Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Gen Physiol Biophys ; 38(4): 335-342, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31219428

ABSTRACT

Ultraviolet-inactivated Sendai virus strain Tianjin (UV-Tianjin) has been proved to have antitumor effects in many kinds of tumor cells. Here, we investigated the anticancer properties of UV-Tianjin on human osteosarcoma HOS cells and the underlying molecular mechanism. Apoptosis, intracellular reactive oxygen species (ROS) levels and mitochondrial membrane potential were determined by flow cytometry analysis. The expression levels of apoptosis-related proteins were tested by western blotting. The results showed that UV-Tianjin concentration-dependently induced apoptosis in HOS cells. UV-Tianjin-induced apoptosis was mediated by the mitochondrial pathway, which was confirmed by mitochondrial dysfunction, downregulation of B-cell lymphoma 2 (Bcl-2), B-cell lymphoma-xL (Bcl-xL) and myeloid cell leukemia-1 (Mcl-1), upregulation of B-cell lymphoma 2 associated X protein (Bax) and Bcl-2 Homologous Antagonist/Killer (Bak), as well as the cleavage of caspase-9 and -3. Further analysis showed that UV-Tianjin augmented the phosphorylation of c-Jun N-terminal kinase, the extracellular-regulated kinase and p38, the major components of mitogen-activated protein kinase (MAPK) pathways, as well as the generation of ROS. Moreover, UV-Tianjin-induced apoptosis was remarkably attenuated by MAPK inhibitors and ROS inhibitor. Taken together, our results indicated that UV-Tianjin exerts antitumor effects by inducing mitochondria-dependent apoptosis involving ROS generation and MAPK pathway in human osteosarcoma HOS cells.


Subject(s)
Apoptosis , Mitochondria/metabolism , Oncolytic Virotherapy , Osteosarcoma/therapy , Osteosarcoma/virology , Sendai virus/classification , Sendai virus/radiation effects , Cell Line, Tumor , Humans , MAP Kinase Signaling System , Membrane Potential, Mitochondrial , Mitochondria/enzymology , Osteosarcoma/pathology , Reactive Oxygen Species/metabolism
2.
J Cell Physiol ; 234(4): 4179-4190, 2019 04.
Article in English | MEDLINE | ID: mdl-30146726

ABSTRACT

Sendai virus strain Tianjin, a novel genotype of Sendai virus, has been proven to possess potent antitumor effect on certain cancer cell types although inactivated by ultraviolet (UV). This study was carried out to investigate the in vitro anticancer properties of UV-inactivated Sendai virus strain Tianjin (UV-Tianjin) on human osteosarcoma cells and the underlying molecular mechanism. Our studies demonstrated UV-Tianjin significantly inhibited the viability of human osteosarcoma cell lines and triggered apoptosis through activation of both extrinsic and intrinsic pathways in MG-63 cells. Meanwhile, autophagy occurred in UV-Tianjin-treated cells. Blockade of autophagy with 3-methyladenine remarkably attenuated the inhibition of cell proliferation by UV-Tianjin, suggesting that UV-Tianjin-induced autophagy may be contributing to cell death. Furthermore, UV-Tianjin induced reactive oxygen species (ROS) production, which was involved in the execution of MG-63 cell apoptosis and autophagy, as evidenced by the result that treatment of N-acetyl-L-cysteine, a ROS scavenger, attenuated both apoptosis and autophagy. In addition, inhibition of apoptosis promoted autophagy, whereas suppression of autophagy attenuated apoptosis. Our results suggest that UV-Tianjin triggers apoptosis and autophagic cell death via generation of the ROS in MG-63 cells, which might provide important insights into the effectiveness of novel strategies for osteosarcoma therapy.


Subject(s)
Apoptosis , Autophagy , Bone Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses , Osteosarcoma/therapy , Oxidative Stress , Reactive Oxygen Species/metabolism , Sendai virus , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Proteins/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/virology , Cell Line, Tumor , Cell Proliferation , Host-Pathogen Interactions , Humans , Oncolytic Viruses/radiation effects , Osteosarcoma/metabolism , Osteosarcoma/pathology , Osteosarcoma/virology , Sendai virus/radiation effects , Ultraviolet Rays
3.
Asian Pac J Cancer Prev ; 15(12): 5023-8, 2014.
Article in English | MEDLINE | ID: mdl-24998581

ABSTRACT

Sendai virus strain Tianjin is a novel genotype. Here, we investigate the antitumor and proapoptotic effects of ultraviolet-inactivated Sendai virus strain Tianjin (UV-Tianjin) on human breast cancer MDA-MB-231 cells in vitro, as well as the involvement of the apoptotic pathway in the mechanism of UV-Tianjin-induced antitumor effects. MTT assays showed that treatment with UV-Tianjin dose-dependently inhibited the proliferation of MDA- MB-231 cells but not normal MCF 10A breast epithelium cells. Hoechst staining and flow cytometric analysis revealed that UV-Tianjin induced apoptosis of MDA-MB-231 cells in a dose-dependent manner. Moreover, UV-Tianjin treatment resulted in reduction in the mitochondria membrane potential (MMP) and release of cytochrome complex (cyt c) via regulation of Bax and Bcl-2, as well as activation of caspase-9, caspase-3, Fas, FasL and caspase-8 in MDA-MB-231 cells. In summary, our study suggests that UV-Tianjin exhibits anticancer activity in human breast cancer MDA-MB-231 cells through inducing apoptosis, which may involve both the endogenous mitochondrial and exogenous death receptor pathways.


Subject(s)
Apoptosis , Breast Neoplasms/pathology , Cell Proliferation , Sendai virus/physiology , Blotting, Western , Breast Neoplasms/immunology , Female , Flow Cytometry , Humans , Membrane Potential, Mitochondrial , Sendai virus/radiation effects , Tumor Cells, Cultured , Virus Inactivation/radiation effects
4.
J Transl Med ; 11: 205, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-24007528

ABSTRACT

BACKGROUND: Ultraviolet-inactivated, replication-defective Sendai virus particles (Z strain) have displayed antitumor effect through enhancing the immune responses or inducing apoptosis in a variety of carcinomas. Sendai virus strain Tianjin was isolated from the lungs of marmoset and proved to be a novel genotype of Sendai virus. In this study, we explored the antitumor effect and its mechanism of ultraviolet-inactivated, replication-defective Sendai virus strain Tianjin (UV-Tianjin) in mice bearing CT26 colon carcinoma. METHODS: Three injections of UV-Tianjin were delivered into CT26 tumors growing on the back of BALB/c mice. Tumor size was measured in a blinded manner and survival rate of mice was calculated. In order to make clear antitumor mechanism of UV-Tianjin, the maturation and interleukin-6 (IL-6) release from murine myeloid dendritic cells (DCs) was examined by flow cytometry or ELISA assay after induced by UV-Tianjin and compared with those of live virus. Moreover, real-time RT-PCR and immunohistochemistry was performed to identify whether UV-Tianjin could induce infiltration of DCs, CD4⁺ and CD8⁺ T cells into tumors. The TUNEL assay was done to observe the apoptosis of CT26 tumor cells after UV-Tianjin injection. RESULTS: In animal model, UV-Tianjin could obviously inhibit the growth of CT26 tumors and prolong the survival of the tumor-bearing mice compared with control group (P < 0.01). In vitro murine DCs stimulated by UV-Tianjin underwent dose-dependent maturation, similar to that elicited by live virus. And the secretion amount of IL-6 from DCs induced by UV-Tianjin was a little lower than that released in the presence of live virus. Real-time RT-PCR and immunohistochemistry revealed that UV-Tianjin induced a remarkable infiltration of DCs, CD4⁺ and CD8⁺ T cells into tumors. The TUNEL assay showed that the apoptosis index of tumor tissues injected with UV-Tianjin was significantly higher than that of control group (P < 0.01). CONCLUSIONS: Our results have demonstrated that UV-Tianjin alone could inhibit the growth of CT26 tumor in mice through enhancing host antitumor immunity and inducing apoptosis of tumor cells. Therefore, UV-Tianjin shows its prospect as a novel drug for carcinoma therapy.


Subject(s)
Apoptosis/immunology , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Immunity/immunology , Sendai virus/genetics , Sendai virus/physiology , Virus Inactivation , Animals , Cell Line, Tumor , Cell Proliferation , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Genotype , Interleukin-6/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred BALB C , Sendai virus/radiation effects , Survival Analysis , Ultraviolet Rays , Virus Inactivation/radiation effects
5.
Clin Cancer Res ; 19(3): 668-79, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23251005

ABSTRACT

PURPOSE: Cancer immunotherapy has encountered many difficulties in the face of the expectation to eradicate cancer, and new breakthroughs are required. We have previously shown that UV-inactivated Sendai virus particles (hemagglutinating virus of Japan envelope; HVJ-E) induce immunity against multiple tumor types. In this study, a novel pseudovirion that stimulates more robust antitumor immunity was designed for cancer treatment. EXPERIMENTAL DESIGN: First, we found that culturing murine splenocytes with HVJ-E in combination with interleukin (IL)-12 resulted in a remarkable increase in IFN-γ production compared with that observed in splenocytes cultured with IL-12 alone. The synergistic effects of HVJ-E and IL-12 on IFN-γ production were caused by viral F proteins independently of HVJ-E fusion activity and not by hemagglutination from hemagglutinin-neuraminidase (HN) proteins. We next constructed HN-depleted HVJ-E expressing the Fc region of immunoglobulin G (IgG) on the envelope and single-chain IL-12 containing the ZZ domain of protein A to produce an IL-12-conjugated HVJ-E particle without hemagglutinating activity. RESULTS: IL-12-conjugated HVJ-E dramatically enhanced the amount of IFN-γ produced by immune cells. Intratumoral injection of IL-12-conjugated HVJ-E eradicated murine melanomas more effectively than injection of wild-type HVJ-E through increased production of melanoma-specific CTLs. IL-12-conjugated HVJ-E preferentially accumulated in the lungs after systemic administration. When small metastatic melanoma foci were formed in the lungs, systemic administration of IL-12-conjugated HVJ-E significantly reduced the number of metastatic foci by inducing local production of IFN-γ in the lungs and generating large numbers of melanoma-specific CTLs. CONCLUSION: IL-12-conjugated HVJ-E is a promising tool for the treatment of cancers, including lung metastasis.


Subject(s)
Interferon-gamma/biosynthesis , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Melanoma/immunology , Melanoma/pathology , Sendai virus/immunology , Virion/immunology , Animals , Cell Line , Cricetinae , Disease Models, Animal , Female , Haplorhini , Immunoglobulin Fc Fragments/immunology , Injections, Intralesional , Interleukin-12/immunology , Interleukin-12/metabolism , Lung Neoplasms/metabolism , Melanoma/metabolism , Mice , Sendai virus/radiation effects , Viral Fusion Proteins/immunology , Virion/radiation effects
6.
Mol Immunol ; 46(3): 393-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19038458

ABSTRACT

The innate cellular response to virus particle entry in non-immune cells requires the transcriptional activity of interferon regulatory factor 3 (IRF-3), but not production of type I interferon (IFN). Here, we characterize the IFN-independent innate cellular response to virus-derived stimuli in Vero cells, a monkey kidney epithelial cell line deficient for IFN production. We provide evidence that Vero cells are deficient in their ability to mount an IRF-3-dependent, IFN-independent antiviral response against either incoming virus particles or polyinosinic:polycytidylic acid (pIC), a dsRNA mimetic. We further demonstrate that abundance of IRF-3 protein is a determinant in the pIC-mediated antiviral signalling pathway. These observations further characterize the permissive nature of Vero cells to viral infection, and highlight the crucial involvement of IRF-3 in the innate antiviral response.


Subject(s)
Herpesvirus 1, Human/immunology , Interferon Regulatory Factor-3/immunology , Interferons/biosynthesis , Sendai virus/immunology , Animals , Chlorocebus aethiops , Cloning, Molecular , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/radiation effects , Humans , Poly I-C/pharmacology , Sendai virus/drug effects , Sendai virus/physiology , Sendai virus/radiation effects , Sequence Analysis, DNA , Ultraviolet Rays , Vero Cells , Virus Inactivation/drug effects , Virus Inactivation/radiation effects , Virus Replication/drug effects , Virus Replication/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...