Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin J Am Soc Nephrol ; 16(7): 1025-1036, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33985991

RESUMO

BACKGROUND AND OBJECTIVES: In the rare disease primary hyperoxaluria type 1, overproduction of oxalate by the liver causes kidney stones, nephrocalcinosis, kidney failure, and systemic oxalosis. Lumasiran, an RNA interference therapeutic, suppresses glycolate oxidase, reducing hepatic oxalate production. The objective of this first-in-human, randomized, placebo-controlled trial was to evaluate the safety, pharmacokinetic, and pharmacodynamic profiles of lumasiran in healthy participants and patients with primary hyperoxaluria type 1. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: This phase 1/2 study was conducted in two parts. In part A, healthy adults randomized 3:1 received a single subcutaneous dose of lumasiran or placebo in ascending dose groups (0.3-6 mg/kg). In part B, patients with primary hyperoxaluria type 1 randomized 3:1 received up to three doses of lumasiran or placebo in cohorts of 1 or 3 mg/kg monthly or 3 mg/kg quarterly. Patients initially assigned to placebo crossed over to lumasiran on day 85. The primary outcome was incidence of adverse events. Secondary outcomes included pharmacokinetic and pharmacodynamic parameters, including measures of oxalate in patients with primary hyperoxaluria type 1. Data were analyzed using descriptive statistics. RESULTS: Thirty-two healthy participants and 20 adult and pediatric patients with primary hyperoxaluria type 1 were enrolled. Lumasiran had an acceptable safety profile, with no serious adverse events or study discontinuations attributed to treatment. In part A, increases in mean plasma glycolate concentration, a measure of target engagement, were observed in healthy participants. In part B, patients with primary hyperoxaluria type 1 had a mean maximal reduction from baseline of 75% across dosing cohorts in 24-hour urinary oxalate excretion. All patients achieved urinary oxalate levels ≤1.5 times the upper limit of normal. CONCLUSIONS: Lumasiran had an acceptable safety profile and reduced urinary oxalate excretion in all patients with primary hyperoxaluria type 1 to near-normal levels. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER: Study of Lumasiran in Healthy Adults and Patients with Primary Hyperoxaluria Type 1, NCT02706886.


Assuntos
Hiperoxalúria Primária/tratamento farmacológico , Oxalatos/urina , RNA Interferente Pequeno/farmacologia , RNA Interferente Pequeno/farmacocinética , Fármacos Renais/farmacologia , Fármacos Renais/farmacocinética , Adolescente , Adulto , Criança , Feminino , Glicolatos/sangue , Humanos , Hiperoxalúria Primária/sangue , Hiperoxalúria Primária/urina , Masculino , RNA Interferente Pequeno/efeitos adversos , Fármacos Renais/efeitos adversos , Método Simples-Cego , Adulto Jovem
2.
N Engl J Med ; 384(13): 1216-1226, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33789010

RESUMO

BACKGROUND: Primary hyperoxaluria type 1 (PH1) is a rare genetic disease caused by hepatic overproduction of oxalate that leads to kidney stones, nephrocalcinosis, kidney failure, and systemic oxalosis. Lumasiran, an investigational RNA interference (RNAi) therapeutic agent, reduces hepatic oxalate production by targeting glycolate oxidase. METHODS: In this double-blind, phase 3 trial, we randomly assigned (in a 2:1 ratio) patients with PH1 who were 6 years of age or older to receive subcutaneous lumasiran or placebo for 6 months (with doses given at baseline and at months 1, 2, 3, and 6). The primary end point was the percent change in 24-hour urinary oxalate excretion from baseline to month 6 (mean percent change across months 3 through 6). Secondary end points included the percent change in the plasma oxalate level from baseline to month 6 (mean percent change across months 3 through 6) and the percentage of patients with 24-hour urinary oxalate excretion no higher than 1.5 times the upper limit of the normal range at month 6. RESULTS: A total of 39 patients underwent randomization; 26 were assigned to the lumasiran group and 13 to the placebo group. The least-squares mean difference in the change in 24-hour urinary oxalate excretion (lumasiran minus placebo) was -53.5 percentage points (P<0.001), with a reduction in the lumasiran group of 65.4% and an effect seen as early as month 1. The between-group differences for all hierarchically tested secondary end points were significant. The difference in the percent change in the plasma oxalate level (lumasiran minus placebo) was -39.5 percentage points (P<0.001). In the lumasiran group, 84% of patients had 24-hour urinary oxalate excretion no higher than 1.5 times the upper limit of the normal range at month 6, as compared with 0% in the placebo group (P<0.001). Mild, transient injection-site reactions were reported in 38% of lumasiran-treated patients. CONCLUSIONS: Lumasiran reduced urinary oxalate excretion, the cause of progressive kidney failure in PH1. The majority of patients who received lumasiran had normal or near-normal levels after 6 months of treatment. (Funded by Alnylam Pharmaceuticals; ILLUMINATE-A ClinicalTrials.gov number, NCT03681184.).


Assuntos
Hiperoxalúria Primária/tratamento farmacológico , Oxalatos/urina , RNA Interferente Pequeno/uso terapêutico , Terapêutica com RNAi , Adolescente , Adulto , Criança , Creatinina/urina , Método Duplo-Cego , Feminino , Taxa de Filtração Glomerular , Humanos , Hiperoxalúria Primária/sangue , Hiperoxalúria Primária/complicações , Hiperoxalúria Primária/urina , Cálculos Renais/prevenção & controle , Masculino , Pessoa de Meia-Idade , Oxalatos/sangue , Oxalatos/metabolismo , RNA Interferente Pequeno/efeitos adversos , Adulto Jovem
3.
Kidney Int ; 93(4): 961-967, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29398133

RESUMO

The clinical diagnosis of inherited renal tubulopathies can be challenging as they are rare and characterized by significant phenotypic variability. Advances in sequencing technologies facilitate the establishment of a molecular diagnosis. Therefore, we determined the diagnostic yield of a next generation sequencing panel assessing relevant disease genes in children followed through three national networks with a clinical diagnosis of a renal tubulopathy. DNA was amplified with a kit provided by the European Consortium for High-Throughput Research in Rare Kidney Diseases with nine multiplex PCR reactions. This kit produced 571 amplicons covering 37 genes associated with tubulopathies followed by massive parallel sequencing and bioinformatic interpretation. Identified mutations were confirmed by Sanger sequencing. Overall, 384 index patients and 16 siblings were assessed. Most common clinical diagnoses were 174 patients with Bartter/Gitelman syndrome and 76 with distal renal tubular acidosis. A total of 269 different variants were identified in 27 genes, of which 95 variants were considered likely, 136 definitely pathogenic and 100 had not been described at annotation. These mutations established a genetic diagnosis in 245 of the index patients. Genetic testing changed the clinical diagnosis in 16 cases and provided insights into the phenotypic spectrum of the respective disorders. Our results demonstrate a high diagnostic yield of genetic testing in children with a clinical diagnosis of a renal tubulopathy, consistent with a predominantly genetic etiology in known disease genes. Thus, genetic testing helped establish a definitive diagnosis in almost two-thirds of patients thereby informing prognosis, management and genetic counseling.


Assuntos
Análise Mutacional de DNA/métodos , Sequenciamento de Nucleotídeos em Larga Escala , Mutação , Erros Inatos do Transporte Tubular Renal/genética , Acidose Tubular Renal/diagnóstico , Acidose Tubular Renal/genética , Adolescente , Fatores Etários , Síndrome de Bartter/diagnóstico , Síndrome de Bartter/genética , Estudos de Casos e Controles , Criança , Pré-Escolar , Europa (Continente) , Feminino , Marcadores Genéticos , Predisposição Genética para Doença , Síndrome de Gitelman/diagnóstico , Síndrome de Gitelman/genética , Hereditariedade , Humanos , Lactente , Recém-Nascido , Masculino , Reação em Cadeia da Polimerase Multiplex , Linhagem , Fenótipo , Valor Preditivo dos Testes , Kit de Reagentes para Diagnóstico , Erros Inatos do Transporte Tubular Renal/diagnóstico , Fatores de Risco
5.
Pediatr Nephrol ; 32(6): 987-996, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28188436

RESUMO

BACKGROUND: Distal renal tubular acidosis (dRTA) is characterized by hyperchloraemic metabolic acidosis, hypokalaemia, hypercalciuria and nephrocalcinosis. It is due to reduced urinary acidification by the α-intercalated cells in the collecting duct and can be caused by mutations in genes that encode subunits of the vacuolar H+-ATPase (ATP6V1B1, ATP6V0A4) or the anion exchanger 1 (SLC4A1). Treatment with alkali is the mainstay of therapy. METHODS: This study is an analysis of clinical data from a long-term follow-up of 24 children with dRTA in a single centre, including a genetic analysis. RESULTS: Of the 24 children included in the study, genetic diagnosis was confirmed in 19 patients, with six children having mutations in ATP6V1B1, ten in ATP6V0A4 and three in SLC4A1; molecular diagnosis was not available for five children. Five novel mutations were detected (2 in ATP6V1B1 and 3 in ATP6V0A4). Two-thirds of patients presented with features of proximal tubular dysfunction leading to an erroneous diagnosis of renal Fanconi syndrome. The proximal tubulopathy disappeared after resolution of acidosis, indicating the importance of following proximal tubular function to establish the correct diagnosis. Growth retardation with a height below -2 standard deviation score was found in ten patients at presentation, but persisted in only three of these children once established on alkali treatment. Sensorineural hearing loss was found in five of the six patients with an ATP6V1B1 mutation. Only one patient with an ATP6V0A4 mutation had sensorineural hearing loss during childhood. Nine children developed medullary cysts, but without apparent clinical consequences. Cyst development in this cohort was not correlated with age at therapy onset, molecular diagnosis, growth parameters or renal function. CONCLUSION: In general, the prognosis of dRTA is good in children treated with alkali.


Assuntos
Acidose Tubular Renal/epidemiologia , Acidose Tubular Renal/genética , Álcalis/uso terapêutico , Transtornos do Crescimento/epidemiologia , Perda Auditiva Neurossensorial/epidemiologia , Túbulos Renais Coletores/metabolismo , Acidose Tubular Renal/tratamento farmacológico , Proteína 1 de Troca de Ânion do Eritrócito/genética , Pré-Escolar , Estudos de Coortes , Comorbidade , Cistos/epidemiologia , Cistos/genética , Análise Mutacional de DNA , Feminino , Seguimentos , Testes Genéticos , Taxa de Filtração Glomerular , Transtornos do Crescimento/genética , Perda Auditiva Neurossensorial/genética , Humanos , Lactente , Recém-Nascido , Medula Renal/patologia , Túbulos Renais Coletores/citologia , Masculino , Mutação , ATPases Vacuolares Próton-Translocadoras/genética
6.
Hum Mol Genet ; 24(18): 5079-92, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26082470

RESUMO

The adaptor protein-2 sigma subunit (AP2σ2) is pivotal for clathrin-mediated endocytosis of plasma membrane constituents such as the calcium-sensing receptor (CaSR). Mutations of the AP2σ2 Arg15 residue result in familial hypocalciuric hypercalcaemia type 3 (FHH3), a disorder of extracellular calcium (Ca(2+) o) homeostasis. To elucidate the role of AP2σ2 in Ca(2+) o regulation, we investigated 65 FHH probands, without other FHH-associated mutations, for AP2σ2 mutations, characterized their functional consequences and investigated the genetic mechanisms leading to FHH3. AP2σ2 mutations were identified in 17 probands, comprising 5 Arg15Cys, 4 Arg15His and 8 Arg15Leu mutations. A genotype-phenotype correlation was observed with the Arg15Leu mutation leading to marked hypercalcaemia. FHH3 probands harboured additional phenotypes such as cognitive dysfunction. All three FHH3-causing AP2σ2 mutations impaired CaSR signal transduction in a dominant-negative manner. Mutational bias was observed at the AP2σ2 Arg15 residue as other predicted missense substitutions (Arg15Gly, Arg15Pro and Arg15Ser), which also caused CaSR loss-of-function, were not detected in FHH probands, and these mutations were found to reduce the numbers of CaSR-expressing cells. FHH3 probands had significantly greater serum calcium (sCa) and magnesium (sMg) concentrations with reduced urinary calcium to creatinine clearance ratios (CCCR) in comparison with FHH1 probands with CaSR mutations, and a calculated index of sCa × sMg/100 × CCCR, which was ≥ 5.0, had a diagnostic sensitivity and specificity of 83 and 86%, respectively, for FHH3. Thus, our studies demonstrate AP2σ2 mutations to result in a more severe FHH phenotype with genotype-phenotype correlations, and a dominant-negative mechanism of action with mutational bias at the Arg15 residue.


Assuntos
Complexo 2 de Proteínas Adaptadoras/genética , Subunidades sigma do Complexo de Proteínas Adaptadoras/genética , Códon , Genes Dominantes , Estudos de Associação Genética , Hipercalcemia/congênito , Mutação , Complexo 2 de Proteínas Adaptadoras/química , Subunidades sigma do Complexo de Proteínas Adaptadoras/química , Adolescente , Adulto , Substituição de Aminoácidos , Biomarcadores , Linhagem Celular , Criança , Pré-Escolar , Diagnóstico Diferencial , Feminino , Expressão Gênica , Humanos , Hipercalcemia/diagnóstico , Hipercalcemia/genética , Lactente , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Linhagem , Fenótipo , Conformação Proteica , Relação Estrutura-Atividade , Adulto Jovem
7.
Clin Sci (Lond) ; 126(10): 721-6, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24266877

RESUMO

The study of families with rare inherited forms of hypo- and hyper-tension has been one of the most successful strategies to probe the molecular pathophysiology of blood pressure control and has revealed dysregulation of distal nephron Na+ reabsorption to be a common mechanism. FHHt (familial hyperkalaemic hypertension; also known as Gordon's syndrome) is a salt-dependent form of hypertension caused by mutations in the regulators of the thiazide-sensitive Na+-Cl- co-transporter NCC [also known as SLC12A3 (solute carrier family 12 member 3)] and is effectively treated by thiazide diuretics and/or dietary salt restriction. Variation in at least four genes can cause FHHt, including WNK1 [With No lysine (=K) 1] and WNK4, KLHL3 (kelch-like family member 3), and CUL3 (cullin 3). In the present study we have identified novel disease-causing variants in CUL3 and KLHL3 segregating in 63% of the pedigrees with previously unexplained FHHt, confirming the importance of these recently described FHHt genes. We have demonstrated conclusively, in two unrelated affected individuals, that rare intronic variants in CUL3 cause the skipping of exon 9 as has been proposed previously. KLHL3 variants all occur in kelch-repeat domains and so probably disrupt WNK complex binding. We have found no evidence of any plausible disease-causing variants within SLC4A8 (an alternative thiazide-sensitive sodium transporter) in this population. The results of the present study support the existing evidence that the CUL3 and KLHL3 gene products are physiologically important regulators of thiazide-sensitive distal nephron NaCl reabsorption, and hence potentially interesting novel anti-hypertensive drug targets. As a third of our non-WNK FHHt families do not have plausible CUL3 or KLHL3 variants, there are probably additional, as yet undiscovered, regulators of the thiazide-sensitive pathways.


Assuntos
Proteínas de Transporte/genética , Proteínas Culina/genética , Predisposição Genética para Doença , Mutação/genética , Pseudo-Hipoaldosteronismo/genética , Proteínas Adaptadoras de Transdução de Sinal , Processamento Alternativo/genética , Segregação de Cromossomos/genética , Análise Mutacional de DNA , Éxons/genética , Família , Feminino , Humanos , Masculino , Proteínas dos Microfilamentos , Linhagem , Fenótipo
8.
Nephrol Dial Transplant ; 27(8): 3191-5, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22391140

RESUMO

BACKGROUND: Mutations in the 4-hydroxy-2-oxoglutarate aldolase (HOGA1) gene have been recently identified in patients with atypical primary hyperoxaluria (PH). However, it was not clearly established whether these mutations caused disease via loss of function or activation of the gene product. METHODS: Whole-gene sequencing of HOGA1 was conducted in 28 unrelated patients with a high clinical suspicion of PH and in whom Types 1 and 2 had been excluded. RESULTS: Fifteen patients were homozygous or compound heterozygous for mutations in HOGA1. In total, seven different mutations were identified including three novel changes: a missense mutation, c.107C > T (p.Ala36Val), and two nonsense mutations c.117C > A (p.Tyr39X) and c.208C > T (p.Arg70X) as well as the previously documented c.860G > T (p.Gly297Val), c.907C > T (p.Arg303Cys) and in-frame c.944_946delAGG (p.Glu315del) mutations. The recurrent c.700 + 5G > T splice site mutation in intron 5 was most common with a frequency of 67%. Expression studies on hepatic messenger RNA demonstrated the pathogenicity of this mutation. CONCLUSIONS: The detection of a patient with two novel nonsense mutations within exon 1 of the gene, c.117C > A (p.Tyr39X) and c.208C > T (p.Arg70X), provides definitive proof that PH Type 3 is due to deficiency of the 4-hydroxy-2-oxoglutarate aldolase enzyme.


Assuntos
Hiperoxalúria Primária/enzimologia , Hiperoxalúria Primária/genética , Mutação , Oxo-Ácido-Liases/deficiência , Oxo-Ácido-Liases/genética , Adulto , Criança , Pré-Escolar , Códon sem Sentido , Análise Mutacional de DNA , Heterozigoto , Homozigoto , Humanos , Hiperoxalúria Primária/classificação , Lactente , Modelos Biológicos , Mutação de Sentido Incorreto , Sítios de Splice de RNA , Deleção de Sequência , Compostos de Espiro , Adulto Jovem
9.
J Pediatr ; 159(6): 1004-11, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21784456

RESUMO

OBJECTIVE: To report new adverse effects of cysteamine. STUDY DESIGN: Detailed clinical information was obtained from the patients' physicians. RESULTS: New adverse events were reported in 8 of 550 patients with cystinosis treated with cysteamine in Europe during the last 5 years. Detailed clinical information was not available for 2 of these patients, 1 of whom died from cerebral ischemia. The 6 evaluable patients developed vascular elbow lesions (6/6), neurologic symptoms (1/6), bone and muscle pain (2/6), and/or skin striae (2/6). Analysis of biopsy specimens from the elbow lesions demonstrated angioendotheliomatosis with irregular collagen fibers. In 3 of the 6 patients, the daily cysteamine dose exceeded the recommended maximum of 1.95 g/m(2)/day. Dose reduction led to improvement of signs and symptoms in all 6 patients, suggesting a causal relationship with cysteamine administration. CONCLUSION: Cysteamine administration can be complicated by the development of skin, vascular, neurologic, muscular, and bone lesions. These lesions improve after cysteamine dose reduction. Doses >1.95 g/m(2)/day should be prescribed with great caution, but underdosing is not advocated.


Assuntos
Cisteamina/toxicidade , Cistinose/tratamento farmacológico , Toxidermias/etiologia , Pré-Escolar , Humanos , Lactente , Masculino
11.
Urol Res ; 37(3): 127-32, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19283374

RESUMO

The primary hyperoxalurias, PH1 and PH2, are inherited disorders caused by deficiencies of alanine:glyoxylate aminotransferase and glyoxylate reductase, respectively. Mutations in either of these enzymes leads to endogenous oxalate overproduction primarily in the liver, but most pathological effects are exhibited in the kidney ultimately leading to end-stage renal failure and systemic oxalosis. To provide a non-invasive means of accessing kidney cells from individuals with primary hyperoxaluria, we have derived primary cultures of renal proximal tubule cells from the urine of these patients. The cells stain positively for the epithelial markers pan-cytokeratin and zonula occludens 1 and the proximal tubule marker gamma-glutamyl transpeptidase. Mutation analysis confirmed that the cultured cells had the same genotype as the leucocytes of the patients and also expressed glyoxylate reductase at the mRNA level, illustrating their potential value as a source of renal material from these individuals.


Assuntos
Hiperoxalúria Primária/metabolismo , Hiperoxalúria Primária/patologia , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Adolescente , Oxirredutases do Álcool/genética , Sequência de Bases , Células Cultivadas , Criança , Pré-Escolar , Primers do DNA/genética , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Genótipo , Humanos , Hiperoxalúria Primária/genética , Hiperoxalúria Primária/urina , Lactente , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Urina/citologia , Adulto Jovem
12.
Kidney Int ; 75(8): 848-55, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19165178

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) caused by mutations in PKD1 is significantly more severe than PKD2. Typically, ADPKD presents in adulthood but is rarely diagnosed in utero with enlarged, echogenic kidneys. Somatic mutations are thought crucial for cyst development, but gene dosage is also important since animal models with hypomorphic alleles develop cysts, but are viable as homozygotes. We screened for mutations in PKD1 and PKD2 in two consanguineous families and found PKD1 missense variants predicted to be pathogenic. In one family, two siblings homozygous for R3277C developed end stage renal disease at ages 75 and 62 years, while six heterozygotes had few cysts. In the other family, the father and two children with moderate to severe disease were homozygous for N3188S. In both families homozygous disease was associated with small cysts of relatively uniform size while marked cyst heterogeneity is typical of ADPKD. In another family, one patient diagnosed in childhood was found to be a compound heterozygote for the PKD1 variants R3105W and R2765C. All three families had evidence of developmental defects of the collecting system. Three additional ADPKD families with in utero onset had a truncating mutation in trans with either R3277C or R2765C. These cases suggest the presence of incompletely penetrant PKD1 alleles. The alleles alone may result in mild cystic disease; two such alleles cause typical to severe disease; and, in combination with an inactivating allele, are associated with early onset disease. Our study indicates that the dosage of functional PKD1 protein may be critical for cyst initiation.


Assuntos
Alelos , Dosagem de Genes , Penetrância , Canais de Cátion TRPP/genética , Cistos/genética , Análise Mutacional de DNA , Saúde da Família , Genótipo , Humanos , Falência Renal Crônica , Mutação de Sentido Incorreto , Linhagem
14.
Pediatr Nephrol ; 21(4): 503-9, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16508773

RESUMO

We have determined levels of glutathione (GSH), ATP, mitochondrial complex activity and apoptosis rate in proximal tubular cells (PTCs) exfoliated from urine in cystinotic (n=9) and control (n=9) children. Intracellular GSH was significantly depleted in cystinotic PTCs compared with controls (6.8 nmol GSH/mg protein vs 11.8 nmol GSH/mg protein; P<0.001), but there were no significant differences in mitochondrial complex activities or ATP levels under basal conditions. Cystinotic PTCs showed significantly increased apoptosis rate. After PTCs had been stressed by hypoxia, there was further depletion of GSH in cystinotic and control PTCs (2.4 nmol GSH/mg protein vs 7.2 nmol GSH/mg protein; P<0.001). Hypoxic stress led to increased complex I and complex IV activities in control but not in cystinotic PTCs. ATP levels were significantly reduced in cystinotic PTCs after hypoxic stress (12.2 nmol/mg protein vs 26.9 nmol/mg protein; P<0.001). GSH depletion occurs in this in vitro model of cystinotic PTCs, is exaggerated by hypoxic stress and may contribute to reduced ATP and failure to increase complex I/IV activities. Apoptotic rate is also increased, and these mechanisms may contribute to cellular dysfunction in cultured, human cystinotic PTCs.


Assuntos
Apoptose , Cistinose/metabolismo , Cistinose/patologia , Glutationa/metabolismo , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Células Cultivadas , Criança , Humanos
15.
Pediatr Nephrol ; 20(8): 1098-105, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15940548

RESUMO

Thin glomerular basement membrane (GBM) disease is generally known to have a good renal prognosis, although renal insufficiency has sometimes been reported and the overlap with Alport syndrome implies that a good prognosis cannot be guaranteed. In order to shed light on long-term prognosis of thin GBM disease we have retrospectively evaluated 22 children with persistent haematuria and biopsy-proven thin GBM. Mean follow up was 7 years (range 2-17 years), mean age at onset was 7 years (range 1.5-15). Biopsies were performed a mean of 3.8 years after detection of hematuria. The light microscopy (LM) and immunofluorescence (IF) findings were essentially unremarkable in all of the children, while electron microscopy (EM) showed thinning of the GBM in all cases and no changes characteristic of Alport syndrome. The family history was positive for renal disease in 17 (77.3%) patients with hematuria in 8 (36.3%) families, and hematuria with renal failure (RF) or deafness in 9 (40.9%). It was completely negative for renal disease in 4 (18.2%) and unavailable in 1 (4.5%). Four patients (18%) showed a decline in renal function after 6, 8, 9 and 12 years of follow-up, and 1 of these also developed hearing impairment. None developed hypertension. Our study suggests that thin GBM disease is not always benign and a child with thin GBM should never be assigned such a prognosis, especially if there is a family history of renal impairment or deafness, where careful follow-up is needed due to the risk of late onset renal failure.


Assuntos
Colágeno Tipo IV/genética , Hematúria/genética , Hematúria/patologia , Glomérulos Renais/patologia , Adolescente , Membrana Basal/patologia , Biópsia , Criança , Pré-Escolar , Feminino , Seguimentos , Taxa de Filtração Glomerular , Humanos , Lactente , Masculino , Estudos Retrospectivos
16.
Pediatr Nephrol ; 20(2): 136-40, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15627171

RESUMO

The renal Fanconi syndrome (FS) is characterised by generalised proximal tubular dysfunction. Cystinosis is the most common genetic cause of the FS and results from defective function of cystinosin, due to mutations of the CTNS gene leading to intralysosomal cystine accumulation. Despite these advances in our understanding of the molecular basis of cystinosis, the mechanisms of proximal tubular cell (PTC) dysfunction are still unknown. We have further characterised an in vitro model of cultured cells exfoliated in patients' urine. We cultured cells from 9 cystinosis children, 4 children with Lowe syndrome and 8 controls. PTC phenotype and homogeneity were studied by lectin staining, immunocytochemistry (using ZO-1 as an epithelial marker) and enzyme cytochemistry (using gamma-glutamyltransferase as a PTC marker). All cultured cells showed PTC phenotype. Cystinosin was stained using anti-cystinosin antibody and co-localised to the lysosomes with LAMP-2 antibody. Additionally, we have demonstrated significantly elevated intracellular cystine levels in cystinotic cell lines (13.8+/-2.3 nmol 1/2 cystine/mg protein, P <0.001) compared with controls. We believe this in vitro model will allow further investigation of cystinosis and other types of the FS.


Assuntos
Cistinose/urina , Síndrome de Fanconi/urina , Túbulos Renais Proximais/fisiopatologia , Células Cultivadas , Criança , Pré-Escolar , Humanos , Túbulos Renais Proximais/patologia , Modelos Biológicos , Síndrome Oculocerebrorrenal
17.
Nephron Clin Pract ; 98(2): c45-8, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15499205

RESUMO

The aetiology of stones in children differs from that in adults. Young children, especially boys, are prone to infective stones, although this type of calculi is decreasing in frequency over time in prosperous countries. Two monogenic causes, cystinuria and hyperoxaluria, each account for 5-15% of paediatric stones. Increased factors for stone formation in children include prematurity, neurological problems, ketogenic diet and reconstructed or augmented bladders. Hypercalciuria is commonly found in paediatric stone formers, is usually idiopathic and is only rarely associated with hypercalcaemia. All children with stones should undergo a metabolic evaluation.


Assuntos
Cálculos Urinários/etiologia , Cálcio/urina , Criança , Cistinúria/complicações , Humanos , Hiperoxalúria/complicações , Purinas/análise , Purinas/metabolismo , Cálculos Urinários/química , Cálculos Urinários/urina , Infecções Urinárias/complicações
18.
Eur J Pediatr ; 163(10): 589-94, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15241688

RESUMO

UNLABELLED: Familial hypocalciuric hypercalcaemia (FHH) is an autosomal dominant condition due to heterozygous loss of function calcium sensing receptor (CaSR) mutations. However, individuals who are homozygous for CaSR mutations have neonatal severe hyperparathyroidism (NSHPT), which unlike the relatively benign and asymptomatic FHH can be fatal without parathyroidectomy. We report three patients with NSHPT associated with marked hypercalcaemia and severe hyperparathyroidism with related skeletal demineralisation. We describe the novel use of intravenous pamidronate in NSHPT, to control severe hypercalcaemia in these patients prior to parathyroidectomy and in one individual as a rescue therapy to stabilise life-threatening demineralisation. Furthermore, a marked phenotypic heterogeneity was observed amongst four members from a large kindred with the same homozygous CaSR mutations: one patient would have died without parathyroidectomy in infancy; a second patient survived infancy but underwent parathyroidectomy in early childhood following severe symptomatic hypercalcaemic episodes; whilst the other two patients have survived to adolescence without parathyroidectomy. Additionally, in contrast to the literature these two individuals suffered minimal morbidity. CONCLUSION: We commend the short-term use of pamidronate in neonatal severe hyperparathyroidism to treat extreme hypercalcaemia and halt hyperparathyroid-driven skeletal demineralisation in preparation for parathyroidectomy. The remarkable degree of phenotypic variation demonstrated remains unexplained without functional studies; this variability highlights the challenge of treating this rare condition.


Assuntos
Hipercalcemia/complicações , Hiperparatireoidismo/complicações , Antineoplásicos/uso terapêutico , Análise Mutacional de DNA , Desidratação/etiologia , Difosfonatos/uso terapêutico , Feminino , Genótipo , Humanos , Hipercalcemia/tratamento farmacológico , Hipercalcemia/genética , Hiperparatireoidismo/genética , Hiperparatireoidismo/cirurgia , Recém-Nascido , Masculino , Pamidronato , Glândulas Paratireoides/cirurgia , Linhagem , Fenótipo , Reação em Cadeia da Polimerase , Receptores de Detecção de Cálcio/genética , Resultado do Tratamento
19.
Pediatr Nephrol ; 18(7): 712-3, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12750979

RESUMO

3-methylglutaconic aciduria is frequently found during urine organic acid analysis and is widely regarded as a marker of a mitochondrial disorder, the clinical features of which are very heterogeneous. We describe two siblings with 3-methylglutaconic aciduria in whom renal ultrasonography showed echogenic medullae consistent with nephrocalcinosis. One patient also developed medullary cysts. In both children renal function was normal and neither had any plasma or urinary evidence of tubulopathy. The presence of nephrocalcinosis and medullary cysts in patients with 3-methylglutaconic aciduria adds to the heterogeneous clinical presentation of this group of disorders.


Assuntos
Glutaratos/urina , Doenças Renais Císticas/complicações , Medula Renal/patologia , Erros Inatos do Metabolismo/complicações , Nefrocalcinose/etiologia , Criança , Feminino , Humanos , Doenças Renais Císticas/diagnóstico por imagem , Medula Renal/diagnóstico por imagem , Erros Inatos do Metabolismo/patologia , Erros Inatos do Metabolismo/urina , Nefrocalcinose/diagnóstico por imagem , Nefrocalcinose/patologia , Ultrassonografia
20.
Pediatr Nephrol ; 18(7): 700-2, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12734749

RESUMO

We report a case of severe nephrocalcinosis related to hypercalcaemia in a newborn with glucose-galactose malabsorption. He presented with poor growth and was noted to have polyuria, which was later recognised to be severe watery diarrhoea. We discuss the possible aetiological factors for nephrocalcinosis in this condition.


Assuntos
Galactose/metabolismo , Glucose/metabolismo , Síndromes de Malabsorção/complicações , Síndromes de Malabsorção/patologia , Nefrocalcinose/complicações , Nefrocalcinose/patologia , Cálcio/urina , Diarreia/etiologia , Transtornos do Crescimento/etiologia , Humanos , Hipercalcemia/etiologia , Hipercalcemia/patologia , Recém-Nascido , Cariotipagem , Rim/diagnóstico por imagem , Rim/patologia , Síndromes de Malabsorção/dietoterapia , Masculino , Nefrocalcinose/dietoterapia , Oxalatos/urina , Poliúria/etiologia , Ultrassonografia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...