Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Hum Mol Genet ; 32(9): 1466-1482, 2023 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-36519761

RESUMO

Abnormal lipid homeostasis has been observed in the brain of Parkinson's disease (PD) patients and experimental models, although the mechanism underlying this phenomenon is unclear. Notably, previous studies have reported that the PD-linked protein Parkin functionally interacts with important lipid regulators, including Sterol Regulatory Element-Binding Proteins (SREBPs) and cluster of differentiation 36 (CD36). Here, we demonstrate a functional relationship between Parkin and lipoprotein lipase (LPL), a triglyceride lipase that is widely expressed in the brain. Using a human neuroblastoma cell line and a Parkin knockout mouse model, we demonstrate that Parkin expression level positively correlates with neuronal LPL protein level and activity. Importantly, our study identified SREBP2, a major regulator of sterol and fatty acid synthesis, as a potential mediator between Parkin and LPL. Supporting this, SREBP2 genetic ablation abolished Parkin effect on LPL expression. We further demonstrate that Parkin-LPL pathway regulates the formation of intracellular lipid droplets, and that this pathway is upregulated upon exposure to PD-linked oxidative stress induced by rotenone. Finally, we show that inhibition of either LPL or SREBP2 exacerbates rotenone-induced cell death. Taken together, our findings reveal a novel pathway linking Parkin, SREBP2 and LPL in neuronal lipid homeostasis that may be relevant to the pathogenesis of PD.


Assuntos
Lipase Lipoproteica , Doença de Parkinson , Proteína de Ligação a Elemento Regulador de Esterol 2 , Ubiquitina-Proteína Ligases , Animais , Humanos , Camundongos , Homeostase , Metabolismo dos Lipídeos/genética , Metabolismo dos Lipídeos/fisiologia , Lipase Lipoproteica/genética , Lipase Lipoproteica/metabolismo , Camundongos Knockout , Neurônios/metabolismo , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Rotenona/efeitos adversos , Transdução de Sinais , Proteína de Ligação a Elemento Regulador de Esterol 2/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
2.
EMBO Rep ; 23(12): e55191, 2022 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-36256516

RESUMO

Autophagy has emerged as the prime machinery for implementing organelle quality control. In the context of mitophagy, the ubiquitin E3 ligase Parkin tags impaired mitochondria with ubiquitin to activate autophagic degradation. Although ubiquitination is essential for mitophagy, it is unclear how ubiquitinated mitochondria activate autophagosome assembly locally to ensure efficient destruction. Here, we report that Parkin activates lipid remodeling on mitochondria targeted for autophagic destruction. Mitochondrial Parkin induces the production of phosphatidic acid (PA) and its subsequent conversion to diacylglycerol (DAG) by recruiting phospholipase D2 and activating the PA phosphatase, Lipin-1. The production of DAG requires mitochondrial ubiquitination and ubiquitin-binding autophagy receptors, NDP52 and optineurin (OPTN). Autophagic receptors, via Golgi-derived vesicles, deliver an autophagic activator, EndoB1, to ubiquitinated mitochondria. Inhibition of Lipin-1, NDP52/OPTN, or EndoB1 results in a failure to produce mitochondrial DAG, autophagosomes, and mitochondrial clearance, while exogenous cell-permeable DAG can induce autophagosome production. Thus, mitochondrial DAG production acts downstream of Parkin to enable the local assembly of autophagosomes for the efficient disposal of ubiquitinated mitochondria.


Assuntos
Ubiquitina-Proteína Ligases , Ubiquitina , Ubiquitina-Proteína Ligases/genética , Lipídeos
3.
Hepatology ; 71(6): 1967-1987, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31539182

RESUMO

BACKGROUND AND AIMS: Hepatocellular carcinoma (HCC) is often accompanied by resistance to immunotherapies despite the presence of tumor-infiltrating lymphocytes. We report that histone deacetylase 6 (HDAC6) represses interleukin-17 (IL-17)-producing helper T (TH 17) cell pathogenicity and the antitumor immune response, dependent on its deacetylase activity. APPROACH AND RESULTS: Adoptive transfer of HDAC6-deficient TH 17 cells impedes HCC growth, dependent on elevated IL-17A, by enhancing the production of antitumor cytokine and cluster of differentiation 8-positive (CD8+) T cell-mediated antitumor responses. Intriguingly, HDAC6-depleted T cells trigger programmed cell death protein 1 (PD-1)-PD-1 ligand 1 expression to achieve a strong synergistic effect to sensitize advanced HCC to an immune checkpoint blocker, while blockade of IL-17A partially suppresses it. Mechanistically, HDAC6 limits TH 17 pathogenicity and the antitumor effect through regulating forkhead box protein O1 (FoxO1). HDAC6 binds and deacetylates cytosolic FoxO1 at K242, which is required for its nuclear translocation and stabilization to repress retinoic acid-related orphan receptor gamma (RoRγt), the transcription factor of TH 17 cell. This regulation of HDAC6 for murine and human TH 17 cell is highly conserved. CONCLUSIONS: These results demonstrate that targeting the cytosolic HDAC6-FoxO1 axis reprograms the pathogenicity and antitumor response of TH 17 cells in HCC, with a pathogenicity-driven responsiveness to facilitate immunotherapies.


Assuntos
Carcinoma Hepatocelular , Desacetilase 6 de Histona/imunologia , Interleucina-17/imunologia , Neoplasias Hepáticas , Animais , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/terapia , Linhagem Celular , Reprogramação Celular/efeitos dos fármacos , Reprogramação Celular/imunologia , Proteína Forkhead Box O1/farmacologia , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Imunoterapia/métodos , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/terapia , Camundongos , Receptor de Morte Celular Programada 1/imunologia , Receptores do Ácido Retinoico/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Receptor gama de Ácido Retinoico
4.
Mol Cell ; 51(6): 819-28, 2013 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-24035499

RESUMO

Aberrant protein aggregation is a dominant pathological feature in neurodegenerative diseases. Protein aggregates cannot be processed by the proteasome; instead, they are frequently concentrated to the aggresome, a perinuclear inclusion body, and subsequently removed by autophagy. Paradoxically, proteasomes are also concentrated at aggresomes and other related inclusion bodies prevalent in neurodegenerative disease. Here, we show that proteasomes are crucial components in aggresome clearance. The disassembly and disposal of aggresomes requires Poh1, a proteasomal deubiquitinating enzyme that cleaves ubiquitinated proteins and releases ubiquitin chains. In Poh1-deficient cells, aggresome clearance is blocked. Remarkably, microinjection of free lysine (K) 63-linked ubiquitin chains restores aggresome degradation. We present evidence that free ubiquitin chains produced by Poh1 bind and activate the deacetylase HDAC6, which, in turn, stimulates actinomyosin- and autophagy-dependent aggresome processing. Thus, unanchored ubiquitin chains are key signaling molecules that connect and coordinate the proteasome and autophagy to eliminate toxic protein aggregates.


Assuntos
Histona Desacetilases/metabolismo , Doenças Neurodegenerativas/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Autofagia , Histona Desacetilases/genética , Humanos , Corpos de Inclusão/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Complexo de Endopeptidases do Proteassoma/genética , Dobramento de Proteína , Proteólise , Ubiquitina/genética , Ubiquitina-Proteína Ligases/metabolismo
5.
EMBO J ; 35(4): 429-42, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26746851

RESUMO

RIG-I is a key cytosolic sensor that detects RNA viruses through its C-terminal region and activates the production of antiviral interferons (IFNs) and proinflammatory cytokines. While posttranslational modification has been demonstrated to regulate RIG-I signaling activity, its significance for the sensing of viral RNAs remains unclear. Here, we first show that the RIG-I C-terminal region undergoes deacetylation to regulate its viral RNA-sensing activity and that the HDAC6-mediated deacetylation of RIG-I is critical for viral RNA detection. HDAC6 transiently bound to RIG-I and removed the lysine 909 acetylation in the presence of viral RNAs, promoting RIG-I sensing of viral RNAs. Depletion of HDAC6 expression led to impaired antiviral responses against RNA viruses, but not against DNA viruses. Consequently, HDAC6 knockout mice were highly susceptible to RNA virus infections compared to wild-type mice. These findings underscore the critical role of HDAC6 in the modulation of the RIG-I-mediated antiviral sensing pathway.


Assuntos
RNA Helicases DEAD-box/metabolismo , Histona Desacetilases/metabolismo , Processamento de Proteína Pós-Traducional , RNA Viral/imunologia , RNA Viral/metabolismo , Animais , Linhagem Celular , Proteína DEAD-box 58 , Modelos Animais de Doenças , Predisposição Genética para Doença , Desacetilase 6 de Histona , Histona Desacetilases/deficiência , Humanos , Camundongos Knockout , Infecções por Vírus de RNA/imunologia , Receptores Imunológicos
6.
Mol Cell ; 47(1): 122-32, 2012 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-22658415

RESUMO

Prolonged deficits in neural input activate pathological muscle remodeling, leading to atrophy. In denervated muscle, activation of the atrophy program requires HDAC4, a potent repressor of the master muscle transcription factor MEF2. However, the signaling mechanism that connects HDAC4, a protein deacetylase, to the atrophy machinery remains unknown. Here, we identify the AP1 transcription factor as a critical target of HDAC4 in neurogenic muscle atrophy. In denervated muscle, HDAC4 activates AP1-dependent transcription, whereas AP1 inactivation recapitulates HDAC4 deficiency and blunts the muscle atrophy program. We show that HDAC4 activates AP1 independently of its canonical transcriptional repressor activity. Surprisingly, HDAC4 stimulates AP1 activity by activating the MAP kinase cascade. We present evidence that HDAC4 binds and promotes the deacetylation and activation of a key MAP3 kinase, MEKK2. Our findings establish an HDAC4-MAPK-AP1 signaling axis essential for neurogenic muscle atrophy and uncover a direct crosstalk between acetylation- and phosphorylation-dependent signaling cascades.


Assuntos
Histona Desacetilases/metabolismo , MAP Quinase Quinase Quinase 2/metabolismo , Músculo Esquelético/metabolismo , Fator de Transcrição AP-1/metabolismo , Acetilação , Animais , Western Blotting , Linhagem Celular , Células HEK293 , Histona Desacetilases/genética , Humanos , MAP Quinase Quinase Quinase 2/genética , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Denervação Muscular , Músculo Esquelético/inervação , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Fosforilação , Ligação Proteica , Interferência de RNA , Fator de Transcrição AP-1/genética
7.
J Cell Sci ; 129(7): 1305-1311, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26869226

RESUMO

HDAC6 is a tubulin deacetylase involved in many cellular functions related to cytoskeleton dynamics, including cell migration and autophagy. In addition, HDAC6 affects antigen-dependent CD4(+)T cell activation. In this study, we show that HDAC6 contributes to the cytotoxic function of CD8(+)T cells. Immunization studies revealed defective cytotoxic activity in vivo in the absence of HDAC6. Adoptive transfer of wild-type or Hdac6(-/-)CD8(+)T cells to Rag1(-/-)mice demonstrated specific impairment in CD8(+)T cell responses against vaccinia infection. Mechanistically, HDAC6-deficient cytotoxic T lymphocytes (CTLs) showed defective in vitro cytolytic activity related to altered dynamics of lytic granules, inhibited kinesin-1-dynactin-mediated terminal transport of lytic granules to the immune synapse and deficient exocytosis, but not to target cell recognition, T cell receptor (TCR) activation or interferon (IFN)γ production. Our results establish HDAC6 as an effector of the immune cytotoxic response that acts by affecting the dynamics, transport and secretion of lytic granules by CTLs.


Assuntos
Grânulos Citoplasmáticos/metabolismo , Citotoxicidade Imunológica/imunologia , Histona Desacetilases/metabolismo , Linfócitos T Citotóxicos/imunologia , Vacínia/imunologia , Animais , Transporte Biológico/fisiologia , Células Cultivadas , Citotoxicidade Imunológica/genética , Complexo Dinactina/antagonistas & inibidores , Desacetilase 6 de Histona , Histona Desacetilases/genética , Interferon gama/metabolismo , Cinesinas/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
8.
J Biol Chem ; 291(10): 5396-405, 2016 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-26763233

RESUMO

The epithelial-to-mesenchymal transition (EMT) is a process by which differentiated epithelial cells reprogram gene expression, lose their junctions and polarity, reorganize their cytoskeleton, increase cell motility and assume a mesenchymal morphology. Despite the critical functions of the microtubule (MT) in cytoskeletal organization, how it participates in EMT induction and maintenance remains poorly understood. Here we report that acetylated α-tubulin, which plays an important role in microtubule (MT) stabilization and cell morphology, can serve as a novel regulator and marker of EMT. A high level of acetylated α-tubulin was correlated with epithelial morphology and it profoundly decreased during TGF-ß-induced EMT. We found that TGF-ß increased the activity of HDAC6, a major deacetylase of α-tubulin, without affecting its expression levels. Treatment with HDAC6 inhibitor tubacin or TGF-ß type I receptor inhibitor SB431542 restored the level of acetylated α-tubulin and consequently blocked EMT. Our results demonstrate that acetylated α-tubulin can serve as a marker of EMT and that HDAC6 represents an important regulator during EMT process.


Assuntos
Transição Epitelial-Mesenquimal , Histona Desacetilases/metabolismo , Processamento de Proteína Pós-Traducional , Tubulina (Proteína)/metabolismo , Acetilação , Anilidas/farmacologia , Animais , Benzamidas/farmacologia , Dioxóis/farmacologia , Células HEK293 , Desacetilase 6 de Histona , Histona Desacetilases/genética , Humanos , Ácidos Hidroxâmicos/farmacologia , Células MCF-7 , Camundongos , Microtúbulos/metabolismo , Fator de Crescimento Transformador beta/farmacologia
9.
J Biol Chem ; 290(15): 9455-64, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25713068

RESUMO

Efficient elimination of misfolded proteins by the proteasome system is critical for proteostasis. Inadequate proteasome capacity can lead to aberrant aggregation of misfolded proteins and inclusion body formation, a hallmark of neurodegenerative disease. The proteasome system cannot degrade aggregated proteins; however, it stimulates autophagy-dependent aggregate clearance by producing unanchored lysine (K)63-linked ubiquitin chains via the proteasomal deubiquitinating enzyme Poh1. The canonical function of Poh1, which removes ubiquitin chains en bloc from proteasomal substrates prior to their degradation, requires intact 26S proteasomes. Here we present evidence that during aggresome clearance, 20S proteasomes dissociate from protein aggregates, while Poh1 and selective subunits of 19S proteasomes are retained. The dissociation of 20S proteasome components requires the molecular chaperone Hsp90. Hsp90 inhibition suppresses 26S proteasome remodeling, unanchored ubiquitin chain production, and aggresome clearance. Our results suggest that 26S proteasomes undergo active remodeling to generate a Poh1-dependent K63-deubiquitinating enzyme to facilitate protein aggregate clearance.


Assuntos
Proteínas de Choque Térmico HSP90/metabolismo , Lisina/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Benzoquinonas/farmacologia , Western Blotting , Linhagem Celular Tumoral , Inibidores de Cisteína Proteinase/farmacologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Desacetilase 6 de Histona , Histona Desacetilases/metabolismo , Humanos , Lactamas Macrocíclicas/farmacologia , Leupeptinas/farmacologia , Microscopia Confocal , Complexo de Endopeptidases do Proteassoma/genética , Agregados Proteicos/efeitos dos fármacos , Interferência de RNA , Transativadores/genética , Transativadores/metabolismo , Ubiquitinação/efeitos dos fármacos
10.
J Biol Chem ; 290(22): 13862-74, 2015 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-25861987

RESUMO

Mutations in PARKIN (PARK2), an ubiquitin ligase, cause early onset Parkinson disease. Parkin was shown to bind, ubiquitinate, and target depolarized mitochondria for destruction by autophagy. This process, mitophagy, is considered crucial for maintaining mitochondrial integrity and suppressing Parkinsonism. Here, we report that under moderate mitochondrial stress, parkin does not translocate to mitochondria to induce mitophagy; rather, it stimulates mitochondrial connectivity. Mitochondrial stress-induced fusion requires PINK1 (PARK6), mitofusins, and parkin ubiquitin ligase activity. Upon exposure to mitochondrial toxins, parkin binds α-synuclein (PARK1), and in conjunction with the ubiquitin-conjugating enzyme Ubc13, stimulates K63-linked ubiquitination. Importantly, α-synuclein inactivation phenocopies parkin overexpression and suppresses stress-induced mitochondria fission, whereas Ubc13 inactivation abrogates parkin-dependent mitochondrial fusion. The convergence of parkin, PINK1, and α-synuclein on mitochondrial dynamics uncovers a common function of these PARK genes in the mitochondrial stress response and provides a potential physiological basis for the prevalence of α-synuclein pathology in Parkinson disease.


Assuntos
Regulação da Expressão Gênica , Mitocôndrias/metabolismo , Proteínas Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , alfa-Sinucleína/metabolismo , Animais , Carbonil Cianeto m-Clorofenil Hidrazona/química , Feminino , Fibroblastos/metabolismo , Inativação Gênica , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Knockout , Microscopia Confocal , Mitofagia , Mutação , Neurônios/metabolismo , Doença de Parkinson/metabolismo , Fosforilação , Ubiquitina/química
11.
J Cell Sci ; 127(Pt 22): 4954-63, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25271058

RESUMO

Fasting and glucose shortage activate a metabolic switch that shifts more energy production to mitochondria. This metabolic adaptation ensures energy supply, but also elevates the risk of mitochondrial oxidative damage. Here, we present evidence that metabolically challenged mitochondria undergo active fusion to suppress oxidative stress. In response to glucose starvation, mitofusin 1 (MFN1) becomes associated with the protein deacetylase HDAC6. This interaction leads to MFN1 deacetylation and activation, promoting mitochondrial fusion. Deficiency in HDAC6 or MFN1 prevents mitochondrial fusion induced by glucose deprivation. Unexpectedly, failure to undergo fusion does not acutely affect mitochondrial adaptive energy production; instead, it causes excessive production of mitochondrial reactive oxygen species and oxidative damage, a defect suppressed by an acetylation-resistant MFN1 mutant. In mice subjected to fasting, skeletal muscle mitochondria undergo dramatic fusion. Remarkably, fasting-induced mitochondrial fusion is abrogated in HDAC6-knockout mice, resulting in extensive mitochondrial degeneration. These findings show that adaptive mitochondrial fusion protects metabolically challenged mitochondria.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Acetilação , Animais , GTP Fosfo-Hidrolases/genética , Desacetilase 6 de Histona , Histona Desacetilases/deficiência , Histona Desacetilases/metabolismo , Camundongos , Dinâmica Mitocondrial , Estresse Oxidativo
12.
EMBO Rep ; 15(11): 1175-83, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25205686

RESUMO

During muscle regeneration, the transcription factor Pax7 stimulates the differentiation of satellite cells (SCs) toward the muscle lineage but restricts adipogenesis. Here, we identify HDAC4 as a regulator of Pax7-dependent muscle regeneration. In HDAC4-deficient SCs, the expression of Pax7 and its target genes is reduced. We identify HDAC4-regulated Lix1 as a Pax7 target gene required for SC proliferation. HDAC4 inactivation leads to defective SC proliferation, muscle regeneration, and aberrant lipid accumulation. Further, expression of the brown adipose master regulator Prdm16 and its inhibitory microRNA-133 are also deregulated. Thus, HDAC4 is a novel regulator of Pax7-dependent SC proliferation and potentially fate determination in regenerating muscle.


Assuntos
Histona Desacetilases/metabolismo , Músculo Esquelético/fisiologia , Fator de Transcrição PAX7/metabolismo , Regeneração , Células Satélites de Músculo Esquelético/metabolismo , Animais , Proteínas Relacionadas à Autofagia , Proliferação de Células , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Histona Desacetilases/genética , Metabolismo dos Lipídeos , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Fator de Transcrição PAX7/genética , Proteínas/genética , Proteínas/metabolismo , Células Satélites de Músculo Esquelético/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
13.
EMBO J ; 30(20): 4142-56, 2011 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-21847094

RESUMO

Histone deacetylases (HDACs) deacetylate histones and non-histone proteins, thereby affecting protein activity and gene expression. The regulation and function of the cytoplasmic class IIb HDAC6 in endothelial cells (ECs) is largely unexplored. Here, we demonstrate that HDAC6 is upregulated by hypoxia and is essential for angiogenesis. Silencing of HDAC6 in ECs decreases sprouting and migration in vitro and formation of functional vascular networks in matrigel plugs in vivo. HDAC6 regulates zebrafish vessel formation, and HDAC6-deficient mice showed a reduced formation of perfused vessels in matrigel plugs. Consistently, overexpression of wild-type HDAC6 increases sprouting from spheroids. HDAC6 function requires the catalytic activity but is independent of ubiquitin binding and deacetylation of α-tubulin. Instead, we found that HDAC6 interacts with and deacetylates the actin-remodelling protein cortactin in ECs, which is essential for zebrafish vessel formation and which mediates the angiogenic effect of HDAC6. In summary, we show that HDAC6 is necessary for angiogenesis in vivo and in vitro, involving the interaction and deacetylation of cortactin that regulates EC migration and sprouting.


Assuntos
Movimento Celular , Cortactina/metabolismo , Histona Desacetilases/metabolismo , Neovascularização Fisiológica , Tubulina (Proteína)/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Acetilação , Animais , Carcinoma Pulmonar de Lewis/enzimologia , Células Cultivadas , Células Endoteliais/metabolismo , Feminino , Desacetilase 6 de Histona , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Camundongos Knockout
14.
Biochem Biophys Res Commun ; 464(4): 1235-1240, 2015 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-26210454

RESUMO

Mitochondria undergo fusion and fission in response to various metabolic stresses. Growing evidences have suggested that the morphological change of mitochondria by fusion and fission plays a critical role in protecting mitochondria from metabolic stresses. Here, we showed that hypoxia treatment could induce interaction between HDAC6 and MFN2, thus protecting mitochondrial connectivity. Mechanistically, we demonstrated that a mitochondrial ubiquitin ligase MARCH5/MITOL was responsible for hypoxia-induced MFN2 degradation in HDAC6 deficient cells. Notably, genetic abolition of HDAC6 in amyotrophic lateral sclerosis model mice showed MFN2 degradation with MARCH5 induction. Our results indicate that HDAC6 is a critical regulator of MFN2 degradation by MARCH5, thus protecting mitochondrial connectivity from hypoxic stress.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Histona Desacetilases/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Estresse Fisiológico/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Hipóxia Celular/fisiologia , Regulação para Baixo , Feminino , Desacetilase 6 de Histona , Humanos , Proteínas de Membrana , Camundongos , Mitocôndrias/ultraestrutura , Oxigênio/metabolismo
15.
Neurodegener Dis ; 15(6): 339-49, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26360702

RESUMO

Aberrant accumulation of protein aggregates is a pathological hallmark of many neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Although a buildup of protein aggregates frequently leads to cell death, whether it is the key pathogenic factor in driving neurodegenerative disease remains controversial. HDAC6, a cytosolic ubiquitin-binding deacetylase, has emerged as an important regulator of ubiquitin-dependent quality control autophagy, a lysosome-dependent degradative system responsible for the disposal of misfolded protein aggregates and damaged organelles. Here, we show that in cell models HDAC6 plays a protective role against multiple disease-associated and aggregation-prone cytosolic proteins by facilitating their degradation. We further show that HDAC6 is required for efficient localization of lysosomes to protein aggregates, indicating that lysosome targeting to autophagic substrates is regulated. Supporting a critical role of HDAC6 in protein aggregate disposal in vivo, genetic ablation of HDAC6 in a transgenic SOD1G93A mouse, a model of ALS, leads to dramatic accumulation of ubiquitinated SOD1G93A protein aggregates. Surprisingly, despite a robust buildup of SOD1G93A aggregates, deletion of HDAC6 only moderately modified the motor phenotypes. These findings indicate that SOD1G93A aggregation is not the only determining factor to drive neurodegeneration in ALS, and that HDAC6 likely modulates neurodegeneration through additional mechanisms beyond protein aggregate clearance.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Agregação Patológica de Proteínas/metabolismo , Ubiquitina/metabolismo , Esclerose Lateral Amiotrófica/genética , Animais , Autofagia/genética , Modelos Animais de Doenças , Histona Desacetilases/metabolismo , Lisossomos/metabolismo , Camundongos Transgênicos
16.
EMBO J ; 29(5): 969-80, 2010 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-20075865

RESUMO

Autophagy is primarily considered a non-selective degradation process induced by starvation. Nutrient-independent basal autophagy, in contrast, imposes intracellular QC by selective disposal of aberrant protein aggregates and damaged organelles, a process critical for suppressing neurodegenerative diseases. The molecular mechanism that distinguishes these two fundamental autophagic responses, however, remains mysterious. Here, we identify the ubiquitin-binding deacetylase, histone deacetylase-6 (HDAC6), as a central component of basal autophagy that targets protein aggregates and damaged mitochondria. Surprisingly, HDAC6 is not required for autophagy activation; rather, it controls the fusion of autophagosomes to lysosomes. HDAC6 promotes autophagy by recruiting a cortactin-dependent, actin-remodelling machinery, which in turn assembles an F-actin network that stimulates autophagosome-lysosome fusion and substrate degradation. Indeed, HDAC6 deficiency leads to autophagosome maturation failure, protein aggregate build-up, and neurodegeneration. Remarkably, HDAC6 and F-actin assembly are completely dispensable for starvation-induced autophagy, uncovering the fundamental difference of these autophagic modes. Our study identifies HDAC6 and the actin cytoskeleton as critical components that define QC autophagy and uncovers a novel regulation of autophagy at the level of autophagosome-lysosome fusion.


Assuntos
Autofagia/fisiologia , Histona Desacetilases/metabolismo , Fagossomos/metabolismo , Ubiquitina/metabolismo , Actinas/metabolismo , Animais , Animais Geneticamente Modificados , Autofagia/genética , Linhagem Celular , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Desacetilase 6 de Histona , Histona Desacetilases/genética , Imuno-Histoquímica , Lisossomos/genética , Lisossomos/metabolismo , Camundongos , Microscopia de Fluorescência , Fagossomos/genética
17.
Adv Sci (Weinh) ; 11(2): e2303489, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37964763

RESUMO

The essential branched-chain amino acids (BCAAs) leucine, isoleucine, and valine play critical roles in protein synthesis and energy metabolism. Despite their widespread use as nutritional supplements, BCAAs' full effects on mammalian physiology remain uncertain due to the complexities of BCAA metabolic regulation. Here a novel mechanism linking intrinsic alterations in BCAA metabolism is identified to cellular senescence and the senescence-associated secretory phenotype (SASP), both of which contribute to organismal aging and inflammation-related diseases. Altered BCAA metabolism driving the SASP is mediated by robust activation of the BCAA transporters Solute Carrier Family 6 Members 14 and 15 as well as downregulation of the catabolic enzyme BCAA transaminase 1 during onset of cellular senescence, leading to highly elevated intracellular BCAA levels in senescent cells. This, in turn, activates the mammalian target of rapamycin complex 1 (mTORC1) to establish the full SASP program. Transgenic Drosophila models further indicate that orthologous BCAA regulators are involved in the induction of cellular senescence and age-related phenotypes in flies, suggesting evolutionary conservation of this metabolic pathway during aging. Finally, experimentally blocking BCAA accumulation attenuates the inflammatory response in a mouse senescence model, highlighting the therapeutic potential of modulating BCAA metabolism for the treatment of age-related and inflammatory diseases.


Assuntos
Aminoácidos de Cadeia Ramificada , Fenótipo Secretor Associado à Senescência , Animais , Camundongos , Aminoácidos de Cadeia Ramificada/metabolismo , Leucina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Metabolismo Energético , Mamíferos/metabolismo
18.
Nature ; 447(7146): 859-63, 2007 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-17568747

RESUMO

A prominent feature of late-onset neurodegenerative diseases is accumulation of misfolded protein in vulnerable neurons. When levels of misfolded protein overwhelm degradative pathways, the result is cellular toxicity and neurodegeneration. Cellular mechanisms for degrading misfolded protein include the ubiquitin-proteasome system (UPS), the main non-lysosomal degradative pathway for ubiquitinated proteins, and autophagy, a lysosome-mediated degradative pathway. The UPS and autophagy have long been viewed as complementary degradation systems with no point of intersection. This view has been challenged by two observations suggesting an apparent interaction: impairment of the UPS induces autophagy in vitro, and conditional knockout of autophagy in the mouse brain leads to neurodegeneration with ubiquitin-positive pathology. It is not known whether autophagy is strictly a parallel degradation system, or whether it is a compensatory degradation system when the UPS is impaired; furthermore, if there is a compensatory interaction between these systems, the molecular link is not known. Here we show that autophagy acts as a compensatory degradation system when the UPS is impaired in Drosophila melanogaster, and that histone deacetylase 6 (HDAC6), a microtubule-associated deacetylase that interacts with polyubiquitinated proteins, is an essential mechanistic link in this compensatory interaction. We found that compensatory autophagy was induced in response to mutations affecting the proteasome and in response to UPS impairment in a fly model of the neurodegenerative disease spinobulbar muscular atrophy. Autophagy compensated for impaired UPS function in an HDAC6-dependent manner. Furthermore, expression of HDAC6 was sufficient to rescue degeneration associated with UPS dysfunction in vivo in an autophagy-dependent manner. This study suggests that impairment of autophagy (for example, associated with ageing or genetic variation) might predispose to neurodegeneration. Morover, these findings suggest that it may be possible to intervene in neurodegeneration by augmenting HDAC6 to enhance autophagy.


Assuntos
Autofagia/fisiologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Histona Desacetilases/metabolismo , Doenças Neurodegenerativas/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Animais , Autofagia/genética , Modelos Animais de Doenças , Drosophila melanogaster/genética , Desacetilase 6 de Histona , Humanos , Transtornos Musculares Atróficos/genética , Transtornos Musculares Atróficos/metabolismo , Doenças Neurodegenerativas/genética , Peptídeos/genética , Peptídeos/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo
19.
J Med Chem ; 66(14): 10080-10091, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37463038

RESUMO

In our previous research, a series of phenylsulfonylfuroxan-based hydroxamates were developed, among which compound 1 exhibited remarkable in vitro and in vivo antitumor potency due to its histone deacetylase (HDAC) inhibitory and nitric oxide (NO)-donating activities. Herein, the in-depth study of compound 1 revealed that this HDAC inhibitor-NO donor hybrid could enduringly increase the intracellular levels of acetyl histones and acetyl α-tubulin, which could be ascribed to its irreversible inhibition toward class I HDACs and HDAC6. Structural modification of compound 1 led to a novel phenylsulfonylfuroxan-based hydroxamate 4, which exhibited considerable HDAC6 inhibitory activity and selectivity. Furthermore, compound 4 could inhibit intracellular HDAC6 both selectively and irreversibly. To the best of our knowledge, this is the first research reporting the irreversible inhibition of HDAC6. It was also demonstrated that compared with ACY-241 (a reversible HDAC6 inhibitor in clinical trials), the irreversible HDAC6 selective inhibitor 4 exhibited not only superior anti-multiple myeloma activity but also improved therapeutic index.


Assuntos
Mieloma Múltiplo , Humanos , Mieloma Múltiplo/tratamento farmacológico , Desacetilase 6 de Histona , Histona Desacetilases/metabolismo , Histonas , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Inibidores de Histona Desacetilases/química , Isoformas de Proteínas , Ácidos Hidroxâmicos/farmacologia , Ácidos Hidroxâmicos/química
20.
J Cachexia Sarcopenia Muscle ; 13(6): 3091-3105, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36059045

RESUMO

BACKGROUND: Cathelicidin, an antimicrobial peptide, plays a key role in regulating bacterial killing and innate immunity; however, its role in skeletal muscle function is unknown. We investigated the potential role of cathelicidin in skeletal muscle pathology resulting from acute injury and Duchenne muscular dystrophy (DMD) in mice. METHODS: Expression changes and muscular localization of mouse cathelicidin-related antimicrobial peptide (Cramp) were examined in the skeletal muscle of normal mice treated with chemicals (cardiotoxin and BaCl2 ) or in dystrophic muscle of DMD mouse models (mdx, mdx/Utrn+/- and mdx/Utrn-/- ). Cramp penetration into myofibres and effects on muscle damage were studied by treating synthetic peptides to mouse skeletal muscles or C2C12 myotubes. Cramp knockout (KO) mice and mdx/Utrn/Cramp KO lines were used to determine whether Cramp mediates muscle degeneration. Muscle pathophysiology was assessed by histological methods, serum analysis, grip strength and lifespan. Molecular factors targeted by Cramp were identified by the pull-down assay and proteomic analysis. RESULTS: In response to acute muscle injury, Cramp was activated in muscle-infiltrating neutrophils and internalized into myofibres. Cramp treatments of mouse skeletal muscles or C2C12 myotubes resulted in muscle degeneration and myotube damage, respectively. Genetic ablation of Cramp reduced neutrophil infiltration and ameliorated muscle pathology, such as fibre size (P < 0.001; n = 6) and fibrofatty infiltration (P < 0.05). Genetic reduction of Cramp in mdx/Utrn+/- mice not only attenuated muscle damage (35%, P < 0.05; n = 9-10), myonecrosis (53%, P < 0.05), inflammation (37-65%, P < 0.01) and fibrosis (14%, P < 0.05) but also restored muscle fibre size (14%, P < 0.05) and muscle force (18%, P < 0.05). Reducing Cramp levels led to a 63% (male, P < 0.05; n = 10-14) and a 124% (female, P < 0.001; n = 20) increase in the lifespan of mdx/Utrn-/- mice. Proteomic and mechanistic studies revealed that Cramp cross-talks with Ca2+ signalling in skeletal muscle through sarcoplasmic/endoplasmic reticulum Ca2+ -ATPase1 (SERCA1). Cramp binds and inactivates SERCA1, leading to the activation of Ca2+ -dependent calpain proteases that exacerbate DMD progression. CONCLUSIONS: These findings identify Cramp as an immune cell-derived regulator of skeletal muscle degeneration and provide a potential therapeutic target for DMD.


Assuntos
Distrofia Muscular de Duchenne , Camundongos , Masculino , Feminino , Animais , Distrofia Muscular de Duchenne/complicações , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Camundongos Endogâmicos mdx , Proteômica , Músculo Esquelético/patologia , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA