Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(24): e2400163121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38830098

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease with a high fatality rate of up to 30% caused by SFTS virus (SFTSV). However, no specific vaccine or antiviral therapy has been approved for clinical use. To develop an effective treatment, we isolated a panel of human monoclonal antibodies (mAbs). SF5 and SF83 are two neutralizing mAbs that recognize two viral glycoproteins (Gn and Gc), respectively. We found that their epitopes are closely located, and we then engineered them as several bispecific antibodies (bsAbs). Neutralization and animal experiments indicated that bsAbs display more potent protective effects than the parental mAbs, and the cryoelectron microscopy structure of a bsAb3 Fab-Gn-Gc complex elucidated the mechanism of protection. In vivo virus passage in the presence of antibodies indicated that two bsAbs resulted in less selective pressure and could efficiently bind to all single parental mAb-escape mutants. Furthermore, epitope analysis of the protective mAbs against SFTSV and RVFV indicated that they are all located on the Gn subdomain I, where may be the hot spots in the phleboviruses. Collectively, these data provide potential therapeutic agents and molecular basis for the rational design of vaccines against SFTSV infection.


Subject(s)
Antibodies, Bispecific , Antibodies, Neutralizing , Antibodies, Viral , Phlebovirus , Animals , Antibodies, Bispecific/immunology , Mice , Antibodies, Neutralizing/immunology , Phlebovirus/immunology , Humans , Antibodies, Viral/immunology , Glycoproteins/immunology , Antibodies, Monoclonal/immunology , Epitopes/immunology , Disease Models, Animal , Severe Fever with Thrombocytopenia Syndrome/immunology , Severe Fever with Thrombocytopenia Syndrome/prevention & control
2.
J Virol ; 98(7): e0076924, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38829138

ABSTRACT

Highly pathogenic viruses from family Phenuiviridae, which are mainly transmitted by arthropods, have intermittently sparked epidemics worldwide. In particular, tick-borne bandaviruses, such as severe fever with thrombocytopenia syndrome virus (SFTSV), continue to spread in mountainous areas, resulting in an average mortality rate as high as 10.5%, highlighting the urgency and importance of vaccine development. Here, an mRNA vaccine developed based on the full-length SFTSV glycoprotein, containing both the receptor-binding domain and the fusion domain, was shown to confer complete protection against SFTSV at a very low dose by triggering a type 1 helper T cell-biased cellular immune response in rodents. Moreover, the vaccine candidate elicited long-term immunity and protection against SFTSV for at least 5 months. Notably, it provided complete cross-protection against other bandaviruses, such as the Heartland virus and Guertu virus, in lethal challenge models. Further research revealed that the conserved epitopes among bandaviruses within the full-length SFTSV glycoprotein may facilitate broad-spectrum protection mediated by the cellular immune response. Collectively, these findings demonstrate that the full-length SFTSV glycoprotein mRNA vaccine is a promising vaccine candidate for SFTSV and other bandaviruses, and provide guidance for the development of broad-spectrum vaccines from conserved antigens and epitopes. IMPORTANCE: Tick-borne bandaviruses, such as SFTSV and Heartland virus, sporadically trigger outbreaks in addition to influenza viruses and coronaviruses, yet there are no specific vaccines or therapeutics against them. mRNA vaccine technology has advantages in terms of enabling in situ expression and triggering cellular immunity, thus offering new solutions for vaccine development against intractable viruses, such as bandaviruses. In this study, we developed a novel vaccine candidate for SFTSV by employing mRNA vaccination technology and using a full-length glycoprotein as an antigen target. This candidate vaccine confers complete and durable protection against SFTSV at a notably low dose while also providing cross-protection against Heartland virus and Guertu virus. This study highlights the prospective value of full-length SFTSV-glycoprotein-based mRNA vaccines and suggests a potential strategy for broad-spectrum bandavirus vaccines.


Subject(s)
Glycoproteins , Phlebovirus , Severe Fever with Thrombocytopenia Syndrome , Viral Vaccines , Animals , Phlebovirus/immunology , Phlebovirus/genetics , Mice , Severe Fever with Thrombocytopenia Syndrome/prevention & control , Severe Fever with Thrombocytopenia Syndrome/immunology , Glycoproteins/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Antibodies, Viral/immunology , Antibodies, Viral/blood , mRNA Vaccines/immunology , Cross Protection/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Female , Immunity, Cellular , Mice, Inbred BALB C
3.
BMC Infect Dis ; 24(1): 476, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714948

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne viral disease caused by the SFTS virus (Dabie bandavirus), which has become a substantial risk to public health. No specific treatment is available now, that calls for an effective vaccine. Given this, we aimed to develop a multi-epitope DNA vaccine through the help of bioinformatics. The final DNA vaccine was inserted into a special plasmid vector pVAX1, consisting of CD8+ T cell epitopes, CD4+ T cell epitopes and B cell epitopes (six epitopes each) screened from four genome-encoded proteins--nuclear protein (NP), glycoprotein (GP), RNA-dependent RNA polymerase (RdRp), as well as nonstructural protein (NSs). To ascertain if the predicted structure would be stable and successful in preventing infection, an immunological simulation was run on it. In conclusion, we designed a multi-epitope DNA vaccine that is expected to be effective against Dabie bandavirus, but in vivo trials are needed to verify this claim.


Subject(s)
Epitopes, T-Lymphocyte , Phlebovirus , Severe Fever with Thrombocytopenia Syndrome , Vaccines, DNA , Viral Vaccines , Vaccines, DNA/immunology , Vaccines, DNA/genetics , Phlebovirus/immunology , Phlebovirus/genetics , Severe Fever with Thrombocytopenia Syndrome/prevention & control , Severe Fever with Thrombocytopenia Syndrome/immunology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Viral Vaccines/immunology , Viral Vaccines/genetics , Humans , Computer-Aided Design , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Animals , Computational Biology
4.
J Microbiol ; 62(4): 327-335, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38635002

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) is a life-threatening viral zoonosis. The causative agent of this disease is the Dabie bandavirus, which is usually known as the SFTS virus (SFTSV). Although the role of vertebrates in SFTSV transmission to humans remains uncertain, some reports have suggested that dogs could potentially transmit SFTSV to humans. Consequently, preventive measures against SFTSV in dogs are urgently needed. In the present study, dogs were immunized three times at two-week intervals with formaldehyde-inactivated SFTSV with two types of adjuvants. SFTSV (KCD46) was injected into all dogs two weeks after the final immunization. Control dogs showed viremia from 2 to 4 days post infection (dpi), and displayed white pulp atrophy in the spleen, along with a high level of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling assay (TUNEL) positive area. However, the inactivated SFTSV vaccine groups exhibited rare pathological changes and significantly reduced TUNEL positive areas in the spleen. Furthermore, SFTSV viral loads were not detected at any of the tested dpi. Our results indicate that both adjuvants can be safely used in combination with an inactivated SFTSV formulation to induce strong neutralizing antibodies. Inactivated SFTSV vaccines effectively prevent pathogenicity and viremia in dogs infected with SFTSV. In conclusion, our study highlighted the potential of inactivated SFTSV vaccination for SFTSV control in dogs.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Dog Diseases , Phlebovirus , Severe Fever with Thrombocytopenia Syndrome , Vaccines, Inactivated , Viral Vaccines , Animals , Dogs , Phlebovirus/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Severe Fever with Thrombocytopenia Syndrome/virology , Severe Fever with Thrombocytopenia Syndrome/prevention & control , Severe Fever with Thrombocytopenia Syndrome/immunology , Severe Fever with Thrombocytopenia Syndrome/veterinary , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Dog Diseases/virology , Dog Diseases/prevention & control , Dog Diseases/immunology , Viremia , Viral Load , Spleen/virology , Spleen/pathology , Spleen/immunology , Adjuvants, Immunologic/administration & dosage , Vaccination/veterinary
5.
Sci Rep ; 11(1): 21831, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34750465

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne infectious disease in China, Japan, and Korea. This study aimed to estimate the monthly SFTS occurrence and the monthly number of SFTS cases in the geographical area in Korea using epidemiological data including demographic, geographic, and meteorological factors. Important features were chosen through univariate feature selection. Two models using machine learning methods were analyzed: the classification model in machine learning (CMML) and regression model in machine learning (RMML). We developed a novel model incorporating the CMML results into RMML, defined as modified-RMML. Feature importance was computed to assess the contribution of estimating the number of SFTS cases using modified-RMML. Aspect to the accuracy of the novel model, the performance of modified-RMML was improved by reducing the MSE for the test data as 12.6-52.2%, compared to the RMML using five machine learning methods. During the period of increasing the SFTS cases from May to October, the modified-RMML could give more accurate estimation. Computing the feature importance, it is clearly observed that climate factors such as average maximum temperature, precipitation as well as mountain visitors, and the estimation of SFTS occurrence obtained from CMML had high Gini importance. The novel model incorporating CMML and RMML models improves the accuracy of the estimation of SFTS cases. Using the model, climate factors, including temperature, relative humidity, and mountain visitors play important roles in transmitting SFTS in Korea. Our findings highlighted that the guidelines for mountain visitors to prevent SFTS transmissions should be addressed. Moreover, it provides important insights for establishing control interventions that predict early identification of SFTS cases.


Subject(s)
Machine Learning , Severe Fever with Thrombocytopenia Syndrome/epidemiology , Severe Fever with Thrombocytopenia Syndrome/transmission , Aged , Climate , Computer Simulation , Epidemiological Models , Female , Humans , Male , Middle Aged , Mountaineering , Regression Analysis , Republic of Korea/epidemiology , Risk Factors , Severe Fever with Thrombocytopenia Syndrome/prevention & control , Travel-Related Illness
6.
Exp Mol Med ; 53(5): 713-722, 2021 05.
Article in English | MEDLINE | ID: mdl-33953322

ABSTRACT

An emerging infectious disease first identified in central China in 2009, severe fever with thrombocytopenia syndrome (SFTS) was found to be caused by a novel phlebovirus. Since SFTSV was first identified, epidemics have occurred in several East Asian countries. With the escalating incidence of SFTS and the rapid, worldwide spread of SFTSV vector, it is clear this virus has pandemic potential and presents an impending global public health threat. In this review, we concisely summarize the latest findings regarding SFTSV, including vector and virus transmission, genotype diversity and epidemiology, probable pathogenic mechanism, and clinical presentation of human SFTS. Ticks most likely transmit SFTSV to animals including humans; however, human-to-human transmission has been reported. The majority of arbovirus transmission cycle includes vertebrate hosts, and potential reservoirs include a variety of both domestic and wild animals. Reports of the seroprevalence of SFTSV in both wild and domestic animals raises the probability that domestic animals act as amplifying hosts for the virus. Major clinical manifestation of human SFTS infection is high fever, thrombocytopenia, leukocytopenia, gastrointestinal symptoms, and a high case-fatality rate. Several animal models were developed to further understand the pathogenesis of the virus and aid in the discovery of therapeutics and preventive measures.


Subject(s)
Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/virology , Disease Susceptibility , Phlebovirus/physiology , Severe Fever with Thrombocytopenia Syndrome/epidemiology , Severe Fever with Thrombocytopenia Syndrome/virology , Animals , Communicable Disease Control , Communicable Diseases, Emerging/prevention & control , Communicable Diseases, Emerging/transmission , Disease Management , Disease Models, Animal , Disease Susceptibility/immunology , Genetic Variation , Host-Pathogen Interactions/immunology , Humans , Phlebovirus/classification , Reassortant Viruses , Seroepidemiologic Studies , Severe Fever with Thrombocytopenia Syndrome/prevention & control , Severe Fever with Thrombocytopenia Syndrome/transmission , Symptom Assessment , Viral Zoonoses
7.
Vet Microbiol ; 257: 109076, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33957572

ABSTRACT

The Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne hemorrhagic zoonotic disease, which is potentially fatal in human with mortality rates ranging from 16.2%-32%. The rabies virus (RABV) LBNSE vector expressing foreign antigens have shown considerable promise as vaccines against viral diseases, which is effective and safe. In the present study, we generated a recombinant RABV rLBNSE-Gn expressing a SFTSV glycoprotein Gn by reverse genetic technology to control rabies and SFTS in animals. An extra insertion of Gn gene did not impact replication of the recombinant virus rLBNSE-Gn in NA and BHK-21 cells compared to the parent rLBNSE strain. The SFTSV Gn gene together with RABV N and G genes were efficiently expressed in rLBNSE-infected Vero cells by immunostaining and immune blots. A single dose of 107 FFU of the rLBNSE-Gn intramuscularly inoculated in BALB/c mice induced rapid and robust humoral responses against both RABV and SFTSV without any signs of disease or weight loss. Compared to the rLBNSE and DMEM groups, the extra Gn expression contributed to the recruitments and/or activations of the dendritic cells and B cells from inguinal lymph nodes of BALB/c mice vaccinated with rLBNSE-Gn. The protective efficacy of rLBNSE-Gn against SFTSV in C57BL/6 mice was evaluated, and the virus loading in the spleens reduced to 10 TCID50/mg at 7 days post SFTSV infections, which indicated that the rLBNSE-Gn conferred efficacious protective immune responses from SFTSV in C57BL/6 mice. All the mice immunization with rLBNSE-Gn and rLBNSE survived after a lethal RABV challenge, suggesting a 100 % protection from RABV. Therefore, the rLBNSE-Gn would be a promising bivalent candidate vaccine against SFTS and rabies in animals.


Subject(s)
Antibodies, Viral/blood , Genetic Vectors , Phlebovirus/immunology , Rabies virus/genetics , Rabies/prevention & control , Severe Fever with Thrombocytopenia Syndrome/prevention & control , Viral Vaccines/immunology , Animals , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phlebovirus/genetics , Rabies Vaccines/administration & dosage , Rabies Vaccines/immunology , Rabies virus/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vaccines, Combined/administration & dosage , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
8.
Parasit Vectors ; 14(1): 237, 2021 May 06.
Article in English | MEDLINE | ID: mdl-33957950

ABSTRACT

BACKGROUND: Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease that is regionally distributed in Asia, with high fatality. Constructing the transmission model of SFTS could help provide clues for disease control and fill the gap in research on SFTS models. METHODS: We built an SFTS transmission dynamics model based on the susceptible-exposed-infectious-asymptomatic-recovered (SEIAR) model and the epidemiological characteristics of SFTS in Jiangsu Province. This model was used to evaluate the effect by cutting off different transmission routes and taking different interventions into account, to offer clues for disease prevention and control. RESULTS: The transmission model fits the reported data well with a minimum R2 value of 0.29 and a maximum value of 0.80, P < 0.05. Meanwhile, cutting off the environmental transmission route had the greatest effect on the prevention and control of SFTS, while isolation and shortening the course of the disease did not have much effect. CONCLUSIONS: The model we have built can be used to simulate the transmission of SFTS to help inform disease control. It is noteworthy that cutting off the environment-to-humans transmission route in the model had the greatest effect on SFTS prevention and control.


Subject(s)
Severe Fever with Thrombocytopenia Syndrome/transmission , Animals , Arachnid Vectors/virology , China/epidemiology , Humans , Incidence , Middle Aged , Models, Theoretical , Severe Fever with Thrombocytopenia Syndrome/epidemiology , Severe Fever with Thrombocytopenia Syndrome/prevention & control , Ticks/virology
9.
Viruses ; 13(4)2021 04 06.
Article in English | MEDLINE | ID: mdl-33917632

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS), which is caused by SFTS virus (SFTSV), is a tick-borne emerging zoonosis with a high case-fatality rate. At present, there is no approved SFTS vaccine, although the development of a vaccine would be one of the best strategies for preventing SFTS. This article focused on studies aimed at establishing small animal models of SFTS that are indispensable for evaluating vaccine candidates, developing these vaccine candidates, and establishing more practical animal models for evaluation. Innate immune-deficient mouse models, a hamster model, an immunocompetent ferret model and a cat model have been developed for SFTS. Several vaccine candidates for SFTS have been developed, and their efficacy has been confirmed using these animal models. The candidates consist of live-attenuated virus-based, viral vector-based, or DNA-based vaccines. SFTS vaccines are expected to be used for humans and companion dogs and cats. Hence for practical use, the vaccine candidates should be evaluated for efficacy using not only nonhuman primates but also dogs and cats. There is no practical nonhuman primate model of SFTS; however, the cat model is available to evaluate the efficacy of these candidate SFTS vaccines on domesticated animals.


Subject(s)
Disease Models, Animal , Phlebovirus/immunology , Severe Fever with Thrombocytopenia Syndrome/prevention & control , Viral Vaccines/immunology , Animals , Cat Diseases/immunology , Cat Diseases/prevention & control , Cat Diseases/virology , Cats , Cricetinae , Dog Diseases/immunology , Dog Diseases/prevention & control , Dog Diseases/virology , Dogs , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phlebovirus/pathogenicity , Primates , Severe Fever with Thrombocytopenia Syndrome/immunology
10.
PLoS Pathog ; 17(2): e1008859, 2021 02.
Article in English | MEDLINE | ID: mdl-33534867

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) caused by a species Dabie bandavirus (formerly SFTS virus [SFTSV]) is an emerging hemorrhagic infectious disease with a high case-fatality rate. One of the best strategies for preventing SFTS is to develop a vaccine, which is expected to induce both humoral and cellular immunity. We applied a highly attenuated but still immunogenic vaccinia virus strain LC16m8 (m8) as a recombinant vaccine for SFTS. Recombinant m8s expressing SFTSV nucleoprotein (m8-N), envelope glycoprotein precursor (m8-GPC), and both N and GPC (m8-N+GPC) in the infected cells were generated. Both m8-GPC- and m8-N+GPC-infected cells were confirmed to produce SFTSV-like-particles (VLP) in vitro, and the N was incorporated in the VLP produced by the infection of cells with m8-N+GPC. Specific antibodies to SFTSV were induced in mice inoculated with each of the recombinant m8s, and the mice were fully protected from lethal challenge with SFTSV at both 103 TCID50 and 105 TCID50. In mice that had been immunized with vaccinia virus strain Lister in advance of m8-based SFTSV vaccine inoculation, protective immunity against the SFTSV challenge was also conferred. The pathological analysis revealed that mice immunized with m8-GPC or m8-N+GPC did not show any histopathological changes without any viral antigen-positive cells, whereas the control mice showed focal necrosis with inflammatory infiltration with SFTSV antigen-positive cells in tissues after SFTSV challenge. The passive serum transfer experiments revealed that sera collected from mice inoculated with m8-GPC or m8-N+GPC but not with m8-N conferred protective immunity against lethal SFTSV challenge in naïve mice. On the other hand, the depletion of CD8-positive cells in vivo did not abrogate the protective immunity conferred by m8-based SFTSV vaccines. Based on these results, the recombinant m8-GPC and m8-N+GPC were considered promising vaccine candidates for SFTS.


Subject(s)
Antigens, Viral/immunology , Nucleoproteins/immunology , Phlebovirus/immunology , Severe Fever with Thrombocytopenia Syndrome/prevention & control , Vaccines, Attenuated/administration & dosage , Vaccines, Synthetic/administration & dosage , Viral Envelope Proteins/immunology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Severe Fever with Thrombocytopenia Syndrome/immunology , Severe Fever with Thrombocytopenia Syndrome/virology
SELECTION OF CITATIONS
SEARCH DETAIL