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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 25(14): 2972-2984, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27206984

RESUMEN

Mutations in PARK2, encoding the E3 ubiquitin protein ligase Parkin, are a common cause of autosomal recessive Parkinson's disease (PD). Loss of PARK2 function compromises mitochondrial quality by affecting mitochondrial biogenesis, bioenergetics, dynamics, transport and turnover. We investigated the impact of PARK2 dysfunction on the endoplasmic reticulum (ER)-mitochondria interface, which mediates calcium (Ca2+) exchange between the two compartments and is essential for Parkin-dependent mitophagy. Confocal and electron microscopy analyses showed the ER and mitochondria to be in closer proximity in primary fibroblasts from PARK2 knockout (KO) mice and PD patients with PARK2 mutations than in controls. Ca2+ flux to the cytosol was also modified, due to enhanced ER-to-mitochondria Ca2+ transfers, a change that was also observed in neurons derived from induced pluripotent stem cells of a patient with PARK2 mutations. Subcellular fractionation showed the abundance of the Parkin substrate mitofusin 2 (Mfn2), which is known to modulate the ER-mitochondria interface, to be specifically higher in the mitochondrion-associated ER membrane compartment in PARK2 KO tissue. Mfn2 downregulation or the exogenous expression of normal Parkin restored cytosolic Ca2+ transients in fibroblasts from patients with PARK2 mutations. In contrast, a catalytically inactive PD-related Parkin variant had no effect. Overall, our data suggest that Parkin is directly involved in regulating ER-mitochondria contacts and provide new insight into the role of the loss of Parkin function in PD development.


Asunto(s)
Retículo Endoplásmico/metabolismo , GTP Fosfohidrolasas/genética , Mitocondrias/metabolismo , Enfermedad de Parkinson/genética , Ubiquitina-Proteína Ligasas/genética , Animales , Señalización del Calcio/genética , Citosol/metabolismo , Retículo Endoplásmico/patología , Fibroblastos , GTP Fosfohidrolasas/biosíntesis , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/patología , Mitofagia/genética , Mutación , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología
2.
Neuropathology ; 38(2): 113-124, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29218765

RESUMEN

Autophagy, the major pathway for protein turnover, is critical to maintain cellular homeostasis and has been implicated in neurodegenerative diseases. The aim of this research was to analyze the expression of autophagy markers in postmortem brains from Machado-Joseph disease (MJD) patients. The expression of autophagy markers in the cerebellum and the oculomotor nucleus from MJD patients and age-matched controls with no signs of neuropathology was inspected postmortem by immunohistochemistry (IHC) and Western blot. Furthermore, autophagy was examined by means of transmission electron microscopy (TEM). Western blot and IHC revealed nuclear accumulation of misfolded ataxin-3 (ATXN3) and the presence of ubiquitin- and p62-positive aggregates in MJD patients as compared to controls. Moreover, the autophagic proteins, autophagy-related gene (Atg) protein (ATG)-7, ATG-12, ATG16L2 and autophagosomal microtubule-associated protein light chain 3 (LC3) were significantly increased in MJD brains relative to controls, while beclin-1 levels were reduced in MJD patients. Increase in the levels of lysosomal-associated membrane protein 2 (LAMP-2) and of the endosomal markers (Rab7 and Rab1A) were observed in MJD patients relatively to controls. In addition, these findings were further confirmed by TEM in brain tissue where large vesicles accumulating electron-dense materials were highly enriched in MJD patients. Postmortem brains with MJD exhibit increased markers of autophagy relative to age-matched control brains, therefore suggesting strong dysregulation of autophagy that may have an important role in the course of MJD pathogenesis.


Asunto(s)
Autofagia , Cerebelo/metabolismo , Enfermedad de Machado-Joseph/metabolismo , Complejo Nuclear Oculomotor/metabolismo , Adulto , Ataxina-3/metabolismo , Beclina-1/metabolismo , Biomarcadores/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Endosomas/metabolismo , Femenino , Proteínas Ligadas a GPI/metabolismo , Humanos , Lisosomas/metabolismo , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sirolimus/metabolismo , Ubiquitina/metabolismo
3.
Neurobiol Dis ; 102: 21-37, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28237315

RESUMEN

Mutations in SPG11 account for the most common form of autosomal recessive hereditary spastic paraplegia (HSP), characterized by a gait disorder associated with various brain alterations. Mutations in the same gene are also responsible for rare forms of Charcot-Marie-Tooth (CMT) disease and progressive juvenile-onset amyotrophic lateral sclerosis (ALS). To elucidate the physiopathological mechanisms underlying these human pathologies, we disrupted the Spg11 gene in mice by inserting stop codons in exon 32, mimicking the most frequent mutations found in patients. The Spg11 knockout mouse developed early-onset motor impairment and cognitive deficits. These behavioral deficits were associated with progressive brain atrophy with the loss of neurons in the primary motor cortex, cerebellum and hippocampus, as well as with accumulation of dystrophic axons in the corticospinal tract. Spinal motor neurons also degenerated and this was accompanied by fragmentation of neuromuscular junctions and muscle atrophy. This new Spg11 knockout mouse therefore recapitulates the full range of symptoms associated with SPG11 mutations observed in HSP, ALS and CMT patients. Examination of the cellular alterations observed in this model suggests that the loss of spatacsin leads to the accumulation of lipids in lysosomes by perturbing their clearance from these organelles. Altogether, our results link lysosomal dysfunction and lipid metabolism to neurodegeneration and pinpoint a critical role of spatacsin in lipid turnover.


Asunto(s)
Metabolismo de los Lípidos/fisiología , Lisosomas/metabolismo , Enfermedad de la Neurona Motora/metabolismo , Proteínas/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Lisosomas/patología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/fisiología , Enfermedad de la Neurona Motora/patología , Neuronas/metabolismo , Neuronas/patología , Fenotipo , Proteínas/genética , Médula Espinal/metabolismo , Médula Espinal/patología
4.
Am J Hum Genet ; 94(2): 268-77, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24388663

RESUMEN

Hereditary spastic paraplegias (HSPs) are clinically and genetically heterogeneous neurological conditions. Their main pathogenic mechanisms are thought to involve alterations in endomembrane trafficking, mitochondrial function, and lipid metabolism. With a combination of whole-genome mapping and exome sequencing, we identified three mutations in REEP2 in two families with HSP: a missense variant (c.107T>A [p.Val36Glu]) that segregated in the heterozygous state in a family with autosomal-dominant inheritance and a missense change (c.215T>A [p.Phe72Tyr]) that segregated in trans with a splice site mutation (c.105+3G>T) in a family with autosomal-recessive transmission. REEP2 belongs to a family of proteins that shape the endoplasmic reticulum, an organelle that was altered in fibroblasts from an affected subject. In vitro, the p.Val36Glu variant in the autosomal-dominant family had a dominant-negative effect; it inhibited the normal binding of wild-type REEP2 to membranes. The missense substitution p.Phe72Tyr, in the recessive family, decreased the affinity of the mutant protein for membranes that, together with the splice site mutation, is expected to cause complete loss of REEP2 function. Our findings illustrate how dominant and recessive inheritance can be explained by the effects and nature of mutations in the same gene. They have also important implications for genetic diagnosis and counseling in clinical practice because of the association of various modes of inheritance to this new clinico-genetic entity.


Asunto(s)
Proteínas de la Membrana/genética , Paraplejía Espástica Hereditaria/genética , Secuencia de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Mapeo Cromosómico , Exoma , Femenino , Heterocigoto , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Mutación Missense , Linaje , Fenotipo , Paraplejía Espástica Hereditaria/patología
5.
Acta Neuropathol ; 128(5): 705-22, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24859968

RESUMEN

There is still no treatment for polyglutamine disorders, but clearance of mutant proteins might represent a potential therapeutic strategy. Autophagy, the major pathway for organelle and protein turnover, has been implicated in these diseases. To determine whether the autophagy/lysosome system contributes to the pathogenesis of spinocerebellar ataxia type 7 (SCA7), caused by expansion of a polyglutamine tract in the ataxin-7 protein, we looked for biochemical, histological and transcriptomic abnormalities in components of the autophagy/lysosome pathway in a knock-in mouse model of the disease, postmortem brain and peripheral blood mononuclear cells (PBMC) from patients. In the mouse model, mutant ataxin-7 accumulated in inclusions immunoreactive for the autophagy-associated proteins mTOR, beclin-1, p62 and ubiquitin. Atypical accumulations of the autophagosome/lysosome markers LC3, LAMP-1, LAMP2 and cathepsin-D were also found in the cerebellum of the SCA7 knock-in mice. In patients, abnormal accumulations of autophagy markers were detected in the cerebellum and cerebral cortex of patients, but not in the striatum that is spared in SCA7, suggesting that autophagy might be impaired by the selective accumulation of mutant ataxin-7. In vitro studies demonstrated that the autophagic flux was impaired in cells overexpressing full-length mutant ataxin-7. Interestingly, the expression of the early autophagy-associated gene ATG12 was increased in PBMC from SCA7 patients in correlation with disease severity. These results provide evidence that the autophagy/lysosome pathway is impaired in neurons undergoing degeneration in SCA7. Autophagy/lysosome-associated molecules might, therefore, be useful markers for monitoring the effects of potential therapeutic approaches using modulators of autophagy in SCA7 and other autophagy/lysosome-associated neurodegenerative disorders.


Asunto(s)
Autofagia/fisiología , Encéfalo/patología , Lisosomas/metabolismo , Lisosomas/patología , Proteínas del Tejido Nervioso/metabolismo , Ataxias Espinocerebelosas/patología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Ataxina-7 , Beclina-1 , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Estudios de Casos y Controles , Línea Celular Transformada , Femenino , Regulación de la Expresión Génica/genética , Humanos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Lisosomas/ultraestructura , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/ultraestructura , Proteínas de Unión a Fosfato , Transducción de Señal/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Ataxias Espinocerebelosas/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Repeticiones de Trinucleótidos/genética
6.
BMC Neurosci ; 14: 135, 2013 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-24192137

RESUMEN

BACKGROUND: Mutations of the gene encoding the major component of Lewy bodies (LB), α-synuclein (α-syn), cause autosomal dominant forms of Parkinson's disease (PD), whereas loss-of-function mutations of the gene encoding the multifunctional E3 ubiquitin-protein ligase Parkin account for autosomal recessive forms of the disease. Parkin overproduction protects against α-syn-dependent neurodegeneration in various in vitro and in vivo models, but it remains unclear whether this process is affected by Parkin deficiency. We addressed this issue, by carrying out more detailed analyses of transgenic mice overproducing the A30P variant of human α-syn (hA30Pα-syn) and with two, one or no parkin knockout alleles. RESULTS: Longitudinal behavioral follow-up of these mice indicated that Parkin depletion delayed disease-predictive sensorimotor impairment due to α-syn accumulation, in a dose-dependent fashion. At the end stage of the disease, neuronal deposits containing fibrillar α-syn species phosphorylated at S129 (PS129α-syn) were the predominant neuropathological feature in hA30Pα-syn mice, regardless of their parkin expression. Some of these deposits colocalized with the LB markers ubiquitin and α-syn truncated at D135 (α-synD135), indicating that PS129α-syn is subjected to secondary posttranslational modification (PTM); these features were not significantly affected by parkin dysfunction. CONCLUSIONS: These findings suggest that Parkin deficiency acts as a protective modifier in α-syn-dependent neurodegeneration, without overtly affecting the composition and characteristics of α-syn deposits in end-stage disease.


Asunto(s)
Encéfalo/patología , Degeneración Nerviosa/genética , Ubiquitina-Proteína Ligasas/genética , alfa-Sinucleína/genética , Animales , Western Blotting , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Humanos , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Destreza Motora , Degeneración Nerviosa/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología
7.
Mol Cell Neurosci ; 47(3): 191-202, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21545838

RESUMEN

Truncating mutations in the SPG11 and SPG15 genes cause complicated spastic paraplegia, severe neurological conditions due to loss of the functions of spatacsin and spastizin, respectively. We developed specific polyclonal anti-spatacsin (SPG11) and anti-spastizin (SPG15) antisera, which we then used to explore the intracellular and tissue localizations of these proteins. We observed expression of both proteins in human and rat central nervous system, which was particularly strong in cortical and spinal motor neurons as well as in retina. Both proteins were also expressed ubiquitously and strongly in embryos. In cultured cells, these two proteins had similar diffuse punctate, cytoplasmic and sometimes nuclear (spastizin) distributions. They partially co-localized with multiple organelles, particularly with protein-trafficking vesicles, endoplasmic reticulum and microtubules. Spastizin was also found at the mitochondria surface. This first study of the endogenous expression of spatacsin and spastizin shows similarities in their expression patterns that could account for their overlapping clinical phenotypes and involvement in a common protein complex.


Asunto(s)
Proteínas Portadoras/metabolismo , Citoplasma/metabolismo , Neuronas Motoras/metabolismo , Proteínas/metabolismo , Animales , Proteínas Portadoras/genética , Línea Celular Tumoral , Células Cultivadas , Citoplasma/genética , Humanos , Ratones , Proteínas/genética , Ratas , Paraplejía Espástica Hereditaria/genética , Paraplejía Espástica Hereditaria/metabolismo
8.
Mov Disord ; 26(9): 1648-56, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21469212

RESUMEN

The locomotor area has recently emerged as a target for deep brain stimulation to lessen gait disturbances in advanced parkinsonian patients. An important step in choosing this target is to define anatomical limits of its 2 components, the pedunculopontine nucleus and the cuneiform nucleus, their connections with the basal ganglia, and their output descending pathway. Based on the hypothesis that pedunculopontine nucleus controls locomotion whereas cuneiform nucleus controls axial posture, we analyzed whether both nuclei receive inputs from the internal pallidum and substantia nigra using anterograde and retrograde tract tracing in monkeys. We also examined whether these nuclei convey descending projections to the reticulospinal pathway. Pallidal terminals were densely distributed and restricted to the pedunculopontine nucleus, whereas nigral terminals were diffusely observed in the whole extent of both the pedunculopontine nucleus and the cuneiform nucleus. Moreover, nigral terminals formed symmetric synapses with pedunculopontine nucleus and cuneiform nucleus dendrites. Retrograde tracing experiments confirmed these results because labeled cell bodies were observed in both the internal pallidum and substantia nigra after pedunculopontine nucleus injection, but only in the substantia nigra after cuneiform nucleus injection. Furthermore, anterograde tracing experiments revealed that the pedunculopontine nucleus and cuneiform nucleus project to large portions of the pontomedullary reticular formation. This is the first anatomical evidence that the internal pallidum and the substantia nigra control different parts of the brain stem and can modulate the descending reticulospinal pathway in primates. These findings support the functional hypothesis that the nigro-cuneiform nucleus pathway could control axial posture whereas the pallido-pedunculopontine nucleus pathway could modulate locomotion.


Asunto(s)
Globo Pálido/fisiología , Vías Nerviosas/fisiología , Formación Reticular/fisiología , Sustancia Negra/fisiología , Animales , Biotina/análogos & derivados , Biotina/metabolismo , Dextranos/metabolismo , Masculino , Microscopía Electrónica de Transmisión/métodos , Vías Nerviosas/metabolismo , Primates , Formación Reticular/anatomía & histología , Aglutinina del Germen de Trigo-Peroxidasa de Rábano Silvestre Conjugada/metabolismo
9.
J Cell Biol ; 174(1): 65-76, 2006 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-16818720

RESUMEN

The pathogenesis of spinocerebellar ataxia type 7 and other neurodegenerative polyglutamine (polyQ) disorders correlates with the aberrant accumulation of toxic polyQ-expanded proteins in the nucleus. Promyelocytic leukemia protein (PML) nuclear bodies are often present in polyQ aggregates, but their relation to pathogenesis is unclear. We show that expression of PML isoform IV leads to the formation of distinct nuclear bodies enriched in components of the ubiquitin-proteasome system. These bodies recruit soluble mutant ataxin-7 and promote its degradation by proteasome-dependent proteolysis, thus preventing the aggregate formation. Inversely, disruption of the endogenous nuclear bodies with cadmium increases the nuclear accumulation and aggregation of mutant ataxin-7, demonstrating their role in ataxin-7 turnover. Interestingly, beta-interferon treatment, which induces the expression of endogenous PML IV, prevents the accumulation of transiently expressed mutant ataxin-7 without affecting the level of the endogenous wild-type protein. Therefore, clastosomes represent a potential therapeutic target for preventing polyQ disorders.


Asunto(s)
Núcleo Celular/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Ataxina-7 , Células COS , Cloruro de Cadmio/farmacología , Células Cultivadas , Chlorocebus aethiops , Humanos , Interferón beta/farmacología , Ratones , Ratones Transgénicos , Complejos Multiproteicos/efectos de los fármacos , Mutación , Proteínas de Neoplasias/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/efectos de los fármacos , Péptidos/genética , Proteína de la Leucemia Promielocítica , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Isoformas de Proteínas/efectos de los fármacos , Isoformas de Proteínas/metabolismo , Factores de Transcripción/efectos de los fármacos , Proteínas Supresoras de Tumor/efectos de los fármacos
10.
Nat Neurosci ; 9(3): 340-8, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16462734

RESUMEN

Vascular endothelial growth factor C (VEGF-C) was first identified as a regulator of the vascular system, where it is required for the development of lymphatic vessels. Here we report actions of VEGF-C in the central nervous system. We detected the expression of the VEGF-C receptor VEGFR-3 in neural progenitor cells in Xenopus laevis and mouse embryos. In Xenopus tadpole VEGF-C knockdowns and in mice lacking Vegfc, the proliferation of neural progenitors expressing VEGFR-3 was severely reduced, in the absence of intracerebral blood vessel defects. In addition, Vegfc-deficient mouse embryos showed a selective loss of oligodendrocyte precursor cells (OPCs) in the embryonic optic nerve. In vitro, VEGF-C stimulated the proliferation of OPCs expressing VEGFR-3 and nestin-positive ventricular neural cells. VEGF-C thus has a new, evolutionary conserved function as a growth factor selectively required by neural progenitor cells expressing its receptor VEGFR-3.


Asunto(s)
Encéfalo/embriología , Diferenciación Celular/fisiología , Factores de Crecimiento Nervioso/metabolismo , Neuronas/metabolismo , Células Madre/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Células Cultivadas , Evolución Molecular , Proteínas de Filamentos Intermediarios/metabolismo , Larva , Ventrículos Laterales/citología , Ventrículos Laterales/embriología , Ventrículos Laterales/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Nestina , Neuronas/citología , Oligodendroglía/citología , Oligodendroglía/metabolismo , Nervio Óptico/citología , Nervio Óptico/embriología , Nervio Óptico/metabolismo , Ratas , Ratas Wistar , Células Madre/citología , Factor C de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Xenopus laevis
11.
J Neurosci ; 28(30): 7624-36, 2008 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-18650339

RESUMEN

White matter axons organize into fascicles that grow over long distances and traverse very diverse environments. The molecular mechanisms preserving this structure of white matter axonal tracts are not well known. Here, we used the optic nerve as a model and investigated the role of TAG-1, a cell adhesion molecule expressed by retinal axons. TAG-1 was first expressed in the embryonic retinal ganglion cells (RGCs) and later in the postnatal myelin-forming cells in the optic nerve. We describe the consequences of genetic loss of Tag-1 on the developing and adult retinogeniculate tract. Tag-1-null embryos display anomalies in the caliber of RGC axons, associated with an abnormal organization of the astroglial network in the optic nerve. The contralateral projections in the lateral geniculate nucleus are expanded postnatally. In the adult, Tag-1-null mice show a loss of RGC axons, with persistent abnormalities of axonal caliber and additional cytoskeleton and myelination defects. Therefore, TAG-1 is an essential regulator of the structure of RGC axons and their surrounding glial cells in the optic nerve.


Asunto(s)
Axones/fisiología , Moléculas de Adhesión Celular Neuronal/fisiología , Vaina de Mielina/metabolismo , Nervio Óptico/metabolismo , Retina/citología , Células Ganglionares de la Retina/citología , Animales , Animales Recién Nacidos , Axones/ultraestructura , Moléculas de Adhesión Celular Neuronal/deficiencia , Células Cultivadas , Contactina 2 , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/fisiología , Complejo de Antígeno L1 de Leucocito/genética , Complejo de Antígeno L1 de Leucocito/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/metabolismo , Nervio Óptico/ultraestructura , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/ultraestructura , Factor de Transcripción Brn-3A/genética , Factor de Transcripción Brn-3A/metabolismo
12.
J Neurochem ; 110(5): 1607-16, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19573020

RESUMEN

We examined the effects of wild-type and mutant atlastin-1 on vesicle transport in the endoplasmic reticulum (ER)-Golgi interface and vesicle budding from ER-derived microsomes using the temperature-sensitive reporter vesicular stomatitis virus glycoprotein (VSV-G), and the ability of purified atlastin-1 to form tubules or vesicles from protein-free phosphatidylserine liposomes. A GTPase domain mutation (T162P) altered the cellular distribution of the ER, but none of the mutations studied significantly affected transport from the ER to the Golgi apparatus. The mutations also had no significant effect on the incorporation of VSV-G into vesicles formed from ER microsomes. Atlastin-1, however, was also incorporated into microsome-derived vesicles, suggesting that it might be implicated in vesicle formation. Purified atlastin-1 transformed phosphatidylserine liposomes into branched tubules and polygonal networks of tubules and vesicles, an action inhibited by GDP and the synthetic dynamin inhibitor dynasore. The GTPase mutations T162P and R217C decreased but did not totally prevent this action; the C-terminal transmembrane domain mutation R495W was as active as the wild-type enzyme. Similar effects were observed in human embryonic kidney cells over-expressing mutant atlastin-1. We concluded that atlastin-1, like dynamin, might be implicated in membrane tubulation and vesiculation and participated in the formation as well as the function of the ER.


Asunto(s)
Vesículas Citoplasmáticas/enzimología , Retículo Endoplásmico/enzimología , GTP Fosfohidrolasas/metabolismo , Lípidos de la Membrana/metabolismo , Microtúbulos/enzimología , Línea Celular , Vesículas Citoplasmáticas/genética , Vesículas Citoplasmáticas/ultraestructura , Dinaminas/genética , Dinaminas/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/ultraestructura , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/fisiología , Proteínas de Unión al GTP , Humanos , Lípidos de la Membrana/genética , Proteínas de la Membrana , Microtúbulos/genética , Microtúbulos/ultraestructura , Transporte de Proteínas/fisiología , Paraplejía Espástica Hereditaria/enzimología , Paraplejía Espástica Hereditaria/genética
13.
J Neurosci ; 27(10): 2483-92, 2007 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-17344386

RESUMEN

Spinocerebellar ataxia 7 (SCA7) is a neurodegenerative disease caused by a polyglutamine (polyQ) expansion in the ataxin 7 (ATXN7) protein, a member of a multiprotein complex involved in histone acetylation. We have created a conditional Drosophila model of SCA7 in which expression of truncated ATXN7 (ATXN7T) with a pathogenic polyQ expansion is induced in neurons in adult flies. In this model, mutant ATXN7T accumulated in neuronal intranuclear inclusions containing ubiquitin, the 19S proteasome subunit, and HSP70 (heat shock protein 70), as in patients. Aggregation was accompanied by a decrease in locomotion and lifespan but limited neuronal death. Disaggregation of the inclusions, when expression of expanded ATXN7T was stopped, correlated with improved locomotor function and increased lifespan, suggesting that the pathology may respond to treatment. Lifespan was then used as a quantitative marker in a candidate gene approach to validate the interest of the model and to identify generic modulators of polyQ toxicity and specific modifiers of SCA7. Several molecular pathways identified in this focused screen (proteasome function, unfolded protein stress, caspase-dependent apoptosis, and histone acetylation) were further studied in primary neuronal cultures. Sodium butyrate, a histone deacetylase inhibitor, improved the survival time of the neurons. This model is therefore a powerful tool for studying SCA7 and for the development of potential therapies for polyQ diseases.


Asunto(s)
Modelos Animales de Enfermedad , Drosophila , Ataxias Espinocerebelosas/genética , Animales , Animales Modificados Genéticamente , Ataxina-7 , Muerte Celular , Células Cultivadas , Discinesias/genética , Glutamina , Humanos , Cuerpos de Inclusión Intranucleares/ultraestructura , Longevidad , Masculino , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Péptidos/genética , Fenotipo , Ratas , Ataxias Espinocerebelosas/metabolismo , Ataxias Espinocerebelosas/patología , Ataxias Espinocerebelosas/fisiopatología , Treonina
14.
J Neurosci ; 27(29): 7827-37, 2007 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-17634376

RESUMEN

A neurodegenerative tauopathy endemic to the Caribbean island of Guadeloupe has been associated with the consumption of anonaceous plants that contain acetogenins, potent lipophilic inhibitors of complex I of the mitochondrial respiratory chain. To test the hypothesis that annonacin, a prototypical acetogenin, contributes to the etiology of the disease, we investigated whether annonacin affects the cellular distribution of the protein tau. In primary cultures of rat striatal neurons treated for 48 h with annonacin, there was a concentration-dependent decrease in ATP levels, a redistribution of tau from the axons to the cell body, and cell death. Annonacin induced the retrograde transport of mitochondria, some of which had tau attached to their outer membrane. Taxol, a drug that displaces tau from microtubules, prevented the somatic redistribution of both mitochondria and tau but not cell death. Antioxidants, which scavenged the reactive oxygen species produced by complex I inhibition, did not affect either the redistribution of tau or cell death. Both were prevented, however, by forced expression of the NDI1 nicotinamide adenine dinucleotide (NADH)-quinone-oxidoreductase of Saccharomyces cerevisiae, which can restore NADH oxidation in complex I-deficient mammalian cells and stimulation of energy production via anaerobic glycolysis. Consistently, other ATP-depleting neurotoxins (1-methyl-4-phenylpyridinium, 3-nitropropionic, and carbonyl cyanide m-chlorophenylhydrazone) reproduced the somatic redistribution of tau, whereas toxins that did not decrease ATP levels did not cause the redistribution of tau. Therefore, the annonacin-induced ATP depletion causes the retrograde transport of mitochondria to the cell soma and induces changes in the intracellular distribution of tau in a way that shares characteristics with some neurodegenerative diseases.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Furanos/farmacología , Lactonas/farmacología , Neuronas/efectos de los fármacos , Proteínas tau/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Encéfalo/citología , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Femenino , Microscopía Inmunoelectrónica/métodos , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/metabolismo , Neuronas/ultraestructura , Paclitaxel/farmacología , Embarazo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Moduladores de Tubulina/farmacología
15.
Brain ; 130(Pt 4): 1062-75, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17347251

RESUMEN

Charcot-Marie-Tooth disease is a genetically heterogeneous group of hereditary motor and sensory neuropathies. Three loci for the axonal autosomal recessive subgroup (ARCMT2) have been reported in 1q21 (CMT2B1, LMNA), 8q21 (CMT4A and CMT2K, GDAP1) and 19q13 (CMT2B2). We report here a clinical, electrophysiological, pathological and genetic study in 13 Moroccan families with ARCMT2 phenotypes. Clinical and electrophysiological examinations were performed in all index cases and 64 'at-risk' relatives. Thirty-one patients were clinically affected. A peroneal nerve biopsy was obtained from three patients. Four families were linked to the 1q21 locus, all had the LMNA R298C mutation. Six families were linked to the 8q21 locus, all had the GDAP1 S194X mutation. Founder effects for both mutations were suggested by the analysis of microsatellite markers close to the genes. The three remaining families were excluded from the three known loci. The electrophysiological findings were consistent with an axonal neuropathy. The clinical data show that in CMT2B1 the disease began most often in the second decade and progressed gradually from distal to proximal muscles. Three of our patients with the longest disease durations (>24 years) had also severe impairment in the scapular muscles. Reported here for the first time, this might be a hallmark of CMT2B1. Patients with CMT4A/2K had onset most often before the age of 2 years. Most had severe clubfoot from the beginning, one of the hallmarks of CMT4A/2K. None of our patients with CMT4A/2K had vocal cord paralysis. The clinical phenotype of the three families that are not linked to the three known loci presented some particularities that were not seen in those with known genetic defects. One family was characterized by late onset of the disease (>20 years) or a mild neuropathy that was diagnosed only when the family was examined. In a second family, dorsal scoliosis was the most prominent symptom. In the third family, symptoms began in the second decade with a moderate neuropathy associated with a pronounced scoliosis. These families illustrate the extent of clinical and genetic heterogeneity in ARCMT2.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Potenciales de Acción/fisiología , Adolescente , Adulto , Axones/fisiología , Enfermedad de Charcot-Marie-Tooth/etnología , Enfermedad de Charcot-Marie-Tooth/fisiopatología , Niño , Preescolar , Consanguinidad , Electromiografía , Femenino , Efecto Fundador , Genes Dominantes/genética , Genotipo , Humanos , Lamina Tipo A/genética , Escala de Lod , Masculino , Marruecos , Mutación/genética , Proteínas del Tejido Nervioso/genética , Conducción Nerviosa/fisiología , Linaje , Nervio Peroneo/patología , Fenotipo
16.
J Neurosci ; 25(16): 4159-68, 2005 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-15843619

RESUMEN

Increased levels of mitochondrial-free calcium have been associated with several cell-death paradigms, such as excitotoxicity and ceramide-mediated neuronal death. In the latter, calcium is transferred from the endoplasmic reticulum to mitochondria by a mechanism that is only partly understood. We show here that CDK5 (cyclin-dependent kinase 5) plays a role. Free calcium levels in the endoplasmic reticulum and mitochondria were measured with fluorescent markers in C2-ceramide-treated primary cultures of mesencephalic neurons and differentiated pheochromocytoma PC12 cells. Calcium levels decreased in the endoplasmic reticulum as they increased in mitochondria. Both changes were blocked by the pharmacological and molecular CDK5 inhibitors roscovitine and a dominant-negative form of CDK5. Although the kinase did not mediate the transfer of calcium per se, which required the proapoptotic Bcl-2 family protein t-Bid (the truncated form of Bid), it facilitated the transfer by inducing the clustering of endoplasmic reticulum and mitochondria around the centrosome where they formed close contacts, as shown by immunocytochemistry and electron microscopy. Organelle clustering resulted from CDK5-dependent phosphorylation of the microtubule-associated protein tau on threonine 231. This caused its release from microtubules into the soluble fraction of cellular proteins, which appears to favor retrograde transport of the organelles. Mutation of threonine 231 to alanine, so that tau could not be phosphorylated at this site, prevented the ceramide-induced release of tau from microtubules, organelle clustering, the increase in mitochondrial-free calcium levels, and neuronal death, demonstrating the importance of the CDK5-dependent signaling cascade in this calcium-dependent cell-death mechanism.


Asunto(s)
Calcio/metabolismo , Quinasa 5 Dependiente de la Ciclina/fisiología , Retículo Endoplásmico/metabolismo , Mitocondrias/metabolismo , Proteínas tau/metabolismo , Análisis de Varianza , Animales , Western Blotting/métodos , Recuento de Células/métodos , Muerte Celular/efectos de los fármacos , Células Cultivadas , Ceramidas/farmacología , Clonación Molecular/métodos , Citocalasina D/farmacología , Interacciones Farmacológicas , Embrión de Mamíferos , Retículo Endoplásmico/ultraestructura , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Compuestos Heterocíclicos con 3 Anillos , Inmunohistoquímica/métodos , Masculino , Mesencéfalo/citología , Microscopía Electrónica de Transmisión/métodos , Mitocondrias/ultraestructura , Neuronas/efectos de los fármacos , Neuronas/fisiología , Neuronas/ultraestructura , Nocodazol/farmacología , Fosforilación , Embarazo , Purinas/farmacología , Ratas , Ratas Wistar , Roscovitina , Factores de Tiempo , Transfección/métodos , Tubulina (Proteína)/metabolismo , Proteínas tau/genética
17.
J Neurosci ; 24(31): 7007-14, 2004 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-15295036

RESUMEN

Although L-dopa remains the most effective treatment of Parkinson disease, its long-term administration is hampered by the appearance of dyskinesia. Hypersensitivity of dopamine D1 receptors in the striatum has been suggested to contribute to the genesis of these delayed adverse effects. However, D1 receptor amounts are unchanged in Parkinson disease, suggesting alterations of downstream effectors. In rodents, striatal D1 receptors activate adenylyl cyclase through olfactory type G-protein alpha subunit (Galphaolf) and G-protein gamma 7 subunit (Ggamma7). We found that Galphaolf was enriched in human basal ganglia and was markedly diminished in the putamen of patients with Huntington disease, in relation with the degeneration of medium spiny neurons. In contrast, in the putamen of patients with Parkinson disease, Galphaolf and Ggamma7 levels were both significantly increased. In the rat, the degeneration of dopamine neurons augmented Galphaolf levels in the striatal neurons, specifically at the plasma membrane, an effect accounting for the increase of D1 response on cAMP production in dopamine-depleted striatum. In lesioned rats, Galphaolf levels were normalized by a 3 week treatment with l-dopa or a D1 agonist but not with aD2-D3 agonist, supporting a Galphaolf regulation by D1 receptor usage. In contrast, the increases of Galphaolf levels in patients were not affected by the duration of l-dopa treatment but correlated with duration of disease. In conclusion, our results revealed in the parkinsonian putamen a prolonged elevation of Galphaolf levels that may lead to a persistent D1 receptor hypersensitivity and contribute to the genesis of long-term complications of L-dopa.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Enfermedad de Parkinson/metabolismo , Putamen/metabolismo , Receptores de Dopamina D1/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Antiparkinsonianos/farmacología , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Femenino , Humanos , Levodopa/farmacología , Masculino , Persona de Mediana Edad , Oxidopamina/farmacología , Enfermedad de Parkinson/patología , Putamen/patología , Ratas , Ratas Sprague-Dawley , Simpaticolíticos/farmacología
18.
Arch Neurol ; 60(4): 598-604, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12707075

RESUMEN

BACKGROUND: The first locus for demyelinating autosomal recessive Charcot-Marie-Tooth (ARCMT) disease was identified in 8q13, where mutations in GDAP1 have been found. Mutations in the same gene have been detected in families with axonal ARCMT disease. OBJECTIVE: To determine the clinical, electrophysiologic, and morphologic characteristics of a consanguineous Moroccan family with ARCMT disease associated with the S194X mutation in the GDAP1 gene. METHODS: Four patients from a consanguineous Moroccan family were examined clinically and electrophysiologically. In one patient, a morphometric and ultrastructural study of a peroneal nerve biopsy sample was performed. Mutation in the coding region of the GDAP1 gene was identified by direct sequencing. RESULTS: Neuropathy was evident early in childhood, walking was delayed in one patient, and onset of symptoms occurred before 18 months in the others. The phenotype was severe: foot deformities and disabilities involving the hands and feet developed toward the end of the first decade, followed by involvement of proximal muscles in the lower limbs, leading to loss of autonomy. Electrophysiologic findings were consistent with an axonal form of CMT disease: motor nerve conduction velocities, recordable in one patient only, were greater than 40 m/sec. Sensory nerve action potentials were either abolished or substantially reduced in amplitude. The morphologic data supported the diagnosis of axonal neuropathy, showing a marked reduction in myelinated fibers and signs of axonal regeneration, including frequent pseudo-onion bulb formations. The 4 patients in this family were homozygous for the S194X mutation in the GDAP1 gene. CONCLUSION: Electrophysiologic and pathological findings support the hypothesis of an axonal disorder in this ARCMT family with the S194X mutation in the GDAP1 gene.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/etnología , Enfermedad de Charcot-Marie-Tooth/genética , Genes Recesivos/genética , Mutación , Adolescente , Adulto , Enfermedad de Charcot-Marie-Tooth/patología , Preescolar , Consanguinidad , Análisis Mutacional de ADN , Electrofisiología , Femenino , Humanos , Lactante , Escala de Lod , Masculino , Marruecos/etnología , Linaje , Fenotipo , Serina/genética
19.
Mov Disord ; 17(6): 1174-9, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12465054

RESUMEN

Levodopa therapy in Parkinson's disease is mediated by dopamine receptors and, in a recent study, we showed that a Dl full agonist can induce an internalization of D1 dopamine receptors. The aim of the present study was to determine whether levodopa or a dopamine agonist such as ropinirole can also induce the internalization of D1 dopamine receptors in the striatum of control and hemiparkinsonian rats. The distribution of D1 dopamine receptors was analyzed by immunohistochemistry using a specific antibody. In control animals and 6-hydroxydopamine (6-OHDA)-lesioned animals treated with saline, D1 dopamine receptors were localized at the level of the plasma membrane. In contrast, in both lesioned and nonlesioned animals receiving a single dose of levodopa, but not in animals receiving ropinirole, D1 dopamine receptors were internalized in the cytoplasm. This result is likely explained by the fact that ropinirole binds to non-D1 dopamine receptors, whereas levodopa, which increases dopamine levels, indirectly acts on both D1 and D2 receptors. Ropinirole is consequently less likely to desensitize D1 dopamine receptors than levodopa and, thus, to reduce the efficacy of the treatment.


Asunto(s)
Antiparkinsonianos/farmacología , Carbidopa/farmacología , Cuerpo Estriado/efectos de los fármacos , Citoplasma/efectos de los fármacos , Indoles/farmacología , Levodopa/farmacología , Trastornos Parkinsonianos/patología , Receptores de Dopamina D1/efectos de los fármacos , Membranas Sinápticas/efectos de los fármacos , Animales , Cuerpo Estriado/patología , Citoplasma/patología , Combinación de Medicamentos , Masculino , Microscopía Inmunoelectrónica , Neuronas/efectos de los fármacos , Neuronas/patología , Oxidopamina , Trastornos Parkinsonianos/inducido químicamente , Ratas , Ratas Sprague-Dawley , Membranas Sinápticas/patología , Tirosina 3-Monooxigenasa/metabolismo
20.
Hum Mol Genet ; 12(5): 517-26, 2003 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-12588799

RESUMEN

Parkin gene mutations have been implicated in autosomal-recessive early-onset parkinsonism and lead to specific degeneration of dopaminergic neurons in midbrain. To investigate the role of Parkin in neuronal cell death, we overproduced this protein in PC12 cells in an inducible manner. In this cell line, neuronally differentiated by nerve growth factor, Parkin overproduction protected against cell death mediated by ceramide, but not by a variety of other cell death inducers (H(2)O(2), 4-hydroxynonenal, rotenone, 6-OHDA, tunicamycin, 2-mercaptoethanol and staurosporine). Protection was abrogated by the proteasome inhibitor epoxomicin and disease-causing variants, indicating that it was mediated by the E3 ubiquitin ligase activity of Parkin. Interestingly, Parkin acted by delaying mitochondrial swelling and subsequent cytochrome c release and caspase-3 activation observed in ceramide-mediated cell death. Subcellular fractionation demonstrated enrichment of Parkin in the mitochondrial fraction and its association with the outer mitochondrial membrane. Together, these results suggest that Parkin may promote the degradation of substrates localized in mitochondria and involved in the late mitochondrial phase of ceramide-mediated cell death. Loss of this function may underlie the degeneration of nigral dopaminergic neurons in patients with Parkin mutations.


Asunto(s)
Grupo Citocromo c/metabolismo , Ligasas/metabolismo , Mitocondrias/metabolismo , Animales , Caspasa 3 , Caspasas/metabolismo , Muerte Celular/fisiología , Ceramidas/metabolismo , Humanos , Neuronas/metabolismo , Ratas , Ubiquitina-Proteína Ligasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA