Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Viruses ; 13(2)2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33672327

RESUMO

The Bunyavirales order accommodates related viruses (bunyaviruses) with segmented, linear, single-stranded, negative- or ambi-sense RNA genomes. Their glycoproteins form capsomeric projections or spikes on the virion surface and play a crucial role in virus entry, assembly, morphogenesis. Bunyavirus glycoproteins are encoded by a single RNA segment as a polyprotein precursor that is co- and post-translationally cleaved by host cell enzymes to yield two mature glycoproteins, Gn and Gc (or GP1 and GP2 in arenaviruses). These glycoproteins undergo extensive N-linked glycosylation and despite their cleavage, remain associated to the virion to form an integral transmembrane glycoprotein complex. This review summarizes recent advances in our understanding of the molecular biology of bunyavirus glycoproteins, including their processing, structure, and known interactions with host factors that facilitate cell entry.


Assuntos
Infecções por Bunyaviridae/metabolismo , Orthobunyavirus/metabolismo , Receptores Virais/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Animais , Infecções por Bunyaviridae/genética , Infecções por Bunyaviridae/virologia , Humanos , Orthobunyavirus/química , Orthobunyavirus/genética , Ligação Proteica , Processamento de Proteína Pós-Traducional , Receptores Virais/genética , Proteínas do Envelope Viral/genética
2.
Viruses ; 12(9)2020 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-32872451

RESUMO

Severe fever with thrombocytopenia syndrome virus (SFTSV) is an emerging human pathogen, endemic in areas of China, Japan, and the Korea (KOR). It is primarily transmitted through infected ticks and can cause a severe hemorrhagic fever disease with case fatality rates as high as 30%. Despite its high virulence and increasing prevalence, molecular and functional studies in situ are scarce due to the limited availability of high-titer SFTSV exposure stocks. During the course of field virologic surveillance in 2017, we detected SFTSV in ticks and in a symptomatic soldier in a KOR Army training area. SFTSV was isolated from the ticks producing a high-titer viral exposure stock. Through the use of advanced genomic tools, we present here a complete, in-depth characterization of this viral stock, including a comparison with both the virus in its arthropod source and in the human case, and an in vivo study of its pathogenicity. Thanks to this detailed characterization, this SFTSV viral exposure stock constitutes a quality biological tool for the study of this viral agent and for the development of medical countermeasures, fulfilling the requirements of the main regulatory agencies.


Assuntos
Infecções por Bunyaviridae/virologia , Febres Hemorrágicas Virais/virologia , Phlebovirus/isolamento & purificação , Adulto , Animais , Infecções por Bunyaviridae/genética , Infecções por Bunyaviridae/metabolismo , Feminino , Genoma Viral , Humanos , Masculino , Camundongos , Phlebovirus/fisiologia , Filogenia , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , República da Coreia , Carrapatos/virologia
3.
J Virol ; 94(17)2020 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-32522852

RESUMO

Schmallenberg virus (SBV) is an insect-transmitted orthobunyavirus that can cause abortions and congenital malformations in the offspring of ruminants. Even though the two viral surface glycoproteins Gn and Gc are involved in host cell entry, the specific cellular receptors of SBV are currently unknown. Using genome-wide CRISPR-Cas9 forward screening, we identified 3'-phosphoadenosine 5'-phosphosulfate (PAPS) transporter 1 (PAPST1) as an essential factor for SBV infection. PAPST1 is a sulfotransferase involved in heparan sulfate proteoglycan synthesis encoded by the solute carrier family 35 member B2 gene (SLC35B2). SBV cell surface attachment and entry were largely reduced upon the knockout of SLC35B2, whereas the reconstitution of SLC35B2 in these cells fully restored their susceptibility to SBV infection. Furthermore, treatment of cells with heparinase diminished infection with SBV, confirming that heparan sulfate plays an important role in cell attachment and entry, although to various degrees, heparan sulfate was also found to be important to initiate infection by two other bunyaviruses, La Crosse virus and Rift Valley fever virus. Thus, PAPST1-triggered synthesis of cell surface heparan sulfate is required for the efficient replication of SBV and other bunyaviruses.IMPORTANCE SBV is a newly emerging orthobunyavirus (family Peribunyaviridae) that has spread rapidly across Europe since 2011, resulting in substantial economic losses in livestock farming. In this study, we performed unbiased genome-wide CRISPR-Cas9 screening and identified PAPST1, a sulfotransferase encoded by SLC35B2, as a host entry factor for SBV. Consistent with its role in the synthesis of heparan sulfate, we show that this activity is required for efficient infection by SBV. A comparable dependency on heparan sulfate was also observed for La Crosse virus and Rift Valley fever virus, highlighting the importance of heparan sulfate for host cell infection by bunyaviruses. Thus, the present work provides crucial insights into virus-host interactions of important animal and human pathogens.


Assuntos
Infecções por Bunyaviridae/genética , Infecções por Bunyaviridae/virologia , Sistemas CRISPR-Cas , Orthobunyavirus/genética , Orthobunyavirus/fisiologia , Animais , Bunyaviridae , Chlorocebus aethiops , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Europa (Continente) , Técnicas de Inativação de Genes , Células HEK293 , Heparitina Sulfato/metabolismo , Humanos , Gado , Glicoproteínas de Membrana/genética , Orthobunyavirus/patogenicidade , Vírus da Febre do Vale do Rift , Transportadores de Sulfato/metabolismo , Sulfotransferases/metabolismo , Células Vero , Ligação Viral
4.
Cell Rep ; 30(13): 4370-4385.e7, 2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32234474

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) virus (SFTSV) is an emerging tick-borne virus that carries a high fatality rate of 12%-50%. In-depth understanding of the SFTSV-induced pathogenesis mechanism is critical for developing effective anti-SFTS therapeutics. Here, we report transcriptomic analysis of blood samples from SFTS patients. We observe a strong correlation between inflammatory responses and disease progression and fatal outcome. Quantitative proteomic analysis of SFTSV infection confirms the induction of inflammation and further reveals virus-induced mitochondrial dysfunction. Mechanistically, SFTSV infection triggers BCL2 antagonist/killer 1 (BAK) upregulation and BAK/BCL2-associated X (BAX) activation, leading to mitochondrial DNA (mtDNA) oxidization and subsequent cytosolic release. The cytosolic mtDNA binds and triggers NLRP3 inflammasome activation. Notably, the BAK expression level correlates with SFTS disease progression and fatal outcome. These findings provide insights into the clinical features and molecular underpinnings of severe SFTS, which may aid in patient care and therapeutic design, and may also be conserved during infection by other highly pathogenic viruses.


Assuntos
Infecções por Bunyaviridae/metabolismo , DNA Mitocondrial/metabolismo , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Phlebovirus/fisiologia , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo , Adulto , Animais , Infecções por Bunyaviridae/genética , Infecções por Bunyaviridae/virologia , Linhagem Celular , Modelos Animais de Doenças , Progressão da Doença , Feminino , Humanos , Inflamação/genética , Interleucina-1beta/metabolismo , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Modelos Biológicos , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Receptor 8 Toll-Like/metabolismo , Transcriptoma/genética
5.
J Virol ; 94(6)2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-31852783

RESUMO

Nuclear factor erythroid 2-related factor 2 (Nrf2) dissociates from its inhibitor, Keap1, upon stress signals and subsequently induces an antioxidant response that critically controls the viral life cycle and pathogenesis. Besides intracellular Fc receptor function, tripartite motif 21 (TRIM21) E3 ligase plays an essential role in the p62-Keap1-Nrf2 axis pathway for redox homeostasis. Specifically, TRIM21-mediated p62 ubiquitination abrogates p62 oligomerization and sequestration activity and negatively regulates the Keap1-Nrf2-mediated antioxidant response. A number of viruses target the Nrf2-mediated antioxidant response to generate an optimal environment for their life cycle. Here we report that a nonstructural protein (NSs) of severe fever with thrombocytopenia syndrome virus (SFTSV) interacts with and inhibits TRIM21 to activate the Nrf2 antioxidant signal pathway. Mass spectrometry identified TRIM21 to be a binding protein for NSs. NSs bound to the carboxyl-terminal SPRY subdomain of TRIM21, enhancing p62 stability and oligomerization. This facilitated p62-mediated Keap1 sequestration and ultimately increased Nrf2-mediated transcriptional activation of antioxidant genes, including those for heme oxygenase 1, NAD(P)H quinone oxidoreductase 1, and CD36. Mutational analysis found that the NSs-A46 mutant, which no longer interacted with TRIM21, was unable to increase Nrf2-mediated transcriptional activation. Functionally, the NS wild type (WT), but not the NSs-A46 mutant, increased the surface expression of the CD36 scavenger receptor, resulting in an increase in phagocytosis and lipid uptake. A combination of reverse genetics and assays with Ifnar-/- mouse models revealed that while the SFTSV-A46 mutant replicated similarly to wild-type SFTSV (SFTSV-WT), it showed weaker pathogenic activity than SFTSV-WT. These data suggest that the activation of the p62-Keap1-Nrf2 antioxidant response induced by the NSs-TRIM21 interaction contributes to the development of an optimal environment for the SFTSV life cycle and efficient pathogenesis.IMPORTANCE Tick-borne diseases have become a growing threat to public health. SFTSV, listed by the World Health Organization as a prioritized pathogen, is an emerging phlebovirus, and fatality rates among those infected with this virus are high. Infected Haemaphysalis longicornis ticks are the major source of human SFTSV infection. In particular, the recent spread of this tick to over 12 states in the United States has increased the potential for outbreaks of this disease beyond Far East Asia. Due to the lack of therapies and vaccines against SFTSV infection, there is a pressing need to understand SFTSV pathogenesis. As the Nrf2-mediated antioxidant response affects viral life cycles, a number of viruses deregulate Nrf2 pathways. Here we demonstrate that the SFTSV NSs inhibits the TRIM21 function to upregulate the p62-Keap1-Nrf2 antioxidant pathway for efficient viral pathogenesis. This study not only demonstrates the critical role of SFTSV NSs in viral pathogenesis but also suggests potential future therapeutic approaches to treat SFTSV-infected patients.


Assuntos
Infecções por Bunyaviridae/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Phlebovirus/metabolismo , Ribonucleoproteínas/metabolismo , Proteína Sequestossoma-1/metabolismo , Transdução de Sinais , Proteínas não Estruturais Virais/metabolismo , Animais , Infecções por Bunyaviridae/genética , Infecções por Bunyaviridae/patologia , Células HEK293 , Humanos , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Camundongos , Camundongos Knockout , Fator 2 Relacionado a NF-E2/genética , Phlebovirus/genética , Ribonucleoproteínas/genética , Proteína Sequestossoma-1/genética , Proteínas não Estruturais Virais/genética
6.
J Virol ; 94(6)2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-31852787

RESUMO

Severe fever with thrombocytopenia syndrome virus (SFTSV) is a newly identified phlebovirus associated with severe hemorrhagic fever in humans. While many viruses subvert the host cell cycle to promote viral growth, it is unknown whether this is a strategy employed by SFTSV. In this study, we investigated how SFTSV manipulates the cell cycle and the effect of the host cell cycle on SFTSV replication. Our results suggest that cells arrest at the G2/M transition following infection with SFTSV. The accumulation of cells at the G2/M transition did not affect virus adsorption and entry but did facilitate viral replication. In addition, we found that SFTSV NSs, a nonstructural protein that forms viroplasm-like structures in the cytoplasm of infected cells and promotes virulence by modulating the interferon response, induces a large number of cells to arrest at the G2/M transition by interacting with CDK1. The interaction between NSs and CDK1, which is inclusion body dependent, inhibits formation and nuclear import of the cyclin B1-CDK1 complex, thereby leading to cell cycle arrest. Expression of a CDK1 loss-of-function mutant reversed the inhibitive effect of NSs on the cell cycle, suggesting that this protein is a potential antiviral target. Our study provides new insight into the role of a specific viral protein in SFTSV replication, indicating that NSs induces G2/M arrest of SFTSV-infected cells, which promotes viral replication.IMPORTANCE Severe fever with thrombocytopenia syndrome virus (SFTSV) is a tick-borne pathogen that causes severe hemorrhagic fever. Although SFTSV poses a serious threat to public health and was recently isolated, its pathogenesis remains unclear. In particular, the relationship between SFTSV infection and the host cell cycle has not been described. Here, we show for the first time that both asynchronized and synchronized SFTSV-susceptible cells arrest at the G2/M checkpoint following SFTSV infection and that the accumulation of cells at this checkpoint facilitates viral replication. We also identify a key mechanism underlying SFTSV-induced G2/M arrest, in which SFTSV NSs interacts with CDK1 to inhibit formation and nuclear import of the cyclin B1-CDK1 complex, thus preventing it from regulating cell cycle progression. Our study highlights the key role that NSs plays in SFTSV-induced G2/M arrest.


Assuntos
Infecções por Bunyaviridae/metabolismo , Proteína Quinase CDC2/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular , Phlebovirus/fisiologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Infecções por Bunyaviridae/genética , Infecções por Bunyaviridae/patologia , Proteína Quinase CDC2/genética , Ciclina B1/genética , Ciclina B1/metabolismo , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Proteínas não Estruturais Virais/genética
7.
Sci Rep ; 9(1): 12957, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31506463

RESUMO

Carcinus maenas is in the top 100 globally invasive species and harbours a wide diversity of pathogens, including viruses. We provide a detailed description for a novel bunyavirus (Carcinus maenas Portunibunyavirus 1) infecting C. maenas from its native range in the Faroe Islands. The virus genome is tripartite, including large (L) (6766 bp), medium (M) (3244 bp) and small (S) (1608 bp) negative sense, single-stranded RNA segments. Individual genomic segments are flanked by 4 bp regions of similarity (CCUG). The segments encode an RNA-dependent RNA-polymerase, glycoprotein, non-structural protein with a Zinc-Finger domain and a nucleoprotein. Most show highest identity to the 'Wenling Crustacean Virus 9' from an unidentified crustacean host. Phylogenomics of crustacean-infecting bunyaviruses place them across multiple bunyavirus families. We discuss the diversity of crustacean bunyaviruses and provide an overview of how these viruses may affect the health and survival of crustacean hosts, including those inhabiting niches outside of their native range.


Assuntos
Braquiúros/virologia , Infecções por Bunyaviridae/virologia , Variação Genética , Genética Populacional , Genoma Viral , Orthobunyavirus/crescimento & desenvolvimento , Filogenia , Sequência de Aminoácidos , Animais , Braquiúros/genética , Infecções por Bunyaviridae/genética , Orthobunyavirus/genética , Orthobunyavirus/isolamento & purificação , Homologia de Sequência
8.
J Virol ; 93(14)2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31043534

RESUMO

Several host and viral processes contribute to forming infectious virions. Polyamines are small host molecules that play diverse roles in viral replication. We previously demonstrated that polyamines are crucial for RNA viruses; however, the mechanisms by which polyamines function remain unknown. Here, we investigated the role of polyamines in the replication of the bunyaviruses Rift Valley fever virus (vaccine strain MP-12) and La Crosse virus (LACV). We found that polyamine depletion did not impact viral RNA or protein accumulation, despite significant decreases in titer. Viral particles demonstrated no change in morphology, size, or density. Thus, polyamine depletion promotes the formation of noninfectious particles. These particles interfere with virus replication and stimulate innate immune responses. We extended this phenotype to Zika virus; however, coxsackievirus did not similarly produce noninfectious particles. In sum, polyamine depletion results in the accumulation of noninfectious particles that interfere with replication and stimulate immune signaling, with important implications for targeting polyamines therapeutically, as well as for vaccine strategies.IMPORTANCE Bunyaviruses are emerging viral pathogens that cause encephalitis, hemorrhagic fevers, and meningitis. We have uncovered that diverse bunyaviruses require polyamines for productive infection. Polyamines are small, positively charged host-derived molecules that play diverse roles in human cells and in infection. In polyamine-depleted cells, bunyaviruses produce an overabundance of noninfectious particles that are indistinguishable from infectious particles. However, these particles interfere with productive infection and stimulate antiviral signaling pathways. We further find that additional enveloped viruses are similarly sensitive to polyamine depletion but that a nonenveloped enterovirus is not. We posit that polyamines are required to maintain bunyavirus infectivity and that polyamine depletion results in the accumulation of interfering noninfectious particles that limit infectivity. These results highlight a novel means by which bunyaviruses use polyamines for replication and suggest promising means to target host polyamines to reduce virus replication.


Assuntos
Poliaminas Biogênicas/imunologia , Infecções por Bunyaviridae/imunologia , Vírus Defeituosos/fisiologia , Vírus da Encefalite da Califórnia/fisiologia , Vírus da Febre do Vale do Rift/fisiologia , Vírion/fisiologia , Replicação Viral/imunologia , Infecções por Bunyaviridae/genética , Infecções por Bunyaviridae/patologia , Linhagem Celular Tumoral , Humanos
9.
J Gen Virol ; 100(3): 392-402, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30720418

RESUMO

The Nairoviridae family within the Bunyavirales order comprise tick-borne segmented negative-sense RNA viruses that cause serious disease in a broad range of mammals, yet cause a latent and lifelong infection in tick hosts. An important member of this family is Crimean-Congo haemorrhagic fever virus (CCHFV), which is responsible for serious human disease that results in case fatality rates of up to 30 %, and which exhibits the most geographically broad distribution of any tick-borne virus. Here, we explored differences in the cellular response of both mammalian and tick cells to nairovirus infection using Hazara virus (HAZV), which is a close relative of CCHFV within the CCHFV serogroup. We show that HAZV infection of human-derived SW13 cells led to induction of apoptosis, evidenced by activation of cellular caspases 3, 7 and 9. This was followed by cleavage of the classical apoptosis marker poly ADP-ribose polymerase, as well as cellular genome fragmentation. In addition, we show that the HAZV nucleocapsid (N) protein was abundantly cleaved by caspase 3 in these mammalian cells at a conserved DQVD motif exposed at the tip of its arm domain, and that cleaved HAZV-N was subsequently packaged into nascent virions. However, in stark contrast, we show for the first time that nairovirus infection of cells of the tick vector failed to induce apoptosis, as evidenced by undetectable levels of cleaved caspases and lack of cleaved HAZV-N. Our findings reveal that nairoviruses elicit diametrically opposed cellular responses in mammalian and tick cells, which may influence the infection outcome in the respective hosts.


Assuntos
Apoptose , Infecções por Bunyaviridae/fisiopatologia , Nairovirus/metabolismo , Proteínas do Nucleocapsídeo/metabolismo , Carrapatos/virologia , Motivos de Aminoácidos , Animais , Infecções por Bunyaviridae/enzimologia , Infecções por Bunyaviridae/genética , Infecções por Bunyaviridae/virologia , Caspase 3/genética , Caspase 3/metabolismo , Caspase 7/genética , Caspase 7/metabolismo , Linhagem Celular , Interações Hospedeiro-Patógeno , Humanos , Nairovirus/química , Nairovirus/genética , Proteínas do Nucleocapsídeo/química , Proteínas do Nucleocapsídeo/genética , Processamento de Proteína Pós-Traducional
10.
Virus Genes ; 55(2): 123-126, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30600431

RESUMO

In recent reports about the molecular epidemiology of Schmallenberg virus (SBV), an orthobunyavirus affecting ruminants, it was proposed that the observed sequence variability within the viral M-segment might be higher in sheep than in cattle. However, these analyses are highly biased by the sample material from which the publicly available sequences were generated. While from cattle predominantly blood samples from acutely infected animals were studied, the vast majority of ovine samples originate from malformed fetuses or newborn lambs. Therefore, the observed sequence variability is misinterpreted since the samples from malformed fetuses and lambs do not reflect circulating SBV.


Assuntos
Infecções por Bunyaviridae/virologia , Orthobunyavirus/genética , Proteínas da Matriz Viral/genética , Animais , Animais Recém-Nascidos/virologia , Infecções por Bunyaviridae/genética , Infecções por Bunyaviridae/veterinária , Bovinos , Orthobunyavirus/patogenicidade , Ruminantes/virologia , Ovinos/virologia , Proteínas da Matriz Viral/química
11.
PLoS Negl Trop Dis ; 12(6): e0006547, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29939989

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease that is caused by a novel bunyavirus, SFTSV. We assessed whether the single nucleotide polymorphisms (SNPs) in the tumor necrosis factor-alpha (TNF-α) were associated with risk to severity of SFTS. Five TNF-α SNPs (SNP1: T-1031C; SNP2: C-863A; SNP3: C-857T; SNP4: G-308A; SNP5: G-238A) were genotyped in 987 hospitalized SFTS patients and 633 asymptomatic/mild SFTSV-infected subjects of Chinese Han origin. Multivariate logistic regression analysis was used to calculate adjusted odds ratios (ORs) and 95% confidence intervals (95% CIs). The hospitalized SFTS patients had significantly lower frequency of G-238A A allele than those with mild/asymptomatic infection (P = 0.006). Furthermore, T-1031C C allele (P < 0.001) and G-238A A allele (P < 0.001) were significantly associated with decreased risk of death. Multiple haplotypes were significantly associated with decreased risk of SFTS hospital admission (SNP1-2, CC; SNP1-3, CCC; SNP1-4, CCCG; SNP1-5, CCCGA; SNP2-4, CCGA; SNP3-5, CGA; SNP4-5, GA) and death (SNP1-2, CA; SNP1-3, CAG; SNP1-4, CACG; SNP1-5, CACGG; SNP2-3, AC; SNP2-4, ACG; SNP2-5, ACGG) after correction for multiple comparisons. By using the ELISA assay, we observed that TNF-α concentration of hospitalized patients was significantly increased in acute phase than in convalescent phase (P < 0.001). Elevated TNF-α concentration was also revealed from fatal patients (P < 0.001). The -238A allele was associated with decreased serum TNF-α levels in SFTS patients in acute phase (P = 0.01). Our findings suggest that polymorphisms in TNF-α gene may play a role in mediating the risk to disease severity of SFTS in Chinese Han population.


Assuntos
Infecções por Bunyaviridae/genética , Haplótipos/genética , Polimorfismo de Nucleotídeo Único/genética , Regiões Promotoras Genéticas/genética , Trombocitopenia/genética , Fator de Necrose Tumoral alfa/genética , Idoso , Alelos , Povo Asiático/genética , Povo Asiático/estatística & dados numéricos , Infecções por Bunyaviridae/sangue , Infecções por Bunyaviridae/virologia , Feminino , Estudos de Associação Genética , Humanos , Masculino , Pessoa de Meia-Idade , Risco , Trombocitopenia/sangue , Trombocitopenia/virologia
12.
PLoS Negl Trop Dis ; 12(5): e0006508, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29813068

RESUMO

Oropouche Virus is the etiological agent of an arbovirus febrile disease that affects thousands of people and is widespread throughout Central and South American countries. Although isolated in 1950's, still there is scarce information regarding the virus biology and its prevalence is likely underestimated. In order to identify and elucidate interactions with host cells factors and increase the understanding about the Oropouche Virus biology, we performed microRNA (miRNA) and target genes screening in human hepatocarcinoma cell line HuH-7. Cellular miRNAs are short non-coding RNAs that regulates gene expression post-transcriptionally and play key roles in several steps of viral infections. The large scale RT-qPCR based screening found 13 differentially expressed miRNAs in Oropouche infected cells. Further validation confirmed that miR-217 and miR-576-3p were 5.5 fold up-regulated at early stages of virus infection (6 hours post-infection). Using bioinformatics and pathway enrichment analysis, we predicted the cellular targets genes for miR-217 and miR-576-3p. Differential expression analysis of RNA from 95 selected targets revealed genes involved in innate immunity modulation, viral release and neurological disorder outcomes. Further analysis revealed the gene of decapping protein 2 (DCP2), a previous known restriction factor for bunyaviruses transcription, as a miR-217 candidate target that is progressively down-regulated during Oropouche infection. Our analysis also showed that activators genes involved in innate immune response through IFN-ß pathway, as STING (Stimulator of Interferon Genes) and TRAF3 (TNF-Receptor Associated Factor 3), were down-regulated as the infection progress. Inhibition of miR-217 or miR-576-3p restricts OROV replication, decreasing viral RNA (up to 8.3 fold) and virus titer (3 fold). Finally, we showed that virus escape IFN-ß mediated immune response increasing the levels of cellular miR-576-3p resulting in a decreasing of its partners STING and TRAF3. We concluded stating that the present study, the first for a Peribunyaviridae member, gives insights in its prospective pathways that could help to understand virus biology, interactions with host cells and pathogenesis, suggesting that the virus escapes the antiviral cellular pathways increasing the expression of cognates miRNAs.


Assuntos
Infecções por Bunyaviridae/metabolismo , MicroRNAs/metabolismo , Orthobunyavirus/fisiologia , Provírus/genética , Infecções por Bunyaviridae/genética , Infecções por Bunyaviridae/virologia , Linhagem Celular , Biologia Computacional , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , MicroRNAs/genética , Orthobunyavirus/genética , Provírus/fisiologia , Replicação Viral
13.
Acta Trop ; 178: 61-67, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29111139

RESUMO

Schmallenberg virus (SBV) is a recently identified member of the genus Orthobunyavirus of the family Bunyaviridae. It is an arbovirus transmitted by different members of Culicoides spp of biting midges. The virus is more recognized for its effect on reproductive disorders in ruminants characterised by abortion, stillbirth and birth of congenitally defective newborns with hydranencephaly-arthrogryposis syndrome. The current study was undertaken with the objectives of exploring the presence of SBV exposure and identification of factors affecting its distribution among dairy cattle in Ethiopia. A cross-sectional study was conducted on 1379 dairy cattle sampled from 149 dairy herds in central, southern and western Ethiopia during September 2011 to May 2012. Serum samples were examined using competitive enzyme linked immunosorbent assay (cELISA). Data on hypothesised risk factors were collected from farm records where available and semi-structured questionnaire-based interview. The apparent seroprevalence of exposure to SBV was 56.6% (95% confidence interval (CI): 53.9-59.3). True prevalence adjusted for sensitivity and specificity of the cELISA kit used was 58.3% (95% CI 55.7-60.9). Among the sampled herds, 82.6% (95% CI: 75.5-88.3) had at least one seropositive animal. Seropositive cattle were found in all of the 15 conurbations studied. Adult dairy cows [odds ratio (OR)=1.6] were more commonly affected than young heifers. Dairy cattle kept in commercial (OR=1.6) and breeding farms (OR=3.5) and Midland agroecology (OR=2.5) showed statistically significant seroconversion than cattle kept under small-holder dairy farms and Highland agroecology respectively (p<0.05). Reproductive disorders including abortion, retention of the fetal membranes, and metritis were associated with serostatus of SBV. In conclusion, the seroprevalence of SBV is high and widely distributed in the studied parts of Ethiopia. This being the first study of its kind on SBV in Ethiopia, further longitudinal studies on isolation of the virus and its impact on reproductive disorders are recommended.


Assuntos
Infecções por Bunyaviridae/genética , Doenças dos Bovinos/genética , Ceratopogonidae/virologia , Orthobunyavirus/genética , Orthobunyavirus/isolamento & purificação , Animais , Infecções por Bunyaviridae/epidemiologia , Bovinos , Doenças dos Bovinos/epidemiologia , Estudos Transversais , Etiópia/epidemiologia , Geografia , Estudos Soroepidemiológicos
14.
Indian J Med Res ; 145(5): 601-610, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28948950

RESUMO

Newly emerging and re-emerging viral infections are of major public health concern. Bunyaviridae family of viruses comprises a large group of animal viruses. Clinical symptoms exhibited by persons infected by viruses belonging to this family vary from mild-to-severe diseases i.e., febrile illness, encephalitis, haemorrhagic fever and acute respiratory illness. Several arthropods-borne viruses have been discovered and classified at serological level in India in the past. Some of these are highly pathogenic as the recent emergence and spread of Crimean-Congo haemorrhagic fever virus and presence of antibodies against Hantavirus in humans in India have provided evidences that it may become one of the emerging diseases in this country. For many of the discovered viruses, we still need to study their relevance to human and animal health. Chittoor virus, a variant of Batai virus; Ganjam virus, an Asian variant of Nairobi sheep disease virus; tick-borne viruses such as Bhanja, Palma and mosquito-borne viruses such as Sathuperi, Thimiri, Umbre and Ingwavuma viruses have been identified as the members of this family. As Bunyaviruses are three segmented RNA viruses, they can reassort the segments into genetically distinct viruses in target cells. This ability is believed to play a major role in evolution, pathogenesis and epidemiology of the viruses. Here, we provide a comprehensive overview of discovery, emergence and distribution of Bunyaviruses in India.


Assuntos
Infecções por Bunyaviridae/virologia , Febre Hemorrágica da Crimeia/virologia , Orthobunyavirus/patogenicidade , Animais , Infecções por Bunyaviridae/epidemiologia , Infecções por Bunyaviridae/genética , Vírus da Febre Hemorrágica da Crimeia-Congo/patogenicidade , Febre Hemorrágica da Crimeia/epidemiologia , Humanos , Índia/epidemiologia , Orthobunyavirus/classificação , Filogenia , RNA Viral/genética , Ovinos/virologia
15.
J Biol Chem ; 292(40): 16722-16733, 2017 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-28848048

RESUMO

Heartland virus (HRTV) is a pathogenic phlebovirus related to the severe fever with thrombocytopenia syndrome virus (SFTSV), another phlebovirus causing life-threatening disease in humans. Previous findings have suggested that SFTSV can antagonize the host interferon (IFN) system via viral nonstructural protein (NSs)-mediated sequestration of antiviral signaling proteins into NSs-induced inclusion bodies. However, whether and how HRTV counteracts the host innate immunity is unknown. Here, we report that HRTV NSs (HNSs) also antagonizes IFN and cytokine induction and bolsters viral replication, although no noticeable inclusion body formation was observed in HNSs-expressing cells. Furthermore, HNSs inhibited the virus-triggered activation of IFN-ß promoter by specifically targeting the IFN-stimulated response element but not the NF-κB response element. Consistently, HNSs blocked the phosphorylation and nuclear translocation of IFN regulatory factor 3 (IRF3, an IFN-stimulated response element-activating transcription factor). Reporter gene assays next showed that HNSs blockades the antiviral signaling mediated by RIG-I-like receptors likely at the level of TANK-binding kinase 1 (TBK1). Indeed, HNSs strongly interacts with TBK1 as indicated by confocal microscopy and pulldown analyses, and we also noted that the scaffold dimerization domain of TBK1 is required for the TBK1-HNSs interaction. Finally, pulldown assays demonstrated that HNSs expression dose-dependently diminishes a TBK1-IRF3 interaction, further explaining the mechanism for HNSs function. Collectively, these data suggest that HNSs, an antagonist of host innate immunity, interacts with TBK1 and thereby hinders the association of TBK1 with its substrate IRF3, thus blocking IRF3 activation and transcriptional induction of the cellular antiviral responses.


Assuntos
Infecções por Bunyaviridae/imunologia , Imunidade Inata , Fator Regulador 3 de Interferon/imunologia , Interferon beta/imunologia , Phlebovirus/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Transdução de Sinais/imunologia , Proteínas não Estruturais Virais/imunologia , Animais , Infecções por Bunyaviridae/genética , Chlorocebus aethiops , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Fator Regulador 3 de Interferon/genética , Interferon beta/genética , Phlebovirus/genética , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais/genética , Células Vero , Proteínas não Estruturais Virais/genética
16.
PLoS One ; 12(2): e0172744, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28234991

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease caused by a novel bunyavirus (SFTS virus, SFTSV). At present there is still no specific antiviral treatment for SFTSV; To understand which cells support SFTSV life cycle and whether SFTSV infection activates host innate immunity, four different cell lines (Vero, Hela, Huh7.5.1, and Huh7.0) were infected with SFTSV. Intracellular/extracellular viral RNA and expression of IFNα, and IFNß were detected by real-time RT- PCR following infection. To confirm the role of non-structural protein (NSs) of SFTSV in exogenous IFNα-induced Jak/STAT signaling, p-STAT1 (Western Blot), ISRE activity (Luciferase assay) and ISG expression (real-time PCR) were examined following IFNα stimulation in the presence or absence of over-expression of NSs in Hela cells. Our study showed that all the four cell lines supported SFTSV life cycle and SFTSV activated host innate immunity to produce type I IFNs in Hela cells but not in Huh7.0, Huh7.5.1 or Vero cells. NSs inhibited exogenous IFNα-induced Jak/STAT signaling as shown by decreased p-STAT1 level, suppressed ISRE activity and down-regulated ISG expression. Suppression of the exogenous Type I IFN-induced Jak/STAT signaling by NSs might be one of the mechanisms of SFTSV to evade host immune surveillance.


Assuntos
Infecções por Bunyaviridae/genética , Interferon Tipo I/genética , Phlebovirus/genética , Proteínas não Estruturais Virais/genética , Animais , Infecções por Bunyaviridae/virologia , Chlorocebus aethiops , Células HeLa , Interações Hospedeiro-Parasita/genética , Humanos , Imunidade Inata/genética , Interferon Tipo I/antagonistas & inibidores , Phlebovirus/patogenicidade , Transdução de Sinais/genética , Células Vero
17.
J Virol ; 91(3)2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27881648

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne disease endemic in parts of Asia. The etiologic agent, SFTS virus (SFTSV; family Bunyaviridae, genus Phlebovirus) has caused significant morbidity and mortality in China, South Korea, and Japan, with key features of disease being intense fever, thrombocytopenia, and leukopenia. Case fatality rates are estimated to be in the 30% range, and no antivirals or vaccines are approved for use for treatment and prevention of SFTS. There is evidence that in human cells, SFTSV sequesters STAT proteins in replication complexes, thereby inhibiting type I interferon signaling. Here, we demonstrate that hamsters devoid of functional STAT2 are highly susceptible to as few as 10 PFU of SFTSV, with animals generally succumbing within 5 to 6 days after subcutaneous challenge. The disease included marked thrombocytopenia and inflammatory disease characteristic of the condition in humans. Infectious virus titers were present in the blood and most tissues 3 days after virus challenge, and severe inflammatory lesions were found in the spleen and liver samples of SFTSV-infected hamsters. We also show that SFTSV infection in STAT2 knockout (KO) hamsters is responsive to favipiravir treatment, which protected all animals from lethal disease and reduced serum and tissue viral loads by 3 to 6 orders of magnitude. Taken together, our results provide additional insights into the pathogenesis of SFTSV infection and support the use of the newly described STAT2 KO hamster model for evaluation of promising antiviral therapies. IMPORTANCE: Severe fever with thrombocytopenia syndrome (SFTS) is an emerging viral disease for which there are currently no therapeutic options or available vaccines. The causative agent, SFTS virus (SFTSV), is present in China, South Korea, and Japan, and infections requiring medical attention result in death in as many as 30% of the cases. Here, we describe a novel model of SFTS in hamsters genetically engineered to be deficient in a protein that helps protect humans and animals against viral infections. These hamsters were found to be susceptible to SFTSV and share disease features associated with the disease in humans. Importantly, we also show that SFTSV infection in hamsters can be effectively treated with a broad-spectrum antiviral drug approved for use in Japan. Our findings suggest that the new SFTS model will be an excellent resource to better understand SFTSV infection and disease as well as a valuable tool for evaluating promising antiviral drugs.


Assuntos
Infecções por Bunyaviridae/virologia , Modelos Biológicos , Phlebovirus/fisiologia , Amidas/farmacologia , Animais , Animais Geneticamente Modificados , Antivirais/farmacologia , Infecções por Bunyaviridae/tratamento farmacológico , Infecções por Bunyaviridae/genética , Infecções por Bunyaviridae/mortalidade , Cricetinae , Modelos Animais de Doenças , Suscetibilidade a Doenças , Genótipo , Humanos , Fenótipo , Pirazinas/farmacologia , Fator de Transcrição STAT2/genética
18.
PLoS Negl Trop Dis ; 10(10): e0005076, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27760141

RESUMO

OBJECTIVE: Severe fever with thrombocytopenia syndrome (SFTS) is an emerging hemorrhagic fever caused by a tick-borne bunyavirus (SFTSV) in East Asian countries. The role of human leukocyte antigen (HLA) in resistance and susceptibility to SFTSV is not known. We investigated the correlation of HLA locus A, B and DRB1 alleles with the occurrence of SFTS. METHODS: A total of 84 confirmed SFTS patients (patient group) and 501 unrelated non-SFTS patients (healthy individuals as control group) from Shandong Province were genotyped by PCR-sequence specific oligonucleotide probe (PCR-SSOP) for HLA-A, B and DRB1 loci.Allele frequency was calculated and compared using χ2 test or the Fisher's exact test. A corrected P value was calculated with a bonferronis correction. Odds Ratio (OR) and 95% confidence intervals (CI) were calculated by Woolf's method. RESULTS: A total of 11 HLA-A, 23 HLA-B and 12 HLA-DRB1 alleles were identified in the patient group, whereas 15 HLA-A, 30 HLA-B and 13 HLA-DRB1 alleles were detected in the control group. The frequencies of A*30 and B*13 in the SFTS patient group were lower than that in the control group (P = 0.0341 and 0.0085, Pc = 0.5115 and 0.252). The ORs of A*30 and B*13 in the SFTS patient group were 0.54 and 0.49, respectively. The frequency of two-locus haplotype A*30-B*13 was lower in the patient group than in the control group(5.59% versus 12.27%, P = 0.037,OR = 0.41, 95%CI = 0.18-0.96) without significance(Pc>0.05). A*30-B*13-DRB1*07 and A*02-B*15-DRB1*04 had strong associations with SFTS resistance and susceptibility respectively (Pc = 0.0412 and 0.0001,OR = 0.43 and 5.07). CONCLUSION: The host HLA class I polymorphism might play an important role with the occurrence of SFTS. Negative associations were observed with HLA-A*30, HLA-B*13 and Haplotype A*30-B*13, although the associations were not statistically significant. A*30-B*13-DRB1*07 had negative correlation with the occurrence of SFTS; in contrast, haplotype A*02-B*15-DRB1*04 was positively correlated with SFTS.


Assuntos
Infecções por Bunyaviridae/genética , Frequência do Gene , Predisposição Genética para Doença , Antígenos HLA-A/genética , Antígenos HLA-B/genética , Cadeias HLA-DRB1/genética , Phlebovirus , Adulto , Idoso , Idoso de 80 Anos ou mais , Infecções por Bunyaviridae/sangue , Infecções por Bunyaviridae/virologia , Feminino , Genótipo , Haplótipos , Humanos , Masculino , Pessoa de Meia-Idade , Razão de Chances , Phlebovirus/isolamento & purificação , Reação em Cadeia da Polimerase , Polimorfismo Genético
19.
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
20.
Oncotarget ; 7(22): 33340-9, 2016 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-27147565

RESUMO

BACKGROUND: Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease caused by a novel bunyavirus named SFTS virus (SFTSV). We hypothesize that host genetic variations may contribute to susceptibility to SFTS. RESULTS: Compared with the rs1800818 AA genotype, AG + GG genotypes were significantly associated with increased susceptibility to SFTS (odds ratio, 1.66, 95% confidence interval = 1.28-2.16; P < 0.001). By using the ELISA assay, we observed that PDGF-BB concentration was significantly reduced in acute phase of patients than in the controls (P < 0.001) and recovered patients at 6 month (P = 0.007) and 12 month (P = 0.003). A persistently reduced PDGF-BB was also revealed from the SFTSV-infected C57BL/6J mice (P < 0.001). The rs1800818 G allele was associated with decreased serum PDGF-BB levels in SFTS patients at their early infection (P = 0.015). In accordance, the relative mRNA levels of the at-risk G allele of 1800818 were lower than those of the A allele in heterozygous cell from acute phase of SFTS patients. PDGF-B rs1800818 conferred no susceptibility to severe or fatal outcome in SFTS patients. MATERIALS AND METHODS: An initially small-scale case-control association study guided the selection of platelet derived growth factor-B (PDGF-B) rs1800818 in 1020 SFTS patients and 1353 controls. Functional analyses were conducted to verify the biological significance of rs1800818 polymorphism. CONCLUSIONS: Our findings suggest that the PDGF-B rs1800818 polymorphism might play a role in mediating the susceptibility to SFTS.


Assuntos
Infecções por Bunyaviridae/genética , Febre/genética , Polimorfismo de Nucleotídeo Único , Proteínas Proto-Oncogênicas c-sis/genética , Trombocitopenia/genética , Animais , Povo Asiático/genética , Becaplermina , Infecções por Bunyaviridae/sangue , Infecções por Bunyaviridae/etnologia , Infecções por Bunyaviridae/virologia , Estudos de Casos e Controles , China/epidemiologia , Modelos Animais de Doenças , Feminino , Febre/sangue , Febre/etnologia , Febre/virologia , Frequência do Gene , Estudos de Associação Genética , Predisposição Genética para Doença , Heterozigoto , Homozigoto , Humanos , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Fenótipo , Proteínas Proto-Oncogênicas c-sis/sangue , Fatores de Risco , Índice de Gravidade de Doença , Síndrome , Trombocitopenia/sangue , Trombocitopenia/etnologia , Trombocitopenia/virologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...