Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Med Virol ; 96(1): e29346, 2024 01.
Article in English | MEDLINE | ID: mdl-38178580

ABSTRACT

Orthohantaviruses, etiological agents of hemorrhagic fever with renal syndrome (HFRS) and hantavirus cardiopulmonary syndrome, pose a critical public health threat worldwide. Hantaan orthohantavirus (HTNV) outbreaks are particularly endemic in Gyeonggi Province in northern area of the Republic of Korea (ROK). Small mammals were collected from three regions in the Gyeonggi Province during 2017 and 2018. Serological and molecular prevalence of HTNV was 25/201 (12.4%) and 10/25 (40%), respectively. A novel nanopore-based diagnostic assay using a cost-efficient Flongle chip was developed to rapidly and sensitively detect HTNV infection in rodent specimens within 3 h. A rapid phylogeographical surveillance of HTNV at high-resolution phylogeny was established using the amplicon-based Flongle sequencing. In total, seven whole-genome sequences of HTNV were newly obtained from wild rodents collected in Paju-si (Gaekhyeon-ri) and Yeoncheon-gun (Hyeonga-ri and Wangnim-ri), Gyeonggi Province. Phylogenetic analyses revealed well-supported evolutionary divergence and genetic diversity, enhancing the resolution of the phylogeographic map of orthohantaviruses in the ROK. Incongruences in phylogenetic patterns were identified among HTNV tripartite genomes, suggesting differential evolution for each segment. These findings provide crucial insights into on-site diagnostics, genome-based surveillance, and the evolutionary dynamics of orthohantaviruses to mitigate hantaviral outbreaks in HFRS-endemic areas in the ROK.


Subject(s)
Hantaan virus , Hemorrhagic Fever with Renal Syndrome , Orthohantavirus , Animals , Phylogeny , Hantaan virus/genetics , Orthohantavirus/genetics , Rodentia , Mammals , Republic of Korea/epidemiology
2.
PLoS Pathog ; 17(1): e1009179, 2021 01.
Article in English | MEDLINE | ID: mdl-33471866

ABSTRACT

Primary effusion lymphoma (PEL) is an aggressive B cell lymphoma that is etiologically linked to Kaposi's sarcoma-associated herpesvirus (KSHV). Despite standard multi-chemotherapy treatment, PEL continues to cause high mortality. Thus, new strategies to control PEL are needed urgently. Here, we show that a phosphodegron motif within the KSHV protein, latency-associated nuclear antigen (LANA), specifically interacts with E3 ubiquitin ligase FBW7, thereby competitively inhibiting the binding of the anti-apoptotic protein MCL-1 to FBW7. Consequently, LANA-FBW7 interaction enhances the stability of MCL-1 by preventing its proteasome-mediated degradation, which inhibits caspase-3-mediated apoptosis in PEL cells. Importantly, MCL-1 inhibitors markedly suppress colony formation on soft agar and tumor growth of KSHV+PEL/BCBL-1 in a xenograft mouse model. These results strongly support the conclusion that high levels of MCL-1 expression enable the oncogenesis of PEL cells and thus, MCL-1 could be a potential drug target for KSHV-associated PEL. This work also unravels a mechanism by which an oncogenic virus perturbs a key component of the ubiquitination pathway to induce tumorigenesis.


Subject(s)
Antigens, Viral/metabolism , F-Box-WD Repeat-Containing Protein 7/metabolism , Herpesvirus 8, Human/physiology , Lymphoma, Primary Effusion/virology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Nuclear Proteins/metabolism , Sarcoma, Kaposi/virology , Amino Acid Sequence , Animals , Antigens, Viral/genetics , Apoptosis , Cell Proliferation , F-Box-WD Repeat-Containing Protein 7/genetics , Female , Humans , Lymphoma, Primary Effusion/genetics , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Nuclear Proteins/genetics , Phosphorylation , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/pathology , Tumor Cells, Cultured , Ubiquitination , Xenograft Model Antitumor Assays
3.
J Med Virol ; 95(7): e28894, 2023 07.
Article in English | MEDLINE | ID: mdl-37386895

ABSTRACT

Severe fever with thrombocytopenia syndrome virus (SFTSV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can cause the hyperproduction of inflammatory cytokines, which have pathological effects in patient including severe or fatal cytokine storms. To characterize the effect of SFTSV and SARS-CoV-2 infection on the production of cytokines in severe fever with thrombocytopenia syndrome (SFTS) and COVID-19 patients, we performed an analysis of cytokines in SFTS and COVID-19 patients and also investigated the role of interleukin-10 (IL-10) in vitro studies: lipopolysaccharide-induced THP-1-derived macrophages, SFTSV infection of THP-1 cells, and SARS-CoV-2 infection of THP-1 cells. In this study, we found that levels of both IL-10 and IL-6 were significantly elevated, the level of transforming growth factor-ß (TGF-ß) was significantly decreased and IL-10 was elevated earlier than IL-6 in severe and critical COVID-19 and fatal SFTS patients, and inhibition of IL-10 signaling decreased the production of IL-6 and elevated that of TGF-ß. Therefore, the hyperproduction of IL-10 and IL-6 and the low production of TGF-ß have been linked to cytokine storm-induced mortality in fatal SFTS and severe and critically ill COVID-19 patients and that IL-10 can play an important role in the host immune response to severe and critical SARS-CoV-2 and fatal SFTSV infection.


Subject(s)
COVID-19 , Severe Fever with Thrombocytopenia Syndrome , Humans , Cytokine Release Syndrome , Cytokines , Interleukin-10 , Interleukin-6 , SARS-CoV-2 , Transforming Growth Factor beta
4.
J Med Virol ; 95(9): e29099, 2023 09.
Article in English | MEDLINE | ID: mdl-37702580

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne disease with high mortality in Eastern Asia. The disease is caused by the SFTS virus (SFTSV), also known as Dabie bandavirus, which has a segmented RNA genome consisting of L, M, and S segments. Previous studies have suggested differential viral virulence depending on the genotypes of SFTSV; however, the critical viral factor involved in the differential viral virulence is unknown. Here, we found a significant difference in viral replication in vitro and virulence in vivo between two Korean isolates belonging to the F and B genotypes, respectively. By generating viral reassortants using the two viral strains, we demonstrated that the L segment, which encodes viral RNA-dependent RNA polymerase (RdRp), is responsible for the enhanced viral replication and virulence. Comparison of amino acid sequences and viral replication rates revealed a point variation, E251K, on the surface of RdRp to be the most significant determinant for the enhanced viral replication rate and in vivo virulence. The effect of the variation was further confirmed using recombinant SFTSV generated by reverse genetic engineering. Therefore, our results indicate that natural variations affecting the viral replicase activity could significantly contribute to the viral virulence of SFTSV.


Subject(s)
Severe Fever with Thrombocytopenia Syndrome , Humans , Virulence , DNA-Directed RNA Polymerases/genetics , Virus Replication , RNA-Dependent RNA Polymerase/genetics
5.
Clin Infect Dis ; 75(4): 596-603, 2022 09 10.
Article in English | MEDLINE | ID: mdl-34893799

ABSTRACT

BACKGROUND: Middle East respiratory syndrome (MERS) is a highly lethal respiratory disease caused by a zoonotic betacoronavirus. The development of effective vaccines and control measures requires a thorough understanding of the immune response to this viral infection. METHODS: We investigated cellular immune responses up to 5 years after infection in a cohort of 59 MERS survivors by performing enzyme-linked immunospot assay and intracellular cytokine staining after stimulation of peripheral blood mononuclear cells with synthetic viral peptides. RESULTS: Memory T-cell responses were detected in 82%, 75%, 69%, 64%, and 64% of MERS survivors from 1-5 years post-infection, respectively. Although the frequency of virus-specific interferon gamma (IFN-γ)-secreting T cells tended to be higher in moderately/severely ill patients than in mildly ill patients during the early period of follow-up, there was no significant difference among the different clinical severity groups across all time points. While both CD4+ and CD8+ T cells were involved in memory T-cell responses, CD4+ T cells persisted slightly longer than CD8+ T cells. Both memory CD4+ and CD8+ T cells recognized the E/M/N proteins better than the S protein and maintained their polyfunctionality throughout the period examined. Memory T-cell responses correlated positively with antibody responses during the initial 3-4 years but not with maximum viral loads at any time point. CONCLUSIONS: These findings advance our understanding of the dynamics of virus-specific memory T-cell immunity after MERS-coronavirus infection, which is relevant to the development of effective T cell-based vaccines.


Subject(s)
Coronavirus Infections , Middle East Respiratory Syndrome Coronavirus , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Humans , Immunologic Memory , Leukocytes, Mononuclear , Memory T Cells , Survivors
6.
J Korean Med Sci ; 37(8): e67, 2022 Feb 28.
Article in English | MEDLINE | ID: mdl-35226425

ABSTRACT

We investigated the kinetics of the neutralizing antibody responses to the severe acute respiratory syndrome-coronavirus-2 delta variant over the course of 1 year in 16 patients infected at the beginning of the pandemic. In patients with severe disease, neutralizing responses to the delta variant were detectable, albeit at lower levels than responses to the wild type. Neutralizing responses to the delta variant were undetectable, however, in asymptomatic persons. This finding implies that the vaccination strategy for persons with past natural infection should depend on the severity of the previous infection.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19/immunology , SARS-CoV-2/immunology , Adult , Aged , COVID-19/prevention & control , COVID-19 Vaccines/immunology , Female , Humans , Kinetics , Male , Middle Aged , Severity of Illness Index , Vaccination , Young Adult
7.
Int J Mol Sci ; 24(1)2022 Dec 30.
Article in English | MEDLINE | ID: mdl-36614125

ABSTRACT

The use of vaccines is the most effective and reliable method for the prevention of viral infections. However, research on evaluation of effective therapeutic agents for use in treatment after infection is necessary. Zanamivir was administered through inhalation for treatment of pandemic influenza A/H1N1 in 2009. However, the emergence of drug-resistant strains can occur rapidly. Alloferon, an immunomodulatory drug developed as an NK cell activator, exerts antiviral effects against various viruses, particularly influenza viruses. Therefore, alloferon and zanamivir were administered in combination in an effort to improve the antiviral effect of zanamivir by reducing H1N1 resistance. First, we confirmed that administration of combined treatment would result in effective inhibition of viral proliferation in MDCK and A549 cells infected with H1N1. Production of IL-6 and MIP-1α in these cells and the activity of p38 MAPK and c-Jun that are increased by H1N1 were inhibited by combined treatment. Mice were then infected intranasally with H1N1, and examination of the antiviral efficacy of the alloferon/zanamivir combination was performed. The results showed that combined treatment after infection with H1N1 prevented weight loss, increased the survival rate, and improved lung fibrosis. Combined treatment also resulted in reduced infiltration of neutrophils and macrophages into the lungs. Combined treatment effectively inhibited the activity of p38 MAPK and c-Jun in lung tissue, which was increased by infection with H1N1. Therefore, the combination of alloferon/zanamivir effectively prevents the development of H1N1-mediated inflammation in the lungs by inhibiting the production of inflammatory mediators and migration of inflammatory cells into lung tissue.


Subject(s)
Antiviral Agents , Orthomyxoviridae Infections , Zanamivir , Animals , Humans , Mice , Antiviral Agents/pharmacology , Drug Resistance, Viral , Influenza A Virus, H1N1 Subtype , Neuraminidase , Oseltamivir/pharmacology , Zanamivir/pharmacology , Orthomyxoviridae Infections/drug therapy
8.
Clin Infect Dis ; 73(3): e550-e558, 2021 08 02.
Article in English | MEDLINE | ID: mdl-32898238

ABSTRACT

BACKGROUND: Zoonotic coronaviruses have emerged as a global threat by causing fatal respiratory infections. Given the lack of specific antiviral therapies, application of human convalescent plasma retaining neutralizing activity could be a viable therapeutic option that can bridges this gap. METHODS: We traced antibody responses and memory B cells in peripheral blood collected from 70 recovered Middle East respiratory syndrome coronavirus (MERS-CoV) patients for 3 years after the 2015 outbreak in South Korea. We also used a mouse infection model to examine whether the neutralizing activity of collected sera could provide therapeutic benefit in vivo upon lethal MERS-CoV challenge. RESULTS: Anti-spike-specific IgG responses, including neutralizing activity and antibody-secreting memory B cells, persisted for up to 3 years, especially in MERS patients who suffered from severe pneumonia. Mean antibody titers gradually decreased annually by less than 2-fold. Levels of antibody responses were significantly correlated with fever duration, viral shedding periods, and maximum viral loads observed during infection periods. In a transgenic mice model challenged with lethal doses of MERS-CoV, a significant reduction in viral loads and enhanced survival was observed when therapeutically treated with human plasma retaining a high neutralizing titer (> 1/5000). However, this failed to reduce pulmonary pathogenesis, as revealed by pathological changes in lungs and initial weight loss. CONCLUSIONS: High titers of neutralizing activity are required for suppressive effect on the viral replication but may not be sufficient to reduce inflammatory lesions upon fatal infection. Therefore, immune sera with high neutralizing activity must be carefully selected for plasma therapy of zoonotic coronavirus infection.


Subject(s)
Coronavirus Infections , Middle East Respiratory Syndrome Coronavirus , Animals , Antibodies, Neutralizing , Antibodies, Viral , Coronavirus Infections/drug therapy , Humans , Mice , Republic of Korea , Spike Glycoprotein, Coronavirus
9.
J Virol ; 94(24)2020 11 23.
Article in English | MEDLINE | ID: mdl-32967955

ABSTRACT

Middle East respiratory syndrome coronavirus (MERS-CoV) causes severe respiratory illness and has a high mortality of ∼34%. However, since its discovery in 2012, an effective vaccine has not been developed for it. To develop a vaccine against multiple strains of MERS-CoV, we targeted spike glycoprotein (S) using prime-boost vaccination with DNA and insect cell-expressed recombinant proteins for the receptor-binding domain (RBD), S1, S2, SΔTM, or SΔER. Our S subunits were generated using an S sequence derived from the MERS-CoV EMC/2012 strain. We examined humoral and cellular immune responses of various combinations with DNA plasmids and recombinant proteins in mice. Mouse sera immunized with SΔER DNA priming/SΔTM protein boosting showed cross-neutralization against 15 variants of S-pseudovirions and the wild-type KOR/KNIH/002 strain. In addition, these immunizations provided full protection against the KOR/KNIH/002 strain challenge in human DPP4 knock-in mice. These findings suggest that vaccination with the S subunits derived from one viral strain can provide cross-protection against variant MERS-CoV strains with mutations in S. DNA priming/protein boosting increased gamma interferon production, while protein-alone immunization did not. The RBD subunit alone was insufficient to induce neutralizing antibodies, suggesting the importance of structural conformation. In conclusion, heterologous DNA priming with protein boosting is an effective way to induce both neutralizing antibodies and cell-mediated immune responses for MERS-CoV vaccine development. This study suggests a strategy for selecting a suitable platform for developing vaccines against MERS-CoV or other emerging coronaviruses.IMPORTANCE Coronavirus is an RNA virus with a higher mutation rate than DNA viruses. Therefore, a mutation in S-protein, which mediates viral infection by binding to a human cellular receptor, is expected to cause difficulties in vaccine development. Given that DNA-protein vaccines promote stronger cell-mediated immune responses than protein-only vaccination, we immunized mice with various combinations of DNA priming and protein boosting using the S-subunit sequences of the MERS-CoV EMC/2012 strain. We demonstrated a cross-protective effect against wild-type KOR/KNIH/002, a strain with two mutations in the S amino acids, including one in its RBD. The vaccine also provided cross-neutralization against 15 different S-pseudotyped viruses. These suggested that a vaccine targeting one variant of S can provide cross-protection against multiple viral strains with mutations in S. The regimen of DNA priming/Protein boosting can be applied to the development of other coronavirus vaccines.


Subject(s)
Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Cross Protection , Middle East Respiratory Syndrome Coronavirus/immunology , Spike Glycoprotein, Coronavirus/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Coronavirus Infections/mortality , Coronavirus Infections/virology , Disease Models, Animal , Female , Humans , Immunity, Cellular , Immunization, Secondary , Immunogenicity, Vaccine , Mice , Plasmids/administration & dosage , Plasmids/genetics , Plasmids/immunology , Spike Glycoprotein, Coronavirus/genetics , Vaccination , Vaccines, DNA/administration & dosage , Viral Vaccines/administration & dosage
10.
Emerg Infect Dis ; 26(8): 1878-1881, 2020 08.
Article in English | MEDLINE | ID: mdl-32687023

ABSTRACT

Serologic and molecular surveillance of serum collected from 152 suspected scrub typhus patients in Myanmar revealed Orientia tsutsugamushi of genotypic heterogeneity. In addition, potential co-infection with severe fever with thrombocytopenia syndrome virus was observed in 5 (3.3%) patients. Both scrub typhus and severe fever with thrombocytopenia syndrome are endemic in Myanmar.


Subject(s)
Coinfection , Orientia tsutsugamushi , Scrub Typhus , Thrombocytopenia , Coinfection/epidemiology , Humans , Myanmar/epidemiology , Orientia , Orientia tsutsugamushi/genetics , Scrub Typhus/complications , Scrub Typhus/diagnosis , Scrub Typhus/epidemiology
11.
Biotechnol Bioeng ; 117(7): 1990-2007, 2020 07.
Article in English | MEDLINE | ID: mdl-32297972

ABSTRACT

High-quality antibody (Ab) production depends on the availability of immunologically relevant antigens. We present a potentially universal platform for generating soluble antigens from bacterial hosts, tailored to immunized animals for Ab production. A novel RNA-dependent chaperone, in which the target antigen is genetically fused with an RNA-interacting domain (RID) docking tag derived from the immunized host, promotes the solubility and robust folding of the target antigen. We selected the N-terminal tRNA-binding domain of lysyl-tRNA synthetase (LysRS) as the RID for fusion with viral proteins and demonstrated the expression of the RID fusion proteins in their soluble and native conformations; immunization predominantly elicited Ab responses to the target antigen, whereas the "self" RID tag remained nonimmunogenic. Differential immunogenicity of the fusion proteins greatly enriched and simplified the screening of hybridoma clones of monoclonal antibodies (mAbs), enabling specific and sensitive serodiagnosis of MERS-CoV infection. Moreover, mAbs against the consensus influenza hemagglutinin stalk domain enabled a novel assay for trivalent seasonal influenza vaccines. The Fc-mediated effector function was demonstrated, which could be harnessed for the design of next-generation "universal" influenza vaccines. The nonimmunogenic built-in antigen folding module tailored to a repertoire of immunized animal hosts will drive immunochemical diagnostics, therapeutics, and designer vaccines.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antibodies, Viral/biosynthesis , Antigens, Viral/chemistry , Coronavirus Infections/diagnosis , Hybridomas/immunology , Middle East Respiratory Syndrome Coronavirus/immunology , Molecular Chaperones , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/immunology , Antibodies, Viral/isolation & purification , Antigens, Viral/genetics , Antigens, Viral/immunology , Coronavirus Infections/blood , Enzyme-Linked Immunosorbent Assay , Female , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunization , Influenza Vaccines , Lysine-tRNA Ligase/chemistry , Lysine-tRNA Ligase/genetics , Mice , Mice, Inbred BALB C , Protein Conformation , Protein Domains , Protein Folding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Serologic Tests , Solubility
12.
Emerg Infect Dis ; 25(5): 1029-1031, 2019 05.
Article in English | MEDLINE | ID: mdl-31002059

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS), a tickborne viral disease, has been identified in China, South Korea, and Japan since 2009. We found retrospective evidence of SFTS virus (SFTSV) infection in Vietnam, which suggests that SFTSV infections also occur in Vietnam, where the virus has not been known to be endemic.


Subject(s)
Bunyaviridae Infections/epidemiology , Bunyaviridae Infections/virology , Phlebovirus , Thrombocytopenia/epidemiology , Thrombocytopenia/virology , Tick-Borne Diseases/epidemiology , Tick-Borne Diseases/virology , Adult , Bunyaviridae Infections/diagnosis , Female , Genes, Viral , Humans , Male , Phlebovirus/classification , Phlebovirus/genetics , Phlebovirus/isolation & purification , Phylogeny , Public Health Surveillance , Symptom Assessment , Thrombocytopenia/diagnosis , Tick-Borne Diseases/diagnosis , Vietnam/epidemiology
13.
Emerg Infect Dis ; 25(6): 1161-1168, 2019 06.
Article in English | MEDLINE | ID: mdl-30900977

ABSTRACT

The unexpectedly large outbreak of Middle East respiratory syndrome in South Korea in 2015 was initiated by an infected traveler and amplified by several "superspreading" events. Previously, we reported the emergence and spread of mutant Middle East respiratory syndrome coronavirus bearing spike mutations (I529T or D510G) with reduced affinity to human receptor CD26 during the outbreak. To assess the potential association of spike mutations with superspreading events, we collected virus genetic information reported during the outbreak and systemically analyzed the relationship of spike sequences and epidemiology. We found sequential emergence of the spike mutations in 2 superspreaders. In vivo virulence of the mutant viruses seems to decline in human patients, as assessed by fever duration in affected persons. In addition, neutralizing activity against these 2 mutant viruses in serum samples from mice immunized with wild-type spike antigen were gradually reduced, suggesting emergence and wide spread of neutralization escapers during the outbreak.


Subject(s)
Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/virology , Coronavirus Infections/epidemiology , Coronavirus Infections/virology , Middle East Respiratory Syndrome Coronavirus/genetics , Mutation , Adult , Aged , Antibodies, Neutralizing/immunology , Communicable Diseases, Emerging/history , Communicable Diseases, Emerging/immunology , Coronavirus Infections/history , Coronavirus Infections/immunology , Disease Outbreaks , Female , Genotype , History, 21st Century , Humans , Male , Middle Aged , Middle East Respiratory Syndrome Coronavirus/immunology , Neutralization Tests , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology
14.
Biochim Biophys Acta Mol Cell Res ; 1864(1): 1-11, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27751885

ABSTRACT

Members of the herpesviral family use multiple strategies to hijack infected host cells and exploit cellular signaling for their pathogenesis and latent infection. Among the most intriguing weapons in the arsenal of pathogenic herpesviruses are the constitutively active virally-encoded G protein-coupled receptors (vGPCRs). Even though vGPCRs contribute to viral pathogenesis such as immune evasion and proliferative disorders, the molecular details of how vGPCRs continuously activate cellular signaling are largely unknown. Here, we report that the vGPCR of Herpesvirus saimiri (HVS), an oncogenic γ2-herpesvirus, constitutively activates T cells via a heteromeric interaction with cellular CXCR4. Constitutive T cell activation also occurs with expression of the vGPCR of Kaposi's sarcoma-associated herpesvirus (KSHV), but not the vGPCR of Epstein-Barr virus. Expression of HVS vGPCR down-regulated the surface expression of CXCR4 but did not induce the degradation of the chemokine receptor, suggesting that vGPCR/CXCR4 signaling continues in cytosolic compartments. The physical association of vGPCR with CXCR4 was demonstrated by proximity ligation assay as well as immunoprecipitation. Interestingly, the constitutive activation of T cells by HVS vGPCR is independent of proximal T cell receptor (TCR) signaling molecules, such as TCRß, Lck, and ZAP70, whereas CXCR4 silencing by shRNA abolished T cell activation by vGPCRs of HVS and KSHV. Furthermore, previously identified inactive vGPCR mutants failed to interact with CXCR4. These findings on the positive cooperativity of vGPCR with cellular CXCR4 in T cell activation extend our current understanding of the molecular mechanisms of vGPCR function and highlight the importance of heteromerization for GPCR activity.


Subject(s)
Herpesvirus 2, Saimiriine/metabolism , Herpesvirus 8, Human/metabolism , Receptors, CXCR4/genetics , Receptors, Chemokine/genetics , T-Lymphocytes/virology , Gene Expression Regulation , HEK293 Cells , Herpesvirus 2, Saimiriine/genetics , Herpesvirus 2, Saimiriine/growth & development , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/metabolism , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/growth & development , Host-Pathogen Interactions , Humans , Lymphocyte Activation , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Primary Cell Culture , Protein Binding , Protein Multimerization , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , ZAP-70 Protein-Tyrosine Kinase/genetics , ZAP-70 Protein-Tyrosine Kinase/immunology , ZAP-70 Protein-Tyrosine Kinase/metabolism
15.
Emerg Infect Dis ; 24(11): 2103-2105, 2018 11.
Article in English | MEDLINE | ID: mdl-30334706

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) was reported in China in 2009 and in South Korea in 2012. We found retrospective evidence of SFTS virus infection in South Korea in 2010, suggesting that infections in South Korea occurred before previously reported and were more concurrent with those in China.


Subject(s)
Bunyaviridae Infections/epidemiology , Phlebovirus/isolation & purification , Thrombocytopenia/epidemiology , Bunyaviridae Infections/virology , Humans , Phlebovirus/genetics , Phylogeny , Republic of Korea/epidemiology , Retrospective Studies , Thrombocytopenia/virology
16.
Microbiol Immunol ; 62(9): 574-584, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30117617

ABSTRACT

Middle East respiratory syndrome coronavirus (MERS-CoV) is a single-stranded RNA virus that causes severe respiratory disease in humans with a high fatality rate. Binding of the receptor binding domain (RBD) of the spike (S) glycoprotein to dipeptidyl peptidase 4 is the critical step in MERS-CoV infection of a host cell. No vaccines or clinically applicable treatments are currently available for MERS-CoV. Therefore, rapid diagnosis is important for improving patient outcomes through prompt treatment and protection against viral outbreaks. In this study, the aim was to establish two ELISA systems for detecting antigens and antibodies against MERS-CoV. Using a recombinant full-length S protein, an indirect ELISA was developed and found to detect MERS-CoV-specific antibodies in animal sera and sera of patient with MERS. Moreover, MAbs were induced with the recombinant S protein and RBD and used for sandwich ELISA to detect the MERS-CoV S protein. Neither ELISA system exhibited significant intra-assay or inter-assay variation, indicating good reproducibility. Moreover, the inter-day precision and sensitivity were adequate for use as a diagnostic kit. Thus, these ELISAs can be used clinically to diagnose MERS-CoV.


Subject(s)
Antibodies, Viral/isolation & purification , Antigens, Viral/isolation & purification , Coronavirus Infections/diagnosis , Coronavirus Infections/immunology , Immunologic Tests/methods , Middle East Respiratory Syndrome Coronavirus/immunology , Spike Glycoprotein, Coronavirus/immunology , Animals , Antibodies, Monoclonal , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antigens, Viral/immunology , Baculoviridae/genetics , Coronavirus Infections/virology , Enzyme-Linked Immunosorbent Assay/methods , Escherichia coli/genetics , Gene Expression , Humans , Middle East Respiratory Syndrome Coronavirus/genetics , Protein Binding/genetics , Protein Binding/immunology , Protein Interaction Domains and Motifs , Rats , Rats, Wistar , Receptors, Virus/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Reproducibility of Results , Sensitivity and Specificity , Sf9 Cells , Spike Glycoprotein, Coronavirus/genetics , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology
17.
Biochim Biophys Acta ; 1853(3): 619-31, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25549939

ABSTRACT

Cystamine and its reduced form cysteamine showed protective effects in various models of neurodegenerative disease, including Huntington's disease and Parkinson's disease. Other lines of evidence demonstrated the cytotoxic effect of cysteamine on duodenal mucosa leading to ulcer development. However, the mechanism for cystamine cytotoxicity remains poorly understood. Here, we report a new pathway in which cystamine induces apoptosis by targeting apoptosis-inducing factor (AIF). By screening of various cell lines, we observed that cystamine and cysteamine induce cell death in a cell type-specific manner. Comparison between cystamine-sensitive and cystamine-resistant cell lines revealed that cystamine cytotoxicity is not associated with unfolded protein response, reactive oxygen species generation and transglutaminase or caspase activity; rather, it is associated with the ability of cystamine to trigger AIF nuclear translocation. In cystamine-sensitive cells, cystamine suppresses the levels of intracellular glutathione by inhibiting γ-glutamylcysteine synthetase expression that triggers AIF translocation. Conversely, glutathione supplementation completely prevents cystamine-induced AIF translocation and apoptosis. In rats, cysteamine administration induces glutathione depletion and AIF translocation leading to apoptosis of duodenal epithelium. These results indicate that AIF translocation through glutathione depletion is the molecular mechanism of cystamine toxicity, and provide important implications for cystamine in the neurodegenerative disease therapeutics as well as in the regulation of AIF-mediated cell death.


Subject(s)
Apoptosis Inducing Factor/physiology , Apoptosis/drug effects , Cystamine/pharmacology , Glutathione/metabolism , Animals , Apoptosis/genetics , Duodenal Ulcer/metabolism , Duodenal Ulcer/pathology , Female , HeLa Cells , Humans , MCF-7 Cells , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured , Up-Regulation/drug effects
18.
J Nanobiotechnology ; 14(1): 76, 2016 Nov 26.
Article in English | MEDLINE | ID: mdl-27887623

ABSTRACT

BACKGROUND: Zinc oxide nanoparticle (ZNP) has been applied in various biomedical fields. Here, we investigated the usage of ZNP as an antigen carrier for vaccine development by combining a high affinity peptide to ZNP. RESULTS: A novel zinc oxide-binding peptide (ZBP), FPYPGGDA, with high affinity to ZNP (K a  = 2.26 × 106 M-1) was isolated from a random peptide library and fused with a bacterial antigen, ScaA of Orientia tsutsugamushi, the causative agent of scrub typhus. The ZNP/ZBP-ScaA complex was efficiently phagocytosed by a dendritic cell line, DC2.4, in vitro and significantly enhanced anti-ScaA antibody responses in vivo compared to control groups. In addition, immunization with the ZNP/ZBP-ScaA complex promoted the generation of IFN-γ-secreting T cells in an antigen-dependent manner. Finally, we observed that ZNP/ZBP-ScaA immunization provided protective immunity against lethal challenge of O. tsutsugamushi, indicating that ZNP can be used as a potent adjuvant when complexed with ZBP-conjugated antigen. CONCLUSIONS: ZNPs possess good adjuvant potential as a vaccine carrier when combined with an antigen having a high affinity to ZNP. When complexed with ZBP-ScaA antigen, ZNPs could induce strong antibody responses as well as protective immunity against lethal challenges of O. tsutsugamushi. Therefore, application of ZNPs combined with a specific soluble antigen could be a promising strategy as a novel vaccine carrier system.


Subject(s)
Antigens, Bacterial/immunology , Metal Nanoparticles/chemistry , Orientia tsutsugamushi/metabolism , Scrub Typhus/prevention & control , Zinc Oxide/chemistry , Amino Acid Sequence , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Biocompatible Materials/chemistry , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Female , Mice , Mice, Inbred C57BL , Orientia tsutsugamushi/immunology , Peptides/chemistry , Phagocytosis , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Scrub Typhus/veterinary , Vaccination
19.
J Appl Toxicol ; 35(6): 593-602, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25572658

ABSTRACT

Therapeutic cancer vaccines promote immune responses by delivering tumour-specific antigens. Recently, we developed iron oxide (Fe3 O4 )-zinc oxide (ZnO) core-shell nanoparticles (CSNPs) as carriers for antigen delivery into dendritic cells (DCs), and the CSNPs were injected subcutaneously into C57BL/6 mice to examine the systemic toxicity, tissue distribution and excretion of the CSNPs. The doses injected were 0, 4, 20 and 200 mg kg(-1) weekly for 4 weeks. No significant changes were observed after the CSNPs administration with respect to mortality, clinical observations, body weight, food intake, water consumption, urinalysis, haematology, serum biochemistry,and organ weights. A dose-dependent increase in granulomatous inflammation was observed at the injection site of the CSNP-treated animals, but no other histopathological lesions in other organs could be attributed to the CSNPs. The Zn concentration, which is an indicator for CSNPs, was not significantly higher in the sampled tissues, urine, or faeces after the CSNP injection. In contrast, the Zn concentration at the subcutaneous skin of the site injected with the CSNPs increased in a dose-dependent manner, along with a macroscopic deposition of the CSNPs. The CSNP residue at the injection site resulted in a foreign body response with the appearance of macrophage infiltration, but otherwise did not show any systemic distribution or toxicity at up to 200 mg kg(-1) during this study. In conclusion, CSNPs could be used as good antigen carriers for DC-based immunotherapy, although further study is needed to completely clear the residue of the CSNPs at the injection site.


Subject(s)
Ferric Compounds/toxicity , Metal Nanoparticles/toxicity , Zinc Oxide/toxicity , Animals , Female , Ferric Compounds/administration & dosage , Ferric Compounds/pharmacokinetics , Foreign-Body Reaction/chemically induced , Foreign-Body Reaction/pathology , Injections, Subcutaneous , Metal Nanoparticles/administration & dosage , Mice , Mice, Inbred C57BL , Tissue Distribution , Zinc Oxide/administration & dosage , Zinc Oxide/pharmacokinetics
20.
Biochem Biophys Res Commun ; 447(1): 192-6, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24704446

ABSTRACT

Calcium acts as a second messenger and plays a crucial role in signaling pathways involved in cell proliferation. Recently, calcium channels related to calcium influx into the cytosol of epithelial cells have attracted attention as a cancer therapy target. Of these calcium channels, TRPV6 is overexpressed in prostate cancer and is considered an important molecule in the process of metastasis. However, its exact role and mechanism is unclear. NUMB, well-known tumor suppressor gene, is a novel interacting partner of TRPV6. We show that NUMB and TRPV6 have a reciprocal positive regulatory relationship in PC-3 cells. We repeated this experiment in two other prostate cancer cell lines, DU145 and LNCaP. Interestingly, there were no significant changes in TRPV6 expression following NUMB knockdown in DU145. We revealed that the presence or absence of PTEN was the cause of NUMB-TRPV6 function. Loss of PTEN caused a positive correlation of TRPV6-NUMB expression. Collectively, we determined that PTEN is a novel interacting partner of TRPV6 and NUMB. These results demonstrated a novel relationship of NUMB-TRPV6 in prostate cancer cells, and show that PTEN is a novel regulator of this complex.


Subject(s)
Calcium Channels/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , PTEN Phosphohydrolase/deficiency , Prostatic Neoplasms/physiopathology , TRPV Cation Channels/metabolism , Calcium Channels/drug effects , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Male , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , PTEN Phosphohydrolase/physiology , Prostatic Neoplasms/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL