Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Brain ; 17(1): 26, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38778381

RESUMO

Aggregation of misfolded α-synuclein (α-syn) is a key characteristic feature of Parkinson's disease (PD) and related synucleinopathies. The nature of these aggregates and their contribution to cellular dysfunction is still not clearly elucidated. We employed mass spectrometry-based total and phospho-proteomics to characterize the underlying molecular and biological changes due to α-syn aggregation using the M83 mouse primary neuronal model of PD. We identified gross changes in the proteome that coincided with the formation of large Lewy body-like α-syn aggregates in these neurons. We used protein-protein interaction (PPI)-based network analysis to identify key protein clusters modulating specific biological pathways that may be dysregulated and identified several mechanisms that regulate protein homeostasis (proteostasis). The observed changes in the proteome may include both homeostatic compensation and dysregulation due to α-syn aggregation and a greater understanding of both processes and their role in α-syn-related proteostasis may lead to improved therapeutic options for patients with PD and related disorders.


Assuntos
Neurônios , Doença de Parkinson , Agregados Proteicos , Proteômica , Proteostase , alfa-Sinucleína , alfa-Sinucleína/metabolismo , Animais , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Neurônios/metabolismo , Neurônios/patologia , Camundongos , Mapas de Interação de Proteínas , Proteoma/metabolismo
2.
SLAS Discov ; 29(2): 100137, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38128829

RESUMO

Aberrant protein aggregation is a pathological cellular hallmark of many neurodegenerative diseases, such as Alzheimer's disease (AD) and frontotemporal dementia (FTD), where the tau protein is aggregating, forming neurofibrillary tangles (NFTs), and propagating from neuron to neuron. These processes have been linked to disease progression and a decline in cognitive function. Various therapeutic approaches aim at the prevention or reduction of tau aggregates in neurons. Human induced pluripotent stem cells (hiPSCs) are a very valuable tool in neuroscience discovery, as they offer access to potentially unlimited amounts of cell types that are affected in disease, including cortical neurons, for in vitro studies. We have generated an in vitro model for tau aggregation that uses hiPSC - derived neurons expressing an aggregation prone, fluorescently tagged version of the human tau protein after lentiviral transduction. Upon addition of tau seeds in the form of recombinant sonicated paired helical filaments (sPHFs), the neurons show robust, disease-like aggregation of the tau protein. The model was developed as a plate-based high content screening assay coupled with an image analysis algorithm to evaluate the impact of small molecules or genetic perturbations on tau. We show that the assay can be used to evaluate small molecules or screen targeted compound libraries. Using siRNA-based gene knockdown, genes of interest can be evaluated, and we could show that a targeted gene library can be screened, by screening nearly 100 deubiquitinating enzymes (DUBs) in that assay. The assay uses an imaging-based readout, a relatively short timeline, quantifies the extent of tau aggregation, and also allows the assessment of cell viability. Furthermore, it can be easily adapted to different hiPSC lines or neuronal subtypes. Taken together, this complex and highly relevant approach can be routinely applied on a weekly basis in the screening funnels of several projects and generates data with a turnaround time of approximately five weeks.


Assuntos
Doença de Alzheimer , Células-Tronco Pluripotentes Induzidas , Humanos , Proteínas tau/genética , Proteínas tau/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Doença de Alzheimer/metabolismo , Neurônios/metabolismo
3.
Eur J Pharmacol ; 934: 175301, 2022 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-36191631

RESUMO

In this study we aimed to reduce tau pathology, a hallmark of Alzheimer's Disease (AD), by activating mTOR-dependent autophagy in a transgenic mouse model of tauopathy by long-term dosing of animals with mTOR-inhibitors. Rapamycin treatment reduced the burden of hyperphosphorylated and aggregated pathological tau in the cerebral cortex only when applied to young mice, prior to the emergence of pathology. Conversely, PQR530 which exhibits better brain exposure and superior pharmacokinetic properties, reduced tau pathology even when the treatment started after the onset of pathology. Our results show that dosing animals twice per week with PQR530 resulted in intermittent, rather than sustained target engagement. Nevertheless, this pulse-like mTOR inhibition followed by longer intervals of re-activation was sufficient to reduce tau pathology in the cerebral cortex in P301S tau transgenic mice. This suggests that balanced therapeutic dosing of blood-brain-barrier permeable mTOR-inhibitors can result in a disease-modifying effect in AD and at the same time prevents toxic side effects due to prolonged over activation of autophagy.


Assuntos
Doença de Alzheimer , Animais , Camundongos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Proteínas tau/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Camundongos Transgênicos , Encéfalo , Sirolimo/farmacologia , Modelos Animais de Doenças
4.
Sci Rep ; 11(1): 2879, 2021 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-33536571

RESUMO

Alzheimer's disease (AD) is a common neurodegenerative disease with poor prognosis. New options for drug discovery targets are needed. We developed an imaging based arrayed CRISPR method to interrogate the human genome for modulation of in vitro correlates of AD features, and used this to assess 1525 human genes related to tau aggregation, autophagy and mitochondria. This work revealed (I) a network of tau aggregation modulators including the NF-κB pathway and inflammatory signaling, (II) a correlation between mitochondrial morphology, respiratory function and transcriptomics, (III) machine learning predicted novel roles of genes and pathways in autophagic processes and (IV) individual gene function inferences and interactions among biological processes via multi-feature clustering. These studies provide a platform to interrogate underexplored aspects of AD biology and offer several specific hypotheses for future drug discovery efforts.


Assuntos
Doença de Alzheimer/genética , Autofagia/genética , Modelos Genéticos , Agregação Patológica de Proteínas/genética , Proteínas tau/metabolismo , Doença de Alzheimer/patologia , Encéfalo/patologia , Sistemas CRISPR-Cas/genética , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Engenharia Genética , Humanos , Aprendizado de Máquina , Mitocôndrias/genética , Mitocôndrias/patologia , Neurônios , Agregação Patológica de Proteínas/patologia , Transdução de Sinais/genética
5.
Neuropharmacology ; 108: 229-37, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27039042

RESUMO

Neurosteroids such as progesterone and allopregnanolone have been shown to exert neuroprotective effects under a variety of pathological or insult conditions, and there is evidence that the neurosteroid system is perturbed in Multiple Sclerosis (MS) patients. Neurosteroids are synthesized in the central nervous system (CNS) through a series of metabolic transformations, beginning with a rate-limiting step of cholesterol transport through the outer mitochondrial membrane via the transporter translocator protein (TSPO). We examined the effects of etifoxine and XBD-173, two different brain penetrant TSPO agonists, for their ability to ameliorate clinical signs in two different experimental autoimmune encephalitis (EAE) models. Etifoxine, as previously reported, was efficacious in EAE, while XBD-173 was not. Surprisingly, XBD-173, but not etifoxine elevated relevant neurosteroids in brain of female rats and differed in its ability to exert anti-inflammatory and direct neuroprotective effects in vitro as compared to etifoxine. We conclude that the neurosteroid elevations produced in brain by XBD-173 are not sufficient to ameliorate EAE and suggest that etifoxine may have additional mechanisms of action that provide therapeutic benefit in this model system.


Assuntos
Modelos Animais de Doenças , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/metabolismo , Oxazinas/metabolismo , Purinas/metabolismo , Receptores de GABA/metabolismo , Animais , Linhagem Celular Tumoral , Células Cultivadas , Relação Dose-Resposta a Droga , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Oxazinas/uso terapêutico , Purinas/uso terapêutico , Ratos , Resultado do Tratamento
6.
Nat Commun ; 6: 8045, 2015 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-26289944

RESUMO

Autophagy is an important degradation pathway, which is induced after starvation, where it buffers nutrient deprivation by recycling macromolecules in organisms from yeast to man. While the classical pathway mediating this response is via mTOR inhibition, there are likely to be additional pathways that support the process. Here, we identify Annexin A2 as an autophagy modulator that regulates autophagosome formation by enabling appropriate ATG9A trafficking from endosomes to autophagosomes via actin. This process is dependent on the Annexin A2 effectors ARP2 and Spire1. Annexin A2 expression increases after starvation in cells in an mTOR-independent fashion. This is mediated via Jun N-terminal kinase activation of c-Jun, which, in turn, enhances the trans-activation of the Annexin A2 promoter. Annexin A2 knockdown abrogates starvation-induced autophagy, while its overexpression induces autophagy. Hence, c-Jun-mediated transcriptional responses support starvation-induced autophagy by regulating Annexin A2 expression levels.


Assuntos
Anexina A2/metabolismo , Autofagia/fisiologia , Regulação da Expressão Gênica/fisiologia , Animais , Anexina A2/genética , Proteínas Relacionadas à Autofagia , Fibroblastos , Genes jun , Células HeLa , Humanos , MAP Quinase Quinase 4 , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
7.
Hum Mol Genet ; 22(22): 4528-44, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23804751

RESUMO

Inhibition of the insulin/insulin-like growth factor signalling pathway increases lifespan and protects against neurodegeneration in model organisms, and has been considered as a potential therapeutic target. This pathway is upstream of mTORC1, a negative regulator of autophagy. Thus, we expected autophagy to be activated by insulin-like growth factor-1 (IGF-1) inhibition, which could account for many of its beneficial effects. Paradoxically, we found that IGF-1 inhibition attenuates autophagosome formation. The reduced amount of autophagosomes present in IGF-1R depleted cells can be, at least in part, explained by a reduced formation of autophagosomal precursors at the plasma membrane. In particular, IGF-1R depletion inhibits mTORC2, which, in turn, reduces the activity of protein kinase C (PKCα/ß). This perturbs the actin cytoskeleton dynamics and decreases the rate of clathrin-dependent endocytosis, which impacts autophagosome precursor formation. Finally, with important implications for human diseases, we demonstrate that pharmacological inhibition of the IGF-1R signalling cascade reduces autophagy also in zebrafish and mice models. The novel link we describe here has important consequences for the interpretation of genetic experiments in mammalian systems and for evaluating the potential of targeting the IGF-1R receptor or modulating its signalling through the downstream pathway for therapeutic purposes under clinically relevant conditions, such as neurodegenerative diseases, where autophagy stimulation is considered beneficial.


Assuntos
Autofagia/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 1/genética , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Células HeLa , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Macrolídeos/farmacologia , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/patologia , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
8.
J Cell Biol ; 196(4): 483-96, 2012 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-22351926

RESUMO

Macroautophagy (in this paper referred to as autophagy) and the ubiquitin-proteasome system are the two major catabolic systems in cells. Autophagy involves sequestration of cytosolic contents in double membrane-bounded vesicles called autophagosomes. The membrane source for autophagosomes has received much attention, and diverse sources, such as the plasma membrane, Golgi, endoplasmic reticulum, and mitochondria, have been implicated. These may not be mutually exclusive, but the exact sources and mechanism involved in the formation of autophagosomes are still unclear. In this paper, we identify a positive role for the small G protein Arf6 in autophagosome formation. The effect of Arf6 on autophagy is mediated by its role in the generation of phosphatidylinositol 4,5-bisphosphate (PIP(2)) and in inducing phospholipase D (PLD) activity. PIP(2) and PLD may themselves promote autophagosome biogenesis by influencing endocytic uptake of plasma membrane into autophagosome precursors. However, Arf6 may also influence autophagy by indirect effects, such as either by regulating membrane flow from other compartments or by modulating PLD activity independently of the mammalian target of rapamycin.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Autofagia , Fagossomos/fisiologia , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfolipase D/metabolismo , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/genética , Animais , Proteínas Relacionadas à Autofagia , Western Blotting , Células CHO , Proteínas de Transporte/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cricetinae , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Imunofluorescência , Proteínas Ativadoras de GTPase/metabolismo , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Imunossupressores/farmacologia , Fagossomos/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo
9.
Cell ; 146(2): 303-17, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21784250

RESUMO

Autophagy is a catabolic process in which lysosomes degrade intracytoplasmic contents transported in double-membraned autophagosomes. Autophagosomes are formed by the elongation and fusion of phagophores, which can be derived from preautophagosomal structures coming from the plasma membrane and other sites like the endoplasmic reticulum and mitochondria. The mechanisms by which preautophagosomal structures elongate their membranes and mature toward fully formed autophagosomes still remain unknown. Here, we show that the maturation of the early Atg16L1 precursors requires homotypic fusion, which is essential for subsequent autophagosome formation. Atg16L1 precursor homotypic fusion depends on the SNARE protein VAMP7 together with partner SNAREs. Atg16L1 precursor homotypic fusion is a critical event in the early phases of autophagy that couples membrane acquisition and autophagosome biogenesis, as this step regulates the size of the vesicles, which in turn appears to influence their subsequent maturation into LC3-positive autophagosomes.


Assuntos
Autofagia , Fagossomos/metabolismo , Proteínas Relacionadas à Autofagia , Proteínas de Transporte/metabolismo , Vesículas Citoplasmáticas/metabolismo , Células HeLa , Humanos , Membranas Intracelulares/metabolismo , Proteínas R-SNARE/metabolismo , Proteínas SNARE/metabolismo
10.
Physiol Rev ; 90(4): 1383-435, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20959619

RESUMO

(Macro)autophagy is a bulk degradation process that mediates the clearance of long-lived proteins and organelles. Autophagy is initiated by double-membraned structures, which engulf portions of cytoplasm. The resulting autophagosomes ultimately fuse with lysosomes, where their contents are degraded. Although the term autophagy was first used in 1963, the field has witnessed dramatic growth in the last 5 years, partly as a consequence of the discovery of key components of its cellular machinery. In this review we focus on mammalian autophagy, and we give an overview of the understanding of its machinery and the signaling cascades that regulate it. As recent studies have also shown that autophagy is critical in a range of normal human physiological processes, and defective autophagy is associated with diverse diseases, including neurodegeneration, lysosomal storage diseases, cancers, and Crohn's disease, we discuss the roles of autophagy in health and disease, while trying to critically evaluate if the coincidence between autophagy and these conditions is causal or an epiphenomenon. Finally, we consider the possibility of autophagy upregulation as a therapeutic approach for various conditions.


Assuntos
Autofagia/fisiologia , Células Eucarióticas/metabolismo , Mamíferos/fisiologia , Animais , Células Eucarióticas/patologia , Humanos , Fagossomos/metabolismo , Transdução de Sinais , Estresse Fisiológico
11.
J Cell Biol ; 190(6): 1023-37, 2010 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-20855506

RESUMO

Parkinson's disease (PD) is characterized pathologically by intraneuronal inclusions called Lewy bodies, largely comprised of α-synuclein. Multiplication of the α-synuclein gene locus increases α-synuclein expression and causes PD. Thus, overexpression of wild-type α-synuclein is toxic. In this study, we demonstrate that α-synuclein overexpression impairs macroautophagy in mammalian cells and in transgenic mice. Our data show that α-synuclein compromises autophagy via Rab1a inhibition and Rab1a overexpression rescues the autophagy defect caused by α-synuclein. Inhibition of autophagy by α-synuclein overexpression or Rab1a knockdown causes mislocalization of the autophagy protein, Atg9, and decreases omegasome formation. Rab1a, α-synuclein, and Atg9 all regulate formation of the omegasome, which marks autophagosome precursors.


Assuntos
Autofagia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , alfa-Sinucleína/metabolismo , Animais , Linhagem Celular Tumoral , Drosophila melanogaster/metabolismo , Técnicas de Silenciamento de Genes , Complexo de Golgi/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Biológicos , Fagossomos/metabolismo , Transporte Proteico , Vesículas Secretórias/metabolismo , Proteínas rab1 de Ligação ao GTP/metabolismo
12.
Autophagy ; 6(8): 1184-6, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20861674

RESUMO

The membrane origin of autophagosomes has long been a mystery and it may involve multiple sources. In this punctum, we discuss our recent finding that the plasma membrane contributes to the formation of pre-autophagic structures via clathrin-mediated endocytosis. Our study suggests that Atg16L1 interacts with clathrin heavy-chain/AP2 and is also localized on vesicles (positive for clathrin or cholera toxin B) close to the plasma membrane. Live-cell imaging studies revealed that the plasma membrane contributes to Atg16L1-positive structures and that this process and autophagosome formation are impaired by knockdowns of genes regulating clathrin-mediated endocytosis.


Assuntos
Autofagia , Membrana Celular/metabolismo , Fagossomos/metabolismo , Animais , Endocitose , Endossomos/metabolismo , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Biológicos
13.
Nat Cell Biol ; 12(8): 747-57, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20639872

RESUMO

Autophagy is a catabolic process in which lysosomes degrade intracytoplasmic contents transported in double-membraned autophagosomes. Autophagosomes are formed by the elongation and fusion of phagophores, which derive from pre-autophagosomal structures. The membrane origins of autophagosomes are unclear and may involve multiple sources, including the endoplasmic reticulum and mitochondria. Here we show in mammalian cells that the heavy chain of clathrin interacts with Atg16L1 and is involved in the formation of Atg16L1-positive early autophagosome precursors. Atg16L1 associated with clathrin-coated structures, and inhibition of clathrin-mediated internalization decreased the formation of both Atg16L1-positive precursors and mature autophagosomes. We tested and demonstrated that the plasma membrane contributes directly to the formation of early Atg16L1-positive autophagosome precursors. This may be particularly important during periods of increased autophagosome formation, because the plasma membrane may serve as a large membrane reservoir that allows cells periods of autophagosome synthesis at levels many-fold higher than under basal conditions, without compromising other processes.


Assuntos
Autofagia/fisiologia , Membrana Celular/metabolismo , Fagossomos/metabolismo , Proteínas Relacionadas à Autofagia , Western Blotting , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Membrana Celular/ultraestrutura , Clatrina/genética , Clatrina/metabolismo , Células HeLa , Humanos , Imuno-Histoquímica , Microscopia Eletrônica de Transmissão , Modelos Biológicos , Fagossomos/ultraestrutura , Ligação Proteica , Deleção de Sequência/genética
15.
Autophagy ; 5(5): 585-9, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19411822

RESUMO

Recent years have seen the realization that macroautophagy (which we will call autophagy) is not only important in yeast but is necessary for diverse functions in plants and animals. Importantly, autophagy can have an impact on human pathologies including infectious diseases, cancers, and neurodegenerative conditions. Thus, we need to be able to measure autophagy accurately in order to understand how it can be regulated physiologically and with exogenous agents.


Assuntos
Autofagia/fisiologia , Animais , Autofagia/efeitos dos fármacos , Humanos , Macrolídeos/farmacologia , Proteínas Associadas aos Microtúbulos/metabolismo , Fagossomos/efeitos dos fármacos , Fagossomos/metabolismo
16.
Methods Enzymol ; 453: 83-110, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19216903

RESUMO

Autophagy has emerged as a field of rapidly growing interest with implications in several disease conditions, such as cancer, infectious diseases, and neurodegenerative diseases. Autophagy is a major degradation pathway for aggregate-prone, intracytosolic proteins causing neurodegenerative disorders, such as Huntington's disease and forms of Parkinson's disease. Up-regulating autophagy may be a tractable therapeutic intervention for clearing these disease-causing proteins. The identification of autophagy-enhancing compounds would be beneficial not only in neurodegenerative diseases but also in other conditions where up-regulating autophagy may act as a protective pathway. Furthermore, small molecule modulators of autophagy may also be useful in dissecting pathways governing mammalian autophagy. In this chapter, we highlight assays that can be used for the identification of autophagy regulators, such as measuring the clearance of mutant aggregate-prone proteins or of autophagic flux with bafilomycin A(1). Using these methods, we recently described several mTOR-independent autophagy-enhancing compounds that have protective effects in various models of Huntington's disease.


Assuntos
Autofagia/fisiologia , Degeneração Neural/metabolismo , Proteínas/metabolismo , Animais , Western Blotting , Eletroforese em Gel de Poliacrilamida , Células PC12 , Proteínas Quinases/metabolismo , Ratos , Serina-Treonina Quinases TOR
17.
J Cell Sci ; 121(Pt 10): 1649-60, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18430781

RESUMO

Huntington disease (HD) is caused by a polyglutamine-expansion mutation in huntingtin (HTT) that makes the protein toxic and aggregate-prone. The subcellular localisation of huntingtin and many of its interactors suggest a role in endocytosis, and recently it has been shown that huntingtin interacts indirectly with the early endosomal protein Rab5 through HAP40. Here we show that Rab5 inhibition enhanced polyglutamine toxicity, whereas Rab5 overexpression attenuated toxicity in our cell and fly models of HD. We tried to identify a mechanism for the Rab5 effects in our HD model systems, and our data suggest that Rab5 acts at an early stage of autophagosome formation in a macromolecular complex that contains beclin 1 (BECN1) and Vps34. Interestingly chemical or genetic inhibition of endocytosis also impeded macroautophagy, and enhanced aggregation and toxicity of mutant huntingtin. However, in contrast to Rab5, inhibition of endocytosis by various means suppressed autophagosome-lysosome fusion (the final step in the macroautophagy pathway) similar to bafilomycin A1. Thus, Rab5, which has previously been thought to be exclusively involved in endocytosis, has a new role in macroautophagy. We have previously shown that macroautophagy is an important clearance route for several aggregate-prone proteins including mutant huntingtin. Thus, better understanding of Rab5-regulated autophagy might lead to rational therapeutic targets for HD and other protein-conformation diseases.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Transporte/metabolismo , Doença de Huntington/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Animais , Autofagia , Células COS , Chlorocebus aethiops , Modelos Animais de Doenças , Drosophila , Células HeLa , Humanos , Transfecção
18.
Methods Mol Biol ; 445: 195-211, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18425452

RESUMO

The accumulation of mutant aggregate-prone proteins is a feature of several human disorders, collectively referred to as protein conformation disorders or proteinopathies. We have shown that autophagy, a cytosolic, non-specific bulk degradation system, is an important clearance route for many cytosolic toxic, aggregate-prone proteins, like mutant huntingtin and mutant alpha-synucleins. Induction of autophagy enhances the clearance of both soluble and aggregated forms of the mutant protein, and protects against toxicity caused by these mutations in cell, fly, and mouse models. Inhibition of autophagy has opposite effects. Thus, the autophagic pathway may represent a possible therapeutic target in the treatment of certain protein conformation disorders.


Assuntos
Autofagia/fisiologia , Proteínas Mutantes/metabolismo , Animais , Western Blotting , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Eletroforese em Gel de Poliacrilamida , Proteína Huntingtina , Doença de Huntington/metabolismo , Imuno-Histoquímica , Proteínas Mutantes/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ratos , Sinucleínas/genética , Sinucleínas/metabolismo
19.
Curr Top Dev Biol ; 76: 89-101, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17118264

RESUMO

Intracellular protein misfolding/aggregation are features of many late-onset neurodegenerative diseases, called proteinopathies. These include Alzheimer's disease, Parkinson's disease, tauopathies, and polyglutamine expansion diseases [e.g., Huntington's disease; and various spinocerebellar ataxias (SCAs), like SCA3]. There are no effective strategies to slow or prevent the neurodegeneration resulting from these diseases in humans. The mutations causing many proteinopathies (e.g., polyglutamine diseases and tauopathies) confer novel toxic functions on the specific protein, and disease severity frequently correlates with the expression levels of the protein. Thus, the factors regulating the synthesis and clearance of these aggregate-prone proteins are putative therapeutic targets. The proteasome and autophagy-lysosomal pathways are the major routes for mutant huntingtin fragment clearance. While the narrow proteasome barrel precludes entry of oligomers/aggregates of mutant huntingtin (or other aggregate-prone intracellular proteins), such substrates can be degraded by macroautophagy (which we will call autophagy). We showed that the autophagy inducer rapamycin reduced the levels of soluble and aggregated huntingtin and attenuated its toxicity in cells, and in transgenic Drosophila and mouse models. We extended the range of intracellular proteinopathy substrates that are cleared by autophagy to a wide range of other targets, including proteins mutated in certain SCAs, forms of alpha-synuclein mutated in familial forms of Parkinson's disease, and tau mutants that cause frontotemporal dementia/tauopathy. In this chapter, we consider the therapeutic potential of autophagy upregulation for various proteinopathies, and describe how this strategy may act both by removing the primary toxin (the misfolded/aggregate-prone protein) and by reducing susceptibility to apoptotic insults.


Assuntos
Autofagia , Citosol/metabolismo , Proteínas/metabolismo , Animais , Apoptose , Humanos , Doença de Huntington/metabolismo , Doença de Huntington/terapia , Modelos Biológicos , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/terapia , Complexo de Endopeptidases do Proteassoma/metabolismo , Dobramento de Proteína , Proteínas Quinases/metabolismo , Estrutura Quaternária de Proteína , Proteínas/química , Transdução de Sinais , Serina-Treonina Quinases TOR , Ubiquitina/metabolismo
20.
Mol Aspects Med ; 27(5-6): 520-7, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16973207

RESUMO

Macroautophagy (henceforth referred to simply as autophagy) is a bulk degradation process involved in the clearance of long-lived proteins, protein complexes and organelles. A portion of the cytosol, with its contents to be degraded, is enclosed by double-membrane structures called autophagosomes/autophagic vacuoles, which ultimately fuse with lysosomes where their contents are degraded. In this review, we will describe how induction of autophagy is protective against toxic intracytosolic aggregate-prone proteins that cause a range of neurodegenerative diseases. Autophagy is a key clearance pathway involved in the removal of such proteins, including mutant huntingtin (that causes Huntington's disease), mutant ataxin-3 (that causes spinocerebellar ataxia type 3), forms of tau that cause tauopathies, and forms of alpha-synuclein that cause familial Parkinson's disease. Induction of autophagy enhances the clearance of both soluble and aggregated forms of such proteins, and protects against toxicity of a range of these mutations in cell and animal models. Interestingly, the aggregates formed by mutant huntingtin sequester and inactivate the mammalian target of rapamycin (mTOR), a key negative regulator of autophagy. This results in induction of autophagy in cells with these aggregates.


Assuntos
Autofagia , Doença de Huntington/metabolismo , Doença de Huntington/terapia , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Animais , Humanos , Doença de Huntington/genética , Mutação/genética , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA