Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
N Engl J Med ; 370(2): 129-38, 2014 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-24401050

RESUMO

BACKGROUND: In renal Fanconi's syndrome, dysfunction in proximal tubular cells leads to renal losses of water, electrolytes, and low-molecular-weight nutrients. For most types of isolated Fanconi's syndrome, the genetic cause and underlying defect remain unknown. METHODS: We clinically and genetically characterized members of a five-generation black family with isolated autosomal dominant Fanconi's syndrome. We performed genomewide linkage analysis, gene sequencing, biochemical and cell-biologic investigations of renal proximal tubular cells, studies in knockout mice, and functional evaluations of mitochondria. Urine was studied with the use of proton nuclear magnetic resonance ((1)H-NMR) spectroscopy. RESULTS: We linked the phenotype of this family's Fanconi's syndrome to a single locus on chromosome 3q27, where a heterozygous missense mutation in EHHADH segregated with the disease. The p.E3K mutation created a new mitochondrial targeting motif in the N-terminal portion of EHHADH, an enzyme that is involved in peroxisomal oxidation of fatty acids and is expressed in the proximal tubule. Immunocytofluorescence studies showed mistargeting of the mutant EHHADH to mitochondria. Studies of proximal tubular cells revealed impaired mitochondrial oxidative phosphorylation and defects in the transport of fluids and a glucose analogue across the epithelium. (1)H-NMR spectroscopy showed elevated levels of mitochondrial metabolites in urine from affected family members. Ehhadh knockout mice showed no abnormalities in renal tubular cells, a finding that indicates a dominant negative nature of the mutation rather than haploinsufficiency. CONCLUSIONS: Mistargeting of peroxisomal EHHADH disrupts mitochondrial metabolism and leads to renal Fanconi's syndrome; this indicates a central role of mitochondria in proximal tubular function. The dominant negative effect of the mistargeted protein adds to the spectrum of monogenic mechanisms of Fanconi's syndrome. (Funded by the European Commission Seventh Framework Programme and others.).


Assuntos
Síndrome de Fanconi/genética , Túbulos Renais Proximais/metabolismo , Mitocôndrias/metabolismo , Mutação de Sentido Incorreto , Enzima Bifuncional do Peroxissomo/genética , Sequência de Aminoácidos , Animais , População Negra , Cromossomos Humanos Par 3 , Modelos Animais de Doenças , Síndrome de Fanconi/etnologia , Feminino , Ligação Genética , Humanos , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Linhagem , Enzima Bifuncional do Peroxissomo/química , Enzima Bifuncional do Peroxissomo/metabolismo , Fenótipo , Análise de Sequência de DNA
2.
Pediatr Nephrol ; 30(6): 945-51, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25526929

RESUMO

BACKGROUND AND OBJECTIVES: Nephropathic cystinosis is a lysosomal storage disorder characterized by renal tubular Fanconi syndrome in infancy and glomerular damage leading to renal failure at ∼10 years of age. Therapy with the cystine-depleting agent cysteamine postpones renal failure, but the degree of compliance with this treatment has not been correlated with preservation of kidney function. METHODS: We assessed leucocyte cystine depletion by cysteamine and created the composite compliance score that incorporates the extent of leucocyte cystine depletion, as well as duration of cysteamine treatment, into a single integer. Age at renal failure was used to gauge preservation of renal function, and the Fanconi syndrome index (FSI), a measure of aminoaciduria, was used to assess renal tubular Fanconi syndrome. RESULTS: Age at renal failure varied directly and linearly with the composite compliance score (y = 0.3x +8.8; R(2) = 0.61). The slope indicated that for every year of excellent cystine depletion, nearly 1 year of renal function was preserved. Age at renal failure correlated roughly with mean leucocyte cystine level, but not with mean cysteamine dosage. There was no correlation between the FSI and the composite compliance score. CONCLUSIONS: Greater compliance with oral cysteamine therapy yields greater preservation of renal glomerular, but not tubular, function. Oral cysteamine therapy should be given at the maximum tolerated dose, within the recommended limits.


Assuntos
Cisteamina/uso terapêutico , Eliminadores de Cistina/uso terapêutico , Cistina/sangue , Cistinose/tratamento farmacológico , Síndrome de Fanconi/tratamento farmacológico , Falência Renal Crônica/prevenção & controle , Rim/efeitos dos fármacos , Leucócitos/efeitos dos fármacos , Adesão à Medicação , Administração Oral , Adolescente , Adulto , Fatores Etários , Biomarcadores/sangue , Criança , Cisteamina/administração & dosagem , Eliminadores de Cistina/administração & dosagem , Cistinose/sangue , Cistinose/complicações , Cistinose/diagnóstico , Cistinose/fisiopatologia , Síndrome de Fanconi/diagnóstico , Síndrome de Fanconi/etiologia , Síndrome de Fanconi/fisiopatologia , Feminino , Humanos , Rim/fisiopatologia , Falência Renal Crônica/diagnóstico , Falência Renal Crônica/etiologia , Falência Renal Crônica/fisiopatologia , Testes de Função Renal , Leucócitos/metabolismo , Modelos Lineares , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Estudos Retrospectivos , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento , Adulto Jovem
3.
Gastroenterology ; 144(1): 112-121.e2, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23041322

RESUMO

BACKGROUND & AIMS: Autosomal recessive polycystic kidney disease (ARPKD), the most common ciliopathy of childhood, is characterized by congenital hepatic fibrosis and progressive cystic degeneration of kidneys. We aimed to describe congenital hepatic fibrosis in patients with ARPKD, confirmed by detection of mutations in PKHD1. METHODS: Patients with ARPKD and congenital hepatic fibrosis were evaluated at the National Institutes of Health from 2003 to 2009. We analyzed clinical, molecular, and imaging data from 73 patients (age, 1-56 years; average, 12.7 ± 13.1 years) with kidney and liver involvement (based on clinical, imaging, or biopsy analyses) and mutations in PKHD1. RESULTS: Initial symptoms were liver related in 26% of patients, and others presented with kidney disease. One patient underwent liver and kidney transplantation, and 10 others received kidney transplants. Four presented with cholangitis and one with variceal bleeding. Sixty-nine percent of patients had enlarged left lobes on magnetic resonance imaging, 92% had increased liver echogenicity on ultrasonography, and 65% had splenomegaly. Splenomegaly started early in life; 60% of children younger than 5 years had enlarged spleens. Spleen volume had an inverse correlation with platelet count and prothrombin time but not with serum albumin level. Platelet count was the best predictor of spleen volume (area under the curve of 0.88905), and spleen length corrected for patient's height correlated inversely with platelet count (R(2) = 0.42, P < .0001). Spleen volume did not correlate with renal function or type of PKHD1 mutation. Twenty-two of 31 patients who underwent endoscopy were found to have varices. Five had variceal bleeding, and 2 had portosystemic shunts. Forty-percent had Caroli syndrome, and 30% had an isolated dilated common bile duct. CONCLUSIONS: Platelet count is the best predictor of the severity of portal hypertension, which has early onset but is underdiagnosed in patients with ARPKD. Seventy percent of patients with ARPKD have biliary abnormalities. Kidney and liver disease are independent, and variability in severity is not explainable by type of PKHD1 mutation; ClinicalTrials.gov number, NCT00068224.


Assuntos
Hipertensão Portal/fisiopatologia , Cirrose Hepática/congênito , Cirrose Hepática/patologia , Rim Policístico Autossômico Recessivo/genética , Receptores de Superfície Celular/genética , Adolescente , Adulto , Fosfatase Alcalina/sangue , Criança , Pré-Escolar , Colangiopancreatografia por Ressonância Magnética , Endoscopia Gastrointestinal , Varizes Esofágicas e Gástricas/etiologia , Feminino , Humanos , Hipertensão Portal/sangue , Hipertensão Portal/complicações , Lactente , Transplante de Rim , Cirrose Hepática/diagnóstico por imagem , Cirrose Hepática/genética , Transplante de Fígado , Masculino , Pessoa de Meia-Idade , Mutação , Tamanho do Órgão , Contagem de Plaquetas , Rim Policístico Autossômico Recessivo/complicações , Pressão na Veia Porta , Tempo de Protrombina , Albumina Sérica , Índice de Gravidade de Doença , Esplenomegalia/diagnóstico por imagem , Ultrassonografia Doppler em Cores , Adulto Jovem , gama-Glutamiltransferase/sangue
4.
Development ; 137(15): 2587-96, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20627962

RESUMO

Costeff Syndrome, which is caused by mutations in the OPTIC ATROPHY 3 (OPA3) gene, is an early-onset syndrome characterized by urinary excretion of 3-methylglutaconic acid (MGC), optic atrophy and movement disorders, including ataxia and extrapyramidal dysfunction. The OPA3 protein is enriched in the inner mitochondrial membrane and has mitochondrial targeting signals, but a requirement for mitochondrial localization has not been demonstrated. We find zebrafish opa3 mRNA to be expressed in the optic nerve and retinal layers, the counterparts of which in humans have high mitochondrial activity. Transcripts of zebrafish opa3 are also expressed in the embryonic brain, inner ear, heart, liver, intestine and swim bladder. We isolated a zebrafish opa3 null allele for which homozygous mutants display increased MGC levels, optic nerve deficits, ataxia and an extrapyramidal movement disorder. This correspondence of metabolic, ophthalmologic and movement abnormalities between humans and zebrafish demonstrates a phylogenetic conservation of OPA3 function. We also find that delivery of exogenous Opa3 can reduce increased MGC levels in opa3 mutants, and this reduction requires the mitochondrial localization signals of Opa3. By manipulating MGC precursor availability, we infer that elevated MGC in opa3 mutants derives from extra-mitochondrial HMG-CoA through a non-canonical pathway. The opa3 mutants have normal mitochondrial oxidative phosphorylation profiles, but are nonetheless sensitive to inhibitors of the electron transport chain, which supports clinical recommendations that individuals with Costeff Syndrome avoid mitochondria-damaging agents. In summary, this paper introduces a faithful Costeff Syndrome model and demonstrates a requirement for mitochondrial OPA3 to limit HMG-CoA-derived MGC and protect the electron transport chain against inhibitory compounds.


Assuntos
Glutaratos/metabolismo , Proteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Proteínas/genética , Proteínas de Peixe-Zebra/genética , Acil Coenzima A/metabolismo , Alelos , Erros Inatos do Metabolismo dos Aminoácidos/genética , Animais , Modelos Animais de Doenças , Transporte de Elétrons , Proteínas de Membrana/genética , Mitocôndrias/genética , Modelos Biológicos , Modelos Genéticos , Atrofia Óptica/genética , Fosforilação , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
5.
Nat Genet ; 36(9): 999-1002, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15286787

RESUMO

Hartnup disorder, an autosomal recessive defect named after an English family described in 1956 (ref. 1), results from impaired transport of neutral amino acids across epithelial cells in renal proximal tubules and intestinal mucosa. Symptoms include transient manifestations of pellagra (rashes), cerebellar ataxia and psychosis. Using homozygosity mapping in the original family in whom Hartnup disorder was discovered, we confirmed that the critical region for one causative gene was located on chromosome 5p15 (ref. 3). This region is homologous to the area of mouse chromosome 13 that encodes the sodium-dependent amino acid transporter B(0)AT1 (ref. 4). We isolated the human homolog of B(0)AT1, called SLC6A19, and determined its size and molecular organization. We then identified mutations in SLC6A19 in members of the original family in whom Hartnup disorder was discovered and of three Japanese families. The protein product of SLC6A19, the Hartnup transporter, is expressed primarily in intestine and renal proximal tubule and functions as a neutral amino acid transporter.


Assuntos
Doença de Hartnup/genética , Mutação , Sequência de Aminoácidos , Sistemas de Transporte de Aminoácidos Neutros/genética , Animais , Sequência de Bases , Mapeamento Cromossômico , Humanos , Rim/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Linhagem
6.
J Am Soc Nephrol ; 22(3): 443-8, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21183592

RESUMO

The Lowe oculocerebrorenal syndrome is an X-linked disorder characterized by congenital cataracts, cognitive disability, and proximal tubular dysfunction. Both this syndrome and Dent Disease 2 result from loss-of-function mutations in the OCRL gene, which encodes a type II phosphatidylinositol bisphosphate 5-phosphatase. Ocrl-deficient mice are unaffected, however, which we believe reflects a difference in how humans and mice cope with the enzyme deficiency. Inpp5b and INPP5B, paralogous autosomal genes that encode another type II phosphoinositide 5-phosphatase in mice and humans, respectively, might explain the distinct phenotype in the two species because they are the closest paralogs to Ocrl and OCRL in their respective genomes yet differ between the two species with regard to expression and splicing. Here, we generated Ocrl(-/-) mice that express INPP5B but not Inpp5b. Similar to the human syndromes, all showed reduced postnatal growth, low molecular weight proteinuria, and aminoaciduria. Thus, we created an animal model for OCRL and Dent Disease 2 tubulopathy by humanizing a modifier paralog in mice already carrying the mutant disease gene.


Assuntos
Doença de Dent/genética , Modelos Animais de Doenças , Túbulos Renais/fisiopatologia , Síndrome Oculocerebrorrenal/genética , Animais , Feminino , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Monoéster Fosfórico Hidrolases/genética
7.
Mol Genet Metab ; 103(4): 307-14, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21620748

RESUMO

Alkaptonuria is a rare, autosomal recessive disorder of tyrosine degradation due to deficiency of the third enzyme in the catabolic pathway. As a result, homogentisic acid (HGA) accumulates and is excreted in gram quantities in the urine, which turns dark upon alkalization. The first symptoms, occurring in early adulthood, involve a painful, progressively debilitating arthritis of the spine and large joints. Cardiac valvular disease and renal and prostate stones occur later. Previously suggested therapies have failed to show benefit, and management remains symptomatic. Nitisinone, a potent inhibitor of the second enzyme in the tyrosine catabolic pathway, is considered a potential therapy; proof-of-principle studies showed 95% reduction in urinary HGA. Based on those findings, a prospective, randomized clinical trial was initiated in 2005 to evaluate 40 patients over a 36-month period. The primary outcome parameter was hip total range of motion with measures of musculoskeletal function serving as secondary parameters. Biochemically, this study consistently demonstrated 95% reduction of HGA in urine and plasma over the course of 3 years. Clinically, primary and secondary parameters did not prove benefit from the medication. Side effects were infrequent. This trial illustrates the remarkable tolerability of nitisinone, its biochemical efficacy, and the need to investigate its use in younger individuals prior to development of debilitating arthritis.


Assuntos
4-Hidroxifenilpiruvato Dioxigenase/antagonistas & inibidores , Alcaptonúria/tratamento farmacológico , Cicloexanonas/uso terapêutico , Nitrobenzoatos/uso terapêutico , Adulto , Alcaptonúria/sangue , Alcaptonúria/urina , Ácido Homogentísico/sangue , Ácido Homogentísico/urina , Humanos , Pessoa de Meia-Idade , Estudos Prospectivos , Tirosina/metabolismo
8.
Mol Genet Metab ; 103(2): 128-34, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21420888

RESUMO

Hermansky-Pudlak syndrome (HPS) is a rare disorder of oculocutaneous albinism, platelet dysfunction, and in some subtypes, fatal pulmonary fibrosis. There is no effective treatment for the pulmonary fibrosis except lung transplantation, but an initial trial using pirfenidone, an anti-fibrotic agent, showed promising results. The current, randomized, placebo-controlled, prospective, double-blind trial investigated the safety and efficacy of pirfenidone for mild to moderate HPS-1 and 4 pulmonary fibrosis. Subjects were evaluated every 4 months at the National Institutes of Health Clinical Center, and the primary outcome parameter was change in forced vital capacity using repeated measures analysis with random coefficients. Thirty-five subjects with HPS-1 pulmonary fibrosis were enrolled during a 4-year interval; 23 subjects received pirfenidone and 12 received placebo. Four subjects withdrew from the trial, 3 subjects died, and 10 serious adverse events were reported. Both groups experienced similar side effects, especially gastroesophageal reflux. Interim analysis of the primary outcome parameter, performed 12 months after 30 patients were enrolled, showed no statistical difference between the placebo and pirfenidone groups, and the study was stopped due to futility. There were no significant safety concerns. Other clinical trials are indicated to identify single or multiple drug regimens that may be effective in treatment for progressive HPS-1 pulmonary fibrosis.


Assuntos
Síndrome de Hermanski-Pudlak/tratamento farmacológico , Fibrose Pulmonar/tratamento farmacológico , Piridonas/uso terapêutico , Adulto , Feminino , Síndrome de Hermanski-Pudlak/diagnóstico por imagem , Síndrome de Hermanski-Pudlak/mortalidade , Humanos , Masculino , Pessoa de Meia-Idade , Cooperação do Paciente , Fibrose Pulmonar/diagnóstico por imagem , Fibrose Pulmonar/mortalidade , Radiografia , Resultado do Tratamento
9.
Mol Genet Metab ; 99(2): 160-73, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19914852

RESUMO

PKHD1, the gene mutated in autosomal recessive polycystic kidney disease (ARPKD)/congenital hepatic fibrosis (CHF), is an exceptionally large and complicated gene that consists of 86 exons and has a number of alternatively spliced transcripts. Its longest open reading frame contains 67 exons that encode a 4074 amino acid protein called fibrocystin or polyductin. The phenotypes caused by PKHD1 mutations are similarly complicated, ranging from perinatally-fatal PKD to CHF presenting in adulthood with mild kidney disease. To date, more than 300 mutations have been described throughout PKHD1. Most reported cohorts include a large proportion of perinatal-onset ARPKD patients; mutation detection rates vary between 42% and 87%. Here we report PKHD1 sequencing results on 78 ARPKD/CHF patients from 68 families. Differing from previous investigations, our study required survival beyond 6 months and included many adults with a CHF-predominant phenotype. We identified 77 PKHD1 variants (41 novel) including 19 truncating, 55 missense, 2 splice, and 1 small in-frame deletion. Using computer-based prediction tools (GVGD, PolyPhen, SNAP), we achieved a mutation detection rate of 79%, ranging from 63% in the CHF-predominant group to 82% in the remaining families. Prediction of the pathogenicity of missense variants will remain challenging until a functional assay is available. In the meantime, use of PKHD1 sequencing data for clinical decisions requires caution, especially when only novel or rare missense variants are identified.


Assuntos
Variação Genética , Cirrose Hepática/congênito , Cirrose Hepática/genética , Rim Policístico Autossômico Recessivo/complicações , Rim Policístico Autossômico Recessivo/genética , Receptores de Superfície Celular/genética , Adolescente , Adulto , Sequência de Bases , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Cirrose Hepática/complicações , Masculino
10.
Am J Med Genet A ; 152A(10): 2640-5, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20818665

RESUMO

OFD I is an X-linked dominant male-lethal ciliopathy characterized by prominent external features including oral clefts, hamartomas or cysts of the tongue, and digital anomalies. Although these external features are easy to recognize and often lead to diagnosis in early childhood, visceral findings in OFD I, especially the fibrocystic liver and pancreas disease, are under-recognized. In addition, while the occurrence of polycystic kidney disease (PKD) in OFD I is well known, few patients are evaluated and monitored for this complication. We report on two adult females diagnosed with OFD I in infancy, but not evaluated for visceral involvement. In adulthood, they were incidentally found to have severe hypertension and chronic renal insufficiency due to undiagnosed PKD. A pancreatic cystic lesion, also discovered incidentally, was thought to be malignant and led to consideration of major surgery. We present NIH evaluations, including documentation of OFD I mutations, extreme beading of the intrahepatic bile ducts, pancreatic cysts, and tabulate features of reported OFD I cases having hepatic, pancreatic, and renal cystic disease. Liver and pancreas are not routinely evaluated in OFD I patients. Increased awareness and lifelong monitoring of visceral complications, particularly involving the liver, pancreas, and kidney, are essential for timely and accurate treatment.


Assuntos
Fibrose Cística/genética , Cirrose Hepática/genética , Rim Policístico Autossômico Dominante/genética , Proteínas/genética , Anormalidades Múltiplas/genética , Adulto , Fibrose Cística/complicações , Éxons/genética , Feminino , Humanos , Cirrose Hepática/complicações , Masculino , Pessoa de Meia-Idade , Deleção de Sequência , Língua/anormalidades
11.
Hum Mutat ; 30(12): 1611-9, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19862842

RESUMO

Alkaptonuria (AKU) is a rare autosomal recessive metabolic disorder, characterized by accumulation of homogentisic acid, leading to darkened urine, pigmentation of connective tissue (ochronosis), joint and spine arthritis, and destruction of cardiac valves. AKU is due to mutations in the homogentisate dioxygenase gene (HGD) that converts homogentisic acid to maleylacetoacetic acid in the tyrosine catabolic pathway. Here we report a comprehensive mutation analysis of 93 patients enrolled in our study, as well as an extensive update of all previously published HGD mutations associated with AKU. Within our patient cohort, we identified 52 HGD variants, of which 22 were novel. This yields a total of 91 identified HGD variations associated with AKU to date, including 62 missense, 13 splice site, 10 frameshift, 5 nonsense, and 1 no-stop mutation. Most HGD variants reside in exons 3, 6, 8, and 13. We assessed the potential effect of all missense variations on protein function, using five bioinformatic tools specifically designed for interpretation of missense variants (SIFT, POLYPHEN, PANTHER, PMUT, and SNAP). We also analyzed the potential effect of splice-site variants using two different tools (BDGP and NetGene2). This study provides valuable resources for molecular analysis of alkaptonuria and expands our knowledge of the molecular basis of this disease.


Assuntos
Alcaptonúria/enzimologia , Alcaptonúria/genética , Homogentisato 1,2-Dioxigenase/genética , Mutação/genética , Alcaptonúria/diagnóstico por imagem , Alcaptonúria/patologia , Estudos de Coortes , Éxons/genética , Genótipo , Homogentisato 1,2-Dioxigenase/urina , Ácido Homogentísico/urina , Humanos , National Institutes of Health (U.S.) , Fenótipo , Polimorfismo de Nucleotídeo Único/genética , Radiografia , Irmãos , Estados Unidos
12.
Invest Ophthalmol Vis Sci ; 59(12): 4945-4952, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30347088

RESUMO

Purpose: Oral nitisinone has been shown to increase fur and ocular pigmentation in a mouse model of oculocutaneous albinism (OCA) due to hypomorphic mutations in tyrosinase (TYR), OCA1B. This study determines if nitisinone can improve ocular and/or fur pigmentation in a mouse model of OCA type 3 (OCA3), caused by mutation of the tyrosinase-related protein 1 (Tyrp1) gene. Methods: Mice homozygous for a null allele in the Tyrp1 gene (C57BL/6J-Tyrp1 b-J/J) were treated with 8 mg/kg nitisinone or vehicle every other day by oral gavage. Changes in fur and ocular melanin pigmentation were monitored. Mature ocular melanosome number and size were quantified in pigmented ocular structures by electron microscopy. Results: C57BL/6J-Tyrp1 b-J/J mice carry a novel c.403T>A; 404delG mutation in Tyrp1, predicted to result in premature truncation of the TYRP1 protein. Nitisinone treatment resulted in an approximately 7-fold increase in plasma tyrosine concentrations without overt toxicity. After 1 month of treatment, no change in the color of fur or pigmented ocular structures was observed. The distribution of melanosome cross-sectional area was unchanged in ocular tissues. There was no significant difference in the number of pigmented melanosomes in the RPE/choroid of nitisinone-treated and control groups. However, there was a significant difference in the number of pigmented melanosomes in the iris. Conclusions: Treatment of a mouse model of OCA3 with oral nitisinone did not have a favorable clinical effect on melanin production and minimally affected the number of pigmented melanosomes in the iris stroma. As such, treatment of OCA3 patients with nitisinone is unlikely to be therapeutic.


Assuntos
Albinismo Oculocutâneo/tratamento farmacológico , Cicloexanonas/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Nitrobenzoatos/uso terapêutico , Administração Oral , Albinismo Oculocutâneo/sangue , Albinismo Oculocutâneo/genética , Albinismo Oculocutâneo/patologia , Animais , Western Blotting , Modelos Animais de Doenças , Técnicas de Genotipagem , Melaninas/metabolismo , Melanossomas/patologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Microscopia Eletrônica de Transmissão , Oxirredutases/genética , Reação em Cadeia da Polimerase em Tempo Real , Resultado do Tratamento , Tirosina/sangue
13.
N Engl J Med ; 347(26): 2111-21, 2002 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-12501223

RESUMO

BACKGROUND: Alkaptonuria, caused by mutations in the HGO gene and a deficiency of homogentisate 1,2-dioxygenase, results in an accumulation of homogentisic acid (HGA), ochronosis, and destruction of connective tissue. There is no effective therapy for this disorder, although nitisinone inhibits the enzyme that produces HGA. We performed a study to delineate the natural history of alkaptonuria. METHODS: We evaluated 58 patients with alkaptonuria (age range, 4 to 80 years), using clinical, radiographic, biochemical, and molecular methods. A radiographic scoring system was devised to assess the severity of spinal and joint damage. Two patients were treated with nitisinone for 10 and 9 days, respectively. RESULTS: Life-table analyses showed that joint replacement was performed at a mean age of 55 years and that renal stones developed at 64 years, cardiac-valve involvement at 54 years, and coronary-artery calcification at 59 years. Linear regression analysis indicated that the radiographic score for the severity of disease began increasing after the age of 30 years, with a more rapid increase in men than in women. Twenty-three new HGO mutations were identified. In a 51-year-old woman, urinary HGA excretion fell from 2.9 to 0.13 g per day after a 10-day course of nitisinone (7 days at a dose of 0.7 mg per day and 3 days at 2.8 mg per day). In a 59-year-old woman, urinary HGA fell from 6.4 g to 1.7 g per day after nine days of treatment with nitisinone (0.7 mg per day). Plasma tyrosine levels in these patients rose from approximately 1.1 mg per deciliter (60 micromol per liter) in both to approximately 12.8 mg per deciliter (700 micromol per liter) and 23.6 mg per deciliter (1300 micromol per liter), respectively, with no clinical signs or symptoms. CONCLUSIONS: The reported data on the natural history of alkaptonuria provide a basis for the evaluation of long-term therapies. Although nitisinone can reduce HGA production in humans with homogentisate 1,2-dioxygenase deficiency, the long-term safety and efficacy of this treatment require further evaluation.


Assuntos
Alcaptonúria , Dioxigenases , Oxigenases/genética , 4-Hidroxifenilpiruvato Dioxigenase/antagonistas & inibidores , Adolescente , Adulto , Idade de Início , Idoso , Idoso de 80 Anos ou mais , Alcaptonúria/classificação , Alcaptonúria/complicações , Alcaptonúria/genética , Alcaptonúria/metabolismo , Criança , Pré-Escolar , Doenças do Tecido Conjuntivo/etiologia , Cicloexanonas/uso terapêutico , Análise Mutacional de DNA , Progressão da Doença , Inibidores Enzimáticos/uso terapêutico , Feminino , Doenças das Valvas Cardíacas/etiologia , Homogentisato 1,2-Dioxigenase , Humanos , Artropatias/etiologia , Cálculos Renais/etiologia , Tábuas de Vida , Modelos Lineares , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Mutação , Nitrobenzoatos/uso terapêutico , Índice de Gravidade de Doença , Tirosina/metabolismo
14.
Medicine (Baltimore) ; 84(3): 137-146, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15879904

RESUMO

Nephropathic cystinosis is a rare, autosomal recessive lysosomal storage disorder caused by mutations in the CTNS gene that codes for a cystine transporter in the lysosomal membrane. Affected patients store 50-100 times the normal amounts of cystine in their cells, and suffer renal tubular and glomerular disease, growth retardation, photophobia, and other systemic complications, including a myopathy and swallowing dysfunction. Using videofluoroscopy and ultrasound examinations, we assessed the swallowing function of 101 patients with nephropathic cystinosis on their most recent admission to the National Institutes of Health Clinical Center between 1987 and 2004. These patients ranged in age from 6 to 45 years; more than half had significant complaints of swallowing difficulty. On examination of barium swallow, the oral, pharyngeal, and esophageal phases of swallowing were abnormal in 24%, 51%, and 73% of patients, respectively. The frequency of dysfunction increased with age for each phase of swallowing. Both the Swallowing Severity Score (a measure of dysfunction on barium swallow) and the Oral Muscle Composite Score (a reflection of vocal strength, oral-facial movement, and tongue and lip function) increased (that is, worsened) with the number of years that a patient was not receiving treatment with cysteamine, the cystine-depleting agent of choice in cystinosis. The severity scores decreased with the number of years on cysteamine therapy. The Swallowing Severity Score varied directly with the severity of muscle disease, but was not correlated with the presence or absence of the 57-kb CTNS deletion that commonly occurs in nephropathic cystinosis patients. We conclude that swallowing dysfunction in cystinosis presents a risk of fatal aspiration, correlates with the presence of muscle atrophy, and, based on cross-sectional data, increases in frequency with age and number of years without cysteamine treatment. Cystine-depleting therapy with cysteamine should be considered the treatment of choice for both pre- and posttransplant cystinosis patients.


Assuntos
Cisteamina/uso terapêutico , Cistinose/complicações , Transtornos de Deglutição/etiologia , Adolescente , Adulto , Fatores Etários , Sistemas de Transporte de Aminoácidos Neutros , Criança , Cistinose/tratamento farmacológico , Cistinose/fisiopatologia , Transtornos de Deglutição/prevenção & controle , Feminino , Glicoproteínas/genética , Humanos , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana Transportadoras , Pessoa de Meia-Idade , Mutação , Índice de Gravidade de Doença
15.
Metabolism ; 54(6): 719-28, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15931605

RESUMO

Alkaptonuria, a rare autosomal recessive disorder caused by mutations in the HGD gene and deficiency of homogentisate 1,2 dioxygenase, is characterized by ochronosis, arthritis, and daily excretion of gram quantities of homogentisic acid (HGA). Nitisinone, an inhibitor of the enzyme 4-hydroxyphenylpyruvate dioxygenase, can drastically reduce urinary excretion of HGA in individuals with alkaptonuria. We investigated the safety and the HGA-depleting efficacy of nitisinone in an open-label, single-center study of 9 alkaptonuria patients (5 women, 4 men; 35-69 years of age) over the course of 3 to 4 months. Each patient received nitisinone in incremental doses, 0.35 mg bid followed by 1.05 mg bid, and remained on this dosage and a regular diet for 3 months. Nitisinone reduced urinary HGA levels from an average of 4.0 +/- 1.8 (SD) g/day to 0.2 +/- 0.2 g/day ( P < .001). The average plasma tyrosine concentration, initially 68 +/- 18 mmicro mol/L, rose to 760 +/- 181 micro mol/L ( P < .001). During the final week of the study, 5 patients adhered to a protein-restricted diet (40 g/day), and their mean plasma tyrosine level fell from 755 +/- 167 to 603 +/- 114 mu mol/L. Six of the 7 patients who received nitisinone for more than 1 week reported decreased pain in their affected joints. Weekly ophthalmologic examinations showed no signs of corneal toxicity. Adverse events included the passing of kidney stones, the recognition of symptoms related to aortic stenosis, and elevation of liver transaminase levels. We conclude that low-dose nitisinone effectively reduced urinary HGA levels in patients with alkaptonuria. Future long-term clinical trials are planned to determine the benefits of nitisinone in preventing joint deterioration and providing pain relief, and its long-term side effects.


Assuntos
Alcaptonúria/tratamento farmacológico , Cicloexanonas/uso terapêutico , Nitrobenzoatos/uso terapêutico , Adulto , Idoso , Alcaptonúria/metabolismo , Cicloexanonas/efeitos adversos , Cicloexanonas/sangue , Proteínas Alimentares/administração & dosagem , Feminino , Ácido Homogentísico/sangue , Ácido Homogentísico/urina , Humanos , Masculino , Pessoa de Meia-Idade , Nitrobenzoatos/efeitos adversos , Nitrobenzoatos/sangue , Tirosina/sangue
16.
Mayo Clin Proc ; 79(3): 410-2, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15008615

RESUMO

We describe a 49-year-old woman in whom ocular cystinosis was diagnosed on the basis of a routine eye examination 12 years previously. Conjunctival biopsy was reported to support the diagnosis. The patient described photophobia for the past 5 years and reported a 2-fold increase in her serum IgG level for the past 12 years. On ophthalmic examination, corneal crystals were evident in the epithelium and superficial stromal layers, rather than throughout the corneal epithelium and the entire stroma as in ocular cystinosis. The patient's serum protein level was elevated at 8.7 g/dL; protein electrophoresis showed an elevated gamma-globulin peak, and the IgG level was twice that of normal at 2820 mg/dL. Bone marrow biopsy confirmed the diagnosis of multiple myeloma. This case illustrates that multiple myeloma can mimic corneal findings of cystinosis.


Assuntos
Doenças da Córnea/diagnóstico , Cistinose/diagnóstico , Mieloma Múltiplo/diagnóstico , Doenças da Córnea/complicações , Cistinose/complicações , Diagnóstico Diferencial , Feminino , Humanos , Imunoglobulina G/sangue , Pessoa de Meia-Idade , Fotofobia/etiologia
17.
Gene ; 499(2): 323-5, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22450360

RESUMO

Cystinosis is an autosomal recessive disorder characterized by defective transport of cystine across the lysosomal membrane and resulting in renal, ophthalmic, and other organ abnormalities. Mutations in the CTNS gene cause a deficiency of the transport protein, cystinosin. We performed mutation analysis of CTNS in six cystinosis patients from four families in Thailand. Using PCR sequencing of the entire coding regions, we identified all eight mutant alleles, including two mutations, p.G309D and p.Q284X, that have not been previously reported. This study expands the mutational and population spectrum of nephropathic cystinosis.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/genética , Cistinose/genética , Mutação de Sentido Incorreto , Mutação Puntual , Adolescente , Criança , Pré-Escolar , Análise Mutacional de DNA , Humanos , Lactente , Tailândia
18.
J Clin Invest ; 121(10): 3914-23, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21968110

RESUMO

Mutation of the tyrosinase gene (TYR) causes oculocutaneous albinism, type 1 (OCA1), a condition characterized by reduced skin and eye melanin pigmentation and by vision loss. The retinal pigment epithelium influences postnatal visual development. Therefore, increasing ocular pigmentation in patients with OCA1 might enhance visual function. There are 2 forms of OCA1, OCA-1A and OCA-1B. Individuals with the former lack functional tyrosinase and therefore lack melanin, while individuals with the latter produce some melanin. We hypothesized that increasing plasma tyrosine concentrations using nitisinone, an FDA-approved inhibitor of tyrosine degradation, could stabilize tyrosinase and improve pigmentation in individuals with OCA1. Here, we tested this hypothesis in mice homozygous for either the Tyrc-2J null allele or the Tyrc-h allele, which model OCA-1A and OCA-1B, respectively. Only nitisinone-treated Tyrc-h/c-h mice manifested increased pigmentation in their fur and irides and had more pigmented melanosomes. High levels of tyrosine improved the stability and enzymatic function of the Tyrc-h protein and also increased overall melanin levels in melanocytes from a human with OCA-1B. These results suggest that the use of nitisinone in OCA-1B patients could improve their pigmentation and potentially ameliorate vision loss.


Assuntos
Albinismo Oculocutâneo/tratamento farmacológico , Cicloexanonas/uso terapêutico , Cor de Olho/efeitos dos fármacos , Nitrobenzoatos/uso terapêutico , Pigmentação da Pele/efeitos dos fármacos , Albinismo Oculocutâneo/genética , Albinismo Oculocutâneo/fisiopatologia , Animais , Modelos Animais de Doenças , Inibidores Enzimáticos/uso terapêutico , Estabilidade Enzimática/efeitos dos fármacos , Estabilidade Enzimática/genética , Cor de Olho/genética , Cor de Olho/fisiologia , Feminino , Humanos , Melaninas/metabolismo , Melanócitos/efeitos dos fármacos , Melanócitos/metabolismo , Melanócitos/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Microscopia Eletrônica de Transmissão , Modelos Moleculares , Monofenol Mono-Oxigenase/química , Monofenol Mono-Oxigenase/deficiência , Monofenol Mono-Oxigenase/genética , Mutação , Gravidez , Pigmentação da Pele/genética , Pigmentação da Pele/fisiologia , Tirosina/metabolismo
19.
Clin J Am Soc Nephrol ; 5(6): 972-84, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20413436

RESUMO

BACKGROUND AND OBJECTIVES: Renal function and imaging findings have not been comprehensively and prospectively characterized in a broad age range of patients with molecularly confirmed autosomal recessive polycystic kidney disease (ARPKD). DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Ninety potential ARPKD patients were examined at the National Institutes of Health Clinical Center. Seventy-three fulfilled clinical diagnostic criteria, had at least one PKHD1 mutation, and were prospectively evaluated using magnetic resonance imaging (MRI), high-resolution ultrasonography (HR-USG), and measures of glomerular and tubular function. RESULTS: Among 31 perinatally symptomatic patients, 25% required renal replacement therapy by age 11 years; among 42 patients who became symptomatic beyond 1 month (nonperinatal), 25% required kidney transplantation by age 32 years. Creatinine clearance (CrCl) for nonperinatal patients (103 +/- 54 ml/min/1.73 m(2)) was greater than for perinatal patients (62 +/- 33) (P = 0.002). Corticomedullary involvement on HR-USG was associated with a significantly worse mean CrCl (61 +/- 32) in comparison with medullary involvement only (131 +/- 46) (P < 0.0001). Among children with enlarged kidneys, volume correlated inversely with function, although with wide variability. Severity of PKHD1 mutations did not determine kidney size or function. In 35% of patients with medullary-only abnormalities, standard ultrasound was normal and the pathology was detectable with HR-USG. CONCLUSIONS: In ARPKD, perinatal presentation and corticomedullary involvement are associated with faster progression of kidney disease. Mild ARPKD is best detected by HR-USG. Considerable variability occurs that is not explained by the type of PKHD1 mutation.


Assuntos
Genes Recessivos , Rim/patologia , Rim/fisiopatologia , Doenças Renais Policísticas/genética , Receptores de Superfície Celular/genética , Adolescente , Adulto , Biomarcadores/sangue , Biomarcadores/urina , Criança , Pré-Escolar , Creatinina/urina , Cistatina C/sangue , Análise Mutacional de DNA , Progressão da Doença , Feminino , Predisposição Genética para Doença , Taxa de Filtração Glomerular , Humanos , Lactente , Estimativa de Kaplan-Meier , Rim/diagnóstico por imagem , Túbulos Renais/patologia , Túbulos Renais/fisiopatologia , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , National Institutes of Health (U.S.) , Tamanho do Órgão , Linhagem , Fenótipo , Doenças Renais Policísticas/patologia , Doenças Renais Policísticas/fisiopatologia , Doenças Renais Policísticas/terapia , Estudos Prospectivos , Medição de Risco , Fatores de Risco , Índice de Gravidade de Doença , Fatores de Tempo , Ultrassonografia , Estados Unidos , Adulto Jovem
20.
Clin J Am Soc Nephrol ; 1(3): 555-62, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-17699259

RESUMO

Cystinosis, an autosomal recessive disorder of lysosomal cystine accumulation, results from mutations in the CTNS gene that encodes the lysosomal cystine transporter, cystinosin. Renal tubular Fanconi syndrome occurs in infancy, followed by rickets, growth retardation, photophobia, and renal failure, which requires renal transplantation at approximately 10 yr of age. Treatment with cysteamine decreases cellular cystine levels, retards renal deterioration, and allows for normal growth. Patients with a history of inadequate cystine depletion therapy may survive, after renal transplantation, into the third to fifth decades but will experience other, extrarenal complications of the disease. Routine chest and head computed tomography scans of 41 posttransplantation patients with cystinosis were reviewed for vascular calcification. The radiologic procedures had been performed to examine lung and brain parenchyma, so there was little ascertainment bias. Thirteen of the 41 patients had vascular calcification, including 11 with coronary artery calcification. One 25-yr-old man required three-vessel coronary artery bypass graft surgery. There were no significant differences between the 13 patients with calcification and the 28 without calcification in the following parameters: Time on dialysis, frequency of transplantation, hypertension, hypercholesterolemia, homozygosity for the 57-kb deletion in CTNS, serum creatinine, and calcium-phosphate product. However, the finding of vascular calcification correlated directly with duration of life without cysteamine therapy and inversely with duration of life under good cystine-depleting therapy. The accumulation of intracellular cystine itself maybe a risk factor for vascular calcifications, and older patients with cystinosis should be screened for this complication.


Assuntos
Calcinose/diagnóstico por imagem , Calcinose/etiologia , Doença da Artéria Coronariana/diagnóstico por imagem , Doença da Artéria Coronariana/etiologia , Cistinose/etiologia , Transplante de Rim/efeitos adversos , Tomografia Computadorizada por Raios X , Adolescente , Adulto , Criança , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa