Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 156
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 202(3): 841-856, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30598516

RESUMO

Induction of type I IFNs during viral infection is crucial for host defense. IRF 3 and IRF7 play a critical role as key transcription factors in the activation of the IFN induction. Viruses have evolved a variety of strategies to evade innate immunity. Our previous studies have shown that the nonstructural protein (NSs) of the severe fever with thrombocytopenia syndrome virus (SFTSV) can suppress the IFN-ß induction through its interaction with tank-binding kinase-1 and sequestering the inhibitor of nuclear factor kappa B kinase(IKK) complex into the inclusion bodies formed by NSs. In this study, we characterized the unique function of IRF7 in innate immunity and its role in inducing IFN-α in particular, regulated by NSs during the SFTSV infection in several cell types of human origin. Whereas IRF3 is constitutively expressed, IRF7 was significantly induced differentially in various cell types in response to SFTSV infection, promoted the induction of IFN-α2 and -α4, and further induced IFN-ß, thus contributing to suppressing the viral replication. Our data indicate that NSs directly interacted with and sequestered IRF7 into the inclusion bodies, which is different from IRF3 indirectly interacting with NSs. Although interaction of NSs with IRF7 did not inhibit IRF7 phosphorylation, p-IRF7 was trapped in the inclusion bodies, resulting in a significant reduction of the IFN-α2 and -α4 induction and therefore enhanced viral replication. Interaction of the viral NSs with both IRF7 and IRF3 and subsequent sequestration of these transcription factors into viral inclusion bodies, a unique strategy used by this phlebovirus, may ensure effective evasion and suppression of host innate immunity.


Assuntos
Corpos de Inclusão Viral/imunologia , Fator Regulador 7 de Interferon/imunologia , Interferon-alfa/imunologia , Interferon beta/imunologia , Phlebovirus/imunologia , Proteínas não Estruturais Virais/imunologia , Células HEK293 , Células HeLa , Células Hep G2 , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Fator Regulador 3 de Interferon/genética , Fator Regulador 7 de Interferon/genética , Transdução de Sinais , Células THP-1 , Replicação Viral
2.
J Biol Chem ; 294(12): 4290-4303, 2019 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-30737282

RESUMO

Most neutralizing antibodies against highly pathogenic avian influenza A virus H5N1 recognize the receptor-binding site (RBS) on the globular head domain and the stem of H5N1 hemagglutinin (HA). Through comprehensive analysis of multiple human protective antibodies, we previously identified four vulnerable sites (VS1-VS4) on the globular head domain. Among them, the VS1, occupying the opposite side of the RBS on the same HA, was defined by the epitope of antibody 65C6. In this study, we report the crystal structures of two additional human H5N1 antibodies isolated from H5N1-infected individuals, 3C11 and AVFluIgG01, bound to the head at 2.33- and 2.30-Å resolution, respectively. These two new antibody epitopes have large overlap with and extend beyond the original VS1. Site-directed mutagenesis experiments identified eight pivotal residues (Ser-126b, Lys-165, Arg-166, Ser-167, Tyr-168, Asn-169, Thr-171, and Asn-172) critical for 65C6-, 3C11-, and AVFluIgG01-binding and neutralization activities. These residues formed a unique "Y"-shaped surface on H5N1 globular head and are highly conserved among H5N1 viruses. Our results further support the existence of a vulnerable site distinct from the RBS and the stem region of H5N1 HA, and future design of immunogens should take this particular site into consideration.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Virus da Influenza A Subtipo H5N1/metabolismo , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Sítios de Ligação , Cristalografia por Raios X , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Virus da Influenza A Subtipo H5N1/imunologia , Virus da Influenza A Subtipo H5N1/patogenicidade , Mutagênese Sítio-Dirigida , Conformação Proteica , Reprodutibilidade dos Testes
3.
Proc Natl Acad Sci U S A ; 114(36): E7564-E7573, 2017 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-28827346

RESUMO

Severe fever with thrombocytopenia syndrome virus (SFTSV) and Rift Valley fever virus (RVFV) are two arthropod-borne phleboviruses in the Bunyaviridae family, which cause severe illness in humans and animals. Glycoprotein N (Gn) is one of the envelope proteins on the virus surface and is a major antigenic component. Despite its importance for virus entry and fusion, the molecular features of the phleboviruse Gn were unknown. Here, we present the crystal structures of the Gn head domain from both SFTSV and RVFV, which display a similar compact triangular shape overall, while the three subdomains (domains I, II, and III) making up the Gn head display different arrangements. Ten cysteines in the Gn stem region are conserved among phleboviruses, four of which are responsible for Gn dimerization, as revealed in this study, and they are highly conserved for all members in Bunyaviridae Therefore, we propose an anchoring mode on the viral surface. The complex structure of the SFTSV Gn head and human neutralizing antibody MAb 4-5 reveals that helices α6 in subdomain III is the key component for neutralization. Importantly, the structure indicates that domain III is an ideal region recognized by specific neutralizing antibodies, while domain II is probably recognized by broadly neutralizing antibodies. Collectively, Gn is a desirable vaccine target, and our data provide a molecular basis for the rational design of vaccines against the diseases caused by phleboviruses and a model for bunyavirus Gn embedding on the viral surface.


Assuntos
Anticorpos Neutralizantes/metabolismo , Epitopos/metabolismo , Glicoproteínas/química , Glicoproteínas/metabolismo , Phlebovirus/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Animais , Infecções por Bunyaviridae/virologia , Linhagem Celular , Cristalografia por Raios X , Epitopos/química , Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/metabolismo , Células Sf9 , Internalização do Vírus
4.
J Biol Chem ; 293(42): 16503-16517, 2018 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-30154240

RESUMO

The highly pathogenic avian influenza virus H5N1 is a major threat to global public health and therefore a high-priority target of current vaccine development. The receptor-binding site (RBS) on the globular head of hemagglutinin (HA) in the viral envelope is one of the major target sites for antibody recognition against H5N1 and other influenza viruses. Here, we report the identification and characterization of a pair of human RBS-specific antibodies, designated FLD21.140 and AVFluIgG03, that are mutually complementary in their neutralizing activities against a diverse panel of H5N1 viruses. Crystallographic analysis and site-directed mutagenesis revealed that the two antibodies share a similar RBS-binding mode, and their individual specificities are governed by residues at positions 133a, 144, and 145. Specifically, FLD21.140 preferred Leu-133a/Lys-144/Ser-145, whereas AVFluIgG03 favored Ser-133a/Thr-144/Pro-145 residue triplets, both of which perfectly matched the most prevalent residues in viruses from epidemic-originating regions. Of note, according to an analysis of 3758 H5 HA sequences available in the Influenza Virus Database at the National Center for Biotechnology Information, the residues Leu-133a/Ser-133a and Ser-145/Pro-145 constituted more than 87.6 and 99.3% of all residues at these two positions, respectively. Taken together, our results provide a structural understanding for the neutralizing complementarity of these two antibodies and improve our understanding of the RBS-specific antibody response against H5N1 infection in humans.


Assuntos
Anticorpos Neutralizantes/metabolismo , Virus da Influenza A Subtipo H5N1/metabolismo , Receptores Virais/imunologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Cristalografia por Raios X , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Humanos , Especificidade da Espécie
5.
BMC Infect Dis ; 19(1): 476, 2019 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-31138131

RESUMO

BACKGROUND: Severe fever with thrombocytopenia syndrome (SFTS) is a newly identified severe infectious disease caused by SFTS phlebovirus (SFTSV). SFTS monitoring has been carried out since 2010 in mainland China. We analysed the detection results of SFTSV RNA and antibody in SFTS surveillance cases to provide basic data for SFTS diagnosis. METHODS: This study was conducted in Shandong Province. Sera of SFTS surveillance cases were collected to detect SFTSV RNA and antibody by real-time RT-PCR and enzyme-linked immunosorbent assay, respectively. Detection rates were calculated. SPSS 18.0 (Chicago, IL, USA) was used for statistical analysis to compare the detection rates of SFTSV RNA and antibodies among different sera groups. RESULTS: A total of 374 SFTS surveillance cases were enrolled. Overall, 93.3% (349/374) of the sera samples were collected within 2 weeks after onset, and 6.7% (25/374) were collected between 15 days and 45 days. Of these, 183 (48.9%) were positive for SFTSV RNA. The SFTSV RNA-positive rate peaked (52.2%) in samples collected ≤7 days after onset and then showed a decreasing trend. The detection rate of SFTSV-specific IgM antibody was 30.5% (46/151) and was highest in samples collected between 8 and 14 days (43.3%, 26/60). The positive rate of SFTSV-specific IgG antibody (17.9%, 27/151) showed an increasing trend with the specimen collection time. In total, 74.8% (113/151) of sera samples had the same SFTSV RNA and IgM antibody detection results. However, 23.2% (29/125) of SFTSV RNA-negative cases were IgM antibody-positive, and 8.6% (9/105) of IgM antibody-negative cases were SFTSV RNA-positive. CONCLUSIONS: SFTSV RNA detection was preferred for SFTSV infection during disease surveillance. For highly suspected SFTS cases, IgM antibody is suggested to make a comprehensive judgement.


Assuntos
Anticorpos Antivirais/sangue , Infecções por Bunyaviridae/epidemiologia , Infecções por Bunyaviridae/virologia , Phlebovirus/genética , RNA Viral/sangue , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , China/epidemiologia , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Febre/virologia , Humanos , Imunoglobulina M/sangue , Masculino , Pessoa de Meia-Idade , Phlebovirus/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Trombocitopenia/epidemiologia , Trombocitopenia/virologia
6.
J Virol ; 91(14)2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28468880

RESUMO

Severe complications of Zika virus (ZIKV) infection might be caused by inflammation, but how ZIKV induces proinflammatory cytokines is not understood. In this study, we show opposite regulatory effects of the ZIKV NS5 protein on interferon (IFN) signaling. Whereas ZIKV and its NS5 protein were potent suppressors of type I and type III IFN signaling, they were found to activate type II IFN signaling. Inversely, IFN-γ augmented ZIKV replication. NS5 interacted with STAT2 and targeted it for ubiquitination and degradation, but it had no influence on STAT1 stability or nuclear translocation. The recruitment of STAT1-STAT2-IRF9 to IFN-ß-stimulated genes was compromised when NS5 was expressed. Concurrently, the formation of STAT1-STAT1 homodimers and their recruitment to IFN-γ-stimulated genes, such as the gene encoding the proinflammatory cytokine CXCL10, were augmented. Silencing the expression of an IFN-γ receptor subunit or treatment of ZIKV-infected cells with a JAK2 inhibitor suppressed viral replication and viral induction of IFN-γ-stimulated genes. Taken together, our findings provide a new mechanism by which the ZIKV NS5 protein differentially regulates IFN signaling to facilitate viral replication and cause diseases. This activity might be shared by a group of viral IFN modulators.IMPORTANCE Mammalian cells produce three types of interferons to combat viral infection and to control host immune responses. To replicate and cause diseases, pathogenic viruses have developed different strategies to defeat the action of host interferons. Many viral proteins, including the Zika virus (ZIKV) NS5 protein, are known to be able to suppress the antiviral property of type I and type III interferons. Here we further show that the ZIKV NS5 protein can also boost the activity of type II interferon to induce cellular proteins that promote inflammation. This is mediated by the differential effect of the ZIKV NS5 protein on a pair of cellular transcription factors, STAT1 and STAT2. NS5 induces the degradation of STAT2 but promotes the formation of STAT1-STAT1 protein complexes, which activate genes controlled by type II interferon. A drug that specifically inhibits the IFN-γ receptor or STAT1 shows an anti-ZIKV effect and might also have anti-inflammatory activity.


Assuntos
Interferon gama/metabolismo , Proteínas não Estruturais Virais/imunologia , Zika virus/imunologia , Linhagem Celular , Humanos , Ligação Proteica , Fator de Transcrição STAT2/metabolismo , Transdução de Sinais
7.
BMC Infect Dis ; 18(1): 66, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29402229

RESUMO

BACKGROUND: Severe fever with thrombocytopenia syndrome (SFTS) is a severe viral disease caused by SFTSV. It is important to estimate the rate of missed SFTS diagnosis and to further understand the actual incidence in high endemic areas in China. METHODS: This study was conducted in two high SFTS endemic provinces in 2015. Patients hospitalized in 2014 or within 1 year before investigation were selected after considering their clinical manifestations, specifically, fever, platelet, and white blood cell. During retrospective investigation, sera were collected to detect SFTSV antibodies to assess SFTSV infection. To further understand SFTSV infection, acute phase sera were detected; SFTSV infection rate among a healthy population was also investigated to determine the basic infection level. RESULTS: In total, 246 hospitalized cases were included, including 83 cases (33.7%) with fever, thrombocytopenia and leukopenia, 38 cases (15.4%) with fever and thrombocytopenia but without leukopenia, and 125 cases (50.8%) without fever but with thrombocytopenia and leukopenia. In total, 13 patients (5.3%) were SFTSV IgM antibody-positive, 48 (19.5%) were IgG-positive. Of the 13 IgM-positive cases, 11 (84.6%) were IgG-positive (9 with titres ≥1:400). Seropositive rates of antibodies were high (8.4% for IgM and 30.1% for IgG) in patients with fever, thrombocytopenia and leukopenia. Furthermore, among IgG-positive cases in this group, 76% (19/25) of patients' IgG antibody titres were ≥1:400. Additionally, 28 of 246 cases were initially diagnosed with suspected SFTS and were then excluded, and 218 patients were never diagnosed with SFTS; the seropositive rates of IgM and IgG in these two groups were 25% and 67.9% and 2.8% and 13.3%, respectively. These rates were 64.3% and 21.4% in 14 sera collected during acute phase of the 28 cases mentioned above. Seropositive rate of SFTSV IgG was only 1.3% in the patient-matched healthy group, and no IgM antibody was detected. A preliminary estimate of 8.3% of SFTS cases were missed in SFTS high endemic provinces. CONCLUSIONS: The actual SFTS incidence was underestimated. Effective measures such as adding a new SFTS case category - "SFTS clinical diagnosis cases" or using serological detection methods during acute phase should be considered to avoid missed diagnoses.


Assuntos
Infecções por Bunyaviridae/epidemiologia , Febre/epidemiologia , Trombocitopenia/epidemiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Antivirais/sangue , China/epidemiologia , Feminino , Febre/complicações , Hospitalização , Humanos , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Incidência , Leucopenia/complicações , Leucopenia/epidemiologia , Masculino , Pessoa de Meia-Idade , Phlebovirus/imunologia , Estudos Retrospectivos , Trombocitopenia/complicações , Adulto Jovem
8.
J Biol Chem ; 291(31): 16138-49, 2016 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-27226560

RESUMO

Synaptogyrin-2 is a non-neuronal member of the synaptogyrin family involved in synaptic vesicle biogenesis and trafficking. Little is known about the function of synaptogyrin-2. Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease characterized by high fever, thrombocytopenia, and leukocytopenia with high mortality, caused by a novel tick-borne phlebovirus in the family Bunyaviridae. Our previous studies have shown that the viral nonstructural protein NSs forms inclusion bodies (IBs) that are involved in viral immune evasion, as well as viral RNA replication. In this study, we sought to elucidate the mechanism by which NSs formed the IBs, a lipid droplet-based structure confirmed by NSs co-localization with perilipin A and adipose differentiation-related protein (ADRP). Through a high throughput screening, we identified synaptogyrin-2 to be highly up-regulated in response to SFTS bunyavirus (SFTSV) infection and to be a promoter of viral replication. We demonstrated that synaptogyrin-2 interacted with NSs and was translocated into the IBs, which were reconstructed from lipid droplets into large structures in infection. Viral RNA replication decreased, and infectious virus titers were lowered significantly when synaptogyrin-2 was silenced in specific shRNA-expressing cells, which correlated with the reduced number of the large IBs restructured from regular lipid droplets. We hypothesize that synaptogyrin-2 is essential to promoting the formation of the IBs to become virus factories for viral RNA replication through its interaction with NSs. These findings unveil the function of synaptogyrin-2 as an enhancer in viral infection.


Assuntos
Infecções por Bunyaviridae/metabolismo , Phlebovirus/fisiologia , Sinaptogirinas/metabolismo , Doenças Transmitidas por Carrapatos/metabolismo , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/fisiologia , Animais , Infecções por Bunyaviridae/genética , Chlorocebus aethiops , Células HeLa , Humanos , Corpos de Inclusão Viral/genética , Corpos de Inclusão Viral/metabolismo , Corpos de Inclusão Viral/virologia , RNA Viral/biossíntese , RNA Viral/genética , Sinaptogirinas/genética , Doenças Transmitidas por Carrapatos/genética , Células Vero , Proteínas não Estruturais Virais/genética
9.
Virol J ; 14(1): 133, 2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28720142

RESUMO

BACKGROUND: Hantaan and Seoul viruses, in the Hantavirus genus, are known to cause hemorrhagic fever with renal syndrome (HFRS). The plaque reduction neutralization test (PRNT), as conventional neutralization test for hantaviruses, is laborious and time-consuming. Alternatives to PRNT for hantaviruses are required. METHODS: In this study, the methods for Hantaan and Seoul viruses serological typing including microneutralization test (MNT), pseudoparticle neutralization test (PPNT) and immunofluorescence assay based on viral glycoproteins (IFA-GP) were developed and compared with PRNT using a panel of 74 sera including 44 convalescent sera of laboratory confirmed HFRS patients and 30 patients sera of non-hantavirus infection. Antibody titres and serotyping obtained with different methods above were analyzed by paired-t, linear correlation, McNemar χ2 and Kappa agreement tests. RESULTS: Antibody titres obtained with MNT50, PPNT50 and IFA-GP were significantly correlated with that obtained with PRNT50 (p < 0.001). GMT determined by PPNT50 was statistically higher than that determined by PRNT50 (p < 0.001), while GMT determined by MNT50 and IFA-GP were equal with (p > 0.05) and less than (p < 0.001) that obtained with PRNT50 respectively. Serotyping obtained with MNT50 and PRNT50, PPNT50 and PRNT50 were highly consistent (p < 0.001), whereas that obtained with IFA-GP and PRNT50 were moderately consistent (p < 0.001). There were no significant differences for serotyping between PRNT50 and MNT50, as well as PRNT50 and PPNT50 (p > 0.05). IFA-GP was less sensitive than PRNT50 and MNT50 for serotyping of hantaviruses infection (p < 0.05). However, for 79.5% (35/44) samples, serotyping determined by IFA-GP and PRNT50 were consistent. CONCLUSIONS: MNT50 and PPNT50 both can be used as simple and rapid alternatives to PRNT50, and MNT50 is more specific while PPNT50 is more sensitive than other assays for neutralizing antibody determination. So far, this work has been the most comprehensive comparison of alternatives to PRNT.


Assuntos
Anticorpos Antivirais/sangue , Vírus Hantaan/imunologia , Vírus Seoul/imunologia , Sorotipagem/métodos , Humanos , Sensibilidade e Especificidade
10.
Biologicals ; 46: 92-98, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28173977

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) is caused by a phlebovirus of the Bunyaviridae family, which is designated as SFTS virus (SFTSV). To our knowledge, no efficient SFTSV vaccine exists. Here, we report the identification of a standard virus strain for the eight major SFTSV strains circulating in China for use in evaluating the SFTSV vaccine. Rabbits were immunized with the SFTSV strains and the cross-neutralization capacities of SFTSV anti-sera were determined in microculture cytopathic effect (CPE)-inhibition assays. The mean cross-neutralization capacity of the eight SFTSV anti-sera ranged from 62.4 to 142.6%, compared to autologous strains. The HB29 strain demonstrated strong cross-reactivity with heterologous antibodies, and 33 serum samples from SFTS patients efficiently neutralized HB29, suggesting its broad cross-reactivity. In addition, HB29 demonstrated good replication in Vero and MRC-5 cells (8.0 and 6.0 lg 50% cell culture-infectious dose/mL, respectively) and significant CPE, which satisfied the requirements for a standard virus strain. The HB29 isolate was proven identical to the reported HB29 strain by DNA sequencing, and showed high homology in the S segments with other SFTSV strains (94.8-99.7%). Our results suggest that HB29 may be the best candidate standard strain for use in SFTS vaccine development in China.


Assuntos
Infecções por Bunyaviridae/imunologia , Testes de Neutralização/métodos , Phlebovirus/imunologia , Vacinas Virais/imunologia , Sequência de Aminoácidos , Animais , Povo Asiático , Infecções por Bunyaviridae/etnologia , Infecções por Bunyaviridae/virologia , Linhagem Celular , China , Chlorocebus aethiops , Reações Cruzadas/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Phlebovirus/genética , Phlebovirus/fisiologia , Filogenia , Controle de Qualidade , Coelhos , Padrões de Referência , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Células Vero , Proteínas do Envelope Viral/classificação , Proteínas do Envelope Viral/genética , Vacinas Virais/normas
11.
J Virol ; 89(6): 3026-37, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25552716

RESUMO

UNLABELLED: Severe fever with thrombocytopenia syndrome virus (SFTSV) is an emerging tick-borne pathogen that was first reported in China in 2009. Phylogenetic analysis of the viral genome showed that SFTS virus represents a new lineage within the Phlebovirus genus, distinct from the existing sandfly fever and Uukuniemi virus groups, in the family Bunyaviridae. SFTS disease is characterized by gastrointestinal symptoms, chills, joint pain, myalgia, thrombocytopenia, leukocytopenia, and some hemorrhagic manifestations with a case fatality rate of about 2 to 15%. Here we report the development of reverse genetics systems to study STFSV replication and pathogenesis. We developed and optimized functional T7 polymerase-based M- and S-segment minigenome assays, which revealed errors in the published terminal sequences of the S segment of the Hubei 29 strain of SFTSV. We then generated recombinant viruses from cloned cDNAs prepared to the antigenomic RNAs both of the minimally passaged virus (HB29) and of a cell culture-adapted strain designated HB29pp. The growth properties, pattern of viral protein synthesis, and subcellular localization of viral N and NSs proteins of wild-type HB29pp (wtHB29pp) and recombinant HB29pp viruses were indistinguishable. We also show that the viruses fail to shut off host cell polypeptide production. The robust reverse genetics system described will be a valuable tool for the design of therapeutics and the development of killed and attenuated vaccines against this important emerging pathogen. IMPORTANCE: SFTSV and related tick-borne phleboviruses such as Heartland virus are emerging viruses shown to cause severe disease in humans in the Far East and the United States, respectively. Study of these novel pathogens would be facilitated by technology to manipulate these viruses in a laboratory setting using reverse genetics. Here, we report the generation of infectious SFTSV from cDNA clones and demonstrate that the behavior of recombinant viruses is similar to that of the wild type. This advance will allow for further dissection of the roles of each of the viral proteins in the context of virus infection, as well as help in the development of antiviral drugs and protective vaccines.


Assuntos
Febre por Flebótomos/virologia , Phlebovirus/genética , Genética Reversa/métodos , Sequência de Aminoácidos , Sequência de Bases , China , Feminino , Genoma Viral , Humanos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Phlebovirus/química , Phlebovirus/metabolismo , Alinhamento de Sequência , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo
12.
J Infect Dis ; 211(6): 915-25, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25326554

RESUMO

SFTS virus (SFTSV) is a highly pathogenic bunyavirus that causes severe fever with thrombocytopenia syndrome (SFTS), an emerging infectious disease in China. Laboratory mice have been reported to be susceptible to SFTSV infection, but the infection in nonhuman primates has not been investigated. This study is the first to report that, in rhesus macaques, SFTSV does not cause severe symptoms or death but causes fever, thrombocytopenia, leukocytopenia, and increased levels of transaminases and myocardial enzymes in blood. Viremia, virus-specific immunoglobulin M and immunoglobulin G antibodies, and neutralizing antibodies were identified in all infected macaques. Levels of the cytokines interferon γ, eotaxin, tumor necrosis factor α, and macrophage inflammatory protein 1ß were significantly elevated in the blood. Minor pathological lesions were observed in the liver and kidney during the late stages of infection. Overall, SFTSV infection in rhesus macaques resembled mild SFTS in humans.


Assuntos
Infecções por Bunyaviridae/veterinária , Macaca mulatta/virologia , Doenças dos Macacos/virologia , Phlebovirus/imunologia , Animais , Anticorpos Antivirais/sangue , Infecções por Bunyaviridae/sangue , Infecções por Bunyaviridae/imunologia , Citocinas/sangue , Modelos Animais de Doenças , Feminino , Humanos , Rim/patologia , Rim/virologia , Fígado/patologia , Fígado/virologia , Macaca mulatta/imunologia , Camundongos , Doenças dos Macacos/sangue , Doenças dos Macacos/imunologia , RNA Viral/sangue
14.
J Gen Virol ; 96(11): 3204-3211, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26353965

RESUMO

Severe fever with thrombocytopenia syndrome virus (SFTSV) is an emerging tick-borne pathogen causing significant morbidity and mortality in Asia. NSs protein of SFTSV is known to perturb type I IFN induction and signalling, but the mechanism remains to be fully understood. Here, we showed the suppression of both type I and type III IFN signalling by SFTSV NSs protein is mediated through inhibition of STAT1 phosphorylation and activation. Infection with live SFTSV or expression of NSs potently suppressed IFN-stimulated genes but not NFkB activation. NSs was capable of counteracting the activity of IFN-α1, IFN-ß, IFN-λ1 and IFN-λ2. Mechanistically, NSs associated with STAT1 and STAT2, mitigated IFN-ß-induced phosphorylation of STAT1 at S727, and reduced the expression and activity of STAT1 protein in IFN-ß-treated cells, resulting in the inhibition of STAT1 and STAT2 recruitment to IFNstimulated promoters. Taken together, SFTSV NSs protein is an IFN antagonist that suppresses phosphorylation and activation of STAT1.


Assuntos
Interferon-alfa/genética , Interferon beta/genética , Interleucinas/genética , Febre por Flebótomos/genética , Phlebovirus/metabolismo , Fator de Transcrição STAT1/metabolismo , Proteínas não Estruturais Virais/metabolismo , Humanos , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Interferons , Interleucinas/metabolismo , Febre por Flebótomos/metabolismo , Febre por Flebótomos/virologia , Phlebovirus/genética , Fosforilação , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT2/genética , Fator de Transcrição STAT2/metabolismo , Transdução de Sinais , Proteínas não Estruturais Virais/genética
15.
J Virol ; 88(6): 3067-76, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24335286

RESUMO

Cells are equipped with pattern recognition receptors (PRRs) such as the Toll-like and RIG-I-like receptors that mount innate defenses against viruses. However, viruses have evolved multiple strategies to evade or thwart host antiviral responses. Viral inclusion bodies (IBs), which are accumulated aggregates of viral proteins, are commonly formed during the replication of some viruses in infected cells, but their role in viral immune evasion has rarely been explored. Severe fever with thrombocytopenia syndrome (SFTS) is an emerging febrile illness caused by a novel phlebovirus in the Bunyaviridae. The SFTS viral nonstructural protein NSs can suppress host beta interferon (IFN-ß) responses. NSs can form IBs in infected and transfected cells. Through interaction with tank-binding kinase 1 (TBK1), viral NSs was able to sequester the IKK complex, including IKKε and IRF3, into IBs, although NSs did not interact with IKKε or IRF3 directly. When cells were infected with influenza A virus, IRF3 was phosphorylated and active phosphorylated IRF3 (p-IRF3) was translocated into the nucleus. In the presence of NSs, IRF3 could still be phosphorylated, but p-IRF3 was trapped in cytoplasmic IBs, resulting in reduced IFN-ß induction and enhanced viral replication. Sequestration of the IKK complex and active IRF3 into viral IBs through the interaction of NSs and TBK1 is a novel mechanism for viral evasion of innate immunity.


Assuntos
Quinase I-kappa B/metabolismo , Evasão da Resposta Imune , Corpos de Inclusão Viral/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Febre por Flebótomos/metabolismo , Phlebovirus/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/imunologia , Imunidade Inata , Corpos de Inclusão Viral/imunologia , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Febre por Flebótomos/imunologia , Febre por Flebótomos/virologia , Phlebovirus/genética , Phlebovirus/metabolismo , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Proteínas não Estruturais Virais/imunologia , Proteínas não Estruturais Virais/metabolismo
16.
FASEB J ; 28(6): 2504-16, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24599967

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) virus is an emerging bunyavirus that causes a hemorrhagic fever with a high mortality rate. The virus is likely tick-borne and replicates primarily in hemopoietic cells, which may lead to disregulation of proinflammatory cytokine induction and loss of leukocytes and platelets. The viral genome contains L, M, and S segments encoding a viral RNA polymerase, glycoproteins G(n) and G(c), nucleoprotein (NP), and a nonstructural S segment (NSs) protein. NSs protein is involved in the regulation of host innate immune responses and suppression of IFNß-promoter activities. In this article, we demonstrate that NSs protein can form viroplasm-like structures (VLSs) in infected and transfected cells. NSs protein molecules interact with one another, interact with NP, and were associated with viral RNA in infected cells, suggesting that NSs protein may be involved in viral replication. Furthermore, we observed that NSs-formed VLS colocalized with lipid droplets and that inhibitors of fatty acid biosynthesis decreased VLS formation or viral replication in transfected and infected cells. Finally, we have demonstrated that viral dsRNAs were also localized in VLS in infected cells, suggesting that NSs-formed VLS may be implicated in the replication of SFTS bunyavirus. These findings identify a novel function of nonstructural NSs in SFTSV-infected cells where it is a scaffolding component in a VLS functioning as a virus replication factory. This function is in addition to the role of NSs protein in modulating host responses that will broaden our understanding of viral pathogenesis of phleboviruses.


Assuntos
Infecções por Bunyaviridae/virologia , Corpos de Inclusão Viral/metabolismo , Phlebovirus/ultraestrutura , Proteínas não Estruturais Virais/metabolismo , Animais , Infecções por Bunyaviridae/imunologia , Chlorocebus aethiops , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Metabolismo dos Lipídeos , Nucleoproteínas/metabolismo , RNA Viral/genética , Células Vero , Replicação Viral/fisiologia
17.
Acta Biochim Biophys Sin (Shanghai) ; 47(10): 815-23, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26188200

RESUMO

Rabies virus (RV) causes a fatal disease in both human and animals. The disease can be prevented by post-exposure prophylaxis in individuals exposed to RV. However, the neutralization effect is limited after the virus enters into the host cells. So, it is important to identify new targets for rabies therapy. In this study, a human antibody RV1A2 specific to RV phosphoprotein (RV-P) was generated from a human naïve immune antibody library. The antibody recognized all forms of the phosphoproteins including the full length (P1) and short length of the P proteins (P2, P3, P4, and P5). The epitope mapping and the molecular docking of antigen-antibody complex showed that the antibody targets at a conserved epitope of 'VLGWV' ranging from amino acid (aa) 262 to 266 at C-terminal domain of the P protein, which locates at a hydrophobic pocket region in the C-terminal of the RV-P. The aa W265 within the epitope is on the flat surface of the domain, suggesting that it may be a critical amino acid for the functions of the P protein. Our results further showed that intracellular antibody RV1A2 which targets at the C-terminal domain of the P protein could effectively inhibit RV propagation 2-4 days post infection. These results suggest that the conserved C-terminal domain may be used as a new target for drug discovery, which highlights an intracellular inhibition of RV propagation and provides a potential novel way to treat RV infection.


Assuntos
Anticorpos Antivirais/imunologia , Fosfoproteínas/imunologia , Fosfoproteínas/metabolismo , Vírus da Raiva/química , Vírus da Raiva/imunologia , Replicação Viral/fisiologia , Anticorpos Antivirais/administração & dosagem , Sítios de Ligação , Células HEK293 , Humanos , Ligação Proteica , Estrutura Terciária de Proteína , Vírus da Raiva/efeitos dos fármacos , Relação Estrutura-Atividade , Replicação Viral/efeitos dos fármacos
18.
Proc Natl Acad Sci U S A ; 109(25): 10053-8, 2012 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-22665769

RESUMO

The discovery of an emerging viral disease, severe fever with thrombocytopenia syndrome (SFTS), caused by SFTS virus (SFTSV), has prompted the need to understand pathogenesis of SFTSV. We are unique in establishing an infectious model of SFTS in C57/BL6 mice, resulting in hallmark symptoms of thrombocytopenia and leukocytopenia. Viral RNA and histopathological changes were identified in the spleen, liver, and kidney. However, viral replication was only found in the spleen, which suggested the spleen to be the principle target organ of SFTSV. Moreover, the number of macrophages and platelets were largely increased in the spleen, and SFTSV colocalized with platelets in cytoplasm of macrophages in the red pulp of the spleen. In vitro cellular assays further revealed that SFTSV adhered to mouse platelets and facilitated the phagocytosis of platelets by mouse primary macrophages, which in combination with in vivo findings, suggests that SFTSV-induced thrombocytopenia is caused by clearance of circulating virus-bound platelets by splenic macrophages. Thus, this study has elucidated the pathogenic mechanisms of thrombocytopenia in a mouse model resembling human SFTS disease.


Assuntos
Febre/virologia , Trombocitopenia/virologia , Viroses/virologia , Animais , Plaquetas/imunologia , Adesão Celular , Modelos Animais de Doenças , Febre/etiologia , Febre/patologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose , RNA Viral/genética , Trombocitopenia/complicações , Trombocitopenia/patologia , Viroses/complicações , Viroses/patologia
19.
N Engl J Med ; 364(16): 1523-32, 2011 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-21410387

RESUMO

BACKGROUND: Heightened surveillance of acute febrile illness in China since 2009 has led to the identification of a severe fever with thrombocytopenia syndrome (SFTS) with an unknown cause. Infection with Anaplasma phagocytophilum has been suggested as a cause, but the pathogen has not been detected in most patients on laboratory testing. METHODS: We obtained blood samples from patients with the case definition of SFTS in six provinces in China. The blood samples were used to isolate the causal pathogen by inoculation of cell culture and for detection of viral RNA on polymerase-chain-reaction assay. The pathogen was characterized on electron microscopy and nucleic acid sequencing. We used enzyme-linked immunosorbent assay, indirect immunofluorescence assay, and neutralization testing to analyze the level of virus-specific antibody in patients' serum samples. RESULTS: We isolated a novel virus, designated SFTS bunyavirus, from patients who presented with fever, thrombocytopenia, leukocytopenia, and multiorgan dysfunction. RNA sequence analysis revealed that the virus was a newly identified member of the genus phlebovirus in the Bunyaviridae family. Electron-microscopical examination revealed virions with the morphologic characteristics of a bunyavirus. The presence of the virus was confirmed in 171 patients with SFTS from six provinces by detection of viral RNA, specific antibodies to the virus in blood, or both. Serologic assays showed a virus-specific immune response in all 35 pairs of serum samples collected from patients during the acute and convalescent phases of the illness. CONCLUSIONS: A novel phlebovirus was identified in patients with a life-threatening illness associated with fever and thrombocytopenia in China. (Funded by the China Mega-Project for Infectious Diseases and others.).


Assuntos
Infecções por Bunyaviridae/virologia , Doenças Transmissíveis Emergentes/virologia , Orthobunyavirus/isolamento & purificação , Trombocitopenia/virologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Anticorpos Antivirais/sangue , Infecções por Bunyaviridae/complicações , Infecções por Bunyaviridae/epidemiologia , China/epidemiologia , Doenças Transmissíveis Emergentes/epidemiologia , Feminino , Febre/virologia , Genoma Viral , Humanos , Ixodidae/virologia , Masculino , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Orthobunyavirus/classificação , Orthobunyavirus/genética , Orthobunyavirus/imunologia , Filogenia , RNA Viral/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
J Virol ; 87(12): 6829-39, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23576501

RESUMO

Severe fever with thrombocytopenia syndrome is an emerging infectious disease caused by a novel bunyavirus (SFTSV). Lack of vaccines and inadequate therapeutic treatments have made the spread of the virus a global concern. Viral nucleocapsid protein (N) is essential for its transcription and replication. Here, we present the crystal structures of N from SFTSV and its homologs from Buenaventura (BUE) and Granada (GRA) viruses. The structures reveal that phleboviral N folds into a compact core domain and an extended N-terminal arm that mediates oligomerization, such as tetramer, pentamer, and hexamer of N assemblies. Structural superimposition indicates that phleboviral N adopts a conserved architecture and uses a similar RNA encapsidation strategy as that of RVFV-N. The RNA binding cavity runs along the inner edge of the ring-like assembly. A triple mutant of SFTSV-N, R64D/K67D/K74D, almost lost its ability to bind RNA in vitro, is deficient in its ability to transcribe and replicate. Structural studies of the mutant reveal that both alterations in quaternary assembly and the charge distribution contribute to the loss of RNA binding. In the screening of inhibitors Suramin was identified to bind phleboviral N specifically. The complex crystal structure of SFTSV-N with Suramin was refined to a 2.30-Å resolution. Suramin was found sitting in the putative RNA binding cavity of SFTSV-N. The inhibitory effect of Suramin on SFTSV replication was confirmed in Vero cells. Therefore, a common Suramin-based therapeutic approach targeting SFTSV-N and its homologs could be developed for containing phleboviral outbreaks.


Assuntos
Proteínas do Nucleocapsídeo/química , Proteínas do Nucleocapsídeo/uso terapêutico , Febre por Flebótomos/tratamento farmacológico , Phlebovirus/efeitos dos fármacos , Suramina/química , Suramina/uso terapêutico , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , Cristalização , Modelos Moleculares , Dados de Sequência Molecular , Proteínas do Nucleocapsídeo/genética , Proteínas do Nucleocapsídeo/metabolismo , Febre por Flebótomos/virologia , Dobramento de Proteína , RNA Viral/genética , RNA Viral/metabolismo , Análise de Sequência de DNA , Relação Estrutura-Atividade , Suramina/metabolismo , Células Vero , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA