Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
PLoS Pathog ; 17(5): e1009587, 2021 05.
Article in English | MEDLINE | ID: mdl-33974679

ABSTRACT

Severe fever with thrombocytopenia syndrome virus (SFTSV) is a tick-borne emerging phlebovirus with high mortality rates of 6.0 to 30%. SFTSV infection is characterized by high fever, thrombocytopenia, leukopenia, hemorrhage and multiple organ failures. Currently, specific therapies and vaccines remain elusive. Suitable small animal models are urgently needed to elucidate the pathogenesis and evaluate the potential drug and vaccine for SFTSV infection. Previous models presented only mild or no pathogenesis of SFTS, limiting their applications in SFTSV infection. Therefore, it is an urgent need to develop a small animal model for the investigation of SFTSV pathogenesis and evaluation of therapeutics. In the current report, we developed a SFTSV infection model based on the HuPBL-NCG mice that recapitulates many pathological characteristics of SFTSV infection in humans. Virus-induced histopathological changes were identified in spleen, lung, kidney, and liver. SFTSV was colocalized with macrophages in the spleen and liver, suggesting that the macrophages in the spleen and liver could be the principle target cells of SFTSV. In addition, histological analysis showed that the vascular endothelium integrity was severely disrupted upon viral infection along with depletion of platelets. In vitro cellular assays further revealed that SFTSV infection increased the vascular permeability of endothelial cells by promoting tyrosine phosphorylation and internalization of the adhesion molecule vascular endothelial (VE)-cadherin, a critical component of endothelial integrity. In addition, we found that both virus infection and pathogen-induced exuberant cytokine release dramatically contributed to the vascular endothelial injury. We elucidated the pathogenic mechanisms of hemorrhage syndrome and developed a humanized mouse model for SFTSV infection, which should be helpful for anti-SFTSV therapy and pathogenesis study.


Subject(s)
Disease Models, Animal , Phlebovirus/pathogenicity , Severe Fever with Thrombocytopenia Syndrome/pathology , Tick-Borne Diseases/pathology , Animals , Blood Platelets/pathology , Blood Platelets/virology , Cell Adhesion Molecules/metabolism , Endothelial Cells/pathology , Endothelial Cells/virology , Female , Humans , Leukocytes, Mononuclear/pathology , Leukocytes, Mononuclear/virology , Macrophages/pathology , Macrophages/virology , Mice , Phosphorylation , Severe Fever with Thrombocytopenia Syndrome/virology , Tick-Borne Diseases/virology
2.
PLoS Pathog ; 16(12): e1009129, 2020 12.
Article in English | MEDLINE | ID: mdl-33284835

ABSTRACT

Bunyavirus ribonucleoprotein (RNP) that is assembled by polymerized nucleoproteins (N) coating a viral RNA and associating with a viral polymerase can be both the RNA synthesis machinery and the structural core of virions. Bunyaviral N and RNP thus could be assailable targets for host antiviral defense; however, it remains unclear which and how host factors target N/RNP to restrict bunyaviral infection. By mass spectrometry and protein-interaction analyses, we here show that host protein MOV10 targets the N proteins encoded by a group of emerging high-pathogenic representatives of bunyaviruses including severe fever with thrombocytopenia syndrome virus (SFTSV), one of the most dangerous pathogens listed by World Health Organization, in RNA-independent manner. MOV10 that was further shown to be induced specifically by SFTSV and related bunyaviruses in turn inhibits the bunyaviral replication in infected cells in series of loss/gain-of-function assays. Moreover, animal infection experiments with MOV10 knockdown corroborated the role of MOV10 in restricting SFTSV infection and pathogenicity in vivo. Minigenome assays and additional functional and mechanistic investigations demonstrate that the anti-bunyavirus activity of MOV10 is likely achieved by direct impact on viral RNP machinery but independent of its helicase activity and the cellular interferon pathway. Indeed, by its N-terminus, MOV10 binds to a protruding N-arm domain of N consisting of only 34 amino acids but proving important for N function and blocks N polymerization, N-RNA binding, and N-polymerase interaction, disabling RNP assembly. This study not only advances the understanding of bunyaviral replication and host restriction mechanisms but also presents novel paradigms for both direct antiviral action of MOV10 and host targeting of viral RNP machinery.


Subject(s)
Host-Pathogen Interactions/physiology , Nucleocapsid Proteins/metabolism , Phlebovirus/pathogenicity , RNA Helicases/metabolism , Virus Replication/physiology , Animals , Cell Line , Female , Humans , Mice , Mice, Inbred C57BL , Ribonucleoproteins/metabolism , Severe Fever with Thrombocytopenia Syndrome/metabolism
3.
Proc Natl Acad Sci U S A ; 116(8): 3118-3125, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30718396

ABSTRACT

Although climate change may expand the geographical distribution of several vector-borne diseases, the effects of environmental temperature in host defense to viral infection in vivo are unknown. Here, we demonstrate that exposure of mice to the high ambient temperature of 36 °C impaired adaptive immune responses against infection with viral pathogens, influenza, Zika, and severe fever with thrombocytopenia syndrome phlebovirus. Following influenza virus infection, the high heat-exposed mice failed to stimulate inflammasome-dependent cytokine secretion and respiratory dendritic cell migration to lymph nodes. Although commensal microbiota composition remained intact, the high heat-exposed mice decreased their food intake and increased autophagy in lung tissue. Induction of autophagy in room temperature-exposed mice severely impaired virus-specific CD8 T cells and antibody responses following respiratory influenza virus infection. In addition, we found that administration of glucose or dietary short-chain fatty acids restored influenza virus-specific adaptive immune responses in high heat-exposed mice. These findings uncover an unexpected mechanism by which ambient temperature and nutritional status control virus-specific adaptive immune responses.


Subject(s)
Adaptive Immunity/immunology , Influenza A virus/immunology , Influenza, Human/immunology , Phlebovirus/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Hot Temperature , Humans , Inflammasomes/immunology , Influenza A virus/pathogenicity , Influenza, Human/prevention & control , Influenza, Human/virology , Lung/immunology , Lung/virology , Mice , Phlebovirus/pathogenicity , Zika Virus/immunology , Zika Virus/pathogenicity , Zika Virus Infection
4.
J Gen Virol ; 102(11)2021 11.
Article in English | MEDLINE | ID: mdl-34726591

ABSTRACT

Phleboviruses (order Bunyavirales, family Phenuiviridae) are globally emerging arboviruses with a wide spectrum of virulence. Sandfly fever Sicilian virus (SFSV) is one of the most ubiquitous members of the genus Phlebovirus and associated with a self-limited, incapacitating febrile disease in travellers and military troops. The phleboviral NSs protein is an established virulence factor, acting as antagonist of the antiviral interferon (IFN) system. Consistently, we previously reported that SFSV NSs targets the induction of IFN mRNA synthesis by specifically binding to the DNA-binding domain of the IFN transcription factor IRF3. Here, we further characterized the effect of SFSV and its NSs towards IFN induction, and evaluated its potential to affect the downstream IFN-stimulated signalling and the subsequent transactivation of antiviral interferon-stimulated genes (ISGs). We found that SFSV dampened, but did not entirely abolish type I and type III IFN induction. Furthermore, SFSV NSs did not affect IFN signalling, resulting in substantial ISG expression in infected cells. Hence, although SFSV targets IRF3 to reduce IFN induction, it is not capable of entirely disarming the IFN system in the presence of high basal IRF3 and/or IRF7 levels, and we speculate that this significantly contributes to its low level of virulence.


Subject(s)
Interferons/immunology , Phlebotomus Fever/genetics , Phlebotomus Fever/virology , Phlebovirus/immunology , Host-Pathogen Interactions , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferons/genetics , Phlebotomus Fever/immunology , Phlebovirus/genetics , Phlebovirus/isolation & purification , Phlebovirus/pathogenicity , Up-Regulation , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology , Virulence
5.
PLoS Pathog ; 15(2): e1007375, 2019 02.
Article in English | MEDLINE | ID: mdl-30707748

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease localized to China, Japan, and Korea that is characterized by severe hemorrhage and a high fatality rate. Currently, no specific vaccine or treatment has been approved for this disease. To develop a therapeutic agent for SFTS, we isolated antibodies from a phage-displayed antibody library that was constructed from a patient who recovered from SFTS virus (SFTSV) infection. One antibody, designated as Ab10, was reactive to the Gn envelope glycoprotein of SFTSV and protected host cells and A129 mice from infection in both in vitro and in vivo experiments. Notably, Ab10 protected 80% of mice, even when injected 5 days after inoculation with a lethal dose of SFTSV. Using cross-linker assisted mass spectrometry and alanine scanning, we located the non-linear epitope of Ab10 on the Gn glycoprotein domain II and an unstructured stem region, suggesting that Ab10 may inhibit a conformational alteration that is critical for cell membrane fusion between the virus and host cell. Ab10 reacted to recombinant Gn glycoprotein in Gangwon/Korea/2012, HB28, and SD4 strains. Additionally, based on its epitope, we predict that Ab10 binds the Gn glycoprotein in 247 of 272 SFTSV isolates previously reported. Together, these data suggest that Ab10 has potential to be developed into a therapeutic agent that could protect against more than 90% of reported SFTSV isolates.


Subject(s)
Antibodies, Neutralizing/metabolism , Phlebovirus/immunology , Adult , Animals , Antibodies, Neutralizing/physiology , Antibodies, Viral/metabolism , Bunyaviridae Infections/therapy , Epitopes/immunology , Female , Fever , Glutamine/immunology , Glutamine/metabolism , Glycoproteins/immunology , HEK293 Cells , Humans , Leukopenia , Male , Mice , Mice, Knockout , Neutralization Tests , Phlebovirus/pathogenicity , Republic of Korea , Thrombocytopenia/immunology , Viral Envelope Proteins/immunology
6.
Crit Rev Microbiol ; 47(1): 112-125, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33245676

ABSTRACT

Severe fever with thrombocytopenia syndrome virus (SFTSV) is a novel bunyavirus. Since 2007, SFTS disease has been reported in China with high fatality rate up to 30%, which drew high attention from Centre for Disease Control and Prevention and government. SFTSV is endemic in the centra l and eastern China, Korea and Japan. There also have been similar cases reported in Vietnam. The number of SFTSV infection cases has a steady growth in these years. As SFTSV could transmitted from person to person, it will expose the public to infectious risk. In 2018 annual review of the Blueprint list of priority diseases, World Health Organisation has listed SFTSV infection as prioritised diseases for research and development in emergency contexts. However, the pathogenesis of SFTSV remains largely unclear. Currently, there are no specific therapeutics or vaccines to combat infections of SFTSV. This review discusses recent findings of epidemiology, transmission pathway, pathogenesis and treatments of SFTS disease.


Subject(s)
Phlebovirus/physiology , Phlebovirus/pathogenicity , Severe Fever with Thrombocytopenia Syndrome/virology , Animals , Asia/epidemiology , Humans , Phlebovirus/genetics , Severe Fever with Thrombocytopenia Syndrome/epidemiology , Severe Fever with Thrombocytopenia Syndrome/mortality , Severe Fever with Thrombocytopenia Syndrome/transmission , Virulence
7.
J Biol Chem ; 294(24): 9503-9517, 2019 06 14.
Article in English | MEDLINE | ID: mdl-31040183

ABSTRACT

Heartland virus (HRTV) is a pathogenic phlebovirus recently identified in the United States and related to severe fever with thrombocytopenia syndrome virus (SFTSV) emerging in Asia. We previously reported that SFTSV disrupts host antiviral responses directed by interferons (IFNs) and their downstream regulators, signal transducer and activator of transcription (STAT) proteins. However, whether HRTV infection antagonizes the IFN-STAT signaling axis remains unclear. Here, we show that, similar to SFTSV, HRTV also inhibits IFN-α- and IFN-λ-mediated antiviral responses. As expected, the nonstructural protein (NSs) of HRTV (HNSs) robustly antagonized both type I and III IFN signaling. Protein interaction analyses revealed that a common component downstream of type I and III IFN signaling, STAT2, is the target of HNSs. Of note, the DNA-binding and linker domains of STAT2 were required for an efficient HNSs-STAT2 interaction. Unlike the NSs of SFTSV (SNSs), which blocks both STAT2 and STAT1 nuclear accumulation, HNSs specifically blocked IFN-triggered nuclear translocation only of STAT2. However, upon HRTV infection, IFN-induced nuclear translocation of both STAT2 and STAT1 was suppressed, suggesting that STAT1 is an additional HRTV target for IFN antagonism. Consistently, despite HNSs inhibiting phosphorylation only of STAT2 and not STAT1, HRTV infection diminished both STAT2 and STAT1 phosphorylation. These results suggest that HRTV antagonizes IFN antiviral signaling by dampening both STAT2 and STAT1 activities. We propose that HNSs-specific targeting of STAT2 likely plays an important role but is not all of the "tactics" of HRTV in its immune evasion.


Subject(s)
Antiviral Agents/pharmacology , Bunyaviridae Infections/immunology , Cell Nucleus/metabolism , Interferon Type I/pharmacology , Interferons/pharmacology , Phlebovirus/immunology , STAT1 Transcription Factor/antagonists & inhibitors , STAT2 Transcription Factor/antagonists & inhibitors , Bunyaviridae Infections/drug therapy , Bunyaviridae Infections/metabolism , Bunyaviridae Infections/virology , Cell Nucleus/drug effects , Host-Pathogen Interactions , Humans , Immune Evasion , Phlebovirus/drug effects , Phlebovirus/pathogenicity , Phosphorylation , Protein Transport , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Signal Transduction , Interferon Lambda
8.
J Virol ; 93(10)2019 05 15.
Article in English | MEDLINE | ID: mdl-30814285

ABSTRACT

Severe fever with thrombocytopenia syndrome virus (SFTSV) is a novel emerging virus that has been identified in China, South Korea, and Japan, and it induces thrombocytopenia and leukocytopenia in humans with a high case fatality rate. SFTSV is pathogenic to humans, while immunocompetent adult mice and golden Syrian hamsters infected with SFTSV never show apparent symptoms. However, mice deficient for the gene encoding the α chain of the alpha- and beta-interferon receptor (Ifnar1-/- mice) and golden Syrian hamsters deficient for the gene encoding signal transducer and activator of transcription 2 (Stat2-/- hamsters) are highly susceptible to SFTSV infection, with infection resulting in death. The nonstructural protein (NSs) of SFTSV has been reported to inhibit the type I IFN response through sequestration of human STAT proteins. Here, we demonstrated that SFTSV induces lethal acute disease in STAT2-deficient mice but not in STAT1-deficient mice. Furthermore, we discovered that NSs cannot inhibit type I IFN signaling in murine cells due to an inability to bind to murine STAT2. Taken together, our results imply that the dysfunction of NSs in antagonizing murine STAT2 can lead to inefficient replication and the loss of pathogenesis of SFTSV in mice.IMPORTANCE Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease caused by SFTSV, which has been reported in China, South Korea, and Japan. Here, we revealed that mice lacking STAT2, which is an important factor for antiviral innate immunity, are highly susceptible to SFTSV infection. We also show that SFTSV NSs cannot exert its anti-innate immunity activity in mice due to the inability of the protein to bind to murine STAT2. Our findings suggest that the dysfunction of SFTSV NSs as an IFN antagonist in murine cells confers a loss of pathogenicity of SFTSV in mice.


Subject(s)
Bunyaviridae Infections/metabolism , Phlebovirus/metabolism , STAT2 Transcription Factor/metabolism , Animals , Antiviral Agents/metabolism , Bunyaviridae Infections/virology , Glycoproteins/metabolism , HEK293 Cells , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Humans , Immunity, Innate/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phlebotomus Fever/virology , Phlebovirus/pathogenicity , Phosphorylation , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/physiology , Species Specificity , Thrombocytopenia/metabolism , Viral Nonstructural Proteins/metabolism , Virulence
9.
J Virol ; 93(10)2019 05 15.
Article in English | MEDLINE | ID: mdl-30842332

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging, highly pathogenic, infectious disease caused by infection with a newly discovered tick-borne phlebovirus, SFTS virus (SFTSV). Limited information on the molecular mechanism of SFTSV infection and pathogenesis impedes the development of effective vaccines and drugs for SFTS prevention and treatment. In this study, an isobaric tag for relative and absolute quantification (iTRAQ)-based quantitative proteomic analysis of SFTSV-infected HEK 293 cells was performed to explore dynamic host cellular protein responses toward SFTSV infection. A total of 433 of 5,606 host proteins involved in different biological processes were differentially regulated by SFTSV infection. The proteomic results highlighted a potential role of endoplasmic reticular stress-triggered unfolded-protein response (UPR) in SFTSV infection. Further functional studies confirmed that all three major branches of the UPR, including the PKR-like endoplasmic reticulum kinase (PERK), the activating transcription factor-6 (ATF6), and the inositol-requiring protein-1 (IRE1)/X-box-binding protein 1 (XBP1) pathways, were activated by SFTSV. However, only the former two pathways play a crucial role in SFTSV infection. Furthermore, expression of SFTSV glycoprotein (GP) alone was sufficient to stimulate the UPR, whereas suppression of PERK and ATF6 notably decreased GP expression. Interestingly, two other newly discovered phleboviruses, Heartland virus and Guertu virus, also stimulated the UPR, suggesting a common mechanism shared by these genetically related phleboviruses. This study provides a global view to our knowledge on how host cells respond to SFTSV infection and highlights that host cell UPR plays an important role in phlebovirus infection.IMPORTANCE Severe fever with thrombocytopenia syndrome virus (SFTSV) is an emerging tick-borne bunyavirus that causes severe fever with thrombocytopenia syndrome in humans, with a mortality rate reaching up to 30% in some outbreaks. There are currently no U.S. Food and Drug Administration-approved vaccines or specific antivirals available against SFTSV. To comprehensively understand the molecular interactions occurring between SFTSV and the host cell, we exploit quantitative proteomic approach to investigate the dynamic host cellular responses to SFTSV infection. The results highlight multiple biological processes being regulated by SFTSV infection. Among these, we focused on exploration of the mechanism of how SFTSV infection stimulates the host cell's unfolded-protein response (UPR) and identified the UPR as a common feature shared by SFTSV-related new emerging phleboviruses. This study, for the first time to our knowledge, provides a global map for host cellular responses to SFTSV infection and highlighted potential host targets for further research.


Subject(s)
Bunyaviridae Infections/metabolism , Phlebovirus/metabolism , Unfolded Protein Response/physiology , Activating Transcription Factor 6/metabolism , Bunyaviridae Infections/virology , Endoribonucleases/metabolism , Glycoproteins/metabolism , HEK293 Cells , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Humans , Phlebovirus/pathogenicity , Protein Serine-Threonine Kinases/metabolism , Proteomics/methods , Thrombocytopenia/metabolism , X-Box Binding Protein 1/metabolism , eIF-2 Kinase/metabolism
10.
Virol J ; 17(1): 122, 2020 08 05.
Article in English | MEDLINE | ID: mdl-32758245

ABSTRACT

BACKGROUND: Severe fever and thrombocytopenia bunyavirus (SFTSV) infection causes severe fever and thrombocytopenia syndrome with high mortality. It is extremely rare that a transmitting tick can be directly captured in bite wounds, and that SFTSV can be isolated from both the captured tick and patient's serum to establish a solid pathogen diagnosis. CASE PRESENTATION: We report a case infected with severe fever and thrombocytopenia bunyavirus. The 69-year-old male patient presented with fever and tenderness on two lymph nodes in the right groin. A visible tick bite mark appeared on right upper quadrant of the patient's abdomen, and a live tick was captured in the bite wound upon physical examination. The virus was detected in both the blood of the patient and in the tick that stayed in the bite wound for 7 days. The phylogenetic analysis indicated that the SFTSV isolated from the tick and the patient's serum sample belonged to type B, in which the L/S segment of these two isolates shared 100% homology, while the M segment had 99.9% homology. The bitten patient was given various supportive care, but eventually died of multiple organ failure. CONCLUSION: The present case provides strong evidence of SFTSV transmission from H. longicornis to humans, and suggests that direct cross-species transmission can occur without additional intermediate hosts.


Subject(s)
Bites and Stings , Phlebovirus/genetics , Phylogeny , Severe Fever with Thrombocytopenia Syndrome/virology , Ticks/virology , Aged , Animals , China , Fatal Outcome , Humans , Male , Multiple Organ Failure , Phlebovirus/classification , Phlebovirus/pathogenicity , RNA, Viral/blood , Severe Fever with Thrombocytopenia Syndrome/diagnosis , Severe Fever with Thrombocytopenia Syndrome/transmission
11.
J Virol ; 92(13)2018 07 01.
Article in English | MEDLINE | ID: mdl-29643242

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease caused by a tick-borne phlebovirus of the family Bunyaviridae, SFTS virus (SFTSV). Wild-type and type I interferon (IFN-I) receptor 1-deficient (IFNAR1-/-) mice have been established as nonlethal and lethal models of SFTSV infection, respectively. However, the mechanisms of IFN-I production in vivo and the factors causing the lethal disease are not well understood. Using bone marrow-chimeric mice, we found that IFN-I signaling in hematopoietic cells was essential for survival of lethal SFTSV infection. The disruption of IFN-I signaling in hematopoietic cells allowed an increase in viral loads in serum and produced an excess of multiple inflammatory cytokines and chemokines. The production of IFN-I and inflammatory cytokines was abolished by deletion of the signaling molecules IPS-1 and MyD88, essential for retinoic acid-inducible gene I (RIG-I)-like receptor (RLR) and Toll-like receptor (TLR) signaling, respectively. However, IPS-1-/- MyD88-/- mice exhibited resistance to lethal SFTS with a moderate viral load in serum. Taken together, these results indicate that adequate activation of RLR and TLR signaling pathways under low to moderate levels of viremia contributed to survival through the IFN-I-dependent antiviral response during SFTSV infection, whereas overactivation of these signaling pathways under high levels of viremia resulted in abnormal induction of multiple inflammatory cytokines and chemokines, causing the lethal disease.IMPORTANCE SFTSV causes a severe infectious disease in humans, with a high fatality rate of 12 to 30%. To know the pathogenesis of the virus, we need to clarify the innate immune response as a front line of defense against viral infection. Here, we report that a lethal animal model showed abnormal induction of multiple inflammatory cytokines and chemokines by an uncontrolled innate immune response, which triggered the lethal SFTS. Our findings suggest a new strategy to target inflammatory humoral factors to treat patients with severe SFTS. Furthermore, this study may help the investigation of other tick-borne viruses.


Subject(s)
Bunyaviridae Infections/immunology , DEAD Box Protein 58/metabolism , Inflammation Mediators/metabolism , Phlebotomus Fever/immunology , Receptor, Interferon alpha-beta/physiology , Thrombocytopenia/immunology , Toll-Like Receptors/metabolism , Animals , Bunyaviridae Infections/metabolism , Bunyaviridae Infections/virology , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , DEAD Box Protein 58/genetics , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phlebotomus Fever/metabolism , Phlebotomus Fever/virology , Phlebovirus/pathogenicity , Severity of Illness Index , Thrombocytopenia/metabolism , Thrombocytopenia/virology , Toll-Like Receptors/genetics , Viral Load
12.
J Virol ; 92(19)2018 10 01.
Article in English | MEDLINE | ID: mdl-30021900

ABSTRACT

The nonstructural protein (NSs) of severe fever with thrombocytopenia syndrome phlebovirus (SFTSV) sequesters TANK-binding kinase 1 (TBK1) into NSs-induced cytoplasmic structures to inhibit the phosphorylation and nuclear translocation of interferon (IFN) regulatory factor 3 (IRF3) and subsequent interferon beta (IFN-ß) production. Although the C-terminal region of SFTSV NSs (NSs66-249) has been linked to the formation of NSs-induced cytoplasmic structures and inhibition of host IFN-ß responses, the role of the N-terminal region in antagonizing host antiviral responses remains to be defined. Here, we demonstrate that two conserved amino acids at positions 21 and 23 in the SFTSV and heartland virus (HRTV) NSs are essential for suppression of IRF3 phosphorylation and IFN-ß mRNA expression following infection with SFTSV or recombinant influenza virus lacking the NS1 gene. Surprisingly, formation of SFTSV/HRTV NSs-induced cytoplasmic structures is not essential for inhibition of host antiviral responses. Rather, an association between SFTSV/HRTV NSs and TBK1 is required for suppression of mitochondrial antiviral signaling protein (MAVS)-mediated activation of IFN-ß promoter activity. Although SFTSV NSs did not prevent the ubiquitination of TBK1, it associates with TBK1 through its N-terminal kinase domain (residues 1 to 307) to block the autophosphorylation of TBK1. Furthermore, we found that both wild-type NSs and the 21/23A mutant (NSs in which residues at positions 21 and 23 were replaced with alanine) of SFTSV suppressed NLRP3 inflammasome-dependent interleukin-1ß (IL-1ß) secretion, suggesting that the importance of these residues is restricted to TBK1-dependent IFN signaling. Together, our findings strongly implicate the two conserved amino acids at positions 21 and 23 of SFTSV/HRTV NSs in the inhibition of host interferon responses.IMPORTANCE Recognition of viruses by host innate immune systems plays a critical role not only in providing resistance to viral infection but also in the initiation of antigen-specific adaptive immune responses against viruses. Severe fever with thrombocytopenia syndrome (SFTS) is a newly emerging infectious disease caused by the SFTS phlebovirus (SFTSV), a highly pathogenic tick-borne phlebovirus. The 294-amino-acid nonstructural protein (NSs) of SFTSV associates with TANK-binding kinase 1 (TBK1), a key regulator of host innate antiviral immunity, to inhibit interferon beta (IFN-ß) production and enhance viral replication. Here, we demonstrate that two conserved amino acids at positions 21 and 23 in the NSs of SFTSV and heartland virus, another tick-borne phlebovirus, are essential for association with TBK1 and suppression of IFN-ß production. Our results provide important insight into the molecular mechanisms by which SFTSV NSs helps to counteract host antiviral strategies.


Subject(s)
Host-Pathogen Interactions/immunology , Interferon Regulatory Factor-3/immunology , Interferon-beta/immunology , Phlebovirus/immunology , Protein Serine-Threonine Kinases/immunology , Viral Nonstructural Proteins/immunology , Amino Acid Sequence , Conserved Sequence , Gene Expression Regulation , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Interferon Regulatory Factor-3/genetics , Interferon-beta/antagonists & inhibitors , Interferon-beta/genetics , Interleukin-1beta/genetics , Interleukin-1beta/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Phlebotomus Fever/genetics , Phlebotomus Fever/immunology , Phlebotomus Fever/pathology , Phlebotomus Fever/virology , Phlebovirus/pathogenicity , Phosphorylation , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , Protein Transport , Sequence Alignment , Severity of Illness Index , Signal Transduction , Ubiquitination , Viral Nonstructural Proteins/genetics , Viruses, Unclassified/immunology , Viruses, Unclassified/pathogenicity
13.
BMC Infect Dis ; 19(1): 885, 2019 Oct 24.
Article in English | MEDLINE | ID: mdl-31651242

ABSTRACT

BACKGROUND: Severe fever with thrombocytopenia syndrome (SFTS) is a tick-borne infection that has recently emerged. This infectious disease is due to the transfer of SFTS virus (SFTSV) from the infected blood of animals to humans. Approximately 30% of patients who are infected with SFTS die from multiorgan failure associated with severe infection, systemic inflammatory response syndrome, or disseminated intravascular coagulation. We treated an elderly Japanese couple (husband and wife) who had genetically identical SFTSV infections and who both developed severe rhabdomyolysis. CASE PRESENTATION: An 80-year-old man presented to the clinic with a fever; his 74-year-old wife presented with a fever 9 days later. Their laboratory results at diagnosis showed severe rhabdomyolysis with significantly elevated creatinine kinase (detected levels: husband, 9546 U/L; wife, 15,933 U/L). The creatinine kinase isozyme was 100% MM type in both patients. In both the husband and wife, SFTSV was identified with real-time polymerase chain reaction analysis. The detected SFTSVs in both the husband and wife were identical according to the genome sequence analysis. The husband's bone marrow indicated macrophage activation syndrome, but he responded to supportive therapy. He was discharged after being hospitalized for 32 days. The wife was admitted to our hospital in critical condition 2 days after SFTS symptom onset. She died of multiorgan failure 8 days after onset, despite being cared for in an intensive care unit. Both of the patients presented with rhabdomyolysis following SFTS symptom onset. The patients' clinical outcomes were different from each other; i.e., the husband survived, and the wife died. CONCLUSIONS: SFTSV infection-associated rhabdomyolysis has been reported in one patient, and simultaneous onset in two related patients has not been described previously. Our findings suggest that similar biological responses occurred, but they resulted in different clinical outcomes in the patients infected by the identical SFTSV isolates. Notably, a patient's clinical outcome depends on their own immune response. We suggest that one component of viral rhabdomyolysis involves immune-mediated responses. Severe immunological responses may adversely affect the treatment outcome, as demonstrated by the wife's clinical course. Our findings demonstrate that a patient's immune response contributes to their prognosis following SFTSV infection.


Subject(s)
Bunyaviridae Infections/etiology , Phlebovirus/genetics , Rhabdomyolysis/etiology , Aged , Aged, 80 and over , Bone Marrow/pathology , Bone Marrow/virology , Bunyaviridae Infections/immunology , Bunyaviridae Infections/therapy , China , Female , Humans , Male , Multiple Organ Failure , Phlebovirus/pathogenicity , Real-Time Polymerase Chain Reaction , Rhabdomyolysis/therapy , Rhabdomyolysis/virology , Spouses
14.
BMC Infect Dis ; 19(1): 174, 2019 Feb 19.
Article in English | MEDLINE | ID: mdl-30782137

ABSTRACT

BACKGROUND: Severe fever with thrombocytopenia syndrome (SFTS) is emerging in Asian 3 countries, China, Japan and Korea, which are scrub typhus endemic areas, and its incidence is increasing. As the two infections overlap epidemiologically and clinically and the accessibility or sensitivity of diagnostic tests is limited, early clinical prediction may be useful for diagnostic and therapeutic purposes. METHODS: Patients aged ≥16 years who were clinically suspected and laboratory-confirmed to be infected with Orientia tsutsugamushi or the SFTS virus in South Korea were enrolled. Clinical and laboratory parameters were compared. Scrub typhus was further subclassified according to the status of eschar and skin rash. An SFTS prediction scoring tool was generated based on a logistic regression analysis of SFTS compared with scrub typhus. RESULTS: The analysis was performed on 255 patients with scrub typhus and 107 patients with SFTS. At initial presentation, subjective symptoms except for gastrointestinal symptoms, were more prominent in scrub typhus patients. In addition to the characteristic eschar and skin rash, headache was significantly more prominent in scrub typhus, while laboratory abnormalities were more prominent in SFTS. Leukopenia (white blood cell count < 4000/mm3; odds ratio [OR] 30.13), thrombocytopenia (platelet count < 80,000 /mm3; OR 19.73) and low C-reactive protein (< 1 mg/dL; OR 67.46) were consistent risk factors for SFTS (all P < 0.001). A prediction score was generated using these 3 variables, and a score ≥ 2 had a sensitivity of 93.1% (95% confidence interval [CI], 87.9-96.4%) and a specificity of 96.1% (95% CI, 93.8-97.6%) for SFTS. CONCLUSION: This prediction scoring tool may be useful for differentiating SFTS from eschar- or skin rash-negative scrub typhus. It is a simple and readily applicable tool with potential for use in primary care settings.


Subject(s)
Bunyaviridae Infections/diagnosis , Scrub Typhus/diagnosis , Adolescent , Aged , Bunyaviridae Infections/virology , Female , Humans , Leukopenia/virology , Male , Middle Aged , Odds Ratio , Orientia tsutsugamushi/genetics , Orientia tsutsugamushi/pathogenicity , Phlebovirus/genetics , Phlebovirus/pathogenicity , Republic of Korea , Risk Factors , Scrub Typhus/epidemiology , Scrub Typhus/virology , Thrombocytopenia/virology
15.
Transpl Infect Dis ; 21(4): e13098, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31009160

ABSTRACT

Tick-borne infections represent a significant health risk each year in the United States. Immunocompromised patients are typically at risk of more severe disease manifestations than their immunocompetent counterparts. Here we report a case of a newly emerging phlebovirus, Heartland virus, in a heart transplant recipient.


Subject(s)
Bunyaviridae Infections/diagnosis , Heart Transplantation/adverse effects , Transplant Recipients , Aged , Humans , Male , Missouri , Phlebovirus/pathogenicity
16.
Traffic ; 17(6): 639-56, 2016 06.
Article in English | MEDLINE | ID: mdl-26990254

ABSTRACT

Bunyaviruses represent a growing threat to humans and livestock globally. The receptors, cellular factors and endocytic pathways used by these emerging pathogens to infect cells remain largely unidentified and poorly characterized. DC-SIGN is a C-type lectin highly expressed on dermal dendritic cells that has been found to act as an authentic entry receptor for many phleboviruses (Bunyaviridae), including Rift Valley fever virus (RVFV), Toscana virus (TOSV) and Uukuniemi virus (UUKV). We found that these phleboviruses can exploit another C-type lectin, L-SIGN, for infection. L-SIGN shares 77% sequence homology with DC-SIGN and is expressed on liver sinusoidal endothelial cells. L-SIGN is required for UUKV binding but not for virus internalization. An endocytosis-defective mutant of L-SIGN was still able to mediate virus uptake and infection, indicating that L-SIGN acts as an attachment receptor for phleboviruses rather than an endocytic receptor. Our results point out a fundamental difference in the use of the C-type lectins L-SIGN and DC-SIGN by UUKV to enter cells, although both proteins are closely related in terms of molecular structure and biological function. This study sheds new light on the molecular mechanisms by which phleboviruses target the liver and also highlights the added complexity in virus-receptor interactions beyond attachment.


Subject(s)
Cell Adhesion Molecules/metabolism , Endocytosis , Lectins, C-Type/metabolism , Phlebovirus/physiology , Receptors, Cell Surface/metabolism , Cell Adhesion Molecules/genetics , Endothelial Cells/metabolism , Endothelial Cells/virology , HeLa Cells , Humans , Lectins, C-Type/genetics , Liver/cytology , Liver/virology , Phlebovirus/pathogenicity , Protein Binding , Receptors, Cell Surface/genetics , Virus Internalization
17.
Vox Sang ; 113(3): 297-299, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29359332

ABSTRACT

The risk of transfusion-transmitted infection (TTI) for severe fever with thrombocytopenia syndrome virus (SFTSV) is a concern because person-to-person transmission resulting from contact with SFTSV-contaminated blood has been reported. To obtain information regarding the risk of TTI-SFTSV, antibody testing was performed for blood samples donated in an severe fever with thrombocytopenia syndrome-endemic area in Japan. No antibody-positive samples were detected among 3990 samples. This finding suggested that there were few cases of SFTSV infection among donors and that the risk of TTI-SFTSV was also estimated low in Japan.


Subject(s)
Bunyaviridae Infections/epidemiology , Phlebovirus/immunology , Transfusion Reaction/epidemiology , Adult , Antibodies, Viral/blood , Blood Donors , Bunyaviridae Infections/blood , Female , Humans , Japan , Male , Phlebovirus/pathogenicity , Transfusion Reaction/blood , Transfusion Reaction/virology
18.
BMC Infect Dis ; 18(1): 181, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29665796

ABSTRACT

BACKGROUND: Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne disease. Haemophysalis longicornis ticks have been considered the vector of severe fever with thrombocytopenia syndrome virus (SFTSV). However, clear data on the transmission of SFTS from ticks to humans are limited. CASE PRESENTATION: We report an 84-year-old woman who presented with fever and altered mentality, which was confirmed as SFTS with encephalopathy by reverse-transcription polymerase chain reaction in blood and cerebrospinal fluid. The SFTSV was also identified in the tick that bit her, H. longicornis. Phylogenetic analyses indicated that the SFTSV from the patient and the tick was identical. The patient gradually recovered with treatments of corticosteroids and immunoglobulin. CONCLUSION: These findings provide further evidence of SFTS viral transmission from H. longicornis to human.


Subject(s)
Brain Diseases/virology , Bunyaviridae Infections/virology , Ixodidae/virology , Phlebovirus/genetics , Aged, 80 and over , Animals , Arachnid Vectors/virology , Brain Diseases/etiology , Bunyaviridae Infections/etiology , Bunyaviridae Infections/therapy , Cerebrospinal Fluid/virology , Female , Humans , Phlebovirus/pathogenicity , Phylogeny
19.
J Infect Chemother ; 24(10): 773-781, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30098914

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) caused by SFTS virus (SFTSV), a novel phlebovirus, was reported to be endemic to central and northeastern PR China and was also to be endemic to South Korea and western Japan. SFTS is an emerging viral infection, which should be categorized as a viral hemorrhagic fever disease as Crimean-Congo hemorrhagic fever (CCHF) is caused by CCHF virus. SFTS is a tick-borne viral infection. SFTSV is maintained between several species of ticks and wild and domestic animals in nature. Patients with SFTS show symptoms of fever, general fatigue, and gastrointestinal symptoms such as bloody diarrhea. The severely ill SFTS patients usually show gastrointestinal hemorrhage and deteriorated consciousness. The case fatality rate of SFTS ranges from 5 to 40%. Pathological studies on SFTS have revealed that the mechanisms behind the high case fatality rate are virus infection-related hemophagocytic syndrome associated with cytokine storm, coagulopathy due to disseminated intravascular coagulation causing bleeding tendency, and multi-organ failure. Favipiravir was reported to show efficacy in the prevention and treatment of SFTSV infections in an animal model. A clinical study to evaluate the efficacy of favipiravir in the treatment of SFTS patients has been initiated in Japan. SFTSV is circulating in nature in PR China, Korea, and Japan, indicating that we cannot escape from the risk being infected with SFTSV. The development of specific therapy and preventive measures is a pressing issue requiring resolution to reduce the morbidity and mortality of SFTS patients.


Subject(s)
Amides/therapeutic use , Antiviral Agents/therapeutic use , Bunyaviridae Infections/drug therapy , Phlebovirus/pathogenicity , Pyrazines/therapeutic use , Thrombocytopenia/drug therapy , Aged, 80 and over , Animals , Bunyaviridae Infections/epidemiology , Bunyaviridae Infections/pathology , Bunyaviridae Infections/prevention & control , China/epidemiology , Female , Humans , Japan/epidemiology , Male , Mice , Mice, Knockout , Middle Aged , Mortality , Republic of Korea/epidemiology , Syndrome , Thrombocytopenia/epidemiology , Thrombocytopenia/pathology , Thrombocytopenia/prevention & control
20.
Uirusu ; 68(1): 41-50, 2018.
Article in Japanese | MEDLINE | ID: mdl-31105134

ABSTRACT

Seven years have passed since the discovery of a novel infectious disease, severe fever with thrombocytopenia syndrome (SFTS) caused by a novel Phlebovirus, SFTS virus (SFTSV), in PR China. It was also confirmed that SFTS was endemic to Japan through an identification of a woman, who died of SFTSV infection in Yamaguchi prefecture in late 2012. Approximately 6 years have passed since the discovery of SFTS-endemicity in Japan. At present, SFTS is endemic to PR China, South Korea and western Japan. SFTSV is maintained between several species of ticks such as Haemaphysalis longicornis and wild and domestic animals in nature. Therefore, we cannot escape from the risk of being infected with SFTSV. Based on the similarity in the characteristics of the clinical symptoms including the high case fatality rate, mode of infection to humans, pathology and virology between SFTS and Crimean-Congo hemorrhagic fever (CCHF), SFTS should be classified as viral hemorrhagic fever. Although the time from the discovery of SFTS is still short, there have been many scientific reports on the epidemiological, clinical, and/or pathological, and virological studies on SFTS. Favipiravir was reported to show an efficacy in the prevention and treatment of SFTSV infections in an animal model. A clinical study to evaluate the efficacy of favipiravir in the treatment of SFTS patients has been initiated in Japan. Specific and effective treatment with antiviral drugs for and preventive measures of SFTS with vaccination shoued be developed through scientific, clinical, and basic research.


Subject(s)
Disease Outbreaks , Phlebotomus Fever/transmission , Phlebotomus Fever/virology , Phlebovirus/pathogenicity , Animals , Antiviral Agents/therapeutic use , Asia, Eastern/epidemiology , Humans , Phlebotomus Fever/drug therapy , Phlebotomus Fever/prevention & control , Phlebovirus/immunology , Ticks/virology , Viral Vaccines
SELECTION OF CITATIONS
SEARCH DETAIL