Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Gen Comp Endocrinol ; 320: 114009, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35227727

RESUMO

Bone marrow-derived mesenchymal/stromal stem cells (MSCs) became a major focus of research since the anti-inflammatory features and the osteogenic commitment of these cells can prevent the inflamm-aging and various form of osteopenia in humans and animals. We previously showed that p62/SQSTM1 plasmid can prompt release of anti-inflammatory cytokines/chemokines by MSC when injected in adult mice. Furthermore, it can enhance osteoblastogenesis at the expense of adipogenesis and ameliorate bone density and bone remodeling. On the other hand, absence of p62 partially exhausted MSC pool caused expansion of fat cells within bone marrow and pro-inflammatory mediator's accumulation. Given the critical function of p62 as molecular hub of MSC dynamics, here, using MSCs from p62 knockout adult mice, we investigated the effect of this protein on MSC survival and bone-forming molecule cascades. We found that the main osteogenic routes are impaired in absence of p62. In particular, lack of p62 can suppress Smads activation, and Osterix and CREBs expression, thus significantly modifying the schedule of MSCs differentiation. MSCs obtained from p62-/- mice have also demonstrate an amplified NFκB/ Smad1/5/8 colocalization along with NFκB activation in the nucleus, which precludes Smads binding to target promoters. Considering the "teamwork" of TGFß, PTH and BMP2 on MSC homeostatic behavior, we consider that p62 exerts an essential role as a hub protein. Lastly, ex vivo pulsing p62-deficient MSCs, which then will be administered to a patient as a cell therapy, may be considered as a treatment for bone and bone marrow disorders.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Proteína Sequestossoma-1 , Animais , Medula Óssea/metabolismo , Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Knockout , Proteína Sequestossoma-1/genética , Proteína Sequestossoma-1/fisiologia
2.
Int J Oncol ; 59(4)2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34414460

RESUMO

Sequestosome 1 (SQSTM1)/p62 is an adapter protein mainly involved in the transportation, degradation and destruction of various proteins that cooperates with components of autophagy and the ubiquitin­proteasome degradation pathway. Numerous studies have shown that SQSTM1/p62 functions at multiple levels, including involvement in genetic stability or modification, post­transcriptional regulation and protein function. As a result, SQSTM1/p62 is a versatile protein that is a critical core regulator of tumor cell genetic stability, autophagy, apoptosis and other forms of cell death, malignant growth, proliferation, migration, invasion, metastasis and chemoradiotherapeutic response, and an indicator of patient prognosis. SQSTM1/p62 regulates these processes via its distinct molecular structure, through which it participates in a variety of activating or inactivating tumor­related and tumor microenvironment­related signaling pathways, particularly positive feedback loops and epithelial­mesenchymal transition­related pathways. Therefore, functioning as a proto­oncogene or tumor suppressor gene in various types of cancer and tumor­associated microenvironments, SQSTM1/p62 is capable of promoting or retarding malignant tumor aggression, giving rise to immeasurable effects on tumor occurrence and development, and on patient treatment and prognosis.


Assuntos
Neoplasias/etiologia , Proteína Sequestossoma-1/fisiologia , Agressão , Transição Epitelial-Mesenquimal , Humanos , Neoplasias/patologia , Proteína Sequestossoma-1/genética , Microambiente Tumoral
3.
Autophagy ; 17(11): 3763-3775, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33719859

RESUMO

Macroautophagy/autophagy plays a critical role in antiviral immunity through targeting viruses and initiating host immune responses. The receptor protein, SQSTM1/p62 (sequestosome 1), plays a vital role in selective autophagy. It serves as a receptor targeting ubiquitinated proteins or pathogens to phagophores for degradation. In this study, we explored the reciprocal regulation between selective autophagy receptor SQSTM1 and Seneca Valley virus (SVV). SVV infection induced autophagy. Autophagy promoted SVV infection in pig cells but played opposite functions in human cells. Overexpression of SQSTM1 decreased viral protein production and reduced viral titers. Further study showed that SQSTM1 interacted with SVV VP1 and VP3 independent of its UBA domain. SQSTM1 targeted SVV VP1 and VP3 to phagophores for degradation to inhibit viral replication. To counteract this, SVV evolved strategies to circumvent the host autophagic machinery to promote viral replication. SVV 3Cpro targeted the receptor SQSTM1 for cleavage at glutamic acid 355, glutamine 392, and glutamine 395 and abolished its capacity to mediate selective autophagy. At the same time, the 3Cpro-mediated SQSTM1 cleavage products lost the ability to inhibit viral propagation. Collectively, our results provide evidence for selective autophagy in host against viruses and reveal potential viral strategies to evade autophagic machinery for successful pathogenesis.Abbreviations: Baf.A1: bafilomycin A1; Co-IP: co-immunoprecipitation; hpi: h post-infection; LIR: LC3-interacting region; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MOI: multiplicity of infection; PB1: N-terminal Phox/Bem1p; Rap.: rapamycin; Seneca Valley virus: SVV; SQSTM1/p62: sequestosome 1; SQSTM1-N355: residues 1 to 355 of SQSTM1; SQSTM1-C355: residues 355 to 478 of SQSTM1; SQSTM1-N392: residues 1 to 392 of SQSTM1; SQSTM1-C392: residues 392 to 478 of SQSTM1; SQSTM1-N388: residues 1 to 388 of SQSTM1; SQSTM1-N397: residues 1 to 397 of SQSTM1; UBA: ubiquitin association; Ubi: ubiquitin.


Assuntos
Autofagia , Picornaviridae/fisiologia , Proteína Sequestossoma-1/metabolismo , Proteínas Estruturais Virais/metabolismo , Replicação Viral , Animais , Linhagem Celular , Células HEK293 , Humanos , Proteína Sequestossoma-1/fisiologia , Especificidade da Espécie , Suínos , Proteínas Estruturais Virais/fisiologia
4.
Autophagy ; 17(11): 3461-3474, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33509017

RESUMO

Macroautophagy/autophagy plays an important role in the control of viral infections and viruses have evolved multiple strategies to interfere with autophagy to avoid destruction and promote their own replication and spread. Here we report that the deubiquitinase encoded in the N-terminal domain of the Epstein-Barr virus (EBV) large tegument protein, BPLF1, regulates selective autophagy. Mass spectrometry analysis identified several vesicular traffic and autophagy related proteins as BPLF1 interactors and potential substrates, suggesting that the viral protein targets this cellular defense during productive infection. Direct binding of BPLF1 to the autophagy receptor SQSTM1/p62 (sequestosome 1) was confirmed by co-immunoprecipitation of transfected BPLF1 and by in vitro affinity isolation of bacterially expressed proteins. Expression of the catalytically active BPLF1 was associated with decreased SQSTM1/p62 ubiquitination and failure to recruit LC3 to SQSTM1/p62-positive aggregates. Selective autophagy was inhibited as illustrated by the accumulation of large protein aggregates in BPLF1-positive cells co-transfected with an aggregate-prone HTT (huntingtin)-Q109 construct, and by a slower autophagy-dependent clearance of protein aggregates upon transfection of BPLF1 in cells expressing a tetracycline-regulated HTT-Q103. The inhibition of aggregate clearance was restored by overexpression of a SQSTM1/p62[E409A,K420R] mutant that does not require ubiquitination of Lys420 for cargo loading. These findings highlight a previously unrecognized role of the viral deubiquitinase in the regulation of selective autophagy, which may promote infection and the production of infectious virus.Abbreviations: BPLF1, BamH1 fragment left open reading frame-1; EBV, Epstein-Barr virus; GFP, green fluorescent protein; HTT, huntingtin; MAP1LC3/LC3, microtubule associated protein 1 light chain 3; PB1, Phox and Bem1 domain; PE, phosphatidylethanolamine; SQSTM1/p62, sequestosome 1; UBA, ubiquitin-associated domain.


Assuntos
Autofagia/fisiologia , Enzimas Desubiquitinantes/fisiologia , Herpesvirus Humano 4/fisiologia , Proteína Sequestossoma-1/fisiologia , Proteínas Virais Reguladoras e Acessórias/fisiologia , Autofagia/genética , Enzimas Desubiquitinantes/genética , Infecções por Vírus Epstein-Barr/patologia , Infecções por Vírus Epstein-Barr/virologia , Células HeLa , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/patogenicidade , Interações entre Hospedeiro e Microrganismos/genética , Interações entre Hospedeiro e Microrganismos/fisiologia , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Macroautofagia/genética , Macroautofagia/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação , Agregados Proteicos/genética , Agregados Proteicos/fisiologia , Proteína Sequestossoma-1/genética , Transfecção , Ubiquitinação , Proteínas Virais Reguladoras e Acessórias/genética
5.
Hepatology ; 69(3): 1046-1063, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30281815

RESUMO

In hepatocellular carcinoma (HCC), dysregulated expression of DDX5 (DEAD box protein 5) and impaired autophagy have been reported separately. However, the relationship between them has not been explored. Here we present evidence to show that, by interacting with autophagic receptor p62, DDX5 promotes autophagy and suppresses tumorigenesis. DDX5 inversely correlated with p62/sequestosome 1 (SQSTM1) expression in hepatitis B virus (HBV)-associated and non-HBV-associated HCCs. Patients with low DDX5 expression showed poor prognosis after tumor resection. We found that DDX5 overexpression induced, while DDX5 knockdown attenuated, autophagic flux in HepG2 and Huh7 cells. DDX5 promoted p62 degradation and markedly reduced the half-life of p62. Moreover, DDX5 overexpression dramatically reduced, while DDX5 knockdown promoted, cancer cell growth and tumorigenesis in vitro and in vivo. We found that DDX5 bound to p62 and interfered with p62/TRAF6 (tumor necrosis factor receptor-associated factor 6) interaction. Further findings revealed that the N-terminal domain of DDX5, involved in the interaction with p62, was sufficient to induce autophagy independent of its RNA binding and helicase activity. DDX5 overexpression decreased p62/TRAF6-mediated lysine 63-linked ubiquitination of mammalian target of rapamycin (mTOR) and subsequently inhibited the mTOR signaling pathway. Knockdown of TRAF6 blocked DDX5-induced autophagy. Furthermore, we showed that miR-17-5p downregulated DDX5 and impaired autophagy. Inhibition of miR-17-5p promoted autophagic flux and suppressed tumor growth in HCC xenograft models. Conclusion: Our findings define a noncanonical pathway that links miR-17-5p, DDX5, p62/TRAF6, autophagy, and HCC. These findings open an avenue for the treatment of HCC.


Assuntos
Autofagia/fisiologia , Carcinogênese , RNA Helicases DEAD-box/fisiologia , Proteína Sequestossoma-1/fisiologia , Humanos , Neoplasias Hepáticas , Células Tumorais Cultivadas
6.
Blood ; 133(2): 168-179, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30498063

RESUMO

Autophagy maintains hematopoietic stem cell integrity and prevents malignant transformation. In addition to bulk degradation, selective autophagy serves as an intracellular quality control mechanism and requires autophagy receptors, such as p62 (SQSTM1), to specifically bridge the ubiquitinated cargos into autophagosomes. Here, we investigated the function of p62 in acute myeloid leukemia (AML) in vitro and in murine in vivo models of AML. Loss of p62 impaired expansion and colony-forming ability of leukemia cells and prolonged latency of leukemia development in mice. High p62 expression was associated with poor prognosis in human AML. Using quantitative mass spectrometry, we identified enrichment of mitochondrial proteins upon immunoprecipitation of p62. Loss of p62 significantly delayed removal of dysfunctional mitochondria, increased mitochondrial superoxide levels, and impaired mitochondrial respiration. Moreover, we demonstrated that the autophagy-dependent function of p62 is essential for cell growth and effective mitochondrial degradation by mitophagy. Our results highlight the prominent role of selective autophagy in leukemia progression, and specifically, the importance of mitophagy to maintain mitochondrial integrity.


Assuntos
Autofagia , Leucemia Experimental/patologia , Leucemia Mieloide Aguda/patologia , Mitofagia , Proteína Sequestossoma-1/metabolismo , Proteína Sequestossoma-1/fisiologia , Animais , Seguimentos , Humanos , Leucemia Experimental/genética , Leucemia Experimental/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Camundongos , Camundongos Knockout , Prognóstico , Taxa de Sobrevida , Células Tumorais Cultivadas
7.
J Cell Sci ; 131(21)2018 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-30397181

RESUMO

SQSTM1 (also known as p62) is a multifunctional stress-inducible scaffold protein involved in diverse cellular processes. Its functions are tightly regulated through an extensive pattern of post-translational modifications, and include the isolation of cargos degraded by autophagy, induction of the antioxidant response by the Keap1-Nrf2 system, as well as the regulation of endosomal trafficking, apoptosis and inflammation. Accordingly, malfunction of SQSTM1 is associated with a wide range of diseases, including bone and muscle disorders, neurodegenerative and metabolic diseases, and multiple forms of cancer. In this Review, we summarize current knowledge regarding regulation, post-translational modifications and functions of SQSTM1, as well as how they are dysregulated in various pathogenic contexts.


Assuntos
Proteínas de Ligação a RNA/fisiologia , Proteína Sequestossoma-1/fisiologia , Animais , Humanos , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Proteína Sequestossoma-1/genética , Proteína Sequestossoma-1/metabolismo , Transdução de Sinais
8.
Autophagy ; 14(12): 2049-2064, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30215534

RESUMO

Glucocorticoids are widely used to treat inflammatory disorders; however, prolonged use of glucocorticoids results in side effects including osteoporosis, diabetes and obesity. Compound A (CpdA), identified as a selective NR3C1/glucocorticoid receptor (nuclear receptor subfamily 3, group C, member 1) modulator, exhibits an inflammation-suppressive effect, largely in the absence of detrimental side effects. To understand the mechanistic differences between the classic glucocorticoid dexamethasone (DEX) and CpdA, we looked for proteins oppositely regulated in bone marrow-derived macrophages using an unbiased proteomics approach. We found that the autophagy receptor SQSTM1 but not NR3C1 mediates the anti-inflammatory action of CpdA. CpdA drives SQSTM1 upregulation by recruiting the NFE2L2 transcription factor to its promoter. In contrast, the classic NR3C1 ligand dexamethasone recruits NR3C1 to the Sqstm1 promoter and other NFE2L2-controlled gene promoters, resulting in gene downregulation. Both DEX and CpdA induce autophagy, with marked different autophagy characteristics and morphology. Suppression of LPS-induced Il6 and Ccl2 genes by CpdA in macrophages is hampered upon Sqstm1 silencing, confirming that SQSTM1 is essential for the anti-inflammatory capacity of CpdA, at least in this cell type. Together, these results demonstrate how off-target mechanisms of selective NR3C1 ligands may contribute to a more efficient anti-inflammatory therapy.


Assuntos
Acetatos/farmacologia , Anti-Inflamatórios/farmacologia , Inflamação/genética , Inflamação/prevenção & controle , Macrófagos/efeitos dos fármacos , Receptores de Glucocorticoides/agonistas , Proteína Sequestossoma-1/fisiologia , Tiramina/análogos & derivados , Animais , Células Cultivadas , Dexametasona/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Glucocorticoides/metabolismo , Proteína Sequestossoma-1/genética , Ativação Transcricional/efeitos dos fármacos , Tiramina/farmacologia
9.
Yakugaku Zasshi ; 138(8): 1059-1065, 2018.
Artigo em Japonês | MEDLINE | ID: mdl-30068847

RESUMO

In the process of recent hit-to-lead studies, not only in industry but also in academia, early evaluation of metabolic properties has been one of the key aspects supporting a higher probability of success in drug discovery. In this review, we introduce the development of chemical seeds targeting the Kelch-like ECH-associated protein-1 (Keap1) as an example of an academic hit-to-lead study considering metabolic stability. Keap1 regulates the function of nuclear factor erythroid 2-related factor 2 (Nrf2), which induces various antioxidative or detoxification proteins. An inhibitor of protein-protein interaction (PPI) between Keap1 and Nrf2 to activate Nrf2 is expected to be a novel target for drug discovery. However, Nrf2 is also activated in several cancers, such as human hepatocellular carcinoma, and causes chemoresistance, which is mediated by phosphorylated p62/Sqstm1 (p-p62), an autophagy-related protein that also undergoes a PPI with Keap1. In this case, an Nrf2 suppressor could be used to attenuate drug resistance. We discovered inhibitors against the Nrf2-Keap1 PPI and p-p62-Keap1 PPI using high-throughput screening and established the synthetic routes for the hit compounds and their derivatives. Furthermore, we assessed the metabolic stability of both of the PPI inhibitors in human liver microsomes and identified the metabolic sites.


Assuntos
1-Naftilamina/análogos & derivados , Fumarato de Dimetilo , Descoberta de Drogas , Isotiocianatos , Proteína 1 Associada a ECH Semelhante a Kelch , Fator 2 Relacionado a NF-E2 , Ácido Oleanólico/análogos & derivados , Mapas de Interação de Proteínas/efeitos dos fármacos , Sulfonamidas , 1-Naftilamina/química , 1-Naftilamina/farmacologia , Autofagia , Fumarato de Dimetilo/química , Fumarato de Dimetilo/farmacologia , Ensaios de Triagem em Larga Escala , Humanos , Isotiocianatos/química , Isotiocianatos/farmacologia , Proteína 1 Associada a ECH Semelhante a Kelch/fisiologia , Microssomos Hepáticos/metabolismo , Fator 2 Relacionado a NF-E2/fisiologia , Ácido Oleanólico/química , Ácido Oleanólico/farmacologia , Fosforilação , Proteína Sequestossoma-1/fisiologia , Sulfonamidas/química , Sulfonamidas/farmacologia , Sulfóxidos
10.
Br J Pharmacol ; 175(3): 440-455, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29148034

RESUMO

BACKGROUND AND PURPOSE: SQSTM1/p62 is a multifunctional, stress-induced, scaffold protein involved in multiple cellular processes including autophagic clearance, regulation of inflammatory responses and redox homeostasis. Its altered function has been associated with different human pathologies, such as neurodegenerative, metabolic and bone diseases (down-regulation), and cancerogenesis (up-regulation). However, its role in the off-target effects of clinically used drugs is still not understood. EXPERIMENTAL APPROACH: We evaluated the expression of p62 in cultured Hep3B cells and their derived ρ° cells (lacking mitochondria), along with markers of autophagy and mitochondrial dysfunction. The effects of efavirenz were compared with those of known pharmacological stressors, rotenone, thapsigargin and CCCP, and we also used transient silencing with siRNA and p62 overexpression. Western blotting, quantRT-PCR and fluorescence microscopy were used to assay these effects and their underlying mechanisms. KEY RESULTS: In Hep3B cells, efavirenz augmented p62 protein content, an effect not observed in the corresponding ρ° cells. p62 up-regulation followed enhanced SQSTM1 expression mediated through the transcription factor CHOP/DDIT3, while other well-known regulators (NF-kB and Nrf2) were not involved. Inhibition of autophagy with 3MA or with transient silencing of Atg5 did not affect SQSTM1 expression in efavirenz-treated cells while p62 overexpression ameliorated the deleterious effect of efavirenz on cell viability. CONCLUSION AND IMPLICATIONS: In our model, p62 exerted a specific, autophagy-independent role and protected against efavirenz-induced mitochondrial ROS generation and activation of the NLRP3 inflammasome. These findings add to the multifunctional nature of p62 and may help to understand the off-target effects of clinically useful drugs.


Assuntos
Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Benzoxazinas/toxicidade , Proteína Sequestossoma-1/fisiologia , Alcinos , Linhagem Celular Tumoral , Ciclopropanos , Relação Dose-Resposta a Droga , Hepatócitos/efeitos dos fármacos , Hepatócitos/fisiologia , Humanos , Espécies Reativas de Oxigênio/metabolismo , Inibidores da Transcriptase Reversa/toxicidade
11.
Trends Endocrinol Metab ; 28(11): 818-830, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28966079

RESUMO

p62, a protein capable of binding both ubiquitin and autophagy substrates, is well established as a key regulator in cancer and neurodegenerative diseases. Recently, there has been accumulating evidence that p62 is also a pivotal regulator in metabolic diseases, such as obesity, T2DM, NAFLD, metabolic bone disease, gout and thyroid disease. This review summarizes the emerging role of p62 on these diseases by considering its functional domains, phenotypes in genetically modified animals, clinically observed alterations, and its effects on downstream metabolic signaling pathways. At the same time, we highlight the need to explore the roles played by p62 in the gastrointestinal environment and immune system, and the extent to which its elevated expression may confer protection against metabolic disorders.


Assuntos
Doenças Metabólicas/genética , Doenças Metabólicas/metabolismo , Proteína Sequestossoma-1/fisiologia , Animais , Autofagia/genética , Humanos , Camundongos , Camundongos Transgênicos , Transdução de Sinais
12.
Neurobiol Aging ; 60: 104-115, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28941726

RESUMO

Many studies reveal that BAG3 plays a critical role in the regulation of protein degradation via macroautophagy. However, it remains unknown whether BAG3 affects the quality control of α-synuclein (SNCA), a Parkinson's disease-related protein. In this study, we demonstrated the increases of BAG3 expression in the ventral midbrain of SNCAA53T transgenic mice and also in MG132-treated PC12 cells overexpressing wild-type SNCA (SNCAWT-PC12). Moreover, we showed that BAG3 overexpression was sufficient to enhance the autophagy activity while knockdown of Bag3 reduced it in SNCAWT-PC12 cells. Immunoprecipitation revealed that BAG3 interacted with heat shock protein 70 and sequestosome 1. The immunostaining also showed the perinuclear accumulation and colocalization of BAG3 with these 2 proteins, as well as with LC3 dots in tyrosine hydroxylase-positive neurons in the midbrain of SNCAA53T mice. BAG3 overexpression was able to modulate SNCA degradation via macroautophagy which was prevented by Atg5 knockdown. Taken together, these results indicate that BAG3 plays a relevant role in regulating SNCA clearance via macroautophagy, and the heat shock protein 70-BAG3-sequestosome 1 complex may be involved in this process.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas Reguladoras de Apoptose/fisiologia , Autofagia/genética , Autofagia/fisiologia , alfa-Sinucleína/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Expressão Gênica , Proteínas de Choque Térmico HSP70/fisiologia , Masculino , Mesencéfalo/metabolismo , Camundongos Transgênicos , Células PC12 , Ratos , Proteína Sequestossoma-1/fisiologia
13.
PLoS One ; 12(1): e0169291, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28076378

RESUMO

One of the proteins most frequently found in neuropathological lesions is the ubiquitin binding protein p62 (sequestosome 1). Post-mortem analysis of p62 is a defining diagnostic marker in several neurodegenerative diseases including amyotrophic lateral sclerosis and inclusion body myositis. Since p62 functions in protein degradation pathways including autophagy, the build-up of p62-positive inclusions suggests defects in protein clearance. p62 was expressed unilaterally in the rat substantia nigra with an adeno-associated virus vector (AAV9) in order to study p62 neuropathology. Inclusions formed within neurons from several days to several weeks after gene transfer. By electron microscopy, the inclusions were found to contain packed 10 nm thick filaments, and mitochondria cristae structure was disrupted, resulting in the formation of empty spaces. In corollary cell culture transfections, p62 clearly impaired mitochondrial function. To probe for potential effects on macroautophagy, we co-expressed p62 with a double fluorescent tagged reporter for the autophagosome protein LC3 in the rat. p62 induced a dramatic and specific dissociation of the two tags. By 12 weeks, a rotational behavior phenotype manifested, consistent with a significant loss of dopaminergic neurons analyzed post-mortem. p62 overexpression resulted in a progressive and robust pathology model with neuronal inclusions and neurodegeneration. p62 gene transfer could be a novel methodological probe to disrupt mitochondrial function or autophagy in the brain and other tissues in vivo.


Assuntos
Corpos de Inclusão/genética , Corpos de Inclusão/patologia , Doenças Neurodegenerativas/genética , Proteína Sequestossoma-1/genética , Substância Negra/metabolismo , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Miosite de Corpos de Inclusão/genética , Miosite de Corpos de Inclusão/patologia , Doenças Neurodegenerativas/patologia , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Proteína Sequestossoma-1/fisiologia
14.
Inflammation ; 39(5): 1603-16, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27271513

RESUMO

The aim of this study was to clarify the role of p62-dependent mitochondrial apoptosis in the initiation of monosodium urate (MSU) crystal-induced inflammation in macrophages. The induction of mitochondrial apoptosis in RAW 264.7 murine macrophages by MSU crystals was measured using western blotting and quantitative real-time polymerase chain reaction for Bax, caspase-3, caspase-9, or PARP1, and by flow cytometric analysis. Immunoprecipitation and western blotting was applied to detect ubiquitination of p62, TRAF6, and caspase-9. Mitochondrial apoptosis, reactive oxygen species (ROS) generation, and cell proliferation were assessed in cells transfected with p62 small interfering RNA (siRNA). Treatment of RAW 264.7 cells with MSU crystals induced activation of Bax, caspase-3, caspase-9, and PARP1 at the early phase, in addition to enhancing IL-1ß expression, but these findings were attenuated at the late phase. MSU crystals induced ubiquitination of p62, followed by ubiquitination of TRAF6 and caspase-9, which were significantly reversed by ascorbic acid. RAW 264.7 cells transfected with p62 siRNA showed attenuated expression of Bax, caspase-3, caspase-9, and PARP1, decreased ROS and IL-1ß production, and increased cell proliferation, compared to controls. The antioxidant ascorbic acid inhibited p62, caspase-9, and IL-1ß expression increased by MSU crystals. p62 may be a crucial mediator for the mitochondrial apoptosis pathway in MSU crystal-induced inflammation, which is linked to the acute inflammatory response during the early phase of gout.


Assuntos
Apoptose , Interleucina-1beta/biossíntese , Macrófagos/metabolismo , Mitocôndrias/metabolismo , Proteína Sequestossoma-1/fisiologia , Ácido Úrico/farmacologia , Animais , Gota/patologia , Inflamação , Redes e Vias Metabólicas , Camundongos , Células RAW 264.7 , RNA Interferente Pequeno , Proteína Sequestossoma-1/genética
15.
J Cell Sci ; 129(4): 817-30, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26743088

RESUMO

The signaling adaptor sequestosome 1 (SQSTM1)/p62 is frequently overexpressed in tumors and plays an important role in the regulation of tumorigenesis. Although great progress has been made, biological roles of p62 and relevant molecular mechanisms responsible for its pro-tumor activity remain largely unknown. Here, we show that p62 knockdown reduces cell growth and the expression of glycolytic genes in a manner that depends on HIF1α activity in renal cancer cells. Knockdown of p62 decreases HIF1α levels and transcriptional activity by regulating mTORC1 activity and NF-κB nuclear translocation. Furthermore, p62 interacts directly with the von Hippel-Lindau (VHL) E3 ligase complex to modulate the stability of HIF1α. Mechanistically, p62 binds to the VHL complex and competes with HIF1α. Expression of p62 inhibits the interaction of DCNL1 (also known as DCUN1D1) with CUL2 and attenuates the neddylation of CUL2, and thus downregulates the VHL E3 ligase complex activity. Functionally, HIF1α expression is required for p62-induced glucose uptake, lactate production and soft agar colony growth. Taken together, our findings demonstrate that p62 is a crucial positive regulator of HIF1α, which is a facilitating factor in p62-enhanced tumorigenesis.


Assuntos
Glucose/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteína Sequestossoma-1/fisiologia , Proteínas Culina/metabolismo , Glicólise , Células HEK293 , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos/metabolismo , NF-kappa B/metabolismo , Domínios e Motivos de Interação entre Proteínas , Serina-Treonina Quinases TOR/metabolismo , Ubiquitinação , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA