Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
Immunity ; 57(9): 2077-2094.e12, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-38906145

RESUMEN

Tissues are exposed to diverse inflammatory challenges that shape future inflammatory responses. While cellular metabolism regulates immune function, how metabolism programs and stabilizes immune states within tissues and tunes susceptibility to inflammation is poorly understood. Here, we describe an innate immune metabolic switch that programs long-term intestinal tolerance. Intestinal interleukin-18 (IL-18) stimulation elicited tolerogenic macrophages by preventing their proinflammatory glycolytic polarization via metabolic reprogramming to fatty acid oxidation (FAO). FAO reprogramming was triggered by IL-18 activation of SLC12A3 (NCC), leading to sodium influx, release of mitochondrial DNA, and activation of stimulator of interferon genes (STING). FAO was maintained in macrophages by a bistable switch that encoded memory of IL-18 stimulation and by intercellular positive feedback that sustained the production of macrophage-derived 2'3'-cyclic GMP-AMP (cGAMP) and epithelial-derived IL-18. Thus, a tissue-reinforced metabolic switch encodes durable immune tolerance in the gut and may enable reconstructing compromised immune tolerance in chronic inflammation.


Asunto(s)
Tolerancia Inmunológica , Interleucina-18 , Macrófagos , Nucleótidos Cíclicos , Interleucina-18/metabolismo , Interleucina-18/inmunología , Animales , Ratones , Nucleótidos Cíclicos/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Humanos , Ratones Endogámicos C57BL , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Ratones Noqueados , Ácidos Grasos/metabolismo , Intestinos/inmunología , Inmunidad Innata , Inflamación/inmunología , Inflamación/metabolismo , Glucólisis , Oxidación-Reducción
2.
Physiol Rev ; 104(3): 1147-1204, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38329422

RESUMEN

The Na+-Cl- cotransporter (NCC; SLC12A3) is a highly regulated integral membrane protein that is known to exist as three splice variants in primates. Its primary role in the kidney is to mediate the cosymport of Na+ and Cl- across the apical membrane of the distal convoluted tubule. Through this role and the involvement of other ion transport systems, NCC allows the systemic circulation to reclaim a fraction of the ultrafiltered Na+, K+, Cl-, and Mg+ loads in exchange for Ca2+ and [Formula: see text]. The physiological relevance of the Na+-Cl- cotransport mechanism in humans is illustrated by several abnormalities that result from NCC inactivation through the administration of thiazides or in the setting of hereditary disorders. The purpose of the present review is to discuss the molecular mechanisms and overall roles of Na+-Cl- cotransport as the main topics of interest. On reading the narrative proposed, one will realize that the knowledge gained in regard to these themes will continue to progress unrelentingly no matter how refined it has now become.


Asunto(s)
Homeostasis , Humanos , Animales , Homeostasis/fisiología , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Equilibrio Hidroelectrolítico/fisiología , Sodio/metabolismo , Riñón/metabolismo
3.
Clin Exp Nephrol ; 28(8): 803-810, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38478191

RESUMEN

BACKGROUND: This study aimed to analyze genotype-phenotype correlations in children with Gitelman syndrome (GS). METHODS: This multicenter retrospective study included 50 Korean children diagnosed with SLC12A3 variants in one or both alleles and the typical laboratory findings of GS. Genetic testing was performed using the Sanger sequencing except for one patient. RESULTS: The median age at the diagnosis was 10.5 years (interquartile range, 6.8;14.1), and 41 patients were followed up for a median duration of 5.4 years (interquartile range, 4.1;9.6). A total of 30 different SLC12A3 variants were identified. Of the patients, 34 (68%) had biallelic variants, and 16 (32%) had monoallelic variants on examination. Among the patients with biallelic variants, those (n = 12) with the truncating variants in one or both alleles had lower serum chloride levels (92.2 ± 3.2 vs. 96.5 ± 3.8 mMol/L, P = 0.002) at onset, as well as lower serum potassium levels (3.0 ± 0.4 vs. 3.4 ± 0.3 mMol/L, P = 0.016), and lower serum chloride levels (96.1 ± 1.9 vs. 98.3 ± 3.0 mMol/L, P = 0.049) during follow-up than those without truncating variants (n = 22). Patients with monoallelic variants on examination showed similar phenotypes and treatment responsiveness to those with biallelic variants. CONCLUSIONS: Patients with GS who had truncating variants in one or both alleles had more severe electrolyte abnormalities than those without truncating variants. Patients with GS who had monoallelic SLC12A3 variants on examination had almost the same phenotypes, response to treatment, and long-term prognosis as those with biallelic variants.


Asunto(s)
Estudios de Asociación Genética , Síndrome de Gitelman , Miembro 3 de la Familia de Transportadores de Soluto 12 , Humanos , Síndrome de Gitelman/genética , Síndrome de Gitelman/diagnóstico , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Niño , Masculino , Estudios Retrospectivos , Femenino , Adolescente , Fenotipo , República de Corea , Preescolar , Mutación , Potasio/sangre , Predisposición Genética a la Enfermedad , Cloruros/sangre
4.
Physiol Genomics ; 55(3): 113-131, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36645671

RESUMEN

Solute carrier 12 (Slc12) is a family of electroneutral cation-coupled chloride (Cl-) cotransporters. Na+/K+/2Cl- (Nkcc) and Na+/Cl- cotransporters (Ncc) belong to the Nkcc/Ncc subfamily. Human and mouse possess one gene for the Na+/Cl- cotransporter (ncc gene: slc12a3), whereas teleost fishes possess multiple ncc genes, slc12a3 (ncc1) and slc12a10 (ncc2), in addition to their species-specific paralogs. Amphibians and squamates have two ncc genes: slc12a3 (ncc1) and ncc3. However, the evolutionary relationship between slc12a10 and ncc3 remains unresolved, and the presence of slc12a10 (ncc2) in mammals has not been clarified. Synteny and phylogenetic analyses of vertebrate genome databases showed that ncc3 is the ortholog of slc12a10, and slc12a10 is present in most ray-finned fishes, coelacanths, amphibians, reptiles, and a few mammals (e.g., platypus and horse) but pseudogenized or deleted in birds, most mammals, and some ray-finned fishes (pufferfishes). This shows that slc12a10 is widely present among bony vertebrates and pseudogenized or deleted independently in multiple lineages. Notably, as compared with some fish that show varied slc12a10 tissue expression profile, spotted gar, African clawed frog, red-eared slider turtle, and horse express slc12a10 in the ovaries or premature gonads. In horse tissues, an unexpectedly large number of splicing variants for Slc12a10 have been cloned, many of which encode truncated forms of Slc12a10, suggesting that the functional constraints of horse slc12a10 are weakened, which may be in the process of becoming a pseudogene. Our results elaborate on the evolution of Nkcc/Ncc subfamily of Slc12 in vertebrates.NEW & NOTEWORTHY slc12a10 is not a fish-specific gene and is present in a few mammals (e.g., platypus and horse), non-avian reptiles, amphibians, but was pseudogenized or deleted in most mammals (e.g., human, mouse, cat, cow, and rhinoceros), birds, and some ray-finned fishes (pufferfishes).


Asunto(s)
Ornitorrinco , Femenino , Bovinos , Animales , Humanos , Caballos , Ratones , Miembro 3 de la Familia de Transportadores de Soluto 12 , Filogenia , Peces/genética , Reptiles/genética , Aves , Anfibios/genética
5.
Clin Genet ; 104(6): 674-678, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37702302

RESUMEN

Gitelman syndrome (GS) is caused by SLC12A3 biallelic variants. A previous study showed that large rearrangements (LRGs) of SLC12A3 accounted for the low sensitivity of genetic testing. However, a systematic screening for LRGs in Chinese GS patients is lacking. Massively parallel sequencing (MPS) and multiplex ligation-dependent probe amplification (MLPA) were performed to sequence the genomic DNA of patients with clinically diagnosed GS. Of 165 index cases, MPS identified 151 cases with two or more affected alleles and 14 cases with one variant allele. LRGs were detected by MLPA in 20 out of 27 cases, including 15 cases with suspected LRGs by MPS. Among these 20 cases with LRGs, the results of MPS and MLPA were identical in only 8 cases. Additional LRGs in 6 cases were detected by MLPA alone. In 6 cases, E4_E6del was identified by MPS, while E4_E5del and Intron6del were identified by MLPA. Among the 102 distinct variants, 30 are novel. LRGs were found in 20 cases (12.1%). LRGs were found in 12.1% of our Chinese GS patients cohort. We show that MPS and MLPA are two complementary techniques with the ability to improve the diagnostic yield of GS.


Asunto(s)
Pueblos del Este de Asia , Síndrome de Gitelman , Humanos , Pueblos del Este de Asia/genética , Pruebas Genéticas , Síndrome de Gitelman/genética , Mutación , Miembro 3 de la Familia de Transportadores de Soluto 12/genética
6.
Clin Endocrinol (Oxf) ; 99(5): 474-480, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-36562655

RESUMEN

OBJECTIVE: Gitelman syndrome (GS) is an autosomal recessive tubulopathy resulting from inactivating mutations in the SLC12A3 gene that encodes the thiazide-sensitive sodium-chloride cotransporter (NCC). To date, more than 500 mutations have been identified in the SLC12A3 gene. In this study, we identified two new mutations in the SLC12A3 gene in two Chinese GS pedigrees. DESIGN, PATIENTS AND MEASUREMENTS: The clinical characteristics and laboratory examination of two suspected GS patients in our hospital were analyzed. In addition, two pedigrees including 11 members and 2 patients underwent SLC12A3 gene analysis. RESULTS: Both patients were middle-aged women with characteristics of hypokalemic metabolic alkalosis, hypomagnesemia, low level of urinary calcium and the elevated levels of renin-angiotensin-aldosterone system. So, they were clinically diagnosed as GS. Patient 2 also had type 2 diabetes and Graves' disease. Both patients were found to carry two mutations of SLC12A3 gene by Sanger direct sequencing, which were all compound heterozygous mutations. We identified three mutations in these two Chinese GS pedigrees, one of which was c.179C>T (Thr60Met). The novel c.2159G>T (p. Gly720Val) and c.2675T>C (p. Leu892Pro) mutations were strongly predicted to be pathogenic using four network programs-Polyphen-2, SIFT, Mutation Taster and LRT. CONCLUSIONS: We identified two novel SLC12A3 genetic variant [c.2159G>T (p.Gly720Val) and c.2675T>C (p.Leu892Pro)] in two Chinese GS pedigrees. The discovery of new mutations has enriched the spectrum of SLC12A3 genotypes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Síndrome de Gitelman , Enfermedad de Graves , Persona de Mediana Edad , Humanos , Femenino , Síndrome de Gitelman/genética , Síndrome de Gitelman/diagnóstico , Linaje , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Mutación
7.
BMC Nephrol ; 24(1): 123, 2023 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-37131142

RESUMEN

A 14-year-old male patient who suffered from limb numbness, fatigue, and hypokalemia was considered Graves' disease (GD) complicated with thyrotoxic periodic paralysis (TPP) at the first diagnosis. Although with the treatment of antithyroid drugs, he developed severe hypokalemia and rhabdomyolysis (RM). Further laboratory tests revealed hypomagnesemia, hypocalciuria, metabolic alkalosis, hyperrenin, and hyperaldosteronemia. Genetic testing revealed compound heterozygous mutations in the SLC12A3 gene (c.506-1G > A, c.1456G > A) encoding the thiazide-sensitive sodium-chloride cotransporter, which presented a definitive diagnosis of Gitelman syndrome (GS). Moreover, gene analysis revealed his mother diagnosed with subclinical hypothyroidism due to Hashimoto's thyroiditis carried the c.506-1G > A heterozygous mutation in the SLC12A3 gene and his father carried the c.1456G > A heterozygous mutation in the SLC12A3 gene. His younger sister who had hypokalemia and hypomagnesemia carried the same compound heterozygous mutations as the proband and was diagnosed with GS as well, but with a much milder clinical presentation and better treatment outcome. This case suggested the potential relationship between GS and GD, clinicians should strengthen the differential diagnosis to avoid missed diagnosis.


Asunto(s)
Síndrome de Gitelman , Enfermedad de Graves , Hipopotasemia , Rabdomiólisis , Masculino , Femenino , Humanos , Adolescente , Síndrome de Gitelman/complicaciones , Síndrome de Gitelman/diagnóstico , Síndrome de Gitelman/genética , Hipopotasemia/etiología , Hipopotasemia/complicaciones , Mutación , Enfermedad de Graves/complicaciones , Enfermedad de Graves/diagnóstico , Enfermedad de Graves/genética , Madres , Rabdomiólisis/complicaciones , Rabdomiólisis/diagnóstico , Miembro 3 de la Familia de Transportadores de Soluto 12/genética
8.
Balkan J Med Genet ; 26(1): 69-74, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37576796

RESUMEN

Gitelman syndrome (GS) is a rare renal tubulopathy with an autosomal recessive mode of inheritance, caused by biallelic pathogenic variants in the SLC12A3 gene. The clinical features may overlap with other disorders, such as Bartter syndrome type 3, HNF1B nephropathy or even mitochondrial disease, but can be distinguished by molecular genetic analysis. Here we report on two preschool brothers, who presented with a several months' history of episodes of carpopedal spasms and muscle aches. The biochemical analyses revealed hypokalemia and hypomagnesemia without metabolic alkalosis. A 24-h urine sample demonstrated hypocalciuria. The molecular analyses showed that both patients were heterozygous for 3 (likely) pathogenic variants in SLC12A3: c.1805_1806del; p. (Tyr602Cysfs*31), c.2660+1G>A and c.2944 A>T; p. (Ile982Phe). Analysis of the parents showed that the mother was heterozygous for the c.2944 A>T p.(Ile982Phe) variant, and the father carried the other 2 variants (c.1805_1806del and c.2660+1G>A). Herein we present two children in a family from N. Macedonia with clinical manifestations and electrolyte imbalances suggestive of GS. The results of the tubulopathy next generation sequencing (NGS) panel confirmed the diagnosis. The boys are treated with a high salt diet and oral potassium and magnesium supplements.

9.
BMC Nephrol ; 23(1): 267, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35883046

RESUMEN

BACKGROUND: Gitelman syndrome (GS) is a rare autosomal recessive inherited salt-losing tubulopathy (SLT). Here, we report, for the first time, a case of GS overlapping nephrotic syndrome (NS) related to PLA2R-associated membranous nephropathy (MN). CASE PRESENTATION: We described a male patient had a 4-year history of recurrent fatigue. Serum biochemistry revealed hypokalemia with renal potassium wasting, hypomagnesemia, metabolic alkalosis, hyperreninemia, hypocalciuria, as well as nephrotic-range proteinuria, hypoalbuminemia, and elevated serum anti-phospholipase A2 receptor (PLA2R) antibody. Gene sequencing identified compound heterozygous mutations in SLC12A3 [c.536T > A(p.V179D) and c.1456G > A(p.D486N)]. The unusual association of SLTs and nephrotic-range glomerular proteinuria prompted us to perform a renal biopsy. Renal biopsy showed idiopathic MN. Due to the potential to activate the sodium-chloride co-transporter (NCC) and cause hyperkalemia, tacrolimus was selected to treat NS. Following treatment with potassium chloride, magnesium oxide, low-dose glucocorticoid combined with tacrolimus, the fatigue significantly improved, and concurrently hypokalemia, hypomagnesemia were corrected and NS was remitted. CONCLUSIONS: Renal biopsy should be warranted for GS patients with moderate to nephrotic-range proteinuria. Tacrolimus was preferred to the management of GS patients with NS.


Asunto(s)
Síndrome de Gitelman , Glomerulonefritis Membranosa , Hipopotasemia , Fatiga , Síndrome de Gitelman/complicaciones , Síndrome de Gitelman/diagnóstico , Síndrome de Gitelman/genética , Glomerulonefritis Membranosa/complicaciones , Glomerulonefritis Membranosa/diagnóstico , Glomerulonefritis Membranosa/genética , Humanos , Hipopotasemia/complicaciones , Magnesio , Masculino , Potasio , Proteinuria/complicaciones , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Tacrolimus/uso terapéutico
10.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 47(3): 401-406, 2022 Mar 28.
Artículo en Inglés, Zh | MEDLINE | ID: mdl-35545335

RESUMEN

Two patients with Gitelman syndrome were admitted to the Department of Endocrinology, Third Xiangya Hospital of Central South University. The genomic DNA from the patients' peripheral blood was extracted and the whole-exome sequencing was performed to detect the possible mutations. The function of the mutation sites was analyzed by bioinformatics software. Through whole-exome sequencing and Sanger sequencing, we have found that 2 patients with Gitelman syndrome carried compound heterozygous mutations of SLC12A3 gene, which were c.486_490delTACGGinsA, p.R943W, p.D486N, and p.R928C. Among them, c.486_490delTACGGinsA insertion deletion mutation causes frame shift and protein truncation. The p.R943W, p.D486N, and p.R928C of SLC12A3 gene were predicted to be pathogenic mutations by SIFT, PolyPhen2, and Mutation Taster. These 4 mutations were all reported, but p.R943W was first reported in Chinese population. Gitelman syndrome is rare in clinic and the rate of missed diagnosis is high. Early genetic analysis in patients with Gitelman syndrome is helpful to determine the etiology and guide the treatment.


Asunto(s)
Síndrome de Gitelman , Pruebas Genéticas , Síndrome de Gitelman/diagnóstico , Síndrome de Gitelman/genética , Humanos , Mutación , Linaje , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Secuenciación del Exoma
11.
Clin Genet ; 99(4): 558-564, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33382082

RESUMEN

Based on the Chinese Children Genetic Kidney Disease Database (CCGKDD), we established a pediatric Gitelman syndrome (GS) cohort to explore the phenotype and genotype characteristics. Thirty-two patients with SLC12A3 gene variants were collected. Five cases (16%) were homozygous, 16 (50%) were compound heterozygous, 10 (31%) carried only a single variant, and the other one harbored two de novo variants beyond classification. p.(T60M) was found in eight patients. The average diagnosis age was 7.79 ± 3.54 years. A total of 31% of the patients were asymptomatic. Muscle weakness was the most common symptom, accounting for 50%. Earlier age of onset (4.06 ± 1.17 yr vs. 8.10 ± 3.46 yr vs. 8.61 ± 3.56 yr, p< 0.05) and lower urinary calcium-creatinine ratio (p = 0.024) were found in the homozygous group than those in the heterozygous and compound heterozygous group. Patients with p.(T60M) variant had an earlier age of onset (4.01 ± 2.83 yr vs. 6.92 ± 3.07 yr, p = 0.025) and lower urinary calcium-creatinine ratio (p = 0.056). Thus, more than 30% of GS children have no clinical symptoms. Homozygous variant and the p.(T60M) variant may be associated with earlier onset and lower urinary calcium excretion in Chinese pediatric GS.


Asunto(s)
Pueblo Asiatico/genética , Síndrome de Gitelman/genética , Adolescente , Edad de Inicio , Calcio/orina , Niño , Preescolar , Creatinina/orina , Enanismo/genética , Femenino , Estudios de Asociación Genética , Síndrome de Gitelman/etnología , Síndrome de Gitelman/orina , Humanos , Hipopotasemia/genética , Lactante , Masculino , Debilidad Muscular/genética , Miembro 3 de la Familia de Transportadores de Soluto 12/genética
12.
J Endocrinol Invest ; 44(3): 471-480, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32642858

RESUMEN

PURPOSE: Gitelman syndrome (GS) is an autosomal recessive renal tubular disease that arises as a consequence of mutations in the SLC12A3 gene, which codes for an Na-Cl cotransporter (NCC) in distal renal tubules. This study was designed to explore the mutations associated with GS in an effort to more fully understand the molecular mechanisms governing GS. METHODS: We analyzed SLC12A3 mutations in a pedigree including a 42-year-old male with GS as well as four related family members over three generations using Sanger and next generation sequencing approaches. We additionally explored the functional ramifications of identified mutations using both Xenopus oocytes and the HEK293T cell line. RESULTS: We found that the subject with GS exhibited characteristic symptoms including sporadic thirst, fatigue, excess urination, and substantial hypokalemia and hypocalciuria, although magnesium levels were normal. Other analyzed subjects in this pedigree had normal laboratory findings and did not exhibit clear signs of GS. Sequencing analyses revealed that the GS subject exhibited a homozygous missense mutation (c.2874C > G, p.N958K) in exon 24 of SLC12A3. Both parents of this GS subject, as well as his older brother and daughter all exhibited heterozygous mutations at this same site. Functional analyses in Xenopus oocytes indicated that this mutated SLC12A3 gene encodes a protein which fails to mediate normal sodium transport, and when this mutant gene was expressed in HEK293T cells, we observed significant increases in endoplasmic reticulum (ER)-stress pathway activation. CONCLUSION: The p.N958K mutation in exon 24 of SLC12A3 can trigger GS at least in part via enhancing ER stress responses.


Asunto(s)
Estrés del Retículo Endoplásmico , Síndrome de Gitelman/patología , Homocigoto , Mutación Missense , Adulto , Femenino , Síndrome de Gitelman/etiología , Síndrome de Gitelman/metabolismo , Humanos , Masculino , Linaje , Pronóstico , Miembro 3 de la Familia de Transportadores de Soluto 12/genética
13.
Scand J Clin Lab Invest ; 81(8): 629-633, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34657521

RESUMEN

Mutations in the SLC12A3 gene have been reported to cause Gitelman syndrome (GS). This study aimed to investigate the genetic mutations and clinical features of patients with GS. Four pedigrees (4 GS patients and 14 family members) were enrolled. The symptoms, laboratory results, management, and genotypes were analyzed. Genomic DNA was screened for gene variations using Sanger sequencing. DNA sequences were compared with reference sequences. The effects of the mutations were predicted using prediction tools (Mutation Taster, PolyPhen-2, SIFT, and PROVEAN). Genetic analysis revealed six genetic variants of SLC12A3, including three novel heterozygous mutations (c.2T > C, c.1609C > T, c.3055G > A) and three previously characterized mutations (c.1456G > A, c.2542G > A, c.1077C > G). These mutations were predicted to exert a damaging effect based on predictive in silico tools. GS patients had low blood pressure and low levels of serum K+, serum Mg2+, and 24-h urinary Ca2+ but high levels of 24-h urinary K+. These clinical manifestations and genotypes were consistent with the diagnostic criteria of GS. The study described the phenotypes and genotypes of 4 pedigrees involving GS patients, demonstrating the importance of SLC12A3 gene screening for GS.


Asunto(s)
Síndrome de Gitelman , Síndrome de Gitelman/diagnóstico , Síndrome de Gitelman/genética , Humanos , Mutación , Linaje , Fenotipo , Miembro 3 de la Familia de Transportadores de Soluto 12/genética
14.
BMC Nephrol ; 21(1): 328, 2020 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-32758178

RESUMEN

BACKGROUND: Gitelman syndrome is a rare salt-losing renal tubular disorder associated with mutation of SLC12A3 gene, which encodes the Na-Cl co-transporter (NCCT). Gitelman syndrome is characterized by hypokalemia, metabolic alkalosis, hypomagnesemia, hypocalciuria, and renin-angiotensin-aldosterone system (RAAS) activation. Different SLC12A3 variants may lead to phenotypic variability and severity. METHODS: In this study, we reported the clinical features and genetic analysis of a Chinese pedigree diagnosed with Gitelman syndrome. RESULTS: The proband exhibited hypokalaemia, hypomagnesemia, metabolic alkalosis, but hypercalciuria and kidney stone formation. The increased urinary calcium excretion made it confused to Bartter syndrome. The persistent renal potassium wasting resulted in renal tubular lesions, and might affect urinary calcium reabsorption and excretion. Genetic analysis revealed mutations of SLC12A3 gene with c.433C > T (p.Arg145Cys), c.1077C > G (p.Asn359Lys), and c.1666C > T (p.Pro556Ser). Potential alterations of structure and function of NCCT protein due to those genetic variations of SLC12A3 are predicted. Interestingly, one sibling of the proband carried the same mutant sites and exhibited similar clinical features with milder phenotypes of hypokalemia and hypomagnesemia, but hypocalciuria rather than hypercalciuria. Family members with at least one wild type copy of SLC12A3 had normal biochemistry. With administration of spironolactone, potassium chloride and magnesium supplement, the serum potassium and magnesium were maintained within normal ranges. CONCLUSIONS: In this study, we identified compound mutations of SLC12A3 associated with varieties of clinical features. Further efforts are needed to investigate the diversity in clinical manifestations of Gitelman syndrome and its correlation with specific SLC12A3 mutations.


Asunto(s)
Síndrome de Gitelman/genética , Adulto , Anciano , Alcalosis/genética , Alcalosis/metabolismo , Síndrome de Bartter/metabolismo , China , Femenino , Genotipo , Síndrome de Gitelman/metabolismo , Humanos , Hipercalciuria/genética , Hipercalciuria/metabolismo , Hipopotasemia/genética , Hipopotasemia/metabolismo , Magnesio/sangre , Masculino , Persona de Mediana Edad , Mutación , Linaje , Fenotipo , Eliminación Renal , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Desequilibrio Hidroelectrolítico/genética , Desequilibrio Hidroelectrolítico/metabolismo
15.
BMC Pediatr ; 20(1): 366, 2020 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-32758191

RESUMEN

BACKGROUND: Hypokalaemia is a common condition among paediatric patients, but severe hypokalaemia is rare and can be life-threatening if not treated properly. The causes of hypokalaemia are complex. Finding the root cause is the key. CASE PRESENTATION: This article reports on a 2-year-old boy with severe hypokalaemia who was diagnosed with pneumonia. The child's lab findings were low blood potassium minimum level of 1.7 mmol/L, hypomagnesemia, and metabolic alkalosis. However, he was without the common features of hypokalaemia, such as respiratory paralysis, severe arrhythmia, weakness and decreased blood pressure. After recovering from pneumonia, his potassium levels did not return to normal. This outcome was suspected to be due to chronic renal loss of potassium. After undergoing second-generation gene sequencing tests, it was discovered he carried the SLC12A3 gene mutation with an Asp486Asn mutation site, which he had inherited from his mother. The final diagnosis was made, confirming the child suffered from Gitelman syndrome. CONCLUSIONS: Genetic predisposition is an important cause of hypokalaemia in children. Children with unexplained persistent hypokalaemia should be examined for the possibility of Gitelman syndrome, which should be distinguished from Bartter syndrome. Genetic testing is the gold standard.


Asunto(s)
Síndrome de Bartter , Síndrome de Gitelman , Hipopotasemia , Síndrome de Bartter/complicaciones , Síndrome de Bartter/diagnóstico , Síndrome de Bartter/genética , Niño , Preescolar , Síndrome de Gitelman/complicaciones , Síndrome de Gitelman/diagnóstico , Síndrome de Gitelman/genética , Humanos , Hipopotasemia/diagnóstico , Hipopotasemia/etiología , Masculino , Mutación , Potasio , Miembro 3 de la Familia de Transportadores de Soluto 12/genética
16.
J Endocrinol Invest ; 42(6): 653-665, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30413979

RESUMEN

CONTEXT: Gitelman syndrome (GS) is clinically heterogeneous. The genotype and phenotype correlation has not been well established. Though the long-term prognosis is considered to be favorable, hypokalemia is difficult to cure. OBJECTIVE: To analyze the clinical and genetic characteristics and treatment of all members of 13 GS pedigrees. METHODS: Thirteen pedigrees (86 members, 17 GS patients) were enrolled. Symptoms and management, laboratory findings, and genotype-phenotype associations among all the members were analyzed. RESULTS: The average ages at onset and diagnosis were 27.6 ± 10.2 years and 37.9 ± 11.6 years, respectively. Males were an average of 10 years younger and exhibited more profound hypokalemia than females. Eighteen mutations were detected. Two novel mutations (p.W939X, p.G212S) were predicted to be pathogenic by bioinformatic analysis. GS patients exhibited the lowest blood pressure, serum K+, Mg2+, and 24-h urinary Ca2+ levels. Although blood pressure, serum K+ and Mg2+ levels were normal in heterozygous carriers, 24-h urinary Na+ excretion was significantly increased. During follow-up, only 41.2% of patients reached a normal serum K+ level. Over 80% of patients achieved a normal Mg2+ level. Patients were taking 2-3 medications at higher doses than usual prescription to stabilize their K+ levels. Six patients were taking spironolactone simultaneously, but no significant elevation in the serum K+ level was observed. CONCLUSION: The phenotypic variability of GS and therapeutic strategies deserve further research to improve GS diagnosis and prognosis. Even heterozygous carriers exhibited increased 24-h Na+ urine excretion, which may make them more susceptible to diuretic-induced hypokalemia.


Asunto(s)
Pueblo Asiatico/genética , Síndrome de Gitelman/genética , Hipopotasemia/etiología , Mutación , Adolescente , Adulto , Niño , Femenino , Estudios de Seguimiento , Estudios de Asociación Genética , Genotipo , Síndrome de Gitelman/complicaciones , Síndrome de Gitelman/patología , Heterocigoto , Humanos , Hipopotasemia/patología , Masculino , Linaje , Fenotipo , Pronóstico , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Adulto Joven
17.
BMC Nephrol ; 20(1): 393, 2019 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-31660880

RESUMEN

BACKGROUND: Diabetic nephropathy is a global common cause of chronic kidney disease and end-stage renal disease. A lot of research has been conducted in biomedical sciences, which has enhanced understanding of the pathophysiology of diabetic nephropathy and has expanded the potential available therapies. An increasing number of evidence suggests that genetic alterations play a major role in development and progression of diabetic nephropathy. This systematic review was focused on searching an association between Arg913Gln variation in SLC12A3 gene with diabetic nephropathy in individuals with Type 2 Diabetes and Gitelman Syndrome. METHODS: An extensive systematic review of the literature was completed using PubMed, EBSCO and Cochrane Library, from their inception to January 2018. The PRISMA guidelines were followed and the search strategy ensured that all possible studies were identified to compile the review. Inclusion criteria for this review were: 1) Studies that analyzed the SLC12A3 gene in individuals with Type 2 Diabetes and Gitelman Syndrome. 2) Use of at least one analysis investigating the association between the Arg913Gln variation of SLC12A3 gene with diabetic nephropathy. 3) Use of a case-control or follow-up design. 4) Investigation of type 2 diabetes mellitus in individuals with Gitelman's syndrome, with a history of diabetic nephropathy. RESULTS: The included studies comprised 2106 individuals with diabetic nephropathy. This review shows a significant genetic association in most studies in the Arg913Gln variation of SLC12A3 gene with the diabetic nephropathy, pointing out that the mutations of this gene could be a key predictor of end-stage renal disease. CONCLUSIONS: The results showed in this systematic review contribute to better understanding of the association between the Arg913Gln variation of SLC12A3 gene with the pathogenesis of diabetic nephropathy in individuals with T2DM and GS.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Nefropatías Diabéticas/genética , Síndrome de Gitelman/genética , Diabetes Mellitus Tipo 2/complicaciones , Variación Genética , Síndrome de Gitelman/complicaciones , Humanos , Fallo Renal Crónico/genética , Mutación , Simportadores del Cloruro de Sodio/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/genética
18.
BMC Pediatr ; 19(1): 114, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30999883

RESUMEN

BACKGROUND: Gitelman syndrome (GS) is an autosomal recessive disorder and mild variant of classic Bartter syndrome. The latter is caused by defects in the genes CLCNKB and/or CLCNKA (chloride voltage-gated channel Ka and Kb). Patients with GS usually have loss-of-function mutations in SLC12A3. No patient has been reported with compound heterozygous mutations in these genes. We report a girl with GS with a paternally inherited heterozygous mutation in SLC12A3, and maternally inherited heterozygous variants in both CLCNKB and CLCNKA. CASE PRESENTATION: In this report, we reported a female patient (8 y and 10 mo) who had growth retardation (111.8 cm, - 1.62 standard deviation height for age) and normal blood pressure, with persistent hypokalemia, hypomagnesemia, hypocalciuria, hypochloremic alkalosis, and elevated levels of plasma renin and aldosterone. Her younger brother, father, and paternal grandmother all had histories of mild low levels of plasma potassium (3.0-3.5 mmol/L), which were rectified by potassium-rich foods. The genomic DNA of the patient, younger brother, parents, and grandparents were screened for gene variations and pedigree analysis using trio whole exome sequencing (WES). The candidate variants were validated by Sanger sequencing. Protein-protein interaction analysis utilized the following databases: Biogrid, MINT, HPRD, STRING, IntAct, iRefIndex, and ppiTrim. The trio WES screening showed that the patient has paternally inherited SLC12A3 p.N359K, and maternally inherited CLCNKB p.L94I. The paternal grandmother and younger brother are both carriers of SLC12A3 p.N359K. According to the STRING database, SLC12A3 and CLCNKB proteins may interact or coexpress with proteins associated with GS. CONCLUSIONS: Based on clinical phenotypes, genetic evidence of the pedigree, and previous reported studies, this case of GS indicates a digenetic inheritance of SLC12A3 and CLCNKB that resulted in renal tubular dysfunction perhaps, due to a genetic double-hit mechanism. The putative pathogenicity of the CLCNKB p.L94I variant requires confirmation.


Asunto(s)
Canales de Cloruro/genética , ADN/genética , Predisposición Genética a la Enfermedad , Síndrome de Gitelman/genética , Mutación , Niño , China , Canales de Cloruro/metabolismo , Análisis Mutacional de ADN , Femenino , Síndrome de Gitelman/diagnóstico , Síndrome de Gitelman/metabolismo , Humanos , Linaje , Fenotipo , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo
19.
BMC Med Genet ; 19(1): 17, 2018 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-29378538

RESUMEN

BACKGROUND: Gitelman syndrome (GS) is an autosomal recessive disorder caused by genic mutations of SLC12A3 (Solute carrier family 12 member 3), which encodes the Na-Cl cotransporter (NCC), and presents with characteristic metabolic abnormalities, including hypokalemia, metabolic alkalosis, hypomagnesemia, and hypocalciuria. In this study, we report a case of a GS pedigree, including analysis of GS-associated gene mutations. METHODS: We performed next-generation sequencing analysis and Sanger sequencing to explore the SLC12A3 mutations in a GS pedigree that included a 35-year-old female patient with GS and five family members within three generations. Furthermore, we summarized their clinical manifestations and analyzed laboratory parameters related to GS. RESULTS: The female proband (the patient with GS) presented with intermittent fatigue and transient periods of tetany, along with significant hypokalemia, hypomagnesemia, and hypocalciuria. All other members of the pedigree had normal laboratory results without obvious GS-related symptoms. Genetic analysis of the SLC12A3 gene identified two novel missense mutations (c.1919A > G, p.N640S in exon 15; c.2522A > G, p.D841G in exon 21) in the patient with GS. Moreover, we demonstrated that her mother, younger maternal uncle, and cousin were carriers of one mutation (c.1919A > G), and her father was the carrier of the other (c.2522A > G). CONCLUSION: This is the first report of these two novel pathogenic variants of SLC12A3 and their contribution to GS. Further functional studies are particularly warranted to explore the underlying molecular mechanisms.


Asunto(s)
Síndrome de Gitelman/diagnóstico , Síndrome de Gitelman/genética , Adulto , Femenino , Pruebas Genéticas , Variación Genética , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Mutación Missense , Linaje , Miembro 3 de la Familia de Transportadores de Soluto 12/genética
20.
Mol Cell Biochem ; 437(1-2): 203-210, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28744814

RESUMEN

Whether the Arg913Gln variation (rs11643718, G/A) of SLC12A3 contributes to diabetic nephropathy (DN) remains controversial. We undertook a case-control study to evaluate the association of the SLC12A3-Arg913Gln variation with the risk of end-stage renal disease (ESRD) in Chinese type 2 diabetes mellitus (T2DM) patients undergoing hemodialysis, and analyzed the genotype-phenotype interaction. Unrelated Chinese T2DM patients (n = 372) with diabetic retinopathy were classified into the non-DN (control) group (n = 151; duration of T2DM >15 years, no signs of renal involvement) and the DN-ESRD group (n = 221; ESRD due to T2DM, receiving hemodialysis). Polymerase chain reaction-direct sequencing was used to genotype the SLC12A3-Arg913Gln variation for all participants. The frequency of the GA+AA genotype in the DN-ESRD group was significantly higher than that of the non-DN group (23.1 vs. 9.9%; adjusted OR 2.2 (95% CI 1.3-4.5), P = 0.019). In the non-DN group, GA+AA carriers had a significantly higher urinary albumin excretion rate (UAER) and diastolic blood pressure compared with GG carriers (both P < 0.05). The SLC12A3-Arg913Gln variation may be associated with increased blood pressure and UAER and, therefore, could be used to predict the development and progression of DN-ESRD in Chinese T2DM patients undergoing hemodialysis.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Nefropatías Diabéticas/genética , Fallo Renal Crónico/genética , Mutación Missense , Anciano , Sustitución de Aminoácidos , Pueblo Asiatico/genética , China , Diabetes Mellitus Tipo 2/terapia , Nefropatías Diabéticas/terapia , Genotipo , Humanos , Fallo Renal Crónico/terapia , Persona de Mediana Edad , Diálisis Renal , Miembro 3 de la Familia de Transportadores de Soluto 12/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda