ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Hypomagnesemia: Causes of hypomagnesemia

Hypomagnesemia: Causes of hypomagnesemia
Literature review current through: Jan 2024.
This topic last updated: Jan 12, 2022.

INTRODUCTION — Hypomagnesemia is a common entity occurring in up to 12 percent of hospitalized patients [1]. The incidence rises to as high as 60 to 65 percent in patients in an intensive care setting in which nutrition, diuretics, hypoalbuminemia, and aminoglycosides may play important roles [2-5]. In addition, serum magnesium concentration is a quantitative trait with substantial polygenic heritability, and a number of genome-wide association studies (GWAS) have identified candidate loci associated with low serum magnesium levels [6-8].

There are two major mechanisms by which hypomagnesemia can be induced: gastrointestinal or renal losses (table 1). Regardless of the cause, hypomagnesemia begins to occur after a relatively small magnesium deficit because there is little rapid exchange of extracellular magnesium with the much larger bone and cell stores.

Hypomagnesemia is often associated with hypokalemia (due to urinary potassium wasting) and hypocalcemia (due both to lower parathyroid hormone secretion and end-organ resistance to its effect). (See "Hypomagnesemia: Clinical manifestations of magnesium depletion".)

The major causes of hypomagnesemia will be reviewed in this topic. The regulation of magnesium balance, the signs and symptoms of hypomagnesemia, and the evaluation and treatment of patients with hypomagnesemia are presented elsewhere:

(See "Regulation of magnesium balance".)

(See "Hypomagnesemia: Clinical manifestations of magnesium depletion".)

(See "Hypomagnesemia: Evaluation and treatment".)

GASTROINTESTINAL LOSSES — Gastrointestinal secretions contain some magnesium, and potential losses are continuous and not regulated. Although the obligatory losses are not large, marked dietary deprivation can lead to progressive magnesium depletion.

Magnesium losses from both the upper and lower gastrointestinal tract can induce hypomagnesemia. In general, magnesium depletion is more commonly due to diarrhea than to vomiting [5]. This is because the magnesium content of lower tract secretions is significantly higher (up to 15 mEq/L versus approximately 1 mEq/L for upper tract). Common settings in which hypomagnesemia may be seen include acute or chronic diarrhea, malabsorption and steatorrhea, and small bowel bypass surgery.

In addition, hypomagnesemia with secondary hypocalcemia can be produced by a familial disorder characterized by a selective defect in magnesium absorption (primary intestinal hypomagnesemia). This disease presents in the neonatal period with hypocalcemia that is responsive to magnesium administration [9]. In some instances, the defect appears to have an X-linked recessive inheritance, but others have described autosomally recessive inheritance with linkage to chromosome 9 [10]. The autosomal recessive form is caused by mutations in the TRPM6 gene [11-15]. The encoded protein, which is expressed both by intestinal epithelia and by the renal distal convoluted tubule, functions as an apical magnesium entry channel [16]. Thus, patients with this disorder have not only decreased intestinal magnesium absorption, but also inappropriate renal magnesium wasting.

Hypomagnesemia can also be seen in acute pancreatitis. The mechanism of hypomagnesemia is presumably similar to the mechanism partially responsible for hypocalcemia in acute pancreatitis: saponification of magnesium and calcium in necrotic fat [17]. The degree of hypocalcemia may be exacerbated by the hypomagnesemia, which can both lower parathyroid hormone secretion and induce end-organ resistance to its effect. (See "Etiology of hypocalcemia in adults" and "Hypomagnesemia: Clinical manifestations of magnesium depletion".)

Proton pump inhibitors — Hypomagnesemia, usually with hypocalcemia, has been described in case reports with the chronic use of omeprazole (usually for more than one year) and other proton pump inhibitors (PPIs) [18-21]. The association of PPIs with lower serum magnesium has also been described in population studies [22-25]. The best data come from a large cohort of 11,490 patients admitted to the intensive care unit at a single center [22]. In this study, the relationship between PPI use and magnesium varied by whether patients concurrently used diuretics (see "Effect of diuretics on magnesium handling by the kidney"):

In patients taking diuretics, concurrent use of PPIs was associated with a 0.028 mg/dL (0.011 mmol/L) lower adjusted serum magnesium. In addition, the prevalence of hypomagnesemia (defined as a serum magnesium less than 1.6 mg/dL [0.66 mmol/L]) was significantly higher in patients taking both drugs as compared with those who only used diuretics (15.6 versus 11 percent).

In patients not taking diuretics, use of PPIs was not associated with the serum magnesium or the prevalence of hypomagnesemia.

In a smaller study of 402 inpatients with hypomagnesemia and matched controls, proton pump inhibitors were not associated with hypomagnesemia [23]. However, duration of use was not examined, and most patients were not taking diuretics; therefore, these findings are potentially consistent with those of the larger cohort study. A meta-analysis of studies through 2014 reported a pooled odds ratio for PPI-associated hypomagnesemia of 1.78 (95% CI 1.08-2.92), although there was substantial heterogeneity among the included studies [24].

The presumed mechanism is impaired absorption of magnesium by intestinal epithelial cells caused by PPI-induced inhibition of transient receptor potential melastatin-6 (TRPM6) and TRPM7 channels [26]. Renal losses are not likely to be involved, since urinary magnesium excretion is appropriately low in patients with hypomagnesemia due to PPIs [20,21]. Some patients have inappropriately low serum parathyroid hormone levels [19], a finding that is well described in patients with hypomagnesemia due to other causes. (See "Hypomagnesemia: Clinical manifestations of magnesium depletion", section on 'Calcium metabolism'.)

In March 2011, the United States Food and Drug Administration (FDA) issued a safety warning suggesting that, in patients expected to be on PPIs for long periods of time and in those taking other medications associated with hypomagnesemia as described below (eg, diuretics), providers measure serum magnesium levels prior to initiation of PPI therapy and periodically during treatment [27]. The hypomagnesemia can be partially or completely corrected by high-dose oral magnesium supplementation [19-21]. Hypomagnesemia resolves with cessation of PPI therapy [18,19,21].

RENAL LOSSES — Urinary magnesium losses can occur via a variety of either acquired or intrinsic mechanisms. (See "Regulation of magnesium balance".)

Medications — Both loop and thiazide diuretics can inhibit net magnesium reabsorption, while potassium-sparing diuretics may enhance magnesium transport and lower magnesium excretion. The degree of hypomagnesemia induced by loop and thiazide diuretics is generally mild, in part because the associated volume contraction will tend to increase proximal sodium, water, and magnesium reabsorption. A more detailed review of how these changes might occur is discussed separately. (See "Effect of diuretics on magnesium handling by the kidney".)

In addition to loop and thiazide diuretics, many nephrotoxic drugs can produce urinary magnesium wasting [28], including but not limited to:

Aminoglycoside antibiotics (see "Aminoglycosides", section on 'Nephrotoxicity')

Amphotericin B (see "Amphotericin B nephrotoxicity")

Cisplatin (see "Cisplatin nephrotoxicity")

Pentamidine (see "Overview of kidney disease in patients with HIV", section on 'Electrolyte disorders')

Calcineurin inhibitors (see "Cyclosporine and tacrolimus nephrotoxicity" and 'Posttransplant patients' below)

Antibodies targeting the epidermal growth factor (EGF) receptor (cetuximab, panitumumab) (see "Nephrotoxicity of molecularly targeted agents and immunotherapy", section on 'Anti-EGFR monoclonal antibodies')

Digoxin [29]

The impairment in loop and distal magnesium reabsorption may occur prior to the onset of, and may persist after the resolution of, the acute tubular necrosis and acute kidney injury (AKI) associated with these nephrotoxins. Studies in the rat, for example, have shown a dose-related and rapidly reversible decrease in renal tubular reabsorption of magnesium and calcium (but not sodium and potassium) within 60 minutes of beginning an infusion of gentamicin [30]. The hypermagnesuria in this setting can be striking, and the resulting hypomagnesemia may be sufficient to produce hypocalcemia. With cisplatin therapy, a reduction in gastrointestinal magnesium absorption may also contribute to the hypomagnesemia [31].

Volume expansion — Expansion of the extracellular fluid volume, by reducing reabsorption of sodium and water, can decrease passive magnesium transport. Mild hypomagnesemia may ensue if this is sustained, as in primary aldosteronism. (See "Pathophysiology and clinical features of primary aldosteronism".)

Alcohol — Hypomagnesemia is common in patients with an alcohol use disorder who are admitted to the hospital; in one study, for example, the prevalence was 30 percent [32]. Excessive urinary excretion of magnesium occurred in 18 of the 38 patients with hypomagnesemia. The defect in urinary excretion appears to reflect alcohol-induced tubular dysfunction that is reversible within four weeks of abstinence [33]. This effect is relatively modest, and other factors are also thought to contribute to hypomagnesemia in these patients, including dietary deficiency, acute pancreatitis, and diarrhea.

Uncontrolled diabetes mellitus — Hypomagnesemia is not uncommon in patients with uncontrolled diabetes mellitus; it appears to be associated with increased urinary magnesium excretion that is reversed by correction of the hyperglycemia with insulin [5,34,35]. It has been proposed, although not proven, that hypomagnesemia may impair glucose disposal and may play a role in the pathogenesis of some of the complications of diabetes.

Posttransplant patients — Hypomagnesemia is common in patients who have received solid organ transplants and may be an important risk factor for new-onset diabetes mellitus after transplantation [36-38].

Hypomagnesemia after transplantation is attributed to renal magnesium wasting [36] and is primarily caused by treatment with calcineurin inhibitors, which are known to downregulate TRPM6 [39]. These drugs may also increase claudin-14 expression, which would inhibit paracellular magnesium transport [40]. (See "Regulation of magnesium balance".)

The incidence of hypomagnesemia after kidney transplantation in patients treated with tacrolimus may be as high as 43 percent [41], which is considerably higher than in patients treated with cyclosporine [42]. In such patients who are converted from a calcineurin inhibitor to a mammalian (mechanistic) target of rapamycin (mTOR) inhibitor, serum magnesium levels increase by approximately 0.2 mg/dL, with a concomitant decrease in the fractional excretion of magnesium [43].

Hypercalcemia — Patients with hypercalcemia due, for example, to hyperparathyroidism can develop mild hypomagnesemia. This finding results at least in part from the fact that calcium and magnesium functionally compete for transport in the thick ascending limb of the loop of Henle. A variety of mechanisms may contribute to the reduction in magnesium reabsorption resulting from hypercalcemia. One well-established mechanism is mediated by the basolateral calcium-sensing receptor (CaSR) in the thick ascending limb:

Calcium binds to the basolateral CaSR.

This leads to generation of prostaglandins and cytochrome P450 metabolites in the cell, which inhibit the apical potassium channel (ROMK) [44,45].

Inhibition of ROMK inhibits sodium chloride reabsorption in the thick ascending limb and reduces paracellular magnesium and calcium reabsorption (figure 1). Inhibition of ROMK in this nephron segment resulting from stimulation of the CaSR is the mechanism underlying type V Bartter syndrome. Thus, hypercalcemia can produce a Bartter-like phenotype. (See "Inherited hypokalemic salt-losing tubulopathies: Pathophysiology and overview of clinical manifestations", section on 'Clinical manifestations'.)

In addition, stimulation of the CaSR decreases paracellular permeability of the thick ascending limb to both magnesium and calcium [46], likely due to increased expression of claudin-14 [47,48], which negatively regulates claudin-16 and claudin-19. (See 'Familial hypomagnesemia with hypercalciuria and nephrocalcinosis (FHHNC)' below.)

Other acquired tubular dysfunction — Magnesium wasting can be seen as part of the tubular dysfunction associated with recovery from acute tubular necrosis, following kidney transplantation, or during a postobstructive diuresis.

Familial renal magnesium wasting — Primary renal magnesium wasting is an unusual disorder that may present sporadically or as a familial disease [49-53]. Several types have been recognized [54].

The diagnosis of primary renal magnesium wasting is one of exclusion. It is established by the demonstration of inappropriately high urinary magnesium excretion in the absence of any other apparent cause. (See "Hypomagnesemia: Evaluation and treatment".)

Gitelman and Bartter syndrome — Gitelman syndrome is the most common form of familial renal magnesium wasting and is usually associated with salt wasting, hypokalemic metabolic alkalosis, and hypocalciuria. It is caused by recessive mutations in the gene coding for the thiazide-sensitive sodium chloride cotransporter (SLC12A3). Although the hypokalemia in this syndrome is usually attributed to decreased sodium chloride transport, direct effects of hypomagnesemia may also contribute [55]. The hypomagnesemia in Gitelman syndrome is significantly more marked than that observed with thiazide administration. Bartter syndrome, particularly the late-onset cases due to mutations in CLCNKB (Bartter syndrome type 3) that resemble Gitelman syndrome, may also present with hypomagnesemia due to defective reabsorption in the thick ascending limb [56]. (See "Inherited hypokalemic salt-losing tubulopathies: Pathophysiology and overview of clinical manifestations".)

EAST (SeSAME) syndrome — EAST syndrome (also known as SeSAME syndrome) is an autosomal recessive disorder that presents in infancy with epilepsy, ataxia, sensorineural deafness, intellectual disability, and a renal salt-losing tubulopathy that resembles Gitelman syndrome and manifests as hypokalemic metabolic alkalosis with hypomagnesemia [57,58]. This syndrome is caused by loss-of-function mutations in the gene encoding the potassium channel, KCNJ10 (Kir4.1), which is expressed on the basolateral membrane of the distal convoluted tubule and regulates expression of the thiazide-sensitive sodium chloride cotransporter [59].

Familial hypomagnesemia with hypercalciuria and nephrocalcinosis (FHHNC) — This form of primary renal magnesium wasting is autosomal recessive and associated with hypercalciuria; affected patients usually present in childhood or adolescence with symptomatic hypocalcemia [60-64]. Recurrent nephrolithiasis and nephrocalcinosis are also seen, and progression to kidney function impairment and an acidification defect are common. The problem with acidification has been attributed to defective ammonia transfer to the deep nephrons and impaired medullary hydrogen ion secretion due to nephrocalcinosis [65]. Affected patients may also have polyuria and polydipsia due to nephrogenic diabetes insipidus [60,64].

Mutations in the claudin-16 gene (also known as paracellin-1) are the major cause of FHHNC [15,64,66-70]. Claudin-16 is a tight junction protein that facilitates the passive, paracellular reabsorption of both magnesium and calcium in the thick ascending limb of the loop of Henle. Some of these patients present with hypokalemia due to either secondary hyperaldosteronism and/or direct effects of hypomagnesemia on potassium transport.

FHHNC may occasionally be due to mutations in claudin-19 [71]. In addition to hypomagnesemia, nephrolithiasis, and nephrocalcinosis, such patients have ocular involvement [72], which may include macular colobomata, nystagmus, and myopia.

Na-K-ATPase mutations — A gene responsible for autosomal dominant isolated renal magnesium wasting in two unrelated Dutch families has been mapped to a region on chromosome 11q23 [73]. The abnormality in this disorder appears to be a dominant negative mutation in the gene encoding the gamma subunit of Na-K-ATPase, resulting in misrouting of the protein [74,75]. This may mimic the magnesuric effects of digoxin. In addition, de novo heterozygous loss-of-function mutations in the ATP1A1 gene, which encodes the a1 subunit of the Na-K-ATPase, have been described in three infants with severe hypomagnesemia due to renal magnesium wasting, refractory seizures, and intellectual disability [76]. (See 'Medications' above.)

Voltage-gated potassium channel — A mutation in the gene (KCNA1) that encodes the voltage-gated potassium channel (Kv1.1) is the cause of isolated autosomal dominant hypomagnesemia discovered in a large Brazilian family [77]. This channel colocalizes with the magnesium channel TRPM6 in the distal collecting tubules. Wild type Kv1.1 appears to control TRPM6 magnesium reabsorption via the creation of an appropriate potential across the luminal membrane.

Hepatocyte nuclear factor-1-beta gene mutations — Hypomagnesemia and renal magnesium wasting have been reported in children with known mutations in the hepatocyte nuclear factor-1-beta (HNF-1-beta) gene [78]. The prevalence of hypomagnesemia in such patients increases with age (100 percent by age 13.5 to 18 years) and is associated with a more generalized dysfunction of the distal convoluted tubule that resembles Gitelman syndrome [79]. HNF-1-beta is a transcription factor that regulates the expression of the gamma subunit of Na-K-ATPase and the potassium channel Kir5.1 [80]. These findings suggest that some inactivating mutations of HNF-1-beta can inhibit the Na-K-ATPase and Kir5.1, thereby causing hypomagnesemia/Gitelman-like syndrome, in association with early-onset diabetes and kidney malformations that include multicystic dysplastic disease and obstructive uropathy. (See "Classification of diabetes mellitus and genetic diabetic syndromes".)

Epidermal growth factor gene mutation — Epidermal growth factor (EGF) appears to stimulate distal tubular magnesium reabsorption by binding to a receptor on the basolateral membrane and activating the magnesium channel TRPM6 in the apical membrane. A point mutation in pro-EGF leads to impaired basolateral membrane sorting, inadequate stimulation of renal EGF receptor, insufficient activation of the magnesium channel, and causes isolated autosomal recessive renal magnesium wasting [81,82]. A similar inhibition of the EGF receptor likely underlies the effects of cetuximab and other EGF receptor inhibitors on magnesium transport as previously mentioned [81,82]. (See 'Medications' above.)

PCBD1 mutation — PCBD1 (pterin-4 alpha-carbinolamine dehydratase/dimerization cofactor of hepatocyte nuclear factor-1 homeobox A) is a protein that acts as an enzyme to regenerate tetrahydrobiopterin, which is a cofactor for phenylalanine hydroxylase. Homozygous mutations in PCBD1 in neonates causes transient hyperphenylalaninemia. In addition, PCBD1 acts as a dimerization cofactor for the transcription factor, hepatocyte nuclear factor-1-beta (HNF1B). Homozygous mutations in PCBD1 in adults have been reported to be associated with hypomagnesemia, renal magnesium wasting, and maturity-onset diabetes of the young, mimicking the effect of HNF1B mutations [83]. (See "Classification of diabetes mellitus and genetic diabetic syndromes", section on 'Hepatocyte nuclear factor-1-beta'.)

Cyclin M2 mutations — Mutations in the cyclin M2 (CNNM2) gene have been implicated in two families with dominant isolated renal magnesium wasting [84]. CNNM2 encodes a transmembrane protein that is localized to the basolateral membrane of the thick ascending limb and distal convoluted tubule; it functions either as a magnesium channel [85] or magnesium-sensitive sodium channel and is upregulated under conditions of magnesium deficiency.

MISCELLANEOUS — Hypomagnesemia has been described in a number of other settings. The following is a partial list of potential causes:

Intravascular chelation can lead to hypomagnesemia and has been described in the following settings:

Following surgery, at least in part due to chelation by circulating free fatty acids [86]

After foscarnet therapy of cytomegalovirus chorioretinitis, often in association with hypocalcemia [87]

Ionized hypomagnesemia during liver transplantation due to transfusion of citrate-rich blood products in the absence of adequate hepatic function to metabolize the citrate [88]

Hypomagnesemia can occur as part of the "hungry bone" syndrome in which there is increased magnesium uptake by renewing bone following parathyroidectomy for hyperparathyroidism, thyroidectomy for hyperthyroidism, or correction of severe metabolic acidosis [89]. (See "Hungry bone syndrome following parathyroidectomy in patients with end-stage kidney disease".)

Hypomagnesemia has been found in up to 10 percent of patients placed on an extremely high-fat diet to induce ketogenesis as the therapy of intractable epilepsy. The mechanism of this effect is not clear but may result from malabsorption of magnesium [90]. (See "Evaluation and management of drug-resistant epilepsy".)

Hypomagnesemia due solely to dietary deprivation is exceedingly rare, since nearly all foods contain some magnesium, and renal adaptation to conserve magnesium is highly efficient. Magnesium deficiency has been described rarely with protein-calorie malnutrition [91]. In addition, it may occur in patients with an alcohol use disorder (see 'Alcohol' above), in whom decreased food intake in combination with renal magnesium losses seems to be responsible. Hypomagnesemia is also observed in patients receiving parenteral nutrition, likely due to refeeding syndrome [92].

Hypomagnesemia, in combination with hypertension and dyslipidemia, was described in a kindred with a mutation in mitochondrial tRNA [93]. The mutation likely resulted in multiple and varied physiologic disruptions because of its effect on energy consumption. With respect to low magnesium levels, distal convoluted tubule cells could be adversely affected because magnesium reabsorption requires significant energy.

Hypomagnesemia has been associated with leptospirosis and may be due at least in part to urinary magnesium wasting [94,95].

SUMMARY

General principles – Hypomagnesemia is a common entity occurring in up to 12 percent of hospitalized patients. The incidence rises to as high as 60 to 65 percent in patients in an intensive care setting. There are two major mechanisms by which hypomagnesemia can be induced: gastrointestinal or renal losses (table 1). (See 'Introduction' above.)

Gastrointestinal losses – Gastrointestinal secretions contain some magnesium. Although the obligatory losses are not large, marked dietary deprivation can lead to progressive magnesium depletion. Magnesium losses from both the upper and lower gastrointestinal tract can induce hypomagnesemia. Common settings in which hypomagnesemia may be seen include acute or chronic diarrhea, malabsorption and steatorrhea, and small bowel bypass surgery. Hypomagnesemia can also be seen in acute pancreatitis. (See 'Gastrointestinal losses' above.)

Proton pump inhibitor use – Hypomagnesemia has been described with the chronic use of proton pump inhibitors (PPIs), usually for more than one year, and is due to impaired intestinal absorption. The United States Food and Drug Administration (FDA) issued a safety warning suggesting that, in patients expected to be on these drugs for long periods of time and in those taking other medications associated with hypomagnesemia (eg, diuretics), providers should measure serum magnesium levels prior to initiation of therapy and periodically during treatment. (See 'Proton pump inhibitors' above.)

Renal losses – Urinary magnesium losses can occur via a variety of either acquired or intrinsic mechanisms. These include:

Medications, including loop and thiazide diuretics and various potential nephrotoxins (see 'Medications' above)

Sustained expansion of the extracellular fluid volume, as with primary aldosteronism (see 'Volume expansion' above)

Alcohol use (see 'Alcohol' above)

Uncontrolled diabetes mellitus (see 'Uncontrolled diabetes mellitus' above)

Hypercalcemia, as in patients with primary hyperparathyroidism (see 'Hypercalcemia' above)

Recovery from acute kidney injury (AKI) and following kidney transplantation (see 'Other acquired tubular dysfunction' above)

Familial renal magnesium wasting, such as with Gitelman syndrome (see 'Familial renal magnesium wasting' above)

ACKNOWLEDGMENTS — The UpToDate editorial staff acknowledges Zalman Agus, MD, and Gunjeet K Kala Ahluwalia, MD, who contributed to an earlier version of this topic review.

  1. Wong ET, Rude RK, Singer FR, Shaw ST Jr. A high prevalence of hypomagnesemia and hypermagnesemia in hospitalized patients. Am J Clin Pathol 1983; 79:348.
  2. Chernow B, Bamberger S, Stoiko M, et al. Hypomagnesemia in patients in postoperative intensive care. Chest 1989; 95:391.
  3. Desai TK, Carlson RW, Geheb MA. Prevalence and clinical implications of hypocalcemia in acutely ill patients in a medical intensive care setting. Am J Med 1988; 84:209.
  4. Ryzen E. Magnesium homeostasis in critically ill patients. Magnesium 1989; 8:201.
  5. Tong GM, Rude RK. Magnesium deficiency in critical illness. J Intensive Care Med 2005; 20:3.
  6. Meyer TE, Verwoert GC, Hwang SJ, et al. Genome-wide association studies of serum magnesium, potassium, and sodium concentrations identify six Loci influencing serum magnesium levels. PLoS Genet 2010; 6.
  7. Chang X, Glessner J, Tin A, et al. Genome-wide association study reveals two loci for serum magnesium concentrations in European-American children. Sci Rep 2015; 5:18792.
  8. Tin A, Köttgen A, Folsom AR, et al. Genetic loci for serum magnesium among African-Americans and gene-environment interaction at MUC1 and TRPM6 in European-Americans: the Atherosclerosis Risk in Communities (ARIC) study. BMC Genet 2015; 16:56.
  9. Paunier L, Radde IC, Kooh SW, et al. Primary hypomagnesemia with secondary hypocalcemia in an infant. Pediatrics 1968; 41:385.
  10. Walder RY, Shalev H, Brennan TM, et al. Familial hypomagnesemia maps to chromosome 9q, not to the X chromosome: genetic linkage mapping and analysis of a balanced translocation breakpoint. Hum Mol Genet 1997; 6:1491.
  11. Schlingmann KP, Weber S, Peters M, et al. Hypomagnesemia with secondary hypocalcemia is caused by mutations in TRPM6, a new member of the TRPM gene family. Nat Genet 2002; 31:166.
  12. Walder RY, Landau D, Meyer P, et al. Mutation of TRPM6 causes familial hypomagnesemia with secondary hypocalcemia. Nat Genet 2002; 31:171.
  13. Huang CL. The transient receptor potential superfamily of ion channels. J Am Soc Nephrol 2004; 15:1690.
  14. Schlingmann KP, Sassen MC, Weber S, et al. Novel TRPM6 mutations in 21 families with primary hypomagnesemia and secondary hypocalcemia. J Am Soc Nephrol 2005; 16:3061.
  15. Guran T, Akcay T, Bereket A, et al. Clinical and molecular characterization of Turkish patients with familial hypomagnesaemia: novel mutations in TRPM6 and CLDN16 genes. Nephrol Dial Transplant 2012; 27:667.
  16. Voets T, Nilius B, Hoefs S, et al. TRPM6 forms the Mg2+ influx channel involved in intestinal and renal Mg2+ absorption. J Biol Chem 2004; 279:19.
  17. Ryzen E, Rude RK. Low intracellular magnesium in patients with acute pancreatitis and hypocalcemia. West J Med 1990; 152:145.
  18. Hess MW, Hoenderop JG, Bindels RJ, Drenth JP. Systematic review: hypomagnesaemia induced by proton pump inhibition. Aliment Pharmacol Ther 2012; 36:405.
  19. Epstein M, McGrath S, Law F. Proton-pump inhibitors and hypomagnesemic hypoparathyroidism. N Engl J Med 2006; 355:1834.
  20. Broeren MA, Geerdink EA, Vader HL, van den Wall Bake AW. Hypomagnesemia induced by several proton-pump inhibitors. Ann Intern Med 2009; 151:755.
  21. Cundy T, Dissanayake A. Severe hypomagnesaemia in long-term users of proton-pump inhibitors. Clin Endocrinol (Oxf) 2008; 69:338.
  22. Danziger J, William JH, Scott DJ, et al. Proton-pump inhibitor use is associated with low serum magnesium concentrations. Kidney Int 2013; 83:692.
  23. Koulouridis I, Alfayez M, Tighiouart H, et al. Out-of-hospital use of proton pump inhibitors and hypomagnesemia at hospital admission: a nested case-control study. Am J Kidney Dis 2013; 62:730.
  24. Park CH, Kim EH, Roh YH, et al. The association between the use of proton pump inhibitors and the risk of hypomagnesemia: a systematic review and meta-analysis. PLoS One 2014; 9:e112558.
  25. Zipursky J, Macdonald EM, Hollands S, et al. Proton pump inhibitors and hospitalization with hypomagnesemia: a population-based case-control study. PLoS Med 2014; 11:e1001736.
  26. Perazella MA. Proton pump inhibitors and hypomagnesemia: a rare but serious complication. Kidney Int 2013; 83:553.
  27. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm245275.htm (Accessed on March 08, 2011).
  28. Shah GM, Kirschenbaum MA. Renal magnesium wasting associated with therapeutic agents. Miner Electrolyte Metab 1991; 17:58.
  29. Abu-Amer N, Priel E, Karlish SJD, et al. Hypermagnesuria in Humans Following Acute Intravenous Administration of Digoxin. Nephron 2018; 138:113.
  30. Foster JE, Harpur ES, Garland HO. An investigation of the acute effect of gentamicin on the renal handling of electrolytes in the rat. J Pharmacol Exp Ther 1992; 261:38.
  31. Lajer H, Kristensen M, Hansen HH, et al. Magnesium and potassium homeostasis during cisplatin treatment. Cancer Chemother Pharmacol 2005; 55:231.
  32. Elisaf M, Merkouropoulos M, Tsianos EV, Siamopoulos KC. Pathogenetic mechanisms of hypomagnesemia in alcoholic patients. J Trace Elem Med Biol 1995; 9:210.
  33. De Marchi S, Cecchin E, Basile A, et al. Renal tubular dysfunction in chronic alcohol abuse--effects of abstinence. N Engl J Med 1993; 329:1927.
  34. Tosiello L. Hypomagnesemia and diabetes mellitus. A review of clinical implications. Arch Intern Med 1996; 156:1143.
  35. White JR Jr, Campbell RK. Magnesium and diabetes: a review. Ann Pharmacother 1993; 27:775.
  36. Ramos EL, Barri YM, Kubilis P, et al. Hypomagnesemia in renal transplant patients: improvement over time and association with hypertension and cyclosporine levels. Clin Transplant 1995; 9:185.
  37. Van Laecke S, Van Biesen W, Verbeke F, et al. Posttransplantation hypomagnesemia and its relation with immunosuppression as predictors of new-onset diabetes after transplantation. Am J Transplant 2009; 9:2140.
  38. Huang JW, Famure O, Li Y, Kim SJ. Hypomagnesemia and the Risk of New-Onset Diabetes Mellitus after Kidney Transplantation. J Am Soc Nephrol 2016; 27:1793.
  39. Nijenhuis T, Hoenderop JG, Bindels RJ. Downregulation of Ca(2+) and Mg(2+) transport proteins in the kidney explains tacrolimus (FK506)-induced hypercalciuria and hypomagnesemia. J Am Soc Nephrol 2004; 15:549.
  40. Gong Y, Hou J. Claudin-14 underlies Ca⁺⁺-sensing receptor-mediated Ca⁺⁺ metabolism via NFAT-microRNA-based mechanisms. J Am Soc Nephrol 2014; 25:745.
  41. Navaneethan SD, Sankarasubbaiyan S, Gross MD, et al. Tacrolimus-associated hypomagnesemia in renal transplant recipients. Transplant Proc 2006; 38:1320.
  42. Margreiter R, European Tacrolimus vs Ciclosporin Microemulsion Renal Transplantation Study Group. Efficacy and safety of tacrolimus compared with ciclosporin microemulsion in renal transplantation: a randomised multicentre study. Lancet 2002; 359:741.
  43. Sánchez-Fructuoso AI, Santín Cantero JM, Pérez Flores I, et al. Changes in magnesium and potassium homeostasis after conversion from a calcineurin inhibitor regimen to an mTOR inhibitor-based regimen. Transplant Proc 2010; 42:3047.
  44. Wang WH, Lu M, Hebert SC. Cytochrome P-450 metabolites mediate extracellular Ca(2+)-induced inhibition of apical K+ channels in the TAL. Am J Physiol 1996; 271:C103.
  45. Wang W, Lu M, Balazy M, Hebert SC. Phospholipase A2 is involved in mediating the effect of extracellular Ca2+ on apical K+ channels in rat TAL. Am J Physiol 1997; 273:F421.
  46. Loupy A, Ramakrishnan SK, Wootla B, et al. PTH-independent regulation of blood calcium concentration by the calcium-sensing receptor. J Clin Invest 2012; 122:3355.
  47. Gong Y, Renigunta V, Himmerkus N, et al. Claudin-14 regulates renal Ca⁺⁺ transport in response to CaSR signalling via a novel microRNA pathway. EMBO J 2012; 31:1999.
  48. Dimke H, Desai P, Borovac J, et al. Activation of the Ca(2+)-sensing receptor increases renal claudin-14 expression and urinary Ca(2+) excretion. Am J Physiol Renal Physiol 2013; 304:F761.
  49. Evans RA, Carter JN, George CR, et al. The congenital "magnesium-losing kidney". Report of two patients. Q J Med 1981; 50:39.
  50. Geven WB, Monnens LA, Willems JL. Magnesium metabolism in childhood. Miner Electrolyte Metab 1993; 19:308.
  51. Freeman RM, Pearson E. Hypomagnesemia of unknown etiology. Am J Med 1966; 41:645.
  52. Runeberg L, Collan Y, Jokinen EJ, et al. Hypomagnesemia due to renal disease of unknown etiology. Am J Med 1975; 59:873.
  53. Booth BE, Johanson A. Hypomagnesemia due to renal tubular defect in reabsorption of magnesium. J Pediatr 1974; 85:350.
  54. Konrad M, Weber S. Recent advances in molecular genetics of hereditary magnesium-losing disorders. J Am Soc Nephrol 2003; 14:249.
  55. Kamel KS, Harvey E, Douek K, et al. Studies on the pathogenesis of hypokalemia in Gitelman's syndrome: role of bicarbonaturia and hypomagnesemia. Am J Nephrol 1998; 18:42.
  56. Seys E, Andrini O, Keck M, et al. Clinical and Genetic Spectrum of Bartter Syndrome Type 3. J Am Soc Nephrol 2017; 28:2540.
  57. Bockenhauer D, Feather S, Stanescu HC, et al. Epilepsy, ataxia, sensorineural deafness, tubulopathy, and KCNJ10 mutations. N Engl J Med 2009; 360:1960.
  58. Scholl UI, Choi M, Liu T, et al. Seizures, sensorineural deafness, ataxia, mental retardation, and electrolyte imbalance (SeSAME syndrome) caused by mutations in KCNJ10. Proc Natl Acad Sci U S A 2009; 106:5842.
  59. Zhang C, Wang L, Zhang J, et al. KCNJ10 determines the expression of the apical Na-Cl cotransporter (NCC) in the early distal convoluted tubule (DCT1). Proc Natl Acad Sci U S A 2014; 111:11864.
  60. Praga M, Vara J, González-Parra E, et al. Familial hypomagnesemia with hypercalciuria and nephrocalcinosis. Kidney Int 1995; 47:1419.
  61. Nicholson JC, Jones CL, Powell HR, et al. Familial hypomagnesaemia--hypercalciuria leading to end-stage renal failure. Pediatr Nephrol 1995; 9:74.
  62. Benigno V, Canonica CS, Bettinelli A, et al. Hypomagnesaemia-hypercalciuria-nephrocalcinosis: a report of nine cases and a review. Nephrol Dial Transplant 2000; 15:605.
  63. Müller D, Kausalya PJ, Bockenhauer D, et al. Unusual clinical presentation and possible rescue of a novel claudin-16 mutation. J Clin Endocrinol Metab 2006; 91:3076.
  64. Konrad M, Hou J, Weber S, et al. CLDN16 genotype predicts renal decline in familial hypomagnesemia with hypercalciuria and nephrocalcinosis. J Am Soc Nephrol 2008; 19:171.
  65. Rodríguez-Soriano J, Vallo A. Pathophysiology of the renal acidification defect present in the syndrome of familial hypomagnesaemia-hypercalciuria. Pediatr Nephrol 1994; 8:431.
  66. Simon DB, Lu Y, Choate KA, et al. Paracellin-1, a renal tight junction protein required for paracellular Mg2+ resorption. Science 1999; 285:103.
  67. Blanchard A, Jeunemaitre X, Coudol P, et al. Paracellin-1 is critical for magnesium and calcium reabsorption in the human thick ascending limb of Henle. Kidney Int 2001; 59:2206.
  68. Weber S, Schneider L, Peters M, et al. Novel paracellin-1 mutations in 25 families with familial hypomagnesemia with hypercalciuria and nephrocalcinosis. J Am Soc Nephrol 2001; 12:1872.
  69. Kausalya PJ, Amasheh S, Günzel D, et al. Disease-associated mutations affect intracellular traffic and paracellular Mg2+ transport function of Claudin-16. J Clin Invest 2006; 116:878.
  70. Godron A, Harambat J, Boccio V, et al. Familial hypomagnesemia with hypercalciuria and nephrocalcinosis: phenotype-genotype correlation and outcome in 32 patients with CLDN16 or CLDN19 mutations. Clin J Am Soc Nephrol 2012; 7:801.
  71. Hou J, Renigunta A, Konrad M, et al. Claudin-16 and claudin-19 interact and form a cation-selective tight junction complex. J Clin Invest 2008; 118:619.
  72. Konrad M, Schaller A, Seelow D, et al. Mutations in the tight-junction gene claudin 19 (CLDN19) are associated with renal magnesium wasting, renal failure, and severe ocular involvement. Am J Hum Genet 2006; 79:949.
  73. Meij IC, Saar K, van den Heuvel LP, et al. Hereditary isolated renal magnesium loss maps to chromosome 11q23. Am J Hum Genet 1999; 64:180.
  74. Meij IC, Koenderink JB, van Bokhoven H, et al. Dominant isolated renal magnesium loss is caused by misrouting of the Na(+),K(+)-ATPase gamma-subunit. Nat Genet 2000; 26:265.
  75. Meij IC, Koenderink JB, De Jong JC, et al. Dominant isolated renal magnesium loss is caused by misrouting of the Na+,K+-ATPase gamma-subunit. Ann N Y Acad Sci 2003; 986:437.
  76. Schlingmann KP, Bandulik S, Mammen C, et al. Germline De Novo Mutations in ATP1A1 Cause Renal Hypomagnesemia, Refractory Seizures, and Intellectual Disability. Am J Hum Genet 2018; 103:808.
  77. Glaudemans B, van der Wijst J, Scola RH, et al. A missense mutation in the Kv1.1 voltage-gated potassium channel-encoding gene KCNA1 is linked to human autosomal dominant hypomagnesemia. J Clin Invest 2009; 119:936.
  78. Adalat S, Woolf AS, Johnstone KA, et al. HNF1B mutations associate with hypomagnesemia and renal magnesium wasting. J Am Soc Nephrol 2009; 20:1123.
  79. Adalat S, Hayes WN, Bryant WA, et al. HNF1B Mutations Are Associated With a Gitelman-like Tubulopathy That Develops During Childhood. Kidney Int Rep 2019; 4:1304.
  80. Kompatscher A, de Baaij JHF, Aboudehen K, et al. Loss of transcriptional activation of the potassium channel Kir5.1 by HNF1β drives autosomal dominant tubulointerstitial kidney disease. Kidney Int 2017; 92:1145.
  81. Wagner CA. Disorders of renal magnesium handling explain renal magnesium transport. J Nephrol 2007; 20:507.
  82. Groenestege WM, Thébault S, van der Wijst J, et al. Impaired basolateral sorting of pro-EGF causes isolated recessive renal hypomagnesemia. J Clin Invest 2007; 117:2260.
  83. Ferrè S, de Baaij JH, Ferreira P, et al. Mutations in PCBD1 cause hypomagnesemia and renal magnesium wasting. J Am Soc Nephrol 2014; 25:574.
  84. Stuiver M, Lainez S, Will C, et al. CNNM2, encoding a basolateral protein required for renal Mg2+ handling, is mutated in dominant hypomagnesemia. Am J Hum Genet 2011; 88:333.
  85. Goytain A, Quamme GA. Functional characterization of ACDP2 (ancient conserved domain protein), a divalent metal transporter. Physiol Genomics 2005; 22:382.
  86. Aglio LS, Stanford GG, Maddi R, et al. Hypomagnesemia is common following cardiac surgery. J Cardiothorac Vasc Anesth 1991; 5:201.
  87. Palestine AG, Polis MA, De Smet MD, et al. A randomized, controlled trial of foscarnet in the treatment of cytomegalovirus retinitis in patients with AIDS. Ann Intern Med 1991; 115:665.
  88. Scott VL, De Wolf AM, Kang Y, et al. Ionized hypomagnesemia in patients undergoing orthotopic liver transplantation: a complication of citrate intoxication. Liver Transpl Surg 1996; 2:343.
  89. Frisch LS, Mimouni F. Hypomagnesemia following correction of metabolic acidosis: a case of hungry bones. J Am Coll Nutr 1993; 12:710.
  90. Kang HC, Chung DE, Kim DW, Kim HD. Early- and late-onset complications of the ketogenic diet for intractable epilepsy. Epilepsia 2004; 45:1116.
  91. Rosen EU, Campbell PG, Moosa GM. Hypomagnesemia and magnesium therapy in protein-calorie malnutrition. J Pediatr 1970; 77:709.
  92. Ziegler TR. Parenteral nutrition in the critically ill patient. N Engl J Med 2009; 361:1088.
  93. Wilson FH, Hariri A, Farhi A, et al. A cluster of metabolic defects caused by mutation in a mitochondrial tRNA. Science 2004; 306:1190.
  94. Sitprija V. Altered fluid, electrolyte and mineral status in tropical disease, with an emphasis on malaria and leptospirosis. Nat Clin Pract Nephrol 2008; 4:91.
  95. Spichler A, Athanazio DA, Furtado J, et al. Case report: severe, symptomatic hypomagnesemia in acute leptospirosis. Am J Trop Med Hyg 2008; 79:915.
Topic 829 Version 46.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟