Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 126(9): 3585-97, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27525439

RESUMO

Huntington's disease (HD) is a progressive, adult-onset neurodegenerative disease caused by a polyglutamine (polyQ) expansion in the N-terminal region of the protein huntingtin (HTT). There are no cures or disease-modifying therapies for HD. HTT has a highly conserved Akt phosphorylation site at serine 421, and prior work in HD models found that phosphorylation at S421 (S421-P) diminishes the toxicity of mutant HTT (mHTT) fragments in neuronal cultures. However, whether S421-P affects the toxicity of mHTT in vivo remains unknown. In this work, we used murine models to investigate the role of S421-P in HTT-induced neurodegeneration. Specifically, we mutated the human mHTT gene within a BAC to express either an aspartic acid or an alanine at position 421, mimicking tonic phosphorylation (mHTT-S421D mice) or preventing phosphorylation (mHTT-S421A mice), respectively. Mimicking HTT phosphorylation strongly ameliorated mHTT-induced behavioral dysfunction and striatal neurodegeneration, whereas neuronal dysfunction persisted when S421 phosphorylation was blocked. We found that S421 phosphorylation mitigates neurodegeneration by increasing proteasome-dependent turnover of mHTT and reducing the presence of a toxic mHTT conformer. These data indicate that S421 is a potent modifier of mHTT toxicity and offer in vivo validation for S421 as a therapeutic target in HD.


Assuntos
Proteína Huntingtina/genética , Doença de Huntington/genética , Serina/química , Alanina/química , Animais , Ácido Aspártico/química , Comportamento Animal , Cromossomos Artificiais Bacterianos , Modelos Animais de Doenças , Progressão da Doença , Feminino , Marcha , Genótipo , Humanos , Proteína Huntingtina/química , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Transgênicos , Mutação , Proteínas do Tecido Nervoso/genética , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/genética , Fenótipo , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo
2.
Sci Transl Med ; 6(268): 268ra178, 2014 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-25540325

RESUMO

Age-related neurodegenerative disorders including Alzheimer's disease and Huntington's disease (HD) consistently show elevated DNA damage, but the relevant molecular pathways in disease pathogenesis remain unclear. One attractive gene is that encoding the ataxia-telangiectasia mutated (ATM) protein, a kinase involved in the DNA damage response, apoptosis, and cellular homeostasis. Loss-of-function mutations in both alleles of ATM cause ataxia-telangiectasia in children, but heterozygous mutation carriers are disease-free. Persistently elevated ATM signaling has been demonstrated in Alzheimer's disease and in mouse models of other neurodegenerative diseases. We show that ATM signaling was consistently elevated in cells derived from HD mice and in brain tissue from HD mice and patients. ATM knockdown protected from toxicities induced by mutant Huntingtin (mHTT) fragments in mammalian cells and in transgenic Drosophila models. By crossing the murine Atm heterozygous null allele onto BACHD mice expressing full-length human mHTT, we show that genetic reduction of Atm gene dosage by one copy ameliorated multiple behavioral deficits and partially improved neuropathology. Small-molecule ATM inhibitors reduced mHTT-induced death of rat striatal neurons and induced pluripotent stem cells derived from HD patients. Our study provides converging genetic and pharmacological evidence that reduction of ATM signaling could ameliorate mHTT toxicity in cellular and animal models of HD, suggesting that ATM may be a useful therapeutic target for HD.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Doença de Huntington/patologia , Proteínas Mutantes/toxicidade , Proteínas do Tecido Nervoso/toxicidade , Proteínas da Membrana Plasmática de Transporte de Serotonina/toxicidade , Adulto , Idoso , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular , Citoproteção/efeitos dos fármacos , Modelos Animais de Doenças , Drosophila melanogaster/metabolismo , Dosagem de Genes , Técnicas de Silenciamento de Genes , Histonas/metabolismo , Humanos , Proteína Huntingtina , Doença de Huntington/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos Mutantes Neurológicos , Pessoa de Meia-Idade , Morfolinas/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Mudanças Depois da Morte , Transdução de Sinais/efeitos dos fármacos , Tioxantenos/farmacologia
3.
J Huntingtons Dis ; 2(3): 279-95, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-25062676

RESUMO

BACKGROUND: Huntington's disease is a neurodegenerative disorder, typically with clinical manifestations in adult years, caused by an expanded polyglutamine-coding repeat in HTT. There are no treatments that delay or prevent the onset or progression of this devastating disease. OBJECTIVE AND METHODS: In order to study its pre-symptomatic molecular progression and provide a large mammalian model for determining natural history of the disease and for therapeutic testing, we generated and previously reported on lines of transgenic sheep carrying a full length human HTT cDNA transgene, with expression driven by a minimal HTT promoter. We report here further characterization of our preferred line, OVT73. RESULTS: This line reliably expresses the expanded human huntingtin protein at modest, but readily detectable levels throughout the brain, including the striatum and cortex. Transmission of the 73 unit glutamine coding repeat was relatively stable over three generations. At the first time-point of a longitudinal study, animals sacrificed at 6 months (7 transgenic, 7 control) showed reduced striatum GABAA α1 receptor, and globus pallidus leu-enkephalin immunoreactivity. Two of three 18 month old animals sacrificed revealed cortical neuropil aggregates. Furthermore, neuronal intranuclear inclusions were identified in the piriform cortex of a single 36 month old animal in addition to cortical neuropil aggregates. CONCLUSIONS: Taken together, these data indicate that the OVT73 transgenic sheep line will progressively reveal early HD pathology and allow therapeutic testing over a period of time relevant to human patients.


Assuntos
Animais Geneticamente Modificados , Córtex Cerebral/patologia , Modelos Animais de Doenças , Doença de Huntington/patologia , Carneiro Doméstico/genética , Animais , Humanos , Proteína Huntingtina , Corpos de Inclusão Intranuclear/patologia , Proteínas do Tecido Nervoso/genética
4.
Nat Chem Biol ; 7(12): 925-34, 2011 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-22037470

RESUMO

Polyglutamine (polyQ) stretches exceeding a threshold length confer a toxic function to proteins that contain them and cause at least nine neurological disorders. The basis for this toxicity threshold is unclear. Although polyQ expansions render proteins prone to aggregate into inclusion bodies, this may be a neuronal coping response to more toxic forms of polyQ. The exact structure of these more toxic forms is unknown. Here we show that the monoclonal antibody 3B5H10 recognizes a species of polyQ protein in situ that strongly predicts neuronal death. The epitope selectively appears among some of the many low-molecular-weight conformational states assumed by expanded polyQ and disappears in higher-molecular-weight aggregated forms, such as inclusion bodies. These results suggest that protein monomers and possibly small oligomers containing expanded polyQ stretches can adopt a conformation that is recognized by 3B5H10 and is toxic or closely related to a toxic species.


Assuntos
Doenças Neurodegenerativas/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Peptídeos/química , Peptídeos/toxicidade , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Morte Celular/efeitos dos fármacos , Células Cultivadas , Epitopos/química , Epitopos/imunologia , Epitopos/toxicidade , Células HEK293 , Humanos , Corpos de Inclusão/química , Peso Molecular , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Peptídeos/imunologia , Relação Estrutura-Atividade , Expansão das Repetições de Trinucleotídeos
5.
Neuron ; 70(3): 427-40, 2011 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-21555070

RESUMO

Huntington's disease-like-2 (HDL2) is a phenocopy of Huntington's disease caused by CTG/CAG repeat expansion at the Junctophilin-3 (JPH3) locus. The mechanisms underlying HDL2 pathogenesis remain unclear. Here we developed a BAC transgenic mouse model of HDL2 (BAC-HDL2) that exhibits progressive motor deficits, selective neurodegenerative pathology, and ubiquitin-positive nuclear inclusions (NIs). Molecular analyses reveal a promoter at the transgene locus driving the expression of a CAG repeat transcript (HDL2-CAG) from the strand antisense to JPH3, which encodes an expanded polyglutamine (polyQ) protein. Importantly, BAC-HDL2 mice, but not control BAC mice, accumulate polyQ-containing NIs in a pattern strikingly similar to those in the patients. Furthermore, BAC mice with genetic silencing of the expanded CUG transcript still express HDL2-CAG transcript and manifest polyQ pathogenesis. Finally, studies of HDL2 mice and patients revealed CBP sequestration into NIs and evidence for interference of CBP-mediated transcriptional activation. These results suggest overlapping polyQ-mediated pathogenic mechanisms in HD and HDL2.


Assuntos
Doença de Huntington , Proteínas de Membrana/genética , Oligodesoxirribonucleotídeos Antissenso/metabolismo , Peptídeos/toxicidade , Expansão das Repetições de Trinucleotídeos/genética , Fatores Etários , Análise de Variância , Animais , Células Cultivadas , Córtex Cerebral/citologia , Imunoprecipitação da Cromatina/métodos , Modelos Animais de Doenças , Embrião de Mamíferos , Regulação da Expressão Gênica/genética , Humanos , Doença de Huntington/tratamento farmacológico , Doença de Huntington/genética , Doença de Huntington/fisiopatologia , Corpos de Inclusão Intranuclear/metabolismo , Corpos de Inclusão Intranuclear/patologia , Camundongos , Camundongos Transgênicos , Atividade Motora/genética , Neurônios/metabolismo , Tamanho do Órgão/genética , Peptídeos/genética , Fatores de Tempo , Transfecção , Ubiquitina/metabolismo
6.
Hum Mol Genet ; 19(19): 3702-20, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20616151

RESUMO

The aggregation of mutant polyglutamine (polyQ) proteins has sparked interest in the role of protein quality-control pathways in Huntington's disease (HD) and related polyQ disorders. Employing a novel knock-in HD mouse model, we provide in vivo evidence of early, sustained alterations of autophagy in response to mutant huntingtin (mhtt). The HdhQ200 knock-in model, derived from the selective breeding of HdhQ150 knock-in mice, manifests an accelerated and more robust phenotype than the parent line. Heterozygous HdhQ200 mice accumulate htt aggregates as cytoplasmic aggregation foci (AF) as early as 9 weeks of age and striatal neuronal intranuclear inclusions (NIIs) by 20 weeks. By 40 weeks, striatal AF are perinuclear and immunoreactive for ubiquitin and the autophagosome marker LC3. Striatal NIIs accumulate earlier in HdhQ200 mice than in HdhQ150 mice. The earlier appearance of aggregate pathology in HdhQ200 mice is paralleled by earlier and more rapidly progressive motor deficits: progressive imbalance and decreased motor coordination by 50 weeks, gait deficits by 60 weeks and gross motor impairment by 80 weeks of age. At 80 weeks, heterozygous HdhQ200 mice exhibit striatal and cortical astrogliosis and a approximately 50% reduction in striatal dopamine receptor binding. Increased LC3-II protein expression, which is noted early and sustained throughout the disease course, is paralleled by increased expression of the autophagy-related protein, p62. Early and sustained expression of autophagy-related proteins in this genetically precise mouse model of HD suggests that the alteration of autophagic flux is an important and early component of the neuronal response to mhtt.


Assuntos
Autofagia , Técnicas de Introdução de Genes , Doença de Huntington/genética , Doença de Huntington/patologia , Animais , Biomarcadores/metabolismo , Modelos Animais de Doenças , Proteína Glial Fibrilar Ácida/metabolismo , Saúde , Heterozigoto , Doença de Huntington/fisiopatologia , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Atividade Motora , Mutação/genética , Neostriado/patologia , Neostriado/fisiopatologia , Neostriado/ultraestrutura , Neurônios/patologia , Neurônios/ultraestrutura , Estrutura Quaternária de Proteína , Transporte Proteico , Receptores Dopaminérgicos/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/química , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Ubiquitina/metabolismo
7.
J Biol Chem ; 285(12): 8808-23, 2010 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-20086007

RESUMO

Huntingtin proteolysis has been implicated in the molecular pathogenesis of Huntington disease (HD). Despite an intense effort, the identity of the pathogenic smallest N-terminal fragment has not been determined. Using a panel of anti-huntingtin antibodies, we employed an unbiased approach to generate proteolytic cleavage maps of mutant and wild-type huntingtin in the HdhQ150 knock-in mouse model of HD. We identified 14 prominent N-terminal fragments, which, in addition to the full-length protein, can be readily detected in cytoplasmic but not nuclear fractions. These fragments were detected at all ages and are not a consequence of the pathogenic process. We demonstrated that the smallest fragment is an exon 1 huntingtin protein, known to contain a potent nuclear export signal. Prior to the onset of behavioral phenotypes, the exon 1 protein, and possibly other small fragments, accumulate in neuronal nuclei in the form of a detergent insoluble complex, visualized as diffuse granular nuclear staining in tissue sections. This methodology can be used to validate the inhibition of specific proteases as therapeutic targets for HD by pharmacological or genetic approaches.


Assuntos
Doença de Huntington/metabolismo , Mutação , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Proteínas Nucleares/genética , Animais , Células COS , Calpaína/química , Núcleo Celular/metabolismo , Chlorocebus aethiops , Citoplasma/metabolismo , Modelos Animais de Doenças , Éxons , Genótipo , Proteína Huntingtina , Camundongos , Estrutura Terciária de Proteína
8.
Neuron ; 64(6): 828-40, 2009 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-20064390

RESUMO

The N-terminal 17 amino acids of huntingtin (NT17) can be phosphorylated on serines 13 and 16; however, the significance of these modifications in Huntington's disease pathogenesis remains unknown. In this study, we developed BAC transgenic mice expressing full-length mutant huntingtin (fl-mhtt) with serines 13 and 16 mutated to either aspartate (phosphomimetic or SD) or alanine (phosphoresistant or SA). Both mutant proteins preserve the essential function of huntingtin in rescuing knockout mouse phenotypes. However, fl-mhtt-induced disease pathogenesis, including motor and psychiatric-like behavioral deficits, mhtt aggregation, and selective neurodegeneration are abolished in SD but preserved in SA mice. Moreover, modification of these serines in expanded repeat huntingtin peptides modulates aggregation and amyloid fibril formation in vitro. Together, our findings demonstrate that serines 13 and 16 are critical determinants of fl-mhtt-induced disease pathogenesis in vivo, supporting the targeting of huntingtin NT17 domain and its modifications in HD therapy.


Assuntos
Predisposição Genética para Doença/genética , Doença de Huntington/genética , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Serina/química , Serina/metabolismo , Alanina/química , Alanina/genética , Alanina/metabolismo , Sequência de Aminoácidos/genética , Substituição de Aminoácidos/genética , Amiloide/genética , Amiloide/metabolismo , Animais , Ácido Aspártico/química , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/genética , Humanos , Proteína Huntingtina , Doença de Huntington/metabolismo , Doença de Huntington/fisiopatologia , Camundongos , Camundongos Transgênicos , Peso Molecular , Mutação/genética , Degeneração Neural/genética , Degeneração Neural/metabolismo , Degeneração Neural/fisiopatologia , Fenótipo , Estrutura Terciária de Proteína/genética , Serina/genética , Expansão das Repetições de Trinucleotídeos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...