Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Curr Biol ; 32(12): 2581-2595.e6, 2022 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-35580604

RESUMEN

Autophagy is a conserved, multi-step process of capturing proteolytic cargo in autophagosomes for lysosome degradation. The capacity to remove toxic proteins that accumulate in neurodegenerative disorders attests to the disease-modifying potential of the autophagy pathway. However, neurons respond only marginally to conventional methods for inducing autophagy, limiting efforts to develop therapeutic autophagy modulators for neurodegenerative diseases. The determinants underlying poor autophagy induction in neurons and the degree to which neurons and other cell types are differentially sensitive to autophagy stimuli are incompletely defined. Accordingly, we sampled nascent transcript synthesis and stabilities in fibroblasts, induced pluripotent stem cells (iPSCs), and iPSC-derived neurons (iNeurons), thereby uncovering a neuron-specific stability of transcripts encoding myotubularin-related phosphatase 5 (MTMR5). MTMR5 is an autophagy suppressor that acts with its binding partner, MTMR2, to dephosphorylate phosphoinositides critical for autophagy initiation and autophagosome maturation. We found that MTMR5 is necessary and sufficient to suppress autophagy in iNeurons and undifferentiated iPSCs. Using optical pulse labeling to visualize the turnover of endogenously encoded proteins in live cells, we observed that knockdown of MTMR5 or MTMR2, but not the unrelated phosphatase MTMR9, significantly enhances neuronal degradation of TDP-43, an autophagy substrate implicated in several neurodegenerative diseases. Our findings thus establish a regulatory mechanism of autophagy intrinsic to neurons and targetable for clearing disease-related proteins in a cell-type-specific manner. In so doing, our results not only unravel novel aspects of neuronal biology and proteostasis but also elucidate a strategy for modulating neuronal autophagy that could be of high therapeutic potential for multiple neurodegenerative diseases.


Asunto(s)
Autofagosomas , Proteínas Tirosina Fosfatasas no Receptoras , Autofagosomas/metabolismo , Autofagia/genética , Neuronas/fisiología , Proteínas Tirosina Fosfatasas no Receptoras/genética , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo
2.
Autophagy ; 18(2): 254-282, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34057020

RESUMEN

Mechanisms of protein homeostasis are crucial for overseeing the clearance of misfolded and toxic proteins over the lifetime of an organism, thereby ensuring the health of neurons and other cells of the central nervous system. The highly conserved pathway of autophagy is particularly necessary for preventing and counteracting pathogenic insults that may lead to neurodegeneration. In line with this, mutations in genes that encode essential autophagy factors result in impaired autophagy and lead to neurodegenerative conditions such as amyotrophic lateral sclerosis (ALS). However, the mechanistic details underlying the neuroprotective role of autophagy, neuronal resistance to autophagy induction, and the neuron-specific effects of autophagy-impairing mutations remain incompletely defined. Further, the manner and extent to which non-cell autonomous effects of autophagy dysfunction contribute to ALS pathogenesis are not fully understood. Here, we review the current understanding of the interplay between autophagy and ALS pathogenesis by providing an overview of critical steps in the autophagy pathway, with special focus on pivotal factors impaired by ALS-causing mutations, their physiologic effects on autophagy in disease models, and the cell type-specific mechanisms regulating autophagy in non-neuronal cells which, when impaired, can contribute to neurodegeneration. This review thereby provides a framework not only to guide further investigations of neuronal autophagy but also to refine therapeutic strategies for ALS and related neurodegenerative diseases.Abbreviations: ALS: amyotrophic lateral sclerosis; Atg: autophagy-related; CHMP2B: charged multivesicular body protein 2B; DPR: dipeptide repeat; FTD: frontotemporal dementia; iPSC: induced pluripotent stem cell; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; PINK1: PTEN induced kinase 1; RNP: ribonuclear protein; sALS: sporadic ALS; SPHK1: sphingosine kinase 1; TARDBP/TDP-43: TAR DNA binding protein; TBK1: TANK-binding kinase 1; TFEB: transcription factor EB; ULK: unc-51 like autophagy activating kinase; UPR: unfolded protein response; UPS: ubiquitin-proteasome system; VCP: valosin containing protein.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Esclerosis Amiotrófica Lateral/metabolismo , Autofagia/fisiología , Demencia Frontotemporal/genética , Humanos , Proteostasis , Respuesta de Proteína Desplegada
3.
J Biol Chem ; 297(3): 101003, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34303705

RESUMEN

Autophagy is an evolutionarily conserved pathway mediating the breakdown of cellular proteins and organelles. Emphasizing its pivotal nature, autophagy dysfunction contributes to many diseases; nevertheless, development of effective autophagy modulating drugs is hampered by fundamental deficiencies in available methods for measuring autophagic activity or flux. To overcome these limitations, we introduced the photoconvertible protein Dendra2 into the MAP1LC3B locus of human cells via CRISPR/Cas9 genome editing, enabling accurate and sensitive assessments of autophagy in living cells by optical pulse labeling. We used this assay to perform high-throughput drug screens of four chemical libraries comprising over 30,000 diverse compounds, identifying several clinically relevant drugs and novel autophagy modulators. A select series of candidate compounds also modulated autophagy flux in human motor neurons modified by CRISPR/Cas9 to express GFP-labeled LC3. Using automated microscopy, we tested the therapeutic potential of autophagy induction in several distinct neuronal models of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). In doing so, we found that autophagy induction exhibited discordant effects, improving survival in disease models involving the RNA binding protein TDP-43, while exacerbating toxicity in neurons expressing mutant forms of UBQLN2 and C9ORF72 associated with familial ALS/FTD. These studies confirm the utility of the Dendra2-LC3 assay, while illustrating the contradictory effects of autophagy induction in different ALS/FTD subtypes.


Asunto(s)
Autofagia , Proteínas Adaptadoras Transductoras de Señales/genética , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/terapia , Autofagia/efectos de los fármacos , Proteínas Relacionadas con la Autofagia/genética , Proteína C9orf72/genética , Sistemas CRISPR-Cas , Proteínas de Unión al ADN/genética , Ensayos de Selección de Medicamentos Antitumorales , Demencia Frontotemporal/genética , Demencia Frontotemporal/terapia , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Proteínas Luminiscentes/genética , Proteínas Asociadas a Microtúbulos/genética , Modelos Biológicos , Neuronas Motoras/metabolismo , Mutación
4.
Cell Rep ; 17(1): 125-136, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27681426

RESUMEN

Spinal and bulbar muscular atrophy (SBMA), a progressive degenerative disorder, is caused by a CAG/glutamine expansion in the androgen receptor (polyQ AR). Recent studies demonstrate that skeletal muscle is an important site of toxicity that contributes to the SBMA phenotype. Here, we sought to identify critical pathways altered in muscle that underlie disease manifestations in AR113Q mice. This led to the unanticipated identification of gene expression changes affecting regulators of carbohydrate metabolism, similar to those triggered by denervation. AR113Q muscle exhibits diminished glycolysis, altered mitochondria, and an impaired response to exercise. Strikingly, the expression of genes regulating muscle energy metabolism is rescued following peripheral polyQ AR gene silencing by antisense oligonucleotides (ASO), a therapeutic strategy that alleviates disease. Our data establish the occurrence of a metabolic imbalance in SBMA muscle triggered by peripheral expression of the polyQ AR and indicate that alterations in energy utilization contribute to non-neuronal disease manifestations.


Asunto(s)
Silenciador del Gen , Atrofia Muscular Espinal/terapia , Oligonucleótidos Antisentido/farmacología , Receptores Androgénicos/genética , Animales , Metabolismo de los Hidratos de Carbono/genética , Ciclo del Ácido Cítrico/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Glucólisis/genética , Humanos , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Mitocondrias/patología , Músculo Esquelético , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/patología , Péptidos/química , Péptidos/metabolismo , Condicionamiento Físico Animal , Receptores Androgénicos/metabolismo
5.
J Clin Invest ; 125(2): 831-45, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25607844

RESUMEN

Expansion of the polyglutamine (polyQ) tract within the androgen receptor (AR) causes neuromuscular degeneration in individuals with spinobulbar muscular atrophy (SBMA). PolyQ AR has diminished transcriptional function and exhibits ligand-dependent proteotoxicity, features that have both been implicated in SBMA; however, the extent to which altered AR transcriptional function contributes to pathogenesis remains controversial. Here, we sought to dissociate effects of diminished AR function from polyQ-mediated proteotoxicity by enhancing the transcriptional activity of polyQ AR. To accomplish this, we bypassed the inhibitory effect of AR SUMOylation (where SUMO indicates small ubiquitin-like modifier) by mutating conserved lysines in the polyQ AR that are sites of SUMOylation. We determined that replacement of these residues by arginine enhances polyQ AR activity as a hormone-dependent transcriptional regulator. In a murine model, disruption of polyQ AR SUMOylation rescued exercise endurance and type I muscle fiber atrophy; it also prolonged survival. These changes occurred without overt alterations in polyQ AR expression or aggregation, revealing the favorable trophic support exerted by the ligand-activated receptor. Our findings demonstrate beneficial effects of enhancing the transcriptional function of the ligand-activated polyQ AR and indicate that the SUMOylation pathway may be a potential target for therapeutic intervention in SBMA.


Asunto(s)
Fibras Musculares de Contracción Lenta/metabolismo , Trastornos Musculares Atróficos/metabolismo , Péptidos/metabolismo , Receptores Androgénicos/metabolismo , Sumoilación , Transcripción Genética , Animales , Ratones , Ratones Transgénicos , Fibras Musculares de Contracción Lenta/patología , Trastornos Musculares Atróficos/genética , Trastornos Musculares Atróficos/patología , Células PC12 , Péptidos/genética , Ratas , Receptores Androgénicos/genética
6.
Endocrinology ; 155(7): 2624-34, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24742193

RESUMEN

Spinal and bulbar muscular atrophy (SBMA) is a late-onset, progressive neurodegenerative disease linked to a polyglutamine (polyQ) expansion in the androgen receptor (AR). Men affected by SBMA show marked muscle weakness and atrophy, typically emerging midlife. Given the androgen-dependent nature of this disease, one might expect AR antagonists to have therapeutic value for treating SBMA. However, current work from animal models suggests otherwise, raising questions about whether polyQ-expanded AR exerts androgen-dependent toxicity through mechanisms distinct from normal AR function. In this study, we asked whether the nonsteroidal AR antagonist flutamide, delivered via a time-release pellet, could reverse or prevent androgen-dependent AR toxicity in three different mouse models of SBMA: the AR97Q transgenic (Tg) model, a knock-in (KI) model, and a myogenic Tg model. We find that flutamide protects mice from androgen-dependent AR toxicity in all three SBMA models, preventing or reversing motor dysfunction in the Tg models and significantly extending the life span in KI males. Given that flutamide effectively protects against androgen-dependent disease in three different mouse models of SBMA, our data are proof of principle that AR antagonists have therapeutic potential for treating SBMA in humans and support the notion that toxicity caused by polyQ-expanded AR uses at least some of the same mechanisms as normal AR before diverging to produce disease and muscle atrophy.


Asunto(s)
Andrógenos/metabolismo , Atrofia Bulboespinal Ligada al X/tratamiento farmacológico , Modelos Animales de Enfermedad , Flutamida/farmacología , Antagonistas de Andrógenos/farmacología , Animales , Western Blotting , Atrofia Bulboespinal Ligada al X/genética , Atrofia Bulboespinal Ligada al X/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Orquiectomía , Péptidos/genética , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Análisis de Supervivencia , Factores de Tiempo , Resultado del Tratamiento , Expansión de Repetición de Trinucleótido/genética
7.
Hum Mol Genet ; 23(5): 1376-86, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24150846

RESUMEN

Spinobulbar muscular atrophy (SBMA) is an inherited neuromuscular disorder caused by the expansion of a CAG repeat encoding a polyglutamine tract in exon 1 of the androgen receptor (AR) gene. SBMA demonstrates androgen-dependent toxicity due to unfolding and aggregation of the mutant protein. There are currently no disease-modifying therapies, but of increasing interest for therapeutic targeting is autophagy, a highly conserved cellular process mediating protein quality control. We have previously shown that genetic manipulations inhibiting autophagy diminish skeletal muscle atrophy and extend the lifespan of AR113Q knock-in mice. In contrast, manipulations inducing autophagy worsen muscle atrophy, suggesting that chronic, aberrant upregulation of autophagy contributes to pathogenesis. Since the degree to which autophagy is altered in SBMA and the mechanisms responsible for such alterations are incompletely defined, we sought to delineate autophagic status in SBMA using both cellular and mouse models. Here, we confirm that autophagy is induced in cellular and knock-in mouse models of SBMA and show that the transcription factors transcription factor EB (TFEB) and ZKSCAN3 operate in opposing roles to underlie these changes. We demonstrate upregulation of TFEB target genes in skeletal muscle from AR113Q male mice and SBMA patients. Furthermore, we observe a greater response in AR113Q mice to physiological stimulation of autophagy by both nutrient starvation and exercise. Taken together, our results indicate that transcriptional signaling contributes to autophagic dysregulation and provides a mechanistic framework for the pathologic increase of autophagic responsiveness in SBMA.


Asunto(s)
Autofagia/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Regulación de la Expresión Génica , Trastornos Musculares Atróficos/genética , Factores de Transcripción/metabolismo , Activación Transcripcional , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Transgénicos , Trastornos Musculares Atróficos/metabolismo , Péptidos/genética , Condicionamiento Físico Animal , Receptores Androgénicos/genética
8.
CNS Neurol Disord Drug Targets ; 12(8): 1146-56, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24040817

RESUMEN

We review the genetic and clinical features of spinobulbar muscular atrophy (SBMA), a progressive neuromuscular disorder caused by a CAG/glutamine tract expansion in the androgen receptor. SBMA was the first polyglutamine disease to be discovered, and we compare and contrast it with related degenerative disorders of the nervous system caused by expanded glutamine tracts. We review the cellular and animals models that have been most widely used to study this disorder, and highlight insights into disease pathogenesis derived from this work. These model systems have revealed critical aspects of the disease, including its hormone dependence, a feature that underlies disease occurrence only in men with the mutant allele. We discuss how this and other findings have been translated to clinical trials for SBMA patients, and examine emerging therapeutic targets that have been identified by recent work.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Terapia Genética/métodos , Trastornos Musculares Atróficos/diagnóstico , Trastornos Musculares Atróficos/terapia , Animales , Ensayos Clínicos como Asunto/métodos , Humanos , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Trastornos Musculares Atróficos/genética , Transducción de Señal/fisiología
9.
Nat Chem Biol ; 9(2): 112-8, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23222885

RESUMEN

We sought new strategies to reduce amounts of the polyglutamine androgen receptor (polyQ AR) and achieve benefits in models of spinobulbar muscular atrophy, a protein aggregation neurodegenerative disorder. Proteostasis of the polyQ AR is controlled by the heat shock protein 90 (Hsp90)- and Hsp70-based chaperone machinery, but mechanisms regulating the protein's turnover are incompletely understood. We demonstrate that overexpression of Hsp70 interacting protein (Hip), a co-chaperone that enhances binding of Hsp70 to its substrates, promotes client protein ubiquitination and polyQ AR clearance. Furthermore, we identify a small molecule that acts similarly to Hip by allosterically promoting Hsp70 binding to unfolded substrates. Like Hip, this synthetic co-chaperone enhances client protein ubiquitination and polyQ AR degradation. Both genetic and pharmacologic approaches targeting Hsp70 alleviate toxicity in a Drosophila model of spinobulbar muscular atrophy. These findings highlight the therapeutic potential of allosteric regulators of Hsp70 and provide new insights into the role of the chaperone machinery in protein quality control.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Péptidos/química , Animales , Relación Dosis-Respuesta a Droga , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacología , Drosophila , Femenino , Células HEK293 , Células HeLa , Humanos , Concentración 50 Inhibidora , Modelos Químicos , Chaperonas Moleculares/química , Trastornos Musculares Atróficos/metabolismo , Neurotoxinas/química , Células PC12 , Estructura Terciaria de Proteína , Proteínas/química , Piridinas/farmacología , Ratas , Receptores Androgénicos/química , Receptores Androgénicos/metabolismo , Tiazoles/farmacología , Ubiquitinación
10.
Toxicol Lett ; 176(2): 149-56, 2008 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-18155859

RESUMEN

The impact of endocrine disrupting chemical (EDC) exposure on human health is receiving increasingly focused attention. The prototypical EDC bisphenol A (BPA) is an estrogenic high-production chemical used primarily as a monomer for the production of polycarbonate and epoxy resins. It is now well established that there is ubiquitous human exposure to BPA. In the general population, exposure to BPA occurs mainly by consumption of contaminated foods and beverages that have contacted epoxy resins or polycarbonate plastics. To test the hypothesis that bioactive BPA was released from polycarbonate bottles used for consumption of water and other beverages, we evaluated whether BPA migrated into water stored in new or used high-quality polycarbonate bottles used by consumers. Using a sensitive and quantitative competitive enzyme-linked immunosorbent assay, BPA was found to migrate from polycarbonate water bottles at rates ranging from 0.20 ng/h to 0.79 ng/h. At room temperature the migration of BPA was independent of whether or not the bottle had been previously used. Exposure to boiling water (100 degrees C) increased the rate of BPA migration by up to 55-fold. The estrogenic bioactivity of the BPA-like immunoreactivity released into the water samples was confirmed using an in vitro assay of rapid estrogen signaling and neurotoxicity in developing cerebellar neurons. The amounts of BPA found to migrate from polycarbonate drinking bottles should be considered as a contributing source to the total "EDC-burden" to which some individuals are exposed.


Asunto(s)
Neuronas/efectos de los fármacos , Fenoles/toxicidad , Cemento de Policarboxilato/química , Embalaje de Productos , Animales , Animales Recién Nacidos , Compuestos de Bencidrilo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cerebelo/citología , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Disruptores Endocrinos/química , Disruptores Endocrinos/metabolismo , Disruptores Endocrinos/toxicidad , Ensayo de Inmunoadsorción Enzimática , Estradiol/farmacología , Estrógenos/toxicidad , Femenino , Humanos , Inmunoquímica , L-Lactato Deshidrogenasa/metabolismo , Neuronas/citología , Neuronas/metabolismo , Fenoles/química , Fenoles/metabolismo , Ratas , Ratas Sprague-Dawley , Temperatura , Factores de Tiempo , Agua/análisis , Agua/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...