Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-37205440

RESUMO

Proteasome activity is crucial for cellular integrity, but how tissues adjust proteasome content in response to catabolic stimuli is uncertain. Here, we demonstrate that transcriptional coordination by multiple transcription factors is required to increase proteasome content and activate proteolysis in catabolic states. Using denervated mouse muscle as a model system for accelerated proteolysis in vivo , we reveal that a two-phase transcriptional program activates genes encoding proteasome subunits and assembly chaperones to boost an increase in proteasome content. Initially, gene induction is necessary to maintain basal proteasome levels, and in a more delayed phase (7-10 d after denervation) it stimulates proteasome assembly to meet cellular demand for excessive proteolysis. Intriguingly, the transcription factors PAX4 and α-PAL NRF-1 control the expression of proteasome among other genes in a combinatorial manner, driving cellular adaptation to muscle denervation. Consequently, PAX4 and α-PAL NRF-1 represent new therapeutic targets to inhibit proteolysis in catabolic diseases (e.g. type-2 diabetes, cancer).

2.
EMBO Rep ; 23(12): e55175, 2022 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-36278411

RESUMO

Maintenance of desmin intermediate filaments (IF) is vital for muscle plasticity and function, and their perturbed integrity due to accelerated loss or aggregation causes atrophy and myopathies. Calpain-1-mediated disassembly of ubiquitinated desmin IF is a prerequisite for desmin loss, myofibril breakdown, and atrophy. Because calpain-1 does not harbor a bona fide ubiquitin-binding domain, the precise mechanism for desmin IF disassembly remains unknown. Here, we demonstrate that the AAA-ATPase, ATAD1, is required to facilitate disassembly and turnover of ubiquitinated desmin IF. We identified PLAA and UBXN4 as ATAD1's interacting partners, and their downregulation attenuated desmin loss upon denervation. The ATAD1-PLAA-UBXN4 complex binds desmin filaments and promotes a release of phosphorylated and ubiquitinated species into the cytosol, presenting ATAD1 as the only known AAA-ATPase that preferentially acts on phosphorylated substrates. Desmin filaments disassembly was accelerated by the coordinated functions of Atad1 and calpain-1, which interact in muscle. Thus, by extracting ubiquitinated desmin from the insoluble filament, ATAD1 may expose calpain-1 cleavage sites on desmin, consequently enhancing desmin solubilization and degradation in the cytosol.


Assuntos
Filamentos Intermediários , Músculos , ATPases Associadas a Diversas Atividades Celulares
3.
FEBS J ; 289(10): 2755-2770, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-33825342

RESUMO

Desmin is the primary intermediate filament (IF) of cardiac, skeletal, and smooth muscle. By linking the contractile myofibrils to the sarcolemma and cellular organelles, desmin IF contributes to muscle structural and cellular integrity, force transmission, and mitochondrial homeostasis. Mutations in desmin cause myofibril misalignment, mitochondrial dysfunction, and impaired mechanical integrity leading to cardiac and skeletal myopathies in humans, often characterized by the accumulation of protein aggregates. Recent evidence indicates that desmin filaments also regulate proteostasis and cell size. In skeletal muscle, changes in desmin filament dynamics can facilitate catabolic events as an adaptive response to a changing environment. In addition, post-translational modifications of desmin and its misfolding in the heart have emerged as key determinants of homeostasis and disease. In this review, we provide an overview of the structural and cellular roles of desmin and propose new models for its novel functions in preserving the homeostasis of striated muscles.


Assuntos
Desmina , Músculo Esquelético , Miofibrilas , Desmina/genética , Desmina/fisiologia , Homeostase , Humanos , Músculo Esquelético/fisiologia , Doenças Musculares/genética , Doenças Musculares/metabolismo , Miofibrilas/fisiologia
5.
Am J Physiol Cell Physiol ; 321(3): C615-C631, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34319828

RESUMO

The size and shape of skeletal muscle fibers are affected by various physiological and pathological conditions, such as muscle atrophy, hypertrophy, regeneration, and dystrophies. Hence, muscle fiber cross-sectional area (CSA) is an important determinant of muscle health and plasticity. We adapted the Imaris software to automatically segment muscle fibers based on fluorescent labeling of the plasma membrane and measure muscle fiber CSA. Analysis of muscle cross sections by the Imaris semiautomated and manual approaches demonstrated a similar decrease in CSA of atrophying muscles from fasted mice compared with fed controls. In addition, we previously demonstrated that downregulation of the Ca2+-specific protease calpain-1 attenuates muscle atrophy. Accordingly, both the Imaris semiautomated and manual approaches showed a similar increase in CSA of fibers expressing calpain-1 shRNA compared with adjacent nontransfected fibers in the same muscle cross section. Although both approaches seem valid for measurements of muscle fiber size, the manual marking method is less preferable because it is highly time-consuming, subjective, and limits the number of cells that can be analyzed. The Imaris semiautomated approach is user-friendly, requires little training or optimization, and can be used to efficiently and accurately mark thousands of fibers in a short period. As a novel addition to the commonly used statistics, we also describe statistical tests that quantify the strength of an effect on fiber size, enabling detection of significant differences between skewed distributions that would otherwise not be detected using typical methods.


Assuntos
Tamanho Celular , Processamento de Imagem Assistida por Computador , Microscopia Confocal , Microscopia de Fluorescência , Fibras Musculares Esqueléticas/patologia , Atrofia Muscular/patologia , Software , Animais , Automação Laboratorial , Calpaína/genética , Calpaína/metabolismo , Modelos Animais de Doenças , Jejum , Imunofluorescência , Masculino , Camundongos Endogâmicos ICR , Fibras Musculares Esqueléticas/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/metabolismo
6.
Biomolecules ; 11(1)2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33467597

RESUMO

Protein degradation maintains cellular integrity by regulating virtually all biological processes, whereas impaired proteolysis perturbs protein quality control, and often leads to human disease. Two major proteolytic systems are responsible for protein breakdown in all cells: autophagy, which facilitates the loss of organelles, protein aggregates, and cell surface proteins; and the ubiquitin-proteasome system (UPS), which promotes degradation of mainly soluble proteins. Recent findings indicate that more complex protein structures, such as filamentous assemblies, which are not accessible to the catalytic core of the proteasome in vitro, can be efficiently degraded by this proteolytic machinery in systemic catabolic states in vivo. Mechanisms that loosen the filamentous structure seem to be activated first, hence increasing the accessibility of protein constituents to the UPS. In this review, we will discuss the mechanisms underlying the disassembly and loss of the intricate insoluble filamentous myofibrils, which are responsible for muscle contraction, and whose degradation by the UPS causes weakness and disability in aging and disease. Several lines of evidence indicate that myofibril breakdown occurs in a strictly ordered and controlled manner, and the function of AAA-ATPases is crucial for their disassembly and loss.


Assuntos
Miofibrilas/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Animais , Humanos , Proteínas Musculares/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
7.
Biochim Biophys Acta Mol Cell Res ; 1867(10): 118788, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32603758

RESUMO

Muscle atrophy is an inevitable sequel of fasting, denervation, aging, exposure to microgravity, and many human diseases including, cancer, type-2 diabetes, and renal failure. During atrophy the destruction of the muscle's fundamental contractile machinery, the myofibrils, is accelerated leading to a reduction in muscle mass, weakness, frailty, and physical disability. Recent findings indicate that atrophy can be a major cause of death in affected individuals, and inhibition of muscle wasting is likely to prolong survival. Major advances in our understanding of the mechanisms for myofibril breakdown in atrophy include the discovery of biological pathways and key components that play prominent roles. On fasting or denervation, degradation of myofibrillar proteins requires an initial dissociation of the desmin cytoskeleton, whose integrity is critical for myofibril stability. This loss of desmin filaments involves phosphorylation, ubiquitination, and subsequent depolymerization by calpain-1, and appears to reduce myofibrils integrity and facilitate their destruction. Consequently, depolymerization of desmin filament in atrophy seems to be an early key event for overall proteolysis. A focus of this review is to discuss these new insights and the specific role of calpain-1 in promoting desmin filaments loss, and to highlight important key questions that merit further study.


Assuntos
Calpaína/metabolismo , Desmina/metabolismo , Atrofia Muscular/metabolismo , Polimerização , Animais , Humanos , Miofibrilas/metabolismo , Ubiquitinação
8.
EMBO Rep ; 21(4): e48791, 2020 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-32133736

RESUMO

PI3K-Akt-FoxO-mTOR signaling is the central pathway controlling growth and metabolism in all cells. Ubiquitination of the protein kinase Akt prior to its phosphorylation is required for PI3K-Akt activity. Here, we found that the deubiquitinating (DUB) enzyme USP1 removes K63-linked polyubiquitin chains on Akt to restrict PI3K-Akt-FoxO signaling in mouse muscle during prolonged starvation. DUB screening platform identified USP1 as a direct DUB for Akt, and USP1 depletion in mouse muscle increased Akt ubiquitination, PI3K-Akt-FoxO signaling, and glucose uptake during fasting. Co-immunoprecipitation and mass spectrometry identified disabled homolog-2 (Dab2), the tuberous sclerosis complex TSC1/TSC2, and PHLPP1 as USP1 bound proteins. During starvation, Dab2 is essential for Akt recruitment to USP1-TSC1-PHLPP1 complex, and for PI3K-Akt-FoxO inhibition. Surprisingly, USP1 limits TSC1 levels to sustain mTOR-mediated basal protein synthesis rates and maintain its own protein levels. We propose that Dab2 recruits Akt to USP1-TSC1-PHLPP1 complex to efficiently terminate the transmission of growth signals when cellular energy level is low.


Assuntos
Proteína Forkhead Box O1 , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Inanição , Proteases Específicas de Ubiquitina/metabolismo , Animais , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Camundongos , Músculos/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proteases Específicas de Ubiquitina/genética , Ubiquitinação
9.
Nat Commun ; 11(1): 1381, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-32170063

RESUMO

Signaling through the insulin receptor governs central physiological functions related to cell growth and metabolism. Here we show by tandem native protein complex purification approach and super-resolution STED microscopy that insulin receptor activity requires association with the fundamental structural module in muscle, the dystrophin glycoprotein complex (DGC), and the desmosomal component plakoglobin (γ-catenin). The integrity of this high-molecular-mass assembly renders skeletal muscle susceptibility to insulin, because DGC-insulin receptor dissociation by plakoglobin downregulation reduces insulin signaling and causes atrophy. Furthermore, low insulin receptor activity in muscles from transgenic or fasted mice decreases plakoglobin-DGC-insulin receptor content on the plasma membrane, but not when plakoglobin is overexpressed. By masking ß-dystroglycan LIR domains, plakoglobin prevents autophagic clearance of plakoglobin-DGC-insulin receptor co-assemblies and maintains their function. Our findings establish DGC as a signaling hub, and provide a possible mechanism for the insulin resistance in Duchenne Muscular Dystrophy, and for the cardiomyopathies seen with plakoglobin mutations.


Assuntos
Distrofina/metabolismo , Glicoproteínas/metabolismo , Músculo Esquelético/metabolismo , Receptor de Insulina/metabolismo , Transdução de Sinais , gama Catenina/metabolismo , Animais , Autofagia , Membrana Celular/metabolismo , Modelos Animais de Doenças , Distroglicanas/metabolismo , Distrofina/genética , Masculino , Camundongos , Camundongos Transgênicos , Distrofia Muscular de Duchenne/metabolismo , Domínios Proteicos
10.
BMC Med ; 18(1): 8, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31959160

RESUMO

BACKGROUND: Dystroglycanopathies are a group of inherited disorders characterized by vast clinical and genetic heterogeneity and caused by abnormal functioning of the ECM receptor dystroglycan (Dg). Remarkably, among many cases of diagnosed dystroglycanopathies, only a small fraction can be linked directly to mutations in Dg or its regulatory enzymes, implying the involvement of other, not-yet-characterized, Dg-regulating factors. To advance disease diagnostics and develop new treatment strategies, new approaches to find dystroglycanopathy-related factors should be considered. The Dg complex is highly evolutionarily conserved; therefore, model genetic organisms provide excellent systems to address this challenge. In particular, Drosophila is amenable to experiments not feasible in any other system, allowing original insights about the functional interactors of the Dg complex. METHODS: To identify new players contributing to dystroglycanopathies, we used Drosophila as a genetic muscular dystrophy model. Using mass spectrometry, we searched for muscle-specific Dg interactors. Next, in silico analyses allowed us to determine their association with diseases and pathological conditions in humans. Using immunohistochemical, biochemical, and genetic interaction approaches followed by the detailed analysis of the muscle tissue architecture, we verified Dg interaction with some of the discovered factors. Analyses of mouse muscles and myocytes were used to test if interactions are conserved in vertebrates. RESULTS: The muscle-specific Dg complexome revealed novel components that influence the efficiency of Dg function in the muscles. We identified the closest human homologs for Dg-interacting partners, determined their significant enrichment in disease-associations, and verified some of the newly identified Dg interactions. We found that Dg associates with two components of the mechanosignaling Hippo pathway: the WW domain-containing proteins Kibra and Yorkie. Importantly, this conserved interaction manages adult muscle size and integrity. CONCLUSIONS: The results presented in this study provide a new list of muscle-specific Dg interactors, further analysis of which could aid not only in the diagnosis of muscular dystrophies, but also in the development of new therapeutics. To regulate muscle fitness during aging and disease, Dg associates with Kibra and Yorkie and acts as a transmembrane Hippo signaling receptor that transmits extracellular information to intracellular signaling cascades, regulating muscle gene expression.


Assuntos
Proteínas de Drosophila/metabolismo , Distroglicanas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Atrofia Muscular/metabolismo , Distrofias Musculares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Envelhecimento/metabolismo , Animais , Modelos Animais de Doenças , Drosophila , Distroglicanas/genética , Feminino , Masculino , Espectrometria de Massas , Camundongos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/patologia , Distrofias Musculares/genética , Distrofias Musculares/patologia , Mutação , Mapas de Interação de Proteínas
11.
J Cell Biol ; 217(10): 3698-3714, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30061109

RESUMO

Myofibril breakdown is a fundamental cause of muscle wasting and inevitable sequel of aging and disease. We demonstrated that myofibril loss requires depolymerization of the desmin cytoskeleton, which is activated by phosphorylation. Here, we developed a mass spectrometry-based kinase-trap assay and identified glycogen synthase kinase 3-ß (GSK3-ß) as responsible for desmin phosphorylation. GSK3-ß inhibition in mice prevented desmin phosphorylation and depolymerization and blocked atrophy upon fasting or denervation. Desmin was phosphorylated by GSK3-ß 3 d after denervation, but depolymerized only 4 d later when cytosolic Ca2+ levels rose. Mass spectrometry analysis identified GSK3-ß and the Ca2+-specific protease, calpain-1, bound to desmin and catalyzing its disassembly. Consistently, calpain-1 down-regulation prevented loss of phosphorylated desmin and blocked atrophy. Thus, phosphorylation of desmin filaments by GSK3-ß is a key molecular event required for calpain-1-mediated depolymerization, and the subsequent myofibril destruction. Consequently, GSK3-ß represents a novel drug target to prevent myofibril breakdown and atrophy.


Assuntos
Calpaína/biossíntese , Desmina/metabolismo , Regulação para Baixo , Regulação da Expressão Gênica no Desenvolvimento , Glicogênio Sintase Quinase 3 beta/metabolismo , Atrofia Muscular/metabolismo , Miofibrilas/metabolismo , Animais , Cálcio/metabolismo , Calpaína/genética , Desmina/genética , Glicogênio Sintase Quinase 3 beta/genética , Masculino , Camundongos , Atrofia Muscular/genética , Atrofia Muscular/patologia , Miofibrilas/genética , Miofibrilas/patologia , Fosforilação/genética
12.
Nat Commun ; 9(1): 1759, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29717115

RESUMO

Autophagy is an intracellular degradation process essential for adaptation to metabolic stress. DAPK2 is a calmodulin-regulated protein kinase, which has been implicated in autophagy regulation, though the mechanism is unclear. Here, we show that the central metabolic sensor, AMPK, phosphorylates DAPK2 at a critical site in the protein structure, between the catalytic and the calmodulin-binding domains. This phosphorylation activates DAPK2 by functionally mimicking calmodulin binding and mitigating an inhibitory autophosphorylation, providing a novel, alternative mechanism for DAPK2 activation during metabolic stress. In addition, we show that DAPK2 phosphorylates the core autophagic machinery protein, Beclin-1, leading to dissociation of its inhibitor, Bcl-XL. Importantly, phosphorylation of DAPK2 by AMPK enhances DAPK2's ability to phosphorylate Beclin-1, and depletion of DAPK2 reduces autophagy in response to AMPK activation. Our study reveals a unique calmodulin-independent mechanism for DAPK2 activation, critical to its function as a novel downstream effector of AMPK in autophagy.


Assuntos
Adenilato Quinase/metabolismo , Autofagia , Proteínas Quinases Associadas com Morte Celular/metabolismo , Estresse Fisiológico , Células A549 , Sequência de Aminoácidos , Animais , Proteína Beclina-1/metabolismo , Catálise , Proteínas Quinases Associadas com Morte Celular/química , Dimerização , Células HCT116 , Células HEK293 , Humanos , Masculino , Camundongos , Mutação , Fosforilação , Homologia de Sequência de Aminoácidos , Serina/metabolismo , Treonina/metabolismo , Proteína bcl-X/metabolismo
13.
Nature ; 547(7662): 179-184, 2017 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-28581497

RESUMO

The adult mammalian heart is non-regenerative owing to the post-mitotic nature of cardiomyocytes. The neonatal mouse heart can regenerate, but only during the first week of life. Here we show that changes in the composition of the extracellular matrix during this week can affect cardiomyocyte growth and differentiation in mice. We identify agrin, a component of neonatal extracellular matrix, as required for the full regenerative capacity of neonatal mouse hearts. In vitro, recombinant agrin promotes the division of cardiomyocytes that are derived from mouse and human induced pluripotent stem cells through a mechanism that involves the disassembly of the dystrophin-glycoprotein complex, and Yap- and ERK-mediated signalling. In vivo, a single administration of agrin promotes cardiac regeneration in adult mice after myocardial infarction, although the degree of cardiomyocyte proliferation observed in this model suggests that there are additional therapeutic mechanisms. Together, our results uncover a new inducer of mammalian heart regeneration and highlight fundamental roles of the extracellular matrix in cardiac repair.


Assuntos
Agrina/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Coração/fisiologia , Regeneração , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Animais Recém-Nascidos , Proteínas de Ciclo Celular , Proliferação de Células , Distroglicanas/metabolismo , Feminino , Camundongos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/citologia , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Sinalização YAP
14.
Proc Natl Acad Sci U S A ; 114(8): E1375-E1384, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28096335

RESUMO

A hallmark of muscle atrophy is the excessive degradation of myofibrillar proteins primarily by the ubiquitin proteasome system. In mice, during the rapid muscle atrophy induced by fasting, the desmin cytoskeleton and the attached Z-band-bound thin filaments are degraded after ubiquitination by the ubiquitin ligase tripartite motif-containing protein 32 (Trim32). To study the order of events leading to myofibril destruction, we investigated the slower atrophy induced by denervation (disuse). We show that myofibril breakdown is a two-phase process involving the initial disassembly of desmin filaments by Trim32, which leads to the later myofibril breakdown by enzymes, whose expression is increased by the paired box 4 (PAX4) transcription factor. After denervation of mouse tibialis anterior muscles, phosphorylation and Trim32-dependent ubiquitination of desmin filaments increased rapidly and stimulated their gradual depolymerization (unlike their rapid degradation during fasting). Trim32 down-regulation attenuated the loss of desmin and myofibrillar proteins and reduced atrophy. Although myofibrils and desmin filaments were intact at 7 d after denervation, inducing the dissociation of desmin filaments caused an accumulation of ubiquitinated proteins and rapid destruction of myofibrils. The myofibril breakdown normally observed at 14 d after denervation required not only dissociation of desmin filaments, but also gene induction by PAX4. Down-regulation of PAX4 or its target gene encoding the p97/VCP ATPase reduced myofibril disassembly and degradation on denervation or fasting. Thus, during atrophy, the initial loss of desmin is critical for the subsequent myofibril destruction, and over time, myofibrillar proteins become more susceptible to PAX4-induced enzymes that promote proteolysis.


Assuntos
Desmina/metabolismo , Proteínas de Homeodomínio/metabolismo , Atrofia Muscular/metabolismo , Miofibrilas/metabolismo , Fatores de Transcrição Box Pareados/metabolismo , Adenosina Trifosfatases/metabolismo , Animais , Citoesqueleto/metabolismo , Regulação para Baixo/fisiologia , Masculino , Camundongos , Denervação Muscular/métodos , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Proteínas Nucleares/metabolismo , Ubiquitina/metabolismo , Ubiquitinação/fisiologia
15.
Nat Rev Drug Discov ; 14(1): 58-74, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25549588

RESUMO

Atrophy occurs in specific muscles with inactivity (for example, during plaster cast immobilization) or denervation (for example, in patients with spinal cord injuries). Muscle wasting occurs systemically in older people (a condition known as sarcopenia); as a physiological response to fasting or malnutrition; and in many diseases, including chronic obstructive pulmonary disorder, cancer-associated cachexia, diabetes, renal failure, cardiac failure, Cushing syndrome, sepsis, burns and trauma. The rapid loss of muscle mass and strength primarily results from excessive protein breakdown, which is often accompanied by reduced protein synthesis. This loss of muscle function can lead to reduced quality of life, increased morbidity and mortality. Exercise is the only accepted approach to prevent or slow atrophy. However, several promising therapeutic agents are in development, and major advances in our understanding of the cellular mechanisms that regulate the protein balance in muscle include the identification of several cytokines, particularly myostatin, and a common transcriptional programme that promotes muscle wasting. Here, we discuss these new insights and the rationally designed therapies that are emerging to combat muscle wasting.


Assuntos
Atrofia Muscular/diagnóstico , Atrofia Muscular/tratamento farmacológico , Qualidade de Vida , Animais , Humanos , Atrofia Muscular/epidemiologia , Miofibrilas/efeitos dos fármacos , Miofibrilas/patologia , Miostatina/farmacologia , Miostatina/uso terapêutico , Sepse/diagnóstico , Sepse/tratamento farmacológico , Sepse/epidemiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Síndrome de Emaciação/diagnóstico , Síndrome de Emaciação/tratamento farmacológico , Síndrome de Emaciação/epidemiologia
16.
Mol Biol Cell ; 25(25): 3988-98, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25318673

RESUMO

The 26S proteasome recognizes a vast number of ubiquitin-dependent degradation signals linked to various substrates. This recognition is mediated mainly by the stoichiometric proteasomal resident ubiquitin receptors S5a and Rpn13, which harbor ubiquitin-binding domains. Regulatory steps in substrate binding, processing, and subsequent downstream proteolytic events by these receptors are poorly understood. Here we demonstrate that mammalian S5a is present in proteasome-bound and free states. S5a is required for efficient proteasomal degradation of polyubiquitinated substrates and the recruitment of ubiquitin-like (Ubl) harboring proteins; however, S5a-mediated ubiquitin and Ubl binding occurs only on the proteasome itself. We identify the VWA domain of S5a as a domain that limits ubiquitin and Ubl binding to occur only upon proteasomal association. Multiubiquitination events within the VWA domain can further regulate S5a association. Our results provide a molecular explanation to how ubiquitin and Ubl binding to S5a is restricted to the 26S proteasome.


Assuntos
Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Fator 4 Ativador da Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Células HEK293 , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Poliubiquitina/metabolismo , Complexo de Endopeptidases do Proteassoma/química , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteólise , Proteínas de Ligação a RNA , Proteínas Ubiquitinadas/metabolismo , Ubiquitinação
17.
J Cell Biol ; 204(5): 747-58, 2014 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-24567360

RESUMO

Activation of the PI3K-Akt-FoxO pathway induces cell growth, whereas its inhibition reduces cell survival and, in muscle, causes atrophy. Here, we report a novel mechanism that suppresses PI3K-Akt-FoxO signaling. Although skeletal muscle lacks desmosomes, it contains multiple desmosomal components, including plakoglobin. In normal muscle plakoglobin binds the insulin receptor and PI3K subunit p85 and promotes PI3K-Akt-FoxO signaling. During atrophy, however, its interaction with PI3K-p85 is reduced by the ubiquitin ligase Trim32 (tripartite motif containing protein 32). Inhibition of Trim32 enhanced plakoglobin binding to PI3K-p85 and promoted PI3K-Akt-FoxO signaling. Surprisingly, plakoglobin overexpression alone enhanced PI3K-Akt-FoxO signaling. Furthermore, Trim32 inhibition in normal muscle increased PI3K-Akt-FoxO signaling, enhanced glucose uptake, and induced fiber growth, whereas plakoglobin down-regulation reduced PI3K-Akt-FoxO signaling, decreased glucose uptake, and caused atrophy. Thus, by promoting plakoglobin-PI3K dissociation, Trim32 reduces PI3K-Akt-FoxO signaling in normal and atrophying muscle. This mechanism probably contributes to insulin resistance during fasting and catabolic diseases and perhaps to the myopathies and cardiomyopathies seen with Trim32 and plakoglobin mutations.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Atrofia Muscular/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ubiquitina-Proteína Ligases/fisiologia , gama Catenina/metabolismo , Animais , Regulação para Baixo , Privação de Alimentos , Técnicas de Silenciamento de Genes , Masculino , Camundongos , Camundongos Endogâmicos , Modelos Biológicos , Receptor de Insulina/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , gama Catenina/genética
18.
J Cell Biol ; 198(4): 575-89, 2012 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-22908310

RESUMO

During muscle atrophy, myofibrillar proteins are degraded in an ordered process in which MuRF1 catalyzes ubiquitylation of thick filament components (Cohen et al. 2009. J. Cell Biol. http://dx.doi.org/10.1083/jcb.200901052). Here, we show that another ubiquitin ligase, Trim32, ubiquitylates thin filament (actin, tropomyosin, troponins) and Z-band (α-actinin) components and promotes their degradation. Down-regulation of Trim32 during fasting reduced fiber atrophy and the rapid loss of thin filaments. Desmin filaments were proposed to maintain the integrity of thin filaments. Accordingly, we find that the rapid destruction of thin filament proteins upon fasting was accompanied by increased phosphorylation of desmin filaments, which promoted desmin ubiquitylation by Trim32 and degradation. Reducing Trim32 levels prevented the loss of both desmin and thin filament proteins. Furthermore, overexpression of an inhibitor of desmin polymerization induced disassembly of desmin filaments and destruction of thin filament components. Thus, during fasting, desmin phosphorylation increases and enhances Trim32-mediated degradation of the desmin cytoskeleton, which appears to facilitate the breakdown of Z-bands and thin filaments.


Assuntos
Citoesqueleto de Actina/metabolismo , Desmina/metabolismo , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Miofibrilas/patologia , Sarcômeros/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação/fisiologia , Actinas/metabolismo , Animais , Jejum/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos , Miofibrilas/metabolismo , Fosforilação/fisiologia , Ubiquitina-Proteína Ligases/genética
19.
J Cell Biol ; 185(6): 1083-95, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19506036

RESUMO

Loss of myofibrillar proteins is a hallmark of atrophying muscle. Expression of muscle RING-finger 1 (MuRF1), a ubiquitin ligase, is markedly induced during atrophy, and MuRF1 deletion attenuates muscle wasting. We generated mice expressing a Ring-deletion mutant MuRF1, which binds but cannot ubiquitylate substrates. Mass spectrometry of the bound proteins in denervated muscle identified many myofibrillar components. Upon denervation or fasting, atrophying muscles show a loss of myosin-binding protein C (MyBP-C) and myosin light chains 1 and 2 (MyLC1 and MyLC2) from the myofibril, before any measurable decrease in myosin heavy chain (MyHC). Their selective loss requires MuRF1. MyHC is protected from ubiquitylation in myofibrils by associated proteins, but eventually undergoes MuRF1-dependent degradation. In contrast, MuRF1 ubiquitylates MyBP-C, MyLC1, and MyLC2, even in myofibrils. Because these proteins stabilize the thick filament, their selective ubiquitylation may facilitate thick filament disassembly. However, the thin filament components decreased by a mechanism not requiring MuRF1.


Assuntos
Proteínas Musculares/metabolismo , Músculo Esquelético , Atrofia Muscular/metabolismo , Miofibrilas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Actomiosina/metabolismo , Animais , Proteínas de Transporte/metabolismo , Denervação , Jejum/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Musculares/genética , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/patologia , Miofibrilas/química , Cadeias Leves de Miosina/metabolismo , Ligação Proteica , Proteínas com Motivo Tripartido , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
20.
Leuk Res ; 29(8): 923-31, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15978943

RESUMO

The JAK-STAT signal transduction cascade participates in various cellular processes, including immune response, cell replication, differentiation and oncogenesis. Here, we report that this cascade is induced in two human myeloid HL-60 leukemia cell variants by the granulocyte differentiation inducer dimethyl sulfoxide (DMSO) and macrophage differentiation inducer phorbol 12-myristate 13-acetate (PMA). DMSO and PMA also induced the expression and catalytic activity of 2'-5' oligoadenylate synthetase (2-5A synthetase), a known interferon (IFN) inducible enzyme. The HL-60 cell variants included HL-205, which is susceptible to DMSO- and PMA-induced differentiation, and HL-525, which is susceptible to DMSO- but not to PMA-induced differentiation. Treatment of HL-205 and HL-525 cells with DMSO and HL-205 cells with PMA-induced JAK1 phosphorylation, JAK1/STAT1 association, formation of STAT1-STAT2 heterodimers, and the binding of the active IFN stimulating growth factor 3 (ISGF3) to the IFN-stimulated response element (ISRE) fragment isolated from the 2-5A synthetase promoter. These events were either reduced or absent in the resistant HL-525 cells treated with PMA. Taken together, our data implicate the above signaling cascade in DMSO- and PMA-induced 2-5A synthetase expression and catalytic activity in the HL-60 cell system.


Assuntos
2',5'-Oligoadenilato Sintetase/genética , Proteínas de Ligação a DNA/metabolismo , Dimetil Sulfóxido/farmacologia , Leucemia Mieloide/enzimologia , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais/fisiologia , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacologia , Transativadores/metabolismo , 2',5'-Oligoadenilato Sintetase/efeitos dos fármacos , 2',5'-Oligoadenilato Sintetase/metabolismo , Catálise , Proteínas de Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21 , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Regulação Enzimológica da Expressão Gênica , Genes bcl-1/efeitos dos fármacos , Genes bcl-1/genética , Células HL-60 , Humanos , Janus Quinase 1 , Leucemia Mieloide/metabolismo , Fator de Transcrição STAT1 , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...