Genes involved in vasoconstriction and vasodilation system affect salt-sensitive hypertension

Lorena Citterio, Marco Simonini, Laura Zagato, Erika Salvi, Simona Delli Carpini, Chiara Lanzani, Elisabetta Messaggio, Nunzia Casamassima, Francesca Frau, Francesca D'Avila, Daniele Cusi, Cristina Barlassina, Paolo Manunta, Lorena Citterio, Marco Simonini, Laura Zagato, Erika Salvi, Simona Delli Carpini, Chiara Lanzani, Elisabetta Messaggio, Nunzia Casamassima, Francesca Frau, Francesca D'Avila, Daniele Cusi, Cristina Barlassina, Paolo Manunta

Abstract

The importance of excess salt intake in the pathogenesis of hypertension is widely recognized. Blood pressure is controlled primarily by salt and water balance because of the infinite gain property of the kidney to rapidly eliminate excess fluid and salt. Up to fifty percent of patients with essential hypertension are salt-sensitive, as manifested by a rise in blood pressure with salt loading. We conducted a two-stage genetic analysis in hypertensive patients very accurately phenotyped for their salt-sensitivity. All newly discovered never treated before, essential hypertensives underwent an acute salt load to monitor the simultaneous changes in blood pressure and renal sodium excretion. The first stage consisted in an association analysis of genotyping data derived from genome-wide array on 329 subjects. Principal Component Analysis demonstrated that this population was homogenous. Among the strongest results, we detected a cluster of SNPs located in the first introns of PRKG1 gene (rs7897633, p = 2.34E-05) associated with variation in diastolic blood pressure after acute salt load. We further focused on two genetic loci, SLC24A3 and SLC8A1 (plasma membrane sodium/calcium exchange proteins, NCKX3 and NCX1, respectively) with a functional relationship with the previous gene and associated to variations in systolic blood pressure (the imputed rs3790261, p = 4.55E-06; and rs434082, p = 4.7E-03). In stage 2, we characterized 159 more patients for the SNPs in PRKG1, SLC24A3 and SLC8A1. Combined analysis showed an epistatic interaction of SNPs in SLC24A3 and SLC8A1 on the pressure-natriuresis (p interaction = 1.55E-04, p model = 3.35E-05), supporting their pathophysiological link in cellular calcium homeostasis. In conclusions, these findings point to a clear association between body sodium-blood pressure relations and molecules modulating the contractile state of vascular cells through an increase in cytoplasmic calcium concentration.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Multidimensional scaling plot of stage…
Figure 1. Multidimensional scaling plot of stage 1 cohort.
Individuals in red circle were identified as outliers and subsequently excluded. The two principal components are plotted on the axis.
Figure 2. Renal pressure-natriuresis relationship during acute…
Figure 2. Renal pressure-natriuresis relationship during acute salt load according to the rs3790261 (SLC24A3), rs11893826 (SLC8A1) genotypes.
Urinary sodium excretion (UNa+) as a function of SBP, at basal level (t 0) and after salt load (t 120). The pressure-natriuresis curve is drawn in the left panel considering SLC8A1, rs11893826 polymorphism (carriers of the non risk allele G, green slope, and carriers of the risk allele A, orange slope), in the genetic background of the non risk SLC24A3 AA genotype at rs3790261. It is drawn in the right panel considering the same genotypes for SLC8A1, but within the genetic context of the risk SLC24A3 G allele at rs3790261. The significantly less steep slope of the orange line in the right panel, compared to the three other slopes indicates the need for BP increase in order to excrete the salt load in carriers of that specific allele combination.

References

    1. Abegunde DO, Mathers CD, Adam T, Ortegon M, Strong K. The burden and costs of chronic diseases in low-income and middle-income countries. Lancet. 2007;370:1929–1938.
    1. Meneton P, Jeunemaitre X, de Wardener HE, MacGregor GA. Links between dietary salt intake, renal salt handling, blood pressure, and cardiovascular diseases. Physiol Rev. 2005;85:679–715.
    1. Haddy FJ. Role of dietary salt in hypertension. Life Sci. 2006;79:1585–1592.
    1. Campese VM. Salt sensitivity in hypertension. Renal and cardiovascular implications. Hypertension. 1994;23:531–550.
    1. Sanders PW. Dietary salt intake, salt sensitivity, and cardiovascular health. Hypertension. 2009;53:442–445.
    1. Hill MA, Meininger GA, Davis MJ, Laher I. Therapeutic potential of pharmacologically targeting arteriolar myogenic tone. Trends Pharmacol Sci. 2009;30:363–374.
    1. Guyton AC, Langston JB, Navar G. Theory for Renal Autoregulation by Feedback at the Juxtaglomerular Apparatus. Circ Res. 1964;15:(SUPPL):187–197.
    1. Cowley AW., Jr Genetic and nongenetic determinants of salt sensitivity and blood pressure. Am J Clin Nutr. 1997;65:587S–593S.
    1. Cusi D, Barlassina C, Azzani T, Casari G, Citterio L, et al. Polymorphisms of alpha-adducin and salt sensitivity in patients with essential hypertension. Lancet. 1997;349:1353–1357.
    1. Manunta P, Lavery G, Lanzani C, Braund PS, Simonini M, et al. Physiological interaction between alpha-adducin and WNK1-NEDD4L pathways on sodium-related blood pressure regulation. Hypertension. 2008;52:366–372.
    1. Barlassina C, Schork NJ, Manunta P, Citterio L, Sciarrone M, et al. Synergistic effect of alpha-adducin and ACE genes causes blood pressure changes with body sodium and volume expansion. Kidney Int. 2000;57:1083–1090.
    1. Manunta P, Messaggio E, Ballabeni C, Sciarrone MT, Lanzani C, et al. Plasma ouabain-like factor during acute and chronic changes in sodium balance in essential hypertension. Hypertension. 2001;38:198–203.
    1. Sober S, Org E, Kepp K, Juhanson P, Eyheramendy S, et al. Targeting 160 candidate genes for blood pressure regulation with a genome-wide genotyping array. PLoS One. 2009;4:e6034.
    1. Hirschhorn JN, Daly MJ. Genome-wide association studies for common diseases and complex traits. Nat Rev Genet. 2005;6:95–108.
    1. Hindorff LA, Sethupathy P, Junkins HA, Ramos EM, Mehta JP, et al. Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc Natl Acad Sci U S A. 2009;106:9362–9367.
    1. WTCCC. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature. 2007;447:661–678.
    1. Org E, Eyheramendy S, Juhanson P, Gieger C, Lichtner P, et al. Genome-wide scan identifies CDH13 as a novel susceptibility locus contributing to blood pressure determination in two European populations. Hum Mol Genet. 2009;18:2288–2296.
    1. Wang Y, O'Connell JR, McArdle PF, Wade JB, Dorff SE, et al. From the Cover: Whole-genome association study identifies STK39 as a hypertension susceptibility gene. Proc Natl Acad Sci U S A. 2009;106:226–231.
    1. Newton-Cheh C, Johnson T, Gateva V, Tobin MD, Bochud M, et al. Genome-wide association study identifies eight loci associated with blood pressure. Nat Genet 2009
    1. Levy D, Ehret GB, Rice K, Verwoert GC, Launer LJ, et al. Genome-wide association study of blood pressure and hypertension. Nat Genet 2009
    1. Price AL, Patterson NJ, Plenge RM, Weinblatt ME, Shadick NA, et al. Principal components analysis corrects for stratification in genome-wide association studies. Nat Genet. 2006;38:904–909.
    1. Yu K, Wang Z, Li Q, Wacholder S, Hunter DJ, et al. Population substructure and control selection in genome-wide association studies. PLoS One. 2008;3:e2551.
    1. Pahl R, Schafer H. PERMORY: an LD-exploiting permutation test algorithm for powerful genome-wide association testing. Bioinformatics. 2010;26:2093–2100.
    1. Weber S, Bernhard D, Lukowski R, Weinmeister P, Worner R, et al. Rescue of cGMP kinase I knockout mice by smooth muscle specific expression of either isozyme. Circ Res. 2007;101:1096–1103.
    1. Feil R, Gappa N, Rutz M, Schlossmann J, Rose CR, et al. Functional reconstitution of vascular smooth muscle cells with cGMP-dependent protein kinase I isoforms. Circ Res. 2002;90:1080–1086.
    1. Lytton J. Na+/Ca2+ exchangers: three mammalian gene families control Ca2+ transport. Biochem J. 2007;406:365–382.
    1. Blaustein MP, Zhang J, Chen L, Song H, Raina H, et al. The pump, the exchanger, and endogenous ouabain: signaling mechanisms that link salt retention to hypertension. Hypertension. 2009;53:291–298.
    1. Dong H, Jiang Y, Triggle CR, Li X, Lytton J. Novel role for K+-dependent Na+/Ca2+ exchangers in regulation of cytoplasmic free Ca2+ and contractility in arterial smooth muscle. Am J Physiol Heart Circ Physiol. 2006;291:H1226–1235.
    1. de la Sierra A, Giner V, Bragulat E, Coca A. Lack of correlation between two methods for the assessment of salt sensitivity in essential hypertension. J Hum Hypertens. 2002;16:255–260.
    1. Blaustein MP, Lederer WJ. Sodium/calcium exchange: its physiological implications. Physiol Rev. 1999;79:763–854.
    1. Karaki H, Ozaki H, Hori M, Mitsui-Saito M, Amano K, et al. Calcium movements, distribution, and functions in smooth muscle. Pharmacol Rev. 1997;49:157–230.
    1. Di Rienzo A, Hudson RR. An evolutionary framework for common diseases: the ancestral-susceptibility model. Trends Genet. 2005;21:596–601.
    1. Tamura N, Itoh H, Ogawa Y, Nakagawa O, Harada M, et al. cDNA cloning and gene expression of human type Ialpha cGMP-dependent protein kinase. Hypertension. 1996;27:552–557.
    1. Michael SK, Surks HK, Wang Y, Zhu Y, Blanton R, et al. High blood pressure arising from a defect in vascular function. Proc Natl Acad Sci U S A. 2008;105:6702–6707.
    1. Feil R, Holter SM, Weindl K, Wurst W, Langmesser S, et al. cGMP-dependent protein kinase I, the circadian clock, sleep and learning. Commun Integr Biol. 2009;2:298–301.
    1. Zhao Q, Wang L, Yang W, Chen S, Huang J, et al. Interactions among genetic variants from contractile pathway of vascular smooth muscle cell in essential hypertension susceptibility of Chinese Han population. Pharmacogenet Genomics. 2008;18:459–466.
    1. Pfeifer A, Klatt P, Massberg S, Ny L, Sausbier M, et al. Defective smooth muscle regulation in cGMP kinase I-deficient mice. Embo J. 1998;17:3045–3051.
    1. Black DL. Mechanisms of alternative pre-messenger RNA splicing. Annu Rev Biochem. 2003;72:291–336.
    1. Bourgeois CF, Lejeune F, Stevenin J. Broad specificity of SR (serine/arginine) proteins in the regulation of alternative splicing of pre-messenger RNA. Prog Nucleic Acid Res Mol Biol. 2004;78:37–88.
    1. Lee GS, Choi KC, Jeung EB. K+-dependent Na+/Ca2+ exchanger 3 is involved in renal active calcium transport and is differentially expressed in the mouse kidney. Am J Physiol Renal Physiol. 2009;297:F371–379.
    1. Visser F, Valsecchi V, Annunziato L, Lytton J. Exchangers NCKX2, NCKX3, and NCKX4: identification of Thr-551 as a key residue in defining the apparent K(+) affinity of NCKX2. J Biol Chem. 2007;282:4453–4462.
    1. Iwamoto T, Kita S, Zhang J, Blaustein MP, Arai Y, et al. Salt-sensitive hypertension is triggered by Ca2+ entry via Na+/Ca2+ exchanger type-1 in vascular smooth muscle. Nat Med. 2004;10:1193–1199.
    1. Navis G, Bakker SJ, van der Harst P. Dissecting the genetics of complex traits: lessons from hypertension. Nephrol Dial Transplant. 2010;25:1382–1385.
    1. Citterio L, Lanzani C, Manunta P, Bianchi G. Genetics of primary hypertension: The clinical impact of adducin polymorphisms. Biochim Biophys Acta 2010
    1. McCarthy MI, Hirschhorn JN. Genome-wide association studies: potential next steps on a genetic journey. Hum Mol Genet. 2008;17:R156–165.
    1. Wang WY, Barratt BJ, Clayton DG, Todd JA. Genome-wide association studies: theoretical and practical concerns. Nat Rev Genet. 2005;6:109–118.
    1. Bianchi G. Genetic variations of tubular sodium reabsorption leading to “primary” hypertension: from gene polymorphism to clinical symptoms. Am J Physiol Regul Integr Comp Physiol. 2005;289:R1536–1549.
    1. Manunta P, Cusi D, Barlassina C, Righetti M, Lanzani C, et al. Alpha-adducin polymorphisms and renal sodium handling in essential hypertensive patients. Kidney Int. 1998;53:1471–1478.
    1. Li Y, Abecasis G. Mach 1.0: Rapid Haplotype Reconstruction and Missing Genotype Inference. Am J Hum Genet. 2006;S79:2290.
    1. Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MA, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81:559–575.
    1. Cartegni L, Wang J, Zhu Z, Zhang MQ, Krainer AR. ESEfinder: A web resource to identify exonic splicing enhancers. Nucleic Acids Res. 2003;31:3568–3571.

Source: PubMed

3
Abonnere