Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Technol Cancer Res Treat ; 22: 15330338221146024, 2023.
Article in English | MEDLINE | ID: mdl-36617975

ABSTRACT

BACKGROUND: RAS homolog family member A (RhoA), a member of the Rho family of small GTPases, and Vav1, a guanine nucleotide exchange factor for Rho family GTPases, have been reported to activate pathways related to the actin cytoskeleton and regulation of cell shape, attachment, and motility. The interaction between these molecules in lymphoma is involved in malignant signaling, but its function in epithelial malignancy is unknown. Here, we investigated the malignant signal of mutant RhoA in gastric cancer and demonstrated the potential of RhoA G17E/Vav1 as a therapeutic target for diffuse gastric cancer. METHODS: The RhoA mutants R5W, G17E, and Y42C were retrovirally transduced into the gastric cancer cell line MKN74. The stably transduced cells were used for morphology, proliferation, and migration/invasion assays in vitro. MKN74 cells stably transduced with ectopic wild-type RhoA and mutant RhoA (G17E) were used in a peritoneal xenograft assay. RESULTS: The RhoA mutations G17E and Y42C induced morphological changes in MKN74. G17E induced Vav1 expression at the mRNA and protein levels and promoted the migration and invasion of MKN74. An RNA interference assay of Vav1 revealed that RhoA G17E enhanced cancer cell invasion via Vav1. Furthermore, immunoprecipitation revealed that Vav1 and RhoA G17E specifically bind and function together through matrix metalloproteinase -9. In a peritoneal xenograft model of nude mice, RhoA G17E promoted peritoneal dissemination, whereas Vav1 knockdown suppressed it. CONCLUSION: Overall, our findings indicate that RhoA G17E is associated with Vav1 and promoted cancer invasion via matrix metalloproteinase -9 in gastric cancer cells. Thus, RhoA G17E/Vav1 signaling in diffuse gastric cancer may be a useful therapeutic target.


Subject(s)
Proto-Oncogene Proteins c-vav , Stomach Neoplasms , rhoA GTP-Binding Protein , Animals , Humans , Mice , Cell Line, Tumor , Cell Movement/genetics , Matrix Metalloproteinase 9/metabolism , Mice, Nude , Neoplasm Invasiveness/genetics , Proto-Oncogene Proteins c-vav/genetics , Proto-Oncogene Proteins c-vav/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism , Stomach Neoplasms/pathology
2.
J Equine Sci ; 32(1): 1-9, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33776534

ABSTRACT

This review briefly describes the virus classification, clinical signs, epidemiology, diagnosis, disinfection, and vaccines related equine group A rotavirus (RVA) infection. Equine RVA is one of the most important pathogens causing diarrhoea in foals. The main transmission route is faecal-oral, and the clinical signs are diarrhoea, fever, lethargy, and anorexia (decreased suckling). Some human RVA rapid antigen detection kits based on the principles of the immunochromatographic assay are useful for the diagnosis of equine RVA infection. The kits are used in daily clinical practice because of their rapidity and ease of handling. Equine RVA is a non-enveloped virus and is more resistant to disinfectants than enveloped viruses such as equine influenza virus and equine herpesvirus. Although amphoteric soaps and quaternary ammonium compounds are commonly used in veterinary hygiene, they are generally ineffective against equine RVA. Alcohol products, aldehydes, and chlorine- and iodine-based compounds are effective against equine RVA. Inactivated vaccines have been used for equine RVA infection in some countries. Pregnant mares are intramuscularly inoculated with a vaccine, and thus their colostrum has abundant antibodies against RVA at the time of birth. According to G and P classification defined in accordance with the VP7 and VP4 genes, respectively, the predominant equine RVAs circulating in horse populations globally are G3P[12] and G14P[12] equine RVAs, but the vaccines contain only the G3P[12] equine RVA strain. Ideally, a G14P[12] equine RVA should be added as a vaccine strain to obtain a better vaccine effect.

3.
Biosci Biotechnol Biochem ; 85(2): 324-331, 2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33604645

ABSTRACT

APS001F is a strain of Bifidobacterium longum genetically engineered to express cytosine deaminase that converts 5-fluorocytosine (5-FC) to 5-fluorouracil. In the present study, antitumor effects of APS001F plus 5-FC (APS001F/5-FC) in combination with anti-PD-1 monoclonal antibody were investigated using a CT26 syngeneic mouse model. Both of dosing of APS001F/5-FC before and after anti-PD-1 mAb in the combination dosing exhibited antitumor effects as well as prolonged survival over the nontreated control. The survival rate in the combination therapy significantly increased over the monotherapy with APS001F/5-FC and that with anti-PD-1 mAb. Regulatory T cells among CD4+ T cells in tumor decreased in the combination therapy, while the ratio of CD8+ T cells was maintained in all groups. Taken these results together, APS001F/5-FC not only demonstrates a direct antitumor activity, but also immunomodulatory effects once localized in the hypoxic region of the tumor, which allows anti-PD-1 mAb to exert potentiated antitumor effects.


Subject(s)
Antibodies, Monoclonal/immunology , Antineoplastic Agents/pharmacology , Bifidobacterium longum/physiology , Flucytosine/pharmacology , Genetic Engineering , Programmed Cell Death 1 Receptor/immunology , Animals , Bifidobacterium longum/genetics , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Mice
4.
Int J Oncol ; 57(5): 1179-1191, 2020 11.
Article in English | MEDLINE | ID: mdl-32901840

ABSTRACT

KRAS­mutant colorectal cancer (CRC) is a highly malignant cancer with a poor prognosis, however specific therapies targeting KRAS mutations do not yet exist. Anti­epidermal growth factor receptor (EGFR) agents, including cetuximab and panitumumab, are effective for the treatment of certain patients with CRC. However, these anti­EGFR treatments have no effect on KRAS­mutant CRC. Therefore, new therapeutic strategies targeting KRAS­mutant CRC are urgently needed. To clarify the direct effect of KRAS gene mutations, the present study transduced mutant forms of the KRAS gene (G12D, G12V and G13D) into CACO­2 cells. A drug­screening system (Mix Culture assay) was then applied, revealing that the cells were most sensitive to the MEK inhibitor trametinib among tested drugs, Cetuximab, Panitumumab, Regorafenib, Vemurafenib, BEZ­235 and Palbociclib. Trametinib suppressed phosphorylated ERK (p­ERK) expression and inhibited the proliferation of KRAS­mutant CACO­2 cells. However, low­dose treatment with trametinib also increased the expression of the anti­apoptotic protein Bcl­xL in a dose­dependent manner, leading to drug resistance. To overcome the resistance of KRAS­mutant CRC to apoptosis, the combination of trametinib and the Bcl­xL antagonist ABT263 was assessed by in vitro and in vivo experiments. Compared with the effects of low­dose trametinib monotherapy, combination treatment with ABT263 had a synergistic effect on apoptosis in mutant KRAS transductants in vitro. Furthermore, in vivo combination therapy using low­dose trametinib and ABT263 against a KRAS­mutant (G12V) xenograft synergistically suppressed growth, with an increase in apoptosis compared with the effects of trametinib monotherapy. These data suggest that a low dose of trametinib (10 nM), rather than the usual dose of 100 nM, in combination with ABT263 can overcome the resistance to apoptosis induced by Bcl­xL expression, which occurs concurrently with p­ERK suppression in KRAS­mutant cells. This strategy may represent a promising new approach for treating KRAS­mutant CRC.


Subject(s)
Colorectal Neoplasms/drug therapy , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , bcl-X Protein/antagonists & inhibitors , Aniline Compounds/pharmacology , Animals , Apoptosis/drug effects , Caco-2 Cells , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Pyridones/pharmacology , Pyrimidinones/pharmacology , Sulfonamides/pharmacology
5.
Am J Pathol ; 190(2): 453-468, 2020 02.
Article in English | MEDLINE | ID: mdl-31734232

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a transcription factor known as a dioxin receptor. Recently, Ahr-/- mice were revealed to develop cecal tumors with inflammation and Wnt/ß-catenin pathway activation. However, whether ß-catenin degradation is AhR dependent remains unclear. To determine whether other signaling pathways function in Ahr-/- cecal tumorigenesis, we investigated histologic characteristics of the tumors and cytokine/chemokine production in tumors and Ahr-/- peritoneal macrophages. AhR expression was also assessed in human colorectal carcinomas. Of the 28 Ahr-/- mice, 10 developed cecal lesions by 50 weeks of age, an incidence significantly lower than previously reported. Cecal lesions of Ahr-/- mice developed from serrated hyperplasia to adenoma/dysplasia-like neoplasia with enhanced proliferation. Macrophage and neutrophil infiltration into the lesions was also observed early in serrated hyperplasia, although adjacent mucosa was devoid of inflammation. Il1b, Il6, Ccl2, and Cxcl5 were up-regulated at lesion sites, whereas only IL-6 production increased in Ahr-/- peritoneal macrophages after lipopolysaccharide + ATP stimulation. Neither Myc (alias c-myc) up-regulation nor ß-catenin nuclear translocation was observed, unlike previously reported. Interestingly, enhanced phosphorylation of extracellular signal-regulated kinase, Src, and epidermal growth factor receptor and Amphiregulin up-regulation at Ahr-/- lesion sites were detected. In human serrated lesions, however, AhR expression in epithelial cells was up-regulated despite morphologic similarity to Ahr-/- cecal lesions. Our results suggest novel mechanisms underlying Ahr-/- cecal tumorigenesis, depending primarily on cecum-specific mitogen-activated protein kinase pathway activation and inflammation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Carcinogenesis/pathology , Cecal Neoplasms/pathology , Colorectal Neoplasms/pathology , Inflammation/pathology , Mitogen-Activated Protein Kinases/metabolism , Receptors, Aryl Hydrocarbon/physiology , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinogenesis/immunology , Carcinogenesis/metabolism , Cecal Neoplasms/immunology , Cecal Neoplasms/metabolism , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Female , Hyperplasia/immunology , Hyperplasia/metabolism , Hyperplasia/pathology , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/genetics , Phosphorylation , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism
6.
BMC Vet Res ; 15(1): 280, 2019 Aug 06.
Article in English | MEDLINE | ID: mdl-31387602

ABSTRACT

BACKGROUND: Equine herpesvirus type 1 (EHV-1) infection is a major cause of pyrexias in winter among Japanese racehorses. In 2014-2015, the Japan Racing Association (JRA) changed the EHV-1 vaccine from an inactivated vaccine to a live vaccine (both produced by Nisseiken). To evaluate the effect of changing the vaccines, the capacities of these vaccines to induce virus-neutralizing (VN) antibodies were compared, and an epizootiological investigation of EHV-1 was performed at the JRA Ritto Training Center during epizootic periods from 2010-2011 to 2016-2017. RESULTS: Three-year-old horses that received the first dose of live vaccine showed higher geometric mean (GM) VN titers (205 and 220) than those that received inactivated vaccine (83, P < 0.05). The response rates after vaccination with the live vaccine (76 and 90%) were higher than that after vaccination with inactivated vaccine (42%, P < 0.05). Four-year-old horses from 2015 to 2017 that had received the live vaccine in the previous epizootic periods had higher GM titers (205 to 246) than those from 2011 to 2014, which had received the inactivated vaccine (139 to 164, P < 0.05). The estimated numbers of horses infected with EHV-1 or EHV-4, or both, in 2011-2012 (29 [95%CI: 21-37]) and 2013-2014 (37 [95%CI: 27-47]) were higher than those in the other periods (7 [95%CI: 2-12] to 16 [95%CI: 9-23]). Likewise, the seroconversion rates to EHV-1 in horses that stayed at the training center in 2011-2012 (66.0%) and 2013-2014 (52.0%) were higher than those in the other periods (12.0 to 28.6%). CONCLUSIONS: The live EHV-1 vaccine is highly immunogenic and provides greater VN antibody responses than the inactivated vaccine. Unlike the period when the policy was to use inactivated vaccine, there was no detectable epizootic EHV-1 infection at the training center during three consecutive periods after the introduction of the live vaccine. These results suggest that the replacement of inactivated vaccine with live vaccine, together with the achievement of high vaccination coverage, reinforced the herd effect, and contributed to better control of EHV-1 epizootics in the training center.


Subject(s)
Antibodies, Viral/blood , Herpesvirus 1, Equid , Horse Diseases/prevention & control , Viral Vaccines/immunology , Animals , Herpesvirus 4, Equid , Horse Diseases/epidemiology , Horse Diseases/virology , Horses , Japan/epidemiology , Seasons , Serologic Tests , Vaccines, Inactivated
7.
Vet Microbiol ; 222: 18-24, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30080668

ABSTRACT

Immune responses were compared after intranasal (IN) and intramuscular (IM) vaccination of horses with a modified live equine herpesvirus type-1 (EHV-1) vaccine, and the protective effect after EHV-1 challenge was evaluated. IN- and IM-vaccinated groups (n = 5 each) showed significant rises in serum virus-neutralizing titers with increased levels of IgGa and IgGb antibodies after the first vaccination (P < 0.05). In nasal secretions, the IN group had significantly increased levels of IgA antibodies after vaccination (P < 0.05), whereas the response of the IM group was dominated by IgGa and IgGb subclasses. After challenge infection, the numbers of pyretic horses from 1 to 4 days post-inoculation were 3/5 in the placebo (PBO) group (n = 5), 0/5 in the IN group, and 1/5 in the IM group. The IN and IM groups had significantly lower levels of virus shedding than the PBO group (P < 0.05). There were no significant between-group differences in the numbers of viremic horses each day. Notably, two horses in the IM group had no virus shedding or viremia, whereas all horses in the other group did. Both IN and IM vaccination induced systemic humoral immunity and mucosal immunity, suppressing virus replication in the nasal mucosa, and partially protected horses from pyrexia, especially early in infection. This study showed a mucosal antibody response was induced, not only by IN vaccination but also by IM vaccination.


Subject(s)
Administration, Intranasal/methods , Herpesviridae Infections/veterinary , Herpesvirus 1, Equid/immunology , Herpesvirus Vaccines/immunology , Injections, Intramuscular/methods , Vaccination/veterinary , Animals , Antibodies, Viral/blood , Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/virology , Herpesvirus Vaccines/administration & dosage , Horse Diseases/immunology , Horse Diseases/prevention & control , Horse Diseases/virology , Horses , Immunity, Mucosal , Immunoglobulin A/blood , Immunoglobulin G/blood , Neutralization Tests , Vaccination/methods , Vaccines, Attenuated/administration & dosage , Virus Shedding/immunology
8.
J Biol Chem ; 293(17): 6326-6336, 2018 04 27.
Article in English | MEDLINE | ID: mdl-29496994

ABSTRACT

Fascin1 is an actin-bundling protein involved in cancer cell migration and has recently been shown also to have roles in virus-mediated immune cell responses. Because viral infection has been shown to activate immune cells and to induce interferon-ß expression in human cancer cells, we evaluated the effects of fascin1 on virus-dependent signaling via the membrane- and actin-associated protein RIG-I (retinoic acid-inducible gene I) in colon cancer cells. We knocked down fascin1 expression with shRNA retrovirally transduced into a DLD-1 colon cancer and L929 fibroblast-like cell lines and used luciferase reporter assays and co-immunoprecipitation to identify fascin1 targets. We found that intracellular poly(I·C) transfection to mimic viral infection enhances the RIG-I/MDA5 (melanoma differentiation-associated gene 5)-mediated dimerization of interferon regulatory factor 3 (IRF-3). The transfection also significantly increased the expression levels of IRF-7, interferon-ß, and interferon-inducible cytokine IP-10 in fascin1-deleted cells compared with controls while significantly suppressing cell growth, migration, and invasion. We also found that fascin1 constitutively interacts with IκB kinase ϵ (IKKϵ) in the RIG-I signaling pathway. In summary, we have identified fascin1 as a suppressor of the RIG-I signaling pathway associating with IκB kinase ϵ in DLD-1 colon cancer cells to suppress immune responses to viral infection.


Subject(s)
Carrier Proteins/metabolism , Colonic Neoplasms/metabolism , DEAD Box Protein 58/metabolism , I-kappa B Kinase/metabolism , Interferon-beta/metabolism , Microfilament Proteins/metabolism , Neoplasm Proteins/metabolism , Signal Transduction , Animals , Carrier Proteins/genetics , Carrier Proteins/immunology , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/virology , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , HEK293 Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Mice , Microfilament Proteins/genetics , Microfilament Proteins/immunology , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Receptors, Immunologic , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/metabolism
9.
Tohoku J Exp Med ; 244(2): 133-144, 2018 02.
Article in English | MEDLINE | ID: mdl-29459573

ABSTRACT

Primary lung cancer is the most frequent cause of cancer-related deaths worldwide. Cisplatin has been used as a key drug in the treatment for patients with lung cancer; however, most of the patients failed to respond to cisplatin within several months, and the mechanisms underlying the cisplatin resistance have not been fully elucidated. Apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) is a key adaptor protein in the formation of inflammasomes. ASC is also involved in apoptotic signaling. Importantly, ASC expression is decreased in lung cancer and various cancers, but its precise function in tumor progression remains unknown. To explore the hitherto unknown role of ASC in lung cancer, we initially searched for lung cancer cell lines with higher expression levels of ASC using Cancer Cell Line Encyclopedia (CCLE) database, thereby identifying the A549 human non-small cell lung cancer cell line. Accordingly, with retroviral shRNA, the expression of ASC was forced to decrease in A549 cells. Stable ASC-knockdown cells, thus established, showed the increased activities of proliferation, motility, and invasion, compared with control cells. Importantly, ASC-knockdown cells also became resistant to cisplatin, but not to other anti-cancer agents, 5-fluorouracil and paclitaxel. Bcl-2 and phospho-Src levels were increased in ASC-knockdown cells. A Bcl-2 inhibitor, ABT-199, induced an apoptotic response in ASC-knockdown cells, and dasatinib, a Src inhibitor, blocked cell invasiveness. Thus, ASC may be involved in tumor suppression and cell death via Bcl-2 and pSrc. Targeting Bcl-2 and Src in ASC-downregulated populations of lung cancer may improve treatment outcome.


Subject(s)
Apoptosis , CARD Signaling Adaptor Proteins/metabolism , Cisplatin/therapeutic use , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , A549 Cells , Apoptosis/drug effects , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Phenotype , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Up-Regulation/drug effects , src-Family Kinases/metabolism
10.
J Vet Med Sci ; 80(2): 382-386, 2018 Mar 02.
Article in English | MEDLINE | ID: mdl-29237998

ABSTRACT

Equine influenza (EI) vaccine has been widely used. However, the causative EI virus (H3N8) undergoes continuous antigenic drift, and the vaccine strains must be periodically reviewed and if necessary, updated to maintain vaccine efficacy against circulating viruses. In 2016, the Japanese vaccine was updated by replacing the old viruses with the Florida sub-lineage Clade (Fc) 2 virus, A/equine/Yokohama/aq13/2010 (Y10). We investigated the virus neutralization (VN) antibody response to Fc2 viruses currently circulating in Europe, after booster or primary immunization with the new vaccine. These European viruses have the amino acid substitution A144V or I179V of the hemagglutinin. In horses that had previously received a primary course and bi-annual boosters with the old vaccine booster, immunization with the updated vaccine increased the VN antibody levels against the European Fc2 viruses as well as Y10. There were no significant differences in the VN titers against Y10 and the Fc2 viruses with A144V or I179V substitution in horses that had received a primary course of the updated vaccine. However, a mixed primary course where the first dose was the old vaccine and the second dose was the updated vaccine, reduced VN titers against the European viruses compared to that against Y10. In summary, the new vaccine affords horses protective level of VN titers against the Fc2 viruses carrying A144V or I179V substitution, but our results suggest that the combination of the old and new vaccines for primary immunization would not be optimum.


Subject(s)
Antibodies, Viral/blood , Horse Diseases/virology , Influenza A Virus, H3N8 Subtype/immunology , Orthomyxoviridae Infections/veterinary , Vaccination/veterinary , Animals , Antibody Formation , Female , Horse Diseases/immunology , Horse Diseases/prevention & control , Horses , Immunization, Secondary/veterinary , Influenza A Virus, H3N8 Subtype/classification , Japan , Male , Neutralization Tests/veterinary , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control
11.
Oncol Rep ; 39(2): 843-850, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29251327

ABSTRACT

Cholangiocarcinoma (CCC) is a strongly aggressive malignancy for which surgical resection is the only potential curative therapy. Sorafenib, a multikinase inhibitor of the RAF/MEK/ERK pathway, is a molecular-targeted drug that is approved for hepatocellular carcinoma (HCC) but not for CCC. The differences in signaling pathway characteristics under sorafenib treatment between HCC (HLF, Huh7, PLC/PRF/5) and CCC (RBE, YSCCC, Huh28) cell lines were therefore investigated using cell proliferation, western blotting, and apoptosis analyses. Sorafenib inhibited cell growth significantly less in CCC cells than in HCC cells, with lower suppression of ERK phosphorylation. Significantly decreased AKT Ser473 phosphorylation in HCC cells, and conversely enhanced phosphorylation of AKT Ser473 and mTORC2 in CCC cells, were observed with sorafenib treatment. Disassembly of the mTORC2 complex in RBE cells with siRNA targeting Rictor resulted in the downregulation of AKT Ser473 phosphorylation and enhanced apoptosis presumably via increased FOXO1, which consequently suppressed RBE cell proliferation. Phosphorylation of mTORC1 and autophagy were not influenced by sorafenib in CCC cells. Simultaneous administration of everolimus to suppress activated mTORC1 in RBE cells revealed that combined everolimus and sorafenib treatment under mTORC2 disassembly could enhance growth inhibition through the suppression of both sorafenib- and everolimus-dependent AKT Ser473 phosphorylation in addition to the inhibition of mTORC1 phosphorylation. Prevention of escape by AKT/mTOR signaling from the RAF/MEK/ERK pathway in sorafenib treatment by suppressing mTORC2 activity may lead to promising new approaches in CCC therapy.


Subject(s)
Bile Duct Neoplasms/metabolism , Cholangiocarcinoma/metabolism , Drug Resistance, Neoplasm , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Bile Duct Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Cholangiocarcinoma/drug therapy , Drug Synergism , Everolimus/pharmacology , Forkhead Box Protein O1/metabolism , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System/drug effects , Niacinamide/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Sorafenib , TOR Serine-Threonine Kinases/metabolism
12.
Mol Clin Oncol ; 7(2): 217-220, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28781788

ABSTRACT

The n-3 fatty acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), found in fish oil, exert a number of beneficial effects, and they are used in the treatment of hyperlipidemia. In recent years, EPA and DHA have been found to affect cancer cell proliferation. In the present study, PC3 cells, which are androgen-independent prostate cancer cells that resemble castration-resistant prostate cancer cells, were used to investigate a possible novel treatment for castration-resistant prostate cancer. The PC3 cells were cultured and incubated with various concentrations of EPA or DHA. Cancer proliferation was confirmed by trypan blue microscopy. Invasion and migration assays were used in the upper chamber in PC3 cells, and serum-free medium and various concentrations of EPA or DHA were placed in the lower chamber in serum-containing medium. EPA and DHA decreased PC3 cell proliferation, invasion and migration. The effect of EPA on PC3 cells was dose-dependent and significant differences were observed at concentrations of 100 and 200 µg/ml. The effect of DHA on PC3 cells was similar to that of EPA. In the migration assay, EPA exerted almost no effects at 25 µg/ml, but migration was reduced at 50 µg/ml. Similar to EPA, DHA exerted almost no effects at 25 µg/ml, but further reduction was observed at the 50 µg/ml concentration. In the invasion assay, EPA at 25 µg/ml was not significantly different from the control, but suppressed invasion at 50 µg/ml. DHA decreased invasion compared with the control at 25 µg/ml, whereas invasion was significantly reduced at a DHA concentration of 50 µg/ml. In conclusion, it was demonstrated that EPA and DHA were effective in decreasing the proliferation, invasion and migration of prostate PC3 cancer cells. However, the detailed underlying mechanisms have not yet been fully elucidated.

13.
J Vet Med Sci ; 79(6): 1061-1063, 2017 Jun 16.
Article in English | MEDLINE | ID: mdl-28458275

ABSTRACT

Equine influenza (EI) is a respiratory disease caused by equine influenza A virus (EIV, H3N8) infection. Rapid diagnosis is essential to limit the disease spread. We previously reported that some rapid antigen detection (RAD) tests are fit for diagnosing EI although their sensitivity is not optimal. Here, we evaluated the performance of the newly developed RAD test using silver amplification immunochromatography (Quick Chaser Auto Flu A, B: QCA) to diagnose EI. The detection limits of QCA for EIVs were five-fold lower than the conventional RAD tests. The duration of virus antigen detection in the infected horses was longer than the conventional RAD tests. We conclude that QCA could be a valuable diagnostic method for EI.


Subject(s)
Chromatography, Affinity/veterinary , Horse Diseases/diagnosis , Influenza A Virus, H3N8 Subtype , Orthomyxoviridae Infections/veterinary , Animals , Chromatography, Affinity/methods , Horse Diseases/virology , Horses , Orthomyxoviridae Infections/diagnosis , Silver , Time Factors
14.
PLoS One ; 12(1): e0169340, 2017.
Article in English | MEDLINE | ID: mdl-28056049

ABSTRACT

ASC (apoptosis-associated speck-like protein containing a CARD) is a key adaptor molecule of inflammasomes that mediates inflammatory and apoptotic signals. Aberrant methylation-induced silencing of ASC has been observed in a variety of cancer cells, thus implicating ASC in tumor suppression, although this role remains incompletely defined especially in the context of closely neighboring cell proliferation. As ASC has been confirmed to be silenced by abnormal methylation in HT1080 fibrosarcoma cells as well, this cell line was investigated to characterize the precise role and mechanism of ASC in tumor progression. The effects of ASC were examined using in vitro cell cultures based on comparisons between low and high cell density conditions as well as in a xenograft murine model. ASC overexpression was established by insertion of the ASC gene into pcDNA3 and pMX-IRES-GFP vectors, the latter being packed into a retrovirus and subjected to reproducible competitive assays using parental cells as an internal control, for evaluation of cell viability. p21 and p53 were silenced using shRNA. Cell viability was suppressed in ASC-expressing transfectants as compared with control cells at high cell density conditions in in vitro culture and colony formation assays and in in vivo ectopic tumor formation trials. This suppression was not detected in low cell density conditions. Furthermore, remarkable progression of apoptosis was observed in ASC-introduced cells at a high cell density, but not at a low one. ASC-dependent apoptosis was mediated not by p21, p53, or caspase-1, but rather by cleavage of caspase-9 as well as by suppression of the NF-κB-related X-linked inhibitor-of-apoptosis protein. Caspase-9 cleavage was observed to be dependent on gap junction formation. The remarkable effect of ASC on the induction of apoptosis through caspase-9 and gap junctions revealed in this study may lead to promising new approaches in anticancer therapy.


Subject(s)
Caspase 9/metabolism , Cytoskeletal Proteins/metabolism , Gap Junctions/metabolism , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , CARD Signaling Adaptor Proteins , Caspase 9/genetics , Cell Communication/genetics , Cell Communication/physiology , Cell Line, Tumor , Cell Survival , Connexin 43/genetics , Cytoskeletal Proteins/genetics , Humans , Immunohistochemistry , In Situ Nick-End Labeling , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/genetics , rho GTP-Binding Proteins/genetics
15.
Influenza Other Respir Viruses ; 10(6): 536-539, 2016 11.
Article in English | MEDLINE | ID: mdl-27465864

ABSTRACT

We previously reported that horse antiserum against the Japanese equine influenza vaccine virus, A/equine/La Plata/1993 (LP93) exhibited reduced cross-neutralization against some Florida sublineage Clade (Fc) 2 viruses, for example, A/equine/Carlow/2011 (CL11). As a result, Japanese vaccine manufacturers will replace LP93 with A/equine/Yokohama/aq13/2010 (Y10, Fc2). To assess the benefit of updating the vaccine, five horses vaccinated with inactivated Y10 vaccine and five vaccinated with inactivated LP93 were challenged by exposure to a nebulized aerosol of CL11. The durations of pyrexia (≥38.5°C) and other adverse clinical symptoms experienced by the Y10 group were significantly shorter than those of the LP93 group.


Subject(s)
Antibodies, Viral/blood , Antigens, Viral/immunology , Horse Diseases/immunology , Influenza A Virus, H3N8 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/veterinary , Animals , Antibodies, Viral/immunology , Antigens, Viral/genetics , Horse Diseases/virology , Horses , Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
16.
Cancer Med ; 5(9): 2487-500, 2016 09.
Article in English | MEDLINE | ID: mdl-27350283

ABSTRACT

Disorders of cytoskeletal remodeling and signal transduction are frequently involved in cancer progression. In particular, apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC) has been reported a proapoptotic molecule that is epigenetically silenced in several human cancers. ASC is a well-characterized adaptor protein involved in the formation of multiprotein oligomers, called inflammasomes, and plays a crucial role in the activation and secretion of interleukin-1ß and interleukin-18 in innate immune cells. However, the function of ASC in the regulation of tumor progression remains elusive. The present investigation examined the involvement of ASC in cancer progression and the acquisition of metastatic ability. To determine the effect of ASC depletion in in vitro and in vivo model systems, ASC was stably knocked down in B16 murine melanoma cell lines using retroviral transduction of shRNA. ASC suppression increased the motility of B16BL6 cells in scratch assays and augmented invasiveness as assessed by a Matrigel-coated transwell system. Invadopodia formation and Src phosphorylation level were markedly enhanced in ASC-knockdown cells as well. Since caspase-8 has been reported to enhance cellular migration by Tyr380 phosphorylation via Src, we examined Tyr380 phosphorylation of caspase-8 in ASC-knockdown cells and found it to be elevated in ASC-knockdown cells but attenuated by z-VAD-fmk or z-IETD-fmk. Moreover, ASC ablation increased pulmonary metastasis in mice after intravenous injection of B16BL6 cells. Our cumulative findings indicate that ASC suppresses cancer metastasis and progression via the modulation of cytoskeletal remodeling and the Src-caspase-8 signaling pathway.


Subject(s)
CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Neoplasms/metabolism , Phenotype , Signal Transduction , Animals , Caspase 8/metabolism , Cell Movement/genetics , Cell Proliferation/genetics , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Knockdown Techniques , Gene Silencing , Lung Neoplasms/secondary , Melanoma, Experimental , Mice , Neoplasm Metastasis , Neoplasms/pathology , Phosphorylation , src-Family Kinases/metabolism
17.
Influenza Other Respir Viruses ; 10(2): 127-33, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26568369

ABSTRACT

BACKGROUND: Equine influenza (EI) is a highly contagious disease caused by viruses of the H3N8 subtype. The rapid diagnosis of EI is essential to reduce the disease spread. Many rapid antigen detection (RAD) tests for diagnosing human influenza are available, but their ability to diagnose EI has not been systematically evaluated. OBJECTIVES: The aim of this study was to compare the performance of 22 RAD tests in the diagnosis of EI. METHODS: The 22 RAD tests were performed on fivefold serial dilutions of EI virus to determine their detection limits. The four most sensitive RAD tests (ImmunoAce Flu, BD Flu examan, Quick chaser Flu A, B and ESPLINE Influenza A&B-N) were further evaluated using nasopharyngeal samples collected from experimentally infected and naturally infected horses. The results were compared to those obtained using molecular tests. RESULTS: The detection limits of the 22 RAD tests varied hugely. Even the four RAD tests showing the best sensitivity were 125-fold less sensitive than the molecular techniques. The duration of virus detection in the experimentally infected horses was shorter using the RAD tests than using the molecular techniques. The RAD tests detected between 27% and 73% of real-time RT-PCR-positive samples from naturally infected horses. CONCLUSIONS: The study demonstrated the importance of choosing the right RAD tests as only three of 22 were fit for diagnosing EI. It was also indicated that even RAD tests with the highest sensitivity serve only as an adjunct to molecular tests because of the potential for false-negative results.


Subject(s)
Horse Diseases/diagnosis , Horse Diseases/virology , Influenza A Virus, H3N8 Subtype/isolation & purification , Orthomyxoviridae Infections/veterinary , Animals , Antibodies, Viral , Antigens, Viral/analysis , Enzyme-Linked Immunosorbent Assay , Horses , Humans , Influenza A Virus, H3N8 Subtype/genetics , Influenza A Virus, H3N8 Subtype/immunology , Influenza, Human/diagnosis , Influenza, Human/virology , Japan , Nasopharynx/virology , Orthomyxoviridae Infections/diagnosis , Orthomyxoviridae Infections/virology , RNA, Viral , Reagent Kits, Diagnostic , Real-Time Polymerase Chain Reaction/standards , Sensitivity and Specificity
18.
Virus Res ; 195: 172-6, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25456403

ABSTRACT

Equine herpesvirus type 1 (EHV-1) has haemagglutination (HA) activity toward equine red blood cells (RBCs), but the identity of its haemagglutinin is unknown. To identify the haemagglutinin of EHV-1, the major glycoproteins of EHV-1 were expressed in 293T cells, and the cells or cell lysates were mixed with equine RBCs. The results showed that only EHV-1 glycoprotein C (gC)-producing cells adsorbed equine RBCs, and that the lysate of EHV-1 gC-expressing cells agglutinated equine RBCs. EHV-1 lacking gC did not show HA activity. HA activity was inhibited by monoclonal antibodies (MAbs) specific for gC, but not by antibodies directed against other glycoproteins. In addition, HA activity was not inhibited by the addition of heparin. These results indicate that EHV-1 gC can bind equine RBCs irrespective of heparin, in contrast to other herpesvirus gC proteins.


Subject(s)
Hemagglutination , Hemagglutinins/metabolism , Herpesvirus 1, Equid/physiology , Viral Envelope Proteins/metabolism , Animals , Erythrocytes/drug effects , Horses
19.
Clin Vaccine Immunol ; 21(8): 1070-6, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24872513

ABSTRACT

Equine herpesvirus type 1 (EHV-1) is a major cause of winter pyrexia in racehorses in two training centers (Ritto and Miho) in Japan. Until the epizootic period of 2008-2009, a vaccination program using a killed EHV-1 vaccine targeted only susceptible 3-year-old horses with low antibody levels to EHV-1 antigens. However, because the protective effect was not satisfactory, in 2009-2010 the vaccination program was altered to target all 3-year-old horses. To evaluate the vaccine's efficacy, we investigated the number of horses with pyrexia due to EHV-1 or equine herpesvirus type 4 (EHV-4) infection or both and examined the vaccination coverage in the 3-year-old population and in the whole population before and after changes in the program. The mean (± standard deviation [SD]) estimated numbers of horses infected with EHV-1 or EHV-4 or both, among pyretic horses from 1999-2000 to 2008-2009 were 105 ± 47 at Ritto and 66 ± 44 at Miho. Although the estimated number of infected horses did not change greatly in the first period of the current program, it decreased from the second period, with means (±SD) of 21 ± 12 at Ritto and 14 ± 15 at Miho from 2010-2011 to 2012-2013. Vaccination coverage in the 3-year-old population was 99.4% at Ritto and 99.8% at Miho in the first period, and similar values were maintained thereafter. Coverage in the whole population increased more gradually than that in the 3-year-old population. The results suggest that EHV-1 epizootics can be suppressed by maintaining high vaccination coverage, not only in the 3-year-old population but also in the whole population.


Subject(s)
Fever/veterinary , Herpesviridae Infections/immunology , Herpesvirus 1, Equid/immunology , Horse Diseases/immunology , Vaccination/veterinary , Animals , Antibodies, Viral/blood , Fever/immunology , Fever/prevention & control , Fever/virology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/virology , Herpesvirus 4, Equid/immunology , Herpesvirus Vaccines/immunology , Horse Diseases/prevention & control , Horse Diseases/virology , Horses , Japan , Mass Vaccination , Vaccination/statistics & numerical data , Vaccines, Inactivated/immunology
20.
Article in English | MEDLINE | ID: mdl-23706920

ABSTRACT

OBJECTIVES: To assess apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) expression in oral squamous cell carcinoma (OSCC) and analyze its clinical and pathological significance. STUDY DESIGN: ASC expression was studied using immunohistochemistry in 119 OSCCs patients. The relationships between ASC expression and clinical and pathological parameters were statistically analyzed. In addition, the relationships between ASC expression and cell differentiation [IVL (involcrin) expression] and apoptosis (TUNEL [TdT-mediated dUTP nick end labeling] positive cell number) were investigated. RESULTS: ASC expression showed significant correlations with parameters including clinical tumor stage, mode of invasion, and histological differentiation, and had a significant impact on survival of OSCC. The distribution of ASC correlated well with that of IVL. ASC expression was significantly correlated with the TUNEL-positive cell number. CONCLUSIONS: Lower ASC expression correlates with clinical and pathological malignancy and, consequently, poor prognosis of OSCC. ASC has a close association with cell differentiation and apoptosis.


Subject(s)
Apoptosis/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , Cytoskeletal Proteins/metabolism , Mouth Neoplasms/pathology , Protein Precursors/metabolism , Adult , Aged , Aged, 80 and over , CARD Signaling Adaptor Proteins , Carcinoma, Squamous Cell/metabolism , Cell Differentiation , Cell Line, Tumor , Female , Fluorescent Antibody Technique , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Mouth Neoplasms/metabolism , Real-Time Polymerase Chain Reaction , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...