Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 114(23): E4676-E4685, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28533375

RESUMO

The activity of the transcription factor nuclear factor-erythroid 2 p45-derived factor 2 (NRF2) is orchestrated and amplified through enhanced transcription of antioxidant and antiinflammatory target genes. The present study has characterized a triazole-containing inducer of NRF2 and elucidated the mechanism by which this molecule activates NRF2 signaling. In a highly selective manner, the compound covalently modifies a critical stress-sensor cysteine (C151) of the E3 ligase substrate adaptor protein Kelch-like ECH-associated protein 1 (KEAP1), the primary negative regulator of NRF2. We further used this inducer to probe the functional consequences of selective activation of NRF2 signaling in Huntington's disease (HD) mouse and human model systems. Surprisingly, we discovered a muted NRF2 activation response in human HD neural stem cells, which was restored by genetic correction of the disease-causing mutation. In contrast, selective activation of NRF2 signaling potently repressed the release of the proinflammatory cytokine IL-6 in primary mouse HD and WT microglia and astrocytes. Moreover, in primary monocytes from HD patients and healthy subjects, NRF2 induction repressed expression of the proinflammatory cytokines IL-1, IL-6, IL-8, and TNFα. Together, our results demonstrate a multifaceted protective potential of NRF2 signaling in key cell types relevant to HD pathology.


Assuntos
Doença de Huntington/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Adulto , Idoso , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Doença de Huntington/genética , Proteína 1 Associada a ECH Semelhante a Kelch/química , Intoxicação por MPTP/metabolismo , Intoxicação por MPTP/prevenção & controle , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Pessoa de Meia-Idade , Fator 2 Relacionado a NF-E2/química , Células-Tronco Neurais/metabolismo , Fármacos Neuroprotetores/farmacologia , Conformação Proteica/efeitos dos fármacos , Ratos , Transdução de Sinais
2.
Cell Chem Biol ; 23(7): 849-861, 2016 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-27427231

RESUMO

There are currently no disease-modifying therapies for the neurodegenerative disorder Huntington's disease (HD). This study identified novel thiazole-containing inhibitors of the deacetylase sirtuin-2 (SIRT2) with neuroprotective activity in ex vivo brain slice and Drosophila models of HD. A systems biology approach revealed an additional SIRT2-independent property of the lead-compound, MIND4, as an inducer of cytoprotective NRF2 (nuclear factor-erythroid 2 p45-derived factor 2) activity. Structure-activity relationship studies further identified a potent NRF2 activator (MIND4-17) lacking SIRT2 inhibitory activity. MIND compounds induced NRF2 activation responses in neuronal and non-neuronal cells and reduced production of reactive oxygen species and nitrogen intermediates. These drug-like thiazole-containing compounds represent an exciting opportunity for development of multi-targeted agents with potentially synergistic therapeutic benefits in HD and related disorders.


Assuntos
Modelos Animais de Doenças , Doença de Huntington/tratamento farmacológico , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Sirtuína 2/antagonistas & inibidores , Tiazóis/farmacologia , Tiazóis/uso terapêutico , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Drosophila , Doença de Huntington/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/uso terapêutico , Ratos , Sirtuína 2/deficiência , Sirtuína 2/metabolismo , Relação Estrutura-Atividade , Tiazóis/química
3.
Sci Rep ; 6: 25626, 2016 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-27172999

RESUMO

We previously reported neuroprotective activity of the botanical anti-cancer drug candidate PBI-05204, a supercritical CO2 extract of Nerium oleander, in brain slice and in vivo models of ischemic stroke. We showed that one component of this neuroprotective activity is mediated through its principal cardiac glycoside constituent, oleandrin, via induction of the potent neurotrophic factor brain-derived neurotrophic factor (BDNF). However, we also noted that the concentration-relation for PBI-05204 in the brain slice oxygen-glucose deprivation (OGD) model is considerably broader than that for oleandrin as a single agent. We thus surmised that PBI-05204 contains an additional neuroprotective component(s), distinct from oleandrin. We report here that neuroprotective activity is also provided by the triterpenoid constituents of PBI-05204, notably oleanolic acid. We demonstrate that a sub-fraction of PBI-05204 (Fraction 0-4) containing oleanolic and other triterpenoids, but without cardiac glycosides, induces the expression of cellular antioxidant gene transcription programs regulated through antioxidant transcriptional response elements (AREs). Finally, we show that Fraction 0-4 provides broad neuroprotection in organotypic brain slice models for neurodegeneration driven by amyloid precursor protein (APP) and tau implicated in Alzheimer's disease and frontotemporal dementias, respectively, in addition to ischemic injury modeled by OGD.


Assuntos
Antineoplásicos/farmacologia , Encéfalo/efeitos dos fármacos , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Doenças Neurodegenerativas/tratamento farmacológico , Extratos Vegetais/farmacologia , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Antineoplásicos/química , Encéfalo/metabolismo , Encéfalo/patologia , Fracionamento Químico/métodos , Modelos Animais de Doenças , Feminino , Glucose/metabolismo , Humanos , Masculino , Nerium/química , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Ácido Oleanólico/química , Ácido Oleanólico/farmacologia , Técnicas de Cultura de Órgãos , Oxigênio/metabolismo , Ratos Sprague-Dawley
4.
J Biol Chem ; 289(10): 6709-6726, 2014 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-24407293

RESUMO

Huntington disease (HD) is an inherited neurodegenerative disease caused by a CAG expansion in the HTT gene. Using yeast two-hybrid methods, we identified a large set of proteins that interact with huntingtin (HTT)-interacting proteins. This network, composed of HTT-interacting proteins (HIPs) and proteins interacting with these primary nodes, contains 3235 interactions among 2141 highly interconnected proteins. Analysis of functional annotations of these proteins indicates that primary and secondary HIPs are enriched in pathways implicated in HD, including mammalian target of rapamycin, Rho GTPase signaling, and oxidative stress response. To validate roles for HIPs in mutant HTT toxicity, we show that the Rho GTPase signaling components, BAIAP2, EZR, PIK3R1, PAK2, and RAC1, are modifiers of mutant HTT toxicity. We also demonstrate that Htt co-localizes with BAIAP2 in filopodia and that mutant HTT interferes with filopodial dynamics. These data indicate that HTT is involved directly in membrane dynamics, cell attachment, and motility. Furthermore, they implicate dysregulation in these pathways as pathological mechanisms in HD.


Assuntos
Doença de Huntington/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Células HEK293 , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Redes e Vias Metabólicas , Camundongos , Células NIH 3T3 , Proteínas do Tecido Nervoso/genética , Pseudópodes/metabolismo
5.
Nat Med ; 19(8): 1030-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23852340

RESUMO

Huntington's disease is caused by an expanded polyglutamine repeat in the huntingtin protein (HTT), but the pathophysiological sequence of events that trigger synaptic failure and neuronal loss are not fully understood. Alterations in N-methyl-D-aspartate (NMDA)-type glutamate receptors (NMDARs) have been implicated. Yet, it remains unclear how the HTT mutation affects NMDAR function, and direct evidence for a causative role is missing. Here we show that mutant HTT redirects an intracellular store of juvenile NMDARs containing GluN3A subunits to the surface of striatal neurons by sequestering and disrupting the subcellular localization of the endocytic adaptor PACSIN1, which is specific for GluN3A. Overexpressing GluN3A in wild-type mouse striatum mimicked the synapse loss observed in Huntington's disease mouse models, whereas genetic deletion of GluN3A prevented synapse degeneration, ameliorated motor and cognitive decline and reduced striatal atrophy and neuronal loss in the YAC128 Huntington's disease mouse model. Furthermore, GluN3A deletion corrected the abnormally enhanced NMDAR currents, which have been linked to cell death in Huntington's disease and other neurodegenerative conditions. Our findings reveal an early pathogenic role of GluN3A dysregulation in Huntington's disease and suggest that therapies targeting GluN3A or pathogenic HTT-PACSIN1 interactions might prevent or delay disease progression.


Assuntos
Comportamento Animal , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/metabolismo , Morte Celular/efeitos dos fármacos , Proteínas do Citoesqueleto , Modelos Animais de Doenças , Deleção de Genes , Células HEK293 , Humanos , Doença de Huntington/fisiopatologia , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Atividade Motora/efeitos dos fármacos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas Mutantes/toxicidade , Neostriado/metabolismo , Neostriado/patologia , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Neuropeptídeos/metabolismo , Fosfoproteínas/metabolismo , Ligação Proteica/efeitos dos fármacos , Estrutura Quaternária de Proteína , Teste de Desempenho do Rota-Rod , Sinapses/efeitos dos fármacos , Sinapses/ultraestrutura
6.
Neurotherapeutics ; 10(3): 400-15, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23700210

RESUMO

Huntington's disease (HD) typifies a class of inherited neurodegenerative disorders in which a CAG expansion in a single gene leads to an extended polyglutamine tract and misfolding of the expressed protein, driving cumulative neural dysfunction and degeneration. HD is invariably fatal with symptoms that include progressive neuropsychiatric and cognitive impairments, and eventual motor disability. No curative therapies yet exist for HD and related polyglutamine diseases; therefore, substantial efforts have been made in the drug discovery field to identify potential drug and drug target candidates for disease-modifying treatment. In this context, we review here a range of early-stage screening approaches based in in vitro, cellular, and invertebrate models to identify pharmacological and genetic modifiers of polyglutamine aggregation and induced neurodegeneration. In addition, emerging technologies, including high-content analysis, three-dimensional culture models, and induced pluripotent stem cells are increasingly being incorporated into drug discovery screening pipelines for protein misfolding disorders. Together, these diverse screening strategies are generating novel and exciting new probes for understanding the disease process and for furthering development of therapeutic candidates for eventual testing in the clinical setting.


Assuntos
Doença de Huntington , Doenças Neurodegenerativas , Peptídeos/metabolismo , Deficiências na Proteostase/complicações , Animais , Modelos Animais de Doenças , Humanos , Proteína Huntingtina , Doença de Huntington/complicações , Doença de Huntington/etiologia , Doença de Huntington/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Peptídeos/genética , Deficiências na Proteostase/genética
7.
Chem Biol ; 19(2): 264-75, 2012 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-22365609

RESUMO

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by the amplification of a polyglutamine stretch at the N terminus of the huntingtin protein. N-terminal fragments of the mutant huntingtin (mHtt) aggregate and form intracellular inclusions in brain and peripheral tissues. Aggregates are an important hallmark of the disease, translating into a high need to quantify them in vitro and in vivo. We developed a one-step TR-FRET-based immunoassay to quantify soluble and aggregated mHtt in cell and tissue homogenates. Strikingly, quantification revealed a decrease of soluble mHtt correlating with an increase of aggregated protein in primary neuronal cell cultures, transgenic R6/2, and HdhQ150 knock-in HD mice. These results emphasize the assay's efficiency for highly sensitive and quantitative detection of soluble and aggregated mHtt and its application in high-throughput screening and characterization of HD models.


Assuntos
Doença de Huntington/metabolismo , Imunoensaio , Proteínas do Tecido Nervoso/análise , Proteínas Nucleares/análise , Animais , Células Cultivadas , Transferência Ressonante de Energia de Fluorescência , Técnicas de Introdução de Genes , Proteína Huntingtina , Doença de Huntington/patologia , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo
8.
Neurobiol Aging ; 33(3): 621.e17-33, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21482444

RESUMO

In Huntington's disease (HD), mutated huntingtin (mhtt) causes striatal neurodegeneration which is paralleled by elevated microglia cell numbers. In vitro corticostriatal slice and primary neuronal culture models, in which neuronal expression of mhtt fragments drives HD-like neurotoxicity, were employed to examine wild type microglia during both the initiation and progression of neuronal pathology. As neuronal pathology progressed, microglia initially localized in the vicinity of neurons expressing mhtt fragments increased in number, demonstrated morphological evidence of activation, and expressed the proliferation marker, Ki67. These microglia were positioned along irregular neurites, but did not localize with mhtt inclusions nor exacerbate mhtt fragment-induced neurotoxicity. Prior to neuronal pathology, microglia upregulated ionized calcium binding adaptor molecule 1 (Iba1), signaling a functional shift. With neurodegeneration, interleukin-6 and complement component 1q were increased. The results suggest a stimulatory, proliferative signal for microglia present at the onset of mhtt fragment-induced neurodegeneration. Thus, microglia effect a localized inflammatory response to neuronal mhtt expression that may serve to direct microglial removal of dysfunctional neurites or aberrant synapses, as is required for reparative actions in vivo.


Assuntos
Proliferação de Células , Doença de Huntington/patologia , Microglia/patologia , Mutação/genética , Proteínas do Tecido Nervoso/genética , Neuritos/patologia , Neurônios/patologia , Proteínas Nucleares/genética , Animais , Animais Recém-Nascidos , Morte Celular/genética , Células Cultivadas , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/metabolismo , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Degeneração Neural/genética , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Proteínas do Tecido Nervoso/biossíntese , Neuritos/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/biossíntese , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley , Sinapses/genética , Sinapses/patologia
9.
Neurobiol Dis ; 43(1): 248-56, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21458569

RESUMO

Huntington's disease (HD) is a late-onset, neurodegenerative disease for which there are currently no cures nor disease-modifying treatments. Here we report the identification of several potential anti-inflammatory targets for HD using an ex vivo model of HD that involves the acute transfection of human mutant huntingtin-based constructs into rat brain slices. This model recapitulates key components of the human disease, including the formation of intracellular huntingtin protein (HTT)-containing inclusions and the progressive neurodegeneration of striatal neurons-both occurring within the native tissue context of these neurons. Using this "high-throughput biology" screening platform, we conducted a hypothesis-neutral screen of a collection of drug-like compounds which identified several anti-inflammatory targets that provided neuroprotection against HTT fragment-induced neurodegeneration. The nature of these targets provide further support for non-cell autonomous mechanisms mediating significant aspects of neuropathogenesis induced by mutant HTT fragment proteins.


Assuntos
Anti-Inflamatórios não Esteroides/metabolismo , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Doença de Huntington/tratamento farmacológico , Degeneração Neural/tratamento farmacológico , Animais , Animais Recém-Nascidos , Anti-Inflamatórios não Esteroides/farmacologia , Corpo Estriado/patologia , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley
10.
PLoS One ; 6(1): e16676, 2011 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-21304966

RESUMO

BACKGROUND: Proteolytic processing of mutant huntingtin (mHtt), the protein that causes Huntington's disease (HD), is critical for mHtt toxicity and disease progression. mHtt contains several caspase and calpain cleavage sites that generate N-terminal fragments that are more toxic than full-length mHtt. Further processing is then required for the degradation of these fragments, which in turn, reduces toxicity. This unknown, secondary degradative process represents a promising therapeutic target for HD. METHODOLOGY/PRINCIPAL FINDINGS: We have used intrabodies, intracellularly expressed antibody fragments, to gain insight into the mechanism of mutant huntingtin exon 1 (mHDx-1) clearance. Happ1, an intrabody recognizing the proline-rich region of mHDx-1, reduces the level of soluble mHDx-1 by increasing clearance. While proteasome and macroautophagy inhibitors reduce turnover of mHDx-1, Happ1 is still able to reduce mHDx-1 under these conditions, indicating Happ1-accelerated mHDx-1 clearance does not rely on these processes. In contrast, a calpain inhibitor or an inhibitor of lysosomal pH block Happ1-mediated acceleration of mHDx-1 clearance. These results suggest that mHDx-1 is cleaved by calpain, likely followed by lysosomal degradation and this process regulates the turnover rate of mHDx-1. Sequence analysis identifies amino acid (AA) 15 as a potential calpain cleavage site. Calpain cleavage of recombinant mHDx-1 in vitro yields fragments of sizes corresponding to this prediction. Moreover, when the site is blocked by binding of another intrabody, V(L)12.3, turnover of soluble mHDx-1 in living cells is blocked. CONCLUSIONS/SIGNIFICANCE: These results indicate that calpain-mediated removal of the 15 N-terminal AAs is required for the degradation of mHDx-1, a finding that may have therapeutic implications.


Assuntos
Calpaína/metabolismo , Éxons/genética , Fragmentos de Imunoglobulinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Sequência de Aminoácidos , Animais , Proteína Huntingtina , Camundongos , Proteínas Mutantes/imunologia , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Estabilidade Proteica , Solubilidade
11.
Chem Biol ; 17(11): 1189-200, 2010 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21095569

RESUMO

Huntington's Disease (HD) is characterized by a mutation in the huntingtin (Htt) gene encoding an expansion of glutamine repeats on the N terminus of the Htt protein. Numerous studies have identified Htt proteolysis as a critical pathological event in HD postmortem human tissue and mouse HD models, and proteases known as caspases have emerged as attractive HD therapeutic targets. We report the use of the substrate activity screening method against caspase-3 and -6 to identify three novel, pan-caspase inhibitors that block proteolysis of Htt at caspase-3 and -6 cleavage sites. In HD models these irreversible inhibitors suppressed Hdh(111Q/111Q)-mediated toxicity and rescued rat striatal and cortical neurons from cell death. In this study, the identified nonpeptidic caspase inhibitors were used to confirm the role of caspase-mediated Htt proteolysis in HD. These results further implicate caspases as promising targets for HD therapeutic development.


Assuntos
Inibidores de Caspase , Inibidores de Cisteína Proteinase/química , Doença de Huntington/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/química , Animais , Apoptose , Caspase 3/metabolismo , Caspase 6/metabolismo , Células Cultivadas , Cumarínicos/química , Cumarínicos/uso terapêutico , Inibidores de Cisteína Proteinase/síntese química , Inibidores de Cisteína Proteinase/uso terapêutico , Modelos Animais de Doenças , Humanos , Proteína Huntingtina , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ratos , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/uso terapêutico , Relação Estrutura-Atividade , Especificidade por Substrato
12.
J Biomol Screen ; 15(7): 806-19, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20581077

RESUMO

Huntington's disease (HD) is a fatal neurodegenerative disease characterized by progressive cognitive, behavioral, and motor deficits and caused by expansion of a polyglutamine repeat in the Huntingtin protein (Htt). Despite its monogenic nature, HD pathogenesis includes obligatory non-cell-autonomous pathways involving both the cortex and the striatum, and therefore effective recapitulation of relevant HD disease pathways in cell lines and primary neuronal monocultures is intrinsically limited. To address this, the authors developed an automated high-content imaging screen in high-density primary cultures of cortical and striatal neurons together with supporting glial cells. Cortical and striatal neurons are transfected separately with different fluorescent protein markers such that image-based high-content analysis can be used to assay these neuronal populations separately but still supporting their intercellular interactions, including abundant synaptic interconnectivity. This assay was reduced to practice using transfection of a mutant N-terminal Htt domain and validated via a screen of ~400 selected small molecules. Both expected as well as novel candidate targets for HD emerged from this screen; of particular interest were target classes with close relative proximity to clinical testing. These findings suggest that composite primary cultures incorporating increased levels of biological complexity can be used for high-content imaging and "high-context" screening to represent molecular targets that otherwise may be operant only in the complex tissue environment found in vivo during disease pathogenesis.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Doença de Huntington/patologia , Neurônios/patologia , Animais , Biomarcadores/metabolismo , Células Cultivadas , Córtex Cerebral/patologia , Técnicas de Cocultura , AMP Cíclico/metabolismo , Inibidores Enzimáticos/análise , Inibidores Enzimáticos/farmacologia , Humanos , Doença de Huntington/metabolismo , Proteínas Luminescentes/metabolismo , Camundongos , Neostriado/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/análise , Bibliotecas de Moléculas Pequenas/farmacologia
13.
J Cell Biol ; 187(7): 1083-99, 2009 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-20026656

RESUMO

Expansion of the polyglutamine repeat within the protein Huntingtin (Htt) causes Huntington's disease, a neurodegenerative disease associated with aging and the accumulation of mutant Htt in diseased neurons. Understanding the mechanisms that influence Htt cellular degradation may target treatments designed to activate mutant Htt clearance pathways. We find that Htt is phosphorylated by the inflammatory kinase IKK, enhancing its normal clearance by the proteasome and lysosome. Phosphorylation of Htt regulates additional post-translational modifications, including Htt ubiquitination, SUMOylation, and acetylation, and increases Htt nuclear localization, cleavage, and clearance mediated by lysosomal-associated membrane protein 2A and Hsc70. We propose that IKK activates mutant Htt clearance until an age-related loss of proteasome/lysosome function promotes accumulation of toxic post-translationally modified mutant Htt. Thus, IKK activation may modulate mutant Htt neurotoxicity depending on the cell's ability to degrade the modified species.


Assuntos
Quinase I-kappa B/fisiologia , Lisossomos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Encéfalo/metabolismo , Linhagem Celular , Humanos , Proteína Huntingtina , Camundongos , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/química , Proteínas Nucleares/análise , Proteínas Nucleares/química , Fosforilação , Estrutura Terciária de Proteína , Ratos , Ratos Sprague-Dawley , Solubilidade , Ubiquitinação
14.
PLoS Genet ; 3(5): e82, 2007 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-17500595

RESUMO

Huntington's disease (HD) is a fatal neurodegenerative condition caused by expansion of the polyglutamine tract in the huntingtin (Htt) protein. Neuronal toxicity in HD is thought to be, at least in part, a consequence of protein interactions involving mutant Htt. We therefore hypothesized that genetic modifiers of HD neurodegeneration should be enriched among Htt protein interactors. To test this idea, we identified a comprehensive set of Htt interactors using two complementary approaches: high-throughput yeast two-hybrid screening and affinity pull down followed by mass spectrometry. This effort led to the identification of 234 high-confidence Htt-associated proteins, 104 of which were found with the yeast method and 130 with the pull downs. We then tested an arbitrary set of 60 genes encoding interacting proteins for their ability to behave as genetic modifiers of neurodegeneration in a Drosophila model of HD. This high-content validation assay showed that 27 of 60 orthologs tested were high-confidence genetic modifiers, as modification was observed with more than one allele. The 45% hit rate for genetic modifiers seen among the interactors is an order of magnitude higher than the 1%-4% typically observed in unbiased genetic screens. Genetic modifiers were similarly represented among proteins discovered using yeast two-hybrid and pull-down/mass spectrometry methods, supporting the notion that these complementary technologies are equally useful in identifying biologically relevant proteins. Interacting proteins confirmed as modifiers of the neurodegeneration phenotype represent a diverse array of biological functions, including synaptic transmission, cytoskeletal organization, signal transduction, and transcription. Among the modifiers were 17 loss-of-function suppressors of neurodegeneration, which can be considered potential targets for therapeutic intervention. Finally, we show that seven interacting proteins from among 11 tested were able to co-immunoprecipitate with full-length Htt from mouse brain. These studies demonstrate that high-throughput screening for protein interactions combined with genetic validation in a model organism is a powerful approach for identifying novel candidate modifiers of polyglutamine toxicity.


Assuntos
Degeneração Neural/genética , Degeneração Neural/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Animais , Drosophila melanogaster/efeitos dos fármacos , Humanos , Proteína Huntingtina , Imunoprecipitação , Camundongos , Modelos Neurológicos , Peptídeos/toxicidade , Ligação Proteica , Mapeamento de Interação de Proteínas , Reprodutibilidade dos Testes
15.
Dev Dyn ; 234(2): 346-54, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16127716

RESUMO

FoxA transcription factors are central regulators of gut development in all animals that have been studied. Here we examine the sole Caenorhabditis elegans FoxA protein, which is called pha-4. We describe the molecular characterization of five pha-4 mutations and characterize their associated phenotypes. Two nonsense mutations are predicted to truncate PHA-4 after the DNA binding domain and remove the conserved carboxyl terminus. Surprisingly, animals harboring these mutations are viable, provided the mutant mRNAs are stabilized by inactivating the nonsense-mediated decay pathway. Two additional nonsense mutations reveal that the DNA binding domain is critical for activity. A missense mutation predicted to alter the PHA-4 amino terminus leads to a dramatic reduction in pha-4 activity even though the protein is expressed appropriately. We suggest that the PHA-4 amino terminus is essential for PHA-4 function in vivo, possibly as a transactivation domain, and can compensate for loss of the carboxyl terminus. We also provide evidence for autoregulation by PHA-4.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/fisiologia , Transativadores/genética , Transativadores/fisiologia , Sequência de Aminoácidos , Animais , Caenorhabditis elegans , Núcleo Celular/metabolismo , Sobrevivência Celular , DNA/química , Heterozigoto , Homozigoto , Modelos Genéticos , Dados de Sequência Molecular , Mutação , Mutação de Sentido Incorreto , Fenótipo , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA