Top Three Pharmacogenomics and Personalized Medicine Applications at the Nexus of Renal Pathophysiology and Cardiovascular Medicine

Murielle Bochud, Michel Burnier, Idris Guessous, Murielle Bochud, Michel Burnier, Idris Guessous

Abstract

Pharmacogenomics is a field with origins in the study of monogenic variations in drug metabolism in the 1950s. Perhaps because of these historical underpinnings, there has been an intensive investigation of 'hepatic pharmacogenes' such as CYP450s and liver drug metabolism using pharmacogenomics approaches over the past five decades. Surprisingly, kidney pathophysiology, attendant diseases and treatment outcomes have been vastly under-studied and under-theorized despite their central importance in maintenance of health, susceptibility to disease and rational personalized therapeutics. Indeed, chronic kidney disease (CKD) represents an increasing public health burden worldwide, both in developed and developing countries. Patients with CKD suffer from high cardiovascular morbidity and mortality, which is mainly attributable to cardiovascular events before reaching end-stage renal disease. In this paper, we focus our analyses on renal function before end-stage renal disease, as seen through the lens of pharmacogenomics and human genomic variation. We herein synthesize the recent evidence linking selected Very Important Pharmacogenes (VIP) to renal function, blood pressure and salt-sensitivity in humans, and ways in which these insights might inform rational personalized therapeutics. Notably, we highlight and present the rationale for three applications that we consider as important and actionable therapeutic and preventive focus areas in renal pharmacogenomics: 1) ACE inhibitors, as a confirmed application, 2) VDR agonists, as a promising application, and 3) moderate dietary salt intake, as a suggested novel application. Additionally, we emphasize the putative contributions of gene-environment interactions, discuss the implications of these findings to treat and prevent hypertension and CKD. Finally, we conclude with a strategic agenda and vision required to accelerate advances in this under-studied field of renal pharmacogenomics with vast significance for global public health.

Figures

Fig. (1)
Fig. (1)
Top three pharmacogenomics applications at the nexus of renal pathophysiology and cardiovascular medicine, namely ACE inhibitor treatment, VDR antagonists and moderate dietary salt intake. Arrows represent the direct links as described in the scientific literature. The light blue band across PXR, CAR, VDR and AHR illustrates the tight interplay that exists among these genes. VIP = very important pharmacogenes; SS = genes that are involved in the blood pressure response to salt intake with various levels of evidence.

References

    1. Levey AS, Atkins R, Coresh J, et al. Chronic kidney disease as a global public health problem: approaches and initiatives - a position statement from Kidney Disease Improving Global Outcomes. Kidney Int. 2007;72:247–59.
    1. Wen CP, Cheng TY, Tsai MK, et al. All-cause mortality attributable to chronic kidney disease: a prospective cohort study based on 462 293 adults in Taiwan. Lancet. 2008;371:2173–82.
    1. Hallan SI, Coresh J, Astor BC, et al. International comparison of the relationship of chronic kidney disease prevalence and ESRD risk. J Am Soc Nephrol. 2006;17:2275–84.
    1. Coresh J, Selvin E, Stevens LA, et al. Prevalence of chronic kidney disease in the United States. JAMA. 2007;298:2038–47.
    1. Chadban SJ, Briganti EM, Kerr PG, et al. Prevalence of kidney damage in Australian adults: The AusDiab kidney study. J Am Soc Nephrol. 2003;14:S131–8.
    1. Najafi I, Attari F, Islami F, et al. Renal function and risk factors of moderate to severe chronic kidney disease in Golestan Province, northeast of Iran. PLoS One. 2010;5:e14216.
    1. Chen W, Wang H, Dong X, et al. Prevalence and risk factors associated with chronic kidney disease in an adult population from southern China. Nephrol Dial Transplant. 2009;24:1205–12.
    1. Zhang L, Zhang P, Wang F, et al. Prevalence and factors associated with CKD: a population study from Beijing. Am J Kidney Dis. 2008;51:373–84.
    1. Agarwal SK, Srivastava RK. Chronic kidney disease in India: challenges and solutions. Nephron Clin Pract . 2009;111:c197–203. discussion c203.
    1. Muntner P, Bowling CB, Gao L, et al. Age-Specific Association of Reduced Estimated Glomerular Filtration Rate and Albuminuria with All-Cause Mortality. Clin J Am Soc Nephrol. 2011;6:2200–7.
    1. Matsushita K, van der Velde M, Astor BC, et al. Association of estimated glomerular filtration rate and albuminuria with all-cause and cardiovascular mortality in general population cohorts: a collaborative meta-analysis. Lancet. 2010;375:2073–81.
    1. Keith DS, Nichols GA, Gullion CM, et al. Longitudinal follow- up and outcomes among a population with chronic kidney disease in a large managed care organization. Arch Intern Med. 2004;164:659–63.
    1. Dalrymple LS, Katz R, Kestenbaum B, et al. Chronic kidney disease and the risk of end-stage renal disease versus death. J Gen Intern Med. 2011;26:379–85.
    1. Peralta CA, Shlipak MG, Fan D, et al. Risks for end-stage renal disease, cardiovascular events, and death in Hispanic versus non-Hispanic white adults with chronic kidney disease. J Am Soc Nephrol. 2006;17:2892–9.
    1. Go AS, Chertow GM, Fan D, et al. Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. N Engl J Med. 2004;351:1296–305.
    1. Quteineh L, Verstuyft C. Pharmacogenetics in immuno- suppressants: impact on dose requirement of calcineurin inhibitors in renal and liver pediatric transplant recipients. Curr Opin Organ Transplant. 2010;15:601–7.
    1. Herrero MJ, Sanchez-Plumed J, Galiana M, et al. Influence of pharmacogenetic polymorphisms in routine immunosuppression therapy after renal transplantation. Transplant Proc. 2010;42:3134–6.
    1. Hesselink DA, Bouamar R, van Gelder T. The pharmacogenetics of calcineurin inhibitor-related nephrotoxicity. Ther Drug Monit. 2010;32:387–93.
    1. Capron A, Mourad M, De Meyer M, et al. CYP3A5 and ABCB1 polymorphisms influence tacrolimus concentrations in peripheral blood mononuclear cells after renal transplantation. Pharmacogenomics. 2010;11:703–14.
    1. Thervet E, Loriot MA, Barbier S, et al. Optimization of initial tacrolimus dose using pharmacogenetic testing. Clin Pharmacol Ther. 2010;87:721–6.
    1. Wang L, McLeod HL, Weinshilboum RM. Genomics and drug response. N Engl J Med. 2011;364:1144–53.
    1. Rahmioglu N, Heaton J, Clement G, et al. Genetic epidemiology of induced CYP3A4 activity. Pharmacogenet Genomics. 2011;21:642–51.
    1. Birkenfeld AL, Jordan J, Hofmann U, et al. Genetic influences on the pharmacokinetics of orally and intravenously administered digoxin as exhibited by monozygotic twins. Clin Pharmacol Ther. 2009;86:605–8.
    1. Jiang Z, Dragin N, Jorge-Nebert LF, et al. Search for an association between the human CYP1A2 genotype and CYP1A2 metabolic phenotype. Pharmacogenet Genomics. 2006;16:359–67.
    1. Kuehl P, Zhang J, Lin Y, et al. Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nat Genet. 2001;27:383–91.
    1. Klein TE, Chang JT, Cho MK, et al. Integrating genotype and phenotype information: an overview of the PharmGKB project. Pharmacogenetics Research Network and Knowledge Base. The pharmacogenomics journal. 2001;1:167–70.
    1. Pharmacogenomics Knowledge Database. Available from: . 2011. [Accessed July 22, 2011].
    1. Bochud M, Eap CB, Elston RC, et al. Association of CYP3A5 genotypes with blood pressure and renal function in African families. J Hypertens. 2006;24:923–9.
    1. Bochud M, Bovet P, Burnier M, et al. CYP3A5 and ABCB1 genes and hypertension. Pharmacogenomics. 2009;10:477–87.
    1. Eap CB, Bochud M, Elston RC, et al. CYP3A5 and ABCB1 genes influence blood pressure and response to treatment, and their effect is modified by salt. Hypertension. 2007;49:1007–14.
    1. Bochud M, Eap CB, Maillard M, et al. Association of ABCB1 genetic variants with renal function in Africans and in Caucasians. BMC Med Genom. 2008;1:21.
    1. Ehret GB, Munroe PB, Rice KM, et al. Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk. Nature. 2011;478:103–9.
    1. Bochud M, Bovet P, Elston RC, et al. High heritability of ambulatory blood pressure in families of East African descent. Hypertension. 2005;45:445–50.
    1. Sanada H, Jones JE, Jose PA. Genetics of salt-sensitive hypertension. Curr Hypertens Rep. 2011;13:55–66.
    1. Zhou SF, Wang B, Yang LP, et al. Structure, function, regulation and polymorphism and the clinical significance of human cytochrome P450 1A2. Drug Metab Rev. 2010;42:268–354.
    1. Landi MT, Sinha R, Lang NP, et al. Human cytochrome P4501A2. IARC Sci Publ. 1999. pp. 173–95.
    1. Aitchison KJ, Gonzalez FJ, Quattrochi LC, et al. Identification of novel polymorphisms in the 5' flanking region of CYP1A2, characterization of interethnic variability, and investigation of their functional significance. Pharmacogenetics. 2000;10:695–704.
    1. Gunes A, Dahl ML. Variation in CYP1A2 activity and its clinical implications: influence of environmental factors and genetic polymorphisms. Pharmacogenomics. 2008;9:625–37.
    1. Nakajima M, Yokoi T, Mizutani M, et al. Genetic polymorphism in the 5'-flanking region of human CYP1A2 gene: effect on the CYP1A2 inducibility in humans. J Biochem. 1999;125:803–8.
    1. Sachse C, Brockmoller J, Bauer S, et al. Functional significance of a C-->A polymorphism in intron 1 of the cytochrome P450 CYP1A2 gene tested with caffeine. Br J Clin Pharmacol. 1999;47:445–9.
    1. Leblond F, Guevin C, Demers C, et al. Downregulation of hepatic cytochrome P450 in chronic renal failure. J Am Soc Nephrol. 2001;12:326–32.
    1. Dani M, Boisvert C, Michaud J, et al. Down-regulation of liver drug-metabolizing enzymes in a murine model of chronic renal failure. Drug Metab Dispos. 2010;38:357–60.
    1. Kottgen A, Glazer NL, Dehghan A, et al. Multiple loci associated with indices of renal function and chronic kidney disease. Nat Genet. 2009;41:712–7.
    1. Boger CA, Chen MH, Tin A, et al. CUBN is a gene locus for albuminuria. J Am Soc Nephrol. 2011;22:555–70.
    1. Nawrot TS, Staessen JA, Roels HA, et al. Cadmium exposure in the population: from health risks to strategies of prevention. Biometals. 2010;23:769–82.
    1. Boger CA, Heid IM. Chronic kidney disease: novel insights from genome-wide association studies. Kidney & blood pressure research. 2011;34:225–34.
    1. Newton-Cheh C, Johnson T, Gateva V, et al. Genome-wide association study identifies eight loci associated with blood pressure. Nat Genet. 2009;41:666–76.
    1. Levy D, Ehret GB, Rice K, et al. Genome-wide association study of blood pressure and hypertension. Nat Genet. 2009;41:677–87.
    1. Fox ER, Young JH, Li Y, et al. Association of genetic variation with systolic and diastolic blood pressure among African Americans: the Candidate Gene Association Resource study. Hum Mol Genet. 2011;20:2273–84.
    1. Zhou TB, Qin YH, Su LN, et al. Relationship between Angiotensin-Converting Enzyme Insertion/Deletion Gene Polymorphism and Susceptibility of Minimal Change Nephrotic Syndrome: A Meta-Analysis. Int J Nephrol. 2011;2011:360357.
    1. Kato N, Takeuchi F, Tabara Y, et al. Meta-analysis of genome-wide association studies identifies common variants associated with blood pressure variation in east Asians. Nat Genet. 2011;43:531–8.
    1. Liu C, Li H, Qi Q, et al. Common variants in or near FGF5, CYP17A1 and MTHFR genes are associated with blood pressure and hypertension in Chinese Hans. J Hypertens. 2011;29:70–5.
    1. Zhou SF, Liu JP, Chowbay B. Polymorphism of human cyto- chrome P450 enzymes and its clinical impact. Drug Metab Rev. 2009;41:89–295.
    1. Ingelman-Sundberg M, Sim SC, Gomez A, et al. Influence of cytochrome P450 polymorphisms on drug therapies: pharma- cogenetic, pharmacoepigenetic and clinical aspects. Pharmacol Ther. 2007;116:496–526.
    1. Hallberg P, Karlsson J, Kurland L, et al. The CYP2C9 genotype predicts the blood pressure response to irbesartan: results from the Swedish Irbesartan Left Ventricular Hypertrophy Investigation vs Atenolol (SILVHIA) trial. J Hypertens. 2002;20:2089–93.
    1. Cornelis MC, El-Sohemy A, Kabagambe EK, et al. Coffee, CYP1A2 genotype, and risk of myocardial infarction. JAMA. 2006;295:1135–41.
    1. Cornelis MC, Monda KL, Yu K, et al. Genome-wide meta-analysis identifies regions on 7p21 (AHR) and 15q24 (CYP1A2) as determinants of habitual caffeine consumption. PLoS Genet. 2011;7:e1002033.
    1. Frary CD, Johnson RK, Wang MQ. Food sources and intakes of caffeine in the diets of persons in the United States. J Am Diet Assoc. 2005;105:110–3.
    1. Higdon JV, Frei B. Coffee and health: a review of recent human research. Crit Rev Food Sci Nutr. 2006;46:101–23.
    1. Noordzij M, Uiterwaal CS, Arends LR, et al. Blood pressure response to chronic intake of coffee and caffeine: a meta-analysis of randomized controlled trials. J Hypertens. 2005;23:921–8.
    1. Klag MJ, Wang NY, Meoni LA, et al. Coffee intake and risk of hypertension: the Johns Hopkins precursors study. Arch Intern Med. 2002;162:657–62.
    1. Winkelmayer WC, Stampfer MJ, Willett WC, et al. Habitual caffeine intake and the risk of hypertension in women. JAMA. 2005;294:2330–5.
    1. Liu YT, Hao HP, Liu CX, et al. Drugs as CYP3A probes, inducers, and inhibitors. Drug Metab Rev. 2007;39:699–721.
    1. Daly AK. Significance of the minor cytochrome P450 3A isoforms. Clin Pharmacokinet. 2006;45:13–31.
    1. Wilkinson GR. Drug metabolism and variability among patients in drug response. New Engl J Med. 2005;352:2211–21.
    1. Xi B, Wang C, Liu L, et al. Association of the CYP3A5 polymorphism (6986G>A) with blood pressure and hypertension. Hypertens Res. 2011. [Epub ahead of print]
    1. Givens RC, Lin YS, Dowling AL, et al. CYP3A5 genotype predicts renal CYP3A activity and blood pressure in healthy adults. J Appl Physiol. 2003;95:1297–300.
    1. Chen X, Wang H, Zhou G, et al. Molecular population genetics of human CYP3A locus: signatures of positive selection and implications for evolutionary environmental medicine. Environment Health Persp. 2009;117:1541–8.
    1. Thompson EE, Kuttab-Boulos H, Witonsky D, et al. CYP3A variation and the evolution of salt-sensitivity variants. Am J Hum Genet. 2004;75:1059–69.
    1. Schuetz EG, Schuetz JD, Grogan WM, et al. Expression of cytochrome P450 3A in amphibian, rat, and human kidney. Arch Biochem Biophys. 1992;294:206–14.
    1. Chowbay B, Zhou S, Lee EJ. An interethnic comparison of polymorphisms of the genes encoding drug-metabolizing enzymes and drug transporters: experience in Singapore. Drug Metab Rev. 2005;37:327–78.
    1. Ho H, Pinto A, Hall SD, et al. Association between the CYP3A5 genotype and blood pressure. Hypertension. 2005;45:294–8.
    1. Roberts PJ, Rollins KD, Kashuba AD, et al. The influence of CYP3A5 genotype on dexamethasone induction of CYP3A activity in African Americans. Drug Metab Dispos. 2008;36:1465–9.
    1. Hukkanen J, Vaisanen T, Lassila A, et al. Regulation of CYP3A5 by glucocorticoids and cigarette smoke in human lung-derived cells. J Pharmacol Exp Ther. 2003;304:745–52.
    1. Schuetz JD, Schuetz EG, Thottassery JV, et al. Identification of a novel dexamethasone responsive enhancer in the human CYP3A5 gene and its activation in human and rat liver cells. Mol Pharmacol. 1996;49:63–72.
    1. Burk O, Koch I, Raucy J, et al. The induction of cytochrome P450 3A5 (CYP3A5) in the human liver and intestine is mediated by the xenobiotic sensors pregnane X receptor (PXR) and constitutively activated receptor (CAR) J Biol Chem. 2004;279:38379–85.
    1. Padmanabhan S, Melander O, Johnson T, et al. Genome-wide association study of blood pressure extremes identifies variant near UMOD associated with hypertension. PLoS Genet. 2010;6:e1001177.
    1. Kivisto KT, Niemi M, Schaeffeler E, et al. CYP3A5 genotype is associated with diagnosis of hypertension in elderly patients: data from the DEBATE Study. Am J Pharmacogenomics. 2005;5:191–5.
    1. Langaee TY, Gong Y, Yarandi HN, et al. Association of CYP3A5 polymorphisms with hypertension and antihypertensive response to verapamil. Clin Pharmacol Ther. 2007;81:386–91.
    1. Fromm MF, Schmidt BM, Pahl A, et al. CYP3A5 genotype is associated with elevated blood pressure. Pharmacogenet Genomics. 2005;15:737–41.
    1. Kreutz R, Zuurman M, Kain S, et al. The role of the cytochrome P450 3A5 enzyme for blood pressure regulation in the general Caucasian population. Pharmacogenet Genomics. 2005;15:831–7.
    1. Lieb W, Bolbrinker J, Doring A, et al. No association of the CYP3A5*1 allele with blood pressure and left ventricular mass and geometry: the KORA/MONICA Augsburg echocardiographic substudy. Clin Sci (Lond) 2006;111:365–72.
    1. Coto E, Tavira B, Marin R, et al. Functional polymorphisms in the CYP3A4, CYP3A5, and CYP21A2 genes in the risk for hyper- tension in pregnancy. Biochem Biophys Res Commun. 2010;397:576–9.
    1. Yazdanpanah M, Aulchenko YS, Hofman A, et al. Effects of the renin-angiotensin system genes and salt sensitivity genes on blood pressure and atherosclerosis in the total population and patients with type 2 diabetes. Diabetes. 2007;56:1905–12.
    1. Watlington CO, Kramer LB, Schuetz EG, et al. Corticosterone 6 beta-hydroxylation correlates with blood pressure in spontaneously hypertensive rats. Am J Physiol. 1992;262:F927–31.
    1. Ghosh SS, Basu AK, Ghosh S, et al. Renal and hepatic family 3A cytochromes P450 (CYP3A) in spontaneously hypertensive rats. Biochem Pharmacol. 1995;50:49–54.
    1. Siest G, Jeannesson E, Visvikis-Siest S. Enzymes and pharma- cogenetics of cardiovascular drugs. Clin Chim Acta. 2007;381:26–31.
    1. Willrich MA, Hirata MH, Hirata RD. Statin regulation of CYP3A4 and CYP3A5 expression. Pharmacogenomics. 2009;10:1017–24.
    1. Jin Y, Wang YH, Miao J, et al. Cytochrome P450 3A5 genotype is associated with verapamil response in healthy subjects. Clin Pharmacol Ther. 2007;82:579–85.
    1. Bhatnagar V, Garcia EP, O'Connor DT, et al. CYP3A4 and CYP3A5 polymorphisms and blood pressure response to amlodipine among African-American men and women with early hypertensive renal disease. Am J Nephrol. 2010;31:95–103.
    1. Kim KA, Park PW, Lee OJ, et al. Effect of CYP3A5*3 genotype on the pharmacokinetics and pharmacodynamics of amlodipine in healthy Korean subjects. Clin Pharmacol Ther. 2006;80:646–56.
    1. Zhang L, Miyaki K, Wang W, et al. CYP3A5 polymorphism and sensitivity of blood pressure to dietary salt in Japanese men. J Hum Hypertens. 2010;24:345–50.
    1. Darbar D, Dell'Orto S, Morike K, et al. Dietary salt increases first-pass elimination of oral quinidine. Clin Pharmacol Ther. 1997;61:292–300.
    1. Darbar D, Fromm MF, Dell'Orto S, et al. Modulation by dietary salt of verapamil disposition in humans. Circulation. 1998;98:2702–8.
    1. Kang HJ, Song IS, Lee SS, et al. Effects of dietary salt on the expression of drug transporters, cytochrome P4503a, and nuclear receptors in rats. Xenobiotica. 2008;38:147–55.
    1. Kosuge K, Chuang AI, Uematsu S, et al. Discovery of osmo- sensitive transcriptional regulation of human cytochrome P450 3As by the tonicity-responsive enhancer binding protein (nuclear factor of activated T cells 5) Mol Pharmacol. 2007;72:826–37.
    1. Wang D, Johnson AD, Papp AC, et al. Multidrug resistance polypeptide 1 (MDR1, ABCB1) variant 3435C>T affects mRNA stability. Pharmacogenet Genomics. 2005;15:693–704.
    1. Kimchi-Sarfaty C, Oh JM, Kim IW, et al. A "silent" polymorphism in the MDR1 gene changes substrate specificity. Science. 2007;315:525–8.
    1. Hauser IA, Koziolek M, Hopfer U, et al. Therapeutic concentrations of cyclosporine A, but not FK506, increase P-glycoprotein expression in endothelial and renal tubule cells. Kidney Int. 1998;54:1139–49.
    1. Thiebaut F, Tsuruo T, Hamada H, et al. Cellular localization of the multidrug-resistance gene product P-glycoprotein in normal human tissues. Proc Natl Acad Sci U S A. 1987;84:7735–8.
    1. Ernest S, Rajaraman S, Megyesi J, et al. Expression of MDR1 (multidrug resistance) gene and its protein in normal human kidney. Nephron. 1997;77:284–9.
    1. Geick A, Eichelbaum M, Burk O. Nuclear receptor response elements mediate induction of intestinal MDR1 by rifampin. J Biol Chem. 2001;276:14581–7.
    1. Burk O, Arnold KA, Geick A, et al. A role for constitutive androstane receptor in the regulation of human intestinal MDR1 expression. Biol Chem. 2005;386:503–13.
    1. Tachibana S, Yoshinari K, Chikada T, et al. Involvement of Vitamin D receptor in the intestinal induction of human ABCB1. Drug Metab Dispos. 2009;37:1604–10.
    1. Saeki M, Kurose K, Tohkin M, et al. Identification of the functional vitamin D response elements in the human MDR1 gene. Biochem Pharmacol. 2008;76:531–42.
    1. Chow EC, Durk MR, Cummins CL, et al. 1Alpha,25-dihydroxyvitamin D3 up-regulates P-glycoprotein via the vitamin D receptor and not farnesoid X receptor in both fxr(-/-) and fxr(+/+) mice and increased renal and brain efflux of digoxin in mice in vivo. J Pharmacol Exp Ther. 2011;337:846–59.
    1. Kim RB. Drugs as P-glycoprotein substrates, inhibitors, and inducers. Drug Metab Rev. 2002;34:47–54.
    1. Ueda K, Okamura N, Hirai M, et al. Human P-glycoprotein transports cortisol, aldosterone, and dexamethasone, but not progesterone. J Biol Chem. 1992;267:24248–52.
    1. Zhou SF. Structure, function and regulation of P-glycoprotein and its clinical relevance in drug disposition. Xenobiotica. 2008;38:802–32.
    1. Uhr M, Holsboer F, Muller MB. Penetration of endogenous steroid hormones corticosterone, cortisol, aldosterone and progesterone into the brain is enhanced in mice deficient for both mdr1a and mdr1b P-glycoproteins. J Neuroendocrinol. 2002;14:753–9.
    1. Bello-Reuss E, Ernest S, Holland OB, et al. Role of multidrug resistance P-glycoprotein in the secretion of aldosterone by human adrenal NCI-H295 cells. Am J Physiol Cell Physiol. 2000;278:C1256–65.
    1. Nakamura T, Okamura N, Yagi M, et al. Effects of ABCB1 3435C>T genotype on serum levels of cortisol and aldosterone in women with normal menstrual cycles. Genet Mol Res. 2009;8:397–403.
    1. Rigalli JP, Ruiz ML, Perdomo VG, et al. Pregnane X receptor mediates the induction of P-glycoprotein by spironolactone in HepG2 cells. Toxicology. 2011;285:18–24.
    1. Parker RB, Yates CR, Laizure SC, et al. P-glycoprotein modulates aldosterone plasma disposition and tissue uptake. J Cardiovasc Pharmacol. 2006;47:55–9.
    1. Huang BS, Wang H, Leenen FH. Chronic central infusion of aldosterone leads to sympathetic hyperreactivity and hypertension in Dahl S but not Dahl R rats. Am J Physiol Circ Physiol. 2005;288:H517–24.
    1. Huang BS, Cheung WJ, Wang H, et al. Activation of brain renin-angiotensin-aldosterone system by central sodium in Wistar rats. Am J Physiol Circ Physiol. 2006;291:H1109–17.
    1. Widder JD, Guzik TJ, Mueller CF, et al. Role of the multidrug resistance protein-1 in hypertension and vascular dysfunction caused by angiotensin II. Arterioscler Thromb Vasc Biol. 2007;27:762–8.
    1. Zolk O, Jacobi J, Pahl A, et al. MDR1 genotype-dependent regulation of the aldosterone system in humans. Pharmacogenet Genomics. 2007;17:137–44.
    1. Tapaninen T, Backman JT, Kurkinen KJ, et al. Itraconazole, a P-glycoprotein and CYP3A4 inhibitor, markedly raises the plasma concentrations and enhances the renin-inhibiting effect of aliskiren. J Clin Pharmacol. 2011;51:359–67.
    1. Tripodi G, Citterio L, Kouznetsova T, et al. Steroid biosynthesis and renal excretion in human essential hypertension: association with blood pressure and endogenous ouabain. Am J Hypertens. 2009;22:357–63.
    1. Weiss J, Sauer A, Divac N, et al. Interaction of angiotensin receptor type 1 blockers with ATP-binding cassette transporters. Biopharmaceutics Drug Dispos. 2010;31:150–61.
    1. Kamiyama E, Nakai D, Mikkaichi T, et al. Interaction of angiotensin II type 1 receptor blockers with P-gp substrates in Caco-2 cells and hMDR1-expressing membranes. Life sciences. 2010;86:52–8.
    1. Stern N, Lustig S, Petrasek D, et al. Cyclosporin A-induced hyperreninemic hypoaldosteronism. A model of adrenal resistance to angiotensin II. Hypertension. 1987;9:III31–5.
    1. Adu D, Turney J, Michael J, et al. Hyperkalaemia in cyclosporin-treated renal allograft recipients. Lancet. 1983;2:370–2.
    1. Woillard JB, Rerolle JP, Picard N, et al. Donor P-gp polymorphisms strongly influence renal function and graft loss in a cohort of renal transplant recipients on cyclosporine therapy in a long-term follow-up. Clin Pharmacol Ther. 2010;88:95–100.
    1. de Denus S, Zakrzewski M, Barhdadi A, et al. Association between renal function and CYP3A5 genotype in heart transplant recipients treated with calcineurin inhibitors. J Heart Lung Transplant. 2011;30:326–31.
    1. Del Moral RG, Olmo A, Osuna A, et al. Role of P-glycoprotein in chronic cyclosporine nephrotoxicity and its relationship to intrarenal angiotensin II deposits. Transplant Proc. 1998;30:2014–6.
    1. Koziolek MJ, Riess R, Geiger H, et al. Expression of multidrug resistance P-glycoprotein in kidney allografts from cyclosporine A-treated patients. Kidney Int. 2001;60:156–66.
    1. Naesens M, Lerut E, de Jonge H, et al. Donor age and renal P-glycoprotein expression associate with chronic histological damage in renal allografts. J Am Soc Nephrol. 2009;20:2468–80.
    1. Foote CJ, Greer W, Kiberd BA, et al. MDR1 C3435T poly- morphisms correlate with cyclosporine levels in de novo renal recipients. Transplant Proc. 2006;38:2847–9.
    1. Karssen AM, Meijer OC, van der Sandt IC, et al. Multidrug resistance P-glycoprotein hampers the access of cortisol but not of corticosterone to mouse and human brain. Endocrinology. 2001;142:2686–94.
    1. Iqbal M, Gibb W, Matthews SG. Corticosteroid regulation of P-glycoprotein in the developing blood-brain barrier. Endocrinology. 2011;152:1067–79.
    1. Goodwin JE, Geller DS. Glucocorticoid-induced hypertension. Pediatr nephrol. 2011. [Epub ahead of print]
    1. Magiakou MA, Smyrnaki P, Chrousos GP. Hypertension in Cushing's syndrome. Best Pract Res Clin Endocrinol Metab. 2006;20:467–82.
    1. Zhou S, Lim LY, Chowbay B. Herbal modulation of P-glycoprotein. Drug Metab Rev. 2004;36:57–104.
    1. Soldner A, Christians U, Susanto M, et al. Grapefruit juice activates P-glycoprotein-mediated drug transport. Pharm Res. 1999;16:478–85.
    1. Morales MM, Capella MA, Sanches MV, et al. Modulation of the mdr-1b gene in the kidney of rats subjected to dehydration or a high-salt diet. Pflugers Arch. 2000;439:356–62.
    1. Schinkel AH, Smit JJ, van Tellingen O, et al. Disruption of the mouse mdr1a P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to drugs. Cell. 1994;77:491–502.
    1. Huls M, Kramers C, Levtchenko EN, et al. P-glycoprotein-deficient mice have proximal tubule dysfunction but are protected against ischemic renal injury. Kidney Int. 2007;72:1233–41.
    1. Chambers JC, Zhang W, Lord GM, et al. Genetic loci influencing kidney function and chronic kidney disease. Nat Genet. 2010;42:373–5.
    1. Kottgen A, Pattaro C, Boger CA, et al. New loci associated with kidney function and chronic kidney disease. Nat Genet. 2010;42:376–84.
    1. Murakami T, Ohmori H, Katoh T, et al. Cadmium causes increases of N-myc and multidrug-resistance gene mRNA in neuroblastoma cells. J UOEH. 1991;13:271–8.
    1. Lee WK, Torchalski B, Kohistani N, et al. ABCB1 protects kidney proximal tubule cells against cadmium-induced apoptosis: roles of cadmium and ceramide transport. Toxicol Sci. 2011;121:343–56.
    1. Thevenod F, Friedmann JM, Katsen AD, et al. Up-regulation of multidrug resistance P-glycoprotein via nuclear factor-kappaB activation protects kidney proximal tubule cells from cadmium- and reactive oxygen species-induced apoptosis. J Biol Chem. 2000;275:1887–96.
    1. Abu-Hayyeh S, Sian M, Jones KG, et al. Cadmium accumulation in aortas of smokers. Arterioscler Thromb Vasc Biol. 2001;21:863–7.
    1. Messner B, Knoflach M, Seubert A, et al. Cadmium is a novel and independent risk factor for early atherosclerosis mechanisms and in vivo relevance. Arterioscler Thromb Vasc Biol. 2009;29:1392–8.
    1. Navas-Acien A, Selvin E, Sharrett AR, et al. Lead, cadmium, smoking, and increased risk of peripheral arterial disease. Circulation. 2004;109:3196–201.
    1. Haussler MR, Norman AW. Chromosomal receptor for a vitamin D metabolite. Proc Natl Acad Sci U S A. 1969;62:155–62.
    1. Kumar R, Schaefer J, Grande JP, et al. Immunolocalization of calcitriol receptor, 24-hydroxylase cytochrome P-450, and calbindin D28k in human kidney. Am J Physiol. 1994;266:F477–85.
    1. Wang TT, Tavera-Mendoza LE, Laperriere D, et al. Large-scale in silico and microarray-based identification of direct 1,25-dihydroxyvitamin D3 target genes. Mol Endocrinol. 2005;19:2685–95.
    1. Schuster I. Cytochromes P450 are essential players in the vitamin D signaling system. Biochim Biophys Acta. 2011;1814:186–99.
    1. Makishima M, Lu TT, Xie W, et al. Vitamin D receptor as an intestinal bile acid sensor. Science. 2002;296:1313–6.
    1. Barthel TK, Mathern DR, Whitfield GK, et al. 1,25-Dihydroxyvitamin D3/VDR-mediated induction of FGF23 as well as transcriptional control of other bone anabolic and catabolic genes that orchestrate the regulation of phosphate and calcium mineral metabolism. J Steroid Biochem Mol Biol. 2007;103:381–8.
    1. Meyer MB, Watanuki M, Kim S, et al. The human transient receptor potential vanilloid type 6 distal promoter contains multiple vitamin D receptor binding sites that mediate activation by 1,25-dihydroxyvitamin D3 in intestinal cells. Mol Endocrinol. 2006;20:1447–61.
    1. Haussler MR, Haussler CA, Bartik L, et al. Vitamin D receptor: molecular signaling and actions of nutritional ligands in disease prevention. Nutr Rev. 2008;66:S98–112.
    1. Holick MF. Vitamin D deficiency. N Engl J Med. 2007;357:266–81.
    1. Holick MF. McCollum Award Lecture, 1994: vitamin D--new horizons for the 21st century. Am J Clin Nutr. 1994;60:619–30.
    1. Dusso A, Brown A, Slatopolsky E. Extrarenal production of calcitriol. Semin Nephrol. 1994;14:144–55.
    1. Khanal R, Nemere I. Membrane receptors for vitamin D metabolites. Crit Rev Eukaryot Gene Expr. 2007;17:31–47.
    1. Mizwicki MT, Norman AW. The vitamin D sterol-vitamin D receptor ensemble model offers unique insights into both genomic and rapid-response signaling. Sci Signal. 2009;2:re4.
    1. Uitterlinden AG, Fang Y, Van Meurs JB, et al. Genetics and biology of vitamin D receptor polymorphisms. Gene. 2004;338:143–56.
    1. Levin A, Bakris GL, Molitch M, et al. Prevalence of abnormal serum vitamin D, PTH, calcium, and phosphorus in patients with chronic kidney disease: results of the study to evaluate early kidney disease. Kidney Int. 2007;71:31–8.
    1. Ravani P, Malberti F, Tripepi G, et al. Vitamin D levels and patient outcome in chronic kidney disease. Kidney Int. 2009;75:88–95.
    1. Wolf M, Betancourt J, Chang Y, et al. Impact of activated vitamin D and race on survival among hemodialysis patients. J Am Soc Nephrol. 2008;19:1379–88.
    1. Li YC. Renoprotective effects of vitamin D analogs. Kidney Int. 2010;78:134–9.
    1. Tan X, Li Y, Liu Y. Paricalcitol attenuates renal interstitial fibrosis in obstructive nephropathy. J Am Soc Nephrol. 2006;17:3382–93.
    1. Makibayashi K, Tatematsu M, Hirata M, et al. A vitamin D analog ameliorates glomerular injury on rat glomerulonephritis. Am J Pathol. 2001;158:1733–41.
    1. Alborzi P, Patel NA, Peterson C, et al. Paricalcitol reduces albuminuria and inflammation in chronic kidney disease: a randomized double-blind pilot trial. Hypertension. 2008;52:249–55.
    1. Agarwal R, Acharya M, Tian J, et al. Antiproteinuric effect of oral paricalcitol in chronic kidney disease. Kidney Int. 2005;68:2823–8.
    1. de Zeeuw D, Agarwal R, Amdahl M, et al. Selective vitamin D receptor activation with paricalcitol for reduction of albuminuria in patients with type 2 diabetes (VITAL study): a randomised controlled trial. Lancet. 2010;376:1543–51.
    1. Fishbane S, Chittineni H, Packman M, et al. Oral paricalcitol in the treatment of patients with CKD and proteinuria: a randomized trial. Am J Kidney Dis. 2009;54:647–52.
    1. Szeto CC, Chow KM, Kwan BC, et al. Oral calcitriol for the treatment of persistent proteinuria in immunoglobulin A nephropathy: an uncontrolled trial. Am J Kidney Dis. 2008;51:724–31.
    1. Kivlighn SD, Dzielak DJ. The role of the renin angiotensin system in chronic renal disease. Expert Opin Investig Drugs. 1997;6:1643–50.
    1. Resnick LM, Muller FB, Laragh JH. Calcium-regulating hormones in essential hypertension. Relation to plasma renin activity and sodium metabolism. Ann Intern Med. 1986;105:649–54.
    1. Li YC, Kong J, Wei M, et al. 1,25-Dihydroxyvitamin D(3) is a negative endocrine regulator of the renin-angiotensin system. J Clin Invest. 2002;110:229–38.
    1. Freundlich M, Quiroz Y, Zhang Z, et al. Suppression of renin-angiotensin gene expression in the kidney by paricalcitol. Kidney Int. 2008;74:1394–402.
    1. Tripathi G, Sharma R, Sharma RK, et al. Vitamin D receptor genetic variants among patients with end-stage renal disease. Ren Fail. 2010;32:969–77.
    1. de Souza CM, Braosi AP, Luczyszyn SM, et al. Association between vitamin D receptor gene polymorphisms and susceptibility to chronic kidney disease and periodontitis. Blood Purif. 2007;25:411–9.
    1. Marco MP, Martinez I, Amoedo ML, et al. Vitamin D receptor genotype influences parathyroid hormone and calcitriol levels in predialysis patients. Kidney Int. 1999;56:1349–53.
    1. Ned RM, Yesupriya A, Imperatore G, et al. Inflammation gene variants and susceptibility to albuminuria in the U.S. population: analysis in the Third National Health and Nutrition Examination Survey (NHANES III), 1991-1994. BMC Med Genet. 2010;11:155.
    1. Ortlepp JR, Metrikat J, Albrecht M, et al. The vitamin D receptor gene variant and physical activity predicts fasting glucose levels in healthy young men. Diabet Med. 2003;20:451–4.
    1. Ortlepp JR, Lauscher J, Hoffmann R, et al. The vitamin D receptor gene variant is associated with the prevalence of type 2 diabetes mellitus and coronary artery disease. Diabet Med. 2001;18:842–5.
    1. Filus A, Trzmiel A, Kuliczkowska-Plaksej J, et al. Relationship between vitamin D receptor BsmI and FokI polymorphisms and anthropometric and biochemical parameters describing metabolic syndrome. Aging Male. 2008;11:134–9.
    1. Oh JY, Barrett-Connor E. Association between vitamin D receptor polymorphism and type 2 diabetes or metabolic syndrome in community-dwelling older adults: the Rancho Bernardo Study. Metabolism. 2002;51:356–9.
    1. Hitman GA, Mannan N, McDermott MF, et al. Vitamin D receptor gene polymorphisms influence insulin secretion in Bangladeshi Asians. Diabetes. 1998;47:688–90.
    1. Ogunkolade BW, Boucher BJ, Prahl JM, et al. Vitamin D receptor (VDR) mRNA and VDR protein levels in relation to vitamin D status, insulin secretory capacity, and VDR genotype in Bangladeshi Asians. Diabetes. 2002;51:2294–300.
    1. Calvo MS, Whiting SJ, Barton CN. Vitamin D fortification in the United States and Canada: current status and data needs. Am J Clin Nutr. 2004;80:1710S–6S.
    1. Elnenaei MO, Chandra R, Mangion T, et al. Genomic and metabolomic patterns segregate with responses to calcium and vitamin D supplementation. Br J Nutr. 2011;105:71–9.
    1. Guessous I, Bochud M, Bonny O, et al. Calcium, Vitamin D and Cardiovascular Disease. Kidney & blood pressure research. 2011;34:404–17.
    1. Pfeifer M, Begerow B, Minne HW, et al. Effects of a short-term vitamin D(3) and calcium supplementation on blood pressure and parathyroid hormone levels in elderly women. J Clin Endocrinol Metab. 2001;86:1633–7.
    1. Wang L, Manson JE, Buring JE, et al. Dietary intake of dairy products, calcium, and vitamin D and the risk of hypertension in middle-aged and older women. Hypertension. 2008;51:1073–9.
    1. Xiang W, Kong J, Chen S, et al. Cardiac hypertrophy in vitamin D receptor knockout mice: role of the systemic and cardiac renin-angiotensin systems. Am J Physiol Endocrinol Metab. 2005;288:E125–32.
    1. Kong J, Qiao G, Zhang Z, et al. Targeted vitamin D receptor expression in juxtaglomerular cells suppresses renin expression independent of parathyroid hormone and calcium. Kidney Int. 2008;74:1577–81.
    1. Bonizzi G, Karin M. The two NF-kappaB activation pathways and their role in innate and adaptive immunity. Trends Immunol. 2004;25:280–8.
    1. Guijarro C, Egido J. Transcription factor-kappa B (NF-kappa B) and renal disease. Kidney Int. 2001;59:415–24.
    1. Richart T, Li Y, Staessen JA. Renal versus extrarenal activation of vitamin D in relation to atherosclerosis, arterial stiffening, and hypertension. Am J Hypertens. 2007;20:1007–15.
    1. He J, Cheng Q, Xie W. Minireview: Nuclear receptor-controlled steroid hormone synthesis and metabolism. Mol Endocrinol. 2010;24:11–21.
    1. Pechere-Bertschi A, Burnier M. Female sex hormones, salt, and blood pressure regulation. Am J Hypertens. 2004;17:994–1001.
    1. Kinuta K, Tanaka H, Moriwake T, et al. Vitamin D is an important factor in estrogen biosynthesis of both female and male gonads. Endocrinology. 2000;141:1317–24.
    1. Yoshizawa T, Handa Y, Uematsu Y, et al. Mice lacking the vitamin D receptor exhibit impaired bone formation, uterine hypoplasia and growth retardation after weaning. Nat Genet. 1997;16:391–6.
    1. Enjuanes A, Garcia-Giralt N, Supervia A, et al. Regulation of CYP19 gene expression in primary human osteoblasts: effects of vitamin D and other treatments. Eur J Endocrinol. 2003;148:519–26.
    1. Hughes PJ, Twist LE, Durham J, et al. Up-regulation of steroid sulphatase activity in HL60 promyelocytic cells by retinoids and 1alpha,25-dihydroxyvitamin D3. Biochem J. 2001;355:361–71.
    1. Velayoudom-Cephise FL, Larifla L, Donnet JP, et al. Vitamin D deficiency, vitamin D receptor gene polymorphisms and cardiovascular risk factors in Caribbean patients with type 2 diabetes. Diabetes Metab. 2011. [Epub ahead of print]
    1. Tworowska-Bardzinska U, Lwow F, Kubicka E, et al. The vitamin D receptor gene BsmI polymorphism is not associated with anthropometric and biochemical parameters describing metabolic syndrome in postmenopausal women. Gynecol Endocrinol. 2008;24:514–8.
    1. Kulah E, Dursun A, Acikgoz S, et al. The relationship of target organ damage and 24-hour ambulatory blood pressure monitoring with vitamin D receptor gene fok-I polymorphism in essential hypertension. Kidney & blood pressure research. 2006;29:344–50.
    1. Muray S, Parisi E, Cardus A, et al. Influence of vitamin D receptor gene polymorphisms and 25-hydroxyvitamin D on blood pressure in apparently healthy subjects. J Hypertens. 2003;21:2069–75.
    1. Vaidya A, Forman JP, Hopkins PN, et al. 25-Hydroxyvitamin D is associated with plasma renin activity and the pressor response to dietary sodium intake in Caucasians. J Renin Angiotensin Aldosterone Syst. 2011;12(3 ):311–9.
    1. Levi M. Nuclear receptors in renal disease. Biochim Biophys Acta. 2011;1812:1061–7.
    1. Zhang B, Xie W, Krasowski MD. PXR: a xenobiotic receptor of diverse function implicated in pharmacogenetics. Pharmacogenomics. 2008;9:1695–709.
    1. Fukuen S, Fukuda T, Matsuda H, et al. Identification of the novel splicing variants for the hPXR in human livers. Biochem Biophys Res Commun. 2002;298:433–8.
    1. Zhou C, Verma S, Blumberg B. The steroid and xenobiotic receptor (SXR), beyond xenobiotic metabolism. Nuclear receptor signaling. 2009;7:e001.
    1. Hustert E, Zibat A, Presecan-Siedel E, et al. Natural protein variants of pregnane X receptor with altered transactivation activity toward CYP3A4. Drug Metab Dispos. 2001;29:1454–9.
    1. Bosch TM, Deenen M, Pruntel R, et al. Screening for polymorphisms in the PXR gene in a Dutch population. Eur J Clin Pharmacol. 2006;62:395–9.
    1. Zhang J, Kuehl P, Green ED, et al. The human pregnane X receptor: genomic structure and identification and functional characterization of natural allelic variants. Pharmacogenetics. 2001;11:555–72.
    1. Koyano S, Kurose K, Ozawa S, et al. Eleven novel single nucleotide polymorphisms in the NR1I2 (PXR) gene, four of which induce non-synonymous amino acid alterations. Drug Metab Pharmacokinet. 2002;17:561–5.
    1. Lim YP, Liu CH, Shyu LJ, et al. Functional characterization of a novel polymorphism of pregnane X receptor, Q158K, in Chinese subjects. Pharmacogenet Genomics. 2005;15:337–41.
    1. King CR, Xiao M, Yu J, et al. Identification of NR1I2 genetic variation using resequencing. Eur J Clin Pharmacol. 2007;63:547–54.
    1. Miki Y, Suzuki T, Tazawa C, et al. Steroid and xenobiotic receptor (SXR), cytochrome P450 3A4 and multidrug resistance gene 1 in human adult and fetal tissues. Mol Cell Endocrinol. 2005;231:75–85.
    1. Synold TW, Dussault I, Forman BM. The orphan nuclear receptor SXR coordinately regulates drug metabolism and efflux. Nat Med. 2001;7:584–90.
    1. Sonoda J, Xie W, Rosenfeld JM, et al. Regulation of a xenobiotic sulfonation cascade by nuclear pregnane X receptor (PXR) Proc Natl Acad Sci U S A. 2002;99:13801–6.
    1. Klaassen CD, Slitt AL. Regulation of hepatic transporters by xenobiotic receptors. Curr Drug Metab. 2005;6:309–28.
    1. Koyano S, Kurose K, Saito Y, et al. Functional characterization of four naturally occurring variants of human pregnane X receptor (PXR): one variant causes dramatic loss of both DNA binding activity and the transactivation of the CYP3A4 promoter/enhancer region. Drug Metab Dispos. 2004;32:149–54.
    1. Orans J, Teotico DG, Redinbo MR. The nuclear xenobiotic receptor pregnane X receptor: recent insights and new challenges. Mol Endocrinol. 2005;19:2891–900.
    1. Xue Y, Moore LB, Orans J, et al. Crystal structure of the pregnane X receptor-estradiol complex provides insights into endobiotic recognition. Mol Endocrinol. 2007;21:1028–38.
    1. Tabb MM, Sun A, Zhou C, et al. Vitamin K2 regulation of bone homeostasis is mediated by the steroid and xenobiotic receptor SXR. J Biol Chem. 2003;278:43919–27.
    1. Landes N, Pfluger P, Kluth D, et al. Vitamin E activates gene expression via the pregnane X receptor. Biochem Pharmacol. 2003;65:269–73.
    1. Reschly EJ, Krasowski MD. Evolution and function of the NR1I nuclear hormone receptor subfamily (VDR, PXR, and CAR) with respect to metabolism of xenobiotics and endogenous compounds. Curr Drug Metab. 2006;7:349–65.
    1. Xie W, Yeuh MF, Radominska-Pandya A, et al. Control of steroid, heme, and carcinogen metabolism by nuclear pregnane X receptor and constitutive androstane receptor. Proc Natl Acad Sci U S A. 2003;100:4150–5.
    1. Zhai Y, Pai HV, Zhou J, et al. Activation of pregnane X receptor disrupts glucocorticoid and mineralocorticoid homeostasis. Mol Endocrinol. 2007;21:138–47.
    1. Hagedorn KA, Cooke CL, Falck JR, et al. Regulation of vascular tone during pregnancy: a novel role for the pregnane X receptor. Hypertension. 2007;49:328–33.
    1. Terzolo M, Borretta G, Ali A, et al. Misdiagnosis of Cushing's syndrome in a patient receiving rifampicin therapy for tuberculosis. Horm Metab Res. 1995;27:148–50.
    1. Hanif K, Bid HK, Konwar R. Reinventing the ACE inhibitors: some old and new implications of ACE inhibition. Hypertens Res. 2010;33:11–21.
    1. Rigat B, Hubert C, Alhenc-Gelas F, et al. An insertion/deletion polymorphism in the angiotensin I-converting enzyme gene accounting for half the variance of serum enzyme levels. J Clin Invest. 1990;86:1343–6.
    1. Gard PR. Implications of the angiotensin converting enzyme gene insertion/deletion polymorphism in health and disease: a snapshot review. Int J Mol Epidemol Genet. 2010;1:145–57.
    1. Sayed-Tabatabaei FA, Oostra BA, Isaacs A, et al. ACE polymorphisms. Circ Res. 2006;98:1123–33.
    1. Kramers C, Danilov SM, Deinum J, et al. Point mutation in the stalk of angiotensin-converting enzyme causes a dramatic increase in serum angiotensin-converting enzyme but no cardiovascular disease. Circulation. 2001;104:1236–40.
    1. Rieder MJ, Taylor SL, Clark AG, et al. Sequence variation in the human angiotensin converting enzyme. Nat Genet. 1999;22:59–62.
    1. Staessen JA, Wang JG, Ginocchio G, et al. The deletion/insertion polymorphism of the angiotensin converting enzyme gene and cardiovascular-renal risk. J Hypertens. 1997;15:1579–92.
    1. Agerholm-Larsen B, Nordestgaard BG, Tybjaerg-Hansen A. ACE gene polymorphism in cardiovascular disease: meta-analyses of small and large studies in whites. Arterioscler Thromb Vasc Biol. 2000;20:484–92.
    1. Ji LD, Zhang LN, Shen P, et al. Association of angiotensinogen gene M235T and angiotensin-converting enzyme gene I/D polymorphisms with essential hypertension in Han Chinese population: a meta-analysis. J Hypertens. 2010;28:419–28.
    1. Johnson AD, Newton-Cheh C, Chasman DI, et al. Association of hypertension drug target genes with blood pressure and hypertension in 86,588 individuals. Hypertension. 2011;57:903–10.
    1. Kuznetsova T, Staessen JA, Wang JG, et al. Antihypertensive treatment modulates the association between the D/I ACE gene polymorphism and left ventricular hypertrophy: a meta-analysis. J Hum Hypertens. 2000;14:447–54.
    1. Zintzaras E, Raman G, Kitsios G, et al. Angiotensin-converting enzyme insertion/deletion gene polymorphic variant as a marker of coronary artery disease: a meta-analysis. Arch Int Med. 2008;168:1077–89.
    1. Tao HM, Shao B, Chen GZ. Meta-analysis of the ACE gene polymorphism in cerebral infarction. Can J Neurol Sci. 2009;36:20–5.
    1. Medica I, Kastrin A, Peterlin B. Genetic polymorphisms in vasoactive genes and preeclampsia: a meta-analysis. Eur J Obstet Gynecol Reprod Biol. 2007;131:115–26.
    1. Mooyaart AL, Valk EJ, van Es LA, et al. Genetic associations in diabetic nephropathy: a meta-analysis. Diabetologia. 2011;54:544–53.
    1. Ruggenenti P, Bettinaglio P, Pinares F, et al. Angiotensin converting enzyme insertion/deletion polymorphism and renoprotection in diabetic and nondiabetic nephropathies. Clin J Am Soc Nephrol. 2008;3:1511–25.
    1. Campbell CY, Fang BF, Guo X, et al. Associations between genetic variants in the ACE, AGT, AGTR1 and AGTR2 genes and renal function in the Multi-ethnic Study of Atherosclerosis. Am J Nephrol. 2010;32:156–62.
    1. Pereira TV, Nunes AC, Rudnicki M, et al. Influence of ACE I/D gene polymorphism in the progression of renal failure in autosomal dominant polycystic kidney disease: a meta-analysis. Nephrol Dial Transplant. 2006;21:3155–63.
    1. Yong D, Qing WQ, Hua L, et al. Association of angiotensin I-converting enzyme gene insertion/deletion polymorphism and IgA nephropathy: a meta-analysis. Am J Nephrol. 2006;26:511–8.
    1. Giner V, Poch E, Bragulat E, et al. Renin-angiotensin system genetic polymorphisms and salt sensitivity in essential hypertension. Hypertension. 2000;35:512–7.
    1. Hiraga H, Oshima T, Watanabe M, et al. Angiotensin I-converting enzyme gene polymorphism and salt sensitivity in essential hypertension. Hypertension. 1996;27:569–72.
    1. Poch E, Gonzalez D, Giner V, et al. Molecular basis of salt sensitivity in human hypertension. Evaluation of renin-angiotensin-aldosterone system gene polymorphisms. Hypertension. 2001;38:1204–9.
    1. Kojima S, Inenaga T, Matsuoka H, et al. The association between salt sensitivity of blood pressure and some polymorphic factors. J Hypertens. 1994;12:797–801.
    1. Sanada H, Yatabe J, Midorikawa S, et al. Single-nucleotide polymorphisms for diagnosis of salt-sensitive hypertension. Clin Chem. 2006;52:352–60.
    1. Arnett DK, Claas SA, Glasser SP. Pharmacogenetics of anti-hypertensive treatment. Vascul Pharmacol. 2006;44:107–18.
    1. Harrap SB, Tzourio C, Cambien F, et al. The ACE gene I/D polymorphism is not associated with the blood pressure and cardiovascular benefits of ACE inhibition. Hypertension. 2003;42:297–303.
    1. Suonsyrja T, Hannila-Handelberg T, Fodstad H, et al. Renin-angiotensin system and alpha-adducin gene polymorphisms and their relation to responses to antihypertensive drugs: results from the GENRES study. Am J Hypertens. 2009;22:169–75.
    1. Arnett DK, Davis BR, Ford CE, et al. Pharmacogenetic association of the angiotensin-converting enzyme insertion/deletion polymorphism on blood pressure and cardiovascular risk in relation to antihypertensive treatment: the Genetics of Hypertension-Associated Treatment (GenHAT) study. Circulation. 2005;111:3374–83.
    1. Brugts JJ, Isaacs A, Boersma E, et al. Genetic determinants of treatment benefit of the angiotensin-converting enzyme-inhibitor perindopril in patients with stable coronary artery disease. Eur Heart J. 2010;31:1854–64.
    1. Parving HH, Jacobsen P, Tarnow L, et al. Effect of deletion polymorphism of angiotensin converting enzyme gene on progression of diabetic nephropathy during inhibition of angiotensin converting enzyme: observational follow up study. BMJ. 1996;313:591–4.
    1. Parving HH, de Zeeuw D, Cooper ME, et al. ACE gene polymorphism and losartan treatment in type 2 diabetic patients with nephropathy. J Am Soc Nephrol. 2008;19:771–9.
    1. van der Kleij FG, Schmidt A, Navis GJ, et al. Angiotensin converting enzyme insertion/deletion polymorphism and short-term renal response to ACE inhibition: role of sodium status. Kidney Int Suppl. 1997;63:S23–6.
    1. van der Kleij FG, de Jong PE, Henning RH, et al. Enhanced responses of blood pressure, renal function, and aldosterone to angiotensin I in the DD genotype are blunted by low sodium intake. J Am Soc Nephrol. 2002;13:1025–33.
    1. Luik PT, Hoogenberg K, Kerstens MN, et al. The influence of the ACE ( I/D) polymorphism on systemic and renal vascular responses to angiotensins in normotensive, normoalbuminuric Type 1 diabetes mellitus. Diabetologia. 2003;46:1131–9.
    1. Toffoli G, De Mattia E. Pharmacogenetic relevance of MTHFR polymorphisms. Pharmacogenomics. 2008;9:1195–206.
    1. Gaughan DJ, Barbaux S, Kluijtmans LA, et al. The human and mouse methylenetetrahydrofolate reductase (MTHFR) genes: genomic organization, mRNA structure and linkage to the CLCN6 gene. Gene. 2000;257:279–89.
    1. van der Put NM, Gabreels F, Stevens EM, et al. A second common mutation in the methylenetetrahydrofolate reductase gene: an additional risk factor for neural-tube defects? Am J Hum Genet. 1998;62:1044–51.
    1. Schwahn B, Rozen R. Polymorphisms in the methylenetetra-hydrofolate reductase gene: clinical consequences. Am J Pharmacogenomics. 2001;1:189–201.
    1. Waheed A. Homocysteine and risk of ischemic heart disease and stroke: a meta-analysis. JAMA. 2002;288:2015–22.
    1. Lewis SJ, Ebrahim S, Davey Smith G. Meta-analysis of MTHFR 677C->T polymorphism and coronary heart disease: does totality of evidence support causal role for homocysteine and preventive potential of folate? BMJ. 2005;331:1053.
    1. Kes P. Hyperhomocysteinemia in end-stage renal failure. Acta Med Croatica. 2000;54:175–81.
    1. Shishehbor MH, Oliveira LP, Lauer MS, et al. Emerging cardiovascular risk factors that account for a significant portion of attributable mortality risk in chronic kidney disease. Am J Cardiol. 2008;101:1741–6.
    1. Tripathi G, Sankhwar SN, Sharma RK, et al. Role of thrombotic risk factors in end-stage renal disease. Clin Appl Thromb Hemost. 2010;16:132–40.
    1. Poduri A, Mukherjee D, Sud K, et al. MTHFR A1298C polymorphism is associated with cardiovascular risk in end stage renal disease in North Indians. Mol Cell Biochem. 2008;308:43–50.
    1. Fung MM, Salem RM, Lipkowitz MS, et al. Methylenetetra-hydrofolate reductase (MTHFR) polymorphism A1298C (Glu429Ala) predicts decline in renal function over time in the African-American Study of Kidney Disease and Hypertension (AASK) Trial and Veterans Affairs Hypertension Cohort (VAHC) Nephrol Dial Transplant. 2011. [Epub ahead of print]
    1. Kerkeni M, Letaief A, Achour A, et al. Endothelial nitric oxide synthetase, methylenetetrahydrofolate reductase polymorphisms, and cardiovascular complications in Tunisian patients with nondiabetic renal disease. Clin Biochem. 2009;42:958–64.
    1. Jamison RL, Shih MC, Humphries DE, et al. Effect of the MTHFR C677T and A1298C polymorphisms on survival in patients with advanced CKD and ESRD: a prospective study. Am J Kidney Dis. 2009;53:779–89.
    1. Koupepidou P, Deltas C, Christofides TC, et al. The MTHFR 677TT and 677CT/1298AC genotypes in Cypriot patients may be predisposing to hypertensive nephrosclerosis and chronic renal failure. Int Angiol. 2005;24:287–94.
    1. Moczulski D, Fojcik H, Zukowska-Szczechowska E, et al. Effects of the C677T and A1298C polymorphisms of the MTHFR gene on the genetic predisposition for diabetic nephropathy. Nephrol Dial Transplant. 2003;18:1535–40.
    1. Goforth RL, Rennke H, Sethi S. Renal vascular sclerosis is associated with inherited thrombophilias. Kidney Int. 2006;70:743–50.
    1. Ninomiya T, Kiyohara Y, Kubo M, et al. Hyperhomocysteinemia and the development of chronic kidney disease in a general population: the Hisayama study. Am J Kidney Dis. 2004;44:437–45.
    1. van Guldener C. Why is homocysteine elevated in renal failure and what can be expected from homocysteine-lowering? Nephrol Dial Transplant. 2006;21:1161–6.
    1. Samuelsson O, Lee DM, Attman PO, et al. The plasma levels of homocysteine are elevated in moderate renal insufficiency but do not predict the rate of progression. Nephron. 1999;82:306–11.
    1. Blom HJ, De Vriese AS. Why are homocysteine levels increased in kidney failure? A metabolic approach. J Lab Clin Med. 2002;139:262–8.
    1. Brosnan JT. Homocysteine and cardiovascular disease: interactions between nutrition, genetics and lifestyle. Can J Appl Physiol. 2004;29:773–80.
    1. Kosmas IP, Tatsioni A, Ioannidis JP. Association of C677T polymorphism in the methylenetetrahydrofolate reductase gene with hypertension in pregnancy and pre-eclampsia: a meta-analysis. J Hypertens. 2004;22:1655–62.
    1. Conen D, Cheng S, Steiner LL, et al. Association of 77 polymorphisms in 52 candidate genes with blood pressure progression and incident hypertension: the Women's Genome Health Study. J Hypertens. 2009;27:476–83.
    1. Takeuchi F, Isono M, Katsuya T, et al. Blood pressure and hypertension are associated with 7 loci in the Japanese population. Circulation. 2010;121:2302–9.
    1. Tomaszewski M, Debiec R, Braund PS, et al. Genetic architecture of ambulatory blood pressure in the general population: insights from cardiovascular gene-centric array. Hypertension. 2010;56:1069–76.
    1. Demirel Y, Dogan S, Uludag A, et al. Combined Effect of Factor V Leiden, MTHFR, and Angiotensin-Converting Enzyme (Insertion/ Deletion) Gene Mutations in Hypertensive Adult Individuals: A Population-Based Study from Sivas and Canakkale, Turkey. Genet Test Mol Biomarkers . 2011. [Epub ahead of print]
    1. Hong KW, Go MJ, Jin HS, et al. Genetic variations in ATP2B1, CSK, ARSG and CSMD1 loci are related to blood pressure and/or hypertension in two Korean cohorts. J Hum Hypertens. 2010;24:367–72.
    1. Sen U, Mishra PK, Tyagi N, et al. Homocysteine to hydro- gen sulfide or hypertension. Cell Biochem Biophys. 2010;57:49–58.
    1. Rodriguez-Esparragon F, Hernandez-Perera O, Rodriguez-Perez JC, et al. The effect of methylenetetrahydrofolate reductase C677T common variant on hypertensive risk is not solely explained by increased plasma homocysteine values. Clin Exp Hypertens. 2003;25:209–20.
    1. Jiang S, Hsu YH, Niu T, et al. A common haplotype on methylenetetrahydrofolate reductase gene modifies the effect of angiotensin-converting enzyme inhibitor on blood pressure in essential hypertension patients--a family-based association study. Clin Exp Hypertens. 2005;27:509–21.
    1. Jiang S, Yu Y, Venners SA, et al. Effects of MTHFR and MS gene polymorphisms on baseline blood pressure and Benazepril effectiveness in Chinese hypertensive patients. J Hum Hypertens. 2011;25:172–7.
    1. Wilson CP, McNulty H, Scott JM, et al. Postgraduate Symposium: The MTHFR C677T polymorphism, B-vitamins and blood pressure. Proc Nutr Soc. 2010;69:156–65.
    1. Horigan G, McNulty H, Ward M, et al. Riboflavin lowers blood pressure in cardiovascular disease patients homozygous for the 677C-->T polymorphism in MTHFR. J Hypertens. 2010;28:478–86.
    1. Williams C, Kingwell BA, Burke K, et al. Folic acid supplementation for 3 wk reduces pulse pressure and large artery stiffness independent of MTHFR genotype. Am J Clin Nutr. 2005;82:26–31.
    1. Lunegova OS, Kerimkulova AS, Turdakmatov NB, et al. [Association of C677T gene polymorphism of methylenetetra-hydrofolate reductase with insulin resistance among Kirghizes] Kardiologiia. 2011;51:58–62.
    1. Chen AR, Zhang HG, Wang ZP, et al. C-reactive protein, vitamin B12 and C677T polymorphism of N-5,10-methylenetetra- hydrofolate reductase gene are related to insulin resistance and risk factors for metabolic syndrome in Chinese population. Clin Invest Med. 2010;33:E290–7.
    1. Movva S, Alluri RV, Venkatasubramanian S, et al. Association of methylene tetrahydrofolate reductase C677T genotype with type 2 diabetes mellitus patients with and without renal complications. Genet Test Mol Biomarkers. 2011;15:257–61.
    1. Rezen T, Rozman D, Pascussi JM, et al. Interplay between cholesterol and drug metabolism. Biochim Biophys Acta. 2011;1814:146–60.
    1. Strazzullo P, D'Elia L, Kandala NB, et al. Salt intake, stroke, and cardiovascular disease: meta-analysis of prospective studies. BMJ. 2009;339:b4567.
    1. Appel LJ, Frohlich ED, Hall JE, et al. The importance of population-wide sodium reduction as a means to prevent cardiovascular disease and stroke: a call to action from the American Heart Association. Circulation. 2011;123:1138–43.
    1. Cook NR, Cutler JA, Obarzanek E, et al. Long term effects of dietary sodium reduction on cardiovascular disease outcomes: observational follow-up of the trials of hypertension prevention (TOHP) BMJ. 2007;334:885–8.
    1. Zhao Q, Gu D, Hixson JE, et al. Common Variants in Epithelial Sodium Channel Genes Contribute to Salt-Sensitivity of Blood Pressure: The GenSalt Study. Circ Cardiovasc Genet. 2011;4:375–80.
    1. Guessous I, Gwinn M, Khoury MJ. Genome-wide association studies in pharmacogenomics: untapped potential for translation. Genome Med. 2009;1:46.

Source: PubMed

3
Předplatit