Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Ther Nucleic Acids ; 11: 79-90, 2018 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-29858092

RESUMO

Huntington's disease (HD) is an intractable neurodegenerative disorder caused by mutant Huntingtin (HTT) proteins that adversely affect various biomolecules and genes. MicroRNAs (miRNAs), which are functional small non-coding RNAs, are also affected by mutant HTT proteins. Here, we show amelioration in motor function and lifespan of HD-model mice, R6/2 mice, by supplying miR-132 to HD brains using a recombinant adeno-associated virus (rAAV) miRNA expression system. miR-132 is an miRNA related to neuronal maturation and function, but the level of miR-132 in the brain of R6/2 mice was significantly lower than that of wild-type mice. Our miR-132 supplemental treatment, i.e., supplying miR-132 to the brain, produced symptomatic improvement or retarded disease progression in R6/2 mice; interestingly, it had little effect on disease-causing mutant HTT mRNA expression and its products. Therefore, the findings suggest that there may be a therapeutic way to treat HD without inhibiting and/or repairing disease-causing HTT genes and gene products. Although miR-132 supplement may not be a definitive treatment for HD, it may become a therapeutic method for relieving HD symptoms and delaying HD progression.

2.
eNeuro ; 4(2)2017.
Artigo em Inglês | MEDLINE | ID: mdl-28374014

RESUMO

Age-associated neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and the polyglutamine (polyQ) diseases, are becoming prevalent as a consequence of elongation of the human lifespan. Although various rodent models have been developed to study and overcome these diseases, they have limitations in their translational research utility owing to differences from humans in brain structure and function and in drug metabolism. Here, we generated a transgenic marmoset model of the polyQ diseases, showing progressive neurological symptoms including motor impairment. Seven transgenic marmosets were produced by lentiviral introduction of the human ataxin 3 gene with 120 CAG repeats encoding an expanded polyQ stretch. Although all offspring showed no neurological symptoms at birth, three marmosets with higher transgene expression developed neurological symptoms of varying degrees at 3-4 months after birth, followed by gradual decreases in body weight gain, spontaneous activity, and grip strength, indicating time-dependent disease progression. Pathological examinations revealed neurodegeneration and intranuclear polyQ protein inclusions accompanied by gliosis, which recapitulate the neuropathological features of polyQ disease patients. Consistent with neuronal loss in the cerebellum, brain MRI analyses in one living symptomatic marmoset detected enlargement of the fourth ventricle, which suggests cerebellar atrophy. Notably, successful germline transgene transmission was confirmed in the second-generation offspring derived from the symptomatic transgenic marmoset gamete. Because the accumulation of abnormal proteins is a shared pathomechanism among various neurodegenerative diseases, we suggest that this new marmoset model will contribute toward elucidating the pathomechanisms of and developing clinically applicable therapies for neurodegenerative diseases.


Assuntos
Animais Geneticamente Modificados , Callithrix , Modelos Animais de Doenças , Doenças Neurodegenerativas , Peptídeos , Envelhecimento/patologia , Envelhecimento/fisiologia , Animais , Ataxina-3/genética , Ataxina-3/metabolismo , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular , Progressão da Doença , Orelha , Fibroblastos/metabolismo , Fibroblastos/patologia , Vetores Genéticos , Humanos , Lentivirus/genética , Masculino , Atividade Motora/fisiologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Doenças Neurodegenerativas/diagnóstico por imagem , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Peptídeos/metabolismo , Fenótipo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Expansão das Repetições de Trinucleotídeos
3.
Proc Natl Acad Sci U S A ; 112(19): E2497-506, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25918398

RESUMO

The heat shock response (HSR), a transcriptional response that up-regulates molecular chaperones upon heat shock, is necessary for cell survival in a stressful environment to maintain protein homeostasis (proteostasis). However, there is accumulating evidence that the HSR does not ubiquitously occur under stress conditions, but largely depends on the cell types. Despite such imbalanced HSR among different cells and tissues, molecular mechanisms by which multicellular organisms maintain their global proteostasis have remained poorly understood. Here, we report that proteostasis can be maintained by molecular chaperones not only in a cell-autonomous manner but also in a non-cell-autonomous manner. We found that elevated expression of molecular chaperones, such as Hsp40 and Hsp70, in a group of cells improves proteostasis in other groups of cells, both in cultured cells and in Drosophila expressing aggregation-prone polyglutamine proteins. We also found that Hsp40, as well as Hsp70 and Hsp90, is physiologically secreted from cells via exosomes, and that the J domain at the N terminus is responsible for its exosome-mediated secretion. Addition of Hsp40/Hsp70-containing exosomes to the culture medium of the polyglutamine-expressing cells results in efficient suppression of inclusion body formation, indicating that molecular chaperones non-cell autonomously improve the protein-folding environment via exosome-mediated transmission. Our study reveals that intercellular chaperone transmission mediated by exosomes is a novel molecular mechanism for non-cell-autonomous maintenance of organismal proteostasis that could functionally compensate for the imbalanced state of the HSR among different cells, and also provides a novel physiological role of exosomes that contributes to maintenance of organismal proteostasis.


Assuntos
Exossomos/metabolismo , Chaperonas Moleculares/metabolismo , Animais , Animais Geneticamente Modificados , Linhagem Celular , Drosophila , Drosophila melanogaster , Proteínas de Choque Térmico HSP40/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Proteínas de Choque Térmico/metabolismo , Homeostase , Camundongos , Microscopia Eletrônica , Doenças Neurodegenerativas/patologia , Peptídeos/química , Dobramento de Proteína , Estrutura Terciária de Proteína , Transdução de Sinais , Transcrição Gênica , Transfecção
4.
J Biol Chem ; 290(3): 1442-53, 2015 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-25480790

RESUMO

Oligomer formation and accumulation of pathogenic proteins are key events in the pathomechanisms of many neurodegenerative diseases, such as Alzheimer disease, ALS, and the polyglutamine (polyQ) diseases. The autophagy-lysosome degradation system may have therapeutic potential against these diseases because it can degrade even large oligomers. Although p62/sequestosome 1 plays a physiological role in selective autophagy of ubiquitinated proteins, whether p62 recognizes and degrades pathogenic proteins in neurodegenerative diseases has remained unclear. In this study, to elucidate the role of p62 in such pathogenic conditions in vivo, we used Drosophila models of neurodegenerative diseases. We found that p62 predominantly co-localizes with cytoplasmic polyQ protein aggregates in the MJDtr-Q78 polyQ disease model flies. Loss of p62 function resulted in significant exacerbation of eye degeneration in these flies. Immunohistochemical analyses revealed enhanced accumulation of cytoplasmic aggregates by p62 knockdown in the MJDtr-Q78 flies, similarly to knockdown of autophagy-related genes (Atgs). Knockdown of both p62 and Atgs did not show any additive effects in the MJDtr-Q78 flies, implying that p62 function is mediated by autophagy. Biochemical analyses showed that loss of p62 function delays the degradation of the MJDtr-Q78 protein, especially its oligomeric species. We also found that loss of p62 function exacerbates eye degeneration in another polyQ disease fly model as well as in ALS model flies. We therefore conclude that p62 plays a protective role against polyQ-induced neurodegeneration, by the autophagic degradation of polyQ protein oligomers in vivo, indicating its therapeutic potential for the polyQ diseases and possibly for other neurodegenerative diseases.


Assuntos
Autofagia , Proteínas de Drosophila/metabolismo , Doenças Neurodegenerativas/metabolismo , Peptídeos/química , Células Fotorreceptoras de Invertebrados/metabolismo , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Fator de Transcrição TFIID/metabolismo , Animais , Citoplasma/metabolismo , Drosophila , Imuno-Histoquímica , Microscopia Eletrônica de Varredura , Fosforilação , Células Fotorreceptoras de Invertebrados/ultraestrutura , Desnaturação Proteica , Dobramento de Proteína , Transgenes , Proteínas Ubiquitinadas/química
5.
Curr Med Chem ; 21(23): 2575-82, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24533807

RESUMO

The polyglutamine (polyQ) diseases including Huntington's disease and spinocerebellar ataxias are a group of inherited neurodegenerative diseases that are caused by an abnormal expansion of the polyQ stretch in disease-causative proteins. The expanded polyQ stretches are intrinsically unstable and are prone to form insoluble aggregates and inclusion bodies. Recent studies have revealed that the expanded polyQ proteins gain cytotoxicity during the aggregation process, which may possibly cause detrimental effects on a wide range of essential cellular functions leading to eventual neuronal degeneration. Based on the pathogenic mechanism of the polyQ diseases, several therapeutic approaches have been proposed to date. Among them, here we focus on peptide-based approaches that target either aggregate formation of the polyQ proteins or abnormal cellular processes induced by the expanded polyQ proteins. Although both approaches are effective in suppressing cytotoxicity of the abnormal polyQ proteins and the disease phenotypes of animal models, the former approach is more attractive since it targets the most upstream change occurring in the polyQ diseases, and is therefore expected to be effective against various downstream functional abnormalities in a broad range of polyQ diseases. One of the major current problems that must be overcome for development of peptide-based therapies of the polyQ diseases is the issue of brain delivery, which is also discussed in this article. We hope that in the near future effective therapies are developed, and bring hope to many patients suffering from the currently untreatable polyQ diseases.


Assuntos
Doenças Neurodegenerativas/tratamento farmacológico , Peptídeos/uso terapêutico , Animais , Humanos , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Peptídeos/química , Peptídeos/metabolismo , Fenótipo , Agregação Patológica de Proteínas/tratamento farmacológico , Agregação Patológica de Proteínas/metabolismo
6.
Neurotherapeutics ; 10(3): 440-6, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23504628

RESUMO

Protein misfolding and aggregation in the brain have been recognized to be crucial in the pathogenesis of various neurodegenerative diseases, including Alzheimer's, Parkinson's, and the polyglutamine (polyQ) diseases, which are collectively called the "protein misfolding diseases". In the polyQ diseases, an abnormally expanded polyQ stretch in the responsible proteins causes the proteins to misfold and aggregate, eventually resulting in neurodegeneration. Hypothesizing that polyQ protein misfolding and aggregation could be inhibited by molecules specifically binding to the expanded polyQ stretch, we identified polyQ binding peptide 1 (QBP1). We show that QBP1 does, indeed, inhibit misfolding and aggregation of the expanded polyQ protein in vitro. Furthermore overexpression of QBP1 by the crossing of transgenic animals inhibits neurodegeneration in Drosophila models of the polyQ diseases. We also introduce our attempts to deliver QBP1 into the brain by administration using viral vectors and protein transduction domains. Interestingly, recent data suggest that QBP1 can also inhibit the misfolding/aggregation of proteins responsible for other protein misfolding diseases, highlighting the potential of QBP1 as a general therapeutic molecule for a wide range of neurodegenerative diseases. We hope that in the near future, aggregation inhibitor-based drugs will be developed and bring relief to patients suffering from these currently intractable protein misfolding diseases.


Assuntos
Oligopeptídeos/uso terapêutico , Peptídeos/metabolismo , Deficiências na Proteostase/terapia , Animais , Animais Geneticamente Modificados , Humanos , Oligopeptídeos/biossíntese , Dobramento de Proteína/efeitos dos fármacos , Transdução Genética
7.
PLoS One ; 7(11): e51069, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23226463

RESUMO

The polyglutamine (polyQ) diseases such as Huntington's disease (HD), are neurodegenerative diseases caused by proteins with an expanded polyQ stretch, which misfold and aggregate, and eventually accumulate as inclusion bodies within neurons. Molecules that inhibit polyQ protein misfolding/aggregation, such as Polyglutamine Binding Peptide 1 (QBP1) and molecular chaperones, have been shown to exert therapeutic effects in vivo by crossing of transgenic animals. Towards developing a therapy using these aggregation inhibitors, we here investigated the effect of viral vector-mediated gene therapy using QBP1 and molecular chaperones on polyQ disease model mice. We found that injection of adeno-associated virus type 5 (AAV5) expressing QBP1 or Hsp40 into the striatum both dramatically suppresses inclusion body formation in the HD mouse R6/2. AAV5-Hsp40 injection also ameliorated the motor impairment and extended the lifespan of R6/2 mice. Unexpectedly, we found even in virus non-infected cells that AAV5-Hsp40 appreciably suppresses inclusion body formation, suggesting a non-cell autonomous therapeutic effect. We further show that Hsp40 inhibits secretion of the polyQ protein from cultured cells, implying that it inhibits the recently suggested cell-cell transmission of the polyQ protein. Our results demonstrate for the first time the therapeutic effect of Hsp40 gene therapy on the neurological phenotypes of polyQ disease mice.


Assuntos
Terapia Genética , Proteínas de Choque Térmico HSP40/genética , Proteínas de Choque Térmico HSP40/uso terapêutico , Doença de Huntington/genética , Doença de Huntington/terapia , Peptídeos/metabolismo , Animais , Encéfalo/patologia , Encéfalo/fisiopatologia , Células Cultivadas , Dependovirus/metabolismo , Modelos Animais de Doenças , Força da Mão/fisiologia , Doença de Huntington/patologia , Doença de Huntington/fisiopatologia , Corpos de Inclusão/metabolismo , Longevidade , Camundongos , Atividade Motora/fisiologia , Neurônios/metabolismo , Neurônios/patologia , Neurônios/virologia , Oligopeptídeos/genética , Oligopeptídeos/uso terapêutico , Fenótipo , Redução de Peso
8.
J Amino Acids ; 2011: 265084, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22312459

RESUMO

Misfolding and abnormal aggregation of proteins in the brain are implicated in the pathogenesis of various neurodegenerative diseases including Alzheimer's, Parkinson's, and the polyglutamine (polyQ) diseases. In the polyQ diseases, an abnormally expanded polyQ stretch triggers misfolding and aggregation of the disease-causing proteins, eventually resulting in neurodegeneration. In this paper, we introduce our therapeutic strategy against the polyQ diseases using polyQ binding peptide 1 (QBP1), a peptide that we identified by phage display screening. We showed that QBP1 specifically binds to the expanded polyQ stretch and inhibits its misfolding and aggregation, resulting in suppression of neurodegeneration in cell culture and animal models of the polyQ diseases. We further demonstrated the potential of protein transduction domains (PTDs) for in vivo delivery of QBP1. We hope that in the near future, chemical analogues of aggregation inhibitor peptides including QBP1 will be developed against protein misfolding-associated neurodegenerative diseases.

9.
Curr Pharm Biotechnol ; 11(2): 188-97, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20166962

RESUMO

Protein misfolding and aggregation in the brain have been implicated as a common molecular pathogenesis of various neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and the polyglutamine (polyQ) diseases. The polyQ diseases are a group of nine hereditary neurodegenerative diseases, including Huntington's disease (HD) and various types of spinocerebellar ataxia (SCA), which are caused by abnormal expansions of the polyQ stretch (> 35-40 repeats) in unrelated disease-causative proteins. The expanded polyQ stretch is thought to trigger misfolding of these proteins, leading to their aggregation and accumulation as inclusion bodies in affected neurons, eventually resulting in neurodegeneration. Misfolding and aggregation of the polyQ protein are the most ideal therapeutic targets since they are the most upstream events in the pathogenic cascade, and therefore, therapeutic approaches using molecular chaperones, which prevent protein misfolding and assist the refolding of misfolded proteins, are being extensively investigated. Indeed, a variety of molecular chaperones such as Hsp70 and Hsp40 have been demonstrated to exert therapeutic effects against various experimental models of the polyQ diseases. Furthermore, toward developing pharmacological therapies, small chemical activators of heat shock transcription factor 1 (HSF1) such as geldanamycin and its derivative 17-AAG, which induce multiple endogenous molecular chaperones, have been proven to be effective not only in polyQ disease models, but also in other neurodegenerative disease models. We hope that brain-permeable molecular chaperone inducers will be developed as drugs against a wide range of neurodegenerative diseases in the near future.


Assuntos
Transtornos Heredodegenerativos do Sistema Nervoso/terapia , Chaperonas Moleculares/biossíntese , Peptídeos/metabolismo , Amiloide/genética , Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Transtornos Heredodegenerativos do Sistema Nervoso/genética , Transtornos Heredodegenerativos do Sistema Nervoso/metabolismo , Humanos , Chaperonas Moleculares/genética , Chaperonas Moleculares/fisiologia , Peptídeos/genética , Dobramento de Proteína
10.
Bioorg Med Chem ; 17(3): 1259-63, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19121945

RESUMO

Aggregation and deposition of expanded polyglutamine proteins in the brain cause neurodegenerative diseases including Huntington disease. This pathogenic process is suppressed and delayed in the presence of polyglutamine binding peptide 1 (QBP1), which we previously identified as an undecapeptide binding to pathogenic polyglutamine proteins from phage display peptide libraries. In this paper, a structure-activity relationship study on QBP1 was conducted to determine the pharmacophores for inhibition of polyglutamine aggregation. Furthermore, a truncation study identified an octapeptide as the minimum structure for suppressing aggregation of polyglutamine proteins, which is equipotent to the parent undecapeptide QBP1.


Assuntos
Oligopeptídeos/química , Sequência de Aminoácidos , Concentração Inibidora 50 , Oligopeptídeos/farmacologia , Biblioteca de Peptídeos , Relação Estrutura-Atividade
11.
Neurosci Lett ; 449(2): 87-92, 2009 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-18603372

RESUMO

The polyglutamine (polyQ) diseases are neurodegenerative diseases caused by proteins with an abnormally expanded polyQ stretch, which triggers abnormal aggregation of these proteins in the brain. We previously showed that the polyQ-binding peptide QBP1 inhibits polyQ aggregation, and further that administration of QBP1 fused with a protein transduction domain (PTD) suppresses polyQ-induced neurodegeneration in Drosophila. As the next step towards developing a therapy using QBP1, we investigated the delivery of PTD-QBP1 to the mouse brain upon its administration. Here we successfully detected delivery of PTD-QBP1 into mouse brain cells upon its single intracerebroventricular injection. In addition, long-term administration of PTD-QBP1 to polyQ disease mice improved their weight loss phenotype, suggesting a possible therapeutic effect. Our study indicates the potential of PTD-mediated delivery of QBP1 as a therapeutic strategy for the currently untreatable polyQ diseases.


Assuntos
Encéfalo/efeitos dos fármacos , Doenças Neurodegenerativas/tratamento farmacológico , Neurônios/efeitos dos fármacos , Oligopeptídeos/farmacologia , Peptídeos/antagonistas & inibidores , Animais , Peso Corporal/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Humanos , Injeções Intraventriculares , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/fisiopatologia , Neurônios/metabolismo , Neurônios/patologia , Oligopeptídeos/uso terapêutico , Peptídeos/metabolismo , Fenótipo , Estrutura Terciária de Proteína/fisiologia , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Resultado do Tratamento
12.
Curr Pharm Des ; 14(30): 3267-79, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19075705

RESUMO

The polyglutamine (polyQ) diseases, including Huntington's disease and spinocerebellar ataxias, are classified as the protein misfolding neurodegenerative diseases like Alzheimer's and Parkinson's diseases, and they are caused by an abnormal expansion of the polyQ stretch in disease-causative proteins. Expanded polyQ stretches have been shown to undergo a conformational transition to a beta-sheet-dominant structure, leading to assembly of the host proteins into insoluble beta-sheet-rich amyloid fibrillar aggregates and their subsequent accumulation as inclusion bodies in affected neurons, eventually resulting in neurodegeneration. Based on cytotoxicity of the soluble beta-sheet monomer of the expanded polyQ protein, we propose the "Exposed beta-sheet hypothesis", in which both the toxic beta-sheet conformational transition and misassembly into amyloid fibrils of the disease-causative proteins contribute to the pathogenesis of the polyQ diseases, and possibly the other protein misfolding neurodegenerative diseases. Among the various therapeutic targets, the toxic conformational changes and aggregation of the expanded polyQ proteins are most ideal since they are the earliest events in the pathogenic cascade, and therapeutic approaches using molecular chaperones, intrabodies, peptides, and small chemical compounds have been developed to date. Furthermore, high-throughput screening approaches to identify polyQ aggregate inhibitors are in progress. We hope that protein aggregate inhibitors which are widely effective not only for the polyQ diseases, but also for many neurodegenerative diseases will be discovered in the near future.


Assuntos
Doenças Neurodegenerativas/tratamento farmacológico , Peptídeos , Animais , Humanos , Modelos Moleculares , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/uso terapêutico , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Peptídeos/uso terapêutico , Dobramento de Proteína , Estrutura Secundária de Proteína , Bibliotecas de Moléculas Pequenas/uso terapêutico
13.
J Biol Chem ; 283(38): 26188-97, 2008 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-18632670

RESUMO

Many neurodegenerative diseases including Alzheimer, Parkinson, and polyglutamine (polyQ) diseases are thought to be caused by protein misfolding. The polyQ diseases, including Huntington disease and spinocerebellar ataxias (SCAs), are caused by abnormal expansions of the polyQ stretch in disease-causing proteins, which trigger misfolding of these proteins, resulting in their deposition as inclusion bodies in affected neurons. Although genetic expression of molecular chaperones has been shown to suppress polyQ protein misfolding and neurodegeneration, toward developing a therapy, it is ideal to induce endogenous molecular chaperones by chemical administration. In this study, we assessed the therapeutic effects of heat shock transcription factor 1 (HSF1)-activating compounds, which induce multiple molecular chaperones, on polyQ-induced neurodegeneration in vivo. We found that oral administration of 17-(allylamino)-17-demethoxygeldanamycin (17-AAG) markedly suppresses compound eye degeneration and inclusion body formation in a Drosophila model of SCA. 17-AAG also dramatically rescued the lethality of the SCA model (74.1% rescue) and suppressed neurodegeneration in a Huntington disease model (46.3% rescue), indicating that 17-AAG is widely effective against various polyQ diseases. 17-AAG induced Hsp70, Hsp40, and Hsp90 expression in a dose-dependent manner, and the expression levels correlated with its therapeutic effects. Furthermore, knockdown of HSF1 abolished the induction of molecular chaperones and the therapeutic effect of 17-AAG, indicating that its therapeutic effects depend on HSF1 activation. Our study indicates that induction of multiple molecular chaperones by 17-AAG treatment is a promising therapeutic approach for a wide range of polyQ diseases and possibly other neurodegenerative diseases.


Assuntos
Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Doenças Neurodegenerativas/metabolismo , Peptídeos/química , Células Fotorreceptoras de Invertebrados/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Animais , Animais Geneticamente Modificados , Benzoquinonas/farmacologia , Relação Dose-Resposta a Droga , Drosophila , Proteínas de Choque Térmico HSP40/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Fatores de Transcrição de Choque Térmico , Lactamas Macrocíclicas/farmacologia , Modelos Biológicos , Células Fotorreceptoras de Invertebrados/efeitos dos fármacos , Ligação Proteica
14.
J Biol Chem ; 282(33): 24039-48, 2007 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-17573338

RESUMO

Abnormal aggregation of misfolded proteins and their deposition as inclusion bodies in the brain have been implicated as a common molecular pathogenesis of neurodegenerative diseases including Alzheimer, Parkinson, and the polyglutamine (poly(Q)) diseases, which are collectively called the conformational diseases. The poly(Q) diseases, including Huntington disease and various types of spinocerebellar ataxia, are caused by abnormal expansions of the poly(Q) stretch within disease-causing proteins, which triggers the disease-causing proteins to aggregate into insoluble beta-sheet-rich amyloid fibrils. Although oligomeric structures formed in vitro are believed to be more toxic than mature amyloid fibrils in these diseases, the existence of oligomers in vivo has remained controversial. To explore oligomer formation in cells, we employed fluorescence correlation spectroscopy (FCS), which is a highly sensitive technique for investigating the dynamics of fluorescent molecules in solution. Here we demonstrate direct evidence for oligomer formation of poly(Q)-green fluorescent protein (GFP) fusion proteins expressed in cultured cells, by showing a time-dependent increase in their diffusion time and particle size by FCS. We show that the poly(Q)-binding peptide QBP1 inhibits poly(Q)-GFP oligomer formation, whereas Congo red only inhibits the growth of oligomers, but not the initial formation of the poly(Q)-GFP oligomers, suggesting that FCS is capable of identifying poly(Q) oligomer inhibitors. We therefore conclude that FCS is a useful technique to monitor the oligomerization of disease-causing proteins in cells as well as its inhibition in the conformational diseases.


Assuntos
Amiloide/química , Oligopeptídeos/análise , Peptídeos/análise , Espectrometria de Fluorescência/métodos , Células Cultivadas , Proteínas de Fluorescência Verde , Humanos , Doenças Neurodegenerativas/etiologia , Dobramento de Proteína
15.
Brain Nerve ; 59(4): 393-404, 2007 Apr.
Artigo em Japonês | MEDLINE | ID: mdl-17447526

RESUMO

The polyglutamine diseases are a group of nine inherited neurodegenerative diseases including Huntington disease, spinocerebellar ataxia type 1, 2, 3, 6, 7 and 17, dentatorubral pallidoluysian atrophy, and spinobulbar muscular atrophy, which are caused by an abnormal expansion of the CAG repeat encoding a polyglutamine stretch in each unrelated disease-causing gene. In the pathogenesis of the polyglutamine diseases, expansion of the polyglutamine stretch causes misfolding and conformational alterations of the disease-causing proteins, leading to pathogenic protein-protein interactions including aggregate formation, and subsequently resulting in their deposition as inclusion bodies in affected neurons. Expression of these expanded polyglutamine proteins has been reported to impair protein degradation, transcriptional regulation, axonal transport, mitochondrial function, etc., which eventually result in neurodegeneration. Currently, a wide variety of research towards establishing therapies targeting each step in the pathogenesis of the polyglutamine diseases is in progress, which includes suppressing mutant gene expression by RNAi, inhibiting protein misfolding/aggregation, promoting protein degradation, activating transcription, activating mitochondrial function, inhibiting neuronal cell death, and neuroprotection by neurotrophic factors. Standardized validation of these preclinical studies and development of sensitive biomarkers for evaluation of therapeutic efficacy in clinical trials will be necessary for development of drugs for the polyglutamine diseases.


Assuntos
Doenças Neurodegenerativas/genética , Peptídeos/genética , Humanos , Doenças Neurodegenerativas/tratamento farmacológico , Peptídeos/uso terapêutico
16.
Nat Struct Mol Biol ; 14(4): 332-40, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17369839

RESUMO

Polyglutamine (polyQ) diseases are classified as conformational neurodegenerative diseases, like Alzheimer and Parkinson diseases, and they are caused by proteins with an abnormally expanded polyQ stretch. However, conformational changes of the expanded polyQ protein and the toxic conformers formed during aggregation have remained poorly understood despite their important role in pathogenesis. Here we show that a beta-sheet conformational transition of the expanded polyQ protein monomer precedes its assembly into beta-sheet-rich amyloid-like fibrils. Microinjection of the various polyQ protein conformers into cultured cells revealed that the soluble beta-sheet monomer causes cytotoxicity. The polyQ-binding peptide QBP1 prevents the toxic beta-sheet conformational transition of the expanded polyQ protein monomer. We conclude that the toxic conformational transition, and not simply the aggregation process itself, is a therapeutic target for polyQ diseases and possibly for conformational diseases in general.


Assuntos
Peptídeos/química , Peptídeos/toxicidade , Amiloide/efeitos dos fármacos , Animais , Células COS , Morte Celular/efeitos dos fármacos , Chlorocebus aethiops , Microscopia de Força Atômica , Microscopia Eletrônica de Transmissão , Modelos Biológicos , Oligopeptídeos/metabolismo , Peptídeos/metabolismo , Estrutura Quaternária de Proteína/efeitos dos fármacos , Estrutura Secundária de Proteína/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo , Solubilidade/efeitos dos fármacos
17.
Mol Ther ; 15(2): 303-9, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17235308

RESUMO

Many neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and the polyglutamine (polyQ) diseases share common features including abnormal aggregation of misfolded proteins and their deposition as inclusion bodies in the brain. The polyQ diseases are caused by abnormal expansion of a polyQ stretch in each disease-causing protein, which triggers these proteins to form aggregates. We previously showed that genetic expression of the aggregate inhibitor peptide polyQ binding peptide 1 (QBP1) suppresses polyQ-induced neurodegeneration in Drosophila. However, to establish a molecular therapy using QBP1, QBP1 needs to be delivered into cells by its administration. In this study, we employed protein transduction domains (PTDs) to enable the efficient intracellular delivery of QBP1. We show here that fusion with a PTD enables the efficient intracellular delivery of QBP1, and that PTD-QBP1 treatment suppressed polyQ-induced cytotoxicity in cultured cells. Most importantly, oral administration of PTD-QBP1 successfully suppressed polyQ-induced premature death as well as polyQ inclusion body formation in a Drosophila model of the polyQ diseases, demonstrating its therapeutic effect against polyQ-induced neurodegeneration in vivo. Our study indicates that PTD-mediated delivery of aggregate inhibitor peptides is a promising therapeutic strategy for neurodegenerative diseases with abnormal aggregation of misfolded proteins.


Assuntos
Degeneração Neural/tratamento farmacológico , Oligopeptídeos/farmacologia , Peptídeos/metabolismo , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Células COS , Chlorocebus aethiops , Modelos Animais de Doenças , Drosophila/efeitos dos fármacos , Drosophila/genética , Drosophila/metabolismo , Corpos de Inclusão/efeitos dos fármacos , Corpos de Inclusão/metabolismo , Substâncias Macromoleculares/metabolismo , Dados de Sequência Molecular , Degeneração Neural/genética , Degeneração Neural/metabolismo , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Peptídeos/química , Peptídeos/genética , Ligação Proteica
18.
FEBS Lett ; 579(17): 3842-8, 2005 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-15978579

RESUMO

Heat shock transcription factor 1 (HSF1) mediates the induction of heat shock proteins in response to various types of stress. Although HSF1 activity is regulated by its post-translational modifications, alterations in mRNA expression have also been suggested. We here identified three new alternatively spliced isoforms of Drosophila HSF (dHSF) mRNA, named dHSFb, dHSFc, and dHSFd. We found that the ratio of dHSFb increases upon heat exposure, while that of dHSFd increases upon cold exposure. The dHSFc and dHSFd isoforms showed greater transcriptional activity than the other isoforms. Our findings suggest that alternative splicing regulates the transcriptional activity of dHSF.


Assuntos
Processamento Alternativo/fisiologia , Temperatura Baixa , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Resposta ao Choque Térmico/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Células Cultivadas , Drosophila melanogaster/metabolismo , Regulação da Expressão Gênica , Fatores de Transcrição de Choque Térmico , Humanos , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/análise , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...