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












Base de datos
Intervalo de año de publicación
1.
Neurobiol Dis ; 82: 495-503, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26392287

RESUMEN

Diminished lysosomal function can lead to abnormal cellular accumulation of specific proteins, including α-synuclein, contributing to disease pathogenesis of vulnerable neurons in Parkinson's disease (PD) and related α-synucleinopathies. GBA1 encodes for the lysosomal hydrolase glucocerebrosidase (GCase), and mutations in GBA1 are a prominent genetic risk factor for PD. Previous studies showed that in sporadic PD, and in normal aging, GCase brain activity is reduced and levels of corresponding glycolipid substrates are increased. The present study tested whether increasing GCase through AAV-GBA1 intra-cerebral gene delivery in two PD rodent models would reduce the accumulation of α-synuclein and protect midbrain dopamine neurons from α-synuclein-mediated neuronal damage. In the first model, transgenic mice overexpressing wildtype α-synuclein throughout the brain (ASO mice) were used, and in the second model, a rat model of selective dopamine neuron degeneration was induced by AAV-A53T mutant α-synuclein. In ASO mice, intra-cerebral AAV-GBA1 injections into several brain regions increased GCase activity and reduced the accumulation of α-synuclein in the substantia nigra and striatum. In rats, co-injection of AAV-GBA1 with AAV-A53T α-synuclein into the substantia nigra prevented α-synuclein-mediated degeneration of nigrostriatal dopamine neurons by 6 months. These neuroprotective effects were associated with altered protein expression of markers of autophagy. These experiments demonstrate, for the first time, the neuroprotective effects of increasing GCase against dopaminergic neuron degeneration, and support the development of therapeutics targeting GCase or other lysosomal genes to improve neuronal handling of α-synuclein.


Asunto(s)
Neuronas Dopaminérgicas/enzimología , Terapia Genética/métodos , Glucosilceramidasa/genética , Mesencéfalo/enzimología , Enfermedades Neurodegenerativas/terapia , alfa-Sinucleína/metabolismo , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Dopamina/metabolismo , Neuronas Dopaminérgicas/patología , Femenino , Vectores Genéticos , Glucosilceramidasa/metabolismo , Humanos , Masculino , Mesencéfalo/patología , Ratones Transgénicos , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/patología , Ratas Sprague-Dawley , alfa-Sinucleína/genética
2.
Ann Neurol ; 78(4): 568-83, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26174152

RESUMEN

OBJECTIVE: A noncoding hexanucleotide repeat expansion in C9orf72 is the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). It has been reported that the repeat expansion causes a downregulation of C9orf72 transcripts, suggesting that haploinsufficiency may contribute to disease pathogenesis. Two protein isoforms are generated from three alternatively spliced transcripts of C9orf72; a long form (C9-L) and a short form (C9-S), and their function(s) are largely unknown owing to lack of specific antibodies. METHODS: To investigate C9orf72 protein properties, we developed novel antibodies that recognize either C9-L or C9-S. Multiple techniques, including Western blot, immunohistochemistry, and coimmunoprecipitation, were used to determine the expression levels and subcellular localizations of C9-L and C9-S. RESULTS: Investigation of expression of C9-L and C9-S demonstrated distinct biochemical profiles, region-specific changes, and distinct subcellular localizations in ALS tissues. In particular, C9-L antibody exhibited a diffuse cytoplasmic staining in neurons and labeled large speckles in cerebellar Purkinje cells. In contrast, C9-S antibody gave very specific labeling of the nuclear membrane in healthy neurons, with apparent relocalization to the plasma membrane of diseased motor neurons in ALS. Coimmunoprecipitation experiments revealed an interaction of the C9-isoforms with both Importin ß1 and Ran-GTPase, components of the nuclear pore complex. INTERPRETATION: Using these antibodies, we have shown that C9orf72 may be involved in nucleocytoplasmic shuttling and this may have relevance to pathophysiology of ALS/FTLD. Our antibodies have provided improved detection of C9orf72 protein isoforms, which will help elucidate its physiological function and role in ALS/FTLD.


Asunto(s)
Esclerosis Amiotrófica Lateral/diagnóstico , Anticuerpos/análisis , Proteínas/análisis , Anciano , Anciano de 80 o más Años , Secuencia de Aminoácidos , Esclerosis Amiotrófica Lateral/metabolismo , Anticuerpos/metabolismo , Proteína C9orf72 , Cerebelo/química , Cerebelo/metabolismo , Cerebelo/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Neuronas Motoras/química , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Isoformas de Proteínas/análisis , Isoformas de Proteínas/biosíntesis , Proteínas/metabolismo
3.
Antioxid Redox Signal ; 23(6): 550-64, 2015 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-26094487

RESUMEN

AIMS: Loss-of-function mutations in GBA1, which cause the autosomal recessive lysosomal storage disease, Gaucher disease (GD), are also a key genetic risk factor for the α-synucleinopathies, including Parkinson's disease (PD) and dementia with Lewy bodies. GBA1 encodes for the lysosomal hydrolase glucocerebrosidase and reductions in this enzyme result in the accumulation of the glycolipid substrates glucosylceramide and glucosylsphingosine. Deficits in autophagy and lysosomal degradation pathways likely contribute to the pathological accumulation of α-synuclein in PD. In this report we used conduritol-ß-epoxide (CBE), a potent selective irreversible competitive inhibitor of glucocerebrosidase, to model reduced glucocerebrosidase activity in vivo, and tested whether sustained glucocerebrosidase inhibition in mice could induce neuropathological abnormalities including α-synucleinopathy, and neurodegeneration. RESULTS: Our data demonstrate that daily systemic CBE treatment over 28 days caused accumulation of insoluble α-synuclein aggregates in the substantia nigra, and altered levels of proteins involved in the autophagy lysosomal system. These neuropathological changes were paralleled by widespread neuroinflammation, upregulation of complement C1q, abnormalities in synaptic, axonal transport and cytoskeletal proteins, and neurodegeneration. INNOVATION: A reduction in brain GCase activity has been linked to sporadic PD and normal aging, and may contribute to the susceptibility of vulnerable neurons to degeneration. This report demonstrates that systemic reduction of GCase activity using chemical inhibition, leads to neuropathological changes in the brain reminiscent of α-synucleinopathy. CONCLUSIONS: These data reveal a link between reduced glucocerebrosidase and the development of α-synucleinopathy and pathophysiological abnormalities in mice, and support the development of GCase therapeutics to reduce α-synucleinopathy in PD and related disorders.


Asunto(s)
Complemento C1q/metabolismo , Glucosilceramidasa/antagonistas & inhibidores , Inositol/análogos & derivados , Microglía/fisiología , Agregación Patológica de Proteínas/enzimología , alfa-Sinucleína/metabolismo , Animales , Autofagia , Transporte Axonal , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Activación de Complemento , Glucosilceramidasa/metabolismo , Inositol/farmacología , Masculino , Ratones , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/enzimología , Agregación Patológica de Proteínas/inducido químicamente , Proteínas/metabolismo , Transmisión Sináptica
4.
Am J Hum Genet ; 96(6): 962-70, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-26004200

RESUMEN

An expanded G4C2 repeat in C9orf72 represents the most common known genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). However, the lower limit for pathological expansions is unknown (the suggested cutoff is 30 repeats). It has been proposed that the expansion might have occurred only once in human history and subsequently spread throughout the population. However, our present findings support a hypothesis of multiple origins for the expansion. We report a British-Canadian family in whom a ∼70-repeat allele from the father (unaffected by ALS or FTLD at age 89 years) expanded during parent-offspring transmission and started the first generation affected by ALS (four children carry an ∼1,750-repeat allele). Epigenetic and RNA-expression analyses further discriminated the offspring's large expansions (which were methylated and associated with reduced C9orf72 expression) from the ∼70-repeat allele (which was unmethylated and associated with upregulation of C9orf72). Moreover, RNA foci were only detected in fibroblasts from offspring with large expansions, but not in the father, who has the ∼70-repeat allele. All family members with expansions were found to have an ancient known risk haplotype, although it was inherited on a unique 5-Mb genetic backbone. We conclude that small expansions (e.g., 70 repeats) might be considered "pre-mutations" to reflect their propensity to expand in the next generation. Follow-up studies might help explain the high frequency of ALS- or FTLD-affected individuals with an expansion but without a familial history (e.g., 21% among Finnish ALS subjects).


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Expansión de las Repeticiones de ADN/genética , Degeneración Lobar Frontotemporal/genética , Degeneración Lobar Frontotemporal/patología , Proteínas/genética , Southern Blotting , Proteína C9orf72 , Canadá , Metilación de ADN/genética , Haplotipos/genética , Humanos , Linaje , Reacción en Cadena de la Polimerasa
5.
PLoS One ; 10(3): e0121072, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25815475

RESUMEN

Dopaminergic neurons in the substantia nigra pars compacta (SNpc) are characterized by the expression of genes required for dopamine synthesis, handling and reuptake and the expression of these genes is largely controlled by nuclear receptor related 1 (Nurr1). Nurr1 is also expressed in astrocytes and microglia where it functions to mitigate the release of proinflammatory cytokines and neurotoxic factors. Given that Parkinson's disease (PD) pathogenesis has been linked to both loss of Nurr1 expression in the SNpc and inflammation, increasing levels of Nurr1 maybe a promising therapeutic strategy. In this study a novel Nurr1 agonist, SA00025, was tested for both its efficiency to induce the transcription of dopaminergic target genes in vivo and prevent dopaminergic neuron degeneration in an inflammation exacerbated 6-OHDA-lesion model of PD. SA00025 (30mg/kg p.o.) entered the brain and modulated the expression of the dopaminergic phenotype genes TH, VMAT, DAT, AADC and the GDNF receptor gene c-Ret in the SN of naive rats. Daily gavage treatment with SA00025 (30mg/kg) for 32 days also induced partial neuroprotection of dopaminergic neurons and fibers in rats administered a priming injection of polyinosinic-polycytidylic acid (poly(I:C) and subsequent injection of 6-OHDA. The neuroprotective effects of SA00025 in this dopamine neuron degeneration model were associated with changes in microglial morphology indicative of a resting state and a decrease in microglial specific IBA-1 staining intensity in the SNpc. Astrocyte specific GFAP staining intensity and IL-6 levels were also reduced. We conclude that Nurr1 agonist treatment causes neuroprotective and anti-inflammatory effects in an inflammation exacerbated 6-OHDA lesion model of PD.


Asunto(s)
Dopamina/biosíntesis , Imidazoles/administración & dosificación , Inflamación/tratamiento farmacológico , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Enfermedad de Parkinson Secundaria/tratamiento farmacológico , Piridinas/administración & dosificación , Receptor Toll-Like 3/biosíntesis , Animales , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Expresión Génica , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Masculino , Microglía/metabolismo , Microglía/patología , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/patología , Neuroprotección/efectos de los fármacos , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/agonistas , Oxidopamina/toxicidad , Enfermedad de Parkinson Secundaria/metabolismo , Enfermedad de Parkinson Secundaria/patología , Porción Compacta de la Sustancia Negra/efectos de los fármacos , Porción Compacta de la Sustancia Negra/metabolismo , Poli I-C/administración & dosificación , ARN Bicatenario , Ratas , Receptor Toll-Like 3/genética
6.
J Neurochem ; 131(5): 588-601, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25113441

RESUMEN

Peripherin is a type III intermediate filament protein, the expression of which is associated with the acquisition and maintenance of a terminally differentiated neuronal phenotype. Peripherin up-regulation occurs during acute neuronal injury and in degenerating motor neurons of amyotrophic lateral sclerosis. The functional role(s) of peripherin during normal, injurious, and disease conditions remains unknown, but may be related to differential expression of spliced isoforms. To better understand peripherin function, we performed a yeast two-hybrid screen on a mouse brain cDNA library using an assembly incompetent peripherin isoform, Per-61, as bait. We identified new peripherin interactors with roles in vesicular trafficking, signal transduction, DNA/RNA processing, protein folding, and mitochondrial metabolism. We focused on the interaction of Per-61 and the constitutive isoform, Per-58, with SNAP25 interacting protein 30 (SIP30), a neuronal protein involved in SNAP receptor-dependent exocytosis. We found that peripherin and SIP30 interacted through coiled-coil domains and colocalized in cytoplasmic aggregates in SW13vim(-) cells. Interestingly, Per-61 and Per-58 differentially altered the subcellular distribution of SIP30 and SNAP25 in primary motor neurons. Our findings suggest a novel role of peripherin in vesicle trafficking.


Asunto(s)
Periferinas/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Fracciones Subcelulares/metabolismo , Técnicas del Sistema de Dos Híbridos , Animales , Línea Celular Transformada , Humanos , Inmunoprecipitación , Ratones , Mutación/genética , Periferinas/genética , Isoformas de Proteínas/fisiología , Estructura Terciaria de Proteína , Receptores de Lisoesfingolípidos/genética , Transfección
7.
Exp Neurol ; 261: 217-29, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24907400

RESUMEN

Intracellular proteinaceous inclusions are well-documented hallmarks of the fatal motor neuron disorder amyotrophic lateral sclerosis (ALS). The pathological significance of these inclusions remains unknown. Peripherin, a type III intermediate filament protein, is upregulated in ALS and identified as a component within different types of ALS inclusions. The formation of these inclusions may be associated with abnormal peripherin splicing, whereby an increase in mRNA retaining introns 3 and 4 (Per-3,4) leads to the generation of an aggregation-prone isoform, Per-28. During the course of evaluating peripherin filament assembly in SW-13 cells, we identified that expression of both Per-3,4 and Per-28 transcripts formed inclusions with categorically distinct morphology: Per-3,4 was associated with cytoplasmic condensed/bundled filaments, small inclusions (<10µM), or large inclusions (≥10µM); while Per-28 was associated with punctate inclusions in the nucleus and/or cytoplasm. We found temporal and spatial changes in inclusion morphology between 12 and 48h post-transfected cells, which were accompanied by unique immunofluorescent and biochemical changes of other ALS-relevant proteins, including TDP-43 and ubiquitin. Despite mild cytotoxicity associated with peripherin transfection, Per-3,4 and Per-28 expression increased cell viability during H2O2-mediated oxidative stress in BE(2)-M17 neuroblastoma cells. Taken together, this study shows that ALS-associated peripherin isoforms form dynamic cytoplasmic and intranuclear inclusions, effect changes in local endogenous protein expression, and afford cytoprotection against oxidative stress. These findings may have important relevance to understanding the pathophysiological role of inclusions in ALS.


Asunto(s)
Estrés Oxidativo/genética , Periferinas/genética , Agregación Patológica de Proteínas/genética , Isoformas de Proteínas/genética , Carcinoma/patología , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Peróxido de Hidrógeno/farmacología , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/metabolismo , Estrés Oxidativo/efectos de los fármacos , Periferinas/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/metabolismo , Factores de Tiempo , Transfección , Ubiquitina/metabolismo , Vimentina/metabolismo
8.
Neurosci Lett ; 576: 73-8, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24882721

RESUMEN

Adeno-associated viral (AAV) gene transfer holds great promise for treating a wide-range of neurodegenerative disorders. The AAV9 serotype crosses the blood-brain barrier and shows enhanced transduction efficiency compared to other serotypes, thus offering advantageous targeting when global transgene expression is required. Neonatal intravenous or intracerebroventricular (i.c.v.) delivery of recombinant AAV9 (rAAV9) have recently proven effective for modeling and treating several rodent models of neurodegenerative disease, however, the technique is associated with variable cellular tropism, making tailored gene transfer a challenge. In the current study, we employ the human synapsin 1 (hSYN1) gene promoter to drive neuron-specific expression of green fluorescent protein (GFP) after neonatal i.c.v. injection of rAAV9 in mice. We observed widespread GFP expression in neurons throughout the brain, spinal cord, and peripheral nerves and ganglia at 6 weeks-of-age. Region-specific quantification of GFP expression showed high neuronal transduction rates in substantia nigra pars reticulata (43.9±5.4%), motor cortex (43.5±3.3%), hippocampus (43.1±2.7%), cerebellum (29.6±2.3%), cervical spinal cord (24.9±3.9%), and ventromedial striatum (16.9±4.3%), among others. We found that 14.6±2.2% of neuromuscular junctions innervating the gastrocnemius muscle displayed GFP immunoreactivity. GFP expression was identified in several neuronal sub-types, including nigral tyrosine hydroxylase (TH)-positive dopaminergic cells, striatal dopamine- and cAMP-regulated neuronal phosphoprotein (DARPP-32)-positive neurons, and choline acetyltransferase (ChAT)-positive motor neurons. These results build on contemporary gene transfer techniques, demonstrating that the hSYN1 promoter can be used with rAAV9 to drive robust neuron-specific transgene expression throughout the nervous system.


Asunto(s)
Adenoviridae/genética , Encéfalo/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Neuronas/metabolismo , Médula Espinal/metabolismo , Sinapsinas/genética , Transgenes , Animales , Animales Recién Nacidos , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Humanos , Inyecciones Intraventriculares , Ratones , Regiones Promotoras Genéticas , Distribución Tisular
9.
Hum Mol Genet ; 23(17): 4510-27, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24728190

RESUMEN

A long-term goal of modeling Huntington's disease (HD) is to recapitulate the cardinal features of the disease in mice that express both mutant and wild-type (WT) huntingtin (Htt), as HD commonly manifests as a heterozygous condition in humans, and loss of WT Htt is associated with loss-of-function. In a new heterozygous Q175 knock-in (KI) mouse model, we performed an extensive evaluation of motor and cognitive functional deficits, neuropathological and biochemical changes and levels of proteins involved in synaptic function, the cytoskeleton and axonal transport, at 1-16 months of age. Motor deficits were apparent at 6 months of age in Q175 KI mice and at that time, postmortem striatal gamma-aminobutyric acid (GABA) levels were elevated and mutant Htt inclusions were present throughout the brain. From 6 months of age, levels of proteins associated with synaptic function, including SNAP-25, Rab3A and PSD-95, and with axonal transport and microtubules, including KIF3A, dynein and dynactin, were altered in the striatum, motor cortex, prefrontal cortex and hippocampus of Q175 KI mice, compared with WT levels. At 12-16 months of age, Q175 KI mice displayed motor and cognitive deficits, which were paralleled at postmortem by striatal atrophy, cortical thinning, degeneration of medium spiny neurons, dense mutant Htt inclusion formation, decreased striatal dopamine levels and loss of striatal brain-derived neurotrophic factor (BDNF). Data from this study indicate that the heterozygous Q175 KI mouse represents a realistic model for HD and also provides new insights into the specific and progressive synaptic, cytoskeletal and axonal transport protein abnormalities that may accompany the disease.


Asunto(s)
Transporte Axonal , Conducta Animal , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Sinapsis/metabolismo , Envejecimiento/patología , Animales , Atrofia/genética , Atrofia/patología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Heterocigoto , Cuerpos de Inclusión/metabolismo , Metaboloma , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Neostriado/metabolismo , Neostriado/patología , Neurotransmisores/metabolismo , Receptor trkB/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo
10.
Sci Transl Med ; 4(141): 141ra90, 2012 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-22764206

RESUMEN

Parkinson's disease (PD) is a common neurodegenerative disorder caused by genetic and environmental factors that results in degeneration of the nigrostriatal dopaminergic pathway in the brain. We analyzed neural cells generated from induced pluripotent stem cells (iPSCs) derived from PD patients and presymptomatic individuals carrying mutations in the PINK1 (PTEN-induced putative kinase 1) and LRRK2 (leucine-rich repeat kinase 2) genes, and compared them to those of healthy control subjects. We measured several aspects of mitochondrial responses in the iPSC-derived neural cells including production of reactive oxygen species, mitochondrial respiration, proton leakage, and intraneuronal movement of mitochondria. Cellular vulnerability associated with mitochondrial dysfunction in iPSC-derived neural cells from familial PD patients and at-risk individuals could be rescued with coenzyme Q(10), rapamycin, or the LRRK2 kinase inhibitor GW5074. Analysis of mitochondrial responses in iPSC-derived neural cells from PD patients carrying different mutations provides insight into convergence of cellular disease mechanisms between different familial forms of PD and highlights the importance of oxidative stress and mitochondrial dysfunction in this neurodegenerative disease.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Neuronas/citología , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Humanos , Indoles/uso terapéutico , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Neuronas/efectos de los fármacos , Fenoles/uso terapéutico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Sirolimus/uso terapéutico , Ubiquinona/uso terapéutico
11.
Neurobiol Dis ; 47(2): 258-67, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22549133

RESUMEN

While studying transgenic mice that overexpress human wildtype alpha-synuclein (Thy1-ASO, ASO) for typical brain alpha-synucleinopathy and central nervous system neuropathology, we observed progressive functional changes in the gastrointestinal and other peripheral organs. A more systematic study revealed that the gastrointestinal tract in ASO mice showed severe distension and blockage of the large intestine by 9-12 months of age. Functional assessments demonstrated a reduction in fecal water content and fecal pellet output, and increased whole gut transit time, in ASO mice compared to wildtype littermates, indicative of constipation, a symptom commonly reported by Parkinson's disease (PD) patients. Food intake was increased and body weight was decreased in 12 month old ASO mice, suggestive of metabolic abnormalities. Post-mortem histological analyses showed that human alpha-synuclein protein was robustly expressed in axonal fibers and in occasional cell bodies of the enteric nervous system, and in the heart of ASO mice. Accumulation of proteinase-K insoluble alpha-synuclein, reminiscent of neurodegenerative processes in PD was also observed. The functional and pathological changes we document here in ASO mice could relate to the autonomic deficits also seen in idiopathic and alpha-synuclein-mediated genetic forms of PD. These experimental data provide a foundation for therapeutic modeling of autonomic changes in PD and related alpha-synucleinopathies.


Asunto(s)
Enfermedades del Sistema Nervioso Autónomo/genética , Enfermedades del Sistema Nervioso Autónomo/metabolismo , Tracto Gastrointestinal/patología , Regulación de la Expresión Génica , Vejiga Urinaria/patología , alfa-Sinucleína/biosíntesis , Animales , Enfermedades del Sistema Nervioso Autónomo/patología , Tracto Gastrointestinal/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Vejiga Urinaria/metabolismo , alfa-Sinucleína/genética
12.
Mol Cell Neurosci ; 49(2): 230-9, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22155155

RESUMEN

Alternative splicing is a complex post-transcriptional process that can be regulated by cis-acting elements located within genomic non-coding regions. Recent studies have identified that polymorphic variations in non-coding regions of the α-synuclein gene (SNCA) locus are associated with an increased risk for developing Parkinson's disease (PD). The underlying mechanism(s) for this susceptibility may involve changes in α-synuclein mRNA expression and alternative splicing. As a first step towards understanding the biology of α-synuclein splice variants in PD, we characterized the levels of the full-length SNCA-140 mRNA transcript and SNCA-126, -112, and -98 alternatively spliced variants in different neuronal regions from PD patients or transgenic mice overexpressing human α-synuclein (ASO). In human post-mortem tissue, α-synuclein spliced transcripts were expressed in a region-specific manner in the cortex, substantia nigra, and cerebellum. We observed increased nigral SNCA-140 and SNCA-126 transcript levels in PD patients when compared to neurologically unaffected cases. Human α-synuclein splicing changes were also found to occur in a region-specific manner in ASO mice. Here, SNCA-126, -112, and -98 transcript levels did not increase proportionally with SNCA-140 levels, or parallel the region-specific mouse transcript ratios seen in wild-type (WT) littermates. While most transcripts were elevated in ASO mice when compared to WT mice, the most prominent increase was found in the ventral midbrain of 15-month-old ASO mice. These results demonstrate region-specific human α-synuclein transcript level abnormalities in PD patients and in a transgenic mouse model of α-synucleinopathy. This study is relevant to understanding the normal, adaptive, or pathological role(s) of α-synuclein splice variants.


Asunto(s)
Empalme Alternativo/genética , Enfermedad de Parkinson/genética , Sustancia Negra/metabolismo , alfa-Sinucleína/metabolismo , Secuencia de Aminoácidos , Animales , Cerebelo/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Neuronas/metabolismo , alfa-Sinucleína/genética
13.
Brain Res ; 1422: 57-65, 2011 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-21985957

RESUMEN

The type III intermediate filament peripherin is found associated with pathological inclusions present within motor neurons of patients with amyotrophic lateral sclerosis (ALS). Peripherin intra-isoform associations contribute to filament network formation at defined stoichiometric ratios. Distinct biochemical signatures characterize peripherin isoform expression in traumatic neuronal injury and motor neuron disease, while disruptions to peripherin alternative splicing or translation are associated with inclusion formation. In our efforts to identify pathological relationships between peripherin isoform expression and inclusion formation, we provide evidence of peripherin isoform-specific expression and ratio changes with concomitant, dose-dependent inclusion formation in response to oxidative stress. Upon increasing exposure to physiologically relevant levels of hydrogen peroxide in Neuro-2a cells, we observed a significant increase and decrease in peripherin isoforms Per-58 and Per-45, respectively, with peripherin-specific perikaryal aggregation of filaments 10-15 µm in diameter. Interestingly, peripherin-immunoreactive inclusions showed no overt carbonylation, suggesting that aggregation may serve a physiologically relevant role during oxidative stress. These findings provide novel insight into the biological significance of peripherin isoforms and inclusion formation, with relevance to the pathology of ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Cuerpos de Inclusión/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Glicoproteínas de Membrana/metabolismo , Neuronas Motoras/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuroblastoma/metabolismo , Estrés Oxidativo/fisiología , Animales , Línea Celular Tumoral , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/patología , Proteínas de Filamentos Intermediarios/química , Proteínas de Filamentos Intermediarios/genética , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Ratones , Neuronas Motoras/efectos de los fármacos , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Neuroblastoma/química , Periferinas , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/química , Isoformas de Proteínas/genética
14.
J Neurosci ; 28(8): 1833-40, 2008 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-18287500

RESUMEN

The neuronal intermediate filament protein peripherin is a component of ubiquitinated inclusions and of axonal spheroids in amyotrophic lateral sclerosis (ALS). Overexpression of peripherin causes motor neuron degeneration in transgenic mice and variations within the peripherin gene have been identified in ALS cases. We have shown previously the abnormal expression of a neurotoxic peripherin splice variant in transgenic mice expressing mutant superoxide dismutase-1. These findings indicated that abnormalities of peripherin splicing may occur in ALS. In the current study, peripherin splice variants were identified by reverse transcription-PCR of human neuronal RNA and comparisons in expression made between control and ALS spinal cord using Western blot analysis and immunocytochemistry. Using this approach we have identified a novel peripherin transcript retaining introns 3 and 4 that results in a 28 kDa splice isoform, designated Per 28. Using an antibody specific to Per 28, we show that this isoform is expressed at low stoichiometric levels from the peripherin gene, however causes peripherin aggregation when its expression is upregulated. Importantly we show an upregulation of Per 28 expression in ALS compared with controls, at both the mRNA and protein levels, and that Per 28 is associated with disease pathology, specifically round inclusions. These findings are the first to establish that peripherin splicing abnormalities occur in ALS, generating aggregation-prone splice isoforms.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Proteínas de Filamentos Intermediarios/biosíntesis , Proteínas de Filamentos Intermediarios/genética , Intrones/fisiología , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Regulación hacia Arriba/genética , Adolescente , Adulto , Anciano , Esclerosis Amiotrófica Lateral/metabolismo , Línea Celular Tumoral , Células Cultivadas , Femenino , Humanos , Proteínas de Filamentos Intermediarios/fisiología , Masculino , Glicoproteínas de Membrana/fisiología , Persona de Mediana Edad , Proteínas del Tejido Nervioso/fisiología , Periferinas , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología
15.
Glia ; 52(1): 59-69, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15920739

RESUMEN

Neurofilament (NF) aggregate formation within motor neurons is a pathological hallmark of both the sporadic and familial forms of amyotrophic lateral sclerosis (ALS). The relationship between aggregate formation and both microglial and astrocytic proliferation, as well as additional neuropathological features of ALS, is unknown. To examine this, we have used transgenic mice that develop NF aggregates, through either a lack of the low-molecular-weight NF subunit [NFL (-/-)] or the overexpression of human NFL [hNFL (+/+)]. Transgenic and wild-type C57bl/6 mice were examined from 1 month to 18 months of age, and the temporal pattern of motor neuron degeneration, microglial and astrocytic proliferation, and heat shock protein-70 (HSP-70) expression characterized. We observed three overlapping phases in both transgenic mice, including transient aggregate formation, reactive microgliosis, and progressive motor neuron loss. However, only NFL (-/-) mice demonstrated significant astrogliosis and HSP-70 upregulation in both motor neurons and astrocytes. These in vivo models suggest that the development of NF aggregates in motor neurons leads to motor neuron death, but that the interaction between the degenerating motor neurons and the adjacent non-neuronal cells may differ significantly depending on the etiology of the NF aggregate itself.


Asunto(s)
Esclerosis Amiotrófica Lateral/fisiopatología , Gliosis/fisiopatología , Degeneración Nerviosa/fisiopatología , Proteínas de Neurofilamentos/genética , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Astrocitos/metabolismo , Astrocitos/patología , Caspasa 3 , Caspasas/metabolismo , Muerte Celular/fisiología , Proliferación Celular , Modelos Animales de Enfermedad , Gliosis/genética , Gliosis/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/metabolismo , Cuerpos de Inclusión/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microglía/metabolismo , Microglía/patología , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Factores de Tiempo
16.
J Neurobiol ; 61(3): 392-9, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15490479

RESUMEN

Genetic analysis of the Drosophila larval neuromuscular junction has identified some of the key molecules that regulate synaptic plasticity. Among these molecules, the expression level of Fasciclin II (FasII), a homophilic cell adhesion molecule, is critically important for determining the final form of the neuromuscular junction. Genetic reduction of FasII expression by 50% yields more elaborate nerve terminals, while a greater reduction in expression, to 10% of wild-type, yields a substantial reduction in the nerve terminal morphology. Importantly, regulation of FasII expression seems to be the final output for several genetic manipulations that transform NMJ morphology. In an effort to understand the importance of this regulatory pathway in the normal animal, we have undertaken studies to identify environmental cues that might be important for initiating FasII-dependent changes in synaptic plasticity. Here we report on the relationship between larval population density and synaptic morphology, synaptic strength, and FasII levels. We raised Drosophila larvae under conditions of increasing population density and found an inverse exponential relationship between population density and the number of synaptic boutons, the number of branches, and the length of branches. We also observed population-dependent alteration in FasII levels, with lower densities having less FasII at the synapse. The correlation between density and morphological change was abrogated in larvae constitutively expressing FasII, and in wild-type larvae grown on soft culture medium. Together these data show that environmental cues can induce regulation of FasII. Interestingly, however, the quantal content of synaptic transmission was not different among the different population densities, suggesting that other factors contribute to maintaining synaptic strength at a defined level.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Unión Neuromuscular/fisiología , Sinapsis/fisiología , Animales , Drosophila , Estimulación Eléctrica , Potenciales Evocados/fisiología , Inmunohistoquímica/métodos , Larva , Músculos/fisiología , Músculos/efectos de la radiación , Unión Neuromuscular/crecimiento & desarrollo , Densidad de Población , Terminales Presinápticos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...