Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 15: 1378359, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38779662

RESUMO

Skin tissue-resident memory T (Trm) cells are produced by antigenic stimulation and remain in the skin for a long time without entering the peripheral circulation. In the healthy state Trm cells can play a patrolling and surveillance role, but in the disease state Trm cells differentiate into various phenotypes associated with different diseases, exhibit different localizations, and consequently have local protective or pathogenic roles, such as disease recurrence in vitiligo and maintenance of immune homeostasis in melanoma. The most common surface marker of Trm cells is CD69/CD103. However, the plasticity of tissue-resident memory T cells after colonization remains somewhat uncertain. This ambiguity is largely due to the variation in the functionality and ultimate destination of Trm cells produced from memory cells differentiated from diverse precursors. Notably, the presence of Trm cells is not stationary across numerous non-lymphoid tissues, most notably in the skin. These cells may reenter the blood and distant tissue sites during the recall response, revealing the recycling and migration potential of the Trm cell progeny. This review focuses on the origin and function of skin Trm cells, and provides new insights into the role of skin Trm cells in the treatment of autoimmune skin diseases, infectious skin diseases, and tumors.


Assuntos
Plasticidade Celular , Homeostase , Memória Imunológica , Células T de Memória , Dermatopatias , Pele , Humanos , Homeostase/imunologia , Células T de Memória/imunologia , Células T de Memória/metabolismo , Pele/imunologia , Pele/patologia , Plasticidade Celular/imunologia , Animais , Dermatopatias/imunologia , Antígenos CD/metabolismo , Antígenos CD/imunologia
2.
Int Immunopharmacol ; 121: 110487, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37364328

RESUMO

In this study, we explored the possible mechanism of tumor tolerance induced by multiple repeated immunizations with a tumor vaccine (MUC1-MBP fusion protein plus CpG2006). We first analyzed the mechanism of tolerance by immunizing tumor-bearing mice 2, 5, or 8 times and found that compared with five immunizations with the M-M vaccine, eight immunizations increased tumor volume and weight and Treg levels, while the proportions of Th1 and Tc1 cells in the spleen and lymph nodes were decreased. In particular, the M-M vaccine induced PD-L1 expression in CD11c + DCs and decreased their CD80/PD-L1 ratio. Therefore, the mechanism of tolerance induction by multiple immunizations with the M-M vaccine was investigated by focusing on the CD80/PD-L1 ratio, and an anti-PD-L1 antibody (αPD-L1) and the M-M vaccine were used in combination to treat melanoma. The results showed that αPD-L1 increased the CD80/PD-L1 ratio and enhanced the maturation of cDC1s by blocking PD-L1 on DCs, which potentially increased the activity of Th1 and Tc1 cells. Furthermore, the combination of the M-M vaccine with αPD-L1 decreased the activity and proportion of Tregs, which reversed the immune tolerance induced by eight immunizations with the vaccine. This study reveals the mechanism of the combination of M-M and αPD-L1 and provides a new combination strategy for improving the therapeutic effect of the M-M vaccine, laying a theoretical basis for the clinical application of the vaccine.


Assuntos
Vacinas Anticâncer , Melanoma , Camundongos , Animais , Linfócitos T Reguladores , Imunização , Melanoma/tratamento farmacológico , Tolerância Imunológica
3.
Int J Mol Sci ; 23(18)2022 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-36142800

RESUMO

Our previous study found that CpG oligodeoxynucleotides 1826 (CpG 1826), combined with mucin 1 (MUC1)-maltose-binding protein (MBP) (M-M), had certain antitumor activity. However, this combination is less than ideal for tumor suppression (tumors vary in size and vary widely among individuals), with a drawback being that CpG 1826 is unstable. To solve these problems, here, we evaluate MF59/CpG 1826 as a compound adjuvant with M-M vaccine on immune response, tumor suppression and survival. The results showed that MF59 could promote the CpG 1826/M-M vaccine-induced tumor growth inhibition and a Th1-prone cellular immune response, as well as reduce the individual differences of tumor growth and prolonged prophylactic and therapeutic mouse survival. Further research showed that MF59 promotes the maturation of DCs stimulated by CpG1826/M-M, resulting in Th1 polarization. The possible mechanism is speculated to be that MF59 could significantly prolong the retention time of CpG 1826, or the combination of CpG 1826 and M-M, as well as downregulate IL-6/STAT3 involved in MF59 combined CpG 1826-induced dendritic cell maturation. This study clarifies the utility of MF59/CpG 1826 as a vaccine compound adjuvant, laying the theoretical basis for the development of a novel M-M vaccine.


Assuntos
Vacinas Anticâncer , Neoplasias , Adjuvantes Imunológicos/farmacologia , Animais , Antígenos , Células Dendríticas , Interleucina-6 , Proteínas Ligantes de Maltose , Camundongos , Camundongos Endogâmicos C57BL , Mucina-1/genética , Neoplasias/tratamento farmacológico , Oligodesoxirribonucleotídeos/uso terapêutico , Polissorbatos , Fator de Transcrição STAT3/metabolismo , Esqualeno
4.
Int J Mol Sci ; 21(16)2020 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-32823603

RESUMO

We explored the effect of a recombinant mucin1-maltose-binding protein vaccine, including immunization cycles of recombinant mucin1-maltose-binding protein (MUC1-MBP) and CpG 2006 on T cell responses to human MUC1-overexpressing mouse melanoma B16 cells (B16-MUC1) melanoma in mice. We found that the vaccine had a significant antitumor effect, with the most obvious tumor-suppressive effect being observed in mice immunized five times. After more than five immunizations, the tumor inhibition rate decreased from 81.67% (five immunizations) to 43.67% (eight immunizations). To study the possible mechanism, Mucin-1(MUC1)-specific antibodies, IFN-γ secretion by lymphocytes, and cytotoxic T lymphocyte (CTL) cytotoxicity were measured by enzyme-linked immunosorbent assay (ELISA) and a real-time cell analyzer (RTCA). T cell subsets and immunosuppressive cells in the mouse spleen and tumor microenvironment were analyzed by FACS. These results showed that five immunizations activated MUC1-specific Th1 and CTL and reduced the ratio of myeloid-derived suppressor cells (MDSCs) and Th17 in mice more significantly than eight immunizations, indicating that excessive frequency of the immune cycle leads to the increased numbers of immunosuppressive cells and decreased numbers of immunostimulatory cells, thereby inhibiting antitumor immune activity. This data provide an experimental foundation for the clinical application of a recombinant MUC1-MBP vaccine.


Assuntos
Imunização , Proteínas Ligantes de Maltose/imunologia , Melanoma Experimental/imunologia , Mucina-1/imunologia , Linfócitos T/imunologia , Vacinas Sintéticas/imunologia , Animais , Proliferação de Células , Modelos Animais de Doenças , Feminino , Humanos , Melanoma Experimental/prevenção & controle , Camundongos Endogâmicos C57BL , Baço/patologia , Microambiente Tumoral , Regulação para Cima
5.
Mol Med Rep ; 20(4): 2990-3002, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31432147

RESUMO

Gene expression and DNA methylation levels affect the outcomes of patients with cancer. The present study aimed to establish a multigene risk model for predicting the outcomes of patients with cervical cancer (CerC) treated with or without radiotherapy. RNA sequencing training data with matched DNA methylation profiles were downloaded from The Cancer Genome Atlas database. Patients were divided into radiotherapy and non­radiotherapy groups according to the treatment strategy. Differently expressed and methylated genes between the two groups were identified, and 8 prognostic genes were identified using Cox regression analysis. The optimized risk model based on the 8­gene signature was defined using the Cox's proportional hazards model. Kaplan­Meier survival analysis indicated that patients with higher risk scores exhibited poorer survival compared with patients with lower risk scores (log­rank test, P=3.22x10­7). Validation using the GSE44001 gene set demonstrated that patients in the high­risk group exhibited a shorter survival time comprared with the low­risk group (log­rank test, P=3.01x10­3). The area under the receiver operating characteristic curve values for the training and validation sets were 0.951 and 0.929, respectively. Cox regression analyses indicated that recurrence and risk status were risk factors for poor outcomes in patients with CerC treated with or without radiotherapy. The present study defined that the 8­gene signature was an independent risk factor for the prognosis of patients with CerC. The 8­gene prognostic model had predictive power for CerC prognosis.


Assuntos
Regulação Neoplásica da Expressão Gênica , Interleucina-8/biossíntese , Modelos Biológicos , Proteínas de Neoplasias/biossíntese , Neoplasias do Colo do Útero , Adulto , Intervalo Livre de Doença , Feminino , Humanos , Interleucina-8/genética , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Valor Preditivo dos Testes , Taxa de Sobrevida , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/mortalidade , Neoplasias do Colo do Útero/radioterapia
6.
Cell Immunol ; 325: 56-63, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29452695

RESUMO

Our previous study demonstrated that maltose-binding protein (MBP) activated Th1 through the TLR2-mediated MyD88-dependent pathway and the TLR4-mediated TRIF-dependent pathway. The combination of MBP and BCG synergistically induced Th1 activation, and the TLR2/9-mediated MyD88-dependent pathway is involved in this process. To further explore this mechanism, we stimulated purified mouse CD4+ T cells with MBP and BCG in vitro. The results demonstrated that MBP combined with BCG synergistically increased IFN-γ production and TLR2/4/9 expression, suggesting the involvement of TLR2/4/9 in the combination-induced Th1 activation. Next, TLRs 2/4/9 were blocked to analyze the effects of TLRs on Th1 activation. The results demonstrated that MBP induced a low level of Th1 activation by upregulating TLR2-mediated MyD88-TRAF6 and TLR4-mediated TRIF-TRAF3 expression, whereas MBP combined with BCG induced synergistic Th1 activation, which was not only triggered by strong upregulation of TLR2/9-mediated MyD88-TRAF6 expression but also by shifting TLR4-mediated TRIF-TRAF3 into the TRIF-TRAF6 pathway. Moreover, we observed that a TLR4 antibody upregulated MyD88 expression and a TLR9 inhibitor downregulated TRIF expression, indicating that there was cross-talk between TLRs 2/4/9 in MBP combined with BCG-induced Th1 activation. Our findings may expand the knowledge regarding TLR cross-talk involved in regulating the Th1 response.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Citocinas/metabolismo , Regulação para Baixo , Proteínas Ligantes de Maltose/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator 3 Associado a Receptor de TNF/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Células Th1/imunologia , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética , Ativação Transcricional , Regulação para Cima
7.
Int Immunopharmacol ; 50: 338-344, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28750349

RESUMO

MBP (maltose-binding protein) is a component of Escherichia coli. Our previous study found that MBP directly induces the activation of Th1 (T helper type 1), but the molecular mechanism remains unclear. In the present study, CD4+T cells were purified from the spleens of normal mice using antibody-coated immunomagnetic beads by negative selection. CD4+T cells activated with a CD3/CD28 antibody were stimulated with MBP. The results indicated that MBP elevated IFN-γ mRNA levels in activated CD4+T cells and promoted IFN-γ production from activated CD4+T cells. To explore TLR2/TLR4 signaling involved in the mechanism of MBP-induced activation of Th1, we further detected downstream molecules of TLR2/TLR4 signaling. We found that MBP increased the mRNA levels of MyD88, TRAF6, TRIF and TRAF3 expressed in CD4+T cells. The results suggested that downstream molecules of TLR2/TLR4 signaling may be involved in MBP-induced activation of CD4+T cells. Furthermore, MyD88, TRIF, TRAF3 and TRAF6 expressed in activated CD4+T cells blocked with anti-TLR2 antibody or anti-TLR4 antibody followed by treatment with MBP were detected via RT-PCR and western blotting, respectively. MBP decreased the production of IFN-γ in CD4+T cells in the presence of anti-TLR2, accompanied by the down-regulated expression of MyD88 and TRAF6. However, MBP increased the production of IFN-γ in CD4+T cells in the presence of anti-TLR4 antibody accompanied by the up-regulated expression of MyD88 and the down-regulated expression of TRIF, TRAF6 and TRAF3. The results suggested that the MyD88-dependent pathway of TLR2 and TRIF-dependent pathway are involved in the mechanism of Th1 activation induced by MBP. Our study has contributed to the clarification of the molecular mechanism of MBP-induced activation of CD4+T cells.


Assuntos
Infecções por Escherichia coli/imunologia , Proteínas de Escherichia coli/metabolismo , Escherichia coli/imunologia , Proteínas Ligantes de Maltose/metabolismo , Células Th1/imunologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Células Cultivadas , Proteínas de Escherichia coli/genética , Interferon gama/metabolismo , Ativação Linfocitária , Proteínas Ligantes de Maltose/genética , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais , Fator 3 Associado a Receptor de TNF/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Receptor 4 Toll-Like/metabolismo
8.
Mediators Inflamm ; 2017: 1953680, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28293065

RESUMO

To explore whether TLR2/TLR4 could be involved in the maturation of dendritic cells and polarization of CD4+ T cells induced by dendritic cells stimulated with MBP and BCG, in vitro and in vivo experiments using TLR2-/- or TLR4-/- mice were employed. MBP and BCG elevated CD80, CD86 and MHC class II expressed on dendritic cells and increased IL-12 protein, induced DC maturation, and indirectly promoted Th1 activation. Moreover, MBP and BCG upregulated costimulatory molecules on DCs in a TLR2- and TLR4-dependent manner. The levels of IFN-γ, IL-4, and IL-10 in CD4+ T cells cocultured with dendritic cells from different types of mice were determined with ELISPOT or ELISA method. TLR2/TLR4 is important in the maturation and activation of dendritic cells and the activation of Th1 cells induced by stimulation with MBP and BCG. In conclusion, TLR2 and TLR4 play an important role in the upregulation of costimulatory molecules and MHC class II molecules on dendritic cells and the activation of Th1 cells induced by stimulation with MBP and BCG. The results above indicate that the combination of MBP and BCG induced the maturation and activation of dendritic cells and promoted Th1 activation via TLR2/TLR4.


Assuntos
Células Dendríticas/metabolismo , Proteínas Ligantes de Maltose/farmacologia , Mycobacterium bovis/fisiologia , Células Th1/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Linfócitos T CD4-Positivos/metabolismo , Citometria de Fluxo , Interferon gama/metabolismo , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
9.
Mol Immunol ; 79: 32-37, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27693915

RESUMO

Our previous study demonstrated that maltose-binding protein (MBP) combined with BCG induced synergistic mouse Th1 activation in vivo. Here, to explore the mechanism of MBP combined with BCG on Th1 activation, mouse purified CD4+ T cells were stimulated with MBP and BCG in vitro. The results showed that MBP combined with BCG synergistically increased IFN-γ production, accompanied with the upregulation of TLR2/9 expressions, suggesting that TLR2/9 were involved in the combination-induced Th1 activation. Next, TLR2 antibodies and TLR9 inhibitor were used to further analyze the effects of TLRs in Th1 activation. Results showed TLR2 antibody partly decreased MBP combined with BCG-induced IFN-γ production, MyD88 expression and IκB phosphorylation, indicating that TLR2-mediated MyD88-dependent pathway was involved in the MBP combined with BCG-induced Th1 activation. Moreover, MBP combined with BCG-induced Th1 activation was completely abrogated by TLR9 inhibitor, suggesting that TLR9-mediated MyD88-dependent pathway played a more important role than TLR2 in the combination-induced Th1 activation. Further study showed that TLR9 inhibitor downregulated TLR2 expression, suggesting that TLR9 signaling regulated TLR2 activation to favor Th1 resonse induced by MBP combined with BCG. Collectively, we demonstrated for the first time that the cross-talk of TLR2 and TLR9 triggered Th1 activation collaboratively and our findings provided valuable information about designing more effective adjuvant for cancer therapy.


Assuntos
Ativação Linfocitária/imunologia , Células Th1/imunologia , Receptor 2 Toll-Like/imunologia , Receptor Toll-Like 9/imunologia , Animais , Western Blotting , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Proteínas Ligantes de Maltose/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium bovis/imunologia , Reação em Cadeia da Polimerase , Receptor Cross-Talk
10.
Int J Nanomedicine ; 10: 6075-87, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26491285

RESUMO

Recently, gold nanoparticles (AuNPs) have shown promising biological applications due to their unique electronic and optical properties. However, the potential toxicity of AuNPs remains a major hurdle that impedes their use in clinical settings. Mesoporous silica is very suitable for the use as a coating material for AuNPs and might not only reduce the cytotoxicity of cetyltrimethylammonium bromide-coated AuNPs but might also facilitate the loading and delivery of drugs. Herein, three types of rod-like gold-mesoporous silica nanoparticles (termed bare AuNPs, core-shell Au@mSiO2NPs, and Janus Au@mSiO2NPs) were specially designed, and the effects of these AuNPs on cellular uptake, toxic behavior, and mechanism were then systematically studied. Our results indicate that bare AuNPs exerted higher toxicity than the Au@mSiO2NPs and that Janus Au@mSiO2NPs exhibited the lowest toxicity in human breast cancer MCF-7 cells, consistent with the endocytosis capacity of the nanoparticles, which followed the order, bare AuNPs > core-shell Au@mSiO2NPs > Janus Au@mSiO2NPs. More importantly, the AuNPs-induced apoptosis of MCF-7 cells exhibited features that were characteristic of intracellular reactive oxygen species (ROS) generation, activation of c-Jun-N-terminal kinase (JNK) phosphorylation, an enhanced Bax-to-Bcl-2 ratio, and loss of the mitochondrial membrane potential. Simultaneously, cytochrome c was released from mitochondria, and the caspase-3/9 cascade was activated. Moreover, both ROS scavenger (N-acetylcysteine) and JNK inhibitor (SP600125) partly blocked the induction of apoptosis in all AuNPs-treated cells. Taken together, these findings suggest that all AuNPs induce apoptosis through the ROS-/JNK-mediated mitochondrial pathway. Thus, Janus Au@mSiO2NPs exhibit the potential for applications in biomedicine, thus aiding the clinical translation of AuNPs.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Ouro/química , Nanopartículas/administração & dosagem , Dióxido de Silício/química , Animais , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caspase 3/metabolismo , Caspase 9/metabolismo , Células Cultivadas , Citocromos c/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células MCF-7 , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Nanopartículas/química , Fosforilação/efeitos dos fármacos , Ratos , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
11.
Oncotarget ; 6(22): 19264-78, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26057631

RESUMO

Mucin1 (MUC1), as an oncogene, plays a key role in the progression and tumorigenesis of many human adenocarcinomas. In this study, wound-healing, transwell migration and matrigel invasion assays showed that MUC1 promotes human hepatocellular carcinoma (HCC) cell migration and invasion by MUC1 gene silencing and overexpressing. Treatment with exogenous transforming growth factor beta (TGF-ß)1, TGF-ß type I receptor (TßRI) inhibitor, TGF-ß1 siRNAs, or activator protein 1 (AP-1) inhibitor to MUC1-overexpressing HCC cells revealed that MUC1-induced autocrine TGF-ß via JNK/AP-1 pathway promotes the cell migration and invasion. In addition, the migration and invasion of HCC cells were more significantly inhibited by JNK inhibitor compared with that by TßRI inhibitor or TGF-ß1 siRNAs. Further studies demonstrated that MUC1-mediated JNK activation not only enhances the phosphorylation of Smad2 C-terminal at Ser-465/467 site (Smad2C) through TGF-ß/TßRI, but also directly enhances the phosphorylation of Smad2 linker region at Ser-245/250/255 site (Smad2L), and then both of them collaborate to upregulate matrix metalloproteinase (MMP)-9-mediated cell migration and invasion of HCC. These results indicate that MUC1 is an attractive target in liver cancer therapy.


Assuntos
Carcinoma Hepatocelular/genética , Movimento Celular/fisiologia , Neoplasias Hepáticas/metabolismo , Sistema de Sinalização das MAP Quinases , Mucina-1/metabolismo , Proteína Smad2/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Mucina-1/genética , Fosforilação , Proteína Smad2/genética , Transfecção
12.
Int J Mol Sci ; 16(5): 9896-909, 2015 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-25941931

RESUMO

Maltose-binding protein (MBP) is a critical player of the maltose/maltodextrin transport system in Escherichia coli. Our previous studies have revealed that MBP nonspecifically induces T helper type 1 (Th1) cell activation and activates peritoneal macrophages obtained from mouse. In the present study, we reported a direct stimulatory effect of MBP on RAW264.7 cells, a murine macrophage cell line. When stimulated with MBP, the production of nitric oxide (NO), IL-1ß, IL-6 and IL-12p70, and the expressions of CD80, MHC class II and inducible nitric oxide synthase (iNOS) were all increased in RAW264.7 cells, indicating the activation and polarization of RAW264.7 cells into M1 macrophages induced by MBP. Further study showed that MBP stimulation upregulated the expression of TLR2 and TLR4 on RAW264.7 cells, which was accompanied by subsequent phosphorylation of IκB-α and p38 MAPK. Pretreatment with anti-TLR2 or anti-TLR4 antibodies largely inhibited the phosphorylation of IκB-α and p38 MAPK, and greatly reduced MBP-induced NO and IL-12p70 production, suggesting that the MBP-induced macrophage activation and polarization were mediated by TLR2 and TLR4 signaling pathways. The observed results were independent of lipopolysaccharide contamination. Our study provides a new insight into a mechanism by which MBP enhances immune responses and warrants the potential application of MBP as an immune adjuvant in immune therapies.


Assuntos
Polaridade Celular/efeitos dos fármacos , Proteínas de Escherichia coli/farmacologia , Macrófagos/citologia , Macrófagos/metabolismo , Proteínas Periplásmicas de Ligação/farmacologia , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Biomarcadores/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Citocinas/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Mediadores da Inflamação/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Camundongos , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Pinocitose/efeitos dos fármacos , Células RAW 264.7 , Receptor 2 Toll-Like/efeitos dos fármacos , Receptor 4 Toll-Like/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Oncotarget ; 6(6): 4253-65, 2015 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-25714018

RESUMO

Mucin1 (MUC1) is a transmembrane glycoprotein that acts as an oncogene in human hepatic tumorigenesis. Hepatocellular carcinoma (HCC) cells often gain advantage by reducing the tumor-suppressive activity of transforming growth factor beta (TGF-ß) together with stimulation of its oncogenic activity as in MUC1 expressing HCC cells; however, molecular mechanisms remain largely unknown. Type I TGF-ß receptor (TßRI) and c-Jun NH2-terminal kinase (JNK) differentially phosphorylate Smad3 mediator to create 2 phosphorylated forms: COOH-terminally phosphorylated Smad3 (pSmad3C) and linker-phosphorylated Smad3 (pSmad3L). Here, we report that MUC1 overexpression in HCC cell lines suppresses TßRI-mediated pSmad3C signaling which involves growth inhibition by up-regulating p21(WAF1). Instead, MUC1 directly activates JNK to stimulate oncogenic pSmad3L signaling, which fosters cell proliferation by up-regulating c-Myc. Conversely, MUC1 gene silencing in MUC1 expressing HCC cells results in preserved tumor-suppressive function via pSmad3C, while eliminating pSmad3L-mediated oncogenic activity both in vitro and in vivo. In addition, high correlation between MUC1 and pSmad3L/c-Myc but not pSmad3C/p21(WAF1) expression was observed in HCC tissues from patients. Collectively, these results indicate that MUC1 shifts Smad3 signaling from a tumor-suppressive pSmad3C/p21(WAF1) to an oncogenic pSmad3L/c-Myc pathway by directly activating JNK in HCC cells, suggesting that MUC1 is an important target for HCC therapy.


Assuntos
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Mucina-1/metabolismo , Transdução de Sinais/fisiologia , Proteína Smad3/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Citometria de Fluxo , Imunofluorescência , Xenoenxertos , Humanos , Imunoprecipitação , MAP Quinase Quinase 4/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Confocal , Proteínas Proto-Oncogênicas c-myc/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
14.
Immunobiology ; 220(6): 782-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25601391

RESUMO

Maltose-binding protein (MBP), a component of the maltose transport system of Escherichia coli, has been commonly thought to have minimal bioactivity. Our previous studies demonstrated that MBP could significantly enhance Bacillus Calmette-Guerin (BCG)-induced T helper 1 (Th1) cell activation in mice. In the present study, we analyzed the effect of MBP on mouse T cells and found that MBP promoted the proliferation and IFN-γ production of CD4(+) T cells, suggesting that MBP directly induces Th1 activation. To explore the mechanism of Th1 activation, the expression of Toll-like receptor 2/4 (TLR2/4) on purified mouse CD4(+) T cells was detected. The results showed that MBP up-regulated TLR2 while down-regulated TLR4 expression, accompanied by a clear increase in MyD88 expression and IκB phosphorylation. Notably, the addition of anti-TLR2 antibody abrogated the MBP-induced CD4(+) T cells proliferation, IFN-γ secretion and MyD88 expression, whereas the addition of anti-TLR4 antibody exhibited a contradictive effect. Besides, the block of either TLR2 or TLR4 both reduced IκB phosphorylation. These results above suggest that TLR2-mediated MyD88-dependent pathway contributes to MBP-induced Th1 activation, while TLR4 appears to counteract this effect via MyD88-independent pathway.


Assuntos
Proteínas de Escherichia coli/imunologia , Ativação Linfocitária/imunologia , Proteínas Ligantes de Maltose/imunologia , Células Th1/imunologia , Células Th1/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Citocinas/metabolismo , Regulação da Expressão Gênica , Proteínas I-kappa B/metabolismo , Imunofenotipagem , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , Fosforilação , Baço/citologia , Baço/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética
15.
Int J Biochem Cell Biol ; 59: 116-25, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25526895

RESUMO

In a previous study, we observed by global gene expression analysis that oncogene mucin1 (MUC1) silencing decreased transforming growth factor beta (TGF-ß) signaling in the human hepatocellular carcinoma (HCC) cell line SMMC-7721. In this study, we report that MUC1 overexpression enhanced the levels of phosphorylated Smad3 linker region (p-Smad3L) (Ser-213) and its target gene MMP-9 in HCC cells, suggesting that MUC1 mediates TGF-ß signaling. To investigate the effect of MUC1 on TGF-ß signaling, we determined TGF-ß secretion in MUC1 gene silencing and overexpressing cell lines. MUC1 expression enhanced not only TGF-ß1 expression at the mRNA and protein levels but also luciferase activity driven by a TGF-ß promoter, as well as elevated the activation of c-Jun N-terminal kinase (JNK) and c-Jun, a member of the activation protein 1 (AP-1) transcription factor family. Furthermore, pharmacological reduction of TGF-ß receptor (TßR), JNK and c-Jun activity inhibited MUC1-induced autocrine TGF-ß signaling. Moreover, a co-immunoprecipitation assay showed that MUC1 directly bound and activated JNK. In addition, both MUC1-induced TGF-ß secretion and exogenous TGF-ß1 significantly increased Smad signaling and cell migration, which were markedly inhibited by either TßR inhibitor or small interfering RNA silencing of TGF-ß1 gene in HCC cells. The high correlation between MUC1 and TGF-ß1 or p-Smad3L (Ser-213) expression was shown in tumor tissues from HCC patients by immunohistochemical staining analysis. Collectively, these results indicate that MUC1 mediates autocrine TGF-ß signaling by activating the JNK/AP-1 pathway in HCC cells. Therefore, MUC1 plays a key role in HCC progression and could serve as an attractive target for HCC therapy.


Assuntos
Comunicação Autócrina , Carcinoma Hepatocelular/enzimologia , Neoplasias Hepáticas/enzimologia , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Mucina-1/metabolismo , Fator de Transcrição AP-1/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Movimento Celular , Ativação Enzimática , Técnicas de Silenciamento de Genes , Inativação Gênica , Células HEK293 , Humanos , Neoplasias Hepáticas/patologia , Mucina-1/genética , Fosforilação , Ligação Proteica , Proteína Smad3/metabolismo
16.
Int Immunopharmacol ; 21(1): 171-80, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24825603

RESUMO

Maltose-binding protein (MBP) is a component of the maltose transport system of Escherichia coli. Our previous study found that MBP combined with Bacillus Calmette-Guerin (BCG) increases the percentage of activated macrophages in the spleen and the pinocytic activity of peritoneal macrophages in vivo. However, the effect of MBP alone on macrophages remains unclear. In the present study, the results showed that MBP enhanced LPS-stimulated macrophage activity in vivo. Subsequently, we investigated the regulatory effect of MBP on mouse peritoneal macrophages in vitro and the possible underlying mechanism. The results showed that MBP directly promoted macrophage phagocytic activity and increased the production of NO, IL-1ß and IL-6. Notably, macrophage phenotypic analysis showed that MBP significantly increased iNOS, IL-12p70 and CD16/32. In contrast, MBP decreased the secretion of IL-10 and slightly decreased Arg-1 mRNA and CD206 protein expression. These results suggested that MBP activated macrophages and polarized them into M1 macrophages. Further study found that MBP directly bound to macrophages and upregulated TLR2 mRNA expression. This process was accompanied by a clear increase in MyD88 expression and phosphorylation of p38 MAPK and IκB-α, but these effects were largely abrogated by pretreatment with anti-TLR2 or anti-TLR4 antibodies. The effects of MBP on macrophage NO production were also partially inhibited by anti-TLR2 and/or anti-TLR4 antibodies. Furthermore, the effect of MBP on IL-12 and IL-10 secretion was largely influenced by the NF-κB inhibitor PDTC and the p38 MAPK inhibitor SB203580. These results suggest that MBP directly activates macrophages and induces M1 polarization through a process that may involve TLR2 and TLR4.


Assuntos
Escherichia coli/metabolismo , Macrófagos Peritoneais/imunologia , Proteínas Ligantes de Maltose/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Anticorpos Bloqueadores/farmacologia , Antígenos CD/metabolismo , Arginase/genética , Arginase/metabolismo , Diferenciação Celular , Células Cultivadas , Citocinas/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Imidazóis/farmacologia , Técnicas In Vitro , Macrófagos Peritoneais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/antagonistas & inibidores , Óxido Nítrico/metabolismo , Fagocitose , Prolina/análogos & derivados , Prolina/farmacologia , Piridinas/farmacologia , Tiocarbamatos/farmacologia , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/imunologia , Receptor 4 Toll-Like/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
17.
Oncol Rep ; 31(6): 2811-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24737121

RESUMO

Mucin1 (MUC1) is a transmembrane glycoprotein that plays a key role as an oncogene in the tumorigenesis of many human adenocarcinomas. However, the role of MUC1 in human hepatocellular carcinoma (HCC) progression remains unclear. In the present study, we silenced MUC1 to investigate its effect on the human HCC cell line SMMC-7721 and found that knockdown of MUC1 significantly inhibited cell proliferation, enhanced cell-cell aggregation and induced apoptosis. No significant differences were found in in vitro migration or invasion. We also observed that knockdown of MUC1 decreased the translocation of ß­catenin to the nucleus, reduced the activity of T cell factor and blocked the expression of cyclin D1 and c-Myc. In addition, MUC1 knockdown enhanced the expression of E-cadherin, a molecular chaperone of ß­catenin that plays an important role in cell-cell aggregation. In vivo assays demonstrated that there was no tumor growth in mice injected with MUC1-silenced cells. Global gene expression analysis showed that a series of genes encoding molecules in the Wnt/ß­catenin, nuclear factor-κB (NF-κB), mitogen-activated protein kinase (MAPK), insulin, transforming growth factor ß (TGF-ß) and vascular endothelial growth factor (VEGF) signaling pathways were all influenced by the knockdown of MUC1, and these may contribute to the phenotypic alterations observed. Collectively, our results indicate that MUC1 plays a key role in HCC tumorigenesis.


Assuntos
Carcinoma Hepatocelular/genética , Transformação Celular Neoplásica , Neoplasias Hepáticas/genética , Mucina-1/genética , Animais , Apoptose/genética , Caderinas , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...