Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Pediatr Nephrol ; 37(8): 1905-1914, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34999988

RESUMEN

BACKGROUND: Post-transplantation immunosuppressive therapy reduces the risk of graft rejection but raises the risk of infection and malignancy. A biomarker of the level of immunosuppression can be helpful in monitoring immunosuppressive therapy. Inverse correlation between Torque teno virus (TTV) from the Anelloviridae (AV) family load and immune competence was described in previous studies. The aim of this study was to analyze the association between AV family viruses' kinetics and the risk for graft rejection in the first year after kidney transplantation in children. METHODS: The titers of three genera (TTV, TTMDV, and TTMV) from the AV family were monitored by real-time PCR in consecutive samples from children before and after kidney transplantation. RESULTS: Twenty-one children who underwent kidney transplantation were enrolled. Five out of 21 patients experienced acute graft rejection within a year from transplantation. We found that in patients who experienced graft rejection, the median titers of TTV and total AV titers at 5-6 months post-transplantation were lower than in those who did not. Using a threshold determined by ROC analysis, significant differences in TTV and total AV load were found between patients who had or did not have graft rejection (p = 0.002 and 0.004, respectively). No association was found between the dominance of any AV genus titer and the likelihood of rejection. CONCLUSION: This pilot study suggests that children after kidney transplantation with low TTV and total AV titers 5-6 months post-transplantation are at increased risk for graft rejection within a year after transplantation. A higher resolution version of the Graphical abstract is available as Supplementary information.


Asunto(s)
Anelloviridae , Trasplante de Riñón , Torque teno virus , Niño , ADN Viral , Rechazo de Injerto , Humanos , Trasplante de Riñón/efectos adversos , Proyectos Piloto , Complicaciones Posoperatorias/diagnóstico , Complicaciones Posoperatorias/etiología , Torque teno virus/genética , Carga Viral
2.
Int J Mol Sci ; 23(2)2022 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-35055190

RESUMEN

Hydroxyproline is one of the most prevalent amino acids in animal proteins. It is not a genetically encoded amino acid, but, rather, it is produced by the post-translational modification of proline in collagen, and a few other proteins, by prolyl hydroxylase enzymes. Although this post-translational modification occurs in a limited number of proteins, its biological significance cannot be overestimated. Considering that hydroxyproline cannot be re-incorporated into pro-collagen during translation, it should be catabolized following protein degradation. A cascade of reactions leads to production of two deleterious intermediates: glyoxylate and hydrogen peroxide, which need to be immediately converted. As a result, the enzymes involved in hydroxyproline catabolism are located in specific compartments: mitochondria and peroxisomes. The particular distribution of catabolic enzymes in these compartments, in different species, depends on their dietary habits. Disturbances in hydroxyproline catabolism, due to genetic aberrations, may lead to a severe disease (primary hyperoxaluria), which often impairs kidney function. The basis of this condition is accumulation of glyoxylate and its conversion to oxalate. Since calcium oxalate is insoluble, children with this rare inherited disorder suffer from progressive kidney damage. This condition has been nearly incurable until recently, as significant advances in substrate reduction therapy using small interference RNA led to a breakthrough in primary hyperoxaluria type 1 treatment.


Asunto(s)
Hidroxiprolina/metabolismo , Hiperoxaluria Primaria/genética , ARN Interferente Pequeño/farmacología , Animales , Evolución Molecular , Predisposición Genética a la Enfermedad , Glioxilatos/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Hiperoxaluria Primaria/tratamiento farmacológico , Hiperoxaluria Primaria/metabolismo , Mitocondrias/metabolismo , Peroxisomas/metabolismo , ARN Interferente Pequeño/uso terapéutico
3.
J Urol ; 205(5): 1394-1399, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33350326

RESUMEN

PURPOSE: The etiology of calcium-oxalate kidney stone formation remains elusive. Biallelic mutations in HOGA1 are responsible for primary hyperoxaluria type 3 and result in oxalate overproduction and kidney stone disease. Our previous study showed that carriers of HOGA1 mutations have elevated urinary levels of oxalate precursors. In this study we explored the possibility that mutations in HOGA1 confer a dominant phenotype in the form of kidney stone disease or hyperoxaluria. MATERIALS AND METHODS: An observational analytic case control study was designed to determine the prevalence of pathogenic HOGA1 mutations among adults with calcium-oxalate kidney stone disease. Given the high prevalence of HOGA1 mutations among Ashkenazi Jews, this group was evaluated separately. Carrier frequency of any of the 52 reported pathogenic mutations was compared to data derived from gnomAD for the corresponding ethnic group. Sanger sequencing of HOGA1 gene was performed on DNA samples from the following groups: 60 Ashkenazi Jews and 86 nonAshkenazi calcium-oxalate stone formers, 150 subjects with low and 150 with high urinary oxalate levels. RESULTS: The carrier prevalence of pathogenic mutations among the Ashkenazi Jews was 1.7% compared to 2.8% in the corresponding control group (p=0.9 OR=0.6 95% CI 0.01-3.51). We did not detect any mutation among the nonAshkenazi study group. No correlation was detected between hyperoxaluria and HOGA1 variants. CONCLUSIONS: This study shows that mutations in HOGA1 do not confer a dominant phenotype in the form of calcium-oxalate kidney stone disease or hyperoxaluria.


Asunto(s)
Oxalato de Calcio , Hiperoxaluria/genética , Cálculos Renales/genética , Mutación , Oxo-Ácido-Liasas/genética , Fenotipo , Adulto , Anciano , Oxalato de Calcio/análisis , Estudios de Casos y Controles , Estudios de Cohortes , Femenino , Humanos , Cálculos Renales/química , Masculino , Persona de Mediana Edad
4.
Pediatr Nephrol ; 36(9): 2593-2606, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33156410

RESUMEN

Loss-of-function mutations in three genes, involved in the metabolic pathway of glyoxylate, result in increased oxalate production and its crystallization in the form of calcium oxalate. This leads to three forms of primary hyperoxaluria-an early-onset inherited kidney disease with wide phenotypic variability ranging from isolated kidney stone events to stage 5 chronic kidney disease in infancy. This review provides a description of metabolic processes resulting in oxalate overproduction and summarizes basic therapeutic approaches. Unfortunately, current treatment of primary hyperoxaluria does not allow the prevention of loss of kidney function or to substantially diminish other symptoms in most patients. However, latest breakthroughs in biotechnology provide new promising directions for drug development. Some of them have already progressed to the level of clinical trials; others are just at the stage of proof of concept. Here we review the most advanced technologies including those that have been harnessed as possible therapeutic modalities.


Asunto(s)
Hiperoxaluria Primaria , Humanos , Hiperoxaluria Primaria/terapia
5.
J Cell Sci ; 133(24)2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33177075

RESUMEN

Eukaryotic cells have evolved organelles that allow the compartmentalization and regulation of metabolic processes. Knowledge of molecular mechanisms that allow temporal and spatial organization of enzymes within organelles is therefore crucial for understanding eukaryotic metabolism. Here, we show that the yeast malate dehydrogenase 2 (Mdh2) is dually localized to the cytosol and to peroxisomes and is targeted to peroxisomes via association with Mdh3 and a Pex5-dependent piggybacking mechanism. This dual localization of Mdh2 contributes to our understanding of the glyoxylate cycle and provides a new perspective on compartmentalization of cellular metabolism, which is critical for the perception of metabolic disorders and aging.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Secuencia de Aminoácidos , Citosol/metabolismo , Glioxilatos , Malato Deshidrogenasa/genética , Malato Deshidrogenasa/metabolismo , Peroxisomas/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
6.
J Mol Med (Berl) ; 96(7): 621-630, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29777253

RESUMEN

Primary hyperoxaluria type 1 is a severe kidney stone disease caused by abnormalities of the peroxisomal alanine-glyoxylate aminotransferase (AGT). The most frequent mutation G170R results in aberrant mitochondrial localization of the active enzyme. To evaluate the population of peroxisome-localized AGT, we developed a quantitative Glow-AGT assay based on the self-assembly split-GFP approach and used it to identify drugs that can correct mislocalization of the mutant protein. In line with previous reports, the Glow-AGT assay showed that mitochondrial transport inhibitors DECA and monensin increased peroxisomal localization of the mutant. Here, we demonstrate that prolonged treatment with the translation elongation inhibitor emetine, a medicinal alkaloid used in treatment of amoebiasis, corrected G170R-AGT mislocalization. Furthermore, emetine reduced the augmented oxalate level in culture media of patient-derived hepatocytes bearing the G170R mutation. A distinct translation inhibitor GC7 had a similar effect on the mutant Glow-AGT relocalization indicating that mild translation inhibition is a promising therapeutic approach for primary hyperoxaluria type 1 caused by AGT misfolding/mistargeting. KEY MESSAGES: • There is no effective conservative treatment to decrease oxalate production in PH1 patients. • Chemical chaperones rescue mislocalization of mutant AGT and reduce oxalate levels. • We have developed an assay for precise monitoring of the peroxisomal AGT. • Inhibition of translation by emetine reroutes the mutant protein to peroxisome. • Mild translation inhibition is a promising cure for conformational disorders.


Asunto(s)
Mutación , Biosíntesis de Proteínas , Transaminasas/genética , Transaminasas/metabolismo , Animales , Biomarcadores , Células CHO , Supervivencia Celular , Cricetulus , Hepatocitos/metabolismo , Hiperoxaluria/tratamiento farmacológico , Hiperoxaluria/genética , Hiperoxaluria/metabolismo , Espacio Intracelular/metabolismo , Mitocondrias/metabolismo , Terapia Molecular Dirigida , Oxalatos/metabolismo , Peroxisomas/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Transporte de Proteínas
7.
Pediatr Nephrol ; 33(8): 1443-1446, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29705963

RESUMEN

BACKGROUND: Primary hyperoxaluria type 3 (PH3) is a recently described cause of childhood renal calculi. It results from mutations in the HOGA1 gene and most cases have been diagnosed after clinical ascertainment, exclusion of other genetic hyperoxalurias and mutation testing. Metabolite testing has not been widely applied but holds promise for the rapid screening and diagnosis of patients who are not specifically suspected to have PH3. CASE-DIAGNOSIS/TREATMENT: Two cases presented with renal calculi. Urine metabolite testing by tandem mass spectrometry was performed as part of the routine diagnostic work-up for this condition. Both had significantly increased levels of the PH3 urine marker 4-hydroxyglutamate and related metabolites. The diagnosis of PH3 was confirmed by the finding of bi-allelic damaging HOGA1 mutations. CONCLUSIONS: Urine screening by tandem mass spectrometry is a rapid, high-throughput test that can detect PH3 cases that may otherwise not be diagnosed.


Asunto(s)
Glutamatos/orina , Hiperoxaluria Primaria/diagnóstico , Ácidos Cetoglutáricos/orina , Cálculos Renales/etiología , Oxalatos/orina , Adolescente , Femenino , Glutamatos/metabolismo , Humanos , Hiperoxaluria Primaria/complicaciones , Hiperoxaluria Primaria/genética , Hiperoxaluria Primaria/orina , Lactante , Ácidos Cetoglutáricos/metabolismo , Cálculos Renales/terapia , Cálculos Renales/orina , Litotricia , Masculino , Metabolómica/métodos , Oxo-Ácido-Liasas/genética , Oxo-Ácido-Liasas/metabolismo , Recurrencia , Espectrometría de Masas en Tándem
8.
JIMD Rep ; 15: 1-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-24563386

RESUMEN

Primary hyperoxaluria type 3 (PH3) is a recently identified inborn error of 4-hydroxyproline metabolism causing kidney stone disease. Diagnosis to date has relied on mutation detection. The excretion of 4-hydroxyglutamate (4OHGlu) was investigated in controls and a cohort of nine patients with PH3 and their parents using flow injection tandem mass spectrometry. 4OHGlu was stable in acidified urine samples and was not influenced by diet. Its measurement was readily incorporated into an existing multi-analyte panel for comprehensive screening for inborn errors of metabolism. There was a steady decline with age in 4OHGlu levels, expressed as µmol/mmol of creatinine, in controls. Levels in patients with PH3 ranged from 6.5 to 98 µmol/mmol of creatinine and were all significantly increased when compared to age-matched controls (<4.2). Levels in eight parents (obligatory carriers of the corresponding mutation) were moderately, but significantly increased, ranging from 0.6 to 2.5 (age-matched controls <1.4, p = 0.03). Urine 4OHGlu screening was used to prospectively diagnose PH3 in an 18-month-old boy with calcium oxalate kidney stone disease associated with hyperoxaluria. 4OHGlu was also increased in a stored newborn screening dried blood spot sample from this child (37 µmol/L, controls <2.53). 4OHGlu testing provides a robust and high-throughput biochemical screen for PH3.

9.
J Med Genet ; 51(8): 526-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24996905

RESUMEN

BACKGROUND: The primary hyperoxalurias are a group of recessive kidney diseases, characterised by extensive accumulation of calcium oxalate that progressively coalesces into kidney stones. Oxalate overproduction is facilitated by perturbations in the metabolism of glyoxylate, the product of glycolate oxidation, and the immediate precursor of oxalate. Glycolic aciduria associated with hyperoxaluria is regarded as the hallmark of type 1 primary hyperoxaluria. The genetic basis of isolated glycolic aciduria is reported here. METHODS AND RESULTS: Two brothers, born to consanguineous healthy parents of Arab descent, were evaluated for psychomotor delay associated with triple-A-like syndrome (anisocoria, alacrima and achalasia). The proband showed markedly increased urinary glycolic acid excretion with normal excretion of oxalate, citrate and glycerate. Abdominal ultrasound showed normal-sized kidneys with normal echotexture. The genetic nature of triple-A-like syndrome in this kindred was found to be unrelated to this metabolic abnormality. Direct DNA sequencing of glycolate oxidase gene (HAO1) revealed a homozygous c.814-1G>C mutation in the invariant -1 position of intron 5 splice acceptor site. Since HAO1 is a liver-specific enzyme, the effect of this novel mutation on splicing was validated by an in vitro hybrid-minigene approach. We confirmed the appearance of an abnormal splice variant in cells transfected with mutant minigene vector. CONCLUSIONS: Our results pinpoint the expression of defective splice variant of glycolate oxidase as the cause of isolated asymptomatic glycolic aciduria. This observation contributes to the development of novel approaches, namely, substrate reduction, for the treatment of primary hyperoxaluria type I.


Asunto(s)
Oxidorreductasas de Alcohol/genética , Hiperoxaluria , Errores Innatos del Metabolismo , Insuficiencia Suprarrenal , Niño , Acalasia del Esófago , Glicolatos/orina , Glioxilatos/metabolismo , Humanos , Hiperoxaluria/etiología , Hiperoxaluria/genética , Masculino , Errores Innatos del Metabolismo/complicaciones , Errores Innatos del Metabolismo/genética
10.
J Mol Med (Berl) ; 90(12): 1497-504, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22729392

RESUMEN

Perturbations in glyoxylate metabolism lead to the accumulation of oxalate and give rise to primary hyperoxalurias, recessive disorders characterized by kidney stone disease. Loss-of-function mutations in HOGA1 (formerly DHDPSL) are responsible for primary hyperoxaluria type III. HOGA1 is a mitochondrial 4-hydroxy-2-oxoglutarate aldolase catalyzing the fourth step in the hydroxyproline pathway. We investigated hydroxyproline metabolites in the urine of patients with primary hyperoxaluria type III using gas chromatography-mass spectroscopy. Significant increases in concentrations of 4-hydroxy-2-oxoglutarate and its precursor and derivative 4-hydroxyglutamate and 2,4-dihydroxyglutarate, respectively, were found in all patients as compared to carriers of the corresponding mutations or healthy controls. Despite a functional block in the conversion of hydroxyproline to glyoxylate--the immediate precursor of oxalate--the production of oxalate increases. To explain this apparent contradiction, we propose a model of glyoxylate compartmentalization in which cellular glyoxylate is normally prevented from contact with the cytosol where it can be oxidized to oxalate. We propose that HOGA1 deficiency results in the accumulation of 4-hydroxy-2-oxoglutarate in the mitochondria and its transport into the cytosol where it is converted to glyoxylate by a different cytosolic aldolase. In human hepatocyte cell lines, we detected a cytosolic 4-hydroxy-2-oxoglutarate aldolase activity not due to HOGA1. These studies provide a diagnostic tool for primary hyperoxaluria type III and shed light on glyoxylate metabolism and the pathogenesis of primary hyperoxalurias.


Asunto(s)
Glioxilatos/metabolismo , Hiperoxaluria Primaria/metabolismo , Butileno Glicoles/metabolismo , Butiratos/metabolismo , Línea Celular , Hepatocitos/metabolismo , Humanos , Oxalatos/metabolismo , Oxo-Ácido-Liasas/metabolismo
11.
RNA Biol ; 9(1): 33-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22258151

RESUMEN

Mutations in human mitochondrial tRNA genes are associated with a number of multisystemic disorders. These single nucleotide substitutions in various domains of tRNA molecules may affect different steps of tRNA biogenesis. Often, the prominent decrease of aminoacylation and/or steady-state levels of affected mitochondrial tRNA have been demonstrated in patients' tissues and in cultured cells. Similar effect has been observed for pathogenic mutations in nuclear genes encoding mitochondrial aminoacyl-tRNA-synthetases, while over-expression of mitochondrial aminoacyl-tRNA synthetases or elongation factor EF-Tu rescued mutated tRNAs from degradation. In this review we summarize experimental data concerning the possible regulatory mechanisms governing mitochondrial tRNA steady-state levels, and propose a hypothesis based on the tRNA channelling principle. According to this hypothesis, interaction of mitochondrial tRNA with proteins ensures not only tRNA synthesis, maturation and function, but also protection from degradation. Mutations perturbing this interaction lead to decreased tRNA stability.


Asunto(s)
Mitocondrias/genética , ARN de Transferencia/genética , ARN/genética , Aminoacil-ARNt Sintetasas/genética , Aminoacil-ARNt Sintetasas/metabolismo , Aminoacilación , Genes Mitocondriales , Genoma Humano , Humanos , Mitocondrias/enzimología , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Mutación , Factor Tu de Elongación Peptídica/genética , Factor Tu de Elongación Peptídica/metabolismo , Polimorfismo de Nucleótido Simple , ARN/metabolismo , Procesamiento Postranscripcional del ARN , Estabilidad del ARN , ARN Mitocondrial , ARN de Transferencia/metabolismo , Transcripción Genética
12.
Clin J Am Soc Nephrol ; 6(9): 2289-95, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21896830

RESUMEN

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.


Asunto(s)
Oxalato de Calcio/metabolismo , Predisposición Genética a la Enfermedad , Hiperoxaluria Primaria/genética , Oxo-Ácido-Liasas/genética , Urolitiasis/etiología , Humanos , Mutación , Oxo-Ácido-Liasas/metabolismo , Factores de Riesgo , Urolitiasis/genética
13.
Am J Hum Genet ; 88(2): 193-200, 2011 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-21255763

RESUMEN

An uncharacterized multisystemic mitochondrial cytopathy was diagnosed in two infants from consanguineous Palestinian kindred living in a single village. The most significant clinical findings were tubulopathy (hyperuricemia, metabolic alkalosis), pulmonary hypertension, and progressive renal failure in infancy (HUPRA syndrome). Analysis of the consanguineous pedigree suggested that the causative mutation is in the nuclear DNA. By using genome-wide SNP homozygosity analysis, we identified a homozygous identity-by-descent region on chromosome 19 and detected the pathogenic mutation c.1169A>G (p.Asp390Gly) in SARS2, encoding the mitochondrial seryl-tRNA synthetase. The same homozygous mutation was later identified in a third infant with HUPRA syndrome. The carrier rate of this mutation among inhabitants of this Palestinian isolate was found to be 1:15. The mature enzyme catalyzes the ligation of serine to two mitochondrial tRNA isoacceptors: tRNA(Ser)(AGY) and tRNA(Ser)(UCN). Analysis of amino acylation of the two target tRNAs, extracted from immortalized peripheral lymphocytes derived from two patients, revealed that the p.Asp390Gly mutation significantly impacts on the acylation of tRNA(Ser)(AGY) but probably not that of tRNA(Ser)(UCN). Marked decrease in the expression of the nonacylated transcript and the complete absence of the acylated tRNA(Ser)(AGY) suggest that this mutation leads to significant loss of function and that the uncharged transcripts undergo degradation.


Asunto(s)
Alcalosis Respiratoria/genética , Hipertensión Pulmonar/genética , Hiperuricemia/genética , Mitocondrias/enzimología , Mutación/genética , Insuficiencia Renal/genética , Serina-ARNt Ligasa/genética , Alcalosis Respiratoria/patología , ADN Mitocondrial/genética , Femenino , Humanos , Hipertensión Pulmonar/patología , Hiperuricemia/patología , Lactante , Recién Nacido , Masculino , Linaje , Polimorfismo de Nucleótido Simple , Aminoacil-ARN de Transferencia/genética , Aminoacil-ARN de Transferencia/metabolismo , Insuficiencia Renal/patología , Síndrome
14.
Am J Hum Genet ; 87(3): 392-9, 2010 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-20797690

RESUMEN

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.


Asunto(s)
Hiperoxaluria Primaria/genética , Mutación/genética , Oxo-Ácido-Liasas/genética , Proteínas/genética , Alelos , Secuencia de Aminoácidos , Secuencia de Bases , Niño , Preescolar , Análisis Mutacional de ADN , Familia , Femenino , Humanos , Hidroxiprolina/metabolismo , Lactante , Recién Nacido , Judíos/genética , Masculino , Redes y Vías Metabólicas , Datos de Secuencia Molecular , Oxalatos/metabolismo , Oxo-Ácido-Liasas/química , Linaje , Proteínas/química
15.
Pediatr Nephrol ; 24(12): 2369-73, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19806368

RESUMEN

Dent's disease is an X-linked proximal tubulopathy. It often manifests in childhood with symptoms of Fanconi syndrome and low-molecular-weight proteinuria. We describe four boys from three unrelated families whose only presenting symptoms of Dent's disease were nephrotic-range proteinuria and histological findings of focal segmental and/or global glomerulosclerosis. In all families, a causal mutation in the CLCN5 gene, encoding a voltage-gated chloride transporter and chloride-proton exchanger, was identified. All three mutations are pathogenic: two are novel (p.Asp727fs and p.Trp122X), and one is a recurrent mutation, p.R648X. Given the atypical phenotype of these patients with Dent's disease, it is possible that this clinical entity is markedly underdiagnosed and that our report represents only the tip of the iceberg. The diagnosis of Dent's disease should be considered in all patients with nephrotic-range proteinuria without hypoalbuminemia or edema. Establishing the diagnosis of Dent's disease will prevent the administration of unnecessary immunosuppressive medications with their undesirable side effects.


Asunto(s)
Canales de Cloruro/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Glomeruloesclerosis Focal y Segmentaria/patología , Proteinuria/genética , Biopsia , Calcio/orina , Niño , Preescolar , Codón sin Sentido , Creatinina/orina , ADN/genética , ADN/aislamiento & purificación , Análisis Mutacional de ADN , Glomeruloesclerosis Focal y Segmentaria/diagnóstico , Humanos , Riñón/cirugía , Masculino , Tasa de Depuración Metabólica
16.
Am J Hum Genet ; 84(1): 80-4, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19110212

RESUMEN

The spondylo-meta-epiphyseal dysplasia [SMED] short limb-hand type [SMED-SL] is a rare autosomal-recessive disease, first reported by Borochowitz et al. in 1993.(1) Since then, 14 affected patients have been reported.(2-5) We diagnosed 6 patients from 5 different consanguineous Arab Muslim families from the Jerusalem area with SMED-SL. Additionally, we studied two patients from Algerian and Pakistani ancestry and the parents of the first Jewish patients reported.(1) Using a homozygosity mapping strategy, we located a candidate region on chromosome 1q23 spanning 2.4 Mb. The position of the Discoidin Domain Receptor 2 (DDR2) gene within the candidate region and the similarity of the ddr2 knockout mouse to the SMED patients' phenotype prompted us to study this gene(6). We identified three missense mutations c.2254 C > T [R752C], c. 2177 T > G [I726R], c.2138C > T [T713I] and one splice site mutation [IVS17+1g > a] in the conserved sequence encoding the tyrosine kinase domain of the DDR2 gene. The results of this study will permit an accurate early prenatal diagnosis and carrier screening for families at risk.


Asunto(s)
Calcinosis/genética , Predisposición Genética a la Enfermedad , Deformidades Congénitas de la Mano/genética , Osteocondrodisplasias/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Mitogénicos/metabolismo , Secuencia de Aminoácidos , Calcinosis/enzimología , Cromosomas Humanos Par 1/genética , Consanguinidad , Receptores con Dominio Discoidina , Deformidades Congénitas de la Mano/enzimología , Humanos , Datos de Secuencia Molecular , Osteocondrodisplasias/enzimología , Adulto Joven
17.
Pediatr Nephrol ; 24(2): 403-6, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18791744

RESUMEN

Congenital analbuminemia is a rare autosomal recessive disease in which albumin is not synthesized. Patients with this disorder generally have minimal symptoms despite complete absence of the most abundant serum protein. We report a family in which the proband presented with acute glomerulonephritis and was found to have underlying congenital analbuminemia. Consequently, the patient's two older sisters were diagnosed with the same condition. Sequencing of the human serum albumin gene was performed, and a homozygous mutation in exon 3 was found in all three patients. Together with these three patients of Arab ethnicity, this mutation, known as Kayseri, is the most frequently described mutation in congenital analbuminemia. This article discusses clinical features and diagnostic challenges of this disorder, particularly in this case, where concomitant renal disease was present.


Asunto(s)
Glomerulonefritis/complicaciones , Glomerulonefritis/diagnóstico , Hipoalbuminemia/complicaciones , Hipoalbuminemia/diagnóstico , Albúmina Sérica/genética , Enfermedad Aguda , Adolescente , Adulto , Salud de la Familia , Femenino , Genes Recesivos , Humanos , Hipoalbuminemia/congénito , Hipoalbuminemia/genética , Masculino , Adulto Joven
18.
Int J Oncol ; 27(1): 143-8, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15942653

RESUMEN

Since early diagnosis of many types of cancer greatly improves the chances for successful treatment, high-quality methods for cancer detection are necessary. Our laboratory develops chimeric proteins for targeted therapy, such as gonadotropin releasing hormone (GnRH)-based chimeric proteins for the targeted therapy of adenocarcinomas in humans. For chimeric proteins to cause specific cell death, they must first recognize specific receptors/binding sites expressed on the surface of target cells. Thus, we examined whether we could exploit these binding sites not only as targets for the killing of specific cells but also as a diagnostic marker for identifying adenocarcinomas, using the same chimeric proteins. In this report, we show that one such GnRH-based chimeric protein, GnRH-Caspase3, can indeed serve as a diagnostic tool. GnRH-Caspase3 was able to specifically bind adenocarcinoma cells, as measured by FACS analysis and demonstrated with the aid of confocal microscopy and specific antibodies. Moreover, we found a correlation between cell sensitivity to treatment and the binding level of the chimeric protein to the cells. Hence, we suggest that in addition to their therapeutic potential, GnRH-based chimeric proteins can be used as a diagnostic tool for the detection of adenocarcinomas.


Asunto(s)
Adenocarcinoma/diagnóstico , Hormona Liberadora de Gonadotropina/química , Proteínas Recombinantes de Fusión/química , Adenocarcinoma/metabolismo , Sitios de Unión , Caspasa 3 , Caspasas/metabolismo , Línea Celular Tumoral , Separación Celular , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Microscopía Confocal , Plásmidos/metabolismo , Unión Proteica , Proteínas Recombinantes de Fusión/metabolismo , Sensibilidad y Especificidad
19.
Clin Immunol ; 110(1): 89-99, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14962800

RESUMEN

We previously constructed a pro-apoptotic Fcepsilon-Bak chimeric protein, targeted against cells expressing the IgE high affinity receptor (FcepsilonRI). We demonstrated that the chimeric protein is internalized by target mast cells and kills them. These results, which constitute a promising basis for applying this approach to antiallergic therapy, raise some theoretical questions with respect to two major issues: (a) is the monomeric Fcepsilon-Bak-FcepsilonRI complex able to undergo endocytosis, and (b) does the receptor binding domain of human IgE (Fcepsilon) react with rodent FcepsilonRI? In an attempt to answer these questions, we have now thoroughly investigate the interaction of human (h) and mouse (m) Fcepsilon-Bak with FcepsilonRI-positive cells. Using established cultures of rodent and human origin, as well as a primary mouse mast cell culture, we demonstrate that binding of the chimeric protein to the membrane is followed by quick endocytosis, leading to the apoptosis of specific cells. We also confirm that this interaction depends on FcepsilonRI and not on other IgE receptors. We found that the effect of Fcepsilon-Bak on the cells depends on the level of surface FcepsilonRI expression, but not on the origin of the target cells or of the Fcepsilon moiety. We suggest that endocytosis of the monomeric Fcepsilon-Bak-FcepsilonRI complex results from the inability of Fcepsilon-Bak to transduce signals, characteristic of the monomeric IgE-FcepsilonRI complex due to the absence of the variable portion of the IgE molecule. Our results also indicate that at least the Fc fragment of human IgE is able to interact with both human and rodent FcRI.


Asunto(s)
Inmunoglobulina E/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de IgE/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Animales , Apoptosis/inmunología , Apoptosis/fisiología , Degranulación de la Célula/inmunología , Humanos , Inmunoglobulina E/inmunología , Mastocitos/inmunología , Mastocitos/metabolismo , Proteínas de la Membrana/genética , Ratones , Receptores de IgE/genética , Receptores de IgE/inmunología , Proteínas Recombinantes de Fusión/genética , Proteína Destructora del Antagonista Homólogo bcl-2
20.
Biochem Biophys Res Commun ; 290(1): 332-8, 2002 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-11779174

RESUMEN

We recently designed and constructed chimeric proteins for the elimination of specific cell populations. These chimeric proteins are composed of a targeting component fused to an apoptotic protein as the killing moiety. However, chimeric proteins can serve not only to eliminate cell populations, but also as "biological tools" for studying the fate of endogenous proteins. We show here that upon entering their target cell, a variety of chimeric proteins composed of an endogenous protein as their killing moiety reach the subcellular location of their endogenous counterpart. In contrast, bacterial-based killing domains head for the subcellular site of their substrate. Moreover, the chimeric protein acts similarly to the endogenous protein, while causing the cell to die. Therefore, chimeric proteins may serve as a unique tool for investigating cellular proteins and their intracellular localization, without the need to overexpress them.


Asunto(s)
Técnicas de Laboratorio Clínico/métodos , Proteínas Proto-Oncogénicas c-bcl-2 , Proteínas Recombinantes de Fusión/química , Animales , Apoptosis , Línea Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Escherichia coli/metabolismo , Colorantes Fluorescentes/farmacología , Humanos , Interleucina-2/biosíntesis , Linfocitos , Microscopía Confocal/métodos , Microscopía Fluorescente/métodos , Proteínas Proto-Oncogénicas/biosíntesis , Células Tumorales Cultivadas , Proteína X Asociada a bcl-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...