Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Int J Cancer ; 134(12): 2865-77, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24265099

ABSTRACT

Ovarian cancer is the most frequent cause of gynecological cancer-related mortality as a majority of patients are diagnosed at an advanced stage with intraperitoneal dissemination because of the absence of initial symptoms. Granulocyte-macrophage colony-stimulating factor (GM-CSF) plays an important role in the maturation of specialized antigen-presenting cells. In this study, we utilized a herpes simplex virus (HSV) amplicon expressing murine GM-CSF combined with HF10 (mGM-CSF amplicon), a highly attenuated HSV type 1 strain functioning as a helper virus to strengthen anti-tumor immune response, for the treatment of ovarian cancer with intraperitoneal dissemination. A mouse ovarian cancer cell line, OV2944-HM-1 (HM-1), was intraperitoneally injected, following which HF10 only or the mGM-CSF amplicon was injected intraperitoneally three times. HF10 injection prolonged survival and decreased intraperitoneal dissemination, but to a lesser extent than the mGM-CSF amplicon. Although HF10 replication was not observed in HM-1 cells, expression of VP5, a late gene coding the major capsid protein of HSV, was detected. Moreover, mGM-CSF production was detected in transfected HM-1 cells. Immunohistochemical staining revealed the infiltration of CD4- and CD8-positive cells into the peritoneal tumor(s). A significantly increased CD4+ T cell concentration was observed in the spleen. Murine splenic cells after each treatment were stimulated with HM-1 cells, and the strongest immune response was observed in the mice that received mGM-CSF amplicon injections. These results suggested that the mGM-CSF amplicon is a promising agent for the treatment of advanced ovarian cancer with intraperitoneal dissemination.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Herpesvirus 1, Human/genetics , Oncolytic Virotherapy/methods , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Capsid Proteins/biosynthesis , Cell Line, Tumor , Cell Movement/immunology , Chlorocebus aethiops , Female , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Herpesvirus 1, Human/immunology , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Survival , Tumor Necrosis Factor-alpha/metabolism , Vero Cells
2.
Front Microbiol ; 3: 158, 2012.
Article in English | MEDLINE | ID: mdl-22557998

ABSTRACT

Genital herpes is an intractable disease caused mainly by herpes simplex virus (HSV) type 2 (HSV-2), and is a major concern in public health. A previous infection with HSV type 1 (HSV-1) enhances protection against primary HSV-2 infection to some extent. In this study, we evaluated the ability of HF10, a naturally occurring replication-competent HSV-1 mutant, to protect against genital infection in mice caused by HSV-2. Subcutaneous inoculation of HF10-immunized mice against lethal infection by HSV-2, and attenuated the development of genital ulcer diseases. Immunization with HF10 inhibited HSV-2 replication in the mouse vagina, reduced local inflammation, controlled emergence of neurological dysfunctions of HSV-2 infection, and increased survival. In HF10-immunized mice, we observed rapid and increased production of interferon-γ in the vagina in response to HSV-2 infection, and numerous CD4(+) and a few CD8(+) T cells localized to the infective focus. CD4(+) T cells invaded the mucosal subepithelial lamina propria. Thus, the protective effect of HF10 was related to induction of cellular immunity, mediated primarily by Th1 CD4(+) cells. These data indicate that the live attenuated HSV-1 mutant strain HF10 is a promising candidate antigen for a vaccine against genital herpes caused by HSV-2.

3.
J Virol ; 86(1): 492-503, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22013039

ABSTRACT

Tankyrase 1 is a poly(ADP-ribose) polymerase (PARP) which localizes to multiple subcellular sites, including telomeres and mitotic centrosomes. Poly(ADP-ribosyl)ation of the nuclear mitotic apparatus (NuMA) protein by tankyrase 1 during mitosis is essential for sister telomere resolution and mitotic spindle pole formation. In interphase cells, tankyrase 1 resides in the cytoplasm, and its role therein is not well understood. In this study, we found that herpes simplex virus (HSV) infection induced extensive modification of tankyrase 1 but not tankyrase 2. This modification was dependent on extracellular signal-regulated kinase (ERK) activity triggered by HSV infection. Following HSV-1 infection, tankyrase 1 was recruited to the nucleus. In the early phase of infection, tankyrase 1 colocalized with ICP0 and thereafter localized within the HSV replication compartment, which was blocked in cells infected with the HSV-1 ICP0-null mutant R7910. In the absence of infection, ICP0 interacted with tankyrase 1 and efficiently promoted its nuclear localization. HSV did not replicate efficiently in cells depleted of both tankyrases 1 and 2. Moreover, XAV939, an inhibitor of tankyrase PARP activity, decreased viral titers to 2 to 5% of control values. We concluded that HSV targets tankyrase 1 in an ICP0- and ERK-dependent manner to facilitate its replication.


Subject(s)
Cell Nucleus/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Herpes Simplex/enzymology , Herpesvirus 1, Human/physiology , Immediate-Early Proteins/metabolism , Tankyrases/metabolism , Ubiquitin-Protein Ligases/metabolism , Virus Replication , Animals , Cell Line , Cell Nucleus/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Herpes Simplex/genetics , Herpes Simplex/virology , Herpesvirus 1, Human/enzymology , Herpesvirus 1, Human/genetics , Humans , Immediate-Early Proteins/genetics , Phosphorylation , Protein Binding , Protein Transport , Tankyrases/genetics , Ubiquitin-Protein Ligases/genetics
4.
Cancer Sci ; 103(2): 375-81, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22017376

ABSTRACT

Epstein-Barr virus (EBV), which infects B cells, T cells, and natural killer (NK) cells, is associated with multiple lymphoid malignancies. Recently, histone deacetylase (HDAC) inhibitors have been reported to have anticancer effects against various tumor cells. In the present study, we evaluated the killing effect of valproic acid (VPA), which acts as an HDAC inhibitor, on EBV-positive and -negative T and NK lymphoma cells. Treatment of multiple T and NK cell lines (SNT13, SNT16, Jurkat, SNK6, KAI3 and KHYG1) with 0.1-5 mM of VPA inhibited HDAC, increased acetylated histone levels and reduced cell viability. No significant differences were seen between EBV-positive and -negative cell lines. Although VPA induced apoptosis in some T and NK cell lines (SNT16, Jurkat and KHYG1) and cell cycle arrest, it did not induce lytic infection in EBV-positive T or NK cell lines. Because the killing effect of VPA was modest (1 mM VPA reduced cell viability by between 22% and 56%), we tested the effects of the combination of 1 mM of VPA and 0.01 µM of the proteasome inhibitor bortezomib. The combined treated of cells with VPA and bortezomib had an additive killing effect. Finally, we administered VPA to peripheral blood mononuclear cells from three patients with EBV-associated T or NK lymphoproliferative diseases. In these studies, VPA had a greater killing effect against EBV-infected cells than uninfected cells, and the effect was increased when VPA was combined with bortezomib. These results indicate that VPA has antitumor effects on T and NK lymphoma cells and that VPA and bortezomib may have synergistic effects, irrespective of the presence of EBV.


Subject(s)
Antineoplastic Agents/pharmacology , Herpesvirus 4, Human/physiology , Killer Cells, Natural/drug effects , Lymphoma, Non-Hodgkin/drug therapy , T-Lymphocytes/drug effects , Valproic Acid/pharmacology , Adolescent , Boronic Acids/pharmacology , Bortezomib , Cell Cycle Checkpoints , Cell Line , Cell Survival/drug effects , Child , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Infections/virology , Female , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Killer Cells, Natural/virology , Leukocytes, Mononuclear/drug effects , Lymphoma, Non-Hodgkin/pathology , Lymphoma, Non-Hodgkin/virology , Male , Pyrazines/pharmacology , T-Lymphocytes/virology
5.
Virol J ; 8: 257, 2011 May 25.
Article in English | MEDLINE | ID: mdl-21609490

ABSTRACT

BACKGROUND: Herpes simplex viruses (HSVs) rapidly shut off macromolecular synthesis in host cells. In contrast, global microarray analyses have shown that HSV infection markedly up-regulates a number of host cell genes that may play important roles in HSV-host cell interactions. To understand the regulatory mechanisms involved, we initiated studies focusing on the zinc finger transcription factor insulinoma-associated 1 (INSM1), a host cell protein markedly up-regulated by HSV infection. RESULTS: INSM1 gene expression in HSV-1-infected normal human epidermal keratinocytes increased at least 400-fold 9 h after infection; INSM1 promoter activity was also markedly stimulated. Expression and subcellular localization of the immediate early HSV protein ICP0 was affected by INSM1 expression, and chromatin immunoprecipitation (ChIP) assays revealed binding of INSM1 to the ICP0 promoter. Moreover, the role of INSM1 in HSV-1 infection was further clarified by inhibition of HSV-1 replication by INSM1-specific siRNA. CONCLUSIONS: The results suggest that INSM1 up-regulation plays a positive role in HSV-1 replication, probably by binding to the ICP0 promoter.


Subject(s)
Herpesvirus 1, Human/physiology , Host-Pathogen Interactions , Immediate-Early Proteins/metabolism , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , DNA, Viral/metabolism , Gene Expression Regulation , Humans , Keratinocytes/virology , Promoter Regions, Genetic , Protein Binding , Up-Regulation
6.
Virol J ; 7: 179, 2010 Aug 03.
Article in English | MEDLINE | ID: mdl-20682038

ABSTRACT

BACKGROUND: Herpes simplex virus type 2 (HSV-2) is one of many viruses that exploits and modifies the cellular ubiquitin system. HSV-2 expresses the tegument protein UL56 that has been implicated in cytoplasmic transport and/or release of virions, and is a putative regulatory protein of Nedd4 ubiquitin ligase. In order to elucidate the biological function of UL56, this study examined the interaction of UL56 with the Nedd4-family ubiquitin ligase Itch and its role in the regulation of Itch. Additionally, we assessed the similarity between UL56 and regulatory proteins of Itch and Nedd4, Nedd4-family-interactins proteins (Ndfip). RESULTS: UL56 interacted with Itch, independent of additional viral proteins, and mediated more striking degradation of Itch, compared to Nedd4. Moreover, it was suggested that the lysosome pathway as well as the proteasome pathway was involved in the degradation of Itch. Other HSV-2 proteins with PY motifs, such as VP5 and VP16, did not mediate the degradation of endogenous Itch. Ndfip1 and Ndfip2 were similar in subcellular distribution patterns to UL56 and colocalized with UL56 in co-transfected cells. CONCLUSIONS: We believe that this is the first report demonstrating the interaction of a HSV-specific protein and Itch. Thus, UL56 could function as a regulatory protein of Itch. The mechanism, function and significance of regulating Itch in HSV-2 infection remain unclear and warrant further investigation.


Subject(s)
Protein Interaction Mapping , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Viral Proteins/metabolism , Animals , Carrier Proteins/genetics , Cell Line , Chlorocebus aethiops , Gene Expression Regulation , Humans , Lysosomes/metabolism , Membrane Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Sequence Homology, Amino Acid , Viral Proteins/genetics
7.
Curr Gene Ther ; 8(3): 208-21, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18537595

ABSTRACT

Oncolytic HSV-1 has been developed as a novel anticancer agent. According to the properties and functions of HSV-1 encoded proteins, several genes have been targeted for engineering of oncolytic HSV-1. As a result, a variety of strategies have been applied to the engineering of oncolytic HSV-1. Success in cancer therapy for solid tumors requires a maximal oncolytic effect; however, recombinant HSV-1 that has been adapted to meet neurotoxicity requirements for the treatment of brain tumors may be too highly attenuated for effective use in solid tumors outside the brain. Recently, there has been renewed interest in the high potency of naturally oncolytic viruses. In this review, we will overview the engineered oncolytic HSV developed thus far, as well as its mechanism of selectivity and its mode of spreading within tumors. We also discuss the preclinical and clinical studies of HF-10, a non-engineered oncolytic HSV-1 virus, and its potential for use in cancer gene therapy.


Subject(s)
Herpesvirus 1, Human/genetics , Neoplasms/therapy , Oncolytic Virotherapy/methods , Animals , Clinical Trials as Topic , Female , Genes, Immediate-Early , Genetic Engineering , Genetic Therapy/methods , Humans , Male , Mice , Mutation , Viral Fusion Proteins/genetics
8.
Microbes Infect ; 10(4): 405-13, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18403238

ABSTRACT

Herpes simplex virus types 1 (HSV-1) and 2 (HSV-2) induce similar responses in infected cells and animals but differ in several significant respects. Previous studies have shown that defects in the US3-encoded protein kinase greatly affect both viruses in their interactions with cells and hosts. To investigate the impact of infection with HSV-1, HSV-2 and their US3-deficient mutants (DeltaUS3) on cellular transcriptional responses, we performed a global microarray analysis on human epithelial HEp-2 cells that were mock-infected, or infected with wild-type (WT) HSV-1, HSV-2 and their DeltaUS3 mutants. Among 54,765 probe sets examined, only 1156 (approximately 2.1%) and 2006 (approximately 3.7%) genes increased by at least fourfold at 9h postinfection in WT HSV-1 and HSV-2-infected cells, respectively. Unexpectedly, HSV-2 infection increases mRNA levels for a larger number of cellular genes than HSV-1 infection. Additionally, DeltaUS3 infection upregulated the expression of a larger number of cellular genes than WT infection. The genes affected by HSV infection were assigned to various groups of functional classes and cellular pathways. We have thus identified cellular genes whose expression was similarly or differently changed by infection with each virus.


Subject(s)
Herpesvirus 1, Human/physiology , Herpesvirus 2, Human/physiology , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases/genetics , Viral Proteins/genetics , Cell Line , Epithelial Cells , Gene Deletion , Herpesvirus 1, Human/genetics , Herpesvirus 2, Human/genetics , Humans , Time Factors , Up-Regulation
9.
Biochem Biophys Res Commun ; 349(4): 1372-7, 2006 Nov 03.
Article in English | MEDLINE | ID: mdl-16979593

ABSTRACT

To elucidate the regulation of IL-27p28 gene, we analyzed the promoter region of the gene in DC2.4 cells with or without lipopolysaccharide (LPS)-treatment. The results indicate that a region (-648 to -364) of p28 promoter was responsible for LPS-induction. EMSA with DNA probes within the region reveals that binding of GATA motif bound proteins was decreased by LPS-treatment. We identified one of the proteins as non-POU domain-containing octamer binding protein (NonO). Taken together, LPS-induced activation of IL-27p28 gene can be accounted for by the displacement of bound NonO protein from the IL-27p28 promoter.


Subject(s)
Dendritic Cells/metabolism , Interleukins/metabolism , Lipopolysaccharides/pharmacology , Animals , Cell Line , Dendritic Cells/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Interleukins/genetics , Mice , Recombinant Proteins/metabolism
10.
Biochem Biophys Res Commun ; 338(4): 1773-8, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16288719

ABSTRACT

Lewis and Brown Norway rats are entirely different with respect to the polarization of their immune responses (Th1 and Th2, respectively). We found that naive Lewis rat splenocytes treated in vitro with heat-killed Mycobacterium tuberculosis (Mtb) upregulate the expression of both subunits of IL-27 (IL-27p28 and EBI3). Mtb treatment caused naive Lewis rat splenocytes to express 4.6-fold more IL-27p28 than Mtb-treated Brown Norway rat splenocytes 6h after the treatment. Although WSX-1, the IL-27 receptor, was not induced by Mtb treatment in splenocytes from either rat strain, Lewis rats expressed significantly higher levels of the IL-27 signal transducers T-bet and IL-12Rbeta2 than Brown Norway rats. Flow cytometric analysis of dendritic cells from bone marrow cells revealed Lewis rats had more IL-27p28-positive cells. Thus, early in the immune response, Lewis rats appear to produce higher levels of IL-27 than Brown Norway rats, resulting in polarization towards Th1-immunity.


Subject(s)
Dendritic Cells/physiology , Interleukins/biosynthesis , Rats, Inbred Lew/immunology , Th1 Cells/immunology , Animals , Histocompatibility Antigens Class II/immunology , Interleukins/physiology , Male , Minor Histocompatibility Antigens , Mycobacterium tuberculosis/immunology , Protein Subunits/biosynthesis , Rats , Rats, Inbred BN/immunology , Receptors, Interleukin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL