You are on page 1of 410

Gout

David S. Newcombe

Gout
Basic Science and Clinical Practice

Edited by Dwight R. Robinson


Author Editor
David S. Newcombe, M.D. Dwight R. Robinson, M.D.
Weston Department of Rheumatology
Massachusetts Massachusetts General Hospital
USA Boston
USA

ISBN 978-1-4471-4263-8 ISBN 978-1-4471-4264-5 (eBook)


DOI 10.1007/978-1-4471-4264-5
Springer London Heidelberg New York Dordrecht

Library of Congress Control Number: 2012950015

© Springer-Verlag London 2013


This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or
part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way,
and transmission or information storage and retrieval, electronic adaptation, computer software,
or by similar or dissimilar methodology now known or hereafter developed. Exempted from this
legal reservation are brief excerpts in connection with reviews or scholarly analysis or material
supplied specifically for the purpose of being entered and executed on a computer system, for
exclusive use by the purchaser of the work. Duplication of this publication or parts thereof is
permitted only under the provisions of the Copyright Law of the Publisher’s location, in its
current version, and permission for use must always be obtained from Springer. Permissions for
use may be obtained through RightsLink at the Copyright Clearance Center. Violations are liable
to prosecution under the respective Copyright Law.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are
exempt from the relevant protective laws and regulations and therefore free for general use.
While the advice and information in this book are believed to be true and accurate at the date of
publication, neither the authors nor the editors nor the publisher can accept any legal responsibility
for any errors or omissions that may be made. The publisher makes no warranty, express or
implied, with respect to the material contained herein.

Printed on acid-free paper

Springer is part of Springer Science+Business Media (www.springer.com)


This book is dedicated to Dr. Newcombe’s daughters:
Catherine L. Newcombe, Kirsten N. Shilling and Sarah N. Faucett.
Preface

One might ask why a book devoted to a single disorder like gout is necessary
since standard textbooks adequately review the usual clinical findings and
management of this disorder. The answer to that question is multifaceted.
Gout is now clearly associated with several different clinical presentations
that are often not described in detail in modern textbooks. Familial juvenile
hyperuricemic nephropathy, autosomal dominant polycystic kidney disease,
glycogen storage disease, and other disorders associated with hyperuricemia
and gout are dysfunctions that need to be recognized early to avoid serious
consequences. These and other clinical subsets of gout, in some cases, have
also been defined at the molecular level. Such advances in molecular biology
provide another rationale for amplifying a physician’s knowledge of gout and
the mechanisms of inflammation associated with this disease. Furthermore,
the inflammatory and anti-inflammatory mediators, their mechanisms of
action, and their real and putative roles in gout have recently been elucidated
in greater detail and contribute to a better understanding of the pathogenesis
of gout. Such new discoveries also provide a perspective on the capacity of
the host to generate natural anti-inflammatory molecules, and such data may
lead to new therapeutic initiatives. Uric acid, the precursor of gout, has also
begun to attain more significance in relation to renal disease, hypertension,
and obesity, and these interrelationships have been characterized in specialty
journals but are not linked to textbook discussions of hyperuricemia and gout.
These findings alert the physician to evaluate uric acid associated disorders
such as hypertension and obesity more extensively than has been the custom
previously.
Perhaps of greatest significance to the subjects of hyperuricemia and gout
are the epidemiological studies showing that fewer than 10% of patients with
gout are referred to rheumatologists. This circumstance leads to multiple
errors in the management of this disease by physicians as well as patients. For
example, the literature clearly documents the misuse of allopurinol since a
number of patients are prescribed this agent for inappropriate reasons.
Treating asymptomatic hyperuricemia with allopurinol in the absence of a
sound rationale for the reduction in uric acid levels places patients at risk for
the serious side effects of this agent including renal failure and even death. In
addition, allopurinol has also been used as the sole agent for the treatment of
acute gouty arthritis in elderly patients suffering from diuretic-induced gout
as well as in other inappropriate settings where allopurinol is not indicated.
Although uric acid is often the precursor of acute gouty arthritis, allopurinol

vii
viii Preface

has no role in the management of acute gouty arthritis since it has no effect
on an acute inflammatory response. Patients with hand deformities due to
extensive tophaceous deposits may often be mistakenly diagnosed as rheu-
matoid arthritis, and the need for the reduction of tophaceous deposits com-
pletely ignored. Neglecting the treatment of chronic tophaceous gout may
have serious consequences such as the collapse of a urate-laden vertebra or
aseptic necrosis of the hip.
Another important aspect of gout in modern times is the increasing
identification of genetic dysfunctions associated with hyperuricemia and
gout. Thus, a family history becomes a critical part of the evaluation of the
hyperuricemic patient and avoids the pitfalls related to a delay in the early
detection and treatment of affected family members. Often a cooperative
patient who researches the family for underlying disorders may provide the
physician with a most rewarding experience and be most useful in the early
treatment of family members.
The contents of this monograph provide the practicing physician with a
ready source of information concerning hyperuricemia and gout as well as
discussions of the management goals for the optimal treatment of these
disorders.
About the Author

David Sugden Newcombe, M.D., graduated from Amherst College in 1952,


cum laude in Biology. He received his medical degree from McGill University
in 1956. Dr. Newcombe did his postdoctoral training at Boston City Hospital,
Duke University, Boston University, and the Peter Brent Brigham Hospital at
Harvard. He completed his military service in Korea in 1958.
From 1965 to 1967, he was an Assistant Professor of Medicine at the
University of Virginia. He then served as an Associate Professor of Medicine
at the University of Vermont where he subsequently became the Director of
Rheumatology. In 1977, he went to The Johns Hopkins University where he
rose to the rank of Professor of Environmental Health Science in the
University’s School of Hygiene and Public Health and held a Joint
Appointment in the Department of Medicine in the University’s School of
Medicine.
Upon retiring from Johns Hopkins in 1992, he moved to Massachusetts
where he was Associate Chief of Staff at the Bedford Veterans Administration
Hospital until 1999. He then served as an Associate Physician at the Federal
Medical Center (Devens) Massachusetts until he retired in 2001.
Dr. Newcombe wrote two books: Inherited Biochemical Disorders and
Uric Acid Metabolism (1975 University Park Press) and Clinical
Immunotoxicology (1992 Raven Press). He also authored over one hundred
medical and scientific articles.
At the time of his retirement, Dr. Newcombe began the project that eventu-
ally became this book on gout. He had a life-long interest in the disease and
its complexities. The 1,000 page manuscript was completed just five days
before his death. Almost a year later his family requested that Dr. Robinson
arrange for the publication of his work. The manuscript was then modified
and updated. At this time the book represents the most comprehensive publi-
cation available on gout, which is the world’s most common inflammatory
rheumatic disease.
The Editor, Dwight R. Robinson, M.D., is a rheumatologist at the
Massachusetts General Hospital and Professor of Medicine, Harvard Medical
School.

ix
Acknowledgment

The Newcombe family is profoundly grateful to Dr. Dwight Robinson.


Dr. Newcombe’s manuscript on gout would never have been published had it
not been for Dr. Robinson’s expert knowledge of this disease and his unwav-
ering dedication to the substantial task of editing the valuable piece of medi-
cal scholarship that Dr. Newcombe produced.

xi
Contents

1 A Brief History of Gout ................................................................. 1


Egyptian Archeology ....................................................................... 1
Byzantine Empire ............................................................................. 2
Seventeenth Century ........................................................................ 2
Eighteenth Century........................................................................... 2
Nineteenth Century .......................................................................... 4
Twentieth Century ............................................................................ 4
Famous Sufferers.............................................................................. 6
References ........................................................................................ 7
2 The Prevalence and Risk Factors for Gout .................................. 9
Prevalence of Gout ........................................................................... 9
Diet, Disease States, and Heredity Associated with Gout ............... 9
Genetic Abnormalities in Renal Tubular Transport Leading
to Reduced Renal Excretion of Urate............................................... 13
References ........................................................................................ 18
3 Purine Biochemistry....................................................................... 25
Structure ........................................................................................... 25
Nucleic Acid Degradation ................................................................ 25
De Novo Purine Nucleotide Synthesis ............................................. 30
The Key Intermediate, PRPP ........................................................... 33
Biosynthesis of AMP and GMP ....................................................... 33
Nucleotide Interconversions and Catabolism ................................... 34
Purine Salvage Pathways.................................................................. 34
Phosphoribosylpyrophosphate Synthetase ....................................... 36
PRPP Amidotransferase ................................................................... 40
Hypoxanthine-Guanine Phosphoribosyltransferase (HPRTase)....... 42
Adenine Phosphoribosyltransferase ................................................. 47
Isotopes in the Evaluation of Uric Acid Metabolism ....................... 54
References ........................................................................................ 55
4 Uric Acid Metabolism in Humans ................................................ 69
Overview .......................................................................................... 69
Purine Biosynthesis .......................................................................... 69
Miscible Pool of Urate ..................................................................... 70
Urate Turnover Rate ......................................................................... 72
Mechanisms of Hyperuricemia and Gout ........................................ 72
Elevated PRPP Levels ...................................................................... 72

xiii
xiv Contents

Decreased Hypoxanthine Reutilization ............................................ 73


Glutamine Hypothesis ...................................................................... 75
Accelerated ATP Degradation .......................................................... 75
Increased Nucleic Acid Turnover..................................................... 81
Accelerated Nucleotide Degradation ............................................... 83
Excessive Purine Intake.................................................................... 84
Elimination of Uric Acid.................................................................. 84
Renal Mechanisms of Hyperuricemia and Gout .............................. 85
Renal Mechanisms of Hyperuricemia and Secondary Gout ............ 86
References ........................................................................................ 87
5 Clinical Aspects of Gout and Associated Disease States ............. 91
Introduction ...................................................................................... 91
Clinical Gout ............................................................................... 91
Intercritical Gout ......................................................................... 93
Chronic (Tophaceous) Gouty Arthritis ........................................ 94
Differential Diagnosis ...................................................................... 96
Diagnostic Criteria for Hyperuricemia and Gout ............................. 99
Urinary Uric Acid Measurements .................................................... 102
Overproduction of Uric Acid ........................................................... 102
Lesch-Nyhan Syndrome .............................................................. 103
Kelley-Seegmiller Syndrome ...................................................... 104
Phosphoribosylpyrophosphate Synthetase Overactivity ............. 106
Idiopathic Gout ............................................................................ 106
Secondary Hyperuricemia and Gout due to Overproduction
of Purines..................................................................................... 107
Glycogen Storage Diseases (GSD).............................................. 108
Fatty Acid Oxidation Deficiencies .............................................. 113
Decreased Uric Acid Excretion........................................................ 114
Introduction ................................................................................. 114
Primary Underexcretor Gout ....................................................... 114
Secondary Hyperuricemia and Gout due to the Underexcretion
of Uric Acid ................................................................................. 114
Urate Nephropathy ...................................................................... 115
Acute Uric Acid Nephropathy..................................................... 117
Autosomal Dominant Polycystic Kidney Disease (ADPKD) ..... 118
Medullary Cystic Disease (MCD) ............................................... 119
Familial Juvenile Gouty Nephropathy (FJGN) ........................... 120
Nephrogenic Diabetes Insipidus (NDI) ....................................... 121
Uric Acid Nephrolithiasis ................................................................ 122
Introduction ................................................................................. 122
Heritable Causes of Uric Acid Nephrolithiasis ........................... 124
Acquired Causes of Uric Acid Nephrolithiasis ........................... 128
Nephrolithiasis Mimicking Uric Acid Stones ............................. 129
Drug-Induced Hyperuricemia and Gout........................................... 135
Diuretics ...................................................................................... 135
Salicylates .................................................................................... 136
Ethanol ........................................................................................ 136
Contents xv

Cytotoxic Agents ......................................................................... 136


Allopurinol-Induced Gout ........................................................... 136
Antituberculous Drugs ................................................................ 137
Cyclosporine................................................................................ 137
Nicotinic Acid ............................................................................. 139
Fructose-Induced Hyperuricemia ................................................ 139
Megaloblastic Anemia ................................................................. 140
Summary of Drug-Induced Hyperuricemia and Gout ................. 141
Saturnine Gout (Lead-Induced Gout)............................................... 141
Gaucher Disease (Glucocerebrosidase Deficiency) ......................... 144
Gout in Females ............................................................................... 144
Juvenile Hyperuricemia and Gout ............................................... 144
Premenopausal Hyperuricemia and Gout .................................... 145
Hyperuricemia and Gout in Pregnancy ....................................... 146
Postmenopausal Hyperuricemia and Gout .................................. 146
Associated Disorders........................................................................ 147
Obesity ........................................................................................ 147
Hyperlipidemia ............................................................................ 148
Glucose Intolerance ..................................................................... 152
Hypertension ............................................................................... 152
Atherosclerosis ............................................................................ 153
Critical Illnesses .......................................................................... 153
References ........................................................................................ 154

6 Diagnostic Procedures in the Management of Gout ................... 187


Introduction ...................................................................................... 187
Hyperuricemia .................................................................................. 187
Synovial Fluid Analysis ................................................................... 188
Arthrocentesis Techniques .......................................................... 188
Synovial Fluid Culture ................................................................ 189
Synovial Fluid Gram Stain .......................................................... 190
Synovial Fluid White Blood Cell Counts .................................... 190
Synovial Fluid Differential Cell Count ....................................... 191
Synovial Fluid Wet Preparations for Crystal Identification ........ 191
Additional Clinical Data Concerning Synovial Fluid ................. 192
References ........................................................................................ 194

7 Roentgenographic Findings and Musculoskeletal Ultrasound .. 199


Introduction ...................................................................................... 199
Soft Tissue Swelling and Joint Effusions ......................................... 199
Bony Erosions .................................................................................. 200
Extra-articular Tophi ........................................................................ 201
Uric Acid Calculus ........................................................................... 201
Avascular Necrosis ........................................................................... 202
Chondrocalcinosis ............................................................................ 202
Musculoskeletal Ultrasound ............................................................. 203
Dual-Energy Computed Tomography (DECT) ................................ 203
References ........................................................................................ 204
xvi Contents

8 Mechanisms of the Acute Attack of Gout and Its Resolution .... 207
Overview .......................................................................................... 207
Synovial Membrane ......................................................................... 208
Monosodium Urate Crystallization .................................................. 211
Inflammasome: Innate Immunity; Inflammation
in Gout Is Mediated by the Innate Immune System......................... 213
Introduction ................................................................................. 213
Interleukin-1b and Interleukin-18 ............................................... 216
Mechanisms of NLRP3 Activation ............................................. 217
Neutrophil Chemoattractants ........................................................... 218
Overview ..................................................................................... 218
Proteinases and Neutrophil Migration......................................... 219
The Complement Fragments: C5a and C5a des Arg ................... 220
C5a Receptor (C5aR) .................................................................. 221
Platelet-Activating Factor (PAF) ................................................. 222
PAF Receptor ................................................................................... 227
Biological Functions of PAF ....................................................... 227
Regulation of PAF: Acetylhydrolase ........................................... 228
Leukotriene B4 (LTB4) ............................................................... 229
LTB4 Biosynthesis via Phospholipase/Lipoxygenase
and Its Release ............................................................................. 230
LTA4 Hydrolase .......................................................................... 231
Transcellular Metabolism and LTB4 Production ........................ 231
Phospholipase Classification and Functions ............................... 232
Interleukin-8 (IL-8) ..................................................................... 235
Cell Sources................................................................................. 236
IL-8 Gene .................................................................................... 236
IL-8, MSU Crystals, and Gout .................................................... 237
Adhesion Molecules ......................................................................... 238
An Overview of Selectins and Integrins ...................................... 238
Selectin Structures ....................................................................... 238
P-Selectin Glycoprotein Ligand-1 ............................................... 239
L-Selectin .................................................................................... 239
Unique L-Selectin Properties ...................................................... 240
E-Selectin .................................................................................... 242
Integrins ....................................................................................... 243
Integrin Structure ......................................................................... 243
Ligands for Integrins ................................................................... 245
Oxygen Radical Generation ............................................................. 247
NADPH Oxidase Assembly ........................................................ 249
Oxidants and Oxidant-Mediated Destructive Responses ............ 250
Myeloperoxidase ......................................................................... 251
Nitric Oxide and Bacterial Killing .............................................. 251
Oxidants and Uric Acid Destruction ........................................... 252
Proinflammatory Mediators ............................................................. 253
Non-chemotactic, Proinflammatory Cell Mediators ................... 253
Vasoactive Arachidonic Acid Metabolites .................................. 253
Contents xvii

Bradykinin ................................................................................... 254


Tumor Necrosis Factor ................................................................ 254
Monosodium Urate Crystal-Induced Inflammation ......................... 255
Proinflammatory Mediators and MSU Crystals .......................... 255
Regulation of Synovial Fibroblast-Like Cells ............................. 256
MAP Kinase and MSU Crystal-Induced Neutrophil Activation . 256
Phospholipase D and MSU Crystals ........................................... 257
MSU Crystals and Other Phospholipids Metabolizing Enzymes 258
Other Pathways Stimulated by MSU Crystals ............................ 258
MSU Crystal-Induced Proinflammatory Mediators .................... 259
MSU Crystals and Cell Membrane Interactions.......................... 259
Neutrophil and Macrophage Apoptosis ........................................... 260
Overview ..................................................................................... 260
Endogenous and Exogenous Anti-inflammatory Agents ................. 260
Overview ..................................................................................... 260
Anti-inflammatory Eicosanoids ....................................................... 262
Overview ..................................................................................... 262
Lipoxin Biosynthesis........................................................................ 263
Anti-inflammatory Effects of Lipoxins ....................................... 265
Proinflammatory to Anti-inflammatory Switch........................... 266
Resolvin Biosynthesis ................................................................. 266
Resolvin Receptors ...................................................................... 266
PAF Acetylhydolase and Anti-inflammation............................... 270
Apoptosis and Anti-inflammation ............................................... 271
Nitric Oxide and Anti-inflammation ........................................... 272
Drugs ........................................................................................... 274
Glucocorticoid .................................................................................. 274
Overview ..................................................................................... 274
Mode of Action ........................................................................... 275
Histone Acetylation and Glucocorticoids.................................... 277
Annexins and Glucocorticoids .................................................... 278
References ........................................................................................ 278
9 Management of Hyperuricemia and Gout ................................... 291
Overview .......................................................................................... 291
Management of Asymptomatic Hyperuricemia ............................... 292
Symptomatic Drug-Induced Hyperuricemia .................................... 294
Hyperuricemia Associated with the Treatment
of Neoplastic Disease ....................................................................... 295
Treatment of Acute Gouty Arthritis ................................................. 295
Colchicine Use in Acute and Chronic Gout ..................................... 296
Treatment Schedules ................................................................... 296
Mechanism of Action .................................................................. 297
Pharmacokinetics of Colchicine .................................................. 298
Colchicine Toxicity ..................................................................... 299
Nonsteroidal Anti-inflammatory Drugs (NSAIDs) .......................... 301
Toxicity of NSAIDs..................................................................... 304
Other Toxicities of NSAIDs ........................................................ 305
xviii Contents

Indomethacin (Indocin) .................................................................... 313


Treatment Schedules ................................................................... 314
Toxicity ........................................................................................ 314
Naproxen .......................................................................................... 315
Glucocorticoids ................................................................................ 316
Therapeutic Regimens ................................................................. 320
Interleukin 1 Inhibitors..................................................................... 322
Urate-Lowering Therapy: Uricosuric
and Other Hypouricemic Drugs ....................................................... 323
Uricosuric Drugs: Sulfinpyrazone, Probenecid,
and Benzbromarone ......................................................................... 325
Sulfinpyrazone ............................................................................. 325
Probenecid ................................................................................... 326
Salicylates .................................................................................... 327
Benzbromarone ........................................................................... 328
Xanthine Oxidase Inhibitors: Allopurinol and Febuxostat .............. 329
Allopurinol .................................................................................. 329
Determination of Renal Function ................................................ 334
Newer Urate-Lowering Drugs ..................................................... 335
Low-Purine Diet ............................................................................... 339
Special Treatment Regimens ............................................................ 339
Management of Acute Uric Acid Nephropathy .......................... 339
Management of Tophaceous Deposits ......................................... 340
Lesch-Nyhan Syndrome .............................................................. 341
Glycogen Storage Disease ........................................................... 341
Hereditary Fructose Intolerance (HFI) ........................................ 341
Refractory Gout ........................................................................... 342
Pegloticase ................................................................................... 344
Other Agents with Urate-Lowering Effects ..................................... 345
Fenofibrate ................................................................................... 345
Losartan ....................................................................................... 345
Management of Nephrolithiasis ....................................................... 346
Allopurinol Treatment of Stones ...................................................... 348
The Role of Diet and Comorbidities
and Their Management in Gout ....................................................... 348
Hypertension ............................................................................... 349
Summary .......................................................................................... 353
Summary of Principles of Management........................................... 354
Lifestyle ....................................................................................... 354
Acute Attacks .............................................................................. 354
Chronic Gout ............................................................................... 354
References ........................................................................................ 355
Index ....................................................................................................... 387
A Brief History of Gout
1

Ancient Greek and Roman civilization Gout Aulus Cornelius Celsus (25 B.C.–50 A.D.),
appeared to be well recognized both by ancient author of the most comprehensive Roman medical
Greek and Roman writers as well as physicians text of the time (De medicina), first proposed
even though confusion with other forms of venesection at the outset of a gouty episode both
arthritis may have existed. The term gout arose to initiate a cure and to utilize as a prophylactic
from the Latin word, gutta, which means drop measure. Aretaeus the Cappadocian, another sec-
and was selected as a term to explain the con- ond century physician and contemporary of
cept that bad humors entered the afflicted part Galen, was the first to propose that a specific toxic
drop by drop [1]. The first clinical descriptions substance in the blood was the cause for the gout.
of gout are attributed to Hippocrates (460– He also provided a precise clinical description of
377 B.C.) who documented the rarity of gout in gouty arthritis: “The joints begin to be affected in
premenopausal women, young boys (ante usum this way: pain seizes the great toe then the fore-
veneris – prepubertal), and eunuchs [2]. Indeed, part of the heel on which we rest; next it comes
the same author termed the affliction in the big into the arch of the foot, but the ankle-joint swells
toe, podagra, based on the topographical loca- last of all. All sufferers at first wish to blame the
tion of the affliction rather than as an etiological wrong cause - some function of a new shoe, oth-
concept. Chiagra and gonagra were the terms ers a long walk, others an accident, or being trod-
given to those with gout affecting the wrist or den upon ---but the true cause is seldom believed
knee. Hippocrates also noted the periodicity of by the patient when he hears it from the physi-
gouty episodes and the recurrence of gout in the cian.” Galen (130–200 A.D.) provided a more
spring and fall. In the early descriptions of gout, radical approach to the disorder; bleeding and
there was a general concept that prior to the purgation along with local applications were used
deposition of chalk (tophi), the disease could be as treatments. He was also the first to describe
controlled, but after the occurrence of such tophi, the term arising from the Greek word mean-
deposits, the disease became more difficult to ing rough crumbling rock. Galen is also credited
control. The elder Garrod summarized the role with the first description of the tophus [4].
of Greek physicians in describing the diagnosis
and management of gout [3]. As the Augustan
poet, Publius Ovid (43 B.C.), wrote in his Pontic Egyptian Archeology
epistles, “Tollere nodosam nescit medicina
pologram.” Translated from the Latin, this Two findings of archeological studies are of
means that the gouty swellings defied the art of interest to the historical analysis of gout. Smith
medicine. and Jones [5] discovered a large mass of urates

D.S. Newcombe, Gout, 1


DOI 10.1007/978-1-4471-4264-5_1, © Springer-Verlag London 2013
2 1 A Brief History of Gout

in the great toe of an elderly male buried in a [9]. In his writings, he differentiated acute and
cemetery in Upper Egypt. The chemical compo- chronic gouty arthritis for the first time and
sition of this mass was shown to be urates by recorded this classical description:
chemical analysis. A renal calculus found in an Towards the end of January or the beginning of
Egyptian mummy on analysis was shown to February suddenly, and with scarcely any premon-
contain a nucleus of uric acid [6]. The age of itory feelings, the disease breaks out. Its only fore-
this mummy was estimated to be about runner is indigestion and crudity of the stomach,
which troubles the patient for some weeks previ-
7,000 years old. Thus, these studies documented ous to the attack. His body also feels swollen,
chronic tophaceous gout and urate calculi as heavy, and windy - symptoms which increase from
disorders of antiquity. day to day until the fit breaks out. But a few days
before this torpor comes on, and a feeling of flatus
along the legs and thighs. Besides this, there is a
spasmodic affection, whilst the day before the fit,
Byzantine Empire the appetite is unnaturally hearty. The victim goes
to bed in good health and sleeps. About two o’clock
Alexander of Tralles (525–605 A.D.), a Byzantine in the morning he is awakened by a severe pain,
generally in the great toe, more rarely in the heel,
physician, and his contemporary, Aetius, were ankle, or instep. This pain is like that of a disloca-
the first to indicate the usefulness of colchicum in tion of the bones of these parts, and is accompa-
the treatment of gout [2, 7]. “Hermodactylon (the nied by a sensation as of chilly water poured over
finger of Hermes) confestim minuit dolores,” the membranes of the suffering joint. Then follows
chills and shivers and a little fever. The pain, which
noted Alexander. There has been speculation that at first moderated, becomes gradually more intense,
these early Byzantine physicians were referring and while it increases the chills and shivers die out.
to Colchicum variegatum rather than Colchicum Every hour that passes finds it greater, until at
autumnale, the present-day source of colchicine. length at nighttime it reaches its worst intensity,
and insinuates itself with most exquisite cruelty
In fact, the use of this medication dates back to among the numerous small bones of the tarsus and
the Ebers Papyrus of 1500 B.C., but historical metatarsus, in the ligaments of which it is lurking.
records document its use in more detail in pub- Now it is a violent stretching and tearing of the
lished works in Latin [8]. ligaments, now it is gnawing pain, and now a pres-
sure and tightening. So exquisite and lively mean-
while is the feeling of the part affected that it
cannot bear the weight of the bedclothes nor the jar
Seventeenth Century of a person walking in the room. Hence the night is
passed in torture and a restless rolling first to one
side, then to the other, of the suffering limb, with
The actual usage of the word gout has been attrib- perpetual change of posture, the tossing about of
uted to Geoffroi de Villehardouin writing in the the body being as incessant as the pain of the tor-
1200s that Count Hugues de Saint Paul suffered tured joint, and being at its worst as the fit is com-
from “une grant maladie de gote.” To the present ing on. Hence the vain efforts by change of posture,
both in the body and the limb affected, to obtain an
day, gout is used in most languages (French – abatement of the pain.
goutte, Spanish – gota, Italian – gotta, and
German – gicht) to describe this malady. The Thus, Sydenham documented the presence of
modern history of gout probably begins with Sir low-grade fever in association with involvement
Thomas Sydenham (1624–1689) who was of the distal extremities and related to overeating.
afflicted with the disease and provided physicians
with one of the best clinical descriptions of the
disease. This talented English physician is Eighteenth Century
responsible for differentiating gout from other
forms of arthritis. In his book entitled, A Treatise Anton van Leeuwenhoek (1632–1723), the Dutch
on Gout and Dropsy, he provided the literature biologist who became the father of microscopy,
with a remarkably accurate description of gouty discovered that the gouty tophus was made up of
arthritis based on his own personal experiences crystals, but the actual nature of the crystal
Eighteenth Century 3

remained unknown. The discovery of uric acid as him to let me have some of the chalk, which he
a component of tophi was discovered by the willingly granted me, mentioning the time when he
would bandage his arm. For this purpose I brought
English physician turned chemist, William two small glasses in which there had never been
Wollaston, a nephew of William Heberden whose anything, to put the chalk in; the substance which
name is now associated with osteoarthritis of the issued from the arm was not only chalk but was
distal interphalangeal joints [10, 11]. In the pub- mixed with some pus and a little blood, as also with
a viscous fluid in which there were many whitish
lished letters of van Leeuwenhoek, the following spots, some of which were so small that they could
remarkable description of crystals from a gouty hardly be seen by the naked eye. All this substance
tophus is recorded. I separated as well as I could with the point of a
knife, in order to be able to distinguish it all the
In your previous letter you ask me to examine the better. First of all I observed the solid matter which
chalk which gouty persons have on their joints or to our eyes resembles chalk, and saw to my great
which will break out. Now, although I am familiar astonishment that I was mistaken in my opinion,
with many people suffering severely from gout, the for it consisted of nothing but long, transparent lit-
joints of whose fingers are very thick, I never tle particles, many pointed at both ends and about 4
observed a perforation. Consequently, I always “axes” of the globules in length, others shorter and
thought that I could not discover anything worth a few only half as long. And the proportion of their
noting in the chalky matter, imagining that the thickness as compared to their length, I cannot bet-
chalk would consist only of globules, because, if it ter describe than by supposing that we saw with
should contain pointed particles, the gouty patient our naked eye pieces from a horse-tail cut to a
would never be free from pain, whereas we observe length of one sixth of an inch. In a quantity of mat-
the contrary to be the fact. ter of the size of a course grain of sand, there lay
A certain gentleman who is related to me and who is some thousands of these long figures, mixed with a
severely affected by gout, having heard about three small quantity of fluid. And roundabout this chalk
years ago that I had some Moxa and that I had burnt there flowed a small quantity of a thin fluid which
myself with it for research, asked me for some of it I judged to be the watery fluid of our blood, mixed
in order to burn himself with it. I was glad to give it with globules of blood and globules which were
to him, after which he went to Utrecht where he very white, which I judged to be also blood and
caused himself to be burnt several times, but which is generally called pus. In some places the
obtained no relief from it. When this gentleman above-mentioned long particles lay in regular
returned to this place after having lived in another order, one next to the other, and not in a mass as if
town for some time, I was told that he had had a very they formed one body, but in a manner in which
sudden and severe attack. This gentleman passing glands lie. The clear matter was so viscous and
my house a few days afterwards, I inquired after his tough that I would not have thought that such a
health upon which he told me about his last illness, humid matter could be found in our body, the more
which I will now describe for your information. so because after several observations I could not
For two days consecutively he had gone to stool but judge that it consisted of closely joined glob-
many times, after which he felt so weak that he had ules, in many places mixed with the above men-
a doctor sent for. After this he fainted and had ner- tioned long figures which in one place lay very
vous twitches, reason why only cordials were pre- close together and in another place lay widely scat-
scribed, because it was supposed that his life would tered. These were whitish spots that I had observed
last but a short time. He next was seized with cramp with my native eye in the clear matter, and which
in many limbs, so that, as he complained to me, if were whitest where they lay closest together. For
he moved a limb in order to rub the cramped part all transparent parts, lying one upon the other with-
he was at once seized again with cramp in the arm out being closely united, look white, such as snow,
or leg which he moved. The stools ceased sponta- pounded glass or resin, sugar, paper, etc.
neously and all his joints became so thin as if all
the gouty swellings had been carried off in the This description, of course, provided no clue
stools. But he complained that they already began as to the chemical composition of the so-called
to grow again. A few days later I asked this gentle-
man if it had ever happened to him that chalk came chalk and was completely dependent on the sub-
out of his joints; he told me that some time ago the sequent chemical characterization of uric acid by
chalk in the heel of his foot, in which there was a Carl Scheele (1742–1786), a Swedish chemist
large hole, came out in such quantity that he formed and pharmacist who is responsible for an unparal-
almost a new heel, and that there was also a hole in
his arm on his elbow from which the chalk had leled description of new chemicals [12]. The char-
come during quite six months on end, but not so acterization of the material from a tophus as uric
much by far, nor so thick as from his heel. I asked acid was reported by W. H. Wollaston in 1797 and
4 1 A Brief History of Gout

marked the end of the humoral theories of gout formation of uric acid in the system, or which lead
and the introduction of the chemical hypothesis as to its retention in the blood. (The discussion of the
eighth proposition is of much interest and consid-
the explanation for the etiology of gout [11]. erable importance, for if we can prove the truth of
the statement that the predisposing causes are of
different kinds, one leading to the increased forma-
Nineteenth Century tion of the morbid matter, the other to its retention
in the blood, we at once have a clue to the varieties
of the disease, popularly known as the rich and the
In the mid-1800s, Sir Alfred B. Garrod published his poor man’s gout).
identification of increased levels of uric acid in the Ninthly, the causes exciting a gouty fit, are those
blood of gouty patients using the murexide test and which induce a less alkaline condition of the blood,
or which greatly augment for the time, the forma-
subsequently the “thread” test [3, 13–15]. He sum- tion of uric acid, or such as temporarily check the
marized his clinical conclusions as follows: eliminating power of the kidney.
Tenthly, in no disease but true gout is there a depo-
First, in true gout, uric acid, in the form of urate of sition of urate of soda in the inflamed tissues.
soda, is invariably present in the blood in abnormal
quantities, both prior to and at the period of the
seizure, and is essential to its production; but this Garrod’s propositions set the stage for the
acid may occasionally exist largely in the circulat- investigations that characterize many of the
ing fluid without the development of inflammatory
aspects of clinical gout as clinicians know it today.
symptoms, as for example in some cases of lead
poisoning, and a few other instances. Its mere pres- Parallel with the descriptions of the clinical
ence, therefore, does not explain the occurrence of aspects of gout, the biochemical and pharmaco-
the gouty paroxysm. logic aspects of purine metabolism and their rela-
Secondly, the investigations recently made in the
tion to gout were developing. In 1898, Emil
morbid anatomy of gout prove incontestably that
true gouty inflammation is always accompanied with Fischer (1852–1919), a Nobel laureate, first estab-
a deposition of urate of soda in the inflamed part lished that uric acid was a purine compound and
(This fact I wish to impress forcibly on the minds of related to the nucleic acid constituents, adenine
my readers, because in the constancy of such deposi-
and guanine [16]. Folin and Denis in 1913
tion, lies the clue which has long been wanting: the
occurrence of the deposit is perfectly pathogno- described a colorimetric method for the measure-
monic, and at once separates gout from other disease ment of blood uric acid levels, which led the way
which at first sight may appear allied to it). to clinical and metabolic studies of patients with
Thirdly, the deposit is crystalline and interstitial,
gout [17]. After its early discovery as a therapeu-
and when once the cartilages and ligamentous
structures become infiltrated, such deposition tic modality, colchicine was rediscovered in the
remains for a lengthened time. mid-1700s by Baron Anton de Storck, the physi-
Fourthly, the deposited urate of soda may be looked cian for Empress Maria Theresa of Austria who
upon as the cause, and not the effect, of gouty
used it principally for the treatment of dropsy and
inflammation.
Fifthly, the inflammation which occurs in the gouty not gout [8]. Benjamin Franklin (1706–1790)
paroxysm tends to the destruction of the urate of brought a concoction called L’Eau d’Husson from
soda in the blood of the inflamed part, and conse- France for the treatment of his gouty episodes,
quently of the system generally.
and in 1814, Dr. James Want determined the active
Sixthly, the kidneys are implicated in gout, proba-
bly in its early, and certainly in its chronic stages, ingredient of Nicolas Husson’s elixir was colchi-
and the renal affection, perhaps only functional at cine [18, 19]. Thus, Franklin is credited with the
first, subsequently becomes structural; the urinary introduction of colchicine to the United States.
secretion is also altered in composition.
Seventhly, the impure state of the blood, arising
principally from the presence of urate of soda, is
the probable cause of the disturbance which pre- Twentieth Century
cedes the seizure, and of many of the anomalous
symptoms to which gouty subjects are liable.
For more than a century, little progress was made
Eighthly, the causes which predispose to gout,
independently of those connected with individual in the pharmacologic treatment of gout until
peculiarity, are either such as produce an increased probenecid, Benemid, was synthesized by Beyer
Twentieth Century 5

and his colleagues as an analog of carinamide and purine metabolism [33]. Two decades later,
a putative inhibitor of penicillin secretion [20, 21]. DeWitt Stetten and his colleagues at the National
The objective of this investigative program was to Institutes of Health initiated studies using purine
find an organic acid inhibitor of the renal tubular isotopes to investigate the synthesis and disposal
secretion of penicillin that would serve to increase or uric acid in the human [34]. At the same time,
the blood levels of this antibiotic and delay its John Buchanan and his students at the
excretion. Subsequently, it was determined that Massachusetts Institute of Technology and others
probenecid was a competitive inhibitor of many purified and characterized the enzymes of the
organic acids, and its primary effect of uric acid de novo purine biosynthetic pathway. More
was to inhibit the renal tubular reabsorption of recently, Zalkin and Smith and their colleagues at
filtered urate. Alexander Gutman and his col- Purdue University have extended the biochemis-
league, Tsai F. Yu, as well as John Talbott’s group try of the purine enzymes. A number of eminent
contributed to the present-day understanding of physician-scientists including Alexander Gutman,
the clinical usefulness of this drug in the manage- Jay Seegmiller, and James Wyngaarden sought
ment of chronic tophaceous gout [22–27]. the mechanisms by which gouty patients differed
The next major breakthrough in the manage- from nongouty individuals [35–44]. These stud-
ment of gout came when the recent Nobel laure- ies focused on the purine pathway to investigate
ates, George Hitchings and Gertrude Elion, in the the regulation of urate synthesis and on the renal
early 1960s studied the effect of allopurinol on handling of urate to evaluate purine excretion.
the thiopurine metabolism, hyperuricemia, and In 1965, Michael Lesch, a medical student at
gout [28]. Allopurinol, a hypoxanthine analog, the time, and William Nyhan, a pediatrician,
was originally synthesized as a potential chemo- first described a metabolic abnormality of
therapeutic agent [29, 30]. Although it had no purine metabolism that now bears their name
effect on experimental tumors, it was found to be [45]. The Lesch-Nyhan syndrome is character-
a very effective inhibitor of xanthine oxidase ized at the molecular level by the complete
[31]. The initial clinical usage of allopurinol was absence of the purine salvage enzyme, hypoxan-
as an adjunct in the treatment of leukemia for thine-guanine phosphoribosyltransferase, and,
patients receiving 6-mercaptopurine. The marked interestingly, usually presents with evidence of a
reduction in serum and urinary uric acid levels marked increase in purine production, self-muti-
observed by investigators at Duke University lation, choreoathetosis, but no stigmata of gouty
(James Wyngaarden, Wayne Rundles, and their arthritis. It was determined subsequently that this
colleagues) and the Burroughs Wellcome group disorder was inherited as an X-linked recessive
(George Hitchings, Gertrude Elion, and their col- trait [46, 47]. Soon after this publication, William
leagues) in 1963 led to trials of this drug in the Kelley and Jay Seegmiller and their colleagues at
treatment of gout [32]. This agent is now rou- the National Institutes of Health described adults
tinely used and perhaps overused as an effective with gouty arthritis, renal calculi, and purine
agent for the treatment of certain forms of gout overproduction who were found to have a partial
and uric acid lithiasis and for the prevention of deficiency of the same purine salvage enzyme
obstructive uropathies due to uric acid in patients [48, 49]. Then, in 1972, Oded Sperling and his
with tumors sensitive to chemotherapeutic or colleagues described another X-linked recessive
radiotherapeutic modalities. disorder, phosphoribosylpyrophosphate syn-
With the advent of biochemical genetics and thetase overactivity, in patients with purine over-
the postulate put forth in 1931 by Sir Archibald production, gout, and uric acid lithiasis [50, 51].
E. Garrod, the distinguished Regius Professor of Although there have been a few reports of other
Medicine at Oxford and the son of Alfred Garrod, inherited forms of gout, these are rarer than the
that gout represented an inherited disorder and an ones discussed here and less well characterized
inborn error of metabolism ushered in the mod- from a mechanistic standpoint. Studies of purine
ern era of molecular medicine as it applied to metabolism continue today focusing on the early
6 1 A Brief History of Gout

detection and treatment of heritable disorders of


purine metabolism and the characterization of the
molecular basis of human purine mutants.

Famous Sufferers

Gout has been described as the disease of the


aristocracy since it has been an affliction of
statesmen, scientists, warriors, theologians, art-
ists, and men of letters through the centuries.
John Hunter (founder of experimental and surgi-
cal pathology), William Harvey (discoverer of
blood circulation), Charles Darwin (father of
modern biology), Carl Linnaeus (Swedish natu-
ralist and physician), and Isaac Newton (mathe-
matician and inventor of the telescope) as well as
others noted previously were all victims of the
disorder as were the warriors, Lord Howe,
Marshal Saxe (commander in the war of Polish
and Austrian succession), Albrecht Wallenstein
(Bohemian commander), Charlemagne (emperor
of the Holy Roman Empire), and Alexander the
Great (conqueror of Asia Minor and commander
of the Macedonian army) who also suffered from
the disease. Queen Anne (queen of England, Fig. 1.1 Consolation in the gout (Reproduced with per-
Scotland, and Ireland), Lord Beaverbrook mission from the American College of Rheumatology)
(Canadian-born millionaire, newspaper publisher,
and member of the British parliament), John citing an apt recipe to avoid the misfortune of the
Calvin (theologian and author), Benjamin disease when he wrote,
Franklin (American author and inventor), The rule of not too much, by temperance taught
Alexander Hamilton (American political leader), In what thou eat’st and drink’st, seeking from thence
Cotton Mather (fomenter of witchcraft hysteria Due nourishment, not gluttonous delight
in Salem and preacher), William Pitt the Elder
and the Younger (British statesmen and prime or as John Dryden described gout afflicting the
ministers), George Mason (American statesman hands,
and drafter of the constitution), Cardinal Wolsey Knots upon his gouty joints appear,
(the last medieval prince of the church), Count and Chalk is in his crippled fingers found.
Nikolaus Ludwig von Zinzendorff (German theo-
or the cure recommended by Charles Dickens,
logian), and other statesmen were afflicted with
gout. A number of distinguished artists and ‘The gout, sir,’ replied Mr. Weller, ‘The gout is a
authors were plagued by the disease including complaint as arises from too much ease and com-
fort. If ever you’re attacked with the gout, sir, just
John Milton, John Dryden, William Congreve,
you marry a widder as has got a good loud voice,
Henry Fielding, Johann Wolfgang von Goethe, with a decent notion of usin’ it and you’ll never
Samuel Johnson, Leonardo da Vinci, Thomas have the gout agin. It’s a capital prescription, sir. I
Gray, Ben Johnson, Martin Luther, and John takes it reg’lar, and I can warrant it to drive any
illness as is caused by too much jollity.’ Having
Wesley (English evangelist and theologian). Not
imparted this valuable secret, Mr. Weller drained
surprisingly, some writers transcribed their his glass once more, produced a laboured wink,
thoughts about gout as Milton may have done in sighed deeply and slowly retired. (Fig. 1.1)
References 7

References 23. Gutman AB, Yu TF. Current principles of manage-


ment of gout. Am J Med. 1952;13:744.
24. Yu TF, Dayton PG, Gutman AB. Mutual suppression
1. Hartung EF. Historical considerations. Metabolism.
of the uricosuric effects of sulfinpyrazone and salicy-
1957;6:196.
late: a study in interaction between drugs. J Clin
2. Hippocrates: The genuine works of Hippocrates, vol.
Invest. 1963;42:1330.
I and II, translated from the Greek with a preliminary
25. Gutman AB. Uricosuric drugs, with special reference
discourse and annotations by Francis Adams. New
to probenecid and sulfinpyrazone. Adv Pharmacol.
York: Wood; 1886.
1966;4:91.
3. Garrod AB. The nature and treatment of gout and
26. Yu TF, Berger L, Gutman AB. Hypoglycemic and uri-
rheumatic gout. London: Walton and Maberly; 1859.
cosuric properties of acetohexamide and hydroxyhex-
4. Neuwirth E. Milestones in the diagnosis and treat-
amide. Metabolism. 1968;17:309.
ment of gout. Arch Intern Med. 1943;72:377.
27. Talbott JH. Gout. 3rd ed. New York: Grune and
5. Smith GE, Jones FW. The archeological survey of
Stratton, Inc; 1967.
Nubia, report for 1907–8. vol. 2, Cairo, National
28. Rundles RW, Wyngaarden JB, Hitchings GW, et al.
Printing Department; 1910. p. 44 and p. 269.
Effects of a xanthine oxidase inhibitor on thiopurine
6. Kittredge WE, Downs R. The role of gout in the for-
metabolism, hyperuricemia, and gout. Trans Assoc
mation of urinary calculi. J Urol. 1952;67:841.
Am Phys. 1963;76:126.
7. Ghalioungui P. Rheumatic disorders in ancient
29. White FR. 4-Aminopyrazolo(3,4-d)pyrimidine and
Egyptian papyri. Egypt Rheumatol. 1964;1:4.
three derivatives. Cancer Chemother. 1959;3:26.
8. von Storch A. An assay on the use and effects of the
30. Shaw RK, Shulman RN, Davidson JK, et al. Studies
root of the Colchicum autumnale, or meadow saffron,
with the experimental antitumor agent 4-aminopyra-
translated from the Latin, T. Becket and PA deHonet;
zolo (3,4-d)pyrimidine. Cancer. 1960;13:482.
1764.
31. Feigelson P, Davidson JK, Robins PK. Pyrazol-
9. Sydenham T. A treatise of the gout and dropsy.
opyrimidines as inhibitors and substrates of xanthine
London: GGJ, J Robinson, W Otridge, S Hayes and
oxidase. J Biol Chem. 1957;226:993.
E Newbery; 1683.
32. Wyngaarden JB, Rundles RW, Silberman HR, et al.
10. McCarty DJ. A historical note: Leeuwenhoek’s
Control of hyperuricemia with hydroxypyrazolopy-
description of crystals from a gouty tophus. Arthritis
rimidine, a purine analogue which inhibits uric acid
Rheum. 1970;13:414.
synthesis. Arthritis Rheum. 1963;6:306.
11. Wollaston WH. On gouty and urinary concretions.
33. Garrod AE. The inborn factors in disease: an
Philos Trans R Soc Lond. 1797;87:386.
essay. London/New York: Oxford University Press
12. Scheele KW. Examen chemicum calculi urinarii.
(Clarendon); 1931.
Opuscula. 1776;2:73.
34. Wyngaarden JB, Stetten Jr D. Uricolysis in normal
13. Garrod AB. Observations on certain pathological
man. J Biol Chem. 1953;203:9.
conditions of the blood and urine in gout. Trans
35. Gutman AB, Yu TF. Renal function in gout with a
M-Chir Soc (Edinb). 1848;31:83.
commentary on the renal regulation of urate excre-
14. Garrod AB. On the blood and effused fluids in gout,
tion, and the role of the kidney in the pathogenesis of
rheumatism and Bright’s disease. Trans M-Chir Soc
gout. Am J Med. 1957;23:600.
(Edinb). 1854;37:49.
36. Gutman AB, Yu TF, Black H, et al. Incorporation of
15. Garrod AB. The nature and treatment of gout and
glycine-1–C14, glycine-2–C14, and glycine-N15 into
rheumatic gout. 2nd ed. London: Walton and Maberly;
uric acid in normal and gouty subjects. Am J Med.
1863.
1958;25:917.
16. Fischer E. Untersuchungen in der puringruppe. Berlin/
37. Sperling O, Wyngaarden JB, Starmer CF. The kinetics
New York: Springer; 1907.
of intramolecular distribution of 15N-glycine: a rein-
17. Folin O, Denis W. A new (colorimetric) method for
terpretation of the basis for the hypothesis of an
the determination of uric acid in the blood. J Biol
abnormality of glutamine metabolism in primary
Chem. 1912–1913;13:469.
gout. J Clin Invest. 1973;52:2468.
18. Schnitker MA. A history of the treatment of gout. Bull
38. Wyngaarden JB, Blair AE, Hilley L. On the mecha-
Inst Hist Med. 1936;4:89.
nism of overproduction in patients with primary gout.
19. Want J. The use of Colchicum autumnale in rheuma-
J Clin Invest. 1958;37:579.
tism. Med Physiol J (Lond). 1814;32:312.
39. Wyngaarden JB. Overproduction of uric acid as the
20. Beyer KH, Miller K, Russo HF, et al. The inhibitory
cause of hyperuricemia in primary gout. J Clin Invest.
effect of carinamide on the renal elimination of peni-
1957;36:1508.
cillin. Am J Physiol. 1947;149:355.
40. Seegmiller JE, Grayzel AI, Howell RR, et al. The
21. Beyer KH, Russo HF, Schuchardt DS, et al. 3-hydroxy-
renal excretion of uric acid in gout. J Clin Invest.
2-phenylcinchoninic acid (HPC): its absorption, excre-
1962;41:1094.
tion, and its effect of certain renal functions and
41. Gutman AB, Yu TF. An abnormality of glutamine
enzyme systems. J Pharmacol Exp Ther. 1951;103:79.
metabolism in primary gout. Am J Med. 1963;35:820.
22. Gutman AB, Yu TF. Benemid, (p-[di-n-propylsulfamyl]
42. Seegmiller JE, Klinenberg JR, Miller J, et al.
-benzoic acid) as uricosuric agent in chronic gouty
Suppression of glycine-N15 incorporation into urinary
arthritis. Trans Assoc Am Phys. 1951;64:279.
8 1 A Brief History of Gout

uric acid by adenine-8-C14 in normal and gouty 48. Kelley WN, Greene ML, Rosenbloom JF, et al.
subjects. J Clin Invest. 1968;47:1193. A specific enzyme defect in gout associated with
43. Kelley WN, Rosenbloom FM, Seegmiller JE. The overproduction of uric acid. Proc Natl Acad Sci USA.
effect of azathioprine (Imuran) on purine synthesis in 1967;57:1735.
clinical disorders of purine metabolism. J Clin Invest. 49. Kelley WN, Greene ML, Rosenbloom JF, et al.
1967;46:1518. Hypoxanthine-guanine phosphoribosyltransferase
44. Seegmiller JE, Laster L, Stetten Jr D. Incorporation of deficiency in gout. Ann Intern Med. 1969;70:155.
4-amino-5-imidazolecarboxamide-4-C13 into uric acid 50. Sperling O, Eilam G, Persky-Brosh S, et al.
in the normal human. J Biol Chem. 1955;216:653. Accelerated erythrocyte 5’-phosphoribosylpyrophos-
45. Lesch M, Nyhan WL. A familial disorder of uric acid phate synthesis. A familial abnormality associated
metabolism and central nervous system function. Am with excessive uric acid production and gout. Biochem
J Med. 1964;36:561. Med. 1972;6:310.
46. Shapiro SL, Sheppard GL, Dreifuss FE, Newcombe 51. Sperling O, Persky-Brosh S, Boer P, et al. Human
DS. X-linked recessive pattern of inheritance of a syn- erythrocyte phosphoribosylpyrophosphate synthetase
drome of mental retardation with hyperuricemia. Proc mutationally altered in regulatory properties. Biochem
Soc Exp Biol Med. 1966;122:609. Med. 1973;7:389.
47. Nyhan WL, Pesek J, Sweetman L, et al. Genetics of
an X-linked disorder of uric acid metabolism and
cerebral function. Pediatr Res. 1967;1:5.
The Prevalence and Risk Factors
for Gout 2

Prevalence of Gout Diet, Disease States, and Heredity


Associated with Gout
Gout is the most common inflammatory rheu-
matic disease, at least in the Western world, and Scientific research has now changed the way uric
several surveys have shown that the prevalence acid is characterized as to its role in the human,
of gout continues to increase into the twenty- and this new evidence has altered the clinical
first century. Estimates from the United States assessment and follow-up evaluations in patients
National Health and Nutrition Examination with gout. Originally, uric acid was considered
Survey (NHANES III) showed that the preva- a nitrogenous waste product of human metabo-
lence of gout in the USA more than doubled in lism that was excreted by the kidney and bowel.
the last half of the twentieth century. And further Despite a host of hypotheses generated to the
studies indicate the prevalence of gout continues contrary, this nonfunctional concept of uric acid
to increase. A study of data from the NHANES has remained in vogue until recent studies have
2007–2008 estimated that the prevalence of gout begun to provide the scientific evidence with
among US adults was 3.9 % or 8.3 million indi- respect to the potential biological functions
viduals. Among men, the prevalence was 5.9 % of uric acid [3–7]. A variety of investigations
and among women 2.0 %. The prevalence of have now linked the causes of hypertension, salt
hyperuricemia was also increasing. This sur- retention, hypertriglyceridemia, cardiovascular
vey led to the conclusions that over the past two adverse events, and obesity to increased serum
decades, the prevalence of gout had increased by uric acid levels [3, 8–40]. In addition to clinical
1.2 % and hyperuricemia by 3.2 %. If hyperu- data implicating uric acid levels with cardiovas-
ricemia is defined as levels over 7.0 mg/dl, the cular events like hypertension, the pathological
prevalence of hyperuricemia was 13.2 % of US mechanisms for such associations including the
adults. Most of these increases were attributed to development of renal disease, renin-angiotensin
the increasing prevalences of obesity and hyper- system activation, and endothelial dysfunction
tension [1]. Moreover, the increasing prevalence have been characterized [12, 14–18, 27, 41–48].
has been found in several countries other than the Although the data relating to the generation of
USA. A survey in the UK found that the preva- reactive oxygen species (ROS) and nitric oxide
lence of gout there had increased threefold in the and the production of endothelial dysfunction
two decades prior to 1991, and similar studies remains controversial, there is an increasing body
from New Zealand, China, and African countries of evidence that links hyperuricemia with hyper-
have also found similar increases [1, 2]. tension and renal damage [11, 12, 27, 41, 49–52].

D.S. Newcombe, Gout, 9


DOI 10.1007/978-1-4471-4264-5_2, © Springer-Verlag London 2013
10 2 The Prevalence and Risk Factors for Gout

Despite the fact that absolute correlations do not is strikingly high (62.8 % in US adults and 43.6 %
exist between uric acid and hypertension and its in Korean adults) as compared with a general
pathologic mechanisms, sufficient data do exist population without gout (25.4 % in the USA and
to encourage the careful monitoring for the onset 5.2 % in Korea) [71, 72]. This association between
of hypertension in gouty patients. Furthermore, gout and the metabolic syndrome has been
evidence also suggests that the hyperuricemic confirmed in other studies as well [73, 74].
state accompanying gout should be treated with Cardiovascular disease in gouty patients at high
hypouricemic agents along with the aggressive risk, those with type 2 diabetes mellitus, stroke,
management of hypertension [53]. Although uric congestive heart failure, or coronary heart dis-
acid and hyperuricemia have been associated ease, showed an independent association of
with these morbidities, the role of uric acid in the gout with cardiovascular disease and mortality
pathogenesis of these disease states remains to be [75, 76]. Low-risk gouty patients without these
determined in most cases. However, some evi- accompanying findings have determined that
dence does indicate that gout is an independent serum uric acid levels are poor predictors of car-
risk factor for cardiovascular disease [2, 54]. diovascular morbidity and mortality [36, 76].
Gout has also been documented to be associ- There are a number of publications that address
ated with obesity, dyslipidemia, and hyperglyce- components and complications of the metabolic
mia along with hypertension [55–59]. With syndrome in connection with the risk of vascular
respect to obesity and the insulin resistance syn- disease, the occurrence of insulin resistance, the
drome, elevated serum uric acid levels have presence of ischemic heart disease, cardiovascu-
been observed with these clinical findings as well lar mortality, and the presence of accompanying
[60–62]. Further evidence of the association disorders at the time of the initial diagnosis of
between gout and obesity comes from studies gout [49, 73, 77–89]. Hyperuricemia is associ-
showing that the risk of gout diminishes with ated with an increased risk for vascular disease
weight reduction [63, 64]. Recently, obesity, in some studies, but other investigations only
hypertension, lipidemia, and hyperglycemia have demonstrate a link between gout and death from
been considered as a cluster of findings charac- cardiovascular diseases but not with hyperurice-
teristic of a syndrome of multiple interrelated mia [78, 81]. The prevalence of insulin resistance
conditions called the metabolic syndrome [65]. is higher in patients with gout than in normal
This so-called metabolic syndrome is diagnosed healthy control groups [79]. Patients with gout in
on the basis of the following revised criteria: one study were all shown to manifest the meta-
abdominal obesity (waist circumference of >102 bolic syndrome, and 16 % had ischemic heart
cm in men and >88 cm in women), hypertriglyc- disease [77]. A recent review of the clinical sta-
eridemia (>150 mg/dl or 1.69 mmol/l), low high- tus of patients with respect to their first episode
density lipoprotein (HDL) cholesterol (<40 mg/ of gout and the diagnosis of the metabolic syn-
dl or 1.04 mmol/l in men and <50 mg/dl or 1.29 drome and its components and complications
mmol/l in women), a blood pressure reading of showed that 90 % of the first attacks of gout pre-
less than 130/80 mm of HG, and a fasting glucose ceded the diagnosis of the metabolic syndrome,
level of more than 100 mg/dl or more than 5.6 its components, and its complications [80]. At
mmol/l [66]. Two facts stress the importance of the time of the inclusion of gouty patients into
identifying the metabolic syndrome in gouty this study (a mean of 13.7 years after the first
patients. First, the metabolic syndrome increases attack of gout), 93 % of these patients had at least
the risk of cardiovascular disease up to three one associated disorder. The most common asso-
times and the risk for type 2 diabetes mellitus up ciated disorders included hypertriglyceridemia,
to five times [67, 68]. It also causes an increase in 63 %; obesity, 54 %; hypertension, 45.6 %; meta-
mortality from cardiovascular diseases as well as bolic syndrome, 40 %; hyperglycemia, 37 %;
all-cause mortality [60–70]. Second, the preva- low HDL, 17 %, diabetes mellitus, 15 %, chronic
lence of the metabolic syndrome in gouty patients renal failure, 17 %; and ischemic heart disease,
Diet, Disease States, and Heredity Associated with Gout 11

6.6 %. These data emphasize the need for care- gout [31, 95, 96]. Fructose-induced hyperurice-
ful follow-up evaluations of patients with gout. mia has been known since the 1970s, but it has
Another study showed a 2:1 ratio of the preva- only recently been implicated in the causation of
lence of essential hypertension, hyperlipidemia, the metabolic syndrome and gout [97–100]. Two
diabetes mellitus without complications, and mechanisms have been proposed as the means by
coronary atherosclerosis in patients with gout which fructose causes the hyperuricemia of the
as compared with subjects without gout [90]. A metabolic syndrome. The need for fructose to be
strong correlation has also been shown to exist phosphorylated by ATP during its metabolism to
between diminished urinary uric acid levels and form fructose-1-phosphate leads to the break-
the presence of the metabolic syndrome [89]. down of ATP and ultimate production of uric
The proposed mechanism for this abnormality of acid. The presence of low phosphorus levels
uric acid excretion is an impairment of renal uric impedes the regeneration of ATP and favors the
acid excretion mediated by hyperinsulinemia- degradation of adenosine monophosphate. The
enhanced proximal tubule sodium reabsorption second mechanism postulates that tissue isch-
and the diminished uric acid excretion [49, 88]. emia leads to the degradation of ATP causing an
Although medications are usually the means increase in uric acid synthesis. In addition, the
by which recurrent gouty episodes are controlled, enzyme, xanthine dehydrogenase, is converted to
there are certain dietary restrictions that may be xanthine oxidase in the ischemic state, and the
useful as well. It has been shown that meat and latter enzyme uses molecular oxygen as an elec-
seafood intake are associated with an increased tron donor leading to the formation of superoxide
risk of gout. Meat including beef, lamb, and pork anion and hydrogen peroxide (oxygen free radi-
as well as bologna, sausage, salami, bacon, hot cals). Thus, tissue ischemia leads to an increased
dogs, chicken, turkey, hamburger, chicken liver, production of both oxygen free radicals and uric
and beef liver are the most well-characterized acid. Recent data have also shown that excessive
offenders as inducers of gouty episodes. Seafood fructose ingestion causes not only the metabolic
associated with an increase risk of gout include syndrome but also a hypertensive response [31].
tuna, dark fish, shrimp, lobster, clams, and scal- In the foregoing study, reducing uric acid levels
lops. There are also some purine-rich vegetables also decreases the blood pressure, and this is per-
like peas, lentils, spinach, mushrooms, oatmeal, tinent to the data showing that hyperuricemia
and cauliflower, but they do not appear to increase may have a role in the production of hypertension
the risk of gout [56, 91–94]. In contrast these in man. Fructose ingestion has also been associ-
high-purine-containing foods, there are some ated with an increased risk of gout and kidney
foods with low purine content such as milk, stones as well as its capacity to increase post-
cheese, yogurt, ice cream, breads, pasta, vegeta- prandial levels of triglycerides [95].
bles other than those with a high purine content, Two studies have examined the dietary intake
fruits, nuts, sugars, and sweets. Of course, it is of fructose as it relates to the risks of the devel-
important to recognize that beef, cheese, ice opment of gout [101, 102]. Sugar-sweetened
cream, and some other foods are not appropriate soft drink ingestion significantly increases the
for those with lipid abnormalities. Usually, gout serum uric acid levels, and such increments
is associated with type IV hyperlipidemia or were entirely independent of age, sex, body
hypertriglyceridemia. These dyslipidemias occur mass index, alcohol use, renal function, hyper-
in about 25–60 % of gouty patients [55, 56]. It is tension, and diuretic use. For example, the dif-
also worth remembering that roughly two-thirds ference between the serum uric acid levels and
of the purines in the human are produced by cell the extreme categories of sugar-sweetened soft
and tissue turnover, whereas only one-third of drink intake was 0.4 mg/dl. The latter increment
purines are contributed by dietary intake. in serum uric acid level was equal to the con-
Excessive fructose intake has been implicated sumption of one beer/day and caused a 50 %
in the causation of the metabolic syndrome and increase in the risk of incident gout [103, 104].
12 2 The Prevalence and Risk Factors for Gout

In fact, another study showed the risk of incident taken after a 12-h fast. Patients who have abnor-
gout was 85 % higher among men who ingested mal blood lipids should reduce their alcohol use,
two or more servings of sugar-sweetened soft should institute a weight reducing diet contain-
drinks daily as compared to those who con- ing only 10–20 % fat, and be treated with either
sumed less than one serving per month [102]. fenofibrate or gemfibrozil to reduce their trig-
Furthermore, fructose-induced hyperuricemia is lyceride levels. These dietary restrictions should
greater in those patients with gout or hyperuri- also be accompanied by an exercise program to
cemia [105–108]. These data suggest that increase HDL levels. Treatment regimens for
patients with gout might improve their health hypertriglyceridemia and the risk of mortality
and well-being by avoiding the excessive use of have been published and can be consulted
sugar-sweetened soft drinks or shift to diet soft for additional information [123–128]. Finally,
drinks that do not increase serum uric acid lev- blood sugar levels should be maintained below
els [101]. There is another means by which fruc- 100 mg/dl.
tose intake may also affect humans since it Alcohol ingestion in excess and its association
contributes to insulin resistance, glucose intol- with gout has been known since antiquity [56,
erance, and hyperinsulinemia [109–113]. In 129–132]. Beer has a special capacity to raise the
summary, fructose intake increases insulin lev- serum uric acid levels and put patients at risk for
els, enhances insulin resistance, fosters obesity, gouty episodes since it contains guanosine, a
and causes significant increments in serum uric purine that is easily converted enzymatically to
acid levels, especially in gouty patients [57, 71, uric acid. Finally, many patients will tell their
114, 115]. In animal studies, female sex hor- physician that particular foods or alcoholic bev-
mones protect women against fructose-induced erages will trigger an attack of gout. In these
hyperuricemia, but hyperinsulinemia causes a cases, physicians should heed the patient’s infor-
decrease in renal urate excretion and is corre- mation and ask the patient to restrict their intake
lated with the increase in serum uric acid levels of the offending agent (Fig. 2.1).
[55, 116–120]. One final point should be made There are four general rules that are useful
regarding the metabolic syndrome. A recent when considering dietary modifications for
study has proposed that hyperuricemia is patients with gout. Overweight patients should
strongly associated with the metabolic syn- make a concerted effort to lose weight by diet and
drome and should be included as a parameter of exercise. Measurement of fasting blood lipids is
the metabolic syndrome [121]. This fact coupled essential to exclude hyperlipidemia as a contrib-
with the frequency of the metabolic syndrome uting factor to underlying health problems.
in patients with gout (63 %) provides the impe- Appropriate diets and medications should be
tus to assess and monitor patients with gout for instituted to decrease the low-density lipoprotein
the metabolic syndrome [71, 122]. Therefore, it (LDL) levels (bad cholesterol) and increase the
is recommended that blood pressure levels of high-density lipoprotein (HDL) levels (good cho-
greater than 130/80 mmHg be treated aggres- lesterol) if hyperlipidemia exists. Gouty patients
sively to maintain blood pressure levels lower should also be encouraged to eat a balanced diet
than this. Abdominal obesity with a waist cir- with less red meat and less seafood than usual. In
cumference of greater than 102 cm in men or general, the three foods/beverages that place
greater than 88 cm in women should be treated patients at risk for gout are red meat, beer, and
with weight reduction. Patients with hypertrig- fructose, a sugar commonly found in sugar-
lyceridemia who manifest triglyceride levels of sweetened soft drinks. Finally, patients with gout
greater than 150 mg/dl and/or low high-density should decrease their use of hard liquor and beer
lipoprotein levels (less than 40 mg/dl in men with a substitution of wine if necessary.
and less than 50 mg/dl in women) should be Hyperuricemia is the precursor of gout and
treated to alter these elevated lipid levels. Blood kidney stones, and therefore, the mechanisms
lipid levels should always be measured in samples that increase serum uric acid levels become
Genetic Abnormalities in Renal Tubular Transport Leading to Reduced Renal Excretion of Urate 13

Fig. 2.1 Champaign driving away real pain (Reproduced with permission from the American College of
Rheumatology)

important to understand and modify, if necessary. overproduction, and many studies have indicated
Serum uric acid levels represent a balance genetic abnormalities of several renal tubular
between cell and tissue breakdown, the synthesis proteins that are involved in urate transport in
of uric acid and its precursors by the de novo addition to the lifestyle issues discussed above.
purine and salvage pathways, the renal elimina-
tion of urate, and the dietary intake of purines and
other chemicals known to influence urinary uric Genetic Abnormalities in Renal
acid excretion. Genetic parameters have been Tubular Transport Leading to
described in monogenic disorders including Reduced Renal Excretion of Urate
hypoxanthine-guanine phosphoribosyltransferase
deficiency, phosphoribosylpyrophosphate syn- Recently, genome-wide association studies have
thetase overactivity, familial juvenile hyperurice- been applied to assess serum uric acid concentra-
mic nephropathy, and glycogen storage disease, tions [133–140]. Studies have begun to link an
but these disorders account for only a very small excessive fructose intake, hyperuricemia, and a
proportion of patients with gout. The majority of transport system in the kidney. The gene involved
patients with gout have a drug-induced or in this process, SLC2A9, encodes a glucose/fruc-
undefined alteration in their renal elimination of tose transporter [133, 138–146]. This gene con-
uric acid. Most gout therefore is associated with tains 14 exons and 12 transmembrane helical
reduced renal excretion of uric acid rather than domains and is highly expressed in the liver and
14 2 The Prevalence and Risk Factors for Gout

kidney [147, 148]. There are two isoforms of this mutation in the SLC2A9 gene (heterozygous C to
gene, a long isoform (GLUT9L) and a short iso- G alteration in nucleotide 1296 in exon 11 leads
form (GLUT9S), and the gene itself is localized to a change from proline 412 to arginine 412 –
to chromosome 4p15.3-16 [144]. The gene that CCC to CGC). When examined in experimental
maps to chromosome 4p15.3-16 consists of 12 systems, this mutant significantly reduced urate
exons and encodes a 540 amino acid protein transport activity. The foregoing data leave under
[147]. A splice variant of this gene has also been consideration mutant apical renal membrane sites
cloned from human kidney cDNA, and this vari- for SLC2A9, mutant basolateral membrane sites
ant consists of 13 exons and encodes a protein of for SLC2A9, or single nucleotide polymorphisms
512 amino acids. This splice variant has been of this gene as putative mediators of hyperurice-
termed GLUT9DN to delineate that GLUT9 and mia [147, 150, 151].
GLUT9DN only differ in their N terminus. Genetic polymorphisms of SLC2A9/GLUT9
GLUT9 was found to be present in the kidney, have been identified from genome-wide associa-
placenta, liver, and leukocytes, whereas tion studies either by genetic variations in the
GLUT9DN has been localized to the kidney and human genome sequence, single nucleotide poly-
placenta. In the Madrin-Darby canine kidney, morphisms (SNPs), or HapMap databases to be
GLUT9 was localized in the basolateral mem- related to low uric acid levels or low fractional
brane of the proximal renal tubule epithelial cells, clearance of urate by the kidney (elevated serum
and GLUT9DN localizes to the apical membrane uric acid levels) [133, 135, 137, 138, 140, 143].
of the proximal tubule epithelial cells. The SNPs associated with gout include the fol-
Recent studies using GLUT9 isoforms 1 and 2 lowing: rs 12510549, rs 1014290, rs 6449213, rs
have proposed that SLC2A9 (isoform 2) is cou- 7442295, rs 6855991, rs 737267, and rs 16890979.
pled with URAT1, the principal renal urate reab- The long isoform of GLUT9 (GLUT9L) is local-
sorption transporter, to cause the excretion of ized to the basolateral membranes of the proxi-
urate from the tubule cells [149]. GLUT9, in these mal tubule epithelial cells, whereas the short
experiments, manifested saturable kinetics, and splice variant (GLUT9S) is localized to the apical
the Eadie-Hofstee plot showed a Km value of membranes of the proximal tubule epithelial
365 ± 42 uM and a Vmax value of 5,521 ± 291 cells. It is now well accepted that GLUT9L is the
pmol/h/oocyte (mean ± S.E.). Experimental data principal mediator of urate transport from the
supported a voltage sensitivity transport system intracellular compartment of the renal tubular
when external potassium ions depolarized the cell epithelial cells to the interstitium/blood space,
membrane and the net negative charge remained and it is postulated that GLUT9S is responsible
with the cell. The uptake of urate by GLUT9 was for the secretion of urate into the lumen of the
not influenced by glucose, fructose, lactate, nico- renal tubules. These SNPs associated with gout
tinamide, orotate, or ketone bodies like acetoace- showed no significant associations with coronary
tate or b-hydroxybutyrate. However, probenecid, artery disease or myocardial infarction [146].
benzbromarone, and losartan did manifest mod- Additional studies of New Zealand populations
erate inhibition of urate uptake. These data sug- of Maoris and Pacific Islanders also showed a
gest that GLUT9 confined to the basolateral strong correlation between specific SNPs and
membrane of the proximal tubules may be a new gout [152]. The SNPs evaluated for this associa-
target for uricosuric agents. As is known, muta- tion were the following: rs 16890979, rs 5028843,
tions in URAT1 can result in a hereditary disease rs 11942223, and rs 12510549. However, the data
characterized by an inborn error in kidney cell published in this study suggest that additional
membrane transport leading to a defect in urate parameters may also contribute to the increased
reabsorption and resulting in hypouricemia. frequency of gout in these population groups
However, not all patients manifesting idiopathic known to have an increased frequency of gout.
hypouricemia express mutations in URAT1. The A recent study has also related glucose and
recent report of a hypouricemic patient with a fructose transport to the SLC2A9 gene [145].
Genetic Abnormalities in Renal Tubular Transport Leading to Reduced Renal Excretion of Urate 15

These investigations evaluated two variants of of some interest to indicate that IL-1b, the
SLC2A9 (SNPs – rs 7442295 and rs 13113918). proinflammatory cytokine produced by the
These SNPs were delineated as SLC2A9a and inflammasome in gout, stimulates the transport
SLC2A9b in the publication. Both of these SNPs of glucose in articular cartilage. Since GLUT9
demonstrated urate fluxes with identical kinetic mediates urate transport at very high rates and at
analyses and provided a Km value of 981 uM and rates significantly faster than its facilitated trans-
a Vmax value of 304 pmol/oocyte/20 min. port of either glucose or fructose, this SLC2A9
Additional experiments were done to determine gene product transporter may be a key to the
whether any compounds altered urate transport. deposition of monosodium urate crystals in the
Benzbromarone showed a dose–response inhibi- joint space and cartilage. In addition, since IL-1b
tion of urate uptake between concentrations of enhances glucose transport, it would be of inter-
10 uM (34 % inhibition) and 100 uM (80 % est to determine what effect this proinflammatory
inhibition), and a Dixon plot for benzbromarone cytokine might have on urate transport by carti-
inhibition of urate transport by SLC2A9 showed lage via this protein transporter.
a Ki value of 27 uM. The only other inhibitor of Although the identification of monogenic
urate transport was determined to be lactate, and mutations in the human has clearly delineated
1 mM concentrations of this metabolite were the biochemical rationales behind the increase in
found to be a very weak inhibitor of urate trans- serum uric acid levels associated with these
port. Benemid, pyruvate, butyrate, and phloretin human mutations, there is no question that serum
(a hexose transport inhibitor) caused no inhibi- uric acid levels may also be dependent, in many
tion of urate transport. cases, on the renal elimination of uric acid. This
An evaluation of hexose transport (glucose renal determinant becomes increasingly impor-
and fructose) by SLC2A9 showed that urate tant since it contributes to the development of
transport was 40- to 60-fold faster than hexose gout and kidney stones, but it is also linked to the
transport. When urate uptake was examined in metabolic syndrome and cardiovascular disease
the face of d-glucose, d-fructose, and l-glucose, [50, 157–159]. The renal transport of urate has
the presence of glucose or fructose accelerated been found to be mediated by transmembrane
urate transport. d-glucose increased urate trans- proteins localized to the proximal renal tubular
port by sevenfold, whereas fructose enhanced cells of the kidney [159–161]. The direction of
urate transport by threefold. Thus, sugars like urate transport in the kidney is primarily depen-
fructose enhance urate transport. These experi- dent on the concentration gradients of the trans-
ments also determined that glucose and fructose ported substrates and their countertransported
bind to a different site on the transporter protein substrates even though transporter proteins such
than urate. These data relating fructose and urate as URAT1 are described as being primarily
transport support population studies showing that responsible for urate reabsorption by the proxi-
the consumption of sugar-sweetened soft drinks mal renal tubules [160]. However, mutations in
and fructose are strongly associated with elevated the transport proteins of the kidney and single
serum uric acid levels and an increased risk of nucleotide polymorphisms in these same trans-
gouty arthritis [101, 102, 153]. Finally, in addi- port proteins have begun to characterize the roles
tion to gout, excessive fructose intake has also these transmembrane proteins play in the gener-
been implicated as a cause of the metabolic syn- ation of hyperuricemia and gout. In addition to
drome in association with gout. One final point the SNPs associated with gout in the SLC2A9
about the protein product of the SLC2A9 gene is gene as discussed previously, there are other
the fact that this gene is also expressed in chon- mutations in the renal transport proteins as well
drocytes [140, 147, 150, 154–156]. This capacity as SNPs that are associated with hyperuricemia
of cartilage to transport urate may contribute to and gout. A homozygous frameshift mutation
the clinical expression of gout since cartilage is has been described in URAT1 (SLC22A12 gene)
often the site of urate crystal deposition. It is also in an individual that manifested hyperuricemia
16 2 The Prevalence and Risk Factors for Gout

and gout in contrast to the usual function of trations and is also presumed to function as a
URAT1 as principally a transporter of urate reab- urate secretor in the kidney [136, 170, 171]. This
sorption in exchange for lactate at the apical T806C genotype in the voltage-dependent urate
membrane of the proximal tubules [162]. transporter NPT1 was found to be associated with
Furthermore, defects in URAT1 cause hypouri- a significant degree of weight loss in obese
cemia because the kidney cannot reabsorb urate Japanese men [172]. The authors of the foregoing
[163–165]. Thus, these are two different muta- publication also suggest that SLC17A1 polymor-
tions, one of which enhances urate urinary excre- phisms are associated with the development of
tion through its failure to reabsorb urate leading gout. Thus, these SNPs and mutations in the
to hypouricemia and the other appears to enhance transmembrane proteins localized to the proxi-
urate reabsorption leading to hyperuricemia and mal tubules of the kidney demonstrate clearly
gout. Several drugs and metabolites effect the that understanding the mechanisms of urate trans-
excretion of uric acid through interactions with port in the kidney are likely to make inroads into
URAT1, leading to either urate retention or the genetics of gout and emphasize the significant
increased urate excretion. For example, lactate, contributions of the kidney to the generation of
nicotinate, and pyrazinoate interact with URAT1 hyperuricemia and gout. Such studies related to
to increase the reabsorption of uric acid. On the the renal transport of urate also provide new tar-
other hand, the uricosuric agents, benzbroma- gets for the design of drugs for the treatment of
rone, probenecid, and losartan, interact with hyperuricemia. The further definition of SNPs
URAT1 to inhibit urate reabsorption and thus related to hyperuricemia and gout will also lead
increase urate excretion [166]. to a better understanding of at-risk individuals
In addition to these mutations causing an and populations. Finally, more emphasis has been
alteration in the renal transport of urate, there are placed on the exclusion of parameters associated
functional polymorphisms associated with hype- with the metabolic syndrome because of its high
ruricemia and gout. The GLUT9 or SLC2A9 prevalence in gouty patients. Components of the
gene associated with glucose and fructose inges- metabolic syndrome should probably be evalu-
tion has already been discussed in relation to ated annually in patients with gout and treatment
fructose intake, but other polymorphisms have instituted when appropriate.
also been characterized. The ATP-binding cas- The handling of urate by the proximal renal
sette subfamily G member 2 (ABCG2) gene has tubule is summarized in the diagram in Fig. 2.2.
been identified as influencing serum urate levels Monocarboxylates are transported from the
and the occurrence of gout [137, 167]. This trans- lumen into the cell through the monocarboxylate
porter causes the export of uric acid out of the transporters SLC5A8 and SLC 5A12. Urate is
proximal tubules and decreases the intracellular transported into the cell through URAT1 in
urate concentration over time [168]. The intro- exchange for monocarboxylates and in exchange
duction of the mutation Q141K (glutamine-141 for dicarboxylates by OAT4. Urate also enters the
lysine) in the ABCG2 encodes a common SNP cell through SLC2A9v2 and is transported from
(rs 2231142), and this polymorphism causes a the cell into the blood through SLC2A9v1. Urate
54 % reduction in the urate transport rates com- may be secreted into the lumen by MRP4 and
pared to the wild-type ABCG2 gene. These inves- OATv1, and OAT 1 and OAT 3 may also be
tigations also determined that about 10 % of the involved in urate transport. As noted above, the
individuals with gout in white males could be presence of glucose and fructose may facilitate
attributed to this polymorphism. This SNP also the tubular reabsorption of urate, through the
appears to be responsible for gout in the Chinese SCLA transporters, and this may contribute to
Han male population [169]. This same polymor- the association with excessive intakes of these
phism also accounts for much of the gout in Asian substances with gout.
populations. Another SNP involves the SLC17A1 A report of a meta-analysis of genome-wide
gene that is associated with serum urate concen- association studies (GWAS) found significant
Genetic Abnormalities in Renal Tubular Transport Leading to Reduced Renal Excretion of Urate 17

The uric acid transportasome.

Basolateral Apical

Uric acid
reabsorption

Na+
SLC5A8
SLC5A12
Monocarboxylate Monocarboxylate
Uric acid
URAT1
Uric acid
OAT1
OAT3 Uric acid
Dicarboxylates Dicarboxylate OAT4
SLC13A3
Na+
Uric acid
SLC2A9v2 Glucose
Fructose
Uric acid
Glucose SLC2A9v1
Frucose Uric acid
MRP4 secretion
Uric acid
OATv1
(NPT1)

Fig. 2.2 The uric acid transportasome. Current under- through basolateral SLC2A9v1. For efflux of uric acid
standing of uric acid transport in the proximal renal tubule. into the lumen, MRP4 and a voltage-driven organic anion
Monocarboxylates accumulate in the tubular cell through transporter (vOAT1/NPT1) are candidates. OAT1 and
sodium-dependent monocarboxylate transporters SLC5A8 OAT3 are known to transport uric acid, although the direc-
and SLC5A12 and dicarboxylates through SLC13A3. tion of transport is not clear (Reproduced with permission
Uric acid enters the cell in exchange for monocarboxylate from Dalbeth and Merriman [173]. ©The Author 2008.
via apical URAT1 and for dicarboxylate via apical OAT4. Published by Oxford University Press on behalf of the
Apical SLC2A9v2 plays a significant role in uric acid British Society for Rheumatology)
reabsorption, the reabsorbed uric acid exiting the cell

associations of single nucleotide polymorphisms but not with blood pressure, chronic kidney dis-
at eight genetic loci with serum uric acid levels. ease, or coronary heart disease [173]. These
However, only two loci, one in SLC2A9 and one observations suggest that some genetic factors
in ABCG2 showed significant associations with that predispose to gout are not responsible for
gout. A genetic urate score showed significant other disease states that are often associated
associations with serum urate levels and gout, with gout.
18 2 The Prevalence and Risk Factors for Gout

References 19. Mercure G, Vitale C, Cerquetani E, et al. Effect of


hyperuricemia upon endothelial function in patients at
increased cardiovascular risk. Am J Cardiol. 2004;94:
1. Choi HK, Ahu Z, Mount DB. Genetics of gout. Curr
932.
Opin Rheumatol. 2010;22:144–51.
20. Corry DB, Eslami P, Yamamoto K, et al. Uric acid
2. Zhu Y, Pandya BJ, Choi HK. Prevalence of gout and
stimulates vascular smooth muscle cell proliferation
hyperuricemia in the US general population. Arthritis
and oxidative stress via the vascular renin-angiotensin
Rheum. 2011;63:3136–41.
system. J Hypertens. 2008;26:269.
3. Watanabe S, Kang DH, Feng L, et al. Uric acid, homi-
21. Brown CM, Dulloo AG, Yepuri G, Montani JP.
noid evolution, and the pathogenesis of salt-sensitivity.
Fructose ingestion acutely elevates blood pressure in
Hypertension. 2002;40:355.
healthy young humans. Am J Physiol Regul Integr
4. Ames BN, Cathcart R, Schwiers E, Hochstein P. Uric
Comp Physiol. 2008;294:R730.
acid provides antioxidant defense in humans against
22. Wexler BC, Greenberg BP. Effect of increased serum
oxidant- and radical-caused aging and cancer: a
urate levels on virgin rats with no arteriosclerosis ver-
hypothesis. Proc Natl Acad Sci USA. 1981;78:6858.
sus breeder rats with preexisting arteriosclerosis.
5. Proctor P. Similar functions of uric acid and ascorbate
Metabolism. 1977;26:1309.
in man? Nature. 1970;228:868.
23. Wexler BC. Allantoxamide-induced myocardial
6. Johnson RJ, Gaucher EA, Sautin YY, et al. The plan-
necrosis in Sprague–Dawley vs spontaneously hyper-
etary biology of ascorbate and uric acid and their rela-
tensive rats. Proc Soc Exp Biol Med. 1982;170:476.
tionship with the epidemic of obesity and
24. Kanby M, Ozkarar A, Selcoki Y, et al. Effect of treat-
cardiovascular disease. Med Hypotheses. 2008;71:22.
ment of hyperuricemia with allopurinol on blood
7. Orawan E. The origin of man. Nature. 1955;175:683.
pressure, creatinine clearance, and proteinuria in
8. Alper Jr AB, Chen W, Yau L, et al. Childhood uric
patients with normal renal function. Int Urol Nephrol.
acid predicts adult blood pressure: the Bogalusa Heart
2007;39:127.
Study. Hypertension. 2005;45:34.
25. Kang DH, Park SK, Lee IK, Johnson RJ. Uric acid-
9. Mellen PB, Bleyer AJ, Erlinger TP, et al. Serum uric
induced C-reactive protein expression: implication on
acid predicts incident hypertension in a biethnic
cell proliferation and nitric oxide production of human
cohort: the Atherosclerosis Risk in Communities
vascular cells. J Am Soc Nephrol. 2005;16:3553.
Study. Hypertension. 2006;48:1037.
26. Toma I, Kang JJ, Meer EJ, Peti-Peterdi J. Uric acid
10. Perlstein TS, Gumieniak O, Williams GH, et al. Uric
triggers renin release via a macula densa-dependent
acid and the development of hypertension: the
pathway. J Am Soc Nephrol. 2007;18:156A.
Normative Aging Study. Hypertension. 2006;48:1031.
27. Khosla UM, Zharikov S, Finch JL, et al. Hyperuricemia
11. Sundstrom J, Sullivan L, D’Agostino RB, et al.
induces endothelial dysfunction. Kidney Int.
Relations of serum uric acid to longitudinal blood
2005;67:1739.
pressure tracking and hypertension incidence.
28. Talaat KM, EI-Sheikh AR. The effect of mild hyperu-
Hypertension. 2005;45:28.
ricemia on urinary transforming growth factor beta
12. Mazzali M, Hughes J, Kim YG, et al. Elevated uric
and the progression of chronic kidney disease. Am J
acid increases blood pressure in the rat by a novel
Nephrol. 2007;27:435.
crystal-independent mechanism. Hypertension. 2001;
29. Masuo K, Kawaguchi H, Mikami H, et al. Serum uric
38:1101.
acid and plasma norepinephrine concentrations pre-
13. Gruskin AB. The adolescent with essential hyperten-
dict subsequent weight gain and blood pressure eleva-
sion. Am J Kidney Dis. 1985;6:86.
tion. Hypertension. 2003;42:474.
14. Erdogan D, Gullu H, Caliskan M, et al. Relationship
30. Ouyang X, Cerillo P, Sautin Y, et al. Fructose con-
of serum uric acid to measures of endothelial function
sumption as a risk factor for non-alcohol fatty liver
and atherosclerosis in healthy adults. Int J Clin Pract.
disease. J Hepatol. 2008;48:993.
2005;59:1276.
31. Perez-Pozo SE, Schold J, Nakagawa T, et al. Excessive
15. Saito I, Saruta T, Kondo K, et al. Serum uric acid and
fructose intake induces the features of the metabolic
the renin-angiotensin system in hypertension. J Am
syndrome in healthy adult men: role of uric acid in the
Geriatr Soc. 1978;26:241.
hypertensive response. Int J Obes. 2009;84:354.
16. Zoccali C, Maio R, Mallamaci F, et al. Uric acid and
32. Feig Dl, Soletsky B, Johnson RJ. Effect of allopurinol
endothelial dysfunction in essential hypertension.
on blood pressure of adolescents with newly diag-
J Am Soc Nephrol. 2006;17:1466.
nosed essential hypertension: a randomized trial.
17. Doehner W, Schoene N, Rauchhaus M, et al. Effects
JAMA. 2009;301:270.
of xanthine oxidase inhibition with allopurinol on
33. Cannon PJ, Stason WB, Demartini FE, et al.
endothelial function and peripheral blood flow in
Hyperuricemia in primary and renal hypertension. N
hyperuricemic patients with chronic heart failure:
Engl J Med. 1966;275:457.
results from 2 placebo-controlled studies. Circulation.
34. Perlstein TS, Gumienieah O, Williams GH, et al. Uric
2002;105:2619.
acid and the development of hypertension: the
18. Farquharson CA, Butler R, Hill A, et al. Allopurinol
Normative Aging Study. Hypertension. 2006;48:1031.
improves endothelial dysfunction in chronic heart
35. Krishnan E, Svendsen K, Neaton JD, et al. MRFIT
failure. Circulation. 2002;106:221.
Research Group. Long-term cardiovascular mortality
References 19

among middle-aged men with gout. Arch Intern Med. 53. Strazzulo P, Puig JG. Uric acid and oxidative stress:
2008;168:1104. relative impact on cardiovascular risk? Nutr Metab
36. Baker JF, Krishnan E, Chen L, Schumacher HR. Cardiovasc Dis. 2007;17:409.
Serum uric acid and cardiovascular disease: recent 54. Saag KG, Choi HK. Epidemiology, risk factors, and
developments, and where do they leave us? Am J lifestyle modifications in gout. Arthritis Res Ther.
Med. 2005;118:816. 2006;8 Suppl 1:S2.
37. Karagiannis A, Mikhailidis DP, Tzimalos K, et al. 55. Schlesinger N. Dietary factors and hyperuricaemia.
Serum uric acid as an independent prediction of early Curr Pharm Res. 2005;11:4133.
death after acute stroke. Circ J. 2007;71:1120. 56. Fam AG. Gout, diet and the insulin resistance syn-
38. Syamala S, Li J, Shankar A. Association between drome. J Rheumatol. 2002;29:1350.
serum uric acid and prehypertension among US adults. 57. Choi HK, Atkinson K, Karlson EW, Curhan G.
J Hypertens. 2007;25:1583. Obesity, weight change, hypertension, diuretic use,
39. Viazzi F, Parodi D, Leoncini G, et al. Serum uric acid and risk of gout in men: the Health Professional’s
and target organ damage in primary hypertension. Follow-up Study. Arch Intern Med. 2005;165:742.
Hypertension. 2005;45:991. 58. Roubenoff R, Klag MJ, Mead LA, et al. Incidence and
40. Iwashima Y, Horio T, Kamide K, et al. Uric acid, left risk factors for gout in white men. JAMA. 1991;266:
ventricular mass index, and risk of cardiovascular disease 3004.
in essential hypertension. Hypertension. 2006;47:195. 59. Chen SY, Shen ML. Juvenile gout in Taiwan associ-
41. Feig DI, Johnson RJ. Hyperuricemia in childhood pri- ated with family history and overweight. J Rheumatol.
mary hypertension. Hypertension. 2003;42:247. 2007;34:2308.
42. Sanchez-Lozada LG, Tapia E, Soto V, et al. Treatment 60. Glynn RJ, Campion EW, Silbert JE. Trends in serum uric
with the xanthine oxidase inhibitor: febuxostat lowers acid levels 1961–1980. Arthritis Rheum. 1983;26:87.
uric acid and alleviates systemic and glomerular 61. Rathmann W, Funkhouser E, Dyer AR, Roseman JM.
hypertension in experimental hyperuricaemia. Relations of hyperuricemia with the various compo-
Nephrol Dial Transplant. 2008;23:1179. nents of the insulin resistance syndrome in young
43. Sanchez-Lozada LG, Tapia E, Santamaria J, et al. black and white adults: the Cardia Study. Coronary
Mild hyperuricemia induces glomerular hypertension Artery Risk Development in Young Adults. Ann
in normal and remnant kidney rats. Kidney Int. Epidemiol. 1998;8:250.
2005;67:237. 62. Lee J, Sparrow D, Vokonas PS, et al. Uric acid and coro-
44. Kang DH, Nakagawa T, Feng L, et al. A role for uric nary heart disease risk: evidence for a role of uric acid in
acid in the progression of renal disease. J Am Soc the obesity-insulin resistance syndrome. The Normative
Nephrol. 2002;13:2888. Aging Study. Am J Epidemiol. 1995;142:288.
45. Iseki K, Oshira S, Tozawa M, et al. Significance of 63. Dessein PH, Shipton EA, Stanwix AE, et al. Beneficial
hyperuricemia on the early detection of renal failure effects of weight loss associated with moderate calo-
in a cohort of screened subjects. Hypertens Res. rie/carbohydrate restriction, and increased propor-
2001;24:691. tional intake of protein and unsaturated fat on serum
46. Siu YP, Leung KT, Tong MK, et al. Use of allopurinol urate and lipoprotein levels in gout: a pilot study. Ann
in slowing the progression of renal disease through its Rheum Dis. 2000;59:539.
ability to lower serum uric acid level. Am J Kidney 64. Yamashita S, Matsuzawa Y, Tokunaga K, et al. Studies on
Dis. 2006;47:51. the impaired metabolism of uric acid in obese subjects:
47. Weimert NA, Tanke WF, Sims JJ. Allopurinol as a marked reduction of renal urate excretion and its improve-
cardioprotectant during coronary artery bypass graft ment by a low-calorie diet. Int J Obes. 1986;10:255.
surgery. Ann Pharmacother. 2003;37:1708. 65. Third Report of the National Cholesterol Education
48. Butler R, Morris AD, Belch JJF, et al. Allopurinol Program (NCEP) Expert Panel on detection, educa-
normalizes endothelial dysfunction in type 2 diabetics tion, and treatment of high blood cholesterol in adults
with mild hypertension. Hypertension. 2000;35:746. (adult treatment panel 111) final report. Circulation.
49. Strazzullo P, Barbato A, Galletti F, et al. Abnormalities 2002;106:3143.
of renal sodium handling in the metabolic syndrome: 66. Grundy SM, Brower Jr HB, Gleeman JI, et al.
results of the Olivetti Heart Study. J Hypertens. Definition of metabolic syndrome: report of the
2006;24:1633. National, Heart, Lung and Blood Institute/American
50. Nakagawa T, Kang DH, Feig D, et al. Unearthing uric Heart Association conference on scientific issues
acid: an ancient factor with recently found significance related to definition. Circulation. 2004;109:433.
in renal and cardiovascular disease. Kidney Int. 67. Grundy SM, Gleeman JI, Daniels SR, et al. Diagnosis
2006;69:1722. and management of the metabolic syndrome: an
51. Nieto FJ, Iribarren C, Gross MD, et al. Uric acid and American Heart Association/National Heart, Lung,
serum antioxidant capacity: a reaction to atheroscle- and Blood Institute Scientific Statement. Circulation.
rosis? Atherosclerosis. 2000;148:131. 2005;112:2735, e297, e298.
52. Zweir JL, Kuppusamy P, Lutty GA. Measurement of 68. Pearson TA, Blair SN, Daniels SR, et al. AHA
endothelial cell free radical generation: evidence for a guidelines for primary prevention of cardiovascular
central mechanism of free radical injury in postisch- disease and stroke: Consensus panel guide to com-
emic tissue. Proc Natl Acad Sci USA. 1988;85:4046. prehensive risk reduction for adult patients without
20 2 The Prevalence and Risk Factors for Gout

coronary or other atherosclerotic vascular diseases. 87. Balakrishnan VS, Coles GA, Williams JD. A role for
Circulation. 2002;106:388–391. endogenous adenosine in control of human glomeru-
69. Trevian M, Liu J, Bahsas FB, et al. Syndrome X and lar and tubular function. Am J Physiol. 1993;265:
mortality: a population-based study. Am J Epidemiol. F504.
1998;148:958. 88. Puig JG, Ruilope LM. Uric acid as a cardiovascular
70. Ford ES. Risks for all-cause mortality, cardiovascular risk factor in arterial hypertension. J Hypertens.
disease, and diabetes associated with the metabolic 1999;17:869.
syndrome. Diabetes Care. 2005;28:1769. 89. Lopez-Suarez A, Elvira-Gonzalez J, Bascunana-
71. Choi HK, Ford ES, Li C, Curhan G. Prevalence of the Quirell A, et al. Serum urate levels and urinary uric
metabolic syndrome in patients with gout: the Third acid excretion in subjects with the metabolic syn-
National Health and Nutrition Examination Survey. drome. Med Clin. 2006;126:321.
Arthritis Rheum. 2007;57:109. 90. Novak S, Melkonian AK, Patel PA, et al. Metabolic
72. Rho YH, Choi SJ, Lee YH, et al. The prevalence of syndrome-related conditions among people with and
metabolic syndrome in patients with gout: a multi- without gout: prevalence and resource use. Curr Med
center study. J Korean Med Sci. 2005;20:1029. Res Opin. 2007;23:623.
73. Puig JG, Martinez MA. Hyperuricemia, gout and the 91. Fam AG. Gout, excess calories, purines, and beyond.
metabolic syndrome. Curr Opin Rheumatol. 2008;20: Response to a urate-lowering diet. J Rheumatol.
187. 2005;32:773.
74. Chen SY, Chen CL, Shen ML. Manifestations of met- 92. Choi HK, Lin S, Curhan G. Intake of purine-rich
abolic syndrome associated with male gout in differ- foods, protein and dairy products and relationship to
ent age strata. Clin Rheumatol. 2007;26:1453. serum levels of uric acid. The Third National Health
75. Choi HK, Curhan G. Independent impact of gout on and Nutrition Examination Survey. Arthritis Rheum.
mortality and risk for coronary heart disease. 2005;52:283.
Circulation. 2007;116:894. 93. Emmerson BT. The management of gout. N Engl J
76. Krishnan E, Baker JF, Furst DE, Schumacher HR. Med. 1996;334:445.
Gout and the risk of acute myocardial infarction. 94. Gibson T, Rodgers AV, Simmonds HA, et al. A con-
Arthritis Rheum. 2006;54:2688. trolled study of diet in patients with gout. Ann
77. Vazez-Mellado J, Garcia CG, Vazquez SG, et al. Rheum Dis. 1983;42:123.
Metabolic syndrome and ischemic heart disease in 95. Bantle JP. Dietary fructose and metabolic syndrome
gout. J Clin Rheumatol. 2004;10:105. and diabetes. J Nutr. 2009;139:1263S.
78. Hjortnaes J, Algra A, Olijhoek J, et al. Serum uric acid 96. Seegmiller JE, Dixon RM, Kemp GJ, et al. Fructose-
levels and risk for vascular disease in patients with the induced aberration of metabolism in familial gout
metabolic syndrome. J Rheumatol. 2007;34:1882. identified by 31 P magnetic resonance spectroscopy.
79. Yoo HG, Lee SI, Chase HJ, et al. Prevalence of insulin Proc Natl Acad Sci USA. 1990;87:8326.
resistance and metabolic syndrome in patients with 97. Maenpaa PH, Raivio KO, Kekomaki MP. Liver ade-
gouty arthritis. Rheumatol Int. 2011;31(4):485–91. nine nucleotides: fructose-induced depletion and its
Epub 2009 Dec 20. effect on protein synthesis. Science. 1968;161:1253.
80. Hernandez-Cuevas CB, Roque LH, Huerta-Sil G, et al. 98. Bode JC, Zelder O, Rumpelt HJ, Wittkamp U.
First acute attacks commonly precede features of the Detection of liver adenosine phosphates and meta-
metabolic syndrome. J Clin Rheumatol. 2009;15:65. bolic effects of intravenous infusion of fructose or
81. Kuo CF, See LC, Luo SF, et al. Gout: an independent sorbitol in man and in the rat. Eur J Clin Invest.
risk factor for all-cause and cardiovascular mortality. 1973;3:436.
Rheumatology. 2010;49:141. 99. Hultman E, Nilsson LH, Sahlin K. Adenine nucle-
82. Mitsuhashi H, Yatsuya H, Matsushita K, et al. Uric otide content of human liver. Normal values and
acid and left ventricular hypertrophy in Japanese men. fructose-induced depletion. Scand J Clin Lab Invest.
Circ J. 2009;73:667. 1975;35:245.
83. Enomoto A, Kimura H, Chairoungdua A, et al. 100. Nakagawa T, Tuttle K, Short RA, Johnson RJ.
Molecular identification of a renal urate anion exchanger Hypothesis: fructose-induced hyperuricemia as a
that regulates blood urate levels. Nature. 2002;417:447. causal mechanism for the epidemic of the metabolic
84. Bakker SJ, Gana RO, ter Maaten JC, et al. The potential syndrome. Nat Clin Pract Nephrol. 2005;1:80.
role of adenosine in the pathophysiology of the insulin 101. Choi JW, Ford ES, Gao X, Choi HK. Sugar-
resistance syndrome. Atherosclerosis. 2001;155:283. sweetened soft drinks, diet soft drinks, and serum
85. Fransen R, Koomans HA. Adenosine and renal sodium uric acid level: the Third National Health and
handling: direct natriuresis and renal nerve-mediated Nutrition Examination Survey. Arthritis Rheum.
antinatriuresis. J Am Soc Nephrol. 1995;6:1491. 2008;59:109.
86. Balakrishnan VS, Coles GA, Williams JD. Effects of 102. Choi HK, Curhan G. Soft drinks, fructose consump-
intravenous adenosine on renal function in healthy tion, and the risk of gout in men: perspective cohort
human subjects. Am J Physiol. 1996;271:F374. study. BMJ. 2008;336:309.
References 21

103. Choi HK, Atkinson K, Karlson EW, et al. Alcohol 120. Ter Maaten JC, Voorburg A, Heine RJ, et al. Renal
intake and risk of incident gout in men: a prospective handling of urate and sodium during acute physio-
study. Lancet. 2004;363:1277. logical hyperinsulinemia in healthy subjects. Clin
104. Choi HK, Curhan G. Beer, liquor, and wine con- Sci. 1997;92:51.
sumption and serum uric acid levels: the Third 121. Lin WY, Liu CS, Li TC, et al. In addition to insulin
National Health and Nutrition Survey. Arthritis resistance and obesity, hyperuricemia is strongly
Rheum. 2005;51:1023. associated with metabolic syndrome using definitions
105. Fox IH, Kelley N. Studies of the mechanism of in Chinese populations: a population based study
fructose-induced hyperuricemia in man. Metabolism. (Taichung Community Health Study). Ann Rheum
1972;21:713. Dis. 2008;67:432.
106. Perheentupa J, Raivio K. Fructose-induced hyperuri- 122. Marasini B, Massarotti M. What rheumatologists
caemia. Lancet. 1967;2:528. should know about gout and cardiovascular disease.
107. Stirpe F, Della Corte E, Bonetti E, et al. Fructose- J Rheumatol. 2009;36:854.
induced hyperuricaemia. Lancet. 1970;2:1310. 123. Capel WH, Eckel RH. Severe hypertriglyceridemia
108. Emmerson BT. Effect of oral fructose on urate pro- with a history of treatment failure. Nat Clin Pract
duction. Ann Rheum Dis. 1974;33:276. Endocrinol Metab. 2005;1:53.
109. Blakely SR, Hallfresch J, Reiser S, Prather ES. Long- 124. Consensus conference: treatment of hypertriglyceri-
term effects of moderate fructose feeding on glucose demia. JAMA. 1984;251:1196
tolerance parameters in rats. J Nutr. 1981;111:307. 125. Yadav D, Pitchumoni CS. Issues in hyperlipidemic
110. Rizkalla SW, Boillot J, Trecottet V, et al. Effects of pancreatitis. J Clin Gastroenterol. 2003;36:54.
chronic dietary fructose with and without copper 126. Illingworth DR. Treatment of hyperlipidaemia. Br
supplementation on glycaemic control, adiposity, Med Bull. 1990;46:1025.
insulin binding to adipocytes and glomerular base- 127. Santamarina-Fojo S. The familial chylomicronemia
ment membrane thickness in normal rats. Br J Nutr. syndrome. Endocrinol Metab Clin North Am. 1998;
1993;70:199. 27:551.
111. Thorburn AW, Storlien LH, Jenkins AB, et al. 128. Baron TH, Morgan DE. Acute necrotizing pancreati-
Fructose-induced in vivo insulin resistance and ele- tis. N Engl J Med. 1999;340:1412.
vated triglyceride levels in rats. Am J Clin Nutr. 129. Snaith M. Gout and alcohol. Rheumatology.
1989;49:1155. 2004;43:1208.
112. Zavaroni I, Sander S, Scott S, Reaven GM. Effect of 130. Snaith M. A (very) short history of diets for gout.
fructose feeding on insulin secretion and insulin Rheumatology. 2004;43:1054.
action in the rat. Metabolism. 1980;29:970. 131. Johnson RJ, Rideout BA. Uric acid and diet- insights
113. Wu T, Giovannucci E, Pischon T, et al. Fructose, gly- into the epidemic of cardiovascular disease. N Engl J
cemic load, and quantity and quality of carbohydrate Med. 2004;350:1071.
in relation to plasma C-peptide concentrations in US 132. Sharpe CR. A case–control study of alcohol con-
women. Am J Clin Nutr. 2004;80:1043. sumption and drinking behavior in patients with
114. Gross LS, Li L, Ford ES, Liu S. Increased consump- acute gout. Can Med Assoc J. 1984;131:563.
tion of refined carbohydrates and the epidemic of 133. Li S, Sanna S, Maschio A, et al. The GLUT9 gene is
type 2 diabetes in the United States: an ecologic associated with serum uric acid levels in Sardinia
assessment. Am J Clin Nutr. 2004;79:774. and Chianti cohorts. PLoS Genet. 2007;3:e194.
115. Bray GA, Nielsen SJ, Popkin BM. Consumption of 134. Zumunik T, Boban M, Lane G, et al. Genome-wide
high-fructose corn syrup in beverages may play a association study of biochemical traits in Korcula
role in the epidemic of obesity. Am J Clin Nutr. Island. Croat Med J. 2009;50:23.
2007;79:537. 135. Wallace C, Newhouse SJ, Braund P, et al. Genome-
116. Galipeau D, Verma S, McNeill JH. Female rats are wide association study identifies genes for biomark-
protected against fructose-induced changes in ers of cardiovascular disease: serum urate and
metabolism and blood pressure. Am J Heart Circ dyslipidemia. Am J Hum Genet. 2008;82:139.
Physiol. 2002;283:H2478. 136. Kotz M, Johnson T, Sanna S, et al. Meta-analysis of
117. Vasudevan H, Xiang H, McNeill JH. Differential 28,141 individuals identifies common variants within
regulation of insulin resistance and hypertension by five new loci that influence uric acid concentrations.
sex hormones in fructose-fed male rats. Am J Physiol PLoS Genet. 2009;5:e1000504.
Heart Circ Physiol. 2005;289:H1335. 137. Dehghan A, Kottgen A, Yang O, et al. Association of
118. Muscelli E, Natali A, Bianchi S, et al. Effect of insu- three genetic loci with uric acid concentration and
lin on renal sodium and uric acid handling in essen- risk of gout: a genome-wide association study.
tial hypertension. Am J Hypertens. 1996;9:746. Lancet. 2008;372:1953.
119. Horton TJ, Gayles EC, Prach PA, et al. Female rats 138. Doring A, Gieger C, Mehta D, et al. SLC2AO
do not develop sucrose-induced insulin resistance. influences uric acid concentrations with pronounced
Am J Physiol. 1997;272:R1571. sex-specific effects. Nat Genet. 2008;40:430.
22 2 The Prevalence and Risk Factors for Gout

139. McArdle PF, Parsa A, Chang YP, et al. Association 153. Gao X, Oi L, Qiao N, et al. Intake of added sugar and
of a common monosynonymous variant in GLUT9 sugar-sweetened drink and serum uric acid concen-
and serum uric acid levels in old order Amish. tration in US men and women. Hypertension.
Arthritis Rheum. 2008;58:2874. 2007;50:306.
140. Vitart V, Rudan I, Haywood C, et al. SLC2A9 is a 154. Richardson S, Neama G, Phillips T, et al. Molecular
newly identified urate transporter influencing serum characterization and partial eDNA cloning of facili-
urate concentration, urate excretion and gout. Nat tative glucose transporters expressed in human artic-
Genet. 2008;40:437. ular chondrocytes: stimulation of 2- deoxyglucose
141. Preitner F, Bonny O, Laverriere A, et al. Glut9 is a uptake by IGF-1 and elevated MMP-2 secretion by
major regulator of urate homeostasis and its genetic glucose deprivation. Osteoarthritis Cartilage.
inactivation induces hyperuricosuria and urate neph- 2003;11:92.
ropathy. Proc NatlAcad Sci USA. 2009;106:15501. 155. Shikhman AR, Brinson DC, Valbracht J, Lotz MK.
142. Cheeseman C. Solute carrier family 2, member 9 and Cytokine regulation of facilitated glucose transport
uric acid homeostasis. Curr Opin Nephrol Hypertens. in human articular chondrocytes. J Immunol. 2001;
2009;18:428. 167:7001.
143. Brandstatter A, Kiechl S, Kollerits B, et al. Sex- 156. Mobasheri A, Neama G, Bell S, et al. Human articu-
specific association of the putative fructose trans- lar chondrocytes express three facilitative glucose
porter SLC2A9 variants with uric acid levels is transporter isoforms: GLUT1, GLUT3 and GLUT9.
modified by BMI. Diabetes Care. 2008;31:1662. Cell Biol Int. 2002;26:297.
144. Le MT, Shafiu M, Mu W, Johnson RJ. SLC2A9 – a 157. Wu XW, Muzny OM, Lee CC, Caskey CT. Two
fructose transporter identified as a novel uric acid independent mutational events in the loss of urate
transporter. Nephrol Dial Transplant. 2008;23:2746. oxidase during hominoid evolution. J Mol Evol.
145. Caufield MJ, Munroe PB, O’Neill D, et al. SLC2A9 1992;34:78.
is a high-capacity urate transporter in humans. PLoS 158. Feig DL, Mazzali M, Kang DH, et al. Serum uric
Med. 2008;5:e197. acid: a risk factor and a target for treatment? J Am
146. Stark K, Reinhard W, Neureuther K, et al. Association Soc Nephrol. 2007;17:569.
of common polymorphisms in GLUT9 gene with 159. Mount DB. Molecular physiology and the four-com-
gout but not with coronary artery disease in a large ponent model of renal urate transport. Curr Opin
case–control study. PLoS One. 2008;3:e1948. Nephrol Hypertens. 2005;14:460.
147. Augustin R, Carayannopoulos MO, Dowd LO, et al. 160. Hediger MA, Johnson RJ, Miyazaki H, Endou H.
Identification and characterization of human glucose Molecular physiology of urate transport. Physiology.
transporter-like protein-9 (GLUT9): alternative 2005;20:125.
splicing alters trafficking. J Biol Chem. 2004;279: 161. Rafey MA, Lipowitz MS, Leal-Pinto E, Abramson
16229. RG. Uric acid transport. Curr Opin Nephrol
148. Salas-Burgos A, Iserovich P, Zuniga F, et al. Hypertens. 2003;12:511.
Predicting the three-dimensional structure of the 162. Vasquez-Mellado J, Alvarado-Romano V, Burgos-
human facilitative glucose transporter glut1 by a Vargas R, et al. Homozygous frameshift mutation in
novel evolutionary homology strategy: insights on the SLC22A12 gene in a patient with primary gout
the molecular mechanism of substrate migration, and high levels of uric acid. J Clin Pathol.
and binding sites for glucose and inhibitory mole- 2007;60:947.
cules. Biophys J. 2004;87:2990. 163. Ichida K, Hosoyamada M, Hisatome I, et al. Clinical
149. Anzai N, Ichida K, Jutabha F, et al. Plasma urate and molecular analysis of patients with renal hypou-
level is directly regulated by a voltage-driven urate ricemia in Japan- influence of URAT1 gene on uri-
efflux transporter URATv1 (SLC2A9) in humans. nary urate excretion. J Am Soc Nephrol. 2004;15:
J Biol Chem. 2008;283:26834. 164.
150. Phay JE, Hussain HB, Moley JF. Cloning and expres- 164. Iwai N, Mino Y, Hosoyamada M, et al. A high preva-
sion analysis of a novel member of the facilitative lence of renal hypouricemia caused by inactive
glucose transporter family, SLC2A9 (GLUT9). SLC22A12 in Japanese. Kidney Int. 2004;66:935.
Genomics. 2000;66:217. 165. Taniguchi A, Urano W, Yamanaka M, et al. A com-
151. Choi HK, Zhu Y, Mount DB. Genetics of gout. Curr mon mutation in an organic anion transporter gene
Opin Rheumatol. 2010;22:144. SLC22A12, is a suppressing factor for the develop-
152. Hollis-Moffatt JE, Xu X, Dalbeth N, et al. Role of ment of gout. Arthritis Rheum. 2005;52:2576.
the urate transporter SLC2A9 gene in susceptibility 166. Zhang W, Doherty M, Bardin T, et al. EULAR evi-
to gout in New Zealand Maori, Pacific Island, and dence-based recommendations for gout. Part II:
Caucasian case–control sample sets. Arthritis management. Report of a task force of the EULAR
Rheum. 2009;60:3485. Standing Committee for International Clinical
References 23

Studies Including Therapeutics (ESCISIT). Ann renal sodium phosphate transport protein and its
Rheum Dis. 2006;65:1312. assignment to chromosome 6p21-p23. Genomics.
167. Huls M, Brown CD, Windass AS, et al. The breast 1993;18:255.
cancer resistance protein transporter ABCG2 is 171. Uchino H, Tamai I, Yamashita K, et al.
expressed in the human kidney proximal tubule api- P-aminohippuric acid transport at renal apical mem-
cal membrane. Kidney Int. 2008;73:270. brane mediated by human inorganic phosphate trans-
168. Woodward OM, Kottgen A, Coresh J, et al. porter NPT1. Biochem Biophys Res Commun.
Identification of a urate transporter, ABCG2, with a 2000;270:254.
common functional polymorphism causing gout. 172. Urano W, Taniguchi A, Anzai N, et al. Sodium-
Proc Natl Acad Sci USA. 2009;106:10338. dependent phosphate cotransporter type 1 (NPT1)
169. Wang B, Miao Z, Liu S, et al. Genetic analysis of sequence polymorphisms in male patients with gout.
ABCG2 gene C421A polymorphism with gout dis- Ann Rheum Dis. 2010;69:932.
ease in Chinese Han male population. Hum Genet. 173. Dalbeth N, Merriman T. Crystal ball gazing: new
2010;127:245. therapeutic targets for hyperuricaemia and gout.
170. Chong SS, Kristjannson K, Zoghbi HY, et al. Rheumatology. 2009;48:222–6.
Molecular cloning of the eDNA encoding a human
Purine Biochemistry
3

Structure and which then becomes position 4 in the pyrazo-


lopyrimidine ring (Fig. 3.2). This structural ana-
Emil Fischer and his colleagues were the first to log of hypoxanthine has become an important
interrelate the various purines discovered prior to therapeutic agent for gout since it lowers serum
their work and to devise the correct formula for uric acid levels (hypouricemia) through its capac-
uric acid. The purine ring is the product of a ity to inhibit the enzyme, xanthine oxidase, which
fusion between an imidazole ring and a pyrimi- catalyzes the oxidation of hypoxanthine and xan-
dine in which the carbon atoms 4 and 5 are shared. thine to uric acid (2,6,8-trioxopurine).
Its chemical name is 7(9)-H-imidazole (4,5-d) The other by-product from these studies of
pyrimidine. The purine ring and numbering sys- purine metabolism was the chemical synthesis of
tem are shown in Fig. 3.1, and the hexagonal rep- 6-mercaptopurine by the treatment of hypoxan-
resentation conforms closely to the structure thine with phosphorus pentasulfide. This purine
found by X-ray diffraction studies. Although analog was subsequently found to have antitumor
Fischer also determined the actual structure of properties and also, as will be discussed subse-
the purine base, adenine (6-aminopurine), it was quently, is a feedback inhibitor of de novo purine
not recognized that this base was a constituent of biosynthesis when converted from its free base to
deoxyribonucleic acid (DNA) until it was iso- its corresponding nucleotide. A number of gen-
lated from beef pancreas by Kossel in 1885. eral references are available for more detailed
Kossel also recognized guanine (2-amino-6- information concerning purine synthesis [1–4]
hydroxypurine) as a constituent of DNA. Further, (Fig. 3.3).
Kossel, his colleagues, and others were also
responsible for isolating the pyrimidine constitu-
ents of DNA including thymine, cytosine, uracil, Nucleic Acid Degradation
5-methylcytosine, and 5-hydroxymethylcytosine.
In studies in the late 1800s, both xanthine Gout is an inherited metabolic disorder, and
(2,6-dihydroxypurine) and hypoxanthine (6-hydro- understanding its pathogenesis requires knowl-
xypurine) were characterized. Another purine edge of both the endogenous and exogenous
base of significance to the understanding of sources of uric acid. There are three sources for
gout is the hypoxanthine analog, allopurinol which uric acid can be generated in the human:
(4-hydroxypyrazolo (3,4-d) pyrimidine). The dietary intake, tissue turnover, and de novo purine
carbon in position 8 of the purine ring and nucleotide biosynthesis from small molecules.
the nitrogen in position 7 are transposed in the Dietary purines consist mainly of free purine
allopurinol molecule, and a hydroxy group is bases and purine nucleotides that are ingested
attached to what was position 6 of the purine ring and then broken down to uric acid. Tissue turnover

D.S. Newcombe, Gout, 25


DOI 10.1007/978-1-4471-4264-5_3, © Springer-Verlag London 2013
26 3 Purine Biochemistry

Fig. 3.1 The purine ring H


is a fusion between a C
five-membered imidazole 6 N
N 5 7
ring and a six-membered 1
pyrimidine ring. The atoms 8 CH
of the purine ring are 2 4
HC 3 9
numbered as shown in the top N
figure. The pyrazolopyrimi- N
H
dine ring is numbered in a
different manner from the
purine ring as shown in the Purine ring
middle figure. The middle
figure represents the structure OH
of the hypoxanthine analog,
allopurinol. As can be seen
from the structure, the carbon
atom at position 8 and 4
N 3
nitrogen atom at position 7 of 5
the purine ring are transposed 2 N
and a hydroxy group 6 1
substituted in position 6 of 7
N
the purine ring to form N
4-hydroxypyrazolo(3,4-d) H
pyrimidine (allopurinol). The
bottom figure shows the
origin of the purine ring 4-Hydroxypyrazolo (3,4-d)pyrimidine
atoms from their parent small
molecules. N5, N10-MTHFA
is an abbreviation for N5,
N10-methenyltetrahydrofolic CO2
acid. Glycine (NH2-CH2- glycine
COOH) contributes carbon
atoms 4 and 5 as well as
nitrogen atom 7 to the purine H
ring C
N
Aspartic acid N C
5 10
CH N , N - MTHFA
5 10
N , N − MTHFA HC C
N
N
H

Amide nitrogen of glutamine

Sources of the atoms in the purine ring

represents the process by which cells and tissue chains, and by exonucleases that hydrolyze suc-
components are broken down and subsequently cessive terminal bonds. These nucleases have
renewed. The cell and tissue purine metabolites specificity for DNA, RNA, or both. The action of
from the degradative process are catabolized fur- most nucleases leads to the formation of 5¢-nucle-
ther by various enzymes to the purine end prod- otides. The general structure of such nucleotides
uct, uric acid. Nucleic acids (DNA and RNA) are is a purine base conjugated to either ribose phos-
hydrolyzed both by endonucleases, which attack phate or deoxyribose phosphate. The pathway for
chemical bonds in the middle of polynucleotide degradation of ribose nucleotides such as guanylic
Nucleic Acid Degradation 27

Fig. 3.2 Allopurinol is a Hypoxanthine and its analog allopurinol


structural analog of hypoxanthine
in which the nitrogen atom in the
seventh position and the carbon HO OH
atom in the eighth position of H
hypoxanthine are transposed. The
chemical name for allopurinol is N
N N
4-hydroxy(3,4-d)pyrimidine. It is H N
converted to oxipurinol by the
enzyme, xanthine oxidase. N H N
H N N
Oxipurinol is an analog of
xanthine and its chemical name H H
is 4, 6-dihydroxypyrazolo(3,4-d)
pyrimidine Hypoxanthine 4-Hydroxypyrazolo(3,4-d)pyrimidine

Fig. 3.3 Two of the most Structure of the purine analogs


commonly used purine
analogs are 6-mercaptopurine SH
and azathioprine. They have
both anticancer and
immunosuppressive N
activities. The rationale for N
synthesizing azathioprine was
to protect the sulfhydryl CH
group of 6-mercaptopurine
from rapid methylation and
N
oxidation. The slow liberation N
of 6-mercaptopurine from
azathioprine gives the H
molecule increased potency
as an immunosuppressive 6-Mercaptopurine
drug in comparison with
6-mercaptopurine
S C N CH3

N
N N
O2N
CH

N
N
H

Azathioprine

and adenylic acids is as follows. Adenylic acid is hypoxanthine. Hydrolytic deamination of guanine
deaminated by adenylate deaminase to inosinic forms xanthine, and oxidation of hypoxanthine
acid, and inosinic and guanylic acids are then catalyzed by xanthine oxidase converts hypoxan-
hydrolyzed by phosphomonoesterases to their thine to xanthine. Finally, xanthine oxidation by
respective nucleosides. These nucleosides are xanthine oxidase leads to the formation of uric
then cleaved by nucleoside phosphorylases to acid (Fig. 3.4a, b). Thus, excessive tissue break-
generate the free purine bases, guanine, and down as one might observe clinically with tumor
28 3 Purine Biochemistry

Nucleic acid degradation via adenine nucleotides

Nucleic acid (DNA/RNA)

Nucleases

NH2 OH

N Adenylate deaminase N
N N
CH CH

N N
N N

P-R P-R
Inosinic acid
Adenylic acid

5˙- Nucleotidase

OH OH

N Purine nucleoside N
N phosphorylase N
CH CH

N N
N N
H
Hypoxanthine R
Inosine

Xanthine oxidase

OH OH

N N
N Xanthine oxidase N
CH OH

N N
HO N N
H HO H

Uric acid
xanthine

Fig. 3.4 (a, b) Nucleic acids (DNA and RNA) are hypoxanthine to xanthine and uric acid. Guanylic acid is
hydrolyzed by deoxyribonucleases or ribonucleases and converted to guanosine by 5¢-nucleotidase, and guanosine
phosphodiesterases to 5¢-nucleotides. Adenosine mono- is then converted to its free base, guanine, by purine
phosphate is then converted to inosinic acid by adenylate nucleoside phosphorylase. Guanine deaminase converts
deaminase. Inosinic acid is subsequently converted to guanine to xanthine, and xanthine is oxidized by xanthine
inosine by 5¢-nucleotidases and to hypoxanthine by purine oxidase to uric acid
nucleoside phosphorylase. Xanthine oxidase then oxidizes
Nucleic Acid Degradation 29

Nucleic acid degradation via guanylic acid

Nucleic acids (DNA/RNA)

Nucleases

OH OH

N N
N 5′ - Nucleotidase N
CH CH

N N
N H2N N
H2N
R
P-R
Guanosine
Guanylic acid
Purine
nucleoside
phosphorylase

OH OH

N N
N Guanine deaminase N
CH CH

N N
N H2N N
HO H H

Xanthine Guanine

Xanthine oxidase

OH

N
N
OH

N
HO N
H
Uric acid

Fig. 3.4 (continued)


30 3 Purine Biochemistry

destruction by chemotherapeutic agents often (Fig. 3.6). This inversion takes place during the
leads to an excess production of uric acid. This reaction to form 5-phospho-b-d-ribosylamine
catabolic process may also be complicated by (PRA), and the resultant amino group attached to
excessive quantities of sparsely soluble uric acid ribose forms the beginning of the purine ring. The
being presented to the kidney. The inability to next step in de novo purine biosynthesis converts
excrete excessive loads of uric acid may aggra- phosphoribosylamine to 5¢-phosphoribosylg-
vate the hyperuricemia associated with increased lycineamide (GAR) by attaching a glycyl group to
rates of cell and tissue turnover. the amino group with the energy required for this
step supplied by ATP. The enzyme, phosphoribo-
sylglycineamide synthetase, catalyzes this reac-
De Novo Purine Nucleotide Synthesis tion. The next reaction adds a formyl group from
5, 10-methenyltetrahydrofolate to the free amino
The de novo synthesis of purine nucleotides is group of the glycyl residue. It is catalyzed by
another key to understanding the biochemical phosphoribosylglycineamide formyl transferase
basis of gout. The precursors of the carbon and and forms 5¢-phospho-ribosyl-N-formylglycine
nitrogen atoms in the purine are small molecules (FGAR). Another nitrogen is then transferred
that contribute to the complete biosynthesis of from the amide group of l-glutamine with the
the purine ring (Fig. 3.5a, b). Glycine contributes energy for reaction provided by ATP, and this
carbon atoms 4 and 5 and nitrogen atom 7. converts FGAR to 5¢-phosphoribosyl-N-formylg-
Formate provides carbon atoms 2 and 8, carbon lycinamidine (FGAM). This reaction is catalyzed
atom 6 arises from carbon dioxide, and nitrogen by phosphoribosyl-formylglycineamidine syn-
atoms 3 and 9 come from the amide nitrogen of thetase. Another ATP is then consumed to catalyze
glutamine. Nitrogen atom 1 comes from aspartic the closure of the imidazole ring by the enzyme,
acid. The complete synthesis of the purine nucle- phosphoribosylaminoimidazole synthetase, to form
otide components of nucleic acids, adenosine 5¢-phosphoribosyl-5-aminoimidazole (AIR). The
monophosphate and guanosine monophosphate, next enzyme in the pathway, phosphoribosylami-
requires the consumption of six high-energy noimidazole carboxylase, catalyzes the formation
phosphates in the form of adenosine triphosphate of 5¢-phosphoribosyl-5-aminoimidazole-4-carbox-
(ATP), and this requirement for high-energy ylate (CAIR) using carbon dioxide as the source
phosphate compounds is a sufficient rationale for for the carboxylate group. Subsequently, the amino
conserving purine synthesis. As will be discussed group of l-aspartate is transferred to form 5¢-phos-
subsequently, the purine salvage pathways con- phoribosyl-5-aminoimidazole-4-(N-succino) car-
vert hypoxanthine, guanine, and adenine to their boxamide (succino-AICAR) utilizing ATP as the
respective nucleotides, thus conserving both the energy to create the carboxamide. This reaction is
purine-free bases synthesized by the de novo bio- catalyzed by phosphoribosylaminoimidazole-suc-
synthetic pathway without the utilization of addi- cinocarboxamide synthetase. Adenylosuccinase
tional ATP molecules. then cleaves the molecule into fumarate and
The first irreversible reaction in purine syn- 5¢-phosphoribosyl-5-aminoimidazole-4-carbox-
thesis is the transfer of the amide group of amide (AICAR). 10-formyl-tetrahydrofolate then
l-glutamine to phosphoribosylpyrophosphate supplies the final carbon atom to the ring catalyzed
with the liberation of inorganic pyrophos- by phosphoribosylaminoimidazolecarboxamide
phate. This reaction is catalyzed by the enzyme, formyl transferase to form 5¢-phosphoribosyl-5-
phosphoribosylpyrophosphate amidotransferase. formamidoimidazole-4-carboxamide (Formyl-
5-phospho-a-d-ribosyl pyrophosphate (PRPP) AICAR). Inosinocase removes water from the
has an a-configuration, and the ribosylamine preceding molecule to form the parent purine com-
of phosphoribosylamine has a b-configuration pound, inosine-5¢-phosphate (IMP).
De Novo Purine Nucleotide Synthesis 31

De novo purine biosynthesis

2+ PP-ribose-P + AMP
A. Ribose 5-phosphate + ATP + Mg

2+
α-PP-ribose-P + glutamine + H2O + Mg β-Phosphoribosylamine + glutamic acid + PP

B. Phosphoribosylamine + glycine + ATP + Mg2+ 5’ - phosphoribosylglycineamide + ADP + Pi

5 10
Phosphoribosylglycineamide + N , N -methenyl-THFA + H2O Phosphoribosyl-α-N-
+
formylglycineamide + THFA + H

5’ -Phosphoribosylformylglycineamide + glutamine + ATP + H2O + Mg2+ 5’-


phosphoribosyl-α-N-formylglycineamidine + glutamic acid + ADP + Pi

5’ -phosphoribosylglycineamidine + ATP + Mg2+ + K+ 5’ -phosphoribosyl-5-aminoimidazole


+ ADP + Pi

5’ -phosphoribosylaminoimidazole + CO2 5’ - phosphoribosyl-5-amino-4-


imidazolecarboxylate

5’ -phosphoribosyl-5-amino-4-imidazolecarboxylate + aspartate + ATP + Mg2+ 5’ -


phosphoribosyl-5-amino-4-imidazolesuccinocarboxamixe + ADP + Pi

5’ -phosphoribosyl-amino-4-imidazolesuccinocarboxamide fumarate + 5”-


phosphoribosyl-5-amino-4-imidazolecarboxamide

10 +
5’ -phosphoribosyl-amino-4-imidazolecarboxamide + N -formyl-THFA + K 5’-
phosphoribosyl-5-formamido-4-imidazolecarboxamide
+ THFA

5’-phosphoribosyl-5-formamido-4-imidazolecarboxamide inosinic acid (IMP) + H2O

Biosynthesis of AMP and GMP

2+
Inosinic acid (IMP) + L-aspartate + GTP + Mg AMP-S + GDP + Pi

AMP-S AMP + fumarate

Inosinic acid (IMP) + NAD+ + H2O + k+


+
XMP + NADH + H

2+
XMP + glutamine + ATP + Mg GMP + glutamic acid + AMP + Pi

Fig. 3.5 (a) De novo purine biosynthesis. The second enzyme into the larger inactive form. The first thick arrow
thick arrow identifies the principal regulatory enzyme of indentifies the enzyme, PRPP synthetase, which is subject
de novo purine biosynthesis, phosphoribosylpyrophos- to mutation in humans and may result in an increase in the
phate (PRPP) amidotransferase. It is the first irreversible rate of purine synthesis. (b) The biosynthesis of adenosine
step in purine synthesis and is the site of feedback inhibi- monophosphate and guanine monophosphate. Two addi-
tion by various purine end products (AMP, GMP, and tional high energy phosphates are utilized to generate
6-mercaptopurine ribonucleotide). PRPP amidotrans- AMP and GMP from IMP. Adenosine monophosphate
ferase exists as an inactive enzyme form (molecular and guanine monophosphate serve not only as precursors
weight of 270,000) and a catalytic species (molecular of nucleic acids but also act as feedback inhibitors of
weight of 133,000). PRPP converts the 270,000 enzyme de novo purine biosynthesis. XMP stands for xanthosine
form to the catalytically active smaller form, whereas monophosphate
purine ribonucleotides convert the small molecular weight
32 3 Purine Biochemistry

De novo purine biosynthesis: key reactions

H2O3POCH2
O

+ ATP

OH

OH OH

D-Ribose-5-phosphate

2+ PRPP Synthetase
Mg

H2O3POCH2
O

O O
+ L-glutamine
O-P-O-P-OH

OH OH
OH OH

Alpha-5-phospho-D-ribosyl-1-pyrophosphate (PRPP)

2+
PRPP amidotransferase
Mg

H2O3POCH2 NH2
O

OH OH

Beta-5-Phosphoribosyl-1-amine

Fig. 3.6 The rate of purine biosynthesis is controlled by sylpyrophosphate amidotransferase into its large inactive
alterations in two enzymes, phosphoribosylpyrophosphate form. Variations in purine nucleotides, especially adenos-
synthetase and phosphoribosylpyrophosphate amidotrans- ine diphosphate (ADP) and guanosine diphosphate (GDP),
ferase. Fine control of purine biosynthesis is mediated by also competitively inhibit phosphoribosylpyrophosphate
the latter enzyme through its response to changes in purine synthetase activity with respect to Mg-adenosine triphos-
nucleotides and the concentration of phosphoribosylpyro- phate concentrations. Similarly, 2,3-diphosphoglycerate
phosphate. Phosphoribosylpyrophosphate binds to the (2,3-DPG) competitively inhibits phosphoribosylpyro-
enzyme, phosphoribosylpyrophosphate amidotransferase, phosphate synthetase activity with respect to its substrate,
and induces activation of the enzyme by converting the ribose-5-phosphate. The regulation of phosphoribosylpy-
enzyme into small active monomers, whereas purine rophosphate synthetase by purine nucleotides is less sen-
nucleotides binding at a different site from where phos- sitive than the inhibition of phosphoribosylpyrophosphate
phoribosylpyrophosphate binds converts phosphoribo- amidotransferase by purine nucleotides
Biosynthesis of AMP and GMP 33

Phosphoribosylpyrophosphate

H2O3POCH2
O

O O

O P O P OH

OH OH
OH OH

Fig. 3.7 The rate of de novo purine nucleotide biosyn- activity of the oxidative pentose phosphate pathway gen-
thesis is directly proportional to the intracellular concen- erates increased levels of ribose-5-phosphate, the immedi-
tration of phosphoribosylpyrophosphate (PRPP), and this ate precursor of PRPP. Speculation states that carbohydrate
metabolite is one of the key regulators of uric acid produc- excess and increased lipogenesis lead to an increased rate
tion via the de novo purine biosynthetic pathway. Patients of pentose phosphate shunt activity, increased ribose-5-
with phosphoribosylpyrophosphate synthetase overactiv- phosphate, and PRPP production. Such a mechanism
ity overproduce PRPP, and this increases the rate of could explain the hyperuricemia associated with hyper-
de novo purine biosynthesis and urate production. Patients triglyceridemia and obesity. Glucose-6-phosphatase
with hypoxanthine-guanine phosphoribosyltransferase deficiency and the resultant accumulation of glucose-6-
deficiency underutilize PRPP, and the resultant excess phosphate may stimulate the pentose phosphate pathway
PRPP activates the enzyme, PRPP amidotransferase, the leading to increased levels of ribose-5-phosphate and
first irreversible step in de novo purine biosynthesis lead- PRPP. Such a mechanism may contribute to the hyperuri-
ing to an acceleration in uric acid production. Excessive cemia observed in this type of glycogen storage disease

The Key Intermediate, PRPP contribute to the increased rate of purine synthe-
sis in such cells. Conversely, allopurinol, orotic
5-phosphoribosyl-1-pyrophosphate (PRPP) is acid, adenine, and 2,6-diaminopurine through
synthesized from ATP and ribose-5-phosphate their capacity to consume PRPP may reduce the
utilizing ATP as a source of energy and the intracellular concentrations of this substrate and
enzyme, ribose phosphate pyrophosphokinase reduce the rate of de novo purine biosynthesis by
(PRPP synthetase), as the catalyst to transfer diminishing the availability of PRPP for conver-
the pyrophosphate group from ATP to the one sion to PRA.
carbon of ribose-5-phosphate. PRPP has an
a-configuration (Fig. 3.7). Ribose-5-phosphate
arises from either the pentose phosphate path- Biosynthesis of AMP and GMP
way or ribose-1-phosphate. This key interme-
diate, PRPP, is a significant component of two Inosinic acid is the parent purine compound
critical reactions of purine synthesis: the conver- since it serves as the intermediate in the synthe-
sion of PRPP to phosphoribosylamine (PRA) sis of adenosine-5¢-monophosphate (AMP) and
and the conversion of free purine bases to ribo- guanosine-5¢-monophosphate (GMP) which are
nucleotides via the so-called salvage pathways. subsequently converted to nucleic acids. AMP is
When present in excess, this metabolite (PRPP) synthesized from IMP in two steps: the conversion
increases de novo purine biosynthesis through its of IMP to adenylosuccinate (N6-succinoadenosine
capacity to act as a substrate for the first irrevers- 5¢-phosphate) and the subsequent conversion of
ible step in purine synthesis, the conversion of adenylosuccinate to adenosine monophosphate
PRPP to PRA. PRPP levels are increased in cells (Fig. 3.5b).
deficient in hypoxanthine-guanine phosphoribo-
syltransferase activity [5], and these molecules IMP + L - aspartate + GTP ® AMP - S + GDP + Pi
34 3 Purine Biochemistry

This reaction is catalyzed by the enzyme, ade- be converted to AMP by the enzyme, adenosine
nylosuccinate synthetase, and requires magne- kinase, that utilizes ATP as an energy source.
sium as a cofactor. GTP is utilized as a source of Adenosine + H 2 O ® inosine + NH 3
energy. The second step is catalyzed by the
enzyme, adenylosuccinase, that also catalyzes Adenosine + ATP ® AMP + ADP
the conversion of SAICAR to AICAR. Guanine can be converted to xanthine through
the activity of the enzyme, guanase.
AMP - S « AMP + fumarate
Guanine + H 2 O ® xanthine + NH 3
The conversion of IMP to GMP also occurs in
two steps: the oxidation of IMP to xanthosine
monophosphate (XMP) and the subsequent ami- Purine Salvage Pathways
nation of XMP to form GMP. The first step is
catalyzed by the enzyme, inosine 5¢-phosphate Six ATP molecules (nine high-energy phosphate
dehydrogenase, and requires NAD+ as a hydro- groups if one includes the two ATP molecules
gen acceptor. The second step uses the amide necessary to synthesize glutamine and for the
group of l-glutamine as the specific amino donor, activation of formate) are consumed in the syn-
consumes ATP as a source of energy, and requires thesis of one molecule of inosinic acid, the parent
magnesium as a cofactor. It is catalyzed by the purine base, via the de novo purine pathway. This
enzyme, guanosine-5¢-phosphate synthetase. enormous expenditure of energy would be waste-
ful if the free bases were simply excreted in the
IMP + NAD + + H 2 O ® XMP + NADH + H + urine. For this reason, salvage pathways or purine
reutilization pathways are available to preserve
XMP + L -glutamine + ATP the purine bases synthesized by the de novo path-
® GMP + glutamic acid + AMP + PPi way. Four salvage pathways have been identified
of varying importance in the human. First, nucle-
oside phosphorylase catalyzes nucleoside forma-
Nucleotide Interconversions tion from purine bases and ribose-1-phosphate.
and Catabolism The reaction equilibrium is toward nucleoside
formation rather than nucleoside degradation.
Purine mononucleotides are converted to nucleo- Salvaged nucleosides can then be phosphorylated
sides by the action of 5¢-nucleotidases or to the nucleotide by the activity of the enzyme,
nonspecific phosphatases. purine kinase. Even though these two reactions
consume an ATP molecule, they require much
Purine mononucleotide + H 2 O fewer high-energy phosphates than the de novo
® purine nucleoside + Pi purine pathway.

Purine nucleosides, except for adenosine, are Purine base + ribose - 1 - phosphate
cleaved phosphorolytically to form a free purine « purine nucleoside
base and ribose-1-phosphate. The enzyme, nucle- Purine nucleoside + ATP
oside phosphorylase, is active with inosine or ® purine nucleotide + ADP
guanosine as substrates and slightly effective
with xanthosine as the substrate. The intermediate in the de novo pathway,
5-amino-4-imidazolecarboxamide (AIC) or
Purine nucleoside + Pi → purine base + ribose-1-P (ZMP)-5¢- monophosphate, can also be salvaged
by conversion to ZTP by the enzyme, PRPP
Adenosine is converted to inosine by the synthetase. This is the only reaction known to
enzyme, adenosine deaminase, and adenosine can favor the synthesis of a nucleoside triphosphate
Purine Salvage Pathways 35

Adenine phosphoribosyl transferase

NH2 NH2

Adenine phosphoribosyltransferase
N N
N N
PRPP + CH CH
2+
Mg
N N
N N
H
R-P
5′ - Nucleotidase

Fig. 3.8 Adenine phosphoribosyltransferase catalyzes tive inhibitor of adenine. PPi is a noncompetitive inhibi-
the salvage of adenine by its condensation with phospho- tor of both substrates. The enzyme has an absolute
ribosylpyrophosphate (PRPP) to form 5-adenosine requirement for divalent cations (Mg2+) and may catalyze
monophosphate (AMP). This enzyme is inhibited by the formation of nucleotides from specific adenine ana-
both its products, AMP and PPi. AMP is a competitive logs as well
inhibitor of the substrate, PRPP, but it is a noncompeti-

utilizing an enzyme that usually catalyzes reac- (Figs. 3.8 and 3.9). These reactions require mag-
tions that favor the synthesis of PRPP. ZTP is nesium as a cofactor.
normally not found in human erythrocytes
except in the case of individuals with a complete Purine base + PRPP
deficiency of the enzyme, hypoxanthine-gua- ® purine mononucleotide + PPi
nine phosphoribosyltransferase (Lesch-Nyhan
syndrome), in which elevated levels of ZTP Two enzymes of this type exist in the human;
have been measured [6]. These patients also one converts adenine to AMP (adenine phosphori-
excrete large quantities of 5-amino-4-carbox- bosyltransferase), and the other converts
amide in their urine [7]. Since folate is required hypoxanthine or guanine to IMP or GMP, respec-
for the conversion of this metabolite to IMP, this tively (hypoxanthine-guanine phosphoribosyl-
metabolite is also increased in folate-deficient transferase). The latter enzyme also converts
patients. 6-thioguanine and allopurinol to their respective
The adenosine cycle may be physiologically ribonucleotides. The conversion of 6-thioguanine
significant in the regulation of cellular energy or 6-mercaptopurine by this enzyme to their nucle-
and for the synthesis of adenosine utilized for cell otide forms is essential for the activity of these
receptors [8]. In this cycle, adenosine is formed antimetabolites. Thus, cells with a deficiency in
from adenosine monophosphate (AMP) through hypoxanthine-guanine phosphoribosyltransferase
the action of the enzyme, 5¢-nucleotidase, and are unaffected by these antimetabolites. In in vitro
subsequently, adenosine is phosphorylated by the studies, using cells in tissue culture can utilize this
activity of the enzyme, adenosine kinase, which property as a mechanism by which cells deficient
requires ATP as an energy source. in this enzyme can be selected. Cells with an intact
enzyme are killed in the presence of these metabo-
AMP ® adenosine + Pi lites, whereas cells deficient in this enzyme are
Adenosine + ATP ® AMP + ADP unable to convert the free base to its active nucle-
otide. Heritable deficiencies have been described
The most significant purine salvage pathways in both these enzymes, and alterations in the
in the human are those catalyzed by the purine enzyme, hypoxanthine-guanine phosphoribosyl-
phosphoribosyltransferases, adenine phosphori- transferase, are of significance to metabolic errors
bosyltransferase (APRTase), and hypoxanthine- in uric acid metabolism. In addition, patients
guanine phosphoribosyltransferase (HGPRTase) with adenine phosphoribosyltransferase deficiency
36 3 Purine Biochemistry

Ribose-5-P + ATP
3

5-Phosphoribosyl-1-Pyrophosphate (PRPP) + Glutamine

1 Fe
tion ed
ibi 5-Phosphoribosyl-1-amine ba
k inh c
ac

k
Glycine

inh
b
ed

ibi
Fe

tio
n
Formate

Nucleic acids Nucleic acid

Guanylic acid Inosinic acid Adenylic acid


7 7 7

PRPP
2 5 4
Guanosine Inosine Adenosine
P Adenine
PR

P 6
PR
PP

6
Guanine Hypoxanthine

8 2.8 dioxyadenine
Xanthine

8
Uric acid

Fig. 3.9 Summary of de novo purine synthesis and degradation

may mimic those with urate overproduction and subunits [9], and the enzyme has an absolute
renal calculus formation since the 2,8-dihydroxy- requirement for inorganic phosphorus as an allos-
adenine stones formed in this disorder react to teric activator. One of its substrates, magnesium
some test reagents in a similar fashion to the way ATP, causes aggregation of the enzyme into its
urate stones react. active, high molecular weight form (MW –
65,000–1,040,000) [10]. The enzyme possesses
three identifiable regulatory sites: a magnesium
Phosphoribosylpyrophosphate ATP site, a ribose-5-phosphate site, and a nucle-
Synthetase otide site. ADP acts as a competitive inhibitor of
magnesium ATP, and 2,3-diphosphoglycerate
Three enzymes are of cardinal importance (2,4-DPG) acts as a competitive inhibitor of
to an understanding of hyperuricemia and ribose-5-phosphate. A variety of nucleotides
gout: phosphoribosylpyrophosphate synthetase including ATP, GTP, NAD, FAD, AMP, ADP,
(PRPS), phosphoribosylpyrophosphate amido- GDP, IDP, ITP, TDP, and NADPH inhibit the
transferase, and hypoxanthine-guanine phos- enzyme in a noncompetitive manner with respect
phoribosyltransferase. The enzyme, PRPS, to its substrates. Nucleotide diphosphates and
transfers the terminal pyrophosphate group of triphosphates are the most potent inhibitors of the
ATP to the one carbon of ribose-5-phosphate and nucleotide site. This enzyme resides on the X
is subject to allosteric regulation. The human chromosome [11], and as will be discussed subse-
erythrocyte form of the enzyme has a molecular quently, mutations in this enzyme are inherited as
weight of 65,000. It is constructed of two identical X-linked recessive traits in the human (Fig. 3.10).
Phosphoribosylpyrophosphate Synthetase 37

Regulation of phosphoribosylpyrophosphate synthetase

MgATP Ribose-5-phosphate

Adenosine diphosphate 2,3-Diphosphoglycerate


(competitive inhibitor) (competitive inhibitor)

PRPP Synthetase

H2O3POCH2
O

O O

O-P-O-P-OH
OH OH
OH OH

Purine nucleotids - ADP and GDP


(noncompetitive inhibitors)

Fig. 3.10 Phosphoribosylpyrophosphate (PRPP) syn- with respect to R-5-P. Magnesium and inorganic phospho-
thetase is an allosteric enzyme that utilizes two substrates, rous are activators of the enzyme. PRPP is a substrate
magnesium adenosine triphosphate (MgATP) and ribose- used in the biosynthesis of purine, pyrimidine, and pyri-
5-phosphate (R-5-P), for the synthesis of PRPP. MgATP is dine nucleotides as well as certain synthetic compounds
essential for the aggregation and activation of the enzyme, (6-mercaptopurine, 6-thioguanine, allopurinol, and
and adenosine diphosphate (ADP) is a potent competitive azaadenine), and it is an important regulator of pyrimidine
inhibitor of this substrate. ADP and GDP are noncompeti- and purine nucleotide synthesis via orotate phosphoribo-
tive inhibitors of this enzyme. 2,3-diphosphoglycerate syltransferase and PRPP amidotransferase activities
(2,3-DPG) is a competitive inhibitor of PRPP synthetase

The product of PRPS activity, a-phosphori- phosphoribosyltransferase, and in such settings,


bosyl-1-pyrophosphate (PRPP), is an important purine biosynthesis is increased. The Km for
regulator of the first committed enzyme step in PRPP, the substrate of PRPP amidotransferase,
de novo purine synthesis, the conversion of PRPP adenine phosphoribosyltransferase, and hypox-
to phosphoribosylamine by the enzyme, PRPP anthine-guanine phosphoribosyltransferase mea-
amidotransferase. PRPP is also utilized as a sub- sured in lymphoblasts is 0.30, 0.03, and
strate in the purine salvage pathways, another 0.07 mmol/l, respectively. At physiological con-
important purine biosynthetic regulatory step. centrations, these enzymes are incompletely sat-
Phosphoribosylpyrophosphate levels are elevated urated with PRPP, the intracellular concentration
in cells deficient in hypoxanthine-guanine of which ranges between 1.0 and 5.0 umol/l [12].
38 3 Purine Biochemistry

Therefore, the increased PRPP concentrations and/or function of the nervous system. Recognition
found in erythrocytes, lymphoblasts, and fibroblasts of these disorders of overactivity and underactiv-
from patients with increased PRPS activity are ity requires measurements of enzyme activity in
accompanied by an increased rate of purine syn- both erythrocytes and fibroblasts since PRPS
thesis [13]. The rate of purine synthesis can also be activity is usually normal or low in human eryth-
reduced through the use of alternative substrates rocytes of patients with overactivity of the enzyme,
for the salvage pathway such as allopurinol, orotic while it is elevated in fibroblasts from the same
acid, or 2,6-diaminopurine. In the clinical setting, patients [18]. As noted previously, those patients
it is allopurinol that is utilized to reduce purine with a deficiency of PRPS only express the lesion
biosynthesis and uric acid production. in their erythrocytes [28, 29].
Since the first description of human mutants Evidence now supports the presence of two
with PRPS overactivity [14], more than 30 fami- major types of PRPS mutations: allosteric regula-
lies with this exceedingly rare metabolic error in tory defects and pretranslational regulatory
purine synthesis have been documented [15–22]. defects. Just as the clinical expression of this dis-
Patients with this altered enzyme usually suffer order shows heterogeneity in its clinical manifes-
from gouty arthritis and uric acid lithiasis begin- tations, the biochemical mechanisms of PRPS
ning in early adulthood [15, 18]. The clinical pre- overactivity are heterogeneous as well [30, 31]. A
sentation characterized by PRPS overactivity and variety of kinetic abnormalities of the enzyme
gouty arthritis is the adult phenotype; the infantile have been described. These allosteric regulatory
phenotype is more severe and occurs in childhood. defects include enzymes with altered affinity for
In the latter phenotype, uric acid overproduction the enzyme’s activator, phosphate, and enzymes
together with growth and motor retardation and with an increased affinity for PRPP [16–18, 23,
sensorineural deafness [15–17, 19, 21, 22] are the 25, 26]. The index cases of this PRPS overactivity
principal clinical findings. Since the genes for this were shown to have an accelerated rate of PRPP
disorder are on the X chromosome, the disease is synthesis that was due to the insensitivity of the
transmitted to hemizygous males whose X chro- enzyme to inhibition by guanosine diphosphate
mosome expresses the mutation. Heterozygous (GDP), adenosine diphosphate (ADP), adenosine
females with the adult form of the disease are monophosphate (AMP), and 2,3-diphosphoglyc-
asymptomatic [14, 18, 21, 23–26]; however, erate (2,3-DPG) [14, 31–33]. In those patients
female heterozygotes carrying the infantile phe- with allosteric regulatory defects, PRPS1 is found
notype may manifest clinical abnormalities albeit to have point mutations in its cDNA encoding
less severe than in the affected hemizygotes [27]. enzymes with altered allosteric properties [34].
Enzyme deficiencies are usually the classical These widely dispersed point mutations in the
finding in those patients with inherited metabolic X-chromosome-linked PRPS1 gene lead to dis-
abnormalities; however, PRPS overactivity is the tinctive base substitutions and resultant amino
first inherited human disorder characterized by an acid alterations in PRPS1 isoforms. Such altera-
increase in enzyme activity [27]. Interestingly tions cause changes in enzyme regulation by
enough, patients have also been documented with purine nucleotide feedback inhibition as well as
a deficiency of this same enzyme that is restricted enzyme activation by inorganic phosphate.
to the erythrocyte and not present in the fibroblasts Thus, in the case of mutations causing insensi-
of such patients [28, 29]. Patients with PRPS tivity to nucleotides, the enzyme is resistant to con-
deficiency present with hypouricemia, mental trol by intracellular nucleotides and their dampening
retardation, megaloblastic bone marrow, and effect on enzyme activity. Other individuals with
increased excretion of orotic acid. Clinical disor- defects in this enzyme include those with an
ders representing PRPS overactivity and deficiency increased affinity of phosphoribosylpyrophosphate
are both associated with abnormalities of the ner- synthetase for its substrate, ribose-5-phosphate
vous system suggesting that the enzyme and its [33]. In this setting, the enzyme catalyzes the syn-
products are likely to have a role in the development thesis of PRPP more rapidly than normal for any
Phosphoribosylpyrophosphate Synthetase 39

concentration of ribose-5-phosphate below amidotransferase activity. The latter enzyme is


enzyme-saturating concentrations. An additional more susceptible to purine nucleotide inhibition
kindred has been shown to have a threefold increase since PRPP activation is also antagonized. Despite
in erythrocyte PRPP synthetase activity as com- the existence of these feedback regulatory controls
pared to controls [17, 23]. This mutant enzyme of purine nucleotide synthesis, the pretranslational
was shown to manifest increased activity at all mutant causing overexpression of PRPS1 is the
concentrations of its activator, inorganic phos- primary effector of purine nucleotide synthesis
phate, and responded normally to nucleotide end- and increased uric acid production.
product inhibition. Further, the enzyme showed At the present time, the following molecular
normal affinities for its substrates. Other variants aspects of the enzyme, phosphoribosylpyrophos-
of mutant PRPS have been shown to have both phate synthetase, have been characterized. Recently,
regulatory and catalytic defects [17], and some two PRPS enzymes (PRPS1 and PRPS2) have been
have been shown to have a defective response to demonstrated in the rat [40], and subsequently two
inhibition by ADP and the enzyme activator, inor- enzymes have been identified in humans as well
ganic phosphate [35]. Analysis of these mutations [41]. From these and other studies, it has been
at the level of DNA is incomplete, but there is some determined that PRPS1 maps to the long arm of the
evidence that a single base transition accounts for X chromosome in the interval Xq22–q24 [42], and
some of the phenotypic changes observed [36]. PRPS2 maps to the short arm of the X chromosome
The second major PRPS1 enzyme defect (pre- in the interval Xp22.2–p22.3 [42]. Both PRPS1
translational regulation) remains unknown at and PRPS2 specify proteins with 317 amino acids
present. Although the rate of transcription of the [40, 43, 44]. Another PRPS enzyme has recently
PRPS1 gene is increased in these mutant forms, been identified that is localized to testicular tissue
there is no structural change in the PRPS1 gene and has been termed, PRPS3 [45, 46]. This tissue-
itself. Thus, there is an increased rate of tran- specific enzyme has been mapped to the short arm
scription of the gene, but no alteration in the of chromosome 7 in the interval 7p15–p21.1 [47].
enzyme’s amino acid sequence. The only This enzyme shows greater than 90 % homology
identifiable enzyme alteration is an increase in its with PRPS1 [43]. Recent studies examining the
maximal velocity. In addition, investigations of physical and kinetic properties of PRPS1 and
PRPS1 DNA showed no changes in the region 5¢ PRPS2 from recombinant isoforms purified to
to the consensus transcription initiation site. The homogeneity have shown almost complete amino
identification of normal base sequences in these acid sequence homology [48]. However, the physi-
regions excludes any defect in promoter genes or cal and kinetic properties of these isoforms differ
immediate PRPS1 flanking sequences [34, 37]. significantly. PRPS1 is more heat labile than
Speculation now suggests that a mutation may PRPS2, and the latter isoform is less sensitive to
exist in a different promoter region such as in an purine nucleotide inhibition. The PRPS2 isoform is
intronic enhancer or suppressor, in a 3¢-flanking easier to dissociate into subunits that are inactive in
DNA sequence, or in a trans-acting gene [34]. the absence of Mg2+ and ATP than PRPS1. The iso-
Thus, the actual mutation causing the increased electric points of these two isoforms are also differ-
PRPS1 transcription remains to be identified. ent suggesting a charge difference. The pH optimum
One additional key point needs mention with of PRPS1 is 8.8, whereas it is 7.4 for PRPS1.
regard to the pretranslational regulatory defect. The mutational sites in PRPS1 cDNA have
Even though normal allosteric regulation of PRPS1 been identified for two patients who manifest
activity exists in this mutant and there are increased altered feedback inhibition by purine nucleotides
purine nucleotide pools, the rate of PRPP synthe- [17, 27]. Both patients demonstrate a single base
sis is sufficient to activate the enzyme, PRPP ami- substitution. One patient has an A to G transition
dotransferase, causing an increased rate of de novo at nucleotide 341 that results in an asparagine to
purine nucleotide synthesis [34, 38, 39]. Thus, serine transition in the enzyme protein. The other
purine nucleotides inhibit both PRPS1 and PRPP patient shows a G to C transversion at nucleotide
40 3 Purine Biochemistry

547 that results in an aspartic acid to histidine the conversion of IMP to adenylosuccinate and
change at amino acid 182. When normal and then AMP and the conversion of IMP to xanthy-
mutant PRPS1 cDNAs were cloned into a plas- late and GMP, are also subject to feedback inhibi-
mid expression vector and introduced into an E. tion. AMP inhibits the conversion of inosinic
coli strain in which expression of recombinant acid to adenylosuccinate, and GMP inhibits the
hPRPS1 could be induced, the mutant phenotype conversion of inosinic acid to xanthylate. Thus,
showed properties identical to those observed the purine salvage pathways and PRPP synthetase
with the patient’s enzyme [49]. Thus, the mutant are key regulators of purine synthesis, and when
PRPS enzymes that have been examined demon- their activity and/or regulation are defective, an
strate only point mutations. Further work needs overproduction of uric acid occurs.
to be done to evaluate the molecular basis of Human glutamine phosphoribosylpyrophos-
the other types of mutations observed in this phate amidotransferase, also known as PRPP
enzyme. amidotransferase, has been shown to exist in a
small and large molecular weight form [55], and
the two forms purified from human placenta have
PRPP Amidotransferase molecular weights of 133,000 and 270,000 [56].
The conversion of the large form to the small
The second significant reaction in purine biosyn- form occurs in the presence of PRPP, and the lat-
thesis is the first committed step in purine synthe- ter is the active form of the enzyme. Purine ribo-
sis that converts phosphoribosylpyrophosphate nucleotides acting as feedback inhibitors convert
(PRPP) and l-glutamine to 5-phosphoribosylam- the small form to the large inactive form. The
ine and is catalyzed by the enzyme, PRPP amido- human enzyme has been determined in one
transferase. The latter enzyme is of significance experiment to be localized to the pter → q21
not only because it represents the first irreversible region of chromosome 4 [57]. Although no
reaction in the de novo synthesis of purines but mutant forms of this enzyme have been charac-
also because it is subject to feedback regulation terized in humans, there is evidence of mutants
by the purine nucleotides, IMP, AMP, and GMP, existing in bacteria, Ehrlich ascites cell lines, and
in an allosteric fashion (Fig. 3.11). Thus, in the Chinese hamster ovary cells [58–60]. The fact
absence of a normal complement of GMP, as that these mutants are resistant to feedback inhi-
exists with a deficiency of hypoxanthine-guanine bition suggest that human mutants of this same
phosphoribosyltransferase, there would be less type might exist. At this time, no human mutants
feedback inhibition of PRPP amidotransferase, have been described; however, it is not unreason-
and there would be more PRPP for utilization by able to suspect that some overproducers of uric
the latter enzyme. The net result of the reduction acid could have alterations in this enzyme result-
in feedback inhibitors is that the rate of de novo ing in less feedback inhibition by nucleotides or
purine pathway increases and uric acid is over- increased responsiveness to PRPP which could
produced. In contrast to the regulation of PRPP result in increased purine production.
synthetase, purine monophosphates have greater The most recent studies of PRPP amido-
inhibitory activity than diphosphates and triphos- transferase have focused on its catalytic and
phates [50–52] on PRPP amidotransferase, and inhibitory mechanisms using microbial enzymes
phosphoribosylpyrophosphate synthetase is less [61–74]. Even though these elegant structure-
sensitive to purine nucleotide feedback inhibition function studies have, for the most part, been
than PRPP amidotransferase [53]. Further, syner- accomplished using purified microbial enzymes,
gism occurs between the various feedback inhibi- they are pertinent to the human enzyme since the
tors of PRPP amidotransferase activity in contrast cDNA of human amidophosphoribosyltransferase
to its absence in the case of PRPP synthetase [50, and its derived amino acid sequence has a high
52, 54]. It should also be recognized that the degree of identity with enzymes purified from
branch points in the synthesis of AMP and GMP, other species [66]. Of even greater significance to
PRPP Amidotransferase 41

Fig. 3.11 The rates of De novo purine biosynthesis: regulation


de novo purine biosynthesis
are regulated by
phosphoribosylpyrophosphate
(PRPP) and purine nucleotide H2O3POCH2
end products, adenosine O
monophosphate (AMP) and + Glutamine
O O
guanosine monophosphate
(GMP). Purine nucleotides
are negative feedback O-P-O-P-OH
regulators of both PRPP OH
OH
amidotransferase and PRPP OH OH
synthetase activities, whereas
PRPP is an allosteric activator
of PRPP amidotransferase. PRPP amidotransferase
Elevated concentrations of
PRPP (e.g. HGPRTase Mg2+
deficiency) overcome purine
nucleotide inhibition and
increase rates of de novo
purine biosynthesis.
Decreased PRPP
concentrations slow the rate
of de novo purine H2O3POCH2 NH2
biosynthesis O

OH OH

Inosine 5’-Phosphate

Adenosine 5’-phosphate Guanosine 5’-phosphate

the correlation between the microbial and human must be removed from the human enzyme before
enzymes is the conservation of the amino acids enzyme activity can occur. Mutations in certain
essential for catalysis and feedback regulation propeptide amino acid residues do not permit
[61, 75]. On the basis of these and other studies, the processing of the enzyme. The synthesis of
the following structural and functional aspects phosphoribosylamine by PRPP amidotransferase
of PRPP amidotransferase have now been char- occurs by two-half reactions at different sites
acterized. The PRPP amidotransferase gene has on the enzyme [72]. Glutamine is hydrolyzed to
been mapped to the q12 region of chromosome 4 glutamate and ammonia at the N-terminal glu-
[75]. A propeptide consisting of 11 amino acids taminase site on the enzyme. PRPP binding to
42 3 Purine Biochemistry

its C-terminal helical site is a requirement for the It seems unlikely that human PRPP amido-
activation of the glutaminase site. PRPP binding transferase mutants causing an overproduction of
lowers the Km for glutamine by 100-fold. These urate will be identified since the enzyme reaction
changes serve to form an ammonia channel to pro- requires complex substrate interactions as well as
vide the ammonia from glutamine for its reaction conformational changes. Nonetheless, the possi-
with PRPP to form phosphoribosylamine. These bility exists that human enzyme mutants with
reaction mechanisms are generated through con- diminished feedback inhibition by purine nucle-
formational changes in the amidotransferase such otides could occur. Another amidotransferase
that the distance between the PRPP and glutamine- mutant was isolated from E. coli grown in mini-
binding sites are drawn closer together [72]. The mal essential media and 6-diazonorleucine, an
mechanism for interdomain signaling between inhibitor of PRPP amidotransferase [76]. This
the PRPP and glutamine sites has yet to be com- bacterial mutant utilized ammonia as a substrate
pletely established, but the key amino acids for rather than glutamine for de novo purine nucle-
substrate binding and the ammonia channel have otide synthesis when grown in the presence of
been defined [72]. Tyrosine is the key to the estab- 6-diazonorleucine, a glutamine analog and irre-
lishment of the interactions between the NH2- versible inhibitor of PRPP amidotransferase
terminal glutamine site and the COOH-terminal activity. This mutant grew very slowly in mini-
PRPP site. Glutamine binding is regulated in part mal essential media and clearly would not have
by arginine and aspartic acid [73–75]. shown increased rates of purine synthesis. Thus,
With regard to the feedback inhibitor sites on no human PRPP amidotransferase mutants have
PRPP amidotransferase, adenine and guanine been characterized at this time, but there is a
nucleotides as well as other less common nucle- remote possibility that they could exist.
otides (6-mercaptopurine nucleotide) bind to an
allosteric site (A-site) and a catalytic site (C-site)
on the enzyme [62, 65, 67]. Feedback inhibitors Hypoxanthine-Guanine
(adenosine monophosphate-AMP and guanosine Phosphoribosyltransferase (HPRTase)
monophosphate-GMP) of de novo purine nucle-
otide biosynthesis may act in concert to cause Since the de novo purine nucleotide biosyn-
more than additive inhibition of the amidotrans- thetic pathway utilizes several small precursor
ferase. Even though both nucleotides can interact molecules (glycine, aspartic acid, glutamine,
with either the A-site or the C-site, evidence 10-formyl-H4-folate, and bicarbonate) and six
derived from microbial PRPP amidotransferase moles of adenosine triphosphate for the gen-
has determined that GMP prefers binding to the eration of each purine end product, recycling
A-site and that AMP prefers binding to the C-site. of purine bases rather than excreting them con-
The A-site on the enzyme is localized adjacent to serves cellular energy and precursor molecules.
the catalytic site in between the enzyme subunits. The salvage enzyme, hypoxanthine-guanine
Four intersubunit allosteric sites exist for each phosphoribosyltransferase (HGPRTase), serves
amidotransferase tetramer. Nucleotide reactions this purpose by reutilizing hypoxanthine or gua-
with the A-site or C-site keep the native enzyme nine. Its counterpart, adenine phosphoribosyl-
in an inactive conformation. Although nucleotide transferase (APRTase), performs a comparable
feedback control is the major rate-limiting factor task by reusing the purine base, adenine. These
of the de novo purine nucleotide biosynthetic two enzymes are part of a family of related phos-
pathway, regulation of this pathway is also depen- phoribosyltransferases that catalyze the synthesis
dent on another key enzyme, PRPP synthetase. of pyrimidines, histidine, nicotinates, nicotin-
As will be discussed subsequently, it appears that amides, quinolates, and tryptophan in a variety of
fine control of de novo purine biosynthesis is organisms. In most higher organisms, HGPRTase
regulated primarily by interactions with PRPP has its highest activities in the testes and brain.
amidotransferase. In the latter organ, it is primarily concentrated
Hypoxanthine-Guanine Phosphoribosyltransferase (HPRTase) 43

in the basal ganglia. Some have suggested that hypoxanthine, and PRPP from published reports
there is an inverse relationship between the range between 4.0–21, 9.0–17, and 220–250 uM,
de novo purine nucleotide pathway and the sal- respectively [85, 93–95]. The human enzyme is
vage enzymes so that in those organs where the modified by posttranslational changes that
de novo pathway is most active, the salvage path- account for its electrophoretic heterogeneity [96–
ways are least active. This appears to be the case 99]. Several additional studies have utilized
in the liver where the de novo pathway is most mutant enzymes and analogs to characterize the
active and the salvage enzymes are least active. role of certain structural components of the
Similarly, in the brain where the de novo pathway enzyme. The following articles should be con-
is nonexistent, the salvage enzymes have their sulted for more structure-function relationships
highest activity. HGPRTase also has higher activ- [100–103]. A comparison of the cDNA from the
ity in the gray matter of the brain than in white human, rat, Chinese hamster, and mouse HPRTase
matter [77–80]. Both these enzymes are localized shows a high degree of similarity approaching
to the soluble cytoplasmic compartment of cells 70–90 % [104]. In fact, the primary structures of
even though some evidence suggests that there the human hypoxanthine-guanine phosphoribo-
may be a loose binding between these enzymes syltransferase and two bacterial phosphoribosyl-
and the cell membranes of erythrocytes and lym- transferases (E. coli PRPP amidotransferase and
phocytes [81–83]. The activity of this enzyme is S. typhimurium adenosine triphosphate phospho-
also increased in tissues that have a rapid turn- ribosyltransferase) show a common amino acid
over such as embryonic cells [77, 84]. domain between the E. coli and the human
Hypoxanthine-guanine phosphoribosyltrans- enzyme but no shared domain with the S. typh-
ferase catalyzes the transfer of phosphoribose imurium enzyme [105]. Similarly, another
from dimagnesium PRPP to the nine positions of 20-amino-acid domain common to mouse,
hypoxanthine or guanine resulting in the forma- human, and bacterial phosphoribosyltransferase
tion of IMP or GMP. Adenine phosphoribo- proteins has also been described [106, 107].
syltransferase transfers the same moiety Analysis of these common amino acid spans led
(phosphoribose) from dimagnesium PRPP to the to a prediction that a dinucleotide-binding fold
nine positions of adenine to form AMP. existed which could be related to substrate bind-
Magnesium ions are competitive inhibitors of the ing [105]. Subsequent data confirmed the pres-
dimagnesium PRPP complex [85, 86]. Human ence of hypoxanthine/guanine and PRPP-binding
HGPRTase is reported to exist as a dimer or sites to this fold [106–112].
tetramer depending on the ionic strength and pH The cloning of the HGPRTase gene has pro-
of its surrounding milieu. The monomers of the vided additional information as to the gene, itself,
dimer or tetramer are identical in composition its pseudogenes, and the regulation of its expres-
and contain 217 amino acids [87–89]. Each sion [113]. The gene for hypoxanthine-guanine
monomer has a molecular weight of 24,470 [87, phosphoribosyltransferase has been localized to
89, 90]. The N-terminus of the original 218 amino the X chromosome at Xq26–q27 [114–116], and
acid protein, methionine, is removed leaving ala- fine structure analysis has defined the order of the
nine as the N-terminus. The alanine N-terminus various gene markers in the region of the
is subsequently acetylated [87]. The enzyme is HGPRTase gene [117, 118]. In addition to the
composed of ten b strands and six a helices, and locale for the principal gene, there are four
the active site of the enzyme has been character- HGPRTase pseudogenes that have been deter-
ized as well as other structural components of the mined to reside on chromosomes 5, 3, and 11
normal and mutant enzymes [91, 92]. These stud- [119]. Two of the four pseudogenes reside on
ies also determined the crystal structure of the chromosome 11, one is on chromosome 3, and
enzyme in complex with GMP and that the reac- the other is in the region of 5p13–q11. These
tion mechanisms strongly favor the formation of pseudogenes are nonfunctional copies of the
the nucleotide product. The Km values for guanine, functional gene. The HGPRTase gene contains
44 3 Purine Biochemistry

39.8 kb from its initiator methionine codon (ATG) known to exist for many years in bacteria and cul-
to its termination codon, TAA [120]. The gene tured mammalian cells [127–129], the first human
consists of nine exons. When the amino acid mutant of this enzyme was described in 1967.
sequences of human, rat, Chinese hamster, and This mutant was determined in association with a
mouse HGPRTase are compared, only 16 amino clinical syndrome characterized by hyperurice-
acids differences are found between the four spe- mia, hyperuricaciduria, severe neurological
cies [104]. The regions of the HGPRTase gene deficits including in some cases mental retarda-
necessary for the transcription of its DNA into tion [130], and soon thereafter, a second mutant
mRNA, the promoter region, have been deter- was described in adults with gouty arthritis [131].
mined. This region has the characteristics of a These two human enzyme deficiencies were sub-
housekeeping gene since it has guanosine-cyto- sequently described in many human tissues from
sine-rich areas and no TATA or CCAAT sequences affected individuals including amniotic fluid
[120]. These GC-rich regions are found surround- cells, cultured human fibroblasts, and lympho-
ing promoter regions and have been called blasts. Initially, mutations in the enzyme were
“housekeeping genes” because they encode pro- identified by differences in their thermolability,
teins essential for cell viability. In the HGPRTase substrate affinity, electrophoretic mobility, and
gene, there are four copies of the GC-rich hexa- decreased reactivity to anti-hypoxanthine guanine
nucleotide sequence, 5¢-GGCGG-3¢, on the 5¢ phosphoribosyltransferase antibodies [94, 132–
side of the first exon and three copies on the 3¢ 139]. With the determination of the entire amino
side [120]. This sequence has been determined to acid sequence of the enzyme, peptide sequences
be a binding site for the transcription factor, Sp1, between normal and mutant enzymes could be
in dihydrofolate reductase and may have the same assessed, and with this methodology, structural
function in the enzyme, hypoxanthine-guanine variations were observed in some mutants [108].
phosphoribosyltransferase [121]. Since these All these mutant detection methods are depen-
GC-rich areas are located on both sides of exon dent on the presence of enzyme protein, and since
one, sequences of significance to the regulation over 70 % of Lesch-Nyhan patients have no
of the HGPRTase gene may exist in both these detectable enzyme protein, these methods failed
locations as well. Multiple transcription start sites to provide any information concerning the type of
have also been identified in association with this mutants in these patients [140]. Cloning of mutant
gene [122, 123]. Other regulatory regions for the cDNA from affected individuals has revealed
HGPRTase gene have also been defined includ- significant heterogeneity in the characteristics of
ing negative control elements, but the complete the mutations including point mutations, substitu-
regulatory aspects of the gene remain to be char- tions, deletions, and duplications spanning the
acterized [124–126]. In addition to the fine struc- whole enzyme protein, splicing site mutations,
ture of the HGPRTase gene, several other and conversions to stop codons [110, 141–151].
parameters of this gene are of significance. Its Many of these mutations have been catalogued
role in understanding X-chromosome inactiva- and characterized [152]. There is a clear spectrum
tion, the mechanism by which it causes an of clinical subsets of hypoxanthine-guanine phos-
increase in de novo purine biosynthesis, the other phoribosyltransferase deficiency ranging from
biochemical changes associated with mutations patients with the classical Lesch-Nyhan syn-
in the HGPRTase gene, and the characteristics of drome who have choreoathetosis, self-mutilation,
HGPRTase mutants all have contributed to the spasticity, developmental retardation, muscle
knowledge base of human genetics. hypotonia, and massive uricaciduria to those with
The principal interest in the HGPRTase the Lesch-Nyhan syndrome without self-mutila-
enzyme has been the various mutant forms of this tion, to those with late onset Lesch-Nyhan syn-
phosphoribosyltransferase that have been drome with slight neurological involvement, and
described and the diseases associated with them. to patients with the Kelley-Seegmiller syndrome
Although mutants of this enzyme have been (partial deficiency of HGPRTase) who have gouty
Hypoxanthine-Guanine Phosphoribosyltransferase (HPRTase) 45

arthritis, urate stones, uricaciduria, hyperurice- cells; it is only in the erythrocyte where this
mia, and renal disease. Unfortunately, with the mechanism appears to result in increased adenine
exception of finding partial enzyme activity in phosphoribosyltransferase activity.
patients with the Kelley-Seegmiller syndrome Another enzyme, inosine monophosphate
and little or no enzyme activity in those with the (IMP) dehydrogenase, which catalyzes the con-
Lesch-Nyhan syndrome, no correlation has been version of IMP to xanthosine monophosphate
identified between the molecular abnormality and (XMP) in the presence of DPN+ as a hydrogen
the degree of the neurological deficit. To date, acceptor, is also found in elevated concentra-
well over 100 separate and distinct mutations tions in the erythrocytes of hypoxanthine-guanine
have been described for HGPRTase. Of some phosphoribosyltransferase-deficient patients
interest, mutations affecting arginine at codon 51 [173, 180]. The mechanism by which this enzyme
when converted to glycine result in gouty arthritis activity is increased has been proposed to be the
[109], whereas if the same amino acid is con- result of the high concentration of 2,3-diphospho-
verted to proline, the result is a patient with the glycerate in erythrocytes [181]. When the level of
Lesch-Nyhan syndrome [141]. A variety of new this metabolite is reduced by prolonged dialysis,
mutants have been identified since those summa- IMP dehydrogenase activity is comparable to the
rized in the 1998 edition of McKusick’s Mendelian level of activity in normal erythrocytes [181]. In a
Inheritance in Man including data on the study of Lesch-Nyhan patients with no HGPRTase
identification of heterozygotes and the mutation activity, increased nicotinamide adenine dinucle-
in a female with the Lesch-Nyhan syndrome otide (NAD) levels have been determined in
[153–168]. One issue that remains to be resolved erythrocytes [182]. Elevated activities of nicotinic
is the relationship between these various muta- acid phosphoribosyltransferase and NAD syn-
tions and the different clinical presentations of thetase were also found in these patients. The Km
HPRTase deficiencies. for the substrates of these enzymes was within the
In addition to the primary defect in the normal range, but the Vmax for these enzymes was
enzyme, hypoxanthine-guanine phosphoribosyl- increased. Thus, the elevated NAD levels in eryth-
transferase, several secondary biochemical alter- rocytes can be accounted for by the increased
ations have also been described in patients and activity of these biosynthetic enzymes. Finally,
cells expressing this mutation. In patients with monoamine oxidase, an enzyme that catalyzes
either the Lesch-Nyhan or Kelley-Seegmiller the degradation of biogenic amines, has been
syndrome, the erythrocyte activity of the enzyme, found to have lower activity in skin fibroblasts
adenine phosphoribosyltransferase, is increased from patients with the Lesch-Nyhan syndrome
[130, 169–173]. The mechanism for this altered and in HGPRT-deficient neuroblastoma cells than
activity has been proposed to result from the in control neuroblastoma cells or normal skin
increased levels of erythrocyte PRPP which fibroblasts [183–187]. Since the genes for mono-
has been determined to stabilize the enzyme, amine oxidases A and B have been determined to
adenine phosphoribosyltransferase [174–177]. reside on the X chromosome, the relationship
Interestingly, adenine phosphoribosyltransferase between these enzymes and hypoxanthine-gua-
activity is completely within the normal range nine phosphoribosyltransferase remains to be
in fibroblasts, lymphoblasts, and neuroblastoma determined [188]. Thus, mutant hypoxanthine-
cells in specimens recovered from patients with guanine phosphoribosyltransferases result in
hypoxanthine-guanine phosphoribosyltrans- changes in other enzymes. At this time, no patho-
ferase deficiencies [12, 178, 179]. Even though genic relationships have been defined between
the levels of PRPP are increased in these cells, the primary mutation and these secondary
there is no concomitant increase in adenine phos- changes.
phoribosyltransferase activity. Thus, the elevated In addition to the characterization of mutant
levels of PRPP do not stabilize the latter enzyme HGPRTase and the secondary enzyme effects
in lymphoblasts, fibroblasts, and neuroblastoma in association with hypoxanthine-guanine
46 3 Purine Biochemistry

phosphoribosyltransferase deficiency, the mech- dotransferase. This latter mechanism is probably


anism for the overproduction of purines and uric the major factor in the increased purine synthe-
acid needs to be discussed. The deficiency in the sis observed in patients who are hypoxanthine-
activity of hypoxanthine-guanine phosphoribo- guanine phosphoribosyltransferase-deficient.
syltransferase leads to an increase in the avail- A relative deficiency of the nucleotides inosine
ability of its substrates, hypoxanthine, guanine, diphosphate, inosine triphosphate, guanosine
and PRPP, since the capacity of the enzyme to sal- diphosphate, and guanosine triphosphate may
vage purine bases is diminished and fewer of its contribute to the increased rate of purine synthe-
substrate molecules are utilized. This decreased sis through an absence of their inhibition of the
reutilization of the free bases, hypoxanthine, and enzyme, PRPP synthetase.
guanine, for ribonucleotide synthesis to inosine Since the HGPRTase gene has been localized
monophosphate (IMP) and guanosine mono- to the X chromosome, it has been used as a model
phosphate (GMP), results in a diminished intra- to understand the dosage compensation of
cellular concentration of these nucleotides, and X-linked genes in male and female and the ran-
fewer nucleotides are available to exert feedback dom inactivation of female somatic cell X chro-
inhibition on PRPP amidotransferase, the enzyme mosomes. There now exists sufficient evidence to
catalyzing rate-limiting step in de novo purine bio- postulate that DNA methylation participates in
synthesis. This mechanism for increased de novo the regulation of X-chromosome inactivation.
purine biosynthesis is a reasonable explanation The evidence can be summarized as follows.
for the increased synthesis of IMP in hypox- Active X-chromosome DNA can provide
anthine-guanine phosphoribosyltransferase- HGPRTase enzyme activity to an HGPRTase-
deficient human fibroblasts [189]. In addition, deficient recipient cell. Inactive X-chromosome
comparative measurements of urinary hypox- DNA cannot provide an HGPRTase-deficient
anthine and xanthine in hypoxanthine-guanine recipient cell with HGPRTase enzyme activity.
phosphoribosyltransferase-deficient subjects The use of 5-azacytidine to cause hypomethyla-
as compared to normal subjects and those with tion of cytosine in DNA is able to reactivate genes
primary gout and no known enzyme defect of an inactive X chromosome, and altered pat-
show striking increases in uric acid, hypoxan- terns of DNA methylation exist between active
thine, and xanthine in hypoxanthine-guanine and inactive X chromosomes [191–205].
phosphoribosyltransferase-deficient subjects. Evidence that supports the role of DNA methyla-
Data from the literature illustrate these changes; tion in X-chromosome activation is found in the
urinary uric acid (mg 24 h−1 1.73 m−2), hypoxan- differential methylation of the human HGPRTase
thine (umol/g creatinine), and xanthine (umol/g genes [192, 205–207]. On the first intron of the
creatinine) were 1,906 ± 645, 544 ± 356, and HGPRTase gene, there is an unmethylated site
223 ± 167,respectively, in HPRTase-deficient that is present on an active X chromosome, and
subjects as compared to levels of 418 ± 160, this same site is methylated when it is on an inac-
29 ± 18, and 17 ± 12 in patients with primary tive X chromosome [192, 193, 205]. Another
gout and 446 ± 86, 47 ± 14, and 29 ± 9 in normal area of the gene demonstrates just the reverse,
subjects [190]. A second mechanism resulting in methylation in the active and hypomethylation in
increased de novo purine biosynthesis is based on the inactive X chromosome [192, 193, 205]. In
the fact that PRPP, the other substrate for the sal- the 5¢ region of the gene, reactivated X chromo-
vage enzyme, is present in excess since it is not somes are demethylated [193]. Since the mouse
being utilized in the reaction catalyzed by hypox- gene for HPRTase provides evidence that the
anthine-guanine phosphoribosyltransferase. This changes in methylation occur after the X
excess PRPP drives de novo purine biosynthesis chromosome has been inactivated, it is likely that
since it is a substrate for the rate-limiting step the methylation-demethylation process is not the
of purine synthesis, the conversion of PRPP to entire event related to X-chromosome inactivation,
phosphoribosylamine by the enzyme, PRPP ami- but may be involved in the maintenance of the
Adenine Phosphoribosyltransferase 47

inactivation process [208]. X-chromosome inac- 2,8-dihydroxyadenine. These renal calculi have
tivation may be either a random or a nonrandom often been confused with uric acid calculi, and
process; in the former case, females who are for this reason, autosomal recessive APRTase
heterozygotes for an X-linked genetic trait have deficiency is sometimes misidentified as a defect
two different cell types depending on which X in uric acid metabolism. The discussion that fol-
chromosome has been inactivated and are, there- lows is designed to prevent this confusion.
fore, mosaics. Since significant variation is Second, APRTase is deficient in some families
observed with respect to which X chromosome is whose members have familial juvenile gouty
inactivated, this variation may account for the nephropathy [217, 218], and its pathogenic role
different degrees of symptom expression related in this disorder remains incompletely defined.
to an X linked mutation. This mechanism might Since this association may relate to alterations in
be responsible for the occurrence of gout in uric acid metabolism, a discussion of the bio-
female patients who carry the X-linked HGPRTase chemistry of APRTase is provided to assist the
mutation [209]. A summary of the most recent reader in understanding future studies that may
evidence suggests that DNA methylation plays a characterize this relationship more fully. Finally,
major role in X-chromosome inactivation and this enzyme is of significance to understanding
that the position and length of the methylated the feedback regulation of de novo purine bio-
regions as well as the chromatin structure may synthesis since adenosine monophosphate is a
interact to repress housekeeping promoters feedback inhibitor of PRPP amidotransferase. A
on the inactive X chromosome [210–213]. deficiency in the activity of the salvage enzyme,
Nonetheless, some genes on the X chromosome adenine phosphoribosyltransferase, could affect
escape inactivation, and the mechanism for this the rate of de novo purine biosynthesis by reduc-
escape is incompletely understood at this time ing the cellular levels of adenosine monophos-
[214–216]. Alterations in X-chromosome inacti- phate, a negative feedback inhibitor of the first
vation and escape mechanisms may have impli- committed enzyme in purine synthesis. As will
cations for carrier females and disease. be discussed subsequently, this is not the case.
One final issue related to the HGPRTase gene The salvage enzyme, adenine phosphoribosyl-
is completely unrelated to the subject matter of transferase (APRTase), catalyzes the conversion
this book but has relevance to the use of informa- of adenine and phosphoribosylpyrophosphate
tion that the foregoing studies have provided to (PRPP) to the mononucleotide, 5¢-adenosine
the general scientific community. Since the gene monophosphate (AMP), and inorganic pyrophos-
has been cloned, it has been used as a target for phate (PPi). In humans, this is the only system
potential mutagens. There are numerous publica- available for salvaging the free purine base, ade-
tions showing the utility of this gene in the study nine. The Km for its substrate, adenine, using
of mutagenesis and its role in the identification of human red blood cells as a source of the enzyme
environmental and drug mutagens. Thus, knowl- ranges between 1.1 and 2.7 uM [219–222] and for
edge of the HGPRTase nucleotide and amino acid PRPP between 6 and 29 uM [219, 221–223]. The
sequence in normal individuals permits an analy- latter substrate is Mg2+-dependent although other
sis of alterations caused by mutagens. metals may serve as substitutes but with much less
efficiency. Both end products of the reaction
inhibit substrate conversion by the enzyme to its
Adenine Phosphoribosyltransferase end products. Inhibition of APRTase activity by
PPi is noncompetitive with respect to both sub-
There are several reasons why a deficiency in strates (adenine/PRPP), whereas inhibition by 5¢-
adenine phosphoribosyltransferase (APRTase) AMP is noncompetitive with respect to adenine
is discussed in a treatise on gout (Fig. 3.12). but competitive for PRPP [224, 225]. Several ade-
First, APRTase-deficient patients form kidney nine analogs can also act as substrates for APRTase
stones composed of the adenine metabolite, including 4-amino-5-imidazole-carboxamide
48 3 Purine Biochemistry

Fig. 3.12 The counterpart to Adenine metabolism in the presence and absence of adenine
hypoxanthine-guanine Phophoribosyl transferase
phosphoribosyltransferase
is adenine phosphoribosyl- NH2
NH2
transferase (APRTase), the
enzyme that recycles adenine
rather than excreting this N N
N Mg2+ N
purine metabolite. When
+ PRPP
adenine phosphoribosyltrans-
ferase deficiency exists, N
N N
adenine is oxidized by the N H
enzyme, xanthine oxidase, to R-P
form 8-hydroxyadenine and Adenine
2,8-dihydroxyadenine
(2,8-DHA). 2,8-DHA is Xanthine oxidase
nephrotoxic due to its
insolubility and may cause
NH2
renal failure but usually
results in the formation of
2,8-dihydroxyadenine kidney N
stones. 2,8-dihydroxyadenine N
is indistinguishable from uric OH
acid by routine tests, and the
N
stones formed from this N H
metabolite are also
radiolucent just as uric acid 8-Hydroxyadenine
stones are. Uric acid and
2,8-DHA can be
distinguished from each other
by specific tests Xanthine oxidase

NH2

N
N
OH

N
HO N H

2, 8-Dihydroxyadenine

(AIC), 2,6-diaminopurine (DAP), 4-aminopyra- form survive and are selected for their absence of
zole (3,4-d) pyrimidine, 8-azaadenine, 6,meth- APRTase activity. None of these analogs have a
ylpurine, 6-methylaminopurine, 2-fluoroadenine, higher affinity for APRTase than adenine [226].
and 4-caramoyl-imidazololium-5-olate [226– Adenine phosphoribosyltransferase has been
228]. DAP and 8-azaadenine are used to select purified to near homogeneity from human eryth-
for cells resistant to these agents and deficient in rocytes [229], and its structure determined to be a
APRTase by methods similar to those used for dimer with 179 amino acids per subunit [230].
the selection of HGPRTase-deficient cells. The The molecular weight of the enzyme has been
addition of antimetabolites to tissue culture media calculated to be 38,962 in total or 19,481 per sub-
along with cells permits the conversion of these unit. cDNA sequencing has predicted a polypep-
antimetabolites to their active form by normal tide of 180 amino acids which is consistent with
APRTase activity, and these products are then the amino acid sequence determined from the
lethal to the normal cell. Cells without the capac- purified enzyme since the second amino acid,
ity to convert these antimetabolites to their active alanine, is acetylated and becomes the N-terminal
Adenine Phosphoribosyltransferase 49

amino acid. The gene is composed of 5 exons A survey of APRTase mutations in Japan, Korea,
[231]. The human gene for APRTase has been and Taiwan gave a heterozygote frequency of 1.1
mapped to chromosome 16q24 [232–234], and % for the Japanese and 0.53 % for the Koreans.
fine mapping has shown the location of the gene No heterozygotes were identified in the Taiwanese
to be distal to the haptoglobin (HP) gene and the [278]. On the basis of the presence of this muta-
fragile site (FRA16D) so the resultant gene order tion in Okinawans and Koreans, it has been esti-
is HP-FRA16D-APRTase-qter [234]. Hamster or mated that the origin of this mutation occurred
mouse APRTase gene probes have been used to prior to 300 BC [278]. Type I patients develop
isolate the human gene from a genomic lambda symptoms of the disease at an earlier time than
bacteriophage library [235–237]. A BamHI frag- type II patients. The biochemical abnormalities
ment has been used as a probe to isolate APRTase associated with these mutations are principally
cDNA clones from human cDNA libraries [237, related to the resultant increase in the urine of
238]. The APRTase gene has been determined to adenine and its metabolites, 8-hydroxyadenine
have five exons and four introns, and the amino (8-HA) and 2,8-dihydroxyadenine (2,8-DHA)
acid sequence obtained from the gene cDNA is [277]. Increased levels of these latter metabolites
consistent with the sequence from peptide map- arise because of the inability of cells to salvage
ping studies [230]. Restriction-fragment-length adenine and the subsequent oxidation of adenine
polymorphism (RFLP) sites have also been by xanthine oxidase to 8-HA and 2,8-DHA. The
identified, and the frequencies of these sites have ratio of these urinary purines has been determined
been determined for Caucasian and Japanese to be 1.0:0.03:1.5 for adenine, 8-HA, and 2,8-
populations [237–241]. The promoter region for DHA, respectively [277]. In patients on low-
the APRTase gene like that for the HGPRTase purine diets, adenine compounds represent 20–30
gene does not contain any CCAAT or TATA % of the total urinary purine end products (uric
sequences [242, 243]. There are also G- and acid, oxypurines, and adenine metabolites)
C-rich regions of the gene located in the promoter excreted [250, 253, 263, 265, 266, 289].
region that may represent binding sites for Sp1 or Heterozygotes with a deficiency in this enzyme
other components that assist in the binding of have no alterations in adenine metabolite excre-
RNA polymerase to DNA [242, 244]. Like the tion. Fast and relatively simple screening meth-
HGPRTase gene, the APRTase gene has a rather ods have been developed for the detection of
unique distribution of CpG dinucleotides that are 2,8-dihydroxyadenine in untreated urine samples
likely to be methylation sites that may control by capillary zone electrophoresis [290]. Using
gene expression [245, 246]. this methodology, 2,8-dihydroxyadenine is sepa-
As with HPRTase-deficient patients, the rated within 8 min, and its characteristic ultravio-
mutant forms of APRTase have been of greatest let spectra can be used to confirm its presence.
interest to the geneticist and clinician. Two major As indicated previously, type I APRTase
types of mutations have been identified through deficiency is manifested by a complete absence
clinical and biochemical studies: type I deficiency of APRTase activity, whereas the type II lesion
(no APRTase enzyme activity in cell extracts) is only a partial deficiency of the enzyme. The
and type II deficiency (partial APRTase enzyme latter defect is found almost exclusively in the
activity in cell extracts). The type I defect has Japanese population [278–288], whereas the for-
been described in patients from many ethnic mer has been described in patients from many
backgrounds [247–277], whereas the type II different countries including Japan [247–277]. In
alteration has only been identified, at this time, in the type I defect, enzyme activity in the erythro-
Japanese patients or those with Japanese heritage cyte is either very low or absent in homozygotes
[278–288]. In fact, the most common mutation in [247–277], and heterozygotes of this type show
the Japanese is a mutation of the trinucleotide about 25 % of normal enzyme activity [219, 220,
ATG (methionine) to ACG (threonine) at codon 223, 247–277, 291]. The proposed mechanism
136, and this mutation causes 68 % of the for the absence or very low enzyme activity is
disease-causing genes among the Japanese [278]. that either little or no enzyme is synthesized,
50 3 Purine Biochemistry

or the enzyme once synthesized is degraded When one considers assays to distinguish the
at a rapid rate. A variety of mutations in the heterozygotes of both types I and II, the heterozy-
structural gene for APRTase could cause such gous types cannot be differentiated by their resis-
alterations, but detailed characterization of such tance to adenine analogs, their uptake of
structural changes at the molecular level remains radioactive adenine, or the level of enzyme activ-
to be described. In the heterozygotes with 25 % ity in cell lysates [279–284]. Both types of
enzyme activity and not the expected 50 % activ- heterozygotes generate DAP-resistant clones of
ity, it has been postulated that random aggrega- T lymphocytes, but no resistant clones arise in
tion of the two dimers making up the complete individuals who do not carry any abnormality in
APRTase enzyme occurs leading to three pos- APRTase. The DAP-resistant clones from type I
sible combinations. These would include a dimer heterozygotes have no detectable enzyme activ-
enzyme made up of two normal monomers, a ity, whereas those from type II heterozygotes
dimer containing one normal monomer and a show significant APRTase activity.
mutant monomer, and a dimer consisting of two The properties of types I and II mutant
mutant monomers. If only the normal-normal enzymes are quite different. As noted previously,
dimer is active, then only 25 % of the normal type I mutant enzymes from homozygotes show
APRTase activity would result in the other forms. APRTase activity and immunoreactive protein
No explanation has yet been found for the occur- values of less than 1 % of the normal enzyme.
rence of 50 % of the normal APRTase activity APRTase activity from heterozygotes is about
in lymphoblasts and fibroblasts obtained from 25 % of normal; however, the immunoreactive
heterozygotes for the type I deficiency. Possible enzyme protein levels in heterozygotes range
explanations include the fact that the hybrid from 22 to 100 %. In lymphoblasts from heterozy-
dimers are more stable in those cells, greater gotes, the corresponding values are 46 and 41 %,
quantities of the normal allele are present, or the respectively. In those with type II deficiency, the
mutant allele is unstable [292]. Cells from type enzyme has a reduced affinity for PRPP with a
I homozygotes fail to survive in azaserine (glu- normal affinity for adenine [282, 294]. The
tamine analog)-alanosine-adenine medium but mutant enzyme shows the kinetics of an allosteric
have a normal survival in DAP medium [260, enzyme with respect to PRPP. A sigmoidal curve
281, 284, 293]. In contrast, cells from type I is generated when its substrate (PRPP) concen-
heterozygotes survive in azaserine-alanosine- tration is plotted as a function of enzyme satura-
adenine medium, whereas they do not in DAP tion. The Hill coefficient is nearly 2.0 for some
medium [258, 281, 284, 293]. families who possess the mutant enzyme, whereas
In the type II defect, erythrocyte hemolysates it is 1.0 for the normal enzyme. In addition, the
have approximately 25 % of the normal APRTase S0.5 value for PRPP is 2.9 uM for the normal
activity, and T lymphocyte extracts show about enzyme and in the range of 47–82 uM in the
50 % of the normal activity [278–281, 288]. mutant enzyme. The enzyme from those with the
Patients homozygous for the type II defect cannot type II defect has altered heat stability when
be distinguished from type I heterozygotes using compared to the normal enzyme [282, 294, 295].
only enzyme assays conducted on cell lysates Normal APRTase activity and type II mutant
[280]. Cells from type II homozygotes are resis- enzyme can be distinguished using starch gel
tant to the adenine analogs, 6-methylpurine and electrophoresis since the mutant enzyme is found
DAP, indicating that although enzyme activity is closer to the anode than the normal enzyme [287,
detected in these patients, the enzyme is nonfunc- 296]. Type II heterozygotes show three bands on
tional in the intact cell [281, 282]. In contrast, starch gel electrophoresis as represented by the
cells manifesting the type I heterozygosity are wild, hybrid, and mutant. It is important to recog-
sensitive to these analogs. Further, intact erythro- nize the fact that recent blood transfusions lead-
cytes from type II homozygotes incorporate ing to admixtures of erythrocytes may provide
minimal amounts of radioactive adenine [280].
Adenine Phosphoribosyltransferase 51

misleading results in patients with these muta- lithiasis from 2,8-dihydroxy adenine stones, and
tions [289]. misclassified patients may have been excluded.
In summary, there are two different mutant Finally, some mutations may be lethal, and lethal
forms of the APRT enzyme that cannot be distin- mutations may be excluded from ascertainment
guished from each other by assaying erythrocyte calculations as well. Molecular studies of non-
lysates alone. Patients and heterozygotes are only Japanese and Japanese patients with this disorder
distinguishable when several different assays are have shown that only three mutations exist in
used. These assays include (1) measurements of Japanese patients. The most common mutation in
APRTase activity in lysed and intact erythrocytes type II deficiency is a substitution of a threonine
and cultured lymphocytes [283, 297, 298], (2) (ACG) for methionine (ATG) at codon 136 desig-
assessment of the sensitivity of cultured cells to nated as M136T. In type I deficiencies in this
adenine analogs [281, 285, 293, 299], (3) analy- same population, two mutations have been
sis of urinary adenine metabolites by high-per- identified, a TGG to TGA missense mutation in
formance liquid chromatography or other codon 98 designated as W98X [288, 306–308]
methods [270, 299, 300–302], and (4) assays for and a four base pair duplication, CCGA, at codon
adenine uptake using intact cells [260, 298–300]. 87 [288]. These three mutations represent 96 %
Erythrocyte lysate assays for APRTase are used of the mutations observed in the Japanese with
to identify type I defects in homozygotes and the mutant, M136T, representing a little less than
heterozygotes. Intact erythrocyte studies using 70 % of the total. The M136T mutation appears
radioactive adenine to assay uptake differentiate to have occurred within the codons for the PRPP-
type I heterozygotes from homozygotes for the binding region of the enzyme and could account
type II defect since the former incorporate for the changes in the kinetic properties of this
significant quantities of the isotope, whereas the enzyme (increased Km for PRPP) [202]. There is
latter only incorporate negligible amounts. Their sufficient evidence based on the relationships
resistance to DAP or 6-methylpurine [303] dis- between the M136T mutant and a TaqI polymor-
tinguishes homozygous type II patients. The so- phic site in intron 2 to suggest that a crossover
called compound heterozygotes composed of has occurred between the mutation and SphI
enzymes carrying the mutant phenotype for type polymorphic site [211]. The TaqI restriction-
II defects and the type I defect showing no fragment-length polymorphism (RFLP) site
enzyme activity (null allele) are only differenti- results in a 2.1-kb and a 2.7-kb fragment when
ated by the use of analog-containing media. the gene is exposed to TaqI. The frequency of the
As noted previously, APRTase deficiencies are alleles in the Caucasian population is 0.79 for the
inherited as autosomal recessive disorders [191, smaller fragment and 0.21 for the larger frag-
191, 194, 291, 297, 303], and the frequency of ment, whereas in the Japanese population, the
heterozygotes has been estimated to range allele frequency is about 0.50 [209]. The SphI
between 0.4 and 1.1 % in various Caucasian pop- RFLP is 2.8 kb upstream from the TaqI site in
ulations [190, 263, 291, 297, 304, 305]. intron 2, and it results in an 8- and 12-kb frag-
Heterozygote frequency in the Japanese popula- ments with allele frequencies of about 0.80 and
tions varies between 0.5 and 1.2 % [283, 299]. 0.20 in the Caucasian population and of about
These estimates are higher than the actual 0.40 and 0.60 in the Japanese population [81,
observed frequencies, and at this time, one can 211]. In the Japanese population, all the M136T
only speculate as to why there is a variance mutants (194 alleles) have been associated with
between the observed and estimated data. 2.7-kb TaqI fragment, and 42 of these alleles are
Ascertainment may be one factor that accounts for associated with the 8-kb SphI fragment and 6
some of these differences. Asymptomatic patients with the 12-kb SphI fragment. These data along
have been described, and such patients may not be with the population studies cited previously sug-
included in the ascertainment estimates. Confusion gest that a single ancestral mutation may well
has existed in the differentiation of uric acid account for the type II mutation [211, 278].
52 3 Purine Biochemistry

Similar molecular studies have been disorders, N-acetylgalactosamine-6-sulfate sul-


performed to assess the basis of the type I non- fatase (GALNS) deficiency and adenine phos-
Japanese patients, and several different mutations phoribosyltransferase deficiency, may be more
have been identified. The most common mutation common than previously recorded since both
is a splicing defect localized to intron 4 [309, genes for these enzymes reside on the chromo-
310]. Insertion of a T nucleotide at the intron 4 some location 16q24.3 [317–320]. Morquio syn-
splice donor site replaces an A nucleotide three drome presents with a variety of organ
bases downstream from the splice site. A purine involvements, and such patients may have short
nucleotide is necessary at that site for normal stature, dense calvaria, corneal opacities, hearing
splicing to occur [311]. Some of the patients with loss, hepatomegaly, and different bony abnormal-
this lesion are homozygous for the 2.1-kb TaqI ities. The latter may include platyspondyly, odon-
RFLP, whereas others are homozygous for the toid hypoplasia, cervical subluxation, cervical
8-kb SphI RFLP [309, 312]. Another patient has myelopathy, pectus carinatum, genu valgum, or
been described with a transversion from G to T at hip dysplasia. Fibroblasts and amniocytes (pre-
the intron 4 splice donor site that probably results natal diagnosis) have a deficiency in galac-
in defective splicing. In the Icelandic patients, a tosamine-6-sulfatase, and leukocytes and
single missense mutation results in the replace- fibroblasts show metachromasia [321]. These
ment of an aspartic acid (GAC) by valine (GTC) patients also have an increased urinary excretion
at codon 65 and has been designated D65V [310]. of keratin sulfate. Like APRTase deficiency,
In this abbreviation, D is the conventional one- Morquio syndrome is inherited as an autosomal
letter symbol for aspartic acid and V is for valine. recessive. As noted previously, APRTase
All the patients described thus far with this muta- deficiency presents primarily with renal calculi
tion have been found to be homozygous for the or renal failure due to the deposition of the insol-
2.1-kb Taq1 RFLP, and all the Icelandic patients uble adenine metabolite, 2,8-dihydroxyadenine,
have been determined to be homozygous for the in the kidney. The mutation in this individual
8-kb SphI RFLP. The single English patient in with two genetic errors has been determined to be
the group has been found to be heterozygous at the result of a deletion in the genes on chromo-
the SphI site [310, 313, 314]. Two other patients some 16 and a subsequent fusion between
showed an insertion of a T nucleotide in exon 2 sequences distal to the GALNS exon 2 and proxi-
that is likely to cause a frameshift after isoleucine mal to the APRT exon 3. The size of the deleted
at codon 61 [310]. The remainder of the non-Jap- region between GALNS intron 2 and APRT
anese patients have been shown to have princi- intron 2 is approximately 100 kb [317].
pally deletions in exon 3 or exon 5 or single base Studies of the clinical biochemistry of
changes including R67Q (R = arginine, Q = glu- APRTase deficiency have shown that approxi-
tamine), R87X (R = arginine, X = any amino acid mately 14 % of patients carrying the defect are
to which arginine can be mutated), L110P asymptomatic and only identified through family
(L = leucine, P = proline), I112F (I = isoleucine, studies in which a propositus has been docu-
F = phenylalanine), C153R (C = cysteine, R = argi- mented to have the disorder [248, 253, 259, 266,
nine), R89Q (R = arginine, Q = glutamine), and 283, 322, 323]. The inability of patients with this
M1V (M = methionine, V = valine) [310]. Most of defect to salvage adenine and its subsequent oxi-
the adenine phosphoribosyltransferase mutants dation to hydroxylated adenine metabolites by
have been reviewed in McKusick’s catalogue of xanthine oxidase results in the generation of the
human inheritance, and compound heterozygotes metabolites, 8-hydroxyadenine and 2,8-dihy-
have been recently characterized in Japan [151, droxyadenine. The latter metabolite is responsi-
315, 316]. ble for the pathologic consequences of the
Clinical investigations have recently described disease. 2,8-DHA is insoluble at any pH and par-
an unusual patient who presented with 2,8-dihy- ticularly at a near neutral pH (6.5) where its
droxyadenine urolithiasis and the Morquio syn- solubility is 1.53 ± 0.04 mg/l [323]. At pH 5.0
drome [317]. The presence of these two genetic (the approximate pH of urine), its solubility is
Adenine Phosphoribosyltransferase 53

2.68 ± 0.84 mg/l, and at an alkaline pH (7.8), its APRTase deficiency [325–327]. Through the cre-
solubility is little improved (4.97 ± 1.49 mg/l) ation of mice that are homozygous for a null
[323]. The urine contains unknown factors that APRTase allele, a mouse model for APRTase
permit supersaturation of the urine to occur, and deficiency has been developed [325, 326]. These
levels of adenine metabolites between 40 and mice excrete adenine and 2,8-dihydroxyadenine
90 mg/l have been determined [323]. just as humans do with this disorder, and the
The kidney is the primary target of the insolu- resultant renal histopathology is characterized by
ble adenine metabolite, and it is often deposited renal tubular dilation, inflammation, necrosis,
in renal tubules eliciting an inflammatory and fibrosis. Birefringent crystalline deposits are
response consisting of lymphocyte infiltrates, found in the kidney of these mice as are renal cal-
giant cell formation, and increased interstitial culi composed primarily of 2,8-DHA, the toxic
connective tissue associated with atrophied adenine metabolite. Treatment with allopurinol
tubules. In addition to the deposition of 2,8-DHA improves the life expectancy of the affected mice
in the kidney tubules, renal calculi composed of as well as the kidney function. Of interest, male
this same metabolite are often observed clini- mice show significantly more renal damage than
cally. The spectrum of stone formation ranges females, and at this time, no sound explanation
from crystalluria to the formation of urinary tract for this difference has been determined [328].
calculi and acute anuric renal failure secondary to Thus, this mouse model of nephrolithiasis, inter-
urinary tract obstruction. Yellow-brown, round stitial nephritis, and chronic renal failure provides
crystals of 2,8-DHA may be seen in the urine or a model for the investigation of interstitial nephri-
be suspected when diapers are stained with tis generated from renal calculi and more
brownish spots [314]. Stones recovered from specifically, a model that mimics human APRTase
patients react in a fashion identical to uric acid deficiency.
when tested using routine biochemical analyses. Another recent clinical finding is that patients
Colorimetric analysis with phosphomolybdate, with the autosomal dominant disorder, familial
murexide testing with nitric acid, and the alkaline juvenile hyperuricemic nephropathy (FJHN),
spectrum give identical reactions for 2,8-DHA have been found to have reduced APRTase activ-
and uric acid. When a sample of 2,8-DHA on ity in their erythrocyte lysates [187, 329, 330].
filter paper is wet with liquid nitrogen and then This disorder is characterized by the early onset
exposed to ultraviolet light, it is intensely phos- of gout and/or hyperuricemia in affected females
phorescent [324]. Ultraviolet spectra in acid and and males. The molecular basis for this APRTase
alkali, mass spectrometry, and high-performance deficiency has been identified as a single base
liquid chromatography are the usual techniques substitution in exon 3, codon 89, with the replace-
used to provide absolute identification of this ment of the normal amino acid, arginine, by glu-
metabolite and to differentiate it from uric acid tamine. This mutation has been found in two
[299, 301, 302]. The principal treatment for brothers with FJHN in one family, an affected
patients with APRTase deficiency is the hypoxan- female in another family, and a mother and son
thine analog, allopurinol, which causes a redistri- from a family with severe combined
bution of the urinary metabolites since it inhibits immunodeficiency disease, and an unaffected
xanthine oxidase and blocks the conversion of mother and her son in another family with FJHN.
adenine to its hydroxylated metabolites. Thus, a single base substitution in the APRTase
Therefore, patients treated with allopurinol gene has been identified in three of nine families
excrete principally adenine rather than the toxic with FJHN. A survey of 500 randomly selected
metabolite 2,8-DHA. No alkalinization is required DNA samples did not identify this abnormality,
to increase the solubility of the small quantities but it remains unclear as to whether this genetic
of the hydroxylated adenine metabolites since alteration has any clinical significance or whether
alkali does not alter the solubility of these com- it is simply an incidental finding and bears no
pounds. Two recent findings show promise for a relation to the underlying disorder in purine
better understanding of the renal pathology in metabolism.
54 3 Purine Biochemistry

The other issue related to purine metabolism genetic defect, the increased APRTase activity
is the role of the enzyme, APRTase, in the control has been determined to be related to the increased
of the first committed step in de novo purine bio- synthesis of APRTase rather than diminished
synthesis. Since adenosine monophosphate degradation [334]. For these reasons, it may be
(AMP) is a product of the salvage enzyme, reasonable to reexamine the proposed role of
APRTase, and this product is also a negative increased APRTase stabilization by PRPP.
feedback inhibitor of PRPP amidotransferase, the
absence of APRTase activity should lead to the
reduced formation of AMP and increased quanti- Isotopes in the Evaluation of Uric Acid
ties of PRPP available for other reactions. Metabolism
Reduced levels of AMP and increased availabil-
ity of PRPP could also increase purine synthesis Before leaving the biochemistry of purines, sev-
by the de novo pathway. The latter could occur eral additional aspects should be discussed. Since
through the mechanism of either decreased feed- glycine supplies carbon atoms 4 and 5 and nitro-
back regulation of the first committed enzyme, gen atom 7 of the purine ring, this simple amino
PRPP amidotransferase, or the use of the excess acid can be used to assess the rate of purine syn-
PRPP as a substrate for the conversion of phos- thesis in vivo or in vitro. By measuring the rate of
phoribosylpyrophosphate to phosphoribosylam- incorporation of labeled glycine-1-C14 or glycine
ine. No patients with APRTase deficiencies have alpha-N15 into uric acid or other purine end prod-
been shown to excrete increased quantities of ucts, an estimation of the rate of purine synthesis
uric acid, and this observation would suggest that can be determined. Isotopes can also be utilized
decreased quantities of AMP do not have a to determine uric acid turnover rates, purine deg-
significant role in the regulation of the de novo radation rates, and the rates of flow through
pathway in these patients. Further, the increased nucleotide pools. Studies using these isotope
quantities of PRPP available as a result of its methodologies were the first to suggest that
underutilization for adenine salvage appear not to beyond measuring the quantity of uric acid in the
drive purine synthesis more rapidly via the urine, the rate of purine synthesis could be shown
de novo pathway. Thus, the deficiencies of ade- to be increased in patients with gout who excreted
nine phosphoribosyltransferase observed in more than the normal quantity of uric acid. A
patients with familial juvenile hyperuricemic variety of isotopes have been used to measure
nephropathy and their relationship to the defect purine production including glycine-2-C14, for-
in uric acid metabolism in these families remain mate-C14, 4-aminoimadazole-5-carboxamide-C14,
to be determined. It would also appear that the and ammonium-N15, whereas hypoxanthine-8-C14
adenine salvage pathway plays no significant role and adenine-8-C14 have been utilized to measure
in the regulation of de novo purine biosynthesis. purine degradation and flow through the nucle-
Further, there is evidence that the increased level otide pools. Radiolabeled adenine incorporated
of adenine phosphoribosyltransferase activity into the adenine nucleotide pool permits the mea-
observed in those patients with diminished or surement of adenine degradation through the
absent hypoxanthine-guanine phosphoribosyl- quantitation of radiolabeled products in the urine
transferase activity may not be solely related to [335, 336].
stabilization of the former enzyme by phosphori- To summarize the significance of the de novo
bosylpyrophosphate. Erythrocyte APRTase activ- purine biosynthetic pathway and its regulation in
ity and PRPP levels are elevated in patients with humans with gout, accelerated production of uric
purine nucleoside phosphorylase deficiency and acid can occur on the basis of three fundamental
defective cell-mediated immunity [331, 332], and mechanisms: (1) increased concentrations of the
erythrocyte APRTase activity is also elevated in specific substrate, PRPP, available to the rate-
patients with the pyrimidine abnormality, heredi- limiting step in purine synthesis catalyzed by the
tary orotic aciduria [333, 334]. In the latter enzyme, PRPP amidotransferase, (2) alterations
References 55

in the regulatory properties of the PRPP amido- 3. Kelley WN, Weiner IM, editors. Uric acid. Berlin:
transferase enzyme causing an increase in its Springer; 1978.
4. Elliott K, Fitzsimons DW, editors. Purine and pyrimi-
activity, or (3) alterations in the concentrations of dine metabolism, Ciba Foundation symposium 48.
regulatory nucleotides, adenylic or guanylic acid, Amsterdam: Elsevier; 1977.
resulting either in an imbalance in the two or a 5. Rosenbloom FM, Henderson JF, Caldwell JC, Kelley
decrease in their concentrations. Although the WN, Seegmiller JE. Biochemical bases of accelerated
purine biosynthesis de novo in human fibroblasts
activities of glutamine synthetase and glutami- lacking hypoxanthine-guanine phosphoribosyltrans-
nase have been examined as putative contributors ferase. J Biol Chem. 1968;243:1166.
to an increased rate of purine synthesis, no abnor- 6. Sidi Y, Mitchell BS. Z-nucleotide accumulation in
malities have been identified, and it is unlikely erythrocytes from Lesch-Nyhan patients. J Clin Invest.
1985;76:2416.
that alterations in l-glutamine, the other substrate 7. Newcombe DS, Lapes M, Thomson C, Wright EY.
for PRPP amidotransferase, are responsible for Urinary excretion of 4-amino-5-imidazolecarboxam-
the gouty diathesis. As will be discussed in some ide in X-linked primary hyperuricemia. Clin Res.
detail in the clinical chapters of this book, the 1967;15:45.
8. Fox IH, Kelley WN. The role of adenosine and deoxy-
abnormalities in the enzymes, PRPP synthetase adenosine in mammalian cells. Annu Rev Biochem.
and hypoxanthine-guanine phosphoribosyltrans- 1978;47:655.
ferase, that are associated with gout and those 9. Becker MA, Meyer LJ, Huisman WH, Lazar C,
individuals with deficiencies in glucose-6- Adams WB. Human erythrocyte phosphoribosylpyro-
phosphate synthetase: subunit analysis and states of
phophatase (glycogen storage disease, type I) fall subunit association. J Biol Chem. 1977;252:3911.
into the first mechanism cited since these defects 10. Meyer LJ, Becker MA. Human erythrocyte phospho-
lead to increased amounts of PRPP for utilization ribosylpyrophosphate synthetase: dependence of
in the de novo pathway. Even in the clinical set- activity on state of subunit association. J Biol Chem.
1977;252:3919.
ting where there is an increased turnover of 11. Becker MA, Yen RCK, Itkin P. Regional localization
nucleic acids such as one might observe in an of the gene for human phosphoribosylpyrophosphate
aggressive malignancy, the increased rate of synthetase on the X-chromosome. Science. 1979;203:
incorporation of nucleotides into polynucleotides 1016.
12. Wood AW, Becker MA, Seegmiller JE. Purine nucle-
leads to a dampening of feedback inhibition via otide synthesis in lymphoblasts cultured from normal
the PRPP amidotransferase enzyme as a result of subjects and a patient with Lesch-Nyhan syndrome.
the overutilization of nucleotides for polynucle- Biochem Genet. 1973;9:261.
otide synthesis. This is consistent with the second 13. Losman MJ, Hecker S, Woo S, Becker MA. Diagnostic
evaluation of phosphoribosylpyrophosphate syn-
mechanism cited. The third mechanism is illus- thetase activities in hemolysates. J Lab Clin Med.
trated by the disorders associated with a deficiency 1984;103:932.
in the activity of the enzyme, hypoxanthine-gua- 14. Sperling O, Eilam G, Persky-Brosh S, de Vries A.
nine phosphoribosyltransferase, and the dimin- Accelerated erythrocyte 5-phosphoribosyl-1-pyro-
phosphate synthesis. A familial abnormality associ-
ished production of GMP and IMP, feedback ated with excessive uric acid production and gout.
inhibitors of purine synthesis. The role of adenine Biochem Med. 1972;6:310.
phosphoribosyltransferase in familial juvenile 15. Nyhan WL, James JA, Teberg AJ, Sweetman L,
hyperuricemic nephropathy remains unexplained Nelson LG. A new disorder of purine metabolism
with behavioral manifestations. J Pediatr. 1969;
at this time. It may simply be an incidental finding 74:20.
or it may have some pathogenetic significance 16. Rosenberg AL, Bergstrom L, Troost BT,
yet to be defined. Bartholomew BA. Hyperuricemia and neurologic
deficits. N Engl J Med. 1970;282:992.
17. Becker MA, Raivio KO, Bakay B, Adams WB, Nyhan
WL. Variant human phosphoribosylpyrophosphate
References synthetase altered in regulatory and catalytic func-
tions. J Clin Invest. 1980;65:109.
1. Colowick SP, Kaplan NO. Methods in enzymology, 18. Becker MA. Phosphoribosylpyrophosphate synthetase
vol. 6. New York: Academic; 1963. superactivity: detection, characterization of underly-
2. Chargaff E, Davidson JN. The nucleic acids, vol. 3. ing defects and treatment. Adv Exp Med Biol. 1984;
New York: Academic; 1963. 165A:91.
56 3 Purine Biochemistry

19. Simmonds HA, Webster DR, Wilson J, Lingham S. 33. Henderson JF, Rosenbloom FM, Kelley WN,
An X-linked syndrome characterized by hyperurice- Seegmiller JE. Variations in purine metabolism of
mia, deafness, and neurological abnormalities. Lancet. cultured skin fibroblasts from patients with gout.
1982;2:68. J Clin Invest. 1968;47:1511.
20. Becker MA, Losman MJ, Rosenberg AL, Mehlman I, 34. Becker MA, Smith PR, Taylor W, Mustafi R,
Levinson DJ, Holmes EW. Phosphoribosylpyro- Switzer RL. The genetic and functional basis of purine
phosphate synthetase superactivity. Arthritis Rheum. nucleotide feedback-resistant phosphoribosylpyro-
1986;29:880. phosphate synthetase superactivity. J Clin Invest.
21. Becker MA, Puig JG, Mateos FA, Jimenez ML, 1995;906:2133–41.
Kim M, Simmonds HA. Inherited superactivity of 35. Lejeune E, Bouvier M, Mousson B, Llorca G,
phosphoribosylpyrophosphate synthetase: association Baltassat P. Anomalies de la phosphoribosylpyro-
of uric acid overproduction and sensorineural deaf- phosphate synthetase dans deux cas de goutte a debut
ness. Am J Med. 1988;85:383. precose. Rev Rhum. 1979;46:457.
22. Christen H-J, Hanefeld F, Duley JA, Simmonds HA. 36. Roessler BJ, Golovoy N, Palella TD, Heidler S,
Distinct neurological syndrome in two brothers with Becker MA. Identification of distinct PRS1 mutations
hyperuricaemia. Lancet. 1992;340:1167. in two patients with X-linked phosphoribosylpyro-
23. Becker MA, Meyer LJ, Seegmiller JE. Gout with phosphate synthetase overactivity. Adv Exp Med Biol.
purine overproduction due to increased phosphoribo- 1991;309B:125.
sylpyrophosphate synthetase activity. Am J Med. 37. Ishizuka T, Iizasa T, Taiara M, Ishijuma S, Sonada T,
1973;55:232. Shimada H, Nagatake N, Tatibana M. Promoter
24. Becker MA. Patterns of phosphoribosylpyrophos- regions of the human X-linked housekeeping gene
phate and ribose-5-phosphate concentration and gen- PRPS1 and PRPS2 encoding phosphoribosylpyro-
eration in fibroblasts from patients with gout and phosphate synthetase subunit I and II isoforms.
purine over-production. J Clin Invest. 1976;57:308. Biochim Biophys Acta. 1992;1130:139–48.
25. Akaoka I, Fujimori S, Kamatani N, Takeuchi F, 38. Becker MA, Losman MJ, Kim M. Mechanisms of
Yano E, Nishida Y, et al. A gouty family with increased accelerated purine nucleotide synthesis in human
phosphoribosylpyrophosphate synthetase activity: fibroblasts with superactive phosphoribosylpyrophos-
case reports, family studies, and kinetic studies of the phate synthetase. J Biol Chem. 1987;262:5596–602.
abnormal enzyme. J Rheumatol. 1981;8:563. 39. Becker MA, Taylor W, Smith PR, Ahmed M.
26. Becker MA, Losman MJ, Itkin P, Simkin PA. Gout Overexpression of the normal phosphoribosylpyro-
with superactive phosphoribosylpyrophosphate syn- phosphate synthetase 1 isoform underlies catalytic
thetase due to increased enzyme catalytic rate. J Lab superactivity of human phosphoribosylpyrophosphate
Clin Med. 1982;99:485. synthetase. J Biol Chem. 1996;271:19894–9.
27. Becker MA, Losman MJ, Wilson J, Simmonds HA. 40. Taira M, Ishijima S, Kita K, Yamada K, Iizasa T,
Superactivity of human phosphoribosylpyrophosphate Tatibana M. Nucleotide and deduced amino acid
synthetase due to altered regulation by nucleotide sequences of two distinct cDNAs for rat phosphoribo-
inhibitors and inorganic phosphate. Biochim Biophys sylpyrophosphate synthetase. J Biol Chem. 1987;262:
Acta. 1986;882:168. 14867.
28. Wada Y, Nishimura Y, Tanabu M, Yoshimura Y, 41. Taira M, Kudoh J, Minoshima S, Ikzasa T, Shimada H,
Iinuma K, Yoshida T, Arakawa T. Hypouricemic, Shimizu Y, Tatibana M, Shimizu N. Localization of
mentally retarded infant with a defect of 5-phosphori- human phosphoribosylpyrophosphate synthetase sub-
bosyl-1-pyrophosphate synthetase of erythrocytes. unit I and II genes (PRPS1 and PRPS2) to different
Tohoku J Exp Med. 1974;113:149. regions of the X chromosome and assignment of two
29. Iinuma K, Wada Y, Onuma A, Tanabu M. PRPS1-related genes to autosomes. Somat Cell Mol
Electroencephalographic study of an infant with phos- Genet. 1989;15:29.
phoribosylpyrophosphate synthetase deficiency. 42. Becker MA, Heidler SA, Bell GI, Seino S,
Tohoku J Exp Med. 1975;116:53. Le Beau MM, Westbrook CA, Neuman W, Shapiro
30. Becker MA, Kostel PJ, Meyer LJ, Seegmiller JE. LJ, Mohandas TK, Roessler BJ, Palella TD. Cloning
Human phosphoribosylpyrophosphate synthetase: of cDNAs for human phosphoribosylpyrophosphate
increased enzyme specific activity in a family with synthetases 1 and 2 and X chromosome localization
gout and excessive purine synthesis. Proc Natl Acad of PRPS1 and PRPS2 genes. Genomics. 1990;8:555.
Sci USA. 1973;70:2749. 43. Sonoda T, Taira M, Ishijima S, Ishjizuka T, Iizasa T,
31. Hershko A, Hershko C, Mager J. Increased formation Tatibana M. Complete nucleotide sequence of human
of 5¢-phosphoribosyl-1-pyrophosphate in red blood phosphoribosylpyrophosphate synthetase subunit I
cells of some gouty patients. Isr J Med Sci. 1968; (PRPS1) cDNA and a comparison with human and rat
4:939. PRPS gene families. J Biochem. 1991;109:361.
32. Sperling O, Persky-Brosh S, Boer P, De Vreis A. 44. Iizasa T, Taira M, Shimada H, Ishijima S, Tatibana M.
Human erythrocyte phosphoribosylpyrophosphate Molecular cloning and sequencing of human cDNA
synthetase mutationally altered in regulatory proper- for phosphoribosylpyrophosphate synthetase subunit
ties. Biochem Med. 1973;7:389. II. FEBS Lett. 1989;244:47.
References 57

45. Taira M, Iizasa T, Yamada K, Shimada H, Tatibana M. 59. Henderson JF, Caldwell IE, Paterson ARP. Decreased
Tissue-differential expression of two distinct genes feedback inhibition of a 6-methylmercaptopurine
for phosphoribosylpyrophosphate synthetase and resistant tumor. Cancer Res. 1967;27:1773.
existence of the testis-specific transcript. Biochim 60. Oates DC, Vannais D, Patterson D. A mutant of CHO-
Biophys Acta. 1989;1007:203. K1 cells deficient in two nonsequential steps of
46. Taira M, Iizasa T, Shimada H, Kudoh J, Shimizu N, de novo purine biosynthesis. Cell. 1980;20:757.
Tatibana M. A human testis-specific mRNA for phos- 61. Kim JH, Krahn JM, Tomchick DR, Smith JL, Zalkin H.
phoribosylpyrophosphate synthetase that initiates Structure and function of the glutamine phosphoribo-
from a non-AUG codon. J Biol Chem. 1990;265: sylpyrophosphate amidotransferase glutamine site
16491. and communication with the phosphoribosylpyro-
47. Becker MA, Roessler BJ. Hyperuricemia and gout. phosphate site. J Biol Chem. 1996;271:15549–57.
In: Scriver CR, Beudet AL, Sly WS, Valle D, editors. 62. Kim JH, Wolle D, Haridas K, Parry RJ, Smith JL,
The metabolic and molecular bases of inherited dis- Zalkin H. A stable carbocyclic analog of 5-phosphor-
ease, vol. II. New York: McGraw-Hill, Inc.; 1995. ibosyl-1-pyrophosphate to probe the mechanism of
1670p. catalysis and regulation of glutamine phosphoribo-
48. Nosal JM, Switzer RL, Becker MA. Overexpression, sylpyrophosphate amidotransferase. J Biol Chem.
purification, and characterization of recombinant 1995;270:17394–9.
human 5-phosphoribosyl-1-pyrophosphate synthetase 63. IwahanaH,ItakuraM.Amidophosphoribosyltransferase.
isozymes I and II. J Biol Chem. 1993;268:10168. Nippon Rinsho. 1996;54:3202–6.
49. Roessler BJ, Nosal JM, Smith PR, Heidler SA, Palella 64. Smith JL. Enzymes of nucleotide synthesis. Curr Opin
TD, Switzer RL, Becker MA. Human X-linked phos- Struct Biol. 1995;5:752–7.
phoribosylpyrophosphate synthetase superactivity is 65. Smith JL, Zaluzec EJ, Wery JP, Niu L, Switzer RL,
associated with distinct point mutations in the PRPS1 Zalkin H, Satow Y. Structure of the allosteric regula-
gene. J Biol Chem. 1993;268:26476. tory enzyme of purine biosynthesis. Science.
50. Holmes EW, McDonald JA, McCord JM, Wyngaarden 1994;264:1427–33.
JW, Kelley WN. Human glutamine phosphoribosylpy- 66. Iwahana H, Oka J, Mizusawa N, Kudo E, Ii S,
rophosphate amidotransferase: kinetic and regulatory Yoshimoto K, Holmes EW, Itakura M. Molecular
properties. J Biol Chem. 1973;248:144. cloning of human amidophosphoribosyltransferase.
51. Holmes EW. Kinetic, physical, and regulatory proper- Biochem Biophys Res Commun. 1993;190:192–200.
ties of amidophosphoribosyltransferase. Adv Enzyme 67. Chen S, Tomchick DR, Walle D, Hu P, Smith JL,
Regul. 1981;44:215. Switzer RL, Zalkin H. Mechanism of the synergistic
52. Caskey CT, Ashton DM, Wyngaarden JB. The enzy- end-product regulation of Bacillus subtilis glutamine
mology of feedback inhibition of glutamine phospho- phosphoribosylpyrophosphate amidotransferase by
ribosylpyrophosphate amidotransferase by purine nucleotides. Biochemistry. 1997;36:10718–26.
nucleotides. J Biol Chem. 1964;239:2570. 68. Krahn JM, Kim JH, Burns MR, Parry RJ, Zalkin H,
53. Becker MA, Kim M. Regulation of purine synthesis Smith JL. Coupled formation of an amidotransferase
de novo in human fibroblasts by purine nucleotides interdomain ammonia channel and phosphoribosyl-
and phosphoribosylpyrophosphate. J Biol Chem. transferase active site. Biochemistry. 1997;36:11061–8.
1987;262:14531. 69. Meschmore CR, Krahn JM, Kim JH, Zalkin H,
54. Wood AW, Seegmiller JE. Properties of 5-phosphori- Smith JL. Crystal structure of glutamine phosphoribo-
bosyl-1-pyrophosphate amidotransferase from human sylpyrophosphate amidotransferase from Escherichia
lymphoblasts. J Biol Chem. 1973;248:138. coli. Protein Sci. 1998;7:39–51.
55. Singer SC, Holmes EW. Human glutamine phosphori- 70. Zalkin H, Smith JL. Enzymes utilizing glutamine as
bosylpyrophosphate amidotransferase: hysteretic an amide donor. Adv Enzymol Relat Areas Mol Biol.
properties. J Biol Chem. 1977;252:7959. 1998;72:87–144.
56. Holmes EW, Wyngaarden JB, Kelley WN. Human 71. Smith JL. Glutamine PRPP amidotransferase: snap-
glutamine phosphoribosylpyrophosphate amidotrans- shots of an enzyme in action. Curr Opin Struct Biol.
ferase: two molecular forms interconvertible by purine 1998;8:686–94.
ribonucleotides and phosphoribosylpyrophosphate. 72. Bera AK, Chen S, Smith JL, Zalkin H. Interdomain sig-
J Biol Chem. 1973;248:6035. naling in glutamine phosphoribosylpyrophosphate
57. Stanley W, Chu EHY. Assignment of the gene for amidotransferase. J Biol Chem. 1999;274:36498–504.
phosphoribosylpyrophosphate amidotransferase to 73. Bera AK, Smith JL, Zalkin H. Dual role for the glu-
the pter → q21 region of human chromosome 4. tamine phosphoribosylpyrophosphate amidotrans-
Cytogenet Cell Genet. 1978;22:228. ferase ammonia channel. Interdomain signaling and
58. Nagy M. Regulation of the biosynthesis of purine intermediate channeling. J Biol Chem. 2000;275:
nucleotides in Schizosaccharomyces pombe. I. Properties 7975–9.
of phosphoribosylpyrophosphate: glutamine amido- 74. Li S, Smith JL, Zalkin H. Mutational analysis of
transferase of the wild strain and of a mutant desensi- Bacillus subtilis glutamine phosphoribosylpyrophos-
tized towards feedback modifiers. Biochim Biophys phate amidotransferase propeptide processing.
Acta. 1970;198:471. J Bacteriol. 1999;181:1403–8.
58 3 Purine Biochemistry

75. Brayton KA, Chen Z, Zhon G, Nagy P, Gavalas A, 90. Holden JA, Kelley WN. Human hypoxanthine-guanine
Trent JM, Deaven LL, Dixon JE, Zalkin H. Two genes phosphoribosyltransferase. Evidence for tetrameric
for de novo purine nucleotide synthesis on human structure. J Biol Chem. 1978;253:4459.
chromosome 4 are closely linked and divergently 91. Eads JC, Scapin G, Xu Y, Grubmeyer C, Sacchettini
transcribed. J Biol Chem. 1994;269:5313–21. JC. The crystal structure of human hypoxanthine-
76. Newcombe DS. Unpublished observations. guanine phosphoribosyltransferase with bound
77. Gutensohn W, Guroff G. Hypoxanthine guanine phos- GMP. Cell. 1994;78:325–34.
phoribosyltransferase from rat brain (purification, 92. Xu Y, Eads J, Sacchettini JC, Grubmeyer C. Kinetic
kinetic properties, development, and distribution). mechanism of human hypoxanthine-guanine phos-
J Neurochem. 1972;19:2139. phoribosyltransferase: rapid phosphoribosyl transfer
78. Adams A, Harkness RA. Developmental changes in chemistry. Biochemistry. 1997;36:3700–12.
purine phosphoribosyltransferases in human and rat 93. Henderson JF, Brox LW, Kelley WN, Rosenbloom
tissues. Biochem J. 1976;100:565. FM, Seegmiller JE. Kinetic studies of hypoxanthine-
79. Rosenbloom FM, Kelley WN, Miller J, Henderson JF, guanine phosphoribosyltransferase. J Biol Chem.
Seegmiller JE. Inherited disorder of purine metabo- 1968;243:2514.
lism: correlation between central nervous system dys- 94. McDonald JA, Kelley WN. Lesch-Nyhan syndrome:
function and biochemical defects. JAMA. 1967; altered kinetic properties of mutant enzyme. Science.
202:175. 1971;171:689.
80. Watts RWE, Harkness RA, Spellacy E, Taylor NF. 95. Keough DT, McConachie LA, Gordon RB, de Jersey
Lesch-Nyhan syndrome: growth delay, testicular atro- J, Emmerson BT. Human hypoxanthine phosphori-
phy a partial failure of the 11b-hydroxylation of ste- bosyltransferase. Development of a spectrometric
roids. J Inherit Metab Dis. 1987;10:210. assay and its use in detection and characterization of
81. DeBruyn CHMM, Oei TL. Purine phosphoribosyl- mutant forms. Clin Chim Acta. 1987;163:301.
transferase in human erythrocyte ghosts. Adv Exp 96. Bakay B, Nyhan WL. Heterogeneity of hypoxan-
Med Biol. 1977;76A:139. thine guanine phosphoribosyltransferase from
82. Yip LC, Chang V, Balis EM. Membrane-associated human erythrocytes. Arch Biochem Biophys. 1975;
purine metabolizing enzyme activities of human 168:26.
peripheral blood cells. Biochemistry. 1982;21:6972. 97. Olsen AS, Milman G. Subunit molecular weight of
83. Melton DW, Konecki DS, Ledbetter DH, Hejtmancik human hypoxanthine guanine phosphoribosyltrans-
JF, Caskey CT. In vitro translation of hypoxanthine ferase. J Biol Chem. 1974;249:4038.
guanine phosphoribosyltransferase mRNA: character- 98. Olsen AS, Milman G. Human hypoxanthine phos-
ization of a mouse neuroblastoma cell line that has phoribosyltransferase: purification and properties.
elevated levels of hypoxanthine guanine phosphoribo- Biochemistry. 1977;21:960.
syltransferase protein. Proc Natl Acad Sci USA. 99. Davies MR, Dean BM. The heterogeneity of eryth-
1981;78:6977. rocyte IMP: pyrophosphate phosphoribosyltrans-
84. Allsop J, Watts RWE. Activities of amidophosphori- ferase and purine nucleoside phosphorylase by
bosyltransferase (EC 2.4.2.14) and the purine phos- isoelectric focusing. FEBS Lett. 1971;18:283.
phoribosyltransferases (EC 2.4.2.7 and 2.4.2.8) and 100. Shi W, Li CM, Tyler PC, Furneaux RH, Grubmeyer
the phosphoribosylpyrophosphate content of rat cen- C, Schramm VL, Almo SC. The 2.0 A structure of
tral nervous system at different stages of development. human hypoxanthine-guanine phosphoribosyltrans-
J Neurol Sci. 1980;46:221. ferase in complex with a transition-state analog
85. Krenitsky TA, Papaioannou R, Elion GB. Human inhibitor. Nat Struct Biol. 1999;6:588–93.
hypoxanthine phosphoribosyltransferase I. Purification, 101. Lee CC, Craig 3rd SP, Eakin AE. A single amino
properties and specificity. J Biol Chem. 1969;244:1263. acid substitution in the human and a bacterial hypox-
86. Salerno C, Giacomello A. Human hypoxanthine gua- anthine phosphoribosyltransferase modulates
nine phosphoribosyltransferase. The role of magne- specificity for the binding of guanine. Biochemistry.
sium ion in a phosphoribosylpyrophosphate-utilizing 1998;37:3491–8.
enzyme. J Biol Chem. 1981;256:3671. 102. Xu Y, Grubmeyer C. Catalysis in human hypoxan-
87. Wilson JM, Tarr GE, Mahoney WC, Kelley WN. thine-guanine phosphoribosyltransferase: Asp 137
Human hypoxanthine-guanine phosphoribosyltrans- acts as a general acid/base. Biochemistry. 1998;37:
ferase deficiency. Complete amino acid sequence of 4114–24.
the erythrocyte enzyme. J Biol Chem. 1982;257: 103. Balendiran GK, Molina JA, Xu Y, Torres-Martinez J,
10987. Stevens R, Focia PJ, Eakin AE, Sacchettini JC, Craig
88. Strauss M, Behlke J, Ampers F, Goeri M. Evidence 3rd SP. Ternary complex structure of human
against the existence of real isoenzymes of hypoxan- HGPRTase, PRPP, Mg2+, and the inhibitor HPP
thine phosphoribosyltransferase. Eur J Biochem. reveals the involvement of the flexible loop in sub-
1978;90:89. strate binding. Protein Sci. 1999;8:1023–31.
89. Johnson GG, Eisenberg LR, Migeon BR. Human and 104. Chiaverotti TA, Battula N, Monnat Jr RJ. Rat hypox-
mouse hypoxanthine guanine phosphoribosyltrans- anthine phosphoribosyltransferase cDNA cloning
ferases dimers and tetramers. Science. 1979;203:174. and sequence analysis. Genomics. 1991;11:1158.
References 59

105. Argos P, Hanei M, Wilson JM, Kelley WN. A pos- 117. Nicklas JA, Hunter TC, O’Neill JP, Albertini RJ.
sible nucleotide-binding domain in the tertiary fold Fine structure mapping of the hypoxanthine-guanine
of phosphoribosyltransferases. J Biol Chem. 1983; phosphoribosyltransferase (HPRT) gene region of
258:6430. the human X chromosome (Xq26). Am J Hum
106. Dush MK, Sikela JM, Khan SA, Tischfield JA, Genet. 1991;49:267.
Stambrook PJ. Nucleotide sequence and organiza- 118. Reilly DS, Lewis RA, Nussbaum RL. Genetic and
tion of the mouse adenine phosphoribosyltransferase physical mapping of Xq24-q26 markers flanking the
gene: presence of a coding region common to animal Lowe oculocerebrorenal syndrome. Genomics.
and bacterial phosphoribosyltransferases that has 1990;8:62.
variable intron/exon arrangement. Proc Natl Acad 119. Patel PI, Nussbaum RL, Framson PE, Ledbetter DH,
Sci USA. 1985;82:2731. Caskey CT, Chinault AC. Organization of the HPRT
107. Hershey HV, Taylor MW. Nucleotide sequence and gene and related sequences in the human genome.
deduced amino acid sequence of Escherichia coli Somat Cell Mol Genet. 1984;10:483.
adenine phosphoribosyltransferase and comparison 120. Edwards A, Voss H, Rice P, Civitello A, Stegemann J,
with other analogous enzymes. Gene. 1986;43:287. Schwager C, Zimmermann J, Edrfle H, Caskey CT,
108. Wilson JM, Young AB, Kelley WN. Hypoxanthine- Ansorge W. Automated DNA sequencing of the
guanine phosphoribosyltransferase deficiency. The human HPRT locus. Genomics. 1990;6:593.
molecular basis of the clinical syndromes. N Engl J 121. Dynan WS, Sazer S, Tjian R, Schimke RT.
Med. 1983;309:900. Transcription factor Spl recognizes a DNA sequence
109. Wilson JM, Kobayashi R, Fox IH, Kelley WN. in the mouse dihydrofolate reductase promoter.
Human hypoxanthine-guanine phosphoribosyltrans- Nature. 1986;319:246.
ferase. Molecular abnormality in a mutant form of 122. Patel PI, Framson PE, Caskey CT, Chinault AG. Fine
the enzyme (HPRTToronto). J Biol Chem. 1983;258: structure of the human hypoxanthine phosphoribo-
6458. syltransferase gene. Mol Cell Biol. 1986;6:393.
110. Davidson BL, Tarle SA, Palella TD, Kelley WN. 123. Kim SH, Moores JC, David D, Respess JG, Jolly DJ,
Molecular basis of hypoxanthine-guanine phospho- Freidmann T. The organization of the human HPRT
ribosyltransferase deficiency in ten subjects deter- gene. Nucleic Acids Res. 1986;14:3103.
mined by direct sequencing of amplified transcripts. 124. Rincon-Limas DE, Krueger DA, Patel PI. Functional
J Clin Invest. 1989;84:342. characterization of the human hypoxanthine phospho-
111. Wilson JM, Kelley WN. Human hypoxanthine- ribosyltransferase gene promoter: evidence for a nega-
guanine phosphoribosyltransferase. Structural alter- tive regulatory element. Mol Cell Biol. 1991;11:4157.
ation in a dysfunctional enzyme variant (HPRT 125. Johnson P, Friedmann T. Limited bidirectional activ-
Munich) isolated from a patient with gout. J Biol ity of two housekeeping promoters: human HPRT
Chem. 1984;259:27. and PGK. Gene. 1990;88:207.
112. Wilson JM, Tarr GE, Kelley WN. Human hypoxan- 126. Sykes RC, Lin D, Hwang SJ, Framson PE, Chinault
thine (guanine) phosphoribosyltransferase: an amino AC. Yeast ARS function and nuclear matrix associa-
acid substitution in a mutant form of the enzyme iso- tion coincide in a short sequence from the human
lated from a patient with gout. Proc Natl Acad Sci HPRT locus. Mol Gen Genet. 1988;212:301.
USA. 1983;89:870. 127. Davidson JD, Bradley TR, Roosa RA, Law LW.
113. Jolly DJ, Okayama H, Berg P, Esty AC, Filpula D, Purine nucleotide pyrophosphorylases in 8-azagua-
Bohlen P, Johnson GG, Shively JE, Hunkapillar T, nine-sensitive and resistant P 388 leukemias. Natl
Friedmann T. Isolation and characterization of a full- Cancer Inst Monogr. 1962;29:789.
length expressible cDNA for human hypoxanthine 128. Littlefield JW. The inosinic acid pyrophosphorylase
phosphoribosyltransferase. Proc Natl Acad Sci USA. activity of mouse fibroblasts partially resistant to
1983;80:477. 8-azaguanine. Proc Natl Acad Sci USA. 1963;50:
114. Francke U, Taggart RT. Comparative gene mapping: 568.
order of loci on the X chromosome is different in 129. Brockman RW, Debovadi CS, Stutts P, Hutchinson
mice and humans. Proc Natl Acad Sci USA. 1980;77: DJ. Purine ribonucleotide pyrophosphorylase and
3595. resistance to purine analogues in Streptococcus
115. Shows TB, Brown JA. Localization of genes coding faecalis. J Biol Chem. 1961;236:1471.
for PGK, HPRT, and G6PD on the long arm of the X 130. Seegmiller JE, Rosenbloom FM, Kelley WN. An
chromosome in somatic hybrids. Cytogenet Cell enzyme defect associated with a sex-linked human
Genet. 1975;14:426. neurological disorder and excessive purine synthe-
116. Pai GS, Sprenkle JA, Do TT, Mareni CE, Migeon sis. Science. 1967;155:1682.
BR. Localization of loci for hypoxanthine phospho- 131. Kelley WN, Rosenbloom FM, Henderson JR,
ribosyltransferase and glucose-6-phosphate dehy- Seegmiller JE. A specific enzyme defect in gout
drogenase and biochemical evidence of nonrandom associated with overproduction of uric acid. Proc
X chromosome expression from studies of a human Natl Acad Sci USA. 1967;57:1735.
X-autosome translocation. Proc Natl Acad Sci USA. 132. Bakay B, Nyhan WL. Activation of variants of
1980;77:2810. hypoxanthine-guanine phosphoribosyltransferase by
60 3 Purine Biochemistry

the normal enzyme. Proc Natl Acad Sci USA. 147. Tarle SA, Davidson BL, Wu VC, Zidar FJ,
1972;69:2523. Seegmiller JE, Kelley WN, Palella TD. Deter-
133. Bakay B, Nyhan WL. Electrophoretic properties of mination of the mutations responsible for the
hypoxanthine-guanine phosphoribosyltransferase in Lesch-Nyhan syndrome in 17 subjects. Genomics.
erythrocytes of subjects with Lesch-Nyhan syn- 1991;10:499.
drome. Biochem Genet. 1972;6:139. 148. Wehnert M, Herrmann FH. Characterization of three
134. Kelley WN, Meade JC. Studies on hypoxanthine- new deletions at the 5¢ end of the HPRT structural
guanine phosphoribosyltransferase in fibroblasts gene. J Inherit Metab Dis. 1990;13:178.
from patients with the Lesch-Nyhan syndrome: evi- 149. Igarashi T, Minami M, Nishada Y. Molecular analy-
dence for genetic heterogeneity. J Biol Chem. sis of hypoxanthine-guanine phosphoribosyltrans-
1971;246:2953. ferase mutations in five unrelated Japanese patients.
135. Arnold WJ, Meade JC, Kelley WN. Hypoxanthine- Acta Paediatr Jpn. 1989;31:303.
guanine phosphoribosyltransferase: characteristics 150. Gibbs RA, Nguyen PN, McBride LJ, Koepf SM,
of the mutant enzyme in erythrocytes from patients Caskey CT. Identification of mutations leading to the
with the Lesch-Nyhan syndrome. J Clin Invest. Lesch-Nyhan syndrome by automated direct DNA
1972;51:1805. sequencing of in vitro amplified cDNA. Proc Natl
136. Benke PM, Herrick N. Azaguanine-resistance as a Acad Sci USA. 1989;86:1919.
manifestation of a new form of metabolic overpro- 151. Yang TP, Patel PI, Chinault AC, Jackson LG,
duction of uric acid. Am J Med. 1972;52:547. Hildebrand BM, Caskey CT. Molecular evidence for
137. Gutensohn W, Jahn H. Partial deficiency of hypox- new mutation at the hprt locus in Lesch-Nyhan
anthine-phosphoribosyltransferase: evidence for a patients. Nature. 1984;310:412.
structural mutation in a patient with gout. Eur J Clin 152. McKusick VA, Francomano CA, Antonarakis SE.
Invest. 1979;9:43. Mendelian inheritance in man. Catalogs of auto-
138. Sperling O, Boer P, Eilam G, DeVries A. Altered somal dominant, autosomal recessive, and X-linked
kinetic properties of erythrocyte phosphoribosylpy- phenotypes. 10th ed. Baltimore: The Johns Hopkins
rophosphate synthetase in excessive purine produc- University Press; 1992.
tion. Eur J Clin Biol Res. 1972;17:703. 153. Kim KJ, Yamada Y, Suzumori K, Choi Y, Yang SW,
139. Balis ME, Yip LC, Yu TF, Gutman AB, Cox R, Cheong HI, Hwang YS, Goto H, Ogasawara N.
Dancis J. Unstable HPRTase in subjects with abnor- Molecular analysis of hypoxanthine guanine phos-
mal urinary oxypurine excretion. In: Sperling O, phoribosyltransferase (HPRT) gene in five Korean
DeVries A, Wyngaarden JB, editors. Purine metabo- families with Lesch-Nyhan syndrome. J Korean Med
lism in man. New York: Plenum; 1974. p. 195. Soc. 1997;12:332–9.
140. Wilson JM, Stout JT, Palella TD, Davidson BL, 154. Torres Jimenez R, Mateos Anton F, Molano Mateos J,
Kelley WN, Caskey CT. A molecular survey of Garcia Puig J. The genetic diagnosis of hypoxan-
hypoxanthine-guanine phosphoribosyltransferase thine-guanine phosphoribosyltransferase (HGPRT)
deficiency in man. J Clin Invest. 1986;77:188. deficiency. A study of 12 cases. Med Clin (Barc).
141. Davidson BL, Tarle SA, van Antwerp M, Gibbs DA, 1997;108:344–8.
Watts RWE, Kelley WN, Palella TD. Identification 155. Maruta K, Ohi T, Yamada Y, Goto H, Ogasawara N,
of 17 independent mutations responsible for human Matsukura S. A Japanese family with Lesch-Nyhan
hypoxanthine-guanine phosphoribosyltransferase syndrome resulting from a new point mutation in
(HPRT) deficiency. Am J Hum Genet. 1991;48:951. hypoxanthine-guanine phosphoribosyltransferase
142. Gordon RB, Dawson PA, Sculley DG, Emmerson BT, gene. No To Shinkei. 1997;49:1009–13.
Caskey CT, Gibbs RA. The molecular characteriza- 156. Hidalgo-Laos RI, Kedar A, Williams CA, Neiberger
tion of HPRTCHERMSIDE and HPRTCOORPAROO: RE. A new point mutation in a hypoxanthine
two Lesch-Nyhan patients with reduced amounts of phosphoribosyltransferase-deficient patient. Pediatr
mRNA. Gene. 1991;108:299. Nephrol. 1997;11:645–8.
143. Gibbs RA, Nguyen PN, Edwards A, Civitello AB, 157. Hikita M, Hosoya T, Ichida K, Okabe H, Saji M,
Caskey CT. Multiplex DNA deletion detection and Ohno I, Kuriyama S, Tomonari H, Hayashi F,
exon sequencing of the hypoxanthine-guanine phos- Fujimori S, Yamaoka N, Sakuma R. Partial deficiency
phoribosyltransferase gene in Lesch-Nyhan families. of hypoxanthine-guanine phosphoribosyltransferase
Genomics. 1990;7:235. manifesting as acute renal damage. Intern Med.
144. Ogasawara N, Stout JT, Goto H, Sonta SI, Matsumoto 1998;37:945–9.
A, Caskey CT. Molecular analysis of a female Lesch- 158. Tvrdik T, Marcus S, Hou SM, Falt S, Noori P,
Nyhan patient. J Clin Invest. 1989;84:1024. Podlutskaja N, Hanefeld F, Stromme P, Lambert B.
145. Yamada Y, Goto H, Ogasawara N. Identification of Molecular characterization of two deletion events
two independent Japanese mutant HPRT genes using involving Alu-sequences, one novel base substitu-
the PCR technique. Adv Exp Med Biol. 1991; tion and two tentative hotspot mutations in hypoxan-
309B:121. thine phosphoribosyltransferase (HPRT) gene in five
146. Stout JT, Caskey CT. HPRT: gene structure, expres- patients with Lesch-Nyhan syndrome. Hum Genet.
sion, and mutation. Annu Rev Genet. 1985;19:127. 1998;103:311–8.
References 61

159. Kamatani N. Partial hypoxanthine phosphoribosyl- hypoxanthine-guanine phosphoribosyltransferase.


transferase deficiency: unrecognized until adult ages. J Lab Clin Med. 1971;77:33.
Intern Med. 1998;37:905–6. 173. Sweetman L, Nyhan WL. Further studies of the
160. Liu G, Aral B, Zabot MT, Kamoun P, Ceballos-Picot enzyme composition of mutant cells in X-linked uric
I. The molecular basis of hypoxanthine-guanine aciduria. Arch Intern Med. 1974;130:214.
phosphoribosyltransferase deficiency in French fam- 174. Gordon RB, Thompson L, Emmerson BT. Erythrocyte
ilies; report of two novel mutations. Hum Mutat. phosphoribosylpyrophosphate concentrations in
1998;Suppl 1:S88–90. heterozygotes for hypoxanthine-guanine phosphori-
161. Khattak FH, Morris IM, Harris K. Kelley-Seegmiller bosyltransferase deficiency. Metabolism. 1974;23:
syndrome: a case report and review of the literature. 921.
Br J Rheumatol. 1998;37:580–1. 175. Yip LC, Dancis J, Methieson B, Balis ME. Age-
162. Puig JG, Mateos FA, Torres RJ, Buno AS. Purine induced changes in adenosine monophosphate:
metabolism in female heterozygotes for hypoxan- pyrophosphate phosphoribosyltransferase from nor-
thine-guanine phosphoribosyltransferase deficiency. mal and Lesch-Nyhan erythrocytes. Biochemistry.
Eur J Clin Invest. 1998;28:950–7. 1974;13:2558.
163. Chang SJ, Chang JG, Chen CJ, Wang JC, Ou TT, 176. Fox IH, Kelley WN. Phosphoribosylpyrophosphate
Chang KL, Ko YC. Identification of a new single in man: biochemical and clinical significance. Ann
nucleotide substitution on the hypoxanthine-guanine Intern Med. 1971;74:424.
phosphoribosyltransferase gene (HPRT(Tsou)) from 177. Greene ML, Boyle JA, Seegmiller JE. Substrate sta-
a Taiwanese aboriginal family with severe gout. bilization: genetically controlled reciprocal relation-
J Rheumatol. 1999;26:1802–7. ship of 2 human enzymes. Science. 1970;167:337.
164. O’Neill P, Trombley L, Gundel M, Hunter T, 178. Nuki G, Lever J, Seegmiller JE. Biochemical charac-
Nicklas JA, Ferreira ML, Bugallo MJ, Farias AC, teristics of 8-azaguanine resistant human lympho-
Lohr A, Diamantopoulos M, Raskin S. Identification blast mutants selected in vitro. In: Sperling O,
of a new Lesch-Nyhan syndrome mutation DeVries A, Wyngaarden JB, editors. Purine metabo-
(HPRTBrasil) and analysis of potentially heterozy- lism in man. New York: Plenum; 1975. p. 255.
gous females. Arq Neuropsiquiatr. 1999;57:907–11. 179. Wood AW, Becker MA, Minna JD, Seegmiller JE.
165. Willers I, Bolz H, Wehnert M, Gal A. Eighteen novel Purine metabolism in normal and thioguanine-resis-
mutations in patients with Lesch-Nyhan syndrome tant neuroblastoma. Proc Natl Acad Sci USA.
or partial hypoxanthine phosphoribosyltransferase 1973;70:3880.
deficiency. J Inherit Metab. 1999;22:845–6. 180. Pehlke DM, McDonald JA, Holmes EW, Kelley WN.
166. De Gregorio L, Nyhan WL, Serafin E, Chamoles Inosinic acid dehydrogenase activity in the Lesch-
NA. An unexpected affected female patient in a clas- Nyhan syndrome. J Clin Invest. 1972;51:1398.
sical Lesch-Nyhan family. Mol Genet Metab. 181. Lommen EJP, De Abreu RA, Trijbels JMF, Edam A.
2000;69:263–8. The IMP dehydrogenase catalysed reaction in eryth-
167. Torres RJ, Mateos FA, Molano J, Gathoff BS, rocytes of normal individuals and patients with
O’Neill JP, Gundel RM, Trombley L, Puig JG. hypoxanthine-guanine phosphoribosyltransferase
Molecular basis of hypoxanthine-guanine phospho- deficiency. Acta Paediatr Scand. 1974;63:140.
ribosyltransferase deficiency in thirteen Spanish 182. Micheli V, Sestini S, Rocchigiani M, Jacomelli G,
families. Hum Mutat. 2000;15:383. Manzoni F, Peruzzi L, Gathof BS, Zammarchi E,
168. Zoref-Shani E, Feinstein S, Frishberg Y, Bromberg Y, Pompucci G. Hypoxanthine-guanine phosphoribo-
Sperling O. Kelley-Seegmiller syndrome due to a syltransferase deficiency and erythrocyte synthesis
unique variant of hypoxanthine-guanine phosphori- of pyridine coenzymes. Life Sci. 1999;64:2479–87.
bosyltransferase: reduced affinity for 5-phosphori- 183. Skaper SD, Seegmiller JE. Hypoxanthine-guanine
bosyl-1-pyrophosphate manifested only at low, phosphoribosyltransferase mutant glioma cells:
physiological substrate concentrations. Biochim diminished monoamine oxidase activity. Science.
Biophys Acta. 2000;500:197–203. 1976;194:1171.
169. Lesch M, Nyhan WL. A familial disorder of uric 184. Roth JA, Breakefield XO, Castiglione CM. Monoamine
acid metabolism and central nervous system func- oxidase and catechol-o-methyltransferase in cultured
tion. Am J Med. 1964;36:561. human skin fibroblasts. Life Sci. 1976;19:1705.
170. Rubin CS, Balis ME, Piomelli S, Berman PH, 185. Breakefield XO, Castiglione CM, Edelstein SB.
Dancis J. Elevated AMP pyrophosphorylase activity Monoamine oxidase activity decreased in cells lack-
in congenital IMP pyrophosphorylase deficiency ing hypoxanthine phosphoribosyltransferase activ-
(Lesch-Nyhan disease). J Lab Clin Med. 1969;74: ity. Science. 1976;192:1018.
732. 186. Edelstein SB, Castiglione CM, Breakefield XO.
171. Kelley WN. Hypoxanthine-guanine phosphoribosyl- Monoamine oxidase activity in normal and Lesch-
transferase deficiency in the Lesch-Nyhan syndrome Nyhan fibroblasts. J Neurochem. 1978;31:1247.
and gout. Fed Proc. 1968;27:1047. 187. Singh S, Willer I, Klus EM, Goedde HW. Monoamine
172. Kelley WN. Studies on the adenine phosphoribosyl- oxidase and catechol-o-methyltransferase activity in
transferase enzyme in human fibroblasts lacking cultured fibroblasts from patients with maple syrup
62 3 Purine Biochemistry

urine disease, Lesch-Nyhan syndrome and healthy 202. Hors-Cayla MC, Heuertz S, Frezal H. Coreactivation
controls. Clin Genet. 1979;15:153. of four inactive X genes in a hamster x human hybrid
188. Kochersperger LM, Parker EL, Siciliano M, and persistence of late replication of reactivated X
Darlington GJ, Denney IM. Assignment of genes for chromosome. Somatic Cell Genet. 1983;9:645.
the human monoamine oxidase A and B to the X 203. Keith DH, Singer-Sam J, Riggs AD. Active X chro-
chromosome. J Neurosci Res. 1986;16:601. mosome DNA is unmethylated at eight CCGG sites
189. Zoref-Shani E, Sperling O. Dependence of the meta- clustered in a guanine-plus-cytosine-rich island at
bolic fate of IMP on the rate of total IMP synthesis. the 5¢ end of the gene for phosphoglycerate kinase.
Studies in cultured fibroblasts from normal subjects Mol Cell Biol. 1986;6:4122.
and from purine-overproducing mutant patients. 204. Wolf SF, Dintzis T, Toniolo D, Persico G, Lunnen KD,
Biochim Biophys Acta. 1980;607:503. Axelman J, Migeon BR. Complete concordance
190. Puig JG, Mateos FA. Clinical and biochemical between glucose-6-phosphate dehydrogenase activ-
aspects of uric acid overproduction. Pharm World ity and hypomethylation of 3¢ CpG clusters:
Sci. 1994;16:40. Implications for X chromosome dosage compensa-
191. Locke LF, Takagi N, Martin GR. Methylation of the tion. Nucleic Acids Res. 1984;12:9333.
HPRT gene on the inactive X occurs after chromo- 205. Wolf SF, Jolly DJ, Lunnen KD, Friedman T,
some inactivation. Cell. 1987;48:39. Migeon BR. Methylation of the hypoxanthine phos-
192. Yen PH, Patel PI, Chinault AL, Mohandas T, phoribosyltransferase locus on the human X chromo-
Shapiro LJ. Differential methylation of hypoxan- some: implications for X-chromosome inactivation.
thine phosphoribosyltransferase genes on active and Proc Natl Acad Sci USA. 1984;81:2806.
inactive human X-chromosomes. Proc Natl Acad Sci 206. Wolf SF, Migeon BR. Clusters of CpG dinucleotides
USA. 1984;81:1759. implicated by nuclease hypersensitivity as control
193. Locke LF, Melton DW, Caskey CT, Martin GR. elements of housekeeping genes. Nature. 1985;314:
Methylation of the mouse HPRT gene differs on the 467.
active and inactive X chromosomes. Mol Cell Biol. 207. Migeon BR, Holland MM, Driscoll DJ, Robinson JC.
1986;6:914. Programmed demethylation in CpG islands during
194. Lester SC, Korn NJ, DeMars R. Derepression of human fetal development. Somat Cell Mol Genet.
genes on the human inactive X chromosome: evi- 1991;17:159.
dence for differences in locus-specific rates of dere- 208. Lock LF, Takagi N, Martin GR. Methylation of the
pression and rates of transfer of active and inactive Hprt gene on the inactive X occurs after chromo-
genes after DNA-mediated transformation. Somatic some inactivation. Cell. 1987;48:39.
Cell Genet. 1982;8:265. 209. Marcus S, Steen AM, Andersson B, Lambert B,
195. Chapman VM, Kratzer PG, Siracusa LD, Kristoffersson U, Francke U. Mutation analysis and
Quarantillo BA, Evans R, Liskay RM. Evidence for prenatal diagnosis in a Lesch-Nyhan family showing
DNA modification in the maintenance of non-random X inactivation interfering with carrier
X-chromosome inactivation of adult mouse tissues. detection. Hum Genet. 1992;89:395.
Proc Natl Acad Sci USA. 1982;79:5357. 210. Hornstra IK, Yang TP. Multiple in vivo footprints are
196. Liskay RM, Evans RJ. Inactive X chromosome DNA specific to the active allele of the X-linked human
does not function in DNA-mediated cell transforma- hypoxanthine phosphoribosyltransferase gene 5¢
tion for the hypoxanthine phosphoribosyltransferase region: implications for X chromosome inactivation.
gene. Proc Natl Acad Sci USA. 1980;77:4895. Mol Cell Biol. 1992;12:5345–54.
197. Graves JAM. 5-Azacytidine-induced re-expression 211. Hornstra IK, Yang TP. High-resolution methylation
of alleles on the inactive X chromosome in a hybrid analysis of the human hypoxanthine phosphoribo-
mouse cell line. Exp Cell Res. 1982;141:99. syltransferase gene 5¢ region on the active and inac-
198. Venolia L, Gartler SM. Comparison of transforma- tive X chromosomes: correlation with binding sites
tion efficiency of human active and inactive for transcription factors. Mol Cell Biol. 1994;14:
X-chromosomal DNA. Nature. 1983;302:82. 1419–30.
199. Mohandas T, Sparkes RS, Shapiro LJ. Reactivation 212. Park JG, Chapman VM. CpG island promoter region
of an inactive human X chromosome: evidence for X methylation patterns of the inactive-X-chromosome
inactivation by DNA methylation. Science. 1981;211: hypoxanthine phosphoribosyltransferase (Hprt)
393. gene. Mol Cell Biol. 1994;14:7975–83.
200. Paterno GD, Adra CN, McBurney MW. X chromo- 213. Subramanian PS, Chinault AC. Replication timing
some reactivation in mouse embryonal carcinoma properties of the human HPRT locus on active, inac-
cells. Mol Cell Biol. 1985;5:2705. tive and reactivated X chromosomes. Somat Cell
201. Toniolo D, D’Urso M, Martini G, Persico M, Mol Genet. 1997;23:97–109.
Tufano V, Battistuzzi G, Luzzatto L. Specific 214. Disteche CM. Escape from X inactivation in human
methylation pattern at the 3¢ end of the human house- and mouse. Trends Genet. 1995;11:17–22.
keeping gene for glucose 6-phosphate dehydroge- 215. Disteche CM. Escapees on the X chromosome. Proc
nase. EMBO J. 1984;3:1987. Natl Acad Sci USA. 1999;96:14180–2.
References 63

216. Carrel L, Cottle AA, Goglin KC, Willard HF. A first- chromosome 16 by mouse-human somatic cell
generation X-inactivation profile of the human X chro- hybridization. Proc Natl Acad Sci USA. 1974;
mosome. Proc Natl Acad Sci USA. 1999;96:14440–4. 71:45.
217. Moro F, Ogg CS, Simmonds HA, Cameron JS, 234. Fratini A, Simmers RN, Callen DF, Hyland VJ,
Chantler C, McBride MB, Duley JA, Davies PM. Tischfield JA, Stambrook PJ, Sutherland GR. A new
Familial juvenile gouty nephropathy with renal urate location for the human adenine phosphoribosyltrans-
hypoexcretion preceding renal disease. Clin Nephrol. ferase gene (APRT) distal to the haptoglobin (HP)
1991;35:263. and fra (16)(q23)(FRA16D) loci. Cytogenet Cell
218. Simmonds HA. 2,8-dihyroxyadenine lithiasis – epi- Genet. 1986;43:10.
demiology, pathogenesis and therapy. In: 235. Van Acker KJ, Simmonds HA, Potter CF, Sahota A.
Verhandlungen der Deutschen Gesellschaft fur Inheritance of adenine phosphoribosyltransferase
Innere Medizin, 92 band. Munchen: JF Bergman (APRT) deficiency. Adv Exp Med Biol. 1980;122A:
Verlag; 1986. p. 503. 349.
219. Kelley WN, Levy RI, Rosenbloom FM, Henderson JF, 236. Murray AM, Drobetsky E, Arrand JE. Cloning the
Seegmiller JE. Adenine phosphoribosyltransferase complete human adenine phosphoribosyltransferase
deficiency – a previously undescribed genetic defect gene. Gene. 1984;31:233.
in man. J Clin Invest. 1968;47:2281. 237. Stambrook PJ, Dush MK, Trill JJ, Tischfield JA.
220. Delbarre F, Auscher C, Amor B, DeGery A, Cartier P, Cloning of a functional human adenine phosphori-
Hamlet M. Gout with adenine phosphoribosyltrans- bosyltransferase (APRT) gene: Identification of a
ferase deficiency. Biomedicine. 1974;21:82. restriction fragment length polymorphism and pre-
221. Henderson JF, Miller HR, Kelley WN, liminary analysis of DNAs from APRT-deficient
Rosenbloom FM, Seegmiller JE. Kinetic studies of families and cell mutants. Somat Cell Mol Genet.
mutant human adenine phosphoribosyltransferase. 1984;10:359.
Can J Biochem. 1968;46:703. 238. Tischfield JA, Trill JJ, Dush MK, Stambrook PJ.
222. Dean BM, Watts RWE, Westwick WJ. Human eryth- Polymorphism within the human adenine transferase
rocyte AMP pyrophosphate phosphoribosyltrans- (APRT) locus. Genetics. 1984;107:s107.
ferase (EC 2.4.2.7). FEBS Lett. 1968;1:179. 239. Ogasawara N, Goto H. Restriction fragment length
223. Fox IH, Meade JC, Kelley WN. Adenine phosphori- polymorphisms of HPRT and APRT genes in
bosyltransferase deficiency in man. Report of a sec- Japanese population. Adv Exp Med Biol. 1989;253A:
ond family. Am J Med. 1973;55:614. 461.
224. Srivastava SK, Beutler E. Purification and kinetic stud- 240. Arrand JE, Murray AM, Spurr N. Sph I restriction
ies of adenine phosphoribosyltransferase from human fragment length polymorphism on human chromo-
erythrocytes. Arch Biochem Biophys. 1971;142:426. some 16 detected with an APRT gene probe. Nucleic
225. Hori M, Henderson JF. Kinetic studies of adenine phos- Acids Res. 1987;15:9615.
phoribosyltransferase. J Biol Chem. 1966;241:3404. 241. Kamatani N, Kuroshima S, Hakoda M, Palella TD,
226. Krenitsky TA, Neil SM, Elion GB, Hitchings GH. Hidaka Y. Crossovers within a short DNA sequence
Adenine phosphoribosyltransferase from monkey indicate a long evolutionary history of the APRT*J
liver. J Biol Chem. 1969;244:4779. mutation. Hum Genet. 1990;85:600.
227. Taylor MW, Sahota A. Cellular resistance to adenine 242. Broderick TP, Schaff DA, Bertino AM, Dush MK,
analogues. In: Gupta RS, editor. Drug resistance in Tischfield JA, Stambrook PJ. Comparative anatomy
mammalian cells, Antimetabolites and cytotoxic of the human APRT gene and enzyme. Nucleotide
analogs, vol. 1. Boca Raton: CRC; 1989. p. 111. sequence divergence and conservation of a nonran-
228. Koyama H, Kodama H. Adenine phosphoribosyl- dom CpG dinucleotide arrangement. Proc Natl Acad
transferase deficiency in cultured mouse mammary Sci USA. 1987;84:3349.
tumor FM3A cells resistant to 4-carbamoylimidazo- 243. Briggs MR, Kadonaga JT, Bell SP, Tijian R.
lium-5-olate. Cancer Res. 1982;42:4210. Purification and biochemical characterization of the
229. Arnold WJ. Purine salvage enzymes. In: Kelley WN, promoter specific transcription factor, Spl. Science.
Weiner IM, editors. Uric acid. Berlin: Springer; 1986;234:47.
1978. p. 61. 244. Keshet I, Yisraeli J, Cedar H. Effect of regional DNA
230. Wilson JM, O’Toole TE, Argos P, Shewach DS, methylation on gene expression. Proc Natl Acad Sci
Daddona PE, Kelley WN. Human adenine phosphori- USA. 1985;82:2560.
bosyltransferase: complete amino acid sequence of the 245. Frank D, Keshet I, Shani M, Levine A, Razin A,
erythrocyte enzyme. J Biol Chem. 1986;261:13677. Cedar H. Demethylation of CpG islands in embry-
231. Kamatani N. Adenine phosphoribosyltransferase onic cells. Nature. 1991;351:239.
(APRT). Nippon Rinsho. 1996;54:3213–9. 246. Park J-H, Hershey HV, Taylor MW. Housekeeping
232. Kahan B, Held KR, DeMars R. The locus for human genes. In: Simonsen CC, Shepard M, editors.
adenine phosphoribosyltransferase on chromosome Molecular genetics of mammalian cells. New York:
No. 16. Genetics. 1974;78:1143. Macmillan; 1986. p. 79.
233. Tischfield JA, Ruddle FH. Assignment of the gene 247. Simmonds HA, Van Acker KJ, Cameron JS,
for adenine phosphoribosyltransferase to human Snedden W. The identification of 2,8-dihydroxyade-
64 3 Purine Biochemistry

nine, a new component of urinary stones. Biochem J. immunologic and phagocytic investigations. Pediatr
1976;157:485. Res. 1984;18:30.
248. Debray H, Cartier P, Temstet A, Cendron J. Child’s 262. Szonyi P, Bereni M, Toth J. A rare enzyme deficiency
urinary lithiasis revealing a complete deficit in ade- causing formation of 2,8-dihydroxyadenine (purine
nine phosphoribosyltransferase. Pediatr Res. body) calculi. Int Urol Nephrol. 1985;17:231.
1976;10:762. 263. Simmonds HA. 2,8-dihydroxyadenine lithiasis. Clin
249. Van Acker KJ, Simmonds HA, Potter CF, Chim Acta. 1986;160:103.
Cameron JS. Complete deficiency of adenine phos- 264. Manyak MJ, Frensilli FJ, Miller HC. 2,8-dihydroxy-
phoribosyltransferase: report of a family. N Engl J adenine urolithiasis: report of an adult case in the
Med. 1977;297:127. United States. J Urol. 1987;137:312.
250. Barratt TM, Simmonds HA, Cameron JS, Potter CF, 265. Gliklich D, Gruber HE, Matas AJ, Tellis VA, Karwa G,
Rose GA, Arkell DG, Williams DI. Complete Finley K, Salem C, Soberman R, Seegmiller JE.
deficiency of adenine phosphoribosyltransferase. A 2,8-dihydroxyadenine lithiasis: report a case first diag-
third case presenting as renal stones in a young child. nosed after renal transplant. Q J Med. 1988;69:785.
Arch Dis Child. 1979;54:25. 266. Laxdal T, Jonason TA. Adenine phosphoribosyl-
251. Takemoto M, Nagano S. Urolithiasis containing transferase deficiency in Iceland. Acta Med Scand.
2,8-dihydroxyadenine: report of a case. Acta Urol 1988;224:621.
Jpn. 1979;25:265. 267. Chiba P, Zwiauer K, Muller MM. Characterization
252. Osada T, Inoue T, Hirano A, Tanaka K, Ogita Z, Isobe of an adenine phosphoribosyltransferase deficiency.
M, Hayashi S. A case of 2,8-dihydroxyadenine lithia- Clin Chim Acta. 1988;172:141.
sis revealing a complete deficiency of adenine phos- 268. Usenius JP, Ruapuro ML, Usenius R. Adenine phos-
phoribosyltransferase. J Clin Urol Jpn. 1980;71:981. phoribosyltransferase deficiency: 2,8-dihydroxyade-
253. Cartier P, Hamet M, Vincens A, Perignon JL. nine urolithiasis in a 48-year-old woman. Br J Urol.
Complete adenine phosphoribosyltransferase (APRT) 1988;62:521.
deficiency in two siblings: report of a new case. Adv 269. Jung P, Becht E, Ziegler M, Bommert R, Bach K,
Exp Med Biol. 1980;122A:343. Haas HJ. New diagnostic and therapeutic aspects of
254. Schabel F, Doppler W, Hirsch-Kauffmann M, 2,8-dihydroxyadenine lithiasis. Another case of
Glatzl J, Schweiger M, Berger H, Heinz-Erian P. complete adenine phosphoribosyltransferase
Hereditary deficiency of adenine phosphoribosyl- deficiency. Eur Urol. 1988;14:493.
transferase. Paediatr Padol. 1980;15:233. 270. Hesse A, Miersch WD, Classen A, Thon A, Doppler
255. Gault MH, Simmonds HA, Snedden W, Dow D, W. 2,8-dihydroxyadeninuria: laboratory diagnosis
Churchill DN, Penney H. Urolithiasis due to 2,8-di- and therapy control. Urol Int. 1988;43:174.
hydroxyadenine in an adult. N Engl J Med. 1981;305: 271. Lamontagne AE. 2,8-dihydroxyadenine urolithiasis:
1570. report of a woman in the United States. J Urol.
256. Asper R, Schmucki O. Diagnostik und Therapie der 1989;142:369.
2,8-Dihydroxyadenine Lithiasis. In: Gasser G, 272. Gleeson MJ, Griffith DP. Distribution of patients
Vahlensieck W, editors. Pathogenese und Klinik der with 2,8-dihydroxyadenine urolithiasis and adenine
Harnsteine IX. Darmstadt: Steinkopff Verlag; 1982. phosphoribosyltransferase deficiency in Japan.
p. 274. J Urol. 1989;142:834.
257. Joost J, Doppler W. The 2,8-dihydroxyadenine stone 273. Honeke K, Butz M. 2,8-dihydroxyadenin harnstein:
in childhood. Urology. 1982;20:67. die bedeuting der exakten physikalischen steinanal-
258. Witten FR, Morgan JE, Foster JG, Glenn JF. yse. Urologe. 1989;28:361.
2,8-dihydroxyadenine urolithiasis: review of the lit- 274. Coupris L, Champion G, Duverne C, Varlet F,
erature and report of a case in the United States. Rataajczak A. La lithiase 2,8-dihydroxyadenique. 2
J Urol. 1983;130:938. nouvelles observations p’ediatriques d’un de’ficit
259. Simmonds HA, Van Acker KJ. Adenine phosphori- me’tabolique me’conna. Apport de la lithotripsie
bosyltransferase deficiency: 2,8-dihydroxyadenine extra-corporelle. Chir Pediatr. 1990;31:26.
lithiasis. In: Stanbury JB, Wyngaarden JB, 275. Leusmann DB, Schmidt G. Urolithiasis bei 2,8-dihy-
Fredrickson DS, Goldstein JL, Brown MS, editors. droxyadenurie: vorstelling von drei wieteren Fallen.
The metabolic basis of inherited disease. 5th ed. Z Urol Nephrol. 1990;83:383.
New York: McGraw-Hill; 1983. p. 1144. 276. Frick J, Sarica K, Kohle RF, Kunit G. Long-term
260. Chevet D, Le Pogamp P, Gie S, Gary J, Daudon M, follow-up after extracorporeal shock wave litho-
Hamet M. 2,8-dihydroxyadenine (2,8-DHA) uro- tripsy in children. Eur Urol. 1991;19:225.
lithiasis in an adult – complete adenine phosphoribo- 277. Simmonds HA, Sahota AS, Van Acker KJ.
syltransferase deficiency – family study. Kidney Int. Adenine phosphoribosyltransferase deficiency and
1984;26:226. 2,8-dihydroxyadenine lithiasis. In: Scriver CR,
261. Kishi T, Kidani K, Komazawa Y, Sakura N, Beaudet AL, Sly WS, Valle D, editors. The meta-
Matsuura R, Kobayashi M, Tanabe A, Hyodo S, bolic and molecular bases of inherited disease, vol.
Kittaka E, Sakano T. Complete deficiency of adenine II. New York: McGraw-Hill, Inc.; 1995. p. 1707.
phosphoribosyltransferase: report of three cases and
References 65

278. Katamatani N, Terai C, Kim CY, Chen CL, 290. Greenwood MC, Dillon MJ, Simmonds HA,
Yamanaka H, Hakoda M, Totokawa S, Kashiwazaki Barratt TM, Pincott JR, Metrewelli C. Renal failure
S. The origin of the most common mutation of ade- due to 2,8-dihydroxyadenine urolithiasis. Eur J
nine phosphoribosyltransferase among Japanese Pediatr. 1982;138:346.
goes back to a prehistoric era. Hum Genet. 1996; 291. Emmerson BT, Gordon RB, Thompson L. Adenine
98:596–600. phosphoribosyltransferase deficiency: its inheritance
279. Kuroda M, Miki T, Kiyohara H, Usami M, and occurrence in a female with gout and renal dis-
Nakamura T, Kotake T, Takemoto M, Sonoda T. ease. Aust N Z J Med. 1975;5:440.
Urolithiasis composed of 2,8-dihydroxyadenine due 292. O’Toole TE, Wilson JM, Gault MH, Kelley WN.
to partial deficiency of adenine phosphoribosyltrans- Human adenine phosphoribosyltransferase: charac-
ferase. Jpn J Urol. 1980;71:283. terization from subjects with a deficiency of enzyme
280. Takeuchi F, Matsuta K, Miyamoto T, Enomoto S, activity. Biochem Genet. 1983;21:1121.
Fujimori S, Akaoka I, Kamatani N, Nishioka K. Rapid 293. Steglich C, DeMars R. Mutations causing deficiency
method for the diagnosis of partial adenine phosphori- of APRT in fibroblasts cultured from humans
bosyltransferase deficiency causing 2,8-dihydroxy- heterozygous for mutant APRT alleles. Somatic Cell
adenine urolithiasis. Hum Genet. 1985;71:167. Genet. 1982;8:115.
281. Kamatani N, Takeuchi F, Nishida Y, Yamanaka H, 294. Fujimori S, Akaoka I, Takeuchi F, Kanayama H,
Nishioka K, Tatara K, Fujimori S, Kaneko K, Akaoka Tatara K, Nishioka K, Kamatani N. Altered kinetic
I, Tofuku Y. Severe impairment in adenine metabo- properties of a mutant adenine phosphoribosyltrans-
lism with a partial deficiency of adenine phosphori- ferase. Metabolism. 1986;35:187.
bosyltransferase. Metabolism. 1985;34:164. 295. Abe S, Hayasaka K, Narisawa K, Tada K, Okada G,
282. Fujimoro S, Akaoka I, Sakamoto K, Yamanaka H, Koyama H, Kurosa S, Kudoh M, Matushita K. Partial
Nishioka K, Kamatani N. Common characteristics and complete adenine phosphoribosyltransferase
of mutant adenine phosphoribosyltransferase from deficiency associated with 2,8-dihydroxyadenine
four separate Japanese families with 2,8-dihydroxy- urolithiasis: kinetic and immunochemical properties
adenine urolithiasis associated with partial enzyme of APRT. Enzyme. 1987;37:182.
deficiencies. Hum Genet. 1985;7:71. 296. Kamatani N, Kuroshima S, Terai C, Hidaka Y,
283. Kamatani N, Terai C, Kuroshima S, Nishioka K, Palella TD, Nishioka K. Detection of an amino acid
Mikanagi K. Genetic and clinical studies on 19 fami- substitution in the mutant enzyme for a special type
lies with adenine phosphoribosyltransferase of adenine phosphoribosyltransferase (APRT)
deficiencies. Hum Genet. 1987;75:163. deficiency by sequence-specific protein cleavage.
284. Kamatani N, Kuroshima S, Terai C, Kawai K, Am J Hum Genet. 1989;45:325.
Mikanagi K, Nishioka K. Selection of human cells 297. Fox IH, LaCroix S, Planet G, Moore M. Partial
having two different types of mutations in single deficiency of adenine phosphoribosyltransferase in
cells: application to the diagnosis of the heterozy- man. Medicine. 1977;56:515.
gous state for a unique type adenine phosphoribosyl- 298. Simmonds HA, Duley JA, Davies PM. Analysis of
transferase deficiency. Hum Genet. 1987;76:148. purine and pyrimidines in blood, urine, and other
285. Kamatani N, Sonoda T, Nishioka K. Distribution of physiological fluids. In: Hommes F, editor. Techniques
patients with 2,8-dihydroxyadenine urolithiasis and in diagnostic human biochemical genetics. A labora-
adenine phosphoribosyltransferase deficiency in tory manual. New York: Wiley-Liss; 1991. p. 397.
Japan. J Urol. 1988;140:1470. 299. Hakoda M, Yamanaka H, Kamatani N, Kamatani N.
286. Kamatani N, Kuroshima S, Yamanaka H, Nakashe S, Diagnosis of heterozygote states for adenine phos-
Take H, Hakoda M. Identification of a compound phoribosyltransferase deficiency based on detection
heterozygote for adenine phosphoribosyltransferase of in vivo somatic mutants in blood T cells: applica-
deficiency (APRT*J/APRT*Q0) leading to 2,8-dihy- tion to screening of heterozygotes. Am J Hum Genet.
droxyadenine urolithiasis. Hum Genet. 1990;85:500. 1991;48:552.
287. Sahota A, Chen J, Behzadian MA, Ravindra R, 300. Dean BM, Perrett D, Simmonds HA, Sahota A, Van
Takeuchi H, Stambrook PJ, Tischfield JA. 2,8-dihy- Acker KJ. Adenine and adenosine metabolism in intact
droxyadenine lithiasis in a Japanese patient heterozy- erythrocytes deficient in adenosine monophosphate-
gous at the adenine phosphoribosyltransferase locus. pyrophosphate phosphoribosyltransferase: a study of
Am J Hum Genet. 1991;48:983. two families. Clin Sci Mol Med. 1978;55:407.
288. Kamatani N, Hakoda M, Otsuka S, Yoshikawa H, 301. Doppler W, Hirsch-Kauffmann M, Schabel F,
Kashiqazaki S. Only three mutations account for Schweiger M. Characterization of the biochemical
almost all defective alleles causing adenine phos- basis of a complete deficiency of the adenine phos-
phoribosyltransferase deficiency in Japanese phoribosyltransferase (APRT). Hum Genet. 1981;57:
patients. J Clin Invest. 1992;90:130. 404.
289. Sevcik J, Adam T, Mazacova H. A fast and simple 302. Kojima T, Nishina T, Kitamura M, Kamatani N,
screening method for detection of 2,8-dihydroxy- Nishioka K. Reversed-phase high-performance liq-
adenine urolithiasis by capillary zone electrophore- uid-chromatography of 2,8-dihydroxyadenine in
sis. Clin Chim Acta. 1996;245:85–92.
66 3 Purine Biochemistry

serum and urine with electrochemical detection. Clin 314. Laxdal T. 2,8-dihydroxyadenine crystalluria vs. uro-
Chim Acta. 1989;181:109. lithiasis. Lancet. 1992;340:184.
303. Johnson LA, Gordon RB, Emmerson BT. Adenine 315. Ohne T, Fujito A, Koga K, Imaide Y, Uchida M.
phosphoribosyltransferase: a simple spectrophoto- 2,8-dihydroxyadenine urolithiasis due to partial
metric assay and the incidence of mutation in the deficiency of adenine phosphoribosyltransferase: a
normal population. Biochem Genet. 1977;15:256. case report. Hinyokika Kiyo. 1998;44:725–8.
304. Stenzel A, Banholzer P, Reiter S, Grobner W, Zollner 316. Suzuki K, Kobayashi S, Kawamura K, Kuhara T,
N, Hegeman M, Pfab R. Activity of adenine phos- Tsugawa R. Family study of 2,8-dihyroxyadenine
phoribosyltransferase (APRT) in patients with renal stone formation: report of two cases of a compound
failure and urolithiasis. In: Schwille PO, Smith LH, heterozygote for adenine phosphoribosyltransferase
Robertson WG, Vahlensieck W, editors. Urolithiasis deficiency (APRT*J/APRT*Q0). Int J Urol. 1997;4:
and clinical research. New York: Plenum Press; 304–6.
1985. p. 347. 317. Wang L, Ou X, Sebasta I, Vondrak K, Krijt J,
305. Srivastava SK, Villacorte D, Beutler E. Correlation Elleder M, Poupetova H, Ledvinova J, Zeman J,
between adenylate metabolising enzymes and ade- Simmonds HA, Tischfield JA, Sahota A. Combined
nine nucleotide levels of erythrocytes during blood adenine phosphoribosyltransferase and N-acetylga-
storage in various media. Transfusion. 1972;12:190. lactosamin-6-sulfate sulfatase deficiency. Mol Genet
306. Hidaka Y, Tarle SA, Fujimori S, Kamatani N, Kelley Metab. 1999;68:78–85.
WN, Palella TD. Human adenine phosphoribosyl- 318. Baker E, Gua XH, Orsborn AM, Sutherland GR,
transferase deficiency. Demonstration of a single Callen DF, Hopwood JJ, Morris CP. The morquio A
mutant allele common to the Japanese. J Clin Invest. syndrome (mucopolysaccharidosis IVA) gene maps
1988;81:945. to 16q24.3. Am J Hum Genet. 1993;52:96–8.
307. Sahota A, Chen J, Asako K, Takeuchi H, 319. Masuno M, Tomatsu S, Nakashima Y, Hori T,
Stambrook PJ, Tischfield JA. Identification of a com- Fukuda S, Masue M, Sukegawa K, Orii T.
mon nonsense mutation in Japanese patients with Mucopolysaccharidosis IV A: assignment of the
type I adenine phosphoribosyltransferase deficiency. human N-acetylgalactosamine-6-sulfate sulfatase
Nucleic Acids Res. 1990;18:5915. (GALNS) gene to chromosome 16q24. Genomics.
308. Mimori A, Hidaka Y, Wu VC, Tarle SA, Kamatani N, 1993;16:777–8.
Kelley WN, Palella TD. A mutant allele common to 320. Fukuda S, Tomatsu S, Masuno M, Ogawa T,
type I adenine phosphoribosyltransferase deficiency Yamagishi A, Rezvi GM, Sukegawa K, Shimozawa
in Japanese subjects. Am J Hum Genet. 1991;48:102. N, Suzuki Y, Kondo N, Imaizumi K, Kuroki Y,
309. Gathof BS, Sanota A, Gresser U, Chen J, Stambrook Okabe T, Orii T. Mucopolysaccharidosis IVA: sub-
PJ, Tischfield JA, Zollner N. A splice mutation at the microscopic deletion of 16q24.3 and a novel R386C
adenine phosphoribosyltransferase locus in a German mutation of N-acetylgalactosamine-6-sulfate sul-
family. Adv Exp Med Biol. 1991;309B:83. fatase gene in a classical Morquio disease. Hum
310. Sahota A, Chen J, Stambrook PJ, Tischfield JA. Mutat. 1996;7:123–34.
Genetic basis of adenine phosphoribosyltransferase 321. Yuen M, Fensom AH. Diagnosis of classical
deficiency. In: Gresser U, editor. Molecular genetics, Morquio’s disease: N-acetylgalactosamine-6-sulfate
biochemistry and clinical aspects of purine and sulphatase activity in cultured fibroblasts, leuko-
pyrimidine metabolism. Heidelberg: Springer; 1993. cytes, amniotic cells and chorionic villi. J Inherit
p. 54. Metab Dis. 1985;8:80–6.
311. Carothers AM, Urlaub G, Mucha J, Harvey RG, 322. Ceballos-Picot I, Perignon JL, Hamet M, Daudon M,
Chasin LA, Grunberger D. Splicing mutations in the Kamoun P. 2,8-dihydroxyadenine urolithiasis, an
CHO DHFR gene preferentially induced by (+/−) underdiagnosed disease. Lancet. 1992;339:1050.
3a,4b-dihydroxy-1a, 2a-epoxy-1,2,3,4-tetrahydro- 323. Peck CC, Bailey FJ, Moore GL. Enhanced solubility
benzo[c]phenambenzene. Proc Natl Acad Sci USA. of 2,8-dihydroxyadenine (DOA) in human urine.
1990;87:5464. Transfusion. 1977;17:383.
312. Chen J, Sahota A, Martin GF, Hakoda M, Kamatani 324. Maddocks JL, Al-Safi SA. Adenine phosphoribosyl-
N, Stambrook PJ, Tischfield JA. Analysis of ger- transferase deficiency: a simple diagnostic test. Clin
mline and in vivo somatic mutations in the human Sci. 1988;75:217.
adenine phosphoribosyltransferase genes: muta- 325. Engle SJ, Stockelman MG, Chen J, Boivin G, Yum
tional hotspots at the intron 4 splice donor site and at MN, Davies PM, Ying MY, Sahota A, Simmonds
codon 87. Mutat Res. 1993;287:217. HA, Stambrook PJ, Tischfield JA. Adenine
313. Chen J, Sahota A, Laxdal T, Scrine M, Bowman S, phosphoribosyltransferase-deficient mice develop
Cui C, Stambrook PJ, Tischfield JA. Identification of 2,8-dihydroxyadenine nephrolithiasis. Proc Natl
a single missense mutation in the adenine phosphori- Acad Sci USA. 1996;93:5307–12.
bosyltransferase gene from five Icelandic patients and 326. Redhead NJ, Selfridge J, Wu CL, Melton DW. Mice
a British patient. Am J Hum Genet. 1991;49:251. with adenine phosphoribosyltransferase deficiency
References 67

develop fatal 2,8-dihydroxyadenine lithiasis. Hum 331. Cohen A, Doyle D, Martin DW, Ammann AJ.
Gene Ther. 1996;7:1491–502. Abnormal purine metabolism and purine overpro-
327. Estepa-Maurice L, Hennequin C, Marfisi C, duction in a patient deficient in purine nucleoside
Lacour B, Daudon M. Fourier transform infrared phosphorylase. N Engl J Med. 1976;295:1449.
microscopy identification of crystal deposits in tis- 332. Simmonds HA, Fairbanks LD, Morris GS, Morgan G,
sues: clinical importance in various pathologies. Am Watson AR, Timms P. Central nervous system dys-
J Clin Pathol. 1996;105:576–82. function and erythrocyte guanosine triphosphate
328. Stockelman MG, Lorenz JN, Smith FN, Boivin GP, depletion in purine nucleoside phosphorylase
Sahota A, Tischfield JA, Stambrook PJ. Chronic deficiency. Arch Dis Child. 1987;62:385.
renal failure in a mouse model of human adenine 333. Simmonds HA, Webster DR, Becroft DMO,
phosphoribosyltransferase deficiency. Am J Physiol. Potter CF. Purine and pyrimidine metabolism in
1998;275:F154–63. hereditary orotid aciduria: some unexpected effects
329. Tischfield JA. Molecular characterization of a novel of allopurinol. Eur J Clin Invest. 1980;10:33.
mutation in APRT heterozygotes. In: Sahota A, 334. Wilson JM, Daddona PE, Otaadese T, Kelley WN.
Taylor M, editors. Purine and pyrimidine metabo- Adenine phosphoribosyltransferase in patients with
lism in man VIII. New York: Plenum Press; 1995. disorders of purine and pyrimidine metabolism.
p. 675. J Lab Clin Med. 1982;99:163.
330. Sahota A, Bye S, Chen J, Khattar NH, Turker MS, 335. Edwards NL, Recker D, Airozo D, Fox IH. Enhanced
Moro F, Simmonds HA, Emmerson BT, Gordon RB, purine salvage during allopurinol therapy: an impor-
Tischfield JA. Molecular characterization of a novel tant pharmacologic property in humans. J Lab Clin
mutation in APRT heterozygotes. In: Sahota A, Med. 1981;98:673.
Taylor M, editors. Purine and pyrimidine metabo- 336. Edwards NL, Recker D, Fox IH. Overproduction of
lism in man VIII. New York: Plenum Press; 1995. uric acid in hypoxanthine-guanine phosphoribosyl-
p. 675. transferase deficiency. J Clin Invest. 1979;63:922.
Uric Acid Metabolism in Humans
4

Overview extrarenal routes such as the gastrointestinal tract


could exist. In the human, only three of these
The metabolic alterations leading to an elevated mechanisms are clearly associated with gout: the
serum uric acid level are the precursors of the increased production of uric acid via de novo bio-
clinical expression of hyperuricemia and gout, synthesis, the increased production of uric acid as
and therefore, the key to understanding the patho- a result of the breakdown of preformed tissue
genesis and management of gout lies with an nucleic acids, and the decreased elimination of
understanding of the mechanisms that can cause uric acid via the kidney.
these changes. Even though gout is a single clini-
cal entity, it is caused by a variety of different
mechanisms that may merit different treatment Purine Biosynthesis
regimens. The contributors to the pool of uric
acid in the human include preformed purines in The ultimate rate of purine biosynthesis is the
the diet that may be catabolized to uric acid, the result of complex interdependent relationships
de novo biosynthesis of uric acid from small mol- between de novo biosynthesis, dietary purine
ecules, and the turnover and breakdown of tissue intake, purine degradation, and purine excretion.
nucleic acids to uric acid. There is constant Although attempts have been made to measure
turnover of the uric acid pool since uric acid is the rate of purine synthesis in humans on purine-
eliminated via the kidney, gastrointestinal tract, restricted diets, these studies have produced vari-
and other minor routes such as tears, perspira- able rates without providing a single standardized
tion, and saliva. A consideration of the routes of rate for normal, healthy individuals. For this rea-
urate generation and elimination provides a son, assessments of the rates of purine synthesis
framework for postulating the means by which have primarily relied upon the measurements of
hyperuricemia could occur. The following poten- urinary uric acid excretion which are reflections
tial mechanisms need to be considered. First, an of the rate of purine synthesis. On isocaloric,
increased production of uric acid could occur via purine-free diets, 24-h urinary uric acid excretion
the de novo pathway of biosynthesis. Second, an values for normal males have been defined statis-
increased catabolism of tissue purines and the tically as 422 ± 75 mg [1, 2]. Since purine-free
production of urate might be operating. Third, diets are not very palatable and require about
excessive dietary intake of preformed purines a week to establish an equilibrium, such restric-
and their conversion to uric acid could cause tive diets are rarely instituted to estimate purine
hyperuricemia. Fourth, there could be a decrease production in the clinical setting. Instead, it has
in the elimination of uric acid via the kidney. been established that normal individuals excrete
Fifth, a decreased elimination of uric acid via approximately 600–800 mg of uric acid in 24 h

D.S. Newcombe, Gout, 69


DOI 10.1007/978-1-4471-4264-5_4, © Springer-Verlag London 2013
70 4 Uric Acid Metabolism in Humans

on a regular diet. Values of 800–1,000 mg of uri- measured the incorporation of glycine-1-C14 to


nary uric acid/24 h are considered borderline, and assess the rate of purine synthesis. As noted pre-
patients with such values deserve greater scrutiny viously, this small molecule is incorporated into
to determine whether they are truly urate overpro- the purine ring and makes up carbon atoms 4 and
ducers. When values exceed 1,000 mg/24 h, 5 as well as nitrogen atom 7 of the completed
patients are definitely classified as overproducers purine ring. In patients with gout who are urate
of uric acid. In patients with gout, urinary uric overexcretors and most normoexcretors, glycine
acid excretion varies widely from patients who is overincorporated into urinary uric acid when
have values as low as 100–200 mg/24 h to values compared to normal, nongouty subjects. Further,
of more than 2 g/24 h. Low urinary uric acid lev- patients with gout incorporate glycine into uri-
els are observed in those who either have altered nary uric acid more rapidly and to a greater extent
renal function and retention of urates on the basis than normal subjects [3–6]. In some patients,
of renal disease or have specific, yet to be especially those who have lost weight, repeated
identified, renal defects in the excretion of uric measurements of glycine incorporation into uri-
acid. The latter group of patients with diminished nary uric acid after weight reduction have shown
urate excretion probably represents the largest a decrease in the magnitude of the overincorpora-
number of gouty patients. Like all laboratory val- tion [7]. Thus, the degree of overincorporation
ues, urinary uric acid measurements should not can be reduced in some patients, and it has even
be viewed in isolation. Elevated urinary uric acid reverted to normal in one patient [8]. In gouty
levels in the absence of increased levels of serum patients with decreased renal function, the quan-
uric acid may not be a sign of overproduction of tity of uric acid excreted is likely to decline with
urate but may indicate an increased renal clear- a resultant increase in the amount of uric acid
ance of uric acid as might be observed in some eliminated by the gastrointestinal tract. Unless
patients with hereditary renal hypouricemia. the changes in urate kinetics introduced by the
Furthermore, normal urinary uric acid levels do altered renal function are taken into consider-
not exclude the overproduction of urate since ation, overincorporation of the radiolabel into
renal impairment may result in a larger fraction of urinary uric acid may be disguised. If diminished
urate being shunted to extrarenal disposal sites. renal function is considered, then gouty subjects
Documented overexcretion of uric acid on who manifest glycine incorporation within the
more than a single occasion may denote an normal range will show an increased incorpora-
increased synthesis of purines by the de novo tion when corrections are made for the decreased
pathway and indicates the need both to evaluate renal function.
such patients for the underlying cause of the
overproduction and to institute therapeutic
modalities for the prevention of the complica- Miscible Pool of Urate
tions of this metabolic abnormality. It is esti-
mated that probably less than 5–10 % of patients The dietary intake of preformed purines and uric
with primary gout are overproducers of urate. acid, de novo purine nucleotide biosynthesis cen-
Since this small percentage of patients is docu- tered in the liver and blood forming organs, and
mented for those with primary gout, it excludes the various catabolic pathways resulting in the
those with secondary gout due to other disor- breakdown of nucleic acids and/or purine nucle-
ders such as malignancies, psoriasis, and other otides and nucleosides represent the sources of
diseases that may be associated with urate over- uric that contribute to the pool of uric acid
production. The pathophysiological mecha- (Fig. 4.1). The miscible pool, by definition, is the
nisms for primary and secondary gout are quantity of uric acid in the body by which isotopi-
discussed subsequently. cally labeled uric acid administered intravenously
In addition to the measurements of urinary is immediately diluted. The miscible pool does
uric acid excretion, a number of studies have not measure tissue resident urate deposits (tophi)
Miscible Pool of Urate 71

Pathogenesis of gout

De novo purine Dietray intake Cell and tissue


Overproducers biosynthesis turnover

Miscible uric acid pool


Males: 800 − 1,500 mg
Females: 500 − 1,000 mg

Daily urate pool turnover


Males: 480 − 1,050 mg (0 − 0.7 pools/day)
Females: 300 − 700 mg (0.6 − 0.7 pools/day)

Underexcretors Renal uric acid Intestinal Other sources


excretion uricolysis (sweat, tears, etc.)

Fig. 4.1 Serum uric acid levels are a function of endog- and phosphoribosylpyrophosphate synthetase overactiv-
enous and exogenous sources of this trihydroxy purine ity). Increased purine nucleotide turnover is observed in
and its rate of urinary excretion. The miscible pool or some malignant disorders and other metabolic dysfunc-
freely exchangeable uric acid is a reflection of the serum tions leading to an increase in the miscible pool of uric
urate level. De novo purine biosynthesis is increased prin- acid and serum uric acid levels. The regulation of urinary
cipally in patients with heritable enzyme defects (hypox- uric acid excretion also plays a significant role in the con-
anthine-guanine phosphoribosyltransferase deficiency trol of serum uric acid concentrations

which are often not freely exchangeable with the where less freely exchangeable uric acid resides
body pool of urate. In males, the miscible (freely as tissue deposits (tophi). Therefore, the values
exchangeable) pool has an average value of obtained by the one-compartment miscible pool
1,200 mg [9, 10], and in females, values for this methodology underestimate the actual pool size.
pool range from 541 to 647 mg. If body weight is This concept has been confirmed by experiments
considered, then the average pool size in males on tophi removed from patients who have been
is about 16.3 mg/kg of body weight [11]. administered 15N-labeled uric acid. These experi-
In patients with gout, the miscible pool of ments showed that only the peripheral layers of
urate is significantly larger than it is in normal, the tophi were exchangeable with urate in the
healthy subjects. In those without tophaceous soluble pool [12]. In fact, if a two-compartment
deposits, it ranges from 2,000 to 4,000 mg [10, model is considered where the quantity of uric
11], whereas in those with tophi, the pool size acid in tissue deposits like tophi only slowly
may range from 18,000 to 31,000 mg [12]. exchange, if at all, with the soluble pool of uric
All miscible pool measurements are based on acid, the slowly exchangeable urate is estimated
a one-compartment model with urate freely to be 300-fold larger than the freely exchange-
exchangeable and exclude a second compartment able pool [13].
72 4 Uric Acid Metabolism in Humans

Urate Turnover Rate Elevated PRPP Levels

The rate of disappearance of radiolabeled uric In those patients with primary gout that excrete
acid from the miscible pool has been utilized to more than 590 mg/day, the cumulative incorpo-
calculate the turnover rate of uric acid even ration is always above the normal range [15]. As
though the one-compartment model has the limi- noted previously, only a small number of patients
tations noted previously. In nongouty subjects, with primary gout, less than 5–10 %, overpro-
the mean turnover rate for uric acid has been duce uric acid, and the mechanisms by which
reported to be 693 ± 112 mg/24 h [11]. The turn- this overproduction occurs have been defined in
over rates in gouty subjects, as one might guess, two clinical settings, PRPP synthetase overac-
are higher than those in normal subjects, and the tivity and HGPRTase deficiency, and remain
average turnover rate in one study has been undefined in others who overproduce uric acid
found to be 800 ± 130 mg/24 h [11]. However, in and have no recognizable enzyme abnormality.
some gouty subjects who have a miscible pool The mechanism for the overproduction in PRPP
within the normal range, the turnover rates have synthetase overactivity relates to the overpro-
also been found to be within the normal range. duction of phosphoribosylpyrophosphate (PRPP)
Thus, in gouty patients with normal 24-h uric which is the substrate for the rate-controlling
acid excretion and no clinical evidence of tophi, step in de novo purine biosynthesis, the conver-
the uric acid turnover rates are likely to be sion of phosphoribosylpyrophosphate and
entirely normal or only slightly above the nor- l-glutamine to phosphoribosylamine (PRA) by
mal range. Usually, turnover rates are highest in the enzyme, PRPP amidotransferase (Fig. 4.2).
those subjects who overexcrete uric acid, and An excess of the substrate (PRPP) increases the
such rates may reach levels of 2,000 mg/24 h in conversion of PRPP to PRA and subsequently
some overexcretors, whereas normoexcretors leads to an increase in the end product of
have a range of turnover rates from normal to the purine pathway, uric acid. The mechanism
elevated [14]. for urate overproduction in hypoxanthine-gua-
nine phosphoribosyltransferase (HGPRTase)
deficiency is the underutilization of PRPP as a
Mechanisms of Hyperuricemia substrate for the conversion of either hypoxan-
and Gout thine or guanine to inosinic acid or guanosinic
acid, respectively, catalyzed by the enzyme,
A number of studies have been performed to hypoxanthine-guanine phosphoribosyltrans-
measure the rate of purine biosynthesis using ferase. This leaves an excess of PRPP for use as
labeled precursors of uric acid. The most accept- the substrate for PRPP amidotransferase and its
able technique for the measurement of the rate of conversion to PRA (Fig. 4.3). This conversion
uric acid biosynthesis de novo is to use 1-14C- accelerates purine synthesis via the de novo
glycine in tracer amounts and to quantify the pathway. The evidence for this mechanism
radiolabel incorporated into urinary uric acid. comes from experiments showing an increased
Various studies using this method have been aver- production of uric acid as well as increased con-
aged and show that in normal subjects, the mean centrations of PRPP in erythrocytes and
value for incorporation of glycine into urinary fibroblasts from patients with PRPP synthetase
uric acid is 0.17 % in 7 days, whereas in gouty overactivity [16–20]. Similarly, elevations in the
subjects, the average incorporation is 0.28 % over cellular levels of PRPP have also been deter-
the same time period [15]. About one-half the mined in HGPRTase-deficient patients [20].
gouty patients who excrete less than 590 mg/day Cellular concentrations of PRPP are elevated in
have a cumulative incorporation into urinary uric some heterozygote carriers of HGPRTase
acid above the normal range [15]. deficiencies who may, in some cases, develop
Decreased Hypoxanthine Reutilization 73

Regulation of phosphoribosylpyrophosphate synthetase

MgATP Ribose-5-Phosphate

Adenosine diphosphate 2,3-Diphosphoglycerate


(competitive inhibitor) (competitive inhibitor)

PRPP
synthetase

H2O3POCH2
O
O O

O-P-O-P-OH
OH OH
OH OH

Purine nucleotides - ADP and GDP


(noncompetitive inhibitors)

Fig. 4.2 Phosphoribosylpyrophosphate synthetase super- of phosphoribosylpyrophosphate synthetase superactivity


activity leads to an increased production of phosphoribo- has been attributed to defective regulation of this enzyme
sylpyrophosphate, and the excess of this metabolite by its allosteric effectors, catalytic overactivity, combined
activates the rate-limiting enzyme of de novo purine regulatory and catalytic defects, or an increased affinity of
nucleotide biosynthesis, phosphoribosylpyrophosphate the substrate, ribose-5-phosphate, for the enzyme.
amidotransferase. These changes result in the overproduc- Catalytic overactivity is the most common defect, and the
tion of uric acid by the de novo pathway and cause an cause for this mutation has yet to be determined
increased excretion of uric acid via the kidney. The cause

gout. Since some heterozygotes for this enzyme Decreased Hypoxanthine


deficiency also have normal levels of PRPP Reutilization
despite an increased excretion of uric acid and
hyperuricemia, erythrocyte PRPP levels may It is known that decreased reutilization of
not always reflect the levels of PRPP in other hypoxanthine to generate inosinic acid occurs
tissues such as the primary sites of purine syn- in patients with HGPRTase deficiency, and as a
thesis (liver) or may indicate that other factors consequence, more hypoxanthine is available
contribute to the increased production of urate for oxidation to uric acid via xanthine oxidase
in HGPRTase-deficient individuals. In some (Fig. 4.3). This mechanism produces more oxy-
individuals, elevated PRPP levels have been purines including uric acid in those patients with
documented in the absence of an enzyme defect, HGPRTase deficiency and contributes additional
and the mechanism responsible for this change oxypurines to the increased pool of urate gen-
remains unidentified. erated by the accelerated urate production via
74 4 Uric Acid Metabolism in Humans

Purine salvage pathways

OH
OH
HGPRTASE
N
N N
N CH
CH
N
N
N
H2N N
R-P

Guanine
Inosinic acid
+ MgPRPP

OH
OH

N
N N
N CH
CH
N
N
N N H2N
HGPRTASE
R-P
Hypoxanthine
Guanylic acid

Xanthine oxidase

OH OH

N Xanthine oxidase N
N N
CH C OH

N N
N N
HO H HO H

Xanthine Uric acid

Fig. 4.3 In conjunction with the increased levels of phosphoribosyltransferase activity. This mechanism
phosphoribosylpyrophosphate observed in hypoxanthine- together with the increased rate of de novo purine nucle-
guanine phosphoribosyltransferase-deficient patients, the otide biosynthesis driven by the excess phosphoribosylpy-
hypoxanthine formed has no other metabolic route to fol- rophosphate levels accounts for the hyperuricemia in these
low other than its oxidation to uric acid when it is not patients
converted to inosinic acid (IMP) by hypoxanthine-guanine

the de novo pathway. How the abnormality in remains to be determined. Other possibilities
hypoxanthine reutilization might contribute, if at exist to account for those urate overproducers
all, to the unidentified defect(s) in urate overpro- in whom no identifiable defect has been char-
ducers without an identified enzyme deficiency acterized. These include possible abnormalities
Accelerated ATP Degradation 75

in the feedback inhibition sites on the enzyme, in that correction of hypoglycemia results in the
PRPP amidotransferase, and aberrations in the suppression of glucagon levels, a correction of
pentosemonophosphate shunt producing more the hypoglycemia, and a reversal of the hyperuri-
ribose-5-phosphate for the conversion to PRPP, cemia in individuals with glucose-6-phosphatase
the primary substrate for the first step of de novo deficiency [21, 23–30]. In contrast to these exper-
purine biosynthesis. imental results, the infusion of glucagon increases
serum uric acid levels in patients with glycogen
storage disease, type I, as well as increasing the
Glutamine Hypothesis urinary concentrations of oxypurines and uric
acid. Indeed, if labeled ATP is administered to
Although alterations in l-glutamine metabolism, such individuals, glucagon causes an increase in
the other substrate for the rate limiting enzyme, the urinary radiolabeled metabolites originating
PRPP amidotransferase, have been proposed as from ATP. Further, the infusion of somatosta-
possible contributors to enhanced purine synthe- tin to suppress glucagon levels results not only
sis, no evidence has been generated to support in the lowering of the glucagon levels but also
this mechanism of purine overproduction. reductions in the serum urate, urinary uric acid,
and oxypurine levels. There is also a reduction
in the radiolabeled purine products if isotopically
Accelerated ATP Degradation labeled ATP has been infused. These experiments
serve to confirm the hypothesis that increased
In secondary gout, quite different biochemi- serum uric acid and urinary oxypurines in gly-
cal mechanisms cause urate overproduction. cogen storage disease, type I, occur as a result of
These mechanisms include increased adenosine ATP degradation. It is important to recognize that
triphosphate degradation, decreased adenosine there are several forms of type I glycogen storage
triphosphate synthesis, increased nucleic acid disease based on the hepatic microsomal glu-
turnover, and accelerated nucleotide degradation. cose-6-phosphatase complex [31]. The enzyme,
Hyperuricemia and gout are the primary long- glucose-6-phosphatase, is located in the lumen
term complications of glucose-6-phosphatase of the endoplasmic reticulum of hepatic cells in
deficiency (glycogen storage disease, type I), and association with a calcium-dependent regulatory
the mechanism by which hyperuricemia occurs protein and transport proteins (T1, T2, and T3)
in these patients has been shown to be the result which transport glucose-6-phosphate, phosphate,
of an accelerated rate of adenosine triphosphate and glucose, respectively, across the endoplas-
turnover and degradation [21]. In glycogen mic reticulum membrane [31]. The T1 protein
storage disease, hypoglycemia stimulates the transports glucose-6-phosphate to glucose-6-
release of glucagon, and glucagon activates the phosphatase which then hydrolyzes the phos-
hepatic enzyme, glycogen phosphorylase, which phorylated glucose, and the transport proteins,
degrades glycogen to glucose-6-phosphate. The T2 and T3, return the hydrolyzed phosphate and
absence of the enzyme, glucose-6-phosphatase, glucose to the cytosol. Since some patients with
prevents the further catabolism of this metabolite the clinical picture of glycogen storage disease
except the small quantities that may be converted have completely normal glucose-6-phosphatase
to glucose by debrancher enzyme activity [22], activity, alternative explanations for the clini-
and the increasing formation of phosphorylated cal symptoms had to be defined. Recent studies
sugars traps inorganic phosphate and depletes have characterized three additional subsets of the
ATP stores (Fig. 4.4). The depletion of ATP stores type I glycogen storage disease [32]. Type Ia is
leads to the formation of purine nucleoside mono- the classical form of glycogen storage disease
phosphates which are then degraded to inosine, (von Gierke’s disease), type Ib is recognized as
hypoxanthine, xanthine, and uric acid (Fig. 4.5). a defect in the T1 protein or translocase, and type
Experimental evidence confirms this mechanism Ic has been documented as a deficiency of the
76 4 Uric Acid Metabolism in Humans

Glycogenolysis

Glycogen

ATP Phosphorylase
Oligo-1,4-1,4 glucantransferase
Amylo-1,6-gucosidase

ADP

Glucose-1-phosphate

Phosphoglucomutase

Glucose-6-phosphatase

6-phosphogluconate Glucose-6-phosphate Glucose

Phosphohexose isomerase

Fructose-6-phosphate
ATP

Ribulose-5-phosphate Phosphofurctokinase

ADP AMP

Furctose-1,6-diphosphate

Ribose-5-phosphate
+
ATP
Uric acid
Lactate

PRPP + Glutamine PRA Inosinic acid Hypoxanthine

Fig. 4.4 In the absence of glucose-6-phosphatase (glycogen of glucose-6-phosphate to fructose-6-phosphate and even-
storage disease type I), phosphate esters (glucose-6-phos- tually to lactate. The latter small molecule inhibits the
phate) accumulate and inorganic phosphorous levels excretion of urate by the kidney and results in an increase
decrease. Since inorganic phosphorus is a potent inhibitor in serum uric acid levels. Excess glusoce-6-phosphate may
of adenosine monophosphate deaminase, this enzyme is also increase its conversion to 6-phospho-d-gluconolactone
more active with the decrease in inorganic phosphorus and the eventual conversion to phosphoribosylpyrophos-
levels and catalyzes the breakdown of adenosine mono- phate, an activator of de novo purine nucleotide biosynthe-
phosphate to uric acid (see Fig. 4.5). In the absence of sis and an increased production of uric acid
glucose-6-phosphatase, there is an increased conversion
Accelerated ATP Degradation 77

Adenine nucleotide catabolism

Adenosine kinase Adenylate kinase


Adenosin + ATP AMP + ATP 2 ADP

AMP deaminase

Inosine monophophate

Cytoplasmic 5’-nucleotidase

Inosine

Nucleoside phosphorylase

Hypoxanthine

Xanthine oxidase

Xanthine

Xanthine oxidase

Uric acid

Fig. 4.5 Adenine nucleotide catabolism is the final com- 6-diphosphatase deficiency. Decreased adenosine triphos-
mon pathway for the production of hyperuricemia in con- phate synthesis may also lead to hyperuricemia through
ditions where increased adenosine triphosphate the acceleration of adenine nucleotide degradation in
degradation occurs. Such conditions include strenuous conditions such as tissue hypoxia (severe illnesses) and
muscle exercise, ethanol ingestion, glucose-6-phosphatase metabolic myopathies (glycogen storage disease types III,
deficiency (glycogen storage disease), hereditary fructose V, and VII)
intolerance (aldolase B deficiency), and fructose-1,
78 4 Uric Acid Metabolism in Humans

pyrophosphate transport protein (T2). There may carnitine palmityl transferase deficiency, with
also be additional defects to be described includ- muscle phosphoglycerate mutase deficiency, and
ing those with a defect in the glucose transport a patient with a variant form of phosphofructoki-
protein (T3) and possibly the regulatory protein nase deficiency [33–38]. Gouty arthritis has been
for glucose-6-phosphatase. With the exception documented in 30% of the patients with glycogen
of type Ib which is often associated with altera- storage disease, type VII, as well as in a patient
tions in the number (neutropenia) and mobil- with muscle phosphoglycerate mutase deficiency
ity of neutrophils, all these type I subsets have and one with phosphofructokinase deficiency
similar clinical presentations, but they may vary [33]. Tophi have been observed in a patient with
in their severity. Patients with type Ib may have phosphoglycerate mutase deficiency [37]. These
recurrent bacterial infections based on the afore- alterations in purine metabolism associated with
mentioned alterations in the neutrophil. Similar metabolic myopathies may not be recognized in
mechanisms for increased ATP degradation have the absence of exercise testing and the measure-
also been proposed as the cause for the hyperuri- ment of the metabolites, hypoxanthine, inosine,
cemia observed in patients with hereditary fruc- lactate, and uric acid before, during, and after
tose intolerance and fructose-1,6-diphosphatase exercise. Further, myogenic hyperuricemia and
deficiency. gout may be a clue to an underlying disorder in
Several other types of glycogen storage dis- muscle metabolism if muscle pain and weak-
eases have been shown to be associated with ness occur after exercise. Indeed, some patients
hyperuricemia on the basis of alterations in ATP with primary muscle disease will give a his-
metabolism. Glycogen storage diseases, type III tory of pigmented urine due to unrecognized
(debrancher enzyme deficiency), V (muscle phos- myoglobinuria.
phorylase deficiency), and VII (muscle phospho- Ethyl alcohol ingestion has always been
fructokinase deficiency), have all been studied to associated with hyperuricemia and the gouty
characterize the mechanism by which hyperu- diathesis; however, the pathogenesis of this asso-
ricemia occurs. Experiments using leg exercise ciation has not been well understood until rela-
mediated via a bicycle ergometer to measure tively recently. Experiments using radiolabeled
the effects of exercise on purine metabolism in adenine administration to label the adenine pool
such patients clearly document an increase in with the subsequent infusion of ethanol result in
plasma concentrations of ammonia, inosine, and an increase in urine radioactivity by 150 % and
hypoxanthine after a modest degree of exercise in the excretion of urinary oxypurines by 400 %.
[33]. After a delay, the plasma urate concentra- The proposed mechanism for this occurrence is
tion increases, and later, the urinary excretion of the oxidation of ethanol to acetaldehyde by the
inosine, hypoxanthine, and urate also increases. enzyme, alcohol dehydrogenase, and, subse-
These changes are the result of an accelerated quently, the conversion of acetaldehyde to ace-
degradation of muscle purine nucleotides, and tate catalyzed by aldehyde dehydrogenase. Both
the inosine and hypoxanthine produced in the these oxidation reactions result in an increase in
muscle then serve as substrates for the synthesis NADH concentrations at the expense of NAD
of uric acid giving rise to the elevated plasma and cause an increase in the ratio of NADH to
urate levels and the increment in uric acid excre- NAD. The change in that ratio alters the balance
tion (Fig. 4.6). Blood cell hemolysis also occurs between the redox pairs, lactate/pyruvate and
in GSD, type VII, and may contribute to the hype- b-hydroxybutyrate/acetoacetate. The equilibrium
ruricemia on the basis of an accelerated turnover is shifted toward lactate and b-hydroxybutyrate,
of nucleic acids. In addition to these glycog- metabolites that interfere with the excretion of
enoses, other less common disorders of muscle uric acid in the renal tubules. Acetate is then
metabolism have also been associated with hype- converted to acetyl coenzyme A by the enzyme
ruricemia and accelerated ATP degradation with acetyl-CoA synthetase which consumes ATP
exercise. These include reports of patients with in the process of synthesizing acetyl-AMP. The
Accelerated ATP Degradation 79

Nucleotide metabolism in muscle

Adenosine triphosphate (atp)

Glucose
Muscular oxidation, fatty
Activity acid oxidation,
and oxygen

Adenosine diphosphate (adp)

Adenylate kinase

Adenosine Adenosine monophosphate (amp) + atp

Amp deaminase

Inosinic acid (imp)


Adenosine
deaminase 5'-nucleotidase

Inosine + inorganic phosphorus

Purine nucleoside phosphorylase

Hypoxanthine (Hx)

Xanthine oxidase

Xanthine (X)

Xanthine oxidase

Uric acid (UA)

Fig. 4.6 Muscular activity utilizes adenosine triphos- from the conversion of adenosine diphosphate (ADP) to
phate (ATP) as an energy source, and glucose oxidation ATP and AMP may be converted to inosinic acid (IMP) by
along with oxygen and inorganic phosphorus are require- AMP deaminase or to adenosine by 5¢-nucleotidase.
ments for the regeneration of ATP. Metabolic myopathies, Subsequently, adenosine is converted to inosine by ade-
strenuous exercise, and other conditions that limit glucose nosine deaminase and IMP is converted to hypoxanthine
oxidation or fatty acid oxidation may lead to the degrada- by purine nucleoside phosphorylase. Hypoxanthine
tion of ATP. Two routes are available for the catabolism of released by muscle is then oxidized in the liver to xanthine
adenosine monophosphate (AMP). The AMP generated and uric acid by xanthine oxidase

AMP generated from ATP consumption may be converted to hypoxanthine, xanthine, and uric
either be recycled to ATP or degraded to IMP acid by the action of the enzyme, xanthine oxi-
and subsequently to inosine. Inosine may then dase (Figs. 4.5 and 4.7). Experiments utilizing
80 4 Uric Acid Metabolism in Humans

Fig. 4.7 The association Ethanol metabolism


between ethanol ingestion and
hyperuricemia has been
proposed to occur as a result
of an increased degradation of CH3CH2OH
adenosine triphosphate (ATP).
Ethanol is metabolized to NAD+
acetate by a series of
enzymatic reactions (alcohol Alcohol dehydrogenase
dehydrogenase, aldehyde
dehydrogenase, and acetyl- +
NADH+ + H
CoA synthetase). The
conversion of acetate to
acetyl-CoA consumes ATP CH3CHO
and results in the formation of
adenosine monophosphate NAD+
(AMP). The latter metabolite
may then be converted to ATP
Aldehyde dehydrogenase
or deaminated by AMP
+
deaminase to form inosinic NADH+ + H
acid (IMP). Subsequently,
IMP is converted to inosine _
CH3COO
by the enzyme, 5¢-nucleoti-
dase, and inosine is further ATP
converted to hypoxanthine by
purine nucleoside phosphory- Acetyl-coA synthetase
lase activity. Hypoxanthine is
then sequentially converted to PPi
xanthine and uric acid by NH2
xanthine oxidase. This
pathway is shown in Fig. 4.5.
This metabolic model has N
N _
been confirmed by the O O
infusion of acetate into human CH
subjects and documenting the H2C P O C CH3
N
increased ATP degradation N
O
and oxypurine excretion O
H H

H H
OH OH

Acetyl-coA synthetase

Acetyl-coA + Aenosine monophosphate

[14C] adenine in normal subjects have also docu- The other clinical settings in which ATP
mented increased ATP degradation and increased synthesis is decreased and in which hyperurice-
oxypurine excretion [39]. Thus, ethanol induces mia occurs are those associated with tissue
hyperuricemia and perhaps gout by increasing hypoxia resulting either from ischemia or hypox-
ATP degradation and by inhibiting tubular secre- emia [40–44]. Elevated levels of hypoxanthine,
tion of urate on the basis of the generation of lac- xanthine, and inosine have been measured in
tate and b-hydroxybutyrate. patients with hypoxemia and are a reflection of
Increased Nucleic Acid Turnover 81

accelerated ATP degradation [43, 44]. Tissue maximal labeling of urinary uric acid on day
hypoxia and an impaired blood supply limit the 15 in polycythemia, on day 5–10 in secondary
resynthesis of ATP. Thus, the flux of metabolites polycythemia, and on days 3 and 12 in myeloid
is toward uric acid in these patients, and the level metaplasia (two peaks) [45–48]. Additional stud-
of serum uric acid appears to be a biomarker of a ies of patients with myeloid metaplasia who have
poor prognosis [43]. been administered 1-14C-glycine show marked
early and greater labeling of urinary adenine,
guanine, and 7-methylguanine than of urinary
Increased Nucleic Acid Turnover hypoxanthine, xanthine, and uric acid [49, 50].
Such results have suggested that turnover rates
In addition to these examples of secondary hype- for adenosine monophosphate, guanosine mono-
ruricemia and gout, two other mechanisms have phosphate, and other nucleotides are more rapid
been shown to cause these metabolic diseases: than normal. In disorders like polycythemia, uri-
increased nucleic acid turnover and accelerated nary uric acid labeling that peaks at 5–10 days
nucleotide degradation. Increased nucleic acid may reflect an augmented turnover of erythroid
turnover is a major component of lymphoprolif- cells. The peaks in labeling observed with myelo-
erative and myeloproliferative disorders, myeloid proliferative disorders at 12–15 days after the
metaplasia, pernicious anemia, certain hemo- administration of radiolabeled glycine most likely
globinopathies, thalassemia and other chronic indicate augmented turnover in myeloid cells. In
hemolytic anemias, multiple myeloma, and poly- polycythemia vera, the erythrocytes manifest an
cythemia (primary or secondary), and these dis- increased activity of the pentose phosphate path-
eases may also be associated with hyperuricemia, way, augmented generation of phosphoribosylpy-
gout, and hyperuricaciduria. Increased turnover rophosphate (PRPP), increased incorporation
of nucleic acids and bone marrow activity char- of 14C-adenine into nucleotides, and increased
acterize all these disorders, and hyperuricemia activity of adenine phosphoribosyltransferase.
is the common expression of these metabolic If these erythroid precursor cells have an intact
abnormalities. Gout does occur in these condi- de novo purine biosynthetic pathway, the excess
tions but is much less common than the occur- PRPP levels could increase the conversion of
rence of hyperuricemia. The occurrence of gout PRPP to phosphoribosylamine (PRA), the first
and/or hyperuricemia is of clinical significance irreversible step in purine synthesis leading to an
since they may herald the onset of the underlying increased production of uric acid. It is important
disease, and alterations in uric acid metabolism to recognize that alterations in uric acid produc-
can be an early clinical indicator of a hematopoi- tion may first be detected after chemotherapeu-
etic neoplasm. The mechanism for the derange- tic agents have been used to treat the underlying
ment in uric acid metabolism in neoplams is an malignancy and the destruction of cells leads to
accelerated turnover of nucleic acid synthesis the release of massive quantities or nucleotides
since these malignancies utilize large quantities for catabolism to uric acid. Similarly, patients
of nucleotides. Excessive purine nucleotide uti- with untreated pernicious anemia may manifest
lization leads to a reduction in tissue nucleotide altered uric acid metabolism when treatment
concentrations, to the subsequent relaxation of begins and ineffective erythropoiesis ceases.
feedback inhibition of PRPP amidotransferase The hyperuricemia observed in association
activity by nucleotides, and to an increased rate with psoriasis is another example of how
of de novo purine biosynthesis (Fig. 4.8). These increased nucleic acid turnover may contribute to
foregoing changes in purine metabolism result an increased pool of uric acid [51–54]. Studies of
in hyperuricemia, hyperuricaciduria, and, at this skin disease have focused on the increased
times, gouty arthritis. Radiolabeled glycine has epidermal turnover that characterizes this disor-
been used to characterize the urinary purines in der. Increased rates of epidermal turnover may
certain of these disorders and has documented lead to an increased degradation of purines and
82 4 Uric Acid Metabolism in Humans

De novo purine biosynthesis: regulation

H2O3POCH2
O
+ Glutamine
O O

O-P-O-P-OH

OH OH
OH OH

PRPP amidotransferase

Mg2+

H2O3POCH2 NH2
O

OH OH

Inosine 5'-phosphate

Adenosine 5'-phosphate Guanosine 5'-phosphate

Fig. 4.8 The rate of de novo purine nucleotide biosynthe- sylpyrophosphate synthetase by purine nucleotides where
sis is subjected to end-product inhibition by purine nucle- diphosphate and triphosphate nucleotides are more potent
otides (AMP and GMP). Amino- and hydroxy-substituted inhibitors than monophosphate nucleotides. Combinations
purine nucleotides at position 6 of the purine ring bind to of 6-amino and 6-hydroxy substituted purine monophos-
inhibitory sites different from the substrate of the enzyme, phates are synergistic with respect to their inhibition of
phosphoribosylpyrophosphate (PRPP) amidotransferase. PRPP amidotransferase. Purine nucleotide end-product
Monophosphate nucleotides (AMP and GMP) are more inhibition of PRPP amidotransferase is overcome by ele-
potent inhibitors of amidotransferase activity than diphos- vated concentrations of PRPP. Adenosine monosphos-
phate and triphosphate nucleotides (ADP, ATP, GDP, and phate (AMP) also inhibits the conversion of inosinic acid
GTP). This is in contrast to the inhibition of phosphoribo- (IMP) to adenosine monophosphate (AMP)
Accelerated Nucleotide Degradation 83

Amp deaminase

NH2 OH

N N
N AMP deaminase N
CH CH
N
N N N

P-R P-R

Adenosine monophosphate GTP and Pi Inosine monophosphate

Fig. 4.9 A single patient with gout has been character- shown to be insensitive to guanosine triphosphate (GTP)
ized with an alteration in the liver enzyme, adenosine inhibition. This permits the breakdown of AMP to IMP
monophosphate (AMP) deaminase, leading to an acceler- and then the further catabolism of IMP to uric acid (see
ation in nucleotide degradation and an increased produc- Fig. 4.5). In the normal state, AMP deaminase activity is
tion of uric acid. AMP deaminase in this patient was 95 % inhibited by GTP

urate overproduction. However, in some studies, Accelerated Nucleotide Degradation


no correlation has been found between the amount
of skin involvement and serum uric acid concen- Accelerated nucleotide degradation, the other
trations [54–56], and some patients do not mani- proposed mechanism to account for enzyme
fest hyperuricemia. Hyperuricosuria has been abnormalities in primary gout, has only been
documented in many patients with psoriasis, and identified in one patient with gout. Altered deam-
clinical gout has been reported albeit rarely in ination of adenosine monophosphate has been
this disorder. described in a liver specimen obtained from a
Finally, accelerated ATP degradation has been male patient with familial gout. The activity of
documented after strenuous muscular exercise adenosine monophosphate deaminase (AMPD1)
[57–59]. Strenuous exercise in healthy normal assayed in high speed supernatants of human
individuals leads to a depletion in muscle ATP liver documented that enzyme activity was com-
levels by almost one-half and results in a three- pletely inhibited at 0.5 mmol/l GTP in healthy
fold elevation of plasma and urinary oxypurines adults, whereas a residual activity of 28 ± 2 %
[57]. The mechanism by which this occurs is (mean ± SEM of three determinations) was deter-
through the rapid consumption of ATP during mined in the gouty liver specimen. This led the
muscle contractions which surpasses the capacity authors to postulate that a reduction in the sensi-
to regenerate ATP due to the limited supply of tivity of AMP deaminase to GTP was the basis
oxygen and glucose as well as the decreased fatty for increased nucleotide degradation and gout
acid oxidation necessary for its regeneration. The [60] (Fig. 4.9). The hypothesis formulated from
latter metabolic setting leads to the conversion of this single patient is that the increased deamina-
adenosine diphosphate (ADP) to adenosine tion of adenosine monophosphate would provide
monophosphate (AMP) and then to adenosine, greater quantities of inosinic acid for conversion
inosine, hypoxanthine, xanthine, and uric acid to inosine, hypoxanthine, xanthine, and uric acid.
(Fig. 4.6). Although dehydration and hyperlacti- Recent publications have described additional
cacidemia may contribute to the retention of urate parameters of this enzyme that may be pertinent
and an increase in the serum uric acid concentra- to this single report of gout and AMP deaminase
tion, the primary mechanism for these changes in deficiency. A patient with myoadenylate deami-
urate metabolism during muscular exercise is nase deficiency and exertional myalgias, elevated
ATP degradation. creatine kinase, as well as elevated blood lactate
84 4 Uric Acid Metabolism in Humans

levels after ischemic forearm exercise has been Renal handling of uric acid
reported [61]. Is it possible that such patients
might present with hyperuricemia and possibly
gout on the basis of their exertional myalgias and
alterations in uric acid metabolism? Other studies
have shown that AMPD1 mRNA with exon 2
deleted encodes a functional AMPD1 peptide
[62]. This deletion is the result of alternative
splicing, and such alterations modulate the resid-
ual activity of AMPD1 in individuals with mutant 100 %
glomerular
alleles of this enzyme [63]. This is the only case filtration
of abnormal AMP deaminase reported in gout,
and it remains to be seen whether additional cases
will be shown to have this defect. The rescuing
process through alternative splicing may account
for the rarity of the expression of mutations and 98−100 %
clinical findings. reabsorption

Excessive Purine Intake

Certainly, dietary intake of excessive quantities


50 %
of nucleoproteins that can be converted to uric secretion
acid may contribute to an increased pool of uric
acid and hyperuricemia, but it is unlikely that
dietary indiscretions will be the sole factor caus-
ing hyperuricemia.

40−44 %
Elimination of Uric Acid reabsorption

The disposal of uric acid in the human occurs pri-


marily via the kidney and gastrointestinal tract;
minor extrarenal disposal routes also contribute
8−10 %
to the elimination of uric acid such as the saliva, secretion
tears, and perspiration. The renal elimination of
uric acid in the human has been determined to
occur by a four-component system that includes
glomerular filtration, proximal tubular reabsorp- Fig. 4.10 The most significant components of the renal
tion, tubular secretion, and postsecretory reab- handling of uric acid are located in the proximal tubules of
sorption (Fig. 4.10). Of the uric acid presented to the kidney. Urine is completely filtered at the glomerulus and
the renal glomerulus, nearly 100 % is filtered, almost completely reabsorbed in the proximal tubules. About
50 % of the reabsorbed urate is then secreted and a large frac-
and of that amount, about 98–100 % is reab- tion (40–44 %) of the secreted urate is subjected to postsecre-
sorbed by the proximal tubules. Subsequently, at tory reabsorption. Thus, only 6–12 % of the filtered load of
different segments of the proximal tubule, urate urate is excreted in the urine. In the normal adult host on a
is secreted [64]. The product of the plasma urate regular diet, this amounts to 600–800 mg of uric acid per day.
As noted in the text, excretion of quantities of uric acid above
concentration and the glomerular filtration rate is this level usually are classified as overexcretors and may
a reasonable estimate of the filtered load of urate. often represent patients with abnormal purine metabolism
Renal Mechanisms of Hyperuricemia and Gout 85

The actual mechanisms of urate reabsorption and 9


secretion remain incompletely resolved in the
8
human, and their contribution to the urinary uric
acid excretion has yet to be accurately measured 7
in humans. A number of small molecules such as
b-hydroxybutyrate, lactate, acetoacetate, salicy- 6

UUR V (mg/min.)
late, and branched chain amino acids inhibit uri-
5
nary uric acid [65–68], and these alterations in
excretion have been documented to inhibit tubu- 4
lar secretion [68, 69]. These tubular mechanisms
play a significant role in the alterations in serum 3
uric acid observed in certain diseases in which
2
these small molecules are key metabolites. Thus,
renal mechanisms unrelated to gout may cause 1
hyperuricemia in certain patients and can result
in false conclusions regarding the gouty diathe- 0
4 6 8 10 12 14 16 18 20
sis. On the basis of many different studies Plasma uric acid (mg/100 ml)
designed to examine the handling of urate by the
human kidney, the prevailing evidence that best Fig. 4.11 The data in the graph were collected from the
following published studies: Nugent and Tyler [70];
fits what is known to occur is almost complete Seegmiller et al. [71]; Yu et al. [72]; Latham and Rodnan
glomerular filtration of uric acid, almost complete [73]. The open symbols represent data from normal sub-
proximal tubular reabsorption with subsequent, jects and the solid symbols from gouty subjects. Plasma
and perhaps overlapping, tubular secretion and urate levels were either normal values under basal condi-
tions, after RNA feeding, or after infusions of lithium
then postsecretory reabsorption. urate. Data from Latham and Rodnan are represented by
squares, those from Yu are circles, those from Seegmiller
are inverted triangles, and those from Nugent are upright
Renal Mechanisms of Hyperuricemia triangles
and Gout
substrate-velocity curve is postulated to be the
The key abnormality observed in gouty subjects deficit in most patients with primary gout, the
who are not overproducers of uric acid is the fundamental question is what the underlying
requirement of a higher plasma level of urate to cause for this abnormality in the handling of uric
achieve a given clearance of urate when com- acid by the kidney is. Several possibilities have
pared to normal subjects. When urate excretion is been proposed to account for this abnormality
plotted as a function of different plasma urate including decreased glomerular filtration,
levels shows, the S-shaped curve representing the enhanced renal tubular reabsorption, and
data for gouty subjects is shifted to the right as decreased tubular secretion. Data are not avail-
compared to the curve for normal subjects able to confirm any of these possibilities.
(Fig. 4.11). These linear expressions of substrate- Although some have suggested that increased
velocity in normal and gouty subjects show that binding of urate is the cause for this renal defect
gouty patients require serum urate values approx- [74], there is little evidence to support this
imately 1.7 mg/dl higher than normal subjects to hypothesis. On the basis of faulty assumptions
achieve the same uric acid excretion rates. These with regard to the pyrazinamide suppression test
values for gouty subjects are not the result of which is now known to underestimate tubular
prolonged hyperuricemia since patients with leu- urate secretion [75–77], it was originally thought
kemia and hyperuricemia have urate excretion that decreased tubular secretion accounted for the
rates comparable to normal subjects or, in fact, cause of primary underexcretion of urate. Once it
may have increased rates. Since this abnormal was determined that postsecretory reabsorption
86 4 Uric Acid Metabolism in Humans

occurred in the human, it was suggested that control subjects [78]. The published data show-
enhanced reabsorption of urate was the explana- ing that when serum urate levels are decreased,
tion of the hyperuricemia and urate underexcre- the underexcretion of urate returns to normal
tion; however, little direct evidence for this excretion rates have been interpreted as support
mechanism is available. More recent studies pro- for the concept of a defect in tubular secretion of
vide indirect evidence that decreased tubular uric acid [82]. Other studies have utilized pyrazi-
secretion is more likely to account for urate under- namide to analyze postsecretory absorption along
excretion in gouty subjects. Such studies have with probenecid to measure secretory rates since
found that urate secretory rates in gouty subjects it inhibits postsecretory reabsorption. These stud-
who underexcrete uric acid are similar to those of ies documented a decreased uricosuric response
control subjects when plasma urate concentra- in gouty underexcretors [79, 80]. The tubular
tions are decreased [78]. Subsequently, it has been urate secretory rate was unaffected by serum
shown that gouty underexcretors have a decreased urate levels. Despite all these studies, direct evi-
uricosuric response to probenecid and that the dence for a defect in renal tubular reabsorption or
tubular secretion of uric acid is not influenced by secretion of urate in the human remains elusive.
plasma urate concentrations [79, 80]. These data
provide indirect evidence that a defect in urate
renal tubular secretion is responsible for the Renal Mechanisms of Hyperuricemia
underexcretion of urate in gouty subjects. and Secondary Gout
In addition to this renal disposal defect dis-
cussed previously in primary gout, there are For the most part, mechanisms leading to second-
a number of other mechanisms that result in ary hyperuricemia do not lead to gouty arthritis.
decreased urate excretion and could contribute to Exceptions to this generalization include some
the defect in primary gout and to secondary hype- cases of diuretic-induced enhanced reabsorption,
ruricemia and perhaps secondary gout. These perhaps some cases of salicylate-induced decre-
include the following: (1) decreased glomerular ments in tubular secretion, and gout associated
filtration rate, (2) increased tubular reabsorption, with lead intoxication in which the renal mecha-
(3) decreased tubular secretion, and (4) several nism remains incompletely understood. There
unknown mechanisms. In primary gout, although are four mechanisms that account for secondary
altered protein binding of urate has been proposed hyperuricemia in association with decreased
as a mechanism of renal-associated hyperurice- urate excretion: (1) decreased glomerular
mia and could result in a decreased urate filtration filtration rate, (2) increased tubular reabsorption
in the face of a normal glomerular filtration rate, of urate, (3) decreased tubular secretion of urate,
there is little evidence to support abnormalities and (4) unknown mechanisms of decreased renal
in urate binding proteins as a cause for hyperu- excretion of urate.
ricemia and gout in man. Reinterpretation of the One of the most common causes of hyperuri-
pyrazinamide suppression test which originally cemia is observed in patients with chronic renal
did not include any allowance for postsecretory disease and a decreased population of fully func-
urate reabsorption has suggested that enhanced tional nephrons. In order for the failing kidney to
tubular reabsorption of urate could be the expla- maintain homeostasis with respect to uric acid
nation for the hyperuricemia in gouty underex- levels in the plasma, the urate excretion is aug-
cretors [75, 77, 81]. More recent evidence has mented for the reduced number of nephrons indi-
suggested that the site of the defect in the renal cating that glomerular filtration as well as urate
excretion of urate in primary gout is a reduc- tubular reabsorptive and secretory rates is main-
tion in the renal tubular secretion of urate [75, tained until advanced renal disease supervenes.
78]. This concept has been fostered by evidence At that stage, the declining glomerular filtration
showing that plasma urate reduction resulted in rate gives way to a glomerulotubular imbalance
normalization of urate secretory rates to levels of and hyperuricemia results [83]. For this reason
References 87

alone, it is imperative that serum uric acid mea- uric acid, but the exact mechanism by which
surements be accompanied by simultaneous mea- these agents alter this function remains incom-
surements of blood urea nitrogen and serum pletely understood. Nonetheless, with the excep-
creatinine. In this way, the hyperuricemia associ- tion of several of the drugs cited, the alterations
ated with renal failure is indentified promptly, in renal urate clearance caused by these agents
and inappropriate treatment of the hyperuricemia are of little clinical significance.
associated with renal failure is avoided. The Lead is a heavy metal that has been shown to
hyperuricemia of renal failure is probably the cause “saturnine gout,” and intoxication with lead
most common cause of secondary hyperuricemia, leads to impaired urate excretion [99, 100], but
and this mechanism results from a decrement in here again, the mechanism by which this occurs
the glomerular filtration rate. Chronic renal fail- is poorly understood. Of clinical significance
ure is an exceedingly unusual cause for second- with respect to this chemical, secondary gout is a
ary gout. complication of the hyperuricemia resulting from
Diuretic-induced hyperuricemia is probably chronic lead intoxication. For this reason, envi-
almost as common a cause of hyperuricemia as ronmental and occupational sources of lead expo-
renal failure and frequently is a common clinical sure are important to identify in the patient with
setting in which acute gout supervenes especially gouty arthritis.
in the aged female population. The primary mech- In summary, an understanding of uric acid
anism for diuretic-induced hyperuricemia is the metabolism provides the clinician with the ratio-
contraction of the extracellular volume as a result nale for comprehending the biochemical and
of the loss of water and salts causing an increased physiological basis of hyperuricemia and gout.
reabsorption of urate as well as a decreased glom- The vast majority of gouty patients seen by cli-
erular filtration rate [84–87]. Conditions other nicians are likely to have defective renal secre-
than diuretic therapy can also result in hyperu- tion of urate as the underlying cause for their
ricemia by this same mechanism including dia- gout, and a small percentage of such patients
betes insipidus and dehydration. Furosemide, a will manifest an underlying biochemical error in
loop diuretic, also causes lacticacidemia which purine metabolism that will account for their
inhibits urate tubular secretion, and therefore, clinical presentation. The latter group of patients
two mechanisms are operative with this drug. requires a detailed genetic history, a careful
Thus, increased renal tubular reabsorption of investigation of their uric acid metabolism to
urate is the cause for the hyperuricemia observed identify the underlying cause, and treatment to
in clinical settings in which there is contraction prevent complications of the overproduction of
of the extracellular fluid compartment. urate.
As noted previously, a number of organic
acids are secreted by the same mechanism as
urate and may competitively inhibit uric acid
secretion. The most well-defined organic acids
References
including acetoacetate, lactate, b-hydroxybu- 1. Gutman AB, Yu TF. Renal function in gout: with a
tyrate, branched chain keto acids, and low-dose commentary on the renal regulation of urate excretion
salicylate all cause hyperuricemia, and the mech- and the role of the kidney in the pathogenesis of gout.
anism is hypothesized to be the inhibition of Am J Med. 1957;23:600.
2. Seegmiller JE, Grayzel AI, Laster L, Liddle L. Uric
tubular secretion [65, 66, 88]. acid production in gout. J Clin Invest. 1961;40:1304.
A number of other pharmacologic agents 3. Benedict JD, Yu T-F, Bien EJ, Gutman AB, Stetten Jr
including pyrazinamide, cyclosporin A, theo- D. A further study of the utilization of dietary glycine
phylline, warfarin, catecholamines, b-adrenergic nitrogen for uric acid synthesis in gout. J Clin Invest.
1953;32:775.
blockers, methoxyflurane, levodopa, nicotinic 4. Benedict JD, Roche M, Yu T-F, Bien EJ, Gutman AB,
acid, and some nonsteroidal anti-inflammatory Stetten Jr D. Incorporation of glycine nitrogen into uric
drugs [89–98] also alter the renal clearance of acid in normal and gouty man. Metabolism. 1952;1:3.
88 4 Uric Acid Metabolism in Humans

5. Wyngaarden JB. Overproduction of uric acid as the 22. Tsalikian E, Simmons P, Howard C, Haymond MW.
cause for hyperuricemia in primary gout. J Clin Invest. Near normal glucose production in type I GSD.
1957;36:1508. Pediatr Res. 1982;16:265A.
6. Wyngaarden JB. Normal glycine-C14 incorporation into 23. Slonim AE, Lacy WW, Terry A, Greene HL, Burr IM.
uric acid in primary gout. Metabolism. 1958;7:374. Nocturnal intragastric therapy in type I glycogen stor-
7. Emmerson BT. The effect of weight reduction on age disease: effect on hormonal and amino acid
urate metabolism. In: Sperling O, DeVreis A, metabolism. Metab Clin Exp. 1979;28:707.
Wyngaarden JB, editors. Purine metabolism in man, 24. Crigler JF, Folkman J. Glycogen storage disease: new
vol. 41B. New York: Plenum; 1974. p. 429. approaches to therapy. Ciba Found Symp. 1978;55:331.
8. Yu T-F, Roboz J. Long-term follow-up of incorpora- 25. Greene HL, Slonim AE, O’Neill JA, Burr IM.
tion of 15N from glycine into uric acid in gout. Am J Continuous nocturnal intragastric feeding for man-
Med. 1981;70:797. agement of type I glycogen storage disease. N Engl J
9. Wyngaarden JB, Kelley WN. Gout and hyperurice- Med. 1976;294:423.
mia. New York: Grune and Stratton; 1976. p. 1–512. 26. Davisdon AGF, Wong TK, Kirby L, Tze WJ, Rigg JM,
10. Wyngaarden JB, Kelley WN. Gout. In: Stanbury JB, Applegarth DA. Glycogen storage disease type I:
Wyngaarden JB, Fredrickson DS, editors. The meta- effect of continuous nocturnal nasogastric feeding.
bolic basis of inherited disease. 4th ed. New York: Monogr Hum Genet. 1978;9:29.
McGraw-Hill; 1978. p. 916. 27. Ehrlich RM, Robinson BH, Freedman HH, Howard
11. Scott JT, Holloway VP, Glass HI, Arnot RN. Studies NJ. Nocturnal intragastric infusion of glucose in man-
of uric acid pool size and turnover rate. Ann Rheum agement of defective gluconeogenesis with hypogly-
Dis. 1969;28:366. cemia. Am J Dis Child. 1978;132:241.
12. Benedict JD, Forsham PH, Roche M, Sotoway J, 28. Pearlman M, Aker M, Slonim AE. Successful treat-
Stetten Jr D. The effect of salicylates and adrenocorti- ment of severe type I glycogen storage disease with
cotropic hormone on the miscible pool of uric acid in neonatal presentation by nocturnal intragastric feed-
gout. J Clin Invest. 1950;29:1104. ing. J Pediatr. 1979;94:772.
13. Sorenson LB. The pathogenesis of gout. Arch Intern 29. Greene HL, Slonim AE, Burr IM, Moran JR. Type I gly-
Med. 1962;109:379. cogen storage disease: five years of management with
14. Kelley WN, Greene ML, Rosenbloom FM, Henderson nocturnal intragastric feeding. J Pediatr. 1980;96:590.
JF. Hypoxanthine-guanine phosphoribosyltransferase 30. Benke PJ, Gold S. Uric acid metabolism in therapy of
deficiency in gout. Ann Intern Med. 1969;70:155. glycogen storage disease type I. Pediatr Res. 1978;
15. Wyngaarden JB, Kelley WN. Gout. In: Stanbury JB, 12:204.
Wyngaarden JB, Fredrickson DS, editors. The meta- 31. Burchell A. Molecular pathology of glucose-6-
bolic basis of inherited disease. 5th ed. New York: phosphatase. FASEB J. 1990;4:2978.
McGraw-Hill Book Company; 1983. 1078p. 32. Dunger DB, Holton JB. Disorders of carbohydrate
16. DeVries A, Sperling O. Familial gouty malignant uric metabolism. In: Hommes FA, editor. Techniques in
acid lithiasis due to mutant phosphoribosylpyrophos- diagnostic human biochemical genetics: a laboratory
phate synthetase. Urologe A. 1973;12:153. manual. New York: Wiley-Liss; 1991. p. 21.
17. Hershko A, Hershko C, Mager J. Increased formation 33. Mineo I, Kono N, Hara N, Shimizu T, Yamada Y,
of 5¢-phosphoribosyl-1-pyrophosphate in red blood Kawachi M, Kiyokawa H, Wang YL, Tarui S.
cells of some gouty patients. Isr J Med Sci. 1968;4: Myogenic hyperuricemia. A common pathophysio-
939. logic feature of glycogenoses types III, V, and VII. N
18. Becker MA, Losman MJ, Wilson J, Simmonds HA. Engl J Med. 1987;317:75.
Superactivity of human phosphoribosylpyrophosphate 34. Nakagawa C, Mineo I, Kaido M, Fujimura H, Shimizu T,
synthetase due to altered regulation by nucleotide Hamaguchi T, Nakajima H, Tarui S. A new variant case
inhibitors and inorganic phosphate. Biochim Biophys of muscle phosphofructokinase deficiency, coexisting
Acta. 1986;882:168. with gastric ulcer, gouty arthritis, and increased hemol-
19. Becker MA, Meyer LJ, Wood AW, Seegmiller JE. ysis. Muscle Nerve Suppl. 1995;3:S39–44.
Purine overproduction in man associated with 35. Bertorini TE, Shively V, Taylor B, Palmieri GMA,
increased phosphoribosylpyrophosphate synthetase Fox IH. ATP degradation products after ischemic
activity. Science. 1973;179:1123. exercise: hereditary lack of phosphylase or carnitine
20. Becker MA. Patterns of phosphoribosylpyrophos- palmityltransferase. Neurology. 1985;35:1355.
phate and ribose-5-phosphate concentration and gen- 36. Sabina RL, Swain JL, Olanow CW, Bradley WG,
eration in fibroblasts from patients with gout and Fishbein WN, DiMauro S, Holmes EW. Myoadenylate
purine overproduction. J Clin Invest. 1976;57:308. deaminase deficiency. Functional and metabolic
21. Cohen JL, Vinik A, Fuller J, Fox IH. Hyperuricemia abnormalities associated with disruption of the purine
in glycogen storage disease type I: contributions by nucleotide cycle. J Clin Invest. 1984;73:720.
hypoglycemia and hypophosphatemia to increased 37. DiMauro S, Miranda AF, Khan S, Gitlin K,
urate production. J Clin Invest. 1985;78:251. Friedman R. Human muscle phosphoglycerate
References 89

mutase deficiency: newly discovered metabolic myo- 57. Nasralla S, Al-Khalidi V. Nature of purines excreted
pathy. Science. 1981;212:1277. in urine during muscular exercise. J Appl Physiol.
38. DiMauro S, Miranda AF, Hays AP, et al. Myoadenylate 1964;19:246.
deaminase deficiency: muscle biopsy and muscle cul- 58. Knochel JB, Dotin LN, Hamburger RJ. Heat stress,
ture in a patient with gout. J Neurol Sci. 1980;47:191. exercise, and muscle injury: effects on urate metabo-
39. Puig JG, Fox IH. Ethanol induced activation of ade- lism. Ann Intern Med. 1972;81:321.
nine nucleotide turnover. Evidence for a role of ace- 59. Grantham JJ, Chonko AM. Renal handling of organic
tate. J Clin Invest. 1984;74:936. anions and cations; metabolism and excretion of uric
40. Braunwald E, Kloner RA. Myocardial reperfusion: a acid. In: Brenner BM, editor. The kidney. 3rd ed.
double-edged sword? J Clin Invest. 1985;76:1713. Philadelphia: Saunders; 1986. p. 663.
41. Fox IH. Metabolic basis for disorders of purine nucle- 60. van den Berghe G, Hers HG. Abnormal AMP deami-
otide degradation. Metabolism. 1981;30:616. nase in primary gout. Lancet. 1980;2:1090.
42. Grum CM, Simon RH, Dantzker DR, Fox IH. 61. Abe M, Higuchi I, Morisaki H, et al. Myoadenylate
Biochemical indicators of cellular hypoxia in criti- deaminase deficiency with progressive muscle weak-
cally ill patients: evidence for ATP degradation. Chest. ness and atrophy caused by new missense mutation I
1985;88:763. AMPD1 gene: case report of a Japanese patient.
43. Woolliscroft JO, Colfer H, Fox IH. Hyperuricemia in Neuromuscul Disord. 2000;10:472.
acute illness: a poor prognostic sign. Am J Med. 62. Morisaki H, Morisaki T, Newby LK, Holmes EW.
1982;72:58. Alternative splicing: a mechanism for phenotype res-
44. Woolliscroft JO, Fox IH. Increased body fluid purines cue of a common inherited defect. J Clin Invest.
during hypotensive events: evidence for ATP degrada- 1993;91:2275.
tion. Am J Med. 1986;81:472. 63. Morisaki H, Morisaki T, Kariko K, et al. Positive and
45. Yu T-F, Weismann B, Sharney L, et al. On the biosyn- negative elements mediate control of alternative splic-
thesis of uric acid from glycine-N15 in primary and ing in the AMPD1 gene. Gene. 2000;246:365.
secondary polycythemia. Am J Med. 1956;21:901. 64. Goldfinger S, Klinenberg JR, Sigmiller JE. Renal reten-
46. Yu T-F, Wasserman LR, Benedict J, et al. A simulta- tion of uric acid induced by infusion of beta-hydroxybu-
neous study of glycine-N15 incorporation into uric tyrate and acetoacetate. N Engl J Med. 1965;272:351.
acid and heme, and of Fe utilization, in a case of gout 65. Yu T-F, Sirota JH, Berger L, Halpern M, Gutman AB.
associated with polycythemia secondary to congenital Effect of sodium lactate infusion on urate clearance in
heart disease. Am J Med. 1953;15:845. man. Proc Soc Exp Biol (NY). 1957;96:809–13.
47. Lester L, Muller AF. Uric acid production in a case of 66. Schulman JD, Lustberg TJ, Kennedy JL, Museles M,
myeloid metaplasia associated with gouty arthritis, Seegmiller JE. A new variant of maple syrup urine
studied with N15-labeled glycine. Am J Med. 1953; disease (branched chain ketoaciduria): clinical and
15:857. biochemical evaluation. Am J Med. 1970;49:118.
48. Wyngaarden JB. Intermediary purine metabolism and 67. Yu T-F, Gutman AB. Study of the paradoxical effects
the metabolic defects of gout. Metabolism. 1957;6:244. of salicylates in low, intermediate, and high dosage on
49. Wyngaarden JB, Blair AE, Hilley L. On the mecha- the renal mechanisms for excretion of urate in man.
nism of overproduction of uric acid in patients with J Clin Invest. 1959;38:1298.
primary gout. J Clin Invest. 1958;37:579. 68. Gutman AB, Yu T-F. Renal mechanisms for the regula-
50. Bauman RR, Jillson OF. Hyperuricemia and psoriasis. tion of uric acid excretion, with special reference to nor-
J Invest Dermatol. 1961;36:105. mal and gouty man. Semin Arthritis Rheum. 1972;2:1.
51. Eisen AZ, Seegmiller JE. Uric acid metabolism in 69. Campion DW, Olsen RW, Caughey D, Bluestone R,
psoriasis. J Clin Invest. 1961;40:1486. Klinenberg JR. Does increased free serum urate con-
52. Mariani G, Tuoni M, Gianfaldoni R, Molea N, centration cause gout? Clin Res. 1975;23:261.
Giubbolini L, Fusani L, Mian E, Bianchi R. Turnover 70. Nugent CA, Tyler FH. The renal excretion of uric acid
and distribution of 14C-uric acid in psoriatic patients. in patients with gout and in nongouty subjects. J Clin
Adv Exp Med Biol. 1984;165A:277. Invest. 1959;38:1890.
53. Puig JG, Mateos FA, Jimenez ML, Gomez PL, Michan 71. Seegmiller JE, Grayzel AL, Howell RR, et al. The
AA, Vazquez JO. Uric acid metabolism in psoriasis. renal excretion of uric acid in gout. J Clin Invest.
Adv Exp Med Biol. 1986;195A:411–6. 1962;41:1094.
54. Lambert JR, Wright V. Serum uric acid levels in pso- 72. Yu T-F, Berger L, Gutman AB. Renal function in gout.
riatic arthritis. Ann Rheum Dis. 1977;36:264–7. II. Effect of uric acid loading on renal excretion of
55. Brenner W, Gschnait F. Serum uric acid levels in uric acid. Am J Med. 1962;33:829.
untreated and PUVA-treated patients with psoriasis. 73. Latham W, Rodnan JP. Impairment of uric acid excre-
Dermatologica. 1978;157:91–5. tion in gout. J Clin Invest. 1962;41:1955.
56. Sutton JR, Toews CJ, Ward GR, Fox IH. Purine 74. Rieselbach RE, Steele TH. Influence of the kidney
metabolism during strenuous muscle exercise in man. upon urate homeostasis in health and disease. Am
Metabolism. 1980;29:254. J Med. 1974;56:665.
90 4 Uric Acid Metabolism in Humans

75. Diamond HS, Paolino JS. Evidence for a post-secretory 88. Menon RK, Mikhailidis DP, Bell JL, Kernoff PB,
reabsorptive site for uric acid in man. J Clin Invest. Daddona P. Warfarin administration increases uric
1973;52:1491. acid concentrations in the plasma. Clin Chem.
76. Steele TH. Urate secretion in man: the pyrazinamide 1986;32:1557.
suppression test. Ann Intern Med. 1973;79:734. 89. Sugino H, Kagoshima M, Katagiri S. Effects of some
77. Reiselbach RE, Sorenson LB, Shelp WD, Steele TH. drugs on plasma uric acid in rats – actions of cate-
Diminished renal urate secretion per nephron as a cholamines and beta-blocking agents. Nippon
basis for primary gout. Ann Intern Med. 1970;73:359. Yakurigaku Zasshi. 1984;84:293.
78. Puig JG, Anton FM, Jimenez ML, Guitierrez PC. 90. Palestine AG, Nussenblatt RB, Chan CC. Side effects
Renal handling of uric acid in gout: impaired tubular of systemic cyclosporine in patients not undergoing
transport of urate not dependent on serum urate val- transplantation. Am J Med. 1984;77:652.
ues. Metabolism. 1986;35:1147. 91. Hamilton WFD, Robertson GS. Changes in serum
79. Puig JG, Anton FM, Sanz MA, Gaspar G, Lesmes A, uric acid related to the dose of methoxyflurane.
Ramos T, Vazquez OJ. Renal handling of uric acid in Br J Anaesth. 1974;46:54.
normal subjects by means of the pyrazinamide and 92. Weiner IM, Tinker JP. Pharmacology of pyrazin-
probenecid tests. Nephron. 1983;35:183. amide: metabolic and renal function studies related
80. Levinson DJ, Sorenson LB. Renal handling of uric to the mechanism of drug-induced urate retention.
acid in gouty and normal subjects: evidence for a J Pharmacol Exp Ther. 1972;180:411.
4-component system. Ann Rheum Dis. 1980;39:173. 93. Postlethwaite AB, Bartel AG, Kelley WN. Hyperuricemia
81. Ross GR, Seegmiller JE. Hyperuricemia and gout. due to ethambutol. N Engl J Med. 1972;286:761.
N Engl J Med. 1979;300:1459. 94. Gershon SL, Fox IH. Pharmacologic effects of nico-
82. Steele TH, Rieselbach RE. The contribution of resid- tinic acid on human purine metabolism. J Lab Clin
ual nephrons within the chronically diseased kidney to Med. 1974;84:179.
urate homeostasis in man. Am J Med. 1967;43:876. 95. Becker MA, Raivio KO, Meyer LJ, Seegmiller JE.
83. Feinstein EI, Quion VH, Kaptein EM, Massry SG. The effects of nicotinic acid on human purine metab-
Severe hyperuricemia in patients with volume deple- olism. Clin Res. 1973;21:616.
tion. Am J Nephrol. 1984;4:77. 96. Honda H, Gindin RA. Gout while receiving levodopa
84. Steele TH. Evidence for altered renal urate reabsorp- for Parkinsonism. JAMA. 1972;219:55.
tion during changes in volume of the extracellular 97. Morita Y, Nishida Y, Kamatani N, Miyamoto T.
fluid. J Lab Clin Med. 1969;74:228. Theophylline increases serum uric acid levels.
85. Suki WN, Eknoyan G, Martinez-Maldonado M. J Allergy Clin Immunol. 1984;74:707.
Tubular sites and mechanisms action of diuretics. 98. Titinen S, Nissila M, Ruutsalo HM, Isomaki H.
Annu Rev Pharmacol. 1973;13:91. Effect of nonsteroidal anti-inflammatory drugs on
86. Steele TH, Oppenheimer S. Factors affecting urate the renal excretion of uric acid. Clin Rheumatol.
excretion following diuretic administration in man. 1983;2:233.
Am J Med. 1969;47:564. 99. Batuman V, Maesaka JK, Haddad B, Tepper E,
87. Goldfinger S, Klinenberg JR, Seegmiller JE. Renal Landy E, Wedeen RP. The role of lead in gout neph-
retention of uric acid induced by infusion of beta-hy- ropathy. N Engl J Med. 1981;304:520.
droxybutyrate and acetoacetate. N Engl J Med. 100. Ball GV, Sorenson LB. Pathogenesis of hyperurice-
1965;272:351. mia in saturnine gout. N Engl J Med. 1969;280:1199.
Clinical Aspects of Gout
and Associated Disease States 5

Introduction gouty patients, are important to recognize early


since the initiation of treatment may impede
Deposition of a variety of microcrystals including not only the progression of the disease itself
monosodium urate in the synovium and other soft but also some of its complications. Patients
tissues causes acute and chronic arthritis. The with hyperuricemia and gout can be classified
most common clinical disorders associated with into two broad groups: those with increased
microcrystalline synovitis are gout and pseudog- purine biosynthesis or urate production and
out. The latter two disorders may occur as sepa- those with decreased renal clearance of urate.
rate entities or may coexist. The definitive The key target organs affected by gout include
diagnosis of gout depends on the identification of not only the skeletal system and soft tissues but
specific microcrystals in synovial fluid aspirates also the kidney. Although the terms primary
or tissue deposits (tophi). and secondary gout are often used to classify
Gout is the primary clinical expression of gout, the recent literature has increased the
many different underlying dysfunctions, and a complexity of using these terms as a means of
discussion of its clinical presentation should classifying gout. It may be more reasonable to
address both the classical articular disease as well classify and discuss gout in terms of the over-
as the heritable and acquired causes that predis- production and underexcretion of uric acid
pose individuals to gouty arthritis. Heritable and realizing that these two groupings may include
acquired disorders that predispose an individual both primary or innate forms of gout as well as
to gout often have additional clinical findings that secondary forms of gout.
assist the clinician in the identification of an
underlying cause for the gouty arthritis.
Primary gout is most often a disease of adult Clinical Gout
males and includes those individuals who have as
their principal deficit an abnormality in purine Clinical gout in the human may include any one
metabolism. Secondary gout occurs as a result of or more of the following characteristics: ele-
an underlying disorder that leads to an alteration vated serum uric acid concentrations; episodes
in uric acid metabolism and is often associated of an acute, painful mono-, oligo-, or polyar-
with drug administration (diuretics, cyclosporine) thritis; monosodium urate monohydrate crystals
or with disorders of increased nucleic acid turnover observable in synovial fluid aspirates; soft tis-
(myeloproliferative diseases, multiple myeloma) sue deposits of monosodium urate monohydrate
and affects both sexes. Those patients with pri- (tophi) primarily, but not solely, in the vicinity of
mary gout resulting from heritable disorders, even joints; chronic arthritis with deformities mimick-
though they represent only a small percentage of ing rheumatoid arthritis; kidney disease related

D.S. Newcombe, Gout, 91


DOI 10.1007/978-1-4471-4264-5_5, © Springer-Verlag London 2013
92 5 Clinical Aspects of Gout and Associated Disease States

to the deposition of urates or defective tubular


transport; and uric acid nephrolithiasis. Gout can
be separated into three principal stages: acute
gouty arthritis, intercritical gout, and chronic
tophaceous gout. Each of these clinical states
has typical presentations and treatment consider-
ations. Acute gouty arthritis has an abrupt onset
and manifests itself as an exquisitely painful joint
usually confined to the lower extremity. The joint
pain and inflammation typically progress rapidly,
and the patient may be crippled by an acute, pain-
ful arthritis within 4–6 h after the onset of joint
symptoms. As has been often said, the patient
may retire at night in perfect health only to be
awakened with an increasingly painful joint.
Classical gout is a distal monoarticular or oli-
goarticular arthritis often involving the first meta-
tarsophalangeal joint of the big toe (podagra).
This particular joint is involved in 90 % of patients
at some time during the life of their disease. In
addition to the great toe, the arthritis may involve
the instep, ankle, wrist, elbow, fingers, or knee.
Acute and chronic tophaceous gout rarely occurs
in other synovial joints but has been reported to
involve the hips, shoulders, sternoclavicular
joints, and sacroiliac and spinal joints [1–7].
Although involvement of the spine is usually
related to chronic tophaceous gout and is rarely a
component of acute gouty arthritis, back pain
may occur as a feature of gout and can present
infrequently as an acute process. Typically, acute
gout involves one joint but may involve as many
as three (oligoarticular) or more than seven (pol-
yarticular) joints. Only 10–15 % of affected indi-
viduals have polyarticular gout, and this form of Fig. 5.1 Acute gouty arthritis in the first metatarsopha-
gout has a predilection for postmenopausal langeal joint, a podagra. Also acute gout in the ankle joint.
females. It is also not unusual for acute gout to Gout may involve more than one joint simultaneously
involve a bursa with or without accompanying
joint involvement, and the olecranon bursa is the the presence of severe joint pain and fever may
most typical site affected (Figs. 5.1 and 5.2). suggest the presence of a septic joint. In fact, sep-
Joints affected by acute gout are extremely tic joints have been reported to occur in the pres-
painful (too painful to touch), swollen, hot to ence of an acute gouty episode. The pain in acute
palpation, and are red-blue in color at the peak gout is often so severe that the patient will not
of their inflammatory response. Fever and bear weight on the involved part and will even
chills may accompany the inflamed joints. The avoid the weight of bedclothes or a stocking over
inflammation often extends above and below the an involved joint. The skin over affected joints is
involved joint suggesting a cellulitis-like or tense, shiny, and dry. If the acute arthritis goes
superficial thrombophlebitis-like lesion. Further, untreated, the inflammatory response resolves in
Introduction 93

Fig. 5.2 Patient with long-standing chronic tophaceous gout. Urate-lowering therapy was unavailable because of renal
disease and an allergy to allopurinol, before febuxostat and pegloticase were available

Table 5.1 Common factors documented to precipitate


reasonable to start urate-lowering drugs at a lower
acute gout
dose and proceed gradually over days to the fully
Purine-rich diet
recommended dose. If patients give a history of
Rapid weight loss (ketosis)
specific foods as precipitating factors, then they
Alcohol
should be avoided or maintenance colchicine
Micro- or macrotrauma
Acute infections
used if such foods are eaten. Prophylactic colchi-
Post-arthroscopy, cystoscopy, or gastroscopic studies
cine therapy should be used in gouty patients if
Myocardial infarction they are to undergo any surgical procedure. In the
Hemodialysis postsurgical patient with suspected gouty arthri-
Diuretic therapy tis, arthrocentesis must be undertaken to exclude
Intravenous nitroglycerin infection.
Antihyperuricemic treatment without colchicine
Cyclosporine treatment
Postsurgery Intercritical Gout

During an acute episode of gout, the clinical pic-


3–12 days. As the redness and pain subside, there ture and synovial fluid analysis should lead to the
is superficial desquamation of the overlying skin. appropriate diagnosis, but in the asymptomatic
The patient is soon left with no residual symp- intercritical phase of gout, a clinical diagnosis
toms or signs of the acute arthritis. must be based, for the most part, on history alone.
A number of events may serve to precipitate Such a paucity of clinical signs and a poor histo-
acute episodes of gout (Table 5.1). Several gen- rian may lead to an erroneous diagnosis [8, 9].
eral rules can be drawn from these precipitating Recent studies have shown that arthrocentesis of
factors. As a rule, it is best to accompany urate- asymptomatic knees or first metatarsophalangeal
lowering drug therapy with low-dose colchicine joints accompanied by synovial fluid analysis of
to prevent precipitating an acute attack of gout the aspirate using polarized light microscopy
during the time that the serum urate-lowering with a first-order red compensator identified
drug is decreasing the urate pool. It may also be monosodium urate (MSU) crystals in patients
94 5 Clinical Aspects of Gout and Associated Disease States

with asymptomatic gout who were not being


treated with hypouricemic agents [10]. Negatively
birefringent MSU crystals are usually not
observed in gouty patients receiving hypourice-
mic agents or who have low serum uric acid lev-
els. Crystals have also been demonstrated in
patients with gout in joints that have never been
the site of inflammation. Such crystals have also
been reported in joints of patients with hyperuri-
cemia and renal failure [11–13]. Several points
can be made regarding these studies. First, the
mean cell count in MSU crystal-containing syn-
ovial fluids (449 cells/mm3) is often significantly
higher than those synovial fluids without crystals
(64 cells/mm3), and those with crystals contain
more polymorphonuclear leukocytes than those
without crystals. Thus, low-grade inflammation
exists during the intercritical phase of the disease
in the joints that contain crystals. Second, sequen-
tial arthrocentesis showed that once hypourice-
mic drug therapy is initiated to reduce the serum
Fig. 5.3 Radiograph of patient in Fig. 5.1, earlier in her
uric acid levels, it takes months before the joint course. Note tophi in the soft tissues of the index and mid-
crystals disappear. Finally, in the two patients fingers
studied with hyperuricemia, renal failure, and
MSU crystals in their joints, the underlying kid- more severely than the joints of the feet. The dis-
ney disease was not defined, but the types of renal tribution of the joint involvement is asymmetric
disease investigated are of interest. One patient and often affects the big toe, olecranon bursa,
had sarcoidosis, two patients had polycystic kid- prepatellar bursa, or finger joints. On rudimentary
ney disease, one had glomerulonephritis, and in examination, severe tophaceous gout of the hands
five, no etiology could be determined. It would may resemble chronic rheumatoid arthritis.
be of some significance if the five patients with Grossly altered joints often have red shiny skin
MSU crystals had either polycystic kidney dis- over the involved joints, and areas of small whit-
ease or were young individuals with familial ish-yellow subcutaneous tissue deposits of urate
juvenile gouty nephropathy. Follow-up studies of may often be observed, especially if the joints are
patients with intrasynovial MSU crystals and no flexed. In some instances, large urate deposits
history of gout would help determine whether the drain white chalky material that is often mistaken
presence of crystals is a predictor of subsequent for a draining abscess. These draining tophi are
gout. seen most frequently at sites such as the olecra-
non bursa or big toe (Figs. 5.3, 5.4, and 5.5).
Accompanying these bone and joint changes,
Chronic (Tophaceous) Gouty Arthritis one may also see extra-articular urate deposits.
These extra-articular tophi often involve bursae
Chronic gouty arthritis occurs primarily in (especially the olecranon and prepatellar bursae),
patients who have deposits of urate in the bones ear cartilage, and tendons and soft tissue adjacent
adjacent to joints that cause a destructive arthrop- to joints. Tophaceous deposits in certain anatomi-
athy and secondary degenerative arthritis. Distal cal sites may also cause other rheumatic condi-
joints are the principal targets of the disease with tions. Deposits of urate in the wrist have caused
the proximal and distal finger joints often affected carpal tunnel syndrome, and deposits in the hip
Introduction 95

have lead to avascular necrosis of the femoral and finger pads making the diagnosis somewhat
head. Tophi may also be confused with rheuma- confusing [14].
toid nodules, especially when they are found on Chronic gouty arthritis has a more rheuma-
the extensor surfaces of the forearm. Often, the toid-like presentation since the acute inflammatory
gouty tophus is grittier to palpation than the rheu- and severely painful monoarticular attacks of
matoid nodule; however, the presence of morning acute gouty arthritis are replaced by morning
stiffness and rheumatoid-like deformities of the stiffness in the affected joints, swelling, and
hands may require sterile aspiration, if feasible, deformity of multiple joints as well as polyarticu-
of a superficial whitish plaque to demonstrate lar pain. Although this picture may mimic rheu-
urate crystals by polarizing microscopy. Tophi matoid arthritis, these two disorders are rarely
have been reported in visceral tissues as well seen together. Thus, the monoarticular presenta-
including the larynx, heart, and bronchi; deposi- tion with severe inflammation of acute gout pro-
tions in disc spaces may cause paraplegia and gresses over time to a polyarticular condition
other neurological deficits [1–7]. It is important with subacute inflammation (Figs. 5.1 and 5.6).
to recognize that such visceral deposits of urate Organ transplantation and posttransplant ther-
may occur without any sign of peripheral tophi. apies to avoid organ rejection have been associ-
Tophi may also be present in Heberden’s nodes ated in their recipients with a marked increase in
hyperuricemia and tophaceous gout. The two
drugs most frequently associated with the induc-
tion of gout in these patients are diuretics and
cyclosporine. Diuretics reduce extracellular fluid
volume, which results in hyperuricemia, whereas
cyclosporine impairs the renal tubular secretion
of urate [15, 16]. Transplant gout represents a
specific clinical subset of tophaceous gout for the
following reasons. The use of cyclosporine and,
at times, diuretics has resulted in the occurrence
of gout as one of the most common complica-
tions of transplantation. Cyclosporine therapy
results in the rapid production of tophaceous gout
and is also associated with the deposition of urate
in unusual locations such as intramuscularly [17,
Fig. 5.4 Radiograph of patient in Fig. 5.1, after 3 years 18]. Perhaps the most difficult problem is the
of untreated gout

Fig. 5.5 Draining tophus in


the olecranon bursa
96 5 Clinical Aspects of Gout and Associated Disease States

Fig. 5.6 Another patient similar to patient in Fig. 5.1, after many years of untreated gout, leading to chronic pain and
severe joint destruction. Acute gout in a large tophus as well

treatment of tophaceous gout in transplant recipi- that should be excluded from consideration are
ents. Allopurinol, the best drug for the treatment bacterial arthritis (septic arthritis), calcium pyro-
of tophaceous gout, causes the accumulation of phosphate crystal deposition disease (pseudog-
6-mercaptopurine, a drug often used to avoid out), traumatic arthritis, or severe degenerative
organ rejection. Such 6-mercaptopurine retention arthritis. Chronic tophaceous gout is mistaken for
may lead to myelosuppression and its complica- chronic rheumatoid-like arthritis, and often drain-
tions [19]. Any reduction of 6-mercaptopurine ing tophi, especially of the great toe, are treated
dosage to accommodate allopurinol therapy risks as draining abscesses. Both septic arthritis and
the rejection of the graft. In addition to these drug pseudogout may occasionally be accompanied by
interactions, the use of colchicine to prevent acute gouty arthritis, and examination of the arth-
recurrent gouty episodes in transplant patients rocentesis specimen may reveal uric acid crystals
often appears to precipitate colchicine myopathy, along with either bacteria or calcium pyrophos-
especially when the drug is administered along phate crystals. Thus, synovial fluid aspirates from
with cyclosporine [20–22]. Recent studies of the joints where infection or pseudogout are sus-
uricosuric drug, benzbromarone, may provide the pected as the underlying process should be care-
means of treating transplant gout effectively and fully examined with polarization microscopy for
without significant side effects [15, 23]. At the negatively birefringent uric acid crystals.
time of this writing, this drug is not generally Predisposing factors should be evaluated when
available in the United States. septic arthritis is considered in the differential
diagnosis. These include patients in the older age
group; joints damaged by arthritis, especially
Differential Diagnosis rheumatoid arthritis; the presence of a prosthetic
joint; intravenous drug abuse; corticosteroid treat-
A variety of acute monoarticular and polyarticu- ment; and the immunocompromised host. In addi-
lar inflammatory joint disorders may mimic acute tion, patients with diabetes mellitus, Charcot’s
gout, but by in large, the most frequent arthritides arthropathy, cancer, hemarthroses, and chronic
Differential Diagnosis 97

liver disease may also be at increased risk for sep- Almost 90 % of joint infections are caused
tic arthritis. As is well known, patients with an by Gram-positive organisms, whereas about
indwelling venous catheter or those undergoing 10–15 % are the result of Gram-negative infec-
chronic hemodialysis are also at increased risk for tions. S. aureus, Streptococci, and Gram-negative
infection including septic arthritis. bacteria are the most common organisms
The key to the diagnosis of bacterial arthritis observed in septic joints. Gram-negative organ-
is the aspiration of the affected joint and a careful isms causing a septic arthritis include E. coli,
analysis of the aspirate. Except for the culture Pseudomonas aeruginosa, and Serratia marc-
and Gram stain, the results of the synovial fluid escens. Staphylococci are the most common
analysis often are similar, if not identical, to organisms found in intravenous drug abusers,
those observed in both pseudogout and gout. The but Gram-negative infections are also seen. Such
synovial fluid white blood cell count is usually patients must be checked for HIV disease and
above 50,000/mm3 with a predominance of poly- syphilis. Although both Gram-positive and Gram-
morphonuclear leukocytes (neutrophils). Cell negative organisms occur in infected prosthetic
counts of 100,000/mm3 or greater must be joints, low virulence S. epidermidis infections
assumed to be septic until proven otherwise. are often encountered. Anaerobic organisms are
Synovial fluid glucose levels are reduced, but most frequently seen following trauma or bites.
they are only valid if a matching serum sample is Suspected joint infections require immediate
drawn, and the aspiration of the joint is done 2 h intravenous antibiotic therapy with revision of
after a meal. Low levels of synovial fluid glucose the antibiotic regimen, if necessary, once specific
are related to glucose consumption by synovial culture and sensitivity results are available.
fluid cells, but similar levels are observed in Calcium pyrophosphate dihydrate (Ca2P2O7 ·
rheumatoid arthritis and crystal-induced arthri- 2H2O – CPPD) crystal deposition is a multifac-
tis. None of these findings are diagnostic of eted disorder associated either with a heritable
infection. Lactic acid in the synovial fluid may condition, a metabolic disorder, joint trauma
also be elevated but is often normal in Gram- including surgery, or no identifiable cause. Like
negative infections. The Gram stain and synovial gout, CPPD crystal deposition disease presents
fluid culture hold the key to the diagnosis of sep- as a self-limiting inflammatory joint disorder. It
tic joints. For the most reliable results, the aspi- may often follow surgery and may follow a seri-
rate should be immediately plated on blood agar, ous illness such as a stroke or myocardial infarc-
chocolate agar, and in anaerobic culture media. tion. The knee is the joint most often affected,
We prefer to do this at the bedside, but alterna- and the acute attack is similar in character to the
tively, the aspirate may be deposited in blood acute gouty episode except that it is often slightly
culture bottles at the bedside or can be taken to less painful. This disorder may be accompanied
the microbiology laboratory and plated on cul- by degenerative joint changes as determined by
ture media immediately. Gram stains are produc- clinical and radiographic examination. Flexion
tive of an organism about 50–70 % of the time. contractions of the knee are common, and such
Such stains will distinguish Gram-positive from changes are unusual in gout. Patients may also
Gram-negative organisms but will not differenti- present with polyarticular involvement. In addi-
ate staphylococci from streptococci. Blood cul- tion to knee involvement, wrists, metacarpopha-
tures should be done simultaneously with the langeal joints, hips, shoulders, ankles, and elbows
synovial fluid cultures, and if N. gonorrhoeae is may also be affected. The disease may also have
the suspected organism, cultures of the oral cav- other forms of presentation that mimic rheuma-
ity, skin lesions, penile discharge, and vaginal toid arthritis, osteoarthritis, and rarely ankylosing
cavity should be done as well. The presence of spondylitis. Nonetheless, acute CPPD crystal
crystals should not deter the clinician from con- deposition disease, the mimicker of gout, has
sidering infection as the primary cause for the been termed pseudogout for purposes of remind-
joint disease. ing the examiner of its similarity to gout. The
98 5 Clinical Aspects of Gout and Associated Disease States

most typical picture by X-ray is one of linear or and erythematous joint. These acute neuropathic
punctate calcification of the fibrocartilage of the joints usually involve nonweight-bearing joints
knee menisci, symphysis pubis, acetabular and may be painful despite the underlying neuro-
labrum, triangular ligament of the wrist joint, and logical deficit. Chronic neuropathic arthropathies
rarely the annulus fibrosus of the spine. These may also mimic infectious processes and gout.
deposits have been termed chondrocalcinosis, Over time affected joints present with large effu-
and they do not make the absolute diagnosis of sions, are warm to the touch, but manifest little
CPPD crystal deposition disease in the absence or no redness. The tarsometatarsal and midfoot
of CPPD crystals observed in a synovial fluid are the most commonly affected joints in dia-
aspirate. betics, whereas knees are usually the targets for
As with acute episodes of gout, CPPD crystal those with joint disease associated with tabes
deposition disease usually presents with a pain- dorsalis. Upper extremity joints are involved in
ful swollen joint associated with local warmth syringomyelia. Spontaneous fractures and infec-
and redness. It may also be polyarticular mim- tions often complicate neuropathic arthropathies.
icking polyarticular gout. Peripheral blood leu- Radiographic findings are not useful in making a
kocytosis and fever may coexist with the acute specific diagnosis. In acute neuropathic arthrop-
arthritis simulating an infected joint. If CPPD athies, soft tissue swelling and bone resorp-
crystal deposition disease becomes the definitive tion are seen, whereas in chronic neuropathic
diagnosis, then it is reasonable to exclude under- joints, the characteristic picture is one of severe
lying diseases associated with CPPD crystals. osteoarthritis. Synovial fluid analyses are usually
These disorders include hyperparathyroidism, noninflammatory in nature with low white blood
hemochromatosis, hypomagnesemia, and hypo- cell counts and a normal differential count, but
phosphatasia. In patients where management of joint fluids with findings similar to those in sep-
these disorders could alter the prognosis, serum tic joints have been reported so infection must be
calcium, serum parathormone, serum iron, serum excluded. Trauma to such pain-free joints often
ferritin, alkaline phosphatase, and serum magne- results in serosanguinous synovial fluids.
sium are measured to identify these underlying Traumatic arthritis is usually easy to diagnose
diseases. Synovial fluid aspiration and wet syn- since there is a history of an acute injury. In cases
ovial fluid preparations of the aspirate in CPPD where there is an underlying fracture, the joint
crystal deposition disease show variably sized may be painful and warm. Synovial fluid exami-
rhombic and/or needle-shaped crystals. Using a nation usually shows a normal total cell and dif-
polarized microscope with a red-plate compen- ferential count, and no crystals are apparent on
sator, these CPPD crystals show weakly positive careful analysis of a synovial fluid aspirate.
birefringence. Identification of the latter crystals Cultures of the joint fluid are negative.
together with an inflammatory joint or joints pro- There are a number of additional acute arthri-
vides the diagnosis of CPPD crystal deposition tides that may mimic a microcrystalline process
disease. In this setting, it is important to recognize or a septic joint. These include Reiter’s syndrome,
that CPPD crystals are observed in the face of an psoriatic arthritis, acute rheumatic fever, and oth-
infected joint, and the diagnosis and management ers, but septic arthritis, pseudogout due to CPPD
of the infection takes precedence over the treat- crystals, and gouty arthritis remain as the major
ment of CPPD crystal deposition disease. CPPD diseases that need to be excluded when the joint
crystals disappear when wet preparations dry. disease appears to be gout.
There are other clinical disorders that may be Chronic tophaceous gout should never be
associated with CPPD crystals and may infre- diagnosed today, but surprisingly, patients with
quently mimic gout. Charcot joints secondary to deforming tophaceous gout of the hands may
neuropathies associated with diabetes mellitus, infrequently present to physicians for care of
tabes dorsalis, syringomyelia, and other rarer neu- their deformity. In these cases, the hands usually
rological disorders present with a swollen, warm, manifest deformities that may at first glance look
Diagnostic Criteria for Hyperuricemia and Gout 99

like the deformities seen in chronic rheumatoid 12 additional criteria include: (1) monoarticular
arthritis. However, flexing the joints easily dem- arthritis, (2) peak inflammation of a joint within
onstrates the subcutaneous whitish to yellow uric 24 h, (3) joint redness, (4) repetitive episodes of
acid tophaceous deposits. Such deforming lesions acute arthritis, (5) asymmetric swelling of a joint,
of the hands may also simulate psoriatic arthritis, (6) swelling and pain in the first metatarsopha-
systemic lupus erythematosus, hemochromato- langeal joint (podagra), (7) unilateral tarsal joint
sis, and multicentric reticulohistiocytosis. A sim- inflammation, (8) suspected or proven tophus, (9)
ple, sterile aspiration of the whitish subcutaneous elevated serum uric acid, (10) negative synovial
lesions will demonstrate the uric acid crystal fluid culture during an acute episode of joint
when examined by polarized microscopy. inflammation, (11) asymmetrical joint swelling
Another common misdiagnosis seen in in a single joint by X-ray, and (12) radiographic
patients with chronic tophaceous gout is the treat- demonstration of subcortical bone cysts without
ment of a draining tophaceous deposit as a septic erosion. If 6 of these 12 criteria are met or a
arthritis. This is a frequent occurrence in the proven tophus has been found, then one can
emergency room setting where a simple evalua- strongly suspect that the joint disease is gout.
tion of the discharge from the tophus would pro- By definition hyperuricemia is present when
vide the true diagnosis. Of course, draining tophi the solubility of uric acid in plasma is exceeded.
can become secondarily infected, but this is the Age and sex have the most influence on the varia-
exception rather than the rule. tions found in humans. If one evaluates the solu-
bility of uric acid in aqueous solutions with a
sodium ion concentration similar to plasma at a
Diagnostic Criteria for Hyperuricemia temperature of 37 °C, these solutions are satu-
and Gout rated with uric acid at a concentration of 6.8 mg/
dl [24]. If one compares males and females in
The absolute criteria for the diagnosis of gout are large populations, uric acid concentrations at the
based on the documentation of uric acid crystals upper end of the reference range rise in both sexes
in joint fluid with their characteristic strong nega- at puberty with values for females prior to meno-
tive birefringence using compensated polarized pause about 1 mg/dl lower than the value for
light microscopy with a first-order red-plate com- males. In the postmenopausal female, uric acid
pensator. In this case, the needle-like uric acid concentrations at the upper end of the reference
crystals are bright yellow when they are parallel range increase approaching those of the male.
to the red compensator and blue when they are Thus, for adults, most investigators accept the
perpendicular to it. A tophus aspirated under ster- normal serum uric acid levels for males to be
ile conditions may also reveal the characteristic 7.0 mg/dl or less and for females, 6.0 mg/dl or
negatively birefringent uric acid crystals. Either less. Values above this level in adults constitute
of these findings, uric acid crystals from the syn- hyperuricemia. In children, the normal ranges for
ovial fluid, or a tophus along with typical articu- serum uric acid vary with age, and these values
lar symptoms and signs provides an absolute have been reviewed (see Reference 332 for a
diagnosis of gout. Such crystals may be observed complete listing of the publications dealing with
to be present inside cells (intracellular) or outside this subject). Reference ranges for uric acid con-
cells (extracellular). In patients with acute gouty centrations should be obtained by the clinician
arthritis, synovial fluid aspirates show that the from the servicing laboratory, and the number of
major portion of crystals reside within cells. observations required in defining these references
There are other clinical, laboratory, and radio- ranges should be large, age and sex specific, and
graphic findings that, if sufficient in number, may based on a specific method of measurement.
permit the clinician to diagnose gout, but crystal Since hyperuricemia may represent either a tran-
identification is still the preferable means by sient abnormality or a laboratory error, more than
which one can be certain of the diagnosis. These a single uric acid measurement should be obtained
100 5 Clinical Aspects of Gout and Associated Disease States

Table 5.2 Common causes of asymptomatic in uric acid metabolism characterized so that the
hyperuricemia various causes can be segregated (overproduction
Azotemia 20 % and underexcretion).
Acidosis 20 % A brief review of certain factors related to
Diuretics 20 % acute and chronic disease processes and hyperu-
Hypertension/obesity 18 % ricemia such as dehydration, moderate degrees of
Gout 12 % ketosis, or alcohol ingestion is presented here,
Other conditions 10 %
and more extensive discussions are given subse-
quently [26, 27]. Appropriate treatment of these
before more expensive evaluations are under- acute disorders may result in a reversal of an ele-
taken to identify the cause of this biochemical vated serum uric acid level to normal. The patho-
abnormality. genesis of the hyperuricemia of acute disease is
In the absence of gouty arthritis, uric acid variable. A combination of increased nucleic acid
tophi, urolithiasis, or urate nephropathy, elevated turnover, a compensatory increase in purine
serum uric acid levels are classified as asymp- nucleotide synthesis, and an increased produc-
tomatic hyperuricemia. To determine whether or tion of metabolites of purine degradation account
not such a laboratory abnormality is secondary to for the hyperuricemic state of acute leukemia.
an underlying disease or the use of drugs affect- Articular complaints in patients with leukemia
ing urate metabolism, knowledge of those dis- are not always the result of hyperuricemia [28].
eases and drugs associated with changes in the The hyperuricemia associated with diabetic
serum uric acid is essential. Common causes of ketoacidosis, exercise, starvation, and toxemia of
asymptomatic hyperuricemia have been delin- pregnancy is, for the most part, related to the
eated, as have those conditions associated with inhibitory effects of small organic molecules
acute and chronic illnesses (Tables 5.1 and 5.2). (lactate, beta-hydroxybutyrate, and acetoacetate)
Careful evaluation of patients with persistent on the renal excretion of urate. The cause of the
hyperuricemia is necessary to exclude the pres- hyperuricemia observed with acute myocardial
ence of underlying disease and to determine fac- infarction is a decreased excretion of urate [29].
tors such as urinary urate excretion that would Decreased renal perfusion is the primary factor
influence decisions to treat asymptomatic hype- implicated in the hyperuricemia of myocardial
ruricemia. A family history of gout or renal cal- infarction, but hypoxia and increased production
culi is often helpful. Of course, the variations in of uric acid secondary to the breakdown of tissue
serum uric acid levels with age, sex, and race must nucleotides may also contribute to the hyperuri-
be considered before concluding that asymptom- cemia observed in association with insults to the
atic hyperuricemia exists. Hyperuricemia has myocardium, vigorous exercise, grand mal sei-
been documented in between 2 and 18 % of adult zures, severe respiratory acidosis, and congestive
populations evaluated [25]. Certain simple labo- heart failure [30–40] (Table 5.3).
ratory tests may further distinguish the causes of Ethanol consumption has long been associated
persistent asymptomatic hyperuricemia. These with an increased prevalence of hyperuricemia
include measurements of serum creatinine, uri- and gout, and populations with a high incidence
nary ketones (diabetes mellitus and starvation), of gout often have increased numbers of alco-
blood lactate levels, blood gases and pH (beryl- hol imbibers [41, 42]. Ethanol-induced hyperu-
liosis and glycogen storage disease), and fasting ricemia results from two different mechanisms.
blood lipid levels (hyperlipoproteinemia). When Early studies have clearly shown that ethanol
a reasonable suspicion of lead exposure exists, the ingestion resulted in elevated blood lactate levels
determination of blood and urinary lead levels are which were proposed as the cause for hyperuri-
indicated (see section “Saturnine gout”). The per- cemia since lactate competitively inhibits uric
sistence of significant hyperuricemia (10–13 mg/ acid clearance by the proximal renal tubule [43].
dl) mandates that 24-h urinary uric acid concen- Subsequently, it has been shown that ethanol is
trations be determined and the underlying defect converted to acetate through oxidative pathways.
Diagnostic Criteria for Hyperuricemia and Gout 101

Table 5.3 Acute pathologic processes associated with denotes the sudden impairment of kidney func-
hyperuricemia tion without regard to cause or mechanism. It may
Acute leukemia be due to prerenal (diminished renal blood flow
Acute myocardial infarction from a variety of causes), postrenal (obstruction
Diabetic ketoacidosis to urine flow), or intrinsic renal disease involving
Ethanol intoxication glomeruli, tubules, interstitium, vasculature, or a
Exercise combination of these sites. With the exception of
Grand mal seizures
acute rhabdomyolysis, the hypercatabolic states
Heat stress nephropathy
associated with lymphoproliferative or myelo-
Infectious hepatitis
proliferative disorders, or chemotherapy-induced
Infectious mononucleosis
Severe respiratory distress
cell lysis, patients with acute renal failure all
Starvation with ketosis manifest increased plasma uric acid levels. Such
Toxemia of pregnancy increments are usually about 1–2 mg/dl above
Acute renal failure normal. Such levels rarely exceed 15 mg/dl since
intestinal disposal of urate and urate hydroly-
sis by gut bacteria increase and usually prevent
Ethanol-induced acetate is then metabolized to greater increments in the serum levels. The incre-
acetylcoenzyme A using high-energy phosphates ments in plasma urate observed in acute renal
derived from adenosine triphosphate. The lat- failure result either from decreased renal perfu-
ter molecules are then converted to adenosine sion, diminished urate excretion by the kidney, or
monophosphate in the process [44]. Precisely a combination of the two. If the serum or plasma
two moles of ATP are used for every mole of urate level exceeds 20 mg/dl, then rhabdomyoly-
ethanol oxidized. Since acetate infusions in man sis including muscle necrosis secondary to severe
markedly increase urinary oxypurine and plasma status epilepticus, cell lysis, or a combination of
hypoxanthine levels, there is strong support for fasting and strenuous physical exercise are likely
the concept that the adenosine monophosphate to be the causes [31, 35]. The serum uric acid
generated during ethanol metabolism is degraded concentration increases rapidly in acute renal
to uric acid accounting for the hyperuricemia failure as renal function progressively deterio-
observed with ethanol ingestion. It appears that rates, and a marked catabolic state often occurs
blood alcohol levels of less than 150 mg/dl are not with increased uric acid synthesis.
associated with hyperlacticacidemia, and in such Hyperuricemia in rhabdomyolysis results
a setting, adenosine monophosphate degrada- from the activation of 5¢-nucleotidase when mus-
tion accounts for the hyperuricemia. When blood cle cell ATP is significantly lowered which then
alcohol levels exceed 200 mg/dl, uric acid excre- reduces the adenylate pool and increases inosine
tion is impaired and hyperlacticacidemia contrib- synthesis and in turn uric acid production. As a
utes to the ethanol-induced hyperuricemia. result of muscle cell necrosis, nucleoproteins are
In recent years, episodes of acute gout have released which are subsequently converted to uric
been observed in association with the adminis- acid by the liver. Thus, rhabdomyolysis causes
tration of intravenous nitroglycerin for unstable increased nucleoprotein catabolism and uric acid
angina [45]. Since intravenous nitroglycerin synthesis from inosine. The latter mechanism
preparations at the time contained significant may also be operative during vigorous exercise.
quantities of ethanol (Tridil containing 50 mg In patients combining weight reduction and
of nitroglycerin per 10 ml of 30 % ethanol and fasting, the acetoacetate and b-hydroxybutyrate
Nitrostat containing 5 % ethanol by volume), produced by fasting and the lactate generated by
reports of acute gouty arthritis observed during and physical exercise interfere with the secretion of
after intravenous nitroglycerin have been attributed urate by the renal tubules and results in hyperuri-
to ethanol-induced hyperuricemia with the subse- cemia [32, 36]. Rarely, acute renal failure has
quent occurrence of gouty arthritis [46]. Acute been observed in the management of solid tumors
renal failure, another cause of hyperuricemia, as contrasted with its more common association
102 5 Clinical Aspects of Gout and Associated Disease States

with leukemias and lymphomas treated with radi- process without the need for a 24-h urinary uric
ation or chemotherapy. One such case has been acid collection and analysis. As will become clear
reported in a patient who received streptozotocin from the following discussions, there are a num-
treatment for an islet cell carcinoma of the pan- ber of disorders of uric acid metabolism that
creas [47]. The significance of this case is that uric require a more extensive evaluation in order to
acid nephrolithiasis and acute renal failure make a prudent decision about the management
occurred in the absence of any apparent acute and treatment of the underlying alteration in
tumor cell lysis accompanying the onset of oligu- metabolism. A 24-h urinary uric acid measure-
ria and in the presence of normal serum uric acid ment is the key to assessing whether a patient is
levels. Streptozotocin, a nitrosourea derivative, is an overproducer or underexcretor of uric acid;
known to be associated with a variety of renal such information is essential in some cases of
tubular dysfunctions including Fanconi syndrome gout in order to identify the underlying disorder
with hyperuricosuria and could have been respon- and to initiate appropriate treatment.
sible for the induction of a defect in the renal tubu- Certain precautions are necessary to ensure
lar reabsorption of uric acid in this patient [47]. the proper 24-h urine collection for uric acid
The mechanism for the hyperuricemia observed measurements. Urine should not be refrigerated
after seizures (grand mal type) is related to the during the collection since uric acid solubility
production of lactic acid from intense muscular decreases at low temperatures and precipitation
activity and a subsequent decrease in the renal of urate crystals may result in the underestima-
tubular elimination of urate [40]. Correction of tion of urinary uric acid levels. A small quantity
carbon dioxide retention in severe respiratory aci- of toluene (5 ml) used to be added to the urine
dosis often results in the reduction of serum uric collection bottle to prevent bacterial growth and
acid levels and increased urinary uric acid excre- uricolysis by bacteria. Today, a different noncarci-
tion. The mechanism for the hyperuricemia in heat nogenic preservative is added to prevent bacterial
stress nephropathy, infectious hepatitis, and infec- growth. The uricase method for uric acid deter-
tious mononucleosis is less well defined [48]. mination should be used since this methodology
In summary, a number of acute and chronic avoids interference from urinary chromagens.
illnesses are associated with elevated concentra- The recent use of X-ray contrast material should
tions of serum uric acid, and these conditions also be avoided prior to the urine collection since
must be excluded before embarking on a more they may increase urinary excretion of uric acid
extensive analysis of the causes for this labora- (see section “Uric acid nephrolithiasis”). Further,
tory abnormality. The measurement of serum uric radiocontrast materials can cause acute renal fail-
acid should never be ordered as a sole parameter ure and hyperuricemia, even after relatively small
and should always be evaluated in conjunction amounts of these substances have been adminis-
with some measurement of renal function such as tered [49–54]. Finally, a careful history of the
serum creatinine or blood urea nitrogen. Such a drug usage including the use of nonprescription
practice identifies renal disease as a cause for the drugs containing salicylates must be sought to
hyperuricemic state. Finally, a more extensive avoid erroneous measurements secondary to the
discussion of drug-induced causes of hyperurice- influence of drugs on uric acid excretion.
mia is included in the section “Drug-induced
hyperuricemia and gout”.
Overproduction of Uric Acid

Urinary Uric Acid Measurements Even though gout presents as a single clinical
entity, there are multiple causes of the disease.
Information from the medical history, physical Each of these separate causes may have special
examination, and laboratory data may be sufficient and distinct clinical findings that help to identify
to establish the etiology of the hyperuricemic it and may denote the need for additional
Overproduction of Uric Acid 103

therapeutic initiatives. This section places empha- choreoathetosis, spasticity, aggressive behavior,
sis on the clinical aspects of the different genetic and mental retardation. Affected patients mani-
and acquired disorders leading to the overproduc- fest the complete absence of the enzyme, hypox-
tion of uric acid resulting in hyperuricemia and anthine-guanine phosphoribosyltransferase, and
gout (Table 5.2). Some of these inherited disor- this enzyme deficiency is inherited as an X-linked
ders (Lesch-Nyhan syndrome, Kelley-Seegmiller recessive trait. On the basis of the pattern of
syndrome, phosphoribosylpyrophosphate syn- inheritance, this disorder affects primarily males,
thetase superactivity) can be classified as primary but a few females have been reported who have
gout, whereas others (glycogen storage diseases, inherited separate mutations in this enzyme from
fatty oxidation deficiencies, tissue hypoxia, etha- father and mother. All patients with the Lesch-
nol ingestion, blood diseases, malignancies, pso- Nyhan syndrome manifest markedly excessive
riasis, and dietary excesses) are clearly secondary uric acid production and may suffer from renal
forms of hyperuricemia and gout. Disorders asso- calculi as the major complication of their altered
ciated with overproduction of uric acid probably uric acid metabolism. These abnormalities of
account for no more than 5–10 % of all the cases purine metabolism may also cause hyperurice-
of gout. It is important to recognize that the inher- mia, uric acid crystalluria, and obstructive
ited diseases have different modes of inheritance, uropathy.
and these patterns of inheritance may assist in Nonspecific signs of central nervous system
determining the underlying genetic disease. dysfunction may be present at birth in affected
The principal mechanism for those disorders infants, and the most common of these is general-
characterized by increased production of uric ized hypotonia. The first clear appearance of
acid in primary hyperuricemia and gout is an specific neurological findings usually begins
increased intracellular level of phosphoribosylpy- between 3 and 6 months of age [55–59].
rophosphate (PRPP). Such elevated intracellular Established neurological dysfunction usually
levels of PRPP enhance purine synthesis by makes itself known at age 1 or 2 years, and
increasing the rate of conversion of PRPP to affected children may be diagnosed with cerebral
phosphoribosylamine by the first enzyme in palsy until self-mutilation makes its appearance.
de novo purine biosynthesis. Idiopathic forms of Both signs of pyramidal and extrapyramidal tract
gout include those individuals who overproduce involvement may be observed. Athetoid move-
uric acid for which no mechanism has been char- ments of the extremities that may become chore-
acterized. Secondary types of hyperuricemia and iform over time characterize the extrapyramidal
gout with increased uric acid production include manifestations. Some patients are hypotonic and
those diseases in which increased ATP catabo- develop hyperkinetic movements with dystonia
lism or decreased ATP synthesis occur, and those [60–62]. Hyperreflexia, ankle clonus, extensor
disorders associated with increased nucleic acid plantar responses, leg scissoring, and spasticity,
turnover. Secondary hyperuricemia and gout may if present, are manifestations of pyramidal tract
also occur in those extremely rare instances in involvement.
which the patient ingests excessive quantities of Dysarthria is also a prominent feature of the
purines. Lesch-Nyhan syndrome, and its presence makes
communication with these patients most difficult.
Although patients may be able to communicate
Lesch-Nyhan Syndrome single words, full sentences are almost always an
impossibility. This speech defect along with a
Despite the profound changes in uric acid metab- moderate degree of mental retardation makes
olism observed in the Lesch-Nyhan syndrome, clinical management of these patients exceed-
the clinical presentation is not usually one of ingly complex [57, 63]. A rare patient may have
gouty arthritis but is characterized by a severe normal intelligence, and so the absence of mental
neurological disorder including self-mutilation, retardation does not exclude the Lesch-Nyhan
104 5 Clinical Aspects of Gout and Associated Disease States

syndrome [64]. Those patients with extensive aggressive and self-mutilating behavior is char-
neurological disease are usually confined to a acteristic of the Lesch-Nyhan syndrome and does
wheelchair and are often incontinent. In addition not occur in those individuals who have a partial
to these neurological deficits, growth may be deficiency of the enzyme, hypoxanthine-guanine
retarded in some affected individuals. phosphoribosyltransferase. Self-injurious behav-
Several key symptoms of the Lesch-Nyhan ior is a problem in some autistic and non-autistic
syndrome provide additional clues to the under- retarded children, but it is most commonly a clin-
lying diagnosis. Some infants may have episodic ical problem associated with the Lesch-Nyhan
irritability associated with red-orange crystals on syndrome and Tourette’s syndrome [66]. Recent
their diapers. Subsequently, urinary tract infec- studies have documented a reduction in dopamine
tions and obstructive uropathy may occur as a transporters in the basal ganglia of Lesch-Nyhan
manifestation of uric acid crystalluria and uric patients. Further, radiographic studies may pro-
acid nephrolithiasis. Hematuria and colicky vide a method for the recognition of these basal
abdominal pain may occur in association with ganglia alterations [67–69]. Although specific
these findings, but pure urate stones, themselves, therapy for self-injurious behavior is unavailable,
are radiolucent making their visualization impos- anticonvulsants and carbamazepine have offered
sible on a simple flat plate of the abdomen. some relief for this symptomatology [70].
Retrograde urography is necessary for their rec- Hematologic abnormalities have also been
ognition as cyst-like structures. These urinary reported in the Lesch-Nyhan syndrome, but their
tract features are the result of the massive excre- pathophysiology is unclear [71–73]. Usually
tion of uric acid (25–140 mg/kg × day) as com- affected patients are anemic, and the anemia is
pared to the normal value (18 mg/kg × day) [65]. macrocytic, but hemolytic anemia has also been
Acute renal failure as a result of renal urate depo- reported [74]. Some patients have low folate lev-
sition may also occur, and kidney stones may be els as a result of the consumption of this metabo-
the only initial manifestation of this disorder. lite by the increased rate of de novo purine
Thus, although gouty arthritis is a rare sign of the synthesis, but the megaloblastic anemia may be
Lesch-Nyhan syndrome, the excessive uric acid unresponsive to folate treatment [75]. No other
excretion leads to deleterious effects on the hematologic abnormalities of clinical significance
kidney. have been reported. Thus, severe neurological
Along with these neurological and urinary diseases characterized by choreoathetosis, spas-
tract findings, the most distinguishing and unusual ticity, and self-injurious behavior along with uri-
feature of the Lesch-Nyhan syndrome is the com- nary tract problems are the most typical clinical
pulsion for self-injury that occurs in affected findings in the Lesch-Nyhan syndrome
patients. These patients manifest intermittent, (Table 5.4).
compulsive mutilation, principally of their lips,
buccal mucosa, fingers, and hands. Stress such as
hospitalization seems to exacerbate or increase Kelley-Seegmiller Syndrome
the self-injurious behavior. At times, no manifes-
tations of self-mutilation are present, and no cor- Although the Kelley-Seegmiller syndrome, a par-
relation with clinical findings has been defined to tial deficiency of hypoxanthine-guanine phos-
account for these periods of freedom from their phoribosyltransferase, is an uncommon cause of
self-mutilating behavior. Along with this aber- gout, recognition of its clinical presentation can
rant behavior, Lesch-Nyhan patients also exhibit lead to the prevention of its complications through
a bizarre form of aggressiveness. They have been appropriate early treatment. Like the complete
known to strike others, spit at them, or kick them. deficiency of hypoxanthine-guanine phosphori-
They may also destroy any implements that are bosyltransferase, it is also inherited as an X-linked
within their environment or use them as weapons recessive trait. Its primary clinical parameters
against other individuals or other objects. This involve the musculoskeletal system, urinary tract,
Overproduction of Uric Acid 105

Table 5.4 Classification of hyperuricemia and gout birth, the resultant altered urate metabolism usu-
I. Increased production of urate ally does not give rise to gout until after the age
A. Primary hyperuricemia and/or gout of 20. Tophaceous deposits in these patients may
1. Elevated PRPP develop in unusual locations. The rapid develop-
(a) Lesch-Nyhan syndrome ment of tophi is a marker of the overproduction
(b) Kelley-Seegmiller syndrome of uric acid and the body’s inability to handle the
(c) PRPP synthetase variants increased load of urate.
2. Idiopathic Kidney stones may be the first manifestation
B. Secondary hyperuricemia and/or gout of disordered urate metabolism, and uric acid
1. Increased ATP catabolism
crystalluria and/or nephrolithiasis may occur
(a) GSD types I, III, V, and VII
in the teens or early twenties. In some, kidney
(b) Muscle phosphoglycerate mutase (?)
stones have been passed in patients younger than
(c) Tissue hypoxia
8 years of age [76]. Accompanying this manifes-
(d) Ethanol
2. Increased nucleic acid turnover
tation of urate overproduction are chronic urinary
(a) Blood diseases tract infections and renal insufficiency. In fact,
(b) Malignancies acute renal failure has been reported as an ini-
(c) Psoriasis tial event in patients with partial hypoxanthine-
3. Excessive purine intake guanine phosphoribosyltransferase deficiency
II. Decreased excretion of urate [77, 78]. Several patients have undergone neph-
A. Primary hyperuricemia and/or gout rectomy as a result of severe infection. These
1. Decreased filtered load (?) episodes of renal insufficiency and ultimate fail-
2. Increased tubular reabsorption (?) ure are treatable disorders in children and young
3. Decreased tubular secretion adults, if recognized early in the course of this
B. Secondary hyperuricemia genetic disorder. The recognition of this enzyme
1. Reduced glomerular filtration rate deficiency is absolutely essential if treatment is
(a) Chronic renal failure to be successful.
2. Increased renal tubular reabsorption
Neurological dysfunction in these patients is
(a) Dehydration
variable, and its manifestations are heteroge-
(b) Diabetes insipidus
neous. The neurologic findings include nystag-
(c) Diuretics
mus, hyperactive deep tendon reflexes, extensor
3. Decreased tubular secretion
(a) Disorders with increased ß-hydroxybutyrate
plantar responses, spastic movements, and dysto-
(b) Disorders with increased acetoacetate nia. Peripheral nervous system alterations may
(c) Starvation result from compression neuropathies (carpal
(d) Diabetic ketoacidosis tunnel syndrome) secondary to tophaceous
(e) Branched chain ketoaciduria deposits. Mental retardation of a modest degree
(f) Disorders with hyperlacticacidemia is also seen in this disorder in some patients. The
(g) Toxemia majority of patients with partial deficiency of
(h) Acute alcoholism hypoxanthine-guanine phosphoribosyltransferase
(i) Tissue hypoxia have no neurological deficits, and the reasons for
the appearance of central nervous system disease
in some must await the complete linkage of the
and kidney. In some patients, central nervous sys- biochemical changes with the nervous system
tem dysfunctions may also be apparent, but no symptoms and signs.
self-injurious behavior occurs. Affected patients The clinical features of this enzyme deficiency
clearly suffer from recurrent and severe gouty and its concomitant alterations in purine metabo-
arthritis, and they may develop tophaceous depos- lism are reflected in the changes in uric acid
its shortly after the onset of gouty arthritis. metabolism. The serum uric acid concentrations
Despite the presence of the mutant enzyme from in most of the affected patients are in the range of
106 5 Clinical Aspects of Gout and Associated Disease States

10 mg/dl, and most patients excrete more than g excreted/24 h), and neurodevelopmental abnor-
600 mg of uric acid per day in their urine. In addi- malities including ataxia, mental slowness,
tion, the uric acid to creatinine ratio in 24-h urine sensineural deafness, hypotonia, and delayed
specimens is usually greater than 2. motor development. The neurological defect
Just as some patients with a complete identified most often in these patients is a high-
deficiency of hypoxanthine-guanine phosphori- frequency deafness [81–83]. The mechanisms
bosyltransferase have a megaloblastic anemia, causing these neurological deficits and their link-
some patients with a partial deficiency of the age to purine biosynthesis are unknown. The
same enzyme also manifest a megaloblastic ane- juvenile or early adult onset phenotype is also
mia often with an accompanying low serum folate characterized by uric acid overproduction, gout,
level. The pathogenesis of this finding remains to and/or uric acid nephrolithiasis, but no neurologi-
be completely understood. cal deficits have been found in these patients [81].
Finally, as one might suspect, patients with There is no definite single mutation that segre-
the partial deficiency are also resistant to aza- gates these two phenotypes except for the fact
thioprine treatment because of their inability to that the childhood form signals a markedly exces-
form the therapeutically active nucleotide. Thus, sive rate of urate overproduction and usually
if such patients require treatment with antime- involves a mutant with a defect in regulation or
tabolites like azathioprine, alternative drugs an abnormality in regulation combined with
that do not require hypoxanthine-guanine phos- another defect.
phoribosyltransferase for their activity should The biochemical mechanism leading to these
be selected. Even though these human mutants abnormalities in uric acid production results from
probably represent less than 1 % of the patients the overproduction of phosphoribosylpyrophos-
with gout, their identification and treatment will phate. Increased phosphoribosylpyrophosphate
reduce morbidity and mortality from this treat- levels, whether they result from underutilization
able disorder. of this metabolite in purine salvage reactions or
by overproduction by phosphoribosylpyrophos-
phate synthetase activity, activate phosphoribo-
Phosphoribosylpyrophosphate sylpyrophosphate amidotransferase and increase
Synthetase Overactivity de novo purine nucleotide biosynthesis and
excessive uric acid production.
Increased activity of the enzyme, phosphoribo- The keys to suspecting this enzymatic abnor-
sylpyrophosphate synthetase, has been described mality are the presence of a neurological deficit,
in small number of kindreds and is characterized especially sensineural deafness, associated with
by uric acid overproduction, gout, and urolithia- signs of uric acid overproduction such as topha-
sis, and in some families, neurodevelopmental ceous deposits or kidney stones. In fact, topha-
deficits have been documented [77–84]. Like ceous gout has been described in an 8-year-old
hypoxanthine-guanine phosphoribosyltransferase boy with this abnormality [81]. In the absence of
deficiency, phosphoribosylpyrophosphate syn- neurological signs, it is difficult if not impossible
thetase superactivity is inherited as an X-linked to differentiate this enzyme abnormality from
recessive trait [85, 86]. Even though phosphori- partial hypoxanthine-guanine phosphoribosyl-
bosylpyrophosphate synthetase superactivity is transferase deficiency.
inherited in this manner, heterozygous female
carriers of the mutation have been reported with
gout and sensineural deafness [81–83, 87]. There Idiopathic Gout
are two somewhat different clinical expressions
of this enzyme abnormality. One phenotype is There are a small number of patients with primary
characterized by an early age of onset in child- gout who are overproducers of uric acid but have
hood, gross overproduction of uric acid (1.0–2.4 no identifiable genetic error in purine metabolism.
Overproduction of Uric Acid 107

There is the possibility that such patients may convulsions, and coma [93, 94]. The laboratory
have a mutation in the rate-limiting enzyme of parameters of this disorder include hypoglyce-
de novo purine biosynthesis, phosphoribosylpy- mia, ketosis, and acidosis. In addition to elevated
rophosphate amidotransferase. Alterations in the blood levels of lactate, ketones, and alanine,
kinetic properties of this enzyme could also lead serum uric acid levels are also increased. This
to excessive rate of purine synthesis. Such abnor- hyperuricemia is reflected in the appearance of
malities have been described in Ehrlich ascites excess uric acid in the urine [95–99]. Patients
cell lines and bacterial species due to a decrease with fructose-1,6-diphosphatase deficiency, like
in the sensitivity of these cells to inhibition of hereditary fructose intolerance, manifest an
purine nucleotides [88, 89]. No such mutants increase in serum uric acid levels after a fructose
have been identified to date in the human, but load [100]. This enzyme deficiency must be dif-
mutations in the human amidotransferase may be ferentiated from other causes of lactic acidosis
characterized in the future. such as pyruvate carboxylase deficiency, pyru-
vate dehydrogenase deficiency, and phosphoenol
pyruvate carboxykinase deficiency.
Secondary Hyperuricemia and Gout Decreased ATP synthesis is observed in two
due to Overproduction of Purines conditions: tissue hypoxia and metabolic myopa-
thies. The latter disorders are discussed subse-
As noted previously, secondary hyperuricemia quently; tissue hypoxia on the basis of ischemia
with increased urate production can be caused by or hypoxemia may result in massively elevated
four different mechanisms: decreased synthesis serum uric acid levels [90, 101–104].
of ATP, increased ATP degradation, increased Hyperuricemia and elevated oxypurine levels
nucleic acid turnover, or excessive ingestion of have been observed in cardiogenic shock, revers-
purines. When the supply of inorganic phospho- ible cardiac arrest, perinatal distress, and pulmo-
rus, oxygen, and nicotinamide adenine dinucle- nary disorders [101, 105–117]. Gout has been
otide is limited, the synthesis of ATP in the reported in rare cases of emphysema and chronic
mitochondria may be decreased. Further, if obstructive pulmonary disease [118, 119]. The
energy sources such as glucose or fatty acids are coexistence of these two disorders probably rep-
restricted, ATP synthesis may also be diminished. resents the coincidental occurrence of two com-
When the need for ATP exceeds the capacity of mon disorders with the gout being aggravated by
the cell to synthesize ATP, then ATP degradation an increased production of urate.
occurs and adenosine diphosphate and adenosine The other group of disorders associated with
monophosphate are rapidly converted to inosine, impairment of ATP synthesis is the metabolic
hypoxanthine, xanthine, and uric acid [90–92]. myopathies. In these conditions, ATP is utilized
Some of the disorders associated with ATP in muscular activity, but the source of carbohy-
degradation are also associated with gout, while drates or fatty acids is limited by deficiencies in
others only produce hyperuricemia. Glycogen the generation of substrates for ATP synthesis.
storage diseases and hereditary fructose intoler- As will be discussed subsequently, this group of
ance have been documented as causes of both diseases causing myogenic hyperuricemia and, in
gout and hyperuricemia. These disorders are dis- some instances, gout represents disorders of both
cussed subsequently. Ethanol ingestion and fruc- glycogen and fatty acid metabolism. The limited
tose-induced hyperuricemia are discussed with capacity of these deficient individuals to provide
the drug-induced causes of hyperuricemia even substrates for ATP synthesis leads to an acceler-
though the mechanism for the induction of hype- ated degradation of adenine nucleotides to inos-
ruricemia in these cases is via ATP degradation. ine, hypoxanthine, xanthine, and uric acid.
Fructose-1,6-bisphosphatase deficiency is an Increased nucleic acid turnover resulting in
autosomal recessive trait observed primarily in hyperuricemia occurs in conditions where exces-
newborns that present with hyperventilation, sive nucleic acid synthesis is required or when
108 5 Clinical Aspects of Gout and Associated Disease States

there is a release of excessive quantities of nucleic The other rare enzyme defect associated with
acids due to the lysis of cells. Excessive nucleic a single case report of gout is the accelerated
acid synthesis is observed most often in hemato- nucleotide degradation observed in a patient with
logical malignancies and results in altered purine liver adenosine monophosphate deaminase
nucleotide concentrations. Excessive nucleic acid deficiency [129]. The conversion of adenosine
synthesis in support of malignant cell growth monophosphate to inosine monophosphate by the
leads a decrease in cellular nucleotide content enzyme adenosine deaminase is the rate-limiting
with the resultant decrease in the feedback inhi- step for the degradation of adenine nucleotides,
bition of de novo purine biosynthesis and an and in this single case of gout, AMP deaminase
increase in de novo purine synthesis. Treatment was determined to be insensitive to its physiolog-
regimens for hematological malignancies or other ical inhibition by inorganic phosphorus and
types of cancer can also cause cell destruction guanosine triphosphate. Thus, AMP deaminase is
with the resultant release of large quantities of released from inhibition, and increased quantities
nucleotides. These excess nucleotides are subse- of adenosine monophosphate are converted to
quently converted to uric acid. There are a variety inosinic acid and subsequently to uric acid. Since
of acquired disorders that have been reported in liver biopsies are rarely, if ever, analyzed in
association with gout and altered nucleotide turn- patients with gout, the frequency of this enzyme
over including hemolytic anemias, sickle cell dis- abnormality in gouty subjects is unknown. This
ease, multiple myeloma, polycythemia, leukemia, enzyme and its alteration in purine metabolism
and myeloproliferative disorders [120–128]. are discussed in greater detail in the section deal-
Excessive ingestion of purines is an unusual ing with uric acid metabolism in the human.
cause of hyperuricemia, but such dietary excesses
may be observed in individuals with unusual eat-
ing habits. In these individuals, dietary indiscre- Glycogen Storage Diseases (GSD)
tions may either cause hyperuricemia by itself or
contribute to the hyperuricemia caused by other Although glycogen storage diseases are usually
mechanisms. Such dietary indiscretions usually recognized in childhood and treatment instituted
aggravate other causes of hyperuricemia and are at that time, most patients survive into adulthood,
not the sole cause of this metabolic change. and some subsets of this disorder are first recog-
Finally, there are two unusual and rare causes nized in adulthood [134–137]. Hyperuricemia
of gout that have been reported in the literature and/or gouty arthritis are frequently observed as
[129–132]. Both these enzyme abnormalities complications of glycogen storage disease types
lead to the overproduction of uric acid. In myo- I, III, V and VII, and their recognition necessi-
adenylate deaminase deficiency, patients experi- tates a careful analysis of the symptoms and signs
ence muscle fatigue and myalgias after exercise. of these adult presentations if the true underly-
The defect in this enzyme results in a deficiency ing cause of this purine dysfunction is to be
of ATP synthesis with subsequent ATP degrada- identified.
tion to uric acid when exercise places increased In the adult, type I glycogen storage disease
demands on ATP utilization that cannot be (glucose-6-phosphatase deficiency) is often
achieved in those with this enzyme deficiency. manifested by musculoskeletal disease (gout,
Since more than 200 patients have been described short stature, osteopenia), hepatic and renal dys-
with this muscle enzyme deficiency and roughly functions (altered liver function, enlarged liver,
2 % of muscle biopsy specimens submitted for hepatic adenomas, microalbuminuria, renal cal-
analysis have this enzyme deficiency, the single culi, and nephrocalcinosis), anemia, and hyper-
case of gout reported in the literature probably lipidemia. If type Ib GSD (glucose-6-phosphate
represents the occurrence of two common disor- translocase deficiency) is the underlying prob-
ders rather than a specific genetic error causing lem, then neutropenia and recurrent bacterial
gout [133]. infections are also observed in conjunction with
Overproduction of Uric Acid 109

the findings associated with classical type I gly- dysfunctions, serum triglycerides and cholesterol
cogen storage disease [138]. Acute and chronic levels are elevated due to the increased rate of
tophaceous gout result from the increased degra- hepatic triglyceride synthesis and decreased
dation of adenine nucleotides to uric acid peripheral lipolysis [149]. These lipid abnormali-
(Fig. 5.1) and the increased lactate production ties give rise to pancreatitis and atheromatous
that inhibits uric acid excretion by the kidney. blood vessel disease.
These two disorders (type I and 1b) are a major In those patients with type Ib GSD, neutrope-
cause of morbidity from gout especially if treat- nia and frequent bacterial infections occur because
ment of these disorders has been ineffective for of impaired neutrophil function [150, 151]. These
any reason. Associated with this articular dis- recurrent bacterial infections require appropriate
ease is an increased incidence of osteopenia antibacterial treatment and attention to personal
including bone fractures and osteonecrosis hygiene to prevent further infections including
[134]. The pathogenesis of these bone disorders the prevention of unnecessary exposures to other
is incompletely understood, and they have not infected individuals.
been evaluated by modern quantitative method- Recognition of the underlying disease process
ologies [134, 139] and the institution of appropriate treatment
Renal problems are frequently found in asso- (allopurinol and high fluid intake) are likely to
ciation with these musculoskeletal disorders prevent the morbidity related to renal calculi,
including calculi that are usually composed of nephrocalcinosis, and recurrent gouty attacks.
calcium oxalate although stones formed from The foregoing descriptions of symptoms and
uric acid may also be observed [140–143]. At the signs unrelated to gouty arthritis can serve to alert
present time, no biochemical rationale has been the clinician to an underlying disease. Although
proposed that links oxalate metabolism with the patient may be aware of the underlying GSD,
defective glycogen metabolism. Nephrocalcinosis, he or she may not make the connection between
glomerulosclerosis, and recurrent pyelonephritis nephrolithiasis, gout, and glycogen storage dis-
in the absence of structural abnormalities of the ease. For this reason, a detailed and careful history
urinary tract occur in more than half the patients is essential for the identification of the complicat-
with GSD type I. Gross proteinuria (>1,000 mg/ ing disease process.
day) and microalbuminuria are also often pres- Type III GSD (debranching enzyme deficiency/
ent. These abnormalities of the kidney may or amylo-1,6-glucosidase deficiency) is clinically
may not be accompanied by a diminished creati- similar to type I GSD in its childhood presenta-
nine clearance and hypertension. Allopurinol tion with hepatomegaly, hyperlipidemia, hypogly-
treatment is recommended to impair the contri- cemia, and stunted growth as the most consistent
butions of hyperuricemia to the progression of features. In adults, liver disease usually resolves
the renal disease. Renal tubular dysfunctions and hepatomegaly may remain as a feature, but it
may also be observed in patients with type 1 gly- is not a common finding. Weakness and muscle
cogen storage disease including renal tubular wasting are the most predominant clinical findings
acidosis, hypercalciuria, and hypocitraturia in adults with the disorder. Muscle atrophy is often
[143]. A Fanconi type syndrome with aminoaci- especially prominent in the legs and small mus-
duria, proteinuria (low molecular weight), phos- cles of the hands, and electromyographic studies
phaturia, and bicarbonaturia has also been show widespread evidence of myopathic changes
described [144, 145]. [152–158]. Electrocardiograms may show ven-
Hepatomegaly is a uniform finding in patients tricular hypertrophy [159–161]. Hepatic disease,
with type I GSD, and many of these patients may as noted previously, is not a prominent feature
also acquire multiple hepatic adenomas [146– of the adult disease although hepatic fibrosis is
148]. These adenomas may rarely undergo malig- often present.
nant degeneration, and for this reason are Laboratory parameters may include moder-
important to identify. In addition to these organ ately increased levels of transaminases, elevated
110 5 Clinical Aspects of Gout and Associated Disease States

serum triglyceride and cholesterol levels, and not [170, 171]. As a result of the muscle pain and
infrequently increased serum uric acid levels elevated creatine phosphokinase levels, this myo-
[152, 153, 162]. Serum creatine phosphokinase pathic disorder has been confused with polymyo-
levels may or may not be elevated as a manifesta- sitis [172]. Although not a common cause of
tion of the underlying muscle disease. A review hyperuricemia and gout, excessive adenine nucle-
of the literature does not reveal any reported otide degradation in muscle does occur in type V
patients who developed gout despite their ele- GSD when intense or sustained exercise is under-
vated serum urate levels. In many patients, lactate taken, and such occurrences suggest the need for
and urate concentrations are within the normal the evaluation of those suspected of this disorder
range and may account for the rarity of gouty with exercise testing [163, 170].
arthritis in this type of GSD. When type III GSD Fewer than 50 cases of type VII GSD
patients exercise, an increase in the plasma con- (Tarui’s disease or muscle phosphofructokinase
centrations of hypoxanthine and uric acid occurs deficiency) have been reported in the literature;
as compared to their concentrations in the non- however, this myopathy is frequently associ-
exercising patient [163]. Thus, hyperuricemia ated with gouty arthritis and/or hyperuricemia
may be especially prominent after exercise. Since [173–177]. Although this disorder primarily
this disorder may be selective in its expression presents as a myopathy with fatigue and mus-
involving liver and/or muscle, diagnostic studies cle cramps associated with exercise along with
must include evaluations of the debranching myoglobinuria, it may have significant variation
enzyme in both muscle and liver. This type of in its clinical characteristics. Late onset of this
GSD is often found in non-Ashkenazic Jews myopathy may only have muscle weakness and
from North Africa, and patients from this ethnic the complete absence of cramps and myoglobi-
background deserve special attention. The skel- nuria [174, 178, 179]. An infantile form of this
etal manifestations may lead to confusion with disease has also been reported which is fatal and
Charcot-Marie-Tooth disease because of the is of no concern to the subject matter discussed
wasting of the small muscles of the hands and herein [180, 181]. Tarui’s disease may be difficult
feet as well as the peripheral limbs seen in both to distinguish from type V GSD since they both
diseases. present with myopathic symptoms associated
Type V GSD (muscle phosphorylase with exercise. Some aspects of the clinical pre-
deficiency) is clearly associated with hyperurice- sentation may assist in segregating these two
mia and gouty arthritis although in some cases, disorders. Tarui’s disease is often associated with
the gout is thought to be coincidental [164, 165]. more severe muscle symptoms than type V GSD,
Type V GSD, although present from birth, mani- and the former patients may also complain of
fests a wide range of heterogeneity with respect nausea and vomiting accompanying acute epi-
to its clinical findings, yet the enzyme defect per sodes of muscle cramps. Hemolytic anemia with
se appears not to be linked with its varying clini- hyperbilibubinemia, reticulocytosis, and reduced
cal presentations. The classical presentation is erythrocyte 2,3-biphosphoglycerate levels is seen
one of exercise-induced muscle cramps with in type VII GSD but not in type V GSD [182,
pink- or burgundy-colored urine as a result of 183]. In some patients with type VII GSD, hemo-
rhabdomyolysis and myoglobinuria [166, 167]. lysis may occur in the absence of any muscle
As noted, the clinical dysfunctions may occur in symptoms. The degree of hemolysis in type VII
childhood, but more commonly, they manifest GSD is mild, and leukopenia may accompany the
themselves at age 20 or 30 and sometimes not hemolysis if the patient has splenomegaly. The
until late in life [168, 169]. Laboratory findings mechanism for this hereditary nonspherocytic
include elevated levels of creatine phosphokinase hemolytic anemia is not completely resolved, but
that can be significantly increased further with it has been suggested that the accumulation of
exercise. Exercise also increases the levels of red blood cell intermediate metabolites may alter
blood ammonia, hypoxanthine, and uric acid the erythrocyte membrane. Thus, if hemolytic
Overproduction of Uric Acid 111

anemia is present in type VII GSD, this compo- phosphoribosylamine, the first step in de novo
nent may differentiate this disorder from type V purine biosynthesis.
GSD in which no hemolysis has been recorded in There are a group of heritable myopathies that
affected patients. must be differentiated from types V and VII
If one considers type III, V, and VII glyco- GSDs since they are characterized in their classi-
gen storage diseases, the latter disorder (type cal presentations by exercise intolerance, muscle
VII GSD) induces the most aggressive reduc- cramps, and myoglobinuria [166, 187]. These
tion in adenine nucleotides with its resultant metabolic myopathies can be divided into those
rise in serum uric acid levels. The magnitude of affecting glycogen metabolism, lipid metabo-
the hyperuricemia is much greater than that lism, or mitochondrial metabolism. Since purine
observed in types III and V GSD, and this may degradation, hyperuricemia, and/or gout have
account for the more frequent association of only been recorded in some of these disorders, it
gout with type VII GSD. Meals high in carbo- is unclear how common alterations in serum uric
hydrate are known to aggravate the exercise- acid levels and gouty arthritis may be in these
induced changes in muscle and their associated rare myopathies. For this reason, brief but not
symptoms [184]. The glucose provided by high detailed clinical descriptions of these disorders
carbohydrate meals cannot be utilized due to are included.
the deficiency of phosphofructokinase activity, Hyperuricemia or gout has been reported in
and this results in a decrease in blood fatty some patients with muscle phosphoglycerate
acids, a key source of muscle fuel. In contrast to mutase deficiency and myoadenylate deaminase
this alteration in glucose metabolism, patients deficiency, but how common these associations
with type V GSD can metabolize glucose, and are is unknown [131, 188]. Documentation of
their exercise intolerance improves with high excessive purine degradation in muscle has been
carboyhydrate diets. Muscle biopsy in type VII reported in these disorders so that it is likely that
GSD shows a PAS-positive abnormal polysac- hyperuricemia and perhaps gout would accom-
charide [174, 175]. pany these diseases [132, 189].
The diagnosis of type VII GSD requires the As for the glycolytic metabolic abnormalities
demonstration of the enzymatic defect in muscle, that give rise to exercise intolerance, recurrent
but exercise-induced muscle symptoms, hyperu- myoglobinuria, myalgias, crampy muscle pain,
ricemia, and/or gout should suggest to the astute limb weakness with elevated creatine phosphoki-
clinician that the underlying disease may be one nase levels, and rarely acute renal failure, their
of the glycogen storage diseases affecting mus- clinical findings most often appear in the early
cle. Although adenine nucleotide catabolism and stages of exercise or associated with intense
the formation of inosine monophosphate, an physical exertion. Thus, the muscle symptoma-
immediate precursor of uric acid, probably repre- tology of types V, VI, VII, VIII, IX, X, and XI
sent the primary source of the increased serum glycogen storage diseases occurs when the energy
urate levels, two other aspects of the disease may source for muscle is primarily dependent on car-
contribute to the hyperuricemia. The low-grade bohydrates. At times, these patients may give a
hemolysis and its accompanying turnover of red history of a second burst of energy after their ini-
blood cell nucleic acids may, when present, result tial symptoms that probably relates to a shift in
in increments in uric acid levels. In addition, the the muscle energy source from carbohydrates to
accumulation of glucose-6-phosphate and fruc- fatty acids.
tose-6-phosphate resulting from the inability to The clinical parameters of type V and VII
convert fructose-6-phosphate to fructose-1,6-bis- GSD have already been described, and only a
phosphate may stimulate the pentose phosphate brief description of the other glycolytic disorders,
shunt and generation of ribose-5-phosphate and the fatty oxidation abnormalities, and the other
phosphoribosylpyrophosphate [185, 186]. The heritable disorders of exercise intolerance and
latter compound is an immediate precursor of myoglobinuria are summarized here since the
112 5 Clinical Aspects of Gout and Associated Disease States

extent of their association with hyperuricemia Table 5.5 Heritable disorders of exercise intolerance
and gout is unknown. Muscle-specific phospho- and myoglobinuria
rylase kinase deficiency is a rare autosomal reces- Glycolytic abnormalities
sive trait presenting with muscle cramps and Phosphorylase deficiency (type V GSD)
myoglobinuria associated with exercise or mus- Phosphofructokinase deficiency (type VII GSD)
cle weakness and atrophy [190–198]. Like other Phosphoglycerate kinase deficiency (type IX GSD)
muscle diseases, the presentation of this illness is Phosphoglycerate mutase deficiency (type X GSD)
Lactate dehydrogenase deficiency (type XI GSD)
heterogeneous in that some patients may not have
Muscle-specific phosphorylase kinase deficiency
muscle cramps, may have muscle cramps without
Fatty acid oxidation abnormalities
myoglobinuria, or may have muscle weakness
Carnitine palmitoyltransferase II deficiency
alone. Muscle biopsy shows a vacuolar myopathy Long-chain acyl-CoA dehydrogenase deficiency
with glycogen accumulation [190, 193, 198]. Very-long-chain acyl-CoA dehydrogenase deficiency
Ischemic exercise shows an elevation in blood Short-chain L-3-hydroxyacyl-CoA dehydrogenase
ammonia but no abnormality in lactate. Since deficiency
there are different types of phosphorylase b Miscellaneous
kinase deficiency affecting the liver only, muscle Glucose-6-phosphate dehydrogenase deficiency
and liver, muscle only, and heart muscle, it is Myoadenylate deaminase deficiency
imperative that diagnostic studies include enzyme Duchenne and Becker muscular dystrophy
analysis of these various tissues [197]. Enzyme Familial recurrent myoglobinuria
analyses of erythrocytes and/or leukocytes alone
may miss the enzyme deficit if muscle tissue is
not analyzed directly. expression of this heritable disorder is heteroge-
An X-linked form of glycogen storage disease neous [201–206]. From the standpoint of clinical
has been reported with muscle involvement and heterogeneity, patients may present with hemo-
gouty arthritis [199]. The clinical presentation of lytic anemia alone, myopathy alone, no central
this disorder in males is one of muscle weakness nervous system changes, or without any symp-
in childhood, enlargement of the liver, growth toms at all. Phosphoglycerate kinase deficiency
retardation, and a delay in sexual maturation. may mimic types V or VII GSD in its presenta-
Hyperuricemia and gouty arthritis have been tion. Therefore, an X-linked recessive pattern of
observed in adults with this disorder. Although inheritance by pedigree analysis may provide a
the precise enzyme deficit remains unknown, significant clue to the underlying cause of myo-
the authors excluded deficiencies in glucose-6- pathic symptoms. Ischemic forearm exercise test-
phosphatase, debrancher enzyme, phosphory- ing shows no elevation of venous lactate levels.
lase, or phosphorylase kinase as the cause for the Phosphoglycerate kinase activity must be mea-
disorder. sured in muscle to make a definitive diagnosis
Phosphoglycerate kinase deficiency is an (Table 5.5).
X-linked recessive trait usually presenting in its Muscle phosphoglycerate mutase deficiency
most complete form as a nonspherocytic hemo- is a rare autosomal recessive disorder associated
lytic anemia, mental retardation, seizures, muscle with exercise intolerance, muscle cramps with
cramps with exercise, myoglobinuria, and renal exercise, myoglobinuria, and elevated creatine
failure. It is possible that the X-linked trait with phosphokinase levels [207–212]. Muscle biop-
muscle involvement and gout described by sies from such patients show increased PAS stain-
Keating and his colleagues represented a case of ing and increased muscle glycogen content. For
phosphoglycerate kinase deficiency [199]. The purposes of classification, this disorder has been
latter disorder has been termed type IX glycogen called type X GSD. Studies of anaerobic glycoly-
storage disease, and electron microscopy of sis have shown decreased lactate production in
muscle specimens shows elevated muscle glyco- this disorder, and in one middle-aged male with a
gen content [200]. The clinical and molecular deficiency of this enzyme, gouty tophi have been
Overproduction of Uric Acid 113

observed [213]. It is not known whether the pres- fasting, emotional stress, cold exposure or an
ence of gout in association with this heritable infectious process may trigger symptoms in those
myopathy is coincidental or the result of exercise patients with defective fatty acid oxidation.
intolerance and hyperuricemia in a patient since The principal fatty acid oxidation defect is the
adolescence. The enzyme, phosphoglycerate hereditary deficiency of carnitine palmitoyltrans-
mutase, is a dimer composed of a slow-migrating ferase II. The classical form of this disorder pri-
muscle isoenzyme and a fast-migrating brain marily affects adult males even though there is a
isoenzyme. In enzyme-deficient patients, assays severe, fatal infantile form [223–226]. This
for enzyme activity in muscle may detect small enzyme deficiency is inherited as an autosomal
quantities of the brain isoenzyme and not the pre- recessive trait, and the reason for the preponder-
dominant muscle isoenzyme. ance of males with the disorder remains to be
Lactate dehydrogenase-A deficiency is the determined. Renal failure secondary to myo-
final error in glycolysis to be associated with globinuria is not an unusual finding in this dis-
exertional muscle pain and myoglobinuria. It has ease and coupled with the fact that this enzyme
been classified as glycogen storage disease type deficiency is a common metabolic cause of recur-
XI and is accompanied by an increase in serum rent myoglobinuria makes its identification
creatine phosphokinase levels [214–219]. This imperative [227–235]. Although permanent mus-
enzyme deficiency is inherited as an autosomal cle weakness is rarely observed, it has been
recessive trait, and in addition to its myopathic reported with elevated creatine phosphokinase
features, erythematosquamous lesions of the skin levels and myopathic changes by electromyogra-
on the extensor surfaces of the limbs may be phy [236]. When patients with this disorder are
observed [220–222]. In contrast to psoriasis, subjected to ischemic forearm testing in the fast-
these skin lesions are often worse in the summer. ing state, plasma purine levels are determined to
As in other metabolic myopathies, acute renal be elevated. These findings clearly document the
failure secondary to rhabdomyolysis is the most increased ATP degradation and uric acid produc-
serious complication of this heritable enzyme tion associated with heritable palmitoyltrans-
deficiency. ferase deficiency [189]. In patients with adult
onset disease, tissue fibroblasts and leukocytes
may express the enzyme deficiency, and this
Fatty Acid Oxidation Deficiencies avoids the need for a muscle biopsy. The latter
procedure, if necessary for the demonstration of
Heritable defects in fatty acid oxidation are the enzyme deficiency, may show an increase in
clearly associated with hyperuricemia, but so far lipid droplets by oil red O staining. As noted,
as can be determined, no cases of gouty arthritis ATP degradation products and hyperuricemia are
have been documented in the literature. All the present in this disorder, but no reports of gout
disorders discussed herein present with exercise have been documented in the literature.
intolerance and recurrent myoglobinuria. In most As can be seen from the foregoing discussion,
instances, the clinical symptoms of myalgias, a wide variety of genetic diseases are associated
cramping, or stiffness of muscles appear after with gout and urate overproduction. In addition,
prolonged exercise. The musculoskeletal symp- there are a number of disorders in which ATP
toms are often accompanied by pigmenturia and metabolism is altered causing hyperuricemia and,
may rarely cause acute renal failure resulting in some, gout (Table 5.3). These so-called over-
from rhabdomyolysis. During episodes of exer- producer types of hyperuricemia and gout account
cise intolerance, creatine phosphokinase levels for about 5–10 % of the total cases of acute gout,
are usually elevated, but they return to normal yet careful clinical analyses of patients with gout
levels on recovery. In muscles, carbohydrates may provide clues to an underlying heritable dis-
serve as the main energy source during the initial ease and prevent the morbidity related to unde-
phases of exercise, whereas 1–4 h of exercise, tected family members.
114 5 Clinical Aspects of Gout and Associated Disease States

Decreased Uric Acid Excretion Primary Underexcretor Gout

Introduction Primary underexcretor gout is defined as a patient


with no identifiable underlying acquired disorder,
In addition to the abnormalities in the production no pharmacologic treatment with agents known
of uric acid that cause hyperuricemia or gout, to cause hyperuricemia or gout, underexcretion
alterations in the elimination of uric acid by the of urinary uric acid (<250–300 mg/24 h on a
kidney may also cause hyperuricemia and gout. purine-free diet), and a lower uric acid clearance
In general, urinary uric acid accounts for about rate than normal nongouty subjects [248–254].
70 % of the uric acid eliminated in a day, whereas The underexcretion of uric acid by gouty subjects
the remainder is eliminated by intestinal uricoly- was further clarified when the urinary uric acid
sis and other minor routes such as tears and per- excretion rate was plotted as a function of the
spiration [237]. Theoretically, a decrease in plasma urate concentration [249, 253, 255, 256].
either intestinal uricolysis or renal excretion In these experiments, the values for gouty sub-
could cause hyperuricemia, but in fact, altera- jects were shifted to the right when compared
tions in intestinal uricolysis do not cause hyperu- with nongouty subjects. The conclusion from
ricemia [238]. such changes is that gouty subjects require serum
To characterize the errors in renal excretion uric acid concentrations 1.7 mg/dl higher than
leading to hyperuricemia or gout, the renal han- nongouty subjects to achieve uric acid excretion
dling of uric acid must be understood. The four- rates comparable to nongouty subjects. The same
component system of uric acid handling by the alteration is found when the clearanceurate/clear-
kidney is the presently accepted model [239– anceinulin is evaluated in gouty and nongouty sub-
242]. At the glomerulus, uric acid is completely jects [248, 253, 256]. When the plasma urate
filtered, and it is almost completely reabsorbed concentration is increased by exogenous RNA or
by the proximal tubules. Proximal renal tubular by urate infusions and the clearance of urate is
urate reabsorption occurs by an active transport evaluated, gouty subjects require a higher plasma
process [243, 244]. Subsequently, about 50 % of urate concentration to achieve a clearance identi-
the reabsorbed urate is secreted by the proximal cal to nongouty subjects [251, 257]. Even though
tubules, and the location of urate secretion the mechanism to account for these differences in
remains to be completely defined in the human. the handling of uric acid by the kidney remain
Then, about 40 % of the secreted urate is reab- unknown, primary underexcretor gout most likely
sorbed by a postsecretory mechanism. Again, the represents an abnormality (perhaps inherited) in
sites for urate secretion and presecretory reab- the renal tubular transport of uric acid.
sorption have not been localized in the human.
Evidence can be cited that different mechanisms
account for presecretory and postsecretory reab- Secondary Hyperuricemia and Gout due
sorption [245, 246]. Despite the role that increased to the Underexcretion of Uric Acid
purine synthesis and degradation may play in the
causation of hyperuricemia and gout, the most In addition to patients with primary underexcre-
common cause of these disorders is a decrease in tor hyperuricemia and gout, there are a number of
the excretion of uric acid. This may occur as a clinical settings in which renal function changes
primary phenomenon with no underlying acquired impair the excretion of uric acid as a result of the
disease or with a pharmacologic agent as the effects of an underlying disease or the use of a
cause or as a secondary phenomenon where the medication. For example, the reduced nephron
development of hyperuricemia and gout occurs population in chronic renal disease and the decline
as a result of another disease. Primary gout or pri- of glomerular filtration rates cause a reduction
mary underexcretor gout is observed in about 85 in the fractional excretion of urate [258]. Thus,
% of patients with gout [247]. the hyperuricemia of renal failure results from
Decreased Uric Acid Excretion 115

a decreased glomerular filtration rate. Diuretic- of uric acid at such low levels are considered.
induced hyperuricemia and gout are primarily Thus, clinical settings where the quantity of uric
caused by increased salt and water loss with a acid presented to the kidney is excessive and
resultant volume contraction and increased urate exceeds the capacity of renal tubules, renal pelvis
reabsorption by the proximal tubules [259–261]. or ureters to handle this increased urate load, uric
This disorder is discussed further in the section acid may precipitate. The kidney is also the site
“Drug-induced hyperuricemia and gout.” Finally, of uric acid deposits in some diseases associated
decreased renal tubular secretion of uric acid and directly with gout and in other diseases in which
secondary hyperuricemia occur when various uric acid deposits occur as the result of massive
organic acids compete with urate tubular secre- tissue destruction and urate production that
tory sites and inhibit uric acid secretion. Such exceeds the kidney’s capacity to excrete it. Thus,
secondary renal hyperuricemia has been docu- urate nephropathy is defined as the deposition of
mented in association with elevated blood lev- microtophi within the kidney parenchyma and is
els of lactate, ß-hydroxybutyrate, acetoacetate, most frequently observed in conditions where
branched chain keto acids, and with low serum there is marked overproduction of uric acid like
concentrations of salicylates [262–265]. the Lesch-Nyhan syndrome.
Although older studies have documented urate
nephropathy as a cause of death in gout, more
Urate Nephropathy recent studies have questioned the common
occurrence of this pathologic abnormality [266–
Even though it is clear that the skeletal system is 268]. It is not appropriate to maintain the concept
the major pathologic recipient of abnormal purine that the development of irreversible kidney dys-
metabolism and gout, the classic studies of function is the primary cause of death in 10–25 %
Talbott and Terplan have also established that the of patients with gout as the older literature has
kidney is also a target organ in gout [266]. If one cited [266, 269, 270]. The change in the fre-
considers human purine metabolism, the kidney quency of this end-stage renal lesion and the
is the primary excretory route for the uric acid cause of death in gout are most likely related to
generated from biosynthetic and degradative an earlier recognition of those entities causing
pathways as well as from the catabolism of urate deposition in the kidney and the use of uri-
ingested purines and nucleic acids. About two- cosuric and hypouricemic agents to protect the
thirds of the daily production of uric acid is kidney from the formation of renal urate depos-
filtered at the glomerulus and subsequently sub- its. Further, the early recognition and effective
jected to transtubular reabsorption and secretion. treatment of coexistent disorders such as hyper-
A relatively small amount is then eliminated by tension, ischemic heart disease, preexistent renal
the kidney. The most recent models of the han- disease, and rarely chronic lead exposure have
dling of uric acid by the kidney indicate that four served to decrease the incidence of renal disease
stages are involved in the excretion of urate: [268, 271–274]. Several population studies have
glomerular filtration, tubule reabsorption, tubular confirmed the fact that hyperuricemia has only a
secretion, and postsecretory reabsorption modest effect on the deterioration of renal func-
(Fig. 5.1). To emphasize this key renal role in the tion. In the Framingham study, only 2 % of the
excretion of urate, the intestinal tract excretes 240 patients evaluated with serum uric acid levels
only about one-third of the daily production of greater than 7 mg/dl had clinical evidence of
uric acid, and the kidney excretes the remaining renal disease to which hyperuricemia might have
two-thirds. Uric acid in the intestine is degraded contributed [275]. A subsequent study of 1,356
by microbial uricase so that very little uric acid, men aged 60–69 with asymptomatic hyperurice-
per se, is actually eliminated by the bowel. The mia or gout showed relatively modest elevations
role of the kidney becomes even more significant in their serum creatinine over a 10-year period,
if the low pH of the urine and the poor solubility and no deaths could be attributed to renal disease
116 5 Clinical Aspects of Gout and Associated Disease States

caused by hyperuricemia [276, 277]. Finally, the for such crystalline deposits are the papillae,
Normative Aging Study of 1,658 male veterans renal pyramids, and the medullary interstitium.
with asymptomatic hyperuricemia evaluated over Pathological examination may also show glom-
a 15-year period showed less than 1 % of the par- eruli hyalinization, vascular changes of hyperten-
ticipants to have serum creatinine levels of 2 mg/ sion, interstitial fibrosis, tubular atrophy, and
dl or more [278]. On the basis of these studies, other nonspecific changes. To confirm the chemi-
hyperuricemia per se appears in many clinical cal structure of the renal crystalline deposits,
settings to have little or no deleterious effect on X-ray diffraction analyses of kidneys from gouty
the kidney. patients have been examined [282]. Monosodium
Despite these findings supporting the rarity of urate monohydrate crystals were detected in
kidney-related deaths in those with hyperurice- patients with gouty arthritis, and in addition,
mia, urate nephropathy does occur in certain some gouty patients showed calcium oxalate
clinical settings associated with gout, and there monohydrate crystals. The latter finding again
are changes in kidney function and pathology demonstrates the poorly understood linkage
that occur in these situations. Intermittent or per- between uric acid metabolism and calcium
sistent albuminuria may occur in 20–40 % of oxalate crystals and calculi. In contrast to these
patients with gout, and glomerular filtration rates findings, evaluation of patients at autopsy with
are often decreased [248, 279, 280]. Further, acute leukemia and increased nucleic acid turn-
there is often loss of the capacity to generate a over, the crystals were composed of uric acid.
maximally concentrated urine. These findings Some investigators have indicated that the glom-
may be present even when hypertensive patients erular changes are distinctive for gout and differ-
are excluded from consideration. Of course, these ent from those of nephrosclerosis and diabetic
laboratory abnormalities are nonspecific and glomerulosclerosis, whereas others do not agree
offer no clue to their underlying causation. The with this conclusion [281, 283, 284]. Despite the
gross examination of kidneys from patients with identification of uric acid crystals in pathological
gout may show small white specks with a granu- specimens from the kidney, several studies docu-
lar feel at the corticomedullary junction, and the ment the difficulties in making the diagnosis of
renal pelvis may contain uric acid calculi. If gout based on postmortem findings [285, 286]. In
pyelonephritis has been superimposed, the kid- one postmortem study of individuals succumbing
neys may be small and contracted. The classic to renal insufficiency, only four patients had a
pathologic finding in gout is the tophus that is history of acute gouty arthritis, whereas 17
represented by a monosodium urate deposit sur- patients manifested renal medullary microtophi
rounded by chronic inflammatory cells (lympho- [285]. In another postmortem study, 26 % of the
cytes, macrophages, and plasma cells) and foreign individuals had medullary microtophi with no
body giant cells. In order to preserve urate crys- underlying cause discernable [286]. In contrast to
tals in tissues, proper fixation and staining are these studies of medullary microtophi, investiga-
necessary. Fixing tissues in absolute alcohol tions of kidney biopsies, and postmortem exami-
rather than using aqueous fixation that dissolves nations of patients with familial juvenile gouty
the crystals preserves crystalline deposits. If tis- nephropathy, a disease to be discussed subse-
sues are then viewed by polarizing microscopy, quently, only occasional crystals have been
the crystals are intensely anisotropic. With the de observed [287, 288]. These studies all support the
Galantha silver stain after alcohol fixation, urate notion that urate microtophi are not diagnostic of
crystals appear brown-black, whereas with hema- gout per se, and they raise the question as to
toxylin-eosin staining after formalin fixation, the whether chronic hyperuricemia causes significant
crystals are a deep blue color [281]. Without renal damage. As a practical matter, patients with
proper fixation or with aqueous fixation, only serum uric acid levels in excess of 10 mg/dl in
clefts may be observed where crystals have been females or 13 mg/dl in males should be carefully
deposited. The most characteristic locations evaluated and treated, if necessary, to prevent
Decreased Uric Acid Excretion 117

renal damage. It is imperative that measurements report indicates that NSAID therapy may be a
of uric acid be accompanied by laboratory evalu- risk factor for this disorder since it has the capac-
ations of renal function since serum uric acid val- ity to alter vascular tone through the impairment
ues rise in the face of impaired renal function. In of vasodilatory substances [303]. In addition to
prepubertal children, the serum uric acid may the hyperuricemia, this syndrome may be accom-
remain in the normal range despite gross over- panied by hyperphosphatemia, hyperkalemia,
production of uric acid since the renal clearance and hypocalcemia [297].
is high in children. In those patients in whom The diagnosis should be suspected in those
elevated levels of uric acid are of concern, the clinical settings known to be associated with the
inherited abnormalities in purine metabolism, syndrome, the presence of a serum uric acid level
hypoxanthine-guanine phosphoribosyltransferase in the range of 10–30 mg/dl in the adult, and olig-
deficiency, and phosphoribosylpyrophosphate uria or anuria. Urinalysis may or may not reveal
synthetase superactivity must be excluded. the presence of uric acid crystals. The ratio of
uric acid to creatinine in the urine usually exceeds
1.0, whereas in other causes of renal failure, it is
Acute Uric Acid Nephropathy less than one [304]. The management of this dis-
order requires its early recognition, the use of
Acute uric acid nephropathy is usually not asso- allopurinol prophylaxis, the judicious use of
ciated with gout due to overproduction of uric sodium bicarbonate to alkalinize the urine, and
acid, but it is frequently observed in clinical set- increase urate solubility. Fluid loading to main-
tings where treatment regimens cause massive tain a urine output in the range of 2–3 l/day and
tissue destruction, nucleic acid catabolism, and the use of loop diuretics are helpful in the treat-
the presentation of large quantities of urate to ment of this syndrome. If diuresis cannot be
the kidney. As a result, the collecting tubules, established, then hemodialysis must be initiated
renal pelvis, and ureters become obstructed with to remove the toxic metabolites.
uric acid crystals. A variety of conditions with Finally, there are four disease associated with
increased turnover of nucleic acids and uric acid gout for which the mechanism(s) of the hyperuri-
formation can result in the precipitation of uric cemia remains incompletely understood. Both
acid crystals in the collecting ducts and ureters autosomal dominant polycystic kidney disease
leading to acute obstruction of urine outflow and and medullary cystic disease have been reported
renal failure. Acute uric acid nephropathy is the in association with gout and uric acid calculi, but
result of uric acid overproduction and overex- the frequency of this association is probably quite
cretion. It is commonly observed in association low. Familial juvenile gouty nephropathy is an
with malignancies and rapid cell destruction unusual kidney disease that is seen with gout and
such as myeloproliferative disease, lymphomas, renal failure, yet urate microtophi are exceed-
leukemias, polycythemia, megaloblastic ane- ingly rare in this disease. This disorder may
mias, hemolytic anemias, epileptic seizures, and become a more prominent cause of gout as more
thalassemia [289–298]. Other disorders that physicians realize that this familial nephropathy
have been reported in association with hyperuri- may not express gout as an early manifestation of
cemic acute renal failure include metastatic the disease. Gout may be observed in family
seminoma, metastatic medulloblastoma, ductal members before the propositus with renal failure
carcinoma of breast cancer, gout, heritable expresses the articular symptoms of gout.
deficiencies of hypoxanthine-guanine phospho- Nephrogenic diabetes insipidus and some other
ribosyltransferase, and gout with markedly types of diabetes insipidus may present with
acidic urine [294, 299–302]. In malignancies, hyperuricemia, and the mechanism by which this
acute renal failure usually arises in course of occurs is thought to be through changes in the
chemotherapy or radiation if the patient is not urinary collecting system caused by the increased
hydrated and pretreated with allopurinol. One volume of urine excreted in this disorder.
118 5 Clinical Aspects of Gout and Associated Disease States

Autosomal Dominant Polycystic Kidney malignancy. Examination of the urine for casts
Disease (ADPKD) and crystals is critical to exclude the presence of
red blood cell casts and the rare occurrence of
Autosomal dominant polycystic kidney disease is glomerulonephritis as well as the presence of cal-
a relatively common heritable kidney disease that cium oxalate stones. If pain persists, is severe,
may not be apparent clinically until the fourth or and none of the above conditions are shown to
fifth decade of life [305]. Further, only about 50 exist, it may simply be the result of an enlarge-
% of the recipients of the disease progress to ment of one or more of the renal cysts. Cyst aspi-
renal failure [306]. Surprisingly, ADPKD is one ration and the use of a sclerosing agent may
of the causes of chronic renal failure that is asso- improve this clinical problem [313]. Care must
ciated with gouty arthritis [266, 307, 308]. be taken to prevent any deterioration in kidney
Furthermore, this heritable nephropathy is asso- function as a result of these painful conditions.
ciated with nephrolithiasis, and the renal calculi This makes careful evaluations of such painful
are composed of uric acid and/or calcium oxalate episodes a prerequisite to preventing the progres-
[309, 310]. The frequency of the occurrence of sion of renal failure.
polycystic kidney disease, renal calculi, and gout A comprehensive review documents the high
is not known, and kidney transplantation for frequency of urinary tract infections as a compli-
polycystic kidneys with the use of cyclosporine cation of ADPKD, especially in females [314].
to prevent organ rejection may prevent an accu- Both parenchymal and cyst infections are possi-
rate ascertainment of this association. Nonetheless, ble in this disorder, and cyst infections are often
the presence of gouty arthritis and/or uric acid more difficult to diagnose because of the absence
nephrolithiasis in a patient with impaired renal of systemic signs (fever, bacteremia, leukocyto-
function should alert the clinician to the possibil- sis, and pyuria) of infection. Cyst infection is
ity that polycystic kidney disease may be the often unilateral, unaccompanied by pyelonephri-
underlying condition. Certain clinical parameters tis or bacteriuria/bacteremia, and may only mani-
may increase the chances that polycystic kidneys fest itself by tenderness or pain on palpation over
are present. Nocturia and inability to concentrate the infected cyst. Pinpointing the offending
the urine maximally are, in some patients, early organism may also be difficult and, at times, may
signs of the disease [311, 312]. A careful abdom- require aspiration and culture of the cyst contents.
inal examination may reveal a palpable mass. Removal of any renal pelvic calculi is imperative
Pain and hematuria are also frequent clinical since infections may persist if stones are not
signs of the presence of polycystic kidneys. The removed. Infected cysts may also be imperme-
character of the pain is variable as is the cause. In able to some lipid-soluble antibiotics making the
most cases, no cause for the aching or heaviness use of nonpolar, lipid-soluble antibiotics a neces-
in the flank is identifiable, but in some cases, sity. As noted previously, calcium oxalate and
bleeding into the cyst or perinephric tissues may uric acid stones also occur in about 20 % of
precipitate complaints of a painful abdomen or patients with ADPKD and may complicate the
back. The pain is usually more severe with peri- treatment of infections [310].
nephric hemorrhages and may be stabbing in Another common complication of ADPKD is
character. Since kidney infection and renal cal- the development of hepatic cysts that occur in
culi are often components of ADPKD, pain of more than 50 % of patients, especially with
more than several days duration must be care- increasing age [315]. Women are more prone to
fully evaluated. Renal malignancy is a rare com- such cysts, especially in the post-pregnancy state
plication and should be suspected when weight or in the course of estrogen therapy [316]. These
loss, fever, and anemia are also present. Gross or facts suggest that hormone therapy such as the
microscopic hematuria may result from a variety use of birth control medications must be carefully
of causes including cyst rupture into the kidney considered in patients with ADPKD. Despite the
pelvis, renal stones, infected cysts, or rarely gross hepatic enlargement that may occur in
Decreased Uric Acid Excretion 119

polycystic kidney disease, liver function is usu- explain the absence of gout and uric acid stones
ally in the normal range except in those rare situ- in this patient group.
ations in which hepatic venous obstruction occurs In summary, the presence of flank masses by
or another hepatic disorder coexists [317, 318]. If palpation and hypertension in association with
the liver enlargement becomes a burden to the pain, hematuria, urinary tract infection, nephro-
patient as a result of pain or abdominal disten- lithiasis, or obstructive uropathy and a history of
sion, hepatic cysts can be drained percutaneously gout should alert the clinician to the possible
or partial hepatectomy performed [319]. Other existence of polycystic kidney disease. The key
gastrointestinal manifestations include pancreatic to the diagnosis in most cases is a family history
and splenic cysts, but these are infrequent com- of kidney disease and, in some cases, early death
plications. The occurrence of diverticulosis may from kidney failure. Since about 10 % of those
be a complication of ADPKD, but whether it is a undergoing chronic renal dialysis have ADPKD,
part of the kidney disease or a coincidental finding dialysis may be a starting point to identify poly-
has not been established [320, 321]. cystic kidney disease in other affected family
The most serious complications of ADPKD members. As can be seen from the complex pre-
are those involving the vascular system. sentations of ADPKD, the management and
Hypertension that most often precedes the onset ongoing care of such patients is not simple. In
of renal failure is a significant risk factor for the fact, the management of the articular disease is
progression of the renal insufficiency and its car- probably the least challenging aspect of the
diovascular alterations [322, 323]. Recognition underlying disorder.
of elevated blood pressure is most important in Since cystic disease occurs as an acquired dis-
males with genetic mutations in chromosome 16 order in those patients undergoing hemodialysis
(PKD1 – polycystic kidney disease 1) since the or peritoneal dialysis, it is important to exclude
male gender and PKD1 gene mutations increase this possibility when evaluating the incidence of
the risk of the progression to renal insufficiency ADPKD in dialysis populations [338, 339]. To
[323–325]. Other cardiac manifestations includ- differentiate the presence of simple cysts and
ing mitral and aortic valve lesions, coronary avoid false-negative results, criteria have been
artery, and abdominal aortic aneurysms have also established for the diagnosis of ADPKD by ultra-
been reported in association with ADPKD, but sonography used in conjunction with DNA test-
their presence may or may not be directly linked ing [306, 340, 341]. These criteria propose that
to the kidney disease. They may be related to the the adult over 60 years of age is required to have
hypertension or, in fact, coincidental findings at least four cysts in each kidney for the diagnosis
[326–331]. Of this group of vascular complica- of ADPKD, whereas those between the ages of
tions, cerebral aneurysms are the most serious 39 and 59 years require only two cysts per kid-
and life-threatening problems [332–335]. They ney. In those younger than 30 years of age, diag-
occur in about 5 % of patients with ADPKD. To nosis requires at least two cysts in one kidney for
avoid the disastrous consequences of aneurysmal diagnosis.
rupture, rigorous control of hypertension in all
patients and screening of patients with a family
history of cerebral aneurysms or those with cen- Medullary Cystic Disease (MCD)
tral nervous system symptomatology should be
undertaken. It should also be noted that an auto- Medullary cystic disease is another autosomal
somal recessive form of polycystic kidney dis- dominant trait manifesting kidney disease that
ease exists, but it has not been reported in has been reported in association with gout [266].
association with gout [336, 337]. Although this The frequency of the latter association is unknown.
form of cystic disease may permit survival beyond The disease is difficult to identify in clinical
the first two decades of life, most patients die in settings until its azotemic phase since flank
the newborn period. This short survival time may pain, hematuria, hypertension, and nephrolithiasis
120 5 Clinical Aspects of Gout and Associated Disease States

infrequently accompany the disorder. The kidneys been identified in a number of families, and more
are not enlarged or palpable. Cysts are present recently this constellation of findings has been
at the corticomedullary margin, deep medulla, given the name familial juvenile gouty nephropa-
and papilla. Tubular atrophy, glomerular hyalini- thy [348–369]. This heritable renal disease is also
zation, basement membrane thickening, and inherited as an autosomal dominant trait and may
patchy interstitial fibrosis are also associated be misdiagnosed as a familial nephropathy before
with these cysts. Sparse infiltration with chronic the appearance of gouty arthritis in a member of
inflammatory cells is also observed [342–345]. a family under evaluation. The disease often
Like ADPKD, an early sign of medullary cystic manifests itself as progressive renal failure. Since
disease is a loss of maximal concentrating capac- clinical gout and hypertension are inconsistent
ity. Salt wasting, hyponatremia, and extracellular features of the disease, some clinicians have
fluid contraction are often transient manifesta- omitted gout from the name and use the term
tions of this disease, but here again, a family his- familial juvenile hyperuricemic nephropathy
tory of kidney disease may be first clue to the [348, 366, 367, 370]. The onset of this disease is
presence of MCD. Further, clinical signs may not usually in the adolescent years or in early adult-
be recognized until azotemia occurs. Since the hood, but clinical gout has been reported in a
frequency of gout in patients with MCD may 9-year-old female [359]. As is clearly apparent,
be very low, such underlying pathology should gout in a female at this young age is distinctly
be excluded when one is presented with a patient unusual, and such an occurrence should alert cli-
with renal failure and gout. Thus, MCD, ADPKD, nicians to the possible presence of FJGN or one
and familial juvenile gouty nephropathy are of the other heritable disorders causing gout.
autosomal dominant traits that need to be con- Thus, the occurrence of gout at or before puberty
sidered when patients present with gout and a and especially in a female is a clinical marker for
family history of renal insufficiency. Of course, FJGN. Hyperuricemia out of proportion to the
all the inherited X-linked recessive traits associ- degree of renal failure should also raise the pos-
ated with gout may also present with renal fail- sibility of FJGN. Characteristically, the renal dis-
ure, and such patients must also be carefully ease progresses rapidly, and end-stage renal
assessed to determine the true underlying cause disease usually occurs in the third or fourth
of their gout. decade of life. Hypertension is not an early com-
ponent or consistent feature of the disease, but
when it occurs, the renal disease is aggravated.
Familial Juvenile Gouty Nephropathy Aggressive treatment of the hypertension and
(FJGN) hyperuricemia is essential if the deleterious
effects of these abnormalities on the progression
Hyperuricemia is a common feature of renal fail- of the renal disease are to be impeded.
ure from any cause, but gout in association with Renal biopsies from patients with FJGN are
chronic renal disease is unusual except in asso- most interesting because they rarely, if ever, show
ciation with specific renal diseases [346, 347]. deposition of urate crystals in the renal tissue.
Renal failure may complicate the Lesch-Nyhan Such biopsies do show severe tubulointerstitial
syndrome, the Kelley-Seegmiller syndrome, and nephropathy with sclerotic glomeruli and peri-
phosphoribosylpyrophosphate synthetase super- glomerular fibrosis. A patchy distribution of
activity. This complication usually occurs when tubular atrophy is also seen along with interstitial
these disorders are unrecognized or have compli- fibrosis and chronic inflammatory cell infiltrates.
cations secondary to renal calculi or infection. Basement membranes of the tubular cells may be
The usual clinical parameters of these genetic thickened as well [361, 363, 365, 369, 371].
disorders have been discussed previously. Several In addition to the characteristic progres-
additional heritable disorders need discussion sive renal insufficiency with its exaggerated
here. Gout, renal disease, and hyperuricemia have hyperuricemic response, a significantly decreased
Decreased Uric Acid Excretion 121

fractional excretion of uric acid (FEurate) is pres- urate secretion, a possible structural defect related
ent, and this abnormality may precede the onset to interstitial nephritis, thickened tubular base-
of the renal disease [367]. The mean clearance of ment membrane and atrophic tubules, or a defect
uric acid relative to creatinine (FEurate) in those in the basolateral or brush-border membrane pro-
patients with FJGN is usually less than 5 %, teins regulating urate transport. Recent studies
whereas normal values for this measurement have identified a urate transporter in rats [372–
usually range between 7 and 12 %. This fact 374]. Although the interactions between urate
emphasizes the need to evaluate carefully every transport and other organic anions in the human
member of an affected family since early recog- are complex and variations in transport mecha-
nition and treatment of this disorder can prevent nisms among various animal species make
the progression of the renal disease. One addi- extrapolations to the human difficult, the cloning
tional parameter of this disease is the occurrence of this urate transporter in rats may provide a
in some families of neonatal death or death in stepping stone to the understanding of human
utero [213]. Although the frequency of this pos- urate transport and perhaps the abnormality
sible complication of FJGN is unknown and the responsible for FJGN. Studies of this transport
cause for it is also presently unknown, the pos- system might also provide clues to the more com-
sible mating of two FJGN heterozygotes leading mon lesion in urate underexcretors documented
to the occurrence of a lethal homozygote cannot in primary gout. In the absence of a molecular
be excluded at this time. probe for FJGN, clinicians must diagnose the dis-
One final parameter of this disorder is of great ease from clinical findings in association with
significance and emphasizes the need for the rec- relatively sophisticated renal excretory studies
ognition of this disease. Recent data have deter- measuring the decrease in the fractional excre-
mined that FJGN is the most common disorder tion of urate. The latter studies must exclude a
studied by the Purine Research Laboratory at variety of other causes of decreased fractional
Guy’s Hospital in London [358]. It accounts for excretion of urate including drugs, toxic sub-
32 % of the cases of defective purine metabolism stances, severe respiratory acidosis, diabetic
evaluated in this laboratory over the past decade. ketoacidosis, metabolic alkalosis, and lead intox-
If one adds to this percentage, the frequency of ication [263, 375–379].
hypoxanthine-guanine phosphoribosyltransferase
deficiency, these two diseases, FJGN and
HGPRTase deficiency, make up more than 50 % Nephrogenic Diabetes Insipidus (NDI)
of the cases evaluated by this group. Like
HGPRTase deficiency, FJGN appears to have a Nephrogenic diabetes insipidus is a rare cause of
worldwide distribution since reports of the disor- gout since it has only been reported in three
der have been recorded in Asia, Australia, Europe, patients from different families [380, 381].
and the United States. Hyperuricemia has been reported in transient
In summary, the autosomal dominant trait, nephrogenic diabetes insipidus in association
familial juvenile gouty nephropathy, should be with pregnancy and hepatic failure, central dia-
considered in any case of documented familial betes insipidus, and nephrogenic diabetes insipi-
renal disease, and the occurrence of gout in any dus unrelated to pregnancy [381–385]. In
individual, male or female, of less than 30 years nephrogenic diabetes insipidus, the presence of a
of age should alert the clinician to the possibility lowered urate to creatinine clearance in associa-
of this progressive renal disease associated with tion with a normal 24-h urinary uric acid level
gout. The molecular cause for this disorder has suggested that the hyperuricemia in this
remains unknown; however, several hypotheses disorder is the result of renal handling of urate
have been proposed as potential contributors to [381]. This may result from the dilated renal col-
its pathogenesis. These include the presence of lecting system characteristically observed in this
an unrecognized metabolite capable of inhibiting X-linked recessive trait or the high filtration
122 5 Clinical Aspects of Gout and Associated Disease States

fraction known to occur in this disease. The rarity 143 meq/l, primary polydipsia can be excluded as
of gout may be due to better diagnostic methods a consideration.
and earlier recognition of the disorder. Diabetes In summary, gouty arthritis has rarely been
insipidus has been discussed as a clinical entity reported in association with diabetes insipidus;
associated with gouty arthritis, but its occurrence however, hyperuricemia is not an uncommon
in the modern literature is almost never reported. association with this disorder.
The diagnosis of diabetes insipidus is made by
documenting the abnormally large volumes of
dilute urine (>50 ml/kg body weight in 24 h) with Uric Acid Nephrolithiasis
a urine specific gravity of less than 1.010 or an
osmolality of less than 300 mOsmol/kg body Introduction
weight [386]. The presence of a serum sodium
concentration of greater than 143 meq/l and a Of the three types of renal afflictions (urate neph-
low specific gravity (<1.010) is highly suggestive ropathy, acute uric acid nephropathy, and uric
of diabetes insipidus if there is no history of a acid nephrolithiasis) associated with gout, renal
high sodium intake. The absolute diagnosis of calculi are probably the most common. Since uric
NDI is confirmed by the failure to concentrate acid lithiasis may occur in a variety of heritable
the urine in the presence of high plasma vaso- and acquired disorders, patients with such calculi
pressin concentrations and after the parental must be carefully evaluated to determine the
administration of vasopressin or desmopressin underlying cause and to institute the proper pre-
[387]. The chronic excretion of large volumes of ventative measures. This is especially important
urine in untreated patients causes hydronephro- in the absence of a stone for chemical analysis or
sis, hydroureter, and megacystis, all of which in the presence of a radiolucent filling defect by
may contribute to the hyperuricemic state [388]. radiographic examination. In these clinical set-
Female carriers of X-linked NDI may be com- tings, the following considerations must be taken
pletely asymptomatic or have varying degrees of into account. Pure uric acid stones are far less
polyuria and polydipsia [389, 390]. common than either pure or mixed stones com-
The differential diagnosis of NDI includes posed of calcium oxalate or calcium phosphate.
a deficiency of the synthesis of the antidiuretic Uric acid stones may be composed of pure uric
hormone, arginine vasopressin, in the supraop- acid or occur in combination with calcium
tic nuclei or a deficiency in the secretion of the oxalate, calcium phosphate, or magnesium phos-
hormone by the posterior pituitary. The cause phate. Pure uric acid stones are radiolucent (non-
for these so-called neurogenic types of diabetes opaque) and not visible on plain X-rays, whereas
insipidus may either be genetic (autosomal domi- their combination with calcium salts may make
nant inheritance of a mutant prepro-arginine- them visible on plain X-rays. Chemical analysis
vasopressin-neurophysin II gene) or acquired. is used to determine the primary component of
The acquired forms include defects due to trauma, any stone. In the United States, about 5–10 % of
malignancies, infections, granulomatous disease, all the reported kidney stones are composed of
autoimmune disorders, or vascular abnormali- uric acid [391–397]. In other areas of the world,
ties. Acquired forms of NDI may also result from the frequency of uric acid stones is much higher.
lithium treatment, hypokalemia, cystic renal For example, the prevalence of uric acid stones is
disease, or urinary tract obstruction. Primary 38 % in Iran, and in West Algeria, uric acid stones
polydipsia is the result of psychiatric illness and account for 31 % of the calculi seen in children
altered thirst mechanisms. It can arise as a result [398, 399]. In addition to this propensity for the
of compulsive water drinking or other mental frequency of urate stones to be increased in hot
aberrations. If there is a high water intake and a climates, the prevalence of such stones appears to
plasma osmolality of greater than 295 mOsmol/ be increased in workers exposed to heat stress in
kg body weight or a serum sodium of greater than their occupations [400]. Thus, climate, chronic
Uric Acid Nephrolithiasis 123

dehydration, and ethnic factors must be consid- Table 5.6 Uric acid nephrolithiasis
ered when evaluating uric acid nephrolithiasis. Heritable causes
At physiological pHs (7.4), uric acid is almost Lesch-Nyhan syndrome (X-linked recessive trait)
completely ionized and is in its soluble form, as Kelley-Seegmiller syndrome (X-linked recessive trait)
it exists in normal plasma. At low pH values, Phosphoribosylpyrophosphate synthetase
uric acid is quite insoluble. As the pH drops superactivity (X-linked recessive trait)
from 7.4, a greater proportion of the molecule is Polycystic kidney disease (autosomal dominant trait)
Heritable uric acid lithiasis (autosomal dominant trait)
converted to its non-ionized form. Free (non-
Glycogen storage disease type I (autosomal recessive
ionized) uric acid is much less soluble than its trait)
ionized counterpart. At the pKa point of uric Cystinuria (autosomal recessive trait)
acid (pH – 5.75), there are equal parts of ionized Hemoglobinopathies (autosomal recessive trait)
and non-ionized forms of the molecule. At a pH Thalassemias (autosomal recessive trait)
of 5.2, only about 20 % of uric acid is ionized. Hereditary renal hypouricemia (autosomal recessive
On the basis of these properties, the lower the trait)
urine pH, the less soluble uric acid becomes, Acquired causes
and the more likely for it to precipitate. A num- Myeloproliferative disorders
ber of studies have confirmed these parameters Lymphoproliferative disorders
by measuring urine pH in uric acid stone form- Multiple myeloma
Polycythemia
ers. In patients with gout and uric acid stones,
Pernicious anemia
the urine pH was less than 5.0 in 48 %, whereas
Uricosuric induced
in the matched controls, only 15 % had a urine
Simulators of uric acid lithiasis
pH below 5.0 [401]. In idiopathic uric acid
Iatrogenic xanthine lithiasis
nephrolithiasis, a disorder shown to have nor- Oxypurinol calculi
mal plasma and urinary uric acid levels, low 2, 8-Dihydroxyadenine lithiasis
urine pH has been documented as a primary Hereditary xanthinuria
contributor to stone formation [402–404]. Hyperuricosuric calcium oxalate lithiasis
Additional evidence for the role of urinary pH in
the formation of uric acid stones comes from
patients with chronic acidosis or chronic diar- levels of urinary uric acid of less than 300 mg/
rhea. The classic example is observed in patients day, the prevalence of uric acid stones is 11 %,
with an ileostomy who manifest increased uri- whereas this prevalence figure increases to 50 %
nary uric acid excretion, a decreased urinary when the urinary uric acid is more than 1,000 mg/
pH, and a decreased volume of urine. Such day [407]. Data similar to these have also been
patients are at increased risk for the formation reported from studies of patients with the inher-
of uric acid lithiasis [405, 406]. In addition to ited disorders of purine metabolism, HGPRTase
low urinary pH, dehydration from any cause deficiency, and PRPP synthetase superactivity. In
resulting in a low urine volume with increased both these genetic errors, the urinary uric acid
urinary uric acid concentrations and a low uri- concentration usually exceeds 1,000 mg/day
nary pH may create a setting for uric acid stone [408–410]. Thus, low urinary pH and increased
formation. urinary uric acid concentrations are the most fre-
Probably the most significant parameter caus- quent predisposing factors leading to uric acid
ing the generation of uric acid stones is an nephrolithiasis. For discussion of the clinical dis-
increased urinary uric acid concentration (hyper- orders associated with renal calculi, the causes of
uricosuria). Adult males usually excrete about kidney stones have been divided into heritable
800 mg of uric acid in a 24-h urine on a standard and acquired diseases associated with uric acid
diet, whereas females excrete between 700 and stones and those kidney stones that may mimic
750 mg/24 h. Data on the frequency of uric acid uric acid stones. This classification is summa-
nephrolithiasis show that in those patients with rized in Table 5.6.
124 5 Clinical Aspects of Gout and Associated Disease States

Heritable Causes of Uric Acid xanthine stones, phosphoribosylpyrophosphate


Nephrolithiasis synthetase superactivity with uric acid stones,
and glycogen storage disease with either uric acid
Any disorder associated with uric acid calculi or calcium oxalate stones. Even if the diagnosis is
demands the early identification of the underly- known and the patient is receiving allopurinol
ing cause so that the morbidity and, at times, the therapy, the possibility of iatrogenic xanthine or
mortality from the kidney stones and their com- oxypurinol calculi also needs to be considered
plications can be prevented. This is especially [414–416]. In the two other inherited X-linked
true for infants and children suffering from the recessive traits, Kelley-Seegmiller syndrome
Lesch-Nyhan syndrome, a disorder in which uric (partial hypoxanthine-guanine phosphoribosyl-
acid stones are quite common. Patients with this transferase deficiency) and phosphoribosylpyro-
disease are often difficult to manage and may be phosphate synthetase superactivity, uric acid
confused with cerebral palsy patients. They are calculi have also been reported. More than three-
often uncooperative and their central nervous quarters of the patients with Kelley-Seegmiller
system disease makes their surgical management syndrome have a history of renal stones. Although
even more difficult. In fact, the author lost a renal calculi also occur in the PRPP synthetase
patient who required nephrectomy secondary to defect, data for the frequency of stone in this
kidney stones and a treatment-resistant kidney enzyme abnormality are lacking. Nonetheless, if
infection. The post-nephrectomy course was the serum uric acid is greater than 10–13 mg/dl or
hampered by the movement disorder and the the urinary uric acid excretion is greater 1,000 mg/
inability to control the patient’s attempts to dam- day, an error in purine metabolism or a disorder
age the operative site with its drain. The patient causing an increased turnover of nucleic acids
eventually succumbed to an overwhelming must be excluded. Lower levels of these parame-
infection. ters do not exclude the presence of a heritable
In children with the Lesch-Nyhan syndrome, disorder in purine metabolism.
urinary uric acid excretion ranges between 40 Uric acid calculi may or may not be associated
and 70 mg of uric acid per kilogram of body with gout. In some cases, the kidney stone may
weight per day [411, 412]. Gouty arthritis cannot precede the onset of acute gouty arthritis. For
be used as a marker for the presence of this example, a urinary calculus may be the first sign
X-linked disorder since gout is rare in the Lesch- of the Kelley-Seegmiller syndrome or phosphori-
Nyhan syndrome. Further, hyperuricemia may bosylpyrophosphate synthetase superactivity.
not be a manifestation of the disease since chil- Kidney stones or painful crystalluria may also be
dren have a high renal clearance of this metabo- the presenting sign of the Lesch-Nyhan syn-
lite. If renal failure is not present, uric acid drome, and in this disorder, gout is rare.
excretion relative to creatinine is usually elevated Even though HGPRTase deficiency and PRPP
to levels twice or four times normal [348, 358]. In synthetase superactivity are probably the most
the presence of renal failure, these findings may common heritable diseases associated with gout
be masked [358, 413]. In this setting, the serum and uric acid calculi, several other inherited dis-
uric acid is usually markedly elevated in the range eases have been documented as causes of gout
of 13–17 mg/dl. Renal ultrasonography is a use- and/or uric acid calculi. About 20 % of the
ful noninvasive tool that may detect crystal neph- patients with autosomal dominant polycystic kid-
ropathy. Of course, the absolute diagnosis must ney disease (ADPKD) develop renal stones com-
be determined by enzyme analysis. Urolithiasis posed of either uric acid or calcium oxalate [310,
and crystal nephropathy with or without renal 417–420]. Calcifications within the kidney paren-
failure are also observed in other inherited disor- chyma may also be observed in ADPKD and
ders such as adenine phosphoribosyltransferase must be differentiated from stones within the uri-
deficiency with 2,8-dihydroxyadenine stone for- nary collecting system. The best method for
mation, xanthine dehydrogenase deficiency with accomplishing this differentiation is by computed
Uric Acid Nephrolithiasis 125

tomography (CT) scanning of the kidneys [340]. localized to different chromosomes depending on
Like all disorders associated with nephrolithiasis, the amino acid excretion pattern [435–437].
it is essential to identify stones that are not passed Recent publications have also documented an
and may result in urinary tract infection, obstruc- increase in uric acid excretion in some cystinuric
tive uropathy, and deterioration of kidney func- patients [438]. In this series of patients, hypercal-
tion. CT scanning or magnetic resonance imaging ciuria and hypocitraturia have also been demon-
is useful for identifying and excluding retained strated in 22 and 44 % of the patients, respectively.
stones. The “abnormality” of uric acid excretion is of
Renal disease including stone formation is interest for several reasons. These data confirm
also common in adult glycogen storage disease an association between increased urinary uric
[421, 422]. Contrary to common opinion, most acid and stone formation as will be discussed
renal calculi in this disorder are composed of cal- subsequently in calcium oxalate stone formers. It
cium oxalate, but uric acid stones have also been also emphasizes the concept that acid urines,
reported. Alterations in uric acid metabolism known to occur in both gout and cystinuria, pro-
occur in glycogen storage disease as a result of vide a setting for the formation of stones in meta-
the decreased renal urate clearance due to lacti- bolic abnormalities other than those associated
cacidemia and ketonemia as well as the increased with errors of purine metabolism. In a more spec-
production of uric acid secondary to ATP degra- ulative realm, there is the possibility that altera-
dation and increased pentose phosphate shunt tions in renal transport proteins such as those
activity [90, 423–425]. Distal renal tubular acido- mutated in cystinuria may affect other renal trans-
sis, hypocitraturia, nephrocalcinosis, and hyper- port mechanisms including those responsible for
calcemia have also been described in glycogen uric acid transport. In summary, cystinuria is a
storage disease, and these abnormalities may also very rare cause of uric acid stones, and this asso-
contribute to stone formation [143]. ciation may be related to defects in transport
Even though pure cystine stones are the most proteins.
common renal calculi observed in homozygous One report of uric acid nephrolithiasis has
cystinuria, a few individuals with this relatively been observed in three independent families in
rare autosomal recessive disorder of renal tubule the absence of hyperuricemia and gout [439].
and gastrointestinal amino acid transport may This form of uric acid lithiasis is inherited as an
also develop calcium oxalate or uric acid stones autosomal dominant trait and is not reported to be
[426, 427]. The occurrence of hyperuricemia, associated with any other abnormalities. No fur-
normal renal function, and cystinuria has been ther reports of this familial form of uric acid stone
reported in the older literature [428–431]. The formation have appeared in the literature. Rarely,
presence of uric acid nephrolithiasis in cystinuric uric acid nephrolithiasis has been observed in
patients has also been cited in a review of the dis- specific hemoglobinopathies and thalassemias,
eases associated with uric acid stones [432]. especially in the presence of severe hemolysis.
The primary clinical expression of cystinuria Finally, in contrast to these disorders associ-
is the formation of renal/bladder stones and their ated with uric acid overproduction, hypouricemia
complications including urinary tract infections and defective urate reabsorption are components
and urine outflow obstruction. Urinary excretion of hereditary renal hypouricemia, and the latter
of cystine as well as arginine, lysine, and orni- disorder often gives rise to uric acid kidney
thine in various patterns is increased. The genetic stones. Neither this disease nor cystinuria is
defect in the renal tubule has been localized to a accompanied by gouty arthritis. Hereditary renal
human gene whose protein, a type II membrane hypouricemia is a rare autosomal recessive trait
glycoprotein, is a carrier protein for the reabsorp- with a defect in postsecretory reabsorption of
tion of cystine and other dibasic amino acids urate producing hypouricemia, increased renal
across the proximal brush border membrane clearance of urate, hyperuricosuria, and/or hyper-
[433, 434]. The cystine transport gene has been calciuria. In 25 % of the homozygotes with this
126 5 Clinical Aspects of Gout and Associated Disease States

disorder, calcium oxalate or uric acid stones have 7 and 12 %, whereas in those with hereditary
been reported [440–444]. In other reported cases renal hypouricemia, this value is increased to lev-
where hypouricemia exists in the absence of any els between 30 and 90 %. Even though the inter-
pattern of inheritance, calcium oxalate stones pretation of data acquired using pyrazinamide to
have recovered [445]. These cases show quite localize the renal tubular transport of urate is
different dysfunctions in tubular transport, and imprecise, both pyrazinamide and probenecid
only one of the patients has been documented to tests have been used to determine the defects in
have a defect in presecretory urate reabsorption. renal tubular transport. As discussed previously,
Interestingly, the latter patient had multiple normal renal handling of uric acid by the kidney
myeloma associated with type 2 renal tubular aci- includes 100 % filtration at the glomerulus,
dosis. The other patients described in this publi- 98–100 % reabsorption in the proximal tubules,
cation could be classified as idiopathic renal 50 % distal proximal tubular secretion, 40–45 %
hypercalciuria [446]. Thus, some patients with postsecretory reabsorption, and 5–10 % secre-
hypouricemia may present with calcium oxalate tion. On the basis of these analyses of human
stones but do not fit into the true definition of renal urate transport, five different defects of
hereditary renal hypouricemia even though they urate transport have been postulated: a complete
have an increased clearance of uric acid. transport deficiency (no uric acid secretion or
One useful key to the recognition of patients at reabsorption), a complete urate reabsorption
the greatest risk for uric acid stones is the quanti- deficiency, a presecretory reabsorption defect, a
tation of urinary uric acid since those with stones postsecretory defect, and secretory superactivity.
usually excrete more than 900–1,000 mg of uric These postulated defects express themselves to
acid/day. Some patients with hereditary renal variable degrees as a result of individual varia-
hypouricemia also manifest hypercalciuria [440, tion, overlapping renal tubular functions, and
444, 447, 448]. The mechanism for this hypercal- probably the existence of partial deficiencies in
ciuria has been shown to be due to increased transport proteins. When FCurate values and the
intestinal reabsorption in some, but in others, the alterations in pyrazinamide and probenecid are
cause for the hypercalciuria remains unknown tabulated from the reports of hereditary renal
[440, 444]. The degree of hypercalciuria in sev- hypouricemia documented in the literature, the
eral patients has also been shown to lead to a following tubular transport deficiencies have
decreased bone density and osteoporosis [447, been identified. More than half the subjects
449]. There has been a suggestion that the mas- reported can be classified as having defective pre-
sive increase in urate clearance may lead to acute secretory urate reabsorption [440, 447, 448, 456–
uric acid nephropathy with associated renal 462]. Other reported cases of hypouricemia and
parenchymal deposition of urate, but absolute renal transport defects fit the pattern of different
proof of such lesions in hereditary renal hypouri- transport abnormalities [449, 459, 462–464].
cemia is lacking [450, 451]. To differentiate hereditary renal hypouricemia
The key to the diagnosis of hereditary renal from a variety of other genetic and acquired dis-
hypouricemia lies initially with the identification orders is not a simple task since there are a large
of a low serum urate level that is less than 2.5 mg/ number of conditions that can give rise to hypou-
dl in adult males and less than 2.1 mg/dl in adult ricemia (Table 5.4). Since there are fewer than 50
females [452–455]. Such low serum urate levels, cases of hereditary renal hypouricemia in the
as noted previously, could be the result of either a world’s literature, more frequent causes of hypou-
low production of uric acid or an excessively high ricemia need to be excluded initially. Probably
rate of urate clearance. In the case of hereditary the most common cause of hypouricemia is
renal hypouricemia, it is the latter process that related to the use of pharmacologic agents that
leads to the low serum urate levels. In normal affect renal tubular urate transport. Salicylates
individuals, the fractional urate clearance (clear- (aspirin) are known to have a paradoxical effect
anceurate/glomerular filtration rate) ranges between on uric acid excretion since this drug causes uric
Uric Acid Nephrolithiasis 127

acid retention by inhibiting uric acid secretion at arthralgias and arthritis similar to gouty arthritis
low doses (serum salicylate levels of 5–10 mg/ have been reported in some cases of xanthinu-
dl), whereas at high doses (serum salicylate lev- ria [477, 484, 489, 491–494]. It has been pos-
els of >15 mg/dl), the drug inhibits uric acid reab- tulated that this is a crystal-induced arthropathy
sorption and causes uricosuria [262, 465]. Some since animal studies have documented that xan-
radiocontrast agents (iapanoic acid, megulamine thine crystals can cause an inflammatory arthri-
iodipamide, and sodium diatrizoate) have urico- tis [484]. In some cases of xanthinuria, muscle
suric properties and can also lower serum uric inflammatory responses have been documented
acid levels [466, 467]. Glyceryl guaiacolate also in association with muscle aches and pains [484,
causes hypouricemia [453, 468]. The less com- 489, 493–497]. The significance of this recessive
monly used uricosuric drugs such as probenecid, trait, hereditary xanthinuria, is that it may present
sulfinpyrazone, and phenylbutazone may also with a migratory arthritis and a radiolucent kid-
reduce serum uric acid levels. Dicoumarol and ney stone under clinical circumstances mimicking
outdated tetracycline also reduce serum uric acid the Lesch-Nyhan syndrome, adenine phospho-
levels; the latter drug causes a Faconi-like syn- ribosyltransferase deficiency with its radiolu-
drome [469]. Thiazides and loop diuretics, known cent 2,8-dihydroxyadenine stones, and in a few
commonly for their capacity to cause hyperurice- other disorders. For this reason, identification of
mia, increase the fractional excretion of urate the chemical constituents of any kidney stone is
acutely, whereas when the volume of extracellu- critical for diagnosing the underlying disorder.
lar fluid is contracted from their use, diuretics Xanthine, uric acid, and 2,8-dihydroxyadenine
decrease the fractional excretion of uric acid. stones all give a purple color in the murexide test
Mannitol, when used as an osmotic diuretic, also [498]. Thus, the murexide test may provide a clue
causes uricosuria and hypouricemia. More to an underlying defect in purine metabolism, but
recently, the angiotensin II receptor antagonist, an ultraviolet spectrum at pH 2.0 and 10.0, col-
losartan, has been shown to be a potent uricosuric umn chromatography, infrared or mass spectrom-
[470–474]. In fact, losartan is a more potent etry, or X-ray crystallography are necessary for
inhibitor of urate reabsorption than probenecid the precise identification of xanthine and these
on the basis of its high affinity for a human urate/ other causes of stones.
anion renal tubular exchanger. Finally, high-dose One child with hypouricemia and mental retar-
ascorbic acid (>4 g/day) causes hypouricemia. dation has been reported [499–501]. Presumably,
There are some rare heritable enzymatic this child acquired the hypouricemic state as a
deficiencies that also cause hypouricemia. result of a decreased level of PRPP accompa-
Classical xanthinuria type 1 arises as a result of nied by a significant decrease in de novo purine
a mutation in the xanthine dehydrogenase gene, nucleotide biosynthesis. The remainder of the
and such a mutation leads to the inability of the inherited hypouricemic disorders is those asso-
human host to convert hypoxanthine and xanthine ciated with the Fanconi syndrome and defec-
to uric acid. This heritable disorder is discussed tive proximal renal tubular urate reabsorption.
briefly here and in more detail in the subsection These hypouricemic disorders include Wilson’s
dealing with those disorders that simulate uric disease, galactosemia, hereditary fructose intol-
acid nephrolithiasis. Patients with xanthinuria erance, cystinosis, and the Hartnup syndrome
may be totally asymptomatic or present with uro- [502–509].
lithiasis and its complications [475–483]. Renal There are a number of acquired causes of
damage may be severe leading to chronic renal hypouricemia that need to be excluded before
failure in some patients while in others, the sever- hereditary renal hypouricemia can be invoked as
ity of the kidney problem has resulted in nephrec- the cause for the hypouricemia in an isolated patient
tomy and terminal uremia [484–490]. In addition without a family history of an inherited renal tubu-
to xanthine stones that are nonopaque by plain lar disorder. Malignancies including pulmonary
X-rays unless they contain calcium oxalate, neoplasms, lymphomas (Hodgkin’s disease), and
128 5 Clinical Aspects of Gout and Associated Disease States

multiple myeloma are often associated with with hematological and chronic hemolytic ane-
hypouricemia and a Fanconi-like renal tubular mias are documented in the older literature, but
disorder [510–515]. Hypouricemia may also be a the recent literature is marked by the absence of
marker for the inappropriate secretion of antidi- such case reports [407, 432, 529]. The lack of
uretic hormone (ADH) and the resultant expan- reports in the modern literature probably results
sion of the extracellular volume [516–518]. In from an earlier recognition of hematological dis-
addition to its role as a marker of the possible orders and the use of allopurinol to prevent acute
presence of an underlying malignancy, the degree uric acid nephropathy and nephrolithiasis.
of hypouricemia appears to correlate with the In some instances, uric acid stones may com-
activity of the disease in Hodgkin’s lymphoma plicate non-hematological cancers and are asso-
[510, 511, 519]. The remaining disorders causing ciated either with increased tumor cell turnover
renal hypouricemia are the renal tubular lesions or the lysis of tumor cells induced by therapeutic
of heavy metal intoxication and liver disease with agents [530]. When primary and secondary gout
jaundice [520–523]. A search of the literature are considered, the incidence of renal calculi in
does not reveal any cases of nephrolithiasis in primary gout (no underlying disease other than
renal hypouricemia except for those observed in gout) is only 22 %, whereas this percentage is
hereditary renal hypouricemia. roughly doubled in secondary gout (gout second-
In this group of heritable diseases associated ary to another disorder or drug use) [407]. Thus,
with nephrolithiasis, the pattern of inheritance is the causes of secondary gout with the potential
variable and may assist in identifying the under- for uric acid nephrolithiasis include myeloprolif-
lying disease. X-linked inheritance is observed erative and lymphoproliferative disorders, multi-
with the Lesch-Nyhan syndrome, Kelley- ple myeloma, secondary polycythemia, pernicious
Seegmiller syndrome, and phosphoribosylpyro- anemia, chronic hemolytic anemias, certain
phosphate synthetase superactivity, whereas hemoglobinopathies, polycythemia vera, myeloid
autosomal dominant inheritance occurs with metaplasia, Gaucher disease, chronic myelocytic
polycystic kidney disease, heritable uric acid lith- anemia, and thalassemias [125, 398, 530].
iasis, and medullary cystic disease. The latter dis- Several unusual and rare causes of uric acid
ease can present with kidney stones, but such an nephrolithiasis exist in addition to those associ-
occurrence is rare. Finally, a variety of autosomal ated with hematological diseases. Uric acid lithia-
recessive diseases including glycogen storage sis has been reported in association with the
disease, thalassemia, hemoglobinopathies, hered- uncommon occurrence of a calculus forming in
itary renal hypouricemia, and cystinuria may also patients with functioning renal transplants [531,
be associated with uric acid lithiasis. 532]. A variety of causes may contribute to this
clinical problem including excessive purine intake
and cyclosporine treatment [532, 533]. Uric acid
Acquired Causes of Uric Acid calculi in this clinical setting represent only one
Nephrolithiasis type of calculus seen in renal transplant patients
since stones composed of other substances have
Acquired disorders of uric acid lithiasis are prob- been reported in renal transplant patients with
ably of greater significance to recognize in the hyperparathyroidism, hyperoxaluria, and even
pediatric population than in adults. Children are adenine phosphoribosyltransferase deficiency
susceptible to urolithiasis in association with [534, 535]. Another rare cause of uric acid neph-
hematological malignancies either as a result of rolithiasis has been observed in some patients who
chemotherapy or as an initial event due to have abused laxatives and, as a result, acquired
increased nucleic acid turnover [524–527]. Uric hypokalemia, extracellular volume depletion, sys-
acid nephrolithiasis may also occur in children temic alkalosis, and elevated angiotensin levels
due to nonmalignant conditions [528]. In adults, [536, 537]. This complication is seen most fre-
reports of uric acid nephrolithiasis in association quently in females and should be suspected when
Uric Acid Nephrolithiasis 129

an ammonium urate calculus is found in conjunc- sophisticated stone analyses are necessary to
tion with a sterile urine. Urine specimens in these characterize the chemical composition of the
cases may test positive for phenolphthalein. stone [545–548]. The use of allopurinol to con-
Finally, a single case of Prader-Willi syndrome trol gross endogenous uric acid overproduction
has been reported with uric acid urolithiasis causes significant elevations in plasma xanthine
[538]. According to the authors of this case levels. The latter state may be greatly exagger-
report, the stone resulted from the hyperphagia ated when HGPRTase is deficient, and the host is
that occurs in Prader-Willi syndrome. The latter unable to reutilize hypoxanthine. The renal clear-
syndrome is characterized by hypotonia in ance of xanthine approaches and may exceed the
infancy, hyperphagia, mental retardation, hypog- glomerular filtration rate resulting in an elevated
onadotrophic hypogonadism, and small hands urinary xanthine concentration that may exceed
and feet. Despite the relatively common occur- the limited solubility of this metabolite in urine
rence of this genetic syndrome, this is the only [475, 495, 549]. In fact, the urinary concentration
report of a uric acid calculus associated with this of xanthine may approach or exceed 1 mmol/
or other hyperphagic syndromes [539–541]. Even mmol of creatinine in patients treated aggres-
though this is an inherited genetic disorder, it is sively for urate overproduction or malignancies
discussed under acquired disorders since the as a result of the markedly elevated serum xan-
cause for the uric acid stone is overeating. thine concentrations and the high renal clearance
of xanthine. Normally, the urinary xanthine con-
centration is less than 0.01 mmol/mmol of creati-
Nephrolithiasis Mimicking Uric nine. The calculated solubility of xanthine in the
Acid Stones urine at pH 5.0 and 7.0 is 5 and 13 mg/dl (~ 0.03–
0.07 mmol/dl), respectively, and these values can
A variety of renal calculi composed of non-urate often be exceeded during allopurinol therapy in
metabolites may mimic uric acid stones due to certain clinical settings [550]. For these reasons,
their nonopaque appearance on plain radiographs xanthine crystalluria, xanthine stones, and acute
or as filling defects on excretory urograms, and at renal failure may occur in patients treated with
times, non-urate stones occur in patients with dis- allopurinol, and these complications are most
orders that may resemble dysfunctions similar to likely to be observed in the Lesch-Nyhan syn-
those observed with alterations in uric acid drome, the Kelley-Seegmiller syndrome, and in
metabolism. The differential diagnosis of such patients being treated aggressively with
filling defects includes uric acid calculi, xanthine allopurinol to protect them from excessive tissue
calculi, 2,8-dihydroxyadenine stones, indinavir- nucleic acid breakdown and increased uric acid
associated lithiasis, blood clots (especially in production during chemotherapy or radiotherapy
those with hemophilia), renal cysts, inflammatory of malignancies [545, 548, 551–556].
processes, and renal carcinoma [542]. Since Except for the characteristics of the stones
unexplained filling defects observed radiographi- themselves, the remainder of the parameters of
cally in the renal pelvis and elsewhere are nondi- allopurinol-induced xanthine stone formation is
agnostic, CT scanning has been recommended as defined by laboratory findings. Xanthine stones
a tool for diagnosing nonopaque calculi [543, are usually brown or orange brown in color and
544]. The principal simulators of uric acid calculi can easily be transected. The cut surface is lami-
are stones composed of xanthine, 2,8-dihydroxy- nated and may contain a core of calcium oxalate
adenine, and oxypurinol. [476–478, 489]. They may be detected by renal
ultrasonography [416]. Since the renal clear-
Iatrogenic Xanthinuria and Xanthine ance of oxypurine is so high, the serum oxypu-
Lithiasis rine concentration may only rise to 0.5–2.0 mg/
Xanthine calculi have been described in patients dl, whereas the normal plasma oxypurine
with disorders requiring allopurinol therapy, and concentrations are in the range of 0.1–0.3 mg/dl
130 5 Clinical Aspects of Gout and Associated Disease States

[550, 557–560]. The ratio of xanthine to hypox- urine [485, 563–570]. To assure the validity of
anthine during allopurinol therapy is roughly the results, a second method of xanthine analysis
2:1 [561]. The serum uric acid in the course of is usually performed. One simple method for
maximal allopurinol therapy may be reduced to assuring that a peak is truly xanthine is to rerun
2–3 mg/dl in the presence of normal renal func- the sample after digestion of the xanthine with an
tion. The urinary excretion of oxypurines is often excess of xanthine oxidase. Another method is to
the key to identifying the potential for crystalline run the sample using two different separation
deposits of xanthine or the formation of xanthine systems for the detection of the metabolite.
stones. Urinary oxypurines in patients receiving The clinical management of xanthine stones is
allopurinol are usually in the range of 50 to over similar to the management of any other kidney
100 mg/day in contrast to the normal urinary con- stone. In cases where the overproduction of urate
centration of less than 20 mg/day. In patients who requires high doses of allopurinol to control,
develop acute renal failure, urinary xanthine con- allopurinol dose reduction and alternative means
centrations may be as high as 152 mg/mmol of to regulate urate production such as diet and/or
creatinine (~ 1.5 g/g of creatinine), whereas the alternative drugs should be considered.
normal urinary xanthine concentration usually
less than 1.5 mg/mmol of creatinine (15 mg/g of Hereditary Xanthinuria
creatinine). Hereditary xanthinuria is a rare autosomal reces-
An animal model of xanthine crystalluria has sive trait that may affect the kidney and/or mus-
been used to evaluate the course of renal damage culoskeletal system. There are two subtypes of
in pigs fed a high purine diet in the form of gua- this disorder that have been identified by differ-
nine along with allopurinol [562]. This model ences in their capacity to oxidize allopurinol
demonstrated the extensive renal tubular damage [571]. Subsequently, these variations have been
associated with the deposition of xanthine crys- expanded to include the ability or inability to oxi-
tals in the pig kidney. The crystalline deposits dize pyrazinamide [572, 573]. Type I xanthinuria
were found predominantly in the distal renal represents a deficiency of xanthine dehydroge-
tubules and collecting ducts and caused wide- nase (XDH) with normal activity of aldehyde
spread tubular epithelial damage including inter- oxidase (AOX) and sulfite oxidase. The absence
stitial edema and inflammation. Such renal of XDH results in the inability to oxidize hypox-
inflammatory lesions and scarring may well be anthine and xanthine to uric acid [571–573]. This
the precursor of permanent renal damage such as deficiency also results in the inability to oxidize
in the case in those patients who manifest renal pyrazinoic acid to 5-hydroxypyrazinoic acid.
insufficiency during the course of allopurinol Since type I-deficient patients have aldehyde oxi-
therapy. dase activity, allopurinol is converted to oxy-
The differential diagnosis of allopurinol-in- purinol, thiopurinol to oxythiopurinol, and
duced xanthinuria includes a number of disorders pyrazinamide to 5-hydroxypyrazinamide [571–
associated with the renal handling of urate as 576]. Type II xanthinuria results from a deficiency
well as hereditary xanthinuria. Clues to the pres- of both xanthine dehydrogenase and aldehyde
ence of these disorders include the presence of oxidase. These enzyme deficiencies block the
hypouricemia and the use of allopurinol. However, oxidation of N-methylnicotinamide to 2- to
reduced levels of serum uric acid may not be 4-pyridonecarboxamides, allopurinol to oxy-
observed in the face of renal failure when purinol, and pyrazinamide to 5-hydroxypyrazin-
allopurinol is prescribed. Thus, iatrogenic xanthi- amide [480, 495, 570, 571, 577, 578]. Roughly,
nuria with stone formation is probably best diag- 50 % of the patients with classical xanthinuria
nosed by ultrasonography and the measurement will also manifest a deficiency in aldehyde
of urinary purine metabolites. A variety of sensi- oxidase.
tive methods are available for the identification of The principal clinical manifestations of classi-
hypoxanthine and xanthine in the plasma and cal xanthinuria are related to the formation of
Uric Acid Nephrolithiasis 131

xanthine stones or acute renal failure secondary patients with hereditary xanthinuria for reasons
to xanthine crystalluria. About 40 % of the that are not clear at this time [484, 566, 571,
patients with classical xanthinuria are diagnosed 583–586]. Other diseases observed in associa-
on the basis of urolithiasis or its associated com- tion with hereditary xanthinuria appear to be
plications such as renal colic, urinary tract infec- coincidental findings.
tions, hematuria, or other clinical difficulties The absolute diagnosis of hereditary xanthi-
associated with crystalluria. Another 20 % of the nuria requires an in vitro assay for xanthine dehy-
cases are completely asymptomatic and often drogenase/xanthine oxidase activity in duodenal
discovered through family studies by the pres- or jejunal mucosa biopsies. Liver biopsies may
ence of hypouricemia. Although urolithiasis and also be assayed for enzyme activity, but the inher-
crystalluria may occur at any age from birth ent dangers in closed liver biopsies obviate the
through the eighth decade, most of the cases are use of the liver as a source of the enzyme unless
first observed in infants who express the disease laparotomy is planned or other reasons necessi-
by nonspecific signs such as irritability, insom- tate liver biopsy. Sample preparations and assay
nia, vomiting, and poor weight gain [479]. Like conditions are discussed along with references to
the Lesch-Nyhan syndrome and APRTase detailed assays in other resources [587]. Analysis
deficiency, hereditary xanthinuria may lead to of muscle tissue from affected patients may be
serious renal disease and even requiring nephrec- suggestive of the underlying condition on the
tomy. Indeed, some patients may die from uremia basis of their elevated concentrations of hypox-
[481, 485–490]. Children may also present with anthine and xanthine [484, 493, 580, 582].
significant signs of kidney damage such as Control values for hypoxanthine were less than
clubbed calices or hydronephrosis. Renal tubular 30 ng/mg dry weight of muscle, and values from
acidosis and hypercalciuria have also been affected muscle were 8–12 times higher than the
reported in association with xanthinuria [482]. control value. Control values for xanthine were
Approximately 10 % of xanthinurics may have less than 50 ng/mg dry weight of muscle, whereas
either articular symptoms or myalgias. Pain, in affected muscle, the values were six to nine
tightness, and stiffness in the distal upper and times higher than the controls.
lower limbs are the usual complaints of adults Since the enzyme, xanthine dehydrogenase/
with this presentation [477, 484, 489, 493, 497, xanthine oxidase, converts hypoxanthine and
579, 580]. In some of those patients with muscle xanthine to uric acid, in its absence, plasma and
symptoms, xanthine and hypoxanthine crystals urinary uric acid levels are low and often are
have been observed as crystal deposits in skeletal undetectable. Plasma levels of oxypurines vary
muscle biopsy specimens [581, 582]. There is between 0.01 and 0.07 mg/dl in healthy individu-
speculation that muscle exercise induces lactic als. Urinary oxypurine concentrations vary
acid production, a fall in tissue pH, and the depo- depending on the purine dietary intake, but males
sition of oxypurines as a result of the insolubility in the United Kingdom evaluated on a low purine
of these metabolites in an acid environment. diet have been reported to excrete between 4 and
Recurrent migratory polyarthritis is also 10 mg/24 h of hypoxanthine and a similar quan-
observed in a small number of patients with xan- tity of xanthine [587]. In xanthinuria, plasma
thinuria [477, 484, 489, 491–493]. Since xan- xanthine concentrations are elevated to levels
thine crystals can cause arthritis in animal between 0.15 and 0.61 mg/dl, whereas plasma
models, it has been hypothesized that crystal hypoxanthine concentrations are less than
deposition may cause the synovitis observed in 0.068 mg/dl [479, 485, 566, 567, 569, 576].
humans with xanthinuria [484]. However, to Urinary excretion of oxypurines in hereditary
date, no hypoxanthine or xanthine crystals have xanthinuria is dominated by xanthine in ratio of
been found in synovial fluid or tissues from xan- 4:1 (xanthine/hypoxanthine), and the total oxy-
thinuric patients. There also appears to be an purine concentration in the urine ranges between
increased frequency of duodenal ulcers in 300 and 450 mg/24 h.
132 5 Clinical Aspects of Gout and Associated Disease States

The differential diagnosis of hereditary xan- their radiolucency [597, 598]. The unusual
thinuria depends in large part on what parameter occurrence of a radiopaque 2,8-dihydroxyade-
is selected for comparison with other diseases. If nine stone emphasizes the need to investigate
stone formation is the clinical marker, then a stone composition by sophisticated analyses
large variety of causes of urolithiasis must be [599]. 2,8-DHA urolithiasis results from a com-
considered. However, if the stone is radiolucent, plete or partial deficiency of the purine salvage
then the differential diagnosis can be restricted to enzyme, adenine phosphoribosyltransferase
causes related to stones composed of uric acid, (APRTase). This deficiency is characterized by
xanthine, cystine, or 2,8-dihydroxyadenine. It is a spectrum of clinical findings related to the
important to recognize that about 85 % of stones excretion of the insoluble metabolite, 2,8-dihy-
contain calcium so that completely radiolucent droxyadenine, by the kidney. Despite the neph-
stones are the exception rather than the rule. rotoxicity of 2,8-DHA, roughly 15 % of patients
Perhaps the best method for differentiating these with this defect remain completely asymptom-
disorders is to measure the serum uric acid levels atic. When crystals or stones of 2,8-DHA are
and the urinary uric acid levels. If hyporuricemia formed, urinary tract infections, hematuria, dys-
is present (serum uric acid levels of less than uria, colicky pain, obstructive uropathy, or, in a
2.5 mg/dl) and is selected as the clinical marker few clinical settings, acute anuric renal failure
for comparison, two mechanisms can be evalu- may occur [600]. Unfortunately, if unrecog-
ated to assess its causation: decreased uric acid nized in a timely fashion and untreated, some
production and increased excretion of urinary patients will develop chronic renal insufficiency
uric acid. Further, low urinary uric acid levels requiring chronic dialysis and/or renal trans-
identify those patients with decreased uric acid plantation [601–605]. The presence of radiolu-
production such as hereditary xanthinuria, severe cent renal calculi and chemical analyses of
liver disease, phosphoribosylpyrophosphate syn- kidney stones have led to the misdiagnosis of
thetase deficiency, and allopurinol/oxipurinol uric acid stones [601, 602, 606–608]. This con-
administration (iatrogenic xanthinuria) [453, 499, fusion resulted primarily because simple chemi-
588–590]. A complete classification of hypouri- cal analyses such as the phosphomolybdate and
cemia is tabulated in Table 5.7. Population sur- murexide tests, the ultraviolet spectrum in alka-
veys have determined the frequency of line solutions, and thermogravimetric analyses
hypouricemia to be in the range of 0.5–1.0 % give identical results for uric acid and 2,8-dihy-
[453, 455, 591–594]. The frequency of hypouri- droxyadenine [601, 602, 609–612]. However,
cemia appears to be significantly lower in the these stones can be distinguished from uric acid
Japanese population [595, 596]. These popula- by performing an ultraviolet spectrum of the
tion surveys document the low frequency of stone components in both alkali and acid solu-
hypouricemia and the fact that most hypourice- tions, by using infrared analysis, by mass spec-
mic patients manifest urate hypersecretion in trometry, or by X-ray crystallography [605, 606,
contrast to those with hyposecretion. Further, 609, 610, 613].
hypouricemia is frequently associated with the APRTase deficiency is inherited as an auto-
use of medications. somal recessive trait, and heterozygotes are usu-
ally completely asymptomatic [614–619]. Two
2,8-Dihydroxyadenine Lithiasis homozygote subsets of this disorder can be dis-
Not all radiolucent stones are composed of uric tinguished by enzymatic analysis. Type I
acid and 2,8-dihydroxyadenine (2,8-DHA), and deficiency is characterized by a complete absence
xanthine stones are also nonopaque and may be of adenine phosphoribosyltransferase activity in
difficult on clinical grounds alone to differentiate erythrocyte lysates, whereas patients with type II
from uric acid stones. 2,8-Dihydroxyadenine cal- deficiency have about 10–25 % of normal activity
culi have often been misdiagnosed as uric acid in their lysates. Patients with type I deficiency
stones on the basis of their chemical analysis or have been identified worldwide, whereas those
Uric Acid Nephrolithiasis 133

Table 5.7 Causes of hyperuricemia 8-hydroxyadenine, and 2,8-dihydroxyadenine is


I. Decreased uric acid production 1.0:0.03:1.5, respectively. These urinary purines
A. Hereditary xanthinuria are excreted because of the inability to salvage
B. Severe liver disease adenine by the enzyme, adenine phosphoribo-
C. Phosphoribosylpyrophosphate synthetase deficiency syltransferase. In the absence of this enzyme,
D. Allopurinol/oxipurinol administration adenine is converted by xanthine oxidase to
II. Increased excretion of uric acid 8-hydroxyadenine and 2,8-dihydroxyadenine.
A. Inborn error of membrane urate transport The latter compound can be analyzed by the
1. Hereditary renal hypouricemia methods cited previously or by high performance
B. Acquired renal hypouricemia
liquid chromatography [565, 620, 621]. Affected
1. Inappropriate ADH secretion
individuals vary in their capacity to solubi-
2. Multiple myeloma
lize 2,8-dihydroxyadenine in their urine, and in
3. Lymphomas (Hodgkin’s disease)
some, the urine may be supersaturated with this
4. Pulmonary neoplasms
C. Fanconi syndrome
metabolite. An increased capacity to solubilize
1. Idiopathic this metabolite may explain the asymptomatic
2. Wilson’s disease subjects with adenine phosphoribosyltransferase
3. Cystinosis deficiency.
4. Type I glycogen storage disease Treatment of this metabolic error can be
5. Galactosemia undertaken by restricting purine intake in the
6. Hereditary fructose intolerance diet, prescribing a high fluid intake, and using
7. Heavy metal intoxication allopurinol at 10 mg/kg body weight in a child or
8. Outdated tetracycline use 300 mg/day in an adult. Allopurinol dose reduc-
9. Liver disease and jaundice tion is essential in patients with renal insufficiency,
10. Neoplasms and plasma oxypurinol levels should be moni-
D. Drugs tored to avoid the toxicity of this allopurinol
1. Benzbromarone metabolite. Since 40 % of the cases of APRTase
2. Calcium ipodate
deficiency are reported in children, and such
3. Dicoumerol
affected subjects may develop chronic renal
4. Estrogens
insufficiency, it is critical to consider this disease
5. Ethyl biscoumacetate
in any child presenting with crystalluria, radiolu-
6. Glyceryl guaiacholate
7. Iodopyracet
cent stones, or renal failure. Although APRTase
8. Iopanoic acid deficiency is a rare cause of nephrolithiasis in the
9. Phenylbutazone adult, it has been identified in patients in their
10. Probenecid seventh decade.
11. Salicylates
12. Sodium diatrizoate Oxipurinol Calculi
13. Sulfinpyrazone Disorders with hyperuricosuria are often treated
14. Carbamazepine with the hypoxanthine analog, allopurinol
15. Beta-lactamases (4-hydroxypyrazolo (3,4d)-pyrimidine), to
reduce the serum and urinary uric acid levels
[557, 558]. Allopurinol not only inhibits xan-
with type II deficiency have been confined to thine oxidase activity to prevent the conversion
those of Japanese ancestry. of hypoxanthine and xanthine to uric acid, but
Laboratory analyses of affected patients dem- allopurinol itself is also converted by xanthine
onstrate that there is no alteration in serum or uri- oxidase to a less soluble metabolite, oxipurinol
nary uric acid levels. The urine contains adenine (4,6-dihydroxypyrazolo (3,4d) pyrimidine)
and two of its metabolites: 8-hydroxyadenine [622]. Since oxipurinol is an analog of xanthine,
and 2,8-dihydroxyadenine. The ratio of adenine, it shares certain of its chemical properties with
134 5 Clinical Aspects of Gout and Associated Disease States

xanthine [549]. Oxipurinol is much less soluble Hyperuricosuric Calcium Oxalate


than its parent compound, allopurinol. At 37 °C, Nephrolithiasis
allopurinol has a solubility of 0.75 mg/ml in Several additional problems may lead to confu-
water, whereas oxipurinol has a solubility of sion when the diagnosis of a renal calculus is
0.35 mg/ml under the same conditions [623]. On entertained. First, gouty subjects also have an
the basis of its relative insolubility, oxipurinol increased frequency of calcium oxalate stones
crystalluria and stone formation have been as well as other stone constituents [626, 627].
observed under certain circumstances. In one Second, other radiolucent stones (xanthine and
patient with purine overproduction who was 2,8-dihydroxyadenine) may be confused with
treated with both oxipurinol and allopurinol, nonopaque uric acid stones, and third, the use
oxipurinol lithiasis has been reported [624]. A of uricosuric agents or hypouricemic drugs
second case of oxipurinol lithiasis has been may give rise to uric acid stones or in the case
reported in a woman with a long history of of allopurinol, xanthine, or oxipurinol stones.
regional enteritis and recurrent uric acid stones Finally, hyperuricosuric calcium oxalate nephro-
[625]. This patient was initially treated with lithiasis occurs in recurrent calcium stone form-
600 mg of allopurinol, and subsequently the ers, and the increased urinary uric acid levels may
dose was increased to 900 mg a day. At a dose of lead to confusion with an abnormality in purine
300 mg of allopurinol a day, this patient excreted metabolism and gout.
in her urine 47 mg of xanthine and 243 mg of Calcium oxalate is the most common constitu-
oxipurinol over a 24-h time period. On 900 mg ent of kidney stones, and it would not be surpris-
of allopurinol, these same urinary outputs ing to find stones containing this metabolite in
increased to 145 mg of xanthine and 440 mg of patients with gout. In fact, a survey of gouty
oxipurinol for a 24-h time period. Chemical patients showed that 80 % of stones are com-
analysis of the nonopaque calculus passed by posed of pure uric acid, whereas 12 % of the
this patient documented its major component to stones contained only calcium oxalate [407].
be oxipurinol with a minor fraction being Mixed stones composed of uric acid and calcium
xanthine. oxalate have been observed as have stones com-
posed of uric acid and calcium phosphate and
Uncommon Cause of Nonopaque Stones uric acid and magnesium ammonium phosphate.
One uncommon cause of uric acid calculi and In calcium stone formers, there is a poorly under-
non-urate radiopaque stones deserves mention. stood link between uric acid excretion and stone
Indinavir sulfate, an effective protease inhibitor formation in that the higher the urinary uric acid
of human immunodeficiency virus type 1, has excretion, the more likely stone formation will
been recognized as a cause of radiolucent stone occur. If the data related to calcium stone formers
formation [626–629]. These calculi, like uric acid are analyzed roughly 26 % manifest hyperurico-
stones, are not visible on plain radiographs, and suria and 12 % have hypercalciuria [628].
CT scans are also nondiagnostic [626]. Excretory Analyses of such patients show that in part, the
urograms and/or renal ultrasound must be used to increased urinary uric acid excretion in affected
visualize these stones. Indinavir stones are individuals is related to a high purine intake;
observed in about 10–20 % of patients taking this however, in other cases, the urinary uric acid
medication, and a history of AIDS is an impor- excretion remained at higher than normal levels
tant historical clue to the possibility of such a even after 7 days on a purine-free diet [629].
stone. Infrared spectrophotometry provides a Thus, both excessive dietary purines and uric acid
structural analysis of indinavir calculi that is overproduction from endogenous purine synthe-
compatible with indinavir monohydrate [627]. sis play a role in the generation of increased uri-
Some reports have indicated that HIV-positive nary uric acid levels. Further, patients with
hemophiliacs are at increased risk for indinavir- hyperuricosuric calcium oxalate nephrolithiasis
associated nephrolithiasis [629]. have more serious disease as measured by the
Drug-Induced Hyperuricemia and Gout 135

rate of cystoscopy and other surgical procedures, induced by drugs is by an alteration in the renal
and it appears that their stone formation peaks at tubular excretion of urate, but some drugs are
age 40–45 in contrast to other stone formers known to increase the formation of uric acid. The
whose disease peaks at age 25–30 years [630]. It most important drugs causing hyperuricemia
appears to be a predominantly a disease of males include diuretics, salicylates, pyrazinamide, nic-
which may account for the increased intake of otinic acid, ethambutol, ethanol, fructose, cyto-
purines in the diet. The most critical evidence toxic agents, and cyclosporine.
linking uric acid metabolism with recurrent cal-
cium oxalate stone formation is the capacity of
allopurinol to reduce new stone formation [628, Diuretics
631–634]. Since allopurinol has no effect on cal-
cium oxalate crystal growth or nucleation, these Diuretics represent a significant cause of hyperu-
studies conclude that the mechanism of allopurinol ricemia and gout because of their common usage
action must be through a reduction in urate excre- in clinical therapeutics. The most frequently
tion. Further, in those patients who have hyper- implicated agents are furosemide, ethacrynic
calciuria in association with hyperuricosuria, the acid, and benzothiadiazines. Some diuretics such
use of allopurinol together with thiazide treat- as spironolactone, amiloride, triamterene, and
ment has been shown to be effective in reducing organomercurials do not result in hyperuricemia.
stone formation. For patients who cannot tolerate Two primary mechanisms contribute to the hype-
allopurinol, it has been shown that potassium cit- ruricemic response: volume contraction of the
rate therapy reduces the rate of stone recurrence extracellular fluid leading to an increase in solute
[635]. The rationale for this treatment is the fact (urate) reabsorption or decreased tubular urate
that citrate lowers calcium oxalate supersatura- secretion [261, 639–644].
tion and may block urate-induced crystallization The clinical features of diuretic-induced gout
of calcium oxalate [636]. The most compelling do not differ from other causes of gout, but epide-
issue related to this disorder is at what level of miological studies have characterized diuretics as a
urinary uric acid should treatment be initiated. risk factor for the development of gout [645–653].
Urinary uric acid levels of 800 mg/day or greater Diuretics have also been documented as a
in men and levels of 600 mg/day in women are significant contributing factor to gouty arthritis in
probably reasonable measurement points when females [645, 647–650, 653]. Most females who
allopurinol therapy should be considered. If have gout are postmenopausal, and many have an
dietary indiscretions are suspected as contribu- underlying disease such as renal insufficiency and/
tors to the disease process, suggesting a reduction or hypertension. Both females and males who
in high purine-containing foods might be useful. develop gout may be receiving diuretics and
There have been studies to suggest that uric cyclosporine after kidney transplantation. In con-
acid promotes the crystallization of calcium trast to the increased frequency of diuretic use in
oxalate from urine, but these studies have been females who develop gout, males most often have
refuted by others [637, 638]. Thus, the relation- chronic alcohol abuse as an associated condition
ship between uric acid and calcium oxalate stone [652]. In those females who develop gout in the
formation remains unresolved. premenopausal state, anorexia nervosa may be the
underlying disease and may be associated with
diuretic abuse [646]. One must recognize that in
Drug-Induced Hyperuricemia this setting, the patient may try to deceive the phy-
and Gout sician by denying the use of diuretics or other drugs
to lose weight. Postmenopausal diuretic-induced
A wide variety of pharmacologic agents can gout occurs frequently at age 50 or above, whereas
cause hyperuricemia and, in some, gouty arthri- gout observed in association with renal allografts
tis. The principal mechanism of hyperuricemia is most often seen in younger patients. Thus, in
136 5 Clinical Aspects of Gout and Associated Disease States

diuretic-induced gout, there is often another medi- occlusion, there does not appear to be a significant
cal condition that results in the impairment of urate increase in the frequency of gouty arthritis in this
excretion. If necessary, diuretic-induced hyperuri- population.
cemia can easily be treated and controlled with
uricosuric agents [654]. It is not reasonable to treat
diuretic-induced hyperuricemia unless there is a Ethanol
rationale for such treatment such as recurrent acute
gouty arthritis or renal insufficiency. If for some Chronic alcohol use, hyperuricemia, and gout
reason uricosuric agents are contraindicated, have been known to be interrelated since the rec-
allopurinol can be used [655, 656]. ognition of gout as a metabolic disease. The cause
for ethanol-induced hyperuricemia is multifacto-
rial. Hyperlacticacidemia leading to a suppres-
Salicylates sion of renal urate excretion, fasting causing
ketonemia and urate retention, increased muscle
Salicylates have a paradoxical effect on the han- activity causing lactate production, and the inges-
dling of uric acid by the kidney. At doses of less tion beer with its high purine content may all
than 2.5 g/day, salicylates cause the retention of contribute to the hyperuricemic state induced by
uric acid, whereas at doses greater than this, they alcohol [662–665]. Despite the frequent associa-
are uricosuric [657–659]. In fact, salicylates at tion of alcohol consumption and hyperuricemia,
high doses were used as uricosurics prior to the alcohol use infrequently results in gouty arthritis
marketing and use of probenecid/Benemid [660]. [647, 652, 653, 666].
The pathophysiological effects of salicylates on
uric acid metabolism have been investigated in
some detail. At low doses of salicylate (<1.2 g/ Cytotoxic Agents
day), serum uric acid levels are increased by 1.0–
2.0 mg/dl [659]. Uric acid retention has been Therapeutic agents that lead to the destruction of
shown to have its maximal effect at plasma sali- cells will routinely cause hyperuricemia unless
cylate concentrations of 5 mg/dl, whereas urico- the host is protected by allopurinol prior to treat-
suria occurs at plasma concentrations greater ment with such cytotoxic agents. The neoplastic
than 10 mg/dl [262]. The most precise measure- diseases prone to this complication have been
ment for assessing the effect of salicylate on the discussed previously in the section “Acute uric
renal tubular handling of urate has been shown to acid nephropathy.” These cytotoxic agents cause
be the concentration of free salicylate in the renal the release of cellular nucleic acids and purine
tubular urine [262]. An excretion rate of free sali- nucleotides that are subsequently metabolized to
cylate of less than 0.5 mg/min is uricoretentive uric acid. The latter metabolite, if produced in
whereas at rates above this level, salicylates are sufficient quantity, may markedly increase serum
uricosuric. It has been proposed that low-dose uric acid levels and may also cause acute uric
salicylates block the renal tubular secretion of acid nephropathy. The onset of gout may coin-
urate whereas at high doses, it inhibits both renal cide with the initiation of chemotherapy or radia-
tubular secretion and reabsorption. In addition to tion therapy [530]. As discussed in the Chap. 4,
these renal tubular effects of salicylates, it is “Nephrolithiasis,” renal calculi are not uncom-
important to recognize that salicylates at any dose mon in patients with gout secondary to myelo-
abrogate the uricosuric effects of probenecid and proliferative disorders.
sulfinpyrazone [661].
Even though salicylates may increase serum
urate concentrations, this effect does not appear Allopurinol-Induced Gout
to be a significant precipitating factor for gout. If
one considers the large number of elderly adults It is not uncommon for an acute gouty episode to
consuming low-dose aspirin to prevent vascular occur in the course of the initiation of allopurinol
Drug-Induced Hyperuricemia and Gout 137

treatment. The precipitation of acute gout in this The exact mechanism for the hyperuricemic effect
setting is the result of a rapid change in the serum caused by ethambutol has yet to be determined
uric acid concentration along with a dramatic [678]. It has been established that hyperlacticaci-
alteration in the uric acid pool. Gouty attacks demia and volume contraction do not explain its
have been reported to occur with serum uric acid hyperuricemic effect. The increment in serum
levels as low as 2 mg/dl in association with a uric acid concentration and the decrease in renal
significantly decreased urinary uric acid excre- clearance of uric acid occur by mechanisms that
tion [558]. This fact serves to emphasize that an are different from similar changes in uric acid
elevated serum uric acid is not necessary for the metabolism induced by diuretics, ethanol, pyrazi-
clinician to entertain the diagnosis of acute gouty namide, and acetylsalicylic acid [678].
arthritis. In fact, either a rapid increase or decrease
in the serum uric acid level may trigger an attack
of acute gout. Since allopurinol may cause acute Cyclosporine
gout, it is reasonable to make the patient aware of
this possibility and to use lower doses of Since the early 1960s, hyperuricemia and gout
allopurinol at the start of therapy while progress- have been observed as a complication of
ing to the full dose of 300 mg/day. The use of cyclosporine therapy for immunosuppression
colchicine in low dosage (0.5–1.0 mg/day) may after organ transplantation [679–683]. In clinical
also prevent acute gouty episodes during the ini- studies of cyclosporine therapy, hyperuricemia
tiation of hypouricemic therapy. occurs in 80–90 % of patients being treated with
this immunosuppressant, and roughly 10–20 %
of these patients develop acute gouty arthritis
Antituberculous Drugs [16, 684–686]. As discussed previously in the
discussion of chronic tophaceous gout,
The resurgence of tuberculosis emphasizes the cyclosporine-induced gout is an accelerated form
importance of understanding the role of ethamb- of gout with tophi occurring over a relatively
utol and pyrazinamide in altering the renal han- short time period and in unusual sites [14, 684].
dling of uric acid. Both these drugs continue to be Clearly, cyclosporine causes a decrease in
used in the treatment of tuberculosis. Pyrazinamide renal urate clearance, but the exact mechanism for
has been recognized as a potent uricoretentive this decrement remains incompletely understood
drug since 1956 [667]. Not only does this drug and may be a multifactorial cause. Alterations
cause hyperuricemia via a renal mechanism, but in glomerular filtration rates, renal tubular func-
it may also induce acute gouty attacks [667–669]. tions, or renal vasculature all have been impli-
The mechanism by which pyrazinamide alters cated in the nephrotoxicity of this drug leading
uric acid metabolism is thought to be by impair- to hyperuricemia [15, 687–691]. Hypertension,
ing both secretion and reabsorption with the net renal insufficiency, and diuretic use may also
result being a reduction in the renal clearance of contribute to the changes in uric acid metabolism
uric acid [257, 670, 671]. From these and other caused by cyclosporine. Tophaceous deposits
studies, it became clear that the deaminated should be sought in unusual locations including
pyrazinamide metabolite, pyrazinoic acid, was soft tissues, intraspinal sites, and the spine itself
the primary mediator of the hyperuricemic effect [18, 692, 693].
[671, 672]. Pyrazinamide-induced hyperuricemia The management of cyclosporine-induced gout
does not respond to uricosuric therapy, but it can in transplant recipients is complex and difficult.
easily be controlled with allopurinol [672–674]. Allopurinol prevents the accumulation of uric
Acetylsalicylic acid at low dosage and p-amin- acid, but its use in patients receiving 6-mercap-
osalicylic acid can reverse the uricoretentive topurine or its derivatives causes the accumulation
effects of pyrazinamide [675, 676]. of 6-mercaptopurine by preventing its metabolism
Subsequent to these observations, ethambutol via xanthine oxidase to an innocuous metabolite,
was also shown to cause hyperuricemia [677]. thiouric acid [694]. The accumulation of either
138 5 Clinical Aspects of Gout and Associated Disease States

6-mercaptopurine or azathioprine potentiates the probenecid [703]. Like other uricosuric agents,
suppression of the myeloid cells in the bone mar- the uricosuric effect of benzbromarone is antago-
row [19]. Another complication of allopurinol nized by pyrazinamide and salicylates, but the
therapy is that renal insufficiency leads to the salicylate effect does not completely abrogate the
accumulation of allopurinol itself, and this drug uricosuric effect [704]. Benziodarone, the iodine-
is relatively insoluble at high concentrations. containing uricosuric drug, is used infrequently
Reduction in azathioprine or 6-mercaptopurine since it alters human thyroid function [705].
dosages to accommodate the use of allopurinol Another less commonly used drug to counteract
in the treatment of cyclosporine-induced hype- cyclosporine-induced hyperuricemia is the enzyme,
ruricemia may reduce their immunosuppressive urate oxidase, derived from a variety of extracts
effects leading to graft rejection [695, 696]. including Aspergillus flavus, Arthrobacter proto-
Even though no absolute methods for the man- formiae, and Candida utilis [706–710]. Although
agement of cyclosporine-induced gout have been the initial studies of this enzyme used purified
determined, several alternative drugs have been urate oxidase, subsequent studies used urate oxi-
used with some success. ACTH injections have dase covalently attached to polyethylene glycol to
been used, but they have a tendency to induce reduce antibody formation in the host [711–713].
rebound attacks of gout after the effect of injec- At the present time, a recombinant urate oxidase is
tions have ceased. The inosine dehydrogenase being tested and may be made available soon for a
inhibitor, mycophenolate, has been used in place wider distribution and use [714].
of azathioprine in kidney transplant recipients The mechanism by which this enzyme acts is
with a resultant modest reduction in the hyperuri- through the conversion of uric acid to the highly
cemia and the capacity to use full doses of water-soluble metabolite, allantoin and carbon
allopurinol [697]. dioxide. This mode of therapy requires an intrave-
Probably the most effective agent for the man- nous or intramuscular injection and is useful in
agement of cyclosporine-induced hyperuricemia the treatment and pretreatment of patients at risk
and gout is the uricosuric drug, benzbromarone, for the tumor lysis syndrome [715, 716]. Even
since it is effective in those patients with renal though in most cases urate oxidase therapy is a
insufficiency [698]. There are two disadvantages safe and efficient means of treating cyclosporine-
to the use of this drug. First, it is not available induced hyperuricemia and gout as well as other
on a general basis in the United Kingdom or causes of gout, it is not without side effects.
the United States, and second, it is ineffective if Anaphylactoid reactions, hypersensitivity skin
the creatinine clearance is less than 20 ml/min. rashes, bronchospasm, and hemolytic anemia
Several publications have attested to the efficacy have been reported with its use. Additional studies
of benzbromarone or its iodinated counterpart, may deem this agent as an attractive option for the
benziodarone, in the treatment of cyclosporine- management of severe gouty arthritis in patients
induced hyperuricemia and gout [15, 23, 699, with renal insufficiency, in those at risk for tumor
700]. Both these drugs were effective in normal- lysis syndrome, and in transplant recipients.
izing serum uric acid levels without significant One additional issue related to transplant
complications. In this small number of published recipients and gout needs to be discussed. In
reports (less than 100 patients), a few patients some patients, maintenance colchicine therapy
have developed uric acid calculi of the kidney, has been used to prevent the recurrence of gouty
and one case of drug-induced hepatitis requir- attacks or to abrogate the onset of gout in the face
ing liver transplantation has been reported after of treatments known to trigger acute gout. Such
benzarone use [699, 701, 702]. Less serious regimens are often not the best way to prevent
side effects of the drug include gastrointesti- acute gout in renal transplant recipients since
nal disturbances and hypersensitivity rashes. In colchicine often induces a myopathy [20, 21,
comparison to probenecid, benzbromarone in a 717]. The onset of muscle weakness (usually
dose of 100 mg/day is equivalent to 1.0 g/day of painless), distal sensory changes, and elevated
Drug-Induced Hyperuricemia and Gout 139

creatine phosphokinase levels is especially likely of uric acid, some investigators have proposed
to occur in patients with renal insufficiency that nicotinic acid and its amide derivative, nico-
treated with colchicine and cyclosporine [20, 22]. tinamide, may also increase purine biosynthesis
The measurement of creatinine clearance has [727–729]. Other evidence has been presented to
been proposed as a means of analyzing the risk of document a decrease in phosphoribosylpyrophos-
colchicine myotoxicity [718]. This investigation phate levels associated with nicotinic acid admin-
determined that patients at the highest risk for istration [721, 730].
colchicine-induced myotoxicity had a creatinine In summary, nicotinic acid clearly induces
clearance of less than 50 ml/min. hyperuricemia and may occasionally precipitate
Thus, cyclosporine-induced hyperuricemia an attack of acute gout. The present literature
and gout occur frequently and are difficult to treat suggests that it should not be commonly used
since the pharmacologic options may interfere since effective drugs such as the statins and fibric
with the therapy to prevent graft rejection. It acid derivatives are available.
appears that newer agents available in Europe
such as benzbromarone and urate oxidase may
offer safer and more effective therapy for this Fructose-Induced Hyperuricemia
complicated drug-induced disease.
Fructose-induced hyperuricemia is of significance
since its mechanism of action on uric acid metab-
Nicotinic Acid olism is well characterized, and the possible
connection between fructose ingestion, hyperu-
Nicotinic acid has been used as a hypolipidemic ricemia, and gout in the heterozygous carriers of
drug since it reduces the levels of plasma total phosphofructoaldolase deficiency [731–736]. The
cholesterol, plasma triglyceride, and very low initial report of the effects of intravenous fruc-
density lipoprotein cholesterol and increases the tose clearly demonstrated that 0.5 g of fructose
high density lipoprotein cholesterol level [719]. per kilogram of body weight results in hyperuri-
Unfortunately, in patients with type II diabetes cemia and hyperuricosuria both in patients with
mellitus, it increases insulin resistance and blood hereditary fructose intolerance and in normal
sugar concentrations [720]. In addition to this children [734]. Subsequent reports have shown
relative contraindication to its clinical use, it also that 200 mg of fructose/kg body weight admin-
causes flushing, gastric irritation, hepatotoxicity, istered intravenously also induces hyperuricemia
and hyperuricemia [721–725]. The primary effect in healthy children and adults [733]. Even doses
of this agent on uric acid metabolism is to of fructose as low as 160 mg/kg body weight/h
decrease the clearance of uric acid by the kidney will cause hyperuricemia in critically ill patients
[721, 726]. In an attempt to localize the site of [737]. Other investigators have demonstrated that
action of nicotinic acid on the renal tubular han- measurements of urinary uric acid as a function
dling of uric acid and characterize any drug-drug of urinary creatinine (mg of uric acid per mg
interactions, the effect of nicotinic acid on creatinine) may be a more precise indicator of
sulfinpyrazone, iopanoic acid, and acetylsalicylic the changes in uric acid metabolism induced by
acid has been evaluated [721]. Acetylsalicylic fructose than serum uric acid levels per se [738,
acid does not reverse the nicotinic acid-induced 739]. In addition to increased urinary uric acid
hyperuricemia in contrast to its reversal of pyrazi- concentrations, fructose also induces an increase
noic acid’s effect on the kidney. Nicotinic acid in uric acid precursors (inosine, hypoxanthine,
inhibits the uricosuric effects of sulfinpyrazone and xanthine) in the urine [735, 740, 741]. The
and iopanoic acid. The latter action indicates the association of the heterozygous state for aldolase
necessity for allopurinol use if nicotinic acid-in- B (fructose 1,6-bisphosphate aldolase) deficiency
duced hyperuricemia needs to be treated. In addi- and gout is discussed in detail in the chapter on
tion to the alteration in the renal tubular handling genetics. Suffice it to say that some evidence sug-
140 5 Clinical Aspects of Gout and Associated Disease States

gests that familial gout may be associated with the AMP is converted by the same nucleotidase
carriers of the mutant gene for hereditary fructose to adenosine that is subsequently converted to
intolerance since a number of such heterozygotes inosine by adenosine deaminase. Inosine is then
were shown to have gouty arthritis. converted to hypoxanthine by nucleoside phos-
The mechanism for the increments in serum phorylase. Xanthine oxidase acts on the latter
and urinary uric acid in hereditary fructose intol- substrate and its immediate product, xanthine, to
erance is via the degradation of adenine nucle- convert these metabolites to uric acid. These reac-
otides to uric acid [742, 743]. Studies of the tions are significantly enhanced by the release of
liver after fructose infusions have demonstrated AMP deaminase inhibition by the fall in inor-
a decrease in adenosine triphosphate (ATP), a ganic phosphorus levels and GTP concentrations
decrease in inorganic phosphorus, and an increase induced by fructose infusions [749]. As a result
in fructose-1-phosphate. The decrease in ATP is of the catabolism of adenine nucleotides and the
the result of its use in the conversion of fructose decreased concentration of nucleotides causing
to fructose-1-phosphate, and the decrease in inor- feedback inhibition of the enzyme, phosphoribo-
ganic phosphorus results from the rephosphory- sylpyrophosphate (PRPP) amidotransferase, the
lation of ADP in the mitochondria. In hereditary rate-limiting enzyme in de novo purine biosynthe-
fructose intolerance, the accumulation of fruc- sis, the rate of purine synthesis is also increased
tose-1-phosphate results from the absence of [753, 754]. In isolated rat hepatocytes, fructose
the enzyme, aldolase B, and the inability to split induces a transient (60 min) increase in PRPP
fructose-1-phosphate into d-glyceraldehyde and and ribose-5-phosphate concentrations [755].
dihydroxyacetone phosphate. These changes in Although there is speculation that this change
the liver have been documented by 31P magnetic relates to an increased PRPP synthetase activity,
resonance spectroscopy [731, 732, 744–746]. the complete mechanism has as yet to be defined.
These changes in metabolite concentrations are Another contributor to fructose-induced hyperu-
diminished in the presence of hepatocellular ricemia is the elevated blood lactate concentra-
disease as a result of the decreased capacity to tions observed after fructose infusions. The latter
phosphorylate fructose [747, 748]. In comparing metabolite blocks urate excretion and results in
patients with chronic hepatitis and healthy adults, hyperuricemia [265, 739, 756].
the alterations in phosphomonoesters in healthy In summary, fructose infusion triggers the
adults after the infusion of 0.5 g of fructose/kg catabolism of adenine nucleotides that lead to the
body weight increased in 15 min to 338 ± 76 % generation of uric acid. This metabolic change is
of the preadministration value in contrast to an especially prominent in patients with hereditary
identical analysis of patients with chronic hepa- fructose intolerance. Further, such metabolic
titis where the increase was only 151 ± 49 % of changes may be a factor in the association of gout
the preadministered value. In addition to the with patients who carry the mutant gene for
decrease in ATP concentration after fructose infu- hereditary fructose intolerance.
sions, the concentration of GTP also decreases
[749]. A similar depletion of high-energy phos-
phate also occurs after fructose infusion in the Megaloblastic Anemia
kidney and small intestine [750–752]. Thus,
the fructose-induced catabolism of ATP, ADP, Hyperuricemia, hyperuricosuria, and acute gouty
and AMP begins by the conversion of fructose arthritis have been observed during the treatment
to fructose-1-phosphate with the generation of of megaloblastic anemias [757–760]. A variety
ADP. Adenosine diphosphate is then converted to of markers of increased purine production have
ATP by adenylate kinase with the generation of been investigated in patients during vitamin
AMP. The latter metabolite is converted to inos- replacement therapy in pernicious anemia, and
ine monophosphate by AMP deaminase. Then, the increase in purines appears to be correlated
inosine monophosphate is converted to inosine with the peak reticulocyte count on days 5–7 of
by cytoplasmic 5¢-nucleotidase, or alternatively, the treatment program [761].
Saturnine Gout (Lead-Induced Gout) 141

The mechanism of these changes in uric acid examples of lead-induced gout or so-called satur-
metabolism is related in part to the increased nine gout have been reported from Queensland,
synthesis of purine nucleotides via the elevated Australia, and the southern part of the United
activity of adenine phosphoribosyltransferase, States [769, 770]. The sources of lead in these
hypoxanthine-guanine phosphoribosyltrans- two examples are quite different, but they empha-
ferase, and PRPP synthetase as well as the increase size an unusual source of lead in one case and a
in the urinary excretion of aminoimidazolecar- long delay in the expression of gout in the other.
boxamide, an intermediate in the de novo purine In semitropical Queensland, the lead source was
biosynthetic pathway, and increased levels of uri- fences painted with a lead-based paint in proxim-
nary oxypurines [761–763]. Another somewhat ity to children’s play areas. Lead intoxication
confusing observation is the occurrence of mac- resulted from the ingestion of flakes of lead from
rocytes and megaloblastosis in the bone marrow the paint. In the southern part of the United States,
of patients with the Lesch-Nyhan syndrome and the source of the lead was the solder and old car
the Kelley-Seegmiller syndrome [72, 764–768]. radiators used in the construction of illicit stills
The increased rate of de novo purine biosynthe- for the purpose of distilling and refining alcohol.
sis and the reduction in the serum folate concen- The product of this distillation process has been
trations due to its increased usage to form the given the name moonshine, and the gouty arthri-
purine ring undoubtedly contribute to the mega- tis associated with the use of this product has
loblastosis, but the megaloblastic anemia appears been called Dixie gout. Lead-containing paints
not to respond to folate therapy [767, 768]. Why are still in use today, but the primary exposures to
these divergent facts occur remains puzzling, but lead-containing paint occur most frequently in
factors other than folate must play a role in the rundown inner city housing where children are
megaloblastic anemia of some Lesch-Nyhan syn- exposed to the flakes of the old paint that contain
drome patients. lead [771]. Other potential exposures to lead in
children include parental sources of lead from
employment in an industry using lead and living
Summary of Drug-Induced near industrial sites that emit lead [772–774].
Hyperuricemia and Gout Industrial lead exposures occur mainly in the
manufacture of automobile batteries, the produc-
From the foregoing facts, it is clear that many tion of alloys like solder, manufacture of plastics
pharmacologic agents can cause hyperuricemia, and ammunition, scrap metal salvaging, lead
and in some patients, several of the drugs dis- smelting, and the manufacture and use of various
cussed may also precipitate attacks of acute gouty lead-containing pigments. A complete list of haz-
arthritis. The marked overuse of allopurinol for ardous exposure to lead in industrial settings has
the treatment of hyperuricemia and the clinical been published [775].
myth that all hyperuricemic patients need to be In the southern part of the United States, ele-
treated should lead the clinician to consider drug- vated blood lead levels have been documented
induced causes of hyperuricemia and gout. The in individuals drinking illegally distilled alcohol
use of alternative pharmacologic agents and the [776–784]. This modern-day association
control of alcohol intake could reduce the inci- between gout and illicitly distilled alcohol also
dence of these abnormalities in purine has a counterpart in history since fortified
metabolism. English wines from the years of 1770 and 1820
contained lead concentrations of 320–1,900
ug/l, values above allowable lead concentrations
Saturnine Gout (Lead-Induced Gout) [784]. These findings may reflect the many car-
toons from the era depicting Englishmen with
The association between lead exposure and their bandaged foot on a footstool with a bottle
the occurrence of gout has been documented of wine on a nearby table. Lead-tainted bever-
from the early 1800s. The clearest documented ages date back centuries since Sir George Baker
142 5 Clinical Aspects of Gout and Associated Disease States

first reported lead colic from the ingestion of disease in Queensland. In Queensland, the kid-
contaminated cider in 1767 [785]. Such events ney disease precedes the onset of gout, whereas
continue to be recorded in the literature [786, renal disease usually does not predate the gout in
787]. It has been suggested that the popularity the southern part of the United States. Dixie gout
of so-called moonshine whiskey is its low cost affects primarily black males from rural areas,
to the consumer, but the major reason for its use whereas in Queensland gout, there is almost an
is the sweet taste imparted by the lead, which equal distribution between the sexes, and the
attracts these advocates of home brew. Thus, in females with gout often contract their disease
the southern regions of the United States and prior to the menopause. In addition, there is no
elsewhere, solder used in homemade stills, lead- significant history of alcohol use in the Queensland
glazed receptacles used to process grapes, and patients [794–796].
even lead crystal wine glasses can be ready To date, there remains a degree of controversy
sources of lead and lead poisoning [788, 789]. regarding the role of lead in the generation of kid-
Other potential exposures that might cause lead ney disease. In all the cases of saturnine gout, the
poisoning include exposures occurring in con- following characteristics have been documented.
struction workers, deleaders, house painters, The serum uric acid levels usually exceed those
firing range users, and do-it-yourself home ren- observed in other forms of chronic disease [770,
ovators [790]. Indeed, in the 1800s, Garrod 792]. The renal clearance of uric acid is reduced
observed that 25 % of his patients with gout in saturnine gout, and some evidence has been
were either painters or artisans who had lead presented to suggest that this change is the result
poisoning [791]. of increased renal tubular reabsorption [792].
From a clinical standpoint, Queensland and Prevailing published data indicate that the renal
Dixie (moonshine) gout present with somewhat disease in Dixie gout is less severe than the
different clinical findings. Chronic lead nephrop- kidney disease observed in Queensland gout.
athy is a characteristic feature of Queensland Tophaceous deposits have reported to be unusual
gout, and death from chronic renal failure was a in saturnine gout, but other studies have not
common occurrence in Queensland in the early confirmed this finding [795, 797]. Although lead
1900s [769]. The studies of this Australian type nephropathy exists as an entity with nonspecific
of lead intoxication demonstrated a clear associa- renal pathology, its pathogenesis and associated
tion between contracted granular kidneys found clinical findings remain incompletely resolved;
at autopsy in affected patients and an increased additional work may resolve the remaining
lead concentration in bone. This finding offered dilemmas [798–814].
suggestive proof that lead was the cause of the No consistent historical feature or clinical
nephropathy. Subsequently, the use of EDTA finding except for the history of lead exposure is
chelation of lead demonstrated an increased lead characteristic of saturnine gout. Since the most
excretion in the urine of these patients, and recent studies of gout document a marked vari-
despite the absence of any symptoms or signs of ability in the occurrence of lead-induced gout, it
recent lead exposure, these results documented is unclear from the literature when gouty patients
the past exposure of these patients to lead [792]. should be screened for this disorder [782, 795,
Of those with chronic lead nephropathy and no 803–805, 815, 816]. It is reasonable to ask every
persistent signs of lead poisoning, 50 % devel- patient with gout about exposure to lead in the
oped gouty arthritis [793]. In contrast to these home or at work. If a positive history of lead
findings, a history of childhood lead intoxication exposure is obtained, then further investigation of
leading to chronic renal disease and ultimately the body stores of lead is indicated. Patients with
death from the kidney disease has not been renal disease of no apparent cause and gout cer-
observed in children with lead poisoning in the tainly require a careful evaluation for the pres-
United States. This leads to the idea that there ence of underlying lead intoxication as a cause
may be additional factors contributing to the renal for the abnormal kidney findings.
Saturnine Gout (Lead-Induced Gout) 143

Laboratory evaluations of patients at risk for of lead. The excretion of 500–600 ug of lead in a
lead exposure should include analyses for the 24-h urine collection in a lead-free container is
detection of both acute and chronic lead intoxica- considered abnormal [793, 795]. There are ques-
tion. The total body burden of lead is distributed tions concerning the effect of the EDTA mobili-
throughout the body and includes a rapidly zation test as to the deposition of the mobilized
exchangeable pool in the blood and a more stable lead in the human [821–823]. Even though EDTA
pool in the skeleton [817]. The rapidly exchange- mobilization tests continue to be used, the possi-
able blood pool of lead represents only 2 % of the ble redistribution of lead to the brain during such
total body burden of lead, whereas the skeletal tests has raised issues about the overall safety of
pool stores more than 90 % of the body lead in its this procedure, and newer pharmacologic agents
stable pool. Thus, blood lead concentrations mea- for mobilizing lead and X-ray fluorescence are
sure the lead recently absorbed by the body, and likely to replace this older test for the measure-
such concentrations do not reflect the total body ment of total body lead burden. A number of pub-
burden of lead. Measurements of skeletal lead lished investigations of K x-ray fluorescence have
concentrations provide an assessment of cumula- documented the use of this technology for the
tive lead absorption from past exposures. Blood assessment of bone lead accumulation [799, 824–
lead concentrations and zinc protoporphyrin con- 831]. Noninvasive X-ray fluorescence appears to
centrations are the two most commonly used tests be an accurate method for determining cumula-
for biologic monitoring and to measure current tive lead exposure and may be further refined to
lead absorption [818–820]. Presently, a blood assess future health effects of the stable pool of
lead level of <40 ug/dl is the standard for protect- lead in bone.
ing workers from excessive lead exposure even Finally, once lead intoxication has been
though most adults have a blood lead level of <15 detected or the total body burden of lead is found
ug/dl and many have levels of <10 ug/dl. It is to be elevated, the issue becomes the treatment of
important to recognize that lead-induced neph- this condition. The key to any treatment program
ropathy and saturnine gout do not occur after is to remove the individual from continued expo-
short-term exposures to lead but are caused by sure to lead. This may obviate the need to take
chronic, long-term exposures. The other tests any further steps to remove lead from the body.
used by some to monitor exposure are the free Nonetheless, this initial therapeutic step may be
erythrocyte porphyrin (FEP) or zinc protopor- difficult to accomplish in moonshine users, but as
phyrin (ZPP) levels. In adults, the FEP or ZPP an alternative in this population of patients, the
levels are usually less than 35 ug/dl. Levels higher continuous monitoring of body stores of lead
than this are usually correlated with blood lead may be easier to accomplish. Once the individual
levels of greater than 50 ug/dl. Since elevated has been removed from lead exposure, chelation
ZPP levels are also observed in iron deficiency therapy may be necessary to reduce the body bur-
anemia, they are not specific for lead exposure. den of lead. The need for chelation therapy is
The anemia of lead poisoning is normocytic, nor- based on the blood lead concentration, the total
mochromic, whereas iron deficiency anemia is body burden of lead, clinical symptoms, labora-
usually a macrocytic, hypochromic anemia. tory abnormalities, and the duration of the expo-
Collections of blood for lead analyses require sure. Urinary excretion of lead should be
special lead-free glass tubes. measured before and after chelation therapy to
In cases where lead intoxication and saturnine assess the quantity of lead released from the body
gout are suspected, it is necessary to determine stores. In addition to EDTA as a chelating agent,
the total body burden of lead. Two methods have new, nontoxic drugs are now available that can be
been used to make this determination: EDTA administered by mouth [832–850]. The dose of
mobilization and K x-ray fluorescence. In the these newer agents for both adults and children
case of the EDTA mobilization test, the total body are provided in the various published studies
burden of lead is assessed as the urinary excretion [832, 834–840, 844, 847]. While chelation is
144 5 Clinical Aspects of Gout and Associated Disease States

ongoing, the patient should be carefully moni- in Gaucher disease must await additional docu-
tored for potential side effects of drugs used in mentation of this association and more detailed
the treatment protocol. study of the affected individuals in this report.
In summary, saturnine gout with or without
nephropathy may be a cause of gouty arthritis,
especially in moonshine users and in patients Gout in Females
from industrial settings where lead is being used.
Lead-associated gout is not a common cause of Hyperuricemia and gout are unusual in females,
gout, but in specific patients, it is valuable to ask and when these clinical and laboratory findings
about exposure to lead. The suspicion of satur- are observed in women, they deserve special
nine gout demands specific testing for the body attention in order to define any underlying condi-
burden of lead and treatment plans to reduce this tion. In population studies in the United States
burden. elsewhere, the percentage of females with gout
varies from publication to publication, but on the
average, females represent about 5–10 % of any
Gaucher Disease (Glucocerebrosidase large series of gouty patients [125, 871–877]. For
Deficiency) purposes of this discussion, it is useful to classify
hyperuricemia and gout in the female into four
Gaucher disease, a lysosomal glycolipid storage categories: juvenile, premenopausal, pregnancy,
disorder resulting from mutations in the lyso- and postmenopausal forms.
somal hydolase, has recently been shown to be
associated with gouty arthritis [851]. Since the
actual cause of the defect in purine metabolism Juvenile Hyperuricemia and Gout
remains unresolved at this time, the reason for
this abnormality in uric acid metabolism cannot Juvenile gout was reviewed in the 1970s prior to
be classified into an overproduction or underex- the complete assessment of some genetic disor-
cretion defect. In the past, Gaucher disease has ders like familial juvenile gouty nephropathy
been reported in association with gout when it (FJGN), so the small group of females reported,
was observed in the presence of a myeloprolifer- two of whom had renal disease, are hard to char-
ative disorder [125, 852]. The occurrence of gout, acterize metabolically by today’s standards [352].
Gaucher disease, and lymphoproliferative disor- Subsequent to this review, a number of clinical
ders is consistent with the apparent increased disorders have been documented that must be
frequency of such lymphoproliferative disorders considered in juveniles who develop gout.
in Gaucher disease. Even though there are more Unfortunately, the frequency of these rare forms
than 100 citations in the literature documenting of hyperuricemia or gout has not been determined
the association between Gaucher disease and since the total numbers of affected females are
malignancies, proof of the significance of these quite small, but gout occurs very infrequently in
observations remains to be determined [853–866]. this age group. Nonetheless, two disorders,
Large splenic infarcts can give rise to an acute HGPRTase deficiency and FJGN, are known to
abdomen with metabolic acidosis and hyperuri- affect females and have been documented to
cemia, but such an occurrence would not account account for roughly 50 % of the cases referred to
for the recent report of Gaucher disease and gout the Purine Research Laboratory in London [358].
[851, 867]. As indicated by the recent report of Familial juvenile gouty nephropathy represented
Gaucher disease and gout, the Ashkenazic Jewish about 32 % of the total cases seen by this group.
population is especially prone to Gaucher disease The latter disease is important to recognize since
[868–870]. At the present time, the mechanism(s) early recognition and treatment may forestall the
for the occurrence of the recent report of gout in progression of the associated renal disease. Since
association with Gaucher disease are not known. gout is not a consistent manifestation of this dis-
Further concepts about the pathogenesis of gout order, some investigators have elected to use the
Gout in Females 145

term, familial juvenile hyperuricemic nephropa- ordered, serum creatinine and/or blood urea
thy, to describe the disease. We have elected to nitrogen must also be measured to ensure the
use the term, familial juvenile gouty nephropa- absence of renal dysfunction as the cause for the
thy, to emphasize that this disease may mimic hyperuricemia. Second, a variety of inherited dis-
other forms of familial nephropathies and search- orders may manifest hyperuricemia including
ing pedigrees for a history of gouty arthritis may glycogen storage diseases, carnitine palmitoyl-
reveal a useful marker for FJGN as the underly- transferase II deficiency, cystinuria, and fatty
ing disease. Thus, a family history of renal dis- acid oxidation deficiencies. Third, hyperurice-
ease, a low fractional excretion of uric acid, and mia secondary to an increased turnover of
hyperuricemia may suggest that the underlying nucleic acids may account for the origin of the
disease is FJGN, and a history of gout in the fam- elevated uric acid levels. Fourth, unusual causes
ily provides stronger evidence for FJGN. Such of hyperuricemia must also be considered includ-
patients may or may not be hypertensive, and ing hypothyroidism, lead nephropathy, infectious
urate crystals are not often found in kidney biop- mononucleosis, and others.
sies from such individuals. Less common herita-
ble diseases that can be associated with gout are
the autosomal recessive disorders such as glyco- Premenopausal Hyperuricemia
gen storage diseases and muscle phosphoglycer- and Gout
ate mutase deficiency as well as autosomal
dominant disorders such as autosomal dominant Since most gouty episodes occur in postmeno-
polycystic kidney disease and medullary cystic pausal females, gout is rare in the post-juvenile,
disease of the kidney. Only in very rare instances premenopausal female. As noted previously,
does the Lesch-Nyhan syndrome occur in females female carriers of HGPRTase deficiency and
when the affected female has inherited a mutated PRPP synthetase superactivity have both been
gene from each parent. Gout has been reported in reported to develop clinical gout. Indeed, exces-
female carriers of HGPRTase deficiency and sive urate production has been documented in
PRPP synthetase superactivity [82, 83, 878]. The heterozygous female carriers of HGPRTase
carriers of X-linked recessive disorders are more deficiency when sophisticated isotopic analyses
likely to express clinical gout at an age beyond of purine synthesis have been measured [879].
their juvenile years. Although hyperuricemia and Autosomal dominant nephropathies associated
gout are complications of organ transplantation with hyperuricemia and gout such as familial
in the adult, gout was not thought to be a compli- juvenile gouty nephropathy, polycystic kidney
cation of transplantation in the pediatric age disease, and medullary cystic disease of the kid-
group until recently. Gout has now been reported ney are also observed in females in this age group.
in three males and two females below the age of Drug-induced gout is also certainly seen in
18 years who underwent renal transplantation females in their premenopausal years. The most
[686]. Cyclosporine and diuretic therapy appear prominent expression of gout is observed in those
to be the causative agents in these cases. Thus, females undergoing organ transplantation and
drug-induced gout does occur in juveniles of both subsequent treatment with cyclosporine and
sexes. diuretics. Even though the use of diuretics is most
Hyperuricemia may be present in juvenile often associated with transplantation, diuretics
females, but its presence is characterized by a used for other reasons have also been observed to
large number of underlying causes, and they will trigger gout in the premenopausal female. Chronic
not be discussed in detail here. However, several lead intoxication and saturnine gout have also
points should be made concerning juvenile hype- been reported in females in this age group.
ruricemia in the female. First, the renal retention Finally, gout secondary to myeloproliferative dis-
of nitrogenous wastes due to renal disease must orders is more common in women than primary
be excluded as a cause of the hyperuricemia. gout [125, 530]. Renal calculi and tophaceous
Therefore, if an arthritis package of tests is deposits are frequent in females with blood
146 5 Clinical Aspects of Gout and Associated Disease States

dyscrasias and may antedate the first episode of unknown [896–902]. Even though the mecha-
gout [880]. This is one cause of gout in this age nism remains unknown, hyperuricemia is a use-
group that must be carefully evaluated in any ful marker for preeclampsia. In addition to
female with gout so that early intervention can preeclampsia, there are other more unusual clini-
occur if such a disorder is identified. cal settings in which hyperuricemia occurs. Acute
An unusual cause of hyperuricemia and gout fatty liver of pregnancy (Sheehan’s syndrome) is
has been reported in a few females abusing laxa- a rare complication of pregnancy associated with
tives [536]. These patients presented with gener- vomiting, abdominal pain, and hyperuricemia
alized weakness, hypokalemia, and systemic [903]. This disorder may be confused with tox-
alkalosis. Two of the patients had hyperuricemia emia of pregnancy, and early diagnosis is para-
while two had recurrent episodes of gout. This mount if one is to avoid encephalopathy, liver and
disorder is self-sustaining since laxative-induced kidney failure, and a fatal outcome. Another dis-
hypokalemia reduces intestinal motility and trig- order that may also be confused with a toxemia-
gers further laxative use and abuse. Hyperuricemia like syndrome is the occurrence of transient
also occurs with a higher frequency in certain vasopressin-resistant diabetes insipidus in asso-
ethnic groups [881–886]. Females in these ethnic ciation with elevated liver function tests,
groups may also have a higher incidence of gouty decreased renal function, and hyperuricemia
arthritis than other females of similar age. The [383]. Some of the cases with this constellation
pathogenetic mechanisms have not been com- of findings have been associated with hepatitis.
pletely defined for the alteration in uric acid The mechanism for the hyperuricemia in this dis-
metabolism in these ethnic groups (Filipinos, order may be related to the changes in uric acid
Polynesians, Micronesians, and Aborigines), but metabolism observed in diabetes insipidus [381–
dietary habits, plasma factors, and/or renal dys- 385]. Finally, hyperuricemia has been observed
function may contribute to the increased fre- in one case of acute uric acid nephropathy during
quency of hyperuricemia and gout in these pregnancy [300]. The diagnosis in this case was
groups. made on the basis of the marked hyperuricemia
and an elevated uric acid to creatinine ratio as has
been discussed previously in the section “Renal
Hyperuricemia and Gout in Pregnancy disease and gout.” Conventional treatment of this
disorder leads to the reversal of the renal failure
Gout is an extremely unusual and rare occurrence and resulted in a healthy newborn.
in pregnant females, but gout has been docu- In summary, gout is an extremely rare compli-
mented during and after pregnancy in the older cation of pregnancy. If gouty arthritis does mani-
literature [887–894]. It is difficult to discuss the fest itself during pregnancy, an underlying disease
pathogenesis of gout in these reports since these is likely to have triggered this abnormality in uric
cases were described before genetic causes of acid metabolism leading to gout. Hyperuricemia
gout were identified. Nonetheless, it appears from is a more common finding, and preeclampsia is
the reported cases and some textbooks that gouty probably the most frequent cause. Nonetheless,
arthritis in pregnant females is probably in most diuretic use, hepatic dysfunction, and even endo-
cases secondary to another disease associated crine disturbances may result in elevated serum
with elevated serum uric acid levels or multifac- uric acid levels in pregnant females.
torial in its etiology [895].
Hyperuricemia is much more common during
pregnancy than gout since it is a marker for preec- Postmenopausal Hyperuricemia
lampsia, a condition occurring not infrequently and Gout
during pregnancy. Despite its occurrence in
preeclampsia, the mechanism(s) for the altered Most of the female patients with gouty arthritis
uric acid metabolism in this condition remains have been reported during the postmenopausal
Associated Disorders 147

years [645, 904–906]. Perhaps contributing to the reveals no simple relationships exist between
increased incidence of gout in this age group is obesity, hyperlipidemia, hypertension, diabetes
the fact that serum uric acid levels approach or mellitus, and atherosclerosis despite their fre-
are equal to those in males [275, 455, 907–910]. quent coexistence with hyperuricemia and gout.
A large percentage of postmenopausal females
with gout are being treated with diuretics [645,
880]. Although the clinical characteristics of gout Obesity
in the female vary from report to report, the fol-
lowing differences between males and females Obesity has been recognized as a condition asso-
have been noted in some publications. Most ciated with hyperuricemia both from population
reports identify the female as having the onset of studies and from investigations of the metabolic
their disease about a decade later than males with consequences of obesity [915–920]. In addition
gout [904, 906]. Some investigators have found to the correlation between obesity and hyperuri-
that renal insufficiency is more common in cemia, surveys of gouty patients have also found
females with gout than males [645, 880]. A more a higher percentage of obesity than in control
universal finding is the increased incidence of subjects [875, 876, 921]. Further, excess weight
hematologic malignancies in females compared gain has been shown to increase the risk of gout
to males [880]. This cause of gout is important to among patients with hyperuricemia [652]. As a
identify early so that treatment of the underlying corollary to these studies, weight loss has been
marrow disease can be initiated. Such patients documented to reduce gouty episodes and lower
may present with renal calculi and tophi, and serum uric acid levels [922].
these complications may precede the onset of There remains controversy over the role of
articular disease [880]. Although the joint hyperuricemia as a risk factor for cardiovascular
involvement is similar in males and females with disease despite the fact that some studies suggest
gout, a few investigations have suggested that the a positive correlation between hyperuricemia and
involvement of finger and ankle joints is more cardiovascular disease [923–926]. This point is
common in females than in males [906, 911]. In discussed further in the section “Atherosclerosis.”
both the premenopausal and postmenopausal However, there appears to be a correlation
female with gout, it is reasonable to assume that between obesity, hyperuricemia, and other car-
any underlying condition leading to hyperurice- diovascular risk factors such as hypertension and
mia such as psoriasis, infectious mononucleosis, hyperlipidemia. For this reason, it is reasonable
and sickle cell disease may be sufficient to cause to initiate therapies aimed at the control of excess
gouty arthritis [912–914]. weight, hypertension, and elevated blood lipids
In summary, gout is most common in post- in hyperuricemic subjects.
menopausal females as compared with the pre- Modern studies have investigated obesity and
menopausal and juvenile female. Even though hyperuricemia using modern tools such as CT
underlying diseases, especially renal insufficiency, scanning to evaluate fat distribution [927–929].
may be an accompanying disorder, diuretic use The results of these investigations define differ-
and malignant disease represent the most impor- ent mechanisms for the hyperuricemia depending
tant conditions that need to be excluded in the on the body distribution of fat [929]. Clearly,
postmenopausal female with gout. these mechanistic studies need further evaluation
to determine how the distribution of fat affects
uric acid metabolism. There have also been cor-
Associated Disorders relations made between the adipocyte hormone,
leptin, and serum uric acid levels [930]. Future
There are a number of disorders that occur fre- studies may uncover relationships between uric
quently in association with hyperuricemia and acid metabolism, leptin concentration, hyperin-
gout. Assessment of these associated disorders sulinemia, and fat distribution [931–934]. Since
148 5 Clinical Aspects of Gout and Associated Disease States

more than 5 % of the national health expenditure presence of previous renal disease. Removal of
in the United States is related to the costs of obe- NSAIDS from their treatment protocol and the
sity and its complications, ongoing studies aimed control of their serum uric acid levels resulted in
at understanding and defining risk factors of obe- a significant improvement in their creatinine
sity and its related disorders are of great clearance of greater than 10 ml/min. Thus, these
significance [935]. results suggest that the control of vascular risk
factors may improve renal function.
A few studies have documented the occur-
Hyperlipidemia rence of hypercholesterolemia in those with
hyperuricemia and gout, but no linkage has been
Documentation of a hyperlipidemic state is established between hypercholesterolemia and
important for several reasons. First, the older lit- hyperuricemia or gout [938, 939, 948–951].
erature indicates that a strikingly high percentage Even though the frequency of hypertriglyceri-
of patients with gout (72–84 %) have hypertrig- demia in patients with hyperuricemia and gout is
lyceridemia [936–941]. Further, many patients probably related to lifestyle factors (obesity, diet,
with hypertriglyceridemia have hyperuricemia as diabetes mellitus, and alcohol intake) and drugs
well [937]. More recent studies have described (diuretics, estrogen therapy, or beta-adrenergic
the lipoprotein phenotypes in patients with pri- blockers), the importance of hyperlipidemia to
mary gout [942]. Less than 15 % of these gouty the overall health of the patient makes its recog-
subjects manifested types IIa and IIb hyperlipo- nition and treatment of significance to those car-
proteinemia (hypercholesterolemia, low density ing for gouty patients [952, 953]. For this reason,
lipoproteins, and combined hyperlipidemia – as well as the high frequency of hypertriglyceri-
LDL plus very low density lipoproteins), whereas demia in those with hyperuricemia or gout, a
most (almost 70 %) manifest type IV hyperlipi- brief review of both inherited and acquired causes
demia (endogenous hyperlipemia – VLDL). of hypertriglyceridemia is discussed. An addi-
Other studies have shown decreases in high den- tional rationale for such a discussion is the fact
sity lipoprotein cholesterol and alterations in very that hypertriglyceridemia is a common ailment
low density apolipoprotein C-III [943–946]. All affecting 10 % of males in the age group between
these studies document the occurrence of hyper- 35 and 39 [954].
lipemia in association with hyperuricemia and Even though no mechanisms have been clearly
gout, but they often do not evaluate either the defined for the association between purine and
newer clinical subsets and mutants causing hyper- triglyceride metabolism, hypertriglyceridemia is
lipemia or the clinical subsets of gout. more frequent than gout. The latter has a preva-
Second, the identification of lipid abnormali- lence of 0.2–0.3 per 100 individuals as contrasted
ties and their treatment are a preventive measure with 1 per 100 for hypertriglyceridemia. The her-
that often decrease morbidity from atherosclero- itable lipid abnormalities associated with elevated
sis and prolong life. The results of preventive triglyceride levels include familial combined
treatment and the documented association hyperlipidemia and its variant, familial lipopro-
between gout and hyperlipidemias provide a rea- tein lipase deficiency, familial apolipoprotein
sonable rationale for screening for both heritable C-II deficiency, familial inhibitor of lipoprotein
and acquired disorders of lipid metabolism. lipase, monogenic hypertriglyceridemia (type V
Third, a recent paper has documented a deficit hyperlipoproteinemia), lecithin/cholesterol acyl-
in renal function in patients with chronic gout transferase deficiency, fish-eye disease, type III
who were using nonsteroidal anti-inflammatory hyperlipoproteinemia (dysbetalipoproteinemia),
drugs (NSAIDS) for the treatment of their gout and the DNA polymorphisms of apo A-I, apo
[947]. These studies showed that low creatinine C-III, and apo A-IV. The acquired disorders are
clearance in their gouty subjects was related to discussed with type III hyperlipoproteinemia
age, hypertension, hypertriglyceridemia, and the with which they are associated.
Associated Disorders 149

Familial Combined Hyperlipidemia present and superimposed on other diseases such


Familial combined hyperlipidemia occurs in as diabetes mellitus, estrogen therapy, lipid-rais-
about 1–2 % of the population in North America ing drugs, or alcohol use [972].
and Europe. It may occur as a single entity or in
combination with hypertension, obesity, or insu- Familial Apolipoprotein C-II Deficiency
lin resistance [955–961]. This disorder is the Apolipoprotein C-II is a cofactor for the activa-
most common genetic hyperlipidemia identified tion of lipoprotein lipase, and like familial lipo-
in survivors of myocardial infarctions, but to protein lipase deficiency, a defect in this cofactor
date, its complete genetic composition remains for lipase activation alters the hydrolysis of trig-
unknown [957]. It is also said to cause 10–20 % lycerides and the transfer of fatty acids to tissues.
of the cases of premature coronary heart disease The clinical hallmark of apolipoprotein C-II
[962]. The pattern of inheritance is compatible deficiency is pancreatitis associated with marked
with an autosomal dominant trait [957]. Its clini- chylomicronemia and triglyceridemia [973].
cal presentation is most effectively defined by Since this disease simulates familial lipoprotein
evaluating patients with premature coronary lipase deficiency (hyperlipoproteinemia type I),
artery disease. Xanthomas are rare, and the lipo- it has also been referred to as hyperlipoproteine-
protein patterns may change in an affected indi- mia type IB.
vidual over time. Lipoprotein patterns may also
vary within family members. No single genetic Familial Lipoprotein Lipase Inhibitor
marker has been characterized for familial com- Little is known about familial lipoprotein lipase
bined hyperlipidemia, but some inroads have inhibitor since only a single family has been
been made into its genetic identity [963–969]. investigated [974]. Clinically, affected patients
In familial combined hyperlipidemia, the have markedly elevated triglyceride levels
plasma triglyceride levels are elevated ranging (>2,000 mg/dl), eruptive xanthoma, and abdomi-
between 200 and 400 mg/dl or even higher. The nal pain with pancreatitis. The inhibitor from
LDL cholesterol levels are usually higher than affected patients blocks adipose tissue lipopro-
100 mg/dl, and total serum apo B levels are often tein lipase activity and postheparin lipase.
higher than 85 mg/dl. The latter apolipoprotein is
often used as a marker for this disorder. Monogenic Hypertriglyceridemia
Monogenic hypertriglyceridemia (type V hyper-
Familial Lipoprotein Lipase Deficiency lipoproteinemia) may present with moderate or
Familial lipoprotein lipase deficiency, familial severe elevations in triglyceride levels. For pur-
apolipoprotein C-II deficiency, and familial lipo- poses of classification, patients with chylomi-
protein lipase inhibitor are rare genetic causes of cronemia are separated into those with elevated
hypertriglyceridemia. The first two disorders are VLDL levels (type V) and those with no eleva-
inherited as autosomal recessive traits, whereas tion in their VLDL levels along with the absence
familial inhibitor of lipoprotein lipase appears to of postheparin plasma lipoprotein lipase activity
be inherited as an autosomal dominant trait. The [975]. Type V hyperlipoproteinemic patients usu-
full-blown clinical picture of familial lipoprotein ally present with hypertriglyceridemia and have
lipase deficiency includes abdominal pain due to chylomicronemia after an overnight fast, whereas
pancreatitis, hepatosplenomegaly, eruptive xan- type IV hyperlipoproteinemic patients have
thomas, and lactescent plasma. As noted, this dis- hypertriglyceridemia but do not have chylomi-
order is very rare, but in some populations, it may crons in their plasma in the fasting state.
occur at a higher frequency [970, 971]. Except for a very few cases, familial hyper-
Heterozygotes with familial lipoprotein lipase triglyceridemia is usually a manifestation of a
deficiency may or may not have elevated plasma primary disorder in association with a secondary
triglyceride levels, but hypertriglyceridemia may cause of hyperlipidemia [953, 976–981].
occur in patients when secondary factors are Secondary causes for hypertriglyceridemia
150 5 Clinical Aspects of Gout and Associated Disease States

include obesity, alcohol intake, and diabetes mel- receptors or a decrease in receptor-mediated lipo-
litus [976]. Drugs including diuretics, beta-adren- protein uptake as in hypothyroidism [999]. The
ergic blockers, estrogens, and isotretinoin decrease in LDL receptor number observed in
(accutane) treatment may elevate triglycerides. In heterozygotes with familial hypercholesterolemia
the case of accutane, about 25 % of patients along with a mutant apo-E allele has also been
treated with this agent develop hyperlipidemia. shown to trigger the expression of type III hyper-
As inferred previously, hypertriglyceridemia may lipoproteinemia [1000]. Dietary alterations that
exist without any accompanying acquired disor- increase cholesterol and plasma lipoproteins and
der, but the nature of the primary cause remains downregulate LDL receptors may also increase
unknown [978]. In some studies examining type type III hyperlipoproteinemia expression
V hyperlipoproteinemic patients and their first- [992–995].
degree relatives, no increase in atherosclerosis The secondary genetic parameters that have
has been observed [976, 982]. been shown to contribute to the expression of
type III hyperlipoproteinemia include those dis-
Type III Hyperlipoproteinemia orders with an overproduction of lipoproteins or
Type III hyperlipoproteinemia or dysbetalipopro- elevated levels of cholesteryl ester transfer pro-
teinemia is an inherited disorder in which patients tein [1001–1003]. Thus, autosomal recessive
manifest elevated concentrations of both plasma forms of type III hyperlipoproteinemia are repre-
cholesterol and triglycerides along with choles- sented by a mutant apo-E together with a second-
terol-rich remnants (ß-VLDL) derived from chy- ary acquired or genetic factor.
lomicrons secreted by the intestine and VLDL Type III hyperlipoproteinemia may also occur
secreted by the liver [983, 984]. Subsequent stud- as an autosomal dominant trait. The mutants
ies documented the role of apo-E, a protein that identified as mediators of the dominant form of
normally mediates the interaction of apo-E-con- type III hyperlipoproteinemia all affect the bind-
taining lipoprotein and binding to lipoprotein ing of apo-E to LDL receptors [989, 1004, 1005].
receptors. Mutant apo-E in type III hyperlipopro- These mutants include alterations in amino acid
teinemia alters binding to lipoprotein receptors sequences in the receptor-binding domain of
[985–987]. apo-E (residues 136–150). Mutations identified
The most common mutant form of apo-E is at this time are 142Arg → Cys, 145Arg → Cys,
apo-E-2, and this mutant results in type III hyper- 146Lys → Gln, 146Lys → Glu substitutions, or a
lipoproteinemia with a single amino acid substi- seven amino acid insertion at residues 121–127.
tution of cysteine for arginine at residue 158 in In addition to defective receptor binding, the
apo-E. Other rare variants have also been 142Arg → Cys and the seven amino acid inser-
described [985, 988, 989]. Overt hyperlipopro- tion mutants show a preference for triglyceride-
teinemia occurs rarely in patients with mutant rich lipoproteins [1006, 1007]. Further, residues
apo-E-2 or its variants, and most patients with 136–150 have an increased affinity for heparin
homozygous apo-E-2 are normolipodemic or binding which is important for lipolytic process-
even hypolipidemic. This low frequency of overt ing and lipoprotein remnant uptake [1008, 1009].
hyperlipidemia indicates that additional genetic Thus, mutants altered in their capacity to bind to
or environmental factors are necessary for the LDL receptors or heparin binding sites with a
full expression of type III hyperlipoproteinemia. preference for triglycerides regulate the dominant
The secondary environmental factors most often inheritance of type III hyperlipoproteinemia.
implicated are hypothyroidism, glucose intoler- These abnormalities lead to defective uptake and
ance, obesity, aging, estrogen deficiency, processing of triglycerides.
increased intake of dietary cholesterol, alcohol The clinical findings in patients with type III
intake, and some drugs [984, 990–998]. For the hyperlipoproteinemia usually include an adult
most part, these secondary factors in type III onset of the disease with xanthomas, premature
hyperlipoproteinemia cause a reduction in LDL coronary artery, and peripheral vascular disease
Associated Disorders 151

as well as asymptomatic hyperuricemia. The geo- Low concentrations of plasma cholesteryl esters
graphic locations of the xanthomas are of special and lysophosphatidylcholine are characteristic of
significance since palmar xanthoma occurring in the disorder [1023]. It is important to recognize
the palmar creases is pathognomonic of type III that not all patients with LCAT deficiency have
hyperlipoproteinemia [1010, 1011]. About one- hypertriglyceridemia.
half the patients with untreated type III hyperli-
poproteinemia manifest this diagnostic sign, Fish-Eye Disease
whereas other xanthomas (tuberous and tubero- Fish-eye disease also represents a deficiency
eruptive) are also encountered on the elbows, (partial) in LCAT activity, but these patients only
knees, buttocks, and knuckles. The latter are non- manifest corneal opacities with no other associ-
diagnostic of type III hyperlipoproteinemia since ated findings. Most patients with this disorder
they are also observed in other hyperlipidemias. have elevated plasma triglycerides, low HDL
Peripheral vascular disease is almost as common cholesterol levels, and normal plasma cholesteryl
as coronary artery disease in type III patients, and ester levels [1020, 1024, 1025].
vascular disease is observed in about 50 % of
such patients [984, 1012–1015]. Asymptomatic DNA Polymorphisms
hyperuricemia has been reported in about 50 % Finally, a number of investigations have been
of patients with this disorder, but only a small directed toward an evaluation of genetic markers
percentage of such patients (4 %) develop gout to determine whether such determinants are
[984]. As can be determined from this discussion, capable of identifying those patients at risk for
the diagnosis of type III hyperlipoproteinemia is atherosclerosis [1026–1045]. Although no simple
not a straightforward process since the disease means for the detection of DNA polymorphisms
expression is variable. The following diagnostic that confer susceptibility to hyperlipidemia and
parameters are the most useful: serum triglycer- coronary artery disease exist at the present time,
ide and cholesterol levels in the range of 400 mg/ such investigations are eventually likely to define
dl, the presence of ß-VLDL, xanthomas (espe- a panel of DNA polymorphisms that will be use-
cially palmar crease xanthomas), premature vas- ful for the characterization of patients at increased
cular disease, and the occurrence of the apo E-2/2 risk for coronary artery disease.
phenotype. As noted at the outset of this section
“Hyperlipidemias”, the pathogenesis of the inter-
Lecithin/Cholesterol Acyltransferase actions between hypertriglyceridemia and hyper-
(LCAT) Deficiency uricemia remains unresolved. Some investigators
Both lecithin/cholesterol acyltransferase have shown that no change occurs in the serum
deficiency and fish-eye disease manifest hyper- uric acid level, urinary uric acid level, or oxypu-
triglyceridemia and are inherited as autosomal rine excretion when serum triglyceride levels are
recessive traits [1016–1022]. In familial LCAT dramatically increased [1046]. Other investiga-
deficiency, there is an absence of lecithin/choles- tors have documented an inverse correlation
terol acyltransferase activity in the plasma. The between serum triglyceride levels and fractional
clinical manifestations of the disorder include cor- urate excretion as well as a positive correlation
neal opacities, normochromic anemia, bone mar- between serum triglyceride and serum uric acid
row foam cells, proteinuria with red blood cell, levels [1047–1050]. In summary, there is an
and hyaline casts, and in some patients, athero- established interrelationship between uric acid
sclerosis with tendon and planar xanthomas are metabolism and hyperlipidemia, usually hyper-
observed. Serum uric acid levels are elevated in triglyceridemia, but the cause for these interac-
some patients, and one reported patient had gout. tions is unknown. It seems reasonable to assess
An abnormal pattern of lipoproteins is present fasting lipids (12 h fast) in patients with hyperuri-
including elevated plasma triglycerides, cemia and patients with gout. This concept is rein-
unesterified cholesterol, and phosphatidylcholine. forced by the role that treatment and disease-related
152 5 Clinical Aspects of Gout and Associated Disease States

prevention play in those with an identifiable Thus, a complete understanding of the mecha-
abnormality if their plasma lipids are lowered. nism causing hyperuricemia in syndrome X
Analyses of the complex lipid abnormalities are remains unresolved, but it appears to be a multi-
not a simple task; however, the clinical factorial problem.
identification of xanthomas (especially palmar Two unusual findings reported in association
crease xanthomas) and lenticular opacities can with syndrome X deserve brief mention. First, a
easily characterize some of the inherited hyper- review of the patients with gonadal dysgenesis
lipidemias. In addition, appropriate attention to (Turner syndrome) has documented an increased
secondary causes of hyperlipidemias is usually frequency of syndrome X including hyperurice-
easy to recognize clinically and can be treated in mia in these patients [1063]. As would be
a straightforward manner. expected, these patients have an increased inci-
dence of diabetes mellitus (NIDDM and IDDM)
as well as hypertension, stroke, and ischemic
Glucose Intolerance heart disease. The other unusual clinical finding
in syndrome X is the case of a 64-year-old female
There are two separate components that relate to with severe tophaceous gout [1064]. This patient
glucose intolerance and alterations in uric acid also suffered from NIDDM, obesity, hyperten-
metabolism. The first is the multifaceted meta- sion, and dyslipidemia, all components of syn-
bolic syndrome X or insulin resistance syndrome drome X. This rare association between gout and
that is associated with insulin resistance, hyperin- syndrome X emphasizes the need to recognize
sulinemia, dyslipidemia, essential hypertension, the clinical diversity associated with hyperurice-
visceral obesity, glucose intolerance or noninsu- mia and gout.
lin-dependant diabetes mellitus, abnormalities in The second component is the relationship
blood coagulation (elevated levels of plasmino- between hyperuricemia, gout, and diabetes mel-
gen activator inhibitor type 1 and fibrinogen), litus. Despite continued interest in these relation-
microalbuminuria, and hyperuricemia [1051– ships, there is little evidence in support of any
1058]. Some investigators have added polycystic pathophysiological process relating modest
ovaries as a component of this syndrome as well. degrees of hyperglycemia to uric acid levels in
The lipid abnormalities in syndrome X overlap diabetes mellitus [916, 1065, 1066]. Of course,
with the dyslipidemia observed in some patients diabetic ketoacidosis is associated with hyperuri-
with hyperuricemia and gout. In syndrome X, the cemia, and the pathogenesis of this metabolic
lipid abnormalities include elevated serum trig- derangement relates to the capacity of ketoacids
lycerides, small LDL particles, low high density to inhibit renal tubular urate secretion [263].
lipoprotein (HDL) cholesterol, and hypercholes-
terolemia. These lipid parameters along with
hypertension and the prothrombotic state increase Hypertension
the risks for coronary artery disease. Since there
is no question about the association between Clearly, hypertension and gout are linked since
insulin resistance and hyperuricemia, the patho- the older literature documents the fact that
genesis of this abnormality in uric acid metabo- 25–50 % of gouty subjects have hypertension
lism and its relation to insulin needs to be [1067, 1068]. Hyperuricemia has also been
investigated [1055, 1059]. As might be expected, reported in about one-third of the patients with
the pathogenesis of hyperuricemia in syndrome untreated hypertension [1069–1071]. More recent
X is multifaceted. It has been related to diuretic studies confirm the association between hyper-
use, obesity, enhanced renal sodium retention, tension and hyperuricemia; the frequency of this
and hypertriglyceridemia [1057, 1060–1062]. association in these studies was about 60 %
Weight reduction definitely lowers serum uric [1072]. Since hypertension, obesity, insulin resis-
acid and serum triglyceride levels [1057, 1062]. tance, and hypertriglyceridemia often coexist,
Associated Disorders 153

diagnostic studies should identify these addi- compound is a potent antioxidant capable of free
tional risk factors as well [1073–1077]. Some radical scavenging [1091–1102]. The literature
other studies have emphasized the role of hyperu- now has determined that uric acid is capable of
ricemia as a marker of renal disease in hyperten- scavenging singlet oxygen, hypochlorous acid,
sive patients [1078–1084]. Hyperuricemia and other radicals, and its plasma concentration
observed in association with hypertension is often (300 uM/dl or greater–5.0 mg/dl or greater) is
the result of decreased renal urate excretion, and significantly greater than ascorbate, another major
some investigators have proposed that intrarenal plasma antioxidant [1091, 1097]. Uric acid has
ischemia caused by vascular contraction and been shown to be produced by the microvascular
increased lactate production may contribute to a endothelium and blocks the oxidative inactiva-
decrease in urate secretion by the anion-exchange tion of cyclooxygenase and angiotensin-convert-
tubular transport mechanism [1078]. As a corol- ing enzyme [1099]. This latter activity protects
lary to the pathogenesis of hypertension-related coronary-sized vessels from vasoconstriction
hyperuricemia, reduction in serum urate levels in and permits vasodilation in the face of oxida-
the hypertensive gouty subject may prevent addi- tive stress. These properties of uric acid may be
tional impairment of kidney function. related to the role of uric acid in cardiovascular
disease [1099]. Such concepts are further sup-
ported by animal studies showing vasorelaxation
Atherosclerosis of the rat thoracic aorta after uric acid scavenges
peroxynitrite [1103]. Further, oxidized LDL
Cerebrovascular and cardiovascular disease are (low density lipoprotein) has been shown to be
major causes of death in gouty patients treated associated with coronary artery disease [1104].
with antihyperuricemic drugs, but it is clear that Even though the evidence linking atherosclero-
hyperuricemia by itself is not an independent risk sis, uric acid, and oxidized LDL is incomplete,
factor for atherosclerosis, cerebrovascular dis- these possible interrelationships add confusion
ease, or coronary artery disease [1085–1089]. to the role of hyperuricemia in atherosclerosis.
Even though hyperuricemia as an independent It is clear that resident vascular cells, oxidative
risk factor of atherosclerosis remains controver- stress, and inflammation stimulate the formation
sial, obesity, hyperlipidemia, and hypertension, of oxidized LDL. Resident vascular cells pro-
frequent components of the clinical presentation duced so-called minimally modified LDL [1105].
of gout, are known risk factors for atherosclerosis This altered LDL stimulates vascular cells to pro-
and its accompanying disorders [1090]. duce monocyte chemotactic protein 1 as well as
Thus, of the associated disorders discussed granulocyte and macrophage colony stimulating
herein, it is reasonable to screen gouty patients factors [1106]. These reactions stimulate further
for lipid disorders and to treat obesity and hyper- peroxidation of LDL, and such oxidative stress
tension aggressively to prevent vascular disease generates completely oxidized LDL. Receptors
including any deterioration in renal function. on the macrophage internalize the oxidized LDL
Evaluating hyperuricemia in patients with gout is to create foam cells, primary components of ath-
discussed in the section “Asymptomatic erosclerotic lesions [1107, 1108]. Since oxidized
hyperuricemia.” LDL cellular uptake is not regulated by feed-
back inhibition, massive quantities of cholesterol
are ingested by these macrophages to become
Critical Illnesses foam cells. Oxidized LDL is chemotactic for
monocytes and enhances monocyte binding to
There is now a large body of evidence to con- endothelium [1109, 1110]. Once these lipid-laden
tradict the thesis that uric acid is a metaboli- macrophages/monocytes enter the subendothelial
cally inert end product of purine metabolism space, their egress is inhibited by oxidized LDL
and to support the premise that this ubiquitous [1111]. The cytotoxicity of oxidized LDL also
154 5 Clinical Aspects of Gout and Associated Disease States

damages vascular cells leading to the release of of renal function and enhance hypoxanthine sal-
lysosomal enzymes. These alterations cause the vage as a result of xanthine oxidase inhibition by
progression of atherosclerotic lesions [1112, allopurinol [1121]. Clearly, the role of hyperuri-
1113]. As further proof of the role of oxidized cemia as a marker of serious illness and as a
LDL in atherosclerosis, antibodies against oxi- prognostic factor remains incompletely under-
dized LDL react with atherosclerotic lesions stood and somewhat controversial. Its role as an
in blood vessels but not with normal arteries in antioxidant and its capacity to reduce atheroscle-
the absence of atherosclerotic lesions [1114]. In rosis are also incompletely resolved issues.
addition, autoantibodies to oxidized LDL are at In summary, although there are no direct
higher concentrations in patients with carotid mechanistic associations between uric acid
atherosclerosis than in age-matched normal con- metabolism and obesity, glucose intolerance,
trol subjects [1115]. Similarly, immunoreactive hyperlipidemia, atherosclerosis, hypertension,
oxidized LDL plasma concentrations are elevated and critical illnesses, sufficient evidence exists
in patients with acute myocardial infarctions as to evaluate some of these associations in patients
compared to normal control subjects [1116]. with hyperuricemia and/or gout. At the present
Despite all this evidence linking the reduced time, it appears reasonable for physicians caring
oxidation of LDL and atherosclerosis, the role of for patients with gout to treat obesity with weight
uric acid as an antioxidant and as a factor causing reduction, to evaluate and manage hypertension
reduced atherosclerosis remains an incomplete aggressively to protect renal function deteriora-
story. A number of investigations support the tion, and to evaluate and treat these same patients
antioxidant activity of uric acid against lipopro- for any identifiable dyslipidemias. It must be
tein oxidation, but whether this is the sole or clear from this discussion that gout is the clini-
major factor acting to reduce the severity of ath- cal expression of many different underlying
erosclerosis remains an open question [1117– disorders.
1120]. As the mechanism(s) of atherosclerosis
become better understood, a reevaluation of the
role that uric acid plays in this pathological pro- References
cess is necessary.
Finally, in contrast to these hypothesized pro- 1. Hausch R, Wilkerson M, Singh E, Reyes C, Harrington
T. Tophaceous gout of the thoracic spine presenting as
tective roles of uric acid in atherosclerosis,
back pain and fever. J Clin Rheumatol. 1999;6:335–41.
marked hyperuricemia is related to tissue hypoxia 2. Marsaudon E, Bouchard C, Languard D. Compression
in severely ill patients and often indicates a poor radiculaire par un tophus goutteux vertebral: a propos
prognosis [101, 1121]. Subsequent studies have d’un cas et revue de la literature. Rev Med Interne.
1999;20:253–7.
questioned the use of uric acid levels as well as
3. Kao MC, Huang SC, Chiu CT, Yao YT. Thoracic cord
adenosine, inosine, hypoxanthine, and lactic acid compression due to gout: a case report and literature
levels as specific markers of prognosis even review. J Formos Med Assoc. 2000;98:572–5.
though such metabolic changes were increased as 4. Mekelburg K, Rahimi AR. Gouty arthritis of the spine:
clinical presentation and effective treatments.
clinical deterioration progressed [1122].
Geriatrics. 2000;55:71–4.
Nonetheless, the hypothesis that tissue hypoxia 5. Paquette S, Lach B, Guiot B. Lumbar radiculopathy
results in increased production of uric acid via secondary to gouty tophi in the filum terminale in a
the depletion and catabolism of adenosine patient with systemic gout: case report. Neurosurgery.
2000;46:986–8.
triphosphate remains valid. In addition, patients
6. Gines R, Bates DJ. Tophaceous lumbar gout mimick-
with hypoxia also have a decreased uric acid ing an epidural abscess. Am J Emerg Med. 1998;
excretion. Some investigations have also sug- 16:216.
gested that the hyperuricemia observed in severe 7. Vargo J, Giampaolo C, Goldenberg DC. Tophaceous
gout of the spine in a patient with no peripheral tophi:
illnesses may only reflect the degree of renal dys-
case report. Arthritis Rheum. 1985;28:1312–5.
function [1123]. Some have even suggested that 8. Pascual E. The diagnosis of gout and CPPD crystal
allopurinol treatment may prevent deterioration arthropathy. Br J Rheumatol. 1996;35:306–8.
References 155

9. Wolfe F, Cathey MA. The misdiagnosis of gout and 28. Weinberger A, Schumacher HR, Schimmer BM, et al.
hyperuricemia. J Rheumatol. 1991;18:1232–4. Arthritis in acute leukemia. Clinical and histopatho-
10. Pascual E, Batele-Gualda E, Martinez A, Rosas J, logical observations. Arch Intern Med. 1981;141:1183.
Vela P. Synovial fluid analysis for diagnosis of inter- 29. Dosman JA, Crawhall JC, Klassen GA. Uric acid
critical gout. Ann Intern Med. 1999;131:756–9. kinetic studies in the immediate post-myocardial
11. Weinberger A, Schumacher HR, Agudelo CA. Urate infarction period. Metabolism. 1975;24:473.
crystals in asymptomatic metatarsophalangeal joints. 30. Levine SA, Gordon B, Derick CL. Some changes in
Ann Intern Med. 1991;91:56–7. the chemical constituents of the blood following a
12. Ronault T, Caldwell DS, Holmes EW. Aspiration of marathon race. JAMA. 1924;82:1778.
the asymptomatic metatarsophalangeal joints in gout 31. Schrier RW, Haus J, Keller HI, et al. Renal, metabolic
patients and hyperuricemic controls. Arthritis Rheum. and circulatory responses to heat and exercise. Ann
1982;25:209–12. Intern Med. 1970;73:213.
13. Pascual E. Persistence of monosodium urate crystals 32. Knochel JP, Dotin LN, Hamburger RJ. Heat stress,
and low-grade inflammation in the synovial fluid of exercise, and muscle injury: effects on urate metabo-
untreated gout. Arthritis Rheum. 1991;34:141–5. lism and renal function. Ann Intern Med. 1974;81:321.
14. Chopra KF, Schneiderman P, Grossman ME. Finger 33. Quick AJ. The effect of exercise on excretion of uric
pad tophi. Cutis. 1999;64:233–6. acid. J Biol Chem. 1935;110:107.
15. Marcen R, Gallego N, Orofino L, et al. Impairment of 34. Castenfors J. Renal function during exercise. Acta
tubular secretion of urate in renal transplant patients Physiol Scand Suppl. 1967;70:7.
on cyclosporine. Nephron. 1995;70:307–13. 35. Knochel JP. Biochemical, electrolyte, and acid–base
16. Lin HY, Rocher LL, McQuilan MA, et al. disturbances in acute renal failure. In: Brenner BM,
Cyclosporine-induced hyperuricemia and gout. N Lazarus JM, editors. Acute renal failure. New York:
Engl J Med. 1989;321:287–92. Churchill Livingstone; 1988. p. 682–3.
17. Baethge BA, Work J, Landreneau MD, McDonald JC. 36. Lacocq FR, McPhaul JJ. Effects of starvation, high fat
Tophaceous gout in patients with renal transplants diets and ketone infusions on uric acid balance.
treated with cyclosporine A. J Rheumatol. 1993; Metabolism. 1965;14:186.
20:2171. 37. Ravioko KO. Neonatal hyperuricemia. J Pediatr.
18. Chaoui A, Garcia J, Kurt AM. Gouty tophus simulat- 1976;88:625.
ing soft tissue tumor in a heart transplant recipient. 38. Valat JP, Lamisse F, LeChevallier PL, et al. Les varia-
Skeletal Radiol. 1997;26:626–8. tions de l’uricemie au cours de’ compensations respi-
19. Kennedy DT, Hayney MS, Lake KD. Azathioprine ratories aigues des bronchopathies chroniques. Rev
and allopurinol: the price of an avoidable drug inter- Rhum Mal Osteoartic. 1974;41:179.
action. Ann Pharmacother. 1996;30:951–3. 39. Bergeaux G, Klein RC. Hyperuricemia following
20. Jagose JT, Bailey RR. Muscle weakness due to colchi- smoke inhalation. Am Rev Respir Dis. 1974;109:145.
cine in a renal transplant recipient. N Z Med J. 40. Oliva PB. Lactic acidosis. Am J Med. 1970;48:209.
1997;110:343. 41. Saker BM, Tofler OB, Burvill MJ, Reilly KA. Alcohol
21. Tapal MF. Colchicine myopathy. Scand J Rheumatol. consumption and gout. Med J Aust. 1967;1:1213.
1996;25:105. 42. Pell S, D’Alonzo CA. The prevalence of chronic dis-
22. Gruberg L, Har-Zahav Y, Agranat O, Freimark D. ease among problem drinkers. Arch Environ Health.
Acute myopathy induced by colchicine treated heart 1968;16:679.
transplant recipient: possible role of the multidrug 43. Beck LH. Clinical disorders of uric acid metabolism.
resistance transporter. Transplant Proc. 1999;31: Med Clin North Am. 1981;65:401.
2157–8. 44. Puig JG, Fox IH. Ethanol-induced activation of ade-
23. Zurcher RM, Bock HA, Thiel G. Excellent uricosuric nine nucleotide turnover. Evidence for a role of ace-
efficacy of benzbromarone in cyclosporine-A treated tate. J Clin Invest. 1984;74:936.
renal transplant patients: a prospective study. Nephrol 45. Shergy WJ, Gilkeson GS, German DC. Acute gouty
Dial Transplant. 1994;9:549–51. arthritis and intravenous nitroglycerin. Arch Intern
24. Loeb JN. The influence of temperature of the solubility Med. 1988;148:2505.
of monosodium urate. Arthritis Rheum. 1972;15:189. 46. Daly RJ, Camann WR, Dawley D, O’Rourke ME.
25. Kelley WN. Approach to the patient with hyperurice- “Cocktail”-coronary care. N Engl J Med. 1984;310:1123.
mia. In: Kelley WN, Harris Jr ED, Ruddy S, Sledge 47.
CB, editors. Textbook of rheumatology. Philadelphia: 48. Dylewski JS, Gerson M. Hyperuricemia in patients
WB Saunders; 1981. p. 494. with infectious mononucleosis. Can Med Assoc J.
26. Adler R, Robinson R, Pazdral P, Grushkin C. 1985;132:1169.
Hyperuricemia in diarrheal dehydration. Am J Dis 49. Byrd L, Sherman RL. Radiocontrast-induced acute
Child. 1969;136:564. renal failure: a clinical and pathophysiologic review.
27. Faller J, Fox IH. Ethanol-induced hyperuricemia. Medicine. 1979;58:270.
Evidence for increased urate production by activation 50. Alexander RD, Barkes SL, Abuelo G. Contrast media-
of adenine nucleotide turnover. N Engl J Med. induced oliguric renal failure. Arch Intern Med.
1980;307:1598. 1978;138:381.
156 5 Clinical Aspects of Gout and Associated Disease States

51. Krumlovsky FA, Simon N, Santhanam S, et al. Acute 69. Roach ES, Delgado M, Anderson L, et al. Carbama-
renal failure associated with administration of radio- zepine trial for Lesch-Nyhan self-mutilation. J Child
graphic contrast material. JAMA. 1978;239:125. Neurol. 1996;11:476–8.
52. Fang LS, Sirota RA, Ebert TH, Lichenstein NS. Low 70. Hernandez-Nieto L, Brito-Barraso ML, Nyhan WL.
fractional excretion of sodium with contrast media- Megaloblastic anemia in Lesch-Nyhan disease.
induced acute renal failure. Arch Intern Med. 1980; Sangre. 1984;29:476.
140:531. 71. Van der Zee SPM, Monnens LAH, Schretlen EDAM.
53. Shafi T, Chou S-Y, Porush JS, Shapiro WB. Infusion Hereditary disorder of purine metabolism with cere-
intravenous pyelography and renal function. Effects bral affection and megaloblastic anemia. Ned Tijdschr
in patients with chronic renal insufficiency. Arch Geneeskd. 1968;112:1475.
Intern Med. 1978;138:1218. 72. Kelley WN, Greene ML, Rosenbloom FM, et al.
54. Older RA, Korobkin M, Cleeve DM, et al. Contrast- Hypoxanthine-guanine phosphoribosyltransferase in
induced acute renal failure. Persistent nephrogram as gout. Ann Intern Med. 1969;70:155.
a clue to early detection. AJR Am J Roentgenol. 73. Schneider W. The Lesch-Nyhan syndrome as a rare
1980;134:339. cause of hemolytic anemia. Acta Med Austriaca.
55. Lesch M, Nyhan WL. A familial disorder of uric acid 1979;6:202.
metabolism and central nervous system function. Am 74. Van der Zee SPM, Schretlen EDAM, Monnens LAH.
J Med. 1964;36:561. Megaloblastic anemia in the Lesch-Nyhan syndrome.
56. Hoefnagel D, Andrew ED, Mireault NG, Berndt WO. Lancet. 1968;1:1427.
Hereditary choreoathetosis, self-mutilation, and hyperu- 75. Zoref-Shani E, Feinstein S, Frishberg Y, et al. Kelley-
ricemia in young males. N Engl J Med. 1965;273:130. Seegmiller syndrome due to a unique variant of hypox-
57. Michener WM. Hyperuricemia and mental retardation anthine-guanine phosphoribosyltransferase: reduced
with athetosis and self-mutilation. Am J Dis Child. affinity for 5-phosphoribosyl-1-pyrophosphate mani-
1967;113:195. fested only at low, physiological substrate concentra-
58. Dreifuss FE, Newcombe DS, Shapiro SL, Sheppard tions. Biochim Biophys Acta. 2000;1500:197–203.
GL. X-linked primary hyperuricemia (hypoxanthine- 76. Wingen RM, Loffler W, Waldherr R, Scharer K. Acute
guanine phosphoribosyltransferase deficiency enceph- renal failure in an infant with partial deficiency of
alopathy). J Ment Defic Res. 1968;12:100. hypoxanthine-guanine phosphoribosyltransferase.
59. Munsat TL, Klinenberg J, Carrel RE, Menkes J. Proc Eur Dial Transplant Assoc Eur Ren Assoc.
Defects in purine metabolism and neurologic disease. 1985;21:751–5.
Bull Los Angeles Neurol Soc. 1968;33:101. 77. Sperling O, Boer P, Persky-Brosh S, et al. Altered
60. Berman PH, Balis ME, Dancis J. Congenital hyperu- kinetic property of erythrocyte phosphoribosyl-pyro-
ricemia, an inborn error of purine metabolism associ- phosphate synthetase in excessive purine production.
ated with psychomotor retardation, athetosis, and Rev Eur Etud Clin Biol. 1972;17:703.
self-mutilation. Arch Neurol. 1969;20:44. 78. Becker MA, Losman MJ, Kim M. Mechanisms of
61. Rosenberg D, Monnet P, Mamelle JL, Colonbel M, accelerated purine nucleotide synthesis in human
et al. Encephalopathie avec troubles due metabolisme fibroblasts with superactive phosphoribosylpyrophos-
des purines. Presse Med. 1968;76:2333. phate synthetases. J Biol Chem. 1987;262:5596.
62. Riley JD. Gout and cerebral palsy in a three-year-old 79. Zoref E, de Vries A, Sperling O. Mutant feed-
boy. Arch Dis Child. 1960;35:293. back-resistant phosphoribosylpyrophosphate syn-
63. Partington MW, Hennen BKE. The Lesch-Nyhan syn- thetase with purine overproduction and gout.
drome: self-destructive biting, mental retardation, Phosphoribosylpyrophosphate and purine metabolism
neurological disorder and hyperuricemia. Dev Med in cultured fibroblasts. J Clin Invest. 1975;56:1093.
Child Neurol. 1967;9:563. 80. Ahmed M, Taylor W, Smith PR, Becker MA.
64. Scherzer AL, Ilson JB. Normal intelligence in the Accelerated transcription of PRS1 in x-linked overac-
Lesch-Nyhan syndrome. Pediatrics. 1969;44:116. tivity of normal human phosphoribosylpyrophosphate
65. Buitelaar JK. Self-injurious behavior in retarded chil- synthetase. J Biol Chem. 1999;274:7482.
dren: clinical phenomena and biological mechanisms. 81. Becker MA, Puig JG, Mateos FA, et al. Inherited
Acta Paedopsychiatr. 1993;56:105–11. superactivity of phosphoribosylpyrophosphate syn-
66. Wong DF, Harris JC, Naidu S, et al. Dopamine trans- thetase: association of uric acid overproduction and
porters are markedly reduced in Lesch-Nyhan disease sensorineural deafness. Am J Med. 1988;85:393.
in vivo. Proc Natl Acad Sci USA. 1996;93:5539–43. 82. Becker MA, Raivio KO, Bakay B, et al. Variant human
67. Harris JC, Lee RR, Jinnah HA, et al. Craniocerebral phosphoribosylpyrophosphate synthetase altered in
magnetic resonance imaging measurement and regulatory and catalytic functions. J Clin Invest.
findings in Lesch-Nyhan syndrome. Arch Neurol. 1980;65:109.
1998;55:547–53. 83. Simmonds HA, Webster DR, Lingham S, Wilson J.
68. Visser JE, Bar PR, Jinnah H. Lesch-Nyhan disease An inborn error of purine metabolism, deafness and
and the basal ganglia. Brain Res Brain Res Rev. neurodevelopmental abnormality. Neuropediatrics.
2000;32:449–75. 1985;16:106.
References 157

84. Becker MA, Smith PR, Taylor W, et al. The genetic 100. Steinmann B, Gitzelmann R. Fruktose und sorbitol
and functional basis of purine nucleotide feedback infusions flussigkeiten sind nicht immer hamlos. Int
resistant phosphoribosylpyrophosphate synthetase J Vitam Nutr Res Suppl. 1976;15:239.
superactivity. J Clin Invest. 1995;96:2133. 101. Woolliscroft JO, Colfer H, Fox IH. Hyperuricemia in
85. Zoref E, de Vries A, Sperling O. Metabolic coopera- acute illness: a poor prognostic sign. Am J Med.
tion between human fibroblasts with normal and with 1982;72:58.
mutant superactive phosphoribosylpyrophosphate 102. Woolliscroft JO, Fox IH. Increased body fluid
synthetase. Nature. 1976;260:787. purines during hypotensive events: evidence for ATP
86. Yen RCK, Adams WB, Lazar C, Becker MA. degradation. Am J Med. 1986;81:472.
Evidence for x-linkage of human phosphoribosyl- 103. Grum CM, Simon RH, Dantzker DR, Fox IH.
pyrophosphate synthetase. Proc Natl Acad Sci USA. Biochemical indicators of cellular hypoxia in criti-
1978;75:482. cally ill patients: evidence for ATP degradation.
87. Simmonds HA, Webster DR, Wilson J, Lingham S. Chest. 1985;88:763.
An X-linked syndrome characterized by hyperuricae- 104. Braunwald E, Kloner RA. Myocardial reperfusion: a
mia, deafness, and neurodevelopmental abnormali- double-edged sword? J Clin Invest. 1985;76:1713.
ties. Lancet. 1982;2:68. 105. Christensen EF, Jacobsen J, Anker-Moller E, et al.
88. Henderson JF, Caldwell IC, Patterson ARP. Increased urinary loss of uric acid in adults with
Decreased feedback inhibition in a 6-methylmercap- acute respiratory failure requiring mechanical venti-
topurine ribonucleoside-resistant tumor. Cancer Res. lation. Chest. 1992;102:556.
1967;27:1773. 106. Braghiroli A, Sacco C, Erbetta M, et al. Overnight
89. Nagy M. Regulation of the biosynthesis of purine urinary uric acid: creatinine ration for detection of
nucleotides in Schizosaccharomyces pombe. I. sleep hypoxemia. Validation study in chronic
Properties of the phosphoribosylpyrophosphate: glu- obstructive pulmonary disease and obstructive sleep
tamine amidotransferase of the wild strain and of a apnea before and after treatment with nasal continu-
mutant desensitized towards feedback modifiers. ous positive airway pressure. Am Rev Respir Dis.
Biochim Biophys Acta (Amst). 1971;198:471. 1993;148:173.
90. Fox IH. Metabolic basis for disorders of purine nucle- 107. Mateos AF, Puig JG, Gomez Fernandez P, et al.
otide degradation. Metabolism. 1981;30:616. Degradation of purine nucleotides in patients with
91. Fox IH, Palella TD, Kelley WN. Hyperuricemia: a marker chronic obstructive airflow. Med Clin (Barc). 1989;
for cell energy crisis. N Engl J Med. 1987;317:111. 92:328.
92. Fox IH. Adenosine triphosphate degradation in 108. Karlsson J, Willerson JT, Leshin SJ, et al. Skeletal
specific disease. J Lab Clin Med. 1985;106:101. muscle metabolites in patients with cardiogenic
93. Alexander D, Assaf M, Khudr A, et al. Fructose-1,6- shock or severe congestive heart failure. Scand J
diphosphatase deficiency: diagnosis using leukocytes Clin Lab Invest. 1975;35:73.
and detection of heterozygotes with radiochemical 109. Buhl L, Vilhelmsen KN, Nielsen JR. Oxypurine release
and spectrophotometric methods. J Inherit Metab Dis. in cardiac disease. Acta Med Scand. 1981;209:83.
1985;8:147. 110. Sangstad OD. Hypoxanthine as a measurement of
94. Gitzelmann R, Steinmann B, Van den Berghe G. hypoxia. Pediatr Res. 1975;9:158.
Disorders of fructose metabolism. In: Scriver CR, 111. Harkness RA, Simmonds RJ, Coade SB, Lawrence
Beaudet AL, Sly WS, Valle D, editors. The metabolic CR. Ratio of the concentration of hypoxanthine to
and molecular bases of inherited disease. New York: creatinine in urine from newborn infants: a possible
McGraw-Hill, Inc; 1995. p. 905–34. indicator for the metabolic damage due to hypoxia.
95. Melancon SB, Khachadurian AK, Nadler HL, Brown Br J Obstet Gynaecol. 1983;90:447.
BI. Metabolic and biochemical studies in fructose- 112. Harkness RA, Whitelow AGL, Simmonds RJ.
1,6-diphosphatase deficiency. J Pediatr. 1973;82:650. Intrapartum hypoxia: the association between neuro-
96. Baerlocher K, Gitzelmann R, Nassli R, Dumermuth logical assessment of damage and abnormal excre-
G. Infantile lactic acidosis due to hereditary fructose- tion of ATP metabolites. J Clin Pathol. 1982;35:999.
1,6-diphosphatase deficiency. Helv Paediatr Acta. 113. Manzke H, Dorner K, Grunitz J. Urinary hypoxan-
1971;26:489. thine, xanthine and uric acid excretion in newborn
97. Hopwood NJ, Holzman I, Drash AL. Fructose-1,6- infants with perinatal complications. Acta Paediatr
diphosphatase deficiency. Am J Dis Child. 1977; Scand. 1977;66:713.
131:418. 114. Jensen MH, Brinklov MM, Lillquist K. Urinary loss
98. Corbell L, Eggermont E, Eeckels R, et al. Recurrent of oxypurines in hypoxic premature neonates. Biol
attacks of ketotic acidosis associated with fructose- Neonate. 1980;38:40.
1,6-diphosphatase deficiency. Acta Paediatr Belg. 115. Raivio KO. Neonatal hyperuricemia. J Pediatr.
1976;29:29. 1976;88:625.
99. Corbell LM, Eggermont E, Bettens W, et al. Fructose 116. Swanstrom S, Bratteby L. Hypoxanthine as a test of
intolerance with normal liver aldolase. Helv Paediatr perinatal hypoxia as compared to lactate, base deficit,
Acta. 1970;25:626. and pH. Pediatr Res. 1982;16:156.
158 5 Clinical Aspects of Gout and Associated Disease States

117. Thiringer K. Cord plasma hypoxanthine as a mea- catalytically inactive mutant M-type subunit. Am
sure of foetal asphyxia. Acta Paediatr Scand. J Hematol. 1982;12:215–25.
1983;72:231. 137. Tarui S, Okuno G, Ikura Y, et al. Phosphofructokinase
118. Fruhmann G, Fritz H, Bergstermann H. Homozygous deficiency in skeletal muscle: a new type of glycog-
inherited alpha1-antitrypsin deficiency with emphy- enosis. Biochem Biophys Res Commun. 1963;19:
sema of the lung, cor pulmonale, and gout. Klin 517–23.
Wochenschr. 1974;52:80. 138. Ambrose DR, McCabe ER, Anderson D, Beaudet A,
119. Kohkhar N. Hyperuricemia and gout in secondary et al. Infections and bleeding complications in
polycythemia due to chronic obstructive pulmonary patients with glycogenosis Ib. Am J Dis Child. 1985;
disease. J Rheumatol. 1980;7:114. 139:691–7.
120. Ball GV, Sorensen LB. Pathogenesis of hyperurice- 139. Soejimak K, Landing BH, Roe TF, Swanson VL.
mia and gout in sickle cell anemia. Arthritis Rheum. Pathologic studies of the osteoporosis of Von
1970;13:846. Gierke’s disease (glycogenosis Ia). Pediatr Pathol.
121. March HW, Schylen SM, Schwartz SE. Mediterranean 1985;3:307–19.
hemopathic syndromes (Cooley’s anemia) in adults: 140. De Parscau L, Guiband P, Labrime P, Odievre M.
study of a family with unusual complications. Am Long-term course of hepatic glycogenosis. A retro-
J Med. 1952;13:46. spective study of 76 cases. Arch Fr Pediatr.
122. Paik CH, Alavi I, Dunea G, Weiner L. Thalassemia 1988;45:641–5.
and gouty arthritis. JAMA. 1970;213:296. 141. Chen YT, Coleman RA, Scheinman JI, Kolbeck PC,
123. Hickling RA. Gout, leukemia, and polycythaemia. Sidbury JB. Renal disease in type I glycogen storage
Lancet. 1953;1:57. disease. N Engl J Med. 1988;318:7–11.
124. Talbott JH. Gout. New York: Grune & Stratton; 142. Baker L, Dahlem S, Goldfarb S, et al. Hyperfiltration
1957. and renal disease in glycogen storage disease, type I.
125. Yu TF. Secondary gout associated with myeloprolif- Kidney Int. 1989;5:1345–50.
erative disease. Arthritis Rheum. 1965;8:765. 143. Restaino I, Kaplan BS, Stanley C, et al.
126. Bronsky D, Bernstein A. Acute gout secondary to Nephrolithiasis, hypocitraturia, and a distal renal
multiple myeloma: a case report. Ann Intern Med. tubular acidification defect in type 1 glycogen stor-
1954;41:820. age disease. J Pediatr. 1993;122:392.
127. Somerville J. Gout in cyanotic congenital heart dis- 144. Matsuo N, Tsuchiya Y, Cho H, et al. Proximal renal
ease. Br Heart Med. 1961;23:31. tubular acidosis in a child with type 1 glycogen stor-
128. Zimmer JG, Demus DJ. Associations between gout, age disease. Acta Paediatr Scand. 1986;75:332.
psoriasis and sarcoidosis: with consideration of 145. Chen YT, Scheinman JI, Park HK, et al. Amelioration
their pathologic significance. Ann Intern Med. of proximal renal tubular dysfunction in type 1 gly-
1966;64:786. cogen storage disease with dietary therapy. N Engl
129. Van den Berghe G, Hers HG. Abnormal AMP deam- J Med. 1990;323:590.
inase in primary gout. Lancet. 1980;2:1090. 146. Howell RR, Stevenson RE, Ben-Menachem Y,
130. Hers HG, Van den Berghe G. Enzyme defect in pri- Phyliky RL, Berry DH. Hepatic adenomata with
mary gout. Lancet. 1979;1:585. type I glycogen storage disease. JAMA.
131. DiMauro S, Miranda AF, Hays AP, et al. 1976;236:1481.
Myoadenylate deaminase deficiency – muscle biopsy 147. Limmer J, Fleig WE, Leupold D, et al. Hepatocellular
and muscle culture in a patient with gout. J Neurol carcinoma in type I glycogen storage disease.
Sci. 1980;47:191–202. Hepatology. 1988;8:531.
132. Sabina RL, Swain JL, Olanow CW, et al. 148. Fink AS, Appelman HD, Thompson NW.
Myoadenylate deaminase deficiency. Functional and Hemorrhage into a hepatic adenoma and type Ia gly-
metabolic abnormalities associated with disruption cogen storage disease: a case report and review of
of the purine nucleotide cycle. J Clin Invest. the literature. Surgery. 1985;97:117.
1984;73:720. 30. 149. Levy E, Thibault LA, Roy CC, et al. Circulating lip-
133. Sabina RL, Holmes EW. Myoadenylate deaminase ids and lipoproteins in glycogen storage disease type
deficiency. In: Scriver CR, Beaudet AL, Sly WS, I with nocturnal intragastric feeding. J Lipid Res.
Valle D, editors. The metabolic and molecular bases 1988;29:215.
of inherited disease. New York: McGraw-Hill, Inc; 150. Beaudet AL, Anderson DC, Michelis VV, Arion WJ,
1995. p. 1769–80. Lange AJ. Neutropenia and impaired neutrophil
134. Talente GM, Coleman RA, Alter C, et al. Glycogen migration in type Ib glycogen storage disease.
storage disease in adults. Ann Intern Med. 1994;120: J Pediatr. 1980;97:906.
218. 151. Bartram CR, Przyrembel H, Wendel U, et al.
135. Abarbanel JM, Potashnik R, Frisher S, et al. Glycogenosis type Ib complicated by severe granu-
Myophosphorylase deficiency: the course of an locytopenia resembling inherited neutropenia. Eur
unusual congenital myopathy. Neurology. 1987;37: J Pediatr. 1981;137:81.
316. 152. Murase T, Ikeda H, Muro T, Nakao K, Sugita H.
136. Zanella A, Mariani M, Meola G, Fagnani G, Sirchia Myopathy associated with type III glycogenosis.
G. Phosphofructokinase (PFK) deficiency due to a J Neurol Sci. 1973;20:287.
References 159

153. Brunberg JA, McCormick WF, Schochet SS. Type 172. Higgs JB, Blaivas M, Albers JW. McArdle’s disease
III glycogenosis. An adult with diffuse weakness presenting as treatment resistant polymyositis.
and muscle wasting. Arch Neurol. 1971;25:171. J Rheumatol. 1989;16:1588.
154. Moses SW, Gadoth N, Bashan N, et al. Neuromuscular 173. Vora S, Davidson M, Seaman C, et al. Heterogeneity
involvement in glycogen storage disease type III. of the molecular lesions in inherited phosphofruc-
Acta Paediatr Scand. 1986;75:289. tokinase deficiency. J Clin Invest. 1983;72:
155. Moses SW, Wanderman KL, Myroz A, Frydman M. 1995–2006.
Cardiac involvement in glycogen storage disease 174. Hays AP, Hallet M, Delfs J, et al. Muscle phospho-
type III. Eur J Pediatr. 1989;148:764. fructokinase deficiency: abnormal polysaccharide in
156. Coleman RA, Winter HA, Wolf B, et al. Glycogen a case of late-onset myopathy. Neurology (NY).
storage disease type III (glycogen debranching 1981;31:1077–86.
enzyme deficiency): correlation of biochemical 175. Agamanolis DP, Askari AD, DiMauro S, et al.
defects with myopathy and cardiomyopathy. Ann Muscle phosphofructokinase deficiency: two cases
Intern Med. 1992;116:896. with unusual polysaccharide accumulation and
157. DiMauro S, Hartwig GB, Hays A, et al. Debrancher immunologically active enzyme protein. Muscle
deficiency: neuromuscular disorder in 5 adults. Ann Nerve. 1980;3:456–67.
Neurol. 1979;5:422. 176. Zanella A, Mariani M, Meola G, et al.
158. Cornelio F, Bresolin N, Singer PA, et al. Clinical Phosphofructokinase (PFK) deficiency due to a cata-
varieties of neuromuscular disease in debrancher lytically inactive mutant M-type subunit. Am J
deficiency. Arch Neurol. 1971;25:171. Hematol. 1982;12:215–25.
159. Labrime PL, Huguet P, Odievre M. Cardiomyopathy 177. Nakagawa C, Mineo I, Kaido M, et al. A new variant
in glycogen-storage disease type III: clinical and case of muscle phosphofructokinase deficiency,
echocardiographic study of 18 patients. Pediatr coexisting with gastric ulcer, gouty arthritis, and
Cardiol. 1991;12:161. increased hemolysis. Muscle Nerve. 1995;3:
160. Olson LJ, Reeder GS, Noller KL, et al. Cardiac 539–44.
involvement in glycogen storage disease III: mor- 178. Danon MJ, Servidei S, DiMauro S, Vora S. Late-
phologic and biochemical characterization with onset muscle phosphofructokinase deficiency.
endomyocardial biopsy. Am J Cardiol. 1984; Neurology. 1988;38:956.
53:980. 179. Serratrice G, Monges A, Roux H, et al. Forme myo-
161. Miller CG, Alleyne GA, Brooks SEH. Gross cardiac pathique du deficit en phosphofructokinase. Rev
involvement in glycogen storage disease type III. Br Neurol. 1969;120:271.
Heart J. 1972;34:862. 180. Servidei S, Bonilla E, Diedrick RG, et al. Fatal
162. van Crevold S. The Blackader lecture, 1961: the infantile form of muscle phosphofructokinase
clinical course of glycogen disease. Can Med Assoc deficiency. Neurology. 1986;36:1465.
J. 1963;88:1–15. 181. Danon MJ, Carpenter S, Manaligod JR, Schlisefeld LH.
163. Mineo I, Kono N, Hara N, et al. Myogenic hyperuri- Fatal infantile glycogen storage disease: deficiency
cemia. A common pathologic feature of glycogeno- of phosphofructokinase and phosphorylase b kinase.
sis type III, V and VII. N Engl J Med. 1987;317: Neurology. 1981;31:1303.
75–80. 182. Vora S, Corash L, Engel WK, et al. The molecular
164. Puig JG, de Miguel E, Mateos FA, et al. McArdle’s mechanism of the inherited phosphofructokinase
disease and gout. Muscle Nerve. 1992;15:822–8. deficiency associated with hemolysis and myopathy.
165. Hardiman O, Farrell M, McElvaney G, et al. Blood. 1980;55:629–35.
Hyperuricemia in type V glycogenosis. Neurology. 183. Shimizu T, Kono N, Kiyokawa H, et al. Erythrocyte
1987;37:728–9. glycolysis and its marked alterations by muscular
166. Servidei S, DiMauro S. Disorders of glycogen exercise in type VII glycogenosis. Blood. 1988;71:
metabolism of muscle. Neurol Clin. 1989;7:159–78. 1130.
167. Moses SW. Muscle glycogenosis. J Inherit Metab 184. Haller RG, Lewis SF. Glucose-induced exertional
Dis. 1990;13:452. fatigue in muscle phosphofructokinase deficiency. N
168. Pourmand R, Sanders DB, Corwin HM. Late-onset Engl J Med. 1991;324:364.
McArdle’s disease with unusual electromyographic 185. Tarui S, Kono N, Kuwajima M, Ikura Y. Type VII
findings. Arch Neurol. 1983;40:374. glycogenosis (muscle and erythrocyte phosphofruc-
169. Hewlett RH, Gardner-Thorpe C. McArdle’s Disease – tokinase deficiency). Monogr Hum Genet. 1978;9:
what limit to the age of onset? S Afr Med J. 42–7.
1978;53:60. 186. Tarui S, Kono N, Nasu T, Nishikawa M. Enzymatic
170. Mineo I, Kono N, Shimizu T, et al. Excess purine basis for the coexistence of myopathy and hemolytic
degradation in exercising muscles of patients with disease in inherited phosphofructokinase deficiency.
glycogen storage disease types V and VII. J Clin Biochem Biophys Res Commun. 1969;34:77–83.
Invest. 1985;76:556–60. 187. Tem I. Metabolic myopathies. Pediatr Neurol.
171. Jinnai K, Kono N, Yamamoto Y, et al. Glycogenosis 1996;3:53–98.
type V (McArdle’s disease) with hyperuricemia. Eur 188. DiMauro S, Miranda AF, Khan S, et al. Human
Neurol. 1993;33:204–7. muscle phosphoglycerate mutase deficiency and
160 5 Clinical Aspects of Gout and Associated Disease States

myoadenylate deaminase deficiency. Science. 205. Fujii H, Miwa S. Other erythrocyte enzyme
1981;212:1277–9. deficiencies associated with non-haematological
189. Bertorini TE, Shively V, Taylor B, et al. ATP degra- symptoms: phosphoglycerate kinase and phospho-
dation products after ischemic exercise: hereditary fructokinase deficiency. Baillieres Best Pract Res
lack of phosphorylase or carnitine palmitoyltrans- Clin Haematol. 2000;13:141–8.
ferase. Neurology. 1985;35:1355–7. 206. Aasly J, van Diggelen OP, Boer AM, Bronstad G.
190. Abaranel JM, Bashan N, Potashnik R, et al. Adult Phosphoglycerate kinase deficiency in two brothers
phosphorylase b kinase deficiency. Neurology. with McArdle-like clinical symptoms. Eur J Neurol.
1986;36:560. 2000;7:111–3.
191. Servidei S, Metlay LA, Booth FA, et al. Clinical and 207. Toscano A, Tsujino S, Vita G, et al. Molecular basis
biochemical heterogeneity of phosphorylase kinase of muscle phosphoglycerate mutase (PGAM-M)
deficiency. Neurology. 1987;37 Suppl 1:139. deficiency in an Italian kindred. Muscle Nerve.
192. Clemens PR, Yamamoto M, Engel AG. Adult phos- 1996;19:1134–7.
phorylase b kinase deficiency. Ann Neurol. 1990; 208. Bresolin N, Ro Y, Reyes M, et al. Muscle phospho-
28:529. glycerate mutase (PGAM) deficiency: a second case.
193. Laforet P, Eymard B, Lombes A, et al. Intolerance a Neurology. 1983;33:1049–53.
l’effort par deficit en phosphorylase b kinase muscu- 209. Tsujino S, Shanske S, Sakoda S, et al. The molecular
laire. Apport des investigations metaboliques in vivo. genetic basis of muscle phosphoglycerate mutase
Rev Neurol (Paris). 1996;152:458–64. (PGAM) deficiency. Am J Hum Genet. 1993;52:472–7.
194. Fukuda T, Sugie H, Sugie Y, et al. Five cases of 210. Poulton KR, Khan AA, Rossi ML, Riddoch D.
phosphorylase b kinase deficiency affecting muscle Muscle phosphoglycerate mutase deficiency: a study
or liver: clinical symptoms and diagnosis. No To of a family with metabolic myopathy. Funct Neurol.
Hattatsu. 1994;26:493–7. 1994;9:47–58.
195. Wilkinson DA, Tonin P, Shanske S, et al. Clinical 211. DiMauro S, Miranda AF, Olarte M, et al. Muscle
and biochemical features of 10 adult patients with phosphoglycerate mutase deficiency. Neurology.
muscle phosphorylase kinase deficiency. Neurology. 1982;32:584–91.
1994;44:461–6. 212. Tsujino S, Shanske S, Sakoda S, et al. Molecular
196. Carrier H, Maire I, Vial C, et al. Myopathic evolu- genetic studies in muscle phosphoglycerate mutase
tion of an exertional muscle pain syndrome with (PGAM-M) deficiency. Muscle Nerve. 1995;3:S50–3.
phosphorylase b kinase deficiency. Acta Neuropathol 213. DiMauro S, Miranda AF, Khan S, et al. Human mus-
(Berlin). 1990;81:84–8. cle phosphoglycerate mutase deficiency: newly dis-
197. Van den Berg IF, Berger R. Phosphorylase b kinase covered metabolic myopathy. Science. 1981;212:
deficiency in man: a review. J Inherit Metab Dis. 1277–9.
1990;13:442–51. 214. Kanno T, Sudo K, Takeuchi I, et al. Hereditary
198. Madloni M, Berley GT, Cohen PT, Marrian VJ. deficiency of lactate dehydrogenase M-subunit. Clin
Phosphorylase b kinase deficiency in a boy with gly- Chim Acta. 1980;108:267–76.
cogenosis affecting both liver and muscle. Eur J 215. Miyajima H, Shimizu T, Kaneko E. Gene expression
Pediatr. 1989;149:52–3. in lactate dehydrogenase-A subunit deficiency.
199. Keating JP, Brown BI, White NH, DiMauro S. Rinsho Shinkeigaku. 1992;32:1087–92.
X-linked glycogen storage disease. A cause of hypo- 216. Miyajima H, Takahashi Y, Suzuki M, et al. Molecular
tonia, hyperuricemia, and growth retardation. Am J characterization of gene expression in human lactate
Dis Child. 1985;139:609–13. dehydrogenase-A deficiency. Neurology. 1993;43:
200. Sugie H, Sugie Y, Nishida M, et al. Recurrent myo- 1414–9.
globinuria in a child with mental retardation: phos- 217. Miyajima H, Takahashi Y, Kaneko E. Characterization
phoglycerate kinase deficiency. J Child Neurol. of the oxidative metabolism in lactate dehydroge-
1989;4:95–9. nase A deficiency. Intern Med. 1995;34:502–6.
201. Tonin P, Shanske S, Miranda AF, et al. Phosphogly- 218. Miyajima H, Takahashi Y, Kaneko E. Characterization
cerate kinase deficiency: biochemical and molecular of the glycolysis in lactate dehydrogenase-A
genetic studies in a new myopathic variant (PGK deficiency. Muscle Nerve. 1995;18:874–8.
Alberta). Neurology. 1993;43:387–91. 219. Takahashi Y, Miyajima H, Kaneko E. Genetic analy-
202. Rosa R, George C, Fardeau M, et al. A new case of sis of a family of lactate dehydrogenase-A subunit
phosphoglycerate kinase deficiency: PGK Creteil deficiency. Intern Med. 1995;34:326–9.
associated with rhabdomyolysis and lacking hemo- 220. Takahashi Y, Fujiwara S, Waki T. Hereditary lactate
lytic anemia. Blood. 1982;60:84–91. dehydrogenase M-subunit deficiency: lactate dehy-
203. Tsujino S, Shanske S, DiMauro S. Molecular genetic drogenase activity in skin lesions and in hair folli-
heterogeneity of phosphoglycerate kinase (PGK) cles. J Am Acad Dermatol. 1991;24:339–42.
deficiency. Muscle Nerve. 1995;8:545–9. 221. Yoshikuni K, Tagami H, Yamada M, et al.
204. DiMauro S, Dalakas M, Miranda AF. Phosphoglycerate Erythematosquamous skin lesions in hereditary lac-
kinase deficiency: another cause of recurrent myo- tate dehydrogenase M-subunit deficiency. Arch
globinuria. Ann Neurol. 1983;13:11–9. Dermatol. 1986;122:1420–4.
References 161

222. Kanno T, Maekawa M. Lactate dehydrogenase 239. Steele TH, Boner G. Origins of the uricosuric
M-subunit deficiencies: clinical features, metabolic response. J Clin Invest. 1973;52:168.
background, and genetic heterogeneities. Muscle 240. Diamond HS, Paolino JS. Evidence for a post-secre-
Nerve. 1995;3:S54–60. tory reabsorptive site for uric acid in man. J Clin
223. DiMauro S, DiMauro PMM. Muscle carnitine palm- Invest. 1973;52:1491.
itoyltransferase deficiency and myoglobinuria. 241. Steele TH. Urate secretion in man: the pyrazinamide
Science. 1973;182:929. suppression test. Ann Intern Med. 1973;79:734.
224. Demaugre F, Bonnefont JP, Colonna M, et al. 242. Rieselbach RE, Steele TH. Influence of the kidney
Infantile form of carnitine palmitoyltransferase II upon urate homeostasis in health and disease. Am
deficiency with hepatomuscular symptoms and sud- J Med. 1974;56:665.
den death Physiopathological approach to carnitine 243. de Rougement D, Henchoz M, Roch-Rumel F. Renal
palmitoyltransferase II deficiency. J Clin Invest. urate excretion at various plasma concentrations in
1991;87:859. the rat: a free-flow micropuncture study. Am
225. Hug G, Bove KE, Soukup S. Lethal neonatal multio- J Physiol. 1976;231:387.
rgan deficiency of carnitine palmitoyltransferase II. 244. Weinman EJ, Senekjian HO, Sansom SC, et al.
N Engl J Med. 1991;325:1862. Evidence for active and passive urate transport in the
226. Zinn AB, Zurcher VL, Krans F, et al. Carnitine rat proximal tubule. Am J Physiol. 1981;240:F90.
palmitoyltransferase B (CPT B) deficiency: a heri- 245. Sorensen LB, Levinson DJ. Isolated defect in postse-
table cause of neonatal cardiomyopathy and dysgen- cretory reabsorption of uric acid. Ann Rheum Dis.
esis of the kidney. Pediatr Res. 1991;29:73A. 1980;39:180.
227. Katzir Z, Hookman B, Biro A, et al. Carnitine palmi- 246. Gibson T, Sims HP, Jiminez SA. Hypouricemia and
toyltransferase deficiency: an underdiagnosed condi- increased renal urate clearance associated with hyper-
tion. Am J Nephrol. 1996;16:162–6. parathyroidism. Ann Rheum Dis. 1976;35:372.
228. Faigel HC. Carnitine palmitoyltransferase deficiency 247. Wyngaarden JB, Kelley WN. Gout. In: Stanbury JB,
in a college athlete: a case report and literature Wyngaarden JB, Fredrickson DS, Goldstein JL,
review. J Am Coll Health. 1995;44:51–4. Brown MS, editors. The metabolic basis of inherited
229. Stavem K, Bjerke G, Skullerud K, Bohmer T. disease. 5th ed. New York: McGraw-Hill, Inc; 1983.
Rhabdomyolysis in carnitine palmitoyltransferase p. 1043–5.
deficiency. Tidsskr Nor Laegeforen. 1994;114: 248. Gutman AB, Yu TF. Renal function in gout: with a
2398–9. commentary on the renal regulation of urate excre-
230. Kiechl S, Kohlendorfer U, Paetzke I, et al. tion and the role of the kidney in the pathogenesis of
Myoglobinuria and palmitoyltransferase deficiency. gout. Am J Med. 1957;23:600.
Diagnostic procedure and differential diagnosis. 249. Simkin PA. Uric acid excretion in patients with gout.
Wien Klin Wochenschr. 1994;106:174–7. Arthritis Rheum. 1979;22:98.
231. Werneck LC, Boer CA, Papadimitriou A, DiMauro S. 250. Nugent CA, MacDiarmid WD, Tyler FH. Renal
Myopathy due to carnitine palmitoyltransferase excretion of urate in patients with gout. Arch Intern
deficiency. Report of 2 cases with enzymatic analy- Med. 1964;113:115.
ses on muscle tissue. Arq Neuropsiquiatr. 1983;41: 251. Houpt JB, Ogryzlo MA. Persistence of impaired uric
377–84. acid excretion in gout during reduced synthesis with
232. Herman J, Nadler HL. Recurrent myoglobinuria and allopurinol. Arthritis Rheum. 1964;7:316.
muscle carnitine palmitoyltransferase deficiency. 252. Snaith ML, Scott JT. Uric acid clearance in patients
J Pediatr. 1977;91:247–50. with gout and normal subjects. Ann Rheum Dis.
233. Kieval RI, Sotrel A, Weinblatt ME. Chronic myopa- 1971;30:285.
thy with a partial deficiency of the carnitine palmitoyl- 253. Seegmiller JE, Grayzel AI, Howell RR, Plato C. The
transferase enzyme. Arch Neurol. 1990;46:575–6. renal excretion of uric acid in gout. J Clin Invest.
234. Carroll JE, Brooke MH, DeVivo DC, et al. 1962;41:1094.
Biochemical and physiologic consequences of carni- 254. Lathem W, Rodnan GP. Impairment of uric acid
tine palmitoyltransferase deficiency. Muscle Nerve. excretion in gout. J Clin Invest. 1962;41:1955.
1978;1:103–10. 255. Wyngaarden JB. Gout. Adv Metab Disord.
235. Layzer RB, Havel RJ, McIlroy MB. Partial deficiency 1965;2:2.
of carnitine palmitoyltransferase: physiologic and 256. Simkin PA. Urate excretion in normal and gouty
biochemical consequences. Neurology. 1980;30: men. In: Miller MM, Kaiser E, Seegmiller JE, edi-
627–33. tors. Purine metabolism in man II. New York:
236. Gieron MA, Karthais JK. Carnitine palmitoyltrans- Plenum; 1977. p. 41.
ferase deficiency with permanent weakness. Pediatr 257. Yu TF, Berger L, Stone DJ, et al. Effects of pyrazin-
Neurol. 1987;3:51–3. amide and pyrazinoic acid on urate clearance and
237. Wyngaarden JB, Kelley WN. Gout and hyperuricemia. other discrete renal functions. Proc Soc Exp Biol
New York: Grune & Stratton, Inc.; 1976. p. 111–7. Med. 1957;96:264.
238. Sorensen LB. The pathogenesis of gout. Arch Intern 258. Steele TH, Reiselbach RE. The contribution of resid-
Med. 1962;109:379. ual nephrons within the chronically diseased kidney
162 5 Clinical Aspects of Gout and Associated Disease States

to urate homeostasis in man. Am J Med. 1967;43: 279. Berger L, Yu TF. Renal function in gout. IV. An
876. analysis of 524 gouty subjects including long-term
259. Steele TH. Evidence for altered renal urate reabsorp- follow-up studies. Am J Med. 1975;59:605.
tion during changes in volume of the extracellular 280. Palella TD, Fox IH. Hyperuricemia and gout. In:
fluid. J Lab Clin Med. 1969;74:228. Scriver CR, Beaudet AL, Sly WS, Valle D, editors.
260. Suki WN, Eknoyan G, Martinez-Maldonado M. The metabolic basis of inherited disease. New York:
Tubular sites and mechanisms of diuretic action. McGraw-Hill, Inc.; 1989. p. 971.
Annu Rev Pharmacol. 1973;13:91. 281. Heptinstall RH. Pathology of the kidney. Boston:
261. Steele TH, Oppenheimer S. Factors affecting urate Little, Brown; 1966.
excretion following diuretic administration in man. 282. Seegmiller JE, Frazier PD. Biochemical consider-
Am J Med. 1969;47:564. ations of the renal damage of gout. Ann Rheum Dis
262. Yu TF, Gutman AB. Study of the paradoxical effects (Suppl). 1966;25:668–72.
of salicylate in low, intermediate and high dosage on 283. Gonick HC, Rubini ME, Gleason IO, Sommers SC.
the renal mechanisms for excretion of urate in man. The renal lesion in gout. Ann Intern Med.
J Clin Invest. 1959;38:1298. 1965;62:667.
263. Goldfinger S, Klinenberg JR, Seegmiller JE. Renal 284. Pardo V, Perez-Stable E, Fisher ER. Ultrastructure
retention of uric acid induced by infusion of beta- studies in hypertension. III. Gouty nephropathy. Lab
hydroxybutyrate and acetoacetate. N Engl J Med. Invest. 1968;18:143.
1965;272:351. 285. Verger D, Leroux-Robert GP, Richet G. Depots
264. Schulman JD, Lustberg TJ, Kennedy JL, et al. A new d’urate intrarenaux chez les insuffisants renaux chro-
variant of maple syrup urine disease (branched chain niques hyperuricemiques. Urol Nephrol. 1967;73:
ketoaciduria). Clinical and biochemical evaluation. 314.
Am J Med. 1970;49:118. 286. Linnane JW, Burry AF, Emmerson BT. Urate depos-
265. Yu TF, Sirota JH, Berger L, et al. Effect of sodium its in the renal medulla: prevalence and associations.
lactate infusion on urate clearance in man. Proc Soc Nephron. 1981;27:216–22.
Exp Biol Med. 1957;96:809. 287. Mukarami T, Kawakami H, Nakatsuda K, et al.
266. Talbott JH, Terplan KL. The kidney in gout. Underexcretory-type hyperuricemia, disproportion-
Medicine. 1960;39:405–62. ate to the reduced glomerular filtration rate, in two
267. Beck LH. Requiem for gouty nephropathy. Kidney boys with mild proteinuria. Nephron. 1990;56:
Int. 1986;30:280. 439–42.
268. Foley RJ, Weinman EJ. Review: urate nephropathy. 288. Farebrother DA, Pincott JR, Simmonds HA, et al.
Am J Med Sci. 1984;288:208. Uric acid crystal-induced nephropathy: evidence for
269. Gudzent F. Gicht and rheumatismus. Berlin: a specific renal lesion in a gouty family. J Pathol.
Springer; 1928. 1981;135:159–68.
270. Mayne JG. Pathological study of the renal lesion 289. Iwata J. Hyperuricemia in hematological disorders.
found in 27 patients with gout. Ann Rheum Dis. Nippon Rinsho. 1996;54:3349–53.
1955;15:61. 290. Agha-Razii M, Amyot SL, Pichette V, et al.
271. Yu TF, Berger L, Dorph DJ. Renal function in gout. Continuous veno-venous hemodiafiltration for the
V. Factors influencing renal hemodynamics. Am J treatment of spontaneous tumor lysis syndrome
Med. 1979;67:766. complicated by acute renal failure and severe hype-
272. Yu TF, Berger L. Renal function in gout. Its associa- ruricemia. Clin Nephrol. 2000;54:59–63.
tion with hypertensive vascular disease and intrinsic 291. Cohen LF, Balow JE, Magrath IT, et al. Acute tumor
renal disease. Am J Med. 1982;72:95. lysis syndrome. A review of 37 patients with
273. Campbell BC, Moore MR, Goldberg A. Subclinical Burkitt’s lymphoma. Am J Med. 1980;68:486–91.
lead exposure: a possible cause of gout. Br Med J. 292. Dubovsky D, Jacobs P. Acute uric acid nephropathy
1978;2:1403. in thalessaemia. S Afr Med J. 1975;49:243–4.
274. Batuman V, Maesaka JK, Haddad B, et al. The role 293. Conger JD. Acute uric acid nephropathy. Med Clin
of lead in gout nephropathy. N Engl J Med. North Am. 1990;74:859–71.
1981;304:520. 294. Barton JC. Tumor lysis syndrome in nonhematopoi-
275. Hall AP, Barry DP, Dawber TR, McNamara PM. etic neoplasms. Cancer. 1989;64:738–40.
Epidemiology of gout and hyperuricemia; a long- 295. Hande KR, Garrow GC. Acute tumor lysis syndrome
term population study. Am J Med. 1967;42:27. in patients with high grade non-Hodgkin’s lym-
276. Fessel WJ. Renal outcomes in gout and hyperurice- phoma. Am J Med. 1993;94:133–8.
mia. Am J Med. 1979;67:74. 296. Kjellstrand CM, Campbell DC, von Hartikch B,
277. Fessel WJ, Barr GD. Uric acid, lean body weight, Buselmeier TJ. Hyperuricemic acute renal failure.
and creatinine interactions. Results from regression Arch Intern Med. 1974;133:349.
analyses of 78 variables. Semin Arthritis Rheum. 297. Chastyl RC, Liu-Yin JA. Acute tumor lysis syn-
1970;7:115. drome. Br J Hosp Med. 1993;49:488–92.
278. Campion EW, Glynn RJ, DeLabry LO. Asymptomatic 298. Warren DJ, Leitch AG, Leggett RJE. Hyperuricaemic
hyperuricemia. Risks and consequences in the acute renal failure after epileptic seizures. Lancet.
Normative Aging Study. Am J Med. 1987;82:421. 1975;2:385–7.
References 163

299. Hess B, Binswanger U. Acute uric acid nephropathy 317. Torres VE, Rastogi S, King BF, et al. Hepatic venous
in two gouty patients with moderate hyperuricemia outflow obstruction in autosomal dominant polycys-
and high urine acidity. Klin Wochenschr. 1990;68: tic kidney disease. J Am Soc Nephrol. 1994;5:
874–9. 1186–92.
300. Alexopoulos E, Tampakoudis P, Bili H, Mantalenakis 318. Everson GT, Scherzinger A, Berger-Leff N, et al.
S. Acute uric acid nephropathy in pregnancy. Obstet Polycystic liver disease: quantitation of parenchymal
Gynecol. 1992;80:488–9. and cyst volume from computed tomography and
301. Tomlinson GC, Solberg Jr LA. Acute tumor lysis clinical correlates of hepatic cysts. Hepatology.
syndrome with metastatic medulloblastoma. A case 1988;8:1627.
report. Cancer. 1984;53:1783. 319. Que F, Nagorney DM, Gross JB, Torres VE. Liver
302. Andreoli SP, Clark JH, McGuire WA, Bergstein JM. resection and cyst fenestration in the treatment of
Purine excretion during turnover lysis in children severe polycystic liver disease. Gastroenterology.
with acute lymphocytic leukemia receiving 1995;108:487–94.
Allopurinol: relationship to acute renal failure. 320. Scheff RT, Zuckerman G, Harter H, et al. Diverticular
J Pediatr. 1986;109:292. disease in patients with chronic renal failure due to
303. Albertazzi A, Cappelli P, Paolo B. Nonsteroidal anti- polycystic kidney disease. Ann Intern Med.
inflammatory drugs as risk factor for renal failure 1980;92:202.
from acute uric acid nephropathy. Nephron. 1987; 321. Dominguez Fernandez E, Albrecht KH, Heeman U,
46:98. et al. Prevalence of diverticulosis and incidence of
304. Kelton J, Kelley WN, Holmes EW. A rapid method bowel perforation after kidney transplantation in
for the diagnosis of acute uric acid nephropathy. patients with polycystic kidney disease. Transpl Int.
Arch Intern Med. 1978;138:612–5. 1998;11:28–31.
305. Gabow PA. Definition and history of polycystic kid- 322. Gabow PA, Johnson AM, Kaehny WD, et al. Factors
ney disease. In: Watson ML, Torres VE, editors. affecting the progression of renal disease in auto-
Polycystic kidney disease. Oxford: Oxford University somal dominant polycystic kidney disease. Kidney
Press; 1996. p. 333–55. Int. 1992;41:1311.
306. Parfrey PS, Bear JC, Morgan J, et al. The diagnosis 323. Johnson AM, Gabow PA. Identification of patients
and prognosis of autosomal dominant polycystic with autosomal dominant polycystic disease at high-
kidney disease. N Engl J Med. 1990;323:1085–90. est risk for end-stage renal disease. J Am Soc
307. Newcombe DS. Gouty arthritis and polycystic kid- Nephrol. 1997;8:1560–7.
ney disease. Ann Intern Med. 1973;79:605–6. 324. Reeders ST, Breuning MH, Davies ICE, et al. A
308. Martinez-Maldonado M. Polycystic kidney disease highly polymorphic marker linked to adult polycys-
and hyperuricemia. Ann Intern Med. 1974;80:116. tic kidney disease on chromosome 16. Nature.
309. Torres VE, Wilson DM, Hattery RR, Segura JW. 1985;317:542–4.
Renal stone disease in autosomal dominant polycys- 325. Gretz N, Zeier M, Geberth S, et al. Is gender a deter-
tic kidney disease. Am J Kidney Dis. 1993;22:513. minant for evaluation of renal failure? A study of
310. Torres VE, Erickson SB, Smith LH, et al. The asso- autosomal dominant polycystic kidney disease. Am
ciation of nephrolithiasis and autosomal dominant J Kidney Dis. 1989;14:178.
polycystic kidney disease. Am J Kidney Dis. 1988; 326. Leier CV, Baker PB, Kelman JW, Wooley CF.
11:318. Cardiovascular abnormalities associated with auto-
311. Martinez-Maldonado M, Yium JJ, Eknovan G, Suki somal dominant polycystic kidney disease (ADPKD).
WN. Adult polycystic kidney disease: studies of the Kidney Int. 1987;31:203.
defect in urine concentration. Kidney Int. 1972;2:107. 327. Hadimeri H, Lamm C, Nyberg G. Coronary aneu-
312. Gabow PA, Kaehny WB, Johnson AM, et al. The rysms in patients with autosomal dominant polycys-
clinical utility of renal concentrating capacity in tic kidney disease. J Am Soc Nephrol. 1998;9:
polycystic kidney disease. Kidney Int. 1989;35: 8370841.
675–80. 328. Chapman JR, Hilson AJW. Polycystic kidneys and
313. Elzinga LW, Barry JM, Torres VE, et al. Cyst decom- abdominal aortic aneurysms. Lancet. 1980;1:646.
pression for autosomal dominant polycystic kidney 329. Montolini J, Torras A, Revert L. Polycystic kidneys
disease. J Am Soc Nephrol. 1992;2:1219. and abdominal aortic aneurysms. Lancet.
314. Schwab SJ, Bander SJ, Klahr S. Renal infection in 1982;1:1133.
autosomal dominant polycystic kidney disease. Am 330. Torra R, Nicolau C, Badenas C, et al. Abdominal
J Med. 1987;82:714–8. aortic aneurysms and autosomal dominant polycys-
315. Everson GT. Hepatic cysts in autosomal dominant tic kidney disease. J Am Soc Nephrol. 1996;7:
polycystic kidney disease. Am J Kidney Dis. 2483–6.
1993;22:250. 331. Hossock KF, Leddy CL, Schrier RW, Gabow PA.
316. Sherstha R, McKinley C, Russ P, et al. Post meno- Incidence of cardiac abnormalities associated with
pausal estrogen therapy selectively stimulates autosomal dominant polycystic kidney disease
hepatic enlargement in women with autosomal dom- (ADPKD). Kidney Int. 1987;31:203.
inant polycystic kidney disease. Hepatology. 1997; 332. Chapman AB, Johnson AM, Gabow PA. Intracranial
26:1282–6. aneurysms in patients with autosomal dominant
164 5 Clinical Aspects of Gout and Associated Disease States

polycystic kidney disease: how to diagnose and who 350. Duncan H, Dixon StJ. Gout, familial hyperuricae-
to screen. Am J Kidney Dis. 1993;22:256. mia, and renal disease. Q J Med. 1960;29:127–36.
333. Chapman AB, Rubinstein D, Hughes R, et al. 351. Rosenbloom FM, Kelley WN, Carr AA, Seegmiller
Intracranial aneurysms in autosomal dominant poly- JE. Familial nephropathy and gout in a kindred. Clin
cystic kidney disease. N Engl J Med. 1992;327:916. Res. 1967;15:270.
334. Levey AS, Pauker SG, Kassirer JP. Occult intracra- 352. Treadwell BLJ. Juvenile gout. Ann Rheum Dis.
nial aneurysms in polycystic kidney disease. N Engl 1971;30:279–84.
J Med. 1983;308:986. 353. Bennett RM, Chait A, Lewis B. Familial hyperuri-
335. Torres VE, Wiebers DO, Forbes GS. Cranial com- caemia and hypertriglyceridaemia. Ann Rheum Dis.
puted tomography and magnetic resonance imaging 1973;32:497–500.
in autosomal dominant polycystic kidney disease. 354. Fessel WJ. Renal outcomes of gout and hyperurice-
J Am Soc Nephrol. 1990;1:84. mia. Am J Med. 1979;67:74–82.
336. Blyth H, Ockenden BG. Polycystic disease of kid- 355. Thompson GR, Weiss JJ, Goldman RT, Rigg GA.
neys and liver presenting in childhood. J Med Genet. Familial occurrence of hyperuricemia, gout and
1971;8:257. medullary cystic disease. Arch Intern Med. 1978;138:
337. Cole BR, Conley SB, Stapleton FB. Polycystic kid- 1614–7.
ney disease in the first year of life. J Pediatr. 1987; 356. Van Goor W, Kooiker CJ, Dorhout Mees EJ. An
111:693. unusual form of renal disease associated with gout
338. Maatoo TK, Griefer I, Geva P, Spitzer A. Acquired and hypertension. J Clin Pathol. 1971;24:354–9.
renal cystic disease in children and young adults on 357. Simmonds HA, Warren DJ, Cameron JS, et al.
maintenance dialysis. Pediatr Nephrol. 1997;11: Familial gout and renal failure in young women.
447–50. Clin Nephrol. 1980;14:176–82.
339. Ishikawa I. Acquired cystic disease: mechanisms 358. Cameron JS, Moro F, Simmonds HA. Gout, uric acid
and manifestations. Semin Nephrol. 1991;11:671. and purine metabolism in paediatric nephrology.
340. Gabow PA, Kimberling WJ, Strain JD, et al. Utility Pediatr Nephrol. 1993;7:105–18.
of ultrasonography in the diagnosis of autosomal 359. Simmonds HA, Cameron JS, Potter CF, et al. Renal
dominant polycystic kidney disease in children. failure in young subjects with familial gout. Adv
J Am Soc Nephrol. 1997;8:105–10. Exp Med Biol. 1980;122A:15–20.
341. Ravine D, Gibson R, Walker R, et al. Evaluation of 360. Massari PU, Hsu CH, Barnes RV, et al. Familial
ultrasonographic diagnostic criteria for autosomal hyperuricemia and renal disease. Arch Intern Med.
dominant polycystic kidney disease. Lancet. 1980;140:680–4.
1994;343:824–7. 361. Richmond JM, Kincaid-Smith P, Whitworth JA,
342. Matsubara K, Susuki K, Lin YW, et al. Familial Becker GJ. Familial urate nephropathy. Clin Nephrol.
juvenile nephronophthisis in two siblings: histologi- 1981;16:163–8.
cal findings in an early stage. Acta Paediatr Jpn. 362. Leumann EP, Wegmann W. Familial nephropathy with
1991;33:482. hyperuricemia and gout. Nephron. 1983;34:51–7.
343. Sherman FE, Studnicki FM, Fetterman GH. Renal 363. Hollingworth P, Scott JT. Familial gout, hyperurice-
lesions of familial juvenile nephronophthisis exam- mia and renal impairment. Ann Rheum Dis. 1983;42
ined by microdissection. Am J Clin Pathol. Suppl 1:87–8.
1971;55:391. 364. Yarom A, Rennebohm RM, Strife F, Levinson JE.
344. Pascal RR. Medullary cystic disease of the kidney: Juvenile gouty arthritis. Am J Dis Child. 1984;138:
study of a case with scanning and transmission elec- 955–7.
tron microscopy. Am J Clin Pathol. 1973;59:659. 365. Foreman JW, Yudkoff M. Familial hyperuricemia and
345. Zollinger HU, Mihatsch MJ, Edfonti A, et al. renal insufficiency. Child Nephrol Urol. 1990;10:115–8.
Nephronophthisis (medullary cystic disease of the 366. Puig JG, Miranda ME, Mateos ML, et al. Familial
kidney): a study using electron microscopy, nephropathy and gout: which comes first? Adv Exp
immunofluorescence, and a review of the morpho- Med Biol. 1991;309A:195–8.
logical findings. Helv Paediatr Acta. 1980;35:509. 367. Moro F, Ogg CS, Simmonds HA, et al. Familial
346. Sarre H, Mertz DP. Gout secondary to renal juvenile gouty nephropathy with renal urate hypoex-
insufficiency. Klin Wochenschr. 1965;43:1134–40. cretion preceding renal disease. Clin Nephrol.
347. Richet G, Mignon F, Ardaillon R. Gout secondary to 1991;35:263–9.
chronic renal disease. Presse Med. 1965;73:633–8. 368. McBride MB, Raman V, Ogg CS, et al. A new kin-
348. Calabrese G, Simmonds HA, Cameron JS, Davies dred with familial juvenile gouty nephropathy. Adv
PM. Precocious familial gout with reduced fractional Exp Med Biol. 1991;309A:191–4.
excretion of urate and normal purine enzymes. 369. Saeki A, Hosoya T, Okabe H, et al. Newly discov-
Q J Med. 1990;75:441–50. ered familial juvenile gouty nephropathy in a
349. Yokota N, Yamanaka H, Fujimoto S, et al. Autosomal Japanese family. Nephron. 1995;70:359–66.
dominant transmission of gouty arthritis with renal 370. Cameron JS, Moro F, Simmonds HA. What is the
disease in a large Japanese family. Ann Rheum Dis. pathogenesis of familial juvenile gouty nephropathy.
1991;50:108–11. Adv Exp Med Biol. 1991;309A:185–91.
References 165

371. Puig JG, Miranda ME, Mateos FA, et al. Hereditary dus in females heterogeneous for a vasopressin type
nephropathy associated with hyperuricemia and 2 receptor mutation. Hum Genet. 1995;96:70–8.
gout. Arch Intern Med. 1993;153:357–65. 391. Nicholas HO. Urinary calculi. III. Further observa-
372. Knorr BA, Lipkowitz MS, Potter BJ. Isolation and tions on calculi from patients in the southeast Texas
immunolocalization of a rat renal cortical membrane area. Clin Chem. 1961;7:175–7.
urate transporter. J Biol Chem. 1994;269:6759. 392. Leonard RH. Quantitative composition of kidney
373. Leal-Pinto E, London RD, Knorr BA, Abramson stones. Clin Chem. 1961;7:546–51.
RG. Reconstitution of hepatic uricase in planar lipid 393. Hughes J, Coppridge WM, Roberts LC, Mann VI.
bilayer reveals a functional organic anion channel. Oxalate urinary tract stones. JAMA. 1960;172:774–6.
J Membr Biol. 1995;146:123. 394. Prien EL. Crystallographic analysis of urinary cal-
374. Leal-Pinto E, Tao W, Rappaport J, et al. Molecular culi; a 25 year survey study. J Urol. 1963;89:917–24.
cloning and functional reconstitution of a urate 395. Melick RA, Henneman PH. Clinical and laboratory
transporter channel. J Biol Chem. 1997;272:617. studies of 207 consecutive patients in a kidney stone
375. Isomaki H, Kreus KE. Serum and urinary uric acid in clinic. N Engl J Med. 1958;259:307–14.
respiratory acidosis. Acta Med Scand. 1968;184:293. 396. Herring LC. Observations on the analysis of 10,000
376. Padova J, Bendersky G. Hyperuricemia in diabetic urinary calculi. J Urol. 1962;88:545–62.
ketoacidosis. N Engl J Med. 1962;267:530. 397. Yu TF. Uric acid nephrolithiasis. Handb Exp
377. Manual MA, Steele TH. Changes in renal urate han- Pharmacol. 1978;51:397–422.
dling after prolonged thiazide treatment. Am J Med. 398. Halabe A, Sperling O. Uric acid nephrolithiasis.
1974;57:741. Miner Electrolyte Metab. 1994;20:424–31.
378. Demartini FE. Hyperuricemia induced by drugs. 399. Harrache D, Mesai A, Addou A, et al. Urolithiasis in
Arthritis Rheum. 1965;8:823. children in West Algeria. Ann Urol (Paris). 1997;
379. Stewart RJ, Chonko AM. Pharmacologic inhibition of 31:84.
urate transport across perfused and non-perfused rabbit 400. Borghi L, Meschi T, Amato F, et al. Hot occupation
proximal straight tubules. Kidney Int. 1981;19:258. and nephrolithiasis. J Urol. 1993;150:1757.
380. Cutler RE, Kleeman CR, Maxwell MH, et al. 401. Gutman AB, Yu TF. Urinary ammonium excretion in
Physiologic studies in nephrogenic diabetes insipi- primary gout. J Clin Invest. 1965;44:1474–81.
dus. J Clin Endocrinol Metab. 1962;22:827–38. 402. Henneman PH, Wallach S, Dempsey EF. The meta-
381. Gorden P, Robertson GL, Seegmiller JE. Hyperuri- bolic defect responsible for uric acid stone forma-
cemia, a concomitant of congenital vasopressin- tion. J Clin Invest. 1962;41:537–42.
resistant diabetes insipidus in the adult. Studies of 403. Woeber KA, Ricca L, Hills AG. Pathogenesis of uric
uric acid metabolism and plasma vasopressin. acid urolithiasis. Clin Res. 1962;10:45.
N Engl J Med. 1971;284:1057–60. 404. Gutman AB, Yu TF. An abnormality of glutamine
382. Mizuno O. Transient nephrogenic diabetes insipidus metabolism in primary gout. Am J Med. 1963;35:
associated with acute hepatic failure in pregnancy. 820–31.
Endocrinol Jpn. 1987;34:449–55. 405. Clarke AM, McKenzie RG. Ileostomy and the risk
383. Harper M, Hatjis CG, Appel RG, Austin WE. of urinary uric acid stones. Lancet. 1969;2:395–7.
Vasopressin-resistant diabetes insipidus, liver dys- 406. Gigax JH, Leach JR. Uric acid calculi associated
function, hyperuricemia and decreased renal func- with ileostomy for ulcerative colitis. J Urol. 1971;
tion. A case report. J Reprod Med. 1987;32:862–5. 105:797–9.
384. Yokoyoshi Y, Saito S. Abnormal serum uric acid 407. Yu TF, Gutman AB. Uric acid nephrolithiasis in
level in endocrine disorders. Nippon Rinsho. gout. Predisposing factors. Ann Intern Med.
1996;54:3360–3. 1967;67:1133–48.
385. Decaux G, Prospert F, Namias B, Soupart A. 408. Sperling O, Eilam G, Persky-Brosh S, DeVries A.
Hyperuricemia as a clue for central diabetes insipi- Accelerated erythrocyte 5-phosphoribosyl-1-pyro-
dus (lack of V1 effect) in the differential diagnosis of phosphate synthesis: a familial abnormality associ-
polydipsia. Am J Med. 1997;103:376–82. ated with excessive uric acid production and gout.
386. Robertson GL. Diabetes insipidus. Endocrinol Metab Biochem Med. 1972;6:310.
Clin North Am. 1995;24:549–72. 409. DeVries A, Sperling O. Familial gouty malignant
387. van Lieburg AF, Knoers NV, Monnens LA. Clinical uric acid lithiasis due to mutant phosphoribosylpyro-
presentations and follow-up of 30 patients with con- phosphate synthetase. Urologie A. 1973;12:153.
genital diabetes insipidus. J Am Soc Nephrol. 410. Kelley WN, Wyngaarden JB. Clinical syndromes
1999;10:1958–64. associated with hypoxanthine-guanine phosphoribo-
388. Uribarri J, Kaskas M. Hereditary nephrogenic diabe- syltransferase deficiency. In: Stanbury JB, Wyngaarden
tes insipidus and bilateral nonobstructive hydro- JB, Fredrickson DS, Goldstein JL, Brown MS, edi-
nephrosis. Nephron. 1993;65:346–9. tors. The metabolic basis of inherited diseases. New
389. Carter C, Simpkiss M. The “carrier” state in nephro- York: McGraw-Hill, Inc.; 1983. p. 1115.
genic diabetes insipidus. Lancet. 1956;2:1069–73. 411. Michener WM. Hyperuricemia and mental retarda-
390. van Lieburg AF, Verrdijk MA, Schoute F, et al. tion with athetosis and self-mutilation. Am J Dis
Clinical phenotype of nephrogenic diabetes insipi- Child. 1967;113:155.
166 5 Clinical Aspects of Gout and Associated Disease States

412. Nyhan WL. The Lesch-Nyhan syndrome. Annu Rev acid transport and has sequence similarity to glu-
Med. 1973;24:41. cosidases. Proc Natl Acad Sci USA. 1992;89:
413. Cameron JS, Simmonds HA, Webster DR, et al. 5596–600.
Problems of diagnosis in an adolescent with hypox- 434. Lee WS, Wells RG, Subbog RV, et al. Cloning and
anthine guanine phosphoribosyltransferase deficiency. chromosomal localization of a human kidney cDNA
Adv Exp Med Biol. 1984;165A:7–13. involved in cystine, dibasic, and neutral amino acid
414. Stone TW, Simmonds HA. Purines: basic and clini- transport. J Clin Invest. 1993;91:1959–63.
cal aspects. London: Kluwer; 1991. 435. Pras E, Arber N, Aksentijevich I, et al. Localization
415. Brock WA, Golden J, Kaplan GW. Xanthine calculi in of a gene causing cystinuria to chromosome 2p. Nat
the Lesch-Nyhan syndrome. J Urol. 1983;130:157–9. Genet. 1995;56:1297–303.
416. Kenney IJ. Renal sonography in long standing Lesch- 436. Wartenfeld R, Golomb E, Katz G, et al. Molecular
Nyhan syndrome. Clin Radiol. 1991;43:39–41. analysis of cystinuria in Libyan Jews: exclusion of
417. Grampsas SA, Chandhoke PS, Fan J, et al. Anatomic the SLC3A1 gene and mapping of a new locus on
and metabolic risk factors for nephrolithiasis in 19q. Am J Hum Genet. 1997;60:617–24.
patients with autosomal dominant polycystic kidney 437. Bisceglia L, Calonge MJ, Totaro A, et al. Localization,
disease. Am J Kidney Dis. 2000;36:53. by linkage analysis, of the cystinuria type III gene to
418. Dimitrakov D, Simeonov S. Studies on nephrolithia- chromosome 19q13.1. Am J Hum Genet. 1997;60:
sis in patients with autosomal dominant polycystic 611–6.
kidney disease. Folia Med (Plovdiv). 1994;36:27. 438. Sakhaee K, Poindexter JR, Pak CYC. The spectrum
419. Ng CS, Yost A, Streem SB. Nephrolithiasis with auto- of metabolic abnormalities in patients with cystine
somal dominant polycystic kidney disease: contempo- nephrolithiasis. J Urol. 1989;141:819–21.
rary urological management. J Urol. 2000;163:726. 439. de Vries A, Frank M, Atsmon A. Inherited uric acid
420. Steg A, Aboulker P, Reziciner S, Bernard E. The lithiasis. Am J Med. 1962;33:880–92.
association of polycystic kidney and uric acid lithia- 440. Greene ML, Marcus R, Aurbach GD, et al.
sis. Ann Med Intern (Paris). 1970;121:431. Hypouricemia due to isolated renal tubular defect.
421. Chen YT. Type I glycogen storage disease: kidney Am J Med. 1972;53:361.
involvement, pathogenesis and its treatment. Pediatr 441. Benjamin D, Sperling O, Weinberger A, et al.
Nephrol. 1991;5:71–6. Familial hypouricemia due to isolated renal tubular
422. Chen YT, Van Hove JL. Renal involvement in type I defect. Nephron. 1977;18:220.
glycogen storage disease. Adv Nephrol Necker 442. Tachibana S, Wakatsuki A, Kamei O, et al. A case of
Hosp. 1995;24:357–65. idiopathic hypouricemia with recurrent renal stones.
423. Howell RR. The interrelationship of glycogen stor- Nish J Urol. 1982;44:795.
age disease and gout. Arthritis Rheum. 1965;8:780. 443. Hedley JM, Phillips PJ. Familial hypouricemia and
424. Howell RR, Ashton DM, Wyngaarden JB. Glucose-6- uric acid calculi. Case report. J Clin Pathol.
phosphatase deficiency glycogen storage disease. 1980;33:971.
Studies on the interrelationship of carbohydrate, lipid 444. Frank M, Many M, Sperling O. Familial renal hypou-
and purine abnormalities. Pediatrics. 1982;29:553. ricemia: two additional cases with uric acid lithiasis.
425. Kelley WM, Rosenbloom FM, Seegmiller JE, Howell Br J Urol. 1979;51:88.
RR. Excessive production of uric acid in type I glyco- 445. Gaspar GA, Puig TG, Mateos FA, et al. Hypouricemia
gen storage disease. J Pediatr. 1968;72:488. due to renal urate wasting: different types of tubular
426. Evans WP, Resnick MJ, Boyce WH. Homozygous transport defects. Adv Exp Med Biol. 1986;195A:357.
cystinuria – evaluation of 35 patients. J Urol. 1982; 446. Pak CYC, Kaplan RA, Bone H, et al. A simple test
127:707. for the diagnosis of absorptive, resorptive and renal
427. Resnick MJ, Goodman HO, Boyce WH. hypercalciuria. N Engl J Med. 1975;292:497–500.
Heterozygous cystinuria and calcium oxalate neph- 447. Sperling O, Weinberger A, Oliver I, et al.
rolithiasis. J Urol. 1979;122:52. Hypouricemia, hypercalciuria and decreased bone
428. King Jr JS, Wainer A. Cystinuria with hyperuricemia density: a hereditary syndrome. Ann Intern Med.
and methioninuria. Biochemical study of a case. Am 1974;80:482.
J Med. 1967;43:125–30. 448. Weitz R, Sperling O. Hereditary renal hypouricemia:
429. Krizek V. Uricemia in cystinuria. Horm Metab Res. Isolated tubular defect of urate reabsorption.
1972;4:51–8. J Pediatr. 1980;96:850.
430. Meloni CR, Canary JJ. Cystinuria with hyperurice- 449. Takeda E, Kuroda T, Ito M, et al. Hereditary renal
mia. JAMA. 1967;200:257–9. hypouricemia in children. J Pediatr. 1985;107:71.
431. Vergis JG, Walker AR. Cystinuria, hyperuricemia 450. Ishikawa I, Sakurai Y, Maasuzaki S, et al. Exercise-
and uric acid nephrolithiasis: case report. Nephron. induced acute renal failure in 3 patients with renal
1970;7:577–9. hypouricemia. Jpn J Nephrol. 1990;32:923.
432. Gutman AB, Yu TF. Uric acid nephrolithiasis. Am 451. Erley CMM, Hirschberg RR, Hoefer W, Schaefer K.
J Med. 1968;45:756–79. Acute renal failure due to uric acid nephropathy in a
433. Wells RG, Hediger MA. Cloning of a rat kidney patient with renal hypouricemia. Klin Wochenschr.
cDNA that stimulates dibasic acid neutral amino 1989;67:308.
References 167

452. Dwash IL, Roncari DAK, Marliss E, Fox IH. 471. Roch-Ramel F, Guisan B. Effect of uricosuric drugs
Hypouricemia in disease: a study of different mecha- and pyrazinoate on urate uptake by human brush-bor-
nisms. J Lab Clin Med. 1977;90:153. der membrane vesicles. Eperientia. 1995;51:A93.
453. Ramsdell C, Kelley WN. The clinical significance of 472. Roch-Ramel F, Werner D, Guisan B. Urate transport
hypouricemia. Ann Intern Med. 1973;78:239–42. in brush-border membrane of human kidney. Am
454. Wyngaarden JB. The use and interpretation of labo- J Physiol. 1994;266:F797.
ratory derived data. In: Beeson PB, McDermott W, 473. Burnier M, Rutshmann B, Nussberger J, et al. Salt-
editors. Textbook of medicine. Philadelphia: dependent renal effects of an angiotensin II antago-
Saunders; 1975. p. 1880. nist in healthy subjects. Hypertension. 1993;22:339.
455. Mikkelsen WM, Dodge HJ, Valkenburg H. The dis- 474. Sweet CS, Bradstreet DC, Berman RS, et al.
tribution of serum uric acid values in a population Pharmacodynamic activity of intravenous E-3174,
unselected as to gout or hyperuricemia: Tecumseh, an angiotensin II antagonist, in patients with essen-
Michigan, 1959–1960. Am J Med. 1965;39:242–51. tial hypertension. Am J Hypertens. 1994;7:1035.
456. Tofuku Y, Ito M, Takasaki H, et al. A case of familial 475. Dickinson CJ, Smellie JM. Xanthinuria. Br Med J.
renal hypouricemia associated with acute renal fail- 1959;2:1217.
ure. Purine Pyrimidine Metab (Jpn). 1990;14:8. 476. Srivastava RN, Hussainy MAA, Goel RO, Rose GA.
457. Gafter U, Zuta A, Frydman M, et al. Hypouricemia Bladder stone disease in children in Afghanistan.
due to familial isolated renal tubular uricosuria – Br J Urol. 1986;58:374.
evaluation with combined pyrazinamide-probenecid 477. Kenawi MM. Xanthinuria and xanthine calculous
test. Miner Electrolyte Metab. 1989;15:309. pyonephrosis. J R Coll Surg Edinb. 1976;21:95.
458. Histome I, Ogino K, Saito M, et al. Renal hypourice- 478. De Vries A, Sperling O. Implications of purine
mia due to an isolated renal defect of urate transport. metabolism for the kidney and urinary tract. Ciba
Nephron. 1988;49:81. Found Symp. 1977;48:179.
459. Matsuda O, Shiigai T, Ito Y, et al. A case of familial 479. Carpenter TO, Lebowitz RL, Nelson D, Bauer S.
renal hypouricemia associated with increased secre- Hereditary xanthinuria presenting in infancy with
tion of PAH and idiopathic edema. Nephron. nephrolithiasis. J Pediatr. 1986;109:307.
1982;30:178. 480. Henderson MJ, Bradbury M, Brocklebank JT,
460. Fujiwara J, Takamitsue J, Ueda N, et al. Hypouricemia Simmonds HA. Xanthinuria: presentation with acute
due to an isolated defect in renal tubular urate reab- renal failure in a nine month old girl. J Inherit Metab
sorption. Clin Nephrol. 1980;13:44. Dis. 1992;15:302.
461. Delexelle F, Trombert JC, Bouvier MF, Canarelli G. 481. Maynard J, Benson P. Hereditary xanthinuria in 2
Idiopathic renal hypouricemia I. Observation. Presse Pakistani sisters: asymptomatic in one with b-thala-
Med. 1980;35:2578. ssemia but causing xanthine stone, obstructive urop-
462. Akaoka I, Nishizawa T, Yano E, et al. Renal urate athy and hypertension in the other. J Urol.
excretion in five cases of hypouricemia with an iso- 1988;139:338.
lated renal defect of urate transport. J Rheumatol. 482. Fildes RD. Hereditary xanthinuria with severe uro-
1977;4:86. lithiasis occurring in infancy as renal tubular acido-
463. Shichiri M, Iwamoto H, Maeda M, et al. sis and hypercalciuria. J Pediatr. 1989;115:277.
Hypouricemia due to subtotal defect in the urate 483. Dent CE, Philpot GR. Xanthinuria, an inborn error
transport. Clin Nephrol. 1987;28:300. (or deviation) of metabolism. Lancet. 1954;1:182.
464. Nakajima H, Gomi M, Iida S, et al. Familial renal 484. Salti IS, Mouradian M, Frayha RA. Hereditary xan-
hypouricemia with intact reabsorption of uric acid. thinuria. Arab J Med. 1982;1:5.
Nephron. 1987;45:40. 485. Simmonds HA, Stutchbury JH, Webster DR, et al.
465. Gutman AB, Yu TF. A three-component system for Pregnancy in xanthinuria: demonstration of fetal uric
regulations of renal excretion of uric acid in man. acid production? J Inherit Metab Dis. 1984;7:77.
Trans Assoc Am Physicians. 1961;74:353. 486. Dwosh IL, Roncari DAK, Marliss E, Fox IH.
466. Mudge GH. Uricosuric action of cholecystographic Hypouricemia in disease: a study of different mecha-
agents. N Engl J Med. 1971;284:929–33. nisms. J Lab Clin Med. 1977;90:153.
467. Postlethwaite AE, Kelley WN. Uricosuric effect of 487. Frezal J, Malassenet R, Cartier P, et al. Sur un cas de
radiocontrast agents. Ann Intern Med. 1971;74:845–52. xanthinurie. Arch Fr Pediatr. 1967;24:129.
468. Ramsdell CM, Postlethwaite AE, Kelley WN. 488. Cifuentes-Delatte L, Castro-Mendoza HJ.
Uricosuric effect of glyceryl guaiacolate. J Rheum. Xanthinuria familiar. Rev Clin Esp. 1967;107:244.
1974;1:114–6. 489. Kennedy JH, Semmence AM. Xanthinuria: a new
469. Gross JM. Fanconi syndrome (adult type) develop- case. Clin Chem. 1980;26:1758.
ing secondary to the ingestion of outdated tetracy- 490. Rodriguez GF, Servan PR, Rodriguez JP. Un nuevo
cline. Ann Intern Med. 1963;48:523. caso de xanthinuria. Asociacion con SIDA. Ann
470. Nakashima M, Uematsu T, Kosuge K, Kanamaru M. Intern Med (Madrid). 1989;6:219.
Pilot study of the uricosuric effect of uP-753, a new 491. Delbarre F, Weissenbach R, Auscher C, De Gery A.
angiotensin II receptor antagonist, in healthy sub- Acess de gout chez xanthinurique. Nouv Presse
jects. Eur J Clin Pharmacol. 1992;42:333. Med. 1973;2:2465.
168 5 Clinical Aspects of Gout and Associated Disease States

492. Wilson DM, Topia HR. Xanthinuria in a large kin- 509. Levy HL. Hartnup disorder. In: Scriver CR, Beaudet
dred. Adv Exp Med Biol. 1974;41A:343. AL, Sly WS, Valle D, editors. The metabolic and
493. Isaacs H, Heffron JJA, Berman L, et al. Xanthine, molecular bases of inherited disease. New York:
hypoxanthine and muscle pain. Histochemical and McGraw-Hill, Inc.; 1995. p. 3629.
biochemical observations. S Afr Med J. 1975;49: 510. Bennett JS, Bond J, Singer I, Gottlieb AJ. Hypouricemia
1035. in Hodgkin’s disease. Ann Intern Med. 1972;76:751.
494. De Vooght HJ, Von de Kamp JJP, Van Gerderen HH, 511. Tykarski A. Mechanism of hypouricemia in
et al. Een xanthinsteen by een kind met xanthinuie: Hodgkin’s disease. Isolated defect in postsecretory
Enkele beschonasingen over een Zeldaure “inborn reabsorption of uric acid. Nephron. 1988;50:217.
error of metabolism”. Ned Tijdschr Geneeskd. 512. Weinstein B, Irreverre F, Watkins DM. Lung carci-
1973;117:976. noma, hypouricemia and aminoaciduria. Am J Med.
495. Engleman K, Watts RWE, Klinenberg JR, et al. 1965;39:520.
Clinical, physiological and biochemical studies of a 513. Gorshein D, Asbell S. Ectopic production of hor-
patient with xanthinuria and pheochromocytoma. mones in tumors. JAMA. 1976;235:2716.
Am J Med. 1964;37:839–61. 514. Cooper DS. Oat-cell carcinoma and severe hypouri-
496. Parker R, Snedden W, Watts RWE. The quantitative cemia. N Engl J Med. 1973;288:321.
determination of hypoxanthine and xanthine (“oxy- 515. Smithline N, Kassirer JP, Cohen JJ. Light-chain
purines”) in skeletal muscle from two patients with nephropathy. N Engl J Med. 1976;294:71.
congenital xanthine oxidase deficiency (xanthinu- 516. Beck IH. Hypouricemia in the syndrome of inappro-
ria). Biochem J. 1970;116:317. priate secretion of antidiuretic hormone. N Engl
497. Chalmers RA, Johnson M, Pallis C, Watts RWE. J Med. 1979;301:528.
Xanthinuria with myopathy (with some observations 517. Weinberger A, Santo M, Shalit M, et al. Abnormality
on the renal handling of oxypurines in the disease). in renal urate handling in the syndrome of inappro-
Q J Med. 1969;38:493. priate secretion of antidiuretic hormone. Isr J Med
498. Simmonds HA, Cameron JS, Barrat TM, et al. Purine Sci. 1982;18:711.
enzyme defects as a cause of acute renal failure in 518. Osterlind K, Hansen M, Dombernowsky P.
childhood. Pediatr Nephrol. 1989;3:433. Hypouricemia and inappropriate secretion of antidi-
499. Wada Y, Nishimura Y, Tanabu M, et al. Hypouricemic, uretic hormone in small cell bronchogenic carci-
mentally retarded infant with a defect of 5-phospho- noma. Acta Med Scand. 1981;209:289.
ribosyl-1-pyrophosphate synthetase of erythrocytes. 519. Kay NE, Gottlieb AJ. Hypouricemia in Hodgkin’s
Tohoku J Exp Med. 1974;113:149–57. disease. Cancer. 1973;32:1508–11.
500. Inuma K, Wada Y, Onuma A, Tanabu M. 520. Chisholm Jr JJ, Harrison HC, Everlein WR, Harrison
Electroencephalographic study of an infant with HE. Amino-aciduria, hypophosphatemia and rickets
phosphoribosylpyrophosphate synthetase deficiency. in lead poisoning. Am J Dis Child. 1955;89:159.
Tohoku J Exp Med. 1975;116:53–5. 521. Clarkson TW, Keneh JE. Urinary excretion of amino
501. Imaeda H, Wada Y. PRPP synthetase deficiency. acids by men absorbing heavy metals. Biochem J.
Ryoikibetsu Shokogun Shirizu. 1998;18:435–7. 1965;62:361.
502. Wilson DB, Goldstein NP. Renal urate excretion in 522. Michelis MF, Warms PC, Fusco RD, Davis BB.
patients with Wilson’s disease. Kidney Int. 1973;4: Hypouricemia and hyperuricosuria in Laennec cir-
331. rhosis. Arch Intern Med. 1974;134:681.
503. Leu MI, Strickland GT, Gutman AB. Renal function 523. Schlosstein L, Kippen I, Bluestone R, et al.
in Wilson’s disease: response to penicillamine ther- Association between hypouricemia and jaundice.
apy. Am J Med Sci. 1970;250:381. Ann Rheum Dis. 1974;33:308.
504. Morgan HG, Steewart WK, Lowe KG, et al. Wilson’s 524. Pui CH, Roy 3rd S, Noe HN. Urolithiasis in child-
disease and the Fanconi syndrome. Q J Med. 1962; hood acute leukemia and non-Hodgkin’s lymphoma.
31:361. J Urol. 1986;136:1052.
505. Elsas LJ, Hayslett JP, Spargo BH, et al. Wilson’s dis- 525. Pochedly C. Hyperuricemia in leukemia and lym-
ease with reversible renal tubular function. Ann phoma. I. Clinical findings and pathophysiology.
Intern Med. 1971;75:127. N Y State J Med. 1973;73:1085.
506. Cusworth DC, Dent CE, Flynn FV. The amino-aci- 526. Moe PJ. Formation of urinary calculi and uric acid
duria in galactosaemia. Arch Dis Child. 1955; findings in children with leukemia. Tidsskr Nor
30:150. Laegeforen. 1970;90:1201.
507. Lamiere N, Mussche M, Baele G, et al. Hereditary 527. Teluk J, Reiszkowski J. Case of chronic myeloid leu-
fructose intolerance: a difficult diagnosis in the adult. kemia complicated by nephrolithiasis. Wiad Lek.
Am J Med. 1978;65:416. 1976;29:1577.
508. Schneider JA, Schulman JD, Seegmiller JE. 528. Hsu AC, Kooh SW, Izukawa T, Fox IH. Uric acid
Cystinosis and the Fanconi syndrome. In: Stanbury lithiasis in an infant with cyanotic congenital heart
JB, Wyngaarden JB, Fredrickson DS, editors. The disease. J Pediatr. 1977;91:1021.
metabolic basis of inherited disease. 4th ed. New 529. Yu TF. Review article. Urolithiasis in hyperuricemia
York: McGraw-Hill, Inc; 1978. p. 1660. and gout. J Urol. 1981;126:424.
References 169

530. Lynch EC. Uric acid metabolism in proliferative 549. Klinenberg JR, Goldfinger S, Miller J, et al. The
diseases of the marrow. Arch Intern Med. 1962; effectiveness of a xanthine oxidase inhibitor in the
109:639. treatment of gout. Arthritis Rheum. 1963;6:779–80.
531. Lancina Martin JA, Garcia Buitron JM, Diaz 550. Klinenberg JR, Goldfinger SF, Seegmiller JE.
Bermudez J, et al. Urinary lithiasis in transplanted The effectiveness of xanthine oxidase inhibitor
kidney. Arch Esp Urol. 1997;50:141. allopurinol in the treatment of gout. Ann Intern Med.
532. Norlen BJ, Hellstrom M, Nisa M, Robertson WG. 1965;62:639–47.
Uric acid stone formation in a patient after kidney 551. Sorensen LB. Seminars on the Lesch-Nyhan syn-
transplantation – metabolic and therapeutic consid- drome: management and treatment. Discussion. Fed
erations. Scand J Urol Nephrol. 1995;29:335. Proc. 1968;27:1097.
533. Cantarell MC, Capdevila L, Morlens M, Piera L. 552. Greene ML, Fujimoto WY, Seegmiller JE. Urinary
Uric acid calculus in renal transplant patients treated xanthine stones – a rare complication of allopurinol
with cyclosporine. Clin Nephrol. 1991;35:288. therapy. N Engl J Med. 1969;280:426.
534. Harper JM, Samuell CT, Hallson PC, et al. Risk fac- 553. Mizuno T, Segawa M, Kurumada T. Clinical and
tors for calculus formation in patients with renal therapeutic aspects of the Lesch-Nyhan syndrome in
transplants. Br J Urol. 1994;74:147. Japanese children. Neuropaediatrie. 1970;2:38.
535. Glicklich D, Gruber HE, Matas AJ, et al. 554. Manzke H. Xanthine stone formation subsequent to
2,8-Dihydroxyadenine urolithiasis: report of a case allopurinol therapy. Dtsch Med Wochenschr.
first diagnosed after renal transplant. Q J Med. 1974;99:918.
1988;68:785. 555. Wyngaarden JB. Allopurinol and xanthine nephrop-
536. Adam O, Goebel FD. Secondary gout and pseudo- athy. N Engl J Med. 1970;283:371–2.
Bartter syndrome in females with laxative abuse. 556. Sperling O, Brosh S, Boer P, et al. Urinary xanthine
Klin Wochenschr. 1987;65:833. stones in an allopurinol treated gouty patient with
537. Dick WH, Lingeman JE, Preminger JE, et al. partial deficiency of hypoxanthine-guanine phospho-
Laxative abuse as a cause for ammonium urate cal- ribosyltransferase. Israel J Med Sci. 1978;14:288.
culi. J Urol. 1990;143:244. 557. Rundles RW, Wyngaarden JB, Hitchings G, et al.
538. Asanuma H, Nagatsuma K, Baba S, Murai M. A case Effects of xanthine oxidase inhibitor on thiopurine
of Prader-Willi syndrome accompanied with a renal metabolism, hyperuricemia and gout. Trans Assoc
stone. Hinyokika Kiyo. 1998;44:37. Am Physicians. 1963;76:126–40.
539. Cassidy SB. Prader-Willi syndrome. Curr Probl 558. Yu TF, Gutman AB. Effect of allopurinol (4-hydroxy-
Pediatr. 1984;14:1. pyrazolo (3,4-d)-pyrimidine) on serum and urinary
540. Holm VJ, Cassidy SB, Butler MG, et al. Prader-Willi uric acid in primary and secondary gout. Am J Med.
syndrome: consensus diagnostic criteria. Pediatrics. 1964;37:885–98.
1993;912:398. 559. Segal S, Wyngaarden JB. Plasma glutamine and
541. Butler MG. Prader-Willi syndrome: current under- oxypurine content in patients with gout. Proc Soc
standing of cause and diagnosis. Am J Med Genet. Exp Biol Med. 1955;88:342–5.
1990;35:319. 560. Jorgensen S. Hypoxanthine and xanthine accumu-
542. Leistenschneider W, Nagel R. Experiences with lated in stored human blood: determination of rela-
lavage cytology from renal pelvis and ureter. Urologe tive amounts by spectrophotometry. Acta Pharmacol
A. 1977;16:230. Toxicol. 1955;11:265–76.
543. Dean TE, Harrison NW, Bishop NL. CT scanning in 561. Elion GB. Allopurinol and other inhibitors of urate
the diagnosis and management of radiolucent uri- synthesis. Handb Exp Pharmacol. 1978;51:485–514.
nary calculi. Br J Urol. 1988;62:405. 562. Farebrother DA, Hatfield P, Simmonds HA, et al.
544. Nakada SY, Hoff DG, Attai S, et al. Determination Experimental crystal nephropathy (one year study in
of stone composition by noncontrast spiral computed the pig). Clin Nephrol. 1975;4:243–50.
tomography in the clinical setting. Urology. 563. Reiter S, Simmonds HA, Zollner N, et al. Demonstration
2000;55:816. of a combined deficiency of xanthine oxidase and
545. Band PR, Silverberg DS, Henderson JF. Xanthine aldehyde oxidase in xanthinuric patients not forming
nephropathy in a patient with lymphosarcoma treated oxipurinol. Clin Chim Acta. 1990;187:221.
with allopurinol. N Engl J Med. 1970;283:354–7. 564. Van Gennip AH, van Noordenburg Huistra D, de
546. Ogawa A, Watanabe K, Minejima N. Renal xanthine Bree PK, Wadman SK. Two-dimensional thin layer
stones in Lesch-Nyhan syndrome treated with chromatography for the screening of disorders of
allopurinol. Urology. 1985;26:56. purine and pyrimidine metabolism. Clin Chim Acta.
547. Stevens SK, Parker BR. Renal oxypurine deposition 1978;96:7.
in Lesch-Nyhan syndrome: sonographic evaluation. 565. Simmonds HA, Duley JA, Davies PM. Analyses of
Pediatr Radiol. 1989;19:479. purines and pyrimidines in blood, urine, and other
548. Ablin A, Stephens BG, Hirata T, et al. Nephropathy, physiological fluids. In: Hommes F, editor.
xanthinuria, and orotic aciduria complicating Techniques in diagnostic human biochemical genet-
Burkitt’s lymphoma treated with chemotherapy and ics. A Laboratory Manual. New York: Wiley-Liss;
allopurinol. Metabolism. 1972;21:771–8. 1991. p. 397.
170 5 Clinical Aspects of Gout and Associated Disease States

566. Bennett MJ, Carpenter KH, Hill PG. Asymptomatic xanthine (“oxypurines”) in skeletal muscle from two
xanthinuria detected as a result of routine analysis of patients with congenital xanthine oxidase deficiency
serum for urate. Clin Chem. 1985;31:492. (xanthinuria). Biochem J. 1969;115:103–8.
567. Boulieu R, Bory C, Baltassat P, Divry P. Hypoxanthine 583. Crawhall JC, Itiaba K, Katz S. Separation and quan-
and xanthine concentrations determined by high per- titation of oxypurines by isocratic high pressure liq-
formance liquid chromatography in biological fluids uid chromatography: application to xanthinuria and
from patients with xanthinuria. Clin Chim Acta. the Lesch-Nyhan syndrome. Biochem Med.
1984;142:83. 1983;30:261.
568. Harkness RA, Coade SB, Walton KR, Wright D. 584. Kawachi M, Kono N, Mineo I, et al. Decreased xan-
Xanthine oxidase deficiency and “Dalmation” thine oxidase activities and increased urinary oxypu-
hypouricemia: Incidence and effect of exercise. rines in heterozygotes for hereditary xanthinuria.
J Inherit Metab Dis. 1983;6:114. Clin Chim Acta. 1990;88:137.
569. Costello J, Al-Dabagh E. A new rapid spectrophoto- 585. Castro-Liendoza HJ, Cifuentes-Delatte LC, Rapudo
metric method for the detection of xanthinuria. In: A. Una nueva observacion de xanthinuria familiar.
Schwille PO, Smith LH, Robertson WG, Vahlensieck Rev Clin Esp. 1972;124:341.
W, editors. Urolithiasis and related clinical research. 586. Kutter D, Humbel R, Bisdorff J. Biochemische
New York: Plenum; 1985. p. 681. untersuchungen bei einem typischen Fall von xan-
570. Holmes EW, Wyngaarden JB. Hereditary xanthinu- thinurie. Dtsch Med Wochenschr. 1970;95:1269.
ria. In: Scriver CR, Beaudet AL, Sly WS, Valle D, 587. Simmonds HA, Reiter S, Nishino T. Hereditary xan-
editors. The metabolic basis of inherited disease. thinuria. In: Scriver CR, Beaudet AL, Sly WS, Valle
New York: McGraw-Hill, Inc; 1989. p. 1090. D, editors. The metabolic and molecular bases of
571. Simmonds HA, Levin B, Cameron JS. Variations in inherited disease. New York: McGraw-Hill, Inc.;
allopurinol metabolism by xanthinuric subjects. Clin 1995. p. 1789.
Sci Mol Med. 1974;47:173. 588. Ullmann H. Zur frage der harnsaureausscheidung in
572. Yamamoto T, Higashino K, Kono N, et al. Metabolism urin bei ikteruskranken. Klin Wochenschr. 1923;2:
of pyrazinamide and allopurinol in hereditary xan- 2174–5.
thine oxidase deficiency. Clin Chim Acta. 1989; 589. Matz R, Christodoulou J, Vianna N, et al. Renal
180:169. tubular dysfunction associated with alcoholism and
573. Auscher C, Pasquier C, Pehuet P, Delbarre F. Study liver disease. N Y State J Med. 1969;69:1312–4.
of urinary pyrazinamide metabolites and their action 590. Passero G, Masini G. L’ipouricemia negli itteri colu-
on renal excretion of xanthine and hypoxanthine in a rici. Minerva Med. 1958;49:3155–8.
xanthinuric patient. Biomedicine. 1978;28:129. 591. Van Peenen HJ. Causes of hyperuricemia. Ann Intern
574. Holmes EW, Mason DH, Goldstein LI, et al. Med. 1973;78:977–8.
Xanthine oxidase deficiency: studies of a previously 592. Casas E, Serrano C, Daimiel E, et al. Prevalence,
unreported case. Clin Chem. 1974;20:1076. physiopathology and processes associated with
575. Auscher C, Pasquier C, Amory N, et al. The effect of hypouricemia in a hospitalized population: analysis
weight reduction on plasma and urinary levels of of 27,987 analytic determinations. Rev Clin Esp.
oxypurines in an obese xanthinuric patient. Adv Exp 1990;186:211–5.
Med Biol. 1980;122A:241. 593. Diaz Curiel M, Zea Mendoza A, Rapado A, Gonzalez
576. Kojima T, Nishina T, Kitamura M, et al. Biochemical Villasante J. Significacion clinica de la hipouicemia
studies on the purine metabolism of four cases with en 14,685 determinaciones del autoanalizador. Rev
hereditary xanthinuria. Clin Chim Acta. 1984; Clin Esp. 1975;139:365.
137:189. 594. Sperling O, Weinberger A, Pinkhas J, de Vries A.
577. Hillebrand G, Reiter S. Hypourikamie – ein differen- Frequency and causes of hypouricemia in hospital
tialdiagnostisches problem. Internist. 1991;32:226. patients. Isr J Med Sci. 1977;13:529.
578. Reiter S, Simmonds HA, Zollner N, et al. 595. Hisatome I, Ogino K, Kotake H, et al. Cause of per-
Demonstration of a combined deficiency of xanthine sistent hypouricemia in outpatients. Nephron.
oxidase and aldehyde oxidase forming oxipurinol. 1989;51:13.
Clin Chim Acta. 1990;187:221. 596. Yanasze M, Nakahama H, Mikami H, et al.
579. Laudaas S, Borch K, Aagaard E. A new case with Prevalence of hypouricemia in apparently normal
hereditary xanthinuria: response to exercise. Clin population. Nephron. 1988;48:80.
Chim Acta. 1989;181:119. 597. Simmonds HA, Van Acker KJ. Adenine phosphori-
580. Engelman K, Watts RWE, Klinenberg JR, et al. bosyltransferase deficiency: 2,8-dihydroxyadenine
Clinical, physiological and biochemical studies of a lithiasis. In: Stanbury JB, Wyngaarden JB,
patient with xanthinuria and pheochromocytoma. Fredrickson DS, Goldstein JL, Brown MS, editors.
Am J Med. 1964;37:839. The metabolic basis of inherited disease. 5th ed.
581. Chalmers RA, Watts RWE, Pallis C, et al. Crystalline New York: McGraw-Hill, Inc; 1983. p. 1144–56.
deposits in striped muscle in xanthinuria. Nature. 598. Coupris L, Champion G, Duverne C, et al. 2,8-dihy-
1969;221:170–1. droxyadenine lithiasis. Two new pediatric cases of this
582. Parker R, Snedden W, Watts RWE. The mass spec- misdiagnosed metabolic abnormality. The value of
trophotometric identification of hypoxanthine and extracorporeal lithotripsy. Chir Pediatr. 1989;30:253.
References 171

599. Yagisawa T, Yamazaki Y, Toma H, Kamatani N. and occurrence in a female with gout and renal dis-
Radiopaque 2,8-dihydroxyadenine lithiasis. Int Urol ease. Aust N Z J Med. 1975;5:440.
Nephrol. 1999;31:141. 617. Kelley WN, Levy RI, Rosenbloom FM, et al.
600. Simmonds HA, Sahota AS, Van Acker KJ. Adenine Adenine phosphoribosyltransferase deficiency – a
phosphoribosyltransferase deficiency and 2,8-dihy- previously undescribed genetic defect in man. J Clin
droxyadenine lithiasis. In: Scriver CR, Beaudet AL, Invest. 1968;47:2281.
Sly WS, Valle D, editors. The metabolic and molec- 618. Fox IH, Meade JC, Kelley WN. Adenine phosphori-
ular bases of inherited disease. New York: McGraw- bosyltransferase deficiency in man. Am J Med.
Hill, Inc; 1995. p. 1704. 1973;55:614.
601. Ceballos-Picot I, Perignon JL, Hamet M, et al. 619. Delbarre F, Auscher C, Amor B, et al. Gout with
2,8-dihydroxyadenine urolithiasis, an underdiag- adenine phosphoribosyltransferase deficiency.
nosed disease. Lancet. 1992;339:1050. Biomedicine. 1974;21:82.
602. Takemoto M, Nagano S. Urolithiasis containing 620. Kojima T, Nishina T, Kamatani N, Nishioka K.
2,8-dihydroxyadenine: report of a case. Acta Urol Reversed-phase high-performance liquid chroma-
(Jpn). 1979;25:265. tography of 2,8-dihydroxyadenine in serum and
603. Gliklich D, Gruber HE, Matas AJ, et al. 2,8-dihy- urine with electrochemical detection. Clin Chim
droxyadenine lithiasis. Report of a case first diag- Acta. 1989;181:109.
nosed after renal transplant. Q J Med. 1988;69:785. 621. Doppler W, Hirsch-Kauffmann M, Schabel F,
604. Greenwood MC, Dillon MJ, Simmonds HA, et al. Schweiger M. Characterization of the biochemical
Renal failure due to 2,8-dihydroxyadenine urolithia- basis of a complete deficiency of the adenine phospho-
sis. Eur J Pediatr. 1982;138:346. ribosyltransferase (APRT). Hum Genet. 1984;57:404.
605. Schabel F, Doppler W, Hirsch-Kauffmann M, et al. 622. Feigleson P, Davidson JD, Robins RK. Pyrazolopy-
Hereditary deficiency of adenine phosphoribosyl- rimidines as inhibitors and substrates of xanthine
transferase. Paediatr Paedol. 1980;15:233. oxidase. J Biol Chem. 1957;226:993–1000.
606. Joost J, Doppler W. The 2,8-dihydroxyadenine stone 623. Krenitsky TA, Elion GB, Strelitz RA, Hitchings GH.
in childhood. Urology. 1982;20:67. Ribonucleosides of allopurinol and oxoallopurinol.
607. Laxdal T, Jonasson TA. Adenine phosphoribosyl- J Biol Chem. 1967;242:2675–82.
transferase deficiency in Iceland. Acta Med Scand. 624. Langrebe AR, Nyhan WL, Coleman M. Urinary
1988;224:621. tract stones resulting from the excretion of oxipurinol.
608. Gleeson MJ, Griffith DP. Distribution of patients N Engl J Med. 1975;292:626–7.
with 2,8-dihydroxyadenine urolithiasis and adenine 625. Stote RM, Smith LH, Dubb JW, et al. Oxipurinol
phosphoribosyltransferase deficiency in Japan. J Urol. nephrolithiasis in regional enteritis secondary to
1989;142:834. allopurinol therapy. Ann Intern Med. 1980;92:
609. Cartier P, Hamet M. Une nouvelle maladie metabo- 384–5.
lique: le deficit complet en adenine-phosphoribosyl- 626. Prien EL, Prien Jr EL. Composition and structure of
transferase avec lithiasise de 2,8-dihydroxyadenine. urinary stone. Am J Med. 1968;45:654–72.
C R Acad Sci (Paris). 1974;279:883. 627. Talbott JH. Gout. New York: Grune & Stratton;
610. Simmonds HA, Van Acker KJ, Cameron JS, 1957. p. 205.
Snedden W. The identification of 2,8-dihydroxyade- 628. Coe FL. Treated and untreated recurrent calcium
nine, a new component of urinary stones. Biochem J. nephrolithiasis in patients with idiopathic hypercal-
1986;157:485. ciuria, hyperuricosuria, or no metabolic disorder.
611. Simmonds HA, Potter CF, Sahota A, et al. Adenine Ann Intern Med. 1977;87:404–10.
phosphoribosyltransferase deficiency presenting 629. Coe FL, Moran E, Kavalach AG. The contribution of
with supposed uric acid stones; pitfalls of diagnosis. dietary protein over-consumption to hyperuricosuria
J R Soc Med. 1978;71:791. in calcium oxalate stone formers. J Chronic Dis.
612. Simmonds HA. 2,8-dihydroxyadeninuria, or when is 1976;29:793–800.
a uric acid stone not a uric acid stone? Clin Nephrol. 630. Coe FL. Association of calcium nephrolithiasis with
1979;12:195. disorders of uric acid metabolism. Handb Exp
613. Reveillaud RJ, Daudon M, Protat MF, et al. Lithiase Pathol. 1978;51:423.
2,8-dihydroxyadenique: un nouveau cas depiste par 631. Coe FL, Raisen L. Allopurinol treatment of uric-acid
analyse infra-rouge. Nouv Presse Med. 1979;8:2965. disorders in calcium-stone formers. Lancet.
614. Johnson LA, Gordon RB, Emmerson BT. Adenine 1973;1:129–31.
phosphoribosyltransferase: a simple spectrophoto- 632. Coe FL, Kavalach AG. Hypercalciuria and hyperuri-
metric assay and the incidence of mutation in the cosuria in patients with calcium nephrolithiasis.
normal population. Biochem Genet. 1977;15:256. N Engl J Med. 1974;291:1344–50.
615. Fox IH, LaCroix S, Planet G, Moore M. Partial 633. Smith MJV. Placebo versus allopurinol for renal cal-
deficiency of adenine phosphoribosyltransferase in culi. J Urol. 1977;117:690–2.
man. Medicine. 1977;56:515. 634. Ettinger B, Tang A, Citron JT, et al. Randomized
616. Emmerson BT, Gordon RB, Thompson L. Adenine trial of allopurinol in the prevention of calcium
phosphoribosyltransferase deficiency: its inheritance oxalate calculi. N Engl J Med. 1986;315:1386–9.
172 5 Clinical Aspects of Gout and Associated Disease States

635. Pak CYC, Peterson R. Successful treatment of hype- patients treated with chlorothiazide. N Engl J Med.
ruricosuric calcium oxalate nephrolithiasis with potas- 1970;282:133.
sium citrate. Arch Intern Med. 1986;146:863–7. 655. Brest AN, Heider C, Mehbod H, et al. Drug control
636. Pak CYC, Sakhaee K, Fuller C. Successful manage- of diuretic-induced hyperuricemia. JAMA. 1966;
ment of uric acid nephrolithiasis with potassium cit- 195:132.
rate. Kidney Int. 1986;30:422–8. 656. Emmerson BT. A comparison of uricosuric agents in
637. Noda S, Hayashi K, Eto K. Oxalate crystallization in gout with special reference to sulfinpyrazone. Med
the kidney in the presence of hyperuricemia. J Aust. 1963;1:839.
Scanning Microsc. 1989;3:829. 657. Yu TF, Gutman AB. Paradoxical retention of uric
638. Gover PK, Ryall RL, Marshall VR. Dissolved urate acid by uricosuric drugs in low dosage. Proc Soc
promotes calcium oxalate crystallization: epitaxy is Exp Biol Med. 1955;90:542.
not the cause. Clin Sci (Colch). 1993;85:303. 658. Gutman AB, Yu TF. Effect of salicylate in varying
639. Suki WN, Hull AR, Rector Jr FC, et al. Mechanism dosage on urinary urate excretion in gouty subjects.
of the effect of thiazide diuretics on calcium and uric Ann Rheum Dis. 1955;14:444.
acid. J Clin Invest. 1967;46:1121. 659. Grayzel AI, Liddle L, Seegmiller JE. Diagnostic
640. Wyngaarden JB. Diuretics and hyperuricemia. significance of hyperuricemia in arthritis. N Engl
N Engl J Med. 1970;283:1170. J Med. 1961;265:763.
641. Cannon PJ, Heinemann H, Stason WB, et al. 660. See G. Etudes sur l’acid salicylique et lest salicy-
Ethacrynic acid. Effectiveness and mode of diuretic lates: traitment du rheumatisme aigu et chronique,
action in man. Circulation. 1965;31:5. de la goutte, et de diverses affections du systeme
642. Steele TH. Evidence for altered renal urate reabsorp- nerveux sensitif par les salicylates. Bull Acad Med
tion during changes in volume of the extracellular Paris. 1877;6:689, 717, 926, 937, 1024.
fluid. J Lab Clin Med. 1969;74:288. 661. Yu TF, Dayton PG, Gutman AB. Mutual suppression
643. Schirmeister J, Man NK, Hallauer W. Lactate and of the uricosuric effects of sulfinpyrazone and sali-
uric acid following oral diuretic drug administration cylate: a study in interactions between drugs. J Clin
in man. Klin Wochenschr. 1967;45:1219. Invest. 1963;42:1330.
644. Schirmeister J, Man NK, Warning D, Hallauer W. 662. Lieber CS, Davidson CS. Some metabolic effects of
On the question of an extrarenal cause for uric acid ethyl alcohol. Am J Med. 1962;33:319.
retention after furosemide. Verh Dtsch Ges Inn Med. 663. Lieber CS, Jones DP, Losowsky MS, et al.
1967;73:1025. Interrelation of uric acid and ethanol metabolism in
645. Park YB, Park YS, Song J, et al. Clinical manifesta- man. J Clin Invest. 1962;41:1863.
tions of Korean female gouty patients. Clin 664. MacLachlan MJ, Rodnan JP. Effects of food, fast
Rheumatol. 2000;19:142. and alcohol on serum uric acid and acute attacks of
646. Hayem G, Delahousse M, Meyer O, et al. Female gout. Am J Med. 1967;42:38.
premenopausal tophaceous gout induced by long- 665. Newcombe DS. Ethanol metabolism and uric acid.
term diuretic abuse. J Rheumatol. 1996;23:2166. Metabolism. 1972;21:1193.
647. Puig JG, Mechan AD, Jiminez ML, et al. Female 666. Olin JA, Devenyi P, Weldon KL. Uric acid in alco-
gout. Clinical spectrum and uric acid metabolism. holics. Q J Stud Alcohol. 1973;34:1202.
Arch Intern Med. 1991;151:726. 667. Cullen JH, Early LJA, Fiore JM. The occurrence of
648. Meyers OL, Monteagudo FS. A comparison of gout hyperuricemia during pyrazinamide-isoniazide ther-
in men and women. A 10-year experience. S Afr apy. Am Rev Tuberc. 1956;74:289.
Med J. 1986;70:721. 668. Inoue T, Iheda N, Kurasawa T, et al. Hyperuricemia
649. Lally EV, Ho Jr G, Kaplan SR. The clinical spectrum and arthralgias during pyrazinamide treatment.
of gouty arthritis in women. Arch Intern Med. Nihon Kokyuki Gakkai Zasshi. 1999;37:115.
1986;146:2221. 669. Koumbaniou C, Nicopoulaaaos G, Vassilion M,
650. Meyers OL, Monteagudo FS. Gout in females: an anal- et al. Is pyrazinamide really the third drug of choice
ysis of 92 patients. Clin Exp Rheumatol. 1985;3:105. in the treatment of tuberculosis. Int J Tuberc Lung
651. Scott JT, Higgins CS. Diuretic induced gout: a mul- Dis. 1998;2:675.
tifactorial condition. Ann Rheum Dis. 1992;51:259. 670. Steele TH, Rieselbach RE. The renal mechanism for
652. Lin KC, Lin HY, Chou P. The interaction between urate homeostasis in normal man. Am J Med.
uric acid level and other risk factors on the develop- 1967;43:868.
ment of gout among asymptomatic hyperuricemic 671. Gutman AB, Yu TF, Berger L. Renal function in
men in a prospective study. J Rheumatol. gout. III. Estimation of tubular secretion and reab-
2000;27:1501. sorption of uric acid by use of pyrazinamide (pyrazi-
653. Lin KC, Lin HY, Chou P. Community based epide- noic acid). Am J Med. 1969;47:575.
miological study on hyperuricemia and gout in 672. Weiner IM, Tinker JP. Pharmacology of pyrazin-
Kin-Hu, Kinmen. J Rheumatol. 2000;27:1045. amide: metabolic and renal function studies related
654. Nicotero JA, Scheib ET, Martinez R, et al. Prevention to the mechanism of drug-induced urate retention.
of hyperuricemia by allopurinol in hypertensive J Pharmacol Exp Ther. 1972;180:411.
References 173

673. Shapiro M, Hyde L. Hyperuricemia due to pyrazin- 692. Peeters P, Sennesael J. Low back pain caused by spi-
amide. Am J Med. 1957;23:596. nal tophus: a complication of gout in a kidney trans-
674. Kanner O, Jacobs P. Elevation of blood serum uric acid plant recipient. Nephrol Dial Transplant. 1998;13:
levels with chemotherapy. Transactions. 1957;16:244. 3245.
675. Wood RHN. Salicylates. Bull Rheum Dis. 693. Hausch R, Wilkerson M, Singh E, et al. Tophaceous
1963;13:297. gout of the thoracic spine presenting as back pain
676. Petty TL, Dalrymple GV. Inhibition of pyrazinamide and fever. J Clin Rheumatol. 1999;6:335.
hyperuricemia by small doses of acetylsalicylic acid. 694. Chan GLC, Canafax DM, Johnson CA. The thera-
Ann Intern Med. 1964;60:898. peutic use of azathioprine in renal transplantation.
677. Postlethwaite AE, Bartel AG, Kelley WN. Pharmacotherapy. 1987;7:165.
Hyperuricemia due to ethambutol. N Engl J Med. 695. Cummins D, Sekar M, Halil M, Banner N.
1972;286:761. Myelosuppression associated with azathioprine-al-
678. Postlethwaite AE, Bartel AG, Kelley WN. lopurinol interaction after heart and lung transplan-
Hyperuricemia induced by ethambutol. Adv Exp tation. Transplantation. 1996;61:1661.
Med Biol. 1974;41B:763. 696. Byrne PAC, Fraser AG, Pritchard MH. Treatment of
679. Palestine AG, Nussenblat RB, Chan CC. Side effects gout following cardiac transplantation. Br J Rheumatol.
of systemic cyclosporine in patients not undergoing 1996;35:1329.
tranplantation. Am J Med. 1984;77:652. 697. Jacobs F, Mamzer-Bruneel MF, Skhiri H, et al.
680. Tiller DJ, Hall BM, Hovrath JS, et al. Gout and Safety of the mycophenolate mofetil-allopurinol
hyperuricemia in patients on cyclosporine and combination in kidney transplant recipients with
diuretics. Lancet. 1985;1:453. gout. Transplantation. 1997;64:2087.
681. Chapman JR, Griffiths D, Harding NG, Morris PJ. 698. Perez Ruiz F, Calaboro M, Ferenendez Lopez J,
Reversibility of cyclosporine nephrotoxicity after et al. Treatment of chronic gout in patients with renal
three month’s treatment. Lancet. 1985;1:128. function impairment. J Clin Rheumatol. 1999;5:49.
682. VanHooff JP, Leunissen KML, vande Staak W. 699. Flury W, Ruch HR, Montandon A. The treatment of
Cyclosporin, uric acid, and the kidney. Lancet. hyperuricemia after kidney transplantation. Schweiz
1985;1:201. Med Wochenschr. 1977;107:1339.
683. Najarian JS, Fryd DS, Strand M, et al. A single insti- 700. Imanishi M, Ikegami M, Ishii T, et al. Clinical stud-
tution randomized prospective trial of cyclosporine ies on hyperuricemia and gout after transplantation.
versus azathioprine-antilymphocyte globulin for Hinyokika Kiyo. 1990;36:893.
immunosuppression in renal allografts recipients. 701. Matzkies F. Effects and side effects of Benzbromarone
Ann Surg. 1985;201:142. in the initial treatment of hyperuricemia and gout.
684. Burack DA, Griffith BP, Thompson ME, Kohl LE. Results of a field study of 3899 patients. Fortschr
Hyperuricemia and gout among heart transplant Med. 1978;96:1619.
recipients receiving cyclosporine. Am J Med. 702. Gehenot M, Horsmans V, Rahier J, Geubel AP.
1992;92:141. Subfulminant hepatitis requiring liver transplanta-
685. Clive DM. Renal transplant-associated hyperurice- tion after benzarone administration. J Hepatol.
mia and gout. J Am Soc Nephrol. 2000;11:974. 1994;20:842.
686. Pela I, Seracini D, Lavoratti G, Materassi M. Acute 703. Diamond HS. Uricosuric drugs. Handb Exp
gouty arthritis in adolescents with renal transplants. Pharmacol. 1977;51:459.
Pediatr Med Chir. 1999;21:135. 704. Sinclair DS, Fox IH. The pharmacology of the
687. Johnson DW, Saunders HJ, Johnson FJ, et al. hypouricemic effect of Benzbromarone. J Rheumatol.
Fibrogenic effects of cyclosporin A on the tubuloint- 1975;2:437.
erstitium: role of cytokines and growth factors. Exp 705. Camus JP, Prier A, Kartun P, et al. Thyreotoxicose et
Nephrol. 1999;7:470. benziodarone. Rev Rhum. 1973;40:2, 148.
688. Hansen JM, Fogh-Andersen N, Leyssac PP, 706. Davis S, Park YK, Abuchowski A, Davis FF.
Strandgaard S. Glomerular and tubular function in Hypouricemic effect of polyethylene glycol modified
renal transplant patients treated with and without urate oxidase. Lancet. 1987;2:281.
cyclosporin A. Nephron. 1998;80:450. 707. Chua CC, Greenberg LL, Viau AT, et al. Use of poly-
689. Edvardsson VO, Kaiser BA, Polinsky MS, et al. ethylene glycol-modified uricase (PEG-uricase) to
Natural history and etiology of hyperuricemia fol- treat hyperuricemia in a patient with non-Hodgkin
lowing pediatric renal transplantation. Pediatr lymphoma. Ann Intern Med. 1988;109:114.
Nephrol. 1995;9:57. 708. Ippoliti G, Negri M, Campana C, Vigano M. Urate
690. Laine J, Holmberg C. Mechanisms of hyperuricemia oxidase in hyperuricemic heart transplant recipients
in cyclosporine-treated renal transplanted children. treated with azathioprine. Transplantation. 1997;63:
Nephron. 1996;74:318. 1370.
691. Zurcher RM, Bock HA, Thiel G. Hyperuricaemia in 709. Rozenberg S, Koeger AC, Bourgeois P. Urate-
cyclosporine-treated patients: GFR-related effect. oxidase for gouty arthritis in cardiac transplant
Nephrol Dial Transplant. 1996;11:153. recipients. J Rheumatol. 1973;20:2171.
174 5 Clinical Aspects of Gout and Associated Disease States

710. Rozenberg S, Roche B, Dorent R, et al. Urate- tion in man during chronic administration of nicotinic
oxidase for the treatment of tophaceous gout in heart acid. Metabolism. 1971;20:1031.
transplant recipients. Rev Rhum Engl Ed. 727. Becker MA, Raivio KO, Meyer LJ, et al. Effects of
1995;65:392. nicotinic acid on human purine metabolism. Clin
711. Royer R, Lamarche M, Kissel P. Etude de l’action Res. 1973;21:616.
d’une urate-oxydase fongique sur l’uricemie et 728. Shuster L, Abraham G. The effect of nicotinamide
l’excretion de l’azote urique chez l’homme. Therapie. on incorporation in vivo of formate-C14. J Biol
1967;22:1113. Chem. 1959;234:129.
712. Brogard JM, Coumaros D, Frankhauser J, et al. 729. Shuster L, Goldin A. The incorporation of C14 glu-
Enzymatic uricolysis: a study of the effect of a fun- cose and C14 ribose into mouse diphosphopyridine
gal urate-oxydase. Eur J Clin Biol Res. 1972;17: nucleotide. J Biol Chem. 1958;230:873.
890. 730. Boyle JA, Raivio KO, Becker MA, et al. Effects of
713. Mourad G, Cristol JP, Chong G, et al. Role of pre- nicotinic acid on human fibroblast purine biosynthe-
cipitating anti-urate oxidase antibodies in urate oxi- sis. Biochim Biophys Acta. 1972;269:179.
dase-resistant hyperuricemia. Presse Med. 1984;13: 731. Oberhaemsli RD, Rajagopalan B, Taylor DJ. Study of
2585. hereditary fructose intolerance by use of 31-P mag-
714. Wolf G, Hegewisch-Becher S, Hossfeld DK, Stahl netic resonance spectroscopy. Lancet. 1987;2:931.
RA. Hyperuricemia and renal insufficiency associ- 732. Seegmiller JE, Dixon RM, Kemp GJ, et al. Fructose-
ated with malignant disease: urate oxidase as an induced aberration of metabolism in familial gout
efficient therapy? Am J Kidney Dis. 1999;34:E20. identified by 31-P magnetic resonance spectroscopy.
715. Pui CH, Relling MV, Lascombe F, et al. Urate oxi- Proc Natl Acad Sci USA. 1990;87:8326.
dase in prevention and treatment of hyperuricemia 733. Steinmann B, Gitzelmann R. The diagnosis of hered-
associated with lymphoid malignancies. Leukemia. itary fructose intolerance. Helv Paediatr Acta.
1997;11:1813. 1981;36:297.
716. Seidemann K, Meyer U, Jansen P, et al. Impaired 734. Perheentupa J, Raivio K. Fructose-induced hyperuri-
renal function and tumor lysis syndrome in pediatric cemia. Lancet. 1967;2:528.
patients with non-Hodgkin’s lymphoma and B-ALL. 735. Fox IH, Kelley WN. Studies on the mechanism of
Observations from the BFM trials. Klin Padiatr. fructose-induced hyperuricemia in man. Metabolism.
1998;210:279. 1972;21:713.
717. Rana SS, Guiliani MJ, Oddis ChV, Lacomis D. 736. Narins RG, Weisberg JS, Myers AR. Effects of car-
Acute onset of colchicine myoneuropathy in cardiac bohydrates on uric acid metabolism. Metabolism.
transplant recipients: case studies of three patients. 1974;23:455.
Clin Neurol Neurosurg. 1997;99:266. 737. Peaston MJT. Dangers of intravenous fructose.
718. Wallace SL, Singer JZ, Duncan GJ, et al. Renal func- Lancet. 1973;1:266.
tion predicts colchicine toxicity: guidelines for the 738. Kogut MD, Roe TF, Ng W, Nonnel GN. Fructose-
prophylactic use of colchicine in gout. J Rheumatol. induced hyperuricemia: observations in normal
1991;18:264. children and in patients with hereditary fructose
719. Garg G, Grundy SM. Nicotinic acid as therapy for intolerance and galactosemia. Pediatr Res.
dyslipidemia in non-insulin-dependent diabetes mel- 1975;9:774.
litus. JAMA. 1990;264:723. 739. Sahebjami DH, Scalettar R. Effect of fructose infu-
720. Kreisberg RA. Diabetic dyslipidemia. Am J Cardiol. sion on lactate and uric acid metabolism. Lancet.
1998;82:67u, 85u. 1971;1:366.
721. Gershon SL, Fox IH. Pharmacologic effects of nico- 740. Edwards NL, Gelfand EW, Biggar D, Fox IH. Partial
tinic acid on human purine metabolism. J Lab Clin deficiency of purine nucleoside phosphorylase: stud-
Med. 1974;84:179. ies of purine and pyrimidine metabolism. J Lab Clin
722. Schwartz ML. Severe reversible hyperglycemia as a Med. 1978;91:736.
consequence of niacin therapy. Arch Intern Med. 741. Kurtz TW, Kabra PM, Booth BE, et al. Liquid chro-
1993;153:2050. matographic measurements of inosine, hypoxanthine
723. Christensen A, Achor WP, Berge KG, et al. Nicotinic and xanthine in studies of fructose-induced degrada-
acid treatment of hypercholesterolemia. JAMA. tion of adenine nucleotides in humans and rats. Clin
1961;177:546. Chem. 1986;32:782.
724. Parsons WB. Studies of nicotinic acid use in hyperc- 742. Bode JC, Zelder O, Rumpelt HJ, Wittkamp U.
holesterolemia. Arch Intern Med. 1961;107:653. Depletion of liver adenosine phosphates and meta-
725. Berge KG, Achor WP, Christensen NA, et al. bolic effects of intravenous infusion of fructose and
Hypercholesterolemia and nicotinic acid: a long sorbitol in man and in the rat. Eur J Clin Invest.
term study. Am J Med. 1961;31:25. 1973;3:436.
726. Gant ZN, Pocelinko R, Solomon AM, et al. Oral glu- 743. Hultman E, Nilsson LH, Sahlin K. Adenine nucle-
cose tolerance, plasma insulin, and uric acid excre- otide content of human liver. Normal values and
References 175

fructose-induced depletion. Scand J Clin Lab Invest. after hemolysis by phenylhydrazine hydrochloride. J
1975;35:245. Biol Chem. 1929;83:409.
744. Buchli R, Meier D, Martin E, Boesiger P. Assessment 760. Sears WG. The occurrence of gout during treatment
of absolute metabolite concentrations in human tis- of pernicious anemia. Lancet. 1933;1:24.
sue by 31P MRS in vivo. Part II. Muscle, liver, kid- 761. Fox IH, Dotten DA, Marchant PJ. Alterations of
ney. Magn Reson Med. 1994;32:453. human purine metabolism in megaloblastic anemia.
745. Boesiger P, Buchli R, Meier D, et al. Changes of Adv Exp Med Biol. 1977;76B:249.
liver metabolite concentrations in adults with disor- 762. Luhby AL, Cooperman JM. Aminoimidazole-
ders of fructose metabolism after intravenous fruc- carboxamide excretion in vitamin-B12 and folic acid
tose by 31P magnetic resonance spectroscopy. deficiencies. Lancet. 1962;2:1381.
Pediatr Res. 1994;36:436. 763. Newcombe DS. The urinary excretion of aminoimi-
746. Oberhaensli RD, Galloway GJ, Taylor DJ, et al. dazolecarboxamide in the Lesch-Nyhan syndrome.
Assessment of human liver metabolism by phospho- Pediatrics. 1970;46:508.
rus-31 magnetic resonance spectroscopy. Br J Radiol. 764. Hernandez Nieto L, Brito Barroso ML, Nyhan WL.
1986;59:695. Anemia megaloblastica en la enfremedad de Lesch-
747. Dufour JF, Stoupis C, Lazeyras F, et al. Alterations Nyhan. Sangre (Barc). 1984;29:476.
in hepatic fructose metabolism in cirrhotic patients 765. Manzke H. Hyperuricamie mit cerebralparese syn-
demonstrated by dynamic 31phosphorus spectros- drom eines hereditaren purinstoffwechselleidens.
copy. Hepatology. 1992;15:835. Helv Paediatr Acta. 1967;22:258.
748. Sukuma H, Itabashi K, Takeda K, et al. Serial P-31 MR 766. Marie J, Royer P, Rappaport R. Hyperuricemie con-
spectroscopy after fructose infusion in patients with genitale avec troubles neurologiques, renaux et san-
chronic hepatitis. J Magn Reson Imaging. 1991;1:701. guins. Arch Fr Pediatr. 1967;24:401.
749. Van den Berghe G, Bronfman M, Vanneste R, Hers 767. van der Zee SPM, Monnens LAH, Schretlen EDAM.
HG. The mechanism of adenosine triphosphate Een hereditaire purinestoffwisselsstoormis met een
depletion in the liver after a load of fructose. Biochem cerebrale aandoening en megaloblastaire anemie
J. 1977;162:601. (syndroom von Lesch en Nyhan). Ned Tijdschr
750. Lamers JMJ, Hulsmann WC. The effect of fructose Geneeskd. 1968;112:1475.
on the stores of energy-rich phosphate in rat jejunum 768. van der Zee SPM, Lommen EJP, Trijbels JMF,
in vivo. Biochim Biophys Acta. 1973;313:1. Schretlen EDAM. The influence of adenine on the
751. Burch HB, Lowry OH, Meinhardt L, et al. Effect of clinical feature and purine metabolism in the
fructose, dihydroxyacetone, glycerol, and glucose Lesch-Nyhan syndrome. Acta Paediatr Scand.
on metabolites and related compounds in liver and 1970;59:259.
kidney. J Biol Chem. 1970;245:2092. 769. Henderson DA. The aetiology of chronic nephritis in
752. Morris RC, Nigon K, Reed EB. Evidence that the Queensland. Med J Aust. 1958;1:377.
depletion of inorganic phosphate determines the 770. Ball GV, Sorensen LB. Pathogenesis of hyperurice-
severity of the disturbance of adenine nucleotide mia in saturnine gout. N Engl J Med. 1968;
metabolism in the liver and renal cortex of the fruc- 280:1199.
tose-loaded rat. J Clin Invest. 1978;61:209. 771. Schwartz J, Levin R. The risk of lead toxicity in
753. Raivio KO, Becker MA, Meyer LJ, et al. Stimulation homes with lead paint hazard. Environ Res. 1991;
of human purine synthesis de novo by fructose infu- 54:1.
sion. Metabolism. 1975;24:861. 772. Delcourt JL, Hamrick HJ, O’Tauma LA, et al.
754. Itakura M, Sabina RL, Heald PW, Holmes EW. Basis Increased lead burden in children of battery workers:
for the control of purine biosynthesis by purine ribo- asymptomatic exposure resulting from contaminated
nucleotides. J Clin Invest. 1981;67:994. work clothing. Pediatrics. 1978;62:563.
755. Vincent MF, Van den Berghe G, Hers HG. Increase 773. Garrettson LK. Childhood lead poisoning in radiator
in phosphoribosyl pyrophosphate induced by ATP mechanics’ children. Vet Hum Toxicol. 1988;30:
and Pi depletion in hepatocytes. FASEB J. 112.
1989;3:1862. 774. Kaye WE, Novotny TE, Tucker M. New ceramics-
756. Kaye R, Williams ML, Barbero G. Comparative study related industry implicated in elevated blood lead
of glucose and fructose metabolism in infants with levels in children. Arch Environ Health. 1987;42:
reference to utilization and to the accumulation of gly- 161.
colytic intermediates. J Clin Invest. 1958;37:752. 775. Committee on Lead in the Human Environment:
757. Riddle MC. Endogenous uric acid metabolism in Environmental studies board, Commission on
pernicious anemia. J Clin Invest. 1929;8:69. Natural Resources, National Research Council,
758. Opsahl R. Hematopoiesis and endogenous uric acid. National Academy of Sciences; 1980.
Acta Med Scand. 1939;102:611. 776. Crutcher JE. Clinical manifestations and therapy of
759. Krafka Jr J. Endogenous uric acid and hematopoie- acute lead intoxication due to the ingestion of illic-
sis. II. Uric acid, reticulocytosis and erythrocytes itly distilled alcohol. Ann Intern Med. 1963;59:707.
176 5 Clinical Aspects of Gout and Associated Disease States

777. CDC. Elevated blood lead levels associated with among middle aged and elderly men: the normative
illicitly distilled alcohol – Alabama, 1990–1991. aging study. J Rheumatol. 2000;27:1708.
MMWR Morb Mortal Wkly Rep. 1992;41:294. 800. Richet G, Albahary C, Ardaillou R, et al. The kidney
778. Mustajoki P. Lead poisoning from illicit alcohol. in chronic lead poisoning. Rev Fr Etud Clin Biol.
Duodecim. 1978;94:993. 1964;50:188.
779. Ellis T, Lacy R. Illicit alcohol (moonshine) con- 801. Richet G, Albahary C, Morel-Maroger L, et al. Renal
sumption in West Alabama revisited. South Med J. changes in 23 cases of occupational lead poisoning.
1998;91:858. Bull Mem Soc Hop Paris. 1966;117:441.
780. Lacy R, Winternitz WW. Moonshine consumption in 802. Craswell PW, Price J, Boyle PD, et al. Patterns of
West Alabama. Ala J Med Sci. 1984;21:364. lead excretion in patients with gout and chronic renal
781. Pegues DA, Hughes BJ, Woernle CH. Elevated blood failure: a comparative German and Australian study.
lead levels associated with illegally distilled alcohol. Sci Total Environ. 1987;66:17.
Arch Intern Med. 1993;153:1501. 803. Wright LF, Saylor RP, Cecere FA. Occult lead intox-
782. Halla JT, Ball GV. Saturnine gout: a review of 42 ication in patients with gout and kidney disease.
patients. Semin Arthritis Rheum. 1982;11:307. J Rheumatol. 1984;11:517.
783. Montgomery R, Finkenbine R. A brief review of 804. Craswell PW, Price J, Boyle PD, et al. Chronic renal
moonshine use. Psychiatr Serv. 1999;50:1088. failure with gout: a marker of chronic lead poison-
784. Ball GV. Two epidemics of gout. Bull Hist Med. ing. Kidney Int. 1984;26:319.
1971;45:401. 805. Colleoni N, D’Amico G. Chronic lead accumulation
785. Baker G. An essay concerning the cause of the endemial as a possible cause of renal failure in gouty patients.
colic of Devonshire. London: J. Hughs; 1767. Nephron. 1986;44:32.
786. Sherlock JC, Pickford CJ, White GF. Lead in alco- 806. Albahary C, Richet G, Guillaume J, Morel-Maroger
holic beverages. Food Addit Contam. 1986;3:347. L. The kidney in occupational lead poisoning. Arch
787. Molinini R, Caravella R, Carino M, Nuzzaco A. Mal Prof. 1965;26:5.
Three cases of poisoning caused by wine contami- 807. Sanchez-Fructuoso AI, Torralbo A, Arroyo M, et al.
nated with lead. G Ital Med Lav. 1985;7:101. Occult lead intoxication as a cause of hypertension and
788. Hight SC. Lead migration from lead crystal wine renal failure. Nephrol Dial Transplant. 1996;11:1775.
glasses. Food Addit Contam. 1996;13:747. 808. Loghman-Adham M. Renal effects of environmental
789. Probst-Hensch N, Braun-Fabrlaender C, Bodenmann A, and occupational lead exposure. Environ Health
Ackermann-Liebrich U. Alcohol consumption and Perspect. 1997;105:928.
other lifestyle factors: avoidable sources of excess lead 809. Perazella MA. Lead and the kidney: nephropathy,
exposure. Soz Praventivmed. 1993;38:43. hypertension, and gout. Conn Med. 1996;60:521.
790. Tumpowsky CM, Davis LK, Rabin R. Elevated 810. Batuman V. Lead nephropathy, gout, and hyperten-
blood lead levels among adults in Massachusetts, sion. Am J Med Sci. 1993;30:241.
1991–1995. Public Health Rep. 2000;115:364. 811. Cledes J, Allain P. Chronic lead nephropathy.
791. Garrod AB. The nature and treatment of gout and Epidemiology and diagnosis. Presse Med. 1992;
rheumatic gout. London: Walton & Maberg; 1859. 21:759.
792. Emmerson BT. Chronic lead nephropathy. Kidney 812. Nolan CV, Shaikh ZA. Lead nephrotoxicity and
Int. 1973;4:1. associated disorders: biochemical mechanisms.
793. Emmerson BT. Chronic lead nephropathy: the diag- Toxicology. 1992;73:127.
nostic uses of calcium EDTA and the association 813. Peitzman SJ, Bodison W, Ellis I. Moonshine drink-
with gout. Australas Ann Med. 1963;12:310. ing among hypertensive veterans in Philadelphia.
794. Emmerson BT. The clinical differentiation of lead Arch Intern Med. 1985;145:632.
gout from primary gout. Arthritis Rheum. 1968; 814. Miranda ME, Puig JG, Mateos FA, et al. The role of
11:623. lead in gout nephropathy reviewed: pathogenic or
795. Reynolds PR, Knapp MJ, Baraf HSB, Holmes EW. associated factor? Adv Exp Med Biol. 1991;309A:
Moonshine and lead. Relationship to the pathogene- 209.
sis of hyperuricemia and gout. Arthritis Rheum. 815. Cullen MR, Robins JM, Eskenazi B. Adult inorganic
1983;26:1057. lead intoxication: presentation of 31 new cases and a
796. Batuman V, Maesako JK, Haddad B, et al. The role review of recent advances in the literature. Medicine.
of lead in gout nephropathy. N Engl J Med. 1981; 1983;62:221.
304:520. 816. Batuman V, Landy E, Maesaka JR, Weeden RP.
797. Poor G, Mituszova M. Saturnine gout. Baillieres Contribution of lead to hypertension with renal
Clin Rheumatol. 1989;3:51. impairment. N Engl J Med. 1983;309:17.
798. Cooper WC, Wong O, Kheifets L. Mortality among 817. Rabinowitz MB, Wetherill GW, Kopple JD. Kinetic
employees of lead battery plants and lead-producing analysis of lead metabolism in healthy humans.
plants. Scand J Work Environ Health. 1985;11:331. J Clin Invest. 1976;58:260.
799. Shadick NA, Kim R, Weiss S, et al. Effect of low 818. Rempel D. The lead-exposed worker. JAMA.
level lead exposure on hyperuricemia and gout 1989;262:532.
References 177

819. Occupational exposure to lead. Final standard. 43 837. Tutunji MF, al-Mahasneh QM. Disappearance of
Federal Register 220:52952–53014 and 225:54353– heme metabolites following chelation therapy with
54616, 1981, 46 Federal Register 238:60758–60766, meso 2,3-dimercaptosuccinic acid (DMSA). J
1981. Toxicol Clin Toxicol. 1994;32:267.
820. 29 CFR 1910.1025 and 1926.62. 838. Aposhian HV, Maiorino RM, Gonzalez-Ramirez D,
821. Chisholm Jr JJ. Mobilization of lead by calcium et al. Mobilization of heavy metals by newer, thera-
disodium edetate. Am J Dis Child. 1987;141:1256. peutically useful chelating agents. Toxicology.
822. Weinberger HL, Post EM, Schneider T, et al. An 1995;97:23.
analysis of 248 initial mobilization tests performed 839. Parru S, Alessio L. The use of chelating agents in
on an ambulatory basis. Am J Dis Child. 1987; occupational lead poisoning. Occup Med (Lond).
141:1266. 1996;46:41.
823. Cory-Slechta DA, Weiss B, Cox C. Mobilization 840. Lifshitz M, Hashkanazi R, Phillip M. The effect of
and redistribution of lead over the course of CaEDTA 2,3-dimercaptosuccinic acid in the treatment of lead
chelation therapy. J Pharmacol Exp Ther. 1987; poisoning in adults. Ann Med. 1997;29:83.
243:804. 841. Berlin Jr CM. Lead poisoning in children. Curr Opin
824. Hu H, Aro A, Payton M, et al. The relationship of Pediatr. 1997;9:173.
bone and blood lead to hypertension. The normative 842. Besunder JB, Super DM, Anderson RL. Comparison
aging study. JAMA. 1996;275:1171, 1996;276: of dimercaptosuccinic acid and calcium disodium eth-
1038. ylenediaminetetraacetic acid versus dimercaptopro-
825. Hu H, Raabinowitz M, Smith D. Bone lead as a bio- panol and ethylenediaminetetraacetic acid in children
logical marker in epidemiologic studies of chronic with lead poisoning. J Pediatr. 1997;130:966.
toxicity: conceptual paradigms. Environ Health 843. Miller AL. Dimercaptosuccinic acid (DMSA), a
Perspect. 1998;106:1. non-toxic, water-soluble treatment for heavy metal
826. Todd AC. Calculating bone-lead measurement vari- toxicity. Altern Med Rev. 1998;3:199.
ance. Environ Health Perspect. 2000;108:383, 844. Restek-Samarzija N, Blanusa M, Pizent A, et al.
2000;108:A298. Meso-2,3-dimercaptosuccinic acid in the treatment
827. Wedeen RP, Ty A, Udasin I, et al. Clinical applica- of occupationally exposed lead workers. Arh Hig
tion of in vivo tibial K-XRF for monitoring lead Rada Toksikol. 1998;49:137.
stores. Arch Environ Health. 1995;50:355. 845. The Treatment of Lead-Exposed Children (TLC)
828. Bateman SN, Pejovic-Milic A, Stronach IM, et al. trial: design and recruitment for a study of the
Performance appraisals of digital spectroscopy sys- effect of oral chelation on growth and development
tems for the measurement of bone lead. Appl Radiat in toddlers. Paediatr Perinat Epidemiol. 1998;
Isot. 2000;53:647. 12:313.
829. Ambrose TM, Al-Lozi M, Scott MG. Bone lead con- 846. Lever SZ, Parsons TL. The presence of lead decreases
centrations assessed by in vivo x-ray fluorescence. the availability of meso-2,3-dimercaptosuccinic acid
Clin Chem. 2000;46:1171. for analysis in the monobromobimane assay. Chem
830. Wedeen RD. In vivo tibial XRF measurements of Res Toxicol. 1999;12:1057.
bone lead. Arch Environ Health. 1990;45:69. 847. Smith DR, Calacsan C, Woolard D, et al. Succimer
831. Rosen JF, Markowitz ME, Bijur PE, et al. L-line and the urinary excretion of essential elements in a
x-ray fluorescence of cortical bone lead compared primate model of childhood lead exposure. Toxicol
with CaNa2EDTA test in lead-toxic children: public Sci. 2000;54:473.
health implications. Proc Natl Acad Sci USA. 848. Chisholm Jr JJ. Safety and efficacy of meso-2,3-di-
1989;86:685. mercaptosuccinic acid (DMSA) in children with
832. Friedheim E, Graziano JH, Popovac D, et al. elevated blood lead concentrations. J Toxicol Clin
Treatment of lead poisoning by 2,3-dimercaptosuc- Toxicol. 2000;38:365.
cinic acid. Lancet. 1978;2:1234. 849. Ellis MR, Kane KY. Lightening the lead load in chil-
833. Graziano JH, Siris ES, Lolacono N, et al. dren. Am Fam Physician. 2000;62:545, 559.
2,3-Dimercaptosuccinic acid as an antidote for lead 850. Markowitz M. Lead poisoning. Pediatr Rev.
intoxication. Clin Pharmacol Ther. 1985;37:431. 2000;21:327.
834. Chisholm Jr JJ, Thomas DJ. Use of 2,3-dimercapto- 851. Eisner ER, Schumacher HR, Sidransky E. Gout and
propane-1-sulfonate in treatment of lead poisoning Gaucher disease. Arthritis Rheum. 2000;43:S122.
in children. J Pharmacol Exp Ther. 1985;235:665. 852. Chang-Lo M, Yam LT, Rubenstone AI, Schwartz
835. Graziano JH, Lolacono NJ, Meyer P. Dose–response SO. Gaucher’s disease associated with chronic lym-
of oral 2,3-dimercaptosuccinic acid in children with phocytic leukemia, gout and carcinoma. J Pathol.
elevated blood lead concentrations. J Pediatr. 1975;116:203–7.
1988;113:751. 853. Liel Y, Hausmann MJ, Moses M. Case report: seren-
836. Chisholm Jr JJ. BAL, EDTA, DMSA and DMPS in dipitous Gaucher’s disease presenting as elevated
the treatment of lead poisoning in children. J Toxicol erythrocyte sedimentation rate due to monoclonal
Clin Toxicol. 1992;30:493. gammopathy. Am J Med Sci. 1991;301:393.
178 5 Clinical Aspects of Gout and Associated Disease States

854. Shoenfeld Y, Berliner S, Pinkhas J, Beutler E. The 873. Scudmore C. A treatise of the nature and course
association of Gaucher’s disease and dysproteine- of gout and gravel. 4th ed. London: Mallett; 1823.
mias. Acta Haematol (Basel). 1980;64:241. p. 64.
855. Turesson T, Rausing A. Gaucher’s disease and 874. Williamson CS. Gout: a clinical study of one hun-
benign monoclonal gammopathy. A case report with dred and sixteen cases. JAMA. 1920;74:1625.
immunofluorescence study of bone marrow and 875. Brochner-Mortensen K. One hundred gouty patients.
spleen. Acta Med Scand. 1975;197:507. Acta Med Scand. 1941;106:81.
856. Pratt PW, Estren F, Kochwa S. Immunoglobin abnor- 876. Grahame R, Scott JT. Clinical survey of 354 patients
malities in Gaucher’s disease. Report of a case. with gout. Ann Rheum Dis. 1970;29:461.
Blood. 1968;31:633. 877. McCracken JP, Owen PS, Pratt JH. Gout: still a for-
857. Mark T, Dominguez C, Rywlin AM. Gaucher’s dis- gotten disease. JAMA. 1946;131:367.
ease associated with chronic lymphocytic leukemia. 878. Seegmiller JE. Diseases of purine and pyrimidine
South Med J. 1982;75:361. metabolism. In: Bondy PK, Rosenberg LE, editors.
858. Kaufman S, Rozenfeld V, Yona R, Varon M. Metabolic control and disease. 8th ed. Philadelphia:
Gaucher’s disease associated with chronic lympho- Saunders; 1980. p. 777.
cytic leukaemia. Clin Lab Haematol. 1986;8:321. 879. Emmerson BT, Wyngaarden JB. Purine metabolism
859. Bruckstein AH, Karanas A, Dire JJ. Gaucher’s dis- in heterozygous carriers of hypoxanthine-guanine
ease associated with Hodgkin’s disease. Am J Med. phosphoribosyltransferase deficiency. Science.
1980;68:610. 1969;166:1533.
860. Cho SY, Sastre M. Coexistence of Hodgkin’s disease 880. Yu TF. Some unusual features of gouty arthritis in
and Gaucher’s disease. Am J Clin Pathol. 1976; females. Arthritis Rheum. 1977;6:247.
65:103. 881. Burch TA, O’Brien WM, Reed R, et al. Hyperuricemia
861. Paulson JA, Marti GI, Fink JK, et al. Richter’s trans- and gout in Mariana Islands. Ann Rheum Dis.
formation of lymphoma complicating Gaucher’s dis- 1966;25:114.
ease. Hematol Pathol. 1989;3:91. 882. Reed D, Labarthe D, Stallones R. Epidemiologic
862. Gal R, Gukovsky-Oren S, Floru S, et al. Sequential studies of serum uric acid levels among Micronesians.
appearance of breast carcinoma, multiple myeloma Arthritis Rheum. 1972;15:381.
and Gaucher’s disease. Haematologica. 1979;73:63. 883. Prior IAM, Rose BS, Harvey HPB, et al.
863. Garfinkel D, Sidi Y, Ben-Bassat M, et al. Coexistence Hyperuricemia, gout, and diabetic abnormality in
of Gaucher’s disease and multiple myeloma. Arch Polynesian people. Lancet. 1966;1:333.
Intern Med. 1982;142:2229. 884. Lennane GAQ, Rose BS, Isdale IC. Gout in Maori.
864. Benjamin D, Joshua H, Djaldetti M, et al. Ann Rheum Dis. 1960;19:120.
Nonsecretory IgD-kappa multiple myeloma in a 885. Healey LA, Jones KW. Hyperuricemia in American
patient with Gaucher’s disease. Scand J Haematol. Samoans. Arthritis Rheum. 1971;14:283.
1979;22:179. 886. Chang SJ, Ko YC, Wang TN, et al. High prevalence
865. Lamon J, Miller W, Tavassoli M, et al. Specialty of gout and related risk factors in Taiwan’s
conference: multiple myeloma complicating Aborigines. J Rheumatol. 1997;24:1364.
Gaucher’s disease. West J Med. 1982;136:122. 887. Talbott JH. Gout. New York: Grune & Stratton;
866. Ruestow PC, Levinson DJ, Catchatourian R, et al. 1964. p. 128.
Coexistence of IgA myeloma and Gaucher’s disease. 888. Greenhut IJ, Silver RA, Campbell JA. Occurrence of
Arch Intern Med. 1980;140:1115. gout in a female: report of an unusual case. Radiology.
867. Beutler E, Grabowski GA. Gaucher disease. In: 1953;60:257.
Scriver CR, Beaudet AL, Sly WS, Valle D, editors. 889. Hill IC. Gout. Lancet. 1938;1:826.
The metabolic and molecular bases of inherited dis- 890. Weingold AB. Gout and pregnancy. Obstet Gynecol.
ease. New York: McGraw-Hill, Inc; 1995. p. 2643. 1960;16:309.
868. Beutler E, Gelbart T, Kuhl W, et al. Mutations in 891. Lee FI, Loeffler FE. Gout and pregnancy. J Obstet
Jewish patients with Gaucher disease. Blood. Gynaecol Br Comm. 1962;69:299.
1992;79:1662. 892. Friedman EA, Little WA. Pregnancy and gout: a case
869. Beutler E, Gelbart T, Kuhl W, et al. Identification of report. Am J Obstet. 1958;76:913.
the second common Jewish Gaucher disease muta- 893. Batt RE, Cirksena WJ, Lebherz TB. Gout and salt
tion makes population based screening for the wasting renal disease during pregnancy; Diagnosis,
heterozygote state. Proc Natl Acad Sci USA. management, and follow-up. JAMA. 1963;186:835.
1991;88:10544. 894. Mertz DP, Henninges D, Schwoerer P. Gout and
870. Beutler E. Gaucher disease: new molecular pregnancy. An unusual case with severe hyperlipo-
approaches to diagnosis and treatment. Science. proteinaemia. Munch Med Wochenschr. 1972;
1992;256:794. 114:658.
871. Hadler NM, Franck WA, Bress NM, et al. Acute pol- 895. Kelsall JT, O’Hanlon DP. Gout during pregnancy.
yarticular gout. Am J Med. 1974;56:715. J Rheumatol. 1994;21:1365.
872. Hench PS. Gout and gouty arthritis. In: Cecil RL, 896. Lee IS, Hsu CD. Placental pathologies are not asso-
Loeb RF, editors. Textbook of medicine. 9th ed. ciated with hyperuricemia in preeclamptic pregnan-
Philadelphia: Saunders; 1955. p. 643. cies. Conn Med. 1999;63:459.
References 179

897. Fadel HE, Northrop G, Misenhimer HR. 918. Gertler MM, Garn SM, Levine SA. Serum uric acid
Hyperuricemia in pre-eclampsia. A reappraisal. Am in relation to age and physique in health and in coro-
J Obstet Gynecol. 1976;125:640. nary artery disease. Ann Intern Med. 1951;34:
898. Riedel H, Eisenbach GM, Henkel E, et al. The clini- 1421.
cal significance of hyperuricemia for the prognosis 919. Fessel JW, Siegelaub AB, Johnson ES. Correlates
of pregnancy toxemia. Fortschr Med. 1978;96:58. and consequences of asymptomatic hyperuricemia.
899. Chesley LC. Diagnosis of preeclampsia. Obstet Arch Intern Med. 1973;132:44.
Gynecol. 1985;65:423. 920. Loenen HM, Eshuis H, Lowik MR, et al. Serum uric
900. Mauy A, Hubel CA, Roberts JA. Hyperuricemia and acid correlates in elderly men and women with spe-
xanthine oxidase in preeclampsia, revisited. Am cial reference to body composition and dietary intake
J Obstet Gynecol. 1996;174:288. (Dutch Nutrition Surveillance System). J Clin
901. Mustaphi R, Gopalan S, Dhaliwal L, Surkar AK. Epidemiol. 1990;43:1297.
Hyperuricemia and pregnancy induced hypertension 921. Emmerson BT, Knowles BR. Triglyceride concen-
– reappraisal. Indian J Med Sci. 1996;50:68. trations in primary gout and gout of chronic lead
902. Merviel P, Ba R, Beaufils M, et al. Lone hyperurice- nephropathy. Metabolism. 1971;20:721.
mia during pregnancy: maternal and fetal outcomes. 922. Heydon S. The workingman’s diet. II. Effect of
Eur J Obstet Gynecol Reprod Biol. 1998;77:145. weight reduction in obese patients with hyperten-
903. Hsiung R, Hasselmann M, Lutun P, Gordji M. Acute sion, diabetes, hyperuricemia and hyperlipidemia.
fatty liver of pregnancy. Diagnostic value of hyperu- Nutr Metab. 1978;22:141.
ricemia in the pre-jaundice stage. J Gynecol Obstet 923. Berchtold P, Berger M, Grieser E, et al. Cardiovascular
Biol Reprod (Paris). 1988;17:901. risk factors in gross obesity. Int J Obes. 1977;1:219.
904. Nishioka N, Mikanagi K. Clinical features of 4,000 924. Fessel WJ. High uric acid as an indicator of cardio-
gouty subjects in Japan. Adv Exp Med Biol. vascular disease. Independence from obesity. Am
1980;122A:47. J Med. 1980;68:401.
905. Cornelius R, Schneider HJ. Gouty arthritis in the 925. Bonora E, Targher G, Zenere MB, et al. Relationship
adult. Radiol Clin North Am. 1988;26:1267. of uric acid concentration to cardiovascular risk fac-
906. Deesomchok U, Tumrasvin T. A clinical comparison tors in young men. Role of obesity and central fat
of females and males with gouty arthritis. J Med distribution. The Verona Young Men Atherosclerosis
Assoc Thai. 1989;72:510. Risk Factor Study. Int J Obes Relat Metab Disord.
907. Hauge M, Harvold B. Heredity in gout and hyperuri- 1996;20:975.
cemia. Acta Med Scand. 1955;152:247. 926. Chu NF, Wang DJ, Liou SH, Shieh SM. Relationship
908. O’Sullivan JB. Gout in a New England town. A prev- between hyperuricemia and other cardiovascular dis-
alence study in Sudbury, Massachusetts. Ann Rheum ease risk factors among adult males in Taiwan. Eur
Dis. 1969;31:166. J Epidemiol. 2000;16:13.
909. Popert AJ, Hewitt JV. Gout and hyperuricemia in 927. Yamashita S, Matsuzawa Y, Tokunaga K, et al.
rural and urban populations. Ann Rheum Dis. Studies on the impaired metabolism of uric acid in
1962;21:154. obese subjects: marked reduction of renal urate
910. Smyth CJ, Stecher RM, Wolfson WQ. Genetic and excretion and its improvement by a low-calorie diet.
endocrine determinants of the plasma urate level. Int J Obes. 1986;10:255.
Science. 1948;108:524. 928. Takahashi S, Yauramoto T, Tsutsumi Z, et al. Close
911. Delbarre F, Braun S, St. Georges-Chaumet F. La correlation between visceral fat accumulation and
goutte feminine (Analyse de quarante observations). uric acid metabolism in healthy men. Metabolism.
Semin Hop (Paris). 1967;43:623. 1997;46:1162.
912. Cowdry SC. Hyperuricemia in infectious mononu- 929. Matsuura F, Yamashita S, Nakamura T, et al. Effect
cleosis. JAMA. 1966;196:319. of visceral fat obesity on uric acid metabolism in
913. Nessan VJ, Geerken RC, Ulvilla J. Uric acid excre- male obese subjects: visceral fat obesity is linked
tion in infectious mononucleosis: a function of more closely to overproduction of uric acid than
increased purine turnover. J Clin Endocrinol Metab. subcutaneous fat obesity. Metabolism. 1998;
1974;38:652. 47:929.
914. Diamond H, Sharon E, Holden D. Renal handling of 930. Fruehwald-Schultes B, Peters A, Kern W, et al.
uric acid in sickle cell anemia. In: Sperling O, Serum leptin is associated with serum uric acid con-
DeVries A, Wyngaarden JB, editors. Purine metabo- centrations in humans. Metabolism. 1999;48:677.
lism in man. New York: Plenum; 1973. p. 759. 931. Monocci A, Savia G, Lucantoni R, et al. Leptin
915. Glynn RJ, Campion EW, Silbert JE. Trends in serum plasma concentrations are dependent on body fat
uric acid levels. 1961–1980. Arthritis Rheum. distribution in obese patients. Int J Obes Relat Metab
1983;26:87. Disord. 2000;24:1139.
916. Myers A, Epstein FH, Dodge HJ, Mikkelsen WM. 932. Shmulewitz D, Auerbach SB, Lehner T, et al.
The relationship of serum uric acid to risk factors in Epidemiology and factor analysis of obesity, type II
coronary heart disease. Am J Med. 1968;45:520. diabetes, hypertension, and dyslipidemia (syndrome
917. Krizek V. Serum uric acid in relation to body weight. X) on the island of Kosrae. Federation states of
Ann Rheum Dis. 1966;25:456. Micronesia. Hum Hered. 2001;51:8.
180 5 Clinical Aspects of Gout and Associated Disease States

933. Leyva F, Godsland IF, Ghatei M, et al. 953. Fallat RW, Glueck CJ. Familial and acquired type V
Hyperleptinemia as a component of a metabolic syn- hyperlipoproteinemia. Atherosclerosis. 1976;23:41.
drome of cardiovascular risk. Arterioscler Thromb 954. Lipid Research Clinics Programs: the prevalence
Vasc Biol. 1998;18:928. study. In: Population studies data book I. Bethesda:
934. Fried SK, Ricci MR, Russell CD, Laferrere B. US Department of Health and Human Services,
Regulation of leptin production in humans. J Nutr. National Institutes of Health, Publication 80–1527;
2000;130:3127. 1980.
935. Khaodhiar L, McCowen KC, Blackburn GL. Obesity 955. Hunt SC, Wu LL, Hopkins PN, et al. Apolipoprotein,
and its comorbid conditions. Clin Cornerstone. low density lipoprotein subfraction and insulin asso-
1999;2:17. ciations with familial combined hyperlipidemia.
936. Benedek TG. Correlations of serum uric acid and Study of Utah patients with familial dyslipidemic
lipid concentrations in normal, gouty and atheroscle- hypertension. Arteriosclerosis. 1989;9:335.
rotic men. Ann Intern Med. 1967;66:851. 956. Williams RR, Hunt SC, Hopkins PN, et al. Familial
937. Berkowitz D. Gout, hyperlipidemia and diabetes dyslipidemic hypertension: Evidence from 58 Utah
interrelationships. JAMA. 1966;197:77. families for a syndrome present in approximately
938. Berkowitz D. Blood lipid and uric acid interrelation- 12% of patients with essential hypertension. JAMA.
ships. JAMA. 1964;190:856. 1988;259:3579.
939. Feldman EB, Wallace SL. Hypertriglyceridemia in 957. Goldstein JL, Schrott HG, Hazzard WL, et al.
gout. Circulation. 1964;29:508. Hyperlipidemia in coronary artery disease. II.
940. Barlow KA. Hyperlipidemia in primary gout. Genetic analysis of lipid levels of 176 families and
Metabolism. 1968;17:289. delineation of a new inherited disorder, combined
941. Gibson T, Grahame R. Gout and hyperlipidemia. hyperlipidemia. J Clin Invest. 1973;52:1544.
Ann Rheum Dis. 1974;33:298. 958. Rose HG, Kranz P, Weinstock M, et al. Inheritance
942. Jiao S, Kameda K, Matsuzawa Y, Tarui S. of combined hyperlipoproteinemia. Evidence for
Hyperlipoproteinaemic phenotype and influence of a new lipoprotein phenotype. Am J Med. 1973;
alcohol intake and obesity in Japan. Ann Rheum Dis. 54:148.
1986;44:390. 959. Vaverkova H, Weinberg AO, Horcicika V, et al.
943. Ferns GA, Lanham J, Stocks J, et al. The measure- Familial combined hyperlipidemia. Part I. Lipid val-
ment of high density lipoprotein subfractions in ues and the lipoprotein pattern. Acta Univ Palacki
patients with primary gout using a simple precipita- Olomuc Fac Med. 1986;113:193.
tion method. Ann Clin Biochem. 1985;22:526. 960. Austin MA, Brunzell J, Fitch WL, Krauss RM.
944. Ulreich A, Kostner GM, Pfeiffer KP, et al. Serum Inheritance of low density lipoprotein subclass pat-
lipids and lipoproteins in patients with primary gout. terns in familial combined hyperlipidemia.
Rheumatol Int. 1985;5:73. Arteriosclerosis. 1990;10:520.
945. Jacobelli S, Artega A, Bidegain F. Cholesterol distri- 961. Austin MA, King M-C, Vranizan KM, Krauss RM.
bution among lipoprotein fractions in patients with Atherogenic lipoprotein phenotype: a proposed
gout and normal controls. J Rheumatol. 1986; genetic marker for coronary heart disease.
13:774. Circulation. 1990;82:495.
946. Phillipi T, Barrett-Connor E. Fasting plasma glu- 962. Pitkanen OP, Nuutila P, Raitakari OT, et al. Coronary
cose, uric acid, and triglycerides as predictors of the flow reserve in young men with familial combined
ration of total cholesterol to HDLC. Am J Clin hyperlipidemia. Circulation. 1999;99:1678.
Pathol. 1984;82:329. 963. Pajukanta P, Porkka KV. Genetics of familial com-
947. Perez-Ruiz F, Calabozo M, Herrero-Beites AM, bined hyperlipidemia. Curr Atheroscler Rep. 1999;
et al. Improvement of renal function in patients with 1:79.
chronic gout after proper control of hyperuricemia 964. Gehrisch S, Kostka H, Tiebel M, et al. Mutations of
and gouty bouts. Nephron. 2000;86:287. the hepatic lipase gene in patients with combined
948. Harris-Jones JN. Hyperuricemia and essential hyper- hypertriglyceridemia/hyperalphalipoproteinemia
cholesterolemia. Lancet. 1957;1:857. and in patients with familial combined hyperlipi-
949. Salvini L, Verdi G. Statistical study on correlation demia. J Mol Med. 1999;77:728.
between blood level of cholesterol beta/alpha lipo- 965. Jarvik GP, Beaty TH, Gallagher PR, et al. Genotype
protein ratio and uric acid of normal and arterioscle- at a major locus with large effects on apolipoprotein
rotic subjects. Gerontologia. 1959;3:327. B levels predicts familial combined hyperlipidemia.
950. Jensen J, Blankenhorn DH, Dornerup V. Blood uric Genet Epidemiol. 1993;10:257.
acid levels in familial hypercholesterolemia. Lancet. 966. Ribalta J, La Ville AE, Heras M, et al. Familial com-
1966;1:298. bined hyperlipidemia: detection and characterization
951. Strejcek J, Kucerova L. Idiopathic hyperlipidemia of the hyperlipidemic profile among children and
and gout. Acta Rheumatol Scand. 1968;14:95. adolescents. Med Clin (Barc). 1997;109:161.
952. Brunzele JD, Bierman EL. Chylomicronemia syn- 967. Aouizerat BE, Allayee H, Bodnar J, et al. Novel
drome: Interaction of genetic and acquired hyper- genes for familial combined hyperlipidemia. Curr
triglyceridemia. Med Clin North Am. 1982;66:455. Opin Lipidol. 1999;10:113.
References 181

968. Sasaki J. Familial combined hyperlipidemia. Nippon 986. Havel RJ. Familial dysbetalipoproteinemia. New
Rinsho. 1999;57:2776. aspects of pathogenesis and diagnosis. Med Clin
969. East C, Bilheimer DW, Grundy SM. Combination North Am. 1982;66:441.
drug therapy for familial combined hyperlipidemia. 987. Mahley RW, Weisgraber KH, Innerarity TL, Rall Jr
Ann Intern Med. 1988;109:25. SC. Genetic defects in lipoprotein metabolism:
970. Fredrickson DS, Goldstein JS, Brown MS. The elevation of atherogenic lipoproteins caused by
familial hyperlipoproteinemias. In: Stanbury JB, impaired catabolism. JAMA. 1991;265:78.
Wyngaarden JB, Fredrickson DS, editors. The meta- 988. Mahley RW, Hussain MM. Chylomicron and chy-
bolic basis of inherited disease. 4th ed. New York: lomicrons remnant catabolism. Curr Opin Lipidol.
McGraw-Hill, Inc; 1978. p. 604. 1991;2:170.
971. Bergeron J, Normand T, Bharucha A, et al. 989. Rall Jr SC, Mahley RW. The role of apolipoprotein
Prevalence, geographical distribution and genealogi- E genetic variants in lipoprotein disorders. J Intern
cal investigations of mutation 188 of lipoprotein Med. 1992;231:653.
lipase gene in French Canadian population of 990. Hazzard WR, Bierman EL. Aggravation of broad-ß
Quebec. Clin Genet. 1992;41:206. disease (type 3 hyperlipoproteinemia) by hypothy-
972. Wilson DE, Emi M, Iverius P-H, et al. Phenotypic roidism. Arch Intern Med. 1972;130:822.
expression of heterozygous lipoprotein lipase 991. Feussner G, Ziegler R. Expression of type III hyper-
deficiency in the extended pedigree of a proband lipoproteinemia in a subject with secondary hypo-
homozygous for a missense mutation. J Clin Invest. thyroidism bearing the apolipoprotein E2/2
1990;86:735. phenotype. J Intern Med. 1991;230:183.
973. Breckenridge WC, Little JA, Steiner G, et al. 992. Brown MS, Goldstein JL. Lipoprotein receptors in
Hypertriglyceridemia associated with deficiency of the liver. Control of signals for plasma cholesterol
apolipoprotein C-II. N Engl J Med. 1978;298:1265. traffic. J Clin Invest. 1983;72:743.
974. Brunzell JD, Miller NE, Alaupovic P, et al. Familial 993. Angelin B, Raviola CA, Innerarity TL, Mahley
Chylomicronemia due to a circulating inhibitor of RW. Regulation of hepatic lipoprotein receptors in
lipoprotein lipase activity. J Lipid Res. 1983;24:12. the dog. Rapid regulation of apolipoprotein B, E
975. Beaumont JL, Carlson LA, Cooper GR, et al. receptors, but not of apolipoprotein E receptors, by
Classification of hyperlipidemias and hyperlipopro- intestinal lipoproteins and bile acids. J Clin Invest.
teinemias. Bull World Health Organ. 1971;43:891. 1983;71:816.
976. Greenberg BH, Blackwelder WC, Levy RI. Primary 994. Myant NB. Cholesterol metabolism, LDL, and IDL
type V hyperlipoproteinemia. A descriptive study of receptors. San Diego: Academic; 1990.
32 families. Ann Intern Med. 1977;87:526. 995. Havel RJ, Goldstein JL, Brown MS. Lipoproteins
977. Chait A, Robertson HT, Brunzell JD. Chylomicronemia and lipid transport. In: Bondy PK, Rosenberg LE,
syndrome in diabetes mellitus. Diabetes Care. 1981; editors. Metabolic control and disease. 8th ed.
4:343. Philadelphia: Saunders; 1980. p. 393.
978. Chait A, Brunzell JD. Severe Hypertriglyceridemia: 996. Mahley RW, Hui DY, Innerarity TL, Weisgraber
role of familial and acquired disorders. Metabolism. KH. Two independent lipoprotein receptors on
1983;32:209. hepatic membranes of dog, swine, and man. Apo-B,
979. Brunzell JD, Chait A. Lipoprotein metabolism. In: E and apo-E receptors. J Clin Invest. 1981;
Rifkin H, Porte Jr D, editors. Ellenberg and Rifkin’s 68:1197.
diabetes mellitus. New York: Elsevier; 1989. 997. Falko JM, Schonfeld G, Witztum JL, et al. Effects
980. Brunzell JD, Bierman EL. Chylomicronemia syn- of estrogen therapy on apolipoprotein E in type III
drome: interaction of genetic and acquired hyper- hyperlipoproteinemia. Metabolism. 1979;78:1171.
triglyceridemia. Med Clin North Am. 1982;66:455. 998. Kushwaha RS, Hazzard WR, Gagne C, et al. Type
981. Brunzell JD, Schrott HG. The interaction of familial III hyperlipoproteinemia: paradoxical hypolipi-
and secondary causes of hypertriglyceridemia: role demic response to estrogen. Ann Intern Med.
of pancreatitis. Trans Assoc Am Physicians. 1973; 1977;87:517.
86:245. 999. Thompson GR, Soutar AK, Spengel FA, et al.
982. Brunzell JD, Schrott HG, Motulsky AG, Bierman Defects of receptor-mediated low density lipopro-
EL. Myocardial infarction in the familial forms of tein catabolism in homozygous familial hypercho-
hypertriglyceridemia. Metabolism. 1976;25:313–20. lesterolemia and hypothyroidism in vivo. Proc Natl
983. Fredrickson DS, Levy RI, Lees KS. Fat transport in Acad Sci USA. 1981;78:2591.
lipoproteins – an integrated approach to mechanisms 1000. Hopkins PN, Wu LL, Schumacher MC, et al. Type
and disorders. N Engl J Med. 1967;276:34. III dyslipoproteinemia in patients heterozygous for
984. Morganroth J, Levy RI, Fredrickson DS. The bio- familial hypercholesterolemia and apolipoprotein
chemical, clinical, and genetic features of type III E2. Evidence for a gene-gene interaction.
hyperlipoproteinemia. Ann Intern Med. 1975;82:158. Arterioscler Thromb. 1991;11:1137.
985. Mahley RW, Apolipoprotein E. Cholesterol transport 1001. Hazzard WR, Warnick GR, Utermann G, Albers JJ.
protein with expanding role in cell biology. Science. Genetic transmission of isoapolipoprotein E pheno-
1988;240:622. types in a large kindred: Relationship to dysbetali-
182 5 Clinical Aspects of Gout and Associated Disease States

poproteinemia and hyperlipidemia. Metabolism. 1019. Carlson LA, Philipson B. Fish-eye disease. A new
1981;30:79. familial condition with massive corneal opacities
1002. Chait A, Albers JJ, Brunzell JD. Very low density and dyslipoproteinemia. Lancet. 1979;2:921.
lipoprotein overproduction in genetic forms of hyper- 1020. Carlson LA. Fish eye disease: a new family condi-
triglyceridaemia. Eur J Clin Invest. 1980;10:17. tion with massive corneal opacities and dyslipopro-
1003. Tall A, Granot E, Brocia R, et al. Accelerated trans- teinaemia. Eur J Clin Invest. 1982;12:41.
fer of cholesteryl esters in dyslipidemic plasma. 1021. Gjone E. Familial lecithin:cholesterol acyltrans-
Role of cholesteryl ester transfer protein. J Clin ferase deficiency: a clinical survey. Scand J Clin
Invest. 1987;79:1217. Lab Invest. 1974;33 Suppl 137:73.
1004. Wardell MR, Weisgraber KH, Havekes LM, Rall Jr 1022. Assmann G, Von Eckardstein A, Funke H.
SC. Apolipoprotein E 3-Leiden contains a Lecithin:cholesterol acyltransferase deficiency and
seven-amino acid insertion that is a tandem repeat of fish eye disease. Curr Opin Lipidol. 1991;2:110.
residues 121 to 127. J Biol Chem. 1989;264:21205. 1023. Glomset JA, Norum KR. The metabolic role of
1005. van den Maagdenberg AMJM, de Kuijff P, lecithin:cholesterol acyltransferase: perspectives
Stalenhoef AFH, et al. Apolipoprotein E 3-Leiden from pathology. Adv Lipid Res. 1973;11:1.
allele results from a partial gene duplication in exon 1024. Klein H-G, Santamarina-Fojo S, Duverger N, et al.
4. Biochem Biophys Res Commun. 1989;165:851. Fish-eye syndrome: a molecular defect in the
1006. Horie Y, Fazio S, Westerlund JR, et al. The func- lecithin:cholesterol acyltransferase (LCAT) gene
tional characteristics of a human apolipoprotein E associated with normal alpha-LCAT-specific activ-
variant (cysteine at residue 142) may explain its ity: implications for classification and prognosis.
association with dominant expression of type III J Clin Invest. 1993;92:479.
hyperlipoproteinemia. J Biol Chem. 1992;267:1962. 1025. Forte TM, Carlson LA. Electron microscopic struc-
1007. Fazio S, Horie Y, Weisgraber KH, et al. Preferential ture of serum lipoproteins from patients with fish
association of apolipoproteins E Leiden with very eye disease. Arteriosclerosis. 1984;4:130.
low density lipoproteins of human plasma. J Lipid 1026. Tybjaerg-Hansen A, Nordestgaard BG, Gerdes LU,
Res. 1993;34:447. et al. Genetic markers in the apo A1-CIII-AIV gene
1008. Ji Z-S, Fazio S, Mahley RW. Variable heparan sulfate cluster for combined hyperlipidemia, hypertriglyc-
proteoglycan binding of apolipoprotein E variants eridemia, and predisposition to atherosclerosis.
may modulate the expression of type III hyperlipo- Atherosclerosis. 1993;100:157.
proteinemia. J Biol Chem. 1994;269:13421. 1027. Moennig G, Wiebusch H, Enbergs A, et al.
1009. Weisgraber KH, Rall Jr SC, Mahley RW, et al. Detection of missense mutations in the genes for
Human apolipoprotein E. Determination of the lipoprotein lipase and hepatic triglyceride lipase in
heparin binding sites of apolipoprotein E 3. J Biol patients with dyslipidemia undergoing coronary
Chem. 1986;261:2608. angiography. Atherosclerosis. 2000;149:395.
1010. Parker F. Xanthomas and hyperlipidemias. J Am 1028. Holzl B, Kraft HG, Wiebusch H, et al. Two novel
Acad Dermatol. 1985;13:1. mutations in the lipoprotein lipase gene in a family
1011. Haber C, Kwiterovich Jr PO. Dyslipoproteinemia with marked hypertriglyceridemia in heterozygous
and xanthomatosis. Pediatr Dermatol. 1984;1:261. carriers. Potential interaction with the polymorphic
1012. Borrie P. Type III hyperlipoproteinaemia. Br Med J. marker D1S104 on chromosome 1q21-q23. J Lipid
1969;2:665. Res. 2000;41:234.
1013. Mishkel MA. Type III hyperlipoproteinaemia with 1029. Ordovas JM, Civeira F, Genest Jr J, et al. Restriction
xanthomatosis. In: Peters H, editor. Protides of the fragment length polymorphisms of the apolipopro-
biological fluids. Oxford: Pergamon; 1972. p. 283. tein A-1, C-III, A-IV gene locus. Relationships
1014. Vermeer BJ, Van Gent CM, Goslings B, Polano with lipids, apolipoproteins, and premature coro-
MK. Xanthomatosis and other clinical findings in nary artery disease. Atherosclerosis. 1991;87:75.
patients with elevated levels of very low density 1030. Rees A, Shoulders CC, Stocks J, et al. DNA poly-
lipoproteins. Br J Dermatol. 1979;100:657. morphism adjacent to human apoprotein A-1 gene:
1015. Stuyt PMJ, Van’t Laar A. Clinical features of type III relation to hypertriglyceridemia. Lancet. 1983;1:444.
hyperlipoproteinaemia. Neth J Med. 1983;26:104. 1031. Aalto-Setala K, Kontula K, Sane T, et al. DNA poly-
1016. Norum KR, Gjone E. Familial plasma morphisms of apolipoprotein A-1/C-III and insulin
lecithin:cholesterol acyltransferase deficiency. genes in familial hypertriglyceridemia and coronary
Biochemical study of a patient with a new inborn heart disease. Atherosclerosis. 1987;66:145.
error of metabolism. Scand J Clin Lab Invest. 1032. Hayden MR, Kirk H, Clark C, et al. DNA polymor-
1967;20:231. phisms in and around the apo A1-CIII genes and
1017. Gjone E, Norum KR. Familial serum-cholesterol genetic hyperlipidemias. Am J Hum Genet. 1987;
ester deficiency: clinical study of a patient with a 40:421.
new syndrome. Acta Med Scand. 1968;183:387. 1033. Rees A, Stocks J, Sharpe CR, et al. Deoxyribonucleic
1018. Torsvik H, Gjone E, Norum KR. Familial plasma acid polymorphism in the apolipoprotein A-1-C-III
cholesterol ester deficiency: clinical studies in a gene cluster. Association with hypertriglyceri-
family. Acta Med Scand. 1968;183:107. demia. J Clin Invest. 1985;76:1090.
References 183

1034. Chamberlain JC, Thorn JA, Oka K, et al. DNA 1049. Matsubara K, Matsuzawa Y, Jiao S, et al.
polymorphisms at the lipoprotein lipase gene: asso- Relationship between hypertriglyceridemia and
ciation in normal and hypertriglycerideaemic sub- uric acid production in primary gout. Metabolism.
jects. Atherosclerosis. 1989;79:85. 1989;38:698.
1035. Dammerman M, Sandkuijl LA, Halaas JL, et al. An 1050. Tinahones FJ, Collantes E, C-Soriguer FJ, et al.
apolipoprotein CIII haplotype protective against Increased VLDL levels and diminished renal
hypertriglyceridemia is specified by promoter and excretion of uric acid in hyperuricaemic-hyper-
3’ untranslated region polymorphisms. Proc Natl triglyceridaemic patients. Br J Rheumatol. 1995;
Acad Sci USA. 1993;90:4562. 34:920.
1036. Needham EW, Mattu RK, Rees A, et al. A poly- 1051. Timar O, Sester F, Levy E. Metabolic syndrome X:
morphism in the human apolipoprotein A1 pro- a review. Can J Cardiol. 2000;16:779.
moter region: a study in hypertriglyceridaemic 1052. Wassef GN. Lipoprotein (a) in android obesity and
patients. Hum Hered. 1994;44:94. NIDDM: a new member in “the metabolic syn-
1037. Barre DE, Guerra R, Verstraete R, et al. Genetic drome”. Biomed Pharmacother. 1999;53:462.
analysis of a polymorphism in the human apolipo- 1053. Miccoli R, Ceraudo AM, Manfredi SG, et al.
protein A-1 gene promoter: effect on plasma HDL Atherogenic dyslipidemia, metabolic syndrome
cholesterol levels. J Lipid Res. 1994;35:1292. and cardiovascular risk. Cardiologia. 1999;44:885.
1038. Tsai MY, Hanson NQ, Copeland KR, et al. 1054. Huth K, Burkard M, Goebel T. Dyslipoproteinemia
Determination of a T/G polymorphism at nucle- and diabetes mellitus in a metabolic syndrome.
otide 3206 of the apolipoprotein CIII gene by Fortschr Med. 1992;110:200.
amplification refractory mutation system. Clin 1055. Vuorinen-Markkola H, Yki-Jaarvinen H. Hyperuri-
Chem. 1994;40:2235. cemia and insulin resistance. J Clin Endocrinol
1039. Zeng Q, Dammerman M, Takada Y, et al. An apoli- Metab. 1994;78:25.
poprotein CIII marker associated with hypertrig- 1056. Steinmetz A, Schafer JR. Secondary disorders of
lyceridemia in Caucasians also confers increased lipid metabolism, metabolic syndrome and familial
risk in a west Japanese population. Hum Genet. combined hyperlipidemia. Wien Med Wochenschr.
1995;95:371. 1994;144:299.
1040. Zhang Q, Liu Y, Liu BW, et al. Common genetic 1057. Reaven GM. The kidney: an unwilling accomplice
variants of lipoprotein lipase and apolipoproteins in syndrome X. Am J Kidney Dis. 1997;30:928.
A1-CIII that relate to coronary artery disease: a 1058. Grundy SM. Hypertriglyceridemia, atherogenic
study in Chinese and European subjects. Mol Genet dyslipidemia and the metabolic syndrome. Am
Metab. 1998;64:177. J Cardiol. 1998;81:18B.
1041. Zhang Q, Liu B, Liu Y, et al. DNA polymorphisms 1059. Wasada T, Katsumori K, Saeki A, Iwatani M.
of apolipoprotein A1 gene in Chinese endogenous Hyperuricemia and insulin resistance. Nippon
hypertriglyceridemics. Hua His I Ko Ta Hsueh Rinsho. 1996;54:3293.
Hsueh Pao. 1997;28:233. 1060. Rathmann W, Funkhouser E, Dyer AR, Roseman
1042. Schonfeld G, George PK, Miller J, et al. JM. Relations of hyperuricemia with the various
Apolipoprotein C-II and C-III levels in hyperlipo- components of the insulin resistance syndrome in
proteinemia. Metabolism. 1979;28:1001. young black and white adults: the CARDA study.
1043. Ghiselli G, Schaefer EJ, Zech LA, et al. Increased Coronary Artery Risk Development in Young
prevalence of apolipoprotein E4 in type V hyperli- Adults. Ann Epidemiol. 1998;8:250.
poproteinemia. J Clin Invest. 1982;70:474. 1061. Dessein PH, Shipton EA, Stanwix AE, et al.
1044. Kuusi T, Taskinen M-R, Solakivi T, Kauppinen- Beneficial effects of weight loss associated with
Makelin R. Role of apolipoproteins E and C in type moderate calorie/carbohydrate restriction, and
V hyperlipoproteinemia. J Lipid Res. 1988;29:293. increased proportional intake of protein and unsatu-
1045. Hegele RA, Breslow JL. Apolipoprotein genetic rated fat on serum urate and lipoprotein levels in
variation in the assessment of atherosclerosis sus- gout: a pilot study. Ann Rheum Dis. 2000;59:539.
ceptibility. Genet Epidemiol. 1987;4:163. 1062. Gerhardt U, Grosse Huttmann M, Hohage H.
1046. Fox IH, John D, De Bruyne S, et al. Hyperuricemia Influence of hyperglycemia and hyperuricemia on
and hypertriglyceridemia: metabolic basis for the long-term transplant survival in kidney transplant
association. Metabolism. 1985;34:741. recipients. Clin Transplant. 1999;13:375.
1047. Giacomello A, Sciascio N, Quaratino CP. Relation 1063. Gravholt CH, Juul S, Naeraa RW, Hansen J.
between serum triglyceride level, serum urate concen- Morbidity in Turner syndrome. J Clin Epidemiol.
tration, and fractional urate excretion. Metabolism. 1998;51:147.
1977;46:1085. 1064. Iannello S, Cavaliere G, Ferro G, et al. Tophaceous
1048. Tinahones JF, Perez-Lindon G, C-Soriguer FJ, et al. gout in plurimetabolic syndrome. Minerva Med.
Dietary alterations in plasma very low density lipo- 1998;89:419.
protein levels modify renal excretion of urates in 1065. Wien YT, Chan CS, Liu CS. Hyperuricaemia and
hyperuricemic-hypertriglyceridemic patients. J Clin type 2 diabetes mellitus. Diabetes Nutr Metab.
Endocrinol Metab. 1997;82:1188. 1999;12:286.
184 5 Clinical Aspects of Gout and Associated Disease States

1066. Lai SW, Tan CK, Ng KC. Epidemiology of hyperg- 1084. Ruilope LM, Rodicio JL. Renal surrogates in essen-
lycemia in elderly persons. J Geront A Biol Sci tial hypertension. Clin Exp Hypertens. 1999;21:609.
Med Sci. 2000;55:M257. 1085. Nishioka K, Mikanagi K. A retrospective study of
1067. Wyngaarden JB, Kelley WN. Gout. In: Stanbury the cause of death, in Japan, of patients with gout.
JB, Wyngaarden JB, Fredrickson DS, editors. The Ryumachi. 1981;21:29.
metabolic basis of inherited disease. 4th ed. New 1086. Brand FN, McGee DL, Kannel WB, et al. Original
York: McGraw-Hill, Inc; 1978. p. 916. contributions: hyperuricemia as a risk factor of
1068. Wyngaarden JB, Kelley WN. Gout and hyperurice- coronary heart disease: the Framingham study. Am
mia. New York: Grune & Stratton; 1976. p. 32. J Epidemiol. 1985;121:11. Peterson B, Trell E.
1069. Cannon PJ, Stason WB, Demartini FE. Raised serum urate concentrations as risk factor for
Hyperuricemia in primary and renal hypertension. premature mortality in middle aged men. Br Med J.
N Engl J Med. 1966;275:457. 1983;287:7.
1070. Garrick R, Bauer GE, Evan CE. Serum uric acid in 1087. Reunanen A, Takkunen H, Knekt P, Aromaa A.
normal and hypertensive Australian subjects: from Hyperuricemia as a risk factor for cardiovascular
a continuing epidemiological survey on hyperten- mortality. Acta Med Scand. 1982;668:49.
sion commenced in 1955. Aust N Z J Med. 1972; 1088. Beard JT. Serum uric acid and coronary heart dis-
2:351. ease. Am Heart J. 1983;106:397.
1071. Breckenridge A. Hypertension and hyperuricemia. 1089. Peterson B, Trell E. Raised serum urate concentra-
Lancet. 1966;1:15. tions as a risk factor for premature mortality in
1072. Saggiani F, Pilati S, Targher G, et al. Serum uric middle aged men. Br Med J. 1983;287:7.
acid and related factors in 500 hospitalized patients. 1090. Tsutsumi Z, Yamamoto T, Takahashi S, et al. Gout
Metabolism. 1996;45:1557. and atherosclerosis. Nippon Rinsho. 1996;54:3297.
1073. Kannel WB, Wilson PW, Zhang TJ. The epidemiol- 1091. Ames BN, Cathcart R, Schwiers E, Hochstein P. Uric
ogy of impaired glucose intolerance and hyperten- acid provides an antioxidant defense in humans against
sion. Am Heart J. 1991;121:1268. oxidant- and radical-caused aging and cancer: a
1074. Zanchetti A. Hyperlipidemia in the hypertensive hypothesis. Proc Natl Acad Sci USA. 1981;78:6858.
patient. Am J Med. 1994;96:35. 1092. Cutler RG. Urate and ascorbate: their possible roles
1075. Berchtod P, Berger M, Jorgens V, et al. as antioxidants in determining longevity of mam-
Cardiovascular risk factors and HDL-cholesterol malian species. Arch Gerontol Geriatr. 1984;3:321.
levels in obesity. Int J Obes. 1981;5:1. 1093. Sevanian A, Davies KJ, Hochstein P. Conservation
1076. Dollery CT. Risk predictors, risk indicators, and of vitamin C by uric acid in blood. J Free Radic
benefit factors in hypertension. Am J Med. Biol Med. 1985;1:117.
1987;82:2. 1094. Davies KJ, Sevanian A, Muakkassah-Kelly SF,
1077. Heyden S, Borhani NO, Tyroler HA, et al. The rela- Hochstein P. Uric acid-iron ion complexes. A new
tionship of weight change to changes in blood pres- aspect of the antioxidant functions of uric acid.
sure, serum uric acid, cholesterol and glucose in the Biochem J. 1986;235:747.
treatment of hypertension. J Chronic Dis. 1985; 1095. Wayner DD, Burton GW, Ingold KU, et al. The
38:281. relative contributions of vitamin E, urate, ascorbate,
1078. Johnson RJ, Kivlighn SD, Kim YG, et al. and proteins to the total peroxyl radical-trapping
Reappraisal of the pathogenesis and consequences antioxidant activity of human blood plasma.
of hyperuricemia in hypertension, cardiovascular Biochim Biophys Acta. 1987;924:408.
disease, and renal disease. Am J Kidney Dis. 1096. Maples KR, Mason RP. Free radical metabolite of
1999;33:225. uric acid. J Biol Chem. 1988;263:1709.
1079. Messerli FH, Frohlich ED, Dreslinski GR, et al. 1097. Peden DB, Hohman R, Brown ME, et al. Uric acid
Serum uric acid in essential hypertension: an indi- is a major antioxidant in human nasal airway secre-
cator of renal vascular involvement. Ann Intern tions. Proc Natl Acad Sci USA. 1990;87:7638.
Med. 1980;93:817. 1098. Sevanian A, Davies KJ, Hochstein P. Serum urate
1080. Saito I, Saruta T, Kondo K. Serum uric acid and the as an antioxidant for ascorbic acid. Am J Clin Nutr.
renin-angiotensin system in hypertension. J Am 1991;54(suppl):1129S.
Geriatr Soc. 1978;26:241. 1099. Becker BF. Towards the physiological function of
1081. Prebis JW, Gruskin AB, Polinsky MS. Uric acid in uric acid. Free Radic Biol Med. 1993;14:615.
childhood essential hypertension. J Pediatr. 1100. Leyton ME, Wood JG, Yan ZY, Forster J. Ischemia/
1981;98:702. reperfusion alters uric acid and ascorbic acid levels
1082. Simon NM, Smucker JE, O’Connor Jr VJ. in liver. J Surg Res. 1996;64:1.
Differential uric acid excretion in essential and 1101. Sahnown Z, Jamoussi K, Zeghal KM. Free radicals
renal hypertension. Circulation. 1969;39:121. and antioxidants: human physiology, pathology and
1083. Donskov AS, Balkarov IM, Fadina ZM, et al. Urate therapeutic aspects. Therapie. 1997;52:251.
affection of kidneys and metabolic disturbances in 1102. Rosell M, Regnstrom J, Kallner A, Hellenius ML.
hypertensive patients. Ter Arkh. 1999;71:53. Serum urate determines antioxidant capacity in
References 185

middle-aged men – a controlled, randomized diet 1112. Schwartz CJ, Valente AJ, Sprague EA. The patho-
and exercise intervention study. J Intern Med. genesis of atherosclerosis: an overview. Clin
1999;246:219. Cardiol. 1991;14(suppl):11.
1103. Skinner KA, White CR, Patel R, et al. Nitrosation of 1113. Cathcart MK, Morel DW, Chisholm III GM.
uric acid by peroxynitrite Formation of a vasoactive Monocytes and neutrophils oxidize low density
nitric oxide donor. J Biol Chem. 1998;273:24491. lipoprotein making it cytotoxic. J Leukoc Biol.
1104. Holvoet P. Endothelial dysfunction, oxidation of 1985;38:341.
low-density lipoprotein, and cardiovascular dis- 1114. Palinski W, Rosenfeld ME, Yla-Herttuala S, et al.
ease. Ther Apher. 1999;3:287. Low density lipoprotein undergoes oxidative
1105. Navab M, Berliner JA, Watson AD, et al. The yin modification in vivo. Proc Natl Acad Sci USA.
and yang of oxidation in the development of the 1989;86:1372.
fatty streak: a review based on the 1994 George 1115. Salonen T, Yla-Herttuala S, Yamamoto R, et al.
Lyman Duff Memorial Lecture. Arterioscler Autoantibody against oxidized LDL and progres-
Thromb Vasc Biol. 1996;16:831. sion of carotid atherosclerosis. Lancet.
1106. Parhami F, Fang ZT, Fogelman AM, et al. Minimally 1992;339:883.
modified low density lipoprotein-induced 1116. Holvoet P, Perez G, Zhao Z, et al. Malondialdehyde-
inflammatory responses in endothelial cells are modified low density lipoproteins in patients with
mediated by cyclic adenosine monophosphate. atherosclerotic disease. J Clin Invest. 1995;
J Clin Invest. 1993;92:471. 95:2611.
1107. Henriksen T, Mahoney EM, Steinberg D. Enhanced 1117. Abuja PM. Ascorbate prevents prooxidant effects
macrophage degradation of low density lipoprotein of urate in oxidation of human low density lipopro-
previously incubated with cultured endothelial tein. FEBS Lett. 1999;446:305.
cells: recognition by receptors for acylated low 1118. Schlotte V, Sevanian A, Hochstein P, Weithmann
density lipoproteins. Proc Natl Acad Sci USA. KU. Effect of uric acid and chemical analogues on
1981;78:6499. oxidation of human low density lipoprotein in vitro.
1108. Kaplan M, Aviram M. Oxidized low density Free Radic Biol Med. 1998;25:839.
lipoprotein: atherogenic and proinflammatory 1119. Nieto FJ, Iribarren C, Gross MD, et al. Uric acid
characteristics during macrophage foam cell for- serum antioxidant capacity: a reaction to athero-
mation. An inhibitory role for nutritional antioxi- sclerosis? Atherosclerosis. 2000;148:131.
dants and serum paraoxonase. Clin Chem Lab Med. 1120. Hasegawa T, Kuroda M. A new role of uric acid as
1999;37:777. an antioxidant in human plasma. Rinsho Byori.
1109. Quinn MT, Parthasarathy S, Steinberg D. 1989;37:1020.
Lysophosphatidylcholine: a chemotactic factor for 1121. Boda D. Role of hyperuricemia in critically ill
human monocytes and its potential role in athero- patients especially newborns. Acta Paediatr Hung.
genesis. Proc Natl Acad Sci USA. 1988;85:2805. 1984;25:23.
1110. Frostegard J, Haegerstrand A, Gidlund M, Nilsson 1122. Jabs CM, Sigurdsson GH, Neglen P. Plasma levels
J. Biologically modified LDL increases the adhe- of high-energy compounds compared with severity
sive properties of endothelial cells. Atherosclerosis. of illness in critically ill patients in the intensive
1991;90:119. care unit. Surgery. 1998;124:65.
1111. Quinn MT, Parthasarathy S, Fong LG, Steinberg D. 1123. MacKinnon KL, Molnar Z, Lowe D, et al. Measures
Oxidatively modified low density lipoproteins: a of total free radical activity in critically ill patients.
potential role in recruitment and retention of mono- Clin Biochem. 1999;32:263.
cyte/macrophages during atherogenesis. Proc Natl
Acad Sci USA. 1987;84:2995.
Diagnostic Procedures
in the Management of Gout 6

Introduction of 6.45 mg %. The monosodium salt of uric acid


is slightly more soluble in water than the free
The objective of this chapter is to provide the acid, and the free acid has two ionization con-
reader with the methodologies used to investigate stants (pKa1 = 5.75 and pKa2 = 10.3). As the pH
abnormalities in purine synthesis and metabo- approaches the second pKa, the molecule is more
lism. As one might suspect, there are many pub- soluble, and therefore, alkalinity enhances the
lished variations of specific methods, and no solubility of uric acid. Solubility in plasma is
attempt has been made to include all these differ- somewhat greater than in water, and supersatura-
ences. Methodologies that have become some- tion occurs at 7.0 mg %. As calculated from
what universal in their usage have been selected Allen’s data by Loeb [1, 2], uric acid has a solu-
for inclusion. Further, an attempt has been made bility of 6.8 mg % at 37 °C in the presence of
to include only those methodologies that have sodium ions (140 mM). On the basis of these cal-
primary significance to the clinician-investigator culations, supersaturation of the plasma occurs at
who is faced with the task of the diagnosis and concentrations of uric acid above 7.0 mg %.
management of patients who may have abnor- Thus, the physicochemical definition of hyperu-
malities in purine metabolism. ricemia is a plasma concentration above 7.0 mg
%. There are data in the literature that higher
concentrations of monosodium urate in plasma
Hyperuricemia can be attained in supersaturated solutions [3–5],
but such supersaturated solutions are usually
The chemical properties of the uric acid of great- unstable and over time monosodium urate pre-
est significance to the production of disease are cipitates. The factor or factors governing the
its capacity to form crystals and its relative insol- enhanced solubility of urate in plasma remain
ubility in nonalkaline solutions. Crystal forma- incompletely understood, even though some
tion leads to the initiation of an acute inflammatory plasma macromolecules including albumin, low-
response in synovial joints and ultimately an density beta-lipoprotein, beta-2 macroglobulin,
acute arthritis. The insolubility of uric acid often and an alpha-1, alpha-2-globulin have been pro-
leads to the formation of kidney stones in the host posed and their affinity for urate characterized in
who is excreting large quantities of uric acid and some cases [6–12]. In summary, there are macro-
also may result in the deposition of urates in tis- molecules that bind urate in plasma; however, the
sue sites such as tophaceous deposits. In water, a binding of urate to plasma proteins at 37 °C is
saturated solution of uric acid at 37 °C is likely to be of little physiological or clinical
0.38 mM, which corresponds to a concentration significance.

D.S. Newcombe, Gout, 187


DOI 10.1007/978-1-4471-4264-5_6, © Springer-Verlag London 2013
188 6 Diagnostic Procedures in the Management of Gout

Synovial Fluid Analysis with soap and water and subsequently swabbed
with iodine and alcohol. One should determine
Arthrocentesis and synovial fluid analysis are that the patient is not sensitive to iodine before
procedures essential for making an absolute diag- treating the skin with iodine. Alcohol cleansing
nosis of either a septic joint or a crystal-induced alone can be used in patients with iodine sensitiv-
arthritis. In addition, synovial fluid culture and ity. The skin over the site of aspiration may be
sensitivity must be done in cases where septic anesthetized either with Xylocaine or ethyl chlo-
arthritis is suspected. Except for synovial fluid ride spray. Deep infiltration with Xylocaine
analysis including culture, there is no reliable should be avoided to prevent injecting anesthetic
means of differentiating septic arthritis from into the synovial fluid and thereby altering the
crystal-induced arthritis. As a general rule, it is results of the subsequent synovial fluid analysis.
reasonable to culture all synovial fluids recovered Small-sized needles may be used for Xylocaine
from patients with an acute arthritis to exclude infiltration (25 gauge), but 16- to 18-gauge nee-
infection. dles should be used for the aspiration of synovial
Any synovial fluid analysis that has a white fluid. The syringe should be wet with sodium
blood cell count of greater than 100,000 cells/ heparin to avoid clotting of the aspirated fluid.
mm3 represents a possible septic joint until proven Although the author prefers to use heparin as an
otherwise by culture results. In contrast, a syn- anticoagulant, EDTA (ethylenediaminetetraa-
ovial fluid count of less than 50,000 white blood cetic acid) or sodium citrate solutions can also be
cells/mm3, although less suspicious of infection, used. Anticoagulants to avoid are powdered
does not exclude a septic joint from consider- oxalate and lithium heparin, since these prepara-
ation. Immunocompromised hosts, especially tions form crystals that lend confusion to the
those undergoing treatment to avoid organ rejec- analysis [13–15]. Talc from surgical gloves may
tion or to suppress systemic connective tissue also give rise to Maltese crosses in synovial fluid
diseases, are often unable to mount a sufficient leading to confusion when the fluid is examined
neutrophil response to an infection or crystal- for true crystals [16].
induced inflammatory response. The identification For diagnostic aspirations, only 1–5 ml of
of monosodium urate crystals in synovial fluid fluid is required. If one elects to perform a com-
with accompanying findings supportive of gout plete aspiration for relief of tension in the joint, a
provides the clinician with an absolute criterion hemostat attached to the hub of the needle per-
for the diagnosis of gout. Nonetheless, such crys- mits an easy exchange of syringes should that be
tals may also be associated with an infectious necessary. In knee aspirations, fluid can be pushed
process or calcium pyrophosphate dihydrate toward the needle by compressing the side oppo-
(CPPD) crystals. site from the site of needle entry. The knee may
be aspirated from a lateral or medial site.
Loculated accumulations of pus, especially in
Arthrocentesis Techniques gonococcal infections, may be encountered, and
both lateral and medial approaches to the knee
The technique for joint aspiration is straightfor- may be necessary in such settings to enter these
ward when the aspiration of a relatively large loculated accumulations of pus. The wrist is usu-
joint (ankle, knee, wrist) is the objective. Smaller ally aspirated from its dorsal surface at the site of
joints (finger) and hips or shoulders may be more the radiocarpal joint. After identifying the joint
difficult to aspirate. If one lacks experience with space, the ankle may be aspirated anterior to the
joint aspirations, the task should be assigned to malleolus. The shoulder may be aspirated using
an experienced rheumatologist or orthopedist. either an anterior or posterior approach. The ante-
Initially, the aspiration site should be cleansed rior approach is done by inserting the needle just
Synovial Fluid Analysis 189

lateral to the coracoid process with the needle white blood cell count decreases by about 1,000–
pointing toward the glenohumeral joint. The pos- 4,000 cells/mm3, and after 24 h, the number of
terior approach to the shoulder joint is accom- CPPD crystals decreases significantly and they
plished by inserting the needle about 1 in. below become more difficult to recognize. Artifactual
the posterolateral boundary of the acromion pro- crystals also form in the synovial fluid over time
cess. Finger joints are best aspirated using a dor- [21]. Finally, although monosodium urate crystals
solateral approach to the joint. Several excellent remain present in synovial fluid samples for
books and CDs are available to guide the clini- 8 weeks, they are fewer in number, less birefrin-
cian in joint aspirations, and these informative gent, and smaller in size than in the original sam-
sources may be consulted for further information ple. Another comparable difficulty in the
on joint aspirations [17–19]. collection and analysis of synovial fluid is the
If one uses sterile techniques during joint aspi- delay in processing specimens for culture. For
rations, there are rarely any complications from this reason, it is often useful to use blood culture
this procedure. The most common complications, bottles at the bedside for the immediate culture of
when they occur, are pain, hemarthroses, and specimens. In both adults and children, cultures
infections. With respect to pain, adequate local may be positive in only 50–70 % of cases, and
anesthesia and avoidance of pain-sensitive joint processing synovial fluid samples for culture at
structures are sufficient to prevent significant dis- the bedside may increase the chances of recover-
comfort during the procedure. Occasionally, ing an organism [22–25].
patients may faint during or after the procedure,
so it is essential to have somebody stay with the
patient when he or she arises after the completion Synovial Fluid Culture
of the joint tap. If the patient is known to have a
significant bleeding diathesis, it is necessary to In acute synovitis, where gout and septic arthritis
perform arthrocentesis after treating the coagula- are the primary clinical considerations, four syn-
tion disorder. Infections relating to arthrocentesis ovial fluid diagnostic tests are often required: cul-
are rare. It is inappropriate to aspirate a joint at a ture and sensitivity of the fluid, synovial fluid
site where cutaneous infections or skin lesions smears for Gram stain and white blood cell dif-
exist, because infectious agents from such lesions ferential counts, total white blood cell count, and
may be carried into the synovial cavity during wet preparations for crystal analyses. In both
aspiration and result in a septic joint. adults and children, cultures of synovial fluid
A recent randomized controlled trial of dry may not be positive in 100 % of cases, and pro-
taps indicated that failed arthrocentesis was most cessing specimens at the bedside may increase
frequently the result of the presence of extremely the chances of recovering an organism. In fact, in
viscous synovial fluid, adipose tissue, or lipoma one recent publication, 70 % of children with
arborescens [20]. Lipoma arborescens (thickened clinical findings indicative of septic arthritis had
synovium infiltrated with fat) and fluid viscosity negative synovial fluid cultures [26]. Certain
may also be a problem with dry taps in chronic clinical settings increase the risk of infection such
synovial effusions. The observations in these as the elderly patient or a child, the presence of a
studies suggest that a lateral approach for knee prosthetic joint, prior arthrocentesis, prior corti-
aspirations may be more rewarding than a medial costeroid injection, and certain medical condi-
approach. There is also objective evidence that a tions. The latter include diabetes mellitus,
delay in synovial fluid analysis may alter the rheumatoid arthritis, systemic lupus erythemato-
findings even if the fluid is stored at 4 °C prior to sus, sickle cell disease, chronic liver disease,
analysis [21]. Even if the delay is a relatively intravenous drug abuse, and the administration of
short period of time (5–6 h), the synovial fluid immunosuppressive drugs.
190 6 Diagnostic Procedures in the Management of Gout

Since a delay in processing synovial fluid in joint functions caused by the infectious
specimens for culture decreases the yield of posi- process.
tive cultures, the use of aerobic and anaerobic
blood culture bottles at the bedside for the imme-
diate culture of specimens is a good practice. N. Synovial Fluid White Blood Cell Counts
gonorrhoeae are especially difficult to culture
from synovial fluid, and most physicians elect to Synovial fluid white blood cell counts of 2,000
plant aspirates from those suspected of infection cells/mm3 or less are usually considered
with this organism on chocolate agar or Thayer- noninflammatory, but total white blood cell counts
Martin culture media at the bedside. Once the in this range are observed in some systemic
specimen is plated on culture media, it should be articular disorders such as systemic lupus erythe-
grown in 10 % CO2 for at least 10 days at 37 °C. matosus, enteric arthritis, systemic sclerosis,
Joint infections due to anaerobic bacteria are osteoarthritis, and rarely crystal-induced synovi-
exceedingly unusual, but certain clinical settings tis [27–29]. Other articular diseases may also
increase the chance of such infections. Penetrating present with noninflammatory total white blood
trauma, surgical procedures, prosthetic joints, cell counts in their synovial fluid including hyper-
and a compromised host are the most common trophic pulmonary osteoarthropathy, sickle cell
predisposing risk factors for anaerobic infections. disease, hypothyroidism, amyloidosis, neuro-
When this class of bacteria is suspected, culture pathic arthropathy, and hyperlipoproteinemia. It
media appropriate for anaerobic organisms is also imperative to remember that individuals
should be used. who are immunocompromised may express low
total white blood cell counts when infection or
crystal-induced arthritis is the underlying cause
Synovial Fluid Gram Stain of their joint inflammation.
At the other extreme, synovial fluid total white
Gram stains of the synovial fluid may provide an blood cell counts in the range of 50,000–100,000
early clue to the underlying infectious agent and cells/mm3 are the usual findings in septic arthritis
permit the institution of antibiotic therapy before or crystal-induced arthritis [30]. However, this
culture results are available. Gram stains usually degree of synovial fluid leukocytosis may also be
reveal an organism in 50–70 % of cases, and they observed in rheumatoid arthritis and other non-
can easily distinguish Gram-positive from Gram- septic arthritis. Nonetheless, data from a series of
negative bacteria. Unfortunately, Gram stains synovial fluid tests confirm the sensitivity and
cannot distinguish staphylococcal from strepto- specificity of synovial fluid white blood cell
coccal infections and cannot, by themselves, counts and the percentage of neutrophils as keys
determine whether an infectious arthritis is com- to discriminating between inflammatory and
plicated by a crystal-induced arthritis. noninflammatory diseases of the joints [31].
It is essential to clean all glass slides and Thus, although synovial fluid levels of protein,
cover slips prior to their use to avoid visualizing glucose, lactic acid dehydrogenase, and other
artifacts in the Gram stained specimens or in wet chemical analyses of the fluid may be helpful, the
preparations of synovial fluid for crystal exami- total white blood cell count, differential cell
nation. The application of acetone to slides and count, and culture are the most critical tests for
cover slips and subsequent drying in a dust-free characterizing infectious and crystal-induced
environment removes most contaminating mate- arthritides.
rial. Initiating antibiotic therapy on the basis of Two critical issues need to be recognized
Gram stain results serves to forestall the amount regarding white blood cell counts. First, the
of joint destruction and its resultant alterations proper diluent for synovial fluid cell counts is
Synovial Fluid Analysis 191

normal saline and not acetic acid, the diluent used used. Centrifuging the synovial fluid specimen at
for peripheral blood cell counts. If acetic acid is 3,000 rpm for 10 min may form such a cell pellet.
used as the diluent for synovial fluid, clots of After decanting the supernatant, the cell pellet
mucin and other components of the fluid occur may be resuspended in several drops of the super-
and white blood cells are trapped in this precipi- nate. The cell concentrate can then be used both
tated material. When such acetic acid-treated for preparing smears for differential counts and
fluids are subjected to automated cell counters, for crystal examination using a wet cell
cell clumping and other debris dramatically preparation.
reduce total white blood cell counts. The author In most septic joints, the synovial fluid dif-
has experienced this laboratory error a number of ferential cell count reveals more than 95 %
times, especially when inexperienced night staff neutrophils. Crystal-induced arthritis usually
are operating the laboratory. has a slightly lower percentage of neutrophils.
Second, the color and turbidity of a fluid often As a general rule, infected synovial fluids show
suggests an underlying infection but is not always neutrophil percentages of 75 or more, and
a foolproof sign. Certainly, purulent-appearing crystal-induced inflammatory fluids have 50 % or
fluid with a green, yellow, or white color is most more neutrophils in their differential count.
likely to indicate an infection with the variation
in color imparted by the type of infecting organ-
ism. Several cases of acute gouty arthritis have Synovial Fluid Wet Preparations
been reported with thick, white synovial fluid on for Crystal Identification
aspiration [32, 33]. Such milky effusions appear
to be most common in alcohol abusers, and such The most significant test in the evaluation of an
effusions have been found to result from massive acute inflammatory process in a joint other than
accumulations of uric acid crystals. The large synovial fluid Gram stains and cultures is the
number of crystals interferes with either the accu- search for crystals by light and compensated
mulation of leukocytes or their accurate count- polarized microscopy [34, 35]. Specimens should
ing. In fact, one reported patient with such a be examined promptly after the wet preparation
milky effusion had a total white blood cell count is prepared, and the need for a careful and tech-
of 6,750 WBC/mm3 with 91 % neutrophils. nologically sound examination of such prepara-
Interestingly, not all chalky-white synovial fluids tions cannot be overemphasized [36–38].
represent purulent effusions or crystal-induced To prepare a wet preparation, two drops of
inflammation. Lipid-laden synovial fluids are synovial fluid are placed on a clean glass slide
orange-white in color, and large numbers of rice and covered with a clean cover slip. The edges of
bodies in synovial fluid may mimic pus. Lipid- this wet preparation are then sealed with clear
laden fluids are usually observed in patients with nail polish or Permount. Sealing prevents the dry-
chronic, long-standing effusions. Thus, not all ing of the specimen and the movement of the
purulent-appearing fluids represent infection. contents to be examined. Wet preparations from
pelleted cells and repeated synovial fluid analy-
ses may be necessary for the identification of
Synovial Fluid Differential Cell Count crystals [39]. Light microscopy appears to iden-
tify calcium pyrophosphate dihydrate (CPPD)
Using clean cover slips and glass slides, a thin crystals more readily than compensated polarized
smear of synovial fluid is made to examine the microscopy since only 20 % of CPPD crystals are
differential white cell count after staining the birefringent [40].
smear with Wright’s stain. If the total cell count Crystals can be visualized by light, phase, and
is low, a concentrated sediment of fluid can be compensated polarized microscopy, but the latter
192 6 Diagnostic Procedures in the Management of Gout

method is the most definitive way to identify


monosodium urate crystals. In addition to the
characteristics of crystals seen by compensated
polarized microscopy, crystal morphology is
helpful in distinguishing CPPD from MSU crys-
tals. MSU crystals are linear and needle shaped
with pointed ends, whereas CPPD crystals are
linear and rhomboid in shape with blunt ends.
These crystals may be found both in intracellular
and extracellular locations. As one might expect,
uricase dissolves MSU crystals but not CPPD
crystals. Recently, a commercial staining method
(Diff Quik) has been useful for the identification
of MSU and CPPD crystals when preparations
are examined both by compensated polarized
microscopy and transmitted light microscopy
[41]. In the absence of a first-order red plate
compensator, birefringent material can be visual-
ized as white colored objects on a black micro-
scopic field when the second polarizing lens is
rotated 90° from the first polarizer and the sam-
ple for examination is placed between the two
polarizers. Such dark field examinations are use-
ful for a preliminary search for birefringent mate-
rial, but the absence of the color changes in Fig. 6.1 Monosodium urate crystals visualized using
crystals seen with a first-order red compensator polarized light microscopy. Crystals are needle shaped,
strongly birefringent, and have a negative sign of birefrin-
in place makes a positive identification of the gence, appearing yellow when parallel to the axis of the
crystal type impossible. With a first-order red red plate, and blue when perpendicular. The upper panel
compensator in place and a rose-colored back- shows crystals under plane polarized light. The lower
ground, MSU crystals have a bright yellow color panel shows crystals under plane polarized light using a
red plate to identify the sign of birefringence
when the crystals are parallel to the axis of the
line of slow vibration of the compensator and
blue when perpendicular to the line of slow viewing many synovial fluid specimens. In addi-
vibration. On the basis of these findings, MSU tion to MSU and CPPD crystals, hydroxyapatite,
crystals have strong negative birefringence. In calcium hydrogen phosphate dihydrate, calcium
contrast, CPPD crystals are blue when they are oxalate, cholesterol, lipid inclusions, and immu-
parallel to the axis of the line of slow vibration of noglobulin cryoprecipitates may occur in various
the compensator and yellow when they are at forms of synovitis and may be seen by compen-
right angles to this axis. CPPD crystals show sated polarized microscopy as birefringent objects
weak positive birefringence and, in many cases, [42–49]. Further, many artifacts are also birefrin-
may show no birefringence (Figs. 6.1 and 6.2). gent and lead to confusion in the interpretation of
Since other birefringent crystals and refractile synovial fluid specimens. The most common
artifacts can be observed using compensated refractile artifacts include the anticoagulants, lith-
polarized microscopy, there is no substitution for ium heparin and calcium oxalate, talc from surgi-
training with someone who is experienced in crys- cal gloves, and corticosteroid crystals [14, 15, 50,
tal identification and for increasing one’s skills by 51]. The latter crystals may remain in a joint for
Synovial Fluid Analysis 193

Fig. 6.2 Calcium pyrophosphate dehydrate crystals. on the right is under polarized light alone, and the two
Using polarized light microscopy. These crystals are usu- panels on the left are seen with polarized light and a red
ally rhomboid, weakly birefringent, and have a positive plate
sign of birefringence, opposite to urate crystals. The panel

prolonged periods of time (months) or be intro- emphasis with regard to crystal-induced disorders.
duced into the joint by withdrawing synovial It is now well established that asymptomatic gouty
fluid with a needle that has been used for thera- patients may have MSU or CPPD crystals in
peutic steroid injections. Less common refractile noninflamed joints during the intercritical phase
components found in the synovial fluid include of their disease. MSU crystals in such joints are
debris from methyl methacrylate or polyethylene most frequently recovered from the knee or first
used for prosthetic joint replacement, nail polish metatarsophalangeal joint and may also be recov-
used to seal wet preparations, fibers from lens ered from joints that have never been the site of
cleaning paper, and crystals contained in immer- an acute episode of gout. Such crystals are more
sion oil used for microscopy [52]. likely to be recovered from noninflamed joints of
those patients who have not been treated with
hypouricemic drugs and who maintain a degree
Additional Clinical Data Concerning of hyperuricemia [53–55]. MSU crystals have
Synovial Fluid also been identified in the noninflamed joints of
patients with renal failure and hyperuricemia [43,
Although the foregoing discussion details the role 49]. Thus, the presence of MSU crystals in syn-
of the clinical laboratory in the use of synovianal- ovial fluid from a noninflamed joint does not
ysis for the diagnosis of acute monarticular arthri- make an absolute diagnosis of intercritical gout
tis, there are several additional facets that need [56]. It is also worth reemphasizing the fact that
194 6 Diagnostic Procedures in the Management of Gout

MSU crystals may be observed in the presence of their usefulness [72–74]. Probably the most
a septic joint as well as in the presence of other promising technique for excluding infection
crystals [57, 58]. resides in the use of polymerase chain reactions
Gout rarely complicates systemic connective and DNA amplification of synovial fluid samples
tissue disorders, but it has been reported in to identify and characterize infectious organisms
patients with systemic sclerosis and in both [75–81]. However, at the present time, none of
females and males with systemic lupus erythe- these techniques including those based on bacte-
matosus [59–65]. The coexistence of gout and rial DNA provides foolproof characterization of
systemic connective tissue diseases may lead to specific organisms. Nonetheless, as more becomes
an erroneous diagnosis if a synovial fluid analysis known about the molecular structure of various
is not performed and a search for crystals under- bacterial species, bacterial DNA analyses of syn-
taken. Errors in diagnosis may be particularly ovial fluid aspirates may provide a rapid, specific,
prominent when immunosuppressive agents are and sensitive means of excluding bacterial
used in the treatment of systemic connective tis- arthritides.
sue diseases and serve to suppress the acute syno- In summary, aspiration of synovial fluid and
vitis associated with gout. In cases where gout its subsequent culture and analysis represents on
complicates lupus, hyperuricemia, kidney dis- the most useful diagnostic tests for distinguishing
ease, and diuretic use may provide clues to avoid various articular diseases. When the diagnosis of
misinterpreting the synovitis as the arthritis of a swollen joint is in question, clinicians should
SLE [64]. not hesitate to tap the joint and examine the con-
Until recently, there was a commonly held tents of the aspirate. Although general rules
belief that gout was unusual in blacks; however, always have significant exceptions, a synovial
recent surveys have disposed of this notion fluid white blood cell count between 50,000 and
[66–70]. These studies document that gout is not 100,000 cells/mm3 with 90 % or more neutrophils
that unusual in blacks and that many associated in its differential cell count often indicates either
factors such as hypertension, diuretic use, renal the presence of an infection or a crystal-induced
disease, and alcohol abuse are observed as comor- arthritis. Bedside culturing of synovial fluid aspi-
bid conditions in blacks with gout. rates increases the likelihood of recovering the
Finally, there are ongoing attempts to develop offending organism, and infection must always
sensitive, rapid, and specific tests for the diagno- be a consideration when the arthritis is associated
sis of bacterial arthritis. Such investigations have either with another disease or a host prone to
also been linked to hypotheses suggesting that infectious complications. A paucity of white
bacteria may have a role in certain systemic blood cells in a synovial specimen does not
arthritides such as rheumatoid arthritis and the exclude the diagnosis of gout [82]. Careful prep-
spondyloarthropathies. These tests have primar- aration of slides for crystal analysis and search-
ily been aimed at discriminating between the ing a number of microscopic fields by
components of the inflammatory responses compensated polarized microscopy usually docu-
observed in septic arthritis versus those in crystal- ments the presence of crystals when they are
induced arthritis or in defining specific bacterial present.
products not observed in crystal-induced arthri-
tis. Measurements of cytokines and acute phase
reactants in the synovial fluid have not been able
to define specific differences between septic References
arthritides and crystal-induced arthritis [71].
Although extensive investigations of synovial 1. Allen DJ, Milosovich G, Mattocks AM. Inhibition of
monosodium urate crystal growth. Arthritis Rheum.
fluid lactic acid levels have been reported, the
1965;8:1123.
inability of these tests to differentiate nonseptic 2. Loeb JN. The influence of temperature on the solubility
joint disorders from bacterial arthritides limits of monosodium urate. Arthritis Rheum. 1972;15:189.
References 195

3. Seegmiller JE. The acute attack of gouty arthritis. 23. Fink CW, Nelson JD. Septic arthritis and osteomyeli-
Arthritis Rheum. 1965;8:714. tis in children. Clin Rheum Dis. 1986;12:423.
4. Gold GL, Fritz RD. Hyperuricemia associated with 24. Goldenberg DL, Reed JI. Bacterial arthritis. N Engl
the treatment of leukemia. Ann Intern Med. 1957; J Med. 1985;312:764.
62:428. 25. Goldenberg DL. Infectious arthritis complicating
5. Kjellstrand CM, Campbell III DC, von Hartitizseh B, rheumatoid arthritis and other chronic rheumatic dis-
et al. Hyperuricemic acute renal failure. Arch Intern orders. Arthritis Rheum. 1989;32:496.
Med. 1974;133:349. 26. Lyon RM, Evanich JD. Culture-negative septic arthri-
6. Alvsaker JO. Uric acid in human plasma. V. Isolation tis in children. J Pediatr Orthop. 1999;19:655.
and identification of plasma proteins interacting with 27. Pascual E. Persistence of monosodium urate crystals
urate. Scand J Clin Lab Invest. 1966;18:228. and low-grade inflammation in the synovial fluid of
7. Alvsaker JO. Uric in human plasma. III. Investigations patients with untreated gout. Arthritis Rheum. 1991;
on the interaction between the urate ion and human 34:141.
albumin. Scand J Clin Lab Invest. 1965;17:467. 28. Louthrenoo W, Sieck M, Clayburne G, et al. Supravital
8. Alvsaker JO. Uric acid in human plasma. IV. staining of cells in non-inflammatory synovial fluids:
Investigations on the interactions between urate and analysis of the effect of crystals on cell populations.
the macromolecular fraction in plasma from healthy J Rheumatol. 1991;18:409.
individuals and patients with diseases associated with 29. de Ceulaer K, Forbes M, Roper D, Serjeant GR. Non-
hyperuricemia. Scand J Clin Lab Invest. 1965; gouty arthritis in sickle cell disease: report of 37 con-
17:476. secutive cases. Ann Rheum Dis. 1984;43:599.
9. Aakesson I, Aakesson JO. The urate-binding alpha-1- 30. Krey P, Bailen D. Synovial fluid leukocytosis: a study
alpha-2-globulin. Isolation and characterization of the of extremes. Am J Med. 1979;67:436.
protein from human plasma. Eur J Clin Invest. 1971; 31. Shmerling RH, Delbanco TL, Tosteson AN, Trentham
1:281. DE. Synovial fluid tests. What should be ordered?
10. Sletten K, Aakesson I, Alvsaker JO. Presence of orni- JAMA. 1990;264:1009.
thine in the urate-binding alpha-1-alpha-2-globulin. 32. Horowitz MD, Abbey L, Sirota DK, Spiera H.
Nat New Biol. 1971;231:118. Intraarticular noninflammatory free urate suspension
11. Katz WA, Ehrlich GE. The solubility of monosodium (urate milk) in 3 patients with painful joints.
urate in serum and connective tissue fractions. J Rheumatol. 1996;17:712.
Arthritis Rheum. 1968;11:492. 33. Fam AG, Reis MD, Szalai JP. Acute gouty synovitis
12. Skeikh MI, Moller JV. Binding of urate to proteins of associated with “urate” milk. J Rheumatol. 1997;
human and rabbit plasma. Biochim Biophys Acta. 25:2285.
1968;158:456. 34. Pascual E, Tovar J, Ruiz MT. The ordinary light
13. Schumacher HR. Intracellular crystals in synovial microscope: an appropriate tool for the provisional
fluid anticoagulated with oxalate. N Engl J Med. detection and identification of crystals in synovial
1966;274:1372. fluid. Ann Rheum Dis. 1989;48:983.
14. Gatter RA, McCarty DJ. Synovianalysis, a rapid clini- 35. Gordon C, Swann A, Dieppe P. Detection of crystals
cal diagnostic procedure. Rheumatism. 1964;20:2. in synovial fluids by light microscopy: sensitivity and
15. Tanphaechitr K, Spilberg I, Hahn B. Lithium heparin reliability. Ann Rheum Dis. 1989;48:737.
crystals simulating calcium pyrophosphate dihydrate 36. Dieppe P, Swann A. Identification of crystals in syn-
crystals in synovial fluid. Arthritis Rheum. 1976;19:966. ovial fluid. Ann Rheum Dis. 1999;38:201.
16. Gatter RA. A practical handbook of joint fluid analy- 37. Segal JB, Albert D. Diagnosis of crystal-induced
sis. Philadelphia: Lea and Febiger; 1984. p. 43. arthritis by synovial fluid examination for crystals:
17. Steinbrocker O, Neustadt D. Aspiration and injection lessons from an important test. Arthritis Care Res.
therapy in arthritis and musculoskeletal diseases. 1999;12:376.
Hagerstown: Harper and Row; 1972. 38. Schlesinger N, Baker DG, Schumacher HR. How well
18. Moskowitz RW. Clinical rheumatology: a problem- have diagnostic tests and therapies for gout been eval-
oriented approach. Philadelphia: Lea & Febiger; 1975. uated? Curr Opin Rheumatol. 1999;11:441.
19. Moore GF. Arthrocentesis [computer file]/Moore, 39. Leader R. Repeated synovial fluid analysis may be
Wigden, Tape. St. Louis: Mosby; 1999. needed to find crystals in gout. Clin Rheumatol.
20. Roberts WN, Hayes CW, Breitbach SA, Owen Jr DS. 1998;17:549.
Dry taps and what to do about them: a pictorial essay 40. Ivorra J, Rosas J, Pascual E. Most calcium pyrophos-
on failed arthrocentesis of the knee. Am J Med. phate crystals appear as non-birefringent. Ann Rheum
1996;100:1. Dis. 1999;58:582.
21. Kerolus G, Clayburne G, Schumacher Jr HR. Is it 41. Salvi E, Manganelli S, Catenaccio M, et al. Diff Quik
mandatory to examine synovial fluids promptly after staining method for detection and identification of
arthrocentesis? Arthritis Rheum. 1989;32:271. monosodium urate and calcium pyrophosphate crys-
22. Morrissy RT. Bone and joint infection in the neonate. tals in synovial fluids. Ann Rheum Dis. 2001;
Pediatr Ann. 1989;18:33. 60:194.
196 6 Diagnostic Procedures in the Management of Gout

42. Schumacher HR, Somlyo AP, Tse RL, Maurer K. temic lupus erythematosus. Ann Rheum Dis.
Apatite crystals associated with arthritis. Ann Intern 1984;43:498.
Med. 1977;87:411. 61. McMillen MA, Cunningham ME, Schoen R, Fikrig E.
43. Hoffman GS, Schumacher HR, Paul H, et al. Calcium Gout in patients with systemic lupus erythematosus.
oxalate microcrystalline-associated arthritis in end- Br J Rheumatol. 1994;33:595.
stage renal disease. Ann Intern Med. 1982;97:38. 62. Veerapen K, Schumacher HRJ, van Linhoudt D, et al.
44. Langlands DR, Dawkins RL, Matz LR, et al. Arthritis Tophaceous gout in young patients with systemic lupus
associated with a crystallizing cryoprecipitable IgG erythematosus. J Rheumatol. 1993;20:721.
paraprotein. Am J Med. 1980;68:461. 63. DeCastro P, Jorizzo JL, Solomon AR, et al. Coexistent
45. Skinner M, Cohen AS. Calcium pyrophosphate dihy- systemic lupus erythematosus and tophaceous gout. J
drate crystal deposition disease. Arch Intern Med. Am Acad Dermatol. 1985;13:650.
1969;123:636. 64. Frocht A, Leck JC, Robbins DL. Gout and hyperuri-
46. Zuckner J, Uddin J, Gantner GE, Dorner RW. caemia in systemic lupus erythematosus. Br J
Cholesterol crystals in synovial fluid. Ann Intern Rheumatol. 1987;26:303.
Med. 1964;60:436. 65. Kurita Y, Tsuboi R, Numata K, Ogawa H. A case of
47. Newcombe DS, Cohen AS. Chylous synovial effusion multiple urate deposition, without gouty attacks, in a
in rheumatoid arthritis. Am J Med. 1965;38:156. patient with systemic lupus erythematosus. Cutis.
48. Tourliere D, Benhamou CL. Lipid microcrystals and 1989;43:273.
other rare crystals in the joints. Rev Prat. 1994; 66. Kowalec JK, Krey PR. Gout in an urban setting. J Med
44:197. Soc N J. 1978;75:617.
49. Verbruggen LA, Bourgain C, Verbeelen D. Late pre- 67. Lowenthal MN, Dymond D. Gout and hyperuricae-
sentation and microcrystalline arthropathy in primary mia in blacks. S Afr Med J. 1977;52:832.
hyperoxaluria. Clin Exp Rheumatol. 1989;7:631. 68. Mody GM, Naidoo PD. Gout in South African blacks.
50. Kahn C, Hollander J, Schumacher HR. Corticosteroid Ann Rheum Dis. 1984;43:394.
crystals in synovial fluid. JAMA. 1970;211:607. 69. Mijiyawa M. Gout in patients attending the
51. Menard HA, de Medicis R, Lussier A, Brown J. Rheumatology unit of Lome Hospital. Br J Rheumatol.
Charcot-Leyden, not corticosteroid, crystals. Arthritis 1995;34:843.
Rheum. 1983;26:1533. 70. Cassim B, Mody GM, Deenadayalu VK, Hammond
52. Crugnola A, Schiller A, Radin E. Polymeric debris in MG. Gout in South Africans: a clinical and genetic
synovium after total hip replacement: histological study. Ann Rheum Dis. 1994;53:759.
identification. J Bone Joint Surg. 1977;59A:860. 71. Soderquist B, Jones J, Fredlund H, Vikerforst T.
53. Pascual E, Batlle-Gualda E, Martinez A, et al. Bacterial or crystal-associated arthritis? Discriminating
Synovial fluid analysis for diagnosis of intercritical ability of serum inflammatory markers. Scand J Infect
gout. Ann Intern Med. 2000;131:756. Dis. 1998;30:591.
54. Kennedy TD, Higgens CS, Woodrow DF, Scott JT. 72. Arthur RE, Stern M, Galeazzi M, et al. Synovial fluid
Crystal deposition in the knee and great toe joints of lactic acid in septic and nonseptic arthritis. Arthritis
asymptomatic gout patients. J R Soc Med. 1984; Rheum. 1983;26:1499.
77:747. 73. Curtis GD, Newman RJ, Slack MP. Synovial fluid lac-
55. Johnson JR. Diagnosis of intercritical gout. Ann tate and the diagnosis of septic arthritis. J Infect.
Intern Med. 2000;132:843. 1983;6:239.
56. Rouault T, Caldwell DS, Holmes EW. Aspiration of 74. Kortekangas P, Peltola O, Toivanen A, Aro HT. Synovial-
the asymptomatic metatarsophalangeal joint in gout fluid D-lactic acid in bacterial and other acute joint effu-
patients and hyperuricemic controls. Arthritis Rheum. sions. Scand J Rheumatol. 1994;23:203.
1982;25:209. 75. Wilbrink B, van der Heijden IM, Schouls LM, et al.
57. Jaccard YB, Gerster JC, Calame L. Mixed monoso- Detection of bacterial DNA in joint samples from
dium urate and calcium pyrophosphate crystal- patients with undifferentiated arthritis and reactive
induced arthropathy. A review of seventeen cases. arthritis, using polymerase chain reaction with univer-
Rev Rhum Engl Ed. 1996;63:331. sal 16S ribosomal RNA primers. Arthritis Rheum.
58. O’Connell PG, Milburn BM, Nashel DJ. Coexistent 1998;41:2276.
gout and septic arthritis: a report of two cases and lit- 76. van der Heijden IM, Wilbrink B, Schouls LM, et al.
erature review. Clin Exp Rheumatol. 1985;3:265. Detection of mycobacteria in joint samples from
59. Durback MA, Schumacher Jr HR. Acute gouty arthri- patients with arthritis using a genus-specific poly-
tis in 4 patients with systemic sclerosis. J Rheumatol. merase chain reaction and sequence analysis.
1988;15:1503. Rheumatology. 1999;38:547.
60. Rodriguez MA, Paul H, Abadi I, et al. Multiple 77. Wilkinson NZ, Kingsley GH, Jones HW, et al. The
microcrystal deposition disease in a patient with sys- detection of DNA from a range of bacterial species with
References 197

a variety of arthritides using a nested, broad-range poly- and characterization of bacterial species in arthritis syn-
merase chain reaction. Rheumatology. 1999;38:260. ovial tissue. Infect Immun. 2000;68:6012.
78. Louie JS, Liebling MR. The polymerase chain reac- 81. van der Heijden IM, Wilbrink B, Vije AE, et al.
tion in infectious and post-infectious arthritis. A Detection of bacterial DNA in serial synovial samples
review. Rheum Dis Clin North Am. 1998;24:227. obtained during antibiotic treatment from patients
79. Jalava J, Skurnik M, Toivanen P, Eerola E. Bacterial with septic arthritis. Arthritis Rheum. 1999;42:2198.
PCR in the diagnosis of joint infection. Ann Rheum 82. Ortel RW, Newcombe DS. Acute gouty arthritis and
Dis. 2001;60:287. response to colchicine in the virtual absence of
80. Kempsell KE, Cox CJ, Hurle M, et al. Reverse tran- synovial-fluid leukocytes. N Engl J Med. 1974;290:
scriptase-PCR analysis of bacterial rRNA for detection 1363.
Roentgenographic Findings
and Musculoskeletal Ultrasound 7

Introduction that may show an elevated serum uric acid level,


and therefore a precise and unequivocal diagno-
The diagnosis of gout is based primarily on the sis of gout is essential as the first step in the man-
medical history and clinical findings. Radiological agement of gout. The gold standard for the
examinations of patients with gout should most diagnosis of gout has become the identification
frequently be utilized either to exclude other of monosodium urate (MSU) crystals obtained
diagnoses or to identify tissue urate deposits. The from an inflamed joint or a tophus (see Chap. 6).
latter findings assist the clinician in the For these reasons, there has been a real need for a
identification of those patients that require inter- noninvasive means to identify uric acid crystal
vention with hypouricemic agents. The three pri- deposits without the need for synovial fluid
mary radiographic manifestations of gout are: aspirations.
joint and/or bursal effusions with soft tissue
swelling, bony erosions, extra-articular tophi,
and uric acid stones. Less common findings Soft Tissue Swelling and Joint
include the rare finding of avascular necrosis due Effusions
to tophaceous deposits interfering with blood
supply to bone, enlarged kidneys secondary to Acute gouty arthritis is manifested by an intense
polycystic kidney disease, enlarged liver shadows inflammatory reaction of the involved joint or
due to alcoholism or glycogen storage disease, joints with associated soft tissue swelling and
and massive hydronephrosis, hydroureter, and often an effusion in the affected joint(s).
dilation of the bladder in nephrogenic diabetes Although X-rays of the involved joints are not
insipidus associated with hyperuricemia and likely to change the clinical impression and will
gout. In addition, urate crystal deposits may be not exclude the presence of intra-articular sep-
visualized by both musculoskeletal ultrasound sis, an X-ray of the involved joint may exclude
and dual-energy computerized tomography, and complicating disorders such as fractures, osteo-
these techniques will be discussed after some myelitis, or chondrocalcinosis. Clinical exami-
consideration of the use of ordinary radiography. nation is able to detect large effusions of involved
The absence of physicians skilled in arthro- joints, and often a positive bulge sign can iden-
centesis or the unwillingness to refer patients to tify small effusions in joints. Arthrocentesis is
those with such skills, and often, the lack of an essential to exclude infection and to identify
appropriate microscope for visualizing uric acid urate microcrystals. The failure of an attempt at
crystals under polarizing light may lead to arthrocentesis can be due to several factors
unsound clinical judgments. It is important to including a medial patellofemoral compartment
recognize that gout is only one of many disorders fat pad, the presence of lipoma arborescens, a

D.S. Newcombe, Gout, 199


DOI 10.1007/978-1-4471-4264-5_7, © Springer-Verlag London 2013
200 7 Roentgenographic Findings and Musculoskeletal Ultrasound

medial plica, viscous synovial fluid, or needle Bony Erosions


blockage by rice bodies or other debris [1]. In
obese patients, fat deposits may simulate small Cyst-like lesions (tophi) containing aggregated,
effusions and can also be a reason for the failure radiolucent deposits of uric acid occur adjacent
to withdraw synovial fluid [1]. These causes for to the margins of the articular cortex and erode
the apparent absence of joint fluid on arthrocen- the underlying bone in gout. They may also local-
tesis have been confirmed by magnetic resonance ize to juxtachondral areas and simulate the ero-
imaging supplemented by intravenous gadoli- sions observed in other inflammatory arthritides
nium contrast [1]. such as rheumatoid arthritis. These lesions, simi-
If clinical suspicions mandate the need for lar to the cystic changes observed in rheumatoid
arthrocentesis, and fluid cannot be detected with arthritis, are not usually associated with para-
certainty on physical examination, a magnetic res- articular osteoporosis caused by the inflammatory
onance image can define the precise anatomical rheumatoid pannus. In contrast, the gouty tophus
location of an effusion for purposes of directing is a sharply marginated, punched-out defect in
the physician in the performance of the aspira- the joint margins of the small bones of the hands
tion. This need for an MRI occurs very infre- and feet. These tophi often have thin sclerotic
quently but may arise in patients where infection margins with the characteristic overhanging mar-
is a strong consideration, and the identification of gin of bone [4]. The marginal deposition of urate
the infectious agent is necessary to initiate treat- tophi also causes periosteal new bone formation
ment promptly. It is often most advantageous to localized to the outer margin of the tophus. Such
perform a synovial aspiration of the lateral knee changes often appear as an elevated overhanging
compartment and to have an assistant compress edge. This overhanging edge is frequently
the areas of the knee opposite from the point of observed in association with the gouty tophus but
aspiration. may also be seen in sarcoidosis and psoriatic
Distal peripheral joints in an asymmetric pat- arthritis. Frequently, tophaceous deposits are also
tern represent the usual distribution of acute associated with osteoarthritis of the distal inter-
gouty arthritis. The characteristic joints involved phalangeal joints that is consistent with the idea
are the ankle, the first metatarsophalangeal, inter- that tophi are found in areas of tissue damage.
phalangeal, carpal, and elbow joints [2, 3]. The Although tophi can be observed in many differ-
olecranon and prepatellar bursae are often ent sites, anteroposterior views of the hands and
inflamed and manifest associated soft tissue feet are the most rewarding X-rays for the delin-
swelling with bursal effusions. Acute gouty eation of tophi. In untreated patients with gout,
arthritis may also involve the knees that may these tophaceous deposits enlarge, destroy more
show marginal erosions of the femorotibial or bone, and in a few instances, where extensive
patellofemoral joints. In the early stages of gout, bony destruction has occurred, may appear like a
joint space width is preserved in the presence of tumor that has invaded bone (Fig. 7.1).
bony erosions, whereas the late stages of the dis- Tophi may also be identified in X-rays as soft
ease show nonuniform articular cartilage narrow- tissue masses, frequently localized to the olecra-
ing in association with degenerative joint disease. non bursa. Such masses may or may not contain
Rarely, gouty arthritis may involve the spine, punctate deposits of opaque calcium interspersed
hips, or sacroiliac joints. These sites are almost throughout the tophus. In addition to the cystic
never involved in the acute process but are more lesions of subchondral bone, tophi extending into
likely to show changes consistent with chronic soft tissue may show increased density as com-
tophaceous gout. In chronic tophaceous gout pared with the adjacent soft tissues. Upon occa-
associated with repetitive acute gouty episodes, sion, tophaceous deposits extend through the
osteopenia is observed from disuse of the overlying skin and drain granular material.
involved joint and the effect of inflammatory Unfortunately, these draining tophi are some-
mediators on bone. times mistaken for infectious processes, and
Uric Acid Calculus 201

of large quantities of urate secondary to tumor


lysis and breakdown of purine precursors is pres-
ent. Such patients are prone to acute renal failure
resulting from the accumulation of urate sludge.
Finally, infants may present with acute renal fail-
ure as a result of underlying enzyme deficiencies
predisposing them to stone formation, obstruc-
tive nephropathy, and postrenal failure. Although
postrenal failure is the least common cause of
acute renal failure when compared to prerenal
and intrinsic causes of failure, the potential for
curing postrenal failure by removing an obstruc-
tion mandates that radiographic studies be done
to evaluate the presence of urinary tract outflow
Fig. 7.1 Tophi causing erosions in early gout. Note the obstruction. Since more than 80 % of urinary cal-
lucent areas in the distal metatarsal bone and the proximal culi contain calcium, a simple plain film of the
phalanx of the great toe abdomen (KUB) is the first radiologic diagnos-
tic procedure to be performed in such settings.
clinicians may treat such lesions as the result of a However, this procedure is of little use in patients
spreading osteomyelitis with associated abscess who have disorders of purine metabolism since
formation. Examination of the exudate using stones in these diseases are usually radiolucent.
polarizing microscopy serves to identify easily The second procedure of choice used to be intra-
the cause for the lesion. venous urography. In the case of uric acid calculi,
this procedure will demonstrate a filling defect in
the pelvocalyceal system denoting the presence
Extra-articular Tophi of radiolucent uric acid stones or other opaque
structures. This procedure has now been largely
Tophaceous deposits may occur in juxtaposition replaced by ultrasonography, a technique that
to tendons and ligaments and cause weakening of avoids the risks associated with the use of con-
those structures eventually leading to rupture. trast media even though such risks are relatively
Furthermore, the deposition of urate in confined low [7]. Such risks include a spectrum of minor
spaces such as the carpal tunnel may give rise to to severe reactions such as nausea, vomiting,
an entrapment neuropathy [5, 6]. urticaria, nephrotoxicity, pulmonary edema, con-
gestive heart failure, bronchospasm, and anaphy-
laxis. Ultrasonography may reveal the presence
Uric Acid Calculus of hydronephrosis as the sole manifestation of
obstructive uropathy or may also show the calcu-
Uric acid nephrolithiasis occurs commonly in lus itself. Since most kidney stones are located at
several clinical settings. Patients with the onset the ureteropelvic or ureterovesical junction, ultra-
of gouty arthritis between the ages of 16 and 35 sound evaluations must be carefully performed to
who may or may not have a family history of gout include examinations of these locations. Partial
are at the highest risk for a genetic form of gout obstructions by a stone or forniceal ruptures may
associated with urate overproduction and the pro- result in an unrecognized presence of a stone
pensity to form urate stones. In the unusual cir- since ultrasonography may miss a kidney abnor-
cumstance of patients who have not been hydrated mality in the absence of hydronephrosis. In such
or pretreated with allopurinol and undergoing cases, if a stone is strongly suspected, evaluation
radiotherapy or chemotherapy, an increased risk by intravenous urography should be undertaken.
for urinary tract obstruction due to the excretion Nonetheless, ultrasonography as a noninvasive
202 7 Roentgenographic Findings and Musculoskeletal Ultrasound

procedure has been extremely useful for the most sensitive and specific method for the diag-
assessment of infants who present with acute nosis of osteonecrosis. The MRI can distinguish
renal failure and have been subsequently shown normal marrow and bone from unrepaired dead
to have obstructive uropathy due to stones. It is bone with marrow replaced by debris along with
important to recognize that not all urate stones areas of repair [11–13]. The high signal intensity
are radiolucent since they may contain flecks of of necrotic bone and marrow together with the
calcium that gives a speckled radiodense appear- dark striations of subchondral bone gives the
ance. Furthermore, there are other renal calculi characteristic serpiginous pattern of the MRI in
that are radiolucent including stones composed this condition. It should be noted that osteonecro-
of xanthine, 2,8-dihydroxyadenine, cystine, and sis can be seen in other disorders associated with
oxipurinol. Oxipurinol stones arise as a result of gout such as sickle cell disease, leukemia, lym-
the inappropriate use of allopurinol and are usu- phoma, organ transplantation (drug therapy), and
ally iatrogenic. alcohol abuse.

Avascular Necrosis Chondrocalcinosis

Rarely tophi in the right location can contribute Since pseudogout and gout are at times difficult
to the obstruction of the blood supply to a bone to differentiate from each other and the two con-
and result in the occurrence of avascular necrosis ditions can occur simultaneously [14–17], chon-
(osteonecrosis). In gout, osteonecrosis is com- drocalcinosis, the radiographic hallmark of
monly localized to the head of the femur [8]. This pseudogout, is helpful although not diagnostic in
disorder progresses through five stages that have cases where a microcrystalline arthritis is sus-
been defined radiographically [9]. Stage 0 repre- pected. The presence of chondrocalcinosis is of
sents the patient who is asymptomatic and has a no diagnostic significance to patients with gout
normal plain radiograph of the hips but has except for the possible concurrent occurrence of
necrotic marrow or bone defined by high signal pseudogout. The presence of punctate and linear
intensity in both T1 and T2 magnetic resonance densities in articular hyaline or fibrocartilage is
images. Stage 1 represents the patient with clini- the characteristic finding in chondrocalcinosis.
cal manifestations (pain in the affected joint) and The typical locations for calcification include the
a normal plain radiograph of the joint. Stage 2 articular cartilage and menisci of the knee, the
represents the patient who has an abnormal plain articular cartilage of the hip joint, the fibrocartilage
X-ray of the involved joint showing areas of of the symphysis pubis, the articular disc of the
osteopenia and osteosclerosis. Stage 3 represents wrist, and the annulus fibrosus of the interverte-
patients with early bone collapse on a plain film bral discs, and X-rays of these structures often
of the involved joint and may have the so-called reveal the presence of chondrocalcinosis. The
crescent sign delineating an area of dead bone as presence of chondrocalcinosis does not indicate
a translucent subcortical area of bone. Stage 4 the presence of calcium pyrophosphate dihydrate
represents patients with clearly identified bone (CPPD) crystals that must be identified in a joint
collapse manifested as flattening of the articular aspirate using compensated polarized light
surface with or without joint incongruity. If avas- microscopy. This microscopic examination will
cular necrosis is suspected in a patient, a plain identify CPPD crystals by their characteristic
roentgenogram of the involved joint or a neutral shape and their weakly positive birefringence.
anteroposterior and frog leg views of the hips Studies have indicated that anteroposterior pro-
should be obtained [10, 11]. Bone scintigraphy jections of both knees, posteroanterior views of
can also be used to assess the uptake of the radio- the hands, and anteroposterior projections of the
pharmaceutical in the same joint on both sides of pelvis will identify 80–90 % of the cases of chon-
the body [10]. As noted previously, an MRI is the drocalcinosis. In tissues where the deposits are
Dual-Energy Computed Tomography (DECT) 203

faint, fine detail radiographs of thin bones and a convenient, noninvasive method of identify-
magnification of thick bones may be useful. ing urate deposits in and around joints and also
Finally, gout and pseudogout can occur together, may be used to demonstrate the disappearance of
and radiographic signs of one do not exclude the tophi in response to therapy [18–20].
other.

Dual-Energy Computed Tomography


Musculoskeletal Ultrasound (DECT)

Musculoskeletal Ultrasound is a procedure that Dual-energy computed tomography (DECT) rep-


is now widely used in rheumatology practice. It resents another technique that permits specific
is a noninvasive technique that may be used to and quantitative visualization of urate deposits in
visualize articular and other connective tissue the human host [21–23]. DECT is a reliable, non-
structures as well as an aid to guide the aspiration invasive method that can surpass the clinical
of joint fluid and the injection of glucocorticoids examination in the detection of uric acid depos-
into joints, bursae, and tendon sheaths. It is use- its. The recent use of dual-energy computed
ful for visualizing the deposits of monosodium tomography (DECT) scans that are color-coded
urate in tissues through its features that appear to identify uric acid crystal deposits may provide
to be specific for urate deposits. The features of another noninvasive technique for the early rec-
urate deposits include the hyperechoic enhance- ognition of such tissue crystal deposits [21–24].
ment of the outer surface of hyaline cartilage or Although the dimensions of DECT’s sensitivity
the double contour sign, the so-called “soft” and for the identification of MSU deposits have yet
“hard” tophi, and the hyperechoic spots within to be completely defined, early studies have
the synovial fluid. Tophi in tissues are hyper- confirmed its use in the accurate diagnosis of
echoic, heterogeneous, and have poorly defined gout and for the quantitative evaluation of the
contours. They are surrounded by an anechoic body burden of MSU deposits. Dual-energy com-
halo (Figs. 7.2 and 7.3). This technique provides puted tomography scans are likely over time to

Fig. 7.2 Extended image in the longitudinal plane (P arrow demonstrates the continuity of tophi with the ole-
proximal, D distal) of the distal upper arm and elbow cranon bursa. The contours of the bone surface of the pos-
(outer curve), showing multiple grouped heterogeneous terior humerus are marked with open arrowheads.
hypoechoic tophi (TH) at the insertion site (arrowheads) Subcutaneous tissue is marked with an asterisk. The mus-
of the triceps tendon (T) on the olecranon (arrows). Open cle belly of the triceps is marked with an M
204 7 Roentgenographic Findings and Musculoskeletal Ultrasound

Fig. 7.3 Extended image of the posterior proximal fore- bright line underneath is the cortical bone of the ulna
arm in the longitudinal plane (P proximal, D distal) show- (arrows). A hypoechoic peripheral echo pattern encom-
ing multiple grouped hyperechoic heterogeneous tophi passes the tophi (arrowheads)
with imprecise contours (dotted lines between s). The

replace the need for an invasive arthrocentesis or important to this discussion, DECT not only
tophus needling that are essential for the permits an accurate, noninvasive diagnosis of
identification of MSU crystals. It is also likely to gout but also provides a technique for monitoring
be a more accurate method to monitor the the progression of gout and the effectiveness of
response of gouty patients to treatment regimens hypouricemic treatments. At this stage of knowl-
designed to reduce and eliminate MSU deposits edge, it would seem reasonable to perform DECT
rather than using clinical observation alone. examinations to determine the extent of uric acid
Dual-energy (DE) computed tomography deposits in gouty patients and to monitor treat-
(CT) using a Siemens SOMATOM definition ment in patients who demonstrate crystal depos-
dual source CT scanner has been able to acquire its and avoid the loss of joint functions. DECT
images that demonstrate clearly uric acid and cal- examinations of hands, feet, and elbows would
cium crystal deposition [24–27]. Clearly, addi- be a useful selection of body parts to examine for
tional and larger studies are needed to determine crystal deposition.
the sensitivity, specificity, and the lower limits for
the detection of uric acid crystals.
One additional parameter of DECT is its References
capacity to detect urinary tract stones composed
of uric acid [28–30]. These additional parameters 1. Roberts WN, Hayes CW, Breitbach SA, Owen Jr DS.
will undoubtedly have an enormous impact not Dry taps and what to do about them: a pictorial essay
on failed arthrocentesis of the knee. Am J Med.
only for uric acid overproducers and their pro- 1996;100:461.
pensity to form stones but also for those with 2. Bloch C, Hermann G, Yu T-F. A radiologic re-evalua-
genetic defects in purine metabolism leading to tion of gout: a study of 2,000 patients. AJR Am
uric acid stone formation. J Roentgenol. 1980;134:781–7.
3. Resnick D. The radiographic manifestations of gouty
To summarize these early DECT studies to arthritis. CRC Crit Rev Diag Imaging. 1977;9:265–335.
detect uric acid deposits, one can conclude that 4. Martel W. The overhanging margin of bone: a roent-
DECT is an effective tool for diagnosing gout genologic manifestation of gout. Radiology. 1981;91:
and demonstrating uric acid crystals. It can also 755–6.
5. Ogilvie C, Kay NR. Fulminating carpal tunnel syn-
differentiate atypical forms of inflammatory drome due to gout. J Hand Surg. 1988;13:42–4.
arthritis from gouty arthritis by showing the 6. Pai CH, Tseng CH. Acute carpal tunnel syndrome caused
absence of uric acid crystals. Finally, most by tophaceous gout. J Hand Surg. 1993;18:667–9.
References 205

7. Caro JJ, Trindale E, McGregor M. The risks of death 20. Thiele RG, Schlesinger N. Diagnosis of gout by ultra-
and of severe nonfatal reactions with high- vs low- sound. Rheumatology. 2007;46:1116–21.
osmolality contrast media: a meta-analysis. AJR Am 21. Nicolaou S, Yong-Hing CJ, Galea-Soler S, et al.
J Roentgenol. 1991;156:825. Dual-energy CT as a potential new diagnostic tool in
8. Mielants H, Veys EM, DeBussere A, et al. Avascular the management of gout in the acute setting. Am
necrosis and its relationship with lipid and purine J Roentgenol. 2010;194:1072.
metabolism. J Rheumatol. 1975;2:430. 22. Artmann A, Ratzenback M, Noszian I, Trieb K. Dual
9. Arlet J, Ficat P. Diagnostic de l’osteonecrose femoro- energy CT – a new perspective in the diagnosis of
capitale primitive au stade I (Stade preradiologic). gout. Rofo. 2010;182:261.
Rev Chir Orthop. 1968;54:637. 23. Choi HK, AI-Arfaj AM, Eftekhari A, et al. Dual
10. Mankin HJ. Nontraumatic necrosis of bone (osteone- energy computed tomography in tophaceous gout.
crosis). N Engl J Med. 1992;326:1473. Ann Rheum Dis. 2009;68:1609.
11. Lee MJ, Corrigan J, Stack JP, et al. A comparison of 24. Johnson TRC, Weckbach S, Kellner H, et al. Clinical
modern imaging modalities in osteonecrosis of the image: dual-energy computed tomographic molecular
femoral head. Clin Radiol. 1990;42:427. imaging of gout. Arthritis Rheum. 2007;56:2809.
12. Jergesen HE, Lang P, Moseley M, et al. Magnetic 25. Choi HK, AI-Arfaj A, Eftekhori A, et al. Dual energy
resonance imaging in osteonecrosis of the femoral computed tomography in tophaceous gout. Ann
head. Orthop Clin North Am. 1985;16:705. Rheum Dis. 2009;68:1609.
13. Jergesen HE, Lang P, Moseley M, et al. Histologic cor- 26. Dalbeth N, McQueen FM. Use of imaging to evaluate
relation in magnetic resonance imaging of femoral head gout and other crystal deposition disorders. Curr Opin
osteonecrosis. Clin Orthop Relat Res. 1978;253:150. Rheumatol. 2009;21:124.
14. Dodds WJ, Steinbach HL. Gout associated with 27. Bacani AK, McCollough CH, Glazebrook KN, et al.
calcification of cartilage. N Engl J Med. 1966;275:745. Dual energy computed tomography for quantification
15. Reginato MA, Valenzuela RF, Matinez CV, et al. of tissue urate deposits in tophaceous gout: help from
Polyarticular and familial chondrocalcinosis. Arthritis modern physics in the management of an ancient dis-
Rheum. 1970;13:197. ease. Rheumatol Int. 2012;32(1):235–9. Epub 2009
16. Skinner M, Cohen AS. Calcium pyrophosphate dihy- Dec 17.
drate crystal deposition disease. Arch Intern Med. 28. Graser A, Johnson TR, Bader M, et al. Dual energy
1969;123:636. CT characterization of urinary calculi: initial in vitro
17. Smith JR, Phelps P. Septic arthritis, gout, pseudogout and clinical experience. Invest Radiol. 2008;43:112.
and osteoarthritis in the knee of a patient with multi- 29. Vandervaeren D, Kirsch J. Dual energy x-ray absorp-
ple myeloma. Arthritis Rheum. 1972;15:89. tiometry: progress in the radiological identification of
18. Fernandez E, Kubota ES, Sandim GB, et al. Ultrasound uric acid stones. Prog Urol. 2008;18:562.
features of tophi in chronic tophaceous gout. Skeletal 30. Boll DT, Patil NA, Paulson EK, et al. Renal stone
Radiol. 2011;40:309–15. assessment with dual-energy multidetector CT and
19. Thiele RG. Role of ultrasound and other advanced advanced postprocessing techniques: improved char-
imaging in the diagnosis and management of gout. acterization of renal stone composition – a pilot study.
Curr Rheumatol Rep. 2011;13:146–53. Radiology. 2009;250:813.
Mechanisms of the Acute Attack
of Gout and Its Resolution 8

Overview inflammatory cells from the postcapillary venules


of the synovial membrane into the synovial cav-
The hallmarks of an acute inflammatory response ity, the phagocytosis of MSU crystals, and the
in the synovial joint space are the presence of eventual propagation of those factors inducing
swelling, redness, warmth, pain, and loss of full the resolution of the acute gouty episode as well
function of the involved joint. These external as other mechanisms that may contribute to the
findings are reflected in the joint space by the cessation of the acute inflammatory response.
recruitment and emigration of polymorphonu- Each of the foregoing processes is complex
clear leukocytes from the intravascular compart- and only partially characterized in gout. In gen-
ment into the joint space and the accompanying eral, only the biomarkers recovered from the syn-
vascular leakage via the postcapillary venules of ovial fluid can be assayed during an acute episode
proteinaceous fluids containing proinflammatory of gout since the molecular pathology of the
mediators. This complex process requires the human synovial membrane in the course of an
production and release of both cell- and plasma- acute attack of gout is almost never available for
derived mediators as well as endothelial cell and analysis. However, the injection of MSU crystals
neutrophil responses to cause the transmigration into an artificially constructed or actual joint
of inflammatory cells from the intravascular com- space using animal models of acute gouty arthri-
partment to the synovial compartment and the tis provides information on the pathogenesis of
subsequent generation of the crystal-induced acute gout in the human. In fact, the dog model
inflammation associated with acute gout. shows that edema, vasodilation, and neutrophil
Acute gouty arthritis is the model for all types migration are the earliest events in crystal-in-
of crystal-induced synovitis, and many differ- duced synovitis [1]. One may assume that similar
ent factors contribute to the inflammatory and changes occur in the human during the early
anti-inflammatory events that result from the stages of an acute attack of gout.
precipitation of monosodium urate (MSU) crys- In addition to the gross changes associated
tals, the subsequent intra-articular inflammatory with the inflammatory response to MSU crystals,
response, and the eventual resolution of the molecular alterations also occur at the level of a
acute inflammatory process. Specific interre- single cell (neutrophil or macrophage) to induce
lated events are essential to the pathogenesis of and regulate a variety of cellular processes. These
acute gouty arthritis including the formation and include those changes required by the cell to
precipitation of urate crystals, the generation phagocytose and completely engulf the MSU
and release of neutrophil chemoattractants and crystal or the apoptotic neutrophil, to alter the
proinflammatory mediators from both humoral shape of the cells necessary to accomplish these
and cellular sources, the transmigration of tasks, to trigger and regulate the intracellular

D.S. Newcombe, Gout, 207


DOI 10.1007/978-1-4471-4264-5_8, © Springer-Verlag London 2013
208 8 Mechanisms of the Acute Attack of Gout and Its Resolution

changes essential for the transportation of the Many areas of investigation are necessary
phagocytosed components to intracellular sites to arrive at a complete picture of the diverse
for degradation, and to generate anti-inflammatory and complex processes necessary to address
agents necessary for the resolution of the acute the acute gouty episode and its resolution. The
process. identification of possible sites for pharmacologic
Although few studies have examined the intervention of these pathological responses may
resolution of acute gouty arthritis, the molecular also reveal more specific targets for modifying
events involved in the spontaneous resolution of the acute process. Further, knowledge obtained
the acute inflammatory response associated with from the detailed study of gout will also iden-
gout are likely to be of great significance to the tify parameters of pertinence to other forms of
physiological management of acute gout. This crystal-induced arthritis and acute inflammatory
resolution process includes the clearance of neu- reactions.
trophils from the synovial space, the suppression Since major advances have been made in
of the activity of proinflammatory mediators, defining the molecular details of an acute
the release of anti-inflammatory mediators, and inflammatory response, the complexity of these
the return of the synovial membrane vascula- processes have increased dramatically. To iden-
ture to its normal tone and blood flow. A num- tify putative target sites for pharmacologic agents
ber of molecular events are likely to participate designed to alter this acute reaction and to develop
in this process including neutrophil apoptosis, more rational types of therapy, the intricacy of
receptor desensitizations, the generation of anti- the attraction and migration of cellular compo-
inflammatory mediators, and other regulatory nents of the inflammatory response as well as the
processes. The molecular pathways and require- generation of inflammatory mediators and the
ments for the resolution of an acute inflammatory expression of cell receptors for them must also be
reaction like acute gout are likely to be as com- fully characterized. Moreover, the handling of
plex and interdependent as the generation of an ligands associated with the inflammatory process
acute inflammatory response. and the internal signaling to transduce their mes-
The principal reasons for providing a detailed sages provides a molecular picture of their mech-
molecular description of these inflammatory- anisms of action and also identifies potential
provoking processes as well as the putative pharmacologic targets for the inhibition or
resolution events is to identify more clearly the enhancement of these activities. In addition to
mechanisms that initiate, sustain, and eventu- describing the role of cells and mediators in the
ally resolve the acute gouty episode. A variety generation of an inflammatory response, little
of endogenous and exogenous anti-inflammatory attention has been given to the mechanism by
agents have been described, and their mechanisms which the acute gouty episode resolves on its
of action offer a means of understanding the reso- own without any therapy. In this discussion, some
lution of inflammatory responses. Through an of the components that might propagate this reso-
understanding of the molecular pathogenesis of lution of acute gout are also discussed.
acute gout and its resolution, one may be able to
delineate possible mechanisms for the preven-
tion of acute episodes of gout or to define ways to Synovial Membrane
shorten the duration of the acute episode. There are
many complex processes that are part of the initia- The synovial membrane is a delicate structure
tion, propagation, and resolution of an acute gouty that lines synovial joints, bursa, and tendon
episode. Cell migration, phagocytosis, apoptosis, sheaths, and its intimal layer of cells is only one
and internal cell regulatory mechanisms, and the to three cell layers thick. This delicate, undulat-
control of these and other processes are incom- ing membrane present in synovial joints is made
pletely characterized and can only be described as up of the so-called synovial membrane intima
fragmented pieces of a large puzzle. which overlies a subsynovial layer of fibroblasts
Synovial Membrane 209

and adipocytes intermixed with collagen fibers the type A and B cell [4]. The two primary syn-
and proteoglycans [2]. Vascular endothelial cells ovial cell types (A and B) can be differentiated by
with basement membranes, unmyelinated nerve cytochemical staining to identify nonspecific
endings, and lymphatic channels are also found esterase activity in type A cell membranes,
in the subsynovial tissue, but these components whereas uridine diphosphoglucose dehydroge-
are mostly absent from the synovial membrane nase and prolyl hydroxylase activities are used to
intima. detect type B synovial membrane cells [12, 13].
Ultrastructural studies of the synovial mem- The absence of leukocyte and endothelial cell
brane intima have demonstrated the presence of antigens by immunohistochemical staining
two principal cell types: type A and type B syn- confirms these cell types. There are a number of
ovial lining cells [3–6]. The type A cell has an publications that have dealt with changes in these
appearance similar to a tissue macrophage with synovial membrane lining cells derived from
highly developed Golgi complexes, smooth- synovial membranes recovered from patients
walled vacuoles, mitochondria with an electron- with various pathological disorders, but no such
dense matrix and irregular cristae, and filopodia. studies have been done in patients with acute
The nucleus of this macrophage-like cell is dense, gouty arthritis and few studies have investigated
rich in heterochromatin, and surrounded by abun- normal synovial membrane for the components
dant cytoplasm containing a paucity of rough cited in the following references [14–22]. Thus,
endoplasmic reticulum and ribosomes. Studies the synovial membrane lining cells in acute gouty
have documented the bone marrow derivation of arthritis remain to be completely characterized.
type A cells using mouse radiation chimeras [7]. The synovial membrane has three principal
The membrane properties of these type A cells functions: the production of joint lubricant, the
such as the presence of nonspecific esterase, Ia removal of debris, and the repair of joint damage.
surface antigens, and rosette formation with IgG- The type 8 cell is constructed for biosynthetic
coated sheep red blood cells also provide further activities and is the source of hyaluronic acid that
credence for their monocytic origin [8]. On the is responsible for the friction-free motion of hya-
basis of their origin from monocytes, these type line articular cartilage. It also produces the col-
A phagocytic synovial macrophages may pro- lagen for joint remodeling. The vascular adhesion
duce IL-8 and LTB4, potent chemoattractants. molecule-1 (VCAM-1) has also been detected on
Type A synovial membrane lining cells (mac- type 8 synoviocytes as well as in the vascular
rophage-like synoviocytes) also stain with mono- wall outside the endothelial cell layer [23]. In
clonal antibodies for annexin-1, the proposed addition, the ligand for LFA-1 (leukocyte func-
mediator of the anti-inflammatory activity of glu- tion antigen) and intercellular adhesion mole-
cocorticoids [9]. Annexin-11 staining has also cule-1 (ICAM-1) have also been demonstrated on
been identified in perivascular tissues and synovial cells [24]. As noted, the synovial mem-
endothelial cells. The latter cells stain strongly brane intimal fibroblasts produce hyaluronan,
for annexin-IV and annexin-VI. The type B syn- fibronectin, type IV collagen, laminin, and chon-
ovial membrane cell (fibroblast-like synoviocyte) droitin-6-sulfate-bearing proteoglycans [12, 25–
has a pale nucleus with an abundance of rough 28]. Synovial fibroblasts (type 8 cells) also
endoplasmic reticulum, a few ribosomes in its express high levels of complement decay-accel-
cytoplasm, and a paucity of cytoplasmic Golgi erating factor (OAF) [12, 29–32]. Type B syn-
complexes and vacuoles. The type B synovial ovial cells may also synthesize lubricant,
membrane cell has the ultrastructural appearance phospholipids, unique carbohydrates, and the
of a connective tissue fibroblast, and these cells potent chemoattractant, IL-8 [33–35].
are derived from mesenchyme [10, 11]. Some The type A cell is constructed for the removal
investigators have described the presence of a of both cellular and particulate debris. It is also
type C synovial membrane cell resident in the likely to be involved in the phagocytosis of urate
intima with ultrastructural components of both and CPPD crystals. The synovial membrane is
210 8 Mechanisms of the Acute Attack of Gout and Its Resolution

also the site of fenestrated blood vessels as well Table 8.1 Principal cellular distribution and expression
as nerves and lymphatic channels. The type A of cell adhesion molecules
synovial lining cells have not been cultured Adhesion molecules and ligands M G EC p
in vitro so their functions have been inferred from L-selectin (CD62L) + +1 +1
studies of blood mononuclear cells from which P-selectin (CD62P)
these type A synovial cells are derived. E-selectin (CD62E) +1
Studies have shown that normal and diseased GlyCAM-1 CD62L ligand +
human synovium can be engrafted into subcu- PSGL-1 CD62P ligand + +
ESL-1 CD62E ligand + +
taneous pouches in the ears of severe combined
Integrin LFA-1 (CD11a/CD18) + +
immunodeficient mice [36]. These xenografts
Integrin Mac-1 (CD11b/CD18) + +
retain the identical cell composition of the nor-
Integrin CD11c/CD18 + +
mal synovium including endothelial cells. Such
Integrin CD11d/CD18 + +
synovial grafts also respond to cytokines (tumor Integrin ligand ICAM-1(CD54) + +1
necrosis factor-alpha) and express intercellu- Integrin ligand ICAM-2 (CD102) + + +
lar adhesion molecule-1 (ICAM-1) as well as Integrin ligand ICAM-3 (CD50) + +
vascular adhesion molecule-1 (VCAM-1) and Integrin ligand VCAM-1 (CD106) + +1
E-selectin. The expression of the latter two mole- Integrin ligand VLA-4 (CD49d) +
cules decreases over time. These grafts composed PECAM (CD31) + + + +
of human synovium may permit a more inte- G granulocyte, M monocyte/macrophage, EC endothelial
grated view of the early stages of monosodium cell, P platelet
urate crystal-induced inflammatory responses. 1. Endothelial cells must be activated to express these
components
As indicated above, the synovium is enriched
with capillaries and postcapillary venules. The
superficial vessels are fenestrated, whereas the endothelial cells. At this point, the neutrophils
deeper vessels in the subsynovial layer have con- are prepared to migrate across the blood vessel
tinuous endothelium. A few studies have exam- wall and into the tissue parenchyma. This com-
ined the fine structure and functions of the synovial plex process of neutrophil adherence requires
membrane, but investigations of the molecular both non-chemotactic cytokines (tumor necrosis
biology and detailed biochemical analyses have factor, interleukin-1, and gamma interferon) and
lagged behind those of other tissues since access chemoattractants (C5a, PAF, LTB4, and IL-8).
to the human synovium in acute inflammatory The distribution and expression of the cell adhe-
processes is prohibited [37–43]. Thin-walled lym- sion molecules on endothelial cells, monocytes,
phatics have also been identified close to the deep platelets, and neutrophils are summarized in
synovial membrane capillaries and venules [44]. Table 8.1. To begin their emigration, neutrophils
The synovial membrane capillaries and venules use selectin receptors to form loose attachments
are lined with endothelial cells that are of great with endothelial cells and to start rolling along
significance to an inflammatory process since the endothelium. Subsequently, neutrophils are
these microvessels regulate the trafficking of anchored tightly to the endothelium by b2 integ-
inflammatory cells. Endothelial cells and their rins before embarking on their migration through
functions of significance to the inflammatory pro- venule walls to the inflammatory site.
cesses are described in a subsequent section. There are a number of cell surface adhesive
Circulating neutrophils emigrating from the molecules that play an essential role in the migra-
intravascular compartment to make contact with tion of neutrophils into the joint cavity during
endothelial cells and then gain access to the site an inflammatory reaction. The so-called inter-
of inflammation must first be marginated from the cellular adhesion molecules (ICAMs) are pres-
flowing blood. After margination occurs and when ent on endothelial cells and fibroblasts and are
the neutrophils are rolling along the endothelial upregulated in inflammatory processes [45–47].
surface, they then become firmly adherent to In addition, selectins and integrins mediate the
Monosodium Urate Crystallization 211

interactions between endothelial cells and leuko- Monosodium Urate Crystallization


cytes [48–61]. A number of recent publications
have now documented the expression of cell Central to the acute attack of gouty arthritis is
adhesion molecules in the normal synovium [23, the formation and precipitation of uric acid
62–66]. Such investigations have documented crystals in the synovial joint cavity. That micro-
the expression of both selectin and integrin mol- crystals of monosodium urate are essential to the
ecules in normal human synovial membranes and inflammatory process has been known since 1899
provide evidence for the mechanisms by which when acute gouty episodes and characteristic
inflammatory cells migrate from intravascular tophi were produced by the injection of sodium
compartments to sites of inflammation. The func- urate into animals [70–72]. After a hiatus of more
tion and significance of selectins and integrins in than a half-century, microcrystals of urate were
the inflammatory process are discussed in some documented in synovial aspirates from patients
detail in a subsequent section. The subsynovial with acute gouty arthritis, and their usefulness
vascular channels also permit the migration of in the diagnosis of acute gout was confirmed
precursors of the type A synovial phagocyte from [73]. It is now clear that negatively birefringent,
the bone marrow and peripheral blood to proceed needle-shaped monosodium urate crystals have
to their final residence in the synovial membrane. been identified in the synovial aspirates of most
In this same subsynovial compartment, unmyeli- patients with acute gouty arthritis, and the pres-
nated nerves exist that provide the sensory inner- ence of such crystals is the only absolute criterion
vation of the synovial membrane and contain a for the diagnosis of acute gout.
neuropeptide, substance P, that has been found in The mechanism(s) that results in the precipita-
the synovial fluid [67, 68]. Rare mast cells also tion of uric acid in the synovial space is incom-
reside in the synovium, and the rat experimen- pletely understood; however, a sudden change in
tal model of monosodium urate crystal-induced the serum urate level, either an increase or a
inflammation has been attenuated by pretreat- decrease, is almost always associated with the
ment with antihistamine drugs [69]. These lat- onset of an acute episode of gouty arthritis in the
ter drugs have little of no effect on acute gouty susceptible individual [74]. Hyperuricemia is fre-
arthritis in the human. quently a precondition to the precipitation of
In summary, the delicate synovial membrane urate crystals but is not always present since
contains all the structures necessary for the prop- patients undergoing therapy with hypouricemic
agation of the acute inflammatory process associ- agents (allopurinol) to lower their serum uric acid
ated with gout and some of the factors involved in levels may also develop acute gout. Further,
protecting the joint from the deposition of crys- hyperuricemia is not always an essential precur-
tals. The intimal cells of the synovium may syn- sor of acute gout since patients with normal
thesize and release proinflammatory cytokines serum uric acid levels (below the saturation con-
that mediate the early stages of the intrasynovial centration of 6.8 mg %) can and do develop acute
reaction to urate crystals. In fact, the type A cell gout. Hyperuricemia may also be present in indi-
may ingest crystals and form, over time, a tophus viduals who may never experience an episode of
that could also be the source of the intrasynovial gout. The latter is the case with those patients
cavity deposition of monosodium urate crystals who have certain types of chronic renal disease
in acute gouty episodes. It is also likely that the and hyperuricemia but not gout [75]. These facts
cells of the synovial membrane synthesize and lead to two conclusions. First, hyperuricemia
release proinflammatory cytokines that mediate may be helpful in the differential diagnosis of an
the early stages of the intrasynovial reaction to acute synovitis, but an elevated serum uric acid
crystals. As will be discussed in detail subse- level alone is not diagnostic of acute gouty arthri-
quently, the synovial membrane also contributes tis. Second, supersaturation of the synovial fluid
to those parameters that result in the limitation with uric acid is only one of the factors that may
and resolution of the acute gouty episode. contribute to the onset of acute gout. Obviously,
212 8 Mechanisms of the Acute Attack of Gout and Its Resolution

decreased solubility of uric acid in the synovial in vitro have documented the enhanced forma-
fluid could contribute to the precipitation of tion of urate crystals in the presence of gamma
monosodium urate crystals. Plasma and synovial globulin and insoluble type I collagen fibers
fluid uric acid concentrations are usually compa- [86]. Further, synovial fluid from gouty patients
rable in patients with gout, but plasma has been added to supersaturated solutions of sodium
determined to be a better solvent for monosodium urate under physiological conditions significantly
urate than synovial fluid [76–78]. increases urate nucleation, but synovial fluids
In addition, low intra-articular temperatures from patients with degenerative joint disease
may also contribute to the insolubility of uric acid. only increase nucleation to a moderate degree
In fact, there is a temperature gradient between and fluids from rheumatoid patients have little
the central body temperature, the more proximal effect on urate nucleation [87]. These facts lead
joints, and the peripheral joints. This temperature to the conclusion that both synovial fluid compo-
gradient has been confirmed by the direct mea- nents and physical properties contribute to
surement of intra-articular temperatures [79–81]. changes in urate solubility and the formation of
For example, in the knee joint, the temperature microcrystals.
has a range between 30.5 and 32.8 °C, whereas The interaction between monosodium urate
the ankle joint has a lower temperature (29 °C) and macromolecules has also been investigated.
[79–81]. The solubility of uric acid in normal The sites of urate deposition contain large quanti-
human plasma (maximum equilibrium concentra- ties of polysaccharides [88]. In fact, total serum
tion) in the presence of 140 mM Na+ is 6.8 mg % glycosaminoglycans have been reported to be
at 37 °C, whereas the solubility falls dramatically three times their normal level in patients with
to half that value (3.3 mg %) at 25 °C [82]. This gout [89]. Chondroitin sulfate concentrations
differential in joint temperatures from the more have also been shown to influence in vitro urate
central joints to the peripheral joints may contrib- crystal formation and crystal growth in synovial
ute to the crystallization of urate and the propen- fluids, and cartilage is known to have an affinity
sity for acute gouty arthritis to affect peripheral for absorbing urate in vitro [90, 91]. Urate is also
joints more frequently than more central joints. more soluble in aggregated than non-aggregated
However, this explanation does not hold for all proteoglycans [92]. In summary, although many
types of crystals since pseudogout, a disorder due connective tissue components may contribute to
to the precipitation of calcium pyrophosphate the crystallization of uric acid, no single element
dihydrate crystals, is observed more frequently has been identified as the key to this initial pro-
in the knee joint than the toe joint. There is also cess essential to the development of acute gouty
speculation that the increased temperature gener- arthritis.
ated by the inflammatory response in microcrys- In addition to the influence of connective tis-
talline processes may enhance the solubility of sue components on the solubility of urate, plasma
precipitated urate and in that way, contribute to proteins have also been implicated in the crystal-
the resolution of the acute episode [82]. The pro- lization and processing of urate, but little evi-
pensity for the first metatarsophalangeal joint to dence supports the interaction between plasma
be afflicted with gout (podagra) may result from proteins and serum uric acid under physiological
the constant microtrauma to which this joint is conditions. The older literature described a
subjected and may have little to do with the tem- deficiency in urate binding to an ornithine-con-
perature of the joint. taining plasma protein that migrated as an
The disruption of tissue continuity as occurs a1-globulin in gouty kindreds [93]. The same
in the presence of diseased tissues may also con- group of investigators had postulated a role for
tribute to the acute gouty episode since gout is this plasma protein in the maintenance of uric
known to occur in Heberden’s nodes and erosive acid solubility [94], but little information con-
osteoarthritis [83–85]. Experiments performed cerning this urate-binding protein is available in
Inflammasome: Innate Immunity; Inflammation in Gout Is Mediated by the Innate Immune System 213

the modern literature. In fact, some investigations Inflammasome: Innate Immunity;


have failed to detect alterations in urate binding Inflammation in Gout Is Mediated
in patients with gout [95]. by the Innate Immune System
Monosodium urate crystals isolated from the
synovial fluid and tophi of gouty patients are Introduction
coated with immunoglobulins [96, 97]. The prin-
cipal immunoglobulin coating these crystals is The inflammatory reaction in gout is mediated by
IgG, and its binding to urate crystals occurs the innate immune system. The innate immune
through the F(ab), antigen-binding fragment of system is a primitive defense mechanism evolved
the immunoglobulin, leaving the Fe crystalliz- to protect from noxious agents, especially infec-
able fragment of immunoglobulin G (IgG) free tious agents including bacteria, viruses, and fun-
for interaction with the Fe receptor present on gal organisms. It responds to dangerous substances
phagocytes. In some cases, it has been shown that and organisms, and is immediately available to
the immunoglobulin coating uric acid crystals is the host. It differs from the adaptive immune sys-
a polyclonal IgG antibody specific for urate crys- tem, which depends on the development of anti-
tals [98]. These IgG antibodies isolated from syn- bodies and specific lymphocytes that then protect
ovial fluid aspirates of patients with gout against infections and other insults.
accelerate crystal formation in supersaturated Around the turn of this century, the term
urate solutions in vitro. In contrast to this effect, “autoinflammatory” was applied to several dis-
IgG antibodies isolated from patients with ease states that differed from autoimmune dis-
osteoarthritis, rheumatoid arthritis, or pseudog- eases in that they were apparently unprovoked
out do not have this property. In fact, IgG anti- inflammatory states that were not associated with
bodies from gouty synovial fluid have a autoimmunity and were not associated with any
concentration-dependent effect on monosodium obvious infectious organisms. Several disorders
urate monohydrate nucleation. The higher the prompted this concept, initially several periodic
concentration of the IgG antibodies specific for fever syndromes including familial Mediterranean
urate, the greater the number of crystals formed. fever, the TNF receptor-associated periodic syn-
Investigations that are more recent have shown drome (TRAPS), each of which are associated
that the hydrophilic surfaces of the urate crystal with periodic fever, serositis, arthritis, and skin
also react with human serum albumin through rash, without any evidence for autoimmunity,
protein carboxylate groups [99]. Thus, proteins such as is seen with diseases such as systemic
are clearly absorbed to the negatively charged lupus erythematosus, which can cause similar
surface of the urate crystal and appear to serve manifestations.
the purpose of enhancing the capacity of such Another group of diseases that supported this
crystals to be phagocytosed. In addition, IgG concept are three syndromes that were found to be
antibodies specific for urate crystals may enhance associated with mutations in the gene, NLRP3/
crystallization since these immunoglobulin mol- CIAS1, that encodes a protein previously called
ecules have a structure like the surface of the cryopyrin, now NLRP3, an activator of interleu-
urate crystal that serves as a nucleating matrix. kin-1b, IL-1b (Fig. 8.1 and 8.2). These syndromes
Despite all these factors that may contribute to vary in their manifestations and severity of illness,
the precipitation of uric acid and the formation of depending on the location of their mutations within
crystals, no single factor has been determined as the NLRP3 gene, and all include periodic fever,
the key to the initiation of crystal formation. rashes, and in the more severe forms, bone and
Thus, a variety of factors in the surrounding joint, and neurologic disease. Subsequently, the
milieu are likely to be responsible for MSU crys- concept of autoinflammatory disease has been
tal formation including some components that extended to include many both rare and common
may protect the host from crystal deposition. diseases extending beyond the periodic fever
214 8 Mechanisms of the Acute Attack of Gout and Its Resolution

DAMP/
PAMP NLRP1 inflammasome
K
+ DAMP/PAMP
ATP Pannexin-1 NLRP1 PYD NACHT LRR
Crystalline/
P2X7
particulate
ligands CARD FIIND

1 CASP1 CARD
CYTOPLASM

2 p20 p10
3
Reactive oxygen NLRP3 inflammasome
species Lysosomal
R rupture NLRP3 PYD NACHT LRR
LR

LR
R

ASC PYD
NACHT
NACHT

NLARP3

CARD
D
PY

PY CASP1 CARD p20 p10


D

D ASC
PY

PY
D
D
CARD

CARD
D

IPAF inflammasome
CAR
CAR

IPAF CARD NACHT LRR


CASP
CASP
NUCLEUS

CASP1 CASP1 CARD


Cathepsin B
NLRP3 inflammasome inhibitor p20 p10
assembly (Ca-074-me) ASC?
NAIP?

CASP AIM2 inflammasome


Activated
CASP Caspase-1 AIM2 PYD HIN
ASC PYD
IL-1ß
Pro-IL-1ß
“Priming” by CARD
proinflammatory stimuli
CASP1 CARD p20 p10
e.g. TLRs, TNF, IL-1ß

Fig. 8.2 Minimal NLRP1, NLRP3, IPAF, and AIM2


inflammasomes For simplicity, the unoligomerized
IL-1ß inflammasome complexes are depicted. Removal of the
CARD domain and processing of the caspase domain of
caspase-1 by autocleavage at the indicated sites results in
Fig. 8.1 NLRP3 inflammasome activation. Three major the formation of the active caspase-1 p10/p20 tetramer. It
models for NLRP3 inflammasome activation are favored should be noted that although human NLRP1 contains a
in the field, which may not be exclusive: (1) The NLRP3 PYD, mouse NLRP1 proteins do not harbor functional
agonist, ATP, triggers P2X7-dependent pore formation by PYDs. Human NLRP1 can also recruit a second caspase,
the pannexin-1 hemichannel, allowing extracellular caspase-5, to the complex (not shown). Maximal caspase-1
NLRP3 agonists to enter the cytosol and directly engage activation in response to IPAF agonists can require ASC
NLRP3. (2) Crystalline or particulate NLRP3 agonists are or NAIP, depending on the stimulus. The interaction of
engulfed, and their physical characteristics lead to lyso- these proteins with the IPAF inflammasome activation is
somal rupture. The NLRP3 inflammasome senses lyso- currently unclear. Domains: CARD, caspase recruitment
somal content in the cytoplasm, for example, via domain; FIIND, domain with function to find; HIN, HIN-
cathepsin-B-dependent processing of a direct NLRP3 200/IF120x domain; LRR, leucine-rich repeat; NACHT,
ligand. (3) All danger-associated molecular patterns nucleotide-binding and oligomerization domain; PYD,
(DAMPs) and pathogen-associated molecular patterns pyrin domain (From Schroder and Tschopp [100].
(PAMPs), including ATP and particulate/crystalline acti- Reproduced with permission)
vators, trigger the generation of reactive oxygen species
(ROS). A ROS-dependent pathway triggers NLRP3 inflam-
masome complex formation. Caspase-1 clustering induces
syndromes. While several of the periodic fever
autoactivation and caspase-1-dependent maturation and syndromes involve genetic mutations of the NLRP3
secretion of proinflammatory cytokines, such as interleu- gene, a protein which associates with other pro-
kin-1b (IL-1b) and IL-18 teins to form the NLRP3 inflammasome, others
Inflammasome: Innate Immunity; Inflammation in Gout Is Mediated by the Innate Immune System 215

have mutations in other proteins that activate and the induction of the components necessary to
inflammasomes, and others disease have known respond to the attack on the host’s tissues cre-
mutations in other proteins, and in some, the defect ated by the crystals. In order to detect and mount
is unknown. In addition to the inflammasome dis- a response to such dangerous substances, the
orders, five other categories have been proposed. human host has developed both extracellular and
These include: NF-kB activation disorders, pro- intracellular sensors that identify microbial and
tein-misfolding disorders, complement disorders, viral components by generating pattern-recogni-
cytokine-signaling diseases, and macrophage acti- tion receptors (PRRs) called pathogen-associated
vation syndromes [101, 102]. molecular patterns (PAMPs) [105–107, 109–118].
Finally, a substantial body of evidence has In the case of MSU or CPPD crystals, the sensor
shown that certain particulate matter induces has been called danger-associated molecular pat-
inflammatory reactions through activation of the terns (DAMPs), and these crystal stimuli have
NALP3 inflammasome. These include monoso- been demonstrated to cause the activation of the
dium urate and calcium pyrophosphate dehydrate NALP3, now called the NLRP3, inflammasome
(CPPD) crystals, as well as asbestos and silica. that is involved in the inflammatory response
Studies with urate and CPPD crystals in normal to these crystals [119]. Several types of innate
mouse macrophages demonstrate caspase activation immune signaling receptors have been identified
and IL-1b release, and this response is abrogated in [120]. Toll-like receptors (TLRs) and C-type lectin
Nlrp3 knockout mice [103]. Studies by others have receptors are transmembrane molecules localized
shown that urate-induced inflammation was reduced to the plasma membrane and to the membranes of
in mice deficient in the inflammasone component, lysosomes, endosomes, and endoplasmic reticu-
Asc or in the IL-1 receptor, or in the IL-1 receptor lum. Receptors found in the cell cytosol include
component, MyD88, but the inflammatory response the retinoic acid-inducible gene-I-like helicases or
was not reduced in mice deficient in each of several RIG-I-like receptors (RLRs), and the nucleotide-
toll-like receptors [104]. binding domain leucine-rich repeat-containing
Recent investigations have determined that crys- receptors or Nod (nucleotide oligomerization
talline structures like monosodium urate (MSU), domain)-like receptors (NLRs) [121–126]. These
calcium pyrophosphate dihydrate (CPPD), silica, innate immune-signaling receptors have diverse
and asbestos create an inflammatory response functional capacities in that they can recognize
through their capacity to interact with the innate molecules associated with microbes, molecules
immune system that identifies certain foreign sub- related to tissue damage, molecules not synthe-
stances in a manner similar to the mechanism by sized by the host such as lipopolysaccharide, and
which it detects invading microbes [105–115]. The molecules related to diseases such as monosodium
innate immune system is a more primitive system urate (MSU) crystals in gout, calcium pyrophos-
relative to the adaptive immune system, and it gen- phate dihydrate (CPPD) crystals in pseudogout,
erates a nonspecific response when recognizing silica in silicosis, and asbestos in asbestosis.
and responding to dangerous signals like MSU or The sensors of the danger to the host in the
CPPD crystals. Once these dangerous substances case of uric acid crystals interact with intracellu-
are recognized by their receptors, a complex lar innate immune receptors that trigger the
protein scaffolding called an inflammasome is construction of a complex protein called the
constructed, and its formation leads to the attrac- inflammasome. The nucleotide-binding domain
tion of a caspase enzyme that converts inactive leucine-rich repeat-containing receptors or NLRs
pro-IL-1 to its active proinflammatory cytokine form a group of intracellular innate immune
IL-1b. Once the latter cytokine is released from receptors of which over 20 have now been
the cell cytoplasm into the extracellular milieu, it identified in humans. These NLRs in man have
acts as a major mediator of inflammatory reactions certain common features related to their
including the recruitment of neutrophils along with C-terminus but differ in their N-terminal domain.
the production of other proinflammatory cytokines A standardized nomenclature for the NLR family
216 8 Mechanisms of the Acute Attack of Gout and Its Resolution

has now been established and is used in most of the cell cytoplasm are incompletely understood
the recent publications related to inflammasomes at this time, these processes are beginning to be
[127]. What is significant to this discussion of the unraveled [137]. It is likely that when these pro-
microcrystalline disorder, gout, is the fact that cesses are completely understood, additional tar-
NLRs not only sense microbial, viral, and fungal gets will become available to prevent the
components but may also sense nonmicrobial inflammatory reaction engendered by NLRP3.
danger signals and are dependent on the forma-
tion of large (~ 700 kDa) multiprotein, cytoplas-
mic scaffoldings called inflammasomes. These Interleukin-1b and Interleukin-18
protein complexes promote the recruitment of
caspase-1 and cause the conversion of pro-IL-1b Interleukin-1 is a multifunctional cytokine whose
to its active, proinflammatory form, IL-1b. The two active forms, IL-1a and IL-1b, have similar
NLR family member of significance to gout is the functions that include the upregulated expression
NLPR3 inflammasome that has also been of adhesion molecules, the generation of other
described by other nomenclature in the early lit- proinflammatory cytokines like IL-6, arachidonic
erature (CIASI, PYPAF 1, cryopyrin, or NALP3). acid metabolites, and chemokines with the capac-
The two commonly used terms for this molecular ity to recruit neutrophils [138]. IL-1 also causes
complex are NALP3 and NLRP3 that stand for angiogenesis, fibroblast proliferation, and fever. It
NACHT-LRR-PYD3 or NLR family pyrin family is a pleiotropic molecule acting on many cell types
containing 3. In any event, the overall structure of including leukocytes, endothelial cells, adipose
this complex (NLRP3) is PYD-NACHT-NAD- tissue, hepatocytes, and other cell types [138].
LRR. The NLR family members (NLRP1, When IL-1 is administered to humans, significant
NLRP3, and NLRC3) lead to the activation of local pain, erythema, and swelling occur at the
caspases, and therefore, they have been classified subcutaneous injection site heralding an inflammatory
as inflammasomes [128, 129]. In its inactive state, reaction [139, 140]. A number of other physical
the NLRP3 inflammasome prevents the associa- findings associated with IL-1 exposure also occur
tion of ASC (apoptosis-associated speck-like including chills, fever, and hypotension [139, 141–
protein containing a CARD) or CARDINAL with 143]. IL-1b injection also causes a significant
the PYD domain of NLRP3 because of the pre- increase in cortisol release [143, 144]. Many excel-
sumed self-association of the LRR domain. When lent, recent reviews of the functions of IL-1 and
NLRP3 is activated by nucleotides (ATP, deoxy- another cytokine produced and released by cas-
ATP, GTP, or uric acid crystals), the molecule pase-1 activity, interleukin (IL-18), have been
unfolds permitting the association of ASC with published and may be evaluated for additional
NLRP3 [130]. parameters of these inflammasome-mediated
Aspartic acid-specific cysteine proteases are activities [138, 145–153].
major players in the inflammatory response. The principle sources of IL-18 are mac-
Inflammatory initiators (caspase-1, caspase-4, rophages and dendritic cells [147]. IL-18 or
and caspase-5) and especially caspase-1 is interferon-g (IFN-g)-inducing factor was first
responsible for the processing of pro-IL-1b to its described two decades ago and was considered
active molecule, IL-1b. Procaspase-1 conversion an immunomodulatory cytokine because of its
is induced by macromolecular oligomerization of properties as an inducer of IFN-g [147]. IL-18
the proenzyme onto the complex called the or interferon-g (IFN-g)-inducing factor was first
inflammasome [125, 128, 131–134]. This process described two decades ago and was considered
is mediated by the recruitment domains in pro- an immunoregulatory cytokine because of its
caspase-1 and inflammasome adaptor proteins properties as an inducer of IFN-g [154]. IL-18 is
like ASC [135, 136]. Although the mechanisms a potent proinflammatory cytokine that mediates
involved in NLRP3 activation, procaspase-1 con- the production of some inflammatory cytokines,
version to caspase-1, and IL-1b transport out of chemokines, nitric oxide, and IFN-g, a product
Inflammasome: Innate Immunity; Inflammation in Gout Is Mediated by the Innate Immune System 217

that activates macrophages [155]. It also induces mediated macrophage production of reactive
the production of IL-1b from monocytes and oxygen species (ROS) via ROS-dependent PI3K
macrophages, and IL-6 by LPS-treated periph- activation and caspase-1 activation with the resul-
eral blood mononuclear cells [156, 157]. It can tant processing of pro-IL-1 beta, and phagosomal
also produce chemokines for the recruitment of destabilization leading to lysosomal disruption
monocytes and macrophages such as macrophage and NLRP3 activation [71, 110, 118, 131,
inflammatory protein-1 alpha (MIP-1 alpha) and 171–188]. Although the mechanisms for NLRP3
monocyte chemotactic protein-1 (MCP-1) when activation are incompletely understood, there are
exposed to peripheral blood mononuclear cells some data in support of these hypotheses.
as well as the adhesion molecule, ICAM-1 [158]. However, knowledge of the complete activation
IL-18 differs from IL-1 and TNF-a since it does process remains incomplete at this time.
not produce fever in humans [159, 160]. It also There is a good case to be made that the leak-
differs from IL-1 and TNF-a since it does not age of intracellular K+ from cells is a necessary
lead to the induction of cyclooxygenase-2 and, component for NLRP3 activation. It is well char-
therefore, does not produce PGE2 [145, 161]. acterized that ATP, asbestos, and MSU all acti-
In addition to its proinflammatory properties, vate NLRP3, but they also cause an efflux of
IL-18 is a key immunomodulating molecule potassium at the same time [110, 131, 179]. In
[162–170]. As noted previously, IL-1b is a potent support of the fact that K+ efflux is essential for
proinflammatory molecule that manifests a broad NLRP3 activation are the data showing that phys-
array of effects including an increased expression iological levels of potassium block the activation
of cytokines like TNF-a, MIP-a, and IFN-g as of NLRP3 [131, 176]. However, the mechanisms
well as proinflammatory mediators like cycloox- of NLRP3 activation are still incomplete regard-
ygenase-2, phospholipase A2, inducible nitric ing how this potassium efflux works to cause
oxide synthase, and endothelin, adhesion mol- inflammasome activation. Another mechanism
ecules like ICAM-1, VCAM-1, and ELAM1, that has some validity is NLRP3 activation by
hepatic acute phase reactants like CRP, xanthine reactive oxygen species [110, 118, 131, 179].
oxidase, lysozyme, and other inflammatory and Human monocytes have been shown to release
tissue remodeling components [138]. As a result ATP, and these cells are related to macrophages
of its increased expression when IL-1 is injected, that are known to phagocytose MSU crystals
it causes the following biological effects: fever, [189–192]. The evidence used to support this
hypotension, lactic acidosis, hyperglycemia, concept is the fact that ROS inhibition reduces
neutrophilia, increased corticosterone synthesis, the formation of mature IL-1 beta when cells are
increased release of arachidonic acid prostanoids exposed to ATP, MSU crystals, asbestos, or silica
and eicosanoids, increased neutrophils in the joint [118, 179]. Several studies have supported the
spaces when injected into joints, and many other concept that lysosomes and/or their contents may
components associated with inflammation [138]. cleave certain substrates and contribute to the
activation of NLRP3 and IL-1b production [173].
The lysosomal protease, cathepsin B, when inhib-
Mechanisms of NLRP3 Activation ited partially decreases IL-1 beta generation and
release [170, 171, 173, 174, 187, 193]. In addi-
Several hypotheses based on data derived from tion, cathepsin B-deficient macrophages also
the study of NLRP3 activators have been pro- show a reduction of IL-1b production when
posed for the mechanisms of NLRP3 activation treated with NLRP3 activators. A cathepsin
including a decrease in intracellular potassium B inhibitor also has been shown to block cas-
concentrations, ligand-receptor interactions that pase-1 activation by the lethal toxin of anthrax in
allow the DAMP to bind to a LRR receptor site another NLRP system [194].
causing a change in molecular conformation of Recent investigations have shown that NALP1
the NLRP3 and resulting in its activation, ATP- is activated by a two-step mechanism and requires
218 8 Mechanisms of the Acute Attack of Gout and Its Resolution

muranyl dipeptide (MDP) and subsequently cess of neutrophil chemotaxis must be rigorously
a ribonucleoside triphosphate to cause caspase-1 regulated. A variety of neutrophil chemoattrac-
activation of NALP1 [184]. The postulate pro- tants have been described in the human, but the
posed in this study was that the synthetic MDP contributions of each of these neutrophil chemot-
molecule, muramyl dipeptide, comprised of actic agents in acute gout remains incompletely
L-alanyl and d-isoglutamine (MDP-LD) induces defined. The chemoattractants commonly impli-
a conformational change in NALP1. This step cated in acute gout include the complement cas-
permits NALP1 to bind ribonucleotide triphos- cade product, C5a, the product of phospholipase
phates in the second step. This latter step causes A2 stimulation, leukotriene B4 (LTB4), the phos-
oligomerization of the NALP1 with the subse- phocholine lipid, platelet-activating factor (PAF),
quent generation and release of IL-1 beta. and the potent peptide neutrophil chemoattrac-
MSU crystals added to cultured monocytes tant, interleukin-8 (IL-8) [1, 195]. A number of
from controls released both IL-1 and caspase-1, neutrophil chemoattractants with structures simi-
but this response was significantly decreased in lar to IL-8 have also been characterized and des-
monocytes obtained from mice with mutations in ignated as chemokines, a term derived from the
their NALP3 molecule or ASC. When MSU crys- words, chemotactic cytokines. Growth-regulating
tals were injected into the peritoneal cavity of oncogenes (GRO-a, GRO-J3, and GRO-y)
control and mutant mice with ASC mutations, the or melanocyte growth-stimulating activity
inflammatory response was significantly reduced (MSGA), neutrophil-activating peptide-2 (NAP-
in the mutant mice as compared to the control 2), epithelial cell-derived neutrophil-activating
mice [108]. Finally, mice lacking a receptor for peptide (ENA-78), and granulocyte chemotactic
IL-1 also showed a diminished inflammatory peptide (GCP-2) are also chemokines that act as
response [105]. All the foregoing studies indi- chemoattractants for neutrophils, but no descrip-
cated that IL-1 and the inflammasome play a key tion of their role in acute gout exists [196].
role in the generation of an inflammatory response The origin of the crystals that initiate the
to MSU crystals, and discovery of its mechanisms inflammatory reaction in acute gout is unknown,
of action should be a key objective of future but several sources are possible. Urate crystal
studies. deposits, so-called synovial membrane tophi,
The proof of the principle that the inflammatory have been identified and could be a possible
reaction induced by MSU crystals activates IL-1 source of crystals initiating an acute gouty
through the inflammasome lies in the demonstra- episode. Such monosodium urate crystals might
tion that three IL-1 inhibitors have been shown to simply precipitate from the synovial fluid as
have anti-inflammatory effects on acute gouty a result of an elevated concentration of urate
arthritis, and these results are described in the exceeding its solubility in this compartment.
chapter on therapy (Chap. 9). These crystals might also have their origin in
the cartilagenous deposits (tophi) in joint carti-
lage. Urate crystals, once precipitated, then must
Neutrophil Chemoattractants make contact with a phagocytic cell to generate
chemoattractants such as IL-8 or LTB4 or activate
Overview the classical or alternative complement pathway
to generate the complement-derived chemotactic
Neutrophil chemotaxis is a critical element of the factor, C5a. Phagocytic cells capable of gener-
acute inflammatory response in gout. The recruit- ating chemotactic agents in the synovial space
ment of neutrophils must be sufficient to address include the small number of neutrophils and/or
the capture and ingestion of the inflammation- monocytes resident in the normal synovial fluid,
provoking urate crystals but insufficient to cause the monocyte-derived type A synovial membrane
permanent tissue damage from the release of toxic cell, or the type 8 fibroblast-like synovial mem-
metabolites from the neutrophil. Thus, the pro- brane cell. Acute gouty episodes can occur in
Neutrophil Chemoattractants 219

the virtual absence of neutrophils in the synovial enhance the chemoattractant effect [197]. This
fluid, and such data suggest that synovial mem- chemotactic-mediated integration of neutrophil
brane cells may also play a key role in the gen- chemotaxis is, in large part, regulated by chemot-
eration of inflammatory mediators under certain actic receptors on the neutrophil plasma mem-
conditions. brane. Fourth, some chemoattractants can block
The chemoattractants, C5a (a product of the the effect of another chemotactic agent by their
complement cascade), platelet-activating factor ability to desensitize the receptor of the other
(PAF, a product of activated endothelial cells), chemoattractant. This phenomenon has been
leukotriene B4 (LTB4, a product of stimulated termed heterologous desensitization, and the
phospholipid metabolism in inflammatory cells), chemoattractant, C5a, is an especially competent
and the chemokine, interleukin-8 (IL-8), are the effector of this form of receptor downregulation.
most likely contributors to the chemotaxis of This process is in contrast to homologous desensi-
neutrophils in acute gout [197]. Although the tization which occurs when a ligand desensitizes
sentinel neutrophil chemoattractant in gouty its own receptor. A full discussion of desensiti-
arthritis has not been positively identified, IL-8 zation is provided subsequently since this pro-
has been characterized as the principal neutrophil cess may contribute to the resolution of uric acid
chemoattractant in acute gout. Further support crystal-induced inflammation. Finally, chemoat-
for the role of certain chemoattractants has been tractant receptors stimulate a variety of differ-
derived from the detection of IL-8 and LT84 in ent cellular functions such as degranulation and
the synovial fluid of patients with acute gout. release of lysosomal enzymes, upregulation of
Although scientific rationales can be cited to sug- adhesion molecules, and initiation of a respiratory
gest that other neutrophil chemoattractants might burst. An example of these differential effects of
be involved in acute gout, no other chemotaxins chemoattractants is illustrated by the fact that the
have been measured in the synovial effusions in neutrophil respiratory burst is only dependent on
acute gouty arthritis. the IL-8 receptor, CXCR-1, whereas the intracel-
The availability of many different chemoat- lular elevation of calcium and the phosphorylation
tractants (LTB4, IL-8, C5a, PAF, and perhaps oth- of mitogen-activated protein kinase are mediated
ers) raises the question as to why such redundancy by both IL-8 receptors (CXCR-1 and CXCR-2).
exists. Different explanations have been offered to These latter receptor-mediated effects regulate or
justify this redundancy. First, different chemoat- trigger specific cellular functions [198].
tractants arise from different cellular sources
and the redundancy of these agents assures that
no matter what tissue is affected, the recruitment Proteinases and Neutrophil Migration
of neutrophils to the inflammatory site will be
effective and prompt. Second, IL-8 is secreted Even though the effect of proteinases on neu-
by many different tissues and inflammatory cells trophil migration remains controversial, there
after such cells and/or tissues are activated by is some evidence that the neutrophil granule
proinflammatory stimuli. This may explain the enzymes such as elastase (present in azurophil
principal role of this chemokine in gout since granules), gelatinase (present in specific and
different cells associated with the synovial mem- gelatinase granules), and cathepsin G (present
brane and joint space fluid are able to secrete this in azurophil granules) play a role in the regu-
chemoattractant agent in response to monoso- lation of neutrophil migration in a variety of
dium urate crystals. Third, neutrophil chemoat- ways. Elastase regulates the quantity of the vita-
tractants can integrate different chemoattractants min d-binding protein (DBP) or Gc-globulin, a
in a variety of ways including responses to the multifunctional 56-kDa protein that enhances
vector sum of the orienting signals, prioritizing C5a chemotactic activity. Elastase performs this
chemoattractant signals based on the memory function by mediating the shedding of the DBP
of their signals, or by agonist combinations to binding site, a cell-surface chondroitin sulfate
220 8 Mechanisms of the Acute Attack of Gout and Its Resolution

proteoglycan. Cathepsin G or elastase result in the the resolution of the acute gouty episode. Thus,
proteolysis of the P-selectin glycoprotein ligand-1 an elevated concentration of C5a in the synovial
(PSGL-1) and causes the downregulation of neu- fluid of resolving gouty arthritis might provide
trophil adhesion to P-selectin. The proteolysis of evidence of the role of this chemoattractant in the
P-selectin counterreceptor decreases adhesion of modification of the acute episode.
the neutrophil and, subsequently, enhances the
migration of these inflammatory cells to the site
of tissue injury. The role of various proteinases in The Complement Fragments:
the migration of neutrophils has also been inves- C5a and C5a des Arg
tigated using proteinase inhibitors. These studies
demonstrate a reduction in neutrophil chemotaxis Human C5a is a cationic glycoprotein containing
and/or migration in the presence of proteinase 74 amino acids with a carbohydrate moiety
inhibitors. In contrast to these findings and the attached to an asparagine residue shown in bold
absence of direct evidence for the role of protei- in the amino acid structure. Its amino acid
nases in neutrophil migration, elastase-deficient sequence is shown below. The carbohydrate moi-
mice clearly demonstrate that neither neutrophil ety is an oligosaccharide containing glucosamine,
elastase, matrix metalloproteinase-9, nor cathep- sialic acid, mannose, and galactose.
sin G are essential for neutrophil transendothelial Thr-Leu-Gin-Lys-Lys-lle-Giu-Giu-lle-
migration [199]. These data may indicate differ- Aia-Aia-Lys-Tyr-Lys-His-Ser-Vai-Val-
ences between the human and murine systems or Lys-Lys-Cys-Cys-Tyr-Asp-Giy-Aia-
the lack of identification of the true proteinases Cys-Vai-Asn-Asn-Asp-Giu-Thr-
supporting neutrophil chemotaxis and migra- Cys·Giu-Gln-Arg-Aia-Aia-Arg-lle-
tion. Thus, the cellular sources of chemoattrac- Ser-Leu-Giy-Pro-Arg-Cys-lle-Lys-
tants, their role in biological functions other than Aia-Phe-Thr-Giu-Cys-Cys-Vai-Vai-
chemotaxis, and their effect on chemoattractant Aia-Ser-Gin-Leu-Arg-Aia-Asn-lle-
receptors are factors that must be considered in Ser-His-Lys-Asp-Met-Gin-Leu-Giy-
assessing neutrophil chemoattraction. Arg
Since the chemoattractants C5a, LTB4, and C5a des Arg has a structure identical to C5a
IL-8, and perhaps PAF, are likely participants except for the absence of a carboxy-terminal
in neutrophil chemotaxis in gout, each of these arginyl residue. This modified C5a molecule is
mediators is discussed in greater detail here. generated by the action of carboxypeptidase on
Except for the investigations of the C5a recep- the native molecule. The neutrophil chemotactic
tor and heterologous receptor desensitization by activity of C5a has been documented in several
C5a, little has been published concerning the rote experimental systems and has the capacity to
of the C5a chemotaxin in gout since IL-8 appears attract human neutrophils at concentrations as
to be the major chemoattractant in that disorder. low as 0.1 nM. C5a concentrations greater than
Nonetheless, since the generation of C5a is not 1 nM can cause desensitization of the C5a recep-
cell dependent, and since it induces the synthesis tor and decrease neutrophil chemotaxis. The C5a
and secretion of the proinflammatory cytokines, derivative, C5a des Arg, is formed through the
IL-1, IL-6, IL-8, and TNF, the activation of C5a action of serum carboxypeptidase N which con-
by monosodium urate crystals could be the early verts C5a to C5a des Arg. The latter metabolite is
reaction triggering the release of inflammatory much less potent as a neutrophil chemotaxin
mediators associated with acute gout [200]. (about 10–20 times less), but it shows compara-
Furthermore, the fact that C5a can regulate neu- ble neutrophil chemoattractant activity when
trophil chemotaxis via heterologous receptor combined with its cochemotaxin, Gc-globulin or
desensitization may provide this complement fac- vitamin d-binding protein [201]. C5a also trig-
tor with the capacity to modify neutrophil migra- gers actions other than neutrophil chemotaxis
tion to inflammatory sites and may be involved in such as the secretion of lysosomal enzymes,
Neutrophil Chemoattractants 221

eicosanoids, PAF, and IL-6, the increased contains an extracellular amino terminal domain,
expression of cell adhesion molecules, and the seven transmembrane-spanning domains, and an
increased production of superoxide anion. intracellular carboxy-terminal domain for the
Monosodium urate crystals are known to acti- coupling of a G protein (a heterotrimeric GTP-
vate both the classical and alternative complement binding or hydrolyzing protein). The seven trans-
pathway. These crystals mediate, either in a membrane-spanning domains, because of their
C-reactive protein-dependent or in an IgG- coiled appearance, have led to the use of the term
dependent manner, the depletion of plasma com- “serpentine receptor” since the G protein-coupled
plement with the production of many polypeptides receptors resemble a coiled snake. When C5a, the
including C1r, C1s, fibronectin, C1q, fibrinogen, receptor ligand, binds to C5aR, it causes a change
lysosomal enzymes, IgG, and apoproteins. What in the receptor conformation that results in the
part the 74 amino acid protein C5a, a neutrophil activation of C5aR and catalyzes the exchange of
chemoattractant, plays in the neutrophil emigra- guanosine triphosphate (GTP) for guanosine
tion in acute gout remains incompletely resolved diphosphate (GDP) on the alpha subunit of the
and somewhat controversial. The levels of com- heterotrimeric (a, b, and g) protein. This interac-
plement in the joint fluid from patients with acute tion causes the dissociation of the a subunit from
gout have been found to be normal using a hemo- the bg dimer, and the free bg dimer then activates
lytic assay for the measurement of complement. the effector channels for the transduction of
Further, heating urate crystals to temperatures the C5a messages necessary for the stimulation
where their capacity to activate complement is lost of chemotaxis, granule enzyme release, and
has no effect on the ability of such heated crystals superoxide anion production. C5a triggers the
to initiate an inflammatory response. Additional initiation of a number of intracellular processes
evidence that complement fragments may not play including the activation of phospholipase C and
a central role in the mediation of the inflammatory phosphatidylinositol 3-kinase as well as the
response to urate crystals has been generated from secretion of TNF-a, IL-1, and IL-8. The human
animal experiments. Cobra venom factor, a sub- receptor for C5a has been cloned and sequenced
stance known to deplete complement components [197, 198]. The eDNA sequence for the C5a
when injected into joints, has no effect on the leu- receptor encodes a structure with seven trans-
kocyte response to urate crystals injected into the membrane domains, a long intracellular carboxy-
same joints. Whether the initial chemotactic factor terminus, three cytoplasmic loops, an extracellular
production results from the activation of the syn- amino terminus, and three extracellular loops.
ovial fluid complement cascade with the genera- Additional structural and functional characteris-
tion of C5a, the synthesis and release of LTB4 from tics of this receptor can be found on the website
synovial lining cells, the release of a-chemokines for G protein-coupled receptors (www.gpcs.
by the synovial lining cells, or a combination of org/7tm/). Recent evidence has documented the
these and other events remains to be firmly estab- role of the NH2-terminal domain to the binding
lished. Nonetheless, the process of neutrophil of C5a to C5aR, but work continues to under-
chemotaxis is likely to be amplified once neutro- stand fully the binding of C5a to its receptor and
phils arrive in the synovial cavity. In addition, C5 the transduction of its message [202]. Studies of
can be converted to the potent chemotaxin, C5a, or this receptor have identified some binding, acti-
its less potent derivative, C5a-des-arg, by C5 con- vation, and transduction sites. Tyrosine-sulfated
vertase formed on the urate crystal surface. residues at position 11 and 14 of the NH2-
terminus of the receptor have been shown to
mediate the initial docking of C5a to its receptor
C5a Receptor (C5aR) but appear not to mediate its receptor activation
site. This sulfation reaction occurs as a posttrans-
The C5a receptor belongs to the large family lational modification of C5aR and is essential for
of human G protein-coupled receptors, and it C5a binding to C5aR. The gene for the C5aR has
222 8 Mechanisms of the Acute Attack of Gout and Its Resolution

been localized to the chromosome site, 19q13.3- Platelet-activating factor


q13.4. The C5aR has a protein structure contain-
ing 350 amino acids and has a molecular weight
H3C—O—(CH2)15—CH3
of 39,320. About 50,000–100,000 C5a molecules O
bind per cell with a Kd of approximately 2 nM.
Although C5a has been shown to play an H3C-C—O.CH O
important role in some forms of inflammatory
arthritis, its role in gout has not been fully evalu-
H2C—O –P—O—CH2—CH2—N(CH3)3
ated [203]. Recent studies have investigated the
presence of the C5a receptor on human synovio-
O-
cytes. In synoviocytes recovered from patients
with rheumatoid arthritis and osteoarthritis, Fig. 8.3 The structure of PAF (1-O-alkyl-2-acetyl-sn-
mRNA for C5aR has been identified and its glycero-3-phosphocholine) usually contains hexadecanol
expression determined to be on the surface of in the sn-1 position, and changing the length of this fatty
alcohol diminishes the potency of the molecule. The ace-
synoviocytes from synovial membranes of
tate at position sn-2 is essential for PAF activity since its
patients with osteoarthritis and rheumatoid arthri- removal abolishes the biologic activity of the molecule.
tis. When C5a and lipopolysaccharide are added The polar head group at position sn-3 is also critical for
to cultured synoviocytes, tumor necrosis factor- the function of the molecule since its alteration decreases
the potency of the molecule. The actual stereochemistry
alpha is released. These studies merit a reexami-
for binding to PAF receptors is not illustrated in this
nation of the role of C5a and its receptor in acute diagram
inflammatory forms of arthritis like gout.

factor-like molecules are derived from oxidized


Platelet-Activating Factor (PAF) low-density lipoproteins [204]. Although the
role of these oxidized lipoproteins has not been
Platelet-activating factor (1-ortho-alkyl-2-acetyl- explored in gout or other inflammatory synovit-
sn-glycero-3-phosphocholine) and leukotriene ides, low-density lipoproteins and other lipopro-
84 (LTB4) are the two lipid chemoattractants teins have been isolated from the synovial fluid
for neutrophils. Although other fatty alcohols in some articular diseases. Since lipoproteins
may occur in the sn-1 position of PAF, the major are altered in some patients with gouty arthritis
metabolite in humans is constructed with a hexa- and oxidized low-density lipoproteins induce
decanol in the sn-1 position. The molecular 5-lipoxygenase activity in leukocytes, this mech-
structure of PAF is as shown below (Fig. 8.3). anism for the production of monocyte chemoat-
Changes in the ether linkage of alkyl group at tractant protein-1 as well as LTB4 may contribute
the sn-1 position of the glycerol backbone, the to the generation of these proinflammatory medi-
short chain acyl residue at the sn-2 position, or ators in acute gout. Further, these oxidized, PAF-
the phosphocholine head group at the sn-3 posi- Iike molecules stimulate monocytes, leukocytes,
tion, all reduce the biological potency of the and platelets; cause monocyte and neutrophil
native molecule. PAF and receptors for this lipid chemotaxis; and enhance leukocyte adhesion to
have been identified by several groups. Other endothelium [205]. Like authentic PAF, these
chemical substances related to PAF have also oxidized lipoproteins act via leukocyte PAF
been isolated including a molecule identical to receptor. PAF may also be referred to as AGEPC
the hexadecanol derivative but containing alkyl (alkyl glycerol ether phosphoryl choline) or PAF-
chains of different lengths in the sn-1 position. acether in the literature, but PAF is the term most
Investigations have detected PAF species with widely accepted.
alkyl derivatives including 17:0, 18:0, and 18:1 In support of the fact that PAF may have a role
groups in the sn-1 position. In addition to these in the pathogenesis of acute gout, PAF has been
cell products, inflammatory platelet-activating identified as a constituent of synovial fluid
Neutrophil Chemoattractants 223

leukocytes and some inflammatory joint diseases phosphocholine (lyso-PAF). Usually, this first
in animal models [205]. step is designated as phospholipase A2 acting on
Furthermore, PAF has been recovered in the 1-0-alkyl-2-arachidonoyl glycerophosphocholine
synovial fluid from inflammatory arthritides. The to form lyso-PAF and free arachidonate. The lat-
most compelling evidence related to the associa- ter compound can then be converted to various
tion between gout and PAF comes from an ani- arachidonate metabolites. The second step in the
mal models of acute uric acid crystal-induced remodeling pathway is catalyzed by the enzyme,
arthritis that has been evaluated using uric acid acetylcoenzyme A: lyso-PAF acetyltransferase.
crystals and an inhibitor of the PAF receptor. This enzyme catalyzes the addition of an acetyl
Pretreatment with a PAF receptor antagonist group to the sn-2 position of lyso-PAF to form
before the injection of urate crystals significantly 1-0-alkyl-2-acetyl-sn-glycero-3-phosphocholine
reduced the following inflammatory parameters or PAF. The enzymatic activity of both cytosolic
as compared to controls (no antagonist present): phospholipase A2 and lyso-PAF-acetyltrans-
synovial fluid volume, the number of cells in the ferase is regulated by phosphorylation-dephos-
synovial fluid and synovial membrane, and the phorylation reactions [207]. The cytosolic
concentration of PGE2 and IL-6 in synovial fluid. phospholipase A2 is regulated by a G protein-
Another interesting facet that could relate to syn- coupled pathway that causes the activation of
ovial joint inflammation in gout is the synthesis protein kinase C and subsequently, the phospho-
of PAF by monocyte-derived macrophages when rylation of PLA2 by mitogen-activated protein
stimulated by serum-opsonized zymosan. From kinase.
such data, one might infer that monocyte-derived Cytosolic phospholipase A2 activity is asso-
synoviocytes could phagocytose uric acid crys- ciated with increments in intracellular calcium
tals and produce PAF. These investigations pro- concentrations and by phosphorylation of mul-
vide a rationale for the discussion of PAF as tiple serines in the enzyme. The calcium ions
a putative mediator in the pathogenesis of acute interact with the amino terminal C2-domain of
gout. The principal sources of PAF are neutro- the cytosolic phospholipase A2 and phosphory-
phils, monocytes, eosinophils, and endothelial lation may occur at serine-505, serine-515, and/
cells. In some cases, PAF is not released from its or serine-727. The phosphorylation of serine-505
cell source but remains on the plasma membrane is mediated by mitogen-activated protein kinases
of the cell [206]. This is an especially prominent (MAPK), phosphorylation of serine-515 by cal-
state in endothelial cells since cell surface PAF cium-/calmodulin-dependent protein kinase II
signals neutrophil adhesion. The quantity of (CAMK II), and phosphorylation of serine-727
membrane-associated PAF is dependent on the by MAPK-interacting kinase Mnk1 or another
experimental conditions. related kinase [208]. The mechanism by which
Platelet-activating factor is not stored in cells cytosolic phospholipase A2a increases its
and is synthesized only in activated cells. Two enzyme activity remains unresolved, but a con-
pathways can lead to the synthesis of PAF: the formational change in the enzyme has been sug-
remodeling pathway and the de novo pathway gested [209]. In the neutrophil, it has been clearly
(Figs. 8.4, 8.5, and 8.6). The former pathway is shown that p38 mitogen-activated protein kinase
well characterized, accounts for the generation of phosphorylates acetyltransferase and increases its
PAF for immune and inflammatory responses, enzymatic activity. A coenzyme A-independent
and is the major biosynthetic pathway for PAF transacylase also plays a role in PAF synthesis
synthesis in neutrophils, endothelial cells, and [210]. This pathway causes the generation of free
monocytes/macrophages. The first step in the arachidonate and lyso-phosphatidylethanolamine
remodeling pathway is catalyzed by a cytosolic from the PLA2 substrate, arachidonate-contain-
phospholipase A2 that hydrolyzes the sn-2 fatty ing plasmalogen phosphatidylethanolamine.
acid from alkyl-choline phosphoglycerides to Lyso-phosphatidylethanolamine then acts as an
form an intermediate, 1–0-alkyl-sn-glycero-3- acceptor of the free arachidonate derived from the
224 8 Mechanisms of the Acute Attack of Gout and Its Resolution

Fig. 8.4 The synthesis Remodeling pathway A


of platelet-activating factor
(PAF) by the remodeling
pathway A occurs in two
steps. The first step is H2C –O -CH2 —R
catalyzed by phospholipase
A2 which hydrolyzes the sn-2 20:4 — CH O
fatty acid from alkyl-choline
phosphoglycerides to form +
H2C — O — PH —CH2 —N(CH2)2
an intermediate, 1-O-alkyl-
sn-glycero-3-phosphocholine
(lyso-PAF). The second step O-
is catalyzed by the enzyme,
acetylcoenzyme A: lyso-PAF 1-0-alkyl-2-arachidonyl glycerophosphocholine
acetyltransferase, which
converts lyso-PAF to PAF
(1-O-alkyl-2-acetyl-sn- Phospholipase A2
glycero-3-phosphocholine).
The degradation of PAF is
catalyzed by an intracellular
PAF acetylhydrolase that Arachidonic acid + H2C —— O —CH2 — CHR
converts PAF to lyso-PAF.
The lyso-PAF molecule is
then reacylated with a long HO—CH O
chain fatty acid by the
activity of an acyltransferase. +
H2C — O —P—CH2 —N(CH3)3
Thus, the remodeling
pathway designated as
pathway A results in PAF O-
synthesis by the activity
of phospholipase A2 Lyso-platele-activating factor
catalyzing the conversion
of 1-O-alkyl-2-arachidonoyl +
glycerophosphocholine to ACETYL CoA
lyso-PAF and free
arachidonic acid. The latter
serves as a substrate for
eicosanoid production. The Lyso-PAF-acetyltransferase
lyso-PAF is then acetylated
through the activity of a
specific enzyme, lyso-PAF
acetyltransferase, and acetyl
coenzyme A donates the
acetyl group. The enzyme, O
lyso-PAF is the rate-limiting H2C — O —CH2 —CHR
enzyme for the pathway
H3C — C— O——— CH O

+
H2C — O —— P —— CH2 —— N(CH3)3

O-
Platelet-activating factor
Neutrophil Chemoattractants 225

Remodeling pathway B

H2C – O — CH CHR H2C — o —CH2R

HO— CH 20:4 — CH
O
+
H2C — O — P — O — CH2CH2NH2 H2C — O — P -CH2CH2N(CH3)3

O- O-

PLA2
Coenzyme A-indpendent transacylase (CoA-IT) PLA2

H2C — O — CH CHR
H2C — O — CH2R

20:4 — CH O
HO— CH
O
H2C — O — P — O — CH2CH2NH2 +
H2C — O — P — O — CH2CH2N(CH3)3

O-
O-

Lyso-PAF-acetyltransferase

O H2C — O — CH2R

CH3— C—— O— CH
O

+
H3C — O — P — O — CH2CH2N(CH3)3

O-

Fig. 8.5 The other remodeling pathway B uses phos- by the activity of the enzyme, lyso-PAF acetyltransferase.
phatidylethanolamine as the source of arachidonic acid by The major storage compound for arachidonate in human
phospholipase A2 hydrolysis rather than phosphatidylcho- neutrophils is the ethanolamine plasmalogen, 1-O-alk-1’-
line. The increased concentrations of 1-O-alk-1¢-enyl-2- enyl-2-acyl-sn-glycero-3-phosphoethanolamine.
lyso-sn-glycero-3-phosphoethanolamine and the presence Stimulated neutrophils trigger phospholipase A2 activity
of the enzyme, coenzyme A-independent transacylase and cause the hydrolysis of glycerol-3-phosphoethano-
(CoA-IT) provides evidence for the hydrolysis of arachi- lamines containing arachidonic acid. The products of this
donate-containing plasmalogen phosphatidylethanolamine hydrolysis are alkenyl-lyso-GPE and free arachidonic
by phospholipase A2 and the stimulation by lyso-phos- acid. Alkenyl-lyso-GPE is a substrate for arachidonate
phatidylethanolamine of the transfer of arachidonate from derived from 1-O-arachidonoyl-GPC. This reaction is
phosphatidylcholine to lysophosphatidylethanolamine catalyzed by CoA-independent transacylase and leads to
catalyzed by the transacylase enzyme with the formation the formation of lyso-PAF which is then converted to PAF
of lyso-PAF. The latter molecule is then converted to PAF by the enzyme acetyl-CoA:lyso-PAF-acetyltransferase
226 8 Mechanisms of the Acute Attack of Gout and Its Resolution

De novo pathway

H2C — OR O

HO — CH + CH2C — SCoA

H2C — O — P — OH

O-
1-Alkyl-2-lyso-sn-glycero-3-phosphate Acetyl-CoA

De novo acetyltransferase
H2C — O — R
O

+ CoASH
CH3C—— O —CH

H2C—— O —P

1-Alkyl-2-acetyl-sn-glycero-3-phosphate Coenzyme A

Phosphohydrolase

H2C —— O— R
O
H2PO4
CH3C — O—— CH +

H2COH

1-Alkyl-2-acetyl-sn-glycerol Phosphate

Cholinephosphotransferase
+

+
CDP-CH2CH2N(CH3)3

PAF

+ CDP

Fig. 8.6 The de novo biosynthetic pathway primarily has its cofactor, acetyl CoA, is converted to 1-O-alkyl-2-
been characterized as a less significant source of PAF than acetyl-sn-glycero-3-phosphate and coenzyme A by the
the remodeling pathways. It is found to operate in the enzyme, de novo alkylglycerophosphate acetyltransferase.
kidney and central nervous system. The immediate pre- In the second step, 1-O-alkyl-2-acetyl-sn-glycero-3-
cursors of PAF in this pathway are the 1-akyl-2-acetyl-sn- phosphate is converted to 1-O-alkyl-2-acetyl-sn-glycerol
glycerols that arise from the acetylation-dephosphorylation and phosphate by the enzyme alkylacetylglycero-P phos-
reactions catalyzed by acetylCoA:1-alkyl-2-lyso-sn-glyc- phohydrolase. The last step is the conversion of 1-O-alkyl-
ero-3-phosphate acetyltransferase and by 1-alkyl-2-acetyl- 2-acetyl-sn-glycerol and cytidine diphosphocholine to
sn-glycero-3-phosphate phosphohydrolase. In this de novo PAF and phosphate catalyzed by the enzyme CDP-
pathway, 1-O-alkyl-2-acetyl-sn-glycero-3-phosphate with choline:alkylacetylglycerol cholinephosphotransferase
PAF Receptor 227

PLA2 substrate, 1–0-alkyl-2-arachidonoyl glyc- and activate b2 integrins than it is as an effector


erophosphocholine and catalyzed by the enzyme, of chemotaxis, degranulation, or reactive oxygen
coenzyme A-independent transacylase (CoA-IT). generation. PAF concentrations necessary for
Both these biosynthetic pathways require phos- neutrophil degranulation and oxygen generation
pholipase A2. The de novo pathway is utilized are usually in the range of 100 nM, whereas con-
mainly in the kidney and brain. This pathway centrations for neutrophil priming, chemotaxis,
consists of three steps. Initially, 1-0-alkyl- and b2 integrin activation are in the range of the
sn-glycero-3-phosphate is acetylated by the Kd for the high-affinity receptor for PAF (0.2–1.0
enzyme, 1-0-alkyl-sn-glycero-3-phosphate:acetyl nM). PAF, as wm be discussed subsequently,
CoA:acetyltransferase. The product of this reac- primes neutrophils for enhanced superoxide gen-
tion is then transformed by a specific phospho- eration when such primed cells are stimulated by
hydrolase to 1-0-alkyl-2-acetyl-sn-glycerol. The other agonists. PAF also primes neutrophils for
latter metabolite is then converted by a dithiothre- aggregation, granule enzyme release, and actin
itol-insensitive CDP-cholinephosphotransferase polymerization, and it also stimulates the synthe-
to PAF. In contrast to the remodeling pathway, sis of the chemoattractant, LTB4, and stimulates
the regulation of this de novo pathway is only its own (PAF) synthesis in neutrophils [211, 212].
dependent on the availability of substrates. The link between the action of PAF on arachi-
donic acid metabolism and the production of the
potent chemoattractant, LTB4, demonstrates the
PAF Receptor redundancy in the generation of chemotactic
agents. PAF is probably most efficient as a medi-
Like the C5a receptor, the PAF receptor is a G ator of cell adhesion-dependent functions. LFA-1
protein-coupled receptor containing seven trans- (lymphocyte function antigen) and MAC-1, the
membrane domains, three extracellular loops, an integrin adhesive molecules enhancing the bind-
extracellular amino terminus, and an intracellular ing of neutrophils to endothelial cells are upregu-
carboxy-terminus. The human gene for this lated by PAF [213–215]. It also mediates
receptor has been cloned, and it encodes a protein neutrophil aggregation and adhesion to specific
containing 342 amino acids and has a molecular matrix proteins [216]. The increment in the sur-
weight of 39,200. The nucleotide sequence for face expression of these PAF-induced integrins is
this gene has been mapped to human chromo- caused by the recruitment of integrins via neutro-
some 1p35-p34.3. Extensive investigations con- phil exocytosis. To reemphasize the interdepen-
tinue to identify and characterize the ligand dencies that occur in the inflammatory response
binding sites for PAF responsible for desensitiza- has been shown to potentiate PAF-stimulated
tion. The effects of PAF receptor desensitization neutrophil degranulation .
will be discussed subsequently, but the complete Finally, PAF has been shown to induce neutro-
picture of the regulation and activation of this phil chemotaxis. It is clear from the investiga-
receptor remains to be completely characterized. tions cited and other studies that the C16:0
molecule is more potent than the C18:0 or C18:1
molecules. Neutrophil chemotaxis by PAF is
Biological Functions of PAF mediated specific receptors on the neutrophil
plasma membrane. These PAF-specific binding
PAF stimulates a variety of different functions in sites on neutrophils have been confirmed using
human neutrophils including !32 integrin activa- PAF-specific receptor antagonists. Since LTB4
tion, chemotaxis, degranulation, oxygen radical and interleukin-8 (IL-8) are also potent neutro-
generation, and changes in cell shape. PAF also phil chemoattractants and are found in the syn-
primes neutrophils to enhance the response of ovial fluid of patients with acute gout, it is
this cell to other agonists. PAF is more effective reasonable to compare the potency of LTB4, ll-8,
in its capacity to prime cells, change their shape, and PAF neutrophil chemoattractants [101].
228 8 Mechanisms of the Acute Attack of Gout and Its Resolution

The only published data comparing the potency PAF. Furthermore, PAF binding to its receptor
of these three chemoattractants comes from the activates phosphatidylinositol-specific phospho-
pediatric literature and uses newborn neutrophils lipase C that causes an increment in intracellular
as the target. These studies show a rank order of calcium and the formation of diacylglycerol. The
potency of lL-8 > lTB4 > PAF and combining increase in calcium causes the activation of pro-
these three chemoattractants increases chemot- tein kinase C and the catabolism of phosphatidyl-
axis over the average of each agent alone. The choline to form diacylglycerol and phosphatidic
latter increments were due to either the combina- acid [217]. Such phospholipid changes also lead
tion of IL-8 and PAF or LT84 and IL-8. Although to the production of arachidonic acid and the pro-
no data exist for the adult human neutrophil duction of proinflammatory arachidonate
responses to these chemoattractants, it is reason- metabolites.
able to assume that the rank order of potency for
adult neutrophils would be similar to newborn
neutrophils. Regulation of PAF: Acetylhydrolase
The significance of PAF to acute gouty arthri-
tis is its biological role as a mediator that increases To counter the activation of cytosolic PLA2
microvascular permeability, that stimulates the and lyso-PAF acetyltransferase and their genera-
adherence of neutrophils to endothelial cells and tion of the proinflammatory mediator, PAF, the
that may cause the chemotaxis and migration of rapid hydrolysis of PAF and PAF-Iike bioactive
neutrophils to endothelial cell monolayers. In lipids is catalyzed by PAF acetylhydrolases.
those endothelial cells stimulate to synthesize Thus, PAF acetylhydolases downregulate the
PAF, this mediator is transferred to the surface of proinflammatory signals generated by PAF and
the cell where it causes neutrophil adhesion and PAF-like lipids, and in a sense, this enzyme
triggers the induction of the activation-dependent serves as an anti-inflammatory agent. PAF acetyl-
upregulation of the adhesion molecules, b2 inte- hydolases with their anti-inflammatory properties
grins (CD11/CD18, CD11a/CD18, CD11b/ could serve as a modulator of acute gouty arthri-
CD18), in neutrophil plasma membrane and sub- tis [218]. Two forms of PAF acetylhydrolase
endothelial matrix. In addition to regulating these exist: plasma (secreted) PAF acetylhydrolase and
key leukocyte-endothelial cell interactions, PAF intracellular PAF acetylhydrolase. The principal
also stimulates the degranulation and generation sources of plasma PAF acetylhydrolases are mac-
of active oxygen metabolites from human neutro- rophage-containing tissues [218]. In addition, the
phils. These functional responses occur at con- differentiation of peripheral blood monocytes
centrations much higher than the subnanomolar into macrophages in culture causes an increase in
concentrations necessary for the induction of intracellular PAF acetylhydrolase activity, and
neutrophil adhesion to endothelial cells. Even such changes may have significance with respect
though PAF is a weak agonist for the generation to the monocyte-derived type A synoviocyte.
of oxygen radicals, it primes cells for other stim- During differentiation of monocytes and HL-60
ulants of this response. Thus, PAF plays two key cells, PAF acetylhydrolase is secreted. Thus, PAF
roles in its interaction with endothelial cells and may induce the synthesis and release of PAF
neutrophils: neutrophil chemotaxis and activa- acetylhydrolase and, in this way, serve as a feed-
tion of endothelial cell receptors for neutrophils. back mechanism to protect the host from exces-
Several additional facets are significant as to the sive PAF activity. The active site amino acids
generation and biological effects of PAF. Both critical for plasma PAF acetylhydrolase activity
tumor necrosis factor and interleukin-1a, two has been identified as Ser-273, Asp-296, and His-
potent proinflammatory mediators, have the 351. The human promoter region for plasma PAF
capacity to stimulate various cells to produce acetylhydrolase has also been identified and the
PAF. Thus, these mediators may augment the transcriptional activator for the gene character-
inflammatory process via their production of ized. Intracellular PAF acetylhydrolases also play
PAF Receptor 229

a regulatory role in the accumulation of PAF, involved in the increase in MCP-1 expression.
especially in macrophages. Several intracellular Another monocyte-related function of LTB4 is to
PAF acetylhydrolases have been cloned and char- trigger an increase in monocyte adhesion to
acterized [219]. At this time, some intracellular endothelial cells as a component of the recruit-
PAF acetylhydrolases appear to have activity ment of monocytes to inflammatory sites.
against both PAF and PAF-Iike molecules, Although some studies have reported an upregu-
whereas others are specific only for PAF. In sum- lation of surface b2-integrin expression on mono-
mary, PAF acetylhydrolases probably play a key cytes, recent investigations examining human
role in the expression and regulation of the bio- monocytes under flow conditions determined that
logical effects of PAF. This may be especially no change in the levels of b1- and b2- integrins
significant if the type A monocyte-derived occurred in response to LTB4. Under physiologi-
synoviocyte is shown to express and secrete cal flow conditions, LTB4 caused no change in
this enzyme. Furthermore, any effect of acety- integrin expression on human monocytes but did
lhydrolases might have on acute gouty arthritis cause an increased avidity of b1- and b2-integrin
and its resolution will be dependent on the role, for their counterligand. Such changes enhanced
if any, PAF plays in the propagation of acute the recruitment of monocytes.
gout. Of significance to gout, monosodium urate
crystals incubated with human neutrophils and
arachidonic acid produce LTB4. If such assays
Leukotriene B4 (LTB4) are dependent on endogenous supplies of arachi-
donic acid, neutrophils also produce LTB4 but at
Leukotriene B4 is a powerful neutrophil chemo- lower levels than when exogenous arachidonic
attractant derived from the lipoxygenation of acid is in the incubation [102]. The genera-
arachidonic acid. As its structure shows, LTB4 tion of LTB4 has been shown to result from the
belongs to the lipid class of chemoattractants. capacity of monosodium urate crystals to induce
Leukotriene B4 phospholipase A2 activity and the synthesis of
phospholipase A2-activating protein/5-lipoxyge-
OH OH
nase-activating protein (FLAP).
COOH A diversity of pathways are activated by LTB4
to regulate its biological functions. The genera-
tion of pseudopods and their extension on migrat-
ing cells is essential for their chemotactic activity,
and the rate of pseudopod extension is dependent
LTB4 Biological Functions on the LTB4 concentrations presented to the cell.
In addition to its role as a neutrophil chemoat- Such changes are regulated by the activation of
tractant and aggregator, LTB4 also causes mono- RhoA and its effector kinase, ROCK. These same
cyte adhesion to endothelial cells, neutrophil effects occur with other chemoattractants includ-
degranulation, cation fluxes, and the release of ing IL-8, C5a, PAF, and fMLP. The inhibition of
lysosomal enzymes and reactive oxygen species. pseudopod formation and extension is mediated,
A less well-characterized effect of LTB4 is the in part, by compounds that block phosphati-
induction of monocyte chemoattractant protein-1 dylinositol 3-kinase activity. The dependence of
(MCP-1) in human monocytes. MCP-1 induction the rate of pseudopod extension on the concentra-
by this mechanism is mediated via the LTB4 tion of a chemoattractant like LTB4 as well as
receptor, BLT1. This receptor-mediated effect other chemoattractants such as IL-8, PAF, C5a,
also requires the activation of the ERK1/2 or JNK and fMLP at equimolar concentrations suggests
mitogen-activated protein kinase to cause a dose- that the polymerization of cytoskeletal F-actin is
and time-dependent increment in MCP-1 tran- regulated by a common rate-limiting step or
scription. NF-kappa B expression may also be steps. LTB 4 also induces the release of human
230 8 Mechanisms of the Acute Attack of Gout and Its Resolution

neutrophil granules as measured by the release of receptor-mediated chemotaxis of neutrophils such


elastase. Elastase release is dependent on p38 as the presence of fMLP has no effect on LTB4
MAP kinase activity. Studies using neutrophils production. This feedback inhibition appears to
obtained from mice demonstrate that the release be a mechanism for limiting LTB4 production
of primary granules such as myeloperoxidase and and preventing the harmful effects of excessive
elastase are regulated by the Rho GTPase, Rac 2. LTB4. Finally, LTB4 activates other neutrophil
These observations have not been confirmed functions of significance to the inflammatory
using human neutrophils. LTB4 clearly activates response including the upregulation of the com-
human neutrophils, and at the same time, this plement receptor, the generation of superoxide,
chemoattractant delays apoptosis [220, 221]. and the increase in cell calcium levels [224]. In
Although the pathways for such activation and summary, the biological roles of LTB4 are criti-
apoptosis retardation remain to be completely cal for inflammatory reactions including acute
characterized, they appear to be regulated sepa- gout, and its function as a recruiter of monocytes
rately. A key role for neutrophils and mac- may point to its putative action in the resolution
rophages in gout is their capacity to phagocytose of acute gouty episodes.
immunoglobulin-coated uric acid crystals, and
LTB4 has been shown to augment FcyR-mediated
phagocytosis by both macrophages and neutro- LTB4 Biosynthesis via Phospholipase/
phils. These observations are supported by the Lipoxygenase and Its Release
fact that human neutrophils treated with different
leukotriene inhibitors exhibit a reduction in their Leukotriene B4 (5S,12R-dihydroxy-6,14-cis-8-
capacity for phagocytosis. LTB4 also activates ,10-trans-eicosatetraenoic acid), a potent lipid
the protein tyrosine kinase, Syk, a molecule mediator, requires for its synthesis the activation
essential for phagocytosis via the Fey receptor of cells with the release of arachidonic acid by
[222]. It accomplishes its actions by causing an the activity of cytosolic phospholipase A2 and
increment in intracellular calcium, and this acti- the subsequent activation of 5-lipoxygenase. 5-li-
vates a number of kinases including protein poxygenase in association with 5-lipoxygenase-
kinase C, mitogen-activated protein kinase, phos- activating protein (FLAP) converts arachidonic
phatidylinositol 3-kinase, and protein tyrosine acid into the unstable, reactive epoxide, LTA4.
kinase [223]. The unstable allylic epoxide intermediate, LTA4
Another property of LTB4 is its capacity to act is then hydrolyzed by the activity of LTA4 hydro-
as a feedback inhibitor of LTB4 under specific lase to form the dihydroxy acid, LTB4 [225].
conditions. Exogenous LTB4 inhibits the pro- Phospholipase A2 enzymes are the rate-limiting
duction of LTB4 by neutrophils when cells are steps for several pathways leading to the synthe-
exposed to the mediator prior to initiating the sis of biologically active lipids. These enzymes
phagocytosis of opsonized zymosan. It also catalyze the hydrolysis of the sn-2 position of
inhibits the synthesis and release of 5-hydroxy- membrane glycerophospholipids and lead to the
6,8,11,14-eicosatetraenoic acid as well as the release of fatty acids and lysophospholipids.
release of arachidonic acid by phagocytosing Among the fatty acids released by these enzymes,
neutrophils. Furthermore, exogenously deliv- arachidonic acid is an especially important
ered LTB4 metabolites, 20-hydroxy-LTB4 and product since it is subsequently converted enzy-
3(S)-hydroxy-LTB4, also reduce LTB4 synthe- matically to mediators like prostaglandins and
sis and release by neutrophils phagocytosing leukotrienes. Even the other products of phos-
opsonized zymosan. These inhibitory functions pholipid A2 activity, lysophospholipids, are
are all blocked when an LTB4 receptor inhibi- themselves biologically active and can be con-
tor is present. In addition, this reduction in the verted to the active proinflammatory mediator,
production of LTB4 by exogenous LTB4 and its platelet-activating factor. Specific phospholipases
metabolites appears to be specific since other may also be involved in the generation of
PAF Receptor 231

mediators that are keys to the resolution of acute the conversion of substrates to specific products.
inflammatory responses. Only LTA4, LTA 3, and LTA5 are substrates for
the enzyme, and the latter two compounds are
relatively poor substrates. These leukotriene
LTA4 Hydrolase epoxides inactivate leukotriene hydrolase during
the formation of LTB4 or the products of other
The final step in the biosynthesis of LTB4 is cata- substrates by a mechanism characterized as
lyzed by a zinc metalloenzyme with anion-depen- suicide inactivation. The latter mechanism is
dent aminopeptidase activity called LTA4 characterized in greater detail in the following
hydrolase. This enzyme has been cloned and citations. LTA4 hydrolase has dual catalytic
characterized from human sources. The gene for activities: epoxide hydrolase and aminopepti-
this approximately 69-kDa enzyme has been dase. The enzymatic catalytic sites are separate
mapped to chromosome 12q22. This gene con- but overlapping sites. As noted, the epoxide
tains 19 exons and has a promoter region upstream hydrolase transforms LTA4 to LTB 4, and the
of its transcription initiation site. In most studies, aminopeptidase cleaves a variety of peptides yet
LTA4 hydrolase has been isolated from the solu- its endogenous substrate(s) remain unknown.
ble protein fraction of cells, but one notable The active sites of these catalytic activities (epox-
exception is the alveolar macrophage. As is obvi- ide hydrolase and aminopeptidase) have been
ous, no data are available regarding the location identified and characterized.
of these enzymes in the synovial membrane mac-
rophage-like cells. In the alveolar macrophage,
despite the preponderance of LTA4 hydrolase in Transcellular Metabolism and LTB4
the nucleus, smaller quantities of this enzyme Production
exist in the cytoplasm. In the neutrophil, LTA4
hydrolase remains in the soluble fraction regard- Finally, LTA4 transcellular metabolism occurs in
less of the cell’s state of activity. Since LTA4 cells that do not contain the enzyme, 5-lipoxyge-
hydrolase is found in both the cytoplasm and nase, but do contain the widely distributed, LTA4
nucleus of alveolar macrophages, the idea that hydrolase, These data implicate the latter enzyme
LTA4 hydrolase migrates from the cytoplasm to as a significant regulator of the inflammatory
the nucleus may or may not be a reasonable response. The absence of 5-lipoxygenase and the
assumption. If that concept is reasonable, then presence of LTA4 hydrolase in endothelial cells
import sites must exist on the enzyme. It has been and platelets demonstrates the capacity of these
proposed that either basic amino acid clusters or cells to contribute LTB4, LTC4, and even lipoxins
armadillo repeats on the LTA4 hydrolase might be to neutrophil-dependent inflammation by using
responsible for the import of this enzyme to the the LT secreted by endothelial cells or platelets to
nucleus. The details of the import of LTA4 hydro- synthesize LTB 4 via LTA4 hydrolase. Of impor-
lase to the nucleus of macrophages such as those tance to the inflammatory reactions observed in
residents of the alveolus remain to be resolved, joint disease is the interaction between neutro-
and additional characterizations of the enzyme phils and endothelial cells. Neutrophils release
can be found in the following citations [226]. LTA4 at a level greater than 50 % of the total
A few characteristics of the enzyme bear quantity synthesized. This quantity of released
emphasis. First, the enzyme is a metalloenzyme LTA4 results in the additional formation of LTB4
that contains one zinc atom per enzyme molecule, and the omega-oxidized metabolites of LTB4,
and chelation of this metallic atom by 1,10- 20-hydroxy- and 20-carboxy-leukotriene B4.
phenanthroline generates an apoenzyme without Furthermore, such interactions may also result
activity. Second, the enzyme has a very selective in the synthesis of LTC4, LTD4, and LTE4.
substrate specificity since the double bond geom- Such transcellular metabolism between neutro-
etry of its substrates limits its capacity to catalyze phils and endothelial cells may contribute to the
232 8 Mechanisms of the Acute Attack of Gout and Its Resolution

maintenance of an elevated gradient of LTB4 studies also demonstrate the complexity of trans-
for the recruitment of additional neutrophils. location and the present inability to identify
LTB4 at high concentrations may also cause specific protein sequences responsible for this
the generation of superoxide, induce neutrophil translocation or lack thereof. The status of 15-li-
degranulation, and enhance neutrophil adher- poxygenase compartmentalization has not been
ence to vascular endothelium. Both LTC4 and determined in human macrophages. Recently, an
LTD4 significantly modify microvascular per- increased risk of myocardial infarction and stroke
meability. Such transcellular metabolism is not has been determined to be associated with a sin-
confined to the leukotrienes since other reactive gle-nucleotide polymorphism at the locus of the
intermediates may also be transferred and uti- arachidonate 5-lipoxygenase-activating protein
lized for eicosanoid biosynthesis. Thus, transcel- (ALOX5AP) gene at the chromosomal locus
lular metabolism represents another route for the 13q12-13 [227]. These patients with this poly-
host to generate LTB4 and other proinflammatory morphism produce increased quantities of LTB4.
mediators. In addition, transcellular metabolism, Since cardiovascular disease, gout, and hyperten-
as will be discussed subsequently, also leads to sion often coexist, this polymorphism might also
the synthesis of anti-inflammatory eicosanoids. identify patients with gout who may be at
Furthermore, it emphasizes the significance of increased risk for cardiovascular and cerebrovas-
the enzyme, LTA4 hydrolase, to the inflammatory cular events. Thus, both 5-lipoxygenase and leu-
process. This enzyme, because of its central role kotriene hydrolase activities are essential for an
in the generation of LTB4, also becomes a rea- acute inflammatory reaction and their inhibition
sonable target for inhibitors that block its bio- can modify such responses.
synthetic capacities. Recently, investigations of
the structure of leukotriene hydrolase complexed
to several different inhibitors have provided the Phospholipase Classification
basis for understanding inhibitor-leukotriene and Functions
hydrolase interactions and for designing inhibi-
tors based on such structural interactions. A vari- Since the release of arachidonic acid, the precur-
ety of potent inhibitors have now been developed sor of proinflammatory and anti-inflammatory
and tested for their specificity. mediators, is a key to the synthesis of these
A few additional concepts are worthy of com- mediators, the enzymes regulating the release of
ment here. The mechanism by which leukotriene this metabolite play a central role in the devel-
hydrolase is inactivated by its substrate and iso- opment of acute gout and its resolution. These
mers has been characterized. Such data may be enzymes are classified here and will be discussed
useful in the design of leukotriene inhibitors. subsequently for their role in shifting from an
Colchicine, a drug often used in the treatment of acute inflammatory response to the initiation of
acute gout, has been shown to block phospholi- the resolution phase of an inflammatory reac-
pase A2 activity and the processing of arachi- tion. The phospholipase A2 family of enzymes
donic acid by 5-lipoxygenase. This agent that includes plasma, secreted, and intracellular
disrupts microtubules may, via its alteration in forms of phospholipase with a unique substrate
arachidonic acid metabolism, contribute to its affinity for platelet-activating factor. Thus, the
anti-inflammatory effects. It is important to rec- latter phospholipases (PAF acetylhydrolases)
ognize that not all the lipoxygenase enzymes serve to downregulate the proinflammatory sig-
translocate to the nuclear envelope, and this doc- nals derived from PAF. Investigations into the
uments the fact that not all lipoxygenases are role of phospholipase A2 enzymes are ongoing,
localized to the same cellular compartments. In and at this time, this family of enzymes can be
the foregoing citation investigating a mouse mac- separated into three principal groups: cytosolic
rophage cell, RAW 267.4, 15-lipoxygenase was PLA2 (cPLA2), intracellular PLA2 (iPLA2), and
not translocated to the nuclear envelope. These secretory PLA2 (sPLA2) [228]. There are ten
PAF Receptor 233

secretory phospholipase A2 enzymes that have activation of cPLA2a is regulated by phosphory-


been identified at this time, and they have been lation. This phospholipase can be phosphorylated
classified into ten groups: group IB, group IIA, at Ser 505, Ser-515, or Ser-727. Phosphorylation
group IIC, group IID, group IIE, group IIF, group of Ser-505 is mediated by mitogen-activated pro-
III, group V, group X, and group XIIA [229]. At tein kinase, whereas calcium-/calmodulin-depen-
least four cytosolic PLA2s have been identified. dent protein kinase II catalyzes the phosphorylation
The four cytosolic enzymes have been termed of Ser-515. Cytosolic PLA2a may also be phos-
cPLA2a, cPLA2b, cPLA2g, and cPLA2d. These phorylated at Ser-727 by MNK1-related protein
phospholipases are classified as group IV phos- kinase. The basis for the phosphorylation of these
pholipases. Finally, the calcium-independent different serines is dependent on the cell type and
phospholipase A2 enzymes are classified as the stimulus, but phosphorylation increases the
group VI. The group VIA PLA2 in the human enzyme activity regardless of the mechanism of
has been mapped to chromosome 22q13.1, and phosphorylation. The amino acid residues critical
this gene encodes five splice variants classified for the catalytic activities of cPLA2a are Arg-
as group VIA-1 (GPVIA-1), group VIA-2 200, Ser-288, Asp-549, and Arg-566. These same
(GPVIA-2), group VIA-3 (GPVIA-3), group amino acids are conserved in all the cPLA2s.
VIA-ankyrin-1 (GPVIA-ankyrin-1), and group Further details relating to these cytosolic phos-
VIA-ankyrin-2 (GPVIA-ankyrin-2). Additional pholipases can be found in a recent review.
calcium-independent PLA2s continue to be char- Recently, cytosolic phospholipase A2g, although
acterized with functions related to energy mobi- it does not show a preference for arachidonic acid
lization and storage for the maintenance of lipid release, causes spontaneous and stimulus induced
homeostasis [230]. (ll-1) arachidonic acid release. Critical for its
Having characterized the various phospholi- activity is Ser-82 and its C-terminal prenylation
pases, the question remains as to what specific site. The arachidonate released by this phospholi-
functions these multiple enzymes carry out. This pase has been shown to produce COX-1 and
has been a difficult problem since most cells con- COX-2 products.
tain more than one PLA2 and the use of inhibi- Other studies have begun to explore the role of
tors and antisense molecules to determine their phospholipase A2s in phagocytosis. Using mouse
functions often lack specificity. The present dis- macrophage-like P38801 cells, it has been deter-
cussion will be limited to those phospholipases mined that zymosan-induced arachidonic acid
that are known to play an essential role in the release may be mediated, in part, by calcium-in-
activation and resolution of inflammatory pro- dependent phospholipase A2. It has been pro-
cesses like gout. It is also entirely possible that posed that this cPLA2 pathway is likely to be
additional phospholipases will be found to con- protein kinase (PK) dependent. Recently, the
tribute to this process in the future. At present, receptor for the Fc portion of the immunoglobu-
the group IV calcium-dependent cytosolic PLA2a lin molecule found on neutrophils, monocytes/
is the principal phospholipase with a preference macrophages, and other cells has been shown to
for the hydrolysis of glycerophospholipids con- couple with calcium-independent PLA2b in
taining an arachidonyl residue at their sn2 posi- U937 cells and to release arachidonic acid lead-
tion. This phospholipase is induced by cell stimuli ing to the subsequent generation of prostaglandin
and the resultant activation of such cells causes a E2 and leukotriene B4. The activation of iPLA2
calcium-dependent translocation of the enzyme in this setting was PKC dependent.
from the cytoplasm to perinuclear and nuclear Investigations have also focused on the role of
membranes. Such translocation places this phos- phospholipases in apoptosis, a cellular function
pholipase in the location of the arachidonic acid- active in the resolution of an acute gouty episode.
metabolizing enzymes like cylcooxygenases and A host of publications have addressed the inter-
lipoxygenases [231]. In addition to the control of action between apoptosis and phospholipases,
cPLA2a by increments in intracellular calcium, but the entire details of this process remain
234 8 Mechanisms of the Acute Attack of Gout and Its Resolution

incomplete. What is clear from the foregoing been detected at increased levels in some
citations is that the connections between phos- inflammatory disorders and in the synovial fluid
pholipases and apoptotic pathways are still under of some inflammatory arthritides [232]. Secretory
investigation, but it is reasonably well established PLA2-IIA, sPLA2-V, and sPLA2-X all have
that cytosolic phospholipase plays a role in some a deliterious effect on pulmonary surfactant.
of the apoptotic pathways. In general, TNFa- Group II phospholipase A2s also manifest potent
induced cell death appears to be mediated by bacteriocidal properties via their degradation of
cytoplasmic PLA2 since caspase-3 causes bacterial phospholipids. Despite the role of these
a cleavage of cPLA2 at Asp-525 producing an phospholipases in inflammatory reactions, few
inactive N-terminal cPLA2 fragment This inac- investigations of these enzymes have been con-
tive cPLA2 fragment localizes to the membrane ducted in patients with acute and resolving gout.
fraction of the cell and competes with native The design of specific inhibitors of these enzymes
cPLA2. It acts as a dominant negative mutant and may provide tools for regulating the acute
prevents A23187 and IL·1 from releasing arachi- inflammatory response in gout. Furthermore, the
donic acid in significant quantities. This cPLA2 key role both cPLA2 and iPLA2 play in the
fragment also selectively reduces the catalytic chemotaxis of monocytes to the monocyte
activity of other PLA2s such as sPLA2 IIA and chemoattractant protein-1 emphasize the impor-
iPLA2. Recent data also suggest that iPLA2 is tance of these enzymes to the resolution of acute
also cleaved by caspases at Asp-183. This iPLA2 gout [233]. Thus, phospholipase A2 activity not
fragment accelerates membrane phospholipid only provides arachidonic acid for further metab-
remodeling including the translocation of olism to the potent chemoattractant, LTB4, but
phosphatidylserine, an activity associated with also may play a key role in other proinflammatory
apoptosis. Further investigations are necessary to and anti-inflammatory responses essential for the
define the pathways relating PLA2s to the apop- propagation and resolution of an acute gouty
totic process and what signals trigger these episode.
events. Arachidonic acid released by PLA2 activity is
In summary, cPLA2 is the major enzyme then converted to the unstable epoxide, LTA4 by
responsible for the generation of arachidonic 5-lipoxygenase with the help of 5-lipoxygenase-
acid used for the synthesis of inflammatory activating protein (FLAP). 5-Lipoxygenase cata-
mediators like PGE2 and LTB4. The activity of lyzes the hydroperoxidation of arachidonic acid
cPLA2 IVA is regulated positively by increases to form 5-(S)-hydroperoxy-6,8,11,14-eicosatet-
in intracellular calcium and by serine phosphory- raenoic acid (5-HPETE) followed by a dehydrase
lation. The release of arachidonic acid during reaction to form the unstable epoxide, LTA4. It is
phagocytosis by cell lines appears to be regulated now clear that all the key enzymes in eicosanoid
by calcium-independent PLA2, and arachidonic biosynthesis (cPLA2, cyclooxygenase, and lipox-
acid release as well as the onset of apoptosis ygenase) are translocated to the nuclear envelope
require cPLA2 or iPLA2 depending on whether where eicosanoid biosynthesis occurs [234]. In
FAS or TNF-a are inducing apoptosis. Secretory addition to these enzymes, the 18-kDa mem-
PLA2s also have a diversity of functions includ- brane-bound protein, 5-lipoxygenase-activating
ing the production of lipid mediators such as protein (FLAP), required for 5-lipoxygenase
eicosanoids and lysophospholipids that contrib- activity and the in vivo formation of leukotrienes,
ute to the inflammatory process as well as tissue is also localized to the nuclear envelope. The lat-
repair and the removal of apoptotic cells. These ter site is where arachidonic acid metabolism
secretory PLA2 (sPLA2) enzymes are of low occurs. These studies demonstrate that FLAP
molecular weight (14–18 vs. 85 kDa for cPLA2s), remains localized to the nucleus in polymorpho-
require millimolar levels of calcium for their nuclear leukocytes, peripheral blood monocytes,
activity, are cysteine rich, and are globular in and alveolar macrophages, whereas the 5-lipoxy-
structure. This group of secretory PLA2s have genase, cytosolic phospholipase A2, prostaglandin
PAF Receptor 235

endoperoxide H synthase, and prostacyclin syn- Amino Acid Structure of CSa


thase require translocation to the nuclear enve- Thr-Leu-Gin-Lys-Lys-lle-Giu-Giu-lle-Aia
lope for the synthesis of their metabolites in Aia-Lys-Tyr-Lys-His-Ser-Vai-Vai-Lys-
activated cells. Investigations are now focused on LysCys-Cys-Tyr-Asp-Giy-Aia-Cys-
determining and characterizing the import and Vai-Asn-AsnAsp-Giu-Thr-Cys-Giu-
export sequences that regulate the translocation Gln-Arg-Aia-Aia-Arglle-Ser-Leu-
of these enzymes [235]. The identification of Giy-Pro-Arg-Cys-lle-Lys-AiaPhe-
these translocation signals, especially those con- Thr-Giu-Cys-Cys-Vai-Vai-Aia-Ser-
trolling import, might be useful if specific inhibi- GinLeu-Arg-Aia-Asn-lle-Ser-His-
tors could be designed to abrogate their capacity Lys-Asp-MetGln-Leu-Giy-Arg
to translocate. Although the mechanisms remain Interleukin-8 is a member of the a- and
incompletely resolved, recent investigations have b-chemokine superfamily. The a-chemokine
demonstrated that cyclic AMP-elevating agents family includes IL-8, GROa (growth-related
and protein kinase A activators inhibit leukot- oncogene a), GROb, GROg, NAP-2 (neutrophil-
riene biosynthesis and 5-lipoxygenase transloca- activating peptide-2), ENA-78 (epithelial cell-
tion in stimulated cells [236, 237]. Further studies derived neutrophil-activating peptide), GCP-2
related to enzyme translocation and the subse- (granulocyte chemotactic protein), PBP (plate-
quent synthesis of prostaglandins and leukot- let basic protein), CTAP-III (connective tissue
rienes may reveal ways in which the synthesis of activating protein-Ill), and bTG (beta thrombo-
these inflammatory mediators can be downregu- globulin). Some of these chemokines contain
lated by therapeutic agents. a unique amino acid triad (glutamic acid-leucine-
arginine) at their amino terminus preceding the
first cysteine residue of the primary structure.
Interleukin-8 (IL-8) This amino acid sequence is visualized in bold
type in the amino acid structure shown above.
Interleukin-8, proposed as the primary chemoat- Chemokine receptors with this ELR (Giu-Leu-
tractant in acute gout, is a 72-amino acid poly- Arg) motif induce the recruitment of human
peptide with sulfide bridges between cysteines 7 neutrophils in vivo. There are seven human
and 34 and between cysteines 9 and 50. Its amino chemokines with the ELR motif: interleukin-8,
acid sequence is quite different from the 74-amino growth-regulated oncogene a (GROa), growth-
acid peptide that comprises the other peptide regulated oncogene b (GROb), growth-regulated
chemoattractant, C5a. A comparison of these two oncogene g (GROg), neutrophil-activating pep-
polypeptide sequences is shown below. Of course, tide-2 (NAP-2), epithelial cell-derived neutrophil-
these polypeptide chemoattractants are not struc- activating peptide-78 (ENA-78), and granulocyte
turally related in any way to the lipid chemoat- chemoattractant protein-2 (GCP-2). This struc-
tractants, LTB4 and PAF. tural element termed the ELR motif indicates an
enhanced neutrophil binding capacity as well as
Amino Acid Structure of IL-8 the capacity to induce neutrophil migration and
Ser-Aia-Lys-Giu-Leu-Arg-Cys-Gin- angiogenesis [237, 238]. Some a-chemokines
Cys-llelys-Thr-Tyr-Ser-Lys-Pro- lack the ELR motif, and for this reason, they do
Phe-His-Pro-lysPhe-lle-Lys-Giu- not attract mature neutrophils. Such non-neutro-
Leu-Arg-Val-lle-Giu-SerGly-Pro- phil chemoattractants include platelet factor-4
His-Cys-Aia-Asn-Thr-Glu-lle-lle (PF-4), interferon-gamma (IFN-g)-inducible pro-
Vai-Lys-Leu-Ser-Asp-Giy-Arg-Giu- tein-10 (IP-10), monokine induced by interferon-
Leu-Cys gamma (MIG), stromal cell-derived factor-1
Leu-Asp-Pro-Lys-Giu-Asn-Trp-Vai-Gin-Arg (SDF-1), and interferon-inducible T cell alpha
Vai-Vai-Giu-Lys-Phe-Leu-Lys-Arg- chemoattractant (I-TAC). All the alpha- and beta-
Aia-GiuAsn-Ser chemokines have conserved cysteines that form
236 8 Mechanisms of the Acute Attack of Gout and Its Resolution

disulfide bonds. The a-chemokines induce both Cell Sources


neutrophil activation and migration [239]. The
alpha-chemokines are 8- to 10-kDa proteins with Interleukin-8 continues to be the most widely
the positions of the first two cysteines separated investigated alpha-chemokine and is a very
by an amino acid, and for this reason, they are potent neutrophil chemoattractant. A variety of
designated as CXC chemokines based on their cells can produce IL-8 including endothelial
cysteine spacing [239]. In the beta-chemokines, cells, fibroblasts, monocytes, macrophages,
the conserved cysteines are adjacent to each other some epithelial cells, mitogen-stimulated T cells,
and designated as CC chemokines. There are addi- and neutrophils. Synovial type B fibroblast-like
tional chemokines designated as C and CX3C, cells can also synthesize IL-8, and the type A
but they have no relevance to this discussion. In synovial cell derived from monocytes may also
modern terminology, the following nomenclature have the capacity to generate IL-8 as well since
has been used for these ligands: GROa = CXCL1, it is derived from monocytes [240]. The diversity
GR0b = CXCL2, GROg = CXCL3, ENA- of cells capable of IL-8 synthesis and release
78 = CXCL5, GCP-2 = CXCL6, NAP-2 = CXCL7, (macrophages, neutrophils, fibroblasts, and
and IL-8 = CXCL8. Of some interest to this dis- endothelial cells) provides a basis for speculat-
cussion are some of the CC chemokines that ing that one of the initial events in acute gout
relate to monocyte/macrophage activities such may be the production of IL-8 by the interaction
as monocyte chemoattractant protein (MCP)-1, between a monosodium urate crystal and one of
and macrophage inflammatory protein (MIP)-a these cells in the synovium or synovial fluid. It
and MIP-13. These ligands have been identified has also been shown that IL-8 occurs in a precur-
by the following nomenclature: CCL2 = MCP-1, sor form containing 77 amino acids rather than
CCL3 = MIP-1a, and CCL4 = MIP-1b. the mature, 72 amino acid form. Recent studies
The beta-chemokines attract monocytes as have documented the cleavage of the 77 amino
well as eosinophils, basophils, and lymphocytes acid form by human hepatic cathepsin L to form
and are potent inhibitors of angiogenesis. The the mature 72 amino acid form of IL-8 [241].
monocyte chemoattractants in the b-chemokine This converting enzyme is also the secretory
family may have a role in the late phases of product of human fibroblasts, and the mature
the acute gouty episode. The CC or beta- IL-8 form is a much more potent chemoattrac-
chemokines include the monocyte chemotactic tant then the 77 amino acid form. Animal studies
proteins (MCP-1, MCP-2, MCP-3, and MCP-4), have employed 99mTc-labeled interleukin-8 to
RANTES (regulated on activation normal T cell detect pulmonary infections and colitis. Such
expressed and secreted), HCC-1 (hemofiltrate techniques may be useful in detecting hidden
CC chemokine-1), eotaxin (eosinophil chemoat- sites of inflammation but lack the specificity for
tractant), MIP-1a (macrophage inflammatory differentiating bacterial versus crystal-induced
protein-1a), MIP-1. MIP-1g, TARC (thymus- inflammation.
and activation-related hemokine), 1–309, 6
chemokine (chemokine with six cysteines), MIP-
3b (macrophage inflammatory protein-3 ). MDC IL-8 Gene
(macrophage-derived hemokine), and MIP-3a/
LARC (liver- and activated-related chemokine)/ The gene that codes for IL-8 has been mapped to
Exodus-1. In addition to their chemoattractant chromosome 4q12-21. It contains four exons and
capacity for monocytes, CC chemokines are also three introns and has a structure similar to other
chemotaxins for T cells, B cells, dendritic cells, neutrophil chemoattractants like GRO and giP-10.
eosinophils, basophils, and natural killer cells. Both IL-1 and TNF, proinflammatory cytokines
None of the latter cells play a prominent part in present in inflamed joints, can induce the synthe-
acute gout. sis of IL-8 mRNA. In addition to inducing IL-8
PAF Receptor 237

mRNA, IL-1 appears to result in a more stable substitute for IL-8 activities when its action is
IL-8 mRNA than in its absence. An AU-rich blocked, and may have different potencies
sequence in the IL-8 gene contributes to the post- depending on many factors.
translational regulation of IL-8 and increased The most acceptable mechanism for the
quantities of these sequences in the 3¢-untrans- transendothelial emigration of neutrophils, if
lated regions of the gene result in lower levels of not via fenestrae, is for these cells to migrate
IL-8 mRNA [242]. between endothelial cells at the vascular-
endothelial (VE)-cadherin junctions. Both
in vivo and in vitro studies have demonstrated
IL-8, MSU Crystals, and Gout that antibodies against VE-cadherin increased
neutrophil transmigration. Neutrophils also
A body of in vitro and in vivo evidence exists to have the capacity to degrade VE-cadherin and
document the role of MSU crystals in the pro- 13-catenin, major proteins of these cell junc-
duction of IL-8. Peripheral blood monocytes and tions. Furthermore, elastase inhibitors block
cultured synovial lining cells (fibroblast-like) this degradation, and the presence of elastase
incubated in vitro with MSU crystals cause the inhibitors such as a 1-proteinase inhibitor can
release of IL-8. The IL-8 release from MSU block the activity of neutrophil elastase. In addi-
crystal-stimulated synovial fibroblast-like cells tion, neutrophil oxygen metabolites can inacti-
occurs in a dose- and time-dependent manner. vate this elastase inhibitor permitting elastase
IL-8 has also been measured in synovial fluid to induce the proteolysis of VE-cadherin junc-
aspirates recovered from patients with acute gout tions. More recent studies have shown that
and compared with aspirates from those with neutrophil elastase is bound to the neutrophil
uncomplicated osteoarthritis. The latter speci- membrane and is catalytically active but resis-
mens showed no leukocytosis in contrast to those tant to inhibition by circulating antiproteases.
aspirates from gouty subjects. Synovial fluid Additional evidence also demonstrates that neu-
gouty aspirates had mean IL-8 levels of 8.4 ng/ trophil migration is regulated by an endothelial
ml as compared to 1.5 ng/ml in the aspirates from cell-dependent process causing the phospho-
patients with osteoarthritis (p value = 0.006). The rylation of myosin light chain kinase with the
role of IL-8 has also been confirmed in animal resultant retraction of adjacent endothelial cells
models of MSU crystal-induced synovitis and and gap formation. These reactions are calcium-
cytokine responses of synovial cells [243]. IL-8 and calmodulin-dependent processes and are
has also been measured in the synovial fluid of inhibited by calcium chelation or calmodulin
other inflammatory forms of synovitis reinforc- inhibition.
ing its role in the generation of inflammatory In normal human synovium, two adhesion
responses. molecules that have a prominent role in the
There are no data regarding the role of other early steps of the transendothelial migration of
a-chemokine neutrophil chemoattractants such as neutrophils have been identified: selectins and
the ELR ± C-X-C chemokines other than IL-8 in integrins. Both so-called E-selectin expressed on
acute gouty arthritis. ENA-78 has been identified endothelial cells and intercellular adhesion mol-
in synovial tissues, but its role in gout has not ecule (ICAM-1) have been localized to synovial
been determined. Other a-chemokine neutrophil venules. E-selectin was a prominent component
chemoattractants have been found in synovial of the superficial venules, whereas ICAM-1 had
fluids and joint tissues from nongouty articular a more prominent expression on large venules
disorders, but their role in gout remains undefined located in the deeper layers of the synovium.
[244]. There is some evidence from in vitro stud- The biological role of these adhesion mole-
ies suggesting that the a-chemokines may have cules and others are discussed in greater detail
a coordinated effect on neutrophil migration, may subsequently.
238 8 Mechanisms of the Acute Attack of Gout and Its Resolution

Adhesion Molecules provides the setting for neutrophil transmigration


to the inflammatory site.
An Overview of Selectins and Integrins

The migration of neutrophils from the intravas- Selectin Structures


cular space to the intrasynovial space is essential
for the development of the inflammatory response The structure of all three selectins is similar.
associated with acute gouty arthritis. A complex Selectins function like lectins and bind to car-
set of molecular interactions moderated by adhe- bohydrate ligands. Each selectin molecule has
sive proteins and leukocyte activation regulate an NH2 extracellular terminus composed of
these initial events and the subsequent tight bind- 120 amino acids that is similar to a domain in
ing of neutrophils to the endothelium. The initial the C-type animallectins. This amino terminal
step in this process is the tethering and rolling domain is linked to an amino acid sequence of
of neutrophils along the surface of endothelial variable length (35–40 amino acids) that has been
cells lining the postcapillary venules. The selec- designated as the epidermal growth factor (EGF)
tin family of cell adhesion molecules and their domain due to its similarity to repeat sequences
glycoconjugated ligands initiates this rolling in the EGF molecule. These EGF domains con-
and tethering process. E (endothelium)-selectin tain six cysteines in positions comparable to
(CD62E) is expressed on activated endothe- those in EGF. Attached to this EGF domain are
lial cells, and P(platelet)-selectin (CD62P) is repetitive amino acid sequences (short consensus
expressed on platelets and endothelial cells. repeats-SCRs) that resemble complement regula-
L(leukocyte)-selectin (CD62L) is expressed on tory proteins and have been designated as com-
most leukocytes. P-selectin resides in storage plement binding (CB) elements. These repetitive
granules (a-granules) in platelets and endothelial elements in the CB domain are about 60 amino
cells (Weibei-Palade bodies) and are mobilized acids long, and the number of CB domains varies
to the cell surface by various secretagogues such depending on the selectin. For example, human
as thrombin, histamine, and others. E-selectin is L-selectin contains two such domains, whereas
induced by inflammatory cytokines such as TNF- human P-selectin has nine and E-selectin has
a, IL-113. and others. L-selectin is constitutively six such domains. The membrane region of each
expressed on myeloid cells and lymphocytes, and selectin is anchored by a short transmembrane
its main function is to participate in lymphocyte domain linked to a short cytoplasmic tail. The
recirculation and the migration of neutrophils structure of the cytoplasmic tail varies depend-
into inflamed tissue sites. The tethering of neu- ing on the selectin analyzed. The cytoplasmic
trophils to endothelial cell surfaces is regulated tail of human P-selectin contains 35 amino acids,
by interactions between P-selectin, E-selectin, whereas E-selectin contains 32 amino acids, and
and L-selectin and their respective ligands. L-selectin contains only 17 amino acids. Each
Neutrophils roll along the surface of endothe- selectin molecule has a separate binding part-
lial cells and recognize chemokines such as IL-8 ner or ligand with the construct of a glycopro-
on the endothelial cell surface. Recognition of tein. With the exception of E-selectin ligand-1
the chemokines, PAF, LTB 4 as well as selec- (ESL-1), the ligand for E-selectin, the two other
tins and their ligands by the rolling neutrophil selectin ligands are sialomucins or have a sialo-
triggers the activation of integrins, and binding mucin domain. E-selectin and P-selectin expres-
then occurs between leukocyte integrins and the sion require activation of endothelial cells.
immunoglobin superfamily to cause firm attach- Therefore, in the absence of proinflammatory
ment of neutrophils to the endothelium [245]. mediators, adhesion molecules on the endothe-
This cascade of molecular events (tethering, roll- lium are absent. The necessity for the induc-
ing, chemokine recognition, integrin activation, tion of endothelial activation before interactions
and firm neutrophil-endothelium attachment) between leukocytes and the blood vessel intima
Adhesion Molecules 239

protects the endothelium from attracting neutro- N-terminus of the PSGL-1 polypeptide. Further,
phils in the absence of inflammation. removing a short peptide (10 amino acids) from
Ligands or binding partners for selectins are the N-terminus of PSGL-1 disrupts the binding
carbohydrates that bind to the lectin domain of between the ligand and P-selectin. Other key
the selectins. Lectin receptors are different from components of PSGL-1 for binding to P-selectin
the usual agonist-receptor recognition site since have been identified by site-directed mutagene-
they represent triads composed of a receptor, sis of PSGL-1 or removal of the extracellular
a ligand, and a carrier. The ligands for selectins domain of PSGL-1 enzymatically using 0-sia-
are oligosaccharides, and the carriers are the scaf- lylglycoprotease. Recent publications suggest
folding on which these oligosaccharides are that PSGL-1 contributes significantly to neutro-
assembled and modified for their binding to the phil adhesions to vascular endothelium via all
lectin domains. The physiological ligands for the three selectins. The identification of PSGL-1 as
selectins are glycoproteins with attached oligo- a critical ligand for the tethering and rolling of
saccharides that are subject to posttranslational leukocytes for all three selectins has been char-
modifications. acterized only in mice, but the description of the
structural requirements of L-selectin binding to
neutrophils and their role in primary leukocyte-
P-Selectin Glycoprotein Ligand-1 endothelial interactions between circulating and
adherent leukocytes indicate that future prog-
P-selectin glycoprotein ligand-1(PSGL-1) is the ress will resolve the binding of ligands to their
high affinity counterligand for P-selectin and is selectins [246].
the principle ligand for P-selectin on human neu-
trophils and other leukocytes including T cells.
PSGL-1 is a membrane polypeptide localized to L-Selectin
neutrophil surface microvilli containing a serine-
threonine-proline-rich extracellular domain, a L-selectin, present on the microvillus-like projec-
transmembrane domain, and a cytoplasmic tail. tions on the surface of leukocytes, is constitu-
Leukocytes express a homodimer of two identi- tively expressed on myeloid cells, and the
cal 120 kDa subunits with a tyrosine sulfation tethering and rolling mediated by it is much more
motif on the N-terminus of the extracellular rapid than that directed by P- and E-selectin. This
domain and a distal polypeptide with 15 deca- tethering and rolling function of selectins through
meric sequence repeats. The serine and threonine contact with their ligands is a major area of con-
residues in the molecule are potential 0-glycosy- tinuing investigation aimed at understanding the
lation sites generated by glucosaminyltransferase physical and topographical forces required for
enzymes. PSGL-1 like other selectin ligands is a the association-dissociation reactions between
sialomucin and has been cloned from a human selectins and their ligands [247]. Since this inter-
HL-60 cell line. Even though L-selectin binding action only slows the rolling of leukocytes, it is
occurs initially, it is short-lived and P-selectin clear that the bonds between selectins and their
binding causes a slower rolling velocity and a counterligands must be only temporary and eas-
longer contact between neutrophils and endothe- ily broken. For the most part, selectin ligands
lial cells. Although sufficient evidence exists for have been characterized most completely in
the demonstration of E-selectin binding to murine cells, and it is unclear in every case what
PSGL-1, such interactions may be the result of the composition of the human homologues is.
leukocyte-leukocyte effects and secondary to Furthermore, little is known about the ligands for
tethering rather than a direct interaction between selectins on the synovial membrane venules. The
E-selectin and PSGL-1. principal ligands for L-selectin have been investi-
The rolling of neutrophils is blocked by gated extensively in mice and humans in relation
monoclonal antibodies against epitopes on the to the homing of lymphocytes to the high endothelial
240 8 Mechanisms of the Acute Attack of Gout and Its Resolution

venules in peripheral lymph nodes [248]. The mediators such as chemoattractants (IL-8, LTB4,
most well-defined L-selectin ligands include and C5a) and activating factors (tumor necro-
GlyCAM-1 (glycosylation-dependent cell adhe- sis factor and granulocyte-macrophage colony
sion molecule-1), MadCAM-1 (mucosal addressin stimulating factor) also induces the shedding of
cell adhesion molecule-1), podocalyxin-like pro- L-selectin from the neutrophil membranes. Most
tein, endoglycan, and Sgp 200 [249]. of these activators have been identified in the
GlyCAM-1 is a transmembrane glycosylated synovial fluid recovered from patients with acute
protein molecule with an 0-linked sugar and sia- gouty arthritis. The transmembrane metallopro-
lyl groups. This molecule has been cloned from teinase, tumor necrosis factor TNF-a convert-
human cells, has been localized to chromosome ing enzyme (TACE), is responsible for releasing
12q13.2, and supports the rolling of lymphocytes soluble TNF-a, L-selectin, transforming growth
on endothelial cells [250]. Mucosal addressin cell factor-a, and TNF receptor from cells. TACE or
adhesion molecule-1, another L-selectin ligand, ADAM-17 (a disintegrin and metalloproteinase)
has been primarily characterized in high endothe- is a zinc metalloproteinase that is membrane-
lial venules from lymph nodes. The human gene associated and has an extracellular domain, a
for MadCAM-1 has been cloned and character- transmembrane domain, and an intracellular
ize.. It has been localized to chromosome 19 at C-terminal domain. Blocking this receptor and
p13.3 near the genes for intercellular adhesion membrane-shedding process indicates the key
molecule-1 and -3 that are ligands for integrins. role played by TACE activity in the inflammatory
The gene has five exons, and each exon encodes process. TACE inhibitors decrease the roll-
a separate domain of the complete molecule (sig- ing velocity of neutrophils, upregulate Mac-1,
nal peptide, two immunoglobin domains, and a increase binding to ICAM-1, and increase the
mucin domain). Exon five encodes the transmem- firm adhesion of neutrophils to endothelial cells
brane domain, the cytoplasmic domain, and a [252]. As a corollary, L-selectin shedding limits
3¢-untranslated region. Little is known about the neutrophil recruitment to inflammatory sites by
other L-selectin ligands except for their role in decreasing the functions noted to be altered by
immune reactions. The complex structure to these TACE inhibitors. L-selectin shedding decreases
ligands for the selectins and their possible varia- leukocyte binding and subsequent transmigra-
tions provides diversity for the recognition of dif- tion since it decreases signaling to rolling leuko-
ferent cells and mediating their capture by cytes, decreases b2-integrin adhesive responses,
endothelial cells. and alters the accessibility to endothelial activa-
tion signals [253]. Data show that calmodulin is
associated with L-selectin in resting leukocytes
Unique L-Selectin Properties and the dissociation of these molecules leads to
L-selectin shedding. Speculation suggests that
L-selectin is a somewhat unique member of the changes in intracellular calcium concentrations
selectin family of adhesion molecules since it has and phosphorylation of L-selectin may trigger
two additional properties. It may cause shedding this dissociation [254]. Of relevance to acute gout,
of membrane L-selectin, and it also mediates sec- neutrophils shed L-selectin during their transend-
ondary neutrophil capture. L-selectin mediates othelial migration into inflamed lung tissue, and
the accumulation of neutrophils along the vascu- such studies suggest that L-selectin may also par-
lar endothelium in response to inflammation by ticipate in the transmigration of neutrophils into
invoking both neutrophil-endothelium and neu- the synovial joint space. Furthermore, painful
trophil-neutrophil interactions. Perhaps of greater stimuli cause L-selectin shedding and reduce neu-
significance to the cell adhesion process is that trophil migration into inflamed joints in a rat model
neutrophil activation causes L-selectin shedding, of arthritis. Increased concentrations of soluble
and this activity regulates leukocyte recruitment L-selectin also decrease inflammation(cytokine)-
[251]. Endothelial activation by inflammatory mediated leukocyte adherence and vascular
Adhesion Molecules 241

leakage in vivo. These studies document the since their measurement represents a noninvasive
key role L-selectin shedding plays in regulating means of defining what proinflammatory media-
neutrophil emigration to sites of inflammation. tors are active in specific diseases and what stage
Furthermore, such data support the development of the process is evolving. Evaluating such data
of pharmacologic agents that can act as selec- avoids direct, invasive assays of the synovial
tin antagonists or increase L-selectin shedding. membrane microvasculature.
Some of the human genes for L-selectin ligands Although few of the soluble cytokines have
have been cloned and characterized. been measured in acute gout, many of these
Since the cell membrane shedding process has mediators have been assayed in articular disor-
a key role in the regulation of cell interactions, ders. Since most of these cytokines are adhesion
the role of the soluble mediators such as the non- molecules residing on cell surfaces, the mecha-
chemotactic cytokines and chemoattractants nisms by which they become circulating, soluble
deserves further discussion. Tumor necrosis fac- mediators is of significance to the underlying
tor-a and its receptor when shed are no longer pathology as well as to the development of pos-
able to activate the expression of selectins and sible pharmacologic agents to alter the progression
integrins on cells and to promote the production of the process. For the most part, these soluble
of PAF, IL-8, and ICAM-1. IL-1 is also able to adhesive molecules arise either by secretion or by
mediate some of these cell changes. The soluble proteolytic action against these surface-expressed
chemoattractant molecules, as noted previously, molecules. In addition, some soluble adhesion
have a key role in activating neutrophils and molecules arise from variations in mRNA splic-
endothelial cells and the subsequent expression ing that result in the synthesis of molecules with-
of adhesion molecules. Although there is out a transmembrane domain. Other soluble
significant redundancy in the chemotactic agents cytokines (E-selectin, VCAM-1, ICAM-1, and
that may be active in regulating the migration of ICAM-3) have a molecular size consistent with
neutrophils to sites of inflammation, the presen- the absence of transmembrane and cytoplasmic
tation of activators and chemotaxins in their cor- domains, but they are also generated by prote-
rect sequence is necessary for neutrophil olytic mechanisms. The most well-characterized
migration. For example, IL-8 and C5a can cause proteolytic shedding mechanism is the cleavage
desensitization of cells to their actions through of tumor necrosis factor-a by tumor necrosis fac-
their own receptors, and they can also desensitize tor-a converting enzyme (TACE/ADAM-17)
cells to each other by acting through each other’s [255]. This metalloproteinase-disintegrin also
receptors as well as through the PAF receptor. releases the tumor necrosis factor p75 receptor,
Such functions document the complexity of transforming growth factor, interleukin-1 recep-
chemoattractant activity and the necessity for tor, p55 tumor necrosis factor-a receptor, and
concentration- and time-dependent regulation of l-selectin. TACE is a membrane-anchored trans-
their actions and emphasize the regulation that membrane protein that contains a proenzyme
must exist between the sequence of neutrophil domain, a catalytic domain (zinc-dependent met-
and endothelial cell activation, the expression of alloproteinase), a cysteine-rich sequence of 197
adhesion molecules, and the interactions between amino acids, a transmembrane domain, and a
various cell receptors. Such processes and their cytoplasmic tail. Calmodulin appears to act as a
regulation also suggest molecular mechanisms negative regulator of TACE activity [256]. The
for altering the recruitment of neutrophils and latent TACE proenzyme can be activated to its
interrupting the inflammatory process. active form by oxidizing a cysteine residue in the
Before discussing the tight adhesion of neu- TACE inhibitory peptide that masks the zinc atom
trophils to endothelial cells mediated by cell in the catalytic domain [256]. Tumor necrosis
membrane integrins, the role of soluble adhesion factor-a has been shown to induce the release of
molecules as biomarkers and regulators of the its receptors and also triggers the release of reac-
inflammatory process needs to be documented tive oxygen species. Thus, TNF and reactive
242 8 Mechanisms of the Acute Attack of Gout and Its Resolution

oxygen species can activate TACE to release sol- mitogen-activated protein kinases (MAPK).
uble mediators. In addition, nitric oxide can also These findings also provide a basis for the devel-
activate this enzyme. Monosodium urate crystals opment of therapeutic agents designed to prevent
are potent stimuli for the generation of interleu- cellular receptor binding or to mimic/inhibit
kin-1 (IL-1) and TNF-a production by mono- biological functions generated by these soluble
cytes and macrophages, and phagocytosis of adhesion molecules.
such crystals induces the generation of reactive On the basis of the foregoing facts, it may be
oxygen species [257]. Further, soluble TNF and reasonable to reexamine some of the other solu-
its receptors have been found in the serum and ble mediators found in synovial fluid to deter-
synovial fluid of patients with inflammatory mine if there are any additional biological
forms of arthritis including gout. Although little functions performed by these molecules. In addi-
is known about the role of nitric oxide in gout, tion to the soluble adhesion molecules cited
this mediator has been documented as a modula- above, intercellular adhesion molecule-1
tor of leukocyte transmigration in inflammatory (ICAM-1), vascular cellular adhesion molecule-1
joint disorders. (VCAM-1), soluble interleukin-6 (IL-6) receptor
The significance of either proinflammatory or as well as other mediators such as IL-6, IL-4,
anti-inflammatory soluble mediators in an acute IL-113, and IL-10 have been detected in synovial
inflammatory process such as gout has yet to be fluid and serum from patients with inflammatory
completely characterized, but recent findings indi- arthritides [258]. Recent publications have also
cate that these soluble cytokines are likely to have documented the use of soluble cytokine receptors
an important role as regulators of inflammation. as therapeutic agents and as regulators of joint
For example, soluble TNF and its soluble recep- inflammation.
tors may bind to each other and prevent excessive Another unique property of L-selectin is its
TNF activity which might be harmful to the host. ability to mediate secondary capture of leuko-
The competition between cellular receptors for cytes. Primary capture in this setting refers to the
TNF and soluble TNF receptors could prevent the initial rolling of leukocytes along endothelium,
injurious effects of excessive TNF activities act- whereas secondary capture defines a process
ing via cellular receptors. The second significant where freely flowing leukocytes interact with
point regarding soluble cellular adhesion mole- rolling leukocytes and subsequently attach to the
cules is the observation that such molecules may endothelium. The ligand for L-selectin in this
have a different role as soluble agents in contrast leukocyte-leukocyte interaction is P-selectin gly-
to their membrane bound actions. At this time, coprotein ligand-1. Although primary leukocyte
only vascular adhesion molecule 1 (VCAM-1) capture is the predominant mechanism causing
and E-selectin as soluble components have been leukocyte rolling, a low rate of secondary capture
shown to express such variations. In case of both does occur in venules.
these soluble adhesion molecules, monocyte
chemotaxis is one biological function generated
by these soluble mediators. Although monocyte E-Selectin
chemotaxis via this mechanism has only been
shown to occur with these soluble adhesion mol- The major ligand for E-selectin is E-selectin
ecules isolated from the synovial fluid obtained ligand-1 (ESL-1). E-selectin like P-selectin is
from patients with rheumatoid arthritis, a simi- expressed on activated endothelial cells. ESL-1
lar effect might also take place in the late stages binding to E-selectin requires N-glycans [259].
of acute gout when monocytes begin to appear ESL-1 has been cloned and characterized from
in the synovial fluid. The stimulation of mono- mice. A protein designated as MG160 has been
cytes by soluble E-selectin demonstrated a time- isolated and cloned from the Golgi apparatus
dependent increase in tyrosine phosphorylation and is homologous to fibroblast growth factor
of cellular proteins via the Src kinases and the and the E-selectin ligand, ESL-1. Mouse ESL-1
Adhesion Molecules 243

has also been localized to the Golgi apparatus, Integrins are transmembrane glycoprotein
and its role in this cell organelle is incompletely receptors that mediate cell-matrix or cell-cell
understood. In addition to E-selectin, L-selectin adhesion. They are heterodimers consisting of an
has been proposed as an E-selectin counterrecep- a and b subunit in a 1:1 stoichiometric ratio which
tor as has P-selectin glycoprotein ligand-1[260]. are bound noncovalently to each other. In 2000,
As noted previously, the selectin-mediated bind- 19 known a subunits and 8b subunits were
ing to selectin ligands results in the tethering and reported in mammals, and these subunits can
rolling of neutrophils along the endothelium. form at least 25 ab heterodimers. Since this dis-
Such initial interactions between neutrophils cussion focuses primarily on neutrophils the
and endothelium result in weak and reversible emphasis is on b1, b2, and b3 integrins that are
recognition of selectin ligands. The transition expressed by these cells [261]. The b2 integrin
from neutrophil rolling to firm adhesion to the subunit is the key structure regulating neutrophil
endothelium is mediated by a change from selec- binding to endothelium, and the trivial names for
tin-dependent adhesion to CD18 [132]-integrin- these integrin subunits are Mac-1 (macrophage
dependent firm adhesion between neutrophils antigen-1), LFA-1 (lymphocyte-function-associ-
and the endothelium. ated antigen-1), and GP150,95. The common
designations for these molecules are aMb2
(Mac-1), aL b2 (LFA-1), and axb2 (GP150,95),
Integrins and the CD nomenclature is as follows: CD11a
(aL). CD11b (aM), CD11c (ax), and CD18 (b2).
After the selectins initiate neutrophil tethering, The human a subunits have been localized in
firm adhesion of these cells to the endothelium a cluster on chromosome 16p13.1-p11. The
is mediated by integrins. Integrin is a term origi- human gene for the b2 subunit has been localized
nally coined to describe membrane receptors that to chromosome 21q22.3 [262].
integrate the extracellular matrix or cells with
the intracellular cytoskeleton. Since this original
terminology was defined, integrin functions have Integrin Structure
been extended to include cell differentiation,
motility, growth, and cell polarity. Of significance The structure of the human b2 chain (Mr of
to acute gouty arthritis is the fact that this glyco- 95,000) and the alpha subunits (aL, aM, and ax)
protein family regulates neutrophil trafficking. have all been determined. The a subunits vary in
The principal interest in integrins as they relate their molecular weight with human aL having a
to the inflammatory response is their capacity Mr of 177,000, aM with an Mr of 165,000, and ax
both to cause firm adhesion of neutrophils to the with an Mr of 150,000. Although alpha-4 beta-1
endothelium in preparation for their migration integrin has been documented as a functional inte-
from the blood vessel to the inflammatory site grin on murine and rodent neutrophils as well as
but also to transduce messages regulating neutro- on human neutrophils treated with LTB4, C5a, or
phil shape and ligand binding affinity. The rate TNF-a, its presence on human neutrophils is con-
at which cells migrate depends on the integrin troversial because the antibody utilized to identify
levels of expression induced by the inflammatory this integrin on human neutrophils is nonspecific.
mediators that cause the transport of b2-integrins The a subunit contains at its extracellular
from the peroxidase negative granules of neu- N-terminus a seven blade 13-propeller structure
trophils and monocytes to the cell surface. The attached to a b barrel or four to six 13-sandwich
substratum ligand levels for integrins and integ- domains and an I domain between the second and
rin-binding affinities also contribute significantly third propeller blades. The cytoplasmic tail of the
to the rate of cell migration. All integrins consist a subunits contains a GFFKR peptide sequence
of one alpha subunit and one beta subunit, and as (glycine-phenylalanine-phenylalanine-lysine-
such, they are heterodimers. arginine) and a phosphorylation site. Models of
244 8 Mechanisms of the Acute Attack of Gout and Its Resolution

these structural elements have been constructed. by b2 integrins requires signaling and activation
The b2 subunits also contain an 1-like domain of neutrophils. A variety of complement peptides
near their N-terminus and a cysteine-rich peptide (principally C5a), chemokines (primarily IL-8),
in their extracellular domain. A phosphorylation lipid factors, and other inflammatory mediators
site is also present on their intracellular cyto- such as IL-1, GM-CSF, PAF, and TNF-a can
plasmic tail. The configuration of these al3 het- serve as activators of neutrophils. The process of
erodimers changes in response to ligand binding activating neutrophils involves a complex series
as has been characterized by binding studies and of cell and mediator interactions that lead to neu-
in models of such binding sites [263]. Structure- trophil activation and adhesion. Interleukin-1 and
function studies of specific al3 heterodimers tumor necrosis factor activate endothelial cells.
are ongoing, but several conclusions have been These activated endothelial cells then produce
drawn from published analyses. First, the domain PAF in response to the aforementioned mediators
of the b2 subunit is the primary element involved as well as granulocyte-macrophage colony-stim-
in ligand binding via their metal ion-dependent ulating factor (GM-CSF), IL-1, and IL-8. These
adhesion site (MIDAS) motifs [264]. Second, the molecules (IL-1, PAF, GM-CSF, and IL-8) then
divalent cation binding sites on the ab heterodi- activate neutrophils and leukocyte adhesion. IL-1
mers have a critical role in ligand binding, and and TNF activate endothelial cells to produce
these sites may either promote or inhibit ligand mediators such as GM-CSF, IL-1, PAF, and IL-8
binding [265, 266]. Finally, the molecular varia- that in turn activate neutrophils.
tions in the integrins may be increased by changes Neutrophil activation by the foregoing media-
in gene splicing, posttranslational modifications, tors causes neutrophil degranulation and the
and molecular alterations induced by reactions translocation of receptors for adhesion molecules
with other cells or intracellular molecules. on the endothelium as well as enzymes from
The complete molecular characterization of specific intracellular neutrophil granules to the
neutrophil firm adhesion remains to be defined, cell surface [267]. Integrins are rapidly mobilized
but many of the component, parts of this complex by the degranulation step from secondary and
series of steps that mediate the transmigration of tertiary granules of neutrophils and are expressed
neutrophils are understood. The selectin-medi- at seven to tenfold greater levels on the cell sur-
ated tethering of leukocytes places these cells in face than they are in the quiescent neutrophil.
a setting in which neutrophils are activated to The neutrophil compartments that store pre-
express the b2 integrins, Mac-1 and LFA-1. The formed integrins (Mac-1 and LFA-1) include
sequence of events responsible for generating secretory vesicles, specific granules, and
firm adhesion to post-capillary venules includes gelatinase-containing granules [268]. Recently,
the activation of neutrophils to mobilize and clus- increased ceramide levels in neutrophils have
ter ab heterodimers (Mac-1 and LFA-1) adhesive been shown to induce cells to degranulate and to
molecules on neutrophil membrane and a subse- increase the expression of integrins on the neu-
quent change in the conformation of these mem- trophil plasma membrane [269]. Thus, TNF and
brane-expressed molecules to increase their chemoattractants cause neutrophil degranulation
avidity for their opposing ligand, intercellular and translocation of the b2 integrins to the neu-
adhesion molecule-1 (ICAM-1). The latter adhe- trophil plasma membrane, and the function of
sion molecule is an immunoglobin-like molecule these molecules remains quiescent until integrins
whose expression on endothelial cells is mark- are activated by intracellular and extracellular
edly increased by inflammatory cytokines. The signals that cause a conformational alteration in
loose adhesion of neutrophils to endothelial cells the molecule to expose its ligand binding sites.
by reversible bonds between the selectins and There are several factors that enhance the binding
their counterligands does not require neutrophil of integrins to their counterligands including a lipid
activation. In contrast to selectin-mediated teth- molecule given the name, integrin-modulating
ering, tight adhesion to the endothelium mediated factor and a cytoplasmic protein, cytohesin-1
Adhesion Molecules 245

[270]. These enhancing molecules and other [272]. ICAM-3 has now been recognized as the
mediators developed by so-called outside-in cell preferred counterligand for leukocyte aLb2 integ-
signaling pathways are responsible for the b2 rin and is found on the surface of neutrophils as
integrin avidity for. its counterligand and for well [273]. ICAM-3 binding occurs in the region
spreading of the neutrophil on the endothelial where aLb2 integrin causes neutrophil aggrega-
cell surface in preparation for its transmigration. tion and induces changes in the neutrophil
Such changes are mediated by interactions cytoskeleton to enhance adhesion. After these
between neutrophil integrins and the cell’s interactions between Mac-1, LFA-1, and integ-
cytoskeleton [271]. rins, neutrophils are tightly adherent to venule
endothelial cells and ready for their migration
through the venule wall to the inflammatory site.
Ligands for Integrins The genes for ICAM-1, ICAM-2, and ICAM-3
have been mapped to chromosome 19p13.3-
Tight adhesions between endothelial cells and p13.2, chromosome 17q23-25, and chromosome
neutrophils are dependent on the binding of these 19p13.3-p13.2, respectively. The gene for
neutrophil integrins with their ligands. The prin- VCAM-1 has been mapped to chromosome
ciple ligands for LFA-1 and Mac-1 are the 1p32-p31.
endothelial ligands designated as intercellular The selectin family of adhesion molecules
adhesion molecules (ICAMs). These ICAMs regulates the initial step of the interaction between
belong to the immunoglobulin gene superfamily neutrophils and endothelium, and the second step
(lgSF). ICAM-1 (CD54) contains five extracel- culminating in the firm arrest of neutrophils
lular immunoglobulin domains, a hydrophobic requires interactions between neutrophil integrins
transmembrane domain, and a short cytoplasmic and the endothelial immunoglobulin superfamily.
tail at its C terminal end. ICAM-2 (CD102) has a Inflammatory mediators including chemoattrac-
structure similar to ICAM-1 except that it con- tants and TNF-a stimulate an increase in b2 inte-
tains only two extracellular immunoglobulin grins on the surface of the neutrophil. Two types
domains in contrast to the five in ICAM-1. of adhesion occur via b2 integrins. One type
ICAM-3 is another member of the immunoglob- involves the recruitment of neutrophils from the
ulin gene superfamily with a structure similar to flowing blood stream to neutrophils already bound
ICAM-1 since it has five extracellular immuno- to the endothelium of the vessel wall [274]. These
globulin domains. TNF, IL-1 and other mediators neutrophil-neutrophil interactions have been
markedly increase the expression of ICAM-1 called neutrophil homotypic adhesions, and they
during inflammatory states. Another related mol- are dependent upon interactions between LFA-1
ecule, vascular cell adhesion molecule-1 (VCAM- (CD11a/CD18) and intercellular adhesion mole-
1) is an inducible endothelial cell immunoglobulin cute-3. ICAM-3 is expressed at high concentra-
superfamily member and an effective ligand for tions on the surface of neutrophils, monocytes,
integrin a9b1. VCAM-1 interaction with a9b1 inte- and lymphocytes, and this molecule differs
grin supports the adhesion of neutrophils to TNF- significantly in its transmembrane and cytoplas-
a-activated endothelial cells. However, the mic domains from ICAM-1 and ICAM-2.
primary role of this cell surface glycop- ICAM-3 is also a counterreceptor for the recently
rotein(VCAM-1) is to mediate the adhesion of characterized aDb2 integrin. ICAM-3 expression
monocytes and lymphocytes to cytokine-acti- on neutrophils exceeds the expression of ICAM-1
vated endothelial cells. Vascular cell adhesion and can activate neutrophils to a proadhesive
molecule-1 is a cell surface glycoprotein contain- state in the absence of stimuli to recruit b2 inte-
ing 9 exons and spanning approximately 25 kb of grins from intracellular storage sites. Activation
DNA and a binding site domain 1. Integrin aLb2 of neutrophils via ICAM-3 causes a reorganiza-
binds to domain 1 of the ICAM-1 molecule, and tion of the neutrophil cytoskeleton and the syn-
integrin aMb2 binds to domain 3 of ICAM-1 thesis and secretion of chemokines (IL-8). Such
246 8 Mechanisms of the Acute Attack of Gout and Its Resolution

neutrophil-neutrophil-mediated interactions along after 1 min in experimental systems, whereas


with IL-8 secretion amplify the recruitment of Mac-1 adhesions are more sustained. In this
neutrophils and the magnitude of the inflammatory experimental system, Mac-1 translocates to the
response. Furthermore, the binding between uropod, whereas LFA-1 remains bound to lamel-
LFA-1 and ICAM-3 is much weaker than the lipodal regions. In TNF-a-activated endothelial
binding between LFA-1 and ICAM-1, and such cells, Mac-1 accumulates at the region of contact
differences may favor neutrophil-endothelial cell between the neutrophil and the endothelial cell,
interactions. Interleukin-8-stimulated neutrophils and as the neutrophil spreads on the endothelial
cause a redistribution of ICAM-3 and moesin, a cell, Mac-1 translocates to the leading edge of
molecule linking plasma membrane proteins and the neutrophil. Thus, Mac-1 moves from the con-
actin cytoskeleton, to the distal portion of neutro- tact area between neutrophil and endothelial cell
phil uropods (a projection of the neutrophil in the to the leading edge of the migrating neutrophil
rear of the migrating cell). Thus, ICAM-3 may as it moves to make its way across the endothe-
play a key role in the recruitment and amplification lium. The LFA-1 affinity for endothelial ligands
of the migration of neutrophils to an inflamed site. is normally induced by allosteric activation of
The ligand, vascular cell adhesion molecule-1 CD18 and amplified by chemotactic signals.
(VCAM-1), has also been identified on human Phosphatidylinositol 3-kinase activity is a key to
endothelial cells and is induced by TNF-a and the mobility of active CD18 and the formation of
IL-1, inflammatory mediators present in acute high avidity patches of CD18. During the change
gout [275]. Integrins a4b1and a9b1 on neutrophils from inactive binding to an active ligand-binding
bind to this endothelial ligand. These integrins state, integrins cluster and alter their conforma-
and their endothelial ligand, VCAM-1, represent tion. As can be determined by these changes in
a b2-integrin independent neutrophil adhesion neutrophil-endothelial binding both inside-out
mechanism that can contribute to neutrophil-en- signaling and outside-in signaling are required
dothelial cell adhesion and neutrophil transmi- for optimal binding. Chemoattractants activate b2
gration through the endothelium. This pathway integrins. This b2 integrin activation and ligand
may be of significance to an arthritic disorder like binding is mediated by inside-out signaling. Once
gout since neutrophils expressing a4-integrin ligand occupancy occurs then signal transduction
have been identified in rats with adjuvant arthri- into the cell takes place via outside-in signaling.
tis. The complete role of VCAM-1 and the b1- The latter process governs endothelial gap for-
integrins in human articular inflammatory mation and increased endothelial permeability,
conditions remains to be determined. and the transmembrane signaling by neutrophils
The second type of adhesion occurring via the causes an elevation of endothelial cell free cal-
b2-integrins involves interactions between LFA-1 cium, a rearrangement of their actin filaments,
(aLb2) and Mac-1 (aMb2) integrins and their ligands, a conformational change in their cytoskeleton,
ICAM-1 and ICAM-2 [276]. Chemoattractants endothelial cell contraction, and gap formation
like LTB4 and IL-8 activate b2 integrins (CD11a/ [277]. Vascular permeability factor/vascular
CD18 and CD11b/CD18) and result in the sequen- endothelial cell growth factor (VPFNEGF) and
tial binding of these integrins to ICAM-1 and its generation by neutrophils is a key mediator
ICAM-2 on endothelial cells. LFA-1 is involved of inflammation and neutrophil transmigration.
primarily in the capture of neutrophils and the This permeability factor promotes the produc-
establishment of sustained contact of neutrophils tion of fenestrations that are known to exist in
with endothelium. Subsequently, Mac-1 interac- the capillaries of the synovium. VEGF, present
tion with ICAM-1 and ICAM-2 causes the stable in the specific granule fraction of neutrophils,
adhesion of neutrophils to the endothelium. The is secreted by human neutrophils stimulated by
chemoattractants, IL-8 and LTB4, induce optimal TNF-a [278]. The permeability of endothelial
rates of LFA-1-dependent adhesion at subnano- cells is associated with changes in the endothe-
molar concentrations. Furthermore, LFA-1 adhe- lial cytoskeleton. Such changes are calcium-
sions to endothelium are brief and begin to decay dependent and lead to endothelial cell contraction
Oxygen Radical Generation 247

and intercellular gap formation Tyrosine kinase is protein kinase (MAPK). Inside-out signaling or
required for the changes in cell shape accompa- the integrin activation pathway regulates ligand
nying the formation of gaps, the reorganization binding and may increase the affinity or avidity
of actin filaments, and the formation of F actin of the receptor for its ligand. Such complex sig-
are likely to participate in these changes in cell naling pathways mediated by leukocyte integrins
shape. have been reviewed in recent publications, but the
Since the absence of the cytoplasmic domain complete picture of integrin signaling remains an
of ICAM-1 inhibits neutrophil transmigration area of active research. The significance of these
but adhesion remains intact, signals generated by integrin signaling functions has been demon-
ICAM-1 are of importance to neutrophil migra- strated through the use of antagonists and mono-
tion. Furthermore, the migration of neutrophils on clonal antibodies against integrin molecules,
the surface of endothelial cells towards endothe- and such studies have documented a variety of
lial junctions is inhibited by the presence of p38 functional defects in cells treated with these inte-
mitogen-activated protein kinase inhibitors. These grin modifiers. The goal of investigations using
inhibitors also block endothelial cytoskeleton integrin antagonists is to develop therapeutic
changes and stiffening of these cells. The means agents that could modify the inflammatory reac-
by which ICAM-1 ligation mediates signals to the tion and its progress by altering the transmigra-
endothelial cell is not fully understood, but some tion of inflammatory cells. Since the pathological
aspects are known. ICAM-1 is associated with changes induced by urate crystals are in some
an actin-binding protein, a-actinin. The cyto- part repaired by inflammatory responses in the
plasmic domain of ICAM-1 can bind phosphati- tissues, the dose of therapeutic integrin antago-
dylinositol 4,5-bisphosphate and ezrin/radixin/ nists would have to be carefully adjusted to
moesin proteins. Finally, neutrophil adhesion maintain tissue repair while still modifying the
induces increments in endothelial cell calcium, inflammatory response.
and chelation of such calcium increments in the In conclusion, several regulators of neutrophil-
endothelial cell does not alter neutrophil adhe- endothelial cell adhesion have a significant role
sion but inhibits neutrophil transmigration across in the modulation of neutrophil migration. There
endothelial cells [279]. Future investigations will are some observations in mouse mutants deficient
identify more fully the endothelial cell signaling in adhesive molecules that b2 integrins and selec-
pathways and their specific role in the functions tins collaborate to produce the ideal conditions
related to neutrophil migration. for firm adhesion of neutrophils to endothelial
One final principal regarding the function of cells. In mice with a deficiency of CD18, leuko-
integrins has significance to their role in neutro- cyte rolling velocities are significantly increased,
phil transmigration as well as the identification and such data support a role for CD18 in the
of possible therapeutic targets to impede regulation of leukocyte rolling velocity [280–
inflammatory reactions. In addition to their 282]. Thus, in murine species, b2 integrins may
capacity to mediate cell migration in response to decrease leukocyte rolling velocities before leu-
tissue injury, integrins also act as signal transmit- kocytes rolling on endothelial cells establish firm
ters capable of transducing information in both adhesions.
directions across the plasma membrane of cells.
These bidirectional message transduction routes
have been characterized as outside-in or inside- Oxygen Radical Generation
out signaling. The outside-in pathway appears to
control the cell shape of the neutrophil and other NADPH oxidase and the generation of reactive
cytoskeletal alterations as well as triggering the oxygen radicals During phagocytosis by neutro-
synthesis of different biochemical mediators to phils, there is a burst of oxygen consumption.
signal these changes. Prominent effectors of out- This burst of oxygen consumption is related to
side-in signaling include tyrosine kinases, phos- the generation of oxygen radicals by the enzyme
phatidylinositol 3-kinase, and mitogen-activated complex, NADPH oxidase and the resultant
248 8 Mechanisms of the Acute Attack of Gout and Its Resolution

superoxide ( O -2 ) production via this enzyme superoxide and hydrogen peroxide are produced
which is necessary for the destruction of a vari- from the NADPH oxidase complex residing on
ety of microorganisms. The significance of this the plasma and phagolysosomal membranes and
enzyme’s activity is documented by the suscep- using NADPH as an electron donor.
tibility of patients with chronic granulomatous
disease to infections because their NADPH 2O 2 + NADPH ® O -2 + NADP + + H +
oxidase is inactive. Since oxidants generated
by NADPH oxidase may also induce damage Most of the superoxide produced by this
to nearby tissues, its ·enzyme activity must be membrane-localized reaction reacts with itself by
tightly regulated to avoid such damage. In fact, a dismutation reaction in a spontaneous manner
the translocation of the enzyme’s components to form hydrogen peroxide. As diagramed below,
and its activation prior to the closure of the pha- this dismutation reaction is catalyzed by the
gosome may account for the local production of enzyme, superoxide dismutase.
oxidants capable of causing local tissue destruc-
tion. Further, when neutrophils attempt to ingest 2O -2 + 2H + ® H 2 O 2
an antibody- or complement-coated surface too
large to phagocytose, reactive oxygen species In addition to these oxidants, a variety of other
may be released in a process known as frustrated superoxide-dependent reactive oxygen species
phagocytosis. These oxygen-derived molecules may also be produced including singlet oxygen,
can amplify Fey receptor signaling and thereby hydroxyl radical, hypochlorous acid, chloramine,
increase the phagocytic capacity of the neutro- nitric oxide, and peroxynitrite.
phil. Furthermore, an imbalance between pro- Under quiescent circumstances in neutrophils,
tein tyrosine phosphatase (CD45) and protein the enzyme complex is dormant and its compo-
tyrosine kinase in favor of decreased phospho- nent parts are stored in different cellular compart-
rylation causes a decrease in calcium mobiliza- ments to maintain their inactivity. Six oxidase
tion, the respiratory burst, and phagolysosome subunits are necessary to construct the complete
degranulation. Evidence suggests that the imbal- enzyme, and they are designated by their appar-
ance between tyrosine kinases and tyrosine phos- ent molecular mass in kilodaltons. The term phox
phatases is, in part, created by oxidant-mediated is an eponym for phagocyte oxidase and is also
inactivation of tyrosine phosphatases. The mech- used to designate these component parts. The
anism by which oxidants inactivate tyrosine membrane-localized components are made up of
phosphatase activity appears to be via cysteine a b-type cytochrome constructed as a heterodi-
residues in the catalytic site of the enzyme. Thus, mer containing a 91 kDa glycosylated protein
reactive oxygen intermediates, especially hydro- (g91-phox) and a nonglycosylated 22 kDa sub-
gen peroxide, amplify FcgR-mediated phago- unit (p22-phox). The cytoplasmic components
cytosis in both neutrophils and monocytes by consist of p47-phox, p40-phox, p67-phox, and
increasing receptor-mediated tyrosine phospho- the low molecular weight GTPase, Rac1 or Rac2
rylation of FcgR-Iinked ITAMs. [283]. The p47-phox, p67-phox, and p40-phox
To avoid the generation of oxygen radicals in exist as a complex in neutrophil cytoplasm [284].
resting neutrophils, the components necessary The gene for p47-phox has been localized to
for neutrophil activation and the generation of chromosome 7q11.23, and the gene for p67-phox
oxygen radicals are localized in separate subcel- has been mapped to chromosome 1q25. The gene
lular compartments. The p40phox, p47phox, and for the membrane-localized gp91-phox is located
p67phox occur in the cytosol of neutrophils as on the x chromosome at p21.1, and the gene for
a complex, and p22phox and gp91phox are localized p22-phox has been mapped to chromosome
to the membranes of secretory vesicles and 16q24.
specific granules. The following equations repre- The component essential for electron trans-
sent the biochemical mechanisms by which port from cytoplasmic NADPH to oxygen is
Oxygen Radical Generation 249

gp91-phox, and the other membrane subunit, phox, of course, links these molecules with
p22-phox, is a regulatory molecule necessary for cytochrome b558. Phosphatidylinositol kinases
the activation· of the oxidase and the recruitment and phosphatases are required or membrane
of other cytosolic subunits. The gp91-phox sub- localization of the cytoplasmic PHOX constitu-
unit contains a NADPH binding site, a noncova- ents. The conformational changes in the p47-
lently bound flavin adenine dinucleotide (FAD), phox upon activation also lead to the interaction
and two non-identical heme groups integrated by between the phox homology (PX) domain of
two histidine residues. A variety of soluble and p47-phox and the activated PI3K [287]. P47-
particulate material can stimulate the assembly phox binds predominantly to phosphatidylinosi-
and activation of NADPH oxidase in phagocytic tol 3,4-bisphosphate. The present suggestion is
cells including chemokines, opsonized microor- that these interactions lead to membrane localiza-
ganisms, arachidonic acid metabolites, and cal- tion of p47-phox. It is clear that the cytosolic pro-
cium. The assembly of a functional NADPH teins, p47-phox and p67-phox form a complex
oxidase and its activation include a variety of sig- with p40-phox in the cytoplasm of the neutrophil.
nal transductions induced by membrane receptor Phosphorylation of p47-phox at a critical serine
binding as well as the signals and changes neces- residue assists in the formation of a p47-phox,
sary for NADPH oxidase assembly. Receptor p40-phox, and p67-phox complex. The other crit-
binding triggers the generation of two significant ical component for the formation of active
second messengers, diacylglycerol and inositol NADPH oxidase is Rac2. Stimulation by the ini-
1,4,5-triphosphate. These phospholipid deriva- tiation of phagocytosis leads to the dissociation
tives cause the release of calcium from its intrac- of the RhoGTPase, Rac2, from its inhibitor
ellular stores and activate protein kinase C. This RhoGDI, and Rac2 then interacts with the cell
enzyme along with tyrosine kinases, serine membrane via its prenylated C-terminus. The
kinases, threonine kinases and mitogen-activated binding of Rac2 to the membrane-localized
kinases may all change the phosphorylation of flavocytochrome b558 forms a binding site for
neutrophil proteins. Guanine nucleotide-binding p67-phox. P67-phox also contains a so-called
proteins as well as GTPases are also involved in activation domain in residues 187–210 that inter-
the regulation of phagocytosis. acts with gp91-phox and regulates the electron
flow from NADPH to FAD [288]. The role of the
cytoplasmic components of NADPH oxidase in
NADPH Oxidase Assembly the final construct is incompletely resolved, but
p40-phox increases the affinity of p47-phox for
The key to the assembly of an active NADPH cytochrome in in vitro systems. In any event, the
oxidase resides in the cytoplasmic p47-phox. p40-phox SH3 domain interacts with the proline-
Exposure of neutrophils to a wide variety of stim- rich domain of p47-phox and its PC domain
uli leads to the translocation and integration of interacts with the PDI domain of p67-phox. The
the cytosolic subunits with the membrane sub- p40-phox is not necessary for NADPH oxidase
units of NADPH oxidase [285]. P47-phox is activity when the components are reconstituted
inhibited in resting cells through the intramolecu- in vitro, and genetically altered p40-phox does
lar binding of two SH3 groups in the N-terminal not lead to chronic granulomatous disease. Rac2
portion of a polybasic sequence of the C-terminus is essential for reconstituted NADPH oxidase
of this molecule. Protein kinase C, Akt, or MAP activity in vitro and in vivo. Rac translocates to
kinases can phosphorylate this region of the intra- the cell membrane independent of p47-phox,
molecular binding, block the autoinhibition of p67-phox, p40-phox, and gp91-phox. All these
the molecule, and open binding sites in the subcomponents of NADPH oxidase and their
exposed N-terminal SH3 domains for interaction functions are the subject of recent reviews, and
with the proline-rich domain of membrane-local- additional details can be acquired from this pub-
ized p22-phox [286]. This interaction of p47- lication [289, 290].
250 8 Mechanisms of the Acute Attack of Gout and Its Resolution

As can be inferred from the foregoing discus- alkaline phase causes a transient increase in pH
sion, NADPH oxidase is found on the surface of to 7.8–8.0. The second phase causes a decrease in
the cell’s plasma membrane but when phagocyto- pH to 6.0 over time through the activities of a
sis occurs, the cell’s external plasma membrane Na+/H+ antiport, H± ATPase, and an H+ conduc-
becomes shifted to the internal portion of the tance channel. It has been postulated that the ini-
phagosome via membrane invagination and can tial increase in phagolysosomal pH results from
release its oxidants directly on the ingested for- the consumption of hydrogen ions by the reaction
eign body. In the resting neutrophil, a small por- catalyzed by superoxide dismutase. This reaction
tion of the oxidase (10 %) is localized at the generates hydrogen peroxide by the following
plasma membrane and remainder (90 %) is local- reaction:
ized in the secretory vesicles and specific gran-
ules. In contrast to the neutrophil, NADPH 2O 2 + 2H + ¾¾¾¾¾¾
superoxide dismutase
® H 2 O2
oxidase resides exclusively on the cell plasma
membrane in the macrophage. Phagosomal acidification is not only an essen-
tial component of neutrophil microbicidal
responses but also provides the acid environment
Oxidants and Oxidant-Mediated for the acid hydrolases resident in the lysosomes.
Destructive Responses Further, these oxidants may inactivate enzymes
that have an oxidant-sensitive reactive group
Since this discussion is concerned with the inges- required for catalysis.
tion and destruction of uric acid crystals and at The generation of the strong oxidant, hydroxy
times, a complicating infectious organism, a more radical, has been the subject of much speculation
complete discussion of reactive oxygen species since the original papers by Fenton and the eluci-
and oxygen-independent mechanisms of micro- dation of the Haber-Weiss reaction. These chemi-
bicidal activity is warranted. Some of the oxi- cal reactions are as follows:
dants suspected of causing oxidation are either Fenton Reaction
unlikely to have a major role as oxidants or may
be very short-lived and difficult to detect. The H 2 O 2 + Fe 2 + ® Fe3+ + OH - + • OH
most prominent oxidants produced by neutro-
phils is superoxide, and most of the oxygen con- Haber-Weiss Reaction
sumed by the oxygen burst of phagocytes ends up
as hydrogen peroxide. H 2 O 2 + O 2- ® O 2 + OH - + • OH
When two superoxide molecules interact with
each other spontaneous dismutation occurs. One Now it is known that small amounts of • OH
superoxide molecule is oxidized, and the other are formed in neutrophils by the MPO-H2O2-Cl−
reduced with the resultant formation of oxygen system. Although very small amounts of
and hydrogen peroxide. This reaction occurs pre- hydroxyl radical are formed, its reaction with
dominantly at pHs of less than 5.0, but as the pH carbon dioxide to form the HCO3. radical may
rises, the reaction to form hydrogen peroxide is create a microbicide. Singlet oxygen, 1O2, is also
more dependent on superoxide dismutase. The formed in neutrophils via the myeloperoxidase-
latter enzyme may be provided by ingested H2O2-chloride system. There is also evidence
microorganisms. Phagosomal pH is also regu- that ozone can be generated by oxidation of
lated by NADPH oxidase through the transfer of water by singlet oxygen to produce ozone (O3)
reducing equivalents across the cell membrane to and hydrogen peroxide. Ozone is highly reactive
oxygen, and the subsequent acidification of the and has a relatively long half-life (t1/2 = 1 min in
cytoplasm. The neutrophil phagosome undergoes water at 37 °C. The production of ozone by neu-
a biphasic response with respect to its pH. trophils requires antibody (IgG), singlet oxygen,
Although incompletely understood, the initial or and water [291].
Oxygen Radical Generation 251

Myeloperoxidase peroxide and ferric iron (Fe2+) [209]. Compound


I then reacts with a halide to form hypohalous
A key to the microbicidal system of phagocytes acid with the generation of ferrous iron (Fe3+).
is myeloperoxidase (MPO) that together with Compound I can also be formed through the reac-
hydrogen peroxide, and a halide, especially chlo- tion of MPO and hypochlorous acid (HOCI).
ride, form hypochlorous acid and subsequently Compound II is formed when an excess of hydro-
chlorine, chloramines, hydroxyl radicals, singlet gen peroxide is present, and this compound can-
oxygen, and ozone. MPO resides in the azuro- not form hypochlorous acid. Superoxide reacts
phil granules of granulocytes and in the primary with both MPO and compound II. In the latter
lysosomes of the monocyte. MPO granules in the case, superoxide reduces compound II to its
monocyte are fewer than in the neutrophil. There active form (compound I) with the capacity to
is heterogeneity amongst the MPO enzymes found form HOCI again. How effective the superoxide-
in neutrophil granules. The human gene for MPO mediated conversion of compound II to com-
has been cloned and localized to chromosome pound I is unresolved since other reducing agents
17q12-24. The enzyme is enzymatically altered appear to be more efficient than superoxide.
during its processing from its initial translation to Superoxide can react directly with myeloperoxi-
its mature 140 kDa protein. Mature MPO consists dase to form compound III, and this conversion is
of a pair of a subunits (59 kDa) and two smaller slower than the reaction of myeloperoxidase and
b subunits (13 kDa). The light subunits contain hydrogen peroxide. The conversion of native
112 amino acids and the heavy subunits have MPO to compound III is not as rapid as the con-
467 amino acids. The heavy subunits are linked version of MPO and hydrogen peroxide despite
by disulfide bonds via cysteine 319, mannose- the presence of compound III in neutrophils and
rich carbohydrates, and two hemes that are cova- its reactivity with many compounds.
lently bound to the heavy subunit. The reduction Despite all these possible reactions to form
and alkylation of MPO creates a hemi-myeloper- various compounds, the significance of MPO is
oxidase consisting of a heavy (a) and light (b) its antimicrobial activity via its production of
subunit. This hemi-myeloperoxidase retains its hypochlorous acid and the monochloramines
enzymatic and bactericidal activities. MPO is a [292]. As can be determined from the foregoing
basic protein with an isoelectric point of greater citations, a vast array of oxidant-mediated reac-
than 10, and on the basis of this chemical property, tions can occur leading to changes in bacteria and
it has the propensity to bind to negatively charged subsequently microbial destruction. Such changes
molecules. As has been known for many years, cause alterations in microbial membrane trans-
MPO is found in cytoplasmic granules and is dis- port, disruption of membrane electron transport,
charged into phagosomes. Although myeloperoxi- destruction of membrane nucleotide energy
dase may find its way into the extracellular milieu pools, suppression of bacterial DNA synthesis,
(frustrated phagocytosis and an unclosed phago- and oxidation of iron and sulfur necessary for
some), it can be removed from the extracellular microbial viability. There is now extensive evi-
compartment via a mannose receptor-mediated dence that microbicidal activity is MPO-
process by macrophages. When bacteria associ- dependent, and the presence of 3-chlorotyrosine
ated with myeloperoxidase or eosinophil peroxi- and 3,5-dichlorotyrosine in bacterial proteins
dase are taken up by macrophages, the destruction inside phagosomes provides documentation of
of microorganisms is enhanced. After many puta- these MPO-dependent mechanisms [293–296].
tive hypotheses, in the late 1960s, it was shown
that MPO, H2O2, and iodide, bromine, chloride,
or thiocyanate ions formed a potent antimicrobial Nitric Oxide and Bacterial Killing
system in neutrophils.
The mechanism of action of MPO is to form Another oxidant-dependent system in the human
initially compound I in the presence of hydrogen is the generation of nitric oxide (NO) from the
252 8 Mechanisms of the Acute Attack of Gout and Its Resolution

oxidation of L-arginine to L-citrulline. The reaction More recent investigations with peroxidase
is catalyzed by nitric oxide synthase and and hydrogen peroxide used to oxidize uric acid
requires molecular oxygen. Human neutrophils demonstrated the following products of this reac-
and macrophages contain an inducible nitric oxide tion: urea, carbon dioxide, alloxan, alloxan
synthase. Nitric oxide reacts with superoxide to monohydrate, allantoin, 5-hydroxyhydantoin-5-
form peroxynitrite ion (ONO−2). Peroxynitrite ion carboxamide and parabanic acid. Thus, oxidation
can be converted to peroxynitrous acid (ONO2H) by myeloperoxidase and hydrogen peroxide not
at acid pHs, and this compound then converts to a only kills microbes but also breaks down uric
strong oxidant. Both these compounds have acid into soluble products for disposal and
microbicidal properties [297]. Despite the forma- excretion.
tion of these reactive nitrogen intermediates, Although oxygen-independent mechanisms
a question remains as to their role in phagosome- exist that also contribute to the microbicidal
related bacterial killing [298]. activity of phagocytes, these phagocyte resident
molecules are of no relevance to the disposal of
uric acid. They may, however, contribute to the
Oxidants and Uric Acid Destruction bactericidal activity of these phagocytes. Since
septic arthritis may accompany acute gout in
Even though bacterial infections sometimes some patients, a brief overview of these oxy-
accompany acute gouty arthritis, the main prem- gen-independent mechanisms is provided. The
ise for discussing oxidants is to determine how major oxygen-independent bactericidal proteins
the host disposes of ingested uric acid crystals. in neutrophils include cathepsin G, cationic
The older literature confirms that humans can antimicrobial protein (CAP37 or azurocidin),
metabolize uric acid despite the absence of proteinase 3, elastase, bactericidal/permeabil-
uricase. In addition to these experiments showing ity-increasing protein (BPI or CAP57), defensins
uricolysis in the intact human host, studies have (HNP-1, HNP-2, HNP-3, and HNP-4), and
also shown that verdoperoxidase (MPO) is capa- lysozyme. These bactericidal proteins reside,
ble of oxidizing uric acid. In these experiments, for the most part, in the neutrophil azurocidin
MPO and hydrogen peroxide yielded urea and granules, but some (lysozyme) are also found in
allantoin. specific granules of the neutrophil. They also
An additional problem arises in the manage- have a spectrum of antibacterial activity against
ment of gout relating to the clearance of uric acid gram-positive and gram-negative organisms.
crystals and bacteria, if infection triggers the The defensins have the broadest spectrum of
acute gouty episode. There is a need to clear such activity since gram-positive and gram-negative
foreign agents before initiating potent therapeutic bacteria as well as many fungi and some viruses
initiatives that may obviate the ingestion and deg- are susceptible to its activity. CAP37, protei-
radation of the crystals or bacteria. To make such nase 3, elastase, and bactericidal/permeability-
agents more appropriate for use, the development increasing protein are effective against
of synovial fluid markers are needed to determine gram-negative bacteria. The effects of some of
reasonably and accurately the stage of the these proteins are also dependent on intrapha-
inflammatory reaction. A most effective marker gosomal pH. However, studies of the intrapha-
would be one that indicates with some degree of gosomal pH have provided data showing a delay
certainty the point at which there is a shift from in the acidification of this structure. These data
an acute process to a resolving one. The use of may be subject to error if the chlorination of a
such markers would provide an opportunity to probe is used to measure pH. Despite these
use agents such as 15d-PGJ2 or TGF-13, as well variable results, it does appear that intraphago-
as natural molecules that enhance the resolution somal pH decreases during and after phago-
of an inflammatory response. cytosis.
Proinflammatory Mediators 253

Proinflammatory Mediators activate inflammatory cells since interactions


between the plasma membranes of cells and crys-
Non-chemotactic, Proinflammatory Cell tals may trigger cellular activation and the release
Mediators of prostaglandins [299]. Thus, prostaglandins
and leukotrienes are likely to play a role in the
The key components of an acute inflammatory earliest manifestations of acute gouty arthritis
response in the synovial space are the recruitment and probably account, in part, for the vasodila-
and emigration of polymorphonuclear leukocytes tory response and the pain of acute gouty
from the blood into the joint space as well as the episodes.
accompanying vascular leakage via the postcap- Interleukins Interleukins have also been
illary venules. This complex process requires the detected in synovial fluid and are produced by
production and release of both cellular and synovial and cellular components known to be
plasma mediators as well as endothelial cell and involved in acute gouty arthritis. Two forms
neutrophil responses to generate the crystal- of interleukin-1 exist, IL-1a and IL-1b, and
induced inflammatory reactions observed in gout. they have identical biological functions. These
The inflammatory properties of urate crystals interleukin molecules are significant media-
have been documented as mediators of an acute tors of the inflammatory response in acute gout.
synovitis as well as an inflammatory response in Interleukin-1, an endogenous pyrogen, stimulates
skin and subcutaneous tissues. Cytokines and the production of PGE2 by synovial fibroblasts
synovial fluid-derived mediators that cause the and induces the synthesis of other cytokines
inflammatory response are produced in the syn- including interleukin-6, a mediator of acute phase
ovial compartment along with neutrophil reactants like fibrinogen, C-reactive protein, and
chemoattractants. Since a variety of cells includ- serum amyloid A as well as the potent neutrophil
ing neutrophils, synoviocytes, endothelial cells, chemoattractant, interleukin-8. A key role for the
and others as well as plasma may produce media- acute phase reactant, serum amyloid A (SAA),
tors of inflammation, the exact sequence and has recently been described in acute inflammatory
temporal pattern of mediator synthesis and release processes [300]. SAA, at a concentration of 100
in gouty arthritis remains incompletely defined. ug/ml, induces a 75- to 400-fold increase in
However, the proinflammatory mediators respon- TNF-a, IL-113. and IL-8 over basal levels. Thus,
sible for the symptoms and signs of acute gout SAA may be one of the early mediators of IL-8
can be postulated on the basis of their biological release, and the latter molecule is a potent neu-
properties as well as what molecules have been trophil chemoattractant and a major mediator of
isolated from the synovial fluid in acute gouty the neutrophil influx observed in gouty synovi-
arthritis. tis. IL-8 also enhances exocytosis, stimulates the
respiratory burst, and increases the expression
of membrane adhesion receptors in neutrophils.
Vasoactive Arachidonic Acid Thus, these three interleukins (IL-1, IL-6, and
Metabolites IL-8) are likely contributors to the early manifes-
tations of acute gout with interleukin-1 providing
Vasoactive prostaglandins (prostaglandin E2 the vasodilatory effects through its capacity to
[PGE2] and prostaglandin I2 [PGI2]) as well as the generate PGE2 and its pyrogenic effect leading to
potent neutrophil chemoattractant, leukotriene the low-grade fever sometimes observed during
B4 [LTB4], and hydroxyeicosatetraenoic acids acute gout. Interleukin-6 undoubtedly also con-
are synthesized by synovial cells and other tributes to the generation of acute phase reactants
inflammatory cells found in the synovial fluid like C-reactive protein which is elevated during
during acute gouty episodes. In addition, MSU the acute gouty episode and interleukin-8 which
crystals do not need to induce membranolysis to is the principal neutrophil chemotaxin in gout.
254 8 Mechanisms of the Acute Attack of Gout and Its Resolution

Bradykinin as a gamma globulin, has a molecular weight


of 88,000, and has been purified from human
Bradykinin is also a likely contributor to the plasma. The structure of the circulating molecule
localized heat, redness, and pain of acute gout. is a single polypeptide chain consisting of 619
Synovial fluid contains components of the con- amino acids, and cleavage of this molecule at an
tact system (Hageman factor, prekallikrein, high- arginine-isoleucine bond results in the generation
molecular-weight kininogen, and coagulation of a disulfide-bonded molecule consisting of a
factor XI) from which bradykinin is derived. heavy chain of 371 amino acids from the amino
Since the injection of monosodium urate crystals terminus of the molecule and a light chain of
into a dog joint causes the rapid generation of 248 amino acids from the carboxy-terminus. The
joint pain and heat peaking within 10 min, and active site of the molecule is localized in the light
bradykinin is a more potent vasoactive agent than chain, and the binding site for high-molecular-
high doses of prostaglandin E1 (0.1 M) or hista- weight kininogen is on the heavy chain [303].
mine (0.01 M), bradykinin may be a key media- Plasma kallikrein cleaves high-molecular-weight
tor of the early stages of acute gout. In fact, kininogen, a single-chain glycoprotein with Mr
intra-articular injections of MSU crystals in of 120,000 Da to generate the vasodilatory nona-
a patient with the gouty diathesis triggered an peptide, bradykinin.
acute episode of gout in association with a rapid In summary, the initial event in this sequence
increase in the synovial fluid concentration of of events is the activation of Hageman factor
bradykinin. which then interacts with prekallikrein bound to
The mechanism for the generation and release kininogen that serves as a substrate for factor
of bradykinin from high-molecular-weight XIIa leading to the generation of the active pro-
kininogen is well understood. The negatively tease, kallikrein. The latter then cleaves kinino-
charged surface of monosodium urate crystals gen to form the vasoactive peptide, bradykinin.
binds Hageman factor (HF/factor XII) through This small peptide increases vascular permeabil-
the positively charged amino acids in its heavy ity, dilates blood vessels, and mediates pain
chain and is subsequently activated, and this responses. Kallikrein also acts as a stimulus for
interaction initiates further interactions between neutrophil degranulation. A recent publication
factor XII, prekallikrein, and high-molecular- has indicated that bradykinin formation from
weight kininogen [301]. This process is known autoactivated factor XII on the surface of endothe-
as contact activation. Factor XII migrates as a lial cells occurs placing this vasoactive mediator
beta globulin by electrophoresis, and cleavage of close to its target, the blood vessel [304]. Contact
this molecule and the generation of activated HF activation appears not to be a component of the
(factor XIIa) occurs from exposure to plasma kal- inflammatory response of gouty arthritis since
likrein or through spontaneous autoactivation on gout has been reported in patients with a
negatively charged surfaces like uric acid [302]. deficiency of the Hageman factor. However, a
The structure of Hageman factor has been eluci- poorly characterized tissue pathway may also
dated from purified human sources and its direct generate bradykinin. Thus, bradykinin functions
amino acid sequence characterized. It consists of to cause venular dilation and activation of phos-
596 amino acids and is activated to factor XIIa pholipase A2 to increase arachidonic acid metab-
by the cleavage of a bond between arginine-353 olism and the release of prostaglandins.
and valine-354 resulting in a heavy chain of 353
amino acids and a light chain of 243 amino acids.
The heavy chain contains the site for binding Tumor Necrosis Factor
to negatively charged surfaces. Activated factor
XIIa catalyzes the conversion of prekallikrein Tumor necrosis factor (TNF) is a pleiotropic
to kallikrein. Plasma prekallikrein migrates 17-kDa protein produced primarily by monocytes
Monosodium Urate Crystal-Induced Inflammation 255

and macrophages, but other cell types including monocytes and synovial membrane lining cells
neutrophils also produce it. This mediator has may also trigger the release of proinflammatory
been documented in synovial fluids associated mediators when challenged by MSU crystals.
with acute inflammatory processes and has been Such interactions between monocytes and MSU
shown to be released from cultured monocytes crystals cause the release of IL-8, IL-1, IL-6, and
and synovial lining cells by monosodium urate tumor necrosis factor-a [103, 104, 123].
crystals. Its biological functions include the Interactions are also likely to occur between the
induction of human neutrophil degranulation, type A (monocyte-derived) synovial lining cell
and it is now well established that the p55 TNF and uric acid crystals, but these cells have not
receptor is responsible for the respiratory burst of been isolated and cultured in vitro so that their
neutrophils. This mediator also stimulates the molecular and functional capacities remain
release of secretory phospholipase A2 (sPLA2), incompletely defined.
the enzyme that regulates the synthesis of eico- A large body of data exists relating to human
sanoids and platelet-activating factor (PAF), and fibroblast-like synovial lining cells including
this action results in the rapid release of prostacy- receptors on these cells, their transduction path-
clin (PGI2), another vasoactive substance. In ways, and the effects of therapeutic agents on
addition, TNF also has a direct action on the these cells. However, the publications describing
endothelial cell adhesion molecules, ICAM-1 these parameters have been primarily restricted
(intracellular adhesion molecule) and VCAM-1 to synovial cells recovered from patients with
(vascular cell adhesion molecule), that in turn rheumatoid arthritis and osteoarthritis. Since
play a role in the interactions between leukocytes these synoviocytes may have structural and func-
and endothelium. These cell adhesion molecules tional differences from normal synoviocytes, it is
are an essential part of the mechanisms involved probably inappropriate to assume that patients
in the migration of neutrophils through blood with gout will have completely identical charac-
vessel walls during inflammatory processes. teristics to those synoviocytes recovered from
patients with nongouty forms of arthritis.
Despite these shortcomings, uric acid crystals
Monosodium Urate Crystal-Induced do activate fibroblast-like synoviocytes in culture
Inflammation and stimulate the production of proinflammatory
cytokines (IL-6, IL-8, TNF-a, and IL-1) [104,
Proinflammatory Mediators and MSU 125]. These same human cells challenged with
Crystals MSU crystals also release matrix metallopro-
teinase, collagenase as well as arachidonic acid
The deposition of monosodium urate crystals and metabolites including prostaglandins. MSU
their interaction with neutrophils are central to crystals also produce neutral proteases from
the inflammatory process associated with acute human synovial fibroblast-like cells. In addition
gouty arthritis. Such neutrophil-MSU crystal to the direct action of MSU crystals on human
interactions not only lead to the production of synoviocytes, other proinflammatory cytokines
proinflammatory mediators but also trigger the as well as MSU-induced neutrophil metabo-
activation of the biochemical pathways that regu- lites that have the capacity to modulate the
late these reactions. Such crystals can generate inflammatory response can interact with human
complement peptides, kinins, histamine, vasoac- fibroblast-like synoviocytes. Thus, the presence
tive prostaglandins, platelet-activating factor as of proinflammatory cytokines or modulators of
well as cytokines such as IL-1, TNF-a, IL-8, IL-6, synoviocyte-mediated inflammatory events in the
oxygen free radicals, and phospholipase synovial fluid have the potential to regulate syn-
A2-activating protein (FLAP) [102, 104, 124]. In ovial fibroblast responses associated with acute
addition to neutrophii-MSU crystal interactions, gouty episodes and their subsequent resolution.
256 8 Mechanisms of the Acute Attack of Gout and Its Resolution

Regulation of Synovial inappropriate to conclude that such findings can


Fibroblast-Like Cells be assumed to occur in synovial fibroblast-like
cells in patients with gout. On the other hand, the
As noted previously, IL-1 and TNF-a are the main findings in synovial fibroblast-like cells from
proinflammatory cytokines generated in acute patients with osteoarthritis and musculoskeletal
gout, and these cytokines regulate a number of trauma are not likely to retain fixed lesions in
type B synoviocyte functions. Tumor necrosis their structure and function. Thus, some of the
factor-a induces the expression of vascular cell data cited in the literature is based on findings
adhesion molecule (VCAM) on human syn- from synovial fibroblast-like cells recovered from
ovial fibroblasts and triggers the synthesis and/ patients with osteoarthritis and musculoskeletal
or expression of PGE2, IL-6, hyaluronic acid, trauma and may of may not reflect those responses
interleukin-1, granulocyte colony-stimulating present in the “normal” synovial fibroblast.
factor (G-CSF), granulocyte-macrophage col-
ony-stimulating factor (GM-CSF), macrophage
colony-stimulating factor (M-CSF), intercel- MAP Kinase and MSU Crystal-Induced
lular adhesion molecule (ICAM)-1, monocyte Neutrophil Activation
chemoattractant protein-1 (MCP-1), collagenase,
and interleukin-10. TNF-a also regulates the The most complex and significant aspect of
expression of the constitutive gene for I kappa b crystal-induced reactions is the identification
kinase-1 (IKK-1) in human fibroblast-like syn- and characterization of the pathways leading to
oviocytes [305]. This kinase plays a central role in neutrophil activation, the mechanisms by which
the regulation of nuclear factor-kappa B (NF-kB) activation is regulated, and the cellular functions
that controls expression of genes of significance that are mediated by such activation. Particulate
to the inflammatory response. Interleukin-1, phagocytosis and proinflammatory cytokines
another key effector of human fibroblast-like syn- such FMLP and TNF-a have been identified
oviocyte function, stimulates the synthesis and/or as activators of p38 mitogen-activated protein
expression of many proinflammatory components kinase (MAPK), but a complete picture of this
similar to those affected by TNF-a such as colla- pathway and its functional role remains to be
genase, PGE2, GM-CSF, IL-6, G-CSF, M-CSF, determined. Evidence supports a role for MAP
IL-10, IL-10R, and MCP-1 [306]. Interleukin-1 kinases in chemoattractant-induced degranula-
also regulates the expression of stromelysin- tion responses, the assembly of NADPH oxidase
1(MMP-3) and tissue inhibitor of metallopro- and the subsequent activation of the respiratory
teinases (TIMP-1). Like TNF-a, interleukin-1 burst, the synthesis and expression of IL-8 activ-
also regulates I kappa B kinase activity. Further, ity, and neutrophil apoptosis [307]. The regula-
IL-1 and TNF-a act synergistically to induce the tion of human neutrophil IL-8 synthesis requires
synthesis and release of IL-6. Interleukin-1 and p38 MAP kinase for the TNF-a-mediated produc-
bradykinin also act synergistically to increase the tion of IL-8. Since TNF-a induces the activation
synthesis and release of PGE2. and nuclear translocation of the transcription fac-
No detailed studies of “normal” synovial tor, NF-kB, and the IL-8 gene has NF-kB binding
fibroblast-like cells have been undertaken, but sites essential for the transcriptional activation
numerous investigations have been published response to TNF-a, it seems reasonable to pro-
with respect to the functions and responses of pose that TNF-a appears to activate MAP kinase
human synovial fibroblast-like cells recovered which then activates NF-kB to cause IL-8 gene
from patients with rheumatoid arthritis, osteoar- expression. Thus, MAP kinase is a key regulator
thritis, and musculoskeletal trauma. Many of of gene expression [308]. Even though the MAP
these publications have focused on rheumatoid kinase and other pathways regulate neutrophil
synoviocytes and have documented many diverse functions known to be triggered by MSU crys-
findings related to these cells. Without any nor- tals, the direct relationship of these pathways to
mal standard to compare with these cells, it is such crystals has not been defined.
Monosodium Urate Crystal-Induced Inflammation 257

Phospholipase D and MSU Crystals the regulation of TNF-a-induced apoptosis by


CPPD crystals have begun to be characterized,
MSU and CPPD crystals also generate messages but pathways for the delay of apoptosis by MSU
via phospholipase-specific phospholipase D. This crystals have yet to be completely defined. Fifth,
enzyme hydrolyzes phosphatidylcholine and phospholipase D activity and ADP-ribosylation
forms phosphatidic acid, a significant lipid medi- factor 1 (ARF1) are associated with lysosomal
ator, and the water soluble product, choline. One membranes in neutrophils and HL-60 neutrophil-
product of this enzyme reaction, phosphatidic like cells. ARF1 is able to restore lysosomal
acid (PA), may also be converted to other second secretion. More recent investigations have postu-
messengers such as 1,2-diacylglycerol and lyso- lated that ADP-ribosylation factor 6 (ARF6) is
phosphatidic acid. Mammalian cells contain two the key metabolite in receptor-mediated NADPH
separate genes for phospholipase D designated oxidase activation in a PLD-dependent manner.
phospholipase D1 (PLD1) and phospholipase D2 The role of other ARF isomers in phagocyte
(PLD2). Two splice variants of PLD1 have also function remains to be determined, but it has
been identified and designated as PLD1a and recently been shown that ARF has a major role in
PLD1b. Another oleate-activated phospholipase the signaling between the fMLP receptor and
D isoform has also been identified, but it has yet NADPH oxidase [312]. Thus, phospholipase D
to be cloned and characterized further. has a regulatory role in the assembly and activa-
Phospholipase D activity has been implicated tion of NADPH oxidase as well as an incom-
in a variety of cellular functions including cell pletely characterized role in lysosomal enzyme
secretion, superoxide generation, and immune secretion. Further, phospholipase D is directly or
responses. As indicated previously, MSU crystals indirectly associated with the generation of sec-
trigger a number of reactions essential for neutro- ond messengers such as calcium, phosphatidic
phil functions. First, MSU crystals increase the acid, and diacylglycerol that control specific neu-
intracellular free calcium ion concentration that trophil functions.
causes the hydrolysis of phosphatidylinositol Since the phospholipase D of human neutro-
4,5-biphosphate by a specific phospholipase C phils is activated by MSU crystals and plays a role
with the resultant formation of inositol [1, 4,] in neutrophil functions triggered by MSU crys-
triphosphate. Second, the product of phospholi- tals, factors regulating phospholipase D activity
pase D, phosphatidic acid, can be further metabo- are of significance. The key regulators of PLD
lized to diacylglycerol (DAG), and these two activity are small GTPases such as ADP-
lipid second messengers (DAG and PA) interact ribosylation factor 1 and its isomers as well as
directly with NADPH oxidase components to Rho proteins and Ral 1. Recent data have also
cause the assembly of this enzyme and its subse- shown that phospholipase D1 is phosphorylated
quent activation [309]. These same metabolites by a casein-kinase-2-like serine kinase. This
also regulate phosphatidic acid-activated protein casein kinase-2-mediated phosphorylation of
kinase and protein kinase C isoforms. Third, phospholipase D1 occurs on cell membranes and
phosphatidic acid has also been shown to regu- is probably responsible for the localization of the
late a specific tyrosine kinase (Fgr), but the func- enzyme (PLD1), its substrates, and its regulators,
tional role of this tyrosine kinase is incompletely to the cellular membranes. Since PLD via its
characterized. Even though the role of the tyrosine immediate and more distal products (phosphatidic
kinase, Fgr, is not known, MSU crystals are acid, diacylglycerol, and lysophosphatidic acid)
known to induce tyrosine phosphorylation and regulate the respiratory burst, ligand-induced
perhaps this enzyme is involved [310]. Fourth, secretion, prostaglandin synthesis and release,
phospholipase D activity appears to result in a cytokine release, and cytoskeletal rearrangements,
prolonged survival of neutrophils by delaying this enzyme has been and will continue to be a
apoptosis [311]. Studies of the latter phenomenon target for potential therapeutic agents with PLD
show that MSU crystals inhibit both spontaneous inhibitory activity. Neutrophils are also stimulated
and TNF-a-induced apoptosis. The pathways for by protein kinase C isoforms such as PKC-a and
258 8 Mechanisms of the Acute Attack of Gout and Its Resolution

PKCb11 [313]. Thus, phospholipase D and its chemokine and cytokine synthesis triggered by
products play a key role in mediating certain neu- MSU crystals. MSU crystals also induce CD18
trophil functions as well as generating second (b2 integrin)-independent and selectin-indepen-
messengers for neutrophil-associated responses dent adhesion of neutrophils to endothelial cells.
Thus, ARF nucleotide binding site opener
(ARNO) and perhaps cytohesion-1, another gua-
MSU Crystals and Other Phospholipids nine nucleotide exchange factor, support the con-
Metabolizing Enzymes version of nucleotide-bound ARF and in this way
activate phospolipase D. This activation sequence
Both MSU and CPPO crystals also activate phos- then activates protein kinase C causing the cou-
phatidylinositol 3-kinase activity, and such acti- pling of FcyRI to phospholipase D1. The exact
vation is a key to a transduction pathway for relationships between ARNO, cytohesion-1,
neutrophil respiratory bursts and degranulation ARF1, ARF6, PKC-a, and MSU crystal-induced
responses. MSU crystals also stimulate phospho- neutrophil phagocytosis remains to be completely
lipase A2 activity with the resultant production of resolved. Recent studies have shown that the gua-
proinflammatory eicosanoids. Investigations have nine exchange factor, ARNO, is stimulated by the
also shown that MSU crystals induce the synthe- chemotactic factor, fMLP, in HL-60 cells, and it
sis of PLA2-activating protein (FLAP), a modu- is likely that similar reactions will occur with
lator of arachidonic acid-mediated inflammatory other chemotactic factors such as IL-8.
responses. Recent studies have demonstrated that Another pathway of interest is the protein
PLA2 and phosphatidic acid cause the fusion of tyrosine phosphatase MEG2 (megakaryo-
neutrophil granules and plasma membranes. cyte 2) that regulates secretory vesicles in
Thus, phospholipid metabolism, especially that hematopoietic cells and is also associated with
mediated by phospholipases A2 and D, may play diphosphate > phosphatidylinositol 3,4,5-triphos-
a central role in neutrophil degranulation. phate > phosphatidylinositol 4-phosphate) and is
Furthermore, U73122, a specific inhibitor of stimulated by opsonized zymosan and phorbol
phospholipase C, inhibits MSU crystal-induced 12-myristate 13-acetate. This enzyme, protein
superoxide generation and neutrophil degranula- tyrosine phosphatase MEG2, is unaffected by
tion. It has been postulated that PLC gamma2 is the chemoattractant, fMLP. On this basis, there
the PLC isoform involved in this signal transduc- is speculation that protein tyrosine phosphatase
tion pathway. Unopsonized MSU crystals can MEG2 may regulate, in some manner, phagocy-
also activate neutrophils by inducing pertussis tosis, but the complete signaling pathway and its
toxin-insensitive phosphatidylinositol bisphos- ultimate role in neutrophil function remains to be
phate (PIP2) hydrolysis with the generation of determined.
inositol [1, 4,] triphosphate and the mobilization Tec kinases represent a large group of non-
of calcium. This pathway modulates superoxide receptor tyrosine kinases that may be involved in
generation and release. the transduction of cell surface receptor mes-
sages. The structure of these cytoplasmic tyrosine
kinases includes proline-rich regions and pleck-
Other Pathways Stimulated by MSU strin homology (PH) domains for their regulation
Crystals and activation. The PH domain is able to bind
phospholipids and heterotrimeric G proteins.
Two additional MSU crystal-induced reactions These Tec kinases are activated by interaction
have been characterized. First, MSU crystals with many cell-surface receptors, integrins, and
stimulate the phosphorylation of SAM68 (sarc- G protein-coupled receptors [314]. The interac-
associated in mitosis 68 kDa). This component is tions with cell-surface receptors have recently
involved in the signal transduction and activation been shown to include chemotactic agents.
of RNA and may regulate specific patterns of Since Tec kinase activation is blocked by
Monosodium Urate Crystal-Induced Inflammation 259

phosphatidylinositol 3-kinase inhibitors, the neutrophii-MSU crystal interactions, monocytes


product of this enzyme, phosphatidylinositol and synovial lining cells also trigger the release
3,4,5-triphosphate, may be critical for Tec kinase of inflammatory mediators when challenged by
activation. This is consistent with data showing MSU crystals. Such interactions between mono-
the binding of such phosphoinositides to the PH cytes/synovial lining cells cause the release of
region of some Tec kinases. In addition to their IL-8, IL-1, IL-6, and tumor necrosis factor-a.
role in neutrophil chemotactic activity, Tec
kinases may signal the activation of MAP kinases,
actin rearrangement, and transcription activities. MSU Crystals and Cell Membrane
All these MSU crystal-induced reactions as Interactions
well as other transduction pathways described in
the neutrophil may be potential sites for interac- MSU crystals are rapidly phagocytosed by neu-
tion with modulators that can modify the biologi- trophils and engulfed by phagolysosomes. Since
cal effects of these pathways. Although excellent lysosomes contain proteolytic enzymes capable
pharmacologic agents are already available for of digesting the proteins coating crystals, the neg-
the treatment of acute gout, newer agents may ative surface of crystals can hydrogen bond with
offer better treatment regimens for the patient the phospholipid head groups of the lysosomal
intolerant to presently available drugs or those membranes [314]. Monosodium urate crystals
patients with contraindications to the standard that are weakly acidic appear to hydrogen bond
drugs used to treat gout. Of course, there is always with the phosphate esters of phospholipids and
the possibility that newer drugs might be more cause the perforation of the organelle with the
potent or more rapidly acting agents than those resultant release of lysosomal enzymes and cell
therapeutic agents presently available. cytoplasmic contents. Studies of serum proteins
absorbed to MSU crystals clearly demonstrate
why immunoglobin-coated crystals do not cause
MSU Crystal-Induced Proinflammatory membranolysis of the plasma membrane of neu-
Mediators trophils since protein-coated crystals have a pro-
tective effect on plasma membranes via these
In addition to oxidants and the exocytosis of absorbed proteins. MSU crystal-induced neutro-
phagolysosomal contents such as lysosomal phil membranolysis and cytolysis raises the ques-
enzymes, MSU crystals can generate a number of tion of the fate of the urate crystals released by
proinflammatory mediators including complement this process. There are at least two enzymes, per-
peptides, kinins, histamine, vasoactive prosta- oxidase and cytochrome oxidase, that have the
glandins, platelet-activating factor, and cytokines capacity to digest uric acid. Probably the major
such as IL-1, TNF-a, IL-8, IL-6, and phospho- route for MSU crystal destruction is via the per-
lipase A2-activating protein (FLAP) via MSU oxidative presence of a monosodium urate tophus
crystal-neutrophil interactions. The deposition of in the synovial membrane may delineate another
monosodium urate crystals and their interaction possible site for the removal and storage of crys-
with neutrophils are central to the inflammatory tals [42]. Finally, free uric acid crystals are often
process associated with acute gouty arthritis. Such observed in the synovial fluid long after the acute
neutrophil-MSU crystal interactions not only lead episode of gout has subsided. Why these free
to the production of proinflammatory products urate crystals do not evoke an acute inflammatory
but also trigger the activation and regulation of the response is presently unknown, but perhaps it is
biochemical pathways that regulate these reac- the absence of an immunoglobin coating and the
tions. The proinflammatory products generated by inability to interact with Fe receptors.
MSU crystal-neutrophil interactions include lyso- In summary, the phagocytosis of IgG-coated
somal enzymes, interleukin-1, interleukin-8, oxy- uric acid crystals is an exceedingly complex
gen free radicals, and eicosanoids. In addition to process and remains incompletely characterized.
260 8 Mechanisms of the Acute Attack of Gout and Its Resolution

It involves a complex set of molecular interactions is the key to the resolution of inflammation and
by the neutrophil to surround the crystal and even- fibrosis. Even though apoptosis occurs as an
tually ingest it fully. Such a response necessitates abnormal balance between cell survival and cell
the use of actin filaments to guide the changes in death in some diseases, it is, for the most part,
the cell protrusions in association with contractile considered as a process of physiologic cell death.
proteins, and to surround the crystal completely The absence of inflammatory responses in asso-
involves fusion and fission of the membranes. The ciation with apoptosis distinguishes it from
key components for the ingestion and disposal necrosis and other forms of cell death. Caspases
of uric acid crystals are dependent on NADPH are cysteine proteases that play a central role in
oxidase, myeloperoxidase, and hydrogen per- the execution of the apoptotic process [315]. Two
oxide. These oxidant-producing systems lead to major pathways can be utilized by neutrophils
the breakdown of the purine ring and the genera- and macrophages to activate caspases and cause
tion of more soluble molecules such as allantoin cell death. The first pathway or so-called death
and urea once the phagosome has fused with receptor pathway is an extrinsic route to cell
the lysosome and the release of such oxidants is death by the interaction of certain cytokines of
initiated. If bacterial infections accompany the the TNF/nerve growth factor family with their
acute episode of gout, the killing of bacteria is, in cognate receptors (TNFR, CD95R, and TNF-
part, dependent on these same systems. It seems related apoptosis-inducing ligand receptor). Once
unlikely that therapeutic agents to alter these pro- these interactions take place, an adaptor molecule
cesses would lead to significant improvements in (TNF receptor-associated death domain or FAS-
the treatment of gout. Nonetheless, an increment associated protein with a death domain) binds to
in the disposal of apoptotic neutrophils and their the receptor, and these complexes can bind and
ingested uric acid crystals might enhance the res- activate specific caspases directly The second
olution of the acute gouty episode. Thus, a dis- pathway is an intrinsic one and is mediated by the
cussion of apoptosis, the key process involved in release of cytochrome c from the cell mitochon-
the phagocytosis of such dying cells, is described dria into the cytoplasm. Cytosolic cytochrome c
in the succeeding sections. interacts with apoptotic protease-activating fac-
tor 1 (Apaf-1) to result in the oligomerization of
Apaf, and this complex then recruits caspase-9.
Neutrophil and Macrophage Once this so-called apoptosome (Apaf 1 and pro-
Apoptosis caspase-9) is activated, it can then interact with
and activate other caspases. The third pathway
Overview that can activate caspases is of no significance to
this discussion since it involves the use of gran-
Apoptosis is a term used to describe a morpho- zyme B from the granules of cytotoxic T cells/
logical process characterizing cell death by a par- natural killer cells which are not involved in the
ticular series of events quite different from cell acute gouty attack. The key to the destructive
necrosis. The apoptotic cell appears shrunken processes associated with apoptosis are the
with membrane blebbing, has nuclear chromatin cysteine proteases called caspases.
condensation and DNA fragmentation into
nucleosome-length fragments, and these changes
in cell membranes cause phosphatidylserine and Endogenous and Exogenous
oxidized phospholipids to take their place on the Anti-inflammatory Agents
external surface of the cell. The term, apoptosis,
comes from the Greek meaning “to fall away Overview
from.” This apoptotic phenomenon is a carefully
orchestrated and genetically controlled event of Immune and inflammatory reactions represent
significance to the inflammatory process observed physiological mechanisms by which the human
in gout and other inflammatory disorders since it host responds to tissue and organ-damaging
Endogenous and Exogenous Anti-inflammatory Agents 261

stimuli. Since excessive expression or inappro- including blocking the activation of the transcrip-
priate activation of these reactions may cause cell tion factor, nuclear factor KB (NF-KB), that reg-
and tissue damage, homeostatic mechanisms ulates proinflammatory cytokines, chemokines,
exist to protect the host from such damaging and cell adhesion molecules [319]. The activa-
effects and to regulate tightly the expression of tion of NF-KB requires phosphorylation and pro-
these inflammatory and immune responses. Acute teolytic degradation of its inhibitor, IKB, with the
episodes of gout are known to resolve without subsequent release of NF-KB, its translocation to
treatment suggesting the presence of endogenous the nucleus, and its binding to DNA sequences in
anti-inflammatory mechanisms. The literature the promoters of the target genes to initiate their
supports the view that the human host has the transcription. The proteolytic degradation of IKB
capacity to turn off acute inflammatory processes occurs via the proteasome pathway, and the inhi-
and to cause their resolution [316, 317]. Uric acid bition of this pathway would, in turn, stabilize
crystals and in some settings, accompanying bac- IKB and maintain its capacity to bind to NF-KB
terial pathogens, are the stimuli that trigger the in the cytoplasm preventing NF-KB activation.
acute inflammatory response in acute gout. The Since proteasome inhibitors have been utilized as
critical elements of the proinflammatory mecha- NF-kB activation inhibitors, a brief review of this
nisms associated with these stimuli are primarily mechanism follows. However, these small, non-
the gene transcription factors, nuclear factor- selective NF-KB inhibitors may activate NF-KB
kappa S (NF-KB), activator protein-1 (AP-1), in some cells and may also inhibit other activities
and STATs (signal transducers and activators of unrelated to the inflammatory response [320].
transcription (STATs) [318]. These transcription The nonspecific nature of these proteasome
factors regulate a wide variety of genes associ- inhibitors suggests that their use may be limited
ated with inflammatory and immune responses in their capacity to impair acute inflammatory
including proinflammatory cytokines, cytokine responses.
receptors, chemotactic proteins, and adhesion Antisense technologies represent a more
molecules. The presence of sites for TNF-a, potent means of investigating gene function for
IL-1b, IL-8, macrophage chemotactic protein-1 the future design of more specific treatment
(MCP-1), granulocyte-macrophage colony stim- approaches for diseases like gout through their
ulating factor (GM-CSF), intercellular adhesion capacity to block the expression of mRNAs
molecule-1 (ICAM-1), vascular adhesion mole- associated with inflammation. Such initiatives
cule-1 (VCAM-1), E-selectin, and others on the are entirely dependent on functional genomics
promoter or regulatory motifs of these transcrip- since a specific treatment must be based on the
tion factors emphasize the key role of transcrip- inhibition of a specific mRNA. The major groups
tion factors in the regulation of inflammation. of sequence-specific nucleic acids designed to
Several different approaches have also been block mRNA expression include antisense oli-
utilized to understand how an inflammatory gonucleotides (ODNs), ribozymes, DNAzymes,
response can be modulated to cause its resolu- and RNA interference (RNAi). Such antisense
tion. Homeostatic mechanisms that regulate the technologies must be delivered efficiently to a
response of the human host to inflammatory cell, have stability and resistance to nuclease
stimuli identify the pathways utilized to suppress activity, have little, if any, activity against sites
inflammation as well as the key components of other than the specific target mRNA, must be
such pathways. As has been discussed, receptor nontoxic to the cell, and must reduce the target
desensitization to proinflammatory agonists is an mRNA specifically. Here again, such antisense
effective mechanism by which inflammatory technology as well as antigene strategies using
responses can be suppressed. In addition, the DNA triplex technology (vide infra) may be lim-
nuclear factor-kappa B represents an ideal target ited in their use in acute gout since fulfilling all
for inhibition since it activates inflammatory gene the criteria for an effective and efficient anti-
expression. Several different mechanisms have sense or antigene strategy may be difficult to
been shown to suppress inflammatory responses achieve.
262 8 Mechanisms of the Acute Attack of Gout and Its Resolution

The most promising technology using nucleic protein (RIP) and IKKy and their role in Nt = −KB
acids is gene silencing via RNA interference. activation, or by causing the destruction of cal-
Since many organisms use double-stranded RNA pastatin and permitting calpain to activate apop-
to silence genes during their developmental tosis. These mechanisms are reviewed in the
phases and to protect cells from viral invasion, following discussions.
transposon jumping, and for silencing introduced Finally, a variety of anti-inflammatory drugs
transgenes, this naturally occurring system and have the capacity to alter NF-KB activation. In
its effector, small interfering RNA (siRNA), has addition to the nonsteroidal anti-inflammatory
been utilized to administer exogenous siRNAs to drugs (NSAIDS), glucocorticoids also have a
cause gene-specific silencing or to provide vec- significant role in modulating the inflammatory
tor-induced expressions of siRNAs in cells as a response since they have the capacity both
means of gene silencing. Although RNAi has yet to counteract the synthesis and release of
to reach its full potential, improved virus- and proinflammatory mediators and to cause the pro-
plasmid-based delivery systems are likely to per- duction of anti-inflammatory cytokines like IL-10,
mit the delivery of siRNAs to appropriate tissue IL-4, TGF-b, and annexin-1. Glucocorticoids
sites at the right time and to characterize the role and the compounds that are induced by them
of specific genes in the expression of acute gouty represent a major negative feedback mechanism
arthritis. Such information may provide better to prevent the overexpression and activation of
targets for anti-inflammatory agents. proinflammatory cytokines and other components
Anti-inflammatory eicosanoids such as lipox- that may result in organ and tissue damage.
ins (lipoxygenase interaction products.), cyclo-
pentenone prostaglandins derived from PGD2
and its metabolites, and the resolvins derived Anti-inflammatory Eicosanoids
from eicosapentaenoic acid (C20:5) and docosa-
hexaenoic acid (C22:6) in the presence of acety- Overview
lated COX-2 and 5-lipoxygenase activity, both
represent endogenously generated eicosanoids Acute gouty arthritis is an intense inflammatory
that are involved in the resolution of inflammatory disorder primarily mediated by the precipitation
responses. These endogenously synthesized anti- of monosodium urate crystals in the intrasynovial
inflammatory agents also emphasize the role space and the recruitment of neutrophils and their
transcellular biosynthesis plays in the downregu- proinflammatory products to the involved joint.
lation of the inflammatory process. Stimulating The initial phase of this inflammatory process
the generation of these anti-inflammatory eico- is regulated by the generation of chemotactic
sanoids is a physiological means of suppressing signals for neutrophils and the expression of
inflammatory responses, and the development of chemotactic cell-surface receptors. This segment
analogs of these suppressive molecules with sim- of the discussion places emphasis not on the
ilar biological activity could be used, in the proinflammatory aspects but on those factors at
future, as therapeutic agents. play during the resolution of this inflammatory
Another means of modulating an acute response. A characteristic of acute gout not often
inflammatory response is to enhance apoptosis, a discussed as a result of the availability of effective
process that clears cellular debris from an drugs for the treatment and suppression of acute
inflamed site. For the most part, these apoptosis- gouty episodes is the fact that the human host is
enhancing mechanisms rely primarily on imped- able to resolve over time the acute inflammatory
ing the activation of NF-KB and its induction of response of gout in the absence of any treatment.
antiapoptotic molecules. This can be accom- This reparative or resolution process provides
plished by altering the capacity of IKB to release strong evidence for the presence of endogenous
NF-KB and its subsequent activation, by impair- molecular system(s) with the capacity to coun-
ing the interactions between receptor-interacting teract proinflammatory mediators and to generate
Lipoxin Biosynthesis 263

proresolution mediators. Such anti-inflammatory a molecule exhibiting both proinflammatory and


processes must account for the downregulation of anti-inflammatory effects and regulates the shift
the synthesis and release of neutrophil chemoat- from the eicosanoid biosynthesis of LTB4 and
tractants, the generation and clearance of apop- 5-lipoxygenase-derived products to the induction
totic neutrophils in a nonphlogistic manner, the of 15-lipoxygenase activity and the release LXA4.
upregulation of the synthesis and release of mono- PGE2 also upregulates the macrophage produc-
cyte/macrophage chemotactic agents, the phago- tion of IL-10, another anti-inflammatory media-
cytosis of apoptotic neutrophils by macrophages, tor [322]. The presence of four PGE receptors
and the eventual disappearance of the phagocytic (EP1 EP2, EP3, and EP4) that subtend different
macrophages from the intrasynovial space via the signal transduction pathways may contribute to
lymphatics. As might be predicted from the tasks this shift to the 15-lipoxygenase pathway. The
necessary for resolution, this reparative phase EP2 and EP4 receptors stimulate adenylate
is just as complex as the generation of an acute cyclase and increase the intracellular levels of
inflammatory reaction. If one examines the list of cyclic AMP, whereas the EP1 receptor activates
endogenous anti-inflammatory agents the resolu- phospholipase C and phosphatidylinositol turn-
tion factors are multiple, often redundant in their over. The EP3 is more complex since it has mul-
functions, and will probably result in as complex tiple isoforms as a result of EP3 gene splicing.
a process as the expression of molecules engaged Nonetheless, PGE2 via its capacity to increase
in acute inflammation. intracellular cyclic AMP stimulates phosphory-
Like proinflammatory reactions, proresolution lated CREB-1-mediated activation of 15-lipoxy-
responses have different effects, in some cases, genase transcription.
depending on the cells and tissues targeted. In Accompanying these changes induced by
addition, although animal models may be excel- PGE2 there is an alteration in the activities of
lent tools for examining the role of mediators in phospholipase A2 isoforms from a predomi-
inflammatory settings, there is no guarantee that nance of the type VI calcium-independent PLA2
identical findings will always be determined in in the acute inflammatory response to the expres-
humans. The conversion to a proresolution state sion of types IIA and V secretory phospholipase
is mediated principally by a shift from the A2s and subsequently the expression of type IV
proinflammatory prostaglandins and leukotrienes cytosolic phospholipase A2 in the resolution
to the proresolution lipoxins and cyclooxygenase- phase. In parallel with this transition to the reso-
2-derived mediators from eicosapentaenoic acid lution phase, there is an increased expression of
(EPA) and docosahexaenoic acid termed resolvins, cyclooxygenase-2 [602]. The calcium-indepen-
docosatrienes, and neuroprotectins. Additional dent PLA2 is responsible for the generation of
endogenous anti-inflammatory factors may also the proinflammatory mediators PGE2, LTB4,
contribute to this resolution process such as heme IL-1b, and PAF, whereas the type IV PLA2 in
oxygenase-1, IL-10, and others, but the principal conjunction with COX-2 generates PGD2 and
proresolution mediators identified to date are 15-deoxy-D12¢14-PGJ2, mediators that have
those derived from the prostaglandin and omega-3 anti-inflammatory activities.
polyunsaturated fatty acid pathways [321].
Two factors, prostaglandin E2 and phospholi-
pases, appear to mediate the resolution of Lipoxin Biosynthesis
inflammation by the induction of 15-lipoxyge-
nase activity and the synthesis and release of The lipoxins are lipid mediators derived from
LXA4as well as by the alteration in phospholi- cell-cell interactions and the activity of either 15-
pase isoforms leading to the expression of secre- and 5-lipoxygenase or 5- and 12-lipoxygenase
tory PLA2s types IIA and V and cytoplasmic [323]. The term, lipoxins, is derived from lipoxy-
PLA2 (type IV) in conjunction with an increase genase interaction products and indicates the
in cyclooxygenase-2 activity. Prostaglandin E2 is endogenous production of these molecules from
264 8 Mechanisms of the Acute Attack of Gout and Its Resolution

lipoxygenases. In contrast to the proinflammatory Table 8.2 Major endogenous anti-inflammatory factors
lipid mediators generated from arachidonic acid Annexin-1 and annexin-derived peptides
via cyclooxygenase-1 and cyclooxygenase-2 to Lipoxins and 15-epi-lipoxins Interleukin-10
form prostaglandins or via 5-lipoxygenase to Resolvins, docasatrienes, and neuroprotectins
form leukotrienes, the lipoxins are produced by Anti-inflammins
transcellular biosynthesis and are endogenous PGD2 and PGA1
stop signals for inflammatory responses. The lat- Inosine monophosphate
ter lipids are trihydroxytetraene-containing mol- PAF acetylhydrolase (PAF-AH) Epoxyeicosatrienoic
acids (EET) Nitric oxide, peroxynitrite
ecules synthesized in the vascular lumen by the
TGF-a (Transforming growth factor-a) IL-1 receptor
interaction between platelets and leukocytes or in antagonist (IL-1ra)
tissues via endothelial cell and leukocyte IL-13
interaction. IL-4
The first lipoxin biosynthetic pathway was High-density lipoproteins
identified in monocytes and epithelial cells and Heme oxygenase-1 and carbon monoxide
catalyzed the insertion of oxygen into arachi-
donic acid mediated by 15-lipoxygenase to
form 15S-hydroperoxyeicosatetraenoic acid or 15-epi-LXA and 15-epi-LXB4. These are the
its reduced form, 15S-hydroxyeicosatetraenoic 15R-enantiomers of LXA4 LXB4, and the lat-
acid (15S-HETE). The latter compound then ter lipoxins have been given the acronym, ATL,
serves as a substrate for neutrophil 5-lipoxy- for aspirin-triggered lipoxins. These lipox-
genase to form 5,6-epoxytetraene. The latter ins are rapidly synthesized, often enhanced by
compound is unstable and leads to the genera- proinflammatory cytokines and their upregula-
tion of trihydroxytetraenes via leukocyte epox- tion of 5-lipoxygenase or cyclooxygenase, and
ide hydrolases. Two trihydroxytetraenes result are rapidly inactivated by oxireductases after
from this biosynthetic pathway: lipoxin A4 their local effect has occurred [324].
(5S, 6R, 15S-trihydroxy-7,9,13-trans-11-cis- Lipoxins have documented to be present in
eicosatetraenoic acid or lipoxin A4) and lipoxin inflammatory disorders, and a variety of anti-
B4 (5S, 14R, 15S-trihydroxy-6,0,12- trans-8-cis- inflammatory actions have been determined for
eicosatetraenoic acid or LXB4). these molecules and their stable analogs using
A second pathway for the synthesis of lipox- both in vitro and in vivo systems. Such in vitro
ins is through the generation of leukotriene and in vivo effects are summarized in Table 8.2.
from arachidonic acid catalyzed by neutrophil The in vivo studies of lipoxins have addressed
5-lipoxygenase. LTA4 taken up by platelets and primarily organ-specific changes and do not
interacts with platelet 12-lipoxygenase leading include any evaluations of the synovial mem-
to the formation of 5S,6S,15S-epoxytetraene brane, but it is likely that such lipoxin-induced
and subsequently to and LXA4 and LXB4. The changes may well be relevant to gout. The data
third major pathway for lipoxin synthesis is in clearly demonstrate that LXs, ATLs, and their
perhaps the most interesting since it involves nontoxic analogs inhibit leukocyte trafficking
another anti-inflammatory agent, aspirin. In the via an alteration in chemotaxis, cell adhesion,
presence of aspirin, cyclooxygenase-2 (COX-2) and neutrophil transmigration. They also enhance
in endothelial cells, epithelial cells or mono- the phagocytic clearance of apoptotic cells. Such
cytes is acetylated and blocks the formation of in vitro and in vivo data document the role of
proinflammatory prostaglandins. This inhibi- these agents in resolving inflammatory responses
tory activity shifts the conversion of arachi- and emphasize the broad therapeutic effects of
donic acid from prostaglandin synthesis to aspirin. In summary, lipoxins have been identified
15R-hydroxyeicosatetraenoic acid. Leukocyte as endogenous regulators of inflammation since
5-lipoxygenase then converts 5-HETE to they appear to act as brakes for the inflammatory
5S,6S,15R-epoxytetraene and subsequently to response.
Lipoxin Biosynthesis 265

Anti-inflammatory Effects of Lipoxins by mononcyte-derived macrophages LXA4-


triggered phagocytosis has been shown to be
The anti-inflammatory effects of lipoxins are optimal at 10–9 M concentrations (EC50 = ~ 0.5
summarized in Table 8.2 and such data empha- × 10 − 9 M), and this concentration is consistent
size the broad spectrum of effects mediated by with the EC50 for LXA4 of 8.3 × 10−10 observed
these molecules. A few of these effects deserve in the THP-1 cell line derived from acute mono-
further discussion. As noted previously, another cytic leukemia cells. Thus, lipoxin(LX), aspirin-
interesting offshoot of the inflammation regula- triggered lipoxin (ATL), 15-epi-lipoxin B4
tory aspects of lipoxins is the induction of (15-epi-LXB4), and a stable synthetic analog
15-epi-lipoxin A4. by aspirin [325]. Aspirin is of lipoxin, 15(R/S)-methyl-LXA4, all promote
well known to acetylate the active site of cycloox- macrophage phagocytosis of apoptotic human
ygenase-2 in endothelial and epithelial cells neutrophils in vivo.
[326]. Such COX-2 acetylation leads to the syn- This macrophage phagocytic process is not
thesis of (15R) hydroxy-eicosatetraenoic acid dependent on phosphatidylserine membrane
(15R-HETE). The latter molecule is then con- expression but depends on the expression of avb3-
verted to 15-epi-lipoxin by leukocyte 5-lipoxyge- CD36 complex. This adds significance to the
nase by a transcellular mechanism. Such a redundancy of receptors utilized for the inges-
mechanism appears to be present in humans since tion of apoptotic cells by the macrophage. Now
aspirin causes an increase in urinary 15-epi- two receptors for lipoxins have been described :
lipoxin levels in humans [213]. This latter metab- FPRL-1 and FPRL-2. Incubation of macrophages
olite (15-epi-lipoxin) causes an increase in the with apoptotic neutrophils also causes a sixfold
plasma levels of nitrite. This increment results increase in the release of TGFb, another anti-
from both constitutive nitric oxide synthase inflammatory mediator. Furthermore, PKC, Pl3-
(eNOS) and inducible nitric oxide synthase kinase, and phosphatase inhibition decreases
(iNOS) [214]. Aspirin has also been found to macrophage phagocytosis of apoptotic neutro-
reduce the inflammatory response in correlation phils. Finally, the fact that in some instances,
with an increase in plasma nitric oxide in rats lipoxins suppress neutrophil chemotaxis, yet
with carrageenin-induced pleurisy. Since nitric they activate monocyte chemotaxis gives rise
oxide appears to have a biphasic effect, the level to the question as to how such differences in
of NO may have a significant role on its regula- chemotaxis occur. One explanation offered for
tory effects. At low levels of NO produced by these opposite actions on the monocyte and neu-
eNOS, NF-kappa B is activated, whereas at high trophil is the possibility that they utilize different
levels of NO typical of iNOS induction, receptors. This receives support from the fact that
NF-kappaB is inhibited. Thus, the use of stable FPRL-2 is not present on the neutrophil but is
analogs of 15-epi-lipoxin holds promise as a way expressed on the monocyte/macrophage. Recent
to cause anti-inflammation when such agents are investigations have characterized some of the
under strictly controlled delivery [214]. anti-inflammatory effects of LXA4. on human
Of some interest to this discussion of lipoxins synovial fibroblasts and determined their action
and the resolution of the inflammatory response occurred via the LXA4receptor. In these stud-
is the fact that LXA4, at very low concentrations, ies, IL-1b and TGF-b2 upregulated the expres-
decreases LTB4-and fMLP-mediated calcium sion of LXA4R mRNA. In addition, the effects
mobilization, and such interference with cal- of LXA4 were inhibited by antibodies against its
cium mobilization impedes neutrophil chemot- receptors.
axis, transmigration, adhesion, degranulation, Another role for the arachidonic acid
and superoxide generation. Of even greater metabolite, 15-hydroperoxyeicosatetraenoic acid
interest to the resolution of gout is the fact that (15-HPETE) has recently been described [215].
lipoxins and their stable analogs stimulate non- These studies suggest that 15-HPETE appears
phlogistic phagocytosis of apoptotic neutrophils to destabilize TNFmRNA by interacting with
266 8 Mechanisms of the Acute Attack of Gout and Its Resolution

a protein_bound to the AU-rich element of acid (C22:6) and aspirin. As has been shown
TNFmRNA. In summary, the existence of anti- arachidonic acid is metabolized by aspirin-acety-
inflammatory molecules with the potential for lated cyclooxygenase/lipoxygenase interactions
modulating the inflammatory response in humans to the epi-lipoxins. When eicosapentaenoic acid
is confirmed by the foregoing studies. Neutrophils (EPA) or docosahexaenoic acid (DHA) are substi-
clearly are programmed for cell death based on tuted for arachidonic acid, DHA is converted to
their short lifespan, but this short lifespan may be 17R-hydroxydocosahexaenoic acid (17R-H(p)
prolonged by proinflammatory substances and DHA) and subsequently reduced to its correspond-
antiapoptotic molecules. The balance between ing alcohol. The action of leukocyte 5-lipoxyge-
proapoptotic and antiapoptotic proteins is the key naseleadstothegenerationof7(8)-epoxy-17R-DHA
to the route the neutrophil takes either for sur- or 4(5)-epoxy-17R-HDHA. These two precursors
vival or death. It is likely that the induction of lead to the formation of four different resolvins:
proapoptotic proteins or the design of nontoxic, 7S, 8, 17R-triHDHA and 7S, 16, 17R-triHDHA
stable analogs of these proapoptotic proteins will (resolving D1 and D2, respectively) from
be useful as agents to enhance the clearance of 7(8)-epoxy-17R-DHA and 4S, 11, 17R-triHDHA
neutrophils from the inflamed site and may be of and 4S, 5, 17R-triDHA (resolvins D3 and D4,
benefit shortening the acute episode of gout. respectively) from 4(5)-epoxy-17R-HDHA. In the
case of human endothelial cells treated with aspi-
rin in the presence of EPA, the latter molecule is
Proinflammatory to Anti-inflammatory then converted to 18R-hydroxyeicosapentaenoic
Switch acid and 15R-hydroxyeicosapentaenoic acid.
These molecules are further metabolized to gener-
There are two phases to the inflammatory ate 15R-LX5 and 5,12,18R-hydroxyeicosapen-
response with respect to lipid mediators as defined taenoic acid. The latter compound has been given
by the murine air pouch model of inflammation. the common name, resolvin E1. A number of
All these experiments support the theory that recent publications have reviewed these biosyn-
15d-PGJ2 and perhaps other PGD2 metabolites thetic pathways and the properties of their prod-
have anti-inflammatory effects on the cellular ucts [316, 327].
responses known to be associated with resolving
gout. Additional experiments using animal mod-
els of acute crystal-induced gout need to be com- Resolvin Receptors (Fig. 8.8)
pleted so that chondrocyte and synoviocyte
responses can be evaluated in the presence of The specific receptors for the resolvins are desig-
15d-PGJ2 injected into crystal-induced (MSU) nated as resolvin D receptors (ResoDR1), resol-
inflammation in animal joints. vin E receptors (ResoER1), and neuroprotectin D
receptors (NPDR). The characterization of such
receptors is in its early stages, but the definition
Resolvin Biosynthesis (Fig. 8.7a, b) of such receptors holds the promise of the devel-
opment of stable agonists that would trigger the
The trivial name, resolvin, arises from the fact that synthesis of resolvins and enhance the resolution
they are resolution phase interaction products. The of an acute inflammatory process. The resolvins
compounds derived from eicosapentaenoic acid and protectins are a new family of lipid mediators
are designated as the E series of resolvins, whereas derived from omega-3 polyunsaturated fatty acids
the resolvins derived from docosahexaenoic acid that promote the resolution of inflammatory reac-
are designated as the D series of resolvins. The tions. These publications document the potent
biosynthesis of the so-called resolvins occurs in anti-inflammatory activities of these resolvins
the presence of omega-3 fatty acids such as eicos- and protectins, and the design of stable analogs
apentaenoic acid (C20:5) and docosahexaenoic of these endogenous anti-inflammatory agents as
Lipoxin Biosynthesis 267

well as their transductive pathways offers the mediators such as IL-1b, TNF-a, and IL-8. Both
possibility of acquiring potent anti-inflammatory resolvin D and E also have the capacity to sup-
therapeutic drugs. press neutrophil recruitment to inflammatory
In summary, the resolution of an acute sites. In addition, annexin 1, a protein induced by
inflammatory process is a complex, multifaceted glucocorticoids, and its N-terminal peptides also
process that requires three critical parameters: diminish neutrophil extravasation and suppress
the suppression of neutrophil recruitment, the the inflammatory response. An intriguing aspect
disposal of apoptotic cells including other dead of these anti-inflammatory molecules is that one
cells and cellular debris, and the suppression of receptor, ALXR, is utilized by both annexin 1
antiapoptotic mechanisms. Lipoxins and their and lipoxin A4 as well as other endogenous pro-
stable analogs inhibit neutrophil infiltration from teins that regulate the pathways engaged by these
the vascular compartment to inflamed sites as anti-inflammatory vectors. Interestingly, the
well as blocking the expression of proinflammatory resolvin receptors await complete characteriza-

a COOH

Eicosapentaenoic acid

HOOC

Fig. 8.7 (a, b) In addition to O(O)H


arachidonic acid, omega-3 18-Hydroperoxyiecosapentaenoic acid
fatty acids such as
COOH
docosahexaenoic acid (DHA)
and eicosapentaenoic acid
(EPA) are converted by
acetylated COX-2 to potent
anti-inflammatory agents
designated as resolvins.
Human neutrophils via this
reaction yield 17R-DHA, and OH
O
endothelial cells by the same
reaction yield 18R-EPE. The
following nomenclature has
been adopted for these 5s, 6R-epoxy, 18R-hydroxy-eicosapentaenoic acid
trihydroxy products: 7S, 8, COOH
17R-triHDHA is resolvin D1; HO
7S, 16, 17R-triHDHA is
resolvin D2; 4S, 11,
17R-trihydroxyHDHA is
resolvin D3; 4S, 5,
17R-trihydroxyHDHA is OH
resolvin D4; and 5S, 12R,
18R-trihydroxy-EPE is
resolvin E1. 4(5),
17R-HDHA is OH
4(5)-epoxy-17R-HDHA 5S,12R,18R-trihydroxyeicosapentaenoic acid
268 8 Mechanisms of the Acute Attack of Gout and Its Resolution

b
COOH

Docosahexaenoic acid

HOOC

O(O)H
17R-H(p) DHA

COOH

4(5)-17R-H DHA

HO
COOH COOH

OH

OH OH
OH

HO

4S, 5,17R-triHDHA 4S, 11,17R-triHDHA

Fig. 8.7 (continued)


Lipoxin Biosynthesis 269

Specialized Pro-Resolving Mediators

Stop PMN transmigration and Non-phlogistic monocyte recrutment


chemotaxis, brake eosinophils
SPM Uptake and removal of apoptotic PMN and
General Actions Block prostaglandins and leukotrienes microbial particles by macrophages
Reduce cytokine release and function Enhance anti-microbial defense mechanisms
(TNFα) and clearance at mucosal surfaces

Precursors Arachidonic acid Eicosapentaenoic acid Docosahexaenoic acid


(AA) (EPA) (DHA)

E- Series D- Series Protectins/


Families Lipoxins Resolvins Resolvins Neuroprotectins

HO COOH
OH OH
HO OH
COOH COOH
HO OH OH COOH
OH OH

OH COOH HO HO OH
LXA RvE1 RvD1 RvD2 PD1/
NPD1
PMN-medicated tissue damage DC IL-12 production
Pain signals DC migration Adhesion receptors PMN adhesion to NF-KB and COX-2 expression
Angiogenesis and Phosphorylation signals ROS generation & endothelial cells Renal fibrosis
cell proliferation inhibit NF-κB reportor Pro-Inflammatory Nitric oxide and prostacyclin T-cell migration
l/R injury gene activation cytokines (TNFα, IL-8) in endothelial cells TLR-mediated Mφ activation
SPM PMN transmigration
ROS extracellular release Block PMN chemotaxis Microbial kiling and TNF and IFNγ release
Specific Actions Adhesion PGE2 production
Mucosal clearance of clearance Protection of retinal
Pro-inflammatory cytokines PMN by CD55 Neovascularization pigment epithelial cells
(TNFα, IL-12) PMN detachment Microglial cell Neuroprotective actions
DC-lymphocyte interactions cytokine expression
LXA4 production CCR5 expression on T-cells
(immune synapse)
ROS intracellular
Phagocytosis and IL-10
production Microbial killing

Fig. 8.8 The genus of specialized proresolving media- lipoxins, E-series resolvins, D-series resolvins, and protec-
tors: structures and actions. The SPM genus is defined by tins. The main structures of key SPM genus members are
reduction or limiting further PMN infiltration and reduc- depicted; the complete stereochemistry of each has been
tion of lipid mediators and cytokines. SPM also stimulate determined, and their physical properties and bioactions
the nonphlogistic recruitment of mononuclear cells and have been confirmed by total organic synthesis (Source:
the stimulation of macrophages to phagocytose apoptotic Serhan [328]. Copyright © 2010 American Society for
PMN microbes and microbial particles. The family pre- Investigative Pathology Terms and Conditions)
cursors are substrates for their respective conversion to

tion and their relationship to the AXL receptor the recognition of apoptotic neutrophils, the sup-
remains to be defined. pression of proinflammatory cytokines as well as
The clearance of leukocytes and macrophages the release of immunosuppressive cytokines like
from inflamed sites may occur by several routes. IL-10 and TGF-b1 by macrophages, and apop-
Leukocytes can emigrate from an inflamed site totic cells contribute to the anti-inflammatory
back to the systemic circulation [329]. Cells effects of these processes. As noted previously,
from inflammatory sites may also migrate to glucocorticoids and lipoxins can increase the
lymph nodes via the lymphatic drainage system. capacity of macrophages to ingest apoptotic cells.
Finally, cells may be cleared by phagocytosis Apoptosis is also of significance to the switch
after cell death by necrosis or apoptosis. The lat- from proinflammatory to anti-inflammatory
ter process is the most efficient means of clearing processes as well as to the enhancement of the
cells and limiting tissue damage since it is a non- expression of death receptors and the induction
phlogistic process. Cells such as neutrophils in of apoptosis. PGE2 via its capacity to increase
an inflammatory site must undergo apoptosis cellular cyclic AMP can suppress the capacity of
and subsequent phagocytosis by macrophages if macrophages to ingest apoptotic neutrophils, yet
clearance in a nonphlogistic manner is to occur. lipoxin and annexin 1 can induce apoptosis and
A variety of mechanisms have been described for enhance the ingestion of apoptotic neutrophils
270 8 Mechanisms of the Acute Attack of Gout and Its Resolution

by macrophages [330]. Although incompletely as the primary provider of arachidonic acid for
resolved, elevated prostaglandin levels such as the synthesis of proinflammatory mediators.
PGE 2 appear to be involved in the switch from Calcium-independent PLA2 is also necessary for
the generation of proinflammatory eicosanoids IgG-mediated phagocytosis and for the process-
to the synthesis of proresolving eicosanoids like ing of IL-1b. In the resolution phase of rat car-
lipoxin. The following mechanism, as noted pre- ageenin-induced pleuritis, sPLA2 types IIa and
viously, has been proposed as a means by which IV are expressed at 36 and 48 h after the onset
prostaglandins and phospholipases regulate this of inflammation, and they synthesize lipoxins
switch. Type VI calcium-independent phospho- and PAF. Lipoxins suppress leukocyte functions
lipase A2 triggers the synthesis of PGE2, PAF, including inflammatory cell infiltration, whereas
LTB4, and IL-1b, the proinflammatory mediators PAF enhances phagocytosis of apoptotic neutro-
that are observed in the early phases of an acute phils by macrophages. Finally, in the late phase
inflammatory response. During the resolution of of carageenin-induced pleuritis, lipoxins and
an inflammatory response, type IIa and type V PAF trigger the expression of type IV cytoplas-
secretory PLA2 and type IV cytoplasmic PLA2 mic PLA2 and cyclooxygenase-2. This leads to
are the predominant isoforms expressed. These the synthesis of PGD2 and its cyclopentenone
PLA2 isoforms are either not detectable or mini- prostaglandins to enhance resolution).
mally detectable at the onset of an inflammatory It is reasonable to assume that a complete pic-
reaction. These studies were performed on rats ture of the changes in phospholipases and the
that had the inflammation-provoking substance, resultant genesis of anti-inflammatory agents
carageenin, injected into their pleural cavity to does not exist at this time. Nonetheless, several
create an inflammatory pleuritis. Pleural exu- components of importance to these changes have
dates were then examined for their distribution of been identified. Endogenous glucocorticoids
phospholipases using the cells recovered from the appear to have a role since the early phases of
exudate as the phospholipase source. Although inflammation are regulated by type VI calcium-
only this pleuritis model has been evaluated, it independent phospholipase A2 which is resistant
is presumed that this experimental system would to suppression by glucocorticoids, and once
hold for other inflammatory reactions. Since cor- endogenous glucocorticoid levels are reduced to
ticosteroids regulate inflammatory responses, the normal, type IIa and type IV secretory phospholi-
level of corticosterone was measured during the pases come into play. The signal for the activa-
onset and resolution of this carageenin-induced tion of these secretory PLA2s remains to be
pleuritis. In carageenin-induced pleuritis, cor- identified, but the cytoplasmic type IV phospho-
ticosterone levels were the highest at the outset lipase is triggered by lipoxins. Two additional
but declined to normal levels and below dur- factors may have a role in this switching process:
ing the resolution phase. Interestingly, experi- PAF acetylhydrolase and nitric oxide/apoptosis.
mental data show that dexamethasone has little
effect on the suppression of IL-1b-stimulated
type VI calcium-independent PLA2, whereas PAF Acetylhydolase
dexamethasone reduces the type IV cytosolic and Anti-inflammation
PLA2 increments triggered by IL-1b. These
data suggest that type VI calcium-independent Human plasma PAF acetylhydrolase (AH) is
PLA2 appears to be resistant to glucocorticoid composed of 441 amino acids and has been
PLA2 suppression and that the expression of mapped in the human to chromosome 6p12-21.1.
PLA2 is regulated by endogenous glucocorti- During the differentiation of monocytes to mac-
coids. It has also been postulated that the inhi- rophages, the latter cells synthesize and release
bition of secretory and cytoplasmic PLA2 leads PAF-AH [331]. Although PAF-AH shows both
to the expression of calcium-independent PLA2 apoptotic and antiapoptotic activity, PAF-AH
Lipoxin Biosynthesis 271

regulates inflammation by diminishing the apoptotic protease-activating factor 1 (APAF-1)


proinflammatory stimuli of PAF and deficiencies and with the addition of deoxyadenosine triphos-
in PAF-AH give rise to several inflammatory phate/adenosine triphosphate (dATP/ATP) forms
states. Although additional insight into the role the apoptosome. The latter heptameric complex
of PAF-AH is needed, the fact that macrophages activates caspase-9 which then activates the
derived from monocytes synthesize and release effector caspases (caspase-3, caspase-6, and cas-
of PAF-AH suggests a role for this enzyme in pase-7). This pathway can also be interrupted by
modifying PAF and its proinflammatory actions. inhibitors and emphasizes the need to shut down
all the proapoptotic molecules.
Two parameters have been described
Apoptosis and Anti-inflammation that have pertinence to the resolution of
inflammation: the regulation of the pro apop-
The other significant pathways regulating the totic and antiapoptotic pathways by NF-KB
resolution of inflammatory responses are closely and the role of epi-lipoxins and nitric oxide in
linked to the apoptotic cell response. Two the anti-inflammatory response. Several factors
major routes can be manipulated to suppress are of significance with respect to NF-KB. The
inflammation: pathways that induce apoptosis inhibition of NF-KB DNA binding in the late
and pathways that suppress the expression of phase of an inflammatory response impedes
antiapoptotic genes. Two pathways are known the resolution of inflammation. COX-2 inhibi-
to trigger apoptosis: the extrinsic pathway or the tion also impedes the resolution. An analysis of
death receptor pathway and the intrinsic path- leukocyte gene expression in the early phase (6
way or the mitochondrial death pathway. The h) and the late phase (48 h) of a carageenin-
extrinsic pathway is triggered by the activation of induced pleuritis demonstrated the presence
transmembrane death receptors secondary to the of proinflammatory cytokines (lymphotoxin
binding of CD95, Fas, and tumor necrosis factor- TNF-a, and the antiapoptotic protein, Bcl2) in
a to them. The binding of these receptors leads the early phase, whereas anti-anti-inflammatory
to the activation of caspase-8 and/or caspase-10. cytokines (TGF-and the proapoptotic Bcl2
These caspases cleave procaspase-3 to generate homolog, Bax) are expressed in the late phase.
caspase-3 leading ultimately to apoptosis. The Recent evidence implicates the IKKa subunit
caspases are proteolytic enzymes activated by of the IKK complex as a negative regulator of
proapoptotic stimuli. C-FLIP (FADD-Iike ICE inflammation [332]. IKKa suppresses NF-kB
(caspase-8) inhibitory protein, ARC (apoptosis activity by increasing the turnover of the RelA
repressor with caspase recruitment domain), and and c-Rel subunits of the NF-kB and their
IAP (inhibitors of apoptosis) may impede the removal from proinflammatory gene promoters.
progress of apoptosis and emphasize the need In addition, p50-p50 homodimers appear to be
to block their activity. The mitochondrial death the principal NF-kB activity in the late phase
pathway is dependent upon the release of cyto- of inflammation. This p50-p50 homodimer is
chrome-c from the mitochondrial intermembrane known to suppress proinflammatory gene tran-
area to the cytoplasm of the cell. The mecha- scription. To confirm these experimental data,
nism by which cytochrome-c is released from inhibition of NF-kB at the time of resolution
the mitochondria is not completely resolved but of inflammation has been shown to prolong
a number of mechanisms have been proposed. the inflammatory response and prevent apopto-
Furthermore, staurosporine, actinomycin D, sis. These data also suggest that studies of the
peroxide, and ultraviolet radiation may also dis- IKKa subunit of NF-kB and its regulation are
rupt the mitochondrial membrane and cause the likely to reveal approaches to the induction of
release of cytochrome-c. Once released from the the resolution of inflammation and the means
mitochondrial membrane, cytochrome-c binds the by which apoptosis can be induced.
272 8 Mechanisms of the Acute Attack of Gout and Its Resolution

Nitric Oxide and Anti-inflammation via the generation of nitric oxide. Nonetheless,
nitric oxide does regulate components like
Finally, a series of investigations have related P-selectin and CD11/CD18 expression that
nitric oxide, NF-kB activity, and aspirin.. Aspirin clearly affect inflammatory responses, and it may
acetylates COX-2 residing in either endothelial account, in some part, for the anti-inflammatory
cells or leukocytes which then leads to the syn- effects of aspirin and aspirin-triggered lipoxins.
thesis of 15-epi-lipoxin A4. The latter compound In addition to its role in impairing leukocyte
then induces nitric oxide synthesis via constitu- transmigration, nitric oxide can also modulate
tive endothelial nitric oxide synthase and induc- apoptosis. The general concept proposed at this
ible nitric oxide synthase. The nitric oxide time is that low levels of nitric oxide produced by
production triggered by aspirin then impairs leu- the neuronal and endothelial cell isoforms of
kocyte-endothelium interactions causing an anti- nitric oxide synthases are cytoprotective, whereas
inflammatory effect. Investigations have also high levels of nitric oxide such as those produced
shown that nitric oxide has a biphasic effect on by inducible nitric oxide synthase cause apopto-
NF-kB activity. At low levels of nitric oxide gen- sis. The latter concept has some experimental
erated by endothelial nitric oxide synthase, support since endogenous- and exogenous-
NF-kB activity is activated, whereas at higher derived nitric oxide from inducible nitric oxide
levels triggered by inducible nitric oxide syn- synthase induce apoptosis in murine macrophage
thase, NF-kB activity is inhibited. It has also been cell lines. Human macrophages also undergo
shown that under some circumstances, endothe- apoptosis when exposed to exogenous donors of
lial nitric oxide synthase causes the expression of nitric oxide. There are, however, some conflicting
inducible nitric oxide synthase via soluble gua- reports as to apoptosis induced by nitric oxide
nylate cyclase. The existence of this nitric oxide including the role of peroxynitrite formed from
guanylate cyclase pathway has been confirmed in the interaction between nitric oxide and the
human mesangial cells. These same investiga- superoxide anion [334]. Nonetheless, experi-
tions showed that IL-1- or TNF-a-triggered nitric ments indicate that activated macrophages can
oxide production inhibited by NG-nitro-L- induce apoptosis by the release of nitric oxide.
arginine methyl ester (L-NAME) causes a In general, it is now proposed that low concen-
significant reduction in inducible nitric oxide trations of nitric oxide activating soluble guany-
synthase expression. Such experiments demon- late cyclase with the formation of cyclic GMP
strate the dependence of iNOS on eNOS activity, protects the cell from apoptosis. In contrast to
and such findings are reversed by providing a this, rabbit macrophages exposed to high concen-
nitric oxide donor. trations of nitric oxide become apoptotic by
Evidence for the effects of lipoxins and aspi- a guanylate cyclase-independent pathway that is
rin-triggered epi-lipoxins on leukocyte properties not altered by cyclic GMP-dependent kinases or
and leukocyte-endothelial interactions have also cyclic GMP analogs. Superoxide dismutase
been described. These anti-inflammatory lipox- antagonizes peroxynitrite-induced apoptosis in
ins impair leukocyte-endothelial cell interactions, these experiments, and therefore, the balance
diminish leukocyte diapedesis, and block leuko- between superoxide and nitric oxide may be
cyte chemotaxis. A variety of publications have a significant regulator of apoptosis. Thus, nitric
documented the role of lipoxins and their stable oxide generation and inducible nitric oxide syn-
analogs as well as 15-epi-lipoxin A4 on cell adhe- thase play a key role in the regulating apoptosis.
sion molecules [333]. It has also been shown that Finally, a brief discussion of the soluble epox-
inhibitors of lipoxin receptors or 15-epi-lipoxin ide hydrolases is warranted since this enzyme
A4 synthesis significantly impair the inhibition family converts biologically active compounds
of the effects of aspirin on neutrophil-endothelial like epoxyeicosatrienoic acids (EETs), hydroxye-
cell interactions. It remains to be determined icosatrienoic acids (HETEs), and epoxyoctade-
whether lipoxins alter cell adhesion directly or cenoic acids (EpOMEs) to their respective diols.
Lipoxin Biosynthesis 273

These EETs possess anti-inflammatory activity cellular and tissue targets for the development of
and inhibiting their further metabolism via epox- agents to control this phase of inflammation.
ide hydrolases may serve to preserve these anti- This discussion also describes in some detail
inflammatory effects. There is now experimental the pathophysiological responses that have been
evidence that soluble epoxide hydrolases inhibited or might be observed in acute and resolving gout.
by either 12-(3-adamantane-1-yl-ureido)-dode- Although not all the parameters discussed have
canoic acid butyl ester (AUDA-BE) or 1-ada- been definitively determined in gout, it is clear
mantane-3-(5-(2-(2-ethylethoxy)ethoxy)pentyl) that MSU crystal deposition, neutrophil chemot-
urea (compound 950) diminishes levels of axis, neutrophil-MSU phagocytosis, and the gen-
proinflammatory cytokines and nitric oxide eration of the inflammatory mediators necessary
metabolites but enhances the formation of lipox- to accomplish the ingestion of MSU crystals are
ins. Such inhibitors may also alter the expression essential for the clearance of these inflammatory-
of NF-kB and IkB kinase via the persistence of provoking foreign crystals. Much of the attention
epoxyeicosatrienoic acids. These studies have concerning the management of inflammatory
been extended by evaluating the changes in conditions has focused on agents that block the
tobacco smoke-induced inflammation in rats in signaling controlled by NF-kB expression. This
the presence and absence of the soluble epoxide approach is based on a large group of studies show-
hydrolase inhibitor, 12-(3-adamantane-1-yl- ing that many anti-inflammatory agents inhibit
ureido)-dodecanoic acid n-butyl ester. When NF-kB. Such inhibitors of NF-kB include aspi-
either the inhibitor or the inhibitor plus EETs was rin, sodium salicylate, NSAIDS, glucocorticoids,
injected subcutaneously to mice prior to smoke antioxidants, proteasome inhibitors, antisense
exposure, there was a significantly reduced num- oligodeoxynucleotides, green tea polyphenols,
ber of bronchoalveolar lavage cells including curcumin, resveratrol, and some cell-penetrating
a reduction in neutrophils and macrophages when peptides. Aspirin and sodium salicylate bind to
compared to controls receiving no inhibitor/EETs and block the ATP binding site of IKKb, the
[335]. These investigations also showed that the phosphorylator of IkB and its subsequent degra-
12-(3-adamantane-1-yl-ureido)-dodecanoic acid dation. Thus, NF-kB is no longer constrained by
n-butyl ester was rapidly converted to 12-(3-ada- its inhibitor, IkB. Some NSAIDS inhibit NF-kB
mantane-1-yl-ureido)-dodecanoic acid in vivo by activation, whereas others do not. Even acet-
an esterase, and the latter compound appeared to aminophen blocks the binding of NF-kB to DNA
be the active ingredient. Furthermore, the combi- in certain cells under specific conditions.
nation of this inhibitor and epoxyeicosatrienoic A variety of mechanisms have been published
acids reduced the effect on bronchoalveolar with respect to the way in which glucocorticoids
lavage cell accumulation more than the inhibitor impair NF-KB activation and the expression of
alone. Such data provide further evidence of the the genes it regulates. These include the increased
anti-inflammatory actions of EETs. synthesis of IkBa (the inhibitor of NF-kB),
The foregoing summary provides a bird’s eye a direct interaction between NF-kB and the glu-
view of the complexity of the resolution of cocorticoid receptor, the inhibition of RNA poly-
inflammatory responses. In addition to this, the merase II phosphorylation, the sequestration of
role of glucocorticoids in the suppression of RelA (a component of NF-KB), and the recruit-
inflammation is also. discussed subsequently in ment of histone deacetylase 2 [336]. The effects
another section. As will be seen, glucocorticoid- of glucocorticoids are discussed in greater detail
induced anti-inflammatory effects mediated by in a separate section subsequently. Selected anti-
annexin 1 continue to provide significant insights oxidants also impair NF-kB activation. The inhi-
into the suppression of inflammation. It is bition of prosteasome-dependent degradation of
expected that investigations of the resolution of the natural inhibitor of NF-kB, IkB, results in
inflammation and its regulation will continue to the inability of NF-kB to free itself from its
provide useful information and will also generate inhibitor and translocate to the nucleus. Several
274 8 Mechanisms of the Acute Attack of Gout and Its Resolution

proteasome inhibitors have been characterized activation. Sulfasalazine has been shown to be
as effective NF-kB inhibitors via this mecha- a specific inhibitor of NF-kB activation but its
nism. Both antisense oligodeoxynucleotides and two metabolites, sulfapyridine and 5-aminosali-
decoy oligodeoxynucleotides have also been cylic acid (5-ASA), do not have any effect on
found to impair NF-kB-mediated gene expres- NF-kB activation. Sulfasalazine also blocks
sion [337]. In addition, natural dietary com- TNF-, endotoxin-, and phorbol ester-induced
pounds including green tea polyphenols, NF-kB activation. Some evidence also exists to
resveratrol, curcumin, and capsaicin also impair show that gold compounds may also impede
NF-kB expression, usually by blocking IKK NF-kB activation. Since reactive oxygen species
activity [338]. Despite the clear evidence that (ROS) can activate NF-kB, antioxidants such as
NF-kB inhibition provides a means of suppress- dithiocarbamates and N-acetylcysteine have been
ing inflammatory responses, the lack of target evaluated as suppressors of NF-kB activation.
specificity raises some problems with the use of Both dithiocarbamate and N-acetylcysteine sup-
such agents. For example, the inhibition of press NF-kB activation. These data support the
cyclooxygenase activity is clearly beneficial in concept that NF-KB activation is a key to the
the treatment of inflammation, but the nonse- generation and expression of proinflammatory
lective activity of these inhibitors may be haz- genes, and the inhibition of this process repre-
ardous to the gastrointestinal tract, kidney, or sents a significant means of suppressing
cardiovascular system. However, the joint cav- inflammatory reactions.
ity represents an excellent site for the activity
of such agents to prevent inflammation since
compounds can be designed to act only at the Glucocorticoid
local site and have a delayed rate of absorption.
The use of natural substances also avoids the Overview
issue of the immunogenicity of therapeutic
agents. Although receptor desensitization and lipoxin
generation are natural mechanisms for the
induction of anti-inflammatory responses with
Drugs the subsequent resolution of acute arthritic
attacks in gout, the mechanisms by which
The dependence of proinflammatory gene expres- anti-inflammatory drugs affect inflammatory
sion on transcription factors like NF-kB has led responses may also provide additional ratio-
to an evaluation of the role of anti-inflammatory nales for suppressing inflammation. One
drugs as modulators of NF-kB. Aspirin and of the most efficient means of suppressing
sodium salicylate at relatively high doses (mM) inflammatory responses is through the use of
impede the degradation of IkB, and by this mech- glucocorticoids. These drugs upregulate lipoco-
anism, they inhibit the activation of IKK. These rtin-1 and p11/calpactin binding protein which
agents bind to IKKb but not to IKKa to inhibit its act to suppress the release of arachidonic acid,
kinase activity. The use of aspirin inhibits IL-1- a precursor of many proinflammatory mediators.
and TNF-induced NF-kB activation as well as Glucocorticoids also induce secretory leukocyte
the expression of IL-6 and IL-8 in synovial protease inhibitor (SLP1), b-adrenergic recep-
fibroblasts. tors, and interleukin-1 type II receptor (IL-IRA).
A variety of nonsteroidal anti-inflammatory Glucocorticoid-dependent induction processes
drugs (NSAIDs) all inhibit IkBa kinase causing take 24–48 h to develop, and for this reason, they
a decrease in its degradation and a decrease in are likely to regulate the late stages of an acute
NF-kB-regulated genes. inflammatory reaction. Of greater pertinence
A few additional anti-inflammatory and anti- to the regulation of immediate inflammatory
arthritic medications are also inhibitors of NF-kB responses are the glucocorticoid-dependent
Glucocorticoid 275

mechanisms that negatively control gene tran- receptor (GR) are generated by alternative splic-
scription. Such mechanisms include DNA bind- ing of the human GR gene in its ninth exon at
ing-dependent transrepression, transrepression the site of the GR pre-mRNA [340]. The GRa
without DNA binding, and gene regulation via and GRb receptors diverge in their structure at
posttranscriptional and translational mecha- their carboxyl terminus. The last 50 amino acids
nisms [339]. Although glucocorticoid-mediated in the GRa are replaced by a nonhomologous,
induction of proteins may contribute to the late 15 amino acid sequence in GRb. The GRa
anti-inflammatory effects of glucocorticoids, when bound to a glucocorticoid molecule results
glucocorticoid-mediated actions may also occur in the translocation of the GRa from the cyto-
by nongenomic actions such as the activation plasm to the nucleus. In the nucleus, this hor-
of mitogen-activated protein kinases, adenylate mone-receptor complex binds to specific DNA
cyclase, protein kinase C, and heterotrimeric response elements in the promoter region of the
guanosine-binding proteins (G proteins). Many glucocorticoid-responsive genes to enhance the
glucocorticoid-mediated anti-inflammatory transcription of anti-inflammatory genes. The
changes may also act by decreasing gene expres- direct binding of activated glucocorticoid recep-
sion. In fact, the major anti-inflammatory action tor alpha to the glucocorticoid response elements
of glucocorticoids occurs via the inhibition of (GREs) present on inducible anti-inflammatory
proinflammatory transcription factors such as genes is called transactivation. The other mech-
activator protein (AP-1), signal transducers and anism by which the GRa receptor regulates
activators of transcription (STATs), nuclear fac- proinflammatory gene expression is by a process
tor of activated T cells (NF-AT), and nuclear termed transrepression. By this mechanism, acti-
factor (NF)-kappa B. In general, multiple mecha- vated GRa has a direct interaction with different
nisms for positive and negative regulation of proinflammatory transcription factors like AP-1
glucocorticoid-mediated gene expression exist, and NF-kB to transrepress transcription factor-
and as will discussed in detail subsequently, the inducible inflammatory gene activation. The
specific sites to which the glucocorticoid-gluco- GRb receptor behaves quite differently from the
corticoid receptor complex binds can either acti- GRa since GRb, in contrast to the GRa, reacts
vate (transactivation) or repress (transrepression) weakly with heat shock proteins, is transcription-
gene transcription. ally inactive and does not bind glucocorticoids.
In addition, GRb has a longer half-life than
GRa and has been related to glucocorticoid-
Mode of Action resistant states in several inflammatory cell types
and diseases, and its expression is enhanced by
Glucocorticoids cause their effect on cells in the proinflammatory cytokines such as TNF-a, IL-1,
following manner. Lipophilic glucocorticoids a combination of IL-2 and IL-4, and IL-13. The
diffuse across the cell membrane and react with prevailing hypothesis is that GRa and GRb form
intracellular cytoplasmic glucocorticoid recep- a heterodimer complex, and this heterodimer
tors (GRs). In the resting state, the glucocorti- inhibits GRa nucleus shuttling in response to
coid receptors are associated with proteins like steroids. Although the role of the GRb receptor
heat shock proteins that enhance their stability still remains controversial, a significant data base
and block their nuclear localization sequences. exists in support of the concept that GRb plays an
However, when GR binds to its ligand, it dis- important role in the inhibition of GRa expres-
sociates from hsp (heat shock protein) 90 with sion [341, 342]. Therefore, evidence supports
the resultant exposure of nuclear localiza- the fact that the human glucocorticoid receptor
tion sequences. Glucocorticoid receptors are beta is a natural dominant negative inhibitor of
encoded by a single gene, but two variants have human glucocorticoid receptor alpha-induced
been described: an alpha and beta form. These transactivation of glucocorticoid-responsive
two homologous isoforms of the glucocorticoid genes. The N-terminal domain of this receptor
276 8 Mechanisms of the Acute Attack of Gout and Its Resolution

contains serines and threonines where kinases The primary purposes of the following discus-
may phosphorylate the receptor [343]. Such sion are twofold: to characterize the mechanisms
phosphorylation may alter nuclear export and for the activation and repression of inflammatory
import, glucocorticoid binding affinity, recep- genes and to delineate the mechanisms by which
tor stability, or transactivating capacity. The glucocorticoids cause the suppression of acute
glucocorticoid receptor-ligand complex binds inflammatory responses like gout. Fulfilling these
to specific sequences in the nuclear DNA pro- two objectives not only identifies targets for the
moter region called glucocorticoid response development of anti-inflammatory drugs but also
elements (GREs). Glucocorticoid-ligand com- characterizes putative pharmacological interven-
plexes bind as homodimers to nuclear DNA. tion sites with the possibility of developing addi-
These GREs located in the promoter region tional drugs with enhanced potency and fewer
are known to regulate steroid-sensitive genes, side effects than the drugs available today.
and activated glucocorticoid receptors regulate the Glucocorticoids can regulate genes through direct
transcription of genes either directly or indirectly. transcriptional control, indirect transcriptional
Several points should be recognized concern- regulation by interfering with other transcription
ing the functions of glucocorticoid-mediated factors, and by posttranscriptional effects. Like
gene regulation. First, glucocorticoid-mediated transactivation, negative regulation of transcrip-
positive regulation of gene transcription or trans- tion (transrepression) can occur via many differ-
activation controls the side effects of steroid ent mechanisms, and models for glucocorticoid
treatment, whereas the regulation of anti- receptor transcriptional modulation have been
inflammatory effects of steroids occurs via pro- generated and supported by investigative findings.
cesses of negative regulation of gene transcription In the first model, homodimers of the glucocorti-
or transrepression. Second, as noted previously, coid receptor repress transcription by interacting
two forms of the human glucocorticoid receptor negatively with glucocorticoid response ele-
have been described. The human GRa isoform ments. Such negative GRE sites are not common
contains 777 amino acids, whereas a somewhat and regulate only a few genes. In the second
smaller, dominant negative, and non-hormone model, a composite site exists for GR and factors
binding human GRb has also been characterized. required for gene transcription. Such competitive
Third, both GR transactivation and GR transre- binding prevents transcriptional factors from
pression appear to occur via multiple different binding to GRE. This setting occurs when the
mechanisms. For example, genes that are posi- cyclic AMP response element binding protein
tively regulated by GREs may activate transcrip- (CREB) cannot bind to GRE because it has over-
tion when homodimers of GR bind cooperatively lapping sites with GR binding sites. In the third
to GRE sites. Another mechanism of positive model, GR interacts with a second transcription
transactivation occurs when GR and another tran- factor and prevents gene transcription by a
scription factor both bind to DNA and activate method that does not require GR binding to DNA.
transcription from composite binding sites. An In the last model, both GR and· a second tran-
additional mechanism involves the interaction of scription factor repress transcription through
GR with a second transcription factor, and composite DNA binding sites. This model
together they activate transcription without the requires binding of both GR and the second tran-
requirement that DNA bind to GRE. Fourth, GR scription factor to DNA. In summary, glucocorti-
transactivation regulates genes involved in gluco- coids regulate inflammation by transrepression of
neogenesis including tyrosine aminotransferase, transcription factors such as activator protein-1,
alanine aminotransferase, and phosphoenolpyru- nuclear factor-kappa S, and nuclear factor-AT.
vate carboxykinase. It is presently believed that Thus, the primary but not the only means by
the genes responsible for the side effects of glu- which glucocorticoids repress inflammation is by
cocorticoid therapy are also regulated by GR interfering with transcription factors. Since
transactivation. inflammatory response genes are primarily regu-
Glucocorticoid 277

lated by NF-kB and AP-1 then it is the interfer- TNF-a, chemokines, and leukocyte adhesion
ence with these transcription factors by molecules by blocking the effects of activated
glucocorticoids that causes the repression of glucocorticoid receptors on transcription factors
the inflammatory response. As the mechanisms like NF-kB and AP-1. In addition, glucocorti-
by which inflammatory genes are expressed and coids inhibit histone acetyltransferase activity and
repressed are discussed in the following recruit histone deacetylase [346]. This inhibitory
paragraphs, the relationships between glucocorti- and recruitment activity prevents the unwind-
coids and these mechanisms will be characterized ing of DNA and access to transcription factors.
if they apply. Another way in which glucocorticoids cause anti-
At this time, the mechanisms by which glu- inflammatory effects is via its interference with
cocorticoids suppress inflammation are com- the phosphorylation of the C-terminal domain
plex and continue to be evaluated, yet these of RNA polymerase II, and thereby, reducing its
agents remain as the most common drugs used interaction with NF-kappa B-regulated promoter
to modify inflammatory responses. Even though regions and subsequent gene expression [347].
the anti-inflammatory effects of glucocorticoids Finally, there is evidence in some cell types, espe-
are complex and data regarding their mecha- cially lymphocytes and monocytes, that dexam-
nisms of action are incomplete, multiple ways ethasone triggers the synthesis of the NF-kappa
for such anti-inflammatory effects to occur have B inhibitor, I-kappa S-a [348].
been described. The glucocorticoid effects on
inflammatory disorders are mediated by the inter-
actions between the ligand glucocorticoid and the Histone Acetylation and Glucocorticoids
glucocorticoid receptor a and that complex then
serves as a transcription factor. There are three The major mechanism by which inflammatory
ways in which such glucocorticoid-glucocorti- genes are repressed is by altering histone acetyla-
coid receptor complexes mediate their effects: tion. The cell chromatin in the nucleus of the cell
activation or inhibition of transcription by RNA contains a complex of DNA, histones, and non-
polymerase II, interactions between the recep- histone proteins. Genes are repressed when DNA
tor complex and other transcription factors such is bound tightly to core histone proteins (H2A,
as NF-kB or AP-1, and nongenomic pathways H28, H3, and H4), and the RNA polymerase II
whose activity is generated via membrane-associ- enzyme cannot bind to DNA and generate
ated receptors and their second messengers [344]. mRNAs. Histone core proteins contain lysine
Another regulatory component of the glucocorti- molecules in their long N-terminal tail that can be
coid receptor is its posttranslational modifications acetylated by the enzyme histone acetyltrans-
based on serine phosphorylations mediated by ferase. Acetylation of histones causes the tightly
cyclin-dependent kinases and mitogen-activated bound DNA to unwind and allows the RNA poly-
protein kinases [345]. In fact, ubiquitin-related merase II along with transcription factors to bind
modifiers interacting with the glucocorticoid to DNA Such acetylation reduces the charge on
receptor may increase the transcriptional activ- histones and results in an activated form of chro-
ity of the receptor. Glucocorticoids can result matin ready for transcription factor binding. The
in the expression of anti-inflammatory proteins proinflammatory transcription factors like
such as lipocortin, IL-1 receptor antagonist, and NF-kappaB bind to specific DNA sequences and
the anti-inflammatory mediator, IL-10, by induc- then interact with coactivator molecules. The
ing the transcription of genes near the glucocor- principal coactivator molecules are cyclic ade-
ticoid response elements, by the recruitment of nosine monophosphate response element-binding
co-activators such as CREB-binding protein/ protein (p300/CREB) and p300/CBP-associated
p300, or by the action of RNA polymerase. factor (PCAF). These coactivator molecules are
Glucocorticoids also inhibit the expression of switches that control gene transcription, and they
many proinflammatory cytokines such as IL-1, have intrinsic histone acetyltransferase activity to
278 8 Mechanisms of the Acute Attack of Gout and Its Resolution

transform the chromatin structure from a closed glucocorticoid-inducible 37 kDa protein origi-
conformation to an open, activated form [349]. nally designated as lipocortin but now called
Thus, histone acetylation and coactivator associa- annexin 1 (ANXA1). This inducible protein
tions result in the unwinding of DNA, the binding inhibits phospholipase A2 activity and as a result,
of TATA box-binding protein (TBP), and the suppresses prostaglandin biosynthesis in per-
binding of coactivators to the TBP. TBP brings fused lungs and activated macrophages. A
together the TATA box that identifies the start site significant impetus to the increase in its role in
of gene transcription and the coactivator factors inflammation resulted from the cloning of the
and their related proteins as well as RNA poly- glucocorticoid-inducible protein, annexin, and
merase II. The acetylation of histones, therefore, its capacity to inhibit acute inflammatory
initiates the expression of inflammatory genes. responses.
Histone deacetylases (HDACs) cause the deacety- In general, annexins are a family of intracel-
lation of acetylated histones and result in the lular proteins with a unique structure of their cal-
repression of genes. It is also known that differ- cium binding sites permitting them to interact
ent HDACs, of which there are 12, interact with with negatively charged phospholipids [350].
different histone acetylation patterns. It is also Although annexins are mainly components of the
well known that the inhibition of HDAC activity intracellular milieu including elevated concentra-
causes an increase in histone acetyltransferase tions in the cytosol of neutrophils, the adherence
activity and increased inflammatory gene expres- of neutrophils to the endothelium results in the
sion. In addition to these HDAC activities that export of annexin 1 to the cell surface. How
suppress gene expression, other means of gene annexin 1 is transported to its extracellular sites
repression exist including the modification of remains incompletely resolved, but several dif-
core histones by methylation, phosphorylation, ferent concepts have been proposed [351].
ubiquitination, inhibition of mRNA stabilizers, Annexin 1 is stored in the gelatinase granules of
and inhibition of activators of transcription fac- the human neutrophil and is transported to the
tors such the c-Jun N-terminal kinase and p38 cell surface when neutrophils adhere to endothe-
MAP kinase. Glucocorticoid receptors may either lium. Glucocorticoids induce annexin 1 expres-
bind directly to activators and inhibit histone sion and/or the secretion of annexin.
acetyltransferase activity or may recruit histone
deacetylases to reverse histone acetylation and
suppress the expression of proinflammatory
genes. As noted, there are also other mechanisms References
that play a role in the expression and repression
1. Phelps P. Appearance of nondialyzable chemotactic
of proinflammatory genes including the induc-
activity following intraarticular injection of monoso-
tion and expression of anti-inflammatory genes dium urate crystals. Arthritis Rheum. 1969;12:324.
that lead to the release of anti-inflammatory 2. Hermanns W, Schulz LC. Enzyme histochemical stud-
molecules. ies of the homogeneity of the mononuclear phagocyte
system with special reference to the synovium. Agents
Actions. 1982;11:117.
3. Barland P, Novikoff AB, Hamerman D. Electron
Annexins and Glucocorticoids microscopy of the human synovial membrane. J Cell
Biol. 1962;14:207.
4. Ghadially FN. Fine structure of joints. In: Sokoloff L,
At this juncture, the significant role of a gluco-
editor. The joints and synovial fluid. New York:
corticoid-inducible protein is discussed since it Academic; 1978. p. 110–20.
plays a major role in the anti-inflammatory effects 5. Graabaek PM. Ultrastructural evidence for two distinct
of glucocorticoids. The first anti-inflammatory types of synoviocytes in rat synovial membrane. J
Ultrastruct Res. 1982;78:321.
agents to be identified in humans and animals
6. Okada Y, Nakanishi I, Kajikawa K. Ultrastructure of
were natural and synthetic glucocorticoids. This the mouse synovial membrane. Arthritis Rheum.
early work was extended by the discovery of a 1981;24:835.
Glucocorticoid 279

7. Ghadially FN, Roy S. Ultrastructure of synovial joints 23. Hou WS, Li Z, Gordon RE, et al. Cathepsin k is a
in health and disease. London: Butterworths; 1969. critical protease in synovial fibroblast-mediated col-
8. Edwards JC. The origin of type A synovial lining lagen degradation. Am J Pathol. 2001;159:2167.
cells. Immunobiology. 1982;161:227. 24. Wilkinson LS, Edwards JC, Poston RN, Haskard DO.
9. Edwards JC, Sedgewick AD, Willoughby DA. Expression of vascular cell adhesion molecule-1 in
Membrane properties and esterase activity of synovial normal and inflamed synovium. Lab Invest. 1993;
lining cells: further evidence for a mononuclear 68:82.
phagocyte subpopulation. Ann Rheum Dis. 1982; 25. Mentzer SJ, Rothlein R, Springer TA, Faller DV.
41:282. Intercellular adhesion molecule-1 (ICAM-1) is
10. Goulding NJ, Dixey J, Morand EF, et al. Differential involved in the cytolytic T lymphocyte interaction
distribution of annexins-1, -11, -IV, and -VI in syn- with human synovial cells. J Cell Physiol. 1988;
ovium. Ann Rheum Dis. 1995;54:841. 137:173.
11. Edwards JCW. Fibroblast biology: development and 26. Jonathan CW, Edwards JC. Fibroblast biology.
differentiation of synovial fibroblasts in arthritis. Development and differentiation of synovial
Arthritis Res. 2002;2:344. fibroblasts in arthritis. Arthritis Res. 2000;2:344.
12. Pap T, Muller-Ladner U, Gay RE, Gay S. Fibroblast 27. Revell PA, AI-Saffar N, Fish S, Osei D. Extracellular
biology: role of synovial fibroblasts in the pathogen- matrix of the synovial intimal cell layer. Ann Rheum
esis of rheumatoid arthritis. Arthritis Res. 2000; Dis. 1995;54:404.
2:361. 28. Waggett AD, Kielty CM, Shuttleworth CA.
13. Wilkinson LS, Pitsillides AA, Worrall JG, Edwards Microfibrillar elements in the synovial joint: presence
JC. Light microscopic characterization of the of type VI collagen and fibrillin-containing
fibroblast-like synovial intimal cell (synoviocyte). microfibrils. Ann Rheum Dis. 1993;52:449.
Arthritis Rheum. 1992;35:1179. 29. Worrall JG, Bayliss MT, Edwards JCW. Zonal dis-
14. Pitsillides AA, Wilkinson LS, Mehdszadeh S. Uridine tribution of sulphated proteoglycans in normal and
diphosphoglucose dehydrogenase activity in normal rheumatoid synovium. Ann Rheum Dis. 1994;
and rheumatoid synovium: the description of a spe- 53:35.
cialized synovial lining cell. Int J Exp Pathol. 30. Edwards JCW. Fibroblast-like synovial lining cells.
1993;74:27. In: Henderson B, Edwards JCW, Pettifer ER, editors.
15. Davis LS. A question of transformation. The synovial Mechanisms and models in rheumatoid arthritis. New
fibroblast in rheumatoid arthritis. Am J Pathol. York: Academic; 1995. p. 153–62.
2003;162:1399. 31. Medof ME, Walter EL, Rutgers JL, et al. Identification
16. Konttinen YT, Ainola M, Valleala H, et al. Analysis of of the complement decay accelerating factor on epi-
16 different matrix metalloproteinases (MMP-1 to thelium and glandular cells and in body fluids. J Exp
MMP-20) in the synovial membrane: different profiles Med. 1987;165:848.
in trauma and rheumatoid arthritis. Ann Rheum Dis. 32. Rice GE, Munro JM, Corless C, Bevilacqua MP.
1999;58:691. Vascular and non-vascular expression of INCAM-
17. Konttinen YT, Li TF, Hukkanen M, et al. Fibroblast 110; a target for mononuclear leucocyte adhesion in
biology: signals targeting the synovial fibroblast in normal and inflamed human tissues. Am J Pathol.
arthritis. Arthritis Res. 2000;2:348. 1991;138:385.
18. Konttinen YT, Li TF, Xu JW, et al. Expression of 33. Edwards JCW, Leigh RD, Cambridge G. Expression
laminins and their integrin receptors in different of molecules involved in B lymphocyte survival and
conditions of synovial membrane and synovial differentiation by synovial fibroblasts. Clin Exp
membrane-like interface tissue. Ann Rheum Dis. Immunol. 1997;108:407.
1999;58:683. 34. Jay GO. Characterization of a bovine synovial fluid
19. Pirila L, Aho H, Roivainene A, et al. Identification of lubricating factor. I. Chemical, surface activity and
alpha6beta1 integrin positive cells in synovial lining lubricating properties. Connect Tissue Res. 1992;
layer as type B synoviocytes. J Rheumatol. 2001; 28:71.
28:478. 35. Dobbie JW. Surfactant protein A and lamellar bodies:
20. Tolboom TCA, Pieterman E, van der Laan WH, et al. a homologous secretory function of peritoneum, syn-
Invasive properties of fibroblast-like synoviocytes: ovium, and lung. Perit Dial Int. 1996;16:574.
correlation with growth characteristics and expression 36. EI-Gabalawy H, King R, Bernstein C, et al. Expression
of MMP-1, MMP-3, and MMP-10. Ann Rheum Dis. of N-acetyl-0-galactosamine associated epitope in
2002;61:975. synovium: a potential marker of glycoprotein produc-
21. Muller-Ladner U, Gay S. MMPs and rheumatoid syn- tion. J Rheumatol. 1997;24:1355.
ovial fibroblasts: siamese twins in joint destruction? 37. Proudman SM, Cleland LG, Fusco M, Mayrhofer G.
Ann Rheum Dis. 2002;61:957. Accessible xenografts of human synovium in the sub-
22. Hou WS, Li W, Keyszer G, et al. Comparison of cutaneous tissues of the ears of SCID mice. Immunol
cathepsin K and S expression within the rheumatoid Cell Biol. 1989;59:1203.
and osteoarthritic synovium. Arthritis Rheum. 2002; 38. Hamerman D, Barland P. Structure and function of the
46:663. synovial membrane. Bull Rheum Dis. 1966;16:396.
280 8 Mechanisms of the Acute Attack of Gout and Its Resolution

39. Wilkinson LS, Edwards JC. Microvascular distribu- 57. Hemler ME, Crouse C, Sonnenberg A. Association of
tion in normal human synovium. J Anat. 1989; the VLA alpha 6 subunit with a novel protein. A pos-
167:129. sible alternative to the common VLA beta 1 subunit of
40. Schumacher Jr HR. Ultrastructure of the synovial certain cell lines. J Biol Chem. 1989;264:6529.
membrane. Ann Clin Lab Sci. 1975;5:489. 58. Cheresh DA, Smith JW, Cooper HM, Quaranta V. A
41. Levick JR. Microvascular architecture and exchange novel vitronectin receptor integrin (avJ3x) is respon-
in synovial joints. Microcirculation. 1975;2:217. sible for distinct properties of carcinoma cells. Cell.
42. Matsubara T, Ziff M. Basement membrane thickening 1989;57:59.
of postcapillary venules and capillaries in rheumatoid 59. Vogel BE, Ryan G, Giancotti FG, Ruoslahti E. A
synovium: immunoelectron microscopic and electron novel fibrinectin receptor with an unexpected subunit
microscopic morphometric analysis. Arthritis Rheum. composition (alphaVB1). J Biol Chem. 1990;265:
1987;30:18. 5934.
43. Agudelo C, Schumacher HR. The synovitis of acute 60. Ramaswamy H, Hemler ME. Cloning, primary struc-
gouty arthritis: a light and electron microscopic study. ture and properties of a novel human integrin beta
Hum Pathol. 1973;4:265. subunit. EMBO J. 1990;9:1561.
44. Neal C, Read N, Goodwin D, Edwards JC. Fenestration 61. Carter WG, Ryan MC, Gahr PJ. Epiligrin, a new cell
of tenosynovial capillaries. Br J Rheumatol. 1989; adhesion ligand for integrin alpha3beta1 in epithelial
28:31. basement membranes. Cell. 1991;65:599.
45. Wilkinson LS, Edwards JC. Demonstration of lym- 62. Isberg RR, Leong JM. Multiple B1 chain integrins
phatics in human synovial tissue. Rheumatol Int. are receptors for invasin, a protein that promotes bac-
1991;11:151. terial penetration into mammalian cells. Cell. 1990;
46. Osborn L, Hession C, Tizard R, et al. Direct expres- 65:861.
sion cloning of vascular cell adhesion molecule 1, a 63. Demaziere A, Athanasou NA. Adhesion receptors of
cytokine-induced endothelial protein that binds to intimal and subintimal cells of the normal synovial
lymphocytes. Cell. 1989;59:1203. membrane. J Pathol. 1992;168:209.
47. Rice GE, Munro JM, Bevilacqua MP. Inducible cell 64. Fairburn K, Kunaver M, Wilkinson LS, et al.
adhesion molecule 110 (INCAM-110) is an endothe- Intercellular adhesion molecules in normal synovium.
lial receptor for lymphocytes. A CD 11/CD 18-inde- Br J Rheumatol. 1993;32:302.
pendent adhesion mechanism. J Exp Med. 1990; 65. Szekanecz Z, Haines GK, Lin TR, et al. Differential
171:1369. distribution of intercellular adhesion molecules
48. Pober JS, Lapierre LA, Stolpen AH, et al. Activation (ICAM-1, ICAM-2, and ICAM-3) and MS-1 antigen
of cultured human endothelial cells by recombinant in normal and diseased synovia. Their possible patho-
lymphotoxin: comparison with tumor necrosis factor genetic and clinical significance in rheumatoid arthri-
and interleukin 1 species. J Immunol. 1987;138:3319. tis. Arthritis Rheum. 1994;37:221.
49. Osborn L. Leukocyte adhesion to endothelium in 66. Wilkinson LS, Edwards JC, Posston LN, Haskard DO.
inflammation. Cell. 1990;62:3. Expression of vascular cell adhesion molecule-1 in nor-
50. Stoolman LM. Adhesion molecules controlling lym- mal and inflamed synovium. Lab Invest. 1993;68:82.
phocyte migration. Cell. 1989;56:907. 67. Kaneko M, Inoue H, Nakazawa R, et al. Pirfenidone
51. McEver RP. Seclectins: novel receptors that mediate induces intercellular adhesion molecule-1 (ICAM-1)
leukocyte adhesion during inflammation. Thromb down-regulation on cultured synovial fibroblasts. Clin
Haemost. 1991;65:223. Exp Immunol. 1998;113:72.
52. Hynes RO. Lntegrins: a family of cell surface recep- 68. Pernow P. Substance P. Pharmacol Rev. 1983;35:85.
tors. Cell. 1987;48:549. 69. Inman RD, Chin B, Marshall KW. Substance P and
53. Werb Z, Tremble PM, Behrendtsen O, et al. Signal arthritis. Analysis of plasma and synovial fluid levels.
transduction through the fibrinectin receptor induces Arthritis Rheum. 1986;29:S9.
collagenase and stromelysin gene expression. J Cell 70. Webster ME, Maling HM, Zweig MH, et al. Urate
Biol. 1989;109:877. crystal induced inflammation in the rat: evidence for
54. Ginsberg MH, Loftus JC, Ryckwaert J-J, et al. the combined actions of kinins, histamine and compo-
Immunochemical and amino-terminal sequence com- nents of complement. Immunol Commun. 1972;1:185.
parison of two cytoadhesions indicates they contain 71. Freudweiler M. Experimentalle untersuchungen uber
similar or identical beta subunits and distinct alpha dase wesen der gichtknoten. Dtsch Arch Klin Med.
subunits. J Biol Chem. 1987;262:5437. 1899;63:266.
55. Holzmann B, Weissman IL. Peyer’s patch-specific 72. Freudweiler M. Experimentalle untersuchungen uber
lymphocyte homing receptors consist of a VLA-4- die enstchung der gichtknoten. Dtsch Arch Klin Med.
Iike a chain associated with either of two J3 chains, 1909;69:155. English translation: Experimental inves-
one of which is novel. EMBO J. 1989;8:1735. tigations into the origin of gouty tophi. Arthritis
56. Kajiji S, Tamura RN, Quaranta V. A novel integrin Rheum. 1965;1:270.
(aEJ3 4) from human epithelial cells suggests a fourth 73. His WH. Schicksal und wirkungend des sauren harn-
family of integrin adhesion receptors. EMBO J. sauren natrons in bauch und gelenkohle des kan-
1989;8:673. inchens. Dtsch Arch Klin Med. 1900;67:81.
Glucocorticoid 281

74. McCarty Jr OJ, Hollander JL. Identification of urate globulin in individuals from two gout kindred. J Clin
crystals in gouty synovial fluid. Ann Intern Med. Invest. 1968;47:1254.
1961;54:452. 95. Aakesson I, Alvsaker JO. The urate-binding alpha
75. Maclachlan MJ, Rodnan GP. Effects of food, fast and 1–2 globulin. Isolation and characterization of the
alcohol on serum uric acid and acute attacks of gout. protein from human plasma. Eur J Clin Invest.
Am J Med. 1967;42:38. 1971;1:281.
76. Richet G, Mignon F, Ardaillon R. Goutte secondaire der 96. Klinenberg JR, Kippen I. The binding of urate to
nephropathies chroniques. Presse Med. 1965;73:633. plasma proteins determined by equilibrium dialysis.
77. Ropes MW, Bauer W. Synovial fluid changes in joint J Lab Clin Med. 1970;75:503.
disease. Cambridge: Harvard University Press; 1953. 97. Hasselbacher P, Schumacher HR. Immunoglobulin in
p. 150. tophi and on the surface of monosodium urate crys-
78. Seegmiller JE. Serum uric acid. In: Cohen AS, editor. tals. Arthritis Rheum. 1978;21:353.
Laboratory procedures in the rheumatic diseases. 2nd 98. Cherian PV, Schumacher HR. Immunochemical and
ed. Boston: Little Brown & Company; 1974. p. 216. ultrastructural characterization of serum proteins
79. Dorner RW, Weiss TE, Baldassare AR, et al. Plasma associated with monosodium urate crystals in syn-
and synovial fluid as solvents for monosodium urate. ovial fluid cells from patients with gout. Ultrastruct
Ann Rheum Dis. 1981;40:70. Pathol. 1986;10:209.
80. Horvath SM, Hollander JL. Intra-articular tempera- 99. Kam M, Peri-Treves D, Caspi D, Addadi L.
ture as a measure of joint reaction. J Clin Invest. Antibodies against crystals. FASEB J. 1992;6:2608.
1949;28:469. 100. Schroder K, Tschopp J. The inflammasomes. Cell.
81. Hollander JL, Stoner EK, Brown Jr EM, et al. Joint 2010;140:821–32.
temperature measurements in the evaluation of antiar- 101. Masters SL, et al. Horror autoinflammaticus: the
thritic agents. J Clin Invest. 1951;30:701. molecular pathophysiology of autoinflammatory
82. Hollander JL, Horvath SM. The influence of physical disease (*). Annu Rev Immunol. 2009;27:621–68.
therapy procedures on intra-articular temperature of 102. Kastner DL, et al. Autoinflammatory disease
normal and arthritic subjects. Am J Med Sci. 1949; reloaded: a clinical perspective. Cell. 2010;140:
218:543. 784–90.
83. Loeb JN. The influence of temperature on the solubil- 103. Martinon F, et al. Gout-associated uric acid crystals
ity of monosodium urate. Arthritis Rheum. activate the NALP3 inflammasome. Nature. 2006;440:
1972;15:189. 237–41.
84. Simkin PA, Campbell PM, Larson EB. Gout in 104. Chen C-J, et al. MyD88-dependent IL-1 receptor
Heberden’s nodes. Arthritis Rheum. 1983;26:94. signaling is essential for gouty inflammation stimu-
85. Parhami N, Greenstein N, Juozevicius JL. Erosive lated by monosodium urate crystals. J Clin Invest.
osteoarthritis and gout. Gout in 36 joints. J Rheumatol. 2006;116:2262–71.
1986;11:469. 105. Akahoshi T, Murakami Y, Kitasato H. Recent
86. Fam AG, Stein J, Rubenstein J. Gouty arthritis in advances in crystal-induced acute inflammation.
nodal osteoarthritis. J Rheumatol. 1996;23:684. Curr Opin Rheumatol. 2007;19:146.
87. McGill NW, Dieppe PA. The role of serum and syn- 106. Gersch MS, Johnson RJ. Uric acid and the immune
ovial fluid components in the promotion of urate crys- response. Nephrol Dial Transplant. 2006;
tal formation. J Rheumatol. 1991;18:1042. 21:3046.
88. Tak H-K, Cooper SM, Wilcox WR. Studies on the 107. Petrilli V, Martinon F. The inflammasome, autoin-
nucleation of monosodium urate at 37°C. Arthritis flammatory diseases, and gout. Joint Bone Spine.
Rheum. 1980;23:574. 2007;74:571.
89. Sokoloff L. The pathology of gout. Metabolism. 108. Martinon F, Petrilli V, Mayor A, et al. Gout-
1957;6:230. associated uric acid crystals activate NALP3
90. Katz WA, Schubert M. The solubility of monosodium inflammasome. Nature. 2006;440:237.
urate in connective tissue components. J Clin Invest. 109. Dostert C, Petrilli V, Van Bruggen R, et al. Innate
1970;49:1783. immune activation through NALP3 inflammasome
91. Burt HM, Dutt YC. Growth of monosodium urate sensing of asbestos and silica. Science. 2008;
monohydrate crystals: effect of cartilage and synovial 320:674.
fluid components on in vitro growth. Ann Rheum Dis. 110. Kaisho T, Akira S. Toll-like receptors as adjuvant
1986;45:858. receptors. Biochim Biophys Acta. 2002;1589:1.
92. Brugsch T, Citron J. Ueber die absorption der harn- 111. Kyburz D, Rethage J, Seibl R, et al. Bacterial pepti-
saure durch knorpel. Z Exp Pathol Ther. 1908;5:401. doglycans but not CpG oligodeoxynucleotides acti-
93. Perricone E, Brandt KD. Enhancement of urate solu- vate synovial fibroblasts by toll-like receptor
bility by connective tissue: 1. Effect of proteoglycan signaling. Arthritis Rheum. 2003;48:642.
aggregates and buffer cation. Arthritis Rheum. 112. Medvedev AE, Kopydlowski KM, Vogel SN.
1978;21:453. Inhibition of lipopolysaccharide-induced signal
94. Alvsaker JO. Genetic studies in primary gout: investi- transduction in endotoxin-tolerized mouse mac-
gations on the plasma levels of the uratebinding alpha- rophages: dysregulation of cytokine, chemokine,
282 8 Mechanisms of the Acute Attack of Gout and Its Resolution

and toll-like receptor 2 and 4 gene expression. J independently of TLR-associated signal adaptor
Immunol. 2000;164:5564. molecules. Genes Cells. 2004;9:1055.
113. Gasse P, Riteau N, Charron S, et al. Uric acid is a 133. Martinon F, Tschopp J. Inflammatory caspases: link-
danger signal activating NALP3 inflammasome in ing an intracellular innate immune system to
lung injury inflammation and fibrosis. Am J Respir autoinflammatory diseases. Cell. 2004;117:561.
Crit Care Med. 2009;179:903. 134. Masumoto J, Taniguchi S, Ayukawa K, et al. ASC, a
114. Winzer M, Tausche AK, Aringer M. Crystal-induced novel 22 kDa protein, aggregates during apoptosis of
activation of the inflammasome: gout and pseudog- human promyelocytic leukemia HL-60 cells. J Biol
out. Z Rheumatol. 2009;68(9):733–9. Chem. 1999;274:33835.
115. Janeway Jr CA, Medzhitov R. Innate immune recog- 135. Taniguchi S, Sagara J. Regulatory molecules
nition. Annu Rev Immunol. 2002;20:197. involved in inflammasome formation with special
116. Akira S, Uematsu S, Takeuchi O. Pathogen recogni- reference to a key mediator protein, ASC. Semin
tion and innate immunity. Cell. 2006;124:783. Immunopathol. 2007;29:231.
117. Cassel SL, Eisenbarth SC, Iyer SS, et al. The NALP3 136. Eder C. Mechanisms of interleukin-1beta release.
inflammasome is essential for the development of Immunobiology. 2009;214(7):543–53.
silicosis. Proc Natl Acad Sci USA. 2008;105:9035. 137. Dinarello CA. Biologic basis for interleukin-1 in dis-
118. Medzhitov R, Janeway Jr CA. An ancient system of ease. Blood. 1996;87:2095.
host defense. Curr Opin Immunol. 1998;10:12. 138. Laughlin MJ, Kirkpatrick G, Sabiston N, et al.
119. Stutz A, Golenbock DT, Latz E. Inflammasomes: too Hematopoietic recovery following high-dose com-
big to miss. J Clin Invest. 2009;119:3502. bined alkylating-agent chemotherapy and autolo-
120. Meylan E, Tschopp J, Karin M. Intracellular pattern- gous bone marrow support in patients in phase 1
recognition receptors in the host response. Nature. clinical trials of colony stimulating factors: G-CSF,
2006;442:39. GM-CSF, IL-1, IL-2 and M-CSF. Ann Hematol.
121. Kawai T, Akira S. TLR signaling. Semin Immunol. 1993;67:267.
2007;19:24. 139. Kitamura T, Takaku A. A preclinical and phase 1
122. Werts C, Girardin SE, Philpott OJ. TIR, CARD and clinical trial of IL-1. Exp Med. 1989;7:170.
PYRIN: three domains for an antimicrobial triad. 140. Nemunaitis J, Applebaum FR, Lilliby K, et al. Phase
Cell Death Differ. 2006;13:798. 1 study of recombinant interleukin-1 in patients
123. Fritz JH, Ferraro RL, Philpott OJ, Girardin SE. Nod- undergoing autologous bone marrow transplantation
like proteins in immunity, inflammation and disease. for acute myelogenous leukemia. Blood. 1994;83:
Nat Immunol. 2006;7:1250. 3473.
124. Mariathasan S, Monack OM. Inflammasome adap- 141. Tewari A, Buhles Jr WC, Starnes Jr HF. Preliminary
tors and sensors: intracellular regulators of infection report: effects of interleukin-1 on platelet counts.
and inflammation. Nat Rev Immunol. 2007;7:31. Lancet. 1990;336:712.
125. Martinon F, Tschopp J. NLR join TLRs as innate 142. Crown J, Jakubowski A, Kemeny N, et al. A phase 1
sensors of pathogens. Trends Immunol. 2005;20: trial of recombinant interleukin-1 alone and in combi-
447. nation with myelosuppressive doses of 5-fluorouracil
126. Ting JP-Y, Lovering RC, Alnemri ES, et al. The in patients with gastrointestinal cancer. Blood.
NLR gene family: a standard nomenclature. 1991;78:1420.
Immunity. 2008;28:285. 143. Starnes HF. Biological effects of interleukin-1.
127. Martinon F, Burns K, Tschopp J. The inflammasome: Semin Hematol. 1991;28:43.
a molecular platform triggering activation of 144. Dinarello CA. Immunological and inflammatory
inflammatory caspases and processing proll-beta. functions of the interleukin-1 family. Annu Rev
Mol Cell. 2002;10:417. Immunol. 2009;27:519.
128. Martinon F, Mayor A, Tschopp J. The inflammasome: 145. Lee JK, Kim SH, Lewis EC, et al. Differences in sig-
guardians of the body. Annu Rev Immunol. naling pathways by IL-1 and IL-18. Proc Natl Acad
2009;27:229. Sci USA. 2004;101:8815.
129. Church LD, Cook GP, McDermott MF. Primer: 146. Dinarello CA. Interleukin 1 and interleukin 18 as
inflammasomes and interleukin 1 in inflammatory mediators of inflammation and the aging process.
disorders. Nat Clin Pract Rheumatol. 2008;4:34. Am J Clin Nutr. 2006;83 Suppl 1:4475.
130. Petrilli V, Papin S, Dostert C, et al. Activation of the 147. Dinarello CA. Therapeutic strategies to reduce IL-1
NALP3 inflammasome is triggered by low intracel- activity in treating local and systemic inflammation.
lular potassium concentration. Cell Death Differ. Curr Opin Pharmacol. 2004;4:378.
2007;14:1583. 148. Dinarello CA. Blocking IL-1 in systemic
131. Stehlik C, Lee H, Dorfleutner A, et al. Apoptosis- inflammation. J Exp Med. 2005;201:1355.
associated speck-like protein containing a caspase 149. Apte RN, Voronov E. Interleukin-1 – a major pleio-
recruitment domain is a regulator of procaspase-1 tropic cytokine in tumor-host interactions. Semin
activation. J Immunol. 2003;171:6154. Cancer Biol. 2002;12:277.
132. Yamamoto M, Yaginama K, Tsutsui H, et al. ASC is 150. Martin MU, Wesche H. Summary and comparison of
essential for LPS-induced activation of procaspase-1 the signaling mechanisms of the Toll/interleukin-1
Glucocorticoid 283

receptor family. Biochim Biophys Acta. 2002; leukin (IL)-18, IL-12, and IL-15 in severe melioido-
1592:265. sis. J Infect Dis. 1999;180:1878.
151. Montovani A, Locati M, Polenanitti N, et al. 166. Vasakayalapati R, Wizel B, Weis SE, et al. Production
Extracellular and intracellular decoys in the timing of interleukin-18 in human tuberculosis. J Infect Dis.
of inflammatory cytokines and Toll-like receptors: 2000;180:234.
the new entry TIRS/SIGIRR. J Leukoc Biol. 167. Netea MG, Fantuzzi G, Kullberg BJ, et al.
2004;75:738. Neutralization of IL-18 reduces neutrophil tissue
152. O’Neill LA. Signal transduction pathways activated accumulation and protects mice against lethal
by the IL-1 receptor/toll-like receptor superfamily. Excherichia coli and Salmonella typhimurium endo-
Top Microbial Immunol. 2002;270:47. toxemia. J Immunol. 2000;164:2644.
153. Nakamura K, Okamura H, Wada M, et al. Endotoxin- 168. Tsutsui H, Matsui K, Okamura H, Nakanishi K.
induced serum factor that stimulates gamma inter- Pathophysiological roles of interleukin-18 in
feron production. Infect Immun. 1989;57:590. inflammatory liver diseases. Immunol Rev. 2000;
154. Lebei-Binay S, Berger F, Zinzindohoue PH, et al. 174:192.
Interleukin-18: biological properties and clinical 169. Puren AJ, Fantuzzi G, Dinarello CA. Gene expres-
implications. Eur Cytokine Netw. 2000;11:15. sion, synthesis, and secretion of interleukin 18 and
155. Puren AJ, Razeghi P, Fantuzzi G, Dinarello CA. interleukin 1 beta are differentially regulated in
Interleukin-18 enhances lipopolysaccharideinduced human blood mononuclear cells and mouse spleen
interferon-gamma production in human whole blood cells. Proc Natl Acad Sci USA. 1999;96:2256.
cultures. J Infect Dis. 1998;178:1830. 170. Duncan JA, Gao X, Huang MT, et al. Neisseria
156. Puren AJ, Fantuzzi G, Gu Y, et al. lnterleukin-18 gonorrhoeae activates the proteinase cathepsin B to
(IFN gamma-inducing factor) induces IL-8 and mediate the signaling activities of the NLRP3 and
IL-1beta via TNF-alpha productions from non- ASC-containing inflammasome. J Immunol. 2009;
CD14+ human blood mononuclear cells. J Clin 182:6460.
Invest. 1999;101:711. 171. Allen IC, Scull MA, Moore CB, et al. The NLRP3
157. Kohka H, Yoshino T, Iwagaki H, et al. Interleukin-18/ inflammasome mediates in vivo innate immunity to
interferon-gamma-inducing factor, a novel cytokine, influenza A virus through recognition of viral RNA.
upregulates ICAM-1 (CD54) expression in KG-1 Immunity. 2009;30:556.
cells. J Leukoc Biol. 1998;64:519. 172. Kanneganti TD, Lamkanfi M, Kim YG, et al.
158. Gatti S, Beck J, Fantuzzi G, et al. Effect of interleu- Pannexin-1-mediated recognition of bacterial mole-
kin-18 on mouse core body temperature. Am J cules activates the cryopyrin inflammasome inde-
Physiol Regul Integr Comp Physiol. 2002; pendent of Toll-like receptor signaling. Immunity.
282:R702. 2007;26:433.
159. Li S, Goorha S, Ballou LR, Blatteis CM. 173. Hornung V, Bauernfeind F, Halle A, et al. Silica
Intracerebroventricular interleukin-6, macrophage crystals and aluminum salts activate the NALP3
inflammatory protein-1 beta and IL-18: pyrogenic inflammasome through phagosomal destabilization.
and PGE(2)-mediated? Brain Res. 2003;992:76. Nat Immunol. 2008;9:847, 2008;9:83.
160. Reznekov LL, Kim SH, Westcott JY, et al. IL-18 bind- 174. Sharp EA, Ruane D, Claass B, et al. Uptake of par-
ing protein increases spontaneous and IL-1 induced ticulate vaccine adjuvants by dendritic cells activates
prostaglandin production via inhibition of IFN- the NALP3 inflammasome. Proc Natl Acad Sci
gamma. Proc Natl Acad Sci USA. 2000;97:2174. USA. 2009;106:870.
161. Arndt PG, Fantuzzi G, Abraham E. Expression of 175. Franchi L, Kanneganti TD, Dubyak GR, Nunez GJ.
interleukin-18 in the lung after endotoxemia or hem- Differential requirement of P2X7 receptor and intra-
orrhage-induced acute lung injury. Am J Respir Cell cellular bacteria. J Biol Chem. 2007;282:188.
Mol Biol. 2000;22:708. 176. Fernandez-Ainemri T. The pyroptosome: a supramo-
162. Yoshimato T, Tsutsui H, Tominaga K, et al. IL-18, lecular assembly of ASC dimmers mediating
although antiallergic when administered with IL-12, inflammatory cell death via caspase-1 activation.
stimulates IL-4 and histamine release by basophils. Cell Death Differ. 2007;14:1590.
Proc Natl Acad Sci USA. 1999;96:13962. 177. Forman HJ, Torres M. Reactive oxygen species and
163. Daemen MA, van’t Veer C, Wolfs TG, Buurman cell signaling: respiratory burst in macrophage sig-
WA. Ischemia/reperfusion-induced IFNgamma up- naling. Am J Respir Crit Care Med. 2002;166:S4.
regulation: involvement of IL-12 and IL-18. J 178. Sekiyama A, Ueda H, Kashiwamura S, et al. A
Immunol. 1999;162:5506. stress-induced, superoxide-mediated caspase-1 acti-
164. Yamada G, Shijubo N, Shigehara K, et al. Increased vation pathway causes IL-18 upregulation. Immunity.
levels of circulating interleukin-18 in patients with 2005;22:669.
advanced tuberculosis. Am J Respir Crit Care Med. 179. Cruz CM, Rinna A, Forman HJ, et al. ATP activates
2000;161:1786. a reactive organ species-dependent oxidative
165. Lauw FN, Simpson AF, Prins JM, et al. Elevated stress response and secretion of proinflammatory
plasma concentrations of interferon (IFN) gamma cytokines in macrophages. J Biol Chem. 2007;
and the interferon-gamma inducing cytokines inter- 282:2871.
284 8 Mechanisms of the Acute Attack of Gout and Its Resolution

180. Walev I, Reske K, Palmer M, et al. Potassium- 194. Newman ZL, Lappla SH, Moayeri M. CA074 Me
inhibited processing of IL-1 beta in human mono- protection against anthrax lethal toxin. Infect Immun.
cytes. EMBO J. 1995;14:1607. 2009;77:4327.
181. Perregaux D, Gabel C. Interleukin-1 beta maturation 195. Bazzoni F, Cassatella MA, Rossi F, et al.
and release in response to ATP and nigericin. Phagocytosing neutrophils produce and release high
Evidence that potassium depletion mediated by these amounts of neutrophil-activating peptide-1/interleu-
agents is a necessary and common feature of their kin-8. J Exp Med. 1991;173:771.
activity. J Biol Chem. 1994;269:15195. 196. Ben-Baruch A, Michiel DF, Oppenheim JJ. Signals
182. Colomar A, Marty V, Medina C, et al. Maturation and receptors involved in recruitment of inflammatory
and release of interleukin-1 beta by lipopolysaccha- cells. J Biol Chem. 1995;270:11703.
ride-primed mouse Schwann cells require the stimu- 197. Foxman EF, Kunkel EJ, Butcher EC. Integrating
lation of P2X7 receptors. J Biol Chem. 2003; conflicting chemotactic signals: the role of memory
278:30732. in leukocyte navigation. J Cell Biol. 1999;147:577.
183. Cain K, Langlais C, Sun XM, et al. Physiological 198. Ahuja SK, Murphy PM. The CXC chemokines
concentrations of K+ inhibit cytochrome cdependent growth-regulated oncogene (GRO)a, GR0!3, GROy,
formation of the apoptosome. J Biol Chem. 2001; neutrophil-activating peptide-78 and epithelial
276:41985. cell-derived neutrophil-activating peptide-78 are
184. Faustin B, Lartigue L, Bruey JM, et al. Reconstituted potent agonists for the type B, but not the type A,
NALP1 inflammasome reveals two-step mechanism human interleukin-8 receptor. J Biol Chem. 1996;
of caspase-1 activation. Mol Cell. 2007;25:713. 271:20545.
185. Marina-Garcia N, Franchi L, Kim YG, et al. 199. Allport JR, Lim YC, Shipley JM, et al. Neutrophils
Pannexin-1-mediated intracellular delivery of mura- from MMP-9- or neutrophil elastase-deficient mice
nyl dipeptide induces caspase-1 activation via cry- show no defect in transendothelial migration under
opyrin/NLRP3 independently of Nod2. J Immunol. flow in vitro. J Leukoc Biol. 2002;71:821.
2008;180:4050. 200. Hsu MH, Wang M, Browning DD, et al. NF-KB acti-
186. Meissner F, Malawi K, Zychlinsky A. Superoxide vation is required for C5a-induced interleukin-8
dismutase 1 regulates caspase-1 and endotoxin gene expression in mononuclear cells. Blood.
shock. Nat Immunol. 2008;9:866. 1999;93:3241.
187. Heutze H, Lin XY, Choi MS, Porter AG. Critical role 201. Robbins RA, Hamel FG. Chemotactic factor inacti-
for cathepsin B in mediating caspase-1- dependent vator interaction with Gc-globulin (vitamin Dbinding
interleukin-18 maturation and caspase-1-indepen- protein). A mechanism of modulating the chemotac-
dent necrosis triggered by the microbial toxin nigeri- tic activity of C5a. J Immunol. 1990;144:2371.
cin. Cell Death Differ. 2003;10:956. 202. Gerber BO, Meng EC, Dotsch V, et al. An activation
188. Bauernfeind FC, Horvath G, Stutz A, et al. Cutting switch in the ligand binding pocket of the C5a recep-
edge: NF-kappa B activating pattern recognition and tor. J Biol Chem. 2001;276:3394.
cytokine receptors license NLRP3 inflammasome 203. Braun L, Christophe T, Boulay F. Phosphorylation of
activation by regulating NLRP3 expression. J key serine residues is required for internalization of
Immunol. 2009;183:787. the complement 5a (C5a) anaphylatoxin receptor via
189. Filippini A, Taffs RE, Sitkovsky MV. Extracellular a 13-arrestin, dynamin, and clathrin-dependent path-
ATP in T-lymphocyte activation: possible role in way. J Biol Chem. 2003;278:4277.
effector functions. Proc Natl Acad Sci USA. 204. Mcintyre TM, Zimmerman GA, Prescott SM.
1990;87:8267, 1991;88:6899. Biologically active oxidized phospholipids. J Biol
190. Netea MG, Nold-Petry CA, Nold MF, et al. Chem. 1999;274:25189.
Differential requirement for the activation of the 205. Shen Y, Sultana C, Arditi M, et al. Endotoxin-
inflammasome for processing and release of IL-1 induced migration of monocytes and PECAM-1
beta in monocytes and macrophages. Blood. phosphorylation are abrogated by PAF receptor
2009;113:2324. antagonists. Am J Physiol. 1998;275:E479.
191. Ferrari D, Chiozzi P, Falzoni S, et al. Purinergic 206. Prescott SM, Zimmerman GA, Mcintyre TM. Platelet-
modulation of interleukin-1 beta release from micro- activating factor. J Biol Chem. 1990;265:17381.
glial cells stimulated with bacterial endotoxin. J Exp 207. Nieto ML, Velasco S, Sanchez Crespo M.
Med. 1997;185:579. Biosynthesis of platelet activating factor in human
192. Piccini A, Carta S, Tassi S, et al. ATP is released by polymorphonuclear leukocytes. Involvement of the
monocytes stimulated with pathogen sensing recep- choline-phosphotransferase pathway in response to
tor ligands and induces IL-1 beta and IL-18 secretion the phorbol esters. J Biol Chem. 1988;263:2217.
in an autocrine way. Proc Natl Acad Sci USA. 208. Baker PR, Owen JS, Nixon AB, et al. Regulation of
2008;105:8067. platelet-activating factor synthesis in human neutro-
193. Koo IC, Wang C, Raghavan S, et al. ESX-1- phils by MAP kinases. Biochim Biophys Acta.
dependent cytolysis is in lysosome secretion and 2002;1592:175.
inflammasome activation during mycobacterial 209. Marquez LA, Huang JT, Dunford HR. Spectral and
infection. Cell Microbiol. 2008;10:1866. kinetic studies on the formation of myeloperoxidase
Glucocorticoid 285

compounds I and II: roles of hydrogen peroxide and 225. Haeggstrom JZ. Leukotriene hydrolase/aminopepti-
superoxide. Biochemistry. 1994;33:1447. dase, the gatekeeper of chemotactic leukotriene B4
210. Venable ME, Zimmerman GA, Mcintyre TM, biosynthesis. J Biol Chem. 2004;279:50639.
Prescott SM. Platelet-activating factor: a phospho- 226. Rybins IV, Feinmark SJ. Alteration of human leukot-
lipid autacoid with diverse actions. J Lipid Res. riene A4 hydrolase activity after sitedirected muta-
1993;34:691. genesis: serine-415 is a regulatory residue. Biochim
211. Chilton FH, O’Fiaherty JT, Walsh CE, et al. Platelet- Biophys Acta. 1999;1438:199.
activating factor. Stimulation of the lipoxygenase 227. Mandai AK, Skoch J, Baeskai BJ, et al. The mem-
pathway in polymorphonuclear leukocytes by 1-0- brane organization of leukotriene synthesis. Proc
alkyl-2-0-acetyl-sn-glycero-3- phosphocholine. J Natl Acad Sci USA. 2004;101:6587.
Biol Chem. 1982;257:5402. 228. Helgadoltir A, Manolexcu A, Thorleifson G, et al.
212. Springer TA. Adhesion receptors of the immune sys- The gene encoding 5-lipoxygenase activating pro-
tem. Nature. 1990;346:425. tein confers risk of myocardial infarction and stroke.
213. Mitchell S, Thomas G, Harvey K, et al. Lipoxins, Nat Genet. 2004;36:233.
aspirin-triggered epi-lipoxins, liposin stable ana- 229. Kudo I. Diversity of phospholipase A2 enzymes.
logues, and the resolution of inflammation: stimula- Biol Pharm Bull. 2004;27:1157.
tion of macrophage phagocytosis of apoptotic 230. Jenkins CM, Mancuso DJ, Yan W, et al. Identification,
neutrophils in vivo. J Am Soc Nephrol. 2002; cloning, expression, and purification of three novel
13:2497. human calcium-independent phospholipase A2 fam-
214. Paul-Clark MJ, van Cao T, Moradi-Bidhendi N, et al. ily members possessing triacylglycerollipase and
15-epi-lipoxin A,-mediated induction of nitric oxide acylglycerol transacylase activities. J Biol Chem.
explains how aspirin inhibits acute inflammation. 2004;48968.
J Exp Med. 2004;200:69. 231. Murakami M, Kudo I. Secretory phospholipase A2.
215. Ferrante JV, Ferrante A. Cutting edge: novel role of Biol Pharm Bull. 2004;27:1158.
lipoxygenases in the inflammatory response: promo- 232. Smith ML, DeWitt DL, Garavito RM.
tion of TNF mRNA decay by 15-hydroperoxyei- Cyclooxygenases: structural, cellular, and molecular
cosatetraenoic acid in a monocytic cell line. biology. Annu Rev Biochem. 2000;69:145.
J Immunol. 2005;174:3169. 233. Six DA, Dennis EA. The expanding superfamily of
216. Ruchand-Sparagano MH, Walker TR, Rossi AG, phospholipase A(2) enzymes: classification and
et al. Soluble E-selectin acts in synergy with platelet- characterization. Biochim Biophys Acta. 2000;
activating factor to activate neutrophill32-integrins. 1488:1.
J Biol Chem. 2000;275:15758. 234. Carnevale KA, Cathcart MK. Calcium-independent
217. Exton JH. Signaling through phosphatidylcholine phospholipase A2 is required for human monocyte
breakdown. J Biol Chem. 1990;265:1. chemotaxis to monocyte chemoattractant protein 1. J
218. Tjoelker LW, Wilder C, Eberhardt C, et al. Anti- Immunol. 2001;167:3414.
inflammatory properties of a platelet-activating fac- 235. Jones SM, Luo M, Healy AM, et al. Structural and
tor acetylhydrolase. Nature. 1995;374:549. functional criteria reveal a new nuclear import
219. Hattori K, Adachi H, Matsuzawa A, et al. eDNA sequence on the 5-lipoxygenase protein. J Biol
cloning and expression of intracellular platelet acti- Chem. 2002;277:38550.
vating factor (PAF) acetylhydrolase. II. Its homol- 236. Hanaka H, Shimizu T, Izumi T. Nuclear-localization-
ogy with plasma PAF acetylhydrolase. J Biol Chem. signa-dependent mechanisms determine the local-
1996;271:33032. ization of 5-lipoxygenase. Biochem J. 2002;
220. Newcombe DS. Leukotrienes: regulation of biosyn- 361:505.
thesis, metabolism and bioactivity. J Clin Pharmacol. 237. Flamand N, Surette ME, Picard S, et al. Cyclic
1988;28:530. AMP-mediated inhibition of 5-lipoxygenase translo-
221. Murray J, Ward C, O’Fiaherty JT, et al. Role of the cation and leukotriene biosynthesis in human neu-
leukotrienes in the regulation of human granulocyte trophils. Mol Pharmacol. 2002;62:250.
behaviour: dissociation between agonist-induced 238. Oppenheim JJ, Zachariae OC, Mukaida N, et al.
activation and retardation of apoptosis. Br J Properties of the novel proinflammatory supergene
Pharmacol. 2003;139:388. “intercrine” cytokine family. Annu Rev Immunol.
222. Turner M, Schweighoffer E, Colucci F, et al. Tyrosine 1991;9:617.
kinase SYK: essential functions for immunoreceptor 239. Luster AD. Chemokines-chemotactic cytokines that
signalling. Immunol Today. 2000;21:148. mediate inflammation. N Engl J Med. 1998;338:436.
223. Canetti C, Hu B, Curtis JL, Peters-Golden M. Syk 240. Nanki T, Nagasaka K, Hayashida K, et al.
activation is a leukotriene B4-regulated event Chemokines regulate IL-6 and IL-8 production by
involved in macrophage phagocytosis of IgG-coated fibroblast-like synoviocytes from patients with rheu-
targets but not apoptotic cells. Blood. 2003; matoid arthritis. J Immunol. 2001;167:5381.
102:1877. 241. Ohashi K, Naruto M, Nakaki T, Sana E. Identification
224. Droge W. Free radicals in the physiological control of interleukin-8 converting enzyme as cathepsin L.
of cell function. Physiol Rev. 2002;82:47. Biochim Biophys Acta. 2003;1649:30.
286 8 Mechanisms of the Acute Attack of Gout and Its Resolution

242. Yu Y, Chadee K. The 3¢-untranslated region of 260. Humphries MJ. Integrin structure. Biochem Soc
human interleukin-8 mRNA suppresses IL-8 gene Trans. 2000;28:34.
expression. Immunology. 2001;102:498. 261. Harris ES, Mcintyre TM, Prescott SM, Zimmerman
243. de Bruin T, de Rooster H, van Bree Hand, Cox E. GA. The leukocyte integrins. J Biol Chem. 2000;275:
lnterleukin-8 mRNA expression in synovial fluid of 23409.
canine stifle joints. Vet Immunol Immunopathol. 262. You TJ, Maxwell DS, Kogan TP, et al. A 3D struc-
2005;108:387. ture model of integrin a4131 complex: I. Construction
244. Conti P, Reale M, Barbacane RC, et al. Differential of a homology model of 131 and ligand binding
production of RANTES and MCP-1 in synovial fluid analysis. Biophys J. 2002;82:447.
from the inflamed human knee. Immunol Lett. 263. Mould AP, Askari JA, Barton S, et al. Integrin acti-
2002;80:105. vation involves a conformational change in the a1
245. Vestweber D, Blanks JE. Mechanisms that regulate helix of the subunit A-domain. J Biol Chem.
the function of selectins and their ligands. Physiol 2002;277:19800.
Rev. 1999;79:181. 264. Schneider D, Engelman DM. Involvement of trans-
246. Bernimoulin MP, Zeng XL, Abbal C, et al. Molecular membrane interactions in signal transduction by
basis of leukocyte rolling on PSGL-1:predominant integrins. J Biol Chem. 2004;279:9840.
role of core-20-glycans and of tyrosine sulfate resi- 265. Salas A, Shimaoka A, Phan U, et al. Transition from
due 51. J Biol Chem. 2003;278:37. rolling to firm adhesion can be mimicked by exten-
247. Hammer DA. Leukocyte adhesion: what’s the catch? sion of integrin a1b2 in an intermediate affinity
Curr Biol. 2005;15:R96. state. J Biol Chem. 2006;281:10876.
248. Rinko LJ, Lawrence MB, Guilford WH. The molecular 266. Chen J, Yang W, Kim M, et al. Regulation of out-
mechanics of P- and L-selectin lectin domains bind- side-in signaling and affinity by the beta2 I domain
ing to PSGL-1. Biophys J. 2004;86:544. of integrin alphaLbeta2. Proc Natl Acad Sci USA.
249. Berg EL, McEvoy LM, Berlin C, et al. L-selectin- 2006;103:13062.
mediated lymphocyte rolling on MAdCAM-1. 267. Zimmerman GA, Mcintyre TM, Prescott SM.
Nature. 1993;366:695. Adhesion and signaling in vascular cell-cell interac-
250. von Andrian UH, Mempel TR. Homing and cellular tions. J Clin Invest. 1996;98:1699.
traffic in lymph nodes. Nat Rev Immunol. 2003; 268. Robinson JM, Kobayashi T. A novel intracellular
3:867. compartment with unusual secretory properties in
251. Hafezi-Moghadam A, Thomas KL, Prorock AJ, human neutrophils. J Cell Biol. 1991;113:743.
et al. L-selectin shedding regulates leukocyte recruit- 269. Lupher ML, Harris EAS, Beals CR, et al. Cellular
ment. J Exp Med. 2001;193:863. activation of leukocyte function-associated antigen-1
252. Walcheck B, Kahn J, Fisher JM, et al. Neutrophil and its affinity are regulated at the I domain allos-
rolling altered by inhibition of L-selectin shedding teric site. J Immunol. 2001;167:1431.
in vitro. Nature. 1996;380:720. 270. Dib K, Melander F, Axelsson L, et al. Down-
253. Matala E, Alexander SR, Kishimoto TK, Walcheck regulation of Rae activity during b2 integrin-
B. The cytoplasmic domain of L-selectin participates medi ated adhesion of human neutrophils. J Biol
in regulating L-selectin endoproteolysis. J Immunol. Chem. 2003;278:24181.
2001;167:1617. 271. Werner E, Werb Z. lntegrins engage mitochondrial
254. Wagner JG, Roth RA. Neutrophil migration mecha- function for signal transduction by a mechanism
nisms, with an emphasis on the pulmonary vascula- dependent on Rho GTPases. J Cell Biol. 2002;
ture. Pharmacol Rev. 2000;52:349. 158:357.
255. Smalley DM, Ley K. L-selectin: mechanisms and 272. Van der Vieren M, Le Trong H, Wood CL, et al. A
physiological significance of ectodomain cleavage. novelleukointegrin, ad132 binds preferentially to
J Cell Mol Med. 2005;9:255. ICAM-3. Immunity. 1995;3:683.
256. Gonzales PE, Solomon A, Miller AB, et al. Inhibition 273. Neelamegham S, Taylor AD, Shankaran H, et al.
of tumor necrosis factor-a-converting enzyme by its Shear and time-dependent changes in Mac-1, LFA-1,
prodomain. J Biol Chem. 2004;279:31638. and ICAM-3 binding regulate neutrophil homotypic
257. Dal Secco D, Moreira AP, Freitas A, et al. Nitric adhesion. J Immunol. 2000;164:3798.
oxide inhibits neutrophil migration by a mechanism 274. Serrander JM, Vicente-Manzanares M, Calvo J, et al.
dependent on ICAM-1: role of soluble guanylate A novel serine-rich motif in the intercellular adhe-
cyclase. Nitric Oxide. 2006;15:77–86. sion molecule 3 is critical for its ezrin/radixin/
258. Deng GM, Zheng L, Chan FK, Lenardo M. moesin-directed subcellular targeting. J Biol Chem.
Amelioration of inflammatory arthritis by targeting 2002;277:10400.
the pre-ligand assembly domain of tumor necrosis 275. Lehmann JCU, Jablonski-Westrich D, Haubold U,
factor receptors. Nat Med. 2005;11:1066. et al. Overlapping and selective roles of endothelial
259. Jutila MA, Kurk S, Jackiw L, et al. L-selectin serves intercellular adhesion molecule-1 (ICAM-1) and
as an E-selectin ligand on cultured T lymphoblasts. ICAM-2 in lymphocyte trafficking. J Immunol.
J Immunol. 2002;169:1768. 2003;171:2588.
Glucocorticoid 287

276. Lum AFH, Green CE, Lee GR, et al. Dynamic regu- 290. Groemping Y, Lapouge K, Smerdon SJ, Rittinger K.
lation of LFA-1 activation and neutrophil arrest on Molecular basis of phosphorylationinduced activa-
intercellular adhesion molecule 1 (ICAM-1) in shear tion of the NADPH oxidase. Cell. 2003;113:343.
flow. J Biol Chem. 2002;277:20660. 291. Kettle AJ, Clark BM, Winterboum CC. Superoxide
277. Alevriadou BR. CAMs and Rho small GTPases: converts indigo carmine to isatin sulfonic acid:
gatekeepers for leukocyte transendothelial migra- implications for the hypothesis that neutrophils pro-
tion. Focus on “VCAM-1-mediated Rae signaling duce ozone. J Biol Chem. 2004;279:18521.
controls endothelial cell-cell contacts and leukocyte 292. Zgliczynski J, Stelmaszynska T. Chlorinating ability
transmigration”. Am J Physiol Cell Physiol. 2003; of human phagocytosing leukocytes. Eur J Biochem.
285:C250. 1995;56:157.
278. Nick JA, Young SK, Arndt PG, et al. Selective sup- 293. Henderson JP, Byun J, Heinecke JW. Molecular
pression of neutrophil accumulation in ongoing pul- chlorine generated by the myeloperoxidase hydro-
monary inflammation by systemic inhibition of p38 gen peroxide-chlorine system of phagocytes pro-
mitogen-activated protein kinase. J Immunol. 2002; duces 5-chlorocytosine in bacterial RNA J. Biol
169:5260. Chem. 1999;274:33440.
279. Watts GM, Beurskens FJM, Martin-Podura I, et al. 294. Peskin AV, Wlnterboum CC. Histamine chloramine
Manifestations of inflammatory arthritis are criti- reactivity with thiol compounds, ascorbate and
cally dependent on LFA-1. J Immunol. 2005; methionine and with intracellular glutathione. Free
174:3668. Radic Biol Med. 2003;35:1252.
280. Heuertz RM, Schneider GP, Potempa LA, Webster 295. Rosen H, Crowley JR, Heinecke JW. Human
RO. Native and modified C-reactive protein bind neutrophils use the myleperoxidase-hydrogen
different receptors on human neutrophils. Int J peroxide-chloride system to chlorinate but not nitrate
Biochem Cell Biol. 2005;37:320. bacterial proteins during phagocytosis. J Biol Chem.
281. Khreiss T, Jozsef L, Potempa LA, Filep JG. 2002;277:30463.
Conformational rearrangement in C-reactive protein 296. Klebanoff SJ. Myeloperoxidase: friend or foe.
is required for proinflammatory actions on human J Leukoc Biol. 2005;77:1.
endothelial cells. Circulation. 2004;109:2016. 297. Lymar SV, Khairutdinov RF, Hurst JK. Hydroxyl
282. Fecteau M-H, Honaare J-C, Plante M, et al. radical formation by 0–0 bond homolysis in per-
Endothelin-1 (1–31) is an intermediate in the pro- oxynitrous acid. Inorg Chem. 2003;42:5259.
duction of endothelin-1 after big endothelin-1 298. Brennan ML, Wu W, Fu X, et al. A tale of two con-
administration in vivo. Hypertension. 2005;46:87. troversies: defining both the role of peroxidases in
283. Nauseef WM. The NADPH-dependent oxidase of nitrotyrosine formation in vivo using eosinophil
phagocytes. Proc Assoc Am Physicians. 1999; peroxidase and myeloperoxidase-generated reac-
111:373. tive nitrogen species. J Biol Chem. 2002;
284. Tsunawaki S, Kagara S, Yoshikawa K, et al. 277:L17415.
Involvement of p40phox in activation of phagocytic 299. Bomalaski JS, Baker DG, Brophy LM, Clark MA.
NADPH oxidase through association of its carboxyl- Monosodium urate crystals stimulate phospholipase
terminal, but not its amino-terminal, with p67phox. A2 enzyme activities and the synthesis of a phospho-
J Exp Med. 1996;184:893. lipase A2-activating protein. J Immunol. 1990;
285. Fontayne A, Dang PM, Gougerot-Pocidalo MA, 145:3391.
EI-Benna J. Phosphorylation of p47phox sites by 300. Furlaneto CJ, Campa A. A novel function of serum
PKC alpha, beta II, delta, and zeta: effect on binding amyloid A: a potent stimulus for the release of tumor
to p22phox and on NADPH oxidase activation. necrosis factor-alpha, interleukin-1beta, and inter-
Biochemistry. 2002;41:7743. leukin-8 by human blood neutrophils. Biochem
286. Chen Q, Powell OW, Rune MJ, et al. Akt phosphory- Biophys Res Commun. 2000;268:405.
lates p47phox and mediates respiratory burst activity 301. Ginsberg MH, Jaques B, Cochrane GG, et al. Urate
in human neutrophils. J Immunol. 2003;170:5302. crystal-dependent cleavage of Hageman factor in
287. Sarfstein R, Gozzalczany Y, Mizrahi A, et al. Dual human plasma and synovial fluid. J Lab Clin Med.
role of Rae in the assembly of NADPH oxidase, teth- 1980;95:497.
ering to the membrane and activation of p67phox. 302. Wiggins RC, Cochrane GG. The autoactivation of
J Biol Chem. 2004;279:16007. rabbit Hageman factor. J Exp Med. 1979;150:1122.
288. Ago T, Kuribayashi F, Hiroaki H, et al. Phospho- 303. van Der Graaf F, Tans G, Bouma BN, Griffin JH.
rylation of p47phox directs phox homology domain Isolation and functional properties of the heavy and
from SH3 domain toward phosphoinositides, leading light chains of human plasma kallikrein. J Biol
to phagocyte NADPH oxidase activation. Proc Natl Chem. 1982;257:14300.
Acad Sci USA. 2003;100:4474. 304. Joseph K, Ghebrehiwet B, Peerschke EIB, et al.
289. Werner E. GTPases and reactive oxygen species: Identification of the zinc-dependent endothelial cell
switches for killing and signaling. J Cell Sci. 2004; binding protein for high molecular weight kininogen
117:143. and factor XII: identify with the receptor that binds
288 8 Mechanisms of the Acute Attack of Gout and Its Resolution

to the globular “heads” of C1q (gCiq-R). Proc Natl 321. Serhan CN. A search for endogenous mechanisms of
Acad Sci USA. 1996;93:8552. anti-inflammation uncovers novel chemical media-
305. Aupperle KR, Yamanishi Y, Bennett BL, et al. tors: missing links to resolution. Histochem Cell
Expression and regulation of inducible lkappaB Biol. 2004;122:305.
kinase (IKK-i) in human fibroblast-like synovio- 322. Bannenberg GL, Chiang N, Ariel A, et al. Molecular
cytes. Cell Immunol. 2001;214:54. circuits of resolution: formation and actions of
306. Morand EF, Hall P, Hutchinson P, Yang YH. resolvins and protectins. J Immunol. 2005;174:4345.
Regulation of annexin I in rheumatoid synovial cells 323. Straus OS, Glass CK. Cyclopentenone prostaglan-
by glucocorticoids and interleukin-1. Mediators dins: new insights on biological activities and cellu-
Inflamm. 2006;2006:1. lar targets. Med Res Rev. 2001;21:185.
307. Bechoa S, Daniel LW. Phospholipase D is required 324. Serhan CN, Maddox JF, Petasis NA, et al. Design of
in the signaling pathway leading to p38 MAPK acti- lipoxin stable analogs that block transmigration and
vation in neutrophil-like HL-60 cells, stimulated by adhesion of human neutrophils. Biochemistry.
N-formyl-methionyl-leucylphenylalanine. J Biol Chem. 1995;34:14609.
2001;276:31752. 325. Serhan CN, Fierro IM, Chiang N, Pouliot M. Cutting
309. Exton JH. New developments in phospholipase D. J edge: nociceptin stimulates neutrophil chemotaxis
Biol Chem. 1997;272:15579. and recruitment inhibition by aspirin-triggered-15-
309. Liscovitch M, Czarny M, Fiucci G, Tang X. epi-lipoxin. J Immunol. 2001;166:3650.
Phospholipase D: molecular and cell biology of 326. Serhan C. Lipoxins and aspirin-triggered 15 epi-
a novel gene family. Biochem J. 2000;345:401. lipoxin biosynthesis: an update and role on anti-
310. Sergeant S, Waite KA, Heravi J, McPhail LC. inflammatory and proresolution. Prostaglandins
Phosphatidic acid regulates tyrosine phosphorylating Other Lipid Mediat. 2002;68–69:433.
activity in human neutrophils. J Biol Chem. 2001; 327. Ariel A, Serhan CN. Resolvins and protectins in the
276:4737. termination program of acute inflammation. Trends
311. Desaulniers P, Fernandes M, Gilbert C, et al. Crystal- Immunol. 2007;28:176.
induced neutrophil activation. VII. Involvement of 328. Serhan CN. Novel lipid mediators and resolution
Syk in the response to monosodium urate crystals. mechanisms in acute inflammation: to resolve or
J Leukoc Biol. 2001;70:659. not? Am J Pathol. 2010;177(4):1576–91.
312. Kaldi K, Szeberenyi J, Rada BK, et al. Contribution doi:10.2353/ajpath.2010.100322.
of phospholipase D and a brefeldin A-sensitive ARF 329. Martin C, Burdon PCE, Bridger G, et al. Chemokine
to chemoattractant-induced superoxide production acting via CXCR2 and CXCR4 control the release of
and secretion of human neutrophils. J Leukoc Biol. neutrophils from the bone marrow and their return
2002;71:695. following senescence. Immunity. 2003;19:583.
313. Oka M, Hitomi T, Okada T, et al. Dual regulation of 330. Gomez PF, Pillinger MH, Attur M, et al. Resolution
phospholipase D1 by protein kinase C alpha in vivo. of inflammation: prostaglandin E2 dissociates
Biochem Biophys Res Commun. 2002;294:1109. nuclear trafficking of individual NF-KB subunits
314. Takesono A, Finkelstein LD, Schwartzberg PL. (p65, p50) in stimulated rheumatoid synovial
Beyond calcium: new signaling pathways for Tec fibroblasts. J Immunol. 2005;175:6924.
family kinases. J Cell Sci. 2002;115:3039. 331. Wu X, Zimmerman GA, Prescott SM, Stafforini
315. Hengartner MO. The biochemistry of apoptosis. DM. The p38 MAPK pathway mediates transcrip-
Nature. 2000;407:770. tional activation of plasma platelet-activating factor
316. Serhan CN. Resolution phases of inflammation: acetylhydrolase gene in macrophages stimulated
novel endogenous anti-inflammatory and proresolv- with lipopolysaccharide. J Biol Chem. 2004;279:
ing lipid mediators and pathways. Annu Rev 36158.
Immunol. 2007;25:101. 332. Lawrence T, Bebien N, Lin GY, et al. IKKa limits
317. Rose OM, Liu-Bryan R. Innate immunity in trigger- macrophage NF-KB activation and contributes to the
ing and resolution of acute gouty inflammation. Curr resolution of inflammation. Nature. 2005;434:1138.
Rheumatol Rep. 2006;8:209. 333. Fiorucci S, Distrutti E, Mencarelli A, et al. Evidence
318. Lodige I, Marg A, Wiesner B, et al. Nuclear export that 5-lipoxygenase and acetylated cyclooxygenase
determines the cytokine sensitivity of STAT tran- 2-derived eicosanoids regulate leukocyte-endothelial
scription factors. J Biol Chem. 2005;280:43087. adherence in response to aspirin. Br J Pharmacol.
319. Gilmore TO, Herscovitch M. Inhibitors of 2003;139:1351.
NF-kappaB signaling: 785 and counting. Oncogene. 334. Taylor EL, Rossi AG, Shaw CA, et al. GEA3162
2006;25:6887. decomposes to co-generate nitric oxide and superox-
320. Isomura I, Morita A. Regulation of NF-KB signaling ide and induces apoptosis in human neutrophils via a
by decoy oligodeoxynucleotides. Microbiol Immunol. peroxynitrite-dependent mechanism. Br J Pharmacol.
2006;50:559. 2004;143:179.
Glucocorticoid 289

335. Smith KR, Pinkerton KE, Watanabe T, et al. Attenuation 343. Choi BR, Kwon JH, Gong SJ, et al. Expression of
of tobacco smoke-induced lung inflammation by treat- glucocorticoid receptor mRNAs in glucocorticoid-
ment with a soluble epoxide hydrolase inhibitor. Proc resistant nasalpolyps. Exp Mol Med. 2006;38:466.
Natl Acad Sci USA. 2005;102:2186. 344. Hafezi-Moghadam A, Simoncini T, Yang Z, et al.
336. Doucas V, Shi Y, Miyamoto S, et al. Cytoplasmic Acute cardiovascular effects of corticosteroids are
catalytic subunit of protein kinase A mediates cross- mediated by non-transcriptional activation of endothe-
repression by NF-kappa Band the glucocorticoid lial nitric oxide synthase. Nat Med. 2002;8:473.
receptor. Proc Natl Acad Sci USA. 2000;97:11893. 345. Ismaili N, Garabedian MJ. Modulation of glucocor-
337. Tomita T, Takeuchi E, Tomita N, et al. Suppressed ticoid receptor function via phosphorylation. Ann N
severity of collagen-induced arthritis by in vivo Y Acad Sci. 2004;1024:86.
transfection of nuclear factor kappa B decoy oli- 346. Ichijo T, Vontetakis A, Cotrim AP, et al. The Smad6-
godeoxy nucleotides as a gene therapy. Arthritis histone deacetylase 3 complex silences the tran-
Rheum. 1999;42:2532. scriptional activity of the glucocorticoid receptor
338. Yamamoto Y, Gaynor RB. Therapeutic potential of Potential clinical implications. J Biol Chem. 2005;
inhibition of the NF-kappa B pathway in the treat- 280:42067.
ment of inflammation and cancer. J Clin Invest. 347. Hampsey M, Reinberg D. Tails of intrigue: phospho-
2001;107:135. rylation of RNA polymerase II mediates histone
339. Almawi WY, Melemedjian OK. Negative regulation methylation. Cell. 2003;113:429.
of nuclear factor-KB activation and function by glu- 348. Bird G, Zorio DAR, Bentley DL. RNA polymerase
cocorticoids. J Mol Endocrinol. 2002;28:69. II carboxy-terminal domain phosphorylation is
340. La NZ, Cidlowski JA. The origin and functions of required for cotranscriptional pre-mRNA splicing
multiple human glucocorticoid receptor isoforms. and 3¢-end formation. Mol Cell Biol. 2004;
Ann N Y Acad Sci. 2004;1024:102. 24:8963.
341. Li L, Leung DYM, Hall CF, Goleva E. Divergent 349. Roth SY, Denu JM, Allis CD. Histone acetyltrans-
expression and function of glucocorticoid receptor ferases. Annu Rev Biochem. 2001;70:81.
13 in human monocytes and T cells. J Leukoc Biol. 350. Gerke V, Moss SE. Annexins: from structure to func-
2006;79:818. tion. Physiol Rev. 2001;82:331.
342. Pujols L, Mullol J, Roca-Ferrer J, et al. Expression 351. Danielsen EM, Van Deurs B, Hansen GH.
of glucocorticoid receptor a- and 13-isoforms in “Nonclassical” secretion of annexin A2 to the lumi-
human cells and tissues. Am J Physiol Cell Physiol. nal side of the enterocyte brush border membrane.
2002;283:C1324. Biochemistry. 2003;42:14670.
Management of Hyperuricemia
and Gout 9

Overview mia. Corticosteroids may have a modest uricosu-


ric effect but are never used to regulate uric acid
Management plans for the treatment of gout have metabolism or to reduce the serum uric acid level.
to be designed to meet the requirements of each In contrast to anti-inflammatory drugs, agents
individual patient and to control both gout and its used to control hyperuricemia and hyperuricaci-
associated disorders. The objectives of such plans duria have absolutely no anti-inflammatory activ-
are as follows: (1) to initiate the resolution of an ity and, in fact, may trigger an episode of acute
acute episode of gout by using the appropriate gout associated with the changes in the uric acid
anti-inflammatory drugs, (2) to assess by history, pool induced by this drug. Furthermore, the con-
physical examination, and laboratory analyses trol of acute gout with anti-inflammatory drugs is
any underlying pathophysiological abnormalities absolutely required for the well-being of the
associated with hyperuricemia or gout, (3) to patient, whereas the reduction in the serum uric
treat any pathophysiological abnormalities com- acid level may not always be required.
plicating the gouty diathesis such as hyperlipi- Second, prescribing uric acid-lowering ther-
demia, hypertension, obesity, and others, (4) to apy requires that the patient is in a quiescent
prevent, when possible, any abnormality associ- (intercritical) phase of their gout (inactive gouty
ated with the deposition of monosodium urate arthritis) and that a careful and thorough history
such as tophaceous deposits, nephrolithiasis, and has been taken, a complete physical examination
renal damage, and (5) to educate patients as to the has been performed, and appropriate laboratory
management parameters for which they have the analyses have been undertaken to evaluate the
primary responsibility (diet, alcohol use, etc.) possible underlying disorders that might be asso-
and to describe the potential side effects and ciated with gout. Individuals who overproduce
proper use of the prescribed drugs used for the uric acid and have genetic abnormalities in purine
treatment. metabolism (PRPP synthetase overactivity and
There are two important principles with hypoxanthine-guanine phosphoribosyltransferase
respect to these management plans that need deficiency) merit treatment with urate-lowering
emphasis. First, a distinct difference exists drugs. In addition, patients with uric acid nephro-
between those pharmacologic agents used in the lithiasis and uric acid-induced renal disease who
treatment of acute gout and those drugs used as require hypouricemic therapy should also be
uricosuric agents (e.g., probenecid) or xanthine treated with inhibitors of uric acid synthesis,
oxidase inhibitors (e.g., allopurinol). Drugs like rather than uricosuric agents, since the latter may
colchicine, NSAIDs, corticosteroids, and others increase the risks of nephrolithiasis. Certainly,
are anti-inflammatory agents with little or no patients with antimetabolite-sensitive tumors
therapeutic effect on the control of hyperurice- deserve pretreatment with allopurinol to prevent

D.S. Newcombe, Gout, 291


DOI 10.1007/978-1-4471-4264-5_9, © Springer-Verlag London 2013
292 9 Management of Hyperuricemia and Gout

urinary tract obstruction resulting from purine Table 9.1 Allopurinol treatment of asymptomatic hype-
breakdown and the precipitation of uric acid in ruricemia: indications
the renal tubules. Finally, patients with a solitary Heritable enzyme deficiencies
kidney also deserve a careful clinical evaluation Hypoxanthine-guanine phosphoribosyltransferase
if they suffer from acute or chronic gout since PRPP synthetase overactivity
their treatment regimens may need adjustment Glycogen storage disease (?)
based on the function of the solitary kidney. Renal calculus
Calcium oxalate
Any approach to identify the cause(s) of hype-
Uric acid
ruricemia must include a determination as to
Increased risk for renal calculus or tophaceous deposits
whether or not treatment should be instituted to
Urinary uric acid excretion of more than 1,000 g/day
reverse this biochemical derangement. Even
Serum uric acid greater than 8–10 mg/dl
though a solid scientific basis exists from which Single functioning kidney
rational therapeutic decisions can be made, phy- Prophylaxis for cytotoxic chemotherapy or
sicians frequently mismanage hyperuricemia. In radiotherapy
general, hyperuricemia should only be treated Secondary hyperuricemia
when urate-induced tissue damage has occurred Urinary uric acid greater than 1,000 mg/day
or is likely to occur from persistent, untreated Serum uric acid of 8 mg/dl or more
hyperuricemia. An approach based on mecha- Renal disease (?)
nisms of altered urate metabolism is presented
here. The significance of the two foregoing prin-
ciples concerning anti-inflammatory drugs and acid since these patients are at higher risk for the
hypouricemic agents requires careful consider- development of renal calculi and/or tophaceous
ation by attending physicians, and for this reason gout. Urinary uric acid excretion greater than
the discussion of such principles is expanded in 1,000 mg/day places these patients at risk for the
the following text. development of these complications, and
allopurinol significantly reduces the risk of such
complications (Table 9.1).
Management of Asymptomatic The issue of whether individuals with chronic
Hyperuricemia renal disease need treatment to reduce elevated
serum uric acid levels remains controversial.
In the case of non-drug-related hyperuricemia, Chronic renal disease is almost always associated
there are relatively few indications for the treat- with hyperuricemia, but the serum uric acid levels
ment of asymptomatic hyperuricemia (Table 9.1), are usually less than 10 mg/dl, and the renal dis-
and the clinical settings in which treatment is ease usually progresses very slowly. There is little
necessary are rare. A history of renal calculi, evidence to suggest that treatment with allopurinol
composed of either uric acid or calcium oxalate, slows the rate of kidney deterioration in these
is a clear indication for the introduction of patients [1]. Nonetheless, in patients with a soli-
allopurinol therapy to reduce the recurrence of tary functioning kidney and marked changes in
calculi. With respect to calcium oxalate calculi, uric acid metabolism (serum uric acid greater than
an increased uric acid excretion (>1 g/day) is 10 mg/dl and a urinary uric acid level of greater
generally accepted as the urinary uric acid level than 1,000 mg/day), hypouricemic therapy may
requiring the institution of allopurinol treatment be warranted. As one might suspect, there are no
since the recurrence of calcium oxalate stones hard and fast rules for these treatment regimens,
decreases with such a treatment regimen. and there are exceptions to these recommenda-
Certainly, allopurinol therapy is indicated in indi- tions. In addition, the levels of serum uric acid
viduals who express any of the heritable enzyme considered to be indicative of the need for treat-
deficiencies associated with abnormalities of ment have been selected in an arbitrary fashion
purine metabolism and overproduction of uric and may vary from physician to physician. Their
Management of Asymptomatic Hyperuricemia 293

selection is not based on experimentally derived been observed equally in patients both with no
data. renal impairment and those with decreased renal
Drug-induced hyperuricemia is probably the function. Chlorthalodine is a more potent antihy-
most common cause of elevated serum uric acid pertensive than hydrochlorothiazide, and it also
levels observed in patients if one excludes those causes elevations of the serum uric acid levels.
with chronic renal disease and an associated Patients with asymptomatic, drug-induced
diminished excretion of nitrogenous waste prod- hyperuricemia and an associated impairment of
ucts. Asymptomatic hyperuricemia induced by renal function present a special set of problems.
drugs, especially diuretics, rarely requires treat- First, it is difficult to determine what fraction of
ment to control the serum uric acid levels with the retained urate is related to renal dysfunction
several important exceptions that are discussed and what quantity can be attributed to a drug-in-
subsequently. The mechanisms by which diuretics duced alteration in the renal handling of urate.
cause hyperuricemia are variable. Volume con- Second, it is often difficult to distinguish between
traction secondary to diuresis may stimulate prox- urate-induced renal disease and non-urate-in-
imal renal tubular reabsorption of urate leading to duced nephropathy. Third, there are no carefully
elevated serum uric acid levels [2, 3]. Both furo- controlled studies of the effects of antihyperuri-
semide and diazoxide induce hyperlacticacidemia cemic drugs on the course of chronic renal
which reduces renal tubular excretion of urate disease.
resulting in an elevation of serum urate levels [4, Thus, whether the combination of diuretic-in-
5]. Furosemide and its congeners are bound to duced hyperuricemia and an underlying nonurate
plasma proteins and are not filtered to a significant nephropathy should be treated in a different man-
degree at the glomerulus. They are secreted into ner from diuretic-induced hyperuricemia in the
the renal tubular fluid of the proximal tubules by absence of renal disease remains an unresolved
an organic transport system, and probenecid, a problem. Controlled studies need to be done to
uricosuric agent, blocks furosemide activity by determine what effects the additional burden of
preventing its entry to the tubular fluid. diuretic-induced hyperuricemia might have on
Hypertension and gout frequently occur the kidneys with a compromised capacity to
together, and patients with gout are at higher risk excrete urate. In the absence of such data, the
for the development of hypertensive disease and case for treatment is made on the basis of an
its complications than the general population [6– accelerated loss of residual renal function from
8]. Since diuretics are often used for the treatment the hyperuricemic state and the concept that treat-
of hypertension, gouty episodes in patients with ment will retain the residual function. Those
established gout may be aggravated by diuretic opposing treatment argue that the potential risks
therapy for hypertension, and hypertensive of hypouricemic drugs far outweigh their benefits.
patients may experience a new episode of acute Such arguments are reinforced by long-term fol-
gout in association with the use of diuretics. low-up studies of diuretic-induced hyperuricemia
Recent studies have defined predictive factors for that show little evidence of progressive renal dys-
the onset of gout in hypertensive populations to function [10–12]. Nonetheless, some physicians
include an increased consumption of alcohol, consider treatment of hyperuricemia when the
obesity, male sex, and the use of loop diuretics serum urate levels are in the range of 8–10 mg/dl
[9]. Of interest is the fact that thiazide diuretics in patients without hemoconcentration. An iden-
are more commonly prescribed for hypertension tical therapeutic regimen should be utilized for
than loop diuretics, but the latter drugs are more patients with drug-induced hyperuricemia and an
frequently associated with gout in the hyperten- associated urate nephropathy. A serum urate level
sive patient than thiazides. Further, the associa- of 10–13 mg/dl or greater may justify hypourice-
tion of gout and the use of loop diuretics have mic drug treatment.
been shown to be related to reduced renal func- There are several special problems that arise in
tion since the increased frequency of gout has the management of drug-induced asymptomatic
294 9 Management of Hyperuricemia and Gout

hyperuricemia. Patients with only a single func- The use of cytotoxic agents in the treatment of
tioning kidney and drug-induced hyperuricemia neoplasia is also an important indication for
should probably be treated early and adequately allopurinol pretreatment in order to avoid the
with antihyperuricemic drugs. Any complicating occurrence of uric acid nephropathy.
clinical issue such as dehydration or urinary tract
infections in which the renal excretion of urate is
impaired should also be treated aggressively to Symptomatic Drug-Induced
prevent deterioration of renal function. Drug- Hyperuricemia
induced hyperuricemia in an asymptomatic
patient with a family history of severe gout prob- A general scheme for the management of drug-
ably should be treated promptly to reduce serum induced, symptomatic hyperuricemia includes
urate levels. Either a uricosuric agent or a xan- the identification of the offending drug, institu-
thine oxidase inhibitor would be an appropriate tion of a treatment regimen appropriate for gouty
drug in the absence of renal disease; a xanthine arthritis, if present, and the use of antihyperurice-
oxidase inhibitor is the drug of choice when renal mic agents to normalize the serum uric acid con-
dysfunction exists. If drug-induced serum urate centration when tophi, calculi, and/or gouty
levels are in the 9- to 10-mg/dl range or greater in nephropathy are present.
the male or female, a careful evaluation must be Drug-induced hyperuricemia is of course most
undertaken to detect underlying disorders that easily managed by substituting a drug that does
may be contributing to the altered uric acid not cause hyperuricemia. In certain cases, the
metabolism. offending drug is unnecessary and can be discon-
In asymptomatic hyperuricemic patients being tinued altogether. For example, some patients use
treated with nondiuretic-hyperuricemic drugs, aspirin, aspirin-containing drugs, or over-the-
the mechanism inducing hyperuricemia is more counter NSAIDs for nonspecific illnesses on a
complex. Salicylates at low doses (<1.2 g/day) chronic basis, and this habit can frequently be
cause urate retention by the kidney, but at higher interrupted by the physician. Acute gouty arthri-
doses (2–4 g/day), salicylates are uricosuric tis as a complication of drug-induced hyperurice-
[13–15]. Low-dose salicylates rarely cause incre- mia is managed by pharmacologic agents that
ments in serum urate concentrations greater than reduce the acute intra-articular inflammatory
1 mg/dl, but the use of salicylates in conjunction response. Tophi as a complication of drug-
with uricosuric agents abolishes the effect of the induced hyperuricemia are relatively rare (see
latter drug [16, 17]. the chapter on clinical gout and the subsection
Pyrazinamide-induced hyperuricemia is entitled cyclosporine), but their presence is an
unresponsive to uricosuric drugs, but high doses absolute indication for antihyperuricemic ther-
of salicylates (2–4 g/day) reverse the hyperuri- apy. In the absence of a simple, noninvasive
cemia caused by this drug [18, 19]. Nicotinic means for the detection of urate nephropathy and
acid in doses of 3–6 g/day causes hyperuricemia clinical data that clearly define the effects of
and inhibits the uricosuric effect of both hyperuricemia on renal function, recommenda-
sulfinpyrazone (Anturane) and iopanoic acid (a tions regarding the use of antihypertensive drugs
radiocontrast dye) [20]. For the latter reason, to prevent renal insufficiency remain empirical.
nicotinic acid-induced hyperuricemia is usually Most attention should be devoted to the treatment
treated with allopurinol when therapy is indi- of any underlying medical condition predispos-
cated. Rarely is treatment needed for such a con- ing to renal dysfunction. Thus, the aggressive
dition. Ethambutol (12–19 mg/kg body wt/day) management of hypertension, obesity, hyper-
is likely to increase serum uric acid concentra- lipidemia, and diabetes mellitus should take
tions, and elevated serum urate levels respond precedence over the use of antihyperuricemic
either to uricosuric agents or xanthine oxidase drugs. Nonetheless, when serum uric acid levels
inhibitors [21]. exceed 8–10 mg/dl, allopurinol is sometimes
Treatment of Acute Gouty Arthritis 295

used to prevent deterioration of renal function, acetazolamide (Diamox) also increases the solu-
although as noted above, there is little evidence bility of urate and allopurinol and its metabolites,
to support this assumption. Allopurinol dosage but alkalinization is usually not necessary in the
should at first be lowered when serum creatinine setting of antineoplastic treatment unless acute
levels are persistently elevated; such adjustments uric acid nephropathy occurs. Acetazolamide is
in dose prevent the retention of allopurinol and associated with a rebound acidosis, which is the
its oxidation product, oxypurinol [22, 23]. reason why preference is given to sodium bicar-
However, the dose of allopurinol should be bonate for urine alkalinization [25]. If acute uric
adjusted upward sufficient to bring the serum acid nephropathy does occur, one must first deter-
urate levels to <6 mg/dl in order to prevent mine whether the patient is anuric or not. In the
sodium urate crystal deposition. Both allopurinol anuric patient, hemodialysis is the most effective
and oxypurinol are relatively insoluble and may means of treatment [26]. Ureteral lavage with
be deposited in the kidney parenchyma [24]. alkaline solutions and surgery to remove the urate
sludge are indicated only if the ureters are
obstructed. Conservative management including
Hyperuricemia Associated with the measures to increase urine flow, solubilize urates,
Treatment of Neoplastic Disease and reduce uric acid production should be uti-
lized in patients who are able to maintain an ade-
Drug-induced hyperuricemia associated with an quate urine output. This conservative regimen
increased production of uric acid represents a necessitates careful monitoring of the volume of
common problem for patients undergoing treat- urine excreted, urinary pH, plasma electrolytes,
ment for neoplastic disorders. Acute uric acid arterial pH, and ketones when underlying chronic
nephropathy is a serious complication of antine- pulmonary disease, renal disease, or diabetes
oplastic treatment regimens resulting from cell mellitus are present. Inhibition of carbonic anhy-
destruction, purine catabolism, and the subse- drase with acetazolamide to increase bicarbonate
quent excretion of excessive quantities of urate excretion by the kidney is contraindicated when
by the kidney. For this reason, it is customary to arterial pH values are less than 7.2 or lactic acid
initiate allopurinol therapy in those patients who contributes to the acidosis. Since allopurinol
are scheduled to undergo chemotherapy or radio- blocks the degradation of 6-mercaptopurine, aza-
therapy for tumors. Allopurinol therapy, if possi- thioprine, and other purine analogs and increases
ble, should precede the use of antineoplastic the toxicity of these antimetabolites, their dose
therapy and should always be accompanied by should be reduced by one-third or more to avoid
fluid hydration to ensure adequate solubility of the toxic potentiation of the antimetabolite effects
uric acid as well as the pyrazolopyrimidines caused by allopurinol [27]. Further, careful moni-
(allopurinol and its metabolites). To induce a toring of the patient on such a dual regimen has to
rapid inhibition of xanthine oxidase, allopurinol be undertaken to diagnose bone marrow suppres-
should be administered at a dose of 300 mg/m2/ sion and secondary infections as a result of the
day in the first 48 h and followed by a dose of increased toxicity of these antimetabolites.
300 mg daily. Twice-a-day dosage is the usual
method for dividing the total dose; a practical
regimen for most antineoplastic treatment settings Treatment of Acute Gouty Arthritis
is to administer allopurinol 300 mg twice a day
for the initial period and follow this by a dose of In this section, we will discuss the pharmacology
150 mg twice a day. Hydration may be accom- and the therapeutic uses of agents used to manage
plished either by mouth or by the intravenous gout. The drugs used to treat the acute attacks of
route. Three to 5 l of fluid over the normal gout are discussed first. These include colchicine,
daily intake is an appropriate therapeutic goal. NSAIDs, glucocorticoids, and finally the emerg-
Urine alkalinization with sodium bicarbonate or ing use of IL-1 inhibitors. Next, the management
296 9 Management of Hyperuricemia and Gout

of the chronic phase of gout is considered, that is, H3CO


the treatment of hyperuricemia, the factor that H3CO
O
underlies the acute attack and the chronic OCH3
inflammation and tissue destruction which occur
O
in gout. The important underlying principle is H OCH3
that the pathologic consequences of gout arise N
from the supersaturation of the body fluids with H3C H
monosodium urate that leads to precipitation of
Fig. 9.1 The chemical structure of colchicine. This alka-
monosodium urate crystals in tissues. It cannot loid was originally obtained from the plant Colchicum
be emphasized too strongly that adequate preven- autumnale
tion of gout requires that the concentration of uric
acid in the blood, which is nearly all in the form
of urate monoanion, must be reduced below the in nearly all patients. More recently, a study has
saturation level of 6.8 mg/dl (357 meq/l) in order been published that compares the standard colchi-
to prevent the precipitation of sodium urate in tis- cine schedule with a lower dose consisting of
sues. Effective treatment of gout can only be 1.2 mg followed by 0.6 mg 1 h later [40].
achieved when this condition is met. From a prac- Observations at 24 h after initiating these treat-
tical point of view, physicians should aim to ments demonstrated that the therapeutic responses
reduce the serum levels of uric acid to less than of both groups of patients were equivalent. This
6.0 mg/dl. Failure to adhere to this principle is study leads to the conclusion that the high-dose
responsible for the widespread failure of the schedule is inappropriate because of its toxicity
management of gout in clinical practice, both by and that similar responses are obtained on the
the specialist and the generalist. lower-dose schedule with significantly fewer
toxic side effects. However, responses were only
reported after the first 24 h of treatment, and it
Colchicine Use in Acute has not been shown that the responses to high-
and Chronic Gout and low-dose therapy are similar throughout the
entire duration of the acute attack, and more study
Treatment Schedules of this issue is needed (Fig. 9.1).
Even low doses of colchicine may be associ-
Colchicine is effective in the treatment of acute ated with significant gastrointestinal toxicity in
gouty arthritis and can act prophylactically to some patients, and alternative forms of treatment
reduce the number of recurrent episodes of gouty may be needed, as discussed below. In addition,
arthritis. In the past, responses to this drug have patients with gout and an associated serious intes-
been used as a diagnostic test for gout, but the tinal disorder, such as Crohn’s disease, should be
therapeutic response of pseudogout and other treated with alternative drugs to prevent these
articular disorders (sarcoidosis, hydroxyapatite gastrointestinal side effects. Acute gouty arthritis
deposition, and familial Mediterranean fever) to may become less responsive to colchicine after
colchicine treatment has decreased the validity of joint symptoms have been present for 48–72 h, as
this pharmacologic test as a diagnostic tool is the case with other forms of treatment.
[28–39]. The standard dosage schedule for the It should be kept in mind that when an acute
treatment of acute gout with colchicine has been arthritis occurs, even in a patient with a history of
to administer one 0.5 or 0.6 mg tablet by mouth gout, alternative diagnoses to gout may be pres-
every hour to a maximum of 6–8 mg in adults. ent, especially septic arthritis. Therefore, aspira-
However, administration of colchicine according tion of the joint is often indicated, both to
to this schedule produces significant gastrointes- document the diagnosis of gout by crystal
tinal symptoms of nausea, vomiting, and diarrhea identification and culture to exclude infection.
Colchicine Use in Acute and Chronic Gout 297

Mechanism of Action Table 9.2 Pharmacologic properties of colchicine


Colchicine does not alter serum urate levels
Colchicine is a lipid soluble drug that crosses cell Colchicine does not alter urinary uric acid levels
membranes easily; its structure and pharmaco- Colchicine binds to tubulin in microtubules
logic properties are shown in Fig. 9.1 and Colchicine (in vitro) inhibits chemotaxis and neutrophil
Table 9.2. Any discussion of the mechanism of adhesion
action of colchicine must correlate plasma and Colchicine (in vitro) impairs lysosome degranulation
and cell motility
tissue levels of the drug attained in clinical usage
Colchicine blocks the release of a chemotactic
with concentrations of colchicine shown to have substance
pharmacologic activity against specific cellular Colchicine under certain conditions may impair
functions such as inflammatory cell chemotaxis neutrophil phagocytosis
and chemotactic peptide generation [41–44]. To Colchicine does not bind to plasma proteins
date, only these two functions in addition to
changes in neutrophil tyrosine phosphorylation
have been shown to be impaired by colchicine Table 9.3 Colchicine concentrations required for the
concentrations in the range found in the plasma inhibition of specific cell functions
after oral or intravenous dosages [41–45]. Other Function I Inhibitory concentration
cellular functions proposed as mechanisms by Leukocyte locomotion 40,000 ug % (10−3 M)
which colchicine is effective in acute gouty Lysosomal degranulation 100–500 ug % (10−5 M)
arthritis require higher concentrations than those Kinin release 400 ug % (10−5 M)
attainable in vivo with pharmacologic doses of Cell metabolism during 50–100 ug %
phagocytosis (2 × 10−6 M)
the drug (Table 9.3) [46–52]. Colchicine clearly
Release of chemotactic 0.4–40 ug %
diminishes the inflammatory response to urate peptide (10−6–10−8 M)
crystals in vivo [52]. In vitro, the drug also binds Chemotaxis 0.4–4 ug % (10−7–10−8 M)
avidly to neutrophil microtubules and inhibits
stimulus-induced lysosomal enzyme release
[53–55]. Since both the release of chemotactic required to demonstrate these effects may be
peptides and chemotaxis are proposed as the pri- above those achieved in serum, leukocytes may
mary pharmacologic targets of colchicine, stud- concentrate colchicine intracellularly, allowing
ies of the mechanism of action of colchicine have these effects to occur. Since urate crystals are
been focused on these parameters. Investigations now thought to initiate acute gout by activating
have shown that a glycoprotein with chemotactic the NLRP inflammasome, colchicine may either
properties for neutrophils and mononuclear leu- impair the uptake of urate crystals by the
kocytes is synthesized by neutrophils exposed to inflammatory cells, neutrophils and macrophages,
monosodium urate crystals, and colchicine inhib- and synovial cells. Whether there are direct
its the production and release of this glycoprotein effects of colchicine on the inflammasome itself
[56]. Neutrophils treated with colchicine (10−5 M) remains unclear (Tables 9.2 and 9.3).
also show a significant decrease in their capacity Although polymorphonuclear neutrophils and
to synthesize and release leukotrienes B4 and inflammatory mediators such as lysosomal
other lipoxygenase products such as hydroxyei- enzymes, oxygen-derived free radicals, interleu-
cosatetraenoic acids [57]. Since LTB4 is a potent kin-1, crystal-induced chemotactic factor, and
neutrophil chemotaxin, some authors have sug- 5-lipoxygenase products contribute to the acute
gested that colchicine alters the production of intrasynovial response to uric acid and calcium
leukotrienes B4 in the intact neutrophil which pyrophosphate dehydrate (CPPD) crystals, the
subsequently decreases the chemotaxis of neutro- actual mechanism by which colchicine alters
phils to the site of monosodium urate crystals acute crystal-induced arthritis still remains
[57]. Although the concentrations of colchicine incompletely understood.
298 9 Management of Hyperuricemia and Gout

It is also noteworthy that studies over the past tion of microtubule assembly and for the promo-
few years have led to the concept that crystal- tion of concanavalin cap formation in human
induced arthropathies, gout and pseudogout, neutrophils [65–70]. Some other anti-
belong to the category of disorders called inflammatory drugs with some similarities to the
autoinflammatory diseases. These diseases differ structure of colchicine, the NSAIDs, do not affect
from autoimmune diseases in that inflammation crystal-induced tyrosine phosphorylation. This
employs the innate immune system, as opposed crystal-induced reaction is presently somewhat
to the adaptive immune system. Adaptive immu- specific for CPPD and monosodium urate (MSU)
nity requires antibody and T cell-specific activ- crystals since other particulates including unop-
ity such as that seen in autoimmune diseases, sonized zymosan, latex beads, heat-killed staphy-
systemic lupus erythematosus and rheumatoid lococcus, and silica crystals cause qualitatively
arthritis, whereas the innate immune system is different responses. It appears from these studies
independent of these mechanisms. Over the past that MSU and CPPD crystals activate kinases or
several years, an increasing number of phosphatases associated with microtubules or
inflammatory diseases are now considered to be tubulin causing a characteristic tyrosine phos-
disorders which employ the innate immune sys- phorylation that is modulated by colchicine. As
tem. It is also intriguing that colchicine is an discussed elsewhere, a major mechanism for the
effective therapy for autoinflammatory diseases induction of inflammation by sodium urate and
that are based on unrelated principles. Colchicine other crystalline materials, calcium pyrophos-
is effective in the treatment of gout and pseudog- phate dehydrate, and silica is now thought to
out, both of which are induced by microcrystals, reside in the activation of the inflammasome
as well as in familial Mediterranean fever, a dis- NLRP3.
ease related to genetic defects in the protein
pyrin, which induces recurrent bouts of fever,
arthritis, and serositis. The explanation for the Pharmacokinetics of Colchicine
effectiveness of colchicine in these apparently
disparate diseases remains unexplained at this In healthy subjects, colchicine is rapidly absorbed
time [58]. from the gastrointestinal tract after oral adminis-
Zero time plasma colchicine level is 1.79 ± 0.7 tration and reaches peak plasma concentrations
ug % (4.5 × 10−8 M) after 2 mg of colchicine is within one-half to two hours. Absorption in the
administered intravenously. Highest mean colchi- gut presumably takes place in the jejunum and
cine level is 0.323 ± 0.7 ug % (7 × 10−9 M) after ileum since large doses of colchicine alter func-
1 mg of colchicine is administered orally. tions known to be carried out by these segments
Recent investigations have demonstrated that of the small bowel such as vitamin B12 absorption
exposure of neutrophils to monosodium urate or [71, 72]. The pharmacokinetic properties of
CPPD crystals causes a rapid increase in the cyto- colchicine include a short half-life, a large appar-
plasmic concentration of free calcium, the forma- ent volume of distribution (2.19 ± 0.8 l/kg body
tion of 1,4,5-triphosphate, the activation of weight), and little or no plasma protein binding
phosphatidylcholine-specific phospholipase D, (Table 9.2) [53]. Urinary and pulmonary excre-
and the tyrosine phosphorylation of a 70-kD pro- tions do not explain the short half-life of colchi-
tein [59–62]. Colchicine specifically inhibits this cine, and the large volume of distribution, which
phosphorylation reaction at 10−7–10−6 M concen- is greater than the extracellular volume, suggests
trations that are similar to colchicine concentra- a high affinity of the drug for tissues. In fact, data
tions observed in patients receiving therapeutic show that polymorphonuclear leukocytes con-
doses of the drug [63, 64]. Interestingly, the centrate colchicine, and these cells are also an
colchicine concentrations and the reaction times essential element of the inflammatory response to
for the inhibition of tyrosine phosphorylation are monosodium urate crystals [63]. After 3 mg of
almost identical to those required for the inhibi- colchicine intravenously, healthy volunteers
Colchicine Use in Acute and Chronic Gout 299

Table 9.4 Colchicine kinetics and metabolism Table 9.5 Colchicine toxicity
Oral colchicine is absorbed via the gastrointestinal tract Full therapeutic doses of colchicine are toxic to the GI
Average peak plasma levels are 0.32 ug/dl after oral tract in 80 % of its recipients
colchicine (1 mg) Crampy abdominal pain, hyperperistalsis, diarrhea,
Peak plasma concentrations are attained in 0.5–2 h nausea, and vomiting are common side effects
Intravenous colchicine has a half-life of 20 min Chronic colchicine use may lead to bone marrow
Intravenous colchicine (3 mg) reaches a peak level in depression, peripheral neuritis, and hair loss
white blood cells of 43 ug/dl after 10 min Other toxic effects of chronic use include amenorrhea,
Oral colchicine (1 mg) gives a serum colchicine level dysmenorrhea, oligospermia, and azoospermia
of 0.03–0.24 ug/dl Overdoses may lead to hemorrhagic gastroenteritis,
Colchicine is excreted in the bile, feces, and urine metabolic acidosis, electrolyte disturbances, renal
failure, shock, Gram-negative septicemia,
hepatocellular failure, and CNS dysfunctions

manifest a peak concentration of colchicine of 43


ug/dl at 10 min, and significant levels of colchi- Colchicine Toxicity
cine are still measurable in leukocytes (11 ug/dl)
from these individuals after 72 h [63]. These leu- Life-threatening toxic reactions of the central
kocyte concentrations of colchicine exceed nervous system, bone marrow, muscle, and liver
plasma concentrations and are comparable to the are the most likely to occur in patients with renal
drug concentrations necessary to inhibit those or hepatic dysfunction who are taking colchicine
specific leukocyte functions accounting for the [79–82]. Excretion of colchicine and its metabo-
drug’s mechanism of action in acute gout. lites is impaired with renal and hepatic disease,
Colchicine is also released slowly from rat peri- and the drug should be avoided in these clinical
toneal mast cells, rat peritoneal macrophages, settings [83, 84]. Alopecia, bone marrow sup-
and human synovial cells [73–76]. Its primary pression, and transient mental confusion have
excretory route in the human is the kidney, but rarely been reported during colchicine therapy;
the lung accounts for an unknown fraction of usually they are associated with toxic responses
excreted drug [52, 77]. The kinetics and metabo- to the drug [52, 79, 85, 86]. Although colchicine-
lism of colchicine are shown in Table 9.4. The induced myopathy has been described as a result
recent observations concerning the inhibition of of drug overdoses, recent clinical data suggest
tyrosine phosphorylation by colchicine may char- that colchicine myoneuropathy may be a rela-
acterize the biochemical basis for the activity of tively common complication of gouty patients
this ancient drug [62]. In patients with chronic with diminished renal function [86–88]. Such
liver disease, colchicine levels in the plasma are myoneuropathies may be observed when modest
measured at higher initial levels than normal, but doses (1–1.5 mg) of colchicine are being used
the drug disappears from the plasma more rap- [88] (Table 9.5).
idly than normal [64]. Further, in those patients The clinical findings of this myopathy include
without associated renal dysfunction, significantly proximal muscle weakness that at times is associ-
increased quantities of colchicine are excreted in ated with a mild peripheral neuropathy [88].
the urine. In patients with severe renal disease, Laboratory findings demonstrate an increase in
the rate of plasma colchicine disappearance is serum creatinine (>1.6 mg/dl), significant eleva-
slower than normal, and the quantity of drug tions of creatine kinase activity (>150 I.U./l), and
excreted by the kidney is markedly decreased an abnormal electromyographic pattern [88].
[64, 78]. Surprisingly, patients with gout and Muscle biopsy specimens show a distinctive lys-
moderate renal dysfunction have patterns of osomal, vacuolar myopathy with vacuoles
plasma colchicine disappearance similar to identified as lysosomal by their acid phosphatase
patients with severe renal disease but excrete nor- reactivity [88]. Peripheral nerve biopsies show a
mal quantities of the drug in their urine [64] loss of myelinated axons [88]. Reduction or dis-
(Table 9.4). continuance of colchicine results in a dramatic
300 9 Management of Hyperuricemia and Gout

Table 9.6 Colchicine contraindications Table 9.7 Relative contraindications to colchicine therapy
Inflammatory bowel disease Neutropenia
Pregnancy Renal failure
Nursing mothers Hepatocellular disease
Hepatic dysfunction Gastrointestinal disease (mucosal lesions)
Decreased renal function Agranulocytosis
Aplastic anemia
Infertility (azoospermia)
reversal of the muscular weakness that correlates Myopathy
Elderly patients with cardiovascular, renal, or
with a return to normal of the elevated creatine
gastrointestinal diseases
kinase activity. The toxic responses to colchicine
are summarized in Table 9.5.
A recent case report raises concerns regarding if the creatinine clearance is 30 ml/min and avoided
the use of colchicine in renal transplant recipients completely when the estimated glomerular filtration
who are being treated with immunosuppressive rate is less than 30 ml/min. Colchicine should not
drugs like cyclosporine and azathioprine [89]. In be used in the presence of other drugs which are
this reported patient, a severe neuromyopathy metabolized by the cytochrome P450 3A4 path-
developed in association with markedly elevated way. These include clarithromycin, cyclosporine,
creatine kinase (phosphokinase) activity (600 certain calcium channel blockers, and ketocon-
I.U./l), aldolase activity (45 I.U./l), SGOT activ- azole and related drugs.
ity (929 I.U./l), and SGPT activity (288 I.U./l). Intravenous colchicine is rapid and an effec-
An EMG showed prominent fibrillations, posi- tive treatment of acute gout that avoids the gas-
tive sharp waves, pronounced high-frequency trointestinal toxicity of oral colchicine, but
discharges, early recruitment, and myopathic serious toxic reactions and death have been
units. Nerve conduction velocity tests were con- reported in association with intravenous colchi-
sistent with a sensorimotor polyneuropathy, and cine therapy [83]. Toxic reactions to intravenous
muscle biopsy showed the pathognomic features colchicine are also observed more frequently in
of a colchicine-induced neuromyositis. The patients with renal and/or hepatic disease and in
mechanisms by which this patient developed those patients who have been receiving oral
colchicine myopathy are unknown, but the colchicine as maintenance therapy. Local reac-
authors speculate on the possible interactions tions to intravenous colchicine include skin
between cyclosporine and colchicine as the prime necrosis and phlebitis at the site of local injec-
mediators responsible for the neuromyopathy tion, and median nerve neuritis has been reported
and the associated hepatic dysfunctions observed secondary to the extravasation of colchicine into
in this patient. Another unusual toxic effect of soft tissues [91]. Because of its toxic potential,
colchicine has been described in a patient receiv- intravenous colchicine has fallen into disuse, and
ing 1 mg of colchicine daily in conjunction with intravenous preparations of colchicine are no lon-
cyclosporine-induced gout [90]. This patient ger available in the United States.
experienced a reversible azoospermia. As previously used, intravenous colchicine was
The generally accepted contraindications to usually administered in 20 ml of saline (0.9 %)
colchicine usage are shown in Tables 9.6 and 9.7. containing 2 mg of the drug. Repeat doses of 1 mg
Tests using d-xylose as a measure of intestinal may be given every 6 h to a total dose of 4 mg. We
absorption may be inaccurate in patients taking believe that intravenous colchicine should rarely,
colchicine since the drug reduces xylose absorp- if ever, be used for the management of gout. When
tion [71] (Tables 9.6 and 9.7). Colchicine should be intravenous colchicine therapy is used, the patient
used in reduced doses in the presence of even mild should be monitored in the hospital to avoid drug
to moderately reduced renal function and generally overdoses, and elderly patients should never
should be used with caution and in reduced doses receive more than 2 mg intravenously for a single
Nonsteroidal Anti-inflammatory Drugs (NSAIDs) 301

attack of gout. Extreme care should be exercised cyclooxygenase accounts for the therapeutic and
to avoid intravenous colchicine in patients already the toxic effects of these agents. The NSAIDs are
receiving maintenance colchicine. As noted previ- in wide use as analgesic and anti-inflammatory
ously, colchicine is excreted slowly, and cells that drugs for a large number of inflammatory condi-
have been exposed to colchicine are more sensi- tions. These drugs are also highly effective in
tive to its effects since the drug persists in cells for relieving the severe pain and swelling of acute
long time periods. gouty arthritis. We will begin with a general dis-
The relative contraindications to colchicine cussion of the mechanism of action and clinical
treatment are shown in Table 9.6. uses of the NSAIDs as a class and then discuss
In summary, oral colchicine may be used in low two drugs, indomethacin and naproxen, in detail
doses to treat acute gouty arthritis without serious since these two agents are most commonly used
gastrointestinal toxicity. Colchicine may also be for treatment of acute gout, and controlled clini-
used in low dosage, 0.5 mg once or twice a day, to cal trials have demonstrated their effectiveness in
protect patients undergoing surgical procedures gout. However, comparison studies have shown
from precipitating a postsurgical episode of gout. that several other NSAIDs are effective in acute
It may be used in low doses also for acute gouty gout.
episodes [40]. This colchicine dosage regimen has Eicosanoids are the products of several differ-
been used in a randomized, placebo-controlled ent enzymes. They are derived from 20-carbon
trial where the low-dose colchicine group utilized polyunsaturated fatty acids, primarily arachi-
1.8 mg of colchicine over 1 h (1.2 mg of colchi- donic acid. These fatty acids are major constitu-
cine initially and 0.6 mg 1 h later) to treat acute ents of phospholipids in all cell membranes. The
gouty attacks within the first 12 h of its onset. This prostaglandins and thromboxanes are the prod-
experimental group was compared with a high- ucts of the action of cyclooxygenases on arachi-
dose colchicine group (4.8 mg of colchicine over 6 donic acid, and the cyclooxygenases are the
h). The high-dose group manifested a significant enzymes that are inhibited by the NSAIDs
degree of diarrhea and vomiting (77 and 17 %, (Fig. 9.2). There are two enzymes (cyclooxyge-
respectively), whereas in the low-dose group, diar- nase-1 [COX-1] and cyclooxygenase-2 [COX-2])
rhea occurred in only 23 % and no one in the group responsible for the conversion of arachidonic
vomited. Thus, low-dose colchicine may be used acid to the prostaglandin endoperoxides (PGG2
to treat acute gouty episodes, but there remains the and PGH2) The eicosanoid products of COX-1
issue of diarrhea in about one-quarter of the recipi- are generally considered to mediate protective
ents of the drug. Such data do not resolve the issue functions of prostaglandins, and COX-2 produces
of colchicine’s toxicity to the gastrointestinal tract mediators of inflammatory reactions. This has led
and the selection of patients to avoid such compli- to the hypothesis that specific inhibitors of COX-2
cations. It also has some serious drug-drug inter- would be effective inhibitors of inflammation,
actions, especially with cyclosporine usage, that without producing toxicity by inhibiting the
make it a dangerous choice for kidney transplant COX-1 enzyme that mediates the protective func-
patients with gout. tions of prostaglandins and thromboxanes.
Subsequently, specific cyclooxygenase-2 inhibi-
tors were developed that block the elevated levels
Nonsteroidal Anti-inflammatory of prostaglandins in inflammatory states [92–97].
Drugs (NSAIDs) The significance of COX-2 to inflammatory
responses lays in the fact that exposure of cells to
The second category of drugs that is useful for inflammatory stimuli causes an increased expres-
the treatment of acute gouty arthritis is that of sion of cyclooxygenase-2 [98–100]. It has also
NSAIDs. These drugs have the common mecha- been shown that COX-1 is involved in the release
nism of action of inhibiting eicosanoid synthesis of prostaglandins for physiological processes and
by the cyclooxygenase enzymes. Inhibition of is expressed in many different tissues, whereas
302 9 Management of Hyperuricemia and Gout

Membrane phospholipids

Phospholipase A2
Diverse physical,chemical,
inflammatory, and mitogenic stimuli

Arachidonic Coxibs
acid

Prostaglandin G2 COX Prostaglandin G2


Prostaglandin G/H Prostaglandin G/H
synthase 1 synthase 2
(cyclooxygenase-1) Prostaglandin H2 HOX (cyclooxygenase-2)
Prostaglandin H2

Tissue-specific isomerases

Prostanoids Prostacyclin Thromboxane A2 Prostaglandin D2 Prostaglandin E2 Prostaglandin F2α

Receptors IP TPa, TPβ DP1, DP2 EP1, EP2, EP3, EP4 FPα, FPβ

Platelets,
Brain, kidney, Uterus, airways,
Endothelium, vascular smooth- Mast cells,
vascular smooth- vascular smooth-
kidney, muscle cells, brain,
muscle cells, muscle cells,
platelets, brain macrophages, airways
platelets eye
kidney

Fig. 9.2 Production and actions of prostaglandins and inhibit cyclooxygenase-2. Prostaglandin H2 is converted
thromboxane. Arachidonic acid, a 20-carbon fatty acid by tissue-specific isomerases to multiple prostanoids.
containing four double bonds, is liberated from the sn2 These bioactive lipids activate specific cell-membrane
position in membrane phospholipids by phospholipase receptors of the superfamily of G-protein–coupled recep-
A2, which is activated by diverse stimuli. Arachidonic tors. Some of the tissues in which individual prostanoids
acid is converted by cytosolic prostaglandin G/H syn- exert prominent effects are indicated. IP denotes prostacy-
thases, which have both cyclooxygenase (COX) and clin receptor, TP thromboxane receptor, DP prostaglandin
hydroperoxidase (HOX) activity, to the unstable interme- D2 receptor, EP prostaglandin E2 receptor, FP prostaglan-
diate prostaglandin H2. The synthases are colloquially din F2a receptor (Reprinted from N Engl J Med with
termed cyclooxygenases and exist in two forms, cycloox- permission)
ygenase-1 and cyclooxygenase-2. Coxibs selectively

COX-2 is induced by inflammatory mediators competitive inhibitors of cyclooxygenases. All


acting on cells involved in inflammatory responses these nonselective NSAIDs inhibit the physio-
(neutrophils, macrophages, endothelial cells, and logical functions regulated by COX-1, and this
fibroblasts) [101]. Furthermore, glucocorticoids, action leads to an increase in gastrointestinal
potent anti-inflammatory agents, block the intolerance including peptic ulcer disease and
induction of COX-2 but have no effect on COX-1 bleeding as well as the possibility of renal
[102, 103]. Most of the available nonsteroidal insufficiency [104, 105].
anti-inflammatory agents are nonselective with Studies have compared the inhibitory effect of
respect to their inhibition of cyclooxygenases. nonselective NSAIDs on COX-1 and COX-2
Ibuprofen (Motrin), naproxen (Naprosyn), and activities [237, 238]. Ideally, one would expect
piroxicam (Feldene) are competitive inhibitors of the most effective and least toxic anti-
cyclooxygenases, and indomethacin (Indocin) inflammatory agents to be the most potent inhibi-
and diclofenac (Voltaren) are time-dependent, tors of COX-2 with the least activity against
Nonsteroidal Anti-inflammatory Drugs (NSAIDs) 303

COX-1. In contrast, the most potent inhibitors of COX-2 inhibitors remain but do have a tendency
COX-1 activity would be the most likely to cause to cause cardiovascular toxicity. Studies have
ulcerations and bleeding from the gastrointestinal also shown that most of the nonselective COX
tract. When S-ibuprofen, meclofenamate, and inhibitors also have risks of cardiovascular toxic-
flurbiprofen were investigated in vitro for their ity, although the mechanisms are not known. The
preference as an inhibitor of COX-2 as compared inhibition on both classes of cyclooxygenases or
to COX-1, it was found that S-ibuprofen has a other secondary effects such as renal function
twofold preference for COX-2, Meclofen a sev- and blood pressure could be contributing factors.
enfold preference, and flurbiprofen was in Regardless of mechanisms, there are differences
between these levels of preference. When sulin- in the relative risks of cardiovascular diseases
dac sulfide, indomethacin, and piroxicam were produced by individual NSAIDs. For example,
compared for their inhibitory capacity against diclofenac has one of the higher risks compared
these same enzymes, it was found that these drugs to other NSAIDs. Naproxen has been found to
were 10–30 times more inhibitory of COX-1 than have the least association with cardiovascular
COX-2. The only drug investigated in this study risk. The reason for the apparent variation in car-
that was a more potent inhibitor of COX-2 activ- diovascular risk with different NSAIDs is
ity as compared to COX-1 was nabumetone unknown, but it may make agents such as
(Relafen) that was shown to be a sevenfold more naproxen a good choice of NSAIDs from this
potent inhibitor of COX-2 as compared to COX-1 point of view [109, 110].
[237]. However, the selectivity of different NSAIDs like sulindac (Clinoril), naproxen
NSAIDs for COX-1 and COX-2 may vary some- (Naprosyn), fenoprofen (Nalfon), ibuprofen
what depending on the assay system. For exam- (Motrin), ketoprofen, and fenamates (flufenamic,
ple, in whole blood studies, indomethacin, mefenamic, and meclofenamic acids) have also
ibuprofen, and naproxen had similar activities been used in the treatment of gout, but none
against COX-1 and COX-2 [106–108, 111]. appeared to be more effective than indomethacin
Prior to the development of the highly selec- [112–118]. Phenylbutazone is no longer avail-
tive COX-2 inhibitors, some investigators postu- able for use due to its bone marrow toxicity, but
lated that these drugs might promote it was a very effective drug for acute gout, and its
cardiovascular toxicity. It was suggested that structure may provide a means of designing less
intravascular clotting was in part prevented by toxic compounds for the treatment of gout.
the production of prostacyclin (PGI2) by endothe- Sulindac is similar to indomethacin in structure,
lial cells. PGI2 causes vasodilatation and inhibits whereas naproxen is related to arylpropionic
platelet aggregation, thereby helping to maintain acid NSAIDs, and fenamates are
vascular flow, and that this counteracted the N-phenylanthranilic acid-substituted NSAIDs.
effects of thromboxane B2 by platelets, which The pharmacokinetics and metabolism of some
both promotes platelet aggregation and vasocon- nonsteroidal anti-inflammatory agents are shown
striction. It had been suggested that selective in Table 9.8. For the most part, these drugs have
inhibition of COX-2 from the endothelium would side effects in the human similar to those
leave the vasoconstrictive effects of thrombox- described for indomethacin, but a few distinctive
ane produced by COX-1 in platelets unopposed toxicities are emphasized subsequently for each
and predispose to vascular occlusion that could of these drugs. Their pharmacokinetic properties
cause coronary occlusion and strokes. Subsequent are also similar with the exception of sulindac
studies have shown that this hypothesis was at and naproxen which have longer half-lives (8–17
least to some extent correct. Numerous studies h) than the other agents which have much shorter
have shown that COX-2-specific inhibitors are half-lives (1–3 h) [118–120]. In addition to its
associated with untoward cardiovascular events, NSAID-like toxicities, sulindac should not be
and one of these, rofecoxib, has been removed used in patients who cannot tolerate prolonged
from the market because of this toxicity. Other bleeding times.
304 9 Management of Hyperuricemia and Gout

Table 9.8 Nonsteroidal anti-inflammatory agents: pharmacokinetics and metabolism


Colchicine Sulindac Indocin Naproxen Motrin
GI absorption (%) Small bowel 88 Rapid Complete/rapid Rapid
Plasma protein binding (%) Not detected 93 98–99 99 99
Peak plasma levels (h) 0.5–2a 1–2 0.75b 2–4 1–2
1–2c
Half-life (h) 0.33d Biphasice 8 and 18 Biphasicf 1 and 9 17 2.5
Metabolism Renal (hepatic) Hepatic (renal) Hepatic (renal) Hepatic Hepatic
Volume of distribution (L/kg) 2.2 – 0.3–1.6 0.09 0.15
Excretory routes Urine/lung Urine > stool Urine > stool Urine Urine
Data for fenoprofen and ketoprofen are comparable to ibuprofen (Motrin)
a
After 1 mg of colchicine
b
Peak plasma levels depend on food intake but are rapid in the fasting state
c
Peak plasma levels are delayed in the fed state
d
After 2 mg of IV colchicine
e
Sulindac undergoes extensive enterohepatic recirculation
f
Indomethacin undergoes partial enterohepatic recirculation

Toxicity of NSAIDs lndomethcin


CH3O CH2CO2H
The potential toxicities of the NSAIDs are impor-
tant to recognize since these are such a widely N CH3
used group of drugs, especially since they are
readily available as nonprescription drugs in C O
many cases. The most frequent toxicity of these
drugs is from their effects on the upper GI tract.
The nonselective NSAIDs inhibit the enzyme
cyclooxygenase 1 in the gastric mucosa that pro- CI
duces PGE2. PGE2 helps to maintain the protec- Sulindac
tive gastric mucosal barrier and inhibits gastric
acid secretion, thus reducing the tendency for CH2CO2H
F
gastric mucosal erosions and their complications
of bleeding, perforation, and obstruction. One of
the major benefits of the selective COX-2 inhibi- CH3
tors is their sparing of COX-1, which reduces the CH3
development of upper GI toxicity. A second
major toxic effect of the NSAIDs is their renal O
effects which can lead to renal failure and hyper-
CH3S
tension, and this toxicity is shared by both classes
of COX inhibitors. Fig. 9.3 The structures of indomethacin and sulindac.
Physicians should become familiar with the Indomethacin is an indole acetic acid with a pKa of 4.5. It
use of one or two NSAIDs useful for the manage- is stable in neutral or acidic environments. Sulindac is
ment of acute gout and use them routinely for prodrug; the sulfide metabolite is the active moiety. As
can be seen, sulindac is an indene acetic acid derivative
acute gouty arthritis. The structure and properties that is chemically related to indomethacin
of naproxen and other commonly used anti-
inflammatory drugs are shown in Tables 9.13,
9.14, and 9.15 and Fig. 9.3. ment of acute gout, the fact that monosodium urate
Even though there are few published reports of crystals induce COX-2 expression in monocytes
the use of COX-2-specific inhibitors in the treat- with the resultant production of proinflammatory
Nonsteroidal Anti-inflammatory Drugs (NSAIDs) 305

prostaglandins provides a rationale for their use in decreased effective intravascular volume, such as
gout [121]. The two most commonly used COX-2 in congestive heart failure [146–150]. In animal
inhibitors, celecoxib (Celebrex) and, before its studies, there is evidence that angiotensin II inhib-
removal from the market, rofecoxib (Vioxx), have itors upregulate renal cortical COX-2 in volume-
proven to be effective anti-inflammatory agents depleted states [151]. These early investigations
for the treatment of rheumatoid arthritis and of COX-2-specific inhibitors indicate that caution
osteoarthritis, and these data suggest that they should be exercised with the use of these drugs in
might be very effective in other joint diseases as those patients with underlying renal disease,
well. Another premise for the use of COX-2 inhib- patients who are elderly, those with volume-
itors is that these specific inhibitors reduce depleted states, and those on low-sodium diets. A
inflammation but do not alter the protective prosta- few studies have also emphasized additional
glandins in the stomach and kidney produced by advantages of the COX-2 inhibitors including the
COX-1 activity [122]. However, investigations absence of effects on platelet aggregation, throm-
have shown that COX-2-specific inhibitors spare boxane B2 production, and methotrexate renal
the COX-1 prostaglandin-dependent mucosal pro- clearance and pharmacokinetics [152, 153].
tective mechanisms active in the gastrointestinal Further, studies in rats have shown that blocking
tract but do not preserve all the renal functions mineralocorticoid receptors with spironolactone
controlled by prostaglandins [123]. Many investi- causes an upregulation of renal cortex COX-2
gations have now confirmed that COX-2 inhibitors [154]. Recent data have shown that COX-2 inhib-
significantly reduce the morbidity and mortality itors may cause adverse cardiovascular events,
from the gastrointestinal effects caused by the use and these toxic parameters are reviewed at the end
of conventional NSAIDs [124–130]. of this section.
The relative lack of upper GI toxicity seen
with the selective COX-2 inhibitors has led to
their use in some patients receiving anticoagu- Other Toxicities of NSAIDs
lants, such as warfarin and heparin. Nonselective
COX inhibitors are hazardous in patients receiv- Magnesium aluminum hydroxide does not affect
ing anticoagulants because of a high risk of upper ketoprofen absorption [155, 156]. Thus, these
GI bleeding. Nonetheless, patients receiving anti- antacids are favored if one is seeking maximal
coagulants and selective COX-2 inhibitors should absorption of this class of NSAID along with
be monitored carefully. While most experience is reduction in gastrointestinal disturbances. At the
related to warfarin anticoagulation, it is logical to doses of ibuprofen and ketoprofen used in gout,
assume that the same precautions will hold for warfarin is displaced from its binding site on
the newer anticoagulants, antithrombin, and anti- plasma proteins resulting in increased anticoagu-
factor X agents that are coming into wide use. lant effects [119, 157]. Similar effects are observed
With respect to the effects of COX-2-specific with naproxen [158–160, 163–165]. In addition,
inhibitors on the kidney, the results of many stud- patients taking anticoagulants are at increased
ies have determined that COX-2 inhibitors induce risk of bleeding generally, and anticoagulants
a decrease in glomerular filtration rates as well as increased the risks of NSAID-induced bleeding
sodium and potassium retention. Thus, COX-2- from the upper GI tract in particular. In general,
specific inhibitors induce renal complications NSAIDs should be avoided in patients on all anti-
comparable to those observed with the use of non- coagulants. The COX-2-specific inhibitors may
selective NSAIDs [121–145]. Cyclooxygenase-2 be used in patients on anticoagulants cautiously
has been localized to the macula densa of the kid- since these drugs have minimal effects on the gas-
ney and the adjacent cortical thick ascending limb tric mucosa and on platelets. Aspirin in combina-
in the renal cortex, and it functions in regulating tion with ibuprofen, fenoprofen, naproxen, or
renin content and maintaining normal renal func- ketoprofen decreases plasma concentrations of
tion in volume-depleted states, or in patients with the NSAID, does not enhance clinical efficacy,
306 9 Management of Hyperuricemia and Gout

and is likely to cause greater toxicity [131, 161– hypoglycemic drugs, and phenytoin [167–178].
165]. Fenemates cause significant gastrointestinal On the other hand, indomethacin protein binding
toxicities, but unlike other NSAIDs, diarrhea is is decreased by ibuprofen (Motrin) [179].
the most prominent side effect [132–135]. Indomethacin also appears to increase the free
Naproxen like indomethacin has the capacity to drug levels of prednisone even though the total
increase significantly the level of prednisolone levels of this agent are unchanged [180].
even though total concentrations of this corticos- Indomethacin may also interfere with the clear-
teroid are unaffected [136]. Ibuprofen, like indo- ance of other drugs such as lithium, blood levels
methacin, can also cause hyperkalemia and acute of which are increased when it is administered
renal insufficiency [137]. Piroxicam (Feldene) concomitantly with an NSAID [181]. Conversely,
has the same propensity [138–140]. as noted previously, probenecid interferes with
The antithrombotic effect of aspirin may be the clearance of indomethacin [169, 170, 182].
inhibited by NSAIDs. NSAIDs are also said to interfere with the antihy-
Some NSAIDs may interfere with the antico- pertensive effects of some b-adrenergic blocking
agulant effect of low doses of aspirin. Aspirin at agents [183, 184]. It has been postulated that such
once daily doses of 81 or 325 mg is commonly an effect is due to the inhibition of endogenous
given to patients with cardiovascular disease to prostaglandin production and the ultimate vasodi-
prevent further myocardial infarction and strokes. latory effects of certain prostanoids [184]. The
Studies have shown conclusively that aspirin initial studies suggesting this effect were per-
lowers the incidence of these vascular disorders. formed in rabbits, but some documentation of this
It is not generally recognized that NSAIDs may problem in humans has been published [183].
prevent this important effect of aspirin. In partic- Interactions between aspirin and indometha-
ular, ibuprofen given before aspirin enters the cin have been the subject of a continuing contro-
active site of platelet cyclooxygenase (COX-1) versy, which remains incompletely resolved.
and blocks the access of aspirin. Thus, it is impor- Indomethacin levels after both a single dose of
tant that aspirin be given at least one-half hour the drug and chronic treatment with this drug are
prior to the use of ibuprofen and probably other increased when aspirin is given concurrently
low molecular weight NSAIDs in order to achieve [185, 186]. Such increments are roughly 10–20
the prophylactic effect of aspirin on cardiovascu- % greater than in the absence of salicylate treat-
lar disease. In addition, it should be noted that if ment. Chronic aspirin therapy also reduces the
ibuprofen is used several times daily, there may bioavailability of indomethacin to a level of 80 %
be sufficient quantities of the drug in the blood to suggesting a decrease in indomethacin absorp-
prevent the effect of aspirin on platelets, even if tion. Renal clearance of indomethacin is also
aspirin is given before the first dose of ibuprofen decreased in the presence of chronic aspirin ther-
in the morning [166]. However, this does not apy [185]. Despite the fact that aspirin decreases
apply to COX-2 inhibitors. The larger size of the the absorption and clearance of indomethacin,
COX-2 inhibitors prevents their access to the there is little evidence to indicate that any thera-
active site of COX-1 enzymes, and thus these peutic differences occur when these pharmaco-
drugs do not inhibit the prophylactic effect of logic agents are used together. In fact, the only
aspirin on platelets. documented study in the literature found no dif-
Several additional drug interactions may occur ference in the therapeutic effect when these drugs
with the use of indomethacin as well as other were used together, but a significant increase in
NSAIDs. These drugs (NSAIDs) displace other adverse effects provides the rationale for not
pharmacologic agents from plasma protein-bind- using these drugs together [187]. The pharmaco-
ing sites resulting in an increase in the fraction of logic activity of hydrochlorothiazide is not altered
circulating free drug [167, 168]. By this mecha- by indomethacin, so this diuretic represents a
nism, indomethacin and other NSAIDs may useful alternative to furosemide whose effect is
increase the toxicity of methotrexate, sulfonylurea, dampened by indomethacin [188]. Finally, the
Nonsteroidal Anti-inflammatory Drugs (NSAIDs) 307

Table 9.9 Classification of cyclooxygenase inhibitors The dynamic interaction between the vasodilatory
COX inhibitor Inhibitor type IC50 (COX-1/ forces of the prostaglandins and the vasoconstric-
COX-2) ratio tive responses mediated by angiotensin II and nor-
Aspirin Selective COX-1 0.01 epinephrine maintains normal renal hemodynamics,
Ibuprofen Nonselective COX 0.50 but in the absence of the vasodilatory prostaglan-
Naproxen Nonselective COX 0.56 dins, blood flow to the kidney is decreased and
S-ketoprofen Nonselective COX 0.61 renal function is impaired [189]. Thus, prosta-
Flurbiprofen Nonselective COX 1.00 glandins I2 and E2 have a significant role in renal
Sodium Nonselective COX 1.03
vasodilatory responses, renin secretion, and
salicylate
Indomethacin Nonselective COX 1.90
sodium and water excretion, but in the presence of
Piroxicam Nonselective COX 3.12 prostaglandin inhibitors, there is an increase in
Meloxicam Relatively selective 11.16 vascular tone, an antinatriuretic response, an anti-
COX-2 renin effect, and an antidiuretic response. The
Nimesulide Relatively selective 17.69 most effective way to prevent NSAID-induced
COX-2 changes in renal hemodynamics leading to renal
Diclofenac Relatively selective 18.90 insufficiency is to recognize the predisposing fac-
COX-2
tors that are likely to cause these deleterious renal
Celecoxib Selective COX-2 >30
responses. Those predisposing conditions include
Rofecoxib Selective COX-2 >400
(Vioxx) patients with congestive heart failure, cirrhosis,
These results are modified from published data contained
cirrhosis and ascites, underlying renal disease,
in the following citations: [286, 301] diuretic therapy, septicemia, hypertension, and
Aspirin inhibits COX-1 irreversibly and may cause GI shock. Patients over 65 years of age and postop-
bleeding. Sodium salicylate does not cause gastric dam- erative patients with fluid accumulation in a third
age. The phenylpropionic acids inhibit both COX-1 and
COX-2 and may cause GI and renal side effects.
space are also at increased risk for NSAID-
Nimesulide and meloxicam at high doses inhibit COX-1 induced renal insufficiency. Renal failure induced
more than COX-2. Vioxx has been removed from the mar- by indomethacin is usually rapid in its onset (24–
ket due to its adverse cardiovascular effects. Selective 48 h after indomethacin administration) and is
COX-2 inhibitors have the same effects on renal prosta-
glandins as nonselective NSAIDs
associated with oliguria. Serum creatinine levels
rise, fractional sodium excretion decreases (usu-
ally <1 %), and serum potassium levels increase.
effect of indomethacin on the gastrointestinal Of all the NSAIDs that may cause renal failure,
tract is enhanced by ethanol. This potential for indomethacin is the most common cause of this
the augmentation of the ulcerogenic properties of condition [190–192]. The combination of indo-
indomethacin also occurs with other NSAIDs as methacin and triamterene should be avoided since
well. These drug-drug interactions are summa- acute renal failure has been reported in subjects
rized in Table 9.9. with normal renal function taking this combina-
The single most significant side effect other tion of drugs [193]. Rather high doses of triam-
than gastrointestinal bleeding is the different terene were administered in these studies and may
nephrotoxic responses that may be observed with have been responsible for the marked renal distur-
indomethacin and other NSAIDs. These renal bances observed. However, on the basis of this
dysfunctions are important to recognize early report, the use of this diuretic in association with
since they are often reversible. Nephrotoxic disor- indomethacin should be avoided. Early recogni-
ders secondary to NSAID usage include acute tion of renal insufficiency and discontinuation of
renal failure, interstitial nephritis, hyperkalemia, the NSAID usually lead to improvement in renal
salt and water retention, and hypertension. The function, but short-term dialysis may be necessary
clinical syndromes associated with these nephro- [194]. Rarely, some patients may require chronic
toxic responses primarily result from the inhibi- dialysis [195]. As might be expected, hyper-
tion of renal prostaglandin synthesis by NSAIDs. kalemia is a common problem accompanying
308 9 Management of Hyperuricemia and Gout

NSAID-induced renal failure, and the fractional greater than the level observed in normal indi-
excretion of sodium may be low as well in a small viduals. Recently, parenteral ketorolac has also
percentage of patients with NSAID-related acute been associated with renal insufficiency [213,
renal failure [196]. 214]. A retrospective analysis of ketorolac has
Special consideration must be given to those concluded that short-term usage (<5 days) does
patients with underlying renal disease since not cause renal insufficiency [215]. Even though
NSAIDs often cause increased impairment of NSAID-induced renal failure is a common occur-
their already somewhat compromised renal func- rence, it is often a reversible state so that its early
tion [197–199]. In the presence of underlying recognition is important. For this reason, avoid-
renal disease, the kidney may manifest an ing the use of NSAIDs in high-risk patients and
increased production of prostaglandins to main- carefully monitoring those patients in whom
tain a normal hemodynamic state, and the inhibi- these drugs must be used are essential.
tion of prostaglandin synthesis may significantly Acute renal papillary necrosis is another
diminish renal blood flow and glomerular NSAID-induced syndrome rarely observed in
filtration rate in the prostaglandin-dependent association with these drugs [216–220]. This dis-
state. A number of mechanisms may contribute order may occur as a solitary finding related to
to the increased risk of NSAID-induced renal NSAID use or as a result of combined therapy
failure in the elderly, an important patient popu- with aspirin. Other studies have investigated the
lation that often receives NSAIDs. They may frequency of renal papillary necrosis in associa-
have low serum albumin levels with a resultant tion with chronic use of NSAIDs [221, 222]. As
decrease in NSAID protein binding and an a general rule, combinations of analgesics includ-
increase in the free drug level. Similarly, dimin- ing NSAIDs should be avoided, and patients
ished total body water, a state often observed in using NSAIDs on a chronic basis should be care-
the elderly, causes an increase in NSAID concen- fully monitored for symptoms and signs of renal
tration, and decreased hepatic metabolism of disease [223].
NSAIDs in the elderly may also lead to increased Although NSAID-induced interstitial nephri-
NSAID concentrations. Finally, any state which tis with or without an accompanying nephrotic
reduces the effective intravascular volume, such syndrome is not a disorder that intermittent
as congestive heart failure, may predispose NSAID users are prone to develop, it may be seen
patients to increased cardiac failure and fluid in patients who use NSAIDs for long periods of
retention, and these drugs should be avoided time (>6 months). It rarely occurs after short-
under these circumstances. term NSAID use. The primary features of this
To avoid these nephrotoxic effects, some have renal disease are proteinuria (usually >3 g/day),
proposed the use of NSAIDs that are less depen- nonoliguric renal failure, and focal interstitial
dent on the kidney for their metabolism [200]. cellular infiltrates with patchy fibrosis on renal
The rationale for the use of sulindac is that its biopsy [224–227]. In addition to these constant
inactive prodrug, sulindac sulfoxide, requires the features, some kidney biopsies have shown vari-
liver for conversion to its active metabolite, sulin- able immunofluorescent staining of the intersti-
dac sulfide. However, the kidney is also capable tial membranes with IgG, IgM, IgA, and C3, and
of activating this prodrug as well [201, 202]. electron-dense deposits in the mesangium may
Although sulindac spares the kidney to some also be present in a few patients. Elderly females
extent, clearly, renal failure and hyperkalemia appear to be particularly susceptible to this con-
can occur when this drug is used [197, 203–211]. dition, and peripheral edema, oliguria, anuria,
Another problem with sulindac leading to renal skin rashes, proteinuria, microscopic hematuria
problems is its long half-life that results in the and granular casts, and eosinophilia are the most
accumulation of its active agent, sulindac sulfide, common clinical and laboratory features of this
in the plasma over time [212]. In fact, plasma lev- drug-induced syndrome of renal failure [228].
els of the drug may be increased twofold or Not all these findings appear in every patient, but
Nonsteroidal Anti-inflammatory Drugs (NSAIDs) 309

renal insufficiency with significant proteinuria (spironolactone, triamterene, and amiloride),


and changes in the urine sediment are the most beta-adrenergic blockers, potassium-containing
common findings in such patients. The etiology salt substitutes, and inhibitors of angiotensin
of interstitial nephritis remains unclear, but a 1-converting enzymes. Extremely careful moni-
delayed hypersensitivity response to NSAIDs, toring of serum potassium levels is imperative if
altered arachidonic acid metabolism, and preex- indomethacin is used in these settings. Alternative
isting renal insufficiency has all been proposed as diuretics may be used to decrease the risk associ-
possible contributors to this form of renal disease ated with the potassium-sparing agents [193,
[229–234]. 256]. It is prudent to follow patients closely who
Other common complications of NSAID ther- need NSAID treatment and have serum potas-
apy are sodium retention and hypertension [235– sium levels at the upper limits of normal
241]. Edema due to salt retention occurs in (>5 meq/l) and monitor them for the possible
roughly 25 % of patients receiving NSAIDs, and occurrence of superimposed hyperkalemia. Since
careful monitoring of patients at risk for pulmo- NSAIDs may antagonize the effects of vasopres-
nary edema is essential if NSAIDs are required as sin, urinary dilution may be impaired and hypona-
therapy in these high-risk patients. Further, in tremia may ensue [257, 258].
clinical settings where parental diuretics are to be In summary, even though the prevalence of
used, the potential for NSAIDs to block diuresis NSAID-induced nephrotoxicity is relatively low,
with the resultant retention of sodium needs to be the extensive use of these agents for gout and
carefully monitored as well. NSAIDs have also many other inflammatory conditions places many
been documented to increase blood pressure both patients at risk for the renal diseases and electro-
in normotensive patients and in those with preex- lyte imbalances associated with NSAID therapy.
isting hypertension. In hypertensive patients The sale of over-the-counter NSAIDs further
receiving b-blockers and diuretics, this NSAID- complicates the problem. To identify and monitor
induced hypertensive effect is very prevalent. those patients at risk for the renal complications
Patients at highest risk for such events are those of NSAID therapy, those patients with hyperten-
with low-renin hypertension such as patients in sion, diabetes mellitus, cirrhosis, and heart dis-
the older age groups and blacks [242]. Although ease should be identified as well as those using
the effects of NSAIDs on hypertension are often diuretics and antihypertensive drugs since those
modest (6–10 mmHg), such small changes may patients deserve careful attention when they have
be of significance in patients with labile blood been prescribed NSAIDs. It is also reasonable to
pressure or hypertension, and mechanism for this measure renal function (serum creatinine), serum
NSAID-induced hypertensive effect remains sodium, and serum potassium levels prior to the
incompletely understood, but the inhibition of institution of NSAID therapy. A review of the
vasodilatory prostaglandins is certainly a promi- nephrotoxicity of NSAIDs, their diagnosis, and
nent factor [242–249]. management is available for additional consider-
Two additional less common electrolyte dis- ations regarding these disorders [259].
turbances, hyperkalemia and hyponatremia, may Other toxic effects of NSAIDs are seen in
be observed in association with NSAID usage pregnancy. Several studies have shown that the
[250–254]. Hyperkalemia following indometha- use of NSAIDs in pregnancy leads to a significant
cin therapy has occurred in both patients with increase in the incidence of abortion [260]. A
normal renal function and in those with preexist- large nested case-controlled study found that sev-
ing renal disease [228, 255]. The clinical picture eral nonaspirin NSAIDs led to an increased inci-
is identical to that observed with hyporeninemic dence of spontaneous abortions in the first
hypoaldosteronism [174, 255]. Patients at risk 20 weeks of pregnancy with an overall odds ratio
for hyperkalemia associated with indomethacin of 2.43. Other studies have reported an increased
therapy include those with renal dysfunction risk of congenital anomalies associated with the
and those taking potassium-sparing diuretics use of NSAIDs in pregnancy.
310 9 Management of Hyperuricemia and Gout

The toxicity of NSAIDs with respect to their ula densa of the renal cortex and cells adjacent to
mechanisms of action and possible alternative them [276–281]. This enzyme has also been
inhibitors of prostaglandin production deserves detected in interstitial cells in the inner medulla
further discussion since the advent of data show- and renal papilla, in the inner medullary collect-
ing adverse cardiovascular events associated ing duct cells, in the renal cortex, and in the
with COX-2 inhibitors. First, the recent demon- medullary vasa recta as well as in human podo-
stration of two isoforms of the cyclooxygenase cytes and arteriolar smooth muscle cells [275,
enzyme designated as COX-1 and COX-2 that 277, 279, 281–284]. Although there are addi-
lead to the conversion of arachidonic acid to the tional organs that may fall prey to the use of
prostaglandin endoperoxides (PGG2 and PGH2) NSAID inhibitors of prostaglandins and other
and subsequently to PGE2 and other prostanoids biochemical mediators, the gastrointestinal tract,
has significantly modified the views of the roles the cardiovascular system, and the kidney are the
of prostaglandins in both inflammatory and toxic major organ systems to be evaluated before using
responses [261]. COX-1 is constitutively either COX-1 or COX-2 inhibitors for the man-
expressed and mediates physiological responses. agement of inflammatory arthritides. Since
It catalyzes the synthesis of cytoprotective pros- COX-2 is an inducible enzyme in inflamed sites,
taglandins in thrombocytes, vascular endothe- selective COX-2 inhibitors were developed to
lium, stomach mucosa, kidneys, pancreas, act as therapeutic agents in inflammatory disor-
seminal vesicles, and brain tissue [262, 263]. ders like gout. The use of such selective COX-2
The COX-1 products (PGE2 and PGI2) reduce inhibitors was also hypothesized as a means of
gastric acid secretion, increase the thickness of avoiding the gastrointestinal toxicity of the
the mucus layer of the stomach, stimulate bicar- COX-1 inhibitors like the traditional NSAIDs.
bonate secretion, and enhance mucosal blood COX-1 is the predominant cyclooxygenase iso-
flow [263–266]. PGE2 also increases mucus form detected in the kidney and may be a key
secretion via the activation of cyclic AMP in producer of mediators regulating renal function
gastric epithelial cells [267]. These functions of such as intrarenal plasma flow and electrolyte
the COX-1 product, PGE2, clearly demonstrate balance [285, 286]. Since the selectivity of
why inhibition of COX-1 activity may impair cyclooxygenase inhibitor varies between selec-
gastroprotective functions. Such COX-1 func- tive COX-1 inhibitors and selective COX-2
tions demonstrate why the pursuit of COX-2 inhibitors, a method has been devised to classify
inhibitors appeared to be a means of preventing NSAIDs into four categories: selective COX-1
inflammatory responses and avoiding gastroin- inhibitors, nonselective COX inhibitors, rela-
testinal side effects. tively selective COX-2 inhibitors, and highly
Although COX-1 and COX-2 isoforms have selective COX-2 inhibitors. This classification
similar amino acid sequences, they have quite has been derived from published data, and the
different functions [263, 268]. COX-2 is respon- separation into these categories is based on
sible for the production of prostanoids involved in vitro whole blood assays using NSAIDs to
in pathological processes like inflammation, and determine IC50 values for COX-1 and COX-2
this enzyme is induced by various proinflammatory inhibition [283–288]. Using this IC50 COX-1/
agents and endotoxins [269–273]. The other COX-2 ratio, the following drugs can be classified
important organ system related to NSAIDs and into those four categories (Table 9.9). As can be
their effects on organ function is the kidney. determined from the data, the selective COX-1
COX-1 is constitutively expressed in the kidney and nonselective COX inhibitors have COX-1/
and has been localized to mesangial cells, arte- COX-2 ratios between 0.5 and 3.5, whereas the
riolar smooth muscle cells, endothelial cells, selective (relatively) and highly selective COX-2
parietal epithelial cells of Bowman’s capsule, inhibitors have IC50 values between 10 and 400.
and the cortical and medullary collecting ducts Gastrointestinal and cardiovascular toxicities of
[274, 275]. COX-2 has been detected in the mac- the NSAIDs are delineated.
Nonsteroidal Anti-inflammatory Drugs (NSAIDs) 311

Although there are a variety of toxic responses pertinent to those patients who are elderly, have
to NSAIDs, the three most significant organ sys- preexisting renal dysfunction, or who have hyper-
tems affected by these agents are the gastrointes- tension [306, 318–320].
tinal tract, the kidney, and the cardiovascular Many human and animal studies have investi-
system. Nonsteroidal anti-inflammatory drugs, gated the role of prostaglandins in renal hemody-
especially those NSAIDs that are nonselective namics [150, 277, 321–333]. In animals, COX-2
inhibitors of cyclooxygenase, are known to place inhibitors have been shown to decrease plasma
patients at increased risk for gastrointestinal renin levels, renal renin release, and COX-2
mucosal damage [289–302]. Thus, a variety of mRNA expression in certain high-renin settings
esophageal, gastric, and small bowel lesions can [150, 325–332]. Humans utilizing a low-sodium
occur such as esophagitis, esophageal stricture, diet and/or receiving furosemide demonstrated
gastritis, gastric mucosal erosions, gastric bleeds, an inhibition of renin release when treated with
peptic ulceration, small bowel erosions, and large COX-2 inhibitors [324, 333]. Some COX-2
bowel erosions [303–305]. Increased risks of inhibitors have also been shown to block the
gastrointestinal disease are well documented in hyperreninemia associated with antenatal
patients using NSAIDs as well as in patients older Bartter’s syndrome/hyperprostaglandin E syn-
than 65 years of age; in those with a history of drome [334, 335]. There is also evidence to sug-
peptic ulcer disease, GI bleeding, or the use of gest that COX-2 inhibition impairs the
antacids; the concomitant use of steroids and vasodilatory responses mediated by COX-2 prod-
NSAIDs; the presence of comorbid conditions ucts that regulate renal blood flow and glomeru-
like cardiovascular disease; high doses of lar filtration rates, especially in volume-depleted
NSAIDs; and combinations of NSAIDs. states [336, 337]. In addition to these findings
It has also been well recognized that nonselec- and others, COX-2 inhibitors have been reported
tive NSAIDs have adverse effects on the kidneys to cause type IV renal tubular acidosis/acute isch-
since they may cause salt retention and increase emic renal insufficiency and hyperkalemia [337–
in blood pressure and a reduction in glomerular 341]. It is clear, then, from reviews of the renal
filtration rates in some patients. Such findings effects of COX-1 and COX-2 inhibitors that treat-
were a part of the impetus for evaluating COX-2 ment with these inhibitors may be associated
inhibitors that might avoid these side effects with edema, hypertension, and acute renal failure
[306]. Despite the concept that COX-1-derived in some patients [342–353].
PGE2 regulates salt and water balance and COX-2 Despite all the data concerning COX-1 and
inhibitors do not influence sodium excretion, COX-2 prostanoid production and their role in
there is now evidence that renal medullary inter- the functions of the kidney in health and disease,
stitial cells express COX-2, and inhibitors of the it remains to be completely resolved whether
production of prostanoids by COX-2 reduce salt COX-1 or COX-2 is the predominant regulator of
excretion and lead to salt-dependent hyperten- renal functions. However, there is no question
sion [144, 307–312]. In addition to these changes that there are risks to the kidney when either
in salt and water regulation by prostaglandins, COX-1 or COX-2 inhibitors are used. In the kid-
COX-1 inhibitors may also impair the diuretic ney, COX-1 provides the kidney with PGE2, PGI2,
effects of furosemide and the antihypertensive and TXA2, whereas COX-2 generates PGE2, PGI2,
effects of other antihypertensive medications TXA2, and PGD2. The receptors for these metab-
including angiotensin-converting enzyme inhibi- olites have been localized to a variety of renal cell
tors, b-blockers, and diuretics [313–317]. On the types [344]. The PGE2 receptor subtypes have
basis of the foregoing data as well as other stud- been found in the following cells: EP1 in the pod-
ies, both COX-1 and COX-2 inhibitors need to be ocytes and collecting duct; EP2 in the vasculature,
carefully monitored for changes in blood pres- interstitial cells, and macula densa; EP3 in the dis-
sure and sodium balance in patients treated with tal tubule, collecting duct, and vasculature; and
these agents. These risk factors are especially EP4 in the vasculature, podocytes, glomerulus,
312 9 Management of Hyperuricemia and Gout

mesangial cells, macula densa, interstitial cells, healthy host should also be monitored for changes
and proximal tubule. The IP receptor for PGI2 is in blood pressure, electrolytes, or other nephro-
found in the vasculature, mesangial cells, macula toxic responses.
densa, distal tubule, collecting duct, and proximal The last and perhaps the most significant toxic
tubule. TP, the receptor for TXA2, is found in manifestation related most specifically to the use
most renal cells, and FP, the receptor for PGF2a, of COX-2 inhibitors is the increased risk of car-
has been localized to the podocytes, distal tubule, diovascular events such as myocardial infarction,
collecting duct, and fibroblasts. The EP receptors cerebrovascular disease, and peripheral vascular
mediate the effects of PGE2, IP receptor mediates disease as well as hyperlipidemia. COX-2 inhibi-
the PGI2 effects, the TP receptor mediates the tors also increase the risk of atherothrombotic
effects of thromboxane A2, and the FP receptor events in diabetic patients. The initial studies of
mediates the effects of PGF2a. Little is known COX-2 inhibitors showed that these agents were
about the DP receptor for PGD2 in the kidney, but as effective as COX-1 inhibitors in the manage-
evidence does show the presence of its product ment of inflammation and pain [364–367]. The
and its effects in animal kidney cells [354–358]. second conclusion concerning COX-2 inhibitors
These prostaglandin receptors are associated with is that these drugs have fewer gastrointestinal
a variety of functions. The EP1 receptor is involved side effects than standard or traditional NSAIDs
with hemodynamics and transport, and the EP2 in many but not all studies [130, 368–376]. The
receptor is a mediator of renin release. The EP3 most critical element with respect to cardiovascu-
receptor is involved in vasoconstrictive responses lar and vascular diseases has been related princi-
and transport functions, whereas the EP4 receptor pally but not completely to the functions of the
is associated with transport functions, vasodila- two prostanoids, PGI2 and TXA2 [377]. One
tion, hemodynamics, renin release, antiapoptosis, method that confirms the importance of PGI2 and
cytoskeletal functions, and growth. The IP recep- TXA2 to vascular functions is the measurement
tor mediates a variety of functions including of the TXA2 and PGI2 metabolites in the blood
vasodilation, hemodynamics, transport, matrix and urine. The serum levels of 6-keto-PGF1a and
synthesis, renin release, antiapoptosis, fibrosis, the urinary levels of 2,3-dinor 6-keto-PGF1a rep-
and growth. The TP receptor is involved with resent indices of PGI2 production, whereas serum
vasoconstriction, hemodynamics, transport, TXB2 levels and urinary levels of 11-dehydro
matrix synthesis, apoptosis, fibrosis, and growth. TXB2 represent indices of TXA2 production.
Finally, the FP receptor is associated with trans- Many studies have now shown that both COX-1
port and cell transformation. As can be deter- and COX-2 inhibitors significantly reduce the
mined from the functions subtended by their levels of PGI2 metabolites [378–383]. Thus, the
prostaglandin receptors, many different functions reduction of PGI2 and the increase in unopposed
are mediated by these receptors and the balance TXA2 have been proposed as the means by which
between such functional parameters is incom- cardiovascular events associated with NSAIDs
pletely characterized at this point in time. This occur. There is now a significant body of evi-
picture of renal functions is also complicated by dence that implicates an increased frequency of
species differences in the localization of such thrombotic events including myocardial infarc-
receptors and their functions [359–363]. These tion and strokes in association with the use of
studies demonstrate the differences in the pros- both traditional NSAIDs and the newer COX-2
tanoids and their functions between species and inhibitors [368, 384–404].
indicate the difficulties in drawing simple conclu- The most recent publications and reviews sort
sions across species. In conclusion, the use of out the role of COX-1 and COX-2 inhibitors on
prostaglandin inhibitors in humans with preexist- the risk of cardiovascular events but do not
ing renal disease, hypertension, or electrolyte resolve the issue and do not conclude with a
imbalance needs to be carefully monitored. These strong recommendation for a single agent to
COX-1 and COX-2 inhibitors when used in the circumvent these toxic events [302, 388, 394,
Indomethacin (Indocin) 313

404, 405]. The removal of some of the COX-2 inhibitor on the market presently since it shows
inhibitors from the market has helped to simplify no increased risk for hypertension or edema over
the decisions to use these drugs or not. A wide a dose range of 200–300 mg/day. Celecoxib also
variety of studies have evaluated cardiovascular carries the lowest risk for gastrointestinal side
and gastrointestinal risks including long-term effects. There is some evidence that acetamino-
placebo-controlled trials, comparative trials phen, nonselective NSAIDs, or selective COX-2
between COX-2-specific inhibitors and tradi- inhibitors used for more than 2 weeks in females
tional NSAIDs, and epidemiological studies may increase the risk of hypertension or cardio-
comparing users of COX-2-specific inhibitors vascular events [411]. Finally, three additional
and NSAIDs and nonusers of anti-inflammatory facets with respect to NSAIDs are worth empha-
drugs [392]. The initial observations of the link sizing. First, all the available NSAIDs have the
between myocardial infarction and NSAID use propensity to increase the risk of cardiovascular
were determined in the year 2000 and appeared events. Second, the lowest efficacious dose of any
to be associated with the commencement of NSAID should be used to avoid any potential side
NSAID therapy [406]. A review of this subject in effects. Third, if hypertension or edema occurs
subsequent studies confirmed that the COX-2 during the use of an NSAID, this complication
inhibitor rofecoxib caused an increase in myocar- should be treated aggressively.
dial infarction in 12–14 studies evaluating the use We will review two NSAIDs in detail, indo-
of this drug. Furthermore, the increased risk of methacin and naproxen, since these two agents
myocardial infarction in other studies varies are among the most frequently used and both
depending on the COX inhibitor. When celecoxib have been validated for the treatment of acute
was evaluated, only 4 of 15 studies demonstrated gouty arthritis in randomized controlled clinical
a significant risk for myocardial infarction [407– trials. Controlled trials have been performed with
410]. The general conclusion from all these dif- other NSAIDs but only as comparators with indo-
ferent studies permits the following conclusions. methacin or naproxen.
Rofecoxib, now off the market, yields a high risk
for myocardial infarction. Celecoxib studies
reveal some risks for myocardial infarction. It has Indomethacin (Indocin)
also been determined that indomethacin,
diclofenac, and meloxicam may have cardiovas- The structure of this NSAID is shown in Fig. 9.3.
cular risk factors similar to rofecoxib. There are Indomethacin is as effective as phenylbutazone in
some shortcomings in these population studies the treatment of acute gout and is much less likely
where smoking, alcohol use, and obesity were to cause aplastic anemia. For these reasons, it is
not considered, and these variables may influence often the drug of choice for the management of
the occurrence of cardiovascular and gastrointes- acute gout. Indomethacin is an indole acetic acid
tinal toxicities [411]. Thus, further studies includ- whose chemical structure is similar to the sulfox-
ing analyses of the foregoing factors may define ide sulindac but not to other nonsteroidal anti-
a more complex set of risk factors for cardiovas- inflammatory drugs. Its pharmacologic properties
cular events. are listed in Table 9.10. In addition to these prop-
In summary, nonspecific inhibitory NSAIDs erties, it should be recognized that this drug
together with a proton pump inhibitor or other crosses the placental membrane, and significant
gastroprotective agents may circumvent the use of concentrations of the drug have been measured in
COX-2 inhibitors and avoid gastrointestinal com- fetal cord blood. The half-life of indomethacin is
plications from these drugs. This strategy is espe- prolonged in newborns and infants (>15 h) as
cially useful in elderly patients with cardiovascular compared to adults. Indomethacin is recirculated
risk factors or those who have had previous myo- via the biliary tract making its serum half-life
cardial infarctions or other atherothrombotic epi- difficult to measure accurately. This drug is
sodes. Celecoxib appears to be the safest COX-2 significantly bound to plasma albumin where a
314 9 Management of Hyperuricemia and Gout

Table 9.10 Indomethacin: pharmacologic properties Table 9.12 Relative contraindications to indomethacin
therapy
Peak fasting concentrations occur in 45 min after an
oral dose Peptic ulcer disease
Protein binding is 98–99 % Inflammatory bowel disease
Bioavailability is 90–98 % Renal or cardiac failure
Biological half-life is 5–10 h Cirrhosis with ascites
Indomethacin has one high-affinity protein-binding site Psychiatric disorders
as well as secondary binding sites Epilepsy
Ibuprofen decreases indomethacin protein binding Parkinson’s disease
Indomethacin increases plasma lithium levels Platelet dysfunctions
Indomethacin interferes with antihypertensive effects Salt-dependent hypertension
of b-adrenergic agents Potassium-retaining diuretics
Probenecid decreases biliary clearance of indomethacin Furosemide therapy (antagonist to natriuresis)
Probenecid increases clinical effectiveness of Thiazide diuretics
indomethacin
Beta-adrenergic agents
Synovial fluid indomethacin levels are equal to or
above serum levels for 9 h after one dose Aspirin sensitivity
Aspirin decreases indomethacin absorption and renal Probenecid therapy (indomethacin excretion is
clearance blocked)
Aspirin increases biliary clearance of indomethacin

until the attack has resolved. In each case, the


Table 9.11 Indomethacin metabolism treatment schedule may have to be prolonged
Indomethacin undergoes enterohepatic recirculation with severe flares of gout [412].
Indomethacin is metabolized by O-demethylation, There are relative contraindications to indo-
N-deacylation, and glucuronidation to inactive methacin, and these are summarized in
metabolites Table 9.12.
After an oral dose about, 16 % of indomethacin is The uricosuric drug probenecid inhibits the
found in the urine as a free drug, and 82.5 % is
metabolized or conjugated, and 1.5 % appears as a free tubular excretion of indomethacin by the kidney
drug in the feces and may interfere with the biliary clearance of
indomethacin which may enhance the clinical
efficacy of the drug and increase its toxicity
single high-affinity site has been characterized [169–171, 182]. Thus, a lower dose of indo-
with seven secondary sites to which indometha- methacin may be used in patients taking the uri-
cin also binds [179]. Extensive metabolism of the cosuric drug probenecid (Benemid).
drug occurs in vivo including O-demethylation,
N-deacylation, and glucuronidation (Table 9.11).
In the range of 20 % of the free drug is excreted Toxicity
by the kidney, so renal disease has the propensity
to alter its excretion and increase its toxicity. The toxicity of indomethacin is similar to the tox-
icity of all nonselective cyclooxygenase inhibi-
tors. Gastrointestinal and renal toxicity are
Treatment Schedules probably the most common. In addition, central
nervous system symptoms such as confusion,
Recommended regimens for the treatment of inability to concentrate, headache, drowsiness,
acute gouty arthritis include the following: 50 mg tremor, and even psychosis occur in a frequency
orally three times daily for 2 days and followed comparable to the gastrointestinal side effects of
by 25 mg three times daily for 3 days or, alterna- the drug. Skin rashes, hepatocellular damage,
tively, 50 mg three times daily for 3 days and fol- coagulation disturbances, and bone marrow apla-
lowed by 25 mg three times daily for 4–7 days or sia rarely occur (Table 9.13).
Naproxen 315

Table 9.13 Indomethacin toxicity Table 9.16 Naproxen toxicity


Gastrointestinal – dyspepsia, nausea, diarrhea, Gastrointestinal – heartburn, GI bleeding, and
abdominal pain, and peptic ulcer abdominal discomfort
CNS – headache and dizziness CNS – headache, drowsiness, and lightheadedness
Skin – rashes Skin – rarely urticaria, ecchymoses, and vasculitis
Cardiovascular – tachycardia Renal – rarely causes acute interstitial nephritis or renal
Hematological – anemia, rarely aplastic anemia papillary necrosis
Hepatic – rarely jaundice
Pulmonary – naproxen-induced asthma
Table 9.14 Pharmacology of naproxen
Naproxen is 99 % bound to plasma proteins (albumin)
Its mean plasma half-life is 14 h might have been predicted that these agents
Its volume of distribution is 0.09 l/kg would only have minor effects of acute gout since
Its total clearance is 0.004 l/kg/h several mediators are thought to be involved in
It is metabolized to glucuronide conjugates this inflammatory reaction. Nonetheless, NSAIDs
Ten percent is eliminated as the unchanged drug are very effective therapeutic agents for acute
Sixty percent is eliminated as conjugated naproxen gout, suggesting that prostaglandins are major
Twenty-eight percent is converted to mediators here. The NSAIDs are thought to be
6-desmethylnaproxen most effective when given early in the acute
Urinary excretion is primarily as the conjugated drug or
attack, indicating that other mediators than
its metabolite 6-desmethylnaproxen
cyclooxygenase products may become important
later in the attack.
Table 9.15 Naproxen: drug-drug interactions There are additional prospects for the treat-
Naproxen may alter prothrombin time ment of the acute inflammation associated with
Naproxen may increase the effects of warfarin gouty arthritis that may become a reality in the
compounds future. It is possible that leukotriene B4 receptor
Aspirin increases the clearance of naproxen
antagonists might be of use in acute gout; how-
Benemid increases naproxen half-life and retards its
clearance
ever, the redundancy of the neutrophil chemot-
axins makes it unlikely that these inhibitors
would be useful. More likely candidates are the
phospholipase inhibitors that block the genera-
Naproxen tion of the substrate for the proinflammatory
prostaglandins, arachidonic acid. However, these
Another NSAID frequently used for the treat- inhibitors must be designed to inhibit specific
ment of acute gouty arthritis is naproxen. Two phospholipases and not impair those prostaglan-
treatment regimens include either 500 mg twice din-generating molecules critical for physiologi-
daily for 5 days or 375–500 mg twice daily for cal purposes. Since monosodium urate crystals
3 days, followed by 250–375 mg orally twice have been shown to stimulate phospholipase A2
daily for 4–7 days or until the attack resolves activities and the synthesis of a phospholipase
[412]. A2-activating protein, inhibitors of phospholi-
Side effects of naproxen are similar to those of pases might be useful for the suppression of the
other NSAIDs described above. However, the proinflammatory cytokines synthesized by this
cardiovascular toxicity of myocardial infarction route. In addition, it appears that there are differ-
and strokes that are seen with both COX-1 and ent arachidonic acid sources arising from differ-
COX-2 inhibitors have been found to be lower ent phospholipases. On a speculative basis, the
with naproxen than any other NSAID in epide- development of specific phospholipase inhibi-
miologic studies (Tables 9.14, 9.15, and 9.16). tors in the future may have some relevance to the
Since the major effect of NSAIDs appears to treatment of gout [413–415]. Many different
be the inhibition of cyclooxygenase products, it studies have now been published on the role of
316 9 Management of Hyperuricemia and Gout

phospholipases in the release of arachidonic acid Glucocorticoids


and the regulation of these enzymes [416–419].
Thus, the contributions of phospholipases to the Glucocorticoids are used in the treatment of a
synthesis of inflammatory mediators represent a wide variety of inflammatory and immune disor-
complex process since it necessitates interac- ders since they suppress the generation and
tions between cytoplasmic phospholipase A2s, release of mediators of inflammation. They also
secretory phospholipase A2s, and the cyclooxy- modify the functions of cells recruited to
genases. Different stimuli coupled to a variety of inflammatory sites. In the human, endogenous
prostaglandin biosynthetic enzymes act on dif- levels of cortisol in the plasma vary between 5
ferent cellular arachidonic acid pools at different and 25 ug of cortisol/100 ml of plasma, and
locations to give rise to immediate and delayed almost all the hormone (90 %) circulates bound
arachidonic acid-derived mediators. These com- to plasma proteins. About 80 % is bound to a-
plex reactions are in the early stages of investi- globulin transcortin (transcortin-corticosteroid-
gation with respect to the synovial compartment binding globulin) through a high-affinity binding
and its cellular components in healthy and dis- site, whereas a low-affinity site exists on albumin
eased tissues [420–423]. The significance of the [426, 427]. Since this discussion is concerned
phospholipases to the generation of inflammatory with exogenously administered corticosteroids,
mediators is clear, and in the future, the path- emphasis is placed on the absorption, metabo-
ways responsible for the hydrolysis of mem- lism, and excretion of those agents commonly
brane phospholipids and the production of used as anti-inflammatory corticosteroids such as
arachidonic acid for mediator synthesis may hydrocortisone, prednisone, prednisolone, and
become therapeutic targets [424]. In fact, anti- dexamethasone. These drugs are absorbed pri-
bodies specific for secretory phospholipase A2 marily in the upper jejunum, and peak plasma
isoforms have been constructed and utilized to levels are attained within 1 h or 2 after their inges-
determine the tissue location and cell-specific tion. Low serum albumin and transcortin levels
functions of these phospholipases [425]. If such as a result of hepatic disease cause an increase in
antibodies against phospholipases are deter- free drug levels [428]. The plasma half-life of
mined to have specificity for specific tissues, synthetic glucocorticoids such as prednisone is
they could also be used as therapeutic agents to about 1 h, whereas the biological half-life of the
block mediator production in specific tissues. drug lasts much longer than the plasma half-life.
Thus, as the phospholipase pathways leading to Glucocorticoids are metabolized primarily by the
the generation of prostaglandins, leukotrienes, liver where they are reduced and conjugated with
and other lipid mediators are better character- glucuronic acid and subsequently excreted in the
ized, the inhibition of such reactions may pro- urine. As noted previously, liver disease may
vide additional potent therapeutic tools for the increase free drug levels and prolong the half-life
management of acute inflammatory responses of the drug. In terms of relative potency with
like those observed in gout. It is important to hydrocortisone designated as one, prednisone
recognize that there are a number of physiologi- and prednisolone have four times the potency of
cal compounds that have recently been discov- hydrocortisone [427]. The pharmacokinetic and
ered from investigations of the resolution of metabolic parameters of some steroids are shown
acute inflammatory processes, and these com- in Tables 9.17, 9.18, and 9.19, and the structure
pounds may well have properties useful for the of some common steroids is shown in Figs. 9.4
resolution of an acute attack of gout. These com- and 9.5.
pounds are reviewed in Chap. 8. There are other The mechanism of action of corticosteroids is
new initiatives related to the discovery of more a complex process incompletely characterized at
effective drugs against inflammatory responses, this time. Glucocorticoids enter cells via simple
and they are discussed at the end of this or facilitated diffusion. They induce changes in
chapter. cyclic nucleotides, membrane fluidity, and ion
Glucocorticoids 317

Table 9.17 Steroidal anti-inflammatory agents: pharma- Table 9.19 Pharmacokinetics of intravenous prednisone
cokinetics and metabolism and prednisolone at different doses
Pharmacokinetics Prednisone/prednisolone Parameters Plasma Plasma
GI absorption (%) Rapid/variable prednisone prednisolone
Plasma protein binding (%) 65–70 5-mg dose
Peak plasma levels (h) Dose dependent T ½ (h) 2.7 ± 0.4 3.0 ± 2.1
Half-life (hrs) 2.5–3 MRT (h) 3.8 ± 0.5 5.1 ± 3.2
Metabolism Hepatic Dose PDL/AUC (PDL) 111 ± 5.0 712 ± 256
Volume of distribution (l/kg) 0.4–1.0 ml/min/1.73 m2 (calculated)
Excretory route Urine AUC (PDL)/AUC (PD) 4.99 –
20-mg dose
T ½ (h) 2.6 ± 0.5 3.2 ± 1.0
Table 9.18 Pharmacokinetics of oral prednisone/predni- MRT (h) 3.9 ± 0.6 5.8 ± 1.3
solone at different doses
Dose PDL/AUC (PDL) 157 ± 25 1,436.8 ± 520.1
Parameters Plasma Plasma ml/min/1.73 m2
prednisone prednisolone (calculated)
5-mg dose AUC (PDL)/AUC (PD) 7.2 –
T ½ (h) 3.5 ± 2.5 2.3 ± 0.3 40-mg dose
MRT (h) 4.9 ± 0.9 3.9 ± 0.4 T ½ (h) 3.5 ± 0.51 4.2 ± 1.0
Dose PD/AUC (PDL) 572 ± 155 116 ± 25 MRT (h) 4.6 ± 0.5 6.6 ± 1.2
ml/min/1.73 m2 Dose PDL/AUC (PDL) 194 ± 29 2,028.8 ± 363.2
AUC (PDL)/AUC (PD) 4.7 ± 1.1 – ml/min/1.73 m2
20-mg dose AUC (PDL)/AUC (PD) 10.4 –
T ½ (h) 3.3 ± 1.1 2.9 ± 0.6 These data are all modified from data published by Rose
MRT (h) 6.5 ± 0.8 5.5 ± 0.9 [1048]
Dose PD/AUC (PDL) 1,034 ± 139 185 ± 23 Examining the AUC ratios demonstrates a nonlinear dose
ml/min/1.73 m2 response. The rapid conversion of PD to PDL is consistent
with first-pass extraction or presystemic biotransforma-
AUC (PDL)/AUC (PD) 6.1 ± 0.8 –
tion. The nonlinear pharmacokinetics is most likely due to
50-mg dose concentration-dependent binding of PD to transcortin and
T ½ (h) 3.4 ± 1.1 3.4 ± 1.0 albumin, the interconversion of PD and PDL, and the
MRT (h) 7.1 ± 2.2 5.9 ± 1.8 concentration-dependent clearance
Dose PD/AUC (PDL) 2,271 ± 791 233 ± 28 The abbreviations used in this table and Table 9.18 are
ml/min/1.73 m2 MRT (mean residence time of drug in the body), PD pred-
AUC (PDL)/AUC (PD) 10 ± 4.0 – nisone, PDL prednisolone, and AUC area under the curve

channels, but their principal effect of significance where it binds to glucocorticoid response ele-
to their anti-inflammatory properties is to bind to ments (GRE) in the chromatin. Binding of the
intracellular glucocorticoid receptors with the steroid receptor complex to GRE in the promoter
subsequent induction of cellular responses lead- region of genes and interactions with other tran-
ing to new protein synthesis [429–437]. The scription factors result in the transcription of
emphasis in this discussion is on the glucocorti- DNA and the formation of specific mRNAs
coid receptor-mediated effects since they are the [440–443].
most relevant to the acute inflammation observed Since the identification of an inducible form of
in gout. cyclooxygenase-2 and its linkage to inflammatory
Intracytoplasmic glucocorticoid receptors are responses, the inhibition of eicosanoid biosynthe-
present in many different cell types including sis by glucocorticoids has been clearly demon-
neutrophils and monocytes [433, 438, 439]. Once strated in many studies and its significance to the
glucocorticoids interact with these intracytoplas- inflammatory process vigorously pursued [94,
mic receptors, the glucocorticoid-receptor com- 444–446]. Glucocorticoids inhibit both in vivo
plex is then translocated to the nucleus of the cell and in vitro prostaglandin production in cells and
318 9 Management of Hyperuricemia and Gout

Naproxen CH3 concentration-dependent manner [453]. These


studies have now been supplemented by data
CHCOOH showing glucocorticoid-induced inhibition of
cyclooxygenase synthesis in human dermal
CH3O fibroblasts, differentiated human macrophage cell
lines (U937), blood monocytes, cultured vascular
Fig. 9.4 The structure of naproxen (Naprosyn). The cells, and mouse peritoneal macrophages [449,
chemical name for naproxen is (S)-6-methoxy-a-methyl- 450, 454–456]. It has now been documented that
2-naphthyleneacetic acid. It is a propionic acid derivative
related to the arylacetic acid group of nonsteroidal anti- glucocorticoids are only effective as inhibitors of
inflammatory drugs and is soluble in lipids and water at a COX synthesis in cells in which COX can be
high pH. It is insoluble in water at a low pH. This drug has induced by IL-1, LPS, or other inducing agents.
anti-inflammatory, analgesic, and antipyretic properties Although many of the effects of glucocorti-
coids are related to their binding to the intracel-
Steroid Nucleus lular glucocorticoid receptor, binding which is
saturated at relatively low concentrations, other
12 17 studies have indicated that there are effects of
11 16 glucocorticoids which are apparently not
accounted for by this receptor binding [457]. This
1
2 9 14 15 may be for some of the actions of glucocorticoids
10 8 seen at high concentrations of these agents. It
3 7 may provide a rationale for the use of pulses of
5
4 6 intravenous glucocorticoids at high concentra-
CH2OH tions that are employed under various circum-
Cortisol (hydrocortisone) stances [457].
C O The key to understanding the role of the glu-
HO OH cocorticoid-induced anti-inflammatory proteins
and the possible means by which therapeutic
agents mimicking their effects lies with the char-
acterization of the glucocorticoid-induced pro-
teins. The discovery of the annexin family of
O glucocorticoid-inducible proteins that bind
anionic phospholipids in a calcium-dependent
Fig. 9.5 The structure of the steroid nucleus and the cor-
tisol molecule. The attachment of the two-carbon chain at manner represented a major breakthrough [458].
position 17 of the steroid nucleus gives rise to the C-21 The fact that annexins including annexin-1 have
steroids. Glucocorticoid activity is completely dependent been found in synovial fluid provides further evi-
on the presence of a hydroxyl group at position 11 of the dence for their role in arthritic processes [459,
steroid nucleus. The addition of a double bond between
atoms 4 and 5 of the steroid nucleus increases the anti- 460]. The finding that sealed the association
inflammatory action of the molecule between arthritis and annexins was the anti-
inflammatory effect of annexin (lipocortin-1) in
the experiment of arthritides [461–463]. From
tissues exposed to these drugs [93, 94, 99, 102, these studies, it was clear that annexin-1 is one of
447–452]. Prednisone administered orally to the key endogenous regulators involved in
human volunteers at a dose of 60 mg for 7 days inflammatory arthritides.
has been shown to inhibit zymosan-induced The mechanism of action of the 37-kD gluco-
eicosanoid release from alveolar macrophages corticoid-inducible protein annexin is through
[453]. Treatment of cultured macrophages with its export to the cell surface and its interaction
hydrocortisone has also been shown to inhibit with specific binding sites that alter phospho-
the release of eicosanoids in a time- and lipid release [464]. Subsequently, a number of
Glucocorticoids 319

Prednisone CH2OH coids suppress total immunoglobulin production


(especially IgA and IgG subtypes and IgE);
C O
decrease the alternate complement pathway pro-
O teins (C3 and factor B); reduce the biosynthesis
OH
of leukotriene B4, prostaglandin E2, and prosta-
glandin F2a; suppress the induction of cytokine
mRNAs by IL-1 for IL-1b, IL-6, granulocyte/
O
macrophage-colony-stimulating factor; suppress
the induction of cytokine mRNAs by lipopoly-
Prednisolone CH2OH saccharide for human monocyte-derived neutro-
phil and T cell chemotactic factors (IL-8);
C O
suppress histamine release; and enhance the pro-
HO OH duction of IL-1 and IL-6 receptors as well as the
acute phase reactants induced by IL-1 and IL-6
[477–495]. Glucocorticoids also cause a periph-
eral blood neutrophilia with a neutropenia at the
O
site of inflammation, a depletion of peripheral
Fig. 9.6 The structure of prednisone and prednisolone. blood monocytes, and a depletion of eosinophils
These are two commonly used glucocorticoids with anti- and basophils as well [494, 496–502]. These ste-
inflammatory properties roids also inhibit the chemotaxis of professional
phagocytes, inhibit phagocytosis and pinocyto-
investigations have demonstrated that annexins sis, suppress oxidative antimicrobial functions,
and their analogs reduce phospholipase activity, and decrease specific granule release from baso-
the source of arachidonic acid used for the syn- phils. These hormonal agents also inhibit anti-
thesis of proinflammatory mediators [460, 465– gen-induced cell proliferation and suppress the
474] (Fig. 9.6). production of lymphokines [496]. The multiplic-
Recent studies have examined more critically ity of glucocorticoid effects including their direct
glucocorticoid-induced annexin in leukocyte effect on inflammatory cells clearly establishes
subsets and measured their intracellular content the mechanisms of action for these potent anti-
as well as their capacity to secrete annexins inflammatory agents that would alter the acute
[475–477]. Dexamethasone at a concentration of inflammatory response to monosodium urate
10−6 M induced the secretion of annexin-1 from crystals.
human-cultured monocytes [477]. This secretory The questions concerning the relationships
process is rapid, occurring within 15 min after between glucocorticoid-induced functions,
exposure to this corticosteroid. Constitutive lipo- annexin, and inflammatory responses have now
cortin-1 concentrations were found to be highest begun to be characterized in greater detail. In a
in neutrophils and monocytes and lowest in B rodent model of inflammation, lipopolysaccha-
lymphocytes [475]. Annexin binding sites are ride has recently been shown to induce the adhe-
increased in neutrophils following adhesion to sion and subsequent emigration of neutrophils
endothelial cells [476]. Such investigations set from the postcapillary venules [503]. Annexin-1
the stage for understanding the role that annexin or dexamethasone reduced the adhesion and
(lipocortin) plays in inflammatory cells and their transmigration of neutrophils induced by lipopoly-
functional responses. Since glucocorticoids mod- saccharides in this animal model. Other animal
ulate both noncyclooxygenase functions, cellular studies have characterized the action of annexins
components of the inflammatory and immune in more detail as to their capacity to alter an
responses, and cyclooxygenase-dependent func- inflammatory reaction [504–512]. Dexamethasone
tions, a wide variety of cellular activities could be has been shown to inhibit leukocyte transmigra-
regulated by annexins. For example, glucocorti- tion in the hamster cheek pouch model, but it does
320 9 Management of Hyperuricemia and Gout

not alter leukocyte rolling or adhesion [507]. human. The focus on identifying the sites of
Thus, lipocortin inhibits leukocyte diapedesis action of glucocorticoids and their functional
directly. The mechanism by which transendothe- responses as well as their mechanism of action
lial passage of neutrophils is blocked results from may lead to the development of glucocorticoid-
the mobilization of lipocortin and its externaliza- like molecules with fewer or no side effects.
tion after neutrophils adhere to venular endothe- Further, the use of glucocorticoid-induced pro-
lium [509]. Subsequently, studies of mouse teins, like the annexins, with anti-inflammatory
mesenteric postcapillary venules using phago- properties may lead to the development of a
cytic stimulus, zymosan, definitively demon- totally new class of therapeutic modalities.
strated that recombinant human lipocortin
provoked the detachment of adherent neutrophils
from activated postcapillary endothelium [508]. Therapeutic Regimens
In addition, glucocorticoids via the lipocortin
mechanism have been shown to reduce IL-1 beta- Oral corticosteroids are frequently useful for the
induced neutrophil adhesion and the expression treatment of acute gout when there is a contrain-
of intercellular adhesion molecule-1 (ICAM-1) in dication to the use of other medications.
postcapillary venules [506]. Glucocorticoids also Glucocorticoids are particularly useful for the
significantly reduce IL-1- and IL-8-induced neu- treatment of acute gout under circumstances
trophil migration and tissue infiltration [504]. where NSAIDs and colchicine are contraindi-
Corticosteroids also suppress the rate of particle cated, such as patients with renal failure or car-
phagocytosis and monocyte recruitment via lipo- diovascular disease. One recommended schedule
cortin induction [510]. Very recent studies have is 30–35 mg of prednisone of prednisolone orally
also identified the site of lipocortin action as the daily for 5 days. However, although there may
formyl peptide receptor on human neutrophils be an initial response, subsequent flares may be
and determined that neutrophils are desensitized seen. Alternatively, the acute attack may be
to chemoattractants by annexins [512]. Work is treated with 30–60 mg prednisone or predniso-
now beginning to identify the pathways by which lone for 2 days (depending on the severity of the
intercellular processing of lipocortin occurs [505]. attack), followed by tapering the dose by 5–10 mg
Clearly, all these studies have pertinence to the every 2 days over a 10-day period [412]. Two
monosodium urate crystal-induced inflammatory randomized placebo-controlled studies have been
response since it involves phagocytosis, IL-1- and reported using oral prednisone for a 5-day period,
IL-8-mediated inflammatory reactions, and 30 mg/day in one study and 35 mg/day in the
transendothelial migration of inflammatory cells. other. Prednisolone was equivalent to standard
In addition to these lipocortin-dependent pro- courses of indomethacin compared to predniso-
cesses, glucocorticoids also inhibit vasodilation lone 30 mg/day [517] and to naproxen compared
of the microcirculation and prevent increases in to the 35 mg/day dose [518].
vascular permeability [513]. Hydrocortisone also The intra-articular injection of steroids (20–
inhibits the generation of reactive oxygen species 40 mg of prednisolone, methylprednisolone, tri-
(ROS) from mononuclear cells [514]. Finally, amcinolone, or other long-acting glucocorticoids)
annexin-1 inhibits bradykinin-induced plasma is especially effective in patients when oral medi-
extravasation in the rat knee joint [513]. Since cations are contraindicated or cannot be effectively
bradykinin is known to interact with synovial administered to a patient. Thus, patients with acute
cells, this may be another inflammatory response gout involving one or two joints after major sur-
impeded by annexins [515, 516]. gery can be effectively treated with intra-articular
It is likely that annexins play a significant role steroids. Polyarticular gout, a clinical syndrome
in the acute inflammatory responses generated by most frequently seen in older age groups, is effec-
monosodium urate crystals, and clearly, gluco- tively treated by oral glucocorticoid therapy.
corticoids can suppress such responses in the Under such circumstances, prednisone at doses
Glucocorticoids 321

beginning at 40–60 mg orally per day and tapering in the elderly, the activity of these oxidases is
over 10 days to 2 weeks is a reasonable treatment diminished by age alone. If one assumes that tar-
regimen. Low-dose intra-articular triamcinolone get organ responses are not altered in such
(10 mg) has also been used successfully to treat patients, reduced doses of prednisolone are indi-
acute gout in small- and medium-sized joints cated in these clinical settings. In hyperthyroid-
including knees, ankles, metatarsophalangeal ism, glucocorticoids are catabolized at an
joints, and wrists [519]. Clearly, such therapy is a increased rate resulting in the decreased systemic
short-term solution to what may be a recurrent availability of the active drug. Thus, hyperthyroid
problem, and other treatment regimens need to be patients may require increased doses of glucocor-
considered over the long term. ticoids. The other pharmacokinetic properties of
Glucocorticoids are absorbed rapidly and prednisone and prednisolone are shown in
attain therapeutic levels in a short period of time Tables 9.17 and 9.18 and document the short
(Table 9.17). Here again, the major risk attendant half-life of these agents. The pharmacokinetics of
to such a treatment regimen is the possible dis- intravenous methylprednisolone is shown in
semination of a microbial agent from an undiag- Table 9.19. Both prednisone and prednisolone
nosed septic arthritis presenting as a mimicker of increase tissue catabolism and may aggravate
acute gout. Delayed wound healing may also azotemia. The toxicities observed with short-term
pose a problem in steroid-treated patients. usage of these drugs include sodium retention,
Recently, mini-pulse methylprednisolone hypertension, and glucose intolerance. In severe
therapy has been used in the management of liver diseases, conversion of prednisone to pred-
patients either with chronic disabling polyarticu- nisolone, the active glucocorticoid, is slightly
lar gout or postoperative gout in which oral treat- impaired, but this altered rate of conversion is
ment may be contraindicated. In these settings, counterbalanced by a reduced catabolism of
intravenously administered methylprednisolone prednisone to its 6b-hydroxyprednisolone metab-
(100 mg in 5 % dextrose) over a 2- or 3-h time olite [520, 521]. The fraction of prednisone
period for three consecutive days is very effec- excreted in the urine as the 6b-hydroxypredniso-
tive. In the small number of patients in which this lone metabolite decreases as a function of increas-
regimen has been used, the frequency of rebound ing hepatic dysfunction [521]. Thus, hepatic
gouty attacks has been low and prevented by the 6b-hydroxylase activity is decreased in the pres-
use of maintenance colchicine in those with pol- ence of impaired hepatic function, whereas the
yarticular gout. The induction of microsomal 11b-hydroxysteroid dehydrogenase enzymes that
enzymes by other drugs (barbiturates, phenytoin, convert prednisone to prednisolone and vice versa
or rifampin) enhances the metabolism of corti- appear to be relatively well maintained in the face
costeroids to inactive metabolites and decreases of liver disease [520–522].
the effectiveness of the total dose of steroid [520]. Adrenocorticotrophic hormone (ACTH) may
In contrast, estrogens potentiate the biological be a useful means of treating gout when other
effects of prednisolone by inhibiting its drugs are contraindicated or cannot be tolerated
microsomal oxidation to 6b-hydroxylated pred- [523]. It is especially useful when oral medica-
nisolone derivatives [516]. Women taking estrogen- tions are contraindicated or when active gastroin-
containing oral contraceptives and elderly testinal bleeding, not associated with steroid
subjects (>65 years of age) have a decreased rate treatment, is the primary problem associated with
of 6b-hydroxyprednisolone formation, and such the acute gouty episode. ACTH (40 units) can be
individuals are likely to require lower doses of given every 6 h for 3 days, then every 8 h for 1
methylprednisolone or prednisolone on the basis day, and then every 12 h for 1 day. Rebound
of these changes in pharmacokinetics and drug arthritis may necessitate additional treatment, but
clearance rates [520]. Estrogens block the activ- recent studies have not found this complication
ity of microsomal liver oxidase preventing the to be a significant problem since the relapse rate
conversion of prednisolone to its catabolites, and for acute gout was similar to that observed with
322 9 Management of Hyperuricemia and Gout

Table 9.20 Pharmacokinetics of intravenous acid or urinary uric acid levels. On the other
methylprednisolone hand, ACTH has been shown to be a uricosuric
Parameters Methylprednisolone agent with a modest capacity to increase urate
5-mg dose excretion by the kidney (Table 9.21).
T ½ (h) 2.8 (13.9)
MRT (h) 4.3 (12.7)
Dose/AUC ml/h/kg 336.5 (30.2) Interleukin 1 Inhibitors
20-mg dose
T ½ (h) 2.7 (13.9)
Finally, a new class of anti-inflammatory drugs is
MRT (h) 4.3 (11.7)
being investigated for the treatment of acute
Dose/AUC ml/h/kg 330 (22.2)
gouty arthritis. The recognition of gout as an
40-mg dose
T ½ (h) 2.6 (7.6)
autoinflammatory disease that is mediated by the
MRT (h) 4.0 (7.4) interactions of sodium urate crystals with the
Dose/AUC ml/h/kg 345 (17.1) inflammasome NLRP3 in inflammatory cells has
The figures in parentheses are the coefficients of variation
indicated that urate-mediated inflammation is
in percent. These data are modified from those published dependent on IL-1 activation, as discussed in
by Szefler et al. [1049] detail elsewhere in this book. Thus, IL-1 block-
The pharmacokinetic data for prednisone, prednisolone, ing agents would be expected to be effective
and methylprednisolone show dose-dependent pharma-
cokinetics as a result of gut metabolism, first-pass metab-
agents for the treatment of acute gouty arthritis,
olism, reversible metabolism, and concentration-dependent and in fact several trials have been carried out
elimination. Methylprednisolone has a low affinity for that demonstrate the proof of this principle.
binding to transcortin and demonstrates linear nonspecific However, the role of these agents in the manage-
protein binding. For these reasons, methylprednisolone
shows more linear pharmacokinetics than either predni-
ment of gout is uncertain for several reasons.
sone or prednisolone First, there are already several effective drugs
available for the treatment of acute gout. Second,
Table 9.21 Relative contraindications to treatment with the studies of IL-1 inhibitors are preliminary and
ACTH leave questions about their effectiveness. Third,
Tuberculosis (including latent disease) many inflammatory mediators aside from IL-1
Psychiatric disorders are involved in the pathogenesis of acute gout.
Cirrhosis (sodium retention) Fourth, the IL-1 inhibitors are expensive. Finally,
Salt-dependent disorders (e.g., hypertension) their long-term toxicity has been incompletely
Hypokalemia studied [524, 525].
Infection The three IL-1 inhibitors currently available
Peptic ulcer disease (?) are anakinra, canakinumab, and rilonacept.
Studies of each of these agents in the treatment of
gouty arthritis have been published. Each of these
indomethacin treatment [523]. There are rela- agents has been approved by the US FDA for
tively few contraindications to ACTH treatment, treatment of patients with CAPS, the cryopyrin-
but the issue of aggravation of gastrointestinal associated periodic syndromes discussed else-
bleeding has not been completely resolved where in this book. Anakinra has been approved
(Table 9.20). ACTH dosage is more difficult to by the FDA for rheumatoid arthritis. Each of
regulate than orally or intravenously adminis- these agents has been demonstrated to be effec-
tered steroids. The time for complete pain relief tive in treatment of CAPS.
after a single dose of ACTH (40 units) was 3 ± 1 h, Anakinra is a recombinant form of the human
whereas after 50 mg of indomethacin four times IL-1 receptor antagonist. It differs from the native
a day, the time interval was 24 ± 10 h [523]. It is human IL-1 receptor antagonist by having a sin-
important to recognize that neither NSAIDs nor gle methionine at its amino terminus. It blocks
colchicine has any effect on either serum uric the action of IL-1b and IL-1a by competitively
Urate-Lowering Therapy: Uricosuric and Other Hypouricemic Drugs 323

inhibiting the interaction of IL-1 with the type 1 Finally, in a study of the prophylactic effects of
interleukin-1 receptor. Anakinra was shown to rilonacept, patients initiating allopurinol received
have some anti-inflammatory effects in acute and either rilonacept or placebo for 16 weeks. In this
chronic gout in a small pilot study, but clearly study, the frequency of acute gout flares was
further studies are needed [526]. reduced by as much as 80 % compared to pla-
Canakinumab is a recombinant human mono- cebo. However, efficacy of rilonacept was not
clonal antibody with specificity for human IL-1b. compared to standard regimens [528].
It belongs to the IgG1/k subclass. A single-blind In summary, the role of the IL-1 blockers in
dose-finding phase II study of canakinumab for the treatment of acute gout and for the prophy-
the treatment of acute gouty arthritis in patients laxis of acute gout during urate-lowering therapy
with intolerance to NSAIDs and colchicine was has not been established at this time. These agents
reported [527]. Five different doses of canaki- may expose patients to risks of serious infections,
numab were compared to treatment with a single and expense remains an issue. Furthermore, it is
40-mg IM dose of triamcinolone, in all patients logical to assume that they must be used early in
within 5 days of the onset of an attack of acute the course of an acute attack of gout since IL-1
gout. Patients receiving all doses of canakinumab appears to be an early mediator of gouty
were reported to have had less pain than controls inflammation. Later in the course of acute gout,
receiving triamcinolone, but the difference was other mediators become important, and these
only statistically significant with the highest dose would not be influenced by IL-1 blockers. In
of 150 mg of canakinumab. In addition, at addition, IL-1 independent mediators such as
8 weeks, patients receiving 150 mg of canaki- endothelial adherence factors and complement
numab had a 94 % reduction of recurrent gout components may play a role in acute gout.
flare, compared to the patients receiving triamci-
nolone. As pointed out by Neogi [528] in an edi-
torial accompanying this report, the findings are Urate-Lowering Therapy: Uricosuric
promising, but the study has several shortcom- and Other Hypouricemic Drugs
ings which must be considered before this drug
can be recommended as a standard practice Ultimately, the effective treatment of gout
option. The choice of comparator drug, 40 mg requires that the concentrations of urate in the
triamcinolone, has not been shown to be optimal extracellular fluid must be reduced below the
therapy. Furthermore, the timing of entry of solubility limit of monosodium urate. Since
patients into the study may not have been ideal, the solubility of monosodium urate in extracel-
risks of infection from IL-1 suppression remain a lular fluid under physiological conditions is
consideration, and the expense of the drug are all 6.8 mg/dl, the concentrations of urate in the
factors requiring further study. serum must be reduced below this level to pre-
Rilonacept is an IL-1 trap. It is a dimeric vent precipitation of sodium urate crystals in tis-
fusion protein consisting of the extracellular sues and to promote the dissolution of sodium
domains of the human IL-1 type 1 receptor with urate crystals from body tissues. Since the patho-
its IL-1 binding domain and the IL-1 receptor logical consequences of gout arise from the pres-
accessory protein both linked to the Fc portion of ence of urate crystals in tissues, primarily in and
human IgG1. Rilonacept acts as a soluble decoy around joints, these crystals must be dissolved
receptor, binding both IL-1a and IL-1b. It also from the tissues and prevented from forming.
binds the IL-1 receptor antagonist although with From a practical viewpoint, the therapeutic goal
a lower affinity than IL-1a and b. In a pilot study should be to reduce the serum urate concentra-
of acute gout, rilonacept was less effective than a tion to less than 6.0 mg/dl. The other major path-
standard regimen of indomethacin [529]. In ological consequence of urate in the body is the
another small study of chronic active gout, formation of uric acid stones in the urinary tract.
rilonacept reduced pain more than placebo [530]. Since uric acid stones are formed in the usually
324 9 Management of Hyperuricemia and Gout

Table 9.22 Criteria for treatment with uricosuric drugs Probenecid


Chronic tophaceous gout
CH3CH2CH2
Diuretic-induced hyperuricemia (serum uric acid in the
range of 8–10 mg/dl) NSO2 COOH
CH3CH2CH2
Absence of renal disease or factors known to damage
the kidney Allopurinol
Absence of uric acid/calcium oxalate lithiasis
OH
Absence of enzyme abnormalities associated with
gout N
N
Urinary uric acid excretion of greater than1 g/24 h N
Recurrent gouty episodes despite colchicine N N
prophylaxis
H
Intolerance to allopurinol
Fig. 9.7 Structures of probenecid and allopurinol and
febuxostat. Probenecid (p-(di-n-propylsulfamyl) benzoic
acid) was originally developed to sustain high blood levels
acid environment of the urine, urate deposits as of penicillin by inhibiting its renal tubular excretion.
unionized uric acid rather than ionized urate. Probenecid (Benemid) increases uric acid clearance by
Risk factors for uric acid stone formation are uri- inhibiting its reabsorption by the kidney, and low doses of
nary acidity and the quantities of urate excreted salicylate abrogate this activity. Allopurinol (4-hydroxy-
pyrazolo (3, 4-d) pyrimidine) was first synthesized for use
in the urine. Therefore, prevention of uric acid as a chemotherapeutic agent but was determined to block
stones is promoted by increased urine output and the synthesis of uric acid by xanthine oxidase. Its major
alkalinization of the urine. These considerations metabolic product oxypurinol (4, 6-dihydroxypyrozolo
should be borne in mind when choosing between (3, 4-d) pyrimidine has a long half-life (28 h) and is the
primary inhibitor of xanthine oxidase activity. Allopurinol
urate-lowering agents which either inhibit urate and oxypurinol are analogs of hypoxanthine and xanthine,
formation or promote urate excretion by the respectively
kidney.
Criteria for the use of allopurinol are reasonably
straightforward, and by exclusion, indications for N O
uricosuric drugs can be derived (Tables 9.21 and N
9.22). The structures of probenecid, sulfinpyrazone, S
O
allopurinol, and febuxostat are shown in Figs. 9.7
O
and 9.8. The pharmacokinetics and metabolism of
the uricosuric and hypouricemic drugs are shown
in Table 9.23. As can be determined by the criteria Fig. 9.8 The structure of febuxostat, a xanthine oxidase
for uricosuric and hypouricemic treatment criteria, inhibitor that is not a purine analogue
no mention has been made of the use of these drugs
in the management of asymptomatic hyperurice-
mia. The latter condition is a frequently encoun- Table 9.23 Indications for allopurinol therapy
tered laboratory abnormality, and its clinical Uric acid nephrolithiasis
significance must be carefully evaluated. As noted Heritable enzyme defects (HGPRTase and PRPP
previously, there is a multiplicity of causes for synthetase)
hyperuricemia, and asymptomatic hyperuricemic Calcium oxalate nephrolithiasis with hyperuricosuria
patients should be educated as to the associations Adenosine phosphoribosyltransferase deficiency
(complete)
between hyperuricemia and fasting, obesity, and
Chemotherapy with cytolytic agents or radiotherapy
excessive alcohol use. The indications for the treat- Gouty nephropathy (serum uric acid usually above 8
ment of asymptomatic hyperuricemia have been or 9 mg/dl)
summarized in Table 9.1 and have been discussed Allergic reactions to uricosuric drugs
in the chapter on clinical gout and hyperuricemia. Acute uric acid nephropathy
There remains the enigma as to whether patients Ineffective control of hyperuricemia by uricosuric
with significant renal impairment and asymptomatic drugs
Uricosuric Drugs: Sulfinpyrazone, Probenecid, and Benzbromarone 325

Table 9.24 Uricosuric and hypouricemic drugs: pharmacokinetics and metabolism


Probenecid Sulfinpyrazone Allopurinol
GI absorption (%) 100 rapid 100 rapid 80
Plasma half-life (h) 4–8a 2.5–3.5b 2–3c
Peak blood levels (h) 4 1 0.5–1.0
Plasma protein binding (%) 85–95 98 –
Volume of distribution (%) 0.15 ± 0.02 0.6 0.5
Excretory route Kidney Kidney Kidney
a
The half-life of probenecid is dose dependent
b
The sulfide metabolite of sulfinpyrazone has a half-life of 14.5 h
c
Seventy percent of allopurinol is converted to oxypurinol that has a half-life of 25 h

hyperuricemia should be treated with allopurinol countries abroad, benzbromarone has been used
to prevent urate-induced renal parenchymal dam- extensively as a uricosuric agent and in combina-
age. No carefully controlled studies have been per- tion with allopurinol, the xanthine oxidase inhibi-
formed to permit an objective analysis of this tor. As will be discussed subsequently,
problem. Since most chronic renal diseases prog- benzbromarone has now been withdrawn from
ress slowly and serum uric acid levels often do not the market because of its toxicity. Sulfinpyrazone
exceed 10–12 mg/dl, allopurinol therapy is not impairs platelet aggregation and prolongs platelet
indicated. However, renal function should be mea- survival; these properties make the use of this
sured periodically to ensure that renal function is drug particularly attractive in patients with gout
not deteriorating at an unacceptable rate. The labo- who are at high risk for serious cardiovascular
ratory parameters that have been used to define disorders and their platelet-mediated complica-
those patients with chronic renal disease who are at tions [531–537]. Both sulfinpyrazone and its
risk for urate-induced renal disease are probably sulfide metabolite inhibit platelet cyclooxygenase
patients with serum uric acid levels in the range of activity; the sulfide metabolite has a more potent
10–13 mg/dl, urinary uric acid to creatinine ratios and prolonged effect on prostaglandin generation
of greater than one, urinary uric acid concentra- than the parent drug [533]. The plasma half-life
tions of greater than 1,000 mg/24 h, and serum uric of sulfinpyrazone is approximately 3 h, whereas
acid increments of 2.5 mg/dl/day or greater. its sulfide metabolite has a half-life of greater
Whether patients with polycystic kidney disease than 14 h. Controversy exists as to whether such
and hyperuricemia deserve early treatment for their alterations in platelet functions have any role in
hyperuricemic state remains an unresolved ques- the prevention of thromboembolism of the myo-
tion, but there appears to be a higher frequency of cardium or brain. Studies indicating their thera-
gout in this disorder as compared with other causes peutic value in such conditions have shown that
of renal failure. Such data may provide a cogent the drug must be given in dosages of 800 mg/day
rationale for using drugs to diminish serum uric for 7 days to have clinically relevant effects on
acid concentrations in individuals with this disor- platelet function [538, 539].
der (Table 9.24). Sulfinpyrazone is also a more potent uricosu-
ric drug than probenecid on a mole for mole basis,
and studies reported three decades ago deter-
Uricosuric Drugs: Sulfinpyrazone, mined that the former agent is better tolerated by
Probenecid, and Benzbromarone patients than the latter drug [540–542]. The fail-
ure of Anturane to control hyperuricemia is usu-
Sulfinpyrazone ally related to the concomitant use of salicylates
or renal insufficiency. Salicylates antagonize the
The two uricosuric drugs generally used in uricosuric effects of probenecid and sulfinpyrazone
the United States are probenecid (Benemid) by abolishing the renal tubular elimination of
and sulfinpyrazone (Anturane). In some other urate [543, 544]. The antituberculous drug
326 9 Management of Hyperuricemia and Gout

pyrazinamide also antagonizes the uricosuric Table 9.25 Pharmacologic properties of probenecid
effects of Anturane and Benemid [543]. Probenecid is rapidly and completely absorbed
Sulfinpyrazone competes with para-aminohippu- Peak blood levels are obtained at approximately 4 h
ric acid (PAH), phenolsulfonphthalein (PSP), Uricosuric effects occur in about 40 min after
salicylates, and probenecid for their renal tubular administration of the drug
transport systems [545–548]. Thus, laboratory Serum half-life (mean) is 4–12 h
tests using PAH or PSP as agents for the measure- Probenecid is 90–95 % bound to serum albumin
Probenecid is metabolized in the liver to an
ments of specific membrane transport functions
acylglucuronide
are not accurate in the presence of these uricosu- Hydroxylated and carboxylated derivatives of
ric agents, and the competition of probenecid and probenecid are also formed
sulfinpyrazone for same of transport system
delays the excretion of the latter drug prolonging
its uricosuric effect. Sulfinpyrazone may also The only relative contraindications to the use of
potentiate the effects of sulfa drugs and oral this drug are the presence of peptic ulcer disease,
hypoglycemic agents [549]. The renal transport skin rashes, and, of course, known allergies to the
systems for uric acid are discussed in detail in drug. The recent use of benzbromarone in Europe
chapter 2. before its removal from the market may have
Sulfinpyrazone is largely excreted in the urine caused sulfinpyrazone to be used less frequently.
unchanged, and the remainder is excreted as the In addition, this drug is infrequently used for the
glucuronide conjugate of its sulfone metabolite management of gout since allopurinol is the most
(>30 %) as p-hydroxysulfinpyrazone (10 %) frequently prescribed drug for lowering the serum
and as other metabolites [550, 551]. Since uric acid levels.
sulfinpyrazone is extensively bound to plasma
proteins, it is not filtered at the glomerulus but
eliminated via the renal tubules. Its uricosuric Probenecid
properties result from interference with the post-
secretory reabsorption of uric acid via the renal The uricosuric drug probenecid has been used for
tubules. Both sulfinpyrazone and its metabolite, decades and is usually not associated with serious
p-hydroxysulfinpyrazone, are potent uricosurics toxicity. It decreases the renal excretion of many
[552]. These drugs are highly selective since they drugs by competing for a common organic acid
only interfere with the trihydroxypurine, uric secretory mechanism [552]. Its structure is shown
acid, and do not alter the excretion of the dihy- in Fig. 9.7. By this competition with other drugs,
droxypurine and monohydroxypurine, xanthine, it raises the serum levels of antibiotics (penicillin,
and hypoxanthine. Probenecid interferes with synthetic penicillins, rifampin, and dapsone), anti-
the secretion of sulfinpyrazone, whereas viral agents (acyclovir), anti-inflammatory agents
sulfinpyrazone does not block the secretion of (indomethacin, naproxen, salicylic acid), and
probenecid by the kidney [546]. Thus, the urico- diuretics (acetazolamide, furosemide, thiazides)
suric effect of sulfinpyrazone enhances the effect [552–561]. Probenecid is rapidly and completely
of probenecid. absorbed and reaches peak plasma concentrations
Sulfinpyrazone causes gastrointestinal toxicity within 4 h (Table 9.25). Its uricosuric effects begin
in approximately 10–15 % of patients, and skin early (40 min after its administration), and its
rashes occur rarely (about 3 % of patients) with half-life in plasma increases in a dose-dependent
the use of this drug. Sulfinpyrazone is a phenylb- fashion. Even though approximately 70 % of the
utazone analog, and like the latter drug, it may drug disappears from the plasma in 24 h, less than
suppress bone marrow activity. Although uncom- 5 % of the native molecule appears in the urine
mon, the toxic response may account for its [562]. The majority of the ingested probenecid is
decreased clinical usage despite its advantageous metabolized by the liver to an acylglucuronide
properties and its few clinical contraindications. and to various hydroxylated and carboxylated
Uricosuric Drugs: Sulfinpyrazone, Probenecid, and Benzbromarone 327

derivatives [562–565]. The latter derivatives are Table 9.26 Probenecid toxicity
also uricosuric. The mechanism of action of GI intolerance
probenecid is similar, if not identical, to Skin rashes
sulfinpyrazone. Like sulfinpyrazone, it does not Fever
alter xanthine or hypoxanthine excretion by the Hypersensitivity reactions
kidney. It does block the reabsorption of oxy- Flank pain and stone passage
purinol, the active metabolite of allopurinol, Acute gout
which prolongs its half-life [566]. Nonetheless, Rarely nephrotic syndrome
probenecid and allopurinol when used together
have an additive effect on the elimination of uric treated with probenecid for hyperuricemia (serum
acid and lowering serum uric acid levels [566]. It uric acid of 15.6 mg/dl) developed pancytopenia
is important to recognize that probenecid is inef- (total white blood cell count of 1,700 cells/mm3,
fective as a uricosuric when the glomerular hemoglobin of 7.1 g/dl, and a platelet count of
filtration rate is less than 50 ml/min [567]. Acute 13,000 cells/mm3) [464]. A bone marrow aspirate
gouty arthritis and uric acid nephrolithiasis are the from this patient showed marked hypocellularity,
most common complications of probenecid ther- most apparent in the myeloid elements (M:E ratio
apy. The frequency of acute gout in association of 1:5). The bone marrow also showed megalo-
with probenecid therapy can be significantly blastic changes with an MCV of 90. Cessation of
reduced by the concomitant administration of all medications including probenecid and the use
colchicine or indomethacin. Nephrolithiasis sec- of platelet and packed cell transfusions as well as
ondary to probenecid can be prevented by initiat- the administration of folic acid (1 mg/day) led to
ing treatment with probenecid at a low dose, the recovery of the bone marrow. Such a catastro-
encouraging a high fluid intake (about 2–3 l/day), phe emphasizes the need for documenting drug-
and using sodium bicarbonate (2–6 g/day) to drug interactions prior to the use of any drug in
increase the urine pH and the solubility of uric patients and the adherence to appropriate criteria
acid in the urine. Similar complications can occur for the clinical indications for such drugs like
with sulfinpyrazone therapy and can be controlled allopurinol and probenecid. It is worth pointing
in an identical manner. out that gout is extremely rare in association with
Drug toxicity from probenecid includes gas- established rheumatoid arthritis.
trointestinal side effects, hypersensitivity reac- Standard treatment regimens for Benemid
tions to the drug, skin rashes, and drug-induced begin with a dose of 250 mg twice a day for 1
fever (Table 9.26), and its pharmacologic proper- week increasing the dose to 500 mg twice a day
ties are shown in Table 9.25. Such toxic reactions to attain the appropriate reduction in serum uric
are not common, only occurring in about 10 % of acid levels. As much as 2 g of Benemid may be
patients, and there are relatively few contraindi- administered as 100- or 200-mg doses twice a
cations to the use of probenecid such as peptic day for 1 week increasing the dose to 200 mg
ulcer disease, skin rashes (penicillin-like), neph- four times a day if necessary to control hyperuri-
rotic syndrome, and epilepsy. More serious reac- cemia (Table 9.26).
tions to this drug such as the nephrotic syndrome
and hepatocellular damage are rare.
One serious complication of probenecid ther- Salicylates
apy is its interaction with methotrexate [568].
Probenecid potentiates methotrexate toxicity by Salicylates are potent uricosuric drugs at high
inhibiting the renal tubular excretion of metho- doses (4–5 g/day), but few patients are able to
trexate and its metabolites, by displacing metho- tolerate these doses. The low serum urate levels
trexate bound to plasma proteins and by inhibiting seen in the past when patients with rheumatoid
methotrexate secretion into the bile [569–571]. A arthritis were treated with high-dose salicylates
single patient with rheumatoid arthritis who was were the result of the uricosuric properties of
328 9 Management of Hyperuricemia and Gout

acetylsalicylic acid and its derivatives. Aspirin zarone and benzarone are conjugated to glucuronic
should only be prescribed when toxicity prevents acid and excreted in the bile (80 %) and urine
the use of all other antihyperuricemic drugs, and (20 %). Although the serum half-life of benzbro-
such situations probably almost never occur. marone is roughly 12 h, enterohepatic recircula-
Since many individuals use aspirin as a preventa- tion of its dehalogenated metabolite, benzarone,
tive measure for cardiac insults and for many permits the detection of this metabolite for over
other medical conditions including over-the- 48 h [575]. The drug is strongly bound to plasma
counter preparations, any effect of very low-dose proteins. A variety of benzbromarone metabo-
aspirin may have a negative impact on renal func- lites have been isolated from the urine and their
tion and alter the 24-h urinary uric acid excretion. structure characterized [576–579].
A recent study has demonstrated that aspirin at The hypouricemic and uricosuric properties of
doses of less than 500 mg/day results in a benzbromarone were first reported in the 1980s,
significant decrease in the rate of uric acid excre- and subsequently a large number of publications
tion (~ 15 % decrease in the rate of uric acid have confirmed this drug’s role in the manage-
excretion) with a concomitant elevation in serum ment of gout [580–587]. The mechanism of
uric acid levels in an elderly population (ages action of benzbromarone is via the selective inhi-
61–94) [572]. These effects were more pro- bition of tubular reabsorption of uric acid by the
nounced in hospitalized patients that had low renal tubules. Recent evidence has demonstrated
serum albumin concentrations (<3 g/dl) and those the binding of benzbromarone to the urate/anion
receiving long-term diuretic therapy (furosemide exchanger at the brush-border membrane vesicles
20–40 mg/day). Even though very low-dose aspi- of the proximal renal tubules in humans [588]. It
rin does not cause dramatic changes in renal has been used extensively in Europe until recently
function or serum uric acid levels, such data are in the treatment of gout and hyperuricemia and
worth considering when assessing renal function, has been most effective in certain special clinical
urinary uric acid concentrations, and serum urate settings. Benzbromarone is an effective alterna-
levels. tive drug for the reduction of serum urate levels
In addition to probenecid and sulfinpyrazone, in patients who are allergic to allopurinol [589,
other uricosuric drugs are being developed. For 590]. The drug is especially useful in patients
example, during the treatment of patients with who develop cyclosporine-induced hyperurice-
HIV infection with the nonnucleoside reverse mia after undergoing organ transplantation [591].
transcriptase inhibitor RDEA806, this agent was Two published benzbromarone/benziodarone-
noted to have uricosuric effects due to inhibition treated patients have documented the success of
of the renal tubular uric acid transporter URAT 1 this agent in organ-transplant patients [591, 592].
[573]. In one series of 25 patients, 100 mg of benzbro-
marone decreased the serum uric acid levels from
mean values of 9.72 ± 0.3–5.26 ± 0.4 mg/dl with
Benzbromarone no side effects, and in another series of 20
patients, 50–100 mg of benziodarone reduced the
Benzbromarone is a halogenated benzofuran mean serum uric acid levels from 10.1 ± 1.8 to
derivative with potent uricosuric properties avail- 5.1 ± 1.4 mg/dl without significant adverse reac-
able in Europe until very recently. Micronized tions [591]. In these cases, allopurinol was con-
benzbromarone is rapidly absorbed attaining traindicated because of its interaction with
peak blood levels in 2–3 h [574]. The drug is azathioprine, whereas benzbromarone at a dose
dehalogenated in the liver to bromobenzarone of 100 mg/day normalized the serum uric acid
after the removal of a single bromine atom and level and increased the urinary uric acid excre-
subsequently to benzarone with both bromine tion without any effect on azathioprine. It is valu-
atoms removed. About 60 % of the native mole- able to reemphasize the rapidity with which
cule is absorbed, and derivatives of bromoben- tophaceous gout can develop in organ-transplant
Xanthine Oxidase Inhibitors: Allopurinol and Febuxostat 329

patients taking cyclosporine and the unusual a dose of 100 mg of benzbromarone has been
clinical presentations such patients may have shown to reduce serum uric acid levels more than
[593–596]. The drug has also been used success- 1 g of Benemid or 300 mg of allopurinol [609].
fully in the treatment of familial juvenile gouty Thus, this drug appears to be an efficacious agent
nephropathy and in a small number of patients for patients with hyperuricemia and gout in which
with renal insufficiency [585, 597]. The drug is uricosuric agents are indicated and is especially
only effective as a uricosuric agent when the cre- useful in those patients who have undergone organ
atinine clearance is greater than 20 ml/min [598]. transplantation and are receiving cyclosporine.
Combination drugs containing allopurinol and As noted previously, all this data concerning
benzbromarone have also been available in benzbromarone are moot since its toxicity has
Europe, but they do not appear to confer any dis- resulted in its removal from the European and US
tinct advantages over the use of single agents markets; however, the drug is still available in
[599]. As might be expected, the combination of some countries, and its properties make it a desir-
drugs is more effective in lowering the serum uric able agent for the treatment of some types of
acid levels than allopurinol alone [600, 601]. One gout. Therefore, its structure may provide a
report in the literature documented a concomitant model for other modified drugs that might be
reduction of serum lipids (triglycerides and cho- synthesized in the future. Benzbromarone is a
lesterol) and serum uric acid during benzbroma- benzofuran derivative that reduces the proximal
rone use [602]. tubular reabsorption of uric acid [610].
Benzbromarone is a potent competitive inhibi- Benzbromarone and its metabolite benzarone
tor of the 7-hydroxylation of the anticoagulant have a similar structure to amiodarone, and the
S-warfarin [603]. Thus, benzbromarone prolongs latter drug causes toxicity to liver mitochondria
the anticoagulant action of S-warfarin. [610–612]. Recent studies have also shown that
Pyrazinamide and low-dose aspirin abrogate the benzarone and benzbromarone inhibit the mito-
uricosuric effect of benzbromarone, and the latter chondrial respiratory chain and b-oxidation. They
drug appears to have no effect on the elimination also uncouple phosphorylation [613]. These
of penicillin [604–606]. It is also has little or no agents also trigger reactive oxygen species pro-
effect on the diuresis induced by thiazide drugs duction and mitochondrial swelling that leads to
[604–606]. Like other uricosuric drugs, benzbro- apoptosis and cellular necrosis. The use of benz-
marone is ineffective at low glomerular filtration bromarone can lead to severe hepatotoxicity and
rates (<25 ml/min) [607]. Like other uricosuric death [614–616]. In view of these life-threatening
drugs, the most common side effects are the pre- toxicities, benzbromarone was withdrawn from
cipitation of an acute episode of gouty arthritis or the European market in 2003.
the formation of uric acid calculi [606, 608].
Utilizing colchicine prophylaxis, using low doses
of benzbromarone at the outset of therapy, Xanthine Oxidase Inhibitors:
encouraging high fluid intake, and alkalinizing Allopurinol and Febuxostat
the urine can easily avoid these complications.
Gastrointestinal intolerance occurs in about 10 % Allopurinol
of patients taking the drug, and skin rashes are
rarely observed [583]. The hypoxanthine analog allopurinol inhibits the
Benzbromarone at a dose of 40 mg is equiva- conversion of the oxypurines hypoxanthine and
lent to 1 or 1.5 g of Benemid or 400–800 mg of xanthine to uric acid [617–619]. Both allopurinol
Anturane [606]. A single dose of 80 mg of benz- and its oxidative product alloxanthine (oxy-
bromarone causes a reduction of serum uric acid purinol) inhibit xanthine oxidase activity. Both
levels by one-third to two-thirds [582, 606]. these drugs have structures similar to their natu-
Benzbromarone at 40 mg/day for 1 week reduced ral purine analogs, hypoxanthine and xanthine,
serum uric acid levels by one-third [606]. Finally, except for the reversal of the carbon and nitrogen
330 9 Management of Hyperuricemia and Gout

atoms at position 7 and 8 of the purine ring Table 9.27 Pharmacologic properties of allopurinol and
(Fig. 9.6). Allopurinol is a competitive inhibitor oxypurinol
of xanthine oxidase since its structure is almost Peak serum concentrations occur in 2–3 h for
identical to the natural substrate of this enzyme, allopurinol or oxypurinol
hypoxanthine [620]. Allopurinol is a much more Neither drug is protein bound
Oral allopurinol is easily absorbed (~ 80 %)
potent inhibitor of xanthine oxidase activity (five-
Oral oxypurinol is absorbed less easily (~ 40 %)
to ten-fold) than oxypurinol in vitro, but this
Allopurinol has a half-life of 2–3 h
mechanism may not be operative in vivo since
Oxypurinol has a longer half-life of 18–33 h
oxypurinol appears to be the major xanthine oxi- Allopurinol is filtered and excreted by the kidney
dase inhibitor on the basis of its prolonged half- Oxypurinol is filtered and reabsorbed by the kidney
life (>25 h) [621]. In addition to the decrease in Minor metabolic products include allopurinol and
uric acid synthesis via the xanthine oxidase path- oxypurinol ribonucleosides and their corresponding
way, these hypouricemic agents also inhibit nucleotides
de novo purine biosynthesis presumably as a
result of a combination of feedback inhibition by
adenine and guanine nucleotides on the first irre- [23]. In contrast, oxypurinol has a much longer
versible step in de novo purine biosynthesis half-life than allopurinol and is filtered at the
(phosphoribosylpyrophosphate amidotransferase) glomerulus and reabsorbed by the tubules such
and the depletion of its substrate phosphoribo- that it is cleared at a rate that is only 10–20 % of
sylpyrophosphate (PRPP) [622–624]. There is an the glomerular filtration rate [23]. Diminished
absolute requirement for the enzyme hypoxan- renal function decreases oxypurinol clearance,
thine-guanine phosphoribosyltransferase since it whereas uricosuric agents enhance its clearance
converts free purine bases and allopurinol into [23]. Since 6-mercaptopurine and azathioprine
their respective nucleotides that are known to be are inactivated by oxidation via the xanthine oxi-
feedback and pseudofeedback inhibitors, respec- dase pathway, allopurinol therapy significantly
tively, of de novo purine biosynthesis [625, 626]. increases the activity of these antimetabolites by
Both allopurinol and oxypurinol also alter delaying their breakdown to inactive metabolites
pyrimidine synthesis by inhibiting orotidine-5- [618]. The activity of cyclophosphamide is also
monophosphate decarboxylase causing an enhanced by allopurinol: The mechanism for this
increase in the urinary excretion of orotidine and latter effect is poorly understood, but it may be
orotic acid [623, 626–628]. These effects on related to an effect on pyrimidine metabolism
pyrimidine metabolism have not been associated [627, 628]. Allopurinol also blocks hepatic drug-
with any adverse effects, but their interference metabolizing enzymes and slows the degradation
with pyrimidine metabolism may play a role in of drugs such as antipyrine and bishydroxycou-
the increased bone marrow toxicity of cyclophos- marin (dicoumarol) that are metabolized via the
phamide observed in patients taking allopurinol microsomal enzyme system [632]. Drugs metab-
[629, 630]. Both allopurinol and oxypurinol have olized by this pathway should not be used in con-
also been used as therapeutic agents, and they junction with allopurinol. Reduced drug dosage
reach peak serum concentrations rapidly and careful monitoring of in vivo drug levels
(Table 9.27). Oxypurinol is not absorbed as well should be undertaken if allopurinol and other
as allopurinol (80 versus 40 %), and higher doses drugs metabolized by these microsomal hepatic
(twofold) of the former drug must be used to enzymes are being used in combination.
achieve changes in the serum and urinary uric Standard treatment regimens for allopurinol
acid levels comparable to those observed when use 100 mg of the drug daily for 1 week and then
allopurinol is used [23, 631]. The conversion increasing the dose to 300 mg daily that may be
of allopurinol to oxypurinol in vivo is rapid (1–2 given in a single dose. In severe tophaceous gout,
h), and the rate of allopurinol excretion by the 400–600 mg of the drug may be necessary to
kidney approaches the glomerular filtration rate control the metabolic derangement in uric acid
Xanthine Oxidase Inhibitors: Allopurinol and Febuxostat 331

metabolism. Prophylactic colchicine at a dose of for the determination of the range of concentra-
0.5–0.6 mg twice a day is recommended when tions causing adverse reactions [644, 645]. The
allopurinol therapy is being initiated. long-term use of this combined therapy has not
In general, the activity of allopurinol as a xan- been evaluated, and the possible adverse reactions
thine oxidoreductase inhibitor is mediated in part from such a treatment regimen remain to be
by its active metabolite oxypurinol. Nonetheless, assessed.
allopurinol dosage should be individualized to Several potential clinical complications may
attain a sufficient decrease in plasma urate con- occur as a result of allopurinol therapy. These
centrations, and such adjustments may require a include the precipitation of acute gouty arthritis
dose higher than the recommended daily dose of as a result of the rapid changes in the mobilization
300 mg/day. When this strategy is selected, care- of tissue secondary to the reduction in serum uric
ful monitoring of patients must be done to reduce acid levels. Both prophylactic colchicine and low
the chance of adverse reactions, especially to pre- initial doses of allopurinol reduce the frequency
vent xanthine/oxypurinol stone formation or of such gouty episodes. Urinary oxypurine excre-
allopurinol hypersensitivity responses [633, 634]. tion increases significantly in the presence of
Perhaps, a less hazardous regimen would be the allopurinol, and the relative insolubility of the
use of allopurinol in combination with probenecid. increments in serum xanthine levels may cause
Such a regimen has been evaluated in both healthy xanthine renal calculi or crystalluria. Oxypurinol
humans and those with gout [635, 636]. In healthy has also been identified with renal stones and uri-
humans, allopurinol (300 mg/day) and probenecid nary tract sludge in rare patients [646, 647]. The
(1,000 mg/day) had a greater hypouricemic effect dioxygen oxidative product of allopurinol, oxy-
than either drug alone. This occurred even though purinol, is also insoluble and has been demon-
the combination of these drugs reduced the steady- strated along with hypoxanthine and xanthine in
state oxypurinol concentration (5.1 ± 1 mg/l) more muscle biopsy specimens from patients treated
than allopurinol alone (9.7 ± 2.1 mg/l). In the with allopurinol after its initial release as a thera-
gouty patients under treatment, the goal was to peutic agent [648]. No specific clinical problems
attain a serum uric acid level of less than were related to the changes observed in these
0.36 mmol/l since recurrent gouty episodes are biopsies, and subsequent use of this drug has not
better prevented at this level of serum uric acid, been associated with such muscle pathology. In
and tophaceous deposits are dissolved more read- elderly patients, the elimination of the allopurinol
ily [637–643]. Of the 32 patients treated with metabolite oxypurinol is reduced probably as a
allopurinol alone at a mean dose of 256 mg/day, result of diminished renal function in the aged
only 25 % attained a serum uric acid level of less population [649]. This metabolite also appears to
than 0.30 mmol/l, and only 53 % reached a level have less inhibitory action against xanthine oxi-
of less than 0.36 mmol/l. In the 14 patients receiv- dase in the elderly. The resultant increase in oxy-
ing both drugs with the mean dose of allopurinol purinol levels in the elderly has been proposed as
of 243 mg/day and probenecid at 1,000 mg/day, a mechanism for the increased allopurinol toxic-
86 % attained a serum uric acid of less than ity in older patients and perhaps those patients
0.30 mmol/l and 100 % achieved a level of less with renal insufficiency.
than 0.36 mmol/l. Thus, these data suggest that Even though allopurinol is well tolerated and
the combination of allopurinol and probenecid useful as a therapeutic agent in certain clinical
might be an effective way to reduce serum uric settings, it is important to emphasize that in a
acid levels and prevent recurrent acute gouty epi- series of patients who manifested serious toxic
sodes in patients with no contraindications to uri- reactions, more than 20 % died [650]. For this
cosuric drugs. Since simple methods are available reason, therapy with this drug should not be
to monitor allopurinol and oxypurinol in human undertaken without appropriate indications.
serum, this methodology might be very useful for Toxic reactions such as gastrointestinal symp-
the detection of the toxic levels of these drugs and toms, skin rashes, hypersensitivity of the
332 9 Management of Hyperuricemia and Gout

Table 9.28 Relative contraindications to allopurinol than 20 % of the patients for whom allopurinol
therapy was prescribed had a definite indication for the
Anticoagulant therapy use of the drug. By definition in this study, neph-
Skin rashes rolithiasis, chronic renal insufficiency, idiopathic
Asthma (theophylline) uric acid overproduction, tophaceous gout, pso-
Antineoplastic agents (6-mercaptopurine/azathioprine) riasis, or hematologic cancers represented the
Renal failure
disorders for which allopurinol should have been
Leukopenia
prescribed. However, over 80 % of the 254
Hepatocellular diseases
patients were treated with the drug without
fulfilling these defined criteria. Of even greater
Stevens-Johnson type, alopecia, bone marrow significance, joint aspiration was performed in
suppression, hepatocellular dysfunction, and vas- approximately 4 % of the patients, only 20 % of
culitis occur in about 10 % of patients taking the patients had radiographic studies to charac-
allopurinol. Such reactions are most frequently terize tophaceous deposits, and just 3 patients of
observed in patients with renal insufficiency and the 209 patients that did not have a definite indi-
in association with thiazide drug use [650]. Skin cation for allopurinol had their 24-h urinary uric
rashes seem to be more common in patients receiv- acid levels measured. More than 50 % of the
ing allopurinol together with ampicillin [651]. patients treated with allopurinol have serum crea-
Allopurinol dose reduction and symptomatic tinine levels documented prior to their treatment.
treatment may cause an abatement of skin rashes. As one might suspect, many of the patients treated
There are certain relative contraindications to had diuretic-induced hyperuricemia which in
the use of the drug (Table 9.28). Discontinuance some cases was associated with obesity. Such
of allopurinol is a necessity when skin manifesta- data, if they are representative of the prescribing
tions such as toxic epidermal necrolysis and/or habits of physicians in general, demonstrate a
vasculitis occur. In view of the increased morbid- deficiency in physician education as to the proper
ity and mortality associated with allopurinol use of allopurinol and place the recipient patient
hypersensitivity reactions, there is a need to iden- at increased risk for unnecessary adverse drug
tify patients at risk for this syndrome. Recent data reactions. Further, the increased cost of prescrib-
suggest that intradermal skin challenge and lym- ing a drug for which there is no clear indication
phocyte transformation with allopurinol or oxy- or therapeutic rationale seems inappropriate to
purinol may detect those patients at risk for say the least.
hypersensitivity reactions to these drugs [652]. On rare occasions, patients with severe topha-
Further, six of these nine patients had significant ceous gout and normal renal function may be
blastogenic responses when their mononuclear candidates for a combined therapeutic program
cells were exposed in vitro to oxypurinol but not using allopurinol and Benemid. Allopurinol
allopurinol. Despite the positive results from extends the half-life of Benemid and potentiates
these in vitro tests, further studies of this method- its uricosuric effect, whereas Benemid increases
ology showed that this testing did not identify all the clearance of oxypurinol diminishing the inhi-
those patients with hypersensitivity to allopurinol bition of xanthine oxidase [608, 655]. These
or its metabolite. A modest increase in the risk mechanisms would suggest that the usual doses
for cataracts has been determined in patients who of allopurinol should be increased, whereas stan-
have used allopurinol for more than 3 years or at dard doses of Benemid should be decreased.
a dose of more than 400 mg/day [653]. Nonetheless, such combined drug therapy is more
To emphasize the importance of using defined effective than either drug alone in reducing serum
criteria when allopurinol treatment is contem- uric acid levels.
plated, a hospital-based Veterans Administration Hypersensitivity reactions deserve some fur-
study determined the frequency of the inappro- ther discussion because recent findings may assist
priate use of allopurinol [654]. In that study, less the physician more directly in the detection of at
Xanthine Oxidase Inhibitors: Allopurinol and Febuxostat 333

risk patients [656]. Since the first description of Table 9.29 Allopurinol hypersensitivity: diagnostic
allopurinol hypersensitivity was reported in 1970, criteria
more than 100 cases of this syndrome have been Major criteria
recorded in the literature [657]. This condition is Diffuse maculopapular rash, exfoliative dermatitis,
said to be relatively infrequent since there are an erythema multiforme, toxic epidermal necrolysis
Decreasing renal function/acute hepatic toxicity
estimated 250 million doses of allopurinol pre-
Minor Criteria
scribed annually as calculated for the year 1985
Fever
[658]. Recent calculations indicate that between
Eosinophilia/leukocytosis
2 and 5 % of the patients prescribed allopurinol Lack of exposure to drug that causes a hypersensitiv-
develop an adverse cutaneous reaction [659, 660]. ity reaction
More recent publications have reported the fre-
quency of the Stevens-Johnson syndrome (STS)
Table 9.30 Symptoms and signs of possible severe cuta-
and toxic epidermal necrolysis (TEN) have neous reactions
increased with respect to the use of allopurinol
Confluent erythema
[661]. Severe adverse cutaneous reactions such as
Facial edema
the allopurinol hypersensitivity syndrome occur
Painful skin
in about 0.4 % of patients receiving allopurinol Palpable purpura
[662]. However, the major issue with respect to Necrotic skin
allopurinol and hypersensitivity reactions is the Blisters
mortality rate that can reach a level of 26 % of Skin detachment
patients being treated with allopurinol [657, 658]. Mucous membrane erosions
In addition to STS and TEN, there is another drug Urticaria
hypersensitivity syndrome associated with Swollen tongue
allopurinol called drug rash with eosinophilia and Lymphadenopathy
systemic syndrome (DRESS) [663–666]. This Fever (>40 °C)
rare idiosyncratic reaction is also associated with Arthralgias/arthritis
a high mortality rate (10 %) [667–669]. Wheezing
Cutaneous drug eruptions or allergic reactions Dyspnea
to drugs are common in both hospitalized patients Hypotension
Eosinophilia (>1,000/mm3)
and outpatients [670–673]. The major diagnostic
Lymphocytosis with atypical lymphocytes
effort with respect to these skin reactions should
Abnormal liver function tests
be on the early recognition of severe drug reac-
Abnormal renal function
tions and the withdrawal of the causative drug.
Most drug reactions present as morbilliform or
exanthematous rashes and may be confined to the Table 9.31 General manifestations of a drug hypersensi-
tivity syndrome
trunk or pressure areas. A number of factors
should alert the clinician to the possibility of a Fever and rash
severe cutaneous reaction (Table 9.29). The two Hepatitis
most common severe cutaneous reactions are the Mucous membrane lesions
Stevens-Johnson syndrome and toxic epidermal Lymphadenopathy
Facial edema
necrolysis [674–676]. These two conditions have
Renal involvement
now been determined to be components of a sin-
Pulmonary symptoms or signs
gle disease with common causes and mechanisms
[667, 677–680]. The principal difference between
these two disorders is the extent of the skin frequent clinical findings of these disorders are
detachment since it is limited in the Stevens- shown in their decreasing order of their frequency
Johnson syndrome but much more extensive in in Table 9.30. Common signs of a drug hypersen-
toxic epidermal necrolysis [667, 679]. The most sitivity syndrome are recorded in Table 9.31.
334 9 Management of Hyperuricemia and Gout

The original report of Stevens-Johnson syn- raises the issue of age and the clearance of oxy-
drome was described in patients with febrile ero- purinol since decreased renal function decreases
sive stomatitis, severe ocular involvement, and oxypurinol clearance by the kidney, and renal
disseminated, discrete dark red macules with an function is often diminished in elderly popula-
occasional necrotic center [681]. Much later toxic tions [698]. Since diminished renal function may
epidermal necrolysis was described with exten- prolong the half-life of oxypurinol and increase
sive loss of the epidermis that gave the underly- the risk of hypersensitivity, creatinine clearance
ing skin a scalded appearance [682]. Mucosal should probably be measured in patients who are
lesions are common in both disorders and may candidates for allopurinol treatment [699].
lead to painful micturition, photophobia, and
difficulty swallowing [673–685]. As can be imag-
ined, with extensive skin loss, there is massive Determination of Renal Function
fluid loss (3–4 l/day), electrolyte imbalance, and
prerenal azotemia [676]. Skin biopsies can dif- Estimating creatinine clearance can be accom-
ferentiate these two conditions from bullous dis- plished by the Cockcroft and Gault formula as
orders unrelated to drugs, and infection is a defined here. Creatinine clearance (ml/min) is
common complication of Stevens-Johnson syn- equal to 1.2 × (140 – age in years) × weight (kg)/
drome and toxic epidermal necrolysis. serum creatinine (umol/l). This formula calcu-
Although most drug reactions appear to occur lates creatinine clearance for males, and when
as manifestations of T cell-mediated delayed females are to be assessed, the result should be
hypersensitivity with the production of cytokines, multiplied by 0.85 [700]. Some investigators
however, the complete pathogenesis of such reac- have also indicated that the use of thiazide and
tions remains incomplete [686–688]. It is also allopurinol together may prolong the half-life of
now apparent that genetic factors play a role in oxypurinol, the metabolite of allopurinol [697,
such adverse drug reactions [689–695]. It has 701, 702]. Other literature emphasizes that thiaz-
been established in some patients with allopurinol ides have no effect on allopurinol in the presence
hypersensitivity that lymphocytes from hyper- of normal renal function [703–705]. Renal
sensitive patients are moderately stimulated by insufficiency may occur in the face of allopurinol
allopurinol (increased 3H-thymidine uptake), and therapy. Acute interstitial nephritis, focal seg-
oxypurinol markedly stimulated lymphocytes mental glomerulonephritis, and vasculitis have
from hypersensitive patients when compared to all been described in patients treated with
allopurinol-treated patients without hypersensi- allopurinol [706–709].
tivity and normal subjects [688]. Such testing has The primary challenge for these severe cuta-
also been found in patients who are hypersensi- neous reactions is to determine a method for
tive to drugs other than allopurinol [687]. Despite detecting which patients might be at risk for
these lymphocyte tests, this methodology is not allopurinol hypersensitivity. Early studies recog-
diagnostic of drug hypersensitivity and should nized that T cells were a primary route for drug
not be used to detect hypersensitive patients. recognition, and subsequently, they may trigger
There are some distinct clinical features of hypersensitivity [710]. These studies also showed
allopurinol hypersensitivity. Renal dysfunction is major histocompatibility complex relationships
a prominent abnormality observed with in drug-induced hypersensitivity.
allopurinol hypersensitivity as compared to other The presence of two major criteria or one
causes of drug hypersensitivity (84 % of patients major and one minor criterion makes the diagno-
with allopurinol hypersensitivity versus 40 % of sis of allopurinol hypersensitivity
patients with other drug hypersensitivities in one Recently, studies have reported strong associ-
study) [696]. Elevated levels of oxypurinol have ations between histocompatibility locus antigen
also been correlated with the risk of the develop- (HLA) molecules and drug-induced hypersensi-
ment of allopurinol hypersensitivity [697]. This tivity reactions [711–717]. HLA-A29, HLA-B12,
Xanthine Oxidase Inhibitors: Allopurinol and Febuxostat 335

and HLA-DR7 are observed more frequently in for a high-risk patient for the development of a
patients with sulfonamide-induced Stevens- severe cutaneous reaction to allopurinol, but its
Johnson syndrome, and HLA-A2 and HLA-B12 presence should not obviate the use of allopurinol
are found more frequently in NSAID-induced since many allopurinol-tolerant patients express
Stevens-Johnson syndrome [711, 712]. When eye this allele as well.
involvement occurs in Stevens-Johnson syn- Clearly, patients who manifest a toxic response
drome, the HLA-B59 allele is frequently observed to allopurinol should have the drug withdrawn,
[713]. Carbamazepine-induced Stevens-Johnson delivered supportive therapy for their condition,
syndrome is often associated with HLA-B*1502 and monitored carefully for possible organ fail-
[715]. Abacavir hypersensitivity is associated ures. Recent retrospective studies in Europe
with HLA-B*5701 as well as a haplotype Hsp (France and Germany) provided data to show that
70-Hom variant [716–718]. HLA-AW33 and neither intravenous immunoglobulins nor corti-
HLA-B17/BW58 have been found frequently in costeroids had beneficial effects on Stevens-
association with allopurinol drug eruptions [714]. Johnson syndrome or toxic epidermal necrolysis
However, the most striking HLA association with when compared to patients receiving supportive
a severe cutaneous adverse reaction and care [723]. Even though corticosteroids are
allopurinol treatment is the strong association of administered to patients with drug-induced severe
HLA-B*5801 in the Han Chinese of Taiwan cutaneous reactions, the beneficial effects of such
[656]. This allele was found in 100 % of Chinese agents remain a question.
affected with allopurinol-induced hypersensitiv- Patients who are either allergic to uricosuric
ity syndrome, Stevens-Johnson syndrome, and agents or have reduced renal function may be
toxic epidermal necrolysis. The calculated odds candidates for allopurinol therapy. When the
ratio in this population exceeds that for the pres- glomerular filtration rate is less than 30 ml/min,
ence of HLA-B27 and ankylosing spondylitis uricosuric agents are ineffective, and allopurinol
[719, 720]. The HLA-B*5801 allele also exists at is the alternative. Similarly, when uricosuric
a frequency of less than 10 % in African, drugs fail to reduce the serum urate concentra-
Caucasian, and Asian Indian populations [721]. tions to less than 7 mg/dl in patients with normal
Although HLA-B*5801 represents a striking renal function, allopurinol may be added to the
association with severe adverse cutaneous reac- treatment regimen.
tion in a Taiwanese Chinese population, it is not
as high in association with skin reactions in other
populations sensitive to allopurinol [656, 722]. Newer Urate-Lowering Drugs
HLA-B*5801 was present in 15 % of the
allopurinol-tolerant subjects, and it was also Febuxostat
observed in 20 % of the general population. Thus, The most well-evaluated xanthine oxidase inhibi-
in this Chinese population, HLA-B*5801 is tor is allopurinol, but recently, several nonpurine
sufficient to be closely associated with severe inhibitors of xanthine oxidase have been
cutaneous responses to allopurinol, but it is not described. In a rat model, a group of 1-phenylpyra-
the only factor involved since some HLA- zoles have shown xanthine oxidase inhibitory
B*5801-positive subjects are allopurinol tolerant. activity [725–729]. The compound showing the
In Europeans, the HLA-B*5901 allele was asso- most potent activity in this capacity in rats was
ciated with Stevens-Johnson syndrome or toxic chosen for further evaluation in the human. The
epidermal necrolysis and allopurinol treatment molecule fitting this criterion was 1-(3-cyano-4-
only at a level of 55 %. Therefore, the HLA- neopentyloxyphenyl) pyrazole-4-carboxylic acid
B*5801 allele is not as strong a risk factor in this (Y-700), and it causes long-lasting hypouricemic
population as in the Taiwanese population. Thus, effects and is excreted primarily via the liver
it would appear that in the Taiwanese Chinese, [701–703]. In studies of healthy Japanese volun-
the HLA-B*5801 allele might be used as a marker teers, this compound was rapidly absorbed after
336 9 Management of Hyperuricemia and Gout

oral administration and had a half-life of 20–40 Gouty arthritis flares occurred during treatment
h. Excretion in the urine was less than 1–1.5 % of with this drug, and such episodes were reduced
the ingested dose. Adverse effects included by the administration of colchicine. Adverse
abdominal cramps, abdominal pain, and effects occurring with this agent have a low fre-
flatulence. Serum uric acid levels were reduced in quency, but abnormalities in liver function tests
a time- and dose-dependent manner. Its principal (primarily transaminases), diarrhea, gastrointes-
route of metabolism is by the liver with relatively tinal complaints, and headaches do occur [736,
little excreted by the kidney. This makes it an 740]. Since allopurinol alters the activities of
ideal agent for patients with renal disease. other enzymes involved in purine and pyrimidine
Febuxostat (2-[3-cyano-4-isobutoxphenyl]-4- metabolism, it was important to assess febuxostat
methylthiazole-5-carboxylic acid) is a nonpurine for its activities against these enzymes as well
selective inhibitor of xanthine oxidase and is a [696, 730, 736, 741]. Examining the effect of
potent inhibitor of both the oxidized and reduced febuxostat on guanine deaminase, hypoxanthine-
forms of xanthine oxidase [730]. Its structure is guanine phosphoribosyltransferase, orotate phos-
shown in Fig. 9.8. This agent is more potent, and phoribosyltransferase, orotidylic acid
its effects are of longer duration than allopurinol decarboxylase, and purine nucleoside phospho-
[730–736]. Studies of the crystal structure of the rylase activities demonstrated no change in
xanthine oxidase-febuxostat (enzyme-inhibitor) enzyme activity in the presence of febuxostat
complex and the mechanism of inhibition showed concentrations of 1, 10, and 100 mm.
a mixed type of inhibition with Ki and Ki ‘values In summary, febuxostat is a more selective
of 1.2 ± 0.05 × 10−10 and 9.0 ± 0.05 × 10−10, respec- inhibitor of xanthine oxidase than allopurinol and
tively [730]. Febuxostat inhibits the enzyme for its metabolites. It diminishes serum and urinary
long time periods, whereas oxypurinol, the uric acid levels in the dose range of 40–80 mg/
allopurinol metabolite that inhibits xanthine oxi- day. It has few side effects in the human and is
dase activity, is relatively rapidly reactivated with useful in patients with decreased renal function
a t1/2 time of 300 min at 25 °C. Furthermore, since the kidney plays only a very minor role in
febuxostat and its metabolites have been studied febuxostat metabolism [738, 739]. In a study
in humans with mild (CCR of 50–80 ml/min), comparing allopurinol with febuxostat, four
moderate (CCR of 30–49 ml/min), and severe (CCR deaths occurred in subjects receiving the latter
of 10–29 ml/min) renal impairment. Febuxostat drug at doses of 80 and 120 mg. These deaths did
therapy showed no change in the effectiveness of not appear to be related to the drug (congestive
this drug to lower the serum uric acid levels in the heart failure and respiratory failure in one patient,
face of renal failure [737–739]. This is in contrast retroperitoneal bleeding in a patient receiving
to allopurinol where a dose reduction of anticoagulants, metastatic colon cancer in another
allopurinol is advised when renal failure is pres- patient, and cardiac arrest in another) [736, 740].
ent. Using three different doses of febuxostat (40, No serious life-threatening rashes have been
80, and 120 mg), the levels of serum uric acid reported in contrast to the deaths reported with
were reduced significantly when compared with allopurinol hypersensitivity reactions [637, 657,
patients receiving placebo [740]. 742]. Febuxostat undergoes glucuronidation and
A febuxostat dose of 40 mg was less effective oxidation in the liver and is excreted either as an
in reducing serum uric acid levels than the higher unchanged molecule or as its metabolites. It can
doses, and on this basis, it was concluded that be used in patients with mild or moderate liver
a dose of 80–120 mg was probably the appropri- disease without complications but cannot be used
ate dose range for clinical use. Furthermore, the in the presence of severe liver disease.
presence of elevated serum levels of hypoxan- The significance of this agent for the treatment
thine and xanthine during febuxostat treatment of gout relates to its differences from allopurinol.
documents xanthine oxidase inhibition as the pri- Febuxostat resides in a channel leading to the
mary mechanism of action of this drug [740]. active site of the enzyme xanthine oxidase and
Xanthine Oxidase Inhibitors: Allopurinol and Febuxostat 337

impedes the access of the substrate for the enzyme and both drugs reach peak concentrations within
to interact with the active site of the enzyme, 1 h [747, 749, 750]. The metabolites of febuxo-
whereas allopurinol and oxypurinol bind directly stat are excreted equally in the stool and urine. As
to the active site of xanthine oxidase [724, 730, noted previously, allopurinol causes a rash in
743–746]. Allopurinol is rapidly oxidized to its about 2 % of patients; about 10 % of patients
active metabolite oxypurinol by xanthine oxi- manifest intolerance to the drug due to abnormal
dase. Oxypurinol has a longer tissue half-life hepatic enzymes, gastrointestinal side effects, or
(14–30 h) than allopurinol (2–3 h) and accounts central nervous system effects; and it is contrain-
for the principal pharmacological effects of these dicated in patients receiving 6-mercaptopurine or
two compounds. The longer half-life of oxy- azathioprine due to the allopurinol-induced delay
purinol is the result of its renal reabsorption in the metabolism of these antimetabolites that
[747]. Allopurinol binds to the oxidized form of increases their toxicity. Alterations in the renal
xanthine oxidase at the Mo6+-pterin coenzyme, elimination of oxypurinol may lead to oxypurinol
whereas febuxostat blocks the access of xanthine calculus formation. Finally, the most significant
oxidase substrates to the molybdenum-pterin side effect is the allopurinol-induced hypersensi-
moiety. At low concentrations, both allopurinol tivity syndrome that occurs in roughly 1 in a
and its active metabolite oxypurinol are competi- 1,000 patients taking allopurinol. This syndrome
tive inhibitors of xanthine oxidase. At higher is associated with a high mortality rate [748].
concentrations, both compounds are noncompeti- Recent studies have also determined that some
tive inhibitors of xanthine oxidase. Oxypurinol severe cutaneous adverse reactions caused by
binds to the reduced form of the enzyme xanthine allopurinol are linked to the genetic marker HLA-
oxidase with high affinity, but it is released when B*5801 haplotype [656]. These studies have only
the enzyme is reoxidized via its sulfhydryl resi- been reported in Chinese subjects and need to be
dues or by proteolysis. The potent nonpurine evaluated in other population groups.
xanthine oxidoreductase inhibitor febuxostat The most common adverse reactions associ-
exhibits a mixed type inhibition with Ki and K¢i of ated with febuxostat treatment are abnormal liver
1.2 ± 0.05 × 10−10 and 9.0 ± 0.05 × 10−10 M, respec- function tests, diarrhea, headache, arthralgias,
tively. In contrast to allopurinol, febuxostat inhib- musculoskeletal pain and stiffness, vomiting, diz-
its both the oxidized and reduced forms of ziness and dysgeusia, asthenia and fatigue, and
xanthine oxidase [724, 730]. The latter drug also peripheral edema [736, 751]. Of all the adverse
has only minor effects on purine and pyrimidine reactions associated with febuxostat therapy,
enzymes except for xanthine oxidase in contrast abnormal liver function tests and gouty flares are
to allopurinol that inhibits not only xanthine oxi- the most frequent when doses of 80–120 mg/day
dase activity but also blocks orotidine-5’-mono- are used. On the basis of the foregoing findings,
phosphate decarboxylase and purine nucleoside febuxostat appears to have some distinct advan-
phosphorylase activities [741, 748]. Febuxostat tages when compared with allopurinol in specific
has no effect on guanine deaminase, hypoxan- clinical settings, but a better evaluation of febuxo-
thine-guanine phosphoribosyltransferase, and stat will become available when the drug is
orotate phosphoribosyltransferase activities and released for general usage. Since febuxostat is
does not increase PRPP levels. The other key dif- metabolized by the liver and not dependent on the
ference between allopurinol and febuxostat is kidney for its metabolism, it may be a useful
that the latter compound is principally metabo- agent for the treatment of patients with impaired
lized by the liver and not the kidney [749, 750]. renal function, but sufficient data are not avail-
Hepatic metabolism converts febuxostat to an able at this time to assess the drug in patients with
acyl-glucuronide metabolite and to an oxidative impaired renal function [752]. If febuxostat ther-
metabolite. These metabolites have been desig- apy proves efficacious in the presence of renal
nated as 67 M-1, 67 M-2, and 67 M-3. Both failure, this would be a major advance for the
allopurinol and febuxostat are rapidly absorbed, management of hyperuricemia and gout as well
338 9 Management of Hyperuricemia and Gout

as other clinical settings associated with impaired lar disease as compared to rats exposed to oxonic
renal function. Febuxostat at a dose of 80 mg/day acid and placebo. These febuxostat-treated rats
appears to be more efficient drug than allopurinol also showed a decrease in mesangial matrix
300 mg/day for lowering the serum urate levels expansion and in the development of preglomer-
[736]. Both allopurinol and febuxostat at doses of ular arteriolar disease. Thus, these animal studies
300 and 80 mg/day, respectively, reduce the num- suggest that the hypertension commonly observed
ber of gout flares and the size of tophaceous in some gouty patients may respond to febuxostat
deposits, but the number of patients achieving therapy, but proof of such a therapeutic effect
serum urate levels of less than 6 mg % was greater must await the use of febuxostat in hypertensive
in the febuxostat 80–120 mg/day groups than in gouty patients.
the allopurinol 300 mg/day group. Pharmacoki- Finally, since febuxostat as well as other med-
netic data show that febuxostat may be used ical disorders can lead to abnormalities in hepatic
either with indomethacin or naproxen without functions, evaluations of the use of febuxostat in
any dose adjustment of febuxostat [753]. Antacids humans need to be preceded by an examination
have been shown to have no effect on the absorp- of the patient for preexisting liver abnormalities.
tion of febuxostat, and even though food decreased Recent studies have reported the effect of febuxo-
febuxostat absorption, it has no effect on febuxo- stat on patients with normal liver function and
stat pharmacodynamics. those with mild and moderately impaired hepatic
In addition to these studies in humans, two functions [757]. At a dose of 80 mg/day of
different animal studies are relevant to the use of febuxostat, no statistically significant difference
febuxostat, if these results are confirmed in in plasma pharmacokinetics has been determined
human subjects. A high-fructose diet in rats leads for febuxostat or its metabolites in patients with
to hyperuricemia, hypertension, triglyceridemia, normal hepatic functions or those patients with
and hyperinsulinemia [754, 755]. These studies mild or moderately impaired liver functions.
compared rats on a high-fructose diet with pla- There was, however, a statistically significant
cebo in their drinking water with rats on the same difference in the mean percentage decrease in
diet but with febuxostat in their drinking water. serum uric acid when those with hepatic impair-
Febuxostat-treated rats manifested a significantly ment were compared to those with normal
lower blood pressure, serum uric acid, triglycer- hepatic functions. These differences turned out
ide, and insulin levels. In addition, the febuxo- to be relatively small (13–14 % less in patients
stat-treated rats also had significantly reduced with hepatic impairment), and those changes
glomerular pressure, renal vasoconstriction, and were considered to be insignificant by the authors
afferent arteriolar area when compared to place- of the study. Furthermore, there was an increase
bo-treated rats. Thus, febuxostat not only normal- in mean serum and urinary xanthine values in
izes the serum uric acid levels but also decreases those with impaired hepatic functions. Although
the metabolic, morphologic, and glomerular this small group of individuals manifested no
hemodynamic changes associated with the fruc- apparent adverse reactions from this increment
tose-induced metabolic syndrome. These studies in serum xanthine levels, a larger number of
suggest a potential role for febuxostat in the man- patients might detect the occurrence of xanthine
agement of the human metabolic syndrome, but stones in such febuxostat-treated patients with
investigations of humans with this syndrome hepatic dysfunction. As documented by the fore-
have yet to be conducted. The second study used going discussion, febuxostat appears to have
oxonic acid-induced hyperuricemia in rats to properties that would be useful to patients with
evaluate the associated hypertension and renal gout and renal disease. It may also have a role in
alterations that can be prevented by allopurinol the treatment of gout associated with hyperten-
therapy [756]. Rats treated with oxonic acid and sion and the metabolic syndrome. More persua-
febuxostat showed a reduction in their serum uric sive documentation of the clinical use of
acid levels and improvement in systemic arterio- febuxostat must await more extensive clinical
Special Treatment Regimens 339

trials with careful documentation of adverse Table 9.32 Dietary restrictions for a low-purine diet
reactions to this agent. Absolute restrictions
A number of studies clearly document the No meats or poultry
association of increased serum uric acid levels No fish or seafood
with cardiovascular disorders, and there has been No herring, sardines, or anchovies
a concerted interest in improving the risk of car- No organ foods (liver, kidney, or heart)
diovascular disease by lowering uric acid levels, No meat extracts or gravies
although it has not been proven that hyperurice- No yeast or yeast products
mia itself is a causative factor in cardiovascular No beer, ale, or alcohol
diseases or whether hyperuricemia is simply No asparagus, beans, cauliflower, lentils, mush-
rooms, oatmeal, or spinach
associated with other risk factors for cardiovas-
Unrestricted foods
cular disease, such as obesity [758–771]. Refined cereals and cereal products
However, several drugs designed for the treat- White bread, flour, sago, arrowroot, and tapioca
ment of cardiovascular diseases, such as Milk, milk products, cheese, and eggs
fenofibrate and losartan, may also be useful in the Sugar and sweets
management of refractory gout since they reduce Gelatin
serum uric acid levels. Butter, polyunsaturated margarine, and fats of any type
Fruit, nuts, and peanut butter
Lettuce, tomatoes, and green vegetables except for
Low-Purine Diet those cited under restricted foods
Beverages
There are relatively few clinical circumstances in Water, fruit juices, and carbonated drinks
Tea, coffee, and cocoa should be excluded if uricase
which strict dietary purine restriction is necessary
or HPLC methods are not used to measure uric acid
since effective urate-lowering drugs are available. since methylxanthines interfere with the accurate
Nonetheless, where it is important to know the measurement of uric acid by colorimetric methods
absolute metabolism and excretion of urate, such
dietary restrictions are used (Table 9.32). Further,
one may wish to inform patients who are at and green vegetables except those noted under
increased risk for gouty episodes and who are restricted foods are all permitted. Vegetables and
prone to overeat foods containing large quantities cream soups without meat or meat stock are also
of purines to avoid such foods. In addition, some permitted, but such a purine-free diet is still not a
patients with gout can identify specific foods or very healthy or palatable diet.
beverages that trigger an acute attack of gout, and Water, fruit juices, carbonated drinks, tea, cof-
if these are identified, patients may wish to avoid fee, and cocoa are permitted ad libitum. If uricase
these foods or beverages. or high-performance liquid chromatography is
The absolute restrictions for purine-free diets used to measure uric acid in the urine, methylx-
include no meats or poultry; no fish or seafood; no anthines do not interfere with uric acid assays.
herring, sardines, or anchovies; no organ foods
(liver, kidney, or heart); no meat extracts or gra-
vies; no yeast or yeast products; no beer, ale, or Special Treatment Regimens
alcohol; and no asparagus, beans, cauliflower, len-
tils, mushrooms, oatmeal, or spinach. As one can Management of Acute Uric Acid
imagine from this list, such a diet is almost unpal- Nephropathy
atable. Refined cereals and cereal products, white
bread, flour, sago, arrowroot, tapioca, milk, milk Uric acid nephropathy is only one of a myriad of
products, cheese, eggs, sugar and sweets, gelatin, intraluminal, intramural, and extrinsic causes that
butter, polyunsaturated margarine, fats of any can lead to urinary tract obstructions. Uric acid
type, fruits, nuts, peanut butter, lettuce, tomatoes, nephropathy is usually a complication of the use
340 9 Management of Hyperuricemia and Gout

of alkylating agents in the treatment of malignant Hydration with intravenous fluids, if no signs of
neoplasms of the hematopoietic system [772]. volume overload exist, should be undertaken
Rarely, this complication has also been reported immediately after the diagnosis has been
in association with disseminated adenomatous confirmed and urine alkalinization accomplished
carcinoma of the gastrointestinal tract [773]. by the infusion of base. Treatment regimens for
Usually, acute uric acid nephropathy is evident urinary alkalinization have been described in
because of the use of alkylating agents, but in the detail in the previous section dealing with the
case of disseminated carcinoma, it may present as management or urinary uric acid calculi. Urinary
an unexpected event. The hallmarks of managing pH should be monitored at regular intervals to
this disorder rest with a careful physical examina- ensure the alkalinity of the urine. Further, careful
tion accompanied by a laboratory and radiologic monitoring of urine output, electrolyte imbal-
evaluation. Assessment of extracellular volume ance, and other metabolic parameters should be
depletion or volume overload as well as a search instituted to prevent fluid overload and other clin-
for features of urinary tract malfunction is essen- ical problems (metabolic alkalosis) associated
tial before appropriate treatment is undertaken. with overhydration and alkalinization. Allopurinol
Urinalysis should include a careful examination is the conventional drug utilized to decrease the
of the urinary sediment for the presence of micro- urate load. Since both allopurinol and xanthine, a
scopic hematuria or crystals (uric acid or others). product of xanthine oxidase inhibition by
A white blood cell count may indicate or confirm allopurinol, are relatively insoluble, care must be
an underlying hematopoietic malignancy. Serum taken to maintain a high urine flow rate during
electrolytes, blood urea nitrogen and creatinine the administration of this hypouricemic drug.
concentrations, serum calcium, magnesium, uric In patients who cannot be diuresed, hemodi-
acid, phosphorus, and albumin are measured to alysis or peritoneal dialysis can be utilized to
assess the presence of prerenal azotemia or intrin- remove significant quantities of urates [779,
sic renal failure. Since ultrasonography has a high 780]. Peritoneal uric acid clearance is about
sensitivity and specificity for the detection of 11 ml/min and may be sufficient to reduce ele-
hydronephrosis, this is the procedure of choice as vated serum urate levels. When the serum uric
the initial radiologic evaluation [774–776]. If a acid level is 15 mg/dl or greater, hemodialysis
stone is suspected, then a flat plate of the abdo- should be used. Hemodialysis should not only be
men will reveal a radiopaque stone. In cases directed at removing excess urate but also at cor-
where ultrasonography and a flat plate are unre- recting the azotemic state. If uric acid sludge
warding, then spiral computed tomography is the obstructs the urine flow, then surgical irrigation,
procedure of choice [777, 778]. which has been discussed in the section on the
Acute uric acid nephropathy causes a tubular management of uric acid nephrolithiasis, should
interstitial nephritis, which must be treated be undertaken.
promptly if this form of acute renal failure is to
be reversed. Prevention is the best approach to
this clinical problem, but in the clinical setting in Management of Tophaceous Deposits
which this disorder is suspected, three principles
govern its management: early recognition, induc- Surgery to remove tophaceous deposits is per-
tion of a high urine flow rate, and reduction of the fectly reasonable when such tophi are large, and
urate load to the kidney. The criteria for early rec- their removal will decrease the uric acid load
ognition have been discussed in the chapter on excreted by the kidney, increase the rate of nor-
clinical gout and will not be reiterated here. malization of the serum uric acid levels, and
Both the induction of a high urine flow and the improve the cosmetic appearance and function of
reduction of urate loads to the kidney should be a joint. Carpal and tarsal tunnel syndromes caused
undertaken concurrently. Neither procedure alone by tophaceous deposits do occur, and surgical
can reverse this form of acute renal failure. repair is certainly indicated to prevent functional
Special Treatment Regimens 341

disturbances that are likely to occur from persis- containing stones. The key to the therapy of
tent nerve/muscle damage. these nonurate stones is adequate hydration, and
when renal colic or renal failure occurs in asso-
ciation with xanthine stones, then shock wave
Lesch-Nyhan Syndrome lithotripsy is favored over lithotomy. Of course,
renal failure must be treated appropriately before
The treatment of Lesch-Nyhan disease entails a more extensive procedures like lithotripsy are
complex set of therapeutic measures that include undertaken.
the treatment of the purine metabolic alterations,
the management of the neurological dysfunctions,
and the control of the behavioral manifestations. Glycogen Storage Disease
The treatment of the purine abnormalities is
accomplished with allopurinol and monitoring of Type I glycogen storage disease is often compli-
the serum uric acid levels. The objective is to cated by gouty arthritis in its late stages [786].
reduce the level of serum uric acid to as near to Such patients may also have proteinuria and a
normal as possible without inducing the compli- reduced capacity to concentrate their urine that
cations associated with allopurinol therapy and to may lead one to conclude that these renal abnor-
prevent the formation of renal calculi and their malities are related to the presence of gouty neph-
complications. The dose of allopurinol to accom- ropathy. However, some patients with glycogen
plish these tasks varies depending on the age and storage disease have been reported to have proxi-
weight of the affected individual, but it is usually mal renal tubular defects with b2-microglobuli-
in the range of 100–600 mg/day in divided doses. nuria; generalized aminoaciduria with markedly
Uric acid stones are treated with a high fluid intake elevated levels of alanine, citrulline, cystine, and
and urine alkalinization. If this regimen fails, then lysine; reduced renal tubular phosphate reabsorp-
lithotripsy or surgery is necessary [781]. tion; and proteinuria [787]. Institution of a regi-
There are risks to the use of allopurinol includ- men that includes frequent feedings, nocturnal
ing hypersensitivity reactions, xanthine stones, glucose infusion, and uncooked cornstarch
and oxypurinol stones [657, 782, 783]. In patients significantly reduces serum uric acid levels and
with hypersensitivity syndromes to allopurinol, improves proximal tubular abnormalities observed
desensitization may be a useful approach to this initially in patients with untreated or poorly man-
problem; otherwise, uricosuric agents may be aged glycogen storage disease [787–789]. When
helpful, but they result in an increased risk for severe hepatic disease is observed in association
uric acid calculi. Some of the newer pharmaco- with glycogen storage disease, fresh frozen
logic agents, such as febuxostat and urate oxi- plasma or exchange transfusions may be used to
dase, may be useful in this. Increased hydration is correct the defective clotting and complement
an absolutely critical component of all treatment deficiencies incurred by liver disease [790].
regimens as a means of reducing the formation of
renal calculi. In addition, to prevent the deteriora-
tion of kidney function from nephrocalcinosis or Hereditary Fructose Intolerance (HFI)
recurrent stones, regularly scheduled renal ultra-
sound evaluations should be done [781, 784]. Since gout has been observed in patients with
Xanthine stones are difficult to treat since hereditary fructose intolerance, diagnosis and
they are not very soluble, even with urine alka- treatment of the underlying carbohydrate abnor-
linization [785]. Although it has been suggested mality will improve the alterations in uric acid
that increasing the dose of allopurinol may favor metabolism as a result of the defective fructose
the production of the more soluble purine prod- metabolism observed in such patients [791, 792].
uct hypoxanthine, such increments in allopurinol Such treatment has the potential to significantly
doses may lead to the formation of oxypurinol- decrease the episodes of gout or completely cure
342 9 Management of Hyperuricemia and Gout

the gouty arthritis that accompanies hereditary attainment and maintenance of serum uric acid
fructose intolerance. levels below 6.0 mg/dl have been documented in
Appropriate treatment of HFI requires the many publications as a means of preventing
complete elimination of all sources of sucrose recurrent episodes of acute gout [638, 798, 799].
and fructose form the diet [791–794]. Even slight Thus, the documentation of repetitive episodes of
relapses in such dietary restrictions to permit the gout suggests an absence of the reduction of
ingestion of 250 mg/kg of fructose per day have serum uric acid to appropriate levels. Second, the
been shown to cause a sustained increase in serum disappearance of tophi observed by physical
and urinary uric acid levels [795]. The fructose examination or radiographically is an indicator of
and sucrose content of various fruits and vegeta- the improvement of chronic tophaceous gout.
bles has been documented, and these published Studies have also shown that a failure to regulate
tables of food composition illustrate the broad recurrent gouty episodes not only causes a pro-
distribution of these sugars in fruits, nuts, fruit gression of the disease but also impairs the life-
juices, and vegetables [793, 794]. In the absence style of affected patients [800–803]. Another
of dietary sources of fructose and sucrose, HFI parameter worth considering with respect to
patients thrive and live a long life [795, 796]. refractory gout is that approximately 20 % of
Since solutions containing sorbitol, fructose, and patients prescribed 300 mg of allopurinol/day
other invert sugars are still used in some hospi- attain and maintain serum uric acid levels of
tals, patients with HFI should be warned as to the 6 mg/dl [736, 801, 804]. However, if most patients
use of such feeding solutions and the need to alert (80 %) treated with 300 mg of allopurinol fail to
physicians to their carbohydrate intolerance. obtain therapeutic serum uric acid levels sufficient
These admonitions to patients should not be taken to reduce the number of recurrent gouty episodes,
lightly since fatalities have occurred when such other therapeutic initiatives need to be consid-
parental therapy has been administered to patients ered. It is also important to recognize that 90 %
with HFI in hospital settings [797]. of the patients with gout are prescribed allopurinol
at a dose of 300 mg/day. Another significant fac-
tor that clearly influences the control of acute
Refractory Gout gouty episodes is the number of patients who are
noncompliant with respect to their prescribed
A small group of patients suffers from recurrent urate-lowering drug regimens. Such noncompli-
flares of gouty arthritis and progressive topha- ance appears also to increase over time [805,
ceous deposits with an associated chronic arthri- 806]. The presence of chronic kidney disease also
tis despite a variety of available medications for influences the management of gout. A general
lowering serum uric acid levels and for prevent- principle regarding the treatment of gout in
ing the progression of gout. Affected patients patients with diminished kidney function has
have been described by the term “refractory been to reduce the allopurinol dose to avoid pre-
gout,” and they represent therapeutic challenges. cipitating an allopurinol hypersensitivity syn-
A number of underlying causes for this disorder drome and/or triggering the formation of
have been documented as responsible for this oxypurinol stones. An allopurinol dosing regi-
refractory condition including the delayed insti- men has been published based on the half-life of
tution of urate-lowering drugs, no use of such oxypurinol that has been designed to prevent
urate-lowering drugs, medication intolerances allopurinol-induced complications in patients
(allopurinol), incomplete patient responses with chronic renal disease [650]. However, recent
despite appropriate treatment regimens, and data show that such modified dosing regimens do
patient noncompliance. To assess the regulation not appear to control allopurinol-induced hyper-
of the number of gouty episodes and the abroga- sensitivity reactions [807, 808]. Furthermore, the
tion of the progression of chronic tophaceous reduction in allopurinol dosage based on the
gout, two key markers are utilized. First, the Hande algorithm not only fails to protect patients
Special Treatment Regimens 343

from the allopurinol hypersensitivity syndrome (rasburicase) has now been approved for short-
but also fails to reduce the serum uric acid levels term use in patients treated for malignancies that
sufficiently for the control of acute episodes of are expected to induce the tumor lysis syndrome
gout in the face of chronic renal disease. [818].
The issues raised by the foregoing data are Uricase preparations have been utilized as
what are the most effective means for managing treatment for uric acid accumulations in disease
refractory gout and, more specifically, what ther- states for a number of additional reasons. Urate
apeutic regimens can be used to reduce serum oxidase (uricase) catalyzes the degradation of
uric acid levels to prevent recurrent gouty epi- uric acid to a metastable intermediate, 5-hydroxy-
sodes. As the subsequent discussion will describe, isourate, and then this intermediate is spontane-
a number of different drugs have been utilized to ously converted to allantoin [819]. The latter
manage refractory gout. Such treatment regimens compound is roughly five to ten times more solu-
not only have shown that modified allopurinol ble than uric acid [820]. Uricase preparations
doses can effectively lower serum uric acid levels resolve not only the deposition of newly synthe-
in refractory gout but a variety of other drugs sized urate but also long-standing tissue deposits,
including uricase, losartan, fenofibrate, febuxo- whereas allopurinol only impairs the synthesis of
stat, and anakinra have been proposed or actually uric acid and has no effect on tissue deposits of
used for the management of such patients. urate. Rasburicase, the recombinant form of the
Purified uricase has been approved in the urate oxidase enzyme, has a half-life of 16–22 h
United States for the treatment of tumor lysis in the human [821, 822].
syndrome. It has also been used successfully in The advantages of rasburicase are its potency,
some patients with refractory gout [809–811]. its more rapid action compared with allopurinol,
However, the availability of pegylated recombi- its activity against uric acid directly rather than
nant mammalian uricase pegloticase is the pre- the inhibition of only newly synthesized uric acid,
ferred form of uricase for the treatment of and the very low rate of dialysis needed to reverse
refractory gout. The other therapeutic approaches the nephrotoxicity of uric acid in rasburicase-
to refractory gout include the use of losartan and treated patients. The absence of the need for dial-
fenofibrate, two uricosuric agents that have a ysis usually only applies to the use of rasburicase
modest uricosuric effect [812–815]. Finally, in the tumor lysis syndrome. Comparisons have
febuxostat, a nonpurine xanthine oxidase inhibi- been made between the use of intravenous rasbu-
tor that is metabolized in the liver and appears to ricase (0.20 mg/kg) and oral allopurinol [823].
be safe in patients with chronic renal insufficiency, With rasburicase, an 86 % reduction in plasma
is a safe, effective urate-lowering agent and is uric acid levels occurs in 4 h after the first dose,
particularly useful in patients who are allergic to whereas the reduction in uric acid levels after oral
or do not tolerate allopurinol. allopurinol showed only a 12 % reduction. Thus,
The original studies reporting on the use of rasburicase is a more rapid acting agent in terms
uricase as a therapeutic agent in humans were of uric acid level reduction than allopurinol.
conducted in France in 1975 and in Italy in 1984 Finally, allopurinol inhibits the enzyme xanthine
[816]. Initially, the uricase used was a nonrecom- oxidase, so it blocks the conversion of hypoxan-
binant agent purified from Aspergillus flavus. thine and xanthine to uric acid and has no effect
This agent led to a variety of hypersensitivity on the uric acid molecule itself. This mode of
reactions with skin rashes, urticaria, and angioe- action is quite different from that of uricase which
dema occurring about 5 % of the treated patients. acts directly on the uric acid molecule. The disad-
More recently, this enzyme molecule has been vantages to the use of rasburicase relate princi-
obtained using recombinant DNA technology pally to allergic reactions, and such hypersensitivity
from a strain of Aspergillus flavus and marketed responses vary depending on the publication
under the name of Elitek in the United States under review. The most recently published data
[817]. This recombinant-based urate oxidase concerned with severe adverse reactions shows
344 9 Management of Hyperuricemia and Gout

such responses to occur in less than 1 % of the reductions in tender and swollen joint counts, and
population evaluated [824]. The most severe changes in reported pain. All of the secondary
reactions to this drug are anaphylaxis, skin rashes, end points were achieved in more patients in the
hemolysis, and methemoglobinemia [825, 826]. pegloticase-treated groups than in the placebo-
Published data show skin rashes occur in about treated groups, and all but the swollen joint count
1.4 % of patients undergoing treatment, broncho- reached statistical significance.
spasm occurs in less than 1 % of patients, and Serious adverse events were common and
anaphylactic shock also occurs in less than 1 % of occurred more often in the pegloticase-treated
patients. Other reactions such as urticaria, dysp- patients. Gout flares occurred in approximately
nea, and hypoxemia also occur rarely [825]. Other 80 % of all patients and occurred more frequently
documented adverse reactions include fever in the pegloticase-treated patients. Infusion reac-
(~ 7 %), neutropenia with fever (4 %), respiratory tions occurred in 26 % of biweekly and 42 % of
distress (3 %), sepsis (3 %), neutropenia (2 %), monthly treated patients and 5 % in placebo
mucositis (2 %), nausea (1.7 %), vomiting treated. Anaphylaxis occurred in five patients
(1.4 %), headache (0.9 %), diarrhea (0.9 %), and treated with pegloticase, but there were no related
abdominal pain [827]. fatalities. Seven deaths occurred during the trial,
four among pegloticase-treated and three in the
placebo group, but none of these were attributed
Pegloticase to the treatments. Antibodies to pegloticase were
detected in 89 % of pegloticase-treated patients,
A more effective form of uricase for the treat- and these tended to correlate with infusion reac-
ment of refractory gout is pegloticase, a mamma- tion, especially if high titers of antibody were
lian recombinant uricase conjugated to found. It was noteworthy that most of the treated
monomethoxypoly(ethylene glycol). This drug patients experiencing infusion reactions had an
was approved by the FDA for the management of associated loss of urate-lowering effect. Infusions
refractory gout in September 2010. A compre- were given with infusion-reaction prophylaxis
hensive study of this agent has been published. In which may have reduced the frequency and sever-
this study, two replicate, randomized, double- ity of the reactions.
blind placebo-controlled trials were carried out in Pegloticase (Krystexxa) is recommended for
multiple centers in the USA, Canada, and Mexico. the treatment of refractory gout, which is esti-
A total of 225 patients were studied, approxi- mated to be approximately 3 % of patients with
mately half of that number in each of the two tri- gout. The recommended procedure is to adminis-
als, over a period of 6 months. All patients had ter 8 mg every 2 weeks intravenously over a
severe refractory gout. The primary end point of period of at least 2 h, carefully monitoring during
the trial was plasma urate levels of less than and after the infusion for signs of infusion reac-
6.0 mg/dl, for at least 80 % of the time, at both tions. Patients should be premedicated with a
3 months and 6 months into the trial. Patients glucocorticoid and an antihistamine for each
were divided into three groups: one group receiv- infusion. Serum uric acid levels should be mea-
ing pegloticase every 2 weeks, one group every sured prior to each infusion both to document the
month, and the third group received placebo. efficacy of the treatment, and because the loss of
When the results of the two trials were pooled, the urate-lowering effect of the drug is associated
the primary end point was achieved in 42 % of with antibodies to pegloticase, and an increased
patients in the biweekly treated group and 35 % frequency of infusion reactions. Treatment should
in the monthly treated group. None of the patients be discontinued if the serum uric acid level is
in the placebo-treated group achieved the primary >6 mg/dl on two occasions. Extra caution should
end point. be used in administering pegloticase to patients
Secondary end points in this study included with active cardiovascular comorbidities, espe-
tophus resolution, reductions in gouty flares, cially with congestive heart failure.
Other Agents with Urate-Lowering Effects 345

Pegloticase is contraindicated in patients with renal clearance of urate in humans [766, 813,
glucose-6-phosphate deficiency because of hemo- 815, 829, 837, 838]. Fenofibrate at a dose of
lysis and methemoglobinemia. Patients of African 150 mg three times a day increases the fractional
or Mediterranean descent should be screened for clearance of xanthine, uric acid, and oxypurinol
G6PD deficiency. [839]. Thus, fenofibrate therapy should be con-
Finally, because gout flares are associated sidered in gouty subjects with triglyceridemia. It
with any urate-lowering therapy and have been may also be useful in patients who need an addi-
seen frequently with pegloticase treatment, all tional drug to reduce their serum uric acid level to
patients should have prophylaxis with either a range where recurrent gouty episodes do not
colchicine 0.6 mg once or twice daily or with an occur.
NSAID.

Losartan
Other Agents with Urate-Lowering
Effects A number of published investigations have evalu-
ated the antihypertensive effects and urate-lower-
Fenofibrate ing effects of losartan potassium, a subtype 1
(AT1)-selective angiotensin II (AII) receptor
Fenofibrate is a fibric acid derivative that has a antagonist (ARB). Losartan has a relatively mod-
major effect on lipids but also reduces serum uric est uricosuric effect [588, 813]. In large popula-
acid levels [815, 828–830]. Reports have docu- tion studies (29,850 subjects), there are relatively
mented that fenofibrate reduces serum uric acid few adverse effects associated with losartan ther-
levels in healthy subjects, hypertensives, and dia- apy [840]. Only 46 patients in this study experi-
betic patients by 20–46 % [829, 831]. The other enced major adverse effects including dizziness,
pharmacologic property of fenofibrate of electrolyte disturbances, hepatic function abnor-
significance to gout is its capacity to reduce blood malities, headache, anemia, cough, blood creati-
lipid concentrations. It lowers total and LDL cho- nine phosphokinase increases, and blood pressure
lesterol by roughly 15 % and increases HDL cho- decreases (hypotension). Among the foregoing,
lesterol by about the same amount, but its cough and hyperkalemia are probably the most
principal antilipid activity is against triglycerides frequent side effects of ARBs. One additional
[828]. Since there are associations between hype- adverse effect has been reported in a patient with
ruricemia and hyperlipidemia, hypertriglyceri- idiopathic renal hypouricemia who suffered from
demia, and gout, fenofibrate may be useful as a exercise-induced acute renal failure while being
therapeutic agent in patients with hyperuricemia treated with losartan and trichloromethiazide
and gout associated with hyperlipidemia [841]. The patient’s acute renal failure improved
[832–835]. Of more pertinence to the present dis- after the cessation of both losartan and the thiaz-
cussion on the management of refractory gout, ide diuretic. The authors suggested that the epi-
fenofibrate (200 mg/day) causes a significant sode of exercise-induced acute renal failure may
reduction in serum uric acid levels (~ 20 %) in have been related to the use of losartan and thiaz-
patients utilizing allopurinol [836]. Of additional ide-induced diuresis. The rationale for suggest-
significance is the fact that patients in that study ing losartan as the possible vector of the acute
had no flares of their gout while being treated renal failure is the fact that uric acid is an antioxi-
with allopurinol and fenofibrate. In general, dant and prevents oxygen free radical-induced
fenofibrate has been shown to reduce serum uric kidney damage, and the lowering of uric acid lev-
acid to levels as low as 46 % in healthy volun- els may have reduced the antioxidant effects of
teers, hypertensive, and diabetic patients as well uric acid [842]. Another less likely explanation
as in gouty patients on specific urate-lowering for acute renal failure in patients with renal
drugs. Fenofibrate also causes an increase in the hypouricemia is the increase in urinary uric acid
346 9 Management of Hyperuricemia and Gout

concentrations related to the uricosuric effects of Table 9.33 The frequency of specific types of renal
losartan and the occurrence of acute uric acid calculi
nephropathy [843]. Calcium oxalate 60 %
The uricosuric effect of losartan has been doc- Calcium phosphate or calcium oxalate 20 %
and calcium phosphate
umented in a number of publications [588, 844–
Uric acid 8%
849]. In one publication, the reduction of serum
Magnesium ammonium phosphate 8%
uric acid levels was demonstrated to be in the
Other types 4%
range of 0.05 mmol/l [892]. The use of losartan
also counterbalances the rise in serum uric acid
levels induced by thiazide diuretics [850, 851]. chemical composition of the stone is unknown.
Losartan is also useful in the management of Renal calculi are relatively common in the United
hypertensive cyclosporine-treated renal trans- States since about 12 % of males and 5 % of
plant patients [852]. In these patients, 50 mg of females have an attack of renal colic by the age of
losartan caused an increase in the fractional 70 [856]. One cannot assume that all renal calculi
excretion of uric acid (17 %) and a modest reduc- associated with the clinical and laboratory fea-
tion in serum uric acid levels (8 %). Of even tures of derangements in uric acid metabolism
greater significance to the management of serum are due to uric acid. In fact, although uric acid
uric acid reductions, a combination of fenofibrate stones are not uncommon, other stones are much
and losartan caused a greater decrease in serum more common and may be associated with abnor-
uric acid levels than either agent alone [813, 830, malities in uric acid metabolism (Table 9.33).
853]. The combination of losartan and fenofibrate This discussion emphasizes the general principles
results in an additive urate-lowering effect in related to the management of uric acid nephro-
comparison to either agent alone, and losartan lithiasis, whereas a more extensive discussion of
may protect against uric acid lithiasis by its urine the treatment of uric acid stones is provided in
alkalinization effect [854]. The literature recom- Chap. 5.
mends that allopurinol therapy be initiated before The most common risk factors associated with
adding fenofibrate and losartan to treatment regi- renal stone formation are a low urine volume,
mens to avoid urolithiasis due to the induced hypercalciuria, hyperuricosuria, hyperoxaluria,
changes in uric acid metabolism by allopurinol hypocitric aciduria, altered urinary pH, and cysti-
[813]. Losartan also increases the urinary excre- nuria. These risk factors must be identified if
tion of xanthine and oxypurinol [839]. The use of recurrent stones are to be prevented [857]. Which
losartan in combination with fenofibrate certainly patients deserve a comprehensive evaluation after
reduces serum uric acid levels, has a positive their first renal calculus remains a debatable sub-
effect on lowering serum triglycerides, and would ject. Some have suggested that a simple regimen
likely reduce the risk factors caused by uric acid of a high fluid intake and the avoidance of dietary
in cardiovascular events, renal disease, hyperten- excesses are sufficient to decrease the recurrence
sion, and diabetes [855]. The foregoing effects of of stones [858]. Others have provided data to
losartan and fenofibrate therapy document the suggest that in men, more than 50 % have a recur-
potential usefulness of these drugs in refractory rence of stone formation within 7 years of their
gout. first stone and need diagnostic evaluations and
treatment to prevent such recurrences [859].
There is general agreement that individuals
Management of Nephrolithiasis with metabolically active stones (calculi that
increase in size over time) and significant risk
The management of nephrolithiasis is reasonably factors (family history of stones, age of onset of
straightforward when the chemical composition less than 30, the presence of small or large bowel
of the stone is known, but more often than not, disease, urinary tract infections, osteoporosis,
only a history of renal colic is obtained, and the gout, and pathological or minimal trauma
Management of Nephrolithiasis 347

Table 9.34 Medical evaluation of high-risk stone Table 9.35 Renal calculus without risk factors: labora-
formers tory and radiological evaluations
Historical data Blood and urine tests
Dietary history (oxalate and vitamin C) Fasting serum calcium (ionized)
Immobilization Fasting serum bicarbonate
Medications Fasting serum phosphorus
Furosemide Serum electrolytes
Thiazide Urinalysis and urine culture
Uricosurics 24-h urine
Alkali Total volume
Vitamin A Uric acid
Vitamin D Calcium
Methoxyflurane Creatinine
Acetazolamide Serum uric acid and creatinine
Radiological examination Radiological analyses
Nephrocalcinosis Plain film of the abdomen for nephrocalcinosis to
Stones (radiodense/radiolucent) assess the possible presence of RTA, primary
Fasting serum concentrations hyperparathyroidism, medullary sponge kidney, or
polycystic kidneys
Calcium (ionized and total)
Renal ultrasonography to assess the presence of
Phosphorus
hydronephrosis or stone
Bicarbonate
CT scan
Chloride
Other radiological procedures if necessary (IVP)
Creatinine
Uric acid
Albumin suspected of having stones composed of cystine,
Urinalysis
uric acid, or magnesium ammonium phosphate
pH by nitrazine paper
(struvite); have recurrent stones; or have risk fac-
Urine culture for urea-splitting organisms
tors noted previously. It should be recognized
24-h urine specimen on a defined diet of 1 g of
calcium, 1 g of protein/kg, and 100 mg of sodium for that risk factors such as hypercalciuria, hyperuri-
24 h for the following measurements cosuria, hyperoxaluria, and hypocitraturia do not
Calcium just occur as solitary findings. For example, of
Sodium the patients with hyperuricosuria (4,800 umol/
Uric acid day in men and 4,400 umol/day in women),
Creatinine approximately 56 % will have hypercalciuria
Phosphorus (>7 mmol/24 h), 22 % will have hypocitraturia
Oxalate (<1.2 mmol/24 h), and 19 % will have hyperox-
Cystine aluria (>0.625 mmol/24 h) [857]. Hyperuricosuria
Optional tests
is also frequently observed in association with
Ammonium chloride load (0.1 g/kg)
hypercalciuria and hyperoxaluria. For example,
Parathormone assay
47 % of patients with hypercalciuria have been
24-h urinary citrate
shown to have hyperuricosuria [857]. Such data
emphasize the need for a careful evaluation of
fractures) require a complete medical evaluation. patients who present with renal calculi of
Further, all children with stones deserve a careful unknown composition. It is also clear that abnor-
and complete evaluation. malities in serum and urinary uric acid are not
Simple outpatient medical evaluations can be absolute indicators of underlying gout since they
performed in those patients without significant may also be observed in patients with hypercal-
risk factors (Tables 9.34 and 9.35). More exten- ciuria and hyperoxaluria due to causes quite apart
sive evaluations are required when patients are from a defect in purine metabolism [857, 860, 861].
348 9 Management of Hyperuricemia and Gout

Such patients often need treatment with colchicine (0.5–1 mg/day) are recommended for
allopurinol but for reasons somewhat different patients beginning allopurinol therapy. The 1-mg
from the rationale for the use of this drug in dose is given in divided dose. Second, initiating
gout. allopurinol at a low dose and gradually increas-
When the composition of a stone is known to ing the dose over several weeks to 300 mg/day
be pure uric acid or primarily uric acid, then the also reduce the likelihood of sudden large changes
diagnostic studies and management of such in the uric acid pool giving rise to acute gouty
patients are relatively simple. As noted, uric acid arthritis. Occasionally, patients overindulge in
nephrolithiasis is most often observed in patients high-purine foods and require modification of
with secondary gout (especially hematologic dis- their diets to reduce their uric acid intake
orders), primary idiopathic gout, inherited defects (Table 9.32).
in uric acid metabolism (hypoxanthine-guanine
phosphoribosyltransferase deficiency and phos-
phoribosylpyrophosphate synthetase overactivity The Role of Diet and Comorbidities
and glucose-6-phosphatase deficiency), the use and Their Management in Gout
of uricosuric agents (probenecid, high-dose sali-
cylates, and X-ray contrast materials), persis- In terms of the management of gout, one must
tently acid urine with or without gout, idiopathic also consider dietary factors and lifestyles that
uric acid stones, chronic inflammatory bowel dis- can modify the gouty diathesis. Several facets of
ease, patients with ileostomies, or dehydration the diet are important to the reduction of the risks
associated with these clinical problems. of a gouty episode: meat and seafood intake,
Uric acid calculi associated with gout require alcohol use, and obesity. Although many physi-
specific treatment regimens quite different from cians still advise patients to reduce their dietary
their use to manage acute and chronic gouty intake of purines, purine-restricted diets only
arthritis. Uric acid nephrolithiasis observed in reduce mean serum urate levels by 0.6 mM/l, and
association either with enzyme mutations in such diets are relatively unpalatable [864–866].
purine biosynthesis, acquired uric acid overpro- Other studies have also documented decreases in
duction, or abnormal urate excretion are managed the urinary uric acid excretion by approximately
by increasing the urine volume, decreasing uri- 200–400 mg/day and mean serum uric acid levels
nary uric acid excretion, and alkalinization of the by roughly 1–2 mg/dl when the patients were
urine. restricting their purine intake [864–866]. For the
There is no rationale for a rigid restriction of moment, the best data related to diet comes from
dietary purines and proteins in the management a recent study of 47,150 men over a 12-year
of gout or nephrolithiasis associated with gout. period that shows meat and seafood increase the
However, alcohol, especially beer, can increase risk of gout [867]. In contrast, dairy products,
uric acid excretion [862]. High alcohol intake has especially low-fat dairy products, are able to
also been associated with uric acid stones [863]. decrease the risk of gout in men. In addition to
Such data indicate that curbing alcohol intake food, alcohol is another significant risk factor for
may be a useful therapeutic maneuver in the man- gout. The relationship between gout and alcohol
agement of uric acid nephrolithiasis. is more complex. Acute alcohol intoxication in
individuals documents a higher plasma urate
concentration when urate levels are measured
Allopurinol Treatment of Stones after individuals have become sober [868]. Such
urate alterations may also occur as a result of not
Allopurinol at a dose of 300 mg/day is used to eating and the presence of ketosis. The latter state
reduce the urinary excretion of uric acid, and two and its circulating ketones and lactate block urate
strategies exist for preventing the onset of acute excretion causing hyperuricemia. In normal
gout when this drug is used. First, low doses of individuals, elevated plasma levels of ethanol
The Role of Diet and Comorbidities and Their Management in Gout 349

(> 200 mg/dl) cause a reduction in urinary urate consumption does not increase the risk of gout
levels [869, 870]. When moonshine whiskey was [920, 921].
produced from lead-soldered stills, saturnine gout
or lead-induced gout was a common occurrence
in the southern part of the United States [871]. Hypertension
Finally, beer contains a purine (guanosine) that
can be metabolized to uric acid [872–880]. Recent Initial studies attributed the association between
studies of 14,809 individuals in the Third National the hyperuricemic state and hypertension either
Health and Nutrition Examination Survey to reduced renal function, the use of diuretics, the
(NHANES) examined the role of beer, wine, and presence of hyperinsulinemia and oxidative
hard liquor in relation to uric acid [881, 882]. In stress, or elevated renal vascular resistance [922].
this study, beer and hard liquor intake were posi- These relationships between hypertension and
tively correlated with hyperuricemia. It has been hyperuricemia led to the conclusion that hyperu-
proposed that a glass of wine as a replacement for ricemia was not a true risk factor for hyperten-
beer or hard liquor might reduce the incidence of sion [923, 924]. However, more recent evidence
gout [874, 883]. Both alcohol intake and obesity has reported that hyperuricemia is an indepen-
can cause an increase in urate production and a dent risk factor for the development of hyperten-
decrease in its elimination by the kidney [884– sion [925–929]. A greater percentage of
902]. When the body mass index (BMI) is evalu- adolescents have been determined to manifest
ated with respect to the relative risk of gout on an hyperuricemia in association with essential
age-adjusted basis, the following data have been hypertension (89 %) in contrast to a few (30 %)
obtained. For a BMI of 23–24.9 kg/m2, there was with secondary hypertension, none with essen-
a relative risk for gout of 1.4, and for a BMI of tially normal blood pressure, and none with
25–29.9 kg/m2, the relative risk increased to 2.35. white-collar hypertension [930]. These human
When the BMI was 30–34.9 kg/m2, the relative studies have been supplemented by animal stud-
risk for gout was 3.26, and for a BMI of greater ies using oxonic acid to inhibit uricase activity
than 35 kg/m2, the relative risk for gout was 4.41 and to increase uric acid levels in the serum of
when compared to individuals with a BMI of rats [931, 932]. In these animal models, the
21–22.9 kg/m2 [891, 903–905]. As noted, obesity hypertensive response has been attributed to a
increases the urate production and decreases reduction in endothelial nitric oxide levels medi-
renal urate excretion. As further proof of the ated by uric acid and a stimulation of renin
association between obesity and uric acid levels, expression [933, 934]. Studies demonstrating
weight reduction results in a decrease in serum uric acid-mediated alterations in endothelial
urate levels. It is also important to remember that function and renin levels have now been shown to
hyperuricemia and gout are also associated with be present in humans as well [935–938]. Finally,
the metabolic syndrome. The incidence of this xanthine oxidase inhibition and the lowering of
cluster of clinical findings (central obesity, dys- serum uric acid levels have been shown to
lipidemia, hypertension, insulin resistance, glu- improve endothelial cell function in chronic heart
cose intolerance/type 2 diabetes mellitus, failure and other cardiovascular conditions
hyperuricemia, and a proinflammatory and pro- [939–941].
thrombotic state) associated with the metabolic The foregoing data provided the impetus to
syndrome has a prevalence of 20–30 % in some evaluate the possible role of antihyperuricemic
populations [904–919]. Recently, an additional drugs like allopurinol on hypertension. A small
dietary factor has been discovered that may have group of children have now been evaluated in
an impact on the risk of gout [920, 921]. Sugar- double-blind, placebo-controlled crossover study
sweetened soft drinks containing fructose are to determine whether allopurinol therapy admin-
strongly associated with an increased risk of gout istered to lower the uric acid levels has any effect
and hyperuricemia in men, but diet soft drink on the essential hypertension manifested by these
350 9 Management of Hyperuricemia and Gout

adolescents [942]. The results of this therapeutic xanthine oxidase inhibitors and reduce serum
trial demonstrated a mean change in 24-h ambu- uric acid levels [943, 944]. Studies aimed at
latory blood pressure of 6.3 mmHg with improving endothelial function in heart failure
allopurinol treatment versus 0.8 mmHg with pla- patients showed that in contrast to allopurinol,
cebo treatment. Similarly, the mean 24-h ambula- the uricosuric agent probenecid did not improve
tory diastolic blood pressure was 4.6 mmHg with endothelial function but that reactive oxygen spe-
allopurinol therapy and only 0.3 mmHg with pla- cies mediate the effects of endothelial function
cebo treatment. These decreases in blood pres- [945–948]. Despite all the possible shortcomings
sure with allopurinol treatment matched the of this small, preliminary study in a group of ado-
decrement in blood pressure induced by standard lescents whose elevated blood pressure responded
antihypertensive medications. Of the 30 patients well to the short-term treatment with allopurinol
enrolled in this study and treated with 200 mg of (4 weeks), the notion that uric acid may play a
allopurinol twice a day, 20 attained normal blood role in the mediation of hypertension without
pressure levels while being treated with question defines the need for additional investi-
allopurinol, and only 1 patient achieved a normal gations of hypertension and its association with
blood pressure on placebo. hyperuricemia. It also clearly documents the role
Several aspects of this small, preliminary that xanthine oxidase inhibitors might have in the
study are important to recognize since they may management of essential hypertension in its early
bear on the putative suggestion that this therapeu- stages. Finally, these data emphasize the impor-
tic initiative might be applied to the general pop- tance of newer xanthine oxidase inhibitors and
ulation with essential hypertension and elevated the significant role they might play in the man-
serum uric acid levels. First, 70 % of the adoles- agement of hypertension. Such data also empha-
cents studied were obese. Thus, even though size the need for the early detection and
blood pressure cuff size was carefully selected to management of hypertension in gouty subjects.
monitor the blood pressure, obesity is not a uni- These reductions in both casual and ambulatory
versal factor representative of the general hyper- blood pressure in the allopurinol-treated patients
tensive population. On the other hand, many were similar to those found when conventional
gouty subjects are obese and manifest hyperten- therapy with b-blockers, a-blockers, and angio-
sion. Such gouty patients with hypertension may tensin-converting enzyme inhibitors were evalu-
warrant treatment with allopurinol even though ated in adults with mild hypertension [949].
they often do not meet the criteria for the use of Scientific research has now changed the way
allopurinol. Since severe and long-standing uric acid is characterized as to its role in the
hypertension in patients has been documented to human, and this new evidence has altered the
produce renal vascular changes, such vascular clinical assessment and follow-up evaluations in
alterations cause renal- and salt-dependent mech- patients with gout. Originally, uric acid was
anisms to drive the hypertensive response [925]. considered a nitrogenous waste product of
These data suggest that allopurinol therapy is not human metabolism that was excreted by the kid-
likely to modify well-established hypertension. ney and bowel. Despite a host of hypotheses
In contrast, allopurinol may be useful in the man- generated to the contrary, this nonfunctional
agement of new-onset essential hypertension and concept of uric acid has remained in vogue until
may also suggest its use early after the onset of recent studies have begun to provide the scientific
hypertension in gouty subjects. evidence with respect to the potential and true
Of course, the most serious consequence of biological functions of uric acid [950–953]. A
allopurinol therapy is the capacity of this drug to variety of investigations have now linked the
cause life-threatening adverse reactions. causes of hypertension, salt retention, hypertrig-
Nonetheless, the results of this preliminary study lyceridemia, cardiovascular adverse events, and
of allopurinol therapy as an antihypertensive drug obesity to increased serum uric acid levels [871,
focus attention on the newer agents that act as 925–927, 929, 931, 936–941, 954–973]. In addi-
The Role of Diet and Comorbidities and Their Management in Gout 351

tion to clinical data implicating uric acid levels patients. First, the metabolic syndrome increases
with cardiovascular events like hypertension, the risk of cardiovascular disease up to three
the pathological mechanisms for such associa- times and the risk for type 2 diabetes mellitus up
tions including the development of renal dis- to five times [992, 993]. It also causes an increase
ease, renin-angiotensin system activation, and in mortality from cardiovascular diseases as well
endothelial dysfunction have been characterized as all-cause mortality [994, 995]. Second, the
[930, 931, 936–940, 943, 960, 974–979]. prevalence of the metabolic syndrome in gouty
Although the data relating to the generation of patients is strikingly high (62.8 % in US adults
ROS and nitric oxide and the production of and 43.6 % in Korean adults) as compared with a
endothelial dysfunction remains controversial, general population without gout (25.4 % in the
there is an increasing body of evidence that links USA and 5.2 % in Korea) [911, 915]. This asso-
hyperuricemia with hypertension and renal dam- ciation between gout and the metabolic syndrome
age [929–931, 933, 960, 980–982]. Despite the has been confirmed in other studies as well [908,
fact that absolute correlations do not exist 912]. Cardiovascular disease in gouty patients at
between uric acid and hypertension and its high risk, those with type 2 diabetes mellitus,
pathologic mechanisms, sufficient data do exist stroke, congestive heart failure, or coronary heart
to encourage the careful monitoring for the onset disease, showed an independent association of
of hypertension in gouty patients. Furthermore, gout with cardiovascular disease and mortality
evidence also suggests that the hyperuricemic [996, 997]. Low-risk gouty patients without these
state accompanying gout should be treated with accompanying findings have determined that
hypouricemic agents along with the aggressive serum uric acid levels are poor predictors of car-
management of hypertension [983]. diovascular morbidity and mortality [969, 998].
Gout has also been documented to be associ- There are a number of publications that
ated with obesity, dyslipidemia, and hyperglyce- address components and complications of the
mia along with hypertension [832, 865, 891, 984, metabolic syndrome in connection with the risk
985]. With respect to obesity and the insulin of vascular disease, the occurrence of insulin
resistance syndrome, elevated serum uric acid resistance, the presence of ischemic heart dis-
levels have been observed with these clinical ease, cardiovascular mortality, and the presence
findings as well [986–988]. Further evidence of of accompanying disorders at the time of the ini-
the association between gout and obesity comes tial diagnosis of gout [910, 913, 999–1011].
from studies showing that the risk of gout dimin- Hyperuricemia is associated with an increased
ishes with weight reduction [897, 989]. Recently, risk for vascular disease in some studies, but
obesity, hypertension, lipidemia, and hyperglyce- other investigations only demonstrate a link
mia have been considered as a cluster of findings between gout and death from cardiovascular dis-
characteristic of a syndrome of multiple interre- eases but not with hyperuricemia [999, 1002].
lated conditions called the metabolic syndrome The prevalence of gout as well as insulin resis-
[990]. This so-called metabolic syndrome is diag- tance is higher in patients with gout than in nor-
nosed on the basis of the following revised crite- mal healthy control groups [1000]. Patients with
ria: abdominal obesity (waist circumference of gout in one study were all shown to manifest the
>102 cm in men and >88 cm in women), hyper- metabolic syndrome, and 16 % had ischemic
triglyceridemia (>150 mg/dl or 1.69 mmol/l), low heart disease [913]. A recent review of the clini-
high-density lipoprotein (HDL) cholesterol cal status of patients with respect to their first epi-
(<40 mg/dl or 1.04 mmol/l in men and <50 mg/dl sode of gout and the diagnosis of the metabolic
or 1.29 mmol/l in women), a blood pressure read- syndrome and its components and complications
ing of less than 130/80 mmHg, and a fasting glu- showed that 90 % of the first attacks of gout pre-
cose level of more than 100 mg/dl or more than ceded the diagnosis of the metabolic syndrome,
5.6 mmol/l [991]. Two facts stress the importance its components, and its complications [1001]. At
of identifying the metabolic syndrome in gouty the time of the inclusion of gouty patients into
352 9 Management of Hyperuricemia and Gout

this study (a mean of 13.7 years after the first Usually, gout is associated with type IV hyper-
attack of gout), 93 % of these patients had at least lipidemia or hypertriglyceridemia. These dyslipi-
one associated disorder. The most common asso- demias occur in about 25–60 % of gouty patients
ciated disorders included hypertriglyceridemia, [832, 865]. It is also worth remembering that
63 %; obesity, 54 %; hypertension, 45.6 %; meta- roughly two-thirds of the purines in the human
bolic syndrome, 40 %; hyperglycemia, 37 %; low are produced by cell and tissue turnover, whereas
HDL, 17 %; diabetes mellitus, 15 %; chronic only one-third of purines are contributed by
renal failure, 17 %; and ischemic heart disease, dietary intake.
6.6 %. These data emphasize the need for careful Alcohol ingestion in excess and its association
follow-up evaluations of patients with gout. with gout has been known since antiquity [865,
Another study showed a 2:1 ratio of the preva- 868, 1014–1016]. Beer has a special capacity to
lence of essential hypertension, hyperlipidemia, raise the serum uric acid levels and put patients at
diabetes mellitus without complications, and cor- risk for gouty episodes since it contains guanos-
onary atherosclerosis in patients with gout as ine, a purine that is easily converted enzymati-
compared with subjects without gout [910]. A cally to uric acid. Finally, many patients will tell
strong correlation has also been shown to exist their physician that particular foods or alcoholic
between diminished urinary uric acid levels and beverages will trigger an attack of gout. In these
the presence of the metabolic syndrome [1010]. cases, physicians should heed the patient’s infor-
The proposed mechanism for this abnormality of mation and ask the patient to restrict their intake
uric acid excretion is an impairment of renal uric of the offending agent.
acid excretion mediated by hyperinsulinemia- There are some general rules that are useful
enhanced proximal tubule sodium reabsorption when considering dietary modifications for
and the diminished uric acid excretion [1009, patients with gout. Overweight patients should
1011]. make a concerted effort to lose weight by diet and
Although medications are usually the means exercise. Measurement of fasting blood lipids is
by which recurrent gouty episodes are controlled, essential to exclude hyperlipidemia as a contrib-
there are certain dietary restrictions that may be uting factor to underlying health problems.
useful as well. It has been shown that meat and Appropriate diets and medications should be
seafood intake are associated with an increased instituted to decrease the low-density lipoprotein
risk of gout. Meat including beef, lamb, and pork (LDL) levels (bad cholesterol) and increase the
as well as bologna, sausage, salami, bacon, hot high-density lipoprotein (HDL) levels (good cho-
dogs, chicken, turkey, hamburger, chicken liver, lesterol) if hyperlipidemia exists. Gouty patients
and beef liver are the most well-characterized should also be encouraged to eat a balanced diet
offenders as inducers of gouty episodes. Seafood with less red meat and less seafood than usual. In
associated with an increase risk of gout include general, the three foods/beverages that place
tuna, dark fish, shrimp, lobster, clams, and scal- patients at risk for gout are red meat, beer, and
lops. There are also some purine-rich vegetables fructose, a sugar commonly found in sugar-
like peas, lentils, spinach, mushrooms, oatmeal, sweetened soft drinks. Finally, patients with gout
and cauliflower, but they do not appear to increase should decrease their use of hard liquor and beer
the risk of gout [865, 866, 882, 1012, 1013]. In with a substitution of wine if necessary.
contrast to these high-purine-containing foods, Hyperuricemia is the precursor of gout and
there are some foods with low-purine content kidney stones, and therefore, the mechanisms
such as milk, cheese, yogurt, ice cream, breads, that increase serum uric acid levels become
pasta, vegetables other than those with a high- important to understand and modify, if necessary.
purine content, fruits, nuts, sugars, and sweets. Serum uric acid levels represent a balance
Of course, it is important to recognize that beef, between cell and tissue breakdown, the synthesis
cheese, ice cream, and some other foods are not of uric acid and its precursors by the de novo
appropriate for those with lipid abnormalities. purine and salvage pathways, the renal elimination
Summary 353

of urate, and the dietary intake of purines and agent such as diuretics that may account for the
other chemicals known to influence urinary uric presence of hyperuricemia and/or gout. Finally, a
acid excretion. Genetic parameters have been vague history of joint complaints with an elevated
described in monogenic disorders including serum uric acid does not constitute a diagnosis of
hypoxanthine-guanine phosphoribosyltransferase gout, but intermittent episodes of an acute
deficiency, phosphoribosylpyrophosphate syn- inflammatory joint process with redness and
thetase overactivity, familial juvenile hyperurice- severe pain are suspicious of an attack of acute
mic nephropathy, and glycogen storage disease, gouty arthritis. Further evidence in support of the
but these disorders only represent about 10 % of shortcomings associated with the diagnosis of the
the patients with gout. The majority of patients causes of hyperuricemia and gout are apparent
with gout have a drug-induced or undefined alter- when the combination of clinical analysis and
ation in their renal elimination of uric acid. imaging data as proposed by the American
College of Rheumatology (ACR) criteria is used
as an alternative to the absolute diagnosis pro-
Summary vided by the detection of MSU crystals. The fact
is that the validity of the ACR criteria as well as
Despite the foregoing knowledge base, many the Rome and New York criteria leads to less than
patients with hyperuricemia and gout remain optimal performance results [1029, 1030]. To
improperly diagnosed, inappropriately treated for emphasize further this defect in the use of clinical
a disease they often do not have, insufficiently criteria alone for the diagnosis of gout, the ACR
managed by the inadequate use of available med- criteria only have a sensitivity of 68 % and a
ications, and poorly followed to ensure the pri- specificity of 78 %. As a supplement to the his-
mary objective of eliminating urate crystal torical data, a thorough physical examination is
deposits from their joints and other tissues and to required including a search for tophi and an
prevent the occurrence of gouty episodes [1017– abdominal examination to elicit enlargements of
1026]. Gout represents the end expression of the liver, spleen, or kidney; an estimation of renal
many different causes, and such etiologic factors function (serum creatinine); a careful neurologi-
are often not evaluated. This is especially true for cal examination to exclude Lesch-Nyhan vari-
comorbid conditions like the metabolic syndrome ants; and a synovial fluid or tophus aspiration to
and its components (hypertension, insulin resis- document the presence or absence of MSU
tance, dyslipidemia, and abdominal obesity) crystals.
[911, 1027, 1028]. Furthermore, many physicians With respect to the physical examination, it is
only treat acute gouty episodes and fail to recog- important to recognize that the involvement of the
nize and manage the deformities and functional first metatarsal phalangeal (MTP) joint may not
impairments caused by tophaceous deposits and represent podagra but infection, other crystal-
their potential to suppress kidney function. induced arthropathies, or psoriatic arthritis
A careful and complete history should provide [1031–1034]. Although most patients with gout
information concerning other family members manifest classical podagra at some time during
with gout, the potential for or the presence of the lifetime of their disease, it is essential to rec-
renal disease including nephrolithiasis, and the ognize that many unusual sites such as the tem-
presence of any neurological deficits in patients poromandibular joint, manubriosternal joint,
or his/her family. In addition, age and sex of the sternoclavicular joint, acromioclavicular joint,
patient and affected family members are likely to sacroiliac joint, lumbar spine, hip joints, symphy-
provide clues to any underlying genetic disorder sis pubis joint, and flexor tenosynovitis with car-
associated with gout. The latter is especially true pal tunnel syndrome may be involved in acute and
if young males or females are documented to chronic gout [1035–1043]. Finally, tophaceous
have hyperuricemia or gout. A careful history of gout may mimic other disorders as a result of its
medications may often reveal a likely causative unusual location and its tumor-like appearance
354 9 Management of Hyperuricemia and Gout

[1044–1046]. A most significant reason for per- Glucocorticoids are effective in acute gout and
forming a synovial fluid aspiration is the possible are the drugs of choice in patients with significant
concomitant occurrence of a septic arthritis along renal disease and other comorbidities. They may
with gout [1047]. be given orally in doses beginning with 40–60 mg/
day of prednisone or its equivalent tapering off
over a period of about 10 days. Intra-articular glu-
Summary of Principles cocorticoids are effective and safe for the treat-
of Management ment of acute gout, especially when monoarticular.
At the same time, samples of synovial fluid may
Lifestyle be obtained for crystal analysis and culture.

Attention should be directed to obesity, hyperten-


sion and renal insufficiency, diabetes, alcohol Chronic Gout
intake, and drugs which might elevate serum uric
acid levels. Diuretics should be avoided for treat- Urate-lowering treatment is indicated in patients
ment of hypertension if possible. Losartan may with recurrent acute attacks of gout, tophi on
be a good choice since it has a modest uricosuric physical examination, or radiographic changes of
effect. Fenofibrate also has some urate-lowering gout. The therapeutic goal of urate-lowering ther-
effect, making it a good choice for treating hyper- apy is to reduce the serum uric acid to levels
lipidemia in patients with gout. below the saturation level of monosodium urate
Treatment decisions also depend on the num- in order to dissolve monosodium urate that is
ber of previous acute attacks of gout, age of the present in tissues and prevent further precipita-
patient, presence of tophi, and radiographic tion. Practically, the therapeutic goal is a serum
signs. uric acid level of <6.0 mg/dl or <360 mmol/l. It is
very important to achieve this goal with adequate
doses of the urate-lowering drug, or treatment
Acute Attacks will be ineffective. It is generally recommended
that urate-lowering therapy not be started during
Oral colchicine and NSAIDs are the first-line an acute attack. However, if a patient is already
agents for the treatment of acute attacks of gout. taking urate-lowering therapy when an attack
Colchicine has been shown to be effective in occurs, the drug may be continued during the
lower doses than commonly used in the past, with attack.
less toxicity. A study of 1.0 mg followed by The most commonly used urate-lowering drug
0.5 mg 1 h later compared to 1.0 mg followed by is the xanthine oxidase inhibitor allopurinol. It
0.5 mg every 2 h until GI toxicity has been pub- should be started at a dose of 100 mg/day in
lished. There was equal anti-inflammatory effec- patients with normal renal function and increased
tiveness of the two regimens at 48 h with reduced every 2–4 weeks until the target level of serum
toxicity in the low-dose group. uric acid is reached. The dose needed is variable
The NSAIDs are also effective agents for from patient to patient. Doses as high as 800 mg/
acute gout. Studies have shown that several day have been approved by the US Food and
NSAIDs have comparable effectiveness. Upper Drug Administration. It is also important to warn
GI toxicity with bleeding is a major concern with patients of possible side effects since up to 5 % of
these agents. This risk may be reduced with con- patients may develop allergy to allopurinol, and a
comitant use of proton pump inhibitors or with much lower number can develop the potentially
Cox-2 selective inhibitors. Cardiovascular risks life-threatening allopurinol hypersensitivity
occur with NSAIDs, but naproxen appears to syndrome.
have less cardiovascular risk than other Another useful urate-lowering drug is febuxo-
NSAIDs. stat. This is a particularly useful drug for patients
References 355

who are allergic to allopurinol, and since this 6. Kuzell WC, Schaffarzick RW, Naugler WE, et al.
drug has been available, allopurinol desensitiza- Some observations on 520 gouty patients. J Chronic
Dis. 1955;2:645.
tion is rarely necessary. Furthermore, it can be 7. Barlow KA, Beilin LJ. Renal disease and primary
used in patients with moderate renal insufficiency gout. Q J Med. 1968;37:79.
since it is metabolized by the liver. It should be 8. Rapado A. Relationship between gout and arterial
started at a dose of 40 mg/day and increased to hypertension. In: Sperling O, DeVries A, Wyngaarden
JB, editors. Purine metabolism in man, vol. 41B. New
120 mg/day if needed. York: Plenum Press; 1974. p. 451.
Alternative urate-lowering drugs are the uri- 9. Waller PC, Ramsay LE. Predicting gout in diuretic-
cosuric agents. The most commonly used is treated hypertensive patients. J Hum Hypertens.
probenecid, and sulfinpyrazone is used less com- 1989;3:457.
10. Laut A. Diuretics: clinical pharmacology and thera-
monly. However, normal or only slightly impaired, peutic use (part II). Drugs. 1985;29:162.
renal function is required, and these drugs should 11. Steele TH. Diuretic-induced hyperuricemia. Clin
not be used if the patient has had uric acid renal Rheum Dis. 1950;2:37.
stones. As with other urate-lowering drugs, the 12. Langford HS, Blaufox MD, Borhani NO, et al. Is thi-
azide-produced uric acid elevation harmful? Analysis
doses of uricosuric drugs must be adequate to of data from the Hypertension Detection and
reduce the serum uric acid to the target levels. Follow-up Program. Arch Intern Med. 1987;147:645.
Prophylaxis against acute attacks is required 13. Klemperer F, Bauer W. Influence of aspirin on urate
for at least the first few months of urate-lowering excretion. J Clin Invest. 1944;23:950.
14. Crone C, Lassen W. The effect of salicylic acid and
therapy, regardless of which drug is used. The acetylsalicylic acid on uric acid excretion and plasma
most common agent is colchicine, 0.5 or 0.6 mg uric acid concentration in normal human subjects.
once or twice daily as tolerated. If colchicine is Acta Pharmacol. 1955;11:355.
not tolerated, an NSAID may be used, although 15. Yu T-F, Gutman AB. Paradoxical retention of uric
acid by uricosuric drugs in low dosage. Proc Soc Exp
the evidence for efficacy of NSAID prophylaxis Biol Med. 1955;90:542.
is limited. While a period of 6 months of prophy- 16. Gutman AB, Yu T-F. Benemid (p-[di-N-propylsulfamyl]
laxis is sometimes recommended, it may be benzoic acid) as a uricosuric agent in chronic gouty
required for much longer periods since it may arthritis. Trans Assoc Am Physicians. 1951;64:279.
17. Steele TH, Bonar G. Origins of uricosuric response. J
take longer periods to dissolve all of the tissue Clin Invest. 1973;52:1368.
urate, especially in patients with tophaceous 18. Shapiro M, Hyde L. Hyperuricemia due to pyrazin-
gout. amide. Am J Med. 1957;23:596.
19. Wood RHN. Salicylates. Bull Rheum Dis.
1963;13:297.
20. Gershon SL, Fox IH. Pharmacologic effects of nico-
tinic acid on human purine metabolism. J Lab Clin
References Med. 1974;84:179.
21. Postlewaite AE, Kelley WN. Studies on the mecha-
1. Rosenfeld JB. Effect of long-term allopurinol on serial nism of ethambutol-induced hyperuricemia. Arthritis
GFR in normotensive and hypertensive hyperuricemic Rheum. 1972;15:403.
subjects. Adv Exp Med Biol. 1974;41B:581. 22. Elion GB, Kovensky A, Hitchings GH, et al. Metabolic
2. Steele TH, Oppenheimer S. Factors affecting urate studies of allopurinol, an inhibitor of xanthine oxi-
excretion following diuretic administration in man. dase. Biochem Pharmacol. 1966;15:863.
Am J Med. 1969;47:564. 23. Elion GB, Yu T-F, Gutman AB, Hitchings GH. Renal
3. Steele TH. Evidence of altered renal urate reabsorption clearance of oxypurinol, the chief metabolite of
during changes in volume of the extracellular fluid. J allopurinol. Am J Med. 1968;45:69.
Lab Clin Med. 1969;74:288. 24. Elion GB. Allopurinol and other inhibitors of urate
4. Schirmeister J, Man NK, Hallaner W. Study of renal synthesis. In: Kelley WN, Weiner IM, editors. Uric
and extrarenal factors involved in hyperuricemia acid. New York: Springer; 1978. p. 485–514.
induced by furosemide. In: Peters G, Roch-Ramal F, 25. Maisey DN, Brown RD. Acetazolamide and symp-
editors. Progress in nephrology. New York: Springer; tomatic metabolic acidosis in mild renal failure. Br
1969. p. 59. Med J. 1981;283:1527.
5. Schultz G, Gesaft G, Losert W, et al. Biochemische 26. Nolph KD, Sarkin MI. Peritoneal dialysis in acute
grundlagen der Diazoxid-hyperglykamie. Naunyn renal failure. In: Brenner BM, Lazarus JM, editors.
Schmeidebergs Arch Pharmacol Exp Pathol. 1966; Acute renal failure. New York: Churchill Livingstone;
253-A:372. 1988. p. 809–38.
356 9 Management of Hyperuricemia and Gout

27. Rundles RW. The development of allopurinol. Arch of tyrosine phosphorylation by colchicine. J Clin
Intern Med. 1985;145:1492. Invest. 1993;92:1722.
28. Zemer D, Revach M, Pras M. A controlled trial of 46. Kitagawa S, Takaku F. Effect of microtubule disrupt-
colchicine in preventing attacks of familial ing agents on superoxide production in human poly-
Mediterranean fever. N Engl J Med. 1976;291:832. morphonuclear leukocytes. Biochim Biophys Acta.
29. Taborn J, Bole GG, Thompson GR. Colchicine sup- 1982;719:589.
pression of local and systemic inflammation due to 47. Okamura N, Hanakura K, Kodakarai M, Ishibashi S.
calcinosis universalis in chronic dermatomyositis. Cooperation of cytochalasin D and anti-microtubular
Ann Intern Med. 1979;89:648. agents in stimulating superoxide anion production in
30. Tabatai MR, Cummings NA. Intravenous colchicine polymorphonuclear leukocytes. J Biochem (Tokyo).
in the treatment of acute pseudogout. Arthritis Rheum. 1980;88:139.
1980;23:370. 48. Phelps P. Polymorphonuclear leukocyte motility
31. Ravid M, Shapira J, Kedar I, Feigl D. Regression of in vitro. II. Stimulating effect on monosodium urate
amyloidosis secondary to granulomatous ileitis fol- crystals and urate in solution; partial inhibition by
lowing surgical resection and colchicine administra- colchicine and indomethacin. Arthritis Rheum.
tion. Acta Hepatogastroenterol. 1979;26:513. 1969;12:189.
32. Ravid M, Robson M, Kedar I. Prolonged colchicine 49. Spilberg I, Gallacher A, Mandell B, Rosenberg D. A
treatment in four patients with amyloidosis. Ann mechanism for action of non-steroidal anti-inflammatory
Intern Med. 1977;87:568. agents in calcium pyrophosphate dehydrate (CPPD)
33. Mizushima Y, Matumira NS, Mosi M, et al. Colchicine crystal-induced arthritis. Agents Actions. 1977;7:153.
in Behcet’s disease. Lancet. 1977;2:1037. 50. Zurier RB, Hoffstein S, Weissman G. Mechanism of
34. Miyachi Y, Taniguchi S, Ozaki M, Horio T. Colchicine lysosomal enzyme release from human leukocytes. I.
in the treatment of cutaneous manifestations of Effect of cyclic nucleotides and colchicine. J Cell
Behcet’s disease. Br J Dermatol. 1981;104:67. Biol. 1973;58:27.
35. Meed SD, Spilberg I. Successful use of colchicine in 51. Wallace SL, Ertel NH. Pharmacology of drugs used in
acute polyarticular pseudogout. J Rheumatol. the treatment of acute gout. In: Kelley WN, Weiner
1981;8:689. IM, editors. Uric acid. New York: Springer; 1978.
36. Kaplan H. Further experience with colchicine in the p. 525–40.
treatment of sarcoid arthritis. N Engl J Med. 52. Wallace SL. Colchicine. Semin Arthritis Rheum.
1963;268:761. 1974;3:369.
37. Ainarella CA, Wolff SM, Goldfinger SE. Colchicine 53. Borisy CG, Taylor EW. The mechanism of action of
therapy for familial Mediterranean fever. A double- colchicine: binding of colchicine-3H to cellular pro-
blind study. N Engl J Med. 1976;291:934. tein. J Cell Biol. 1967;34:525.
38. Aktulga E, Altac M, Muftinoglu A, et al. A double- 54. Schelanski MC, Taylor EW. Isolation of protein sub-
blind study of colchicine in Behcet’s disease. unit from microtubules. J Cell Biol. 1967;34:549.
Haematologica. 1980;65:399. 55. Wright DG, Malawista SE. The mobilization and
39. Hazen PG, Michel B. Management of necrotizing extracellular release of granular enzymes from human
vasculitis with colchicine. Improvement in patients leukocytes during phagocytosis. Arthritis Rheum.
with cutaneous lesions and Behcet’s syndrome. Arch 1971;14:425.
Dermatol. 1979;115:1303. 56. Spilberg I, Mandell B, Mehta J, et al. Mechanism of
40. Terkeltaub RA, Furst DE, Bennett K, et al. High ver- colchicine action in acute urate crystal-induced arthri-
sus low dosing of oral Colchicine for early acute gout tis. J Clin Invest. 1979;64:775.
flare. Arthritis Rheum. 2010;62:1060–8. 57. Serhan CN, Lundberg U, Weissman G, Samuelsson
41. Tse RL, Phelps P. Polymorphonuclear leukocyte B. Formation of leukotrienes and hydroxyl acids by
motility in vitro. V. Release of chemotactic activity human neutrophils and platelets exposed to monoso-
following phagocytosis of calcium pyrophosphate dium urate. Prostaglandins. 1984;27:563.
crystals, diamond dust, and urate crystals. J Lab Clin 58. Kastner DL, Aksentijevich I, Goldbach-Mansky R.
Med. 1970;76:403. Autoinflammatory disease reloaded: a clinical per-
42. Trung PH. Chemotaxis of human leukocytes. II. spective. Cell. 2010;140:784–90.
Effects of lectins, colchicine, cytochalasin B, cyclic 59. Poubelle PE, de Medicis R, Naccache PH.
nucleotides and immuno-stimulatory products. Monosodium urate and calcium pyrophosphate crys-
Biomedicine. 1977;30:121. tals differentially activate the excitation-coupling
43. Phelps P, McCarty Jr DJ. Crystal-induced arthritis. sequence of human neutrophils. Biochem Biophys
Postgrad Med. 1969;45:84. Res Commun. 1987;149:649.
44. Maleich HL, Root RK, Gallin JI. Structural analysis 60. Onello E, Traynor-Kaplan A, Sklar L, Terkeltaub R.
of human neutrophil migration. Centriole, microtu- Mechanism of neutrophil activation by an unop-
bule and microfilament orientation and function dur- sonized inflammatory particulate. Monosodium urate
ing chemotaxis. J Cell Biol. 1977;75:666. crystals induce pertussis toxin-insensitive hydrolysis
45. Roberge CJ, Gaudry M, de Medicis R, et al. Crystal- of phosphatidylinositol 4,5-bisphosphate. J Immunol.
induced neutrophil activation. IV. Specific inhibition 1991;146:4289.
References 357

61. Naccache PH, Bourgoin S, Plante E, et al. Crystal- 80. Bruns BJ. Colchicine toxicity. Australas Ann Med.
induced neutrophil activation. II. Evidence for the 1969;17:341.
activation of a phosphatidylcholine-specific phospho- 81. Carr AA. Colchicine toxicity. Arch Intern Med.
lipase D. Arthritis Rheum. 1993;36:117. 1965;115:29.
62. Gaudry M, Roberge CJ, de Medicis R, et al. Crystal- 82. Kontos HA. Myopathy associated with chronic colchi-
induced neutrophil activation. III. Inflammatory cine toxicity. N Engl J Med. 1962;266:38.
microcrystals induce a distinct pattern of tyrosine 83. Roberts WN, Liang MH, Stern SH. Colchicine in
phosphorylation in human neutrophils. J Clin Invest. acute gout. Reassessment of risks and benefits. JAMA.
1993;91:1649. 1987;257:1920.
63. Ertel NH, Wallace SL. Measurement of colchicine in 84. Malawista SE. Colchicine: a common mechanism for
urine and peripheral leukocytes. Clin Res. 1971;19:348. its anti-inflammatory and anti-meiotic effects.
64. Wallace SL, Omokoku B, Ertel NH. Colchicine Arthritis Rheum. 1968;11:191.
plasma levels: Implications as to pharmacology and 85. Auerbach R, Orentrick N. Alopecia and ichthyosis sec-
mechanism of action. Am J Med. 1970;48:443. ondary to allopurinol. Arch Dermatol. 1968;98:104.
65. Keibman J, Haines KA, Rich AM, et al. Colchicine 86. Riggs JE, Schochet SS, Gutmann L, et al. Chronic
inhibits ionophore-induced formation of leukotriene human colchicine neuropathy and myopathy. Arch
B4 by human neutrophils: the role of microtubules. J Neurol. 1986;43:521.
Immunol. 1986;136:1027. 87. Anonymous. Colchicine myoneuropathy. Lancet. 1987;
66. Rich AM, Hoffstein ST. Inverse correlation between 1:668.
neutrophil microtubule numbers and enhanced ran- 88. Kuncl RW, Duncan G, Watson D, et al. Colchicine
dom migration. J Cell Sci. 1981;48:181. myopathy and neuropathy. N Engl J Med. 1987;
67. Oliver JM, Albertini DF, Berlin RD. Effects of gluta- 316:1562.
thione-oxidizing agents on microtubule assembly and 89. Rieger EH, Halasz NA, Wahlstrom HE. Colchicine
microtubule-dependent surface properties of human neuromyopathy after renal transplantation.
neutrophils. J Cell Biol. 1976;71:921. Transplantation. 1990;49:1196.
68. Malawista SE, Oliver JM, Rudolf SA. Microtubules 90. Kirchin VS, Southgate HJ, Beard RC. Colchicine: an
and cyclic AMP in human leukocytes: on the order of unusual cause of a reversible azoospermia. BJU Int.
things. J Cell Biol. 1978;77:881. 1999;83:156.
69. Olmstead SL, Borisy GG. Microtubules. Annu Rev 91. Wyngaarden JB, Kelley WN. Gout and hyperurice-
Biochem. 1973;42:507. mia. New York: Grune & Stratton; 1976.
70. Wallace SL. Trimethylcolchicinic acid in the treat- 92. Smith WL, Garavito RM, DeWitt DL. Prostaglandin
ment of acute gout. Ann Intern Med. 1961;54:274. endoperoxides H synthases (cyclooxygenases)-1 and −2.
71. Webb DJ, Chodos RB, Mahar CQ, Faloon WW. J Biol Chem. 1996;271:33157.
Mechanism of vitamin B12 malabsorption in patients 93. Hla T, Neilson K. Human cyclooxygenase-2 cDNA.
receiving colchicine. N Engl J Med. 1980;279:845. Proc Natl Acad Sci USA. 1992;89:7384.
72. Rubulis A, Rubert M, Faloon WW. Cholesterol lower- 94. O’Banion MK, Winn VD, Young DA. cDNA cloning
ing, fecal bile acid and sterol changes during neomy- and functional activity of a glucocorticoid regulated
cin and colchicine. J Clin Nutr. 1970;23:1251. inflammatory cyclooxygenase. Proc Natl Acad Sci
73. Gillespie E, Levine RJ, Malawista SE. Histamine USA. 1992;89:4888.
release from rat peritoneal mast cells: Inhibition by 95. Jones DA, Carlton DP, McIntyre TM, et al. Molecular
colchicine and potentiation by deuterium oxide. J cloning of human prostaglandin endoperoxide syn-
Pharmacol Exp Ther. 1968;164:158. thase type II and demonstration of expression in
74. Bhisey AN, Freed JH. Ameboid movement induced in response to cytokines. J Biol Chem. 1993;268:
cultured macrophages by colchicine and vinblastine. 9049.
Exp Cell Res. 1971;64:419. 96. Percival MD, Ouillet M, Vincent CJ, et al. Purification
75. Harris ED, Krane SM. Effect of colchicine on collage- and characterization of recombinant cyclooxyge-
nase in cultures of rheumatoid synovium. Arthritis nase-2. Arch Biochem Biophys. 1994;315:111.
Rheum. 1971;14:669. 97. O’Neill G, Ford-Hutchinson A. Expression of mRNA
76. Robinson DR, Smith H, Levine L. Prostaglandin syn- for cyclooxygenase-1 and cyclooxygenase-2 in human
thesis by human synovial cultures and its stimulation tissues. FEBS Lett. 1992;330:156.
by colchicine. Arthritis Rheum. 1973;16:129. 98. O’Sullivan MG, Chilton FH, Huggins EMJ, et al.
77. Walaszek EJ, Kocsis JJ, Leroy GV, Geiling EMK. Lipopolysaccharide priming of alveolar macrophages
Studies on the excretion of radioactive colchicine. for enhanced synthesis of prostanoids involves induc-
Arch Int Pharmacodyn Ther. 1960;125:371. tion of a novel prostaglandin H synthase. J Biol Chem.
78. Smallwood JI, Malawista SE. Colchicine, crystals, 1992;267:14547.
and neutrophil tyrosine phosphorylation. J Clin Invest. 99. Sano H, Hla T, Maier JAM, et al. In vivo cyclooxyge-
1993;92:1602. nase expression in synovial tissue of patients with
79. Gaultier M, Kaufer A, Bismuth C, et al. Donnees rheumatoid arthritis and osteoarthritis and rats with
actuelles sur l’intoxication aigue par la colchicine. adjuvant and streptococcal cell wall arthritis. J Clin
Ann Med Interne. 1969;120:605. Invest. 1992;89:97.
358 9 Management of Hyperuricemia and Gout

100. Crofford LJ, Wilder RL, Ristimaki AP, et al. 118. Eberl R, Dunky A. Meclofenamate sodium in the
Cyclooxygenase-1 and −2 expression in rheumatoid treatment of acute gout. Arzneimittelforschung.
synovial tissues: effects of interleukin-1b. J Clin 1983;33:641.
Invest. 1994;93:1095. 119. Brogen RN, Heil RC, Speight TM, Avery GS.
101. Smith WL, DeWitt DL. Prostaglandin endoperoxides Sulindac: a review of its pharmacological properties
H synthase-1 and −2. Adv Immunol. 1996;62:167. and therapeutic efficacy in rheumatic diseases.
102. Masferrer JL, Zweifel BS, Seibert K, et al. Drugs. 1978;16:97.
Endogenous glucocorticoids regulate an inducible 120. Duggan DE, Hare LE, Ditzler CA, et al. The disposi-
cyclooxygenase enzyme. Proc Natl Acad Sci USA. tion of sulindac. Clin Pharmacol Ther. 1977;21:326.
1992;89:3917. 121. Pouliot M, James MJ, McColl SR, et al. Monosodium
103. Masferrer JL, Zweifel BS, Seibert K, et al. Selective urate microcrystals induce cyclooxygenase-2 in
inhibition of inducible cyclooxygenase 2 in vivo is human monocytes. Blood. 1998;91:1769.
anti-inflammatory and nonulcerogenic. Proc Natl 122. Vance JR, Bakhle YS, Botting RM. Cyclooxygenase
Acad Sci USA. 1994;91:3228. 1 and 2. Annu Rev Pharmacol Toxicol. 1998;38:97.
104. Langman MJS, Weil J, Wainwright P, et al. Risk of 123. Breyer MD, Harris RC. Cyclooxygenase 2 and the
bleeding peptic ulcer associated with individual non- kidney. Curr Opin Nephrol Hypertens. 2001;10:89.
steroidal anti-inflammatory drugs. Lancet. 1994;343: 124. Giercksky KE, Haglund U, Rask-Madsen J. Selective
1075, 1302. inhibitors of COX-2 – are they safe for the stomach?
105. Murray MD, Brater DC. Renal toxicity of the non- Scand J Gastroenterol. 2000;35:1121.
steroidal anti-inflammatory drugs. Annu Rev 125. Emery P. Cyclooxygenase-2: a major therapeutic
Pharmacol Toxicol. 1993;32:435. advance? Am J Med. 2001;110:42S.
106. Meade EA, Smith WL, DeWitt DL. Differential 126. Furst DE. Meloxicam: selective COX-2 inhibition in
inhibition of prostaglandin endoperoxides synthase clinical practice. Semin Arthritis Rheum. 1997;
(cyclooxygenase) isozymes by aspirin and other 26(Suppl):21.
nonsteroidal anti-inflammatory drugs. J Biol Chem. 127. Brzozowski T, Konturek PC, Konturek SJ, et al.
1993;268:6610. Involvement of cyclooxygenase (COX)-2 products in
107. Mitchell JA, Akarasereenont P, Theimermann C, acceleration of ulcer healing by gastrin and hepatocyte
et al. Selectivity of nonsteroidal anti-inflammatory growth factor. J Physiol Pharmacol. 2000;51(Pt 1):751.
drugs as inhibitors of constitutive and inducible 128. Buttgereit F, Burmester GR, Simon LS. Gastrointestinal
cyclooxygenases. Proc Natl Acad Sci USA. 1994;90: toxic side effects on nonsteroidal anti-inflammatory
11693. drugs and cyclooxygenase-2-specific inhibitors. Am J
108. FitzGerald GA, Patrono C. The coxibs, selective Med. 2001;110(Suppl 3A):13S.
inhibitors of cyclooxygenase-2. N Engl J Med. 129. Everts B, Wahrborg P, Hedner T. COX-2-specific
2001;345:433–42. inhibitors – the emergence of a new class of analge-
109. Solomon EH, Glynn RJ, Rothman KJ, et al. Subgroup sic and anti-inflammatory drugs. Clin Rheumatol.
analyses to determine cardiovascular risk associated 2000;19:331.
with nonsteroidal antiinflammatory drugs and coxibs 130. Silverstein FE, Faich G, Goldstein JL, et al.
in specific patient groups. Arthritis Rheum. Gastrointestinal toxicity with celecoxib vs nonsteroi-
2008;59:1097–104. dal anti-inflammatory drugs for osteoarthritis and
110. Amer M, Bead VR, Bathon J, et al. Usse of nonsteroi- rheumatoid arthritis: the CLASS study: a random-
dal anti-inflammatory drugs in patients with cardio- ized controlled trial. Celecoxib long-term arthritis
vascular disease. Cardiol Rev. 2010;18:204–12. safety study. JAMA. 2000;284:1247.
111. Karachalios GN, Donas G. Sulindac in the treatment of 131. Williams RL, Upton RA, Buskin JN, Jones RM.
acute gouty arthritis. Int J Tissue React. 1982;4:297. Ketoprofen-aspirin interactions. Clin Pharmacol
112. Schweitz MC, Nashel DJ, Alepa FP. Ibuprofen in the Ther. 1981;30:226.
treatment of acute gouty arthritis. JAMA. 1978; 132. Eberl R. Long-term experience with meclofenamate
289:34. sodium. Arzeimittelforschung. 1983;33:667.
113. Wanasukapent S, Lestratenakul Y, Rubenstein HM. 133. Marks JS. Steatorrhea complicating therapy with
Effect of fenoprofen calcium on acute gout. Arthritis mefenamic acid. Br Med J. 1975;4:442.
Rheum. 1976;19:933. 134. Multz CV, Brobyn RD, Caldwell JR. Sodium
114. Wilkens RF, Case JB, Huix FJ. Treatment of acute meclofenamate (meclomen) vs. aspirin for rheuma-
gout with Naprosyn. J Clin Pharmacol. 1976;15:363. toid arthritis. Curr Ther Res. 1978;23:572.
115. Bluestone RH. Safety and efficacy of piroxicam in 135. Preston SN. Safety of sodium meclofenamate
the treatment of gout. Am J Med. 1979; (Meclomen). Curr Ther Res. 1978;23:S107.
72(Suppl):66. 136. Garg V, Jusko WJ. Effect of indomethacin on the
116. Tamisier JN. Ketoprofen. Clin Rheum Dis. 1979; pharmacokinetics and pharmacodynamics of predni-
5:381. solone in rats. J Pharm Sci. 1994;83:747.
117. Petera P, Gainmayer K, Tausch G. Treatment of acute 137. Kimberly RP, Bowden RE, Keiser HR, et al.
gout attacks with tolmetin. Wein Med Wochenschr. Reduction of renal function by newer nonsteroidal
1982;132:43. anti-inflammatory drugs. Am J Med. 1978;64:804.
References 359

138. Miller KP, Lazar EJ, Fotino S. Severe hyperkalemia out concomitant administration of aluminum phos-
during piroxicam therapy. Arch Intern Med. phate. Eur J Clin Pharmacol. 1981;19:305.
1984;144:2414. 156. Segre EJ, Sevelius H, Varady J. Effects of antacids
139. Frais MA, Burgess ED, Mitchell LB. Piroxicam- on naproxen absorption. N Engl J Med. 1974;
induced renal failure and hyperkalemia. Ann Intern 291:582.
Med. 1983;99:129. 157. Slattery JT, Levy G. Effect of ibuprofen on protein-
140. Mitnick PD, Klein Jr WJ. Piroxicam-induced renal binding of warfarin in human serum. J Pharm Sci.
disease. Arch Intern Med. 1984;144:63. 1977;66:1060.
141. Chiolero A, Wurzner G, Burnier M. Selective inhibi- 158. Jain A, McMahon FG, Slattery JT, Levy G. Effect of
tors of type 2 cyclooxygenase: less renal effects than naproxen on steady state serum concentration and
the classical nonsteroidal anti-inflammatory drugs anticoagulant activity of warfarin. Clin Pharmacol
on renal function? Nephrologie. 2000;21:425. Ther. 1979;25:61.
142. Brater DC. Effects of Nonsteroidal anti-inflammatory 159. Slattery JT, Levy G, Jain A, McMahon FG. Effect of
drugs on renal function: focus on cyclooxygenase-2- naproxen on the kinetics of elimination and the anti-
selective inhibition. Am J Med. 1999;107:655. coagulant activity of a single dose of warfarin. Clin
143. Stichtenoth DO, Frohlich JC. COX-2 and the kid- Pharmacol Ther. 1979;25:51.
neys. Curr Pharm Des. 2000;6:1737. 160. Yacobi A, Levy G. Effect of naproxen on protein
144. Swan SK, Rudy DW, Lassater KC, et al. Effect of binding of warfarin in human serum. Res Commun
cyclooxygenase-2 inhibition on renal function in Chem Pathol Pharmacol. 1976;15:369.
elderly persons receiving a low-salt diet. A random- 161. Chaplin MD, Chu NI, Rice BG, Hama KM. Effect of
ized, controlled trial. Ann Intern Med. 2000;133:1. repeated dosing with aspirin on plasma levels of
145. Whelton A, Schulman G, Wallemark C, et al. Effects naproxen (d-2-(6-methoxy-2-naphthyl) propionic
of celecoxib and naproxen on renal function in the acid) in rats. Proc West Pharmacol Soc. 1975;18:62.
elderly. Arch Intern Med. 2000;160:1465. 162. Segre E, Chaplin M, Forchielli E, et al. Naproxen
146. Harris RC, Wang JL, Cheng HF. Prostaglandins in mac- aspirin interactions in man. Clin Pharmacol Ther.
ula densa function. Kidney Int Suppl. 1998;67:S49. 1974;15:374.
147. Harding P, Sigmon DH, Alfie ME, et al. 163. Stitt FW, Vaughan JH. Naproxen-aspirin drug inter-
Cyclooxygenase-2 mediates increased renal renin action studied with response surface analysis. Clin
content induced by low sodium diet. Hypertension. Exp Pharmacol Physiol. 1977;4:216.
1997;29:297. 164. Grennan DM, Ferry DG, Ashworth ME, et al. The
148. Venturi CM, Isakson P, Needleman P. Nonsteroidal aspirin-ibuprofen interaction in rheumatoid arthritis.
anti-inflammatory drug-induced renal failure: a brief Br J Clin Pharmacol. 1979;8:497.
overview of the role of cyclooxygenase isoforms. 165. Rubin A, Rodda BE, Warwick P, et al. Interactions of
Curr Opin Nephrol Hypertens. 1998;7:79. aspirin with non-steroidal anti-inflammatory drugs
149. Traynor TR, Smart A, Briggs JP, Schnermann J. in man. Arthritis Rheum. 1973;16:635.
Inhibition of macula densa-stimulated renin secre- 166. Catella-Lawson F, Reilly MP, Kapoor SC, et al.
tion by pharmacological blockade of cyclooxyge- Cyclooxygenase inhibitors and the anti-platelet
nase-2. Am J Physiol. 1999;277:F706. effects of aspirin. N Engl J Med. 2001;345:
150. Harding P, Curretero OA, Beierwaltes WH. Chronic 1809–17.
cyclooxygenase-2 inhibition blunts low sodium- 167. Hayes AH. Therapeutic implications of drug interac-
stimulated renin without changing renal haemody- tions and acetaminophen and aspirin. Arch Intern
namics. J Hypertens. 2000;18:1107. Med. 1981;141:301.
151. Cheng HF, Wang JL, Zhang MZ, et al. Angiotensin 168. Klotz U. Interaction of analgesics with other drugs.
II attenuates renal cortical cyclooxygenase-2 expres- Am J Med. 1983;75:133.
sion. J Clin Invest. 1999;103:953. 169. Sinclair H, Gibson T. Interaction between probenecid
152. Karim A, Tolbert DS, Hunt TL, et al. Celecoxib, a and indomethacin. Br J Rheumatol. 1986;25:316.
specific COX-2 inhibitor, has no significant effect on 170. Helleberg L. Clinical pharmacokinetics of indo-
methotrexate pharmacokinetics in patients with methacin. Clin Pharmacokinet. 1981;6:245.
rheumatoid arthritis. J Rheumatol. 1999;26:2539. 171. Skeith MD, Simkin P, Healey LA. The renal excre-
153. Greenberg HE, Gottesdiener K, Huntington M, et al. tion of indomethacin and its inhibition by probenecid.
A new cyclooxygenase-2 inhibitor, rofecoxib Clin Pharmacol Ther. 1968;9:89.
(VIOXX) did not alter the antiplatelet effects of low- 172. Dunn MJ. Nonsteroidal anti-inflammatory drugs and
dose aspirin in healthy volunteers. J Clin Pharmacol. renal function. Annu Rev Med. 1984;35:411.
2000;40:1509. 173. Smith DE, Brater DC, Lin ET, Benet LZ. Attenuation
154. Zhang MZ, Harris RC, McKanna JA. Regulation of of furosemide’s diuretic effect by indomethacin: phar-
cyclooxygenase-2 (COX-2) in rat renal cortex by macokinetic evaluation. J Pharmacokinet Biopharm.
adrenal glucocorticoids and mineralocorticoids. Proc 1978;7:265.
Natl Acad Sci USA. 1999;96:15280. 174. Miller KP, Lazar EJ, Fotino S. Severe hyperkalemia
155. Brazier J, Tamisier JN, Ambert D, Bannier A. during piroxicam therapy. Arch Intern Med. 1984;
Bioavailability of ketoprofen in man with and with- 144:2414.
360 9 Management of Hyperuricemia and Gout

175. Finding JW, Beckstrom D, Rawthorne L, et al. 193. Favre L, Glasson P, Vallotton MB. Reversible acute
Indomethacin-induced hyperkalemia in three patients renal failure from combined triamterene and indo-
with gouty arthritis. JAMA. 1980;244:1127. methacin. A study of healthy subjects. Ann Intern
176. Galler M, Shapiro R, Schlondorff D. Reversible Med. 1982;96:317.
acute renal insufficiency and hyperkalemia follow- 194. Garella S, Matarese RA. Renal effects of prostaglan-
ing indomethacin therapy. JAMA. 1981;246:154. din and clinical adverse effects of nonsteroidal anti-
177. Tan SY, Shapiro R, Franco R, et al. Indomethacin- inflammatory agents. Medicine. 1984;63:165.
induced prostaglandin inhibition and hyperkalemia. 195. Berheim JL, Rorzets Z. Indomethacin-induced renal
Ann Intern Med. 1979;90:783. failure. Ann Intern Med. 1979;91:792.
178. Niki G. Nonsteroidal analgesic and antiinflammatory 196. Galler M, Folkert VW, Schlondorf D. Reversible
drugs. Br Med J. 1983;287:39. acute renal insufficiency and hyperkalemia follow-
179. Mason RW, McQueen EG. Protein binding of indo- ing indomethacin therapy. JAMA. 1981;246:154.
methacin. Pharmacology. 1974;12:12. 197. Whelton A, Stout RL, Spilman PS, et al. Renal
180. Rae SA, Williams IA, English J, Baylis EM. effects of ibuprofen, piroxicam, and sulindac in
Alteration of plasma prednisone levels by indo- patients with asymptomatic renal failure. A prospec-
methacin and naproxen. J Clin Pharmacol. 1982; tive, randomized crossover comparison. Ann Intern
14:459. Med. 1990;112:568.
181. Herschberg SN, Sierles FS. Indomethacin-induced 198. Gurwitz JH, Avorn J, Ross-Deghan D, et al.
lithium toxicity. Am Fam Physician. 1983;28:155. Nonsteroidal anti-inflammatory drug-associated
182. van der Donk W, Tsai A, Kulmacz R. The cyclooxy- azotemia in the very old. JAMA. 1990;264:471.
genase reaction mechanism. Biochemistry. 199. Simon LS, Basch CM, Young DY, et al. Effects of
2002;41:15451. naproxen on renal function in older patients with
183. Salvetti A, Arzilli F, Pedriaelli R, et al. Interaction mild to moderate renal dysfunction. Br J Rheumatol.
between oxprenolol and indomethacin on blood 1992;31:163.
pressure in essential hypertensive patients. Eur J 200. Bunning RD, Barth WF. Sulindac: a potentially
Clin Pharmacol. 1982;22:197. renal-sparing nonsteroidal anti-inflammatory drug.
184. Durao V, Prata MM, Goncalves LMP. Modification JAMA. 1982;248:2864.
of antihypertensive effect of b-adrenergic-blocking 201. Brater DC, Anderson S, Baird B, Campbell WB.
agents by inhibition of endogenous prostaglandin Effects of ibuprofen, naproxen, and sulindac on
synthesis. Lancet. 1978;2:1005. prostaglandins in men. Kidney Int. 1985;27:66.
185. Kwan KC, Breault GO, Davis RL, et al. Effects of 202. Miller MJS, Bednar MM, McGiff JC. Renal metabo-
concomitant aspirin administration on the pharma- lism of sulindac: functional implications. J Pharmacol
cokinetics of indomethacin in man. J Pharmacokinet Exp Ther. 1984;231:449.
Biopharm. 1978;6:451. 203. Eriksson L-O, Sturfelt G, Thysell H, et al. Effects of
186. Furst DE, Block KA, Cassel S, et al. A controlled sulindac on prostaglandin excretion in patients with
study of concurrent therapy with a non-acetylated impaired renal function and rheumatoid arthritis.
salicylate and naproxen in rheumatoid arthritis. Am J Med. 1990;89:313.
Arthritis Rheum. 1987;30:146. 204. Daskalopoulos G, Kronberg I, Katkov W, et al.
187. Brooks PM, Walker JJ, Bell MA. Indomethacin aspi- Sulindac and indomethacin suppresses the diuretic
rin interaction. A clinical appraisal. Br Med J. 1974; action of furosemide in patients with cirrhosis and
11:69. ascites: evidence that sulindac affects renal prosta-
188. Williams PL, Davies RO, Berman RS. glandins. Am J Kidney Dis. 1985;6:217.
Hydrochlorothiazide pharmacokinetics and pharma- 205. Klassen DK, Stout RL, Spilman PS, et al. Sulindac
cologic effects: the influence of indomethacin. J Clin kinetics and effects on renal function and prostaglan-
Pharmacol. 1982;22:32. din excretion in renal insufficiency. J Clin Pharmacol.
189. Henrich WL. Nephrotoxicity of the non-steroidal 1989;29:1037.
anti-inflammatory agents. In: Schrier R, Gottshalk C, 206. Dixey JJ, Noormohamed FH, Lant AF. The effects of
editors. Diseases of the kidney. 5th ed. Boston: Little naproxen and sulindac on renal function and their
Brown; 1992. p. 1201–18. interaction with hydrochlorothiazide and piretanide
190. McCarthy JT, Torres VE, Romero JC, et al. Acute in man. Br J Clin Pharmacol. 1987;23:55.
intrinsic renal failure induced by indomethacin: roles 207. Swanson CP, Griffiths P. Acute and chronic effects
of prostaglandin synthetase inhibition. Mayo Clin of sulindac on renal function in chronic renal dis-
Proc. 1982;52:289. ease. Clin Pharmacol Ther. 1985;37:298.
191. Kimberly RP, Brandstetter RD. Exacerbation of phe- 208. Quintero E, Gines P, Arroyo V. Sulindac reduces the
nylbutazone-related renal failure by indomethacin. urinary excretion of prostaglandins and impairs renal
Ann Intern Med. 1978;138:1711. function in cirrhosis and ascites. Nephron.
192. Brezin JH, Katz SM, Schwartz AB, et al. Reversible 1986;42:298.
renal failure and nephrotic syndrome associated with 209. Cibattoni G, Cinotti GA, Pierucci A. Effects of
nonsteroidal anti-inflammatory drugs. N Engl J Med. sulindac and ibuprofen in patients with chronic
1979;301:1271. glomerular disease: evidence for the dependence of
References 361

renal function on prostacyclin. N Engl J Med. 228. Blackshear JL, Napier JS, Davidman M, Stillman
1984;310:279. MT. Renal complications of nonsteroidal anti –
210. Laffi G, Daskalopoulos G, Kronberg I, et al. Effects inflammatory drugs: identification and monitoring of
of sulindac and ibuprofen in patients with cirrhosis those at risk. Semin Arthritis Rheum. 1985;14:163.
and ascites. An explanation for the renal-sparing 229. Chen CY, Pang VF, Chen CS. Pathological and bio-
effect of sulindac. Gastroenterology. 1986;90:182. chemical modifications of renal function in ibupro-
211. Zipser RD, Hoefs JC, Speckart PE, et al. fen-induced interstitial nephritis. Ren Fail. 1996;
Prostaglandins: modulators of renal function and 18:31.
pressor resistance in chronic liver disease. J Clin 230. Goetzl E. Selective feedback inhibition of the
Endocrinol Metab. 1979;48:895. 5-lipoxygenation of the arachidonic acid in human
212. Schlondorff D. Renal complications of nonsteroidal T-lymphocytes. Biochem Biophys Res Commun.
anti-inflammatory drugs. Kidney Int. 1993;44:643. 1981;101:344.
213. Corelli RL, Gericke KR. Renal insufficiency associ- 231. Torres VE. Present and future of the nonsteroidal
ated with intramuscular administration of ketorolac anti-inflammatory drugs in nephrology. Mayo Clin
tromethamine. Ann Pharmacother. 1993;27:1055. Proc. 1982;57:389.
214. Pearce CJ, Gonzalez FM, Wallin JD. Renal failure 232. Siegl M, McConnel R, Porter N, et al. Arachidonate
and hyperkalemia associated with ketorolac metabolism via lipoxygenase and 12-L-hydroper-
tromethamine. Arch Intern Med. 1993;153:1000. oxy-5,8,10,14-eicosatetraenoic acid peroxidase sen-
215. Feldman HI, Kinman JL, Berlin JA, et al. Parental sitive to anti-inflammatory drugs. Proc Natl Acad
ketorolac: the risk for acute renal failure. Ann Intern Sci USA. 1980;77:308.
Med. 1997;126:193. 233. Goetzl E. Mediators of immediate hypersensitivity
216. Lourie SH, Denman SJ, Schroeder ET. Association derived from arachidonic acid. N Engl J Med. 1980;
of renal papillary necrosis and ankylosing spondyli- 303:822.
tis. Arthritis Rheum. 1977;20:917. 234. Bhattacherjee P, Hammond B, Salmon JA, et al.
217. Atta MG, Whelton A. Acute renal papillary necrosis Chemotactic response to some arachidonic acid
induced by ibuprofen. Am J Ther. 1997;4:55. lipoxygenase products in the rabbit eye. Eur J
218. Morales A, Steyn J. Papillary necrosis following phe- Pharmacol. 1981;73:21.
nylbutazone ingestion. Arch Surg. 1971;103:420. 235. Schooley RT, Wagley PF, Lietman PS. Edema asso-
219. Abraham PA, Keane WF. Glomerular and interstitial ciated with ibuprofen therapy. JAMA. 1977;237:
disease induced by nonsteroidal anti-inflammatory 1716.
drugs. Am J Nephrol. 1984;4:1. 236. Nies AS, Gel J, Fadul S, et al. Indomethacin-
220. Gokal R, Matthews DR. Renal papillary necrosis furosemide interaction: the importance of renal
after aspirin and alclofenac. Br Med J. 1977;2:1517. blood flow. J Pharmacol Exp Ther. 1983;226:27.
221. Segasothy M, Samad SA, Zulfiquar A, Bennett WM. 237. Williamson HE, Bourland UA, Marchand GR.
Renal dysfunction and renal papillary necrosis with Inhibition of furosemide-induced increase in renal
long-term use of NSAIDS as a sole or predominant blood flow by indomethacin. Proc Soc Exp Biol
analgesic: a new form of analgesic nephropathy. Am Med. 1975;148:164.
J Soc Nephrol. 1993;4:259. 238. Berg KJ. Acute effects of acetylsalicylic acid in
222. Segasothy M, Chin GL, Sia KK, et al. Chronic neph- patients with chronic renal insufficiency. Eur J Clin
rotoxicity of anti-inflammatory drugs used in the Pharmacol. 1977;11:111.
treatment of arthritis. Br J Rheumatol. 1995;34:162. 239. Patak RV, Moorkejeree BK, Bentzel CJ, et al.
223. Henrich WL, Agodoa LE, Barrett B, et al. Antagonism of the effects of furosemide by indo-
Analgesics and the kidney: summary and recom- methacin in normal and hypertensive men.
mendations to the scientific advisory board of the Prostaglandins. 1975;10:649.
national kidney foundation from an ad hoc commit- 240. Tiggeler RG, Koene RA, Wijdeveld PG. Inhibition
tee of the national kidney foundation. Am J Kidney of furosemide-induced natriuresis by indomethacin
Dis. 1996;27:162. in patients with nephrotic syndrome. Clin Sci Mol
224. Finklestein A, Fraley D, Stachura I, et al. Fenoprofen Med. 1977;2:149.
nephropathy. Lipid nephrosis and interstitial nephri- 241. Mirouze D, Zisper RD, Reynolds TB. Effect of
tis. A possible T lymphocyte disorder. Am J Med. inhibitors of prostaglandin synthesis on induced cir-
1982;72:81. rhosis. Hepatology. 1983;3:50.
225. Katz S, Capaldo R, Everts E, et al. Association with 242. Houston MC. Nonsteroidal anti-inflammatory drugs
reversible renal failure and acute interstitial nephri- and antihypertensives. Am J Med. 1991;90:42S.
tis. JAMA. 1981;246:243. 243. Luscher TF. Imbalance of endothelium-derived
226. Morgenstern SJ, Burns FJ, Fraley DS, et al. relaxing factor and contracting factors. Am J
Ibuprofen-associated lipid nephrosis without inter- Hypertens. 1990;3:317.
stitial nephritis. Am J Kidney Dis. 1989;16:50. 244. Diederich D, Yang ZH, Buhler FR, et al. Impaired
227. Schwartzman M, Dagati V. Spontaneous relapse of endothelium-dependent relaxations in hypertensive
naproxen related nephrotic syndrome. Am J Med. resistance arteries involve cyclooxygenase pathway.
1987;82:329. Am J Physiol. 1990;258:H445.
362 9 Management of Hyperuricemia and Gout

245. Minuz P, Barrow SE, Cockcroft JR, et al. Prostacyclin inhibitors on prostanoid release from various rat
and thromboxane synthesis in mild essential hyper- organs. J Pharmacol Exp Ther. 2000;292:1161.
tension. Hypertension. 1990;15:469. 264. Hawkey CJ. Nonsteroidal anti-inflammatory drug
246. Vane JR, Anggard EE, Botting M. Regulatory func- gastropathy. Gastroenterology. 2000;119:521.
tions of the vascular endothelium. N Engl J Med. 265. Verburg KM, Maziasz TJ, Weiner E, et al. COX-2-
1990;323:27. specific inhibitors: definition of a new therapeutic
247. Panzer JA, Quyyumi AA, Brush JE, et al. Abnormal concept. Am J Ther. 2001;8:41.
endothelium-dependent vascular relaxation in 266. Takeuchi K, Kagawa S, Mimaki H, et al. COX and
patients with essential hypertension. N Engl J Med. NOS isoforms involved in acid-induced duodenal
1990;323:22. bicarbonate secretion in rats. Dig Dis Sci. 2002;
248. Linder L, Wolfgang K, Buhler FR, et al. Indirect evi- 47:2116.
dence for release of endothelium-derived relaxing 267. Tani S, Suzuki T, Kano S, et al. Mechanisms of gas-
factor in human forearm circulation in vivo. tric mucus secretion from cultured rat gastric epithe-
Circulation. 1990;81:1762. lial cells induced by carbachol, cholecystokinin
249. Kato T, Iwama Y, Okumura K, et al. Prostaglandin octapeptide, secretion, and prostaglandin E2. Biol
H2 may be the endothelium-derived contracting fac- Pharm Bull. 2002;25:14.
tor released by acetylcholine in the aorta of the rat. 268. Maricic N, Ehrlich K, Gretzer B, et al. Selective
Hypertension. 1990;15:475. cyclooxygenase-2 inhibition aggravates ischemia-
250. Walshe JJ, Venuto RC. Acute oliguric renal failure reperfusion injury in the rat stomach. Br J Pharmacol.
induced by indomethacin: possible mechanism. Ann 1999;128:1659.
Intern Med. 1979;91:47. 269. Vane JR, Mitchell JA, Appleton T, et al. Inducible
251. Tan SY, Franco R, Stockard H, Luilrow RJ. isoforms of cyclooxygenase and nitric oxide syn-
Indomethacin-induced prostaglandin inhibition with thase in inflammation. Proc Natl Acad Sci USA.
hyperkalemia. A reversible cause of hyporeninemic 1994;91:2046.
hypoaldosteronism. Ann Intern Med. 1979;90:783. 270. Ferraz JG, Sharkey KA, Reuter BK, et al. Induction
252. Nicholls MG, Espiner EA. Indomethacin-induced of cyclooxygenase-1 and −2 in rat stomach during
azotemia and hyperkalaemia: a case study. N Z Med endotoxemia: role in resistance to damage.
J. 1981;94:377. Gastroenterology. 1997;113:195.
253. Kutyrina JM, Andosova SO, Tareyeva IE. 271. Siegle I, Klein T, Backman JT, et al. Expression of
Indomethacin-induced hyporeninaemic hypoaldos- cyclooxygenase-2 in human synovial tissue. Arthritis
teronism. Lancet. 1979;1:785. Rheum. 1998;41:122.
254. Goldszer RC, Coodley EL, Rosner MJ, et al. 272. Michaluart P, Masferrer JL, Carothers AM, et al.
Hyperkalemia associated with indomethacin. Arch Inhibitory effects of caffeic acid on the activity and
Intern Med. 1981;141:802. expression of cyclooxygenase-2 in human oral epi-
255. Clive DM, Stoff JS. Renal syndromes associated thelial cells and in a rat model of inflammation.
with nonsteroidal anti-inflammatory drugs. N Engl J Cancer Res. 1999;59:2347.
Med. 1984;310:563. 273. Mitchell JA, Belvisi MG, Akarasereenont P, et al.
256. Navis GJ, de Jong PE, de Zeeuw D. Serum uric acid, Induction of cyclooxygenase-2 by cytokines in
ace inhibitors and natriuresis. Lancet. 1985;2:156. human pulmonary epithelial cells: regulation by dex-
257. Dunn MJ, Hood VL. Prostaglandins and the kidney. amethasone. Br J Pharmacol. 1994;113:1008.
Am J Physiol. 1977;233:F169. 274. Smith WL, Bell TG. Immunohistochemical localiza-
258. Dunn MJ, Zambraski E. Renal effects of drugs that tion of the prostaglandin-forming cyclooxygenase in
inhibit prostaglandin synthesis. Kidney Int. renal cortex. Am J Physiol. 1978;235:F451.
1980;18:609. 275. Komhoff M, Crone HJ, Klein T, et al. Localization of
259. Whelton A. Nephrotoxicity of nonsteroidal anti- cyclooxygenase-1 and −2 in adult and fetal human
inflammatory drugs: physiologic foundations and kidney: implication for renal function. Am J Physiol.
clinical implications. Am J Med. 1999;106:135. 1977;272:F460.
260. Nakhai-Pour HR, Perrine B, Sheehy OS, Berard A. 276. Komhoff M, Jeck NO, Seyberth HW, et al.
Use of nonaspirin nonsteroidal anti-inflammatory Cyclooxygenase-2 expression is associated with the
drugs during pregnancy and the risk of spontaneous renal macula densa of patients with Bartter-like syn-
abortion. CMAJ. 2011;183:1713–20. drome. Kidney Int. 2000;58:2420.
261. Xie W, Chipman JG, Robertson DL, et al. Expression 277. Harris RC, McKanna JA, Akai Y, et al.
of mitogen-responsive gene encoding prostaglandin Cyclooxygenase-2 is associated with macula densa
synthase is regulated by mRNA splicing. Proc Natl of rat kidney and increases with salt restriction. J
Acad Sci USA. 1991;88:2692. Clin Invest. 1994;94:2504.
262. Mitchell JA, Warner TD. Cyclooxygenase-2: phar- 278. Khan KN, Venturini CM, Bunch RT, et al.
macology, physiology, biochemistry and relevance Expression of cyclooxygenase-2 in the macula
to NSAID therapy. Br J Pharmacol. 1999;128:1121. densa of human kidney in hypertension, congestive
263. Tegeder I, Neuper TW, Guhring H, Geisslinger G. heart failure, and diabetic nephropathy. Ren Fail.
Effects of selective and unselective cyclooxygenase 2001;23:321.
References 363

279. Nantel F, Meadows E, Denis D, et al. lmmunolocal- with individual nonsteroidal anti-inflammatory drugs:
ization of cyclooxygenase-2 in the macula densa of results of a collaborative meta analysis. Br Med J.
human elderly. FEBS Lett. 1999;457:475. 1996;312:1563.
280. Kahn KN, Venturini CM, Bunch RT, et al. lnterspe- 295. Minocha A, Greenbaum OS. Pill-esophagitis caused
cies differences in renal localization of cyclooxyge- by nonsteroidal anti-inflammatory drugs. Am J
nase isoforms: implications in nonsteroidal Gastroenterol. 1991;86:1086.
anti-inflammatory drug-related nephrotoxicity. 296. Fries JF, Williams CA, Block DA, Michel BA.
Toxicol Pathol. 1998;26:612. Nonsteroidal anti-inflammatory drug associated gas-
281. Guan Y, Chang M, Cho W, et al. Cloning, expression tropathy: incidence and risk factor models. Am J
and regulation of rabbit cyclooxygenase-2 in renal Med. 1991;91:213.
medullary interstitial cells. Am J Physiol. 297. Shallcross TM, Healey RV. Effect of nonsteroidal
1997;273:F18. anti-inflammatory drugs and esophageal injury. Br
282. Adegboyega PA, Olalade O. Immunohistochemical Med J. 1990;300:368.
expression of cyclooxygenase-2 in normal kidneys. 298. Semble F, Wu WC, Castell DO. Nonsteroidal anti-
Appl Immunohistochem Mol Morphol. 2004;12:71. inflammatory drugs and esophageal injury. Semin
283. Fergusan S, Hebert RL, Laneuville O. NS-398 Arthritis Rheum. 1989;19:99.
upregulates constitutive cyclooxygenase-2 expres- 299. Graham DY, Chan FKL. NSAIDS, risks, and gastro-
sion in the M1 cortical collecting duct cell line. J Am protective strategies: current status and future.
Soc Nephrol. 1999;10:2261. Gastroenterology. 2008;134:1240.
284. Yang T, Schnermann JB, Briggs JP. Regulation of 300. Moore RA, Derry S, McQuay HJ. Cyclooxygenase-2
cyclooxygenase-2 expression in renal medulla by selective inhibitors and nonsteroidal anti
tonicity in vivo and in vitro. Am J Physiol. 1999; inflammatory drugs: balancing gastrointestinal and
277:F1. cardiovascular risk. BMC Musculoskelet Disord.
285. Furst DE. Are there differences among nonsteroidal 2007;8:73.
anti-inflammatory drugs? Comparing acetylated sal- 301. Sciulli MG, Capone ML, Tacconelli S, Patrignani P.
icylates, nonacetylated salicylates, and nonacety- The future of traditional nonsteroidal anti
lated nonsteroidal anti-inflammatory drugs. Arthritis inflammatory drugs and cyclooxygenase-2 inhibi-
Rheum. 1994;37:1. tors in the treatment of inflammation and pain.
286. Suleyman H, Demircan B, Karagoz Y. Anti- Pharmacol Rep. 2005;57:66.
inflammatory and side effects of cyclooxygenase 302. Simon LS. The COX2 selective inhibitors. What the
inhibitors. Pharmacol Rep. 2007;54:247. newspapers have not told you. Bull NYU Hosp Jt
287. Cryer B, Feldman M. Cyclooxygenase-1 and Dis. 2007;65:229.
cyclooxygenase-2 selectivity of widely used non- 303. Mielants H, Goemaere S, DeVos M, et al. Intestinal
steroidal anti-inflammatory drugs. Am J Med. mucosal permeability in inflammatory rheumatic
1998;104:413. diseases. I. Role of anti-inflammatory drugs. J
288. Warner TD, Giuliano F, Vojnovic L, et al. Nonsteroid Rheumatol. 1991;18:389.
drug selectivities for cyclo-oxygenase-1 rather than 304. Mielants H, DeVos M, Goemaere S, et al. Intestinal
cyclo-oxygenase-2 are associated with human gas- mucosal permeability in inflammatory rheumatic dis-
trointestinal toxicity: a full in vitro analysis. Proc eases. II. Role of disease. J Rheumatol. 1991;18:394.
Natl Acad Sci USA. 1999;96:7563. 305. Allison MC, Howatson AG, Torance CJ.
289. Fries JF, Muller SR, Spitz PW, et al. Toward an epi- Gastrointestinal damage associated with the use of
demiology of gastropathy associated with nonsteroi- nonsteroidal anti-inflammatory drugs. N Engl J Med.
dal anti-inflammatory drug use. Gastroenterology. 1992;327:749.
1989;96:647. 306. Johnson AG. NSAIDS and increased blood pressure.
290. Griffin MR, Piper JM, Daugherty JR, et al. What is the clinical experience? Drug Saf.
Nonsteroidal anti-inflammatory drug use and 1977;17:277.
increased risk for peptic ulcer disease in elderly per- 307. Gross JM, Dwyer JE, Know FG. Natriuretic response
sons. Ann Intern Med. 1991;114:257. to increased pressure is preserved with COX-2
291. Gabriel SE, Jaaklimainen L, Bombadier C. Risk of inhibitors. Hypertension. 1999;34:1163.
serious gastrointestinal complications related to the 308. Catella-Lawson F, McAdam B, Morrison BW, et al.
use of nonsteroidal anti-inflammatory drugs: a meta Effects of specific inhibition of cyclooxygenase-2 on
analysis. Ann Intern Med. 1991;115:787. sodium balance, hemodynamics, and vasoactive
292. Morris AJ, Madhok R, Sturrock RD, et al. eicosanoids. J Pharmacol Exp Ther. 1999;289:735.
Enteroscopic diagnosis of small bowel ulceration in 309. Zewde T, Mattson DL. Inhibition of cyclooxyge-
patients receiving nonsteroidal anti-inflammatory nase-2 in the rat renal medulla leads to sodium-
drugs. Lancet. 1991;337:520. sensitive hypertension. Hypertension. 2004;44:424.
293. Eng J, Sabanathan S. Drug-induced esophagitis. Am 310. Rossat RJ, Maillard M, Nussberger J, et al. Renal
J Gasteroenterol. 1991;86:1127. effects of selective cyclooxygenase-2 inhibition in
294. Henry D, Lim LL, Garcia Rodriguez LA, et al. normotensive salt-depleted subjects. Clin Pharmacol
Variability in the risk of gastrointestinal complications Ther. 1999;66:76.
364 9 Management of Hyperuricemia and Gout

311. Stokes JB, Kokko JP. Inhibition of sodium transport expression in fetal lambs. Am J Physiol Regul Integr
by prostaglandin E2 across the isolated, perfused Comp Physiol. 2003;284:K1012.
rabbit collecting tubule. J Clin Invest. 327. Harris RC, Zhang MZ, Cheng HF. Cyclooxygenase-2
1977;59:1099. and the renal renin-angiotensin system. Acta Physiol
312. Hebert RL, Jacobson HR, Breyer MD. Prostaglandin Scand. 2004;181:543.
E2 inhibits sodium transport in rabbit cortical col- 328. Cheng HF, Wang SW, Zhang MZ, et al. Prostaglandins
lecting duct by increasing intracellular calcium. J that increase renin production in response to ACE inhi-
Clin Invest. 1991;87:1992. bition are not derived from cyclooxygenase-1. Am J
313. Lopez-Parra M, Claria J, Planaguma A, et al. Physiol Regul Integr Comp Physiol. 2002;283:R638.
Cyclooxygenase-1 derived prostaglandins are involved 329. Wang JL, Cheng HF, Harris RC. Cyclooxygenase-2
in the maintenance of renal function in rats with cir- inhibition decreases renin content and lowers blood
rhosis and ascites. Br J Pharmacol. 2002;135:82. pressure in a model of renovascular hypertension.
314. Qi Z, Hao CM, Langenbach RL, et al. Opposite Hypertension. 1999;34:96.
effects of cyclooxygenase-1 and −2 activity on the 330. Castrop H, Schweda F, Schumacher K, et al. Role of
pressure response to angiotensin II. J Clin Invest. renocortical cyclooxygenase-2 for renal vascular
2001;110:61. resistance and macular densa control of renin secre-
315. Athirakul K, Kim HS, Audoly LP, et al. Deficiency tion. J Am Soc Nephrol. 2001;12:867.
of COX-1 causes natriuresis and enhanced sensitiv- 331. Traynor TR, Smart A, Briggs JP, Schnermann J.
ity to ACE inhibition. Kidney Int. 2001;60:2324. Inhibition of macula densa-stimulated renin secre-
316. Johnson AG, Nguyen TV, Day RO. Do nonsteroidal tion by pharmacological blockade of cyclooxyge-
anti-inflammatory drugs affect blood pressure? A nase-2. Am J Physiol. 1999;277:F706.
meta analysis. Ann Intern Med. 1994;121:289. 332. Cheng HF, Wang JL, Zhang MZ, et al. Genetic dele-
317. de Leeuw PW. Nonsteroidal anti-inflammatory drugs tion of COX-2 prevents increased renin expression
and hypertension. The risks in perspective. Drugs. in response to ACE inhibition. Am J Physiol Renal
1996;51:179. Physiol. 2001;280:F449.
318. Whelton A, Fort JG, Puma JA, et al. Cyclooxygenase- 333. Strichtenoth DO, Wagner B, Frolich JC. Effect of
2-specific inhibitors and cardiorenal function: a ran- selective inhibition of the inducible cyclooxygenase
domized, control trial of celecoxib and rofecoxib in on renin release in healthy volunteers. J Investig
older hypertensive osteoarthritis patients. Am J Ther. Med. 1998;46:290.
2001;8:85. 334. Reinalter SC, Jeck N, Brochhausen C, et al. Role of
319. Whelton A, White WB, Bello AE, et al. Effects of cyclooxygenase-2 in hyperprostaglandin E syn-
celecoxib and rofecoxib on blood pressure and drome/antenatal Bartter syndrome. Kidney Int.
edema in patients > or = 65 years of age with sys- 2002;62:253.
temic hypertension and osteoarthritis. Am J Cardiol. 335. Kleta K, Basoglu C, Kuwertz-Broking E. New treat-
2002;90:959. ment options for Bartter’s syndrome. N Engl J Med.
320. Sowers JR, White WB, Pitt B, et al. The effects of 2000;343:661.
cyclooxygenase-2 inhibitors and nonsteroidal anti- 336. Dunn MJ. Prostaglandin 12 and the kidney. Arch
inflammatory therapy on 24-hour blood pressure in Mal Coeur Vaiss. 1989;82:27.
patients with hypertension, osteoarthritis, and type 2 337. Perazella MA, Tray K. Selective cyclooxygenase-2
diabetes mellitus. Arch Intern Med. 2005;165:161. inhibitors. A pattern of nephrotoxicity similar to tra-
321. Cheng HF, Wang JL, Zhang MZ, et al. Role of p38 in ditional nonsteroidal anti-inflammatory drugs. Am J
the regulation of renal cortical cyclooxygenase-2 Med. 2001;111:64.
expression by extracellular chloride. J Clin Invest. 338. Ahmad SR, Kortepeter C, Brinker A, et al. Renal
2000;106:681. failure associated with the use of celecoxib and rofe-
322. Cheng HF, Harris RC. Cyclooxygenase-2 expression coxib. Drug Saf. 2002;25:537.
in cultured cortical thick ascending limb of Henle 339. Papaioannides D, Bouropoulas C, Sinapides D, et al.
increases in response to decreased extracellular ionic Acute renal dysfunction associated with selective
content by both transcriptional and post-transcrip- COX-2 enzyme inhibitor therapy. Int Urol Nephrol.
tional mechanisms. Role of p38-mediated pathways. 2001;33:609.
J Biol Chem. 2002;277:45638. 340. Woywodt A, Schwarz A, Mengel M, et al.
323. Yang T, Park JM, Arend L, et al. Low chloride stim- Nephrotoxicity of selective COX-2 inhibitors. J
ulation of mouse macula densa cell line. J Biol Rheumatol. 2003;28:2133.
Chem. 2000;275:37922. 341. Phelan KM, Mosholder AD, Lu S. Lithium interac-
324. Kammer MC, Nusing RM, Schweda F, et al. Low tion with the cyclooxygenase 2 inhibitors rofecoxib
sodium and furosemide-induced stimulation of the and celecoxib and other nonsteroidal anti-inflammatory
renin system in man is mediated by cyclooxygenase- drugs. J Clin Psychiatry. 2003;64:1328.
2. Clin Pharmacol Ther. 2001;70:468. 342. Harris RC, Breyer MD. Update on cyclooxygenase-2
325. Harris RC. The macula densa: recent developments. inhibitors. Clin J Am Soc Nephrol. 2006;1:236.
J Hypertens. 1996;14:815. 343. Nasrallah R, Clark J, Hebert RL. Prostaglandins in
326. Mertz HL, Liu J, Valego NK, et al. Inhibition of the kidney: developments since Y2K. Clin Sci.
cyclooxygenase-2: effects on renin secretion and 2007;113:297.
References 365

344. Cheng HF, Harris RC. Cyclooxygenases, the kidney 360. Bek M, Nusing RM, Kowark P, et al. Characterization
and hypertension. Hypertension. 2004;43:525. of prostanoids receptors in podocytes. J Am Soc
345. Knights KM, Tsoutsikos P, Miners JO. Novel mech- Nephrol. 1999;10:2084.
anisms of nonsteroidal anti-inflammatory drug- 361. Nasrallah R, Zimplemann J, Singh S, Hebert RL.
induced renal toxicity. Expert Opin Drug Metab Molecular and biochemical characterization of pros-
Toxicol. 2005;1:399. tacyclin (IP) receptors in the rat kidney. Am J Physiol
346. Cheng HF, Harris RC. Renal effects of non-steroidal Renal Physiol. 2001;280:F266.
anti-inflammatory drugs and selective cyclooxyge- 362. Oida H, Nambat T, Sugimoto Y, et al. In situ hybrid-
nase-2 inhibitors. Curr Pharm Des. 2005;11:1795. ization studies of prostacyclin receptor mRNA
347. Harris RC. COX-2 and the kidney. J Cardiovasc expression in various mouse organs. Br J Pharmacol.
Pharmacol. 2006;47 Suppl 1:837. 1995;116:2828.
348. Gambaro G, Perazella MA. Adverse renal effects of 363. Komhoff M, Lesener B, Nakao K, et al. Localization
anti-inflammatory agents: evaluation of selective of the prostacyclin receptor in human kidney. Kidney
and nonselective cyclooxygenase inhibitors. J Intern Int. 1998;54:1899.
Med. 2003;253:643. 364. Day R, Morrison B, Luza A, et al. A randomized
349. Nasrallah R, Hebert RL. Prostacyclin signaling in trial of the efficacy and tolerability of the COX-2
the kidney: implications for health and disease. Am inhibitor rofecoxib vs. ibuprofen in patients with
J Physiol Renal Physiol. 2005;289:235. osteoarthritis. Rofecoxib/lbuprofen Comparator
350. Lomnicka M, Karouni K, Sue M, et al. Effects of Study Group. Arch Intern Med. 2000;160:1781.
nonsteroidal anti-inflammatory drugs on prostacy- 365. Morrison BW, Christensen S, Yuan W, et al.
clin and thromboxane in the kidney. Pharmacology. Analgesic efficacy of the cyclooxygenase-2-specific
2003;68:147. inhibitor rofecoxib in post-dental surgery pain: a ran-
351. Qi Z, Cai H, Morrow JD, Breyer MD. Differentiation domized, controlled trial. Clin Ther. 1999;21:943.
of cyclooxygenase-1 and −2-derived prostanoids in 366. Cannon GW, Caldwell JR, Holt P, et al. Rofecoxib, a
mouse kidney and aorta. Hypertension. 2006;48:323. specific inhibitor of cyclooxygenase 2, with clinical
352. Warford-Woolgar L, Peng CY, Tahiti J, et al. efficacy comparable with that of diclofenac sodium:
Selectivity of cyclooxygenase isoform activity and results of a one-year, randomized, clinical trial in
prostanoid production in normal and diseased Han: patients with osteoarthritis of the knee and hip.
SPRD-cy kidneys. Am J Physiol Renal Physiol. Rofecoxib Phase Ill Protocol 035 Study Group.
2006;290:F897. Arthritis Rheum. 2000;43:978.
353. Hetu PO, Riendeau D. Cyclo-oxygenase-2 contrib- 367. Riecin A, Brown J, Jove M, et al. Efficacy of single-
utes to constitutive prostanoid production in the rat dose and multidose rofecoxib in the treatment of
kidney and brain. Biochem J. 2005;391:561. post-orthopedic pain. Am J Orthop. 2001;30:40.
354. Neuhofer W, Holzapfel K, Frack ML, et al. Chronic 368. Bombardier C, Laine L, Reicin A, et al. Comparison
COX-2 inhibition reduces medullary HSP70 expres- of upper gastrointestinal toxicity of rofecoxib and
sion and induces papillary apoptosis in dehydrated naproxen in patients with rheumatoid arthritis. VIGOR
rats. Kidney Int. 2004;65:431. Study Group. N Engl J Med. 2000;343:1520.
355. Kang DS, Kwon CH, Park JY, et al. 15-Deoxy-12, 369. Lai KC, Chu KM, Hui WM, et al. Celecoxib com-
14-prostaglandin J2 induces renal epithelial cell pared with lansoprazole and naproxen to prevent
death through NF-KB-dependent and MAPK- gastrointestinal ulcer complications. Am J Med.
independent mechanism. Toxicol Appl Pharmacol. 2005;118:1271.
2006;216:426. 370. Schnitzer TJ, Burmester GR, Mysler E, et al.
356. Chana RS, Lewington AJ, Brunskill NJ. Differential Comparison of lumiracoxib with naproxen and ibu-
effects of peroxisome proliferation activated recep- profen in the Therapeutic Arthritis Research and
tor-y (PPAR-y) ligands in proximal tubular cells: Gastrointestinal Event Trial (TARGET), reduction in
thiazolidinediones are partial PPARy agonists. ulcer complications: randomized controlled trial.
Kidney Int. 2004;65:2081. Lancet. 2004;364:665.
357. Sanchez-Gomez FJ, Cemuda-Morollon E, Stamatakis 371. Chan FK, Hung LC, Suen BY, et al. Celecoxib ver-
K, Perez-Sala D. Protein thiol modification by sus diclofenac and omeprazole in reducing the risk
15-deoxy-12, 14-prostaglandin J2 addition in mesan- of recurrent ulcer bleeding in patients with arthritis.
gial cells: role in the inhibition of pro-inflammatory N Engl J Med. 2002;347:2104.
genes. Mol Pharmacol. 2006;66:1349. 372. Laine L, Curtis SP, Cryer B, et al. Assessment of
358. Stamatakis K, Sanchez-Gomez FJ, Perez-Sala D. upper gastrointestinal safety of etoricoxib and
Identification of novel protein targets for modification diclofenac in patients with osteoarthritis and rheu-
by 15-deoxy-12, 14-prostaglandin J2 in mesangial matoid arthritis in the Multinational Etoricoxib and
cells reveals multiple interactions with the cytoskel- Diclofenac Arthritis Long-term (MEDAL) pro-
eton. J Am Soc Nephrol. 2006;17:89. gramme. A randomized comparison. Lancet.
359. Hebert RL, O’Connor T, Neville C, et al. Prostanoid 2007;369:465.
signalling, localization and expression of IP recep- 373. Juni P, Rutjes AW, Dieppe PA. Are selective COX-2
tors in rat thick ascending limb cells. Am J Physiol. inhibitors superior to traditional nonsteroidal anti-
1998;275:F904. inflammatory drugs? BMJ. 2002;324:1287.
366 9 Management of Hyperuricemia and Gout

374. Fenton C, Keating GM, Wagstaff AJ. Valdecoxib: a 389. Hawkey CJ, Hawkey GM, Everett S, et al. Increased
review of its use in the management of osteoarthritis, risk of myocardial infarction as first manifestation of
rheumatoid arthritis, dysmenorrheal, and acute pain. ischaemic heart disease and nonselective nonsteroi-
Drugs. 2004;64:1231. dal anti-inflammatory drugs. Br J Clin Pharmacol.
375. Sikes DH, Agrawal NM, Zhao WW, et al. Incidence 2006;61:730.
of gastroduodenal ulcers associated with valdecoxib 390. Grosser T. The pharmacology of selective inhibition
compared with that of ibuprofen and diclofenac in of COX-2. Thromb Haemost. 2006;96:393.
patients with osteoarthritis. Eur J Gastroenterol 391. Grosser T, Fries S, Fitzgerald GA. Biological bases
Hepatol. 2002;14:1101. for the cardiovascular consequences of COX-2 inhi-
376. Hunt RH, Harper S, Watson OJ, et al. The gastroin- bition: therapeutic challenges and opportunities. J
testinal safety of the COX-2 selective inhibitor etori- Clin Invest. 2006;116:4.
coxib assessed by both endoscopy and analysis of 392. Howes LG. Selective COX-2 inhibitors, NSAIDS
upper gastrointestinal events. Am J Gastroenterol. and cardiovascular events – is celecoxib the safest
2003;98:1725. choice? Ther Clin Risk Manag. 2007;3:831.
377. Davidge ST. Prostaglandin H synthase and vascular 393. Caldwell B, Aldington S, Weatherall M, et al. Risk of
function. Circ Res. 2001;89:650. cardiovascular events and celecoxib: a systematic
378. Castella-Lawson F, McAdam B, Morrison BW, et al. review and meta-analysis. J R Soc Med. 2006;99:132.
Effects of specific inhibition of cyclooxygenase-2 on 394. McGettigan P, Henry D. Cardiovascular risk and
sodium balance, hemodynamics, and vasoactive inhibition of cyclooxygenase. A systematic review
eicosanoids. J Pharmacol Exp Ther. 1999;289:735. of the observational studies of selective and nonse-
379. Belton O, Byrne D, Kearney D, et al. lective inhibitors of cyclooxygenase 2. JAMA.
Cyclooxygenase-1 and −2-dependent prostacyclin 2006;296:1633.
formation in patients with atherosclerosis. 395. Zhang J, Ding EL, Song Y. Adverse effects of
Circulation. 2000;102:840. cyclooxygenase 2 inhibitors of renal and arrhythmia
380. McAdam SF, Castella-Lawson F, Mardini IA, et al. events. JAMA. 2006;296:1619.
Systemic biosynthesis of prostacyclin by cyclooxy- 396. Helm HK, Broich K. Selective COX-2 inhibitors and
genase (COX)-2: the human pharmacology of a risk of thromboembolic events – regulatory aspects.
selective inhibitor of COX-2. Proc Natl Acad Sci Thromb Haemost. 2006;96:423.
USA. 1999;96:272. 397. Krotz F, Schiele TM, Klauss V, Sohn HY. Selective
381. Belton OA, Duffy A, Toomey S, Fitzgerald OJ. COX-2 inhibitors and risk of myocardial infarction.
Cyclooxygenase isoforms and platelet vessel wall J Vasc Res. 2005;42:312.
interactions in the apolipoprotein E knockout mouse 398. Sutton JA. Nonselective nonsteroidal anti-
model of atherosclerosis. Circulation. 2003;108:3017. inflammatory drugs and increased cardiovascular
382. Buerkle MA, Lehrer S, Solon HY, et al. Selective events: stress could be the explanation. Br J Clin
inhibition of cyclooxygenase-2 enhances platelet Pharmacol. 2006;63:501.
adhesion in hamster arterioles in vivo. Circulation. 399. Knights KM, Mangoni A, Miners JO. Non-selective
2004;110:2053. anti-inflammatory drugs and cardiovascular events:
383. Kearney D, Byrne A, Cream P, et al. Optimal sup- is aldosterone the silent partner in crime? Br J Clin
pression of thromboxane A2 formation by aspirin Pharmacol. 2006;61:738.
during percutaneous transluminal coronary angio- 400. Kearney PM, Baigent C, Godwin J, et al. Do selec-
plasty: no additional effect of a selective cyclooxy- tive cyclo-oxygenase-2 inhibitors and traditional
genase-2 inhibitor. J Am Coll Cardiol. 2004;43:526. non-steroidal anti-inflammatory drugs increase the
384. Bresalier RS, Sandler RS, Quan H, et al. risk of atherothrombosis? Meta-analysis of random-
Cardiovascular events associated with rofecoxib in a ized trials. BMJ. 2006;332:1302.
colorectal adenoma chemoprevention trial. N Engl J 401. Gislason GH, Jacobsen S, Rasmussen JN, et al. Risk
Med. 2005;352:1092. of death or reinfarction associated with the use of
385. Solomon SD, McMurray JJ, Pfeiffer MA, et al. selective cyclooxygenase-2 inhibitors and nonselec-
Cardiovascular risk associated with celecoxib in a tive nonsteroidal anti-inflammatory drugs after acute
clinical trial for colorectal adenoma prevention. N myocardial infarction. Circulation. 2006;113:2906.
Engl J Med. 2005;352:1071. 402. McGettigan P, Han P, Henry D. Cyclooxygenase-2
386. Furberg CD, Psaty BM, Fitzgerald GA. Parecoxib, inhibitors and coronary occlusionexploring dose–
valdecoxib, and cardiovascular risk. Circulation. response relationships. Br J Clin Pharmacol. 2006;
2005;111:249. 62:358.
387. Hochman JS, Shah NR. What price pain relief? 403. Andersohn F, Schade R, Suissa S, Garbe E.
Circulation. 2006;113:2868. Cyclooxygenase-2 selective nonsteroidal anti-
388. Rahme E, Nedjar H. Risks and benefits of COX-2 inflammatory drugs and the risk of ischemic stroke.
inhibitors vs. non-selective NSAIDS: does their car- A nested case–control study. Stroke. 2006;37:1725.
diovascular risk exceed their gastrointestinal benefit? 404. Hinz B, Renner B, Brune K. Drug insight: cyclo-
A respective cohort study. Rheumatology. 2007; oxygenase-2 inhibitors- a critical appraisal. Nat Clin
46:435. Pract Rheumatol. 2007;3:552.
References 367

405. Brune K. Do case control studies on coxibs tell us 420. Balk SH, Kwon TK, Lim JH, et al. Secretory phos-
anything new? Rheumatology. 2007;46:730. pholipase A2-potentiated inducible nitric oxide syn-
406. Garcia Rodriguez LA, Varas C, Patrano C. thase expression by macrophages requires NF-kappa
Differential effects of aspirin and non-aspirin non- B activation. J Immunol. 2000;164:6359.
steroidal anti-inflammatory drugs in the primary pre- 421. Roshak AK, Jackson JR, McGough K, et al.
vention of myocardial infarction in postmenopausal Manipulation of distinct NF kappa B proteins alters
women. Epidemiology. 2000;11:382. interleukin-1 beta-induced human rheumatoid syn-
407. Andersohn F, Suissa S, Garber E. Use of first and ovial fibroblast prostaglandin E2 formation. J Biol
second generation cyclooxygenase-2- selective Chem. 1996;271:31496.
nonsteroidal anti-inflammatory drugs and risk of 422. Angel J, Berenbaum F, LeDenmat C, et al.
acute myocardial infarction. Circulation. 2006; Interleukin-1-induced prostaglandin E2 biosynthesis
113:1950. in human synovial cells involves the activation of
408. Johnson SP, Larsson H, Tarone RE, et al. Risk of cytosolic phospholipase A2 and cyclooxygenase-2.
hospitalization for myocardial infarction among Eur J Biochem. 1994;226:125.
users of rofecoxib, celecoxib, and other NSAIDS: a 423. Bidgood MJ, Jamal OS, Cunningham AM, et al.
population-based case–control study. Arch Intern Type IIA secretory phospholipase A2 up-regulates
Med. 2005;165:978. cyclooxygenase-2 and amplifies cytokine-mediated
409. Matsko SP, Rascati KL, Busti AJ, et al. Temporal prostaglandin production in human rheumatoid syn-
relationship between use of NSAIDS, including oviocytes. J Immunol. 2000;165:2790.
selective COX-2 inhibitors, and cardiovascular risk. 424. Vedgar S, Lichtenberg D, Schnitzer E. Inhibition of
Drug Saf. 2006;29:621. phospholipase A(2) as a therapeutic target. Biochim
410. Singh G, Lanes S, Triadafilopoulas G. Risk of seri- Biophys Acta. 2000;1488:182.
ous upper gastrointestinal and cardiovascular throm- 425. Munoz NM, Kim KP, Han SK, et al. Characterization
boembolic complications with meloxicam. Am J of monoclonal antibodies specific for 14-kDa human
Med. 2004;117:100. group V secretory phospholipase A2 (hVPLA2).
411. Forman JP, Rimm EB, Curhan GC. Frequency of Hybridoma. 2000;19:171.
analgesic use and risk of hypertension among men. 426. Morris HG, DeRoche G, Caro CM. Detection of
Arch Intern Med. 2007;167:394. synthetic corticosteroid analogues by the competi-
412. Neogi T. Gout. N Engl J Med. 2011;364:443–52. tive protein-binding assay. Steroids. 1973;22:445.
413. Kalgutkar AS, Marnett AB, Crews BC, et al. Ester 427. Schleimer RP. Glucocorticoids: their mechanisms of
and amide derivatives of the nonsteroidal anti- action and use in allergic diseases. In: Middleton Jr
inflammatory drug, indomethacin, as selective E, Reed CE, Ellis EF, Adkinson Jr NF, Yunginger
cyclooxygenase-2 inhibitors. J Med Chem. 2000; JW, editors. Allergy principles and practice, vol. 1.
43:2860. 3rd ed. St. Louis: CV Mosby; 1989. p. 739–65.
414. Bomalaski JS, Baker DG, Brophy LM, Clark MA. 428. Lewis GP, Jusko WJ, Burke CW, Graves L.
Monosodium urate crystals stimulate phospholipase Prednisone side-effects and serum-protein levels.
A2 enzyme activities and the synthesis of phospholi- Lancet. 1971;2:778.
pase A2-activating protein. J Immunol. 1990; 429. Bray R, Abrams S, Brahmi Z. Studies on the mecha-
145:3391. nism of human natural killer cell-mediated cytolysis.
415. Reddy ST. Prostaglandin synthase-1 and prostaglan- I. Modulation by dexamethasone and arachidonic
din synthase-2 are coupled to distinct phospholipases acid. Cell Immunol. 1983;78:100.
for the generation of prostaglandin D2 in activated 430. Indiveri F, Scudeletti M, Pende D, et al. Inhibitory
mast cells. J Biol Chem. 1997;272:3231. effect of a low dose of prednisone on PHA-induced
416. Marshall J, Krump E, Lindsay T, et al. Involvement Ia antigen expression by human T cells and on pro-
of cytosolic phospholipase A2 and secretory phos- liferation of T cells stimulated with autologous
pholipase A2 in arachidonic acid release from human PHA-T cells. Cell Immunol. 1983;84:311.
neutrophils. J Immunol. 2000;164:2084. 431. Gerrard TL, Cupps FR, Jurgensen CH, Fauci AS.
417. Suzuki N, Ishizaki J, Yokota Y, et al. Structures, Increased expression of HLA-DR antigens in hydrocor-
enzymatic properties, and expression of novel human tisone-treated monocytes. Cell Immunol. 1984;84:311.
and mouse secretory phospholipase A(2)s. J Biol 432. Gerrard TL, Cupps FR, Jurgensen CH, Fauci AS.
Chem. 2000;275:5785. Hydrocortisone-mediated inhibition of monocyte
418. Murakami M, Naraba H, Tanioka T, et al. Regulation antigen presentation: dissociation of inhibitory effect
of prostaglandin E2 biosynthesis by inducible mem- and expression of DR antigens. Cell Immunol. 1984;
brane-associated prostaglandin E2 synthase that acts 85:330.
in concert with cyclooxygenase-2. J Biol Chem. 433. Munck A, Mendel DB, Smith LI, Orti E. Gluco-
2000;275:32783. corticoid receptors and actions. Am Rev Respir Dis.
419. Balk SH, Kwon TK, Lim JH, et al. Secretory phos- 1990;141:S2.
pholipase A2-potentiated inducible nitric oxide syn- 434. Blackwell GJ, Carnuccio R, DiRosa M, et al. Macro-
thase expression by macrophages requires NF-kappa cortin: a polypeptide causing the antiphospholipase
B activation. J Immunol. 2000;164:6359. effect of corticosteroids. Nature. 1980;287:147.
368 9 Management of Hyperuricemia and Gout

435. Hirata F, Axelrod J. Phospholipid methylation and bio- 452. Kujubu DA, Reddy ST, Fletcher BS, Herschman H.
logical signal transmission. Science. 1980;20:1082. Expression of the protein product of the prostaglan-
436. Gupta C, Goldman AS. Dexamethasone-induced din synthase 2/TIS10 gene in mitogen-stimulated
phospholipase A2-inhibitory proteins (PLIP) Swiss 3T3 cells. J Biol Chem. 1993;268:5425.
influenced by H-2 histocompatibility region. Proc 453. Sebaldt RJ, Sheller JR, Oates JA, et al. Inhibition of
Soc Exp Biol Med. 1985;178:29. eicosanoid biosynthesis by glucocorticoids in
437. Gupta C, Katsumata M, Goldman AS, et al. humans. Proc Natl Acad Sci USA. 1990;87:6974.
Glucocorticoid-induced phospholipase A2-inhibitory 454. Goppelt-Struebe M, Wolter D, Resch K.
proteins mediate glucocorticoid teratogenicity Glucocorticoids inhibit prostaglandin synthesis not
in vitro. Proc Natl Acad Sci USA. 1984;81:1140. only at the level of phospholipase A2 but also at the
438. Webel ML, Ritts RE, Taswell HF, et al. Cellular level of cyclooxygenase/PGE isomerase. Br J
immunity after intravenous administration of meth- Pharmacol. 1989;98:1287.
ylprednisolone. J Lab Clin Med. 1974;83:383. 455. Bailey JM, Makheja AN, Pash J, Verma M.
439. Peterson AP, Altman LC, Hill JS, et al. Glucocorticoid Corticosteroids suppress cyclooxygenase messenger
receptors in normal human eosinophils: comparison RNA levels and prostanoids synthesis in cultured
with neutrophils. J Allergy Clin Immunol. 1981; vascular cells. Biochem Biophys Res Commun.
62:212. 1988;157:1159.
440. van der Velden VH. Glucocorticoids: mechanisms of 456. Seibert K, Masferrer JL, Fu J-Y, et al. The biochemi-
action and anti-inflammatory potential in asthma. cal and pharmacological manipulation of cellular
Mediators Inflamm. 1998;7:229. cyclooxygenase (COX) activity. Adv Prostaglandin
441. Beato M, Candau R, Chavez S, et al. Interaction of Thromboxane Leukot Res. 1990;21:45.
steroid hormone receptors with transcription factors 457. Neogi T. Interleukin-1 antagonism in acute gout: is
involves chromatin remodeling. J Steroid Biochem targeting a single cytokine the answer? Arthritis
Mod Biol. 1996;56:47. Rheum. 2011;62:2845–9.
442. Biola A, Pallardy M. Mode of action of glucocorti- 458. Goulding NJ, Pan L, Wardwell K, et al. Evidence for
coids. Presse Med. 2000;29:215. specific annexin-1-binding proteins on human mono-
443. Weigel NL. Steroid hormone receptors and their cytes. Biochem J. 1996;316:593.
regulation by phosphorylation. Biochem J. 1996; 459. Goulding NJ, Dixey J, Morand EF, et al. Differential
319:657. distribution of annexins-1, -II, -IV, and –VI in syn-
444. Xie W, Robertson DL, Simmons DL. Mitogen- ovium. Ann Rheum Dis. 1995;54:841.
inducible prostaglandin G/H synthase: new target for 460. Sampey AV, Hutchinson P, Morand EF. Annexin 1
nonsteroidal anti-inflammatory drugs. Drug Dev surface binding sites and their regulation of human
Res. 1992;25:249. fibroblast-like synoviocytes. Arthritis Rheum. 2000;
445. Maier JA, Hla T, Maciag T. Cyclooxygenase is an 43:2537.
intermediate-early gene induced by interleukin-1 in 461. Yang Y, Leech M, Hutchinson P, et al.
human endothelial cells. J Biol Chem. 1990; Antiinflammatory effect of lipocortin 1 in experi-
265:10805. mental arthritis. Inflammmation. 1997;21:583.
446. Lee SH, Soyoola E, Chanmugam P, et al. Selective 462. Yang YH, Hutchinson P, Santos LL, Morand EF.
expression of mitogen-inducible cyclooxygenase in Glucocorticoid inhibition of adjuvant arthritis syn-
macrophages stimulated with lipopolysaccharide. J ovial nitric oxide production: role of lipocortin 1.
Biol Chem. 1992;267:25934. Clin Exp Immunol. 1998;111:117.
447. Szczepanski A, Moatter T, Carley WW, Gerritsen 463. Yang Y, Hutchinson P, Morand EF. Inhibitory effect
ME. Induction of cyclooxygenase II in human syn- of annexin 1 on synovial inflammation in rat adju-
ovial microvessel endothelial cells by interleukin-1. vant arthritis. Arthritis Rheum. 1999;42:1538.
Arthritis Rheum. 1994;37:495. 464. Solito E, Nuti S, Parente L. Dexamethasone-induced
448. O’Banion MK, Sadowski HB, Winn V, Young DA. translocation of lipocortin (annexin1) to the cell
A serum- and glucocorticoid-regulated 4-kilobase membrane of U-937 cells. Br J Pharmacol. 1994;
mRNA encodes a cyclooxygenase-related protein. J 112:347.
Biol Chem. 1991;266:23261. 465. Bailey JM. New mechanisms for effects of anti-
449. Raz A, Wyche A, Needleman P. Temporal and phar- inflammatory glucocorticoids. Biofactors. 1991;3:97.
macological division of fibroblast cyclooxygenase 466. Bastian BC, Romisch J, Paques EP, Burg G. Lipocortins
expression into transcriptional and translational and phospholipases: new aspects in the physiology of
phases. Proc Natl Acad Sci USA. 1989;86:1657. glucocorticoid effect. Hautarzt. 1991;42:417.
450. Fu J-Y, Masferrer JL, Seibert K, et al. The induction 467. Masferrer JL, Siebert K. Regulation of prostaglandin
and suppression of prostaglandin H synthase synthesis by glucocorticoids. Receptor. 1994;4:25.
(cyclooxygenase) in human monocytes. J Biol Chem. 468. Croxtall JD, Choudhury Q, Tokumoto T, Flower RJ.
1990;265:16737. Lipocortin-1 and the control of arachidonic release in
451. Kujubu DA, Herschman H. Dexamethasone inhibits cell signalling. Glucocorticoids (changed from glu-
mitogen induction of TIS10 prostaglandin synthase corticoids) inhibit G protein-dependent activation of
(cyclooxygenase) gene. J Biol Chem. 1992;267:7991. cPLA2 activity. Biochem Pharmacol. 1995;50:465.
References 369

469. Minghetti L, Nicolini A, Polazzi E, et al. Down- IL-1 or tumor necrosis factor. J Immunol. 1990;
regulation of microglial cyclooxygenase-2 and 146:1212.
inducible nitric oxide synthase expression by lipoco- 485. Snyers L, Wit LD, Content J. Glucocorticoid upreg-
rtin 1. Br J Pharmacol. 1999;126:1307. ulation of high affinity interleukin-6 receptors on
470. Buckland AG, Wilton DC. Inhibition of human cyto- human epithelial cells. Proc Natl Acad Sci USA.
solic phospholipase A2 by human annexin V. 1990;87:2838.
Biochem J. 1998;329:369. 486. Rose B, Brown JSL. The effect of adrenalectomy on
471. Buckland AG, Wilton DC. Inhibition of secreted phos- the histamine content of the tissues of the rat. Am J
pholipases A2 by annexin V. Competition for anionic Physiol. 1941;131:589.
phospholipid interfaces allows an assessment of the 487. Daynes RA, Araneo BA. Contrasting effects of glu-
relative interfacial affinities of secreted phospholi- cocorticoids on the capacity of T cells to produce the
pases A2. Biochim Biophys Acta. 1998;1391:367. growth factors interleukin 2 and interleukin 4. Eur J
472. Koumanov K, Wolf C, Bereziat G. Modulation of Immunol. 1989;19:2319.
human type II secretory phospholipase A2 by sphin- 488. Chensue SW, Terebuh PD, Remick DG, et al. In vivo
gomyelin and annexin VI. Biochem J. 1997;326:227. biologic and immunohistochemical analysis of inter-
473. Mira JP, Dubois T, Oudinet JP, et al. Inhibition of leukin-1a, b and tumor necrosis factor during exper-
cytosolic phospholipase A2 by annexin V in differ- imental endotoxemia. Kinetics, Kupffer cell
entiated permeabilized HL-60 cells. Evidence of expression and glucocorticoid effects. Am J Pathol.
crucial importance of domain I type II Ca2+-binding 1991;138:395.
site in the mechanism of inhibition. J Biol Chem. 489. Makaida N, Shiroo M, Matsushima K. Genomic
1997;272:10474. structure of the human monocyte-derived neutrophil
474. Goppelt-Struebe M. Molecular mechanisms involved chemotactic factor, IL-8. J Immunol. 1989;143:1366.
in the regulation of prostaglandin biosynthesis by 490. Hart PH, Whitty GA, Burgess DR, et al. Augmentation
glucocorticoids. Biochem Pharmacol. 1997;53:1389. of glucocorticoid action on human monocytes by
475. Morand EF, Hutchinson P, Hargreaves A, et al. interleukin-4. Lymphokine Res. 1990;9:147.
Detection of intracellular lipocortin 1 in human leu- 491. Valent P, Bevec D, Maurier D, et al. Interleukin-4
kocyte subsets. Clin Immunol Immunopathol. 1995; promotes expression of mast cell ICAM-1 antigen.
76:195. Proc Natl Acad Sci USA. 1991;88:3399.
476. Euzger HS, Flower RJ, Goulding NJ, Perretti M. 492. Durant S, Homo-Delarche F, Duval D, et al. Opposite
Differential modulation of annexin 1 binding sites effects of glucocorticoid and an immunostimulating
on monocytes and neutrophils. Mediators Inflamm. agent on prostaglandin production by two different
1999;8:53. cell types. Adv Prost Thromb Res. 1985;7:117.
477. Comera C, Russo-Marie F. Glucocoriticoid-induced 493. Dunsky EH, Zweiman B, Fishchler E, Levy DA.
annexin 1 secretion by monocytes and peritoneal Early effects of corticosteroids on basophils, leuko-
leukocytes. Br J Pharmacol. 1995;115:1043. cyte histamine and tissue histamine. J Allergy Clin
478. Sebalt RJ, Sheller JR, Oates JA, et al. Inhibition of Immunol. 1979;64:426.
eicosanoid biosynthesis by glucocorticoids in 494. Savendra-Delgado AM, Mathews KP, Pan PM, et al.
humans. Proc Natl Acad Sci USA. 1990;87:6974. Dose–response studies of the suppression of whole
479. Butler WT, Rossen RD. Effects of corticosteroids in blood histamine and basophil counts by prednisone.
immunity in man. I. Decreased serum IgG concen- J Allergy Clin Immunol. 1980;66:464.
tration caused by 3 or 5 days of high doses of meth- 495. Becker J, Grasso RJ. Suppression of phagocytosis by
ylprednisolone. J Clin Invest. 1973;52:2629. dexamethasone in macrophage cultures: inability of
480. Kumar L, Newcomb RW, Ishizaka K, et al. IgE lev- arachidonic acid, indomethacin, and nordihydrogua-
els in sera of children with asthma. Pediatrics. iaretic acid to reverse the inhibitory response mediated
1973;47:848. by a steroid-inducible factor. Int J Immunopharmacol.
481. Bush RK, Geller M, Busse WW, et al. Response to 1985;7:839.
corticosteroids in the hypereosinophilic syndrome: 496. Schleimer RP. Effects of glucocorticoids on
association with increased IgE levels. Arch Intern inflammatory cells relevant to their therapeutic appli-
Med. 1978;138:1244. cations to asthma. Am Rev Respir Dis. 1990;141:S59.
482. Ricketti AJ, Greenberger PA, Patterson R. Serum 497. Ward PA. The chemosuppression of chemotaxis. J
IgE as an important aid in management of allergic Exp Med. 1966;124:209.
bronchopulmonary aspergillosis. J Allergy Clin 498. Wenck U, Speirs R. The effect of cortisone on blood
Immunol. 1984;74:68. leukocytes and peritoneal fluid cells of mice. Acta
483. Dauchel H, Julen N, Lemercier C, et al. Expression of Haematol. 1957;17:193.
complement alternative pathway proteins by endothe- 499. Thompson J, van Furth R. The effect of glucocorti-
lial cells. Differential regulation by interleukin 1 and coids on the kinetics of mononuclear phagocytes. J
glucocorticoids. Eur J Immunol. 1990;20:1669. Exp Med. 1970;131:429.
484. Mukaida N, Zachariae CO, Gusella GL, Matsushima 500. Rindhart JJ, Sagone AI, Balcerzak SP, et al. Effects
K. Dexamethasone inhibits the induction of mono- of corticosteroid therapy on human monocyte func-
cyte chemotactic-activating factor production by tion. N Engl J Med. 1975;292:236.
370 9 Management of Hyperuricemia and Gout

501. Melewicz FM, Zeiger RS, Melton MH, et al. oral indomethacin/paracetamol combination therapy
Increased peripheral blood monocytes with Fc recep- in the treatment of acute goutlike arthritis: a double-
tors for IgE in patients with severe allergic disorders. blind, randomized, controlled trial. Ann Emerg Med.
J Immunol. 1981;126:1592. 2007;49:670–7.
502. Altman LC, Hill JS, Hairfield WM, Mullarkey MF. 518. Janssens HJ, Janssen M, van de Lisdonk EH, van
Effects of corticosteroids on eosinophil chemotaxis Riel PL, van Weel C. Use of oral prednisolone or
and adherence. J Clin Invest. 1981;67:28. naproxen for the treatment of gout arthritis: a dou-
503. Allcock GH, Allegra M, Flower RJ, Perretti M. ble-blind, randomised equivalence trial. Lancet.
Neutrophil accumulation induced by bacterial 2008;371:1854–60.
lipopolysaccharide: effects of dexamethasone and 519. Fernandez C, Nouera R, Gonzalez JA, Pascual E.
annexin 1. Clin Exp Immunol. 2001;123:62. Treatment of acute attacks of gout with a small dose
504. Perretti M, Flower RJ. Cytokines, glucocorticoids of intraarticular triamcinolone acetonide. J
and lipocortins in the control of neutrophil migra- Rheumatol. 1999;26:2285.
tion. Pharmacol Res. 1994;30:53. 520. Frey FJ. Kinetics and dynamics of prednisolone.
505. Oliani SM, Paul-Clark MJ, Christian HC, et al. Endocrinol Rev. 1987;8(453):530.
Neutrophil interaction with inflamed postcapillary 521. Renner E, Harber FF, Jost G, et al. Effect of liver
venule endothelium alters annexin 1 expression. Am function on the metabolism of prednisone and pred-
J Pathol. 2001;158:603. nisolone in humans. Gastroenterology. 1966;90:819.
506. Tailor A, Tomlinson A, Salas A, et al. Dexamethasone 522. Gambertoglio JG, Amend Jr WJC, Benet LZ.
inhibition of leucocyte adhesion to rat mesenteric Pharmacokinetics and bioavailability of prednisone
postcapillary venules: role of intercellular adhesion and prednisolone in health volunteers and patients.
molecule 1 and KC. Gut. 1999;45:705. A review. J Pharmacokinet Biopharm. 1980;8:1.
507. Mancuso F, Flower RJ, Perretti M. Leucocyte trans- 523. Axelrod D, Preston S. Comparison of parental adre-
migration, but not rolling or adhesion, is selectively nocorticotrophic hormone with oral indomethacin in
inhibited by dexamethasone in the hamster post- the treatment of acute gout. Arthritis Rheum. 1988;
capillary venule. Involvement of endogenous lipoco- 31:803.
rtin 1. J Immunol. 1995;155:377. 524. Schroder K, Tschopp J. The inflammasomes. Cell.
508. Lim LH, Solito E, Russo-Marie F, et al. Promoting 2010;140:821–32.
detachment of neutrophils adherent to murine postcap- 525. Dalbeth N, Merriman T. Crystal ball gazing: new
illary venules to control inflammation: effect of lipo- therapeutic targets for hyperuricaemia and gout.
cortin 1. Proc Natl Acad Sci USA. 1998;95:14535. Rheumatology. 2008;48:222–6.
509. Perretti M, Croxtall JD, Wheller SK, et al. Mobilizing 526. So A, De Smedt T, Revaz S, Tschopp J. A pilot study
lipocortin 1 in adherent human leukocytes downreg- of IL-1 inhibition by anakinra in acute gout. Arthritis
ulates their transmigration. Nat Med. 1996;2:1259. Res Ther. 2007;9:R28.
510. Getting SJ, Flower RJ, Perretti M. Inhibition of neu- 527. So A, De Meukemeester M, Pikhlak A, et al.
trophil and monocyte recruitment by endogenous Canakinumab for the treatment of acute flares in
and exogenous lipocortin 1. Br J Pharmacol. 1997; difficult-to-treat gouty arthritis: results of a multi-
120:1075. center, phase II. Dose-Ranging Study. Arthritis
511. Perretti M, Flower RJ. Measurement of lipocortin 1 Rheum. 2010;62:3064–76.
levels in murine peripheral blood leucocytes by flow 528. Martinon F, Petrilli V, Mayor A, et al. Gout-
cytometry: modulation by glucocorticoids and associated uric acid crystals activate the NALP3
inflammation. Br J Pharmacol. 1996;118:605. inflammasome. Nature. 2006;440:237–41.
512. Walther A, Riehemann K, Gerke V. A novel ligand 529. Terkeltaub R, Sundy JS, Schumacher HR, et al. The
of the formyl peptide receptor: annexin 1 regulates interleukin-1 inhibitor rilonacept in treatment of
neutrophil extravasation by interacting with the FPR. chronic gouty arthritis: results of a placebo-controlled,
Mol Cell. 2000;5:831. monosequence crossover, non-randomised. Single-
513. Green PG, Strausbaugh HJ, Levine JD. Annexin 1 is Blind Pilot Study. Ann Rheum Dis. 2009;68:1613–7.
a local mediator in neural-endocrine feedback of 530. Schumacher Jr HR, Sundy JS, Terkeltaub R, et al.
inflammation. J Neurophysiol. 1998;80:3120. Placebo-controlled study of rilonacept for gout flare
514. Dandona P, Thusu K, Hafeez R, et al. Effect of prophylaxis during initiation of urate-lowering ther-
hydrocortisone on oxygen free radical generation by apy (abstract). Arthritis Rheum. 2009;60:S410–1.
mononuclear cells. Metabolism. 1998;47:788. 531. Buchanan MR. Sulfinpyrazone: relationship between
515. Fahey JV, Newcombe DS. Regulation of bradykinin- dose, kinetics, plasma concentration and biological
induced cyclic AMP response by quinacrine and effects. Thromb Res. 1983;4(Suppl):89.
prostaglandin E2 and F2 alpha in human synovial 532. Buchanan MR, Endrengi L, Giles AR, Rosenfeld J.
fibroblasts. Inflammation. 1979;3:235. The effect of aspirin on the pharmacokinetics of
516. Newcombe DS, Fahey JV, Ishikawa Y. Hydrocortisone sulfinpyrazone in man. Thromb Res. 1983;4(Suppl):
inhibition of the bradykinin activation of human syn- 145.
ovial fibroblasts. Prostaglandins. 1977;13:235. 533. Buchanan MR, Rosenfeld J, Hirsch J. The prolonged
517. Man CY, Cheung IT, Cameron PA, Rainer TH. effect of sulfinpyrazone on collagen-induced platelet
Comparison of oral prednisolone/paracetamol and aggregation in vivo. Thromb Res. 1978;13:883.
References 371

534. Wallis RB. Mechanism of action of sulfinpyrazone. and intravenous doses in normal young men. Am J
Thromb Res. 1983;4(Suppl):31. Med Sci. 1965;249:163.
535. Wiley JS, Chesterman CN, Morgan FJ, Castaldi PA. 554. Brater DC. Increase in diuretic effect of chlorothiaz-
The effect of sulfinpyrazone on the aggregation and ide by probenecid. Clin Pharmacol Ther.
release reactions of human platelets. Thromb Res. 1978;23:259.
1979;14:23. 555. Brooks PM, Bell MA, Sturrock RD, et al. The clini-
536. Steele PO, Weily HS, Genton E. Platelet survival and cal significance of indomethacin-probenecid interac-
adhesiveness in recurrent venous thrombosis. N Engl tion. Br J Clin Pharmacol. 1974;1:287.
J Med. 1973;288:1148. 556. Goodwin CS, Sparell G. Inhibition of dapsone excre-
537. Sherry S. Clinical aspects of antiplatelet therapy. tion by probenecid. Lancet. 1969;2:884.
Semin Hematol. 1985;22:105. 557. Honari J, Blair A, Cutler RE. Effects of probenecid
538. Kaegi A, Pineo GF, Shimizo A, et al. Arteriovenous on furosemide kinetics and natriuresis in man. Clin
shunt and thrombosis by sulfinpyrazone. N Engl J Pharmacol Ther. 1977;22:395.
Med. 1974;290:304. 558. Laskin OL, de Miranda P, King DH, et al. Effects of
539. Weilly HS, Genton E. Altered platelet function in probenecid on the pharmacokinetics and elimination
patients with prosthetic mitral valves. Effect of of acyclovir in humans. Antimicrob Agents
sulfinpyrazone therapy. Circulation. 1970;42:967. Chemother. 1982;21:804.
540. Ogryzlo MA, Harrison J. Evaluation of uricosuric 559. Runkel R, Mrosczak E, Chaplin M, et al. Naproxen-
agents in chronic gout. Ann Rheum Dis. 1957;16:425. probenecid interaction. Clin Pharmacol Ther.
541. Emmerson BT. A comparison of uricosuric agents in 1978;24:710.
gout, with special reference to sulfinpyrazone. Med J 560. Schacter D, Manis JG. Salicylate and salicyl conju-
Aust. 1963;1:839. gates: fluorometric estimation, biosynthesis, and
542. Persellin RH, Schmid FR. The use of sulfinpyrazone renal excretion in man. J Clin Invest. 1958;37:800.
in the treatment of gout reduces serum uric acid levels 561. Weiner IM, Washington JA, Mudge GH. Studies on
and diminishes severity of arthritis attacks with free- the renal excretion of salicylate in the dog. Bull
dom from significant toxicity. JAMA. 1961;175:971. Johns Hopkins Hosp. 1959;105:284.
543. Diamond HS, Aolino JS. Evidence for a postsecre- 562. Weiner IM, Washington JA, Mudge GH. On the
tory reabsorption site for uric acid in man. J Clin mechanism of action of probenecid on renal tubular
Invest. 1973;52:1490. secretion. Bull Johns Hopkins Hosp. 1960;106:333.
544. Fanelli Jr GM, Weiner IM. Urate excretion: drug 563. Israili ZH, Perel JM, Cunningham RF, et al.
interactions. J Pharmacol Sup Ther. 1979;210:186. Metabolites of probenecid: chemical, physical and
545. Burns JJ, Yu T-F, Ritterand A, et al. A potent new pharmacological studies. J Med Chem. 1972;15:709.
uricosuric agent, the sulfoxide metabolite of the phe- 564. Perel JM, Dayton PG, Yu T-F, Gutman AB. Studies
nylbutazone analogue, G-25671. J Pharmacol Exp on the renal excretion of probenecid acyl glucuronide
Ther. 1957;119:418. in man. Eur J Clin Pharmacol. 1971;3:106.
546. Perel JM, Dayton PG, Snell MM, et al. Studies of 565. Schacter D. The chemical estimation of aryl
interactions among drugs in man at the renal level: glucuronides and its application to studies on the
probenecid and sulfinpyrazone. Clin Pharmacol metabolism of benzoate and salicylate in man. J Clin
Ther. 1974;10:834. Invest. 1957;36:297.
547. Yu T-F, Dayton PG, Gutman AB. Mutual suppres- 566. Yu T-F, Paton BC, Chenkin T, et al. Effect of
sion of the uricosuric effects of sulfinpyrazone and allopurinol (4-hydroxypyrazole (3, 4-d) pyrimidine)
salicylates: a study of interactions between drugs. J on serum and urinary uric acid in primary and sec-
Clin Invest. 1963;42:1330. ondary gout. Am J Med. 1964;37:885.
548. Domenjoz R. The pharmacology of the phenylbuta- 567. Gutman AB. Uricosuric drugs, with special refer-
zone analogues. Ann N Y Acad Sci. 1960;86:263. ence of probenecid and sulfinpyrazone. Adv
549. Fitzgerald GA, Sherry S. Pharmacology and phar- Pharmacol. 1988;21:337.
macokinetics of platelet-active drugs under current 568. Iven H, Brasch H. The effects of antibiotics and uri-
clinical investigations. Adv Prost Thromb Leuko. cosuric drugs on the renal elimination of methotrex-
1982;10:107. ate and 7-hydroxymethotrexate in rabbits. Cancer
550. Dieterle W, Faigle JW, Mory H, et al. Biotransformations Chemother Pharmacol. 1988;21:337.
and pharmacokinetics of sulfinpyrazone (Anturane) in 569. Paxton JW. Interaction of probenecid with the protein
man. Eur J Clin Pharmacol. 1975;9:135. binding of methotrexate. Pharmacology. 1984;28:86.
551. Mahoney C, Wolfram KM, Nash PV, Bjornsson TD. 570. Kates RE, Tozer TN, Sorby DL. Increased metho-
Kinetics and metabolism of sulfinpyrazone. Clin trexate toxicity due to concurrent probenecid admin-
Pharmacol Ther. 1983;33:491. istration. Biochem Pharmacol. 1976;25:1485.
552. Dayton PG, Sicam LE, Landrau M, Burns JJ. 571. Basin KS, Escalante A, Beardmore TD. Severe pan-
Metabolism of sulfinpyrazone (Anturane) and other cytopenia in a patient taking low dose methotrexate
analogues of phenylbutazone in man. J Pharmacol and probenecid. J Rheumatol. 1991;18:609.
Exp Med. 1961;132:287. 572. Caspi D, Liebart E, Graff E, et al. The effect of mini-
553. Eickhott TC, Kislak JW, Finland M. Sodium ampi- dose aspirin on renal function and uric acid handling
cillin: Absorption and excretion of intramuscular in elderly patients. Arthritis Rheum. 2000;43:103.
372 9 Management of Hyperuricemia and Gout

573. Burns CM, Wortmann RL. Gout therapeutics: new 590. Pascual E. Gout update: from lab to the clinic and
drugs for an old disease. Lancet. 2011;377:165–77. back. Curr Opin Rheumatol. 2000;12:213.
574. Mertz DP. Therapy with allopurinol and benzbroma- 591. Zurcher RM, Bock HA, Thiel G. Excellent uricosu-
rone, single and combined. Renal elimination of ric efficacy of benzbromarone in cyclosporine-A-
lithogenesis and colloid protective substances. Med treated renal transplant patients: a prospective study.
Klin. 1977;72:664. Nephrol Dial Transplant. 1984;9:548.
575. Broekhuysen J, Pacco M, Scon R, et al. Metabolism 592. Marcen R, Gallego N, Orofino L, et al. Impairment
of benzbromarone in man. Eur J Clin Pharmacol. of tubular secretion of urate in renal transplant
1972;4:125. patients on cyclosporine. Nephron. 1995;70:307.
576. de Vries JX, Walter-Sack I, Ittensohn A, Weber E. 593. Hausch R, Wilkerson M, Singh E, et al. Tophaceous
The isolation, identification and structure of a new gout of the thoracic spine presenting as back pain
hydroxylated metabolite of benzbromarone in man. and fever. J Clin Rheumatol. 1999;6:335.
Xenobiotica. 1989;19:1461. 594. Chaoui A, Garcia J, Kurt AM. Gouty tophus simulat-
577. Walter-Sack I, de Vries JX, Ittlesohn A, Weber E. ing soft tissue tumor in a heart transplant recipient.
Rapid and slow benzbromarone elimination pheno- Skeletal Radiol. 1997;26:626.
types in man: benzbromarone and metabolites 595. Peeters P, Sennesael J. Low-back pain caused by spinal
profiles. Eur J Clin Pharmacol. 1990;39:577. tophus – a complication of gout in a kidney transplant
578. Arnold PJ, Guserle R, Luckow V, et al. Liquid chro- recipient. Nephrol Dial Transplant. 1998;13:3245.
matography-mass spectrometry in metabolic 596. Chopra KF, Schneiderman PS, Grossman ME.
research. I. Metabolites of benzbromarone in human Finger pad tophi. Cutis. 1999;64:233.
plasma and urine. J Chromatogr. 1991;554:267. 597. Lhotta K, Gruber J, Sgonc R, et al. Apoptosis of
579. de Vries JX, Walter-Sack I, Voss A, et al. Metabolism tubular epithelial cells in familial juvenile gouty
of benzbromarone in man: structure of new oxidative nephropathy. Nephron. 1998;79:340.
metabolites, 6-hydroxy- and 1’-oxo-benzbromarone, 598. Perez Ruiz F, Calabozo M, Ferenendez Lopez J,
and the enantioselective formation and elimination et al. Treatment of chronic gout in patients with renal
of 1’-hydroxybenzbromarone. Xenobiotica. 1993;23: function impairment. J Clin Rheumatol. 1999;5:49.
1435. 599. Matzkies F. Long lasting normalization of uric acid
580. Delbarre F, Auscher C, Oliver JL, et al. Treatment of after combination therapy with 300 mg of allopurinol
hyperuricemia and gout with derivatives of benzo- and 60 mg of benzbromarone in patients with gout
furan. Sem Hop. 1967;43:1127. and hyperuricemia. Med Klin. 1992;87:460.
581. Sternon J, Kocheleff P, Courturier E, et al. The hypou- 600. Muller FO, Schall R, Groenewoud G, et al. The
ricemic effect of benzbromarone – study of 24 cases effect of benzbromarone on allopurinol/oxypurinol
(preliminary results). Acta Clin Belg. 1967;22:285. kinetics in patients with gout. Eur J Clin Pharmacol.
582. Sorenson LB, Levinson DJ. Clinical evaluation of 1993;44:69.
benzbromarone: a new uricosuric drug. Arthritis 601. Mertz DP, Eichhorn R. Does benzbromarone in ther-
Rheum. 1976;19:183. apeutic doses raise renal excretion of oxypurinol?
583. Heel RC, Brogden RN, Speight TM, Avery GS. Klin Wochenschr. 1984;62:1170.
Benzbromarone: a review of its pharmacological 602. Marinchev L, Kunev K. The effect of benzbromarone
properties and therapeutic use in gout and hyperuri- on hyperlipidemia in gout. Vutr Boles. 1990;29:99.
caemia. Drugs. 1977;14:349. 603. Takahashi H, Sato T, Shimoyama Y, et al. Potentiation
584. Matzkies F. Effects and side effects of benzbroma- of anticoagulant effect of warfarin caused by enanti-
rone in the initial treatment of hyperuricemia and oselective metabolic inhibition by the uricosuric agent
gout. Result of a field study of 3899 patients. Fortschr benzbromarone. Clin Pharmacol Ther. 1999;66:569.
Med. 1978;96:1619. 604. Sinclair D, Fox IH. The pharmacology of hypourice-
585. Kuzmits R, Bresnik W, Muller MM. The effect of mic effect of benzbromarone. J Rheumatol. 1975;
benzbromarone in gout patients with limited kidney 2:437.
function. Fortschr Med. 1979;97:2057. 605. Politta G, Barthoud S, Goudin G, et al. Mechanism
586. Masbernard A, Giudicelli CP. Ten years’ experience of the uricosuric action of benzbromarone. Praxis.
with benzbromarone in the management of gout and 1975;62:1345.
hyperuricemia. S Afr Med J. 1981;59:701. 606. Yu T-F. Pharmacokinetic and clinical studies of a
587. Berg H. Effectiveness and tolerance of long-term uri- new uricosuric agent – benzbromarone. J Rheumatol.
cosuric treatment. Z Gesamte Inn Med. 1990;45:719. 1976;3:305.
588. Roch-Ramel F, Guisan B, Diezi J. Effects of uricosu- 607. Mertz DP. Veanderungen der Serumkoncentration
ric and antiuricosuric agents on urate transport in von harnsaure unter der Wirkung von
human brush-border membrane vesicles. J Pharmacol Benzbromaronum. Munchener Med Wochenschr.
Exp Ther. 1997;280:839. 1969;111:491.
589. Fam AG. Difficult gout and new approaches for con- 608. Jain AK, Ryan JR, McMahon FG, Novack RJ. Effect
trol of hyperuricemia in the allopurinol-allergic of single oral doses of benzbromarone on serum and
patient. Curr Rheumatol Rep. 2001;3:29. urinary uric acid. Arthritis Rheum. 1974;17:149.
References 373

609. Schepers GW. Benzbromarone therapy in hyperuri- 627. Fox RM, Royse-Smith LH, Dubb JW, et al.
cemia: comparison with allopurinol and probenecid. Orotidinuria induced by allopurinol. Science. 1970;
J Int Med Res. 1981;9:511. 168:861.
610. Fromenty B, Fisch C, Labbe G, et al. Amiodarone 628. Kelley WN, Beardmore TD. Allopurinol: alteration
inhibits the mitochondrial beta-oxidation of fatty in pyrimidine metabolism in man. Science. 1970;
acids and produces microvesicular steatosis of the 169:380.
liver in mice. J Pharmacol Exp Ther. 1990;255:1371. 629. Boston Collaborative Drug Surveillance Program.
611. Fromenty B, Fisch C, Berson A, et al. Dual effect of Allopurinol in relation to bone marrow depression.
amiodarone on mitochondrial respiration. Initial pro- JAMA. 1974;227:1036.
tonophoric uncoupling effect followed by inhibition 630. Witten J, Frederickson PL, Mouridsen HT. The phar-
of the respiratory chain at the levels of complex I and macokinetics of cyclophosphamide in man after
complex II. J Pharmacol Exp Ther. 1990;255:1377. treatment with allopurinol. Acta Pharmacol Toxicol.
612. Spaniol M, Bracher R, Ha HR, et al. Toxicity of 1980;46:392.
amiodarone and amiodarone analogues on isolated 631. Rundles RW. Metabolic effects of allopurinol and
rat liver mitochondria. J Hepatol. 2001;35:628. alloxanthine. Ann Rheum Dis. 1966;25:615.
613. Kaufmann P, Torok M, Hanni A, et al. Mechanism of 632. Vessel ES, Passananti GT, Greene FE. Impairment of
benzarone- and benzbromarone-induced hepatic drug metabolism in man by allopurinol and nortrip-
toxicity. Hepatology. 2005;41:925. tyline. N Engl J Med. 1970;283:1484.
614. Hautekeete ML, Henrion J, Naegels S, et al. Severe 633. Heymsfield SB, Arteaga C, McManus C, et al.
hepatotoxicity related to benzarone: a recent report Measurement of muscle mass in humans: validity of
of three cases with two fatalities. Liver. 1995;15:25. the 24-hour urinary creatinine method. Am J Clin
615. Babany G, Larrey D, Pessayre D, et al. Chronic active Nutr. 1983;37:478.
hepatitis caused by benzarone. J Hepatol. 1987; 634. Rowe JW, Andres R, Tobin JD, et al. The effect of
5:332. age on creatinine clearance in men: a cross-sectional
616. van der Klauw MM, Hautman PM, Stricker BH, and longitudinal study. J Gerontol. 1976;31:155.
Spoelstra P. Hepatic injury caused by benzbroma- 635. Levey AS, Berg RL, Gassman JJ, et al. Creatinine
rone. J Hepatol. 1994;20:376. filtration, secretion and excretion during progressive
617. Klinenberg JR, Goldfinger SE, Seegmiller JE. The renal disease. Kidney Int. 1989;36 Suppl 27:573.
effectiveness of xanthine oxidase inhibitor in the 636. Shemesh O, Golbetz H, Kriss JP, Myers BD.
treatment of gout. Ann Intern Med. 1965;62:639. Limitations of creatinine as a filtration marker in
618. Rundles RW, Wyngaarden JB, Hitchings GH, et al. glomerulonephritis patients. Kidney Int. 1985;28:830.
Effects of a xanthine oxidase inhibitor in thiopurine 637. Terkeltaub RA. Gout. N Engl J Med. 2003;
metabolism, hyperuricemia, and gout. Trans Assoc 349:1647.
Am Physicians. 1963;76:126. 638. Shoji A, Yamanaka H, Kamatani N. A retrospective
619. Spector T. Inhibition of urate production by study of the relationship between serum urate and
allopurinol. Biochem Pharmacol. 1977;26:355. recurrent attacks of gouty arthritis: evidence for
620. Elion GB. Enzymatic and metabolic studies with reduction of recurrent gouty arthritis with antihype-
allopurinol. Biochem Pharmacol. 1977;26:355. ruricemic therapy. Arthritis Rheum. 2004;51:321.
621. Kelley WN. Effects of drugs on uric acid in man. 639. Perez-Ruiz F, Calabozo M, Pijoan JE, et al. Effect of
Annu Rev Pharmacol Toxicol. 1975;15:327. urate-lowering therapy on the velocity of size reduc-
622. Fox IH, Wyngaarden JB, Kelley WN. Depletion of tion of tophi in chronic gout. Arthritis Rheum.
erythrocyte phosphoribosylpyrophosphate in man, a 2002;47:356.
newly observed effect of allopurinol. N Engl J Med. 640. Rott KT, Agudelo CA. Gout. JAMA. 2003;289:2857.
1970;283:1177. 641. Yamanaka H, Tagashi R, Hakoda M, et al. Optimal
623. McCollister RJ, Gilbert Jr WR, Ashton DM, range of serum urate concentrations to minimize risk
Wyngaarden JB. Pseudofeedback inhibition of of gouty attacks during anti-hyperuricemic treat-
purine synthesis by 6-mercaptopurine ribonucleotide ment. Adv Exp Med Biol. 1998;431:13.
and other purine analogues. J Biol Chem. 1964; 642. Wortmann RL. Recent advances in the management
239:1560. of gout and hyperuricemia. Curr Opin Rheumatol.
624. Caskey CT, Ashton DM, Wyngaarden JB. The enzy- 2005;17:319.
mology of feedback inhibition of glutamine phos- 643. Li-Yu J, Clayburne G, Sieck M, et al. Treatment of
phoribosylpyrophosphate amidotransferase by purine chronic gout. Can we determine when urate stores
ribonucleotides. J Biol Chem. 1964;239:2570. are depleted enough to prevent attacks of gout? J
625. Kelley WN, Greene ML, Rosenbloom FM, et al. Rheumatol. 2001;28:577.
Hypoxanthine-guanine phosphoribosyltransferase 644. Reinders MK, Nijdam LC, van Roon EN, et al. A
deficiency and gout. Ann Intern Med. 1969;70:155. simple method for quantification of allopurinol and
626. Kelley WN, Rosenbloom FM, Muller J, Seegmiller oxypurinol in human serum by high-performance
JE. An enzymatic basis for variation in the response liquid chromatography with UV-detection. J Pharm
to allopurinol. N Engl J Med. 1968;278:287. Biomed Anal. 2007;45:312.
374 9 Management of Hyperuricemia and Gout

645. Day RO, Graham GG, Hicks M, et al. Clinical phar- 663. Shalom R, Rimbroth S, Rozenman D, Markel A.
macokinetics and pharmacodynamics of allopurinol Allopurinol-induced recurrent DRESS syndrome:
and oxypurinol. Clin Pharmacokinet. 2007;46:623. pathophysiology and treatment. Ren Fail.
646. Landgrebe AR, Nyhan WL, Coleman M. Urinary 2008;30:327.
tract stones resulting from the excretion of oxy- 664. Markel A. Allopurinol-induced recurrent DRESS
purinol. N Engl J Med. 1975;292:626. syndrome. Isr Med Assoc J. 2005;7:656.
647. Stote RM, Smith LH, Dubb JW, et al. Oxypurinol 665. Dia B, Ba-Fall K, Bouldouryre M, et al. DRESS syn-
nephrolithiasis in regional enteritis secondary to drome to allopurinol: a case in Dakar. Dakar Med.
allopurinol therapy. Ann Intern Med. 1980;92:384. 2004;49:114.
648. Watts RWE, Sneeden W, Parker RA. A quantitative 666. Marrakchi C, Kanoun F, Kilani B, et al. Allopurinol-
study of skeletal muscle purines and pyrazolo (3, induced DRESS syndrome. Rev Med Interne.
4-d) pyrimidine. Clin Sci. 1971;41:153. 2004;25:252.
649. Turnheim K, Krivaneck P, Oberbauer R. 667. Roujeau JC, Stern RS. Severe adverse cutaneous
Pharmacokinetics and pharmacodynamics of reactions to drugs. N Engl J Med. 1994;331:1272.
allopurinol in the elderly and young subjects. Br J 668. Sontheimer R, Houpt KR. DIDMOHS: a proposed
Clin Pharmacol. 1999;48:501. consensus nomenclature for the drug induced
650. Hande KR, Noone RM, Stone WT. Severe allopurinol delayed multiorgan hypersensitivity syndrome. Arch
toxicity. Description and guidelines for prevention in Dermatol. 1998;134:874.
patients with renal insufficiency. Am J Med. 1978; 669. Bocquet H, Bagot M. Drug-induced pseudolym-
76:47. phoma and hypersensitivity syndrome (drug rash
651. Boston Collaborative Drug Surveillance Program. with eosinophilia and systemic symptoms: DRESS.
Excess of ampicillin rashes associated with Semin Cutan Med Surg. 1996;15:250.
allopurinol or hyperuricemia. N Engl J Med 1972; 670. Bigby M, Jick S, Jick H, Arndt K. Drug-induced cuta-
286:505. neous reactions: a report from the Boston Collaborative
652. Emmerson BT, Hazelton RA, Frazer IH. Some Drug Surveillance Program on 15,438 consecutive
adverse reactions to allopurinol may be mediated by inpatients, 1975–1982. JAMA. 1986;256:3358.
lymphocyte reactivity to oxypurinol. Arthritis 671. Alanko K, Stubb S, Kauppinen K. Cutaneous drug
Rheum. 1988;31:436. reactions: clinical types and causative agents: a five-
653. Garbe E, Suissa S, LeLorier J. Exposure to allopurinol year survey of in-patients (1981–1985). Acta Derm
and the risk of cataract extraction in elderly patients. Venereol Suppl. 1989;69:223.
Arch Ophthalmol. 1998;116:1652. 672. Ives TJ, Bentz EJ, Gwyther RE. Dermatologic
654. Zell SC, Carmichael JM. Evaluation of allopurinol adverse drug reactions in a family medicine setting.
use in patients with gout. Am J Hosp Pharm. 1989; Arch Fam Med. 1992;1:241.
46:1813. 673. Leape LL, Brennan TA, Laird N, et al. The nature of
655. Tjandramaga TB, Cucinall SD, Israili ZH, et al. adverse drug reactions in hospitalized patients:
Observation of the disposition of probenecid in results of the Harvard Medical Practice Study II. N
patients receiving allopurinol. Pharmacology. 1972; Engl J Med. 1990;324:377.
8:259. 674. Chan HL, Stern RS, Arndt KA, et al. The incidence
656. Hung SI, Chung WH, Liou LB, et al. HLA-B*5801 of erythema multiforme, Stevens-Johnson syndrome,
allele as a genetic marker for severe cutaneous and toxic epidermal necrolysis: a population-based
adverse reactions caused by allopurinol. Proc Natl study with particular reference to reactions caused
Acad Sci USA. 2005;102:4134. by drugs among outpatients. Arch Dermatol. 1991;
657. Arellano F, Sacristan JA. Allopurinol hypersensitiv- 126:43.
ity syndrome: a review. Ann Pharmacother. 1993; 675. Schopf E, Stuhmer A, Rzany B, et al. Toxic epider-
27:337. mal necrolysis and Stevens-Johnson syndrome: an
658. Aubock J, Fritsch P. Asymptomatic hyperuricaemia epidemiologic study from West Germany. Arch
and allopurinol induced toxic epidermal necrolysis. Dermatol. 1991;127:839.
Br Med J. 1985;290:1969. 676. Revuz J, Penso D, Roujeau JC, et al. Toxic epider-
659. Wortmann RL. Gout and hyperuricemia. Curr Opin mal necrolysis: clinical findings and prognosis fac-
Rheumatol. 2002;14:281. tors in 87 patients. Arch Dermatol. 1987;123:1160.
660. Roujeau JC, Kelly JP, Naldi L, et al. Medication use 677. Anquier-Dunant A, Mockenhaupt M, Naldi L, et al.
and the risk of Stevens-Johnson syndrome or toxic Correlations between clinical patterns and causes of
epidermal necrolysis. N Engl J Med. 1995;333:1600. erythema multiforme majus, Stevens-Johnson syn-
661. Halevy S, Ghislain PD, Mockenhaupt M, et al. drome, and toxic epidermal necrolysis: results of an
Allopurinol is the most common cause of Stevens- international prospective study. Arch Dermatol.
Johnson syndrome and toxic epidermal necrolysis in 2002;138:1019.
Europe and Israel. J Am Acad Dermatol. 2008; 678. Roujeau JC. Stevens-Johnson syndrome and toxic
58:25. epidermal necrolysis are severity variants of the
662. Pluim HJ, van Deuren M, Wetzels JF. The allopurinol same disease which differs from erythema multi-
hypersensitivity syndrome. Neth J Med. 1998;52:107. forme. J Dermatol. 1997;24:726.
References 375

679. Bastuji-Garin S, Rzany B, Stern RS, et al. Clinical 698. Turnheim K, Krivanek P, Oberbauer R.
classification of cases of toxic epidermal necrolysis, Pharmacokinetics and pharmacodynamics of
Stevens-Johnson syndrome, and erythema multi- allopurinol in elderly and young subjects. Br J Clin
forme. Arch Dermatol. 1993;129:92. Pharmacol. 1999;48:501.
680. Ruiz-Maldonado R. Acute disseminated epidermal 699. Perez-Ruiz F, Hernando I, Villar I, Nolla JM.
necrosis types 1, 2, and 3: a study of sixty cases. J Correction of allopurinol dosing should be based on
Am Acad Dermatol. 1985;13:623. clearance of creatinine, but not plasma creatinine
681. Stevens AM, Johnson FC. A new eruptive fever levels: Another insight to allopurinol-related toxic-
associated with stomatitis and ophthalmia: report of ity. J Clin Rheumatol. 2005;11:129.
two cases in children. Am J Dis Child. 1922; 700. Cockcroft DW, Gault MH. Prediction of creatinine
24:526. clearance from serum creatinine. Nephron. 1976;
682. Lyell A. Toxic epidermal necrolysis: an eruption 16:31.
resembling scalding of the skin. Br J Dermatol. 701. Hande KR, Noone RM, Stone WJ. Severe allopurinol
1956;68:355. toxicity. Description and guidelines for prevention in
683. Roupe G, Ahlmen M, Fagerberg B, Suurkula M. patients with renal insufficiency. Am J Med. 1984;
Toxic epidermal necrolysis with extensive mucosal 76:47.
erosions of the gastrointestinal and respiratory tracts. 702. Young Jr JL, Boswell RB, Nies AS. Severe
Int Arch Allergy Appl Immunol. 1986;80:145. allopurinol hypersensitivity-associated with thiaz-
684. Chosidow O, Delchier JC, Chaumette MT, et al. ides and prior renal compromise. Arch Intern Med.
Intestinal involvement in drug-induced toxic epider- 1974;134:553.
mal necrolysis. Lancet. 1991;337:928. 703. Loffler W, Landthaler R, de Vries JX, et al.
685. Timsit JF, Mion G, Rouyer N, et al. Bronchopulmonary Interaction of allopurinol and hydrochlorothiazide
distress associated with toxic epidermal necrolysis. during prolonged oral administration of both drugs
Intensive Care Med. 1992;18:42. in normal subjects. I. Uric acid and kinetics. Clin
686. Roujeau JC. Immune mechanisms in drug allergy. Investig. 1994;72:1076.
Allergol Int. 2006;55:27. 704. Hande KR. Evaluation of a thiazide-allopurinol drug
687. Hari Y, Frutig-Schnyder K, Hurni M, et al. T cell interaction. Am J Med Sci. 1986;292:213.
involvement in cutaneous drug eruptions. Clin Exp 705. Breithaupt B, Tittel M. Kinetics of allopurinol after
Allergy. 2001;31:1398. single intravenous and oral doses. Nonintervention
688. Braden GL, Warzynski MJ, Golightly M, Ballow M. with benzbromarone and hydrochlorothiazide. Eur J
Cell-mediated immunity in allopurinol-induced Clin Pharmacol. 1982;22:77.
hypersensitivity. Clin Immunol Immunopathol. 1994; 706. Mills RM. Severe hypersensitivity reactions associ-
70:145. ated with allopurinol. JAMA. 1971;216:799.
689. Pirmohamed M. Genetic factors in the predisposi- 707. Gelbort DR, Weinstein AB, Fajardo LF. Allopurinol-
tion to drug-induced hypersensitivity reactions. induced interstitial nephritis. Ann Intern Med.
AAPS J. 2006;8:E20. 1977;86:197.
690. Melson RD. Familial hypersensitivity to allopurinol 708. Jarzobaki J, Ferry J, Wombelt D, et al. Vasculitis
with subsequent desensitization. Rheumatology. with allopurinol therapy. Am Heart J. 1970;79:116.
1999;38:1301. 709. Grussendorf M, Andrassy K, Waldhern R, Ritz H.
691. Edwards SG, Hubbard V, Aylett S, Wren D. Systemic hypersensitivity to allopurinol with acute
Concordance of primary generalized epilepsy and interstitial nephritis. Am J Nephrol. 1981;
CBZ hypersensitivity in monozygotic twins. 1:105.
Postgrad Med J. 1999;75:680. 710. Zanni MP, von Greyerz S, Schnyder B, et al. HLA-
692. Gennis MA, Vemuri R, Burns EA, et al. Familial restricted, processing- and metabolism-independent
occurrence of hypersensitivity to phenytoin. Am J pathway of drug recognition by human ab T lym-
Med. 1991;91:631. phocytes. J Clin Invest. 1998;102:1591.
693. Johnson-Reagan L, Bahna SL. Severe drug rashes in 711. Roujeau JC, Bracq C, Huyn NT, et al. HLA pheno-
3 siblings simultaneously. Allergy. 2003;58:445. types and bullous cutaneous reactions to drugs.
694. Pellicano R, Selvestris A, Immantriono M, et al. Tissue Antigens. 1986;28:251.
Familial occurrence of fixed drug eruptions. Acta 712. Roujeau JC, Huynh TN, Bracq C, et al. Genetic sus-
Derm Venereol. 1992;72:292. ceptibility to toxic epidermal necrolysis. Arch
695. Peyrierre H, Nicolas J, Seffert M, et al. Dermatol. 1987;123:1171.
Hypersensitivity related to abacavir in 2 members of 713. Shirato S, Kagaya F, Suzuki Y, Joukou S. Stevens-
a family. Ann Pharmacother. 2001;35:1291. Johnson syndrome induced by methazolamide treat-
696. Peyriere H, Dereure O, Breton H, et al. Variability in ment. Arch Ophthalmol. 1997;115:550.
the clinical pattern of cutaneous side-effects with 714. Chan SH, Tan T. HLA and allopurinol drug eruption.
systemic symptoms: does a DRESS syndrome really Dermatologica. 1989;179:32.
exist? Br J Dermatol. 2006;155:422. 715. Chung WH, Hung SI, Hong HS, et al. Medical genet-
697. Cameron JS, Simmonds HA. Use and abuse of ics: a marker for Stevens-Johnson syndrome. Nature.
allopurinol. Br Med J. 1987;294:1504. 2004;428:486.
376 9 Management of Hyperuricemia and Gout

716. Mallal S, Nolan D, Witt C, et al. Association between structure of the enzyme-inhibitor complex and mech-
presence of HLA-B*5701, HLA-DR7, and HLA- anism of inhibition. J Biol Chem. 2003;278:1848.
DQ3 and hypersensitivity to HIV-1 reverse tran- 731. Chatham WW, Saag KG. Is Febuxostat a more effec-
scriptase inhibitor Abacavir. Lancet. 2002;359:727. tive treatment for hyperuricemia and gout than
717. Martin AM, Nolan D, Gaudieri S, et al. Predisposition allopurinol? Nat Clin Pract Rheumatol. 2006;2:240.
to Abacavir hypersensitivity conferred by HLA- 732. Gelber AC. Febuxostat versus allopurinol for gout.
B*5701 and a haplotype Hsp 70-Hom variant. Proc N Engl J Med. 2006;354:1532.
Natl Acad Sci USA. 2004;101:4180. 733. Lustberg ME. Febuxostat versus allopurinol for
718. Chessman D, Kostenko L, Lethborg T, et al. Human gout. N Engl J Med. 2006;354:1532.
leukocyte antigen class 1-restricted activation of CD8+ 734. Okamoto K, Nishino T. Crystal structures of mam-
T cells provides the immunogenetic basis of a sys- malian xanthine oxidoreductase bound with various
temic drug hypersensitivity. Immunity. 2008;28:822. inhibitors: allopurinol, Febuxostat, and FYX-051. J
719. Schlosstein L, Terasaki PI, Bluestone R, Pearson CM. Nippon Med Sch. 2008;75:2.
High association of an HL-A antigen, W27, with 735. Ziegler R. Febuxostat versus allopurinol for hyperu-
ankylosing spondylitis. N Engl J Med. 1973;288:704. ricemia. Med Monatsschr Pharm. 2006;29:384.
720. Brewerton DA, Hart FD, Nicholls A, et al. Ankylosing 736. Becker MA, Schumacher Jr HR, Wortmann RL,
spondylitis and HL-A27. Lancet. 1973;1:904. et al. Febuxostat compared with allopurinol in
721. Geer L, Terasaki PI, Gjertson DW. HLA frequency. patients with hyperuricemia and gout. N Engl J Med.
In: Gjertson DW, Terasaki PI, editors. HLA. Mt. 2005;353:2450.
Laurel: American Society of Histocompatibility 737. Becker MA, Kisicki J, Khosravan R, et al. Febuxostat
Immunogenetics; 1998. p. 352–3. (TMX-67), a novel, non-purine, selective inhibitor
722. Lonjou C, Borot N, Sekula P, et al. A European study of xanthine oxidase, is safe and decreases serum
of HLA-B in Stevens-Johnson syndrome and toxic urate in healthy volunteers. Nucleosides Nucleotides
epidermal necrolysis related to five high-risk drugs. Nucleic Acids. 2004;23:1111.
Pharmacogenet Genomics. 2008;18:99. 738. Mayer MD, Khosravan R, Verniller L, et al.
723. Schneck J, Fagot JP, Sekula P, et al. Effects of treat- Pharmacokinetics and pharmacodynamics of
ments on the mortality of Stevens-Johnson syndrome Febuxostat, a new non-purine selective inhibitor of
and toxic epidermal necrolysis: a retrospective study xanthine oxidase, in subjects with renal impairment.
of patients included in the prospective study Am J Ther. 2005;12:22.
EuroSCAR study. J Am Acad Dermatol. 2008;58:33. 739. Swan S, Khosravan R, Mayer MD, et al. Effect of
724. Horiuchi H, Ota M, Kobayashi M, et al. A compara- renal impairment on pharmacokinetics, pharmaco-
tive study on the hypouricemic activity and potency dynamics, and safety of Febuxostat (TMX-67), a
in renal xanthine calculus formation of two xanthine novel non-purine selective inhibitor of xanthine oxi-
oxidase/xanthine dehydrogenase inhibitors: TEI- dase. Arthritis Rheum. 2003;48 Suppl 9:SS29.
6720 and allopurinol in rats. Res Commun Mol 740. Becker MA, Schumacher HR, Wortmann RI, et al.
Pathol Pharmacol. 1999;104:293. Allopurinol intolerant patients treated with febuxo-
725. Osada Y, Tsuchimoto M, Fakushima H, et al. stat for 4 years. Arthritis Rheum. 2008;54:S646.
Hypouricemic effect of a novel xanthine oxidase 741. Takano Y, Hase-Aoki K, Horiuchi H, et al. Selectivity
inhibitor, TEI-6720, in rodents. Eur J Pharmacol. of febuxostat, a novel non-purine inhibitor of xan-
1993;241:183. thine oxidase. Life Sci. 2005;76:1835.
726. Ishibuchi S, Morimoto H, Oe T, et al. Synthesis and 742. Goldstein M, Anderson LT, Reuben R, Dancis J.
structure activity relationships of 1-phenylpyrazoles Self-mutilation in Lesch-Nyhan disease is caused by
as xanthine oxidase inhibitors. Bioorg Med Chem dopaminergic denervation. Lancet. 1985;1:338.
Lett. 2001;11:879. 743. Pacher P, Nivorozhkin A, Szabo C. Therapeutic
727. Hashimoto T, Fukumari A, Yamada I, et al. Y-700, a effects of xanthine oxidase inhibitors: renaissance
novel inhibitor of xanthine oxidase, suppresses the half a century after the discovery of allopurinol.
development of colon aberrant crypt foci and cell Pharmacol Rev. 2006;58:87.
proliferation in 1, 2-dimethylhyrazine-treated mice. 744. Hitchings GH, Elion GB. Chemical suppression of
Biosci Biotechnol Biochem. 2005;69:209. the immune response. Pharmacol Rev. 1963;15:365.
728. Yamada I, Fukumari A, Osajima T, et al. Pharmaco- 745. Elion GB. The purine path to chemotherapy. Nobel
kinetics/pharmacodynamics of Y-700, a novel xan- Lecture. Research Triangle Park: Burroughs
thine oxidase inhibitor, in rats and man. Nucleosides Wellcome, Co.; 1988.
Nucleotides Nucleic Acids. 2004;23:1123. 746. Elion GB. The quest for a cure. Annu Rev Pharmacol
729. Fukumari A, Okamoto K, Nishino T, et al. Y-700 Toxicol. 1993;33:1.
[1-[3-cyano-4-(2,2-dimethylpropxy)phenyl]-1H- 747. Pea F. Pharmacology of drugs for hyperuricemia:
pyrazole-4-carboxylic acid]: a potent xanthine oxi- mechanisms, kinetics and interactions. Contrib
doreductase inhibitor with hepatic excretion. J Nephrol. 2005;147:35.
Pharmacol Exp Ther. 2004;311:519. 748. Terkeltaub R, Bushinsky DA, Becker MA. Recent
730. Okamoto K, Eger B, Nishino T, et al. An extremely developments in our understanding of the renal
potent inhibitor of xanthine oxidoreductase. Crystal basis of hyperuricemia and the development of
References 377

novel antihyperuricemic therapeutics. Arthritis Res 764. Tapp RJ, Shaw JE, de Courten MP, et al. Foot com-
Ther. 2006;8 Suppl 1:S4. plications in Type 2 diabetes: an Australian popula-
749. Becker MA, Kisicki J, Khosravan R, et al. Febuxostat tion-based study. Diabet Med. 2003;20:105.
(TMX-67), a novel, non-purine, selective inhibitor 765. Newman EJ, Rahman FS, Lees KR, et al. Elevated
of xanthine oxidase, is safe and decreases serum serum urate concentration independently predicts
urate in healthy volunteers. Nucleosides Nucleotides poor outcome following stroke in patients with dia-
Nucleic Acids. 2004;23:1111. betes. Diabetes Metab Res Rev. 2006;22:79.
750. Schumacher jr HR, Becker MA, Lloyd E, et al. 766. Verdecchia P, Schillaci G, Reboldi G, et al.
Febuxostat in the treatment of gout. Rheumatology. Relationship between serum uric acid and risk of
2009;48:188. cardiovascular disease in essential hypertension. The
751. Schumacher Jr HR, Becker MA, Wortmann HR, PIUMA Study. Hypertension. 2000;36:1072.
et al. Effects of febuxostat versus allopurinol and 767. Weir CI, Muir SW, Walters MR, Lee KR. Serum urate
placebo in reducing serum urate. Arthritis Care Res. as an independent predictor of poor outcome and vas-
2008;59:1540. cular events after acute stroke. Stroke. 2003;34:1951.
752. Schumacher Jr HR. Febuxostat: a non-purine, selec- 768. Frause LV, Pahor M, DiBari M, Shorr RI, et al. Serum
tive inhibitor of xanthine oxidase for the manage- uric acid, diuretic treatment and risk of cardiovascu-
ment hyperuricemia in patients with gout. Expert lar events in the systolic hypertension in the elderly
Opin Investig Drugs. 2005;14:893. prgramme (SHEP). J Hypertens. 2000;188:1149.
753. Khosravan R, Wu JT, Joseph-Ridge N, Vernillet L. 769. DeLeeuw PW, Thijs L, Birkenhager WH, et al.
Pharmacokinetic interactions of concomitant admin- Prognostic of renal function in elderly patients with
istration of Febuxostat and NSAIDS. J Clin isolated systolic hypertension: results from the Syst-
Pharmacol. 2006;46:855. Eur trial. J Am Soc Nephrol. 2002;13:2213.
754. Sanchez-Lozada LG, Tapia E, Bautista-Garcia P, et al. 770. Alderman MH, Cohen H, Madhavean S, Kivlighn S.
Effects of Febuxostat on metabolic and renal altera- Serum uric acid and cardiovascular events in suc-
tions in rats with fructose-induced metabolic syn- cessfully treated hypertensive patients. Hypertension.
drome. Am J Physiol Renal Physiol. 2008;294:F710. 1999;34:144.
755. Sanchez-Lozada LG, Tapia E, Jiminez A, et al. 771. Wang JG, Staessen JA, Faggard RH, et al. Prognostic
Fructose-induced metabolic syndrome is associated significance of serum creatinine and uric acid in
with glomerular hypertension and renal microvascu- older Chinese patients with isolated systolic hyper-
lar damage in rats. Am J Physiol Renal Physiol. tension. Hypertension. 2001;37:1069.
2007;292:F423. 772. Conger JD. Acute uric acid nephropathy. Semin
756. Sanchez-Lozada LG, Tapia E, Soto V, et al. Treatment Nephrol. 1981;1:69.
with the xanthine oxidase inhibitor, Febuxostat, low- 773. Crittenden DR, Ackerman GL. Hyperuricemic acute
ers uric acid and alleviates systemic and glomerular renal failure in disseminated carcinoma. Arch Intern
hypertension in experimental hyperuricaemia. Med. 1977;137:97.
Nephrol Dial Transplant. 2008;23:1179. 774. Stuck KJ, White GM, Granke DS, et al. Urinary
757. Wu XW, Lee CC, Muzny DM, et al. Urate oxidase obstruction in azotemic patients: detection by sonog-
primary structure and evolutionary implications. raphy. AJR Am J Roentgenol. 1987;149:1191.
Proc Natl Acac Sci USA. 1989;86:9412. 775. Coleman BG. Ultrasound of the upper genitourinary
758. Baker JF, Krishnan E, Chen L, Schumacher HR. tract. Urol Clin North Am. 1985;12:633.
Recent developments, and where do they leave us? 776. Rao KG, Hackler RH, Woodlief RM, et al. Real-time
Am J Med. 2005;118:816. renal sonography in spinal cord injury patients: pre-
759. Bickel C, Rupprecht HJ, Blankenberg S, et al. Serum spective comparison with excretory urography. J
uric acid as an independent predictor of mortality in Urol. 1986;135:72.
patients with angiographically proven coronary 777. Kaye AD, Pollack HM. Diagnostic imaging approach
artery disease. Am J Cardiol. 2002;89:12. to the patient with obstructive uropathy. Semin
760. Lehto S, Niskanen L, Ronnemaa T, Laakso M. Nephrol. 1982;2:55.
Serum uric acid is a strong predictor of stroke in 778. Bosniak MA, Megibow AJ, Ambos MA, et al.
patients with non-insulin dependent diabetes melli- Computed tomography of ureteral obstruction. AJR
tus. Stroke. 1998;29:635. Am J Roentgenol. 1982;138:1107.
761. Madsen TE, Muhlestein JB, Carlquist JF, et al. 779. Maker JF, Rath CE, Schreiner GE. Hyperuricemia
Serum uric acid independently predicts mortality in complicating leukemia. Treatment with allopurinol
patients with significant, angiographically defined and dialysis. Arch Intern Med. 1969;123:198.
coronary disease. Am J Nephrol. 2005;25:45. 780. Knochel JP, Mason AD. Effect of alkalinization on
762. Tseng CH. Independent association of uric acid levels peritoneal diffusion of uric acid. Am J Physiol.
with peripheral arterial disease in Taiwanese patients 1980;210:1160.
with type 2 diabetes. Diabet Med. 2004;21:724. 781. Morino M, Shiigai N, Kusuyama H, Okada K.
763. Cherubini A, Polidori MC, Bregnocchi M, et al. Extracorporeal shock wave lithotripsy and xanthine
Antioxidant profile and early outcome in stroke calculi in Lesch-Nyhan syndrome. Pediatr Radiol.
patients. Stroke. 2003;31:2295. 1992;22:304.
378 9 Management of Hyperuricemia and Gout

782. Kranen S, Keough D, Gordon RB, Emmerson BT. 799. Perez-Ruiz F, Liote F. Lowering serum uric acid lev-
Xanthine-containing calculi during allopurinol ther- els: what is the optimal target for improving clinical
apy. J Urol. 1985;133:658. outcomes in gout? Arthritis Rheum. 2007;57:1324.
783. Oka T, Utsunomiya M, Ichikawa Y, et al. Xanthine 800. Sundy JS, Becker MA, Schumacher HR, et al.
calculi in the patient with Lesch-Nyhan syndrome Multicenter longitudinal study of disease character-
associated with urinary tract infection. Urol Int. istics in patients with treatment failure gout. Ann
1985;40:138. Rheum Dis. 2006;65:272.
784. Morton WJ. Lesch-Nyhan syndrome. Urology. 1982; 801. Fels E, Sundy JS. Refractory gout: what is it and
20:506. what do we do about it? Curr Opin Rheumatol. 2008;
785. Brock WA, Golden J, Kaplan GW. Xanthine calculi in 20:198.
the Lesch-Nyhan syndrome. J Urol. 1983;130:157. 802. Sundy JS, Schumacher Jr HR, Wortmann RL, et al.
786. Howell RR. The interrelationship of glycogen stor- Quality of life in patients with treatment failure gout.
age disease and gout. Arthritis Rheum. 1965;8:780. Ann Rheum Dis. 2006;65:271.
787. Chen Y-T, Scheinman JI, Park HK, et al. Amelioration 803. Geletka RC, Hershfield MS, Scarlett EL, Sundy JS.
of proximal renal tubular dysfunction in Type I gly- Severe gout is associated with impaired quality of
cogen storage disease with dietary therapy. N Engl J life and functional status. Am Coll Rheumatol, 68th
Med. 1960;323:590. annual meeting, 16–21 Oct 2004.
788. Greene HL, Slonim AE, O’Neill Jr JA, Burr IM. 804. Pal B, Foxtall M, Dysort T, et al. How is gout man-
Continuous nocturnal intragastric feeding for the aged in primary care? A review of current practice and
management of type I glycogen storage disease. N proposed guidelines. Clin Rheumatol. 2000;19:21.
Engl J Med. 1976;294:423. 805. de Klerk E, van Der Heijde D, Landewe R, et al.
789. Chen Y-T, Cornblath M, Sidbury JB. Cornstarch Patient compliance in rheumatoid arthritis, polymyal-
therapy of Type I glycogen-storage disease. N Engl J gia rheumatica, and gout. J Rheumatol. 2003;30:44.
Med. 1984;310:171. 806. Riedel AA, Nelson M, Joseph-Ridge N, et al.
790. Wyke RJ, Rajkovic IA, Eddleston ALWF, Williams Compliance with allopurinol therapy among managed
R. Defective opsonization and complement care enrollees with gout: a retrospective analysis of
deficiency in serum from patients with fulminant administrative claims. J Rheumatol. 2004;31:1575.
hepatic failure. Gut. 1980;21:643. 807. Dalbeth N, Kumar S, Stamp L, Gow P. Dose adjust-
791. Cornblath M, Schwartz R. Disorders of carbohydrate ment of allopurinol according to creatinine clearance
metabolism in infancy. In: Major Problems in Clinical does not provide adequate control of hyperuricemia
Pediatrics, vol. 3. 2nd ed. Philadelphia: Saunders; in patients with gout. J Rheumatol. 2006;33:1646.
1976. 808. Vazquez-Mellado J, Morales EM, Pacheco-Tena C,
792. Odievre M, Gautier M, Rieu D. Intolerance heredi- Burgos-Vargas R. Relation between adverse events
taire au fructose du nourrisson. Evolution des lesions associated with allopurinol and renal function in
histologiques hepatiques sous traitment dietetique patients with gout. Ann Rheum Dis. 2001;60:981.
prolonge (Etude de huit observations). Arch Fr 809. Sundy JS, Ganson NJ, Kelly SJ, et al.
Pediatr. 1969;26:433. Pharmacokinetics and pharmacodynamics of intra-
793. Hardinge MG, Swarner JB, Crooks H. Carbohydrate venous PEGylated recombinant mammalian urate
in foods. J Am Diet Assoc. 1965;46:197. oxidase in patients with refractory gout. Arthritis
794. Somogyi JC, Trautner K. Der glucose-, fructose- und Rheum. 2007;56:1021.
saccharosegehalt verschiedener gemusearten. 810. Vogt B. Urate oxidase (Rasburicase) for treatment of
Schweiz Med Wochenschr. 1974;104:177. severe tophaceous gout. Nephrol Dial Transplant.
795. Mock DM, Perman JA, Thaler MM, Morris RC. 2005;20:431.
Chronic fructose intoxication after infancy in chil- 811. Moolenburgh JD, Reinders MK, Jansen TL.
dren with hereditary fructose intolerance: A cause of Rasburicase treatment in severe tophaceous gout: a
growth retardation. N Engl J Med. 1983;309:764. novel therapeutic option. Clin Rheumatol.
796. Brauman J, Kentos P, Frisque P, et al. Intolerance to 2006;25:749.
hereditaire au fructose chez une femme de 83 ans. 812. Hepburn AL, Kaye SA, Feher MD. Long-term
Acta Clin Belg. 1971;26:65. remission from gout associated with fenofibrate
797. Gitzelmann R, Steinmann B, van den Berghe G. therapy. Clin Rheumatol. 2003;22:73.
Disorders of fructose metabolism. In: Scriver CR, 813. Takahashi S, Moriwaki Y, Yamamoto T, et al. Effects
Beaudet AL, Sly WE, Valle D, editors. The meta- of combination treatment using antihyperuricaemic
bolic basis of inherited disease. New York: McGraw- agents with fenofibrate and/or losartan on uric acid
Hill, Inc; 1989. p. 399–424. metabolism. Ann Rheum Dis. 2003;62:572.
798. Perez-Ruiz F, Alonso-Ruiz A, Calabozo M, et al. 814. Heburn AL, Kaye SA, Feher MD. Fenofibrate: a new
Efficacy of allopurinol and benzbromarone for the treatment for hyperuricaemia and gout? Ann Rheum
control of hyperuricemia: a pathogenic approach to Dis. 2001;60:984.
the treatment of primary chronic gout. Ann Rheum 815. Bastow MD, Durrington PN, Ishola M.
Dis. 1998;57:545. Hypertriglyceridaemia and hyperuricaemia: effects
References 379

of two fibric acid derivatives (bezafibrate and 831. Feher MD, Casloke M, Foxton J, et al. Atherogenic
fenofibrate) in a double-blind, placebo-controlled lipoprotein phenotype in type 2 diabetes: reversal
trial. Metabolism. 1998;37:217. with fenofibrate. Diabetes Metab Res Rev.
816. Navolanic PM, Pui CH, Larson RA, et al. Elitek – 1999;15:395.
rasburicase: an effective means to prevent and treat 832. Emmerson B. Hyperlipidaemia in hyperuricaemia
hyperuricemia associated with tumor lysis syn- and gout. Ann Rheum Dis. 1998;57:509.
drome. Leukemia. 2003;17:499. 833. Feldman EB, Wallace SL. Hypertriglyceridemia in
817. McDonnell AM, Lenz KL, Frei-Lahr DA, et al. gout. Circulation. 1964;29:508.
Single-dose rasburicase 6 mg in the management of 834. Fox IH, John D, DeBruyne S, et al. Hyperuricaemia
tumor lysis syndrome in adults. Pharmacotherapy. and hypertriglyceridaemia: metabolic basis for the
2006;26:806. association. Metabolism. 1985;34:741.
818. de Bont JM, Pieters R. Management of hyperurice- 835. Berkowitz D. Blood lipid and uric acid interrelation-
mia with rasburicase review. Nucleosides Nucleotides ships. JAMA. 1964;190:856.
Nucleic Acids. 2004;23:1431. 836. Feher MD, Hepburn AL, Hogarth MB, et al.
819. Gabison L, Chiadris M, Colloch N, et al. Recapture Fenofibrate enhances urate reduction in men treated
of [S]-allantoin, the product of uric acid, by urate with allopurinol for hyperuricaemia and gout.
oxidase. FEBS Lett. 2006;580:2087. Rheumatology. 2003;42:321.
820. Brogard JM, Coumaros D, Franckhauser J, et al. 837. Hepburn AL, Kaye SA, Feher MD. Fenofibrate: a
Enzymatic uricolysis: a study of the effect of a fun- new treatment for hyperuricaemia and gout? Ann
gal urate-oxidase. Rev Eur Etud Clin Biol. 1972; Rheum Dis. 2001;60:984.
17:890. 838. Lee YH, Lee CH, Lee J. Effect of fenofibrate in com-
821. Legoux R, Delpech B, Dumont X, et al. Cloning and bination with urate lowering agents in patients wth
expression in Escherichia coli of the gene encoding gout. Korean J Intern Med. 2006;21:89.
Aspergillus flavus urate oxidase. J Biol Chem. 839. Yamamoto T, Moriwaki Y, Takahashi S, et al. Effect
1992;267:8565. of fenofibrate on plasma concentration and urinary
822. Pui CH, Malmoud HH, Wiley JM, et al. Recombinant excretion of purine bases and oxypurinol. J
urate oxidase for the prophylaxis or treatment of Rheumatol. 2001;28:2294.
hyperuricemia in patients with leukemia or lym- 840. Naritomi H, Fujita T, Ito S, et al. Efficacy and safety
phoma. J Clin Oncol. 2001;19:697. of long-term losartan therapy demonstrated by a pro-
823. Goldman SC, Holeenberg JS, Finklestein JZ, et al. A spective observational study in Japanese patients
randomized comparison between rasburicase and with hypertension. The Japanese Hypertension
allopurinol in children with lymphoma and leukemia Evaluation with Angiotensin II Antagonist Losartan
at high risk for tumor lysis. Blood. 2001;97:2998. Therapy (J-HEALTH) Study. Hypertens Res.
824. Trujillo M, Morales M. Rasburicase-induced fatal 2008;31:295.
respiratory arrest? Ann Oncol. 2007;18:399. 841. Ito O, Hasegawa Y, Sato K, et al. A case of exercise-
825. Jeha S, Kantarjian H, Irwin D, et al. Efficacy and induced acute renal failure in a patient with idio-
safety of rasburicase, a recombinant urate oxidase pathic renal hypouricemia developed during
(ElitekTM), in the management of malignancy-asso- antihypertensive therapy with losartan and trichlo-
ciated hyperuricemia in pediatric and adult patients, romethiazide. Hypertens Res. 2003;26:509.
final results of a multicenter compassionate use trial. 842. Murakami T, Kawakami H, Fukuda M, Furukawa S.
Leukemia. 2005;19:34. Patients with renal hypouricemia are prone to
826. Browning LA, Kruse JA. Hemolysis and methemo- develop acute renal failure – why? Clin Nephrol.
globinemia secondary to rasburicase administration. 1995;43:207.
Ann Pharmacother. 2005;39:1932. 843. Erley CM, Hirschberg RR, Hoefer W, Schaefer K.
827. Pui CH, Relling MV, Lascombes F, et al. Urate oxi- Acute renal failure due to uric acid nephropathy in a
dase in prevention and treatment of hyperuricemia patient with renal hypouricemia. Klin Wochenschr.
associated with lymphoid malignancies. Leukemia. 1989;67:308.
1997;11:1813. 844. Wurzner G, Gerster JC, Chiolero A, et al.
828. Keating GM, Ormrod D. Micronised fenofibrate: An Comparative effects of losartan and irbesartan on
updated review of its clinical efficacy in the manage- serum uric acid in hypertensive patients with hyper-
ment of dyslipidaemia. Drugs. 2002;62:1909. uricaemia and gout. J Hypertens. 2001;19:1855.
829. Desager JP, Hulloven R, Harvengt C. Uricosuric 845. Puig JG, Torres R, Ruilope LM. AT1 blockers and
effect of fenofibrate in healthy volunteers. J Clin uric acid metabolism: are there relevant differences?
Pharmacol. 1980;20:560. J Hypertens Suppl. 2002;20:529.
830. Elisaf M, Tsimichodimas V, Baeraktari E, 846. Rayner BL, Trinder YA, Baines D, et al. Effect of
Simopoulas KC. Effect of micronized fenofibrate losartan versus candesartan on uric acid, renal func-
and losartan combination on uric acid metabolism in tion, and fibrinogen in patients with hypertension
hypertensive patients with hyperuricaemia. J and hyperuricemia associated with diuretics. Am J
Cardiovasc Pharmacol. 1999;34:60. Hypertens. 2006;19:208.
380 9 Management of Hyperuricemia and Gout

847. Tsunoda K, Abe K, Hagino T, et al. Hypotensive 867. Choi HK, Atkinson K, Karlson EW, et al. Purine-
effect of losartan, a nonpeptide antiotensin II recep- rich foods, dairy and protein intakes, and the risk of
tor antagonist, in essential hypertension. Am J gout in men. N Engl J Med. 2004;350:1093.
Hypertens. 1993;6:28. 868. Snaith M. Gout and alcohol. Rheumatology.
848. Burnier M, Rutschmann B, Nussberger J, et al. Salt- 2004;43:1208.
dependent renal effects of an angiotensin II antago- 869. Rodnan GP. The pathogenesis of Aldermanic gout:
nist in healthy subjects. Hypertension. 1993;22:339. procatarctic role of fluctuations in serum urate con-
849. Nakashima M, Uematsu T, Kosuge K, et al. Pilot centrations in gouty arthritis provoked by feast and
study of the uricosuric effect of DuP-753: a new alcohol. Arthritis Rheum. 1980;23:737.
angiotensin II receptor antagonist, in healthy sub- 870. Lieber CS, Jones DP, Losowsky MS, Davison CS.
jects. Eur J Clin Pharmacol. 1992;42:333. Interrelation of uric acid and ethanol metabolism in
850. Flaherty JT, Edelman J, Kivlighn S. Effects of a low man. J Clin Invest. 1963;41:1863.
dose of thiazide diuretic on serum uric acid: compari- 871. Lin JL, Huang PT. Blood lead stores and urate excre-
son of a losartan-based regimen vs usual care. 71st tion in men with chronic renal disease. J Rheumatol.
scientific session, AHA, Circulation. 1999;I-723. 1994;21:705.
851. Soffer BA, Wright JT, Pratt JH, et al. Effects of 872. Gibson T, Rodgers AV, Simmonds HA, et al. A con-
losartan on a background of hydrochlorothiazide in trolled study of diet in patients with gout. Ann
patients with hypertension. Hypertension. 1995; Rheum Dis. 1983;42:123.
26:112. 873. Gibson T, Rodgers AV, Simmonds HA, Toseland P.
852. Kamper AL, Nielsen AH. Uricosuric effect of losar- Beer drinking and its effect on uric acid. Br J
tan in patients with renal transplants. Transplantation. Rheumatol. 1984;23:203.
2001;72:671. 874. Choi HK, Atkinson K, Karlson EW, et al. Alcohol
853. Ka T, Inokuchi T, Tsutsumi Z, et al. Effects of intake and risk of incident gout in men: a prospective
fenofibrate/losartan combination on the plasma con- study. Lancet. 2004;363:1277.
centration and urinary excretion of purine bases. Int 875. Moriwaki Y, Ka T, Takahashi S, et al. Effect of beer
J Clin Pharmacol Ther. 2006;44:22. ingestion on the plasma concentrations of purine
854. Shohinfor S, Simpson RI, Carides AD, et al. Safety bases: one-month study. Nucleosides Nucleotides
of losartan in hypertensive patients with thiazide-in- Nucleic Acids. 2006;25:1083.
duced hyperuricemia. Kidney Int. 1999;56:1879. 876. Ka T, Moriwaki Y, Takahashi S, et al. Effects of
855. Alderman M, Alyer KJ. Uric acid: role in cardiovas- long-term beer ingestion on plasma concentrations
cular disease and effects of losartan. Curr Med Res and urinary excretion of purine bases. Horm Metab
Opin. 2004;20:369. Res. 2005;37:641.
856. Smith LH. The medical aspects of urolithiasis: an 877. Eastmond CJ, Garton M, Robins S, Riddock S. The
overview. J Urol. 1989;141:707. effects of alcoholic beverages on urate metabolism
857. Wilson DM. Clinical and laboratory approaches for in gout sufferers. Br J Rheumatol. 1995;34:756.
evaluation of nephrolithiasis. J Urol. 1989;141:770. 878. Rodgers AV, Gibson T, Simmonds HA, Toseland P.
858. Hosking DH, Erickson SB, van den Berg CJ, et al. The effect of beer ingestion on plasma and urine uric
The stone clinic effect in patients with idiopathic acid in gout and normouricaemic subjects. Adv Exp
calcium urolithiasis. J Urol. 1963;130:1115. Med Biol. 1984;165 Part A:327.
859. Burns JR, Finlayson B. Strategies for the medical 879. Tofler OB, Woodings TL. A 13-year follow-up of
management of patients with urinary stone disease. social drinkers. Med J Aust. 1981;2:41.
Monogr Urol. 1981;2:198. 880. Saker MB, Tofler OB, Burvill MJ, Reilly KA. Alcohol
860. Ettinger B. Does hyperuricosuria play a role in cal- consumption and gout. Med J Aust. 1967;1:1213.
cium oxalate lithiasis? J Urol. 1989;141:738. 881. Choi HK, Curhan G. Beer, liquor, and wine con-
861. Andres A, Praga M, Bello I, et al. Hematuria due to sumption and serum uric acid levels - The Third
hypercalciuria and hyperuricosuria in adult patients. National Health and Nutrition Examination Survey.
Kidney Int. 1989;36:96. Arthritis Rheum. 2004;50(Suppl):S480.
862. Zechner O, Pfluger H. Oral purine loading for evalu- 882. Choi HK, Lin S, Curhan G. Intake of purine-rich
ation of uric acid excretion in patients with urinary foods, protein and dairy products and relationship to
calculi. Invest Urol. 1980;18:115. serum levels of uric acid. The Third National Health
863. Zechner O, Pfluger H, Scheiber V. Idiopathic uric and Nutrition Examination Survey. Arthritis Rheum.
acid lithiasis: epidemiologic and metabolic aspects. J 2005;52:283.
Urol. 1982;128:1219. 883. Choi HK, Curhan G. Alcohol and gout. Am J Med.
864. Yu TF. Milestones in the treatment of gout. Am J 2007;120:e5.
Med. 1974;56:676. 884. Miao Z, Li C, Chen Y, et al. Dietary and lifestyle
865. Fam AG. Gout, diet and the insulin resistance syn- changes associated with high prevalence of hyperu-
drome. J Rheumatol. 2002;29:1350. ricemia and gout in the Shandong coastal cities of
866. Fam AG. Gout, excess calories, purines, and beyond. eastern China. J Rheumatol. 2008;35:1859.
Response to a urate-lowering diet. J Rheumatol. 885. Mineo I, Kamiya H, Tsukuda A. Practical strategies
2005;32:773. for lifestyle modification in people with hyperuricemia
References 381

and gout treatment through diet, physical activity, and 903. Williams PT. Effects of diet, physical activity and
reduced alcohol consumption. Nippon Rinsho. 2008; performance and body weight on incident gout in
66:736. ostensibly healthy, vigorously active men. Am J Clin
886. Friedman JE, Dallal RM, Lard JL. Gouty attacks Nutr. 2008;87:1480.
occur frequently in postoperative gastric bypass 904. Nan H, Qiao O, Soderberg S, et al. Serum uric acid
patients. Surg Obes Relat Dis. 2008;4:11. and components of the metabolic syndrome in non-
887. Rush Jr RM. Gout and the postoperative bariatric diabetic populations in Mauritian Indians and
surgery patient. Surg Obes Relat Dis. 2008;4:14. Creoles and in Chinese in Quingdao, China. Metab
888. Saag KG, Choi HK. Epidemiology, risk factors, and Syndr Relat Disord. 2008;6:47.
lifestyle modifications in gout. Arthritis Res Ther. 905. Chang HY, Pan WH, Yeh WT, Tsai KS.
2006;8 Suppl 1:S2. Hyperuricemia and gout in Taiwan: results from the
889. Schlesinger N. Dietary factors and hyperuricaemia. Nutritional and Health Survey in Taiwan (1993–96).
Curr Pharm Res. 2005;11:4133. J Rheumatol. 2001;28:1640.
890. Antozzi P, Soto F, Arias F, et al. Development of 906. No authors listed. Metabolic syndrome in gout.
acute gouty attack in the morbidly obese population Vestn Akad Med Nauk 2008;6:29.
after bariatric surgery. Obes Surg. 2005;15:405. 907. Muntner P, Srinvasan S, Menke A, et al. Impact of
891. Choi HK, Atkinson K, Karlson EW, Curhan G. childhood metabolic syndrome components on the
Obesity, weight change, hypertension, diuretic use, risk of elevated uric acid in adulthood: the Bogalusa
and risk of gout in men: the Health Professional’s Heart Study. Am J Med Sci. 2008;335:332.
Follow-up Study. Arch Intern Med. 2005;165:742. 908. Puig JG, Martinez MA. Hyperuricemia, gout and the
892. Tipene-Leach D, Pahau H, Joseph N, et al. Insulin metabolic syndrome. Curr Opin Rheumatol.
resistance in a rural Maori community. N Z Med J. 2008;20:187.
2004;117:U1208. 909. Virsaladze DK, Tetradze W, Dzhavashvili LV, et al.
893. Barskova VG, Eliseev MS, Nasonov EL, et al. Levels of uric acid in serum in patients with meta-
Insulin resistance syndrome in patients with gout bolic syndrome. Georgian Med News. 2007;146:35.
and its influence on formation of clinical characteris- 910. Novak S, Melkonian AK, Patel PA, et al. Metabolic
tics of the disease. Ter Arkh. 2004;76:51. syndrome-related conditions among people with and
894. Lyu LC, Hsu CY, Yeh CY, et al. A case–control study without gout: prevalence and resource use. Curr Med
of the association of diet and obesity with gout in Res Opin. 2007;23:623.
Taiwan. Am J Clin Nutr. 2003;78:690. 911. Choi HK, Ford ES, Li C, Curhan G. Prevalence of
895. Chen SY, Chen CL, Shen ML, Kamatani N. Trends in the metabolic syndrome in patients with gout: the
the manifestations of gout in Taiwan. Rheumatology. Third National Health and Nutrition Examination
2003;42:1529. Survey. Arthritis Rheum. 2007;57:109.
896. Takahashi S, Moriwaki Y, Tsutsumi Z, et al. Increased 912. Chen SY, Chen CL, Shen ML. Manifestations of
visceral fat accumulation further aggravates the risk metabolic syndrome associated with male gout in
of insulin resistance in gout. Metabolism. 2001; different age strata. Clin Rheumatol. 2007;26:1453.
50:393. 913. Vazez-Mellado J, Garcia CG, Vazquez SG, et al.
897. Dessein PH, Shipton EA, Stanwix AE, et al. Metabolic syndrome and ischemic heart disease in
Beneficial effects of weight loss associated with gout. J Clin Rheumatol. 2004;10:105.
moderate calorie/carbohydrate restriction, and 914. Korochina IE, Bagirova GG. Metabolic syndrome
increased proportional intake of protein and unsatu- and rheumatic diseases. Ter Arkh. 2006;78:39.
rated fat on serum urate and lipoprotein levels in 915. Rho YH, Choi SJ, Lee YH, et al. The prevalence of
gout: a pilot study. Ann Rheum Dis. 2000;59:539. metabolic syndrome in patients with gout: a multi-
898. Iwatani M, Wasada T, Iwamoto Y, Kamatani N. center study. J Korean Med Sci. 2005;20:1029.
Insulin sensitizer and urate metabolism. Nippon 916. Iannello S, Cavaliere G, Ferro G, et al. Tophaceous
Rinsho. 2000;58:439. gout in plurimetabolic syndrome. Minerva Med.
899. Vague J. The degree of masculine differentiation of 1998;89:419.
obesities: a factor determining predisposition to dia- 917. Gresser U, Gathof BS, Gross M. Benzbromarone
betes, atherosclerosis, gout, and uric calculus dis- and fenofibrate are lipid lowering and uricosuric: a
ease. Nutrition. 1999;15:89. possible key to metabolic syndrome? Adv Exp Med
900. Tikly M, Bellingan A, Lincoln D, Russell A. Risk Biol. 1994;370:87.
factors for gout: a hospital-based study in urban black 918. Katzmann K. The metabolic syndrome. Z Arztl
South Africans. Rev Rhum Engl Ed. 1998;65:225. Fortbild (Jena). 1991;85:651.
901. Takahashi S, Yamamoto T, Tsutsumi Z, et al. Close 919. Knappe G. Obesity – pacemaker of the metabolic
correlations between visceral fat accumulation and syndrome. Z Arztl Fortbild (Jena). 1992;86:891.
uric acid metabolism in healthy men. Metabolism. 920. Choi JW, Ford ES, Gao X, Choi HK. Sugar-
1997;46:1162. sweetened soft drinks, diet soft drinks, and serum
902. Takahashi S, Yamamoto T, Tsutsumi Z, et al. uric acid level: the Third National Health and
Increased visceral fat accumulation in patients with Nutrition Examination Survey. Arthritis Rheum.
primary gout. Adv Exp Med Biol. 2000;486:131. 2008;59:109.
382 9 Management of Hyperuricemia and Gout

921. Choi HK, Curhan G. Soft drinks, fructose consump- 937. Saito I, Saruta T, Kondo K, et al. Serum uric acid and
tion, and the risk of gout in men: perspective cohort the renin-angiotensin system in hypertension. J Am
study. BMJ. 2008;336:309. Geriatr Soc. 1978;26:241.
922. Messerli FH, Frohlich ED, Dreslinski GR, et al. 938. Zoccali C, Maio R, Mallamaci F, et al. Uric acid and
Serum uric acid in essential hypertension: an indica- endothelial dysfunction in essential hypertension. J
tor of renal vascular involvement. Ann Intern Med. Am Soc Nephrol. 2006;17:1466.
1980;93:817. 939. Doehner W, Schoene N, Rauchhaus M, et al. Effects
923. Chobanian AV, Bakris GL, Black HR, et al. The sev- of xanthine oxidase inhibition with allopurinol on
enth report of the Joint National Committee on pre- endothelial function and peripheral blood flow in
vention, detection, and treatment of high blood hyperuricemic patients with chronic heart failure:
pressure. The JNC 7 report. JAMA. 2003;289:2560. results from 2 placebo-controlled studies. Circulation.
924. Vaccarino V, Krumholz HM. Risk factors for cardio- 2002;105:2619.
vascular disease: one down, many more to evaluate. 940. Farquharson CA, Butler R, Hill A, et al. Allopurinol
Ann Intern Med. 1999;131:62. improves endothelial dysfunction in chronic heart
925. Alper Jr AB, Chen W, Yau L, et al. Childhood uric failure. Circulation. 2002;106:221.
acid predicts adult blood pressure: the Bogalusa 941. Mercure G, Vitale C, Cerquetani E, et al. Effect of
Heart Study. Hypertension. 2005;45:34. hyperuricemia upon endothelial function in patients
926. Mellen PB, Bleyer AJ, Erlinger TP, et al. Serum uric at increased cardiovascular risk. Am J Cardiol.
acid predicts incident hypertension in a biethnic 2004;94:932.
cohort: the Atherosclerosis Risk in Communities 942. Feig DL, Soletsky B, Johnson RJ. Effect of
Study. Hypertension. 2006;48:1037. allopurinol on blood pressure of adolescents with
927. Perlstein TS, Gumieniak O, Williams GH, et al. Uric newly diagnosed essential hypertension: a random-
acid and the development of hypertension: the ized trial. JAMA. 2008;300:924.
Normative Aging Study. Hypertension. 2006;48:1031. 943. Sanchez-Lozada LG, Tapia E, Soto V, et al. Treatment
928. Selby JV, Friedman GO, Quesenberry Jr CP. with the xanthine oxidase inhibitor: Febuxostat low-
Precursors of essential hypertension: pulmonary ers uric acid and alleviates systemic and glomerular
function, heart rate, uric acid, serum cholesterol, and hypertension in experimental hyperuricaemia.
other serum chemistries. Am J Epidemiol. 1990; Nephrol Dial Transplant. 2008;23:1179.
131:1017. 944. Sanchez-Lozada LG, Tapia E, Bautista-Garcia P,
929. Sundstrom J, Sullivan L, D’Agostino RB, et al. et al. Effects of Febuxostat on metabolic and renal
Relations of serum uric acid to longitudinal blood alterations in rats with fructose-induced metabolic
pressure tracking and hypertension incidence. syndrome. Am J Physiol Renal Physiol. 2008;
Hypertension. 2005;45:28. 294:F710.
930. Feig DL, Johnson RJ. Hyperuricemia in childhood 945. George J, Carr E, Davies J, et al. High-dose
primary hypertension. Hypertension. 2003;42:247. allopurinol improves endothelial function by pro-
931. Mazzali M, Hughes J, Kim YG, et al. Elevated uric foundly reducing oxidative stress and not by lower-
acid increases blood pressure in the rat by a novel ing uric acid. Circulation. 2006;114:2508.
crystal-independent mechanism. Hypertension. 2001; 946. Ishihara Y, Sekine M, Hatano A, Shimamoto N.
38:1101. Sustained contraction and endothelial dysfunction
932. Mazzali M, Kanellis J, Han L, et al. Hyperuricemia induced by reactive oxygen species in porcine coro-
induces a primary renal arteriolopathy in rats by a nary artery. Biol Pharm Bull. 2008;31:1667.
blood pressure-independent mechanism. Am J 947. Sanchez-Lozada LG, Soto V, Tapia E, et al. Role of
Physiol Renal Physiol. 2002;282:F991. oxidative stress in the renal abnormalities induced
933. Johnson RJ, Segal MS, Srinivas T, et al. Essential by experimental hyperuricemia. Am J Physiol Renal
hypertension, progressive renal disease, and uric Physiol. 2008;295:F1431.
acid: a pathogenetic link? J Am Soc Nephrol. 2005; 948. Gersch C, Palii SP, Kim KM, et al. Inactivation of
16:1909. nitric oxide by uric acid. Nucleosides Nucleotides
934. Sanchez-Lozada LG, Tapia E, Lopez-Molina R, Nucleic Acids. 2008;27:967.
et al. Effects of acute and chronic L-arginine treat- 949. Neaton JD, Grimm Jr RH, Prineas RJ, et al. Treatment
ment in experimental hyperuricemia. Am J Physiol of Mild Hypertension Study: final results. Treatment
Renal Physiol. 2007;292:F1238. of Mild Hypertension Study Research Group. JAMA.
935. Gruskin AB. The adolescent with essential hyperten- 1993;270(713):962.
sion. Am J Kidney Dis. 1985;6:86. 950. Ames BN, Cathcart R, Schwiers E, Hochstein P.
936. Erdogan D, Gullu H, Caliskan M, et al. Relationship Uric acid provides antioxidant defense in humans
of serum uric acid to measures of endothelial func- against oxidant- and radical-caused aging and can-
tion and atherosclerosis in healthy adults. Int J Clin cer: a hypothesis. Proc Natl Acad Sci USA. 1981;
Pract. 2005;59:1276. 78:6858.
References 383

951. Proctor P. Similar functions of uric acid and ascor- 968. Krishnan E, Svendsen K, Neaton JD, et al. MRFIT
bate in man? Nature. 1970;228:868. Research Group. Long-term cardiovascular mortal-
952. Johnson RJ, Gaucher EA, Sautin YY, et al. The plan- ity among middle-aged men with gout. Arch Intern
etary biology of ascorbate and uric acid and their Med. 2008;168:1104.
relationship with the epidemic of obesity and cardio- 969. Baker JF, Krishnan E, Chen L, Schumacher HR.
vascular disease. Med Hypotheses. 2008;71:22. Serum uric acid and cardiovascular disease: recent
953. Orawan E. The origin of man. Nature. 1955; developments, and where do they leave us? Am J
175:683. Med. 2005;118:816.
954. Corry DB, Eslami P, Yamamoto K, et al. Uric acid 970. Karagiannis A, Mikhailidis DP, Tzimalos K, et al.
stimulates vascular smooth muscle cell proliferation Serum uric acid as an independent prediction of
and oxidative stress via the vascular renin-angio- early death after acute stroke. Circ J. 2007;71:1120.
tensin system. J Hypertens. 2008;26:269. 971. Syamala S, Li J, Shankar A. Association between
955. Brown CM, Dulloo AG, Yepuri G, Montani JP. serum uric acid and prehypertension among US
Fructose ingestion acutely elevates blood pressure in adults. J Hypertens. 2007;25:1583.
healthy young humans. Am J Physiol Regul Integr 972. Viazzi F, Parodi D, Leoncini G, et al. Serum uric
Comp Physiol. 2008;294:R730. acid and target organ damage in primary hyperten-
956. Wexler BC, Greenberg BP. Effect of increased serum sion. Hypertension. 2005;45:991.
urate levels on virgin rats with no arteriosclerosis 973. Iwashima Y, Horio T, Kamide K, et al. Uric acid, left
versus breeder rats with preexisting arteriosclerosis. ventricular mass index, and risk of cardiovascular
Metabolism. 1977;26:1309. disease in essential hypertension. Hypertension.
957. Wexler BC. Allantoxamide-induced myocardial 2006;47:195.
necrosis in Sprague–Dawley vs spontaneously 974. Sanchez-Lozada LG, Tapia E, Santamaria J, et al.
hypertensive rats. Proc Soc Exp Biol Med. 1982; Mild hyperuricemia induces glomerular hyperten-
170:476. sion in normal and remnant kidney rats. Kidney Int.
958. Kang DH, Park SK, Lee IK, Johnson RJ. Uric acid- 2005;67:237.
induced C-reactive protein expression: implication on 975. Kang DH, Nakagawa T, Feng L, et al. A role for uric
cell proliferation and nitric oxide production of human acid in the progression of renal disease. J Am Soc
vascular cells. J Am Soc Nephrol. 2005;16:3553. Nephrol. 2002;13:2888.
959. Toma I, Kang JJ, Meer EJ, Peti-Peterdi J. Uric acid 976. Iseki K, Oshira S, Tozawa M, et al. Significance of
triggers renin release via a macula densa-dependent hyperuricemia on the early detection of renal failure
pathway. J Am Soc Nephrol. 2007;18:156A. in a cohort of screened subjects. Hypertens Res.
960. Khosla UM, Zharikov S, Finch JL, et al. 2001;24:691.
Hyperuricemia induces endothelial dysfunction. 977. Siu YP, Leung KT, Tong MK, et al. Use of allopurinol
Kidney Int. 2005;67:1739. in slowing the progression of renal disease through
961. Talaat KM, EI-Sheikh AR. The effect of mild hype- its ability to lower serum uric acid level. Am J
ruricemia on urinary transforming growth factor beta Kidney Dis. 2006;47:51.
and the progression of chronic kidney disease. Am J 978. Weimert NA, Tanke WF, Sims JJ. Allopurinol as a
Nephrol. 2007;27:435. cardioprotectant during coronary artery bypass graft
962. Masuo K, Kawaguchi H, Mikami H, et al. Serum surgery. Ann Pharmacother. 2003;37:1708.
uric acid and plasma norepinephrine concentrations 979. Butler R, Morris AD, Belch JJF, et al. Allopurinol
predict subsequent weight gain and blood pressure normalizes endothelial dysfunction in type 2 diabetics
elevation. Hypertension. 2003;42:474. with mild hypertension. Hypertension. 2000;35:746.
963. Ouyang X, Cerillo P, Sautin Y, et al. Fructose con- 980. Nakagawa T, Kang DH, Feig D, et al. Unearthing
sumption as a risk factor for non-alcohol fatty liver uric acid: an ancient factor with recently found
disease. J Hepatol. 2008;48:993. significance in renal and cardiovascular disease.
964. Perez-Pozo SE, Schold J, Nakagawa T, et al. Int J Kidney Int. 2006;69:1722.
Obesity. 2009;354. 981. Nieto FJ, Iribarren C, Gross MD, et al. Uric acid and
965. Feig DL, Soletsky B, Johnson RJ. Effect of serum antioxidant capacity: a reaction to atheroscle-
allopurinol on blood pressure of adolescents with rosis? Atherosclerosis. 2000;148:131.
newly diagnosed essential hypertension: a random- 982. Zweir JL, Kuppusamy P, Lutty GA. Measurement of
ized trial. JAMA. 2009;301:270. endothelial cell free radical generation: evidence for
966. Cannon PJ, Stason WB, Demartini FE, et al. a central mechanism of free radical injury in pos-
Hyperuricemia in primary and renal hypertension. N tischemic tissue. Proc Natl Acad Sci USA.
Engl J Med. 1966;275:457. 1988;85:4046.
967. Perlstein TS, Gumienieah O, Williams GH, et al. Uric 983. Strazzulo P, Puig JG. Uric acid and oxidative stress:
acid and the development of hypertension: the relative impact on cardiovascular risk? Nutr Metab
Normative Aging Study. Hypertension. 2006;48:1031. Cardiovasc Dis. 2007;17:409.
384 9 Management of Hyperuricemia and Gout

984. Roubenoff R, Klag MJ, Mead LA, et al. Incidence 1000. Yoo HG, Lee SL, Chase HJ, et al. Prevalence of
and risk factors for gout in white men. JAMA. 1991; insulin resistance and metabolic syndrome in patients
266:3004. with gouty arthritis. Rheumatol Int. 2011;31(4):
985. Chen SY, Shen ML. Juvenile gout in Taiwan associ- 485–91.
ated with family history and overweight. J Rheumatol. 1001. Hernandez-Cuevas CB, Roque LH, Huerta-Sil G,
2007;34:2308. et al. First acute attacks commonly precede features
986. Glynn RJ, Campion EW, Silbert JE. Trends in serum of the metabolic syndrome. J Clin Rheumatol.
uric acid levels 1961–1980. Arthritis Rheum. 1983; 2009;15:65.
26:87. 1002. Kuo CF, See LC, Luo SF, et al. Gout: an indepen-
987. Rathmann W, Funkhouser E, Dyer AR, Roseman dent risk factor for all-cause and cardiovascular
JM. Relations of hyperuricemia with the various mortality. Rheumatology. 2010;49:141.
components of the insulin resistance syndrome in 1003. Mitsuhashi H, Yatsuya H, Matsushita K, et al. Uric
young black and white adults: the Cardia Study. acid and left ventricular hypertrophy in Japanese
Coronary Artery Risk Development in Young Adults. men. Circ J. 2009;73:667.
Ann Epidemiol. 1998;8:250. 1004. Enomoto A, Kimura H, Chairoungdua A, et al.
988. Lee J, Sparrow D, Vokonas PS, et al. Uric acid and Molecular identification of a renal urate anion
coronary heart disease risk: evidence for a role of exchanger that regulates blood urate levels. Nature.
uric acid in the obesity-insulin resistance syndrome. 2002;417:447.
The Normative Aging Study. Am J Epidemiol. 1005. Bakker SJ, Gana RO, ter Maaten JC, et al. The
1995;142:288. potential role of adenosine in the pathophysiology
989. Yamashita S, Matsuzawa Y, Tokunaga K, et al. of the insulin resistance syndrome. Atherosclerosis.
Studies on the impaired metabolism of uric acid in 2001;155:283.
obese subjects: marked reduction of renal urate 1006. Fransen R, Koomans HA. Adenosine and renal
excretion and its improvement by a low-calorie diet. sodium handling: direct natriuresis and renal nerve-
Int J Obes. 1986;10:255. mediated antinatriuresis. J Am Soc Nephrol.
990. Third Report of the National Cholesterol Education 1995;6:1491.
Program (NCEP) Expert Panel on detection, educa- 1007. Balakrishnan VS, Coles GA, Williams JD. Effects
tion, and treatment of high blood cholesterol in of intravenous adenosine on renal function in healthy
adults {adult treatment panel 111) final report. human subjects. Am J Physiol. 1996;271:F374.
Circulation. 2002;106:3143. 1008. Balakrishnan VS, Coles GA, Williams JD. A role
991. Grundy SM, Brower Jr HB, Gleeman JL, et al. for endogenous adenosine in control of human
Definition of metabolic syndrome: report of the glomerular and tubular function. Am J Physiol.
National, Heart, Lung and Blood Institute/American 1993;265:F504.
Heart Association conference on scientific issues 1009. Puig JG, Ruilope LM. Uric acid as a cardiovascular
related to definition. Circulation. 2004;109:433. risk factor in arterial hypertension. J Hypertens.
992. Grundy SM, Gleeman JL, Daniels SR, et al. 1999;17:869.
Diagnosis and management of the metabolic syn- 1010. Lopez-Suarez A, Elvira-Gonzalez J, Bascunana-
drome: an American Heart Association/National Quirell A, et al. Serum urate levels and urinary uric
Heart, Lung, and Blood Institute Scientific Statement. acid excretion in subjects with the metabolic syn-
Circulation. 2005;112:2735, e297, e298. drome. Med Clin. 2006;126:321.
993. American Heart Association. Metabolic syndrome: 1011. Strazzullo P, Barbato A, Galletti F, et al.
new guidelines for prevention and treatment. Abnormalities of renal sodium handling in the met-
URL: http://www.americanheart.org/persenter. abolic syndrome: results of the Olivetti Heart Study.
jhtml?identifier=3033454. J Hypertens. 2006;24:1633.
994. Trevian M, Liu J, Bahsas FB, et al. Syndrome X and 1012. Emmerson BT. The management of gout. N Engl J
mortality: a population-based study. Am J Epidemiol. Med. 1996;334:445.
1998;148:958. 1013. Gibson T, Rodgers AV, Simmonds HA, et al. A con-
995. Ford ES. Risks for all-cause mortality, cardiovascu- trolled study of diet in patients with gout. Ann
lar disease, and diabetes associated with the meta- Rheum Dis. 1983;42:123.
bolic syndrome. Diabetes Care. 2005;28:1769. 1014. Snaith M. A (very) short history of diets for gout.
996. Choi HK, Curhan G. Independent impact of gout on Rheumatology. 2004;43:1054.
mortality and risk for coronary heart disease. 1015. Johnson RJ, Rideout BA. Uric acid and diet-
Circulation. 2007;116:894. insights into the epidemic of cardiovascular dis-
997. Krishnan E, Baker JF, Furst DE, Schumacher HR. ease. N Engl J Med. 2004;350:1071.
Gout and the risk of acute myocardial infarction. 1016. Sharpe CR. A case–control study of alcohol con-
Arthritis Rheum. 2006;54:2688. sumption and drinking behavior in patients with
998. Wannamethee SG. Serum uric acid and risk of coro- acute gout. Can Med Assoc J. 1984;131:563.
nary heart disease. Curr Pharm Dis. 2005;11:4125. 1017. Pascual E, Swera F. Why is gout so poorly man-
999. Hjortnaes J, Algra A, Olijhoek J, et al. Serum uric acid aged? Ann Rheum Dis. 2007;66:1269.
levels and risk for vascular disease in patients with the 1018. Roddy E, Zhang W, Doherty M. Concordance of the
metabolic syndrome. J Rheumatol. 2007;34:1882. management of chronic gout in a UK primary-care
References 385

population with the EULAR gout recommenda- 1033. Boulman N, Slobodin G, Sharif D, et al.
tions. Ann Rheum Dis. 2007;66:1311. Pseudopodagra: a manifestation of infective endo-
1019. Perez-Ruiz F, Pascual E, Carmona L, et al. carditis. Clin Exp Rheumatol. 2005;23:251.
Diagnosis of gout in the rheumatology, hospital 1034. Beaudet F, de Medicis R, Magny P, Lussier A.
based setting lies far from that recommended: Acute apatite podagra with negative birefringent
results from the GEMA (gout evaluation of man- spherulites in the synovial fluid. J Rheumatol.
agement) study. Ann Rheum Dis. 2008;67 Suppl 1993;20:1975.
2:246. 1035. Barthelemy I, Karanas Y, Sannajust JP, et al. Gout
1020. Singh J, Hodges J, Toscano J, Asch S. Quality of of the temperomandibular joint: pitfalls in diagno-
care for gout in the US needs improvement. Arthritis sis. J Craniomaxillofac Surg. 2001;29:307.
Rheum. 2007;57:822. 1036. DeSantis D, Palazzi C, D’Amico E, et al.
1021. Mikuls T, Farrar J, Belker W, et al. Suboptimal phy- Acromioclavicular cyst and “porcupine shoulder’’
sician adherence to quality indicators for the man- in gout. Rheumatology. 2001;40:1320.
agement of gout and asymptomatic hyperuricemia: 1037. Currey H. Letter: primary gout affecting the sterno-
results from the UK General Practice Research clavicular joint. Br Med J. 1976;1:583.
Database (GPRD). Rheumatology. 2005;44:1038. 1038. Perez-Ruiz F, Calabozo M, Alonso-Ruiz A. Gouty
1022. Mikuls T, Curtis J, Allison J, et al. Medication arthritis in the manubiosternal joint. Ann Rheum
errors with the use of allopurinol and colchicines: a Dis. 1997;56:571.
retrospective study of a national anonymous inter- 1039. Kerr R. Radiologic case study. Sacroiliac joint
net-accessable error reporting system. J Rheumatol. involvement by gout and hyperparathyroidism.
2006;33:562. Orthopedics. 1988;11:187.
1023. Perez-Ruiz F, Carmona L, De Miguel E, et al. 1040. Justiniano ME, Colmegna I, Gimenez CR, Espinoza
Variability in the treatment of gout: results from the LR. Tophaceous gout of the symphysis pubis.
GEMA audit of clinical practice. Ann Rheum Dis. Arthritis Rheum. 2005;52:4052.
2008;67 Suppl 2:246. 1041. Parhami N, Feng H. Gout in the hip joint. Arthritis
1024. Annemans L, Spaepen E, Nash C, et al. Medical Rheum. 1993;36:1029.
management of chronic gout in the UK and 1042. Nakajima A, Kato Y, Yamanaka H, et al. Spinal
Germany. Ann Rheum Dis. 2006;65 Suppl 2:430. tophaceous gout mimicking a spinal tumor. J
1025. Sundy JS. Gout management: let’s get it right this Rheumatol. 2004;31:1459.
time. Arthritis Rheum. 2008;59:1535. 1043. Weinzweig J, Fletcher JW, Linburg RM. Flexor ten-
1026. Edwards NL. Treatment failure gout: a moving tar- dinitis and median nerve compression caused by
get. Arthritis Rheum. 2008;58:2587. gout in a patient with rheumatoid arthritis. Plast
1027. Choi HK, Ford ES. Prevalence of the metabolic Reconstr Surg. 2000;106:1570.
syndrome in individuals with hyperuricemia. Am J 1044. Suk KS, Kim KT, Lee SH, et al. Tophaceous gout
Med. 2007;120:442. of the lumbar spine mimicking pyogenic discitis.
1028. Faccini F, Chen YO, Hollenback CB, Reaven GM. Spine J. 2007;7:94.
Relationship between resistance to insulin-medi- 1045. Wazir NN, Moorthy V, Amalourde A, Lim HH.
ated glucose uptake, urinary uric acid clearance, Tophaceous gout causing atlanto-axial subluxation
and plasma uric acid concentration. JAMA. mimicking rheumatoid arthritis: a case report. J
1991;266:3008. Orthop Surg (Hong Kong). 2005;13:203.
1029. Wallace SL, Robinson H, Masi AT, et al. Preliminary 1046. Li TJ, Lue KH, Lin Z, Lee KH. Arthroscopic treat-
criteria for the classification of the acute arthritis of ment for gouty tophi mimicking an intraarticular
primary gout. Arthritis Rheum. 1977;20:895. synovial tumor of the knee. Arthroscopy.
1030. Malik A, Schumacher H, Dinnella J, Clayburne G. 2006;22(910):E1–3.
Clinical diagnostic criteria for gout: comparison 1047. Yu K, Luo S, Liou L, et al. Concurrent septic and
with the gold standard of synovial fluid crystal gouty arthritis. Rheumatology. 2003;42:1062.
analysis. J Clin Rheumatol. 2007;15:22. 1048. Rose JD. J Pharmacokinet Biopharm. 1981;9:389.
1031. Sivera F, Aragon R, Pascual E. First metatarsopha- 1049. Szefler SJ, et al. Eur J Clin Pharmacol.
langeal joint aspiration using a 29-gauge needle. 1986;30:323.
Ann Rheum Dis. 2000;67:273.
1032. Velilla-Moliner J, Martinez-Burgui J, Cabeta-
Garcia JC, Fatah-Baudpey ML. Podagra, is it
always gout? Am J Emerg Surg. 2004;22:320.
Index

A Acute gouty arthritis, 92, 95, 96, 99, 101, 116, 124, 136,
ABCG2. See ATP-binding cassette subfamily G member 137, 140, 141, 191, 199, 200, 207–209, 211,
2 (ABCG2) 212, 218, 219, 228, 229, 237, 238, 240, 243,
Accelerated ATP degradation 252, 253, 255, 259, 262, 294–297, 301, 304,
acetaldehyde, 78 313–315, 322, 323, 327, 331, 348, 353
acetyl-AMP, 78 Acute inflammatory response, 187, 207, 208, 218, 231,
acetyl-CoA synthetase, 78 232, 234, 253, 259, 261–263, 270, 276, 278,
alcohol dehydrogenase, 78 316, 319, 320
b-hydroxybutyrate, 78 Acute uric acid nephropathy
carnitine palmityltransferase deficiency, 78 medullary cystic disease, 117
ethyl alcohol, 78 polycystic kidney disease, 117
glucose-6-phosphatase deficiency, 75 Acute uric acid nephropathy management
glycogen storage diseases, 78 alkylating agents, 340
hypoxemia, 80 allopurinol, 340
inosine, 79 hematopoietic system, 340
lactate, 78 hemodialysis/peritoneal dialysis, 340
muscle metabolism, 78 malignant neoplasms, 340
NADH, 78 tubular interstitial nephritis, 340
nicotinamide adenine dinucleotide ultrasonography, 340
(NAD), 78 urine alkalinization, 340
phosphofructokinase deficiency, 78 Adenine, 4, 25, 77, 107, 249, 330
phosphoglycerate mutase Adenine phosphoribosyltransferase (APRTase), 35, 42,
deficiency, 78 47–54, 131–133
secondary gout, 75 ACG, 51
Von Gierke’s disease, 75 adenosine monophosphate (AMP), 47
xanthine oxidase, 79 APRT enzyme forms, 51
Accelerated nucleotide degradation ATG, 51
AMP deaminase, 84 deficiency, 47
myoadenylate deaminase, 83 de novo, 54
Acute attack, 93, 97, 207–278, 295, 296, 315, 316, 320, 2,8-dihydroxyadenine stones, 51
323, 339, 354, 355 2,8-dihydroxyadenine urolithiasis, 52
Acute attack gout mechanisms familial juvenile hyperuricemic nephropathy
acute gouty arthritis, 207–208, 211, 212, 218, 219, (FJHN), 53
228, 229, 237, 238, 240, 243, 252, 253, 255, gene, 49
259, 262 hereditary orotic aciduria, 54
acute inflammatory response, 207, 208, 218, 231, inhibition of APRTase activity, 47
232, 234, 235, 259, 261–263, 270, 276, 278 Morquio syndrome, 52
apoptosis, 208, 216, 230, 233, 234, 256, 257, 260, mutant forms of APRTase, 49
262, 269–272 TGA, 51
cell migratron, 208, 243, 247 TGG, 51
phagocytosis, 207–209, 230, 233, 234, 242, 247–252, type I APRTase deficiency, 49
256, 258–260, 263, 265, 269, 270, 273 Adenosine deaminase, 34, 79, 108, 140
Acute attacks Adenosine kinase, 34, 35
prophylaxis against, 355 Adenosine monophosphate (AMP), 11, 28, 30, 31,
treatment, 354 33–36, 38, 40–43, 47, 54, 76, 79–84, 101, 107,
urate-lowering treatment, 354 108, 140, 235, 263, 269, 276, 310

D.S. Newcombe, Gout, 387


DOI 10.1007/978-1-4471-4264-5, © Springer-Verlag London 2013
388 Index

Adenylic, 27, 55 Antituberculous drugs, 137, 326


Adenylosuccinase, 33, 34, 40 Anton de Storck, 4
Adenylosuccinate, 33, 34, 40 Anton van Leeuwenhoek, 2
Adhesion molecules, 210, 211, 216, 217, 219, 221, 228, Apoptosis, 208, 216, 230, 233, 234, 256, 257, 260, 262,
237–245, 255, 261, 272, 277 269–272, 312, 329
E-selectin, 242–243 APRTase. See Adenine phosphoribosyltransferase
integrins, 238, 243 (APRTase)
integrins ligand, 245–247 Archibald E.Garrod, 5
integrin structure, 243–245 Arthrocentesis techniques, 188–189
L-selectin, 239–240 Associated disorders
P-selectin glycoprotein ligand-1, 239 DNA Polymorphisms, 151–152
selectins (structures), 238–239 familial apolipoprotein c-II deficiency, 149
unique L-selectin properties, 240–242 familial combined hyperlipidemia, 149
ADPKD. See Autosomal dominant polycystic kidney familial lipoprotein lipase deficiency, 149
disease (ADPKD) familial lipoprotein lipase inhibitor, 149
AIC. See 5-amino-4-imidazolecarboxamide (AIC) fish-eye disease, 151
Albrecht Wallenstein, 6 high density lipoprotein (HDL), 148
Alcohol, 11, 12, 78, 80, 93, 100, 101, 105, 116, 135, 136, lecithin/cholesterol acyltransferase (LCAT)
141, 142, 148–150, 188, 191, 194, 199, 202, deficiency, 151
222, 266, 291, 293, 313, 324, 339, 348, 349, low density lipoproteins (LDL), 148
352, 354 monogenic hypertriglyceridemia, 149–150
Alexander Gutman, 5 nonsteroidal anti-inflammatory drugs
Alexander Hamilton, 6 (NSAIDS), 148
Alexander of tralles, 2 type III hyperlipoproteinemia, 150–151
Alexander the great, 6 very low density lipoproteins (VLDL), 148
Alfred Garrod, 5 Asymptomatic hyperuricemia
Allopurinol chlorthalodine, 293
acute gouty arthritis, 331 chronic renal disease, 292
alloxanthine (oxypurinol), 329 diuretics, 293
azathioprine, 330 drug-induced hyperuricemia, 293
cyclophosphamide, 330 ethambutol, 294
de novo, 330 hypertension, 293
feedback inhibition, 330 nicotinic acid, 294
histocompatibility locus antigen (HLA), 334 pyrazinamide-induced hyperuricemia, 294
hypersensitivity reactions, 332 renal calculi, 292
induced gout, 136–137 salicylates, 293
6-mercaptopurine, 330 Atherosclerosis, 11, 147, 148, 150, 151, 153, 154, 352
orotidine-5-monophosphate decarboxylase, 330 ATP-binding cassette subfamily G member 2 (ABCG2),
oxypurinol, 331 16, 17
prophylactic colchicine, 331 ATP degradation, 75–81, 83, 107, 108, 113, 125
standard treatment regimens, 330 Aulus Cornelius Celsus, 1
Stevens-Johnson syndrome (STS), 333 Autosomal dominant polycystic kidney disease
T cell-mediated delayed hypersensitivity, 334 (ADPKD)
toxic epidermal necrolysis (TEN), 332, 333 cerebral aneurysms, 119
toxic reactions, 331 hypertension, 119
treatment stones, 348 renal failure, 119
vasculitis, 332 urinary tract infections, 118
in vitro, 330, 332 Avascular necrosis, 95, 199, 202 See also Osteonecrosis
in vivo, 330
xanthine oxidase, 330
Amino acid structure of CSa, 235 B
5-amino-4-imidazolecarboxamide (AIC), 48 Bacterial arthritis, 96, 97, 194
AMP. See Adenosine monophosphate (AMP) Beer, 11, 12, 136, 339, 348, 349, 352
Ancient greek and roman civilization, 1 Benjamin Franklin, 4, 6
Annexins, 278 Ben Johnson, 6
Anti-inflammation, 265, 270–271 Benzbromarone, 14–16, 96, 133, 138, 139, 325–326,
Anti-inflammatory eicosanoid, 232, 262–263 328–329
Anti-inflammatory lipoxins effects severe hepatotoxicity, 329
in vivo, 265 Biological functions of PAF, 227–228
Antimetabolites, 35, 48, 106, 291, 295, 330, 337 Blunt ends, 192
Index 389

Body mass index, 11, 349 Colchicum autumnale, 6


Bony erosions, 199–201 Co-morbidities
tophi, 200 alcohol, 348
Bursa, 92, 94, 95, 199, 200, 203, 208 beer, 349
Byzantine empire, 2 dietary factors, 348
wine, 349
Compensated polarized microscopy, 191, 192, 194
C Complement fragments, 220–221
C5a, 210, 218–222, 227, 229, 235, 240, Cotton Mather, 6
241, 243, 244 Count Nikolaus Ludwig von Zinzendorff (German
C5a des Arg, 220–221 theologian), 6
Calcium pyrophosphate dihydrate (CPPD) crystals, 98, Crystal identification, 99, 191, 192, 296
188, 191–193, 202, 209, 215, Cyclooxygenase inhibitors classification
257, 297, 298 cardiovascular events, 312
Cardinal Wolsey (the last medieval prince of the renal hemodynamics, 311
church), 6 Cyclosporine, 91, 93, 95, 96, 118, 128, 135, 137–139,
Cardiovascular disease, 10, 15, 147, 153, 232, 303, 306, 145, 294, 300, 301, 328, 329, 346
311, 320, 339, 351 Arthrobacter protoformiae, 138
C5a receptor, 220–222, 227 Aspergillus flavus, 138
Carl Linnaeus, 6 Candida utilis, 138
Carl Scheele, 3 Cytokines, 15, 194, 210, 211, 214–218, 220, 236–238,
Carpal tunnel syndrome, 94, 105, 353 240–242, 244, 245, 253, 255–262, 264, 269,
Cell membrane, 14, 43, 209, 241, 249, 250, 257, 271, 273, 275, 277, 315, 319, 334
259–260, 275, 297, 301, 302 Cytotoxic agents, 135, 136, 294
Cell migration, 208, 243, 247
Charles Darwin, 6
Chemoattractants, 207, 209, 210, 218–220, 222, D
227–229, 235–237, 240, 241, 244–246, 253, DAMPs. See Inflammasome
263, 320 DAP. See 2,6-diaminopurine (DAP)
Chemotactic peptide generation, 297 Decreased hypoxanthine reutilization, de novo, 74, 75
Cholesterol, 10, 12, 109, 110, 139, 148–153, 329, 345, DECT. See Dual energy computed tomography (DECT)
351, 352 Denis, W., 4
Chondrocalcinosis, 98, 199, 202–203 De novo, 5, 13, 25, 30–34, 37, 39–44, 46, 47, 53–55,
pseudogout, 202 69–76, 81, 82, 103, 104, 106–108, 111, 127,
Chondrocytes, 15, 266 140, 141, 223, 226, 227, 330, 352
Chronic gouty arthritis, 2, 94–96, 348 De novo purine nucleotide synthesis
Chronic renal failure, 10, 53, 87, 105, 118, adenosine monophosphate, 30
127, 142, 352 adenosine triphosphate (ATP), 30
Chronic tophaceous gout, 2, 5, 92–96, 98, 99, 109, 137, adenylosuccinase, 30
200, 324, 342 aspartic acid, 30
Clinical aspects of gout and associated carbon dioxide, 30
disease states de novo, 30
decreased renal clearance of urate, 91 energy phosphate compounds, 30
increased purine biosynthesis, 91 formate, 30
microcrystals, 91 10-formyl-tetrahydrofolate, 30
primary gout, 91 fumarate, 30
pseudogout, 91 glutamine, 30
Secondary gout, 91 glycine, 30
synovium, 91 guanosine monophosphate, 30
uric acid overproduction and underexcretion, 91 inosine-5’-phosphate (IMP), 30
Colchicine 5, 10-methenyltetrahydrofolate, 30
acute gout, 296 5-phospho-a-D-ribosyl pyrophosphate (PRPP), 30
gastrointestinal toxicity, 296 (COMP: Please insert correct symbol.)
mechanism action, 297 phosphoribosylamine, 30
myopathy, 96, 300 5’-phosphoribosyl-5-aminoimidazole
pharmacokinetics, 298–299 (AIR), 30
toxicity 5’-phosphoribosyl-5-aminoimidazole-4-carboxamide
colchicine-induced myopathy, 299 (AICAR), 30
colchicine myoneuropathy, 299 phosphoribosylaminoimidazolecarboxamide formyl
intravenous colchicine, 300 transferase, 30
390 Index

De novo purine nucleotide synthesis (cont.) hypomagnesemia, 98


phosphoribosylaminoimidazole hypophosphatasia, 98
carboxylas, 30 immunocompromised host, 96
5’-phosphoribosyl-5-aminoimidazole-4-carboxylate infected prosthetic joints, 97
(CAIR), 30 joint infections, 97
5’-phosphoribosyl-5-aminoimidazole-4-(N-succino) N. gonorrhoeae, 97
carboxamide (Succino-AICAR), 30 positive birefringence, 98
phosphoribosylaminoimidazole-succinocarboxamide pseudogout, 96, 97
synthetase, 30 Pseudomonas aeruginosa, 97
phosphoribosylaminoimidazole synthetase, 30 psoriatic arthritis, 98
5’-phosphoribosyl-5-formamidoimidazole-4- Reiter’s syndrome, 98
carboxamide (formyl-AICAR), 30 S. aureus, 97
phosphoribosyl-formylglycineamidine synthetase, 30 septic arthritis, 96
5’-phosphoribosyl-glycineamide (GAR), 30 Serratia marcescens, 97
phosphoribosylglycineamide formyl transferase, 30 severe degenerative arthritis, 96
phosphoribosylglycineamide synthetase, 30 synovial fluid
5’-phosphoribosyl-N-formylglycinamidine culture, 97
(FGAM), 30 glucose, 97
5’-phospho-ribosyl-N-formylglycine (FGAR), 30 white blood cell count, 97
phosphoribosylpyrophosphate amidotransferase, 30 syphilis, 97
purine salvage pathways, 30 traumatic arthritis, 96, 98
ribosylamine, 30 2,8-dihydroxyadenine lithiasis
Deoxyribo nucleic acid (DNA), 25, 26, 28, 39, 44, 46, adenine phosphoribosyltransferase, 132
47, 49, 53, 119, 148, 245, 251, 260, 261, 271, adenine phosphoribosyltransferase
273, 275–278, 317, 343 deficiency, 133
Deoxyribose phosphate, 26 2,8-dihydroxyadenine stones, 36, 124, 127, 129, 132
Desquamation, 93 Diuretic(s), 11, 86, 87, 91, 93, 95, 100, 105, 115, 117,
Destructive arthropathy, 94 127, 135–137, 145–147, 148, 150, 152, 194,
DeWitt Stetten, 5 293, 306, 307, 309, 311, 314, 324, 326, 328,
Diabetes mellitus, 10, 11, 96, 98, 100, 139, 147, 149, 332, 345, 346, 349, 353, 354
150, 152, 189, 294, 295, 309, 349, 351, 352 amiloride, 135
2,6-diaminopurine (DAP), 33, 38, 48, 50, 51 benzothiadiazines, 135
Dicarboxylates, 16, 17 diuretic-induced gout, 135
Diet ethacrynic acid, 135
alcohol, 348 furosemide, 135
beer, 349 induced hyperuricemia, 115
dietary factors, 348 spironolactone, 135
wine, 349 triamterene, 135
Dietary restrictions, 11, 12, 339, 342, 352 Drug-induced hyperuricemia and gout, 135
Differential diagnosis Dual energy computed tomography
acute neuropathic joints, 98 (DECT), 203–204
aspiration, 97 Dyslipidemia, 10, 11, 152, 154, 349, 351–353
bacterial arthritis (septic arthritis), 96
calcification, 98
calcium pyrophosphate E
crystal deposition disease, 96 Egyptian archeology, 1–2
dihydrate, 97 Elimination of uric acid
charcot joints, 98 acetoacetate, 85
chondrocalcinosis, 98 b-hydroxybutyrate, 85
chronic neuropathic joints, 98 lactate, 85
chronic tophaceous gout, 98 urate reabsorption mechanisms, 85
E. coli, 97 uric acid renal elimination, 84
gram-negative infections, 97 urinary uric acid excretion, 85
gram-positive organisms, 97 Emil Fischer, 4, 25
gram stain, 97 Empress Maria Theresa of Austria, 4
hemochromatosis, 98 Endonucleases, 26
hemodialysis, 97 E-selectin, 210, 237–239, 241–243, 261
HIV disease, 97 Ethanol, 77, 78, 80, 100, 101, 103, 105, 107, 135–137,
hyperparathyroidism, 98 307, 348
Index 391

Excessive purine intake, 84, 105, 128 intercellularadhesionmolecule-1(ICAM-1), 320


Exonucleases, 26 lipocortin-1, 318
Extra-articular tophi, 94, 199, 201 mechanism, 316
methylprednisolone, 322
neutrophilia, 319
F neutrophil migration, 320
Familial juvenile gouty nephropathy (FJGN), 120–121, phospholipase, 319
144, 145 prednisone, 316
Familial juvenile hyperuricemic nephropathy (FJHN), transcortin, 316
13, 53–55, 120, 145, 353 in vitro, 317
Famous sufferers, 6 in vivo, 317
Fatty acid oxidation deficiencies Glucose/fructose transporter, 13
carnitine palmitoyltransferase II, 113 GLUT9, 14–16
heritable palmitoyltransferase deficiency, 113 Glutamine hypothesis, 72
myoglobinuria, 113 GLUT9DN, 14
Febuxostat GLUT9L, 14
adverse reactions, 337 GLUT9S, 14
hepatic metabolism, 337 Glycogen storage diseases (GSD)
xanthine oxidase, 337 b2-microglobulinuria, 341
Females gout calcium oxalate, 109
hyperuricemia and gout in pregnancy, 146 creatine phosphokinase, 110
juvenile hyperuricemia and gout, 144–145 debranching enzyme
postmenopausal hyperuricemia and gout, 146–147 deficiency, 109
premenopausal hyperuricemia and gout, 145–146 de novo, 111
Fenofibrate generalized aminoaciduria, 341
Fenofibrate, hyperlipidemia, 345 glucose-6-phosphatase deficiency, 108
Fever and chills, 92 glycogen storage disease type XI, 113
FJGN. See Familial juvenile gouty nephropathy (FJGN) hemolytic anemia, 112
FJHN. See Familial juvenile hyperuricemic nephropathy inosine monophosphate, 111
(FJHN) lactate dehydrogenase-A deficiency, 113
Folin, O., 4 metabolic myopathies, 111
Fructose, 11–16, 76–78, 107, 111, 127, 133, 135, muscle phosphoglycerate mutase
139–140, 338, 341–342, 349, 352 deficiency, 112
Fructose-1,6-bisphosphatase deficiency, 78, 107 myoadenylate deaminase deficiency, 111
Fructose-induced hyperuricemia, 11, 12, 107, 139–140 myoglobinuria, 110
de novo, 140 myopathy, 110
Fructose-1-phosphate, 11, 140 nephrocalcinosis, 109
phosphofructokinase activity deficiency, 111
phosphofructokinase deficiency, 110
G phosphoglycerate kinase deficiency, 112
Galen, 1 phosphoglycerate mutase deficiency, 111
Gemfibrozil, 12 phosphorylase kinase deficiency, 112
Generation of reactive oxygen radicals, 247 Tarui’s disease, 110
Genetic abnormalities, 13–17 type III, 110
Genetic polymorphisms of SLC2A9/GLUT9, 14 type V, 110
Genome-wide association studies (GWAS), 13, 14, 16 type VII, 110
George Hitchings, 5 GMP. See Guanosine-5’-monophosphate
George Mason (American statesman and drafter of the (GMP)
constitution), 6 GRE. See Glucocorticoids
Gertrude Elion, 5 GTP, 33, 34, 36, 82, 83, 140, 216, 221
Glucocorticoids, 278 Guanine, 4, 25, 27, 28, 30, 34, 42–44, 46, 72, 81, 130,
annexin, 318 249, 330, 336, 337
chemotaxis, 319 Guanosine-5’-monophosphate (GMP), 31, 33–35, 40–43,
cyclooxygenase synthesis, 318 46, 55, 82, 272
dexamethasone, 316 Guanosinic acid, 72
glucocorticoid-induced inhibition, 318 Guanylic, 26
glucocorticoid-inducible protein, 318 Guanylic acid, 27, 28, 55
glucocorticoid response elements (GRE), 317 GWAS. See Genome-wide association
hydrocortisone, 316 studies (GWAS)
392 Index

H solubility uric acid, 99


HapMap, 14 toxemia pregnancy, 100
Hematological malignancies, 108, 128 urate nephropathy, 100
Henry Fielding, 6 urolithiasis, 100
Hereditary fructose intolerance (HFI), 77, 78, 107, 127, management
133, 139, 140, 341–342 colchicine, 291
Hereditary xanthinuria, migratory polyarthritis, 131 corticosteroids, 291
Heterozygote carriers of HGPRTase deficiencies, 72 NSAIDs, 291
HFI. See Hereditary fructose intolerance (HFI) overview, 291–292
HGPRTase. See Hypoxanthine-guanine proper, 291
phosphoribosyltransferase (HGPRTase) uric acid lowering therapy, 291
High-energy phosphates, 30, 31, 34, 101, 140 uricosuric agents, 291
Hippocrates, 1 Hyperuricemia mechanisms
Histone acetylation and glucocorticoids, 277–278 1-14C-glycine, 72
HLA. See Allopurinol de novo, 72
HPRTase. See Hypoxanthine-guanine Hyperuricemia renal mechanisms
phosphoribosyltransferase (HGPRTase) decreased tubular secretion, 85
Hydrogen peroxide, 11, 248, 250–252, 260 postsecretory reabsorption, 85
Hyperglycemia, 10, 152, 217, 351, 352 pyrazinamide suppression test, 85
Hyperlipidemia, 11, 12, 108, 109, 147–154, 291, 294, urate excretion, 85
312, 345, 352, 354 urate underexcretion, 86
Hypertension Hyperuricosuric calcium oxalate nephrolithiasis, calcium
alcohol, 352 oxalate, 134
de novo, 352 Hypoxanthine, 5, 13, 25, 71, 101, 291
dyslipidemia, 351 Hypoxanthine-guanine phosphoribosyltransferase
meat, 351 (HGPRTase)
metabolic syndrome, 351 adenine phosphoribosyltransferase (APRTase), 42
obesity, 351 deficiency, 41, 72, 73, 106, 121, 123, 124, 144, 145
seafood, 351 de novo, 42–44, 46
vascular disease, 351 dimagnesium PRPP, 43
Hypertriglyceridemia, 9–12, 33, 148–152, 345, 350–352 DNA methylation, 46
Hyperuricemia hypoxanthine-guanine phosphoribosyltransferase
chemical properties of the uric acid, 187 deficiency, 44
uric acid has a solubility, 187 enzyme, 44
Hyperuricemia and gout gene, 44
diagnostic criteria for pseudogenes, 43
acetate, 101 inosine monophosphate (IMP) dehydrogenase, 45
acetoacetate, 101 Kelley-Seegmiller syndrome, 45
acetylcoenzyme A, 101 Lesch-Nyhan syndrome, 45
acute renal failure, 101 NAD synthetase, 45
adenosine monophosphate, 101 nicotinamide adenine dinucleotide (NAD), 45
adenosine triphosphate, 101 nicotinic acid phosphoribosyltransferase, 45
asymptomatic hyperuricemia, 100 X-chromosome activation, 46
b−hydroxybutyrate, 101 X-chromosome inactivation, 46
chemotherapy, 102
compensated polarized light microscopy, 99
ethanol, 101 I
hypercatabolic states, 101 ICAM-1. See Glucocorticoids
hyperlacticacidemia, 101 ICAMs. See Intercellular adhesion molecules (ICAMs)
hyperuricemia, 100 IkB, 261, 262, 273, 274
inosine, 101 IL-8 gene, in vitro, 237
lactate, 101 Immunoglobulin, 192, 213, 230, 233, 245, 319, 335
needle-like uric acid crystals, 99 IMP. See Inosine monophosphate (IMP)
nitroglycerin, 101 Increased cells deficient, 33
normal serum uric acid levels, 99 Increased nucleic acid turnover
nucleoproteins, 101 adenine phosphoribosyltransferase, 81
5’-nucleotidase, 101 ATP degradation, 83
radiation, 102 de novo, 81
rhabdomyolysis, 101 epidermal turnover, 81
seizures, 102 hemoglobinopathies, 81
Index 393

hemolytic anemias, 81 polycystic kidney disease, 94


hyperlacticacidemia, 83 sarcoidosis, 94
hyperuricosuria, 83 Interleukin-8 (IL-8), 209, 210, 218–221, 227–229,
ineffective erythropoiesis, 81 235–238, 240, 241, 244–246, 253, 255, 256,
multiple myeloma, 81 258, 259, 261, 267, 274, 319, 320
muscle contractions, 83 amino acid structure, 235–236
muscular exercise, 83 Amino Acid Structure of CSa, 235
myeloproliferative disorders, 81 Amino Acid Structure
neoplams, 81 of IL-8, 235
nucleotide degradation, 81 Interleukin-18, 216–217
pentose phosphate pathway, 81 Interleukin-1b, 216–217
pernicious anemia, 81 Interleukin-1band interleukin-18, 216–217
phosphoribosylpyrophosphate (PRPP), 81 Interleukin 1 inhibitors
polycythemia, 81 anakinra, 322
psoriasis, 81 canakinumab, 323
Indomethacin decoy receptor, 323
toxicity, 314 rilonacept, 323
in vivo, 314 In vitro, 35, 54, 131, 210, 212, 213, 237, 249, 255, 264,
Inflammasome 297, 303, 310, 317, 330, 332
autoinflammatory, 213, 322 In vivo, 54, 234, 235, 237, 241, 249, 264, 265, 273, 297,
caspase-1, 214, 216–218 314, 317, 330
cryopyrin, 213, 216 Isaac Newton, 6
danger-associated molecular patterns (DAMPs), Ischemic heart disesae, 10, 115, 152, 351, 352
214–215 Isotopes
IL-1b, 15, 213–215, 218 accelerated production of uric
IL-1b, 216 acid, 54
innate immune system, 213–216 de novo, 54, 55
NLRP3, 213–217 estimation, 54
Nod (nucleotide oligomerization domain)-like glycine alpha-N15, 54
receptors (NLRs), 215, 216 glycine-1-C14, 54
pathogen-associated molecular patterns (PAMPs), rate of purine synthesis, 54
214–215 in vitro, 54
periodic fever syndrome, 213, 214 in vivo, 54
periodic fever syndromes, 213, 214
pro-IL-1b, 216, 217
RIG-I-like receptors (RLRs), 215 J
Inflammatory cell chemotaxis, 297 James Wyngaarden, 5
Inflammatory response, 53, 92, 127, 187, 188, 194, 207, Jay Seegmiller, 5
208, 210, 212, 215, 216, 218, 221, 223, 227, Johann Wolfgang von Goethe, 6
230–232, 234, 237, 238, 243, 246, 247, John Buchanan, 5
252–256, 258–267, 270–274, 276–278, 294, John Calvin, 6
297, 298, 301, 302, 310, 316, 317, 319, 320 John Dryden, 6
Inhibitory concentration, 297 John Hunter, 6
Inosine monophosphate (IMP), 30, 31, 33–35, 40, 43, 45, John Milton, 6
46, 55, 74, 79, 80, 82, 83 John Wesley (English evangelist and
Inosine 5’-phosphate dehydrogenase, 34 theologian), 6
Inosinic acid, 27, 28, 33, 34, 40, 72–74, 79, 80, 82, 83, Joint effusions, magnetic resonance
108 image, 200
Insulin resistance, 10, 12, 139, 149, 152, 349, 351, 353 Joint involvement, 92, 94, 147
Insulin resistance syndrome, 10, 152, 351
Integrins, 210, 211, 237, 238
Integrins, 210, 211, 227–229, 237, 238, 240, 241, K
243–247, 258 Kelley-Seegmiller syndrome
Integrin structure, 243–245 hypoxanthine-guanine phosphoribosyltransferase,
Intercellular adhesion molecules (ICAMs), partial deficiency, 104
210, 245 nephrolithiasis, 105
Intercritical gout, 92–94, 193 neurological dysfunction, 105
asymptomatic gout, 94 Kidney stones, 11, 12, 15, 47, 48, 104–106, 122–125,
familial juvenile gouty nephropathy, 94 128, 132, 134, 187, 201, 352
glomerulonephritis, 94 Kossel, 25
394 Index

L Monoarticular or oligoarticular arthritis,


Leonardo da Vinci, 6 92, 95, 96, 99, 354
Lesch-Nyhan, 5, 35, 44, 45, 103–105, 115, 120, 123, Monocarboxylates, 16, 17
124, 127–129, 131, 141, 145, 341, 353 Monogenic mutations, 15
Lesch-Nyhan syndrome Monosodium urate (MSU)
central nervous system dysfunction, 103 crystal-induced neutrophil activation, 256
choreoathetosis, 104 crystallization
de novo, 104 acute gout, 211
hypoxanthine-guanine diagnosis of acute gout, 211
phosphoribosyltransferase, 103 hyperuricemia, 211
nephrolithiasis, 103 immunoglobulins, 213
neurological disorder, 103 intra-articular temperatures, 212
Leukotriene B4 (LTB4), 218, 219, 229–231, 233, 235, serum, 211, 212
253, 315, 319 in vitro, 212, 213
biosynthesis crystals, 93, 94, 191–193, 199, 204, 207, 213, 215,
lipoxygenase, 230–231 217, 218, 237, 253–260, 273, 298, 353
phospholipase A2, 230–231 monohydrate crystals, 91, 116
Lifestyle, 354 Monosodium urate crystal
Ligands for integrins, VEGF, 246 in vitro, 210, 255
Lipoxin biosynthesis Musculoskeletal ultrasound
in vitro, 264 hyperechoic enhancement, 203
in vivo, 264 radiological examinations, 199
Lord Beaverbrook, 6 urate deposits, 203
Lord Howe, 6 Myeloperoxidase (MOP), 230, 250–252, 260
Losartan Myelosuppression, 96
angiotensin II (AII) receptor antagonist Myoadenylate deaminase deficiency,
(ARB), 345 83, 108, 111, 112
uricosuric effect, 346
Low-purine diet, 49, 339
purine-free diets, 339 N
L-selectin, 210, 238–243 NAD+, 34
LTA4 Hydrolase, 230–232 NADPH oxidase assembly
in vitro, 249
in vivo, 249
M Naproxen
MAP kinase, 230, 249, 256, 259, 278 cardiovascular toxicity, 315
Marshal Saxe, 6 treatment regimens, 315
Martin Luther, 6 Neoplastic disease
Mechanisms of NLRP3 activation acute uric acid nephropathy, 295
asbestos, 217 antineoplastic treatment, 295
MSU crystals, 217 Nephrogenic diabetes insipidus (NDI), 121–122
reactive oxygen species (ROS), 217 Nephrolithiasis, uric acid calculi, 348
silica, 217 Neurological deficits, 44, 95, 104–106, 353
Medullary cystic disease (MCD), 119–120 Neutrophil and macrophage apoptosis, 260
Megaloblastic anemia, 104, 106, 117, 140–141 Neutrophil chemoattractants
de novo, 141 chemoattractants, 207, 218–219, 227, 229, 235, 237,
Mercaptopurine, 5, 25, 27, 31, 35, 37, 42, 96, 137, 138, 253, 263
295, 330, 332, 337 chemotactic cytokines, 218, 219
6-mercaptopurine, 5, 25, 27, 31, 35, 37, 42, 96, 137, 138, neutrophil chemotaxis, 218–220, 227
295, 330, 332, 337 Neutrophil migration, 207, 219–220, 235, 237, 240, 241,
Metabolic myopathies, 77–79, 107, 111, 113 247, 320
Metabolic syndrome, 10–12, 15, 16, 152, 338, 349, NF-kB, 215, 256, 261, 262, 271–275, 277
351–353 NHANES III, 9
Michael Lesch, 5 Nicotinic acid, 45, 87, 135, 139, 294
Miscible pool of urate Nitric oxide, 9, 216, 217, 242, 248, 251–252, 264, 265,
de novo, 70 270–274, 349, 351
15
N-labeled uric acid, 71 Nitric oxide and bacterial killing, 251–252
Mode of action NLRP3 activation mechanisms
glucocorticoid receptors, 275 asbestos, 217
glucocorticoids, 277–278 MSU crystals, 217
Index 395

reactive oxygen species (ROS), 217 P


silica, 217 PAF. See Platelet-activating factor (PAF)
NLRs. See Inflammasome PAMPs. See Inflammasome
Nonsteroidal anti-inflammatory drugs Pegloticase
(NSAIDs) adverse events, 344
arachidonic acid, 301 antibodies, 344
cyclooxygenase-1 [COX-1], 301 glucose-6-phosphate deficiency, 345
cyclooxygenase-2 [COX-2]), 301 infusion reactions, 344
eicosanoids, 301 Krystexxa, 344
inhibition of cyclooxygenase, 301 mammalian recombinant uricase, 344
nonselective NSAIDs, 302 Phagocytosis, 207–209, 230, 233, 234, 242, 247–252,
prostaglandin endoperoxides, 301 256, 258–260, 263, 265, 269, 270, 273, 297,
selective COX-2 inhibitors, 303 319, 320
toxicity Phosphatases, 33, 34, 75, 249, 265, 298, 299
acute renal papillary Phosphoenol pyruvate carboxykinase deficiency, 107
necrosis, 308 Phospholipase classification, in vivo, 234
gastrointestinal bleeding, 307 Phospholipase D, 257–258, 298
hyperkalemia, 309 Phospholipids metabolizing enzymes, 258
hypertension, 309 Phosphoribosylamine (PRA), 33, 72, 81
hyponatremia, 309 5-phosphoribosyl-1-pyrophosphate, 33, 37
hyporeninemic hypoaldosteronism, 309 Phosphoribosyl pyrophosphate (PRPP), 5, 13, 30–47, 50,
nephrotoxic disorders, 307 51, 54, 55, 71–76, 81, 82, 103, 105–107, 111,
NSAID-induced interstitial nephritis, 308 117, 120, 123, 124, 127, 128, 132, 133,
NSAID-induced nephrotoxicity, 309 139–141, 145, 291, 292, 330, 337, 348, 353
sodium retention, 309 amidotransferase
upper GI toxicity, 305 adenosine monophosphate (AMP), 40–42
in vitro, 303 de novo, 40–42
NPT1, 16, 17 feedback inhibitors, 40
Nucleases, 26, 261 feedback regulation, 40
Nucleic acids, 4, 26, 28, 30, 31, 33, 55, 69, 70, gene, 41
75, 78, 81–83, 91, 100, 103, 105, 107, glutamine phosphoribosylpyrophosphate
108, 111, 115–117, 124, 128, 129, amidotransferase, 40
136, 145, 261, 262 guanosine monophosphate (GMP), 40–42
Nucleoside, 27, 28, 34, 54, 70, 75, 79, 80, 140, inosine monophosphate (IMP), 40
218, 328, 330, 336, 337 5-phosphoribosylamine, 40
Nucleoside phosphorylase(s), 27, 28, 34, 54, 79, phosphoribosylpyrophosphate (PRPP), 40–42
80, 140, 336, 337 ribonucleotides, 40
5’-nucleotidases, 28, 34, 35, 79, 80, 101, 140 Phosphoribosyl pyrophosphate synthetase (PRPP
Nucleotide, 11, 14, 15, 17, 25, 30–43, 47, 52, 54, 70, 71, synthetase), 5, 13, 32, 33, 36–40, 71, 73, 82,
73–77, 81–84, 100, 106, 108, 110, 127, 214, 103, 106, 117, 120, 123, 124, 128, 132, 133,
215, 227, 232, 249, 251, 258 348, 353
allosteric regulation, 36
amidotransferase, 36
O de novo, 37, 39
Obesity, 9, 10, 12, 33, 100, 147–150, 152–154, 291, 293, hypoxanthine-guanine phosphoribosyltransferase, 36
294, 313, 324, 332, 339, 348–354 overactivity
Oded Sperling, 5 de novo, 106
Olecranon bursa, 92, 94, 95, 200, 203 neurological deficits, 106
Oligoarticular, 92 phosphoribosylpyrophosphate
Organ transplantation, 95, 137, 145, 202, 328, 329 amidotransferase, 106
Orotic acid, 33, 38, 54, 330 pretranslational regulation, 39
Osteonecrosis, 202 Platelet-activating factor (PAF), 210, 218–230, 232, 235,
Oxidants and oxidant-mediated destructive 238, 241, 244, 255, 259, 263, 264, 270, 271
responses acetylhydrolase, 228–229, 270–271
fenton reaction, 250 biological functions, 227–228
haber-weiss reaction, 250 de novo, 223, 230
Oxidants and uric acid destruction, 252 receptor, 222, 223, 227, 241
Oxipurinol calculi, 133 Podagra, 1, 92, 99, 212, 353
Oxygen free radicals, 11, 255, 259 Polyarticular, 92, 95–98, 320, 321
Oxygen radical generation, 227, 247–249 Polyarticular gout, 92, 98, 320, 321
396 Index

Polymerase chain reactions, 194 Renal tubular transport, 13–17


Prepatellar bursae, 94, 200 Resolvin biosynthesis, resolution, 266
Prevalence, 9–17, 100, 122, 123, 148, 309, 349, 351, 352 Resolvin receptors, 266–270
Primary underexcretor gout, 114 Rheumatoid nodules, 95
Probenecid Rhomboid, 192, 193
acute gouty arthritis, 327 Ribo nucleic acid (RNA), 404, 442, 449, 450, 579, 583,
benemid treatment regimens, 327 585, 587
methotrexate toxicity, 327 Ribonucleotides, 31, 33, 35, 40
uric acid nephrolithiasis, 327 Ribose phosphate, 26, 33
Proinflammatory mediators Ribose-5-phosphate, 32, 33, 36–39, 73, 75, 111, 140
bradykinin, 254 Ribose phosphate pyrophosphokinase, 31, 33, 34, 37,
non-chemotactic, proinflammatory cell 39–42, 46, 55, 72, 105, 123, 124, 140, 141,
mediators, 253 145, 291, 292, 324. See also Phosphoribosyl
tumor necrosis factor, 254–255 pyrophosphate synthetase (PRPP synthetase)
vasoactive arachidonic acid metabolites, 253 RLRs. See Inflammasome
Prophylactic colchicine, 93, 331 Roentgenographic findings, 199–204
Proteinases, 219–220, radiological examinations, 199
237, 252
P-selectin glycoprotein ligand-1, 220, 239, 242, 243
Purine(s), 4, 11, 25–55, 69, 91, 187, 201, 260, 291 S
kinase, 34 Salicylates, 102, 115, 126, 133, 135, 136, 138, 294,
nucleotides, 25, 28, 30–34, 37–43, 52, 54, 70, 71, 325–328, 348
73–76, 78–82, 100, 106–108, 127, 136, 141, Salvage pathways, 13, 30, 33–38, 40, 43, 54, 352
336, 337 Samuel Johnson, 6
Purine biosynthesis Secondary gout renal mechanisms
decreased renal function, 70 acetoacetate, 87
de novo, 69, 70 b-adrenergic blockers, 87
glycine, 70 b-hydroxybutyrate, 87
hereditary renal hypouricemia, 70 branched chain keto acids, 87
overproducers of uric acid, 70 catecholamines, 87
secondary gout, 70 chronic renal disease, 86
urate excretion, 70 cyclosporin A, 87
urate overexcretors, 70 decreased glomerular filtration rate, 86
uric acid excretion, 69 dehydration, 87
uric acid overexcretion, 70 diabetes insipidus, 87
urinary uric acid excretion, 70 diuretic-induced hyperuricemia, 87
Pyruvate carboxylase deficiency, 107 furosemide, 87
Pyruvate dehydrogenase deficiency, 107 hyperuricemia of renal failure, 87
lactate, 87
lacticacidemia, 87
Q lead, 87
Queen Anne (queen of England, Scotland), 6 lead intoxication, 86
levodopa, 87
nicotinic acid, 87
R nonsteroidal anti-inflammatory drugs, 87
Reactive oxygen species (ROS), 9, 214, 217, 229, 241, pyrazinamide, 87
242, 248, 250, 274, 320, 329, 350, 351 salicylate, 87
Refractory gout saturnine gout, 87
allantoin, 343 secondary hyperuricemia, 86
allergic reactions, 343 tubular reabsorption of urate, 86
Aspergillus flavus, 343 tubular secretion of urate, 86
hydroxyisourate, 343 Secondary hyperuricemia, 81, 86, 87, 103, 105, 107–108
pegloticase, 343 Selectins (structures), 210, 211, 237–241,
rasburicase, 343 243–245, 247, 258
tumor lysis syndrome, 343 Septic joints, 92, 97, 98, 191
uricase, 343 Single nucleotide polymorphisms (SNPs), 14–17
Renal function determination Sir Alfred B. Garrod, 4
cockcroft formula, 334 Sir Thomas Sydenham, 2
gault formula, 334 SLC2A9, 13–17
Index 397

SLC5A8, 16, 17 Tophi, 1, 3, 70–72, 78, 91, 94–96, 99, 100, 105, 112,
SLC 5A12, 16, 17 137, 147, 199–204, 211, 213, 218, 294, 340,
SLC17A1, 16 342, 353, 354
SNPs. See Single nucleotide polymorphisms (SNPs) Transplant gout, 95, 96
Soft tissue swelling, 98, 199–200 Tsai F.Yu, 5
magnetic resonance image, 200 Type A synovial membrane lining cells (macrophage-like
STS. See Allopurinol synoviocytes), 209, 218
Sugar-sweetened soft drink, 11, 12, 15, 349, 352 Type B synovial membrane cell (fibroblast-like
Superoxide, 11, 221, 227, 230, 232, 248, 250–252, 257, synoviocyte), 209
258, 265, 272
Symptomatic drug-induced hyperuricemia
drug-induced, 294 U
symptomatic hyperuricemia, 294 URAT1, 14–17
Synovial fibroblast-like cells, 237, 255, 256 Urate-lowering therapy, uricosuric drugs, 324
Synovial fluid Urate nephropathy
analysis, 93, 97, 188, 189, 193 microbial uricase, 115
culture, 97, 99, 188–190 microtophi, 115
differential cell, 191 urate nephropathy, 115, 116
gram stain, 190, 191 Urate reabsorption, 14–16, 85, 86, 114, 115,
white blood cell counts, 97, 189–191, 194 125–127, 135
Synovial membrane, 207–211, 218, 219, 223, 231, 239, Urate transport, 13–16, 121, 126, 133
241, 255, 259, 264 Urate turnover, 72
chemoattractants, 207, 209, 210, 218–223, 227–229, Uric acid
235–237, 240, 241, 244–246, 253, 263 calculus
cytokines, 210, 211, 214–218, 220, 236, 238, 241, ultrasonography, 201
242, 244, 253, 255, 256, 259–262, 264, 269, uric acid nephrolithiasis, 201
271, 273, 275, 277, 315 excretion, decrease, 114–121
integrins, 210, 211, 241 metabolism
inter cellular adhesion molecules (ICAMs), 209, 210, de novo, 69
237, 240, 242, 244, 245, 256, 261 gout, 69
selectins, 210, 211, 239, 241, 243 hyperuricemia, 69
type A synovial membrane lining cells, 209, 218 mechanisms, 69
type B synovial membrane cell, 209 nucleic acids, 69
in vitro, 210, 212, 213, 237, 249, 255, 264 uric acid, 69
nephrolithiasis, 122–135
cystine stones, 125
T cystinuria, 125
TEN. See Allopurinol de novo, 127
Therapeutic regimens hemoglobinopathies, 125
ACTH dosage, 322 hereditary renal hypouricemia, 125
adrenocorticotrophic hormone (ACTH), 321 hypercalciuria, 126
estrogens, 321 hypouricemia, 125
hyperthyroidism, 321 Kelley-Seegmiller syndrome, 124
intra-articular injection steroids, 320 Lesch-Nyhan syndrome, 124
intravenous methylprednisolone, 321 malignancies, 127
mini-pulse methylprednisolone, 321 Prader-Willi syndrome, 129
polyarticular gout, 320 xanthinuria, 127
toxicities, 321 overproduction
6-thioguanine, 35, 37 de novo, 103
Thomas Gray, 6 hyperuricemia and gout, secondary
Tissue hypoxia, 77, 80, 81, 103, 105, 107, 154 forms, 103
Tophaceous deposts management Kelley-Seegmiller syndrome, 103
Lesch-Nyhan disease, 341 Lesch-Nyhan syndrome, 103
lithotripsy, 341 phosphoribosylpyrophosphate
oxypurinol stones, 341 (PRPP), 103
surgery, 340 phosphoribosylpyrophosphate synthetase
xanthine stones, 341 superactivity, 103
Tophaceous gout, 2, 5, 92–96, 98, 99, 106, 109, 137, 142, secondary hyperuricemia
152, 200, 292, 324, 328, 330, 332, 342, 353, 355 and gout, 103
398 Index

Urinary uric acid measurements William Heberden, 3


24-h urinary uric acid, 102 William Kelley, 5
24-h urine collection, 102 William Nyhan, 5
overproducer, 102 William Pitt the Elder and the Younger, 6
radiocontrast materials, 102
underexcretor, 102
uricase method, 102 X
Xanthine
dehydrogenase, 11, 124, 127, 130, 131
V lithiasis, 123, 129–130
Vascular disease, 10, 150, 151, 153, 312, 351 oxidase, 5, 11, 25, 27, 28, 48, 49, 52, 53, 73, 79,
Visceral tissues, 95 80, 130, 131, 133, 137, 140, 154, 217, 291,
294, 295, 324, 325, 329–337, 340, 343,
349, 350, 354
W Xanthinuria, 123, 127, 129–133
Wayne Rundles, 5 Xanthosine monophosphate (XMP), 31, 34, 45
William Congreve, 6 XMP. See Xanthosine monophosphate (XMP)

You might also like