You are on page 1of 154

10

Human Cytochrome P450 Enzymes


R Peter Guengerich

1. Background and History of human P450s lAl (ref [6]), 1A2 (ref [7]), 2A6
(ref [8]), 2C9 (ref [9]), 2D6 (refs [7], [10], [11]),
Development of the Field and 3A4 (ref [12]), were isolated in this general
manner. Another general approach that was used
Much of P450 research has always been done was purification from tissue on the basis of
with the view of appUcation to humans, even immunochemical cross-reactivity with animal
when done with experimental animals and P450si3-i5.
microorganisms. Research with the human P450s With the development of recombinant DNA
has been done in several stages, and clinical phar- technology, cDNAs for many of the human P450s
macology has utilized the knowledge at each point were cloned in the 1980s^^ In the late 1980s,
in its development. methods came into use for the heterologous
Aside from in vivo experiments with drugs, expression of P450s, first in mammalian and yeast
human P450 work in the late 1960s and 1970s was systems, and then in baculovirus and (by the mid-
done with tissue samples, primarily biopsies. Some 1990s) in bacterial systems^^^^. In the late 1980s,
data on metabolic patterns and rates were col- the nomenclature system developed by Nebert^^
lected^. In the late 1970s, several groups began to was applied and allowed individual human and
purify P450s from human liver microsomes. The other P450s to be discussed on the basis of their
first purified proteins, selected because of their sequences. (For a guide to some of the earlier
abundance and ease of purification, were probably nomenclature, see ref [22].)
what are recognized now as P450 3A and 2C sub-
By the early 1990s, much of the interest in
family proteins^^. Efforts were shifted to purifying
P450 research had shifted to the human P450
individual P450s on the basis of catalytic activities
enzymes because of the availability of systems for
with the evidence that in some cases a single P450
handling these. In particular, studies in the areas
could be identified in this way; for example, the
of drug metabolism and chemical toxicology/car-
enzyme now known as P450 2D6 was found to be
cinogenesis were facilitated by the knowledge that
under monogenic control^. The approach is techni-
a relatively small number of the P450s account for
cally demanding because of the need to do separa-
a large fraction of the metabolism of the drugs and
tions in the presence of detergents and then remove
other chemicals of interest. In the pharmaceutical
them from individual chromatography fractions
industry, the roles of the major hepatic P450s are
prior to analysis of catal3^ic activity. Nevertheless,
extensively studied in developing predictions

F. Peter Guengerich • Department of Biochemistry and Center in Molecular Toxicology, Vanderbilt University
School of Medicine, 638 Robinson Research Building, Nashville, TN.
Cytochrome P450: Structure, Mechanism, and Biochemistry, 3e, edited by Paul R. Ortiz de Montellano
Kluwer Academic / Plenum Publishers, New York, 2005.

377
378 F. Peter Guengerich

about bioavailability, drug-drug interactions, and utmem.edu/CytochromeP450.html). Much of the


toxicity. Since the publication of the chapter on progress since 1995 has involved extrahepatic
human P450s in the last edition of this book^^, P450s, many with roles in the processing of
apparently all of the remaining human P450 genes "endobiotic" chemicals, for example, sterols and
have been identified, and the number (57) appears vitamins.
to be complete because of more general knowl- The list of the 57 human P450s is presented in
edge about the human genome (html://dmelson. Table 10.1, along with available knowledge about

Table 10.1. Human P450s

Subcellular
P450 Tissue sites localization'^ Typical reaction^

lAl Lung, several ER Benzo[a]pyrene 3-hydroxylation


extrahepatic sites,
peripheral blood cells
1A2 Liver ER Caffeine TV^-demethylation

IBl Many extrahepatic ER 17(3-Estradiol 4-hydroxylation


sites, including lung
and kidney
2A6 Liver, lung, and ER Coumarin 7-hydroxylation
several extrahepatic sites
2A7 ER
2A13 Nasal tissue ER Activation of
4-(methylnitrosamino)-1 -
(3-pyridyl)-1 -butanone
(NNK)
2B6 Liver, lung ER (iS)-Mephenytoin A^-demethylation
2C8 Liver ER Taxol 6a-hydroxylation
2C9 Liver ER Tobutamine methyl hydroxylation
2C18 Liver ER
2C19 Liver ER (5)-Mephenytoin 4'-hydroxylation
2D6 Liver ER^ Debrisoquine 4-hydroxylation
2E1 Liver, lung, other ER Chlorzoxazone
tissues 6-hydroxylation
2F1 Lung ER 3-Methylindole activation
2J2 Lung ER Arachidonic acid oxidations
2R1
2S1 Lung ER
2U1
2W1
3A4 Liver, small intestine ER Testosterone 6p-hydroxylation
3A5 Liver, lung ER Testosterone 6 p-hydroxylation
3A7 Fetal liver ER Testosterone 6 p-hydroxylation
3A43 (mRNA detected in gonads) (ER)

4A11 Liver ER Fatty acid w-hydroxylation


4A22 ER
4B1 Lung ER Laurie acid w-hydroxylation
4F2 Liver ER Leukotriene B^ w-hydroxylation
4F3 Neutrophils ER Leukotriene B^ w-hydroxylation
4F8 Seminal vesicles ER Prostaglandin a)-2 hydroxylation
Human P450s 379

Table 10.1. Continued

Subcellular
P450 Tissue sites localization" Typical reaction*

4F11 Liver ER
4F12 Liver ER Arachidonic acid ci)-,a)-2-hydroxylation
4F22
4V2
4X1
4Z1
5A1 Platelets ER Thromboxane A^ synthase reaction
7A1 Liver ER Cholesterol 7a-hydroxylation
7B1 Brain ER Dehydroepiandrosterone
7a-hydroxylation
8A1 Aorta, others ER Prostacyclin synthase reaction
8B1 Liver ER 7a-hydroxyprogesterone
12-hydroxylation (?)
llAl Adrenals, other Mit Cholesterol side-chain
steroidogenic tissues cleavage
llBl Adrenals Mit 11-Deoxycortisol 11-hydroxylation
11B2 Adrenals Mit Corticosterone 18-hydroxylation
17A1 Steroidogenic tissues ER Steroid 17a-hydroxylation
19A1 Steroidogenic tissues, ER Androgen aromatization
adipose, brain
20A1
21A2 Steroidogenic tissues ER 17-Hydroxyprogesterone
21-hydroxylation
24A1 Kidney Mit 25-Hydroxyvitamin D3 24-hydroxylation
26A1 Several ER Retinoic acid 4-hydroxylation
26B1 Brain ER Retinoic acid 4-hydroxylation
26C1 (ER?)
27A1 Liver Mit Sterol 27-hydroxylation
27B1 Kidney Mit Vitamin D3 1-hydroxylation
27C1
39A1 Liver (?) ER 24-Hydroxycholesterol 7-hydroxylase
46A1 Brain ER Cholesterol 24-hydroxylation (?)
51A1 Liver, testes ER Lanosterol 14a-demethylation

^ER = endoplasmic reticulum (microsomal), Mit = mitochondria.


^If known.
•^Mainly ER, some detected in mitochondria.

sites of tissue expression, subcellular localization, appear to be expressed primarily in the endoplas-
and a typical reaction. In the 1995 edition^^, the mic reticulum and only 6 are located exclusively
list included 31 human P450s, and 2 of these have in mitochondria. (However, work in animal mod-
been dropped as apparent cloning artifacts (2C10, els by Avadhani has clearly demonstrated the
2C17), leaving 29. Thus, the list was only half import of what have been generally considered
complete at that time. It should be emphasized microsomal P450s into mitochondria^"^' ^^. The
that we still have little knowledge about the roles basis of this transport appears to be signals in the
of some of these P450s beyond the genomic infor- N-terminal region^^. Limited information is avail-
mation. Of the 57, most that have been examined able about this phenomenon with human P450s;
380 F. Peter Guengerich

recent collaborative experiments with Avadhani's vary considerably (Figures 10.1 and 10.2).
group indicate that many human liver samples Deficiencies in the expression or catalytic activities
contain immunochemically detectable and catalyt- of most of the P450s involved in steroid metabohsm
ically active P450 2D6 in mitochondrial as well lead to serious diseases (Table 10.3) (included in
as microsomal fractions pST. Avadhani and this hst^^ are P450s IBl [true function unknown]
RR Guengerich, unpublished results].) In many and 24A1 [vitamin D hydroxylation]). In one case,
cases, no direct information is available because a high level of P450 activity (P450 19) can be a
protein studies have not been done, although most problem (estrogen formation in estrogen-dependent
of these "orphan" P450s are predicted to reside tumors) and this P450 is a target for therapeutic
primarily in the endoplasmic reticulum. attenuation^^. The 15 identified xenobiotic-metabo-
Of the P450s with known catalytic activities, 14 lizing P450 are mainly in the families 1-3, and lev-
are clearly involved in steroidogenesis, 4 are els of these vary considerably in humans (Figures
involved in what appear to be important aspects of 10.1 and 10.2). Studies with transgenic (knockout)
metabolism of vitamins (vitamins A and D), 5 are mice do not indicate critical function associated
involved in eicosanoid metabolism, 4 appear to have with the apparent orthologs in the absence of xeno-
fatty acids as their substrates, and 15 catalyze trans- biotic challenge^ ^ Analysis of the lists of drugs in
formation of xenobiotic chemicals (Table 10.2). which individual human P450s are involved^^ indi-
Some of these categories should be considered ten- cates that —75% of the drugs can be oxidized by
tative. This classification accounts for only 42 of the 3 P450s (3A4, 2D6, 2C9), and a set of 6-7 P450s
57 P450s. Many of the xenobiotic-metabolizing will account for 90-95% of all drug metabolism
P450s can also catalyze oxidation of steroids and (Figure 10.3)^^. Similar numbers of P450s are
fatty acids, but these functions do not appear to be involved in the metabolism of chemical carcino-
critical to homeostasis (e.g., testosterone 6p-hydro- gens, although the pattern shifts from that of Figure
xylation by P450 3A4, lauric acid 11-hydroxylation 10.3, with P450s 2C19 and 2D6 being replaced by
by P450 2E1, possibly 17p-estradiol 4-hydroxyla- P450S lAl, IBl, 2A6, and 2E1 (Table 10.4). The
tion by P450 IBl). Most of the steroid-oxidizing relative contributions of these xenobiotic-metaboliz-
enzymes are critical, and the levels of these P450s ing P450s are, to some extent, a function of the rel-
are relatively invariable among individuals, in con- ative abundance (Table 10.5, Figure 10.4), although
trast to the xenobiotic-metabolizing P450s, which there are some important exceptions. Further, the

Table 10.2. Classification of Human P450s based on Major Substrate Class""

Sterols Xenobiotics Fatty acids Eicosanoids Vitamins Unknown

IBl lAl 2J2 4F2 24 2A7


7A1 1A2 4A11 4F3 26A1 2R1
7B1 2A6 4B1 4F8 26B1 2S1
8B1 2A13 4F12 5A1 27B1 2U1
llAl 2B6 8A1 2W1
llBl 2C8 3A43
11B2 2C9 4A22
17 2C18 4F11
19 2C19 4F22
21A2 2D6 4V2
27 A1 2E1 4X1
39 2F1 4Z1
46 3A4 20
51 3A5 26C1
3A7 27C1

''As pointed out in the text, this classification is somewhat arbitrary, for example, P450s IBl and 27A1
could be grouped in two different categories.
Human P450s 381

uoi)e|AxojpAg-,|. |0|ejn^ng

•§ eei
0) ^
^^^^^^H ozv
zzi
g
^^^^^1
^^^^Hjm ezi-
931.
uojiepjxo 3U|djpa^!N I^
CO ^
o>
T—
1
'
8U CO -g
O
CM
^HH^^^HHIH^ 9 U
^^^^^^^^^^^^^H 9U
oLO ^^^"^^^jj^^^^^S* zu
Q. IHHl 01-1-
lull 601.
801-
^^^^^^^9 lOi

^^^^^^^5
^•[
001.
86
68
O O
o
CO
CM
d
d
uojieiAxojpAii-,^ ujO)Au3Ljda|/\|-(s)

uo|)e|AxojpAi|-9 auozexozjomo

ri ^
_o o
"Si
o
> ^
r^
-S CO

X!
o
uo!)eiAi|)adp-o uj^mosajAxoiii^-z 1— s
a
3
>.2 (1>
o> '^
o
382 F. Peter Guengerich

^ 2
1 •• j,„ — , , , , ,
Is
w O
< <| < < < <| <i <
-D
<A
t/) PH

< <
CM CO
o • • • 1 • • •• • • o
in
1
IT) • • • • ^ •••
GL
• • 1•••• •• Q.

D
o
D
1 l_ -1- 1 ..._., 1 . , , 1 1 ..,.

in o o o o
d <0 'sf CM

r 1 1 ' 1 ' 1

< < < \< << < <C 1 << <

< < < <S<< < Q ^ < < B


CM
<< «€iei^< <W<R1

CM m a m iDDa aa • • u. .2 «
o U> B OH

D a • DO D • DDDXCDn Q. •• •
o 3
0. "oS
D ^ >
D Z

1 • 1 - J 1 J 1
1
CM 00 ^ O
>
O
as
c .a

r—T '— 1 — 1 1 1 j '> c c3


T3 (U -^
II op 13
fi -s
"D ^o C/3 O
<< < <$i«i <cmf <m <>5 d N
CM s >
JJ
o • • m • • •• • •• u.
to TS

P ~^-^ t/5

D
T3 ^ S
_ 1 - I .......1 • J
>
O ^ -S
s T3
(U
43 2
^
^
S f^
o
u.|
-D S
s cd (U
0)
CX 00
UL
o
1/3
^1
2
2
o O
c
o V / "O

u
p^ c3 cd
o o o fa
CO CM 1- o
U O b
09l7d |B»01 o/o
uia^ojd leuiosojojui Bui/os^d puiu CM 5 ^
O
T1 iS
£
3
SF2

il 1^
Human P450s 383

Table 10.3. Diseases Associated with Mutations in CYP Genes^^

Gene Disorder

CYPIBI Primary congenital glaucoma (buphthalmos)


CYP4A, 4B^ Defects in salt metabolism, water balance leading to arterial hypertension
CYP5A1, 8A1 Defects leading to clotting and inflammatory disorders, coronary artery disease, and
pulmonary hypertension
CYP7A1 Hypercholesterolemia, resistance to statin drugs
CYP7B1 Severe hyperoxysterolemia and neonatal liver disease
CYPUAl Lipoid adrenal hyperplasia; occasional congenital adrenal hyperplasia (CAH)
CYPllBl Occasional CAH
CYP11B2 Corticosterone methyloxidase deficiency type I, or type II; occasional CAH
CYPllBl, 11B2 Chimeric enzymes causing glucocorticoid-remediable aldosteronism; occasional CAH
CYPUAl Mineralocorticoid excess syndromes, glucocorticoid, and sex hormone deficiencies;
association with increased risk of prostate cancer and benign prostatic hypertrophy;
occasional CAH
CYP19A1 Loss of function: virilization of females, hypervirilization of males, occasional CAH; gain
of function: gynecomastia in young males
CYP21A2 >90%ofallCAH
CYP24A1^ Hypervitaminosis D
CYP27A1 Cerebrotendinous xanthomatosis
CYP27B1 Vitamin D-dependent rickets type I

''Strong evidence of disease in animal models but not yet in clinical studies.

2. General Issues of Variability


and Polymorphism

Variability in patterns of drug metabolism has


been recognized for some time, even before the
discovery of P450s. For instance, the phenomenon
of pharmacogenetic variation had been identified
1A1 by the 1950s^^' ^^ and the early work of Remmer"^^
showed the influence of barbiturates upon drug
metabolism. Further, a number of congenital
defects in steroid metabolism were known and
some could be attributed to alterations in specific
hydroxylations'*^ Much of the subsequent work
on inducibility has been done in experimental ani-
mal models"^^ and later, cell culture.
2C19 In the 1960s and 1970s, a number of accounts
Figure 10.3. Estimated contributions of individual appeared describing variations in rates of metabo-
human P450s to the metabolism of all drugs, based upon lism of drugs in human liver biopsy samples^ The
literature available (adapted from Evans and Relling)^^. first characterization of a monogenic variability in
a human drug-metabolizing P450 was the work of
Smith with debrisoquine^, which was paralleled
patterns reported for human liver (Table 10.5, by the work of Dengler and Eichelbaum on
Figures 10.1, 10.2, and 10.4) do not necessarily sparteine"*^. This polymorphism was first described
apply in other tissues, some of which may be targets in the context of extensive metabolizers (EMs) and
for carcinogens and other toxicants^^. poor metabolizers (PMs) (Figure 10.5). These
384 F. Peter Guengerich

u
o

.-o-I t i l l
.5 .S 2 >. ^
.2 b <u J^

bo^ m
X ^ g ^ o 6 ^ i^ 2
o s^
(U OH

^^
Xi
r'^

H
o
O)
w-i

a
(D
<L>
C/3 T—1

w
^—^ rs|
^
ly,
<==>
u^
a
S
W) ^ CIH
o
.s
u

II 11
t 5> 2 t £
( D O , - — <D ON

4 -^ Z 4 -^

e / S
1
^ 4, ^
00^

6 16 O
3
T3 PU en
1c
rT
1 < c

•S N 6
•c
OH £
a
g 'B t 0)
c
<u
o
o
^ .s
o e s OH
o c
'Jo
it
1
O 6 6 o o o
c c c c 43
c
c N
S rs» "s 6 'o 'B •g "? < ^
•g •g t5
<1 < c
< 1 < 4 < <
<N 1
(N

o o /•

CL|
S

T3
c
cd o
-e
cd
S
PH
o 2^ O
C ^ N

>. 43O 6 1 ^•£a.


rQ
^
13 c
N
'>>
C
'o < O. i n
PQ o.
Human P450s 385

^II
P +1 ^J"

+1 +1
o

:3

u +1 ^ +1 2

u +1 ^

w
0^
OQ +1 :^ +1 p +1 ^
00 o ^ O

'B
+1 ::: +1 ^ +1 ^
o ^^

d
•^ n
12 +1 o +1 o +1 o

•I ^

C «^

U
II
386 F. Peter Guengerich

Substrates
Nifedipine
Mephenytoin Tolbutamide Midazolam Caffeine Debrisoquine
Omeprazole Phenytoin Erytfiromycin Theophyline Sparteine
Coumarin Warfarin Cyclosporin Tacrine Chlorzoxazone

Inhibitors Fluconazole Disulfiram Quinidine


Methoxsalen Sulfaphenazole

Inducers Barbiturates Barbiturates Barbiturates Omeprazole


Rifampicin Rifampicin Rifampicin Tobacco smoke Ethanol
Dexamethasone Isoniazid
Carbamazepine

Figure 10.4. A summary of major human P450s involved in drug metabolism, including major substrates,
inhibitors, and inducers (adapted from Breimer)"^^' ^^. The sizes of the circles indicate the approximate mean
percentages of the total hepatic P450 attributed to each P450 (See also Figure 10.1 and Table 10.5). The overlap of
the circles is to make the point that overlap of catalytic action is often observed, although the overlap does not
necessarily refer to the indicated substrates (or inhibitors).

25

20

^ 15
c
0)

¥ 10 h

5 h

-20 -15 - 1 0 - 5 0 5 10 15 20

log-io Metabolic ratio

Figure 10.5. Frequency distribution histogram of (in vivo) debrisoquine 4-hydroxylation in a Caucasian
population'*'*. The metabolic ratio is the ratio of debrisoquine/4-hydroxydebrisoquine in the urine of individuals
who were administered debrisoquine (10 mg free base) 8 hr previously. The groups are designated PM (poor
metabolizers, solid bars) and EM (extensive metabolizers, gray bars). The group labeled "Ultra" is from retrospective
research"^^ and probably represents gene duplication.
Human P450s 387

polymorphisms were first studied at the level of Several allelic variants clearly lead to the PM
phenotype, that is, pharmacokinetics and in some phenotype, for a variety of reasons. A relatively rare
cases unusual responses to drugs due to reduced case is a gene deletion (*5)^^. The most common
metabolism"*^. The area of pharmacogenetics (now (Caucasian) PM phenotype is an SNP that leads to
also known as—or expanded to—"pharmaco- aberrant RNA splicing (i.e., in splice site) and no
genomics") was facilitated by the identification of mRNA or protein. Other alleles involve partial
the P450 enzymes involved in the drug metabolism deletions, frameshifts, and coding for protein with
phenotypes, and particularly by the development of either intrinsically low catalytic activity or instabil-
molecular biology, which allows the precise charac- ity (reduced halflife). These general patterns have
terization of genetic differences between individu- been seen in other P450s (and other genes). In addi-
als. The majority of the allelic differences are single tion to the EM and PM phenotypes, there is also an
nucleotide polymorphisms (SNPs), or single base "ultrarapid metabolizer" phenotype, due to gene
changes. As anticipated from previous knowledge of duplication. A Swedish family has been identified
pharmacoethnicity, many of these SNPs and poly- with 13 gene copies, leading to 13 times more
morphisms show racial linkage. (A polymorphism enzyme"^^. The level of hepatic P450 2D6 and a
is generally defined as a 1% frequency of an allelic parameter of in vivo debrisoquine metabolism (the
variant in a population; below this frequency, the urinary metabolic ratio = urinary debrisoquine/
terms "rare genetic trait" or "rare allele" are applied 4-hydroxydebrisoquine) vary ~10^ fold among
or, in the case of a very detrimental allele, a mutant people (Figure 10.5). With P450 2D6, and several
or "inborn error of metabolism.") other P450s, the alleles describing the high and low
The debrisoquine polymorphism is now under- levels of metabolism have been described, but the
stood in terms of P450 2D6 and has been a proto- kinetic parameters for many of the alleles have not
type for research in this area. The characterization of been determined by heterologous expression and
the gene^^ led to a basic understanding of the PM measurement of catalytic activity. This is still the
phenotype. The incidence of the PM phenotype is general case with most of the human P450s. P450
about 7% in most Northern European populations, 2D6 is regulated by a hepatic nuclear factor (HNF)
with different phenotypic incidence (and SNPs) in element^^, but is not considered to be inducible by
other racial groups'*"^' '^^~^^. More than 70 allelic xenobiotics. With many other P450s, there is regu-
variants are now known, and 98% of the PMs in lation and variability due to noncoding region
Northern European populations can be accounted SNPs, levels of inducers consumed, and inter-
for by four variant alleles^^' ^^ A nomenclature sys- actions between P450s and transporters, such as
tem has been set up for P450 alleles (using the P-glycoprotein^"^' ^^, may influence the phenotype.
suffixes *1, *2, *3,...) and is maintained by Although the level of P450 2D6 may have a
Oscarson at http://www.imm.ki.se/Cypalleles/. dramatic effect on the metabolism of certain drugs

Table 10.6. Some Major Inducers of Human P450 Enzymes

Class of inducers Some sources Example P450s induced"

Ah ligands Tobacco, broiled Polychlorinated lAl, 1A2


meat, accidental biphenyls
exposures
Barbiturates and similar Drugs, some Diphenylhydantoin 2C, 3A4
compounds polyhalogenated
biphenyls, DDT
PXR ligands Some steroids and Rifampicin 3A4
antibiotics, other
drugs
P450 2E1 inducers Ethanol, isomiazid Ethanol 2E1

^Based on in vivo responses.


388 F. Peter Guengerich

E
(movement
to nucleus?)

BR JR' J , P450 gene


P450gene \~ X^ ^
DNA DNA ^ — ^ ^ — ^
t •
RNA pol
(increased access
to promoter, start site)

Increased transcription

Figure 10.6. Generalized model for regulation of P450 genes by induction. L = ligand, R = receptor,
R' = partner protein for heterodimer of R, Coactiv = coactivator, RNA pol = RNA polymerase.

(Figure 10.5), no other biological changes have seen in humans, they tend to be relatively soon
been reported in PMs. This appears to be the after birth (e.g., P450 3A4, 3A7, see refs [58],
general case for many of the hepatic P450s prima- [59]), and changes in expression seen in the
rily involved in the metabolism of xenobiotics, elderly have not been very dramatic^^"^^.
and few observable physiological effects have
been reported in transgenic mice in which these
genes have been deleted^ ^. As pointed out earlier,
however, deficiencies in some of the steroid-
3. Approaches to Defining
hydroxylating P450s can be very debilitating or Catalytic Specificity of
lethal"* ^ In general, the variation in the levels Human P450s
of these "more critical" P450s is limited in most
of the population, compared to the xenobiotic- Knowledge of the roles of individual P450s in
metabolizing P450s in which an order of magni- specific reactions is critical in the application of
tude variation is not unusual^^. P450 biochemistry to practical issues in drug
The influence of inducers on the expression of metabolism. Originally some of the P450s were
each P450 will be mentioned later (Table 10.6, purified on the basis of their catalytic activities
Figure 10.6). It should be pointed out that several toward certain specific drugs^' ^' "' ^^^ i^y^ gy^j^ jj^
of the P450s can be downregulated by cytokines, those cases, there are the issues of the extent of
and the result has practical significance in the contribution of that form and the involvement of
impairment of drug metabolism in individuals that P450 in other reactions, particularly with new
with colds or flu, or who have received vaccina- drugs. Identification of the individual P450s
tions^^. Another general point to make is that, in contributing to the metabolism of a new drug can-
contrast to some animal models (see Chapter 8)^^, didate is routinely done in the pharmaceutical
human P450 expression shows little if any gender industry. This information is usually required by
differences. When developmental differences are the Food and Drug Administration at the time of
Human P450s 389

application. Identifying P450s involved in oxida- a titration approach (concentration dependence)


tion is important in predicting drug-drug interac- has merit^^.
tions and the extent of variation in bioavailability. Another general issue is the selection of a protein
In general, it is desirable to develop drugs for concentration. Microsomal proteins can bind drugs
which several P450s have a contribution to metab- in a nonselective manner and effectively lower
olism. Drug candidates that are metabolized the free concentration of substrate or inhibitor^^' ^^,
exclusively by a highly polymorphic P450 (e.g., which can influence the interpretation of results.
2D6, 2C19) are usually dropped from further Another point is that the concentration of the P450 of
development. interest should be less than that of the drug and the
A combination of methods involving the use of inhibitor, in order for the basic assumptions about
human tissues and recombinant human P450s is steady-state kinetics to apply (and for the reaction to
usually used to identify P450s involved in a particu- remain linear during the incubation time, although
lar reaction, using an approach outlined earlier^^' ^^. some of the inhibitors are mechanism-based and the
A combination of the following methods is usually loss of activity will be time dependent, requiring pre-
done, not necessarily in a particular order. Lu^^ has incubation). A corollary of these latter points, which
recently reviewed these approaches. also apply to the other approaches that follow, is that
having a very sensitive assay method is very desir-
able. Thus, methods such as HPLC/fluorescence and
particularly HPLC/ mass spectrometry have gained
3.1. Inhibitors
popularity.
The reaction is demonstrated in NADPH- Finally, the choice of an organic solvent is an
fortified human liver microsomes (if the reaction issue. Ideally the substrate should be dissolved in
of interest is restricted to another tissue, then this H2O or very little organic solvent, but this may not
tissue would be used instead). The effects of selec- be possible with many drugs. Several examina-
tive inhibitors on the reaction are examined. A list tions of the effects of individual solvents on
of some of the inhibitors that have been used is human P450s have been published^^' ^^.
presented elsewhere in this volume by Correia In principle, the extent of inhibition of a reac-
(Chapter 7)64,65 tion by a P450-selective inhibitor indicates the
The choice of concentration parameters is fraction of that reaction attributable to that P450.
important in this and some other approaches. For instance, if a 1 |xM concentration of quinidine
Ideally the effect of the substrate concentration on (a P450 2D6 inhibitor) inhibits 50% of a reaction,
the rate of catalytic activity should be determined in then 50% of that reaction may be attributed to
the absence of inhibitor to determine F _ and K P450 2D6. If one desires a more global view than
parameters. If this information is available, the inhi- within a single liver sample, then a pooled set of
bition experiments are best done with a concentra- microsomes (e.g., from 10 samples, balanced on
tion of substrate at or below the K. in order to the basis of liver weight or protein) may be used for
m'
the inhibition assays. However, if one desires to
observe the effect of the inhibitor on the ratio examine the differences among individuals in terms
V^^JK^, which is the parameter usually most rele- of the contribution of a P450, then doing several
vant to human drug metabolism. If the F ^^ and K experiments with individual liver samples is the
o max m
information is not available, an alternative is to approach to use.
select a substrate concentration near that expected
for the in vivo plasma concentration (C ^ ^ or less).
With regard to inhibitor concentration, ideally 3.2. Correlations
a range of concentrations would be used. However,
if a single concentration of the diagnostic inhibitor Another approach with a set of human tissue
is used, it must be selected on the basis of previous microsomal samples is to measure the new reac-
literature because nonselective effects are often tion of interest in each and attempt correlation
observed. For instance, a-naphthoflavone (otNF) with rates of marker activities (for individual
(5,6-benzoflavone) can inhibit P450s other than P450s). Lists are also published in this volume in
1A2 at high concentrations^^ and azoles inhibit Chapter 7 by Correia^^ and elsewhere^ ^.
many P450s at higher concentrations^"^' ^^. Use of
390 F. Peter Guengerich

Correlation can be done by plotting the spe- the antibody raised against the P450. (c) The anti-
cific activity for the new reaction vs the marker body should be shown not to inhibit a reaction
reaction (Figure 10.7). In principle, the correlation known to be attributable to other P450s. Immuno-
coefficient r^ estimates the fraction of the vari- globulin G fractions are generally preferred in that
ance attributable to the relationship between the they produce less nonspecific inhibition than crude
two activities, that is, the fraction of the activity preparations such as sera. Polyclonal antibodies can
catalyzed by the particular enzyme (assuming that vary in their specificity and titer from one animal to
all of the marker activity is catalyzed by this another and from one bleed to another, so constant
enzyme). In some cases, excellent correlations properties cannot necessarily be assumed. In prin-
have been reported^^' ^^. An alternative method of ciple, monoclonal antibodies and antibodies eluted
analysis is the Spearman rank plot, which has from phage display libraries should not vary,
some deficiencies but avoids the overweighting of although this has not always been the case with
unusually high or low values^"^. monoclonals.
Although the approach works well when high In general, antibodies are often selective for
correlation coefficients are generated, the method individual P450 subfamilies, for example, lA vs
is less usefiil when several P450s contribute to a IB vs 2A vs 2B vs 2C, etc., but cross-reaction
reaction, that is, r^ < 0.4. The results should, in all among families can be detected, and in some
cases, be considered in the context of results cases the (P450) sites of cross-reactivity have
obtained with other approaches. been identified^^. Achieving selectivity among
individual P450 subfamily members (e.g., P450
3A4 vs 3A5 vs 3A7) is more difficult. With poly-
clonal antibodies, this can be achieved by cross-
3.3. Antibody Inhibition
adsorption^^; with monoclonals and phage display
The points raised in the Section 3.1, Inhibitors, libraries, this can be done by selection. The point
apply to antibodies as well. Antibodies are used to should be made that any selectivity demonstrated
inhibit activities in human liver (or other tissue) among classes of animal P450s (e.g., rat P450
microsomes and are of several general types: families) cannot be assumed to carry over to
(a) polyclonal antibodies raised against purified human P450s.
animal P450s, (b) polyclonal antibodies raised Antipeptide antibodies have become popular
against purified human P450s, (c) monoclonal in recent years and have two major advantages:
antibodies raised against purified human P450s, (a) peptides can be synthesized and readily puri-
(d) polyclonal antibodies raised against peptide fied by HPLC, avoiding the need to express and
fragments of P450s, and (e) antibody phage dis- rigorously purify P450 proteins (although demon-
play library antibodies selected for recognition of stration of purity by HPLC, capillary electro-
individual P450s. phoresis, and mass spectrometry is still in order),
At this time, almost all antibodies raised against and (b) peptides can be selected for use as anti-
intact P450s have been generated using recombi- gens by sequence comparisons, favoring specific
nant P450s (or against peptides), in contrast to regions.
early work in the field with P450s isolated from Phage display antibody libraries are relatively
liver. Another point to make is that not all antibod- new and have been used in a few P450 applica-
ies inhibit catalytic activity. Further, specificity in tions to date (D.S. Keeney personal communica-
one immunochemical assay (e.g., electrophoretic/ tion). These have a number of advantages,
immunoblotting) does not necessarily implicate including potential selectivity due to the large
specificity in another (immunoinhibition). number of potential antibodies in libraries, the
Three points should be made in designing ability to avoid animal protocols, the immediate
immunoinhibition experiments, (a) The concen- availability of libraries (as opposed to waiting on
tration of antibody should be varied and increased animals to develop antibodies), the consistency of
to the point where the extent of inhibition is con- reproduction of the proteins propagated in bacter-
stant, (b) A nonimmune antibody should be used ial systems, and the ability to include a second
as a control, using the same concentrations as with "epitope tag" for recovery, etc.
Human P450s 391

to

uo!)e|AxojpAq-,|. |0|ejn^ng c
•• T"' o
Q.
« ] "5
£
o
(0
o o
o
r i • **r\« • • ] 1
CD
E
o

[_
O
-• 1

00 CD -^ CJ
E
Q.

0)
B B
uoj)e|AxojpAM-z uueuinoo co
o
oin
Q.

oIf) uo!)e|AL|)8ep-o uueuinooAxoL|)3-z o a


'St

0.
^ C3

13 o
o (Z)
^o
W)
c
o (L)
'rrt
?^ <CU3
^
t:
o
U W)
C
uoi)e|Ai|)aep-o uijnjosajAxomg-z 1^
cr>

o ^
TS
T- C

3
o> (U
U . TS
392 F. Peter Guengerich

3.4. Demonstration of Reaction (and the number is even higher if one moves the
with Recombinant P450 esterase and epoxide hydrolase reactions to the
Phase II group because they are not involved in
In early work in this field, this point would redox reactions). Constructing a figure of this type
have been the demonstration of the reaction of can be somewhat misleading in that the contribu-
interest with an enzyme purified from tissue. tion of each P450 is more difficult to evaluate
Today P450 proteins are generally produced in in vivo than in vitro (for a more original tabulation,
recombinant systems and seldom purified from see ref [32]). The large contributions of P450s
tissue sources. In routine practice in the pharma- 3A(4) and 2C9 are driven to a large extent by the
ceutical industry, new reactions are examined with high levels of expression of these two enzymes in
a battery of the major recombinant human (liver) human liver (and small intestine) and to their broad
P450s, many of which are available from com- substrate specificity. The charts do not necessarily
mercial sources. Systems used for expression reflect all drugs currently in development. A cur-
include bacteria, yeast, baculovirus (-infected rent tendency has been the development of larger
insect cells), and mammalian cells. The P450s molecules as drug candidates, in order to achieve
need not be purified for these comparisons but target specificity and affinity, and a general axiom
must have suitable provision for NADPH-P450 is that these are more readily accommodated by
reductase in a crude system (and cytochrome b^ P450s 3A(4) and 2C9. In recent years, pharmaceu-
[b^] in certain cases). tical companies have tried to avoid developing
Usually activity results obtained with several drug candidates that are substrates (or inhibitors)
of the major P450s are compared to each other for the highly polymorphic P450s 2D6 and 2C19.
and to those obtained with tissue microsomes, in With all of these caveats in hand, the allocation of
order to put the work in context. Ideally assays are the chart in Figure 10.3 is probably a good estimate
done at several substrate concentrations and the and may not change considerably in the near
parameters k^^^ (^max) ^^^ ^cat/^m ^^^ obtained. future. However, a point to be made here is that the
These values should be normalized on the basis of metabolism of many drugs is a function not only of
P450 concentration, in that any values based on P450s but also of other enzymes and, as recog-
milligram protein for the expression system can- nized more in recent years, transporters that alter
not be used for comparisons with tissue micro- the concentrations of drugs within cells. A discus-
somes. In principle, the k^^^ (total P450 basis) sion of drug transporters is outside the scope of
should be at least as high for the recombinant this chapter, and the reader is referred elsewhere^ ^
reaction as for the tissue microsomes. A more The subjects of P450 regulation and polymor-
realistic way to make a comparison is to immuno- phism (or mutation in some cases) have already
quantify the amount of the particular P450 in the been mentioned, and will be treated again, with
tissue microsomes and then use this value in cor- individual P450s. At this point, some general
recting the microsomal k^^^ for comparison with practical considerations will be discussed. If one
the recombinant system. The matter of scaling considers the total concentration of P450 in liver
these parameters to generate predicted microso- samples from different healthy individuals (on a
mal (or in vivo) rates from in vitro experiments milligram protein basis), most individuals fall
with recombinant enzymes is not trivial, but a within a range of ~3-fold'. However, when indi-
number of efforts have been made^'^~^^. vidual "drug-metabolizing" P450s (e.g., families
1, 2, 3) are considered, the variation is consider-
able, with 5-10-fold being common and 40-fold
not unusual, for example, P450 1A2 (ref [73]).
4. Relevance of P450s in In Vivo With P450 1A2, a similar variability (40-fold) is
Drug IVIetabolism seen in in vivo caffeine pharmacokinetics^^. With
highly polymorphic enzymes, the variability in the
P450s are the major enzymes involved in same in vivo pharmacokinetic parameters can be
human drug metabolism. In looking at the fraction as much as lO'^-fold (Figure 10.5).
of the number of drugs processed by "Phase I" Two examples of studies of the variability
enzymes (Figure 10.3), P450s account for >80% among individuals are presented in Figure 10.5
Human P450s 393

(Caucasians) and Figure 10.2 (Caucasian and Extensive Metabolizer (EM, normal)
Japanese). Gender has not been shown to have a
major influence on levels of expression of the
major xenobiotic-metabolizing P450s, and inter-
gender pharmacokinetic differences are probably
due to other influences on bioavailability or vol-
Plasma
ume of distribution^^. Racial differences exist due level of
to allelic variations, which may influence either drug
levels of expression or the inherent catal3^ic activ-
ity of the P450s [ref [49]). Some apparent racial
differences are seen here (Figure 10.2) and have
also been reported in in vivo studies (e.g., 3A4
(ref [83]), 2E1 (ref [84])). Controlling diets is an
issue in many in vivo studies of this type, and
in vitro studies can also be affected. In general, the
differences in activities of a given P450 between Time (arrows show repeated doses)
races are much less than within a race (e.g..
Figure 10.2). Finally, the point made above should Figure 10.8. Significance of low metabolism of a
be noted that the levels of the P450s involved in drug by P450s (or other enzymes). A "typical" pattern is
seen in the upper panel (EM), where the plasma level of
steroid metabolism (e.g., families 11, 17, 19, 21)
the drug is maintained in a certain range when a
vary considerably less than do the xenobiotic- particular repetitive dose is prescribed. Unusually slow
metabolizing P450s (families 1, 2, 3), probably metabolism (lower panel, PM) results in an elevated
due to their well-defined roles in regulation of plasma level of the drug. C ^^ = maximum plasma
physiological processes. concentration; AUC = area under the curve.
Many chemicals are capable of inducing
P450s, as clearly demonstrated in animals and population of interest. The plasma concentration
with cell culture systems^^. In vivo induction rises to a peak (C ^^J following the first dose and
experiments with humans are not as readily done then decreases to a lower level prior to the next
as with animals, but ample evidence for P450 dose. With subsequent doses, the plasma concen-
induction is available, going back to the barbitu- tration remains within this region and yields the
rate observations of Remmer in the 1950s^^. A list desired pharmacological effect. Without prior
of some established P450 inducers is presented in knowledge about a problem with this drug, the
Table 10.6. This list is rather conservative in that PM (lower panel of Figure 10.8) would be admin-
only information is included from studies in istered the same doses. Very limited metabolism
which in vivo evidence has been obtained. Many would occur between doses, and the plasma con-
of the studies have involved pharmacokinetics, but centration of the drug (and presumably the con-
some "moderately invasive" studies have involved centration of the drug in the target tissue) will rise
direct measurement of proteins, mRNA, or enzyme to an unexpectedly high level, with an attendant
activities in peripheral blood cells or small intes- increase in the area under the curve (AUC). The
tinal biopsies; liver biopsy data is rare. Table 10.6 simplest effect would be an exaggerated (and
could probably be expanded considerably if all probably undesirable) pharmacological response.
information from in vitro studies were included, One can also imagine a situation in which metab-
for example, P450s IBl and 2S1 are probably olism is more rapid than expected in the typical
inducible by Ah ligands^^' ^^. The major problem in patient, for example, due to gene amplification or
demonstrating human P450 induction in vivo is the enzyme induction. In this case, C ^^^ and AUC
lack of diagnostic pharmacokinetic parameters for would be smaller than in the case of the EM
many ofP450s. (Figure 10.8, upper panel), and decreased drug
The clinical influence of differences in P450 efficacy would be expected.
activity can be rationalized using the scheme of Some practical situations follow. With regard
Figure 10.8. In this model example, the drug doses to polymorphisms, several are known that can
have been developed with the EMs as the general render some drugs dangerous due to toxicity
394 F. Peter Guengerich

(e.g., perhexiline, leading to peripheral neuropa- is not available, it is possible that blocking the
thy due to lack of metabolism by P450 2D6 oxidation of one drug by a P450 could cause it to
(ref. [88]) or can alter the recommended dose (e.g., accumulate and behave as an inhibitor toward
warfarin/P450 2C9 (refs [89-91]) and omepra- another. A potential example would be decreasing
zole/P450 2C19 (refs [92], [93]). Drug interac- the P450 3A4-catalyzed oxidation of quinidine
tions are a serious problem, and pharmacokinetic and having the accumulated drug inhibit P450
interactions have several molecular bases. One 2D6 (ref [106]). P450 induction could result not
is enzyme induction, which usually results in only in decreased oral availability but also in the
decreased bioavailability. The decreased bioavail- enhanced bioactivation of chemicals. This is a
ability of a drug can be the result of induction by general concern with potential carcinogens, as
that same drug or by another drug. A classic discussed in the next section of this chapter, and
example is the decreased bioavailability of the one of the reasons why regulatory agencies have
oral contraceptive 17a-ethynylestradiol following concerns about P450 lA inducers.
treatment of individuals with rifampicin, barbitu- The phenomenon of P450 stimulation has been
rates, or St. John's wort and consequent P450 3A4 studied in some detail in vitro^^^. By stimulation
induction^^' ^^' ^^. Another aspect of drug-drug we mean the enhancement of P450 catalytic activ-
interactions involves P450 inhibition. The inhibi- ity by the direct addition of another compound,
tion can be of a competitive nature, that is, two outside of a cellular environment in which gene
substrates competing for a limiting amount of regulation is involved. Some aspects of P450 stim-
a P450 or a bona fide inhibitor (no enzymatic trans- ulation will be treated under the topic of P450
formation) competing with substrates. An example 3A4 (Section 6.20.4), with which much of the
here is the antihistamine terfenadine, the metabo- work has been done. An open question is whether
lism of which is inhibited by the P450 3A4 such behavior occurs in humans. At least four
inhibitors, erythromycin and ketoconazole. Another pieces of evidence suggest that such behavior is
major type of P450 inhibition is "mechanism- possible: (a) cooperativity has been reported in
based" (or "suicide") inactivation, in which oxida- hepatocyte cultures'^^, (b) an early experiment
tion of a substrate destroys the P450 (refs [64], with neonatal mice (individual P450s unknown)
[96]). An example here is the inactivation of P450 by Conney's group indicated the immediate
3A4 by bergamottin and other flavones found in enhancement of an activity by flavones'^^; (c) the
grapefruit juice^^ ' ^^. work of Slattery and Nelson with rats showed an
In the above cases, the effects have been dis- interaction between caffeine and acetaminophen
cussed only in terms of altered bioavailability; that that implies such behavior''^; and (d) quinidine
is, with increased clearance of 17a-ethynylestradiol, enhanced the in vivo oxidation of diclofenac in
unexpected menstruation and pregnancies have monkeys, in a manner consistent with in vitro
resulted^^' '^*' ^^^. Some of the drug interaction human work'''. If stimulation does occur in vivo,
problems can be more complex, even when the it is a phenomenon that has been very difficult to
analysis is restricted to pharmacokinetic aspects. predict (even in vitro), and in the case of P450
For instance, in the example mentioned above, ter- 3A4 substrates, the situation would probably
fenadine can be considered a prodrug'^^; in most be further complicated by issues involving P-
individuals, the P450 oxidation (followed by fur- glycoprotein behavior (and P-glycoprotein also
ther oxidation) yields fexofenadine, the circulat- shows cooperativity of its own^'^).
ing form of the drug. Low levels of P450 3A4 In the process of drug development, there are
activity (due to inhibition or other reasons) cause three guiding principles to dealing with P450
the accumulation of the parent (prodrug) terfena- metabolism, aside from details of each specific
dine to toxic levels that can cause arrhythmia^^^' ^^'^. case: (a) use in vitro screening to delete com-
Another possibility is that blocking a primary pounds that will have poor bioavailability (i.e.,
route of metabolism of a drug may favor second- rapid in vitro oxidation); (b) use in vitro screens to
ary pathways that lead to toxicity, for example, avoid obvious problems of toxicity, induction, and
blocking phenacetin 0-deethylation (P450 1A2) inhibition; and (c) seek drug candidates in which
can lead to deacetylation, A^-oxygenation, and the metabolism is the result of several different
methemoglobinemia^^^. Although a good example enzymes and not dependent upon a single one,
Human P450s 395

particularly a highly polymorphic P450 (or difficulties in the earlier phases of the work^^^.
another highly polymorphic enzyme). Many of the early problems have been circumvented
with the ability to measure mRNA expression and
the access to DNA sequences. While evidence for
correlation of P450 lAl mRNA expression with
5. Relevance of P450s in lung cancer incidence has been obtained^^^, an unre-
Toxicology and Cancer Risk solved issue is the nature of any genetic variability.
In contrast to the situation seen in mouse models ^^^,
Historically much of the attention given to P450s the allelic variations in the human Ah receptor
has come from the interest in cancer, going back to (which has apparently considerably lower affinity
some of the first demonstrations of redox reactions for many of the ligands of interest than the mouse
in the metabolism of chemical carcinogens^ ^^ and receptor^^^) do not appear to account for inter-
the inducibility of P450s by carcinogens^^. The individual levels of inducibility of P450 1 Al (refs
interest in P450s was also extended to chemical tox- [127], [128]). Kawajiri's laboratory has presented
icities other than cancer with the demonstration of epidemiological evidence for association of lung
bioactivation of compounds such as the drug aceta- cancer incidence with an Mspl polymorphism of
minophen^ ^"^ and the insecticide parathion^^^' ^^^. P450 1 Al (ref [129]). However, these results, from
Many studies have been done with P450 animal studies done with Japanese, have not been repro-
models, particularly using P450 inducers and ducible in Caucasians (refs [130-132]). Further, the
inhibitors and genetically modified mice, either nat- heterologously expressed human P450 lAl allelic
urally occurring or transgenic. These studies provide variant (V462I) showed only a relatively small
strong evidence that alterations in the activities of change in oxidation of the prototype polycyclic aro-
P450s can modify the sensitivity of mice to various matic hydrocarbon carcinogen benzo[a]pyrene
chemicals. For instance, the Ah locus (which con- diolepoxide^^^' ^^'*. A recent explanation to the
trols P450s lAl, 1A2, and IBl as well as some quandary comes from the work of Kamataki's
Phase II enzymes) can modify the sensitivity in Ah group, who have shown that P450 IBl, not P450
receptor-deficient mice, depending upon the chem- lAl, is the major P450 responsible for the aryl
ical and the organ site^^^. Effects of specific P450 hydrocarbon hydroxylation activity in lymphocytes
knockouts have been reported in transgenic mice as and that it is P450 IBl expression that shows the
well, for example, prevention of acetaminophen tox- classic trimodality, not P450 lAl, (ref [135]).
icity by deleting P450 2E1 (ref [118]) and of Today the field is such that the search for roles
7, 12-dimethylbenz[«]anthracene-induced lym- of a particular P450 in human disease follows a
phomas by deletmg P450 IBl (ref [119]). route similar to that just discussed for P450 lAl,
Despite the strong evidence for effects of vari- that is, the identification of SNPs is a basis for epi-
ability of P450 on chemical toxicity and cancer risk demiological associations with various maladies.
in animals and the knowledge that human P450 lev- This approach is commonly applied to the possible
els vary considerably (Figures 10.1, 10.2, 10.5, and roles of P450s in cancers at various organ sites. The
10.7), demonstrating relationships with human general concept is also utilized for other diseases
disease has been difficult. In the 1960s, the demon- and is the major basis for the Environmental
stration of the inducibility of aryl hydrocarbon Genome Project of the National Institute of
hydroxylase (thought to be what is now known as Environmental Health Sciences (which includes
P450 lAl) by Nebert and Gelboin^^o j ^ ^ ^^ ^^^^ many other gene candidates in addition to
investigations with human samples, particularly P450s)^^^. The positive aspects of this strategy are
peripheral blood cells. The work of Shaw and that we have an extensive knowledge base of allelic
Kellerman^^^' ^^^ suggested that the inducibility of variations of P450s (e.g., http://www.imm.ki.se/
aryl hydrocarbon hydroxylase (now recognized as CYPalleles/), sophisticated and very sensitive bio-
P450 1 Al and IBl under these conditions) is corre- logical tools, and the potential to noninvasively
lated with susceptibility of smokers to lung cancer. analyze large populations, at least in the case of
In the early work, this apparently genetic variability some diseases and P450s. On the negative side,
was trimodal. Subsequently, this phenomenon has the ability to rapidly screen for associations
proven difficult to study, in part due to technical without serious thought about chemical exposure
396 F. Peter Guengerich

levels has lead to many studies with little or only In the process of drug development, the induc-
marginal biological plausibility. Many association tion of P450 1 and P450 2B enzymes (in animals
studies have been difficult to repeat. An example or in human cell or reporter assays) has often been
in point is the reported association of attenuated considered an issue for potential toxicity'^^' ^^'.
lung cancer risk (of smokers) with the P450 2D6 The concern about induction is that the rodents
PM phenotype. Although the initial reports were may be likely to develop liver or other tumors in
quite exciting ^^^, subsequent studies yielded vari- cancer bioassays with these compounds, and any
able results, and meta analysis has not supported association between these inductions and human
an association^^^; moreover, no real experimental cancer is not established; for example, epileptics
support for a biological association was ever with long-term exposure to barbiturates and
found^^^. A recent review by Vineis*^^ concludes hydantoins have not been found to have more can-
that the risks of cancer due to genetics are consid- cer'^^. Likewise, the induction of P450 4A is an
erably less than those associated with smoking or indicator of peroxisomal proliferation, a phenom-
other environmental factors. enon associated with rodent liver tumors but prob-
What associations of P450 have been ade- ably not human'^^. Thus, induction of rodent
quately demonstrated? The list below is short and P450s has been shown to be a means of identify-
not intended to necessarily be totally inclusive, but ing types of potential rodent toxicity^ ^'^, some of
emphasizes some of the more positive associa- which may be relevant to humans, but should not
tions found to date. (The absence of several of the be used as evidence for adverse roles of these
steroid-oxidizing P450s is known to be debilitat- agents in humans.
ing [Table 10.3], but these are not treated here; see
the sections on individual P450s and ref [29].)
The possible association between P450 lAl and
lung cancer has already been discussed above; a 6. Individual Human P450
confounding factor may be expression of P450 Enzymes
IBl. Truncation of P450 IBl is associated with
glaucoma, for unknown reasons^^^; this defect has Each of the 57 human P450s will be covered
not been seen in the P450 IBl-knockout mice^'' ^ •^. here. Clearly much more information is available
Allelic variants in P450 IBl do not appear to have about some than others. Points to be covered with
major effects in the oxidation of carcinogens'"^^; each, when possible, include sites of expression
some differences in cancer risk have been reported and relative abundance, polymorphism and
in the epidemiology literature''^^' ''^^. P450 1A2 inducibility, substrates and reactions, knowledge
activity has been reported to be associated with of important residues and active site characteris-
colon cancer incidence, when the factors of N- tics, inhibitors, and clinical issues.
acetyltransferase and well-done meat intake are
considered''^^; an association has plausibility in
the activation of heterocyclic amines by P450 1A2 6.1. P450 1A1
(ref [146]). One of the strongest associations
6.1.1. Sites of Expression and
reported to date involves that of P450 2A6 with
lung cancer; the association is driven by the data
Abundance
obtained with individuals with the gene dele- The gene has seven exons, and the cDNA
tion'^^. A relationship is plausible due to the region is -^70% identical to that of the closest
demonstrated ability of P450 2A6 to activate relative, P450 1A2. P450 lAl is expressed in fetal
A^-nitrosamines (Table 10.4), and possibly via the liver but not at appreciable levels in adult
decreased smoke intake of null-type individuals ljygj.155-157 P45Q lAl can be induced in primary
due to impaired metabolism of nicotine'"^^ (see human hepatocyte cultures^^^. The dominance of
Section 6.4.6). Although many epidemiological P450 1A2 over 1 Al in vivo may be due to prefer-
studies have been done with SNPs of P450 2E1, ential induction of P450 1A2 > lAl at low doses
any putative changes in P450 2E1 phenotype have of inducers (a phenomenon established in rats^^^)
not been validated with in vivo assays and must be or the presence of factors in liver that are not
considered suspect^'*^. preserved in hepatocyte cultures.
Human P450s 397

P450 lAl is expressed in human lung and has to initiate transcription (Figure 10.6, with R = Ah
been partially purified^. A recent estimate of a receptor), R^ = ARNT, and L = TCDD or other
median level of P450 in human lung is 6.5 pmol/mg inducers. A number of details regarding this
microsomal protein (n = 7) and 16 pmol/mg micro- scheme remain to be elucidated, such as roles of
somal protein for smokers (n = 18)^^^. The varia- coactivators, whether an endogenous ligand exists
tion in levels of P450 lAl is very high and if so what it is, etc. The list of inducers
(> 100-fold)^' ^^^, as suggested from earlier work in reported from in vitro studies includes TCDD and
which only benzo[a]pyrene hydroxylation was used is quite long. The list of compounds for which
as an indicator^^^ in vivo evidence of induction is more limited, but
P450 lAl is also expressed in placenta^^^ and it is generally accepted that it includes cigarette
peripheral blood cells (lymphocytes, monocytes)^^^, smoke, heterocyclic amines, polychlorinated
and these tissues have been used in many studies. biphenyls^^^, and some drugs (e.g., omeprazole^^^).
Expression (at least at the mRNA level) has been
reported in a number of other extrahepatic tissues
including pancreas, thymus, prostate, small intes-
tine, colon, uterus, and mammary gland^^"^. 6.1.3. Substrates and Reactions
This enzyme was first explored in the context
of an aryl hydrocarbon hydroxylase, using fluo-
6.1.2. Regulation and Polymorphism rescence assays that measured primarily the
Polymorphism in the inducibility of benzo[fl] 3-hydroxylation of benzo[fl]pyrene^^^. (It should
P3^ene hydroxylation activity has attracted consid- be noted that the fluorescence assay also detects
erable interest following the reports of Shaw and other fluorescent products, for example, 9-hydroxy-
Kellerman^^^' ^^^ that the induction in lymphocytes benzo[a]pyrene, and that other P450s also cat-
of smokers can be associated with susceptibility alyze the 3-hydroxylation reaction, for example,
to lung cancer. The link to lung cancer has been P450 2C9 in human liver^^^.) Another classic
studied extensively but few general conclusions model reaction used for P450 lAl is 7-ethoxyre-
can be reached. Smoking clearly induces levels of sorufin O-deethylation^^^, i73 Human P450 lAl
lungP450 lAl (refs [124], [160], [165]). Some epi- oxidizes benzo[a]p5n-ene to a variety of prod-
demiological investigations link the *2A (Mspl) ucts^^^' ^'^^. Many other polycyclic hydrocarbons
and *2B (I462V) polymorphisms to lung cancer are substrates for P450 1 Al and have been studied
incidence in Japanese^^^, but this association extensively^^^' ^'^'^. Some heterocyclic and aro-
has not been reproducible in other studies with matic amines can also be activated by P450 lAl
Caucasians^^^' ^^^ These two alleles are in linkage (ref. [178]). P450 lAl does not appear to play a
disequilibrium^ ^^. Two studies with recombinant major role in the metabolism of many drugs, pos-
human P450 lAl have not shown a major differ- sibly because of its locations of expression.
ence in any catal3^ic activities due to the substitu-
tion at codon 462 (refs [133], [134]). Although
there is a general consensus that phenotypic
variation in the inducibility of P450 lAl is 6.1.4. Knowledge about Active Site
observed, extensive searches have not associated Relatively little is known about the active site of
the inducibility with any known polymorphisms in P450 lAl. Early work on pharmacophore models
the P450 lAl, Ah receptor, or arylhydrocarbon for rat P450 1 Al was done by Jerina's group^^^. The
nuclear translocator (ARNT) genes ^^^' ^^^. early modeling of substrates and inhibitors sug-
The induction of P450 lAl has been studied gested that P450 lAl ligands were relatively
extensively and is discussed elsewhere in this planar. Some homology modeling has been done by
book^^^. Briefly, the Ah receptor resides in the Lewis^^^, although little has been done with site-
cytosol and, when activated by binding of an appro- directed mutagenesis on the roles of individual
priate agonist, loses the accessory protein Hsp90 amino acids. The lack of effect of interchanging
and dimerizes with the ARNT protein, moving to Val and Leu at position 462 has already been
the nucleus and interacting with an XRE element mentioned^^^' ^^^.
398 F. Peter Guengerich

6.1.5. Inhibitors 6.2.2. Regulation and Polymorphism


Despite the long interest in this enzyme, the The variability and inducibility of P450 1A2
hst of inhibitors is relatively short, and many have been recognized for some time, indirectly,
inhibitors show overlap with P450s 1A2 and IBl going back to studies on phenacetin metabolism
(ref. [181]). For instance, aNF is often used as by Conney and his associates ^^'^. The characteriza-
an inhibitor but is more effective against P450 tion of P450 1A2 CT450p/') as the low K^
1A2 (refs [181], [182]). Another inhibitor is ellip- phenacetin 0-deethylase^ led to some interpreta-
ticine^"^. 1 -(1' -Prop)aiyl)pyrene and 2-( 1 -propynyl) tion of the earlier results. P450 1A2 was shown to
phenanthrene were found to be selective P450 be the caffeine iV^-demethylase^^, and the 40-fold
1 Al inhibitors when compared with human P450s variation in levels of liver P450 1A2 is reflected in
l A 2 a n d l B l (ref [181]). the 40-fold variation in some in vivo parameters of
caffeine metabolism^^, some of Vesells's earlier
work on the metabolism of antipyrine in twins
6.1.6. Clinical Issues suggests a role for genetic polymorphism in P450
Due to a rather limited role of P450 lAl in 1A2 activity^^^, and a more recent twin study
drug metabolism, there are no real pharmacoki- confirms the strong genetic component of caf-
netic issues. The issue with P450 1 Al is induction feine demethylation^^^. However, elucidating
and a possible role in chemical carcinogenesis. details of any functional polymorphism has been
Work with animal models shows that P450 lAl difficult.
inducers can be co-carcinogens^^' ^^^. Thus, regu- At least 13 allelic variants of P450 1A2 are
latory agencies tend to look unfavorably at induc- now known^^. Of these, at least five have changes
tion of P450 lAl by potential drugs in animal in the coding sequences that cause amino acid
models. However, the point should be made that changes. Recent work in this laboratory with the
there is presently little experimental or epidemio- expressed coding region variants indicates that
logical evidence to support this hypothesis, and most do not differ more than 2-fold in their kinetic
Ah inducers can afford protection from cancer in parameters for several assays (phenacetin O-
some animal models^^^. deethylation and A^-hydroxylation of heterocyclic
amines), although one of the variants (R431W)
did not express holoprotein in Escherichia
coli^^^^. Some (*1C and *1F) have been proposed
6.2. P4501A2 to modify levels of expression^^. However, the
basis for a polymorphism is not clear. One view is
6.2.1. Sites of Expression and
that the variability is not genetic, based upon the
Abundance
lack of modality breaks in analysis of some in vivo
As mentioned earlier, human P450s lAl and parameters of caffeine metabolism^^^.
1A2 both have seven exons and 70% sequence One complication with genetic polymorphism,
identity in their coding regions. Both these genes as with P450 lAl (vide supra), is the inducibility.
show similar patterns of regulation by the Ah Because of the availability of markers of hepatic
receptor system, but P450 1A2 is essentially P450 1A2 function (phenacetin is no longer used
expressed only in the liver^^'^, probably due to the but caffeine and theophylline are), demonstrating
involvement of HNF in its regulation (vide infra). in vivo changes in P450 is relatively easy to do and
Several lines of evidence indicate that the level of the effects are consistently seen, at least quantita-
expression is substantial (Figures 10.1 and 10.2, tively. The mechanism of induction appears to be
Table 10.5), - 1 0 - 1 5 % of the total P450, on the similar to that of P450 lAl (Figure 10.6), with
average, with levels varying ~40-fold among expression restricted to the liver because of the
individuals (Figure 10.4). need for HNF (ref [188]). An interesting observa-
Occasional reports cite mRNA expression in tion made recently in mice is that the inducer
some extrahepatic tissues, for example, colon^^^. 3-methylcholanthrene causes a persistent induc-
Extensive searches have not found expression in tion (of P450 lAl) in liver, lasting beyond the
human lung^^"^. time suggested by pharmacokinetic expectations ^^^.
Human P450s 399

One interpretation is that a P450 lA2-generated other P450s, e.g., 3A4 (ref [203])). Induction of
metabolite is involved. Further details and any P450 1A2 and 2-hydroxylation has been proposed
relevance to humans remain to be established. as a means of preventing oxidation of 17(3-
With animal P450 1A2, one mechanism of induc- estradiol to the potentially more reactive 4- and
tion involves protein stabilization, for example, 16a-hydroxy products^^"^' ^^^.
by isosafrole-derived products *^^. Whether or not
this mechanism is relevant in humans is unknown.
Reported inducers include cigarette smoking, 6.2.4. Knowledge about Active Site
charbroiled food (presumably via polycyclic
hydrocarbons and heterocyclic amines), crucifer- Considerable site-directed mutagenesis work
ous vegetables, vigorous exercise^^\ and the drug has been done with rat P450 1A2 (refs [206-208])
omeprazole (actually a metabolite) ^^^. but relatively little with human P450 1A2. Some
pharmacophore^^^ and homology^^^' •^^^ modeling
work has been reported.
6.2.3. Substrates and Reactions An approach was developed in this laboratory
for doing random mutagenesis of P450 1A2, uti-
P450 1A2 has been expressed in a number of
lizing changes in the rates of formation of muta-
systems and is used in analyses of catalytic selec-
genic hydroxylamines^^^ Changing Glu226 (to He
tivity. Of the P450s, this has one of the highest
or Gin) had the effect of increasing rates of
levels of expression in bacterial systems^^^' ^^^.
phenacetin 0-deethylation 8-fold (effect on ^cat)"^^^-
The list of drug substrates is long^^, and only a
Also of interest is a 100-fold decrease of activity
few of the more well-known reactions are listed in
seen in mutation of the neighboring Phe225 (to He
Table 10.7.
or Tyr)^^^. The mutation of Asp320 (to Ala) also
Many carcinogens are substrates, particularly
decreased activity^^^. Kinetic deuterium isotope
aromatic and heterocyclic amines (Table 10.4).
effect studies suggest little change in rate-limiting
Other carcinogens shown to be substrates include
steps of the 0-dealkylation reaction over a wide
polycyclic hydrocarbons, nitropolycyclic hydro-
range of activities with these mutants, and the
carbons, and some A^-nitrosamines^^^.
mutations do not affect ground-state substrate
The only major endogenous substrates are 17p- binding or rates of P450 reduction^^^. The effects
estradiol and estrone (2-hydroxylation). The phys- of these substitutions have not been interpreted in
iological relevance of this reaction is unknown, terms of specific roles.
particularly because of the wide variation in levels
The issue of cooperativity will be discussed
of P450 1A2 (this reaction is also catalyzed by
later under P450 3A4. Cooperativity has not been
reported for human P450 1A2 but behavior of the
rabbit ortholog has been interpreted in the context
Table 10.7. Some Drug Substrates for Human of multiple, overlapping binding sites^^"^.
P450 1A2^

Drug^ References
6.2.5. Inhibitors
Acetaminophen (3') 195 Several human P450 1A2 inhibitors are known
Antipyrine (4,3-methyl) 196 from clinical work, including fiirafylline (mecha-
Bufuralol(l,4) 197 nism based)^^^ and fuvoxamine. aNF is a
Caffeine (3) 73 commercially available and strong inhibitor of
Clozapine 49
human P450 1A2 (K- ~ 6 nM)^^^ for in vitro work.
Olanzapine 49
Ondansetron (7,8) 198
A number of polycyclic acetylenes are potent
Phenacetin 7 inhibitors of P450 1A2 (ref [181]). With rat
Tacrine 199, 200 P450 1A2, 2,3,7,8-tetrachloro[/?]dibenzodioxin
Theophylline (1,3,8) 201 and some polyhalogenated biphenyls are strong
inhibitors, but these studies have not been extended
"See also Rendic^^. to human P450 1A2 (ref [216]).
400 F. Peter Guengerich

6.2.6. Clinical Issues the levels are of an order of magnitude lower than
for P450 lAl (ref [160]). These low values may
Some drug interactions have been reported. An explain the lack of immunostaining in (nontumor)
older example is that of low activity toward tissues reported by Murray et al?^^ Specific val-
phenacetin favoring a potentially toxic secondary ues for levels of expression in tissues other than
pathway, deacetylation followed by quinoneimine lung have not been published. Recently Chang
formation and methemoglobinemia^^^. Furafylline
et alP^ found traces of P450 IBl mRNA in
was a drug candidate but was never developed
human liver using real-time PCR, but the protein
because of its strong P450 1A2 inhibition and
was undetectable within the limit of sensitivity.
interference with caffeine metabolism^^^. High
levels of P450 1A2 activity have also been associ-
ated with ineffectiveness of theophylline therapy
(forasthma)2i8,2i9
6.3.2. Regulation and Polymorphism
The other concern about P450 1A2 is the same Levels of P450 IBl in human lung vary by at
discussed earlier for P450 1 Al, the co-carcinogenic least one order of magnitude ^^^. An interesting
effect. In this regard, there is some epidemiological observation is that a termination variant of P450
evidence that high P450 1A2 activity (measured as IBl is strongly associated with glaucoma^"^^' ^^^.
in vivo caffeine metabolism) is associated with A similar phenotype has not been seen in P450
enhanced risk of colon cancer, although the effect IBl-knockout mice^^^. Other polymorphisms of
was not seen in the absence of high A^-acetyltrans- (human) P450 IBl are known and are predomi-
ferase activity and high consumption of charbroiled nant in a set of haplotypes involving four varia-
meat^"^^. tions Arg/Gly 48, Ala/Ser 119, Val/Leu 432, and
Asn/Ser 453. Assays involving the metabolism of
17p-estradiol and polycyclic hydrocarbons by
6.3. P450 1B1 recombinant P450 IBl variants show some varia-
tions but have not been particularly dramatic
6.3.1. Sites of Expression and (reviewed by Shimada et al.^"^^).
Abundance In vitro experiments show the inducibility of
P450 IBl in patterns expected for an^/?-responsive
P450 IBl was originally discovered in ker-
gene, which is the way in which the gene was
atinocyte cultures in a search for new dioxin-
found^^. Unlike P450 lAl and particularly 1A2
inducible genes^^ and in work on adrenals in
(vide supra), there is limited direct evidence for
animal models^^^. In contrast to P450 lAl and
inducibility of P450 IBl in vivo because of the low,
1A2 (seven exons), the P450 IBl gene has only
extrahepatic expression and the lack of a diagnostic
three exons and is located on chromosome 2
probe drug. Although the expression of P450 IBl is
instead of 15 (ref [221]). Although most of the
driven by the Ah system, additional factors must be
detailed studies of tissue-specific expression have
involved because of the known tissue and cell line
been done at the mRNA level and not protein,
selectivity of expression. For instance, major differ-
strong responses are seen in fetal kidney, heart,
ences are seen between HepG2, MCF-7, and ACHN
and brain, in that order^^^. In adults (human), there
cells (of liver, breast, and kidney tumor origins,
is little detectable expression in liver, but there is
respectively)^^ ^ With the information available
more detectable expression in kidney, spleen, thy-
today, one would expect the gene to be induced (in
mus, prostate, lung, ovary, small intestine, colon,
extrahepatic tissues) by the compounds that induce
uterus, and mammary gland^^^. Many of these tis-
P450s lAl and 1A2.
sues are of particular interest because of the
tumors that develop there. Immunochemical stain-
ing of P450 IBl has been reported in a variety of
malignant tumors^^^. 6.3.3. Substrates and Reactions
The level of expression (of the protein) in Human P450 IBl, like P450 lAl, has never
human lung has been estimated to be at the level been purified from tissue and all of our informa-
of ~ 1 pmol/mg microsomal protein in nonsmokers tion has come from protein expressed in heterolo-
and 2-4 pmol/mg microsomal protein in smokers. gous systems. 7-Ethoxyresorufin O-deethylation
Human P450s 401

can be used as a model reaction^^^. The catalytic from 7,12-dimethylbenz[a]anthracene is of partic-


activity of P450 IBl is intermediate between ular importance^ ^^.
P450s lAl and 1A2 (ref. [181]). Some other One of the interesting findings with human
model reactions can be used as welP^^. P450 IBl is that this enzyme is an efficient cata-
Much of the interest in P450 IBl has been lyst of np-estradiol hydroxylation and that the
because of its ability to activate a broad spectrum pattern is for 4- > 2-hydroxylation^^^' ^^^' ^^^. This
of chemical carcinogens, including polycyclic pattern is the opposite of that seen for P450s 1A2
hydrocarbons and their oxygenated derivatives, and 3A4 (2- > 4-hydroxylation)^^^' ^^^ and is of
heterocyclic amines, aromatic amines, and significance because 4-hydroxyestradiol is chemi-
nitropolycyclic hydrocarbons^^^ (Table 10.8). cally more reactive with oxygen and more likely
Of particular interest is the observation that to oxidize (to 0-quinone) and bind DNA^^"^. Thus,
human P450 IBl is at least as active as P450 lAl 4-hydroxyestrogens are considered to be candi-
in the conversion of the classic carcinogen dates for causing estrogen-dependent tumors^^^.
benzo[«]pyrene to the 7,8-dihydrodiol, the first The available information indicates that mouse
step in the formation of the diol epoxide^^^. P450 IBl does not catalyze estrogen hydroxyl-
In general, it would appear from the available ation^^ ^' ^^^, providing a potentially important
information that the rodent P450 IBl cnzyrnQS difference with the human enzyme. This apparent
have similar catalytic specificity as human P450 lack of conservation of selectivity has relevance in
toward carcinogens, from the available informa- the use of mouse (and rat) models in some of the
tion^^ ^ If this is a valid view, then the observation biology, for example, the human glaucoma
that P450 IBl-knockout mice do not form tumors mentioned earlier^'*^' ^^^.

Table 10.8. Carcinogens Activated by Human P450 IBl

Substrate References Substrate References

Polycyclic aromatic hydrocarbons IQ 178


Benzo[<a!]pyrene 181 Trp-Pl ' 178
Benzo[fl]pyrene-4,5-diol 178 Trp-P2 178
(+) Benzo[fl]pyrene-7,8-diol 178 PhIP 178
( - ) Benzo[«]pyrene-7,8-diol 178
Aromatic amines
Dibenzo[fl, /]pyrene 226
2-Aminoanthracene 178
Dibenzo[fl, /jpyrene-11,12-diol 178
2 - Aminofluorene 178
Benz[a]anthracene 181
4-Aminobiphenyl 178
Benz[a]anthracene-1,2-diol 178
3-Methoxy-4-aminoazobenzene 178
Benz[Gf]anthracene-c/5-5,6-diol 178
O-Aminoazotoluene 178
7,12-Dimethylbenz [a] anthracene 178
6-Aminochrysene 178
7,12-Dimethylbenz[fl]anthracene-3,4-diol 178
Benzo[c]phenanthrene-3,4-diol 178 Nitropolycyclic hydrocarbons
Fluoranthene-2,3-diol 178 1-Nitropyrene 227
Benzo[6]fluoranthene-9,10-diol 178 2-Nitropyrene 178
Chrysene-1,2-diol 178 6-Nitrochrysene 178
5 -Methylchry sene 226 2-Nitrofluoranthene 227
5-Methylchrysene-1,2-diol 178 3 -Nitrofluoranthene 227
5,6-Dimethylchrysene-1,2-diol 178 6-Nitrobenzo [ajpyrene 227
Benzo[g] chrysene-11,12-diol 178 1,8-Dinitropyrene 227
6-Aminochrysene-1,2-diol 178 1-Aminopyrene 227
Heterocyclic amines Estrogens
MelQ 178 np-Estradiol 228
MelQx 178 Estrone 229
402 F. Peter Guengerich

6.3.4. Knowledge of Active Site light of the number of carcinogens that P450 IBl
activates (Table 10.8). The issue of oxidation of
Very little knowledge about the active site is estrogens to reactive products is one worth con-
available. The general pattern of catalytic speci- sidering, in light of the experimental evidence
ficity, with similarity to P450 lAl and 1A2, supporting a link with cancer in estrogen-
would argue for some similarity. The effects of the dependent tumors. Another matter that has not
allelic variants are probably not strong enough to been addressed is the possible metabolism of the
be of much use in understanding the effects of various estrogens in postmenopausal hormone
those residues ^'^^. Some homology modeling has treatments (e.g., Premarin® by P450 IBl, e.g., see
been done^^^. refs 234 and 241 regarding DNA adducts formed
by some of these estrogens).
6.3.5. Inhibitors
aNF is a strong inhibitor, as in the case of 6.4. P450 2A6
P450 1A2 (ref [181]). Some acetylenes devel-
oped by Alworth's group have been found to selec-
6.4.1. Sites of Expression and
tively inhibit P450 IBl (at least relative to P450s Abundance
lAl and 1A2), including 2-ethynylpyrene^^^ A P450 2A6 (formerly termed IIA3 and 2A322)
potential drawback to these compounds is that was purified from human liver microsomes^ and a
they are rapidly oxidized by P450 IBl. cDNA was isolated from a human liver library^"^^.
Resveratrol is a polyphenol found in red The protein is expressed at medium to low levels
grapes and has been of interest in the context of its in liver (Table 10.5, Figure 10.2). In one study, the
potential to inhibit cancer^^^. This compound is a fraction of total human liver P450 attributed to
noncompetitive inhibitor of P450 IBl, with a K^ P450 2A6 ranged from <0.2% to 13% among
value of 23 |LJLM in model systems^^^ (with selec- individual samples, with a mean of —4%^^. P450
tivity toward P450 lAl). Recently, Potter et alP^ 2A6 was not found in placenta (frill term)^"^^.
reported that P450 IBl oxidizes resveratrol to the P450 2A6 is also expressed in other tissues,
known anticancer agent piceatannol, a tyrosine particularly in the nasopharyngeal region.
kinase inhibitor. Further studies showed that the Expression has been detected in nasal mucosa,
natural product rhapontigenin is a low K^ inhibitor trachea, lung^"*"^, and esophageal mucosa^"*^. These
of P450 lAl (ref [240]). A series of methoxy- sites of expression are of interest regarding certain
substituted trans stilbene compounds of the cancers. In liver cancers, overexpression of P450
resveratrol/rhapontigenin family were prepared 2A6 protein was associated with chronic inflam-
and tested: of these, 2,4,3',5'-tetramethoxystil- mation and cirrhosis^^^.
bene was found to be a strong and selective com-
petitive inhibitor of P450 IBl (K. = 3 nM) and
resisted demethylation^"^^. 6.4.2. Regulation and Polymorphism
The regulation of P450 2A6 expression has
been studied in primary cultures of human hepato-
6.3.6. Clinical Issues
cytes. Expression (mRNA, protein) is inducible by
No issues regarding drug interactions have rifampicin^"^^ and phenobarbitaF"*^ and, to a lesser
been raised. As with the P450 lA subfamily extent, clofibrate, cobalt, griseofiilvin, and pyra-
enzymes, an issue is that induction of P450 IBl zole^"*^. The nuclear receptor HNF-4 is involved in
might increase the activation of procarcinogens. the expression of cultured hepatocytes^'*^.
This issue may be real, although presently there is Many polymorphisms (> 11) are known for the
no epidemiological evidence to support such a CYP2A6 gene^^. These include a splice variant
relationship. Although the coding region polymor- (* 12) in which CYP2A 7 exons are included and the
phisms have only indicated a limited potential protein has lost catalytic activity^^^' ^^^ Another
for contribution to cancer (vide supra), the recent SNP polymorphism (*2), recognized earlier, is the
evidence for trimodal induction^^^ is certainly of L160H change which yields very low catalytic
interest (see Section 5, vide supra), particularly in activity^^^. Also of interest is a gene deletion (*4).
Human P450s 403

The incidence of these polymorphisms is racially (SM-12502)277, 278 and tegafur^^^, 28o^ ^j^ich is
linked^^. P450 2A6 is involved in nicotine oxida- converted to 5-fluorouracil. Halothane is reduc-
tion, and in 1998, Tyndale and her associates tively converted to a free radical by P450 2A6,
reported that individuals with low P450 2A6 activ- which can yield at least two products and initiate
ity smoke less and might have lower cancer risk^"^^. lipid peroxidation^^ ^
This proposal seems reasonable but the findings Some of the catalytic selectivity of P450 2A6
have been questioned. General agreement exists overlaps with that of P450 2E1 {vide infra). One
that defective P450 2A6 genes cause reduced nico- area in which the overlap has been noted is in the
tine metabolism (the presumed basis for reduced oxidation of nitrosamines. P450 2A6 preferen-
smoking)^^^"^^^. Several reports conclude that tially catalyzes the oxidation (and activation) of
deficient P450 2A6 reduces smoking^^^"^^^ and A^-nitro- sodiethylamine, in contrast to P450 2E1
also lung cancer^'^^' ^^^' ^^^ in smokers. The latter which preferentially oxidizes A^-nitrosodimethy-
hypothesis has biological plausibility because lamine^^^' ^^^. P450 2A6 is also involved in the
many carcinogens from tobacco are activated by oxidation of many tobacco-specific nitrosamines,
P450 2A6 (Table 10.4 and vide infra). However, including 4-(methylnitrosamino)-1 -(3 -pyridyl)-
other studies have not revealed any relationship 1-butanone (NNK)283-286 P45Q 2A6 appears
between CYP2A6 genotype and smoking; cancer is to be the major human P450 involved in the
also controversiaP^^"^^^. Some of the discrepan- activation of iV-nitroso-benzylmethylamine^^^,
cies may be raciaP^^ but even this is unclear^^^. iV-nitrosodipropylamine, A^-nitrosobutylamine,
Some problems are attributed to technical short- A^-nitrosophenyl- methylamine, and iV-nitrosonor-
comings in genotype analyses^^^ and a definite nicotine^^^. Fujita and Kamataki^^^ studied the
relationship is still lacking^^^ in Caucasians, but is bacterial mutagenicity of a number of tobacco-
more likely in Asians^^^, where the incidence of specific iV-nitrosamines and concluded that P450
gene deletion is higher. 2A6 is the major human enzyme involved in the
activation of all examined.
6.4.3. Substrates and Reactions P450 2A6 is also involved in the metabolism
of nicotine {vide supra). P450 2A6 is the main cat-
The most characteristic and specific reaction alyst in the oxidation of nicotine to cotinine^^^"^^^.
of P450 2A6 is coumarin 7-hydroxylation^' ^^^. P450 2A6 is also involved in the 3'-hydroxylation
Coumarin 7-hydroxylation has also been used as of cotinine^^^. In addition, P450 2A6 catalyzes 2'-
an in vivo diagnostic assay^^^"^^^. hydroxylation of nicotine, yielding a precursor of
One issue with P450 2A6 is whether b^ is a lung carcinogen^^"^.
required for optimal catalytic activity. Soucek-^^^ P450 2A6 can also A^-demethylate hexa-
demonstrated that a 1:1 ratio of b^ to P450 was methylphosphoramide^^^.
optimal in coumarin 7-hydroxylation catalyzed by Several forms of human P450 catalyze the
the purified recombinant enzyme. The effect of Z?^ 3-hydroxylation of indole^^^, and the product
on catalytic selectivity has not been evaluated in dimerizes to indigo. P450 2A6 was the most active
all reports on P450 2A6. human P450 identified for this activity and could
Coumarin 7-hydroxylation can be used in vivo also catalyze several oxidations of indole^^^.
with humans as a phenotypic assay. An alternative Mutants of P450 2A6 generated from a random-
procedure is to administer caffeine to individuals ized library were shown to catalyze the oxidation
and determine the conversion of 1,7-dimethyl- of several substituted indoles to generate variously
xanthine to 1,7-dimethyluric acid, a reaction cat- colored indigos and indirubins^^^.
alyzed by P450 2A6 (ref [274]).
Some industrial chemicals are substrates for
oxidation by P450 2A6, including alkoxyethers
6.4.4. Knowledge about Active Site
(used as fuel additives, e.g., tert-h\xty\ methyl
ether)^^^ and the vinyl monomer 1,3-butadiene, Relatively little site-directed mutagenesis work
a cancer suspect^^^. has been done on P450 2A6 (although the rodent
Some drugs are also substrates, including (+) P450 2A enzymes were an early target for the
c/5-3,5-dimethyl-2-(3-pyridyl)thiazolidin-4-one approach)^^^. Some information has been gleaned
404 F. Peter Guengerich

from naturally occurring SNPs. In vivo studies are in Asians^^^ but remain controversial in
consistent with a shift from coumarin 7-hydroxyla- Caucasians266,268,269,3i5,3i6^
tion to 3-hydroxylation associated with the L160H Some drugs are P450 substrates, although the
allele^^^, although this phenomenon has not been relative contribution of P450 2A6 is still so small
verified in vitro. The substitution R128Q yields a (Figure 10.3) that P450 2A6 reactions are gener-
protein with half the content of heme, but the ally not included in screens.
inability to bind carbon monoxide (to ferrous iron) P450 2A6 expression has been reported to be
(plus a loss of 98% of the coumarin 7-hydroxyla- induced during infection by (carcinogenic) liver
tion activity)^^^. flukes^ ^^ and downregulated during infection by
Lewis has published several homology models hepatitis A virus^^^.
of P450 2A6 (refs [300-302]) and also attempted
to rationalize the pattern of nicotine oxidation
using molecular orbital calculations^^^. 6-5. P450 2A7
The situation involving the CYP2A7 gene is
6.4.5. Inhibitors complex, and sometimes this has even been
erroneously referred to as a pseudogene^^. Two
Several selective inhibitors of P450 2A6 are
pseudogenes {CYP2A7PTX md CYP2A7PCX) are
known. Diethyldithiocarbamate appears to be a
known. The P450 2A7 mRNA transcript is pro-
mechanism-based inactivator, although the inactiva-
duced in human liver, at roughly the same level as
tion has not been extensively characterized^^^.
that for P450 2A6 (ref [250], [319]). Gonzalez's
Diethyldithiocarbamate and its oxidized form,
laboratory had isolated cDNA clones now recog-
disulfiram, also inhibit P450 2E1 (ref [304]).
nized as 2A6, the 2D6 variant LI60H, and 2A7, and
In vivo single-dose treatment of people with disulfi-
expressed all three in HepG2 cells^"*^. Of the three,
ram inhibits P450 2E1 but not P450 2A6 (ref [305]).
only the "wild type" P450 2A6 incorporated heme.
The vegetable watercress, a source of phenethyl
Others have also expressed P450 2A7 in heterolo-
isothiocyanate, did not inhibit P450 2A6 in vivo^^^.
gous systems but not reported any evidence of a cat-
A number of chemicals have been tested as
alytically active P450 2A7 holoprotein^^^. Whether
inhibitors of P450 2A6 in human liver micro-
or not a fiinctional P450 2A7 is transcribed from
somes^^'^. Of these, the most selective and potent
the mRNA in human tissues is still unclear, and
inhibitors appear to be 8-methoxypsoralen, tranyl-
nothing can be said about catalytic activity.
cypromine, and tryptamine, with K^ values
~ 1 JULM^^^"^^^. The inhibition by the natural prod- Gene conversion events between the CYP2A6
uct 8-methoxypsoralen (in many foods) is mecha- and CYP2A7 genes have been reported, yielding
nism based^^^. 8-Methoxypsoralen (methoxysalen) chimeric proteins in humans^^^' ^^^' ^^^. These pro-
inhibits P450 2A6 in vivo^^^ and has also been teins have some of the coumarin 7-hydroxylation
reported to decrease nicotine metabolism in smok- conferred by the 2A6 component^^^
ers^ ^^ Both of the inhibitors 8- and 5-methoxypso-
ralen were covalently bound to P450 2A6 during
incubation with NADPH^^^. Menthofiiran, another 6.6. P450 2A13
natural product, is also a mechanism-based inacti-
6.6.1. Sites of Expression and
vator of P450 2A6 (ref [313]).
Abundance
Isoniazid has been reported to be a weak
mechanism-based inactivator of P450 2A6 The CYP2A13 gene has been recognized for
(ref [314]). some time^^^ The gene is expressed in human
liver^^^' ^^^ and several other extrahepatic tissues,
including nasal mucosa, lung, trachea, brain, mam-
6.4.6. Clinical Issues mary gland, prostate, testis, and uterus^^^. The
As indicated in Section 6.4.2, the major highest level seems to be in nasal mucosa^^^, which
issue regarding P450 2A6 polymorphisms is is of interest in the context of tobacco-related can-
the effects on lung and esophageal cancers and cers because of some of the catalytic activities
smoking habits, which have good epidemiology toward nitrosamine substrates {vide infra).
Human P450s 405

6.6.2. Regulation and Polymorphism Much of the early work with P450s in experi-
mental animals was focused on the phenobarbital-
Little is known about the regulation and inducible enzymes now recognized to be in the 2B
inducibility of the CYP2A13 gene. Several variant subfamily^^^' ^^^ and a general expectation was
alleles have been identified, including one in the that similar P450s would be prominent in human
coding region (R257C) with somewhat less activ-
liver (and further suggested by immunochemical
ity toward NNK^23
studies^ and early cloning work^^^). However, the
major P450 in human liver (and small intestine)
proved to be P450 3A4 (Figures 10.2 and 10.3).
6.6.3. Substrates and Reactions The mean level of P450 2B6 in human liver has
Recombinant P450 2A13 has much lower been somewhat controversial. One of the prob-
coumarin 7-hydroxylation activity than does P450 lems has been antibody specificity. Antibodies
2A6, but coumarin is also converted to the 3,4- raised against rat P450 2B1 have not been very
epoxide^^"^. P450 2A13 also catalyzes several reac- specific^^^; unfortunately many papers in this area
tions at rates as high or higher than P450 2A6, show only limited sections of gels or actually
including 2,6-dichlorobenzonitrile activation, iV-nitro- show major cross-reactive material. The results
sodiethylamine 7V-deethylation, hexamethylphos- tend to fall into two groups. One set reports levels
phoramide A/-demethylation, 7V,7V-dimethylaniline very low to 80 pmol P450 2B6 per milligram pro-
A/-demethylation, 2'-methoxyacetophenone O-deme- ^gjj^330-332 Another set of reports range from near
thylation, A^-nitrosomethylphenylamine A/-demethy- zero levels to 28 pmol P450 2B6/mg microsomal
lation, and the activation of NNK^^^. The latter protein^^^' ^33-336 However, the mean values differ
reaction is of particular interest with regard to considerably in both the former and latter groups.
tobacco-related cancer because of the localization of While some of the discrepancy may be attributa-
expression of this P450 in nasal mucosa. ble to the differences in liver samples, the main
difference is probably with the antibodies used
and cross-reactivity with other proteins, as well as
6.6.4. Knowledge about Active Site error inherent in other aspects of immunochemi-
cal methods. Our own work is in line with the
No information is presently available beyond lower set of estimates of expression levels (mean
an effect of the R257C variant allele^^s ~ 1 % of total P450, with values rarely exceeding
5% even in samples from individuals administered
inducers)^^^. This level is an order of magnitude
6.6.5. Inhibitors less than for P450 3A4 (Figures 10.2 and 10.4).
No inhibitors have been reported.

6.6.6. Clinical Issues 6.7.2. Regulation and Polymorphism


Until recently, the mechanisms of induction by
P450 2A13 probably does not make a major
barbiturates had been rather vague in humans and
contribution to the metabolism of drugs. The
experimental animals. Studies with HepG2 cells
major interest in P450 2A13 involves a possible
(derived from hepatocytes) show the role of the
role in chemical carcinogenesis^^^.
constitutive androstane receptor (CAR), a member
of the steroid receptor superfamily, and its interac-
tion with the phenobarbital-responsive enhancer
6.7. P450 2B6 module (PBREM) in the region between —1733
and —1683 bp in the 5' flanking region^^^. Other
6.7.1. Sites of Expression and
work with HepG2 cells has implicated the liver-
Abundance
selective transcription factor C/EBPa^^^. Kliewer's
P450 2B6 is expressed primarily in liver, and group^^^ also demonstrated the involvement of
the protein has been partially purified^^^. The pro- another previously orphan receptor, pregnane X
tein has also been detected in human lung^^^. receptor (PXR), in binding to PBREM in primary
406 F. Peter Guengerich

human hepatocytes to induce P450 2B6. PXR is and propofol hydroxylation^^^. The iV-demethylation
active only when hgand-activated but CAR appar- of (iS)-mephenytoin has been used as a marker of
ently acts without an added ligand; both CAR and P450 2B6 in vitro (microsomes)^^^' ^^^' ^^°.
PXR heterodimerize with (liganded) RXR^^^. However, a valid in vivo probe for P450 2B6 is
"Cross-talk" also exists at the PBREM site with the still lacking354,360,361
vitamin D receptor as well as CAR and PXR^"*^' ^^^. As with animal P450 2B enzymes, P450 2B6
The levels of CAR and PXR mRNA in individual can also oxidize some environmental pollutants^^^.
human livers are correlated with the level of P450 Nonhyperbolic kinetics have been reported for
2B6 mRNA^^l The regulation of P450 2B6 has some P450 2B6-catalyzed reactions but these have
considerable similarity to that of P450 3A4 (vide not been extensively characterized^^^.
infra), with some differences. Several recent find-
ings provide some further insight into the mecha-
nism, although several questions persist. CAR 6.7.4. Knowledge about Active Site
does have ligand-activated effects and 6-(4-
Several homology models of P450 2B6 have
chlorophenyl)imidazo[2,l-Z?][l,3]thiazole-5-
been published^^^' ^^'^, including one using molec-
carbaldehyde 0-(3,4-dichlorobenzyl)oxime has
ular dynamics^^^.
been identified as an agonist^^"^. A novel distal
Relatively little site-directed mutagenesis has
enhancer regulated by PXR and CAR has been
been done with P450 2B6. Halpert's laboratory
identified in the CYP2B6 gene^^^.
modified 10 residues and measured some activities,
Alternative splicing in the CYP2B6 gene was although most of the changes were <2-fold^^^.
already identified in 1990^^^, with use of a cryptic Recently, Halpert's laboratory has solved a crys-
exon within introns 3 and a splice site acceptor tal structure of a derivative of the related rabbit
within exon 4. Extensive polymorphism (mainly P450 2B4 (ref [366a]), which should be of rele-
SNPs) has been identified in the CYP2B6 vance in understanding P450 2B6.
gene^^' ^^^' ^^^. Several of the mutants appear to
yield attenuated levels of protein and catalytic
activity^'^^. Some evidence for enhanced catalytic 6.7.5. Inhibitors
activity of a P450 2B6 SNP variant (N172H) has
been reported (~ 2-fold) and attributed to the Lists of the reported inhibitors of P450 2B6
homotropic activation seen in 7-ethoxycoumarin have been compiled by Rendic^^. Orphenadine had
O-deethylation^"*^. been utilized in some work with microsomes but
does not appear to be particularly selective^^^' ^^^.
More recently 2-isopropenyl-2-methyladamantane
and 3-isopropenyl-3-methyldiamantane have been
6.7.3. Substrates and Reactions reported as selective inhibitors of P450 2B6 (ref
Many reactions have now been demonstrated [368]). Triethylenethiophosphoramide has also
to be catalyzed by recombinant P450 2B6, mirror- been reported to be a selective inhibitor of P450
ing the early research in the P450 field with rat 2B6 (ref [369]).
P450 2B1 and rabbit P450 2B4 (refs [350-353]). The oral contraceptive 17a-ethynylestradiol
However, this information must be considered in is a mechanism-based inactivator of P450 2B6
the context of the amount of P450 2B6 present in and modifies the (apo)protein^^^, but the in vivo
liver and intestine, particularly in comparison with relevance of the inhibition has not been
P450 3A4 (vide supra). One estimate has been established.
made that P450 2B6 is involved in —3% of drug
metabolism reactions (Figure 10.3).
6.7.6. Clinical Issues
Lists of P450 2B6 substrates have been pub-
lished elsewhere, for example, refs 32 and 354, No real clinical issues have been identified yet,
and will not be reiterated here. One of the drugs to primarily because of the difficulty in identifying
which P450 2B6 apparently makes a significant reactions catalyzed in vivo due to the lack of spe-
contribution is cyclophosphamide^^^' ^^^. Some cific inhibitors and the overlapping regulatory
other reactions attributed to P450 2B6 involve mechanisms with other enzymes. Some pharma-
anesthetics, e.g., ketamine A^-demethylation^^^ ceutical companies have begun to include P450
Human P450s 407

2B6 in their in vitro screens for individual P450s appears to be inducible by barbiturates^^^ but a
with potential roles, however. PBREM was not found in the 5' untranslated
The phenomenon of barbiturate-like enzyme region of the gene^^ ^.
induction is still an issue in drug development, Several polymorphisms have been reported and
however. The point is not only drug interactions, studied^^^' ^^^' ^^^. Two coding region polymor-
but particularly the prospect of tumor promotion phisms involve the amino acid substitutions I264M
in rodent cancer bioassays, which is probably and K399R, with the latter appearing in a haplo-
unrelated to the P450 induction ^^^. type with R139K (ref [382]). Polymorphisms
upstream of the coding region are also known^^^.
The metabolism of taxol (paclitaxel) is decreased
6.8. P450 2C8 with the *3 allele (K399R/R139K haplotype), but
The P450s in the 2C subfamily have been of the extent of the decrease has been variable in dif-
interest for some time. In retrospect, some of the ferent studies, ranging from 90%^^^ to 25%^^^, 384
first human P450 preparations purified were prob- The *1C polymorphism appears to be associated
ably P450 2C9 (refs [3], [4]). A major impetus for with some attenuation of the mean level of
research in this field was the observed genetic expression^^^.
polymorphism in (5)-mephenytoin 4'-hydroxyla-
tion^^^' ^^^, which led to efforts at purification.
Purified proteins had some catalytic activity
toward mephenytoin^, but subsequent in vivo phar- 6.8.3. Substrates and Reactions
macokinetic^^^ and heterologous expression exper- P450 2C8 does not appear to have the general
iments^''^ demonstrated a distinction between significance of P450 2C9 (or 2C19) in drug metab-
tolbutamide and (*S)-mephenytoin hydroxylation. olism. An important substrate is taxol (pacli-
Genomic analysis indicated the complexity of the taxel)(6a-hydroxylation)^^' ^^^. Another substrate
CYP2C gene subfamily^^^. Subsequently the sub- for P450 2C8 is SilUrans retinoic acid^^^ P450
family was characterized in terms of four P450s: 2C8 also contributes to the oxidation of troglita-
2C8,2C9,2C18, and2C19 (ref [376]). P450 2C19 zone^^^ and verapamil, rosiglitazone, cerivastatin,
is the polymorphic (*S)-mephenytoin 4'-hydroxy- amiodarone, dapsone, and amodiaquine (reviewed
lase^^^' ^^^; P450 2C9 is involved in a considerable in refs [32, 382]).
number of drug oxidations (Figure 10.3). Two pre- In general, P450 2C8 has relatively low catalytic
vious entries, 2C10 and 2C17, are considered activity toward the known substrates of P450s
allelic variants cf other genes or other artifacts and 2C9 and 2C19. However, Mansuy's laboratory has
have been deleted (Table 10.1)^^^. recently synthesized a sulfaphenazole derivative
toward which all of the human P450 2C subfamily
P450s have activity^^^.
6.8.1. Sites of Expression and
Abundance
P450 2C8 was first purified from human
liver^; the enzyme is known to be expressed in 6.8.4. Knowledge about Active Site
liver and kidney^ ^^. The available data indicate Some of the knowledge about the catal5^ic
that the level of expression of P450 2C8 is rela- selectivity of various P450 2C enzymes can be
tively low in liver but may be one of the more sub- interpreted in terms of the active site. The rabbit
stantial P450s in the kidney. Other sites of P450 P450 2C5 structure provides some possible insight
2C8 (mRNA) include adrenal gland, brain, uterus, in this area. Recently, the groups of Johnson
manmiary gland, ovary, and duodenum^^^ and Mansuy^^^ have obtained a three-dimensional
structure with a common P450 2C subfamily
substrate^^^ bound. Two binding modes were
6.8.2. Regulation and Polymorpiiism
observed, one of which corresponds to the
The level of P450 2C8 expression in human observed oxidation^^^. Very recently Johnson's
liver varies at least 20-fold^^^. Rifampicin induces group has obtained a crystal structure of a slightly
P450 2C8 in hepatocyte culture^"^^. The enzyme modified P450 2C8 (ref [389a]).
408 F. Peter Guengerich

6.8.5. Inhibitors allelic variant of P450 2C9, although since no


evidence for the Asp417 mutation has been found
In contrast to P450 2C9, sulfaphenazole is not in population studies, no allele designation has
a strong inhibitor of P450 2C8. Mansuy's group been made; the original assignment had been based
has synthesized some sulfaphenazole-based selec-
on the now unexplained distinct 3' noncoding
tive inhibitors of individual P450 2C enzymes,
sequence^^^. Most of the literature dealing with
including P450 2C8 (refs [390], [391]). The early
P450 2C10 can be interpreted as 2C9.
work on paclitaxel metabolism suggests that high
concentrations of the natural flavonoids narin-
genin, quercitin, and kaempferol and the synthetic 6.9.1. Sites of Expression and
(xNF inhibit^^, but little in vivo inhibition would
Abundance
be expected.
P450 2C9 is primarily a hepatic P450. The
level of expression is probably the highest, on
6.8.6. Clinical Issues the average, except for P450 3A4 (Figures 10.1
Induction and inhibition of P450 2C8 are not and 10.4; Table 10.5)^1 All P450 2C enzymes are
particular issues at this point. Although P450 2C8 absent in fetal liver, including P450 2C9 (ref
may play a prominent role in the hepatic and renal [393]), and levels rise quickly in the first month
oxidation of arachidonic acid and retinoic acid, no after birth^^^. Pharmacokinetic experiments with
disease etiology has been implicated at this point. accepted P450 2C9 substrates indicate that the
The most serious issue is probably any impact on level of hepatic P450 2C9 does not change with
the disposition of the cancer chemotherapeutic age, at least to 68 years^^^.
agent paclitaxel. Polymorphisms may have some P450 2C9 is also expressed in the small
effect on in vivo 6a-hydroxylation^^^' ^^^, although intestine^^^.
any influence may be modulated in part by the
contribution of P450 3A4 to other reactions^^.
6.9.2. Regulation and Polymorphism

6.9. P450 2C9 Early work with human hepatocytes showed


induction of P450 2C9 by barbiturates and
In retrospect, many of the observations regard- rifampicin"^^^, consistent with earlier in vivo work
ing in vivo metabolism of barbiturates^^' ^^^ are on the induction of barbiturate metabolism^^^.
some of the first reports on what is known as P450 Subsequent studies have shown that P450 2C9 is
2C9. P450 2C9 is one of the major enzymes the only P450 2C subfamily enzyme expressed at
involved in drug metabolism (Figure 10.3). a significant level in untreated hepatocytes and
Retrospectively some of the first purified human that expression is induced by rifampicin, dexam-
liver P450s can now be recognized as P450 2C9 ethasone, and phenobarbitaP^^' ^^K The induction
(refs [3], [4]). The protein purified with some involves a glucocorticoid receptor, CAR, and
mephenytoin 4'-hydroxylation activity (MP-1) is PXR, with CAR and PXR apparently competing
also P450 2C9 (ref [9]), and the cDNA corre- at the same site'*^^.
sponds"^^^. Proteins were also purified from liver on Recently, evidence for action of CAR at an
the basis of their oxidation of tolbutamide^^^ and additional site has been presented^^^. It should be
hexobarbitaP^"^' ^^\ The human P450 2C subfamily emphasized that the action of CAR is somewhat
is complex^'^^ and characterization of individual different than other receptors from the steroid
members was not achieved without heterologous receptor superfamily, in that it may be enhanced in
expression and careful analysis of catalytic activi- the absence of a bound ligand and some of the
tjg5374,396 ^ transcript designated as P450 2C10 control is at the level of nuclear translocation'*^'*.
from this laboratory had only two apparent coding Other factors involved are HNF-4 (ref [405]) and
region changes (Cys358 and Asp417) from the C/EBPa (ref [338]), accounting at least in part for
CYP2C9*1 allele, one of which (reported initially hepatic localization.
as Cys358) was subsequently shown to result from The genetic polymorphism of P450 2C9
a sequencing error^^^. This is now thought to be an has been studied extensively and has clinical
Human P450s 409

significance, although P450 2C9 probably does P450 2C9 is the major catalyst of oxidation, and
not have a critical function in normal physiology. polymorphisms affect the in vivo pharmacokinetic
Tolbutamide metabolism had been reported to dis- parameters'^ ^'^^^.
play polymorphism^^^, which was an impetus to Several aspects of P450 2C9 reactions are of
purify the protein catalyzing the hydroxylation^^^. concern regarding interpretation of results, at least
A 6-base deletion in the coding region lowered in in vivo research. One issue is the effect of sol-
catalytic activity in a recombinant enzyme"^^^. vents on catalytic activity'^^. A concentration of
A number of P450 2C9 SNPs have been identi- 1% (v/v) CH3CN markedly inhibited the catalytic
fied"^^^ and their racial linkage has been explored"^^^. activity of P450 2C9 (ref [423]). Another issue is
P450 2C9 polymorphism has been reviewed the enhancement of most reactions by b^ (ref.
recently^^^' ^^^ and the reader is referred to these [424]). Further work also showed that apo-Z)^
reviews and to the website http://www.imm.ki.se/ (devoid of heme) was as effective as ^5 (ref
Cypalleles/ for more details. Of some interest, in [425]), arguing against a need for electron trans-
addition to the *2 and *3 alleles with generally fer. Other work showed that even other P450s
lower catalytic activity, is the *5 allele (of higher fre- could enhance the rates of some P450 2C9 reac-
quency in Africans) with lower catalytic activit)^^^. tions, even though those P450s did not catalyze
Some of the SNPs occur in the 5'-flanking region the reactions themselves"^^"^. These results are rem-
and attenuate the expression of P450 2C9 (ref iniscent of some of the interactions of rabbit
[413]). Also of interest is an unusual phenomenon in P450s 1A2 and 2B4 reported by Backes'^^ and are
which the CYP2C18 exon 1-like locus is fused with still unexplained.
combinations of exons and introns from CYP2C9 to Other work with P450 2C9 has provided evi-
yield chimeric RNA transcripts^^^. Finally, linkage dence for cooperativity in some reactions, although
between CYP2C8 and CYP2C9 genetic polymor- the area has not been as developed as for P450 3A4
phisms has been reported"^^^. {vide infra). Dapsone and some analogs enhance the
binding and 4-hydroxylation of diclofenac"^^^' '^^.
However, the activity of P450 2C9 toward dapsone is
6.9.3. Substrates and Reactions
unaffected by diclofenac, in a situation similar to that
P450 2C9 is one of the major P450s involved of P450 3A4, aflatoxin B^, and aNF'29 jy^^ jj^^er-
in drug metabolism (Figures 10.3 and 10.4). Some pretation that P450 2C9 uses two binding sites in
aspects of substrate specificity have been these interactions is probably valid"^^^, although (as
reviewed by Miners and Birkett^^^. A more exten- with P450 3A4) the mechanism remains to be eluci-
sive recent compendium of substrates has been dated (including the exact nature of the binding).
developed by Rendic^^.
One of the early substrates examined was pheny-
toin, which undergoes 4-hydroxylation^. P450s
6.9.4. Knowledge about Active Site
2C19 and 2C18 (R. Kinobe and E.M.J. Gillam,
personal communication) can also catalyze this The point should be made before detailed con-
reaction but P450 2C9 is the major catalyst"^^^. siderations of site-directed mutagenesis, etc., that
Recently, Mansuy's group has used the P450 changes in particular residues of P450 2C9 yield
2C9 inhibitor sulfaphenazole to build a substrate markedly different effects depending on the sub-
common to all four P450 2C subfamily enzymes^^^. strate and reaction under consideration. For
Some compounds normally in body are instance, the polymorphism *3 (I359L), which
oxidized by P450 2C9, including linoleic acid appears to be very conservative, changed catalytic
(epoxidation)"^^^ and vitamin A (all-^ra«5-retinoic efficiencies of different reactions by factors of
acid, 4-hydroxylation)'*^^, although the physiolog- 3-27-fold {in vitrof^^. Although the *2 and *3
ical significance is unknown. polymorphisms cause considerable changes with
Several reactions have been used as in vivo some substrates, diclofenac metabolism is not
probes, including tolbutamide, warfarin, flurbi- altered^^^ consistent with the in vitro findings.
profen, and losartan^^^. With the above caveats, roles of a number of
One substrate of recent interest is celocoxib, amino acids have been examined with several reac-
a cyclooxygenase (C0X)-2 inhibitor (Celebrex®). tions, although extrapolation to more reactions
410 F. Peter Guengerich

requires caution. Arg97 and Arg98 affected group has also announced a P450 2C9 crystal
activity toward diclofenac in a yeast recombinant structure"^"^^.
system"^^^; in contrast, mutation of Arg97 ablated
hemoprotein expression in a bacterial system
(E.MJ. Gillam, personal communication). Muta- 6.9.5. Inhibitors
tion of Lys72 failed to affect affinity for ibuprofen Sulfaphenazole has been recognized as a
or diclofenac (E.M.J. Gillam, personal communi- highly selective competitive inhibitor of P450 2C9
cation). Asp293 has been shown to have a rela- for some time"^"^^ and has relatively poor affin-
tively general structural role, possibly by bonding ity for other P450 2C subfamily enzymes^^^.
to a partner amino acid or amide^^^. Studies with Mansuy's group has examined some other similar
coumarins suggested two sites, one for Il-stacking compounds as ligands and inhibitors^^^' ^^^.
of aromatic rings and an ionic binding site for
Other inhibitors have been reported, although
organic anions^^"^; many P450 2C9 ligands have an
some have relatively poor affinity"^"^^' ^'^^, including
anionic charge^^^' '^^^.
several warfarin analogs^^^. For a more extensive
P450 2C9 was converted into an enzyme with compilation of inhibitors, see Rendic^^.
(5)-mephenytoin 4'-hydroxylation activity (i.e., Tienilic acid is a mechanism-based inactivator
P450 2C19-like) with a relatively small number of of P450 2C9 (ref [451]). The mechanism involves
changes (I99H, S220P, P221T, S286N, V292A, iS-oxygenation, and the unstable product reacts
F295L). Comparisons with the crystal structure of with P450 2C9 (ref [452]). Subsequently, autoim-
rabbit P450 2C5 suggests that most of these mune antibodies develop in some patients that rec-
residues are unlikely to directly contact the sub- ognize unmodified P450 2C9 (ref [451]). Exactly
strate but probably influence packing of substrate- how (or if) this process is related to the hepatitis
binding sites and substrate-access channels^^^. seen in some individuals who used tienilic acid is
Conversely, P450 2C19 could be transformed to still unclear'*^\ but the phenomenon has raised
an enzyme with warfarin hydroxylation activity concerns about whether such processes might be
similar to that of P450 2C9 (and also sul- associated with other drugs that covalently modify
faphenazole binding) with the changes N286S, proteins and could lead to idiosyncratic drug reac-
I289N, and E241K (ref [438]). Other work iden- tion in patients, one of the major concerns today for
tified roles of residues 292, 295, and 399 plus safety assessment in drug development. Structure-
residues 231-288 (substrate-binding sequence activity relationships have been reported on
[SRS] 3) as important in P450 2C9 activities^^^. thiophenes other than tienilic acid"^^^.
Mansuy's laboratory identified residues 476, 365,
and 114 as being important in diclofenac and sul-
faphenazole binding and in inactivation by tienilic
acid^"^^. Phell4 is proposed to be involved in 6.9.6. Clinical Issues
Il-stacking'^'^^ perhaps serving the role proposed The major issue regarding P450 2C9 is its
in the coumarin studies mentioned earlier^^"^. It role in drug development because of the sizeable
might be speculated that Phel20 in P450 2D6 fraction of drugs oxidized by this enzyme
could serve a similar role in that enzyme (vide (Figure 10.3)^^. Although the polymorphism is not
infmY^K as dramatic as with P450 2C19 or P450 2D6 (vide
Several models of P450 2C9 have been infra), it can be an issue in drug interactions and
published^^^'^^^^*^^. Some of these take experimen- safety.
tal binding studies into consideration in their formu- A general issue with P450 2C9, because of its
lation while others are only based on homology. Of relatively high abundance (Figures 10.1 and 10.4),
interest is the recent work with rabbit P450 2C5 is its role in reducing bioavailability. However,
using P450 2C9 ligands, showing multiple substrate- estimating in vivo pharmacokinetic properties
binding modes^^^. from in vitro data is still not trivial. Houston has
A crystal structure of P450 2C9 with bound reviewed the issue with P450 2C9 recently^^"*.
warfarin has been published recently^"^^^. Goldstein'*^^ has reviewed the clinical relevance
Obviously no information is available regarding of genetic polymorphisms in the P450 2C subfam-
ligand interactions either. Very recently Johnson's ily. One of the most relevant involves warfarin.
Human P450s 411

which has a relatively low therapeutic index"^^^. and protein"^^^ levels. However, expression in lung
(i^)-Warfarin is oxidized by P450 1A2 (6- and and skin appears to be significant^"^"^' ^'^^^ ^'^^.
8-hydroxy) and P450 3A4 (10-hydroxy), and (S)-
warfarin is oxidized primarily by P450 2C9
(7-hydroxy)^^^' ^^^. The metabolism of (*S)-warfarin 6.10.2. Regulation and Polymorpiiism
is competitively inhibited by (i?)-warfarin, but the The variability in levels of expression of P450
converse is not the case"^^^. The hydroxylation of 2C18 in human liver is difficult to assess because
(5)-warfarin by P450 2C9 (ref [459]) is an issue of the very low levels (<2.5 pmol/mg microsomal
because of reduced catalytic efficiency by the *2 protein)"^^-^. The extent of variability in other
and *3 variants^^' ^^^' ^^^ The differences are mani- tissues is not known.
fested in altered toxicity of warfarin (hemorrhaging) Rae et al?"^^ reported that P450 2C18 was not
at a given dose and in an altered optimal dose of inducible by rifampicin in human hepatocytes, in
warfarin^^"^^' ^^^^ ^^^. The issue extends to the analog contrast to P450s 2C8 and 2C9.
acenocoumarol"^^"^. The principles of physiologically Polymorphisms in the CYP2C18 gene have
based pharmacokinetic modeling have been applied been reported"^^^, but the effects on expression and
to the variation of warfarin risk in individuals with catalytic activities are not well characterized. One
different genotypes/phenotypes"^^^; this effort may possible polymorphism has an exon 5 deletion'^^^
serve as a paradigm for other efforts to convert in
vitro data on P450 variability into estimates of risk.
Tolbutamide hydroxylation is another example 6.10.3. Substrates and Reactions
of a manifestation of in vitro knowledge about
P450 2C18 has low catalytic activity in tolbu-
P450 2C9 in clinical pharmacolog/^^^^^ In one
tamide methyl hydroxylation"^^ ^ Limited activity
sense, this is rather logical because the in vitro
toward drugs has been shown, and P450 2C18
work with tolbutamide^^^ was developed from in
probably does not make much contribution in gen-
vivo findings"*^^.
eral drug disposition, in part because of low expres-
In other clinically relevant research involving
sion levels. P450 2C18 is active in phenytoin
P450 2C9, the genotype has been reported to pre-
metabolism, having an enzyme efficiency {k^JK^
dict the blood pressure response to the drug irbe-
for 4-hydroxylation comparable to P450 2C9, and
sartan^^^ a relative to the P450 2C9 substrate (and
being more active in the bioactivation to a reactive
the prodrug losartan)^^^' 47i Although P450 2C9
intermediate (R. Kinobe and E.M.J. Gillam,
is involved in the metabolism of diclofenac, no
personal communication).
relationship of the genotype with the cases of
Minoletti et al.^^^ studied a series of derivatives
diclofenac-induced hepatitis was observed"^^^.
of tienilic acid and characterized an aroylthiophene,
The final issue about P450 2C9 is possible rel-
3-[2,3-dichloro-4-(2-thenoyl)phenoxy]propan-1 -ol,
evance to cancer risk. Some carcinogens are sub-
as a selective substrate for 5-hydroxylation by P450
strates (e.g., benzo[a]pyrene^^^) although many of
2C18 (k. = 125 min-i, K^ = 9 |ULM).
the reactions are probably detoxications. CYP2C9
SNPs have been analyzed in relation to colorectal
cancer. An association was found in one study"^^^, 6.10.4. Knowledge about Active Site
but not a subsequent one"^^"^. In another study, no
association of CYP2C9 SNPs was found with lung Information about the active site of P450 2C18
cancer^^^. is relatively limited beyond the substrates cited
above"^^^, the interaction of other P450 2C proteins
with general 2C substrates^^^ and inhibitors^^^, and
6.10- P450 2C18 inferences from the rabbit P450 2C5 structures^^^.
At least one homology model has been published"*^^.
6.10.1. Sites of Expression and
Abundance
6.10.5. iniiibitors
Of the four human P450 2C subfamily mem-
bers, the level of hepatic expression appears to be P450 2C18 is not appreciably inhibited by sul-
lowest for P450 2C18, at both the mRNA376, 476 faphenazole. Mansuy's group has published on
412 F. Peter Guengerich

some synthetic inhibitors (sulfaphenazole deriva- 6.11.2. Regulation and Polymorphism


tives) that can be used in vitro^^^- ^^^
In vivo work had shown that the enzyme was
inducible by rifampicin^^^. Thus, this P450 dif-
fered from P450 2D6 in that it was both polymor-
6.10.6. Clinical Issues phic and inducible. Analysis of the regulatory
The limited expression and repertoire of cat- system has not been extensive, but studies with
alytic activity of P450 2C18 preclude considera- human hepatocytes have demonstrated induction
tion of clinical issues at this point in time. of P450 2C19 mRNA by rifampicin, dexametha-
sone, and phenobarbital'^^^
The polymorphism is now relatively well
understood. The incidence of the PM phenotype in
6-11. P450 2C19 Caucasians is generally 3-5% but the incidence in
Asians is —20%"^^. On some Pacific islands, the
Interest in P450 2C19 developed from the dis-
incidence is as high as 75%"^^^' ^^^. The major
covery of the pol3miorphic metabolism of the
defect in Caucasians and Japanese was first iden-
iS-isomer of mephenytoin, the first major poly-
tified in an exon 5 mutation that leads to an aber-
morphism to be studied following P450 2D6
rant splice site and yields a truncated protein^^^.
(refs [371], [372]). Initial work led to the purifi-
Other polymorphisms are collected at the website
cation of an enzyme with some (*S)-mephenytoin
http://www.imm.ki.se/CYPalleles/. These are rather
4'-hydroxylation activity^. Exactly how this and
diverse and include a mutation of the initiation
other gene products from the complex P450 2C
codon"^^^ and altered enzymatic properties"^^^.
family^^^' ^^^ were involved was unclear"^^^' ^^^.
Although there were some indications that the
hexobarbital 3'-hydroxylase (P450 2C9) was the
6.11.3. Substrates and Reactions
enzyme of investigation^^^' ^^^, expression of P450
2C9 cDNA^^^ in yeast yielded a protein with (»S)-Mephenytoin 4'-hydroxylation is the classic
activity towards tolbutamide but not (iS)-mepheny- reaction attributed to P450 2C19. Early studies on
tQijj374,396 P450 2C18 had also been suggested to the basis of the polymorphism of tolbutamide
be the enzyme^^^. hydroxylation suggested that the same enzyme
Wrighton^^'^ compared (»S)-mephenytoin 4'- might be responsible for both activities^^^, but in
hydroxylation activity in different liver samples vivo work^^^ and heterologous expression studies^^'*
with a protein gel band recognized by anti-rat distinguished the two activities. Nevertheless,
P450 2B1 and correlated this with P450 2C19, recombinant P450 2C19 has now been shown to
a sequence which had been reported earlier. have some tolbutamide hydroxylation activity^^^.
Subsequently, Goldstein et al?^^ expressed several Extensive lists of reports of P450 2C19 reactions
P450 2C subfamily cDNAs in yeast and identified have been published by Rendic^^ and only a few will
P450 2C19 as having the highest activity be mentioned. The scope of P450 2C19 in drug
metabolism is relatively restricted (Figure 10.3).
One drug of particular interest is the ulcer drug
omeprazole (and related compounds), because indi-
6.11.1. Sites of Expression and
viduals with low enzyme activity show a better
Abundance response to treatment for ulcers^^' ^^. Some of the
Apparently significant expression only occurs early variations seen in warfarin metabolism^^^ can
in the liver. As with all other P450s examined to be explained by the finding that P450 2C19
date, there appears to be no gender difiference"^^^. catalyzes the 8-hydroxylation of (/?)-warfarin'^^'^.
P450 2C19 is a relatively minor P450 in its abun- 18-Methoxycoronaridine is 0-demethylated by
dance, probably accounting for < 5 % of total P450 P450 2C1949^ P450 2C19 is responsible for the 5-
even in EM liver samples (Figure 10.4). and 5'-hydroxylation of thalidomide, an older drug
P450 2C19 and (5)-mephenytoin 4'-hydroxy- notorious for teratogenic effects that has been
lation activity were not detected in fetal liver "rediscovered'"^^^. Whether the polymorphism was
samples^^^. related to the birth defects is unclear.
Human P450s 413

P450 2C19 also oxidizes steroids, including Most pharmaceutical companies and regula-
progesterone 21-hydroxylation and testosterone tory agencies discourage development of a P450
IT-oxidation"^^^. Finally, the organphosphate 2C19 substrate because of potential problems for
insecticide diazinon is activated in human liver by PM individuals. However, several studies indicate
P450 2C19 (ref. [498]). that PM patients may have more effective therapy
(for ulcers) with omeprazole and related com-
pOUnds489, 500-503^
6.11.4. Knowledge about Active Site As with many polymorphisms, epidemiology
As with other P450 2C subfamily enzymes, studies have been done to explore risks to diseases
P450 2C19 activities are usually stimulated by b^ in the absence of information about etiology, sub-
(ref. [425]). In this case, stimulation is not strates, etc. Some of the reports include sugges-
dependent on heme in the b^ so electron transfer tion of more hepatocellular cancer in PMs^^"^ and
cannot be involved"^^^. lack of association of leukemia with polymor-
Homology models of P450 2C19 have been phism^^^. Other possible relationships have been
published302,444 explored but evidence for any associations is
Goldstein's group did chimeric analysis and limited at this time"^^^.
then site-directed mutagenesis on P450 2C9 to
convert it to a protein with P450 2C19-character- 6.12. P450 2D6
istic omeprazole hydroxylation activity^^^. Only
three changes were needed to achieve the activity P450 2D6 is one of the main enzymes involved
of wild-type P450 2C19:199H, S200P, and P221T. in drug metabolism (Figure 10.3). It was the first
However, at least three different mutations were "xenobiotic-metabolizing" P450 recognized to be
needed to convert P450 2C9 to an enzyme with under monogenic regulation^.
(»S)-mephenytoin 4'-hydroxylation activity, even
to a catal3^ic efficiency one third of wild-t3q)e
P450 2C19 (ref [437]). In an opposite experi- 6.12.1. Sites of Expression and
ment, P450 2C19 was converted to a P450 2C9- Abundance
like warfarin hydroxylase with high sensitivity to
P450 2D6 is expressed mainly in liver and was
sulfaphenazole"^^^. Residues 286 and 289 appear
first purified from liver microsomes^' ^^. In the
to be important. However, these residues may
average person, P450 2D6 accounts for —5% of
exert an indirect influence by adjusting the active
total P450 (with wide variation)^^. However, this
site or substrate-access channels"^^^.
enzyme is involved in the oxidation of ~ 2 5 % of
all drugs oxidized by P450s (Figure 10.3).
6.11.5. Inhibitors Developmental studies show little P450 2D6 in
fetal liver and a rapid increase in protein shortly
Relatively little has been published concerning after birth, yielding a peak accumulation in
P450 2C19 inhibitors, although screening may be newborns and decline in adulthood^^^.
done in some pharmaceutical companies. Recently, P450 2D6 is also expressed at low levels in
Mansuy's group has developed some P450- lung (bronchial mucosa and lung parenchyma)^^^.
selective inhibitors for the 2C subfamily enzymes, Another site of P450 2D6 expression is brain,
including P450 2C19 (refs [390], [391]). with localization in large principal neurons^^^.
Higher levels of brain expression have been
reported in alcoholics^^^.
6.11.6. Clinical Issues
The issue is the polymorphism, particularly for
6.12.2. Regulation and Polymorphism
drugs marketed in Asian populations. At least
eight alleles have been associated with the PM AH information available indicates that P450
phenotype"^^^. Desta et al.^^^ have reviewed some 2D6 is not inducible. Some factors are known to
of the drugs for which the 2C19 phenotype is a be involved in constitutive expression, including
problem. C/EBPa338 and HNF-4a53.
414 F. Peter Guengerich

The wide variability in the activity of P450 2D6 result from less stable proteins^ *^, although low
is attributed to genetic variabihty (Figure 10.5). activity P450 2D6 variant proteins have also been
Reduced ability to metabolize the drug debrisoquine reported^*^'^*^. Some of the allelic differences are
was first noted (personally) by Smith in a drug trial. present as haplotypes^*^.
Subsequent work led to the report of polymorphic In addition to the "poor" and "intermediate"
hydroxylation of debrisoquine^, including a pheno- metabolizer phenotypes, an "ultrarapid" metabo-
typic hypotensive response^^^. Racial differences lizer phenotype was identified in early work
were first noted with Africans'^'^. The phenomenon (Figure 10.5). Ingelman-Sundberg's group identi-
of polymorphic debrisoquine hydroxylation^*^ was fied the basis for this as a gene duplication, with up
also reported for sparteine oxidation"^^' ^*^. to 13 copies present in some individuals'*^. The
Purification of the P450 2D6 enzyme^' *^' *' was fol- main form of this phenomenon is a haplotype
lowed by Gonzalez's cloning of the gene'^'^ and iden- resulting from gene duplication"*^' ^^^. The amplifi-
tification of some of the genetic defects as mRNA cation appears to result from unequal segregation
splicing variants^*^. and extrachromosomal replication of the acentric
Today more than 70 alleles of P450 are DNA^^*. As many as 7% of Caucasians show some
known and have been classified with a nomen- of this effect, and the incidence is even higher in
clature system^ ^ Systems for genotyping have some Ethiopian and Middle Eastern populations^^^.
become relatively powerful^^"^ and the "intermedi-
ate metabolizer" phenotype has been character-
6.12.3. Substrates and Reactions
ized^*^. The most significant decreases in activity
for P450 2D6 alleles, aside from mRNA splicing Since the original work with debrisoquine^,
problems and gene deletion^^, are considered to many substrates and reactions have been reported

Figure 10.9. A pharmacophore model for the active site of P450 2D6 (ref [527]). Inhibitors are overlaid to keep
the nitrogen atoms (marked with arrow) in a fixed position.
Human P450s 415

for P450 2D6. In some cases, the role of P450 usually Asp301 in most studies. (Recent work
2D6 is very dominant in vivo and the clinical man- shows a role for Glu216, however, vide infra.)
ifestations of genetic polymorphism are important The use of these models requires some caveats.
and even deadly^^^' ^^^. An extensive list of P450 Although the pK^ of the substrate has been proposed
2D6 substrates has been published recently by to have a dominant influence^^^, work in this labo-
Rendic^^. ratory has shown that the intrinsic pK^ of a substrate
P450 2D6 catalyzes many of the basic kinds can be altered in the active site of P450 2D6 (ref
of oxidative reactions of P450s, for example, [531]). Another issue is that some compounds with
aliphatic and aromatic hydroxylations, heteroatom a single amine nitrogen undergo A/-dealkylation, for
dealkylations, etc^^"^. In early work in this labora- example, deprenyl^^^, which cannot be rationalized
tory^^^, the observation was made that most of the with an amine-oxidation site interatomic distance of
substrates contained a basic nitrogen atom situ- 5-7 A. Some substrates devoid of basic nitrogen
ated ~ 5 A away from the site of oxidation, possi- (and any nitrogen) have been reported, including
bly due to a specific anionic charge in P450 2D6. steroids^^^' ^^^. Spirosulfonamide and several
Subsequently, more detailed pharmacophore mod- analogs are devoid of basic nitrogen and have been
els have been developed^^^"^^^ (Figure 10.9). All shown to be good substrates and ligands for P450
of these are based on the premise that a basic 2D6 (ref [535]) (Figure 10.10).
nitrogen atom in the molecule interacts (coulom- A large fraction of the population is devoid of
bic bond) with an acidic amino acid in P450 2D6, active P450 2D6 but appears to fimction well. This

Bufuralol

1 R = so2NH2 Ka^^\iM 6
/Cd 6.2 ^iM
(spirosulfonamide) Kd 4.0 ^iM

2 R = SO2CH3 Kd 32 |iM
(spirometiiyisulfone)

3 R = CONH2

4 R = CSNH2 /Cd1.7|iM

5 R = C02H (Kd >75 |IM)


Testosterone Progesterone
KdlS^iM /Cd1.5|LiM
CH3
H3C-

a
Ritonavir K^i1.2\ihA

Figure 10.10. Analogs of spirosulfonamide and other P450 2D6 ligands. K^ values were estimated by spectral
titrations^^^.
416 F. Peter Guengerich

information may be interpreted to mean that P450 A list of P450 2D6 residues postulated to form
2D6 has no "physiological" substrate. Nevertheless, the active site includes at least Asp 100, Trp316,
some reactions may be catalyzed by P450 2D6 and Pro371 (ref [539]), Prol03, Ilel06, ThrlOT,
yield physiological responses that yield less than LeullO, Proll4, Serll6, Alal22, Asp301, Ser304,
obvious changes. For instance, overexpression of Ala305, Thr309, Val370, Gly373, Val374, and
human P450 2D6 in transgenic mice produces Phe483 (refs [542], [551]), Phel20, Glu216 (refs
a somewhat lethargic phenotype (F.J. Gonzalez, [441], [541], [543], [544], [550], [552], [553]), and
personal communication). Tryptamine has been G l n i n , Leul21, Leu213, Phe219, and Phe481
proposed as a physiological substrate in one study^^^ (ref [543]). Only six of these residues have been
but discounted in another^^^. Proposed physio- examined experimentally to date. The effects of
logical reactions catalyzed by P450 2D6 are Asp301 have already been mentioned, with caveats
the 0-demethylations of 5-methoxytryptamine, about general changes in the protein"^"^^' ^^^.
5-methoxy-i\yV-dimethyltryptamine, and pinoline Changing Val374 to Met also has an eflfect^'^^' ^^7.
(6-methoxy-l,2,3,4-tetrahydro-p-carboline)^^^' ^^l Mutation at Asp 100 or Ser304 has been reported to
Whether significant catalytic is seen at the low con- have little effect, if any^'^^' ^^\ Mutation of Phe483
centrations that occur in vivo and what the effect is to He produced some alteration of the pattern of
remains to be established. testosterone oxidation by P450 2D6 (ref [551]). A
change in Phe481 yielded a 10-fold lower catalytic
efficiency (k^JK^) toward some substrates but not
6.12.4. Knowledge about Active Site others^^^. The effects of Glu216 have already been
The active site of P450 2D6 has been the sub- mentioned"*"^^' ^^^ and seem to be restricted largely
ject of considerable interest, probably because of to the basic amines'^'*'. Recent models of the P450
the relevance to issues in the pharmaceutical 2D6 active site (Figure 10.11) suggest that both
industry. Some residues have been identified as Asp301 and Glu216 are within bonding distance of
being important, and many homology and phar- amine substrates'^'*'' ^'*^. Another suggestion from
macophore models have been published^^^"^^^' the more recent models'*"*'' ^^^ is that one role of
539-545 (Figure 10.9). Asp301 is to use amide hydrogen bonds to estab-
The original clone reported by Gonzalez"^^ had lish the juxtaposition of Phel20, which may be
Met at position 374 but this now appears to be an involved in hydrophobic bonds with substrates.
artifact and the correct residue is Val^"*^' ^^^. This Site-directed mutagenesis experiments with this
residue appears to be in the active site and affects residue are currently in progress (F.P Guengerich
activity. and E.M.J. Gillam, unpublished results).
In 1995, Ellis et al.^"^^ found that mutation of The work cited above brings up the point that
Asp301 to neutral residues reduced catalytic activ- certain mutations may alter activity toward some
ity toward several substrates and concluded that substrates but not others (e.g., Phe481 (ref [555]),
this acidic residue was involved in docking amine Glu216 (ref [441])). Similar behavior is seen with
substrates through coulombic interaction. Subse- some of the natural allelic variants of P450 2D6 as
quently, all models published until recently have well556.
been based on this view. A caveat about the reduc- Modi et al.^^^ reported differences in product
tion in the catalytic activity of the Asp301 mutants profiles of P450 2D6 reactions supported with
is that heme incorporation is diminished (and artificial oxygene surrogates and NADPH-P450
is completely abolished when basic residues are reductase, and interpreted these as evidence for an
substituted)^^^. Further, as indicated earlier, some allosteric influence of the reductase. Subsequent
P450 2D6 substrates (e.g., spirosulfonamide) are experiments in this laboratory did not support this
devoid of basic nitrogen but the hydroxylations conclusion and are in accord with some differ-
are still attenuated by mutation at Asp301 ences in the chemical mechanisms for the oxygen
(ref [535]). Subsequent work in this laboratory surrogates^^^.
showed that the oxidations of basic amine sub- Detailed experiments have been done on the
strates (and their binding) are dependent upon 0-demethylation of 3- and 4-methoxyphenethy-
Glu216 (Asp216 is also efiFective)'^^^ a result lamine by P450 2D6 (ref [559]). Analysis of kinetic
independently reported by Wolf's group^^^. deuterium isotope effects, kinetic simulation, and
Human P450s 417

Figure 10.11. Model of some residues in the P450 2D6 active site'^'^^ Views of the substrate-binding cavity in the
P450 2D6 homology model are shown with only the relevant residues, plus the heme, I-helix backbone, and other
areas of peptide backbone shown. Hydrogens are not shown except for the amide hydrogens of residues 119 and 120
hypothesized to hydrogen bond to the carboxylate oxygen atoms of Asp301. Side view of the active site from the
perspective of the I-helix: I-helix residues have been cut away excepting the side chains of residues 309, 301, and
313 shown at the front of the view.

other experiments yield evidence that both late steps Rendic^^. Inhibition of P450 2D6 is an undesirable
in O2 activation and C-H bond breaking contribute issue in drug development, and most pharmaceuti-
to k^^^. The exact meaning of X^ is still not defined cal companies have screening programs in place.
with this and most P450 reactions. Some of the The most established inhibitor of P450 2D6 is
P450 2D6 allelic variants show no changes in k^^^ quinidine^^^ The K^ is ~50 nM and inhibition is
for certain reactions but do show K^ differences^^^; competitive. Interestingly, quinidine is not a sub-
these are probably more complex than simple strate for P450 2D6 (refs [106], [559]).
"affinity" for the substrate. Mechanism-based inactivation of P450 2D6 is
known, for example, 5-fluoro-2-[4-[(2-phenyl-
li/-imidazoyl-5-yl)methyl]-1 -piperazinyl]pyrimi-
6.12.5. Inhibitors
dine (SCH66712)^^^. In the case of this compound,
Many inhibitors of P450 2D6 have been covalent binding to protein was detected but the
reported; for a compilation of the literature, see position of attachment has not been identified.
418 F. Peter Guengerich

6.12.6. Clinical Issues Another disease in which P450 2D6 has been
proposed to play a role, on the basis of epide-
The clinical issues regarding P450 2D6 are miology, is Parkinson's disease^^^. Contradictory
considerable due to the large variation in the findings have been reported^^^' ^^^. Although a
genetics in the population (Figures 10.2 and 10.5), hypothesis has been raised that induction of P450
and the contribution of P450 2D6 in the total 2D6 by smoking might explain some discrepan-
scheme of drug metabolism (Figure 10.3). cies^^ ^ this proposal lacks biological plausibility in
Individuals seem to be rather tolerant of the wide light of the known refractory response of P450 2D6
variability in expression with many marketed to induction.
drugs, probably because of generally wide thera- A final issue is that of autoantigens. Auto-
peutic windows selected for in the basic process of antigens (LKMl) that recognize P450 2D6 have
drug development. However, P450 2D6 PMs can been known for some time^^^' ^^^. These antibod-
be at considerable risk when they encounter cer- ies are associated with some cases of hepatitis.
tain drugs, as first observed by Smith^' ^^^. The The exact mechanism of how they arise is still
problem is seen with drugs having a relatively unclear, as is the relationship with hepatitis. The
narrow therapeutic index, for example, debriso- antibodies may arise by molecular mimicry^^"^
quine^, phenformin^^^, captopril^^"^. The effects of or they may result from P450 2D6 translocation
P450 2D6 deficiency are seen not only in short- to the outer plasma membrane^^^' ^^^. These
term treatments but also in long-term therapy^^^. "LKMl" antibodies may serve as diagnostic tools
The issue of ineffectiveness of drugs that are very for particular types of hepatitis^^^' ^^^, but causal
rapidly metabolized by "ultrarapid" metabolizers relationships have never been demonstrated.
is an issue (Figure 10.5). Modeling of the vari-
ability is still an issue^^^ and may be a function of
particular drugs. The issue of whether genotyp-
ing/phenotyping is economical has been consid-
ered, particularly in the case of neuroactive and 6-13. P450 2E1
antipsychotic drugs^^^' ^^^. The overlap between The mixed-function oxidation of ethanol was
P450 2D6 substrates and neuroactive drugs is also reported nearly 40 years ago^^^. The view that
an issue in drug development, largely due to the ethanol could be a P450 substrate was not readily
overlap of these two groups of compounds^^^. accepted because of the hydrophilic nature of the
Another issue with P450 2D6 is the relevance molecule, but Lieber's group characterized the
of the polymorphism to cancer risks. In 1984, enzyme in rat liver^^^' ^^K Collaborative work with
Idle^^^ reported an association of lower risk of Levin led to the isolation of the P450 ("j"), which
lung cancer (in smokers) with the P450 2D6 PM was also found to be inducible by isoniazid^^^.
phenotype. These epidemiology results were Human P450 2E1 was purified by Wrighton
reproduced in some studies^^^, but not others'^^. et al.^^^, and Gonzalez's group characterized the
Attempts were made to resolve the discrepancies human gene^^'*.
on the basis of levels of smoking^^^ Although
some expression of P450 2D6 is detectable in
lung^^^, no clear role for P450 2D6 in carcinogen
6.13.1. Sites of Expression and
activation could be established, even with crude
Abundance
tobacco smoke fractions^^^. The issue of whether
lung cancer is associated with P450 2D6 was not The greatest concentration is in the liver, and
resolved by changing analyses from phenotyping P450 2E1 is a moderately abundant P450
to genotyping. The generally accepted epidemio- (Figure 10.4). The inter-individual variation is an
logical conclusion today is that P450 2D6 is not order of magnitude (Figure 10.2)^^' ^^^. A racial
related to lung cancer^ ^^' ^72-575 difference exists, with Japanese samples having
Other epidemiology studies have suggested rela- mean expression levels less than Caucasians
tionships of P450 2D6 with other cancers^^^' ^^^, but (Figure 10.2)^1
these findings have not been scrutinized as much as P450 2E1 is reported not to be present in fetal
the lung cancer hypothesis. liver but appears within a few hours after birth,
Human P450s 419

regardless of the gestational age^^^. The activity increased mRNA translation efficiency, and
increases during the first year of childhood, and decreased protein degradation^^^.
transcriptional regulation due to hypermethylation HNF-1 is reported to regulate CYP2E1 gene
has been proposed. transcription^^^. Obesity and diabetes are known to
P450 2E1 is expressed in many extrahepatic sites modulate P450 2E1 in rat models. In rat hepatocyte
including lung^^^, esophagus, small intestine^^^, cell culture, insulin attenuated mRNA levels and
brain^^^' ^^^, nasal mucosa^^^, and pancreas^^^ (some glucagon or dibutyryl cyclic AMP elevated mRNA,
of the evidence is extrapolated from rat work but not with the latter effect downregulated by a protein
necessarily extended to humans). kinase A inhibitor^ ^^. mRNA levels are also selec-
P450 2E1 is found mainly in the endoplasmic tively attenuated in mice or cell culture (relative to
reticulum. With heterologous expression in bacte- other P450s) by interleukin-6^^\ interleukin-4^^^, or
ria, (rabbit) P450 2E1 is membrane bound and interleukin-ip or tumor necrosis factor (TNF)a^^^.
catalytically active even when amino acids 3-29 Multiple mechanisms have been invoked, including
are deleted^ ^' ^^^. The same bacterial localization kinase pathways, control of HNF-1 a fimction, and
was seen with human P450 2E1 from which 21 regulation of other transcription factors.
N-terminal residues were deleted^^^. However, Evidence for control at the level of mRNA sta-
P450 2E1 can show some unusual localization in bility and enhanced translation efficiency has
mammalian systems. Ingelman-Sundberg's group been presented by Novak^^"^' ^^^. The 3'-region of
deleted residues 2-29 of rat P450 2E1 and demon- the gene appears to be important in stability. The
strated the presence of a shattered fragment in the relevance of this rat model to human P450 2E1 is
mitochondria of a mouse hepatoma cell line^^"^. still unknown.
Avadhani's group found P450 2E1 intact in rat Another mechanism, generally well accepted
liver mitochondria and reported that it could cou- although not completely understood, involves pro-
ple these with adrenodoxin and adrenodoxin tein stabilization by substrate. Rat studies {in vivo)
reductase there with frill catalytic activity^^^. showed that - 1 / 2 of P450 2E1 was lost in 1 hr,
Subsequent work demonstrated a cryptic mito- and a ubiquitin-linked pathway was invoked^^^.
chondrial targeting signal at positions 21-31 that Similar findings were also reported for human
was activated by cyclic AMP-dependent phospho- P450 2E1 in HepG2 cells^^l An attempt has been
rylation of Serl29 (ref. [26]). Neve et al^^^ found made to estimate the halflife of P450 2E1 in
that the charge of the iV-terminus of (rat) P450 humans in vivo using chlorzoxazone pharmacoki-
2E1 was such that a part is directed to either the netics and a P450 2E1 inhibitor^^^. The halflife
lumen of the endoplasmic reticulum or the outside was estimated at 50 ± 19 hr, but this approach
of the plasma membrane. The relevance of these may not be sensitive enough to detect a short-lived
localizations to human tissues is still unknown P450 2E1 pool. The relevance of substrate stabi-
but likely. lization of P450 2E1 to in vivo parameters has
been addressed by Thunmael and Slattery^^^.
P450 2E1 is polymorphic and, because of the
6.13.2. Regulation and nature of many of the substrates, many efforts have
been made to determine the relevance of SNPs and
Polymorphism
other polymorphisms to disease and risk of injury.
Early work in experimental animals was For a current update on CYP2E1 polymorphisms,
focused on the induction of P450 2E1 in rat see http://www.imm.ki.se/Cypalleles/. A polymor-
liver^^^. Subsequently, many other chemicals, phism in the 5' flanking region was suggested to
including isoniazid and some solvents, were shown be related to the binding of a transcription factor
to induce P450 2E1 (ref [607]). It is also of and related to alcohol intake^^' ^^^. A number of
interest to note that some of the common poly- other polymorphisms have been identified^^' ^^^'
cyclic hydrocarbons and other inducers of P450 1 ^^^. However, the evidence to date indicates that
family enzymes attenuated the level of P450 2E1 these polymorphisms do not seem to have much
(ref [607]). The regulation of P450 2E1 has come significance in terms of their effects on in vitro
to be recognized to be relatively complex, involv- or in vivo activity of P450 2E1 (refs [84], [621],
ing transcriptional activation, mRNA stabilization, [623-625]).
420 F. Peter Guengerich

6.13.3. Substrates and Reactions molecular weight cancer suspects including not
only nitrosamines but also benzene, styrene, CCI4,
P450 2E1 was originally characterized as an CHCI3, CH2CI2, CH3CI, CH3CCI3, 1,2-dichloro-
ethanol-oxidizing enzyme. P450 2E1 can oxidize propane, ethylene dichloride, ethylene dibromide,
some compounds that are present in the body, vinyl chloride, vinyl bromide, acrylonitrile, vinyl
including acetone and possibly other ketones carbamate, ethyl carbamate, and trichloroethyl-
involved in certain physiological syndromes gj^g304 jj^Q oxidations by P450 2E1 all have rele-
(fasting, diabetes)^^^. Transgenic P450 2E1-knock- vance to the activation and detoxication of these
out mice appear to be relatively normal, although compounds and their risk assessments^"*' ^^^.
the blood acetone levels become much higher Another substrate is the gasoline additive methyl
(than in wild-type mice) after fasting^^^. tert-buty\ ether^^l A role of P450 2E1 has been
The role that P450 2E1 plays in ethanol metab- shown in the activation of some of these chemicals
olism has been debated for many years^^^. What in knockout mice^"*"*' ^'*^.
seems to be the general consensus is that alcohol Another substrate for human P450 2E1 is lau-
dehydrogenase is the main enzyme involved in ric acid, which undergoes 1 l-hydroxylation^'*^' ^'*^.
ethanol oxidation. P450 2E1 may make a contri- The physiological relevance of this reaction
bution at very high ethanol concentrations or in is unknown. Indole is oxidized by P450 2E1
individuals with low levels of alcohol dehydroge- (3-hydroxylation, generating indigo) as well as
nase activity. P450 2E1-knockout mice have blood by other P450s, particularly P450 2A6 and 2C19
ethanol levels not significantly different from wild- (refs [296], [648]). The relevance of this reaction to
type animals after administration of ethanol^^^. the urinary excretion of indigoids^"*^ is still unclear.
Acetaldehyde, the product of ethanol oxidation, is
Relatively few drugs are oxidized by P450 2E1
also oxidized to acetic acid by rat and human P450
(Figure 10.3). Chlorzoxazone is one^^"*. Halogenated
2E1 (ref [630-632]).
anesthetics are often metabolized by P450 2E1,
The oxidation of 4-nitrophenol to 4-nitrocate- including halothane^^^ and isoflurane^^^
chol has been used as an in vitro marker of human For more lists of substrates, see Rendic^^.
P450 2E1 (ref [633]). Chlorzoxazone 6-hydroxyla-
tion was demonstrated to be a relatively specific
reaction catalyzed by human P450 2E1; other
6.13.4. Knowledge about Active Site
enzymes (e.g., P450 lAl) can catalyze the reaction
but with poor catalytic eff'iciency^^'*' ^^^. Chlorzo- One of the issues in P450 2E1 reactions is
xazone is a relatively innocuous muscle relaxant and the need for b^, first demonstrated with the rat
the assay can be used in vivo to estimate hepatic enzyme^s^ and also the human enzyme^"*^' ^^^; the
P450 2E1 fimction noninvasively^'^' ^^^. involvement also exists in microsomes^^^. b^ also
One group of substrates of interest is A^- augments P450 2E1 activity in bacterial expres-
nitrosamines, which are carcinogens at many sites sion systems"*^^' ^^^. In contrast to several of the
and can be formed by chemical reactions within P450s, apo-Z?3 (minus heme) does not ftinction,
the body (e.g., stomach acid)^^^. Early research on arguing for a "classic" role of electron donation in
the activation of A^-nitrosodimethylamine (N,N- enhancement of catalysis"^^^' ^^^.
dimethylnitrosamine) indicated biphasic kinetics A number of homology models of human P450
of the activating iV-demethylation reaction and the 2E1 have been published, based upon bacterial
possible contribution of multiple P450s and possi- P450s and rabbit P450 2C5 (refs [302], [656],
bly other enzymes^^^' ^^^. The enzyme involved in [657]). One of the diff'iculties in dealing with mod-
the "low KJ' reaction was shown to be P450 2E1 els for the low molecular weight substrates is that
m
many of these compounds have very little in the
in rat and human liver^^^' ^^^. An in vivo role of
way of features to bond to, other than hydrophobic
P450 2E1 has been confirmed in rats^"^'. However,
residues or halogens. Utilizing these models for
P450 2A6 has a significant share of the role of
both very small substrates (e.g., ethanol, CH3CI)
activation of some more complex nitrosamines,
and larger, more conventional ones (e.g., chlorzox-
even A^-nitrosodiethylamine-^^^' ^^^.
azone, lauric acid) is an issue, unless only parts of
P450 2E1 has been shown to be a major P450
the larger substrates are inserted. One problem is
involved in the oxidation of a number of low
Human P450s 421

that inherent binding affinities are generally isotope effect is expressed in the K^ parameter,
unknown and spin-state changes have not been which includes the C-H bond-breaking step, k^^^ is
very useful with P450 2E1 (refs [652], [658]). governed largely by an enzyme physical step after
Mathematical models have also been devel- oxidation of substrate. In this system, the K^ term
oped for rates of oxidation by P450 2E1 contains k^^^ as a variable^^^' ^^^.
(refs [659], [660]). In essence, these are based on A final point involves a report of the kinetics
chemical reactivity at individual substrate atom of CO binding to human P450 2E1 following flash
sites. In both of the cited examples^^^' ^^^, the photolysis^^^. The kinetics appeared to be
models were used for relatively small sets of monophasic and the rate was decreased in the
related compounds and may have some utility. An presence of (400 mM) ethanol. One interpretation
inherent problem in more extended sets is the dif- of the results is that binding of the substrate makes
ficulty in interpretation of the parameters k^^^ and P450 2E1 more rigid^^l
K^. Thus, the rate-limiting step may not be related
to hydrogen abstraction or a similar chemical step 6.13.5. Inhibitors
involving the substrate {vide infra).
As mentioned earlier, many low molecular
Keefer et al.^^^ reported a kinetic deuterium
weight solvents are substrates for P450 2E1.
isotope effect on the carcinogenicity of iV-nitroso-
These are also inhibitors of P450 2E1 (refs [69],
dimethylamine in rat liver. Subsequent work with
[70]). Such inhibition is a problem in that histori-
rat and liver microsomes indicated that the effect
cally many insoluble P450 substrates have been
of the deuterium substitution was expressed in the
added to enzymes using final solvent concentra-
parameter K^ but not k^^^ (^max)^^^' ^^^ ^^^^ ^^^"
tions of 1% (v/v), which is often —100 mM. Thus,
tope effects on K^ were seen when deuterated
care is needed in analyses. It is possible to dilute
iV-nitrosodimethylamine was used as a competitive
many of the P450 2E1 low molecular weight sub-
inhibitor of other P450 2E1 reactions^^^. These
strates directly in water to add them to incuba-
results were of interest in that deuterium substitu-
tions, for example, methylene chloride has a
tion would not be expected to modify the affinity
solubility of - 1 0 0 mM in H^^^^.
of a substrate for an enzyme. Studies with deuter-
ated and tritiated ethanol in this laboratory also Some of the alcohol and aldehyde dehydroge-
indicated an isotope effect on the oxidation of both nase inhibitors are also inhibitors of P450 2E1,
ethanol and acetaldehyde by recombinant human making interpretations of in vivo ethanol metabo-
P450 2E1, manifested mainly in Kj^^^ ^^^ J^IQ lism studies difficult. 4-Methylpyrazole is an
results are understood in the context of a reaction excellent inhibitor^^^' ^^^ and probably one of the
sequence where burst kinetics are observed, that is, best choices for in vitro experiments at this time.
the first reaction cycle is much faster (—400 min" ^) 3-Amino-l,2,4-triazole^^'^ and diethyldithiocarba-
than the subsequent ones, which control k^^^. Pulse- mate^^"^ are mechanism-based inactivators. The
chase experiments suggest that little of the latter is of interest in that the oxidized form, disul-
acetaldehyde (or its hydrated form CH3CH(OH)2) firam (Antabuse®), is an aldehyde dehydrogenase
dissociates, due to kinetic phenomena. Neither inhibitor used in patients in alcohol aversion ther-
ethanol, acetaldehyde, nor acetic acid has much apy. Many of the early animal and human studies
affinity for P450 2E1. The rate-limiting step on interactions of ethanol and disulfiram with
occurs after product formation (for both ethanol various chemicals can now be rationalized in the
and acetaldehyde oxidations), but is not product context of P450 2E1 (refs [668], [669]).
release per se. This view of the reaction sequence A number of compounds of natural origin have
may apply to some P450 2E1 reactions but not also been examined as P450 2E1 inhibitors, many
others. Recent work in this laboratory with both of which are derived from vegetables such as
P450s 2E1 and 2A6 has shown a kinetic isotope onions, garlic, and cruciferous vegetables^^^' ^^^
effect primarily on X^, for the iV-dealkylation of
iV-nitrosodimethylamine but not A'-nitrosodiethy- 6.13.6. Clinical Issues
lamine^^^, and the kinetic mechanisms remain to The major clinical issues involve the role of
be further elaborated. An interesting point of the P450 2E1 in the oxidation of certain drugs, alco-
deuterated ethanol work is that the intermolecular holism, oxidative stress, and risk from cancer.
422 F. Peter Guengerich

As pointed out earlier, the most generally results appear to be very mixed. An early report
accepted noninvasive human assay involves suggested a link of lung cancer with a polymor-
6-hydroxylation of the muscle relaxant chlorzoxa- phism^^"^, but since then the results have been mixed
zone^^^' ^^^. Studies with humans show little effect for cancers of the lung^^^"^^^, oral cavity^^^' ^^^, and
of diabetes^^^' ^''^, but an effect of body weight/ stomach^^^. In most of these cases, it should be
obesity^^^' ^^^. As mentioned before, genotype has emphasized that there is little information about
shown little impact on the in vivo parameters to exposure and the only relevant etiology is probably
dateH673 tobacco-derived nitrosamines. In a study of workers
Another issue is drug metabolism and toxicity. exposed to vinyl chloride (a P450 2E1 substrate^^"^),
Acetaminophen overdose remains a major cause some association was found between P450 2E1
of liver failure in the United States. Several P450s polymorphisms and p53 mutations^^"^. However, it
are involved in the oxidation to the reactive imi- should be emphasized again that the relevance of
noquinone^^^. Studies with P450 2E1 knockout CYP2E1 polymorphisms to known P450 2E1 reac-
mice indicate that P450 2E1 is a major determi- tions is unclear, particularly in vivo^^^, and it is diffi-
nant of acetaminophen toxicity, because the toxic- cult to define roles of these genetic polymorphisms
ity was considerably attenuated in null animals^^^. in cancer risk; overall P450 2E1 expression due to
P450 2El-null mice have the same blood environmental influences may have a role but is
ethanol levels as wild-type animals after ethanol more difficult to establish.
dosing^^^ suggesting that P450 2E1 activity is not
a major factor in ethanol metabolism, at least in 6.14. P450 2F1
mice. The situation regarding a role for P450 2E1
in alcohol-induced liver injury in other models is This is primarily a lung P450. In 1990,
unclear, with some reports suggesting a link^^"^' ^^^ Nhamburo et al.^"^^ cloned the cDNA from a
and others not^^^' ^^^. Autoantibodies against P450 human lung library. The level of expression
2E1 have been reported in alcoholics^^^ and attrib- appears to be relatively low, as judged by the
uted to hydroxyethyl radicals^^^ (which may arise mRNA abundance. The apparent orthologs 2F2
from lipid peroxidation processes rather than as and 2F3 have been studied in mouse and goat
intermediates in P450-catalyzed oxidation, vide lung, respectively.
supra). P450 2E1 is also a major autoantigen asso- P450 2F1 has been expressed in heterologous
ciated with halothane hepatitis, a rather idiosyn- systems. Catalytic activity was observed for
cratic response^^^. As with other autoimmunities 7-ethoxy- and -propoxycoumarin O-dealkylation
involving P450s, causal associations remain to be and 7-pentoxyresorufin 0-depentylation. The
demonstrated"^^^. enzyme showed modest activation of the lung toxin
Another issue is the contribution of P450 2E1 and (potential drug candidate) 4-ipomeanol^^^.
to oxidative stress. Ingleman-Sundberg reported However, the ability of P450 2F1 to activate the
that P450 2E1 contributed - 2 0 % of the NADPH- potential lung toxicants 3-methylindole, naphtha-
dependent lipid peroxidation in rat liver micro- lene^^^, and styrene^^^ is more impressive (the acti-
somes (and 45% in microsomes prepared from vation of 3-methylindole appears to involve initial
rats treated with acetone to induce P450 2E1)^^^. desaturation^^^).
Transfection of human P450 2E1 into a rat hepatic The basis for the selective expression of P450
stellate cell culture system yielded elevated pro- 2F1 in lung is unknown. Recently, Carr etal.^^^ iso-
duction of reactive species^^^ Cederbaum^^^ has lated the CYP2F1 gene. Using luciferase-based con-
reviewed studies on the relationship of oxidative structs, they identified a specific promoter element
stress to P450 in liver cell models. The exact rele- that binds a protein in the -152 to —182 5' region.
vance to liver injury and alcohol-induced disease This protein is termed a lung specific factor (LSF).
requires more investigation.
Many studies have been reported on the rela- 6.15. P450 2J2
tionship of CYP2E1 polymorphisms to risk of
diseases. Benzene poisoning in Chinese workers The P450 2J2 cDNA was first isolated from a
showed some changes in risk with one genotype but human liver library but was found to be most
only in smokers^^^. With regard to cancers, the highly expressed in heart^^^. Expression (mRNA)
Human P450s 423

has also been found in kidney and muscle^^^, No other information is presently available
lung^^^ and the gastrointestinal tract^^^. about P450 2S1.
Zeldin's group has done most of the work on this
P450, including the initial cloning and analysis of
tissue expression. Incubation of a recombinant P450 6.18. P450 2U1
2 J2 (plus reductase and NADPH) with arachidonic
acid yielded all four epoxides, that is, epoxy- As with some of the other human P450 genes,
eicosatetraenoic acids (EETs)^^^. These EETs were the only information presently available is the
found in heart tissue, and the stereochemistry of the identification of the CYP2U1 gene in the human
,705
recombinant P450 2J2 products was found to match genome
that of the compounds isolated from tissue. A num-
ber of physiological functions have been postulated
for the EETs, reviewed elsewhere^^^. 6.19. P450 2W1
The extent of human variability of expression No information is available at this time except
of P450 2J2 has not been reported. However, for the existence of the CYP2W1 gene in the
Zeldin's group has sequenced CYP2J2 genes and human genome^^^.
found a number of SNPs^^^. One was in the pro-
moter region, eight were exonic regions, five were
in introns, and four were in the 3'-untranslated 6.20. P450 3A4
region. Only four of the SNPs resulted in amino
acid changes. These allelic variants were expressed P450 3A4 is the most abundant P450 in the
in a baculovirus system; all had activity toward body (e.g.. Figures 10.2 and 10.4) and has a dom-
arachidonic and linoleic acids within a 2-fold level inant role in drug metabolism (Figure 10.3). Some
of wild-type P450 2J2, with the N404Y variant of the earliest preparations of human P450
showing only 10% catalytic activity (all assays (refs [3], [4]) were retrospectively found to be
only done at a 100 jxM substrate concentration), P450 3A4. Two approaches led to an extensive
although some qualitative changes in products characterization. Watkins et alP isolated a P450
were seen with the I192N substitution. The physi- from human liver using the criterion of immuno-
ological relevance of these substitutions is presently chemical cross-reactivity with what is now recog-
unknown. nized as a rat 3A subfamily P450; this laboratory
isolated an enzyme from human livers that cat-
alyzed the oxidation of the hypotensive dihy-
6.16. P450 2R1 dropyridine drug nifedipine^^. cDNA cloning
yielded sequences corresponding to CYP3A3
The only information available is the presence (ref. [707]) and CYP3A4 (ref. [708]). (The former
of the CYP2R1 gene in the human genome^^^. differed from CYP3A4 at 14 sites and could be
considered a rare allele, although it has not been
reported again^^^"^^^ and originally came from the
6.17. P450 2S1 same single-liver cDNA library as the CYP3A4
This gene was identified by searching data- clone; CYP3A3 has been dropped from the
bases by Rylander et alJ^^ mRNA and protein nomenclature and earlier references to this should
blotting work indicate highest expression in tra- probably be considered to indicate P450 3A4.)
chea, lung (and fetal lung), stomach, small intes- Subsequently, studies with microsomes, anti-
tine, and spleen. Expression was also relatively bodies, and purified P450 3A4 quickly indicated
abundant (mRNA level) in colon, appendix, liver, that nifedipine was not the only substrate; other
kidney, thymus, substantia nigra, peripheral substrates included other dihydropyridines^^^,
leukocytes, and placenta. Absolute levels of abun- steroids^^' ^^^, quinidine^^^, the oral contraceptive
dance are unknown. 17a-ethynylestradioF^, and the carcinogen afla-
Rivera et alP demonstrated that both mouse toxin Bj (ref. [714]). With more studies and the
and human P450 2S1 mRNA transcripts are application of recombinant systems, the repertoire
inducible by TCDD in cell culture. of substrates expanded rapidly^^^.
424 F. Peter Guengerich

6.20.1. Sites of Expression and extent because of difficulty in finding appropri-


Abundance ately responsive cells to utilize the CYP3A4 gene
and vector constructs derived from it. Guzelian's
P450 3A4 is the most abundant P450 in human laboratory reported that the source of liver cells
liver and in the small intestine. The average frac- was a greater issue than the CYP3A regulatory
tion of the total P450 in liver accounted for by region in comparing interspecies differences in
P450 3A4 is -25-30%28 (Figures 10.2 and 10.4); CYP3A gene regulation^^^, and this result can now
in the small intestine, the fraction attributed to be rationalized in the context of new knowledge
P450 3A4 is even higher. A study with the selec- about receptors (vide infra).
tive inhibitor gestodene, which destroys P450 Although most CYP3A subfamily genes are
3A4, indicates that P450 3A4 can constitute 60% inducible by dexamethasone, the classic glucocor-
of the total hepatic P450 (ref [716]). ticoid receptor was shown not to be involved in rat
P450 3A4 is also expressed in some extra- liver^^^. In early 1998, Maurel reported that the
hepatic tissues, including lung^"^"^' '''^, stomach, macrolide antibiotic rifampicin acted as a non-
colon^^"^, and adrenal (weak)^^l P450 3A4 does not steroid ligand and agonist of the human glucocor-
appear to be expressed in kidney, prostate, testis, or ticoid receptor, providing a possible mechanism
thymus, but other 3A subfamily P450s are^^^. The for regulation and a difference with the rodent sys-
literature is mixed on whether expression occurs in tems^^^. The interpretation of these conclusions
peripheral blood lymphocytes or not^'^' ^'^. was questioned by Ray et alP^.
A gender difference in P450 3A4 expression Shortly thereafter, Kliewer's group character-
does not appear to occur^^ and apparent pharma- ized the human homolog of a mouse receptor
cokinetic differences may be attributable to P- (PXR) that bound steroids and interacted with
glycoprotein, not P450 3A4 (ref. [55]). In fetal CYP3A subfamily genes in the manner expected
liver, P450 3A7 is the most abundant form and for a major regulatory influence^^^' ^^^ (some liter-
P450 3A4 expression is very low^^' ^^^. P450 ature also refers to the human PXR as "SXR").
3A4 expression increases rapidly after birth and This member of the steroid receptor family
reaches 50% of adult levels between 6 and "orphan" group interacted with barbiturates,
12 months of age^^^. Although many general steroids (including dexamethasone), statin drugs,
regulatory concerns have been expressed about macrolide antibiotics, and some organochlorine
additional safety margins for children with drugs pesticides^''^' -^^^
and other chemicals, the evidence in this case Knowledge of the PXR and its cognate binding
indicates that P450 3A4 activity levels in infants site has led to the development of PXR receptor
are slightly higher than in adults^^^. and reporter assays to screen for P450 3A4 induc-
P450 3A4 is expressed in some tumors, tion with new drug candidates^^"^"^^^. The discov-
although the literature is mixed as to reports of ery of the PXR receptor suggested that alleles of
levels lower and higher than the surrounding this receptor might be responsible for the variable
tisSUe^21-723 _
inducibility in different individuals. However, the
SNPs found to date have not been found to control
P450 3A4 induction^^^. The regulation of CYP3A4
6.20.2. Regulation and Polymorphism expression is more complicated than simple load-
The CYP3A4 gene is at chromosome 7q22.1 ing of activated PXR (e.g.. Figure 10.6), as sug-
(ref [724]). Although 3A subfamily enzymes gested by Kliewer's early work showing the roles
were long known to be inducible in animals'^^^ and of coactivators'^^''^^^. However, the glucocorticoid-
considerable literature existed on the in vivo mediated induction of P450 3A4 is mediated by
induction of many activities by barbiturates and elements in addition to the now-canonical PXR
macrolide antibiotics (e.g., rifampicin)"^^, early gjl^g738,739 Some compounds (e.g., ketoconazole)
demonstrations of inducibility were indirect but suppress CYP3A4 gene expression, apparently via
some progress was made^^, A general correlation binding to the PXR and interaction with "co-
between enzymes and mRNA levels could be repressors" (NCoR, SMRT)^^^. CAR (see Section
shown in human livers^^^' '^^^. Defining the 6.7.2) appears to interact with the CYP3A4 gene at
mechanism of regulation was difificult^^^, to some the PXR site and induce^'^^ Further, there is
Human P450s 425

evidence that la,25-dihydroxyvitamin D3 (see as lovastatin (Mevacor®) and other statins^^^, the
Section 6.53) also controls the transcription of prostate hypertrophy inhibitor finasteride
P450 3A4 (ref [742]). This effect is mediated (Proscar®/Propecia®)^^^, the immune suppressant
through the vitamin D receptor^"^^, which has sim- cyclosporin^^^' ^^^, protease inhibitors such as
ilarity to PXR and CAR in the steroid receptor indinavir^^^, and sildenafil (Viagra®)^^"^.
superfamily. Kinases have been shown to modu- In the course of these reactions, P450 3A4 cat-
late the induction of P450 3A4 via the vitamin D alyzes examples of some atypical reactions^^"^
receptor in Caco-2 cells^^^. including desaturation'^^^, oxidative carboxylic
Other factors also contribute to P450 3A4 reg- acid ester cleavage^^^, and oxidation of a nitrile to
ulation. Among these are C/EPPa and DBP^"^"^ and an amide''^^. An unexpected reaction encountered
PINF-4a (ref [745]). Interleukin-6 has been in this laboratory was the oxidation of alkylphenyl
reported to downregulate P450 3 A4 through trans- ether non-ionic detergents, which have been com-
lational induction of the repressive C/EBPp-LIP monly used in enzyme purifications^^^ and also
protein^"^^. Thus, the transcriptional regulation of have some medical and industrial applications^^^.
P450 3A4 expression centers on PXR but involves Methylene hydroxylations yield hemiacetals,
many other aspects. which break down to shorten the chains''^^.
Another aspect of P450 3A4 regulation involves One of the classic (and fastest) reactions cat-
degradation. Troleandomycin, erthyromycin, and alyzed by P450 3A4 is testosterone 6p-hydroxyla-
some related amine macrolide antibiotics form tion^^. However, the physiological significance of
"metabolite complexes" (C-nitroso: iron, R-N= this and other (P450 3A4-catalyzed) steroid
0:Fe) and inactive protein accumulates'^^^' ^^^. hydroxylations^ ^^ is unclear. The significance of
These studies have relevance to in vivo P450 3A4 P450 3A4 in physiology may be questioned,
inhibition by these drugs. given its variability (Figure 10.1 and Table 10.5).
P450 3 A4 appears to be degraded by a ubiquitin- However, some contributions are possible and may
linked pathway^^^. Correia's group also reported be suggested in recent work. Cholesterol is oxidized
that protein kinase C modified P450 3A4 at Thr264 by P450 3A4 to 4p-hydroxycholesterol, a major cir-
and Ser420; the relevance of these phosphorylations culating oxysteroF^^' ^^^. P450 3A4 also catalyzes
to ubiquitin-linked degradation is yet unknown^"^^. the 25-hydroxylation of 5p-cholestane-3a,7a,12a-
The issue of polymorphism is considered in trio^^^' '^^^ The product is a potent PXR agonist,
the context of attempts to explain the population and this system might function as an autoregulatory
variability in P450 3A4 activity, which does not pathway (i.e., excess triol activates PXR and P450
show true modality in its distribution'^^^. A num- 3A4, which reduces the level of trioF'^^).
ber of SNPs and other polymorphisms have been P450 3A4 also functions in the metabolism of
identified, but they have not shown much relation- cancer chemotherapeutic drugs. In addition, atten-
ship to catalytic activities yet'^^^"^^^. tion has been given to activations of drugs and
chemical carcinogens. P450 3A4 activates the
estrogen receptor antagonist tamoxifen to produce
6.20.3. Substrates and Reactions DNA adducts^^^. Another example of carcinogen
Analysis of the catalytic activity of P450 3A4 activation involves aflatoxin B^ which undergoes
and other 3A subfamily enzymes is not always both a detoxicating 3a-hydroxylation and forma-
easy to assess because of nuances about the effects tion of the highly mutagenic 8, 9-6xo-epoxide^^'^'
of the membranes and other proteins, as discussed 774,775 Some other carcinogen substrates of P450
in Section 6.20.4. Wrighton has examined P450s 3A4 are listed in Table 10.4.
3A4, 3A5, and 3A7 under identical conditions and One of the issues with P450 3A4 is which reac-
concluded that P450 3A4 is generally more tion provides the most appropriate index of activ-
catalytically active than 3A4 or 3A7 toward all ity, both in vitro and in vivo. Historically
substrates examined^^^. nifedipine oxidation and testosterone 6p-hydroxy-
P450 3A4 contributes to the metabolism of lation were among the first activities identified^^
—50% of the drugs on the market or under devel- and are still used in vitro^^. Midazolam 1 '-hydrox-
opment (Figure 10.3). For an extensive list, see ylation has also been used^^ in part because of its
Rendic^^. Many of these are important drugs such acceptance for in vivo assays.
426 F. Peter Guengerich

Some higher throughput fluorescence assays 6.20.4. Knowledge about Active Site
have also been developed and gained commercial
appear'^^' ^'^^. One issue regarding these and also Because of the importance of this enzyme in
several other P450 3A4 reactions is that they show drug metabolism (Figure 10.3), many efforts have
variable effects of added chemicals, that is, one dealt with developing a better understanding of its
compound may inhibit a certain P450 3 A4 reaction function and there is hope that intelligent predic-
but stimulate another. Chauret et alP^ reported a tions can be made regarding activities toward new
fluorescence reaction that behaves in a very similar drug candidates. However, as already alluded to,
way to testosterone 6p-hydroxylation. Houston has P450 3 A4 has some unusual properties and a num-
examined the behavior of P450 3A4 probe sub- ber of important issues have not been resolved.
strates in vitro and grouped them into two cate- In the early purifications of P450 3A4 (ref [12]),
gories. Although all of these reactions are catalyzed reconstitution conditions were difficult to optimize
by P450 3A4, they are categorized into two groups and showed some unexplained variations, which
by their behavior in the presence of other com- was also the case with recombinant enzyme^ ^^' ^^^.
pounds, as mentioned above^^^. One group includes A major factor was the composition of the
testosterone, cyclosporin, and erythromycin. The lipid/detergent environment^^^' ^^^. Another issue
second includes midazolam, triazolam, dextro- was the NADPH-P450 reductase. P450 3A4
methorphan, and diazepam. Terfenadine fits in either expressed in yeast showed poor coupling with
group and nifedipine seemed to have properties yeast NADPH-P450 reductase^'^ although P450
unique from both groups^^^. 2C9 had coupled welP^"^. Pompon developed a
yeast system in which yeast NADPH-P450 reduc-
The ambivalence about the variability of probe
tase and Z?3 were eliminated and replaced by the
drugs is even worse for in vivo human experi-
human counterparts, enabling P450 3A4 func-
ments than in vitro, as one might expect. A num-
tion^^^. Studies with purified P450 3A4 and
ber of reactions have been used including
NADPH-P450 reductase have shown that P450
nifedipine oxidation^^^, erythromycin iV-demethy-
3A4 reduction is generally slow in the absence of
lation^^^ lidocaine oxidation^^^, dapsone A^-
substrate and greatly enhanced by substrate^^^.
hydroxylation^^^, midazolam 1'-hydroxylation^^'*,
and quinine 3'-hydroxylation^^^. In most cases, The role ofb^ in P450 3A4 reaction is a some-
the test drug is administered orally for conven- what complex subject. Some reactions of purified
ience, except for some uses of erythromycin and P450 3A4 are stimulated by b^ but others are
midazolam (i.v). The ratio of urinary 63-hydroxy- j^Ql^799,800 With reactions that are influenced by
cortisol to Cortisol has also been used to assess b^ (e.g., nifedipine oxidation and testosterone
P450 3A4 function^^^. Many of the assays reflect 6p-hydroxylation), a role for b^ could be demon-
the activity of P450 3A4 in the small intestine, strated in human liver microsomes using antibod-
particularly with the drugs administered orally. ies^^'; stimulation by b^ can also be demonstrated
The erythromycin breath test (exhaled CO2 pro- with P450 3A4 heterologously expressed in
duced from the HCHO released in the reaction) is bacterial membranes"^^^' ^^^, although the presence
generally used to estimate hepatic P450 3A4 and of Z?3 does not seem as critical for function as with
has been used as an aid in selecting cyclosporin the purified systems^^^' ^^^. Other considerations
doses for liver transplant patients^^'^. The lack of suggest that the amount of b^ in heterologous
correlation of these indicators is still a problem in expression systems should not be an issue in the
the practical analysis of drug interactions^^^"'^^^. development of "relative activity factors" for
Some of the discrepancies are probably inherent in extrapolations from hepatic systems^^^. The
the nature of P450 3A4 itself (i.e., see in vitro mechanism of stimulation of P450 3A4 activities
assays, vide supra). Other issues involve the lack by Z?3 is still not clear. Pompon used a membrane
of coordinate regulation of hepatic and intestinal system and interpreted the results in the context of
P450 3A4 (ref [791]) and the activity of P- a "classical"^^^' ^^^ transfer of an electron to the
glycoprotein^^^, which shows some overlap in Fe02^^ complex^^^. However, the activities of
regulation patterns with P450 3A4 (ref [793]) and purified P450 3A4 were stimulated by apo-Z75
influence the availability of substrates to P450 (devoid of heme) as effectively as by b^
3A4 in both small intestine and liver. (ref [808]), and similar effects have now been
Human P450s 427

reported for several other P450s (refs [425], [809], appear to accumulate in P450 3A4 reactions^^^,
[810]). Although an alternate mechanism involv- these are not well characterized yet. One approach
ing rapid transfer of P450 3A4 heme to apo-Z)^ has to analysis of rate-limiting components of P450
been proposed^ ^^ evidence ruling out this mecha- (and other enzyme) reactions is the use of kinetic
nism has been presented^^^. deuterium isotope effects^^^' ^•^^. Interpretation of
A number of models of the P450 3 A4 protein kinetic isotope effects in enzymatic reactions is a
have been presented^^^' 8i3-8i5^ j^^g^ of which are complex subject, but a simple interpretation of a
based on homology modeling. At the time of this significant intermolecular noncompetitive hydro-
proof (April 2004), the Astex company has pub- gen isotope effect is that C-H bond-breaking is at
licly announced a crystal structure for P450 3A4, least partially rate limiting^^^. Despite the interest
presumably in the absence of ligands, but the in P450 3A4, relatively few kinetic deuterium iso-
information is proprietary and no details have tope effect studies have been reported. Obach^^^
been released. reported an isotope effect of only 1.3 on the
A number of site-directed mutagenesis studies hydroxylation of the drug ezlopitant, although
on the possible roles of individual residues have details regarding k^^^ and K^ are lacking and ftir-
been published. FhQ304^^^ and Ala305 (ref [817]), ther interpretation of this result is diff'icult. Work in
in the putative I-helix, are proposed to control this laboratory with 6-(i2-testosterone has some-
access to the catalytic center. Phe304 was also what surprisingly shown a low competitive isotope
implicated in the partitioning of aflatoxin B^ oxi- effect (^Fand ^(V/K) - 3 , J.A. Krauser and
dation (between 3a-hydroxylation and S,9-exo- F.P Guengerich, unpublished results). Testosterone
epoxidation)^^^. A role for Asn206 was also 6p-hydroxylation is one of the fastest reactions
proposed in the work with aflatoxin B j (ref [818]). catalyzed by P450 3A4 and one might expect less
Leu211 is also postulated to control the size of the masking of an isotope effect in a system in which
active site^^^. other steps are known to be occurring eff'iciently.
Another issue about considerations of predict- In some of the previous sections, compounds
ing sites and rates of P450 3A4 reactions deals have been mentioned that stimulate the catal3^ic
with models based on chemical reactivity. The activities of P450s after direct addition to the
concept has been proposed that P450 3A4 has a enzyme, as opposed to regulation of genes in cells
relatively open active site and that reactions are or animals. This process is referred to as "stimula-
influenced largely by the chemical lability of C-H tion" (as opposed to induction). The phenomenon
bonds^^^, and some commercial software systems has been recognized for some time with P450s
have utilized such concepts. This approach to P450 (refs [107], [109], [827]). Conney's group demon-
catalysis may have some utility, and substrate lin- strated the activation of benzo[a]pyrene 3-hydroxy-
ear free energy relationships have been exploited lation and aflatoxin B^ activation by aNF in human
in our own research with rat P450 2B1 (ref [821]). liver microsomes^^^' ^^^. Johnson also demonstrated
However, there are some concerns about exactly the enhancement of human liver microsomal 17(3-
how appropriate the predictions are beyond spe- estradiol 2-hydroxylation by aNF^^^. Subsequent
cific sets of substrates. Knowledge of rate-limiting work in this laboratory provided evidence that P450
steps in P450 3A4 reactions is still rather rudimen- 3A4 was the human liver P450 most stimulated by
tary, in part because of some of the various com- aNFi46, 714 P45Q 3A4 is the P450 about which
plications discussed here. The rate of transfer of most of the discussion about cooperative behavior
the first electron from NADPH-P450 reductase is has been given, although some reports of coopera-
slow in the absence of substrate, but quite rapid in tivity have appeared regarding P450s 1A2, 2B6,
the presence of substrate and an equimolar con- and 2C9 (Sections 6.7.4 and 6.9.4). In addition,
centration of NADPH-P450 reductase"^^^. In liver P450 2C19 may show stimulation by some
microsomes, the testosterone-stimulated rate of compounds (R. Kinobe, B.D. Hammock, and
P450 reduction appears to be faster than the over- E.M.J. Gillam, personal communication).
all rate of 6p-hydroxylation^^^, but conclusions are In considering cooperativity, two types will be
complicated by the inability to observe only the described, using a convention we"^^^' ^^^ have
P450 3A4 component of the reduction. Although adapted from Kuby^^"^. "Homotropic" cooperativ-
some apparent (FeO) intermediate complexes do ity refers to nonhyperbolic phenomena (either
428 F. Peter Guengerich

steady-state reaction kinetics or binding) seen more substrates), and many of the data can be fit
with the addition of a single compound to an to a model with this much freedom (basically
enzyme. The most typical type is a sigmoidal, or Michaelis-Menten expression with two values
"S-shaped" curve, which when analyzed by a Hill each for k^^^ and K^, or insert of proportionality
plot (v = V* S'^/iS + S^^]) yields a value for « > 1 factors before the parameters)^^^"^"*^.
(S^^ is an approximation of the usual K^, or "pos- Halpert's laboratory has changed a number of
itive cooperativity." Examples of "negative coop- the amino acids in P450 3A4, based mainly upon
erativity" are less common but documented (n < homology modeling, and found that several can
1) as in a case with rabbit P450 1A2 (ref. [214]). alter the homotropic and the heterotropic cooper-
The other phenomenon is "heterotropic coopera- ativity. These residues include Ala305 (ref [844]),
tivity," described above as "stimulation," where Leu211, Asp214 (ref [845]), Serll9, Ala370,
two compounds are added to an enzyme and one Ile301 (ref [846]), and possibly some others as
enhances the catalytic action of the enzyme on well^"^^. A general conclusion from much of this
the other. In some cases, both homotropic and work is that two or possibly three ligands co-
heterotropic cooperativity can be operative^^^. occupy the binding site and alter each other's
Homotropic cooperativity was seen in the oxi- juxtaposition to generate some of the observed
dation of aflatoxin Bj by P450 3A4 (ref [832]). effects. One problem with much of the work in
The previously reported stimulation of P450 3A4 this field is that actual binding phenomena are
activities by aNF^^"* was not seen for some reac- not necessarily investigated. However, binding has
tions, and the A^-oxygenation of 4, 4'-methylene- been analyzed in some of the work^^^' ^^^ and
Z?/5(2-chloroaniline) was inhibited^^^. Subsequently, shown to exhibit homotropic and heterotropic
studies with aflatoxin B^ oxidation showed that cooperativity. Further, combinations of binding
3a-hydroxylation was inhibited and 8,9-(exo)- and inhibition results obtained with several lig-
epoxidation was stimulated by aNF^^^' '^^^. ands in this laboratory were consistent with a
Aflatoxin B^ (or an analog) did not modify the scheme in which three ligand subdomains exist in
5,6-epoxidation of aNF, however. Interestingly, the overall binding site of P450 3A4 (ref [831]),
the positive cooperativity seen in Hill plots for the in agreement with the current hypotheses of
oxidation of aflatoxin B^ (for both reactions) was Halpert^"*^. More evidence consistent with such a
eliminated in the presence of aNF"*^^. The values model is available from a fluorescence study by
for n in the Hill plots (2.1-2.3) are probably the Atkins and Weinkers^"^^, in which pyrene-pyrene
highest for any P450 apparent cooperativity stacking spectra were observed. This work pro-
reported to date. Most are much lower. One tech- vides some of the stronger evidence to support the
nical issue of particular note is that those reactions "multiple-substrate site" model.
that proceed too far at low substrate concentra- A crystal structure of bacterial P450 107A1
tions will show apparently low rates (due to sub- has been solved with two ligand molecules pres-
strate depletion or product inhibition) and ent^"^^. The binding titration shows homotropic
artificial sigmoidicity can be created. cooperativity^"^^ and also some heterotropic coop-
Many seemingly unusual P450 3A4 reactions erativity^^^. Because the redox partner of P450
and patterns have been reported (vide supra). 107A1 is not known, obtaining reasonable cat-
P450 3A4-catalyzed testosterone 6p-hydroxyla- alytic activity is difficult and the relevance to
tion and erythromycin A^-demethylation are not P450 3A4 is still not definite^^^.
very competitive^^^. Hydroxylation of meloxicam Another aspect and possibly another solution
is stimulated by another substrate, quinidine^^^. to the issue comes from work by Friedman using
Lu and his group showed that inhibition patterns flash photolysis kinetics (of CO rebinding after
for several known P450 3A4 reactions were photodissociation from ferrous P450 3A4). The
substrate dependent^^^. Similar discrepancies kinetics were multiphasic and were selectively
were reported by Weinkers' laboratory^^^. The altered by the presence of different substrates^^^
(mechanism-based?) inactivation of P450 3A4 by Heterotropic effects were observed with benzo[<2]-
diclofenac was stimulated by the substrate quini- pyrene and aNF^^^. The interpretation of the
^jj^g838 Qj^g interpretation of some of the results results is that different substrates differentially
is that a single active site accommodates two (or modulate these kinetics by (a) changing the P450
Human P450s 429

conformation to alter the rate, and/or (b) steric already mentioned that different protein conforma-
effects (of ligands) that reduce rates^^^. Both tions exist throughout the catalytic cycle, can differ
effects are possible, although the enhancement of in affinities and substrate orientation, and may not
rates in some cases^^' argues against the general- be in rapid equilibria. Many steady-state kinetic
ity of the latter explanation and in favor of multi- schemes have been proposed but, in considering the
ple conformations for P450 3A4 bound to various possible origins^^"*' ^^^ can never be considered
ligands. The concept advanced is that some lig- unique and do not provide mechanistic answers.
ands act as allosteric factors to "switch" P450 3A4 The availability of a series of three-dimensional
conformations^^"^. Some possibly relevant work X-ray structures of P450 3A4 with various ligands
has been done by Anzenbacherova et al.^^^, who would provide insight into the conformational
did pressure studies on P450 3A4 and found that rigidity of P450 3A4 and the number of modes of
the compressibility of P450 3A4 was less than that binding. Another possible set of experiments
of bacterial P450 102; the compressibility was involves restraining conformational changes
modified by the ligand troleandomycin (TAO). through engineering (e.g., with reversible disulfide
The concept of preexisting multiple conformers bonds) and examination of the effects. Another
of P450 3A4 is an explanation for the flash concern, already expressed here, is that most of the
photolysis work^^^"^^"^ and has support in newer studies in this field have avoided measuring bind-
nonclassical approaches to general protein ing, with some exceptions'^ ^' ^^^^ ^^^. Some attempts
chemistry^^^^^^. This view differs from the more have been made to directly quantify P450 3A4-
general static "lock-and-key" view of enzyme/ ligand interactions (e.g., dialysis and equivalent
substrate complexes and the induced-fit theory in methods), but the technical problems associated
which enzymes are "shaped" by their substrates. with equilibrium binding (e.g., insolubility and
The basic concept is protein dynamics present an nonspecific components) are not trivial. At this
ensemble of structures of an enzyme in solution time a priori prediction of cooperativity is not
and different ligands bind to individual states really possible, except perhaps in extension of
depending upon their complementation^^^^^^. chemical classes already covered. The lack of abil-
Another consideration in this discussion, some- ity to predict P450 3A4 cooperative ligand interac-
what related, is that there is good evidence that tions indicates a deficiency in being able to predict
P450 conformations change during the course of all ligand interactions.
the catal3^ic cycle^^^, and evidence has already Is the cooperativity of P450 3A4 relevant to
been presented that different forms of P450 3A4 any practical issues? Atkins'^^ has discussed the
can differ in their binding of a ligand (e.g., ferric general implications of the issue to toxicology,
and ferrous)^^^ although conclusions are speculative because the
Where does all of the work in this area to date function of P450 3A4 can be good, bad, or not
leave us? A recent review by Atkins et al.^^^ sum- really selected for anyway. Some evidence for an
marizes much of the work in more detail and pres- interaction between caffeine and acetaminophen
ents a cogent analysis. Summarizing and expanding in rat models is suggestive of a heterotropic inter-
on this, there are several major possibilities to action^ ^^' '^^. Dextromethorphan studies in pri-
explain the observed cooperativity of P450 (and the mary hepatocytes also show cooperativity^^'.
other P450s showing this behavior), which are not Cooperativity in human hepatocyte systems has
necessarily exclusive: (a) a "classic" allosteric also been reported for oxidation of midazolam and
model with binding of effectors at a site that then warfarin'^"^. Cooperativity is a possible mecha-
regulates the conformation of substrate binding, nism for a drug interaction between felbamate and
(b) a relatively rigid P450 with a large active site carbamazepine'^^. One of the strongest cases
that can accommodate 2-3 ligands, with the results involves an in vivo study on the enhancement of
depending on the chemical interactions of the two diclofenac in monkeys by quinidine^^^' ^^^, which
ligands with each other and with P450 residues; apparently cannot be attributed to induction. In
and (c) a series of preexisting conformations of summary, there is some evidence for in vivo P450
P450 3A4 that selectively interact with individual 3A4 cooperativity, but at this time, the issue is
ligands^^^^^^. A general concept of induced fit is generally considered to be less of a problem than
related to the third possibility, as in the phenomena enzyme induction or inhibition.
430 F. Peter Guengerich

6.20.5. Inhibitors gestodene^^^. Because of the very low doses of these


contraceptives that are used today, the effects might
Inhibition of P450 3A4 is a major issue in the be expected to be small^^"^ although some in vivo
pharmaceutical industry because of a number of effects have been reported^^^' ^^^.
important drug-drug interactions. One example Finally, some chemicals and also oxidants have
of a problem leading to recall of a drug is that of been shown to cause the covalent crosslinking of
terfenadineio^'104,866
heme to apo-P450 (ref. [877]). Correia's group has
Erythromycin and ketoconazole are two of the characterized the products of the destruction of
most established inhibitors of P450 3A4, based on P450 3A4 with cumene hydroperoxide; the infor-
clinical experience. Ketoconazole, used at ~ 1 JULM, mation is consistent with a dipyrrolic fragment of
is probably the best established P450 3A4 inhibitor heme bound to a fragment of the protein^^^.
for in vitro use^^. Another P450 inhibitor is TAO^^^
which also has clinical implications. TAO has been
used as a diagnostic in vitro inhibitor of P450 3A4,
6.20.6. Clinical Issues
although its mode of action (activation to a nitroso
that complexes P450 iron) requires time for the The major issues involving P450 3A4 in drug
inhibition to occur. development and clinical use are related to the role
A compendium of P450 3A4 inhibitors has been of the enzyme in drug disposition, particularly
compiled by Rendic^^. Only a few other specific bioavailability and drug-drug interactions due to
examples of P450 inhibitors will be mentioned here. induction or inhibition^^^' ^^^. High enzyme activ-
One issue is the inhibition of P450 3A4 by ity toward a drug will reduce bioavailability, and
grapefruit juice, first reported by Bailey^^^. The variations in levels of P450 3A4 can cause clinical
effect was rather specific for grapefruit and a few problems when the therapeutic window is narrow.
other citrus fruits (not orange), and warning labels For instance, low cyclosporin levels will not pre-
now include this contraindication for many vent organ rejection during transplant but high lev-
(jj^gg869 Naringenin has some effect^'^^, but the els cause renal toxicity, so adjustment of the dose
most active principles appear to be the ftirano- can be very useful^^^ Terfenadine has a relatively
coumarins bergamottin and 6',7'-dihydroxyberg- wide window for use but a few serious problems
amottin, which behave as mechanism-based were encountered ^^'^' ^^^. Renwick has considered
inactivators to destroy intestinal P450 3A4 (refs population models of P450 3A4 variability and
[99], [100]). The magnitude of the effect of the inter- concluded that there is more inter-individual
action varies with drugs, with some of the statins, variability from the oral route than i.v, which is
buspirone, terfenadine, astemizole, and amiodarone not surprising in light of the previous discussion of
reported to show the greatest interactions^^^. the intestinal contribution to drug metabolism.
Many of the HIV protease inhibitors are also A "default factor" for adults of 3.2-fold is pre-
potent inhibitors of P450 3A4 as well as substrates sented, but a factor of 12(-fold) was calculated to
in some cases^^'. Because of the variety of drugs be needed to cover 99% of the neonates as well^^^.
that AIDS patients use, the potential for interac- The effbct of disease on P450 3A4 has
tions is considerable. been considered. P450 3A4 expression appears to
The effects of some herbal medicines on P450 be decreased as a result of liver cirrhosis or
3A4 have already been mentioned. In addition to cancer^^^' '^2'' ^^^. P450 3A4 levels were also
P450 3A4 induction (e.g., St. John's wort), some decreased in celiac disease and reversed by a
of these materials also contain inhibitors. For change in diet^^^.
instance, kava-kava extracts produce kavapyrones The interactions of herbal medicines with P450
that inhibit P450 3A4 (ref. [872]). 3 A4 have already been mentioned and are one of the
Oral contraceptives contain acetylenes and worst problems with these mixtures^^^. One of the
can be mechanistic inactivators of P450 3A4. most studied issues is St. John's wort, which induces
Inactivation has been demonstrated for 17a- P450 3A4 as an agonist of the PXR receptor^^^' ^^^.
ethjoiylestradiol, the major estrogenic component The induction of P450 3A4 by St. John's wort has
of oral contraceptives^^' ^^^, and several of been responsible for the loss of the effectiveness of
the progestogenic components, particularly oral contraceptives^^' ^^^. The resulting pregnancies
Human P450s 431

are the result of contraceptive failure due to more expressed^^^. Gonzalez found a liver sample appar-
rapid elimination of 17a-ethynylestradiol, a phe- ently expressing only P450 3A5 and not 3A4, and
nomenon previously reported for P450 3A4 induc- used this to clone the cDNA^^^.
tion by rifampicin and barbiturates^^' ^4, loi P450 3 A5 expression has been reported in liver,
P450 3A4 is also of some interest regarding small intestine, kidney, lung prostate, adrenal
cancer, regarding exogenous carcinogens, drugs gland, and pituitary^ ^^' 902-904 gome researchers
used to treat cancer, and metabolism of steroids or have reported expression of P450 3A5 in periph-
other compounds that may affect cancer risk or eral blood cells (and not P450 3A4)^^^ but others
response to chemotherapy. Some chemical carcino- have not^^^.
gens activated by P450 3A4 are shown in Table 10.4. P450 3A5 is expressed in fetal liver, in contrast
The activation and detoxication of aflatoxin B^ to P450 3 A4, but in a polymorphic manner^^^. The
have already been discussed in the context of overall expression of P450 3A5 (mRNA) as a part
3a-hydroxylation (to aflatoxin Q^) and formation of all P450 3 A subfamily transcripts has been esti-
of the highly reaction exO'S,9-QpoxidQ^^^' '^^^. mated at 2%^^^. However, only about 25% of
However, aflatoxin B^ is a hepatocarcinogen and Caucasians express P450 3A5, and when it is pres-
must reach the liver to cause damage. In a rat ent, the level is usually less than that of P450 3A4.
model, induction of rat P450 led to an increase in However, a few individuals have been identified in
small intestinal DNA adducts, suggesting that acti- which P450 3A5 is the predominant P450 3A sub-
vation of aflatoxin B^ at this site constitutes a family enzyme. The variability in expression levels
detoxication process, in that these cells are rapidly has been linked to a polymorphism (vide infra).
sloughed and do not progress to tumors^^^.
CYP3A4 genotypes have been reported to be
6.21.2. Regulation and Polymorphism
related to leukemias caused by prior treatment
with epipodophyllotoxin^^^ P450 3A4 expres- The regulation of CYP3A5 gene seems to be
sion, measured at the mRNA level, has shown an similar to that of CYP3A4, although P450 3A5
inverse correlation with response of breast cancer does not seem as inducible. The fetal/adult selec-
patients to docetaxel, presumably due to changes tivity of P450 3A4/3A7 is not seen with P450 3A5
in bioavailability^^^. However, no relationships (ref. [906]).
were found for any CYP3A4 genotypes in therapy- Maurel^^^ reported genomic clones and found
related myeloid malignancies^^^. One of the more a CATA box (not TATA) in the promoter. The
controversial issues involves whether CYP3A4 responses to glucocorticoids are probably
genotypes are linked with prostate cancer, with explained by the PXR system^^^. A general con-
reports for and against an association^^"^^^^. The clusion has been reached that P450s 3A4 and 3A5
point should be made that strong evidence for a are co-regulated in the liver and intestine, in terms
change in an accepted P450 3A4 phenotype has of transcriptional control^^^, although other
not been made in many of these cases. factors may alter the expression^^^
The polymorphic variation of P450 3A5 has
6.21. P450 3A5 been studied and several variants have now been
identified (http://www.imm.ki.se/Cypalleles/)^^^.
P450 3A5 has 85% sequence identity with Alternate splicing is a very common phenomenon
P450 3A4 and, although generally accepted to with CYP3A5, with ~50% of liver samples show-
have less importance than P450 3A4, is of interest ing this (E.G. Schuetz, personal communication).
because of its polymorphic and racial distribution Most Caucasians with low P450 3A5 protein
and possible relevance to clinical issues with P450 expression have the *3 allele with an inserted
3A subfamily reactions. intron^^^' ^^^. Interesting, the representation of the
*1 allele is much higher in Africans and they
express active P450 3A5. Other alleles are known,
6.21.1. Sites of Expression and
including changes in the 5'-regulatory region
Abundance where transcription factors bind^^^
P450 3A5 ("Hlp3") was first purified from The in vivo consequences of 3A5 polymor-
human adult liver and found to be polymorphically phism are not clear. For instance, Huang found no
432 F. Peter Guengerich

significant effect of the *3 polymorphism on mentioned previously, Huang^^^ found no signifi-


midazolam pharmacokinetics^^^. cant effect of the *3 allele on midazolam pharma-
cokinetics in Chinese individuals. However, it is
possible that the extrahepatic expression^ ^^ may
6.21.3. Substrates and Reactions influence the course of particular drugs and other
Since the discovery of P450 3A5, the catalytic chemicals.
selectivity has been known to be similar to that of
P450 3A4 (ref [900]), and subsequent compar-
isons with P450 3A4 confirmed this view^^^. 6.22. P450 3A7
However, a general problem with P450 3A sub- Early work in the field of human P450
family enzymes is that they are sensitive to the research was done by Kamataki and his associates
membrane environment and many reactions of with fetal samples which led to the purification of
P450 3A5 (but not all) are stimulated by b^ (refs a P450 termed HFLa, now known as P450 3A7
[425], [800]). In a few cases, the selectivity of P450 (refs [915], [916]). Early research established that
3A5 for different oxidation sites appears to differ this was a major P450 in fetal liver (not in adult
from that of P450 3A4, for example, aflatoxin Bj liver), and that the enzyme could catalyze several
3a-hydroxylation vs 8,9-epoxidation^^^' ^^^. reactions^ ^^.
Recently, as noted above, Wrighton reported
an extensive comparison of many reactions by
recombinant P450s 3A4, 3A5, 3A7 under identi- 6.22.1. Sites of Expression and
cal reconstitution conditions and concluded that Abundance
P450 3 A5 had equal or reduced activity compared
to P450 3A4 in all cases^^l Early work established that P450 3A7 is the
major P450 present in fetal liver^^^ and is also
present in other fetal tissues including kidney,
6.21.4. Knowledge about Active Site adrenal, and lung^'^. Further work by Kamataki's
group showed the existence of some immuno-
Because of the similarity of reactions of P450s
chemically detectable P450 3A7 in gynecologic
3A4 and 3A5, homology models for P450 3A4 are
tumors and in human placenta, but interestingly
probably about as valid for P450 3A5. The avail-
not in cynomologous monkey placenta^^^.
ability of the Astex P450 3A4 three-dimensional
Guzelian's group also reported P450 3A7 protein
structure should improve the understanding of
in human placenta and endometrium, with eleva-
P450 3A5 as well. Relatively little site-directed
tion in the latter site during pregnancy or during
mutagenesis of P450 3A5 has been done, but one
the secretory phase of the menstrual cycle^^^.
study of note is the effort by Correia and Halpert
Subsequently, Sarkar et al?'^^ reported 10-fold
to utilize the differences in reactions with afla-
greater expression of P450 3A7 in endometrium
toxin B, (refs [774], [800]) to probe the effects of
in the proliferative rather than the secretory phase.
changing residues in the putative active site^^^.
Hakkola et alP^^ reported some expression of
P450 3 A7 mRNA in some first trimester placentas
6.21.5. Inhibitors but not in fiill-term placenta^"*^.
With regard to development in fetal tissues,
In general, the P450 3A4 inhibitors also inhibit Juchau's group found expression of P450 3A7 in
P450 3A5. For instance, ketoconazole and flu- early fetal tissue (50-60 days)^^^. Schuetz et al.^^^
conazole inhibited both P450s 3A4 and 3A5 (ref found P450 3A7 mRNA in all fetal liver samples
[914]). The mechanism-based inactivator gesto- analyzed and also reported its presence in one half
dene^^^ also inhibits P450 3A5 (ref [913]). of adult liver samples. However, the issue may
be the level of expression because Kamataki's
group^^ had reported the fetal > adult selectivity.
6.21.6. Clinical Issues
De Wildt et al™ also found fetal specificity and
At this point, the significance of the wide only very low levels of P450 3A7 in adults. P450
variability in P450 3A5 is difficult to assess. As 3A7 expression was high during embryonic and
Human P450s 433

fetal life, and decreased rapidly during the first Some interesting variants of CYP3A7 genes
week of life. Similar findings were reported by have been reported. An mRNA species was found
Hakkola et al?^^ Also, the variability of P450 3A7 that contains exons 2 and 13 of a nearby CYP3A
expression was 5-fold in fetal tissue (and 77-fold pseudogene spliced at the 3 end^^^. The CYP3A7*
in mRNA). In another report^24^ P45Q 3^7 ^^so IC allele is unusual in the sense that a part of the
disappeared rapidly after infancy. CYP3A4 promoter replaces the corresponding
region of CYP3A 7 (ER6 motif) and thus confers
high levels of expression to CYP3A7*1C
(ref [932]).
6.22.2. Regulation and Polymorphism
As in the case of P450 3A4, relatively little
solid evidence is available regarding the ftinc- 6.22.3. Substrates and Reactions
tional relevance of coding region SNPs. However,
the regulation of this gene is complex, as one Early studies with P450 3A7 purified from
might expect after considering the temporal pat- fetal liver established that testosterone 6p-hydrox-
terns of expression during development that were ylation is catalyzed by this enzyme^^^. Another
discussed earlier. early study indicated 16a-hydroxylation of dehy-
Kamataki's group published the cDNA^^^ and droepiandrosterone (DHEA) 3-sulfate^^'*. These
genomic^^^ sequences, which are similar to those activities were later verified with the use of
of P450 3A4. However, more identity (-90%) is recombinant P450 3A7 (ref. [935]).
seen in the coding region than elsewhere^^^' ^^^. In general, P450 3A7 has catalytic activities
Recent work by Koch et aV^^ re-established that rather similar to P450 3A4 and 3A5 (refs [936],
P450 3A7 only accounted for <2% of all P450 937]). Activation of aflatoxin B^ (refs [938-940])
expression in adult human liver; a bimodality of and heterocyclic amines^^^ has been observed in
P450 3A7 expression was seen, however. P450 various recombinant and transgenic systems,
3 A4 and 3 A7 constructs were expressed in various including transgenic mice^^^ Retinoic acid
cell lines by Ourlin et aV^^, who showed differen- 4-hydroxylation by P450 3A7 has also been
tial responses to C/EBPa and DBP As in the case reported^"^^. Wrighton's laboratory has done an
with P450 3A4, P450 3A7 was inducible by extensive comparison of catalytic activities and
rifampicin in cell culture^^^. P450 3A7 has a ftmc- concluded that rates for P450 3A7 are generally
tional PXR element^^s^ ^^ ^^^^ P450 3^4 {vide considerably lower for P450 3A7 than for P450
3A4 or 3A5 under similar conditions^^^.
supra), explaining the rifampicin response. Thus,
one would expect fetal P450 3A7 induction by the
usual P450 3A4 inducers.
Bertilsson et al?^'^ have also reported a distal 6.22.4. Knowledge about Active Site
xenobiotic response enhancer module (XREM) in Much less has been done with P450 3A7 than
the CYP3A7 gene. An 3A7^B element in CYP3A7 with P450s 3A4 and 3A5. Because the catalytic
is inactive in CYP3A4 (ref [930]), and this ele- selectivity of P450 3A7 is similar to P450s 3A4
ment has recently been shown to respond to p53 and 3A5, those models are probably about as
(T. Kamataki, personal communication). CYP3A7 applicable. One point of interest is the work of
expression is regulated by Spl, Sp3, HNF-3P, and Kamataki's group showing that the substitution
upstream stimulatory factor (USF) 1. Far upstream T485P improved holoprotein expression in
(-11 kb) there are HNF-1 and HNF-4 and USFl
elements, which differ from the CYP3A4 gene.
Exactly how these and other sequence differences
are involved in the rapid onset of P450 3A4, and
6.22.5. Inhibitors
decrease in P450 3A7 shortly after birth^^ is still not
totally clear. Recent work in Kamataki's group sug- Inhibitors have not been studied extensively,
gests that after birth, CEB/Pa recruits an uncharac- but presumably all inhibitors of P450 3A4 are eff-
terized protein factor to squelch the 3A7^B site ective with P450 3A7, for example, ketoconazole,
(T. Kamataki, personal communication). troleandomycin, etc.
434 F. Peter Guengerich

6.22.6. Clinical Issues keratinocytes^^^. In cell cultures, P450 4A11


expression has been observed in primary cultures
The general point has already been made that of human kidney proximal tubular cells^^^ and
P450 3A7 is the major human fetal P450 and, HepG2 cells956.
therefore, makes a major contribution to drug A gene originally assigned as CYP4A11 by
metabolism in the fetus. Thus, many, if not most, Kawashima's group^^^ has now been assigned
of the considerations regarding drug interactions as that corresponding to CYP4A22 and replaced
etc. with P450 3A4 should be considered with by the CYP4A11 gene corresponding to the P450
respect to the fetus during pregnancy. 4A11 CDNA952_
Another potentially important aspect is a
report that P450 3A7 expression increases in
hepatocellular carcinoma^"^^, possibly as a part of
6.24.2. Regulation and Polymorphism
dedifferentiation.
The levels of hepatic P450 4A11 vary
~ 10-fold among humans^^^' ^^^. To date no
6.23. P450 3A43 polymorphisms have been reported (April 2004;
In 2001, three groups reported the characteri- http://www.imm.ki.se/CYPalleles/). The regula-
zation of a fourth member of the 3A subfamily, tion of P450 4A11 expression is not well under-
P450 3A43 (refs [945-947]). The sequence iden- stood but has relevance in consideration of the
tity with the other 3A subfamily proteins is peroxisome proliferation system and any rele-
71-76%. Expression could be detected in liver, vance to cancer. Induction of P450 4A11 by per-
kidney, pancreas, and prostate. Rifampicin was oxisome proliferators has not been seen in
reported to induce P450 3A43 in human liver^"^^. primary human hepatocytes cultures, although
The level of expression in liver was generally P450 4A11 expression is readily detected^^^. In
agreed to be very low ( - 0 . 1 % of P450 3A4). confluent HepG2 cell cultures, P450 4A11 is
Heterologous expression was achieved in bac- induced (independently) by peroxisome prolifera-
teria^"^^ but not any of several eukaryotic sys- tors (e.g., Wy 14643) and dexamethasone^^^. The
tems^"^^. The recombinant protein had only very relevance of these results to the induction in vivo
low testosterone 6p-hydroxylation activity^"^^. and the observed variability in expression is
No information is available about polymor- unknown.
phisms, although the transcripts appear very prone
to splicing^'*^. There is a general agreement that
P450 3A43 makes little contribution to drug 6.24.3. Substrates and Reactions
metabolism, but specialized roles in extrahepatic
P450 4A11 is the major lauric acid w-hydrox-
tissues may be possible.
ylase^^^' ^^•' ^^'^^ ^^^' ^^^. The enzyme also catalyzes
(0- and some co-l hydroxylation of myristic and
6.24. P450 4A11 palmitic acids^^^' ^^^. Some papers have reported
arachidonic acid hydroxylation^^^' ^^'' ^^^, but
6.24.1. Sites of Expression and most studies have not associated this activity with
Abundance P450 4A11 at any appreciable level^^^, 957, 960
Oliw's group has reported some hydroxylation
P450 4A11 cDNAs were first cloned from of prostaglandin H2 and analogs by P450 4A11
human kidney cDNA libraries using rodent P450 (ref [963]).
4A probes^'^^-^^^. P450 4A11 is now known to be
the major lauric acid w-hydroxylase in human
liver and kidney^^^' ^^^, a fact of some historical
6.24.4. Knowledge about Active Site
interest in the sense that this was the activity first
utilized in the separation and reconstitution of Some information is available about the active
(rabbit) P450 (ref [953]). The exact level of site from the catalytic selectivity among fatty acid
expression in these tissues is unknown; P450 substrates^^^. Some homology models have been
4A11 expression has also been reported in human presented^o^'964.
Human P450s 435

The interesting observation was made that the 4A22 in human liver^^^. P450 4A22 expression
LI3IF mutant catalyzes only w-l hydroxylation could not be observed in HepG2 cells or PPARa-
and not oo-hydroxylation of lauric acid^^^. Residue overexpressing cells^^^.
131 also controlled access to substituted imida-
zole inhibitors. Interestingly, some of the results
on binding of imidazoles provide evidence that the
ferric enzyme undergoes a conformational change 6.26. P450 4B1
that depends on both reduction of the iron and the
6.26.1. Sites of Expression and
presence of both NADPH-P450 reductase and
Abundance
NADPH959.
Another interesting observation is that P450 P450 4B1 was cloned by Nhamburo et alP^^
4A enzymes, including P450 4A11, show at least from a human lung cDNA library. P450 4B1 expres-
partial covalent heme attachment^^^. Covalent sion has also been reported (in addition to lung)
heme binding involves a conserved I-helix glu- in kidney, bladder^^^, breast^^^, and prostate^^^.
tamic acid (apparently unique in the P450 4A sub- Expression has also been reported in bladder and
family) and covalent heme binding occurs via an breast tumors^^^. Definitive information about the
ester bond to the heme 5-methyl group, mediated level of expression of P450 4B1 is not available.
by an autocatal3^ic process^^^. The extent of effect
of this modification on catalytic activity is diffi-
cult to define, although with animal P450 4A 6.26.2. Regulation and Polymorphism
enzymes, there appears to be some effect.
The extent of variability of P450 4B1 expres-
sion is considerable, at least in bladder where the
6.24.5. Inhibitors variation is two orders of magnitude^^^. Several
SNPs have been reported, including one resulting
Substituted imidazoles have been used as in a premature truncation^^^.
inhibitors in vitro^^^. Presumably some acetylenic No evidence for the inducibility of human 4B1
fatty acids might be inhibitors but no studies have has been presented.
been reported.

6.26.3. Substrates and Reactions


6.24.6. Clinical Relevance
Direct information about the catalytic speci-
The significance of P450 4A11 is not very
ficity of human P450 4B1 has been difficult to
clear. Apparently individuals can vary in their
obtain because of problems in heterologous expres-
expression levels by an order of magnitude^^^.
sion. Following the initial cDNA cloning, expres-
Further, the w-hydroxylation of medium chain
sion in a baculovirus system was unsuccessful and
fatty acids occurs, but its relevance is generally
only yielded inactive cytochrome P420 (ref. [971]).
considered not to be as important as in the case of
The substitution S427P allowed for expression, and
long chain fatty acids.
o)-hydroxylation of lauric acid could be demon-
strated. However, information about the native
human enzyme has not been available (the S427P
6.25. P450 4A22
mutant does not occur naturally^^^).
Relatively little is known about P450 4A22. Imaoka et alP^^ found that functional P450
The originally reported CYP4A11 gene^^^ was 4B1 could be successfully expressed as a fusion
subsequently shown to be CYP4A22 (ref. [952]), protein with NADPH-P450 reductase. They were
but the cDNA and protein have not been reported. also successful in developing transgenic mice in
The similarity of the two genes is 95%. which fiinctional P450 was expressed in liver. The
Johnson's laboratory^^^ has reported that P450 authors postulate that expression in the presence
4A22 is expressed at lower levels than P450 4A11 of auxiliary proteins (NADPH-P450 reductase,
in human liver, as well as kidney^^^. There was no b^) may stabilize P450 4B1 (ref [972]). With
correlation of expression levels of P450 4A11 and these systems, it was possible to demonstrate that
436 F. Peter Guengerich

P450 4B1 catalyzes the iV-hydroxylation of expression was distinct from P450 4F3, which is
2-aminofluorene and w-hydroxylation of lauric acid, restricted to polymorphonuclear leukocytes. P450
as expected from studies with rabbit P450 4B1. 4F2 is found not only in liver but in several extra-
If other results from work with animal P450 4B1 hepatic tissues, however^ •^^, including kidney (S2
enzymes also carry over to the human QnzymQS, one and S3 segments of proximal tubules, in cortex
might expect the reaction 4-ipomeanol activation and outer medulla). The extent of variation of
(epoxidation?), 3-methoxy-4-aminoazobenzene P450 4F2 in human liver was -S-fold^^^.
7V-hydroxylation, 2-aminoanthracene A^-hydroxyla- P450 4F2 catalyzes co-hydroxylation of several
tion, valproic acid hydroxylation (and desatura- lipids, including leukotriene B^ (refs [979], [980]),
tion?), and dehydrogenation of 3-methylindole^^^. arachidonic acid^^^, 6-^ra«5-leukotriene B^, lipoxin
A^, 8-hydroxyeicosatetraenoic acid, 12-hydroxy-
eicosatetraenoic acid, and 12-hydroxystearic
6.26.4. Knowledge about Active Site acid^^^ The physiological relevance of some of
Because of the paucity of information about these reactions is of interest but the effects of vari-
catalytic selectivity (vide supra), little can be said ability of P450 4F2 have not been demonstrated.
about the active site of human P450 4B1. Some Part of the interest lies in the fact that leukotriene
kinetic hydrogen isotope effect work with rabbit B4 is a potent proinflammatory agent^^^' ^^^.
P450 4B1 suggests that the active site is more
restricted than that of P450 2B1 (ref [973]).
Another interesting observation with rabbit P450 6,28. P450 4F3
4B1 is the covalent linking of the heme to the pro-
tein^^^, a phenomenon observed with several of In 1993, Kikuta et al!^^^ cloned a P450 now
the P450 4 family proteins^^^' ^^^ Whether this known as P450 4F3 from a human leukocyte
binding is seen in human P450 4B1 is unknown. cDNA library. The protein was expressed in a
yeast vector system and was shown to catalyze
leukotriene B^ a)-hydroxylation. The K^ (0.7 JULM)
6.26.5. Inhibitors was much lower than that reported for P450 4F2
for this reaction (although the k^^^ and k^JK^
No inhibitors of human P450 4B1 have been
values have not been carefully compared)^^^.
reported.
The gene was cloned in 1998^^^. Interestingly,
the CYP4F3 gene has been shown to use tissue-
6.26.6. Clinical Issues specific splicing and alternate promoters. A 4F3A
form contains exon 4 (but not 3) and is expressed
There are two issues with P450 4B1. First, the in neutrophils; a 4F3B form contains exon 3 (but
enzyme has been shown to activate carcinogens, not 4) and is expressed in fetal and adult liver and
for example, 2-aminofluorene, and could be a risk kidney, trachea, and gastrointestinal tract^^"^' ^^^.
factor in bladder cancer^^^. The level of P450 4B1 The K^ of the 4F3B (liver) form was 26-fold
in tumorous tissue was not higher than in the sur- higher than for the 4F3A (neutrophil) form^^^,
rounding tissue, levels of bladder P450 4B1 were although the significance of this report is quali-
higher in tumor patients than in controls^^^. fied by the absence of k^^^ or k^JK^ parameters.
The other aspect is the use of P450 4B1 as a Further studies by Soberman's group have shown
means of drug delivery. Rabbit P450 4B1 has been that the substitution of exon 3 changes the cat-
utilized as an experimental transgenic activation sys- alytic selectivity from leukotriene B^ to arachi-
tem in the activation of 4-ipomeanol and 2-amino- donic acid (w-hydroxylation in both cases)^^^.
anthracene, to date only in cell culture models^"^^. Again, the usefulness of the observation is limited
by the lack of kinetic parameters. The relevance of
the preferential localization^^^ and altered cat-
6.27. P450 4F2
alytic selectivity are presently unknown but there
The Kusunose laboratory reported the cloning is potential clinical relevance in light of the known
of a human liver cDNA corresponding to the physiological action of both leukotriene B4 and
leukotriene B^ o)-hydroxylase^^^. The site of 20-hydroxyeicosatetraenoic acid.
Human P450s 437

6.29. P450 4F8 20 (ref [991]), hydroxylation of the antihistamine


ebastine^^^, and w-oxidation of leukotriene B^
Bylund et al?^^ used degenerate PCR primers (refs [990], [991]), and w-hydroxylation of some
and isolated a cDNA from human seminal vesi- prostaglandins and prostaglandin analogs^^^.
cles, denoted P450 4F8. This P450 was shown to No further information is yet available about
be a 19-hydroxylase with prostaglandin endoper- the relevance of this enzyme in physiological or
oxides^^^' ^^^ Recombinant P450 4F8 catalyzed the clinical situations.
a)-2 hydroxylation of arachidonic acid and three
stable prostaglandin H2 analogs but prostaglandins
D2, Ej, E2, and F2^ and leukotriene B^ were poor 6.32. P450 4F22
substrates^^^. (19^)-Hydroxy prostaglandins E^
No information is available except the exis-
and E2 are the main prostaglandins of human sem-
tence of the CYP4F22 gene in the human
inal fluid. Bylund et al.^^'^ propose that a)-2
genome^^^.
hydroxylation of prostaglandins H^ and H2 by
P450 4F8 occurs in seminal vesicles, and that iso-
merization to (19/?)-hydroxy prostaglandin E is the 6.33. P450 4V2
result of action of prostaglandin E synthase.
Further investigations by Bylund and Oliw^^^ No further information is available except for
have demonstrated the expression of P450 4F8 the existence of the human CYP4V2 gene^^^.
protein in human epidermis, hair follicles, sweat
glands, corneal epithelium, proximal renal
tubules, and epithelial linings of the gut and uri-
6.34. P450 4X1
nary tract. P450 4F8 was shown to be upregulated Relatively little is known beyond the existence
(mRNA and protein) in the epidermis in psoria- of the human CYP4X1 gene except for one recent
sis^^^. The exact physiological role of P450 4F8 is paper on rat P450 4X1 (ref [993]). mRNA expres-
unclear, although 19-hydroxy prostaglandins do sion was highly selective in brain (cortex, hip-
have several biological activities^^^. pocampus, cerebellum, brainstem). The rat protein
was expressed in yeast but has not been examined
for catalytic activity.
6.30. P450 4F11
P450 4F11 is another member of the CYP4A 6.35. P450 4Z1
gene cluster found on chromosome 19^^^.
Expression has been demonstrated primarily in The only information available is the existence
liver, with some expression also in kidney, heart, of the CYP4Z1 gene in the human genome^^^.
and skeletal muscle. No other information is
presently available, although it might be expected
to be capable of leukotriene hydroxylation based 6.36. P450 5A1
upon its similarity to other P450 4F enzymes. P450 5A1 is the classification of thromboxane
synthase, which converts prostaglandin R^ ^^
thromboxane (Figure 10.12). Thromboxane
6.31. P450 4F12 causes vasoconstriction and platelet aggregation,
P450 4F12 was originally cloned from human which are of considerable interest.
liver^^^ and small intestine^^^ cDNA libraries.
Expression has been demonstrated in liver, kidney, 6.36.1. Sites of Expression and
colon, small intestine, and heart^^^' ^^^ Actual lev-
Abundance
els of abundance are unknown, although this
would appear to be a minor P450. P450 5A1 is expressed in platelets and also
Two groups have expressed P450 4A12 in erythroleukemia cells^^^. The enzyme is also
yeast. Catalytic activities include the hydroxylation found in human monocytes^^^, leukocytes^^^, and
of arachidonic acid at carbons 18 (ref [990]) and kidney interstitial dendritic reticulum cells
438 F. Peter Guengerich

P450 5A1
a
O,,

P450 8A1

Fe'V-Q

'<x: homolytic
0 - 0 cleavage
Fe'v-d

Fe'^a
rearrangement
to C» radical
R2
electron -Fe"
transfer

carbocatlon
formation
+

o.O
stabilization
to product HO"

PGI2

Figure 10.12. Rearrangement of prostaglandin H2 to prostacyclin (PGI2) by P450 8A1 and thromboxane (TXA2)
by P450 5A1 (ref. [994]).

surrounding the tubules^^^. Some expression is factor NF-E2 is critical for both alteration of the
also seen in lung and liver^^^. nucleosomal structure and activation of the P450
5A1 promoter^ooo
Chevalier e^ a/.'^^' identified 11 polymorphic
6.36.2. Regulation and Polymorphism variants in the CYP5A1 gene, including 8 mis-
As one might expect from its function, P450 sense changes in the coding region. The effects of
5A1 is a highly regulated system. Dexamethasone these changes have not been reported yet.
induces P450 5A1 in human monocytes^^^.
Phorbol esters also induce P450 5A1 (e.g., \2-0- 6.36.3. Substrates and Reactions
tetradecanoylphorbol-13-acetate) in human ery-
throleukemia cells^^^. Patients with systemic The thromboxane synthase reaction has been
sclerosis showed 6-fold enhanced levels of leuko- known for many years but was associated with a
cyte P450 5A1 (ref. [997]). P450 by Ullrich and his associates, first in spectral
Promoter analysis indicates a 39-bp core pro- studies ^^^^ and then by purification^^^^. With
moter, containing TATA and initiator elements that the purified enzyme or one expressed in
control transcription. Binding of the transcription a baculovirus system^^^^, prostaglandin H2 was
Human P450s 439

converted to thromboxane A2 and 12-hydroxyhep- search for inhibitors as drug candidates has
tatrienoic acid (HHT) plus malondialdehyde, in continued^^^^.
equimolar amounts ^^^^. Prostaglandin G2 was
transformed to malondialdehyde and the corre-
sponding 15- and 12-hydroperoxy products. 6.36.6. Clinical Issues
Prostaglandin Hj was enzymatically transfor- As indicated earlier, platelet aggregation due
med into 12(Z)-hydroxy-8, 10-heptadecadienoic to the produced thromboxanes is important but
acid, and prostaglandin H3 yielded thromboxane overproduction can yield plugs, so control of
B3 and 12(L)-hydroxy-5,8,10,14-heptadecate- homeostasis is desirable.
traenoic acid^^^^ (Figure 10.12).
These are all rearrangement reactions, not
involving input of O2 or electrons from pyridine 6.37. P450 7A1
nucleotides. The reaction of the "oxygen-surro-
P450 7A1 catalyzes cholesterol 7a-hydroxyla-
gate" iodosylbenzene with a P450 5Al-containing
tion, the rate-limiting step in bile acid synthesis.
preparation and the stable prostaglandin H2 analog
Much has been done in several animal models,
15(*S)-hydroxy-l la,9a-epoxymethano-5(Z), 13(E)-
including purification of the enzyme from rabbit
prostadienoic acid (U46619) yielded three oxida-
and rat liver^^^^' ^^^^. The enzyme was partially
tion products (that could also be formed in a
purified from human liver^^^^ and the cDNA was
similar system using rat liver microsomes)^^^^. cloned by several groups in 19901016-1018
These and other studies led Hecker and Ullrich^^^''
to propose a mechanism involving homolytic
cleavage of the prostaglandin endoperoxide (with 6.37.1. Sites of Expression
the Fe^^ bonded to one oxygen and the other oxy-
gen bearing a radical), transfer of the radical to a Apparently the only site of P450 7A1 expres-
carbon, further electron transfer to generate Fe"^ sion is the liver. The CYP7A1 gene is on chromo-
plus a carbocation, and collapse of the bis-ionic some 8 q l l - q l 2 and contains recognition
structure to yield thromboxane A2 (ref [994]). sequences for a number of liver-specific transcrip-
Fragmentation competes with the electron transfer tion factorsioi9-io2i
step to also yield malondialdehyde and hepta- The level of the enzyme in liver appears to be
trienoic acid^^^. similar to some of the low-to-moderately abun-
dant xenobiotic-metabolizing enzymes in liver.

6.36.4. Knowledge about Active Site


6.37.2. Regulation and Polymorphism
Relatively little is known about the active site
The regulation of the CYP7A1 gene is very
of P450 5A1 beyond th^ information about the
complex, as might be expected from the important
reactions presented above. As indicated, the pro-
physiological role this enzyme plays.
tein does not bind NADPH-P450 reductase.
P450 7A1 activity has long been known to be
Presumably, the active site is rather specific,
upregulated by dietary cholesterol in most animal
although iodosylbenzene could be utilized as an
models^^^^, although there are some excep-
oxygen surrogate.
tions ^^^^. Feeding rats the competitive inhibitor
7-oxocholesterol led to reduced bile acid synthesis
6.36.5. Inhibitors (due to inhibition) and a compensatory increase
in P450 7A1 synthesis^^^s chiangi024 identified
Thromboxane synthase inhibitors have been a a bile acid-responsive element in the CYP7A1
matter of interest for many years because of their promoter.
potential use in preventing plugs of platelets, and Studies with P450 7A1-knockout mice show
efforts at development preceded the characteriza- that this reaction (cholesterol 7a-hydroxylation) is
tion of the enzyme as a P450 (refs [1008-1010]). essential for proper absorption of dietary lipids
Many of these inhibitors have a basic nitrogen and fat-soluble vitamins in newborn mice, but not
atom that binds to the P450 5A1 heme^^i^. The for maintenance of cholesterol and lipid levels^^^^.
440 F. Peter Guengerich

The mice exhibit a complex phenotype with increased (rat) CYP7A1 expression in a reporter
abnormal lipid excretion, skin pathologies, and assay^^"^"*.
behavioral irregularities. The cholesterol levels In addition to the mouse CYP7A1 knockouts,
were not altered. Interestingly, vitamin D3 and E work has been done with overexpression in
levels were low to undetectable. j^j^gi045, 1046 j^Q j^j^,g ^ j ^ jjQt exhibit altered
A new era in the regulation of P450 7A1 began cholesterol levels ^^'^^. The lack of an LXR element
with reports of the involvement of some of the in a region (—56 to -49) of the human promoter
orphan steroid receptors. The proximal promoter may dictate some of the differences seen in mouse
region interacts with LXRa. The oxysterols 24(S)- and human models. With regard to humans, one
hydroxycholesterol and 24(»S)-epoxycholesterol study of biopsy samples from gallstone patients
activate LXRa (and LXRP)^^26 Further, mice led to the conclusion that there was no correlation
devoid of LXRa fail to induce CYP7A1 transcrip- between levels of total bile acids and P450 7A1
tJQj^i027 j^Q other proteins, FXR and CPF, are activity^^'^''. A correlation was seen with levels of
also involved^^^^"^^^^. Chenodeoxycholate, a bile chenodeoxycholic acid.
acid derived from cholesterol, interacts with A long-standing observation from rodent stud-
FXR to suppress CYP7A1 transcription^^^^ ies is the apparent circadian rhythm of P450 7A1
However, the action of FXR has been reported to (ref [1048]). This phenomenon has been sug-
be indirect^^^^ PXR binds lithocholic acid and gested to be indicative of a short halflife of the
downregulates CYP7A1 (ref [1032]). Thus, cho- enzyme^^^^' *^^^. The phenomenon has also been
lesterol metabolites control their synthesis in the reported in nonhuman primates^^^^. The circadian
liver through feedback suppression of CYP7A1 rhythm can be demonstrated at the level of actual
(ref [1028]). Hylemon^^^^ has concluded that the P450 7A1 in rats^^^^. The molecular mechanism
dominant factor is LXRa. CPF binds to the of the rhythm is still not clear. One aspect is the
promoter (as a monomer) and leads to CYP7A1 instability of P450 7A1 in microsomes {in vitro),
transcription^ °^^. with a /,/2 of ~ l - 2 h r in humans and rats^^^^.
Other studies have addressed the role of Alternatively, the mRNA has a short /,/2 and the
PPARa in P450 7A1 downregulation'^"^^. circadian rhythm can be seen at the mRNA
However, differences exist between humans and jgyg|i054 Another unresolved aspect of P450 7A1
mice gene responses have been observed, with the research is the issue of phosphorylation, postu-
mouse gene showing an enhanced response to lig- lated early in the field^^^^. In vitro experiments
ands because of an additional binding site'^^^ (fur- with microsomes show some effects of various
ther, humans have much less PPARa than treatments^^^^' '^^^. More recent work with micro-
rodents^o^^). Chiang^^^^ analyzed the PPARa somes and recombinant proteins also shows
response and provided evidence that the downreg- effects'^^^, although the in vivo significance is yet
ulation by PPARa-agonist complex is due to com- unclear.
petition with HNF-4 for the DR-1 sequence. Polymorphisms in the coding and noncoding
The regulation of P450 7A1 by other factors has regions of the CYP7A1 gene are known'^^^. Some
been considered. Downregulation by TNFa has have been associated with clinical changes'^^^, but
been interpreted in the context of MEKKl, an others have not'^^'.
upstream nitrogen-activated protein kinase, affect-
ing HNF-4 (ref [1038]). The same mechanism may
6.37.3. Substrates and Reactions
be involved in the repression by endotoxin and
interleukin-1 (ref [1039]). A novel CYP7A1 site The classic reaction of P450 7A1 is choles-
appears to be involved in the repression of CYP7A1 terol 7a-hydroxylation^^, and esterified choles-
by thyroid hormone (T3)^^'^^. Studies with rats indi- terol is not a substrate ^^^^. However, recent
cate differences in the regulation of P450 7A1 and experiments have established that the enzyme
P450 27A1, a sterol 27-hydroxylase^^4i Human also catalyzes the 7a-hydroxylation of
CYP7A1 expression is also repressed by insulin and 24-hydroxycholesterol, with preference for the
phorbol esters *^^^. Estrogen (100 juig/kg/week) (»S)-isomer^^^^. 7a-Hydroxylation (with recombi-
increased hepatic cholesterol 7a-hydroxylation 2.7- nant human P450 7A1) was observed with 20(5)-
fold in ovariectomized baboons ^^^^. Retinoic acid hydroxycholesterol, 25-hydroxycholesterol, and
Human P450s 441

27-hydroxycholesterol^^^^. The relevance of interpretation of results of family and experimen-


the activity toward 25(»S)-hydroxycholesterol is tal studies with P450 7A1.
unknown compared to P450 39 (ref. [1065]).

6.37.4. Knowledge about Active Site 6.38- P450 7B1


Relatively little has been done with site- Almost all of the work with P450 7B1 is from
directed mutagenesis or modeling. As indicated rodent models and application to the human
(vide supra), the enzyme only catalyzes 7a- CYP7BI gene system is by inference. A P450 7B1
hydroxylation but is not very sensitive to side- transcript was first characterized in a rat (brain)
chain hydroxyIs. hippocampus cDNA library^^^^. A heterologously
The region 214-227 has been postulated to expressed protein was shown to catalyze the 7a-
interact with the membrane and to serve as a hydroxylation of the steroids DHEA and preg-
substrate-access channel ^^^^. Mutations in this nenolone ^^^^. Expression has also been reported in
region yielded some changes in kinetic parameters liver and kidney^^^^' ^^'^^. Disruption of the mouse
toward cholesterol. CYP7B1 gene yielded animals that were viable
and apparently normal, but ex vivo 7a-hydroxyla-
tion of DHEA and 25-hydroxycholesterol was
6.37.5. Inhibitors blocked in brain, spleen, thymus, heart, lung,
Limited information about inhibitors is avail- prostate, uterus, and mammary gland^^^^.
able. As indicated earlier, 7-oxocholesterol is a (n) Although extrapolation to humans has not
(competitive) inhibitor ^^^^. been reported, P450 7B1 is considered to be a
neurosteroid hydroxylase and have a potentially
important role^^^^' ^^^^. Functional polymorphisms
6.37.6. Clinical Issues in the human CYP7B1 gene have not been
reported, but have been postulated to lead to
P450 7A1 has been a topic of considerable inter-
severe hypercholesterolemia and neonatal liver
est in the areas of hepatology and gastroenterology.
disease^^.
The hypersecretion of cholesterol in obesity
does not appear to be due to reduced 7a-hydroxy-
lation^^^^. Coffee terpenes (e.g., cafestol) inhibit
P450 7A1 and also raise cholesterol levels^^^^, 6.39. P450 8A1
although it is not clear that the two phenomena are
linked. The complex regulation of P450 7A1 Prostacyclin (prostaglandin I2) has strong
makes interpretation of some experiments diffi- vasodilation and anti-aggregation effects on
cult. Overexpression of P450 7A1 in HepG2 platelets, and the imbalance of prostacyclin and
cells increased bile acid synthesis but led to thromboxane A2 (product of P450 5A1) is a factor
decreased hydroxymethylglutarate (HMG) CoA in several diseases, for example, myocardial
reductase activity (rate-limiting step in cholesterol infarction, stroke, atherosclerosis^^^^' ^^^^. The
biosynthesis) ^^^^. reaction yielding prostacyclin from prostaglandin
Alterations in P450 7A1 were not seen in H2 is another "internal" oxygen transfer, without
hypo- or h5q)erthyroidism^^^^. the input of O2 and electrons from NADPH
A 10-week old child with a stop-codon muta- (Figure 10.12), and the involvement of a P450 was
tion and lacking P450 7A1 presented with severe not immediately obvious. Ullrich hypothesized
cholestasis, cirrhosis, and liver synthetic fail- P450 involvement on the basis of spectral interac-
^j.gi060 ^ frameshift leading to (homozygous) tion studies^o^l DeWitt and Smith^^^^ used a
lack of P450 7A1 was associated with high low- monoclonal antibody to purify catalytically active
density lipoprotein (LDL) cholesterol, but not prostacyclin synthase from bovine aorta and
total cholesterol^^^^. Heterozygotes were also demonstrated a P450 Fe^^'CO spectrum.
hyperlipidemic. However, Beigneux et al}^^'^ have Subsequently, P450 8A1 was cloned from bovine
discussed some of the caveats associated with endothelial cells^^^^.
442 F. Peter Guengerich

6.39.1. Sites of Expression and the corresponding prostacyclins ^^^^. Spectral bind-
Abundance ing studies with 9,11-epoxymethano prostaglandins
F2 and F2^ lead to the view that the binding juxta-
Human P450 8A1 was cloned from aorta position is the key determinant in distinguishing the
endothelial cells by Tanabe's group ^^^^. The courses of catalysis by P450s 5A1 and 8A1 (ref
mRNA is widely expressed in human tissues, [1007]). A mechanism consistent with available
including ovary, heart, skeletal muscle, lung, data has been proposed (Figure 10.12)^^"*' ^^^^.
prostate^^''^, and umbilical vein^^^^ More recent
work has shown some localization in the brain,
including neurons^^^^' ^^^^. Another site of expres- 6.39.4. Knowledge about Active Site
sion is fallopian tubes, with expression in luminal
epithelia, tubal smooth muscle, vascular endothe- Mutagenesis of Cys441 (heme binding Cys) or
lial cells, and vascular smooth muscle cells^^^"^. Glu347 or Arg350 (EXXR motif) abolished cat-
alytic activity, suggesting that the placement of
these residues is correct'^^^. Other site-directed
6.39.2. Regulation and Polymorphism mutagenesis studies suggest roles of Ile67, Val76,
Leu384, Pro355, Glu360, and Asp364, which have
P450 8A1 is constitutively expressed in human been suggested in models'°^^. However, the level
endothelial cells^^^^. The human CYP8A1 gene of residual activity was 5-10% and only a single
(chromosome 20) has 10 exons^^^^"^^^^ and has substrate concentration was used; another caveat
consensus sequences for Spl, AP-2, an interferon-7 is that the expression work was done in COS cells
response element, GATA NF^B, a CACCC box, and the level of expression of holoprotein was not
glucocorticoid receptor, and a shear stress respon- measured.
sive element (GAGACC)!^^^ Whether or not all Other work has been on membrane topology,
of these are functional and how they interact to and antibody studies indicate that P450 8A1 is
maintain constitutive expression is not well under- mainly exposed on the cytoplasmic site of the
stood yet. endoplasmic reticulum with a single transmem-
Polymorphisms have been of interest because brane anchor^ ^^^' •^^^. The (unstable) substrate,
of disease relevance. The coding sequence con- prostaglandin H2, is produced in the lumen and
tains at least five alleles^^^^. In the 5'-region, these apparently passes through the membrane to reach
are polymorphisms involving a variable number of P450 8A1. Antibodies raised to the peptides of the
tandem repeats (VNTR) that affect transcription, putative substrate channel (66-75 and 95-116)
as demonstrated in reporter systems in vitro^^^^. At interact only after membrane solubilization,
least nine of these allelic variants are known'^^^. implying that the substrate-access channel is very
An association between this VNTR polymorphism near the membrane'^^^.
and cerebral infarction has been reported •^^^.
A SNP in exon 8 has been reported to be linked
to myocardial infarction, although no amino acid 6.39.5. Inhibitors
change occurs'^^^ However, the VNTR polymor-
phism does not appear to be related to essential Relatively little interest has been shown in the
hypertension'^^^, nor does the 5'-flanking region development of drugs that inhibit P450 8A1
SNP T192G (ref [1093]). However, a novel splic- because inhibition is generally considered to be
ing variation leading to skipping of exon 9 has deleterious.
inhibit^^oo
Phenylbutazone has been reported to
been linked to hypertension'^^'^.
P450 8A1 is slowly inactivated during the
normal reaction itself, apparently by one of the
reactive intermediates in the catalytic cycle
6.39.3. Substrates and Reactions
(Figure lO.ny^'K A )^,,^,,,^,^„ of 0.06 s'^ was
P450 8A1 has a very limited catalytic speci- reported^^^^
ficity, functioning only as the prostacyclin synthase Peroxynitrite is a powerful inhibitor of P450
(Figure 10.12). Prostaglandins G2, H2, 13(5)- 8A1, with a reported K. of 50 nM^^^^
hydroxy H2, 15-keto H2, and H3 are isomerized to Peroxynitrite is formed by the chemical reaction
Human P450s 443

of NO* and O2' (ref. [1103]). The mechanism is Regulation of the gene is of interest, in that
believed to involve tyrosine nitration^ ^^"^j and P450 8B1 catalyzes the synthesis of cholic
recently Tyr430 has been implicated as the site of acid and controls the ratio of cholic acid to
nitration^ *^^. chenodeoxycholic acid in the bile^^^^. HNF-4a
activates human CYP8B1 expression in HepG2
cells^^^^. Bile acids and farnesoid X receptor
6.39.6. Clinical Issues (FXR) downregulate HNFa expression. Inflam-
As mentioned earlier, prostacyclin is a power- mation in liver cells causes increased synthesis of
ful vasodilator and inhibits platelet adhesion and a J-antitrypsin, a serum protease inhibitor, and in a
undesired cell growth. Although this view may be derived peptide (C-36). C-36 appears to interact
overly simplistic, prostacyclins are a counterbal- with the a J-fetoprotein transcription factor (FTF)
ance to thromboxanes in a "yin-yang" relation- site in the human CYP8B1 promoter, inducing a
ship. Thus, the action of P450 8A1 balances that conformational change to lower DNA binding
ofP450 5Al. ability, and suppressing the transcription of the
Decreased expression of P450 8A1 has been CYP8B1 (and CYP7A1) genes^^^^' ^^^^. HNFa
reported in severe pulmonary hypertension^ ^^^. could overcome the inhibitory effects of FTF and
With regard to general cardiovascular disease, a bile acids^i^^ Thus, regulation of P450 8B1 is
study of Japanese subjects associated the VNTR involved in bile acid feedback inhibition.
polymorphism with hypertension (odds ratio
1.9)^^^^. Individuals with 3 - ^ repeats had less
promoter activity and higher risk. In experimen-
tal studies, the overexpression of P450 8A1 in 6.41. P450 11A1
transgenic mice protected against the develop-
P450 11 Al is the enzyme involved in the initi-
ment of hypoxic pulmonary hypertension^ ^^^. In
ation of steroid synthesis (Figures 10.13 and
another study, the expression of human P450
10.14). It catalyzes the conversion of cholesterol
8A1 in the carotid arteries of rats after arterial
to pregnenolone by side-chain cleavage and has
balloon injury (using a virus) led to increased
been referred to in the older literature as P450^^^
synthesis of prostacyclin and to reduced neointi-
or cholesterol desmolase. The enzyme was puri-
mal formation^ ^^^.
fied from bovine adrenal cortex mitochondria^ ^^^.
P450 8A1 also has relevance in cancer treat- The human gene was cloned by Omura and Fujii-
ment. Transfection of colon adenocarcinoma cells Kuriyama in 1987^^^^ and includes nine exons. Of
with P450 8A1 led to slower growth and reduced historical significance is the fact that this P450
vascular development following inoculation into only contains a single cysteine and further estab-
syngeneic mice^^^^. lishes the position of the heme thiolate peptide
Finally, antibodies in the sera of some patients in P450s, extending the work on the location
with hypersensitivity reactions to phenytoin and from the crystal structure of bacterial P450 101
carbamazepine recognize rat P450 3A1 but not (ref [1118]).
human P450 3A (ref [1111]). The antisera also
recognize peptides derived from P450s 8A1 and
5A1, although relationships of etiology and
causality are unclear. 6.41.1. Sites of Expression
P450 11 Al is found primarily in steroidogenic
6.40. P450 8B1 tissues, that is, adrenal cortex and gonads, includ-
ing ovary (corpus luteum^^^^' ^^^^ and theca
P450 8B1 is a sterol 12a-hydroxylase interna cells^^^^ and others^^^^). Of interest are
expressed in the liver. The human CYP8B1 gene recent reports of P450 l l A l in brain^^^^-^^^^ and
was characterized on the basis of the rabbit and pancreas^ ^^^.
mouse orthologs^^^^. Of interest is the finding that P450 l l A l is one of the few P450s localized
this gene is devoid of introns, unique for this gene in the mitochondria (Table 10.1 and Figure 10.14).
among the P450 family^ ^^^. Studies with the bovine enzyme demonstrated that
444 F. Peter Guengerich

P450 21A2 ) (P450 11B1


Progesterone ^ Deoxycorticosterone • Corticosterone

(M5017A1^ ,P45017A1^
I > 4 5 0 21A2;
17-OH Pregnenolone 17-OH Progesterone "—^ 11-Deoxycortisol
18-OH Corticosterone

Cortisol Aldosterone

Testosterone

P45019AO

Estradiol

Figure 10.13. A view of the metabolic pathway of steroidogenesis and the major P450s involved^^.

P45011A1 P45017A1 P45017A1


Cholesterol • Pregnenolone 17-OH Dehydroepi-
Pregnenolone androsterone
3li-0H 3/3-OH 3/3-OH
Steroid D.H. Steroid D.H. Steroid D.H.
P45017A1 P450 17A1
Progesterone • 17-OH T >- Androstenedione
I Pregesterone
P450 21A2 P450 21A2 P45019A1

P450 P450
11B2 11B1 Estrone
18-OH Cortico^^^ Deoxy- 11-Deoxy-
Corticosterone sterone"^ corticosterone cortisol
P450
11B2
P450
11B1
Aldosterone Cortisol

Mitochondrion Endoplasmic reticulum

Figure 10.14. Overview of steroidogenic pathway and cellular compartmentalization^^.

P450 l l A l , synthesized on ribosomes in the efficient mitochondrial import^ ^^^. Miller's group
cytosol, is imported into mitochondria without constructed vectors that could be used to direct
processing of the amino terminal extension pep- P450 l l A l to the endoplasmic reticulum and
tide^'^^. The protein moves to the mitochondrial found that the enzyme was inactive' '^^. The mem-
inner membrane and is then cleaved to yield the brane environment was concluded to be more
mature form^^^^. Alteration of the basic amino important in modulating catalytic function than
acid residues of the A^-terminus resulted in less the electron transfer partners.
Human P450s 445

6.41.2. Regulation and Polymorphism intermediate^ ^^^. Oxidative cleavage of the


diol to pregnenolone and 4-methylpentanal
The regulation of P450 1 lAl is relatively com-
(isocaproic aldehyde) completes the overall
plex, as might be expected for the initial step in
reaction. The mechanism of the last step is not
steroid formation^ ^^^. Moreover, the system must
completely clear, but some proposals have been
be able to respond to signals in many different tis-
presented^ ^'*^~^^^^.
sues. Much of our understanding of the regulation
The rate of electron transfer from adrenodoxin
of P450 1 lAl expression is based on studies with
is important and appears to be the rate-limiting
CYPllAl genes of experimental animals and rein-
step for the enzyme in human placenta^ ^'^^. The
vestigated with human CYPllAl.
redox potential of adrenodoxin can be varied by
P450 l l A l has long been known to be regu-
site-directed mutagenesis, but had little effect on
lated by ACTH and cyclic AMR In the bovine
rates of electron transfer, consistent with the view
CYPllAl gene two Spl-binding sites mediate
that other factors such as protein-protein inter-
cyclic AMP transcription through the protein
actions are more important than the intrinsic ther-
kinase A signaling pathway, utilizing the rather
modynamics ^^ 47 y^YiQii P450s 1 lAl and 1 IBl are
ubiquitous transcription factor Spl (ref [1131]).
expressed together in cells, they can compete for
The steroidogenic factor 1 (SFl) activates
reducing equivalents from adrenodoxin^ ^'^^;
CYPllAl transcription through interaction with
exactly how important the competition is in tis-
protein factors upstream^ ^^^. An upstream CREB-
sues is unclear. Another report indicates interac-
binding region and an AP-1 site are also involved
tion of P450 l l A l with and enhancement by b^
in the cyclic AMP response. Sp3 can also be
(ref [1149]) although the relevance is unclear
involved^ ^^^. The TATA box drives cell type-
because of the compartmental separation of P450
specific cyclic AMP-dependent transcription^ ^^^.
11 Al (mitochondria) and b^ (endoplasmic reticu-
SF-1 also interacts with Spl (refs [1134-1136]).
lum) (Figure 10.14).
Thus, the regulation of the human CYPllAl gene
involves all the above factors plus an AdE ele-
j^gjj^ii22 More recently, the expression of the
human gene has been shown to involve the zinc 6.41.4. Knowledge about
finger protein TreP-132, interacting with both Active Site
CBP/p300 (ref [1137]) and SF-1 (ref [1138]).
Also, salt-inducible kinase (SIK) represses cyclic Knowledge about this enzyme is still relatively
AMP-dependent protein kinase-mediated activa- limited. Studies with bovine P450 l l A l indicated
tion through the CREB basic leucine zipper the significance of Lys377 and Lys381 in adreno-
domain^ ^^^. doxin binding^ ^^^. As indicated earlier, a mutation
at Arg353 was found to attenuate the function
Other recent work with human placenta show
of P450 l l A l in a patient^^^i site-directed
that activating protein-2 (AP-2) assumes the role
mutagenesis of human P450 l l A l (in E. coli)
of SF-1 by binding to an overlapping promoter
indicated that Ile462 had some effect on kinetic
elementii40.
parameters^ ^^^
Mutations are also found in CYPllAl and can
cause congenital adrenal insufficiency. Arg353
was found to be critical in a study with an afflicted
patient^ ^41 6.41.5. Inhibitors
A number of inhibitors of P450 l l A l have
been reported, although some were studied only
with the bovine enzyme ^^^^' ^^^^, including some
6.41.3. Substrates and Reaction
acetylenic mechanism-based inactivators^^^"*.
P450 l l A l appears to be quite specific in With regard to the human enzyme, there is some
using cholesterol as a substrate. The reaction potential for use of inhibitors in treatment of pro-
proceeds in a three-step sequence, with genera- static cancer, and prodrug forms of amino-
tion of (22i?)-20a, 22-dihydroxycholesterol as an glutethimide have been examined^ ^^^.
446 F. Peter Guengerich

Anti-convulsants have been reported to inhibit the early research in this field was done with
P450 1 lAl, but the interaction is not strong^ ^^^. bovine adrenal glands because of the need for
large amounts of material, and the bovine
P450 IIB protein has the function that P450
6.41.6. Clinical Issues l l B l (11-hydroxylation) and the P450 11B2
(11-hydroxylation, 18-hydroxylation, and oxida-
Two major issues are of interest. Because of tion of the 18-alcohol to an aldehyde) have in
the nature of P450 1 lAl in initiating steroidogen- most other species, including humans^^^^. The
esis, deficient P450 11 Al can lead to (congenital) two human genes (CYPllBl, CYP11B2) were
adrenal hyperplasia^^. Rabbit and mouse models characterized and clearly shown in both to be
show the effects^i^^' ^^^l C7P77^7-null mice die essential^ 16^-^ ^^^
shortly after birth, but can be rescued by steroid P450 1 IBl expression has also been deleted in
injection* ^^^. ACTH levels become very high due human fetal adrenal gland, particularly in the
to lack of feedback regulation by glucocorticoids. "fetal zone" (as opposed to neocortex)^^^^.
Male null mice are feminized with female external
genitalia and underdeveloped male accessory sex
organs. These manifestations resemble various 6.42.2. Regulation and Polymorphism
human steroid deficiency syndromes. Much of the background on regulation of P450
Another issue is autoantibodies to P450 11 Al l l B l came from studies with the bovine gene,
(and also P450 17A1) in patients with autoim- which responds to ACTH and has six c/^-acting
mune polyglandular syndrome types I and II, and regulatory elements^^^^. The protein (Ad4BP) that
Addison's disease**^^' **^°. As with other P450s binds to one of these (Ad4) is a member of the
recognized by autoantibodies, causal relationships steroid hormone receptor superfamily^^^^. Other
between immunity and disease are unclear. studies by Omura^^^^ indicated the cooperative
nature of these elements in transcription. Work
with rat CYPllBl showed that ACTH stimulates
transcription by changing composition in AP-1
6.42. P450 11B1 factors (Fos, Jun)*'^^
P450s l l B l and 11B2 differ in only 32 The human gene also has a cyclic AMP
residues. P450 1 IBl catalyzes the 11 p-hydroxyla- response element (CRE)"^^. The Adl element
tion of deoxycortisol to yield Cortisol, which is the bound CRE-binding protein, activating transcrip-
main glucocorticoid in the body. Deficiencies in tion factor-1 (ATF-1), and ATF-2. Steroidogenic
the enzyme are known, causing congenital adrenal factor-1 (SF-1) interacted at the Ad4 site
hyperplasia^^' **^*. (-2427-234) and was required for transcrip-
^JQj^ii72, 1173^ which contrasts with the lack of
response of C7P7752.
Many mutations are known because of the
6.42.1. Sites of Expression
relationship of the gene with congenital adrenal
P450 1 IBl is expressed in the adrenal cortex, hyperplasia''^^ These include a 5-base duplica-
specifically the zonafasciculate/reticularis^^^K In tion"'^'^ and clusters of mutations in exons 6-8
rats, some expression has been detected in brain (ref [1175]). The high similarity and proximity of
but the relevance is not clear. the CYPllBl and CYP11B2 genes appear to lead
P450 l l B l is synthesized in the cytosol and to mutant generated by unequal crossover and
directed to the mitochondria with a 24-residue A^- inactive chimeric product "^^""^^. Splice donor
terminal targeting sequence (where this is lost after site mutations are also known"^^.
entry). As with the other four (exclusively) mito-
chondrial P450s (Table 10.1 and Figure 10.14),
P450 l l B l receives electrons fi-om adrenodoxin
6.42.3. Substrates and Reactions
instead of NADPH-P450 reductase.
The characterization of the CYPllB gene has As indicated previously, the only substrate for
developed considerably in recent years. Much of P450 l l B l is deoxycortisol, which undergoes
Human P450s 447

11 P-hydroxylation to yield Cortisol (Figures 10.13 excess 11^^' ^^^^. Overproduction of glucocorti-
and 10.14). coids, which could have any of several causes
including overactive P450 l l B l , is associated
with Cushing's syndrome^i^i.
6.42.4. Knowledge about Active Site
One of the concerns about studies on the func-
tion of particular residues in site-directed mutagen- 6.43. P450 11B2
esis is that expressions in some cellular systems
lead to competition between P450s 1 lAl and 1 IBl P450 11B2 is highly related to P450 l l B l
for (adrenodoxin) reducing equivalents in cellular {vide supra) and has a somewhat similar function.
systems^ ^'^^. Another issue is that human P450s P450 11B2 catalyzes the 11 (B-hydroxylation of
l l B l and 11B2 have been difficult to express in 11-deoxycorticosterone followed by 18-hydroxy-
bacteria, so that most experiments have relied on lation and 2-electron oxidation of the 18-alcohol
to an aldehyde (Figures 10.13 and 10.14).
mammalian cells {Schizosaccharomyces pombe has
Changes in the gene can lead to corticosterone
provided some success^ ^^^). Nevertheless, much
methyloxidase deficiency and hyperaldostero-
information about function has been obtained from
nism29' 1165, 1192, 1193 i^ tjjg Qi^g^ literature, this
patients' samples ^ ^ ^ I
The close similarity of P450s l l B l and 11B2 P450 is sometimes termed as "P450 , ,^."
(and their reactions) has also facilitated studies.
Making the changes S288G and V320A yielded
an enzyme with both P450 1 IBl and 11B2 activ- 6.43.1. Sites of Expression
ities^^^^. Changes at positions 147 (refs [1182],
[1183]), and 301/355 (ref [1184]) have also had P450 11B2 is expressed in the adrenal cortex
the same effect. (zona glomerulosa) and is involved in the synthe-
sis of aldosterone (the lip-hydroxy, 19-aldehyde
Homology models of P450 l l B l have also
product). It is a mitochondrial P450, as are the
been published^^^^' ^^^^^ ^^^^, although the effects
other 11 family P450s. The cDNA was first cloned
of all of the mutants known to alter function have
from an adrenal tumor of a patient suffering from
not been systematically rationalized.
primary aldosteronism^ ^^'*. Another early study
showed higher levels of P450 11B2 in aldos-
6.42.5. Inhibitors terone-secreting tumors^ ^^^.
There is some evidence for the synthesis
Compared with some of the other steroido- of aldosterone outside of the adrenals, and Li
genic P450s, there is some reason to develop P450 et al}^^^ reported expression of P450 11B2 in
1 IBl inhibitors. High levels of Cortisol are associ- hepatic stellate cells of liver; the activation of
ated with Cushing's syndrome^ ^^^ Cellular these cells is a key event in liver fibrogenesis.
expression systems have been set up to assay for
inhibitors, using measurements of concentrations
o f s t e r o i d s ! 187, 1188

18-Vinylprogesterone and 18-ethynylproges-


6.43.2. Regulation and
terone have been reported to be mechanism-based Polymorphism
inactivators of bovine P450 IIB, but apparently Some of the research on regulation overlaps
have not been tested with human P450 1 IBl (ref that presented for CYPllBl {vide supra). A
[1189]). CRE/Adl element and ATF-1 (and ATF-2?) play a
role with both CYPllBl and CYP11B2 (ref.
[1197]). However, SF-1 does not appear to regulate
6.42.6. Clinical Issues
CYP11B2, in contrast with CYPllBl (ref [1173]).
As indicated previously, the main issue with Many aspects of regulation remain to be further
P450 l l B l is the impaired synthesis of Corti- investigated, including the mechanisms of the
sol and congenital adrenal h3q)erplasia, character- observed Ca^^ and cyclic AMP signalingn^^ and
ized by hypertension and signs of androgen the effects of kinase inhibitors^ ^^^' ^^^^.
448 F. Peter Guengerich

As in the case of CYPllBl, many mutations (ref [1208]), although that paper reported that
have now been defined from clinical studies. The the enzyme used catalyzed l i p - , 18-, and 19-
"crossovers" between P450s 1 IBl and 11B2 yield hvdroxvlatinn
hydroxylation
inactive P450 11B2, as well as P450 l l B l (refs
[1178], [1179], [1201], [1202]). Other mutations
in CYP11B2 were associated with corticosterone 6.43.6. Clinical Issues
methyloxidase I and II deficiency^^^^' ^^^^' ^^^^.
Polymorphisms in CYP11B2 have also been The issues of congenital adrenal hyperplasia
linked to idiopathic hyperaldosteronism, a condi- and Types I and II corticosterone methyloxidase
tion characterized by autonomous production of deficiency in individuals with attenuated P450
aldosterone and arterial hypertension^^^'^. A poly- 11B2 activity have already been mentioned. The
morphism in the promoter region of CYP11B2 other issue also mentioned is elevated aldosterone.
(—344 TK) has been associated with predisposi- Several studies have reported an association
tion to essential hypertension^^^^. between polymorphisms and essential hyperten-
sion, although the measurements of aldosterone
excretion are still lacking in some studies ^^^^.
Other studies show association of the -344C allele
6.43.3. Substrates and Reactions
with increased left ventricular size^^^^"^^^^. The
P450 11B2 catalyzes the three-step conversion hypertension association has been seen in several
of 11-deoxycorticosterone to aldosterone, with 11 (3- studies^^^^' *2'^' ^2^^' ^2^^, but not in a Japanese
hydroxylation, 18-hydroxylation, and 2-electron study^^^^.
oxidation of the 18-carbinol (Figures 10.13 and
10.14). No other substrates are known. Information
about the processivity of the human enzyme 6.44. P450 17A1
(i.e., extent of release of intermediate products) is
not available at this time. 17-Hydroxylation and the 17, 20-lyase reac-
tion C'desmolase") are two important reactions in
steroid biosynthesis (Figures 10.13 and 10.14).
Cloning of a cDNA which, when expressed,
6.43.4. Knowledge about Active Site
yielded both activities that established the role of
Some studies with the closely related P450 1 IBl what is now known as human P450 17A1 (previ-
have already been mentioned. Bemhardt's labora- ously termed P450,7^, etc.)'2»^ The gene^^n
tory found that changes only at positions 320 and showed similarity to CYP21AL The demonstra-
335 conferred some 18-hydroxylation activity to tion of both catalytic activities in a single protein
P450 l l B l (ref [1184]). Homology modeling has established work previously done with purified
also been done''^^' ^^^^. In other site-directed muta- hog protein^^'^. The two activities have long been
genesis work, residues that differed among species known to be regulated by b^ (refs. [1219], [1220]),
were changed and residues 112, 147, and 152 and aspects of this duality of ftinction still remain
were found to have effects'^^^. Modeling suggested unclear.
an indirect effect of residue 147 and that residue
112 might be in the substrate-access channel.
6.44.1. Sites of Expression
Human P450 17A is expressed in steroidogenic
6.43.5. Inhibitors
tissues, including adrenals and gonads. The enzyme
Elevated aldosterone levels can be detrimental has also been reported in fetal kidney, thymus, and
and some interest exists in targeting P450 11B2. A spleen^^^^ The enzyme has also been found in
yeast system has been developed that can be used human (adult) heart^^^^ and adipose tissue^^^^.
for screening for inhibitors ^^^^. P450 17A1 is a microsomal enzyme. A proline
The literature contains one older report of the rich region in the A/-terminus has been found to be
use of an acetylenic P450 19 inhibitor to inhibit important for efficient folding, but not for subse-
the activity of what may have been P450 l l B l quent maintenance of the folded structure ^^^^.
Human P450s 449

6.44.2. Regulation and Polymorphism mechanism of the lyase reaction is not com-
pletely established, but mechanisms have been
As with the other steroidogenic P450s, the reg- proposed using analogs ^244 Lieberman^245 j^^g
ulation of the CYP17A1 gene is relatively com- proposed alternative reactions, although the
plex. Induction of P450 17A1 has long been favored pathway involves what would be a very
known to be cyclic AMP-mediated and the induc- unstable diradical. No other substrates are known
tion is suppressed by testosterone (mouse presently, other than pregnenolone and proges-
model)*^^^, and a cyclic AMP response region was terone and possibly closely related analogs.
mapped in porcine Leydig cells ^^^^. Very recently, Soucy et al}^^^ have provided
With the human CYP17AI gene, the homeo- evidence that human P450 17A1 also converts
domain protein Pbxl was shown to interact with pregnenolone into 5,16-androstadien-3p-ol, a
protein kinase A in the cyclic AMP-dependent "16-ene synthase" reaction (without intermediate
regulation (at -2507-241)^227 YurthQT analysis formation of an alcohol).
showed interaction at a cyclic AMP-related site A key to research on the protein was the devel-
(-80/-40) by SF-1 (ref [1228]). Further, inter- opment of a robust E. coli expression system by
actions were shown for Spl and Sp3 Waterman's group in 1991^^. Further work on the
(-227/-184) and NF-IC ( - 1 0 7 / - 8 5 and differential effects of b^ on individual catalytic
-1787-152)1229 sp_j (yide supra) also interacts activities has been reported^247 j ^ ^ J.^^JQ ^f ^^ ^^
with p54"*, NonO, and protein-associated splic-
P450 is high in testis and this phenomenon might
ing factori23o j^Q ACTH/cyclic AMP response is
regulate the two activities of P450 17A1. Miller's
dependent upon phosphatase activity, as well as group has proposed that phosphorylation of Ser
kinase activity^^si, 1232 jj^g cyclic AMP-depend- and Thr residues in P450 17A1 may alternatively
ent protein kinase enhances transcription via influence the two activities^248^ although any
MKP-1 activation, involving phosphorylation of experimental evidence in support of this hypothe-
SF-1 (ref [1233]). sis is still very limited^248,1249
Polymorphisms of CYP17A1 are known, but A second h^ gene has been identified recently,
most of the attention has been given to mutations and this protein also has the same stimulatory
that result in serious defects in patients ^^34 Most effect on lyase activity^250 Auchus et al.^^^ also
of the mutations have been SNPs in the coding demonstrated that the same stimulatory effect of
region 1234-1236^ ^^^ others include a 2-bp deletion b^ could be obtained with apo-Z?^, arguing against
yielding a frameshift and premature stop the requirement for electron transfer. P450 17A
codon^^^^^ a 4-bp duplication changing the C-ter- enzymes from other species vary in their ability to
minal 28 amino acids^^^^^ and a 5'-splice site catalyze the 17,20-lyase reaction, and compar-
mutationi239 Some of the patients presenting with isons of the rat and human enzymes also led to
symptoms yielded P450 17A1 that, upon heterol- the conclusion that selective enhancement of the
ogous expression, retained 17-hydroxylation but
lyase reaction was not due to changes in electron
not 17,20-lyase activity^^^o. 1241 Mutations of the transfer ^251
latter type led Auchusi242 to propose a model in
The concertedness of the P450 17A1 lyase
which neutralization of positive charges in the
reaction has been examined, and both the studies
redox partner binding surface of P450 17A may
both reached the conclusion that much of the 17a-
block the lyase activity but not 17-hydroxylation.
hydrox3^regnenolone dissociates ^252, 1253 jj^ ^j^^
of the studies^252^ ^^^ authors concluded that the
off-rate was an important factor in determining
the balance between 17-hydroxypregnenolone and
6.44.3. Substrates and Reactions
DHEA. Exactly how b^ would control this rate,
The generally accepted reactions of P450 which was modeled to be rather slow (2.6-
17A are the 17a-hydroxylation of pregnenolone 29min~^), is unclear, unless the effect is on the
to 17-hydroxypregnenolone and of progesterone protein conformation. However, a classic burst
to 17-hydroxyprogesterone. 17-Hydroxypregne- kinetic experiment was not done in the cited work
nolone is also oxidized to DHEA in the lyase and the h)q)othesis remains to be addressed in
reaction (Figures 10.13 and 10.14)^241,1243 J^IQ more detail.
450 F. Peter Guengerich

6.44.4. Knowledge about Active Site 6.44.6. Clinical Issues


Much of the information about the signifi- P450 17A1 has a central role in human
cance of active site residues comes from the steroid metabolism because of its role in regulat-
analysis of mutations in patients presenting with ing steroid flux (Figures 10.13 and 10.14).
diseases (see Section 6.44.2.). The changes Perturbations lead to problems in adrenarche,
H373L (ref [1254]) and P409R (ref [1255]) led to aging, and polycistric ovary syndrome ^^'^^' ^^^^.
a loss of heme incorporation. Mutation at Thr306, Some of the more serious mutations have been
possibly involved in protonation of Fe-00~ or mentioned already; another is a case of pseudo-
O-O cleavage, impaired 17a-hydroxylation more hermaphroditism due to lack of lyase activity^^^^.
than the lyase reaction^^^^. However, the change Some of the other possible disease conditions
R346A selectively abolished lyase activity^^^^, as or risks are being studied in relationship to less
did F417C (ref [1258]). Mutations at Lys83, serious polymorphisms. In most of these cases,
Arg347, Arg358, and Arg449 produced proteins the relationships are more difficult to establish in
that were refractory to b^ stimulation and attenu- the serious diseases. A possible link of CYP17A1
ated in lyase activityi259-i26i o f these, only R347H polymorphism has been made with rheumatoid
and R358Q have been found in patients'^^^. Some arthritis^ ^^^. Little influence of polymorphism was
mutants found in patients do cause the loss of seen on age of menarche^^^^. However, a link was
both 17-hydroxylation and the lyase reaction, made between a particular polymorphism and the
howeveri263, i264^ prediction to use hormone replacement therapy
Some animal P450 17A1 enzymes have (i.e., postmenopausal estrogen therapy)^^^^. No
different ratios of 17-hydroxylation/lyase activity association was found with polycistronic ovarian
and efforts have been made to use these properties syndrome in a study with an SNP at the regulatory
to define more elements controlling the latter Spl sitei29o.
steps, although the results have been limited to Much attention has been given to the possibil-
datel265, 1266^ ity of a link between CYP17A1 allelic SNPs and
Several homology models of human P450 breast cancer risk^^^^ The epidemiology results
17A1 have been published and some of the muta- are mixed at best^^^^"^^^^ and a conclusion in
genesis results can be interpreted^ ^^^' 1267-1271 favor of a relationship cannot be made at this
time'296-1298^
Some positive epidemiology has been pre-
sented for a relationship with prostate cancer^^^^,
6.44.5. Inhibitors although probably not a strong one'^^^. Some pos-
Inhibitors of P450 17A1 have been studied for itive results have also been reported for endome-
some time. Interestingly, ketoconazole inhibits trial cancer and CYP17AI alleles ^ 3^'.
lyase activity but not 17-hydroxylation activ- As with some other P450s, circulating anti-
bodies to P450 17A are seen in some autoimmune
ity^ 7a-Thiospirolactone is a mechanism-
diseases, for example, autoimmune polyglandular
based inhibitor of (guinea pig) P450 17A1 (ref
syndrome and Addison's disease^ ^^^' ^^^^, but no
[1273]).
causal relationship has been demonstrated.
A number of steroidal inhibitors have been
studied, primarily with the goal of treating
cancers ^^^^~'^^^. The enantiomer of progesterone
(e«f-progesterone) is reported to be a competitive
inhibitor of P450 17A {K. = 0.2 |ULM)1279
6.45. P45019A1
Nonsteroidal inhibitors have also been P450 19A1 is the classic "aromatase," often
Studiedl280,1281_ known by that name in endocrinology. This
Molecular modeling (Section 6.44.4) has also enzyme oxidizes the androgens androstandione
been applied to searches for inhibitors ^^^^' ^^^^. and testosterone to the estrogens estrone and
Other approaches utilize P450 17A expressed in 17p-estradiol, respectively (Figure 10.15). This
E. coli to screen for P450 17A inhibition in process is very important in normal physiology
medium-to-high throughput systems^^^^' ^^^'^. and also a target for inhibition in some tumors.
Human P450s 451

Figure 10.15. Aromatization reactions catalyzed by P450 19. The three distinct steps are shown, with the possible
substrates: Testosterone, Rj: -OH, R2: H; androstenedione, Rj.- ^ O , R2: -OH; 16-hydroxytestosterone,
R,—R~ -OH.

6.45.1. Sites of Expression In adipose tissue, the promoter from exon 1.4 is
utilized^^^^. The same exon is utilized in bone and
Estrogens have a number of functions and not skin^^^^, and in leiomyoma tissue derived from
only feminization. Sites of (human) expression myometrium^^^^. This system is regulated with
include the ovaries, testes, placenta, fetal (but not Spl, a glucocorticoid regulatory element, STAT3,
adult) liver, adipose tissue, chondrocytes and and possibly PPAR7^^^^' ^^^^. Pre-adipocytes also
osteoblasts of bone, vasculature smooth muscle, involve regulation with liver receptor homolog-1
and several sites in brain, including parts of the (LRH-l)i309.
hypothalamus, limbic system, and cerebral cor- In placenta exon I.l, an 89 kb upstream ele-
I^g^i303 ^g discussed later, regulatory mechanisms ment is utilized^^^^. This is a strong promoter and
differ considerably in these tissues. P450 19A1 is involves C/EBP-p^^^^. A strong positive enhancer
also expressed in some tumors, particularly those element between —42 and - 5 0 1 is present^^^^.
derived from these tissues. The possibility exists that vitamin D receptor/
The actions of androgens and estrogens in the RXRa heterodimers and PPAR7 may have
gonadal tissues are fairly well understood, but less eflFectsi303.
is known in the brain. Androgens and androgen- Regulation in bone uses exon 1.6 (ref. [1303]).
derived estrogens regulate complementary and The study of regulation in bone is less extensive
interacting genes in many neural networks^^^"^. than in other sites, and 1,25-dihydroxycholecalcif-
erol, interleukins, TNFa, and TGF-p^ have had
stimulatory activity.
6.45.2. Regulation and Polymorphism
Regulation in brain uses exon I.f and has also
The regulation of the CYP19A1 gene is quite not been as extensively studied^^^^. P450 19A1
complex, primarily because of the use of four does seem to be upregulated by androgens.
tissue-selective promoters^^^^' ^^^^. Either the LI, Regulation in fetal liver involves exon 1.4, as
1.4, I.f, or 1.6 sequence is read as exon I and with adipose tissue ^^^^. The same pattern appears
spliced into the mRNA, depending upon the tis- to apply in skin fibroblasts and intestine.
sue. However, exon I does not code for the protein, In cancer cells, alternate regulatory pathways
so the P450 19A1 enzyme is always the same. are utilized^ ^^^.
In preovulatory follicles and corpora lutea of A number of polymorphisms have been
human ovary, the 5'-untranslated region of P450 reported in the CYP19A1 gene. These have been
19A1 transcripts is encoded by exon Ila (ref studied in relationship to breast but without
[1306]). The major operatives here are CRE and convincing relationships {vide infra)\ also, there
SF-1 elementsi303_ was no relationship with breast density^^^^
452 F. Peter Guengerich

The number of cases of serious P450 19A1 Another report suggests generation of
deficiency is apparently only about ten, including inhibitory Michael agents from prostaglandin J2,
two adult males^^^^. but detailed characterization has not been
done^326
Other nonsteroidal inhibitors of P450 19A1 are
6.45.3. Substrates and Reactions also under consideration^^^^.
The reaction involves three steps and has been Breast cancer is the major target area for P450
the subject of considerable mechanistic interest 19A1 inhibition, but other cancers are also under
(Figure 10.15). Androstanedione is converted to investigation^ ^^^.
estrone, testosterone to 17p-estradiol, and 16- The point has been made by Simpson et alP^^
hydroxy DHEA to estriol. The first two steps are that a future goal of P450 19A1 inhibition should
relatively straightforward, for example, RCH3 -> be tissue selectivity. The diverse role of P450
R C H 2 0 H ^ C H 0 (at CI9). The third step was 19A1 in different tissues might indicate that gen-
difficult to rationalize with "classic" FeO^^ chem- eralized inhibition of estrogen synthesis may be
istry, and there has been general acceptance of a less than desirable. Targeted inhibition of P450
FeOO~-based mechanism originally developed by 19A1 could, in principle, be achieved by (a) selec-
Robinson^^^^ and Akhtar^^^^, and further devel- tive targeting of inhibitors of P450 19A1 catalysis
oped in models by Coon and Vaz^^^"^. to tumors/individual organs or (b) targeted down-
regulation of P450 19A1 synthesis in selected
The possibility of utilization of DHEA as a
areas.
substrate for estrone synthesis has been proposed
but not addressed directly^^^^.
6.45.6. Clinical Issues
6.45.4. Knowledge about Active Site Several clinical issues have already been
alluded to. The first issue is congenital aromatase
One of the historic problems in studying
deficiency. Serious cases in adults appear to be
structure-function relationships in P450 19A1
relatively rare'^^^' ^^^^ and have been treated with
has been the availability of expression systems.
estrogen replacement therapy'^^^. However, some
Recently two E. coli systems have been
children are considered to have attenuated P450
developed^^^^' ^3^^.
19A1 activity'3^'.
Site-directed mutagenesis has been done using
Studies with P450 19A1 knockout mice show
mammalian and insect cell-based systems, and
expected reproductive and sexual phenotypes and
models have been in existence for some time'^^^.
also adipose and bone phenotypes'^^^' '^^^, as well
More recent modeling work'^^^ has been done, with
as a sociosexual behavior phenotype'^^^.
an emphasis on docking of inhibitors in SRS-1.
The issue of using P450 19A1 inhibitors in the
treatment of a variety of estrogen-dependent tumors
6.45.5. Inhibitors has already been presented. In addition, there is
consideration of the use of inhibitors for breast
P450 19A1 inhibitors have long been of interest cancer prevention in high-risk individuals'^^'^.
for treatment of estrogen-dependent cancers in a A number of studies have been made on the
variety of tissues^^' ^^^^. Today, the process has relationship of CYP19A1 polymorphisms with
reached the stage of "3rd-generation" inhibitors^^^^ breast cancer, but the evidence has not shown a
moving beyond early drugs such as amino- change in risk'^''' '^^^. No strong association was
glutethimide^^^^. The newer inhibitors are more seen for endometriosis either'^^^.
effective in lowering the body limit of estrogens ^^^^.
One example of a newer drug is exemestane, a
mechanism-based inactivator currently in Phase III
6.46- P450 20A1
clinical trials and being compared with the estrogen
receptor antagonist tamoxifen^^2^~^^^^. The inactiva- At the time of writing this chapter, the only
tion appears to involve generation of a Michael available information was the existence of the
acceptor in the active site^^^^. CYP20 gene in the human genome^^^. The gene
Human P450s 453

appeared to be vertebrate specific and had been hyperplasia, and many mutations are now
speculated to be involved in development. known to be associated with the disease. Many are
the result of recombination with the related
pseudogene^^"^^' ^^^^. Some are in the coding
6.47. P450 21A2 ' and the 5'-flanking region^^'*^. The
region^
P450 21A2 is the enzyme involved in the 21- incidence of carriers of congenital adrenal hyper-
hydroxylation of progesterone and 17-hydroxy- plasia is 1-2% in the population, and many delete-
progesterone, yielding deoxycorticosterone and rious mutations have now been identified^^^^"^^^•^.
11-deoxycortisol from the two substrates, respec-
tively (Figures 10.13 and 10.14). The 21-hydroxy-
6.47.3. Substrates and Reactions
lation reaction is an important step in the synthesis
of glucocorticoids and mineralcorticoids, and The only known substrates are progesterone
deficiencies lead to "salt-wasting syndrome," if and 17-hydroxyprogesterone, which are hydroxy-
not treated, and to congenital adrenal hyperplasia lated only at the 21-position (Figures 10.13 and
in the worst cases. 10.14).

6.47.1. Sites of Expression 6.47.4. Knowledge about Active Site


The major site of expression is the adrenal cor- Homology models have been reported^^^^' ^^^^.
tex. This reaction has been known for some time, The amount of site-directed mutagenesis has been
and many of the early biochemical studies were limited, but the disease has yielded many locations
done with bovine adrenals because of the need for for loss of function because the severity of the dis-
large amounts of tissue ^^^^. ease is (inversely) correlated to the residual 21-
Low amounts of P450 21A2 have been hydroxylation activity. Many of the mutants could
reported in human lymphocytes^^^^ and brain^^^^. be rationalized in the context of a homology
Any specific function in these tissues is unknown model^^^^, although some associated with disease
at this time. are more subtle (e.g., E380D).

6.47.2. Regulation and Polymorphism 6.47.5. Inhibitors


The regulation of P450 21A2 has some similar- Relatively little has been published about
ity to that of P450 17A1, in that both are regulated inhibitors. Detrimental effects of spironolactone
by ACTH. The cyclic AMP responsive sequence in have been attributed to inhibition of 21-hydroxy-
the 5'-flanking region^^"^^ uses adrenal-specific lation^^^^, although further details with this P450
protein factor and an Ad4-like sequence ^^"^^ are lacking. Recently, Auchus^^^^ reported that the
One issue in the regulation of the CYP21A2 gene is enantiomeric form of progesterone (ew^proges-
the neighboring homologous but nonfunctional terone) is a competitive inhibitor of P450 21A2
CYP21A1 pseudogene, which can compete for (although not as effective as with P450 17).
transcription factors and other regulatory pro-
teins ^^^^. In other work, protein kinases A and C
6.47.6. Clinical Issues
and Ca^^ were found to regulate CYP21A2 gene
expression in a human cortical cell line^^^^. As mentioned earlier, the incidence of defects
Another interesting aspect of the regulation of is relatively frequent and the ability to form Corti-
the CYP21A2 gene is that it is located very close sol is a problem. At least 56 different mutations
to the major histocompatibility locus, 2.3 kb have been identified^ ^^^ Patients who cannot syn-
downstream from the C4 gene. Transcriptional thesize sufficient aldosterone may lose sodium
regulatory elements for the CYP21A2 gene lie balance and can develop a fatal "salt-wasting"
within intron 35 kb of the C4 gene^^"^"^. syndrome. Treatment involves administration of
Steroid 21-hydroxylase deficiency is the mineralocorticoids and glucocorticoids. Females
most common cause of congenital adrenal with severe, classic P450 21A2 deficiency are
454 F. Peter Guengerich

exposed to excess androgens prenatally and bom 6.48.2. Regulation and Polymorphism
with virilized external genitalia, but prenatal diag-
nosis permits prenatal treatment of affected The regulation of the CYP24A1 gene appears
females^^^^. Experimental research is being done to be complex, although some phenomena
on gene therapy to transfer active CYP21A2 observed in animal models have not been exam-
genes; work done on mice suggests feasibility^^^^. ined in as much detail in humans. The activity has
long been known to be inducible by vitamin D,
perhaps to relieve the cells of an overload, and
a vitamin D receptor element has been found
6.48. P450 24A1 in the 5'-region of the CYP24A1 gene^^^^' ^^^^
Parathyroid hormone and cyclic AMP both
The next three P450s (24A1, 27A1, 27B1) are enhance induction by the vitamin D receptor^^^^.
involved in vitamin D metabolism (Figure 10.16). In human keratinocytes, P450 24A1 mRNA
All three are mitochrondrial and receive electrons was also elevated by la,25-dihydroxyvitamin D3
from the iron sulfur protein adrenodoxin (via the (ref [1367]). Studies with rat systems indicate that
flavoprotein adrenodoxin reductase) (Table 10.1). this response is also mediated by vitamin D
response elements and that two of these (VDRE-1,
VDRE-2) operate synergistically^^^^. A functional
6.48.1. Sites of Expression and Ras-dependent Ets-binding site is located down-
stream from the proximal VDRE site and was
Abundance
critical; the model indicates transcriptional cooper-
The 24-hydroxylation of 25-hydroxyvitamin ation between Ras-activated Ets proteins and the
D3 has long been known to occur in the kidney vitamin D receptor-RXR complex in mediating
mitochondrial membrane ^^^^ Following the la,25-dihydrox)rvitamin D action on the P450
purification of a rat P450 with this activity^^^2, 24A1 promoter'^^^. The YYl transcription factor
cDNA clones for chicken^^^^ and human^^^"^ has been reported to repress la,25-dihydroxy-
homologs were obtained. vitamin D3-induced transcription in cell cul-
The enzyme is expressed in both proximal and ^gi375 jYiQ isoflavone genistein was reported to
distal kidney tubules^^^^, but has also been found block the transcription of the CYP24A1 gene in
in human nonsmall cell lung carcinomas ^^^^. This human prostatic cancer cells and this block could
would appear to be a relatively low abundance be relieved with the histone deacetylase inhibitor
P450. Expression has also been reported in human trichostatin A*^''^. Finally, the earlier results with
keratinocytes^^^^' ^^^^, colon carcinoma cells^^^^, Ets proteins {vide supra) have been expanded
and prostatic cancer cells'^^^. to show distinct roles of the MAP kinases

,P45027A1 J ( P450 24A1


Vitamin D3 - ^ 25-OH D3 ^^ ^> 24,25-(0H)2 D3
(chloecalciferol)

P450 24A1^
1a,25-(OH)2D3 ^'^^ ^ 1 a,24,25-(OH)3 D3

P450 3A4 regulation - • Vitamin D receptor • Bone Ca^"^ mobilization,


intestinal Ca^"*" absorption

Figure 10.16. Overview of P450s involved in key steps of vitamin D activation^^.


Human P450s 455

HO^'

25(OH)

Figure 10.17. C-23 hydroxylation pathway for 25-hydroxyvitamin D3 (25(OH)) oxidation catalyzed by P450
24A1 (ref. [1378]).

ERK1/ERK2 and ERK5 (ref. [1376]). Induction 6.48.5. Inhibitors


of P450 24A1 by la,25-dihydroxyvitamin D3
involves Ets-1 phosphorylation at Thr38, but Inhibition of P450 24A1 is of some interest in
la,25-dihydrox3rvitamin D3 stimulation of ERKl/ the context of raising levels of active vitamin D
ERK2 required RXRa phosphorylation on Ser260 metabolites. Schuster^^^^' ^^^^' ^^^^ has identified
(ref. [1376]). some inhibitors that differ in their selectivity
between P450 24A1 and P450 27B1 and have
Polymorphisms in P450 24A1 have apparently
not been reported. sub-jxM IC5Q values.

6.48.6. Clinical Issues


6.48.3. Substrates and Reactions
The scheme presented in Figure 10.16 depicts
Both 25-hydroxyvitamin D3 and la,25-dihy- P450 24A1 as an enzyme involved in deactivating
droxyvitamin D3 are substrates for 24-hydroxyla- the activated form of vitamin D. The possibility
tion (Figure 10.16), with the latter being the has been considered that defects in P450 24A1
preferred substrate^^^^. However, human P450 might lead to hypervitaminosis TP^. An overactive
24A1 can also catalyze other side-chain reactions P450 24A1 could lead to vitamin D deficiency.
(Figure 10.17). Studies with side-chain-fluori- Henry^^^^ has reviewed the role of P450 24A1 and
nated vitamin D analogs also provide evidence for made comparisons to other "multistep" P450
some flexibility of this side chain in allowing enzymes. The possibility is raised that P450 24
P450 24A1 to oxidize^^^o, i38i ^^t P450 24A1 dif- could serve to generate products with their own
fers from the human ortholog in taking la,25- biological activities, with P450 24 thus being
dihydroxyvitamin D3 on to calcitroic acid instead involved in an anabolic pathway. Recently, trans-
of the products shown in Figure 10.17^^^^"^^^"^. genic rats overexpressing (rat) P450 24A1 were
found to have low plasma levels of 24,25-dihy-
droxy vitamin 03^^^^, which was unexpected.
6.48.4. Knowledge about Active Site Further, the transgenic rats developed albuminuria
Limited site-directed mutagenesis has been and hyperlipidemia shortly after weaning, and
done with P450 24A1. Expression studies using later developed atherosclerotic lesions in the
E. coli indicate species conservation in the aorta. These results raise the possibility that P450
residues 245-253 (F-helix), and the mutants 24A1 is involved in ftmctions other than vitamin
M246F and F249T showed changes in the ratio of D metabolism^^^^.
24-hydroxylation to other pathways^^^'^.
Studies by Jones' group^^^^ with analogs indi- 6.49. P450 26A1
cate that the site of hydroxylation by P450 24A1
depends on the distance from the ring to the site Retinoic acid is a vitamin A derivative that
(23 or 24), and not by the distance beyond this. plays an important role in gene regulation
Schuster's group has developed pharmacophore and development. The metabolism of retinoic acid
models^^^^. has been a matter of interest for some time, and
456 F. Peter Guengerich

the oxidation of retinoic acid by some hepatic 6.51. P450 26C1


P450s has been demonstrated^^^' ^^2, i39o
However, the relevance of these transformations to The only information presently available about
retinoic acid homeostasis in target tissues is not the CYP26C1 gene is its existence in the human
clear. White et alP^^ probed a panel of mRNAs genome^^^. Any suggestion of a role in retinoid
from mammalian cell lines with a cDNA from metabolism is only speculatory at this time.
a zebrafish P450 shown to be involved in
retinoic acid-inducible retinoic acid oxidation and
characterized P450 26A1 (ref [1391]). The
6.52. P450 27A1
heterologously expressed enzyme converted all- This is a mitochondrial enzyme that was
tmns-rQtinoic acid to the 4-hydroxy-, and 4-oxo-, characterized on the basis of two rather divergent
and 18-hydroxy products. The turnover numbers catal)^ic activities, the 25-hydroxylation of vita-
are unknown because the amount of P450 was min D3 (Figure 10.16) and the oxidation of cho-
not quantified, but the enzyme is clearly able to lesterol at the C27 position (Figure 10.18). Thus,
catalyze the oxidation of sub-iuiM additions of all- the enzyme bridges between hormone (vitamin D)
trans-YQtinoic acid^^^^. Apparently other retinoic and oxysterol pathways, and the clinical relevance
acid isomers are not substrates. of P450 27A1 is considerable.
The enzyme is expressed in cell lines derived
from several different tissues (kidney, lung, liver,
breast)^^^^. P450 26A1 has also been shown to be 6.52.1. Sites of Expression and
expressed in human fetal liver and brain tis- Abundance
sues^^^^' ^^^^, with a pattern differing from P450
26B1. The enzyme is localized in liver mitochondria.
P450 26A1 (and 26B1, vide infra) may func- Confusion existed in the early literature because
tion to protect fetal brain and possibly other tis- some animal species have liver microsomal vita-
sues from excess retinoic acid, which is known to min D3 25-hydroxylases (e.g., hog liver and kid-
be teratogenic, by metabolism. The interplay ney P450 2D25, (refs [1398], [1399])), but not
between P450s 26A1 and 26B1, and the signifi- humans''^^^. The rat and human liver mitochrondr-
cance of changes in their levels to normal tissue ial P450 27A1 recombinant enzymes were clearly
fimction is not yet known. shown to catalyze both vitamin D3 25-hydroxyla-
tion and the 27-hydroxylation of the side chains of
cholesterol and several derivatives^'^^^' ^'^^^.
Expression, at least at the mRNA level,
6.50. P450 26B1 has also been reported in leukocytes ^"^^^j skin
The CYP26B1 gene has 44% sequence identity fibroblasts''^^'^, kidney^"^^^ (and fetal liver and
with CYP26A1; it was found by searches of kidney'"^^^), and the arterial wall''^^^
genomic databases^^^^. The gene expression pat-
tern for P450 26B1 is different from that of P450
26A1 in mice^^^^ and also in human fetal
6.52.2. Regulation and Induction
brain^^^^ The localization of P450 26B1 in adult In a "normal" human population, the variation
human brain differs from that of P450 26A1, with in the steady-state P450 27A1 mRNA level was
a higher level of P450 26B1 mRNA in the cere- reported to be ~25-fold, compared with 60-fold
bellum ^^^^. Exactly how critical this enzyme is in for P450 7A1 in the same study'^"^^ However, at
brain function is unknown^^^^. The catalytic speci- least two polymorphisms ( > 1 % incidence, no dra-
ficity is very similar to that of P450 26A1, acting matic effect) and 42 mutations (rare alleles, usually
only on dd\-tmns retinoic acid and catalyzing the debilitating) are known^'^^'*' ^^^'^. Truncation muta-
formation of 4-hydroxy-, 4-oxo-, and 18-hydroxy tions are known^"^^^, as well as splice variants^'^^^.
(trans) retinoic acid^^^^. Defects in the CYP27A1 gene are associated
As with P450 26A1, frirther studies are needed with a condition known as cerebrotendinous
to define the presence of any defects in the gene xanthomatosis (CTX), a rare, autosomal recessive
and what the consequences might be. disorder characterized by accumulation of
Human P450s 457

J ] /-OH J ] CO2H
^
^ ^ ^ A ^ J ^ ^ P450 27A1^ ^ x l I V P450 27A1^
^.~/L> '
HO'^^---'^
Cholesterol
HO' -CU CXJ
27-OH Cholesterol 3^0H-5-cholestenoic acid

P450 7A1
\ \ ^

rXM rX^" Y-^CO.H


^ - x A ^ J ^

Ho-V.''^"'OH
P450 27A1^ ^ . I l

HO^^ - - ^ • • • O H
V P450 27A1^
Vr^
H O ^- - ^ • • • O H
7a-0H cholesterol 7a,27-(OH)2 cholesterol 3/3,7a-(OH)2-5-cholestenoic acid

]f
' ^ ^ r . ^-

rAH M V /OCP ^
r-vX V°^"
cxc
^ Y S ^ " ^ P^50 27A1^ ^.v^Jv^^Ly P450 27A1^

0^=^V.A.^".OH o<^^.AJ'-^OH 0^
7a-OH-4 cholestene-3-one 7a;27-(OH)2-4'Cholestene-3-one 7a-OH-3-oxo-4-cholestenolc acid

CO2H

O^ " ^ ^ "OH HO '•^^ ^-^ "OH


7c^12a-(OH)2-4-cholestene-3-one Chenodeoxycholic acid

CO2H

5j3-Cholestane-3a,7a,12a-triol 5j3-Cholestane-3o^7a^12(z,27-tetrol ZoL, la, 12c»-(OH)3 coprostanoic acid

P450 3A4
f
OH^

CO2H

5j3-Cholestane-3 0^70^12a^25-tetrol

Figure 10.18. Bile acid synthesis from cholesterol^^^^. The steps shown with dashed arrows are tentative.

cholestanol and cholesterol in many tissues. The signs, myelopathy, and peripheral neuropathy^^'
clinical manifestations include tendon xanthoma, ^^^^.
premature cataracts, juvenile atherosclerosis, and a Several aspects of regulation of the CYP27A1
progressive neurological syndrome involving men- gene have been studied. In rats, the enzyme can be
tal retardation, cerebellar atoxia, pyramidal tract induced by gonadotropin^"^^^. A hamster model
458 F. Peter Guengerich

showed downregulation of the gene in cholestatic (human) P450 2D6 abolished the activity toward
liver^^^^, although human P450 27A1 (used in vitamin 03^"^^^.
HepG2 cells) was not subject to negative feedback
regulation^'^^^
6.52.5. Inhibitors
Apparently little specific work has been done
6.52.3. Substrates and Reactions
on inhibition of this enzyme. Inhibition of this
Expanding on previous discussion, P450 27A1 enzyme by a drug would probably be undesirable.
catalyzes the 25-hydroxylation of vitamin D3
(Figure 10.16), la-hydroxyvitamin D3, vitamin
D2, and la-hydrox3rvitamin D2, and also the 27- 6.52.6. Clinical Issues
hydroxylation of cholesterol and several deriva-
Low serum 25-hydroxyvitamin D3 concentra-
tives (Figure lO.lS)^^!^' ^^^i. The cholesterol
tions have been reported in a variety of other med-
alcohols are further oxidized by the enzyme to
ical conditions and are considered to be potential
aldehydes and then carboxylic acids''^^^. The
problems^'^^^. Although CTX is linked with defec-
available information suggests release of the inter-
tive P450 27A1 (ref [29]), there are a number of
mediates in the pathway^^^^. The regioselectivity
enigmas about the etiology. A heterozygote
of the enzyme is considered to be a function of the
showed frontal lobe dementia and abnormal cho-
distance of the hydroxylation site to the end of the
lesterol metabolism^'^^^. Compound heterozygous
side chain^^^^.
mutations have also been reported to cause a vari-
More detailed analysis of the vitamin D3 reac- ation of CTX ^^^l
tion has been with E. coli recombinant P450
Bjorkhem has recently reviewed the issue of
27A1, with evidence for the following products
whether oxysterols (e.g., hydroxycholesterol) con-
(from vitamin D3): 25-hydroxy, 26-hydroxy, 27-
trol cholesterol homeostasis ^'*^^. Studies with
hydroxy, 24i^-25-dihydroxy, la,25-dihydroxy, 25,
rodents and cultured cells have not been very clear
26-dihydroxy, 25-,27-dihydroxy, 27-oxo, and an
to date. For instance, disruption of the mouse
unidentified dehydrogenated product^'^^^' ^^^^.
CYP27A1 gene yielded reduced bile acid synthe-
sis but apparently caused no change in levels of
cholesterol or la,25-dihydroxyvitamin 03^"*^^
6.52.4. Knowledge about Active Site
P450 27A1 is constitutively expressed in the nor-
Some information about the roles of amino mal artery wall and is substantially upregulated in
acids can be inferred from the knowledge of alle- atherosclerosis, and the possibility has been raised
les involved in CTX; many of these proteins were that P450 27A1 may be a protective mechanism
unstable when attempts were made at heterologous for removing cholesterol ^'*^^. Further, immune
expression^'*^^. Other work by Pikuleva et al}^^^ complexes and IFN-7 decreased P450 27A1
with the putative F and G helices has shown dif- expression in human aortic endothelial cells,
ferences due to substitution of Phe207, Ile211, peripheral blood mononuclear cells, monocytes-
Phe215, Trp235, and Tyr238. Interestingly, the derived macrophages, and a human monoc3^oid
121 IK and F215K mutations affected the regiose- cell line, suggesting downregulation of P450
lectivity and enabled the enzyme to catalyze C-C 27A1 to maintain cholesterol homeostasis in the
bond cleavage. Further work with mutants in this arterial wall^"^^^.
region led to weaker association of P450 27A1 In Cyp27Al~'~ mice, a dramatic increase in the
with the membrane, and some of the nonconserva- level of P450 3A enzymes is seen; some sterols
tive changes yielded impaired catalytic activity ^^^^. accumulate and induce via the mouse PXR sys-
Human P450 27A1 can be contrasted with ^gj^i423 P45Q 3^4 j^^g gQj^g side-chain hydroxyla-
porcine P450 2D25, which also catalyzes vitamin tion activities toward cholesterol-derived
D3 25-hydroxylation. The only human P450 2D sub- sterols^^^ However, elevated P450 3A4 activities
family enzyme which does not have activity toward were not increased in CTX^^^ indicating a differ-
vitamin D is P450 2D6. Further, changing a set of ence in the murine and human systems. Recently,
residues of porcine 2D25 to their counterparts in Escher et al}^'^^ have reported that cholesterol
Human P450s 459

efflux in CHOP cells is enhanced by (heterolo- vitamin D-related activities, including skin (basal
gous) expression of human P450 27A1, and the keratinocytes, hair follicles), lymph nodes (granu-
authors suggest this as part of a protective system lomata), colon (epithelial cells and parasympa-
against atherosclerosis. The basis is probably the thetic ganglia), pancreas (islets), adrenal medulla,
ability of 27-hydroxycholesterol to act as an brain (cerebellum and cerebral cortex) ^^^'^, pla-
endogenous ligand for the liver X receptor in cho- centa (decidual and trophoblastic cells)^^^'^~^^^^,
lesterol-loaded cells ^'^^^. cervix^"^^^, and parathyroid glands^"^^^. Thus, P450
In considering the general question of whether 27B1 may be an intracrine modulator of vitamin D
oxysterols (e.g., 27-hydroxycholesterol) control function in peripheral tissues^'^^'*. The expression
cholesterol homeostasis, the hypothesis is still of P450 27B1 was elevated in parathyroid adeno-
open and the rodent data are not totally clear here. mas, but attenuated in carcinomas, relative to nor-
Bjorkhem^^'*^ has made the point that humans mal parathyroid tissue ^^^^.
lacking P450 27A1 have normal circulating levels
of cholesterol.
6.53.2. Regulation and Polymorphism
Although the CYP27B1 gene is only 5 kb in
6.53- P450 27B1 size, the regulation is quite complex. The pro-
moter is in the -85/+22 region and requires
As discussed earlier, vitamin D is an important a functional CCATT element. Three consensus
hormone. A critical step in activation is la- AP-1 sites are upstream^"^^^. Enzyme activity has
hydroxylation^"^^^ (Figure 10.16). Early work long been known to be stimulated by low phos-
established the P450 nature of the enzyme, local- phorus diets (in animal models)^"^"^^j and more
ized in kidney mitochondria^'*^^. Subsequent work recently, this phenomenon has been linked to a
demonstrated that the la- and 24-hydroxylation growth hormone mechanism^^^^' ^'*'^^; its relevance
activities could be attributed to different in humans is not known.
enzymes^'*^^' ^^'^^. Some early work had suggested
Expression is also regulated by calcium,
that the la- and 25-hydroxylation activities were
parathyroid hormone, and by the product la,25-
associated with the same enzyme^^^^, but later
dihydroxyvitamin 03^^"*^' ^^^. Regulatory regions
work showed that these activities were due to
involving the responses to parathyroid hormone,
P450 27B1 and 27A1, respectively
calcitonin, and la,25-hydroxyvitamin D3 are
located in the region —0.4 to —0.5 kb^"^"^^.
Forskolin also regulates gene expression, and an
6.53.1. Sites of Expression and
Spl site is involved in aspects of regulation^"^"^^' ^^'^'^.
Abundance
Complexity is seen in different models.
The cloning of the human cDNA for what is Parathyroid hormone-related protein and Ca^^
now known as P450 27B1 established the kidney have conflicting actions in a nude rat model of
mitochondrial P450 (27B1) as the vitamin D3 la- humoral hypercalcemia of malignancy^"^"^^. In dif-
hydroxylase^'*^^ The gene has nine exons and ferentiated Caco cells, there is upregulation of
spans only 5 kb^"^^^. P450 27B1 expression by la,25-dihydrox5rvita-
P450 27B1 is expressed in many parts of the min D3 and epidermal growth factor, but down-
human kidney, including the distal convoluted regulation in less differentiated Caco cell lines^^^^.
tubule, the cortical and medullary part of the col- Another aspect of regulation of P450 27B1 is
lecting ducts, and the papillary epithelia^"^^^. genetic. P450 27B1 results in Type I vitamin D-
Lower expression was observed along the thick dependent rickets^'^'*^. The genetics have been
ascending limb of the loop of Henle and established in more than 30 patients involving at
Bowman's capsule. Some weaker expression was least 20 mutations^'^^^' ^^^^. At least 13 missense
observed in glomeruli or vascular structures. In mutations have been observed, none of which
normal humans, the distal nephron is the predom- encode an active protein. Some of the mutants are
inant site of expression^^^^. splicing defects^"^^^. Some mutations in CYP27B1
P450 27B1 is also expressed in many are also involved in what is termed pseudovitamin
extrarenal sites (human) where it is involved in D-deficiency rickets^"^^^' ^^^^. Beyond these
460 F. Peter Guengerich

debilitating mutations, little information is avail- responsible for vitamin D-dependent rickets Type
able about actual polymorphisms. I^"^^^. At least 30 different mutations are known in
patients^'^'^^' ^'^^^. Even the "mild" phenotype of
Type I rickets is due to deficiency in P450 27B1
6.53.3. Substrates and Reactions (ref [1461]).
P450 27B1 can catalyze the la-hydroxylation CYP27B1 knockout mice have been character-
of both 25-hydroxy and 24(i?),25-dihydroxyvita- ized and show a typical rickets phenotype ^'^^^.
min D3^^^^' ^^^^ (Figure 10.16). The intrinsic activ- Another mouse model in which the gene has
ity (^ca/^m) ^^^ ^^^ recombinant human enzyme is been ablated showed skeletal, reproductive, and
better for 24(i?),25-hydroxy vitamin D3, but this immune dysfunction^^^^. Rickets was also observed
does not mean that this is the favored substrate in in a conditional knockout model^"^^^.
the cell, because of the balance of vitamin D Patients with severe renal insufficiency show
metabolites regulated by P450s 24A1 and 27A1 attenuated la-hydroxylation activity^^^^.
(ref [29]). Apparently the 25-hydroxy group is an Another aspect of P450 27B1 research
obligatory requirement^^^^' ^^^^. involves cancer. Increased activity was reported in
parathyroid tumors ^'^^^. Some splice variants of
the CYP27B1 gene (coding for truncated proteins)
6.53.4. Knowledge about Active Site were amplified in human (brain) gliomas ^'^^^.
Reports have also appeared on the relationship of
Some information is available from the natural
P450 27B1 expression to various biological
mutants of P450 27B1, even if the basis for loss of
processes in human nonsmall cell lung carcino-
activity is not obvious. Inouye's group^'^^'^ has pro-
mas'^^^, colon tumors''^^^"^'^^^, and prostate can-
vided evidence that ArglOT, Glyl25, and Pro497
^gj.gi47i, 1472^ generally with decreased expression
are not simply involved in binding substrate but
in tumors.
required for proper folding. It was also suggested
Finally, la,25-dihydroxyvitamin D3 is used to
that Arg389 and Arg453 are involved in heme
treat psoriasis, and patients can develop resist-
binding and that Asp 164 stabilizes the bundle of
ance. An experimental model for therapy involves
the four helices D, E, I, and J. Thr321 is suggested
enhancement of the endogenous production of
to be involved in O2 activation^"^^"^. The natural
la,25-dihydroxyvitamin D3 by gene therapy^'^^^.
mutants L343F and E189G show partial activity
and the individuals bearing these have only mar-
ginal impairment ^'*^^.
6.54. P450 27C1
As with some of the other P450s, the only
6.53.5. Inhibitors knowledge currently available is the existence of
Little has been done because impairment of the CYP27C1 gene in the human genome^^^.
this enzyme is a clinical problem. Some thiavita- Suggestions that this enzyme is involved in vita-
min D analogs have been evaluated in animal min D metabolism are still only speculatory.
models^"^^^.

6.55. P450 39A1


6.53.6. Clinical Issues
An expression-cloning approach was utilized
The significance of the enzyme is due to the to isolate a cDNA from {Cyp7bl~'~) mice that
pleiotropic actions of the active form of vitamin could, when expressed, catalyze the 7a-hydroxyla-
D, la,25-dihydroxyvitamin D3, which include tion of 24-hydroxycholesterol^'^^'^. P450 39 has a
regulation of calcium homeostasis, control of microsomal location with a preference for the sub-
bone cell differentiation, and modification of strate 24-hydroxycholesterol and is expressed in
immune responses^^^^. The la-hydroxylation liver. Presumably these characteristics of mouse
reaction is rate limiting and hormonally con- P450 39 apply to the human ortholog but no fur-
trolled. The expression of the gene is usually ther information is yet available. Potential rele-
tightly regulated (vide supra), but gene defects are vance of this enzyme is in the inactivation of
Human P450s 461

24-hydroxycholesterol, a ligand for the LXR for many years and then demonstrated the reaction
nuclear hormone receptor (see Section 6.56 on in rat liver microsomes in 1994^^^^ Subsequently
P450 46, vide infra), the reaction was also demonstrated in rat brain
microsomes ^^^^.

6.56- P450 46A1


6.57.1. Sites of Expression and
An expression cloning approach was utilized Abundance
to clone cDNAs encoding both murine and human
cholesterol 24-hydroxylase, P450 46A1 (ref. Waterman's group identified the human
[1475]). The mouse and human sequences are CYP51A1 gene and two pseudogenes^"^^^. mRNA
95% identical. Expression is predominantly in blot analysis showed the highest levels in testis,
brain (neurons in several regions). The enzyme is ovary, adrenal, prostate, liver, kidney, and lung. In
in the endoplasmic reticulum; alternate substrates mouse testis, P450 51A1 was localized in both
have not been explored but may be unlikely. round and elongated spermatids ^'^^^. The enzyme
is also found in (rodent) oocytes^"^^^.
The significance of P450 46A1 rests in the
fact that the brain is the most cholesterol-rich
tissue in the body^"^^^. LXRs, members of the 6.57.2. Regulation and Polymorphism
nuclear hormone receptor family, are activated
by 24-hydroxycholesterol. LXRp and P450 46 Polymorphisms in the human CYP51A1 gene
have overlapping expression patterns in brain^'*'^^. have not been reported nor have debilitating
The system may be counter-balanced by P450 39, mutants been defined. However, Kelley et al}^^^
which catalyzes the 7a-hydroxylation of reported a patient with Antley-Bixler syndrome
25-hydroxycholesterol (Section 6.55). and ambiguous genitalia with lanosterol accumu-
lation and an apparent defect in P450 51A1.
Recently, there has been considerable interest
With regard to regulation of the human gene,
in the relationship between P450 46A1 and
primer extension studies indicated predominant
Alzheimer's Disease. P450 46A1 had a marked
transcription initiation sites in liver, lung, and kid-
difference in distribution in the brains of normal
ney, and placenta 250 and 249 bp upstream from
and Alzheimer's patients, with less staining of
the translation start site and a second major site at
neuronal cells but more of glial cells in diseased
-lOObp, with the absence of TATA and CAAT
patients^'^^^. Also, elevated levels of 25(5)-hydroxy-
patterns and a GC-rich sequence in the promoter
cholesterol were found in cerebral spinal fluid in
region^"^^^. Multiple (rat) testis-specific transcripts
the early stages of dementia^^^^. Associations^"^^^
arise from differential polyadenylation site
and lacks of associations ^"^^^ between CYP46
usage ^'*^^.
SNPs and Alzheimer's Disease have been
reported. One of the issues in the research with In human adrenocortical H295R cells (in cul-
P450 46A1 is the apparently very low rate of ture), cholesterol deprivation led to a 2.6-3.8-fold
cholesterol 24-hydroxylation (although an exact induction of P450 51A1 mRNA, which was sup-
rate can be deduced from the published informa- pressed by the addition of 25-hydroxycholes-
tion). Recently, Pikuleva's laboratory has found terol^'^^^. In the liver and other somatic tissues,
that 24-hydroxycholesterol is a much better sub- CYP51 is regulated by a sterol/sterol-regulatory
strate for recombinant P450 46A1 than is choles- element binding protein (SREBP)-dependent
terol, being oxidized to as yet uncharacterized pathway^"^^^. In testis, cAMP/cAMP-responsive
producti480a element modulator (CREM)^-dependent regula-
tion predominates. Spl fiinctions to maximize the
sterol regulatory pathway of P450 51 (ref. [1490]).
6.57. P450 51A1 Insulin is an essential factor in "basal" expres-
sion of P450 51 in rat liver, with possible involve-
Lanosterol is an important intermediate in ment of SREBP-lc involvement^'^^^. In a porcine
cholesterol synthesis, and 14a-demethylation vascular endothelial cell model (and in arterial
has been established as a step in the pathway. wall), LDLs downregulated P450 51 through an
Yoshida's laboratory had studied the yeast enzyme SREBP-2 mechanismi492.
462 F. Peter Guengerich

6.57.3. Substrates and Reactions 6.57.6. Clinical Issues


Stimulation of human P450 51 activity by b^ in Most of the work discussed here is from exper-
a reconstituted system has been reported by imental studies on the possible role of P450 51 in
Kelly's laboratory^'*^^. reproduction, and the translation of phenomena
The normal mammalian substrate for P450 51 fi-om animal models to humans is still somewhat
is 24,25-dihydrolanosteroli^^^, with the 14a- speculatory. However, the very high level of P450
demethylation process proceeding in what are 51 expression in postmeiotic haploid spermatids is
assumed to be three consecutive steps, as with striking. The action of P450 51 is proposed to lead
some other P450s, for example, l l A l , 17A1, to the production of signaling steroids in haploid
19A1. Interestingly, both human and yeast germ cells ^^^^. Meiosis-activating substances
{Candida albicans) P450 51 showed relatively lit- (MAS) are produced by 14-reduction of products of
tle selectivity among a closely related group of the action of P450 51 on lanosterol^^^^. Follicular
analogs^^^'*. It is also interesting to note that even fluid MAS (FF-MAS) is formed from lanosterol in
though this P450 has a relatively defined role in rat spermatids *^^^ Yoshida's group has reported
a physiological process, the kinetic parameters gonadotropin-dependent expression of P450 51 in
are relatively poor among P450s {k^JK^ = 300 rat ovaries and the production of MAS ^^^^.
The reaction and possible physiological signif-
icance of the system in reproduction have been
reviewed recently by Rozman^^^^. Leydig cells
6.57.4. Knowledge about Active Site and acrosomes of spermatids have the highest
A crystal structure of human P450 51 is not P450 51 levels, and primary mouse oocytes and
yet available but high resolution structure of the granulose cells also contain P450 51. The MAS
soluble Mycobacterium tuberculosis P450 51 is may have a role in fertilization •^^^.
(ref [1495]). Two notable features are a bent I
helix and an open conformation of the BC loop.
The bacterial structure has been utilized in con- 7. Concluding Remarks
sideration of mammalian models, and the SRS
predictions do not seem to apply well^"^^^. Further, Some information has been presented about the
the mutation hotspots for known azole-resistant known P450s. Because the genome is nearly com-
C. albicans P450 51 mutants tend to be outside the pleted, no more human P450s are likely to be added
predicted active site and suggest the contribution unless our views of marker sequences change.
of long-range effects on ligand binding''^^^' ^'^^^. Although much of the interest in human P450 is
Studies on Arg448 indicate that, despite directed toward drug metabolism, the majority of
sequence conservation, aspects of the role of this P450s appear to be involved in the metabolism of
and other residues are different for human and endobiotics (Table 10.2). In one sense, the fact that
yeast P450 51 (ref [1498]). about 5 or 6 of the P450s are so dominant in xeno-
biotic metabolism (Figure 10.3) is not surprising
when we recognize that only about 15 of the 57 use
6.57.5. Inhibitors
xenobiotics as substrates (Table 10.1) and consider
Most of the interest in inhibition has been with the dominant levels of expression of a few P450s
fungal P450 51, as a target for antimycotic drugs. that have rather broad selectivity (Figure 10.4).
The goal is to select candidate drugs inhibitory to Aside from the current problems already men-
fungal P450 51, but not human P450 51. tioned, including practical issues, what are some of
Some work on the interaction of azoles with the future challenges and in what areas should
human P450 51 has been published^'^^^. Although work be done? Some of the problems are very
human P450 51 has been suggested as a target for basic, such as the goals of deriving more experi-
cholesterol-lowering drugs, apparently little has mental three-dimensional structures, answering
been done and potential toxicity due to the questions about catalytic mechanisms, and under-
steroidogenic and potential germ cell side effects standing the complexities of regulation of these
{vide infra) could be an issue. genes.
Human P450s 463

NCH3

H3C0. H3CO.

NCH3 NCH3

Neopinone

Figure 10.19. Steps in mammalian morphine synthesis^'

There is considerable opportunity to improve there is an opportunity for the introduction of


screening and predictability for use with new novel approaches.
chemical entities in the pharmaceutical industry. Note added in proof. Recently Pai et alP^^
Another open issue is whether large-scale SNP have demonstrated an unusual phenomenon with
analysis can be practical in drug development. the pseudogene, CYP2D7. A frame shifted allelic
Another area is understanding the relevance of variant yields expression of an enzyme specifi-
variations in P450s (both SNPs and expression cally in the brain, not liver, and this enzyme is
levels) to diseases, not only drug therapies. For more efficient than P450 2D6 in converting
instance, one can ask if some of the "minor" sterol codeine to morphine (Figure 10.19).
products produced by P450 3A4 have any impact
on cardiovascular disease. How important are
some of the P450s that oxidize arachidonic acid to Acknowledgments
various products (in humans)? Does a P450 2D6
deficit have any relevance to long-term health? Research in the author's laboratory has been
Finally, there are two more major problems supported in part by USPHS grants ROl CA90426
in understanding what human P450s do. One and P30 ES00267. Thanks are extended to the
problem is the oxidation reactions, largely involv- many individuals who provided unpublished
ing endobiotics, for which no P450s have been results, to Drs. E.M.J. Gillam, F.F. Kadlubar, and
defined. One example is the synthesis of endoge- M.R. Waterman for comments, and to present and
nous morphine in the body, which appears to past members of the author's laboratory for their
require multiple oxidations and is subject to short- involvement in some of the original research. This
term regulation^^^"^' ^^^^ (Figure 10.19). Although chapter is dedicated to Dr. Tsutomu Shimada, my
P450 2D6 can oxidize codeine to morphine^^^^, long-term collaborator, on the occasion of his
this enzyme does not contribute to the endogenous retirement from the Osaka Prefectural Institute of
formation of morphine ^^^^. What other oxidative Public Health (March 2003), and to my postdoc-
reactions remain to be discovered and character- toral mentor. Prof M.J. Coon, who introduced me
ized? In the past year, the oxidative dealkylation to these enzymes 30 years ago.
in DNA repair was demonstrated to involve an
ct-ketoglutarate-dependent dioxygenase^^^^' ^^^^.
Perhaps similar roles for P450s may be found. References
The other issue is identifying functions for the
remaining human P450s, of which there are at 1. Distlerath, L.M. and F.F. Guengerich (1987).
least 15 (Table 10.2). Doing this on a step-by-step Enzymology of human liver cytochromes P-450.
basis as in the past will not be very efficient, and In F.P. Guengerich (ed.) Mammalian Cytochromes
464 F. Peter Guengerich

P-450, Vol. 1. CRC Press, Boca Raton, FL, Characterization of rat and human liver microsomal
pp. 133-198. cytochrome P-450 forms involved in nifedipine oxi-
2. Beaune, R, R Dansette, J.R Flinois, S. Columelli, dation, a prototype for genetic polymorphism in
D. Mansuy, and J.R Leroux (1979). Partial purifica- oxidative drug metabolism. J. Biol. Chem. 261,
tion of human liver cytochrome P-450. Biochem. 5051-5060.
Biophys. Res. Commun. 88, 826-832. 13. Watkins, RB., S.A. Wrighton, R Maurel,
3. Wang, R, PS. Mason, and F.R Guengerich (1980). E.G. Schuetz, G. Mendez-Picon, G.A. Parker et al.
Purification of human liver cytochrome P-450 and (1985). Identification of an inducible form of
comparison to the enzyme isolated from rat liver. cytochrome P-450 in human liver. Proc. Natl. Acad.
Arch. Biochem. Biophys. 199, 206-219. Sci. 0X4 82,6310-6314.
4. Wang, PR, P. Beaune, L.S. Kaminsky, G.A. Dannan, 14. Wrighton, S.A., C. Campanile, RE. Thomas,
FR Kadlubar, D. Larrey et al. (1983). Purification S.L. Maines, RB. Watkins, G. Parker et al. (1986).
and characterization of six cytochrome P-450 Identification of a human liver cytochrome P-450
isozymes from human liver microsomes. homologous to the major isosafrole-inducible
Biochemistry 11, 5375-5383. cytochrome P-450 in the rat. Mol. Pharmacol. 29,
5. Mahgoub, A., J.R. Idle, L.G. Dring, R. Lancaster, 405^10.
and R.L. Smith (1977). Polymorphic hydroxylation 15. Wrighton, S.A., RE. Thomas, R Willis, S.L. Maines,
of debrisoquine in man. Lancet ii, 584-586. PB. Watkins, W Levin et al. (1987). Purification of
6. Shimada, T., C.-H. Yun, H. Yamazaki, J.-C. Gautier, a human liver cytochrome P-450 immunochemi-
RH. Beaune, and FR Guengerich (1992). cally related to several cytochromes P-450 purified
Characterization of human lung microsomal from untreated rats. J. Clin. Invest. 80, 1017-1022.
cytochrome P-450 lAl and its role in the oxidation 16. Gonzalez, F.J. (1989). The molecular biology of
of chemical carcinogens. Mol. Pharmacol. 41, cytochrome P450s. Pharmacol. Rev. 40, 243-288.
856-864. 17. Larson, J.R., M.J. Coon, and T.D. Porter (1991).
7. Distlerath, L.M., PE.B. Reilly, M.V. Martin, Alcohol-inducible cytochrome P-450IIE1 lacking
G.G. Davis, G.R. Wilkinson, and FP Guengerich the hydrophobic NH2-terminal segment retains cat-
(1985). Purification and characterization of the alytic activity and is membrane-bound when
human liver cytochromes P-450 involved in debriso- expressed in Escherichia coli. J. Biol. Chem. 266,
quine 4-hydroxylation and phenacetin O-deethylation, 7321-7324.
two prototypes for genetic polymorphism in oxidative 18. Li, Y.C. and J.Y.L. Chiang (1991). The expression of
drug metabolism. J. Biol. Chem. 260, 9057-9067. a catalytically active cholesterol 7a-hydroxylase
8. Yun, C.-H., T. Shimada, and FR Guengerich (1991). cytochrome P-450 in Escherichia coli. J. Biol.
Purification and characterization of human liver Chem. 266, 19186-19191.
microsomal cytochrome P-450 2A6. Mol. 19. Barnes, H.J., M.P Arlotto, and M.R. Waterman
Pharmacol. 40, 679-685. (1991). Expression and enzymatic activity of
9. Shimada, T., K.S. Misono, and FP. Guengerich recombinant cytochrome P450 17a-hydroxylase in
(1986). Human liver microsomal cytochrome P-450 Escherichia coli. Proc. Natl. Acad. Sci. USA 88,
mephenytoin 4-hydroxylase, a prototype of genetic 5597-5601.
polymorphism in oxidative drug metabolism. 20. Guengerich, FP, E.M.J. Gillam, and T. Shimada
Purification and characterization of two similar (1996). New applications of bacterial systems to
forms involved in the reaction. J. Biol. Chem. 261, problems in toxicology. Crit. Rev. Toxicol. 26,
909-921. 551-583.
10. Gut, J., T. Catin, R Dayer, T. Kronbach, U. Zanger, 21. Nebert, D.W, M. Adesnik, M.J. Coon,
and U.A. Meyer (1986). Debrisoquine/sparteine- R.W Estabrook, F.J. Gonzalez, FP. Guengerich
type polymorphism of drug oxidation: Purification et al. (1987). The P450 gene superfamily:
and characterization of two functionally different Recommended nomenclature. DNA 6, 1-11.
human liver cytochrome P-450 isozymes involved 22. Nelson, DR., T Kamataki, D.J. Waxman,
in impaired hydroxylation of the prototype substrate FP. Guengerich, R.W. Estabrook, R. Feyereisen
h\xfyxx2\o\. J. Biol. Chem. 261, 11734-11743. et al. (1993). The P450 superfamily: Update on new
11. Birgersson, C , E.T. Morgan, H. Jornvall, and C. von sequences, gene mapping, accession numbers, early
Bahr (1986). Purification of a desmethylimipramine trivial names of enzymes, and nomenclature. DNA
and debrisoquine hydroxylating cytochrome P-450 Cell Biol. 12, 1-51.
from human liver. Biochem. Pharmacol. 35, 23. Guengerich, FR (1995). Human cytochrome P450
3165-3166. enzymes. In PR. Ortiz de Montellano (ed.).
12. Guengerich, RR, M.V Martin, RH. Beaune, Cytochrome P450. Plenum Press, New York,
R Kremers, T. Wolff, and D.J. Waxman (1986). pp. 473-535.
Human P450s 465

24. Niranjan, B.G., N.M. Wilson, C.R. Jefcoate, and Factors in the Clinical Evaluation of Medicines.
N.G. Avadhani (1984). Hepatic mitochondrial Kluwer Academic Publishers, Dordrecht/Boston/
cytochrome P-450 system: Distinctive features of London, pp. 13-26.
cytochrome P-450 involved in the activation of afla- 37. Guengerich, RR and D.C. Liebler (1985). Enzy-
toxin Bj and benzo(a)pyrene. J. Biol. Chem. 259, matic activation of chemicals to toxic metabolites.
12495-12501. Crit. Rev Toxicol. 14, 259-307.
25. Addya, S., H.K. Anandatheerthavarada, G. Biswas, 38. Motulsky, A.G. (1957). Drug reactions, enzymes
S.V Bhagv^at, J. Mullick, and N.G. Avadhani and biochemical genetics. J. Am. Med. Assoc. 165,
(1997). Targeting of NH2-terminal-processed 835-837.
microsomal protein to mitochondria: A novel path- 39. Kalow, W (1962). Pharmacogenetics. W B. Saunders,
way for the biogensis of hepatic mitochondrial Philadelphia, PA.
P450j^T-2- -^ Cell Biol 139, 589-599. 40. Remmer, H. (1957). The acceleration of evipan oxi-
26. Robin, M.A., H.K. Anandatheerthavarada, dation and the demethylation of methylaminopyrine
G. Biswas, N.B. Sepuri, D.M. Gordon, D. Pain et al. by barbiturates. Naunyn-Schmiedebergs Arch. Exp.
(2002). Bimodal targeting of microsomal CYP2E1 Pathol. Pharmakol. 231, 296-307.
to mitochondria through activation of an N-terminal 41. Keeney, D.S. and M.R. Waterman (1993). Regu-
chimeric signal by cAMP-mediated phosphoryla- lation of steroid hydroxylase gene expression:
tion. J. Biol. Chem. 277, 40583-40593. Importance to physiology and disease. Pharmacol.
27. Tateishi, T., Y. Krivoruk, Y.-F. Ueng, A.J.J. Wood, Then 58,301-317.
F.R Guengerich, and M. Wood (1996). Identifi- 42. Conney, A.H., E.G. Miller, and J.A. Miller (1956).
cation of human liver cytochrome P450 3A4 as The metabolism of methylated aminoazo dyes.
the enzyme responsible for fentanyl and sulfentanyl V Evidence for induction of enzyme synthesis in
^-&Q^\ky\dX\on. Anesth. Analg. 82, 167-172. the rat by 3-methylcholanthrene. Cancer Res. 16,
28. Shimada, T., H. Yamazaki, 4. Mimura, Y. Inui, and 450-459.
P.P. Guengerich (1994). Interindividual variations in 43. Eichelbaum, M., N. Spannbrucker, B. Steincke, and
human liver cytochrome P450 enzymes involved in H.J. Dengler (1979). Defective N-oxidation of
the oxidation of drugs, carcinogens, and toxic chem- sparteine in man: A new pharmacogenetic defect.
icals: Studies with liver microsomes of 30 Japanese Eur. J. Clin. Pharmacol. 16, 183-187.
and 30 Caucasians. J. Pharmacol. Exp. Then 270, 44. Woolhouse, N.M., B. Andoh, A. Mahgoub, T.P Sloan,
414^23. J.R. Idle, and R.L. Smith (1979). Debrisoquin
29. Nebert, D.W. and D.W. Russell (2002). Clinical hydroxylation polymorphism among Ghanaians and
importance of the cytochromes P450. Lancet 360, Caucasians. Clin. Pharmacol. Ther 26, 584-591.
1155-1162. 45. Johansson, I., E. Lundqvist, L. Bertilsson,
30. Brodie, A.M.H. (1985). Aromatase inhibition and its M.L. Dahl, R Sjoqvist, and M. Ingelman Sundberg
pharmacologic implications. Biochem. Pharmacol. (1993). Inherited amplification of an active gene in
34,3213-3219. the cytochrome P450 CYP2D locus as a cause of
31. Gonzalez, RJ. and S. Kimura (2003). Study of P450 ultrarapid metabolism of debrisoquine. Proc. Natl.
function using gene knockout and transgenic mice. Acad Sci. USA 90, 11825-11829.
Arch. Biochem. Biophys. 409, 153-158. 46. Smith, R.L., J.R. Idle, A.A. Mahgoub, T.R Sloan,
32. Rendic, S. (2002). Summary of information on and R. Lancaster (1978). Genetically determined
human CYP enzymes: Human P450 metabolism defects of oxidation at carbon centres of drugs.
data. DrugMetab. Rev. 34, 83^48. Lancet i, 943-944.
33. Evans, WE. and M.V Relling (1999). 47. Gonzalez, F.J., R.C. Skoda, S. Kimura, M. Umeno,
Pharmacogenomics: Translating function genomics U.M. Zanger, D.W. Nebert et al. (1988). Characteri-
into rational therapeutics. Science 286, 487^91. zation of the common genetic defect in humans
34. Guengerich, RR andT. Shimada (1991). Oxidation deficient in debrisoquine metabolism. Nature 331,
of toxic and carcinogenic chemicals by human 442^46.
cytochrome P-450 enzymes. Chem. Res. Toxicol. 4, 48. Nakamura, K., R Goto, W.A. Ray, C.B. McAllister,
391^07. E. Jacqz, G.R. Wilkinson et al. (1985). Interethnic
35. Breimer, D.D. and J.H. Schellens (1990). A "cocktail" differences in genetic polymorphism of debrisoquin
strategy to assess in vivo oxidative drug metabolism and mephenytoin hydroxylation between Japanese
in humans. Trends Pharmacol. Sci. 11, 223-225. and Caucasian populations. Clin. Pharmacol. Then
36. Breimer, D.D. (1994). Genetic polymorphisms in 38, 402-408.
drug metabolism; clinical implications and conse- 49. Daly, A.K. (2003). Pharmacogenetics of the major
quences in ADME studies. In S. Walker, C. Lumley, polymorphic metabolizing enzymes. Fundam. Clin.
and N. McAuslane (eds), The Relevance of Ethnic Pharmacol. \1,21-A\.
466 F. Peter Guengerich

50. Nagata, K. and Y. Yamazoe (2002). Genetic isoform identification. Drug Metab. Dispos. 31,
polymorphism of human cytochrome P450 involved 345-350.
in drug metabolism. Drug Metab. Pharmacokinet. 64. Correia, M.A. and PR. Ortiz de Montellano (2004,
17, 167-189. in press). Inhibition and degradation of cytochrome
51. Daly, A.K., J. Brockmoller, F. Broly, M. Eichelbaum, P450 enzymes. In P. R. Ortiz de Montellano (ed.).
W.E. Evans, F.J. Gonzalez et al. (1996). Nomen- Cytochrome P450: Structure, Mechanism, and
clature for human CYP2D6 alleles. Pharmacogenetics Biochemistry. Plenum Press, New York (Chap. 7 of
6, 193-201. this monograph).
52. Gaedigk, A., M. Blum, R. Gaedigk, M. Eichelbaum, 65. Correia, M.A. (2004, in press). Isoform functional
and U.A. Meyer (1991). Deletion of the entire markers, isoform substrate specificities, and
cytochrome P450 CYP2D6 gene as a cause of fluorescent substrate assays. In PR. Ortiz de
impaired drug metabolism in poor metabolizers of Montellano (ed.). Cytochrome P450: Structure,
the debrisoquine/sparteine polymorphism. Am. J. Mechanism, and Biochemistry. Plenum Press,
Hum. Genet. 48, 943-950. New York (Appendix of this monograph).
53. Corchero, J., C.P. Granvil, T.E. Akiyama, 66. Harris, J.W., A. Rahman, B.-R. Kim,
G.P. Hayhurst, S. Pimprale, L. Feigenbaum et al. FP Guengerich, and J.M. Collins (1994).
(2001). The CYP2D6 humanized mouse: Effect of Metabolism of taxol by human hepatic microsomes
the human CYP2D6 transgene and HNF4a on the and liver slices: Participation of cytochrome P450
disposition of debrisoquine in the mouse. Mol. 3A4 and of an unknown P450 enzyme. Cancer Res.
Pharmacol. 60, 1260-1267. 54, 4026-4035.
54. Schuetz, E.G., D.R. Umbenhauer, K. Yasuda, 67. Tran, T.H., L.L. von Moltke, K. Venkatakrishnan,
C. Brimer, L. Nguyen, M.V Relling et al. (2000). B.W. Granda, M.A. Gibbs, R.S. Obach et al. (2002).
Altered expression of hepatic cytochromes P-450 in Microsomal protein concentration modifies the
mice deficient in one or more mdr^ genes. Mol. apparent inhibitory potency of CYP3A inhibitors.
Pharmacol. 57, 188-197. Drug Metab. Dispos. 30, 1441-1445.
55. Cummins, C.L., C.Y Wu, and L.Z. Benet (2002). 68. Austin, R.P, P Barton, S.L. Cockroft,
Sex-related differences in the clearance of M.C Wenlock, and R.J. Riley (2002). The influence
cytochrome P450 3A4 substrates may be caused by of nonspecific microsomal binding on apparent
P-glycoprotein. Clin. Pharmacol. Ther 72, 474-489. intrinsic clearance, and its prediction from physico-
56. Renton, K.W. and L.C. Knickle (1990). Regulation chemical properties. Drug Metab. Dispos. 30,
of hepatic cytochrome P-450 during infectious dis- 1497-1503.
ease. Can. J. Physiol. Pharmacol. 68, 177-lSl. 69. Yoo, J.S.H., R.J. Cheung, CJ. Patten, D. Wade,
57. Kato, R. and Y. Yamazoe (1992). Sex-specific and CS. Yang (1987). Nature of 7V-nitrosodi-
cytochrome P450 as a cause of sex- and species- methylamine demethylase and its inhibitors. Cancer
related differences in drug toxicity. Toxicol. Lett. Res. 47, 3378-3383.
64/65, 661-667. 70. Chauret, N., A. Gauthier, and D.A. Nicoll-Grififith
58. Komori, M., K. Nishio, M. Kitada, K. Shiramatsu, (1998). Effect of common organic solvents on in
K. Muroya, M. Soma et al. (1990). Fetus-specific vitro cytochrome P450-mediated metabolic activi-
expression of a form of cytochrome P-450 in human ties in human liver microsomes. Drug Metab.
livers. Biochemistry 29, 4430^433. Dispos. 26, 1-4.
59. Lacroix, D., M. Sonnier, A. Moncion, G. Cheron, 71. Yuan, R., S. Madani, X.X. Wei, K. Reynolds, and
and T. Cresteil (1997). Expression of CYP3A in the S.M. Huang (2002). Evaluation of cytochrome P450
human liver: Evidence that the shift between probe substrates commonly used by the pharmaceu-
CYP3A7 and CYP3A4 occurs immediately after tical industry to study in vitro drug interactions.
birth. Eur J. Biochem. 247, 625-634. Drug Metab. Dispos. 30, 1311-1319.
60. Loi, CM. and R.E. Vestal (1988). Drug metaboHsm 72. Guengerich, FP (1988). Oxidation of 17a-
in the elderly. Pharmacol. Ther 36, 131-149. ethynylestradiol by human liver cytochrome P-450.
61. Durnas, C., CM. Loi, and B.J. Cusack (1990). Mol. Pharmacol. 33, 500-508.
Hepatic drug metabolism and aging. Clin. 73. Butler, M.A., M. Iwasaki, FP. Guengerich, and
Pharmacokinet. 19, 359-389. FF Kadlubar (1989). Human cytochrome P-450p^
62. George, J., K. Byth, and G.C Farrell (1995). Age (P-450j^2)' the phenacetin 0-deethylase, is prima-
but not gender selectively affects expression of indi- rily responsible for the hepatic 3-demethylation
vidual cytochrome P450 proteins in human liver. of caffeine and N-oxidation of carcinogenic ary-
Biochem. Pharmacol. 50, 727-730. lamines. Proc. Natl. Acad Sci. USA 86, 7696-7700.
63. Lu, A.Y.H., R.W. Wang, and J.H. Lin (2003). 74. Disderath, L.M. and FP Guengerich (1984).
Cytochrome P450 in vitro reaction phenotyping: Characterization of a human liver cytochrome P-450
A re-evaluation of approaches used for P450 involved in the oxidation of debrisoquine and other
Human P450s 467

drugs using antibodies raised to the analogous rat 85. Conney, A.H. (1982). Induction of microsomal
enzyme. Proc. Natl. Acad. Sci. USA 81, 7348-7352. enzymes by foreign chemicals and carcinogenesis
75. Soucek, P., M.V Martin, Y.-F. Ueng, and by polycyclic aromatic hydrocarbons: G. H, A.
F.P. Guengerich (1995). Identification of a common Clowes Memorial Lecture. Cancer Res. 42,
epitope near the conserved heme-binding region 4875-4917.
with polyclonal antibodies raised against 86. Sutter, T.R., K. Guzman, K.M. Dold, and
cytochrome P450 family 2 proteins. Biochemistry W.E Greenlee (1991). Targets for dioxin: Genes for
34, 16013-16021. plasminogen activator inhibitor-2 and interleukin-
76. Thomas, P.E., D. Koreniowski, D. Ryan, and \^. Science 254, 415^\S.
W. Levin (1979). Preparation of monospecific 87. Rivera, S.P, S.T. Saarikoski, and O. Hankinson
antibodies against two forms of rat liver cyto- (2002). Identification of a novel dioxin-inducible
chrome P-450 and quantitation of these antigens cytochrome P450. Mol. Pharmacol. 61, 255-259.
in microsomes. Arch. Biochem. Biophys. 192, 88. Shah, R.R., N.S. Gates, J.R. Idle, R.L. Smith, and
524-532. J.D.E Lockhart (1982). Impaired oxidation of
77. Venkatakrishnan, K., L.L. von Moltke, M.H. Court, debrisoquine in patients with perhexiline neuropa-
J.S. Harmatz, C.L. Crespi, and D.J. Greenblatt thy. Br Med. J. 284, 295-299.
(2000). Comparison between cytochrome P450 89. Steward, D.J., R.L. Haining, K.R. Henne, G. Davis,
(CYP) content and relative activity approaches to T.H. Rushmore, W.E Trager et al. (1997). Genetic
scaling from cDNA-expressed CYPs to human liver association between sensitivity to warfarin and
microsomes: Ratios of accessory proteins as sources expression of CYP2C9*3. Pharmacogenetics 1,
of discrepancies between the approaches. Drug 361-367.
Metab. Dispos. 28, 1493-1504. 90. Aithal, G.P, C.P Day, PJ. Kesteven, and A.K. Daly
78. Venkatakrishnan, K., L.L. von Moltke, and (1999). Association of polymorphisms in the
D.J. Greenblatt (2001). Application of the relative cytochrome P450 CYP2C9 with warfarin dose
activity factor approach in scaling from heterolo- requirement and risk of bleeding complications.
gously expressed cytochromes P450 to human liver Lancet 3,5^, 1X1-1\9.
microsomes: Studies on amitriptyline as a model 91. Daly, A.K., C.P Day, and G.P Aithal (2002).
substrate. J. Pharmacol. Exp. Then 291, 326-337. CYP2C9 polymorphism and warfarin dose require-
79. Venkatakrishnan, K., L.L. von Moltke, and ments. Brit. J. Clin. Pharmacol. 53, 408-409.
D.J. Greenblatt (2001). Human drug metabolism 92. Chiba, K., K. Kobayashi, K. Manabe, M. Tani,
and the cytochromes P450: Application and rele- T. Kamataki, and T. Ishizaki (1993). Oxidative
vance of in vitro models. J. Clin. Pharmacol. 41, metabolism of omeprazole in human liver micro-
1149-1179. somes: cosegregation with 5'-mephenytoin 4'-
80. Soars, M.G., H.V Gelboin, K.W. Krausz, and hydroxylation. J. Pharmacol. Exp. Ther 266,
R.J. Riley (2003). A comparison of relative abun- 52-59.
dance, activity factor and inhibitory monoclonal 93. Karam, W.G., J.A. Goldstein, J.M. Lasker, and
antibody approaches in the characterization of human B.I. Ghanayem (1996). Human CYP2C19 is a major
CYP enzymology. Br J. Clin. Pharmacol. 55, omeprazole 5-hydroxylase, as demonstrated with
175-181. recombinant cytochrome P450 enzymes. Drug
81. Schwab, M., M. Eichelbaum, and M.F. Fromm Metab. Dispos. 24, 1081-1087.
(2003). Genetic polymorphisms of the human mdr^ 94. Bolt, H.M., M. Bolt, and H. Kappus (1977).
drug transporter. Annu. Rev. Pharmacol. Toxicol. 43, Interaction of rifampicin treatment with pharmaco-
285-307. kinetics and metabolism of ethinyloestradiol in
82. Butler, M.A., N.R Lang, J.E Young, N.E. Caporaso, man. Acta Endocrinol. 85, 189-197.
P Vineis, R.B. Hayes et al. (1992). Determination of 95. Schwarz, U.I., B. Buschel, and W Kirch (2003).
CYP1A2 and NAT2 phenotypes in human popula- Unwanted pregnancy on self-medication with
tions by analysis of caffeine urinary metabolites. St. John's wort despite hormonal contraception.
Pharmacogenetics 2, 116-127. Br J. Clin. Pharmacol. 55, 112-113.
83. Ahsan, C.H., A.G. Renwick, B. Macklin, 96. Guengerich, EP (1999). Inhibition of drug
VE Challenor, D.G. Waller, and C.E George (1991). metabolizing enzymes: Molecular and biochemical
Ethnic differences in the pharmacokinetics of oral aspects. In T. E Woolf (ed.), Handbook of
nifedipine. Br J. Clin. Pharmacol. 31, 399^03. Drug Metabolism. Marcel Dekker, New York,
84. Kim, R.B., H. Yamazaki, M. Mimura, T. Shimada, pp. 203-227.
EP Guengerich, K. Chiba et al. (1996). 97. Bailey, D.G., J.D. Spence, B. Edgar, CD. Bayliff,
Chlorzoxazone 6-hydroxylation in Japanese and and J.M.O. Arnold (1990). Ethanol enhances the
Caucasians. In vitro and in vivo differences. J. hemodynamic effects of felodipine. Clin. Invest.
Pharmacol. Exp. Ther 279, 4-11. Med 12, 357-362.
468 F. Peter Guengerich

98. Edgar, B., D.G. Bailey, R. Bergstrand, G. Johnsson, P450 3A4-mediated interaction of diclofenac and
and L. Lurje (1990). Formulation dependent inter- quinidine. Drug Metab. Dispos. 28, 1043-1050.
action between felodipine and grapefruit juice. 112. Shapiro, A.B., K. Fox, R Lam, and V Ling (1999).
Clin. Pharmacol. Ther. 47, 181. Stimulation of P-glycoprotein-mediated drug
99. He, K., R. Iyer, R.N. Hayes, M.W. Sinz, T.F. Woolf, transport by prazosin and progesterone. Eur J.
and RF. Hollenberg (1998). Inactivation of Biochem. 259, 841-850.
cytochrome P450 3A4 by bergamottin, a compo- 113. Mueller, G.C. and J.A. Miller (1948). The metabo-
nent of grapefruit juice. Chem. Res. Toxicol. 11, lism of 4-dimethylaminoazobenzene by rat liver
252-259. homogenates. J. Biol. Chem. 176, 535-544.
100. Schmiedlin-Ren, R, D.J. Edwards, 114. Jollow, D.J., J.R. Mitchell, W.Z. Potter,
M.E. Fitzsimmons, K. He, K.S. Lown, RM. Woster D.C. Davis, J.R. Gillette, and B.B. Brodie (1973).
et al. (1997). Mechanisms of enhanced oral Acetaminophen-induced hepatic necrosis. II. Role
availability of CYP3A4 substrates by grapefruit of covalent binding in vivo. J. Pharmacol. Exp.
constituents. Drug Metab. Dispos. 25, 1228-1233. Ther 187, 195-202.
101. Reimers, D. and A. Jezek (1971). Rifampicin und 115. Neal, R.A. and J. Halpert (1982). Toxicology of
andere antituberkulotika bei gleichzeitiger oraler thiono-sulfur compounds. Annu. Rev. Pharmacol.
kontrzeption. Przx. Pneumolonology 25, 255-262. Toxicol. 22,321-339.
102. Nocke-Finck, L. H. Brewer, and D. Reimers 116. Wing, K.D., A.H. Glickman, and J.E. Casida
(1973). Wirkung van rifampicin auf den menstra- (1983). Oxidative bioactivation of S-alkyl phospho-
tionszylkus und die ostrogenausscheidung bei ein- rothiolate pesticides: Stereospecificity of profeno-
nahme oraler kontrazeptive. Deutsch. Med. fos insecticide activation. Science 219, 63-65.
Wochenschrifften 9H, 1521-1523. 117. Nebert, D.W. (1989). The Ah locus: Genetic differ-
103. Kivisto, K.T., RJ. Neuvonen, and U. Klotz (1994). ences in toxicity, cancer, mutation, and birth
Inhibition of terfenadine metabolism: Pharma- defects. Crit. Rev Toxicol. 20, 153-174.
cokinetic and pharmacodynamic consequences. 118. Lee, S.S.T., J.TM. Buters, T. Pineau, R Fernandez-
Clin. Pharmacokinet. 21, 1-5. Salguero, and F.J. Gonzalez (1996). Role of
104. Thompson, D. and G. Oster (1996). Use of terfe- CYP2E1 in the hepatotoxicity of acetaminophen.
nadine and contraindicated drugs. J. Am. Med. J. Biol. Chem. 271, 12063-12067.
Assoc, lis, 1339-1341. 119. Buters, J.T.M., S. Sakai, T Richter, T. Pineau, D.L.
105. Fischbach, T. and W. Lenk (1985). Additional routes Alexander, 0. Savas et al. (1999). Cytochrome
in the metabolism of phenacetin. Xenobiotica 15, P450 CYPIBI determines susceptibility to 7,12-
149-164. dimethylbenz[fl]anthracene-induced lymphomas.
106. Guengerich, F.R, D. Miiller-Enoch, and LA. Blair Proc. Natl. Acad. Sci. USA 96, 1977-1982.
(1986). Oxidation of quinidine by human liver 120. Nebert, D.W. and H.V. Gelboin (1968).
cytochrome P-450. Mol. Pharmacol. 30, 287-295. Substrate-inducible microsomal arylhydroxylase
107. Halpert, J.R. and FR Guengerich (1997). Enzyme in mammalian cell culture: Assay and proper-
inhibition and stimulation. In F P. Guengerich ties of induced enzyme. J. Biol. Chem. 243,
(ed.). Biotransformation, Vol. 3, Comprehen- 6242-6249.
sive Toxicology. Elsevier Science Ltd., Oxford, 121. Kellerman, G., M. Luyten-Kellerman, and
pp. 21-35. C.R. Shaw (1973). Genetic variation of aryl hydro-
108. Witherow, L.E. and J.B. Houston (1999). Sig- carbon hydroxylase in human lymphocytes. Am. J.
moidal kinetics of CYP3A substrates: An approach Hum. Genet. 25,327-331.
for scaling dextromethorphan metabolism in 122. Kellerman, G., C.R. Shaw, and M. Luyten-
hepatic microsomes and isolated hepatocytes to Kellerman (1973). Aryl hydrocarbon hydroxylase
predict in vivo clearance in rat. J. Pharmacol. Exp. inducibility and bronchogenic carcinoma. A'; Engl.
Then 290, 58-65. J. Med 298, 934-937.
109. Lasker, J.M., M.-T. Huang, and A.H. Conney 123. Paigen, B., E. Ward, A. Reilly, L. Houten,
(1982). In vivo activation of zoxazolamine metab- H.L. Gurtoo, J. Minowada et al (1981). Seasonal
olism by flavone. Science 216, 1419-1421. variation of aryl hydrocarbon hydroxylase activity
110. Lee, C.A., J.H. Lillibridge, S.D. Nelson, and in human lymphocytes. Cancer Res. 41,2757-2761.
J.T. Slattery (1996). Effects of caffeine and theo- 124. Kouri, R.E., C.E. McKinney, D.J. Slomiany,
phylline on acetaminophen pharmacokinetics: D.R. Snodgrass, N.P. Wray, and T.L. McLemore
P450 inhibition and activation. J. Pharmacol. Exp. (1982). Positive correlation between high aryl
Ther 211,2^7-291. hydrocarbon hydroxylase activity and primary
111. Ngui, J.S., W. Tang, R.A. Stearns, M.G. Shou, lung cancer as analyzed in cryopre served lympho-
R.R. Miller, Y. Zhang et al. (2000). Cytochrome cytes. Cancer Res. 42, 5030-5037.
Human P450s 469

125. Chang, C , D.R. Smith, VS. Prasad, C.L. Sidman, hydrocarbon hydroxylase represents CYPIBI, and
D.W. Nebert, and A. Puga (1993). Ten nucleotide not CYPlAl, in human freshly isolated white cells:
differences, five of which cause amino acid Trimodal distribution of Japaneses population
changes, are associated with the Ah receptor locus according to induction of CYPIBI mRNA by envi-
polymorphism of C57BL/6 and DBA/2 mice. ronmental dioxins. Cancer Epidemiol. Biomarkers
Pharmacogenetics 3, 312-321. Prev. 12,219-222.
126. Dolwick, K.M., J.V Schmidt, L.A. Carver, 136. Guengerich, F.P. (1998). The environmental genome
H.I. Swanson, and C.A. Bradfield (1993). Cloning project: Functional analysis of polymorphisms.
and expression of a human Ah receptor cDNA. Environ. Health Perspect. 106, 365-368.
Mol. Pharmacol 44, 911-917. 137. Ayesh, R., J.R. Idle, J.C. Ritchie, M.J. Crothers,
127. Fujii-Kuriyama, Y., M. Ema, J. Mimura, and M.R. Hetzel (1984). Metabolic oxidation phe-
N. Matsushita, and K. Sogawa (1995). Polymorphic notypes as markers for susceptibility to lung can-
forms of the Ah receptor and induction of the cer. A^a^re 312, 169-170.
CYPlAl gene. Pharmacogenetics 5, S149-S153. 138. d'Errico, A., E. Taioli, X. Chen, and P. Vineis
128. Kawajiri, K., J. Watanabe, H. Eguchi, K. Nakachi, (1996). Genetic metabolic polymorphisms and
C. Kiyohara, and S. Hayashi (1995). Polymorphisms the risk of cancer: A review of the literature.
of human Ah receptor gene are not involved in lung BioMarkers 1, 149-173.
cancer. Pharmacogenetics 5, 151-158. 139. Shimada, T and FR Guengerich (1991). Activa-
129. Hayashi, S., J. Watanabe, K. Nakachi, and tion of amino-a-carboline, 2-amino-l-methyl-
K. Kawajiri (1991). Genetic linkage of lung 6-phenylimidazo[4,5-^]pyridine, and a copper
cancer-associated Msp\ polymorphisms with phthalo-cyanine cellulose extract of cigarette
amino acid replacement in the heme binding smoke condensate by cytochrome P-450 enzymes
region of the human cytochrome P450IA1 gene. in rat and human liver microsomes. Cancer Res.
J. Biochem. (Tokyo) 110, 407-^11. 51,5284-5291.
130. Tefre, T., D. Ryberg, A. Haugen, D.W. Nebert, 140. Vineis, P. (2002). The relationship between poly-
V Skaug, A. Brogger et al (1991). Human CYPlAl morphisms of xenobiotic metabolizing enzymes and
(cytochrome Pj450) gene: Lack of association susceptibility to cancer. Toxicology 181, 457^62.
between the Msp I restriction fragment length poly- 141. Stoilov, I., A.N. Akarsu, I. Alozie, A. Child,
morphism and incidence of lung cancer in a M. Barsoum-Homsy, M.E. TuracH et al. (1998).
Norwegian population. Pharmacogenetics 1, Sequence analysis and homology modeling suggest
20-25. that primary congenital glaucoma on 2p21 results
131. Hirvonen, A., K. Husgafvel-Pursiainen, from mutations disrupting either the hinge region
A. Karjalainen, S. Anttila, and H. Vainio (1992). or the conserved core structures of cytochrome
Point-mutational MspX and Ile-Val polymorphisms P4501B1. ylm. J. Hum. Genet. 62, 573-584.
closely linked in the CYPlAl gene: Lack of asso- 142. Shimada, T, J. Watanabe, F.P. Guengerich,
ciation with susceptibility to lung cancer in a K. Inoue, and E.M.J. Gillam (2001). Specificity
Finnish study population. Cancer Epidemiol. of 17p-oestradiol and benzo[a]pyrene oxidation
Biomarkers Prev. 1, 485^89. by polymorphic human cytochrome P450 IBl
132. Wedlund, PJ., S. Kimura, EL Gonzalez, and variants substituted at residues 48, 119, and 432.
D.W. Nebert (1994). I462V mutation in the human Xenobiotica 31, 163-176.
CYPlAl gene: Lack of correlation with either the 143. Watanabe, J., T. Shimada, E.M.J. Gillam, T. Ikuta,
Msp I 1.9kb (M2) allele or CYPlAl inducibility K. Suemasu, Y Higashi et al. (2000). Association
in a three-generation family of East Mediterranean of CYPIBI genetic polymorphism with incidence
descent. Pharmacogenetics 4, 21-26. to breast and lung cancer. Pharmacogenetics 10,
133. Zhang, Z.-Y, M.J. Fasco, L. Huang, F.P. Guengerich, 25-33.
and L.S. Kaminsky (1996). Characterization of puri- 144. Zheng, W, D.W. Xie, F Jin, J.R. Cheng, Q. Dai,
fied recombinant human CYP 1 Al-Ile462 and NdX^^^\ WQ. Wen et al. (2000). Genetic polymorphism of
Assessment of a role for the rare allele in carcino- cytochrome P450-1B1 and risk of breast cancer.
genesis. Cancer Res. 56, 3926-3933. Cancer Epidemiol. Biomarkers Prev. 9, 147-150.
134. Persson, I., I. Johansson, and M. Ingelman-Sundberg 145. Lang, N.R, M.A. Butler, J. Massengill, M. Lawson,
(1997). In vitro kinetics of two human CYPlAl R.C. Stotts, M. Maurer-Jensen etal. (1994). Rapid
variant enzymes suggested to be associated with metabolic phenotypes for acetyltransferase and
interindividual differences in cancer susceptibility. cytochrome P4501A2 and putative exposure to
Biochem. Biophys. Res. Commun. 231, 227-230. food-borne heterocyclic amines increase the risk
135. Toide, K., H. Yamazaki, R. Nagashima, K. Itoh, for colorectal cancer or polyps. Cancer Epidemiol.
S. Iwano, Y Takahashi et al. (2003). Aryl Biomarkers Prev. 3, 675-682.
470 F. Peter Guengerich

146. Shimada, T., M. Iwasaki, M.V Martin, and benzo[^]fluoranthene in human hepatocytes:
F.P. Guengerich (1989). Human liver microsomal CYPlAl or CYP1A2?. Arch. Biochem. Biophys.
cytochrome P-450 enzymes involved in the bioac- 389, 130-134.
tivation of procarcinogens detected by umu gene 159. Fagan, J.B., J.V Pastewka, S.R. Chalberg,
response in Salmonella typhimurium TA1535/ E. Gozukara, F.P. Guengerich, and H.V Gelboin
pSK1002. Cancer Res. 49, 3218-3228. (1986). Noncoordinate regulation of the mRNAs
147. Ariyoshi, N., M. Miyamoto, Y. Umetsu, encoding cytochromes P-450g and P-
H. Kunitoh, H. Dosaka-Akita, Y. Sawamura et al. 450,, , Arch. Biochem. Biophys. 244,261-272.
(2002). Genetic polymorphism of CYP2A6 gene 160. Kim, J.H., M.E. Sherman, EC. Curreiro, FP
and tobacco-induced lung cancer risk in male Guengerich, PT. Strickland, and T.R. Sutter (2004,
smokers. Cancer Epidemiol. Biomarkers Prev. 11, in press). Expression of cytochromes P450 lAl and
890-894. IBl in human lung from smokers, non-smokers,
148. Pianezza, M.L., E.M. Sellers, and R.F. Tyndale and ex-smokers. Toxicol. Appl. Pharmacol.
(1998). Nicotine metabolism defect reduces smok- 161. Prough, R.A., Z. Sipal, and S.W. Jakobsson (1977).
ing. Nature 393, 750. Metabolism of benzo(a)pyrene by human lung
149. Kim, R.B., D. O'Shea, and G.R. Wilkinson (1994). microsomal fractions. Life Sci. 21, 1629-1636.
Relationship in healthy subjects between CYP2E1 162. Fujino, T, K. Gottlieb, D.K. Manchester, S.S. Park,
genetic polymorphism and the 6-hydroxylation of D. West, H.L. Gurtoo et al. (1984). Monoclonal anti-
chlorzoxazone: A putative measure of CYP2E1 body phenotyping of interindividual differences in
activity. Pharmacogenetics 4, 162-165. cytochrome P-450-dependent reactions of single and
150. loannides, C. and D.V Parke (1993). Induction of twin human placenta. Cancer Res. 44, 3916-3923.
cytochrome P4501 as an indicator of potential 163. Robie-Suh, K., R. Robinson, H.V Gelboin, and
chemical carcinogenesis. Drug Metab. Rev. 25, F.P. Guengerich (1980). Aryl hydrocarbon
485-501. hydroxylase is inhibited by antibody to rat liver
151. Rice, J.M., B.A. Diwan, J.M. Ward, R.W. Nims, and cytochrome P-450. Science 208, 1031-1033.
R.A. Lubet (1992). Phenobarbital and related com- 164. Shimada, T, H. Yamazaki, M. Mimura,
pounds: Approaches to interspecies extrapolation. N. Wakamiya, F.P. Guengerich, and Y Inui (1996).
In Relevance ofAnimal Studies to the Evaluation of Characterization of microsomal cytochrome P450
Human Cancer Risk. Wiley-Liss, Inc., pp. 231-249. enzymes involved in the oxidation of xenobiotic
152. Olsen, J.H., J.D. Boice Jr., J.PA. Jensen and chemicals in human fetal livers and adult lungs.
J.F. Fraumeni Jr. (1989). Cancer among epileptic Drug Metab. Dispos. 24, 515-522.
patients exposed to anticonvulsant drugs. J. Natl. 165. McLemore, T.L., S. Adelberg, M.C. Liu,
Cancer Inst. 81,803-808. N.A. McMahon, S.J. Yu, W.C. Hubbard et al
153. Kluwe, WM. (1994). The relevance of hepatic per- (1990). Expression of CYPlAl gene in patients
oxisome proliferation in rats to assessment of with lung cancer: Evidence for cigarette smoke-
human carcinogenic risk for pharmaceuticals. induced gene expression in normal lung tissue and
Regul. Toxicol. Pharmacol. 20, 170-186. for altered gene regulation in primary pulmonary
154. Thomas, R.S., D.R. Rank, S.G. Penn, carcinomas. .( A^a//. Cancer Inst. 82, 1333-1339.
G.M. Zastrow, K.R. Hayes, K. Pande et al. (2001). 166. Anttila, S., X.D. Lei, E. Elovaara, A. Karjalainen,
Identification of toxicologically predictive gene W.M. Sun, H. Vainio et al. (2000). An uncommon
sets using cDNA microarrays. Mol. Pharmacol. phenotype of poor inducibility of CYPlAl in
60, 1189-1194. human lung is not ascribable to polymorphisms in
155. Omiecinski, C.J., C.A. Redlich, and P Costa the AHR, ARNT, or CYPlAl genes. Pharmaco-
(1990). Induction and developmental expression of genetics 10, 741-751.
cytochrome P450IA1 messenger RNA in rat and 167. Anttila, S., P. Tuominen, A. Hirvonen, M. Nurminen,
human tissues: Detection by the polymerase chain A. Karjalainen, O. Hmkmsonetal. (2001). CYPlAl
reaction. Cancer Res. 50, 4315^321. levels in lung tissue of tobacco smokers and poly-
156. Kitada, M., M. Taneda, K. Itahashi, andT. Kamataki morphisms of CYPlAl and aromatic hydrocarbon
(1991). Four forms of cytochrome P-450 in human receptor. Pharmacogenetics 11, 501-509.
fetal liver: Purification and their capacity to activate 168. Bradfield, C.A. (2004, in press). Induction of P450
promutagens. Jpn. J. Cancer Res. 82, 426-432. enzymes: Receptors. In P. R. Ortiz De Montellano
157. Kitada, M. and T. Kamataki (1994). Cytochrome (ed.), Cytochrome P450: Structure, Mechanism,
P450 in human fetal liver: Significance and fetal- and Biochemistry, Plenum, New York (Chap. 8 of
specific expression. Drug Metab. Rev. 26, 305-323. this monograph).
158. Liu, N., Q.Y Zhang, D. Vakharia, D. Dunbar, and 169. Lucier, G.W, K.G. Nelson, R.B. Everson,
L.S. Kaminsky (2001). Induction of CYPIA by T.K. Wong, R.M. Philpot, T. Tiernan et al (1987).
Human P450s 471

Placental markers of human exposure to polychlo- 181. Shimada, T, H. Yamazaki, M. Foroozesch,


rinated biphenyls and polychlorinated dibenzofu- N.E. Hopkins, W.L. Alworth, and F.R Guengerich
rans. Environ. Health Perspect. 76, 79-87. (1998). Selectivity of polycyclic inhibitors for
170. Diaz, D., I. Fabre, M. Daujat, B. Saintaubert, human cytochromes P450 lAl, 1A2, and IBl.
P. Bories, H. Michel et al. (1990). Omeprazole is Chem. Res. Toxicol. 11, 1048-1056.
an aryl hydrocarbon-like inducer of human hepatic 182. McManus, M.E., W.M. Burgess, M.E. Veronese,
cytochrome-P450. Gastroenterology 99, 12>1-1A1. A. Huggett, L.C. Quattrochi, and R.H. Tukey
171. Yun, C.-H., T. Shimada, and F.R Guengerich (1990). Metabolism of 2-acetylaminofluorene and
(1992). Roles of human liver cytochrome P-4502C benzo[(2]pyrene and activation of food-derived het-
and 3A enzymes in the 3-hydroxylation of erocyclic amine mutagens by human cytochromes
benzo[«]pyrene. Cancer Res. 52, 1868-1874. P-450. Cancer Res. 50, 3367-3376.
172. Prough, R.A., M.D. Burke, and R.T. Mayer (1978). 183. Mercurio, M.G., S.J. Shiff, R.A. Galbraith, and
Direct fluorometric methods for measuring mixed- S. Sassa (1995). Expression of cytochrome P450
function oxidase activity. Meth. Enzymol. 52, mRNAs in the colon and the rectum in normal
372-377. human subjects. Biochem. Biophys. Res. Commun.
173. Guo, Z., E.M.J. Gillam, S. Ohmori, R.H. Tukey, 210, 350-355.
and P.P. Guengerich (1994). Expression of modi- 184. Pantuck, E.J., K.-C. Hsiao, A. Maggio,
fied human cytochrome P450 lAl in Escherichia K. Nakamura, R. Kuntzman, and A.H. Conney
coli: Effects of 5' substitution, stabilization, purifi- (1974). Effect of cigarette smoking on phenacetin
cation, spectral characterization, and catalytic metabolism. Clin. Pharmacol. Then 15, 9-17.
properties. Arch. Biochem. Biophys. 312, 436-446. 185. Vesell, E.S. and J.G. Page (1968). Genetic control of
174. Shou, M., K.R. Korzekwa, C.L. Crespi, drug levels in man: Antipyrine. Science 161, 72-73.
F.J. Gonzalez, and H.V Gelboin (1994). The role of 186. Rasmussen, B.B., T.H. Brix, K.O. Kyvik, and
12 cDNA-expressed human, rodent, and rabbit K. Brosen (2002). The interindividual differences
cytochromes P450 in the metabolism of in the 3-demethylation of caffeine alias CYP1A2
benzo[a]pyrene and benzo[fl]pyrene trans-l,S- is determined by both genetic and environmental
dihydrodiol. M?/. Carcinogen. 10, 159-168. factors. Pharmacogenetics 12, 473-478.
175. Bauer, E., Z. Guo, Y.-E Ueng, L.C. Bell, and 186a.Zhou, H., RD. Josephy, D. Kim, and ER
F.R Guengerich (1995). Oxidation of benzo[a]- Guengerich (2004). Functional characterization of
pyrene by recombinant human cytochrome P450 four allelic variants of human cytochrome P450
enzymes. Chem. Res. Toxicol. 8, 136-142. 1A2. Arch. Bioch. Biophys. 422, 23-30.
176. Shou, M., K.W. Krausz, F.J. Gonzalez, and 187. Kalow, W. and B.K. Tang (1993). The use of
H.V Gelboin (1996). Metabolic activation of caffeine for enzyme assays: A critical appraisal.
the potent carcinogen dibenzo[flf,/?]anthracene by Clin. Pharmacol. Ther 53, 503-514.
cDNA-expressed human cytochromes P450. Arch. 188. Quattrochi, L.C, T. Vu, and R.H. Tukey (1994).
Biochem. Biophys. 328, 201-207. The human CYP1A2 gene and induction by
177. Shou, M., K.W. Krausz, EJ. Gonzalez, and 3-methylcholanthrene: A region of DNA that sup-
H.V Gelboin (1996). Metabolic activation of the ports Ah-receptor binding and promoter-specific
potent carcinogen dibenzo[fl,/]pyrene by human induction. J. Biol. Chem. 269, 6949-6954.
recombinant cytochromes P450, lung and liver 189. Kondraganti, S.R., W.W. Jiang, and B. Moorthy
microsomes. Carcinogenesis 17, 2429-2433. (2002). Differential regulation of expression of
178. Shimada, T, C.L. Hayes, H. Yamazaki, S. Amin, hepatic and pulmonary cytochrome P4501A
S.S. Hecht, ER Guengerich et al. (1996). Activation enzymes by 3-methylcholanthrene in mice lacking
of chemically diverse procarcinogens by human the CYP1A2 gene. J. Pharmacol. Exp. Ther 303,
cytochrome P450 IBl. Cancer Res. 56,2979-2984. 945-951.
179. Balani, S.K., H.J.C. Yeh, D.E. Ryan, RE. Thomas, 190. Fisher, G.J., H. Fukushima, and J.L. Gaylor
W. Levin, and D.M. Jerina (1985). Absolute con- (1981). Isolation, purification, and properties of a
figuration of the 5,6-oxide formed from 7,12- unique form of cytochrome P-450 in microsomes
dimethylbenz[a]anthracene by cytochrome P450c. of isosafrole-treated rats. J. Biol. Chem. 256,
Biochem. Biophys. Res. Commun. 130, 610-616. 4388-4394.
180. Lewis, D.EV, B.G. Lake, S.G. George, M. Dickins, 191. Vistisen, K., H.E. Poulsen, and S. Loft (1992).
RJ. Eddershaw, M.H. Tarbit et al. (1999). Foreign compound metabolism capacity in man
Molecular modeling of CYP family enzymes measured from metabolites of dietary caffeine.
CYPlAl, CYP1A2, CYP1A6, and CYPIBI based Carcinogenesis 13, 1561-1568.
on sequence homology with CYP 102. Toxicology 192. Rost, K.L., H. Brosicke, J. Brockmoller,
139, 53-79. M. Scheffler, H. Helg, and L Roots (1992).
472 F. Peter Guengerich

Increase of cytochrome P450IA2 activity by involved in theophylline 8-hydroxylation.


omeprazole: Evidence by the ^^C-[A^-3-methyl]- Biochem. Pharmacol. 50, 205-211.
caffeine breath test in poor and extensive metabo- 202. Yamazaki, H., Y. Oda, Y Funae, S. Imaoka, Y Inui,
Hzers of *S-mephenytoin. Clin. Pharmacol. Then F.P. Guengerich et al. (1992). Participation of rat
52, 170-180. liver cytochrome P450 2E1 in the activation of
193. Fisher, C.W., D.L. Caudle, C. Martin-Wixtrom, A^-nitrosodimethylamine and A^-diethylnitrosamine
L.C. Quattrochi, R.H. Tukey, M.R. Waterman et al to products genotoxic in Salmonella typhimurium
(1992). High-level expression of functional NM2009. Carcinogenesis 13, 979-985.
cytochrome P450 1A2 in Escherichia coli, 203. Yamazaki, H., RM. Shaw, ER Guengerich, and
FASEB J. 6,759-164. T Shimada (1998). Roles of cytochromes P450
194. Sandhu, P., Z. Guo, T. Baba, M.V. Martin, 1A2 and 3A4 in the oxidation of estradiol and
R.H. Tukey, and F.P. Guengerich (1994). Expres- estrone in human liver microsomes. Chem. Res.
sion of modified human cytochrome P450 1A2 in Toxicol. 11,659-665.
Escherichia coli: Stabilization, purification, spec- 204. Michnovicz, J.J., R.J. Hershcopr, H. Naganuma,
tral characterization, and catalytic activities of the H.L. Bradlow, and J. Fishman (1986). Increased
enzyme. ^rcA. Biochem. Biophys. 309, 168-177. 2-hydroxylation of estradiol as a possible mecha-
195. Patten, C.J., RE. Thomas, R.L. Guy, M. Lee, nism for the anti-estrogenic effect of cigarette
RJ. Gonzalez, ER Guengerich et al. (1993). smoking. New Engl. J. Med. 315, 1305-1309.
Cytochrome P450 enzymes involved in acetamin- 205. Bradlow, H.L., R.J. Hershcopf, C.R Martucci, and
ophen activation by rat and human liver micro- J. Fishman (1985). Estradiol 16a-hydroxylation in
somes and their kinetics. Chem. Res. Toxicol. 6, the mouse correlates with mammary tumor inci-
511-518. dence and presence of murine mammary tumor
196. Engel, G., U. Hofmann, H. Heidemann, J. Cosme, virus: A possible model for the hormonal etiology
and M. Eichelbaum (1996). Antipyrine as a probe of breast cancer in humans. Proc. Natl. Acad. Sci.
for human oxidative drug metabolism: Identifica- USA 82, 6295-6299.
tion of the cytochrome P450 enzymes catalyzing 206. Hiroya, K., M. Ishigooka, T. Shimizu, and
4-hydroxyantipyrine, 3-hydroxymethylantipyrine, M. Hatano (1992). Role of Glu318 and Thr319
and norantipyrine formation. Clin. Pharmacol. in the catalytic function of cytochrome P450j
Then 59, 613-623. (P4501A2): Effects of mutations on the methanol
197. Yamazaki, H., Z. Guo, M. Persmark, M. Mimura, hydroxylation. FASEB J. 6, 749-751.
EJ. Gonzalez, C. Sugahara et al. (1994). Bufuralol 207. Ishigooka, M., T. Shimizu, K. Hiroya, and
hydroxylation by cytochrome P450 2D6 and 1A2 M. Hatano (1992). Role of Glu318 at the putative
enzymes in human liver microsomes. Mol. distal site in the catalytic function of cytochrome
Pharmacol. 46, 568-577. P450j. Biochemistry 3\, 1528-1531.
198. Berthou, E, V Carriere, D. Ratanasavanh, 208. Shimizu, T, A.J.M. Sadeque, G.N. Sadeque,
T Goasdufif, E Morel, J.C. Gautier et al. (1993). M. Hatano, and Y Fujii-Kuriyama (1991). Ligand
On the specificity of chlorzoxazone as drug binding studies of engineered cytochrome P-450j
probe of cytochrome P4502E1. In Abstracts, wild type, peroximal mutants, and distal mutants.
5th European ISSXMeeting, Vol. 3. p. 116, 26-29 Biochemistry 30, 1490-1496.
September, Tours, France. 209. Fuhr, U, G. Strobl, E Manaut, E.M. Anders,
199. Woolf, TE, W.E Pool, M. Kukan, S. Bezek, E Sorgel, E. Lopez-de-Brinas et al. (1993).
K. Kunze, and W.E Trager (1993). Characterization Quinolone antibacterial agents: Relationship
of tacrine metabolism and bioactivation using between structure and in vitro inhibition of the
heterologous expression systems and inhibition human cytochrome P450 isoform CYP1A2.
studies: Evidence for CYP1A2 involvement. Mol. Pharmacol. 43, 191-199.
In Abstracts, 5th North American ISSX Meeting, 210. Lozano, J.J., E. L6pez-de-Briiias, N.B. Centeno,
Vol. 4, p. 139, 17-21 October, Tucson, AZ. E Sanz, and R. Guigo (1997). Three-dimensional
200. Benoit, G.G., C.E Naud, M.A. Simard, and modelling of human cytochrome P450 1A2 and
A.L. Astier (1997). Noninterference of cytochrome its interaction with caffeine and MelQ. J. Comput.
P4501A2 in the cytotoxicity of tacrine using genet- Aided Mol. Des. 11, 395^08.
ically engineered V79 Chinese hamster cells for 211. Parikh, A., P.D. Josephy, and F.P. Guengerich
stable expression of the human or rat isoform and (1999). Selection and characterization of human
two human hepatocyte cell lines. Biochem. cytochrome P450 1A2 mutants with altered cat-
Pharmacol. 53, 423-427. alytic properties. Biochemistry 38, 5283-5289.
201. Zhang, Z.Y. and L.S. Kaminsky (1995). 212. Yun, C.-H., G.R Miller, and EP Guengerich
Characterization of human cytochromes P450 (2000). Rate-determining steps in phenacetin
Human P450s 473

oxidation by human cytochrome P450 1A2 and mutations in cytochrome P4501B1 (CYPIBI) as
selected mutants. Biochemistry 39, 11319-11329. the principal cause of primary congenital glaucoma
213. Yun, C.-H., G.P. Miller, and F.P. Guengerich (2001). (Buphthalmos) in families linked to the GLC3A
Oxidations of /7-alkoxyacylanilides by human locus on chromosome 2p21. Hum. Mol. Genet. 6,
cytochrome P450 1A2: Structure-activity relation- 641-647.
ships and simulation of rate constants of individual 225. Shimada, T, E.M.J. Gillam, T.R. Sutter,
steps in catalysis. Biochemistry 40, 4521-4530. PT. Strickland, F.P. Guengerich, and H. Yamazaki
214. Miller, G.P and F.P Guengerich (2001). Binding (1997). Roles of recombinant human cytochrome
and oxidation of alkyl 4-nitrophenyl ethers by P450 IBl in the oxidation of xenobiotic chemicals.
rabbit cytochrome P450 1A2: Evidence for two Drug Metab. Dispos. 25, 617-622.
binding sites. Biochemistry 40, 7262-7272. 226. Shimada, T, Y Oda, E.M.J. Gillam,
215. Racha, J.K., A.E. Rettie, and K.L. Kunze (1998). F.P. Guengerich, and K. Inoue (2001). Metabolic
Mechanism-based inactivation of human cyto- activation of polycyclic aromatic hydrocarbons
chrome P450 1A2 by fiirafylline: Detection of a and their dihydrodiol derivatives and other pro-
1:1 adduct to protein and evidence for the forma- carcinogens by cytochrome P450 lAl and IBl
tion of a novel imidazomethide intermediate. allelic variants and other human cytochrome P450
Biochemistry 37, 7407-7419. enzymes in Salmonella typhimurium NM2009.
216. Voorman, R. and S.D. Aust (1987). Specific bind- Drug Metab. Dispos. 29, 1176-1182.
ing of polyhalogenated aromatic hydrocarbon 227. Yamazaki, H., N. Hatanaka, R. Kizu, K. Hayakawa,
inducers of cytochrome P-450j to the cytochrome N. Shimada, FP Guengerich et al. (2000).
and inhibition of its estradiol 2-hydroxylase activ- Bioactivation of diesel exhaust particle extracts
ity. Toxicol. Appl. Pharmacol 90, 69-78. and their major nitrated polycyclic aromatic hydro-
217. Sesardic, D., A. Boobis, B. Murray, S. Murray, carbon components, 1-nitropyrene and dinitropy-
J. Segura, R. De La Torre et al (1990). FurafyUine is renes, by human cytochrome P450s lAl, 1A2, and
a potent and selective inhibitor of cytochrome P450 IBl. Chem. Res. Toxicol. 411, 129-138.
1A2 in man. Br. J. Clin. Pharmacol. 29, 651-663. 228. Hayes, C.L., D.C. Spink, B.C. Spink, J.Q. Cao,
218. Kappas, A., A.P Alvares, K.E. Anderson, N.J. Walker, and TR. Sutter (1996). 17p-Estradiol
E.J. Pantuck, C.B. Pantuck, R. Chang et al. (1978). hydroxylation catalyzed by human cytochrome P450
Effect of charcoal-broiled beef on antipyrine and IBl. Proc. Natl. Acad Sci. USA 93, 9776-9781.
theophylline metabolism. Clin. Pharmacol. Ther 229. Shimada, T, J. Watanabe, K. Kawajiri, T.R. Sutter,
23, 445-450. F.P Guengerich, E.M.J. Gillam et al. (1999). Cata-
219. Feldman, C.H., VE. Hutchinson, C.E. Pippenger, lytic properties of polymorphic human cytochrome
T.A. Blemenfeld, B.R. Feldman, and W.J. Davis P450 IBl variants. Carcinogenesis 20, 1607-1614.
(1980). Effect of dietary protein and carbohydrate 230. Shimada, T, E.M.J. Gillam, Y. Oda, F. Tsumura,
on theophylline metabolism in children. Pediatrics T.R. Sutter, RP Guengerich et al. (1999). Meta-
66, 956-962. bolism of benzo[fl]pyrene to ^ra«5-7,8-dihydroxy-
220. Otto, S., C. Marcus, C. Pidgeon, and C. Jefcoate 7,8-dihydrobenzo[fl]pyrene by recombinant human
(1991). A novel adrenocorticotropin-inducible cytochrome P450 IBl and purified liver epoxide
cytochrome P450 from rat adrenal microsomes hydrolase. Chem. Res. Toxicol. 12, 623-629.
catalyzes polycyclic aromatic hydrocarbon metab- 231. Savas, U., C.P Carstens, and C.R. Jefcoate (1997).
olism. Endocrinology 129, 970-982. Recombinant mouse CYPIBI expressed in
221. Murray, B.P and M.A. Correia (2001). Ubiquitin- Escherichia coli exhibits selective binding by poly-
dependent 26S proteasomal pathway: A role cyclic hydrocarbons and metabolism which paral-
in the degradation of native human liver CYP3 A4 lels C3H10T1/2 cell microsomes, but differs from
expressed in Saccharomyces cerevisiael Arch. human recombinant CYPIBI. Arch. Biochem.
Biochem. Biophys. 393, 106-116. Biophys. 347, 181-192.
222. Murray, G.I., M.C. Taylor, M.C. McFadyen, 232. Hanna, I.H., S. Dawling, N. Roodi, F.P. Guengerich,
J.A. McKay, W.F Greenlee, M.D. Burke et al. and R Pari (2000). Cytochrome P450 IBl (CYPIBI)
(1997). Tumor-specific expression of cytochrome pharmacogenetics: Association of polymorphisms
P450 CYPIBI. Cancer Res. 57, 3026-3031. with functional differences in estrogen hydroxyla-
223. Chang, TK.H., J. Chen, V Pillay, J.-Y. Ho, and tion activity. Cancer Res. 60, 3440-3444.
S.M. Bandiera (2003). Real-time polymerase chain 233. Shou, M., K.R. Korzekwa, E.N. Brooks,
reaction analysis of CYPIBI gene expression in K.W Krausz, F.J. Gonzalez, and H.V Gelboin
human liver. Toxicol. Sci. 71, 11-19. (1997). Role of human hepatic cytochrome P450
224. Stoilov, I., A.N. Akarsu, and M. Sarfarazi 1A2 and 3A4 in the metabolic activation of
(1997). Identification of three different truncating estrone. Carcinogenesis 18, 207-214.
474 F. Peter Guengerich

234. Bolton, XL., E. Pisha, F. Zhang, and S. Qiu (1998). expression and relevance to nitrosamine metabo-
Role of quinoids in estrogen carcinogenesis. lism. Carcinogenesis 23, 611-616.
Chem. Res. Toxicol. 11, 1113-1127. 246. Raunio, H., R. Juvonen, M. Pasanen, O. Pelkonen,
235. Liehr, J.G., M.J. Ricci, C.R. Jefcoate, R Paakko, and Y Soini (1998). Cytochrome
E.V Hannigan, J.A. Hokanson, and B.T. Zhu P4502A6 (CYP2A6) expression in human hepato-
(1995). 4-Hydroxylation of estradiol by human cellular carcinoma. Hepatology 27, 427-432.
uterine myometrium and myoma microsomes: 247. Rae, J.M., M.D. Johnson, M.E. Lippman, and
Implications for the mechanism of uterine tumori- D.A. Flockhart (2001). Rifampin is a selective,
genesis. Proc. Natl. Acad. Sci. USA 92, 9220-9224. pleiotropic inducer of drug metabolism genes in
236. Lewis, D.F.V, M. Dickins, RJ. Eddershaw, human hepatocytes: Studies with cDNA and
M.H. Tarbit, and RS. Goldfarb (1999). Cyto- oligonucleotide expression arrays. J. Pharmacol.
chrome P450 substrate specificities, substrate Exp. Ther 299, 849-857.
structural templates and enzyme active site geome- 248. Donato, M.T., R Viitala, C. Rodriguez-Antona,
tries. Drug Metabol Drug Interact. 15, 1^9. A. Lindfors, J.V Castell, H. Raunio et al. (2000).
237. Jang, M., L. Cai, G.O. Udeani, K.V Slowing, C.R CYP2A5/CYP2A6 expression in mouse and
Thomas, C.W.W. Beecher et al (1997). Cancer human hepatocytes treated with various in vivo
chemopreventive activity of resveratrol, a natural inducers. Drug Metab. Dispos. 28, 1321-1326.
product derived from grapes. Science 275,218-220. 249. Jover, R., R. Bort, M.J. Gomez-Lechon, and
238. Chun, Y.-J., M.-R Kim, and RR Guengerich (1999). J.V. Castell (2001). Cytochrome P450 regulation
Resveratrol is a selective human cytochrome P450 by hepatocyte nuclear factor 4 in human hepato-
lAl inhibitor. Biochem. Biophys. Res. Commun. cytes: A study using adenovirus-mediated anti-
262, 20-24. sense targeting. Hepatology 33, 668-675.
239. Potter, G.A., L.H. Patterson, E. Wanogho, 250. Ding, S., B.C. Lake, T. Friedberg, and C.R. Wolf
RJ. Perry, PC. Butler, T. Ijaz et al. (2002). The (1995). Expression and alternative sphcing of the
cancer preventative agent resveratrol is converted to cytochrome P-450 CYP2A7. Biochem. J. 306,
the anticancer agent piceatannol by the cytochrome 161-166.
P450 enzyme CYPIBI. Br J. Cancer 86, 774-778. 251. Oscarson, M., R.A. McLellan, V Asp, M. Ledesma,
240. Chun, Y.-J., S. Kim, D. Kim, S.-K. Lee, and M.L. Ruiz, B. Sinues etal. (2002). Characterization
RR Guengerich (2001). A new selective and potent of a novel CYP2A7/CYP2A6 hybrid allele
inhibitor of human cytochrome P450 IBl and its {CYP2A6*]2) that causes reduced CYP2A6 activ-
application to antimutagenesis. Cancer Res. 61, ity Hum. Mutat. 20, 275-283.
8164-8170. 252. Hadidi, H., K. Zahlsen, J.R. Idle, and S. Cholerton
241. Shen, L., S. Qiu, R.B. van Breemen, R Zhang, (1997). A single amino acid substitution
Y. Chen, and J.L. Bolton (1997). Reaction of the (Leul60His) in cytochrome P450 CYP2A6 causes
premarin metabolite 4-hydroxyequilenin semi- switching from 7-hydroxylation to 3-hydroxylation
quinone radical with 2'-deoxyguanosine: Formation of coumarin. Food Chem. Toxicol. 35, 903-907.
of unusual cyclic adducts. J. Am. Chem. Soc. 119, 253. Nakajima, M., J.-T. Kwon, N. Tanaka, T. Zenta,
11126-11127. Y. Yamamoto, H. Yamamoto et al, (2001).
242. Yamano, S., J. Tatsuno, and RJ. Gonzalez (1990). Relationship between interindividual differences in
The CYP2A3 gene product catalyzes coumarin nicotine metabolism and CYP2A6 genetic polymor-
7-hydroxylation in human liver microsomes. phism in humans. Clin. Pharmacol. Ther 69, 72-78.
Biochemistry 19, 1322-1329. 254. Tyndale, R.R and E.M. Sellers (2001). Variable
243. Hakkola, J., M. Pasanen, J. Hukkanen, O. Pelkonen, CYP2A6-mediated nicotine metabolism alters
J. Maenpaa, R.J. Edwards et al. (1996). Expression smoking behavior and risk. Drug Metab. Dispos.
of xenobiotic-metabolizing cytochrome P450 forms 29, 548-552.
in human full-term placenta. Biochem. Pharmacol. 255. Yoshida, R., M. Nakajima, Y Watanabe, J.T. Kwon,
51,403-411. and T. Yokoi (2002). Genetic polymorphisms in
244. Ding, X. and L.S. Kaminsky (2003). Human extra- human CYP2A6 gene causing impaired nicotine
hepatic cytochromes P450: Function in xenobiotic metabolism. Br J. Clin. Pharmacol 54, 511-517.
metabolism and tissue-selective chemical toxicity 256. Gu, D.R, L.J. Hinks, N.E. Morton, and I.N. Day
in the respiratory and gastrointestinal tracts. Annu. (2000). The use of long PCR to confirm three
Rev. Pharmacol. Toxicol. 43, 149-173. common alleles at the CYP2A6 locus and the rela-
245. Godoy W., R.M. Albano, E.G. Moraes, PR. Pinho, tionship between genotype and smoking habit.
R.A. Nunes, E.H. Saito et al. (2002). CYP2A6/2A7 Ann. Hum. Genet. 64, 383-390.
and CYP2E1 expression in human oesophageal 257. Rao, Y, E. Hoffmann, M. Zia, L. Bodin,
mucosa: Regional and inter-individual variation in M. Zeman, E.M. Sellers et al. (2000). Duplications
Human P450s 475

and defects in the CYP2A6 gene: Identification, chromatographic-fluorescence detection method


genotyping, and in vivo effects on smoking. Mol. with a spectrofluorometric method for the determi-
Pharmacol. 58, 747-755. nation of 7-hydroxycoumarin in human urine.
258. Xu, C, S. Goodz, E.M. Sellers, and R.F. Tyndale J. Chromatogr 575, 325-330.
(2002). CYP2A6 genetic variation and potential con- 272. Rautio, A., H. Kraul, A. Kojo, E. Salmela, and
sequences. ^Jv. DrugDeliv. Rev. 54, 1245-1256. O. Pelkonen (1992). Interindividual variability of
259. Tyndale, R.F. and E.M. Sellers (2002). Genetic coumarin 7-hydroxylation in healthy volunteers.
variation in CYP2A6-mediated nicotine metabo- Pharmacogenetics 2, 227-233.
lism alters smoking behavior. Ther. Drug Monit. 273. Soucek, P. (1999). Expression of cytochrome P450
24, 163-171. 2A6 in Escherichia coli: Purification, spectral and
260. Kamataki, T., K. Nunoya, Y. Sakai, H. Kushida, catalytic characterization, and preparation of poly-
and K. Fujita (1999). Genetic polymorphism of clonal antibodies. Arch. Biochem. Biophys. 370,
CYP2A6 in relation to cancer. Mutat. Res. 428, 190-200.
125-130. 274. Nowell, S., C. Sweeney, G. Hammons,
261. Miyamoto, M., Y Umetsu, H. Dosaka-Akita, FF Kadlubar, and N.P Lang (2002). CYP2A6
Y Sawamura, J. Yokota, H. Kunitoh et al. (1999). activity determined by caffeine phenotyping:
CYP2A6 gene deletion reduces susceptibility to Association with colorectal cancer risk. Cancer
lung cancer. Biochem. Biophys. Res. Commun. Epidemiol. Biomarkers Prev. 11, 377-383.
261, 658-660. 275. Le Gal, A., Y Dreano, PG. Gervasi, and F Berthou
262. London, S.J., J.R. Idle, A.K. Daly, and (2001). Human cytochrome P450 2A6 is the major
G.A. Coetzee (1999). Genetic variation of CYP2A6, enzyme involved in the metabolism of three
smoking, and risk of cancer. Lancet 353, 898-899. alkoxyethers used as oxyfuels. Toxicol. Lett. 124,
263. Schulz, T.G., P Ruhnau, and E. Hallier (2001). 47-58.
Lack of correlation between CYP2A6 genotype 276. Duescher, R.J. and A.A. Elfarra (1994). Human
and smoking habits. Adv. Exp. Med. Biol. 500, liver microsomes are efficient catalysts of 1,3-
213-215. butadiene oxidation: Evidence for major roles by
264. Raunio, H., A. Rautio, H. Gullsten, and O. cytochrome P450 2A6 and 2E1. Arch. Biochem.
Pelkonen (2001). Polymorphisms of CYP2A6 and Biophys. 311, 342-349.
its practical consequences. Br J. Clin. Pharmacol. 277. Nunoya, K., T. Yokoi, K. Kimura, K. Inoue,
52,357-363. T. Kodama, M. Funayama et al. (1998). A new
265. Tricker, A.R. (2003). Nicotine metabolism, human deleted allele in the human cytochrome P450 2A6
drug metabolism polymorphisms, and smoking (CYP2A6) gene found in individuals showing poor
behaviour. Toxicology 183, 151-173. metabolic capacity to coumarin and (+)-cis-3,5-
266. Loriot, M.A., S. Rebuissou, M. Oscarson, S. Cenee, dimethyl-2-(3-pyridyl)thiazolidin-4-one hydrochlo-
M. Miyamoto, N. Ariyoshi et al. (2001). Genetic ride (SM-12502). Pharmacogenetics 8, 239-249.
polymorphisms of cytochrome P450 2A6 in a case- 278. Nunoya, K., T. Yokoi, K. Kimura, T. Kainuma,
control study on lung cancer in a French popula- K. Satoh, M. Kinoshita et al. (1999). A new
tion. Pharmacogenetics 11, 3 9 ^ 4 . CYP2A6 gene deletion responsible for the in vivo
267. Zhang, X., K. Amemo, S. Ameno, K. Iw^ahashi, polymorphic metabolism of (+)-c^5-3,5-dimethyl-
H. Kinoshita, T. Kubota et al. (2001). Lack of 2-(3-pyridyl)thiazolidin-4-one hydrochloride in
association between smoking and CYP2A6 gene humans. J. Pharmacol. Exp. Ther 289, 437^42.
polymorphisms in a Japanese population. Nihon 279. Komatsu, T., H. Yamazaki, N. Shimada,
Arukoru Yakuhutsu Igakkai Zasshi 36, 486-490. M. Nakajima, and T. Yokoi (2000). Roles of
268. Oscarson, M. (2001). Genetic polymorphisms cytochromes P450 1A2, 2A6, and 2C8 in 5-
in the cytochrome P450 2A6 (CYP2A6) gene: fluorouracil formation from tegafiir, an anticancer
Implications for interindividual differences in nico- prodrug, in human liver microsomes. Drug Metab.
tine metabolism. Drug Metab. Dispos. 29, 91-95. Dispos. 28, 1457-1463.
269. Nakajima, M., Y Kuroiwa, and T. Yokoi (2002). 280. Ikeda, K., K. Yoshisue, E. Matsushima, S.
Interindividual differences in nicotine metabolism Nagayama, K. Kobayashi, C.A. Tyson et al. (2000).
and genetic polymorphisms of human CYP2A6. Bioactivation of tegafur to 5-fluorouracil is catalyzed
Drug Metab. Rev. 34, 865-877. by cytochrome P-450 2A6 in human liver micro-
270. Daly, A.K., S. Cholerton, W. Gregory, and J.R. Idle somes in vitro, Clin. Cancer Res. 6, 4409-4415.
(1993). Metabolic polymorphisms. Pharmacol. 281. Minoda, Y and E.D. Kharasch (2001). Halothane-
Ther 57, 129-160. dependent lipid peroxidation in human liver micro-
271. Cholerton, S., M.E. Idle, A. Vas, FJ. Gonzalez, and somes is catalyzed by cytochrome P4502A6
J.R. Idle (1992). Comparison of a novel thin-layer (CYP2A6). Anesthesiology 95, 509-514.
476 F. Peter Guengerich

282. Crespi, C.L., B.W. Penman, J.A. Leakey, 291. Messina, E.S., R.F Tyndale, and E.M. Sellers
M.P. Arlotto, A. Stark, A. Parkinson et al. (1990). (1997). A major role for CYP2A6 in nicotine
Human cytochrome P450IIA3:cDNA sequence, C-oxidation by human liver microsomes.
role of the enzyme in the metabolic activation of J. Pharmacol. Exp. Then 282, 1608-1614.
promutagens, comparison to nitrosamine activa- 292. Yamazaki, H., K. Inoue, M. Hashimoto, and
tion by human cytochrome P450IIE1. Carcino- T. Shimada (1999). Roles of CYP2A6 and
genesis 11, 1293-1300. CYP2B6 in nicotine C-oxidation by human liver
283. Yamazaki, H., Y. Inui, C.-H. Yun, M. Mimura, microsomes. Arch. Toxicol. 73, 65-70.
F.P Guengerich, and T. Shimada (1992). 293. Nakajima, M., T. Yamamoto, K. Nunoya, T. Yokoi,
Cytochrome P450 2E1 and 2A6 enzymes as major K. Nagashima, K. Inoue et al. (1996). Characteri-
catalysts for metabolic activation of A^-nitrosodi- zation of CYP2A6 involved in 3'-hydroxylation of
alkylamines and tobacco-related nitrosamines in cotinine in human liver microsomes. J. Pharmacol.
human liver microsomes. Carcinogenesis 13, Exp. Ther 111, 1010-1015.
1789-1794. 294. Hecht, S.S., J.B. Hochalter, PW Villalta, and
284. Smith, T.J., Z. Guo, F.J. Gonzalez, F.P Guengerich, S.E. Murphy (2000). 2'-Hydroxylation of nico-
G.D. Stoner, and C.S. Yang (1992). Metabolism of tine by cytochrome P450 2A6 and human
4-(methylnitrosamino)-1 -(3-pyridyl)-1 -butanone liver microsomes: Formation of a lung carcinogen
(NNK) in human lung and liver microsomes and precursor. Proc. Natl. Acad. Sci. USA 97,
cytochromes P-450 expressed in hepatoma cells. 12493-12497.
Cancer Res. 52, 1757-1763. 295. Su, T., Z.P Bao, Q.Y. Zhang, T.J. Smith, J.Y Hong,
285. Crespi, C.L., B.W. Penman, H.V. Gelboin, and and X.X. Ding (2000). Human cytochrome P450
F.J. Gonzalez (1991). A tobacco smoke-derived CYP2A13: Predominant expression in the res-
nitrosamine, 4-(methylnitrosamino)-1 -(3-pyridyl)- piratory tract and its high efficiency metabolic
1-butanone, is activated by multiple human activation of a tobacco-specific carcinogen, 4-
cytochrome P450s including the polymorphic (methylnitrosamino)-l-(3-pyridyl)-1-butanone.
human cytochrome P4502D6. Carcinogenesis 12, Cancer Res. 60, 5074-5079.
1197-1201. 296. Gillam, E.M.J., L.M. Notley, H. Cai, J.J. DeVoss,
286. Tiano, H.F, R.L. Wang, M. Hosokawa, C. Crespi, and F.P. Guengerich (2000). Oxidation of indole by
K.R. Tindall, and R. Langenbach (1994). Human cytochrome P450 enzymes. Biochemistry 39,
CYP2A6 activation of 4-(methylnitrosamino)-l- 13817-13824.
(3-pyridyl)-l-butanone (NNK): Mutational speci- 297. Nakamura, K., l.H. Hanna, H. Cai, Y Nishimura,
ficity in the gpt gene of AS52 cells. Carcinogenesis K.M. Williams, and FP Guengerich (2001).
15,2859-2866. Coumarin substrates for cytochrome P450 2D6
287. von Weymarn, L.B., N.D. Felicia, X. Ding, and fluorescence assays. Anal. Biochem. 292,280-286.
S.E. Murphy (1999). iV-Nitrosobenzylmethylamine 298. Lindberg, R.L.P and M. Negishi (1989). Alteration
hydroxylation and coumarin 7-hydroxylation: of mouse cytochrome P450^p,^ substrate specificity
Catalysis by rat esophageal microsomes and by mutation of a single amino-acid residue. Nature
cytochrome P450 2A3 and 2A6 enzymes. Chem. 339, 632-634.
Res. Toxicol. 12, 1254-1261. 299. Kitagawa, K., N. Kunugita, M. Kitagawa, and
288. Kushida, H., K. Fujita, A. Suzuki, M. Yamada, T. Kawamoto (2001). CYP2A6''6, a novel polymor-
T. Endo, T. Nohmi et al. (2000). Metabolic activation phism in cytochrome P450 2A6, has a single amino
of A^-alkylnitrosamines in genetically engineered acid substitution (R128Q) that inactivates enzy-
Salmonella typhimurium expressing CYP2E1 or matic activity^ Biol. Chem. 216, 17830-17835.
CYP2A6 together with human NADPH-cytochrome 300. Lewis, D.FV, M. Dickins, B.G. Lake,
P450 reductase. Carcinogenesis 2\, 1227-1232. PJ. Eddershaw, M.H. Tarbit, and PS. Goldfarb
289. Fujita, K. and T. Kamataki (2001). Predicting the (1999). Molecular modeling of the human cyto-
mutagenicity of tobacco-related A^-nitrosamines chrome P450 isoform CYP2A6 and investigations
in humans using 11 strains of Salmonella typhi- of CYP2A substrate selectivity. Toxicology 133,
murium YG7108, each coexpressing a form of 1-33.
human cytochrome P450 along with NADPH- 301. Lewis, D.F. (2002). Molecular modeling of human
cytochrome P450 reductase. Environ. Mol. cytochrome P450-substrate interactions. Drug
Mutagen. 38, 339-346. Metab. Rev. 34, 55-67.
290. Nakajima, M., T. Yamamoto, K. Nunoya, T. Yokoi, 302. Lewis, D.F (2002). Homology modelling of
K. Nagashima, K. Inoue et al. (1996). Role of human CYP2 family enzymes based on the
human cytochrome P4502A6 in C-oxidation of CYP2C5 crystal structure. Xenobiotica 32,
mcoXiwQ. Drug Metab. Dispos. 24, 1212-1217. 305-323.
Human P450s 477

303. Lewis, D.F. and J.W. Gorrod (2002). Molecular cytochrome P450 enzymes in drug abuse and
orbital calculations and nicotine metabolism: A dependence. Pharmacogenomics 3, 185-199.
rationale for experimentally observed metabolite 316. Tan, W, G.E Chen, D.Y. Xing, C.Y Song,
ratios. Drug Metabol. Drug Interact. 19, 29-39. EE Kadlubar, and D.X. Lin (2001). Frequency of
304. Guengerich, F.P., D.-H. Kim, and M. Iwasaki CYP2A6 gene deletion and its relation to risk of
(1991). Role of human cytochrome P-450 IIEl in lung and esophageal cancer in the Chinese popula-
the oxidation of many low molecular weight cancer tion. Int. J. Cancer 95, 96-101.
suspects. Chem. Res. Toxicol. 4, 168-179. 317. Satarug, S., M.A. Lang, P. Yongvanit, P.
305. Kharasch, E.D., D.C. Hankins, P.J. Baxter, and Sithithaworn, E. Mairiang, P. Mairiang et al.
K.E. Thummel (1998). Single-dose disulfiram (1996). Induction of cytochrome P450 2A6 expres-
does not inhibit CYP2A6 activity. Clin. Pharmacol. sion in humans by the carcinogenic parasite infec-
Then 64, 39-45. tion, opisthorchiasis viverrini. Cancer Epidemiol.
306. Murphy, S.E., L.M. Johnson, L.M. Losey, Biomarkers Prev. 5, 795-800.
S.G. Carmella, and S.S. Hecht (2001). Consumption 318. Pasanen, M., Z. Rannala, A. Tooming,
of watercress fails to alter coumarin metabolism in E.A. Sotaniemi, O. Pelkonen, and A. Rautio (1997).
humans. Drug Metab. Dispos. 29, 786-788. Hepatitis A impairs the function of human hepatic
307. Draper, A.J., A. Madan, and A. Parkinson (1997). CYP2A6 in vivo. Toxicology 123, 177-184.
Inhibition of coumarin 7-hydroxylase activity in 319. Koskela, S., J. Hakkola, J. Hukkanen, O. Pelkonen,
human liver microsomes. Arch. Biochem. Biophys. M. Sorri, A. Saranen et al. (1999). Expression
341,47-61. of CYP2A genes in human liver and extrahepatic
308. Zhang, W.J., T. Kilicarslan, R.E Tyndale, and tissues. Biochem. Pharmacol. 57, 1407-1413.
E.M. Sellers (2001). Evaluation of methoxsalen, 320. Oscarson, M., R.A. McLellan, H. GuUsten,
tranylcypromine, and tryptamine as specific and J.A.G. Agundez, J. Benitez, A. Rautio et al.
selective CYP2A6 inhibitors in vitro. Drug Metab. (1999). Identification and characterisation of
Dispos. 26, 897-902. novel polymorphisms in the CYP2A locus:
309. Taavitsainen, P., R. Juvonen, and O. Pelkonen Implications for nicotine metabolism. FEBS Lett.
(2001). In vitro inhibition of cytochrome P450 460, 321-327.
enzymes in human liver microsomes by a potent 321. Fernandez-Salguero, P, S.M.G. Hoffman, S.
CYP2A6 inhibitor. ^ra«^-2-phenylcyclopropy- Cholerton, H. Mohrenweiser, H. Raunio, A. Rautio
lamine (tranylcypromine), and its nonamine ana- et al. (1995). A genetic polymorphism in coumarin
log, cyclopropylbenzene. Drug Metab. Dispos. 29, 7-hydroxylation. Sequence of the human CYP2A
217-222. genes and identification of variant CYP2A 6 aWeles.
310. Koenigs, L.L., R.M. Peter, S.J. Thompson, Am. J. Hum. Genet. 57, 651-660.
A.E. Rettie, and W.F. Trager (1997). Mechanism- 322. Koskela, S., J. Hakkola, J. Hukkanen, O. Pelkonen,
based inactivation of human liver cytochrome M. Sorri, A. Saranen et al. (1999). Expression of
P450 2A6 by 8-methoxypsoralen. Drug Metab. CYP2A genes in human liver and extrahepatic
Dispos. 25, 1407-1415. tissues. Biochem. Pharmacol. 57, 1407-1413.
311. Sellers, E.M., H.L. Kaplan, and R.E Tyndale (2000). 323. Zhang, X., T. Su, Q.Y Zhang, J. Gu, M. Caggana,
Inhibition of cytochrome P450 2A6 increases nico- H. Li et al. (2002). Genetic polymorphisms of the
tine's oral bioavailability and decreases smoking. human CYP2AI3 gene: Identification of single-
Clin. Pharmacol. Then 68, 35-43. nucleotide polymorphisms and functional charac-
312. Koenigs, L.L. and W.F. Trager (1998). Mechanism- terization of an Arg257Cys variant. J. Pharmacol.
based inactivation of P450 2A6 by fiira- Exp. Ther 302, 416^23.
nocoumarins. Biochemistry 37, 10047-10061. 324. Von Weymarn, L.B. and S.E. Murphy (2003).
313. Khojasteh-Bakht, S.C, L.L. Koenigs, R.M. Peter, CYP2A13-catalysed coumarin metabolism: Com-
W.E Trager, and S.D. Nelson (1998). (i?)-(+)- parison with CYP2A5 and CYP2A6. Xenobiotica
Menthofuran is a potent, mechanism-based inacti- 33,73-81.
vator of human liver cytochrome P450 2A6. Drug 325. Mimura, M., T. Baba, Y Yamazaki, S. Ohmori,
Metab. Dispos. 26, 701-704. Y Inui, EJ. Gonzalez et al (1993). Characteri-
314. Wen, X., J.S. Wang, RJ. Neuvonen, and zation of cytochrome P450 2B6 in human liver
J.T Backman (2002). Isoniazid is a mechanism- microsomes. Drug Metab. Dispos. 21, 1048-1056.
based inhibitor of cytochrome P450 1A2, 2A6, 326. Hukkanen, J., A. Pelkonen, J. Hakkola, and
2C19 and 3A4 isoforms in human liver micro- H. Raunio (2002). Expression and regulation of
somes. Eur J. Clin. Pharmacol. 57, 799-804. xenobiotic-metabolizing cytochrome P450 (CYP)
315. Howard, L.A., E.M. Sellers, and R.E Tyndale enzymes in human lung. Crit. Rev. Toxicol. 32,
(2002). The role of pharmacogenetically-variable 391-411.
478 F. Peter Guengerich

327. van der Hoeven, T.A., D.A. Haugen, and MJ. Coon 338. Jover, R., R. Bort, M.J. Gomezlechon, and
(1974). Cytochrome P-450 purified to apparent J.V Castell (1998). Re-expression of C/EBPa
homogeneity from phenobarbital-induced rabbit Hver induces CYP2B6, CYP2C9 and CYP2D6 genes in
microsomes: Catalytic activity and other properties. HepG2 cells. FEBS Lett. 431, 227-230.
Biochem. Biophys. Res. Commun. 60, 569-575. 339. Goodwin, B., L.B. Moore, CM. Stoltz, D.D.
328. Imai, Y. and R. Sato (1974). A gel-electrophoretically McKee, and S.A. Kliewer (2001). Regulation of
homogeneous preparation of cytochrome P-450 from the human CYP2B6 gene by the nuclear pregnane
liver microsomes of phenobarbital-pretreated rabbits. X receptor. Mol. Pharmacol. 60, 427-431.
Biochem. Biophys. Res. Commun. 60, 8-14. 340. Willson, TM. and S.A. KHewer (2002). PXR,
329. Phillips, I.R., E.A. Shephard, A. Ashworth, and CAR and drug metabolism. Nat. Rev. Drug Discov.
B.R. Rabin (1985). Isolation and sequence of a 1,259-266.
human cytochrome P-450 cDNA clone. Proc. Natl. 341. Makinen, J., C. Frank, J. Jyrkkarinne, J. Gynther,
Acad. Sci. USA 82, 983-987. C. Carlberg, and P Honkakoski (2002).
330. Stresser, D.M. and D. Kupfer (1999). Mono- Modulation of mouse and human phenobarbital-
specific antipeptide antibody to cytochrome P-450 responsive enhancer module by nuclear receptors.
2B6. DrugMetah. Dispos. 11, 517-525. Mol. Pharmacol. 62, 366-378.
331. Code, E.L., C.L. Crespi, B.W. Penman, 342. Drocourt, L., J.C. Ourlin, J.M. Pascussi, P. Maurel,
F.J. Gonzalez, T.K.H. Chang, and D.J. Waxman and M.J. Vilarem (2002). Expression of CYP3A4,
(1997). Human cytochrome P4502B6. Interindivi- CYP2B6, and CYP2C9 is regulated by the vitamin
dual hepatic expression, substrate specificity, and D receptor pathway in primary human hepatocytes.
role in procarcinogen activation. Drug Metab. J. Biol. Chem. Ill, 25125-25132.
Dispos. 25, 985-993. 343. Chang, TK., S.M. Bandiera, and J. Chen (2003).
332. Ekins, S., M. VandenBranden, B.J. Ring, Constitutive androstane receptor and pregnane X
J.S. Gillespie, T.J. Yang, H.V Gelboin et al. (1998). receptor gene expression in human liver: Inter-
Further characterization of the expression in liver individual variability and correlation with CYP2B6
and catalytic activity of CYP2B6. J. Pharmacol. mRNA levels. Drug Metab. Dispos. 31, 7-10.
Exp. Ther 286, 1253-1259. 344. Maghch, J.M., D.J. Parks, L.B. Moore, J.L. Collins,
333. Imaoka, S., T. Yamada, T. Hiroi, K. Hayashi, B. Goodwin, A.N. Billin et al. (2003).
T. Sakaki, Y Yabusaki etal. (1996). Multiple forms Identification of a novel human CAR agonist and
of human P450 expressed in Saccharomyces its use in the identification of CAR target genes.
cerevisiae: Systematic characterization and com- J. Biol. Chem. 11%, 17277-17283.
parison with those of the rat. Biochem. Pharmacol. 345. Wang, H., S. Faucette, T. Sueyoshi, R. Moore,
51, 1041-1050. S. Ferguson, M. Negishi et al. (2003). A novel
334. Roy, P, L.J. Yu, C.L. Crespi, and D.J. Waxman distal enhancer module regulated by PXR/CAR
(1999). Development of a substrate-activity based is essential for the maximal induction of CYP2B6
approach to identify the major human liver P-450 gene expression, o^ 5/o/. Chem. 11%, 14146-14152.
catalysts of cyclophosphamide and ifosfamide 346. Miles, J.S., A.W McLaren, F.J. Gonzalez, and
activation based on cDNA-expressed activities and C.R. Wolf (1990). Alternative splicing in the
liver microsomal P-450 profiles. Drug Metab. human cytochrome P450IIB6 gene: Use of a cryp-
Dispos. 27, 655-666. tic exon within intron 3 and splice acceptor site
335. Gervot, L., B. Rochat, J.C. Gautier, within exon 4. Nucleic Acids Res. 18, 189.
F. Bohnenstengel, H. Kroemer, V de Berardinis 347. Zanger, U.M., J. Fischer, K. Klein, andT Lang (2002).
et al. (1999). Human CYP2B6: Expression, Detection of single nucleotide polymorphisms in
inducibility and catalytic activities. Pharmaco- CYP2B6 gene. Meth. Enzymol. 357, 45-53.
genetics 9, 295-306. 348. Lang, T, K. Klein, J. Fischer, A.K. Nussler,
336. Hanna, I.H., J.R. Reed, F.P. Guengerich, and P Neuhaus, U. Hofmann et al. (2001). Extensive
PF. Hollenberg (2000). Expression of human genetic polymorphism in the human CYP2B6 gene
cytochrome P450 2B6 in Escherichia coli: with impact on expression and fiinction in human
Characterization of catalytic activity and expres- liver. Pharmacogenetics 11, 399^15.
sion levels in human liver. Arch. Biochem. 349. Ariyoshi, N., M. Miyazaki, K. Toide, Y Sawamura,
Biophys. 316, 206-216. and T. Kamataki (2001). A single nucleotide
337. Sueyoshi, T., T. Kawamoto, 1. Zelko, polymorphism of CYP2B6 found in Japanese
P Honkakoski, and M. Neigishi (1999). The enhances catalytic activity by autoactivation.
repressed nuclear receptor CAR responds to Biochem. Biophys. Res. Commun. 281, 1256-1260.
phenobarbital in activating the human CYP2B6 350. Haugen, D.A. and M.J. Coon (1976). Properties
gene. J. Biol. Chem. 274, 6043-6046. of electrophoretically homogenous phenobarbital-
Human P450s 479

inducible and p-naphthoflavone-inducible forms 363. Lewis, D.FV, B.G. Lake, M. Dickins,
of liver microsomal cytochrome P-450. J. Biol. RJ. Eddershaw, M.H. Tarbit, and PS. Goldfarb
Chem. 251, 7929-7939. (1999). Molecular modelling of CYP2B6, the
351. Coon, M.J. (1981). The 1980 Bernard B. Brodie human CYP2B isoform, by homology with the sub-
Award lecture. Drug metabolism by cytochrome strate-bound CYP102 crystal structure: Evalua-
P-450: Progress and perspectives. Drug Metab. tion of CYP2B6 substrate characteristics, the
Dispos. 9, \~^. cytochrome b^ binding site and comparisons with
352. Guengerich, P.P. (1977). Separation and purifica- CYP2B1 and CYP2B4. Xenobiotica 29, 361-393.
tion of multiple forms of microsomal cytochrome 364. Lewis, D.F, S. Modi, and M. Dickins (2001).
P-450. Activities of different forms of cytochrome Quantitative structure-activity relationships
P-450 towards several compounds of environ- (QSARs) within substrates of human cytochromes
mental interest. J. Biol. Chem. 252, 3970-3979. P450 involved in drug metabolism. Drug Metabol.
353. Lu, A.Y.H. and S.B. West (1978). Reconstituted Drug Interact. 18, 221-242.
mammalian mixed-function oxidases: Requirements, 365. Bathelt, C , R.D. Schmid, and J. Pleiss (2002).
specificities and other properties. Pharmacol. Then Regioselectivity of CYP2B6: Homology model-
2, 337-338. ing, molecular dynamics simulation, docking.
354. Ekins, S. and S.A. Wrighton (1999). The role of J. Mol. Model. (Online) 8, 327-335.
CYP2B6 in human xenobiotic metabolism. Drug 366. Domanski, T.L., K.M. Schultz, F. Roussel,
Metab. Rev. 31, 719-754. J.C. Stevens, and J.R. Halpert (1999). Structure-
355. Chang, TK.H., G.F. Weber, C.L. Crespi, and function analysis of human cytochrome P-450 2B6
D.J. Waxman (1993). Differential activation of using a novel substrate, site-directed mutagenesis,
cyclophosphamide and ifosphamide by cyto- and molecular modeling. J. Pharmacol. Exp. Then
chromes P-450 2B and 3A in human liver micro- 290, 1141-1147.
somes. Cancer Res. 53, 5629-5637. 366a.Scott, E.E., YA. He, M.A. Wester, C.C. Chin, J.R.
356. Xie, H.J., U. Yasar, S. Lundgren, L. Griskevicius, Halpert, E.R Johnson, and CD. Stout (2003). An
Y Terelius, M. Hassan et al. (2003). Role of poly- open conformation of mammalian cytochrome
morphic human CYP2B6 in cyclophosphamide P450 2B4 at 1.6-A resolution. Proc. Natl. Acad.
bioactivation. Pharmacogenomics J. 3, 53-61. Sci. U.S.A. 100, 13121-13122.
357. Yanagihara, Y, S. Kariya, M. Ohtani, K. Uchino, 367. Guo, Z., S. Raeissi, R.B. White, and J.C. Stevens
T. Aoyama, Y Yamamura et al. (2001). Involve- (1997). Orphenadrine and methimazole inhibit
ment of CYP2B6 in A^-demethylation of ketamine multiple cytochrome P450 enzymes in human
in human liver microsomes. Drug Metab. Dispos. liver microsomes. Drug Metab. Dispos. 25,
29, 887-890. 390-393.
358. Court, M.H., S.X. Duan, L.M. Hesse, 368. Stiborova, M., L. Borek-Dohalska, P Hodek,
K. Venkatakrishnan, and D.J. Greenblatt (2001). J. Mraz, and E. Frei (2002). New selective
Cytochrome P-450 2B6 is responsible for inhibitors of cytochromes P450 2B and their
interindividual variability of propofol hydroxyla- application to antimutagenesis of tamoxifen. Arch.
tion by human liver microsomes. Anesthesiology Biochem. Biophys. 403, 4 1 ^ 9 .
94, 110-119. 369. Rae; J.M., N.V Soukhova, D.A. Flockhart, and
359. Heyn, H., R.B. White, and J.C. Stevens (1996). Z. Desta (2002). Triethylenethiophosphoramide
Catalytic role of cytochrome P4502B6 in the is a specific inhibitor of cytochrome P4502B6:
A^-demethylation of iS-mephenytoin. Drug Metab. Implications for cyclophosphamide metabolism.
Dispos. 24, 948-954. Drug Metab. Dispos. 30, 525-530.
360. Ko, J.W, Z. Desta, and D.A. Flockhart (1998). 370. Kent, U.M., D.E. Mills, R.V Rajnarayanan,
Human A^-demethylation of (5')-mephenytoin by W.L. Alworth, and PR HoUenberg (2002). Effect of
cytochrome P450s 2C9 and 2B6. Drug Metab. 17a-ethynylestradiol on activities of cytochrome
Dispos. 26,115-11^. P450 2B (P450 2B) enzymes: Characterization of
361. Ekins, S., M. VandenBranden, B.J. Ring, and inactivation of P450s 2B1 and 2B6 and identifica-
S.A. Wrighton (1997). Examination of purported tion of metabolites. J. Pharmacol. Exp. Then 300,
probes of human CYP2B6. Pharmacogenetics 7, 549-558.
165-179. 371. Kiipfer, A. and R. Preisig (1984). Pharmacogenetics
362. Ariyoshi, N., K. Oguri, N. Koga, H. Yoshimura, of mephenytoin: A new drug hydroxylation poly-
andY Funae (1995). MetaboUsm of highly persist- morphism in man. Eur J. Clin. Pharmacol. 26,
ent PCB congener, 2, 4,5, 2',4',5'-hexachloro- 753-759.
biphenyl, by human CYP2B6. Biochem. Biophys. 372. Wedlund, P I , W.S. Aslanian, C.B. McAllister,
Res. Commun. Ill, 455-460. G.R. Wilkinson, and R.A. Branch (1984).
480 F. Peter Guengerich

Mephenytoin hydroxylation deficiency in 383. Gerbal-Chaloin, S., J.M. Pascussi, L. Pichard-


Caucasians: Frequency of a new oxidative drug Garcia, M. Daujat, F. Waechter, J.M. Fabre et al.
metabolism polymorphism. Clin. Pharmacol. Ther. (2001). Induction of CYP2C genes in human hepa-
36, 773-780. tocytes in primary culture. Drug Metab. Dispos.
373. Knodell, R.G., S.D. Hall, G.R. Wilkinson, and 29,242-251.
F.P. Guengerich (1987). Hepatic metabolism 384. Soyama, A., Y. Saito, N. Hanioka, N. Murayama,
of tolbutamide: Characterization of the form of O. Nakajima, N. Katori et al. (2001). Non-
cytochrome P-450 involved in methyl hydro- synonymous single nucleotide alterations found in
xylation and relationship to in vivo disposition. the CYP2C8 gene result in reduced in vitro pacli-
J. Pharmacol. Exp. Ther 241, 1112-1119. taxel metabolism. Biol. Pharm. Bull. 24,1427-1430.
374. Brian, W.R., RK. Srivastava, D.R. Umbenhauer, 385. Rahman, A., K.R. Korzekwa, J. Grogan, F.J.
R.S. Lloyd, and F.R Guengerich (1989). Expression Gonzalez, and J.W. Harris (1994). Selective bio-
of a human liver cytochrome P-450 protein with transformation of taxol to 6a-hydroxytaxol by
tolbutamide hydroxylase activity in Saccharomyces human cytochrome P450 2C8. Cancer Res. 54,
cerevisiae. Biochemistry 28, 4993^999. 5543-5546.
375. Ged, C, D.R. Umbenhauer, T.M. Bellew, 386. Leo, M.A., J.M. Lasker, J.L. Raucy, C.I. Kim,
R.W. Bork, PK. Srivastava, N. Shinriki et al. M. Black, and C.S. Lieber (1989). Metabolism
(1988). Characterization of cDNAs, mRNAs, and of retinol and retinoic acid by human liver
proteins related to human liver microsomal cytochrome P450IIC8. Arch. Biochem. Biophys.
cytochrome P-450 (iS')-mephenytoin 4'-hydroxy- 269,305-312.
lase. Biochemistry 27, 6929-6940. 387. Yamazaki, H., A. Shibata, M. Suzuki, M. Nakajima,
376. Romkes, M., M.B. Faletto, J.A. Blaisdell, N. Shimada, FP Guengerich et al (1999). Oxi-
J.L. Raucy, and J.A. Goldstein (1991). Cloning and dation of troglitazone to a quinone-type metabolite
expression of complementary DNAs for multiple catalyzed by cytochrome P450 2C8 and 3A4 in
members of the human cytochrome P450I1C human liver microsomes. Drug Metab. Dispos. 27,
subfamily. Biochemistry 30, 3247-3255. 1260-1266.
377. Wrighton, S.A., J.C. Stevens, G.W. Becker, and 388. Marques-Soares, C , S. Dijols, A.-C. Macherey,
M. VandenBranden (1993). Isolation and charac- M.R. Wester, E.F. Johnson, PM. Dansette et al.
terization of human liver cytochrome P450 2C19: (2003). Sulfaphenazole derivatives as tools for
Correlation between 2C19 and 5-mephenytoin comparing cytochrome P450 2C5 and human
4'-hydroxylation. Arch. Biochem. Biophys. 306, cytochrome P450 2Cs: Identification of a new high
240-245. affinity substrate common to those CYP 2C
378. Goldstein, J.A., M.B. Faletto, M. Romkes-Sparks, enzymes. Biochemistry 42, 6363-6369.
T. Sullivan, S. Kitareewan, J.L. Raucy et al. 389. Wester, M.R., E.F. Johnson, C. Marques-Soares,
(1994). Evidence that CYP2C19 is the major P Dansette, D. Mansuy, and CD. Stout (2003). The
(»S)-mephenytoin 4'-hydroxylase in humans. structure of a substrate complex of mammalian
Biochemistry ^:^, 1743-1752. cytochrome P450 2C5 at 2.3 A resolution:
379. Nelson, D.R., L. Koymans, T. Kamataki, Evidence for multiple substrate binding modes.
J.J. Stegeman, R. Feyereisen, D.J. Waxman Biochemistry 42, 9335-9345.
et al. (1996). P450 superfamily: Update on new 389a.Schoch, G.A., J.K. Yano, M.R. Wester, K.J. Griffin,
sequences, gene mapping, accession numbers, and CD. Stout, and E.F Johnson (2004). Structure
nomenclature. Pharmacogenetics 6, 1-42. of human microsomal cytochrome P450 2C8.
380. Dai, D., D.C. Zeldin, J.A. Blaisdell, B. Chanas, Evidence for a peripheral fatty acid binding site. J.
S.J. Coulter, B.I. Ghanayem et al. (2001). Biol. Chem. 279, 9497-9503.
Polymorphisms in human CYP2C8 decrease 390. Ha-Duong, N.T., S. Dijols, C Marques-Soares,
metabolism of the anticancer drug paclitaxel and C Minoletti, PM. Dansette, and D. Mansuy (2001).
arachidonic acid. Pharmacogenetics 11, 597-607. Synthesis of sulfaphenazole derivatives and their
381. Klose, T.S., J.A. Blaisdell, and J.A. Goldstein use as inhibitors and tools for comparing the active
(1999). Gene structure of CYP2C8 and extra- sites of human liver cytochromes P450 of the 2C
hepatic distribution of the human CYP2Cs. subfamily J. Med. Chem. 44, 3622-3631.
J. Biochem. Mol. Toxicol. 13, 289-295. 391. Ha-Duong, N.T., C Marques-Soares, S. Dijols,
382. Bahadur, N., J.B. Leathart, E. Mutch, D. Steimel- M.A. Sari, PM. Dansette, and D. Mansuy (2001).
Crespi, S.A. Dunn, R. Gilissen et al. (2002). Interaction of new sulfaphenazole derivatives with
CYP2C8 polymorphisms in Caucasians and human liver cytochrome P4502Cs: Structural
their relationship with paclitaxel 6a-hydroxylase determinants required for selective recognition by
activity in human liver microsomes. Biochem. CYP2C9 and for inhibition of human CYP2Cs.
Pharmacol. 64, 1579-1589. Arch. Biochem. Biophys. 394, 189-200.
Human P450s 481

392. Zilly, W., D.D. Breimer, and E. Richter (1975). CYP2C9 by the constitutive androstane receptor:
Induction of drug metabolism in man after Discovery of a new distal binding site. Mol.
rifampicin treatment measured by increased hexo- Pharmacol. 62, 737-746.
barbital and tolbutamide clearance. Eur. J. Clin. 404. Pascussi, J.M., S. Gerbal-Chaloin, L. Drocourt,
Pharmacol. 9,219-227. P. Maurel, and M.J. Vilarem (2003). The expression
393. Umbenhauer, D.R., M.V. Martin, R.S. Lloyd, and of CYP2B6, CYP2C9 and CYP3A4 genes: A tangle
F.P. Guengerich (1987). Cloning and sequence of networks of nuclear and steroid receptors.
determination of a complementary DNA related Biochim. Biophys. Acta 1619, 243-253.
to human liver microsomal cytochrome P-450 405. Ibeanu, G.C. and J.A. Goldstein (1995). Transcrip-
iS-mephenytoin 4-hydroxylase. Biochemistry 26, tional regulation of human CYP2C genes:
1094-1099. Functional comparison of CYP2C9 and CYP2C18
394. Knodell, R.G., R.K. Dubey, G.R. Wilkinson, and promoter regions. Biochemistry 34, 8028-8036.
F.R Guengerich (1988). Oxidative metabolism 406. Scott, J. and PL. Poffenbarger (1978). Pharmaco-
of hexobarbital in human liver: Relationship to genetics of tolbutamide metabolism in humans.
polymorphic 5-mephenytoin 4-hydroxylation. Diabetes 2^, A\-5\.
J. Pharmacol. Exp. Then 245, 845-849. 407. Ohgiya, S., M. Komori, H. Ohi, K. Shiramatsu,
395. Yasumori, T., N. Murayama, Y. Yamazoe, A. Abe, N. Shinriki, and T. Kamataki (1992). Six-base
Y. Nogi, T. Fukasawa et al. (1989). Expression deletion occurring in messages of human cyto-
of a human P-450IIC gene in yeast cells using chrome P-450 in the CYP2C subfamily results in
galactose-inducible expression system. Mol reduction of tolbutamide hydroxylase activity.
Pharmacol. 35, 443-449. Biochem. Int. 27, 1073-1081.
396. Srivastava, RK., C.-H. Yun, RH. Beaune, C. Ged, 408. Goldstein, J.A. and S.M.F Demorais (1994).
and RR Guengerich (1991). Separation of Biochemistry and molecular biology of the human
human liver tolbutamine hydroxylase and (S)- CYP2C subfamily. Pharmacogenetics 4, 285-299.
mephenytoin 4'-hydroxylase cytochrome P-450 409. Inoue, K., H. Yamazaki, K. Imiya, S. Akasaka,
enzymes. Mol. Pharmacol. 40, 69-79. KR Guengerich, andT. Shimada (1997). Relation-
397. Treluyer, J.M., G. Gueret, G. Cheron, M. Sonnier, ship between CYP2C9 and CYP2C19 genotypes
and T. Cresteil (1997). Developmental expres- and tolbutamide methyl hydroxylation and
sion of CYP2C and CYP2C-dependent activities in iS'-mephenytoin 4'-hydroxylation in livers of
the human liver: in-vivo/in-vitro correlation and Japanese and Caucasian populations. Pharma-
inducibility. Pharmacogenetics 7, 441-452. cogenetics 7, 103-113.
398. Brenner, S.S., C. Herrlinger, K. Dilger, 410. Lee, C.R., J.A. Goldstein, and J.A. Pieper (2002).
T.E. Murdter, U. Hoftnann, C. Marx et al (2003). Cytochrome P450 2C9 polymorphisms: A compre-
Influence of age and cytochrome P450 2C9 geno- hensive review of the in-vitro and human data.
type on the steady-state disposition of diclofenac Pharmacogenetics 12, 251-263.
and celecoxib. Clin. Pharmacokinet. 42, 283-292. 411. Xie, H.G., H.C. Prasad, R.B. Kim, and CM. Stein
399. Obach, R.S., Q.Y. Zhang, D. Dunbar, and (2002). CYP2C9 allelic variants: Ethnic distribu-
L.S. Kaminsky (2001). Metabolic characterization tion and functional significance. Adv. Drug Deliv.
of the major human small intestinal cytochrome Rev. 54, 1257-1270.
P450s. DrugMetab. Dispos. 29, 347-352. 412. Dickmann, L.J., A.E. Rettie, M.B. Kneller,
400. Morel, R, RH. Beaune, D. Ratanasavanh, R.B. Kim, A.J. Wood, CM. Stein et al (2001).
J.-P. Flinois, C.-S. Yang, F.P. Guengerich et al. Identification and functional characterization of a
(1990). Expression of cytochrome P-450 enzymes new CYP2C9 variant (CYP2C9*5) expressed among
in cultured human hepatocytes. Eur J. Biochem. African Americans. Mol Pharmacol 60, 382-387.
191,AZl-A^A. 413. Shintani, M., I. leiri, K. Inoue, K. Mamiya,
401. Raucy, XL., L. Mueller, K. Duan, S.W. Allen, H. Ninomiya, N. Tashiro et al (2001). Genetic
S. Strom, and J.M. Lasker (2002). Expression and polymorphisms and functional characterization of
induction of CYP2C P450 enzymes in primary the 5'-flanking region of the human CYP2C9
cultures of human hepatocytes. J. Pharmacol. Exp. gene: In vitro and in vivo studies. Clin. Pharmacol.
Then 302, 475-482. Then 70, 175-182.
402. Gerbal-Chaloin, S., M. Daujat, J.M. Pascussi, 414. Warner, S.C, C Finta, and RG. Zaphiropoulos
L. Pichard-Garcia, M.J. Vilarem, and P. Maurel (2001). Intergenic transcripts containing a novel
(2002). Transcriptional regulation of CYP2C9 gene. human C5^ochrome P450 2C exon 1 spliced to
Role of glucocorticoid receptor and constitutive sequences from the CYP2C9 gene. Mol Biol Evol
androstane receptor. J. Biol. Chem. 211, 209-217. 18, 1841-1848.
403. Ferguson, S.S., E.L. LeCluyse, M. Negishi, and 415. Yasar, U, S. Lundgren, E. Eliasson, A. Bennet,
J.A. Goldstein (2002). Regulation of human B. Wiman, U. de Faire et al (2002). Linkage
482 F. Peter Guengerich

between the CYP2C8 and CYP2C9 genetic 426. Backes, W.L., C.J. Batie, and G.E Cawley (1998).
polymorphisms. Biochem. Biophys. Res. Commun. Interactions among P450 enzymes when combined
299, 25-28. in reconstituted systems: Formation of a 2B4-1A2
416. Miners, J.O. and D.J. Birkett (1998). Cytochrome complex with a high affinity for NADPH cytochrome
P4502C9: An enzyme of major importance in P450 reductase. Biochemistry 37, 12852-12859.
human drug metabolism. Br. J. Clin. Pharmacol. 427. Hutzler, J.M., D. Kolwankar, M.A. Hummel, and
45, 525-538. TS. Tracy (2002). Activation of CYP2C9-
417. Giancarlo, G.M., K. Venkatakrishnan, B.W. Granda, mediated metabolism by a series of dapsone
L.L. von Moltke, and D.J. Greenblatt (2001). analogs: Kinetics and structural requirements.
Relative contributions of CYP2C9 and 2C19 to Drug Metab. Dispos. 30, 1194-1200.
phenytoin 4-hydroxylation in vitro: Inhibition by 428. Hutzler, J.M., L.C. Wienkers, J.L. Wahlstrom,
sulfaphenazole, omeprazole, and ticlopidine. Eur J. T.J. Carlson, and TS. Tracy (2003). Activation of
Clin. Pharmacol. 57, 31-36. cytochrome P450 2C9-mediated metabolism:
418. Draper, A.J. and B.D. Hammock (2000). Mechanistic evidence in support of kinetic obser-
Identification of CYP2C9 as a human liver micro- vations. Arch. Biochem. Biophys. 410, 16-24.
somal linoleic acid epoxygenase. Arch. Biochem. 429. Ueng, Y-E, T Kuwabara, Y.-J. Chun, and
Biophys. 376, 199-205. EP Guengerich (1997). Cooperativity in oxidations
419. McSorley, L.C. and A.K. Daly (2000). Identification catalyzed by cytochrome P450 3A4. Biochemistry
of human cytochrome P450 isoforms that contribute 36,370-381.
to all-/ra«^-retinoic acid 4-hydroxylation. Biochem. 430. Takanashi, K., H. Tainaka, K. Kobayashi,
Pharmacol. 60, 517-526. T Yasumori, M. Hosakawa, and K. Chiba (2000).
420. Lee, C.R., J.A. Pieper, R.F. Frye, A.L. CYP2C9 Ile^^^ and Leu^^^ variants: Enzyme kinetic
Hinderliter, J.A. Blaisdell, and J.A. Goldstein study with seven substrates. Pharmacogenetics 10,
(2003). Tolbutamide, flurbiprofen, and losartan as 95-104.
probes of CYP2C9 activity in humans. J. Clin. 431. Yasar, U, E. Eliasson, C. Forslund-Bergengren,
Pharmacol. 43,84-91. G. Tybring, M. Gadd, E Sjoqvist et al. (2001). The
421. Sandberg, M., U. Yasar, P Stromberg, J.O. Hoog, and role of CYP2C9 genotype in the metabolism of
E. Eliasson (2002). Oxidation of celecoxib by poly- diclofenac in vivo and in vitro. Eur J. Clin.
morphic cytochrome P450 2C9 and alcohol dehy- Pharmacol. 57, 729-735.
drogenase. Br J. Clin. Pharmacol. 54, 423-429. 432. Ridderstrom, M., C. Masimirembwa, S. Trump-
422. Tang, C , M. Shou, T.H. Rushmore, Q. Mei, Kallmeyer, M. Ahlefelt, C. Otter, andTB. Andersson
P Sandhu, E.J. WooXf etal (2001). In-vitro metab- (2000). Arginines 97 and 108 in CYP2C9 are impor-
olism of celecoxib, a cyclooxygenase-2 inhibitor, tant determinants of the catalyticfiinction.Biochem.
by allelic variant forms of human liver microsomal Biophys. Res. Commun. 270, 983-987.
cytochrome P450 2C9: Correlation with CYP2C9 433. Flanagan, J.U., L.A. McLaughlin, M.J. Paine,
genotype and in-vivo pharmacokinetics. M.J. Sutclifife, G.C. Roberts, and C.R. Wolf
Pharmacogenetics 11, 223-235. (2003). Role of conserved Asp^^^ of cytochrome
423. Tang, C.Y, M.G. Shou, and A.D. Rodrigues P450 2C9 in substrate recognition and catalytic
(2000). Substrate-dependent effect of acetonitrile activity Biochem. J. 370, 921-926.
on human liver microsomal cytochrome P4502C9 434. He, M., K.R. Korzekwa, J.P Jones, A.E. Rettie, and
(CYP2C9) activity Drug Metab. Dispos. 28, WF. Trager (1999). Structural forms of phenpro-
567-572. coumon and warfarin that are metabolized at the
424. Yamazaki, H., E.M.J. Gillam, M.-S. Dong, active site of CYP2C9, Arch. Biochem. Biophys.
WW. Johnson, EP Guengerich, and T Shimada 372, 16-28.
(1997). Reconstitution of recombinant human 435. Mancy, A., S. Dijols, S. Poli, EP. Guengerich, and
cytochrome P450 2C9 and comparison with D. Mansuy (1997). Interaction of sulfaphenazole
cytochrome P450 and other forms: Effects of derivatives with human liver cytochromes P450 2C:
cytochrome P450:P450 and cytochrome ?450:b^ Molecular origin of the specific inhibitory effects
interactions. Arch. Biochem. Biophys. 342,329-337. of sulfaphenazole on CYP 2C9 and consequences
425. Yamazaki, H., T. Komatsu, K. Ohyama, M. for the sub-strate binding topology. Biochemistry
Nakamura, S. Asahi, N. Shimada et al. (2002). 35, 16205-16212.
Roles of NADPH-P450 reductase and apo- and 436. Mancy A., P Broto, S. Dijols, PM. Dansette, and
holo-cytochrome b^ on xenobiotic oxidations cat- D. Mansuy (1995). The substrate binding site of
alyzed by 12 recombinant human cytochrome human liver cytochrome P450 2C9: An approach
P450s expressed in membranes of Escherichia using designed tienilic acid derivatives and molec-
coli. Protein Expr Purif. 24, 329-337. ular modeling. Biochemistry 34, 10365-10375.
Human P450s 483

437. Tsao, C.C., M.R. Wester, B. Ghanayem, 447. Veronese, M.E., J.O. Miners, D. Randies,
S.J. Coulter, B. Chanas, E.F. Johnson et al. (2001). D. Gregov, and D.J. Birkett (1990). Validation of
Identification of human CYP2C19 residues that the tolbutamide metabolic ratio for population
confer .S'-mephenytoin 4'-hydroxylation activity to screening with use of sulfaphenazole to produce
CYP2C9. Biochemistry 40, 1937-1944. model phenotypic poor metabolizers. Clin.
438. Jung, E, K.J. Griffin, T.H. Richardson, M. Yang, Pharmacol. Ther 47, 403^11.
and E.F. Johnson (1998). Identification of amino 448. Wen, X., J.S. Wang, J.T. Backman, J. Laitila, and
acid substitutions that confer a high affinity for P.J. Neuvonen (2002). Trimethoprim and sul-
sulphaphenazole binding and a high catalytic effi- famethoxazole are selective inhibitors of CYP2C8
ciency for warfarin metabolism to P450 2C19. and CYP2C9, respectively. Drug Metab. Dispos.
Biochemistry:^!, 16270-16279. 30,631-635.
439. Niwa, T., A. Kageyama, K. Kishimoto, 449. Wen, X., J.S. Wang, K.T Kivisto, PJ. Neuvonen,
Y. Yabusaki, R Ishibashi, and M. Katagiri (2002). and J.T. Backman (2001). In vitro evaluation of
Amino acid residues affecting the activities of valproic acid as an inhibitor of human cytochrome
human cytochrome P450 2C9 and 2C19. Drug P450 isoforms: Preferential inhibition of
Metab. Dispos. 30, 931-936. cytochrome P450 2C9 (CYP2C9). Br J. Clin.
440. Melet, A., N. Assrir, P. Jean, M. Pilar Lopez- Pharmacol. 52, 547-553.
Garcia, C. Marques-Soares, M. Jaouen etal. (2003). 450. Zhang, Z.Y., J. Kerr, R.S. Wexler, H.Y. Li,
Substrate selectivity of human cytochrome P450 A.J. Robinson, PR Harlow et al. (1997). Warfarin
2C9: Importance of residues 476, 365, and 114 in analog inhibition of human CYP2C9-catalyzed S-
recognition of diclofenac and sulfaphenazole and in warfarin 7-hydroxylation. Thromb. Res. 88,
mechanism-based inactivation by tienilic acid. Arch. 389-398.
Biochem. Biophys. 409, 80-91. 451. Beaune, P, PM. Dansette, D. Mansuy, L. Kiffel,
441. Guengerich, EP, I.H. Hanna, M.V Martin, and M. Finck, C Amar et al. (1987). Human anti-
E.M.J. Gillam (2003). Role of glutamic acid 216 endoplasmic reticulum autoantibodies appearing
in cytochrome P450 2D6 substrate binding and in a drug-induced hepatitis are directed against a
catalysis. Biochemistry 42, 1245-1253. human liver cytochrome P-450 that hydroxylates
442. Lewis, D.EV, M. Dickins, R.J. Weaver, the drug. Proc. Natl. Acad Sci. USA 84, 551-555.
PJ. Eddershaw, PS. Goldfarb, and M.H. Tarbit 452. Dansette, PM., D.C Thang, H. El Amri, and
(1998). Molecular modelling of human CYP2C D. Mansuy (1992). Evidence for thiophene-S-
subfamily enzymes CYP2C9 and CYP2C19: oxide as a primary reactive metabolite of thio-
Rationalization of substrate specificity and site- phene in vivo: Formation of a dihydrothiophene
directed mutagensis experiments in the CYP2C sulfoxide mercapturic acid. Biochem. Biophys.
subfamily. Xenobiotica 28, 235-268. Res. Commun. 186, 1624-1630.
443. Afzelius, L., I. Zamora, M. Ridderstrom, 453. Beaune, P., D. Pessayre, P. Dansette, D. Mansuy,
T.B. Andersson, A. Karlen, and CM. Masimirembwa and M. Manns (1994). Autoantibodies against
(2001). Competitive CYP2C9 inhibitors: Enzyme C3^ochromes P450: Role in human diseases. Adv.
inhibition studies, protein homology modeling, and Pharmacol. 30, 199-245.
three-dimensional quantitative structure-activity 454. Carlile, D.J., N. Hakooz, M.K. BayUss, and
relationship analysis. Mol. Pharmacol. 59, 909-919. J.B. Houston (1999). Microsomal prediction of
444. Ridderstr5m, M., I. Zamora, O. Fjellstrom, and in vivo clearance of CYP2C9 substrates in humans.
T.B. Andersson (2001). Analysis of selective regions Br J. Clin. Pharmacol. 47, 625-635.
in the active sites of human cytochromes P450,2C8, 455. Goldstein, J.A. (2001). Clinical relevance of
2C9, 2C18, and 2C19 homology models using genetic polymorphisms in the human CYP2C
GRID/CPCA. J. Med. Chem. 44, 4072^081. subfamily. Br J. Clin. Pharmacol. 52, 349-355.
445. de Groot, M.J., A.A. Alex, and B.C. Jones (2002). 456. Kaminsky, L.S. and Z.Y. Zhang (1997). Human
Development of a combined protein and pharma- P450 metabolism of warfarin. Pharmacol. Ther
cophore model for cytochrome P450 2C9. J. Med. 73, 67-74.
Chem. 45, 1983-1993. 457. Yamazaki, H. and T Shimada (1997). Human
445a.Williams, PA., J. Cosme, A. Ward, H.C. Angove, liver cytochrome P450 enzymes involved in the
D. Matak Vinkovic, and H. Jhoti (2003). Crystal 7-hydroxylation of ^- and »S-warfarin enantiomers.
structure of human cytochrome P450 2C9 with Biochem. Pharmacol. 54, 1195-1203.
bound warfarin. Nature 424, 464^68. 458. Kunze, K.L., A.C Eddy, M. Gibaldi, and
446. Wester, M.R., CD. Stout, G. Schoch, J.K. Yano, W.F. Trager (1991). Metabolic enantiomeric inter-
and E.F. Johnson (2003). Crystallization of human actions: The inhibition of human (S)-warfarin-7-
P450 2C9. FASEB J. 17, A609. hydroxylase by (R)-warfarin. Chirality 3, 24-29.
484 F. Peter Guengerich

459. Rettie, A.E., K.R. Korzekwa, K.L. Kunze, Investigation vs Atenolol (SILVHIA) trial.
R.F. Lawrence, A.C. Eddy, T. Aoyama et al. (1992). J. Hypertens. 20, 2089-2093.
Hydroxylation of warfarin by human cDNA- 470. Yun, C.-H., H.S. Lee, H. Lee, J.K. Rho,
expressed cytochrome P-450: A role for P-4502C9 H.G. Jeong, and F.P Guengerich (1995). Oxidation of
in the etiology of (5)-warfarin-drug interactions. the angiotensin II receptor antagonist losartan (DuP
Chem. Res. Toxicol. 5, 54-59. 753) in human liver microsomes: Role of cytochrome
460. Rettie, A.E., L.C. Wienkers, F.J. Gonzalez, P450 3A(4) in formation of the active metabolite
W.F. Trager, and K.R. Korzekwa (1994). Impaired EXP 3174. DrugMetab. Dispos. 23, 285-289.
(iS)-warfarin metabolism catalysed by the R144C 471. Stearns, R.A., PK. Chakravarty, R. Chen, and
allelic variant of CYP2C9. Pharmacogenetics 4, S.H.L. Chiu (1995). Biotransformation of losartan
39^2. to its active carboxylic acid metabolite in human
461. Yamazaki, H., K. Inoue, and T. Shimada (1998). liver microsomes: Role of cytochrome P4502C
Roles of two allelic variants (Argl44Cys and and 3A subfamily members. Drug Metab. Dispos.
Ile359Leu) of cytochrome P4502C9 in the oxida- 23,207-215.
tion of tolbutamide and warfarin by human liver 472. Aithal, G.P, C.P Day, J.B. Leathart, and A.K. Daly
microsomes. Xenobiotica 28, 103-115. (2000). Relationship of polymorphism in CYP2C9
462. Scordo, M.G., V Pengo, E. Spina, M.L. Dahl, to genetic susceptibility to diclofenac-induced
M. Gusella, and R. Padrini (2002). Influence of hepatitis. Pharmacogenetics 10, 511-518.
CYP2C9 and CYP2C19 genetic polymorphisms 473. Martinez, C , E. Garcia-Martin, J.M. Ladero,
on warfarin maintenance dose and metabolic J. Sastre, F. Garcia-Gamito, M. Diaz-Rubio et al.
clearance. Clin. Pharmacol. Ther 72, 702-710. (2001). Association of CYP2C9 genotypes leading
463. Higashi, M.K., D.L. Veenstra, L.M. Kondo, to high enzyme activity and colorectal cancer risk.
A.K. Wittkowsky, S.L. Srinouanprachanh, Carcinogenesis 12, 1323-1326.
F.M. Farin et al. (2002). Association between 474. Yasar, U, E. Eliasson, and M.L. Dahl (2002).
CYP2C9 genetic variants and anticoagulation- Association of CYP2C9 genotypes leading to
related outcomes during warfarin therapy. J. Am. high enzyme activity and colorectal cancer risk.
Med. Assoc. 287, 1690-1698. Carcinogenesis 23, 665; author reply 667-668.
464. Tassies, D., C. Freire, J. Pijoan, S. Maragall, 475. Garcia-Martin, E., C. Martinez, J.M. Ladero,
J. Monteagudo, A. Ordinas et al. (2002). Pharma- F.J. Gamito, A. Rodriguez-Lescure, and
cogenetics of acenocoumarol: Cytochrome P450 J.A. Agundez (2002). Influence of cytochrome
CYP2C9 polymorphisms influence dose require- P450 CYP2C9 genotypes in lung cancer risk.
ments and stability of anticoagulation. Haemato- Cancer Lett. 180,41^6.
logicaSl, 1185-1191. 476. Minoletti, C , S. Dijols, PM. Dansette, and
465. Gentry, PR., C.E. Hack, L. Haber, A. Maier, and D. Mansuy (1999). Comparison of the substrate
H.J. Clewell, III (2002). An approach for the quan- specificities of human liver cytochrome P450s 2C9
titative consideration of genetic polymorphism and 2C18: Apphcation to the design of a specific
data in chemical risk assessment: Examples with substrate of CYP 2C18. Biochemistry 38,
warfarin and parathion. Toxicol. Sci. 70, 120-139. 7828-7836.
466. Lee, C.R., J.A. Pieper, A.L. Hinderliter, 477. Richardson, T.H., K.J. Griffin, F Jung, J.L. Raucy,
J.A. Blaisdell, and J.A. Goldstein (2002). Evaluation and E.F. Johnson (1997). Targeted antipeptide anti-
of cytochrome P4502C9 metabolic activity with bodies to cytochrome P450 2C18 based on epitope
tolbutamide in CYP2C91 heterozygotes. Clin. mapping of an inhibitory monoclonal antibody to
Pharmacol. Ther 72, 562-571. P450 2C5. Arch. Biochem. Biophys. 338, 157-164.
467. Shon, J.H., Y.R. Yoon, K.A. Kim, YC. Lim, 478. Zaphiropoulos, PG. (1999). RNA molecules con-
K.J. Lee, J.Y Park et al (2002). Effects of CYP2C19 taining exons originating from different members
and CYP2C9 genetic polymorphisms on the disposi- of the cytochrome P450 2C gene subfamily
tion of and blood glucose lowering response to tolbu- (CYP2C) in human epidermis and liver. Nucleic
tamide in humans. Pharmacogenetics 12, 111-119. Acids Res. 27, 2585-2590.
468. Miners, J. (2002). CYP2C9 polymorphism: Impact 479. Mace, K., E.D. Bowman, P Vautravers, PG. Shields,
on tolbutamide pharmacokinetics and response. C.C. Harris, and A.M. Pfeifer (1998). Characteri-
Pharmacogenetics 12, 91-92. sation of xenobiotic-metabolising enzyme expres-
469. Hallberg, R, J. Karlsson, L. Kurland, L. Lind, sion in human bronchial mucosa and peripheral lung
T. Kahan, K. Malmqvist et al (2002). The tissues. Eur J. Cancer 34, 914-920.
CYP2C9 genotype predicts the blood pressure 480. Mizugaki, M., M. Hiratsuka, Y Agatsuma,
response to irbesartan: Results from the Y Matsubara, K. Fujii, S. Kure et al (2000). Rapid
Swedish Irbesartan Left Ventricular Hypertrophy detection of CYP2C18 genotypes by real-time
Human P450s 485

fluorescence polymerase chain reaction. J. Pharm. tolbutamide hydroxylation by human liver micro-
Pharmacol. 52, 199-205. somes. DrugMetab. Dispos. 28, 354-359.
481. Zhu-Ge, J., Y.N. Yu, YL. Qian, and X. Li (2002). 493. Kaminsky, L.S., D.A. Dunbar, PR Wang,
Establishment of a transgenic cell line stably P. Beaune, D. Larrey, F.P, Guengerich et al. (1984).
expressing human cytochrome P450 2C18 and Human hepatic cytochrome P-450 composition as
identification of a CYP2C18 clone with exon 5 probed by in vitro microsomal metabolism of war-
missing. World J. Gastroenterol. 8, 888-892. farin. Drug Metab. Dispos. 12, 470-477.
482. Payne, VA., Y.T. Chang, and G.H. Loew (1999). 494. Wienkers, L.C., C.J. Wurden, E. Storch,
Homology modeling and substrate binding study K.L. Kunze, A.E. Rettie, and W F Trager (1996).
of human CYP2C18 and CYP2C19 enzymes. Formation of (^)-8-hydroxywarfarin in human
Proteins 37,204-211. liver microsomes: A new metabolic marker for the
483. Meier, U.T. and U.A. Meyer (1987). Genetic (5)-mephenytoin hydroxylase, P4502C19. Drug
polymorphism of human cytochrome P-450 (S)- Metab. Dispos. 24, 610-614.
mephenytoin 4-hydroxylase. Studies with human 495. Zhang, W, Y Ramamoorthy, R.F Tyndale,
autoantibodies suggest a functionally altered S.D. Glick, I.M. Maisonneuve, M.E. Kuehne et al.
cytochrome P-450 isozyme as cause of the genetic (2002). Metabolism of 18-methoxycoronaridine,
deficiency. Biochemistry 26, 8466-8474. an ibogaine analog, to 18-hydroxycoronaridine by
484. Wilkinson, G.R., F.P Guengerich, and R.A. Branch genetically variable CYP2C19. Drug Metab.
(1989). Genetic polymorphism of 5-mephenytoin Dispos. 30, 663-669.
hydroxylation. Pharmacol. Then 43, 53-76. 496. Ando, Y, E. Fuse, and WD. Figg (2002).
485. Yasumori, T., N. Murayama, Y Yamazoe, and Thalidomide metabolism by the CYP2C subfam-
R. Kato (1990). Polymorphism in hydroxylation of ily. Clin. Cancer Res. 8, 1964-1973.
mephenytoin and hexobarbital stereoisomers in 497. Yamazaki, H. andT Shimada (1997). Progesterone
relation to hepatic P-450 human-2. Clin. and testosterone hydroxylation by cytochromes
Pharmacol. Ther 47, 313-322. P450 2C19, 2C9, and 3A4 in human liver micro-
486. Kim, M.J., J.S. Bertino, Jr., A. Gaedigk, Y Zhang, somes. Arch. Biochem. Biophys. 346, 161-169.
E.M. Sellers, and A.N. Nafziger (2002). Effect of 498. Kappers, W.A., R.J. Edwards, S. Murray, and
sex and menstrual cycle phase on cytochrome A.R. Boobis (2001). Diazinon is activated by
P450 2C19 activity with omeprazole used as a bio- CYP2C19 in human liver. Toxicol. Appl.
marker. Clin. Pharmacol. Ther 12, 192-199. Pharmacol. Ill, 68-76.
487. Zhou, H.H., L.B. Anthony, A.J. Wood, and 499. Ibeanu, G.C., B.I. Ghanayem, R Linko, L. Li,
G.R. Wilkinson (1990). Induction of polymorphic E.G. Pedersen, and J.A. Goldstein (1996).
4'-hydroxylation of S-mephenytoin by rifampicin. Identification of residues 99, 220, and 221 of
Br I Clin. Pharmacol. 30, 471^75. human cytochrome P450 2C19 as key determi-
488. Blaisdell, J., H. Mohrenweiser, J. Jackson, nants of omeprazole hydroxylase activity. J. Biol.
S. Ferguson, S. Coulter, B. Chanas et al. (2002). Chem. 271, 12496-12501.
Identification and functional characterization of 500. Furuta, T, N. Shirai, M. Takashima, F Xiao,
new potentially defective alleles of human H. Hanai, K. Nakagawa et al. (2001). Effects of
CYP2C19. Pharmacogenetics 12, 703-711. genotypic differences in CYP2C19 status on cure
489. Desta, Z., X. Zhao, J.G. Shin, and D.A. Flockhart rates for Helicobacter pylori infection by dual
(2002). Clinical significance of the cytochrome therapy with rabeprazole plus amoxicillin.
P450 2C19 genetic polymorphism. Clin. Pharma- Pharmacogenetics 11, 341-348.
cokinet. 41, 913-958. 501. Furuta, T, N. Shirai, F Watanabe, S. Honda,
490. de Morals, S.M.F., G.R. Wilkinson, J. Blaisdell, K. Takeuchi, T. lida et al. (2002). Effect of cyto-
K. Nakamura, U.A. Meyer, and J.A. Goldstein chrome P4502C19 genotypic differences on cure
(1994). The major genetic defect responsible for rates for gastroesophageal reflux disease by lanso-
the polymorphism of ^S-mephenytoin metabolism prazole. Clin. Pharmacol. Ther 72, 453-460.
in humans. J. Biol. Chem. 269, 15419-15422. 502. Kita, T., T Sakaeda, N. Aoyama, T. Sakai,
491. Ferguson, R.J., S.M. De Morals, S. Benhamou, Y Kawahara, M. Kasuga et al. (2002). Optimal
C. Bouchardy, J. Blaisdell, G. Ibeanu et al. (1998). dose of omeprazole for CYP2C19 extensive
A new genetic defect in human CYP2C19: metabolizers in anti-Helicobacter pylori therapy:
Mutation of the initiation codon is responsible for Pharmacokinetic considerations. Biol. Pharm.
poor metabolism of S-mephenytoin. J. Pharmacol. Bull. 25, 923-927.
Exp. Ther 2H4, 356-361. 503. Kita, T, T Sakaeda, T. Baba, N. Aoyama,
492. Wester, M.R., J.M. Lasker, E.F. Johnson, and M. Kakumoto, Y Kurimoto et al. (2003). Different
J.L. Raucy (2000). CYP2C19 participates in contribution of CYP2C19 in the in vitro metabolism
486 F. Peter Guengerich

of three proton pump inhibitors. Biol. Pharm. Bull Elucidation of the genetic basis of the common
26, 386-390. 'intermediate metabolizer' phenotype for drug oxi-
504. Chau, T.K., S. Marakami, B. Kawai, K. Nasu, dation by CYP2D6. Pharmacogenetics 10,
T. Kubota, and A. Ohnishi (2000). Genotype analy- 577-581.
sis of the CYP2C19 gene in HCV-seropositive 516. Zanger, U.M., J. Fischer, S. Raimundo, T. Stuven,
patients with cirrhosis and hepatocellular carci- B.O. Evert, M. Schwab et al. (2001). Comprehen-
noma. Life Sci. 67, 1719-1724. sive analysis of the genetic factors determining
505. Roddam, P.L., S. Rollinson, E. Kane, E. Roman, expression and function of hepatic CYP2D6.
A. Moorman, R. Cartwright et al. (2000). Poor Pharmacogenetics 11, 573-585.
metabolizers at the cytochrome P450 2D6 and 517. Tyndale, R., T Aoyama, F Broly, T. Matsunaga,
2C19 loci are at increased risk of developing adult T. Inaba, W. Kalow et al. (1991). Identification of
acute leukaemia. Pharmacogenetics 10, 605-615. a new variant CYP2D6 allele lacking the codon
506. Treluyer, J.M., E. Jacqz-Aigrain, F. Alvarez, and encoding Lys-281: Possible association with the
T. Cresteil (1991). Expression of CYP2D6 in devel- poor metabolizer phenotype. Pharmacogenetics 1,
oping human liver. Eur. J. Biochem. 202, 583-588. 26-32.
507. Lo Guidice, J.M., D. Marez, N. Sabbagh, 518. Yu, A., B.M. Kneller, A.E. Rettie, and
M. LegrandAndreoletti, C. Spire, E. Alcaide et al. R.L. Haining (2002). Expression, purification,
(1997). Evidence for CYP2D6 expression in biochemical characterization, and comparative
human lung. Biochem. Biophys. Res. Commun. function of human cytochrome P450 2D6.1,
241, 79-85. 2D6.2, 2D6.10, and 2D6.17 allelic isoforms.
508. Siegle, I., R Fritz, K. Eckhardt, U.M. Zanger, and J. Pharmacol. Exp. Ther 303, 1291-1300.
M. Eichelbaum (2001). Cellular localization and 519. Fletcher, B., D.B. Goldstein, A.L. Bradman,
regional distribution of CYP2D6 mRNA and pro- M.E. Weale, N. Bradman, and M.G. Thomas
tein expression in human brain. Pharmacogenetics (2003). High-throughput analysis of informative
11,237-245. CYP2D6 compound haplotypes. Genomics 81,
509. Miksys, S., Y. Rao, E. Hoffmann, D.C. Mash, and 166-174.
R.F. Tyndale (2002). Regional and cellular expres- 520. Dahl, M.L., I. Johansson, L. Bertilsson,
sion of CYP2D6 in human brain: Higher levels in M. Ingelman-Sundberg, and F Sjoqvist (1995).
alcoholics. J. Neurochem. 82, 1376-1387. Ultrarapid hydroxylation of debrisoquine in a
510. Idle, J.R., A. Mahgoub, R. Lancaster, and Swedish population. Analysis of the molecular
R.L. Smith (1978). Hypotensive response to genetic basis. J. Pharmacol. Exp. Ther 174,
debrisoquine and hydroxylation phenotype. Life 516-520.
Sci. 11, 979-984. 521. Lundqvist, E., I. Johansson, and M. Ingelman-
511. Alvan, G., C. von Bahr, P. Seideman, and Sundberg (1999). Genetic mechanisms for dupli-
F Sjoqvist (1982). High plasma concentrations of cation and multiduplication of the human CYP2D6
p-receptor blocking drugs and deficient debriso- gene and methods for detection of duplicated
quine hydroxylation. Lancet i, 333. CYP2D6 genes. Gene 226, 327-338.
512. Evans, D.A.P, D. Harmer, D.Y Downham, 522. Lovlie, R., A.K. Daly, A. Molven, J.R. Idle, and
E.J. Whibley, J.R. Idle, J. Ritchie et al. (1983). The V.M. Steen (1996). Ultrarapid metabolizers of
genetic control of sparteine and debrisoquine debrisoquine: Characterization and PCR-based
metabolism in man with new methods of analysing detection of alleles with duplication of the
bimodal distributions. J. Med. Genet. 20, 321-329. CYP2D6 gene. FEBS Lett. 392, 30-34.
513. Skoda, R.C., F.J. Gonzalez, A. Demierre, and 523. Kohnke, M.D., E.U. Griese, D. Stosser, I. Gaertner,
U.A. Meyer (1988). Two mutant alleles of the and G. Barth (2002). Cytochrome P450 2D6 defi-
human cytochrome P-450dbl gene (P450C2D1) ciency and its clinical relevance in a patient treated
associated with genetically deficient metabolism with risperidone. Pharmacopsychiatry 35, 116-118.
of debrisoquine and other drugs. Proc. Natl. Acad. 524. Guengerich, FP. (2001). Common and uncommon
Sci. USA 85, 5240-5243. cytochrome P450 reactions related to metabolism
514. Gaedigk, A., R.R. Gotschall, N.S. Forbes, and chemical toxicity. Chem. Res. Toxicol. 14,
S.D. Simon, G.L. Keams, and J.S. Leeder (1999). 611-650.
Optimization of cytochrome P4502D6 (CYP2D6) 525. Wolff, T, L.M. Distlerath, M.T Worthington,
phenotype assignment using a genotyping algorithm J.D. Groopman, G.J. Hammons, FF Kadlubar et al.
based on allele frequency data. Pharmacogenetics (1985). Substrate specificity of human liver
9, 669-682. cytochrome P-450 debrisoquine 4-hydroxylase
515. Raimundo, S., J. Fischer, M. Eichelbaum, probed using immunochemical inhibition and
E.U. Griese, M. Schwab, and U.M. Zanger (2000). chemical modeling. Cancer Res. 45, 2116-2122.
Human P450s 487

526. Islam, S.A., C.R. Wolf, M.S. Lennard, and substrates reveals that CYP2D6 is a 5-methoxyin-
M.J.E. Sternberg (1991). A three-dimensional dolethylamine O-demethylase. Pharmacogenetics
molecular template for substrates of human 13,307-319.
cytochrome P450 involved in debrisoquine 4- 538. Yu, A.M., J.R. Idle, E.G. Byrd, K.W Krausz,
hydroxylation. Carcinogenesis 12, 2211-2219. A. Kupfer, and F.J. Gonzalez (2003). Regeneration
527. Strobl, G.R., S. von Kruedener, J. Stockigt, of serotonin from 5-methoxytryptamine by poly-
F.P. Guengerich, andT. Wolff (1993). Development morphic human CYP2D6. Pharmacogenetics 13,
of a pharmacophore for inhibition of human liver 173-181.
cytochrome P-450 2D6: Molecular modeling and 539. Koymans, L., N.P.E. Vermeulen, A. Baarslag, and
inhibition studies. J. Med. Chem. 36, 1136-1145. G. Donne-Op den Kelder (1993). A preliminary
528. Koymans, L., N.P.E. Vermeulen, S.A.B.E. van 3D model for cytochrome P450 2D6 constructed
Acker, J.M. te Koppele, J.J.P. Heykants, by homology model building. J. Comput.Aided
K. Lavrijsen et al. (1992). A predictive model for Mol.Des. 7,281-289.
substrates of cytochrome P450-debrisoquine 540. de Groot, M.J., N.RE. Vermeulen, J.D. Kramer,
(2D6). Chem. Res. Toxicol. 5, 211-219. F.A.A. van Acker, and G.M. Donne-Op den Kelder
529. de Groot, M.J., G.J. Bijloo, B.J. Martens, (1996). A three-dimensional protein model for
F.A.A. van Acker, and N.RE. Vermeulen (1997). A human cytochrome P450 2D6 based on the crystal
refined substrate model for human cytochrome structure of P450 101, P450 102, and P450 108.
P450 2D6. Chem. Res. Toxicol. 10, 4 1 ^ 8 . Chem. Res. Toxicol. 9, 1079-1091.
530. Upthagrove, A.L. and W.L. Nelson (2001). 541. Lewis, D.F.V, RJ. Eddershaw, RS. Goldfarb, and
Importance of amine pKa and distribution coeffi- M.H. Tarbit (1997). Molecular modeling of
cient in the metabolism of fluorinated propranolol cytochrome P4502D6 (CYP2D6) based on an
derivatives. Preparation, identification of metabo- alignment with CYP102: Structural studies on spe-
lite regioisomers, and metabolism by CYP2D6. cific CYP2D6 substrate metabolism. Xenobiotica
DrugMetah. Dispos. 29, 1377-1388. 27,319-340.
531. Miller, G.R, I.H. Hanna, Y. Nishimura, and 542. Modi, S., M.J. Paine, M.J. Sutcliffe, L.Y Lian,
F.P. Guengerich (2001). Oxidation of phenylethy- WU. Primrose, C.R. Wolfe/ al. Roberts (1996). A
lamine derivatives by cytochrome P450 2D6: model for human cytochrome P450 2D6 based on
The issue of substrate protonation in binding and homology modeling and NMR studies of substrate
catalysis. Biochemistry 40, 14215-14223. binding. Biochemistry 35, 4540-^550.
532. Grace, J.M., M.T. Kinter, and T.L. Macdonald 543. de Groot, M.J., M.J. Ackland, VA. Home,
(1994). Atypical metabolism of deprenyl and A.A. Alex, and B.C. Jones (1999). A novel
its enantiomer, (*S)-(+)-7V,a-dimethyl-A^-propyny- approach to predicting P450 mediated drug metab-
Iphenethylamine, by cytochrome P450 2D6. Chem. olism. CYP2D6 catalyzed A^dealkylation reac-
Res. Toxicol. 7, 286-290. tions and qualitative metabolite predictions using a
533. Niwa, T., Y. Yabusaki, K. Honma, N. Matsuo, combined protein and pharmacophore model for
K. Tatsuta, F. Ishibashi et al. (1998). Contribution CYP2D6. J. Med. Chem. 42, 4062-4070.
of human hepatic cytochrome P450 isoforms to 544. Kirton, S.B., C.A. Kemp, N.R Tomkinson,
regioselective hydroxylation of steroid hormones. S. St.-Gallay, and M.J. Sutcliffe (2002). Impact
Xenobiotica 28, 539-547. of incorporating the 2C5 crystal structure into
534. Hiroi, T, T. Chow, S. Imaoka, and Y Funae (2000). comparative models of cytochrome P450 2D6.
Catalytic specificity of each CYP2D isoform in rat Pro/ems 49, 216-231.
and human. Abstracts, 13th Int. Sympos. Microsomes 545. Venhorst, J., A.M. ter Laak, J.N. Commandeur,
and Drug Oxidations, 10-14 July, Stresa: p. 113. Y Funae, T. Hiroi, and N.R Vermeulen (2003).
535. Guengerich, EP, G.R Miller, I.H. Hanna, Homology modeling of rat and human cytochrome
M.V Martin, S. Leger, C. Black et al (2002). P450 2D (CYP2D) isoforms and computational
Diversity in the oxidation of substrates by rationalization of experimental ligand-binding
cytochrome P450 2D6. Lack of an obligatory role specificities. J. Med. Chem. 46, 74-86.
of aspartate 301-substrate electrostatic bonding. 546. Crespi, C.L., D.T. Steimel, B.W. Penman,
Biochemistry 41, 11025-11034. K.R. Korzekwa, P. Femandez-Salguero, J.T.M.
536. Martinez, C , J.A. Agundez, G. Gervasini, Buters et al. (1995). Comparison of substrate
R. Martin, and J. Benitez (1997). Tryptamine: A metabolism by wild type CYP2D6 protein and
possible endogenous substrate for CYP2D6. a variant containing methionine, not valine, at
Pharmacogenetics 7, 85-93. position 374. Pharmacogenetics 5, 234-243.
537. Yu, A.M., J.R. Idle, T. Herraiz, A. Kiipfer, and 547. Ellis, S.W, K. Rowland, M.J. Ackland, E. Rekka,
F.J. Gonzalez (2003). Screening for endogenous A.R Simula, M.S. Lennard et al. (1996). Influence
488 F. Peter Guengerich

of amino acid residue 374 of cytochrome P-450 NADPH-cytochrome P450 reductase. Biochemistry
2D6 (CYP2D6) on the regio- and enantio-selective 36, 4461-4470.
metaboUsm of metoprolol. Biochem. J. 316, 558. Hanna, LH., J.A. Krauser, H. Cai, M.-S. Kim, and
647-654. F.P. Guengerich (2001). Diversity in mechanisms
548. ElUs, S.W., G.P. Hayhurst, G. Smith, T. Lightfoot, of substrate oxidation by cytochrome P450 2D6.
M.M.S. Wong, A.R Simula et al (1995). Evidence Lack of an allosteric role of NADPH-cytochrome
that aspartic acid 301 is a critical substrate-contact P450 reductase in catalytic regioselectivity. J. Biol.
residue in the active site of cytochrome P450 2D6. Chem. 276,39553-39561.
J. Biol. Chem. 270, 29055-29058. 559. Guengerich, RP, G.P Miller, LH. Hanna, H. Sato,
549. Hanna, I.H., M.-S. Kim, and F.R Guengerich and M.V Martin (2002). Oxidation of
(2001). Heterologous expression of cytochrome methoxyphenethylamines by cytochrome P450
P450 2D6 mutants, electron transfer, and catalysis 2D6. Analysis of rate-limiting steps. J. Biol. Chem.
of bufiiralol hydroxylation. The role of aspartate 277,33711-33719.
301 in structural integrity. Arch. Biochem. Biophys. 560. Fukuda, T, Y Nishida, S. Imaoka, T Hiroi,
393,255-261. M. Naohara, Y Funae et al. (2000). The decreased
550. Paine, M.J., L.A. McLaughlin, J.U. Flanagan, in vivo clearance of CYP2D6 substrates by
C.A. Kemp, M.J. Sutcliffe, G.C. Roberts et al. CYP2D6* 10 might be caused not only by the low-
(2003). Residues glutamate 216 and aspartate 301 expression but also by low affinity of CYP2D6.
are key determinants of substrate specificity and Arch. Biochem. Biophys. 380, 303-308.
product regioselectivity in cytochrome P450 2D6. 561. Otton, S.V, T Inaba, and W Kalow (1984).
J. Biol. Chem. 278, 4021-4027. Competitive inhibition of sparteine oxidation in
551. Smith, G., S. Modi, 1. Pillai, L.-Y. Lian, human liver by p-adrenoceptor antagonists and
M.J. Sutcliffe, M.P Pritchard et al. (1998). other cardiovascular drugs. Life Sci. 34, 73-80.
Determinants of the substrate specificity of human 562. Palamanda, J.R., C.N. Casciano, L.A. Norton,
cytochrome P-450 CYP2D6: Design and construc- R.P Clement, L.V Favreau, C.C. Lin et al. (2001).
tion of a mutant with testosterone hydroxylase Mechanism-based inactivation of CYP2D6 by 5-
activity Biochem. J. 331, 783-792. fluoro-2-[4-[(2-phenyl-1 //-imidazol-5-yl)methyl]-
552. Wiseman, H. and D.F Lewis (1996). The metabo- 1 -piperazinyl]pyrimidine. Drug Metab. Dispos. 29,
lism of tamoxifen by human cytochromes P450 863-867.
is rationalized by molecular modelling of the 563. Gates, N.S., R.R. Shah, J.R. Idle, and R.L. Smith
enzyme-substrate interactions: Potential impor- (1983). Influence of oxidation polymorphism on
tance to its proposed anti-carcinogenic/carcino- phenformin kinetics and dynamics, Clin.
genic actions. Carcinogenesis 17, 1357-1360. Pharmacol. Then 34, 827-834.
553. Lightfoot, T, S.W Ellis, J. Mahling, M.J. 564. Gates, N.S., R.R. Shah, PL. Drury, J.R. Idle, and
Ackland, EE. Blaney, G.J. Bijloo et al. (2000). R.L. Smith (1982). Captopril-induced agranulocy-
Regioselectivity hydroxylation of debrisoquine by tosis associated with an impairment of debriso-
cytochrome P4502D6: Implications for active site quine hydroxylation. Br J. Pharmacol. 14, 60IP
modelling. Xenobiotica 30, 219-233. 565. Rau, T, R. Heide, K. Bergmann, H. Wuttke,
554. Ellis, S.W, G.P Hayhurst, T. Lightfoot, G. Smith, U. Werner, N. Feifel, and T. Eschenhagen (2002).
J. Harlow, K. Rowland-Yeo et al. (2000). Evidence Effect of the CYP2D6 genotype on metoprolol
that serine 304 is not a key ligand-binding residue metabolism persists during long-term treatment.
in the active site of cytochrome P450 2D6. Pharmacogenetics 12, 465^72.
Biochem. J. ^^5, 565-51 \. 566. Dome, J.L., K. Walton, W Slob, and A.G, Renwick
555. Hayhurst, G.P, J. Harlow, J. Chowdry, E. Gross, (2002), Human variability in polymorphic CYP2D6
E. Hilton, M.S. Lennard et al. (2001). Influence of metabolism: Is the kinetic default uncertainty factor
phenylalanine-481 substitutions on the catalytic adequate? Food Chem. Toxicol. 40, 1633-1656.
activity of cytochrome P450 2D6. Biochem. J. 355, 567. Chou, W.H., EX. Yan, J. de Leon, J, Bamhill,
373-379. T, Rogers, M, Cronin et al. (2000). Extension of a
556. Ramamoorthy, Y, R.F Tyndale, and E.M. Sellers pilot study: Impact from the cytochrome P450 2D6
(2001). Cytochrome P450 2D6.1 and cytochrome polymorphism on outcome and costs associated with
P450 2D6.10 differ in catalytic activity for multi- severe mental illness. J. Clin. Psychopharmacol. 20,
ple substrates. Pharmacogenetics 11, 477^87. 246-251.
557. Modi, S., D.E. Gilham, M.J. SutcHffe, L.-Y. Lian, 568. Dahl, M.L. (2002). Cytochrome P450 phenotyp-
WU. Primrose, C.R. Wolf e/a/. (1997). 1-Methyl- ing/genotyping in patients receiving antipsy-
4-phenyl-l,2,3,6-tetrahydropyridine as a substrate chotics: Useful aid to prescribing?. Clin.
of cytochrome P450 2D6: Allosteric effects of Pharmacokinet. 41, 453^70.
Human P450s 489

569. Scolnick, E.M. (2002). Discovery and develop- 582. Zanger, U.M., H.R Hauri, J. Loeper, J.C Homberg,
ment of antidepressants: A perspective from a and U.A. Meyer (1988). Antibodies against human
pharmaceutical discovery company. Biol. cytochrome P-450^^| in autoimmune hepatitis type
Psychiatry SI, 154-156. II. Proc. Natl. Acad. Sci. USA 85, 8256-2860.
570. Caporaso, N., R.B. Hayes, M. Dosemeci, R. Hoover, 583. Manns, M.P. (1991). Cytochrome P450 enzymes as
R. Ayesh, M. Hetzel et al. (1989). Lung cancer risk, human autoantigens. Immunol. Res. 10, 503-507.
occupational exposure, and the debrisoquine meta- 584. Manns, M.P, K.J. Griffin, K.F Sullivan, and
bolic phenotype. Cancer Res. 49, 3675-3679. E.F. Johnson (1991). LKM-1 autoantibodies recog-
571. Bouchardy, C , S. Benhamou, and R Dayer (1996). nize a short linear sequence in P450IID6, a
The effect of tobacco on lung cancer risk depends cytochrome P-450 monooxygenase. J. Clin. Invest.
on CYP2D6 activity Cancer Res. 56, 251-253. 88, 1370-1378.
572. Shaw, G.L., R.T. Falk, J.N. Frame, B. WeiflFenbach, 585. Loeper, J., V Descatoire, M. Maurice, P. Beaune,
J.V Nesbitt, H.I. Pass et al. (1998). Genetic poly- J. Belghiti, D. Houssin et al. (1993). Cytochromes
morphism of CYP2D6 and lung cancer risk. P-450 in human hepatocyte plasma membrane:
Cancer Epidemiol. Biomarkers Prev. 7, 215—219. Recognition by several autoantibodies.
573. Rostami-Hodjegan, A., M.S. Lennard, H.F. Woods, Gastroenterology 104, 203-216.
and G.T. Tucker (1998). Meta-analysis of studies 586. Vergani, D. (2000). LKM antibody: Getting in
of the CYP2D6 polymorphism in relation to lung some target practice. Gut 46, 449^50.
cancer and Parkinson's disease. Pharmacogenetics 587. Vitozzi, S., P. Lapierre, I. Djilali-Saiah, and
8, 227-238. F. Alvarez (2002). Autoantibody detection in type
574. Legrand-Andreoletti, M., I. Stucker, D. Marez, 2 autoimmune hepatitis using a chimera recombi-
R Galais, J. Cosme, N. Sabbagh et al. (1998). nant protein. J. Immunol. Meth. 262, 103-110.
Cytochrome P450 CYP2D6 gene polymorphism 588. Nolte, W, F Polzien, B. Sattler, G. Ramadori, and
and lung cancer susceptibility in Caucasians. H. Hartmann (1995). Recurrent episodes of acute
Pharmacogenetics 8, 7-14. hepatitis associated with LKM-1 (cytochrome
575. Christensen, P.M., PC. Gotzsche, and K. Brosen P450 2D6) antibodies in identical twin brothers.
(1997). The sparteine/debrisoquine (CYP2D6) J. Hepatol. 23, 734-739.
oxidation polymorphism and the risk of lung can- 589. Orme-Johnson, W.H. and D.M. Ziegler (1965).
cer: A meta-analysis. Eur J. Clin. Pharmacol. 51, Alcohol mixed function oxidase activity of
389-393. mammalian liver micoromes. Biochem. Biophys.
576. Fleming, CM., A. Kaisary, G.R. Wilkinson, Res. Commun. 21, 78—82.
R Smith, and R.A. Branch (1992). The ability to 590. Lieber, C S . and L.M. DeCarU (1970). Hepatic
4-hydroxylate debrisoquine is related to recurrence microsomal ethanol oxidizing system: In vitro
of bladder cancer. Pharmacogenetics 2, 128-134. chracteristics and adaptive properties in vivo.
577. Worrall, S.F, M. Corrigan, A. High, D. Starr, J. Biol. Chem. 245, 2505-2512.
C. Matthias, C.R. Wolfed al. (1998). Susceptibility 591. Teschke, R., Y Hasumura, and C S . Lieber (1974).
and outcome in oral cancer: Preliminary data Hepatic microsomal ethanol-oxidizing system:
showing an association with polymorphism in Solubilization, isolation and characterization.
cytochrome P450 CYP2D6. Pharmacogenetics 8, Arch. Biochem. Biophys. 163, 404-415.
433^*39. 592. Ryan, D.E., L. Ramanathan, S. lida, PE. Thomas,
578. Barbeau, A., M. Roy, S. Paris, T. Cloutier, M. Haniu, J.E. Shively et al (1985).
L. Plasse, and J. Poirier (1985). Ecogenetics of Characterization of a major form of rat hepatic
Parkinson's disease: 4-hydroxylation of debriso- microsomal cytochrome P-450 induced by isoni-
quine. Lancet ii, 1213-1215. azid. J. Biol. Chem. 260, 6385-6393.
579. Armstrong, M., A.K. Daly, S. Cholerton, 593. Wrighton, S.A., PE. Thomas, D.E. Ryan, and
D.N. Bateman, and J.R. Idle (1992). Mutant W. Levin (1987). Purification and characterization
debrisoquine hydroxylation genes in Parkinson's of ethanol-inducible human hepatic cytochrome
disease. Lfl^ce? 339, 1017-1018. P-450HLJ. Arch. Biochem. Biophys. 258, 292-297.
580. Harhangi, B.S., B.A. Oostra, R Heutink, CM. van 594. Umeno, M., O.W McBride, C.-S. Yang,
Duijn, A. Hofman, and M.M. Breteler (2001). H.V Gelboin, and FJ. Gonzalez (1988). Human
CYP2D6 polymorphism in Parkinson's disease: ethanol-inducible P450IIE1: Complete gene
The Rotterdam Study. Mov. Disord. 16, 290-293. sequence, promoter characterization, chromosome
581. Allam, M.F, A. Serrano del Castillo, and mapping, and cDNA-directed expression.
R. Fernandez-Crehuet Navajas (2002). Smoking Biochemistry 21, 9006-9013.
and Parkinson's disease: Explanatory hypothesis. 595. Guengerich, FP and CG. Turvy (1991).
Int. J. Neurosci. 112, 851-854. Comparison of levels of several human microsomal
490 F. Peter Guengerich

cytochrome P-450 enzymes and epoxide hydrolase P450 2E1 to the plasma membrane. J. Biol. Chem.
in normal and disease states using immunochemi- 275, 17130-17135.
cal analysis of surgical liver samples. J. Pharmacol. 607. Thomas, RE., S. Bandiera, S.L. Maines,
Exp. Then 256, 1189-1194. D.E. Ryan, and W. Levin (1987). Regulation of
596. Vieira, I., M. Sonnier, and T. Cresteil (1996). cytochrome P-450J, a high-affinity A^-nitrosodi-
Developmental expression of CYP2E1 in the methylamine demethylase, in rat hepatic micro-
human liver: Hypermethylation control of gene somes. Biochemistry 26, 2280-2289.
expression during the neonatal period. Eur. J. 608. Koop, D.R. and D.J. Tierney (1990). Multiple
Biochem. 238, 476-483. mechanisms in the regulation of ethanol-inducible
597. Ding, X. and L.S. Kaminsky (2003). Human extra- cytochrome P450IIE1. BioEssays 12, 429^35.
hepatic cytochromes P450: Function in xenobiotic 609. Ueno, T. and F.J. Gonzalez (1990). Transcriptional
metabolism and tissue-selective chemical toxicity control of the rat hepatic CYP2E1 gene. Mol. Cell
in the respiratory and gastrointestinal tracts. Annu. Biol. 10, 4495^505.
Rev. Pharmacol. Toxicol. 43, 149-173. 610. Woodcroft, K.J. and R.F Novak (1999). The role of
598. Warner, M. and J.A. Gustafsson (1994). Effect of phosphatidylinositol 3-kinase, Src kinase, and pro-
ethanol on cytochrome P450 in the rat brain. Proc. tein kinase A signaling pathways in insulin and
Natl. Acad. Sci. USA 91, 1019-1023. glucagon regulation of CYP2E1 expression.
599. Upadhya, S.C, RS. Tirumalai, M.R. Boyd, T. Mori, Biochem. Biophys. Res. Commun. 266, 304-307.
and V. Ravindranath (2000). Cytochrome P4502E 611. Siewert, E., R. Bort, R. Kluge, PC. Heinrich,
(CYP2E) in brain: Constitutive expression, induc- J. Castell, and R. Jover (2000). Hepatic
tion by ethanol and localization by fluorescence cytochrome P450 down-regulation during aseptic
in situ hybridization. Arch. Biochem. Biophys. 373, inflammation in the mouse is interleukin 6 depend-
23-34. ent. Hepatology 32, 49-55.
600. Kazakoff, K., P. Iversen, T. Lawson, J. Baron, 612. Lagadic-Gossmann, D , C. Lerche, M. Rissel,
F.P. Guengerich, and P. Pour (1994). Involvement F. Joannard, M. Galisteo, A. Guillouzo et al. (2000).
of cytochrome P450 2El-like isoform in the acti- The induction of the human hepatic CYP2E1 gene
vation of A^-nitrosobis(2-oxopropyl)amine in the by interleukin 4 is transcriptional and regulated by
rat nasal mucosa. Eur J. Cancer 30B, 179-185. protein kinase C. Cell Biol. Toxicol. 16, 221-233.
601. Norton, I.D., M.V Apte, RS. Haber, 613. Hakkola, J., Y Hu, and M. Ingelman-Sundberg (2003).
G.W. McCaughan, R.C. Pirola, and J.S. Wilson Mechanisms of down-regulation of CYP2E1 expres-
(1998). Cytochrome P4502E1 is present in rat pan- sion by inflammatory cytokines in rat hepatoma
creas and is induced by chronic ethanol adminis- cdls. J. Pharmacol. Exp. Then 304, 1048-1054.
tration. Gut 42, 426^30. 614. Kim, S.G. and R.F Novak (1990). Induction of rat
602. Larson, J.R., M.J. Coon, and T.D. Porter (1991). hepatic P450IIE1 (CYP 2E1) by pyridine:
Purification and properties of a shortened form Evidence for a role of protein synthesis in the
of cytochrome P-450 2E1: Deletion of the NH2- absence of transcriptional activation. Biochem.
terminal membrane-insertion signal peptide does Biophys. Res. Commun. 166, 1072-1079.
not alter the catalytic activities. Proc. Natl. Acad. 615. Kocarek, T.A., R.C. Zangar, and R.F Novak
Sci. USA ^^,9\4\-9U5. (2000). Post-transcriptional regulation of rat
603. Gillam, E.M.J., Z. Guo, and F.R Guengerich CYP2E1 expression: Role of CYP2E1 mRNA
(1994). Expression of modified human untranslated regions in control of translational effi-
cytochrome P450 2E1 in Escherichia coli, purifi- ciency and message stability. Arch. Biochem.
cation, and spectral and catalytic properties. Arch. Biophys. 376, 180-190.
Biochem. Biophys. 312, 59-66. 616. Roberts, B.J., B.J. Song, Y Soh, S.S. Park, and
604. Neve, E.RA. and M. Ingelman-Sundberg (1999). S.E. Shoaf (1995). Ethanol induces CYP2E1 by
A soluble NH2-terminally truncated catalytically protein stabilization: Role of ubiquitin conjugation
active form of rat cytochrome P450 2E1 targeted to in the rapid degradation of CYP2E1. J. Biol. Chem.
liver mitochondria. FEBS Lett. 460, 309-314. 270, 29632-29635.
605. Robin, M.A., H.K. Anandatheerthavarada, J.K. Fang, 617. Yang, M.X. and A.I. Cederbaum (1997).
M. Cudic, L. Otvos, and N.G. Avadhani (2001). Characterization of cytochrome P4502E1 turnover
Mitochondrial targeted cytochrome P450 2E1 (P450 in transfected HepG2 cells expressing human
MT5) contains an intact N-terminus and requires CY?2E\.Arch. Biochem. Biophys. 341, 25-33.
mitochondrial specific electron transfer proteins 618. Emery, M.G., C. Jubert, K.E. Thummel, and
for activity J. Biol. Chem. 276, 24680-24689. E.D. Kharasch (1999). Duration of cytochrome P-
606. Neve, E.R and M. Ingelman-Sundberg (2000). 450 2E1 (CYP2E1) inhibition and estimation of
Molecular basis for the transport of cytochrome functional CYP2E1 enzyme half-life after
Human P450s 491

single-dose disulfiram administration in humans. (CYP2E1). Biochem. Biophys. Res. Commun. 179,
J. Pharmacol. Exp. Ther. 291, 213-219. 689-694.
619. Chien, J.Y., K.E. Thummel, and J.T. Slattery 631. Kunitoh, S., S. Imaoka, T. Hiroi, Y. Yabusaki,
(1997). Pharmacokinetic consequences of induc- T. Monna, and Y. Funae (1997). Acetaldehyde as
tion of CYP2E1 by ligand stabilization. Drug well as ethanol is metabolized by human CYP2E1.
Metab. Dispos. 25, 1165-1175. J. Pharmacol. Exp. Ther 280, 527-532.
620. Watanabe, J., S. Hayashi, and K. Kawajiri (1994). 632. Bell-Parikh, L.C. and EP Guengerich (1999).
Different regulation and expression of the human Kinetics of cytochrome P450 2E1-catalyzed oxida-
CYP2E1 gene due to the Rsal polymorphism in tion of ethanol to acetic acid via acetaldehyde.
the 5'-flanking region. J. Biochem. (Tokyo) 116, J. Biol. Chem. 274, 23833-23840.
321-326. 633. Tassaneeyakul, W, M.E. Veronese, D.J. Birkett,
621. Fairbrother, K.S., J. Grove, I. de Waziers, F.J. Gonzalez, and J.O. Miners (1993). Validation
D.T. Steimel, C.P Day, C.L. Crespi et al. (1998). of 4-nitrophenol as an in vitro substrate probe
Detection and characterization of novel polymor- for human liver CYP2E1 using cDNA expression
phisms in the CYP2E1 gene. Pharmacogenetics 8, and microsomal kinetic techniques. Biochem.
543-552. Pharmacol. 46, 1975-1981.
622. Fritsche, E., G.S. Pittman, and D.A. Bell (2000). 634. Peter, R., R.G. Bocker, PH. Beaune, M. Iwasaki,
Localization, sequence analysis, and ethnic distri- EP Guengerich, and C.-S. Yang (1990). Hydro-
bution of a 96-bp insertion in the promoter of the xylation of chlorzoxazone as a specific probe for
human CYP2E1 gene. Mutat. Res. 432, 1-5. human liver cytochrome P-450 IIEl. Chem. Res.
623. Powell, H., N.R. Kitteringham, M. Pirmohamed, Toxicol. 3, 566—573.
D.A. Smith, and B.K. Park (1998). Expression of 635. Yamazaki, H., Z. Guo, and EP Guengerich (1995).
cytochrome P4502E1 in human liver: Assessment Selectivity of cytochrome P450 2E1 in chlorzoxazone
by mRNA, genotype and phenotype. Pharma- 6-hydroxylation. Drug Metab. Dispos. 23, 438^40.
cogenetics 8, 411-^21. 636. Preussmann, R. and B.W Stewart (1984). A^-Nitroso
624. Inoue, K., H. Yamazaki, and T. Shimada (2000). carcinogens. In C.E. Searle (ed.). Chemical
Characterization of liver microsomal 7-ethoxy- Carcinogens, Vol. 2. American Chemical Society,
coumarin O-deethylation and chlorzoxazone 6- Washington, D. C , pp. 643-828.
hydroxylation activities in Japanese and Caucasian 637. Argus, M.E, J.C. Arcos, K.M. Pastor, B.C. Wu,
subjects genotyped for CYP2E1 gene. Arch. and N. Venkatesan (1976). Dimethylnitrosamine-
Toxicol. 74, 372-378. demethylase: Absence of increased enzyme
625. O'Shea, D., S.N. Davis, R.B. Kim, and catabolism and multiplicity of effector sites in
G.R. Wilkinson (1994). Effect of fasting and obe- repression. Hemoprotein involvement. Chem. Biol.
sity in humans on the 6-hydroxylation of chlorox- Interact. 13, 127-140.
azone: A putative probe of CYP2E1 activity. Clin. 638. Lake, B.G., C.E. Heading, J.C. Phillips,
Pharmacol. Then 56, 359-367. S.D. Gangolh, and A.G. Lloyd (1974). Some studies
626. Koop, D.R. and J.R Casazza (1985). Identification on the metabolism in vitro of dimethylnitrosamine
of ethanol-inducible P-450 isozyme 3a as the ace- by rat liver. Biochem. Soc. Transact. 2, 610-612.
tone and acetol monooxygenase of rabbit micro- 639. Levin, W, PE. Thomas, N. Oldfield, and
somes. J. Biol. Chem. 260, 13607-13612. D.E. Ryan (1986). 7V-Demethylation of 7V-nitroso-
627. Bondoc, EY, Z. Bao, W.Y Hu, EJ. Gonzalez, dimethylamine catalyzed by purified rat hepatic
Y Wang, C.S. Yang et al. (1999). Acetone cataboUsm microsomal cytochrome P-450: Isozyme speci-
by cytochrome P450 2E1: Studies with CYP2E1- ficity and role of cytochrome b^. Arch. Biochem.
nuU mice. Biochem. Pharmacol. 105, 83-88. Biophys. 248, 158-165.
628. Kashiwagi, T., S. Ji, J.J. Lemasters, and 640. Wrighton, S.A., PE. Thomas, D.T. Molowa,
R.G. Thurman (1982). Rates of alcohol dehydro- M. Haniu, J.E. Shively, S.L. Maines et al. (1986).
genase-dependent ethanol metabolism in peripor- Characterization of ethanol-inducible human liver
tal and pericentral regions of the perfused rat liver. A^-nitrosodimethylamine demethylase. Biochemistry
Mol. Pharmacol. 21, 438^43. 25,6731-6735.
629. Kono, H., B.U. Bradford, M. Yin, K.K. Sulik, 641. Bastien, M.C. and J.R Villeneuve (1998).
D.R. Koop, J.M. Peters et al. (1999). CYP2E1 is Characterization of cytochrome P450 2E1 activity
not involved in early alcohol-induced liver injury. by the [^"^CJnitrosodimethylamine breath test.
Am. J. Physiol. Ill, G1259-G1267. Can. J. Physiol. Pharmacol. 76, 756-763.
630. Terelius, Y, C. Norsten-Hoog, T. Cronholm, and 642. Raucy, J.L., J.C. Kraner, and J.M. Lasker (1993).
M. Ingelman-Sundberg (1991). Acetaldehyde as a Bioactivation of halogenated hydrocarbons by
substrate for ethanol-inducible cytochrome P450 cytochrome P4502E1. Crit. Rev. Toxicol. 23, 1-20.
492 F. Peter Guengerich

643. Hong, J.Y., C.S. Yang, M. Lee, Y.Y. Wang, mutagenicity of dialkylnitrosamines in methyl-
W. Huang, Y. Tan et ah (1997). Role of cytochromes transferase-deficient strains of Salmonella
P450 in the metabolism of methyl tert-\miy\ ether typhimurium. Mutat. Res. 484, 61-68.
in human livers. Arch. Toxicol. 71, 266-269. 655. Schenkman, LB. and I. Jansson (2003). The many
644. Wang, H., B. Chanas, and B.I. Ghanayem (2002). roles of cytochrome by Pharmacol. Ther 97,
Cytochrome P450 2E1 (CYP2E1) is essential for 139-152.
acrylonitrile metabolism to cyanide: Comparative 656. Tan, Y, S.P White, S.R. Paranawithana, and
studies using CYP2El-null and wild-type mice. C.S. Yang (1997). A hypothetical model for the
DrugMetab. Dispos. 30, 911-917. active site of human cytochrome P4502E1.
645. Hoffler, U., H.A. El-Masri, and B.I. Ghanayem Xenobiotica 27, 287-299.
(2003). Cytochrome P450 2E1 (CYP2E1) is the prin- 657. Lewis, D.F, B.G. Lake, M.G. Bird, G.D. Loizou, M.
cipal enzyme responsible for urethane metabolism: Dickins, and PS. Goldfarb (2003). Homology mod-
Comparative studies using CYP2El-null and wild- elling of human CYP2E1 based on the CYP2C5
type mice. J. Pharmacol Exp. Ther. 305, 557-564. crystal structure: Investigation of enzyme-substrate
646. Clarke, S.E., S.J. Baldwin, J.C. Bloomer, and enzyme-inhibitor interactions. Toxicol. In Vitro
A.D. Ayrton, R.S. Sozio, and R.J. Chenery (1994). 17, 93-105.
Laurie acid as a model substrate for the simultaneous 658. Smith, S.V, A.P Koley, R. Dai, R.C. Robinson,
determination of cytochrome P450 2E1 and 4A in H. Leong, A. Markowitz et al. (2000).
hepatic microsomes. Chem. Res. Toxicol. 7, 836-842. Conformational modulation of human cytochrome
647. Castle, PL, J.L. Merdink, LR. Okita, P450 2E1 by ethanol and other substrates: A CO
S.A. Wrighton, and R.T. Okita (1995). Human flash photolysis study. Biochemistry 39, 5731-5737.
liver lauric acid hydroxylase activities. Drug 659. Yin, H., M.W. Anders, K.R. Korzekwa, L. Higgins,
Metab. Dispos. 23, 1037-1043. K.E. Thummel, E.D. Kharasch et al. (1995).
648. Gillam, E.M.L, A.M. Aguinaldo, L.M. Notley, Designing safer chemicals: Predicting the rates of
D. Kim, R.G. Mundkowski, A. Volkov et al. metabolism of halogenated alkanes. Proc. Natl.
(1999). Formation of indigo by recombinant mam- Acad. Sci. USA 92, 11076-11080.
malian cytochrome P450. Biochem. Biophys. Res. 660. Lewis, D.F, C. Sams, and G.D. Loizou (2003).
Commun. 265, 469-472. A quantitative structure-activity relationship
649. Adachi, J., Y. Mori, S. Matsui, H. Takigami, analysis on a series of alkyl benzenes metabolized
L Fujino, H. Kitagawa et al. (2001). Indirubin and by human cytochrome P450 2E1. J. Biochem. Mol.
indigo are potent aryl hydrocarbon receptor lig- Toxicol. 17, 47-52.
ands present in human urine. J. Biol. Chem. 276, 661. Keefer, L.K., W. Lijinsky, and H. Garcia (1973).
31475-31478. Deuterium isotope effect on the carcinogenicity of
650. Spracklin, D.K., D.C. Hankins, J.M. Fisher, dimethylnitrosamine in rat liver. J. Natl. Cancer
K.E. Thummel, and E.D. KJiarasch (1997). Inst. 51, 299-302.
Cytochrome P450 2E1 is the principal catalyst of 662. Wade, D., C.S. Yang, C.J. Metral, LM. Roman,
human oxidative halothane metabolism in vitro. J.A. Hrabie, C.W. Riggs et al. (1987). Deuterium
J. Pharmacol. Exp. Ther 281, 400-411. isotope effect on denitrosation and demethylation
651. Kharasch, E.D., D.C. Hankins, and K. Cox (1999). of A^-nitrosodimethylamine by rat liver micro-
Clinical isoflurane metabolism by cytochrome somes. Cancer Res. 47, 3373-3377.
P450 2E\. Anesthesiology 90, 766-771. 663. Yang, C.S., H. Ishizaki, M. Lee, D. Wade, and
652. Bell, L.C. and FP Guengerich (1997). Oxidation A. Fadel (1991). Deuterium isotope effect in the
kinetics of ethanol by human cytochrome P450 interaction of A^-nitrosodimethylamine, ethanol,
2E1. Rate-limiting product release accounts for and related compounds with cytochrome
effects of isotopic hydrogen substitution and P-450IIE1. Chem. Res. Toxicol. 4, 408-413.
cytochrome b^ on steady-state kinetics. J. Biol. 664. Calcutt, W and FP Guengerich (2003). Kinetic
C/^ew. 272, 29643-29651. isotope effects in dialkylnitrosamine dealkylations
653. Yamazaki, H., M. Nakano, E.M.J. Gillam, catalyzed by human cytochrome P450s 2A6 and
L.C. Bell, RP Guengerich, andT. Shimada (1996). lEX.FASEBJ. 17,A1325.
Requirements for cytochrome b^ in the oxidation 665. Reitz, R.H., A. Mendrala, and FP Guengerich
of 7-ethoxycoumarin, chlorzoxazone, aniline, and (1989). In vitro metabolism of methylene chloride
A^-nitrosodimethylamine by recombinant cyto- in human and animal tissues: Use in physiologi-
chrome P450 2E1 and by human liver microsomes. cally-based pharmacokinetic models. Toxicol.
Biochem. Pharmacol. 52, 301-309. Appl. Pharmacol. 97, 230-246.
654. Cooper, M.T. and T.D. Porter (2001). Cytochrome 666. Pernecky, S.I, TD. Porter, and M.L Coon (1990).
b^ coexpression increases the CYP2E1-dependent Expression of rabbit cytochrome P-450IIE2 in
Human P450s 493

yeast and stabilization of the enzyme by 4- major antigen in autoantibody formation among
methylpyrazole. Biochem. Biophys. Res. Commun. alcoholics. Gastroenterology 111, 206-216.
Ill, 1331-1337. 679. Bourdi, M., W Chen, R.M. Peter, J.L. Martin,
667. Koop, D.R. (1990). Inhibition of ethanol-inducible J.T.M. Buters, S.D. Nelson et al. (1996). Human
cytochrome P450IIE1 by 3-amino-l, 2,4-triazole. cytochrome P450 2E1 is a major autoantigen asso-
Chem. Res. Toxicol. 3, 377-383. ciated with halothane hepatitis. Chem. Res.
668. Hultmark, D., K. Sundh, L. Johansson, and Toxicol. 9, 1159-1166.
E. Arrhenius (1979). Ethanol inhibition of vinyl 680. Ekstrom, G. and M. Ingelman-Sundberg (1989).
chloride metabolism in isolated rat hepatocytes. Rat liver microsomal NADPH-supported oxidase
Chem. Biol. Interact. 25, 1-6. activity and lipid peroxidation dependent on
669. Wong, L.C.K., J.M. Winston, C.B. Hong, and ethanol-inducible cytochrome P-450 (P-450IIE1).
H. Plotnick (1982). Carcinogenicity and toxicity of Biochem. Pharmacol. 38, 1313-1319.
1,2-dibromoethane in the rat. Toxicol Appl. 681. Nieto, N., S.L. Friedman, P. Greenwel, and
Pharmacol. 63, 155-165. A.I. Cederbaum (1999). CYP2E1-mediated oxida-
670. Kwak, M.K., S.G. Kim, J.Y. Kwak, R.F. Novak, tive stress induces collagen type I expression in rat
and N.D. Kim (1994). Inhibition of cytochrome hepatic stellate cells. Hepatology 30, 987-996.
P4502E1 expression by organosulfur compounds 682. Cederbaum, A.L, D. Wu, M. Mari, and J. Bai
allylsulfide, allylmercaptan and allylmethylsulfide (2001). CYP2E1-dependent toxicity and oxidative
in rats. Biochem. Pharmacol. 47, 531-539. stress in HepG2 cells. Free Radic. Biol. Med. 31,
671. Nakajima, M., R. Yoshida, N. Shimada, 1539-1543.
H. Yamazaki, and T. Yokoi (2001). Inhibition and 683. Wan, J., L Shi, L. Hui, D. Wu, X. Jin, N. Zhao et al.
inactivation of human cytochrome P450 isoforms (2002). Association of genetic polymorphisms in
by phenethyl isothiocyanate. Drug Metab. Dispos. CYP2E1, MPO, NQOl, GSTMl, and GSTTl
29,1110-1113. genes with benzene poisoning. Environ. Health
672. Lucas, D., C. Farez, L.G. Bardou, J. Vaisse, Perspect. 110, 1213-1218.
J.R. Attali, and R Valensi (1998). Cytochrome 684. Uematsu, R, H. Kikuchi, M. Motomiya, T. Abe,
P450 2E1 activity in diabetic and obese patients I. Sagami, T. Ohmachi et al. (1991). Association
as assessed by chlorzoxazone hydroxylation. between restriction fragment length polymorphism
Fundam. Clin. Pharmacol. 12, 553-558. of the human cytochrome P450IIE1 gene and sus-
673. Le Marchand, L., G.R. Wilkinson, and L.R. Wilkens ceptibility to lung cancer. Jpn. J. Cancer Res. 82,
(1999). Genetic and dietary predictors of CYP2E1 254-256.
activity: A phenotyping study in Hawaii Japanese 685. Hirvonen, A., K. Husgafvel-Pursiainen, S. Anttila,
using chlorzoxazone. Cancer Epidemiol. Biomarkers A. Karjalainen, M. Sorsa, and H. Vainio (1992).
Prev. 8, 495-500. Metabolic cytochrome P450 genotypes and assess-
674. Morimoto, M., A.L. Hagbjork, A.A. Nanji, ment of individual susceptibility to lung cancer.
M. Ingelman-Sundberg, K.O. Lindros, P.C. Fu et al. Pharmacogenetics 2, 259-263.
(1993). Role of cytochrome P4502E1 in alcoholic 686. Ingelman-Sundberg, M., I. Johansson, I. Persson,
liver disease pathogenesis. Alcohol 10, 459^64. Q.Y Yue, M.L. Dahl, L. Bertilsson et al. (1992).
675. Morgan, K., S.W. French, andT.R. Morgan (2002). Genetic polymorphism of cytochromes P450:
Production of a cytochrome P450 2E1 transgenic Interethnic differences and relationship to inci-
mouse and initial evaluation of alcoholic liver dence of lung cancer. Pharmacogenetics 2,
damage. Hepatology 36, 122-134. 264-271.
676. Koop, D.R., B. Klopfenstein, Y. limuro, and 687. Persson, L, I. Johansson, H. Bergling, M.L. Dahl,
R.G. Thurman (1997). Gadolinium chloride blocks J., Seidegard, R. Rylander et al. (1993). Genetic
alcohol-dependent liver toxicity in rats treated polymorphism of cytochrome P4502E1 in a
chronically with intragastric alcohol despite the Swedish population: Relationship to incidence of
induction of CYP2E1. Mol. Pharmacol. 51, lung cancer. FEBS Lett. 319, 207-211.
944-950. 688. London, S.J., A.K. Daly, J. Cooper, C.L. Carpenter,
677. Lytton, S.D., A. Helander, Z.Q. Zhang-Gouillon, WC. Navidi, L. Ding et al. (1996). Lung cancer risk
K. Stokkeland, R. Bordone, S. Arico et al. (1999). in relation to the CYP2E1 Rsal genetic polymor-
Autoantibodies against cytochromes P-4502E1 phism among African-Americans and Caucasians
and P-4503A in alcoholics. Mol. Pharmacol. 55, in Los Angeles County. Pharmacogenetics 6,
223-233. 151-158.
678. Clot, P., E. Albano, E. Eliasson, M. Tabone, 689. Kato, S., PG. Shields, N.E. Caporaso,
S. Arico, Y. Israel et al. (1996). Cytochrome H. Sugimura, G.E. Trivers, M.A. Tucker et al
P4502E1 hydroxyethyl radical adducts as the (1994). Analysis of cytochrome P450 2E1 genetic
494 F. Peter Guengerich

polymorphisms in relation to human lung cancer. 700. Wu, S., C.R. Moomaw, K.B. Tomer, J.R. Falck, and
Cancer Epidemiol. Biomarkers Prev. 3, 515-518. D.C. Zeldin (1996). Molecular cloning and expres-
690. Itoga, S., F. Nomura, Y. Makino, T. Tomonaga, sion of CYP2J2, a human cytochrome P450
H. Shimada, T. Ochiai et al. (2002). Tandem repeat arachidonic acid epoxygenase highly expressed in
polymorphism of the CYP2E1 gene: An associa- heart. J. Biol. Chem. Ill, 3460-3468.
tion study with esophageal cancer and lung cancer 701. Zeldin, D.C, J. Foley, J. Ma, IE. Boyle,
Alcohol. Clin. Exp. Res. 26, 15S-19S. J.M.S. Pascual, C.R. Moomaw et al (1996). CYP2J
691. Bouchardy, C , A. Hirvonen, C. Coutelle, P.J. Ward, subfamily P450s in the lung: Expression, localiza-
P. Dayer, and S. Benhamou (2000). Role of alcohol tion, and potential functional significance. Mol
dehydrogenase 3 and cytochrome P-4502E1 geno- Pharmacol. SQ, 1111-1117.
types in susceptibility to cancers of the upper 702. Zeldin, D.C, J. Foley, S.M. Goldsworthy,
aerodigestive tract. Int. J. Cancer 87, 734-740. M.E. Cook, J.E. Boyle, J. Ma et al. (1997). CYP2J
692. Liu, S., J.Y. Park, S.P Schantz, J.C. Stern, and subfamily cytochrome P450s in the gastrointesti-
P Lazarus (2001). Elucidation of CYP2E1 5' reg- nal tract: Expression, localization, and potential
ulatory RsaVPstl allelic variants and their role in functional significance. Mol. Pharmacol. 51,
risk for oral cancer Oral Oncol. 37, 437^45. 931-943.
693. Kato, S., M. Onda, N. Matsukura, A. Tokunaga, 703. Zeldin, D.C. (2001). Epoxygenase pathways of
T. Tajiri, D.Y Kim et al. (1995). Cytochrome arachidonic acid metabolism. J. Biol. Chem. 276,
P4502E1 (CYP2E1) genetic polymorphism in a 36059-36062.
case-control study of gastric cancer and liver dis- 704. King, L.M., J. Ma, S. Srettabunjong, J. Graves, J.A.
ease. Pharmacogenetics 5, S141-S144. Bradbury, L. Li et al. (2002). Cloning of CYP2J2
694. Wong, R.H., C.L. Du, J.D. Wang, C.C. Chan, gene and identification of functional polymor-
J.C. Luo, and T.J. Cheng (2002). XRCCl and phisms. Mol. Pharmacol. 61, 840-852.
CYP2E1 polymorphisms as susceptibility factors 705. Nelson, D.R. (2003). Comparison of P450s from
of plasma mutant p53 protein and anti-p53 anti- human and fugu: 420 million years of vertebrate
body expression in vinyl chloride monomer- P450 evolution. Arch. Biochem. Biophys. 409,
exposed polyvinyl chloride workers. Cancer 18-24.
Epidemiol. Biomarkers Prev. 11, 475^82. 706. Rylander, T, E.P.A. Neve, M. Ingelman-Sundberg,
695. Nhamburo, PT, S. Kimura, O.W. McBride, and M. Oscarson (2001). Identification and tissue
C.A. Kozak, H.V Gelboin, and F.J. Gonzalez (1990). distribution of the novel human cytochrome P450
The human CYP2F gene subfamily: Identification 2S1 (CYP2S1). Biochem. Biophys. Res. Commun.
of a cDNA encoding a new cytochrome P450, 281,529-535.
cDNA-directed expression, and chromosome 707. Molowa, D.T, E.G. Schuetz, S.A. Wrighton,
mapping. Biochemistry 29, 5491-5499. PB. Watkins, P. Kremers, G. Mendez-Picon et al.
696. Czerwinski, M., TL. McLemore, R.M. Philpot, (1986). Complete cDNA sequence of a
PT. Nhamburo, K. Korzekwa, H.V Gelboin et al. cytochrome P-450 inducible by glucocorticoids in
(1991). Metabolic activation of 4-ipomeanol by human liver Proc. Natl. Acad. Sci. USA 83,
complementary DNA-expressed human cytochro- 5311-5315.
mes P-450: Evidence for species-specific metabo- 708. Beaune, PH., D.R. Umbenhauer, R.W. Bork,
lism. Cancer Res. 51, 4636-4638. R.S. Lloyd, and FP Guengerich (1986). Isolation
697. Lanza, D.L., E. Code, C.L. Crespi, F.J. Gonzalez, and sequence determination of a cDNA clone
and G.S. Yost (1999). Specific dehydrogenation of related to human cytochrome P-450 nifedipine oxi-
3-methylindole and epoxidation of naphthalene dase. Proc. Natl. Acad. Sci. USA 83, 8064-8068.
by recombinant human CYP2F1 expressed in 709. Bork, R.W, T Muto, PH. Beaune, PK. Srivastava,
lymphoblastoid cells. Drug Metab. Dispos. 27, R.S. Lloyd, and FP Guengerich (1989).
798-803. Characterization of mRNA species related to
698. Nakajima, T, E. Elovaara, F.J. Gonzalez, human liver cytochrome P-450 nifedipine oxidase
H.V Gelboin, H. Raunio, O. Pelkonen et al. and the regulation of catalytic activity. J. Biol.
(1994). Styrene metabolism by cDNA-expressed Chem. 264, 9\0~9\9.
human hepatic and pulmonary cytochromes P450. 710. Kolars, J., P Schmiedelin-Ren, W Dobbins,
Chem. Res. Toxicol. 7, 891-896. R. Merion, S. Wrighton, and P Watkins (1990).
699. Carr, B.A., J. Wan, R.N. Hines, and G.S. Yost Heterogeneity of P-450 III A expression in human
(2003). Characterization of the human lung gut epithelia. FASEB J. 4, A2242.
CYP2F1 gene and identification of a novel lung- 711. Kolars, J.C, P Schmiedlin-Ren, J.D. Schuetz,
specific binding motif. J. Biol. Chem. 278, C Fang, and PB. Watkins (1992). Identification of
15473-15483. rifampin-inducible P450IIIA4 (CYP3A4) in
Human P450s 495

human small bowel enterocytes. J. Clin. Invest. 90, 723. Hughes, S.J., M.A. Morse, CM. Weghorst,
1871-1878. H. Kim, RB. Watkins, RR Guengerich et al.
712. Bocker, R.H. and F.R Guengerich (1986). (1999). Cytochromes P450 are expressed in prolif-
Oxidation of 4-aryl- and 4-alkyl-substituted erating cells in Barrett's metaplasia. Neoplasia 1,
2,6-dimethyl-3,5-bis(alkoxycarbonyl)-l,4-dihy- 145-153.
dropyridines by human liver microsomes and 724. Inoue, K., J. Inazawa, H. Nakagawa, T. Shimada,
immunochemical evidence for the involvement H. Yamazaki, RR Guengerich et al. (1992).
of a form of C3^ochrome P-450. J. Med. Chem. 29, Assignment of the human cytochrome P450
1596-1603. nifedipine oxidase gene (CYP3A4) to band 7q22.1
713. Waxman, D.J., C. Attisano, F.R Guengerich, and by in situ fluorescence hybridization. Jpn. J. Hum.
D.R Lapenson (1988). Cytochrome P-450 steroid Genet. 37, 133-138.
hormone metabolism catalyzed by human liver 725. Lu, A.Y.H., A. Somogyi, S. West, R. Kuntzman,
microsomes. Arch. Biochem. Biophys. 263, and A.H. Conney (1972). Pregnenolone-16a-
424-436. carbonitrile: A new type of inducer of drug-metab-
714. Shimada, T. and RR Guengerich (1989). Evidence olizing enzymes. Arch. Biochem. Biophys. 152,
for C3^ochrome P-450^p, the nifedipine oxidase, 457-462.
being the principal enzyme involved in the bioacti- 726. Guengerich, RR (1999). Human cytochrome P-450
vation of aflatoxins in human liver. Proc. Natl. 3A4: Regulation and role in drug metabolism.
Acad. Sci. USA 86, A62-A65. Annu. Rev. Pharmacol. Toxicol. 39, 1-17.
715. Brian, W.R., M.-A. Sari, M. Iwasaki, T. Shimada, 727. Barwick, J.L., L.C. Quattrochi, A.S. Mills,
L.S. Kaminsky, and RR Guengerich (1990). C. Potenza, R.H. Tukey, and PS. Guzelian (1996).
Catalytic activities of human liver cytochrome Trans-species gene transfer for analysis of gluco-
P-450 IIIA4 expressed in Saccharomyces cere- corticoid-inducible transcriptional activation of
visiae. Biochemistry 29, 11280-11292. transiently expressed human CYP3A4 and rabbit
716. Guengerich, RR (1990). Mechanism-based inacti- CYP3A6 in primary cultures of adult rat and rabbit
vation of human liver cytochrome P-450 IIIA4 by hepatocytes. Mol. Pharmacol. 50, 10-16.
gestodene. Chem. Res. Toxicol. 3, 363-371. 728. Schuetz, E.G. and PS. Guzelian (1984). Induction
717. Kelley, J.D., D.L. Eaton, RR Guengerich, and of cytochrome P-450 by glucocorticoids in rat
R.A. Coulombe, Jr. (1997). Aflatoxin Bj activation liver: II. Evidence that glucocorticoids regulate
in human lung. Toxicol. Appl. Pharmacol. 144, induction of cytochrome P-450 by a nonclassical
88-95. receptor mechanism. J. Biol. Chem. 259,
718. Koch, I., R. Weil, R. Wolbold, J. Brockmoller, 2007-2012.
E. Hustert, O. Burk et al (2002). Interindividual 729. Calleja, C , J.M. Pascussi, J.C. Mani, R Maurel,
variability and tissue-specificity in the expression and M.J. Vilarem (1998). The antibiotic rifampicin
of cytochrome P450 3A mRNA. Drug Metab. is a nonsteroidal ligand and activator of the human
Dispos. 30, 1108-1114. glucocorticoid receptor. Nat. Med. 4, 92-96.
719. Nakamoto, T., I. Hase, S. Imaoka, T. Hiroi, Y. Oda, 730. Ray, D.W., A.M. Lovering, J.R. Davis, and
A. Asada et al. (2000). Quantitative RT-PCR for A. White (1998). Rifampicin: A glucocorticoid
CYP3A4 mRNA in human peripheral lympho- receptor ligand?. Nat. Med. 4, 1090-1091.
cytes: Induction of CYP3A4 in lymphocytes and 731. Kliewer, S.A., J.T. Moore, L. Wade,
in liver by rifampicin. Pharmacogenetics 10, J.L. Staudinger, M.A. Watson, S.A. Jones et al.
571-575. (1998). An orphan nuclear receptor activated by
720. de Wildt, S.N., G.L. Kearns, J.S. Leeder, and pregnanes defines a novel steroid signaling path-
J.N. van den Anker (1999). Cytochrome P450 way. Cell 92, 73-82.
3A. Ontogeny and drug disposition. Clin. 732. Lehmann, J.M., D.D. McKee, M.A. Watson,
Pharmacokinet. 37, 485-505. TM. Wilson, J.T Moore, and S.A. Kliewer (1998).
721. El Mouelhi, M., M.S. Didolkar, E.G. Elias, The human orphan nuclear receptor PXR is activated
RR Guengerich, and EC. Kauffman (1987). by compounds that regulate CYP3A4 gene expres-
Hepatic drug metabolizing enzymes in primary sion and cause drug interactions. J. Clin. Invest. 102,
and secondary tumors of human liver. Cancer Res. 1016-1023.
47, 460-466. 733. Coumoul, X., M. Diry, and R. Barouki (2002).
722. Fujitaka, K., T. Oguri, T. Isobe, Y Fujiwara, and PXR-dependent induction of human CYP3A4
N. Kohno (2001). Induction of cytochrome P450 gene expression by organochlorine pesticides.
3A4 by docetaxel in peripheral mononuclear cells Biochem. Pharmacol. 64, 1513-1519.
and its expression in lung cancer. Cancer 734. Jones, S.A., L.B. Moore, G.B. Wisely, and
Chemother. Pharmacol. 48, 4 2 ^ 6 . S.A. Kliewer (2002). Use of in vitro pregnane X
496 F. Peter Guengerich

receptor assays to assess CYP3A4 induction CAR-mediated xenobiotic induction of CYP3A4.


potential of drug candidates. Meth. Enzymol. 357, Nat. Med. 9, 220-224.
161-170. 746. Jover, R., R. Bort, M.J. Gomez-Lechon, and
735. Raucy, J., L. Warfe, M.F. Yueh, and S.W. Allen J.V Castell (2002). Down-regulation of human
(2002). A cell-based reporter gene assay for deter- CYP3A4 by the inflammatory signal interleukin-6:
mining induction of CYP3A4 in a high-volume Molecular mechanism and transcription factors
system. J. Pharmacol. Exp. Then 303, 412-423. 'mwo\\Q&. FASEB J. 16, 1799-1801.
736. Luo, G., M, Cunningham, S. Kim, T. Burn, J. Lin, 747. Danan, G., V Descatoire, and D. Pessayre (1981).
M. Sinz et al. (2002). CYP3A4 induction by drugs: Self-induction by erythromycin of its own trans-
Correlation between a pregnane X receptor reporter formation into a metabolite forming an inactive
gene assay and CYP3A4 expression in human complex with reduced cytochrome P-450.
hepatocytes. Drug Metab. Dispos. 30, 795-804. J. Pharmacol. Exp. Ther 218, 509-514.
737. Zhang, J., P Kuehl, E.D. Green, J.W. Touchman, 748. Bensoussan, C , M. Delaforge, and D. Mansuy
PB. Watkins, A. Daly et al. (2001). The human (1995). Particular ability of cytochromes P450 3 A
pregnane X receptor: Genomic structure and iden- to form inhibitory P450-iron-metabolite com-
tification and functional characterization of natural plexes upon metabolic oxidation of amino drugs.
alleHc variants. Pharmacogenetics 11, 555-572. Biochem. Pharmacol. 49, 591-602.
738. Goodwin, B., E. Hodgson, and C. Liddle (1999). 749. Wang, X.Y, K.F. Medzihradszky, D. Maltby, and
The orphan human pregnane X receptor mediates M.A. Correia (2001). Phosphorylation of native
the transcriptional activation of CYP5A4 by and heme-modified CYP3A4 by protein kinase C:
rifampicin through a distal enhancer module. Mol. A mass spectrometric characterization of the phos-
Pharmacol. 56, 1329-1339. phorylated peptides. Biochemistry 40,
739. El-Sankary, W., V Bombail, G.G. Gibson, and 11318-11326.
N. Plant (2002). Glucocorticoid-mediated induc- 750. Sy, S.K., A. Ciaccia, W. Li, E.A. Roberts, A. Okey,
tion of CYP3A4 is decreased by disruption of a W. Kalow et al. (2002). Modeling of human
protein: DNA interaction distinct from the preg- hepatic CYP3A4 enzyme kinetics, protein, and
nane X receptor response element. Drug Metab. mRNA indicates deviation from log-normal distri-
Dispos. 30, 1029-1034. bution in CYP3A4 gene expression. Eur J. Clin.
740. Takeshita, A., M. Taguchi, N. Koibuchi, and Pharmacol. 58, 357-365.
Y. Ozawa (2002). Putative role of the orphan 751. Ball, S.E., J. Scatina, J. Kao, G.M. Ferron, R.
nuclear receptor SXR (steroid and xenobiotic Fruncillo, P. Mayer et al. (1999). Population distri-
receptor) in the mechanism of CYP3A4 inhibition bution and effects on drug metaboHsm of a genetic
by xenobiotics. J. Biol. Chem. Ill, 32453-32458. variant in the 5' promoter region of CYP3A4. Clin.
741. Goodwin, B., E. Hodgson, D.J. D'Costa, Pharmacol. Ther 66, 288-294.
G.R. Robertson, and C. Liddle (2002). 752. Spurdle, A.B., B. Goodwin, E. Hodgson,
Transcriptional regulation of the human CYP3A4 J.L. Hopper, X. Chen, DM. Purdie et al. (2002). The
gene by the constitutive androstane receptor. Mol. CYP3A4*1B polymorphism has no functional sig-
Pharmacol. 62, 359-365. nificance and is not associated with risk of breast or
742. Thummel, K.E., C. Brimer, K. Yasuda, ovarian cancer. Pharmacogenetics 12, 355-366.
J. Thottassery, T. Senn, Y. Lin et al. (2001). 753. Eiselt, R., T.L. Domanski, A. Zibat, R. Mueller,
Transcriptional control of intestinal cytochrome E. Presecan-Siedel, E. Hustert et al. (2001).
P-4503A by la,25-dihydroxy vitamin D3. Mol. Identification and functional characterization of
Pharmacol. 60, 1399-1406. eight CYP3A4 protein variants. Pharmacogenetics
743. Hara, H., Y Yasunami, and T. Adachi (2002). 11, 447-458.
Alteration of cellular phosphorylation state affects 754. Garcia-Martin, E., C. Martinez, R.M. Pizarro,
vitamin D receptor-mediated CYP3A4 mRNA F.J. Garcia-Gamito, H. GuUsten, H. Raunio et al.
induction in Caco-2 cells. Biochem. Biophys. Res. (2002). CYP3A4 variant alleles in white individu-
Commun. 296, 182-188. als with low CYP3A4 enzyme activity. Clin.
744. Ourlin, J.C, Y. Jounaidi, P. Maurel, and Pharmacol. Ther 71, 196-204.
M.J. Vilarem (1997). Role of the liver-enriched 755. Lamba, J.K., YS. Lin, K. Thummel, A. Daly,
transcription factors C/EBPa and DBP in the PB. Watkins, S. Strom et al. (2002). Common allelic
expression of human CYP3A4 and CYP3A7. variants of cytochrome P4503A4 and their preva-
J. Hepatol. 26(Suppl 2), 54-62. lence in different populations. Pharmacogenetics
745. Tirona, R.G., W. Lee, B.F. Leake, L.B. Lan, 12, 121-132.
C.B. Cline, V Lamba et al. (2003). The orphan 756. Dai, D, J. Tang, R. Rose, E. Hodgson,
nuclear receptor HNF4a determines PXR- and R.J. Bienstock, H.W Mohrenweiser et al. (2001).
Human P450s 497

Identification of variants of CYP3A4 and charac- 766. Zhang, Z., Y. Li, R.A. Stearns, RR. Ortiz De
terization of their abihties to metaboHze testos- Montellano, T.A. Baillie, and W. Tang (2002).
terone and chlorpyrifos. J. Pharmacol. Exp. Ther. Cytochrome P450 3A4-mediated oxidative conver-
299,825-831. sion of a cyano to an amide group in the metabo-
757. Lamba, J.K., Y.S. Lin, E.G. Schuetz, and lism of pinacidil. Biochemistry 41, 2712-2718.
K.E. Thummel (2002). Genetic contribution to 767. Hosea, N.A. and FR Guengerich (1998).
variable human CYPSA-mediated metabolism. Oxidation of non-ionic detergents by cytochrome
Adv. DrugDeliv. Rev. 54, 1271-1294. P450 enzymes. Arch. Biochem. Biophys. 353,
758. Williams, J.A., B.J. Ring, VE. Cantrell, D.R. Jones, 365-373.
J. Eckstein, K. Ruterbories et al. (2002). 768. Bodin, K., L. Bretillon, Y Aden, L. Bertilsson,
Comparative metabolic capabilities of CYP3A4, U. Broome, C. Einarsson, and U. Diczfalusy
CYP3A5, and CYP3A7. Drug Metab. Dispos. 30, (2001). Antiepileptic drugs increase plasma levels
883-891. of 4(3-hydroxycholesterol in humans: Evidence for
759. Wang, R.W., RH. Kari, A.Y.H. Lu, RE. Thomas, involvement of cytochrome P450 3A4. J. Biol.
ER Guengerich, and K.R Vyas (1991). Chem. 276, 38685-38689.
Biotransformation of lovastatin. IV Identification 769. Bodin, K., U. Andersson, E. Rystedt, E. Ellis,
of cytochrome P-450 3A proteins as the major M. Norlin, I. Pikuleva et al. (2002). Metabolism of
enzymes responsible for the oxidative metabolism 4(3-hydroxycholesterol in humans. J. Biol. Chem.
of lovastatin in rat and human liver microsomes. 277,31534-31540.
Arch. Biochem. Biophys. 290, 355-361. 770. Furster, C. and K. Wikvall (1999). Identification of
760. Huskey, S.E.W, D.C. Dean, R.R. Miller, CYP3A4 as the major enzyme responsible for 25-
G.H. Rasmusson, and S.H.L. Chiu (1995). hydroxylation of 5p-cholestane-3a,7a,12a-triol in
Identification of human cytochrome P450 isozymes human liver microsomes. Biochim. Biophys. Acta
responsible for the in vitro oxidative metabolism of 1437, 46-52.
finasteride. Drug Metab. Dispos. 23, 1126-1135. 771. Honda, A., G. Salen, Y Matsuzaki, A.K. Batta,
761. Kronbach, T., V Fischer, and U.A. Meyer (1988). G. Xu, E. Leitersdorf et al. (2001). Side chain
Cyclosporine metabolism in human liver: hydroxylations in bile acid biosynthesis catalyzed
Identification of a cytochrome P-450III gene fam- by CYP3A are markedly up-regulated in Cyp27~^~
ily as the major cyclosporine-metabolizing enzyme mice but not in cerebrotendinous xanthomatosis.
explains interactions of cyclosporine with other J. Biol. Chem. 276, 34579-34585.
drugs. Clin. Pharmacol. Ther. 43, 630-635. 772. Dussault, I., H.D. Yoo, M. Lin, E. Wang, M. Fan,
762. Combalbert, J., I. Fabre, G. Fabre, I. Dalet, A.K. Batta et al. (2003). Identification of an
J. Derancoiirt, J.R Cano et al. (1989). Metabolism endogenous ligand that activates pregnane X
of cyclosporin A. IV Purification and identifica- receptor-mediated sterol clearance. Proc. Natl.
tion of the rifampicin-inducible human liver Acad. Sci. USA 100, 833-838.
cytochrome P-450 (cyclosporin A oxidase) as a 773. Boocock, D.J., K. Brown, A.H. Gibbs, E. Sanchez,
product of P450IIIA gene subfamily. Drug Metab. K.W. Turteltaub, and I.N. White (2002).
Dispos. 17, 197-207. Identification of human CYP forms involved in the
763. Koudriakova, T., E. latsimirskaia, I. Utkin, activatidn of tamoxifen and irreversible binding to
E. Gangl, R Vouros, E. Storozhuk et al. (1998). DNA. Carcinogenesis 23, 1897-1901.
Metabolism of the human immunodeficiency virus 774. Ueng, Y.-F, T. Shimada, H. Yamazaki, and
protease inhibitors indinavir and ritonavir by ER Guengerich (1995). Oxidation of aflatoxin B^
human intestinal microsomes and expressed by bacterial recombinant human cytochrome P450
cytochrome P4503A4/3A5: Mechanism-based enzymes. Chem. Res. Toxicol. 8, 218-225.
inactivation of cytochrome P4503A by ritonavir. 775. Iyer, R., B. Coles, K.D. Raney, R. Thier,
Drug Metab. Dispos. 26, 552-561. ER Guengerich, and T.M. Harris (1994). DNA
764. Warrington, J.S., R.I. Shader, L.L. von Moltke, and adduction by the potent carcinogen aflatoxin B^:
D.J. Greenblatt (2000). In vitro biotransformation Mechanistic studies. J. Am. Chem. Soc.116,
of sildenafil (Viagra): Identification of human 1603-1609.
cytochromes and potential drug interactions. Drug 776. Crespi, C.L., VR Miller, and B.W Penman (1997).
Metab. Dispos. 28, 392-397. Microtiter plate assays for inhibition of human,
765. Kudo, S., M.G. Okumura, and T. Ishizaki (1999). drug-metabolizing cytochromes P450. Anal.
Cytochrome P-450 isoforms involved in car- Biochem. 248, 188-190.
boxylic acid ester cleavage of Hantzsch pyridine 777. Stresser, D.M., S.D. Turner, A.R Blanchard,
ester of pranidipine. Drug Metab. Dispos. 27, VR Miller, and C.L. Crespi (2002). Cytochrome
303-308. P450 fluorometric substrates: Identification of
498 F. Peter Guengerich

isoform-selective probes for rat CYP2D2 and human 789. Krivoruk, Y, M.T. Kinirons, A.J.J. Wood, and
CYP3A4. DrugMetab. Dispos. 30, 845-852. M. Wood (1994). Metabohsm of cytochrome
778. Chauret, N., N. Tremblay, R.L. Lackman, P4503A substrates in vivo administered by the
J.-Y. Gauthier, J.M. Silva, J. Marois et al (1999). same route: Lack of correlation between alfentanil
Description of a 96-well plate assay to measure clearance and erythromycin breath test. Clin.
cytochrome P4503A inhibition in human liver Pharmacol. Ther 56, 608-614.
microsomes using a selective fluorescent probe. 790. Kivisto, K.T and H.K. Kroemer (1997). Use of
Anal. Biochem. 276, 215-226. probe drugs as predictors of drug metabolism in
779. Kenworthy, K.E., J.C. Bloomer, S.E. Clarke, and humans. J. Clin. Pharmacol. 37, 40S-48S.
J.B. Houston (1999). CYP3A4 drug interactions: 791. Thummel, K.E., D. O'Shea, M.K Paine, D.D. Shen,
Correlation of 10 in vitro probe substrates. Br. J. K.L. Kunze, J.D. Perkins et al. (1996). Oral first-
Clin. Pharmacol. 48, 716-727. pass elimination of midazolam involves both
780. Schellens, J.H.M., PA. Soons, and D.D. Breimer gastrointestinal and hepatic CYP3A-mediated
(1988). Lack of bimodality in nifedipine plasma metabolism. Clin. Pharmacol. Ther 59, 491-502.
kinetics in a large population of healthy subjects. 792. Paine, M.K, D.A. Wagner, K.A. Hoffmaster, and
Biochem. Pharmacol. 37, 2507-2510. PB. Watkins (2002). Cytochrome P450 3A4 and
781. Lown, K., J. Kolars, K. Turgeon, R. Merion, P-glycoprotein mediate the interaction between an
S.A. Wrighton, and PB. Watkins (1992). The oral erythromycin breath test and rifampin. Clin.
erythromycin breath test selectively measures Pharmacol. Ther 72, 524-535.
P450IIIA in patients with severe liver disease. 793. Schuetz, E.G., WT. Beck, and J.D. Schuetz (1996).
Clin. Pharmacol. Ther 51, 229-238. Modulators and substrates of P-glycoprotein and
782. Gremse, D.A., H.H. A-Kader, T.J. Schroeder, and cytochrome P4503A coordinately up-regulate
W.F. BaHstreri (1990). Assessment of lidocaine these proteins in human colon carcinoma cells.
metabolite formation as a quantitative hver func- Mol. Pharmacol. 49, 311-318.
tion test in children. Hepatology 12, 565-569. 794. Gillam, E.M.J., T Baba, B.-R. Kim, S. Ohmori,
783. May, D.G., J. Porter, G.R. Wilkinson, and and F.P Guengerich (1993). Expression of modi-
R.A. Branch (1994). Frequency distribution of fied human cytochrome P450 3A4 in Escherichia
dapsone yV-hydroxylase, a putative probe for P450 coli and purification and reconstitution of the
3A4 activity, in a white population. Clin. QX\zymQ. Arch. Biochem. Biophys. 305, 123-131.
Pharmacol. Ther 55, 492-500. 795. Imaoka, S., Y. Imai, T. Shimada, and Y Funae
784. Thummel, K.E., D.D. Shen, R.L. Carithers, Jr., (1992). Role of phospholipids in reconstituted
P Hartwell, TD. Podoll, W.F. Trager et al. (1993). cytochrome P450 3A forms and mechanism of
Prediction of in vivo midazolam clearance from their activation of catalytic activity. Biochemistry
hepatic CYP3A content and midazolam 1 '-hydrox- 31, 6063-6069.
ylation activity in liver transplant patients. In 796. Ingelman-Sundberg, M., A.-L. Hagbjork, Y.-F. Ueng,
Abstracts, 5th North American ISSX Meeting, Vol. H. Yamazaki, and F.P Guengerich (1996). High rates
4. p. 235, 17-21 October, Tucson, AZ. of substrate hydroxylation by human cytochrome
785. Wanwimolruk, S., M.F Paine, S.N. Pusek, and P450 3A4 in reconstituted membranous vesicles:
PB. Watkins (2002). Is quinine a suitable probe to Influence of membrane charge. Biochem. Biophys.
assess the hepatic drug-metabolizing enzyme Res. Commun. 221, 318-322.
CYP3A4? Br J. Clin. Pharmacol. 54, 643-651. 797. Peyronneau, M.A., J.P Renaud, G. Truan, P. Urban,
786. Ged, C , J.M. Rouillon, L. Pichard, J. Combalbert, D Pompon, and D. Mansuy (1992). Optimization
N. Bressot, P. Bories et al. (1989). The increase in of yeast-expressed human liver cytochrome-P450
urinary excretion of 6P-hydroxycortisol as a 3A4 catalytic activities by coexpressing NADPH-
marker of human hepatic cytochrome P450IIIA cytochrome P450 reductase and cytochrome b^.
induction. Br J. Clin. Pharmacol. 28, 373-387. Eur J. Biochem. 207, 109-116.
787. Watkins, PB., T.A. Hamilton, T.M. Annesley, 798. Guengerich, FP and WW Johnson (1997).
C.N. Ellis, J.C. Kolars, and J.J. Voorhees (1990). Kinetics of ferric cytochrome P450 reduction by
The erythromycin breath test as a predictor of NADPH-cytochrome P450 reductase: Rapid
cyclosporine blood levels. Clin. Pharmacol. Ther reduction in absence of substrate and variations
48, 120-129. among cytochrome P450 systems. Biochemistry
788. Kinirons, M.T., D. Oshea, T.E. Downing, 36, 14741-14750.
A.T. Fitzwilliam, L. Joellenbeck, J.D. Groopman et 799. Shet, M.S., C.W Fisher, PL. Holmans, and
al. (1993). Absence of correlations among 3 puta- R.W. Estabrook (1993). Human cytochrome P450
tive in vivo probes of human cytochrome P4503 A 3A4: Enzymatic properties of a purified recombi-
activity in young healthy men. Clin. Pharmacol. nant fusion protein containing NADPH-P450 reduc-
Ther 54,621-629. tase. Proc. Natl. Acad. Sci. USA 90, 11748-11752.
Human P450s 499

800. Gillam, E.M.J., Z. Guo, Y.-F. Ueng, H. Yamazaki, CYP4A4 metabolism of prostaglandin E-1 and
I. Cock, P.E.B. Reilly et aL (1995). Expression of arachidonic acid. Arch. Biochem. Biophys. 393,
cytochrome P450 3A5 in Escherichia coli: Effects 329-338.
of 5' modifications, purification, spectral charac- 811. Guryev, O.L., A.A. Gilep, S.A. Usanov, and
terization, reconstitution conditions, and catalytic R.W. Estabrook (2001). Interaction of apo-
activities. Arch. Biochem. Biophys. 317, 374-384. cytochrome b^ with cytochromes P4503A4 and
801. Yamazaki, H., M. Nakano, Y. Imai, Y.-F. Ueng, P45017A: Relevance of heme transfer reactions.
EP. Guengerich, and T. Shimada (1996). Roles of Biochemistry 40, 5018-5031.
cytochrome b^ in the oxidation of testosterone and 812. Yamazaki, H., T. Shimada, M.V Martin, and
nifedipine by recombinant cytochrome P450 3A4 F.P. Guengerich (2001). Stimulation of cytochrome
and by human liver microsomes. Arch. Biochem. P450 reactions by apo-cytochrome b^ Evidence
Biophys. 325, 174-182. against transfer of heme from cytochrome P450
802. Yamazaki, H., M. Nakajima, M. Nakamura, 3A4 to apo-cytochrome b^ or heme oxygenase.
S. Asahi, N. Shimada, E.M.J. Gillam et al. (1999). J. Biol. Chem. 276, 30885-30891.
Enhancement of cytochrome P-450 3A4 catalytic 813. Ekins, S., G. Bravi, J.H. Wikel, and S.A. Wrighton
activities by cytochrome b^ in bacterial mem- (1999). Three-dimensional-quantitative structure
branes. DrugMetab. Dispos. 27, 999-1004. activity relationship analysis of cytochrome P-450
803. Parikh, A., E.M.J. Gillam, and ER Guengerich 3A4 substrates. J. Pharmacol. Exp. Ther 291,
(1997). Drug metabolism by Escherichia coli 424-433.
expressing human cytochromes P450. Nat. 814. Ekins, S., G. Bravi, S. Binkley, J.S. Gillespie,
Biotechnol. 15, 784-788. B.J. Ring, J.H. Wikel et al. (1999). Three- and
804. Blake, J.A.R., M. Pritchard, S. Ding, G.C.M. Smith, four-dimensional quantitative structure activity
B. Burchell, C.R. Wolfe/ al (1996). Coexpression relationship analyses of cytochrome P-450 3A4
of a human P450 (CYP3A4) and P450 reductase inhibitors. J. Pharmacol. Exp. Ther 290,
generates a highly functional monooxygenase sys- 429-438.
tem in Escherichia coli, FEBS Lett. 397, 210-214. 815. Stevens, J.C, TL. Domanski, G.R. Harlow,
805. Nakajima, M., K. Tane, S. Nakamura, N. Shimada, R.B. White, E. Orton, and J.R. Halpert (1999). Use
H. Yamazaki, and T. Yokoi (2002). Evaluation of of the steroid derivative RPR 106541 in combina-
approach to predict the contribution of multiple tion with site-directed mutagenesis for enhanced
cytochrome P450s in drug metabolism using relative cytochrome P-450 3A4 structure/function analysis.
activity factor: Effects of the differences in expres- J. Pharmacol. Exp. Ther 290, 594-602.
sion levels of NADPH-cytochrome P450 reductase 816. Domanski, T.L., YA. He, G.R. Harlow, and
and cytochrome b^ in the expression system and the J.R. Halpert (2000). Dual role of human
differences in the marker activities. J. Pharm. Sci. 91, cytochrome P450 3A4 residue Phe-304 in sub-
952-963. strate specificity and cooperativity. J. Pharmacol.
806. Noshiro, M. and T. Omura (1978). Immuno- Exp. Ther 293,585-591.
chemical study on the electron pathway from 817. Fowler, S.M., R.J. Riley, M.R Pritchard,
NADH to cytochrome P-450 of liver microsomes. M.J. Sutchffe, T Friedberg, and C.R. Wolf (2000).
J. Biochem. (Tokyo) 83, 61-77. Amino acid 305 determines catalytic center acces-
807. Perret, A. and D. Pompon (1998). Electron shuttle sibility in CYP3A4. Biochemistry 39, 4406-4414.
between membrane-bound cytochrome P450 3A4 818. Xue, L., H.F Wang, Q. Wang, G.D. Szklarz,
and 63 rules uncoupling mechanisms. Biochemistry T.L. Domanski, J.R. Halpert et al. (2001).
37, 11412-11424. Influence of P450 3A4 SRS-2 residues on cooper-
808. Yamazaki, H., W.W. Johnson, Y.-F Ueng, ativity and/or regioselectivity of aflatoxin Bj oxi-
T. Shimada, and F.P. Guengerich (1996). Lack of dation. Chem. Res. Toxicol. 14, 483-491.
electron transfer from cytochrome b^ in stimulation 819. Fowler, S.M., J.M. Taylor, T. Friedberg, C.R. Wolf,
of catalytic activities of cytochrome P450 3A4: and R.J. Riley (2002). CYP3A4 active site volume
Characterization of a reconstituted cytochrome modification by mutagenesis of leucine 211. Drug
P450 3A4/NADPH-cytochrome P450 reductase Metab. Dispos. 30, 452^56.
system and studies with apo-cytochrome b^. 820. Smith, D.A. and B.C. Jones (1992). Speculations
J. Biol. Chem. 271, 27438-27444. on the substrate structure-activity relationship
809. Auchus, R.J., T.C. Lee, and W.L. Miller (1998). (SSAR) of cytochrome P450 enzymes. Biochem.
Cytochrome b^ augments the 17,20-lyase activity Pharmacol. 44, 2089-2098.
of human P450cl7 without direct electron transfer. 821. Macdonald, T.L., WG. Gutheim, R.B. Martin, and
J. Biol. Chem. 273, 3158-3165. F.P Guengerich (1989). Oxidation of substituted
810. Aitken, A.E., L.J. Roman, PA, Loughran, M. de la A'.A^-dimethylanilines by cytochrome P-450:
Garza, and B.S.S. Masters (2001). Expressed Estimation of the effective oxidation-reduction
500 F. Peter Guengerich

potential of cytochrome P-450. Biochemistry 28, 834. Wang, R.W., D.J. Newton, T.D. Scheri, and
2071-2077. A.Y.H. Lu (1997). Human cytochrome P450 3A4-
822. Yamazaki, H., Y.-F. Ueng, T. Shimada, and catalyzed testosterone 6p-hydroxylation and eryth-
F.P. Guengerich (1995). Roles of divalent metal romycin A^-demethylation. Drug Metab. Dispos.
ions in oxidations catalyzed by recombinant 25, 502-507.
cytochrome P450 3A4 and replacement of 835. Ludwig, E., J. Schmid, K. Beschke, and T. Ebner
NADPH-cytochrome P450 reductase with other (1999). Activation of human cytochrome P-450
flavoproteins, iron-sulfur proteins, and oxygen sur- 3A4-catalyzed meloxicam 5'-methylhydroxylation
rogates. Biochemistry 34, 8380-8389. by quinidine and hydroquinidine in vitro.
823. Northrop, D.B. (1975). Steady-state analysis of J. Pharmacol Exp. Ther 290, 1-8.
kinetic isotope effects in enzymic reactions. 836. Wang, R.W, D.J. Newton, N. Liu, W.M. Atkins,
Biochemistry 14, 2644-2651. and A.Y.H. Lu (2000). Human cytochrome P-450
824. Kuby, S.A. (1991). A Study of Enzymes, Vol I, 3A4: In vitro drug-drug interaction patterns are
Enzyme Catalysis, Kinetics, and Substrate substrate-dependent. Drug Metab. Dispos. 28,
Binding. CRC Press, Boca Raton, FL. 360-366.
825. Walsh, C. (1979). Enzymatic Reaction Mechanisms. 837. Schrag, M.L. and L.C. Wienkers (2001). Covalent
W. H. Freeman Co, San Francisco, CA. alteration of the CYP3A4 active site: Evidence for
826. Obach, R.S. (2001). Mechanism of cytochrome multiple substrate binding domains. Arch.
P4503A4- and 2D6-catalyzed dehydrogenation of Biochem. Biophys. 391, 49-55.
ezlopitant as probed with isotope effects using five 838. Masubuchi, Y, A. Ose, and T. Horie (2002).
deuterated analogs. Drug Metab. Dispos. 29, Diclofenac-induced inactivation of CYP3A4 and
1599-1607. its stimulation by quinidine. Drug Metab. Dispos.
827. Wiebel, RJ., J.C. Leutz, L. Diamond, and 30, 1143-1148.
H.V Gelboin (1971). Aryl hydrocarbon 839. Shou, M., J. Grogan, J.A. Mancewicz, K.W. Krausz,
(benzo[«]pyrene) hydroxylase in microsomes from FJ. Gonzalez, H.V Gelboin et al (1994). Activation
rat tissues: Differential inhibition and stimulation of CYP3A4: Evidence for the simultaneous binding
by benzoflavones and organic solvents. Arch. of two substrates in a cytochrome P450 active site.
Biochem. Biophys. 144, 78-86. Biochemistry 33, 6450-6455.
828. Kapitulnik, J., PJ. Poppers, M.K. Buening, 840. Korzekwa, K.R., N. Krishnamachary, M. Shou,
J.G. Fortner, and A.H. Conney (1977). Activation A. Ogai, R.A. Parise, A.E. Rettie et al. (1998).
of monooxygenases in human liver by 7,8-ben- Evaluation of atypical cytochrome P450 kinetics
zoflavone. Clin. Pharmacol Ther 11, 475^85. with two-substrate models: Evidence that multiple
829. Buening, M.K., J.G. Fortner, A. Kappas, and substrates can simultaneously bind to cytochrome
A.H. Conney (1978). 7,8-Benzoflavone stimulates P450 active sites. Biochemistry 37, 4137^147.
the metabolic activation of aflatoxin Bj to muta- 841. Shou, M., R. Dai, D. Cui, K.R. Korzekwa,
gens by human liver. Biochem. Biophys. Res. T.A. Baillie, and T.H. Rushmore (2001). A kinetic
Commun. 82, 348-355. model for the metabolic interaction of two sub-
830. Schwab, G.E., J.L. Raucy, and E.F. Johnson strates at the active site of cytochrome P450 3A4.
(1988). Modulation of rabbit and human hepatic J. Biol Chem. 276, 2256-2262.
cytochrome P-450-catalyzed steroid hydroxyla- 842. Kenworthy K.E., S.E. Clarke, J. Andrews, and
tions by a-naphthoflavone. Mol Pharmacol 33, J.B. Houston (2001). Multisite kinetic models for
493-499. CYP3A4: Simultaneous activation and inhibition
831. Hosea, N.A., G.P Miller, and FP Guengerich (2000). of diazepam and testosterone metabolism. Drug
Elucidation of distinct binding sites for cytochrome Metab. Dispos. 29, 1644-1651.
P450 3A4. Biochemistry 39, 5929-5939. 843. Galetin, A., S.E. Clarke, and J.B. Houston (2002).
832. Guengerich, RP, B.-R. Kim, E.M.J. Gillam, and Quinidine and haloperidol as modifiers of
T. Shimada (1994). Mechanisms of enhancement CYP3A4 activity: Multisite kinetic model
and inhibition of cytochrome P450 catalytic activ- approach. Drug Metab. Dispos. 30, 1512-1522.
ity In M. C. Lechner (ed.). Proceedings, 8th 844. Domanski, T.L., J. Liu, G.R. Harlow, and
International Conference on Cytochrome P450: J.R. Halpert (1998). Analysis of four residues
Biochemistry, Biophysics, and Molecular Biology, within substrate recognition site 4 of human
John Libbey Eurotext, Paris, pp. 97-101. cytochrome P450 3A4: Role in steroid hydroxy-
833. Yun, C.-H., T. Shimada, and FP Guengerich (1992). lase activity and a-naphthoflavone stimulation.
Contributions of human liver cytochrome P-450 Arch. Biochem. Biophys. 350, 223-232.
enzymes to the N-oxidation of 4, 4'-methylene- 845. Harlow, G.R. and J.R. Halpert (1998).
bis(2-chloroaniline). Carcinogenesis 13, 217-222. Analysis of human cytochrome P450 3A4
Human P450s 501

cooperativity: Construction and characterization of protein site: A matter of pre-existing populations.


a site-directed mutant that displays hyperboHc Protein Sci. 11, 184-197.
steroid hydroxylation kinetics. Proc. Natl. Acad. 857. Carlson, H.A. (2002). Protein flexibility and drug
Sci. t/&4 95,6636-6641. design: How to hit a moving target. Curr Opin.
846. He, Y.A., F. Roussel, and J.R. Halpert (2003). Chem. Biol. 6, 447^52.
Analysis of homotropic and heterotropic coopera- 858. Carlson, H.A. (2002). Protein flexibility is an
tivity of diazepam oxidation by CYP3A4 using important component of structure-based drug dis-
site-directed mutagenesis and kinetic modeling. covery. Curr Pharm. Des. 8, 1571-1578.
Arch. Biochem. Biophys. 409, 92-101. 859. Schlichting, I., J. Berendzen, K. Chu, A.M. Stock,
847. Domanski, T.L., Y.A. He, K.K. Khan, F. Roussel, S.A. Maves, D.E. Benson et al (2000). The cat-
Q. Wang, and J.R. Halpert (2001). Phenylalanine alytic pathway of cytochrome P450^^j^ at atomic
and tryptophan scanning mutagenesis of CYP3A4 resolution. Science 287, 1615-1622.
substrate recognition site residues and effect 860. Atkins, W.M., R.W. Wang, and A.Y.H. Lu (2001).
on substrate oxidation and cooperativity. Allosteric behavior in cytochrome P450-dependent
Biochemistry ^^, 10150-10160. in vitro drug-drug interactions: A prospective
848. Dabrowski, M.J., M.L. Schrag, L.C. Wienkers, and based on conformational dynamics. Chem. Res.
W.M. Atkins (2002). Pyrene-pyrene complexes at Toxicol. 14, 338-347.
the active site of cytochrome P450 3A4: Evidence 861. Segel, I.H. (1975). Enzyme Kinetics. Behavior and
for a multiple substrate binding site. J. Am. Chem. Analysis of Rapid Equilibrium and Steady-State
Soc. 124, 11866-11867. Enzyme Systems. John Wiley & Sons, New York.
849. Cupp-Vickery, J., R. Anderson, and Z. Hatziris 862. Atkins, W.M., WD. Lu, and D.L. Cook (2002). Is
(2000). Crystal structures of ligand complexes of there a toxicological advantage for non-hyperbolic
P450eryF exhibiting homotropic cooperativity. kinetics in cytochrome P450 catalysis? Functional
Proc. Natl. Acad. Sci. USA 97, 3050-3055. allostery from "distributive catalysis." J. Biol.
850. Khan, K.K., H. Liu, and J.R. Halpert (2003). Chem. Ill, 33258-33266.
Homotropic versus heterotorpic cooperativity of 863. Di Petrillo, K., S. Wood, V Kostrubsky, K. Chatfield,
cytochrome P450eryF: A substrate oxidation and J. Bement, S. Wrighton et al (2002). Effect of caf-
spectral titration study. Drug Metab. Dispos. 31, feine on acetaminophen hepatotoxicity in cultured
356-359. hepatocytes treated with ethanol and isopentanol.
851. Koley, A.P, J.T.M. Buters, R.C. Robinson, Toxicol. Appl. Pharmacol. 185, 91-97.
A. Markowitz, and RK. Friedman (1995). CO 864. Tang, W and R.A. Stearns (2001). Heterotropic
binding kinetics of human cytochrome P450 3A4: cooperativity of cytochrome P450 3A4 and poten-
Specific interaction of substrates with kinetically tial drug-drug interactions. Curr Drug Metab. 2,
distinguishable conformers. J. Biol. Chem. 270, 185-198.
5014-5018. 865. Egnell,A.C., J.B. Houston, andC.S. Boyer (2003). In
852. Koley, A.P, J.T.M. Buters, R.C. Robinson, vivo CYP3A4 heteroactivation is a possible mecha-
A. Markowitz, and F.K. Friedman (1997). nism for the drug interaction between felbamate and
Differential mechanisms of cytochrome P450 inhi- carbamazepine. J. Pharmacol. Exp. Ther. 305,
bition and activation by a-naphthoflavone. J. Biol. 1251-1262.
Chem. 272,3149-3152. 866. Yun, C.-H., R.A. Okerholm, and F.P Guengerich
853. Koley, A.P., R.C. Robinson, and F.K. Friedman (1993). Oxidation of the antihistaminic drug terfe-
(1996). Cytochrome P450 conformation and sub- nadine in human liver microsomes: Role of
strate interactions as probed by CO binding cytochrome P450 3A(4) in N-dealkylation and
kinetics. Biochimie 78, 706-713. C-hydroxylation. Drug Metab. Dispos. 21,
854. Koley, A.P, R.C. Robinson, A. Markowitz, and 403^09.
F.K. Friedman (1997). Drug-drug interactions: 867. Delaforge, M., M. Jaouen, and D. Mansuy (1983).
Effect of quinidine on nifedipine binding to human Dual effects of macrolide antibiotics on rat liver
cytochrome P450 3A4. Biochem. Pharmacol. 53, cytochrome P-450: Induction and formation of
455^60. metabolite-complexes: A structure-activity rela-
855. Anzenbacherova', E., N. Bee, P. Anzenbacher, tionship. Biochem. Pharmacol. 32, 2309-2318.
J. Hudecek, P Soucek, C. Jung et ah (2000). 868. Bailey, D.G., B. Edgar, J.D. Spence, C. Munzo, and
Flexibility and stability of the structure of J.M.O. Arnold (1990). Felodipine and nifedipine
cytochromes P450 3A4 and BM-3. Eur J. interactions with grapefruit juice. Clin. Pharmacol.
Biochem. 261, 2916-2920. Ther 47, 180.
856. Ma, B., M. Shatsky, H.J. Wolfson, and R. Nussinov 869. Greenblatt, D.J., K.C. Patki, L.L. von Moltke, and
(2002). Multiple diverse ligands binding at a single R.I. Shader (2001). Drug interactions with grapefruit
502 F. Peter Guengerich

juice: An update. J. Clin. Psychopharmacol. 21, cyclosporine pharmacokinetics in marrow transplant


357-359. patients. Blood 64, 1277-1279.
870. Guengerich, F.P. and D.-H. Kim (1990). In vitro 882. von Moltke, L.L. D.J. Greenblatt, S.X. Duan,
inhibition of dihydropyridine oxidation and afla- J.S. Harmatz, and R.I. Shader (1994). In vitro pre-
toxin Bj activation in human liver microsomes by diction of the terfenadine-ketoconazole pharmacoki-
naringenin and other flavonoids. Carcinogenesis netic interaction.^ Clin. Pharmacol. 34,1222-1227.
11, 2275-2279. 883. Dome, J.L., K. Walton, and A.G. Renwick (2003).
871. von Moltke, L.L., D.J. Greenblatt, J.M. Grassi, Human variability in CYP3A4 metabolism and
B.W. Granda, S.X. Duan, S.M. Fogelman et al CYP3A4-related uncertainty factors for risk
(1998). Protease inhibitors as inhibitors of human assessment. Food Chem. Toxicol. 41, 201-224.
cytochromes P450: High risk associated with 884. Yang, L.Q., S.J. Li, YE Cao, X.B. Man, W.E Yu,
ritonavir. J. Clin. Pharmacol. 38, 106-111. H.Y Wang et al. (2003). Different alterations of
872. Unger, M., U. Holzgrabe, W. Jacobsen, C. Cummins, cytochrome P450 3A4 isoform and its gene
and L.Z. Benet (2002). Inhibition of cytochrome expression in livers of patients with chronic liver
P450 3A4 by extracts and kavalactones of Piper diseases. World J. Gastroenterol. 9, 359-363.
methysticum (kava-kava). Planta Med. 68, 885. Lang, C.C, R.M. Brown, M.T. Kinirons,
1055-1058. M.-A. Deathridge, F.P. Guengerich, D. Kelleher
873. Lin, H.L., U.M. Kent, and PR Hollenberg (2002). et al. (1996). Decreased intestinal P450 3A4 in
Mechanism-based inactivation of cytochrome celiac sprue: Reversal following successful gluten
P450 3A4 by 17a-ethynylestradiol: Evidence for free diet. Clin. Pharmacol. Ther 59, 4 1 ^ 6 .
heme destruction and covalent binding to protein. 886. Zhou, S., Y Gao, W Jiang, M. Huang, A. Xu, and
J. Pharmacol. Exp. Then 301, 160-167. J.W Paxton (2003). Interactions of herbs with
874. Palovaara, S., K.T. Kivisto, P. Tapanainen, cytochrome P450. Drug Metab. Rev 35, 35-98.
P. Manninen, P.J. Neuvonen, and K. Laine (2000). 887. Moore, L.B., B. Goodwin, S.A. Jones, G.B. Wisely
Effect of an oral contraceptive preparation contain- C.J. Serabjit-Singh, T.M. Willson et al. (2000). St.
ing ethinylestradiol and gestodene on CYP3A4 John's wort induces hepatic drug metabolism
activity as measured by midazolam 1'-hydroxyla- through activation of the pregnane X receptor. Proc.
tion. Br J. Clin. Pharmacol. 50, 333-337. Natl. Acad Sci. USA 97, 7500-7502.
875. Kuhl, H., C. Jung-Hoffmann, and E Heidt (1988). 888. Watkins, R.E., J.M. Maglich, L.B. Moore,
Alterations in the serum levels of gestodene and G.B. Wisely, S.M. Noble, PR. Davis-Searles et al.
SHBG during 12 cycles of treatment with 30 |jLg (2003). 2.1 A crystal structure of human PXR in
ethinylestradiol and 75 |jLg gestodene. complex with the St. John's wort compound hyper-
Contraception 38, 477-486. forin. Biochemistry 42, 1430-1438.
876. Balogh, A., S. Gessinger, U. Svarovsky, M. Hippius, 889. Henderson, L., Q.Y Yue, C. Bergquist, B. Gerden,
U. Mellinger, G. Klinger et al. (1998). Can oral con- and P. Arlett (2002). St. John's wort (Hypericum
traceptive steroids influence the elimination of perforatum): Drug interactions and clinical out-
nifedipine and its primary pryidine metabolite in comes. Br J. Clin. Pharmacol. 54, 349-356.
humans?. Eur J. Clin. Pharmacol. 54, 729-734. 890. Kolars, J.C, P Benedict, P Schmiedlin-Ren, and
877. Guengerich, EP (1986). Covalent binding to PB. Watkins (1994). Aflatoxin B,-adduct forma-
apoprotein is a major fate of heme in a variety of tion in rat and human small bowel enterocytes.
reactions in which cytochrome P-450 is destroyed. Gastroenterology 106, 433-439.
Biochem. Biophys. Res. Commun. 138, 193-198. 891. Felix, C.A., A.H. Walker, B.J. Lange, T.M. Williams,
878. He, K., L.M. Bomheim, A.M. Ealick, D. Maltby N.J. Winick, N.-K.V Cheung et al. (1998).
H. Yin, and M.A. Correia (1998). Identification of Association of CYP3A4 genotype with treatment-
the heme-modified peptides from cumene related leukemia. Proc. Natl. Acad. Sci. USA 95,
hydroperoxide-inactivated cytochrome P450 3A4. 13176-13181.
Biochemistry 2>1, 17448-17457. 892. Miyoshi, Y, A. Ando, Y Takamura, T. Taguchi,
879. Wienkers, L.C. (2001). Problems associated with in Y Tamaki, and S. Noguchi (2002). Prediction of
vitro assessment of drug inhibition of CYP3A4 and response to docetaxel by CYP3A4 mRNA expression
other P-450 enzymes and its impact on drug dis- in breast cancer tissues. Int. J. Cancer 97, 129-132.
covery. J. Pharmacol. Toxicol. Methods 45, 79-84. 893. Blanco, J.G., M.J. Edick, M.L. Hancock,
880. Plant, N.J. and G.G. Gibson (2003). Evaluation of N.J. Winick, T Dervieux, M.D. Amylon etal. (2002).
the toxicological relevance of CYP3A4 induction. Genetic polymorphisms in CYP3A5, CYP3A4 and
Curr Opin. Drug Discov. Devel. 6, 50-56. NQOl in children who developed therapy-related
881. Yee, G.C., M.S. Kennedy R. Storb, and E.D. Thomas myeloid malignancies. Pharmacogenetics 12,
(1984). Effect of hepatic dysfunction on oral 605-611.
Human P450s 503

894. Rebbeck, T.R., J.M. Jaffe, A.H. Walker, A.J. Wein, 905. Janardan, S.K., K.S. Lown, P Schmiedlin-Ren,
and S.B. Malkowicz (1998). Modification of clini- K.E. Thummel, and RB. Watkins (1996). Selective
cal presentation of prostate tumors by a novel expression of CYP3A5 and not CYP3A4 in human
genetic variant in CYP3A4. J. Natl. Cancer Inst. blood. Pharmacogenetics 6, 379-385.
90, 1225-1228. 906. Hakkola, J., H. Raunio, R. Purkunen, S. Saarikoski,
895. Tayeb, M.T., C. Clark, L. Sharp, N.E. Haites, K. Vahakangas, O. Pelkonen et al. (2001).
RH. Rooney, G.I. Murray et al. (2002). CYP3A4 Cytochrome P450 3 A expression in the human fetal
promoter variant is associated with prostate cancer liver: Evidence that CYP3A5 is expressed in only a
risk in men with benign prostate hyperplasia. limited number of fetal livers. Biol. Neonate 80,
Oncol. Rep. 9, 653-655. 193-201.
896. Tayeb, M.T., C. Clark, N.E. Haites, L. Sharp, 907. Jouna'idi, Y, PS. Guzelian, and M.J. Vilarem
G.I. Murray, and H.L. McLeod (2003). CYP3A4 (1994). Sequence of the 5'-flanking region of
and VDR gene polymorphisms and the risk of CYP3A5: Comparative analysis with CYP3A4 and
prostate cancer in men with benign prostate hyper- CYP3A7. Biochem. Biophys. Res. Commun. 205,
plasia. Br. J. Cancer 88, 928-932. 1741-1747.
897. Ando, Y., T. Tateishi, Y Sekido, T. Yamamoto, 908. Schuetz, J.D., E.G. Schuetz, J.V Thottassery,
T. Satoh, Y Hasegawa et al (1999). Modification PS. Guzelian, S. Strom, and D. Sun (1996).
of clinical presentation of prostate tumors by a Identification of a novel dexamethasone respon-
novel genetic variant in CYP3A4. J. Natl. Cancer sive enhancer in the human CYP3A5 gene and its
Inst. 91, 1587-1588. activation in human and rat liver cells. Mol.
898. Kittles, R.A., W. Chen, R.K. Panguluri, Pharmacol. 49, 63-72.
C. Ahaghotu, A. Jackson, C.A. Adebamowo et al. 909. Lin, Y.S., A.L. Dowling, S.D. Quigley, KM. Farin,
(2002). CYP3A4-V and prostate cancer in African J. Zhang, J. Lamba et al. (2002). Co-regulation of
Americans: Causal or confounding association CYP3A4 and CYP3A5 and contribution to hepatic
because of population stratification?. Hum. Genet. and intestinal midazolam metabolism. Mol.
110, 553-560. Pharmacol. 62, 162-172.
899. Wojnowski, L., E. Hustert, K. Klein, M. Goldammer, 910. Hustert, E., M. Haberl, O. Burk, R. Wolbold,
M. Haberl, J. Kirchheiner et al. (2002). Re: modifi- YQ. He, K. Klein et al. (2001). The genetic deter-
cation of clinical presentation of prostate tumors by a minants of the CYP3A5 polymorphism.
novel genetic variant in CYP3A4. J. Natl. Cancer Pharmacogenetics 11, 773-779.
Inst. 94, 630-631. 911. Paulussen, A., K. Lavrijsen, H. Bohets,
900. Wrighton, S.A., B.J. Ring, RB. Watkins, and J. Hendrickx, P Verhasselt, W Luyten et al. (2000).
M. VandenBranden (1989). Identification of a Two linked mutations in transcriptional regulatory
polymorphically expressed member of the human elements of the CYP3A5 gene constitute the major
cytochrome P-450III family. Mol. Pharmacol. 86, genetic determinant of polymorphic activity in
97-105. humans. Pharmacogenetics 10, 415^24.
901. Aoyama, T., S. Yamano, D.J. Waxman, 912. Shih, PS. and J.D. Huang (2002). Pharmacokinetics
D.R Lapenson, U.A. Meyer, V Fischer et al. of midazolam and 1 '-hydroxymidazolam in Chinese
(1989). Cytochrome P-450 hPCN3, a novel with different CYP3A5 genotypes. Drug Metab.
cytochrome P-450 III A gene product that is differ- Dispos. 30, 1491-1496.
entially expressed in adult human liver. J. Biol. 913. Wrighton, S.A., W.R. Brian, M.A. Sari, M. Iwasaki,
Chem. 264, 10388-10395. EP Guengerich, J.L. Raucy et al. (1990). Studies on
902. Murray, G.L, S. Pritchard, W.T. Melvin, and the expression and metabolic capabilities of human
M.D. Burke (1995). Cytochrome P450 CYP3A5 in liver cytochrome P450IIIA5 (HLp3). Mol.
the human anterior pituitary gland. FEBS Lett. Pharmacol. 38, 207-213.
364, 79-82. 914. Gibbs, M.A., K.E. Thummel, D.D. Shen, and
903. Yamakoshi, Y, T. Kishimoto, K. Sugimura, and K.L. Kunze (1999). Inhibition of cytochrome P-
H. Kawashima (1999). Human prostate CYP3A5: 450 3A(CYP3A) in human intestinal and liver
Identification of a unique 5'-untranslated sequence microsomes: Comparison of K- values and impact
and characterization of purified recombinant protein. of CYP3A5 expression. Drug Metab. Dispos. 27,
Biochem. Biophys. Res. Commun. 260, 676-681. 180-187.
904. Hukkanen, J., T. Vaisanen, A. Lassila, R. Piipari, 915. Kamataki, T., M. Sugiura, Y Yamazoe, and R. Kato
S. Anttila, O. Pelkonen et al. (2003). Regulation of (1979). Purification and properties of cytochrome
CYP3A5 by glucocorticoids and cigarette smoke P-450 and NADPH-cytochrome c (P-450) reduc-
in human lung-derived cells. J. Pharmacol. Exp. tase from human liver microsomes. Biochem.
Then 304, 745-752. Pharmacol. 28, 1993-2000.
504 F. Peter Guengerich

916. Kitada, M., T. Kamataki, K. Itahashi, T. Rikihisa, element in the 5' flanking region. Biochim.
R. Kato, and Y. Kanakubo (1985). Purification and Biophys. Acta 1130, 133-138.
properties of cytochrome P-450 from homogenates 927. Greuet, J., L. Pichard, C. Bonfils, J. Domergue,
of human fetal livers. Arch. Biochem. Biophys. and P. Maurel (1996). The fetal specific gene
241, 275-280. CYP3A7 is inducible by rifampicin in adult human
917. Kitada, M., T. Kamataki, K. Itahashi, T. Rikihisa, hepatocytes in primary culture. Biochem. Biophys.
R. Kato, andY. Kanakubo (1985). Immunochemical Res. Commun. 225, 689-694.
examinations of C3^ochrome P-450 in various tissues 928. Pascussi, J.M., Y Jounaidi, L. Drocourt,
of human fetuses using antibodies to human fetal J. Domergue, C. Balabaud, P Maurel et al. (1999).
cytochrome P-450, P-450 HFLa. Biochem. Biophys. Evidence for the presence of a functional pregnane
Res. Commun. 131, 1154-1159. X receptor response element in the CYP3A7 pro-
918. Okajima, Y, N. Inaba, I. Fukazawa, Y Ota, moter gene. Biochem. Biophys. Res. Commun. 260,
Y Hirai, N. Sato etal. (1993). Immunohistochemical 377-381.
and immunoelectron microscopic study of 929. Bertilsson, G., A. Berkenstam, and P. Blomquist
cytochrome P-450 of human fetal livers (P- (2001). Functionally conserved xenobiotic respon-
450HFLa): Implications for an onco-feto-placental sive enhancer in cytochrome P450 3A7. Biochem.
enzyme. Asia Oceania J. Obstet. Gynaecol. 19, Biophys. Res. Commun. 280, 139-144.
329-341. 930. Saito, T., Y Takahashi, H. Hashimoto, and
919. Schuetz, J.D., S. Kauma, and PS. Guzelian (1993). T. Kamataki (2001). Novel transcriptional regula-
Identification of the fetal liver cytochrome tion of the human CYP3A7 gene by Spl and Sp3
CYP3A7 in human endometrium and placenta. through nuclear factor KB-like element. J. Biol.
J. Clin. Invest. 92, 1018-1024. C/zew. 276, 38010-38022.
920. Sarkar, M.A., V Vadlamuri, S. Ghosh, and 931. Finta, C. and PG. Zaphiropoulos (2000). The
D.D. Glover (2003). Expression and cychc vari- human cytochrome P450 3A locus. Gene evolution
ability of CYP3A4 and CYP3A7 isoforms in by capture of downstream exons. Gene 260, 13-23.
human endometrium and cervix during the men- 932. Burk, O., H. Tegude, I. Koch, E. Hustert,
strual cycle. Drug Metab. Dispos. 31, 1-6. R. Wolbold, H. Glaeser, K. et al. (2002). Molecular
921. Hakkola, J., H. Raunio, R. Purkunen, O. Pelkonen, mechanisms of polymorphic CYP3A7 expression
S. Saarikoski, T. Cresteil et al. (1996). Detection of in adult human liver and intestine. J. Biol. Chem.
cytochrome P450 gene expression in human pla- 277, 24280-24288.
centa in first trimester of pregnancy. Biochem. 933. Kitada, M., T. Kamataki, K. Itahashi, T. Rikihisa,
Pharmacol. 52, 379-383. and Y Kanakubo (1987). Significance of
922. Yang, H.Y.L., Q.P Lee, A.E. Rettie, and cytochrome P-450 (P-450 HFLa) of human fetal
M.R. Juchau (1994). Functional cytochrome livers in the steroid and drug oxidations. Biochem.
P4503A isoforms in human embryonic tissues: Pharmacol. 36, 453-^56.
Expression during organogenesis. Mol. 934. Kitada, M., T. Kamataki, K. Itahashi, T. Rikihisa,
Pharmacol. 46, 922-928. and Y Kanakubo (1987). P-450 HFLa, a form
923. Schuetz, J.D., D.L. Beach, and PS. Guzelian of cytochrome P-450 purified from human
(1994). Selective expression of cytochrome P450 fetal livers, is the 16a-hydroxylase of dehy-
CYP3A mRNAs in embryonic and adult human droepiandrosterone 3-sulfate. J. Biol. Chem. 262,
liver. Pharmacogenetics 4, 11-20. 13534-13537.
924. Tateishi, T., H. Nakura, M. Asoh, M. Watanabe, 935. Ohmori, S., N. Fujiki, H. Nakasa, H. Nakamura,
M. Tanaka, T. Kumai et al. (1997). A comparison I. Ishii, K. Itahashi et al. (1998). Steroid hydroxy-
of hepatic cytochrome P450 protein expression lation by human fetal CYP3A7 and human
between infancy and postinfancy. Life Sci. 61, NADPH-cytochrome P450 reductase coexpressed
2567-2574. in insect cells using baculovirus. Res. Commun.
925. Komori, M., K. Nishio, H. Ohi, M. Kitada, and Mol. Pathol. Pharmacol. 100, 15-28.
T. Kamataki (1989). Molecular cloning and 936. Gorski, J.C., S.D. Hall, D.R. Jones,
sequence analysis of cDNA containing entire cod- M. VandenBranden, and S.A. Wrighton (1994).
ing region for human fetal liver cytochrome P-450. Regioselective biotransformation of midazolam by
J. Biochem. (Tokyo) 106, 161-163. members of the human cytochrome P450 3A
926. Itoh, S., T. Yanagimoto, S. Tagawa, H. Hashimoto, (CYP3A) subfamily. Biochem. Pharmacol. 47,
R. Kitamura et al. (1992). Genomic organization 1643-1653.
of human fetal specific P-450IIIA7 (cytochrome 937. Gillam, E.M.J., R.M. Wunsch, Y.-R Ueng,
P-450HFLa)-related gene(s) and interaction of T. Shimada, PE.B. Reilly, T. Kamataki et al.
transcriptional regulatory factor with its DNA (1997). Expression of cytochrome P450 3A7 in
Human P450s 505

Escherichia coli: Effects of 5' modification and Characterization of a cDNA encoding a human
catalytic characterization of recombinant enzymes kidney, cytochrome P-450 4A fatty acid w-hydroxy-
expressed in bicistronic format with NADPH- lase and the cognate enzyme expressed in
cytochrome P450 reductase. Arch. Biochem. Escherichia coli. Biochim. Biophys. Acta 1172,
Biophys. 346, 81-90. 161-166.
938. Hashimoto, H., Y. Yanagawa, M. Sawada, S. Itoh, 949. Bell, D.R., N.J. Plant, C.G. Rider, L. Na, S. Brown,
T. Deguchi, andT. Kamataki (1995). Simultaneous I. Ateitalla et al. (1993). Species-specific induction
expression of human CYP3A7 and A^-acetyltrans- of cytochrome P-450 4A RNAs: PCR cloning
ferase in Chinese hamster CHL cells results in high of partial guinea-pig, human and mouse CYP4A
cytotoxicity for carcinogenic heterocyclic amines. cDNAs. Biochem. J. 294, 173-180.
Arch. Biochem. Biophys. 320, 323-329. 950. Imaoka, S., H. Ogawa, S. Kimura, and F.J. Gonzalez
939. Li, Y, T. Yokoi, M. Katsuki, J.S. Wang, (1993). Complete cDNA sequence and cDNA-
J.D. Groopman, and T. Kamataki (1997). In vivo directed expression of CYP4A11, a fatty acid co-
activation of aflatoxin Bj in C57BL/6N mice car- hydroxylase expressed in human kidney. DNA Cell
rying a human fetus-specific CYP3A7 gene. Biol. 12, 893-899.
Cancer Res. 57, 641-645. 951. Powell, PK., I. Wolf, and J.M. Lasker (1996).
940. Yamada, A., K. Fujita, T. Yokoi, S. Muto, A. Suzuki, Identification of CYP4A11 as the major lauric
Y. Gondo et al. (1998). In vivo detection of muta- acid w-hydroxylase in human liver microsomes.
tions induced by aflatoxin Bj using human Arch. Biochem. Biophys. 335, 219-226.
CYP3A7/HITEC hybrid mice. Biochem. Biophys. 952. Bellamine, A., Y Wang, M.R. Waterman, J.V Gainer,
Res. Commun. 250, 150-153. III, E.R Dawson, N.J. Brown et al. (2003).
941. Li, Y, T. Yokoi, R. Kitamura, M. Sasaki, M. Gunji, Characterization of the CYP4A11 gene, a second
M. Katsuki et al. (1996). Establishment of CYP4A gene in humans. Arch. Biochem. Biophys.
transgenic mice carrying human fetus-specific 409,221-227.
CYP3A7. Arch. Biochem. Biophys. 329, 235-240. 953. Lu, A.Y.H. and M.J. Coon (1968). Role of hemo-
942. Chen, H., A.G. Fantel, and M.R. Juchau (2000). protein P-450 in fatty acid co-hydroxylation in
Catalysis of the 4-hydroxylation of retinoic acids a soluble enzyme system from liver microsomes.
by CYP3A7 in human fetal hepatic tissues. Drug J.Biol. Chem. 243, 1331-1332.
Metab. Dispos. 28, 1051-1057. 954. Gonzalez, M.C., C. Marteau, J. Franchi, and
943. Inoue, E., Y Takahashi, Y Imai, and T. Kamataki D. Migliore-Samour (2001). Cytochrome P450
(2000). Development of bacterial expression sys- 4A11 expression in human keratinocytes: Effects
tem with high yield of CYP3A7, a human fetus- of ultraviolet irradiation. Br J. Dermatol. 145,
specific form of cytochrome P450. Biochem. 749-757.
Biophys. Res. Commun. 269, 623-627. 955. Cummings, B.S., J.M. Lasker, and L.H. Lash
944. Kondoh, N., T. Wakatsuki, A. Ryo, A. Hada, (2000). Expression of glutathione-dependent
T. Aihara, S. Horiuchi et al. (1999). Identification enzymes and cytochrome P450s in freshly isolated
and characterization of genes associated with and primary cultures of proximal tubular cells
human hepatocellular carcinogenesis. Cancer Res. from human kidney. J. Pharmacol. Exp. Then 293,
59, 4990-4996. 677-685.
945. Gellner, K., R. Eiselt, E. Hustert, H. Arnold, 956. Savas, U., M.H. Hsu, and E.E Johnson (2003).
I. Koch, M. Haberl et al. (2001). Genomic organi- Differential regulation of human CYP4A genes by
zation of the human CYP3 A locus: Identification of peroxisome proliferators and dexamethasone.
a new, inducible CYP3A gene. Pharmacogenetics Arch. Biochem. Biophys. 409, 212-220.
II, 111-121. 957. Kawashima, H., T. Naganuma, E. Kusunose,
946. Domanski, T.L., C. Finta, J.R. Halpert, and T. Kono, R. Yasumoto, K. Sugimura et al. (2000).
P.G. Zaphiropoulos (2001). cDNA cloning and ini- Human fatty acid co-hydroxylase. CYP4AJI:
tial characterization of CYP3A43, a novel human Determination of complete genomic sequence and
cytochrome P450. Mol. Pharmacol. 59, 386-392. characterization of purified recombinant protein.
947. Westlind, A., S. Malmebo, I. Johansson, C. Otter, Arch. Biochem. Biophys. 378, 333-339.
T.B. Andersson, M. Ingelman-Sundberg et al. 958. Cattley, R.C., J. DeLuca, C. Elcombe, P Fenner-
(2001). Cloning and tissue distribution of a novel Crisp, B.G. Lake, D.S. Marsman et al. (1998). Do
human cytochrome P450 of the CYP3A subfamily, peroxisome proliferating compounds pose a hepa-
CYP3A43. Biochem. Biophys. Res. Commun. 281, tocarcinogenic hazard to humans?. Regul. Toxicol.
1349-1355. Pharmacol. 27, 47-60.
948. Palmer, C.N., T.H. Richardson, K.L Griffin, 959. Dierks, E.A., Z. Zhang, E.R Johnson, and PR. Ortiz
M.H. Hsu, A.S. Muerhofif, J.E. Clark et al. (1993). de Montellano (1998). The catalytic site of
506 F. Peter Guengerich

cytochrome P4504A11 (CYP4A11) and its L131F P450 CYP4B1: Evidence for a non-functional
mutant. J. Biol. Chem. 273, 23055-23061. allelic variant. Pharmacogenetics 12, 367-374.
960. Hoch, U, Z. Zhang, D.L. Kroetz, and P.R. Ortiz de 971. Zheng, Y.-M., M.B. Fisher, N. Yokotani, Y Fujii-
Montellano (2000). Structural determination of the Kuriyama, and A.E. Rettie (1998). Identification
substrate specificities and regioselectivities of the of a meander region proline residue critical for
rat and human fatty acid w-hydroxylases. Arch. heme binding to cytochrome P450: Implications
Biochem. Biophys. 373, 63-71. for the catalytic function of human CYP4B1.
961. Powell, PK., I. Wolf, R. Jin, and J.M. Lasker Biochemistry'il, 12847-12851.
(1998). MetaboHsm of arachidonic acid to 20- 972. Imaoka, S., K. Hayashi, T. Hiroi, Y Yabusaki,
hydroxy-5,8,ll,14-eicosatetraenoic acid by P450 T. Kamataki, and Y Funae (2001). A transgenic
enzymes in human liver: Involvement of CYP4F2 mouse expressing human CYP4B1 in the liver.
and CYP4A11. J. Pharmacol. Exp. Then 285, Biochem. Biophys. Res. Commun. 284, 757-762.
1327-1336. 973. Henne, K.R., M.B. Fisher, K.R. Iyer, D.H. Lang,
962. Lasker, J.M., W.B. Chen, I. Wolf, B.R Bloswick, WF. Trager, and A.E. Rettie (2001). Active site
PD. Wilson, and PK. Powell (2000). Formation of characteristics of CYP4B1 probed with aromatic
20-hydroxyeicosatetraenoic acid, a vasoactive and ligands. Biochemistry 40, 8597-8605.
natriuretic eicosanoid, in human kidney. J. Biol. 974. Henne, K.R., K.L. Kunze, YM. Zheng, P Christmas,
Chem. 275,4118^126. R.J. Soberman, and A.E. Rettie (2001). Covalent
963. Oliw, E.H., K. Stark, and J. Bylund (2001). linkage of prosthetic heme to CYP4 family P450
Oxidation of prostaglandin W^ and prostaglandin enzymes. Biochemistry 40, 12925-12931.
H2 analogues by human cytochromes P450: 975. Hoch, U and PR. Ortiz de Montellano (2001).
Analysis of w-side chain hydroxy metaboHtes and Covalently linked heme in cytochrome P4504A
four steroisomers of 5-hydroxyprostaglandin Ij by fatty acid hydroxylases. J. Biol. Chem. 276,
mass spectrometry. Biochem. Pharmacol. 62, 11339-11346.
407-^15. 976. Frank, S., S. StefFens, U Fischer, A. Tlolko,
964. Chang, Y.T. and G.H. Loew (1999). Homology N.G. Rainov, and CM. Kramm (2002).
modeling and substrate binding study of human Differential cytotoxicity and bystander effect of
CYP4A11 enzyme. Proteins 34, 403^15. the rabbit cytochrome P450 4B1 enzyme gene by
965. LeBrun, L.A., U. Hoch, and P R. Ortiz de two different prodrugs: Implications for pharma-
Montellano (2002). Autocatalytic mechanism and cogene therapy. Cancer Gene Ther 9, 178-188.
consequences of covalent heme attachment in the 977. Kikuta, Y, E. Kusunose, T. Kondo, S. Yamamoto,
cytochrome P4504A family. J. Biol. Chem. Ill, H. Kinoshita, and M. Kusunose (1994). Cloning
12755-12761. and expression of a novel form of leukotriene B4
966. Nhamburo, PT., F.J. Gonzalez, O.W. McBride, omega-hydroxylase from human liver. FEBS Lett.
H.V Gelboin, and S. Kimura (1989). Identification 348,70-74.
of a new P450 expressed in human lung: Com- 978. Kikuta, Y, E. Kusunose, and M. Kusunose (2002).
plete cDNA sequence, cDNA-directed expression, Prostaglandin and leukotriene w-hydroxylases.
and chromosome mapping. Biochemistry 28, Prostaglandins Other Lipid Medial 68-69, 345-362.
8060-8066. 979. Jin, R., D.R. Koop, J.L. Raucy, and J.M. Lasker
967. Imaoka, S., Y Yoneda, T. Sugimoto, T. Hiroi, (1998). Role of human CYP4F2 in hepatic catabo-
K. Yamamoto, T. Nakatani et al. (2000). CYP4B1 is lism of the proinflammatory agent leukotriene B4.
a possible risk factor for bladder cancer in humans. Arch. Biochem. Biophys. 359, 89-98.
Biochem. Biophys. Res. Commun. Ill, 776-780. 980. Kikuta, Y, Y. Miyauchi, E. Kusunose, and
968. Iscan, M., T. Klaavuniemi, T. Coban, M. Kusunose (1999). Expression and molecular
N. Kapucuoglu, O. Pelkonen, and H. Raunio cloning of human liver leukotriene B4 w-hydroxy-
(2001). The expression of cytochrome P450 lase (CYP4F2) gene. DMA Cell Biol. 18, 723-730.
enzymes in human breast tumours and normal 981. Kikuta, Y, E. Kusunose, and M. Kusunose (2000).
breast tissue. Breast Cancer Res. Treat. 70, 47-54. Characterization of human liver leukotriene B^ w-
969. Finnstrom, N., C. Bjelfman, T.G. Soderstrom, hydroxylase P450 (CYP4F2). J. Biochem. (Tokyo)
G. Smith, L. Egevad, B.J. Norlen et al. (2001). 111, 1047-1052.
Detection of cytochrome P450 mRNA transcripts 982. Kikuta, Y, E. Kusunose, K. Endo, S. Yamamoto,
in prostate samples by RT-PCR. Eur J. Clin. Invest. K. Sogawa, Y Fujii-Kuriyama et al. (1993). A
31, 880-886. novel form of cytochrome P-450 family 4 in
970. Lo-Guidice, J.M., D. Allorge, C. Cauffiez, human polymorphonuclear leukocytes: cDNA
D. Chevalier, J.J. Lafitte, M. Lhermitte et al. (2002). cloning and expression of leukotriene B^ co-
Genetic polymorphism of the human cytochrome hydroxylase. J. Biol. Chem. 268, 9376-9380.
Human P450s 507

983. Kikuta, Y., M. Kato, Y. Yamashita, Y Miyauchi, platelet thromboxane A synthase. Biochem.
K. Tanaka, N. Kamada et al (1998). Human Biophys. Res. Commun. 178, 1479-1484.
leukotriene B^ w-hydroxylase (CYP4F3) gene: 996. Nusing, R. and V Ullrich (1992). Regulation of
Molecular cloning and chromosomal localization. cyclooxygenase and thromboxane synthase in
DNA Cell Biol. 17, 221-230. human monocytes. Eur J. Biochem. 206, 131-136.
984. Christmas, P., S.R. Ursino, J.W. Fox, and 997. Young, V, M. Ho, H. Vosper, J.J. Belch, and
R.J. Soberman (1999). Expression of the CYP4F3 C.N. Palmer (2002). Elevated expression of the
gene. J. Biol. Chem. 274, 21191-21199. genes encoding TNF-a and thromboxane syn-
985. Christmas, P., J.P Jones, C.J. Patten, D.A. Rock, thase in leucocytes from patients with systemic
Y Zheng, S.M. Cheng et al. (2001). Alternative sclerosis. Rheumatology 41, 869-875.
splicing determines the function of CYP4F3 by 998. Nusing, R., RM. Fehr, F. Gudat, E. Kemeny,
switching substrate specificity. J. Biol. Chem. 276, M.J. Mihatsch, and V Ullrich (1994). The local-
38166-38172. ization of thromboxane synthase in normal and
986. Bylund, J., N. Finnstrom, and E.H. Oliw (1999). pathological human kidney tissue using a mono-
Gene expression of a novel cytochrome P450 of clonal antibody Tu 300. Virchows Arch. 424,
the CYP4F subfamily in human seminal vesicles. 69-74.
Biochem. Biophys. Res. Commun. 261, 169-174. 999. Ihara, H., C. Yokoyama, A. Miyata, T. Kosaka,
987. Bylund, J., M. Hidestrand, M. Ingelman- R. Nusing, V Ullrich et al. (1992). Induction of
Sundberg, and E.H. Oliw (2000). Identification of thromboxane synthase and prostaglandin endoper-
CYP4F8 in human seminal vesicles as a prominent oxide synthase mRNAs in human erythroleukemia
19-hydroxylase of prostaglandin endoperoxides. cells by phorbol ester. FEBS Lett. 306, 161-164.
J. Biol. Chem. 275, 21844-21849. 1000. Yaekashiwa, M. and L.H. Wang (2002).
988. Stark, K., H. Torma, M. Cristea, and E.H. Oliw Transcriptional control of the human thrombox-
(2003). Expression of CYP4F8 (prostaglandin H ane synthase gene in vivo and in vitro. J. Biol.
19-hydroxylase) in human epithelia and prominent Chem. 277, 22497-22508.
induction in epidermis of psoriatic lesions. Arch. 1001. Chevaher, D., J.M. Lo-Guidice, E. Sergent,
Biochem. Biophys. 409, 188-196. D. Allorge, H. Debuysere, N. Ferrari et al. (2001).
989. Cui, X., D.R. Nelson, and H.W. Strobel (2000). Identification of genetic variants in the human
A novel human cytochrome P450 4F isoform thromboxane synthase gene (CYP5A1). Mutat.
(CYP4F11): cDNA cloning, expression, and Res. 432, 61-67.
genomic structural characterization. Genomics 68, 1002. Ullrich, V and M. Haurand (1983). Thromboxane
161-166. synthase as a cytochrome P450 enzyme. Adv.
990. Bylund, J., M. Bylund, and E.H. Oliw (2001). Prostaglandin Thromboxane Leukot. Res. 11,
cDNA cloning and expression of CYP4F12, 105-110.
a novel human cytochrome P450. Biochem. 1003. Haurand, M. and V Ullrich (1985). Isolation and
Biophys. Res. Commun. 280, 892-897. characterization of thromboxane synthase from
991. Hashizume, T, S. Imaoka, T. Hiroi, Y Terauchi, human platelets as a cytochrome P-450 enzyme.
T. Fujii, H. Miyazaki et al. (2001). cDNA cloning J. Biol. Chem. 260, 15059-15067.
and expression of a novel cytochrome P450 1004. Yokoyama, C , A. Miyata, K. Suzuki, Y Nishikawa,
(CYP4F12) from human small intestine. Biochem. T. Yoshimoto, S. Yamamoto et al. (1993).
Biophys. Res. Commun. 280, 1135-1141. Expression of human thromboxane synthase using
992. Hashizume, T., S. Imaoka, M. Mise, Y Terauchi, a baculovirus system. FEBS Lett. 318, 91-94.
T. Fujii, H. Miyazaki et al. (2002). Involvement 1005. Hecker, M., M. Haurand, V Ullrich, U. Diczfalusy,
of CYP2J2 and CYP4F12 in the metabolism and S. Hammarstrom (1987). Products, kinetics,
of ebastine in human intestinal microsomes. and substrate specificity of homogeneous
J. Pharmacol. Exp. Ther 300, 298-304. thromboxane synthase from human platelets:
993. Bylund, J., C. Zhang, and D.R. Harder (2002). Development of a novel enzyme assay. Arch.
Identification of a novel cytochrome P450, Biochem. Biophys. 254, 124-135.
CYP4X1, with unique localization specific to the 1006. Hecker, M., W.J. Baader, P Weber, and V Ullrich
brain. Biochem. Biophys. Res. Commun. 296, (1987). Thromboxane synthase catalyses hydrox-
677-684. ylation of prostaglandin H2 analogs in the
994. Ullrich, V (2003). Thoughts on thiolate tethering. presence of iodosylbenzene. Eur J. Biochem.
Tribute and thanks to a teacher. Arch. Biochem. 169, 563-569.
Biophys. 409,45-51. 1007. Hecker, M. and V Ullrich (1989). On the
995. Yokoyama, C , A. Miyata, H. Ihara, V Ullrich, and mechanism of prostacyclin and thromboxane A2
T. Tanabe (1991). Molecular cloning of human biosynthesis. J. Biol. Chem. 264, 141-150.
508 F. Peter Guengerich

1008. Alusy, UD. and S. Hammarstrom (1977). 1021. Wang, D.P and J.Y. Chiang (1994). Structure and
Inhibitors of thromboxane synthase in human nucleotide sequences of the human cholesterol
platelets. FEES Lett. 82, 107-110. 7a-hydroxylase gene {CYP7). Genomics 20,
1009. Gorman, R.R., G.L. Bundy, D.C. Peterson, 320-323.
F.F. Sun, O.V Miller, and F.A. Fitzpatrick (1977). 1022. Xu, G., G. Salen, S. Shefer, G.C. Ness,
Inhibition of human platelet thromboxane L.B. Nguyen, T.S. Parker et al. (1995).
synthetase by 9,ll-azoprosta-5,13-dienoic acid. Unexpected inhibition of cholesterol 7a-hydrox-
Proc. Natl Acad. Sci. USA 74, 4007-4011. ylase by cholesterol in New Zealand white
1010. Vane, J.R. (1978). Inhibitors of prostaglandin, and Watanabe heritable hyperlipidemic rabbits.
prostacyclin, and thromboxane synthesis. Adv. J. Clin. Invest. 95, 1497-1504.
Prostaglandin Thromboxane Res. 4, 2 7 ^ 4 . 1023. Breuer, O., E. Sudjana-Sugiaman, G. Eggertsen,
1011. Hecker, M., M. Haurand, V Ullrich, and S. Terao J.Y. Chiang, and I. Bjorkhem (1993). Cholesterol
(1986). Spectral studies on structure-activity 7a-hydroxylase is up-regulated by the competi-
relationships of thromboxane synthase inhibitors. tive inhibitor 7-oxocholesterol in rat liver. Eur J.
Eur J. Biochem. 157, 217-223. Biochem. 215, 705-710.
1012. Pace-Asciak, C.R., D. Reynaud, P Demin, 1024. Shibata, A., PF Ohneseit, YC. Tsai, C.H. Spruck,
R. Aslam, and A. Sun (2002). A new family of III, PW Nichols, H.S. Chiang et al. (1994).
thromboxane receptor antagonists with secondary Mutational spectrum in the p53 gene in bladder
thromboxane synthase inhibition. J. Pharmacol. tumors from the endemic area of black foot dis-
Exp. r/?er. 301, 618-624. ease in Taiwan. Carcinogenesis 15, 1085-1087.
1013. Miki, N., R. Miura, and Y. Miyake (1987). 1025. Schwarz, M., E.G. Lund, K.D. Setchell,
Purification and characterization of cholesterol H.J. Kayden, J.E. Zerwekh, I. Bjorkhem et al.
7a-hydroxylase cytochrome P-450 of untreated (1996). Disruption of cholesterol 7a-hydroxylase
rabbit liver microsomes. J. Biochem. (Tokyo) 101, gene in mice. II. Bile acid deficiency is overcome
1087-1094. by induction of oxysterol 7a-hydroxylase. J. Biol.
1014. Ogishima, T, S. Deguchi, and K. Okuda (1987). Chem. 271, 18024-18031.
Purification and characterization of cholesterol 1026. Lehmann, J.M., S.A. Kliewer, L.B. Moore,
7a-hydroxylase from rat liver microsomes. T.A. Smith-Oliver, B.B. Oliver, J.L. Su et al.
J. Biol. Chem. 262, 7646-7650. (1997). Activation of the nuclear receptor LXR
1015. Nguyen, L.B., S. Shefer, G. Salen, G. Ness, by oxysterols defines a new hormone response
R.D. Tanaka, V Packin et al. (1990). Purification pathway J Biol. Chem. 272, 3137-3140.
of cholesterol 7a-hydroxylase from human and 1027. Peet, D.J., S.D. Turley W. Ma, B.A. Janowski,
rat liver and production of inhibiting polyclonal J.M. Lobaccaro, R.E. Hammer et al. (1998).
antibodies. J. Biol. Chem. 265, 4541^546. Cholesterol and bile acid metabolism are
1016. Noshiro, M. and K. Okuda (1990). Molecular impaired in mice lacking the nuclear oxysterol
cloning and sequence analysis of cDNA encoding receptor LXRa. Cell 93, 693-704.
human cholesterol 7 a-hydroxylase. FEBS Lett. 1028. Russell, D.W. (1999). Nuclear orphan receptors
268, 137-140. control cholesterol catabolism. Cell 91, 539-542.
1017. Li, Y.C., D.P Wang, and J.Y.L. Chiang (1990). 1029. Makishima, M., A.Y Okamoto, J.J. Repa, H. Tu,
Regulation of cholesterol 7a-hydroxylase in the R.M. Learned, A. Luk et al. (1999).
liver: Cloning, sequencing, and regulation of cho- Identification of a nuclear receptor for bile acids.
lesterol 7a-hydroxylase mRNA. J. Biol. Chem. Science 2M, 1362-1365.
265, 12012-12019. 1030. Nitta, M., S. Ku, C. Brown, A.Y. Okamoto, and
1018. Jelinek, D.F., S. Andersson, C.A. Slaughter, and B. Shan (1999). CPF: An orphan nuclear receptor
D.W. Russell (1990). Cloning and regulation of that regulates liver-specific expression of the
cholesterol 7a-hydroxylase, the rate-limiting human cholesterol 7a-hydroxylase gene. Proc.
enzyme in bile acid biosynthesis. J. Biol. Chem. Natl. Acad Sci. USA 96, 6669-6665.
265,8190-8197. 1031. Chiang, J.Y, R. Kimmel, C. Weinberger, and
1019. Cohen, J.C, J.J. Cah, D.F Jelinek, M. Mehrabian, D. Stroup (2000). Farnesoid X receptor responds
R.S. Sparkes, A.J. Lusis et al. (1992). Cloning to bile acids and represses cholesterol 7a-hydrox-
of the human cholesterol 7a-hydroxylase gene ylase gene (CYP7AJ) transcription. J. Biol. Chem.
(CYP7) and localization to chromosome 8ql l-ql2. 215, 10918-10924.
Genomics 14, 153-161. 1032. Staudinger, J.L., B. Goodwin, S.A. Jones,
1020. Nishimoto, M., M. Noshiro, and K. Okuda (1993). D. Hawkins-Brown, K.I. MacKenzie, A. LaTour
Structure of the gene encoding human liver cho- et al. (2001). The nuclear receptor PXR is a litho-
lesterol 7a-hydroxylase. Biochim. Biophys. Acta cholic acid sensor that protects against liver toxi-
nil, 147-150. city Proc. Natl. Acad Sci. USA 98, 3369-3374.
Human P450s 509

1033. Gupta, S., W.M. Pandak, and P.B. Hylemon ovariectomized baboons. Biochim. Biophys. Acta
(2002). LXRa is the dominant regulator of 1084, 300-302.
CYP7A1 transcription. Biochem. Biophys. Res. 1044. Crestani, M., A. Sadeghpour, D. Stroup, G. Galli,
Commun. 293, 338-343. and J.Y. Chiang (1996). The opposing effects of
1034. Patel, D.D., B.L. Knight, A.K. Soutar, G.F. Gibbons, retinoic acid and phorbol esters converge to a
and D.P. Wade (2000). The effect of peroxisome- common response element in the promoter of the
proliferator-activated receptor-a on the activity of rat cholesterol 7a-hydroxylase gene (CYP7A).
the cholesterol 7a-hydroxylase gene. Biochem. Biochem. Biophys. Res. Commun. 225, 585-592.
J. 351, 747-753. 1045. Goodart, S.A., C. Huynh, W Chen, A.D. Cooper,
1035. Cheema, S.K. and L.B. Agellon (2000). The and B. Levy-Wilson (1999). Expression of the
murine and human cholesterol 7a-hydroxylase human cholesterol 7a-hydroxylase gene in trans-
gene promoters are differentially responsive to genic mice. Biochem. Biophys. Res. Commun.
regulation by fatty acids mediated via peroxisome 266, 454-459.
proliferator-activated receptor a. J. Biol. Chem. 1046. Chen, J.Y, B. Levy-Wilson, S. Goodart, and
215, 12530-12536. A.D. Cooper (2002). Mice expressing the human
1036. Palmer, C.N.A., M.H. Hsu, K.J. Grififin, J.L. Raucy, CYP7A1 gene in the mouse CYP7A1 knock-out
and E.F. Johnson (1998). Peroxisome proliferator background lack induction of CYP7A1 expres-
activated receptor-a expression in human liver. sion by cholesterol feeding and have increased
Mol. Pharmacol. 53, 14-22. hypercholesterolemia when fed a high fat diet.
1037. Marrapodi, M. and J.Y. Chiang (2000). J. Biol. Chem. 277, 42588^2595.
Peroxisome proliferator-activated receptor a 1047. Reihner, E., I. Bjorkhem, B. Angelin, S. Ewerth,
(PPARa) and agonist inhibit cholesterol and K. Einarsson (1989). Bile acid synthesis in
7a-hydroxylase gene (CYP7A1) transcription. humans: Regulation of hepatic microsomal cho-
J. Lipid Res. 41, 514-520. lesterol 7a-hydroxylase activity. Gastroenterology
1038. De Fabiani, E., N. Mitro, A.C. Anzulovich, 97, 1498-1505.
A. Pinelli, G. GalH, and M. Crestani (2001). The 1048. Mayer, D. (1976). The circadian rhythm of syn-
negative effects of bile acids and tumor necrosis thesis and catabolism of cholesterol. Arch.
factor-a on the transcription of cholesterol 7a- Toxicol. 36, 267-276.
hydroxylase gene {CYP7A1) converge to hepatic 1049. Gielen, J., J. Van Cantfort, B. Robaye, and
nuclear factor-4: A novel mechanism of feedback J. Renson (1975). Rat-liver cholesterol 7a-
regulation of bile acid synthesis mediated hydroxylase. 3. New results about its circadian
by nuclear receptors. J. Biol. Chem. 276, rhythm. Eur J. Biochem. 55, 4 1 ^ 8 .
30708-30716. 1050. Danielsson, H. and K. Wikvall (1981). Evidence
1039. Feingold, K.R., D.K. Spady, A.S. Pollock, for a specific cytochrome P-450 with short half-
A.H. Moser, and C. Grunfeld (1996). Endotoxin, life catalyzing 7a-hydroxylation of cholesterol.
TNF, and IL-1 decrease cholesterol 7a-hydroxy- Biochem. Biophys. Res. Commun. 103, 46-51.
lase mRNA levels and activity. J. Lipid Res. 37, 1051. Hulcher, EH., R.D. MargoUs, and D.J. Bowman
223-228. (1978). Circadian rhythm of cholesterol-7a-
1040. Drover, VA., N.C. Wong, and L.B. Agellon hydroxylase and Cortisol in the African green
(2002). A distinct thyroid hormone response ele- monkey (Cercopithecus aethiops). Biochim.
ment mediates repression of the human choles- Biophys. Acta 529, 409-418.
terol 7a-hydroxylase (CYP7A1) gene promoter. 1052. Chiang, J.Y, WE Miller, and G.M. Lin (1990).
Mol. Endocrinol. 16, 14-23. Regulation of cholesterol 7 alpha-hydroxylase in
1041. Bjorkhem, I., Z. Araya, M. Rudling, B. Angelin, the liver. Purification of cholesterol 7a-hydroxy-
C. Einarsson, and K. Wikvall (2002). Differences lase and the immunochemical evidence for the
in the regulation of the classical and the alterna- induction of cholesterol 7a-hydroxylase by
tive pathv^ay for bile acid synthesis in human cholestyramine and circadian rhythm. J. Biol.
liver. No coordinate regulation of CYP7A1 and Chem. 265, 3889-3897.
CYP27A1. J. Biol. Chem. 277, 26804-26807. 1053. Kinowaki, M., S. Tanaka, Y Maeda, S. Higashi,
1042. Wang, D., D. Stroup, M. Marrapodi, M. Crestani, K. Okuda, and T. Setoguchi (2002). Half-life of
G. Galh, and J.Y.L. Chiang (1996). Transcriptional cholesterol 7a-hydroxylase activity and enzyme
regulation of the human cholesterol 7a-hydroxy- mass differ in animals and humans when deter-
lase gene {CYP7A) in HepG2 cells. J. Lipid Res. mined by a monoclonal antibody against human
37,1831-1841. cholesterol 7a-hydroxylase. J. Steroid Biochem.
1043. Kushwaha, R.S. and K.M. Born (1991). Mol. Biol. 81, 377-380.
Effect of estrogen and progesterone on the 1054. Noshiro, M., M. Nishimoto, and
hepatic cholesterol 7a-hydroxylase activity in K. Okuda (1990). Rat liver cholesterol
510 R Peter Guengerich

7a-hydroxylase: Pretranslational regulation for cir- 1067. Stahlberg, D., M. Rudling, B. Angelin, I. Bjorkhem,
cadian rhythm. J. Biol. Chem. 265, 10036-10041. P Forsell, K. Nilsell et al. (1997). Hepatic choles-
1055. Sanghvi, A., E. Grassi, V Warty, W. Diven, terol metabolism in human obesity. Hepatology 25,
C. Wight, and R. Lester (1981). Reversible 1447-1450.
activation-inactivation of cholesterol 7a- 1068. Post, S.M., E.C. de Wit, and H.M. Princen
hydroxylase possibly due to phosphorylation- (1997). Cafestol, the cholesterol-raising factor
dephosphorylation. Biochem. Biophys. Res. in boiled coffee, suppresses bile acid synthesis
Commun. 103, 886-892. by downregulation of cholesterol 7a-hydroxy-
1056. Goodwin, CD., B.W Cooper, and S. Margolis lase and sterol 27-hydroxylase in rat hepato-
(1982). Rat liver cholesterol 7a-hydroxylase: cytes. Arterioscler. Thromb. Vase. Biol. 17,
Modulation of enzyme activity by changes in 3064-3070.
phosphorylation state. J. Biol. Chem. 257, 1069. Pandak, W.M., C. Schwarz, PB. Hylemon,
4469-^472. D. Mallonee, K. Valerie, D.M. Heuman et al.
1057. Holsztynska, E.J. and D.J. Waxman (1987). (2001). Effects of CYP7A1 overexpression on cho-
Cytochrome P-450 cholesterol 7a-hydroxylase: lesterol and bile acid homeostasis. Am. J. Physiol.
Inhibition of enzyme deactivation by structurally Gastrointest. Liver Physiol. 281, G878-G889.
diverse calmodulin antagonists and phosphatase 1070. Sauter, G., M. Weiss, and R. Hoermann (1997).
inhibitors. Arch. Biochem. Biophys. 256, 543-559. Cholesterol 7a-hydroxylase activity in hypothy-
1058. Nguyen, L.B., S. Shefer, G. Salen, J.Y. Chiang, roidism and hyperthyroidism in humans. Horm.
and M. Patel (1996). Cholesterol 7a-hydroxylase Metah. Res. 29, 176-179.
activities from human and rat liver are modulated 1071. Pullinger, C.R., C. Eng, G. Salen, S. Shefer,
in vitro posttranslationally by phosphorylation/ A.K. Batta, S.K. Erickson et al. (2002). Human
dephosphorylation. Hepatology 24, 1468-1474. cholesterol 7a-hydroxylase (CYP7A1) defi-
1059. Karam, WG. and J.Y.L. Chiang (1992). Polymor- ciency has a hypercholesterolemic phenotype.
phisms of human cholesterol 7a-hydroxylase. J. Clin. Invest. 110, 109-117.
Biochem. Biophys. Res. Commun. 185, 588-595. 1072. Beigneux, A., A.F. Hofmann, and S.G. Young
1060. Setchell, K.D., M. Schwarz, N.C. O'Connell, (2002). Human CYP7A1 deficiency: Progress and
E.G. Lund, D.L. Davis, R. Lathe et al. (1998). enigmas. J. Clin. Invest. 110, 29-31.
Identification of a new inborn error in bile acid 1073. Stapleton, G., M. Steel, M. Richardson, J.O.
synthesis: Mutation of the oxysterol 7a-hydroxy- Mason, K.A. Rose, R.G.M. Morris et al. (1995).
lase gene causes severe neonatal liver disease. A novel cytochrome P450 expressed primarily in
J. Clin. Invest. 102, 1690-1703. brain. J. Biol. Chem. 270, 29739-29745.
1061. Hegele, R.A., J. Wang, S.B. Harris, J.H. Brunt, 1074. Rose, K.A., G. Stapleton, K. Dott, M.P Kieny,
T.K. Young, A.J. Hanley et al. (2001). Variable R. Best, M. Schwarz et al. (1997). Cyp7b, a novel
association between genetic variation in the brain cytochrome P450, catalyzes the synthesis of
CYP7 gene promoter and plasma lipoproteins in neurosteroids 7a-hydroxy dehydroepiandros-
three Canadian populations. Atherosclerosis 154, terone and 7a-hydroxy pregnenolone. Proc. Natl.
579-587. Acad Sci. USA 94, 4925-4930.
1062. Balasubramaniam, S., K.A. Mitropoulos, and 1075. Rose, K., A. Allan, S. Gauldie, G. Stapleton,
N.B. Myant (1975). The substrate for cholesterol L. Dobbie, K. Dott et al. (2001). Neurosteroid
7a-hydroxylase. Biochim. Biophys. Acta 398, hydroxylase CYP7B: Vivid reporter activity in
172-177. dentate gyrus of gene-targeted mice and aboli-
1063. Norlin, M., A. Toll, I. Bjorkhem, and K. Wikvall tion of a widespread pathway of steroid and
(2000). 24-hydroxycholesterol is a substrate for oxysterol hydroxylation. J. Biol. Chem. 276,
hepatic cholesterol 7a-hydroxylase (CYP7A). 23937-23944.
J. Lipid Res. 41, 1629-1639. 1076. Bunting, S., S. Moncada, and J.R. Vane (1983).
1064. Norlin, M., U. Andersson, I. Bjorkhem, and The prostacyclin-thromboxane A2 balance:
K. Wikvall (2000). Oxysterol 7a-hydroxylase Pathophysiological and therapeutic implications.
activity by cholesterol 7a-hydroxylase (CYP7A). Br Med Bull. 39,271-216.
J. Biol. Chem. 275, 34046-34053. 1077. Miyata, A., S. Hara, C. Yokoyama, H. Inoue,
1065. Lathe, R. (2002). Steroid and sterol 7-hydroxyla- V Ullrich, andT. Tanabe (1994). Molecular cloning
tion: Ancient pathways. Steroids 67, 967-977. and expression of human prostacyclin synthase.
1066. Nakayama, K., A. Puchkaev, and LA. Pikuleva Biochem. Biophys. Res. Commun. 200, 1728-1734.
(2001). Membrane binding and substrate access 1078. Ullrich, V, L. Castle, and P Weber (1981).
merge in cytochrome P450 7A1, a key enzyme in Spectral evidence for the cytochrome P450 nature
degradation of cholesterol. J. Biol. Chem. 276, of prostacyclin synthetase. Biochem. Pharmacol.
31459-31465. 30,2033-2036.
Human P450s 511

1079. DeWitt, D.L. and W.L. Smith (1983). Purification prostacyclin synthase gene variant with cerebral
of prostacyclin synthase from bovine aorta by infarction. Am. J. Hypertens. 13, 1263-1267.
immunoafifinity chromatography. Evidence that 1091. Nakayama, T., M. Soma, S. Saito, J. Honye,
the enzyme is a hemoprotein. J. Biol. Chem. 258, J. Yajima, D. Rahmutula et al. (2002). Association
3285-3293. of a novel single nucleotide polymorphism of the
1080. Kara, S., A. Miyata, C. Yokoyama, H. Inoue, prostacyclin synthase gene with myocardial
R. Brugger, F. Lottspeich et al. (1994). Isolation infarction. Am. Heart J. 143, 797-801.
and molecular cloning of prostacyclin synthase 1092. Nakayama, T., M. Soma, Y Takahashi,
from bovine endothelial cells. J. Biol Chem. 269, D. Rehemudula, H. Tobe, M. Sato et al. (2001).
19897-19903. Polymorphism of the promoter region of prosta-
1081. Spisni, E., G. Bartolini, M. Orlandi, B. Belletti, cyclin synthase gene is not related to essential
S. Santi, and V Tomasi (1995). Prostacyclin (PGj2) hypertension. Am. J. Hypertens. 14, 4 0 9 ^ 1 1 .
synthase is a constitutively expressed enzyme in 1093. Nakayama, T, M. Soma, D. Rahmutula, H. Tobe,
human endothelial cells. Exp. Cell Res. 219, M. Sato, J. Uwabo et al. (2002). Association
507-513. study between a novel single nucleotide polymor-
1082. Mehl, M., H.J. Bidmon, H. Hilbig, K. Zilles, phism of the promoter region of the prostacyclin
R. Dringen, and V Ullrich (1999). Prostacyclin synthase gene and essential hypertension.
synthase is localized in rat, bovine and human Hypertens. Res. 25, 65-68.
neuronal brain cells. Neurosci. Lett. 271,187-190. 1094. Nakayama, T, M. Soma, Y Watanabe, B. Hasimu,
1083. Siegle, I., T. Klein, M.H. Zou, R Fritz, and M. Sato, N. Aoi et al. (2002). Splicing mutation
M. Komhofif (2000). Distribution and cellular of the prostacyclin synthase gene in a family
localization of prostacyclin synthase in human associated with hypertension. Biochem. Biophys.
brain. J. Histochem. Cytochem. 48, 631-641. Res. Commun. 297, 1135-1139.
1084. Huang, J.C, F Arbab, K.J. Tumbusch, J.S. Goldsby, 1095. Hatae, T, S. Hara, C. Yokoyama, T. Yabuki,
N. Matijevic-Aleksic, and K.K. Wu (2002). Human H. Inoue, V Ullrich et al. (1996). Site-directed
fallopian tubes express prostacyclin (PGI) synthase mutagenesis of human prostacyclin synthase:
and cyclooxygenases and synthesize abundant PGI. Alteration of Cys"^^^ of the Cys-pocket, and
J. Clin. Endocrinol. Metab. 87, 4361^368. Glu347 and Arg350 of the EXXR motif FEBSLett.
1085. Yokoyama, C., T. Yabuki, H. Inoue, Y. Tone, 389, 268-272.
S. Hara, T. Hatae et al. (1996), Human gene 1096. Shyue, S.K., K.H. Ruan, L.H. Wang, and K.K. Wu
encoding prostacyclin synthase (PTGIS): (1997). Prostacyclin synthase active sites.
Genomic organization, chromosomal localization, Identification by molecular modeling-guided site-
and promoter activity. Genomics 36, 296-304. directed mutagenesis. J. Biol. Chem. 272,
1086. Nakayama, T., M. Soma, Y. Izumi, and 3657-3662.
K. Kanmatsuse (1996). Organization of the human 1097. Lin, Y, K.K. Wu, and K.H. Ruan (1998).
prostacyclin synthase gene. Biochem. Biophys. Characterization of the secondary structure and
Res. Commun. Ill, 803-806. membrane interaction of the putative membrane
1087. Wang, L.H. and L. Chen (1996). Organization anchor domains of prostaglandin I2 synthase and
of the gene encoding human prostacyclin cytochrome P450 2C1. Arch. Biochem. Biophys.
synthase. Biochem. Biophys. Res. Commun. 226, 352, 78-84.
631-637. 1098. Lin, YZ., H. Deng, and K.H. Ruan (2000).
1088. ChevaUer, D., C. Cauffiez, C. Bernard, Topology of catalytic portion of prostaglandin I2
J.M. Lo-Guidice, D. AUorge, F. Fazio et al. synthase: Identification by molecular modeling-
(2001). Characterization of new mutations in the guided site-specific antibodies. Arch. Biochem.
coding sequence and 5'-untranslated region of Biophys. 379, 188-197.
the human prostacylcin synthase gene 1099. Deng, H., A. Huang, S.R So, YZ. Lin, and
(CYP8A1). ifi/m. Genet. 108, 148-155. K.H. Ruan (2002). Substrate access channel
1089. Chevalier, D., D. Allorge, J.M. Lo-Guidice, topology in membrane-bound prostacyclin syn-
C. Cauffiez, C. Lepetit, F. Migot-Nabias et al. thase. Biochem. J. 362, 545-551.
(2002). Sequence analysis, frequency and ethnic 1100. Reed, G.A., I.O. Griffin, and T.E. Eling (1985).
distribution of VNTR polymorphism in the 5'- Inactivation of prostaglandin H synthase and
untranslated region of the human prostacyclin prostacyclin synthase by phenylbutazone.
synthase gene (CYP8A1). Prostaglandins Other Requirement for peroxidative metabolism. Mol.
Lipid Mediat. 70, 31-37. Pharmacol. 21, 109-114.
1090. Nakayama, T, M. Soma, D. Rehemudula, 1101. Wade, M.L., N.F Voelkel, and FA. Fitzpatrick
Y Takahashi, H. Tobe, M. Satoh et al. (2000). (1995). "Suicide" inactivation of prostaglandin I2
Association of 5' upstream promoter region of synthase: Characterization of mechanism-based
512 F. Peter Guengerich

inactivation with isolated enzyme and endothelial human and mouse: Eukaryotic cytochrome P-450
cells. Arch. Biochem. Biophys. 321, 453-458. gene devoid of introns. Genomics 56, 184-196.
1102. Zou, M.H. and V Ullrich (1996). Peroxynitrite 1113. Zhang, M. and J.Y Chiang (2001).
formed by simultaneous generation of nitric Transcriptional regulation of the human sterol
oxide and superoxide selectively inhibits bovine 12a-hydroxylase gene (CYPSBl): Roles of
aortic prostacyclin synthase. FEBS Lett. 382, hepatocyte nuclear factor 4a in mediating
101-104. bile acid repression. J. Biol. Chem. 276,
1103. Crow, J.P. and J.S. Beckman (1995). Reactions 41690-41699.
between nitric oxide, superoxide, and peroxyni- 1114. Gerbod-Giannone, M.C., A. Del Castillo-
trite: Footprints of peroxynitrite in vivo. Adv. Olivares, S. Janciauskiene, G. Gil, and
Pharmacol. 34, 1 7 ^ 3 . P.B. Hylemon (2002). Suppression of cholesterol
1104. Zou, M., C. Martin, and V Ullrich (1997). 7a-hydroxylase transcription and bile acid syn-
Tyrosine nitration as a mechanism of selective thesis by an a j-antitrypsin peptide via interaction
inactivation of prostacyclin synthase by peroxyni- with a J-fetoprotein transcription factor. J. Biol.
trite. Biol. Chem. 378, 707-713. Chem. 211, 42973^2980.
1105. Schmidt, P., N. Youhnovski, A. Daiber, A. Balan, 1115. Yang, Y, M. Zhang, G. Eggertsen, and J.Y Chiang
M. Arsic, M. Bachschmid et al. (2003). Specific (2002). On the mechanism of bile acid inhibition
nitration at tyrosine-430 revealed by high resolu- of rat sterol 12a-hydroxylase gene (CYPSBl)
tion mass spectrometry as basis for redox regula- transcription: Roles of a-fetoprotein transcription
tion of bovine prostacyclin synthase. J. Biol. factor and hepatocyte nuclear factor 4a. Biochim.
Chem. 278, 12813-12819. Biophys. Acta 1583, 63-73.
1106. Tuder, R.M., CD. Cool, M.W. Geraci, J. Wang, 1116. Wang, H.P and T. Kimura (1976). Purification
S.H. Abman, L. Wright et al. (1999). Prostacyclin and characterization of adrenal cortex mitochon-
synthase expression is decreased in lungs from drial cytochrome P-450 specific for cholesterol
patients with severe pulmonary hypertension. side chain cleavage activity. J. Biol. Chem. 251,
Am. J. Respir. Crit. Care Med. 159, 1925-1932. 6068-6074.
1107. Iwai, N., T. Katsuya, K. Ishikawa, T. Mannami, 1117. Morohashi, K., K. Sogawa, T Omura, and
J. Ogata, J. Higaki et al. (1999). Human prostacy- Y Fujii-Kuriyama (1987). Gene structure of
clin synthase gene and hypertension: The Suita human cytochrome P-450(SCC), cholesterol
study Circulation 100, 2231-2236. desmolase. J. Biochem. (Tokyo) 101, 8879-8887.
1108. Geraci, M.W, B. Gao, D.C. Shepherd, M.D. Moore, 1118. Poulos, T.L., B.C. Finzel, l.C. Gunsalus,
J.Y. Westcott, K.A. Pagan et al. (1999). Pulmonary G.C. Wagner, and J. Kraut (1985). The 2.6-A crys-
prostacyclin synthase overexpression in transgenic tal structure of Pseudomonas putida cytochrome
mice protects against development of hypoxic P-450. J. Biol. Chem. 260, 16122-16130.
pulmonary hypertension. J. Clin. Invest. 103, 1119. Belfiore, C.J., D.E. Hawkins, M.C. Wiltbank, and
1509-1515. G.D. Niswender (1994). Regulation of
1109. Todaka, T, C. Yokoyama, H. Yanamoto, cytochrome P450g^^ synthesis and activity in the
N. Hashimoto, I. Nagata, T. Tsukahara et al. ovine corpus luteum. J. Steroid Biochem. Mol.
(1999). Gene transfer of human prostacyclin syn- Biol. 51,283-290.
thase prevents neointimal formation after carotid 1120. Matocha, M.F and M.R. Waterman (1986).
balloon injury in rats. Stroke 30, 419-426. Import and processing of P-450g^,^ and P-4501 Ip
1110. Pradono, P., R. Tazawa, M. Maemondo, precursors by corpus luteal mitochondria: A pro-
M. Tanaka, K. Usui, Y Saijo et al. (2002). Gene cessing pathway recognizing homologous and
transfer of thromboxane A2 synthase and heterologous precursors. Arch. Biochem. Biophys.
prostaglandin I2 synthase antithetically altered 250, 456^60.
tumor angiogenesis and tumor growth. Cancer 1121. Sasano, H., M. Okamoto, J.l. Mason,
Res 62, 63-66. E.R. Simpson, C.R. Mendelson, N. Sasano et al.
nil. Leeder, J.S., X. Lu, Y Timsit, and A. Gaedigk (1989). Immunolocalization of aromatase, 17a-
(1998). Non-monooxygenase cytochromes P450 hydroxylase and side-chain-cleavage cytochromes
as potential human auto antigens in anticonvulsant P-450 in the human ovary. J. Reprod. Fertil. 85,
hypersensitivity reactions. Pharmacogenetics 8, 163-169.
211-225. 1122. Chung, B.C., l.C. Guo, and S.J. Chou (1997).
1112. Gafvels, M., M. Olin, B.P Chowdhary, Transcriptional regulation of the CYPllAl and
T. Raudsepp, U Andersson, B. Persson et al. ferredoxin genes. Steroids 62, 3 7 ^ 2 .
(1999). Structure and chromosomal assignment 1123. Walther, B., J.R Ghersi-Egea, A. Minn, and
of the sterol 12a-hydroxylase gene (CYP8B1) in G. Siest (1987). Brain mitochondrial cytochrome
Human P450s 513

P-450g^^: Spectral and catalytic properties. Arch. 1136. Liu, Z. and E.R. Simpson (1999). Molecular
Biochem. Biophys. 254, 592-596. mechanism for cooperation between Spl and
1124. Warner, M. and J.A. Gustafsson (1995). steroidogenic factor-1 (SF-1) to regulate bovine
Cytochrome P450 in the brain: Neuroendocrine CYPl 1A gene expression. Mol. Cell Endocrinol.
functions. Front. Neuroendocrinol. 16, 224-236. 153,183-196.
1125. Beyenburg, S., B. Stoffel-Wagner, M. Watzka, 1137. Gizard, F, B. Lavallee, F DeWitte, and D.W Hum
I. Blumcke, J. Bauer, J. Schramm et al. (1999). (2001). A novel zinc finger protein TReP-132 inter-
Expression of cytochrome P450g^^ mRNA in the acts with CBP/p300 to regulate human CYPllAl
hippocampus of patients with temporal lobe gene expression. J. Biol. Chem. 276, 33881-33892.
epilepsy. Neuroreport 10, 3067-3070. 1138. Gizard, F, B. Lavallee, F DeWitte, E. Teissier,
1126. Watzka, M., F. Bidlingmaier, J. Schramm, B. Staels, and D.W. Hum (2002). The transcrip-
D. KlingmuUer, and B. StofiFel-Wagner (1999). tional regulating protein of 132 kDa (TReP-132)
Sex- and age-specific differences in human brain enhances P450scc gene transcription through
CYPllAl mRNA expression. J. Neuroendocrinol. interaction with steroidogenic factor-1 in human
II, 901-905. adrenal cells. J. Biol. Chem. Ill, 39144-39155.
1127. Morales, A., A. Cuellar, J. Ramirez, F. Vilchis, 1139. Doi, J., H. Takemori, X.Z. Lin, N. Horike,
and V Diaz-Sanchez (1999). Synthesis of steroids Y Katoh, and M. Okamoto (2002). Salt-inducible
in pancreas: Evidence of cytochrome P-450g^^ kinase represses cAMP-dependent protein
activity. Pancreas 19, 39-44. kinase-mediated activation of human cholesterol
1128. Ou, W, A. Ito, K. Morohashi, Y. Fujii-Kuriyama, side chain cleavage cytochrome P450 promoter
and T. Omura (1986). Processing-independent through the CREB basic leucine zipper domain.
in vitro translocation of cytochrome P-450g(^j^ J. Biol. Chem. Ill, 15629-15637.
precursor across mitochondrial membranes. 1140. Ben-Zimra, M., M. Koler, and J. Orly (2002).
J. Biochem. (Tokyo) 100, 1287-1296. Transcription of cholesterol side-chain cleavage
1129. Kumamoto, T., K. Morohashi, A. Ito, and cytochrome P450 in the placenta: Activating
T. Omura (1987). Site-directed mutagenesis of protein-2 assumes the role of steroidogenic
basic amino acid residues in the extension peptide factor-1 by binding to an overlapping promoter
of P-450g(^(-, precursor: Effects on the import of element. Mol. Endocrinol. 16, 1864-1880.
the precursor into mitochondria. J. Biochem. 1141. Katsumata, N., M. Ohtake, T. Hojo, E. Ogawa,
(Tokyo) 102, 833-838. T. Hara, N. Sato et al. (2002). Compound het-
1130. Black, S.M., J.A. Harikrishna, G.D. Szklarz, and erozygous mutations in the cholesterol side-chain
WL. Miller (1994). The mitochondrial environ- cleavage enzyme gene (CYPl 1 A) cause congeni-
ment is required for activity of the cholesterol tal adrenal insufficiency in humans. J. Clin.
side-chain cleavage enzyme, cytochrome P450g^^.. Endocrinol. Metab. 87, 3808-3813.
Proc. Natl. Acad. Sci. USA 91, 7247-7251. 1142. Tuckey, R.C. and K.J. Cameron (1993). Human
1131. Venepally, R and M.R. Waterman (1995). Two placental cholesterol side-chain cleavage:
Spl-binding site mediate cAMP-induced tran- Enzymatic synthesis of (22/?)-20a,22-dihydroxy-
scription of the bovine CYPl 1A gene through the cholesterol. Steroids 58, 230-233.
protein kinase A signaling pathway. J. Biol. 1143. Murray, R.I. and S.G. Sligar (1985). Oxidative
Chem. 270, 25402-25410. cleavage of 1-phenyl-1,2-ethanediol by 4-cyano-
1132. Ahlgren, R., G. Suske, M.R. Waterman, and A^,A^-dimethylaniline N-oxide and chloro
J. Lund (1999). Role of Spl in cAMP-dependent (5,10,15,20-tetraphenylporphinato) chromium
transcriptional regulation of the bovine CYPl 1A (III): A model for cholesterol side-chain cleavage
gene. J. Biol. Chem. 274, 19422-19428. by cytochrome P-450g^,^. J. Am. Chem. Soc. 107,
1133. Guo, I.e. and B.C. Chung (1999). Cell-type 2186-2187.
specificity of human CYPllAl TATA box. 1144. Okamoto, T., K. Sasaki, and S. Oka (1988).
J. Steroid Biochem. Mol. Biol. 69, 329-334. Biomimetic oxidation with molecular oxygen.
1134. Huang, Y, M. Hu, N. Hsu, C.L. Wang, and Selective carbon-carbon bond cleavage of 1,2-
B. Chung (2001). Action of hormone responsive diols by molecular oxygen and dihydropyridine in
sequence in 2.3 kb promoter of CYPllAl. Mol. the presence of iron-porphyrin catalysts. J. Am.
Cell Endocrinol. 175,205-210. Chem. Soc. 110, 1187-1196.
1135. Hu, M.C., N.C. Hsu, C.I. Pai, C.K. Wang, and 1145. Ortiz de Montellano, PR. (1995). Oxygen activa-
B. Chung (2001). Functions of the upstream and tion and reactivity. In RR. Ortiz de Montellano
proximal steroidogenic factor 1 (SF-l)-binding sites (ed.), Cytochrome P450: Structure, Mechanism,
in the CYPllAl promoter in basal transcription and and Biochemistry. Plenum Press, New York,
hormonal response. Mol. Endocrinol. 15, 812-818. pp. 245-303.
514 F. Peter Guengerich

1146. Tuckey, R.C., S.T. Woods, and M. Tajbakhsh 1158. Hu, M.C., N.C. Hsu, N.B. El Hadj, C.I. Pai,
(1997). Electron transfer to cytochrome P-450g^^ H.P Chu, C.K. Wang et al. (2002). Steroid
limits cholesterol-side-chain-cleavage activity in deficiency syndromes in mice with targeted disrup-
the human placenta. Eur. J. Biochem. 244, tion of Cyp] J al. Mol. Endocrinol. 16, 1943-1950.
835-839. 1159. Chen, S., J. Sawicka, C. Betterle, M. Powell,
1147. Beckert, V and R. Bernhardt (1997). Specific L. Prentice, M. Volpato et al. (1996).
aspects of electron transfer from adrenodoxin to Autoantibodies to steroidogenic enzymes in
cytochromes P450g^^ and P450jjg. J. Biol. Chem. autoimmune polyglandular syndrome, Addison's
272, 4883-4888. disease, and premature ovarian failure. J. Clin.
1148. Cao, P. and R. Bernhardt (1999). Interaction of Endocrinol. Metab. 81, 1871-1876.
CYPllBl (cytochrome P-450,,p) with 1160. Seissler, J., M. Schott, H. Steinbrenner,
CYPl 1 Al (cytochrome P-450^ J in COS-1 cells. P Peterson, and WA. Scherbaum (1999).
Eur. J. Biochem. 262, 720-726. Autoantibodies to adrenal cytochrome P450 anti-
1149. Usanov, S.A. and VL. Chashchin (1991). gens in isolated Addison's disease and autoim-
Interaction of cytochrome P-450^^^ with mune polyendocrine syndrome type II. Exp. Clin.
cytochrome b^. FEBS Lett. 278, 279-282. Endocrinol. Diabetes 107, 208-213.
1150. Wada, A. and M.R. Waterman (1992). 1161. Bureik, M., M. Lisurek, and R. Bernhardt (2002).
Identification by site-directed mutagenesis of two The human steroid hydroxylases CYPllBl and
lysine residues in cholesterol side chain cleavage CYP11B2. Biol. Chem. 383, 1537-1551.
cytochrome P450 that are essential for adreno- 1162. Watanuki, M., B.E. Tilley and PE Hall (1978).
doxin binding. J. Biol. Chem. 267, 22877-22882. Cytochrome P-450 for 11(3- and 18-hydroxylase
1151. Woods, S.T, J. Sadleir, T. Downs, activities of bovine adrenocortical mitochondria:
T. Triantopoulos, M.J. Headlam, and R.C. Tuckey One enzyme or two?. Biochemistry 17, 127-130.
(1998). Expression of catalytically active human 1163. Mornet, E., J. Dupont, A. Vitek, and PC. White
cytochrome P450g^^ in Escherichia coli and muta- (1989). Characterization of two genes encoding
genesis of isoleucine-462. Arch. Biochem. human steroid 11 beta-hydroxylase (P-450(ll)p).
Biophys. 353, 109-115. J. Biol. Chem. 264, 20961-20967.
1152. Vickery, L.E. and J.T Kellis (1983). Inhibition of 1164. Kawamoto, T, Y Mitsuuchi, K. Toda,
adrenocortical cytochrome P-450^^^ by (20/?)-20- K. Miyahara, Y Yokoyama, K. Nakao et al.
phenyl-5-pregnene-3(3,20-diol: Mechanism and (1990). Cloning of cDNA and genomic DNA for
implications for the structure of the active site. human cytochrome P-450,,^. FEBS Lett. 269,
J. Biol. Chem. 258, 3832-3836. 345-349.
1153. Vickery, L.E. and J. Singh (1988). 22- 1165. Zhang, G. and WL. Miller (1996). The human
Thio-23,24-bisnor-5-cholen-3 P-ol: An active genome contains only two CYPl IB (P450cll)
site-directed inhibitor of cytochrome P450j,^^. genes. ^ Clin. Endocrinol. Metab. 81, 3254-3256.
J. Steroid Biochem. 29, 539-543. 1166. Kawamoto, T, Y Mitsuuchi, K. Toda,
1154. Olakanmi, O. and D.W Seybert (1990). Modified Y Yokoyama, K. Miyahara, S. Miura et al.
acetylenic steroids as potent mechanism-based (1992). Role of steroid 11 (3-hydroxylase and
inhibitors of cytochrome P-450g^^. J. Steroid steroid 18-hydroxylase in the biosynthesis of glu-
Biochem. 36, 273-280. cocorticoids and mineralocorticoids in humans.
1155. Jarman, M., S.E. Barrie, C.S. Leung, and M.G. Proc. Natl. Acad Sci. USA 89, 1458-1462.
Rowlands (1988). Selective inhibition of choles- 1167. Freije, W.A., V Pezzi, A. Arici, B.R. Carr, and
terol side-chain cleavage by potential pro-drug WE. Rainey (1997). Expression of 11 beta-
forms of aminoglutethimide. Anticancer Drug hydroxylase (CYPllBl) and aldosterone syn-
Design 3, 185-190. thase (CYP11B2) in the human fetal adrenal.
1156. Ohnishi, T and Y. Ichikawa (1997). Direct J. Soc. Gynecol. Investig. 4, 305-309.
inhibitions of the activities of steroidogenic 1168. Morohashi, K., U.M. Zanger, S. Honda, M. Hara,
cytochrome P-450 mono-oxygenase systems by M.R. Waterman, andT Omura (1993). Activation
anticonvulsants. J. Steroid Biochem. Mol. Biol. of CYPl 1A and CYPl 1B gene promoters by the
60, 77-85. steroidogenic cell-specific transcription factor,
1157. Yang, X., K. Iwamoto, M. Wang, J. Artwohl, Ad4BR Mol. Endocrinol. 7, 1196-1204.
J.I. Mason, and S. Pang (1993). Inherited congen- 1169. Honda, S., K. Morohashi, M. Nomura, H. Takeya,
ital adrenal hyperplasia in the rabbit is caused M. Kitajima, and T Omura (1993). Ad4BP regu-
by a deletion in the gene encoding cytochrome lating steroidogenic P-450 gene is a member of
P450 cholesterol side-chain cleavage enzyme. steroid hormone receptor superfamily. J. Biol.
Endocrinology 132, 1977-1982. Chem. 268, 7494-7502.
Human P450s 515

1170. Hashimoto, T., K. Morohashi, K. Takayama, splice donor sites of CYPl IB 1 in congenital
5. Honda, T. Wada, H. Handa et al (1992). adrenal hyperplasia due to 11 p-hydroxylase
Cooperative transcription activation between deficiency. Endocrinol. Res. 26, 797-801.
Adl, a CRE-like element, and other elements in 1181. Mulatero, P., K.M. Curnow, B. Aupetit-Faisant,
the CYPl IB gene promoter. J. Biochem. (Tokyo) M. Foekling, C. Gomez-Sanchez, F. Veglio,
112, 573-575. X. Jeunemaitre et al. (1998). Recombinant
1171. Mukai, K., F. Mitani, R. Agake, and Y. Ishimura CYPl IB genes encode enzymes that can catalyze
(1998). Adrenocorticotropic hormone stimulates conversion of 11-deoxycortisol to Cortisol, 18-
CYPl IB 1 gene transcription through a mecha- hydroxycortisol, and 18-oxocortisol. J. Clin.
nism involving AP-1 factors. Eur. J. Biochem. Endocrinol. Metab. 83, 3996-4001.
256, 190-200. 1182. Fisher, A., E. Davies, R. Eraser, and J.M. Connell
1172. Wang, X.L., M. Bassett, Y Zhang, S. Yin, C. Clyne, (1998). Structure-function relationships of aldos-
PC. White et al. (2000). Transcriptional regu- terone synthase and 11 ^-hydroxylase enzymes:
lation of human 11 ^-hydroxylase QiCYPllBl). Implications for human hypertension. Clin. Exp.
Endocrinology 141, 3587-3594. Pharmacol. Physiol. Suppl. 25, S42-S46.
1173. Bassett, M.H., Y Zhang, C. Clyne, PC. White, 1183. Fisher, A., R. Eraser, J. McConnell, and E. Davies
and WE. Rainey (2002). Differential regulation (2000). Amino acid residue 147 of human aldos-
of aldosterone synthase and 11 (^-hydroxylase terone synthase and 11 (3-hydroxylase plays a key
transcription by steroidogenic factor-1. J. Mol. role in 11 ^-hydroxylation. J. Clin. Endocrinol.
Endocrinol. 28, 125-135. Metab. 85, 1261-1266.
1174. Skinner, C.A. and G. Rumsby (1994). Steroid 11 1184. Bottner, B., K. Denner, and R. Bernhardt (1998).
beta-hydroxylase deficiency caused by a five Conferring aldosterone synthesis to human
base pair duplication in the CYPl IB 1 gene. CYPl IB 1 by replacing key amino acid residues
Hum. Mol. Genet. 3, 377-378. with CYPllB2-specific ones. Eur. J. Biochem.
1175. Curnow, K.M., L. Slutsker, J. Vitek, T. Cole, 252, 458^66.
PW Speiser, M.I. New et al. (1993). Mutations in 1185. Lewis, D.E and P Lee-Robichaud (1998). Mole-
the CYPl IB 1 gene causing congenital adrenal cular modelling of steroidogenic cyto-chromes
hyperplasia and hypertension cluster in exons P450 from families CYPll, CYP17, CYP19 and
6, 7, and 8. Proc. Natl. Acad. Sci. USA 90, CYP21 based on the CYPl02 crystal structure.
4552-4556. J. Steroid Biochem. Mol. Biol. 66, 217-233.
1176. Lifton, R.P, R.G. Dluhy, M. Powers, G.M. Rich, 1186. Belkina, N.V, M. Lisurek, A.S. Ivanov, and
S. Cook, S. UHck et al. (1992). A chimaeric 11 (3- R. Bernhardt (2001). Modelling of three-
hydroxylase/aldosterone synthase gene causes dimensional structures of cytochromes P450
glucocorticoid-remediable aldosteronism and l l B l and 11B2. J. Inorg. Biochem. 87, 197-207.
human hypertension. Nature 355, 262-265. 1187. Denner, K., R. Vogel, W Schmalix, J. Doehmer,
1177. Pascoe, L., K.M. Curnow, L. Slutsker, and R. Bernhardt (1995). Cloning and stable
J.M. Connell, PW. Speiser, M.I. New et al. (1992). expression of the human mitochondrial cyto-
Glucocorticoid-suppressible hyperaldosteronism chrome P45011B1 cDNA in V79 Chinese ham-
results from hybrid genes created by unequal ster cells and their application for testing of
crossovers between CYPl IB 1 and CYP11B2, potential inhibitors. Pharmacogenetics 5, 89-96.
Proc. Natl. Acad Sci. USA 89, 8327-8331. 1188. Johnston, J.O., C.L. Wright, and G.W Holbert
1178. Hampf, M., N.T. Dao, N.T. Hoan, and (1995). Enzyme-activated inhibitors of steroidal
R. Bernhardt (2001). Unequal crossing-over hydroxylases. J. Steroid Biochem. Mol. Biol. 52,
between aldosterone synthase and 11 p-hydroxy- 17-34.
lase genes causes congenital adrenal hyperplasia. 1189. Delorme, C , A. Piffeteau, E Sobrio, and
J. Clin. Endocrinol. Metab. 86, 4445^452. A. Marquet (1997). Mechanism-based inactivation
1179. Portrat, S., P Mulatero, K.M. Curnow, of bovine cytochrome P450jjp by 18-unsaturated
J.L. Chaussain, Y Morel, and L. Pascoe (2001). progesterone derivatives. Eur. J. Biochem. 248,
Deletion hybrid genes, due to unequal crossing 252-260.
over between CYPl IB 1 (11(3-hydroxylase) and 1190. White, PC. (2001). Steroid 11 ^-hydroxylase
CYP11B2 (aldosterone synthase) cause steroid deficiency and related disorders. Endocrinol.
11 p-hydroxylase deficiency and congenital adre- Metab. Clin. North Am. 30, 61-79.
nal hyperplasia. J. Clin. Endocrinol. Metab. 86, 1191. Peter, M., J.M. Dubuis, and WG. Sippell (1999).
3197-3201. Disorders of the aldosterone synthase and steroid
1180. Chabre, O., S. Portrat-Doyen, J. Vivier, Y Morel, 11 ^-hydroxylase deficiencies. Horm. Res. 51,
and G. Defaye (2000). Two novel mutations in 211-222.
516 F. Peter Guengerich

1192. Pascoe, L., K.M. Curnow, L. Slutsker, A. Rosier, corticosterone methyl oxidase II deficiency. Am.
and P.C. White (1992). Mutations in the human J. Hum. Genet. 57, 1037-1043.
CYP11B2 (aldosterone synthase) gene causing 1204. Mulatero, P, D. Schiavone, F. Fallo, F Rabbia,
corticosterone methyloxidase II deficiency. Proc. C. Pilon, L. Chiandussi et al. (2000). CYPl 1B2
Natl. Acad. Sci. USA 89, 4996-5000. gene polymorphisms in idiopathic hyperaldos-
1193. Mitsuuchi, Y., T. Kawamoto, K. Miyahara, teronism. Hypertension 35, 694-698.
S. Ulick, D.H. Morton, Y. Naiki et al. (1993). 1205. Tsukada, K., T. Ishimitsu, M. Teranishi, M. Saitoh,
Congenitally defective aldosterone biosynthesis M. Yoshii, H. Inada et al. (2002). Positive associa-
in humans: Inactivation of the P-450C18 gene tion of CYPl 1B2 gene polymorphism with genetic
(CYPl 1B2) due to nucleotide deletion in CMO I predisposition to essential hypertension. J. Hum.
deficient patients. Biochem. Biophys. Res. Hypertens. 16, 789-793.
Commun. 190, 864-869. 1206. Bechtel, S., N. Belkina, and R. Bernhardt (2002).
1194. Kawamoto, T., Y Mitsuuchi, T. Ohnishi, The effect of amino-acid substitutions II12P, D147E
Y Ichikawa, Y Yokoyama, H. Sumimoto et al. and K152N in CYPl 1B2 on the catalytic activities
(1990). Cloning and expression of a cDNA for of the enzyme. Eur J. Biochem. 269, 1118-1127.
human cytochrome P-450^J^Q as related to 1207. Ehmer, PB., M. Bureik, R. Bernhardt, U. Muller,
primary aldosteronism. Biochem. Biophys. Res. and R.W Hartmann (2002). Development of a
Commun. 173,309-316. test system for inhibitors of human aldosterone
1195. Curnow, K.M., M.T. Tusie-Luna, L. Pascoe, synthase (CYP11B2): Screening in fission yeast
R. Natarajan, J.L. Gu, J.L. Nadler et al. (1991). and evaluation of selectivity in V79 cells.
The product of the CYPl 1B2 gene is required for J. Steroid Biochem. Mol. Biol. 81, 173-179.
aldosterone biosynthesis in the human adrenal 1208. GrifFing, G.T, M. Holbrook, J.C. Melby J. Alberta,
cortex. Mol. Endocrinol. 5, 1513-1522. and N.R. Orme-Johnson (1989). 19-Hydroxylase
1196. Li, X., Y Meng, X.S. Yang, PS. Wu, S.M. Li, and inhibition of adrenal mitochondrial P450
W.Y Lai (2000). CYP11B2 expression in HSCs 1 ip/18/19-hydroxylase by a suicide inhibitor. Am.
and its effect on hepatic fibrogenesis. World J. Med Sci. 298, 83-88.
J. Gastroenterol. 6, 885-887. 1209. Davies, E., CD. Holloway, M.C Ingram,
1197. Bassett, M.H., Y Zhang, PC. White, and G.C Inglis, E.C Friel, C Morrison et al. (1999).
WE. Rainey (2000). Regulation of human Aldosterone excretion rate and blood pressure in
CYPl 1B2 and CYPl IBl: Comparing the role of essential hypertension are related to polymorphic
the common CRE/Adl element. Endocrinol. Res. differences in the aldosterone synthase gene
26,941-951. CYPl 1B2. Hypertension 33, 703-707.
1198. Clyne, CD., PC. White, and WE. Rainey (1996). 1210. White, P C , A. Hautanen, and M. Kupari (1999).
Calcium regulates human CYP11B2 transcrip- Aldosterone synthase (CYP11B2) polymor-
tion. Endocrinol. Res. 22, 485-492. phisms and cardiovascular function. J. Steroid
1199. Bureik, M., A. Zeeh, and R. Bernhardt (2002). Biochem. Mol. Biol. 69, 409-412.
Modulation of steroid hydroxylase activity in sta- 1211. Kupari, M., A. Hautanen, L. Lankinen,
bly transfected V79MZhl 1 Bl and V79MZhl 1B2 P. Koskinen, J. Virolainen, H. Nikkila et al.
cells by PKC and PKD inhibitors. Endocrinol. (1998). Associations between human aldosterone
Res. 28,351-355. synthase (CYP11B2) gene polymorphisms and
1200. J.G. LeHoux, G. Dupuis, and A. Lefebvre (2000). left ventricular size, mass, and function.
Regulation of CYPl 1B2 gene expression by pro- Circulation 97, 569-575.
tein kinase C. Endocrinol. Res. 26, 1027-1031. 1212. Satoh, M., M. Nakamura, H. Saitoh, H. Satoh,
1201. Ise, T., A. Shimoda, H. Takakuwa, T. Kato, T Akatsu, J. Iwasaka et al. (2002). Aldosterone
Y Izumiya, K. Shimizu et al. (2001). A chimeric synthase (CYP11B2) expression and myocardial
CYP11B1/CYP11B2 gene in glucocorticoid- fibrosis in the failing human heart. Clin. Sci.
insuppressible familial hyperaldosteronism. Clin. (Zo«Jo«; 102,381-386.
Endocrinol. (Oxford) 55, 131-134. 1213. Russo, P, A. Siani, A. Venezia, R. lacone,
1202. Jackson, R.V, A. Laflferty, D.J. Torpy, and O. Russo, G. Barba et al. (2002). Interaction
C. Stratakis (2002). New genetic insights in between the C(-344)T polymorphism of CYPl 1B2
familial hyperaldosteronism. Ann. N. Y. Acad. Sci. and age in the regulation of blood pressure and
970, 77-88. plasma aldosterone levels: Cross-sectional and lon-
1203. Zhang, G., H. Rodriguez, C.E. Fardella, gitudinal findings of the Olivetti Prospective Heart
D.A. Harris, and WL. Miller (1995). Mutation Study J. Hypertens. 20, 1785-1792.
T318M in the CYP11B2 gene encoding 1214. Lim, PO., T.M. Macdonald, C Holloway,
P450cllAS (aldosterone synthase) causes E. Friel, N.H. Anderson, E. Dow et al. (2002).
Human P450s 517

Variation at the aldosterone synthase (CYPl 1B2) by an androgen receptor-mediated mechanism.


locus contributes to hypertension in subjects with J. Biol. Chem. 262, 11200-11206.
a raised aldosterone-to-renin ratio. J. Clin. 1226. Zhang, P, X.G. Han, S.H. Mellon, and PR Hall
Endocrinol. Metab. 87, 4398^402. (1996). Expression of the gene for cytochrome
1215. Tsujita, Y., N. Iwai, T. Katsuya, J. Higaki, P-450 17a-hydroxylase/C 17-20 lyase (CYPl7)
T. Ogihara, S. Tamaki et al (2001). Lack of in porcine Leydig cells: Identification of a DNA
association between genetic polymorphism of sequence that mediates cAMP response. Biochim.
CYP11B2 and hypertension in Japanese: The Biophys. Acta 1307, 73-82.
Suita Study. Hypertens. Res. 24, 105-109. 1227. Ogo, A., M.R. Waterman, J.M. McAllister, and
1216. Chung, B., J. Picado-Leonard, M. Haniu, N. Kagawa (1997). The homeodomain protein
M. Bienkowski, PR Hall, J.E. Shively et al. (1987). Pbxl is involved in cAMP-dependent transcrip-
Cytochrome P450cl7 (steroid 17a-hydroxylase/17, tion of human CYPl7. Arch. Biochem. Biophys.
20 lyase): Cloning of human adrenal and testis 348,226-231.
cDNAs indicates the same gene is expressed in both 1228. Bischof, L.J., N. Kagawa, and M.R. Waterman
tissues. Proc. Natl. Acad. Sci. USA 84, 407-411. (1998). The bovine CYPl7 promoter contains
1217. Picado-Leonard, J. and W.L. Miller (1987). Cloning a transcriptional regulatory element coopera-
and sequence of the human gene for P450cl7 tively bound by tale homeodomain proteins.
(steroid 17 a-hydroxylase/17, 20 lyase): Similarity Endocrinol. Res. 24, 489^95.
with the gene for P450c21. DNA 6, 439-448. 1229. Lin, C.J., J.W. Martens, and W.L. Miller (2001).
1218. Nakajin, S., J.E. Shively, PM. Yuan, and PR Hall NP-IC, Spl, and Sp3 are essential for transcrip-
(1981). Microsomal cytochrome P-450 from tion of the human gene for P450cl7 (steroid 17a-
neonatal pig testis: Two enzymatic activities hydroxylase/17, 20 lyase) in human adrenal
(17a-hydroxylase and C17,20-lyase) associated NCI-H295A cells. Mol. Endocrinol. 15,
with one protein. Biochemistry 20, 4037^042. 1277-1293.
1219. Katagiri, M., K. Suhara, M. Shiroo, and 1230. Sewer, M.B., VQ. Nguyen, C.J. Huang,
Y. Rujimura (1982). Role of cytochrome b^ in the PW Tucker, N. Kagawa, and M.R. Waterman
cytochrome P-450-mediated C21-steroid 17,20- (2002). Transcriptional activation of human
lyase reaction. Biochem. Biophys. Res. Commun. CYPl7 in H295R adrenocortical cells depends on
108, 379-384. complex formation among p5'^"'^'^/NonO, protein-
1220. Shinzawa, K., S. Kominami, and S. Takemori associated splicing factor, and SR-1, a complex
(1985). Studies on cytochrome P-450 (P-45017a, that also participates in repression of transcrip-
lyase) from guinea pig adrenal microsomes. tion. Endocrinology 143, 1280-1290.
Dual function of a single enzyme and effect of 1231. Sewer, M.B. and M.R. Waterman (2002).
cytochrome b^. Biochim. Biophys. Acta 833, Adrenocorticotropin/cyclic adenosine 3' ,5' -
151-160. monophosphate-mediated transcription of the
1221. Casey, M.L. and PC. MacDonald (1982). human CYPl7 gene in the adrenal cortex is
Demonstration of steroid 17a-hydroxylase activ- dependent on phosphatase activity. Endocrinology
ity in human fetal kidney, thymus, and spleen. 143, 1769-1777.
Steroids 40, 91-91. 1232. Sewer, M.B. and M.R. Waterman (2002). cAMP-
Mil. Kayes-Wandover, K.M. and PC. White (2000). dependent transcription of steroidogenic genes
Steroidogenic enzyme gene expression in the in the human adrenal cortex requires a dual-
human heart. J. Clin. Endocrinol. Metab. 85, specificity phosphatase in addition to protein
2519-2525. kinase A. J. Mol. Endocrinol. 29, 163-174.
1223. Puche, C , M. Jose, A. Cabero, and A. Meseguer 1233. Sewer, M.B. and M.R. Waterman (2003). cAMP-
(2002). Expression and enzymatic activity of the dependent protein kinase enhances CYPl7
P450cl7 gene in human adipose tissue. Eur transcription via MKP-1 activation in H295R
J. Endocrinol. 146, 223-229. human adrenocortical cells. J. Biol. Chem. 278,
1224. Kusano, K., M. Sakaguchi, N. Kagawa, 8106-8111.
M.R. Waterman, and T Omura (2001). 1234. Yanase, T. (1995). 17a-Hydroxylase/17,20-lyase
Microsomal P450s use specific proline-rich defects. J. Steroid Biochem. Mol. Biol. 53,
sequences for efficient folding, but not for main- 153-157.
tenance of the folded structure. J. Biochem. 1235. Rardella, C.E., D.W Hum, J. Homoki, and
(Tokyo) 129, 259-269. W.L. Miller (1994). Point mutation of Arg440 to
1225. Hales, D.B., L.L. Sha, and A.H. Payne (1987). His in cytochrome P450cl7 causes severe 17a-
Testosterone inhibits cAMP-induced de novo hydroxylase deficiency. J. Clin. Endocrinol.
synthesis of Leydig cell cytochrome P-450(17a) Metab. 79, 160-164.
518 F. Peter Guengerich

1236. Imai, T., H. Globerman, J.M. Gertner, N. Kagawa, 1247. Katagiri, M., N. Kagawa, and M.R. Waterman
and M.R. Waterman (1993). Expression and (1995). The role of cytochrome b^ in the biosyn-
purification of functional human 17a-hydroxy- thesis of androgens by human P450cl7. Arch.
lase/17,20-lyase (P450cl7) in Escherichia coli. Biochem. Biophys. 317, 343-347.
Use of this system for study of a novel form of 1248. Miller, W.L., R.J. Auchus, and D.H. Geller
combined 17a-hydroxylase/17,20-lyase defi- (1997). The regulation of 17, 20 lyase activity.
ciency. J. Biol. Chem. 268, 19681-19689. Steroids 62, 133-142.
1237. Monno, S., Y. Mizushima, N. Toyoda, T. Kashii, 1249. Biason-Lauber, A., B. Kempken, E. Werder,
and M. Kobayashi (1997). A new variant of the M.G. Forest, S. Einaudi, M.B. Ranke et al.
cytochrome P450cl7 (CYP17) gene mutation in (2000). 17a-hydroxylase/17,20-lyase deficiency
three patients with 17a-hydroxylase deficiency. as a model to study enzymatic activity regulation:
Ann. Hum. Genet. 61, 275-279. Role of phosphorylation. J. Clin. Endocrinol.
1238. Kagimoto, K., M.R. Waterman, M. Kagimoto, Metab. H5, 1226-1231.
P. Ferreira, E.R. Simpson, and J.S. Winter (1989). 1250. Soucy, P and V Luu-The (2002). Assessment of
Identification of a common molecular basis the ability of type 2 cytochrome ^5 to modulate
for combined 17a-hydroxylase/17,20-lyase defi- 17,20-lyase activity of human P450cl7. J. Steroid
ciency in two Mennonite families. Hum. Genet. Biochem. Mol. Biol. 80, 71-75.
82, 285-286. 1251. Brock, B.J. and M.R. Waterman (1999).
1239. Yamaguchi, H., M. Nakazato, M. Miyazato, Biochemical differences between rat and human
K. Kangawa, and S. Matsukura (1997). A 5'- cytochrome P450cl7 support the different
splice site mutation in the cytochrome P450 steroidogenic needs of these two species.
steroid 17a-hydroxylase gene in 17a-hydroxy- Biochemistry 3H, 1598-1606.
lase deficiency. J. Clin. Endocrinol. Metab. 82, 1252. Yamazaki, T, T. Ohno, T Sakaki, M. Akiyoshi-
1934-1938. Shibata, Y Yabusaki, T. Imai et al. (1998). Kinetic
1240. Geller, D.H., R.J. Auchus, B.B. Mendonga, and anaylsis of successive reactions catalyzed by
W.L. Miller (1997). The genetic and functional bovine cytochrome P450j7^, . Biochemistry
basis of isolated 17,20-lyase deficiency. Nat. 37, 2800-2806.
Genet. 17,201-205. 1253. Soucy, P and V Luu-The (2000). Conversion of
1241. Miller, W.L., D.H. Geller, and R.J. Auchus pregnenolone to DHEA by human 17a-hydroxy-
(1998). The molecular basis of isolated 17, 20 lase/17,20-lyase (P450c 17). Evidence that DHEA
lyase deficiency. Endocrinol. Res. 24, 817-825. is produced from the released intermediate, 17a-
1242. Auchus, R.J. and M.K. Gupta (2002). Towards hydroxypregnenolone. Eur J. Biochem. 267,
a unifying mechanism for CYP17 mutations 3243-3247.
that cause isolated 17,20-lyase deficiency. 1254. Monno, S., H. Ogawa, T. Date, M. Fujioka, W.L.
Endocrinol. Res. 28, 443-447. Miller, and M. Kobayashi (1993). Mutation of
1243. Kagawa, N. and M.R. Waterman (1995). histidine 373 to leucine in cytochrome P450cl7
Regulation of steroidogenic and related P450s. In causes 17a-hydroxylase deficiency. J. Biol.
PR. Ortiz de Montellano (ed.). Cytochrome Chem. 268,25811-25817.
P450-Structure, Mechanism, and Biochemistry, 1255. Lam, C.W, W Arlt, C.K. Chan, J.W. Honour,
2nd edn. Plenum Press, New York, pp. 419-442. C.J. Lin, S.F Tong et al. (2001). Mutation of
1244. P Lee-Robichaud, A.Z. Shyadehi, J.N. Wright, proline 409 to arginine in the meander region of
M.E. Akhtar, and M. Akhtar (1995). Mechanistic cytochrome p450cl7 causes severe 17a-hydroxy-
kinship between hydroxylation and desaturation lase deficiency Mol. Genet. Metab. 72, 254-259.
reactions: Acyl-carbon bond cleavage pro- 1256. Lee-Robichaud, P, M.E. Akhtar, and M. Akhtar
moted by pig and human CYP17 {?-450^^^, 17a- (1998). An analysis of the role of active site pro-
hydroxylase-17,20-lyase). Biochemistry 34, tic residues of cytochrome P-450s: Mechanistic
14104-14113. and mutational studies on 17a-hydroxylase-
1245. Lieberman, S. and PA. Warne (2001). 17- 17,20-lyase (P-45017a also CYP17). Biochem.
Hydroxylase: An evaluation of the present view J. 330, 967-974.
of its catalytic role in steroidogenesis. J. Steroid 1257. Kitamura, M., E. Buczko, and M.L. Dufau
Biochem. Mol. Biol. 78, 299-312. (1991). Dissociation of hydroxylase and lyase
1246. Soucy, P, L. Lacoste, and V Luu-The (2003). activities by site-directed mutagenesis of the rat
Assessment of porcine and human 16-ene- P450i7^. ^o^- Endocrinol. 5, 1373-1380.
synthase, a third activity of P450cl7, in the 1258. Biason-Lauber, A., E. Leiberman, and
formation of an androstenol precursor. Eur J. M. Zachmann (1997). A single amino acid sub-
Biochem. 210, 1349-1355. stitution in the putative redox partner-binding site
Human P450s 519

of P450cl7 as cause of isolated 17,20-lyase (17a-hydroxylase/17,20-lyase): Insights into


deficiency. J. Clin. Endocrinol Metab. 82, reaction mechanisms and effects of mutations.
3807-3812. Mol. Endocrinol. 13, 1169-1182.
1259. Lee-Robichaud, P., M.E. Akhtar, and M. Akhtar 1270. Ahmed, S. (1999). A novel molecular modelling
(1998). Control of androgen biosynthesis in the study of inhibitors of the 17a-hydroxylase com-
human through the interaction of Arg^^^ and ponent of the enzyme system 17a-hydroxy-
^j.g358 of CYP17 with cytochrome b^ Biochem. J. lase/17,20-lyase (P-450j7^). Bioorg. Med. Chem.
332, 293-296. 7, 1487-1496.
1260. LeeRobichaud, R, M.E. Akhtar, and M. Akhtar 1271. Schappach, A. and H.D. Holtje (2001). Molecular
(1999). Lysine mutagenesis identifies cationic modelling of 17a-hydroxylase-17,20-lyase.
charges of human CYP17 that interact with Pharmazie 56, 435^42.
cytochrome b^ to promote male sex-hormone 1272. Kan, PB., M.A. Hirst, and D. Feldman (1985).
biosynthesis. Biochem. J. 342, 309-312. Inhibition of steroidogenic cytochrome P-450
1261. Auchus, R.J., K. Worthy, D.H. Geller, and enzymes in rat testis by ketoconazole and related
W.L. Miller (2000). Probing structural and func- imidazole anti-fiingal drugs. J. Steroid Biochem.
tional domains of human P450cl7. Endocrinol. 23, 1023-1029.
Res. 26, 695-703. 1273. Kossor, D.C., S. Kominami, S. Takemori, and
1262. Gupta, M.K., D.H. Geller, and R.J. Auchus H.D. Colby (1992). Destruction of testicular
(2001). Pitfalls in characterizing P450cl7 muta- cytochrome P-450 by 7a-thiospironolactone is
tions associated with isolated 17,20-lyase catalyzed by the 17od-hydroxylase. J. Steroid
deficiency. J. Clin. Endocrinol. Metab. 86, Biochem. Mol. Biol. 42, 421^24.
4416-4423. 1274. Potter, G.A., S.E. Barrie, M. Jarman, and
1263. Di Cerbo, A., A. Biason-Lauber, M. Savino, M.G. Rowlands (1995). Novel steroidal inhi-
M.R. Piemontese, A. Di Giorgio, M. Perona et al. bitors of human cytochrome P45017a (17a-
(2002). Combined 17a-hydroxylase/17,20-lyase hydroxylase-Cj7 20"^^^^^)- Potential agents for the
deficiency caused by Phe93Cys mutation in the treatment of prostatic cancer. J. Med. Chem. 38,
CYP17 gene. J. Clin. Endocrinol. Metab. 87, 2463-2471.
898-905. 1275. Li, J.S., Y Li, C. Son, and A.M. Brodie (1996).
1264. Katsumata, N., M. Satoh, A. Mikami, S. Mikami, Synthesis and evaluation of pregnane derivatives
A. Nagashima-Miyokawa, N. Sato et al. (2001). as inhibitors of human testicular 17a-hydroxy-
New compound heterozygous mutation in the lase/Cj7 2o-lyase. J. Med. Chem. 39, 4335-4339.
CYP17 gene in a 46,XY girl with 17a-hydroxy- 1276. Njar, VC. and A.M. Brodie (1999). Inhibitors of
lase/17,20-lyase deficiency. Horm. Res. 55, 17a-hydroxylase/l 7,20-lyase (CYP17): Potential
141-146. agents for the treatment of prostate cancer. Curr.
1265. Brock, B.J. and M.R. Waterman (2000). The use Pharm.Des. 5, 163-180.
of random chimeragenesis to study structure/ 1277. Hartmann, R.W, M. Hector, B.G. Wachall,
function properties of rat and human P450cl7. A. Palusczak, M. Palzer, V Huch et al. (2000).
Arch. Biochem. Biophys. 373, 401^08. Synthesis and evaluation of 17-aliphatic hetero-
1266. Mathieu, A.P., R.J. Auchus, and J.G. LeHoux cycle-substituted steroidal inhibitors of 17a-
(2002). Comparison of the hamster and human hydroxylase/C17-20-lyase (P450 17). J. Med.
adrenal P450cl7 (17a-hydroxylase/17,20-lyase) Chem. 43, 4437-4445.
using site-directed mutagenesis and molecular 1278. Burkhart, J.P, PM. Weintraub, C.A. Gates,
modeling. J. Steroid Biochem. Mol. Biol. 80, R.J. Resvick, R.J. Vaz, D. Friedrich et al (2002).
99-107. Novel steroidal vinyl fluorides as inhibitors of
1267. Lin, D., L.H. Zhang, E. Chiao, and W.L. Miller steroid CI7(20) lyase. Bioorg. Med. Chem. 10,
(1994). Modeling and mutagenesis of the active 929-934.
site of human P450cl7. Mol. Endocrinol. 8, 1279. Auchus, R.J., A. Sampath Kumar, C. Andrew
392^02. Boswell, M.K. Gupta, K. Bruce, N.P Rath et al
1268. Burke, D.F., C.A. Laughton, and S. Neidle (2003). The enantiomer of progesterone {ent-
(1997). Homology modelling of the enzyme P450 progesterone) is a competitive inhibitor of human
17a-hydroxylase/17,20-lyase—a target for pro- cytochromes P450cl7 and P450c21. Arch.
state cancer chemotherapy—from the crystal Biochem. Biophys. 409, 134-144.
structure of P450gj^ 3. Anticancer Drug Des. 12, 1280. Owen, C.P, RJ. NichoUs, H.J. Smith, and
113-123. R. Whomsley (1999). Inhibition of aromatase
1269. Auchus, R.J. and W.L. Miller (1999). (P450^pjj^) by some l-(benzofuran-2-ylmethyl)
Molecular modeling of human P450cl7 imidazoles. J. Pharm. Pharmacol. 51, 427-433.
520 F. Peter Guengerich

1281. Recanatini, M., A. Bisi, A. Cavalli, F. Belluti, A polymorphism in the CYP 17 gene increases the
S. Gobbi, A. Rampa et al. (2001). A new class of risk of breast cancer. Cancer Res. 57, 1063-1065.
nonsteroidal aromatase inhibitors: Design and 1293. Feigelson, H.S., R. McKean-Cowdin,
synthesis of chromone and xanthone derivatives G.A. Coetzee, D.O. Stram, L.N. Kolonel, and
and inhibition of the P450 enzymes aromatase B.E. Henderson (2001). Building a multigenic
and 17a-hydroxylase/C17,20-lyase. J. Med. model of breast cancer susceptibility: CYP 17 and
Chem. 44, 672-680. HSD17B1 are two important candidates. Cancer
1282. Cavalli, A. and M. Recanatini (2002). Looking Res. 61, 785-789.
for selectivity among cytochrome P450 1294. Thompson, PA. and C. Ambrosone (2000).
inhibitors. J. Med. Chem. 45, 251-254. Molecular epidemiology of genetic polymor-
1283. Ehmer, RB., J. Jose, and R.W. Hartmann (2000). phisms in estrogen metabolizing enzymes in
Development of a simple and rapid assay for the human breast cancer. J. Natl. Cancer Inst.
evaluation of inhibitors of human 17cx-hydroxy- Monogr, 125-134.
lase-Cj7 2o"ly^se (P450cl7) by coexpression of 1295. Mitrunen, K., N. Jourenkova, V Kataja,
P450cl7' with NADPH-cytochrome-P450-reduc- M. Eskelinen, V.M. Kosma, S. Benhamou et al.
tase in Escherichia coli. J. Steroid Biochem. Mol. (2000). Steroid metabolism gene CYP17
Biol. 75, 57-63. polymorphism and the development of breast
1284. Grigoryev, D.N., K. Kato, VC. Njar, B.J. Long, cancer. Cancer Epidemiol. Biomarkers Prev. 9,
Y.Z. Ling, X. Wang et al. (1999). Cytochrome 1343-1348.
P450cl7-expressing Escherichia coli as a first- 1296. Ye, Z. and J.M. Parry (2002). The CYP17 M^pAl
step screening system for 17Q:-hydroxylase- polymorphism and breast cancer risk: A meta-
Cj7 2o"^y^^^ inhibitors. Anal. Biochem. 267, analysis. Mutagenesis 17, 119-126.
319-330. 1297. Feigelson, H.S., R. McKean-Cowdin, and
1285. Auchus, R.J. (2001). The genetics, pathophysiol- B.E. Henderson (2002). Concerning the CYP 17
ogy, and management of human deficiencies of MspAl polymorphism and breast cancer risk:
P450cl7. Endocrinol. Metab. Clin. North Am. 30, A meta-analysis. Mutagenesis 17, 445-446.
101-119, vii. 1298. Ambrosone, C.B., K.B. Moysich, H. Furberg,
1286. Kaufman, F.R., G. Costin, U. Goebelsmann, J.L. Freudenheim, E.D. Bowman, S. Ahmed et al.
F.Z. Stanczyk, and M. Zachmann (1983). Male (2003). CYP 17 genetic polymorphism, breast
pseudohermaphroditism due to 17,20-desmolase cancer, and breast cancer risk factors. Breast
deficiency. J. Clin. Endocrinol. Metab. 57, 32-36. Cancer Res. 5, R45-51.
1287. Huang, J., T. Ushiyama, K. Inoue, K. Mori, and 1299. Stanford, J.L., E.A. Noonan, L. Iwasaki, S. Kolb,
S. Hukuda (1999). Possible association of CYP17 R.B. Chadwick, Z. Feng et al. (2002). A poly-
gene polymorphisms with the onset of rheuma- morphism in the CYPl 7 gene and risk of prostate
toid arthritis. Clin. Exp. Rheumatol. 17, 721-724. cancer. Cancer Epidemiol. Biomarkers Prev. 11,
1288. Lai, J., D. Vesprini, W. Chu, H. Jernstrom, and 243-247.
S.A. Narod (2001). CYP gene polymorphisms 1300. Haiman, C.A., M.J. Stampfer, E. Giovannucci,
and early menarche. Mol. Genet. Metab. 74, J. Ma, N.E. Decalo, PW. Kantoff etal. (2001). The
449-457. relationship between a polymorphism in CYP 17
1289. Feigelson, H.S., R. McKean-Cowdin, M.C. Pike, with plasma hormone levels and prostate cancer.
G.A. Coetzee, L.N. Kolonel, A.M. Nomura Cancer Epidemiol. Biomarkers Prev. 10, 743-748.
et al. (1999). Cytochrome P450cl7alpha gene 1301. McKean-Cowdin, R., H.S. Feigelson, M.C. Pike,
(CYP 17) polymorphism predicts use of hormone G.A. Coetzee, L.N. Kolonel, and B.E. Henderson
replacement therapy. Cancer Res 59, 3908-3910. (2001). Risk of endometrial cancer and estrogen
1290. Marszalek, B., M. Lacinski, N. Babych, E. Capla, replacement therapy history by CYP 17 genotype.
J. Biemacka-Lukanty, A. Warenik-Szymankiewicz Cancer Res. 61, 848-849.
et al. (2001). Investigations on the genetic poly- 1302. de Carmo Silva, R., C.E. Kater, S.A. Dib,
morphism in the region of CYP 17 gene encoding S. Laureti, F. Forini, A. Cosentino et al. (2000).
5'-UTR in patients with polycystic ovarian syn- Autoantibodies against recombinant human
drome. Gynecol. Endocrinol. 15, 123-128. steroidogenic enzymes 21-hydroxylase, side-
1291. Kristensen, VN., E.H. Kure, B. Erikstein, chain cleavage and 17a-hydroxylase in Addison's
N. Harada, and A. Borresen-Dale (2001). Genetic disease and autoimmune polyendocrine syn-
susceptibility and environmental estrogen-like drome type 111. Eur J. Endocrinol. 142, 187-194.
compounds. Mutat. Res. 482, 77-82. 1303. Simpson, E.R., C. Clyne, G. Rubin, W.C. Boon,
1292. Feigelson, H.S., G.A. Coetzee, L.N. Kolonel, K. Robertson, K. Britt et al. (2002). Aromatase—
R.K. Ross, and B.E. Henderson (1997). a brief overview. Annu. Rev. Physiol. 64, 93-127.
Human P450s 521

1304. Roselli, C.E. and J.A. Resko (2001). Cytochrome 1315. Takayanagi, R., K. Goto, S. Suzuki, S. Tanaka,
P450 aromatase (CYP19) in the non-human pri- S. Shimoda, and H. Nawata (2002). Dehy-
mate brain: Distribution, regulation, and func- droepiandrosterone (DHEA) as a possible source
tional significance. J. Steroid Biochem. Mol. Biol. for estrogen formation in bone cells: Correlation
79, 247-253. between bone mineral density and serum
1305. Harada, N. (1992). A unique aromatase DHEA-sulfate concentration in postmenopausal
(P-450^j^Qj^) mRNA formed by alternative use women, and the presence of aromatase to
of tissue-specific exons 1 in human skin fibrob- be enhanced by 1,25-dihydroxyvitamin D3 in
lasts. Biochem. Biophys. Res. Commun. 189, human osteoblasts. Mech. Ageing Dev. 123,
1001-1007. 1107-1114.
1306. Hinshelwood, M.M. and C.R. Mendelson (2001). 1316. Zhang, R, D. Zhou, Y.C Kao, J. Ye, and S. Chen
Tissue-specific expression of the human CYP19 (2002). Expression and purification of a recombi-
(aromatase) gene in ovary and adipose tissue of nant form of human aromatase from Escherichia
transgenic mice. J. Steroid Biochem. Mol. Biol. coli. Biochem. Pharmacol. 64, 1317-1324.
79,193-201. 1317. Kagawa, N., Q. Cao, and K. Kusano (2003).
1307. Shozu, M., H. Sumitani, T. Segawa, H.J. Yang, Expression of human aromatase (CYP19) in
K. Murakami, T. Kasai et al. (2002). Over- Escherichia coli by N-terminal replacement and
expression of aromatase P450 in leiomyoma induction of cold stress response. Steroids 68,
tissue is driven primarily through promoter 1.4 205-209.
of the aromatase P450 gene (CYP19). J. Clin. 1318. Graham-Lorence, S., M.W. Khalil, M.C Lorence,
Endocrinol. Metab. 87, 2540-2548. C.R. Mendelson, and E.R. Simpson (1991).
1308. Rubin, G.L., J.H. Duong, CD. Clyne, C.J. Speed, Structure-function relationships of human aro-
Y. Murata, C. Gong et al. (2002). Ligands for the matase cytochrome P-450 using molecular mod-
peroxisomal proliferator-activated receptor eling and site-directed mutagenesis. J. Biol.
gamma and the retinoid X receptor inhibit aro- Chem. 266, 11939-11946.
matase cytochrome P450 (CYPJ9) expression 1319. Conley, A., S. Mapes, CJ. Corbin, D. Greger, and
mediated by promoter II in human breast adipose. S. Graham (2002). Structural determinants of
Endocrinology 143, 2863-2871. aromatase cytochrome P450 inhibition in sub-
1309. Clyne, CD., C.J. Speed, J. Zhou, and E.R. strate recognition site-1. Mol. Endocrinol. 16,
Simpson (2002). Liver receptor homologue-1 1456-1468.
(LRH-1) regulates expression of aromatase in 1320. Marcotte, PA. and CH. Robinson (1982). Design
preadipocytes. J. Biol. Chem. 277, 20591-20597. of mechanism-based inactivators of human pla-
1310. Kamat, A. and CR. Mendelson (2001). cental aromatase. Cancer Res. 42, 3322s-3326s.
Identification of the regulatory regions of the 1321. Brueggemeier, R.W. (2002). Aromatase inhibitors
human aromatase P450 {CYP19) gene involved in in breast cancer therapy. Expert Rev. Anticancer
placenta-specific expression. J. Steroid Biochem. Then 2, 181-191.
Mol. Biol. 79, 173-180. 1322. Lombardi, P. (2002). Exemestane, a new steroidal
1311. Haiman, CA., S.E. Hankinson, I. De Vivo, aromatase inhibitor of clinical relevance.
C Guillemette, N. Ishibe, D.J. Hunter et al. Biochim. Biophys. Acta 1587, 326-337.
(2003). Polymorphisms in steroid hormone path- 1323. Lonning, PE. (2002). The role of aromatase
way genes and mammographic density. Breast inactivators in the treatment of breast cancer. Int.
Cancer Res. Treat. 11, 27-36. J. Clin. Oncol 7, 265-270.
1312. Cole, PA. and CH. Robinson (1988). A peroxide 1324. Lonning, PE. (2002). Aromatase inhibitors and
model reaction for placental aromatase. J. Am. inactivators for breast cancer treatment. Eur
Chem. Soc. 110, 1284-1285. J. Cancer 38(Suppl 6), S47-S48.
1313. Akhtar, M., D. Corina, S. Miller, A.Z. Shyadehi, 1325. Jones, S.A. and S.E. Jones (2000). Exemestane:
and J.N. Wright (1994). Mechanism of the A novel aromatase inactivator for breast cancer.
acyl-carbon cleavage and related reactions Clin. Breast Cancer 1, 211-216.
catalyzed by multifunctional P-450s: Studies 1326. Winnett, G., D. van Hagen, and M. Schrey
on cytochrome P45017a. Biochemistry 33, (2003). Prostaglandin ]^ metabolites inhibit aro-
4410-4418. matase activity by redox-sensitive mechanisms:
1314. Roberts, E.S., A.D.N. Vaz, and M.J. Coon (1991). Potential implications for breast cancer therapy.
Catalysis by cytochrome P-450 of an oxidative Int. J. Cancer 103, 600-605.
reaction in xenobiotic aldehyde metabolism: 1327. Pouget, C , C Fagnere, J.P Basly, G. Habrioux,
Deformylation with olefin formation. Proc. Natl. and A.J. Chulia (2002). Design, synthesis and
Acad Set USA 88, 8963-8966. evaluation of 4-imidazolylflavans as new leads
522 F. Peter Guengerich

for aromatase inhibition. Bioorg. Med. Chem. 1339. Yu, L., D.G. Romero, C.E. Gomez-Sanchez, and
Lett. 12,2859-2861. E.R Gomez-Sanchez (2002). Steroidogenic
1328. Smith, M.R., D. Kaufman, D. George, W.K. Oh, enzyme gene expression in the human brain.
M. Kazanis, J. Manola et al. (2002). Selective Mol. Cell Endocrinol. 190, 9-17.
aromatase inhibition for patients with androgen- 1340. Zanger, U.M., N. Kagawa, J. Lund, and
independent prostate carcinoma. Cancer 95, M.R. Waterman (1992). Distinct biochemical
1864-1868. mechanisms for cAMP-dependent transcription
1329. Murata, Y., K.M. Robertson, M.E. Jones, and of CYP17 and CYP21. FASEB J. 6, 719-723.
E.R. Simpson (2002). Effect of estrogen defi- 1341. Watanabe, N., M. Kitazume, J. Fujisawa,
ciency in the male: The ArKO mouse model. Mol. M. Yoshida, and Y. Fujii-Kuriyama (1993). A
Cell Endocrinol. 193, 7-12. novel cAMP-dependent regulatory region includ-
1330. Herrmann, B.L., B. Sailer, O.E. Janssen, ing a sequence like the cAMP-responsive ele-
P. Gocke, A. Bockisch, H. Sperling et al. (2002). ment, far upstream of the human CYP21A2 gene.
Impact of estrogen replacement therapy in a male Eur J. Biochem. 214, 521-531.
with congenital aromatase deficiency caused by a 1342. Chang, S.E and B.C. Chung (1995). Difference in
novel mutation in the CYP19 gene. J. Clin. transcriptional activity of two homologous
Endocrinol. Metab. 87, 5476-5484. CYP21A genes. Mol. Endocrinol. 9, 1330-1336.
1331. Meinhardt, U. and RE. Mulhs (2002). The aro- 1343. Bird, I.M., J.I. Mason, and WE. Rainey (1998).
matase cytochrome P-450 and its clinical impact. Protein kinase A, protein kinase C, and Ca-^^-
Horm.Res. 57, 145-152. regulated expression of 21-hydroxylase cyto-
1332. Britt, K.L., A.E. Drummond, M. Dyson, chrome P450 in H295R human adrenocortical
N.G. Wreford, M.E. Jones, E.R. Simpson et al. cells. J. Clin. Endocrinol. Metab. 83, 1592-1597.
(2001). The ovarian phenotype of the aromatase 1344. Wijesuriya, S.D., G. Zhang, A. Dardis, and
knockout (ArKO) mouse. J. Steroid Biochem. WL. Miller (1999). Transcriptional regulatory
Mol. Biol. 79, 181-185. elements of the human gene for cytochrome
1333. Bakker, J., S. Honda, N. Harada, and J. Balthazart P450c21 (steroid 21-hydroxylase) lie within
(2002). The aromatase knock-out mouse provides intron 35 of the linked C4B gene. J. Biol. Chem.
new evidence that estradiol is required during 274,38097-38106.
development in the female for the expression of 1345. White, P C , M.T. Tusie-Luna, M.I. New, and
sociosexual behaviors in adulthood. J. Neurosci. PW. Speiser (1994). Mutations in steroid 21-
22,9104-9112. hydroxylase (CYP21). Hum. Mutat. 3, 373-378.
1334. Lonning, RE., L.E. Kragh, B. Erikstein, 1346. M.T. Tusie-Luna, PW. Speiser, M. Dumic,
A. Hagen, T Risberg, E. Schlichting et al. M.l. New, and PC. White (1991). A mutation
(2001). The potential for aromatase inhibition in (Pro-30 to Leu) in CYP21 represents a potential
breast cancer prevention. Clin. Cancer Res. 7, nonclassic steroid 21-hydroxylase deficiency
4423s-4428s; discussion 441 ls^412s. allele. Mol. Endocrinol. 5, 685-692.
1335. Suspitsin, E.N., M.Y. Grigoriev, A.V Togo, 1347. Amor, M., K.L. Parker, H. Globerman, M.I. New,
E.S. Kuligina, E.V. Belogubova, K.M. Pozharisski and PC. White (1988). Mutation in the CYP21B
et al. (2002). Distinct prevalence of the CYP19 gene (He-172—->Asn) causes steroid 21-
A3(TTTA)7 allele in premenopausal versus post- hydroxylase deficiency. Proc. Natl. Acad. Sci.
menopausal breast cancer patients, but not in con- USAH5, 1600-1604.
trol individuals. Eur J. Cancer 38, 1911-1916. 1348. Owerbach, D., L. Sherman, A.L. Ballard, and
1336. Kado, N., J. Kitawaki, H. Obayashi, H. Ishihara, R. Azziz (1992). Pro-453 to Ser mutation in CYP21
H. Koshiba, I. Kusuki et al. (2002). Association is associated with nonclassic steroid 21-hydroxy-
of the CYP17 gene and CYP19 gene polymor- lase deficiency. Mol. Endocrinol. 6, 1211-1215.
phisms with risk of endometriosis in Japanese 1349. Bobba, A., E. Marra, P Lattanzio, A. lolascon, and
women. Hum. Reprod. 17, 897-902. S. Giannattasio (2000). Characterization of the
1337. Bryan, G.T., A.M. Lewis, J.B. Harkins, CYP21 gene 5' flanking region in patients affected
S.F. Micheletti, and G.S. Boyd (1974). by 21-OH deficiency Hum. Mutat. 15, 481.
Cytochrome P450 and steroid 21-hydroxylation 1350. White, PC. and PW. Speiser (2000). Congenital
in microsomes from beef adrenal cortex. Steroids adrenal hyperplasia due to 21-hydroxylase defi-
23, 185-201. ciency. Endocrinol. Rev. 21, 245-291.
1338. Zhou, Z., VR. Agarwal, N. Dixit, P White, and 1351. Lee, H. (2001). CYP21 mutations and congenital
PW. Speiser (1997). Steroid 21-hydroxylase adrenal hyperplasia. Clin. Genet. 59, 293-301.
expression and activity in human lymphocytes. 1352. Krone, N., A. Braun, A.A. Roscher, D. Knorr, and
Mol. Cell Endocrinol. Ill, 11-18. H.P Schwarz (2000). Predicting phenotype in
Human P450s 523

steroid 21-hydroxylase deficiency? Comprehensive a vitamin D3 hydroxylase-associated protein.


genotyping in 155 unrelated, well defined patients J. Biol. Chem. 269, 176-182.
from southern Germany. J. Clin. Endocrinol. 1364. Chen, K.S., J.M. Prahl, and H.F. DeLuca (1993).
Metab. 85, 1059-1065. Isolation and expression of human 1,25-dihy-
1353. Levo, A. and J. Partanen (2001). Novel mutations droxyvitamin D^ 24-hydroxylase cDNA. Proc.
in the human CYP21 gene. Prenat. Diagn. 21, Natl. Acad Sci. USA 90, 4543-4547.
885-889. 1365. Yang, W., PA. Friedman, R. Kumar, J.L. Omdahl,
1354. Koyama, S., T. Toyoura, S. Saisho, B.K. May, M.L. SiuCaldera et al. (1999).
K. Shimozawa, and J. Yata (2002). Genetic analy- Expression of 25(OH)D3 24-hydroxylase in distal
sis of Japanese patients with 21-hydroxylase nephron: Coordinate regulation by 1, 25(OH)2D3
deficiency: Identification of a patient with a and cAMP or PTH. Am. J. Physiol. 276,
new mutation of a homozygous deletion of E793-E805.
adenine at codon 246 and patients without 1366. Jones, G., H. Ramshaw, A. Zhang, R. Cook,
demonstrable mutations within the structural V Byford, J. White et al. (1999). Expression and
gene for CYP21. J. Clin. Endocrinol. Metab. 87, activity of vitamin D-metabolizing cytochrome
2668-2673. P450s (CYPla and CYP24) in human nonsmall
1355. Lee, H.H., D.M. Niu, R.W. Lin, P Chan, and cell lung carcinomas. Endocrinology 140,
C.Y. Lin (2002). Structural analysis of the chimeric 3303-3310.
CYP21P/CYP21 gene in steroid 21-hydroxylase 1367. Chen, M.L., G. Heinrich, Y.I. Ohyama, K. Okuda,
deficiency. J. Hum. Genet. 47, 517-522. J.L. Omdahl, T.C. Chen et al. (1994). Expression
1356. Koppens, PR, T. Hoogenboezem, and of 25-hydroxyvitamin D3-24-hydroxylase mRNA
H.J. Degenhart (2002). Duplication of the in cultured human keratinocytes. Proc. Soc. Exp.
CYP21A2 gene complicates mutation analysis of Biol. Med. 207,57-61.
steroid 21-hydroxylase deficiency: Characteristics 1368. Schuster, I., H. Egger, N. Astecker, G. Herzig,
of three unusual haplotypes. Hum. Genet. I l l , M. Schussler, and G. Vorisek (2001). Selective
405-410. inhibitors of CYP24: Mechanistic tools to
1357. Dain, L.B., N.D. Buzzalino, A. Oneto, S. Belh, explore vitamin D metabolism in human ker-
M. Stivel, T. Pasqualini et al. (2002). Classical atinocytes. Steroids 66, 451-462.
and nonclassical 21-hydroxylase deficiency: 1369. Bareis, P, E. Kallay, M.G. Bischof, G. Bises,
A molecular study of Argentine patients. Clin. H. Hofer, C. Potzi et al. (2002). Clonal differ-
Endocrinol. (Oxford) 56, 239-245. ences in expression of 25-hydroxyvitamin D3-
1358. Mornet, E. and J.F. Gibrat (2000). A 3D model of 1 a-hydroxylase, of 25-hydroxyvitamin D3-24-
human P450c21: Study of the putative effects of hydroxylase, and of the vitamin D receptor in
steroid 21-hydroxylase gene mutations. Hum. human colon carcinoma cells: Effects of epider-
Genet. 106, 330-339. mal growth factor and la,25-dihydroxyvitamin
1359. Menard, R.H., EC. Bartter, and J.R. Gillette D3. Exp. Cell Res. 276, 320-327.
(1976). Spironolactone and cytochrome P-450: 1370. Farhan, H. and H.S. Cross (2002). Transcriptional
Impairment of steroid 21-hydroxylation in the inhibition of CYP24 by genistein. Ann. N. Y.
adrenal cortex. Arch. Biochem. Biophys. 173, Acad Sci. 973, 459-462.
395-402. 1371. Zierold, C , H.M. Darwish, and H.F DeLuca
1360. Tajima, T., T. Okada, X.M. Ma, W. Ramsey, (1994). Identification of a vitamin D-response
S. Bomstein, and G. Aguilera (1999). Restoration element in the rat calcidiol (25-hydroxyvitamin
of adrenal steroidogenesis by adenovirus-mediated D3) 24-hydroxylase gene. Proc. Natl. Acad. Sci.
transfer of human cytochrome P450 21-hydroxy- USA 91, 900-902.
lase into the adrenal gland of 21-hydroxylase-defi- 1372. Ohyama, Y, K. Ozono, M. Uchida, T. Shinki,
cientmice. GeneTher 6, 1898-1903. S. Kato, T. Suda et al. (1994). Identification
1361. Pedersen, J.I., H.H. Shobaki, I. Holmberg, of a vitamin D-responsive element in the 5'-
S. Bergseth, and I. Bjorkhem (1983). 25- flanking region of the rat 25-hydroxyvitamin
Hydroxyvitamin D3-24-hydroxylase in rat kidney D3 24-hydroxylase gene. J. Biol. Chem. 269,
mitochondria. J. Biol. Chem. 258, 742-746. 10545-10550.
1362. Ohyama, Y, S. Hayashi, and K. Okuda (1989). 1373. Kerry, D.M., PP Dwivedi, C.N. Hahn,
Purification of 25-hydroxyvitamin D3 24-hydrox- H.A. Morris, J.L. Omdahl, and B.K. May (1996).
ylase from rat kidney mitochondria. FEBS Lett. Transcriptional synergism between vitamin D-
255, 405^08. responsive elements in the rat 25-hydroxyvitamin
1363. Ettinger, R.A., R. Ismail, and H.F. DeLuca D3 24-hydroxylase (CYP24) promoter. J. Biol.
(1994). cDNA cloning and characterization of Chem. 271,29715-29721.
524 F. Peter Guengerich

1374. Dwivedi, P.P., J.L. Omdahl, I. Kola, D.K. Hume, 1385. Dilworth, KJ., I. Scott, A. Green, S. Strugnell,
and B.K. May (2000). Regulation of rat cyto- YD. Guo, E.A. Roberts et al (1995). Different
chrome P450C24 (CYP24) gene expression. Evi- mechanisms of hydroxylation site selection by
dence for functional cooperation of Ras-activated liver and kidney cytochrome P450 species
Ets transcription factors with the vitamin D (CYP27 and CYP24) involved in vitamin D
receptor in 1,25- dihydroxyvitamin D3-mediated metaboUsm. ^ 5zo/. Chem. 270, 16766-16774.
induction. J. Biol. Chem. 275, 47-55. 1386. Schuster, I., H. Egger, P. Nussbaumer, and
1375. Raval-Pandya, M., P. Dhawan, F. Barletta, and R.T. Kroemer (2003). Inhibitors of vitamin D
S. Christakos (2001). YYl represses vitamin D hydroxylases: Structure-activity relationships.
receptor-mediated 25-hydroxyvitamin D3 24- J. Cell. Biochem. 88, 372-380.
hydroxylase transcription: Relief of repression by 1387. Schuster, I., H. Egger, D. Bikle, G. Herzig,
CREB-binding protein. Mol Endocrinol. 15, G.S. Reddy, A. Stuetz et al. (2001). Selective
1035-1046. inhibition of vitamin D hydroxylases in human
1376. Dwivedi, PR, C.S. Hii, A. Ferrante, J. Tan, keratinocytes. Steroids 66, 409-422.
C.J. Der, J.L. Omdahl et al (2002). Role of MAP 1388. Henry, H.L. (2001). The 25(OH)D3/la,25(OH)2D3-
kinases in the 1,25-dihydroxyvitamin D^-induced 24R-hydroxylase: A catabolic or biosynthetic
transactivation of the rat cytochrome P450C24 enzyme? Steroids 66, 391-398.
(CYP24) promoter. Specific functions for 1389. Kasuga, H., N. Hosogane, K. Matsuoka,
ERK1/ERK2 and ERK5. J. Biol. Chem. Ill, I. Mori, Y Sakura, K. Shimakawa et al. (2002).
29643-29653. Characterization of transgenic rats constitutively
1377. Jones, G. and H.S. Tenenhouse (2002). expressing vitamin D-24-hydroxylase gene.
l,25(OH)2D, the preferred substrate for CYP24. Biochem. Biophys. Res. Commun. 297,1332-1338.
J. Bone Miner. Res. 17, 179-181. 1390. Martini, R. and M. Murray (1993). Participation
1378. Sakaki, T., N. Sawada, K. Komai, S. Shiozawa, of P450 3A enzymes in rat hepatic microsomal
S. Yamada, K. Yamamoto et al. (2000). Dual retinoic acid 4-hydroxylation. Arch. Biochem.
metabolic pathway of 25-hydroxyvitamin D3 cat- Biophys. 303, 57-66.
alyzed by human CYP24. Eur J. Biochem. 267, 1391. White, J.A., B. Beckett-Jones, YD. Guo,
6158-6165. F.J. Dilworth, J. Bonasoro, G. Jones et al.
1379. Beckman, M.J., P Tadikonda, E. Werner, J. Prahl, (1997). cDNA cloning of human retinoic acid-
S. Yamada, and H.F DeLuca (1996). Human 25- metabolizing enzyme (hP450RAI) identifies
hydroxyvitamin D3-24-hydroxylase, a muhicat- a novel family of cytochromes P450 (CYP26).
alytic enzyme. Biochemistry 35, 8465-8472. J. Biol. Chem. 272, 18539-18541.
1380. Miyamoto, Y, T. Shinki, K. Yamamoto, 1392. White, J.A., H. Ramshaw, M. Taimi, W. Stangle,
Y Ohyama, H. Iwasaki, R. Hosotani et al. (1997). A. Zhang, S. Everingham et al. (2000).
1 a,25-dihydroxyvitamin D3-24-hydroxylase Identification of the human cytochrome P450,
(CYP24) hydroxylates the carbon at the end of P450RA1-2, which is predominantly expressed in
the side chain (C-26) of the C-24-fluorinated ana- the adult cerebellum and is responsible for all-
log of la,25-dihydroxyvitamin D3. J. Biol. Chem. trans-XQimo\c acid metabolism. Proc. Natl. Acad.
212, 14115-14119. Sci. USA 97, 6403-6408.
1381. Hayashi, K., M. Akiyoshi-Shibata, T. Sakaki, and 1393. Trofimova-Griffin, M.E. and M.R. Juchau (1998).
Y Tabusaki (1998). Rat CYP24 catalyses 235"- Expression of cytochrome P450RAI (CYP26) in
hydroxylation of 26,26,26,27,27,27-hexafluoro- human fetal hepatic and cephalic tissues.
calcitriol in vitro. Xenobiotica 28, 457^63. Biochem. Biophys. Res. Commun. 252,487-491.
1382. Sakaki, T, N. Sawada, Y Nonaka, Y Ohyama, 1394. Nelson, D.R. (1999). A second CYP26 P450 in
and K. Inouye (1999). Metabolic studies using humans and zebrafish: CYP26B1. Arch. Biochem.
recombinant Escherichia coli cells producing rat Biophys. 371, 345-347.
mitochondrial CYP24. CYP24 can convert 1395. Abu-Abed, S., G. MacLean, V Fraulob,
la,25-dihydroxyvitamin D3 to calcitroic acid. P Chambon, M. Petkovich, and P Dolle (2002).
Eur J. Biochem. 262, 4 3 ^ 8 . Differential expression of the retinoic acid-
1383. Inouye, K. and T. Sakaki (2001). Enzymatic metabolizing enzymes CYP26A1 and CYP26B1
studies on the key enzymes of vitamin D metabo- during murine organogenesis. Mech. Dev. 110,
lism; la-hydroxylase (CYP27B1) and 24-hydrox- 173-177.
ylase (CYP24). Biotechnol. Annu. Rev. 7, 179-194. 1396. Trofimova-Griffin, M.E. and M.R. Juchau
1384. Omdahl, J.L., E.V Bobrovnikova, A. Annalora, (2002). Developmental expression of cytochrome
P Chen, and R. Serda (2003). Expression, CYP26B1 (P450RAI-2) in human cephalic
structure-function, and molecular modeling of tissues. Brain. Res. Develop. Brain Res. 136,
vitamin D P450s. J. Cell. Biochem. 88, 356-362. 175-178.
Human P450s 525

1397. Matsuzaki, Y., B. Bouscarel, T. Ikegami, 1409. Su, P, H. Rennert, R.M. Shayiq, R. Yamamoto,
A. Honda, M. Doy, S. Ceryak et al (2002). Y Zheng, S. Addya et al. (1990). A cDNA encod-
Selective inhibition of CYP27A1 and of chen- ing a rat mitochondrial cytochrome P450 catalyz-
odeoxycholic acid synthesis in cholestatic ham- ing both the 26-hydroxylation of cholesterol and
ster liver. Biochim. Biophys. Acta 1588, 139-148. 25-hydroxylation of vitamin D3: Gonadotropic
1398. Postlind, H., E. Axen, T. Bergman, and K. Wikvall regulation of the cognate mRNA in ovaries. DNA
(1997). Cloning, structure, and expression of a Cell Biol. 9,657-665.
cDNA encoding vitamin D3 25-hydroxylase. 1410. Wikvall, K. (2001). Cytochrome P450 enzymes
Biochem. Biophys. Res. Commun. 241, 491^97. in the bioactivation of vitamin D to its hormonal
1399. Hayashi, S., M. Noshiro, and K. Okuda (1984). form (review). Int. J. Mol. Med. 7, 201-209.
Purification of cytochrome P-450 catalyzing 25- 1411. Pikuleva, I.A., I. Bjorkhelm, and M.R. Waterman
hydroxylation of vitamin D3 from rat liver micro- (1997). Expression, purification, and enzymatic
somes. Biochem. Biophys. Res. Commun. 121, properties of recombinant human cytochrome
994-1000. P450c27 (CYP27). Arch. Biochem. Biophys. 343,
1400. Saarem, K. and J.I. Pedersen (1985). 25- 123-130.
Hydroxylation of la-hydroxyvitamin D3 in rat and 1412. Pikuleva, I.A., A. Babiker, M.R. Waterman, and
human liver. Biochim. Biophys. Acta 840, 117-126. I. Bjorkhem (1998). Activities of recombinant
1401. Akiyoshi-Shibata, M., E. Usui, T. Sakaki, human cytochrome P450c27 (CYP27) which pro-
Y. Yabusaki, M. Noshiro, K. Okuda et al. (1991). duce intermediates of alternative bile acid biosyn-
Expression of rat liver vitamin D3 2 5-hydroxylase thetic pathways, a 5/0/. Chem. 273, 18153-18160.
cDNA in Saccharomyces cerevisiae. FEBS Lett. 1413. Sawada, N., T. Sakaki, M. Ohta, and K. Inouye
280, 367-370. (2000). Metabolism of vitamin D3 by human
1402. Guo, YD., S. Strugnell, D.W. Back, and G. Jones CYP27A1. Biochem. Biophys. Res. Commun.
(1993). Transfected human liver cytochrome 213, 977-984.
P-450 hydroxylates vitamin D analogs at different 1414. Sawada, N., T. Sakaki, S. Kitanaka, S. Kato, and
side-chain positions. Proc. Natl. Acad. Sci. USA K. Inouye (2001). Structure-function analysis of
90, 8668-8672. CYP27B1 and CYP27A1—Studies on mutants
1403. Shiga, K., R. Fukuyama, S. Kimura, K. Nakajima, from patients with vitamin D-dependent rickets
and S. Fushiki (1999). Mutation of the sterol 27- type I (VDDR-I) and cerebrotendinous xan-
hydroxylase gene (CYP27) results in truncation thomatosis (CTX). Eur J. Biochem. 268,
of mRNA expressed in leucocytes in a Japanese 6607-6615.
family with cerebrotendinous xanthomatosis. 1415. Pikuleva, I.A., A. Puchkaev, and I. Bjorkhem
J. Neurol. Neurosurg. Psychiatr. 67, 675-677. (2001). Putative helix F contributes to regio-
1404. Garuti, R., M.A. Croce, R. Tiozzo, M.T. Dotti, selectivity of hydroxylation in mitochon-
A. Federico, S. Bertolini et al. (1997). Four novel drial cytochrome P450 27A1. Biochemistry 40,
mutations of sterol 27-hydroxylase gene in Italian 7621-7629.
patients with cerebrotendinous xanthomatosis. 1416. Murtazina, D., A.V Puchkaev, C.H. Schein,
J. Lipid Res. 38, 2322-2334. N. Oezguen, W Braun, A. Nanavati et al. (2002).
1405. Gascon-Barre, M., C. Demers, O. Ghrab, Membrane-protein interactions contribute to effi-
C. Theodoropoulos, R. Lapointe, G. Jones et al. cient 27-hydroxylation of cholesterol by mito-
(2001). Expression of CYP27A, a gene encoding chondrial cytochrome P450 llAl.J. Biol. Chem.
a vitamin D-25 hydroxylase in human liver and Ill, 37582-37589.
kidney. Clin. Endocrinol. (Oxford) 54, 107-115. 1417. Hosseinpour, F, M. Hidestrand, M. Ingelman-
1406. Shanahan, CM., K.L. Carpenter, and N.R. Gary Sundberg, and K. Wikvall (2001). The
(2001). A potential role for sterol 27-hydroxylase importance of residues in substrate recognition
in atherogenesis. Atherosclerosis 154, 269-276. site 3 for the catalytic function of CYP2D25
1407. Lee, M.J., YC. Huang, M.G. Sweeney, (vitamin D 25-hydroxylase). Biochem. Biophys.
N.W. Wood, M.M. Reilly, and RK. Yip (2002). Res. Commun. 288, 1059-1063.
Mutation of the sterol 27-hydroxylase gene 1418. Wills, M.R. and J. Savory (1984). Vitamin D
(CYP27A1) in a Taiwanese family with cerebro- metabolism and chronic liver disease. Ann. Clin.
tendinous xanthomatosis. J. Neurol. 249, Lab. Sci. 14, 189-197.
1311-1312. 1419. Sugama, S., A. Kimura, W Chen, S. Kubota,
1408. Chen, W., S. Kubota, H. Ujike, T. Ishihara, and Y Seyama, N. Taira et al. (2001). Frontal lobe
Y Seyama (1998). A novel Arg362Ser mutation dementia with abnormal cholesterol metabolism
in the sterol 27-hydroxylase gene (CYP27): and heterozygous mutation in sterol 27-hydroxy-
Its effects on pre-mRNA splicing and enzyme lase gene (CYP27). J. Inherit. Metab. Dis. lA,
activity. Biochemistry 37, 15050-15056. 379-392.
526 F. Peter Guengerich

1420. Bjorkhem, I. (2002). Do oxysterols control cho- 25-hydroxyvitamin D3 l a -hydroxylase. Biochem.


lesterol homeostasis?. J. Clin. Invest. 110, Biophys. Res. Commun. 239, 527-533.
725-730. 1432. Fu, G.K., A.A. Portale, and WL. Miller (1997).
1421. Rosen, H., A. Reshef, N. Maeda, A. Lippoldt, Complete structure of the human gene for the
S. Shpizen, L. Triger et ah (1998). Markedly vitamin D la-hydroxylase, P450cla. DNA Cell
reduced bile acid synthesis but maintained levels Biol. 16, 1499-1507.
of cholesterol and vitamin D metabolites in mice 1433. Zehnder, D., R. Bland, E.A. Walker,
with disrupted sterol 27-hydroxylase gene. A.R. Bradwell, A.J. Howie, M. Hewison et al.
J. Biol. Chem. 273, 14805-14812. (1999). Expression of 25-hydroxyvitamin D3-la-
1422. Reiss, A.B., N.W. Awadallah, S. Malhotra, hydroxylase in the human kidney. J. Am. Soc.
M.C. Montesinos, E.S. Chan, N.B. Javitt et al. Nephrol. 10, 2465-2473.
(2001). Immune complexes and IFN-7 decrease 1434. Zehnder, D., R. Bland, M.C. Williams,
cholesterol 27-hydroxylase in human arterial R.W McNinch, A.J. Howie, RM. Stewart et al.
endothelium and macrophages. J. Lipid Res. 42, (2001). Extrarenal expression of 25-hydroxyvita-
1913-1922. min D3-la-hydroxylase. J. Clin. Endocrinol.
1423. Goodwin, B., K.C. Gauthier, M. Umetani, Metab. 86, 888-894.
M.A. Watson, M.I. Lochansky, J.L. Collins et al. 1435. Zehnder, D., K.N. Evans, M.D. Kilby,
(2003). Identification of bile acid precursors as J.N. Bulmer, B.A. Innes, P.M. Stewart et al.
endogenous ligands for the nuclear xenobiotic (2002). The ontogeny of 25-hydroxyvitamin D3
pregnane X receptor. Proc. Natl. Acad. Sci. USA la-hydroxylase expression in human placenta
100, 223-228. and decidua. Am. J. Pathol. 161, 105-114.
1424. Escher, G., Z. Krozowski, K.D. Croft, and 1436. Diaz, L., C. Arranz, E. Avila, A. HalhaH,
D. Sviridov (2003). Expression of sterol 27- F Vilchis, and F Larrea (2002). Expression and
hydroxylase (CYP27A1) enhances cholesterol activity of 25-hydroxyvitamin D-la-hydroxylase
efflux. J. Biol. Chem. 278, 11015-11019. are restricted in cultures of human syncytiotro-
1425. Fu, X., J.G. Menke, Y. Chen, G. Zhou, phoblast cells from preeclamptic pregnancies.
K.L. MacNaul, S.D. Wright et al. (2001). 27- J. Clin. Endocrinol. Metab. 87, 3876-3882.
Hydroxycholesterol is an endogenous ligand for 1437. Friedrich, M., C. Villena-Heinsen, R. Axt-
liver X receptor in cholesterol-loaded cells. Fliedner, R. Meyberg, W Tilgen, W Schmidt
J. Biol. Chem. 276, 38378-38387. et al. (2002). Analysis of 25-hydroxyvitamin
1426. H.F. DeLuca (1977). Vitamin D as a prohormone. D3-1 a-hydroxylase in cervical tissue. Anticancer
Biochem. Pharmacol. 26, 563-566. Res. 21, 183-186.
1427. Paulson, S.K. and H.F DeLuca (1985). 1438. Segersten, U, P. Correa, M. Hewison, P. Hellman,
Subcellular location and properties of rat renal H. Dralle, T. Carling et al. (2002). 25-hydroxyvi-
25-hydroxyvitamin D3-la-hydroxylase. J. Biol. tamin D3-1 a-hydroxylase expression in normal
Chem. 260, 11488-11492. and pathological parathyroid glands. J. Clin.
1428. M. Burgos-Trinidad, R. Ismail, R.A. Ettinger, Endocrinol. Metab. 87, 2967-2972.
J.M. Prahl, and H.F DeLuca (1992). 1439. Brenza, H.L. and H.F DeLuca (2001). Analysis
Immunopurified 25-hydroxyvitamin D la- of basal regulatory elements in the 25-hydroxyvi-
hydroxylase and 1,25-dihydroxyvitamin D 24- tamin D3 la-hydroxylase gene promoter. Arch.
hydroxylase are closely related but distinct Biochem. Biophys. 388, 121-126.
enzymes. J. Biol. Chem. 267, 3498-3505. 1440. Baxter, L.A. and H.F DeLuca (1976).
1429. Arabian, A., J. Grover, M.G. Barre, and Stimulation of 25-hydroxyvitamin D3-la-hydro-
E.E. Delvin (1993). Rat kidney 25-hydroxyvita- xylase by phosphate depletion. J. Biol. Chem.
min D3 la- and 24-hydroxylases: Evidence for 251,3158-3161.
two distinct gene products. J. Steroid Biochem. 1441. Yoshida, T, N. Yoshida, T. Monkawa, M. Hayashi,
Mol. Biol. 45, 513-516. and T. Saruta (2001). Dietary phosphorus depri-
1430. Axen, E., H. Postlind, H. Sjoberg, and K. Wikvall vation induces 25-hydroxyvitamin D3 la-hydrox-
(1994). Liver mitochondrial cytochrome P450 ylase gene expression. Endocrinology 142,
CYP27 and recombinant expressed human 1720-1726.
CYP27 catalyze la-hydroxylation of 25-hydrox- 1442. Zhang, M.Y, X. Wang, J.T Wang,
yvitamin D3. Proc. Natl. Acad. Sci. USA 91, N.A. Compagnone, S.H. Mellon, J.L. Olson et al.
10014-10018. (2002). Dietary phosphorus transcriptionally reg-
1431. Monkawa, T., T. Yoshida, S. Wakino, T. Shinki, ulates 25-hydroxyvitamin D-la-hydroxylase
H. Anazawa, H.F.DeLuca et al. (1997). Molecular gene expression in the proximal renal tubule.
cloning of cDNA and genomic DNA for human Endocrinology 143, 587-595.
Human P450s 527

1443. Bland, R., E.A. Walker, S.V Hughes, P.M. Stewart, D3 la-hydroxylase gene in patients with
and M. Hewison (1999). Constitutive expression of pseudovitamin D-deficiency rickets. A^. Engl.
25-hydroxyvitamin D3-la-hydroxylase in a trans- J. Med 33H, 653-661.
formed human proximal tubule cell line: Evidence 1454. Smith, S.J., A.K. Rucka, J.L. Berry, M. Davies,
for direct regulation of vitamin D metabolism by S. Mylchreest, C.R. Paterson et al. (1999). Novel
calcium. Endocrinology 140, 2027-2034. mutations in the la-hydroxylase (P450cl) gene
1444. Takeyama, K., S. Kitanaka, T. Sato, M. Kobori, in three families with pseudovitamin D-
J. Yanagisawa, and S. Kato (1997). 25- deficiency rickets resulting in loss of functional
Hydroxyvitamin D3 la-hydroxylase and vitamin enzyme activity in blood-derived macrophages.
D synthesis. Science 111, 1827-1830. J. Bone Miner Res. 14, 730-739.
1445. Murayama, A., K. Takeyama, S. Kitanaka, 1455. Sawada, N., T. Sakaki, S. Kitanaka, K. Takeyama,
Y. Kodera, T. Hosoya, and S. Kato (1998). The S. Kato, and K. Inouye (1999). Enzymatic
promoter of the human 25-hydroxyvitamin D3 properties of human 25-hydroxyvitamin D3 la-
la-hydroxylase gene confers positive and nega- hydroxylase coexpression with adrenodoxin and
tive responsiveness to PTH, calcitonin, and NADPH-adrenodoxin reductase in Escherichia
la,25(OH)2D3. Biochem. Biophys. Res. Commun. coll Eur J. Biochem. 265, 950-956.
249, 11-16. 1456. Wang, X., M.Y Zhang, WL. Miller, and
1446. Kong, X.E, X.H. Zhu, Y.L. Pei, D.M. Jackson, A.A. Portale (2002). Novel gene mutations in
and M.F. Holick (1999). Molecular cloning, char- patients with la-hydroxylase deficiency that con-
acterization, and promoter analysis of the human fer partial enzyme activity in vitro. J. Clin.
25-hydroxyvitamin D3-la-hydroxylase gene. Endocrinol. Metab. 87, 2424-2430.
Proc. Natl. Acad. Sci. USA 96, 6988-6993. 1457. Muralidharan, K.R., M. Rowland-Goldsmith,
1447. Gao, X.H., PR Dwivedi, S. Choe, E Alba, A.S. Lee, G. Park, A.W. Norman, H.L. Henry
H.A. Morris, J.L. Omdahl et al. (2002). Basal and et al. (1997). Inhibitors of 25-hydroxyvitamin D3-
parathyroid hormone induced expression of the la-hydroxylase: Thiavitamin D analogs and bio-
human 25-hydroxyvitamin D la-hydroxylase logical evaluation. J. Steroid Biochem. 62,13-lS.
gene promoter in kidney AOK-B50 cells: Role of 1458. Hewison, M., D. Zehnder, R. Bland, and
Spl, Ets and CCAAT box protein binding sites. PM. Stewart (2000). la-Hydroxylase and the
Int. J. Biochem. Cell Biol. 34, 921-930. action of vitamin D. J. Mol. Endocrinol. 25,
1448. Michigami, T, H. Yamato, H. Suzuki, Y. Nagai- 141-148.
Itagaki, K. Sato, and K. Ozono (2001). Conflicting 1459. Kitanaka, S., K. Takeyama, A. Murayama,
actions of parathyroid hormone-related protein and and S. Kato (2001). The molecular basis of
serum calcium as regulators of 25-hydroxyvitamin vitamin D-dependent rickets type I. Endocrinol.
D3-la-hydroxylase expression in a nude rat J. 48, 427-432.
model of humoral hypercalcemia of malignancy. 1460. Portale, A.A. and W.L. Miller (2000). Human 25-
J. Endocrinol. Ill, 249-257. hydroxyvitamin D-la-hydroxylase: Cloning,
1449. Kato, S., T. Yoshizazawa, S. Kitanaka, mutations, and gene expression. Pediatr Nephrol.
A. Murayama, and K. Takeyama (2002). 14, 620-625.
Molecular genetics of vitamin D-dependent 1461. Kitanaka, S., A. Murayama, T. Sakaki, K. Inouye,
hereditary rickets. Horm. Res. 57, 73-78. Y. Seino, S. Fukumoto et al. (1999). No enzyme
1450. Wang, J.T., C.J. Lin, S.M. Burridge, O.K. Fu, activity of 25-hydroxyvitamin D3 la-hydroxylase
M. Labuda, A.A. Portale et al. (1998). Genetics gene product in pseudovitamin D deficiency rick-
of vitamin D la-hydroxylase deficiency in 17 ets, including that with mild clinical manifesta-
families. Am. J. Hum. Genet. 63, 1694-1702. tion. J. Clin. Endocrinol. Metab. 84, 4111^117.
1451. Portale, A.A. and W.L. Miller (2000). Human 25- 1462. Dardenne, O., J. Prud'homme, A. Arabian,
hydroxyvitamin D-la-hydroxylase: Cloning, EH. Glorieux, and R. St-Arnaud (2001). Targeted
mutations, and gene expression. Pediatr Nephrol. inactivation of the 25-hydroxyvitamin D3-la-
14, 620-625. hydroxylase gene (CYP27B1) creates an animal
1452. Porcu, L., A. Meloni, L. Casula, I. Asunis, model of pseudovitamin D-deficiency rickets.
M.G. Marini, A. Cao et al. (2002). A novel Endocrinology 142,3135-3141.
sphcing defect (IVS6+1G>T) in a patient 1463. Panda, D.K., D. Miao, M.L. Tremblay, J. Sirois,
with pseudovitamin D deficiency rickets. R. Earookhi, G.N. Hendy et al. (2001). Targeted
J. Endocrinol. Invest. 25, 557-560. ablation of the 25-hydroxyvitamin D la-hydrox-
1453. Kitanaka, S., K. Takeyama, A. Murayama, ylase enzyme: Evidence for skeletal, reproduc-
T Sato, K. Okumura, M. Nogami et al. (1998). tive, and immune dysfiinction. Proc. Natl. Acad.
Inactivating mutations in the 25-hydroxyvitamin Sci. USA 98, 7498-7503.
528 F. Peter Guengerich

1464. St-Arnaud, R., O. Dardenne, J. Prud'homme, oxysterol 7a-hydroxylase selective for 24-hydrox-
S.A. Hacking, and KH. Glorieux (2003). ycholesterol. ^ 5/0/. Chem. 275, 16543-16549.
Conventional and tissue-specific inactivation 1475. Lund, E.G., J.M. Guileyardo, and D.W. Russell
of the 25-hydroxyvitamin D-la-hydroxylase (1999). cDNA cloning of cholesterol 24-hydroxy-
(CYP27B1). J. Cell. Biochem. 88, 245-251. lase, a mediator of cholesterol homeostasis in the
1465. Satomura, K., Y. Seino, K. Yamaoka, Y Tanaka, brain. Proc. Natl. Acad Sci. USA 96, 7238-7243.
M. Ishida, H. Yabuuchi et al. (1988). Renal 25- 1476. Russell, D.W. (2000). Oxysterol biosynthetic
hydroxyvitamin D3-I-hydroxylase in patients enzymes. Biochim. Biophys.Acta 1529, 126-135.
with renal disease. Kidney Int. 34, 712-716. 1477. Bogdanovic, N., L. Bretillon, E.G. Lund,
1466. Correa, R, U. Segersten, P. Hellman, U Diczfalusy, L. Lannfelt, B. Winblad et al.
G. Akerstrom, and G. Westin (2002). Increased (2001). On the turnover of brain cholesterol in
25-hydroxyvitamin D3 la-hydroxylase and patients with Alzheimer's disease. Abnormal
reduced 25-hydroxyvitamin D3 24-hydroxylase induction of the cholesterol-catabolic enzyme
expression in parathyroid tumors—^new prospects CYP46 in glial cells. Neurosci. Lett. 314, 45-48.
for treatment of hyperparathyroidism with vitamin 1478. Papassotiropoulos, A., D. Lutjohann, M. Bagli,
D. J. Clin. Endocrinol. Metab. 87, 5826-5829. S. Locatelli, E Jessen, R. Buschfort et al. (2002).
1467. Maas, R.M., K. Reus, B. Diesel, W.I. Steudel, 24iS-hydroxycholesterol in cerebrospinal fluid is
W. Feiden, U. Fischer et al. (2001). Amplification elevated in early stages of dementia. J. Psychiatr
and expression of splice variants of the gene Res. 36, 27-32.
encoding the P450 cytochrome 25-hydroxyvita- 1479. Kolsch, H., D. Lutjohann, M. Ludwig, A. Schulte,
min D3 1 ,a-hydroxylase {CYP 27B1) in human U Ptok, F. Jessen et al. (2002). Polymorphism in
malignant glioma. Clin. Cancer Res. 7, 868-875. the cholesterol 245-hydroxylase gene is associ-
1468. Tangpricha, V, J.N. Flanagan, L.W. Whitlatch, ated with Alzheimer's disease. Mol. Psychiatry 7,
C.C. Tseng, T.C. Chen, PR. Hoh et al. (2001). 25- 899-902.
hydroxyvitamin D-la-hydroxylase in normal and 1480. Desai, P, S.T. DeKosky, and M.I. Kamboh
malignant colon tissue. Lancet 357, 1673-1674. (2002). Genetic variation in the cholesterol 24-
1469. Cross, H.S., P Bareis, H. Hofer, M.G. Bischof, hydroxylase (CYP46) gene and the risk of
E. Bajna, S. Kriwanek et al. (2001). 25- Alzheimer's disease. Neurosci. Lett. 328, 9-12.
Hydroxyvitamin D3-1 a-hydroxylase and vitamin 1480a.Mast, N., R. Norcross, U. Andersson, M. Shou, K.
D receptor gene expression in human colonic Nakayama, I. Bjorkhem, and LA. Pikuleva (2003).
mucosa is elevated during early cancerogenesis. Broad substrate specificity of human cytochrome
Steroids 66, 287-292. P450 46A1 which initiates cholesterol degradation
1470. Ogunkolade, B.W., B.J. Boucher, PD. Fairclough, in the brain. Biochemistry 42, 14284--14292.
G.A. Hitman, S. Dorudi, PJ. Jenkins etal. (2002). 1481. Aoyama, Y, Y Funae, M. Noshiro, T. Horiuchi,
Expression of 25-hydroxyvitamin D-1-a-hydrox- and Y Yoshida (1994). Occurrence of a P450
ylase mRNA in individuals with colorectal cdiX\- showing high homology to yeast lanosterol 14-
CQT. Lancet 359, 1831-1832. demethylase (P450,4p^) in rat liver. Biochem.
1471. Hsu, J.Y, D. Feldman, J.E. McNeal, and DM. Peehl Biophys. Res. Commun. 201, 1320-1326.
(2001). Reduced la-hydroxylase activity in human 1482. Aoyama, Y, T. Horiuchi, and Y Yoshida (1996).
prostate cancer cells correlates with decreased sus- Lanosterol 14-demethylase activity expressed in
ceptibility to 25-hydroxyvitamin Dj-induced rat brain microsomes. J. Biochem. (Tokyo) 120,
growth inhibition. Cancer Res. 61, 2852-2856. 982-986.
1472. Whitlatch, L.W, M.V Young, G.G. Schwartz, 1483. Rozman, D, M. Stromstedt, and M.R. Waterman
J.N. Flanagan, K.L. Burnstein et al. (2002). 25- (1996). The three human cytochrome P450 lanos-
Hydroxyvitamin D-la-hydroxylase activity is terol 14a-demethylase (CYP51) genes reside on
diminished in human prostate cancer cells and is chromosomes 3, 7, and 13: Structure of the two
enhanced by gene transfer. J. Steroid Biochem. retrotransposed pseudogenes, association with a
Mol. Biol. 81, 135-140. line-1 element, and evolution of the human CYP51
1473. Flanagan, J.N., L.W. Whitlatch, T.C. Chen, family. Arch. Biochem. Biophys. 333, 466-474.
X.H. Zhu, M.T Holick, X.R Kong et al. (2001). 1484. Cotman, M., D. Rozma, L. Banek, and D. Jezek
Enhancing la-hydroxylase activity with the (2001). Localisation of lanosterol 14a-demethylase
25-hydroxyvitamin D-la-hydroxylase gene in in round and elongated spermatids of the mouse
cultured human keratinocytes and mouse skin. testis: An immunoelectron microscopic and stereo-
J. Invest. Dermatol. 116, 910-914. logical study PJlugers Arch. 442, R167-R168.
1474. Li-Hawkins, J., E.G. Lund, A.D. Bronson, and 1485. Rozman, D (2000). Lanosterol 14a-demethylase
D.W Russell (2000). Expression cloning of an (CYP51)—a cholesterol biosynthetic enzyme
Human P450s 529

involved in production of meiosis activating 1496. Podust, L.M., J. Stojan, T.L. Poulos, and
sterols in oocytes and testis—a minireview. M.R. Waterman (2001). Substrate recognition
Pflugers Arch. 439, R56-R57. sites in 14 a-sterol demethylase from comparative
1486. Kelley, R.I, L.E. Kratz, R.L. Glaser, analysis of amino acid sequences and X-ray
M.L. Netzloff, L.M. Wolf, and E.W. Jabs (2002). structure of Mycobacterium tuberculosis CYP51.
Abnormal sterol metabolism in a patient with J. Inorg. Biochem. 87, 227-235.
Antley-Bixler syndrome and ambiguous geni- 1497. Marichal, P., L. Koymans, S. Willemsens,
talia. Am. J. Med. Genet. 110, 95-102. D. Bellens, R Verhasselt, W Luyten et al. (1999).
1487. Debeljak, N., M. Fink, and D. Rozman (2003). Contribution of mutations in the cytochrome
Many facets of mammalian lanosterol 14a- P450 14a-demethylase (Ergllp, CYP51p) to
demethylase from the evolutionarily conserved azole resistance in Candida albicans.
cytochrome P450 family CYP51. Arch. Biochem. Microbiology 145, 2701-2713.
Biophys. 409, 159-171. 1498. Lepesheva, G.I., L.M. Podust, A. Bellamine, and
1488. Stromstedt, M., D. Rozman, and M.R. Waterman M.R. Waterman (2001). Folding requirements are
(1996). The ubiquitously expressed human different between sterol 14a-demethylase
CYP51 cDNA encodes lanosterol 14a-demethy- (CYP51) from Mycobacterium tuberculosis and
lase, a cytochrome P450 whose expression is reg- human or fungal orthologs. J. Biol. Chem. 276,
ulated by oxysterols. Arch. Biochem. Biophys. 28413-28420.
329,73-81. 1499. Lamb, D.C., D.E. Kelly, M.R. Waterman,
1489. Rozman, D., M. Fink, G.M. Fimia, P Sassone- M. Stromstedt, D. Rozman, and S.L. Kelly
Corsi, and M.R. Waterman (1999). Cychc (1999). Characteristics of the heterologously
adenosine 3',5'-monophosphate(cAMP)/ cAMP- expressed human lanosterol 14a-demethylase
responsive element modulator (CREM)-depend- (other names: P45014DM, CYP51, P45051) and
ent regulation of cholesterogenic lanosterol inhibition of the purified human and Candida
14a-demethylase (CYP51) in spermatids. Mol. albicans CYP51 with azole antifungal agents.
Endocrinol. 13, 1951-1962. Yeast 15, 755-763.
1490. Haider, S.K., M. Fink, M.R. Waterman, and 1500. Rozman, D. and M.R. Waterman (1998).
D. Rozman (2002). A cAMP-responsive element Lanosterol 14a-demethylase (CYP51) and sper-
binding site is essential for sterol regulation of the matogenesis. DrugMetab. Dispos. 26, 1199-1201.
human lanosterol 14a-demethylase gene 1501. Majdic, G., M. Parvinen, A. Bellamine,
(CYP51). Mol. Endocrinol. 16, 1853-1863. H.J. Harwood, Jr., WW Ku, M.R. Waterman, and
1491. Yamashita, C , M. Kudo, H. Ishida, M. Noshiro, D. Rozman (2000). Lanosterol 14a-demethylase
Y. Aoyama, and Y Yoshida (2000). Insulin is the (CYP51), NADPH-cytochrome P450 reductase
essential factor maintaining the constitutive and squalene synthase in spermatogenesis: Late
expression of hepatic sterol 14-demethylase P450 spermatids of the rat express proteins needed to
(CYP51). J. Biochem. (Tokyo) 128, 93-99. synthesize follicular fluid meiosis activating
1492. Rodriguez, C , J. Martinez-Gonzalez, S. Sanchez- sterol. J. Endocrinol. 166, 463-474.
Gomez, and L. Badimon (2001). LDL downregu- 1502. Yamashita, C , Y Aoyama, M. Noshiro, and
lates CYP51 in porcine vascular endothelial cells Y Yoshida (2001). Gonadotropin-dependent
and in the arterial wall through a sterol regulatory expression of sterol 14-demethylase P450 (CYP51)
element binding protein-2-dependent mecha- in rat ovaries and its contribution to the production
nism. Circ. Res. 88, 268-274. of a meiosis-activating steroid. J. Biochem. (Tokyo)
1493. Lamb, D.C., N.N. Kaderbhai, K. Venkateswarlu, 130, 849-856.
D.E. Kelly, S.L. Kelly, and M.A. Kaderbhai (2001). 1503. Rozman, D., M. Cotman, and R. Frangez
Human sterol 14a-demethylase activity is enhanced (2002). Lanosterol 14a-demethylase and MAS
by the membrane-bound state of cj^ochrome ^^3. sterols in mammalian gametogenesis. Mol. Cell
Arch. Biochem. Biophys. 395, 78-84. Endocrinol. 187, 179-187.
1494. Lamb, D.C., D.E. Kelly, and S.L. Kelly (1998). 1504. Kodaira, H., C.A. Lisek, A. Arimura, I. Jardine,
Molecular diversity of sterol 14a-demethylase and S. Spector (1989). Identification of the con-
substrates in plants, fungi and humans. FEBS vulsant opiate thebaine in mammalian brain.
Lett. 425, 263-265. Proc. Natl. Acad. Sci. USA 86, 716-719.
1495. Podust, L.M., T.L. Poulos, and M.R. Waterman 1505. Kodaira, H. and S. Spector (1988).
(2001). Crystal structure of cytochrome P450 14a- Transformation of thebaine to oripavine, codeine,
sterol demethylase (CYP51) from Mycobacterium and morphine by rat liver, kidney, and brain
tuberculosis in complex with azole inhibitors. microsomes. Proc. Natl. Acad. Sci. USA 85,
Proc. Natl. Acad Sci. USA 98, 3068-3073. 1267-1271.
530 F. Peter Guengerich

1506. Dayer, P., J. Desmeules, T. Leemann, and demethylation by Escherichia coli AlkB
R. Striberni (1988). Bioactivation of the narcotic directly reverts DNA base damage. Nature 12,
drug codeine in human liver is mediated by 174-178.
the polymorphic monooxygenase catalyzing 1509. Welford, R.W., I. Schlemminger, L.A. McNeill,
debrisoquine 4-hydroxylation. Biochem. Biophys. K.S. Hewitson, and C.J. Schofield (2003). The
Res. Commun. 152,411^16. selectivity and inhibition of AlkB. J. Biol. Chem.
1507. Mikus, G., F. Bochner, M. Eichelbaum, P. Horak, 278, 10157-10161.
A.A. Somogyi, and S. Spector (1994). Endo- 1510. Pai, H.V, R.R Kommaddi, S.J. Chinta, T. Mori,
genous codeine and morphine in poor and exten- M.R. Boyd, and V Ravindranath (2004, in press).
sive metabolisers of the CYP2D6 (debrisoquine/ A frame shift mutation and alternate splicing
sparteine) polymorphism. J. Pharmacol. Exp. in human brain generates a functional form of
Then 268,546-551. the pseudogene, cytochrome P4502D7 that
1508. Trewick, S.C, T.K Henshaw, R.R Hausinger, demethylates codeine to morphine. J. Biol. Chem.
T. Lindahl, and B. Sedgwick (2002). Oxidative 39774.

You might also like