Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Inherit Metab Dis ; 45(4): 769-781, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35279850

RESUMEN

Congenital disorders of glycosylation type 1 (CDG-I) comprise a group of 27 genetic defects with heterogeneous multisystem phenotype, mostly presenting with nonspecific neurological symptoms. The biochemical hallmark of CDG-I is a partial absence of complete N-glycans on transferrin. However, recent findings of a diagnostic N-tetrasaccharide for ALG1-CDG and increased high-mannose N-glycans for a few other CDG suggested the potential of glycan structural analysis for CDG-I gene discovery. We analyzed the relative abundance of total plasma N-glycans by high resolution quadrupole time-of-flight mass spectrometry in a large cohort of 111 CDG-I patients with known (n = 75) or unsolved (n = 36) genetic cause. We designed single-molecule molecular inversion probes (smMIPs) for sequencing of CDG-I candidate genes on the basis of specific N-glycan signatures. Glycomics profiling in patients with known defects revealed novel features such as the N-tetrasaccharide in ALG2-CDG patients and a novel fucosylated N-pentasaccharide as specific glycomarker for ALG1-CDG. Moreover, group-specific high-mannose N-glycan signatures were found in ALG3-, ALG9-, ALG11-, ALG12-, RFT1-, SRD5A3-, DOLK-, DPM1-, DPM3-, MPDU1-, ALG13-CDG, and hereditary fructose intolerance. Further differential analysis revealed high-mannose profiles, characteristic for ALG12- and ALG9-CDG. Prediction of candidate genes by glycomics profiling in 36 patients with thus far unsolved CDG-I and subsequent smMIPs sequencing led to a yield of solved cases of 78% (28/36). Combined plasma glycomics profiling and targeted smMIPs sequencing of candidate genes is a powerful approach to identify causative mutations in CDG-I patient cohorts.


Asunto(s)
Trastornos Congénitos de Glicosilación , Trastornos Congénitos de Glicosilación/diagnóstico , Trastornos Congénitos de Glicosilación/genética , Glicómica , Glicosilación , Humanos , Manosa , Manosiltransferasas/genética , N-Acetilglucosaminiltransferasas , Oligosacáridos , Polisacáridos/genética
2.
Hum Mol Genet ; 27(17): 3029-3045, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29878199

RESUMEN

Genomics methodologies have significantly improved elucidation of Mendelian disorders. The combination with high-throughput functional-omics technologies potentiates the identification and confirmation of causative genetic variants, especially in singleton families of recessive inheritance. In a cohort of 99 individuals with abnormal Golgi glycosylation, 47 of which being unsolved, glycomics profiling was performed of total plasma glycoproteins. Combination with whole-exome sequencing in 31 cases revealed a known genetic defect in 15 individuals. To identify additional genetic factors, hierarchical clustering of the plasma glycomics data was done, which indicated a subgroup of four patients that shared a unique glycomics signature of hybrid type N-glycans. In two siblings, compound heterozygous mutations were found in SLC10A7, a gene of unknown function in human. These included a missense mutation that disrupted transmembrane domain 4 and a mutation in a splice acceptor site resulting in skipping of exon 9. The two other individuals showed a complete loss of SLC10A7 mRNA. The patients' phenotype consisted of amelogenesis imperfecta, skeletal dysplasia, and decreased bone mineral density compatible with osteoporosis. The patients' phenotype was mirrored in SLC10A7 deficient zebrafish. Furthermore, alizarin red staining of calcium deposits in zebrafish morphants showed a strong reduction in bone mineralization. Cell biology studies in fibroblasts of affected individuals showed intracellular mislocalization of glycoproteins and a defect in post-Golgi transport of glycoproteins to the cell membrane. In contrast to yeast, human SLC10A7 localized to the Golgi. Our combined data indicate an important role for SLC10A7 in bone mineralization and transport of glycoproteins to the extracellular matrix.


Asunto(s)
Enfermedades del Desarrollo Óseo/etiología , Calcificación Fisiológica , Trastornos Congénitos de Glicosilación/complicaciones , Genómica , Glicómica , Mutación , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Simportadores/genética , Adulto , Animales , Enfermedades del Desarrollo Óseo/metabolismo , Enfermedades del Desarrollo Óseo/patología , Células Cultivadas , Estudios de Cohortes , Exoma , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Glicosilación , Aparato de Golgi/metabolismo , Aparato de Golgi/patología , Humanos , Lactante , Masculino , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Linaje , Fenotipo , Transporte de Proteínas , Simportadores/metabolismo , Adulto Joven , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
3.
Mol Genet Metab ; 126(1): 43-52, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30470562

RESUMEN

Succinate-CoA ligase (SUCL) is a heterodimer consisting of an alpha subunit encoded by SUCLG1, and a beta subunit encoded by either SUCLA2 or SUCLG2 catalyzing an ATP- or GTP-forming reaction, respectively, in the mitochondrial matrix. The deficiency of this enzyme represents an encephalomyopathic form of mtDNA depletion syndromes. We describe the fatal clinical course of a female patient with a pathogenic mutation in SUCLG1 (c.626C > A, p.Ala209Glu) heterozygous at the genomic DNA level, but homozygous at the transcriptional level. The patient exhibited early-onset neurometabolic abnormality culminating in severe brain atrophy and dystonia leading to death by the age of 3.5 years. Urine and plasma metabolite profiling was consistent with SUCL deficiency which was confirmed by enzyme analysis and lack of mitochondrial substrate-level phosphorylation (mSLP) in skin fibroblasts. Oxygen consumption- but not extracellular acidification rates were altered only when using glutamine as a substrate, and this was associated with mild mtDNA depletion and no changes in ETC activities. Immunoblot analysis revealed no detectable levels of SUCLG1, while SUCLA2 and SUCLG2 protein expressions were largely reduced. Confocal imaging of triple immunocytochemistry of skin fibroblasts showed that SUCLG2 co-localized only partially with the mitochondrial network which otherwise exhibited an increase in fragmentation compared to control cells. Our results outline the catastrophic consequences of the mutated SUCLG1 leading to strongly reduced SUCL activity, mSLP impairment, mislocalization of SUCLG2, morphological alterations in mitochondria and clinically to a severe neurometabolic disease, but in the absence of changes in mtDNA levels or respiratory complex activities.


Asunto(s)
Mitocondrias/patología , Enfermedades Mitocondriales/diagnóstico , Mutación , Succinato-CoA Ligasas/genética , Preescolar , ADN Mitocondrial/genética , Resultado Fatal , Femenino , Heterocigoto , Homocigoto , Humanos , Mitocondrias/metabolismo , Fosforilación , Succinato-CoA Ligasas/sangre , Succinato-CoA Ligasas/orina
4.
Hum Mol Genet ; 23(23): 6356-65, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25008109

RESUMEN

Complex III (cytochrome bc1) is a protein complex of the mitochondrial inner membrane that transfers electrons from ubiquinol to cytochrome c. Its assembly requires the coordinated expression of mitochondrial-encoded cytochrome b and nuclear-encoded subunits and assembly factors. Complex III deficiency is a severe multisystem disorder caused by mutations in subunit genes or assembly factors. Sequence-profile-based orthology predicts C11orf83, hereafter named UQCC3, to be the ortholog of the fungal complex III assembly factor CBP4. We describe a homozygous c.59T>A missense mutation in UQCC3 from a consanguineous patient diagnosed with isolated complex III deficiency, displaying lactic acidosis, hypoglycemia, hypotonia and delayed development without dysmorphic features. Patient fibroblasts have reduced complex III activity and lower levels of the holocomplex and its subunits than controls. They have no detectable UQCC3 protein and have lower levels of cytochrome b protein. Furthermore, in patient cells, cytochrome b is absent from a high-molecular-weight complex III. UQCC3 is reduced in cells depleted for the complex III assembly factors UQCC1 and UQCC2. Conversely, absence of UQCC3 in patient cells does not affect UQCC1 and UQCC2. This suggests that UQCC3 functions in the complex III assembly pathway downstream of UQCC1 and UQCC2 and is consistent with what is known about the function of Cbp4 and of the fungal orthologs of UQCC1 and UQCC2, Cbp3 and Cbp6. We conclude that UQCC3 functions in complex III assembly and that the c.59T>A mutation has a causal role in complex III deficiency.


Asunto(s)
Proteínas Portadoras/genética , Citocromos b/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , Proteínas de Saccharomyces cerevisiae/genética , Secuencia de Aminoácidos , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Consanguinidad , Complejo III de Transporte de Electrones/deficiencia , Complejo III de Transporte de Electrones/genética , Estabilidad de Enzimas , Femenino , Fibroblastos/metabolismo , Humanos , Recién Nacido , Proteínas de la Membrana/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Datos de Secuencia Molecular , Mutación Missense
5.
Am J Hematol ; 91(12): E482-E490, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27643674

RESUMEN

TMPRSS6 variants that affect protein function result in impaired matriptase-2 function and consequently uninhibited hepcidin production, leading to iron refractory iron deficiency anemia (IRIDA). This disease is characterized by microcytic, hypochromic anemia and serum hepcidin values that are inappropriately high for body iron levels. Much is still unknown about its pathophysiology, genotype-phenotype correlation, and optimal clinical management. We describe 14 different TMPRSS6 variants, of which 9 are novel, in 21 phenotypically affected IRIDA patients from 20 families living in the Netherlands; 16 out of 21 patients were female. In 7 out of 21 cases DNA sequencing and multiplex ligation dependent probe amplification demonstrated only heterozygous TMPRSS6 variants. The age at presentation, disease severity, and response to iron supplementation were highly variable, even for patients and relatives with similar TMPRSS6 genotypes. Mono-allelic IRIDA patients had a milder phenotype with respect to hemoglobin and MCV and presented significantly later in life with anemia than bi-allelic patients. Transferrin saturation (TSAT)/hepcidin ratios were lower in IRIDA probands than in healthy relatives. Most patients required parenteral iron. Genotype alone was not predictive for the response to oral iron. We conclude that IRIDA is a genotypically and phenotypically heterogeneous disease. The high proportion of female patients and the discrepancy between phenotypes of probands and relatives with the same genotype, suggest a complex interplay between genetic and acquired factors in the pathogenesis of IRIDA. In the absence of inflammation, the TSAT/hepcidin ratio is a promising diagnostic tool, even after iron supplementation has been given. Am. J. Hematol. 91:E482-E490, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Anemia Ferropénica , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Países Bajos , Adulto Joven
6.
N Engl J Med ; 360(22): 2310-8, 2009 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-19474428

RESUMEN

Dehydroepiandrosterone (DHEA) sulfotransferase, known as SULT2A1, converts the androgen precursor DHEA to its inactive sulfate ester, DHEAS [corrected], thereby preventing the conversion of DHEA to an active androgen. SULT2A1 requires 3'-phosphoadenosine-5'-phosphosulfate (PAPS) for catalytic activity. We have identified compound heterozygous mutations in the gene encoding human PAPS synthase 2 (PAPSS2) in a girl with premature pubarche, hyperandrogenic anovulation, very low DHEAS levels, and increased androgen levels. In vitro coincubation of human SULT2A1 and wild-type or mutant PAPSS2 proteins confirmed the inactivating nature of the mutations. These observations indicate that PAPSS2 deficiency is a monogenic adrenocortical cause of androgen excess.


Asunto(s)
Complejos Multienzimáticos/genética , Mutación , Pubertad Precoz/genética , Sulfato Adenililtransferasa/genética , Andrógenos/sangre , Androstenodiona/sangre , Niño , Deshidroepiandrosterona/sangre , Deshidroepiandrosterona/metabolismo , Diagnóstico Diferencial , Femenino , Heterocigoto , Humanos , Complejos Multienzimáticos/deficiencia , Complejos Multienzimáticos/metabolismo , Síndrome del Ovario Poliquístico/diagnóstico , Pubertad Precoz/sangre , ARN Mensajero/metabolismo , Análisis de Secuencia de ADN , Sulfato Adenililtransferasa/deficiencia , Sulfato Adenililtransferasa/metabolismo , Sulfotransferasas/sangre , Sulfotransferasas/metabolismo , Testosterona/sangre
7.
Hum Mol Genet ; 18(18): 3365-74, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19525295

RESUMEN

Contiguous gene syndromes affecting the mitochondrial oxidative phosphorylation system have been rarely reported. Here, we describe a patient with apparent mitochondrial encephalomyopathy accompanied by several unusual features, including dysmorphism and hepatopathy, caused by a homozygous triple gene deletion on chromosome 5. The deletion encompassed the NDUFAF2, ERCC8 and ELOVL7 genes, encoding complex I assembly factor 2 (also known as human B17.2L), a protein of the transcription-coupled nucleotide excision repair (TC-NER) machinery, and a putative elongase of very long-chain fatty acid synthesis, respectively. Detailed evaluation of cultured skin fibroblasts revealed disturbed complex I assembly, depolarization of the mitochondrial membrane, elevated cellular NAD(P)H level, increased superoxide production and defective TC-NER. ELOVL7 mRNA was not detectable in these cells and no alterations in fatty acid synthesis were found. By means of baculoviral complementation we were able to restore the aberrations, thereby establishing causative links between genotype and cell-physiological phenotype. This first chromosomal microdeletion illustrates that beside primary defects in mitochondrial genes also additional genes possibly contribute to the disease phenotype, providing an additional explanation for the broad clinical symptoms associated with these disorders.


Asunto(s)
Anomalías Múltiples/genética , Acetiltransferasas/genética , Enzimas Reparadoras del ADN/genética , Eliminación de Gen , Proteínas Mitocondriales/genética , Chaperonas Moleculares/genética , Factores de Transcripción/genética , Anomalías Múltiples/metabolismo , Resultado Fatal , Elongasas de Ácidos Grasos , Ácidos Grasos/metabolismo , Femenino , Humanos , Recién Nacido , Mitocondrias/metabolismo , Mutación , Oxidación-Reducción , Fosforilación , Unión Proteica
8.
Hum Mol Genet ; 17(7): 986-95, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18156159

RESUMEN

Mutations in mitochondrial DNA (mtDNA) are frequent in cancers but it is not yet clearly established whether they are modifier events involved in cancer progression or whether they are a consequence of tumorigenesis. Here we show a benign tumor type in which mtDNA mutations that lead to complex I (CI) enzyme deficiency are found in all tumors and are the only genetic alteration detected. Actually renal oncocytomas are homogeneous tumors characterized by dense accumulation of mitochondria and we had found that they are deficient in electron transport chain complex I (CI, NADH-ubiquinone oxidoreductase). In this work total sequencing of mtDNA showed that 9/9 tumors harbored point mutations in mtDNA, seven in CI genes, one in complex III, and one in the control region. 7/8 mutations were somatic. All tumors were somatically deficient for CI. The clonal amplification of mutated mtDNA in 8/9 tumors demonstrates that these alterations are selected and therefore favor or trigger growth. No nuclear DNA rearrangement was detected beside mtDNA defects. We hypothesize that functional deficiency of the oxidative phosphorylation CI could create a loop of amplification of mitochondria during cell division, impair substrates oxidation and increase intermediary metabolites availability.


Asunto(s)
Adenoma Oxifílico/genética , ADN Mitocondrial/genética , Complejo I de Transporte de Electrón/genética , Neoplasias Renales/genética , Adenoma Oxifílico/metabolismo , Técnicas de Cultivo de Célula , Núcleo Celular/genética , Proliferación Celular , Citrato (si)-Sintasa/metabolismo , Análisis Mutacional de ADN , ADN Polimerasa gamma , ADN Polimerasa Dirigida por ADN/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Amplificación de Genes , Glucosa/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Renales/metabolismo , NADH Deshidrogenasa/metabolismo , Hibridación de Ácido Nucleico , Fosforilación Oxidativa , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ADN
9.
Eur J Hum Genet ; 23(2): 202-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24781757

RESUMEN

Defects in complex II of the mitochondrial respiratory chain are a rare cause of mitochondrial disorders. Underlying autosomal-recessive genetic defects are found in most of the 'SDHx' genes encoding complex II (SDHA, SDHB, SDHC, and SDHD) and its assembly factors. Interestingly, SDHx genes also function as tumor suppressor genes in hereditary paragangliomas, pheochromocytomas, and gastrointestinal stromal tumors. In these cases, the affected patients are carrier of a heterozygeous SDHx germline mutation. Until now, mutations in SDHx associated with mitochondrial disease have not been reported in association with hereditary tumors and vice versa. Here, we characterize four patients with isolated complex II deficiency caused by mutations in SDHA presenting with multisystem mitochondrial disease including Leigh syndrome (LS) and/or leukodystrophy. Molecular genetic analysis revealed three novel mutations in SDHA. Two mutations (c.64-2A>G and c.1065-3C>A) affect mRNA splicing and result in loss of protein expression. These are the first mutations described affecting SDHA splicing. For the third new mutation, c.565T>G, we show that it severely affects enzyme activity. Its pathogenicity was confirmed by lentiviral complementation experiments on the fibroblasts of patients carrying this mutation. It is of special interest that one of our LS patients harbored the c.91C>T (p.Arg31*) mutation that was previously only reported in association with paragangliomas and pheochromocytomas, tightening the gap between these two rare disorders. As tumor screening is recommended for SDHx mutation carriers, this should also be considered for patients with mitochondrial disorders and their family members.


Asunto(s)
Complejo II de Transporte de Electrones/genética , Enfermedad de Leigh/genética , Leucoencefalopatías/genética , Neoplasias/genética , Secuencia de Aminoácidos , Células Cultivadas , Niño , Preescolar , Complejo II de Transporte de Electrones/química , Fibroblastos/metabolismo , Humanos , Lactante , Enfermedad de Leigh/diagnóstico , Leucoencefalopatías/diagnóstico , Datos de Secuencia Molecular , Mutación Missense , Empalme del ARN
10.
J Neurol Sci ; 326(1-2): 24-8, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23343605

RESUMEN

Coenzyme Q10 (ubiquinone or CoQ10) serves as a redox carrier in the mitochondrial oxidative phosphorylation system. The reduced form of this lipid-soluble antioxidant (ubiquinol) is involved in other metabolic processes as well, such as preventing reactive oxygen species (ROS) induced damage from the mitochondrial membrane. Primary coenzyme Q10 deficiency is a rare, autosomal recessive disorder, often presenting with neurological and/or muscle involvement. Until now, five patients from four families have been described with primary coenzyme Q10 deficiency due to mutations in COQ2 encoding para-hydroxybenzoate polyprenyl transferase. Interestingly, four of these patients showed a distinctive renal involvement (focal segmental glomerular sclerosis, crescentic glomerulonephritis, nephrotic syndrome), which is only very rarely seen in correlation with mitochondrial disorders. The fifth patient deceases due to infantile multi organ failure, also with renal involvement. Here we report a novel homozygous mutation in COQ2 (c.905C>T, p.Ala302Val) in a dizygotic twin from consanguineous Turkish parents. The children were born prematurely and died at the age of five and six months, respectively, after an undulating disease course involving apneas, seizures, feeding problems and generalized edema, alternating with relative stable periods without the need of artificial ventilation. There was no evidence for renal involvement. We would like to raise awareness for this potentially treatable disorder which could be under diagnosed in patients with fatal neonatal or infantile multi-organ disease.


Asunto(s)
Transferasas Alquil y Aril/deficiencia , Transferasas Alquil y Aril/genética , Enfermedades en Gemelos/genética , Enfermedades Metabólicas/genética , Insuficiencia Multiorgánica/genética , Mutación/genética , Secuencia de Aminoácidos , Enfermedades en Gemelos/diagnóstico , Enfermedades en Gemelos/enzimología , Resultado Fatal , Femenino , Homocigoto , Humanos , Lactante , Masculino , Enfermedades Metabólicas/diagnóstico , Enfermedades Metabólicas/enzimología , Datos de Secuencia Molecular , Insuficiencia Multiorgánica/diagnóstico , Insuficiencia Multiorgánica/enzimología
11.
Ann Neurol ; 61(1): 73-83, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17262856

RESUMEN

OBJECTIVE: Mitochondrial complex I deficiency is the commonest diagnosed respiratory chain defect, being genetically heterogeneous. The male preponderance of previous patient cohorts suggested an X-linked underlying genetic defect. We investigated mutations in the X-chromosomal complex I structural genes, NDUFA1 and NDUFB11, as a novel cause of mitochondrial encephalomyopathy. METHODS: We sequenced 12 nuclear genes and the mitochondrial DNA-encoded complex I genes in 26 patients with respiratory chain complex I defect. Novel mutations were confirmed by polymerase chain reaction restriction length polymorphism. Assembly/stability studies in fibroblasts were performed using two-dimensional blue native gel electrophoresis. RESULTS: Two novel p.Gly8Arg and p.Arg37Ser hemizygous mutations in NDUFA1 were identified in two unrelated male patients presenting with Leigh's syndrome and with myoclonic epilepsy and developmental delay, respectively. Two-dimensional blue native gel electrophoresis showed decreased levels of intact complex I with no accumulation of lower molecular weight subcomplexes, indicating that assembly, stability, or both are compromised. INTERPRETATION: Mutations in the X-linked NDUFA1 gene result in complex I defect and encephalomyopathy. Assembly/stability analysis might give an explanation for the different clinical phenotypes and become useful for future diagnostic purposes.


Asunto(s)
Genes Ligados a X , Encefalomiopatías Mitocondriales/genética , NADH Deshidrogenasa/genética , Animales , Arginina/genética , Análisis Mutacional de ADN/métodos , Complejo I de Transporte de Electrón/metabolismo , Electroforesis en Gel Bidimensional/métodos , Femenino , Glicina/genética , Humanos , Masculino , Encefalomiopatías Mitocondriales/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Alineación de Secuencia , Serina/genética
12.
Mol Genet Metab ; 90(1): 10-4, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16996290

RESUMEN

We describe a novel mitochondrial ND2 mutation (T4681C) in a patient presenting with Leigh Syndrome. Biochemical analyses revealed a low isolated complex I activity in patient's fibroblasts, blood and skeletal muscle. Mutant transmitochondrial cybrid clones retained the specific complex I defect, demonstrating the mitochondrial genetic origin of the disease. The mutation leads to a L71P substitution at an evolutionary conserved amino acid stretch. By two-dimensional blue native electrophoresis (2D-BN-SDS-PAGE), decreased complex I levels were observed together with an accumulation of specific assembly intermediates, suggesting that the mutation disturbs the complex I assembly pathway.


Asunto(s)
Complejo I de Transporte de Electrón/genética , Enfermedad de Leigh/enzimología , Enfermedad de Leigh/genética , Proteínas Mitocondriales/genética , Mutación Missense , NADH Deshidrogenasa/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Células Cultivadas , Niño , Complejo I de Transporte de Electrón/deficiencia , Complejo I de Transporte de Electrón/metabolismo , Humanos , Lactante , Enfermedad de Leigh/etiología , Masculino , Proteínas Mitocondriales/fisiología , Datos de Secuencia Molecular , NADH Deshidrogenasa/fisiología
13.
Hum Genet ; 110(3): 264-70, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11935339

RESUMEN

The human mitochondrial NADH:ubiquinone oxidoreductase (complex I), the first complex of the oxidative phosphorylation system, is composed of at least 42 subunits. Little is known about the assembly process of these subunits into the mature complex. Recently, two proteins in Neurospora crassa have been found to be involved in the assembly of complex I. These proteins are not constituent parts of the mature complex but are associated with smaller intermediate complexes of the assembly process and have a chaperone-like function. We have characterized the human homologue of one of these two complex I intermediate associated proteins, named CIA30, and show that expression of the human CIA30 protein is ubiquitous with a slightly higher expression in various heart tissues, kidney, lung and liver. As deletion of the Neurospora crassa CIA genes results in severe disruption of the assembly process, human CIA30 can be considered as a candidate gene related to complex I deficiency. Thirteen patients with an isolated complex I deficiency, but who were ruled out for mutations in the 35 nuclear genes of the complex and mtDNA, were subjected to mutational analysis of the gene coding for the human CIA30 protein. Four new single nucleotide polymorphisms (SNPs) were detected but no functional mutation was found.


Asunto(s)
NADH NADPH Oxidorreductasas/deficiencia , NADH NADPH Oxidorreductasas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Cromosomas Humanos Par 15/genética , ADN Mitocondrial/genética , Complejo I de Transporte de Electrón , Expresión Génica , Humanos , Datos de Secuencia Molecular , Mutación , NADH NADPH Oxidorreductasas/química , NADH NADPH Oxidorreductasas/metabolismo , Polimorfismo de Nucleótido Simple , Subunidades de Proteína , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Distribución Tisular
14.
J Hum Genet ; 48(1): 8-13, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12560872

RESUMEN

Mitochondrial beta-oxidation of long-chain fatty acids requires the concerted action of three tightly integrated membrane-bound enzymes (carnitine palmitoyltransferase I and II and carnitine/acylcarnitine translocase) that transport them into mitochondria. Neonatal onset of carnitine palmitoyltransferase II (CPT II) deficiency is an autosomal recessive, often lethal disorder of this transport. We describe a novel splice-site mutation in the CPT II gene, found in a Moroccan family, of which four out of five children have died from the neonatal form of CPT II deficiency. Mutation detection studies at the mRNA level in the CPT II gene implied that the affected children were homozygous for the previously reported 534T insertion followed by a 25-bp deletion (encompassing bases 534-558). Studies of genomic DNA, however, revealed all patients to be compound heterozygous for this 534T ins/del 25 mutation, and for a new g-->a splice-site mutation in the splice-acceptor site of intron 2. Because of these findings, prenatal diagnosis was performed in chorionic villi of three new pregnancies. This did not reveal new compound heterozygous genotypes, and, after uneventful pregnancies, all children appeared to be healthy. The new mutation is the first splice-site mutation ever identified in CPT II deficiency. The fact that it was not discovered in the patient's cDNA makes this study another example of the incompleteness of mutation detection at the mRNA level in cases where a mutation leads to aberrant splicing or nonsense-mediated messenger decay.


Asunto(s)
Carnitina O-Palmitoiltransferasa/deficiencia , Carnitina O-Palmitoiltransferasa/genética , Mutación , Sitios de Empalme de ARN , Femenino , Humanos , Recién Nacido , Masculino , Linaje , Diagnóstico Prenatal , ARN Mensajero , Análisis de Secuencia de ARN
15.
Eur J Pediatr ; 162(10): 714-8, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12925875

RESUMEN

UNLABELLED: A boy with recurrent episodes of hypoglycaemia and ataxia, microcephaly, mental retardation, permanent lactic acidaemia, intermittent 2-oxoglutaric aciduria as well as elevation of serum branched chain amino acids was diagnosed with dihydrolipoamide dehydrogenase (E3) deficiency. Analysis of genomic DNA revealed compound heterozygosity for two novel mutations: I393T in exon 11, located at the interface domain of the protein and possibly interfering with its dimerisation, and IVS9+1G>A located at a consensus splice site. A heterozygous polymorphism was also detected. In the patient's cDNA the I393T mutation and the polymorphism appeared to be homozygous, indicating that the mRNA coming from the IVS9+1G>A mutant allele is not stable. CONCLUSION: as opposed to the non-neurological phenotype of patients with a homozygous G229C mutation, this patient developed Leigh syndrome. Dihydrolipoamide dehydrogenase and pyruvate dehydrogenase complex activities in muscle were 29% and 14% of the lowest control values, respectively. Pyruvate dehydrogenase complex activity in fibroblasts was normal, however, indicating that the biochemical examination of defects in energy metabolism should be performed in a more energy demanding tissue.


Asunto(s)
Dihidrolipoamida Deshidrogenasa/deficiencia , Dihidrolipoamida Deshidrogenasa/genética , Enfermedad de Leigh/genética , Preescolar , Fibroblastos/enzimología , Heterocigoto , Humanos , Masculino , Músculo Esquelético/enzimología , Mutación Missense , Complejo Piruvato Deshidrogenasa/genética , Sitios de Empalme de ARN
16.
Ann Neurol ; 54(5): 665-9, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14595656

RESUMEN

We describe a novel mutation in the ND6 gene (T14487C) in a patient with Leigh syndrome. Biochemical analyses indicated a low complex I activity in the patient's fibroblasts but normal values in muscle and liver. Cybrid clones showed a specific complex I defect that correlates with the mutant heteroplasmy levels. Additionally, we demonstrate an altered mobility and a decrease in the levels of fully assembled complex I in the patient's fibroblasts and cybrids, suggesting that the mutation has a profound effect on complex I assembly and/or stability.


Asunto(s)
ADN Mitocondrial/genética , Complejo I de Transporte de Electrón/genética , Enfermedad de Leigh/genética , Células Cultivadas , Análisis Mutacional de ADN , Complejo I de Transporte de Electrón/metabolismo , Fibroblastos/fisiología , Humanos , Lactante , Masculino , Mutación Missense , Reacción en Cadena de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA