Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Cell ; 146(2): 303-17, 2011 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-21784250

RESUMEN

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.


Asunto(s)
Autofagia , Fagosomas/metabolismo , Proteínas Relacionadas con la Autofagia , Proteínas Portadoras/metabolismo , Vesículas Citoplasmáticas/metabolismo , Células HeLa , Humanos , Membranas Intracelulares/metabolismo , Proteínas R-SNARE/metabolismo , Proteínas SNARE/metabolismo
2.
Physiol Rev ; 90(4): 1383-435, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20959619

RESUMEN

(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.


Asunto(s)
Autofagia/fisiología , Células Eucariotas/metabolismo , Mamíferos/fisiología , Animales , Células Eucariotas/patología , Humanos , Fagosomas/metabolismo , Transducción de Señal , Estrés Fisiológico
3.
Hum Mol Genet ; 22(22): 4528-44, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23804751

RESUMEN

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.


Asunto(s)
Autofagia/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/genética , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular , Inhibidores Enzimáticos/farmacología , Células HeLa , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Macrólidos/farmacología , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/patología , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
4.
Nat Genet ; 37(7): 771-6, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15980862

RESUMEN

Mutations that affect the dynein motor machinery are sufficient to cause motor neuron disease. It is not known why there are aggregates or inclusions in affected tissues in mice with such mutations and in most forms of human motor neuron disease. Here we identify a new mechanism of inclusion formation by showing that decreased dynein function impairs autophagic clearance of aggregate-prone proteins. We show that mutations of the dynein machinery enhanced the toxicity of the mutation that causes Huntington disease in fly and mouse models. Furthermore, loss of dynein function resulted in premature aggregate formation by mutant huntingtin and increased levels of the autophagosome marker LC3-II in both cell culture and mouse models, compatible with impaired autophagosome-lysosome fusion.


Asunto(s)
Adenina/análogos & derivados , Autofagia , Dineínas/genética , Enfermedad de Huntington/patología , Mutación , Adenina/farmacología , Adenilil Imidodifosfato/farmacología , Animales , Conducta Animal , Encéfalo/patología , Células COS , Chlorocebus aethiops , Cruzamientos Genéticos , Dípteros , Dineínas/antagonistas & inhibidores , Dineínas/metabolismo , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Cuerpos de Inclusión/metabolismo , Ratones , Ratones Mutantes , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Células PC12 , Complejo de la Endopetidasa Proteasomal/metabolismo , Ratas , Sinucleínas
5.
Mol Brain ; 17(1): 26, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38778381

RESUMEN

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.


Asunto(s)
Neuronas , Enfermedad de Parkinson , Agregado de Proteínas , Proteómica , Proteostasis , alfa-Sinucleína , alfa-Sinucleína/metabolismo , Animales , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Neuronas/metabolismo , Neuronas/patología , Ratones , Mapas de Interacción de Proteínas , Proteoma/metabolismo
6.
SLAS Discov ; 29(2): 100137, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38128829

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer , Células Madre Pluripotentes Inducidas , Humanos , Proteínas tau/genética , Proteínas tau/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedad de Alzheimer/metabolismo , Neuronas/metabolismo
7.
Nat Genet ; 36(6): 585-95, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15146184

RESUMEN

Huntington disease is one of nine inherited neurodegenerative disorders caused by a polyglutamine tract expansion. Expanded polyglutamine proteins accumulate abnormally in intracellular aggregates. Here we show that mammalian target of rapamycin (mTOR) is sequestered in polyglutamine aggregates in cell models, transgenic mice and human brains. Sequestration of mTOR impairs its kinase activity and induces autophagy, a key clearance pathway for mutant huntingtin fragments. This protects against polyglutamine toxicity, as the specific mTOR inhibitor rapamycin attenuates huntingtin accumulation and cell death in cell models of Huntington disease, and inhibition of autophagy has the converse effects. Furthermore, rapamycin protects against neurodegeneration in a fly model of Huntington disease, and the rapamycin analog CCI-779 improved performance on four different behavioral tasks and decreased aggregate formation in a mouse model of Huntington disease. Our data provide proof-of-principle for the potential of inducing autophagy to treat Huntington disease.


Asunto(s)
Enfermedad de Huntington/tratamiento farmacológico , Inhibidores de Proteínas Quinasas , Animales , Autofagia , Células COS , Modelos Animales de Enfermedad , Drosophila melanogaster , Femenino , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Sustancias Macromoleculares , Masculino , Ratones , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Péptidos/química , Péptidos/genética , Péptidos/metabolismo , Biosíntesis de Proteínas , Proteínas Quinasas/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR
8.
Eur J Pharmacol ; 934: 175301, 2022 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-36191631

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer , Animales , Ratones , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Proteínas tau/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Ratones Transgénicos , Encéfalo , Sirolimus/farmacología , Modelos Animales de Enfermedad
9.
Sci Rep ; 11(1): 2879, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33536571

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/genética , Autofagia/genética , Modelos Genéticos , Agregación Patológica de Proteínas/genética , Proteínas tau/metabolismo , Enfermedad de Alzheimer/patología , Encéfalo/patología , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Regulación de la Expresión Génica , Ingeniería Genética , Humanos , Aprendizaje Automático , Mitocondrias/genética , Mitocondrias/patología , Neuronas , Agregación Patológica de Proteínas/patología , Transducción de Señal/genética
10.
Nat Neurosci ; 9(6): 824-31, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16699508

RESUMEN

Post-translational modification by the lipid palmitate is crucial for the correct targeting and function of many proteins. Here we show that huntingtin (htt) is normally palmitoylated at cysteine 214, which is essential for its trafficking and function. The palmitoylation and distribution of htt are regulated by the palmitoyl transferase huntingtin interacting protein 14 (HIP14). Expansion of the polyglutamine tract of htt, which causes Huntington disease, results in reduced interaction between mutant htt and HIP14 and consequently in a marked reduction in palmitoylation. Mutation of the palmitoylation site of htt, making it palmitoylation resistant, accelerates inclusion formation and increases neuronal toxicity. Downregulation of HIP14 in mouse neurons expressing wild-type and mutant htt increases inclusion formation, whereas overexpression of HIP14 substantially reduces inclusions. These results suggest that the expansion of the polyglutamine tract in htt results in decreased palmitoylation, which contributes to the formation of inclusion bodies and enhanced neuronal toxicity.


Asunto(s)
Proteínas Portadoras/metabolismo , Corteza Cerebral/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Ácido Palmítico/metabolismo , Aciltransferasas , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos/fisiología , Animales , Animales Recién Nacidos , Células COS , Proteínas Portadoras/genética , Células Cultivadas , Corteza Cerebral/citología , Chlorocebus aethiops , Cisteína/metabolismo , Regulación hacia Abajo/genética , Humanos , Proteína Huntingtina , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/metabolismo , Ratones , Ratones Transgénicos , Mutación/genética , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Proteínas Nucleares/química , Proteínas Nucleares/genética , Péptidos/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Transporte de Proteínas/fisiología , Ratas , Expansión de Repetición de Trinucleótido/genética
11.
Methods Mol Biol ; 445: 195-211, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18425452

RESUMEN

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.


Asunto(s)
Autofagia/fisiología , Proteínas Mutantes/metabolismo , Animales , Western Blotting , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Electroforesis en Gel de Poliacrilamida , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Inmunohistoquímica , Proteínas Mutantes/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ratas , Sinucleínas/genética , Sinucleínas/metabolismo
12.
Mol Aspects Med ; 27(5-6): 520-7, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16973207

RESUMEN

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.


Asunto(s)
Autofagia , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/terapia , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Animales , Humanos , Enfermedad de Huntington/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética
13.
Neuropharmacology ; 108: 229-37, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27039042

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/metabolismo , Oxazinas/metabolismo , Purinas/metabolismo , Receptores de GABA/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Relación Dosis-Respuesta a Droga , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Ligandos , Ratones , Ratones Endogámicos C57BL , Oxazinas/uso terapéutico , Purinas/uso terapéutico , Ratas , Resultado del Tratamiento
14.
Nat Commun ; 6: 8045, 2015 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-26289944

RESUMEN

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.


Asunto(s)
Anexina A2/metabolismo , Autofagia/fisiología , Regulación de la Expresión Génica/fisiología , Animales , Anexina A2/genética , Proteínas Relacionadas con la Autofagia , Fibroblastos , Genes jun , Células HeLa , Humanos , MAP Quinasa Quinasa 4 , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
15.
Int J Biochem Cell Biol ; 36(12): 2541-50, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15325591

RESUMEN

Large cytoplasmic inclusions called aggresomes are seen in many protein conformational diseases including Huntington's disease and Parkinson's disease. The roles of inclusions and aggresomes in these diseases are unresolved critical issues that have been vigorously debated. Two recent studies used microtubule disruption with nocodazole to inhibit aggresome formation and observed increased toxicity of expanded polyglutamines in the context of huntingtin exon 1 and a truncated androgen receptor. Increased toxicity of expanded polyglutamines in the presence of nocodazole was correlated with decreased protein turnover, leading the authors to conclude that aggresomes were cytoprotective and that they directly enhanced clearance of the toxic proteins. Here we show that nocodazole has additional effects, which provide a simple alternative explanation for these previous observations. We confirmed aggresome formation in cells expressing proteins with polyalanine and polyglutamine expansions. As expected, we found a reduction in aggresome formation when microtubule function was disrupted using nocodazole. However, in addition to this effect, nocodazole treatment increased the proportions of cells with nuclear inclusions in PC12 cells expressing huntingtin exon 1 with 74 glutamines. This can be explained as nocodazole inhibits autophagosome-lysosome fusion, a key step in mutant huntingtin exon 1 clearance. This effect alone can explain the previous observations with this compound in polyglutamine diseases and raises doubts about the interpretation of some of the data that have been used to argue that aggresomes protect against polyglutamine mutations.


Asunto(s)
Autofagia/fisiología , Cuerpos de Inclusión/fisiología , Lisosomas/fisiología , Microtúbulos/fisiología , Enfermedades Neurodegenerativas/fisiopatología , Péptidos/fisiología , Animales , Autofagia/efectos de los fármacos , Células COS , Chlorocebus aethiops , Humanos , Enfermedad de Huntington/fisiopatología , Lisosomas/efectos de los fármacos , Fusión de Membrana , Microtúbulos/efectos de los fármacos , Nocodazol/farmacología , Organismos Modificados Genéticamente , Células PC12 , Enfermedad de Parkinson/fisiopatología , Ratas
16.
Neuroreport ; 15(16): 2443-5, 2004 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-15538170

RESUMEN

Protein conformation disorders or proteinopathies are a growing family of human diseases that are characterized by the accumulation of proteins in intracellular aggregates (also known as inclusions) in specific tissues/organs. The role of aggregates in these diseases has been a subject of vigorous debate. However, irrespective of the nature(s) of the toxic species, it is desirable for cells to be able to control the levels of these toxic proteins and restrict their accumulation. Here we discuss how the autophagy-lysosome pathway may regulate protein clearance in some of the protein conformation disorders and why this pathway may represent a possible therapeutic target in such conditions.


Asunto(s)
Autofagia/fisiología , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades Neurodegenerativas/prevención & control , Proteínas/toxicidad , Animales , Humanos , Lisosomas/fisiología , Conformación Proteica , Pliegue de Proteína
17.
J Cell Biol ; 196(4): 483-96, 2012 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-22351926

RESUMEN

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.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Autofagia , Fagosomas/fisiología , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfolipasa D/metabolismo , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/genética , Animales , Proteínas Relacionadas con la Autofagia , Western Blotting , Células CHO , Proteínas Portadoras/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Cricetinae , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Técnica del Anticuerpo Fluorescente , Proteínas Activadoras de GTPasa/metabolismo , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Inmunosupresores/farmacología , Fagosomas/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo
18.
Autophagy ; 6(8): 1184-6, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20861674

RESUMEN

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.


Asunto(s)
Autofagia , Membrana Celular/metabolismo , Fagosomas/metabolismo , Animales , Endocitosis , Endosomas/metabolismo , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Biológicos
19.
Nat Cell Biol ; 12(8): 747-57, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20639872

RESUMEN

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.


Asunto(s)
Autofagia/fisiología , Membrana Celular/metabolismo , Fagosomas/metabolismo , Proteínas Relacionadas con la Autofagia , Western Blotting , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Membrana Celular/ultraestructura , Clatrina/genética , Clatrina/metabolismo , Células HeLa , Humanos , Inmunohistoquímica , Microscopía Electrónica de Transmisión , Modelos Biológicos , Fagosomas/ultraestructura , Unión Proteica , Eliminación de Secuencia/genética
20.
J Cell Biol ; 190(6): 1023-37, 2010 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-20855506

RESUMEN

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.


Asunto(s)
Autofagia , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , alfa-Sinucleína/metabolismo , Animales , Línea Celular Tumoral , Drosophila melanogaster/metabolismo , Técnicas de Silenciamiento del Gen , Aparato de Golgi/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Biológicos , Fagosomas/metabolismo , Transporte de Proteínas , Vesículas Secretoras/metabolismo , Proteínas de Unión al GTP rab1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA