Top Banner
Communication Vol. 269, No. 22, Issue of June 3, pp. 15419-15422, 1994 THE Jnm.a nF Blnmlca CHEMISTRY Printed in U.S.A. The Major Genetic Defect Responsible for the Polymorphism of S-Mephenytoin Metabolism in Humans* (Received for publication, March 15, 1994) Sonia M. F. de MoraisS, Grant R. WilkinsonBI, Joyce Blaisdellt, Koichi Nakamurall, Urs A. Meyer**$$, and Joyce A. Goldstein$$§ From the WIEHS, National Institutesof Health, Research Diangle Park, North Carolina27709, the $Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, the /Department of Clinical Pharmacology and Therapeutics, Medical College of Oita, 879-55 Oita, Japan, and the **Department of Pharmacology, Biozentrum, University of Basel, Basel CH-4056, Switzerland The metabolism of the anticonvulsant drug mepheny- toin exhibits a genetic polymorphism in humans, with the poor metabolizer trait being inherited in an autoso- mal recessive fashion. There are large interracial differ- ences in the frequency of the poor metabolizer pheno- type, with Oriental populations having a 5-fold greater frequency compared to Caucasians. Impaired metabo- lism of mephenytoin and a number of other currently used drugs results from a defect in a cytochrome P450 enzyme recently identified as CYP2C19. Attempts over the past decadeto define the molecular genetic basis of the polymorphism have, however, been unsuccessful. We now report that the principal defect in poor metaboliz- ers is a single base pair (G + A) mutation in exon 5 of CYP2C19, which creates an aberrant splice site. This change alters the reading frame of the mRNA starting with amino acid 215 and produces a premature stop codon 20 amino acids downstream, which results in a truncated, non-functional protein. We further demon- strate that 7/10 Caucasian and 10/17Japanese poor me- tabolizers are homozygous for this defect, indicating that this is the major defect responsible for the poor metabolizer phenotype. Finally, the familial inheritance of the deficient allele was found to be concordantwith that of the phenotypic trait. Severalgeneticpolymorphisms of drugmetabolismhave been documented in humans (1). One of the best characterized is that associated with the 4’-hydroxylation of the S-enanti- omer of the anticonvulsant mephenytoin (2-4). Individuals can * The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “aduertisement” in accordance with 18 U.S.C.Section1734solelyto indicate this fact. The nucleotide sequence(s) reported in this paper has been submitted and L31507. to the GenBankmIEMBL Data Bank with accession number(s) L31506 GM-31304. 1 Supported by United States Public Health Service Grant $$ Supported by the Swiss National Science Foundation. dressed: NIEHS, P. 0. Box 12233, Research Triangle Park, NC 27709. $5 To whom correspondence and reprint requests should be ad- Tel.: 919-541-4495; Fax: 919-541-3647. be characterized as either extensive (EM)’ or poor (PM) me- tabolizers. The latter phenotype is inherited in an autosomal recessive fashion (5,6) with the EM phenotype comprising both the homozygous dominant and heterozygote genotypes. There are marked interracial differences in the frequency of this poly- morphism. For example, the PM phenotype occurs in 2-5% of Caucasian populations but at higher frequencies (18-23%) in Oriental populations (2, 7). This polymorphism affects the me- tabolism of a number of other commonly used drugs, for ex- ample omeprazole (81, proguanil (9), certain barbiturates (10, 111, and citalopram (12). As a result, large interphenotypic differences occur in the disposition of these drugs, which may affect their efficacy and toxicity. The oxidation of propranolol (13), certain tricyclic antidepressants (14-161, and possibly di- azepam (17) is also affected, albeit to a lesser extent. Recent studies have shown that CYP2C19 is the enzyme responsible for the 4’-hydroxylation of S-mephenytoin in hu- man liver and that the levels of CYP2C19 protein correlate with microsomal S-mephenytoin 4‘-hydroxylase activities in human livers (18, 19). However, the molecular basis of the PM phenotype is not known. The purpose of the present study was to determine the molecular genetic mechanism of the defect that is responsible for the polymorphism of S-mephenytoin me- tabolism in humans. MATERIALS AND METHODS Analysis of Human Liver Microsomes-Liver microsomes were pre- pared by differential centrifugation from 13 human liver samples se- lected from organ donors that had been previously characterized in vitro (20) as varying markedly in their S-mephenytoin 4’-hydroxylase activ- ity. For immunoblot analysis of CYP2C19, liver microsomal proteins were separated by SDS-polyacrylamide gel electrophoresis,transferred to nitrocellulose membranes, and developed with a polyclonal antibody to CYP2C9 that also recognizes CYP2C19 using the ECL chemilumi- nescence kit (Amersham Corp.) as previously described (19). Results were confirmed with a specific peptide antibody to CYP2C19. Liver microsomalR- and S-mephenytoin 4‘-hydroxylase activities were meas- ured by high performance liquid chromatography analysis (21). Amplification of CYP2C19 mRNA-Total liver RNA was isolated from liver samples using a single-step method (22) with Tri-reagent (Molecular Research Center Inc.) and reverse-transcribed as previously described (19). In initial experiments, the polymerase chain reaction (PCR) was used to amplify overlapping CYP2C19 cDNA fragments en- compassing the full-length cDNA from three selected human liver samples, which had low microsomal S-mephenytoin 4’-hydroxylase ac- tivity, a high ratio of hydroxylation of the RIS enantiomers of mephe- nytoin (201, and the virtual absence of CYP2C19 by immunoblotting as described above. The cDNA was amplified in 1 x PCR buffer (67 m Tris-HC1,pH8.8, 17 mM (NH,),SO,, 10 mM P-mercaptoethanol, 7 J~M EDTA, 0.2 mg/ml bovine serum albumin) containing 50 p UTP, dCTP, dGTP, and dTTP, 0.25 p PCR primers, 2.5 units of AmpliTaq DNA polymerase (Perkin-Elmer), and 1.0 mM MgC1,. The amplification was performed using a Perkin-Elmer thermocycler for 30 cycles consisting of denaturation at 94 “C for 1 min, annealing at the appropriate tempera- ture for 30 s, and extension at 72 “C for 1 min. An initial denaturation step at 94 “C for 3 min and a final extension step at 72 “C for 10 min were also performed. The PCR fragments were then subcloned into the SmaI site of pBluescript I1 SK’ (Stratagene). Plasmids were purified with Qiagen kits and sequenced with an automated sequencer, using the cycle sequencing reaction employing fluorescence-tagged dye termi- nators (PRISM, AppliedBiosystems). RNA from all 13 human liver donors was subsequently reverse-tran- scribed and amplified using the forward primer 5”ATTGAATGU- The abbreviations used are: EM, extensive metabolizer; PM, poor metabolizer; PCR, polymerase chain reaction; bp, base pair(s). 15419
4

The major genetic defect responsible for the polymorphism of S ...

Jan 02, 2017

Download

Documents

vanbao
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: The major genetic defect responsible for the polymorphism of S ...

Communication Vol. 269, No. 22, Issue of June 3, pp. 15419-15422, 1994 THE J n m . a nF B l n m l c a CHEMISTRY

Printed in U.S.A.

The Major Genetic Defect Responsible for the Polymorphism of S-Mephenytoin Metabolism in Humans*

(Received for publication, March 15, 1994)

Sonia M. F. de MoraisS, Grant R. WilkinsonBI, Joyce Blaisdellt, Koichi Nakamurall, Urs A. Meyer**$$, and Joyce A. Goldstein$$§ From the WIEHS, National Institutes of Health, Research Diangle Park, North Carolina 27709, the $Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, the /Department of Clinical Pharmacology and Therapeutics, Medical College of Oita, 879-55 Oita, Japan, and the **Department of Pharmacology, Biozentrum, University of Basel, Basel CH-4056, Switzerland

The metabolism of the anticonvulsant drug mepheny- toin exhibits a genetic polymorphism in humans, with the poor metabolizer trait being inherited in an autoso- mal recessive fashion. There are large interracial differ- ences in the frequency of the poor metabolizer pheno- type, with Oriental populations having a 5-fold greater frequency compared to Caucasians. Impaired metabo- lism of mephenytoin and a number of other currently used drugs results from a defect in a cytochrome P450 enzyme recently identified as CYP2C19. Attempts over the past decade to define the molecular genetic basis of the polymorphism have, however, been unsuccessful. We now report that the principal defect in poor metaboliz- ers is a single base pair (G + A) mutation in exon 5 of CYP2C19, which creates an aberrant splice site. This change alters the reading frame of the mRNA starting with amino acid 215 and produces a premature stop codon 20 amino acids downstream, which results in a truncated, non-functional protein. We further demon- strate that 7/10 Caucasian and 10/17 Japanese poor me- tabolizers are homozygous for this defect, indicating that this is the major defect responsible for the poor metabolizer phenotype. Finally, the familial inheritance of the deficient allele was found to be concordant with that of the phenotypic trait.

Several genetic polymorphisms of drug metabolism have been documented in humans (1). One of the best characterized is that associated with the 4’-hydroxylation of the S-enanti- omer of the anticonvulsant mephenytoin (2-4). Individuals can

* The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “aduertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

The nucleotide sequence(s) reported in this paper has been submitted

and L31507. to the GenBankmIEMBL Data Bank with accession number(s) L31506

GM-31304. 1 Supported by United States Public Health Service Grant

$$ Supported by the Swiss National Science Foundation.

dressed: NIEHS, P. 0. Box 12233, Research Triangle Park, NC 27709. $5 To whom correspondence and reprint requests should be ad-

Tel.: 919-541-4495; Fax: 919-541-3647.

be characterized as either extensive (EM)’ or poor (PM) me- tabolizers. The latter phenotype is inherited in an autosomal recessive fashion (5,6) with the EM phenotype comprising both the homozygous dominant and heterozygote genotypes. There are marked interracial differences in the frequency of this poly- morphism. For example, the PM phenotype occurs in 2-5% of Caucasian populations but at higher frequencies (18-23%) in Oriental populations (2, 7). This polymorphism affects the me- tabolism of a number of other commonly used drugs, for ex- ample omeprazole (81, proguanil (9), certain barbiturates (10, 111, and citalopram (12). As a result, large interphenotypic differences occur in the disposition of these drugs, which may affect their efficacy and toxicity. The oxidation of propranolol (13), certain tricyclic antidepressants (14-161, and possibly di- azepam (17) is also affected, albeit to a lesser extent.

Recent studies have shown that CYP2C19 is the enzyme responsible for the 4’-hydroxylation of S-mephenytoin in hu- man liver and that the levels of CYP2C19 protein correlate with microsomal S-mephenytoin 4‘-hydroxylase activities in human livers (18, 19). However, the molecular basis of the PM phenotype is not known. The purpose of the present study was to determine the molecular genetic mechanism of the defect that is responsible for the polymorphism of S-mephenytoin me- tabolism in humans.

MATERIALS AND METHODS Analysis of Human Liver Microsomes-Liver microsomes were pre-

pared by differential centrifugation from 13 human liver samples se- lected from organ donors that had been previously characterized in vitro (20) as varying markedly in their S-mephenytoin 4’-hydroxylase activ- ity. For immunoblot analysis of CYP2C19, liver microsomal proteins were separated by SDS-polyacrylamide gel electrophoresis, transferred to nitrocellulose membranes, and developed with a polyclonal antibody to CYP2C9 that also recognizes CYP2C19 using the ECL chemilumi- nescence kit (Amersham Corp.) as previously described (19). Results were confirmed with a specific peptide antibody to CYP2C19. Liver microsomal R- and S-mephenytoin 4‘-hydroxylase activities were meas- ured by high performance liquid chromatography analysis (21).

Amplification of CYP2C19 mRNA-Total liver RNA was isolated from liver samples using a single-step method (22) with Tri-reagent (Molecular Research Center Inc.) and reverse-transcribed as previously described (19). In initial experiments, the polymerase chain reaction (PCR) was used to amplify overlapping CYP2C19 cDNA fragments en- compassing the full-length cDNA from three selected human liver samples, which had low microsomal S-mephenytoin 4’-hydroxylase ac- tivity, a high ratio of hydroxylation of the RIS enantiomers of mephe- nytoin (201, and the virtual absence of CYP2C19 by immunoblotting as described above. The cDNA was amplified in 1 x PCR buffer (67 m Tris-HC1, pH 8.8, 17 mM (NH,),SO,, 10 mM P-mercaptoethanol, 7 J ~ M

EDTA, 0.2 mg/ml bovine serum albumin) containing 50 p UTP, dCTP, dGTP, and dTTP, 0.25 p PCR primers, 2.5 units of AmpliTaq DNA polymerase (Perkin-Elmer), and 1.0 mM MgC1,. The amplification was performed using a Perkin-Elmer thermocycler for 30 cycles consisting of denaturation at 94 “C for 1 min, annealing at the appropriate tempera- ture for 30 s, and extension at 72 “C for 1 min. An initial denaturation step at 94 “C for 3 min and a final extension step at 72 “C for 10 min were also performed. The PCR fragments were then subcloned into the SmaI site of pBluescript I1 SK’ (Stratagene). Plasmids were purified with Qiagen kits and sequenced with an automated sequencer, using the cycle sequencing reaction employing fluorescence-tagged dye termi- nators (PRISM, Applied Biosystems).

RNA from all 13 human liver donors was subsequently reverse-tran- scribed and amplified using the forward primer 5”ATTGAATGU-

The abbreviations used are: EM, extensive metabolizer; PM, poor metabolizer; PCR, polymerase chain reaction; bp, base pair(s).

15419

Page 2: The major genetic defect responsible for the polymorphism of S ...

15420 Polymorphism in Mephenytoin Hydroxylation CATCAGGATTG-3' and the reverse primer 5"GTAAGTCAGCTG- CAGTGATTA-3' and the strategy shown in Fig. 1 to detect aberrant splicing of exon 5. PCR conditions were similar to those described above. PCR products were analyzed on 3% agarose gels and stained with ethidium bromide. Selected PCR products were sequenced directly, after purification using Microcon columns (Amicon).

Phenotyping Procedures-The in vivo phenotype of most of the Swiss subjects was based on their "hydroxylation index" values (2), where a value above 5.6 identifies a PM. The phenotype of American subjects, Japanese subjects, and one Swiss subject was based on the urinary SIR ratio as described previously (3), with a poor metabolizer being defined as having a ratio > 0.9. One American subject was of a rare intermediate phenotype characterized by the extent of 4'-hydroxylation being greater than in PMs, but with the rate of the metabolite's formation being slower than in EMS (23).

Genotyping Procedure-DNA was isolated (24) from human blood of selected Caucasian and Japanese subjects who had been previously phenotyped as described above. These populations contained an in- tended overrepresentation of PMs. PCR conditions were similar to those described previously, except that reactions used 200 ng of genomic DNA, 3 mM MgCI,, and an initial denaturation a t 94 "C for 5 min. The forward primer was 5'-AATTACAACCAGAGC'I"GGC-3' and the reverse primer 5'-TATCACT"TCCATAAAAGCAAG-3'. PCR products were re- stricted with SmaI in the PCR buffer, without purification. Digested PCR products were analyzed on 4% agarose gels stained with ethidium bromide. PCR products of genomic DNA from three individuals who were homozygous-extensive, heterozygous-extensive, and homozygous- poor metabolizers (based on their SmaI restriction digests and their in vivo phenotypes) were sequenced using an automated sequenator (Applied Biosystems). PCR products were purified using Microcon columns and sequenced using the same forward primer used in the PCR reaction.

RESULTS AND DISCUSSION

We initially amplified and sequenced overlapping CYP2C19 cDNA fragments from liver samples of selected human organ donors, which had low microsomal S-mephenytoin 4"hydrox- ylase activity, a high ratio of hydroxylation of the R IS enanti- omers of mephenytoin (20), and the virtual absence of CYP2C19 by immunoblotting. One aberrant CYP2C19 cDNA fragment was identified with a 40-bp deletion at the beginning of exon 5 (from bp 643 to bp 682), which included the deletion of a SmaI restriction site. This alteration results in the deletion of amino acids 215-227 and shifts the reading frame beginning a t amino acid 215, producing a premature stop codon 20 amino acids downstream. The resultant truncated 234-amino acid protein would lack the heme-binding region and, therefore, would be catalytically inactive.

RNA was then reverse-transcribed from a total of 13 liver donors, which were selected based on a wide range of S-mephe- nytoin hydroxylase activities as determined in vitro, and the PCR strategy shown in Fig. lA was used to amplify the region encompassing the 40-bp deletion. Only the aberrantly spliced mRNA (244-bp fragment) was found in sample 35, the charac- teristics of which were most consistent with the PM phenotype (low liver microsomal S-mephenytoin 4'-hydroxylase activity and a ratio of >1.0 for metabolism of the RIS enantiomers) (Fig. 1B ). In contrast, liver samples with the highest S-mephenytoin 4'-hydroxylase activities and RIS ratios of <0.1 contained only the normally spliced mRNA, while several liver samples with intermediate catalytic activities, RIS ratios of 0.1-0.8, and low CYP2C19 levels possessed both types of spliced mRNA. The relatively low liver microsomal S-mephenytoin 4'-hydroxylase activities in some of the heterozygotes may simply reflect the selection process, which included samples with highly variable S-mephenytoin 4'-hydroxylase activities.

The genomic sequence of CYP2C19 is not currently known; hence, primers for the intron 4lexon 5 junction were developed empirically. This involved the use of multiple primers for intron 4 based on the sequence of this region in CYP2C9 (25), which is a closely related gene that shows 95% similarity to CYP2C19 in

A PCR cDNA

B Sample Number

M 4 5 7 9 10 11 13 17 20 21 24 27 35 m"l

re 400 - IL 200- 2 300-

--- I

a, v)

2 0.25 I

0 .-

2 0.5 0

FIG. 1. The splicing patterns of CYP2C19 mRNA in 13 selected human liver donor samples exhibiting a wide range of S-mephe- nytoin 4'-hydroxylase activities and CYP2C19 protein levels. A, diagram of strategy used to amplify CYP2C19 transcripts from human liver samples. This PCR strategy yielded a 284-bp band for the normal cDNA (wtlwt), a 244-bp band for the aberrant cDNA (rnlrn), and both bands with cDNA from heterozygous (wtlm) individuals. The hatched area indicates the 40-bp deletion in exon 5 of the aberrant cDNA. B , relation between genotype as assessed by reverse transcription PCR of human liver mRNA, hepatic content of CYP2C19 protein estimated by immunoblotting, liver microsomal S-mephenytoin 4'-hydroxylase ac- tivities, and the ratio of metabolism of the RIS enantiomers of mephe- nytoin in vitro. Arrows indicate the migration of recombinant CYP2C9 and CYP2C19 protein standards on the immunoblot (19).

the upstream region and several exons: and a specific reverse primer for exon 5 of CYP2C19. One primer pair amplified a DNA fragment with the correct predicted size in both EMS and PMs; however, only the fragment from EMS could be digested with SmaI. Sequencing of this fragment yielded sequence in- formation from which a specific forward primer was generated and used in subsequent PCR reactions to genotype individuals.

DNA from 28 unrelated Swiss and American Caucasian sub- jects whose phenotype had been established in vivo was ampli- fied using specific primers and restricted with SmaI using the strategy outlined in Fig. 2 A . Fig. 2C shows that 11 of 17 EMS were homozygous for the normal CYP2C19,,,, gene and 6 EMS were heterozygous. Among the 10 PMs, 7 were homozygous for

S. M. F. de Morais, J. Blaisdell, and J. A. Goldstein, unpublished data.

Page 3: The major genetic defect responsible for the polymorphism of S ...

Polymorphism in Mephenytoin Hydroxylation 1542 1

A PCR Genomic DNA

FIG. 2. Analysis of genomic DNA. A, diagram showing the strategy used to genotype genomic DNA from human blood. The strategy utilizes a PCR reac- tion followed by SmaI digestion. The pre- dicted sizes of digested DNA fragments are shown. R, diagram of genotypic tree of proband (arrow) and the gel of SmaI- digested PCR products. The genotype agrees with the previously published and indicated phenotype of Family C ( 5 ) . C, analysis of genomic DNA from selected Caucasian subjects from Switzerland and the United States. The phenotype ( E M , IM (intermediate phenotype), or P M ) is indicated by the brackets above the gel. D, analysis of genomic DNA from selected Japanese subjects. M represents the mo- lecular weight markers.

C

Sma I

m- - 120 169bp bP

2C19m e--- I Exon 5 1 Intron 4 Intron 5

* L I 169W '

""

3 - 49 bp

m LEGEND

M

200- - 169 im ' - 120im

50- - 49bp

- 200 - 100 - 50 49m-

D JAPANESE SAMPLES EM PM

- 200 - 100 - 50

the aberrant gene, 1 individual was heterozygous, and 2 were homozygous for the wild-type gene. The individual with the rare intermediate phenotype was also heterozygous. Thus, the mutant CYP2C19," accounted for 15 of the 20 alleles (75%) tested in Caucasian PMs.

Oriental populations have a much greater frequency of the mephenytoin PM phenotype compared to Caucasians (2, 3, 7). Accordingly, we analyzed DNA from 29 unrelated Japanese subjects (Fig. 2 0 ) . Eight of the 12 EMS were homozygous and 4 were heterozygous for CYP2C19,,,,. CYP2C19,, accounted for a similar percentage (74%) of the alleles (25 of 34) in Japanese PMs as found in Caucasian PMs. Ten of 17 PMs were homozy- gous for the mutant allele, and 5 were heterozygous. Thus, the major mutation responsible for the PM phenotype in Japanese is identical to that found in Caucasians. However, the defect

was not present in all PMs regardless of whether they were Caucasian or Japanese. It is therefore likely that additional mutations exist, which result in the PM phenotype in both populations. In a similar manner, a point mutation a t a splice site consensus sequence is the single most common mutation in CYP206, accounting for >75% of mutant alleles in PMs of de- brisoquine (26, 27). However, several minor mutant alleles have also been identified that contribute to this phenotype.

We also genotyped a Japanese family that had been studied previously with respect to the inheritance of the PM trait ( 5 ) (Fig. 2B) . There was complete concordance between the CYP2C19 genotype and the in vivo phenotype consistent with the previously reported Mendelian autosomal recessive mode of inheritance (5 , 6 ) .

DNA from individuals representative of the two CYP2C19

Page 4: The major genetic defect responsible for the polymorphism of S ...

15422 Polymorphism in Mephenytoin Hydroxylation

Aberrant (XlSm) ~ - - - ~ - - - ~ - - - " " ~ " - ~ "". I"fro"d"""- """"""""_" E*oNs-

wtce site

1 t a . f f t a a t . a a f f a t t g f t t t ~ t ~ t t ~ ~ t , t g ~ , * t ~ * t t t t ~ ~ ~ ~ ~ t ~ t ~ ~ t t g ~ t t . t t t ~ ~ ~ ~ ~ A A C C C A l A A C -"

Y Y Y Y Y Y Y Y Y Y Y n c a g G

FIG. 3. Partial sequence of the intron rYexon 6 junction of cYP2C19 in an EM and a PM, showing the correct splice site used in CYP2C19,, and the aberrant splice site used in cYP2C19,. Intron sequences are shown in lowercase and exon se- quences in uppercase letters. The nucleotides deleted in the aberrantly spliced cDNA are indicated in boldface type. The polymorphic SmaI site is underlined in CYP2C19,,. The highly conserved AG residues at the introdexon junction are shown in black bores. The consensus sequence (11YNCAGG) (Y, pyrimidine; N, any base) for the 3"splice site (29) is indicated below the normal and cryptic splice junctions.

genotypes was amplified as described above and then directly sequenced (Fig. 3). The sequence information verified that only CYP2C19 was amplified in the genotyping test. Surprisingly, the sequence of intron 4 of the defective gene was identical to that of the normal gene. The only difference was a G + A substitution in the coding sequence of exon 5 of CYP2C19, corresponding to position 681 in the cDNA. This change pro- duces a cryptic splice site in the exon, which shows a similar degree of homology with the mammalian 3"splice site consen- sus sequence (28) as the normal 3"splice site. There are also potential branch points near this cryptic splice site. The appar- ently complete selection of the cryptic site is somewhat surpris- ing, and the reasons for this are not clear. Comparison of the genotypes of liver samples 13 (extensive), 21 (intermediate), and 35 (poor) with their cDNA analysis patterns (Fig. 1) indi- cated complete agreement between the genotype and splicing pattern. Interestingly, cDNA from CYP2C8 and CYP2C18 have potential 3"splice sites at the same position in exon 5 (291, yet the full-length CYP2C8 protein is present in human liver mi- crosomes (19), indicating that this gene is spliced correctly. Moreover, we have amplified the exon 4exon 5 junction of the cDNA for CW2C18 and CYP2C8 and have seen no evidence for abnormal splicing (data not shown). However, homology to the mammalian consensus sequence for the introdexon junctions and branch points is not the sole determinant of splice site selection. For example, higher order RNA structure can also facilitate cleavage preferentially at a particular site (30) and could explain the preferential selection of the cryptic CYP2C19 splice site.

In conclusion, the present study identifies the primary ge- netic defect (CYP2C19,) that is responsible for the poor me- tabolism of mephenytoin and which also affects metabolism of several other widely used drugs (8-17). CYP2C19, accounts for 75% of the defective alleles in both Caucasian and Japanese PMs. The defect consists of a single base pair substitution in exon 5 of CYP2C19, which produces a cryptic 3"splice site, resulting in an aberrantly spliced mRNA and the absence of the

CYP2C19 protein in livers of PMs. We have developed a simple PCR-based genetic test for the defective CYP2C19, allele, which will be useful in clinical studies investigating the impor- tance of this genetic defect in drug metabolism in humans.

Acknowledgments-We thank Dr. Richard B. Kim and Dr. Diarmuid OShea (Division of Clinical Pharmacology, Vanderbilt University Medi- cal School) for phenotyping some of the individuals used in this study, Dr. Jerome Lasker (Mt. Sinai School of Medicine, New York, NY), for providing the polyclonal antibody to human CYP2C9, and Dr. Gordon Ibeanu (NIEHS) for helpful suggestions during this work.

REFERENCES 1. Daly, A. K., Cholerton, S., Gregory, W. & Idle, J. R. (1993) Pharmacol. Ther. 67,

3. Wedlund, P. J., Aslanian, W. S., McAllister, C. B., Wilkinson, G. R. & Branch, 2. Kiipfer, A. & Preisig, R. (1984) Eur. J. Clin. Pharmacol. 26, 753-759

4. Wilkinson, G. R., Guengerich, F. P. & Branch, R. A. (1989) Pharmacol. Ther. 43,

5. Ward, S. A,, Goto, F., Nakamura, K., Jacqz, E., Wilkinson, G. R. & Branch, R.

6. Inaba, T., Jurima, M. & Kalow, W. (1986)Am. J. Hum. Genet. 38,76%772 7. Nakamura, K. Goto, F., Ray, W. A., McAllister, C. B., Jacqz, E., Wilkinson, G.

8. Andersson, T., Regardh, C.-G., Lou, Y.-C., Zhang, Y., Dahl, M. L. & Bertilsson,

9. Ward, S. A., Helsby, N. A,, Skjelbo, E., Brpsen, K., Gram, L. F. & Breckenridge,

10. Adedoyin, A,, Prakash, C., Blair, J . A. & Wilkinson, G. R. (1994) Pharmacoge-

11. Kiipfer, A. & Branch, R. A. (1985) Clin. Pharmacol. Ther. 38, 41-18 12. Sindrup, S. H., Bwsen, K., Hansen, M. G. J., Aaes-Jorgensen, T., Overo, K. I?

13. Ward, S. A,, Walle, T., Walle, U. R, Wilkinson, G. R. & Branch, R. A. (1989)

14. Baumann, P., Jonzier-Perey, M., Kerb, L., Kiipfer, A,, Tingueley, D. & Schopf,

15. Nielson, K. K., Brgsen, K., Hansen, J. & Gram, L. F. (1991) Acta Physiol.

16. Skjelbo, E., Brmen, K., Hallas, J. & Gram, L. F. (1991) Clin. Pharmacol. Ther.

17. Bertilsson, L., Henthorn, T. K., Sanz, E., Tybring, G., Sawe, J. & VillBn, T. (1989) Clin. Pharmacol. Ther. 45, 34a355

18. Wrighton, S. A,, Stevens, J. C., Becker, G. W. & VandenBranden, M. (1993) Arch. Biochem. Biophys. 306,240-245

19. Goldstein, J. A,, Faletto, M. B., Romkes-Sparks, M., Sullivan, T., Kitareewan, S., Raucy, J. L., Lasker, J. M. & Ghanayem, B. I. (1994) Biochemistry 33,

20. Meier, U. T., Dayer, P., Male, P.-J., Kronbach, T. & Meyer, U. A. (1985) Clin. 1743-1752

Pharmacol. Ther. 38,48&i94

22. Chomczynski, P. & Sacchi, N. (1987) Anal. Biochem. 162, 156159 21. Meier, U. T., Kronbach, T. & Meyer, U. A. (1985)Anal. Biochem. 161,28l%291

23. A r n s , P. A,, Ryder, D., White, L., Wilkinson, G. R. & Branch, R. A. (1990)

24. Sambrook. J., Fritsch, E. F. & Maniatis, T. (1989) Molecular Cloning: A Lab0 Pharmacologist 32, 140

ratory Manual, 2nd Ed., pp. 9.1G9.18, Cold Spring Harbor Laboratory, Cold

129-160

R. A. (1984) Clin. Pharrnacol. Ther. 36,773-780

53-76

A. (1987) Clin. Pharmacol. Ther. 42,9G99

R. & Branch, R. A. (1985) Clin. Pharmacol. Ther. 38,402408

L. (1992) Pharmacogenetics 2,25-31

A. M. (1991) Br. J. Clin. Pharmacol. 31,689492

netics 4, 27-38

& Gram, L. F. (1993) Ther. Drug Monit. 13, 11-17

Clin. Pharmacol. Ther. 45, 72-79

J . (1986) Int. Clin. Psychopharmacol. 1, 102-112

Scand. 14, Suppl. 608, 172

49.18-23

25. de Morais, S. M. F., Schweikl, H., Blaisdell, J . & Goldstein, J. A. (1993) Bio- Spring Harbor, NY

chem. Bioohvs. Res. Commun. 194.194-201 26. Gonzalez, F.'~J:& Meyer, U. A. (1991) Clin. Pharmacol. Ther. KO, 233-238

27. Broly, F., Gaedigk, A,, Heim, M., Eichelbaum, M., Morike, K. & Meyer, U. A.

28. Green, M. R. (1991) Annu. Reu. Cell. B i d . 7, 559-599 29. Romkes, M., Faletto, M. B., Blaisdell, J. A., Raucy, J. L. & Goldstein, J. A.

30. Estes, P. A,, Cooke, N. E. & Liebhaber, S. A. (1992) J. Bid . Chem. 267,14902-

.~~ ~ ~

(1991)

(1991) DNA Cell Biol. 10, 545-558

(1991) Biochemistry 30,3247-3255

14908