Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
1.
J Med Virol ; 96(8): e29854, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39135475

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) has a high mortality rate compared to other infectious diseases. SFTS is particularly associated with a high risk of mortality in immunocompromised individuals, while most patients who die of SFTS exhibit symptoms of severe encephalitis before death. However, the region of brain damage and mechanisms by which the SFTS virus (SFTSV) causes encephalitis remains unknown. Here, we revealed that SFTSV infects the brainstem and spinal cord, which are regions of the brain associated with respiratory function, and motor nerves in IFNAR1-/- mice. Further, we show that A1-reactive astrocytes are activated, causing nerve cell death, in infected mice. Primary astrocytes of SFTSV-infected IFNAR1-/- mice also induced neuronal cell death through the activation of A1-reactive astrocytes. Herein, we showed that SFTSV induces fatal neuroinflammation in the brain regions important for respiratory function and motor nerve, which may underlie mortality in SFTS patients. This study provides new insights for the treatment of SFTS, for which there is currently no therapeutic approach.


Assuntos
Astrócitos , Infecções por Bunyaviridae , Camundongos Knockout , Phlebovirus , Receptor de Interferon alfa e beta , Animais , Astrócitos/virologia , Astrócitos/patologia , Camundongos , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/deficiência , Phlebovirus/genética , Phlebovirus/fisiologia , Phlebovirus/patogenicidade , Infecções por Bunyaviridae/virologia , Infecções por Bunyaviridae/patologia , Infecções por Bunyaviridae/imunologia , Encéfalo/virologia , Encéfalo/patologia , Encéfalo/imunologia , Medula Espinal/virologia , Medula Espinal/patologia , Modelos Animais de Doenças , Neurônios/virologia , Neurônios/patologia , Camundongos Endogâmicos C57BL , Tronco Encefálico/virologia , Tronco Encefálico/patologia , Morte Celular
2.
Arch Virol ; 169(6): 133, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38829449

RESUMO

Akabane virus (AKAV), Aino virus, Peaton virus, Sathuperi virus, and Shamonda virus are arthropod-borne viruses belonging to the order Elliovirales, family Peribunyaviridae, genus Orthobunyavirus. These viruses cause or may cause congenital malformations in ruminants, including hydranencephaly, poliomyelitis, and arthrogryposis, although their pathogenicity may vary among field cases. AKAV may cause relatively severe congenital lesions such as hydranencephaly in calves. Furthermore, strains of AKAV genogroups I and II exhibit different disease courses. Genogroup I strains predominantly cause postnatal viral encephalomyelitis, while genogroup II strains are primarily detected in cases of congenital malformation. However, the biological properties of AKAV and other orthobunyaviruses are insufficiently investigated in hosts in the field and in vitro. Here, we used an immortalized bovine brain cell line (FBBC-1) to investigate viral replication efficiency, cytopathogenicity, and host innate immune responses. AKAV genogroup II and Shamonda virus replicated to higher titers in FBBC-1 cells compared with the other viruses, and only AKAV caused cytopathic effects. These results may be associated with the severe congenital lesions in the brain caused by AKAV genogroup II. AKAV genogroup II strains replicated to higher titers in FBBC-1 cells than AKAV genogroup I strains, suggesting that genogroup II strains replicated more efficiently in fetal brain cells, accounting for the detection of the latter strains mainly in fetal infection cases. Therefore, FBBC-1 cells may serve as a valuable tool for investigating the virulence and tropism of the orthobunyaviruses for bovine neonatal brain tissues in vitro.


Assuntos
Encéfalo , Infecções por Bunyaviridae , Orthobunyavirus , Replicação Viral , Animais , Bovinos , Orthobunyavirus/patogenicidade , Orthobunyavirus/genética , Orthobunyavirus/fisiologia , Orthobunyavirus/classificação , Encéfalo/virologia , Encéfalo/patologia , Linhagem Celular , Infecções por Bunyaviridae/virologia , Infecções por Bunyaviridae/veterinária , Infecções por Bunyaviridae/patologia , Doenças dos Bovinos/virologia , Feto/virologia , Efeito Citopatogênico Viral , Imunidade Inata
3.
Vet Res Commun ; 48(1): 449-457, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37831381

RESUMO

Akabane virus (AKAV) is known as a major teratogenic agent of ruminant fetuses. In this study, we investigated the relationship between porcine abnormal deliveries and AKAV by serology, pathology, and virology investigations using specimens from 16 stillborn fetuses delivered in southern Japan between 2013 and 2015. The major clinical manifestations in stillborn fetuses were hydranencephaly, arthrogryposis, spinal curvature, and both skeletal muscle and subcutaneous edema. Histologic examination of the specimens identified atrophy of skeletal muscle fibers accompanied by adipose replacement. Nonsuppurative encephalomyelitis and decreased neuronal density in the ventral horn of the spinal cord were shown in two separate fetuses, respectively. Neutralizing antibody titers to AKAV were detected in most of the tested fetuses (13/16). The AKAV sequences detected in the affected fetuses in 2013 and 2015 were highly identical and closely related to Japanese AKAV isolates which were isolated in 2013 and sorted into genogroup I of AKAV. Immunohistochemistry visualized AKAV antigens in the neuronal cells of the central nervous system of the fetuses. These findings indicate that AKAV was involved in the birth of abnormal piglets at the affected farm. The clinical manifestations and histopathological features in the stillborn fetuses were very similar to those in ruminant neonates affected by AKAV. To avoid misdiagnosis and to evaluate the precise impact of AKAV on pig reproduction, AKAV should be considered in differential diagnoses of reproductive failures in pigs.


Assuntos
Infecções por Bunyaviridae , Orthobunyavirus , Doenças dos Suínos , Animais , Infecções por Bunyaviridae/diagnóstico , Infecções por Bunyaviridae/veterinária , Infecções por Bunyaviridae/patologia , Feto/patologia , Japão/epidemiologia , Ruminantes , Suínos , Doenças dos Suínos/diagnóstico
4.
Top Companion Anim Med ; 52: 100756, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36586577

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging zoonotic disease caused by the SFTS virus (SFTSV). SFTSV causes severe symptoms both in humans and cats. In this study, we report the clinical and pathological findings of 4 fatal cases of cats with high SFTS viremia levels. These cats showed an acute onset of fever, leukopenia, thrombocytopenia, and increased serum amyloid A and pro-inflammatory cytokine levels. A high viral copy number was detected in the blood, oral swabs, rectal swabs, conjunctiva swabs, and urine. Histopathologically, necrotizing lymphadenitis, splenitis with lymphoblastoid cell proliferation, and hemophagocytosis were observed in all 4 cats. Immunohistochemistry revealed the presence of SFTSV antigen on lymphoblastoid B cells. SFTSV-RNA was detected in systemic tissues, including the brain. The present findings provide useful information for understanding the features of fatal SFTS in cats. To elucidate the mechanisms of severe progress of SFTS cats, as well as its role as a source of human infection, further research is needed.


Assuntos
Infecções por Bunyaviridae , Doenças do Gato , Phlebovirus , Febre Grave com Síndrome de Trombocitopenia , Trombocitopenia , Animais , Gatos , Humanos , Febre Grave com Síndrome de Trombocitopenia/veterinária , Infecções por Bunyaviridae/veterinária , Infecções por Bunyaviridae/patologia , Viremia/veterinária , Phlebovirus/genética , Trombocitopenia/veterinária
5.
Int J Mol Med ; 50(3)2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35856413

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) has been acknowledged as an emerging infectious disease that is caused by the SFTS virus (SFTSV). The main clinical features of SFTS on presentation include fever, thrombocytopenia, leukocytopenia and gastrointestinal symptoms. The mortality rate is estimated to range between 5­30% in East Asia. However, SFTSV infection is increasing on an annual basis globally and is becoming a public health problem. The transmission cycle of SFTSV remains poorly understood, which is compounded by the pathogenesis of SFTS not being fully elucidated. Since the mechanism underlying the host immune response towards SFTSV is also unclear, there are no effective vaccines or specific therapeutic agents against SFTS, with supportive care being the only realistic option. Therefore, it is now crucial to understand all aspects of the host­virus interaction following SFTSV infection, including the antiviral states and viral evasion mechanisms. In the present review, recent research progress into the possible host immune responses against SFTSV was summarized, which may be useful in designing novel therapeutics against SFTS.


Assuntos
Infecções por Bunyaviridae , Phlebovirus , Febre Grave com Síndrome de Trombocitopenia , Trombocitopenia , Infecções por Bunyaviridae/tratamento farmacológico , Infecções por Bunyaviridae/patologia , Humanos , Phlebovirus/fisiologia , Trombocitopenia/patologia
6.
Microbiol Spectr ; 10(3): e0129422, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35612327

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging hemorrhagic fever acquired by tick bites. Whether mast cells (MCs), the body's first line of defense against pathogens, might influence immunity or pathogenesis during SFTS virus (SFTSV) infection remained unknown. Here, we found that SFTSV can cause MC infection and degranulation, resulting in the release of the vasoactive mediators, chymase, and tryptase, which can directly act on endothelial cells, break the tight junctions of endothelial cells and threaten the integrity of the microvascular barrier, leading to microvascular hyperpermeability in human microvascular endothelial cells. Local activation of MCs (degranulation) and MC-specific proteases-facilitated endothelial damage were observed in mouse models. When MC-specific proteases were injected subcutaneously into the back skin of mice, signs of capillary leakage were observed in a dose-dependent manner. MC-specific proteases, chymase, and tryptase were tested in the serum collected at the acute phase of SFTS patients, with the higher level significantly correlated with fatal outcomes. By performing receiver operator characteristic curve (ROC) analysis, chymase was determined as a biomarker with the area under the curve value of 0.830 (95% CI = 0.745 to 0.915) for predicting fatal outcomes in SFTS. Our findings highlight the importance of MCs in SFTSV-induced disease progression and outcome. An emerging role for MCs in the clinical prognosis and blocking MC activation as a potential drug target during SFTSV infection was proposed. IMPORTANCE We revealed a pathogenic role for MCs in response to SFTSV infection. The study also identifies potential biomarkers that could differentiate patients at risk of a fatal outcome for SFTS, as well as novel therapeutic targets for the clinical management of SFTS. These findings might shed light on an emerging role for MCs as a potential drug target during infection of other viral hemorrhagic fever diseases with similar host pathology as SFTS.


Assuntos
Infecções por Bunyaviridae , Febre Grave com Síndrome de Trombocitopenia , Animais , Biomarcadores , Infecções por Bunyaviridae/patologia , Quimases , Células Endoteliais/patologia , Mastócitos/patologia , Camundongos , Peptídeo Hidrolases/uso terapêutico , Permeabilidade , Phlebovirus , Triptases/uso terapêutico
7.
PLoS Negl Trop Dis ; 14(11): e0008863, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33232320

RESUMO

Crimean-Congo Hemorrhagic Fever Virus (CCHFV) and Hazara virus (HAZV) belong to the same viral serotype and family. HAZV has lately been used as a model system and surrogate to CCHFV. However, virus-host cell interaction and level of pathogenicity for these viruses are not well investigated nor compared. In this study, we compared HAZV and CCHFV infection of human polarized epithelial cells to shed light on similarities and differences in virus-host cell interaction between these two viruses. We investigated the pattern of infection of CCHFV and HAZV in fully polarized human cells, the Caco-2 cell line. Polarization of Caco-2 cells lead to difference in expression level and pattern of proteins between the apical and the basolateral membranes. We found that CCHFV virus, in contrast to HAZV, is more likely infecting polarized cells basolaterally. In addition, we found that cytokines/pro-inflammatory factors or other viral factors secreted from CCHFV infected moDC cells enhance the entry of CCHFV contrary to HAZV. We have shown that CCHFV and HAZV early in infection use different strategies for entry. The data presented in this study also highlight the important role of cytokines in CCHFV-host cell interaction.


Assuntos
Células Epiteliais/virologia , Vírus da Febre Hemorrágica da Crimeia-Congo/crescimento & desenvolvimento , Nairovirus/crescimento & desenvolvimento , Ligação Viral , Internalização do Vírus , Infecções por Bunyaviridae/patologia , Células CACO-2 , Linhagem Celular Tumoral , Febre Hemorrágica da Crimeia/patologia , Humanos , Replicação Viral
8.
PLoS Negl Trop Dis ; 14(7): e0008453, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32730341

RESUMO

BACKGROUND: Severe fever with thrombocytopenia syndrome virus (SFTSV) is a novel tick-borne phlebovirus, which is listed in the most dangerous pathogens by the World Health Organization, and has 12-30% fatality rates. SFTSV antibodies were reported in minks that experienced abortion or reproductive failure. The aim of this study was to determine whether SFTSV infection causes an adverse pregnancy outcome in the fetus using a pregnant mouse model. METHODOLOGY/PRINCIPAL FINDINGS: We found SFTSV in the fetus after infection in pregnant mice, and some dams showed adverse pregnancy outcomes after infection with SFTSV including placental damage, fetal reabsorption, and fetal intrauterine growth restriction (IUGR). SFTSV had obvious tropism characteristics in the placenta, especially in the labyrinth. In early-gestation, pregnant mice infected with SFTSV had fetal IUGR and a high viral load in the fetus. The virus widely spread in infected fetuses, including the hindbrain, thymus, heart, spinal cord, and liver. CONCLUSIONS: Our study demonstrated that SFTSV was vertically transmitted to the fetus through the placental barrier of immunocompetent mice, and resulted in adverse pregnancy outcomes.


Assuntos
Infecções por Bunyaviridae/patologia , Feto/anormalidades , Feto/virologia , Phlebovirus , Alquilantes/toxicidade , Animais , Feminino , Hospedeiro Imunocomprometido , Camundongos , Camundongos Endogâmicos C57BL , Mitomicina/toxicidade , Gravidez
9.
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
10.
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
11.
Sci Rep ; 9(1): 17924, 2019 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-31784575

RESUMO

In the present study, patients with acute OROV fever were classified as early seroconverters (IgM/IgG positive at baseline) or late seroconverters (IgM/IgG negative at baseline) and the timeline kinetics of the production of chemokines and cytokines were assessed at 1-3, 4-7, 8-10 and ≥11 days after patients have reported the first symptoms. Regardless immunoglobulin profile, all OROV fever patients presented higher levels of CXCL8, and IFN-α and lower levels of TNF and IL-10 at baseline as compared to healthy donors (HD). Lower levels of CCL2, CXCL10, and IFN-γ and higher levels of CCL2, CXCL10, IL-6, and IL-17A were detected in early and late seroconverters, respectively, as compared to HD. While early seroconverters presented the increasing levels of CCL2 along the timeline, late seroconverters displayed decreasing levels of CCL2, CXCL10, and IL-6 following days of disease onset. Noteworthy was that IFN-α was revealed as universal biomarker of human OROV fever, while CXCL8 & IL-5 and CXCL10 & IL-17 were consistently observed in early and late seroconverters, respectively. Thus, our results suggest that the production of IFN-α, CXCL10, and IL-17 precede the seroconversion bringing novel insights on the immunological events triggered by the OROV disease.


Assuntos
Infecções por Bunyaviridae/sangue , Interferon-alfa/sangue , Soroconversão , Biomarcadores/sangue , Infecções por Bunyaviridae/imunologia , Infecções por Bunyaviridae/patologia , Quimiocinas/sangue , Humanos , Interferon gama/sangue , Interleucina-27/sangue , Interleucina-6/sangue , Testes Sorológicos/métodos , Testes Sorológicos/normas , Tempo
12.
Vet Microbiol ; 236: 108346, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31500732

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) is a recently discovered emerging infectious disease. A zoonotic disease with a high fatality rate in human beings, clinical information on SFTS virus (SFTSV) infection in animals is important. Since 2017, we have diagnosed 24 client-owned cats living in western Japan with SFTS, by genetic and serological testing. In this study, we characterized the clinical features of SFTS in cats and their associated risk factors, by evaluating the clinical parameters retrospectively. A phylogenetic analysis on SFTSV was also conducted. There were no obvious tendencies in age or sex, outdoor cats were commonly at risk of SFTSV infection. All infected cats showed acute onset of clinical signs including anorexia and lethargy, while 68.2% of the cats showed fever and 41.7% showed vomiting. The case fatality rate was 62.5%. Thrombocytopenia, leukopenia, and elevated serum total bilirubin, serum amyloid A, and creatinine phosphokinase concentration were the characteristic findings in the first clinical blood examination. Phylogenic analysis revealed that regional clustered viruses infect both humans and cats. For pet owners and animal hospitals, SFTS in small animals could be an important public health issue.


Assuntos
Infecções por Bunyaviridae/veterinária , Doenças do Gato/virologia , Doenças Transmissíveis Emergentes/veterinária , Phlebovirus/genética , Animais , Infecções por Bunyaviridae/epidemiologia , Infecções por Bunyaviridae/patologia , Infecções por Bunyaviridae/virologia , Doenças do Gato/epidemiologia , Doenças do Gato/patologia , Gatos , Doenças Transmissíveis Emergentes/epidemiologia , Doenças Transmissíveis Emergentes/patologia , Doenças Transmissíveis Emergentes/virologia , Feminino , Japão/epidemiologia , Masculino , Filogenia , Estudos Retrospectivos , Carga Viral
13.
Microb Pathog ; 135: 103627, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31326560

RESUMO

The Guama virus (GMAV) is a member of Peribunyaviridae family, Orthobunyavirus genus. Several strains of the virus were isolated in South and Central Americas from several hosts, such as humans, wild animals, including nonhuman primates, wild rodents and mosquitoes as well as mice used as sentinels. The virus is able to cause febrile disease in humans. Here we describe for the first time pathologic and biochemical findings in golden hamsters (Mesocricetus auratus) infected with the prototype GMAV. Blood and organs of infected and control animals were collected every 24 h after infection from the 1st to the 7th day post infection (dpi) and at 21 dpi when experiment was ended. The tissues were processed for histopathology and immunohistochemistry. The blood and serum were used to determine viremia and biochemical markers plus to detect anti-GMAV antibodies. The viremia was early detected already on the 1st dpi and it was no longer detected on the 3rd dpi. Total anti-GMAV antibodies were detected from the 6th dpi. Hepatic markers as ALT of infected animals were increased and showed statistically significant difference in comparison with control animals, indicating damage of the liver; indeed the liver was the most affected organ, but other organs presented lesions and positive GMAV immunostaining as brain, lung, liver, spleen, and kidney. Our findings indicate that golden hamsters are a good animal model for experimental infection of the GMAV.


Assuntos
Infecções por Bunyaviridae/virologia , Modelos Animais de Doenças , Orthobunyavirus/patogenicidade , Animais , Anticorpos Antivirais/sangue , Infecções por Bunyaviridae/sangue , Infecções por Bunyaviridae/patologia , Rim/patologia , Fígado/patologia , Masculino , Mesocricetus , Baço/patologia , Viremia
14.
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
15.
Transbound Emerg Dis ; 66(3): 1126-1131, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30864252

RESUMO

The insect-transmitted Shuni virus (SHUV) belongs to the Simbu serogroup of orthobunyaviruses and it is known to induce abortions, stillbirths and severe congenital malformations in ruminants and may cause neurological signs in infected horses. Here, SHUV was detected in brain samples of two Israeli cattle, which suffered from severe neurological signs that led to the deaths of the animals. During histopathological examination of the first case, a 5-month-old calf, small perivascular cuffs, composed mainly of neutrophils with few lymphocytes were observed in the brain stem and cerebrum. Similar infiltrates were also found to a lesser extent in the cerebellar meninges leading to the diagnosis of acute-subacute meningoencephalitis. The histological examination of the brainstem from the second case, a 16-month-old heifer, revealed perivascular infiltration composed of equal numbers of macrophages and neutrophils associated with cerebral and meningeal haemorrhages. In this case encephalitis was diagnosed. Viral RNA was extracted from brain samples of both cattle that suffered from severe neurological signs and was subsequently tested by a polymerase chain reaction PCR assay specific for Simbu serogroup viruses and found positive. The presence of SHUV was subsequently confirmed by the isolation of the virus from one sample and sequence analysis of both brain samples. The comparison of the complete sequences of the coding regions of all three genome segments from both cases revealed a close relationship to Shuni viruses detected in tissue samples of aborted or malformed calves or lambs born during the last years in Israel.


Assuntos
Infecções por Bunyaviridae/veterinária , Doenças dos Bovinos/diagnóstico , Orthobunyavirus/isolamento & purificação , Animais , Infecções por Bunyaviridae/diagnóstico , Infecções por Bunyaviridae/patologia , Infecções por Bunyaviridae/virologia , Bovinos , Doenças dos Bovinos/patologia , Doenças dos Bovinos/virologia , Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , Diagnóstico , Feminino , Israel , Masculino , Fases de Leitura Aberta/genética , Orthobunyavirus/genética , Filogenia , Reação em Cadeia da Polimerase/veterinária , RNA Viral/análise
16.
Nat Microbiol ; 4(3): 429-437, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30617349

RESUMO

Severe fever with thrombocytopenia syndrome phlebovirus (SFTSV), listed in the World Health Organization Prioritized Pathogens, is an emerging phlebovirus with a high fatality1-4. Owing to the lack of therapies and vaccines5,6, there is a pressing need to understand SFTSV pathogenesis. SFSTV non-structural protein (NSs) has been shown to block type I interferon induction7-11 and facilitate disease progression12,13. Here, we report that SFTSV-NSs targets the tumour progression locus 2 (TPL2)-A20-binding inhibitor of NF-κB activation 2 (ABIN2)-p105 complex to induce the expression of interleukin-10 (IL-10) for viral pathogenesis. Using a combination of reverse genetics, a TPL2 kinase inhibitor and Tpl2-/- mice showed that NSs interacted with ABIN2 and promoted TPL2 complex formation and signalling activity, resulting in the marked upregulation of Il10 expression. Whereas SFTSV infection of wild-type mice led to rapid weight loss and death, Tpl2-/- mice or Il10-/- mice survived an infection. Furthermore, SFTSV-NSs P102A and SFTSV-NSs K211R that lost the ability to induce TPL2 signalling and IL-10 production showed drastically reduced pathogenesis. Remarkably, the exogenous administration of recombinant IL-10 effectively rescued the attenuated pathogenic activity of SFTSV-NSs P102A, resulting in a lethal infection. Our study demonstrates that SFTSV-NSs targets the TPL2 signalling pathway to induce immune-suppressive IL-10 cytokine production as a means to dampen the host defence and promote viral pathogenesis.


Assuntos
Interações Hospedeiro-Patógeno , MAP Quinase Quinase Quinases/metabolismo , Phlebovirus/patogenicidade , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Proteínas não Estruturais Virais/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Infecções por Bunyaviridae/imunologia , Infecções por Bunyaviridae/patologia , Feminino , Células HEK293 , Células HeLa , Humanos , Interleucina-10/administração & dosagem , Interleucina-10/genética , MAP Quinase Quinase Quinases/imunologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Phlebovirus/efeitos dos fármacos , Proteínas Proto-Oncogênicas/imunologia , Células RAW 264.7 , Genética Reversa
17.
J Vet Med Sci ; 81(2): 204-212, 2019 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-30541984

RESUMO

Schmallenberg virus (SBV), discovered in 2011 in Germany, is associated with clinical manifestations of fever, diarrhea, reduced milk yield, abortions and congenital malformations in ruminants. Despite many studies performed for SBV, there is no detailed research on in vitro apoptotic effect of SBV. This study is aimed to determine apoptosis pathways and role of pro-apoptotic and anti-apoptotic molecules in Vero cells infected with SBV. The study results showed that SBV induced apoptosis via both extrinsic and intrinsic pathways by activating both caspase-8 and caspase-9, respectively. Expression analyses of pro-apoptotic (Bax, Bak and Puma) and anti-apoptotic (Bcl-2 and Bcl-XL) genes revealed that SBV-induced apoptosis causes upregulation of pro-apoptotic genes, dominantly via Puma gene, whereas Bcl-2 and Bcl-XL genes were downregulated. In conclusion, this is the first detailed report about SBV induced apoptosis in the Vero cells via both extrinsic and intrinsic cascades and apoptosis induction is seem to be regulated by Puma.


Assuntos
Apoptose , Infecções por Bunyaviridae/veterinária , Orthobunyavirus , Animais , Infecções por Bunyaviridae/patologia , Infecções por Bunyaviridae/virologia , Caspases/metabolismo , Chlorocebus aethiops , Fragmentação do DNA , Citometria de Fluxo , Genes Virais/genética , Orthobunyavirus/genética , Fosfatidilserinas/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Células Vero/virologia
18.
Clin Infect Dis ; 68(1): 143-145, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29893806

RESUMO

Keystone virus, a California-serogroup orthobunyavirus, was first isolated in 1964 from mosquitoes in Keystone, Florida. There were no prior reports of isolation from humans, despite studies suggesting that ~20% of persons living in the region are seropositive. We report virus isolation from a Florida teenager with a rash and fever.


Assuntos
Infecções por Bunyaviridae/diagnóstico , Infecções por Bunyaviridae/patologia , Exantema/etiologia , Febre/etiologia , Orthobunyavirus/isolamento & purificação , Adolescente , Infecções por Bunyaviridae/virologia , Florida , Humanos , Masculino , Vírus de Plantas
19.
Am J Trop Med Hyg ; 99(6): 1466-1468, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30277197

RESUMO

We describe the case of a patient with severe fever with thrombocytopenia syndrome (SFTS) complicated by SFTS-associated encephalopathy who was successfully treated with 4-day plasma exchange followed by two-time convalescent plasma therapy. During plasma exchange, the plasma cytokines interferon-α and inducible protein-10 gradually decreased without change of plasma viral load. However, plasma viral load gradually decreased after convalescent plasma therapy. This case provides important insights for understanding the mechanisms of experimental therapy in severely affected SFTS patients.


Assuntos
Anticorpos Antivirais/sangue , Encefalopatias/terapia , Infecções por Bunyaviridae/terapia , Phlebovirus/patogenicidade , Troca Plasmática/métodos , Terapias em Estudo/métodos , Idoso , Anticorpos Antivirais/biossíntese , Antivirais/uso terapêutico , Encefalopatias/imunologia , Encefalopatias/patologia , Encefalopatias/virologia , Infecções por Bunyaviridae/imunologia , Infecções por Bunyaviridae/patologia , Infecções por Bunyaviridae/virologia , Quimiocina CXCL10/sangue , Quimiocina CXCL10/imunologia , Convalescença , Estado Terminal , Humanos , Interferon-alfa/sangue , Interferon-alfa/imunologia , Masculino , Phlebovirus/imunologia , República da Coreia , Ribavirina/uso terapêutico , Doadores de Tecidos , Resultado do Tratamento , Carga Viral
20.
J Vet Diagn Invest ; 30(6): 855-861, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30204057

RESUMO

Peaton virus (PEAV; family Peribunyaviridae, genus Orthobunyavirus) appears to be capable of producing congenital malformations in ruminants; however, its pathogenicity remains unknown given its relatively low incidence. We evaluated the relationship between congenital abnormalities of calves and PEAV infection by serologic, epidemiologic, pathologic, and virologic investigations using specimens from 31 malformed calves in the years 1996-2016 in Japan. Antibody testing was carried out for known teratogenic viruses, including Akabane, Aino, Chuzan, and bovine viral diarrhea viruses, in the precolostral sera of these abnormal calves, but all results were negative. However, all 31 malformed calves were positive for antibodies against PEAV. A PEAV-specific gene was amplified from central nervous system tissues from a stillborn calf delivered in April 2007, and its nucleotide sequence was identical with that of PEAV isolated from healthy sentinel cattle in September 2006. These findings indicate that PEAV can cause bovine congenital anomalies.


Assuntos
Infecções por Bunyaviridae/veterinária , Doenças dos Bovinos/patologia , Bovinos/anormalidades , Anormalidades Congênitas/veterinária , Orthobunyavirus/fisiologia , Animais , Anticorpos Antivirais/sangue , Infecções por Bunyaviridae/patologia , Infecções por Bunyaviridae/virologia , Doenças dos Bovinos/virologia , Anormalidades Congênitas/patologia , Anormalidades Congênitas/virologia , Japão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA