Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
1.
J Nephrol ; 36(2): 575-591, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-35994232

RESUMO

Renal tubules play an important role in maintaining water, electrolyte, and acid-base balance. Renal tubule dysfunction can cause electrolyte disorders and acid-base imbalance. Clinically, hypokalemic renal tubular disease is the most common tubule disorder. With the development of molecular genetics and gene sequencing technology, hereditary renal tubular diseases have attracted attention, and an increasing number of pathogenic genes related to renal tubular diseases have been discovered and reported. Inherited renal tubular diseases mainly occur due to mutations in genes encoding various specific transporters or ion channels expressed on the tubular epithelial membrane, leading to dysfunctional renal tubular reabsorption, secretion, and excretion. An in-depth understanding of the molecular genetic basis of hereditary renal tubular disease will help to understand the physiological function of renal tubules, the mechanism by which the kidney maintains water, electrolyte, and acid-base balance, and the relationship between the kidney and other systems in the body. Meanwhile, understanding these diseases also improves our understanding of the pathogenesis of hypokalemia, alkalosis and other related diseases and ultimately promotes accurate diagnostics and effective disease treatment. The present review summarizes the most common hereditary renal tubular diseases (Bartter syndrome, Gitelman syndrome, EAST syndrome and Liddle syndrome) characterized by hypokalemia and alkalosis. Further detailed explanations are provided for pathogenic genes and functional proteins, clinical manifestations, intrinsic relationship between genotype and clinical phenotype, diagnostic clues, differential diagnosis, and treatment strategies for these diseases.


Assuntos
Alcalose , Síndrome de Bartter , Hipopotassemia , Nefropatias , Humanos , Hipopotassemia/etiologia , Hipopotassemia/genética , Síndrome de Bartter/diagnóstico , Síndrome de Bartter/genética , Síndrome de Bartter/terapia , Nefropatias/complicações , Alcalose/diagnóstico , Alcalose/genética , Alcalose/terapia , Água
2.
Curr Opin Nephrol Hypertens ; 31(5): 508-515, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35894287

RESUMO

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.


Assuntos
Alcalose , Síndrome de Bartter , Síndrome de Gitelman , Hipopotassemia , Alcalose/complicações , Alcalose/genética , Síndrome de Bartter/genética , Síndrome de Gitelman/complicações , Síndrome de Gitelman/genética , Humanos , Hipopotassemia/genética , Magnésio/metabolismo , Sódio/metabolismo , Membro 3 da Família 12 de Carreador de Soluto/genética
3.
BMC Nephrol ; 23(1): 170, 2022 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-35509038

RESUMO

BACKGROUND: Gitelman Syndrome (GS) is a hereditary tubulopathy associated with a biallelic inactivating mutations of the SLC12A3 gene encoding the thiazide-sensitive sodium-chloride cotransporter (NCCT). The typical clinical manifestation is a hypokalemic metabolic alkalosis with significant hypomagnesemia, and low urinary calcium excretion. Hypocalciuria is widely believed to be a hallmark of GS that distinguishes it from Barter's syndrome, presenting as hypercalciuria. The pathomechanism of hypocalciuria in GS is not fully elucidated. Up to date, a clinical course of GS with normocalciuria has been reported only in men, while women have a milder course of the disease with typical hypocalciuria, which is believed as the result of sex hormone. Additionally, there is a growing evidence that calcium channels of the distal nephron could be regulated by a variety of hormones, including aldosterone (Aldo). CASE PRESENTATION: We present the case of a 28-year-old Caucasian woman with asymptomatic, chronic hypokalemia, hypomagnesemia, hypochloremic alkalosis and normal urinary calcium excretion. A high renin levels with normal concentration of Aldo in serum have also been found. The values of blood pressure were low. Based on genetic studies, two heterozygous mutations in the trans position were confirmed: c.2186G>T (p.Gly729Val) and c.1247G>C (p.Cys416Ser) in the SLC12A3 gene, which ultimately confirmed the diagnosis of GS. CONCLUSIONS: We report here the first case of genetically confirmed GS manifested as normocalciuria in a Caucasian woman. Thus, our result does not confirm a role of sex hormones on the level of calciuria. Based on the results of normal Aldo concentration despite high renin level in our patient, we hypothesized that Aldo may be connecting with the level of urinary calcium excretion in patients with the GS.


Assuntos
Alcalose , Síndrome de Gitelman , Adulto , Alcalose/genética , Cálcio/metabolismo , Feminino , Síndrome de Gitelman/complicações , Síndrome de Gitelman/diagnóstico , Síndrome de Gitelman/genética , Humanos , Magnésio , Masculino , Mutação/genética , Renina/genética , Membro 3 da Família 12 de Carreador de Soluto/genética
4.
Artigo em Inglês | MEDLINE | ID: mdl-33121425

RESUMO

CASE PRESENTATION: A two-year-old boy visited the doctor for hypokalemia and metabolic alkalosis. Laboratory examination revealed that urinary potassium excretion and serum aldosterone level were increased, with hyperthyroidism and thyroid-related antibodies positive at the same time. Genetic testing showed that there was a complex heterozygous mutation in the SLC12A3 gene, c.1077C>G (p.N359K) and c.1567G>A (p.A523?); the final diagnosis was Gitelman syndrome and autoimmune hyperthyroidism. BACKGROUND: Gitelman syndrome is an autosomal recessive genetic disease caused by the inactivation of mutation of the SLC12A3 gene. The onset age is more than 6 years old; it is mainly manifested as low blood potassium, low blood sodium, low blood chlorine, metabolic alkalosis, increased urine potassium and urine chlorine excretion, and low urine calcium. Autoimmune hyperthyroidism manifests due to autoimmune disorders. The highest incidence rate in children is of Graves' disease, followed by chronic lymphocytic thyroiditis. CONCLUSION: Several cases of Gitelman syndrome with autoimmune hyperthyroidism have been reported, most of which were Asian adults, and the case we identified is the first reported case in children under 14 years with both Gitelman syndrome and autoimmune hyperthyroidism. At the same time, we carried out a high-precision clinical exosome analysis of the gene of this case and further explored the relationship between Gitelman syndrome and autoimmune hyperthyroidism from the perspective of the gene.This case suggests that even children under 6 years with hyperthyroidism and hypokalemia should be suspected of Gitelman syndrome to avoid misdiagnosis.


Assuntos
Síndrome de Gitelman/complicações , Síndrome de Gitelman/genética , Doença de Graves/complicações , Alcalose/complicações , Alcalose/diagnóstico , Alcalose/genética , Pré-Escolar , China , Análise Mutacional de DNA , Testes Genéticos , Síndrome de Gitelman/diagnóstico , Doença de Graves/diagnóstico , Doença de Graves/genética , Humanos , Hipopotassemia/complicações , Hipopotassemia/diagnóstico , Hipopotassemia/genética , Masculino , Polimorfismo de Nucleotídeo Único , Membro 3 da Família 12 de Carreador de Soluto/genética
5.
BMC Nephrol ; 21(1): 328, 2020 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-32758178

RESUMO

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.


Assuntos
Síndrome de Gitelman/genética , Adulto , Idoso , Alcalose/genética , Alcalose/metabolismo , Síndrome de Bartter/metabolismo , China , Feminino , Genótipo , Síndrome de Gitelman/metabolismo , Humanos , Hipercalciúria/genética , Hipercalciúria/metabolismo , Hipopotassemia/genética , Hipopotassemia/metabolismo , Magnésio/sangue , Masculino , Pessoa de Meia-Idade , Mutação , Linhagem , Fenótipo , Eliminação Renal , Membro 3 da Família 12 de Carreador de Soluto/genética , Desequilíbrio Hidroeletrolítico/genética , Desequilíbrio Hidroeletrolítico/metabolismo
8.
Eur J Med Genet ; 62(10): 103728, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31325522

RESUMO

Persistent hypokalemic hypochloremic metabolic alkalosis represents a heterogeneous group of genetic disorders of which the most common is Bartter syndrome (BS). BS is an inherited renal tubulopathy caused by defective salt reabsorption in the thick ascending loop of Henle, which results in persistent hypokalemic hypochloremic metabolic alkalosis. Here we report a 10-year-old girl of a consanguineous family. She presented prenatally with severe polyhydramnios and distended bowel loops. Thereafter, she displayed failure to thrive and had recurrent admissions due to dehydration episodes associated with diarrhea, and characterized by hypokalemia, hypochloremia and metabolic alkalosis. BS was considered her working diagnosis for several years despite negative genetic analysis of the known genes associated with BS. Whole exome sequencing identified a novel homozygous c.1652delT deleterious frameshift mutation in the SLC26A3 gene, which confirmed the diagnosis of congenital chloride diarrhea (CCD), a rare autosomal recessive disease that mimics biochemically BS. A review of twelve additional reported cases of CCD that were initially misdiagnosed as BS, emphasizes CCD in the differential diagnosis of BS, and highlights the clinical discrepancies between these two entities. Taken together, our report further emphasizes the typical clinical features of CCD, and the importance of next generation sequencing in the diagnosis of syndromes with genetic heterogeneity. We suggest including SLC26A3 in the extended BS targeted gene panels.


Assuntos
Alcalose/diagnóstico , Alcalose/genética , Sequenciamento de Nucleotídeos em Larga Escala , Hipopotassemia/sangue , Hipopotassemia/diagnóstico , Alcalose/sangue , Criança , Diagnóstico Diferencial , Feminino , Estudos de Associação Genética , Marcadores Genéticos , Predisposição Genética para Doença , Testes Genéticos , Humanos
9.
Artigo em Inglês | MEDLINE | ID: mdl-31071454

RESUMO

Given that the chemistry of lactate production disproves the existence of a lactic acidosis, there is a need to further reveal and explain the importance of the organic and computational chemistry of pH dependent competitive cation fractional (~) proton (H+) exchange (~H+e). An additional importance of this knowledge is that it could potentially contradict the assumption of the Stewart approach to the physico-chemical theory of acid-base balance. For example, Stewart proposed that chemical reaction and pH dependent H+ dissociation and association do not directly influence the pH of cellular and systemic body fluids. Yet at the time of Stewart's work, there were no data that quantified the H+ exchange during chemical reactions, or from pH dependent metabolite H+ association or dissociation. Consequently, the purpose of this review and commentary was three-fold; 1) to provide explanation of pH dependent competitive cation ~H+e exchange; 2) develop a model of and calculate new data of substrate flux in skeletal muscle during intense exercise; and 3) then combine substrate flux data with the now known ~H+e from chemical reactions of non-mitochondrial energy catabolism to quantify chemical reaction and metabolic pathway ~H+e. The results of purpose 3 were that ~H+ release for the totality of cytosolic energy catabolism = -187.2 mmol·L-1, where total glycolytic ~H+te = -85.0 mmol·L-1. ATP hydrolysis had a ~H+te = -43.1 mmol·L-1. Lactate production provided the largest metabolic ~H+ buffering with a ~H+te = 44.5 mmol·L-1. The total ~H+ release to La ratio = 4.25. The review content and research results of this manuscript should direct science towards new approaches to understanding the cause and source of H+e during metabolic acidosis and alkalosis.


Assuntos
Acidose/genética , Alcalose/genética , Líquidos Corporais/metabolismo , Prótons , Acidose/metabolismo , Alcalose/metabolismo , Bicarbonatos/metabolismo , Glicólise/genética , Humanos , Concentração de Íons de Hidrogênio , Ácido Láctico/metabolismo , Músculo Esquelético/química , Músculo Esquelético/metabolismo
10.
Am J Physiol Renal Physiol ; 315(5): F1271-F1282, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30110571

RESUMO

To better understand the role of the inward-rectifying K channel Kir4.1 (KCNJ10) in the distal nephron, we initially studied a global Kir4.1 knockout mouse (gKO), which demonstrated the hypokalemia and hypomagnesemia seen in SeSAME/EAST syndrome and was associated with reduced Na/Cl cotransporter (NCC) expression. Lethality by ~3 wk, however, limits the usefulness of this model, so we developed a kidney-specific Kir4.1 "knockdown" mouse (ksKD) using a cadherin 16 promoter and Cre-loxP methodology. These mice appeared normal and survived to adulthood. Kir4.1 protein expression was decreased ~50% vs. wild-type (WT) mice by immunoblotting, and immunofluorescence showed moderately reduced Kir4.1 staining in distal convoluted tubule that was minimal or absent in connecting tubule and cortical collecting duct. Under control conditions, the ksKD mice showed metabolic alkalosis and relative hypercalcemia but were normokalemic and mildly hypermagnesemic despite decreased NCC expression. In addition, the mice had a severe urinary concentrating defect associated with hypernatremia, enlarged kidneys with tubulocystic dilations, and reduced aquaporin-3 expression. On a K/Mg-free diet for 1 wk, however, ksKD mice showed marked hypokalemia (serum K: 1.5 ± 0.1 vs. 3.0 ± 0.1 mEq/l for WT), which was associated with renal K wasting (transtubular K gradient: 11.4 ± 0.8 vs. 1.6 ± 0.4 in WT). Phosphorylated-NCC expression increased in WT but not ksKD mice on the K/Mg-free diet, suggesting that loss of NCC adaptation underlies the hypokalemia. In conclusion, even modest reduction in Kir4.1 expression results in impaired K conservation, which appears to be mediated by reduced expression of activated NCC.


Assuntos
Néfrons/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/deficiência , Potássio na Dieta/sangue , Reabsorção Renal , Alcalose/sangue , Alcalose/genética , Alcalose/fisiopatologia , Animais , Aquaporina 3/metabolismo , Técnicas de Silenciamento de Genes , Genótipo , Hipercalcemia/sangue , Hipercalcemia/genética , Hipercalcemia/fisiopatologia , Hiperpotassemia/sangue , Hiperpotassemia/genética , Hiperpotassemia/fisiopatologia , Hipernatremia/sangue , Hipernatremia/genética , Hipernatremia/fisiopatologia , Capacidade de Concentração Renal , Camundongos Endogâmicos C57BL , Camundongos Knockout , Néfrons/fisiopatologia , Fenótipo , Fosforilação , Canais de Potássio Corretores do Fluxo de Internalização/genética , Membro 3 da Família 12 de Carreador de Soluto/metabolismo
11.
Kidney Int ; 93(4): 893-902, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29310825

RESUMO

Kir4.1 in the distal convoluted tubule plays a key role in sensing plasma potassium and in modulating the thiazide-sensitive sodium-chloride cotransporter (NCC). Here we tested whether dietary potassium intake modulates Kir4.1 and whether this is essential for mediating the effect of potassium diet on NCC. High potassium intake inhibited the basolateral 40 pS potassium channel (a Kir4.1/5.1 heterotetramer) in the distal convoluted tubule, decreased basolateral potassium conductance, and depolarized the distal convoluted tubule membrane in Kcnj10flox/flox mice, herein referred to as control mice. In contrast, low potassium intake activated Kir4.1, increased potassium currents, and hyperpolarized the distal convoluted tubule membrane. These effects of dietary potassium intake on the basolateral potassium conductance and membrane potential in the distal convoluted tubule were completely absent in inducible kidney-specific Kir4.1 knockout mice. Furthermore, high potassium intake decreased, whereas low potassium intake increased the abundance of NCC expression only in the control but not in kidney-specific Kir4.1 knockout mice. Renal clearance studies demonstrated that low potassium augmented, while high potassium diminished, hydrochlorothiazide-induced natriuresis in control mice. Disruption of Kir4.1 significantly increased basal urinary sodium excretion but it abolished the natriuretic effect of hydrochlorothiazide. Finally, hypokalemia and metabolic alkalosis in kidney-specific Kir4.1 knockout mice were exacerbated by potassium restriction and only partially corrected by a high-potassium diet. Thus, Kir4.1 plays an essential role in mediating the effect of dietary potassium intake on NCC activity and potassium homeostasis.


Assuntos
Túbulos Renais Distais/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Potássio na Dieta/metabolismo , Alcalose/genética , Alcalose/metabolismo , Alcalose/fisiopatologia , Animais , Modelos Animais de Doenças , Feminino , Homeostase , Hidroclorotiazida/farmacologia , Hipopotassemia/genética , Hipopotassemia/metabolismo , Hipopotassemia/fisiopatologia , Túbulos Renais Distais/efeitos dos fármacos , Túbulos Renais Distais/fisiopatologia , Masculino , Potenciais da Membrana , Camundongos Knockout , Natriurese , Canais de Potássio Corretores do Fluxo de Internalização/deficiência , Canais de Potássio Corretores do Fluxo de Internalização/genética , Eliminação Renal , Sódio/urina , Inibidores de Simportadores de Cloreto de Sódio/farmacologia , Membro 3 da Família 12 de Carreador de Soluto/genética , Membro 3 da Família 12 de Carreador de Soluto/metabolismo
12.
J Am Soc Nephrol ; 28(10): 3118-3128, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28674042

RESUMO

Mice lacking distal tubular expression of CLDN10, the gene encoding the tight junction protein Claudin-10, show enhanced paracellular magnesium and calcium permeability and reduced sodium permeability in the thick ascending limb (TAL), leading to a urine concentrating defect. However, the function of renal Claudin-10 in humans remains undetermined. We identified and characterized CLDN10 mutations in two patients with a hypokalemic-alkalotic salt-losing nephropathy. The first patient was diagnosed with Bartter syndrome (BS) >30 years ago. At re-evaluation, we observed hypocalciuria and hypercalcemia, suggesting Gitelman syndrome (GS). However, serum magnesium was in the upper normal to hypermagnesemic range, thiazide responsiveness was not blunted, and genetic analyses did not show mutations in genes associated with GS or BS. Whole-exome sequencing revealed compound heterozygous CLDN10 sequence variants [c.446C>G (p.Pro149Arg) and c.465-1G>A (p.Glu157_Tyr192del)]. The patient had reduced urinary concentrating ability, with a preserved aquaporin-2 response to desmopressin and an intact response to furosemide. These findings were not in line with any other known salt-losing nephropathy. Subsequently, we identified a second unrelated patient showing a similar phenotype, in whom we detected compound heterozygous CLDN10 sequence variants [c.446C>G (p.(Pro149Arg) and c.217G>A (p.Asp73Asn)]. Cell surface biotinylation and immunofluorescence experiments in cells expressing the encoded mutants showed that only one mutation caused significant differences in Claudin-10 membrane localization and tight junction strand formation, indicating that these alterations do not fully explain the phenotype. These data suggest that pathogenic CLDN10 mutations affect TAL paracellular ion transport and cause a novel tight junction disease characterized by a non-BS, non-GS autosomal recessive hypokalemic-alkalotic salt-losing phenotype.


Assuntos
Alcalose/genética , Claudinas/genética , Hipopotassemia/genética , Erros Inatos do Transporte Tubular Renal/genética , Adolescente , Feminino , Humanos , Masculino , Adulto Jovem
13.
BMJ Case Rep ; 20152015 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-25568271

RESUMO

SLC26A3, a chloride/bicarbonate transporter mainly expressed in the intestines, plays a pivotal role in chloride absorption. We present a 23-year-old woman with a history of congenital chloride diarrhoea (CCD) and renal transplant who was admitted for rehydration and treatment of acute kidney injury after she presented with an acute diarrhoeal episode. Laboratory investigations confirmed metabolic alkalosis and severe hypochloraemia, consistent with her underlying CCD. This contrasts with most other forms of diarrhoea, which are normally associated with metabolic acidosis. Genetic testing was offered and revealed a homozygous non-sense mutation in SLC26A3 (Gly-187-Stop). This loss-of-function mutation results in bicarbonate retention in the blood and chloride loss into the intestinal lumen. Symptomatic management with daily NaCl and KCl oral syrups was supplemented with omeprazole therapy. The loss of her own kidneys is most likely due to crystal-induced nephropathy secondary to chronic volume contraction and chloride depletion. This case summarises the pathophysiology and management of CCD.


Assuntos
Alcalose/genética , Antiportadores de Cloreto-Bicarbonato/genética , Cloretos/metabolismo , Diarreia/congênito , Nefropatias/genética , Erros Inatos do Metabolismo/tratamento farmacológico , Mutação , Omeprazol/uso terapêutico , Adulto , Alcalose/sangue , Alcalose/tratamento farmacológico , Alcalose/etiologia , Bicarbonatos/sangue , Cloretos/uso terapêutico , Diarreia/tratamento farmacológico , Diarreia/genética , Feminino , Humanos , Rim/metabolismo , Rim/cirurgia , Nefropatias/etiologia , Nefropatias/cirurgia , Transplante de Rim , Erros Inatos do Metabolismo/genética , Inibidores da Bomba de Prótons/uso terapêutico , Transportadores de Sulfato , Adulto Jovem
14.
Pediatr Int ; 55(3): 371-3, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23782368

RESUMO

Bartter syndrome (BS) is a group of genetic disorders characterized by hypokalemic metabolic alkalosis, hyponatremia and elevated renin and aldosterone plasma concentrations. BS type II is caused by mutations in the KCNJ1 gene and usually presents with transient hyperkalemia. We report here a novel KCNJ1 mutation in a male neonate, prematurely born after a pregnancy complicated by polyhydramnios. The infant presented with typical clinical and laboratory findings of BS type II, such as hyponatremia, hypochloremic metabolic alkalosis, severe weight loss, elevated renin and aldosterone levels and transient hyperkalemia in the early postnatal period, which were later normalized. Molecular analysis revealed a compound heterozygous mutation in the KCNJ1 gene, consisting of a novel K76E and an already described V315G mutation, both affecting functional domains of the channel protein. Typical manifestations of antenatal BS in combination with hyperkalemia should prompt the clinician to search for mutations in the KCNJ1 gene first.


Assuntos
Síndrome de Bartter/diagnóstico , Síndrome de Bartter/genética , Análise Mutacional de DNA , Canais de Potássio Corretores do Fluxo de Internalização/genética , Alcalose/sangue , Alcalose/diagnóstico , Alcalose/genética , Alelos , Síndrome de Bartter/sangue , Cromossomos Humanos Par 11/genética , Seguimentos , Grécia , Humanos , Recém-Nascido , Masculino , Fenótipo , Reação em Cadeia da Polimerase , Potássio/sangue
15.
Amino Acids ; 44(3): 903-10, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23090292

RESUMO

The aim of this study was to observe the intracellular heat shock protein 72 (HSP72) and heme oxygenase-1 (HSP32) response to prolonged interval cycling following the ingestion of carbohydrates (CHO) and sodium bicarbonate (NaHCO(3)). Six recreationally active males (mean ± SD; age 23.2 ± 2.9 years, height 179.5 ± 5.5 cm, body mass 76.5 ± 6.8 kg, and peak power output 315 ± 36 W) volunteered to complete a 90 min interval cycling exercise on four occasions. The trials were completed in a random and blinded manner following ingestion of either: placebo and an artificial sweetener (P-P), NaHCO(3) and sweetener (B-P), placebo and CHO (P-CHO), and NaHCO(3) and CHO (B-CHO). Both HSP72 and HSP32 were significantly increased in monocytes and lymphocytes from 45 min post-exercise (p ≤ 0.039), with strong relationships between both cell types (HSP72, r = 0.83; HSP32, r = 0.89). Exogenous CHO had no influence on either HSP72 or HSP32, but the ingestion of NaHCO(3) significantly attenuated HSP32 in monocytes and lymphocytes (p ≤ 0.042). In conclusion, the intracellular stress protein response to 90 min interval exercise is closely related in monocytes and lymphocytes, and HSP32 appears to be attenuated with a pre-exercise alkalosis.


Assuntos
Alcalose/metabolismo , Metabolismo dos Carboidratos , Exercício Físico/fisiologia , Proteínas de Choque Térmico HSP72/metabolismo , Heme Oxigenase-1/metabolismo , Adulto , Alcalose/genética , Ingestão de Alimentos , Proteínas de Choque Térmico HSP72/genética , Heme Oxigenase-1/genética , Humanos , Linfócitos/metabolismo , Masculino , Monócitos/metabolismo , Bicarbonato de Sódio/metabolismo , Estresse Fisiológico , Adulto Jovem
16.
World J Pediatr ; 8(1): 25-30, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22282380

RESUMO

BACKGROUND: We aim to review the clinical features of two renal tubular disorders characterized by sodium and potassium wasting: Bartter syndrome and Gitelman syndrome. DATA SOURCES: Selected key references concerning these syndromes were analyzed, together with a PubMed search of the literature from 2000 to 2011. RESULTS: The clinical features common to both conditions and those which are distinct to each syndrome were presented. The new findings on the genetics of the five types of Bartter syndrome and the discrete mutations in Gitelman syndrome were reviewed, together with the diagnostic workup and treatment for each condition. CONCLUSIONS: Patients with Bartter syndrome types 1, 2 and 4 present at a younger age than classic Bartter syndrome type 3. They present with symptoms, often quite severe in the neonatal period. Patients with classic Bartter syndrome type 3 present later in life and may be sporadically asymptomatic or mildly symptomatic. The severe, steady-state hypokalemia in Bartter syndrome and Gitelman syndrome may abruptly become life-threatening under certain aggravating conditions. Clinicians need to be cognizant of such renal tubular disorders, and promptly treat at-risk patients.


Assuntos
Síndrome de Bartter/diagnóstico , Síndrome de Bartter/genética , Síndrome de Gitelman/diagnóstico , Síndrome de Gitelman/genética , Hipopotassemia/genética , Alcalose/genética , Síndrome de Bartter/classificação , Síndrome de Bartter/metabolismo , Síndrome de Bartter/terapia , Constipação Intestinal/genética , Diagnóstico Diferencial , Tontura/genética , Fadiga/genética , Síndrome de Gitelman/metabolismo , Síndrome de Gitelman/terapia , Humanos , Cãibra Muscular/genética , Mutação , Potássio/metabolismo , Sódio/metabolismo
17.
Ann Biol Clin (Paris) ; 69(4): 459-64, 2011.
Artigo em Francês | MEDLINE | ID: mdl-21896412

RESUMO

We report the case of an asymptomatic patient presenting a severe chronic renal hypokalaemia. Once being sure of no diuretics use, two hypothesis can be mentioned for a normotensive patient presenting an hypokalaemia associated with a metabolic alcalosis: Bartter syndrome or Gitelman syndrome. The highlighting of low magnesaemia and hypocalciuria strongly concentrates the diagnosis on Gitelman syndrome. First, this has been strengthened by the results of renal function tests and later it has confirmed by molecular diagnosis with the identification of a known homozygous mutation on SLC12A3 gene. In the patient family, the same chromosomal abnormality has been found in the young sister. For these two patients the treatment ordered is an antikaliuretic diuretic, magnesium and potassium supplements. This case shows the difficulty to diagnose Gitelman syndrome: it is frequently mistaken for Bartter syndrome. The main differences between these two syndromes are magnesaemia and calciuria. Furthemore , patients with Gitelman syndrome are often asymptomatic, this explains why prevalence of this illness is probably underestimated.


Assuntos
Síndrome de Bartter/diagnóstico , Síndrome de Gitelman/diagnóstico , Hipopotassemia/genética , Receptores de Droga/genética , Simportadores/genética , Adulto , Alcalose/genética , Doença Crônica , Diagnóstico Diferencial , Diuréticos/administração & dosagem , Feminino , Síndrome de Gitelman/tratamento farmacológico , Síndrome de Gitelman/genética , Humanos , Magnésio/administração & dosagem , Mutação , Potássio/administração & dosagem , Irmãos , Membro 3 da Família 12 de Carreador de Soluto , Espironolactona/administração & dosagem , Resultado do Tratamento
18.
Eur J Endocrinol ; 165(1): 167-70, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21551164

RESUMO

INTRODUCTION: Pendred syndrome, a combination of sensorineural deafness, impaired organification of iodide in the thyroid and goitre, results from biallelic defects in pendrin (encoded by SLC26A4), which transports chloride and iodide in the inner ear and thyroid respectively. Recently, pendrin has also been identified in the kidneys, where it is found in the apical plasma membrane of non-α-type intercalated cells of the cortical collecting duct. Here, it functions as a chloride-bicarbonate exchanger, capable of secreting bicarbonate into the urine. Despite this function, patients with Pendred syndrome have not been reported to develop any significant acid-base disturbances, except a single previous reported case of metabolic alkalosis in the context of Pendred syndrome in a child started on a diuretic. CASE REPORT: We describe a 46-year-old female with sensorineural deafness and hypothyroidism, who presented with severe hypokalaemic metabolic alkalosis during inter-current illnesses on two occasions, and who was found to be homozygous for a loss-of-function mutation (V138F) in SLC26A4. Her acid-base status and electrolytes were unremarkable when she was well. CONCLUSION: This case illustrates that, although pendrin is not usually required to maintain acid-base homeostasis under ambient condition, loss of renal bicarbonate excretion by pendrin during a metabolic alkalotic challenge may contribute to life-threatening acid-base disturbances in patients with Pendred syndrome.


Assuntos
Alcalose/etiologia , Proteínas de Membrana Transportadoras/genética , Alcoolismo/complicações , Alcalose/genética , Feminino , Bócio Nodular/complicações , Bócio Nodular/genética , Perda Auditiva Neurossensorial/complicações , Perda Auditiva Neurossensorial/genética , Humanos , Hipotireoidismo/genética , Proteínas de Membrana Transportadoras/fisiologia , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Transportadores de Sulfato
19.
J Am Soc Nephrol ; 21(12): 2117-29, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21088294

RESUMO

SeSAME/EAST syndrome is a channelopathy consisting of a hypokalemic, hypomagnesemic, metabolic alkalosis associated with seizures, sensorineural deafness, ataxia, and developmental abnormalities. This disease links to autosomal recessive mutations in KCNJ10, which encodes the Kir4.1 potassium channel, but the functional consequences of these mutations are not well understood. In Xenopus oocytes, all of the disease-associated mutant channels (R65P, R65P/R199X, G77R, C140R, T164I, and A167V/R297C) had decreased K(+) current (0 to 23% of wild-type levels). Immunofluorescence demonstrated decreased surface expression of G77R, C140R, and A167V expressed in HEK293 cells. When we coexpressed mutant and wild-type subunits to mimic the heterozygous state, R199X, C140R, and G77R currents decreased to 55, 40, and 20% of wild-type levels, respectively, suggesting that carriers of these mutations may present with an abnormal phenotype. Because Kir4.1 subunits can form heteromeric channels with Kir5.1, we coexpressed the aforementioned mutants with Kir5.1 and found that currents were reduced at least as much as observed when we expressed mutants alone. Reduction of pH(i) from approximately 7.4 to 6.8 significantly decreased currents of all mutants except R199X but did not affect wild-type channels. In conclusion, perturbed pH gating may underlie the loss of channel function for the disease-associated mutant Kir4.1 channels and may have important physiologic consequences.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Perda Auditiva Neurossensorial/genética , Canal de Potássio Kv1.1/genética , Mutação , Convulsões/genética , Alcalose/genética , Alcalose/fisiopatologia , Análise de Variância , Animais , Ataxia/genética , Ataxia/fisiopatologia , Imunofluorescência , Predisposição Genética para Doença , Células HEK293/metabolismo , Perda Auditiva Neurossensorial/fisiopatologia , Humanos , Hipopotassemia/genética , Hipopotassemia/fisiopatologia , Immunoblotting , Deficiência Intelectual/genética , Deficiência Intelectual/fisiopatologia , Modelos Animais , Biologia Molecular , Oócitos , Convulsões/fisiopatologia , Síndrome , Xenopus laevis
20.
QJM ; 103(10): 741-8, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20650971

RESUMO

Giltelman syndrome (GS) is a recessive salt-losing tubulopathy of children or young adults caused by a mutation of genes encoding the human sodium chloride cotransporters and magnesium channels in the thiazide-sensitive segments of the distal convoluted tubule. The plasma biochemical picture is characterized by hypokalemia, hypomagnesemia, hypocalciuria, metabolic alkalosis and hypereninemic hyperaldosteronism. However, patients with GS present some clinical and biochemical alterations resembling that observed in thiazide diuretics abuse. On the pathophysiological point of view, GS represents a useful and interesting human model to better understand the clinical consequences of plasma hydro-electrolytes and acid-base derangements, associated with multiple hormonal alterations. The impact of this complex disorder involves cardiovascular, muscle-skeletal and some other physiological functions, adversely affecting the patient's quality of life. This review tries to summarize and better explain the linkage between the electrolytes, neurohormonal derangements and clinical picture. Moreover, the differential diagnosis between other similar electrolyte-induced clinical disorders and GS is also discussed.


Assuntos
Alcalose/genética , Síndrome de Gitelman/complicações , Síndrome de Gitelman/diagnóstico , Hipopotassemia/genética , Mutação , Simportadores de Cloreto de Sódio/genética , Adulto , Animais , Cálcio/metabolismo , Criança , Diagnóstico Diferencial , Síndrome de Gitelman/genética , Humanos , Magnésio/metabolismo , Camundongos , Cloreto de Potássio/uso terapêutico , Prognóstico , Erros Inatos do Transporte Tubular Renal/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...