Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Int J Mol Sci ; 23(18)2022 Sep 17.
Article in English | MEDLINE | ID: mdl-36142800

ABSTRACT

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.


Subject(s)
Cancer Vaccines , Neoplasms , Adjuvants, Immunologic/pharmacology , Animals , Antigens , Dendritic Cells , Interleukin-6 , Maltose-Binding Proteins , Mice , Mice, Inbred C57BL , Mucin-1/genetics , Neoplasms/drug therapy , Oligodeoxyribonucleotides/therapeutic use , Polysorbates , STAT3 Transcription Factor/metabolism , Squalene
2.
Int J Mol Sci ; 21(16)2020 Aug 13.
Article in English | MEDLINE | ID: mdl-32823603

ABSTRACT

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.


Subject(s)
Immunization , Maltose-Binding Proteins/immunology , Melanoma, Experimental/immunology , Mucin-1/immunology , T-Lymphocytes/immunology , Vaccines, Synthetic/immunology , Animals , Cell Proliferation , Disease Models, Animal , Female , Humans , Melanoma, Experimental/prevention & control , Mice, Inbred C57BL , Spleen/pathology , Tumor Microenvironment , Up-Regulation
3.
Cell Immunol ; 325: 56-63, 2018 03.
Article in English | MEDLINE | ID: mdl-29452695

ABSTRACT

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.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cytokines/metabolism , Down-Regulation , Maltose-Binding Proteins/metabolism , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , Signal Transduction/drug effects , TNF Receptor-Associated Factor 3/metabolism , TNF Receptor-Associated Factor 6/metabolism , Th1 Cells/immunology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Transcriptional Activation , Up-Regulation
4.
Int J Mol Sci ; 19(3)2018 Mar 20.
Article in English | MEDLINE | ID: mdl-29558459

ABSTRACT

Mucin 1 (MUC1), being an oncogene, is an attractive target in tumor immunotherapy. Maltose binding protein (MBP) is a potent built-in adjuvant to enhance protein immunogenicity. Thus, a recombinant MUC1 and MBP antitumor vaccine (M-M) was constructed in our laboratory. To enhance the antitumor immune activity of M-M, CpG oligodeoxynucleotides 1826 (CpG 1826), a toll-like receptor-9 agonist, was examined in this study as an adjuvant. The combination of M-M and CpG 1826 significantly inhibited MUC1-expressing B16 cell growth and prolonged the survival of tumor-bearing mice. It induced MUC1-specific antibodies and Th1 immune responses, as well as the Cytotoxic T Lymphocytes (CTL) cytotoxicity in vivo. Further studies showed that it promoted the maturation and activation of the dendritic cell (DC) and skewed towards Th1 phenotype in vitro. Thus, our study revealed that CpG 1826 is an efficient adjuvant, laying a foundation for further M-M clinical research.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Mucin-1/immunology , Oligodeoxyribonucleotides/pharmacology , Animals , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Mice , Mice, Inbred C57BL , Th1 Cells/immunology , Toll-Like Receptor 9/agonists
5.
Cancer Sci ; 108(3): 504-511, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28012230

ABSTRACT

Mucin 1 (MUC1), as an oncogene, is overexpressed in hepatocellular carcinoma (HCC) cells and promotes the progression and tumorigenesis of HCC through JNK/TGF-ß signaling pathway. In the present study, RNA interference (RNAi) and JNK inhibitor SP600125, which target MUC1 and/or JNK, were used to treat HCC cells in vitro, and the results showed that both silencing the expression of MUC1 and blocking the activity of JNK inhibited the proliferation of HCC cells. In addition, MUC1-stable-knockdown and SP600125 significantly inhibited the growth of tumors in the subcutaneous transplant tumor models that established in BALB/c nude mice rather than MUC1 or JNK siRNAs transiently transfection. Furthermore, the results from immunohistochemical staining assays showed that the inhibitory effects of MUC1 gene silencing and SP600125 on the proliferation of HCC cells in vivo were through the JNK/TGF-ß signaling pathway. These results indicate that MUC1 and JNK are attractive targets for HCC therapy and may provide new therapeutic strategies for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , Liver Neoplasms/pathology , Mucin-1/genetics , RNA Interference , Animals , Anthracenes/pharmacology , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Neoplasms/genetics , MAP Kinase Signaling System , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Small Interfering/genetics , Transforming Growth Factor beta/metabolism
6.
Mediators Inflamm ; 2017: 1953680, 2017.
Article in English | MEDLINE | ID: mdl-28293065

ABSTRACT

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.


Subject(s)
Dendritic Cells/metabolism , Maltose-Binding Proteins/pharmacology , Mycobacterium bovis/physiology , Th1 Cells/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Flow Cytometry , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
Int J Mol Sci ; 16(5): 9896-909, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25941931

ABSTRACT

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.


Subject(s)
Cell Polarity/drug effects , Escherichia coli Proteins/pharmacology , Macrophages/cytology , Macrophages/metabolism , Periplasmic Binding Proteins/pharmacology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , Biomarkers/metabolism , Cell Survival/drug effects , Cytokines/metabolism , Histocompatibility Antigens Class I/metabolism , Inflammation Mediators/metabolism , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Macrophages/enzymology , Mice , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Pinocytosis/drug effects , RAW 264.7 Cells , Toll-Like Receptor 2/drug effects , Toll-Like Receptor 4/drug effects , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Int Immunopharmacol ; 121: 110487, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37364328

ABSTRACT

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.


Subject(s)
Cancer Vaccines , Melanoma , Mice , Animals , T-Lymphocytes, Regulatory , Immunization , Melanoma/drug therapy , Immune Tolerance
9.
Mol Clin Oncol ; 17(6): 161, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36479256

ABSTRACT

Mucin 1 (MUC1) was the first discovered transmembrane protein of the mucin family; it normally covers epithelial cells of the mucous membrane, providing lubrication and protection. However, aberrant expression of MUC1 is involved in cancer development, invasion and metastasis. It has been reported that MUC1 upregulation is highly associated with the progression of different epithelial cancer types, such as lung, liver, pancreatic and breast cancer. Therefore, MUC1 can be used as a specific marker and a target for immunotherapy in clinical applications, and the detection of MUC1 expression levels can be used to diagnose the occurrence, metastasis, prognosis and recurrence of cancer. The present review summarizes the abnormal expression of MUC1 in different tumours and discusses its clinical significance, thereby highlighting the potential diagnostic and therapeutic significance of MUC1 in cancer.

10.
Int Immunopharmacol ; 107: 108667, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35255300

ABSTRACT

Tumor necrosis factor receptor (TNFR)-associated factor 6 (TRAF6) signaling is a critical positive mechanism for the development, homeostasis and activation of immune cells. We investigated the effect of TRAF6 overexpression on dendritic cells (DCs) maturation. TRAF6-overexpressing DCs had increased expression of costimulatory molecules, major histocompatibility complex (MHC) molecules and IL-12 expression. This indicated that TRAF6 promoted the maturation of DCs and indirectly promoted Th1 activation. The antitumor activities between TRAF6-overexpressing DCs and control DCs were compared by administering DCs pulsed with mucin 1 (MUC1) Ag peptide in a therapeutic human MUC1-overexpressing mouse B16 melanoma cells (B16-MUC1) model. Administration of TRAF6-overexpressing DCs significantly inhibited the growth of B16-MUC1 tumors, accompanied by an increase in MUC1-specific Th1 responses and Tc1 responses, as well as a decrease in Tregs levels. TRAF6 signaling has been found to be involved in DCs maturation and Th1 activation in vitro, as well as therapeutic tumor models in vivo, indicating that TRAF6-overexpressing DCs may be a promising approach for cancer immunotherapy.


Subject(s)
Cancer Vaccines , Melanoma, Experimental , Animals , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Dendritic Cells , Mice , Mice, Inbred C57BL , Mucin-1 , TNF Receptor-Associated Factor 6/metabolism
11.
Immunol Lett ; 245: 38-50, 2022 05.
Article in English | MEDLINE | ID: mdl-35405170

ABSTRACT

In this study, we explored the initiation and regulation mechanism of antigen-specific CTL responses induced by a novel cancer vaccine containing recombinant human mucin1-maltose-binding protein fusion protein (MUC1-MBP) and CpG2006. First, DC subsets were analyzed by flow cytometry in vivo and in vitro. After vaccination, the proportion and maturation of cDC1s in mouse dLNs were upregulated, and the proportion of cDC2s and pDCs was also increased. In vitro studies on vaccine components showed similar changs, which may mainly depend on the activity of CpG2006. Subsequently, the regulatory effect of type Ⅰ IFN signaling on CTL triggering was confirmed through co-culture of sorted DC subsets and T cells and subsequent CTL activity experiments. CTL killing activity exhibited a 61.9% decrease once type I IFN signaling was blocked. Further analysis showed that blocking IFNAR1 on cDC1s but not on CTLs resulted in significant defects in CTL killing activity. Collectively, M-M combined with CpG2006 vaccine promotes MUC1-specific CTL responses by increasing the cDC1 activity in mice, and this is mainly regulated by type Ⅰ IFN signaling in cDC1s.


Subject(s)
Cancer Vaccines , Cross-Priming , Animals , Dendritic Cells , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic
12.
Oncol Lett ; 21(2): 149, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33552267

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are one of the major components of the tumor microenvironment (TME), and are the main mediators of tumor-induced immunosuppression. Recent studies have reported that the survival, differentiation and immunosuppressive activity of MDSCs are affected by the Toll-like receptor (TLR) signaling pathway. However, the regulatory effect of TLR signaling on MDSCs remains controversial. TLR-induced MDSC can acquire different immunosuppressive activities to influence the immune response that can be either beneficial or detrimental to cancer immunotherapy. The present review summarizes the effects of TLR signals on the number, phenotype and inhibitory activity of MDSCs, and their role in cancer immunotherapy, which cannot be ignored if effective cancer immunotherapies are to be developed for the immunosuppression of the TME.

13.
Int Immunopharmacol ; 101(Pt A): 108173, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34607233

ABSTRACT

In previous studies, we have obtained a notable anti-tumor efficacy of the recombinant MUC1-MBP vaccine in the process of mouse B16-MUC1 melanoma treatment. However, the tumor cannot be eliminated completely. We found that the tumor inhibition rate decreased from 81.67% (five immunizations) to 43.67% (eight immunizations) after more than five immunizations, indicating persistent vaccine stimulation may activate immunosuppressive factors. In the present study, we revealed that programmed cell death 1 (PD1), an inhibitory molecule suppressing T cell function, expressed on splenic and tumor-infiltrating T cells were up-regulated by the vaccine. Therefore, to optimize the anti-tumor efficacy of the vaccine, we employed combination immunotherapy with MUC1-MBP vaccine and αPD1 (anti-PD1 antibody). Results showed that combination immunotherapy induced a more remarkable anti-tumor efficacy, the tumor clearance being increased to 80% from 20% which obtain by MUC1-MBP vaccine immunizations. To investigate the possible underlying mechanism, IFN-γ secretion and cytotoxic T lymphocyte (CTL) cytotoxicity were measured by enzyme-linked immunosorbent assay (ELISA) and xCELLigence real-time cell analyzer (RTCA) respectively. T cell subsets and immunosuppressive cells in the mouse spleen and tumor microenvironment were analyzed by FACS. Results showed that the proportion of splenic CD8+T cells and tumor infiltration was increased and the activity of CTL killing, T helper 1 (Th1), Type 1 CD8+T (Tc1) was enhanced, indicating that the anti-tumor efficacy enhanced by combination immunotherapy was mainly through boosting CD8+T cells mediated anti-tumor cellular immunity. Additionally, combination immunotherapy significantly decreased the splenic and tumor-infiltrating myeloid derived suppressor cells (MDSCs). These results demonstrated that combination immunotherapy with MUC1-MBP vaccine and αPD1 was capable to invoke a more potent anti-tumor immune response and provide a foundation for further research.


Subject(s)
Cancer Vaccines/administration & dosage , Immune Checkpoint Inhibitors/pharmacology , Melanoma, Experimental/therapy , Skin Neoplasms/therapy , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor/transplantation , Female , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mucin-1/administration & dosage , Mucin-1/genetics , Mucin-1/immunology , Myelin Basic Protein/administration & dosage , Myelin Basic Protein/genetics , Myelin Basic Protein/immunology , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Th1 Cells/drug effects , Th1 Cells/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
14.
Biomed Res Int ; 2020: 4607197, 2020.
Article in English | MEDLINE | ID: mdl-33294443

ABSTRACT

Tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6), an E3 ubiquitin ligase, is a signal transduction molecule shared by the interleukin-1 receptor (IL-1R)/Toll-like receptor (TLR) family and the TNFR superfamily. TRAF6 has a unique TRAF domain and RING finger domain that mediate intracellular signaling events. In the immune system, TRAF6-mediated signaling has been shown to be critical for the development, homeostasis, and activation of a variety of immune cells, including B cells, T cells, dendritic cells, and macrophages. Although the pathogenesis and etiology of autoimmune diseases and cancer are not fully understood, it is worth noting that existing studies have shown that TRAF6 is involved in the pathogenesis and development of a variety of these diseases. Herein, we reviewed the role of TRAF6 in certain immune cells, as well as the function and potential effect of TRAF6 in autoimmune diseases and cancer. Our review indicates that TRAF6 may be a novel target for autoimmune diseases and cancer.


Subject(s)
Autoimmune Diseases/metabolism , Neoplasms/metabolism , TNF Receptor-Associated Factor 6/metabolism , Humans , Lymphocytes/immunology , Neoplasms/immunology , Proteasome Endopeptidase Complex/metabolism , TNF Receptor-Associated Factor 6/chemistry , TNF Receptor-Associated Factor 6/genetics , Ubiquitination
15.
Cell Immunol ; 255(1-2): 69-75, 2009.
Article in English | MEDLINE | ID: mdl-19091310

ABSTRACT

Activin A is a multifunctional factor of the transforming growth factor-beta (TGF-beta) superfamily and acts as an anti-inflammatory cytokine produced by microglia and macrophages. In this study, we investigated the regulatory effect and possible mechanism of activin A on activation of lipopolysaccharide (LPS)-induced mouse peritoneal macrophages. The results showed that activin A could decrease NO release in LPS-activated mouse peritoneal macrophages, and suppressed phagocytosis and pinocytosis of mouse peritoneal macrophages stimulated by LPS in vitro and in vivo. Furthermore, activin A remarkably inhibited the expressions of CD14 and MHC II on LPS-induced mouse peritoneal macrophages, but had no significant effect on the expression of MHC I and the proliferation of mouse peritoneal macrophages. These findings suggest that activin A can down-regulate inflammatory mediator production and phagocytosis of LPS-activated macrophages via suppressing CD14 expression, and may influence the presentation of exogenous antigens via inhibiting MHC II expression. Thus, activin A might have the potential for treatment of macrophage-mediated inflammatory diseases through modulating both innate and adaptive immune responses.


Subject(s)
Activins/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Phagocytosis/physiology , Activins/genetics , Animals , Cells, Cultured , Female , Genes, MHC Class I , Genes, MHC Class II , HLA Antigens/genetics , HLA Antigens/immunology , Humans , Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/immunology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Pinocytosis/physiology
16.
Gen Comp Endocrinol ; 161(2): 276-82, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19523381

ABSTRACT

Activin plays important roles in reproductive tissues as a stimulator of follicle-stimulating hormone (FSH) secretion. Activin receptor-interacting protein 2 (ARIP2) has been recently identified in mouse tissues as a regulatory protein of activin signal transduction. However, the localization and function of ARIP2 are not well characterized. In this study, we found that ARIP2 mRNA and protein were widely expressed in mouse tissues by reverse transcription-PCR (RT-PCR) and Western blotting. The immunoreactivities of ARIP2 were mainly localized at myocardial cells of heart, Leydig cells in testis, macrophages and epithelial cells of bronchus in lung, renal tubule and collecting tubule, pancreatic islet, adrenal gland, adenohypophysis and hypothalamus by immunohistochemical staining. Furthermore, ARIP2 overexpression down-regulated signal transduction induced by activin A in pituitary gonadotroph LbetaT2 cells and inhibited FSH secretion from primary cultured anterior pituitary cells induced by activin A. These findings suggest that ARIP2 is widely distributed in various tissues and may be a negative regulator of activin action in pituitary cells.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Gene Expression Regulation , Membrane Proteins/physiology , Activins/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adrenal Glands/metabolism , Animals , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Follicle Stimulating Hormone/metabolism , Hypothalamus/metabolism , Immunohistochemistry , In Vitro Techniques , Kidney/metabolism , Lung/metabolism , Macrophages/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Testis/metabolism
17.
Mol Med Rep ; 20(4): 2990-3002, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31432147

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Neoplastic , Interleukin-8/biosynthesis , Models, Biological , Neoplasm Proteins/biosynthesis , Uterine Cervical Neoplasms , Adult , Disease-Free Survival , Female , Humans , Interleukin-8/genetics , Middle Aged , Neoplasm Proteins/genetics , Predictive Value of Tests , Survival Rate , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/mortality , Uterine Cervical Neoplasms/radiotherapy
18.
Cytokine ; 42(1): 85-91, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18321725

ABSTRACT

Activin A is a member of transforming growth factor beta (TGF-beta) superfamily, which is also named restrictin-P, and can inhibit the secretion of nitric oxide (NO) and interleukin-1beta (IL-1beta) from LPS-activated mouse macrophages. In this study, the regulation effect and possible mechanism of activin A as an anti-inflammatory factor on lipopolysaccharide (LPS)-activated macrophages were investigated in vitro. It was observed that activin A could not only decrease the secretion of IL-1beta and NO, as well as the mRNA expressions of IL-1beta and iNOS, but also suppress the pinocytosis of mouse macrophage cell line RAW264.7 cells induced by LPS. In addition, activin A could obviously reduce the expressions of CD68 and CD14, as well as Toll-like receptor 4 (TLR4) on RAW264.7 cells induced by LPS, but could not influence the proliferation of RAW264.7 cells. These findings suggest that activin A may play an important down-regulation role in inflammatory factor production and phagocytosis of the activated macrophages via suppressing the maturation of LPS-induced macrophages or LPS-TLR4 signal transduction.


Subject(s)
Activins/metabolism , Lipopolysaccharides/immunology , Macrophage Activation , Macrophages/metabolism , Activins/genetics , Animals , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Cell Line , Cell Proliferation , Humans , Interleukin-1beta/immunology , Lipopolysaccharide Receptors/immunology , Macrophages/cytology , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/immunology , Pinocytosis/physiology , Signal Transduction/physiology
19.
World J Gastroenterol ; 13(41): 5501-5, 2007 Nov 07.
Article in English | MEDLINE | ID: mdl-17907296

ABSTRACT

AIM: To investigate the regulation of activin receptor-interacting protein 2 (ARIP2) expression and its possible relationships with collagen type IV (collagen IV) in mouse hepatoma cell line Hepal-6 cells. METHODS: The ARIP2 mRNA expression kinetics in Hepal-6 cells was detected by RT-PCR, and its regulation factors were analyzed by treatment with signal transduction activators such as phorbol 12-myristate 13-acetate (PMA), forskolin and A23187. After pcDNA3-ARIP2 was transfected into Hepal-6 cells, the effects of ARIP2 overexpression on activin type II receptor (ActRII) and collagen IV expression were evaluated. RESULTS: The expression levels of ARIP2 mRNA in Hapel-6 cells were elevated in time-dependent manner 12 h after treatment with activin A and endotoxin LPS, but not changed evidently in the early stage of stimulation (2 or 4 h). The ARIP2 mRNA expression was increased after stimulated with signal transduction activators such as PMA and forskolin in Hepal-6 cells, whereas decreased after treatment with A23187 (25.3% +/- 5.7% vs 48.1% +/- 3.6%, P < 0.01). ARIP2 overexpression could remarkably suppress the expression of ActRIIA mRNA in dose-dependent manner, but has no effect on ActRIIB in Hepal-6 cells induced by activin A. Furthermore, we have found that overexpression of ARIP2 could inhibit collagen IV mRNA and protein expressions induced by activin A in Hapel-6 cells. CONCLUSION: These findings suggest that ARIP2 expression can be influenced by various factors. ARIP2 may participate in the negative feedback regulation of signal transduction in the late stage by affecting the expression of ActRIIA and play an important role in regulation of development of liver fibrosis induced by activin.


Subject(s)
Activin Receptors, Type II/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Hepatocellular/metabolism , Collagen Type IV/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Membrane Proteins/metabolism , Signal Transduction , Activin Receptors, Type II/genetics , Activins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adenylyl Cyclases/metabolism , Animals , Calcimycin/pharmacology , Calcium/metabolism , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Colforsin/pharmacology , Collagen Type IV/genetics , Enzyme Activators/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Ionophores/pharmacology , Kinetics , Lipopolysaccharides/pharmacology , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Membrane Proteins/genetics , Mice , Protein Kinase C/metabolism , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Tetradecanoylphorbol Acetate/pharmacology , Transfection
20.
Int Immunopharmacol ; 50: 338-344, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28750349

ABSTRACT

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.


Subject(s)
Escherichia coli Infections/immunology , Escherichia coli Proteins/metabolism , Escherichia coli/immunology , Maltose-Binding Proteins/metabolism , Th1 Cells/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cells, Cultured , Escherichia coli Proteins/genetics , Interferon-gamma/metabolism , Lymphocyte Activation , Maltose-Binding Proteins/genetics , Mice , Myeloid Differentiation Factor 88/metabolism , Signal Transduction , TNF Receptor-Associated Factor 3/metabolism , TNF Receptor-Associated Factor 6/metabolism , Toll-Like Receptor 4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL