Genome-Wide Association Studies of Serum Magnesium, Potassium, and Sodium Concentrations Identify Six Loci Influencing Serum Magnesium Levels

Magnesium, potassium, and sodium, cations commonly measured in serum, are involved in many physiological processes including energy metabolism, nerve and muscle function, signal transduction, and fluid and blood pressure regulation. To evaluate the contribution of common genetic variation to normal physiologic variation in serum concentrations of these cations, we conducted genome-wide association studies of serum magnesium, potassium, and sodium concentrations using ∼2.5 million genotyped and imputed common single nucleotide polymorphisms (SNPs) in 15,366 participants of European descent from the international CHARGE Consortium. Study-specific results were combined using fixed-effects inverse-variance weighted meta-analysis. SNPs demonstrating genome-wide significant (p<5×10−8) or suggestive associations (p<4×10−7) were evaluated for replication in an additional 8,463 subjects of European descent. The association of common variants at six genomic regions (in or near MUC1, ATP2B1, DCDC5, TRPM6, SHROOM3, and MDS1) with serum magnesium levels was genome-wide significant when meta-analyzed with the replication dataset. All initially significant SNPs from the CHARGE Consortium showed nominal association with clinically defined hypomagnesemia, two showed association with kidney function, two with bone mineral density, and one of these also associated with fasting glucose levels. Common variants in CNNM2, a magnesium transporter studied only in model systems to date, as well as in CNNM3 and CNNM4, were also associated with magnesium concentrations in this study. We observed no associations with serum sodium or potassium levels exceeding p<4×10−7. Follow-up studies of newly implicated genomic loci may provide additional insights into the regulation and homeostasis of human serum magnesium levels.

[1]  R. Kleta,et al.  Bartter Syndromes and Other Salt-Losing Tubulopathies , 2006, Nephron Physiology.

[2]  Judy H. Cho,et al.  Finding the missing heritability of complex diseases , 2009, Nature.

[3]  V. Gudnason,et al.  Age, Gene/Environment Susceptibility-Reykjavik Study: multidisciplinary applied phenomics. , 2007, American journal of epidemiology.

[4]  Qiong Yang,et al.  The Third Generation Cohort of the National Heart, Lung, and Blood Institute's Framingham Heart Study: design, recruitment, and initial examination. , 2007, American journal of epidemiology.

[5]  A. Hofman,et al.  Association of three genetic loci with uric acid concentration and risk of gout: a genome-wide association study , 2008, The Lancet.

[6]  Tom Greene,et al.  Calibration and random variation of the serum creatinine assay as critical elements of using equations to estimate glomerular filtration rate. , 2002, American journal of kidney diseases : the official journal of the National Kidney Foundation.

[7]  P. Meneton,et al.  Inactivation of the Na‐Cl Co‐Transporter (NCC) Gene Is Associated With High BMD Through Both Renal and Bone Mechanisms: Analysis of Patients With Gitelman Syndrome and Ncc Null Mice , 2004, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

[8]  Christian Gieger,et al.  New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk , 2010, Nature Genetics.

[9]  A. Folsom,et al.  The Atherosclerosis Risk in Communities (ARIC) Study: design and objectives. The ARIC investigators. , 1989, American journal of epidemiology.

[10]  G. Quamme,et al.  Identification and characterization of a novel family of membrane magnesium transporters, MMgT1 and MMgT2. , 2008, American journal of physiology. Cell physiology.

[11]  R. Swaminathan Magnesium metabolism and its disorders. , 2003, The Clinical biochemist. Reviews.

[12]  D. Günzel,et al.  Targeted deletion of murine Cldn16 identifies extra- and intrarenal compensatory mechanisms of Ca2+ and Mg2+ wasting. , 2010, American journal of physiology. Renal physiology.

[13]  S. Wallach Effects of magnesium on skeletal metabolism. , 1990, Magnesium and trace elements.

[14]  E. Wieben,et al.  Complete primary structure of a human plasma membrane Ca2+ pump. , 1988, The Journal of biological chemistry.

[15]  A. Levey,et al.  A More Accurate Method To Estimate Glomerular Filtration Rate from Serum Creatinine: A New Prediction Equation , 1999, Annals of Internal Medicine.

[16]  Yurii S. Aulchenko,et al.  Multiple loci associated with indices of renal function and chronic kidney disease , 2009, Nature Genetics.

[17]  S. Berg,et al.  Heritabilities for fifteen routine biochemical values: findings in 215 Swedish twin pairs 82 years of age or older , 2009, Scandinavian journal of clinical and laboratory investigation.

[18]  Ian M. Carr,et al.  Mutations in CNNM4 Cause Jalili Syndrome, Consisting of Autosomal-Recessive Cone-Rod Dystrophy and Amelogenesis Imperfecta , 2009, American journal of human genetics.

[19]  H. P. Adamo,et al.  The phosphatase activity of the plasma membrane Ca2+ pump. Activation by acidic lipids in the absence of Ca2+ increases the apparent affinity for Mg2+. , 2007, Biochimica et biophysica acta.

[20]  Andrew D. Johnson,et al.  Genome-wide association study of blood pressure and hypertension , 2009, Nature Genetics.

[21]  K. Nakashima,et al.  [The Rotterdam study]. , 2011, Nihon rinsho. Japanese journal of clinical medicine.

[22]  U. John,et al.  Study of Health in Pomerania (SHIP): A health examination survey in an east German region: Objectives and design , 2005, Sozial- und Präventivmedizin.

[23]  T. Spector,et al.  Genetic contribution to renal function and electrolyte balance: a twin study. , 2002, Clinical science.

[24]  P. Allhoff,et al.  The Framingham Offspring Study , 1991 .

[25]  R. Tyagi,et al.  Characterization of an Evolutionarily Conserved Metallophosphoesterase That Is Expressed in the Fetal Brain and Associated with the WAGR Syndrome* , 2009, Journal of Biological Chemistry.

[26]  M. Devoto,et al.  Estimates of Genetic and Environmental Contribution to 43 Quantitative Traits Support Sharing of a Homogeneous Environment in an Isolated Population from South Tyrol, Italy , 2007, Human Heredity.

[27]  L. Christiansen,et al.  Heritability of biochemical kidney markers and relation to survival in the elderly--results from a Danish population-based twin study. , 2004, Clinica chimica acta; international journal of clinical chemistry.

[28]  W. Kannel,et al.  AN APPROACH TO LONGITUDINAL STUDIES IN A COMMUNITY: THE FRAMINGHAM STUDY , 1963, Annals of the New York Academy of Sciences.

[29]  E. Chouery,et al.  Mutations in CNNM4 cause recessive cone-rod dystrophy with amelogenesis imperfecta. , 2009, American journal of human genetics.

[30]  G. Abecasis,et al.  Heritability of Cardiovascular and Personality Traits in 6,148 Sardinians , 2006, PLoS genetics.

[31]  G. Quamme,et al.  Functional characterization of human SLC41A1, a Mg2+ transporter with similarity to prokaryotic MgtE Mg2+ transporters. , 2005, Physiological genomics.

[32]  T. Gudermann,et al.  TRPM6 and TRPM7--Gatekeepers of human magnesium metabolism. , 2007, Biochimica et biophysica acta.

[33]  G. Quamme,et al.  Functional characterization of ACDP2 (ancient conserved domain protein), a divalent metal transporter. , 2005, Physiological genomics.

[34]  Thomas Meitinger,et al.  A meta-analysis of genome-wide data from five European isolates reveals an association of COL22A1, SYT1, and GABRR2 with serum creatinine level , 2010, BMC Medical Genetics.

[35]  Yurii S. Aulchenko,et al.  BIOINFORMATICS APPLICATIONS NOTE doi:10.1093/bioinformatics/btm108 Genetics and population analysis GenABEL: an R library for genome-wide association analysis , 2022 .

[36]  D. Cole,et al.  Inherited disorders of renal magnesium handling. , 2000, Journal of the American Society of Nephrology : JASN.

[37]  G. Quamme Molecular identification of ancient and modern mammalian magnesium transporters. , 2010, American journal of physiology. Cell physiology.

[38]  W. Kannel,et al.  The association of total cholesterol, triglycerides and plasma lipoprotein cholesterol levels in first degree relatives and spouse pairs. , 1979, American journal of epidemiology.

[39]  Yurii S. Aulchenko,et al.  Twenty bone mineral density loci identified by large-scale meta-analysis of genome-wide association studies , 2009, Nature Genetics.

[40]  A. Hofman,et al.  Reduction of blood pressure with oral magnesium supplementation in women with mild to moderate hypertension. , 1994, The American journal of clinical nutrition.

[41]  E. Haddad,et al.  Hypomagnesemia with secondary hypocalcemia is caused by mutations in TRPM6, a new member of the TRPM gene family , 2002, Nature Genetics.

[42]  K. Lunetta,et al.  Methods in Genetics and Clinical Interpretation Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium Design of Prospective Meta-Analyses of Genome-Wide Association Studies From 5 Cohorts , 2010 .

[43]  J. She,et al.  Molecular cloning and characterization of a novel gene family of four ancient conserved domain proteins (ACDP). , 2003, Gene.

[44]  C. Gieger,et al.  KORA-gen - Resource for Population Genetics, Controls and a Broad Spectrum of Disease Phenotypes , 2005, Gesundheitswesen (Bundesverband der Arzte des Offentlichen Gesundheitsdienstes (Germany)).

[45]  P. Donnelly,et al.  A new multipoint method for genome-wide association studies by imputation of genotypes , 2007, Nature Genetics.

[46]  Monique M. B. Breteler,et al.  The Rotterdam Study: 2016 objectives and design update , 2015, European Journal of Epidemiology.

[47]  V. Sheffield,et al.  Mutation of TRPM6 causes familial hypomagnesemia with secondary hypocalcemia , 2002, Nature Genetics.

[48]  R. Reilly,et al.  Hereditary etiologies of hypomagnesemia , 2008, Nature Clinical Practice Nephrology.

[49]  L. Almasy,et al.  Multipoint quantitative-trait linkage analysis in general pedigrees. , 1998, American journal of human genetics.

[50]  J. Šamaj,et al.  Mrs2p is an essential component of the major electrophoretic Mg2+ influx system in mitochondria , 2003, The EMBO journal.

[51]  S. Harrap,et al.  Antihypertensive Treatments Obscure Familial Contributions to Blood Pressure Variation , 2003, Hypertension.

[52]  M. Stephens,et al.  Imputation-Based Analysis of Association Studies: Candidate Regions and Quantitative Traits , 2007, PLoS genetics.

[53]  R. Kronmal,et al.  The Cardiovascular Health Study: design and rationale. , 1991, Annals of epidemiology.

[54]  G. Quamme,et al.  Functional characterization of NIPA2, a selective Mg2+ transporter. , 2008, American journal of physiology. Cell physiology.

[55]  W. Kannel,et al.  The Framingham Offspring Study. Design and preliminary data. , 1975, Preventive medicine.

[56]  D. Cole,et al.  Genetic determinants of extracellular magnesium concentration: analysis of multiple candidate genes, and evidence for association with the estrogen receptor alpha (ESR1) locus. , 2009, Clinica chimica acta; international journal of clinical chemistry.

[57]  A. Folsom,et al.  Serum and dietary magnesium and the risk for type 2 diabetes mellitus: the Atherosclerosis Risk in Communities Study. , 1999, Archives of internal medicine.

[58]  J. Hoenderop,et al.  Molecular determinants of magnesium homeostasis: insights from human disease. , 2008, Journal of the American Society of Nephrology : JASN.