Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Biochem Biophys Res Commun ; 540: 95-100, 2021 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-33453679

RESUMO

Acquired middle ear cholesteatoma leads to hearing loss, ear discharge, ear pain, and more serious intracranial complications. However, there is still no effective treatment other than surgery. TFIIB-related factor 2 (BRF2) acted as a redox sensor overexpressing in oxidative stress which linked endoplasmic reticulum (ER) stress, while glucose-regulated protein 78 (GRP78) was a biomarker of ER stress in cancer, atherosclerosis and inflammation. In our study, we investigated the roles of BRF2 and GRP78 in acquired middle ear cholesteatoma. Our results revealed that the expression of BRF2 was significant increased in acquired middle ear cholesteatoma, and which was positively correlated with the expression of GRP78. In addition, BRF2 and GRP78 showed colocalization in epithelium of acquired middle ear cholesteatomas and HaCaT cells. Prolongation of LPS stimulation in HaCaT cells escalated the expression of BRF2 and GRP78. To confirm the role of BRF2 and GRP78, we transfected si-BRF2 into HaCaT cells. All results indicated that BRF2 expression positively regulates the expression of GRP78 and may participate in the pathogenesis of acquire middle ear cholesteatoma.


Assuntos
Colesteatoma da Orelha Média/metabolismo , Proteínas de Choque Térmico/metabolismo , Fator de Transcrição TFIIIB/metabolismo , Linhagem Celular Tumoral , Chaperona BiP do Retículo Endoplasmático , Técnicas de Silenciamento de Genes , Proteínas de Choque Térmico/deficiência , Humanos , Lipopolissacarídeos/imunologia , Fator de Transcrição TFIIIB/deficiência , Regulação para Cima
2.
Biochem Biophys Res Commun ; 527(1): 173-179, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32446363

RESUMO

UC is a chronic inflammatory disease of the colonic mucosa and lacks effective treatments because of unclear pathogenesis. Excessive apoptosis of IECs damages the intestinal epithelial barrier and is involved in the progression of UC, but the mechanism is unknown. HSPs are important in maintaining homeostasis and regulate apoptosis through the mitochondrial pathway. In our previous studies, HSF2, an important regulator of HSPs, was highly expressed in UC patients and negatively correlated with inflammation in mice and IECs. Therefore, we hypothesized that HSF2 may protect against intestinal mucositis by regulating the apoptosis of IECs. In this study, a DSS-induced colitis model of hsf2-/- mice was used to explore the relationship between HSF2 and apoptosis in IECs for the first time. The expression of HSF2 increased in the WT + DSS group compared with that in the WT + H2O group. Moreover, the extent of apoptosis was more severe in the KO + DSS group than in the WT + DSS group. The results showed that HSF2 was negatively correlated with apoptosis in vivo. The expression of HSF2 in Caco-2 cells was changed by lentiviral transfection, and the expression of Bax, cytoplasmic Cyto-C, Cleaved Caspase-9 and Cleaved Caspase-3 were negatively correlated with the different levels of HSF2. These results suggest that HSF2 negatively regulates apoptosis of IECs through the mitochondrial pathway. This may be one of the potential mechanisms to explain the protective role of HSF2 in UC.


Assuntos
Apoptose , Colite Ulcerativa/metabolismo , Células Epiteliais/metabolismo , Proteínas de Choque Térmico/metabolismo , Mucosa Intestinal/metabolismo , Mitocôndrias/metabolismo , Fatores de Transcrição/metabolismo , Animais , Apoptose/efeitos dos fármacos , Células CACO-2 , Células Cultivadas , Colite Ulcerativa/patologia , Relação Dose-Resposta a Droga , Células Epiteliais/patologia , Proteínas de Choque Térmico/deficiência , Proteínas de Choque Térmico/genética , Humanos , Mucosa Intestinal/patologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estrutura Molecular , Relação Estrutura-Atividade , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
3.
Int Immunol ; 32(6): 421-432, 2020 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-32154559

RESUMO

Intestinal macrophages participate in the pathogenesis of inflammatory bowel diseases (IBDs) through secreting pro-inflammatory and tissue-damaging factors as well as inducing the differentiation of T helper 1 (Th1) and T helper 17 (Th17) cells. Elucidating the regulatory mechanisms of intestinal macrophage activity in IBDs is important for developing new therapeutic approaches. In the current study, the expression of Sestrins in myeloid cells and lymphocytes in colonic lamina propria (LP) was evaluated in a murine acute colitis model. We found that Sestrin3 was significantly up-regulated in LP macrophages by the colonic LP microenvironment. In the in vitro experiments, lentivirus-mediated Sestrin3 knockdown significantly reduced the production of IL-12 and IL-23 in activated macrophages, in addition to decreasing the expression of classical pro-inflammatory cytokines such as IL-1ß, IL-6 and TNF-α. Additionally, Sestrin3 knockdown impaired macrophage-mediated generation of Th1 and Th17 cells from CD4+ T cells, probably through up-regulating the phosphorylation of mechanistic target of rapamycin complex 1 (mTORC1) in macrophages. In the in vivo experiments, adoptive transfer of Sestrin3-deficient macrophages alleviated the generation of Th1 and Th17 cells in the colonic LP and mesenteric lymph nodes. Furthermore, the adoptive transfer mitigated the severity of colitis, as demonstrated by lower production of pro-inflammatory cytokines and fewer tissue lesions in the colon. Our study suggests that Sestrin3 might be crucial for macrophage-mediated generation of pathogenic Th1 and Th17 cells in IBDs.


Assuntos
Colite/imunologia , Proteínas de Choque Térmico/imunologia , Macrófagos/imunologia , Células Th1/imunologia , Células Th17/imunologia , Animais , Diferenciação Celular/imunologia , Células Cultivadas , Colite/induzido quimicamente , Colite/patologia , Sulfato de Dextrana/administração & dosagem , Modelos Animais de Doenças , Proteínas de Choque Térmico/deficiência , Proteínas de Choque Térmico/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Th1/patologia , Células Th17/patologia
4.
Arch Biochem Biophys ; 676: 108125, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31586554

RESUMO

Intrahepatic cholestasis of pregnancy (ICP) is a liver disorder occurred in pregnant women, and the mechanism for such disease is still unclear. The bioinformatics analysis of our previous study has revealed the abnormal expression of endoplasmic reticulum protein 29 (ERp29) in placental tissue of ICP patients. In this study, the function of ERp29 was further explored using in vitro model of ICP. The results showed that up-regulation of ERp29 occurred in TCA (taurocholic acid)-treated human trophoblast HTR-8/SVeno cells, and ERp29 inhibition reversed TCA toxicity via attenuating G2/M arrest and cell apoptosis. Mechanical study revealed ERp29 inhibition suppressed phosphorylation and kinase activity of p38, thus subsequently affecting expression and phosphorylation of p53 (ser18) as well as the transcriptional activity of p53. The conduction of this study might confirm the important role of ERp29 in ICP and which would be helpful for the development of target therapeutic method for ICP.


Assuntos
Técnicas de Silenciamento de Genes , Proteínas de Choque Térmico/genética , Ácido Taurocólico/toxicidade , Trofoblastos/citologia , Trofoblastos/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular , Colestase Intra-Hepática/genética , Colestase Intra-Hepática/metabolismo , Colestase Intra-Hepática/patologia , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Proteínas de Choque Térmico/deficiência , Humanos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase M do Ciclo Celular/genética , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Gravidez , Complicações na Gravidez/genética , Complicações na Gravidez/metabolismo , Complicações na Gravidez/patologia , Trofoblastos/efeitos dos fármacos
5.
Artigo em Inglês | MEDLINE | ID: mdl-31396489

RESUMO

The host presents an array of environments which induce bacterial stress including changes in pH, antimicrobial compounds and reactive oxygen species. The bacterial envelope sits at the interface between the intracellular and extracellular environment and its maintenance is essential for Salmonella cell viability under a range of conditions, including during infection. In this study, we aimed to understand the contribution of the σH- and σE-regulated small heat shock proteins IbpA, IbpB, and AgsA and the putative σE-regulated stress response protein STM1250 to the Salmonella envelope stress response. Due to shared sequence identity, regulatory overlap, and the specificity of STM1250 and AgsA to Salmonella sp., we hypothesized that functional overlap exists between these four stress response proteins, which might afford a selective advantage during Salmonella exposure to stress. We present here new roles for three small heat shock proteins and a putative stress response protein in Salmonella that are not limited to heat shock. We have shown that, compared to WT, a quadruple mutant is significantly more sensitive to hydrogen peroxide, has a lower minimum bactericidal concentration to the cationic antimicrobial peptide polymyxin B, and is attenuated in macrophages.


Assuntos
Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno , Macrófagos/imunologia , Viabilidade Microbiana , Estresse Oxidativo , Salmonella typhimurium/imunologia , Estresse Fisiológico , Animais , Proteínas de Bactérias/genética , Deleção de Genes , Redes Reguladoras de Genes , Proteínas de Choque Térmico/deficiência , Proteínas de Choque Térmico/metabolismo , Macrófagos/microbiologia , Camundongos , Modelos Biológicos , Células RAW 264.7 , Infecções por Salmonella/imunologia , Infecções por Salmonella/microbiologia , Salmonella typhimurium/fisiologia , Fator sigma/deficiência , Fator sigma/metabolismo , Virulência
6.
Biochem Cell Biol ; 97(4): 471-479, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30620618

RESUMO

Embryos of the crustacean Artemia franciscana may arrest as gastrulae, forming cysts that enter diapause, which is a state of reduced metabolism and enhanced stress tolerance. Diapausing cysts survive physiological stresses for years due, in part, to molecular chaperones. p26, a small heat-shock protein, is an abundant diapause-specific molecular chaperone in cysts, and it affects embryo development and stress tolerance. p26 is therefore thought to influence many proteins in cysts, and this study was undertaken to determine how the loss of p26 by RNA interference (RNAi) affects the diapause proteome of A. franciscana. The proteome was analyzed by shot-gun proteomics coupled to differential isotopic labeling and tandem mass spectrometry. Proteins in the diapause proteome included metabolic enzymes, antioxidants, binding proteins, structural proteins, transporters, translation factors, receptors, and signal transducers. Proteins within the diapause proteome either disappeared or were reduced in amount when p26 was knocked down, or conversely, proteins appeared or increased in amount. Those proteins that disappeared may be p26 substrates, whereas the synthesis of those proteins that appeared or increased may be regulated by p26. This study provides the first global characterization of the diapause proteome of A. franciscana and demonstrates that the sHsp p26 influences proteome composition.


Assuntos
Artemia/metabolismo , Proteínas de Choque Térmico/deficiência , Proteínas de Choque Térmico/metabolismo , Proteoma/metabolismo , Interferência de RNA , Animais , Biologia Computacional , Feminino , Proteínas de Choque Térmico/isolamento & purificação
7.
Cardiovasc Res ; 115(1): 154-167, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29982352

RESUMO

Aims: Inadequate healing after myocardial infarction (MI) leads to heart failure and fatal ventricular rupture, while optimal healing requires timely induction and resolution of inflammation. This study tested the hypothesis that heat shock protein B1 (HSPB1), which limits myocardial inflammation during endotoxemia, modulates wound healing after MI. Methods and results: To test this hypothesis, cardiomyocyte-specific HSPB1 knockout (Hspb1-/-) mice were generated using the Cre-LoxP recombination system. MI was induced by ligation of the left anterior descending coronary artery in Hspb1-/- and wild-type (WT) littermates. HSPB1 was up-regulated in cardiomyocytes of WT animals in response to MI, and deficiency of cardiomyocyte HSPB1 increased MI-induced cardiac rupture and mortality within 21 days after MI. Serial echocardiography showed more aggravated remodelling and cardiac dysfunction in Hspb1-/- mice than in WT mice at 1, 3, and 7 days after MI. Decreased collagen deposition and angiogenesis, as well as increased MMP2 and MMP9 activity, were also observed in Hspb1-/- mice compared with WT controls after MI, using immunofluorescence, polarized light microscopy, and zymographic analyses. Notably, Hspb1-/- hearts exhibited enhanced and prolonged leucocyte infiltration, enhanced expression of inflammatory cytokines, and enhanced TLR4/MyD88/NFκB activation compared with WT controls after MI. In-depth molecular analyses in both mice and primary cardiomyocytes demonstrated that cardiomyocyte-specific knockout of HSPB1 increased nuclear factor-κB (NFκB) activation, which promoted the expression of proinflammatory mediators. This led to increased leucocyte recruitment, thereby to excessive inflammation, ultimately resulting in adverse remodelling, cardiac dysfunction, and cardiac rupture following MI. Conclusion: These data suggest that HSPB1 acts as a negative regulator of NFκB-mediated leucocyte recruitment and the subsequent inflammation in cardiomyocytes. Cardiomyocyte HSPB1 is required for wound healing after MI and could be a target for myocardial repair in MI patients.


Assuntos
Quimiotaxia de Leucócito , Ruptura Cardíaca Pós-Infarto/metabolismo , Proteínas de Choque Térmico/deficiência , Leucócitos/metabolismo , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , NF-kappa B/metabolismo , Remodelação Ventricular , Animais , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Proteínas de Choque Térmico HSP27/deficiência , Proteínas de Choque Térmico HSP27/genética , Ruptura Cardíaca Pós-Infarto/imunologia , Ruptura Cardíaca Pós-Infarto/patologia , Ruptura Cardíaca Pós-Infarto/fisiopatologia , Proteínas de Choque Térmico/genética , Leucócitos/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Chaperonas Moleculares/genética , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/imunologia , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/patologia , Neovascularização Fisiológica , Ratos Sprague-Dawley , Transdução de Sinais , Cicatrização
8.
Sci Transl Med ; 9(396)2017 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-28659443

RESUMO

Follicular lymphoma (FL) is an incurable form of B cell lymphoma. Genomic studies have cataloged common genetic lesions in FL such as translocation t(14;18), frequent losses of chromosome 6q, and mutations in epigenetic regulators such as EZH2 Using a focused genetic screen, we identified SESTRIN1 as a relevant target of the 6q deletion and demonstrate tumor suppression by SESTRIN1 in vivo. Moreover, SESTRIN1 is a direct target of the lymphoma-specific EZH2 gain-of-function mutation (EZH2Y641X ). SESTRIN1 inactivation disrupts p53-mediated control of mammalian target of rapamycin complex 1 (mTORC1) and enables mRNA translation under genotoxic stress. SESTRIN1 loss represents an alternative to RRAGC mutations that maintain mTORC1 activity under nutrient starvation. The antitumor efficacy of pharmacological EZH2 inhibition depends on SESTRIN1, indicating that mTORC1 control is a critical function of EZH2 in lymphoma. Conversely, EZH2Y641X mutant lymphomas show increased sensitivity to RapaLink-1, a bifunctional mTOR inhibitor. Hence, SESTRIN1 contributes to the genetic and epigenetic control of mTORC1 in lymphoma and influences responses to targeted therapies.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Epigênese Genética , Proteínas de Choque Térmico/genética , Linfoma Folicular/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Animais , Deleção Cromossômica , Cromossomos Humanos Par 6/genética , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Inativação Gênica , Testes Genéticos , Genoma Humano , Proteínas de Choque Térmico/deficiência , Humanos , Camundongos , Mutação/genética , Biossíntese de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
9.
Gut ; 64(2): 195-202, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24789843

RESUMO

OBJECTIVE: Stress in the endoplasmic reticulum (ER) leads to activation of the unfolded protein response (UPR). Xbp1, a key component of the UPR has recently been linked to the risk of developing oesophageal squamous cell carcinoma, suggesting an important role for the UPR in the oesophageal epithelium. Here we examined the role of ER stress and the UPR in oesophageal epithelial homoeostasis. DESIGN: We examined the expression of components of the UPR in the oesophageal epithelium. We used a pharmacological approach and a genetic approach to examine the effects of ER stress in vivo in the mouse oesophagus. The oesophagus of these mice was examined using immunohistochemistry and real-time reverse transcription (RT)-PCR. RESULTS: Components of the UPR were heterogeneously expressed in the basal layer of the epithelium. Induction of ER stress by 24-h treatment with thapsigargin resulted in depletion of proliferating cells in the basal layer of the oesophagus and induced differentiation. We next activated the UPR by inducible deletion of the major ER chaperone Grp78 in Ah1Cre-Rosa26-LacZ-Grp78(-/-) mice in which mutant cells could be traced by expression of LacZ. In these mice LacZ-positive mutant cells in the basal layer lost their proliferative capacity, migrated towards the oesophageal lumen and were replaced by LacZ-negative non-mutant cells. We observed no apoptosis in mutant cells. CONCLUSIONS: These results show that ER stress induces epithelial differentiation in precursor cells in the oesophageal epithelium. This UPR induced differentiation may serve as a quality control mechanism that protects against oesophageal cancer development.


Assuntos
Estresse do Retículo Endoplasmático/fisiologia , Esôfago/citologia , Animais , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Retículo Endoplasmático/ultraestrutura , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Células Epiteliais/citologia , Proteínas de Choque Térmico/deficiência , Proteínas de Choque Térmico/fisiologia , Homeostase/fisiologia , Lactonas/farmacologia , Camundongos Endogâmicos C57BL , Sesquiterpenos/farmacologia , Células-Tronco/citologia , Resposta a Proteínas não Dobradas/fisiologia
10.
Brain Pathol ; 25(5): 552-64, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25412696

RESUMO

In Lewy body disease (LBD) such as dementia with LBs and Parkinson's disease, several lines of evidence show that disrupted proteolysis occurs. p62/SQSTM1 (p62) is highly involved with intracellular proteolysis and is a component of ubiquitin-positive inclusions in various neurodegenerative disorders. However, it is not clear whether p62 deficiency affects inclusion formation and abnormal protein accumulation. To answer this question, we used a mouse model of LBD that lacks p62, and found that LB-like inclusions were observed in transgenic mice that overexpressed α-synuclein (Tg mice) with or without the p62 protein. p62 deficiency enhanced α-synuclein pathology with regard to the number of inclusions and staining intensity compared with Tg mice that expressed p62. To further investigate the molecular mechanisms associated with the loss of p62 in Tg mice, we assessed the mRNA and protein levels of several molecules, and found that the neighbor of the brca1 gene (NBr1), which is functionally and structurally similar to p62, is increased in Tg mice without p62 compared with control Tg mice. These findings suggest that p62 and NBR1 affect the pathogenesis of neurodegenerative diseases through the cooperative modulation of α-synuclein aggregation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/deficiência , Encéfalo/patologia , Proteínas de Choque Térmico/deficiência , Corpos de Inclusão/patologia , Doença por Corpos de Lewy/patologia , alfa-Sinucleína/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Encéfalo/metabolismo , Proteínas de Choque Térmico/genética , Humanos , Corpos de Inclusão/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Doença por Corpos de Lewy/metabolismo , Camundongos , Camundongos Transgênicos , Atividade Motora , Proteínas/metabolismo , Proteólise , Proteína Sequestossoma-1 , Estresse Fisiológico , alfa-Sinucleína/genética
11.
J Clin Invest ; 124(8): 3489-500, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25036708

RESUMO

Type 2 diabetes (T2D) is characterized by a deficiency in ß cell mass, increased ß cell apoptosis, and extracellular accumulation of islet amyloid derived from islet amyloid polypeptide (IAPP), which ß cells coexpress with insulin. IAPP expression is increased in the context of insulin resistance, the major risk factor for developing T2D. Human IAPP is potentially toxic, especially as membrane-permeant oligomers, which have been observed to accumulate within ß cells of patients with T2D and rodents expressing human IAPP. Here, we determined that ß cell IAPP content is regulated by autophagy through p62-dependent lysosomal degradation. Induction of high levels of human IAPP in mouse ß cells resulted in accumulation of this amyloidogenic protein as relatively inert fibrils within cytosolic p62-positive inclusions, which temporarily averts formation of toxic oligomers. Mice hemizygous for transgenic expression of human IAPP did not develop diabetes; however, loss of ß cell-specific autophagy in these animals induced diabetes, which was attributable to accumulation of toxic human IAPP oligomers and loss of ß cell mass. In human IAPP-expressing mice that lack ß cell autophagy, increased oxidative damage and loss of an antioxidant-protective pathway appeared to contribute to increased ß cell apoptosis. These findings indicate that autophagy/lysosomal degradation defends ß cells against proteotoxicity induced by oligomerization-prone human IAPP.


Assuntos
Autofagia/fisiologia , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/fisiologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Diabetes Mellitus Tipo 2/patologia , Diabetes Mellitus Tipo 2/fisiopatologia , Proteínas de Choque Térmico/deficiência , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Insulina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Estrutura Quaternária de Proteína , Proteólise , Ratos , Ratos Transgênicos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína Sequestossoma-1
12.
Cancer Cell ; 26(1): 121-135, 2014 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-25002027

RESUMO

The tumor microenvironment plays a critical role in cancer progression, but the precise mechanisms by which stromal cells influence the epithelium are poorly understood. Here we show that p62 levels were reduced in the stroma of several tumors and that its loss in the tumor microenvironment or stromal fibroblasts resulted in increased tumorigenesis of epithelial prostate cancer cells. The mechanism involves the regulation of cellular redox through an mTORC1/c-Myc pathway of stromal glucose and amino acid metabolism, resulting in increased stromal IL-6 production, which is required for tumor promotion in the epithelial compartment. Thus, p62 is an anti-inflammatory tumor suppressor that acts through the modulation of metabolism in the tumor stroma.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Transformação Celular Neoplásica/metabolismo , Metabolismo Energético , Fibroblastos/enzimologia , Proteínas de Choque Térmico/metabolismo , Inflamação/enzimologia , Complexos Multiproteicos/metabolismo , Neoplasias da Próstata/enzimologia , Transdução de Sinais , Células Estromais/enzimologia , Serina-Treonina Quinases TOR/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Aminoácidos/metabolismo , Animais , Comunicação Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Técnicas de Cocultura , Fibroblastos/metabolismo , Glucose/metabolismo , Células HEK293 , Proteínas de Choque Térmico/deficiência , Proteínas de Choque Térmico/genética , Humanos , Inflamação/genética , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Interleucina-6/metabolismo , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multiproteicos/genética , Invasividade Neoplásica , Estresse Oxidativo , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Hiperplasia Prostática/enzimologia , Hiperplasia Prostática/genética , Hiperplasia Prostática/patologia , Neoplasia Prostática Intraepitelial/enzimologia , Neoplasia Prostática Intraepitelial/genética , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Interferência de RNA , Proteína Sequestossoma-1 , Células Estromais/patologia , Serina-Treonina Quinases TOR/genética , Fatores de Tempo , Transfecção , Microambiente Tumoral
13.
Diabetes ; 63(9): 2996-3008, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24740570

RESUMO

Hyperactivation of the mammalian target of rapamycin complex 1 (mTORC1) in ß-cells is usually found as a consequence of increased metabolic load. Although it plays an essential role in ß-cell compensatory mechanisms, mTORC1 negatively regulates autophagy. Using a mouse model with ß-cell-specific deletion of Tsc2 (ßTsc2(-/-)) and, consequently, mTORC1 hyperactivation, we focused on the role that chronic mTORC1 hyperactivation might have on ß-cell failure. mTORC1 hyperactivation drove an early increase in ß-cell mass that later declined, triggering hyperglycemia. Apoptosis and endoplasmic reticulum stress markers were found in islets of older ßTsc2(-/-) mice as well as accumulation of p62/SQSTM1 and an impaired autophagic response. Mitochondrial mass was increased in ß-cells of ßTsc2(-/-) mice, but mitophagy was also impaired under these circumstances. We provide evidence of ß-cell autophagy impairment as a link between mTORC1 hyperactivation and mitochondrial dysfunction that probably contributes to ß-cell failure.


Assuntos
Autofagia/fisiologia , Células Secretoras de Insulina/patologia , Complexos Multiproteicos/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Apoptose , Estresse do Retículo Endoplasmático , Células HEK293 , Proteínas de Choque Térmico/deficiência , Proteínas de Choque Térmico/metabolismo , Humanos , Resistência à Insulina , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Proteína Sequestossoma-1 , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/deficiência
14.
Eur J Immunol ; 44(4): 1084-92, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24374573

RESUMO

Sequestosome1/A170/p62 (SQSTM1) is a scaffold multifunctional protein involved in several cellular events, such as signal transduction, cell survival, cell death, and inflammation. SQSTM1 expression by macrophages is induced in response to environmental stresses; however, its role in macrophage-mediated host responses to environmental stimuli, such as infectious pathogens, remains unclear. In this study, we investigated the role of SQSTM1 in host responses to Legionella pneumophila, an intra-cellular pathogen that infects macrophages, in both an SQSTM1-deficient (SQSTM1(-/-) ) mouse model and macrophages from these mice. Compared with wild-type (WT) macrophages, the production and secretion of the proinflammatory cytokine IL-1ß was significantly enhanced in SQSTM1(-/-) macrophages after infection with L. pneumophila. Inflammasome activity, indicated by the level of IL-18 and caspase-1 activity, was also elevated in SQSTM1(-/-) macrophages after infection with L. pneumophila. SQSTM1 may interact with nucleotide-binding oligomerization domain-like receptor family, caspase recruitment domain-containing 4 and nucleotide-binding oligomerization domain like receptor family, pyrin domain containing 3 proteins to inhibit their self-dimerization. Acute pulmonary inflammation induced by L. pneumophila and silica was enhanced in SQSTM1(-/-) mice with an increase in IL-1ß levels in the bronchoalveolar lavage fluids. These findings suggest that SQSTM1 is a negative regulator of acute pulmonary inflammation, possibly by regulating inflammasome activity and subsequent proinflammatory cytokine production.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas de Choque Térmico/imunologia , Inflamassomos/imunologia , Legionella pneumophila/imunologia , Doença dos Legionários/imunologia , Pneumonia/imunologia , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/imunologia , Líquido da Lavagem Broncoalveolar/microbiologia , Caspase 1/imunologia , Caspase 1/metabolismo , Ensaio de Imunoadsorção Enzimática , Proteínas de Choque Térmico/deficiência , Proteínas de Choque Térmico/genética , Interações Hospedeiro-Patógeno/imunologia , Immunoblotting , Inflamassomos/metabolismo , Interleucina-18/imunologia , Interleucina-18/metabolismo , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Legionella pneumophila/fisiologia , Doença dos Legionários/microbiologia , Doença dos Legionários/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/imunologia , NF-kappa B/metabolismo , Pneumonia/microbiologia , Pneumonia/patologia , Proteína Sequestossoma-1 , Índice de Gravidade de Doença
15.
J Biol Chem ; 288(50): 35886-95, 2013 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-24178293

RESUMO

The association between altered proteostasis and inflammatory disorders has been increasingly recognized, but the underlying mechanisms are not well understood. In this study, we show that deficiency of either autophagy or sequestosome 1 (p62 or SQSTM) led to inflammasome hyperactivation in response to LPS and ATP in primary macrophages and in mice in vivo. Importantly, induction of protein misfolding by puromycin, thapsigargin, or geldanamycin resulted in inflammasome activation that was more pronounced in autophagy- or p62-deficient macrophages. Accumulation of misfolded proteins caused inflammasome activation by inducing generation of nonmitochondrial reactive oxygen species and lysosomal damage, leading to release of cathepsin B. Our results suggest that altered proteostasis results in inflammasome activation and thus provide mechanisms for the association of altered proteostasis with inflammatory disorders.


Assuntos
Homeostase , Inflamassomos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Animais , Autofagia , Proteínas de Transporte/metabolismo , Catepsina B/metabolismo , Linhagem Celular , Proteínas de Choque Térmico/deficiência , Humanos , Lisossomos/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Dobramento de Proteína , Espécies Reativas de Oxigênio/metabolismo , Proteína Sequestossoma-1
16.
Cancer Gene Ther ; 20(11): 606-15, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24201869

RESUMO

Cancer stem cells are refractory to conventional therapy, which result to cancer metastasis and chemo-radioresistance. Grp78 is known to have important roles in cytoprotection and tumorigenesis in several cancers. We therefore examined whether Grp78 can serve as a therapeutic target for refractory stemness phenotype of head and neck cancer (HNC). Six HNC cell lines were used. Fluorescence-activated cell sorting (FACS) analysis was used to sort CD24(-)CD44(+) and Grp78(+) cells. The small interfering RNA (siRNA) knockdown and cDNA transfection were applied to examine the effects of Grp78 on cellular function. Western blot and confocol microscopy were used to determine the effects of downstream protein expressions. Xenografted mouse tumors and immunohistochemistry were used to validate the results. We found that Grp78 regulated the conversion of CD24(-)CD44(+) cells, a characteristic of HNC stem cells. The CD24(-)CD44(+)Grp78(+) cells showed superior chemo-radioresistance and invasion ability compared with CD24(-)CD44(+), Grp78(+) or the parental cells. Silencing Grp78 increased chemo-radiosensitivity, inhibited cell invasion, reverse epithelial-mesenchymal transition, suppressed cancer stemness, withdrew CD24(-)CD44(+) cell conversion and induced differentiated phenotype. Study in xenografted mice further showed that CD24(-)CD44(+)Grp78(+) cells exhibited highest tumorigenesis, compared with CD24(-)CD44(+) CD24(+)CD44(+) or the parental cells. Grp78 knockdown dramatically restrained tumor growth along with the inhibition of stem cell regulatory proteins Oct-4 and Slug. Grp78 may serve as a molecular target that can be further developed for eradication of refractory HNC with stemness phenotype.


Assuntos
Neoplasias de Cabeça e Pescoço/terapia , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Células-Tronco Neoplásicas/patologia , Animais , Antígeno CD24/biossíntese , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Chaperona BiP do Retículo Endoplasmático , Transição Epitelial-Mesenquimal , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Proteínas de Choque Térmico/deficiência , Humanos , Receptores de Hialuronatos/biossíntese , Camundongos , Terapia de Alvo Molecular , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Fenótipo , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Distribuição Aleatória , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Mol Cell Proteomics ; 12(11): 3253-70, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23938469

RESUMO

Stress-inducible phosphoprotein 1 (STI1), a cochaperone for Hsp90, has been shown to regulate multiple pathways in astrocytes, but its contributions to cellular stress responses are not fully understood. We show that in response to irradiation-mediated DNA damage stress STI1 accumulates in the nucleus of astrocytes. Also, STI1 haploinsufficiency decreases astrocyte survival after irradiation. Using yeast two-hybrid screenings we identified several nuclear proteins as STI1 interactors. Overexpression of one of these interactors, PIAS1, seems to be specifically involved in STI1 nuclear retention and in directing STI1 and Hsp90 to specific sub-nuclear regions. PIAS1 and STI1 co-immunoprecipitate and PIAS1 can function as an E3 SUMO ligase for STI. Using mass spectrometry we identified five SUMOylation sites in STI1. A STI1 mutant lacking these five sites is not SUMOylated, but still accumulates in the nucleus in response to increased expression of PIAS1, suggesting the possibility that a direct interaction with PIAS1 could be responsible for STI1 nuclear retention. To test this possibility, we mapped the interaction sites between PIAS1 and STI1 using yeast-two hybrid assays and surface plasmon resonance and found that a large domain in the N-terminal region of STI1 interacts with high affinity with amino acids 450-480 of PIAS1. Knockdown of PIAS1 in astrocytes impairs the accumulation of nuclear STI1 in response to irradiation. Moreover, a PIAS1 mutant lacking the STI1 binding site is unable to increase STI1 nuclear retention. Interestingly, in human glioblastoma multiforme PIAS1 expression is increased and we found a significant correlation between increased PIAS1 expression and STI1 nuclear localization. These experiments provide evidence that direct interaction between STI1 and PIAS1 is involved in the accumulation of nuclear STI1. This retention mechanism could facilitate nuclear chaperone activity.


Assuntos
Astrócitos/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas Inibidoras de STAT Ativados/metabolismo , Animais , Astrócitos/citologia , Astrócitos/efeitos da radiação , Morte Celular/genética , Morte Celular/fisiologia , Morte Celular/efeitos da radiação , Núcleo Celular/metabolismo , Células Cultivadas , Dano ao DNA , Raios gama , Técnicas de Silenciamento de Genes , Células HEK293 , Haploinsuficiência , Proteínas de Choque Térmico/deficiência , Proteínas de Choque Térmico/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Inibidoras de STAT Ativados/antagonistas & inibidores , Proteínas Inibidoras de STAT Ativados/genética , Mapas de Interação de Proteínas , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Estresse Fisiológico , Sumoilação , Técnicas do Sistema de Duplo-Híbrido
18.
J Med Chem ; 56(18): 7177-89, 2013 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-23961953

RESUMO

Inhibition of ClpB, the bacterial representative of the heat-shock protein 100 family that is associated with virulence of several pathogens, could be an effective strategy to develop new antimicrobial agents. Using a high-throughput screening method, we have identified several compounds that bind to different conformations of ClpB and analyzed their effect on the ATPase and chaperone activities of the protein. Two of them inhibit these functional properties as well as the growth of Gram negative bacteria (E. coli), displaying antimicrobial activity under thermal or oxidative stress conditions. This activity is abolished upon deletion of ClpB, indicating that the action of these compounds is related to the stress cellular response in which ClpB is involved. Moreover, their moderate toxicity in human cell lines suggests that they might provide promising leads against bacterial growth.


Assuntos
Anti-Infecciosos/química , Anti-Infecciosos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Avaliação Pré-Clínica de Medicamentos , Proteínas de Choque Térmico/antagonistas & inibidores , Compostos Orgânicos/química , Compostos Orgânicos/farmacologia , Adenosina Trifosfatases/antagonistas & inibidores , Adenosina Trifosfatases/deficiência , Adenosina Trifosfatases/genética , Animais , Anti-Infecciosos/toxicidade , Proteínas de Bactérias/genética , Células CHO , Cricetinae , Cricetulus , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Deleção de Genes , Células HeLa , Proteínas de Choque Térmico/deficiência , Ensaios de Triagem em Larga Escala , Humanos , Ligantes , Compostos Orgânicos/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Temperatura
19.
Am J Respir Crit Care Med ; 187(8): 865-78, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23449689

RESUMO

RATIONALE: Pulmonary arterial hypertension (PAH) is a lethal, female-predominant, vascular disease. Pathologic changes in PA smooth muscle cells (PASMC) include excessive proliferation, apoptosis-resistance, and mitochondrial fragmentation. Activation of dynamin-related protein increases mitotic fission and promotes this proliferation-apoptosis imbalance. The contribution of decreased fusion and reduced mitofusin-2 (MFN2) expression to PAH is unknown. OBJECTIVES: We hypothesize that decreased MFN2 expression promotes mitochondrial fragmentation, increases proliferation, and impairs apoptosis. The role of MFN2's transcriptional coactivator, peroxisome proliferator-activated receptor γ coactivator 1-α (PGC1α), was assessed. MFN2 therapy was tested in PAH PASMC and in models of PAH. METHODS: Fusion and fission mediators were measured in lungs and PASMC from patients with PAH and female rats with monocrotaline or chronic hypoxia+Sugen-5416 (CH+SU) PAH. The effects of adenoviral mitofusin-2 (Ad-MFN2) overexpression were measured in vitro and in vivo. MEASUREMENTS AND MAIN RESULTS: In normal PASMC, siMFN2 reduced expression of MFN2 and PGC1α; conversely, siPGC1α reduced PGC1α and MFN2 expression. Both interventions caused mitochondrial fragmentation. siMFN2 increased proliferation. In rodent and human PAH PASMC, MFN2 and PGC1α were decreased and mitochondria were fragmented. Ad-MFN2 increased fusion, reduced proliferation, and increased apoptosis in human PAH and CH+SU. In CH+SU, Ad-MFN2 improved walking distance (381 ± 35 vs. 245 ± 39 m; P < 0.05); decreased pulmonary vascular resistance (0.18 ± 0.02 vs. 0.38 ± 0.14 mm Hg/ml/min; P < 0.05); and decreased PA medial thickness (14.5 ± 0.8 vs. 19 ± 1.7%; P < 0.05). Lung vascularity was increased by MFN2. CONCLUSIONS: Decreased expression of MFN2 and PGC1α contribute to mitochondrial fragmentation and a proliferation-apoptosis imbalance in human and experimental PAH. Augmenting MFN2 has therapeutic benefit in human and experimental PAH.


Assuntos
GTP Fosfo-Hidrolases/deficiência , Proteínas de Choque Térmico/deficiência , Hipertensão Pulmonar/fisiopatologia , Dinâmica Mitocondrial/fisiologia , Proteínas Mitocondriais/deficiência , Fatores de Transcrição/deficiência , Animais , Apoptose/fisiologia , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Tolerância ao Exercício/efeitos dos fármacos , Hipertensão Pulmonar Primária Familiar , Feminino , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/patologia , Pulmão/citologia , Pulmão/patologia , Proteínas de Membrana/administração & dosagem , Proteínas de Membrana/deficiência , Dinâmica Mitocondrial/genética , Proteínas Mitocondriais/administração & dosagem , Miócitos de Músculo Liso/patologia , Miócitos de Músculo Liso/fisiologia , Atrofia Óptica Autossômica Dominante/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Ratos , Ratos Sprague-Dawley
20.
PLoS One ; 7(8): e43723, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22952748

RESUMO

Artemia franciscana embryos enter diapause as encysted gastrulae, a physiological state of metabolic dormancy and enhanced stress resistance. The objective of this study was to use RNAi to investigate the function of p26, an abundant, diapause-specific small heat shock protein, in the development and behavior of encysted Artemia embryos (cysts). RNAi methodology was developed where injection of Artemia females with dsRNA specifically eliminated p26 from cysts. p26 mRNA and protein knock down were, respectively, confirmed by RT-PCR and immuno-probing of western blots. ArHsp21 and ArHsp22, diapause-related small heat shock proteins in Artemia cysts sharing a conserved α-crystallin domain with p26, were unaffected by injection of females with dsRNA for p26, demonstrating the specificity of protein knock down. Elimination of p26 delayed cyst release from females demonstrating that this molecular chaperone influences the development of diapause-destined embryos. Although development was slowed the metabolic activities of cysts either containing or lacking p26 were similar. p26 inhibited diapause termination after prolonged incubation of cysts in sea water perhaps by a direct effect on termination or indirectly because p26 is necessary for the preservation of diapause maintenance. Cyst diapause was however, terminated by desiccation and freezing, a procedure leading to high mortality within cyst populations lacking p26 and indicating the protein is required for stress tolerance. Cysts lacking p26 were also less resistant to heat shock. This is the first in vivo study to show that knock down of a small heat shock protein slows the development of diapause-destined embryos, suggesting a role for p26 in the developmental process. The same small heat shock protein prevents spontaneous termination of diapause and provides stress protection to encysted embryos.


Assuntos
Artemia/embriologia , Artemia/fisiologia , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Proteínas de Choque Térmico/metabolismo , Estresse Fisiológico , Animais , Artemia/metabolismo , Embrião não Mamífero/fisiologia , Feminino , Técnicas de Silenciamento de Genes , Proteínas de Choque Térmico/deficiência , Proteínas de Choque Térmico/genética , Injeções , RNA de Cadeia Dupla/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA