Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
EMBO J ; 38(14): e100978, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31304625

RESUMO

Viral infection triggers the formation of mitochondrial antiviral signaling protein (MAVS) aggregates, which potently promote immune signaling. Autophagy plays an important role in controlling MAVS-mediated antiviral signaling; however, the exact molecular mechanism underlying the targeted autophagic degradation of MAVS remains unclear. Here, we investigated the mechanism by which RNF34 regulates immunity and mitophagy by targeting MAVS. RNF34 binds to MAVS in the mitochondrial compartment after viral infection and negatively regulates RIG-I-like receptor (RLR)-mediated antiviral immunity. Moreover, RNF34 catalyzes the K27-/K29-linked ubiquitination of MAVS at Lys 297, 311, 348, and 362 Arg, which serves as a recognition signal for NDP52-dependent autophagic degradation. Specifically, RNF34 initiates the K63- to K27-linked ubiquitination transition on MAVS primarily at Lys 311, which facilitates the autophagic degradation of MAVS upon RIG-I stimulation. Notably, RNF34 is required for the clearance of damaged mitochondria upon viral infection. Thus, we elucidated the mechanism by which RNF34-mediated autophagic degradation of MAVS regulates the innate immune response, mitochondrial homeostasis, and infection.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Transporte/metabolismo , Mitocôndrias/metabolismo , Viroses/imunologia , Proteína DEAD-box 58/metabolismo , Células HEK293 , Células HeLa , Humanos , Imunidade Inata , Lisina/metabolismo , Mitofagia , Proteólise , Receptores Imunológicos , Transdução de Sinais , Células THP-1 , Ubiquitinação , Viroses/metabolismo
2.
Breast Cancer Res Treat ; 202(3): 595-606, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37695401

RESUMO

PURPOSE: The overexpression of mitotic kinase monopolar spindle 1 (Mps1) has been identified in many tumor types, and targeting Mps1 for tumor therapy has shown great promise in multiple preclinical cancer models. However, the role played by Mps1 in tamoxifen (TAM) resistance in breast cancer has never been reported. METHODS: The sensitivity of breast cancer cells to tamoxifen was analysed in colony formation assays and wound healing assays. Enhanced transactivational activity of estrogen receptor α (ERα) led by Mps1 overexpression was determined by luciferase assays. The interaction between Mps1 and ERα was verified by co-immunoprecipitation and proximity ligation assay. Phosphorylation of ERα by Mps1 was detected by in vitro kinase assay and such phosphorylation process in vivo was proven by co-immunoprecipitation. The potential phosphorylation site(s) of ERα were analyzed by mass spectrometry. RESULTS: Mps1 determines the sensitivity of breast cancer cells to tamoxifen treatment. Mps1 overexpression rendered breast cancer cells more resistant to tamoxifen, while an Mps1 inhibitor or siMps1 oligos enabled cancer cells to overcome tamoxifen resistance. Mechanistically, Mps1 interacted with estrogen receptor α and stimulated its transactivational activity in a kinase activity-dependent manner. Mps1 was critical for ERα phosphorylation at Thr224 amino acid site. Importantly, Mps1 failed to enhance the transactivational activity of the ERα-T224A mutant. CONCLUSION: Mps1 contributes to tamoxifen resistance in breast cancer and is a potential therapeutic that can overcome tamoxifen resistance in breast cancer.

3.
Hepatology ; 70(3): 851-870, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30723919

RESUMO

The unfolded protein response (UPR) signal in tumor cells activates UPR signaling in neighboring macrophages, which leads to tumor-promoting inflammation by up-regulating UPR target genes and proinflammatory cytokines. However, the molecular basis of this endoplasmic reticulum (ER) stress transmission remains largely unclear. Here, we identified the secreted form of Golgi protein 73 (GP73), a Golgi-associated protein functional critical for hepatocellular carcinoma (HCC) growth and metastasis, is indispensable for ER stress transmission. Notably, ER stressors increased the cellular secretion of GP73. Through GRP78, the secreted GP73 stimulated ER stress activation in neighboring macrophages, which then released cytokines and chemokines involved in the tumor-associated macrophage (TAM) phenotype. Analysis of HCC patients revealed a positive correlation of GP73 with glucose-regulated protein 78 (GRP78) expression and TAM density. High GP73 and CD206 expression was associated with poor prognosis. Blockade of GP73 decreased the density of TAMs, inhibited tumor growth, and prolonged survival in two mouse HCC models. Conclusion: Our findings provide insight into the molecular mechanisms of extracellular GP73 in the amplification and transmission of ER stress signals.


Assuntos
Carcinoma Hepatocelular/patologia , Estresse do Retículo Endoplasmático/genética , Neoplasias Hepáticas/genética , Fosfoproteínas/genética , Microambiente Tumoral/genética , Análise de Variância , Animais , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Chaperona BiP do Retículo Endoplasmático , Xenoenxertos , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos , Transdução de Sinais/genética , Estatísticas não Paramétricas , Análise de Sobrevida , Regulação para Cima/genética
4.
Microbiol Immunol ; 64(11): 768-777, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32902897

RESUMO

The Gram-negative bacterial pathogen Yersinia delivers six effector proteins into the host cells to block the host innate immune response. One of the effectors, YopT, is a potent cysteine protease that causes the disruption of the actin cytoskeleton to inhibit phagocytosis of the pathogen; however, its molecular mechanism and relevance to pathogenesis need further investigation. In this report, we show that RIG-I is a novel target of the YopT protein. Remarkably, YopT interacts with RIG-I and inhibits rat liver homogenate-mediated nuclear factor-κB and interferon regulatory factor-3 activation. Further studies revealed a YopT-dependent increase in the K48-polymerized ubiquitination of RIG-I. These findings suggest that YopT negatively regulates RIG-I-mediated cellular antibacterial response by targeting RIG-I.


Assuntos
Proteínas de Bactérias/metabolismo , Cisteína Endopeptidases/metabolismo , Fator Regulador 3 de Interferon/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/fisiologia , Yersinia/metabolismo , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Cisteína Endopeptidases/genética , Células HEK293 , Humanos , Camundongos , NF-kappa B/genética , Fagocitose , Células RAW 264.7 , Fator de Transcrição RelA , Yersinia/genética
5.
PLoS Pathog ; 13(6): e1006347, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28591144

RESUMO

Estrogen-related receptor α (ERRα) is a member of the nuclear receptor superfamily controlling energy homeostasis; however, its precise role in regulating antiviral innate immunity remains to be clarified. Here, we showed that ERRα deficiency conferred resistance to viral infection both in vivo and in vitro. Mechanistically, ERRα inhibited the production of type-I interferon (IFN-I) and the expression of multiple interferon-stimulated genes (ISGs). Furthermore, we found that viral infection induced TBK1-dependent ERRα stabilization, which in turn associated with TBK1 and IRF3 to impede the formation of TBK1-IRF3, IRF3 phosphorylation, IRF3 dimerization, and the DNA binding affinity of IRF3. The effect of ERRα on IFN-I production was independent of its transcriptional activity and PCG-1α. Notably, ERRα chemical inhibitor XCT790 has broad antiviral potency. This work not only identifies ERRα as a critical negative regulator of antiviral signaling, but also provides a potential target for future antiviral therapy.


Assuntos
Fator Regulador 3 de Interferon/imunologia , Interferon Tipo I/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Receptores de Estrogênio/imunologia , Viroses/imunologia , Células A549 , Animais , Western Blotting , Ensaio de Imunoadsorção Enzimática , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunoprecipitação , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Serina-Treonina Quinases/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Estrogênio/metabolismo , Transdução de Sinais/imunologia , Viroses/metabolismo , Receptor ERRalfa Relacionado ao Estrogênio
6.
J Immunol ; 198(12): 4652-4658, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28476934

RESUMO

Tripartite motif-containing 14 (TRIM14) is a mitochondrial adaptor that facilitates innate immune signaling. Upon virus infection, the expression of TRIM14 is significantly induced, which stimulates the production of type-I IFNs and proinflammatory cytokines. As excessive immune responses lead to harmful consequences, TRIM14-mediated signaling needs to be tightly balanced. In this study, we identify really interesting new gene-type zinc finger protein 125 (RNF125) as a negative regulator of TRIM14 in the innate antiviral immune response. Overexpression of RNF125 inhibits TRIM14-mediated antiviral response, whereas knockdown of RNF125 has the opposite effect. RNF125 interacts with TRIM14 and acts as an E3 ubiquitin ligase that catalyzes TRIM14 ubiquitination. RNF125 promotes K48-linked polyubiquitination of TRIM14 and mediates its degradation via the ubiquitin-proteasome pathway. Consequently, wild-type mouse embryonic fibroblasts show significantly reduced TRIM14 protein levels in late time points of viral infection, whereas TRIM14 protein is retained in RNF125-deficient mouse embryonic fibroblasts. Collectively, our data suggest that RNF125 plays a new role in innate immune response by regulating TRIM14 ubiquitination and degradation.


Assuntos
Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica/imunologia , Imunidade Inata/imunologia , Transativadores/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Ubiquitinação , Animais , Biocatálise , Proteínas de Transporte/imunologia , Fibroblastos/imunologia , Fibroblastos/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Transativadores/imunologia , Proteínas com Motivo Tripartido , Ubiquitina/imunologia , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia
7.
Mol Cell Biochem ; 445(1-2): 35-43, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29280086

RESUMO

Stringent control of the type I interferon signaling pathways is critical to effective host immune responses, however, the molecular mechanisms that negatively regulate these pathways are still poorly understood. Here, we show that apoptosis speck-like protein (ASC), an adaptor protein of inflammasome complex, can inhibit IFN-ß signaling response by interacting with mitochondrial antiviral signaling protein (MAVS). Importantly, ASC-specific siRNA knockdown enhanced virus-induced type I interferon production, with consequent reduction of virus replication. Taken together, these results suggest that ASC, as a negative regulator of the MAVS-mediated innate immunity, may play an important role in host protection upon virus infection.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas Adaptadoras de Sinalização CARD/fisiologia , Imunidade Inata/fisiologia , Animais , Proteínas Adaptadoras de Sinalização CARD/genética , Células Cultivadas , Técnicas de Silenciamento de Genes , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Inflamassomos/fisiologia , Interferon beta/genética , Interferon beta/metabolismo , Células MCF-7 , Camundongos , RNA Interferente Pequeno/genética , Transdução de Sinais/fisiologia , Viroses/imunologia , Viroses/prevenção & controle , Replicação Viral/fisiologia
8.
J Immunol ; 196(3): 1199-208, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26700764

RESUMO

IFN regulatory factors play a pivotal role in many cellular processes, including inflammatory and immune responses. Their activation is tightly regulated by TANK-binding kinase 1 (TBK1). In response to microbial components, TBK1 activates IFN regulatory factor 3 (IRF3) and cytokine expression. In this article, we show that TBK1 is a novel target of the IpaH4.5 protein, a Shigella type III effector possessing E3 ubiquitin ligase activity. Remarkably, IpaH4.5 interacts with TBK1 and promotes its K48-linked polyubiquitylation. Consequently, polyubiquitylated TBK1 undergoes proteasome-dependent degradation, which perturbs the phosphorylation, nuclear translocation, and activation of IRF3. Because IRF3 and TBK1 are required for restricting Shigella growth, we propose that the polyubiquitylation and degradation of TBK1 during Shigella infection are new bacterial strategies to modulate the host antibacterial responses.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Disenteria Bacilar/metabolismo , Interações Hospedeiro-Parasita/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Linhagem Celular , Disenteria Bacilar/imunologia , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Imunoprecipitação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Serina-Treonina Quinases/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Transfecção
9.
Biochim Biophys Acta ; 1863(12): 3148-3159, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27742471

RESUMO

The Yersinia outer protein J (YopJ) plays a pivotal role in evading the host immune response and establishes a persistent infection in host cells after bacterial infection. YopJ is a cysteine protease and can act as a deubiquitinating enzyme that deubiquitinates several targets in multiple signaling pathways. Stimulator of interferon genes (STING) is a critical adapter for the induction of interferon regulatory factor 3 (IRF3) phosphorylation and subsequent production of the cytokines in response to nucleic acids in the cytoplasm. Our studies demonstrate that YopJ targets STING to inhibit IRF3 signaling. Specially, YopJ interacts with STING to block its ER-to-Golgi traffic and remove its K63-linked ubiquitination chains. Deubiquited STING perturbs the formation of STING-TBK1 complex and the activation of IRF3. The 172th cysteine of YopJ mediated STING deubiquitination and IRF3 signaling inhibition. Consequently, mice infected with WT and ΔYopJ/YopJ bacteria induced lower levels of IRF3 and IFN-ß, decreased inflammation and reduced staining of STING as compared to ΔYopJ and ΔYopJ/YopJ C172A strains infection. The data herein reveal a previously unrecognized mechanism by which YopJ modulates innate immune signaling.


Assuntos
Proteínas de Bactérias/genética , DNA/genética , Evasão da Resposta Imune , Fator Regulador 3 de Interferon/genética , Proteínas de Membrana/genética , Yersinia pestis/genética , Animais , Proteínas de Bactérias/imunologia , Linhagem Celular , DNA/imunologia , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/microbiologia , Deleção de Genes , Regulação da Expressão Gênica , Complexo de Golgi/metabolismo , Complexo de Golgi/microbiologia , Células HEK293 , Humanos , Imunidade Inata , Fator Regulador 3 de Interferon/imunologia , Interferon beta/genética , Interferon beta/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Proteínas de Membrana/imunologia , Camundongos , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Transdução de Sinais , Ubiquitinação , Yersinia pestis/crescimento & desenvolvimento , Yersinia pestis/patogenicidade
10.
J Immunol ; 194(10): 4880-90, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25847972

RESUMO

Stringent control of inflammasome signaling pathway is important for maintaining immunological balance, yet the molecular mechanisms responsible for its tight regulation are still poorly understood. In this study, we found that the signaling pathway dependent on mitochondrial antiviral signaling protein (MAVS) was required for the optimal activation of apoptosis-associated specklike protein (ASC)-dependent inflammasome. In particular, TNFR-associated factor 3 was found to be a direct E3 ligase for ASC. Ubiquitination of ASC at Lys(174) was critical for speck formation and inflammasome activation. Deficiency in MAVS or TNFR-associated factor 3 impaired ASC ubiquitination and cytosolic aggregates formation, resulting in reduced inflammasome response upon RNA virus infection. This study has identified a previously unrecognized role of MAVS in the regulation of inflammasome signaling and provided molecular insight into the mechanisms by which ubiquitination of ASC controls inflammasome activity through the formation of ASC specks.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas Reguladoras de Apoptose/imunologia , Inflamassomos/imunologia , Transdução de Sinais/imunologia , Fator 3 Associado a Receptor de TNF/imunologia , Ubiquitinação , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Adaptadoras de Sinalização CARD , Immunoblotting , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Interferência de RNA , Viroses/imunologia
11.
Proc Natl Acad Sci U S A ; 111(5): E601-10, 2014 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-24449872

RESUMO

Resistance to antiestrogens is one of the major challenges in breast cancer treatment. Although phosphorylation of estrogen receptor α (ERα) is an important factor in endocrine resistance, the contributions of specific kinases in endocrine resistance are still not fully understood. Here, we report that an important innate immune response kinase, the IκB kinase-related TANK-binding kinase 1 (TBK1), is a crucial determinant of resistance to tamoxifen therapies. We show that TBK1 increases ERα transcriptional activity through phosphorylation modification of ERα at the Ser-305 site. Ectopic TBK1 expression impairs the responsiveness of breast cancer cells to tamoxifen. By studying the specimens from patients with breast cancer, we find a strong positive correlation of TBK1 with ERα, ERα Ser-305, and cyclin D1. Notably, patients with tumors highly expressing TBK1 respond poorly to tamoxifen treatment and show high potential for relapse. Therefore, our findings suggest that TBK1 contributes to tamoxifen resistance in breast cancer via phosphorylation modification of ERα.


Assuntos
Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Tamoxifeno/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Ciclina D1/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Imunidade Inata/efeitos dos fármacos , Estimativa de Kaplan-Meier , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Tamoxifeno/uso terapêutico , Transcrição Gênica/efeitos dos fármacos , Resultado do Tratamento
12.
J Virol ; 88(19): 11356-68, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25056901

RESUMO

UNLABELLED: Retinoic acid-inducible gene I (RIG-I) is an intracellular RNA virus sensor that induces type I interferon-mediated host-protective innate immunity against viral infection. Although cylindromatosis (CYLD) has been shown to negatively regulate innate antiviral response by removing K-63-linked polyubiquitin from RIG-I, the regulation of its expression and the underlying regulatory mechanisms are still incompletely understood. Here we show that RIG-I activity is regulated by inhibition of CYLD expression mediated by the microRNA miR-526a. We found that viral infection specifically upregulates miR-526a expression in macrophages via interferon regulatory factor (IRF)-dependent mechanisms. In turn, miR-526a positively regulates virus-triggered type I interferon (IFN-I) production, thus suppressing viral replication, the underlying mechanism of which is the enhancement of RIG-I K63-linked ubiquitination by miR-526a via suppression of the expression of CYLD. Remarkably, virus-induced miR-526a upregulation and CYLD downregulation are blocked by enterovirus 71 (EV71) 3C protein, while ectopic miR-526a expression inhibits the replication of EV71 virus. The collective results of this study suggest a novel mechanism of the regulation of RIG-I activity during RNA virus infection by miR-526a and suggest a novel mechanism for the evasion of the innate immune response controlled by EV71. IMPORTANCE: RNA virus infection upregulates the expression of miR-526a in macrophages through IRF-dependent pathways. In turn, miR-526a positively regulates virus-triggered type I IFN production and inhibits viral replication, the underlying mechanism of which is the enhancement of RIG-I K-63 ubiquitination by miR-526a via suppression of the expression of CYLD. Remarkably, virus-induced miR-526a upregulation and CYLD downregulation are blocked by enterovirus 71 (EV71) 3C protein; cells with overexpressed miR-526a were highly resistant to EV71 infection. The collective results of this study suggest a novel mechanism of the regulation of RIG-I activity during RNA virus infection by miR-526a and propose a novel mechanism for the evasion of the innate immune response controlled by EV71.


Assuntos
RNA Helicases DEAD-box/genética , Enterovirus Humano A/genética , Evasão da Resposta Imune , Imunidade Inata , MicroRNAs/genética , Proteínas Virais/genética , Proteases Virais 3C , Animais , Chlorocebus aethiops , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/imunologia , Proteína DEAD-box 58 , RNA Helicases DEAD-box/imunologia , Enzima Desubiquitinante CYLD , Cães , Enterovirus Humano A/imunologia , Regulação da Expressão Gênica , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Macrófagos/imunologia , Macrófagos/virologia , Células Madin Darby de Rim Canino , MicroRNAs/imunologia , Poliubiquitina/genética , Poliubiquitina/imunologia , Receptores Imunológicos , Transdução de Sinais , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/imunologia , Células Vero , Proteínas Virais/imunologia , Replicação Viral
13.
Mol Cell Biochem ; 407(1-2): 69-76, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26002288

RESUMO

MicroRNAs (miRNAs) play vital roles in the regulation of cell cycle, cell growth, apoptosis, and tumorigenesis. Our previous studies showed that miR-526a positively regulated innate immune response by suppressing CYLD expression, however, the functional relevance of miR-526a expression and cell growth remains to be evaluated. In this study, miR-526a overexpression was found to promote cancer cell proliferation, migration, and anchor-independent colony formation. The molecular mechanism(s) of miR-526a-mediated growth stimulation is associated with rapid cell cycle progression and inhibition of cell apoptosis by targeting CYLD. Taken together, these results provide evidence to show the stimulatory role of miR-526a in tumor migration and invasion through modulation of the canonical NF-κB signaling pathway.


Assuntos
MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias/genética , Apoptose , Movimento Celular , Proliferação de Células , Enzima Desubiquitinante CYLD , Células HEK293 , Células Hep G2 , Humanos , Células MCF-7 , NF-kappa B/metabolismo , Invasividade Neoplásica , Neoplasias/metabolismo , Neoplasias/patologia , Transdução de Sinais , Proteínas Supressoras de Tumor/genética
14.
Biochem Biophys Res Commun ; 450(4): 1690-5, 2014 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-25063032

RESUMO

The spindle assembly checkpoint kinase Mps1 is highly expressed in several types of cancers, but its cellular involvement in tumorigenesis is less defined. Herein, we confirm that Mps1 is overexpressed in colon cancer tissues. Further, we find that forced expression of Mps1 in the colon cancer cell line SW480 enables cells to become resistant to both Mps1 inhibition-induced checkpoint depletion and cell death. Overexpression of Mps1 also increases genome instability in tumor cells owing to a weakened spindle assembly checkpoint. Collectively, our findings suggest that high levels of Mps1 contribute to tumorigenesis by attenuating the spindle assembly checkpoint.


Assuntos
Aneuploidia , Proteínas de Ciclo Celular/metabolismo , Neoplasias do Colo/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Fuso Acromático , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Regulação para Baixo , Humanos , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética
16.
ACS Synth Biol ; 12(6): 1686-1695, 2023 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-37196336

RESUMO

Noticeable morbidity and mortality can be caused by influenza A virus in humans. Conventional live attenuated influenza vaccine (LAIV) is one of the main strategies to control the spread of influenza, but its protective efficacy is often limited by its suboptimal immunogenicity and safety. Therefore, a new type of LAIV that can overcome the shortage of existing vaccines is urgently needed. Here, we report a novel method to construct the recombinant influenza A virus (IAV) regulated by small molecules. By inserting 4-hydroxytamoxifen (4-HT)-dependent intein into the polymerase acidic (PA) protein of IAV, a series of 4-HT-dependent recombinant viruses were generated and screened. Among them, the S218 recombinant virus strain showed excellent 4-HT dependent replication characteristics both in vitro and in vivo. Further immunological evaluation indicated that the 4-HT-dependent viruses were highly attenuated in the host and could elicit robust humoral, mucosal, and cellular immunity against the challenge of homologous viruses. The attenuated strategies presented here could also be broadly applied to the development of vaccines against other pathogens.


Assuntos
Vírus da Influenza A , Vacinas contra Influenza , Influenza Humana , Infecções por Orthomyxoviridae , Humanos , Inteínas , Vírus da Influenza A/genética , Vacinas Atenuadas
17.
Front Cell Infect Microbiol ; 13: 1134511, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36936774

RESUMO

Introduction: Inflammation play important roles in the initiation and progression of acute lung injury (ALI), acute respiratory distress syndrome (ARDS), septic shock, clotting dysfunction, or even death associated with SARS-CoV-2 infection. However, the pathogenic mechanisms underlying SARS-CoV-2-induced hyperinflammation are still largely unknown. Methods: The animal model of septic shock and ALI was established after LPS intraperitoneal injection or intratracheal instillation. Bone marrow-derived macrophages (BMDMs) from WT and BPOZ-2 KO mouse strains were harvested from the femurs and tibias of mice. Immunohistology staining, ELISA assay, coimmunoprecipitation, and immunoblot analysis were used to detect the histopathological changes of lung tissues and the expression of inflammatory factors and protein interaction. Results and conclusions: We show a distinct mechanism by which the SARS-CoV-2 N (SARS-2-N) protein targets Bood POZ-containing gene type 2 (BPOZ-2), a scaffold protein for the E3 ubiquitin ligase Cullin 3 that we identified as a negative regulator of inflammatory responses, to promote NLRP3 inflammasome activation. We first demonstrated that BPOZ-2 knockout (BPOZ-2 KO) mice were more susceptible to lipopolysaccharide (LPS)-induced septic shock and ALI and showed increased serum IL-1ß levels. In addition, BMDMs isolated from BPOZ-2 KO mice showed increased IL-1ß production in response to NLRP3 stimuli. Mechanistically, BPOZ-2 interacted with NLRP3 and mediated its degradation by recruiting Cullin 3. In particular, the expression of BPOZ-2 was significantly reduced in lung tissues from mice infected with SARS-CoV-2 and in cells overexpressing SARS-2-N. Importantly, proinflammatory responses triggered by the SARS-2-N were significantly blocked by BPOZ-2 reintroduction. Thus, we concluded that BPOZ-2 is a negative regulator of the NLPR3 inflammasome that likely contributes to SARS-CoV-2-induced hyperinflammation.


Assuntos
Lesão Pulmonar Aguda , COVID-19 , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteínas Nucleares , Choque Séptico , Animais , Camundongos , Lesão Pulmonar Aguda/metabolismo , Proteínas Culina , Inflamassomos/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , SARS-CoV-2/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo
18.
J Mol Endocrinol ; 70(2)2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36394986

RESUMO

Golgi protein 73 (GP73), also called Golgi membrane protein 1 (GOLM1), is a resident Golgi type II transmembrane protein and is considered as a serum marker for the detection of a variety of cancers. A recent work revealed the role of the secreted GP73 in stimulating liver glucose production and systemic glucose homeostasis. Since exaggerated hepatic glucose production plays a key role in the pathogenesis of type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM), GP73 may thus represent a potential therapeutic target for treating diabetic patients with pathologically elevated levels. Here, in this study, we found that the circulating GP73 levels were significantly elevated in T2DM and positively correlated with hemoglobin A1c. Notably, the aberrantly upregulated GP73 levels were indispensable for the enhanced protein kinase A signaling pathway associated with diabetes. In diet-induced obese mouse model, GP73 siRNA primarily targeting liver tissue was potently effective in alleviating abnormal glucose metabolism. Ablation of GP73 from whole animals also exerted a profound glucose-lowering effect. Importantly, neutralizing circulating GP73 improved glucose metabolism in streptozotocin (STZ) and high-fat diet/STZ-induced diabetic mice. We thus concluded that GP73 was a feasible therapeutic target for the treatment of diabetes.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Camundongos , Animais , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Experimental/patologia , Fígado/metabolismo , Glucose/metabolismo , Homeostase
19.
J Immunol ; 185(2): 1158-68, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20554965

RESUMO

Previous studies have shown that both hepatitis A virus and hepatitis C virus inhibit innate immunity by cleaving the mitochondrial antiviral signaling (MAVS) protein, an essential component of the virus-activated signaling pathway that activates NF-kappaB and IFN regulatory factor-3 to induce the production of type I IFN. For human hepatitis B virus (HBV), hepatitis B s-Ag, hepatitis B e-Ag, or HBV virions have been shown to suppress TLR-induced antiviral activity with reduced IFN-beta production and subsequent induction of IFN-stimulated genes. However, HBV-mediated suppression of the RIG-I-MDA5 pathway is unknown. In this study, we found that HBV suppressed poly(deoxyadenylate-thymidylate)-activated IFN-beta production in hepatocytes. Specifically, hepatitis B virus X (HBX) interacted with MAVS and promoted the degradation of MAVS through Lys(136) ubiquitin in MAVS protein, thus preventing the induction of IFN-beta. Further analysis of clinical samples revealed that MAVS protein was downregulated in hepatocellular carcinomas of HBV origin, which correlated with increased sensitivities of primary murine hepatocytes isolated from HBX knock-in transgenic mice upon vesicular stomatitis virus infections. By establishing a link between MAVS and HBX, this study suggests that HBV can target the RIG-I signaling by HBX-mediated MAVS downregulation, thereby attenuating the antiviral response of the innate immune system.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Imunidade Inata/imunologia , Proteínas Mitocondriais/imunologia , Transativadores/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/imunologia , RNA Helicases DEAD-box/metabolismo , Regulação para Baixo , Células Hep G2 , Humanos , Immunoblotting , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Cinética , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Poli dA-dT/genética , Poli dA-dT/imunologia , Poli dA-dT/metabolismo , Ligação Proteica , Receptores Imunológicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia , Transativadores/genética , Transativadores/metabolismo , Transfecção , Células Vero , Proteínas Virais Reguladoras e Acessórias
20.
Front Oncol ; 12: 835603, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35965501

RESUMO

As a critical immune checkpoint molecule, PD-L1 is expressed at significantly higher levels in multiple neoplastic tissues compared to normal ones. PD-L1/PD-1 axis is a critical target for tumor immunotherapy, blocking the PD-L1/PD-1 axis is recognized and has achieved unprecedented success in clinical applications. However, the clinical efficacy of therapies targeting the PD-1/PD-L1 pathway remains limited, emphasizing the need for the mechanistic elucidation of PD-1/PD-L1 expression. In this study, we found that RNF125 interacted with PD-L1 and regulated PD-L1 protein expression. Mechanistically, RNF125 promoted K48-linked polyubiquitination of PD-L1 and mediated its degradation. Notably, MC-38 and H22 cell lines with RNF125 knockout, transplanted in C57BL/6 mice, exhibited a higher PD-L1 level and faster tumor growth than their parental cell lines. In contrast, overexpression of RNF125 in MC-38 and H22 cells had the opposite effect, resulting in lower PD-L1 levels and delayed tumor growth compared with parental cell lines. In addition, immunohistochemical analysis of MC-38 tumors with RNF125 overexpression showed significantly increased infiltration of CD4+, CD8+ T cells and macrophages. Consistent with these findings, analyses using The Cancer Genome Atlas (TCGA) public database revealed a positive correlation of RNF125 expression with CD4+, CD8+ T cell and macrophage tumor infiltration. Moreover, RNF125 expression was significantly downregulated in several human cancer tissues, and was negatively correlated with the clinical stage of these tumors, and patients with higher RNF125 expression had better clinical outcomes. Our findings identify a novel mechanism for regulating PD-L1 expression and may provide a new strategy to increase the efficacy of immunotherapy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA