Skip to main content
Biomedical Journal logoLink to Biomedical Journal
. 2021 Nov 10;45(1):74–87. doi: 10.1016/j.bj.2021.11.002

Clinical and genetic approach to renal hypomagnesemia

Min-Hua Tseng a,b,f, Martin Konrad c, Jhao-Jhuang Ding d, Shih-Hua Lin e,
PMCID: PMC9133307  PMID: 34767995

Abstract

Magnesium (Mg2+) is an important intracellular cation and essential to maintain cell function including cell proliferation, immunity, cellular energy metabolism, protein and nucleic acid synthesis, and regulation of ion channels. Consequences of hypomagnesemia affecting multiple organs can be in overt or subtle presentations. Besides detailed history and complete physical examination, the assessment of urinary Mg2+ excretion is help to differentiate renal from extra-renal (gastrointestinal, tissue sequestration, and shifting) causes of hypomagnesemia. Renal hypomagnesemia can be caused by an increased glomerular filtration and impaired reabsorption in proximal tubular cells, thick ascending limb of the loop of Henle or distal convoluted tubules. A combination of renal Mg2+ wasting, familial history, age of onset, associated features, and exclusion of acquired etiologies point to inherited forms of renal hypomagnesemia. Based on clinical phenotypes, its definite genetic diagnosis can be simply grouped into specific, uncertain, and unknown gene mutations with a priority of genetic approach methods. An unequivocal molecular diagnosis could allow for prediction of clinical outcome, providing genetic counseling, avoiding unnecessary studies or interventions, and possibly uncovering the pathogenic mechanism. Given numerous identified genes responsible for Mg2+ transport in renal hypomagnesemia over the past two decades, several potential and specific molecular and cellular therapeutic strategies to correct hypomagnesemia are promising.

Keywords: Inherited disorders, Hypomagnesemia, Genetic diagnosis


Magnesium (Mg2+), the second-most abundant intracellular cation, is pivotal as a cofactor in the maintenance of numerous cell functions. Deficiency of Mg2+ can lead to muscle weakness, tremor, seizure, arrhythmia, coma, and even death [1,2]. Up to 15% of hospitalized patients and 60% of patients admitted to intensive care unit have hypomagnesemia [[3], [4], [5]]. Gastrointestinal and renal losses are the two major causes of hypomagnesaemia. The acquired renal hypomagnesemia commonly results from medications, alcohol, and osmotic diuresis. Inherited renal hypomagnesemia is defined as hypomagnesemia of renal origin caused by defects of genes responsible for renal Mg2+ handling. Although the inherited renal hypomagnesemia is less common than acquired forms, the inherited renal hypomagnesemia features more variable in characteristics, more refractory in treatment and more severe in consequence. Without timely recognition and appropriate treatment, it can lead to severe complications such as cardiac arrhythmias and death.

Numerous genes encoding proteins involved in Mg2+ transport in the renal tubules have been identified over the last two decades. Such newfound knowledge can potentially enable us to modify and personalize therapeutic strategies. Our aim is to summarize the molecular advances that are linked to renal hypomagnesemia and propose a comprehensive molecular approach for diagnosis and management. Recent advances in molecular and cellular therapies of inherited disorders are also discussed.

Magnesium homeostasis

Magnesium (Mg2+) is a crucial cation involved in a number of biological processes, including cell proliferation, immunity, cellular energy metabolism, protein and nucleic acid synthesis, and regulation of ion channels [1,2]. The serum Mg2+ is tightly maintained within a narrow range. In the body, around 53% are distributed and stored in bones, 27% in muscles, 19% in soft tissues, and only 1% in blood. Around 65–70% of the circulating Mg2+ is in the ionized form which is the active form critical for physiological function. The rest of Mg2+ binds with either proteins or anion such as citrate and phosphate. The extracellular Mg2+ concentration is tightly regulated by gut and kidney, and the serum Mg2+ level reflects the equilibrium between absorption in intestine and urinary excretion.

Gastrointestinal absorption of Mg2+

Mg2+ is acquired through daily food intake, where nearly 35%–80% of the ingested Mg2+ is absorbed [2]. Specifically, around 30–50% of dietary Mg2+ intake is absorbed in the jejunum and ileum by way of passive paracellular route and in the colon through active transcellular route by transient receptor potential melastatin type 6 and 7 (TRPM6 and 7). The absorbed Mg2+ enters the bloodstream by CNNM4 and Na + -Mg2+ exchanger at the basolateral side of cells [6].

Renal regulation of Mg2+ reabsorption

Serum Mg2+ is subsequently excreted in the urine after storage in organs and cellular utilization. The kidney determines the final urinary Mg2+ excretion, and hence plays a major role in Mg2+ homeostasis. In the kidney, only 3–5% of filtered Mg2+ is excreted after reabsorption in renal tubules. As shown in Fig. 1, around 15–25% of filtered Mg2+ is absorbed in the PCT via paracellular pathway. The increasing intratubular Mg2+ concentration triggers the passive reabsorption via claudin 1 and 2 in the late PCT [7,8]. Schlingmann et al. identified children with biallelic variants in KCNJ16, which encodes basolateral potassium channel (Kir5.1) of PCT and DCT, have hypokalemic tubulopathy, salt wasting, disturbed acid-base homeostasis, sensorineural deafness, and hypomagnesemia [9]. In the TALH, about 60–70% of filtered Mg2+ is reclaimed passively by paracellular pathway via claudin 16 and 19 triggered by positive luminal transepithelial voltage. This positive intraluminal voltage is generated by apical Na + -K+-Cl- cotransporter (NKCC2)-mediated Na+, K+, Cl reabsorption, and apical renal outer medullary potassium (ROMK)-mediated parallel K+ excretion [[10], [11], [12], [13]]. Calcium-sensing receptor (CaSR) expressed abundantly in basolateral side of TALH regulates paracellular Mg2+ reabsorption by involving the regulation of salt reabsorption in TAL and the expression of claudin-14 [14,15]. Recent study has shown that children with heterozygous variants in RRAGD, which encodes GTPase RagD of TALH and DCT, has hypomagnesemia, salt wasting, nephrocalcinosis, and dilated cardiomyopathy [16]. The RagD variants leads to constitutive activation of mTOR signaling in vitro and is supposed to interfere with the handling of Mg2+ and other electrolytes in TALH and DCT.

Fig. 1.

Fig. 1

Magnesium reabsorption in proximal convoluted tubule, cortical thick ascending limb of Henle's loop, and distal convoluted tubule. Around 15–25% of filtered Mg2+ is reabsorbed passively via paracellular pathway (probable claudin 1 and 2) in proximal convoluted tubule. The positive luminal voltage generated by apical NKCC2 and ROMK provides the driving force for paracellular reabsorption of 60–70% filtered Mg2+ in thick ascending limb of Henle's loop. The basolateral Na–K-ATPase in provides the driving force of NKCC2. The basolateral CaSR exerts inhibitory effects on paracellular claudin and ROMK. The Mg2+ is reabsorbed actively through TRPM6 in DCT. The apical Kv1.1 provide apical membrane potential by apical excretion of K+. The basolateral Na–K-ATPase establishes the transapical membrane gradient critical for activity of TRPM6. The transcriptional factors including HNF1β and PCBD1 of γ-subunit of Na–K-ATPase, encoded by FXYD2, regulate Mg2+ reabsorption via alteration expression of γ-subunit of Na–K-ATPase. Kv4.1 located on basolateral side recycles the imported by Na–K-ATPase via conducting outward K+ currents. The paracrine action of EGF regulates the activity of TRPM6. The Mg2+ efflux is conducted in Na+-Mg2+ exchanger and possibly also in CNNM2.

Although the amount of reabsorbed Mg2+ in distal convoluted tubule (DCT) is lower than that in PCT and TALH, the DCT plays a crucial role of determining final urinary Mg2+ excretion. The active reabsorption of 5–10% of filtered Mg2+ is tightly regulated in the DCT through transcellular TRPM6 [Fig. 1]. Genetic defects of TRPM6 encoding TRPM6 result in renal Mg2+ wasting and consecutive hypoparathyroidism and hypocalcemia (HSH). The mutations of SLC12A3 encoding thiazide-sensitive sodium chloride cotransporter (NCC) in DCT lead to Na+, K+, and Cl wasting, and reduced expression of TRPM6 [17]. Kv1.1, the apical voltage-gated potassium channel, has been demonstrated to be involved in Mg2+ reabsorption, by the finding that non-functional Kv1.1 results in isolated dominant hypomagnesemia [18]. Kv1.1 facilitates Mg2+ reabsorption by generation of intraluminal positive voltage through apical secretion of potassium in DCT. The binding of epidermal growth factor (EGF) and EGF receptor in the basolateral side of DCT regulates the apical shuttling of TRPM6, and defects in EGF gene have been reported to cause isolated recessive hypomagnesemia, where apical TRPM6 expression is reduced [14,19]. The γ-subunit of the basolateral Na+-K+ ATPase encoded by FXYD2 stabilizes Na + -K+ ATPase. Mutations in FXYD2 have been demonstrated to cause diminished NCC activity and reduced driving force of Mg2+ reabsorption via TRPM6 [20]. The α1-subunit encoded by ATP1A1 represents the exclusive of α-subunits of basolateral Na+-K+ ATPase in kidney [21]. Heterozygous mutations in ATP1A1 recently have been reported to cause renal hypomagnesemia, seizure, and mental retardation [22]. The hepatocyte nuclear factor 1 (HNF1β) and pterin-4a-carinolamine dehydratase (PCBD1), the transcriptional regulatory proteins of γ-subunit of the basolateral Na+-K+ ATPase, have been reported to be associated with renal Mg2+ wasting, and mutations in HNF1B and PCBD1 lead to inherited dominant hypomagnesemia [15,23]. The basolateral Kir4.1 encoded by KCNJ10 (potassium voltage-gated channel subfamily J member 10) has been found to be involved in Mg2+ reabsorption by maintaining the function of Na + -K+ ATPase [24,25]. The genetic defects of KCNJ10 can result in compromised NCC activity and consequent impairment of Mg2+ reabsorption [24]. As far, Na+-Mg2+ exchanger and cyclin M2 are the two reported basolateral proteins that are proposed to act as Mg2+ efflux channels [10,26]. Cyclin M2, encoded by CNNM2, is primarily expressed in both TALH and DCT and is thought to be a basolateral Mg2+ transporter and also act as intracellular Mg2+ sensor by inducing a conformational change upon binding of Mg2+-ATP [27]. Mutations of CNNM2 have been identified to be the causative gene for patients with hypomagnesemia, seizure, intellectual disability, a clinical entity referred to as HSMR syndrome [10].

Clinical manifestations of hypomagnesemia

Hypomagnesemia is defined as total serum Mg2+ concentration less than 1.7 mg/dL (0.7 mM), and the clinical symptoms of hypomagnesemia may not be significant unless it reaches profound hypomagnesemia below 1.2 mg/dL (0.5 mM). Since Mg2+ is involved in many vital physiological functions hypomagnesemia may cause a myriad of manifestations involving multiple organs [Fig. 2]. Neuronal Mg2+ involved in the regulation of N-methyl-d-aspartate (NDMA) and γ-aminobutyric acid (GABA) receptor, and deficiency of Mg2+ leads to hyperexcitability of NMDA and decreased stimulation of GABA [28,29]. In the central nervous system, as shown in Fig. 2, hypomagnesemia can cause tremor and convulsion from neuromuscular hyperexcitabiliy and it is also associated with migraine, brain injury, stroke, and mood disorders [[30], [31], [32]]. In lung, Mg2+ has bronchodilatory and anti-inflammatory effects on airway [33,34]. Hypomagnesaemia has been shown to be associated with asthma and chronic obstructive pulmonary disorder, therefore, Mg2+ is supposed to have therapeutic roles on these disorders. In cardiovascular system, Mg2+ regulates the myocardial contractility and has effects of anti-inflammation, vasodilatation, and inhibitory crystallization of calcium-phosphate [35,36]. Hence, hypomagnesemia may cause arrhythmia, myocardial infarction, vascular calcification, hypertension, and coronary artery diseases. In muscle, Mg2+ acts as Ca2+ antagonist to compete the binding sites of proteins responsible for muscle contraction [37]. Hence, Mg2+ deficiency may lead to hypercontractibility and muscle cramps. In bone, Mg2+ participates in bone formation by stimulating the proliferation of osteoblast and increasing the solubility of minerals. Mg2+ deficiency has been reported to be associated with osteoporosis [38]. Mg2+ may have effects on the secretion of insulin by the findings that patients with Mg2+ deficiency have a decreased insulin secretion [39]. Additionally, patients with diabetes also have low serum Mg2+ levels [40]. In liver, patients with liver cirrhosis and fatty liver are Mg2+ depleted. This may be due to enzymatic reactions in liver that are Mg2+- dependent [41]. Mg2+ has been reported to regulate the proliferation and development of T lymphocytes, and it is associated with X-linked T-cell immunodeficiency [1,42].

Fig. 2.

Fig. 2

Pathophysiology of hypomagnesemia. (A) Clinical manifestations and organ-specific consequences of hypomagnesemia. (B) Cellular physiology of Mg2+. Several transporters are responsible for the cellular Mg2+ homeostasis. Mg2+ stabilizes the structures of DNA and RNA, DNA and RNA polymerases, and their repair in the nucleus. Additionally, Mg2+ also regulated the cell growth and proliferation. In cytosol, Mg2+ is involved in many enzymatic reactions and regulates the glycolysis and ATP synthesis.

Approach to hypomagnesemia

Apart from inadequate dietary intake and malabsorption in gut, defects of renal reabsorption are largely responsible for the dysregulation of Mg2+ homeostasis. In general, causes of hypomagnesemia can be divided into two categories: high renal Mg2+ excretion and low renal Mg2+ excretion including gastrointestinal, tissue sequestration, and shifting origins. Besides detailed medical and family histories, complete physical examination, and urine and serum electrolytes measurement, fractional excretion rate of Mg2+ (FEMg) can provide a rapid differentiation of these two categories [Fig. 3]. In the setting of hypomagnesemia, FEMg >4% indicates renal Mg2+ wasting, while FEMg <2% suggests appropriate renal Mg2+ conservation and Mg2+ wasting of extra-renal origin. Of note, a low glomerular filtration rate may reduce the filtered load of Mg2+, and therefore lowers the cutoff values of FEMg indicating renal wasting. Common causes of gastrointestinal Mg2+ losses include dietary deprivation, diarrhea, malabsorption, use of proton pump inhibitor, and rare primary familial hypomagnesemia caused by molecular defects in TRPM6. Acute pancreatitis, massive blood transfusion, hungry bone syndrome, refeeding syndrome, and cardiopulmonary bypass are common causes of hypomagnesemia secondary to cellular shift and tissue sequestration. Rarely, in hypervolemia and/or overall reduction of serum anion, FEMg may still appear appropriate, despite an overall increase in renal Mg2+ excretion. Renal hypomagnesemia can be caused by an increased glomerular filtration and defective transport in proximal tubular segments, thickening ascending limb of the loop of Henle or distal convoluted tubules.

Fig. 3.

Fig. 3

Congenital and acquired causes of hypomagnesemia.

Identify genetic forms of renal hypomagnesemia

Before establishing the diagnosis of inherited renal hypomagnesemia, the acquired etiologies of renal Mg2+ wasting such as medications should be carefully excluded. Historical clues such as prior hypomagnesemia, associated organ abnormalities in family members, use of drugs affecting hypomagnesemia-loop diuretics, proton pump inhibitor, calcineurin inhibitor, aminoglycoside, foscanet, cisplatin and cetuximab, age of onset, and nephrolithiasis or nephrocalcinosis must be carefully obtained. The diagnosis of inherited renal hypomagnesemia would be made after exclusion the acquired etiologies above-mentioned.

Differential diagnosis of inherited renal hypomagnesemia

The mode of inheritance, assessment of urine and blood biochemistry studies, presence of extra-renal symptoms all aid in determination of further genetic testing. The models of inheritance of inherited renal hypomagnesemia include autosomal dominant, autosomal recessive and maternal inheritance. As shown in Fig. 4, the etiology of inherited renal hypomagnesemia can be divided into lesions in TALH, DCT and PCT by renal calcium excretion. Hypercalciuria points the TALH tubulopathy such as Bartter syndrome, autosomal dominant hypocalcemia, and familial hypercalciuria hypomagnesemia nephrocalcinosis. Absence of hypercalciuria can be further divided into lesions in DCT and PCT. The presence of hypokalemia is cardinal feature for differentiating the Gitelman syndrome-like hypomagnesemia from others DCT tubulopathy [Fig. 4]. Gitelman syndrome-like hypomagnesemia can be further differentiated by the presence of extra-renal manifestations and modes of inheritance. Finally, mitochondrial hypomagnesemia is one of important causes of PCT tubulopathy.

Fig. 4.

Fig. 4

Differential diagnosis of inherited renal hypomagnesemia.

Genetic approach to inherited renal hypomagnesemia

As shown in Fig. 5, direct Sanger sequencing can identify the disease-specific gene assessed by above-mentioned approach [Table 1 and Fig. 4]. Of note, direct Sanger sequencing will miss deep intronic mutation not rare in Gitelman syndrome and large deletion including cBS, familial hypomagnesemia hypercalciuria nephrocalcinosis, HNF1β nephropathy, hypomagnesemia secondary hypocalcemia, and EAST syndrome [43,44]. Therefore, if unable to identify the causative genetic mutation, analysis of cDNA for deep intronic mutation or MLPA (multiplex ligation-dependent probe amplification) for large deletion may be warranted. In patients of diseases with uncertain gene due to the overlapping phenotype and genetic heterogeneity, a gene panel can be considered to assess multiple causal genes simultaneously. The gene panel is also recommended for patients of suspicion of specific disease without detected variants by direct Sanger sequencing. Some patients might have no detected variants by direct Sanger sequence, MLPA, cDNA, and gene panels. In such cases, next generation sequencing (NGS) will aid in the molecular diagnosis. NGS, such as whole exome sequencing (WES) and whole genome sequencing (WGS), provide rapid screen of known genes, as well as modifier genes and epigenetic modification. The bioinformatics analysis and variants databases are essential for determining the pathogenic role of variants identified by NGS. Epigenetic analysis, including DNA methylation, siRNA regulation, and chromatin immunoprecipitation sequencing, may be the further potential tests for patients without identified variants by NGS.

Fig. 5.

Fig. 5

Genetic strategy for diagnosis of inherited renal hypomagnesemia. The diagnostic paradigm for inherited hypomagnesemia diseases is determined by spectrum of specific disease, disorders localized in specific tubules, and disorders with unknown mechanism candidate gene after comprehensive evaluation of phenotypes. The following molecular methods including cDNA analysis multiplex ligation-dependent probe amplification, gene panels, and next generation is considered step by step.

Table 1.

Clinical, biochemical, and genetic characteristics of inherited disorders of renal hypomagnesemia.

Disordersa Involved tubule Inheritance Gene Protein Large deletion Age at onset Serum Ca Serum K Blood pH Urine Mg Urine Ca Extrarenal manifestation Nephrocalcinosis/nephrolithiasis Renal anomaly Early ESRDc Reference
ADH TALH AD CASR CaSR Yes Adolescence/adulthood Hypoparathyroidism Yes No No [65]
FHHNC TALH AR CLDN16, CLDN19 Claudin-16, Caludin-19 Yes Childhood/adolescence ↑↑ ↑↑ Ocular abnormalities hyperparathyroidism Yes No Yes [[66], [67], [68], [69]]
cBS TALH
DCT
AR CLCNKB ClC-Kb Yes Childhood var. ↓↓ - or ↑ No Yes (infrequent) No No [[70], [71], [72], [73]]
BS, type IVa TALH
DCT
AR BSND Barttin Yes Infancy var. ↓↓ - or ↑ Sensorineural deafness Yes (infrequent) No Yes [74]
BS, type IVb TALH
DCT
AR CLCNKA
CLCNKB
ClC-Ka
ClC-Kb
Yes Infancy var. ↓↓ - or ↑ Sensorineural deafness Yes (infrequent) No Yes [75,76]
KICA syndrome TALH, DCT AD RRAGD RagD No Infancy/Childhood ↑ or ↓ Dilated Cardiomyopathy Yes No No [77]
HNF1B nephropathy DCT AD HNF1B HNF1beta Yes Adolescence/adulthood ? ? - or ↑ - or ↓ MODY5, hyperuricemia No Renal cysts, dysplasia, agenesis Yes [[78], [79], [80], [81]]
EAST syndrome DCT AR KCNJ10 Kir4.1 Yes Neonate/infancy Epilepsy, ataxia, sensorineural deafness, intellectual disability No No No [82,83]
HPABH4D DCT AR PCBD1 PCBD1 Yes adolescence/adulthood ? MODY No No No [23,84]
GSd DCT AR SLC12A3 NCC Yes Adolescence/adulthood Chondrocalcinosis No No No [43,[85], [86], [87], [88], [89]]
IDH DCT AD FXYD2 γ subunit of Nab-Kb ATPase No Adolescence/adulthood - or ↑ No No No No [90]
HSMR syndrome 2 DCT AD ATP1A1 α subunit of Nab-Kb ATPase No Neonate/infancy ↑↑ - or ↑ Intellectual disability, epilepsy Yes No No [22]
HSMR syndrome 1 DCT AD/AR CNNM2 Cyclin M2 Yes Infancy/childhood - or ↑ ? Intellectual disability, epilepsy No No No [91]
KCS2 syndrome DCT AD FAM111A FAM111A No Infancy ↓↓ ? ? ? ? Impaired skeletal development, hypocalcemia, hypoparathyroidism No No No [92,93]
EA1 DCT AD KCNA1 Kv1.1 No Childhood ? - or ↑ Episodic ataxia, myokymia, epilepsy, intellectual disability No No No [94]
HSH DCT AR TRPM6 TRPM6 Yes Infancy - or ↑ ;= or ↑ intellectual disability, epilepsy, hypoparathyroidism No No No [95,96]
IRH DCT AR EGF Pro-EGF No Infancy - or ↑ intellectual disability, epilepsy No No No [19]
NISBD2 DCT AR EGFR EGFR No Infancy - or ↑ Severe inflammation of skin and bowel heart abnormalities No Renal dysplasia No [97]
GS phenocopy DCTb Mt MT-TI, MT-TF Mt. tRNAile, phe No Adulthood - or ↑ No No No No [98]] [99]
HUPRA syndrome PCTb AR SARS2 SARS2 No Infancy ? ↑↑ ↓? Hyperuricemia, elevated serum lactate, pulmonary hypertension, prematurity, intellectual disability, diabetes mellitus No No Yes [100]
KSS PCTb Mt Mitochondrial deletion Yes Childhood ↓↓ ↓ or ↑ ↑↑ Brain, eye, ear involvement. Muscle weakness, ataxia, intellectual disability, epilepsy, diabetes mellitus, gonadal failure, thyroid disease, hypoparathyroidism No No No [[101], [102], [103]]
Hypokalemic tubulopathy, salt wasting, disturbed acid-base homeostasis and deafness PCTa TALH
DCT
AR KCNJ16 KCNJ16 No Infancy/Childhood Sensorineural deafness No No No [9]
a

Abbreviations: ADH: autosomal dominant hypocalcemia; BS: Bartter syndrome; cBS: classic Bartter syndrome; DCT: distal convoluted tubule; EA1: episodic ataxia, type I; EAST syndrome: epilepsy, ataxia, sensorineural deafness, and tubulopathy syndrome; ESRD: end stage renal disease; FHHNC: familial primary hypomagnesemia with hypercalciuria and nephrocalcinosis; GS: Gitelman syndrome; HHH syndrome: hypertension, hypercholesterolemia, and hypomagnesemia syndrome; HPABH4D: hyperphenylalaninemia, BH4-deficient, type D; HSH: hypomagnesemia with secondary hypocalcemia; HSMR syndrome: hypomagnesemia, seizures, and intellectual disability syndrome; HUPRA syndrome: hyperuricemia, pulmonary hypertension, and renal failure syndrome; IDH: isolated-dominant hypomagnesemia; IRH: isolated-recessive hypomagnesemia; KCS2 syndrome: Kenny-Caffey syndrome, type 2; KICA syndrome: kidney tubulopathy and cardiomyopathy; KSS: Kearns–Sayre syndrome; NISBD2: neonatal inflammatory skin and bowel disease; PCT: proximal convoluted tubule; TALH: thick-ascending loop of Henle.

b

Lesion in PCT may have characteristics of TALH and DCT tubulopathy.

c

End stage renal disease.

d

Deep intronic mutation.

Management of hypomagnesemia

Delivery of Mg2+ supplementation, avoidance of exacerbated renal Mg2+ wasting, and correction of accompanying metabolic disarrangement is the mainstay treatment in inherited renal hypomagnesemia. The route of administration and dosage of Mg2+ supplement dependents on the severity of hypomagnesemia. In patients with acute symptomatic hypomagnesemia, parenteral Mg2+ supplementation should be considered for alleviating potential complications. Oral Mg2+ supplementation is suitable for patients with non-acute and asymptomatic hypomagnesemia. The common formulations of Mg2+ supplementations and Mg2+-rich food are summarized in Table 2. Although there is no upper limit of dietary Mg2+, oral elemental Mg2+ supplementation less than 350 mg per day is safe for adults and children older than 8 years [45]. The tolerable upper limit of daily elemental Mg2+ supplementation for children 1–3 years and 4–8 years are 65 mg and 110 mg, respectively [46]. Based on the different bioavailability of Mg2+ formula, organic Mg2+ compounds including Mg2+ citrate, Mg2+ aspartate, Mg2+ chloride, and Mg2+ glycerophosphate are superior to Mg2+ oxide and Mg2+ sulfate in correcting hypomagnesemia [47]. Conditions associated with renal hyperfiltration (osmotic diuresis, poor control of hyperglycemia, and massive fluid intake) and increased filterable Mg2+ (loop diuretics, chronic metabolic acidosis, low serum anion including hypoalbuminemia, hypophosphatemia) could aggravate the renal Mg2+ wasting and should corrected appropriately. Accordingly, angiotensin-converting-enzyme (ACE) inhibitors and angiotensin II receptor blockers (ARB) that reduce glomerular filtration can be considered for patients who are in a glomerular hyperfiltrative state. In addition, aldosterone antagonist has been reported to ameliorate the renal Mg2+ wasting and maintain serum Mg2+ levels in patients with congestive heart failure [48]. Aldosterone has been demonstrated to induce renal Mg2+ wasting through increases in intracellular Mg2+ shifts from muscle and bone [49]. Additionally, aldosterone has also been shown to downregulate the activity of renal TRPM6 [50]. Several metabolic disarrangements including hypokalemia, hypocalcemia, hypercalciuria, and hyperglycemia caused by diabetes mellitus may also occur in patients with inherited hypomagnesemia. The condition of hyperglycemia could further exacerbate hypomagnesemia. Hypocalcemia, hypokalemia and hypercalciuria may further deteriorate renal function if left untreated. Therefore, these accompanied metabolic disarrangements should be addressed simultaneously.

Table 2.

Magnesium formulations and magnesium-rich foods.

Supplement Elemental Mg2+ content
Intravenous formulations
 Magnesium sulfate 0.10 mg/mg
Oral formulations
 Magnesium oxide 0.61 mg/mg
 Magnesium hydroxide 0.42 mg/mg
 Magnesium gluconate 0.059 mg/mg
 Magnesium chloride 0.12 mg/mg
 Magnesium carbonate 0.29 mg/mg
 Magnesium lactate 0.12 mg/mg
 Magnesium aspartate hydrochloride 0.10 mg/mg
 Magnesium citrate 0.16 mg/mg
Food sources
 Seeds (pumpkin, chia) 400–560 mg/100 g
 Almonds, dry roasted 286 mg/100 g
 Whole grains 232 mg/100 g
 Dark chocolate 230 mg/100 g
 Peanuts, oil roasted 107 mg/100 ml
 Spinach, boiled 66 mg/100 ml
 Tofu 53 mg/100 g
 Salmon 30 mg/100 g
 Banana 27 mg/100 g
 Chicken breast, roasted 26 mg/100 g
 Soymilk 26 mg/100 ml
 Yogurt 19 mg/100 g
 Milk 10.1–11.4 mg/100 ml

Potential therapy for genetic renal hypomagnesemia

As shown in Fig. 6, molecular therapies that target modifications at the level of DNA, RNA, and proteins are currently being developed and applied on variety of inherited diseases. Successful development of personalized treatment approaches is dependent on the severity of diseases, identification of genetic defects, and understanding the pathophysiological mechanism [[34], [35], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45], [46], [47]]. DNA therapies include gene replacement therapy and genomic editing [51,52]. Gene replacement therapy works by using the therapeutic vectors to insert the normal copy of mutant gene into host cells, and this might be potential for patients with large deletion mutation. Heikkilä et al. succeeded in delivering the COL4A5 gene by adenoviral vector to glomerular cells [53]. Genomic editing allows for precise editing of genomic DNA in vivo or ex vivo by utilizing CRISPR/Cas system and EFNs and TALENs [54,55]. Daga et al. developed two-plasmid approach to achieve a stable variant-specific correction in the X-linked COL4A5 (p.Gly624Asp) and COL4A3 gene (p.Gly856Glu) using CRISP/Cas9 genome editing [56]. RNA therapies are represented by splicing modulation, RNA silence, and RNA editing. The splicing modulation is conducted for correcting aberrant splicing by antisense oligonucleotides, U1 splicesomal RNA, or trans-splicing [[57], [58], [59]]. As shown in Table 1, splice site mutations are identified in several genes responsible for inherited renal hypomagnesemia. Ramsbottom and his colleagues delivered antisense oligonucleotide (ASO)-induced splicing of the mutated exon (CEP20 G1890∗) to restores protein expression in cells from patient with Joubert syndrome [60]. Translation or protein targeting includes translational read-through, restoring proteostasis and pathway-specific therapy. Aminoglycoside and PTC124, the two main drugs of translational read-through, allow the translational machinery to bypass the premature termination code [61]. We had significantly increased survival rate and partially rescued Na+/HCO3- cotransporter 1 (NBCe1) activity ex vivo by delivering PTC124 therapy in NBCe1 p.W516∗ knock-in mice [62]. Cell therapy allows integrating the exogenous delivery of cells and reactivation of cellular function. The induced pluripotent stem cells from somatic cells of patients or embryonic stem cells could be used and differentiated to tubular precursor cells [63]. Of note, the genome-edited cells from patients by CRISPR-engineered cell therapies can be also used for cell therapy and has been successfully applied clinically [56,64]. The above-mentioned potential therapies required appropriate cellular and/or animal models to test the efficacy and safety in the future.

Fig. 6.

Fig. 6

Proposed molecular and cell therapies for inherited renal hypomagnesemia.

Concluding remarks

The etiologies responsible for impaired Mg2+ homeostasis are complex and heterogeneous, therefore, a comprehensive evaluation is essential for accurate diagnosis. After the exclusion of acquired causes, genetic causes can be further classified into defects in TALH, DCT and PCT according to the specific biochemical characteristics and extra-renal manifestations. Strategy for molecular diagnosis of inherited renal hypomagnesemia can be further stratified: patients with specific disease, disease with uncertain gene, and those with unknown gene. Algorithms for genetic testing can be developed based on the knowledge of advantages and limitations of molecular methods including direct Sanger sequencing, cDNA analysis, MLPA, gene panels, and WES. Once the causative gene is detected, prediction of clinical outcome, accurate therapeutic intervention, and genetic consulting are all facets of personalized medicine that ultimately lead to improved outcome within a shorter time frame. Although increasing numbers of genetic defects involving renal Mg2+ homeostasis is being identified and several molecular and cellular therapy is proposed, a clear understanding of underlying biomolecular mechanisms still underpins the basis of management of inherited renal hypomagnesemia.

Conflicts of interest

The authors declare no conflicts of interest.

Acknowledgements

This work was supported by research fund of the Ministry of Science and Technology (MOST109-2314-B-182A-134-MY3).

Footnotes

Peer review under responsibility of Chang Gung University.

References

  • 1.Li F.Y., Chaigne-Delalande B., Kanellopoulou C., Davis J.C., Matthews H.F., Douek D.C., et al. Second messenger role for Mg 2+ revealed by human T-cell immunodeficiency. Nature. 2011;475:471–476. doi: 10.1038/nature10246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.de Baaij J.H.F., Hoenderop J.G., Bindels R.J. Magnesium in man: implications for health and disease. Physiol Rev. 2015;95:1–46. doi: 10.1152/physrev.00012.2014. [DOI] [PubMed] [Google Scholar]
  • 3.Schimatschek H.F., Rempis R. Prevalence of hypomagnesemia in an unselected German population of 16,000 individuals. Magnes Res. 2001;14:283–290. [PubMed] [Google Scholar]
  • 4.Chernow B., Bamberger S., Stoiko M., Vadnais M., Mills S., Hoellerich V., et al. Hypomagnesemia in patients in postoperative intensive care. Chest. 1989;95:391–397. doi: 10.1378/chest.95.2.391. [DOI] [PubMed] [Google Scholar]
  • 5.Escuela M.P., Guerra M., Añón J.M., Martínez-Vizcaíno V., Zapatero M.D., García-Jalón A., et al. Total and ionized serum magnesium in critically ill patients. Intensive Care Med. 2005;31:151–156. doi: 10.1007/s00134-004-2508-x. [DOI] [PubMed] [Google Scholar]
  • 6.Yamazaki D., Funato Y., Miura J., Sato S., Toyosawa S., Furutani K., et al. Basolateral Mg2+ extrusion via CNNM4 mediates transcellular Mg2+ transport across epithelia: a mouse model. PLoS Genet. 2013;9 doi: 10.1371/journal.pgen.1003983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Le Grimellec C., Giocondi M.C., Philippe P. Micropuncture study along the proximal convoluted tubule electrolyte reabsorption in first convolutions. Pflügers Arch Eur J Physiol. 1975;354:133–150. doi: 10.1007/BF00579944. [DOI] [PubMed] [Google Scholar]
  • 8.Quamme G.A., Dirks J.H. The physiology of renal magnesium handling. Ren Physiol. 1986;9:257–269. doi: 10.1159/000173090. [DOI] [PubMed] [Google Scholar]
  • 9.Schlingmann K.P., Renigunta A., Hoorn E.J., Forst A.L., Renigunta V., Atanasov V., et al. Defects in KCNJ16 cause a novel tubulopathy with hypokalemia, salt wasting, disturbed acid-base homeostasis, and sensorineural deafness. J Am Soc Nephrol. 2021;32:1498–1512. doi: 10.1681/ASN.2020111587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Stuiver M., Lainez S., Will C., Terryn S., Günzel D., Debaix H., et al. CNNM2, encoding a basolateral protein required for renal Mg2+ handling, is mutated in dominant hypomagnesemia. Am J Hum Genet. 2011;88:333–343. doi: 10.1016/j.ajhg.2011.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.De Baaij J.H.F., Stuiver M., Meij I.C., Lainez S., Kopplin K., Venselaar H., et al. Membrane topology and intracellular processing of cyclin M2 (CNNM2) J Biol Chem. 2012;287:13644–13655. doi: 10.1074/jbc.M112.342204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Morita K., Furuse M., Fujimoto K., Tsukita S. Claudin multigene family encoding four-transmembrane domain protein components of tight junction strands. Proc Natl Acad Sci Unit States Am. 1999;96:511–516. doi: 10.1073/pnas.96.2.511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Breiderhoff T., Himmerkus N., Stuiver M., Mutig K., Will C., Meij I.C., et al. Deletion of claudin-10 (Cldn10) in the thick ascending limb impairs paracellular sodium permeability and leads to hypermagnesemia and nephrocalcinosis. Proc Natl Acad Sci U S A. 2012;109:14241–14246. doi: 10.1073/pnas.1203834109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Thebault S., Alexander R.T., Tiel Groenestege W.M., Hoenderop J.G., Bindels R.J. EGF increases TRPM6 activity and surface expression. J Am Soc Nephrol. 2009;20:78–85. doi: 10.1681/ASN.2008030327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ferrè S., Veenstra G.J.C., Bouwmeester R., Hoenderop J.G.J., Bindels R.J.M. HNF-1B specifically regulates the transcription of the γ-subunit of the Na+/K+-ATPase. Biochem Biophys Res Commun. 2011;404:284–290. doi: 10.1016/j.bbrc.2010.11.108. [DOI] [PubMed] [Google Scholar]
  • 16.Schlingmann K.P., Jouret F., Shen K., Nigam A., Arjona F.J., Dafinger C., et al. mTOR-activating mutations in RRAGD are causative for kidney tubulopathy and cardiomyopathy. J Am Soc Nephrol. 2021;32:2885–2899. doi: 10.1681/ASN.2021030333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Nijenhuis T., Vallon V., Van Der Kemp A.W.C.M., Loffing J., Hoenderop J.G.J., Bindels R.J.M. Enhanced passive Ca2+ reabsorption and reduced Mg2+ channel abundance explains thiazide-induced hypocalciuria and hypomagnesemia. J Clin Invest. 2005;115:1651–1658. doi: 10.1172/JCI24134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Glaudemans B., Van Der Wijst J., Scola R.H., Lorenzoni P.J., Heister A., Van Der Kemp A.M.W., 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;119:936–942. doi: 10.1172/JCI36948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Groenestege W.M.T., Thébault S., van der Wijst J., van den Berg D., Janssen R., Tejpar S., et al. Impaired basolateral sorting of pro-EGF causes isolated recessive renal hypomagnesemia. J Clin Invest. 2007;117:2260–2267. doi: 10.1172/JCI31680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Meij I.C., Koenderink J.B., van Bokhoven H., Assink K.F.H., Tiel Groenestege W., de Pont J.J.H.H.M., et al. Dominant isolated renal magnesium loss is caused by misrouting of the Na+,K+-ATPase γ-subunit. Nat Genet. 2000;26:265–266. doi: 10.1038/81543. [DOI] [PubMed] [Google Scholar]
  • 21.Lücking K., Nielsen J.M., Pedersen P.A., Jørgensen P.L. Na-K-ATPase isoform (alpha 3, alpha 2, alpha 1) abundance in rat kidney estimated by competitive RT-PCR and ouabain binding. Am J Physiol. 1996;271:F253–F260. doi: 10.1152/ajprenal.1996.271.2.F253. [DOI] [PubMed] [Google Scholar]
  • 22.Schlingmann K.P., Bandulik S., Mammen C., Tarailo-Graovac M., Holm R., Baumann M., et al. Germline de novo mutations in ATP1A1 cause renal hypomagnesemia, refractory seizures, and intellectual disability. Am J Hum Genet. 2018;103:808–816. doi: 10.1016/j.ajhg.2018.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ferre S., de Baaij J.H.F., Ferreira P., Germann R., de Klerk J.B.C., Lavrijsen M., et al. Mutations in PCBD1 cause hypomagnesemia and renal magnesium wasting. J Am Soc Nephrol. 2014;25:574–586. doi: 10.1681/ASN.2013040337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Scholl U.I., Choi M., Liu T., Ramaekers V.T., Häusler M.G., Grimmer J., et al. Seizures, sensorineural deafness, ataxia, mental retardation, and electrolyte imbalance (SeSAME syndrome) caused by mutations in KCNJ10. Proc Natl Acad Sci Unit States Am. 2009;106:5842–5847. doi: 10.1073/pnas.0901749106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Bockenhauer D., Feather S., Stanescu H.C., Bandulik S., Zdebik A.A., Reichold M., et al. Epilepsy, ataxia, sensorineural deafness, tubulopathy, and KCNJ10 mutations. N Engl J Med. 2009;360:1960–1970. doi: 10.1056/NEJMoa0810276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kolisek M., Nestler A., Vormann J., Schweigel-Röntgen M. Human gene SLC41A1 encodes for the Na +/Mg 2+ exchanger. Am J Physiol Cell Physiol. 2012;302:C318–C326. doi: 10.1152/ajpcell.00289.2011. [DOI] [PubMed] [Google Scholar]
  • 27.Arjona F.J., de Baaij J.H.F. CrossTalk opposing view: CNNM proteins are not Na +/Mg 2+ exchangers but Mg 2+ transport regulators playing a central role in transepithelial Mg 2+ (re)absorption. J Physiol. 2018;596:747–750. doi: 10.1113/JP275249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Mayer M.L., Westbrook G.L., Guthrie P.B. Voltage-dependent block by Mg2+ of NMDA responses in spinal cord neurones. Nature. 1984;309:261–263. doi: 10.1038/309261a0. [DOI] [PubMed] [Google Scholar]
  • 29.Möykkynen T., Uusi-Oukari M., Heikkilä J., Lovinger D.M., Lüddens H., Korpi E.R. Magnesium potentiation of the function of native and recombinant GABAA receptors. Neuroreport. 2001;12:2175–2179. doi: 10.1097/00001756-200107200-00026. [DOI] [PubMed] [Google Scholar]
  • 30.Tin A., Grams M.E., Maruthur N.M., Astor B.C., Couper D., Mosley T.H., et al. Results from the Atherosclerosis Risk in Communities study suggest that low serum magnesium is associated with incident kidney disease. Kidney Int. 2015;87:820–827. doi: 10.1038/ki.2014.331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Pham P.C.T., Pham P.M.T., Pham S.V., Miller J.M., Pham P.T.T. Hypomagnesemia in patients with type 2 diabetes. Clin J Am Soc Nephrol. 2007;2:366–373. doi: 10.2215/CJN.02960906. [DOI] [PubMed] [Google Scholar]
  • 32.He K., Liu K., Daviglus M.L., Morris S.J., Loria C.M., Van Horn L., et al. Magnesium intake and incidence of metabolic syndrome among young adults. Circulation. 2006;113:1675–1682. doi: 10.1161/CIRCULATIONAHA.105.588327. [DOI] [PubMed] [Google Scholar]
  • 33.Teragawa H., Matsuura H., Chayama K., Oshima T. Mechanisms responsible for vasodilation upon magnesium infusion in vivo: clinical evidence. Magnes Res. 2002;15:241–246. [PubMed] [Google Scholar]
  • 34.Weglicki W.B., Mak I.T., Phillips T.M. Blockade of cardiac inflammation in Mg2+ deficiency by substance P receptor inhibition. Circ Res. 1994;74:1009–1013. doi: 10.1161/01.res.74.5.1009. [DOI] [PubMed] [Google Scholar]
  • 35.Cheng P.T., Grabher J.J., LeGeros R.Z. Effects of magnesium on calcium phosphate formation. Magnesium. 1988;7:123–132. [PubMed] [Google Scholar]
  • 36.Mubagwa K., Gwanyanya A., Zakharov S., Macianskiene R. Regulation of cation channels in cardiac and smooth muscle cells by intracellular magnesium. Arch Biochem Biophys. 2007;458:73–89. doi: 10.1016/j.abb.2006.10.014. [DOI] [PubMed] [Google Scholar]
  • 37.Greising S.M., Gransee H.M., Mantilla C.B., Sieck G.C. Systems biology of skeletal muscle: fiber type as an organizing principle. Wiley Interdiscip Rev Syst Biol Med. 2012;4:457–473. doi: 10.1002/wsbm.1184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Gür A., Çolpan L., Nas K., Çevik R., Saraç J., Erdoǧan F., et al. The role of trace minerals in the pathogenesis of postmenopausal osteoporosis and a new effect of calcitonin. J Bone Miner Metabol. 2002;20:39–43. doi: 10.1007/s774-002-8445-y. [DOI] [PubMed] [Google Scholar]
  • 39.Murakami M., Ishizuka J., Sumi S., Nickols G.A., Cooper C.W., Townsend C.M., et al. Role of extracellular magnesium in insulin secretion from rat insulinoma cells. Proc Soc Exp Biol Med. 1992;200:490–494. doi: 10.3181/00379727-200-43459. [DOI] [PubMed] [Google Scholar]
  • 40.Rodríguez-Morán M., Guerrero-Romero F. Insulin secretion is decreased in non-diabetic individuals with hypomagnesaemia. Diabetes Metab Res Rev. 2011;27:590–596. doi: 10.1002/dmrr.1206. [DOI] [PubMed] [Google Scholar]
  • 41.Koivisto M., Valta P., Höckerstedt K., Lindgren L. Magnesium depletion in chronic terminal liver cirrhosis. Clin Transplant. 2002;16:325–328. doi: 10.1034/j.1399-0012.2002.01141.x. [DOI] [PubMed] [Google Scholar]
  • 42.Feske S., Skolnik E.Y., Prakriya M. Ion channels and transporters in lymphocyte function and immunity. Nat Rev Immunol. 2012;12:532–547. doi: 10.1038/nri3233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lo Y.F., Nozu K., Iijima K., Morishita T., Huang C.C., Yang S Sen, et al. Recurrent deep intronic mutations in the SLC12A3 gene responsible for Gitelman's syndrome. Clin J Am Soc Nephrol. 2011;6:630–639. doi: 10.2215/CJN.06730810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Heidet L., Decramer S., Pawtowski A., Morinière V., Bandin F., Knebelmann B., et al. Spectrum of HNF1B mutations in a large cohort of patients who harbor renal diseases. Clin J Am Soc Nephrol. 2010;5:1079–1090. doi: 10.2215/CJN.06810909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.McEvoy G.K. American Society of Health System Pharmacists; Bethesda, MD: 1998. AHFS drug information. [Google Scholar]
  • 46.Guerrera M.P., Volpe S.L., Mao J.J. Therapeutic uses of magnesium. Am Fam Physician. 2009;80:157–162. [PubMed] [Google Scholar]
  • 47.Knoers N.V.A.M., Levtchenko E.N. Gitelman syndrome. Orphanet J Rare Dis. 2008;3:22. doi: 10.1186/1750-1172-3-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Barr C.S., Lang C.C., Hanson J., Arnott M., Kennedy N., Struthers A.D. Effects of adding spironolactone to an angiotensin-converting enzyme inhibitor in chronic congestive heart failure secondary to coronary artery disease. Am J Cardiol. 1995;76:1259–1265. doi: 10.1016/s0002-9149(99)80353-1. [DOI] [PubMed] [Google Scholar]
  • 49.Gao X., Peng L., Adhikari C.M., Lin J., Zuo Z. Spironolactone reduced arrhythmia and maintained magnesium homeostasis in patients with congestive heart failure. J Card Fail. 2007;13:170–177. doi: 10.1016/j.cardfail.2006.11.015. [DOI] [PubMed] [Google Scholar]
  • 50.Sontia B., Montezano A.C.I., Paravicini T., Tabet F., Touyz R.M. Downregulation of renal TRPM7 and increased inflammation and fibrosis in aldosterone-infused mice: effects of magnesium. Hypertension. 2008;51:915–921. doi: 10.1161/HYPERTENSIONAHA.107.100339. [DOI] [PubMed] [Google Scholar]
  • 51.Zuris J.A., Thompson D.B., Shu Y., Guilinger J.P., Bessen J.L., Hu J.H., et al. Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nat Biotechnol. 2015;33:73–80. doi: 10.1038/nbt.3081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Bennett J. Taking stock of retinal gene therapy: looking back and moving forward. Mol Ther. 2017;25:1076–1094. doi: 10.1016/j.ymthe.2017.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Heikkilä P., Tibell A., Morita T., Chen Y., Wu G., Sado Y., et al. Adenovirus-mediated transfer of type IV collagen α5 chain cDNA into swine kidney in vivo: deposition of the protein into the glomerular basement membrane. Gene Ther. 2001;8:882–890. doi: 10.1038/sj.gt.3301342. [DOI] [PubMed] [Google Scholar]
  • 54.Gaj T., Gersbach C.A., Barbas C.F. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol. 2013;31:397–405. doi: 10.1016/j.tibtech.2013.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Yao X., Wang X., Hu X., Liu Z., Liu J., Zhou H., et al. Homology-mediated end joining-based targeted integration using CRISPR/Cas9. Cell Res. 2017;27:801–814. doi: 10.1038/cr.2017.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Daga S., Donati F., Capitani K., Croci S., Tita R., Giliberti A., et al. New frontiers to cure Alport syndrome: COL4A3 and COL4A5 gene editing in podocyte-lineage cells. Eur J Hum Genet. 2020;28:480–490. doi: 10.1038/s41431-019-0537-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Hammond S.M., Wood M.J.A. Genetic therapies for RNA mis-splicing diseases. Trends Genet. 2011;27:196–205. doi: 10.1016/j.tig.2011.02.004. [DOI] [PubMed] [Google Scholar]
  • 58.Tanner G., Glaus E., Barthelmes D., Ader M., Fleischhauer J., Pagani F., et al. Therapeutic strategy to rescue mutation-induced exon skipping in rhodopsin by adaptation of U1 snRNA. Hum Mutat. 2009;30:255–263. doi: 10.1002/humu.20861. [DOI] [PubMed] [Google Scholar]
  • 59.Berger A., Lorain S., Joséphine C., Desrosiers M., Peccate C., Voit T., et al. Repair of rhodopsin mRNA by spliceosome-mediated RNA trans-splicing: a new approach for autosomal dominant retinitis pigmentosa. Mol Ther. 2015;23:918–930. doi: 10.1038/mt.2015.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ramsbottom S.A., Molinari E., Srivastava S., Silberman F., Henry C., Alkanderi S., et al. Targeted exon skipping of a CEP290 mutation rescues Joubert syndrome phenotypes in vitro and in a murine model. Proc Natl Acad Sci U S A. 2018;115:12489–12494. doi: 10.1073/pnas.1809432115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Nagel-Wolfrum K., Möller F., Penner I., Baasov T., Wolfrum U. Targeting nonsense mutations in diseases with translational read-through-inducing drugs (TRIDs) BioDrugs. 2016;30:49–74. doi: 10.1007/s40259-016-0157-6. [DOI] [PubMed] [Google Scholar]
  • 62.Fang Y.W., Yang S.S., Chau T., Nakamura M., Yamazaki O., Seki G., et al. Therapeutic effect of prenatal alkalization and PTC124 in Na +/HCO 3 - cotransporter 1 p.W516∗ knock-in mice. Gene Ther. 2015;22:374–381. doi: 10.1038/gt.2015.7. [DOI] [PubMed] [Google Scholar]
  • 63.Mollura D.J., Hare J.M., Rabb H. Stem-cell therapy for renal diseases. Am J Kidney Dis. 2003;42:891–905. doi: 10.1016/j.ajkd.2003.07.018. [DOI] [PubMed] [Google Scholar]
  • 64.Lu Y., Xue J., Deng T., Zhou X., Yu K., Deng L., et al. Safety and feasibility of CRISPR-edited T cells in patients with refractory non-small-cell lung cancer. Nat Med. 2020;26:732–740. doi: 10.1038/s41591-020-0840-5. [DOI] [PubMed] [Google Scholar]
  • 65.Lienhardt A., Garabédian M., Bai M., Sinding C., Zhang Z., Lagarde J.P., et al. A large homozygous or heterozygous in-frame deletion within the calcium-sensing receptor's carboxylterminal cytoplasmic tail that causes autosomal dominant hypocalcemia. J Clin Endocrinol Metab. 2000;85:1695–1702. doi: 10.1210/jcem.85.4.6570. [DOI] [PubMed] [Google Scholar]
  • 66.García-Castaño A., Perdomo-Ramirez A., Vall-Palomar M., Ramos-Trujillo E., Madariaga L., Ariceta G., et al. Novel compound heterozygous mutations of CLDN16 in a patient with familial hypomagnesemia with hypercalciuria and nephrocalcinosis. Mol Genet Genomic Med. 2020;8:1–10. doi: 10.1002/mgg3.1475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Yamaguti P.M., Dos Santos P.A.C., Leal B.S., De Mello Santana V.B.B., Mazzeu J.F., Acevedo A.C., et al. Identification of the first large deletion in the CLDN16 gene in a patient with FHHNC and late-onset of chronic kidney disease: case report. BMC Nephrol. 2015;16:1–6. doi: 10.1186/s12882-015-0079-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Godron A., Harambat J., Boccio V., Mensire A., May A., Rigothier C., 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;7:801–809. doi: 10.2215/CJN.12841211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Claverie-Martín F., Vargas-Poussou R., Müller D., García-Nieto V. Clinical utility gene card for: familial hypomagnesemia with hypercalciuria and nephrocalcinosis with/without severe ocular involvement. Eur J Hum Genet. 2015;23:e1–e4. doi: 10.1038/ejhg.2014.176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Han Y., Lin Y., Sun Q., Wang S., Gao Y., Shao L. Mutation spectrum of Chinese patients with Bartter syndrome. Oncotarget. 2017;8:101614–101622. doi: 10.18632/oncotarget.21355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Seys E., Andrini O., Keck M., Mansour-Hendili L., Courand P., Simian C., et al. Clinical and genetic spectrum of Bartter syndrome type 3. J Am Soc Nephrol. 2017;28:2540–2552. doi: 10.1681/ASN.2016101057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Gorgojo J.J., Donnay S., Jeck N., Konrad M. A Spanish founder mutation in the chloride channel gene, CLCNKB, as a cause of atypical Bartter syndrome in adult age. Horm Res. 2006;65:62–68. doi: 10.1159/000090601. [DOI] [PubMed] [Google Scholar]
  • 73.Nozu K., Fu X.J., Nakanishi K., Yoshikawa N., Kaito H., Kanda K., et al. Molecular analysis of patients with type III Bartter syndrome: picking up large heterozygous deletions with semiquantitative PCR. Pediatr Res. 2007;62:364–369. doi: 10.1203/PDR.0b013e318123fb90. [DOI] [PubMed] [Google Scholar]
  • 74.Birkenhäger R., Otto E., Schürmann M.J., Vollmer M., Ruf E.M., Maier-Lutz I., et al. Mutation of BSND causes Bartter syndrome with sensorineural deafness and kidney failure. Nat Genet. 2001;29:310–314. doi: 10.1038/ng752. [DOI] [PubMed] [Google Scholar]
  • 75.Nozu K., Inagaki T., Fu X.J., Nozu Y., Kaito H., Kanda K., et al. Molecular analysis of digenic inheritance in Bartter syndrome with sensorineural deafness. J Med Genet. 2008;45:182–186. doi: 10.1136/jmg.2007.052944. [DOI] [PubMed] [Google Scholar]
  • 76.Schlingmann K.P., Konrad M., Jeck N., Waldegger P., Reinalter S.C., Holder M., et al. Salt wasting and deafness resulting from mutations in two chloride channels. N Engl J Med. 2004;350:1314–1319. doi: 10.1056/NEJMoa032843. [DOI] [PubMed] [Google Scholar]
  • 77.Schlingmann K.P., Jouret F., Shen K., Nigam A., Arjona F.J., Dafinger C., et al. mTOR-activating mutations in RRAGD are causative for kidney tubulopathy and cardiomyopathy. J Am Soc Nephrol. 2021;32:2885–2899. doi: 10.1681/ASN.2021030333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Li H.J., Groden C., Hoenig M.P., Ray E.C., Ferreira C.R., Gahl W., et al. Case report: extreme coronary calcifications and hypomagnesemia in a patient with a 17q12 deletion involving HNF1B. BMC Nephrol. 2019;20:1–6. doi: 10.1186/s12882-019-1533-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Van Der Made C.I., Hoorn E.J., De La Faille R., Karaaslan H., Knoers N.V.A.M., Hoenderop J.G.J., et al. Hypomagnesemia as first clinical manifestation of ADTKD-HNF1B: a case series and literature review. Am J Nephrol. 2015;42:85–90. doi: 10.1159/000439286. [DOI] [PubMed] [Google Scholar]
  • 80.Faguer S., Decramer S., Chassaing N., Bellanné-Chantelot C., Calvas P., Beaufils S., et al. Diagnosis, management, and prognosis of HNF1B nephropathy in adulthood. Kidney Int. 2011;80:768–776. doi: 10.1038/ki.2011.225. [DOI] [PubMed] [Google Scholar]
  • 81.Okorn C., Goertz A., Vester U., Beck B.B., Bergmann C., Habbig S., et al. HNF1B nephropathy has a slow-progressive phenotype in childhood—with the exception of very early onset cases: results of the German Multicenter HNF1B Childhood Registry. Pediatr Nephrol. 2019;34:1065–1075. doi: 10.1007/s00467-018-4188-8. [DOI] [PubMed] [Google Scholar]
  • 82.Celmina M., Micule I., Inashkina I., Audere M., Kuske S., Pereca J., et al. EAST/SeSAME syndrome: review of the literature and introduction of four new Latvian patients. Clin Genet. 2019;95:63–78. doi: 10.1111/cge.13374. [DOI] [PubMed] [Google Scholar]
  • 83.Abdelhadi O., Iancu D., Tekman M., Stanescu H., Bockenhauer D., Kleta R. Founder mutation in KCNJ10 in Pakistani patients with EAST syndrome. Mol Genet Genomic Med. 2016;4:521–526. doi: 10.1002/mgg3.227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Himmelreich N., Blau N., Thöny B. Molecular and metabolic bases of tetrahydrobiopterin (BH4) deficiencies. Mol Genet Metabol. 2021;133:123–136. doi: 10.1016/j.ymgme.2021.04.003. [DOI] [PubMed] [Google Scholar]
  • 85.Vargas-Poussou R., Dahan K., Kahila D., Venisse A., Riveira-Munoz E., Debaix H., et al. Spectrum of mutations in Gitelman syndrome. J Am Soc Nephrol. 2011;22:693–703. doi: 10.1681/ASN.2010090907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Nozu K., Iijima K., Nozu Y., Ikegami E.I., Imai T., Fu X.J., et al. A deep intronic mutation in the SLC12A3 gene leads to gitelman syndrome. Pediatr Res. 2009;66:590–593. doi: 10.1203/PDR.0b013e3181b9b4d3. [DOI] [PubMed] [Google Scholar]
  • 87.Nozu K., Nozu Y., Nakanishi K., Konomoto T., Horinouchi T., Shono A., et al. Cryptic exon activation in SLC12A3 in Gitelman syndrome. J Hum Genet. 2017;62:335–337. doi: 10.1038/jhg.2016.129. [DOI] [PubMed] [Google Scholar]
  • 88.Chinen T., Saeki E., Mori T., Sohara E., Uchida S., Akimoto T. A case of Gitelman syndrome: our experience with a patient treated in clinical practice on a local island. J Rural Med. 2019;14:258–262. doi: 10.2185/jrm.3014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Glaudemans B., Yntema H.G., San-Cristobal P., Schoots J., Pfundt R., Kamsteeg E.J., et al. Novel NCC mutants and functional analysis in a new cohort of patients with Gitelman syndrome. Eur J Hum Genet. 2012;20:263–270. doi: 10.1038/ejhg.2011.189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.De Baaij J.H.F., Dorresteijn E.M., Hennekam E.A.M., Kamsteeg E.J., Meijer R., Dahan K., et al. Recurrent FXYD2 p.Gly41Arg mutation in patients with isolated dominant hypomagnesaemia. Nephrol Dial Transplant. 2015;30:952–957. doi: 10.1093/ndt/gfv014. [DOI] [PubMed] [Google Scholar]
  • 91.Franken G.A.C., Müller D., Mignot C., Keren B., Lévy J., Tabet A., et al. The phenotypic and genetic spectrum of patients with heterozygous mutations in cyclin M2 (CNNM2) Hum Mutat. 2021;42:473–486. doi: 10.1002/humu.24182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Isojima T., Doi K., Mitsui J., Oda Y., Tokuhiro E., Yasoda A., et al. A recurrent de novo FAM111A mutation causes kenny-caffey syndrome type 2. J Bone Miner Res. 2014;29:992–998. doi: 10.1002/jbmr.2091. [DOI] [PubMed] [Google Scholar]
  • 93.Unger S., Górna M.W., Le Béchec A., Do Vale-Pereira S., Bedeschi M.F., Geiberger S., et al. FAM111A mutations result in hypoparathyroidism and impaired skeletal development. Am J Hum Genet. 2013;92:990–995. doi: 10.1016/j.ajhg.2013.04.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Paulhus K., Ammerman L., Glasscock E. Clinical spectrum of KCNA1 mutations: new insights into episodic ataxia and epilepsy comorbidity. Int J Mol Sci. 2020;21:2802. doi: 10.3390/ijms21082802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Schlingmann K.P., Sassen M.C., Weber S., Pechmann U., Kusch K., Pelken L., et al. Novel TRPM6 mutations in 21 families with primary hypomagnesemia and secondary hypocalcemia. J Am Soc Nephrol. 2005;16:3061–3069. doi: 10.1681/ASN.2004110989. [DOI] [PubMed] [Google Scholar]
  • 96.Naeem M., Hussain S., Akhtar N. Mutation in the tight-junction gene claudin 19 (CLDN19) and familial hypomagnesemia, hypercalciuria, nephrocalcinosis (FHHNC) and severe ocular disease. Am J Nephrol. 2011;34:241–248. doi: 10.1159/000330854. [DOI] [PubMed] [Google Scholar]
  • 97.Lemos M., Gonçalves J.S., Correia C.R., Maria A.T. Neonatal inflammatory skin and bowel disease type 2: a very rare disease associated with EGFR mutation. J Pediatr Neonatal Individ Med. 2021;10:1–5. [Google Scholar]
  • 98.Wilson F.H., Hariri A., Farhi A., Zhao H., Petersen K.F., Toka H.R., et al. A cluster of metabolic defects caused by mutation in a mitochondrial tRNA. Science. 2004;306:1190–1194. doi: 10.1126/science.1102521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Viering D., Schlingmann K.P., Hureaux M., Nijenhuis T., Mallett A., Chan M.M.Y., et al. Gitelman syndrome phenocopy caused by pathogenic variants in mtDNA. J Am Soc Nephrol. 2022;33:305–325. doi: 10.1681/ASN.2021050596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Rivera H., Martín-Hernández E., Delmiro A., García-Silva M.T., Quijada-Fraile P., Muley R., et al. A new mutation in the gene encoding mitochondrial seryl-tRNA synthetase as a cause of HUPRA syndrome. BMC Nephrol. 2013;14:195. doi: 10.1186/1471-2369-14-195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Sabella-Jiménez V., Otero-Herrera C., Silvera-Redondo C., Garavito-Galofre P. Mitochondrial DNA deletion and duplication in kearns–sayre syndrome (KSS) with initial presentation as pearson marrow-pancreas syndrome (PMPS): two case reports in barranquilla, Colombia. Mol Genet Genomic Med. 2020;8:e1509. doi: 10.1002/mgg3.1509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Katsanos K.H., Elisaf M., Bairaktari E., Tsianos E.V. Severe hypomagnesemia and hypoparathyroidism in Kearns-Sayre syndrome. Am J Nephrol. 2001;21:150–153. doi: 10.1159/000046239. [DOI] [PubMed] [Google Scholar]
  • 103.Maceluch J.A., Niedziela M. The clinical diagnosis and molecular genetics of kearns-sayre syndrome: a complex mitochondrial encephalomyopathy. Pediatr Endocrinol Rev. 2006;4:117–137. [PubMed] [Google Scholar]

Articles from Biomedical Journal are provided here courtesy of Chang Gung University

RESOURCES