Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Curr Opin Urol ; 30(2): 171-176, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31895888

RESUMEN

PURPOSE OF REVIEW: The review of potential therapies in the treatment of hyperoxaluria is timely, given the current excitement with clinical trials and the mounting evidence of the importance of oxalate in both kidney stone and chronic kidney disease. RECENT FINDINGS: Given the significant contribution of both endogenous and dietary oxalate to urinary oxalate excretions, it is not surprising therapeutic targets are being studied in both pathways. This article covers the existing data on endogenous and dietary oxalate and the current targets in these pathways. SUMMARY: In the near future, there will likely be therapies targeting both endogenous and dietary oxalate, especially in subsets of kidney stone formers.


Asunto(s)
Hiperoxaluria/metabolismo , Hiperoxaluria/terapia , Oxalatos/efectos adversos , Oxalatos/metabolismo , Adulto , Animales , Dieta/efectos adversos , Humanos , Hiperoxaluria/etiología , Cálculos Renales/química , Cálculos Renales/etiología , Cálculos Renales/metabolismo , Cálculos Renales/terapia , Ratones , Ratas , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/terapia
3.
J Am Soc Nephrol ; 29(6): 1615-1623, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29588429

RESUMEN

Background Endogenous oxalate synthesis contributes to calcium oxalate stone disease and is markedly increased in the inherited primary hyperoxaluria (PH) disorders. The incomplete knowledge regarding oxalate synthesis complicates discovery of new treatments. Hydroxyproline (Hyp) metabolism results in the formation of oxalate and glycolate. However, the relative contribution of Hyp metabolism to endogenous oxalate and glycolate synthesis is not known.Methods To define this contribution, we performed primed, continuous, intravenous infusions of the stable isotope [15N,13C5]-Hyp in nine healthy subjects and 19 individuals with PH and quantified the levels of urinary 13C2-oxalate and 13C2-glycolate formed using ion chromatography coupled to mass detection.Results The total urinary oxalate-to-creatinine ratio during the infusion was 73.1, 70.8, 47.0, and 10.6 mg oxalate/g creatinine in subjects with PH1, PH2, and PH3 and controls, respectively. Hyp metabolism accounted for 12.8, 32.9, and 14.8 mg oxalate/g creatinine in subjects with PH1, PH2, and PH3, respectively, compared with 1.6 mg oxalate/g creatinine in controls. The contribution of Hyp to urinary oxalate was 15% in controls and 18%, 47%, and 33% in subjects with PH1, PH2, and PH3, respectively. The contribution of Hyp to urinary glycolate was 57% in controls, 30% in subjects with PH1, and <13% in subjects with PH2 or PH3.Conclusions Hyp metabolism differs among PH types and is a major source of oxalate synthesis in individuals with PH2 and PH3. In patients with PH1, who have the highest urinary excretion of oxalate, the major sources of oxalate remain to be identified.


Asunto(s)
Glicolatos/orina , Hidroxiprolina/metabolismo , Hiperoxaluria Primaria/metabolismo , Ácido Oxálico/orina , Adulto , Creatinina/orina , Femenino , Humanos , Hiperoxaluria Primaria/orina , Masculino , Persona de Mediana Edad , Adulto Joven
4.
Biochim Biophys Acta ; 1862(6): 1055-62, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26854734

RESUMEN

The hereditary kidney stone disease primary hyperoxaluria type 1 (PH1) is caused by a functional deficiency of the liver-specific, peroxisomal, pyridoxal-phosphate-dependent enzyme, alanine:glyoxylate aminotransferase (AGT). One third of PH1 patients, particularly those expressing the p.[(Pro11Leu; Gly170Arg; Ile340Met)] mutant allele, respond clinically to pharmacological doses of pyridoxine. To gain further insight into the metabolic effects of AGT dysfunction in PH1 and the effect of pyridoxine, we established an "indirect" glycolate cytotoxicity assay using CHO cells expressing glycolate oxidase (GO) and various normal and mutant forms of AGT. In cells expressing GO the great majority of glycolate was converted to oxalate and glyoxylate, with the latter causing the greater decrease in cell survival. Co-expression of normal AGTs and some, but not all, mutant AGT variants partially counteracted this cytotoxicity and led to decreased synthesis of oxalate and glyoxylate. Increasing the extracellular pyridoxine up to 0.3µM led to an increased metabolic effectiveness of normal AGTs and the AGT-Gly170Arg variant. The increased survival seen with AGT-Gly170Arg was paralleled by a 40% decrease in oxalate and glyoxylate levels. These data support the suggestion that the effectiveness of pharmacological doses of pyridoxine results from an improved metabolic effectiveness of AGT; that is the increased rate of transamination of glyoxylate to glycine. The indirect glycolate toxicity assay used in the present study has potential to be used in cell-based drug screening protocols to identify chemotherapeutics that might enhance or decrease the activity and metabolic effectiveness of AGT and GO, respectively, and be useful in the treatment of PH1.


Asunto(s)
Hiperoxaluria Primaria/metabolismo , Oxalatos/metabolismo , Piridoxina/metabolismo , Transaminasas/metabolismo , Animales , Células CHO , Supervivencia Celular , Cricetulus , Glicolatos/metabolismo , Humanos , Hiperoxaluria Primaria/genética , Mutación , Especies Reactivas de Oxígeno/metabolismo , Transaminasas/genética
5.
Biochim Biophys Acta ; 1862(2): 233-9, 2016 02.
Artículo en Inglés | MEDLINE | ID: mdl-26655602

RESUMEN

Excessive endogenous oxalate synthesis can result in calcium oxalate kidney stone formation and renal failure. Hydroxyproline catabolism in the liver and kidney contributes to endogenous oxalate production in mammals. To quantify this contribution we have infused Wt mice, Agxt KO mice deficient in liver alanine:glyoxylate aminotransferase, and Grhpr KO mice deficient in glyoxylate reductase, with (13)C5-hydroxyproline. The contribution of hydroxyproline metabolism to urinary oxalate excretion in Wt mice was 22±2%, 42±8% in Agxt KO mice, and 36%±9% in Grhpr KO mice. To determine if blocking steps in hydroxyproline and glycolate metabolism would decrease urinary oxalate excretion, mice were injected with siRNA targeting the liver enzymes glycolate oxidase and hydroxyproline dehydrogenase. These siRNAs decreased the expression of both enzymes and reduced urinary oxalate excretion in Agxt KO mice, when compared to mice infused with a luciferase control preparation. These results suggest that siRNA approaches could be useful for decreasing the oxalate burden on the kidney in individuals with Primary Hyperoxaluria.


Asunto(s)
Oxidorreductasas de Alcohol/genética , Hidroxiprolina/metabolismo , Hiperoxaluria Primaria/genética , Hiperoxaluria Primaria/terapia , Prolina Oxidasa/metabolismo , Tratamiento con ARN de Interferencia , Oxidorreductasas de Alcohol/metabolismo , Animales , Modelos Animales de Enfermedad , Hiperoxaluria Primaria/metabolismo , Hígado/enzimología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Oxalatos/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico , Tratamiento con ARN de Interferencia/métodos
6.
Hum Mol Genet ; 24(19): 5500-11, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26199318

RESUMEN

Vitamin B6 in the form of pyridoxine (PN) is one of the most widespread pharmacological therapies for inherited diseases involving pyridoxal phosphate (PLP)-dependent enzymes, including primary hyperoxaluria type I (PH1). PH1 is caused by a deficiency of liver-peroxisomal alanine: glyoxylate aminotransferase (AGT), which allows glyoxylate oxidation to oxalate leading to the deposition of insoluble calcium oxalate in the kidney. Only a minority of PH1 patients, mostly bearing the F152I and G170R mutations, respond to PN, the only pharmacological treatment currently available. Moreover, excessive doses of PN reduce the specific activity of AGT in a PH1 cellular model. Nevertheless, the possible effect(s) of other B6 vitamers has not been investigated previously. Here, we compared the ability of PN in rescuing the effects of the F152I and G170R mutations with that of pyridoxamine (PM) and PL. We found that supplementation with PN raises the intracellular concentration of PN phosphate (PNP), which competes with PLP for apoenzyme binding leading to the formation of an inactive AGT-PNP complex. In contrast, PNP does not accumulate in the cell upon PM or PL supplementation, but higher levels of PLP and PM phosphate (PMP), the two active forms of the AGT coenzyme, are found. This leads to an increased ability of PM and PL to rescue the effects of the F152I and G170R mutations compared with PN. A similar effect was also observed for other folding-defective AGT variants. Thus, PM and PL should be investigated as matter of importance as therapeutics for PH1 patients bearing folding mutations.


Asunto(s)
Hiperoxaluria Primaria/genética , Piridoxal/farmacología , Piridoxamina/farmacología , Piridoxina/farmacología , Transaminasas/química , Complejo Vitamínico B/farmacología , Animales , Células CHO , Cricetinae , Cricetulus , Humanos , Hiperoxaluria Primaria/tratamiento farmacológico , Mutación/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Transaminasas/genética
7.
Proc Natl Acad Sci U S A ; 111(40): 14406-11, 2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-25237136

RESUMEN

Primary hyperoxaluria 1 (PH1; Online Mendelian Inheritance in Man no. 259900), a typically lethal biochemical disorder, may be caused by the AGT(P11LG170R) allele in which the alanine:glyoxylate aminotransferase (AGT) enzyme is mistargeted from peroxisomes to mitochondria. AGT contains a C-terminal peroxisomal targeting sequence, but mutations generate an N-terminal mitochondrial targeting sequence that directs AGT from peroxisomes to mitochondria. Although AGT(P11LG170R) is functional, the enzyme must be in the peroxisome to detoxify glyoxylate by conversion to alanine; in disease, amassed glyoxylate in the peroxisome is transported to the cytosol and converted to oxalate by lactate dehydrogenase, leading to kidney failure. From a chemical genetic screen, we have identified small molecules that inhibit mitochondrial protein import. We tested whether one promising candidate, Food and Drug Administration (FDA)-approved dequalinium chloride (DECA), could restore proper peroxisomal trafficking of AGT(P11LG170R). Indeed, treatment with DECA inhibited AGT(P11LG170R) translocation into mitochondria and subsequently restored trafficking to peroxisomes. Previous studies have suggested that a mitochondrial uncoupler might work in a similar manner. Although the uncoupler carbonyl cyanide m-chlorophenyl hydrazone inhibited AGT(P11LG170R) import into mitochondria, AGT(P11LG170R) aggregated in the cytosol, and cells subsequently died. In a cellular model system that recapitulated oxalate accumulation, exposure to DECA reduced oxalate accumulation, similar to pyridoxine treatment that works in a small subset of PH1 patients. Moreover, treatment with both DECA and pyridoxine was additive in reducing oxalate levels. Thus, repurposing the FDA-approved DECA may be a pharmacologic strategy to treat PH1 patients with mutations in AGT because an additional 75 missense mutations in AGT may also result in mistrafficking.


Asunto(s)
Decualinio/farmacología , Hiperoxaluria Primaria/metabolismo , Transaminasas/metabolismo , Animales , Antiinfecciosos Locales/farmacología , Células CHO , Cricetinae , Cricetulus , Evaluación Preclínica de Medicamentos/métodos , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/embriología , Humanos , Hiperoxaluria Primaria/genética , Hiperoxaluria Primaria/prevención & control , Immunoblotting , Microscopía Fluorescente , Mitocondrias/metabolismo , Mutación , Oxalatos/metabolismo , Peroxisomas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Piridoxina/farmacología , Transaminasas/genética , Pez Cebra/embriología
8.
J Biol Chem ; 288(4): 2475-84, 2013 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-23229545

RESUMEN

The gene encoding the liver-specific peroxisomal enzyme alanine:glyoxylate aminotransferase (AGT, EC. 2.6.1.44) exists as two common polymorphic variants termed the "major" and "minor" alleles. The P11L amino acid replacement encoded by the minor allele creates a hidden N-terminal mitochondrial targeting sequence, the unmasking of which occurs in the hereditary calcium oxalate kidney stone disease primary hyperoxaluria type 1 (PH1). This unmasking is due to the additional presence of a common disease-specific G170R mutation, which is encoded by about one third of PH1 alleles. The P11L and G170R replacements interact synergistically to reroute AGT to the mitochondria where it cannot fulfill its metabolic role (i.e. glyoxylate detoxification) effectively. In the present study, we have reinvestigated the consequences of the interaction between P11L and G170R in stably transformed CHO cells and have studied for the first time whether a similar synergism exists between P11L and three other mutations that segregate with the minor allele (i.e. I244T, F152I, and G41R). Our investigations show that the latter three mutants are all able to unmask the cryptic P11L-generated mitochondrial targeting sequence and, as a result, all are mistargeted to the mitochondria. However, whereas the G170R, I244T, and F152I mutants are able to form dimers and are catalytically active, the G41R mutant aggregates and is inactive. These studies open up the possibility that all PH1 mutations, which segregate with the minor allele, might also lead to the peroxisome-to-mitochondrion mistargeting of AGT, a suggestion that has important implications for the development of treatment strategies for PH1.


Asunto(s)
Hiperoxaluria Primaria/genética , Mitocondrias/metabolismo , Mutación , Alanina/genética , Alelos , Animales , Células CHO , Línea Celular , Cricetinae , Cricetulus , Dimerización , Humanos , Cinética , Polimorfismo Genético , Pliegue de Proteína , Transaminasas/genética
9.
Biochim Biophys Acta ; 1832(10): 1776-83, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23597595

RESUMEN

Primary hyperoxaluria type 1 (PH1) is a rare hereditary calcium oxalate kidney stone disease caused by a deficiency of the liver-specific pyridoxal-phosphate-dependent peroxisomal enzyme alanine:glyoxylate aminotransferase (AGT). About one third of patients are responsive to pharmacological doses of pyridoxine (vitamin B6), but its mechanism of action is unknown. Using stably transformed Chinese Hamster Ovary (CHO) cells expressing various normal and mutant forms of AGT, we have shown that pyridoxine increases the net expression, catalytic activity and peroxisomal import of the most common mistargeted mutant form of AGT (i.e. Gly170Arg on the background of the polymorphic minor allele). These multiple effects explain for the first time the action of pyridoxine in the most common group of responsive patients. Partial effects of pyridoxine were also observed for two other common AGT mutants on the minor allele (i.e. Phe152Ile and Ile244Thr) but not for the minor allele mutant AGT containing a Gly41Arg replacement. These findings demonstrate that pyridoxine, which is metabolised to pyridoxal phosphate, the essential cofactor of AGT, achieves its effects both as a prosthetic group (increasing enzyme catalytic activity) and a chemical chaperone (increasing peroxisome targeting and net expression). This new understanding should aid the development of pharmacological treatments that attempt to enhance efficacy of pyridoxine in PH1, as well as encouraging a re-evaluation of the extent of pyridoxine responsiveness in PH1, as more patients than previously thought might benefit from such treatment.


Asunto(s)
Hiperoxaluria Primaria/tratamiento farmacológico , Piridoxina/uso terapéutico , Animales , Células CHO , Cricetinae , Cricetulus , Humanos
10.
Kidney Int ; 86(6): 1074-6, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25427081

RESUMEN

The renal outcome in patients with primary hyperoxaluria type 1 is partly determined by AGXT mutations, including but not limited to the p.Gly170Arg mutation. The study by Mandrile et al. reports on the largest cohort of patients genotyped yet, with long-term renal survival and medical treatment by pyridoxine. In addition to the common p.Gly170Arg mutation, three other mutations were shown to be potentially associated with slower evolution.


Asunto(s)
Hiperoxaluria Primaria/genética , Fallo Renal Crónico/etiología , Mutación , Transaminasas/genética , Femenino , Humanos , Masculino
11.
Biochem Biophys Rep ; 39: 101765, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39040543

RESUMEN

Primary Hyperoxaluria Type 3 (PH3) results from 4-hydroxy-2-oxoglutarate (HOG) aldolase (HOGA) deficiency, which causes an increase in endogenous oxalate synthesis leading to calcium oxalate kidney stone disease. The mechanisms underlying HOG metabolism and increased oxalate synthesis in PH3 are not well understood. We used a Hoga1 knock-out mouse model of PH3 to investigate two aspects of HOG metabolism: reduction to dihydroxyglutarate (DHG), a pathway that may limit oxalate synthesis in PH3, and metabolism to glyoxylate, which is a direct precursor to oxalate. The metabolism of HOG to DHG was highest in liver and kidney cortical tissue, enhanced in the cytosolic compartment of the liver, and preferred NADPH as a cofactor. In the absence of HOGA, HOG to glyoxylate aldolase activity was highest in liver mitoplasts, with no activity present in brain tissue lysates. These findings will assist in the identification of enzymes responsible for the metabolism of HOG to DHG and glyoxylate, which may lead to novel therapeutic approaches to limit oxalate synthesis in those afflicted with PH3.

12.
Nephrol Dial Transplant ; 27(5): 1729-36, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22547750

RESUMEN

Primary hyperoxaluria Type 1 is a rare autosomal recessive inborn error of glyoxylate metabolism, caused by a deficiency of the liver-specific enzyme alanine:glyoxylate aminotransferase. The disorder results in overproduction and excessive urinary excretion of oxalate, causing recurrent urolithiasis and nephrocalcinosis. As glomerular filtration rate declines due to progressive renal involvement, oxalate accumulates leading to systemic oxalosis. The diagnosis is based on clinical and sonographic findings, urine oxalate assessment, enzymology and/or DNA analysis. Early initiation of conservative treatment (high fluid intake, pyridoxine, inhibitors of calcium oxalate crystallization) aims at maintaining renal function. In chronic kidney disease Stages 4 and 5, the best outcomes to date were achieved with combined liver-kidney transplantation.


Asunto(s)
Pruebas Genéticas , Hiperoxaluria Primaria/diagnóstico , Hiperoxaluria Primaria/terapia , Mutación/genética , Transaminasas/genética , Fluidoterapia , Humanos , Hiperoxaluria Primaria/metabolismo , Riñón/diagnóstico por imagen , Trasplante de Riñón , Oxalatos/metabolismo , Citrato de Potasio/uso terapéutico , Ultrasonografía , Vitamina B 6/uso terapéutico
13.
Clin Kidney J ; 15(Suppl 1): i4-i8, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35592619

RESUMEN

Primary hyperoxaluria type 1 (PH1) is a rare genetic form of calcium oxalate kidney stone disease. It is caused by a deficiency in the liver-specific enzyme, alanine:glyoxylate aminotransferase (AGT), a pyridoxal-5'-phosphate (PLP)-dependent enzyme involved in the metabolism of glyoxylate. The excessive endogenous synthesis of oxalate that ensues leads to hyperoxaluria, and the crystallization of the poorly soluble calcium salt of oxalate is responsible for a severe kidney stone disease, which can progress to end-stage renal disease, systemic deposition of oxalate and death. Knowledge about metabolic precursors of glyoxylate and oxalate, molecular pathology of AGT and analytical methods for diagnosis and clinical assessment have allowed a better understanding of the mechanisms underlying PH1 and opened the door to new therapeutic strategies.

14.
Biochem Biophys Rep ; 28: 101138, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34584990

RESUMEN

OBJECTIVE: Hydroxyacylglutathione hydrolase (aka as GLO-2) is a component of the glyoxalase pathway involved in the detoxification of the reactive oxoaldehydes, glyoxal and methylglyoxal. These reactive metabolites have been linked to a variety of pathological conditions, including diabetes, cancer and heart disease and may be involved in the aging process. The objective of this study was to generate a mouse model deficient in GLO-2 to provide insight into the function of GLO-2 and to determine if it is potentially linked to endogenous oxalate synthesis which could influence urinary oxalate excretion. METHODS: A GLO-2 knock out mouse was generated using CRISPR/Cas 9 techniques. Tissue and 24-h urine samples were collected under baseline conditions from adult male and female animals for biochemical analyses, including chromatographic measurement of glycolate, oxalate, glyoxal, methylglyoxal, D-lactate, ascorbic acid and glutathione levels. RESULTS: The GLO-2 KO animals developed normally and there were no changes in 24-h urinary oxalate excretion, liver levels of methylglyoxal, glyoxal, ascorbic acid and glutathione, or plasma d-lactate levels. GLO-2 deficient males had lower plasma glycolate levels than wild type males while this relationship was not observed in females. CONCLUSIONS: The lack of a unique phenotype in a GLO-2 KO mouse model under baseline conditions is consistent with recent evidence, suggesting a functional glyoxalase pathway is not required for optimal health. A lower plasma glycolate in male GLO-2 KO animals suggests glyoxal production may be a significant contributor to circulating glycolate levels, but not to endogenous oxalate synthesis.

15.
Biochim Biophys Acta Mol Basis Dis ; 1867(1): 165981, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33002578

RESUMEN

The Primary Hyperoxalurias (PH) are rare disorders of metabolism leading to excessive endogenous synthesis of oxalate and recurring calcium oxalate kidney stones. Alanine glyoxylate aminotransferase (AGT), deficient in PH type 1, is a key enzyme in limiting glyoxylate oxidation to oxalate. The affinity of AGT for its co-substrate, alanine, is low suggesting that its metabolic activity could be sub-optimal in vivo. To test this hypothesis, we examined the effect of L-alanine supplementation on oxalate synthesis in cell culture and in mouse models of Primary Hyperoxaluria Type 1 (Agxt KO), Type 2 (Grhpr KO) and in wild-type mice. Our results demonstrated that increasing L-alanine in cells decreased synthesis of oxalate and increased viability of cells expressing GO and AGT when incubated with glycolate. In both wild type and Grhpr KO male and female mice, supplementation with 10% dietary L-alanine significantly decreased urinary oxalate excretion ~30% compared to baseline levels. This study demonstrates that increasing the availability of L-alanine can increase the metabolic efficiency of AGT and reduce oxalate synthesis.


Asunto(s)
Alanina/farmacología , Hiperoxaluria Primaria/metabolismo , Oxalatos/metabolismo , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Animales , Células CHO , Cricetulus , Hiperoxaluria Primaria/genética , Hiperoxaluria Primaria/patología , Ratones , Ratones Noqueados , Transaminasas/genética , Transaminasas/metabolismo
16.
Kidney Int ; 77(5): 443-9, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20016466

RESUMEN

We sought to ascertain the long-term outcome and genotype-phenotype correlations available for primary hyperoxaluria type 1 in a large retrospective cohort study. We examined the clinical history of 155 patients (129 families primarily from Western Europe, North Africa, or the Middle East) as well as the enzymatic or genetic diagnosis. The median age at first symptom was 4 years, and at diagnosis 7.7 years, at which time 43% had reached end-stage renal disease. Presentations included: (1) early nephrocalcinosis and infantile renal failure, (2) recurrent urolithiasis and progressive renal failure diagnosed during childhood, (3) late onset with occasional stone passage diagnosed in adulthood, (4) diagnosis occurring on post-transplantation recurrence, and (5) family screening. The cumulative patient survival was 95, 86, and 74% at ages 10, 30, and 50 years, respectively, with the cumulative renal survival of 81, 59, 41, and 10% at ages 10, 20, 30, and 50 years, respectively; 72 patients had undergone a total of 97 transplantations. Among the 136 patients with DNA analysis, the most common mutation was p.Gly170Arg (allelic frequency 21.5%), with a median age at end-stage renal disease of 47 years for homozygotes, 35 years for heterozygotes, and 21 years for other mutations. Our results underscore the severe prognosis of primary hyperoxaluria type 1 and the necessity for early diagnosis and treatment, as well as confirm a better prognosis of the p.Gly170Arg mutation.


Asunto(s)
Hiperoxaluria Primaria/genética , Transaminasas/genética , Sustitución de Aminoácidos , Arginina/metabolismo , Niño , Preescolar , Estudios de Cohortes , Genotipo , Heterocigoto , Homocigoto , Humanos , Hiperoxaluria Primaria/diagnóstico , Lactante , Fallo Renal Crónico/genética , Mutación , Fenotipo , Pronóstico , Estudios Retrospectivos
17.
Pediatr Nephrol ; 25(6): 1081-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20213134

RESUMEN

The deposition of calcium oxalate crystals in the kidney and bone is a hallmark of primary hyperoxaluria type 1 (PH1). We report here an evaluation of the bone status of 12 PH1 children based on bone biomarkers [parathyroid hormone, vitamin D, fibroblast growth factor 23 (FGF23)] and radiological assessments (skeletal age, three-dimensional high-resolution peripheral quantitative computed tomography, HR-pQCT) carried out within the framework of a cross-sectional single-center study. The controls consisted of healthy and children with chronic kidney disease already enrolled in local bone and mineral metabolism studies. The mean age (+ or - standard deviation) age of the patients was 99 (+ or - 63) months. Six children suffered from fracture. Bone maturation was accelerated in five patients, four of whom were <5 years. The combination of new imaging techniques and biomarkers highlighted new and unexplained features of PH1: advanced skeletal age in young PH1 patients, increased FGF23 levels and decreased total volumetric bone mineral density with bone microarchitecture alteration.


Asunto(s)
Biomarcadores/análisis , Huesos/metabolismo , Hiperoxaluria Primaria/metabolismo , Hiperoxaluria Primaria/patología , Tomografía Computarizada por Rayos X/métodos , Adolescente , Niño , Preescolar , Estudios Transversales , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/sangre , Humanos , Lactante , Masculino , Adulto Joven
18.
Pediatr Nephrol ; 25(5): 965-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20091056

RESUMEN

Tacrolimus is known to potentially lead to adverse events in recipients with diarrhoea and/or calcium channel blocker (CCB) co-administration. We report a renal transplant recipient who suffered from severe nephrotoxicity related to a toxic tacrolimus trough concentration in both conditions, diarrhoea and CCB co-administration, and with genotyped CYP3A system and P-glycoprotein (P-gp) polymorphisms. To our knowledge, this is the first case to be investigated for such polymorphisms. Clinicians should be reminded of the possibility of highly increased levels of tacrolimus in situations of diarrhoea and/or co-administration of CCBs. It also highlights the key role in tacrolimus pharmacokinetics of the CYP3A system and P-gp polymorphisms, and their influence in high-risk situations when enzyme activity is already affected by enterocyte damage due to diarrhoea and CCB competition.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Lesión Renal Aguda/inducido químicamente , Bloqueadores de los Canales de Calcio/efectos adversos , Sistema Enzimático del Citocromo P-450/genética , Diarrea/complicaciones , Inmunosupresores/efectos adversos , Trasplante de Riñón , Tacrolimus/efectos adversos , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Lesión Renal Aguda/enzimología , Lesión Renal Aguda/genética , Adolescente , Citocromo P-450 CYP3A , Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Monitoreo de Drogas , Predisposición Genética a la Enfermedad , Humanos , Inmunosupresores/farmacocinética , Masculino , Farmacogenética , Fenotipo , Polimorfismo de Nucleótido Simple , Factores de Riesgo , Tacrolimus/farmacocinética
19.
Curr Opin Organ Transplant ; 15(5): 590-3, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20733487

RESUMEN

PURPOSE OF REVIEW: Primary hyperoxaluria type 1, the most common form of primary hyperoxaluria, is an autosomal recessive disorder caused by a deficiency of the liver-specific enzyme alanine:glyoxylate aminotransferase. This results in increased synthesis and subsequent urinary excretion of the metabolic end-product oxalate and the deposition of insoluble calcium oxalate in the kidney and urinary tract. As glomerular filtration rate decreases due to progressive renal involvement, oxalate accumulates and results in systemic oxalosis. RECENT FINDINGS: Diagnosis is still often delayed. It is mainly established on the basis of clinical and sonographic findings, urinary oxalate ± glycolate assessment, and DNA analysis. SUMMARY: Following specific conservative measures, the ultimate management is based on organ transplantation. Correction of the enzyme defect by liver transplantation should be planned before systemic oxalosis develops to optimize outcomes and may be either simultaneous (immunological benefit) or sequential (biochemical benefit) liver-kidney transplantation depending on disease staging, facilities, and access to deceased or living donors. Allograft and patient survival currently approaches that of transplant patients with kidney transplantation alone and with other diseases requiring combined liver-kidney transplantation. In addition, this strategy has also provided significant improvement in both quality of life and statural growth.


Asunto(s)
Hiperoxaluria Primaria , Trasplante de Riñón , Trasplante de Hígado , Progresión de la Enfermedad , Humanos , Hiperoxaluria/diagnóstico , Hiperoxaluria/mortalidad , Hiperoxaluria/cirugía , Calidad de Vida , Transaminasas/deficiencia , Trasplante Homólogo , Resultado del Tratamiento
20.
Nutrients ; 13(1)2020 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-33379176

RESUMEN

Kidney stone disease is increasing in prevalence, and the most common stone composition is calcium oxalate. Dietary oxalate intake and endogenous production of oxalate are important in the pathophysiology of calcium oxalate stone disease. The impact of dietary oxalate intake on urinary oxalate excretion and kidney stone disease risk has been assessed through large cohort studies as well as smaller studies with dietary control. Net gastrointestinal oxalate absorption influences urinary oxalate excretion. Oxalate-degrading bacteria in the gut microbiome, especially Oxalobacter formigenes, may mitigate stone risk through reducing net oxalate absorption. Ascorbic acid (vitamin C) is the main dietary precursor for endogenous production of oxalate with several other compounds playing a lesser role. Renal handling of oxalate and, potentially, renal synthesis of oxalate may contribute to stone formation. In this review, we discuss dietary oxalate and precursors of oxalate, their pertinent physiology in humans, and what is known about their role in kidney stone disease.


Asunto(s)
Dieta , Oxalatos/metabolismo , Oxalatos/orina , Bacterias , Oxalato de Calcio/metabolismo , Oxalato de Calcio/orina , Microbioma Gastrointestinal/fisiología , Humanos , Riñón , Cálculos Renales/orina , Nefrolitiasis , Oxalobacter formigenes , Urolitiasis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA