Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
Pharmacogenomics J ; 17(2): 137-145, 2017 03.
Article in English | MEDLINE | ID: mdl-26856248

ABSTRACT

Variation in the expression level and activity of genes involved in drug disposition and action ('pharmacogenes') can affect drug response and toxicity, especially when in tissues of pharmacological importance. Previous studies have relied primarily on microarrays to understand gene expression differences, or have focused on a single tissue or small number of samples. The goal of this study was to use RNA-sequencing (RNA-seq) to determine the expression levels and alternative splicing of 389 Pharmacogenomics Research Network pharmacogenes across four tissues (liver, kidney, heart and adipose) and lymphoblastoid cell lines, which are used widely in pharmacogenomics studies. Analysis of RNA-seq data from 139 different individuals across the 5 tissues (20-45 individuals per tissue type) revealed substantial variation in both expression levels and splicing across samples and tissue types. Comparison with GTEx data yielded a consistent picture. This in-depth exploration also revealed 183 splicing events in pharmacogenes that were previously not annotated. Overall, this study serves as a rich resource for the research community to inform biomarker and drug discovery and use.


Subject(s)
Alternative Splicing , Computational Biology , High-Throughput Nucleotide Sequencing , Pharmacogenetics , Pharmacogenomic Variants , Sequence Analysis, RNA , Transcriptome , Adipose Tissue/metabolism , Cell Line , Databases, Genetic , Genotype , Humans , Kidney/metabolism , Liver/metabolism , Myocardium/metabolism , Phenotype
2.
Pharmacogenomics J ; 13(2): 110-20, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22231567

ABSTRACT

Human organic cation transporter 3 (OCT3 and SLC22A3) mediates the uptake of many important endogenous amines and basic drugs in a variety of tissues. OCT3 is identified as one of the important risk loci for prostate cancer, and is markedly underexpressed in aggressive prostate cancers. The goal of this study was to identify genetic and epigenetic factors in the promoter region that influence the expression level of OCT3. Haplotypes that contained the common variants, g.-81G>delGA (rs60515630) (minor allele frequency 11.5% in African American) and g.-2G>A (rs555754) (minor allele frequency>30% in all ethnic groups) showed significant increases in luciferase reporter activities and exhibited stronger transcription factor-binding affinity than the haplotypes that contained the major alleles. Consistent with the reporter assays, OCT3 messenger RNA expression levels were significantly higher in Asian (P<0.001) and Caucasian (P<0.05) liver samples from individuals who were homozygous for g.-2A/A in comparison with those homozygous for the g.-2G/G allele. Studies revealed that the methylation level in the basal promoter region of OCT3 was associated with OCT3 expression level and tumorigenesis capability in various prostate cancer cell lines. The methylation level of the OCT3 promoter was higher in 62% of prostate tumor samples compared with matched normal samples. Our studies demonstrate that genetic polymorphisms in the proximal promoter region of OCT3 alter the transcription rate of the gene and may be associated with altered expression levels of OCT3 in human liver. Aberrant methylation contributes to the reduced expression of OCT3 in prostate cancer.


Subject(s)
DNA Methylation/genetics , Epigenomics , Organic Cation Transport Proteins/genetics , Prostatic Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Pharmacological/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic , Ethnicity/genetics , Female , Gene Expression Regulation , Gene Frequency , Haplotypes , Humans , Male , Middle Aged , Polymorphism, Genetic , Promoter Regions, Genetic , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology
3.
Clin Pharmacol Ther ; 90(5): 674-84, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21956618

ABSTRACT

Multidrug and toxin extrusion 2 (MATE2-K (SLC47A2)), a polyspecific organic cation exporter, facilitates the renal elimination of the antidiabetes drug metformin. In this study, we characterized genetic variants of MATE2-K, determined their association with metformin response, and elucidated their impact by means of a comparative protein structure model. Four nonsynonymous variants and four variants in the MATE2-K basal promoter region were identified from ethnically diverse populations. Two nonsynonymous variants-c.485C>T and c.1177G>A-were shown to be associated with significantly lower metformin uptake and reduction in protein expression levels. MATE2-K basal promoter haplotypes containing the most common variant, g.-130G>A (>26% allele frequency), were associated with a significant increase in luciferase activities and reduced binding to the transcriptional repressor myeloid zinc finger 1 (MZF-1). Patients with diabetes who were homozygous for g.-130A had a significantly poorer response to metformin treatment, assessed as relative change in glycated hemoglobin (HbA1c) (-0.027 (-0.076, 0.033)), as compared with carriers of the reference allele, g.-130G (-0.15 (-0.17, -0.13)) (P=0.002). Our study showed that MATE2-K plays a role in the antidiabetes response to metformin.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacokinetics , Metformin/pharmacokinetics , Organic Cation Transport Proteins/genetics , Adult , Aged , Alleles , Animals , Female , Genetic Variation , Glycated Hemoglobin/metabolism , HCT116 Cells , HEK293 Cells , Haplotypes , Humans , Hypoglycemic Agents/pharmacology , LLC-PK1 Cells , Luciferases/metabolism , Male , Metformin/pharmacology , Middle Aged , Polymorphism, Genetic , Promoter Regions, Genetic , Racial Groups/genetics , Retrospective Studies , Swine , Treatment Outcome
4.
Clin Pharmacol Ther ; 89(4): 571-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21368754

ABSTRACT

Little is known about how genetic variations in enhancers influence drug response. In this study, we investigated whether nucleotide variations in enhancers that regulate drug transporters can alter their expression levels. Using comparative genomics and liver-specific transcription factor binding site (TFBS) analyses, we identified evolutionary conserved regions (ECRs) surrounding nine liver membrane transporters that interact with commonly used pharmaceuticals. The top 50 ECRs were screened for enhancer activity in vivo, of which five--located around ABCB11, SLC10A1, SLCO1B1, SLCO1A2, and SLC47A1--exhibited significant enhancer activity. Common variants identified in a large ethnically diverse cohort (n = 272) were assayed for differential enhancer activity, and three variants were found to have significant effects on reporter activity as compared with the reference allele. In addition, one variant was associated with reduced SLCO1A2 mRNA expression levels in human liver tissues, and another was associated with increased methotrexate (MTX) clearance in patients. This work provides a general model for the rapid characterization of liver enhancers and identifies associations between enhancer variants and drug response.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Methotrexate/pharmacokinetics , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/metabolism , Pharmaceutical Preparations/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/genetics , Alleles , Animals , Binding Sites , Biological Transport , Conserved Sequence , Female , Gene Expression Regulation , Genetic Variation , Genomics/methods , Humans , Liver/metabolism , Male , Mice , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/genetics , Polymorphism, Single Nucleotide , RNA, Messenger/metabolism , Racial Groups/genetics , Transcription Factors
5.
Pharmacogenomics J ; 5(3): 157-65, 2005.
Article in English | MEDLINE | ID: mdl-15738947

ABSTRACT

The human concentrative nucleoside transporter, CNT3 (SLC28A3), plays an important role in mediating the cellular entry of a broad array of physiological nucleosides and synthetic anticancer nucleoside analog drugs. As a first step toward understanding the genetic basis for interindividual differences in the disposition and response to antileukemic nucleoside analogs, we examined the genetic and functional diversity of CNT3. In all, 56 variable sites in the exons and flanking intronic region of SLC28A3 were identified in a collection of 270 DNA samples from US populations (80 African-Americans, 80 European-Americans, 60 Asian-Americans, and 50 Mexican-Americans). Of the 16 coding region variants, 12 had not been previously reported. Also, 10 resulted in amino-acid changes and three of these had total allele frequencies of >/=1%. Nucleotide diversity (pi) at nonsynonymous and synonymous sites was estimated to be 1.81 x 10(4) and 18.13 x 10(4), respectively, suggesting that SLC28A3 is under negative selection. All nonsynonymous variants, constructed by site-directed mutagenesis and expressed in Xenopus laevis oocytes, transported purine and pyrimidine model substrates, except for c. 1099G>A (p. Gly367Arg). This rare variant alters an evolutionarily conserved site in the putative substrate recognition domain of CNT3. The presence of three additional evolutionarily conserved glycine residues in the vicinity of p. Gly367Arg that are also conserved in human paralogs suggest that these glycine residues are critical in the function of the concentrative nucleoside transporter family. The genetic analysis and functional characterization of CNT3 variants suggest that this transporter does not tolerate nonsynonymous changes and is important for human fitness.


Subject(s)
Membrane Transport Proteins/genetics , Vidarabine/analogs & derivatives , Adenosine/metabolism , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Cation Transport Proteins , Cladribine/metabolism , Conserved Sequence , DNA/genetics , Ethnicity , Genetic Variation , Haplotypes , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Oocytes/metabolism , Vidarabine/metabolism , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL