Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Am Soc Nephrol ; 33(2): 305-325, 2022 02.
Article in English | MEDLINE | ID: mdl-34607911

ABSTRACT

BACKGROUND: Gitelman syndrome is the most frequent hereditary salt-losing tubulopathy characterized by hypokalemic alkalosis and hypomagnesemia. Gitelman syndrome is caused by biallelic pathogenic variants in SLC12A3, encoding the Na+-Cl- cotransporter (NCC) expressed in the distal convoluted tubule. Pathogenic variants of CLCNKB, HNF1B, FXYD2, or KCNJ10 may result in the same renal phenotype of Gitelman syndrome, as they can lead to reduced NCC activity. For approximately 10 percent of patients with a Gitelman syndrome phenotype, the genotype is unknown. METHODS: We identified mitochondrial DNA (mtDNA) variants in three families with Gitelman-like electrolyte abnormalities, then investigated 156 families for variants in MT-TI and MT-TF, which encode the transfer RNAs for phenylalanine and isoleucine. Mitochondrial respiratory chain function was assessed in patient fibroblasts. Mitochondrial dysfunction was induced in NCC-expressing HEK293 cells to assess the effect on thiazide-sensitive 22Na+ transport. RESULTS: Genetic investigations revealed four mtDNA variants in 13 families: m.591C>T (n=7), m.616T>C (n=1), m.643A>G (n=1) (all in MT-TF), and m.4291T>C (n=4, in MT-TI). Variants were near homoplasmic in affected individuals. All variants were classified as pathogenic, except for m.643A>G, which was classified as a variant of uncertain significance. Importantly, affected members of six families with an MT-TF variant additionally suffered from progressive chronic kidney disease. Dysfunction of oxidative phosphorylation complex IV and reduced maximal mitochondrial respiratory capacity were found in patient fibroblasts. In vitro pharmacological inhibition of complex IV, mimicking the effect of the mtDNA variants, inhibited NCC phosphorylation and NCC-mediated sodium uptake. CONCLUSION: Pathogenic mtDNA variants in MT-TF and MT-TI can cause a Gitelman-like syndrome. Genetic investigation of mtDNA should be considered in patients with unexplained Gitelman syndrome-like tubulopathies.


Subject(s)
DNA, Mitochondrial/genetics , Gitelman Syndrome/genetics , Mutation , Adolescent , Adult , Aged , Base Sequence , Child , Child, Preschool , Female , Genotype , Gitelman Syndrome/metabolism , Gitelman Syndrome/pathology , HEK293 Cells , Humans , Infant , Kidney/metabolism , Kidney/ultrastructure , Male , Middle Aged , Mitochondria/metabolism , Models, Biological , Nucleic Acid Conformation , Pedigree , Phenotype , Polymorphism, Single Nucleotide , RNA, Transfer, Ile/chemistry , RNA, Transfer, Ile/genetics , RNA, Transfer, Phe/chemistry , RNA, Transfer, Phe/genetics , Solute Carrier Family 12, Member 3/genetics , Young Adult
2.
Curr Opin Nephrol Hypertens ; 31(5): 508-515, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35894287

ABSTRACT

PURPOSE OF REVIEW: Gitelman syndrome is a recessive salt-wasting disorder characterized by hypomagnesemia, hypokalemia, metabolic alkalosis and hypocalciuria. The majority of patients are explained by mutations and deletions in the SLC12A3 gene, encoding the Na+-Cl--co-transporter (NCC). Recently, additional genetic causes of Gitelman-like syndromes have been identified that should be considered in genetic screening. This review aims to provide a comprehensive overview of the clinical, genetic and mechanistic aspects of Gitelman(-like) syndromes. RECENT FINDINGS: Disturbed Na+ reabsorption in the distal convoluted tubule (DCT) is associated with hypomagnesemia and hypokalemic alkalosis. In Gitelman syndrome, loss-of-function mutations in SLC12A3 cause impaired NCC-mediated Na+ reabsorption. In addition, patients with mutations in CLCKNB, KCNJ10, FXYD2 or HNF1B may present with a similar phenotype, as these mutations indirectly reduce NCC activity. Furthermore, genetic investigations of patients with Na+-wasting tubulopathy have resulted in the identification of pathogenic variants in MT-TI, MT-TF, KCNJ16 and ATP1A1. These novel findings highlight the importance of cell metabolism and basolateral membrane potential for Na+ reabsorption in the DCT. SUMMARY: Altogether, these findings extend the genetic spectrum of Gitelman-like electrolyte alterations. Genetic testing of patients with hypomagnesemia and hypokalemia should cover a panel of genes involved in Gitelman-like syndromes, including the mitochondrial genome.


Subject(s)
Alkalosis , Bartter Syndrome , Gitelman Syndrome , Hypokalemia , Alkalosis/complications , Alkalosis/genetics , Bartter Syndrome/genetics , Gitelman Syndrome/complications , Gitelman Syndrome/genetics , Humans , Hypokalemia/genetics , Magnesium/metabolism , Sodium/metabolism , Solute Carrier Family 12, Member 3/genetics
3.
J Am Soc Nephrol ; 32(11): 2885-2899, 2021 11.
Article in English | MEDLINE | ID: mdl-34607910

ABSTRACT

BACKGROUND: Over the last decade, advances in genetic techniques have resulted in the identification of rare hereditary disorders of renal magnesium and salt handling. Nevertheless, approximately 20% of all patients with tubulopathy lack a genetic diagnosis. METHODS: We performed whole-exome and -genome sequencing of a patient cohort with a novel, inherited, salt-losing tubulopathy; hypomagnesemia; and dilated cardiomyopathy. We also conducted subsequent in vitro functional analyses of identified variants of RRAGD, a gene that encodes a small Rag guanosine triphosphatase (GTPase). RESULTS: In eight children from unrelated families with a tubulopathy characterized by hypomagnesemia, hypokalemia, salt wasting, and nephrocalcinosis, we identified heterozygous missense variants in RRAGD that mostly occurred de novo. Six of these patients also had dilated cardiomyopathy and three underwent heart transplantation. We identified a heterozygous variant in RRAGD that segregated with the phenotype in eight members of a large family with similar kidney manifestations. The GTPase RagD, encoded by RRAGD, plays a role in mediating amino acid signaling to the mechanistic target of rapamycin complex 1 (mTORC1). RagD expression along the mammalian nephron included the thick ascending limb and the distal convoluted tubule. The identified RRAGD variants were shown to induce a constitutive activation of mTOR signaling in vitro. CONCLUSIONS: Our findings establish a novel disease, which we call autosomal dominant kidney hypomagnesemia (ADKH-RRAGD), that combines an electrolyte-losing tubulopathy and dilated cardiomyopathy. The condition is caused by variants in the RRAGD gene, which encodes Rag GTPase D; these variants lead to an activation of mTOR signaling, suggesting a critical role of Rag GTPase D for renal electrolyte handling and cardiac function.


Subject(s)
Cardiomyopathy, Dilated/genetics , Hypercalciuria/genetics , Kidney Diseases/genetics , Monomeric GTP-Binding Proteins/genetics , Mutation, Missense , Nephrocalcinosis/genetics , Renal Tubular Transport, Inborn Errors/genetics , TOR Serine-Threonine Kinases/metabolism , Cardiomyopathy, Dilated/metabolism , Female , HEK293 Cells , Humans , Hypercalciuria/metabolism , Kidney Diseases/metabolism , Kidney Tubules, Distal/metabolism , Male , Models, Molecular , Natriuresis/genetics , Nephrocalcinosis/metabolism , Pedigree , Protein Conformation , Renal Tubular Transport, Inborn Errors/metabolism , Seizures/genetics , Seizures/metabolism , Signal Transduction , Exome Sequencing , Whole Genome Sequencing
4.
J Am Soc Nephrol ; 32(6): 1498-1512, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33811157

ABSTRACT

BACKGROUND: The transepithelial transport of electrolytes, solutes, and water in the kidney is a well-orchestrated process involving numerous membrane transport systems. Basolateral potassium channels in tubular cells not only mediate potassium recycling for proper Na+,K+-ATPase function but are also involved in potassium and pH sensing. Genetic defects in KCNJ10 cause EAST/SeSAME syndrome, characterized by renal salt wasting with hypokalemic alkalosis associated with epilepsy, ataxia, and sensorineural deafness. METHODS: A candidate gene approach and whole-exome sequencing determined the underlying genetic defect in eight patients with a novel disease phenotype comprising a hypokalemic tubulopathy with renal salt wasting, disturbed acid-base homeostasis, and sensorineural deafness. Electrophysiologic studies and surface expression experiments investigated the functional consequences of newly identified gene variants. RESULTS: We identified mutations in the KCNJ16 gene encoding KCNJ16, which along with KCNJ15 and KCNJ10, constitutes the major basolateral potassium channel of the proximal and distal tubules, respectively. Coexpression of mutant KCNJ16 together with KCNJ15 or KCNJ10 in Xenopus oocytes significantly reduced currents. CONCLUSIONS: Biallelic variants in KCNJ16 were identified in patients with a novel disease phenotype comprising a variable proximal and distal tubulopathy associated with deafness. Variants affect the function of heteromeric potassium channels, disturbing proximal tubular bicarbonate handling as well as distal tubular salt reabsorption.


Subject(s)
Acid-Base Imbalance/genetics , Hearing Loss, Sensorineural/genetics , Hypokalemia/genetics , Kidney Diseases/genetics , Potassium Channels, Inwardly Rectifying/genetics , Adolescent , Adult , Alleles , Animals , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Kidney Tubules , Loss of Function Mutation , Male , Mice , Nephrons/metabolism , Oocytes , Pedigree , Phenotype , RNA, Messenger/metabolism , Renal Reabsorption/genetics , Salts/metabolism , Exome Sequencing , Xenopus laevis , Young Adult
5.
Kidney Int ; 99(2): 324-335, 2021 02.
Article in English | MEDLINE | ID: mdl-33509356

ABSTRACT

Bartter syndrome is a rare inherited salt-losing renal tubular disorder characterized by secondary hyperaldosteronism with hypokalemic and hypochloremic metabolic alkalosis and low to normal blood pressure. The primary pathogenic mechanism is defective salt reabsorption predominantly in the thick ascending limb of the loop of Henle. There is significant variability in the clinical expression of the disease, which is genetically heterogenous with 5 different genes described to date. Despite considerable phenotypic overlap, correlations of specific clinical characteristics with the underlying molecular defects have been demonstrated, generating gene-specific phenotypes. As with many other rare disease conditions, there is a paucity of clinical studies that could guide diagnosis and therapeutic interventions. In this expert consensus document, the authors have summarized the currently available knowledge and propose clinical indicators to assess and improve quality of care.


Subject(s)
Alkalosis , Bartter Syndrome , Hypokalemia , Bartter Syndrome/diagnosis , Bartter Syndrome/genetics , Bartter Syndrome/therapy , Consensus , Humans , Rare Diseases
6.
Am J Hum Genet ; 103(5): 808-816, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30388404

ABSTRACT

Over the last decades, a growing spectrum of monogenic disorders of human magnesium homeostasis has been clinically characterized, and genetic studies in affected individuals have identified important molecular components of cellular and epithelial magnesium transport. Here, we describe three infants who are from non-consanguineous families and who presented with a disease phenotype consisting of generalized seizures in infancy, severe hypomagnesemia, and renal magnesium wasting. Seizures persisted despite magnesium supplementation and were associated with significant intellectual disability. Whole-exome sequencing and conventional Sanger sequencing identified heterozygous de novo mutations in the catalytic Na+, K+-ATPase α1 subunit (ATP1A1). Functional characterization of mutant Na+, K+-ATPase α1 subunits in heterologous expression systems revealed not only a loss of Na+, K+-ATPase function but also abnormal cation permeabilities, which led to membrane depolarization and possibly aggravated the effect of the loss of physiological pump activity. These findings underline the indispensable role of the α1 isoform of the Na+, K+-ATPase for renal-tubular magnesium handling and cellular ion homeostasis, as well as maintenance of physiologic neuronal activity.


Subject(s)
Intellectual Disability/genetics , Mutation/genetics , Renal Tubular Transport, Inborn Errors/genetics , Seizures/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Child , Child, Preschool , Female , Germ Cells , Heterozygote , Homeostasis/genetics , Humans , Infant , Infant, Newborn , Kidney/pathology , Magnesium/metabolism , Male , Phenotype , Protein Isoforms/genetics
7.
N Engl J Med ; 374(19): 1853-63, 2016 May 12.
Article in English | MEDLINE | ID: mdl-27120771

ABSTRACT

BACKGROUND: Three pregnancies with male offspring in one family were complicated by severe polyhydramnios and prematurity. One fetus died; the other two had transient massive salt-wasting and polyuria reminiscent of antenatal Bartter's syndrome. METHODS: To uncover the molecular cause of this possibly X-linked disease, we performed whole-exome sequencing of DNA from two members of the index family and targeted gene analysis of other members of this family and of six additional families with affected male fetuses. We also evaluated a series of women with idiopathic polyhydramnios who were pregnant with male fetuses. We performed immunohistochemical analysis, knockdown and overexpression experiments, and protein-protein interaction studies. RESULTS: We identified a mutation in MAGED2 in each of the 13 infants in our analysis who had transient antenatal Bartter's syndrome. MAGED2 encodes melanoma-associated antigen D2 (MAGE-D2) and maps to the X chromosome. We also identified two different MAGED2 mutations in two families with idiopathic polyhydramnios. Four patients died perinatally, and 11 survived. The initial presentation was more severe than in known types of antenatal Bartter's syndrome, as reflected by an earlier onset of polyhydramnios and labor. All symptoms disappeared spontaneously during follow-up in the infants who survived. We showed that MAGE-D2 affects the expression and function of the sodium chloride cotransporters NKCC2 and NCC (key components of salt reabsorption in the distal renal tubule), possibly through adenylate cyclase and cyclic AMP signaling and a cytoplasmic heat-shock protein. CONCLUSIONS: We found that MAGED2 mutations caused X-linked polyhydramnios with prematurity and a severe but transient form of antenatal Bartter's syndrome. MAGE-D2 is essential for fetal renal salt reabsorption, amniotic fluid homeostasis, and the maintenance of pregnancy. (Funded by the University of Groningen and others.).


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Antigens, Neoplasm/genetics , Bartter Syndrome/genetics , Genetic Diseases, X-Linked , Mutation , Polyhydramnios/genetics , Female , Fetal Death , Fetal Diseases/genetics , Fetus/metabolism , Humans , Kidney/metabolism , Male , Pedigree , Pregnancy , Premature Birth/genetics , Sequence Analysis, DNA , Sodium Chloride Symporters/metabolism , Solute Carrier Family 12, Member 1/metabolism
8.
J Inherit Metab Dis ; 42(5): 1019-1029, 2019 09.
Article in English | MEDLINE | ID: mdl-31177550

ABSTRACT

Cystinosis is an autosomal recessive storage disease due to impaired transport of cystine out of lysosomes. Since the accumulation of intracellular cystine affects all organs and tissues, the management of cystinosis requires a specialized multidisciplinary team consisting of pediatricians, nephrologists, nutritionists, ophthalmologists, endocrinologists, neurologists' geneticists, and orthopedic surgeons. Treatment with cysteamine can delay or prevent most clinical manifestations of cystinosis, except the renal Fanconi syndrome. Virtually all individuals with classical, nephropathic cystinosis suffer from cystinosis metabolic bone disease (CMBD), related to the renal Fanconi syndrome in infancy and progressive chronic kidney disease (CKD) later in life. Manifestations of CMBD include hypophosphatemic rickets in infancy, and renal osteodystrophy associated with CKD resulting in bone deformities, osteomalacia, osteoporosis, fractures, and short stature. Assessment of CMBD involves monitoring growth, leg deformities, blood levels of phosphate, electrolytes, bicarbonate, calcium, and alkaline phosphatase, periodically obtaining bone radiographs, determining levels of critical hormones and vitamins, such as thyroid hormone, parathyroid hormone, 25(OH) vitamin D, and testosterone in males, and surveillance for nonrenal complications of cystinosis such as myopathy. Treatment includes replacement of urinary losses, cystine depletion with oral cysteamine, vitamin D, hormone replacement, physical therapy, and corrective orthopedic surgery. The recommendations in this article came from an expert meeting on CMBD that took place in Salzburg, Austria, in December 2016.


Subject(s)
Bone Diseases/therapy , Cysteamine/therapeutic use , Cystinosis/drug therapy , Administration, Oral , Bone Diseases/etiology , Cysteamine/administration & dosage , Cystinosis/complications , Disease Management , Fanconi Syndrome/drug therapy , Female , Humans , Male
9.
Am J Hum Genet ; 97(6): 894-903, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26637979

ABSTRACT

SLC39A8 is a membrane transporter responsible for manganese uptake into the cell. Via whole-exome sequencing, we studied a child that presented with cranial asymmetry, severe infantile spasms with hypsarrhythmia, and dysproportionate dwarfism. Analysis of transferrin glycosylation revealed severe dysglycosylation corresponding to a type II congenital disorder of glycosylation (CDG) and the blood manganese levels were below the detection limit. The variants c.112G>C (p.Gly38Arg) and c.1019T>A (p.Ile340Asn) were identified in SLC39A8. A second individual with the variants c.97G>A (p.Val33Met) and c.1004G>C (p.Ser335Thr) on the paternal allele and c.610G>T (p.Gly204Cys) on the maternal allele was identified among a group of unresolved case subjects with CDG. These data demonstrate that variants in SLC39A8 impair the function of manganese-dependent enzymes, most notably ß-1,4-galactosyltransferase, a Golgi enzyme essential for biosynthesis of the carbohydrate part of glycoproteins. Impaired galactosylation leads to a severe disorder with deformed skull, severe seizures, short limbs, profound psychomotor retardation, and hearing loss. Oral galactose supplementation is a treatment option and results in complete normalization of glycosylation. SLC39A8 deficiency links a trace element deficiency with inherited glycosylation disorders.


Subject(s)
Cation Transport Proteins/genetics , Congenital Disorders of Glycosylation/genetics , Dwarfism/genetics , Manganese/blood , Spasms, Infantile/genetics , Amino Acid Sequence , Carbohydrate Sequence , Cation Transport Proteins/deficiency , Cations, Divalent , Congenital Disorders of Glycosylation/blood , Congenital Disorders of Glycosylation/complications , Congenital Disorders of Glycosylation/diet therapy , Dwarfism/blood , Dwarfism/complications , Dwarfism/diet therapy , Female , Galactose/therapeutic use , Gene Expression , High-Throughput Nucleotide Sequencing , Humans , Infant , Ion Transport , Manganese/deficiency , Molecular Sequence Data , Mutation , Pedigree , Sequence Alignment , Spasms, Infantile/blood , Spasms, Infantile/complications , Spasms, Infantile/diet therapy
10.
J Am Soc Nephrol ; 33(7): 1231-1233, 2022 07.
Article in English | MEDLINE | ID: mdl-35728888
12.
PLoS Genet ; 10(4): e1004267, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24699222

ABSTRACT

Intellectual disability and seizures are frequently associated with hypomagnesemia and have an important genetic component. However, to find the genetic origin of intellectual disability and seizures often remains challenging because of considerable genetic heterogeneity and clinical variability. In this study, we have identified new mutations in CNNM2 in five families suffering from mental retardation, seizures, and hypomagnesemia. For the first time, a recessive mode of inheritance of CNNM2 mutations was observed. Importantly, patients with recessive CNNM2 mutations suffer from brain malformations and severe intellectual disability. Additionally, three patients with moderate mental disability were shown to carry de novo heterozygous missense mutations in the CNNM2 gene. To elucidate the physiological role of CNNM2 and explain the pathomechanisms of disease, we studied CNNM2 function combining in vitro activity assays and the zebrafish knockdown model system. Using stable Mg(2+) isotopes, we demonstrated that CNNM2 increases cellular Mg2+ uptake in HEK293 cells and that this process occurs through regulation of the Mg(2+)-permeable cation channel TRPM7. In contrast, cells expressing mutated CNNM2 proteins did not show increased Mg(2+) uptake. Knockdown of cnnm2 isoforms in zebrafish resulted in disturbed brain development including neurodevelopmental impairments such as increased embryonic spontaneous contractions and weak touch-evoked escape behaviour, and reduced body Mg content, indicative of impaired renal Mg(2+) absorption. These phenotypes were rescued by injection of mammalian wild-type Cnnm2 cRNA, whereas mammalian mutant Cnnm2 cRNA did not improve the zebrafish knockdown phenotypes. We therefore concluded that CNNM2 is fundamental for brain development, neurological functioning and Mg(2+) homeostasis. By establishing the loss-of-function zebrafish model for CNNM2 genetic disease, we provide a unique system for testing therapeutic drugs targeting CNNM2 and for monitoring their effects on the brain and kidney phenotype.


Subject(s)
Brain/metabolism , Cyclins/genetics , Intellectual Disability/genetics , Magnesium/metabolism , Mutation, Missense/genetics , Seizures/genetics , Adolescent , Animals , Cation Transport Proteins , Cell Line , Female , HEK293 Cells , Humans , Infant , Infant, Newborn , Intellectual Disability/metabolism , Kidney/metabolism , Male , Phenotype , Seizures/metabolism , Zebrafish/genetics , Zebrafish/metabolism
13.
J Am Soc Nephrol ; 27(2): 604-14, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26047794

ABSTRACT

Idiopathic infantile hypercalcemia (IIH) is characterized by severe hypercalcemia with failure to thrive, vomiting, dehydration, and nephrocalcinosis. Recently, mutations in the vitamin D catabolizing enzyme 25-hydroxyvitamin D3-24-hydroxylase (CYP24A1) were described that lead to increased sensitivity to vitamin D due to accumulation of the active metabolite 1,25-(OH)2D3. In a subgroup of patients who presented in early infancy with renal phosphate wasting and symptomatic hypercalcemia, mutations in CYP24A1 were excluded. Four patients from families with parental consanguinity were subjected to homozygosity mapping that identified a second IIH gene locus on chromosome 5q35 with a maximum logarithm of odds (LOD) score of 6.79. The sequence analysis of the most promising candidate gene, SLC34A1 encoding renal sodium-phosphate cotransporter 2A (NaPi-IIa), revealed autosomal-recessive mutations in the four index cases and in 12 patients with sporadic IIH. Functional studies of mutant NaPi-IIa in Xenopus oocytes and opossum kidney (OK) cells demonstrated disturbed trafficking to the plasma membrane and loss of phosphate transport activity. Analysis of calcium and phosphate metabolism in Slc34a1-knockout mice highlighted the effect of phosphate depletion and fibroblast growth factor-23 suppression on the development of the IIH phenotype. The human and mice data together demonstrate that primary renal phosphate wasting caused by defective NaPi-IIa function induces inappropriate production of 1,25-(OH)2D3 with subsequent symptomatic hypercalcemia. Clinical and laboratory findings persist despite cessation of vitamin D prophylaxis but rapidly respond to phosphate supplementation. Therefore, early differentiation between SLC34A1 (NaPi-IIa) and CYP24A1 (24-hydroxylase) defects appears critical for targeted therapy in patients with IIH.


Subject(s)
Hypercalcemia/genetics , Infant, Newborn, Diseases/genetics , Metabolism, Inborn Errors/genetics , Mutation , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Sodium-Phosphate Cotransporter Proteins/genetics , Animals , Genes, Recessive , Humans , Infant , Infant, Newborn , Mice , Mice, Knockout
14.
N Engl J Med ; 365(5): 410-21, 2011 Aug 04.
Article in English | MEDLINE | ID: mdl-21675912

ABSTRACT

BACKGROUND: Vitamin D supplementation for the prevention of rickets is one of the oldest and most effective prophylactic measures in medicine, having virtually eradicated rickets in North America. Given the potentially toxic effects of vitamin D, the recommendations for the optimal dose are still debated, in part owing to the increased incidence of idiopathic infantile hypercalcemia in Britain in the 1950s during a period of high vitamin D supplementation in fortified milk products. We investigated the molecular basis of idiopathic infantile hypercalcemia, which is characterized by severe hypercalcemia, failure to thrive, vomiting, dehydration, and nephrocalcinosis. METHODS: We used a candidate-gene approach in a cohort of familial cases of typical idiopathic infantile hypercalcemia with suspected autosomal recessive inheritance. Identified mutations in the vitamin D-metabolizing enzyme CYP24A1 were evaluated with the use of a mammalian expression system. RESULTS: Sequence analysis of CYP24A1, which encodes 25-hydroxyvitamin D 24-hydroxylase, the key enzyme of 1,25-dihydroxyvitamin D(3) degradation, revealed recessive mutations in six affected children. In addition, CYP24A1 mutations were identified in a second cohort of infants in whom severe hypercalcemia had developed after bolus prophylaxis with vitamin D. Functional characterization revealed a complete loss of function in all CYP24A1 mutations. CONCLUSIONS: The presence of CYP24A1 mutations explains the increased sensitivity to vitamin D in patients with idiopathic infantile hypercalcemia and is a genetic risk factor for the development of symptomatic hypercalcemia that may be triggered by vitamin D prophylaxis in otherwise apparently healthy infants.


Subject(s)
Hypercalcemia/genetics , Mutation , Steroid Hydroxylases/genetics , Vitamin D/adverse effects , Animals , Cells, Cultured , Cricetinae , Cricetulus , DNA Mutational Analysis , Female , Genes, Recessive , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Hypercalcemia/chemically induced , Infant , Male , Pedigree , Risk Factors , Steroid Hydroxylases/metabolism , Vitamin D/metabolism , Vitamin D/therapeutic use , Vitamin D3 24-Hydroxylase
15.
Nat Genet ; 31(2): 166-70, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12032568

ABSTRACT

Magnesium is an essential ion involved in many biochemical and physiological processes. Homeostasis of magnesium levels is tightly regulated and depends on the balance between intestinal absorption and renal excretion. However, little is known about specific proteins mediating transepithelial magnesium transport. Using a positional candidate gene approach, we identified mutations in TRPM6 (also known as CHAK2), encoding TRPM6, in autosomal-recessive hypomagnesemia with secondary hypocalcemia (HSH, OMIM 602014), previously mapped to chromosome 9q22 (ref. 3). The TRPM6 protein is a new member of the long transient receptor potential channel (TRPM) family and is highly similar to TRPM7 (also known as TRP-PLIK), a bifunctional protein that combines calcium- and magnesium-permeable cation channel properties with protein kinase activity. TRPM6 is expressed in intestinal epithelia and kidney tubules. These findings indicate that TRPM6 is crucial for magnesium homeostasis and implicate a TRPM family member in human disease.


Subject(s)
Hypocalcemia/genetics , Ion Channels/genetics , Magnesium/blood , Mutation , Adult , Female , Haplotypes , Humans , Hypocalcemia/etiology , Infant , Infant, Newborn , Ion Channels/physiology , Male , Molecular Sequence Data , Multigene Family/genetics , Pedigree , Sequence Analysis, DNA , TRPM Cation Channels
16.
Genome Med ; 15(1): 62, 2023 08 23.
Article in English | MEDLINE | ID: mdl-37612755

ABSTRACT

BACKGROUND: Long-read sequencing is increasingly used to uncover structural variants in the human genome, both functionally neutral and deleterious. Structural variants occur more frequently in regions with a high homology or repetitive segments, and one rearrangement may predispose to additional events. Bartter syndrome type 3 (BS 3) is a monogenic tubulopathy caused by deleterious variants in the chloride channel gene CLCNKB, a high proportion of these being large gene deletions. Multiplex ligation-dependent probe amplification, the current diagnostic gold standard for this type of mutation, will indicate a simple homozygous gene deletion in biallelic deletion carriers. However, since the phenotypic spectrum of BS 3 is broad even among biallelic deletion carriers, we undertook a more detailed analysis of precise breakpoint regions and genomic structure. METHODS: Structural variants in 32 BS 3 patients from 29 families and one BS4b patient with CLCNKB deletions were investigated using long-read and synthetic long-read sequencing, as well as targeted long-read sequencing approaches. RESULTS: We report a ~3 kb duplication of 3'-UTR CLCNKB material transposed to the corresponding locus of the neighbouring CLCNKA gene, also found on ~50 % of alleles in healthy control individuals. This previously unknown common haplotype is significantly enriched in our cohort of patients with CLCNKB deletions (45 of 51 alleles with haplotype information, 2.2 kb and 3.0 kb transposition taken together, p=9.16×10-9). Breakpoint coordinates for the CLCNKB deletion were identifiable in 28 patients, with three being compound heterozygous. In total, eight different alleles were found, one of them a complex rearrangement with three breakpoint regions. Two patients had different CLCNKA/CLCNKB hybrid genes encoding a predicted CLCNKA/CLCNKB hybrid protein with likely residual function. CONCLUSIONS: The presence of multiple different deletion alleles in our cohort suggests that large CLCNKB gene deletions originated from many independently recurring genomic events clustered in a few hot spots. The uncovered associated sequence transposition haplotype apparently predisposes to these additional events. The spectrum of CLCNKB deletion alleles is broader than expected and likely still incomplete, but represents an obvious candidate for future genotype/phenotype association studies. We suggest a sensitive and cost-efficient approach, consisting of indirect sequence capture and long-read sequencing, to analyse disease-relevant structural variant hotspots in general.


Subject(s)
Bartter Syndrome , Humans , Haplotypes , Alleles , Genome, Human , Chloride Channels/genetics
17.
Nephrol Dial Transplant ; 27(2): 667-73, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21669885

ABSTRACT

BACKGROUND: Recent identification and characterization of novel renal Mg(2+) transporters and ion channels have greatly increased our understanding of the normal physiology of renal magnesium handling. METHODS: The present study deals with the clinical and molecular characterization of eight Turkish children (median age 10.6 years, range 3-16.2 years, five boys and three girls) with primary hypomagnesaemia from six families. RESULTS: All patients initially presented with tetany and convulsions. Laboratory evaluation yielded severely low serum magnesium levels and low serum calcium levels in all patients. While six patients exhibited inadequately low parathyroid hormone levels, the two remaining patients showed hyperparathyroidism, hypercalciuria and nephrocalcinosis. Genetic studies revealed familial hypomagnesaemia with secondary hypocalcaemia (HSH) due to a TRPM6 mutation in six patients and familial hypomagnesaemia with hypercalciuria and nephrocalcinosis (FHHNC) due to a CLDN16 mutation in one patient. CONCLUSIONS: Among recently identified magnesium-wasting disorders, HSH and FHHNC represent two major entities also in the Turkish population. Besides clinical course and laboratory diagnosis of hypomagnesaemia, the detection of renal calcium wasting and parathyroid function are crucial to differentiate between these most prevalent forms of hereditary magnesium deficiency. While TRPM6 mutations underlying HSH almost uniformly lead to a complete loss of function of the TRPM6 protein, the severity of FHHNC phenotype depends on the residual function of the mutated claudin-16 protein.


Subject(s)
Claudins/genetics , Genetic Predisposition to Disease , Hypercalciuria/epidemiology , Hypercalciuria/genetics , Nephrocalcinosis/epidemiology , Nephrocalcinosis/genetics , Renal Tubular Transport, Inborn Errors/epidemiology , Renal Tubular Transport, Inborn Errors/genetics , TRPM Cation Channels/genetics , Adolescent , Age Distribution , Child , Child, Preschool , Cohort Studies , DNA Mutational Analysis , Female , Humans , Hypercalciuria/diagnosis , Incidence , Infant , Infant, Newborn , Male , Mutation , Nephrocalcinosis/diagnosis , Pedigree , Phenotype , Renal Tubular Transport, Inborn Errors/diagnosis , Risk Assessment , Severity of Illness Index , Sex Distribution , Turkey/epidemiology
18.
Pediatr Nephrol ; 26(10): 1789-802, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21503667

ABSTRACT

Salt-losing tubulopathies with secondary hyperaldosteronism (SLT) comprise a set of well-defined inherited tubular disorders. Two segments along the distal nephron are primarily involved in the pathogenesis of SLTs: the thick ascending limb of Henle's loop, and the distal convoluted tubule (DCT). The functions of these pre- and postmacula densa segments are quite distinct, and this has a major impact on the clinical presentation of loop and DCT disorders - the Bartter- and Gitelman-like syndromes. Defects in the water-impermeable thick ascending limb, with its greater salt reabsorption capacity, lead to major salt and water losses similar to the effect of loop diuretics. In contrast, defects in the DCT, with its minor capacity of salt reabsorption and its crucial role in fine-tuning of urinary calcium and magnesium excretion, provoke more chronic solute imbalances similar to the effects of chronic treatment with thiazides. The most severe disorder is a combination of a loop and DCT disorder similar to the enhanced diuretic effect of a co-medication of loop diuretics with thiazides. Besides salt and water supplementation, prostaglandin E2-synthase inhibition is the most effective therapeutic option in polyuric loop disorders (e.g., pure furosemide and mixed furosemide-amiloride type), especially in preterm infants with severe volume depletion. In DCT disorders (e.g., pure thiazide and mixed thiazide-furosemide type), renin-angiotensin-aldosterone system (RAAS) blockers might be indicated after salt, potassium, and magnesium supplementation are deemed insufficient. It appears that in most patients with SLT, a combination of solute supplementation with some drug treatment (e.g., indomethacin) is needed for a lifetime.


Subject(s)
Bartter Syndrome/pathology , Gitelman Syndrome/pathology , Kidney Diseases/pathology , Kidney Tubules/pathology , Adult , Aldosterone/pharmacology , Aldosterone/physiology , Bartter Syndrome/diagnosis , Bartter Syndrome/therapy , Female , Gitelman Syndrome/diagnosis , Gitelman Syndrome/therapy , Humans , Hyperaldosteronism/complications , Hyperaldosteronism/pathology , Kidney Diseases/diagnosis , Kidney Diseases/therapy , Kidney Tubules, Distal/pathology , Loop of Henle/metabolism , Loop of Henle/pathology , Nephrons/pathology , Prognosis , Salts/metabolism
19.
J Pediatr Endocrinol Metab ; 33(10): 1353-1358, 2020 Aug 31.
Article in English | MEDLINE | ID: mdl-32866123

ABSTRACT

Objectives Both CYP24A1 and SLC34A1 gene mutations are responsible for idiopathic infantile hypercalcemia, whereas loss-of-function mutations in CYP24A1 (25-OH-vitamin D-24-hydroxylase) lead to a defect in the inactivation of active 1.25(OH)2D; mutations in SLC34A1 encoding renal sodium phosphate cotransporter NaPi-IIa lead to primary renal phosphate wasting combined with an inappropriate activation of vitamin D. The presence of mutations in both genes has not been reported in the same patient until today. Case presentation Hypercalcemia was incidentally detected when a 13-month-old boy was being examined for urinary tract infection. After 21 months, hypercalcemia was detected in his six-month-old sister. High dose of vitamin D was not given to both siblings. Both of them also had hypophosphatemia and decreased tubular phosphate reabsorption. Intensive hydration, furosemide and oral phosphorus treatment were given. Bilateral medullary nephrocalcinosis was detected in both siblings and their father. Serum Ca and P levels were within normal limits at follow-up in both siblings. Siblings and their parents all carry a homozygous stop codon mutation (p.R466*) in CYP24A1. Interestingly, both siblings and the father also have a heterozygous splice-site mutation (IVS6(+1)G>A) in SLC34A1. The father has nephrocalcinosis. Conclusions A biallelic loss-of-function mutation in the CYP24A1 gene was identified as responsible for hypercalcemia, hypercalciuria and nephrocalcinosis. In addition, a heterozygous mutation in the SLC34A1 gene, although not being the main pathogenic factor, might contribute to the severe phenotype of both patients.


Subject(s)
Hypercalcemia/genetics , Hypercalcemia/pathology , Mutation , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Vitamin D3 24-Hydroxylase/genetics , Adult , Child , Fathers , Female , Humans , Infant , Male , Siblings
SELECTION OF CITATIONS
SEARCH DETAIL