Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Neurobiol Dis ; 89: 1-9, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26826269

RESUMO

Over-expression of mutant copper, zinc superoxide dismutase (SOD) in mice induces ALS and has become the most widely used model of neurodegeneration. However, no pharmaceutical agent in 20 years has extended lifespan by more than a few weeks. The Copper-Chaperone-for-SOD (CCS) protein completes the maturation of SOD by inserting copper, but paradoxically human CCS causes mice co-expressing mutant SOD to die within two weeks of birth. Hypothesizing that co-expression of CCS created copper deficiency in spinal cord, we treated these pups with the PET-imaging agent CuATSM, which is known to deliver copper into the CNS within minutes. CuATSM prevented the early mortality of CCSxSOD mice, while markedly increasing Cu, Zn SOD protein in their ventral spinal cord. Remarkably, continued treatment with CuATSM extended the survival of these mice by an average of 18 months. When CuATSM treatment was stopped, these mice developed ALS-related symptoms and died within 3 months. Restoring CuATSM treatment could rescue these mice after they became symptomatic, providing a means to start and stop disease progression. All ALS patients also express human CCS, raising the hope that familial SOD ALS patients could respond to CuATSM treatment similarly to the CCSxSOD mice.


Assuntos
Esclerose Lateral Amiotrófica/enzimologia , Cobre/administração & dosagem , Cobre/metabolismo , Chaperonas Moleculares/metabolismo , Medula Espinal/metabolismo , Superóxido Dismutase/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Animais , Modelos Animais de Doenças , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Estimativa de Kaplan-Meier , Camundongos , Camundongos Transgênicos , Chaperonas Moleculares/genética , Superóxido Dismutase/genética
2.
J Biol Chem ; 289(20): 14263-71, 2014 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-24719334

RESUMO

MicroRNA (miRNA) maturation is regulated by interaction of particular miRNA precursors with specific RNA-binding proteins. Following their biogenesis, mature miRNAs are incorporated into the RNA-induced silencing complex (RISC) where they interact with mRNAs to negatively regulate protein production. However, little is known about how mature miRNAs are regulated at the level of their activity. To address this, we screened for proteins differentially bound to the mature form of the miR-1 or miR-133 miRNA families. These muscle-enriched, co-transcribed miRNA pairs cooperate to suppress smooth muscle gene expression in the heart. However, they also have opposing roles, with the miR-1 family, composed of miR-1 and miR-206, promoting myogenic differentiation, whereas miR-133 maintains the progenitor state. Here, we describe a physical interaction between TDP-43, an RNA-binding protein that forms aggregates in the neuromuscular disease, amyotrophic lateral sclerosis, and the miR-1, but not miR-133, family. Deficiency of the TDP-43 Drosophila ortholog enhanced dmiR-1 activity in vivo. In mammalian cells, TDP-43 limited the activity of both miR-1 and miR-206, but not the miR-133 family, by disrupting their RISC association. Consistent with TDP-43 dampening miR-1/206 activity, protein levels of the miR-1/206 targets, IGF-1 and HDAC4, were elevated in TDP-43 transgenic mouse muscle. This occurred without corresponding Igf-1 or Hdac4 mRNA increases and despite higher miR-1 and miR-206 expression. Our findings reveal that TDP-43 negatively regulates the activity of the miR-1 family of miRNAs by limiting their bioavailability for RISC loading and suggest a processing-independent mechanism for differential regulation of miRNA activity.


Assuntos
Proteínas de Ligação a DNA/metabolismo , MicroRNAs/metabolismo , Complexo de Inativação Induzido por RNA/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Animais , Proteínas Argonautas/metabolismo , Drosophila melanogaster/genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Ligação Proteica
3.
J Neurochem ; 118(5): 891-901, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21692800

RESUMO

This study presents the initial characterization of transgenic mice with mutations in a primary zinc-binding residue (H80), either alone or with a G93A mutation. H80G;G93A superoxide dismutase 1 (SOD1) transgenic mice developed paralysis with motor neuron loss, and ubiquitin inclusion-type rather than mitochondrial vacuolar pathology. Unlike G93A SOD1-related disease, the course was not accelerated by over-expression of copper chaperone for SOD1. H80G SOD1 transgenic mice did not manifest disease at levels of SOD1 transgene expressed. The H80G mutation altered certain biochemical parameters of both human wild-type SOD1 and G93A SOD1. The H80G mutation does not substantially change the age-dependent accumulation of G93A SOD1 aggregates and hydrophobic species in spinal cord. However, both H80G;G93A SOD1 and H80G SOD1 lack dismutase activity, the ability to form homodimers, and co-operativity with copper chaperone for SOD1, indicating that their dimerization interface is abnormal. The H80G mutation also made SOD1 susceptible to protease digestion. The H80G mutation alters the redox properties of SOD1. G93A SOD1 exists in either reduced or oxidized form, whereas H80G;G93A SOD1 and H80G SOD1 exist only in a reduced state. The inability of SOD1 with an H80G mutation to take part in normal oxidation-reduction reactions has important ramifications for disease mechanisms and pathology in vivo.


Assuntos
Glutamina/genética , Histidina/genética , Mutação/genética , Superóxido Dismutase/genética , Zinco/metabolismo , Fatores Etários , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Cromoterapia/métodos , Modelos Animais de Doenças , Endopeptidase K/farmacologia , Humanos , Interações Hidrofóbicas e Hidrofílicas/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Oxirredução/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Medula Espinal/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1
4.
Brain ; 133(Pt 2): 375-88, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20145049

RESUMO

The primary biological function of the endogenous cellular prion protein has remained unclear. We investigated its biological function in the generation of cellular immune responses using cellular prion protein gene-specific small interfering ribonucleic acid in vivo and in vitro. Our results were confirmed by blocking cellular prion protein with monovalent antibodies and by using cellular prion protein-deficient and -transgenic mice. In vivo prion protein gene-small interfering ribonucleic acid treatment effects were of limited duration, restricted to secondary lymphoid organs and resulted in a 70% reduction of cellular prion protein expression in leukocytes. Disruption of cellular prion protein signalling augmented antigen-specific activation and proliferation, and enhanced T cell receptor signalling, resulting in zeta-chain-associated protein-70 phosphorylation and nuclear factor of activated T cells/activator protein 1 transcriptional activity. In vivo prion protein gene-small interfering ribonucleic acid treatment promoted T cell differentiation towards pro-inflammatory phenotypes and increased survival of antigen-specific T cells. Cellular prion protein silencing with small interfering ribonucleic acid also resulted in the worsening of actively induced and adoptively transferred experimental autoimmune encephalomyelitis. Finally, treatment of myelin basic protein(1-11) T cell receptor transgenic mice with prion protein gene-small interfering ribonucleic acid resulted in spontaneous experimental autoimmune encephalomyelitis. Thus, central nervous system autoimmune disease was modulated at all stages of disease: the generation of the T cell effector response, the elicitation of T effector function and the perpetuation of cellular immune responses. Our findings indicate that cellular prion protein regulates T cell receptor-mediated T cell activation, differentiation and survival. Defects in autoimmunity are restricted to the immune system and not the central nervous system. Our data identify cellular prion protein as a regulator of cellular immunological homoeostasis and suggest cellular prion protein as a novel potential target for therapeutic immunomodulation.


Assuntos
Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/genética , Inativação Gênica/imunologia , Príons/genética , Receptores de Antígenos de Linfócitos T/fisiologia , Transdução de Sinais/imunologia , Animais , Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/imunologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Príons/imunologia , RNA Interferente Pequeno/genética
5.
Neurobiol Dis ; 40(2): 404-14, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20621187

RESUMO

Familial ALS patients with TDP-43 gene mutations and sporadic ALS patients share common TDP-43 neuronal pathology. To delineate mechanisms underlying TDP-43 proteinopathies, transgenic mice expressing A315T, M337V or wild type human TDP-43 were generated. Multiple TDP-43 founders developed a severe early motor phenotype that correlated with TDP-43 levels in spinal cord. Three A315T TDP-43 lines developed later onset paralysis with cytoplasmic ubiquitin inclusions, gliosis and TDP-43 redistribution and fragmentation. The WT TDP-43 mouse line with highest spinal cord expression levels remains asymptomatic, although these mice show spinal cord pathology. One WT TDP-43 line with high skeletal muscle levels of TDP-43 developed a severe progressive myopathy. Over-expression of TDP-43 in vivo is sufficient to produce progressive motor phenotypes by a toxic gain of function paradigm. Transgenic mouse lines expressing untagged mutant and wild type TDP-43 under the same promoter represent a powerful new model system for studying TDP-43 proteinopathies in vivo.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Doença dos Neurônios Motores/genética , Doença dos Neurônios Motores/metabolismo , Debilidade Muscular/genética , Debilidade Muscular/metabolismo , Animais , Western Blotting , DNA Complementar , Modelos Animais de Doenças , Imunofluorescência , Vetores Genéticos , Humanos , Corpos de Inclusão/metabolismo , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Doença dos Neurônios Motores/fisiopatologia , Debilidade Muscular/fisiopatologia , Mutação de Sentido Incorreto , Reação em Cadeia da Polimerase , Proteinopatias TDP-43/genética , Proteinopatias TDP-43/metabolismo , Proteinopatias TDP-43/fisiopatologia
6.
Hum Mol Genet ; 17(12): 1728-37, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18337307

RESUMO

The CCS copper chaperone is critical for maturation of Cu, Zn-superoxide dismutase (SOD1) through insertion of the copper co-factor and oxidization of an intra-subunit disulfide. The disulfide helps stabilize the SOD1 polypeptide, which can be particularly important in cases of amyotrophic lateral sclerosis (ALS) linked to misfolding of mutant SOD1. Surprisingly, however, over-expressed CCS was recently shown to greatly accelerate disease in a G93A SOD1 mouse model for ALS. Herein we show that disease in these G93A/CCS mice correlates with incomplete oxidation of the SOD1 disulfide. In the brain and spinal cord, CCS over-expression failed to enhance oxidation of the G93A SOD1 disulfide and if anything, effected some accumulation of disulfide-reduced SOD1. This effect was mirrored in culture with a C244,246S mutant of CCS that has the capacity to interact with SOD1 but can neither insert copper nor oxidize the disulfide. In spite of disulfide effects, there was no evidence for increased SOD1 aggregation. If anything, CCS over-expression prevented SOD1 misfolding in culture as monitored by detergent insolubility. This protection against SOD1 misfolding does not require SOD1 enzyme activation as the same effect was obtained with the C244,246S allele of CCS. In the G93A SOD1 mouse, CCS over-expression was likewise associated with a lack of obvious SOD1 misfolding marked by detergent insolubility. CCS over-expression accelerates SOD1-linked disease without the hallmarks of misfolding and aggregation seen in other mutant SOD1 models. These studies are the first to indicate biological effects of CCS in the absence of SOD1 enzymatic activation.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Chaperonas Moleculares/metabolismo , Superóxido Dismutase/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Ativação Enzimática , Humanos , Camundongos , Camundongos Transgênicos , Chaperonas Moleculares/genética , Mutação , Dobramento de Proteína , Superóxido Dismutase/genética , Superóxido Dismutase-1
7.
EMBO Rep ; 9(8): 766-73, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18566599

RESUMO

Elucidating the cross-talk between inflammatory and cell proliferation pathways might provide important insights into the pathogenesis of inflammation-induced cancer. Here, we show that the receptor-interacting protein 1 (RIP1)-an essential mediator of inflammation-induced nuclear factor-kappaB (NF-kappaB) activation-regulates p27(Kip1) levels and cell-cycle progression. RIP1 regulates p27(Kip1) levels by an NF-kappaB-independent signal that involves activation of the phosphatidylinositol 3-kinase (PI3K)-Akt-forkhead pathway. Mouse embryonic fibroblasts (MEFs) from RIP1-knockout mice express high levels of p27(Kip1). Reconstitution of MEFs with RIP1 downregulates p27(Kip1) levels in a PI3K-dependent manner. RIP1 regulates p27(Kip1) at the messenger RNA level by regulating the p27(Kip1) promoter through the forkhead transcription factors. RIP1 expression blocks accumulation of cells in G(1) in response to serum starvation and favours cell-cycle progression. Finally, we show that overexpression of p27(Kip1) blocks the effects of RIP1 on the cell cycle. Thus, our study provides a new insight into how components of inflammatory and immune signalling pathways regulate cell-cycle progression.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Células 3T3 , Animais , Northern Blotting , Western Blotting , Ciclo Celular , Linhagem Celular , Cromonas/farmacologia , Inibidor de Quinase Dependente de Ciclina p27/genética , Fatores de Transcrição Forkhead/metabolismo , Proteínas Ativadoras de GTPase/genética , Humanos , Camundongos , Camundongos Knockout , Morfolinas/farmacologia , NF-kappa B/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-akt/genética
8.
Neuromuscul Disord ; 30(3): 236-240, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32165108

RESUMO

Mutations in heat shock protein B8 were initially identified in inherited neuropathies and were more recently found to cause a predominantly distal myopathy with myofibrillar pathology and rimmed vacuoles. Rare patients also had proximal weakness. Only very few pathogenic variants have been identified in HSPB8. Disruption of the chaperone activity of heat shock protein B8 impairs chaperone-assisted selective autophagy and results in protein aggregation. We report a 23-year-old patient who presented with a 4-year history of predominantly proximal lower limb weakness due to a novel variant in HSPB8. The creatine kinase level was mildly elevated. Electrodiagnostic studies demonstrated a proximal-predominant myopathy without evidence of neuropathy, and muscle histopathology revealed rimmed vacuoles and myofibrillar protein aggregates. Whole exome sequencing identified a de novo frameshift variant in the C-terminal region of HSPB8 (c.577_580dupGTCA, p.Thr194Serfs*23). This case demonstrates that HSPB8-related disorders can present with early onset limb-girdle myopathy without associated neuropathy.


Assuntos
Proteínas de Choque Térmico/genética , Doenças por Armazenamento dos Lisossomos , Chaperonas Moleculares/genética , Doenças Musculares , Distrofia Muscular do Cíngulo dos Membros , Miopatias Congênitas Estruturais , Adulto , Humanos , Doenças por Armazenamento dos Lisossomos/genética , Doenças por Armazenamento dos Lisossomos/fisiopatologia , Masculino , Doenças Musculares/genética , Doenças Musculares/fisiopatologia , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/fisiopatologia , Miopatias Congênitas Estruturais/genética , Miopatias Congênitas Estruturais/fisiopatologia , Adulto Jovem
9.
Neurobiol Dis ; 34(1): 155-62, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19320055

RESUMO

Over-expression of CCS in G93A SOD1 mice accelerates neurological disease and enhances mitochondrial pathology. We studied the effect of CCS over-expression in transgenic mice expressing G37R, G86R or L126Z SOD1 mutations in order to understand factors which influence mitochondrial dysfunction. Over-expression of CCS markedly decreased survival and produced mitochondrial vacuolation in G37R SOD1 mice but not in G86R or L126Z SOD1 mice. Moreover, CCS/G37R SOD1 spinal cord showed specific reductions in mitochondrial complex IV subunits consistent with an isolated COX deficiency, while no such reductions were detected in CCS/G86R or CCS/L126Z SOD1 mice. CCS over-expression increased the ratio of reduced to oxidized SOD1 monomers in the spinal cords of G37R SOD1 as well as G93A SOD1 mice, but did not influence the redox state of G86R or L126Z SOD1 monomers. The effects of CCS on disease are SOD1 mutation dependent and correlate with SOD1 redox susceptibility.


Assuntos
Mitocôndrias/metabolismo , Chaperonas Moleculares/metabolismo , Medula Espinal/metabolismo , Superóxido Dismutase/genética , Animais , Western Blotting , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Mitocôndrias/ultraestrutura , Mutação , Oxirredução , Medula Espinal/ultraestrutura , Superóxido Dismutase-1
10.
JAMA Neurol ; 74(2): 238-241, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27942721

RESUMO

A 26-year-old right-handed man presented with progressive gait imbalance over 6 years. His examination was consistent with cerebellar and upper motor neuronal dysfunction. He had no significant family history. Most of the serum and cerebrospinal fluid studies were unremarkable. Neuroimaging was remarkable for mild cerebellar and noticeable thoracic spinal cord atrophy. The initial differential diagnosis for the patient's presentation was broad, but because of certain clinical characteristics, it was later focused on hereditary ataxias. Detailed analysis of the clinical features in the history, neurologic examination, and neuroimaging studies led to the diagnosis.


Assuntos
Encéfalo/patologia , Progressão da Doença , Deficiência Intelectual/complicações , Doença dos Neurônios Motores/complicações , Espasticidade Muscular/complicações , Atrofia Óptica/complicações , Ataxias Espinocerebelares/complicações , Adulto , Atrofia/patologia , Encéfalo/diagnóstico por imagem , Humanos , Deficiência Intelectual/diagnóstico por imagem , Imageamento por Ressonância Magnética , Masculino , Doença dos Neurônios Motores/diagnóstico por imagem , Espasticidade Muscular/diagnóstico por imagem , Atrofia Óptica/diagnóstico por imagem , Medula Espinal/diagnóstico por imagem , Ataxias Espinocerebelares/diagnóstico por imagem
11.
J Neurosci ; 22(20): 8790-6, 2002 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-12388585

RESUMO

Mutations in the Cu/Zn superoxide dismutase (SOD1) gene cause one form of familial amyotrophic lateral sclerosis, a progressive disorder of motor neurons leading to weakness and death of affected individuals. Experiments using both transgenic mice expressing mutant SOD1 and SOD1 knock-out mice have demonstrated that disease is caused by a toxic gain of function and not by a loss of normal SOD1 activity. Precise mechanisms underlying mutant SOD1 toxicity are unclear but may involve abnormal interactions between zinc and SOD1. The metallothioneins (MTs) represent a family of zinc binding proteins that can function as zinc chaperones for apo-SOD1 in vitro. We hypothesized that manipulation of metallothioneins in vivo might alter the disease phenotype of transgenic mice expressing G93A SOD1 and therefore crossed this line with MT-I and MT-II or MT-III knock-out mice. G93A SOD1 mice deficient of either MT-I and MT-II or MT-III exhibited significant reductions in survival compared with G93A SOD1 mice. In addition, motor dysfunction was markedly accelerated in G93A SOD1 mice deficient in metallothioneins with regard to onset (MT-I and MT-II) or progression (MT-III). These results indicate that the disease course in G93A SOD1 mice is dependent on levels of metallothionein expression. Because MT-I and MT-II are expressed in glia whereas MT-III is found in neurons, these results also indicate that primary changes within non-neuronal cells can affect mutant SOD1-induced disease and do so in ways distinct from primary neuronal changes.


Assuntos
Esclerose Lateral Amiotrófica/fisiopatologia , Metalotioneína/deficiência , Proteínas do Tecido Nervoso/deficiência , Neurônios/metabolismo , Zinco/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Western Blotting , Contagem de Células , Cruzamentos Genéticos , Modelos Animais de Doenças , Progressão da Doença , Proteína Glial Fibrilar Ácida/biossíntese , Humanos , Metalotioneína/genética , Metalotioneína 3 , Camundongos , Camundongos Transgênicos , Atividade Motora/genética , Proteínas do Tecido Nervoso/genética , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/patologia , Fenótipo , Medula Espinal/patologia , Superóxido Dismutase/biossíntese , Superóxido Dismutase/genética , Análise de Sobrevida
12.
J Neurol Sci ; 336(1-2): 1-7, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24269091

RESUMO

Several hypotheses have been proposed for the mechanisms underlying mutant Cu,Zn Superoxide Dismutase-related Amyotrophic Lateral Sclerosis. These include aggregation pathology, mitochondrial dysfunctions, oxidative stress, and glutamate-mediated excitotoxicity. Mitochondrial disease may be a primary event in neurodegeneration, contributing to oxidative stress and apoptosis, or it may be caused by other cellular processes. Mitochondrial structural abnormalities have been detected in the skeletal muscle, lymphoblast and central nervous system of Amyotrophic Lateral Sclerosis patients. The cause or even the extent of mitochondrial defects in spinal cord and brain of patients with Cu,Zn Superoxide Dismutase mutations is difficult to determine because of rapid mitochondrial deterioration in autopsy samples. The focus of this review is how abnormalities in Cu,Zn Superoxide Dismutase redox states, folding and metallation contribute to mitochondrial deficiencies, investigating the differences in mitochondrial defects observed among transgenic mice expressing various Cu,Zn Superoxide Dismutase mutations.


Assuntos
Doenças Mitocondriais/genética , Proteínas Mitocondriais/genética , Chaperonas Moleculares/genética , Mutação/genética , Superóxido Dismutase/genética , Animais , Humanos , Camundongos , Camundongos Transgênicos , Doenças Mitocondriais/enzimologia , Chaperonas Moleculares/fisiologia , Dobramento de Proteína
13.
PLoS One ; 8(8): e71793, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23967244

RESUMO

The identification of proteins which determine fat and lean body mass composition is critical to better understanding and treating human obesity. TDP-43 is a well-conserved RNA-binding protein known to regulate alternative splicing and recently implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS). While TDP-43 knockout mice show early embryonic lethality, post-natal conditional knockout mice show weight loss, fat depletion, and rapid death, suggesting an important role for TDP-43 in regulating energy metabolism. Here we report, that over-expression of TDP-43 in transgenic mice can result in a phenotype characterized by increased fat deposition and adipocyte hypertrophy. In addition, TDP-43 over-expression in skeletal muscle results in increased steady state levels of Tbc1d1, a RAB-GTPase activating protein involved in Glucose 4 transporter (Glut4) translocation. Skeletal muscle fibers isolated from TDP-43 transgenic mice show altered Glut4 translocation in response to insulin and impaired insulin mediated glucose uptake. These results indicate that levels of TDP-43 regulate body fat composition and glucose homeostasis in vivo.


Assuntos
Tecido Adiposo/metabolismo , Adiposidade , Proteínas de Ligação a DNA/metabolismo , Glucose/metabolismo , Homeostase , Tecido Adiposo/crescimento & desenvolvimento , Adiposidade/genética , Animais , Proteínas de Ligação a DNA/genética , Proteínas Ativadoras de GTPase , Expressão Gênica , Transportador de Glucose Tipo 4/genética , Transportador de Glucose Tipo 4/metabolismo , Humanos , Insulina/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Tamanho do Órgão/genética , Transporte Proteico
14.
Arch Neurol ; 68(5): 650-2, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21555641

RESUMO

BACKGROUND: There have been a few case reports of motor neuron disease in association with Huntington disease (HD). OBJECTIVE: To describe a patient presenting with prominent fasciculations, chorea, and possible amyotrophic lateral sclerosis (ALS) in whom genetic testing revealed HD mutation. DESIGN: Case report. SETTING: University of Texas Southwestern Medical Center, Dallas. Patient  A 69-year-old man with chorea and fasciculations. INTERVENTIONS: Genetic and electrophysiologic testing. MAIN OUTCOME MEASURES: Genetic test result, electrophysiologic test result, and physical examination. RESULTS: A 69-year-old man with long-standing depression and failing memory presented with muscle twitches of 8 months' duration. He was found to have choreoathetoid movements and distal weakness on neurological examination. Electrophysiologic studies revealed evidence of motor neuron disease. Genetic test showed CAG repeat of 40 on chromosome 4, confirming the diagnosis of HD. CONCLUSION: Motor neuron disease can rarely occur in patients with HD and could be one of its presenting features.


Assuntos
Cromossomos Humanos Par 4/genética , Doença de Huntington/diagnóstico , Doença de Huntington/genética , Doença dos Neurônios Motores/genética , Idoso , Esclerose Lateral Amiotrófica/genética , Vértebras Cervicais , Diagnóstico Diferencial , Eletrofisiologia , Fasciculação/genética , Testes Genéticos , Humanos , Doença de Huntington/complicações , Doença de Huntington/fisiopatologia , Masculino , Doença dos Neurônios Motores/diagnóstico , Exame Neurológico , Espondilose/complicações , Espondilose/cirurgia , Expansão das Repetições de Trinucleotídeos
16.
Science ; 326(5959): 1549-54, 2009 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-20007902

RESUMO

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by loss of motor neurons, denervation of target muscles, muscle atrophy, and paralysis. Understanding ALS pathogenesis may require a fuller understanding of the bidirectional signaling between motor neurons and skeletal muscle fibers at neuromuscular synapses. Here, we show that a key regulator of this signaling is miR-206, a skeletal muscle-specific microRNA that is dramatically induced in a mouse model of ALS. Mice that are genetically deficient in miR-206 form normal neuromuscular synapses during development, but deficiency of miR-206 in the ALS mouse model accelerates disease progression. miR-206 is required for efficient regeneration of neuromuscular synapses after acute nerve injury, which probably accounts for its salutary effects in ALS. miR-206 mediates these effects at least in part through histone deacetylase 4 and fibroblast growth factor signaling pathways. Thus, miR-206 slows ALS progression by sensing motor neuron injury and promoting the compensatory regeneration of neuromuscular synapses.


Assuntos
Esclerose Lateral Amiotrófica/fisiopatologia , MicroRNAs/metabolismo , Neurônios Motores/fisiologia , Músculo Esquelético/metabolismo , Junção Neuromuscular/patologia , Junção Neuromuscular/fisiologia , Esclerose Lateral Amiotrófica/patologia , Animais , Axônios/fisiologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Fatores de Crescimento de Fibroblastos/metabolismo , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Transgênicos , MicroRNAs/genética , Neurônios Motores/patologia , Denervação Muscular , Músculo Esquelético/inervação , Músculo Esquelético/patologia , Proteína MyoD/genética , Proteína MyoD/metabolismo , Miogenina/genética , Miogenina/metabolismo , Regeneração Nervosa , Junção Neuromuscular/crescimento & desenvolvimento , Interferência de RNA , Transdução de Sinais , Ativação Transcricional , Regulação para Cima
17.
J Biol Chem ; 283(18): 12267-75, 2008 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-18334481

RESUMO

G93A SOD1 transgenic mice overexpressing CCS protein develop an accelerated disease course that is associated with enhanced mitochondrial pathology and increased mitochondrial localization of mutant SOD1. Because these results suggest an effect of mutant SOD1 on mitochondrial function, we assessed the enzymatic activities of mitochondrial respiratory chain complexes in the spinal cords of CCS/G93A SOD1 and control mice. CCS/G93A SOD1 mouse spinal cord demonstrates a 55% loss of complex IV (cytochrome c oxidase) activity compared with spinal cord from age-matched non-transgenic or G93A SOD1 mice. In contrast, CCS/G93A SOD1 spinal cord shows no reduction in the activities of complex I, II, or III. Blue native gel analysis further demonstrates a marked reduction in the levels of complex IV but not of complex I, II, III, or V in spinal cords of CCS/G93A SOD1 mice compared with non-transgenic, G93A SOD1, or CCS/WT SOD1 controls. With SDS-PAGE analysis, spinal cords from CCS/G93A SOD1 mice showed significant decreases in the levels of two structural subunits of cytochrome c oxidase, COX1 and COX5b, relative to controls. In contrast, CCS/G93A SOD1 mouse spinal cord showed no reduction in levels of selected subunits from complexes I, II, III, or V. Heme A analyses of spinal cord further support the existence of cytochrome c oxidase deficiency in CCS/G93A SOD1 mice. Collectively, these results establish that CCS/G93A SOD1 mice manifest an isolated complex IV deficiency which may underlie a substantial part of mutant SOD1-induced mitochondrial cytopathy.


Assuntos
Alanina/genética , Substituição de Aminoácidos , Deficiência de Citocromo-c Oxidase/enzimologia , Glicina/genética , Chaperonas Moleculares/metabolismo , Superóxido Dismutase/metabolismo , Animais , Eletroforese em Gel de Poliacrilamida , Heme/metabolismo , Humanos , Rim/enzimologia , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos , Fosforilação Oxidativa , Subunidades Proteicas/metabolismo , Medula Espinal/enzimologia
18.
Exp Neurol ; 206(1): 53-8, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17482163

RESUMO

The accumulation of protein aggregates is thought to be an important component in the pathogenesis of mutant SOD1-induced disease. Mutant SOD1 aggregates appear to be cleared by proteasomes, at least in vitro, suggesting a potentially important role for proteasome degradation pathways in vivo. G93A SOD1 transgenic mice show an increase in proteasome activity and induction of immuno-proteasome subunits within spinal cord as they develop neurological symptoms. To determine what role immuno-proteasomes may have in mutant SOD1-induced disease, we crossed G93A SOD1 transgenic mice with LMP2-/- mice to obtain G93A SOD1 mice lacking the LMP2 immuno-proteasome subunit. G93A SOD1/LMP2-/- mice show significant reductions in proteasome function within spinal cord compared to G93A SOD1 mice. However, G93A SOD1/LMP2-/- mice show no change in motor function decline, or survival compared to G93A SOD1 mice. These results indicate that the loss of immuno-proteasome function in vivo does not significantly alter mutant SOD1-induced disease.


Assuntos
Esclerose Lateral Amiotrófica/imunologia , Cisteína Endopeptidases/imunologia , Neurônios Motores/imunologia , Complexo de Endopeptidases do Proteassoma/imunologia , Medula Espinal/imunologia , Superóxido Dismutase/metabolismo , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Animais , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neurônios Motores/metabolismo , Mutação/genética , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Medula Espinal/metabolismo , Medula Espinal/fisiopatologia , Superóxido Dismutase/genética , Superóxido Dismutase-1 , Taxa de Sobrevida , Regulação para Cima/fisiologia
19.
Proc Natl Acad Sci U S A ; 104(14): 6072-7, 2007 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-17389365

RESUMO

Cu, Zn superoxide dismutase (SOD1) has been detected within spinal cord mitochondria of mutant SOD1 transgenic mice, a model of familial ALS. The copper chaperone for SOD1 (CCS) provides SOD1 with copper, facilitates the conversion of immature apo-SOD1 to a mature holoform, and influences in yeast the cytosolic/mitochondrial partitioning of SOD1. To determine how CCS affects G93A-SOD1-induced disease, we generated transgenic mice overexpressing CCS and crossed them to G93A-SOD1 or wild-type SOD1 transgenic mice. Both CCS transgenic mice and CCS/wild-type-SOD1 dual transgenic mice are neurologically normal. In contrast, CCS/G93A-SOD1 dual transgenic mice develop accelerated neurological deficits, with a mean survival of 36 days, compared with 242 days for G93A-SOD1 mice. Immuno-EM and subcellular fractionation studies on the spinal cord show that G93A-SOD1 is enriched within mitochondria in the presence of CCS overexpression. Our results indicate that CCS overexpression in G93A-SOD1 mice produces severe mitochondrial pathology and accelerates disease course.


Assuntos
Cobre/metabolismo , Mitocôndrias/patologia , Neurônios Motores/patologia , Superóxido Dismutase/metabolismo , Animais , Fracionamento Celular , Clonagem Molecular , Cruzamentos Genéticos , DNA Complementar , Progressão da Doença , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Mutantes Neurológicos , Camundongos Transgênicos , Mitocôndrias/enzimologia , Mitocôndrias/ultraestrutura , Neurônios Motores/enzimologia , Neurônios Motores/ultraestrutura , Medula Espinal/enzimologia , Medula Espinal/ultraestrutura , Frações Subcelulares , Superóxido Dismutase/genética , Análise de Sobrevida
20.
Mol Cell Biochem ; 287(1-2): 201-11, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16583143

RESUMO

Mutations in SOD1 cause FALS by a gain of function likely related to protein misfolding and aggregation. SOD1 mutations encompass virtually every domain of the molecule, making it difficult to identify motifs important in SOD1 aggregation. Zinc binding to SOD1 is important for structural integrity, and is hypothesized to play a role in mutant SOD1 aggregation. To address this question, we mutated the unique zinc binding sites of SOD1 and examined whether these changes would influence SOD1 aggregation. We generated single and multiple mutations in SOD1 zinc binding residues (H71, H80 and D83) either alone or in combination with an aggregate forming mutation (A4V) known to cause disease. These SOD1 mutants were assayed for their ability to form aggregates. Using an in vitro cellular SOD1 aggregation assay, we show that combining A4V with mutations in non-zinc binding domains (G37R or G85R) increases SOD1 aggregation potential. Mutations at two zinc binding residues (H71G and D83G) also increase SOD1 aggregation potential. However, an H80G mutation at the third zinc binding residue decreases SOD1 aggregation potential even in the context of other aggregate forming SOD1 mutations. These results demonstrate that various mutations have different effects on SOD1 aggregation potential and that the H80G mutation appears to uniquely act as a dominant inhibitor of SOD1 aggregation.


Assuntos
Mutação , Superóxido Dismutase/genética , Esclerose Lateral Amiotrófica/etiologia , Esclerose Lateral Amiotrófica/genética , Animais , Sítios de Ligação/genética , Linhagem Celular , Dimerização , Humanos , Superóxido Dismutase/fisiologia , Superóxido Dismutase-1 , Transfecção , Zinco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA