Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Hum Genet ; 87(3): 392-9, 2010 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-20797690

RESUMO

Primary hyperoxaluria (PH) is an autosomal-recessive disorder of endogenous oxalate synthesis characterized by accumulation of calcium oxalate primarily in the kidney. Deficiencies of alanine-glyoxylate aminotransferase (AGT) or glyoxylate reductase (GRHPR) are the two known causes of the disease (PH I and II, respectively). To determine the etiology of an as yet uncharacterized type of PH, we selected a cohort of 15 non-PH I/PH II patients from eight unrelated families with calcium oxalate nephrolithiasis for high-density SNP microarray analysis. We determined that mutations in an uncharacterized gene, DHDPSL, on chromosome 10 cause a third type of PH (PH III). To overcome the difficulties in data analysis attributed to a state of compound heterozygosity, we developed a strategy of "heterozygosity mapping"-a search for long heterozygous patterns unique to all patients in a given family and overlapping between families, followed by reconstruction of haplotypes. This approach enabled us to determine an allelic fragment shared by all patients of Ashkenazi Jewish descent and bearing a 3 bp deletion in DHDPSL. Overall, six mutations were detected: four missense mutations, one in-frame deletion, and one splice-site mutation. Our assumption is that DHDPSL is the gene encoding 4-hydroxy-2-oxoglutarate aldolase, catalyzing the final step in the metabolic pathway of hydroxyproline.


Assuntos
Hiperoxalúria Primária/genética , Mutação/genética , Oxo-Ácido-Liases/genética , Proteínas/genética , Alelos , Sequência de Aminoácidos , Sequência de Bases , Criança , Pré-Escolar , Análise Mutacional de DNA , Família , Feminino , Humanos , Hidroxiprolina/metabolismo , Lactente , Recém-Nascido , Judeus/genética , Masculino , Redes e Vias Metabólicas , Dados de Sequência Molecular , Oxalatos/metabolismo , Oxo-Ácido-Liases/química , Linhagem , Proteínas/química
2.
Am J Med Genet A ; 158A(9): 2124-30, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22821680

RESUMO

Primary hyperoxaluria type 1 (PH1) is an autosomal recessive disorder caused by mutations in the alanine:glyoxylate aminotransferase (AGXT) gene, located on chromosome 2q37. Mutant AGXT leads to excess production and excretion of oxalate, resulting in accumulation of calcium oxalate in the kidney, and progressive loss of renal function. Brachydactyly mental retardation syndrome (BDMR) is an autosomal dominant disorder, caused by haploinsufficiency of histone deacetylase 4 (HDAC4), also on chromosome 2q37. It is characterized by skeletal abnormalities and developmental delay. Here, we report on a girl who had phenotypes of both PH1 and BDMR. PCR-sequencing of the coding regions of AGXT showed a novel missense mutation, c.32C>G (p.Pro11Arg) inherited from her mother. Functional analyses demonstrated that it reduced the enzymatic activity to 31% of the wild-type and redirected some percentage of the enzyme away from the peroxisome. Microsatellite and array-CGH analyses indicated that the proband had a paternal de novo telomeric deletion of chromosome 2q, which included HDAC4. To our knowledge, this is the first report of PH1 and BDMR, with a novel AGXT mutation and a de novo telomeric deletion of chromosome 2q.


Assuntos
Braquidactilia/genética , Deleção Cromossômica , Cromossomos Humanos Par 2 , Hiperoxalúria Primária/genética , Deficiência Intelectual/genética , Mutação , Transaminases/genética , Pré-Escolar , Feminino , Haplótipos , Humanos , Masculino , Microscopia de Fluorescência , Linhagem , Frações Subcelulares/enzimologia , Transaminases/metabolismo
3.
Circ J ; 74(11): 2403-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20921818

RESUMO

BACKGROUND: In patients with primary hyperoxaluria (PH), oxalate overproduction can result in recurrent urolithiasis and nephrocalcinosis, which in some cases results in a progressive decline in renal function, oxalate retention, and systemic oxalosis involving bone, retina, arterial media, peripheral nerves, skin, and heart. Oxalosis involving the myocardium or conduction system can potentially lead to heart failure and fatal arrhythmias. METHODS AND RESULTS: A retrospective review of our institution's database was conducted for all patients with a confirmed diagnosis of PH between 1/1948 and 1/2006 (n=103). Electrocardiogram (ECG) and echocardiography were used to identify cardiac abnormalities. Ninety-three patients fulfilled the inclusion criteria, 58% were male. Mean follow-up was 11.9 (median 8.8) years. In 38 patients who received an ECG or echocardiography, 31 were found to have any cardiac abnormalities. Cardiac findings correlated with decline in renal function. CONCLUSIONS: Our data suggests that physicians caring for patients with PH should pay close attention to cardiac status, especially if renal function is impaired.


Assuntos
Cardiopatias/etiologia , Hiperoxalúria Primária/complicações , Adolescente , Adulto , Idoso , Biópsia , Criança , Pré-Escolar , Ecocardiografia , Eletrocardiografia , Feminino , Testes Genéticos , Cardiopatias/diagnóstico , Humanos , Hiperoxalúria Primária/diagnóstico , Lactente , Recém-Nascido , Nefropatias/etiologia , Masculino , Pessoa de Meia-Idade , Minnesota , Estudos Retrospectivos , Fatores de Tempo , Adulto Jovem
4.
Hum Mutat ; 30(6): 910-7, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19479957

RESUMO

Primary hyperoxaluria type 1 (PH1) is an autosomal recessive, inherited disorder of glyoxylate metabolism arising from a deficiency of the alanine:glyoxylate aminotransferase (AGT) enzyme, encoded by the AGXT gene. The disease is manifested by excessive endogenous oxalate production, which leads to impaired renal function and associated morbidity. At least 146 mutations have now been described, 50 of which are newly reported here. The mutations, which occur along the length of the AGXT gene, are predominantly single-nucleotide substitutions (75%), 73 are missense, 19 nonsense, and 18 splice mutations; but 36 major and minor deletions and insertions are also included. There is little association of mutation with ethnicity, the most obvious exception being the p.Ile244Thr mutation, which appears to have North African/Spanish origins. A common, polymorphic variant encoding leucine at codon 11, the so-called minor allele, has significantly lower catalytic activity in vitro, and has a higher frequency in PH1 compared to the rest of the population. This polymorphism influences enzyme targeting in the presence of the most common Gly170Arg mutation and potentiates the effect of several other pathological sequence variants. This review discusses the spectrum of AGXT mutations and polymorphisms, their clinical significance, and their diagnostic relevance.


Assuntos
Hiperoxalúria Primária/enzimologia , Hiperoxalúria Primária/genética , Transaminases/genética , Alelos , Animais , Análise Mutacional de DNA , Humanos , Hiperoxalúria Primária/diagnóstico , Polimorfismo Genético , Transaminases/química , Dissomia Uniparental/genética
5.
Am J Kidney Dis ; 52(6): 1096-103, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18951670

RESUMO

BACKGROUND: Urinary oxalate is a major risk factor for calcium oxalate stones. Marked hyperoxaluria arises from mutations in 2 separate loci, AGXT and GRHPR, the causes of primary hyperoxaluria (PH) types 1 (PH1) and 2 (PH2), respectively. Studies of null Slc26a6(-/-) mice have shown a phenotype of hyperoxaluria, hyperoxalemia, and calcium oxalate urolithiasis, leading to the hypothesis that SLC26A6 mutations may cause or modify hyperoxaluria in humans. STUDY DESIGN: Cross-sectional case-control. SETTING & PARTICIPANTS: Cases were recruited from the International Primary Hyperoxaluria Registry. Control DNA samples were from a pool of adult subjects who identified themselves as being in good health. PREDICTOR: PH1, PH2, and non-PH1/PH2 genotypes in cases. OUTCOMES & MEASURES: Homozygosity or compound heterozygosity for SLC26A6 variants. Functional expression of oxalate transport in Xenopus laevis oocytes. RESULTS: 80 PH1, 6 PH2, 8 non-PH1/PH2, and 96 control samples were available for SLC26A6 screening. A rare variant, c.487C-->T (p.Pro163Ser), was detected solely in 1 non-PH1/PH2 pedigree, but this variant failed to segregate with hyperoxaluria, and functional studies of oxalate transport in Xenopus oocytes showed no transport defect. No other rare variant was identified specifically in non-PH1/PH2. Six additional missense variants were detected in controls and cases. Of these, c.616G-->A (p.Val206Met) was most common (11%) and showed a 30% reduction in oxalate transport. To test p.Val206Met as a potential modifier of hyperoxaluria, we extended screening to PH1 and PH2. Heterozygosity for this variant did not affect plasma or urine oxalate levels in this population. LIMITATIONS: We did not have a sufficient number of cases to determine whether homozygosity for p.Val206Met might significantly affect urine oxalate. CONCLUSIONS: SLC26A6 was effectively ruled out as the disease gene in this non-PH1/PH2 cohort. Taken together, our studies are the first to identify and characterize SLC26A6 variants in patients with hyperoxaluria. Phenotypic and functional analysis excluded a significant effect of identified variants on oxalate excretion.


Assuntos
Oxalato de Cálcio , Hipercalciúria/genética , Proteínas de Membrana Transportadoras/genética , Mutação , Nefrolitíase/genética , Adolescente , Adulto , Oxalato de Cálcio/metabolismo , Estudos de Casos e Controles , Pré-Escolar , Estudos Transversais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Nefrolitíase/metabolismo , Linhagem , Fenótipo , Transportadores de Sulfato , Adulto Jovem
6.
Nat Rev Nephrol ; 8(3): 151-62, 2011 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-22183508

RESUMO

Urolithiasis affects approximately 10% of individuals in Western societies by the seventh decade of life. The most common form, idiopathic calcium oxalate urolithiasis, results from the interaction of multiple genes and their interplay with dietary and environmental factors. To date, considerable progress has been made in identifying the metabolic risk factors that predispose to this complex trait, among which hypercalciuria predominates. The specific genetic and epigenetic factors involved in urolithiasis have remained less clear, partly owing to the candidate gene and linkage methods that have been available until now, being inherently low in their power of resolution and in assessing modest effects in complex traits. However, together with investigations of rare, Mendelian forms of urolithiasis associated with various metabolic risk factors, these methods have afforded insights into biological pathways that seem to underlie the development of stones in the urinary tract. Monogenic diseases account for a greater proportion of stone formers in children and adolescents than in adults. Early diagnosis of monogenic forms of urolithiasis is of importance owing to associated renal injury and other potentially treatable disease manifestations, but diagnosis is often delayed because of a lack of familiarity with these rare disorders. In this Review, we will discuss advances in the understanding of the genetics underlying polygenic and monogenic forms of urolithiasis.


Assuntos
Marcadores Genéticos , Predisposição Genética para Doença , Urolitíase/genética , Humanos , Prognóstico , Fatores de Risco , Urolitíase/epidemiologia
7.
Clin J Am Soc Nephrol ; 6(9): 2289-95, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21896830

RESUMO

BACKGROUND AND OBJECTIVES: Primary hyperoxaluria types I and II (PHI and PHII) are rare monogenic causes of hyperoxaluria and calcium oxalate urolithiasis. Recently, we described type III, due to mutations in HOGA1 (formerly DHDPSL), hypothesized to cause a gain of mitochondrial 4-hydroxy-2-oxoglutarate aldolase activity, resulting in excess oxalate. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: To further explore the pathophysiology of HOGA1, we screened additional non-PHI-PHII patients and performed reverse transcription PCR analysis. Postulating that HOGA1 may influence urine oxalate, we also screened 100 idiopathic calcium oxalate stone formers. RESULTS: Of 28 unrelated hyperoxaluric patients with marked hyperoxaluria not due to PHI, PHII, or any identifiable secondary cause, we identified 10 (36%) with two HOGA1 mutations (four novel, including a nonsense variant). Reverse transcription PCR of the stop codon and two common mutations showed stable expression. From the new and our previously described PHIII cohort, 25 patients were identified for study. Urine oxalate was lower and urine calcium and uric acid were higher when compared with PHI and PHII. After 7.2 years median follow-up, mean eGFR was 116 ml/min per 1.73 m(2). HOGA1 heterozygosity was found in two patients with mild hyperoxaluria and in three of 100 idiopathic calcium oxalate stone formers. No HOGA1 variants were detected in 166 controls. CONCLUSIONS: These findings, in the context of autosomal recessive inheritance for PHIII, support a loss-of-function mechanism for HOGA1, with potential for a dominant-negative effect. Detection of HOGA1 variants in idiopathic calcium oxalate urolithiasis also suggests HOGA1 may be a predisposing factor for this condition.


Assuntos
Oxalato de Cálcio/metabolismo , Predisposição Genética para Doença , Hiperoxalúria Primária/genética , Oxo-Ácido-Liases/genética , Urolitíase/etiologia , Humanos , Mutação , Oxo-Ácido-Liases/metabolismo , Fatores de Risco , Urolitíase/genética
8.
J Am Soc Nephrol ; 18(6): 1905-14, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17460142

RESUMO

Mutations in AGXT, a locus mapped to 2q37.3, cause deficiency of liver-specific alanine:glyoxylate aminotransferase (AGT), the metabolic error in type 1 primary hyperoxaluria (PH1). Genetic analysis of 55 unrelated probands with PH1 from the Mayo Clinic Hyperoxaluria Center, to date the largest with availability of complete sequencing across the entire AGXT coding region and documented hepatic AGT deficiency, suggests that a molecular diagnosis (identification of two disease alleles) is feasible in 96% of patients. Unique to this PH1 population was the higher frequency of G170R, the most common AGXT mutation, accounting for 37% of alleles, and detection of a new 3' end deletion (Ex 11_3'UTR del). A described frameshift mutation (c.33_34insC) occurred with the next highest frequency (11%), followed by F152I and G156R (frequencies of 6.3 and 4.5%, respectively), both surpassing the frequency (2.7%) of I244T, the previously reported third most common pathogenic change. These sequencing data indicate that AGXT is even more variable than formerly believed, with 28 new variants (21 mutations and seven polymorphisms) detected, with highest frequencies on exons 1, 4, and 7. When limited to these three exons, molecular analysis sensitivity was 77%, compared with 98% for whole-gene sequencing. These are the first data in support of comprehensive AGXT analysis for the diagnosis of PH1, obviating a liver biopsy in most well-characterized patients. Also reported here is previously unavailable evidence for the pathogenic basis of all AGXT missense variants, including evolutionary conservation data in a multisequence alignment and use of a normal control population.


Assuntos
Testes Genéticos/métodos , Hiperoxalúria Primária/diagnóstico , Hiperoxalúria Primária/genética , Transaminases/genética , Sequência de Aminoácidos , Sequência Conservada , Estudos de Viabilidade , Feminino , Mutação da Fase de Leitura , Genótipo , Humanos , Masculino , Dados de Sequência Molecular , Mutação de Sentido Incorreto
9.
Nephrol Dial Transplant ; 21(8): 2292-5, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16597637

RESUMO

BACKGROUND: Primary hyperoxaluria type 2 (PH2) is a rare monogenic disorder characterized by an elevated urinary excretion of oxalate. Increased oxalate excretion in PH2 patients can cause nephrolithiasis and nephrocalcinosis, and can, in some cases, result in renal failure and systemic oxalate deposition. The disease is due to a deficiency of glyoxylate reductase/hydroxypyruvate reductase (GRHPR) activity. A definitive diagnosis of PH2 is currently made by the analysis of GR activity in a liver biopsy. GRHPR is expressed in virtually every tissue in the body, suggesting that utilization of more readily available cells could be used to determine GRHPR deficiency. In this study, we have evaluated the potential of determining GR and d-glycerate dehydrogenase (DGDH) activity in blood mononuclear cells (BMC) as a diagnostic indicator of PH2. METHODS: Blood samples were obtained from 10 male and 10 female normal subjects, median age 31, range 21-63, at the Wake Forest University Medical Center and from primary hyperoxaluria patients at the Mayo Clinic. The BMC were isolated and GR and DGDH activities measured in cell lysates. RESULTS: An assay of 20 normal individuals indicated that BMC contained a DGDH and GR activity of 0.97+/-0.20 (range 0.62-1.45), and 10.6+/-3.3 (range 8.3-16.6) nmol/min/mg protein, respectively. The intra-assay coefficient of variation for DGDH and GR activity was 8.2 and 11.5%, respectively. The BMC lysates from normal adult subjects and patients with PH1 showed similar GR and DGDH activities. This was confirmed by the presence of immunoreactive GRHPR protein by western blot analysis. In contrast, PH2 BMC lysates did not exhibit DGDH or GR activity, and showed no immunoreactive GRHPR by western blot analysis. CONCLUSION: These results suggest that the assay of DGDH or GR activity in BMC could be used as a minimally invasive diagnostic test for PH2.


Assuntos
Oxirredutases do Álcool/sangue , Ensaios Enzimáticos Clínicos , Hiperoxalúria Primária/diagnóstico , Leucócitos Mononucleares/enzimologia , Espectrofotometria , Adulto , Oxirredutases do Álcool/deficiência , Oxirredutases do Álcool/genética , Western Blotting , Desidrogenases de Carboidrato/sangue , Feminino , Humanos , Hiperoxalúria Primária/classificação , Hiperoxalúria Primária/genética , Masculino , Pessoa de Meia-Idade , Valores de Referência , Reprodutibilidade dos Testes
10.
Am J Nephrol ; 25(2): 183-8, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15849466

RESUMO

BACKGROUND: Marked hyperoxaluria due to liver-specific deficiency of alanine:glyoxylate aminotransferase activity (AGT) characterizes type I primary hyperoxaluria (PHI). Approximately half of PHI patients experience improvement in the degree of hyperoxaluria following pyridoxine (VB6) treatment. Recently, we showed an association between VB6 response and the commonest PHI mutation G170R, with patients possessing one or two copies showing 50% reduction or complete to near complete normalization of oxaluria, respectively. Two patients showed responses varying from this pattern. To further clarify the molecular basis of VB6 response in PHI, we performed additional genotyping. METHODS: 23 PHI patients diagnosed via hepatic enzyme analysis, hyperoxaluria and hyperglycolic aciduria or homozygosity for a known mutation, availability of pre- and post-VB6 24-hour urine oxalate and GFR >40 ml/min/1.73 m2 were included. Data was retrieved retrospectively, oxalate measured by oxalate oxidase, and genotyping performed by PCR-based methods. RESULTS: VB6 response was associated with the G170R and F152I mutations. Eight new sequence changes were detected. CONCLUSIONS: In PHI, two mutations resulting in AGT mistargeting are associated with VB6 response. Whether this favorable effect is specific to the peroxisomal-to-mitochondrial mistargeting caused by these changes or due to another mechanism remains to be determined.


Assuntos
Enzimas/genética , Hiperoxalúria Primária/genética , Piridoxina/uso terapêutico , Vitaminas/uso terapêutico , Estudos de Coortes , Genótipo , Humanos , Hiperoxalúria Primária/tratamento farmacológico , Fenótipo , Estudos Retrospectivos
11.
Kidney Int ; 67(5): 1704-9, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15840016

RESUMO

BACKGROUND: Pyridoxine (VB6) response in type I primary hyperoxaluria (PHI) is variable, with nearly equal numbers of patients showing partial to complete reductions in oxaluria, and resistance. Because high urine oxalate concentrations cause stones and renal injury, reduction in urine oxalate excretion is deemed favorable. Mechanisms of VB6 action on hepatic alanine:glyoxylate aminotransferase (AGT), the deficient enzyme in PHI, and VB6 dose response have not been well-characterized. METHODS: Sequencing or restriction site-generating polymerase chain reaction (PCR) was used for c.508 genotyping in 23 PHI patients. Pre- and post-VB6 24-hour urine oxalate excretion and VB6 dose were ascertained by retrospective chart review. RESULTS: There were six c.508 G>A homozygotes (AA), eight heterozygotes (GA), and nine patients lacking this change (GG). Pre-VB6 urine oxalate excretion was 152 +/- 39, 203 +/- 68 and 206 +/- 74 mg/1.73 m(2)/24 hours, respectively, and did not differ [AA vs. GA (P= 0.07); AA vs. GG (P= 0.07); GA vs. GG, (P= 0.47)]. Post-VB6 urine oxalate excretion was normal in AA (pre- vs. post-VB6) (P < 0.001), partially reduced in GA (P < 0.001), and unchanged in GG (P= 0.06). Urine oxalate excretion attenuation was similar for VB6 doses (mg/kg/day) of 1 to 4.9, 5 to 9.9, and 10 to 14.9 in AA (P= 0.41, P= 0.28, and P= 0.11, respectively) and GA (P= 0.42, P= 0.39, and P= 0.30, respectively) during follow-up. CONCLUSION: Presence of the c.508 G>A allele confers VB6 response in PHI and VB6 doses of 5 mg/kg/day appear sufficient. c.508 genotyping can be used to predict VB6 response and guide treatment in PHI. [c represents cDNA sequence where nucleotide position +1 corresponds to the adenine (A) of the translation start codon ATG. Equivalent positions based on 5' UTR nucleotide numbering are as follows: c.508 G>A = G630A (Gly170Arg), c.32 C>T = C154T (Pro11Leu), and c.454 T>A = T576A (Phe152Ile)], yields highest residual AGT activity. To test whether VB6 response might be attributable to this allele, we performed c.508 genotyping.


Assuntos
Hiperoxalúria Primária/tratamento farmacológico , Hiperoxalúria Primária/genética , Piridoxina/uso terapêutico , Adolescente , Adulto , Alelos , Criança , Pré-Escolar , Relação Dose-Resposta a Droga , Feminino , Genótipo , Heterozigoto , Homozigoto , Humanos , Hiperoxalúria Primária/enzimologia , Masculino , Pessoa de Meia-Idade , Mutação , Piridoxina/administração & dosagem , Transaminases/genética
12.
Am J Nephrol ; 25(3): 290-6, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15961949

RESUMO

BACKGROUND/AIMS: Primary hyperoxaluria (PH) is an inherited disorder that causes calcium urolithiasis and renal failure. Due to its rarity, experience at most centers with this disease is limited. METHODS: A secure, web-based, institutional review board/ethics committee and American Health Insurance Portability and Accountability Act (HIPAA)-compliant registry was developed to facilitate international contributions to a data base. To date 95 PH patients have been entered. RESULTS: PH type was confirmed in 84/95 (PH1 79%, PH2 9%). Mean age +/- SD at symptom onset was 9.5 +/- 10.2 (median 5.5) years whereas age at diagnosis was 15.0 +/- 15.2 (median 10.0) years. Urolithiasis was present at diagnosis in 90% (mean 7, median 1, stones prior to diagnosis) and nephrocalcinosis in 48%. Surprisingly 15% of the patients were asymptomatic at the time of diagnosis. Nineteen of the 95 patients were first recognized to have PH after they had reached end-stage renal disease, with the diagnosis made only after kidney transplantation in 7 patients. Patients were followed for 12.1 +/- 10.6 (median 9.4) years. Thirty-four of 95 progressed to end-stage renal failure, before (19 patients) or after (15 patients) diagnosis. In the PH1 cohort actuarial renal survival was 64% at 30 years of age, 47% at 40 years, and 29% at 50 years. CONCLUSION: We have developed a PH registry, and demonstrated the feasibility of this secure, web-based approach for data entry. By facilitating accumulation of an increasing cohort of patients, this registry should allow more complete characterization of clinical expression of PH, an appreciation of geographic variability, and identification of treatment outcomes.


Assuntos
Hiperoxalúria Primária , Internacionalidade , Sistema de Registros , Adolescente , Adulto , Criança , Pré-Escolar , Estudos de Coortes , Progressão da Doença , Estudos de Viabilidade , Humanos , Hiperoxalúria Primária/complicações , Hiperoxalúria Primária/diagnóstico , Hiperoxalúria Primária/fisiopatologia , Lactente , Rim/fisiopatologia , Falência Renal Crônica/etiologia , Falência Renal Crônica/cirurgia , Transplante de Rim , Estudos Retrospectivos
13.
Kidney Int ; 62(2): 392-400, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12110000

RESUMO

BACKGROUND: Hyperoxaluria may be idiopathic, secondary, or due to primary hyperoxaluria (PH). Hepatic alanine:glyoxylate aminotransferase (AGT) or glyoxylate/hydroxypyruvate reductase (GR/HPR) deficiency causes PHI or PHII, respectively. Hepatic glycolate oxidase (GO) is a candidate enzyme for a third form of inherited hyperoxaluria. METHODS: Six children were identified with marked hyperoxaluria, urolithiasis, and normal hepatic AGT (N = 5) and GR/HPR (N = 4). HPR was below normal and GR not measured in one. Of an affected sibling pair, only one underwent biopsy. GO mutation screening was performed, and dietary oxalate (Diet(ox)), enteric oxalate absorption (EOA) measured using [13C2] oxalate, renal clearance (GFR), fractional oxalate excretion (FE(ox)) in the children, and urine oxalate in first-degree relatives (FDR) to understand the etiology of the hyperoxaluria. RESULTS: Mean presenting age was 19.2 months and urine oxalate 1.3 +/- 0.5 mmol/1.73 m2/24 h (mean +/- SD). Two GO sequence changes (T754C, IVS3 - 49 C>G) were detected which were not linked to the hyperoxaluria. Diet(ox) was 42 +/- 31 mg/day. EOA was 9.4 +/- 3.6%, compared with 7.6 +/- 1.2% in age-matched controls (P = 0.33). GFR was 90 +/- 19 mL/min/1.73 m2 and FE(ox) 4.2 +/- 1.4. Aside from the two brothers, hyperoxaluria was not found in FDR. CONCLUSIONS: These patients illustrate a novel form of hyperoxaluria and urolithiasis, without excess Diet(ox), enteric hyper-absorption, or hepatic AGT, GR/HPR deficiency. Alterations in pathways of oxalate synthesis, in liver or kidney, or in renal tubular oxalate handling are possible explanations. The affected sibling pair suggests an inherited basis.


Assuntos
Hiperoxalúria/genética , Hiperoxalúria/metabolismo , Oxirredutases do Álcool/genética , Cálcio da Dieta/urina , Pré-Escolar , Saúde da Família , Feminino , Glioxilatos/metabolismo , Humanos , Hiperoxalúria Primária , Lactente , Masculino , Dados de Sequência Molecular , Oxalatos/urina , Linhagem , Fosfatos/urina , Cálculos Urinários/genética , Cálculos Urinários/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa