Pharmacogenomics genes show varying perceptibility to microRNA regulation

Objective The aim of pharmacogenomics is to identify individual differences in genome and transcriptome composition and their effect on drug efficacy. MicroRNAs (miRNAs) are short noncoding RNAs that negatively regulate expression of the majority of animal genes, including many genes involved in drug efficacy. Consequently, differences in the miRNA expression among individuals could be an important factor contributing to differential drug response. Pharmacogenomics genes can be divided into drug target genes termed as pharmacodynamics genes (PD) and genes involved in drug transport and metabolism termed as pharmacokinetics genes (PK). To clarify the regulatory potential of miRNAs in pharmacogenomics, we have examined the potential regulation by miRNAs of PK and PD genes. Methods We identified PK and PD genes as annotated by the Pharmacogenomics Knowledge Base and examined miRNA targeting of genes in the two groups according to several miRNA target prediction databases. We furthermore studied how differences between the two groups are reflected in the gene structure and across gene families. Lastly, we studied changes in expression levels of PK versus PD genes in cells depleted for miRNAs by shRNA-mediated knockdown of the miRNA-processing enzyme Dicer. Results Our analysis identify a striking difference in the level of miRNA regulation between PK and PD genes, with the former having less than half predicted conserved miRNA binding sites compared with the latter. Importantly, this finding is reflected in a highly significant difference in the shift in expression levels of PD versus PK genes after depletion of miRNAs. Conclusion Our study emphasizes an intrinsic difference between PK and PD genes and helps clarify the role of miRNAs in pharmacogenomics.

[1]  Noam Shomron,et al.  MicroRNAs and pharmacogenomics. , 2010, Pharmacogenomics.

[2]  Yasuhiro Aoki,et al.  Quantitative Analysis of UDP-Glucuronosyltransferase (UGT) 1A and UGT2B Expression Levels in Human Livers , 2009, Drug Metabolism and Disposition.

[3]  L. Lim,et al.  Transcripts Targeted by the MicroRNA-16 Family Cooperatively Regulate Cell Cycle Progression , 2007, Molecular and Cellular Biology.

[4]  S. Brenner,et al.  Unproductive splicing of SR genes associated with highly conserved and ultraconserved DNA elements , 2007, Nature.

[5]  J. Kim,et al.  Inhibition of cell cycle progression via p27Kip1 upregulation and apoptosis induction by an ethanol extract of Rhus verniciflua Stokes in AGS gastric cancer cells. , 2006, International journal of molecular medicine.

[6]  H. Willard,et al.  Genomic and personalized medicine: foundations and applications. , 2009, Translational research : the journal of laboratory and clinical medicine.

[7]  H. Sültmann,et al.  The human let-7a-3 locus contains an epigenetically regulated microRNA gene with oncogenic function. , 2007, Cancer research.

[8]  J. Fei,et al.  Anti‐miR‐21 oligonucleotide sensitizes leukemic K562 cells to arsenic trioxide by inducing apoptosis , 2010, Cancer science.

[9]  Chunja Lee,et al.  The human cytochrome P450 1A1 mRNA is rapidly degraded in HepG2 cells. , 2000, Archives of biochemistry and biophysics.

[10]  A. Daly,et al.  Genome-wide association studies in pharmacogenomics , 2010, Nature Reviews Genetics.

[11]  Michael Kertesz,et al.  The role of site accessibility in microRNA target recognition , 2007, Nature Genetics.

[12]  M. Niemi,et al.  Membrane transporters in drug development , 2010, Nature Reviews Drug Discovery.

[13]  F. Slack,et al.  Small non-coding RNAs in animal development , 2008, Nature Reviews Molecular Cell Biology.

[14]  D. Begley,et al.  ABC transporters and the blood-brain barrier. , 2004, Current pharmaceutical design.

[15]  Mark Gerstein,et al.  The relationship between the evolution of microRNA targets and the length of their UTRs , 2009, BMC Genomics.

[16]  H. Horvitz,et al.  MicroRNA expression profiles classify human cancers , 2005, Nature.

[17]  H. Yamada,et al.  Changes of micro-RNA expression in rat liver treated by acetaminophen or carbon tetrachloride--regulating role of micro-RNA for RNA expression. , 2007, The Journal of toxicological sciences.

[18]  Brad T. Sherman,et al.  DAVID: Database for Annotation, Visualization, and Integrated Discovery , 2003, Genome Biology.

[19]  Joshua M. Stuart,et al.  Integrating genotype and phenotype information: an overview of the PharmGKB project , 2001, The Pharmacogenomics Journal.

[20]  K. Bock Functions and transcriptional regulation of adult human hepatic UDP-glucuronosyl-transferases (UGTs): mechanisms responsible for interindividual variation of UGT levels. , 2010, Biochemical pharmacology.

[21]  F. Slack,et al.  OncomiR addiction in an in vivo model of microRNA-21-induced pre-B-cell lymphoma , 2010, Nature.

[22]  C. Sander,et al.  A Mammalian microRNA Expression Atlas Based on Small RNA Library Sequencing , 2007, Cell.

[23]  Doron Betel,et al.  The microRNA.org resource: targets and expression , 2007, Nucleic Acids Res..

[24]  J. Schellens,et al.  Increased oral bioavailability of paclitaxel by GF120918 in mice through selective modulation of P-glycoprotein. , 2000, Clinical cancer research : an official journal of the American Association for Cancer Research.

[25]  R. Russell,et al.  Animal MicroRNAs Confer Robustness to Gene Expression and Have a Significant Impact on 3′UTR Evolution , 2005, Cell.

[26]  Daniel J. Blankenberg,et al.  A framework for collaborative analysis of ENCODE data: making large-scale analyses biologist-friendly. , 2007, Genome research.

[27]  Ulrich Pfeffer,et al.  Modulation of microRNA expression by budesonide, phenethyl isothiocyanate and cigarette smoke in mouse liver and lung. , 2010, Carcinogenesis.

[28]  S. Kauppinen,et al.  Therapeutic Silencing of MicroRNA-122 in Primates with Chronic Hepatitis C Virus Infection , 2010, Science.

[29]  Chunxiang Zhang,et al.  MicroRNA-21 in Cardiovascular Disease , 2010, Journal of cardiovascular translational research.

[30]  Mihaela Zavolan,et al.  Effects of Dicer and Argonaute down-regulation on mRNA levels in human HEK293 cells , 2006, Nucleic acids research.

[31]  M. Scally,et al.  The chemical defensome: environmental sensing and response genes in the Strongylocentrotus purpuratus genome. , 2006, Developmental biology.

[32]  Ranit Aharonov,et al.  MicroRNA expression detected by oligonucleotide microarrays: system establishment and expression profiling in human tissues. , 2004, Genome research.

[33]  C. Croce,et al.  MicroRNA signatures in human cancers , 2006, Nature Reviews Cancer.

[34]  O. Kovalchuk,et al.  Induction of microRNAome deregulation in rat liver by long-term tamoxifen exposure. , 2007, Mutation research.

[35]  Brad T. Sherman,et al.  Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources , 2008, Nature Protocols.

[36]  Shingo Takagi,et al.  Human CYP24 Catalyzing the Inactivation of Calcitriol Is Post-Transcriptionally Regulated by miR-125b , 2009, Molecular Pharmacology.

[37]  C. Burge,et al.  Most mammalian mRNAs are conserved targets of microRNAs. , 2008, Genome research.

[38]  A. Patiño-García,et al.  Clinical involvement of catechol-O-methyltransferase polymorphisms in schizophrenia spectrum disorders: influence on the severity of psychotic symptoms and on the response to neuroleptic treatment , 2007, The Pharmacogenomics Journal.

[39]  M. Hirata,et al.  ABCB1 and ABCC1 expression in peripheral mononuclear cells is influenced by gene polymorphisms and atorvastatin treatment. , 2009, Biochemical pharmacology.

[40]  M. Egan,et al.  Catechol-O-methyltransferase val108/158met genotype predicts working memory response to antipsychotic medications , 2004, Biological Psychiatry.

[41]  Hongbing Wang,et al.  Relative Activation of Human Pregnane X Receptor versus Constitutive Androstane Receptor Defines Distinct Classes of CYP2B6 and CYP3A4 Inducers , 2007, Journal of Pharmacology and Experimental Therapeutics.

[42]  J. Beijnen,et al.  Limited oral bioavailability and active epithelial excretion of paclitaxel (Taxol) caused by P-glycoprotein in the intestine. , 1997, Proceedings of the National Academy of Sciences of the United States of America.

[43]  Daniel J. Blankenberg,et al.  Galaxy: A Web‐Based Genome Analysis Tool for Experimentalists , 2010, Current protocols in molecular biology.

[44]  Albert J. Vilella,et al.  EnsemblCompara GeneTrees: Complete, duplication-aware phylogenetic trees in vertebrates. , 2009, Genome research.

[45]  D. Bartel MicroRNAs Genomics, Biogenesis, Mechanism, and Function , 2004, Cell.

[46]  Anjali J. Koppal,et al.  Supplementary data: Comprehensive modeling of microRNA targets predicts functional non-conserved and non-canonical sites , 2010 .

[47]  N. Hewitt,et al.  Induction of hepatic cytochrome P450 enzymes: methods, mechanisms, recommendations, and in vitro–in vivo correlations , 2007, Xenobiotica; the fate of foreign compounds in biological systems.

[48]  P. Provost,et al.  MicroRNAs as a molecular basis for mental retardation, Alzheimer's and prion diseases , 2010, Brain Research.

[49]  C. Burge,et al.  Conserved Seed Pairing, Often Flanked by Adenosines, Indicates that Thousands of Human Genes are MicroRNA Targets , 2005, Cell.

[50]  R. Weinshilboum,et al.  Platelet phenol sulfotransferase and erythrocyte catechol‐O‐methytransferase activities: Correlation with methyldopa metabolism , 1984, Clinical pharmacology and therapeutics.

[51]  Richard M. Weinshilboum,et al.  Pharmacogenomics: Catechol O-Methyltransferase to Thiopurine S-Methyltransferase , 2006, Cellular and Molecular Neurobiology.

[52]  John E. Le Lay,et al.  Hepatic function is preserved in the absence of mature microRNAs , 2009, Hepatology.

[53]  L. Rivera-calimlim,et al.  Catechol‐O‐methyltransferase activity: A determinant of levodopa response , 1980, Clinical pharmacology and therapeutics.