Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 145(6): 863-74, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21640374

RESUMO

Metabolites in the kynurenine pathway, generated by tryptophan degradation, are thought to play an important role in neurodegenerative disorders, including Alzheimer's and Huntington's diseases. In these disorders, glutamate receptor-mediated excitotoxicity and free radical formation have been correlated with decreased levels of the neuroprotective metabolite kynurenic acid. Here, we describe the synthesis and characterization of JM6, a small-molecule prodrug inhibitor of kynurenine 3-monooxygenase (KMO). Chronic oral administration of JM6 inhibits KMO in the blood, increasing kynurenic acid levels and reducing extracellular glutamate in the brain. In a transgenic mouse model of Alzheimer's disease, JM6 prevents spatial memory deficits, anxiety-related behavior, and synaptic loss. JM6 also extends life span, prevents synaptic loss, and decreases microglial activation in a mouse model of Huntington's disease. These findings support a critical link between tryptophan metabolism in the blood and neurodegeneration, and they provide a foundation for treatment of neurodegenerative diseases.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Huntington/tratamento farmacológico , Ácido Cinurênico/análise , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Sulfonamidas/uso terapêutico , Tiazóis/uso terapêutico , Administração Oral , Doença de Alzheimer/fisiopatologia , Animais , Química Encefálica , Modelos Animais de Doenças , Feminino , Humanos , Ácido Cinurênico/sangue , Masculino , Camundongos , Camundongos Transgênicos , Sulfonamidas/administração & dosagem , Tiazóis/administração & dosagem
2.
Biochim Biophys Acta ; 1860(11 Pt A): 2345-2354, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27392942

RESUMO

BACKGROUND: In mammals, the majority of the essential amino acid tryptophan is degraded via the kynurenine pathway (KP). Several KP metabolites play distinct physiological roles, often linked to immune system functions, and may also be causally involved in human diseases including neurodegenerative disorders, schizophrenia and cancer. Pharmacological manipulation of the KP has therefore become an active area of drug development. To target the pathway effectively, it is important to understand how specific KP enzymes control levels of the bioactive metabolites in vivo. METHODS: Here, we conducted a comprehensive biochemical characterization of mice with a targeted deletion of either tryptophan 2,3-dioxygenase (TDO) or indoleamine 2,3-dioxygenase (IDO), the two initial rate-limiting enzymes of the KP. These enzymes catalyze the same reaction, but differ in biochemical characteristics and expression patterns. We measured KP metabolite levels and enzyme activities and expression in several tissues in basal and immune-stimulated conditions. RESULTS AND CONCLUSIONS: Although our study revealed several unexpected downstream effects on KP metabolism in both knockout mice, the results were essentially consistent with TDO-mediated control of basal KP metabolism and a role of IDO in phenomena involving stimulation of the immune system.


Assuntos
Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Cinurenina/metabolismo , Triptofano Oxigenase/metabolismo , Animais , Encéfalo/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Inflamação/etiologia , Inflamação/metabolismo , Cinurenina/sangue , Lipopolissacarídeos/toxicidade , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Triptofano Oxigenase/genética
3.
Nat Rev Neurosci ; 13(7): 465-77, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22678511

RESUMO

The essential amino acid tryptophan is not only a precursor of serotonin but is also degraded to several other neuroactive compounds, including kynurenic acid, 3-hydroxykynurenine and quinolinic acid. The synthesis of these metabolites is regulated by an enzymatic cascade, known as the kynurenine pathway, that is tightly controlled by the immune system. Dysregulation of this pathway, resulting in hyper-or hypofunction of active metabolites, is associated with neurodegenerative and other neurological disorders, as well as with psychiatric diseases such as depression and schizophrenia. With recently developed pharmacological agents, it is now possible to restore metabolic equilibrium and envisage novel therapeutic interventions.


Assuntos
Encéfalo/patologia , Encéfalo/fisiologia , Cinurenina/metabolismo , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/patologia , Transdução de Sinais/fisiologia , Animais , Humanos
4.
J Biol Chem ; 288(51): 36554-66, 2013 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-24189070

RESUMO

Kynurenine 3-monooxygenase (KMO), a pivotal enzyme in the kynurenine pathway (KP) of tryptophan degradation, has been suggested to play a major role in physiological and pathological events involving bioactive KP metabolites. To explore this role in greater detail, we generated mice with a targeted genetic disruption of Kmo and present here the first biochemical and neurochemical characterization of these mutant animals. Kmo(-/-) mice lacked KMO activity but showed no obvious abnormalities in the activity of four additional KP enzymes tested. As expected, Kmo(-/-) mice showed substantial reductions in the levels of its enzymatic product, 3-hydroxykynurenine, in liver, brain, and plasma. Compared with wild-type animals, the levels of the downstream metabolite quinolinic acid were also greatly decreased in liver and plasma of the mutant mice but surprisingly were only slightly reduced (by ∼20%) in the brain. The levels of three other KP metabolites: kynurenine, kynurenic acid, and anthranilic acid, were substantially, but differentially, elevated in the liver, brain, and plasma of Kmo(-/-) mice, whereas the liver and brain content of the major end product of the enzymatic cascade, NAD(+), did not differ between Kmo(-/-) and wild-type animals. When assessed by in vivo microdialysis, extracellular kynurenic acid levels were found to be significantly elevated in the brains of Kmo(-/-) mice. Taken together, these results provide further evidence that KMO plays a key regulatory role in the KP and indicate that Kmo(-/-) mice will be useful for studying tissue-specific functions of individual KP metabolites in health and disease.


Assuntos
Encéfalo/metabolismo , Deleção de Genes , Técnicas de Inativação de Genes , Quinurenina 3-Mono-Oxigenase/genética , Cinurenina/metabolismo , Animais , Cinurenina/análogos & derivados , Cinurenina/sangue , Quinurenina 3-Mono-Oxigenase/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Knockout , Especificidade de Órgãos , Triptofano/metabolismo
5.
Hum Mol Genet ; 21(11): 2432-49, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22357655

RESUMO

The aggregation of α-synuclein (αSyn) is a neuropathologic hallmark of Parkinson's disease and other synucleinopathies. In Lewy bodies, αSyn is extensively phosphorylated, predominantly at serine 129 (S129). Recent studies in yeast have shown that, at toxic levels, αSyn disrupts Rab homeostasis, causing an initial endoplasmic reticulum-to-Golgi block that precedes a generalized trafficking collapse. However, whether αSyn phosphorylation modulates trafficking defects has not been evaluated. Here, we show that constitutive expression of αSyn in yeast impairs late-exocytic, early-endocytic and/or recycling trafficking. Although members of the casein kinase I (CKI) family phosphorylate αSyn at S129, they attenuate αSyn toxicity and trafficking defects by an S129 phosphorylation-independent mechanism. Surprisingly, phosphorylation of S129 modulates αSyn toxicity and trafficking defects in a manner strictly determined by genetic background. Abnormal endosome morphology, increased levels of the endosome marker Rab5 and co-localization of mammalian CKI with αSyn aggregates are observed in brain sections from αSyn-overexpressing mice and human synucleinopathies. Our results contribute to evidence that suggests αSyn-induced defects in endocytosis, exocytosis and/or recycling of vesicles involved in these cellular processes might contribute to the pathogenesis of synucleinopathies.


Assuntos
Leveduras/metabolismo , alfa-Sinucleína/genética , Animais , Caseína Quinase I/genética , Caseína Quinase I/metabolismo , Modelos Animais de Doenças , Retículo Endoplasmático/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Fosforilação , Transporte Proteico , alfa-Sinucleína/metabolismo
6.
Nat Genet ; 37(5): 526-31, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15806102

RESUMO

Huntington disease is a fatal neurodegenerative disorder caused by expansion of a polyglutamine tract in the protein huntingtin (Htt), which leads to its aggregation in nuclear and cytoplasmic inclusion bodies. We recently identified 52 loss-of-function mutations in yeast genes that enhance the toxicity of a mutant Htt fragment. Here we report the results from a genome-wide loss-of-function suppressor screen in which we identified 28 gene deletions that suppress toxicity of a mutant Htt fragment. The suppressors are known or predicted to have roles in vesicle transport, vacuolar degradation, transcription and prion-like aggregation. Among the most potent suppressors was Bna4 (kynurenine 3-monooxygenase), an enzyme in the kynurenine pathway of tryptophan degradation that has been linked directly to the pathophysiology of Huntington disease in humans by a mechanism that may involve reactive oxygen species. This finding is suggestive of a conserved mechanism of polyglutamine toxicity from yeast to humans and identifies new candidate therapeutic targets for the treatment of Huntington disease.


Assuntos
Doença de Huntington/metabolismo , Oxigenases de Função Mista/metabolismo , Saccharomyces cerevisiae/genética , Autofagia/fisiologia , Humanos , Doença de Huntington/tratamento farmacológico , Doença de Huntington/genética , Quinurenina 3-Mono-Oxigenase , Microglia/metabolismo , Oxigenases de Função Mista/genética , Mutação , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/metabolismo
7.
J Neurosci ; 32(50): 18259-68, 2012 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-23238740

RESUMO

Peripheral immune cells and brain microglia exhibit an activated phenotype in premanifest Huntington's disease (HD) patients that persists chronically and correlates with clinical measures of neurodegeneration. However, whether activation of the immune system contributes to neurodegeneration in HD, or is a consequence thereof, remains unclear. Signaling through cannabinoid receptor 2 (CB(2)) dampens immune activation. Here, we show that the genetic deletion of CB(2) receptors in a slowly progressing HD mouse model accelerates the onset of motor deficits and increases their severity. Treatment of mice with a CB(2) receptor agonist extends life span and suppresses motor deficits, synapse loss, and CNS inflammation, while a peripherally restricted CB(2) receptor antagonist blocks these effects. CB(2) receptors regulate blood interleukin-6 (IL-6) levels, and IL-6 neutralizing antibodies partially rescue motor deficits and weight loss in HD mice. These findings support a causal link between CB(2) receptor signaling in peripheral immune cells and the onset and severity of neurodegeneration in HD, and they provide a novel therapeutic approach to treat HD.


Assuntos
Doença de Huntington/imunologia , Doença de Huntington/metabolismo , Leucócitos/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Transdução de Sinais/fisiologia , Animais , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Doença de Huntington/patologia , Interleucina-6/imunologia , Interleucina-6/metabolismo , Leucócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Receptor CB2 de Canabinoide/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
J Neurosci ; 32(1): 133-42, 2012 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-22219276

RESUMO

Huntington's disease (HD) is caused by an expanded polyglutamine tract in the protein huntingtin (htt). Although HD has historically been viewed as a brain-specific disease, htt is expressed ubiquitously, and recent studies indicate that mutant htt might cause changes to the immune system that could contribute to pathogenesis. Monocytes from HD patients and mouse models are hyperactive in response to stimulation, and increased levels of inflammatory cytokines and chemokines are found in pre-manifest patients that correlate with pathogenesis. In this study, wild-type (WT) bone marrow cells were transplanted into two lethally irradiated transgenic mouse models of HD that ubiquitously express full-length htt (YAC128 and BACHD mice). Bone marrow transplantation partially attenuated hypokinetic and motor deficits in HD mice. Increased levels of synapses in the cortex were found in HD mice that received bone marrow transplants. Importantly, serum levels of interleukin-6, interleukin-10, CXC chemokine ligand 1, and interferon-γ were significantly higher in HD than WT mice but were normalized in mice that received a bone marrow transplant. These results suggest that immune cell dysfunction might be an important modifier of pathogenesis in HD.


Assuntos
Doenças Autoimunes do Sistema Nervoso/terapia , Células da Medula Óssea/imunologia , Transplante de Medula Óssea/métodos , Doença de Huntington/imunologia , Doença de Huntington/terapia , Terapia de Imunossupressão/métodos , Animais , Doenças Autoimunes do Sistema Nervoso/fisiopatologia , Modelos Animais de Doenças , Feminino , Humanos , Doença de Huntington/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
9.
J Neurosci ; 32(32): 11109-19, 2012 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-22875942

RESUMO

Huntington's disease (HD) is a devastating neurodegenerative disorder with no disease-modifying treatments available. The disease is caused by expansion of a CAG trinucleotide repeat and manifests with progressive motor abnormalities, psychiatric symptoms, and cognitive decline. Expression of an expanded polyglutamine repeat within the Huntingtin (Htt) protein impacts numerous cellular processes, including protein folding and clearance. A hallmark of the disease is the progressive formation of inclusions that represent the culmination of a complex aggregation process. Methylene blue (MB), has been shown to modulate aggregation of amyloidogenic disease proteins. We investigated whether MB could impact mutant Htt-mediated aggregation and neurotoxicity. MB inhibited recombinant protein aggregation in vitro, even when added to preformed oligomers and fibrils. MB also decreased oligomer number and size and decreased accumulation of insoluble mutant Htt in cells. In functional assays, MB increased survival of primary cortical neurons transduced with mutant Htt, reduced neurodegeneration and aggregation in a Drosophila melanogaster model of HD, and reduced disease phenotypes in R6/2 HD modeled mice. Furthermore, MB treatment also promoted an increase in levels of BDNF RNA and protein in vivo. Thus, MB, which is well tolerated and used in humans, has therapeutic potential for HD.


Assuntos
Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Doença de Huntington/tratamento farmacológico , Azul de Metileno/farmacologia , Azul de Metileno/uso terapêutico , Proteínas do Tecido Nervoso/metabolismo , Análise de Variância , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Modelos Animais de Doenças , Drosophila , Embrião de Mamíferos , Antagonistas de Aminoácidos Excitatórios/toxicidade , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Ácido Cinurênico/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Força Atômica , Mutação/genética , Proteínas do Tecido Nervoso/genética , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/prevenção & controle , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Desempenho Psicomotor , Ratos , Teste de Desempenho do Rota-Rod , Transfecção , Expansão das Repetições de Trinucleotídeos/genética
10.
J Biol Chem ; 287(19): 16017-28, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22433867

RESUMO

Huntington disease is a genetic neurodegenerative disorder that arises from an expanded polyglutamine region in the N terminus of the HD gene product, huntingtin. Protein inclusions comprised of N-terminal fragments of mutant huntingtin are a characteristic feature of disease, though are likely to play a protective role rather than a causative one in neurodegeneration. Soluble oligomeric assemblies of huntingtin formed early in the aggregation process are candidate toxic species in HD. In the present study, we established an in vitro system to generate recombinant huntingtin in mammalian cells. Using both denaturing and native gel analysis, we have identified novel oligomeric forms of mammalian-derived expanded huntingtin exon-1 N-terminal fragment. These species are transient and were not previously detected using bacterially expressed exon-1 protein. Importantly, these species are recognized by 3B5H10, an antibody that recognizes a two-stranded hairpin conformation of expanded polyglutamine believed to be associated with a toxic form of huntingtin. Interestingly, comparable oligomeric species were not observed for expanded huntingtin shortstop, a 117-amino acid fragment of huntingtin shown previously in mammalian cell lines and transgenic mice, and here in primary cortical neurons, to be non-toxic. Further, we demonstrate that expanded huntingtin shortstop has a reduced ability to form amyloid-like fibrils characteristic of the aggregation pathway for toxic expanded polyglutamine proteins. Taken together, these data provide a possible candidate toxic species in HD. In addition, these studies demonstrate the fundamental differences in early aggregation events between mutant huntingtin exon-1 and shortstop proteins that may underlie the differences in toxicity.


Assuntos
Éxons/genética , Proteínas do Tecido Nervoso/genética , Expansão das Repetições de Trinucleotídeos/genética , Animais , Western Blotting , Células Cultivadas , Eletroforese em Gel de Poliacrilamida , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Células HEK293 , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/metabolismo , Camundongos , Microscopia de Força Atômica , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Mutação , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Peptídeos/genética , Conformação Proteica , Multimerização Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo
11.
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
12.
J Biol Chem ; 286(1): 410-9, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21044956

RESUMO

Huntington disease (HD) is a neurodegenerative disorder caused by the expansion of a polyglutamine tract in the huntingtin (htt) protein. To uncover candidate therapeutic targets and networks involved in pathogenesis, we integrated gene expression profiling and functional genetic screening to identify genes critical for mutant htt toxicity in yeast. Using mRNA profiling, we have identified genes differentially expressed in wild-type yeast in response to mutant htt toxicity as well as in three toxicity suppressor strains: bna4Δ, mbf1Δ, and ume1Δ. BNA4 encodes the yeast homolog of kynurenine 3-monooxygenase, a promising drug target for HD. Intriguingly, despite playing diverse cellular roles, these three suppressors share common differentially expressed genes involved in stress response, translation elongation, and mitochondrial transport. We then systematically tested the ability of the differentially expressed genes to suppress mutant htt toxicity when overexpressed and have thereby identified 12 novel suppressors, including genes that play a role in stress response, Golgi to endosome transport, and rRNA processing. Integrating the mRNA profiling data and the genetic screening data, we have generated a robust network that shows enrichment in genes involved in rRNA processing and ribosome biogenesis. Strikingly, these observations implicate dysfunction of translation in the pathology of HD. Recent work has shown that regulation of translation is critical for life span extension in Drosophila and that manipulation of this process is protective in Parkinson disease models. In total, these observations suggest that pharmacological manipulation of translation may have therapeutic value in HD.


Assuntos
Proteínas Fúngicas/biossíntese , Proteínas Fúngicas/genética , Perfilação da Expressão Gênica , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/toxicidade , Proteínas Nucleares/metabolismo , Proteínas Nucleares/toxicidade , Biossíntese de Proteínas , Saccharomyces cerevisiae/genética , Sequência de Bases , Deleção de Genes , Genômica , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Proteínas Mutantes/biossíntese , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Mutantes/toxicidade , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Análise de Sequência com Séries de Oligonucleotídeos , Fases de Leitura Aberta/genética , RNA Ribossômico/genética , Sequências Reguladoras de Ácido Nucleico/genética , Ribossomos/metabolismo , Saccharomyces cerevisiae/citologia , Transgenes/genética
13.
Hum Mol Genet ; 19(1): 65-78, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19825844

RESUMO

Huntington's disease (HD) is a late-onset neurodegenerative disorder that is characterized neuropathologically by the presence of neuropil aggregates and nuclear inclusions. However, the profile of aggregate structures that are present in the brains of HD patients or of HD mouse models and the relative contribution of specific aggregate structures to disease pathogenesis is unknown. We have used the Seprion ligand to develop a highly sensitive enzyme-linked immunosorbent assay (ELISA)-based method for quantifying aggregated polyglutamine in tissues from HD mouse models. We used a combination of electron microscopy, atomic force microscopy (AFM) and sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) to investigate the aggregate structures isolated by the ligand. We found that the oligomeric, proto-fibrillar and fibrillar aggregates extracted from the brains of R6/2 and HdhQ150 knock-in mice were remarkably similar. Using AFM, we determined that the nanometre globular oligomers isolated from the brains of both mouse models have dimensions identical to those generated from recombinant huntingtin exon 1 proteins. Finally, antibodies that detect exon 1 Htt epitopes differentially recognize the ligand-captured material on SDS-PAGE gels. The Seprion-ligand ELISA provides an assay with good statistical power for use in preclinical pharmacodynamic therapeutic trials or to assess the effects of the genetic manipulation of potential therapeutic targets on aggregate load. This, together with the ability to identify a spectrum of aggregate species in HD mouse tissues, will contribute to our understanding of how these structures relate to the pathogenesis of HD and whether their formation can be manipulated for therapeutic benefit.


Assuntos
Encéfalo/patologia , Técnicas de Introdução de Genes , Doença de Huntington/patologia , Filamentos do Neurópilo/patologia , Animais , Bioensaio , Encéfalo/ultraestrutura , Modelos Animais de Doenças , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Éxons/genética , Ligantes , Camundongos , Microscopia de Força Atômica , Microscopia Imunoeletrônica , Filamentos do Neurópilo/ultraestrutura , Peptídeos/metabolismo , Fenótipo , Estrutura Quaternária de Proteína , Proteínas da Membrana Plasmática de Transporte de Serotonina/ultraestrutura
14.
J Biol Chem ; 285(19): 14777-90, 2010 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-20220138

RESUMO

Huntington disease (HD) is caused by an expansion of more than 35-40 polyglutamine (polyQ) repeats in the huntingtin (htt) protein, resulting in accumulation of inclusion bodies containing fibrillar deposits of mutant htt fragments. Intriguingly, polyQ length is directly proportional to the propensity for htt to form fibrils and the severity of HD and is inversely correlated with age of onset. Although the structural basis for htt toxicity is unclear, the formation, abundance, and/or persistence of toxic conformers mediating neuronal dysfunction and degeneration in HD must also depend on polyQ length. Here we used atomic force microscopy to demonstrate mutant htt fragments and synthetic polyQ peptides form oligomers in a polyQ length-dependent manner. By time-lapse atomic force microscopy, oligomers form before fibrils, are transient in nature, and are occasionally direct precursors to fibrils. However, the vast majority of fibrils appear to form by monomer addition coinciding with the disappearance of oligomers. Thus, oligomers must undergo a major structural transition preceding fibril formation. In an immortalized striatal cell line and in brain homogenates from a mouse model of HD, a mutant htt fragment formed oligomers in a polyQ length-dependent manner that were similar in size to those formed in vitro, although these structures accumulated over time in vivo. Finally, using immunoelectron microscopy, we detected oligomeric-like structures in human HD brains. These results demonstrate that oligomer formation by a mutant htt fragment is strongly polyQ length-dependent in vitro and in vivo, consistent with a causative role for these structures, or subsets of these structures, in HD pathogenesis.


Assuntos
Proteínas Mutantes/química , Mutação/genética , Proteínas do Tecido Nervoso/química , Proteínas Nucleares/química , Peptídeos/metabolismo , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Corpo Estriado/citologia , Corpo Estriado/metabolismo , Humanos , Proteína Huntingtina , Técnicas In Vitro , Camundongos , Microscopia de Força Atômica , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Peptídeos/química , Expansão das Repetições de Trinucleotídeos
15.
J Biol Chem ; 285(49): 38183-93, 2010 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-20864533

RESUMO

Inclusion bodies of aggregated mutant huntingtin (htt) fragments are a neuropathological hallmark of Huntington disease (HD). The molecular chaperones Hsp70 and Hsp40 colocalize to inclusion bodies and are neuroprotective in HD animal models. How these chaperones suppress mutant htt toxicity is unclear but might involve direct effects on mutant htt misfolding and aggregation. Using size exclusion chromatography and atomic force microscopy, we found that mutant htt fragments assemble into soluble oligomeric species with a broad size distribution, some of which reacted with the conformation-specific antibody A11. Hsp70 associated with A11-reactive oligomers in an Hsp40- and ATP-dependent manner and inhibited their formation coincident with suppression of caspase 3 activity in PC12 cells. Thus, Hsp70 and Hsp40 (DNAJB1) dynamically target specific subsets of soluble oligomers in a classic ATP-dependent reaction cycle, supporting a pathogenic role for these structures in HD.


Assuntos
Trifosfato de Adenosina/metabolismo , Proteínas de Choque Térmico HSP40/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Corpos de Inclusão/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Multimerização Proteica , Trifosfato de Adenosina/genética , Animais , Bovinos , Proteínas de Choque Térmico HSP40/genética , Proteínas de Choque Térmico HSP70/genética , Humanos , Proteína Huntingtina , Corpos de Inclusão/genética , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Células PC12 , Ratos , Solubilidade
16.
J Neurosci ; 29(28): 9104-14, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19605647

RESUMO

Endogenous protein quality control machinery has long been suspected of influencing the onset and progression of neurodegenerative diseases characterized by accumulation of misfolded proteins. Huntington's disease (HD) is a fatal neurodegenerative disorder caused by an expansion of a polyglutamine (polyQ) tract in the protein huntingtin (htt), which leads to its aggregation and accumulation in inclusion bodies. Here, we demonstrate in a mouse model of HD that deletion of the molecular chaperones Hsp70.1 and Hsp70.3 significantly exacerbated numerous physical, behavioral and neuropathological outcome measures, including survival, body weight, tremor, limb clasping and open field activities. Deletion of Hsp70.1 and Hsp70.3 significantly increased the size of inclusion bodies formed by mutant htt exon 1, but surprisingly did not affect the levels of fibrillar aggregates. Moreover, the lack of Hsp70s significantly decreased levels of the calcium regulated protein c-Fos, a marker for neuronal activity. In contrast, deletion of Hsp70s did not accelerate disease in a mouse model of infectious prion-mediated neurodegeneration, ruling out the possibility that the Hsp70.1/70.3 mice are nonspecifically sensitized to all protein misfolding disorders. Thus, endogenous Hsp70s are a critical component of the cellular defense against the toxic effects of misfolded htt protein in neurons, but buffer toxicity by mechanisms independent of the deposition of fibrillar aggregates.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Choque Térmico HSP72/deficiência , Doença de Huntington/genética , Doença de Huntington/patologia , Proteínas do Tecido Nervoso/genética , Fatores Etários , Análise de Variância , Animais , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/genética , Proteínas de Choque Térmico HSP70/deficiência , Proteínas de Choque Térmico HSP72/classificação , Doença de Huntington/complicações , Doença de Huntington/mortalidade , Corpos de Inclusão/patologia , Estimativa de Kaplan-Meier , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/genética , Transtornos dos Movimentos/etiologia , Transtornos dos Movimentos/genética , Proteínas do Tecido Nervoso/metabolismo , Exame Neurológico/métodos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Expansão das Repetições de Trinucleotídeos/genética , Redução de Peso/genética
17.
J Neurochem ; 113(6): 1416-25, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20236387

RESUMO

Elevated concentrations of neurotoxic metabolites of the kynurenine pathway (KP) of tryptophan degradation may play a causative role in Huntington's disease (HD). The brain levels of one of these compounds, 3-hydroxykynurenine (3-HK), are increased in both HD and several mouse models of the disease. In the present study, we examined this impairment in greater detail using the R6/2 mouse, a well-established animal model of HD. Initially, mutant and age-matched wild-type mice received an intrastriatal injection of (3)H-tryptophan to assess the acute, local de novo production of kynurenine, the immediate bioprecursor of 3-HK, in vivo. No effect of genotype was observed between 4 and 12 weeks of age. In contrast, intrastriatally applied (3)H-kynurenine resulted in significantly increased neosynthesis of (3)H-3-HK, but not other tritiated KP metabolites, in the R6/2 striatum. Subsequent ex vivo studies in striatal, cortical and cerebellar tissue revealed substantial increases in the activity of the biosynthetic enzyme of 3-HK, kynurenine 3-monooxygenase and significant reductions in the activity of its degradative enzyme, kynureninase, in HD mice starting at 4 weeks of age. Decreased kynureninase activity was most evident in the cortex and preceded the increase in kynurenine 3-monooxygenase activity. The activity of other KP enzymes showed no consistent brain abnormalities in the mutant mice. These findings suggest that impairments in its immediate metabolic enzymes jointly account for the abnormally high brain levels of 3-HK in the R6/2 model of HD.


Assuntos
Doença de Huntington/metabolismo , Cinurenina/metabolismo , Doenças Metabólicas/metabolismo , Redes e Vias Metabólicas/fisiologia , Fatores Etários , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Cromatografia Líquida de Alta Pressão/métodos , Modelos Animais de Doenças , Feminino , Proteína Huntingtina , Doença de Huntington/complicações , Doença de Huntington/genética , Doença de Huntington/patologia , Cinurenina/análogos & derivados , Doenças Metabólicas/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Fatores de Tempo , Trítio/administração & dosagem , Trítio/metabolismo , Triptofano/administração & dosagem , Triptofano/metabolismo
18.
J Neurosci ; 28(2): 465-72, 2008 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-18184789

RESUMO

Alpha-synuclein aggregates are a common feature of sporadic Parkinson's disease (PD), and mutations that increase alpha-synuclein abundance confer rare heritable forms of PD. Although these findings suggest that alpha-synuclein plays a central role in the pathogenesis of this disorder, little is known of the mechanism by which alpha-synuclein promotes neuron loss or the factors that regulate alpha-synuclein toxicity. To address these matters, we tested candidate modifiers of alpha-synuclein toxicity using a Drosophila model of PD. In the current work, we focused on phase II detoxification enzymes involved in glutathione metabolism. We find that the neuronal death accompanying alpha-synuclein expression in Drosophila is enhanced by loss-of-function mutations in genes that promote glutathione synthesis and glutathione conjugation. This neuronal loss can be overcome by genetic or pharmacological interventions that increase glutathione synthesis or glutathione conjugation activity. Moreover, these same pharmacological agents suppress neuron loss in Drosophila parkin mutants, a loss-of-function model of PD. Our results suggest that oxidative stress is a feature of alpha-synuclein toxicity and that induction of the phase II detoxification pathway represents a potential preventative therapy for PD.


Assuntos
Desintoxicação Metabólica Fase II/fisiologia , Redes e Vias Metabólicas/fisiologia , Neurônios/fisiologia , Doença de Parkinson/patologia , Doença de Parkinson/fisiopatologia , Fatores Etários , Compostos Alílicos , Animais , Animais Geneticamente Modificados , Morte Celular/genética , Modelos Animais de Doenças , Dissulfetos/farmacologia , Relação Dose-Resposta a Droga , Drosophila , Proteínas de Drosophila/genética , Glutationa/metabolismo , Isotiocianatos , Mutação , Degeneração Neural/fisiopatologia , Proteínas do Tecido Nervoso/genética , Neurônios/patologia , Doença de Parkinson/genética , Sulfóxidos , Tiocianatos/farmacologia , Tirosina 3-Mono-Oxigenase/metabolismo , alfa-Sinucleína/genética
19.
Nat Struct Mol Biol ; 11(12): 1215-22, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15543156

RESUMO

Protein conformational changes that result in misfolding, aggregation and amyloid fibril formation are a common feature of many neurodegenerative disorders. Studies with beta-amyloid (Abeta), alpha-synuclein and other amyloid-forming proteins indicate that the assembly of misfolded protein conformers into fibrils is a complex process that may involve the population of metastable spherical and/or annular oligomeric assemblies. Here, we show by atomic force microscopy that a mutant huntingtin fragment with an expanded polyglutamine repeat forms spherical and annular oligomeric structures reminiscent of those formed by Abeta and alpha-synuclein. Notably, the molecular chaperones Hsp70 and Hsp40, which are protective in animal models of neurodegeneration, modulate polyglutamine aggregation reactions by partitioning monomeric conformations and disfavoring the accretion of spherical and annular oligomers.


Assuntos
Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Epitopos/imunologia , Proteínas de Choque Térmico HSP40 , Microscopia de Força Atômica , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Dodecilsulfato de Sódio/farmacologia , Solubilidade
20.
Methods Mol Biol ; 548: 161-74, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19521824

RESUMO

Huntington's disease (HD) is a devastating neurodegenerative disorder that is inherited in an autosomal dominant fashion and is caused by a polyglutamine expansion in the protein huntingtin (htt). In recent years, modeling of various aspects of HD in the yeast Saccharomyces cerevisiae has provided insight into the conserved mechanisms of mutant htt toxicity in eukaryotic cells. The high degree of conservation of cellular and molecular processes between yeast and mammalian cells have made it a valuable system for studying basic mechanisms underlying human disease. Yeast models of HD recapitulate conserved disease-relevant phenotypes and can be used for drug discovery efforts as well as to gain mechanistic and genetic insights into candidate drugs. Here we provide a detailed overview of yeast models of mutant htt misfolding and toxicity and the molecular and phenotypic characterization of these models. We also review how these models identified novel therapeutic targets and compounds for HD and discuss the benefits and limitations of this model genetic system. Finally, we discuss how yeast may be used to provide further insight into the molecular and cellular mechanisms underlying HD and treatment strategies for this devastating disorder.


Assuntos
Doença de Huntington/genética , Modelos Genéticos , Saccharomyces cerevisiae/genética , Avaliação Pré-Clínica de Medicamentos/métodos , Genômica/métodos , Humanos , Proteína Huntingtina , Doença de Huntington/etiologia , Doença de Huntington/terapia , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/toxicidade , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/toxicidade , Fenótipo , Dobramento de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/toxicidade , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/metabolismo , Expansão das Repetições de Trinucleotídeos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA