Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
J Virol ; : e0049924, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38953631

RESUMO

Tibroviruses are novel rhabdoviruses detected in humans, cattle, and arthropods. Four tibroviruses are known to infect humans: Bas-Congo virus (BASV), Ekpoma virus 1 (EKV-1), Ekpoma virus 2, and Mundri virus. However, since none of them has been isolated, their biological properties are largely unknown. We aimed to characterize the human tibrovirus glycoprotein (G), which likely plays a pivotal role in viral tropism and pathogenicity. Human tibrovirus Gs were found to share some primary structures and display 14 conserved cysteine residues, although their overall amino acid homology was low (29%-48%). Multiple potential glycosylation sites were found on the G molecules, and endoglycosidase H- and peptide-N-glycosidase F-sensitive glycosylation was confirmed. AlphaFold-predicted three-dimensional (3D) structures of human tibrovirus Gs were overall similar. Membrane fusion mediated by these tibrovirus Gs was induced by acidic pH. The low pH-induced conformational change that triggers fusion was reversible. Virus-like particles (VLPs) were produced by transient expression of Gs in cultured cells and used to produce mouse antisera. Using vesicular stomatitis Indiana virus pseudotyped with Gs, we found that the antisera to the respective tibrovirus VLPs showed limited cross-neutralizing activity. It was also found that human C-type lectins and T-cell immunoglobulin mucin 1 acted as attachment factors for G-mediated entry into cells. Interestingly, BASV-G showed the highest ability to utilize these molecules. The viruses infected a wide range of cell lines with preferential tropism for human-derived cells whereas the preference of EKV-1 was unique compared with the other human tibroviruses. These findings provide fundamental information to understand the biological properties of the human tibroviruses. IMPORTANCE: Human tibroviruses are poorly characterized emerging rhabdoviruses associated with either asymptomatic infection or severe disease with a case fatality rate as high as 60% in humans. However, the extent and burden of human infection as well as factors behind differences in infection outcomes are largely unknown. In this study, we characterized human tibrovirus glycoproteins, which play a key role in virus-host interactions, mainly focusing on their structural and antigenic differences and cellular tropism. Our results provide critical information for understanding the biological properties of these novel viruses and for developing appropriate preparedness interventions such as diagnostic tools, vaccines, and effective therapies.

2.
Ann Anat ; 253: 152227, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38336176

RESUMO

BACKGROUND: Vascularization is an essential event for both embryonic organ development and tissue repair in adults. During mouse tooth development, endothelial cells migrate into dental papilla during the cap stage, and form blood vessels through angiogenesis. Megakaryocytes and/or platelets, as other hematopoietic cells, express angiogenic molecules and can promote angiogenesis in adult tissues. However, it remains unknown which cells are responsible for attracting and leading blood vessels through the dental papilla during tooth development. METHODS: Here we analyzed the spatiotemporal expression of c-Mpl mRNA in developing molar teeth of fetal mice. Expression patterns were then compared with those of several markers of hematopoietic cells as well as of angiogenic elements including CD41, erythropoietin receptor, CD34, angiopoietin-1 (Ang-1), Tie-2, and vascular endothelial growth factor receptor2 (VEGFR2) through in situ hybridization or immunohistochemistry. RESULTS: Cells expressing c-Mpl mRNA was found in several parts of the developing tooth germ, including the peridental mesenchyme, dental papilla, enamel organ, and dental lamina. This expression occurred in a spatiotemporally controlled fashion. CD41-expressing cells were not detected during tooth development. The spatiotemporal expression pattern of c-Mpl mRNA in the dental papilla was similar to that of Ang-1, which preceded invasion of endothelial cells. Eventually, at the early bell stage, the c-Mpl mRNA signal was detected in morphologically differentiating odontoblasts that accumulated in the periphery of the dental papilla along the inner enamel epithelium layer of the future cusp region. CONCLUSION: During tooth development, several kinds of cells express c-Mpl mRNA in a spatiotemporally controlled fashion, including differentiating odontoblasts. We hypothesize that c-Mpl-expressing cells appearing in the forming dental papilla at the cap stage are odontoblast progenitor cells that migrate to the site of odontoblast differentiation. There they attract vascular endothelial cells into the forming dental papilla and lead cells toward the inner enamel epithelium layer through production of angiogenic molecules (e.g., Ang-1) during migration to the site of differentiation. C-Mpl may regulate apoptosis and/or proliferation of expressing cells in order to execute normal development of the tooth.


Assuntos
Dente , Fator A de Crescimento do Endotélio Vascular , Animais , Camundongos , Células Endoteliais , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Germe de Dente/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Virus Res ; 340: 199301, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38096954

RESUMO

Heartland virus (HRTV) causes generalized symptoms, severe shock, and multiple organ failure. We previously reported that interferon-α/ß receptor knockout (IFNAR-/-) mice infected intraperitoneally with 1 × 107 tissue culture-infective dose (TCID50) of HRTV died, while those subcutaneously infected with the same dose of HRTV did not. The pathophysiology of IFNAR-/- mice infected with HRTV and the mechanism underlying the difference in disease severity, which depends on HRTV infection route, were analyzed in this study. The liver, spleen, mesenteric and axillary lymph nodes, and gastrointestinal tract of intraperitoneally (I.P.) infected mice had pathological changes; however, subcutaneously (S.C.) infected mice only had pathological changes in the axillary lymph node and gastrointestinal tract. HRTV RNA levels in the mesenteric lymph node, lung, liver, spleen, kidney, stomach, intestine, and blood were significantly higher in I.P. infected mice than those in S.C. infected mice. Chemokine ligand-1 (CXCL-1), tumor necrosis factor (TNF)-α, interleukin (IL)-12, interferon (IFN)-γ, and IL-10 levels in plasma of I.P. infected mice were higher than those of S.C. infected mice. These results indicated that high levels of viral RNA and the induction of inflammatory responses in HRTV-infected IFNAR-/- mice may be associated with disease severity.


Assuntos
Bunyaviridae , Interferon Tipo I , Receptor de Interferon alfa e beta , Animais , Camundongos , Receptor de Interferon alfa e beta/genética , Camundongos Knockout , Interferons , Fígado , Interleucina-12
4.
Monoclon Antib Immunodiagn Immunother ; 41(1): 27-31, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35225659

RESUMO

Rabies is a highly neurotropic disease caused by rabies lyssavirus (RABV). Human rabies vaccines exist for pre- and postexposure prophylaxis; however, after clinical symptoms appear, the disease has an ∼100% mortality rate with no effective treatments available. In our previous study, mouse neuroblastoma cells transfected with a plasmid coding one clone of a single-chain variable fragment (scFv), scFv-P19, against RABV phosphoprotein (RABV-P) derived from an scFv phage-display library, before infection, exhibited reduced viral propagation after infection with the RABV-fixed strain, CVS11. In this study, we conducted epitope mapping of scFv-P19 through indirect fluorescent assay and Western blotting analysis against full-length and N- or C-terminal truncated RABV-P. Our results suggest that scFv-P19 targets a portion containing amino acids 47-52 at the N-terminus, which partially overlaps with the N-terminal nuclear export sequences. This provides insights into the underlying mechanism associated with inhibition of RABV by scFv-P19, while allowing for the design of additional scFv-based therapeutic studies for RABV by integrating appropriate delivery and application systems. Furthermore, the results of this study suggest that scFv-P19 may serve as an effective tool for investigating nuclear trafficking of RABV-P to explore the roles of RABV-P isoforms in rabies pathogenesis.


Assuntos
Vírus da Raiva , Raiva , Anticorpos de Cadeia Única , Animais , Anticorpos Monoclonais/farmacologia , Mapeamento de Epitopos , Camundongos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosfoproteínas/farmacologia , Vírus da Raiva/metabolismo , Anticorpos de Cadeia Única/genética
5.
PLoS Pathog ; 17(2): e1008859, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33534867

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) caused by a species Dabie bandavirus (formerly SFTS virus [SFTSV]) is an emerging hemorrhagic infectious disease with a high case-fatality rate. One of the best strategies for preventing SFTS is to develop a vaccine, which is expected to induce both humoral and cellular immunity. We applied a highly attenuated but still immunogenic vaccinia virus strain LC16m8 (m8) as a recombinant vaccine for SFTS. Recombinant m8s expressing SFTSV nucleoprotein (m8-N), envelope glycoprotein precursor (m8-GPC), and both N and GPC (m8-N+GPC) in the infected cells were generated. Both m8-GPC- and m8-N+GPC-infected cells were confirmed to produce SFTSV-like-particles (VLP) in vitro, and the N was incorporated in the VLP produced by the infection of cells with m8-N+GPC. Specific antibodies to SFTSV were induced in mice inoculated with each of the recombinant m8s, and the mice were fully protected from lethal challenge with SFTSV at both 103 TCID50 and 105 TCID50. In mice that had been immunized with vaccinia virus strain Lister in advance of m8-based SFTSV vaccine inoculation, protective immunity against the SFTSV challenge was also conferred. The pathological analysis revealed that mice immunized with m8-GPC or m8-N+GPC did not show any histopathological changes without any viral antigen-positive cells, whereas the control mice showed focal necrosis with inflammatory infiltration with SFTSV antigen-positive cells in tissues after SFTSV challenge. The passive serum transfer experiments revealed that sera collected from mice inoculated with m8-GPC or m8-N+GPC but not with m8-N conferred protective immunity against lethal SFTSV challenge in naïve mice. On the other hand, the depletion of CD8-positive cells in vivo did not abrogate the protective immunity conferred by m8-based SFTSV vaccines. Based on these results, the recombinant m8-GPC and m8-N+GPC were considered promising vaccine candidates for SFTS.


Assuntos
Antígenos Virais/imunologia , Nucleoproteínas/imunologia , Phlebovirus/imunologia , Febre Grave com Síndrome de Trombocitopenia/prevenção & controle , Vacinas Atenuadas/administração & dosagem , Vacinas Sintéticas/administração & dosagem , Proteínas do Envelope Viral/imunologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Febre Grave com Síndrome de Trombocitopenia/imunologia , Febre Grave com Síndrome de Trombocitopenia/virologia
6.
Virology ; 556: 124-132, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33561699

RESUMO

The heartland virus (HRTV) is a novel phlebovirus that causes severe infections in the USA and closely related to the severe fever thrombocytopenia syndrome virus (SFTSV), a causative agent for SFTS in Asia. The entry mechanisms of HRTV remain unclear. Here, we developed the pseudotyped vesicular stomatitis virus bearing the HRTV glycoprotein (GP) (HRTVpv), and the antigenicity and the entry mechanisms of HRTV were analyzed. HRTVpv was neutralized by anti-SFTSV Gc antibody, but not the anti-SFTSV Gn antibodies. Entry of HRTVpv to cells was inhibited by bafilomycin A1 and dynasore, and but it was enhanced in cells overexpressed with C-type lectins. Production of infectious HRTVpv and SFTSVpv was reduced by Nn-DNJ, α-glucosidase inhibitor. The entry of HRTV occurs via pH- and dynamin-dependent endocytosis. Furthermore, Nn-DNJ may be a possible therapeutic agent against HRTV and SFTSV.


Assuntos
Infecções por Bunyaviridae/virologia , Phlebovirus/patogenicidade , Estomatite Vesicular/virologia , Vesiculovirus/patogenicidade , Proteínas do Envelope Viral/metabolismo , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/farmacologia , Animais , Linhagem Celular , Cricetinae , Haplorrinos , Humanos , Camundongos , Internalização do Vírus
7.
Antiviral Res ; 182: 104926, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32882323

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS), an emerging viral infectious disease with a high case fatality rate, is caused by the SFTS virus (SFTSV). Although several cellular molecules involved in viral entry have been identified, the entry mechanisms of SFTSV remain unclear. In this study, we screened the protein kinase inhibitors in inhibitory effects on the infection of Vero cells with SFTSV using InhibitorSelect™ Protein Kinase Library Series (Merck & Co., Inc., Kenilworth, NJ, USA). Several types of inhibitors targeted to platelet-derived growth factor receptor ß (PDGFRß) inhibited the infection of Vero, Huh7, and NIH3T3 cells with SFTSV in a dose-dependent manner within the noncytotoxic range. In addition, these protein kinase inhibitors also inhibited the infection of the target cells with SFTSV glycoprotein (SFTSV-GP) pseudotyped virus (SFTSVpv). Activation of PDGFRß phosphorylation was detected in SFTSV-treated cells. The infectivities of SFTSVpv were specifically decreased not only in NIH3T3 cells treated with siRNA for PDGFRß but also in NIH3T3 cells treated with anti-PDGFRß neutralizing antibody in a dose-dependent manner. SFTSV growth and entry of SFTSVpv were also inhibited by Akt inhibitors. Activation of Akt phosphorylation was also detected in SFTSV-treated cells. These data indicate that PDGFRß is one of the important host factors in the entry steps of SFTSV.


Assuntos
Phlebovirus/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Febre Grave com Síndrome de Trombocitopenia/virologia , Internalização do Vírus , Animais , Benzimidazóis/farmacologia , Benzotiazóis/farmacologia , Linhagem Celular , Chlorocebus aethiops , Interações entre Hospedeiro e Microrganismos , Humanos , Camundongos , Células NIH 3T3 , Proteína Oncogênica v-akt/metabolismo , Phlebovirus/crescimento & desenvolvimento , Fosforilação , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Células Vero
8.
J Clin Invest ; 130(2): 799-812, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31904586

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging hemorrhagic fever caused by a tick-borne banyangvirus and is associated with high fatality. Despite increasing incidence of SFTS and serious public health concerns in East Asia, the pathogenesis of lethal SFTS virus (SFTSV) infection in humans is not fully understood. Numbers of postmortem examinations to determine target cells of the viral infection have so far been limited. Here we showed that B cells differentiating into plasmablasts and macrophages in secondary lymphoid organs were targets for SFTSV at the end stage of lethal infection, and the majority of SFTSV-infected cells were B cell-lineage lymphocytes. In affected individuals, B cell-lineage lymphocytes with SFTSV infection were widely distributed in both lymphoid and nonlymphoid organs, and infiltration of these cells into the capillaries of the organs could be observed occasionally. Moreover, a human plasmablastic lymphoma cell line, PBL-1, was susceptible to SFTSV propagation and had a similar immunophenotype to that of target cells of SFTSV in fatal SFTS. PBL-1 can therefore provide a potential in vitro model for human SFTSV infection. These results extend our understanding of the pathogenesis of human lethal SFTSV infection and can facilitate the development of SFTSV countermeasures.


Assuntos
Diferenciação Celular/imunologia , Febre por Flebótomos/imunologia , Phlebovirus/imunologia , Plasmócitos/imunologia , Linhagem Celular , Feminino , Humanos , Masculino , Febre por Flebótomos/mortalidade , Febre por Flebótomos/patologia , Plasmócitos/patologia
9.
ACS Omega ; 4(16): 16943-16955, 2019 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-31646241

RESUMO

An acyl-CoA:cholesterol O-acyltransferase-1 (ACAT-1/SOAT-1) inhibitor, K-604 is a promising drug candidate for the treatment of Alzheimer's disease and glioblastoma; however, it exhibits poor solubility in neutral water and low permeability across the blood-brain barrier. In this study, we report the successful delivery of K-604 to the brain via the intranasal route in mice using a hydroxycarboxylic acid solution. In cerebral tissue, the AUC of K-604 after intranasal administration (10 µL; 108 µg of K-604/mouse) was 772 ng·min/g, whereas that after oral administration (166 µg of K-604/mouse) was 8.9 ng·min/g. Thus, the index of brain-targeting efficiency was 133-fold based on the dose conversion. Even with intranasal administration of K-604 once per day for 7 days, the level of cholesteryl esters markedly decreased from 0.70 to 0.04 µmol/g in the mouse brain. Thus, this application will be a crucial therapeutic solution for ACAT-1 overexpressing diseases in the brain.

10.
Sci Rep ; 9(1): 11990, 2019 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-31427690

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging hemorrhagic fever caused by the SFTS phlebovirus (SFTSV). SFTS patients were first reported in China, followed by Japan and South Korea. In 2017, cats were diagnosed with SFTS for the first time, suggesting that these animals are susceptible to SFTSV. To confirm whether or not cats were indeed susceptible to SFTSV, animal subjects were experimentally infected with SFTSV. Four of the six cats infected with the SPL010 strain of SFTSV died, all showing similar or more severe symptoms than human SFTS patients, such as a fever, leukocytopenia, thrombocytopenia, weight loss, anorexia, jaundice and depression. High levels of SFTSV RNA loads were detected in the serum, eye swab, saliva, rectal swab and urine, indicating a risk of direct human infection from SFTS-infected animals. Histopathologically, acute necrotizing lymphadenitis and hemophagocytosis were prominent in the lymph nodes and spleen. Severe hemorrhaging was observed throughout the gastrointestinal tract. B cell lineage cells with MUM-1 and CD20, but not Pax-5 in the lesions were predominantly infected with SFTSV. The present study demonstrated that cats were highly susceptible to SFTSV. The risk of direct infection from SFTS-infected cats to humans should therefore be considered.


Assuntos
Doenças do Gato/virologia , Febres Hemorrágicas Virais/veterinária , Phlebovirus/fisiologia , Animais , Biomarcadores , Biópsia , Doenças do Gato/diagnóstico , Doenças do Gato/mortalidade , Doenças do Gato/transmissão , Gatos , Suscetibilidade a Doenças , Avaliação de Sintomas
11.
Virology ; 535: 102-110, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31299486

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) is an infectious disease with a high fatality rate, caused by SFTS virus (SFTSV). Because little is known about the nature of SFTSV, basic studies are required for the developments of vaccines and effective therapies. In the present study, we identified the amino acid residue important for membrane fusion induced by the SFTSV glycoprotein (GP). Syncytium formations were observed in cells expressing the GPs of SFTSV Japanese strain (YG-1 and SPL030). In contrast, no or only weak syncytium formations were induced in cells expressing GP of SFTSV Chinese strain (HB29). The replacement of arginine at amino acid residue 962 with serine in HB29 GP (R962S) induced membrane fusion, while the replacement of serine at residue 962 with arginine in YG1 GP (S962R) did not. These data indicate that serine at residue 962 in the SFTSV-GP is critical for inducing membrane fusion and viral infection.


Assuntos
Phlebovirus/fisiologia , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus , Substituição de Aminoácidos , Fusão Celular , Células Gigantes/citologia , Células Gigantes/virologia , Mutagênese Sítio-Dirigida , Phlebovirus/genética , Proteínas Virais de Fusão/genética
12.
Cell Mol Biol (Noisy-le-grand) ; 63(11): 119-123, 2017 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-29208183

RESUMO

Thrombopoietin (TPO) and its receptor, c-Mpl, are the central regulators of megakaryocyte development and platelet production and are also crucial to regulate megakaryocytopoiesis. TPO remarkably elevated c-mpl promoter activity, while the protein kinase C (PKC) inhibitors, GF109203, H7 and Calphostin C, clearly reduced the steady level of its promoter activity.  In the present study, motifs crucial for c-mpl promoter activity induced by TPO treatment have been analyzed using a human megakaryoblastic cell line, CMK. Destruction of the -107Sp1 and the -57Sp1 sites in the c-mpl promoter enhancer region resulted in decrease of the promoter activity by 53.1% and 64.4%, respectively, and destruction of -69Ets and -28Ets elements dramatically decreased the promoter activity by 96.4% and 87.8%, respectively, while mutation of -77GATA moderately reduced the activity by 31.4%. The result was in agreement with our previous report that showed the crucial motifs in the c-mpl promoter for the promoter activity induced by PMA-treatment. This indicates that TPO-induced activation of the c-mpl promoter activity is fully modulated by transcription through a PKC-dependent pathway and the two Sp1 and two Ets motifs are crucial for the activation of the c-mpl promoter activity rather than a GATA motif in the c-mpl promoter of CMK cells.


Assuntos
Regiões Promotoras Genéticas/genética , Trombopoetina/genética , Linhagem Celular Tumoral , Humanos , Megacariócitos/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética
13.
Arch Virol ; 162(8): 2421-2425, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28470418

RESUMO

Feline morbillivirus (FeMV), a member of the family Paramyxoviridae, is an emerging virus that was discovered in 2012. Despite the importance of FeMV infection in cats because of its postulated involvement in kidney diseases, no simple serological assay has been reported in its detection. Here, FeMV phosphoprotein (P protein) was expressed and purified as a glutathione-S-transferase (GST)-fusion protein and used for an enzyme-linked immunosorbent assay (ELISA) to detect FeMV-specific antibodies. With a cutoff value determined by immunoblotting, anti-FeMV P protein was detected with this assay in 22 (22%) of the 100 cat plasma samples collected from various regions of Japan. This ELISA is useful for epidemiological and immunological studies, as well as for diagnosis of FeMV infection.


Assuntos
Anticorpos Antivirais/sangue , Doenças do Gato/diagnóstico , Ensaio de Imunoadsorção Enzimática/veterinária , Infecções por Morbillivirus/diagnóstico , Morbillivirus/imunologia , Animais , Doenças do Gato/imunologia , Doenças do Gato/virologia , Gatos , Ensaio de Imunoadsorção Enzimática/métodos , Japão , Morbillivirus/isolamento & purificação , Infecções por Morbillivirus/sangue , Infecções por Morbillivirus/imunologia , Infecções por Morbillivirus/virologia , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia
14.
PLoS One ; 11(7): e0159740, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27448164

RESUMO

Pullulanase, an enzyme that catalyzes the hydrolysis of polysaccharides, has been identified in a broad range of organisms, including bacteria, yeasts, fungi, and animals. The pullulanase (pulB; FTT_0412c) of F. tularensis subspecies tularensis Schu S4 is considered to be a homologue of the type I pullulanase (pulA) of the other Francisella subspecies. The significance of Francisella pullulanase has been obscure until now. In the present study, we characterized a recombinant PulB of F. tularensis SCHU P9, which was expressed as a his-tagged protein in Escherichia coli. The recombinant PulB was confirmed to be a type I pullulanase by its enzymatic activity in vitro. A pulB gene knockout mutant of F. tularensis SCHU P9 (ΔpulB) was constructed using the TargeTron Knockout system and plasmid pKEK1140 to clarify the function of PulB during the growth of F. tularensis in macrophages. The intracellular growth of the ΔpulB mutant in murine macrophage J774.1 cells was significantly reduced compared with that of the parental strain SCHU P9. Expression of PulB in ΔpulB, using an expression plasmid, resulted in the complementation of the reduced growth in macrophages, suggesting that PulB is necessary for the efficient growth of F. tularensis in macrophages. To assess the role of PulB in virulence, the knockout and parent bacterial strains were used to infect C57BL/6J mice. Histopathological analyses showed that tissues from ΔpulB-infected mice showed milder lesions compared to those from SCHU P9-infected mice. However, all mice infected with SCHU P9 and ΔpulB showed the similar levels of bacterial loads in their tissues. The results suggest that PulB plays a significant role in bacterial growth within murine macrophage but does not contribute to bacterial virulence in vivo.


Assuntos
Francisella tularensis/enzimologia , Francisella tularensis/crescimento & desenvolvimento , Glicosídeo Hidrolases/metabolismo , Tularemia/microbiologia , Animais , Modelos Animais de Doenças , Ativação Enzimática , Feminino , Francisella tularensis/genética , Francisella tularensis/patogenicidade , Glicosídeo Hidrolases/genética , Concentração de Íons de Hidrogênio , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Mutação , Temperatura , Tularemia/imunologia , Tularemia/metabolismo , Tularemia/patologia , Virulência
15.
J Virol ; 90(11): 5292-5301, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-26984731

RESUMO

UNLABELLED: Severe fever with thrombocytopenia syndrome (SFTS) is an emerging hemorrhagic fever with a high case fatality rate caused by SFTS virus (SFTSV). Effective vaccines and specific therapies for SFTS are urgently sought, and investigation into virus-host cell interactions is expected to contribute to the development of antiviral strategies. In this study, we have developed a pseudotype vesicular stomatitis virus (VSV) bearing the unmodified Gn/Gc glycoproteins (GPs) of SFTSV (SFTSVpv). We have analyzed the host cell entry of this pseudotype virus and native SFTSV. Both SFTSVpv and SFTSV exhibited high infectivity in various mammalian cell lines. The use of lysosomotropic agents indicated that virus entry occurred via pH-dependent endocytosis. SFTSVpv and SFTSV infectivity was neutralized by serial dilutions of convalescent-phase patient sera. Entry of SFTSVpv and growth of SFTSV were increased in Raji cells expressing not only the C-type lectin dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin (DC-SIGN) but also DC-SIGN-related (DC-SIGNR) and liver and lymph node sinusoidal endothelial cell C-type lectin (LSECtin). 25-Hydroxycholesterol (25HC), a soluble oxysterol metabolite, inhibited the cell entry of SFTSVpv and the membrane fusion of SFTSV. These results indicate that pH-dependent endocytosis of SFTSVpv and SFTSV is enhanced by attachment to certain C-type lectins. SFTSVpv is an appropriate model for the investigation of SFTSV-GP-mediated cell entry and virus neutralization at lower biosafety levels. Furthermore, 25HC may represent a potential antiviral agent against SFTS. IMPORTANCE: SFTSV is a recently discovered bunyavirus associated with SFTS, a viral hemorrhagic fever with a high case fatality rate endemic to China, South Korea, and Japan. Because little is known about the characteristics of the envelope protein and entry mechanisms of SFTSV, further studies will be required for the development of a vaccine or effective therapies. In this study, we investigated the mechanism of SFTSV cell entry using SFTSVpv and the native virus. SFTSV can grow in nonsusceptible cell lines in the presence of certain C-type lectins. Moreover, 25HC, an oxysterol metabolite, may represent a potential therapeutic inhibitor of SFTSV infection.


Assuntos
Glicoproteínas/metabolismo , Phlebovirus/fisiologia , Trombocitopenia/virologia , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/fisiologia , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Animais , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , China , Endocitose , Glicoproteínas/química , Humanos , Concentração de Íons de Hidrogênio , Hidroxicolesteróis/farmacologia , Lectinas Tipo C/metabolismo , Testes de Neutralização , Febre por Flebótomos/virologia , Phlebovirus/química , Receptores de Superfície Celular/metabolismo , Vírus da Estomatite Vesicular Indiana/efeitos dos fármacos , Vírus da Estomatite Vesicular Indiana/crescimento & desenvolvimento
16.
Jpn J Infect Dis ; 69(6): 525-527, 2016 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-26743149

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) is a novel bunyavirus infection caused by the SFTS virus (SFTSV, family Bunyaviridae, genus Phlebovirus) with a high case fatality rate. A previously healthy 72-year-old man showed symptoms of fever, general fatigue, and altered consciousness. He was hospitalized for treatment. On day 3, considering the day on which fever appeared first during the disease course as day 0, he had bloody emesis. An emergency upper gastrointestinal endoscopic examination revealed multiple ulcerative lesions with continuously oozing hemorrhage in the stomach. He died on day 7. He was retrospectively diagnosed as having SFTS, Although it was less likely that the gastric ulcerative lesions were directly induced by SFTSV replication, it was evident that hemorrhagic emesis might occur in the patient in association with the pathophysiology of SFTS. The real-time imaging of gastric ulcerative lesions in a patient with SFTS is reported.


Assuntos
Infecções por Bunyaviridae/diagnóstico , Infecções por Bunyaviridae/patologia , Endoscopia do Sistema Digestório , Phlebovirus/isolamento & purificação , Idoso , Evolução Fatal , Humanos , Masculino , Úlcera Gástrica/etiologia , Úlcera Gástrica/patologia , Vômito/etiologia , Vômito/patologia
17.
Vet Microbiol ; 179(3-4): 197-203, 2015 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-26116524

RESUMO

A novel rhabdovirus was isolated from the serum of a healthy Japanese wild boar (Sus scrofa leucomystax) and identified using the rapid determination system for viral nucleic acid sequences (RDV), next-generation sequencing, and electron microscopy. The virus was tentatively named wild boar rhabdovirus 1 (WBRV1). Phylogenetic analysis of the entire genome sequence indicated that WBRV1 is closely related to Tupaia rhabdovirus (TRV), which was isolated from cultured cells of hepatocellular carcinoma tissue of tree shrew. TRV has not been assigned to any genus of Rhabdoviridae till date. Analysis of the L gene indicated that WBRV1 belongs to the genus Vesiculovirus. These observations suggest that both TRV and WBRV1 belong to a new genus of Rhabdoviridae. Next-generation genome sequencing of WBRV1 revealed 5 open reading frames of 1329, 765, 627, 1629, and 6336 bases in length. The WBRV1 gene sequences are similar to those of other rhabdoviruses. Epizootiological analysis of a population of wild boars in Wakayama prefecture in Japan indicated that 6.5% were positive for the WBRV1 gene and 52% were positive for WBRV1-neutralizing antibodies. Furthermore, such viral neutralizing antibodies were found in domestic pigs in another prefecture. WBRV1 was inoculated intranasally and intraperitoneally into SCID and BALB/c mice and viral RNA was detected in SCID mice, suggesting that WBRV1 can replicate in immunocompromised mice. These results indicate this novel virus is endemic in wild animals and livestock in Japan.


Assuntos
Genoma Viral/genética , Infecções por Rhabdoviridae/veterinária , Rhabdoviridae/classificação , Doenças dos Suínos/virologia , Animais , Sequência de Bases , DNA Viral/química , DNA Viral/genética , Sequenciamento de Nucleotídeos em Larga Escala/veterinária , Japão/epidemiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Dados de Sequência Molecular , Fases de Leitura Aberta/genética , Filogenia , Rhabdoviridae/genética , Rhabdoviridae/isolamento & purificação , Infecções por Rhabdoviridae/epidemiologia , Infecções por Rhabdoviridae/virologia , Análise de Sequência de DNA/veterinária , Sus scrofa , Suínos , Doenças dos Suínos/epidemiologia
18.
Jpn J Infect Dis ; 68(5): 387-93, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25766612

RESUMO

In this study, G proteins of the rabies virus (RABV) Kyoto strain were detected in the cytoplasm but not distributed at the cell membrane of mouse neuroblastoma (MNA) cells. G proteins of CVS-26 were detected in both the cell membrane and perinuclear space of MNA cells. We found that N-glycosylation of street RABV G protein by the insertion of the sequon Asn(204) induced the transfer of RABV G proteins to the cell surface membrane. Fixed RABV budding from the plasma membrane has been found to depend not only on G protein but also on other structural proteins such as M protein. However, the differing N-glycosylation of G protein could be associated with the distinct budding and antigenic features of RABV in street and fixed viruses. Our study of the association of N-glycan of G protein at Asn(204) with the transport of RABV G protein to the cell surface membrane contributes to the understanding of the evolution of fixed virus from street virus, which in turn would help for determine the mechanism underlying RABV budding and enhanced host immune responses.


Assuntos
Antígenos Virais/química , Antígenos Virais/metabolismo , Glicoproteínas/química , Glicoproteínas/metabolismo , Transporte Proteico/fisiologia , Raiva/virologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Animais , Asparagina/química , Asparagina/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Glicosilação , Humanos , Camundongos , Células Tumorais Cultivadas
19.
Sci Rep ; 5: 8850, 2015 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-25743183

RESUMO

We discovered a lethal hemorrhagic syndrome arising from severe thrombocytopenia in Japanese macaques kept at the Primate Research Institute, Kyoto University. Extensive investigation identified that simian retrovirus type 4 (SRV-4) was the causative agent of the disease. SRV-4 had previously been isolated only from cynomolgus macaques in which it is usually asymptomatic. We consider that the SRV-4 crossed the so-called species barrier between cynomolgus and Japanese macaques, leading to extremely severe acute symptoms in the latter. Infectious agents that cross the species barrier occasionally amplify in virulence, which is not observed in the original hosts. In such cases, the new hosts are usually distantly related to the original hosts. However, Japanese macaques are closely related to cynomolgus macaques, and can even hybridize when given the opportunity. This lethal outbreak of a novel pathogen in Japanese macaques highlights the need to modify our expectations about virulence with regards crossing species barriers.


Assuntos
Doenças Transmissíveis Emergentes/complicações , Doenças Transmissíveis Emergentes/virologia , Infecções por Retroviridae/complicações , Infecções por Retroviridae/virologia , Retrovirus dos Símios/classificação , Retrovirus dos Símios/genética , Trombocitopenia/etiologia , Animais , Doenças Transmissíveis Emergentes/diagnóstico , Doenças Transmissíveis Emergentes/transmissão , Feminino , Genoma Viral , Macaca , Metagenômica/métodos , Filogenia , RNA Viral , Infecções por Retroviridae/diagnóstico , Infecções por Retroviridae/transmissão , Retrovirus dos Símios/isolamento & purificação , Retrovirus dos Símios/ultraestrutura , Trombocitopenia/diagnóstico
20.
Okajimas Folia Anat Jpn ; 90(4): 101-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24815109

RESUMO

Thrombopoietin (TPO) and its receptor, c-Mpl, play the crucial role during megakaryocytopoiesis. Previously, we have shown that the promoter activity of c-mpl induced by TPO is modulated by transcription through a PKC-dependent pathway and that GATA(-77) is involved as a positive regulatory element in TPO-induced c-mpl gene expression in the megakaryoblastic CMK cells. In this research, to examine participating possibility of GATA promoter element in TPO- induced c-mpl gene expression through a PKC-independent pathway, the promoter activity of site-directed mutagenesis and the effect of potein kinase C modulator were measured by a transient transfection assay system. Together with our previous results on the TPO-induced c-mpl promoter, this study indicates destruction of -77GATA in c-mpl promoter decreased the activity by 47.3% under existence of GF109203. These results suggest that GATA promoter element plays significant role in TPO-induced c-mpl gene expression through a PKC-independent pathway.


Assuntos
Fatores de Transcrição GATA/metabolismo , Regulação da Expressão Gênica , Células Progenitoras de Megacariócitos/metabolismo , Receptores de Trombopoetina/metabolismo , Trombopoetina/metabolismo , Linhagem Celular Tumoral , Humanos , Regiões Promotoras Genéticas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA