochromes P450 2B and their application to 64. Leemann, T., C. Transon, and P. Dayer (1993).
antimutagenesis of tamoxifen. Arch. Biochem. Cytochrome P450TB (CYP2C): A major monooxy-
Biophys. 403, 4 1 ^ 9 . genase catalyzing diclofenac 4'-hydroxylation in
53. Ekins, S., M. VandenBranden, B.J. Ring, and human liver. Life Sci. 52, 29-34.
S.A. Wrighton (1997). Examination of purported 65. Rettie, A.E., K.R. Korzekwa, K.L. Kunze,
probes of human CYP2B6. Pharmacogenetics 7, R.F. Lawrence, A.C. Eddy, T. Aoyama et al. (1992).
165-179. Hydroxylation of warfarin by human cDNA-
54. Ekins, S. and S.A. Wrighton (1999). The role of expressed cytochrome P-450: A role for P-4502C9
CYP2B6 in human xenobiotic metabolism. Drug in the etiology of (S)-warfarin-drug interactions.
Metab. Rev. 31, 719-754. Chem. Res. Toxicol. 5, 54-59.
55. Rae, J.M., N.V Soukhova, D.A. Flockhart, and 66. Giancarlo, G.M., K. Venkatakrishnan, B.W
Z. Desta (2002). Triethylenethiophosphoramide is a Granda, L.L. von Moltke, and D.J. Greenblatt
specific inhibitor of cytochrome P450 2B6: (2001). Relative contributions of CYP2C9 and
Implications for cyclophosphamide metabolism. 2C19 to phenytoin 4-hydroxylation in vitro:
Drug Metab. Dispos. 30, 525-530. Inhibition by sulfaphenazole, omeprazole, and
56. Faucette, S.R., R.L. Hawke, E.L. Lecluyse, ticlopidine. Eur J. Clin. Pharmacol. 57, 31-36.
S.S. Shord, B. Yan, R.M. Laethem et al. (2000). 67. Relling, M.V, T. Aoyama, F.J. Gonzalez, and
Validation of bupropion hydroxylation as a selective U.A. Meyer (1989). Tolbutamide and mephenytoin
650 M.A. Correia

hydroxylation by human cytochrome P450s in the ific substrate of CYP 2C18. Biochemistry 38,
CYP2C subfamily. J. Pharmacol. Exp. Then 252, 7828-7836.
442^47. 79. Ha-Duong, N.T., C. Marques-Soares, S. Dijols,
68. Brian, W.R., P.K. Srivastava, D.R. Umbenhauer, M.A. Sari, P.M. Dansette, and D. Mansuy
R.S. Lloyd, and F.P. Guengerich (1989). Expression Interaction of new sulfaphenazole derivatives with
of a human liver cytochrome P-450 protein with human liver cytochrome P450 2Cs: Structural
tolbutamide hydroxylase activity in Saccharomyces determinants required for selective recognition by
cerevisiae. Biochemistry 28, 4993-4999. CYP 2C9 and for inhibition of human CYP 2Cs.
69. Srivastava, PK., C.H. Yun, PH. Beaune, C. Ged, Arch. Biochem. Biophys. 394, 189-200.
and F.P. Guengerich (1991). Separation of human 80. Wrighton, S.A., J.C. Stevens, G.W. Becker, and
liver tolbutamine hydroxylase and f5)-mephenytoin M. VandenBranden (1993). Isolation and character-
4'-hydroxylase cytochrome P-450 enzymes. Mol. ization of human liver cytochrome P450 2C19:
Pharmacol. 40, 69-79. Correlation between 2C19 and S-mephenytoin 4'-
70. Miners, J.O. and D.J. Birkett (1996). Use of tolbu- hydroxylation. Arch. Biochem. Biophys. 306,
tamide as a substrate probe for human hepatic cyto- 240-245.
chrome P450 2C9. Meth. Enzymol. 212, 139-145. 81. Goldstein, J.A., M.B. Faletto, M. Romkes-Sparks,
71. Lasker, J.M., M.R. Wester, E. Aramsombatdee, and T. Sullivan, S. Kitareewan, J.L. Raucy etal. (1994).
J.L. Raucy (1998). Characterization of CYP2C19 Evidence that CYP2C19 is the major (S)-mepheny-
and CYP2C9 from human liver: Respective roles toin 4'-hydroxylase in humans. Biochemistry 33,
in microsomal tolbutamide, S-mephenytoin, and 1743-1752.
omeprazole hydroxylation. Arch. Biochem. Biophys. 82. Suzuki, H., M.B. Kneller, R.L. Haining, and
353, 16-24. WF Trager, A.E. Rettie (2002). (+)-N-3-Benzyl-
72. Lopez-Garcia, M.P., P.M. Dansette, and D. Mansuy nirvanol and (-)-N-3-benzyl-phenobarbital: New
(1993). Thiophene derivatives as new mechanism- potent and selective in vitro inhibitors of
based inhibitors of cytochromes P450: Inactivation CYP2C19. Drug Metab. Dispos. 30, 235-239.
of yeast-expressed human liver P450 2C9 by tie- 83. Walsky, R.L. and R.S. Obach (2003). Verification
nilic acid. Biochemistry 33, 166-175. of the selectivity of (+)N-3-benzylnirvanol as a
73. Lopez-Garcia, M.P., P.M. Dansette, P. Valadon, CYP2C19 inhibitor. Drug Metab. Dispos. 31, 343.
C. Amar, PH. Beaune, F.P Guengerich et al. (1993). 84. Ha-Duong, N.T., S. Dijols, A.C. Macherey, J.A.
Human liver P450s expressed in yeast as tools for Goldstein, PM. Dansette, and D. Mansuy (2001).
reactive metabolite formation studies: Oxidative Ticlopidine as a selective mechanism-based
activation of tienilic acid by P450 2C9 and P450 inhibitor of human cytochrome P450 2C19.
2C10. Eur J. Biochem. 213, 223-232. Biochemistry 4(i, 12112-12122.
74. Shimada, T., K.S. Misono, and F.P. Guengerich 85. Ha-Duong, N.T., S. Dijols, A.C. Macherey,
(1986). Human liver microsomal cytochrome PM. Dansette, and D. Mansuy (2001). Inhibition by
P-450 mephenytoin 4-hydroxylase, a prototype of ticlopidine and its derivatives of human liver
genetic polymorphism in oxidative drug metabo- cytochrome P450. Mechanism-based inactivation
lism. Purification and characterization of two of CYP 2C19 by ticlopidine. Adv. Exp. Med. Biol.
similar forms involved in the reaction. J. Biol. 500, 145-148.
Chem. 261,909-921. 86. Gonzalez, F.J., T. Matsunaga, K. Nagata, U.A.
75. Kaminsky, L.S. (1989). Warfarin as a probe of Meyer, D.W Nebert, J. Pastewka et al. (1987).
cytochromes P450 function. Drug Metab. Rev. 20, Debrisoquine 4-hydroxylase: Characterization of a
479-487. new P450 gene subfamily, regulation, chromoso-
76. Kaminsky, L.S., M.J. Fasco, and F.P. Guengerich mal mapping, and molecular analysis of the DA rat
(1981). Production and apphcation of antibodies to polymorphism. DNA 6, 149-161.
rat liver cytochrome P-450. Meth. Enzymol. 74, 87. Gut, J., T. Catin, P Dayer, T. Kronbach, U. Zanger,
262-272. and U.A. Meyer (1986). Debrisoquine/sparteine-
77. Lang, D. and R. Bocker (1995). Highly sensitive type polymorphism of drug oxidation. Purification
and specific high-performance liquid chromato- and characterization of two functionally different
graphic analysis of 7-hydroxywarfarin, a marker human liver cytochrome P-450 isozymes involved
for human cytochrome P-4502C9 activity. J. in impaired hydroxylation of the prototype
Chromatogr Biomed. Appl. 672, 305-309. substrate bufuralol. J. Biol. Chem. 261,
78. Minoletti, C., S. Dijols, RM. Dansette, and 11734-11743.
D. Mansuy (1999). Comparison of the substrate 88. Kronbach, T. (1991). Bufuralol, dextromethorphan,
specificities of human liver cytochrome P450s 2C9 and debrisoquine as prototype substrates for human
and 2C18: Application to the design of a spec- P450IID6. Meth. Enzymol. 206, 509-517.
Human and Rat Liver Cytochromes P450 651

89. Otton, S.V, T. Inaba, and W. Kalow (1984). of cytochrome P450IIE1. Meth. Enzymol. 206,
Competitive inhibition of sparteine oxidation in 595-603.
human Hver by beta-adrenoceptor antagonists and 101. Tassaneeyakul, W., M.E. Veronese, D.J. Birkett,
other cardiovascular drugs. Life Sci. 34, 73-80. EJ. Gonzalez, and J.O. Miners (1993). Validation
90. Bertelsen, K.M., K. Venkatakrishnan, L.L. Von of 4-nitrophenol as an in vitro substrate probe
Moltke, R.S. Obach, and D.J. Greenblatt (2003). for human liver CYP2E1 using cDNA expression
Apparent mechanism-based inhibition of human and microsomal kinetic techniques. Biochem.
CYP2D6 in vitro by paroxetine: Comparison with Pharmacol. 46, 1975-1981.
fluoxetine and quinidine. Drug Metab. Dispos. 31, 102. Guengerich, EP, D.H. Kim, and M. Iwasaki
289-293. (1991). Role of human cytochrome P-450 IIEl in
91. Bloomer, J.C, F.R. Woods, R.E. Haddock, the oxidation of many low molecular weight cancer
M.S. Lennard, and G.T. Tucker (1992). The role of suspects. Chem. Res. Toxicol. 4, 168-179.
cytochrome P4502D6 in the metabolism of parox- 103. Kharasch, E.D., D.C. Hankins, C. Jubert, K.E.
etine by human liver microsomes. Br. J. Clin. Thummel, and J.K. Taraday (1999). Lack of single-
Pharmacol. 33, 521-523. dose disulfiram effects on cytochrome P-450 2C9,
92. Sindrup, S.H., K. Brosen, and L.F. Gram (1992). 2C19, 2D6, and 3A4 activities: Evidence for speci-
Pharmacokinetics of the selective serotonin reup- ficity toward P-450 2E1. Drug Metab. Dispos. 11,
take inhibitor paroxetine: Nonlinearity and relation 717-723.
to the sparteine oxidation polymorphism. Clin. 104. Chang, TK.H., EJ. Gonzalez, and D.J. Waxman
Pharmacol. Ther 51, 288-295. (1994). Evaluation of triacetyloleandomycin, a-
93. Sindrup, S.H., K. Brosen, L.F. Gram, J. Hallas, naphthoflavone and diethyldithiocarbamate as
E. Skjelbo, A. Allen et ai (1992). The relationship selective chemical probes for inhibition of human
between paroxetine and the sparteine oxidation cytochromes P450. Arch. Biochem. Biophys. 311,
polymorphism. Clin. Pharmacol. Ther 51, 437^42.
278-287. 105. Wrighton, S.A., PE. Thomas, D.T. Molowa,
94. Dayer, P, T Leemann, and R. Striberni (1989). M. Haniu, J.E. Shively, S.L. Maines et al. (1986).
Dextromethorphan O-demethylation in liver Characterization of ethanol-inducible human liver
microsomes as a prototype reaction to monitor N-nitrosodimethylamine demethylase. Biochemistry
cytochrome P-450 dbl activity. Clin. Pharmacol. 25,6731-6735.
Ther. 45, 3 4 ^ 0 . 106. Yoo, J.S., EP Guengerich, and C.S. Yang (1988).
95. Rodrigues, A.D. (1996). Measurement of human Metabolism of N-nitrosodialkylamines by human
liver microsomal cytochrome P450 2D6 activity liver microsomes. Cancer Res. 48, 1499-1504.
using [O-methyl-^'^Cldextromethorphan as sub- 107. Yamazaki, H., Y Inui, C.H. Yun, M. Mimura,
strate. Meth. Enzymol 111, 186-195. EP. Guengerich, andT. Shimada (1992). Cytochrome
96. Palamanda, J.R., C.N. Casciano, L.A. Norton, P450 2E1 and 2A6 enzymes as major catalysts for
R.P Clement, L.V Favreau, C. Lin et ai (2001). metabolic activation of A^-nitrosodialkylamines and
Mechanism-based inactivation of CYP2D6 by tobacco-related nitrosamines in human liver micro-
5-fluoro-2-[4-[(2-phenyl-1 H-imidazol-5-yl) methyl]- somes. Carcinogenesis 13, 1789-1794.
l-piperazinyl]pyrimidine. Drug Metab. Dispos. 29, 108. Yamazaki, H., Y. Oda, Y. Funae, S. Imaoka, Y Inui,
863-867. EP Guengerich et al. (1992). Participation of rat
97. Yamazaki, H., Z. Guo, and EP Guengerich (1995). liver cytochrome P450 2E1 in the activation of
Selectivity of cytochrome P4502E1 in chlorzoxa- N-nitrosodimethylamine and N-nitrosodiethyl-
zone 6-hydroxylation. Drug Metab. Dispos. 23, amine to products genotoxic in an acetyltrans-
438^40. ferase-overexpressing Salmonella typhimurium
98. Peter, R., R. Bocker, PH. Beaune, M. Iwasaki, EP strain (NM2009). Carcinogenesis 13, 979-985.
Guengerich, and C.S. Yang (1990). Hydroxylation 109. Waxman, D.J., C. Attisano, EP Guengerich, and
of chlorzoxazone as a specific probe for human liver D.P. Lapenson (1988). Human liver microsomal
cytochrome P-450IIE1. Chem. Res. Toxicol. 3, 566- steroid metabolism: Identification of the major
573. Erratum in: Chem. Res. Toxicol. 4, 389, 1991. microsomal steroid hormone 6 beta-hydroxylase
99. Feierman, D.E. and A.L Cederbaum (1986). cytochrome P-450 enzyme. Arch. Biochem.
Inhibition of microsomal oxidation of ethanol by Biophys. 263, 424^36.
pyrazole and 4-methylpyrazole in vitro. Increased 110. Gillam, E.M., T Baba, B.R. Kim, S. Ohmori, and
effectiveness after induction by pyrazole and EP Guengerich (1993). Expression of modified
4-methylpyrazole. Biochem. J. 239, 671-677. human cytochrome P450 3A4 in Escherichia coli
100. Yang, C.S., C.J. Patten, H. Ishizaki, and J.S.H. Yoo and purification and reconstitution of the enzyme.
(1992). Induction, purification, and characterization Arch. Biochem. Biophys. 305, 123-131.
652 M.A. Correia

111. Guengerich, F.R (1990). Mechanism-based inacti- 122. Stresser, M.D., M.I. Broudy, S.S. Dehal,
vation of human liver cytochrome P-450 IIIA4 by C.J. Patten, and C.L. Crespi (2002). Azamulin is
gestodene. Chem. Res. Toxicol. 3, 363-371. superior to ketoconazole as a selective CYP3A
112. Guengerich, F.R, M.V Martin, RH. Beaune, inhibitor probe. Drug Metab. Rev. Abstracts from
R Kremers, T. Wolff, and D.J. Waxman (1986). the 11th North American ISSX Meeting, Orlando,
Characterization of rat and human liver microso- FL, USA, Oct. 27-Oct. 31, Vol. 34, Abs. 355,
mal cytochrome P-450 forms involved in p. 178.
nifedipine oxidation, a prototype for genetic poly- 123. Jang, G.R., S.A. Wrighton, and L.Z. Benet (1996).
morphism in oxidative drug metabolism. J. Biol. Identification of CYP3A4 as the principal enzyme
C/zem. 261, 5051-5060. catalyzing mifepristone (RU 486) oxidation in
113. Gonzalez, F.J., B.J. Schmid, M. Umeno, O.W. human liver microsomes. Biochem. Pharmacol.
Mcbride, J.R Hardwick, U.A. Meyer et al. (1988). 52,753-761.
Human P450RCN1: Sequence, chromosome local- 124. He, K., T.F Woolf, and RE Hollenberg (1999).
ization, and direct evidence through cDNA expres- Mechanism-based inactivation of cytochrome
sion that P450PCN1 is nifedipine oxidase. DMA 7, P-450-3A4 by mifepristone (RU486). J. Pharmacol.
79-86. Exp. Ther 288, 791-797.
114. Guengerich, F.R, W.R. Brian, M. Iwasaki, 125. Khan, K.K., Y.Q. He, M.A. Correia, and
M.A. Sari, C. Baarnhielm, and P. Bemtsson J.R. Halpert (2002). Differential oxidation of
(1991). Oxidation of dihydropyridine calcium mifepristone by cytochromes P450 3A4 and 3A5:
channel blockers and analogues by human liver Selective inactivation of P450 3A4. Drug Metab.
cytochrome P-450 IIIA4. J. Med. Chem. 34, Dispos. 30, 985-990.
1838-1844. 126. Jones, D.R., J.C. Gorski, M.A. Hamman, B.S.
115. Varhe, A., M.B. Klaus, T. Olkkola, and Mayhew, S. Rider, and S.D. Hall (1999). Diltiazem
P.J. Neuvonen (1994). Oral triazolam is potentially inhibition of cytochrome P-450 3A activity is due
hazardous to patients receiving systemic antimy- to metabolite intermediate complex formation. J.
cotics. Clin. Pharmacol. Ther. 56, 601-607. Pharmacol. Exp. Ther 290, 1116-1125.
116. Watkins, RB., S.A. Wrighton, R Maurel, 127. Wrighton, S.A., W.R. Brian, M.A. Sari,
E.G. Schuetz, G. Mendez-Picon, G.A. Parker et al. M. Iwasaki, F.R Guengerich, J.L. Raucy et al.
(1985). Identification of an inducible form of (1990). Studies on the expression and metaboHc
cytochrome P-450 in human liver. Proc. Natl. capabilities of human Hver cytochrome P450IIIA5
Acad. Sci. USA 82, 6310-6314. (HLp3). Mol. Pharmacol. 38, 207-213.
117. Watkins, RB., S.A. Murray, L.G. Winkelman, 128. Gillam, E.M., Z. Guo, YE Ueng, H. Yamazaki,
D.M. Heuman, S.A. Wrighton, and PS. Guzelian 1. Cock, RE. Reilly et al. (1995). Expression of
(1989). Erythromycin breath test as an assay of cytochrome P450 3A5 in Escherichia coli: Effects
glucocorticoid-inducible liver cytochromes P-450. of 5' modification, purification, spectral character-
J. Clin. Invest. 83, 688-697. ization, reconstitution conditions, and catalytic
118. Thummel, K.E., C.A. Lee, K.L. Kunze, activities. Arch. Biochem. Biophys. 317, 374-384.
S.D. Nelson, and J.T. Slattery (1993). Oxidation of Erratum in: Arch. Biochem. Biophys. 318,498,1995.
acetaminophen to N-acetyl-p-aminobenzoquinone 129. Gorski, J.C, S.D. Hall, D.R. Jones,
imine by human CYP3A4. Biochem. Pharmacol. M. VandenBranden, and S.A. Wrighton (1994).
45, 1563-1569. Regioselective biotransformation of midazolam by
119. Renaud, J.R, C. Cullin, D. Pompon, R Beaune, and members of the human cytochrome P450 3A
D. Mansuy (1990). Expression of human liver (CYP3A) subfamily. Biochem. Pharmacol. 47,
cytochrome P450 IIIA4 in yeast. A functional 1643-1653.
model for the hepatic enzyme. Eur J. Biochem. 130. Gorski, J.C, D.R. Jones, M.A. Hamman, S.A.
194:889-896. Wrighton, and S.D. Hall (1999). Biotrans-
120. Kronbach, T., D. Mathys, M. Umeno, formation of alprazolam by members of the human
F.J. Gonzalez, and U.A. Meyer (1989). Oxidation cytochrome P4503A subfamily. Xenobiotica 29,
of midazolam and triazolam by human liver cyto- 931-944.
chrome P450IIIA4. Mol. Pharmacol. 36, 89-96. 131. Williams, J.A., B.J. Ring, VE. Cantrell, D.R. Jones,
121. Thummel, K.E., D.D. Shen, T.D. PodoU, J. Eckstein, K. Ruterbories et al. (2002). Com-
K.L. Kunze, WE Trager, PS. Hartwell et al. parative metabolic capabilities of CYP3A4,
(1994). Use of midazolam as a human cytochrome CYP3A5, and CYP3A7. Drug Metab. Dispos. 30,
P450 3A probe: I. In vitro-in vivo correlations in 883-891.
liver transplant patients. J. Pharmacol. Exp. Ther 132. Parimoo, B., VM. Mishin, CM. Busch, and
111, 549-556. RE. Thomas (2003). Identification of epitopes on
Human and Rat Liver Cytochromes P450 653

cytochrome P450 3A4/5 recognized by mono- HETE synthesizing enzyme. Br J. Pharmacol.


clonal antibodies. Arch. Biochem. Biophys. 414, 133, 325-329.
244-254. 143. Powell, PK., I. Wolf, R. Jin, and J.M. Lasker
133. Kitada, M., T. Kamataki, K. Itahashi, T. Rikihisa, (1998). Metabolism of arachidonic acid to 20-
and Y. Kanakubo (1987). P-450 HFLa, a form of hydroxy-5,8,ll,14-eicosatetraenoic acid by P450
cytochrome P-450 purified from human fetal enzymes in human liver: Involvement of CYP4F2
livers, is the 16 alpha-hydroxylase of dehydro- and CYP4A11. J. Pharmacol. Exp. Ther 285,
epiandrosterone 3-sulfate. J. Biol. Chem. 161, 1327-1336.
13534-13537. 144. Jin, R., D.R. Koop, J.L. Raucy, and J.M. Asker
134. Nakamura, H., N. Torimoto, I. Ishii, N. Ariyoshi, (1998). Role of human CYP4F2 in hepatic catabo-
H. Nakasa, S. Ohmori et al (2003). CYP3A4 and lism of the proinflammatory agent leukotriene B4.
CYP3A7-mediated carbamazepine 10,11 -epoxida- Arch. Biochem. Biophys. 359, 89-98.
tion are activated by differential endogenous 145. Zhang, X., L. Chen, and J.P Hardwick (2000). Pro-
steroids. Drug Metab. Dispos. 31, 432-^38. moter activity and regulation of the CYP4F2
135. Gillam, E.M., R.M. Wunsch, YE Ueng, T. leukotrieneB(4) omega-hydroxylase gene by per-
Shimada, RE. Reilly, T. Kamataki et al. (1997). oxisomal proliferators and retinoic acid in HepG2
Expression of cytochrome P450 3A7 in cells. Arch. Biochem. Biophys. 378, 364-376.
Escherichia coli: Effects of 5' modification and 146. Shak, S. and I. Goldstein (1984). Omega-oxidation
catalytic characterization of recombinant enzyme is the major pathway for the catabolism of
expressed in bicistronic format with NADPH- leukotriene B4 in human polymorphonuclear
cytochrome P450 reductase. Arch. Biochem. iQukocytQS. J. Biol. Chem. 259, 10181-10187.
Biophys. 346, 81-90. 147. Bylund, I , M. Bylund, and E.H. Oliw (2001).
136. Kawashima, H., E. Kusunose, Y Kikuta, cDNA cloning and expression of CYP4F12, a
H. Kinoshita, S. Tanaka, S. Yamamoto et al. novel human cytochrome P450. Biochem. Biophys.
(1994). Purification and cDNA cloning of human Res. Commun. 280, 892-897.
liver CYP4A fatty acid co-hydroxylase. J. Biochem. 148. Maeda, Y.R., G. Eggertsen, B. Nyberg,
116,74-80. T. Setoguchi, K.I. Okuda, K. Einarsson et al.
137. Powell, PK., I. Wolf, and J.M. Lasker (1996). (1995). Immunochemical determination of human
Identification of CYP4A11 as the major lauric cholesterol 7 alpha-hydroxylase. Eur. J. Biochem.
acid omega-hydroxylase in human liver micro- IIH, 144-148.
somes. Arch. Biochem. Biophys. 335, 219-226. 149. Gafvels, M., M. Olin, B.P Chowdhary,
138. Clarke, S.E., S.J. Baldwin, J.C. Bloomer, T. Raudsepp, U. Andersson, B. Persson et al.
A.D. Ayrton, R.S. Sozio, and R.J. Chenery (1994). (1999). Structure and chromosomal assignment
Lauric acid as a model substrate for the simultaneous of the sterol 12alpha-hydroxylase gene (CYP8B1)
determination of cytochrome P450 2E1 and 4A in human and mouse: Eukaryotic cytochrome
in hepatic microsomes. Chem. Res. Toxicol. 7, P-450 gene devoid of introns. Genomics 56,
836-842. 184-196.
139. Amet, Y, E Berthou, S. Baird, Y Dreano, J.P Bail, 150. Chun, Y.J., S.Y Ryu, T.C. Jeong, and M.Y Kim
and J.E Menez (1995). Validation of the (omega-1)- (2001). Mechanism-based inhibition of human
hydroxylation of lauric acid as an in vitro sub- cytochrome P450 lAl by rhapontigenin. Drug.
strate probe for human liver CYP2E1. Biochem. Metab. Dispos. 29, 389-393.
Pharmacol. 50, 1775-1782. 151. Guengerich, RP, G.A. Dannan, S.T. Wright, M.V
140. Savas, U, M.H. Hsu, and E.E Johnson (2003). Martin, and L.S. Kaminsky (1982). Purification
Differential regulation of human CYP4A genes by and characterization of liver microsomal cyto-
peroxisome proliferators and dexamethasone. chromes P-450: Electrophoretic, spectral, catalytic,
Arch. Biochem. Biophys. 409, 212-220. and immunochemical properties and inducibility
141. Oglivie, B.W., S.M. Otradovec, B.L. Paris, of eight isozymes isolated from rats treated with
J.A. Scheinkoenig, P.W. Carrott, S.P. Loecker et al. phenobarbital or beta-naphthoflavone. Biochemistry
(2000). 10-(imidazolyl)-decanoic acid (10-IDA) is 21, 6019-6030.
a selective and potent inhibitor of CYP4A9/11. 152. Sesardic, D., R.J. Edwards, D.S. Davies, PE.
Drug. Metab. Rev. Abstracts from the 10th North Thomas, W Levin, and A.R. Boobis (1990). High
American ISSX Meeting, Indianapolis, IN, USA, affinity phenacetin O-deethylase is catalysed
Vol. 32 (Suppl. 2), Abs. 188, p. 230. specifically by cytochrome P450d (P450IA2) in the
142. Miyata, N., K. Taniguchi, T. Seki, T. Ishimoto, liver of the rat. Biochem. Pharmacol. 39, 489^98.
M. Sato-Watanabe, Y Yasuda et al. (2001). 153. Sarkar, M.A. and B.J. Jackson (1994).
HET0016, a potent and selective inhibitor of 20- Theophylline N-demethylations as probes for
654 M.A. Correia

P4501A1 and P4501A2. Drug Metab. Dispos. 22, 164. Smith, S.J., K.R. Korzekwa, T. Aoyama,
827-834. F.J. Gonzalez, J.F. Darbyshire, K. Sugiyama et al.
154. Chun, Y.J., S. Kim, D. Kim, S.K. Lee, and (1992). 12 alpha-hydroxytestosterone. A hitherto
F.P. Guengerich (2001). A new selective and potent unidentified testosterone metabolite produced by
inhibitor of human c3^ochrome P450 IBl and its cytochrome P-450 2A2. Drug Metab. Dispos. 20,
application to antimutagenesis. Cancer Res. 61, 566-571.
8164-8170. Erratum in: Cancer Res. 62, 1232, 165. Lunetta, J.M., K. Sugiyama, and M.A. Correia
2002. (1989). Secobarbital-mediated inactivation of rat
155. Shimada, T, H. Yamazaki, M. Foroozesh, liver cytochrome P-450b: A mechanistic reap-
N.E. Hopkins, W.L. Alworth, F.P. Guengerich praisal. Mol. Pharmacol. 35, 10-17.
(1998). Selectivity of polycyclic inhibitors for 166. He, K., A.M. Falick, B. Chen, F Nilsson, and M.A.
human cytochrome P450s lAl, 1A2, and IBl. Correia (1996). Identification of the heme adduct
Chem. Res. Toxicol. 11, 1048-1056. and an active site peptide modified during mecha-
156. Shimada, T., F. Tsumura, E.M. Gillam, nism-based inactivation of rat liver cytochrome
FP Guengerich, and K. Inoue (2000). Roles of P450 2B1 by secobarbital. Chem. Res. Toxicol. 9,
NADPH-P450 reductase in the 0-deethylation of 614-622.
7-ethoxycoumarin by recombinant human cyto- 167. Lubet, R.A., R.T. Mayer, J.W Cameron, R.W. Nims,
chrome P4501B1 variants in Escherichia coli. M.D. Burke, T Wolff e/ al (1985). Dealkylation of
Protein Expr Purif. 20, 73-80. pentoxyresorufin: A rapid and sensitive assay for
157. Badawi, A.F, E.L. Cavalieri, and E.G. Rogan measuring induction of cytochrome(s) P-450 by
(2000). Effect of chlorinated hydrocarbons on phenobarbital and other xenobiotics in the rat. Arch.
expression of cytochrome P450 lAl, 1A2 and IBl Biochem. Biophys. 238, 4 3 ^ 8 .
and 2- and 4-hydroxylation of 17beta-estradiol in 168. Halpert, J., J.Y. Jaw, C. Balfour, and
female Sprague-Dawley rats. Carcinogenesis 21, L.S. Kaminsky (1990). Selective inactivation by
1593-1599. chlorofluoroacetamides of the major phenobarbi-
158. Hayes, C.L., D.C. Spink, B.C. Spink, J.Q. Cao, N.J. tal-inducible form(s) of rat liver cytochrome
Walker, and T.R. Sutter (1996). 17 beta-estradiol P-450. Drug Metab Dispos. 18, 168-174.
hydroxylation catalyzed by human cytochrome 169. Hopkins, N.E., M.K. Foroozesh, and W.L. Alworth
P450 IBl. Proc. Natl. Acad. Sci. USA 93, (1992). Suicide inhibitors of cytochrome P450
9776-9781. lAl and P450 2B1. Biochem. Pharmacol. 44,
159. Levin, W, RE. Thomas, D.E. Ryan, and A.W. 787-796.
Wood (1987). Isozyme specificity of testosterone 170. Roberts, E.S., N.E. Hopkins, M. Foroozesh,
7a-hydroxylation in rat hepatic microsomes: Is W.L. Alworth, J.R. Halpert, and PF Hollenberg
cytochrome P-450a the sole catalyst? Arch. Biochem. et al. (1997). Inactivation of cytochrome P450s
Biophys. 258, 630-635. 2B1, 2B4, 2B6, and 2B11 by arylalkynes. Drug
160. Waxman, D.J., A. Ko, and C. Walsh (1983). Metab Dispos. 25, 1242-1248.
Regioselectivity and stereoselectivity of androgen 171. Swinney, D.C, D.E. Ryan, RE. Thomas, and
hydroxylations catalyzed by cytochrome P-450 W Levin (1987). Regioselective progesterone
isozymes purified from phenobarbital-induced rat hydroxylation catalyzed by eleven rat hepatic
liver. J. Biol. Chem. 258, 11937-11947. cytochrome P-450 isozymes. Biochemistry 26,
161. Waxman, D.J. (1988). Interactions of hepatic 7073-7083.
cytochromes P-450 with steroid hormones. 172. Halpert, J., J.Y. Jaw, and C. Balfour (1989).
Regioselectivity and stereoselectivity of steroid Specific inactivation by 17p-substituted steroids
metabolism and hormonal regulation of rat P-450 of rabbit and rat liver cytochromes P-450 res-
enzyme expression. Biochem. Pharmacol. 37, ponsible for progesterone 21-hydroxylation. Mol.
71-84. Pharmacol. 34, 148-156.
162. Wood, A.W, D.R. Ryan, RE. Thomas, and 173. Kobayashi, K., K. Urashima, N. Shimada, and
W Levin (1983). Regio- and stereoselective K. Chiba (2003). Selectivities of human cyto-
metabolism of two Cjp steroids by five highly puri- chrome P450 inhibitors toward rat P450 isoforms:
fied and reconstituted rat hepatic cytochrome Study with cDNA-expressed systems of the rat.
P450 isozymes. J. Biol. Chem. 258, 8839-8847. Drug Metab. Dispos. 31, 833-836.
163. Arlotto, M.P, J.M. Trant, and R.W Estabrook 174. Masubuchi, Y, A. Ose, and T. Horie (2001).
(1992). Measurement of steroid hydroxylation Mechanism-based inactivation of CYP2C11 by
reactions by high-performance liquid chromatog- diclofenac. Drug Metab. Dispos. 29, 1190-1195.
raphy as indicator of P450 identity and function. 175. Shen, S., S.J. Hargus, B.M. Martin, and L.R. Pohl
Meth. Enzymol 206, 454-462. (1997). Cytochrome P4502C11 is a target of
Human and Rat Liver Cytochromes P450 655

diclofenac covalent binding in rats. Chem. Res. cytochrome P-450 2E1 by diallyl sulfide and its
Toxicol. 10, 420-423. metabolites. Chem. Res. Toxicol. 4, 642-647.
176. MacGeoch, C , E.T. Morgan, J. Halpert, and 187. Jin, L. and TA. BaiUie (1997). Metabohsm of the
J.A. Gustafsson (1984). Purification, characteriza- chemoprotective agent diallyl sulfide to glutathione
tion, and pituitary regulation of the sex-specific conjugates in rats. Chem. Res. Toxicol 10, 318-327.
cytochrome P-450 15 beta-hydroxylase from liver 188. Premdas, PD., R.J. Bowers, and RG. Forkert
microsomes of untreated female rats. J. Biol. (2000). Inactivation of hepatic CYP2E1 by an
Chem. 259, 15433-15439. epoxide of diallyl sulfone. J. Pharmacol. Exp.
177. Wan, J., S. Imaoka, T. Chow, T. Hiroi, Y. Yabusaki, Ther 293, 1112-1120.
and Y. Funae (1997). Expression of four rat 189. Ishizaki, H., J.F Brady, S.M. Ning, and C.S. Yang
CYP2D isoforms in Saccharomyces cerevisiae and (1990). Effect of phenethyl isothiocyanate on micro-
their catalytic specificity. Arch. Biochem. Biophys. somal N-nitrosodimethylamine metabolism and other
348, 383-390. monooxygenase activities. Xenobiotica 20, 255-264.
178. Chow, T., S. Imaoka, T. Hiroi, and Y. Funae (1999). 190. Sonderfan, A.J., M.R Arlotto, D.R. Dutton,
Developmental changes in the catalytic activity S.K. McMillen, and A. Parkinson (1987).
and expression of CYP2D isoforms in the rat liver. Regulation of testosterone hydroxylation by rat
DrugMetab. Dispos. 11, 188-192. liver microsomal cytochrome P-450. Arch.
179. Lin, L.Y., M. Fujimoto, E.W. Distefano, Biochem. Biophys. 255, 27-41.
D.A. Schmitz, A. Jayasinghe, and A.K. Cho 191. Underwood, M.C, J.R. Cashman, and M.A.
(1996). Selective mechanism-based inactivation Correia (1992). Specifically designed thiosteroids
of rat CYP2D by 4-allyloxymethamphetamine. as active site-directed probes for fiinctional dissec-
J. Pharmacol. Exp. Then 111, 595-603. tion of cytochrome P-450 3A isozymes. Chem.
180. Larrey, D., L.M. Distlerath, G.A. Dannan, Res. Toxicol. 5, 42-53.
G.R. Wilkinson, and F.R Guengerich (1984). 192. Wrighton, S.A., R Maurel, E.G. Schuetz,
Purification and characterization of the rat liver RB. Watkins, B. Young, and RS. Guzehan (1985).
microsomal cytochrome P-450 involved in the Identification of the cytochrome P-450 induced by
4-hydroxylation of debrisoquine, a prototype for macrolide antibiotics in rat liver as the glucocorti-
genetic variation in oxidative drug metabolism. coid responsive cytochrome P-450p. Biochemistry
Biochemistry 23, 2787-2795. 24,2171-2178.
181. Tanaka, E. (2001). Chlorzoxazone: A probe drug the 193. Watkins, RB., S.A. Wrighton, E.G. Schuetz,
metabolism of which can be used to monitor R Maurel, and RS. Guzelian (1986). Macrolide
one-point blood sampling in the carbon tetrachloride- antibiotics inhibit the degradation of the glucocor-
intoxicated rat. Hum. Exp. Toxicol. 20, 381-385. ticoid-responsive cytochrome P-450p in rat hepa-
182. Mizuno, D., E. Tanaka, K. Tanno, and S. Misawa tocytes in vivo and in primary monolayer culture.
(2000). Chlorzoxazone: A probe drug whose J. Biol. Chem. 261, 6264-6271.
metabolism can be used to monitor toluene 194. Wang, H., K.L. NapoH, and H.W. Strobel (2000).
exposure in rats. Arch. Toxicol. 74, 139-144. Cytochrome P450 3A9 catalyzes the metabolism
183. Mathews, J.M., A.S. Etheridge, J.H. Raymer, S.R. of progesterone and other steroid hormones. Mol
Black, D.W Pulliam Jr., and J.R. Bucher (1998). Cell. Biochem. 213, 127-135.
Selective inhibition of cytochrome P450 2E1 in 195. Xue, L., V.G. Zgoda, B. Arison, and M.A. Correia
vivo and in vitro with trans-1,2-dichloroethylene. (2003). Rat liver CYP3A9: Structure-function rela-
Chem. Res. Toxicol. 11, 778-785. tionships to its human liver orthologs: Site-directed
184. Yoo, J.S., T.J. Smith, S.M. King, M.J. Lee, mutagenesis to an efficient progesterone dihydrox-
RE. Thomas, and C.S. Yang (1992). Modulation of ylase. Arch. Biochem. Biophys. 409, 113-126.
the levels of cytochromes P450 in rat liver and 196. Gibson, G.G., T.C. Orton, and RR Tamburini
lung by dietary lipid. Biochem. Pharmacol. 43, (1982). Cytochrome P-450 induction by clofibrate:
2535-2542. Purification and properties of a hepatic cyto-
185. Yamazaki, H., Y. Oda, Y Funae, S. Imaoka, Y Inui, chrome P-450 relatively specific for the 12- and
F.P. Guengerich et al. (1992). Participation of rat 11-hydroxylation of dodecanoic acid (lauric acid).
liver cytochrome P450 2E1 in the activation of Biochem. J. 203, 161-168.
N-nitrosodimethylamine and N-nitrosodiethyl- 197. CaJacob, C.A., WK. Chan, E. Shepard, and
amine to products genotoxic in an acetyltrans- RR. Ortiz de Montellano (1988). The catalytic site
ferase-overexpressing Salmonella typhimurium of rat hepatic lauric acid omega-hydroxylase.
strain (NM2009). Carcinogenesis 13, 979-985. Protein versus prosthetic heme alkylation in the
186. Brady, J.F., H. Ishizaki, J.M. Fukuto, M.C. Lin, omega-hydroxylation of acetylenic fatty acids. J.
A. Fadel, J.M. Gapac et al. (1991). Inhibition of Biol. Chem. 263, 18640-18649.
656 M.A. Correia

198. Ortiz de Montellano, P.R. and N.O. Reich (1984). in male Sprague-Dawley rats. Fundam. Appl.
Specific inactivation of hepatic fatty acid hydrox- Toxicol. 19, 43-49.
ylases by acetylenic fatty acids. J. Biol. Chem. 259, 210. Levine, M. and G.D. Bellward (1995). Effect of
4136-4141. cimetidine on hepatic cytochrome P450: Evidence
199. Chiang, J.Y. (1991). Reversed-phase high- for formation of a metabolite-intermediate com-
performance liquid chromatography assay of plex. DrugMetab. Dispos. 23, 1407-1411.
cholesterol 7 alpha-hydroxylase. Meth. Enzymol. 211. Levine, M., E.Y. Law, S.M. Bandiera, T.K. Chang,
206,483-491. and G.D. Bellward (1998). In vivo cimetidine
200. Raucy, J.L., J.M. Lasker, C.S. Lieber, and M. Black inhibits hepatic CYP2C6 and CYP2C11 but not
(1989). Acetaminophen activation by human liver CYPlAl in adult male rats. J. Pharmacol. Exp.
cytochromes P450nEl and P450IA2. Arch. Then 284, 493-499.
Biochem. Biophys. Ill, 270-283. 212. Gerber, M.C, G.A. Tejwani, N. Gerber, and J.R.
201. Sinclair, J., E. JefFery, S. Wrighton, V Kostrubsky, Bianchine (1985). Drug interactions with cimeti-
J. Szakacs, S. Wood, and R Sinclair (1998). dine: An update. Pharmacol. Ther 11, 353-370.
Alcohol-mediated increases in acetaminophen 213. Smith, S.R. and M.J. Kendall (1988). Ranitidine
hepatotoxicity: role of CYP2E and CYP3A. versus cimetidine. A comparison of their potential
Biochem. Pharmacol. 55, 1557-1565. to cause clinically important drug interactions.
202. Imaoka, S., K. Inoue, and Y. Funae (1988). Clin. Pharmacokinet. 15, 44-56.
Aminopyrine metabolism by multiple forms of 214. Correia, M.A., C. Decker, K. Sugiyama, P Caldera,
cytochrome P-450 from rat liver microsomes: L. Bornheim, S.A. Wrighton et al. (1987).
Simultaneous quantitation of four aminopyrine Degradation of rat hepatic cj^ochrome P-450
metabolites by high-performance liquid chromatog- heme by 3,5-dicarbethoxy-2,6-dimethyl-4-
raphy. Arch. Biochem. Biophys. 265, 159-170. ethyl-l,4-dihydropyridine to irreversibly bound
203. Bornheim, L.M., M.C. Underwood, P. Caldera, protein adducts. Arch. Biochem. Biophys. 258,
A.E. Rettie, W.E Trager, S.A. Wrighton et al. 436-451.
(1987). Inactivation of multiple hepatic cj^o- 215. Lesca, P., E. Rafidinarivo, P. LeCointe, and
chrome P-450 isozymes in rats by allyhsopropy- D. Mansuy (1979). A class of strong inhibitors of
lacetamide: Mechanistic implications. Mol. microsomal monooxygenases: The ellipticines.
Pharmacol. 32, 299-308. Chem. Biol. Interact. 24, 189-198.
204. Nebert, D.W (1982). Genetic differences in 216. Tassaneeyakul, W, D.J. Birkett, M.E. Veronese,
the induction of monooxygenase activities by M.E. McManus, R.H. Tukey, L.C. Quattrochi et al.
polycyclic aromatic compounds. In J.B. (1993). Specificity of substrate and inhibitor
Schenkman and D. Kupfer (eds). Hepatic probes for human cytochromes P450 lAl and
Cytochrome P-450 Monooxygenase System. 1A2. J. Pharmacol. Exp. Ther 265, 401^07.
Pergamon Press, New York, NY, pp. 269-291. 217. Hemeryck, A. and FM. Belpaire (2002). Selective
205. Prough, R.A., M.D. Burke, and R.T. Mayer (1978). serotonin reuptake inhibitors and cytochrome
Direct fluorometric methods for measuring mixed- P-450 mediated drug-drug interactions: An update.
fiinction oxidase activity. Meth. Enzymol. 52, Curr Drug. Metab. 3, 13-37.
372-377. 218. Richelson, E. (1997). Pharmacokinetic drug inter-
206. Correia, M.A. and G.J. Mannering (1973). Reduced actions of new antidepressants: A review of the
diphosphopyridine nucleotide synergism of the effects on the metabolism of other drugs. Mayo
reduced triphosphopyridine nucleotide dependent Clin. Proc. 72, 835-847.
mixed function oxidase of hepatic microsomes. 219. Letteron, P., V Descatoire, D. Larrey, M. Tinel,
Role of the Type I drug binding site of cytochrome J. Geneve, and D. Pessayre (1986). Inactivation and
P-450. Mol. Pharmacol. 9, 470-485. induction of cytochrome P-450 by various pso-
207. Ortiz de Montellano, PR. and A.K. Costa (1986). ralen derivatives in rats. J. Pharmacol. Exp. Ther
Dissociation of cytochrome P-450 inactivation 238, 685-692.
and induction. Arch. Biochem. Biophys. 251, 220. Koenigs, L.L. and WF Trager (1998). Mechanism-
514-524. based inactivation of cytochrome P450 2B1 by
208. Mico, B.A., D.A. Federowicz, M.G. Ripple, and 8-methoxypsoralen and several other furano-
W. Kerns (1988). In vivo inhibition of oxidative coumarins. Biochemistry 37, 13184-13193.
drug metabolism by, and acute toxicity of 221. Testa, B. and P Jenner (1981). Inhibitors of
1-aminobenzotriazole (ABT). Biochem. Pharmacol. cytochrome P-450s and their mechanism of action.
37,2515-2519. DrugMetab. Rev. 12, 1-117.
209. Mugford, C.A., M. Mortillo, B.A. Mico, and J.B. 222. Waxman, D.J. and C. Walsh (1983). Cytochrome
Tarloff (1992). 1-Aminobenzotriazole-induced P-450 isozyme 1 from phenobarbital-induced
destruction of hepatic and renal cytochromes P450 rat liver: Purification, characterization, and
Human and Rat Liver Cytochromes P450 657

interactions with metyrapone and cytochrome b5. 235. Philpot, R.M. and E. Hodgson (1972). The effect
Biochemistry 22, 4846-4855. of piperonyl butoxide concentration on the forma-
223. Franklin, M.R. (1972). Inhibition of hepatic oxida- tion of cytochrome P-450 difference spectra in
tive xenobiotic metabolism by piperonyl butoxide. hepatic microsomes from mice. Mol. Pharmacol.
Biochem. Pharmacol. 21, 3287-3299. 8, 204-214.
224. Hodgson, E. and R.M. Philpot (1974). Interaction 236. Franklin, M.R. (1977). Inhibition of mixed-fiinc-
of methylenedioxyphenyl (1,3-benzodioxole) com- tion oxidations by substrates forming reduced
pounds with enzymes and their effects on mam- cytochrome P-450 metabolic-intermediate com-
mals. Drug. Metab. Rev. 3, 231-301. plexes. Pharmacol. Ther. A. 2, 227-245.
225. Buening, M.K. and Franklin, M.R. (1974). The 237. Franklin, M.R. (1991). Cytochrome P450 meta-
formation of complexes absorbing at 455 nm from bolic intermediate complexes from macrolide
cytochrome P-450 and metabolites of compounds antibiotics and related compounds. Meth. Enzymol.
related to SKF 525-A. Drug Metab. Dispos. 2, 206,559-573.
386-390. 238. Larrey, D., M. Tinel, and D. Pessayre (1983).
226. Omura, T. and R. Sato (1964). The carbon monox- Formation of inactive cytochrome P-450
ide-binding pigment of liver microsomes. I. Fe(II)-metabolite complexes with several erythro-
Evidence for its hemoprotein nature. J. Biol. Chem. mycin derivatives but not with josamycin and
239, 2370-2378. midecamycin in rats. Biochem. Pharmacol. 32,
227. Estabrook, R.W., J.A. Peterson, J. Baron, and A.G. 1487-1493.
Hildebrandt (1972). The spectrophotometric 239. Delaforge, M., E. Sartori, and D. Mansuy (1988).
measurement of turbid suspensions of cytochromes In vivo and in vitro effects of a new macrolide
associated with drug metabolism. In C.F. Chignell antibiotic roxithromycin on rat liver cyto-
(ed.). Methods in Pharmacology, Vol. 2. Appleton- chrome P-450: Comparison with troleandomycin
Century-Crofts, New York, NY, pp. 303-350. and erythromycin. Chem. Biol. Interact. 68,
228. Matsubara, T, M. Koike, A. Touchi, Y. Tochino, 179-188.
and K. Sugeno (1976). Quantitative determination 240. Paul, K.G., H. Theorell, and A. Akeson (1953). The
of cytochrome P-450 in rat liver homogenate. molar light absorption of pyridine ferroprotopor-
Anal. Biochem. 75, 596-603. phyrin (Pyridine Haemochromogen). Acta. Chem.
229. Ryan, D.E., RE. Thomas, D. Korzeniowski, and Scand. 7, 1284-1287.
W. Levin (1979). Separation and characterization 241. Waterman, M.R. (1978). Spectral characterization
of highly purified forms of liver microsomal of human hemoglobin and its derivatives. Meth.
cytochrome P-450 from rats treated with poly chlo- Enzymol. 52, 456-463.
rinated biphenyls, phenobarbital, and 3-methyl- 242. Klingenberg, M. (1958). Pigments of rat liver
cholanthrene. J 5?o/. Chem. 254, 1365-1374. microsomes, ^rc/?. Biochem. Biophys. 75, 376-386.
230. Ryan, D.E., S. lida, A.W. Wood, RE. Thomas, 243. Strittmatter, P (1960). The nature of the heme
C.S. Lieber, and W. Levin (1984). Characterization binding in microsomal cytochrome b5. J. Biol.
of three highly purified cytochromes P-450 from Chem. 235, 2492-2497.
hepatic microsomes of adult male rats. J. Biol. 244. Strittmatter, P, P Fleming, M. Connors, and
Chem. 259, 1239-1250. D. Corcoran (1978). Purification of cytochrome b3.
231. Ryan, D.E., RE. Thomas, and W. Levin (1980). Meth. Enzymol. 52, 97-101.
Hepatic microsomal cytochrome P-450 from rats 245. Guengerich, F.P. (1994). Analysis and characteriza-
treated with isosafrole. Purification and character- tion of enzymes. In A.W. Hayes (ed.). Principles
ization of four enzymic forms. J. Biol. Chem. 255, and Methods of Toxicology. Raven Press, Ltd.,
7941-7955. New York, NY. pp. 1259-1313.
232. Jefcoate, C.R. (1978). Measurement of substrate 246. Phillips, A.H. and R.G. Langdon (1962). Hepatic
and inhibitor binding to microsomal cytochrome triphosphopyridine nucleotide-cytochrome c reduc-
P-450 by optical-difference spectroscopy. Meth. tase: Isolation, characterization, and kinetic studies.
Enzymol 52, 258-279. J. Biol. Chem. 237, 2652-2660.
233. Omura, T. and R. Sato (1964). The carbon monox- 247. Strobel, H.W and J.D. Dignam (1978).
ide-binding pigment of liver microsomes. II. Purification and properties of NADPH-
Solubilization, purification, and properties. J. Biol. cytochrome P-450 reductase. Meth. Enzymol. 52,
Chem. 239, 2379-2385. 89-96.
234. Elcombe, C.R., J. Bridges, R.H. Nimmo-Smith, and 248. Wang, H., R. Dick, H. Yin, E. Licad-Coles, D.
J. Werringloer (1975). Cumene hydroperoxide- Kroetz, G. Szklarz et al (1998). Structure-function
mediated formation of inhibited complexes of relationships of human liver cytochromes P450 3 A:
methylenedioxyphenyl compounds with cytochrome Aflatoxin Bl metabolism as a probe. Biochemistry
P450. Biochem. Soc. Trans. 3, 967-970. 37, 12536-12545.
Index

Abiraterone, 292 AflatoxinGl,384


Absorption anisotropy, 93 Age effects on P450, 347, 350, 536, 537
ABT, see 1-Aminobenzotriazole Agrobacterium tumafaciens, 589
Acenocoumarol, 411 Ah receptor. See Aryl hydrocarbon receptor
Acetaldehyde, 28-30, 419 Ah responsive elements, 338
Abscisic acid, 259 AIA, see 2-lsopropyl-4-pentenamide
9'-propargyl-, 259 Aldehyde, as a product, 75
Acetaminophen, 119, 211, 331, 394, 395, 399, 422, 638 Aldehyde oxidation of
cooperativity in interaction, 429 to carboxylic acids, 217, 221, 284, 419
Acetanilide, 632 CO2 loss, 222
Acetone, 419 via deformylation, 221, 222, 284
Acetonitrile, 409 isoform specificity, 221
4-Acetoxyandrost-4-ene-3,17-dione, 289 P450 inactivation, 222, 282, 284
Acetylenes, oxidation of ALDH3A1,338
inactivation of P450, 200, 256, 269, 270 Aldosterone biosynthesis, 135, 292
mechanism, 198 Aldosteronism, P450 link to, 383
metaboHsm of, 198, 200, 270 Alkaloids, 553-555, 557, 571, 572
oxirene, 258 AlkB, 15
rearrangement during, 198, 270, 271 N-Alkylprotoporphyrin IX, see Heme
Acidovorax avenue, 568 see Protoporphyrin IX
Acrylonitrile, 384, 420 Allelochemicals, 553
ACTH, 445, 446, 453 Allene oxide synthase, 158, 555
Activating transcription factor-1 (ATF-1), 446,447 AUenes, 283, 289
Activating transcription factor-2 (ATF-2), 446, 447 Allobarbital, 256
Active site, see P450 Allylisopropylacetamide, see 2-Isopropyl-4-
Acyl CoA, 322 pentenamide
Ad4 Binding protein. See Steroidogenic Factor 1 4-Allyloxymethamphetamine, 636
Adamantane, 28, 31,32 Alprazolam, 622, 630
perdeuterated, 64 Alzheimer's disease, 461
Addison's disease, 446, 450 Amides
AdE element, 445 oxidation potential, 197
Adenosine 2',5'-diphosphate, 127, 128 isotope effect, 197
Adrenal hyperplasia, P450 link, 383,446-448, 453 Amine oxidation, 193
Adrenarche, 450 4-Aminoandrostan-1,4,6-triene-3,17-dione, 289
Adrenodoxin, 116, 134, 454 2-Aminoanthracene, 401, 436
interaction with adrenodoxin reductase, 98, 134, 137 O-Aminoazotoluene, 401
interaction with P450, 135, 137, 419, 445 1-Aminobenzotriazole, 275, 276, 294, 641
maturation, 135 N-benzyl-, 275
structure of, 97, 99, 136, 137 N-(a-methylbenzyl)-, 275
Adrenodoxin reductase, 116, 134, 454 N-(a-ethylbenzyl)-, 275
structure of, 97, 99, 136, 137 4-(4'-Aminobenzyl)-2-oxazolidinones, 288
Adriamycin, 598 4-Aminobiphenyl, 384, 401
AF-1,323 (22/?)-22-Aminocholesterol, 286
AF-2, 333 6-Aminochrysene, 384,401
AFK108,291 -l,2-diol,401
AflatoxinBj, 211,629 2-Aminofluorene, 384, 401, 436
biosynthesis, 224, 225, 603 Aminoglutethimide, 250, 287, 445, 452
oxidation of, 384, 409, 423, 425, 431-433 pyridyl-, 250, 287
enhancement by aNF, 427, 428 2-Amino-6-methyldipyrido[l,2-a:3,2'-</|imidazole, 384

659
660 Index

2-Amino-3-methylimidazo[4,5-/|quinoline, 384 Antibodies contd.


2-Amino-1 -methyl-6-phenylimidazo-[4,5-Z>]pyridine, 384 autoantibodies, 262, 275,410,418,422,443,446,450
3-Amino-1 -methyl-5i/-pyrido[4,3-Z7]indole, 384 inhibition of activities, 390-392
l-Aminopyrene, 401 phage display library, 390
Aminopyrine, 638 selectivity, 390, 405
1 -Amino-2,2,6,6-tetramethylpiperidine, 267 to quantitate P450, 251, 390-392
3-Amino-1,2,4-triazole, 280, 421 Antifungal agents, 585-587, 592, 593, 598, 603,
Amiodarone, 407, 430 604,607
Ammi majus, 223 Antipyrine, 398, 399
Amodiaquine, 407 Antisense, 543
Amphetamine, 285 ttj-Antitrypsin, 443
Amphotericin B, 593, 598-600, 606 AP-1,446,459
Amycolatopsis mediterranei, 595 AP-2, 442, 445
Amycolatopsis orientalis, 106 Apium graveolans (celery), 553
Anastrozole, 287, 288 Apoptosis, 116
Androgen oxidation Aprobarbital, 256
aromatization, 379 ARA9, 336, 337
hormonal effects on P450, 350 Arabidopsis thaliana, 225, 259, 554, 555, 558, 560
2a-hydroxylation, 348 CY51 phylogenetic tree, 591
16a-hydroxylation, 348 CYP61, absence of, 592
Androgens gene absence in, 556, 568
depletion by cisplatin, 362, 363 host for foreign pathways, 562, 564, 566, 571, 573, 574
hormonal effects, 350 Arachidonic acid
imprinting, 349 binding to P450, effects, 132
Androstane receptor (CAR), 323, 326, 327, 406,424 in disease pathophysiology, 531
ligands,328,331 in glycerophospholipids, 535, 541
nuclear translocation, 330 metabolism of, 292, 378, 408, 535-537
nullmice, 330, 331 hydroxylation, 379, 434, 436, 437, 595, 596,
phosphorylation, 329, 331 621,631
role, 405, 408 epoxidation, 423, 531, 539-541, 545
subcellular localization, 329 physiological roles of metabolites, 535, 538, 543-545
5a-Androstane-3a,17p-diol, 3,17-disulfate, 635 PPAR Hgand, 332
Androstanedione, 452 L-Arginine, 16
Androst-1,4-diene-3,17-dione N-hydroxy-, 16
1-methyl-, 289 5-e/7/-Aristone, 569
Androst-4-ene-3,17-dione ARNT, 336, 338, 397
as substrate, 638 Aromatase, see CYP19A1
in sterol biosynthesis, 217,218 Aromatic rings, oxidation of
Androst-5-ene-7,l 7-dione, 289 ipso substitution, 76, 185, 203, 204, 207
5,16-Androstadiene-3 p-ol, 449 isotope effects, 202, 203
1,4,6-Androstatriene-3,17-dione, 289 mechanism, 75, 76
Angiotensin II, 541 •^O studies, 204
Aniline, 639 Arrhythmia, 394
aromatic oxidation, 204 Aryl hydrocarbon receptor, 323, 324, 335
as substrate, 638, 646 Ah locus, 395
N,N-Dimethyl- Ah responsive, 400
N-demethylation, 405 allelic variations, 395, 397
N-oxidation of, 185, 194 see ARNT
isotope effects on, 194-196 see Hsp90
nitrogen radical cation pK^, 195 ligands, 335-337
oxidation potential, 195 mechanism of action, 337, 338
Anisoles, 204 negative P450 regulation, 363
Anthocyanidins, 570 null mice, 338
Anthracene, 363 photoaffinity labeling, 335
Antibodies repressor protein, 338
antipeptide, 390 Aryl migration in P450 reaction, 227, 554, 569
Index 661

Ascorbic acid, 23, 282 Benzo[a]pyrene contd.


Aspergillus 7,8-dihydroxy-7,8-dihydrobenzo[a]pyrene, 384,
fumigatus, 591, 602, 603, 608 395,401
nidulans, 586, 587 4,5-diol, 401
niger, 603 DNA adducts, 207
ochmceous, 586 6-fluoro-, 208
parasiticus, 224 3-hydroxylation, effect of aNF, 427
Astemizole, 430 as inducer, 336
Atamestane, 289 as substrate, 378, 384, 397, 401, 604, 639
Atrazine, 595 sex-dependence of, 347, 348
ATMP, 278, 279 1,2,3-Benzothiadiazole, 5-6-dichloro, 283, 284
Atomic force microscopy, 95 l,4-Benzoxazine-3-one, 567, 574
Aurones, 570 2,4-dihydroxy-, 566, 566
Autoimmune polyglandular syndrome, 450 2,4-dihydroxy-7-methoxy-, 567
Autooxidation, 153, 156 Benzphetamine, 638
Autumnaline, 205 N-3-Benzylnirvanol, 621, 626
Avermectin, 598-601 N-3-Benzylphenobarbital, 621, 626
Azamulin, 622, 627 Benzyne, 275, 276
2-Azido-3-iodo-7,8-dibromodibenzo-/7-dioxin, 335 Berbamunine, 572
7-(a-azolylbenzyl)-1 H-indoles, 288 Berberine, 571,572
Berberis stolonifera, 205
Bacillus Bergamotin, 394, 430
halodurans, 589 6,7-dihydroxy-, 430
megaterium, 95, 115, 118, 131 Bialaphos, 598
subtilis, 99, 131, 135, 138, 212, 589 Bicyclo[2.1.0]pentane, 9, 12, 68, 187, 188
thermoproteolyticus, 135 Bile acid
Baeyer-Villiger reaction, 217, 220, 221, 225 synthesis of, 439, 441, 457
Balhimycin, 206 metabolism, 327
Barbiturates toxicity, 326, 327
and cancer, 396 Bile duct ligation, 364
induction by, 386, 393, 394, 407,424, 431 Bilirubin, 331
metabolism of, 408 Bioavailability, 394, 430
Basic helix-loop-helix domain, 324 Biocatalysts, 133
Benz[a]anthracene, 363, 401 Biotin biosynthesis, 589
l,2-diol,401 /?-Biphenylhydrazine, 274
5,6-diol, 401 Bleomycin, 160
Benzaldehyde, 30, 282 Blood pressure, 543-545
Benzene Blumeria graminis, 591
as inhibitor, 247, 249 BM3R1, 131
oxidation of, 31, 202, 384, 420 Botrotiniafuckeliana, 591
computed mechanism, 75, 76, 79 BRII receptor, plants, 558
oxide, 76, 202 Bradykinin, 542
1,2,4,5-tetramethoxy-, 207 Brassinolide 26-hydroxylation, 555, 558
1-fluoro, 203 Brassinosteroids, 557, 558
l,2-difluoro-,203 biosynthesis, 559, 560
l,3-difluoro-,203 in P450 regulation, 560
poisoning by, 422 Bromotrichloromethane, 281
l,2,3-trifluoro-,203 Bufuralol hydroxylation, 399, 415, 621, 627, 636
1,2,4-trifluoro-, 203 Bupropion hydroxylation, 621, 625
Benzo[6]fluoranthene-9,10-diol, 401 Buspirone, 430
Benzo[c]phenanthrene-3,4-diol, 401 Butadiene, 384, 403
7,8-Benzoflavone, 641 Butanol binding, 646
Benzo[g]chrysene-l 1,12-diol, 401 2-Butene, 185
Benzoic acid oxidation, 226, 603 Buthiobate, 605
Benzo[a]pyrene /er^Butylacetylene, 269
7,8-dihydro-,210 tert-Buty\ methyl ether, 402, 420
662 Index

Butyraldehyde Cannabidiol, 260, 261


ISO-, 221 Capsidiol, 569
2-methyl-, 221 Captopril,418
CAR, see Androstane receptor
Caenorhabditis elegans, reductase null, 605 Carbamazepine
Cafestol, CYP7A1 inhibition, 441 cooperativity in metabolism, 429
Caffeine hypersensitivity to, 443
cooperativity, 429 P450 inhibition by, 250, 262, 263
drug interaction, 394 as substrate, 21, 386
as P450 substrate, 378, 386, 398-400, 621, 624, 633 Carbene
Calcitroic acid, 455 complex, 264, 265, 285
Calcium ring expansion, 14
dependent potassium channel, 538, 541, 542 spin state effects, 11
permeability of membranes, 542 2,3-Z7/5(Carbethoxy)-2,3,-diazabicylo[2.2.0]hex-5-ene,
inroleofEETs, 541 275,277
Caldariomyces fumago, 2 3,5-6w(Carbethoxy)-2,6-dimethyl-4-alkyl-1,4-
Caloric restriction, 365 dihydropyridine
Camalexin, 568 effect on heme biosynthesis, 272
Campestanol, 557, 559 mechanism of oxidation of, 272, 273
Campesterol, 559 metabolism of, 273
Camphene, 66 P450 destruction by, 194, 272, 280, 281, 641
Camphor, 103 Carbofuran, 63, 64
binding, 108, 646 Carbon disulfide, 250
as carbon source, 585 Carbon monoxide
hydroxylation of, 6, 134, 152, 191, 595 inabihty to bind, 404
computational studies, 12, 13, 68-72 asligand,51,264, 605
stereochemistry, 3, 64, 65, 186 as metabohc product, 265, 282, 285
product profile, 65 P450 inhibition by, 248, 641
Campylobacter jejuni, 589 differential sensitivity to, 248
CamR repressor, 595 rebinding kinetics, 421, 428
Canadine synthase, 571 cooperativity in, 428, 429
Cancer spectrum of P450 complex, 644
P450 relation to, 395, 422, 430, 443 Carbon tetrachloride, 280, 281, 283, 285, 364, 384, 420
arachidonic acid metabolism, 531 Carboxylic acid
bladder, 436 decarboxylation, 226, 227, 562
brain, 460 isotope effects on, 226
breast, 287, 289, 431, 450, 452, 554 radical, 228
carcinoma, 434 Carcinogen activation, human, 384
colon, 396, 400, 460 Carveol, 9
endometrial, 450 Carvone, formation of, 569
esophageal, 404 Castasterone, 559, 560
estrogen-dependent, 401, 402 Castration, 350, 351,543
lung, 395-397, 404, 411, 418, 460 Catalase, 162
lymphoma, 395 Compound 1, bond length, 167
parathyroid, 460 computational studies, 62
prostate, 383, 450, 460, 554 as catalyst, 258, 282
risk, decrease in, 403, 418 catalytic cycle, 150, 151
Candida iron-aryl complex, 274
albicans, 462, 589, 591, 593, 602-609 model, 17
dubliniensis, 591 Catalytic cycle
glabrata, 59\,6^^ computed states of, 50
maltosa, 602 gating of cycle, 54, 63
P450 knockout, 602 of P450 enzymes, 3, 48, 49, 150, 153, 155, 184, 606
neoformans, 603 peroxide shunt, 153, 156, 160, 184
tropicalis, 59\, 602 Catalytic species of P450
Candidiasis, 606 see Ferric hydroperoxo
Index

Catalytic species of P450 contd. Cholesterol contd.


see Ferric peroxo anion homeostasis, 458
see Ferryl species conversion to pregnenolone, 135, 211, 212, 379, 445
Catechol, 1 degradation, in P450 knockouts, 119
Catharantheus rosea, 224, 568 20,22-dihydroxy-, 445
Catharanthine, 569 4[3-hydroxylation, 425
Cathasterone, 558-560 7a-hydroxylation, 379, 439, 440, 621, 631, 637
6-deoxy-, 558-560 24-hydroxylation, 379, 461
Cationic intermediate, 8, 11, 28, 186, 192, 193, 226 27-hydroxylation, 456, 457, 458
Cation radical, porphyrin, 155 in P450 regulation, 439
CBP, 324, 333, 445 side-chain cleavage, see CYPl 1 Al
C/EBPa, 361, 405, 408, 413, 425, 433, 451 Cholic acid, 443, 457
Celecoxib, 409 Chorionic gonadotropin, 363
Cembra-2,7,11 -triene-4,6-diol, 554 Chromatin, 324
Cerebrotendinous xanthomatosis, 383, 456 Chrysene
Cerivastatin, 407 5,6-dimethyl-l,2-diol,401
COS 16949A,250,287 -l,2-diol,401
CGS 183208,250 5-methyl,401
Chalcone, 570, 571 5-methyl-l,2-diol,401
Chameleon species, see Ferryl species Cicer arietinum (chickpea), 555
Chaperones, 336 Cimetidine, 249, 641
Chenodeoxycholic acid, 440, 443, 457 Cineol, 595
Chlamydomonas reinhardtii, 592 Cinnamate 4-hydroxylation, 555, 557
Chloramphenicol, 250, 254, 255, 363 Cinnamaldehyde, 197, 282
Chloroeremomycin, 206 Circadian rythm, 440
1 -(2-Chloroethyl)-3-cyclohexyl-1 -nitrosourea, 363 Cirrhosis, 362, 364, 402, 430, 441
Chloroform, 384, 420 Cisplatin, 362, 363, 365
Chloromethane, 420 CIS proteins, 361
/>-Chloro-iV-methylaniline, 639 Citral, 282
we^fl-Chloroperbenzoic acid Citrulline, 16
as donor to P450, 161 Clofibrate, 331, 332, 354, 402
as donor to P450 model, 20, 21, 23, 32 Clopidogrel,253, 621
oxidation of acetylenes, 198 Clorgyline, 265
oxidation of olefins, 198 Clostridium, 121
P450 inactivation, 283 Clotrimazole, 330, 543, 593
Chloroperoxidase Clozapine, 399
axial ligand, 89 Coactivators, 324, 330, 333
catalysis by, 170 Cobalt, as inducer, 402
Compound I, 3, 60 ^^Cobalt source, 157
inactivation, 269 Cochliobolous lunatus, 586
properties of, 1, 156 Codeine, 365, 573
structure, 2, 89 Colchicine biosynthesis, 205
6-(4-Chlorophenyl)imidazo[2.1-b][l,3]thiazole- Colitis, 24
5-carbaldehyde 0-(3,4-dichlorobenzyl oxime, 330 Complestatin, 206, 599
Chlorpromazine, 365 Compound I, 19, 150-153, 156, 160, 167
Chlortoluron, 566 computed, 50
Chlorzoxazone, 378, 386, 419, 420, 621, 627, 636 orbital occupation, 50
5P-Cholestane-3a, 7a, 12a-triol, 425, 457 see also Ferryl complex
5P-Cholestane-3a, 7a, 12a, 25-tetrol, 457 Compoundll, 19, 151, 160
5P-Cholestane-3a, 7a, 12a, 27-tetrol, 457 Compound III, 156
Cholestanol, 457 Compound Q, 15
Cholestasis, 441 Computation, 45
3 3-Hydroxy-5-cholestenoic acid, 457 ab initio, 46, 47
7a-Hydroxy-3-oxo-4-cholestenoic acid, 457 atomic size parameters, 46
Cholesterol, 457 basis set, 46
biosynthesis, 134 bond energy terms, 47
664 Index

Computation contd. CRE/Adl element, 447, 451


CASPT2 methods, 46, 47 Cre/loxP sytem, 119
CCSD(T) methods, 46, 47 ^ra«5-/7-Coumaric acid, 557
complete active space self consistent field (CASSF), Coumarin, 386, 410, 621
46, 47, 54 3-hydroxylation, 404
configuration interaction, 47 7-hydroxylation of, 378, 403, 404, 405, 621, 624
conformational predictions, 105, 108 CREB, 445
density functional theory (DFT), 6, 11, 12, 13, 45, Cryogenic
46,51,66, 183,201,203 approaches, 3, 150, 157, 161, 185
dynamic electron correlation, 47 radiolysis, 5, 153, 158, 164-167
electronic structure, 45, 46 Cryptococcosis, 606
electrostatic effect, 47, 53, 59 Cryptococcus neoformans, 602
force constants, 46 Cubane, methyl, 192
geometry optimization, 51,52 Cumene hydroperoxide
in inhibitor design, 450 as activated oxygen source, 154
Hartree-Fock, 46, 47 heme-protein cross-linking, 281, 430
INDO, 47, 54 Cunninghammela elegans, 591
molecular dynamics simulations, 55, 64, 164, 168 Curyularia sp., 587
molecular mechanic (MM), 45, 46, 63, 65 Cushing's syndrome, 249, 447
Monte Carlo simulations, 63 Cyanide
potential energy surfaces, 47 asligand, 156, 167,248
quantum mechanical (QM), 45, 54, 56, 66, 68 as nucleophile, 258, 259, 260, 280
QM/MM,45,51,61,63,68 as product, 560, 563
SAM1,47,49 Cyanogenic glucosides, 553-555, 557, 560-562, 564,
Schrodinger equation, 46 565
semiempirical, 46, 47 Cyclic AMP, 445, 449, 454, 461
of site reactivity, 66, 421, 427 Cyclic AMP response element (CRE), 446, 449, 453,
of substrate specificity, 64, 65, 66, 68, 404,421 461
spin state, 11, 51, 53 Cycloartenol biosynthesis, 590
strain energies, 46 Cyclobutadiene, 275
valence bond model, 61 Cyclodextrin, 25
van der Waals parameters, 46, 47 Cycloheptenol, 11
wave mechanical approaches, 46, 47 Cyclohexane, hydroxylation of, 7, 28, 30, 31
zero point energy, 71 Cyclohexanecarboxaldehyde, 6, 221
Confocal laser scanning microscopy, 564 Cyclohexanone monooxygenase, 6
Congenital aromatase deficiency, 452 Cyclohexene, 3, 4, 30, 79, 185, 186, 267
Congenital adrenal hyperplasia, 446 3-hydroxymethyl, 11, 15
Conservons, 599, 601 Cyclohexilline, 280
Contraceptive failure, 430, 431 Cyclooctanol, 31
Cooperativity, in P450 Cyclooctanone, 31
early studies, 394 Cyclooctene, 25, 26, 30
heterotropic, 427-429, 428 Cyclopentenyl glycine, 561, 564
homotropic, 427^29 Cyclophosphamide
Coregulators, 333, 335 alteration of P450 regulation, 362, 363, 365
Corepressors, 324 oxidation of, 406
Coronary disease, P450 link, 383 Cyclopropane
Corticosteroid, 585-587, 610 l-aryl-2-alkyl-, 192
Corticosterone 1-methyl, 188, 192
deficiency disease, P450 link, 383 l-methyl-2-phenyl-, 188
18-hydroxylation, 379, 447 l-methyl-2,2-diphenyl-, 188
Cortisol trans-1 -methyl-2-(4-trifluoromethyl)phenyl-, 190
biosynthesis of, 135, 444, 446, 586 /ra«5-l-phenyl-2-vinyl-, 200
metabolism of, 426 Cyclopropylamine, 196, 283
Corynebacterium diptheriae, 594, 596 N-methyl-N-benzyl, 283
Cotinine, 403 17p-(Cyclopropylamino)-androst-5-en-3|3-ol, 292
CPF, 440 Cyclopropylbenzene, 202
Index 665

Cyclosporin, 281, 363, 386,425, 426, 430 CYP2A6 contd.


CYP1A1,377 content in tissues, 382, 385, 391, 402
See Aryl hydrocarbon receptor and cytochrome b5, 403
active site, 397 marker substrate for, 621, 624
in arachidonic acid metabolism, 538 polymorphism, 402-404
content in tissues, 397 regulation/induction, 386, 621
fusion protein with reductase, 133 role in drug metabolism, 386
gene organization, 396 inhibition, 386,404, 621
inhibition of, 397, 632 substrates, 386, 402
interaction with reductase, 129 tissue location, 378, 402
polymorphism of, 395, 397 CYP2A7
reactions, 378 chimeric proteins, 404
regulation/induction, 395, 396 tissue content, 404
in smoking/lung cancer, 395-398 tissue location, 378
substrates, 397, 540, 632 CYP2A13
tissue location, 378, 396, 397 tissue location, 378, 404
CYP1A2, 377 substrates, 405
active site, 399 variants, 405
aldehyde deformylation, 221 CYP2A21,363
in arachidonic acid metabolism, 538 CYP2B 1,560
carcinogen activation by, 384, 396 dehydrogenations by, 209
and colon cancer, 396 disease associations, 404
content in tissues, 381, 382, 385, 391, 398 substrates, 4, 195, 207, 540, 634
cooperativity, 399 and radical clocks, 190
gine organization, 398 regulation/induction, 354, 364
inhibition, 389, 399, 400, 621 threonine, conserved, 302, 256
marker substrates for, 621, 624, 633 CYP2B2, 540
mechanism, 399 regulatory element in, 328
polymorphism in, 398, 400 T302A mutant, 185
prototype inducer, 621 CYP2B4
regulation/induction, 395, 398, 399 aldehyde deformylation, 221
substrates, 207, 399, 540 interaction with b^, 134
tissue location, 378, 398 interaction with reductase, 95, 130
CYPIBI membrane orientation, 129
active site, 402 substrates, 197, 540
carcinogens activated by, 401 structure of, 93
content in tissues, 400 T302A mutant, 191, 222, 258, 282
disease linked to it, 383, 396, 400 CYP2B6
gene organization, 400 active site, 406
inhibition, 402, 633 content in tissues, 382, 385, 405
null mouse, 395, 401 cooperativity, 406
regulation/induction, 393, 395, 400 inhibition, 406, 621
substrates, 380, 400, 401, 402, 633 polymorphism, 406
tissue location, 378,400 regulation/induction, 621, 405
CYP2A1 role in drug metabolism, 386, 406
hormonal regulation of, 349-353, 362, 364 substrates, 406, 621,625
substrates, 633 tissue location, 378,405
CYP2A2 CYP2B10, 328
hormonal regulation of, 349, 350, 353, 354, 363 CYP2B12, 540
male specificity of, 348 cyp2bl9, 540
substrates, 634 cyp2b37, 540
CYP2A6, 377 cyp2b38, 540
active site, 403, 404 cyp2b39, 540
and cancer, 396 cyp2b40, 540
carcinogen activation by, 384, 396 CYP2C family, liver content, 382
diseases associated with, 402 CYP2C 1,540
Index

CYP2C2, 540 CYP2C19co«^^.


CYP2C3,221 polymorphism, 394, 407, 411, 412, 413
CYP2C5 regulation/inducers, 386
crystal structure, 88, 93, 94, 100, 102, 105-108, 183 role in drug metabolism, 386
CYP2C6, 634 substrates, 386, 412, 540, 621, 626
CYP2C7 tissue location, 378, 412
induction, 364 CYP2C23, 540, 541,543
hormonal regulation of, 349, 351, 353, 363 CYP2C24, 540
substrate, 635 CYP2C34, 540
CYP2C8, 540 CYP2D2
active site, 407 inhibition, 636
content in tissue, 407 substrate, 636
inhibition, 386, 408, 621, 625 CYP2D6
polymorphism, 407 activesite, 416, 417
regulation/induction, 386, 407, 621 content of tissues, 381, 382, 385, 391,413
structure, 407 dehydrogenation by, 209
substrates, 386, 407, 621, 625 disease links, 418
tissue location, 378,407 fusion protein with reductase, 133
CYP2C9, 377 inhibition, 386, 389, 394, 417, 621
active site, 409,410 mitochondrial, 380
content in tissues, 385,408 polymorphism, 296, 377, 386, 394, 414, 418
cooperativity, 409 regulation/induction, 386, 413
and cytochrome b5, 134, 409 role in drug metabolism, 380, 383, 386, 413
inhibition, 386, 410, 411,621 substrates, 386, 415, 621, 626
polymorphism, 394, 408, 409, 410 surrogate oxygen donors, differences, 416
regulation/induction, 386, 408, 621 tissue location, 378,413
role in drug metabolism, 380, 383, 386, 392, 410 CYP2E1
structure, 93, 410 acetaldehyde deformylation, 221
substrates, 102, 386, 409, 540, 621, 625 active site, 420
tissue location, 378,408 carcinogen activation, 384, 396
CYP2C11 content of tissues, 381, 382, 385, 391, 419
fusion protein with reductase, 133 cytochrome b5 effects, 420
induction, 364 inhibition, 386, 421, 621, 627, 636
hormonal regulation of, 349-355, 357, 361, 363 null mouse, 395, 420, 422
regioselectivity, 540 polymorphisms, 419, 422
sex specificity, 348 regulation/induction, 364, 386, 419, 621
substrates, 540, 635 role in drug metabolism, 386
CYP2C12 structure model predictions, 66
hormonal regulation, 348, 350, 353-355, 361, 363 substrates, 380, 386, 420, 540, 621, 627, 636
substrate, 635 T303A mutant, 185,269
CYP2C13 tissue location, 278, 418, 419
hormonal regulation of, 348-350, 354, 357 CYP2F1
induction, 364 content of tissues, 422
substrate, 635 tissue location, 378, 422
CYP2C18 CYP2J2
content of tissue, 411 polymorphism, 423
inducers, 621 substrates, 423, 538, 540
inhibitors, 411,412, 621 tissue location, 378
polymorphism, 411 CYP2R1,378,423
substrates, 411, 540, 621, 626 CYP2S1
tissue location, 378, 411 induction, 393, 423
CYP2C19 tissue location, 378
active site, 413 CYP2U1,378,423
content of tissues, 381,412 CYP2W1,378,423
cooperativity, 427 CYP3A1
inhibition, 252, 386, 413, 621 dehydrogenation by, 209
Index 667

CYP3A2 CYP4A4, 534


hormonal regulation of, 349, 353, 363 CYP4A5, 534
substrate, 636 CYP4A6, 534
CYP3A4, 377 CYP4A7, 134,534
active site, 426-428 CYP4A8, 534, 537, 545
C-C cleavage, 228 Cyp4al0,534, 537, 538
carcinogen activation by, 384 CYP4A11,534,538
content in tissues, 381, 382, 384, 391, 424 active site, 434, 435
cooperativity, 427 content of tissues, 434
and cytochrome b5, 115, 134, 426 heme covalent binding, 435
dehydrogenation by, 209 inhibitors, 435, 622, 630
disease links, 430 regulation/induction, 434, 622
fusion protein with reductase, 133 substrates, 434, 622, 630
inhibition, 386,430, 621, 627 tissue location, 378, 434
polymorphism, 394 Cyp4al2, 534, 537, 538, 544
regulation/induction, 386, 394, 424, 621 Cyp4al4, 534, 537, 538, 543, 544
role in drug metaboHsm, 380, 383, 386, 392, 425 CYP4A22, 538
structure, 427 content of tissues, 435
substrates, 380, 386, 425, 426, 621, 627 substrates, 534
tissue location, 378,424 tissue location, 378, 435
CYP3A5 CYP4B1
active site, 432 active site, 436
content of tissues, 431 dehydrogenation by, 209
inhibition, 432 disease links, 383
polymorphism, 431 fusion protein with reductase, 435
regulation/induction, 431 heme covalent binding, 436
substrates, 432, 622, 629 substrates, 435, 436
tissue location, 378, 431 tissue location, 378, 435
CYP3A6,221 CYP4F1,533,535
CYP3A7 CYP4F2, 533
active site, 433 inducers, 622
content of tissues, 432, 433 inhibitor, 622, 631
inhibition, 433 substrates, 436, 535, 538, 622, 631
regulation/induction, 433 tissue location, 378,436
substrates, 433, 622, 630 CYP4F3, 533
tissue location, 378,432 substrates, 436, 535
CYP3A9 tissue location, 436
hormonal regulation, 349, 353 CYP4F3B, 535
substrate, 637 tissue location, 378
CYP3A18 CYP4F4, 533, 535
hormonal regulation, 348, 349, 353 CYP4F5, 533, 535
CYP3A23, 636 CYP4F6, 533, 535
CYP3A43 CYP4F8, 533
content of tissue, 434 substrates, 437
regulation/induction, 434 tissue location, 378, 437
tissue location, 378, 434 CYP4F11,533
CYP4A1,537 tissue location, 379, 437
disease links, 383 CYP4F12, 533
fusion protein with reductase, 133 substrates, 622, 631
inhibition, 271, 272, 637 tissue location, 379,437
substrates, 292, 331, 533, 534, 637 Cyp4fl3,533
CYP4A2, 534, 538 Cyp4fl4, 533
disease link, 543 Cyp4fl5,533
hormonal regulation, 353, 537 Cyp4fl6,533
regulation/induction, 364 Cyp4fl7,533
CYP4A3, 364, 534, 537, 538 CYP4F22, 379, 437, 534
Index

Cyp4f37, 533 CYPllAl i?450scc) contd.


Cyp4f39, 533 substrates, 445
CYP4V2, 379, 437 tissue location, 379, 443
CYP4X1,379,437 CYP11B1(P45011P)
CYP4Z1,379,437 active site, 447
CYP5A1 (thromboxane synthase) adrenodoxin interaction, 447
active site, 439 disease links, 383
disease links, 383 inhibition, 249, 447
inhibition, 439 polymorphism, 446
polymorphism, 438 redox partners, 135
properties, 533 regulation, 446
reaction, 379, 438 substrates, 446, 447
regulation/induction, 438 tissue location, 379, 446
substrates, 438, 439 CYPllB2(P450aldo)
tissue location, 379, 437, 438 active site, 448
CYP7A1,327 disease links, 383
actives site, 441 inhibition, 448
content in tissues, 439 regulation, 447
disease links, 383 substrate, 448
inhibition, 441,637 tissue location, 379, 447
instability of, 440 CYP17A1
phosphorylation, 440 active site, 449, 450
polymorphisms, 440 disease links, 383, 450
regulation/induction, 439, 440, 622 effect of cytochrome b5, 134, 448, 449
substrates, 439, 440, 622, 631, 637 fusion protein with reductase, 133
tissue location, 379, 439 inhibition, 292, 450
CYP7B1 mechanism, 216, 449
disease links, 383 mutants, 217
substrates, 441 pH dependence of products, 217
tissue location, 379, 441 phosphorylation, 449
CYP8A1 (prostacyclin synthase) polymorphism, 449, 450
active site, 442 regulation, 449
disease links, 383, 443 substrates, 215, 449
gene organization, 442 tissue location, 379, 448
inhibition, 442 CYP19A1 (aromatase)
membrane topology, 442 active site, 452
polymorphism, 442 disease linked to it, 289, 383, 452
properties, 533 gene organization, 451
reaction catalyzed, 379, 438 inhibition of, 248, 250, 271, 286, 287, 289, 452
substrates, 441,442 isotope effects in, 218
tissue location, 379, 442 polymorphisms, 451, 452
CYP8B1 reaction, 6, 217, 218, 219, 451, 452
regulation, 443 regulation, 451
substrates, 443, 622, 631 substrates, 452
tissue location, 379, 443 tissue location, 379, 451
CYPllAl (P450scc),211 CYP20A1,379,452
active site, 445 CYP21A2
adrenodoxin interaction, 445 active site, 453
CO sensitivity, 248 disease links, 383, 453
deficiency, 446 inhibition, 386, 453
disease links, 383 regulation/induction, 386, 453
inhibition, 271, 445 role in drug metabolism, 386
interaction with reductase, 135, 136 substrates, 386, 453
polymorphism, 445 tissue location, 379, 453
reactions, 286 CYP24A1
regulation, 445 active site, 455
Index

CYP24A1 contd. CYP61,602,604


disease links, 383 22-desaturation, 586, 592, 601-603
inhibition, 455 CYP64, 603
regulation, 454, 455 CYP71, 9, 555-557, 562-569
substrates, 380, 455 CYP72, 555, 569
tissue location, 379, 454 CYP73A5, 555, 557
CYP26A1 CYP74A1,555
substrate, 456 CYP74B2, 555
tissue location, 379, 456 CYP75B 1,555
CYP26B1 CYP79, 555-557, 562-566, 573, 574
substrate, 456 CYP80, 555
tissue location, 379, 456 CYP83, 555, 574
CYP26C1 CYP84A1,555
tissue location, 379,456 CYP85, 555, 558, 560
CYP27A1 CYP86A1,555
active site, 458 CYP86A8, 555
disease links, 383 CYP88A, 225, 226, 555
polymorphisms, 456 CYP90A1, 555, 558,560
regulation, 440, 456 CYP90B1,555,560
substrates, 457 CYP93C, 226, 227, 555, 571
tissue location, 379,456 CYP98A3, 555
CYP27B1 CYP101,5eeP450,^
active site, 460 CYP102, see P450BJ^.3
disease links, 383 CYP105, 595, 596, 598-601
gene organization, 459 CYP108,5eeP450j^^p
mutations, 459, 460 CYP107A1
regulation, 459 crystal structure, 183
substrates, 460 hydrogen bonding network, 164
tissue location, 379,459 CYP107, 599, 601
CYP27C1,379,460 CYP116,595
CYP39A1,379,460 CYP119,596
CYP46A1,379,461 complex with imidazoles, 100
CYP51 (sterol 14-demethyIase), 555, 590, 594, 601, 602 crystal structure, 88, 91, 100, 183, 601
active site, 462, 609 reaction with peroxides, 3, 161
in azole resistance, 607-609 thermal stability, 91, 601
mutations leading to, 608, 609 CYP121,592,594,598
complementation by, 558 CYP122, 599
and cytochrome bj, 462 CYP123, 594
fusion protein with ferredoxin, 588, 592, 594 CYP124, 594
inhibition, 120,250,603 CYP125, 592, 594, 596, 601
knockout in bacteria, 592, 604 CYP126, 594
phylogram of sequences, 590, 591, 596, 597 CYP128, 594, 598
reaction, 217, 217, 219-221, 379, 585-587 CYP129, 599
regulation, 461 CYP130,594
structure of, 88, 110, 107, 183,462 CYP131,599
substrates, 461 CYP132,594, 598
tissue location, 379,461 CYP135,594,598
CYP52, 602 CYP136,594
CYP54, 603 CYP137, 594, 598
CYP55Al,5eeP450^^^ CYP138, 594
CYP56, 602 CYP139, 594, 598
dityrosine formation, 603 CYP140. 594, 598
CYP57, 603 CYP141.594,598
dityrosine formation, 601 CYP142, 594
CYP58, 603 CYP143, 594, 598
CYP59, 603 CYP144, 594
670 Index

CYP151,598 Cytochrome c peroxidase contd.


CYP152A1 properties, 154
reaction, 170 resting state, 51
structure, 170 Cytochrome oxidase, 152, 165
CYP154, 599,601 Cytochrome P450 reductase, 3
crystal structure, 106, 110, 183, 600 alternative electron acceptors, 120
CYP156,599,601 antibodies to, 222
CYP157, 599, 601 fungal, 602, 605
CYP158,600,601 chemical crosslinking to P450, 129
CYP 160, 599 chromosome location, 119
CYP161,599 Class I, 587
CYP 162, 599 Class II, 115,587,589,596
CYP163, 599 determination of activity, 647'
CYP164, 592, 594, 598 diversity of, 118
CYP 170, 590, 592, 594, 596, 599 electron flow, 124, 125, 127, 128, 131, 133
gene deletion of, 599 flavin orientations, 96, 97
CYP171,599,601 fusion proteins with, 115, 116, 133, 587, 601, 603
CYP 175 A 1,596 gene organization, 118, 119, 599
structure, 92, 601 hydride transfer, 122, 124, 125, 126, 127
CYP176Al,5e^P450cin inhibition, 123,127,266
CYP178,601 interaction
CYP180A1,598,601 with cytochrome b5, 133, 134, 606
CYP181,601 with cytochrome c, 116, 129, 130
CYP504, 586, 587 withP450,95, 116, 128,129, 130
CYP505, 115,603 ionic strength effects, 130
fusion protein, 603 kinetic scheme, 126, 127
CYP701A3, 555 knockout, 119, 120,605
CYP710,592 membrane binding domain, 117, 118, 119, 129
Cysteine ligand, 2, 7, 50, 158 mobility, interdomain, 128-130
bondlength, 57, 59, 61 mutagenesis, 124, 128
effect on iron, 48, 52, 61, 183, 190, 248 NADPH/NADP+ binding, 122, 123, 126-128
in P420, 265 isotope effect, 124, 125
hydrogen bonding to, 48, 89, 186 in CYP55 (P450nor), 587
"push" effect, 48, 52-54, 58, 63, 155 plant, 118, 122,225
and spin state, 48, 157 photoreduction, 96
Cytochrome b5 properties, 117
alternate P450 reductase in yeast, 605, 606 ratio to P450, 129
apocytochrome b5, 134,409, 413, 426, 449 redox properties, 117, 124, 125, 128, 132, 133
complex with P450, 134, 135 regulation/induction, 119, 362, 365
concentration of, 646 structure of, 95, 97, 117, 121, 122, 123, 127
electron transfer to P450, 115 domains, 117, 118, 122, 124, 125, 129, 131
and hemoglobin, 134 truncated forms, 117
interaction with P450 reductase, 120, 133, 134 Cytokines, 388
and lipid biosynthesis, 133, 134
properties of, 133, 134 Dahl Salt Sensitive rat, 543
redox potential, 134 Daidzein, 554, 571
role in vivo, 119, 134 Dapsone, 407, 409, 426
role in individual P450s, 403, 409, 413, 426, 449 Daunorubicin, 599
and uncoupling, 134, 184 DDEP, see 3,5-Z?w(Carbethoxy)-2,6-
Cytochrome b5 reductase, 119, 133, 134, 605, 606 dimethyl-4-ethyl-1,4-dihydropyridine
Cytochrome c, 120 DDMS, 293
crosslinking to P450 reductase, 129 DDT, 330, 332
inapoptosis, 116 5-Deazariboflavin, 96, 127
Cytochrome c peroxidase Debrisoquine, 378, 383, 386, 621, 626, 636
active site structure, 162 polymorphism, 386, 387, 414, 418
Compound I, bond length, 167 Decalin, 31
Index 671

Decalol, 31 ^m(2,3-Dibromopropyl)phosphate, 384


Deforaiylation, 6 2,2-Dichloroacetamides
Dehydration reaction, by P450, 562 N-monosubstituted, 250, 263
Dehydroepiandrosterone hydroxylation, 379,433 2-(/7-bromophenethyl)-, 255
Dehydrogenation, see Desaturation 2-(p-nitrophenethyl)-, 255
5a-Dehydrotestosterone, 544 A^-(l,2-diphenethyl)-,255
Deinococcus radiodumns, 589 2,6-Dichlorobenzonitrile, 405
Delaviridine, 280, 283, 284 1,1-Dichloroethylene, 200, 420, 636
11-Deoxycorticosterone, 447, 453 1,1-Dichloroethylene epoxide, 200
1 l-Deoxycortisol, 453 1,2-Dichloropropane, 420
11-hydroxylation, 379, 446 N-(3,5-Dichloro-4-pyridyl)-4-methoxy-3-(prop-2-
6-Deoxyerythronolide B, 90, 103, 163 ynyloxy)benzamide, 272
Deprenyl,258,259,415 3-[2,3-Dichloro-4-(2-thenoyl)phenoxy]propan-1 -ol,
Desaturation, 210 621,626
acetaminophen, 211 1,4-^/5[2-(3,5-Dichloropyridyloxy)]benzene, 329
cytochrome b5 electron donor, 133 3-[2,3-Dichloro-4-(2-thenoyl)phenoxy]propan-l-ol, 411
dihydropyridine, 211 Diclofenac, 100, 102, 105, 107, 283, 284, 394,
isotope effects, 209, 210 409^11,621,626,635
hydrocarbon, 134, 208-211 cooperativity in metabolism of, 428, 429
lovastatin, 425 Dictyostelium discoideum, 591, 610
mechanism, 209 Dieldrin, 189
3-methylindole, 211,422, 436 Diet, effects on P450, 364, 365, 430, 536, 537, 541, 543
sterol, 210, 586, 592, 601-604, 608 Diethyldithiocarbamate, 250, 404, 421, 621, 627
substrates for, 208 Diethylnitrosamine, 277, 384
see Valproic acid 2,2-Diethyl-4-pentenamide, 268
Desferoxamine, 258 l,2-Difluoro-4-iodobenzene, 17
Desulfuration, 251 Dihydralazine, 275
Detergent, non-ionic, metabolism of, 425 24,25-Dihydrolanosterol, 462
Dexamethasone Dihydroquinolines, 2,2-dialkyl, 273
in decrease of CYP2C11, 363 6' ,7'-Dihydroxybergamotin, 250, 261
as inducer, 324, 325, 326, 386, 408,412, 424,434, 1,25-Dihydroxycholecalciferol, 451
438,621 7a,27-Dihydroxy-4-cholesten-3-one, 457
Dextromethorphan, 265, 426, 621, 627 7a,12a-Dihydroxy-4-cholesten-3-one, 457
cooperativity in metabolism of, 429 3p,7a-Dihydroxy-5-cholestenoic acid, 457
Developmental effects, 388, 432 7a,27-Dihydroxycholesterol, 457
DHEA,441,449 la,25-Dihydroxyvitamin D3, 425, 454, 455
16-hydroxy, 452 24,25-Dihydroxyvitamin D3,460
-3-sulfate, 622, 630 Diltiazam, 622, 629
Dhurrin, 557, 561, 562-564, 573 1,1 -Dimethylallene, 283
Diabetes effects on N,N-Dimethylaniline, see Aniline
drug metabolism, 365,422 7,12-Dimethylbenz[a]anthracene, 363, 395, 401
fatty acid oxidation, 531 3,4-diol,401
P450 expression, 362, 363, 364, 419, 537 N,N-Dimethylbenzamide, 195
manganese porphyrin effect on, 24 4,4-Dimethylbiphenyl, 64
Diallylsulfide, 254, 268 Dimethylnitrosamine, 384, 420
Diallylsulfone, 254, 268, 636 cis-3,4-Dimethyl-2-(3-pyridyl)thiazoHdin-4-one, 403
Dibenzo[<3,/]pyrene, 401 Dimethyl sulfide, 76
ll,12-diol,401 1,7-Dimethylxanthine, 403
2,3-Diazabicyclo[2.2.0]hex-5-ene, 277 1,8-Dinitropyrene, 401
Diazenes, 275 Diol, cleavage of, 211, 212
Diazepam, 426 Dioxin response elements, 337, 338
12,12-Dibromododec-ll-enoic acid, 293 Dioxygenase reductase, 588
2,3-Diazabicyclo[2.2.0]hex-5-ene, 275 Dioxiranes, 222, 223
Diazinon, 413 Disease links to mutations, 383
Dibenz[a,h]anthracene, 363 Disulfiram,386,404,421
1,2-Dibromoethylene, 420 Dityrosine, 601-603
672 Index

l,2-Dithiole-3-thione, 254 Epipodophyllotoxin, 431


Diuron, 566 Epitope mapping, 93
DMZ, 101, 102, 106, 107 Epothilone, 99, 103,595
DNA binding domain, 323 Epoxidation
DNA microarrays, 334 charge transfer, 269
Docetaxel, 431 electronic effects, 73, 185, 203
11-Dodecenoic acid, 211 see Heme adducts
10-Dodecynoic acid, 294 mechanism, 6, 73, 198, 269
11-Dodecynoic acid, 250, 293 see Metalloporphyrin
Doxorubicin, 120 olefins, 185
DR-1,440 orbital diagram of ferryl, 67, 73, 74
Drosophila, 553 see Acetylenes, oxidation of
Drug interactions, 389, 394, 430 see Aromatic rings, oxidation of
DTT, 104 stereochemistry, 19, 198
Dwarfism, in plants, 556, 558, 560 versus hydroxylation, 79
4p,5p-Epoxyandrostenedione, 290
Ebastine, 437 Epoxide hydrolase, 392, 539
Eburicol, 604 24(5)-Epoxycholesterol, 440
Econazole, 593 EPR, 17, 21, 33, 54, 156-160, 165, 167, 185, 191,
EDHF, 540, 542 194,207
EETs Ergosterol biosynthesis, 120, 210, 586, 592,
formation by P450, 292, 423, 533, 539, 541, 545 602-605, 610
in glycerophospholipids, 541, 542 Erythromycin
metabolism of, 553 biosynthesis, 163, 598-600
physiological properties, 293, 541 breath test, 426
role in membrane biology, 293 as inhibitor, 265, 394, 430
5,6-EET, 533, 535, 539, 542, 543 as substrate, 386, 425, 426, 428, 621, 628, 637
8,9-EET, 533, 535, 539 Equilenin, 205
11,12-EET, 535, 538, 539, 540, 542 ERK1,455
14,15-EET,533,545,539 ERK2, 455
EGF signaling, 541 ERK5, 455
Eicosanoids, 531-534, 543 Esterase, 392
metabolism of, 380, 533, 535 Estradiol
Electric field, protein, 51 biosynthesis, 450, 452
Electron correlation, 47 metaboHsm, 204, 378, 380, 384, 399, 400, 401, 633
Electron density, 191 enhancement by aNF, 427
unpaired, 186, 204 in P450 regulation, 363, 364
Electron transfer, 115, 124 Estriol, 452
conformational gating, 128 Estrogen
partners, classes of, 115 biosynthesis, 218
pathway, 96, 127 -dependent tumors, 401, 402
toP450, 183, 124-128, 131-133 in P450 regulation, 351, 440
from radical to iron, 192 Estrone, 204, 399, 401, 450, 452
see SET mechanism Ethane, 69, 71,267
Electrostatic terms, 51 Ethanol
Ellipticine, 398, 641 asinducer, 364, 386, 621
Embryogenesis, 119 as substrate, 418, 419, 421
Endocrine regulation, 347, 348 tolerance of microbes, 586
Endoplastic reticulum proliferation, 328 4-Ethinylbiphenyl, 271
Endotoxin, 440 7-Ethoxycoumarin, 258, 260, 260, 406,422, 621, 639
ENDOR, 17, 161, 165, 185, 191, 192 isotope effect on oxidation, 194
Enhancer elements, 324, 328, 336, 405 7-Ethoxyresorufin, 397, 400, 621, 624
Enthalpies of formation, 47 7-Ethoxy-4-(trifluoromethyl)coumarin, 258, 621, 639
Epidermin, 565 Ethyl carbamate
Epidermophyton spp., 603 activation of, 384, 420
Epiheterodendrin, 564, 565 desaturation of, 211
Index 673

Ethyl methanesulphonate, 556 Ferric hydroperoxo contd.


Ethylbenzene oxidation, 9, 163 bond length, 57
stereochemistry, 9, 25 in amine oxidation, 197
Ethylene in olefin epoxidation, 201
from diethylnitrosamine, 277 dissociation, in peroxidases, 154, 160
computational studies of oxidation, 6, 73, 74, 77, 201 EPR parameters, 159
heme N-alkylation, 74, 199, 255, 267, 268 as P450 intermediate, 3, 5, 48, 56, 151, 153, 156,
Ethylene glycol, in radiolysis, 158 157, 183
2-Ethylhexanoic acid, 209 pH dependence of role, 217
2-(Ethylhexyl) phthalate, 332 pKa, 57, 59
Ethylmorphine, 347, 348, 639, 646 protonation, 54, 56, 58, 63, 155, 158, 164
19-(Ethyldithio)androst-4-ene-3,17-dione, 289 spectrum of, 160
7-Ethynylcoumarin, 250, 258, 259 spin state, 155
17a-Ethynylestradiol, 250, 256, 258, 259, 394, 406, stability of, 157
423,430,431 structure, 57
1-Ethynylnaphthalene, 250 Ferric hydrogen peroxide complex, 51
2-Ethynylnaphthalene, 250, 257, 258, 263, 271 Ferricyanide, 274
2-Ethynylphenanthrene, 271 Ferrocene, 102, 108
3-Ethynylphenanthrene, 271 Ferrous dioxygen complex
9-Ethynylphenanthrene, 271, 625, 634 computational studies, 54-57, 168
np-Ethynylprogesterone, 250, 256, 257 pKa, 56
18-Ethynylprogesterone, 447 as P450 intermediate, 3, 48, 150, 151, 153, 183
1-Ethynylpyrene, 250, 256, 257, 270, 271, 285 protonation of, 56, 57, 63, 155, 156, 161
2-Ethynylpyrene, 402 radiolysis of, 166
Evolutionary relationships, 541, 555, 567, 596 resonance Raman of, 156
EXAFS, 17 spectrum of, 160
Exemestane, 288, 289, 452 stabilization of, 55, 167
Expressed sequence tag (EST), 554, 557, 567, 575 structure of, 167
Ezlopitant, 209, 427 Ferryl species, 6, 48, 59, 183
and active site polarity, 79
FAD, 115, 116, 118 calculations on, 51, 66, 77, 78
affinity for, 122 chameleon species, 60-63, 79
domain, 121, 127 electronic state, 59-62, 66, 67, 72, 74, 77
reduction of, 124, 126, 133 formation of, 3, 58, 155, 156, 161
Fadrozole, 287 electrophilicity of, 77
Farnesoid X receptor (FXR), 440, 443 ligand dependence of properties, 59
Fasting, 331, 334, 537 role in substrate oxidation, 8, 11, 150, 151, 153, 186,
Fatty acid, 131 192
o) and (0-1 hydroxylation, 213, 292, 378, 420, 434, structure, 167
435, 532, 535-537, 539, 545, 555, 621, 631 see also Compound I
w-2, (0-3 hydroxylation, 621, 631, 595, 596 Ferulic acid hydroxylation, 555
metabolism, 330, 331, 380, 603 a J-Fetoprotein transcription factor (FTF), 443
P-oxidation, 539 Fexofenadine, 394
Felbamate, 429 Filipin biosynthesis, 601
Feminization, 446 Finasteride, 425
Fer, 135 FK506,281,598
Ferredoxin, 115, 135 Flash photolysis, 127, 136
fusion protein with, 588, 592, 594 Flavin-containing monooxygenase, 6, 253
Ferredoxin reductase, 115, 118, 135 Flavin hydroperoxide, 6
Ferric peroxo anion, 5, 6, 150, 155, 183, 185, 215, 217, Flavanone, desaturation of, 211
219,220,222 Flavodoxin, 118, 120, 121, 132, 138,213
computational studies, 54, 55 as redox partner to P450cin, 588
reaction with nitrile, 425 as fusion domain on P450, 588
Ferric hydroperoxo Flavodoxin reductase, 588
as oxygen donor, 5, 6, 19, 66, 77, 152, 185, 186, Flavonoid
190-192 biosynthesis, 570
674 Index

Flavonoid contd. Glucosinolates, 553-555, 557, 564, 565, 574


3'-hydroxylase, 555 Glutathione, 24, 539
6-hydroxylase, 569 Glutathione reductase, 98
Flavones, 394 Glutathione transferase, 539
Fluconazole, 250, 290-293, 386, 432, 604, 605 Glycerol, effects of, 128, 158, 159
resistance to, 606, 608, 609 Glycine max (soybean), 566, 569, 571
structure of, 607 /'-Glycoprotein, 394, 424, 426
Fluometuron, 566 Glycyrrhiza echinata (licorice), 571
Fluorescence Gonadal hormones, 350, 366
emission, 126 Grapefruit juice, 261, 262, 394,430
quenching, 428 Green fluorescent protein, 563
Fluorescent reporter group, 103 "Green pigment", 267, 277, 278
Fluoranthene-2,3-diol, 401 GRIP-1,330
15a-Fluorolanost-7-en-3p-ol, 291 Griseofulvin, 278, 279, 402
20-Fluoro-17(20)-pregnenolone, 292 Grotthuss mechanism, 56, 162
5-Fluorouracil, 363 Growth hormone
Flurbiprofen, 409 age-dependence, 350
Fluroxene, 268 in regulation of P450, 350, 351, 354, 359,
Fluvoxamine, 386, 399, 642 363, 459
FMN, 115, 116, 118, 132 receptor, 355-357, 362
affinity for, 121 secretory pattern, 351-353, 355-360
binding domain, 120, 121, 129 alteration by xenobiotics, 363, 364
reduction of, 124 GSTYa, 338
Formic acid, as product, 282, 283 Gynecomastia, P450 link, 383
Formononetin, 571
Forskolin, 459 Halobacterium NRCl, 589
FprA, 125, 127, 135 Halocarbons, 263, 283, 285
Furafylline, 279, 285, 386, 399, 400, 621, 624, 633 Halothane, 285, 403, 420, 422
Furan oxidation, 261, 262, 280 Hammett relationship, 28, 185, 195, 197
Furanocoumarins, 250, 261, 430, 553 HAT mechanism, 196, 197, 204, 211, 228
Fusarium oxysporum, 115, 169, 603 HDL, 541
Fusion proteins, 115, 116, 133, 435, 587, 588, 592, 594, Heart disease, 287
601,603 Heat shock protein. See Hsp90
see P450BM-3 Helianthus tuberus (artichoke), 555, 557
Helicoverpa zea (corn), 553
Galangin,621,624, 633 Helminthosporium turcicum, 568
Gender effects, see Sex-linked differences Hematinic acid, 282
Gene duplication, 414 Heme
Gene knockout mice, see Null mice adducts of, 260, 267, 272, 273, 281, 283
Gene organization, 555, 567 meso-a\ky\-, 222
Gene therapy, 454 N-alkyl/arylation of
Genistein, 454, 554, 571 mechanism, 74, 75, 199, 201, 256, 257, 276
Genomes myoglobin, 274
bacterial, P450 enzymes in, 589, 599 regiochemistry, 268, 270, 274, 278
human, P450 enzymes in, 347, 378 reversibility, 269
Geosmin, 598, 601 stereochemistry, 268
Geraniol, 569 bleaching, 282
Gestodene, 250, 253, 270, 285, 386, 430, 432, 621, 628 carbene complexes, 264, 265, 285
GH, See Growth hormone covalent binding to protein, 435, 436
Giardia lamblia, 590 of fragments, 280, 281, 430
Gibberellafujikuroi, 226, 229, 603 iron-alkyl/aryl complexes, 273, 274, 275, 285
Gibberellin biosynthesis, 225, 228, 229, 603 irradiation to disrupt complexes, 264
Glaucoma, P450 link, 383, 396, 400, 401 nitroso complexes of, 266
Glucocorticoid, 324 quantitation of, 645
receptor, 326, 446 reduction potential, 89
(3-Glucosidase, 560 Heme degradation, 155, 251
Index 675

Heme oxygenase, 120, 150, 151, 153 Horseradish peroxidase


hydroperoxo complex, 159 axial ligand, 51
Hemoglobin, 156, 195,281 catalytic residues, 154
hydroperoxo complex, 158, 159 Compound I, 195
iron-aryl complex, 274 iron-oxygen bond length, 167
Hepatic toxicity, 253, 275 rate constants for formation, 154
Heptamylose sugars, 25 Compound III, 156
Hepatocyte nuclear factors, See HNF computational studies, 62
Hepatitis A, P450 decrease due to, 404 ferric hydroperoxo complex, 158, 159
Hepoxilins, 531 substrate peroxidation, 207
Herbicides, 553, 556, 566, 595, 598, 599 substrate peroxygenation, 196, 198
HET0016,293,622,630 Host-guest complex, 25
HETEs 15-HEPTE, P450 role, 532, 533
formation by lipoxygenases, 531 Hsp90, 336, 337, 397
formation by P450, 292, 536 Humoral hypercalcemia, 459
metabolism by P450, 294, 553, 535 Hydrazines, 274
physiological roles, 293, 294 acyl-, 263
5-HETE, 535 l,l-dialkyl-,263,275
9-HETE, 535 heme destruction by, 273, 275, 281
12-HETE, 535, 536 hemoprotein complexes of, 266, 267, 274
15-HETE, 535 radical formation from, 273
16-HETE, 535 Hydrocortisone, production of, 586, 587
17-HETE, 535 Hydrogen bonds, low energy, 57
18-HETE, 535 Hydrogen peroxide, 281
19-HETE, 535, 538 Hydroperoxide, see Oxygen donors
20-HETE, 535, 538, 543-545 Hydroperoxide lyase, 555
vasoconstrictor, 538, 542, 543 20-Hydroperoxycholesterol, 286
Hexane, 1-hydroperoxy, 5 10-Hydroperoxy-4-estren-3,17-dione, 290
3,4,5,3',4',5'-Hexachlorobiphenyl, 363 Hydrostatic pressure, 165, 429
15-Hexadecynoic acid, 294 N-Hydroxy-2-acetylaminofluorene, 3 84
Hexahydro-l,3,5-trinitro-l,3,5-triazine, 588 4-Hydroxyandrost-4-ene-3,l7-dione, 289, 290
Hexamethylphosphoramide, 403,405 ;7-Hydroxybenzaldehyde, 561
3-Hexene, 267 N-Hydroxy-N' -(4-butyl-2-methylphenyl)formamidine,
Hexobarbital, 347, 348, 408, 412, 646 293
3-Hexyne, 283 5-exo-Hydroxycamphor, 167, 595
HIFla and related, 338 dehydrogenase, 595
Histone deacetylase, 324 7a-Hydroxycholesterol, 457
HIV protease inhibitors, 430 20-Hydroxycholesterol, 440
HNF, 387, 398, 443 24-Hydroxycholesterol, 440
HNFla,361,419,433 7-hydroxylation, 379, 440, 460
HNF3P,361,433 25-Hydroxycholesterol, 440, 441
HNF4a, 326, 330, 361, 402, 408, 413, 425, 433, 440, 27-Hydroxycholesterol, 440, 459
443 6p-Hydroxycortisol, 426, 621
HNF6, 361 8-Hydroxyeicosatetraenoic acid, 436
Homology models, see Sequence, structure models 12-Hydroxyeicosatetraenoic acid, 436
Hordeum vulgare (barley), 555, 561, 564, 566 12-Hydroxyheptatrienoic acid, 439
Hormone 12-Hydroxy-8,10-heptadecadienoic acid, 439
deficiencies, sex hormones, 383 12-Hydroxy-5,8,10-heptadecatrienoic acid, 547
gonadal, 350, 366 12-Hydroxy-5,8,10,14-heptadecatetraenoic acid, 439
see Growth Hormone Hydroxylamines, 266
luteninizing, 363 11 P-Hydroxylase, see CYPl IB
parathyroid, 454, 459, 538 17a-Hydroxylase/C 17,20 lyase, see CYP17A
phytohormones, 554, 556, 558, 569, 574 Hydroxylation
replacement therapy, 450 alkane, 7, 186
status, 347 allylic, 4, 9, 68, 70, 72, 73, 185, 535, 536
see Thyroid hormone barrier height, 12, 47, 70, 71, 72, 161
676 Index

Hydroxylation contd. Indole-3-glycerol phosphate, 567, 568, 574


benzylic, isotope effects, 196 Indolin-2-one, 567
bond strength, 30, 66, 196 3-hydroxy-, 567
hydrogen abstraction, 189, 193, 212 Indomethacin, 227, 285
see Isotope effects, hydroxylation Induction ofP450, 324, 621
mechanism, 186, 191 barbiturates, 386, 393, 394, 407, 424, 431
prediction of site reactivity, 66, 421, 427 charred food, 387, 399, 621
radical intermediate in, 12, 13, 66, 193, 212 clofibrate,331,332,354,402
rearrangements, 9, 68, 186 cruciferous vegetables, 399, 621
rebound mechanism, 192, 193 DDT, 387
stereochemistry, 4, 5, 9, 186, 193 dexamethasone, 324, 386, 408, 424
transition state, 4, 9, 11, 67, 69, 71 diphenylhydantoin, 387, 396
tunneling effects, 14 ethanol, 364, 365, 386, 387
versus epoxidation, 79 exercise, 399
Hydroxyl radical isoniazid,386,418,419, 621
formation, 155 mechanism of, generalized, 388
quenching, 159 PCN, 324
/7-Hydroxymandelonitrile, 561, 562, 563 phenobarbital, 402, 408, 412, 621
a-Hydroxynitrilase, 560 pharmacokinetic effects, 393, 394
^ra«5-4-Hydroxy-2-nonenal, 283 polychlorinated biphenyls, 387, 397
4-Hydroxyphenylacetaldoxime, 555, 557, 561, 562 polycyclic aromatic hydrocarbons, 419
/7-Hydroxyphenylacetonitrile, 561 via protein stabilization, 266, 399, 419, 425
7a-Hydroxyprogesterone 12-hydroxylation, 379 pyrazole, 402
17-Hydroxyprogesterone 21-hydroxylation, 379,453 see Regulation
12-Hydroxystearic acid, 436 rifampicin, 325, 386, 387, 394, 402, 407, 408, 412,
la-Hydroxyvitamin D3, 458 424,431,433,434
2 5-Hydroxyvitamin D3 via RNA increased translation, 419
la-hydroxylation, 460 via RNA stabilization, 419
24-hydroxylation, 379 smoking,386, 397, 399, 621
hydroxylation pathways, 455 St. John's wort, 430, 431
Hyperaldosteronism, 447 troleandomycin, 621, 637
Hypercholesterolemia, P450 link, 383 TCDD, 335, 397, 538
Hypercortisolism, 249 Inflammation, 24
Hyperglycemia, 363 P450 link to, 383, 402
Hyperketonemia, 363 Inhibition, 389
Hyperlipidemia, 363 acetylenes, 199,399
Hyperoxysterolemia, P450 link, 383 AIA, 255, 267, 268, 641
Hypersensitivity, idiosyncratic, 262, 410, 422, 443 aldehydes, 222
Hypertension, 253, 383, 447, 531, 532, 537, 542 1-aminobenzotriazole, 275, 276, 294, 641
Hypervitaminosis D, 455 by antibodies, 390-392
Hypoglycemia, 334 diagnostic, 621
Hypophysectomy, 351, 353, 355, 357, 362 mechanism-based, 247, 394, 430, 447, 450, 452
Hypothalamic-pituitary axis, 351, 363 allosteric stimulation of, 428
Hypothyroidism, 362 heme modification by, 250, 256, 257, 260, 263,
Hypoxia inducible factors, 120, 338 267, 284
protein modification by, 250, 252, 253, 254, 256,
Ibuprofen, 410 259, 262, 263, 267, 271, 404, 406, 410
Imidazole carbon monoxide, 248, 641
binding of, 435 computational studies of binding, 64
in P450 inhibition, 249, 593, 605 cyclopropylamines, 196
structure of P450 complex, 100 dihydropyridines, 272, 641
10-Imidazolyldecanoic acid, 622, 630 glutathione as test of type, 251, 252, 258, 259, 261,
Imipramine, 365 262,275, 280
Indigo, 403, 420 hydrazines, 267
Indole, 403, 420, 567, 568 iron coordination, 247, 274, 543
Indole-3-acetaldoxime, 555 isoform selectivity, 251, 252, 255, 261, 271, 275
Index 677

Inhibition contd. Isotope effects contd.


metabolic intermediate (MI) complex, 262, 263, 264, intrinsic, 188
265, 425, 645 inverse, 198, 202, 203
olefins, 199 masking of, 64
quasi-irreversible, 247, 250, 265 in oxidation of acetylenes, 270
partition ratio, 199, 252, 253, 257, 260, 267, 270, 279 in oxygen activation, 417
substrate competition, 247 solvent, 56, 128, 164, 165
surfactants, 285 Isovaleric acid decarboxylation, 226
sydnones, 277, 278 Issatchenkia orientalis, 591
types, 247 Itraconazole, 593, 605
Insect attractants, 554 resistance to, 608, 609
Insects, 590, 592 structure of, 607
Insecticide resistance, 553
Insulin, 364, 365, 440, 537 Jak2, tyrosine kinase, 357-359
Interferon-7 response element, 442 Jasmonate, 553, 569
Interleukins, 419, 425, 440,451
lodosobenzene, see Oxygen donors Kaempferol, 408
Ion channel regulation, 542 ent-Kaurene oxidase, 555
Ifosphamide, 362, 363, 365 e«^Kaurenoic acid, 225, 555
Indinavir, 425 Kavapyrones, 430
4-Ipomeanol, 422, 436 Ketamine, 406
Irbesartan, 411 Ketene, 257, 258
Iron-sulfiir protein, 115, 116 Ketoconazole, 120, 249, 253, 290, 291, 386, 394, 430,
Isoandrocymbine, 205 432, 433, 593, 604, 621, 642, 450
Isocyanate, a-keto-, 253 resistance to, 605, 606, 609
Isocyanide complexes, 169, 644 structure of, 607, 628, 642
Isoflavone synthase, 227, 228 suppression of P450 expression, 424
Isoflavonoids, 553-555, 569, 570 Ketoprofen, 227
Isoflurane, 420
L-Isoleucine, glucoside precursor, 561, 564, 565 L-754,394, 250, 262
Isoniazid, 266, 386, 404, 418, 419 Langmuir-Blodget, 95
Isonitriles, as product, 285 Lanosterol, 1, 219, 461, 592, 605
Isoporphyrin, 222 Lanosterol 14-demethylase, see CYP51
2-isopropenyl-2-methyladamantane, 406, 621, 625 Laser flash photolysis, 9, 96
3-Isopropenyl-3-methyldiadamantane, 406, 621, 625 Laurie acid oxidation
2-lsopropyl-4-pentenamide, 255, 267, 641 desaturation, 211
i^O-studies, 268 reconstitution of activity, 117
Isosafrole, 264, 282, 399 as substrate, 132, 283, 378, 380, 420, 434-436, 533,
Isothermal titration calorimetry, 98, 135 537, 538, 622, 630, 637
Isothiocyanate, 250 LDL, 541
benzyl-, 254 Leischmeinia, 610
^butyl-, 254 Lens culinaris (lentil), 571
phenyl-, 254 Letrozole, 287, 288
Isotope effects Leucoanthocyanidins, 570
ir-bond oxidation, 207, 270 Leukotriene B4 hydroxylation, 378, 436, 437, 534, 535,
on carcinogenicity of nitrosoamines, 421 621,631
in heteroatom dealkylation, 194, 265, 399, 417 Leukotrienes
in elimination reaction, 223, 272, 283 in physiology, 332
effect of reorientation in site, 64 metaboUsm, 294, 352, 378
on enzyme inactivation, 279, 283 L-Leucine, glucoside precursor, 561, 564
on hydroxylation, 186, 193,427 Lidocaine, 365, 426, 636
acetaldehyde, 421 Ligand, axial
alkyl,4,8,30,68, 164, 189 ammonia, 54
allylic, 4, 9 calculations on, 51, 52, 59, 61
benzylic, 64 catalase, 51
ethanol, 421 see Cysteine ligand
678 Index

Ligand, axial contd. Manganese porphyrin contd.


imidazole, 51 stereoselective oxidation by, 25, 26
protein environment effects, 52 Manihot esculenta (cassava), 557, 561
and spin state, 48, 157 Marmesin, 186,223
thiolate ligand, 48, 49 MDL 19347, 292
effect of structure on, 60, 61 Meander region, 95
effect on iron, 48, 52, 61, 183, 190, 248 Mechanism-based inhibitors, see Inhibition
thiolate ligand equivalents Medicago
methoxide, 62 5fl?/vfl (alfalfa), 555,571
methyl mercaptide, 49-51, 59-61, 69, 70 truncatula, 575
thiolate (SH-), 50, 60, 70 Medicarpin, 571
thiophenoxide, 51 Meloxicam, 428
water as 6th ligand, 48, 65, 192 Membrane
Ligand binding domain, 323 binding of P450 to, 92, 93, 442
Limonene, 9, 569 heme relative to, 93, 95
Linamarin, 564 insertion sequence, 92, 93
Lindane, 210 model phospholipid depth, 95, 129
Linear free energy relationships permeability to calcium, 542
inactivation by cyclopropylamines, 273 phospholipid monolayers, 95, 129
substrate oxidation, 427 targeting to membrane, 92
Linker domain, P450 reductase, 120, 121 targeting to in mitochondria, 92
Linoleic acid topology, 129
oxidation of, 187,409,423 Mentha sp., 569
as PPAR ligand, 332 Menthofuran, 261, 404, 621, 624
Linum usitatissium (flaxseed), 555 Menthol, formation of, 569
Linuron, 566 Mephenytoin, 378, 386, 406-408, 412, 621, 625, 626
Lipase, 117 19-Mercapto-androst-4-ene-3,17-dione, 289
Lipid peroxidation, 403, 422 10-P-Mercaptoestr-4-ene-3,17-dione, 290
Lipoxins,, 436, 531, 535 Metabolic intermediate complexes
Lipoxygenases, 531, 536 properties, 262, 263, 264, 265, 425
Liquiritigenin, 571 spectra, 645
Lithocolic acid, 327, 440 Metabolon, 563, 574
LKMl, see Antibodies Metallocorroles, 19
Loganin, 224, 571 Metalloenzymes, 150
Losartan, 409, 411 Metalloporphyrin, 62
Lotaustralin, 564 activation of dioxygen, 6-10, 155, 157, 159
Lotus japonicus, 571 amphiphilic, 24
Lovastatin, 425 chiral oxidations by, 25, 26
desaturation of, 210 hydroxylation, 17
LRH-1,451 see Manganese porphyrin
LTB4, see Leukotriene B4 olefin epoxidation, 17, 18-21, 26, 535
Lupinus sp., 671 radical cations, 17, 18, 207
Luteinizing hormone, 363 Methane, 11, 68, 69, 71, 72, 590
LXRa,440,461 Methane monooxygenase, 15, 164
LXRP,440,461 Methanosarcinia barkeri, 589
Lycopersicon esculentum (tomato), 555 Methemoglobin reduction, 134
Lysine acylation, 254 Methemoglobinemia, 134, 394, 400
Methimazole, 362
Macarpine, 572 Methionine N-hydroxylation, 555
Magnetic circular dichroism, 156 Methionine synthase, 134
Major histocompatibility locus, 453 reductase, 118, 120, 124, 125, 132
Malondialdehyde, 439 Methoxsalen, 250, 261, 386
Manganese porphyrin, 8, 19-22, 51, 62 2'Methoxyacetophenone, 405
as haloperoxidase, 22, 23 3-Methoxy-4-aminoazobenzene, 401, 436
oxygen exchange in reaction, 19,20, 22 Methoxychlor, 330
peroxynitrite reaction, 23, 24 18-Methoxycoronaridine, 412
Index 679

S-Methoxy-A'^TV-dimethyltryptamine, 416 Mitogen, 541


Methoxylamine, 261 Mitomycin c, 120
3-Methoxyphenethylamine, 416 MKP-1,449
4-Methoxyphenethylamine, 416 Molecular dynamics, see Computation
8-Methoxypsoralen, 250, 404, 621, 642 Monosodium glutamate, 353, 355
Methoxyresorufin, 633 Morpholine, 598
8-Methoxypsoralen, 263 Morphine biosynthesis, 463, 554, 572, 573
5-Methoxytryptamine, 416 Mortierella isabellina, 198
9-Methylanthracene, 207, 208 Mossbauer, 17, 156, 158,267
14a-Methyl-15-aza-D-homosterols, 291 computed parameters, 45, 54
N-Methylcoclaurine 3'-hydroxylation, 557, 571, 572 MSPPOH, 293, 294
3-Methylcholanthrene, 335, 336, 363, 398 Musca domestica, 222
Methylcyclohexane, 31 Mycobacterium
6-Methyleneandrosta-1,4-diene-3,17-dione, 289 avium, 591
4,4'-Methylene-bis(2-chloroaniline), 384, 428 CYPome of, 594, 598
Methylenedioxy carbene complex, 264, 645 bovis, 589, 592, 593
Methylene chloride, 420 CYPome of, 594, 598
Methylenedioxy compounds, 263, 571 chelonei, 592, 593
Methylenedioxy bridge, formation of, 571-573 fortuitum, 592, 593
14-Methylergosta-8,24(28)-dien-3,6-diol, 604, 608 leprae, 589, 592, 594, 596
14-Methylfecosterol, 604, 608 CYPome of, 594, 598
2-Methyl-l-heptene, 267 marinum, 591
3-Methylindole smegmatis, 589, 591-593, 596
desaturation of, 211, 422, 436 CYPome of, 594, 596, 598
P450 inactivation by, 280, 378 phylogenetic tree, 596, 597
4-Methyl-N-methyl-N-(2-phenyl-2H- tuberculosis, 125, 127, 135, 589, 590-593, 596, 609
pyrazol-3-yl)benzenesulfonamide (DMZ), 101 CYPome of, 594, 596
4-(Methylnitrosamino)-1 -(3-pyridyl)-1 -butanol, 3 84 phylogenetic tree, 596, 597
4-(Methylnitrosamino)-1 -(3-pyridyl)-1 -butanone, 378, Mycosphearella graminicola, 591
384, 403 Mycotoxin biosynthesis, 603
Methylococcus capsulatus, 588, 590, 591, 594 Myeloperoxidase, 1
fusion protein with ferredoxin, 592, 594 Myoglobin
4-Methylpentanal, 211, 212, 445 engineering of, 169
4-Methylpyrazole heme modification of, 281
as inhibitor, 421, 621, 627 hydroperoxo complex, 158, 159
0-Methylsterigmatocystin, 224 iron-aryl complex, 274, 275
N-Methylsulfonyl-12,12-dibromododec-11 -enamide, oxygen complexes, 156, 157
293 reaction with peroxides, 281, 154
N-Methylsulfonyl-6-(2- Myristic acid, 434
propargyloxyphenyl)hexanamide, 293, 294 Myzus nicotiana, 554
Methyl thieno[3.2.d][1.2.3]-thiadiazole-6-carboxylate, 283
Methyltrienolone, 351 NADP Sepharose chromatography, 227
Methylvinylmaleimide, 282 Naphthalene, 210,422
Methymycin, 600 dihydro-, 210
Metyrapone, 249, 250, 642 tetrahydro-, 33
Mice a-Naphthoflavone, 389, 398, 399, 402, 408, 409, 538
C57B1/6, 335 as allosteric effector, 427, 428
129SvJ, 538 as inhibitor, 428
Transgenic, 395, 416, 433, 435, 440, 455 2-Naphthylamine, 384
see Null mice 2-Naphthylhydrazine, 274
Miconazole, 250, 290, 291, 293, 592, 593 Narbomycin, 600
Microperoxidase, 154, 160 Naringenin,408,430, 571
Microsporum spp., 603 NcoR, 324,424
Midazolam, 386, 425, 426, 622, 629 Nectria haematococca, 603
cooperativity in metabolism of, 429 Nematode, sterol from diet, 590, 592
Mifepristone, 250, 259, 271, 622 Neomethycin, 600
680 Index

NF-IC, 449 NMR contd.


Neopinone, 573 i^F, 203
Neurospora crassa, 589, 591, 602, 603 ligand binding, 25, 27, 64
NF-1,328 of metalloporphyrins, 17, 157, 267
N F K B , 442 of Pd/PdX complex, 136
Nicotiana tabacum, 554 Nomenclature, See P450
Nicotine NonO, 449
binding to P450 Norbomane, 3, 4, 8
oxidation of, 396, 403 exo-tetradeuterio-, 4
Nifedipine, 134, 386, 423,425, 426, 621, 628, 637 Norbornene
NIH shift, 75, 202, 204 epoxidation, 17,26,269
Nikkomycin, 599 metalloporphyrin adduct, 269
Nitrene, 267 Norcarane, 11, 12,15,188
Nitric oxide, 2 Norepinephrine signaling, 538
as ligand, 51 Nomitrosonicotine, 384
P450 inhibition by, 248 Novobiocin, 599, 600
as product, 16, 571 NRl, 118, 120, 124, 125
Nitric oxide reductase , see CYP55A1 Nuclear receptors, 323, 325
Nitric oxide synthase, 1, 2 enhancer, 328
alternative electron acceptors, 120 phosphorylation, 324
and calmodulin, 16, 118, 120 Null mice, 380, 395, 396
domains, 118 acetyl CoA, 332, 334
structure, 16, 17,89 AHR, 338
mechanism, 16, 127, 150, 159 CAR, 330, 331
reactions, 16, 124 CYP1B1,395,396,400,401
redox properties, 125, 132 CYP2E1,395,419,420,422
spectroscopic properties, 156 cyp4al4, 543, 544
and tetrahydrobiopterin, 16 CYP7A1,439,440
Nitrite, 21 CYP11A1,446
/7-Nitroanisole, 194, 640 CYP19A 1,452
6-Nitrobenzo[a]pyrene, 401 CYP27A1,458
4-Nitrocatechol, 420 CYP27B 1,460
6-Nitrochrysene, 401 P450 reductase, 119, 120. 605
2-Nitrofluoranthene, 401 PPAR, 334, 335, 531, 539
3-Nitrofluoranthene, 401 PXR, 326, 327
Nitrogen oxidation Nystatin, 598, 599
N-dealkylation, 193, 194
methyl vs ethyl, 195 Obtusifoliol 14a-demethylation, 555, 558, 590
electron transfer in, 193 17-Octadecynoic acid, 294, 621, 630, 637
N-oxidation, 194 1-Octene, 33,268
/7-Nitrophenol, 420, 621 /ra/25-[l-2H]-, 198,268
1-Nitropyrene, 401 «-Octylamine, 646
2-Nitropyrene, 401 0-dealkylation, 193, 194
Nitroso complex, 266 17-ODYA, see 17-octadecynoic acid
N-Nitrosobenzylmethylamine, 403 Okadaic acid, 329
N-Nitrosobutylamine, 403 Olanexidine, 214
N-Nitrosodimethylamine, 403, 621, 627, 636 Olanzapine, 399
N-Nitrosomethylphenylamine, 405 Oleandomycin, 599
N-Nitrosophenylmethylamine, 403 Oleic acid, 198
N-Nitrosodipropylamine, 403 Olefin oxidation, see Epoxidation
N-Nitrosodiethylamine, 403, 405, 420 Oligomycin biosynthesis, 601
N-Nitrosonornicotine, 403 Oltipraz, 254
Nitrous oxide, 30 Omeprazole, 386, 394, 397, 399, 412, 621
NMR Ondansetron, 399
of adrenodoxin, 136 Orbital
ofcytochromeP450 reductase, 120, 122 a2„, 50, 59, 62, 66, 77, 189
Index 681

Orbital contd. P450 contd.


d, 48, 50 ordered solvent, 89
0RCA3, 569, 574 water content, 65
Org-30365, 289 branchpoints in catalysis, 156
Org-30958, 289 see Catalytic cycle
Organic anion transporter, 327 conformational dynamics, 102, 106, 428, 429, 435
Oryza sativa, 554 cooperativity in, 394, 427-^29
Orphenadine, 406 distance from iron of oxidized atom, 100
Oryza sativa, 591 entry channel, 47, 65, 105, 108
Osmaronin, 565 expression of isoforms, 336, 392
Osmotic stress, 165, 586, 591 heme binding to protein, 435, 436
Ostrinia nubilatis, 568 human
Ovariectomy, 351 identities, 378, 379
Oxidase, 151-153, 156, 158 classification, 380
Oxidation potential, see Redox potentials diseases, mutation based, 383
Oxidative stress, 422 inducers, major, 387
N-Oxides, see Oxygen donors liver content, 380-382, 385
reduction of, 120 relative importance of isoforms, 383, 386
2,3-Oxidosqualene cyclase, 590, 592 hydrogen bond network, 57, 63, 152, 164
Oximes, dehydration to nitriles, 228, 564 isomerase of hydroperoxides, 5
(19R)-10-oxiranylestr-4-ene-3,17-dione, 290 levels of, 381
7-Oxocholesterol, 439, 441 sex differences, 347
3-Oxodecalin-4-ene-10-carboxaldehyde, 222 mitochondrial, 379, 380, 419, 443, 444, 454, 456
7-Oxo-24,25-dihydrolanosterol, 291 nomenclature, 414, 554, 555, 586
2-Oxoglutarate dioxygenase, 567, 568 phylogenetic trees, 555. 591
Oxonium ion, 265 proteosomal degradation, 282
Oxygen, activation of proton delivery, 63, 152, 154, 155, 161, 162, 165,
mechanism of, 149, 150 167, 169
oxygen-oxygen bond cleavage, 58, 59, 150, 154, 170 see Cysteine ligand
homo-vs heterolytic, 151, 154, 155, 160, 439, 533 quantitation, 644
Oxygen donors rate limiting steps, 154, 427
acyl peroxide, 3, 23, 161 reduction potentials, 54, 134
alkyl peroxides, 3, 154, 185, 254, 263 resting state, calculations on, 51
unsuitability of, 221 RZ values, 645
hydrogen peroxide, 3, 5, 152, 221, 222 spectroscopic maxima, 644
hypochlorite, 23, 28 stoichiometry, P450 catalysis, 184
iodosobenzene, 3, 18, 28, 185, 194, 207, 221, 254, structure fold, overall, 87
439 see Substrate access channel
reversibility of reaction, 17 topography, control of reaction, 73
N-oxides, 30-33, 197 P450Bi„j (CYP107H1), 135, 212, 213, 589
Oxone®, 32 P450^^ (CYPlOl), 585, 586, 595
periodate, 3, 32 active site, 46, 64
persulfide, 23 alternate oxygen donors, 3
product differences vs native system, 416 Asp251, 48, 63, 152, 164, 165, 167
Oxysterols, 458 Ala252, 163
Ozone layer, 30 autooxidation, 183
C357H, 183
P420 computational studies of mechanism, 69-71
formation, 285 D251N, 158
ligand in, 265 entry channel, 65, 136
quantitation, 645, 646 See Ferrous dioxygen complex
spectrum, 645 ferryl species, 3, 46, 59, 60, 167
P450 fusion protein of, 138
active site Gin 360, 48, 60, 61
conserved residues, 598, 601 Gly248, 162
electrostatic potential, 89 Gly359, 48, 60
682 Index

P450cam (CYPlOl) contd. P450eryF (CYP107) contd.


hydrogen bonding network, 57, 90, 152, 162, 167 computational studies, 55, 65
See Ferric hydroperoxo distal hydrogen donor, 89, 90
I-helix, 90 reaction catalyzed, 103
iron-aryl complex, 274 structure, 87-89, 101,428
Leu358, 48, 60 substrate-assisted mechanism, 90, 91, 163
ligand binding, 108, 109, 167 P450^^^(CYP55A1), 587
oxy-complex, 55, 89, 160, 167 ligand complexes, 169
oxygen activation, 55, 56 NADH binding to, 105
potassium binding site, 168 proton delivery, 169
protein radical of, 3 reduction of NO, 105, 169, 603
proton delivery, 48, 57, 152, 162, 165-169 structure of, 169, 183
putidaredoxin interaction with, 100, 135, 136 P450o,y3
rate limiting step, 136 vancomycin biosynthesis, 105
reaction catalyzed, 65, 103 X-ray structure, 206
spin state, 155 P450p^,^,599
solvent isotope effect, 56 P450j^P, 116
substrate binding, 63, 64, 155 P450^^^,5eeCYPllAl
structure, 87-89, 101, 104, 108-110 P450^oy(SoyC), 135
Thr252, 6, 48, 55, 57, 152, 162-168, 185 P450^^^p(CYP108),598
Tyr96, 64, 65, 167, 168 structure, 183
see Uncoupling substrate, 595
water content of active site, 65, 71 P450™c. 599
P450BJ^.3 (CYP102), 585-587, 594, 601 Paclitaxel 6a-hydroxylation, 621, 625
active site, 537 Palmitic acid, 332, 434
aldehyde oxidation, 222, 283 Palmitoleic acid, 100, 105
domains, 118,596 Papaver somniferum, 554
electron transfer, internal, 132, 133 Paraciticein, 569
hydride transfer, 132 Parathion,250,251,395
reaction catalyzed, 11, 103 Parathyroid hormone, 454, 459, 538
redox properties, 125, 133 Parkinson's disease, 418
reductase domain interaction, 95, 96, 98, 115, 120, Paroxetine, 264, 265, 621, 627
130 Paxillin biosynthesis, 603
regulation of, 131 Pbx 1,449
salt bridges in, 92 PAR, see Pregnane X receptor
structure, 87, 88, 106 PAS superfamily, 323, 324, 336
substrate PBREM, 405, 407
access channel, 65, 537 Penicillin biosynthesis, 585, 586, 587
specificity, 537, 540, 595, 596 Penicillium
structure with, 100, 101, 105, 109, 131 chrysogenum, 586, 587
turnover number, 131 digitanum, 607
P450BSP italicum, 591
peroxide dependence, 99 paxilli, 603
fatty acid oxidation, 99 Pentafluorochlorobenzene, 203
structure, 99, 102 Pentalenolacton biosynthesis, 601
P450^^JCYP176A1) Pentobarbital metabohsm, 119
redox partners, 588 7-Pentoxyresorufin, 422, 634
cineol metabolism, 595 Perhexihne, 394
P450j^jj^, See Nitric oxide reductase Peroxidase
P450EP„K axial ligand, 52, 89
function, 99 catalytic cycle, 151
reaction catalyzed, 103, 595 Compound I, 17, 150, 154
structure, 99, 100, 101, 104, 106 dioxygen bond cleavage, 154
P450eryF(CYP107) distal histidine, 154
binding two substrates, 428 electronic state, 52
cooperativity, 428 engineering of, 169
Index 683

Peroxidase contd. Phenylacetaldehyde, 28, 200


pKaofFe-OOH, 154 Phenylacetaldoxime, 555
reduction potentials, 89 Phenylacetate
spin state, 52 growth on, 586
Peroxide shunt, 153, 156, 160, 184 2-hydroxylation, 586
Peroxisome proliferator activated receptor (PPAR), 323, Phenylacetylene, 257, 263, 269, 270, 271
331,545 Phenylalanine
binding protein, 333 in biosynthesis, 561, 569
coregulators, 333, 335 N-hydroxylation of, 555
interaction with genes, 119 1 -Phenyl-1 -butanone, 200
ligands for, 332, 533 Phenylbutazone, 442
null mice, 334, 335 ^ra«5-l-Phenylbutene, 200
PPARa, 119,332,440,539 4-Phenyl-l-butyne, 271
PPARP, 332 Phenylcyclohexylamine, 260
PPAR7, 332,451 1 -(1 -Phenylcyclohexyl)-2,3-dihydro-4-pyridone, 259
species difference, 334 Phenylethyldiazene, 277
structure, 333 Phenethylisothiocyanate, 404
Peroxisome proliferation, 331, 332, 396, 434, 621 6-Phenyl-l-hexyne, 271
hormonal effects on, 354 6-Phenyl-2-hexyne, 271
regulation of P450 reductase, 119 Phenylhydrazine, 273, 274
Peroxygenase N-Phenylhydrazone, 273
activated species, 150 Phenylimidazole
catal)^ic cycle, 151 P450 inhibition by, 249
Peroxynitrite, 23, 24, 442 structure of P450 complex, 100, 104
Persea americana (avocado), 556, 566 Phenylketene, 257
Petroselium crispum (parsley), 555 5-Phenyl-l-pentyne, 271
PGE2, 437, 534 2-Phenylphenanthridinone, 250
PGD2, 427 3-Phenylpropionaldehyde, 282, 283
PGE 1,427 1-Phenyl-1-propyne, 271
PGH synthase, see Prostaglandin H synthase Phenylpropanoid pathway, 570, 574
PGG2, 439 Phenytoin, 252, 386, 409, 443, 621
PGHj, 439 Phlobaphenes, 570
PGH2,441,442,531 Phorbol esters, 438, 440
P450 role in isomerization, 532 Phosphatidylcholine, 117, 541
hydroxylation of, 434, 534 Phosphatidylethanolamine, 541
PGJ2, 452 Phosphatidylinositol, 541
PGI2, 533 Phospholipases, 402, 535
Phanerochaete chrysosporium, 589, 591, 598, 602, 610 Photoacoustic calorimetry, 165
Pharmacogenetics, 387 Photochemical reduction, 157
Pharmacogenomics, 387 Photosystem I, 135
Pharmacokinetics, 347, 387, 393, 394, 409, 419, 424 Phthalate dioxygenase reductase, 138, 589
Pharmacophore models, 397, 399, 414-416, 455 Phylogenetic relationships, 555, 591
Phaseolus aureus (mung bean), 555 Phyllotreta nemorum, 562
Phenacetin, 204, 205, 394, 398^00, 621, 624, 633, 646 Phytoalexins, 553, 566, 568, 571, 603
Phencyclidine, 259, 260 Phytoanticipins, 553, 560
Phenelzine, 273, 274 Phytoattractants, 553
Phenformin, 418 Phytohormones, 554, 556, 558, 569, 574
Phenobarbital Piceatannol, 402
in arachidonate epoxidation, 539 Picia anomola, 591
as inducer, 402, 408, 412, 539, 621 Pikromycin, 599, 600
hormonal effects on, 354 Pimelic acid, 213
induction of P450 reductase, 119 Pinoline, 416
negative regulation of, 363 Piperonyl butoxide, 278, 642
receptor, 325 Pisatin, 603
responsive enhancer module, 328 Pisum sativum (snow pea), 571
Phenol, substituted, 185, 204 Pituitary, 350, 351
684 Index

Plasmalogens, 133 Pregnenolone contd.


Pneumocystis carinii, 603 oxidation of, mechanism, 216, 217
Polychlorinated biphenyls, 336, 397, 399 ^^O studies in oxidation of, 215
Polycistric ovary syndrome, 450 Pregnenolone
Polycyclic aromatic hydrocarbons 16a-carbonitrile, 324-327
binding to DNA, 207 6,16a-dimethyl, 325
fungal hydroxylation of, 602 Premarin®, 402
induction of P450, 335 Proanthocyanidins, 570
ionization potentials, 207 Prodrugs, 394
negative regulation of P450, 363 Progesterone, 415
oxidation of, 384,401 binding to CYP2C5, 108
Polyglandular syndrome, 446 and CAR activity, 329
Polymorphisms, human, 383, 387 21,21-dichloro-,255
see individual P450 isoforms 17a-hydroxy-, 449
influence on drug development, 389, 392, 394, 413 17-hydroxylation, 449
interindividual variation, 392 2a-hydroxylation, 640
single nucleotide (SNP), 387, 395, 402, 406, 409, 6p-hydroxylation, 637, 640
419, 423, 424, 433, 435, 442, 449, 461 15a-hydroxylation, 640
toxicities due to, 394, 396 16a-hydroxylation, 635, 640
ultrarapid metabolizers, 387, 414, 418 21-hydroxylation, 413, 453, 634, 637, 640
Polymorphonuclear leukocytes, 535 e«r-Progesterone, 450, 453
Popupus tremuloides (querken aspen), 555 9'-Propargyl abscisic acid, 259
Porphyria, 278 Propargylamine
Porphyrin N-(2-heptyl)-, 259
binaphthyl, 9, 10,25,26 7V-(2-heptyl)-A^-methyl-, 259
biosynthesis, 278 Propene
see Metalloporphyrin computational studies of oxidation, 68, 70, 71, 74,
octamethyl, 62 77-79
"picnic-basket", 25, 26 in heme N-alkylation, 74, 268
polyfluorinated, 28, 29 Propentdyopents, 282
see Protoporphyrin IX Propionaldehyde, 221
saddling, 62 2-phenyl-, 221
me50-tetraaryl, 17, 18, 20, 29, 195 Propofol, 406
me^o-tetramethyl, 62 7-Propoxycoumarin, 422
"twin-coronet", 26, 27 2-«-Propyl-4-pentenoic acid, 208
Posaconazole, 605, 607 2-n-Propyl-2(£)-pentenoic acid, 209
Potential energy surfaces, 47 4-(l-Propynyl)biphenyl, 271
Potentiometry, 127, 128 10-(2-Propynyl)estr-4-ene-3,17-dione, 290
PPAR, See Peroxisome proliferator activated receptor 2-(l-Propynyl)naphthalene, 271
^^P phosphate source, 157 2-(l-Propynyl)phenanthrene, 271, 398
Prandiol, 223 3-(l-Propynyl)phenanthrene, 271
Pregnancy, P450 induction in, 534 9-Propynylphenanthrene, 271
Pregnane X receptor (PXR), 323, 325 l-(l'-Propynyl)pyrene, 271, 398
in CYP3A induction, 324, 424, 425, 431, 433 Prostacyclin, 438, 441, 531
in CYP2B induction, 330, 405, 406 Prostacyclin synthase, see CYP8A1
in CYP2C induction, 408 Prostaglandin H synthase, 531
in CYP7A inducction, 440 EET oxidation, 533
null mice, 326, 327 model of, 26, 27
Pregn-4,20-diene-3-one, 634 N-dealkylation by, 195
5p-Pregnane-3,20-dione, CAR ligand, 329 reaction catalyzed, 540
Pregnenolone Prostaglandins
21-chloro-,255 biosynthesis, 1, 119
21,21-dichloro-,255 in inflammation, 332
17a-hydroxy-, 449 metabolism of, 294, 378, 437
hydroxylation, 441, 449 Prostanoids, metabolism of, 532, 533
Index 685

Protein kinase A, 445, 449, 453 Radical contd.


Protein kinase C, 425, 453 lifetime, 9, 68, 187, 188
Proteosomal degradation, 282 probes, 9, 11,68-70
Proton inventory, 165 properties of in transition state, 72
Protoporphyrin IX rearrangement, 11, 15, 189
meso-Si\ky\-, 283 recombination, 5, 11, 12, 188
N-alkyl-, 278 spin trapping of, 194, 272, 273
N-(2-hydroxyalkyl)-, 277 trimethylsilyl, 286
N-(2-phenylethenyl)-, 277 tyrosine, 185, 186
N-(2-phenylethyl)-, 277 Radical cation
N-vinyl-, 277 aromatic ring, 203, 207
4-(l-Propynyl)biphenyl, 199 porphyrin, 2, 3, 6, 11, 67, 153
Pseudohermaphroditism, 450 protein, 3
Pseudomona aeruginosa, 589 sulfur, 253
Pseudomonasputida, 3, 15, 87, 134, 585, 595 Radical clock, 9, 11, 68-70, 186-188
Psoralen, 187,223 effects of metals on, 14
Psoriasis, 460 Radiolysis,5, 157, 158, 167, 191
Puberty, 348, 349 Raloxifene, 253, 254
Putidaredoxin, 134, 595 Ralston metallidurans, 589
fusion protein of, 138 Random mutagenesis, 399
interaction with P450^^j^, 135, 136 Rapamycin, 599
interaction with putidaredoxin reductase, 98, 116 Rat
mechanism, 136 Dahl Salt Resistant (DR), 543
structure of, 97, 99, 135 Dahl Salt Sensitive (DS), 543
Putidaredoxin reductase, 134, 595 dwarf, 355
dithiol/disulfide oxidoreductase activity, 138 Fischer 344, 363
fusion protein of, 137 Nude, 459
structure of, 97, 99, 116 Spontaneously Hypertensive (SHR), 543
Pyrene, 428 Sprague-Dawley, 363, 544
Pyridine, 249 Ravuconazole, 605, 607
1 -methyl-4-phenyl-1,2,3,4-tetrahydro-, 196 Reaction barriers, 47
4-aryl(alkyl)-l,4-dihydro-, 211 Rearrangements, oxidative
4-phenyl-^ra«5-1 -(2-phenylcyclopropyl)-1,2,3,6- see Acetylenes
tetrahydro-, 197 see Aryl migration
17-(5-pyrimidyl)androsta-5,16-dien-3P-ol, 292 see Epoxidation
17-(3-pyridyl)androsta-5,16-dien-3(3-ol, 292 see NIH shift
PXR, See Pregnane receptor see Radical clock
Receptor
Quadricyclane, 192 see Aryl hydrocarbon receptor
Quercitin, 408 see Androstane receptor
Quinidine see BRII receptor
as allosteric effector, 428, 429 see Famesoid receptor
as inhibitor, 254, 386, 389, 394, 417, 621, 626 see Glucocorticoid receptor
as substrate, 423 see Growth hormone receptor
Quinine, 426 see Nuclear receptors
Quinone methide, 253 see Peroxisome proliferator activated receptor
see Phenobarbital receptor
R-76713,250,287,288 see Pregnane X receptor
Radiation, 556 see Retinoid X receptor
Radical see Vitamin D receptor
alkoxy, 70,215,217 Redox potentials
alkyl, 11, 12, 14, 189, 265, 272, 273, 275, 277, 280 P450 control of, 89, 131, 132
aryloxy, 26, 206, 207 Reductase partners, classification, 587-589
rer^butoxy, 8, 70, 189, 196 Regulation
caged substrate radical, 8, 9, 14 see Diet, effects on P450
686 Index

Regulation contd. Sequence


downregulation by cytokines, 388 homology model difficulties, 109
endocrine, 347, 541 structure models
see Fasting CYP1A1,397
see Induction CYP1A2, 399
Renex 690 CYP1B1,402
Repeats, 323, 324, 336 CYP2A6, 404
Resonance Raman, 27, 156, 167 CYP2B6, 406
Response elements, 324, 326, 328, 337, 338, 397 CYP2C9,410
Resveratrol, 283, 402 CYP2C18,411
Reticuline, 205, 206, 571-573 CYP2C19,413
Retinoic acid, 119 CYP2D6,416
4-hydroxylation, 349, 379, 407, 409, 433, 455, 456 CYP2E 1,420
in P450 regulation, 440 CYP3A4, 427
Retinoid X receptor (RXR), 324, 328, 451, 454 CYP4A11,434
Rheumatoid arthritis, 450 CYP11B1,434
Rhizopus niger, 586, 587 CYP11B2,448
Rhodococcus sp., 588, 595, 596 CYP17A 1,450
Rhodotorula minuta, 226 CYP19A 1,452
Rhophalosiphum maydis, 568 CYP21A2,453
Rhapontigenin, 283, 284, 402, 632 SET mechanism, 195, 196, 226
Rickets, 459, 460 Sex hormone deficiencies, 383
Rifampicin, 386, 402, 407, 408, 412, 424, 433 Sex-linked differences
Rifampin, 621 androgenic imprinting, 350
Ritonavir, 254, 415 in eicosanoid metabolism, 536
Rosiglitazone, 407 and growth hormone, 350-352
RU486, 325 in hepatic metabolism, 348
Ruthenium P450 levels in rat, 347, 353, 388, 537, 543, 544
P450gj^.3 modification, 96 specific forms, 348
porphyrin, 8, 12, 26, 28-33, 62, 73 Shear stress response element, 442
RZ values, 645 Sildenafil, 425
Simvastatin, desaturation of, 210
Saccharomyces cerevisiae, 585 Sinapoyl malate, 556
azole binding to, 605 Sinorhizobium meliloti, 589
in azole resistance, 608, 603, 605 SKF525-A, 278, 643
14a-demethylase, 558 Smoking, 395-397, 399, 400, 403, 404, 418
A^2-desaturation in, 210 SMRT, 324, 330, 333, 424
P450 enzymes, 589, 591, 601 Na+-K+-ATPase, 536, 538
P450 reduction in, 120, 605. 606 SOCS proteins, 361
Saccharum sp. (sugar cane) 560 Sodium transport, 541, 543
Salen metal complexes, 19 Solarium lycopersicon (potato), 560
Salt, 541, 543 Solvents
Salt-inducible kinase (SIK), 445 see Ethanol
Salt wasting syndrome, 453 induction by, 419
Salutaridine, 205, 206, 572, 573 inhibition of P450 activity, 389, 409
Saperconazole, 250 Soret maximum
SCH66712, 283, 284, 417, 621, 627 alkyl or aryl-iron complex, 273, 275
Schizosacchawmyces pombe, 447, 589, 591, 601 carbene-iron complex, 264, 285
Scoulerine, 571,572 fifth iron ligand effect, 158, 248
Screening, high throughput, 133, 426 MI complex, 265
Secobarbital, 250, 256, 263, 267, 285, 634 red shift, 157
Secologanin, 224, 568, 569 reverse Type I, 248, 646
Sedormid, 256 split, 54, 156
Seminal vesicles, 534 Type I, 646
Senecionine, 384 Type II, 248, 249, 291, 292, 605, 646
Senescence, 349 Sorghum bicolor, 557, 558, 561, 573, 591
Index 687

SoyB, 135 Streptomyces


Spl, 433, 442, 445, 449, 459, 461 avermitilis, 589, 596, 598
Sp3,433, 445, 499 CYPome, 600, 601
Sparteine, 383, 386,414 genome, 599
Spin densities, 61 coelicolor, 99, 589, 590, 594, 596, 600, 601
Spin echo envelope modulation (ESEEM), 51 CYP51 knockout of, 592
Spin state genome, 599
computation of, 47, 50, 51, 80 griseus, 135, 599
doublet, 51, 189,201 Streptomycete P450s, 599
quartet, 11, 12, 21, 49, 189, 190, 201 Strictosidine, 569
high, 48, 136 Styrene epoxidation, 185, 384, 420, 422
ligand effect on, 51, 61, 131 p-chloro-, 25, 28
low, 48, 136 electronic effects on, 28
relation to electron transfer, 131, 248 heme alkylation during, 267
sextet, 11,51 isotope effects on, 198
spectra associated with, 644 /?-methoxy-, 28
spin-orbit coupling, 9 /7-methyl-, 198
substrate binding effect on, 3 cw-|3-methyl, 25, 26
Spiro[2,4]octane, 188 /7-nitro-, 28
Spiro[2,5]octane, 12 /7-phenyl, 198
Spiromethylsulfone, 415 rearrangement during, 30, 200
Spironolactone, 253, 280, 281, 453 stereochemistry of, 27, 33, 65
7a-thio-, 450 Substrate
Spirosulfonamide, 415 access channel, 47, 65, 102, 105, 108, 410, 537, 609
Spontaneously hypertensive rat, 543 exit channel, 47
Squalene, 1,590,592 Substrate binding, 183
SR12813, 325 channeling, in plant pathways, 563
SRC-1,324,330,333 contacting residues, 63
SREBP,461 control of redox/spin states, 3
Starvation, 331,537 determinants of, 63
Stat-3,451 location, 47
Stat-5a, 360 mobility in relation to, 5, 64
Stat-5b, 356-361, 364 of multiple substrates, 428
Sterigmatocystin, 211, 384 spectroscopic changes upon, 556, 646
Steroid water expulsion by, 71, 72
22-desaturation, 586, 592, 601-604 Substrates, functional markers, 621
6p-hydroxylation, 349, 555 Sulfaphenazole, 386, 407^10, 621, 626, 635
7a-hydroxylation, 349 Sulfenic acid, 253
I la-hydroxylation, 586, 587 Sulfinic acid, 253
II (3-hydroxylation, 586 Sulfite reductase, 96, 118
12-hydroxylase, 443 Sulfolobus
17a-hydroxylation, 379 solfataricus, 91, 100, 596, 601
23-hydroxylation, 555 tokodaii, 589
27-hydroxylation, 379 Sulfonic acid, 253
C4-methyl oxidase, 605 Sulforaphane, 254
5a-reductase, 349, 351, 364 Sulfur oxidation
Steroidogenesis pathway, 444 S-dealkylation, 193
Steroidogenic factor 1 (SFl), 445^47, 449, S-oxidation, 76, 197
415,453 sulfur radical cation, 77, 197, 198
Sterol 11(3-/18-hydroxylase, see CYPl IB Superoxide
Sterol 17a-hydroxylase/C 17-20 lyase, see CYPl7 dismutase, 197,258,282
Sterol 21-hydroxylase, see CYP21 reaction with P450, 157
Stigmasterol, 592 uncoupling product, 134
Stilbene, 33, 198, 570 Supersomes®, 252
St. John's wort, 394, 430 Surfactant, 285
Stopped flow studies, 20, 124, 126, 127, 132, 157 Sus scrofa, 591
688 Index

Sutherlandin, 565 Thermus thermophilus, 92, 596, 601


10-SUYS,293,294 Theophylline, 386, 398-400, 633
SXR, see Pregnane X receptor (205)-22-nor-22-Thiacholesterol,286
Sydnones, 275, 277 10-Thiiranylestr-4-ene-3,17-dione, 290
3-(2-phenylethyl)-4-methyl-, 277, 278 Thioester hydrolysis, 253
3-(2-phenylthioethyl)-4-methyl-, 278 Thiophene, 250,252,410
sulfoxide, 251
Tabersonine, 568, 569 Thiosteroids, 254
Tacrine 386, 399 2|3-Thiotestosterone, 637
Tamoxifen, 250, 262, 263, 351, 425, 452 6(3-Thiotestosterone, 636
TAO, see Troleandomycin Thioureas, 250
Tapesia Thlaspi arvense, 566
acuformis, 591 Threonine, conserved, 183, 185, 186, 192, 217
yallundae, 591 Thromboxane, 437-^39, 531
Taxiphyllin, 564 Thromboxane synthase, see CYP5A1
Taxol, 99, 378, 407, 408, 621, 625 Thujone, 210
TCDD, 336 Thyroid hormone
as inducer, 335, 397, 538 in regulation of P450, 349, 362, 365, 440
as inhibitor, 399 in regulation of P450 reductase, 119, 365
in lowering P450 levels, 363 Thyroxine, 363
toxicity, 338 Ticlopidine, 252, 621, 625
Teasterone, 559, 560 Tienilic acid, 250, 251, 252, 410, 411, 621, 625
Tebuconazole, 593 Tirapazamine, 120
Tegafur, 403 Tolbutamide, 378, 386, 407-409, 411, 412, 621, 625
Temperature jump, 127 Toluene hydroxylation, 247
Terconazole, 250 20-(p-Tolyl)-5-pregnen-3p-ol, 212, 213
Terfenadine, 394, 426, 430 Tracazolate, 211
Terpineol, 595, 598 Transcription factors, 323
Testarone, 558 Transcriptome, 605
6-deoxy-, 558 Transgenic
Testosterone mice, 395, 416, 433, 435, 440, 455
dehydrogenation of, 210 plants, 564
depletion by xenobiotics, 363 Transporters, 606
hormonal effects on P450, 350-351, 536 Cdrlp, 607, 608
2-hydroxy-, 635 Cdr2p, 607
6-hydroxy-, 210, 349, 378, 380, 425-428, MDR1,607,608
433, 434 role in drug removal, 392
7-hydroxy-, 210, 349 Tranylcypromine, 404, 621, 624
15-hydroxy, 349 TreP-132,445
16-hydroxy-,349, 635 Tretinoin, 622
17-hydroxylation, 413 Triacetyloleandomycin, see Troleandomycin
in P450 regulation, 364, 449 Triazolam, 426
as substrate, 134, 164, 415, 452, 621, 622, 627, Triazole, in P450 inhibition 249
633-636, 640 Triethylenethiophosphoramide, 406
3,4,5,6-Tetrachlorocyclohexene, 186 Trifoliumpratense (red clover), 571
2,3,78-Tetrachlorodibenzo-p-dioxin, see TCDD Trifolium repens (white clover), 571
Tetracycline, 598 Trichloroacetaldehyde, 200
12-O-Tetradecanoylphorbol 13-acetate, 438, 502 Trichloroacetic acid, 332
Tetrahydrocannabinol, 261 1,1,1 -Trichloroethane, 420
2,3',4,5'-Tetramethoxystilbene, 285, 402, 633 Trichloroethylene, 200, 384, 420
Tetramethylpiperidine cleavage, 228, 229 Trichophyton reubrum, 603
TGF-Pj,451 Trichostatin A, 454
Thalidomide, 412 Triethylenethiophosphoramide, 621
Thaxtomin, 599, 600 Triglochin maritima, 557
Thebaine, 573 2,4,7-Trihydroxy-2H-l ,4-benzoxazin-3(4H)-one, 567
Thermal factors, in crystal structures, 65 Trimethoprim, 621, 625
ThermophiHc P450, 91 Trimethylamine, 195
Index

Trioxsalen, 261 Vancomycin biosynthesis, 206


Triticum aestivum (wheat), 555, 558, 564, 566, 567, 591 Vasoconstriction, 542
Troglitazone, 253, 254, 407 Vasodilation, 541, 542
Troleandomycin Vasopressin, 538
as inducer, 282 VDRE-1,454
as inhibitor, 265, 266, 282, 285, 425, 430, 433, 621, VDRE-2, 454
628, 637 Venturia nashicola, 591
modification of P450 compressibility, 429 Verapamil, 407
Try-P-1 (3-amino-1,4-dimethyl-5H-pyrido(4,5-Z>)indole, Vicia villosa, 571
365 Vigna radiata (mung bean), 560
Trypanosoma brucei, 591, 610 Vinblastin biosynthesis, 568, 569
Tryptamine, 404, 416, 568 Vincristine biosynthesis, 568, 569
Tryptophan as substrate, 555, 569, 574 Vindoline, 569
Tumor necrosis factor a (TNF), 419, 440, 451 Vinylbromide, 384, 420
Tumor promotion, 407 Vinyl carbamate, 384, 420
Type I spectrum, 646 Vinyl chloride, 384, 420, 422
Type I, reverse, spectrum, 248, 646 Vinyl fluoride, 268
Type 11 spectrum, 248, 249, 291, 292, 605, 646 (325)-Vinyllanost-8-en-3p,32-diol, 291
Tylosin, 599 18-Vinylprogesterone, 447
Typhasterol, 559, 560 Virilization, P450 link to, 383,454
Tyrosine Vitamin A deficiency, 364
/>-hydroxy-, 562 Vitamin D, 440
iV-hydroxy-,561,562 deficiency, 455
to /7-hydroxyphenylacetaldoxime, 557 la-hydroxylation, 379, 459
kinase, 356-358 24-hydroxylation, 379
oligomerization by CYP57, 601 25-hydroxylation, 456, 457, 458
as precursor, 561, 564, 572 hypervitaminosis, P450 link to, 383
radical, 185, 186 manifold, overview of, 454
Two-state reactivity (TSR), 11, 66, 69, 73, 80, 188, 190, metabolism, 454
193,201,269 production in yeast, 586
receptor, 406, 425, 451, 454
Vitis vinifera, 574
Ubiquitin,282,419,425 VLDL, 541
10-UDYA, See 10-Undecynoic acid Voriconazole, 605, 606, 607
UGT1A6, 338
UGT85B 1,562-564 Warfarin
Uncoupling, 156 dehydrogenation of, 210
by hydroperoxide dissociation, 5, 58, 161 as substrate, 386, 409, 410-412, 621, 632, 635, 640
cytochrome b5, effect of, 134 cooperativity in metabolism of, 429
oxidase pathway, 152, 156 polymorphism effect, 394, 410
phenomenon of, 152, 184 toxicity, 411
relation to structure, 154 Wound-induced response in plants, 553, 568, 569
and water access, 152, 163, 184 Wy 14643, in peroxisome proliferation, 332,434, 621
10-Undecynoic acid, 250, 256, 257, 263, 293, 621,
630, 637 XAP2, 336
10-Undecynyl sulfate, 293 XRE element, 397
USF 1,433 XREM, 433
Ustilago maydis, 603 Xylenes, 64, 202
XylM, 15, 16
Valeraldehyde, 221
Valeric acid, 226 Yarrowia lipolytica, 602
L-Valine, glucoside precursor, 561, 564 Yellow flourescent protein, 564
Vancomycin, 105 YYl transcription factor, 454
VanderWaals,46, 135
Lennard-Jones potential, 47 Zafirlukast, 280
Valproic acid desaturation, 208, 209, 280, 436 Zea mays (corn), 566, 567
3- and 4-hydroxy-, 209, 436 Zinc finger motif, 323

You might also like