Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
BMC Nephrol ; 21(1): 282, 2020 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-32677916

RESUMO

BACKGROUND: Renal hypouricemia (RHUC) is a genetic disorder caused by mutations in the SLC22A12 gene, which encodes the major uric acid (UA) transporter, URAT1. The clinical course of related, living donor-derived RHUC in patients undergoing kidney transplantation is poorly understood. Here, we report a case of kidney transplantation from a living relative who had an SLC22A12 mutation. After the transplantation, the recipient's fractional excretion of UA (FEUA) decreased, and chimeric tubular epithelium was observed. CASE PRESENTATION: A 40-year-old man underwent kidney transplantation. His sister was the kidney donor. Three weeks after the transplantation, he had low serum-UA, 148.7 µmol/L, and elevated FEUA, 20.8% (normal: < 10%). The patient's sister had low serum-UA (101.1 µmol/L) and high FEUA (15.8%) before transplant. Suspecting RHUC, we performed next-generation sequencing on a gene panel containing RHUC-associated genes. A heterozygous missense mutation in the SLC22A12 gene was detected in the donor, but not in the recipient. The recipient's serum-UA level increased from 148.7 µmol/L to 231.9 µmol/L 3 months after transplantation and was 226.0 µmol/L 1 year after transplantation. His FEUA decreased from 20.8 to 11.7% 3 months after transplantation and was 12.4% 1 year after transplantation. Fluorescence in situ hybridization of allograft biopsies performed 3 months and 1 year after transplantation showed the presence of Y chromosomes in the tubular epithelial cells, suggesting the recipient's elevated serum-UA levels were owing to a chimeric tubular epithelium. CONCLUSIONS: We reported on a kidney transplant recipient that developed RHUC owing to his donor possessing a heterozygous mutation in the SLC22A12 (URAT1) gene. Despite this mutation, the clinical course was not problematic. Thus, the presence of donor-recipient chimerism in the tubular epithelium might positively affect the clinical course, at least in the short-term.


Assuntos
Transplante de Rim , Túbulos Renais/metabolismo , Doadores Vivos , Transportadores de Ânions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/genética , Erros Inatos do Transporte Tubular Renal/genética , Ácido Úrico/metabolismo , Cálculos Urinários/genética , Adulto , Quimerismo , Feminino , Heterozigoto , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Mutação de Sentido Incorreto , Eliminação Renal/genética , Erros Inatos do Transporte Tubular Renal/metabolismo , Irmãos , Cálculos Urinários/metabolismo
2.
Nephrology (Carlton) ; 25(10): 749-757, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32542819

RESUMO

AIM: Gitelman syndrome (GS) is a rare inherited salt-losing renal tubulopathy. Data on clinical features and the pregnancy outcome for female GS patients in a large cohort are lacking. The study was aimed to explore the phenotype and pregnant issue for female GS patients. METHODS: GS cases from the National Rare Diseases Registry System of China (NRSC) were collected, and detailed clinical, laboratory and genetic data were analysed. Articles on pregnancy in GS were also systemically reviewed. RESULTS: A total of 101 GS patients were included; among them, 42.6% were female and 79.2% showed hypomagnesaemia. A lower proportion of female patients presented before 18 years of age, with less frequently reported polyuria, higher serum potassium and less urine sodium and chloride excretions. There was no gender difference in the sodium-chloride cotransporter (NCC) dysfunction evaluated by hydrochlorothiazide test. Twelve of the 43 female GS patients delivered after disease symptom onset, and their pregnancies were generally uneventful. As a group, pregnant GS patients had lower potassium levels in the first-trimester (P = .002) requiring higher potassium supplementation. After delivery, serum potassium (P = .02) and magnesium (P = .03) increased significantly. Both caesarean section and vaginal delivery were safe. CONCLUSION: Female GS patients may have a less severe phenotype with generally favourable outcomes of pregnancy. Intensive monitoring and increased potassium supplementation are necessary during pregnancy, especially in the first-trimester.


Assuntos
Parto Obstétrico , Síndrome de Gitelman , Potássio , Complicações na Gravidez , Membro 3 da Família 12 de Carreador de Soluto/genética , Desequilíbrio Hidroeletrolítico , Adulto , China/epidemiologia , Cloretos/urina , Parto Obstétrico/métodos , Parto Obstétrico/estatística & dados numéricos , Feminino , Síndrome de Gitelman/epidemiologia , Síndrome de Gitelman/genética , Síndrome de Gitelman/fisiopatologia , Síndrome de Gitelman/terapia , Humanos , Recém-Nascido , Magnésio/sangue , Masculino , Mutação , Poliúria/diagnóstico , Poliúria/etiologia , Potássio/sangue , Potássio/uso terapêutico , Gravidez , Complicações na Gravidez/epidemiologia , Complicações na Gravidez/etiologia , Complicações na Gravidez/fisiopatologia , Complicações na Gravidez/terapia , Resultado da Gravidez/epidemiologia , Eliminação Renal/genética , Sódio/urina , Membro 3 da Família 12 de Carreador de Soluto/metabolismo , Desequilíbrio Hidroeletrolítico/sangue , Desequilíbrio Hidroeletrolítico/etiologia , Desequilíbrio Hidroeletrolítico/terapia , Desequilíbrio Hidroeletrolítico/urina
3.
Kidney Int ; 95(5): 1197-1208, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30910378

RESUMO

Urine biomarkers reflecting kidney function and handling of dietary sodium and potassium are strongly associated with several common diseases including chronic kidney disease, cardiovascular disease, and diabetes mellitus. Knowledge about the genetic determinants of these biomarkers may shed light on pathophysiological mechanisms underlying the development of these diseases. We performed genome-wide association studies of urinary albumin: creatinine ratio (UACR), urinary potassium: creatinine ratio (UK/UCr), urinary sodium: creatinine ratio (UNa/UCr) and urinary sodium: potassium ratio (UNa/UK) in up to 218,450 (discovery) and 109,166 (replication) unrelated individuals of European ancestry from the UK Biobank. Further, we explored genetic correlations, tissue-specific gene expression, and possible genes implicated in the regulation of these biomarkers. After replication, we identified 19 genome-wide significant independent loci associated with UACR, 6 each with UK/UCr and UNa/UCr, and 4 with UNa/UK. In addition to 22 novel associations, we confirmed several established associations, including between the CUBN locus and microalbuminuria. We detected high pairwise genetic correlation across the urinary biomarkers, and between their levels and several physiological measurements. We highlight GIPR, a potential diabetes drug target, as possibly implicated in the genetic control of urinary potassium excretion, and NRBP1, a locus associated with gout, as plausibly involved in sodium and albumin excretion. Overall, we identified 22 novel genome-wide significant associations with urinary biomarkers and confirmed several previously established associations, providing new insights into the genetic basis of these traits and their connection to chronic diseases.


Assuntos
Loci Gênicos , Eliminação Renal/genética , Insuficiência Renal Crônica/genética , Biomarcadores/metabolismo , Biomarcadores/urina , Feminino , Estudo de Associação Genômica Ampla , Humanos , Masculino , Pessoa de Meia-Idade , Potássio/metabolismo , Potássio/urina , Receptores Citoplasmáticos e Nucleares/genética , Receptores dos Hormônios Gastrointestinais/genética , Insuficiência Renal Crônica/diagnóstico , Insuficiência Renal Crônica/urina , Albumina Sérica Humana/metabolismo , Albumina Sérica Humana/urina , Sódio/metabolismo , Sódio/urina , Proteínas de Transporte Vesicular/genética
4.
Urolithiasis ; 47(1): 35-42, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30673801

RESUMO

The proximal tubule is responsible for reclaiming water, phosphates and amino acids from the tubular filtrate. There are genetic defects in both phosphate and amino acid transporters leading to nephrolithiasis. This review also explores genetic defects in regulators of phosphate and calcium transport in this nephron segment that lead to stone formation.


Assuntos
Sistemas de Transporte de Aminoácidos/genética , Cálculos Renais/genética , Túbulos Renais Proximais/fisiopatologia , Proteínas de Transporte de Fosfato/genética , Eliminação Renal/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Cálcio/metabolismo , Humanos , Cálculos Renais/fisiopatologia , Túbulos Renais Proximais/patologia , Proteínas de Transporte de Fosfato/metabolismo , Fosfatos/metabolismo
5.
Pediatr Nephrol ; 34(3): 389-403, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-29549463

RESUMO

Nephrotic syndrome is one of the most common glomerular disorders in childhood. Glucocorticoids have been the cornerstone of the treatment of childhood nephrotic syndrome for several decades, as the majority of children achieves complete remission after prednisone or prednisolone treatment. Currently, treatment guidelines for the first manifestation and relapse of nephrotic syndrome are mostly standardized, while large inter-individual variation is present in the clinical course of disease and side effects of glucocorticoid treatment. This review describes the mechanisms of glucocorticoid action and clinical pharmacokinetics and pharmacodynamics of prednisone and prednisolone in nephrotic syndrome patients. However, these mechanisms do not account for the large inter-individual variability in the response to glucocorticoid treatment. Previous research has shown that genetic factors can have a major influence on the pharmacokinetic and dynamic profile of the individual patient. Therefore, pharmacogenetics may have a promising role in personalized medicine for patients with nephrotic syndrome. Currently, little is known about the impact of genetic polymorphisms on glucocorticoid response and steroid-related toxicities in children with nephrotic syndrome. Although the evidence is limited, the data summarized in this study do suggest a role for pharmacogenetics to improve individualization of glucocorticoid therapy. Therefore, studies in larger cohorts with nephrotic syndrome patients are necessary to draw final conclusions about the influence of genetic polymorphisms on the glucocorticoid response and steroid-related toxicities to ultimately implement pharmacogenetics in clinical practice.


Assuntos
Variação Biológica da População/genética , Resistência a Medicamentos/genética , Glucocorticoides/farmacologia , Síndrome Nefrótica/tratamento farmacológico , Medicina de Precisão/métodos , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Adulto , Fatores Etários , Criança , Glucocorticoides/uso terapêutico , Humanos , Síndrome Nefrótica/genética , Polimorfismo Genético , Guias de Prática Clínica como Assunto , Medicina de Precisão/normas , Prednisolona/farmacologia , Prednisolona/uso terapêutico , Prednisona/farmacologia , Prednisona/uso terapêutico , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Indução de Remissão/métodos , Eliminação Renal/genética , Distribuição Tecidual/genética , Resultado do Tratamento
6.
Urolithiasis ; 47(1): 5-10, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30460525

RESUMO

Urinary stones tend to cluster in families. Of the known risk factors, evidence is strongest for heritability of urinary calcium excretion. Recent studies suggest that other stone risk factors may have heritable components including urinary pH, citrate and magnesium excretion, and circulating vitamin D concentration. Several risk factors assumed purely environmental may also have heritable components, including dietary intake and thirst. Thus, future studies may reveal that genetics plays an even stronger role in urinary stone pathogenesis than previously known.


Assuntos
Predisposição Genética para Doença , Cálculos Renais/genética , Eliminação Renal/genética , Citratos/urina , Comportamento Alimentar , Humanos , Rim/metabolismo , Cálculos Renais/sangue , Cálculos Renais/metabolismo , Cálculos Renais/urina , Magnésio/urina , Fatores de Risco , Vitamina D/sangue
7.
Urolithiasis ; 47(1): 43-56, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30460527

RESUMO

Twin and genealogy studies suggest a strong genetic component of nephrolithiasis. Likewise, urinary traits associated with renal stone formation were found to be highly heritable, even after adjustment for demographic, anthropometric and dietary covariates. Recent high-throughput sequencing projects of phenotypically well-defined cohorts of stone formers and large genome-wide association studies led to the discovery of many new genes associated with kidney stones. The spectrum ranges from infrequent but highly penetrant variants (mutations) causing mendelian forms of nephrolithiasis (monogenic traits) to common but phenotypically mild variants associated with nephrolithiasis (polygenic traits). About two-thirds of the genes currently known to be associated with nephrolithiasis code for membrane proteins or enzymes involved in renal tubular transport. The thick ascending limb of Henle and connecting tubules are of paramount importance for renal water and electrolyte handling, urinary concentration and maintenance of acid-base homeostasis. In most instances, pathogenic variants in genes involved in thick ascending limb of Henle and connecting tubule function result in phenotypically severe disease, frequently accompanied by nephrocalcinosis with progressive CKD and to a variable degree by nephrolithiasis. The aim of this article is to review the current knowledge on kidney stone disease associated with inherited defects in the thick ascending loop of Henle and the connecting tubules. We also highlight recent advances in the field of kidney stone genetics that have implications beyond rare disease, offering new insights into the most common type of kidney stone disease, i.e., idiopathic calcium stone disease.


Assuntos
Canais Iônicos/genética , Alça do Néfron/metabolismo , Nefrolitíase/etiologia , Insuficiência Renal Crônica/genética , Erros Inatos do Transporte Tubular Renal/genética , Cálcio/metabolismo , Progressão da Doença , Estudo de Associação Genômica Ampla , Humanos , Canais Iônicos/metabolismo , Alça do Néfron/patologia , Mutação , Nefrocalcinose/etiologia , Nefrocalcinose/patologia , Nefrolitíase/patologia , Eliminação Renal/genética , Insuficiência Renal Crônica/patologia , Reabsorção Renal/genética , Erros Inatos do Transporte Tubular Renal/complicações , Erros Inatos do Transporte Tubular Renal/patologia
8.
Urolithiasis ; 47(1): 23-33, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30446806

RESUMO

Calcium-sensing receptor (CaSR) is a plasma-membrane G protein-coupled receptor activated by extracellular calcium and expressed in kidney tubular cells. It inhibits calcium reabsorption in the ascending limb and distal convoluted tubule when stimulated by the increase of serum calcium levels; therefore, these tubular segments are enabled by CaSR to play a substantial role in the regulation of serum calcium levels. In addition, CaSR increases water and proton excretion in the collecting duct and promotes phosphate reabsorption and citrate excretion in the proximal tubule. These CaSR activities form a network in which they are integrated to protect the kidney against the negative effects of high calcium concentrations and calcium precipitates in urine. Therefore, the CaSR gene has been considered as a candidate to explain calcium nephrolithiasis. Epidemiological studies observed that calcium nephrolithiasis was associated with polymorphisms of the CaSR gene regulatory region, rs6776158, located within the promoter-1, rs1501899 located in the intron 1, and rs7652589 in the 5'-untranslated region. These polymorphisms were found to reduce the transcriptional activity of promoter-1. Activating rs1042636 polymorphism located in exon 7 was associated with calcium nephrolithiasis and hypercalciuria. Genetic polymorphisms decreasing CaSR expression could predispose individuals to stones because they may impair CaSR protective effects against precipitation of calcium phosphate and oxalate. Activating polymorphisms rs1042636 could predispose to calcium stones by increasing calcium excretion. These findings suggest that CaSR may play a complex role in lithogenesis through different pathways having different relevance under different clinical conditions.


Assuntos
Cálcio/metabolismo , Predisposição Genética para Doença , Túbulos Renais/metabolismo , Nefrolitíase/genética , Receptores de Detecção de Cálcio/genética , Regiões 5' não Traduzidas/genética , Genótipo , Humanos , Íntrons/genética , Nefrolitíase/metabolismo , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas/genética , Receptores de Detecção de Cálcio/metabolismo , Eliminação Renal/genética , Reabsorção Renal/genética
9.
Urolithiasis ; 47(1): 57-66, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30515543

RESUMO

Cystinuria, a genetic disorder of cystine transport, is characterized by excessive excretion of cystine in the urine and recurrent cystine stones in the kidneys and, to a lesser extent, in the bladder. Males generally are more severely affected than females. The disorder may lead to chronic kidney disease in many patients. The cystine transporter (b0,+) is a heterodimer consisting of the rBAT (encoded by SLC3A1) and b0,+AT (encoded by SLC7A9) subunits joined by a disulfide bridge. The molecular basis of cystinuria is known in great detail, and this information is now being used to define genotype-phenotype correlations. Current treatments for cystinuria include increased fluid intake to increase cystine solubility and the administration of thiol drugs for more severe cases. These drugs, however, have poor patient compliance due to adverse effects. Thus, there is a need to reduce or eliminate the risks associated with therapy for cystinuria. Four mouse models for cystinuria have been described and these models provide a resource for evaluating the safety and efficacy of new therapies for cystinuria. We are evaluating a new approach for the treatment of cystine stones based on the inhibition of cystine crystal growth by cystine analogs. Our ongoing studies indicate that cystine diamides are effective in preventing cystine stone formation in the Slc3a1 knockout mouse model for cystinuria. In addition to crystal growth, crystal aggregation is required for stone formation. Male and female mice with cystinuria have comparable levels of crystalluria, but very few female mice form stones. The identification of factors that inhibit cystine crystal aggregation in female mice may provide insight into the gender difference in disease severity in patients with cystinuria.


Assuntos
Cistina/metabolismo , Cistinúria/genética , Modelos Animais de Doenças , Cálculos Renais/etiologia , Sistemas de Transporte de Aminoácidos Básicos/genética , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Animais , Quelantes , Ensaios Clínicos como Assunto , Cistina/análogos & derivados , Cistina/uso terapêutico , Cistinúria/complicações , Cistinúria/tratamento farmacológico , Cistinúria/epidemiologia , Desenvolvimento de Medicamentos , Feminino , Humanos , Rim/metabolismo , Cálculos Renais/prevenção & controle , Masculino , Camundongos , Camundongos Knockout , Prevalência , Eliminação Renal/genética , Índice de Gravidade de Doença , Fatores Sexuais
11.
Am J Physiol Renal Physiol ; 314(6): F1138-F1144, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29357420

RESUMO

Many physiological functions have a circadian rhythm, including blood pressure (BP). BP is highest during the active phase, whereas during the rest period, BP dips 10-20%. Patients that do not experience this dip at night are termed "nondippers." Nondipping hypertension is associated with increased risk of cardiovascular disease. The mechanisms underlying nondipping hypertension are not understood. Without the circadian clock gene Per1, C57BL/6J mice develop nondipping hypertension on a high-salt diet plus mineralocorticoid treatment (HS/DOCP). Our laboratory has shown that PER1 regulates expression of several genes related to sodium (Na) transport in the kidney, including epithelial Na channel (ENaC) and Na chloride cotransporter (NCC). Urinary Na excretion also demonstrates a circadian pattern with a peak during active periods. We hypothesized that PER1 contributes to circadian regulation of BP via a renal Na-handling-dependent mechanism. Na-handling genes from the distal nephron were inappropriately regulated in KO mice on HS/DOCP. Additionally, the night/day ratio of Na urinary excretion by Per1 KO mice is decreased compared with WT (4 × vs. 7×, P < 0.001, n = 6 per group). Distal nephron-specific Per1 KO mice also show an inappropriate increase in expression of Na transporter genes αENaC and NCC. These results support the hypothesis that PER1 mediates control of circadian BP rhythms via the regulation of distal nephron Na transport genes. These findings have implications for the understanding of the etiology of nondipping hypertension and the subsequent development of novel therapies for this dangerous pathophysiological condition.


Assuntos
Pressão Sanguínea , Ritmo Circadiano , Hipertensão/metabolismo , Túbulos Renais Distais/metabolismo , Natriurese , Proteínas Circadianas Period/metabolismo , Eliminação Renal , Animais , Pressão Sanguínea/genética , Ritmo Circadiano/genética , Acetato de Desoxicorticosterona , Modelos Animais de Doenças , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/metabolismo , Predisposição Genética para Doença , Hipertensão/genética , Hipertensão/fisiopatologia , Túbulos Renais Distais/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Natriurese/genética , Proteínas Circadianas Period/deficiência , Proteínas Circadianas Period/genética , Fenótipo , Eliminação Renal/genética , Cloreto de Sódio na Dieta , Membro 3 da Família 12 de Carreador de Soluto/genética , Membro 3 da Família 12 de Carreador de Soluto/metabolismo , Fatores de Tempo , Regulação para Cima
12.
Br J Clin Pharmacol ; 84(1): 130-141, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28850715

RESUMO

AIMS: BMS-823778 is an inhibitor of 11ß-hydroxysteroid dehydrogenase type-1, and thus a potential candidate for Type 2 diabetes treatment. Here, we investigated the metabolism and pharmacokinetics of BMS-823778 to understand its pharmacokinetic variations in early clinical trials. METHODS: The metabolism of BMS-823778 was characterized in multiple in vitro assays. Pharmacokinetics were evaluated in healthy volunteers, prescreened as CYP2C19 extensive metabolizers (EM) or poor metabolizers (PM), with a single oral dose of [14 C]BMS-823778 (10 mg, 80 µCi). RESULTS: Three metabolites (<5%) were identified in human hepatocytes and liver microsomes (HLM) incubations, including two hydroxylated metabolites (M1 and M2) and one glucuronide conjugate (M3). As the most abundant metabolite, M1 was formed mainly through CYP2C19. M1 formation was also correlated with CYP2C19 activities in genotyped HLM. In humans, urinary excretion of dosed radioactivity was significantly higher in EM (68.8%; 95% confidence interval 61.3%, 76.3%) than in PM (47.0%; 43.5%, 50.6%); only small portions (<2%) were present in faeces or bile from both genotypes. In plasma, BMS-823778 exposure in PM was significantly (5.3-fold, P = 0.0097) higher than in EM. Furthermore, total radioactivity exposure was significantly higher (P < 0.01) than BMS-823778 exposure in all groups, indicating the presence of metabolites. M1 was the only metabolite observed in plasma, and much lower in PM. In urine, the amount of M1 and its oxidative metabolite in EM was 7-fold of that in PM, while more glucuronide conjugates of BMS-823778 and M1 were excreted in PM. CONCLUSIONS: CYP2C19 polymorphisms significantly impacted systemic exposure and metabolism pathways of BMS-823778 in humans.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/antagonistas & inibidores , Citocromo P-450 CYP2C19/genética , Polimorfismo Genético , Piridinas/farmacocinética , Eliminação Renal/genética , Triazóis/farmacocinética , Adulto , Esquema de Medicação , Genótipo , Glucuronídeos/metabolismo , Voluntários Saudáveis , Hepatócitos/metabolismo , Humanos , Hidroxilação , Masculino , Microssomos Hepáticos/metabolismo , Oxirredução , Piridinas/administração & dosagem , Piridinas/sangue , Piridinas/metabolismo , Triazóis/administração & dosagem , Triazóis/sangue , Triazóis/metabolismo , Adulto Jovem
13.
Kidney Int ; 92(6): 1536-1543, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28888328

RESUMO

The handling of electrolytes by the kidney is essential for homeostasis. However, the heritability of these processes, the first step in gene discovery, is poorly known. To help clarify this, we estimated the heritability of serum concentration, urinary excretion, renal clearance, and fractional excretion of sodium, potassium, magnesium, calcium, phosphate, and chloride in a population-based study. Nuclear families were randomly selected from the general population in Lausanne, Geneva, and Bern, Switzerland, and urine collected over 24-hour periods. We used the ASSOC program (S.A.G.E.) to estimate narrow sense heritability, including sex, age, body mass index, and study center as covariates in the model. The 1128 participants, from 273 families, had a mean age of 47 years, body mass index of 25.0 kg/m2, and an estimated glomerular filtration rate (CKD-EPI) of 98 mL/min/1.73 m2. The heritability of serum concentration was highest for calcium, 37% and lowest for sodium, 13%. The heritability of 24-hour urine clearances, excretions, and fractional excretions ranged from 15%, 10%, and 16%, respectively, for potassium to 45%, 44%, and 51%, respectively, for calcium. All probability values were significant. The heritability for phosphate-related phenotypes was lower than that for calcium. Thus, the serum and urine concentrations as well as urinary excretion and renal handling of electrolytes are heritable in the general adult population. The phenotypic variance attributable to additive genetic factors was variable and was higher for calcium. These results pave the way for identifying genetic variants involved in electrolyte homeostasis in the general population.


Assuntos
Eletrólitos/metabolismo , Homeostase/genética , Rim/fisiopatologia , Eliminação Renal/genética , Adulto , Estudos de Coortes , Biologia Computacional , Eletrólitos/sangue , Eletrólitos/urina , Feminino , Taxa de Filtração Glomerular , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Fenótipo , Software , Suíça
14.
Pediatr Nephrol ; 32(7): 1123-1135, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27234911

RESUMO

Magnesium is essential to the proper functioning of numerous cellular processes. Magnesium ion (Mg2+) deficits, as reflected in hypomagnesemia, can cause neuromuscular irritability, seizures and cardiac arrhythmias. With normal Mg2+ intake, homeostasis is maintained primarily through the regulated reabsorption of Mg2+ by the thick ascending limb of Henle's loop and distal convoluted tubule of the kidney. Inadequate reabsorption results in renal Mg2+ wasting, as evidenced by an inappropriately high fractional Mg2+ excretion. Familial renal Mg2+ wasting is suggestive of a genetic cause, and subsequent studies in these hypomagnesemic families have revealed over a dozen genes directly or indirectly involved in Mg2+ transport. Those can be classified into four groups: hypercalciuric hypomagnesemias (encompassing mutations in CLDN16, CLDN19, CASR, CLCNKB), Gitelman-like hypomagnesemias (CLCNKB, SLC12A3, BSND, KCNJ10, FYXD2, HNF1B, PCBD1), mitochondrial hypomagnesemias (SARS2, MT-TI, Kearns-Sayre syndrome) and other hypomagnesemias (TRPM6, CNMM2, EGF, EGFR, KCNA1, FAM111A). Although identification of these genes has not yet changed treatment, which remains Mg2+ supplementation, it has contributed enormously to our understanding of Mg2+ transport and renal function. In this review, we discuss general mechanisms and symptoms of genetic causes of hypomagnesemia as well as the specific molecular mechanisms and clinical phenotypes associated with each syndrome.


Assuntos
Arritmias Cardíacas/sangue , Hipercalciúria/genética , Deficiência de Magnésio/genética , Magnésio/sangue , Nefrocalcinose/genética , Eliminação Renal/genética , Reabsorção Renal/genética , Erros Inatos do Transporte Tubular Renal/genética , Convulsões/sangue , Arritmias Cardíacas/etiologia , Criança , Bloqueadores do Canal de Sódio Epitelial/uso terapêutico , Homeostase/genética , Humanos , Hipercalciúria/sangue , Hipercalciúria/complicações , Hipercalciúria/tratamento farmacológico , Hipopotassemia/sangue , Hipopotassemia/tratamento farmacológico , Hipopotassemia/etiologia , Hipopotassemia/genética , Túbulos Renais Distais/fisiologia , Alça do Néfron/fisiologia , Magnésio/fisiologia , Magnésio/uso terapêutico , Deficiência de Magnésio/complicações , Deficiência de Magnésio/tratamento farmacológico , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Antagonistas de Receptores de Mineralocorticoides/uso terapêutico , Mitocôndrias/metabolismo , Mutação , Nefrocalcinose/sangue , Nefrocalcinose/complicações , Nefrocalcinose/tratamento farmacológico , Fenótipo , Recomendações Nutricionais , Reabsorção Renal/efeitos dos fármacos , Erros Inatos do Transporte Tubular Renal/sangue , Erros Inatos do Transporte Tubular Renal/complicações , Erros Inatos do Transporte Tubular Renal/tratamento farmacológico , Convulsões/etiologia
15.
Pediatr Nephrol ; 32(10): 1851-1859, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-27757584

RESUMO

BACKGROUND: Dent disease is a rare X-linked recessive proximal tubulopathy caused by mutations in CLCN5 (Dent-1) or OCRL (Dent-2). As a rule, total protein excretion (TPE) is low in tubular proteinuria compared with glomerular disease. Several authors have reported nephrotic-range proteinuria (NP) and glomerulosclerosis in Dent disease. Therefore, we aimed to analyze protein excretion in patients with documented CLCN5 or OCRL mutations in a systematic literature review. DESIGN: PubMed and Embase were searched for cases with documented CLCN5 or OCRL mutations and (semi-)quantitative data on protein excretion. The most reliable data (i.e., TPE > protein-creatinine ratio > Albustix) was used for NP classification. RESULTS: Data were available on 148 patients from 47 reports: 126 had a CLCN5 and 22 an OCRLmutation. TPE was not significantly different between both forms (p = 0.11). Fifty-five of 126 (43.7 %) Dent-1 vs 13/22 (59.1 %) Dent-2 patients met the definition of NP (p = 0.25). Serum albumin was normal in all reported cases (24/148). Glomerulosclerosis was noted in 20/32 kidney biopsies and was strongly related to tubulointerstitial fibrosis, but not to kidney function or proteinuria. CONCLUSION: More than half of the patients with both forms of Dent disease have NP, and the presence of low molecular weight proteinuria in a patient with NP in the absence of edema and hypoalbuminemia should prompt genetic testing. Even with normal renal function, glomerulosclerosis and tubulointerstitial fibrosis are present in Dent disease. The role of proteinuria in the course of the disease needs to be examined further in longitudinal studies.


Assuntos
Doença de Dent/urina , Nefrite Intersticial/urina , Proteinúria/genética , Eliminação Renal/genética , Biópsia , Canais de Cloreto/genética , Doença de Dent/sangue , Doença de Dent/diagnóstico , Doença de Dent/genética , Testes Genéticos , Humanos , Rim/patologia , Rim/fisiopatologia , Mutação , Nefrite Intersticial/sangue , Nefrite Intersticial/diagnóstico , Nefrite Intersticial/genética , Monoéster Fosfórico Hidrolases/genética , Proteinúria/sangue , Proteinúria/diagnóstico , Proteinúria/urina , Albumina Sérica/análise
16.
Clin Exp Hypertens ; 38(4): 352-8, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27149052

RESUMO

OBJECTIVE: We examined an interaction according to the estimated 24-h urinary sodium excretion on the association between ATP2B1 rs17249754 and incident hypertension. METHODS: We assessed the incidence of hypertension in 1780 participants aged 45 to 75 years without cardiovascular diseases. DNA genotyping was performed using the Affymetrix Single Nucleotide Polymorphism (SNP) array 5.0. Because of previously reported associations with hypertension in various populations including Koreans, ATP2B1 rs17249754 was determined. Sodium intake was assessed by estimating the 24-h urinary sodium excretion with a Kawasaki formula from a spot urine sample. We utilized Cox proportional hazard models to test an interaction according to urinary sodium excretion on the association between ATP2B1 rs17249754 and incident hypertension. RESULTS: The incident hypertension was increased as sodium excretion level was increased. We confirmed that the ATP2B1 rs17249754 polymorphism had significant association with hypertension. We found that the association was modified by urinary sodium excretion level (p-value = 0.006); the mutant type (AA allele, previously recognized as a protective allele) with <4 g/day urinary sodium excretion had an inverse association on hypertension compared with the wild types (GG+GA) (HR = 0.39, 95% confidence interval (CI) 0.17-0.90), whereas the mutant type with ≥6 g/day urinary sodium demonstrated a significantly increased risk of hypertension (HR = 1.89, 95% CI 1.05-3.43). CONCLUSIONS: In this 6-year longitudinal study, our findings suggest that excessive sodium intake estimated from urinary sodium excretion significantly modified the risk of developing hypertension associated with ATP2B1 rs17249754 genetic trait. That is, carriers with ATP2B1 rs17249754 homozygote mutant allele may be at higher risk of hypertension, when they consume excessive sodium intake.


Assuntos
Hipertensão , ATPases Transportadoras de Cálcio da Membrana Plasmática/genética , Eliminação Renal/genética , Sódio na Dieta/metabolismo , Sódio/urina , Idoso , Feminino , Genótipo , Humanos , Hipertensão/epidemiologia , Hipertensão/genética , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Incidência , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Modelos de Riscos Proporcionais , República da Coreia/epidemiologia
17.
Drug Metab Pharmacokinet ; 29(2): 208-10, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24025986

RESUMO

Gout, a common disease, is a consequence of hyperuricemia, and increases the risks of hypertension, cardiovascular diseases, cerebrovascular diseases and renal failure. Gout can be classified into 3 types: the renal underexcretion (RUE) type, renal overload type and combined type. RUE type is a major type of gout; however, its genetic causes are still unclear. Since human organic anion transporter 4 (OAT4/SLC22A11) is expressed in the kidney and mediates urate transport, we investigated the effects of a common variant of OAT4/SLC22A11 on the susceptibility to gout. Five hundred and forty-five Japanese male gout cases and 1,115 male individuals as a control group were genotyped with rs17300741, a single nucleotide polymorphism in the OAT4/SLC22A11 gene. The association analysis of rs17300741 showed no significant association for all gout cases; however, there was a slight but significant association for RUE type gout cases (p = 0.049). These results also suggest that OAT4 contributes to urate transport at the apical membrane of renal proximal tubule cells in humans. Our findings make it clear for the first time that a common variant of OAT4/SLC22A11 is associated with RUE type gout, a major gout subtype.


Assuntos
Gota/genética , Gota/metabolismo , Rim/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/genética , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Polimorfismo de Nucleotídeo Único , Eliminação Renal/genética , Ácido Úrico/metabolismo , Povo Asiático/genética , Estudos de Casos e Controles , Distribuição de Qui-Quadrado , Frequência do Gene , Estudos de Associação Genética , Predisposição Genética para Doença , Gota/etnologia , Humanos , Japão , Masculino , Razão de Chances , Fenótipo , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA