Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 31(5): 748-760, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-34559225

RESUMEN

Facioscapulohumeral muscular dystrophy (FSHD) is an inherited myopathy clinically characterized by weakness in the facial, shoulder girdle and upper a muscles. FSHD is caused by chromatin relaxation of the D4Z4 macrosatellite repeat, mostly by a repeat contraction, facilitating ectopic expression of DUX4 in skeletal muscle. Genetic diagnosis for FSHD is generally based on the sizing and haplotyping of the D4Z4 repeat on chromosome 4 by Southern blotting (SB), molecular combing or single-molecule optical mapping, which is usually straight forward but can be complicated by atypical rearrangements of the D4Z4 repeat. One of these rearrangements is a D4Z4 proximally extended deletion (DPED) allele, where not only the D4Z4 repeat is partially deleted, but also sequences immediately proximal to the repeat are lost, which can impede accurate diagnosis in all genetic methods. Previously, we identified several DPED alleles in FSHD and estimated the size of the proximal deletions by a complex pulsed-field gel electrophoresis and SB strategy. Here, using the next-generation sequencing, we have defined the breakpoint junctions of these DPED alleles at the base pair resolution in 12 FSHD families and 4 control individuals facilitating a PCR-based diagnosis of these DPED alleles. Our resultsshow that half of the DPED alleles are derivates of an ancient founder allele. For some DPED alleles, we found that genetic elements are deleted such as DUX4c, FRG2, DBE-T and myogenic enhancers necessitating re-evaluation of their role in FSHD pathogenesis.


Asunto(s)
Distrofia Muscular Facioescapulohumeral , Alelos , Cromatina , Cromosomas Humanos Par 4/genética , Efecto Fundador , Humanos , Distrofia Muscular Facioescapulohumeral/genética , Distrofia Muscular Facioescapulohumeral/metabolismo
2.
Eur J Neurol ; 29(7): 2156-2161, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35253317

RESUMEN

BACKGROUND AND PURPOSE: Ataxia and cough are rare features in hereditary sensory and autonomic neuropathies (HSAN), a group of diseases of mostly unknown genetic cause. Biallelic repeat expansions in RFC1 are associated with cerebellar ataxia, neuropathy, and vestibular areflexia syndrome (CANVAS). This study aimed to investigate the prevalence of RFC1 repeat expansions in a cohort of HSAN patients. METHODS: After unremarkable whole-exome sequencing (WES) analysis, we performed repeat-primed PCR to detect intronic RFC1 expansions in 12 HSAN families, who all presented with chronic cough. RESULTS: In these patients, 75% carried biallelic expansions of the pathogenic AAGGG motif. Compared with RFC1-/- cases, RFC1+/+ cases presented more consistently with positive sensory and autonomic symptoms. Afferent ataxia was more severe in the RFC1+/+ cohort and cerebellar ataxia was a common feature (21%). CONCLUSIONS: We demonstrate that RFC1 is a frequent cause of (WES-negative) HSAN with chronic cough and ataxia. The diagnostic yield of RFC1 repeat-primed PCR was surprisingly high, given that HSAN is genetically poorly understood. This combination of HSAN, ataxia, and chronic cough symptoms represents a new nosological entity within the neuropathy-ataxia spectrum.


Asunto(s)
Vestibulopatía Bilateral , Ataxia Cerebelosa , Neuropatías Hereditarias Sensoriales y Autónomas , Enfermedades del Sistema Nervioso Periférico , Enfermedades Vestibulares , Ataxia , Ataxia Cerebelosa/diagnóstico , Ataxia Cerebelosa/genética , Tos/genética , Neuropatías Hereditarias Sensoriales y Autónomas/genética , Humanos , Enfermedades del Sistema Nervioso Periférico/complicaciones
3.
Clin Genet ; 99(3): 449-456, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33340101

RESUMEN

Pathogenic variants in the RBM10 gene cause a rare X-linked disorder described as TARP (Talipes equinovarus, Atrial septal defect, Robin sequence, and Persistent left vena cava superior) syndrome. We report two novel patients with truncating RBM10 variants in view of the literature, presenting a total of 26 patients from 15 unrelated families. Our results illustrate the highly pleiotropic nature of RBM10 pathogenic variants, beyond the classic TARP syndrome features. Major clinical characteristics include severe developmental delay, failure to thrive, brain malformations, neurological symptoms, respiratory issues, and facial dysmorphism. Minor features are growth retardation, cardiac, gastrointestinal, limb, and skeletal abnormalities. Additional recurrent features include genital and renal abnormalities as well as hearing and visual impairment. Thus, RBM10 loss of function variants typically cause an intellectual disability and congenital malformation syndrome that requires assessment of multiple organ systems at diagnosis and for which provided clinical features might simplify diagnostic assessment. Furthermore, evidence for an RBM10-related genotype-phenotype correlation is emerging, which can be important for prognosis.


Asunto(s)
Pie Equinovaro/genética , Estudios de Asociación Genética , Variación Genética , Cardiopatías Congénitas/genética , Discapacidad Intelectual/genética , Malformaciones del Sistema Nervioso/genética , Fenotipo , Síndrome de Pierre Robin/genética , Proteínas de Unión al ARN/genética , Niño , Preescolar , Humanos , Discapacidad Intelectual/diagnóstico , Mutación con Pérdida de Función , Masculino , Malformaciones del Sistema Nervioso/diagnóstico , Pronóstico
4.
J Synchrotron Radiat ; 27(Pt 1): 185-198, 2020 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-31868751

RESUMEN

Friedreich's ataxia (FRDA) is a neurodegenerative disease characterized by an increase in intracytoplasmic iron concentration. Here the nanoscale iron distribution within single fibroblasts from FRDA patients was investigated using synchrotron-radiation-based nanoscopic X-ray fluorescence and X-ray in-line holography at the ID16A nano-imaging beamline of the ESRF. This unique probe was deployed to uncover the iron cellular two-dimensional architecture of freeze-dried FRDA fibroblasts. An unsurpassed absolute detection capability of 180 iron atoms within a 30 nm × 50 nm nanoscopic X-ray beam footprint was obtained using state-of-the-art X-ray focusing optics and a large-solid-angle detection system. Various micrometre-sized iron-rich organelles could be revealed for the first time, tentatively identified as endoplasmic reticulum, mitochondria and lysosomes. Also a multitude of nanoscopic iron hot-spots were observed in the cytosol, interpreted as chaperoned iron within the fibroblast's labile iron pool. These observations enable new hypotheses on the storage and trafficking of iron in the cell and ultimately to a better understanding of iron-storage diseases such as Friedreich's ataxia.


Asunto(s)
Fibroblastos/química , Ataxia de Friedreich/patología , Holografía/métodos , Hierro/análisis , Análisis de la Célula Individual/métodos , Espectrometría por Rayos X/métodos , Carbono , Citoplasma/química , Fibroblastos/ultraestructura , Liofilización , Humanos , Nanoestructuras , Orgánulos/química , Orgánulos/ultraestructura , Análisis de la Célula Individual/instrumentación , Sincrotrones , Fijación del Tejido/métodos
5.
Clin Genet ; 97(3): 426-436, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31721179

RESUMEN

Biallelic MFSD8 variants are an established cause of severe late-infantile subtype of neuronal ceroid lipofuscinosis (v-LINCL), a severe lysosomal storage disorder, but have also been associated with nonsyndromic adult-onset maculopathy. Here, we functionally characterized two novel MFSD8 variants found in a child with juvenile isolated maculopathy, in order to establish a refined prognosis. ABCA4 locus resequencing was followed by the analysis of other inherited retinal disease genes by whole exome sequencing (WES). Minigene assays and cDNA sequencing were used to assess the effect of a novel MFSD8 splice variant. MFSD8 expression was quantified with qPCR and overexpression studies were analyzed by immunoblotting. Transmission electron microscopy (TEM) was performed on a skin biopsy and ophthalmological and neurological re-examinations were conducted. WES revealed two novel MFSD8 variants: c.[590del];[439+3A>C] p.[Gly197Valfs*2];[Ile67Glufs*3]. Characterization of the c.439+3A>C variant via splice assays showed exon-skipping (p.Ile67Glufs*3), while overexpression studies of the corresponding protein indicated expression of a truncated polypeptide. In addition, a significantly reduced MFSD8 RNA expression was noted in patient's lymphocytes. TEM of a skin biopsy revealed typical v-LINCL lipopigment inclusions while neurological imaging of the proband displayed subtle cerebellar atrophy. Functional characterization demonstrated the pathogenicity of two novel MFSD8 variants, found in a child with an initial diagnosis of juvenile isolated maculopathy but likely evolving to v-LINCL with a protracted disease course. Our study allowed a refined neurological prognosis in the proband and expands the natural history of MFSD8-associated disease.


Asunto(s)
Degeneración Macular/genética , Proteínas de Transporte de Membrana/genética , Lipofuscinosis Ceroideas Neuronales/genética , Niño , Femenino , Variación Genética , Homocigoto , Humanos , Degeneración Macular/diagnóstico por imagen , Degeneración Macular/fisiopatología , Microscopía Electrónica de Transmisión , Mutación , Lipofuscinosis Ceroideas Neuronales/fisiopatología , Retina/diagnóstico por imagen , Retina/fisiopatología , Secuenciación del Exoma
6.
Muscle Nerve ; 61(2): 173-181, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31749205

RESUMEN

INTRODUCTION: Small fiber neuropathies (SFN) are associated with a reduction in quality of life. In adults, epidermal nerve fiber density (END) analysis is recommended for the diagnosis of SFN. In children, END assessment is not often performed. We analyzed small nerve fiber innervation to elucidate the potential diagnostic role of skin biopsies in young patients with pain. METHODS: Epidermal nerve fiber density and sudomotor neurite density (SND) were assessed in skin biopsies from 26 patients aged 7 to 20 years (15 female patients) with unexplained chronic pain. The results were compared with clinical data. RESULTS: Epidermal nerve fiber density was abnormal in 50% and borderline in 35% of patients. An underlying medical condition was found in 42% of patients, including metabolic, autoimmune, and genetic disorders. DISCUSSION: Reduction of epidermal nerve fibers can be associated with treatable conditions. Therefore, the analysis of END in children with pain may help to uncover a possible cause and guide potential treatment options.


Asunto(s)
Dolor Crónico/diagnóstico , Dolor Crónico/patología , Fibras Nerviosas/patología , Piel/patología , Neuropatía de Fibras Pequeñas/patología , Adolescente , Biopsia , Niño , Epidermis/inervación , Epidermis/patología , Femenino , Humanos , Masculino , Neuralgia/diagnóstico , Neuritas/patología , Dimensión del Dolor , Glándulas Sudoríparas/inervación , Glándulas Sudoríparas/patología , Adulto Joven
7.
Am J Hum Genet ; 99(1): 217-27, 2016 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-27374774

RESUMEN

Complex I deficiency is the most common biochemical phenotype observed in individuals with mitochondrial disease. With 44 structural subunits and over 10 assembly factors, it is unsurprising that complex I deficiency is associated with clinical and genetic heterogeneity. Massively parallel sequencing (MPS) technologies including custom, targeted gene panels or unbiased whole-exome sequencing (WES) are hugely powerful in identifying the underlying genetic defect in a clinical diagnostic setting, yet many individuals remain without a genetic diagnosis. These individuals might harbor mutations in poorly understood or uncharacterized genes, and their diagnosis relies upon characterization of these orphan genes. Complexome profiling recently identified TMEM126B as a component of the mitochondrial complex I assembly complex alongside proteins ACAD9, ECSIT, NDUFAF1, and TIMMDC1. Here, we describe the clinical, biochemical, and molecular findings in six cases of mitochondrial disease from four unrelated families affected by biallelic (c.635G>T [p.Gly212Val] and/or c.401delA [p.Asn134Ilefs(∗)2]) TMEM126B variants. We provide functional evidence to support the pathogenicity of these TMEM126B variants, including evidence of founder effects for both variants, and establish defects within this gene as a cause of complex I deficiency in association with either pure myopathy in adulthood or, in one individual, a severe multisystem presentation (chronic renal failure and cardiomyopathy) in infancy. Functional experimentation including viral rescue and complexome profiling of subject cell lines has confirmed TMEM126B as the tenth complex I assembly factor associated with human disease and validates the importance of both genome-wide sequencing and proteomic approaches in characterizing disease-associated genes whose physiological roles have been previously undetermined.


Asunto(s)
Alelos , Complejo I de Transporte de Electrón/deficiencia , Proteínas de la Membrana/genética , Enfermedades Mitocondriales/genética , Mutación/genética , Fenotipo , Adolescente , Adulto , Edad de Inicio , Secuencia de Aminoácidos , Niño , Complejo I de Transporte de Electrón/genética , Femenino , Humanos , Lactante , Masculino , Proteínas de la Membrana/química , Persona de Mediana Edad , Linaje , Adulto Joven
9.
Hum Mol Genet ; 24(5): 1420-31, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25398950

RESUMEN

Giant axonal neuropathy (GAN) is a progressive neurodegenerative disease caused by autosomal recessive mutations in the GAN gene resulting in a loss of a ubiquitously expressed protein, gigaxonin. Gene replacement therapy is a promising strategy for treatment of the disease; however, the effectiveness and safety of gigaxonin reintroduction have not been tested in human GAN nerve cells. Here we report the derivation of induced pluripotent stem cells (iPSCs) from three GAN patients with different GAN mutations. Motor neurons differentiated from GAN iPSCs exhibit accumulation of neurofilament (NF-L) and peripherin (PRPH) protein and formation of PRPH aggregates, the key pathological phenotypes observed in patients. Introduction of gigaxonin either using a lentiviral vector or as a stable transgene resulted in normalization of NEFL and PRPH levels in GAN neurons and disappearance of PRPH aggregates. Importantly, overexpression of gigaxonin had no adverse effect on survival of GAN neurons, supporting the feasibility of gene replacement therapy. Our findings demonstrate that GAN iPSCs provide a novel model for studying human GAN neuropathologies and for the development and testing of new therapies in relevant cell types.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Neuropatía Axonal Gigante/genética , Células Madre Pluripotentes Inducidas/citología , Proteínas de Filamentos Intermediarios/genética , Neuronas Motoras/metabolismo , Axones , Diferenciación Celular , Células Cultivadas , Proteínas del Citoesqueleto/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Terapia Genética/métodos , Vectores Genéticos/genética , Neuropatía Axonal Gigante/terapia , Humanos , Filamentos Intermedios/genética , Filamentos Intermedios/metabolismo , Cariotipificación , Lentivirus/genética , Neuronas Motoras/citología , Mutación , Fenotipo
10.
Am J Hum Genet ; 95(6): 708-20, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25434004

RESUMEN

Respiratory chain deficiencies exhibit a wide variety of clinical phenotypes resulting from defective mitochondrial energy production through oxidative phosphorylation. These defects can be caused by either mutations in the mtDNA or mutations in nuclear genes coding for mitochondrial proteins. The underlying pathomechanisms can affect numerous pathways involved in mitochondrial physiology. By whole-exome and candidate gene sequencing, we identified 11 individuals from 9 families carrying compound heterozygous or homozygous mutations in GTPBP3, encoding the mitochondrial GTP-binding protein 3. Affected individuals from eight out of nine families presented with combined respiratory chain complex deficiencies in skeletal muscle. Mutations in GTPBP3 are associated with a severe mitochondrial translation defect, consistent with the predicted function of the protein in catalyzing the formation of 5-taurinomethyluridine (τm(5)U) in the anticodon wobble position of five mitochondrial tRNAs. All case subjects presented with lactic acidosis and nine developed hypertrophic cardiomyopathy. In contrast to individuals with mutations in MTO1, the protein product of which is predicted to participate in the generation of the same modification, most individuals with GTPBP3 mutations developed neurological symptoms and MRI involvement of thalamus, putamen, and brainstem resembling Leigh syndrome. Our study of a mitochondrial translation disorder points toward the importance of posttranscriptional modification of mitochondrial tRNAs for proper mitochondrial function.


Asunto(s)
Acidosis Láctica/genética , Encefalopatías/genética , Cardiomiopatía Hipertrófica/genética , Proteínas de Unión al GTP/genética , Procesamiento Proteico-Postraduccional , Acidosis Láctica/fisiopatología , Secuencia de Aminoácidos , Encéfalo/patología , Encefalopatías/fisiopatología , Cardiomiopatía Hipertrófica/fisiopatología , Línea Celular , Niño , Preescolar , Consanguinidad , Femenino , Fibroblastos , Proteínas de Unión al GTP/metabolismo , Humanos , Lactante , Recién Nacido , Masculino , Datos de Secuencia Molecular , Mutación , Linaje , Biosíntesis de Proteínas , Interferencia de ARN , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Alineación de Secuencia
11.
Mol Genet Metab ; 122(4): 172-181, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29126765

RESUMEN

Mutations in FARS2 are known to cause dysfunction of mitochondrial translation due to deficient aminoacylation of the mitochondrial phenylalanine tRNA. Here, we report three novel mutations in FARS2 found in two patients in a compound heterozygous state. The missense mutation c.1082C>T (p.Pro361Leu) was detected in both patients. The mutations c.461C>T (p.Ala154Val) and c.521_523delTGG (p.Val174del) were each detected in one patient. We report abnormal in vitro aminoacylation assays as a functional validation of the molecular genetic findings. Based on the phenotypic data of previously reported subjects and the two subjects reported here, we conclude that FARS2 deficiency can be associated with two phenotypes: (i) an epileptic phenotype, and (ii) a spastic paraplegia phenotype.


Asunto(s)
Epilepsia/genética , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Fenotipo , Fenilalanina-ARNt Ligasa/deficiencia , Fenilalanina-ARNt Ligasa/genética , Paraplejía Espástica Hereditaria/genética , Adolescente , Aminoacil-ARNt Sintetasas/metabolismo , Aminoacilación , Encéfalo/diagnóstico por imagen , Células Cultivadas , Exoma , Femenino , Fibroblastos/metabolismo , Heterocigoto , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Mitocondrias/enzimología , Mitocondrias/metabolismo , Músculo Esquelético/patología , Mutación Missense/genética , Consumo de Oxígeno , ARN de Transferencia/metabolismo , Análisis de Secuencia de ADN
12.
Hum Mutat ; 37(7): 653-60, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26931382

RESUMEN

Congenital disorders of glycosylation (CDG) arise from pathogenic mutations in over 100 genes leading to impaired protein or lipid glycosylation. ALG1 encodes a ß1,4 mannosyltransferase that catalyzes the addition of the first of nine mannose moieties to form a dolichol-lipid linked oligosaccharide intermediate required for proper N-linked glycosylation. ALG1 mutations cause a rare autosomal recessive disorder termed ALG1-CDG. To date 13 mutations in 18 patients from 14 families have been described with varying degrees of clinical severity. We identified and characterized 39 previously unreported cases of ALG1-CDG from 32 families and add 26 new mutations. Pathogenicity of each mutation was confirmed based on its inability to rescue impaired growth or hypoglycosylation of a standard biomarker in an alg1-deficient yeast strain. Using this approach we could not establish a rank order comparison of biomarker glycosylation and patient phenotype, but we identified mutations with a lethal outcome in the first two years of life. The recently identified protein-linked xeno-tetrasaccharide biomarker, NeuAc-Gal-GlcNAc2 , was seen in all 27 patients tested. Our study triples the number of known patients and expands the molecular and clinical correlates of this disorder.


Asunto(s)
Trastornos Congénitos de Glicosilación/genética , Manosiltransferasas/genética , Mutación , Polisacáridos/metabolismo , Biomarcadores/metabolismo , Trastornos Congénitos de Glicosilación/metabolismo , Femenino , Genes Letales , Glicosilación , Humanos , Masculino , Análisis de Secuencia de ADN , Análisis de Supervivencia
13.
Biochim Biophys Acta ; 1853(2): 285-98, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25450972

RESUMEN

The tight interrelationship between peroxisomes and mitochondria is illustrated by their cooperation in lipid metabolism, antiviral innate immunity and shared use of proteins executing organellar fission. In addition, we previously reported that disruption of peroxisome biogenesis in hepatocytes severely impacts on mitochondrial integrity, primarily damaging the inner membrane. Here we investigated the molecular impairments of the dysfunctional mitochondria in hepatocyte selective Pex5 knockout mice. First, by using blue native electrophoresis and in-gel activity stainings we showed that the respiratory complexes were differentially affected with reduction of complexes I and III and incomplete assembly of complex V, whereas complexes II and IV were normally active. This resulted in impaired oxygen consumption in cultured Pex5(-/-) hepatocytes. Second, mitochondrial DNA was depleted causing an imbalance in the expression of mitochondrial- and nuclear-encoded subunits of the respiratory chain complexes. Third, mitochondrial membranes showed increased permeability and fluidity despite reduced content of the polyunsaturated fatty acid docosahexaenoic acid. Fourth, the affected mitochondria in peroxisome deficient hepatocytes displayed increased oxidative stress. Acute deletion of PEX5 in vivo using adeno-Cre virus phenocopied these effects, indicating that mitochondrial perturbations closely follow the loss of functional peroxisomes in time. Likely to compensate for the functional impairments, the volume of the mitochondrial compartment was increased several folds. This was not driven by PGC-1α but mediated by activation of PPARα, possibly through c-myc overexpression. In conclusion, loss of peroxisomal metabolism in hepatocytes perturbs the mitochondrial inner membrane, depletes mitochondrial DNA and causes mitochondrial biogenesis independent of PGC-1α.


Asunto(s)
ADN Mitocondrial/metabolismo , Hepatocitos/metabolismo , Mitocondrias/metabolismo , Peroxisomas/metabolismo , Factores de Transcripción/metabolismo , Animales , Compartimento Celular , Proliferación Celular , Respiración de la Célula , Transporte de Electrón , Eliminación de Gen , Hepatocitos/ultraestructura , Lípidos/química , Fluidez de la Membrana , Ratones Noqueados , Mitocondrias/ultraestructura , Oxidación-Reducción , Fosforilación Oxidativa , Estrés Oxidativo , PPAR alfa/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Receptor de la Señal 1 de Direccionamiento al Peroxisoma , Subunidades de Proteína/metabolismo , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/metabolismo
14.
Am J Hum Genet ; 92(5): 767-73, 2013 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-23582646

RESUMEN

Hypomyelination with atrophy of the basal ganglia and cerebellum (H-ABC) is a rare hereditary leukoencephalopathy that was originally identified by MRI pattern analysis, and it has thus far defied all attempts at identifying the causal mutation. Only 22 cases are published in the literature to date. We performed exome sequencing on five family trios, two family quartets, and three single probands, which revealed that all eleven H-ABC-diagnosed individuals carry the same de novo single-nucleotide TUBB4A mutation resulting in nonsynonymous change p.Asp249Asn. Detailed investigation of one of the family quartets with the singular finding of an H-ABC-affected sibling pair revealed maternal mosaicism for the mutation, suggesting that rare de novo mutations that are initially phenotypically neutral in a mosaic individual can be disease causing in the subsequent generation. Modeling of TUBB4A shows that the mutation creates a nonsynonymous change at a highly conserved asparagine that sits at the intradimer interface of α-tubulin and ß-tubulin, and this change might affect tubulin dimerization, microtubule polymerization, or microtubule stability. Consistent with H-ABC's clinical presentation, TUBB4A is highly expressed in neurons, and a recent report has shown that an N-terminal alteration is associated with a heritable dystonia. Together, these data demonstrate that a single de novo mutation in TUBB4A results in H-ABC.


Asunto(s)
Ganglios Basales/patología , Cerebelo/patología , Leucoencefalopatías/genética , Modelos Moleculares , Conformación Proteica , Tubulina (Proteína)/genética , Secuencia de Aminoácidos , Secuencia de Bases , Cristalografía por Rayos X , Exoma/genética , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Humanos , Leucoencefalopatías/patología , Imagen por Resonancia Magnética , Masculino , Modelos Genéticos , Datos de Secuencia Molecular , Neuronas/metabolismo , Análisis de Secuencia de ADN , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo
15.
Mol Genet Metab ; 118(3): 185-189, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27233227

RESUMEN

Acyl-CoA dehydrogenase 9 (ACAD9) is a mitochondrial protein involved in oxidative phosphorylation complex I biogenesis. This protein also exhibits acyl-CoA dehydrogenase (ACAD) activity. ACAD9-mutated patients have been reported to suffer from primarily heart, muscle, liver, and nervous system disorders. ACAD9 mutation is suspected in cases of elevated lactic acid levels combined with complex I deficiency, and confirmed by ACAD9 gene analysis. At least 18 ACAD9-mutated patients have previously been reported, usually displaying severe cardiac involvement. We retrospectively studied nine additional patients from three unrelated families with a wide spectrum of cardiac involvement between the families as well as the patients from the same families. All patients exhibited elevated lactate levels. Deleterious ACAD9 mutations were identified in all patients except one for whom it was not possible to recover DNA. To our knowledge, this is one of the first reports on isolated mild ventricular hypertrophy due to ACAD9 mutation in a family with moderate symptoms during adolescence. This report also confirms that dilated cardiomyopathy may occur in conjunction with ACAD9 mutation and that some patients may respond clinically to riboflavin treatment. Of note, several patients suffered from patent ductus arteriosus (PDA), with one exhibiting a complex congenital heart defect. It is yet unknown whether these cardiac manifestations were related to ACAD9 mutation. In conclusion, this disorder should be suspected in the presence of lactic acidosis, complex I deficiency, and any cardiac involvement, even mild.


Asunto(s)
Acil-CoA Deshidrogenasas/genética , Cardiopatías/genética , Ácido Láctico/sangre , Mutación , Acil-CoA Deshidrogenasas/metabolismo , Adulto , Niño , Femenino , Predisposición Genética a la Enfermedad , Cardiopatías/tratamiento farmacológico , Humanos , Lactante , Recién Nacido , Masculino , Linaje , Estudios Retrospectivos , Riboflavina/uso terapéutico , Resultado del Tratamiento , Adulto Joven
16.
Nat Genet ; 39(4): 534-9, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17384640

RESUMEN

Leukoencephalopathy with brain stem and spinal cord involvement and lactate elevation (LBSL) has recently been defined based on a highly characteristic constellation of abnormalities observed by magnetic resonance imaging and spectroscopy. LBSL is an autosomal recessive disease, most often manifesting in early childhood. Affected individuals develop slowly progressive cerebellar ataxia, spasticity and dorsal column dysfunction, sometimes with a mild cognitive deficit or decline. We performed linkage mapping with microsatellite markers in LBSL families and found a candidate region on chromosome 1, which we narrowed by means of shared haplotypes. Sequencing of genes in this candidate region uncovered mutations in DARS2, which encodes mitochondrial aspartyl-tRNA synthetase, in affected individuals from all 30 families. Enzyme activities of mutant proteins were decreased. We were surprised to find that activities of mitochondrial complexes from fibroblasts and lymphoblasts derived from affected individuals were normal, as determined by different assays.


Asunto(s)
Aspartato-ARNt Ligasa/genética , Ligamiento Genético , Ácido Láctico/metabolismo , Mitocondrias/genética , Degeneraciones Espinocerebelosas/genética , Aspartato-ARNt Ligasa/metabolismo , Marcadores Genéticos , Haplotipos , Humanos , Mitocondrias/enzimología , Enfermedades Mitocondriales/genética , Polimorfismo Genético , Degeneraciones Espinocerebelosas/metabolismo
17.
Hum Mutat ; 36(2): 222-31, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25385316

RESUMEN

A homozygous missense mutation (c.822G>C) was found in the gene encoding the mitochondrial asparaginyl-tRNA synthetase (NARS2) in two siblings born to consanguineous parents. These siblings presented with different phenotypes: one had mild intellectual disability and epilepsy in childhood, whereas the other had severe myopathy. Biochemical analysis of the oxidative phosphorylation (OXPHOS) complexes in both siblings revealed a combined complex I and IV deficiency in skeletal muscle. In-gel activity staining after blue native-polyacrylamide gel electrophoresis confirmed the decreased activity of complex I and IV, and, in addition, showed the presence of complex V subcomplexes. Considering the consanguineous descent, homozygosity mapping and whole-exome sequencing were combined revealing the presence of one single missense mutation in the shared homozygous region. The c.822G>C variant affects the 3' splice site of exon 7, leading to skipping of the whole exon 7 and a part of exon 8 in the NARS2 mRNA. In EBV-transformed lymphoblasts, a specific decrease in the amount of charged mt-tRNA(Asn) was demonstrated as compared with controls. This confirmed the pathogenic nature of the variant. To conclude, the reported variant in NARS2 results in a combined OXPHOS complex deficiency involving complex I and IV, making NARS2 a new member of disease-associated aaRS2.


Asunto(s)
Aspartato-ARNt Ligasa/genética , Mutación Missense , Adulto , Aspartato-ARNt Ligasa/metabolismo , Secuencia de Bases , Células Cultivadas , Consanguinidad , Análisis Mutacional de ADN , Femenino , Estudios de Asociación Genética , Homocigoto , Humanos , Masculino , Enfermedades Musculares/genética , Biosíntesis de Proteínas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Sitios de Empalme de ARN
18.
Hum Mol Genet ; 22(13): 2590-602, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23462291

RESUMEN

Two siblings from consanguineous parents died perinatally with a condition characterized by generalized hypotonia, respiratory insufficiency, arthrogryposis, microcephaly, congenital brain malformations and hyperglycinemia. Catalytic activities of the mitochondrial respiratory complexes I and II were deficient in skeletal muscle, a finding suggestive of an inborn error in mitochondrial biogenesis. Homozygosity mapping identified IBA57 located in the largest homozygous region on chromosome 1 as a culprit candidate gene. IBA57 is known to be involved in the biosynthesis of mitochondrial [4Fe-4S] proteins. Sequence analysis of IBA57 revealed the homozygous mutation c.941A > C, p.Gln314Pro. Severely decreased amounts of IBA57 protein were observed in skeletal muscle and cultured skin fibroblasts from the affected subjects. HeLa cells depleted of IBA57 showed biochemical defects resembling the ones found in patient-derived cells, including a decrease in various mitochondrial [4Fe-4S] proteins and in proteins covalently linked to lipoic acid (LA), a cofactor produced by the [4Fe-4S] protein LA synthase. The defects could be complemented by wild-type IBA57 and partially by mutant IBA57. As a result of the mutation, IBA57 protein was excessively degraded, an effect ameliorated by protease inhibitors. Hence, we propose that the mutation leads to partial functional impairment of IBA57, yet the major pathogenic impact is due to its proteolytic degradation below physiologically critical levels. In conclusion, the ensuing lethal complex biochemical phenotype of a novel metabolic syndrome results from multiple Fe/S protein defects caused by a deficiency in the Fe/S cluster assembly protein IBA57.


Asunto(s)
Encefalopatías/genética , Proteínas Portadoras/genética , Enfermedades Musculares/genética , Mutación , Encéfalo/patología , Encefalopatías/diagnóstico , Proteínas Portadoras/metabolismo , Consanguinidad , Análisis Mutacional de ADN , Transporte de Electrón/genética , Femenino , Fibroblastos/metabolismo , Humanos , Recién Nacido , Imagen por Resonancia Magnética , Masculino , Músculo Esquelético/metabolismo , Enfermedades Musculares/diagnóstico , Linaje , Fenotipo , Hermanos , Piel/metabolismo
19.
Anesthesiology ; 122(2): 343-52, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25296107

RESUMEN

BACKGROUND: Propofol is a short-acting intravenous anesthetic agent. In rare conditions, a life-threatening complication known as propofol infusion syndrome can occur. The pathophysiologic mechanism is still unknown. Some studies suggested that propofol acts as uncoupling agent, others suggested that it inhibits complex I or complex IV, or causes increased oxidation of cytochrome c and cytochrome aa3, or inhibits mitochondrial fatty acid metabolism. Although the exact site of interaction is not known, most hypotheses point to the direction of the mitochondria. METHODS: Eight rats were ventilated and sedated with propofol up to 20 h. Sequential biopsy specimens were taken from liver and skeletal muscle and used for determination of respiratory chain activities and propofol concentration. Activities were also measured in skeletal muscle from a patient who died of propofol infusion syndrome. RESULTS: In rats, authors detected a decrease in complex II+III activity starting at low tissue concentration of propofol (20 to 25 µM), further declining at higher concentrations. Before starting anesthesia, the complex II+III/citrate synthase activity ratio in liver was 0.46 (0.25) and in skeletal muscle 0.23 (0.05) (mean [SD]). After 20 h of anesthesia, the ratios declined to 0.17 (0.03) and 0.12 (0.02), respectively. When measured individually, the activities of complexes II and III remained normal. Skeletal muscle from one patient taken in the acute phase of propofol infusion syndrome also shows a selective decrease in complex II+III activity (z-score: -2.96). CONCLUSION: Propofol impedes the electron flow through the respiratory chain and coenzyme Q is the main site of interaction with propofol.


Asunto(s)
Anestésicos Intravenosos/toxicidad , Propofol/toxicidad , Ubiquinona/metabolismo , Animales , Ciclo del Ácido Cítrico/efectos de los fármacos , Transporte de Electrón/efectos de los fármacos , Masculino , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Ratas , Ratas Wistar , Respiración Artificial , Síndrome
20.
J Inherit Metab Dis ; 38(6): 1147-53, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25971455

RESUMEN

Leukodystrophies are a heterogeneous group of severe genetic neurodegenerative disorders. A multiple mitochondrial dysfunctions syndrome was found in an infant presenting with a progressive leukoencephalopathy. Homozygosity mapping, whole exome sequencing, and functional studies were used to define the underlying molecular defect. Respiratory chain studies in skeletal muscle isolated from the proband revealed a combined deficiency of complexes I and II. In addition, western blotting indicated lack of protein lipoylation. The combination of these findings was suggestive for a defect in the iron-sulfur (Fe/S) protein assembly pathway. SNP array identified loss of heterozygosity in large chromosomal regions, covering the NFU1 and BOLA3, and the IBA57 and ABCB10 candidate genes, in 2p15-p11.2 and 1q31.1-q42.13, respectively. A homozygous c.436C > T (p.Arg146Trp) variant was detected in IBA57 using whole exome sequencing. Complementation studies in a HeLa cell line depleted for IBA57 showed that the mutant protein with the semi-conservative amino acid exchange was unable to restore the biochemical phenotype indicating a loss-of-function mutation of IBA57. In conclusion, defects in the Fe/S protein assembly gene IBA57 can cause autosomal recessive neurodegeneration associated with progressive leukodystrophy and fatal outcome at young age. In the affected patient, the biochemical phenotype was characterized by a defect in the respiratory chain complexes I and II and a decrease in mitochondrial protein lipoylation, both resulting from impaired assembly of Fe/S clusters.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Hierro-Azufre/genética , Leucoencefalopatías/diagnóstico , Leucoencefalopatías/genética , Enfermedades Mitocondriales/diagnóstico , Complejo I de Transporte de Electrón/genética , Resultado Fatal , Heterocigoto , Homocigoto , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Mitocondrias/genética , Mutación , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA