Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Acta Neuropathol ; 135(1): 131-148, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28780615

RESUMEN

Mutations in the small heat shock protein B8 gene (HSPB8/HSP22) have been associated with distal hereditary motor neuropathy, Charcot-Marie-Tooth disease, and recently distal myopathy. It is so far not clear how mutant HSPB8 induces the neuronal and muscular phenotypes and if a common pathogenesis lies behind these diseases. Growing evidence points towards a role of HSPB8 in chaperone-associated autophagy, which has been shown to be a determinant for the clearance of poly-glutamine aggregates in neurodegenerative diseases but also for the maintenance of skeletal muscle myofibrils. To test this hypothesis and better dissect the pathomechanism of mutant HSPB8, we generated a new transgenic mouse model leading to the expression of the mutant protein (knock-in lines) or the loss-of-function (functional knock-out lines) of the endogenous protein Hspb8. While the homozygous knock-in mice developed motor deficits associated with degeneration of peripheral nerves and severe muscle atrophy corroborating patient data, homozygous knock-out mice had locomotor performances equivalent to those of wild-type animals. The distal skeletal muscles of the post-symptomatic homozygous knock-in displayed Z-disk disorganisation, granulofilamentous material accumulation along with Hspb8, αB-crystallin (HSPB5/CRYAB), and desmin aggregates. The presence of the aggregates correlated with reduced markers of effective autophagy. The sciatic nerve of the homozygous knock-in mice was characterized by low autophagy potential in pre-symptomatic and Hspb8 aggregates in post-symptomatic animals. On the other hand, the sciatic nerve of the homozygous knock-out mice presented a normal morphology and their distal muscle displayed accumulation of abnormal mitochondria but intact myofiber and Z-line organisation. Our data, therefore, suggest that toxic gain-of-function of mutant Hspb8 aggregates is a major contributor to the peripheral neuropathy and the myopathy. In addition, mutant Hspb8 induces impairments in autophagy that may aggravate the phenotype.


Asunto(s)
Miopatías Distales/metabolismo , Mutación con Ganancia de Función , Proteínas del Choque Térmico HSP20/genética , Proteínas del Choque Térmico HSP20/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miopatías Estructurales Congénitas/metabolismo , Enfermedades del Sistema Nervioso Periférico/metabolismo , Animales , Atrofia/metabolismo , Atrofia/patología , Autofagia/fisiología , Modelos Animales de Enfermedad , Miopatías Distales/patología , Femenino , Proteínas de Choque Térmico , Ratones Transgénicos , Mitocondrias/metabolismo , Mitocondrias/patología , Chaperonas Moleculares , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Miopatías Estructurales Congénitas/patología , Nervio Ciático/metabolismo , Nervio Ciático/patología
3.
Brain ; 139(Pt 6): 1723-34, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27016404

RESUMEN

The most common form of autosomal recessive hereditary spastic paraplegia is caused by mutations in the SPG11/KIAA1840 gene on chromosome 15q. The nature of the vast majority of SPG11 mutations found to date suggests a loss-of-function mechanism of the encoded protein, spatacsin. The SPG11 phenotype is, in most cases, characterized by a progressive spasticity with neuropathy, cognitive impairment and a thin corpus callosum on brain MRI. Full neuropathological characterization has not been reported to date despite the description of >100 SPG11 mutations. We describe here the clinical and pathological features observed in two unrelated females, members of genetically ascertained SPG11 families originating from Belgium and Italy, respectively. We confirm the presence of lesions of motor tracts in medulla oblongata and spinal cord associated with other lesions of the central nervous system. Interestingly, we report for the first time pathological hallmarks of SPG11 in neurons that include intracytoplasmic granular lysosome-like structures mainly in supratentorial areas, and others in subtentorial areas that are partially reminiscent of those observed in amyotrophic lateral sclerosis, such as ubiquitin and p62 aggregates, except that they are never labelled with anti-TDP-43 or anti-cystatin C. The neuropathological overlap with amyotrophic lateral sclerosis, associated with some shared clinical manifestations, opens up new fields of investigation in the physiopathological continuum of motor neuron degeneration.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Neuronas Motoras/patología , Degeneración Nerviosa/patología , Paraplejía Espástica Hereditaria/patología , Adulto , Encéfalo/patología , Femenino , Ganglios Espinales/patología , Humanos , Lisosomas/ultraestructura , Masculino , Bulbo Raquídeo/patología , Persona de Mediana Edad , Mutación , Proteínas/genética , Paraplejía Espástica Hereditaria/diagnóstico , Paraplejía Espástica Hereditaria/diagnóstico por imagen , Médula Espinal/patología
4.
Am J Med Genet A ; 170(7): 1799-805, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27127007

RESUMEN

Shwachman-Diamond syndrome (SDS) is a recessive ribosomopathy, characterized by bone marrow failure and exocrine pancreatic insufficiency (ePI) often associated with neurodevelopmental and skeletal abnormalities. The aim of this report is to describe a SDS patient with early ichthyosis associated with dermal and epidermal intracellular lipid droplets (iLDs), hypoglycemia and later a distinctive clinical SDS phenotype. At 3 months of age, she had ichthyosis, growth retardation, and failure to thrive. She had not cytopenia. Ultrasonography (US) showed pancreatic diffuse high echogenicity. Subsequently fasting hypoketotic hypoglycemia occurred without permanent hepatomegaly or hyperlipidemia. Continuous gavage feeding was followed by clinical improvement including ichthyosis and hypoglycemia. After 14 months of age, she developed persistent neutropenia and ePI consistent with SDS. The ichthyotic skin biopsy, performed at 5 months of age, disclosed iLDs in all epidermal layers, in melanocytes, eccrine sweat glands, Schwann cells and dermal fibroblasts. These iLDs were reminiscent of those described in Dorfman-Chanarin syndrome (DCS) or Wolman's disease. Both LIPA and CGI-58 analysis did not revealed pathogenic mutation. By sequencing SBDS, a compound heterozygous for a previously reported gene mutation (c.258 + 2T>C) and a novel mutation (c.284T>G) were found. Defective SBDS may hypothetically interfere as in DCS, with neutral lipid metabolism and play a role in the SDS phenotype such as ichthyosis with dermal and epidermal iLDs and hypoglycemia. This interference with neutral lipid metabolism must most likely occur in the cytoplasm compartment as in DCS and not in the lysosomal compartment as in Wolman's disease. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Enfermedades de la Médula Ósea/fisiopatología , Insuficiencia Pancreática Exocrina/fisiopatología , Hipoglucemia/fisiopatología , Ictiosis/fisiopatología , Lipomatosis/fisiopatología , Enfermedades de la Médula Ósea/diagnóstico , Enfermedades de la Médula Ósea/metabolismo , Epidermis/metabolismo , Epidermis/patología , Insuficiencia Pancreática Exocrina/diagnóstico , Insuficiencia Pancreática Exocrina/metabolismo , Femenino , Humanos , Hipoglucemia/diagnóstico , Ictiosis/diagnóstico , Ictiosis/metabolismo , Lactante , Gotas Lipídicas/metabolismo , Gotas Lipídicas/patología , Lipomatosis/diagnóstico , Lipomatosis/metabolismo , Fenotipo , Síndrome de Shwachman-Diamond
5.
Gut ; 64(4): 673-83, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24917551

RESUMEN

OBJECTIVE: No therapy for non-alcoholic steatohepatitis (NASH) has been approved so far. Roux-en-y gastric bypass (RYGB) is emerging as a therapeutic option, although its effect on NASH and related hepatic molecular pathways is unclear from human studies. We studied the effect of RYGB on pre-existent NASH and hepatic mitochondrial dysfunction-a key player in NASH pathogenesis-in a novel diet-induced mouse model nicely mimicking human disease. DESIGN: C57BL/6J mice were fed a high-fat high-sucrose diet (HF-HSD). RESULTS: HF-HSD led to early obesity, insulin resistance and hypercholesterolaemia. HF-HSD consistently induced NASH (steatosis, hepatocyte ballooning and inflammation) with fibrosis already after 12-week feeding. NASH was accompanied by hepatic mitochondrial dysfunction, characterised by decreased mitochondrial respiratory chain (MRC) complex I and IV activity, ATP depletion, ultrastructural abnormalities, together with higher 4-hydroxynonenal (HNE) levels, increased uncoupling protein 2 (UCP2) and tumour necrosis factor-α (TNF-α) mRNA and free cholesterol accumulation. In our model of NASH and acquired mitochondrial dysfunction, RYGB induced sustained weight loss, improved insulin resistance and inhibited progression of NASH, with a marked reversal of fibrosis. In parallel, RYGB preserved hepatic MRC complex I activity, restored ATP levels, limited HNE production and decreased TNF-α mRNA. CONCLUSIONS: Progression of NASH and NASH-related hepatic mitochondrial dysfunction can be prevented by RYGB. RYGB preserves respiratory chain complex activity, thereby restoring energy output, probably by limiting the amount of oxidative stress and TNF-α. These data suggest that modulation of hepatic mitochondrial function contributes to the favourable effect of RYBG on established NASH.


Asunto(s)
Derivación Gástrica , Mitocondrias Hepáticas/fisiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/cirugía , Animales , Humanos , Resistencia a la Insulina , Hepatopatías , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Factor de Necrosis Tumoral alfa
6.
Proc Natl Acad Sci U S A ; 107(8): 3858-63, 2010 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-20133711

RESUMEN

Neuronal cytoplasmic and intranuclear aggregates of RNA-binding protein TDP-43 are a hallmark feature of neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). ALS and FTLD show a considerable clinical and pathological overlap and occur as both familial and sporadic forms. Though missense mutations in TDP-43 cause rare forms of familial ALS, it is not yet known whether this is due to loss of TDP-43 function or gain of aberrant function. Moreover, the role of wild-type (WT) TDP-43, associated with the majority of familial and sporadic ALS/FTLD patients, is also currently unknown. Generating homozygous and hemizygous WT human TDP-43 transgenic mouse lines, we show here a dose-dependent degeneration of cortical and spinal motor neurons and development of spastic quadriplegia reminiscent of ALS. A dose-dependent degeneration of nonmotor cortical and subcortical neurons characteristic of FTLD was also observed. Neurons in the affected spinal cord and brain regions showed accumulation of TDP-43 nuclear and cytoplasmic aggregates that were both ubiquitinated and phosphorylated as observed in ALS/FTLD patients. Moreover, the characteristic approximately 25-kDa C-terminal fragments (CTFs) were also recovered from nuclear fractions and correlated with disease development and progression in WT TDP-43 mice. These findings suggest that approximately 25-kDa TDP-43 CTFs are noxious to neurons by a gain of aberrant nuclear function.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Degeneración Lobar Frontotemporal/genética , Cuerpos de Inclusión/metabolismo , Parálisis/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Degeneración Lobar Frontotemporal/metabolismo , Degeneración Lobar Frontotemporal/patología , Humanos , Cuerpos de Inclusión/genética , Ratones , Ratones Transgénicos , Espasticidad Muscular/genética , Espasticidad Muscular/patología , Mutación Missense , Parálisis/patología
7.
Neuropathology ; 32(3): 234-44, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22017321

RESUMEN

Ataxia-telangiectasia (A-T) is classically characterized by progressive neurodegeneration, oculocutaneous telangiectasia, immunodeficiency and elevated α-fetoprotein levels. Some patients, classified as variant A-T, exhibit a milder clinical course. In the latter patients extrapyramidal symptoms, instead of cerebellar ataxia, tend to be the dominating feature and other classical disease hallmarks, like telangiectasia, appear later or even may be absent. Some patients with variant disease have clinically pronounced anterior horn cell degeneration. Neuropathological studies of genetically proven A-T patients are lacking. The aims of our study were to describe the neuropathology of three A-T patients; in two of them the diagnosis was genetically confirmed. The neuropathological findings were compared with those of all known published autopsy findings in A-T patients up to now. Two classical A-T patients aged 19 and 22 and a 33-year-old patient with variant disease were autopsied. In line with previous reports, our patients had severe cerebellar atrophy, less pronounced degeneration of the dentate nucleus and inferior olive, degeneration of the posterior columns and neurogenic muscular atrophy. In addition, all three had anterior horn cell degeneration, which was most prominent at the lumbar level. Compared to the literature, the degenerative changes in the brain stem of the variant A-T patient were somewhat less than anticipated for his age. Degenerative changes in the cerebellum and spinal cord were comparable with those in the literature. Progeric changes were lacking. In conclusion, compared to classical A-T, the variant A-T patient showed essentially the same, only slightly milder neuropathological abnormalities, except for anterior horn degeneration.


Asunto(s)
Ataxia Telangiectasia/patología , Adulto , Proteínas de la Ataxia Telangiectasia Mutada , Autopsia , Carcinoma Hepatocelular/complicaciones , Causas de Muerte , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Sistema Nervioso Central/patología , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Diabetes Mellitus Tipo 2/complicaciones , Progresión de la Enfermedad , Fenómenos Electrofisiológicos , Resultado Fatal , Femenino , Trastornos Neurológicos de la Marcha/etiología , Genotipo , Humanos , Neoplasias Hepáticas/complicaciones , Linfoma no Hodgkin/complicaciones , Masculino , Mioclonía/etiología , Enfermedades Neuromusculares/etiología , Parálisis/etiología , Neoplasias Faríngeas/complicaciones , Fenotipo , Propiocepción/fisiología , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Pruebas de Función Respiratoria , Infecciones del Sistema Respiratorio/complicaciones , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/genética , Adulto Joven
8.
Am J Med Genet A ; 152A(11): 2802-4, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20949505

RESUMEN

Different missense, nonsense and frameshift mutations in the GAN gene encoding gigaxonin have been described to cause giant axonal neuropathy, a severe early-onset progressive neurological disease with autosomal recessive inheritance. By oligonucleotide array CGH analysis, we identified a 57-131 kb microdeletion affecting this gene in a patient with developmental delay, ataxia, areflexia, macrocephaly, and strikingly frizzy hair. The microdeletion was inherited from the mother and mutation analysis revealed a paternally inherited missense mutation c.1456G>A in exon 9 on the other allele. Our findings illustrate the power of higher resolution array CGH studies and highlight the importance of considering copy number variations in autosomal recessive diseases.


Asunto(s)
Deleción Cromosómica , Proteínas del Citoesqueleto/genética , Neuropatía Axonal Gigante/genética , Heterocigoto , Patrón de Herencia/genética , Mutación/genética , Biopsia , Niño , Preescolar , Femenino , Neuropatía Axonal Gigante/patología , Humanos , Lactante , Recién Nacido , Masculino , Piel/patología , Piel/ultraestructura
9.
Neurology ; 90(8): e658-e663, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29352102

RESUMEN

BACKGROUND: The progressive myoclonic epilepsies (PME) are a heterogeneous group of disorders in which a specific diagnosis cannot be made in a subset of patients, despite exhaustive investigation. C9orf72 repeat expansions are emerging as an important causal factor in several adult-onset neurodegenerative disorders, in particular frontotemporal lobar degeneration and amyotrophic lateral sclerosis. An association with PME has not been reported previously. OBJECTIVE: To identify the causative mutation in a Belgian family where the proband had genetically unexplained PME. RESULTS: We report a 33-year old woman who had epilepsy since the age of 15 and then developed progressive cognitive deterioration and multifocal myoclonus at the age of 18. The family history suggested autosomal dominant inheritance of psychiatric disorders, epilepsy, and dementia. Thorough workup for PME including whole exome sequencing did not reveal an underlying cause, but a C9orf72 repeat expansion was found in our patient and affected relatives. Brain biopsy confirmed the presence of characteristic p62-positive neuronal cytoplasmic inclusions. CONCLUSION: C9orf72 mutation analysis should be considered in patients with PME and psychiatric disorders or dementia, even when the onset is in late childhood or adolescence.


Asunto(s)
Proteína C9orf72/genética , Expansión de las Repeticiones de ADN , Epilepsias Mioclónicas Progresivas/genética , Adulto , Edad de Inicio , Encéfalo/patología , Familia , Femenino , Predisposición Genética a la Enfermedad , Humanos , Persona de Mediana Edad , Epilepsias Mioclónicas Progresivas/patología , Epilepsias Mioclónicas Progresivas/fisiopatología , Epilepsias Mioclónicas Progresivas/psicología , Linaje , Fenotipo
10.
Neuromuscul Disord ; 17(6): 433-42, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17434307

RESUMEN

"Cap myopathy" or "cap disease" is a congenital myopathy characterised by cap-like structures at the periphery of muscle fibres, consisting of disarranged thin filaments with enlarged Z discs. Here we report a deletion in the beta-tropomyosin (TPM2) gene causing cap disease in a 36-year-old male patient with congenital muscle weakness, myopathic facies and respiratory insufficiency. The mutation identified in this patient is an in-frame deletion (c.415_417delGAG) of one codon in exon 4 of TPM2 removing a single glutamate residue (p.Glu139del) from the beta-tropomyosin protein. This is expected to disrupt the seven-amino acid repeat essential for making a coiled coil, and thus to impair tropomyosin-actin interaction. Missense mutations in TPM2 have previously been found to cause rare cases of nemaline myopathy and distal arthrogryposis. This mutation is one not previously described and the first genetic cause identified for cap disease.


Asunto(s)
Eliminación de Gen , Heterocigoto , Mutación Missense/genética , Miopatías Estructurales Congénitas/genética , Tropomiosina/genética , Actinas/genética , Actinas/metabolismo , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , Facies , Regulación de la Expresión Génica , Humanos , Masculino , Datos de Secuencia Molecular , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Debilidad Muscular/patología , Miopatías Estructurales Congénitas/metabolismo , Miopatías Estructurales Congénitas/patología , Insuficiencia Respiratoria/patología , Tropomiosina/metabolismo
11.
Arch Neurol ; 63(12): 1787-94, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17172621

RESUMEN

BACKGROUND: Most mutations in the myelin protein zero gene (MPZ) typically cause a severe demyelinating/dysmyelinating neuropathy that begins in infancy or an adult-onset axonal neuropathy. Axonal degeneration in the late-onset H10P mutation may be caused by the disruption of axoglial interaction. OBJECTIVE: To evaluate sural nerve biopsy samples from a patient with early-onset Charcot-Marie-Tooth disease type 1B caused by an arg69-to-cys (R69C) mutation. DESIGN AND PARTICIPANTS: Biopsies of sural nerves were performed 20 years apart in a patient with an R69C mutation (early onset). In addition, peripheral nerves were obtained from autopsy material from a patient with a T95M mutation (late onset). These nerves were analyzed using light microscopy of semithin sections, teased nerve fiber immunohistochemical analysis, electron microscopy, and immunologic electron microscopy. MAIN OUTCOME MEASURES: Pathological changes in sural nerve. RESULTS: Both R69C biopsy samples showed prominent demyelination and onion bulb formation, unlike the late-onset T95M mutation, which showed primarily axonal degeneration with no onion bulbs. The sural biopsy sample obtained 20 years earlier from the R69C patient showed minimal difference from the present sample, consistent with the lack of clinical progression during the 2 decades. Teased fiber immunohistochemical analysis of R69C revealed voltage-gated sodium channel subtype 1.8 expressions at the nodes of Ranvier around the areas of segmental demyelination. Internodal length in all R69C nerve fibers was invariably short (>94% of all internodes are <150 mum). CONCLUSIONS: Morphologic abnormalities in this early-onset R69C neuropathy were severe in childhood but progressed very slowly after adolescence. The switch to voltage-gated sodium channel subtype 1.8 expression at the nodes may provide clues into the pathogenesis of this case of early-onset neuropathy, and the short internodes may contribute to the extremely slowed conduction velocities in this case (<10 m/s).


Asunto(s)
Canales Iónicos/fisiología , Proteína P0 de la Mielina/genética , Vaina de Mielina/fisiología , Anciano de 80 o más Años , Sustitución de Aminoácidos , Axones/patología , Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/fisiopatología , Progresión de la Enfermedad , Femenino , Humanos , Inmunohistoquímica , Activación del Canal Iónico/fisiología , Canales Iónicos/metabolismo , Masculino , Microscopía Electrónica , Microscopía Inmunoelectrónica , Persona de Mediana Edad , Fibras Nerviosas/patología , Nervio Sural/patología , Nervio Cubital/patología
12.
Neuromuscul Disord ; 16(1): 19-25, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16373086

RESUMEN

Congenital insensitivity to pain with anhidrosis or hereditary sensory and autonomic neuropathy type IV (HSAN IV) is the first human genetic disorder implicated in the neurotrophin signal transduction pathway. HSAN IV is characterized by absence of reaction to noxious stimuli, recurrent episodes of fever, anhidrosis, self-mutilating behavior and often mental retardation. Mutations in the neurotrophic tyrosine kinase, receptor, type 1 (NTRK1) are associated with this disorder. Here we report four homozygous mutations, two frameshift (p.Gln626fsX6 and p.Gly181fsX58), one missense (p.Arg761Trp) and one splice site (c.359+5G>T) mutation in four HSAN IV patients. The splice site mutation caused skipping of exons 2 and 3 in patient's mRNA resulting in an in-frame deletion of the second leucine-rich motif. NTRK1 mutations are only rarely reported in the European population. This report extends the spectrum of NTRK1 mutations observed in patients diagnosed with HSAN IV.


Asunto(s)
Mutación del Sistema de Lectura , Neuropatías Hereditarias Sensoriales y Autónomas/genética , Sitios de Empalme de ARN/genética , Receptor trkA/genética , Estudios de Casos y Controles , Niño , Preescolar , Análisis Mutacional de ADN/métodos , Exones , Femenino , Neuropatías Hereditarias Sensoriales y Autónomas/patología , Neuropatías Hereditarias Sensoriales y Autónomas/fisiopatología , Humanos , Lactante , Desequilibrio de Ligamiento , Masculino , Microscopía Electrónica de Transmisión/métodos , Mutación Missense/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Piel/patología , Piel/ultraestructura
14.
Mitochondrion ; 27: 32-8, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26855408

RESUMEN

Megaconial congenital muscular dystrophy is a disease caused by pathogenic mutations in the gene encoding choline kinase beta (CHKB). Microscopically, the disease is hallmarked by the presence of enlarged mitochondria at the periphery of skeletal muscle fibres leaving the centre devoid of mitochondria. Clinical characteristics are delayed motor development, intellectual disability and dilated cardiomyopathy in half of reported cases. This study describes a patient presenting with the cardinal clinical features, in whom a homozygous nonsense mutation (c.248_249insT; p.Arg84Profs*209) was identified in CHKB and who was treated by heart transplantation. Microscopic evaluation of skeletal and heart muscles typically showed enlarged mitochondria. Spectrophotometric evaluation in both tissues revealed a mild decrease of all OXPHOS complexes. Using BN-PAGE analysis followed by activity staining subcomplexes of complex V were detected in both tissues, indicating incomplete complex V assembly. Mitochondrial DNA content was not depleted in analysed tissues. This is the first report describing the microscopic and biochemical abnormalities in the heart from an affected patient. A likely hypothesis is that the biochemical findings are caused by an abnormal lipid profile in the inner mitochondrial membrane resulting from a defective choline kinase B activity.


Asunto(s)
Colina Quinasa/genética , Codón sin Sentido , Membranas Mitocondriales/fisiología , Miopatías Mitocondriales/patología , Distrofias Musculares/patología , Miocardio/patología , Adenosina Trifosfatasas/análisis , Proteínas Portadoras/análisis , Niño , Trasplante de Corazón , Humanos , Masculino , Proteínas de la Membrana/análisis , Microscopía , Mitocondrias/patología , Miopatías Mitocondriales/diagnóstico , Miopatías Mitocondriales/genética , Miopatías Mitocondriales/terapia , ATPasas de Translocación de Protón Mitocondriales , Distrofias Musculares/diagnóstico , Distrofias Musculares/genética , Distrofias Musculares/terapia , Fosforilación Oxidativa
15.
Neurology ; 87(17): 1777-1786, 2016 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-27664989

RESUMEN

OBJECTIVE: To characterize the clinical and MRI features of 2 families with adult-onset dominant leukoencephalopathy and strokes and identify the underlying genetic cause. METHODS: We applied MRI pattern recognition, whole-exome sequencing, and neuropathology. RESULTS: Based on brain imaging, 13 family members of 40 years or older from 2 families were diagnosed with the disease; in 11 family members of the same age, MRI was normal. In the affected family members, MRI showed a leukoencephalopathy that was disproportionately severe compared to the clinical disease. The clinical picture was dominated by ischemic and hemorrhagic strokes, slow and late cognitive deterioration, and therapy-resistant hypertension. With whole-exome sequencing, we identified one variant shared by both families and segregating with the disease: c.973C>T in CTSA. Haplotype analysis revealed a shared 1,145-kb interval encompassing the CTSA variant on chromosome 20q13.12, suggesting a common ancestor. Brain autopsy of 3 patients showed a leukoencephalopathy that was disproportionately extensive compared to the vascular abnormalities. CTSA encodes cathepsin A. Recessive CTSA mutations cause galactosialidosis. One of the numerous cathepsin A functions is to degrade endothelin-1. In the patients, striking endothelin-1 immunoreactivity was found in white matter astrocytes, correlating with increased numbers of premyelinating oligodendrocyte progenitors. This finding supports a role for endothelin-1 in the leukoencephalopathy through inhibition of oligodendrocyte progenitor maturation. CONCLUSIONS: CARASAL (cathepsin A-related arteriopathy with strokes and leukoencephalopathy) is a novel hereditary adult-onset cerebral small vessel disease. It is of interest that, next to the cerebral vascular abnormalities, endothelin-1 may have a role in the pathogenesis of the extensive leukoencephalopathy.


Asunto(s)
Hemorragia , Leucoencefalopatías , Accidente Cerebrovascular , Malformaciones Vasculares , Adulto , Anciano , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Encéfalo/ultraestructura , Catepsina A/genética , Endotelinas/metabolismo , Salud de la Familia , Femenino , Estudio de Asociación del Genoma Completo , Hemorragia/complicaciones , Hemorragia/diagnóstico por imagen , Hemorragia/genética , Humanos , Leucoencefalopatías/complicaciones , Leucoencefalopatías/diagnóstico por imagen , Leucoencefalopatías/genética , Imagen por Resonancia Magnética , Masculino , Repeticiones de Microsatélite , Persona de Mediana Edad , Mutación/genética , Examen Neurológico , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/diagnóstico por imagen , Accidente Cerebrovascular/genética , Malformaciones Vasculares/complicaciones , Malformaciones Vasculares/diagnóstico por imagen , Malformaciones Vasculares/genética
16.
Neurol Genet ; 2(5): e102, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27668283

RESUMEN

OBJECTIVE: To investigate the molecular basis of a Belgian family with autosomal recessive adult-onset neuronal ceroid lipofuscinosis (ANCL or Kufs disease [KD]) with pronounced frontal lobe involvement and to expand the findings to a cohort of unrelated Belgian patients with frontotemporal dementia (FTD). METHODS: Genetic screening in the ANCL family and FTD cohort (n = 461) was performed using exome sequencing and targeted massive parallel resequencing. RESULTS: We identified a homozygous mutation (p.Ile404Thr) in the Cathepsin F (CTSF) gene cosegregating in the ANCL family. No other mutations were found that could explain the disease in this family. All 4 affected sibs developed motor symptoms and early-onset dementia with prominent frontal features. Two of them evolved to akinetic mutism. Disease presentation showed marked phenotypic variation with the onset ranging from 26 to 50 years. Myoclonic epilepsy in one of the sibs was suggestive for KD type A, while epilepsy was not present in the other sibs who presented with clinical features of KD type B. In a Belgian cohort of unrelated patients with FTD, the same heterozygous p.Arg245His mutation was identified in 2 patients who shared a common haplotype. CONCLUSIONS: A homozygous CTSF mutation was identified in a recessive ANCL pedigree. In contrast to the previous associations of CTSF with KD type B, our findings suggest that CTSF genetic testing should also be considered in patients with KD type A as well as in early-onset dementia with prominent frontal lobe and motor symptoms.

17.
Mol Neurobiol ; 48(1): 22-35, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23475610

RESUMEN

Mutations in TAR DNA-binding protein 43 (TDP-43) are associated with familial forms of amyotrophic lateral sclerosis (ALS), while wild-type TDP-43 is a pathological hallmark of patients with sporadic ALS and frontotemporal lobar degeneration (FTLD). Various in vitro and in vivo studies have also demonstrated toxicity of both mutant and wild-type TDP-43 to neuronal cells. To study the potential additional toxicity incurred by mutant TDP-43 in vivo, we generated mutant human TDP-43 (p.M337V) transgenic mouse lines driven by the Thy-1.2 promoter (Mt-TAR) and compared them in the same experimental setting to the disease phenotype observed in wild-type TDP-43 transgenic lines (Wt-TAR) expressing comparable TDP-43 levels. Overexpression of mutant TDP-43 leads to a worsened dose-dependent disease phenotype in terms of motor dysfunction, neurodegeneration, gliosis, and development of ubiquitin and phosphorylated TDP-43 pathology. Furthermore, we show that cellular aggregate formation or accumulation of TDP-43 C-terminal fragments (CTFs) are not primarily responsible for development of the observed disease phenotype in both mutant and wild-type TDP-43 mice.


Asunto(s)
Sustitución de Aminoácidos/genética , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Animales , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Eosinófilos/metabolismo , Eosinófilos/ultraestructura , Gliosis/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Mutantes/metabolismo , Neuronas/patología , Neuronas/ultraestructura , Fenotipo , Fosforilación , Procesamiento Proteico-Postraduccional , Médula Espinal/patología , Médula Espinal/ultraestructura , Transgenes/genética , Proteínas Ubiquitinadas/metabolismo
18.
Clin Neurol Neurosurg ; 115(7): 1088-93, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23219219

RESUMEN

OBJECTIVE: In the Belgian Fabry Study (BeFaS), the prevalence of Fabry disease was assessed in 1000 young patients presenting with stroke, unexplained white matter lesions or vertebrobasilar dolichoectasia. The results of the BeFaS suggested that Fabry disease may play a role in up to 1% of young patients presenting with cerebrovascular disease. However, the clinical relevance was unclear in all cases. We report on detailed phenotyping in subjects identified with α-galactosidase A (α-Gal A) enzyme deficiency or GLA mutations identified in the BeFaS (n=10), and on the results of family screening in this population. METHODS: Family screening was performed to identify additional mutation carriers. Biochemical and/or clinical evaluation of all subjects (BeFaS index patients and relatives carrying a GLA mutation) was performed. RESULTS: Genetic family screening revealed 18 additional GLA mutation carriers. Bloodspot α-Gal A enzyme activity was normal in all GLA mutation carriers, even in 2 males with the p.A143T mutation. Plasma Gb3 and lyso-Gb3 levels were normal in all subjects. Elevated Gb3 in urine was detected in 2 subjects. Some classic clinical signs of Fabry disease, like angiokeratoma or cornea verticillata, could not be detected in our population. Cardiac symptoms of Fabry disease were found in 6 out of 10 p.A143T carriers. No signs of cerebrovascular disease were found in the relatives with a GLA mutation. CONCLUSIONS: We could not identify mutations causing the classical clinical phenotype of Fabry disease in our cerebrovascular disease population. Enzyme activity analysis in bloodspots and plasma may fail to identify late-onset variants of Fabry disease. We recommend genetic testing when an atypical, late-onset variant of Fabry disease is suspected in a male cerebrovascular disease patient. However, this may lead to the identification of non-disease causing or controversial genetic variants.


Asunto(s)
Enfermedad de Fabry/genética , Mutación/genética , Accidente Cerebrovascular/genética , alfa-Galactosidasa/genética , Adulto , Bélgica/epidemiología , Ecocardiografía , Electrocardiografía , Enfermedad de Fabry/epidemiología , Femenino , Pruebas Genéticas , Glucolípidos/sangre , Glucolípidos/orina , Humanos , Masculino , Mutación/fisiología , Fenotipo , Piel/patología , Esfingolípidos/sangre , Esfingolípidos/orina , Accidente Cerebrovascular/epidemiología , Trihexosilceramidas/sangre , Trihexosilceramidas/orina , Insuficiencia Vertebrobasilar/patología , Adulto Joven , alfa-Galactosidasa/sangre , alfa-Galactosidasa/orina
19.
Histol Histopathol ; 28(8): 1041-54, 2013 08.
Artículo en Inglés | MEDLINE | ID: mdl-23479431

RESUMEN

Tubular aggregates (TAs) are aggregates of densely packed tubules in human skeletal muscle fibers with particular histochemical and ultrastructural features that most probably arise from the sarcoplasmic reticulum. Some studies have shown an additional mitochondrial origin of TAs. We studied the histopathological spectrum and clinical features in a large cohort of patients with TAs in their muscle biopsy (106 biopsies), derived from our muscle biopsy archive (15,412 biopsies in total). In particular, we examined light microscopic, enzyme histochemical, immunohistochemical and ultrastructural features in the muscle biopsies, as well as the patients' clinical data. We found TAs in 0.5% of all muscle biopsies. Based on the size of TAs, we identified two sub-groups: (1) myopathies with large TAs (29 biopsies) in type 2 fibers and sometimes also in type 1 fibers, absence of any other associated disorder, and a familial history in half of the cases, and (2) myopathies with small TAs (77 biopsies), exclusively in type 2 fibers, presence of another associated disease in the majority of patients and mostly no familial history. In the sub-group with large TAs, we observed a high variability of ultrastructural changes. The most frequent clinical symptom in both groups was limb muscle weakness. No significant differences in clinical presentation, age at onset or disease duration at the time of biopsy were found between the two groups. In conclusion, myopathies with TAs can be sub-divided into a group with large TAs, probably corresponding to the so-called primary TA myopathies, and into a group with small TAs as a feature of another underlying condition.


Asunto(s)
Enfermedades Musculares/patología , Adolescente , Adulto , Edad de Inicio , Anciano , Biopsia/métodos , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/patología , Músculo Esquelético/ultraestructura , Linaje , Retículo Sarcoplasmático/metabolismo , Adulto Joven
20.
Neuromuscul Disord ; 22(8): 699-711, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22595202

RESUMEN

Missense mutations in the small heat shock protein HSPB8 cause distal hereditary motor neuropathy (dHMN) and axonal Charcot-Marie-Tooth disease (CMT2L). We previously demonstrated that, despite the ubiquitous expression of HSPB8, motor neurons appear to be predominantly affected by HSPB8 mutations. Here, we studied the effect of mutant HSPB8 in primary fibroblast cultures derived from dHMN patients' skin biopsy. In early passage cultures, we observed in all patients' fibroblasts HSPB8 protein aggregates that were not detected in control cells. After applying heat shock stress on the patients' early passage cultured cells, the protein aggregates coalesced into larger formations, while in control cells a homogenous upregulation of HSPB8 protein expression was seen. We also found a reduction in the mitochondrial membrane potential in the early passage cultures. After three months in culture, the number of cells with aggregates had become indistinguishable from that in controls and the mitochondrial membrane potential had returned to normal. These results emphasize the possible drawbacks of using patients' non-neuronal cells to study neuropathological disease mechanisms.


Asunto(s)
Fibroblastos/fisiología , Proteínas de Choque Térmico/genética , Potencial de la Membrana Mitocondrial/fisiología , Enfermedad de la Neurona Motora/patología , Enfermedad de la Neurona Motora/fisiopatología , Mutación Missense/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas/metabolismo , Adulto , Anciano , Apoptosis , Axones/patología , Biomarcadores/metabolismo , Biopsia , Células Cultivadas , Femenino , Fibroblastos/patología , Humanos , Masculino , Persona de Mediana Edad , Chaperonas Moleculares , Unión Proteica , Piel/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA