Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Cell Dev Biol ; 12: 1363541, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38946797

RESUMO

Introduction: Duchenne muscular dystrophy (DMD) is a genetic disorder caused by mutations in the dystrophin-encoding gene that leads to muscle necrosis and degeneration with chronic inflammation during growth, resulting in progressive generalized weakness of the skeletal and cardiac muscles. We previously demonstrated the therapeutic effects of systemic administration of dental pulp mesenchymal stromal cells (DPSCs) in a DMD animal model. We showed preservation of long-term muscle function and slowing of disease progression. However, little is known regarding the effects of cell therapy on the metabolic abnormalities in DMD. Therefore, here, we aimed to investigate the mechanisms underlying the immunosuppressive effects of DPSCs and their influence on DMD metabolism. Methods: A comprehensive metabolomics-based approach was employed, and an ingenuity pathway analysis was performed to identify dystrophy-specific metabolomic impairments in the mdx mice to assess the therapeutic response to our established systemic DPSC-mediated cell therapy approach. Results and Discussion: We identified DMD-specific impairments in metabolites and their responses to systemic DPSC treatment. Our results demonstrate the feasibility of the metabolomics-based approach and provide insights into the therapeutic effects of DPSCs in DMD. Our findings could help to identify molecular marker targets for therapeutic intervention and predict long-term therapeutic efficacy.

2.
Sci Rep ; 14(1): 12559, 2024 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-38822013

RESUMO

Rabies virus (RABV) causes fatal neurological disease. Pre-exposure prophylaxis (PrEP) and post-exposure prophylaxis (PEP) using inactivated-virus vaccines are the most effective measures to prevent rabies. In Japan, HEP-Flury, the viral strain, used as a human rabies vaccine, has historically been propagated in primary fibroblast cells derived from chicken embryos. In the present study, to reduce the cost and labor of vaccine production, we sought to adapt the original HEP-Flury (HEP) to Vero cells. HEP was repeatedly passaged in Vero cells to generate ten- (HEP-10V) and thirty-passaged (HEP-30V) strains. Both HEP-10V and HEP-30V grew significantly better than HEP in Vero cells, with virulence and antigenicity similar to HEP. Comparison of the complete genomes with HEP revealed three non-synonymous mutations in HEP-10V and four additional non-synonymous mutations in HEP-30V. Comparison among 18 recombinant HEP strains constructed by reverse genetics and vesicular stomatitis viruses pseudotyped with RABV glycoproteins indicated that the substitution P(L115H) in the phosphoprotein and G(S15R) in the glycoprotein improved viral propagation in HEP-10V, while in HEP-30V, G(V164E), G(L183P), and G(A286V) in the glycoprotein enhanced entry into Vero cells. The obtained recombinant RABV strain, rHEP-PG4 strain, with these five substitutions, is a strong candidate for production of human rabies vaccine.


Assuntos
Substituição de Aminoácidos , Vacina Antirrábica , Vírus da Raiva , Animais , Células Vero , Chlorocebus aethiops , Vacina Antirrábica/genética , Vacina Antirrábica/imunologia , Vírus da Raiva/genética , Vírus da Raiva/imunologia , Humanos , Raiva/prevenção & controle , Raiva/virologia , Genoma Viral
3.
Viruses ; 16(5)2024 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-38793581

RESUMO

Rabies is a fatal encephalitic infectious disease caused by the rabies virus (RABV). RABV is highly neurotropic and replicates in neuronal cell lines in vitro. The RABV fixed strain, HEP-Flury, was produced via passaging in primary chicken embryonic fibroblast cells. HEP-Flury showed rapid adaptation when propagated in mouse neuroblastoma (MNA) cells. In this study, we compared the growth of our previously constructed recombinant HEP (rHEP) strain-based on the sequence of the HEP (HEP-Flury) strain-with that of the original HEP strain. The original HEP strain exhibited higher titer than rHEP and a single substitution at position 80 in the matrix (M) protein M(D80N) after incubation in MNA cells, which was absent in rHEP. In vivo, intracerebral inoculation of the rHEP-M(D80N) strain with this substitution resulted in enhanced viral growth in the mouse brain and a significant loss of body weight in the adult mice. The number of viral antigen-positive cells in the brains of adult mice inoculated with the rHEP-M(D80N) strain was significantly higher than that with the rHEP strain at 5 days post-inoculation. Our findings demonstrate that a single amino acid substitution in the M protein M(D80N) is associated with neurovirulence in mice owing to adaptation to mouse neuronal cells.


Assuntos
Substituição de Aminoácidos , Encéfalo , Vírus da Raiva , Raiva , Proteínas da Matriz Viral , Animais , Vírus da Raiva/genética , Vírus da Raiva/patogenicidade , Camundongos , Virulência , Encéfalo/virologia , Encéfalo/patologia , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo , Raiva/virologia , Neurônios/virologia , Neurônios/patologia , Replicação Viral , Linhagem Celular
4.
Gene Ther ; 30(7-8): 641-648, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36977769

RESUMO

Adeno-associated virus (AAV) vector-based gene therapy is potentially curative for various genetic diseases; however, the development of a scalable purification method for full-genome AAV vectors remains crucial to increase productivity and reduce cost of GMP production. In this study, we developed a large-scale short-term purification method for functional full-genome AAV particles by using 2-step cesium chloride (CsCl) density-gradient ultracentrifugation with a zonal rotor. The 2-step CsCl method with a zonal rotor improves separation between empty and full-genome AAV particles, reducing the ultracentrifugation time (4-5 h) and increasing the AAV volume for purification. The highly purified full-genome AAV particles were confirmed by analytical ultracentrifugation (AUC), droplet digital PCR (ddPCR) in the whole region of the AAV vector genome, transduction efficiency in target cells, and transmission electronic microscopy (TEM). The high-purity AAV9 particles were obtained using culture supernatant during vector preparation rather than cell lysate. CsCl could be simply removed by a hydroxyapatite column. Interestingly, ddPCR analysis revealed that "empty" AAV particles contain small fragments of the inverted terminal repeat (ITR), probably due to unexpected packaging of Rep-mediated ITR fragments. This large-scale functional AAV vector purification with ultracentrifugation would be effective for gene therapy.


Assuntos
Dependovirus , Vetores Genéticos , Ultracentrifugação , Dependovirus/genética
5.
Viruses ; 14(10)2022 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-36298707

RESUMO

The rabies virus is widely distributed and vaccines are an important strategy to prevent its spread. The whole-genome sequences of rabies strains in relation to vaccine development provide essential information to maintain vaccine quality and develop new vaccines. However, the genetic characteristics of the purified chick embryo cell culture rabies vaccine, KM Biologics (PCECV-KMB), developed in Japan in the 1970s, have not been explored. In this study, we conducted a genome-wide analysis of the open reading frame regions of rabies strains discovered from the 1940s-1980s and used to develop chick embryo cell-adapted HEP-Flury small plaque-forming (CEF-S) strain, which is a vaccine strain of PCECV-KMB. The genetic characteristic of CEF-S, developed by acclimation of the HEP-Flury-NIID strain to one-day eggs and subsequently to chick embryo cells, were confirmed by comparing the genome identity and revealing the nine amino acid mutations between CEF-S and HEP-Flury-NIID. The efficacy of PCECV-KMB was evaluated using attack strains isolated in Thailand in the 1960s-1970s during vaccine development. Phylogenetic analyses of the attack strains classified them in the same Asian clade as the 2000s imported cases from the Philippines to Japan, suggesting that PCECV-KMB is adequate for preventing the spread of the current rabies virus.


Assuntos
Produtos Biológicos , Vacina Antirrábica , Vírus da Raiva , Raiva , Animais , Humanos , Embrião de Galinha , Vírus da Raiva/genética , Raiva/prevenção & controle , Filogenia , Japão , Desenvolvimento de Vacinas , Anticorpos Antivirais , Aminoácidos
6.
Dis Model Mech ; 14(12)2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34850861

RESUMO

Musculocontractural Ehlers-Danlos syndrome (mcEDS) is caused by generalized depletion of dermatan sulfate (DS) due to biallelic pathogenic variants in CHST14 encoding dermatan 4-O-sulfotransferase 1 (D4ST1) (mcEDS-CHST14). Here, we generated mouse models for mcEDS-CHST14 carrying homozygous mutations (1 bp deletion or 6 bp insertion/10 bp deletion) in Chst14 through CRISPR/Cas9 genome engineering to overcome perinatal lethality in conventional Chst14-deleted knockout mice. DS depletion was detected in the skeletal muscle of these genome-edited mutant mice, consistent with loss of D4ST1 activity. The mutant mice showed common pathophysiological features, regardless of the variant, including growth impairment and skin fragility. Notably, we identified myopathy-related phenotypes. Muscle histopathology showed variation in fiber size and spread of the muscle interstitium. Decorin localized diffusely in the spread endomysium and perimysium of skeletal muscle, unlike in wild-type mice. The mutant mice showed lower grip strength and decreased exercise capacity compared to wild type, and morphometric evaluation demonstrated thoracic kyphosis in mutant mice. The established CRISPR/Cas9-engineered Chst14 mutant mice could be a useful model to further our understanding of mcEDS pathophysiology and aid in the development of novel treatment strategies.


Assuntos
Síndrome de Ehlers-Danlos , Animais , Sistemas CRISPR-Cas/genética , Síndrome de Ehlers-Danlos/genética , Síndrome de Ehlers-Danlos/patologia , Feminino , Genômica , Camundongos , Camundongos Knockout , Gravidez , Sulfotransferases/genética , Sulfotransferases/metabolismo
7.
Front Cell Dev Biol ; 9: 695021, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34708033

RESUMO

Carbohydrate sulfotransferase 14 (CHST14) encodes dermatan 4-O-sulfotransferase 1, a critical enzyme for dermatan sulfate (DS) biosynthesis. Musculocontractural Ehlers-Danlos syndrome (mcEDS) is associated with biallelic pathogenic variants of CHST14 and is characterized by malformations and manifestations related to progressive connective tissue fragility. We identified myopathy phenotypes in Chst14-deficient mice using an mcEDS model. Decorin is a proteoglycan harboring a single glycosaminoglycan chain containing mainly DS, which are replaced with chondroitin sulfate (CS) in mcEDS patients with CHST14 deficiency. We studied the function of decorin in the skeletal muscle of Chst14-deficient mice because decorin is important for collagen-fibril assembly and has a myokine role in promoting muscle growth. Although decorin was present in the muscle perimysium of wild-type (Chst14+/+ ) mice, decorin was distributed in the muscle perimysium as well as in the endomysium of Chst14-/- mice. Chst14-/- mice had small muscle fibers within the spread interstitium; however, histopathological findings indicated milder myopathy in Chst14-/- mice. Myostatin, a negative regulator of protein synthesis in the muscle, was upregulated in Chst14-/- mice. In the muscle of Chst14-/- mice, decorin was downregulated compared to that in Chst14+/+ mice. Chst14-/- mice showed altered cytokine/chemokine balance and increased fibrosis, suggesting low myogenic activity in DS-deficient muscle. Therefore, DS deficiency in mcEDS causes pathological localization and functional abnormalities of decorin, which causes disturbances in skeletal muscle myogenesis.

8.
PLoS One ; 14(10): e0223684, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31589656

RESUMO

Middle East respiratory syndrome-coronavirus (MERS-CoV) is an emerging virus that causes severe disease with fatal outcomes; however, there are currently no approved vaccines or specific treatments against MERS-CoV. Here, we developed a novel bivalent vaccine against MERS-CoV and rabies virus (RV) using the replication-incompetent P-gene-deficient RV (RVΔP), which has been previously established as a promising and safe viral vector. MERS-CoV spike glycoprotein comprises S1 and S2 subunits, with the S1 subunit being a primary target of neutralizing antibodies. Recombinant RVΔP, which expresses S1 fused with transmembrane and cytoplasmic domains together with 14 amino acids from the ectodomains of the RV-glycoprotein (RV-G), was developed using a reverse genetics method and named RVΔP-MERS/S1. Following generation of RVΔP-MERS/S1 and RVΔP, our analysis revealed that they shared similar growth properties, with the expression of S1 in RVΔP-MERS/S1-infected cells confirmed by immunofluorescence and western blot, and the immunogenicity and pathogenicity evaluated using mouse infection experiments. We observed no rabies-associated signs or symptoms in mice inoculated with RVΔP-MERS/S1. Moreover, virus-specific neutralizing antibodies against both MERS-CoV and RV were induced in mice inoculated intraperitoneally with RVΔP-MERS/S1. These findings indicate that RVΔP-MERS/S1 is a promising and safe bivalent-vaccine candidate against both MERS-CoV and RV.


Assuntos
Infecções por Coronavirus/prevenção & controle , Imunogenicidade da Vacina , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Vírus da Raiva/genética , Vacinas Sintéticas/imunologia , Vacinas Virais/imunologia , Replicação Viral , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Vírus da Raiva/fisiologia , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinas Sintéticas/genética , Células Vero , Vacinas Virais/genética
9.
PLoS Negl Trop Dis ; 12(4): e0006398, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29659579

RESUMO

BACKGROUND: Lymphocytic choriomeningitis virus (LCMV) causes a variety of diseases, including asymptomatic infections, meningitis, and congenital infections in the fetus of infected mother. The development of a safe and effective vaccine against LCMV is imperative. This study aims to develop a new candidate vaccine against LCMV using a recombinant replication-incompetent rabies virus (RV) vector. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we have generated a recombinant deficient RV expressing the LCMV glycoprotein precursor (GPC) (RVΔP-LCMV/GPC) which is lacking the RV-P gene. RVΔP-LCMV/GPC is able to propagate only in cells expressing the RV-P protein. In contrast, the LCMV-GPC can be expressed in general cells, which do not express RV-P protein. The ability of RVΔP-LCMV/GPC to protect mice from LCMV infection and induce cellular immunity was assessed. Mice inoculated intraperitoneally with RVΔP-LCMV/GPC showed higher survival rates (88.2%) than those inoculated with the parental recombinant RV-P gene-deficient RV (RVΔP) (7.7%) following a LCMV challenge. Neutralizing antibody (NAb) against LCMV was not induced, even in the sera of surviving mice. CD8+ T-cell depletion significantly reduced the survival rates of RVΔP-LCMV/GPC-inoculated mice after the LCMV challenge. These results suggest that CD8+ T cells play a major role in the observed protection against LCMV. In contrast, NAbs against RV were strongly induced in sera of mice inoculated with either RVΔP-LCMV/GPC or RVΔP. In safety tests, suckling mice inoculated intracerebrally with RVΔP-LCMV/GPC showed no symptoms. CONCLUSIONS/SIGNIFICANCE: These results show RVΔP-LCMV/GPC might be a promising candidate vaccine with dual efficacy, protecting against both RV and LCMV.


Assuntos
Glicoproteínas/imunologia , Coriomeningite Linfocítica/prevenção & controle , Vírus da Coriomeningite Linfocítica/imunologia , Vírus da Raiva/fisiologia , Proteínas Virais/imunologia , Vacinas Virais/imunologia , Animais , Feminino , Expressão Gênica , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Glicoproteínas/administração & dosagem , Glicoproteínas/genética , Humanos , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/genética , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Raiva/genética , Proteínas Virais/administração & dosagem , Proteínas Virais/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Replicação Viral
10.
Virus Res ; 249: 57-65, 2018 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-29548745

RESUMO

The genus Thogotovirus, as represented by Thogoto virus and Dhori virus, comprises a group of arthropod-borne viruses, most members of which are transmitted by ticks. Here we report the genetic and biological characterization of a new thogotovirus, designated Oz virus (OZV), isolated from the hard tick Amblyomma testudinarium in Ehime, Japan. OZV efficiently replicated and induced a cytopathic effect in Vero cells, from which enveloped pleomorphic virus particles were formed by budding. OZV could also replicate in BHK-21 and DH82 cells and caused high mortality in suckling mice after intracerebral inoculation. Phylogenetic analyses of six viral proteins indicated that OZV is clustered with Dhori and related viruses, and is most closely related in glycoprotein (GP) and matrix protein (M) sequences to Bourbon virus, a human-pathogenic thogotovirus discovered recently in the United States. Our findings emphasize the need for understanding the geographic distribution and ecology of OZV and related viruses and for reevaluation of the medical and public health importance of thogotoviruses.


Assuntos
Ixodidae/virologia , Filogenia , Thogotovirus/classificação , Thogotovirus/isolamento & purificação , Animais , Linhagem Celular , Análise por Conglomerados , Efeito Citopatogênico Viral , Modelos Animais de Doenças , Japão , Camundongos , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Análise de Sequência de DNA , Homologia de Sequência , Thogotovirus/genética , Thogotovirus/fisiologia , Proteínas Virais/genética , Cultura de Vírus , Liberação de Vírus , Replicação Viral
11.
Virus Res ; 244: 252-261, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29197549

RESUMO

In Japan, indigenous tick-borne phleboviruses (TBPVs) and their associated diseases first became evident in 2013 by reported human cases of severe fever with thrombocytopenia syndrome (SFTS). In this study, we report a novel member of the genus Phlebovirus designated as Kabuto Mountain virus (KAMV), which was isolated from the ixodid tick Haemaphysalis flava in Hyogo, Japan. A complete viral genome sequencing and phylogenetic analyses showed that KAMV is a novel member of TBPVs, which is closely related to the Uukuniemi and Kaisodi group viruses. However, unlike the Uukuniemi group viruses, the 165-nt intergenic region (IGR) in the KAMV S segment was highly C-rich in the genomic sense and not predicted to form a secondary structure, which are rather similar to those of the Kaisodi group viruses and most mosquito/sandfly-borne phleboviruses. Furthermore, the NSs protein of KAMV was highly divergent from those of other TBPVs. These results provided further insights into the genetic diversity and evolutionary relationships of TBPVs. KAMV could infect and replicate in some rodent and primate cell lines. We evaluated the infectivity and pathogenicity of KAMV in suckling mice, where we obtained a virulent strain after two passages via intracerebral inoculation. This is the first report showing the existence of a previously unrecognized TBPV in Japan, other than the SFTS virus.


Assuntos
Infecções por Bunyaviridae/virologia , DNA Viral/genética , Genoma Viral , Phlebovirus/genética , Filogenia , Animais , Animais Lactentes , Vetores Aracnídeos/virologia , Infecções por Bunyaviridae/mortalidade , Infecções por Bunyaviridae/patologia , Linhagem Celular , Chlorocebus aethiops , DNA Intergênico/genética , Modelos Animais de Doenças , Variação Genética , Humanos , Japão , Mesocricetus , Camundongos , Phlebovirus/classificação , Phlebovirus/isolamento & purificação , Phlebovirus/patogenicidade , Análise de Sequência de DNA , Análise de Sobrevida , Carrapatos/virologia , Células Vero , Virulência , Sequenciamento Completo do Genoma
12.
Virus Res ; 242: 131-140, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28964878

RESUMO

During the course of tick-borne virus surveillance in Japan, three independent isolates of probably the same virus were obtained from three geographically distant populations of the hard tick Haemaphysalis flava. Genome analyses of the three isolates demonstrated that they were closely related but distinct strains of a novel virus, designated Tarumizu tick virus (TarTV), which has a genome of 12 double-stranded RNA segments. The development of the virus-induced cytopathic effects on BHK cells significantly varied according to virus strains. Ten out of 12 segments of TarTV appeared to encode putative orthologs or functional equivalents of viral proteins of Colorado tick fever virus (CTFV) and Eyach virus, suggesting that TarTV is the third member of the genus Coltivirus in the family Reoviridae. This was supported by the facts that the 5'- and 3'-terminal consensus sequences of coltivirus genomes were found also in TarTV genome, and segment 9 of TarTV had sequence and structural features that may mediate a stop codon read-through as observed in that of CTFV. However, segment 7 and 10 of TarTV had no significant sequence similarities to any other proteins of known coltiviruses. Electron microscopic analysis demonstrated that TarTV particle had a non-enveloped bilayer icosahedral structure, and viral inclusion bodies were formed in infected cells. TarTV could infect and replicate in several mammalian cell lines tested, but show no clinical symptoms in intracerebrally inoculated mice. Taken together, our findings provide new insights into genetic diversity and evolution of the genus Coltivirus.


Assuntos
Coltivirus/classificação , Coltivirus/isolamento & purificação , Ixodidae/virologia , Animais , Capsídeo/ultraestrutura , Células Cultivadas , Coltivirus/genética , Cricetinae , Genoma Viral , Corpos de Inclusão Viral/ultraestrutura , Japão , Camundongos , Microscopia Eletrônica de Transmissão , Filogenia , Infecções por Reoviridae/patologia , Infecções por Reoviridae/virologia , Análise de Sequência de DNA , Homologia de Sequência , Vírion/ultraestrutura
13.
Biologicals ; 46: 38-45, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28040390

RESUMO

Potency controls of inactivated rabies vaccines for human use are confirmed by the National Institutes of Health challenge test in which lethal infection with severe neurological symptoms should be observed in approximately half of the mice inoculated with the rabies virus. Weight loss, decreased body temperature, and the presence of rabies-associated neurological signs have been proposed as humane endpoints. The potential for reduction of animal suffering by introducing humane endpoints in the potency test for inactivated rabies vaccine for human use was investigated. The clinical signs were scored and body weight was monitored. The average times to death following inoculation were 10.49 and 10.99 days post-inoculation (dpi) by the potency and challenge control tests, respectively, whereas the average times to showing Score-2 signs (paralysis, trembling, and coma) were 6.26 and 6.55 dpi, respectively. Body weight loss of more than 15% appeared at 5.82 and 6.42 dpi. The data provided here support the introduction of obvious neuronal signs combined with a body weight loss of ≥15% as a humane endpoint to reduce the time of animal suffering by approximately 4 days.


Assuntos
Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Raiva/imunologia , Vacinação/métodos , Potência de Vacina , Animais , Peso Corporal/imunologia , Embrião de Galinha , Feminino , Humanos , Camundongos , Raiva/mortalidade , Raiva/virologia , Análise de Sobrevida , Taxa de Sobrevida , Fatores de Tempo , Vacinas de Produtos Inativados/imunologia , Redução de Peso/imunologia
14.
Trop Med Health ; 44: 6, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27433125

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging disease in East Asia. It is thought that the SFTS virus (SFTSV) circulates between ticks and animals in nature and that the virus is transmitted to humans by tick bites. SFTS is endemic to Nagasaki in western Japan; however, epidemiological information regarding SFTSV in Nagasaki is not known. In this study, we performed SFTSV IgG ELISAs and neutralization antibody assays for a seroepidemiological survey using samples from wild boars captured in six areas of Nagasaki. SFTSV seropositive animals were found in three areas. Our findings provide epidemiological information on the distribution of SFTSV in Nagasaki.

15.
Clin Vaccine Immunol ; 23(8): 689-97, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27307452

RESUMO

Yellow fever (YF) is an acute hemorrhagic viral infection transmitted by mosquitoes in Africa and South America. The major challenge in YF disease detection and confirmation of outbreaks in Africa is the limited availability of reference laboratories and the persistent lack of access to diagnostic tests. We used wild-type YF virus sequences to generate recombinant envelope protein in an Escherichia coli expression system. Both the recombinant protein and sucrose gradient-purified YF vaccine virus 17D (YF-17D) were used to immunize BALB/c mice to generate monoclonal antibodies (MAbs). Eight MAbs were established and systematically characterized by indirect enzyme-linked immunosorbent assay (ELISA), Western blot analysis, and immunofluorescence assay (IFA). The established MAbs showed strong reactivity with wild-type YF virus and recombinant protein with no detectable cross-reactivity to dengue virus or Japanese encephalitis virus. Epitope mapping showed strong binding of three MAbs to amino acid positions 1 to 51, while two MAbs mapped to amino acid positions 52 to 135 of the envelope protein. The remaining three MAbs did not show reactivity to envelope fragments. The established MAbs exert no neutralization against wild-type YF and 17D viruses (titer of <10 for both strains). The applicability of MAbs 8H3 and 3F4 was further evaluated using IgM capture ELISA. A total of 49 serum samples were analyzed, among which 12 positive patient and vaccinee samples were correctly identified. Using serum samples that were 2-fold serially diluted, the IgM capture ELISA was able to detect all YF-positive samples. Furthermore, MAb-based antigen detection ELISA enabled the detection of virus in culture supernatants containing titers of about 1,000 focus-forming units.


Assuntos
Anticorpos Monoclonais/isolamento & purificação , Anticorpos Antivirais/isolamento & purificação , Antígenos Virais/análise , Testes Diagnósticos de Rotina/métodos , Ensaio de Imunoadsorção Enzimática/métodos , Febre Amarela/diagnóstico , Vírus da Febre Amarela/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Imunoglobulina M/sangue , Fatores Imunológicos , Camundongos Endogâmicos BALB C , Ligação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
16.
Sci Rep ; 6: 20213, 2016 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-26863911

RESUMO

Ixodid ticks transmit several important viral pathogens. We isolated a new virus (Tofla virus: TFLV) from Heamaphysalis flava and Heamaphysalis formsensis in Japan. The full-genome sequences revealed that TFLV belonged to the genus Nairovirus, family Bunyaviridae. Phylogenetic analyses and neutralization tests suggested that TFLV is closely related to the Hazara virus and that it is classified into the Crimean-Congo hemorrhagic fever group. TFLV caused lethal infection in IFNAR KO mice. The TFLV-infected mice exhibited a gastrointestinal disorder, and positron emission tomography-computed tomography images showed a significant uptake of (18)F-fluorodeoxyglucose in the intestinal tract. TFLV was able to infect and propagate in cultured cells of African green monkey-derived Vero E6 cells and human-derived SK-N-SH, T98-G and HEK-293 cells. Although TFLV infections in humans and animals are currently unknown, our findings may provide clues to understand the potential infectivity and to develop of pre-emptive countermeasures against this new tick-borne Nairovirus.


Assuntos
Arbovírus/genética , Infecções por Bunyaviridae/virologia , Genoma Viral , Nairovirus/genética , Filogenia , Carrapatos/virologia , Animais , Arbovírus/classificação , Arbovírus/patogenicidade , Infecções por Bunyaviridae/mortalidade , Infecções por Bunyaviridae/patologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Monitoramento Epidemiológico , Fluordesoxiglucose F18/metabolismo , Trato Gastrointestinal/patologia , Trato Gastrointestinal/virologia , Células HEK293 , Humanos , Japão , Camundongos , Camundongos Knockout , Nairovirus/classificação , Nairovirus/patogenicidade , Neuroglia/patologia , Neuroglia/virologia , Neurônios/patologia , Neurônios/virologia , Testes de Neutralização , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Análise de Sequência de RNA , Análise de Sobrevida , Células Vero
17.
Virology ; 482: 19-27, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25817401

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging viral disease that is endemic in China, Korea and Japan. No effective vaccine or specific treatment for SFTS is currently available. Here, we used a mouse model to examine the effects of ribavirin, site-1 protease inhibitor PF-429242, steroids, and combination of minocycline and ciprofloxacin (MC) on SFTS infection. The antiserum from a patient who recovered from SFTS was also examined for its effect on mice. Administration of antiserum completely protected mice against lethal infection with SFTSV. It could also protect mice from showing clinical signs of the disease due to non-lethal infection. MC-treatment resulted in prolonged survival times during lethal infection. Although other agents had no significant protective effects, they did not provide detrimental effects that could lead to progression of the disease in mice. Our results suggest that antiserum treatment may be clinically useful for post-exposure prophylaxis against SFTSV infection.


Assuntos
Anticorpos Antivirais/uso terapêutico , Infecções por Bunyaviridae/terapia , Imunoglobulinas Intravenosas/uso terapêutico , Phlebovirus/imunologia , Profilaxia Pós-Exposição/métodos , Animais , Antivirais/uso terapêutico , Modelos Animais de Doenças , Humanos , Camundongos , Análise de Sobrevida , Resultado do Tratamento
18.
J Gen Virol ; 95(Pt 6): 1390-1395, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24646751

RESUMO

In 2005, we isolated a new species of virus from mosquitoes in the Philippines. The virion was elliptical in shape and had a short single projection. The virus was named Tanay virus (TANAV) after the locality in which it was found. TANAV genomic RNA was a 9562 nt+poly-A positive strand, and polycistronic. The longest ORF contained putative RNA-dependent RNA polymerase (RdRP); however, conserved short motifs in the RdRP were permuted. TANAV was phylogenetically close to Negevirus, a recently proposed taxon of viruses isolated from haemophagic insects, and to some plant viruses, such as citrus leprosis virus C, hibiscus green spot virus and blueberry necrotic ring blotch virus. In this paper, we describe TANAV and the permuted structure of its RdRP, and discuss its phylogeny together with those of plant viruses and negevirus.


Assuntos
Culicidae/virologia , Vírus de Insetos/isolamento & purificação , Vírus de RNA/isolamento & purificação , Vírus não Classificados/isolamento & purificação , Sequência de Aminoácidos , Animais , Culex/virologia , Genoma Viral , Vírus de Insetos/classificação , Vírus de Insetos/genética , Microscopia Eletrônica de Transmissão , Dados de Sequência Molecular , Fases de Leitura Aberta , Filipinas , Filogenia , Vírus de Plantas/genética , Vírus de RNA/classificação , Vírus de RNA/genética , RNA Polimerase Dependente de RNA/genética , Homologia de Sequência de Aminoácidos , Proteínas Virais/genética , Vírion/ultraestrutura , Vírus não Classificados/classificação , Vírus não Classificados/genética
19.
Vaccine ; 28(50): 7939-46, 2010 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-20933564

RESUMO

A formalin-inactivated West Nile Virus (WNV) vaccine (WN-VAX) derived from the WNV-NY99 strain was tested for its safety, efficacy, dilution limit for complete protection, and cross-neutralization. Safety tests performed with experimental animals, bacteria, or cultured cell lines showed no evidence of short- or long-term adverse effects. WN-VAX also protected 100% of 4-week-old mice against a lethal challenge from the WNV-NY99 strain after two doses of intraperitoneal inoculation-even when the vaccine was diluted to 3.2ng/dose. Moreover, very limited cross-neutralization activity against Japanese encephalitis virus, Dengue virus, Murray Valley encephalitis virus, Yellow fever virus or St. Louis encephalitis virus was observed. Therefore, the WN-VAX satisfies the requirements for human trials planned to be done in Japan.


Assuntos
Proteção Cruzada , Febre do Nilo Ocidental/prevenção & controle , Vacinas contra o Vírus do Nilo Ocidental/imunologia , Animais , Linhagem Celular , Chlorocebus aethiops , Cães , Feminino , Formaldeído/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Testes de Mutagenicidade , Testes de Neutralização , Coelhos , Ratos , Testes de Toxicidade , Vacinas de Produtos Inativados/imunologia , Células Vero , Vacinas contra o Vírus do Nilo Ocidental/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...