Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Cell Biochem ; 122(3-4): 326-334, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33053230

RESUMO

The Skp1-Cul1-F-box protein (SCF) E3 ligase complex is one of the largest ubiquitin E3 ligase families. FBXL19, a F-box protein in SCFFBXL19 E3 ligase complex, regulates a variety of cellular responses including cell migration. We have shown that FBXL19 is not stable and its degradation is mediated by the ubiquitin-proteasome system, while the ubiquitin E3 ligase for FBXL19 ubiquitination and degradation has not been identified. In the study, we discovered that a new ubiquitin E3 ligase, SCFFBXW17 , ubiquitinates and induces FBXL19 degradation. Exogenous FBXW17 targets FBXL19 for its ubiquitination and degradation. Lysine 114 in FBXL19 is a potential ubiquitin acceptor site. Acetylation of FBXL19 attenuated SCFFBXW17 -mediated FBXL19 degradation. SCFFBXL19 E3 ligase reduced Rac1 levels and cell migration, while the effects were attenuated by exogenous FBXW17. Downregulation of FBXW17 attenuated lysophosphatidic acid-induced lamellipodia formation and Rac1 accumulation at migration leading edge. Taken together with our previous studies, FBXL19 is degraded by the ubiquitin-proteasome system and its site-specific ubiquitination is mediated by SCFFBXW17 E3 ligase, which promotes cell migration.


Assuntos
Movimento Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas F-Box/metabolismo , Acetilação , Animais , Linhagem Celular , Movimento Celular/genética , Proteínas de Ligação a DNA/genética , Proteínas F-Box/genética , Immunoblotting , Imunoprecipitação , Camundongos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ubiquitinação/genética , Ubiquitinação/fisiologia , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
2.
J Immunol ; 194(2): 795-802, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25472995

RESUMO

IL-33, a relatively new member of the IL-1 cytokine family, plays a crucial role in allergic inflammation and acute lung injury. Long form ST2 (ST2L), the receptor for IL-33, is expressed on immune effector cells and lung epithelia and plays a critical role in triggering inflammation. We have previously shown that ST2L stability is regulated by the ubiquitin-proteasome system; however, its upstream internalization has not been studied. In this study, we demonstrate that glycogen synthase kinase 3ß (GSK3ß) regulates ST2L internalization and IL-33 signaling. IL-33 treatment induced ST2L internalization, and an effect was attenuated by inhibition or downregulation of GSK3ß. GSK3ß was found to interact with ST2L on serine residue 446 in response to IL-33 treatment. GSK3ß binding site mutant (ST2L(S446A)) and phosphorylation site mutant (ST2L(S442A)) are resistant to IL-33-induced ST2L internalization. We also found that IL-33 activated focal adhesion kinase (FAK). Inhibition of FAK impaired IL-33-induced GSK3ß activation and ST2L internalization. Furthermore, inhibition of ST2L internalization enhanced IL-33-induced cytokine release in lung epithelial cells. These results suggest that modulation of the ST2L internalization by FAK/GSK3ß might serve as a unique strategy to lessen pulmonary inflammation.


Assuntos
Quinase 1 de Adesão Focal/imunologia , Quinase 3 da Glicogênio Sintase/imunologia , Interleucinas/imunologia , Receptores de Interleucina-1/imunologia , Transdução de Sinais/imunologia , Substituição de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Quinase 1 de Adesão Focal/genética , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Interleucina-33 , Interleucinas/genética , Camundongos , Mutação de Sentido Incorreto , Ligação Proteica , Receptores de Interleucina-1/genética , Transdução de Sinais/genética
3.
Am J Physiol Cell Physiol ; 307(9): C893-9, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25163517

RESUMO

E-cadherin is essential for the integrity of adherens junctions between lung epithelial cells, and the loss of E-cadherin allows cell motility and is thought to promote lung cancer metastasis. While the downregulation of E-cadherin expression has been well characterized and is seen with transforming growth factor-ß1 (TGF-ß1) exposure, few studies have focused on E-cadherin upregulation. Here, we show that serum starvation causes increased E-cadherin expression via the activation of c-Src kinase in non-small-cell lung cancer A549 cells. Serum starvation increased E-cadherin protein levels in a time- and dose-dependent manner. E-cadherin mRNA transcripts were unchanged with starvation, while protein translation inhibition with cycloheximide attenuated E-cadherin protein induction by starvation, suggesting that E-cadherin is regulated at the translational level by serum starvation. c-Src is a nonreceptor tyrosine kinase known to regulate protein translation machinery; serum starvation caused early and sustained activation of c-Src in A549 cells followed by E-cadherin upregulation. Furthermore, overexpression of a dominant negative c-Src attenuated the induction of E-cadherin by serum deprivation. Finally, we observed that TGF-ß1 treatment attenuated the serum activation of c-Src as well as E-cadherin expression when cells were deprived of serum. In conclusion, our data demonstrate that the c-Src kinase is activated by serum starvation to increase E-cadherin expression in A549 cells, and these phenomena are antagonized by TGF-ß1. These novel observations implicate the c-Src kinase as an upstream inducer of E-cadherin protein translation with serum starvation and TGF-ß1 diametrically regulating c-Src kinase activity and thus E-cadherin abundance in A549 cells.


Assuntos
Caderinas/biossíntese , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Caderinas/genética , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Células Cultivadas , Meios de Cultura Livres de Soro , Ativação Enzimática , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Biossíntese de Proteínas , Fator de Crescimento Transformador beta1/farmacologia , Regulação para Cima/efeitos dos fármacos
4.
Mol Cancer ; 13: 76, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24684802

RESUMO

BACKGROUND: Rac3 is a small GTPase multifunctional protein that regulates cell adhesion, migration, and differentiation. It has been considered as an oncogene in breast cancer; however, its role in esophageal cancer and the regulation of its stability have not been studied. F-box proteins are major subunits within the Skp1-Cullin-1-F-box (SCF) E3 ubiquitin ligases that recognize particular substrates for ubiquitination and proteasomal degradation. Recently, we have shown that SCFFBXL19 targets Rac1 and RhoA, thus regulating Rac1 and RhoA ubiquitination and degradation. Here, we demonstrate the role of FBXL19 in the regulation of Rac3 site-specific ubiquitination and stability. Expression of TGFß1 is associated with poor prognosis of esophageal cancer. TGFß1 reduces tumor suppressor, E-cadherin, expression in various epithelial-derived cancers. Here we investigate the role of FBXL19-mediated Rac3 degradation in TGFß1-induced E-cadherin down-regulation in esophageal cancer cells. METHODS: FBXL19-regulated endogenous and over-expressed Rac3 stability were determined by immunoblotting and co-immunoprecipitation. Esophageal cancer cells (OE19 and OE33) were used to investigate TGFß1-induced E-cadherin down-regulation by Immunoblotting and Immunostaining. RESULTS: Overexpression of FBXL19 decreased endogenous and over-expressed Rac3 expression by interacting and polyubiquitinating Rac3, while down-regulation of FBXL19 suppressed Rac3 degradation. Lysine166 within Rac3 was identified as an ubiquitination acceptor site. The FBXL19 variant with truncation at the N-terminus resulted in an increase in Rac3 degradation; however, the FBXL19 variant with truncation at the C-terminus lost its ability to interact with Rac3 and ubiquitinate Rac3 protein. Further, we found that Rac3 plays a critical role in TGFß1-induced E-cadherin down-regulation in esophageal cancer cells. Over-expression of FBXL19 attenuated TGFß1-induced E-cadherin down-regulation and esophageal cancer cells elongation phenotype. CONCLUSIONS: Collectively these data unveil that FBXL19 functions as an antagonist of Rac3 by regulating its stability and regulates the TGFß1-induced E-cadherin down-regulation. This study will provide a new potential therapeutic strategy to regulate TGFß1 signaling, thus suppressing esophageal tumorigenesis.


Assuntos
Caderinas/biossíntese , Proteínas de Ligação a DNA/genética , Neoplasias Esofágicas/genética , Proteínas F-Box/genética , Fator de Crescimento Transformador beta1/metabolismo , Caderinas/genética , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , Neoplasias Esofágicas/patologia , Proteínas F-Box/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Proteólise , Ubiquitinação , Proteínas rac de Ligação ao GTP/genética
5.
Sci Signal ; 10(483)2017 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-28611184

RESUMO

Cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB)-binding protein (CBP) is a histone acetyltransferase that plays a pivotal role in the control of histone modification and the expression of cytokine-encoding genes in inflammatory diseases, including sepsis and lung injury. We found that the E3 ubiquitin ligase subunit FBXL19 targeted CBP for site-specific ubiquitylation and proteasomal degradation. The ubiquitylation-dependent degradation of CBP reduced the extent of lipopolysaccharide (LPS)-dependent histone acetylation and cytokine release in mouse lung epithelial cells and in a mouse model of sepsis. Furthermore, we demonstrated that the deubiquitylating enzyme USP14 (ubiquitin-specific peptidase 14) stabilized CBP by reducing its ubiquitylation. LPS increased the stability of CBP by reducing the association between CBP and FBXL19 and by activating USP14. Inhibition of USP14 reduced CBP protein abundance and attenuated LPS-stimulated histone acetylation and cytokine release. Together, our findings delineate the molecular mechanisms through which CBP stability is regulated by FBXL19 and USP14, which results in the modulation of chromatin remodeling and the expression of cytokine-encoding genes.


Assuntos
Proteína de Ligação a CREB/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas F-Box/metabolismo , Histonas/química , Pneumonia/metabolismo , Ubiquitina Tiolesterase/metabolismo , Acetilação , Animais , Brônquios/metabolismo , Linhagem Celular , Citocinas/metabolismo , Histona Acetiltransferases/genética , Histonas/metabolismo , Humanos , Inflamação , Lipopolissacarídeos/química , Camundongos , Camundongos Endogâmicos C57BL , Células RAW 264.7 , Ubiquitinação/efeitos dos fármacos
6.
EBioMedicine ; 10: 195-203, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27448760

RESUMO

Lysophosphatidic acid receptor 1 (LPA1) is a druggable target for treating pulmonary inflammatory diseases. However, the molecular regulation of LPA1 stability, a factor that critically impacts its biological activity, remains largely unknown. Here we identify two enzymes that regulate the balance of LPA1 ubiquitination and deubiquitination. Ubiquitin E3 ligase Nedd4L targets LPA1 for its site specific ubiquitination and degradation in the lysosome. Nedd4L negatively regulates LPA-LPA1-mediated cytokine release. The stability of LPA1 is up-regulated by ubiquitin-specific protease 11 (USP11), which deubiquitinates LPA1 and enhances LPA1-mediated pro-inflammatory effects. LPA1 is associated with USP11 in quiescent cells, while LPA treatment triggers LPA1 dis-association with USP11 and in turn binding to Nedd4L. Knockdown or inhibition of USP11 reduces LPA1 stability, levels of LPA1, and LPA1-CD14 interaction complex; thereby diminishing both LPA- and LPS-induced inflammatory responses and lung injury in preclinical murine models. Thus, our findings identify an ubiquitin E3 ligase and a deubiquitinating enzyme responsible for regulation of LPA1 stability and biological activities. This study provides potential targets for the development of anti-inflammatory molecules to lessen lung injury.


Assuntos
Citocinas/metabolismo , Lesão Pulmonar/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Animais , Linhagem Celular , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Humanos , Receptores de Lipopolissacarídeos/metabolismo , Lesão Pulmonar/genética , Lesão Pulmonar/patologia , Camundongos , Mitoxantrona/farmacologia , Ubiquitina-Proteína Ligases Nedd4 , Ligação Proteica , Estabilidade Proteica/efeitos dos fármacos , Proteólise , Receptores de Ácidos Lisofosfatídicos/genética , Transdução de Sinais , Tioléster Hidrolases/metabolismo , Receptor 4 Toll-Like/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
7.
Cell Signal ; 26(11): 2406-11, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25025571

RESUMO

The lysophosphatidic acid receptor 1 (LPA1), a G-protein coupled receptor, regulates cell proliferation, migration, and cytokine release. Here, we investigate the molecular signature of LPA1 trafficking to the cell surface. The overexpressed LPA1 with a C-terminal V5 tag (LPA1-V5) is majorly expressed on the cell surface, while two deletion mutants (C320 and ∆84-87) failed to be trafficked to the cell surface. Further, site-directed mutagenesis analysis of the LPA1 revealed that Ile325, Tyr85, and Leu87 within these two fragments regulate LPA1 maturation and trafficking to the cell surface. Over-expression of Sar1, a component of coat protein complex II (COPII), enhances glycosylation of LPA1 wild type, but not these mutants. The mutants of LPA1 are majorly localized in the endoplasmic reticulum (ER) and exhibit a higher binding affinity to heat shock protein 70 (Hsp70), when compared to the LPA1 wild type. Further, we found that all these mutants failed to increase phosphorylation of Erk, and the cytokine release in response to LPA treatment. These results suggest that Ile325, Tyr85, and Leu87 within LPA1 are essential for LPA1 protein properly folding in the ER.


Assuntos
Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Dobramento de Proteína , Receptores de Ácidos Lisofosfatídicos/metabolismo , Animais , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/genética , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/metabolismo , Linhagem Celular , Membrana Celular/genética , Retículo Endoplasmático/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glicosilação , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Camundongos , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Fosforilação/fisiologia , Transporte Proteico/fisiologia , Receptores de Ácidos Lisofosfatídicos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA