Hantaan Virus

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 1575 Experts worldwide ranked by ideXlab platform

Connie S. Schmaljohn - One of the best experts on this subject based on the ideXlab platform.

  • inhibition of tnf α induced activation of nf κb by hantaVirus nucleocapsid proteins
    Annals of the New York Academy of Sciences, 2009
    Co-Authors: Shannon L Taylor, Ryan L Krempel, Connie S. Schmaljohn
    Abstract:

    HantaViruses cause two human diseases thought to be immune-mediated: hemorrhagic fever with renal syndrome (HFRS) and hantaVirus pulmonary syndrome (HPS). We recently reported that the nucleocapsid (N) protein of HFRS-causing Hantaan Virus (HTNV) inhibits tumor necrosis factor-alpha (TNF-alpha) activation of nuclear factor-kappaB (NF-kappaB). Here we measured the ability of other hantaviral N proteins to similarly interfere with the inflammatory process of TNF-alpha. We found that like HTNV N, the N proteins of HFRS-causing Seoul and Dobrava Viruses inhibited TNF-alpha activation of NF-kappaB and translocation of the NF-kappaB p65 subunit, but did not interfere with degradation of inhibitor of NF-kappaB (IkappaB). In contrast, the HFRS-causing Puumala Virus and the HPS-causing Andes and Sin Nombre Viruses did not prevent TNF-alpha activation of NF-kappaB or nuclear translocation of p65. These studies provide evidence that hantaViruses differ in their abilities to interfere with host innate immune responses.

  • Hantaan Virus nucleocapsid protein binds to importin alpha proteins and inhibits tumor necrosis factor alpha induced activation of nuclear factor kappa b
    Journal of Virology, 2009
    Co-Authors: Shannon L Taylor, Natalia Friasstaheli, Adolfo Garciasastre, Connie S. Schmaljohn
    Abstract:

    HantaViruses such as Hantaan Virus (HTNV) and Andes Virus cause two human diseases, hemorrhagic fever with renal syndrome and hantaVirus pulmonary syndrome, respectively. For both, disease pathogenesis is thought to be immunologically mediated and there have been numerous reports of patients with elevated levels of proinflammatory and inflammatory cytokines, including tumor necrosis factor alpha (TNF-alpha), in their sera. Multiple Viruses have developed evasion strategies to circumvent the host cell inflammatory process, with one of the most prevalent being the disruption of nuclear factor kappa B (NF-kappaB) activation. We hypothesized that hantaViruses might also moderate host inflammation by interfering with this pathway. We report here that the nucleocapsid (N) protein of HTNV was able to inhibit TNF-alpha-induced activation of NF-kappaB, as measured by a reporter assay, and the activation of endogenous p65, an NF-kappaB subunit. Surprisingly, there was no defect in the degradation of the inhibitor of NF-kappaB (IkappaB) protein, nor was there any alteration in the level of p65 expression in HTNV N-expressing cells. However, immunofluorescence antibody staining demonstrated that cells expressing HTNV N protein and a green fluorescent protein-p65 fusion had limited p65 nuclear translocation. Furthermore, we were able to detect an interaction between HTNV N protein and importin alpha, a nuclear import molecule responsible for shuttling NF-kappaB to the nucleus. Collectively, our data suggest that HTNV N protein can sequester NF-kappaB in the cytoplasm, thus inhibiting NF-kappaB activity. These findings, which were obtained using cells transfected with cDNA representing the HTNV N gene, were confirmed using HTNV-infected cells.

  • mixing of m segment dna vaccines to Hantaan Virus and puumala Virus reduces their immunogenicity in hamsters
    Vaccine, 2008
    Co-Authors: Kristin Spik, Catherine V Badger, Iacob Mathiessen, Torunn Elisabeth Tjelle, Jay W Hooper, Connie S. Schmaljohn
    Abstract:

    To determine if DNA vaccines for two hantaViruses causing hemorrhagic fever with renal syndrome, Hantaan Virus and Puumala Virus, are immunogenic when given in combination, we delivered them to hamsters separately or as mixtures by gene gun or by electroporation. Both vaccines elicited neutralizing antibodies when given alone but when they were delivered as a mixture, antibodies to only one of the two hantaViruses could be detected. In contrast, if the DNAs were given as separate vaccinations to a single animal, responses to both were observed. These studies suggest that the two DNA vaccines will need to be given as separate administrations.

  • immunogenicity of combination dna vaccines for rift valley fever Virus tick borne encephalitis Virus Hantaan Virus and crimean congo hemorrhagic fever Virus
    Vaccine, 2006
    Co-Authors: Kristin Spik, Jay W Hooper, Amy C Shurtleff, Anita K Mcelroy, Mary C Guttieri, Connie S. Schmaljohn
    Abstract:

    DNA vaccines for Rift Valley fever Virus (RVFV), Crimean Congo hemorrhagic fever Virus (CCHFV), tick-borne encephalitis Virus (TBEV), and Hantaan Virus (HTNV), were tested in mice alone or in various combinations. The bunyaVirus vaccines (RVFV, CCHFV, and HTNV) expressed Gn and Gc genes, and the flaviVirus vaccine (TBEV) expressed the preM and E genes. All vaccines were delivered by gene gun. The TBEV DNA vaccine and the RVFV DNA vaccine elicited similar levels of antibodies and protected mice from challenge when delivered alone or in combination with other DNAs. Although in general, the HTNV and CCHFV DNA vaccines were not very immunogenic in mice, there were no major differences in performance when given alone or in combination with the other vaccines.

  • dna vaccination with the Hantaan Virus m gene protects hamsters against three of four hfrs hantaViruses and elicits a high titer neutralizing antibody response in rhesus monkeys
    Journal of Virology, 2001
    Co-Authors: Jay W Hooper, David Custer, E A Thompson, Connie S. Schmaljohn
    Abstract:

    Four hantaViruses-Hantaan Virus (HTNV), Seoul Virus (SEOV), Dobrava Virus (DOBV) and Puumala Virus-are known to cause hemorrhagic fever with renal syndrome (HFRS) in Europe and Asia. HTNV causes the most severe form of HFRS (5 to 15% case-fatality rate) and afflicts tens of thousands of people annually. Previously, we demonstrated that DNA vaccination with a plasmid expressing the SEOV M gene elicited neutralizing antibodies and protected hamsters against infection with SEOV and HTNV. Here, we report the construction and evaluation of a DNA vaccine that expresses the HTNV M gene products, G1 and G2. DNA vaccination of hamsters with the HTNV M gene conferred sterile protection against infection with HTNV, SEOV, and DOBV. DNA vaccination of rhesus monkeys with either the SEOV or HTNV M gene elicited high levels of neutralizing antibodies. These are the first immunogenicity data for hantaVirus DNA vaccines in nonhuman primates. Because a neutralizing antibody response is considered a surrogate marker for protective immunity in humans, our protection data in hamsters combined with the immunogenicity data in monkeys suggest that hantaVirus M gene-based DNA vaccines could protect humans against the most severe forms of HFRS.

Jin-won Song - One of the best experts on this subject based on the ideXlab platform.

Linfeng Cheng - One of the best experts on this subject based on the ideXlab platform.

  • In-Cell Western Assays to Evaluate Hantaan Virus Replication as a Novel Approach to Screen Antiviral Molecules and Detect Neutralizing Antibody Titers.
    Frontiers in cellular and infection microbiology, 2017
    Co-Authors: Hesong Chen, Linfeng Cheng, Liang Zhang, Tie-jian Nie, Peijun Han, Yingfeng Lei
    Abstract:

    HantaViruses encompass rodent-borne zoonotic pathogens that cause severe hemorrhagic fever disease with high mortality rates in humans. Detection of infectious Virus titer lays a solid foundation for virology and immunology researches. Canonical methods to assess viral titers rely on visible cytopathic effects (CPE), but Hantaan Virus (HTNV, the prototype hantaVirus) maintains a relatively sluggish life cycle and does not produce CPE in cell culture. Here, an in-cell Western (ICW) assay was utilized to rapidly measure the expression of viral proteins in infected cells and to establish a novel approach to detect viral titers. Compared with classical approaches, the ICW assay is accurate and time- and cost-effective. Furthermore, the ICW assay provided a high-throughput platform to screen and identify antiviral molecules. Potential antiviral roles of several DExD/H box helicase family members were investigated using the ICW assay, and the results indicated that DDX21 and DDX60 reinforced IFN responses and exerted anti-hantaviral effects, whereas DDX50 probably promoted HTNV replication. Additionally, the ICW assay was also applied to assess NAb titers in patients and vaccine recipients. Patients with prompt production of NAbs tended to have favorable disease outcomes. Modest NAb titers were found in vaccinees, indicating that current vaccines still require improvements as they cannot prime host humoral immunity with high efficiency. Taken together, our results indicate that the use of the ICW assay to evaluate non-CPE Hantaan Virus titer demonstrates a significant improvement over current infectivity approaches and a novel technique to screen antiviral molecules and detect NAb efficacies.

  • Novel Identified HLA-A*0201-Restricted Hantaan Virus Glycoprotein Cytotoxic T-Cell Epitopes Could Effectively Induce Protective Responses in HLA-A2.1/Kb Transgenic Mice May Associate with the Severity of Hemorrhagic Fever with Renal Syndrome
    Frontiers Media S.A., 2017
    Co-Authors: Kang Tang, Linfeng Cheng, Chunmei Zhang, Yusi Zhang, Xuyang Zheng, Yun Zhang, Ran Zhuang, Boquan Jin, Fanglin Zhang
    Abstract:

    Hantaan Virus (HTNV) infections can cause severe hemorrhagic fever with renal syndrome (HFRS) in humans, which is associated with high fatality rates. Cytotoxic T cell (CTL) responses contribute to Virus elimination; however, to date, HLA class I allele-restricted HTNV glycoprotein (GP) epitopes recognized by CTLs have not been reported, limiting our understanding of CTL responses against HTNV infection in humans. In this study, 34 HTNV GP nine-mer epitopes that may bind to HLA-A*0201 molecules were predicted using the BIMAS and SYFPEITHI database. Seven of the epitopes were demonstrated to bind to HLA-A*0201 molecules with high affinity via the T2 cell binding assay and were successfully used to synthesize peptide/HLA-A*0201 tetramers. The results of tetramer staining showed that the frequencies of each epitope-specific CTL were higher in patients with milder HFRS, which indicated that the epitopes may induce protective CTL responses after HTNV infection. IFN-γ-enzyme-linked immunospot analysis further confirmed the immunoreactivity of epitopes by eliciting epitope-specific IFN-γ-producing CTL responses. In an HTNV challenge trial, significant inhibition of HTNV replication characterized by lower levels of antigens and RNA loads was observed in major target organs (liver, spleen, and kidneys) of HLA-A2.1/Kb transgenic mice pre-vaccinated with nonapeptides VV9 (aa8–aa16, VMASLVWPV), SL9 (aa996–aa1004, SLTECPTFL) and LL9 (aa358–aa366, LIWTGMIDL). Importantly, LL9 exhibited the best ability to induce protective CTL responses and showed a prominent effect on the kidneys, potentially preventing kidney injury after HTNV infection. Taken together, our results highlight that HTNV GP-derived HLA-A*0201-restricted epitopes could elicit protective CTL responses against the Virus, and that epitope LL9 functions as an immunodominant protective epitope that may advance the design of safe and effective CTL-based HTNV peptide vaccines for humans

  • Incorporation of GM-CSF or CD40L Enhances the Immunogenicity of Hantaan Virus-Like Particles.
    Frontiers in cellular and infection microbiology, 2016
    Co-Authors: Linfeng Cheng, Fang Wang, Liang Zhang, Ziyu Liu, Ying Qikang, Fanglin Zhang
    Abstract:

    A safe and effective Hantaan Virus (HTNV) vaccine is highly desirable because HTNV causes an acute and often fatal disease (hemorrhagic fever with renal syndrome, HFRS). Since the immunity of the inactivated vaccine is weak and the safety is poor, HTNV Virus-like particles (VLPs) offer an attractive and safe alternative. These particles lack the viral genome but are perceived by the immune system as Virus particles. We hypothesized that adding immunostimulatory signals to VLPs would enhance their efficacy. To accomplish this enhancement, we generated chimeric HTNV VLPs containing glycosylphosphatidylinositol (GPI)-anchored granulocyte macrophage colony-stimulating factor (GM-CSF) or CD40 ligand (CD40L) and investigated their biological activity in vitro. The immunization of mice with chimeric HTNV VLPs containing GM-CSF or CD40L induced stronger humoral immune responses and cellular immune responses compared to the HTNV VLPs and Chinese commercial inactivated hantaVirus vaccine. Chimeric HTNV VLPs containing GM-CSF or CD40L also protected mice from an HTNV challenge. Altogether, our results suggest that anchoring immunostimulatory molecules into HTNV VLPs can be a potential approach for the control and prevention of HFRS.

  • Structure and Function of HLA-A*02-Restricted Hantaan Virus Cytotoxic T Cell Epitope that Mediates Effective Protective Responses in HLA-A2.1/Kb Transgenic Mice
    Frontiers Media S.A., 2016
    Co-Authors: Linfeng Cheng, Kang Tang, Chunmei Zhang, Yusi Zhang, Yun Zhang, Ran Zhuang, Bin Yuan, Lihua Chen, Kun Yang
    Abstract:

    HantaVirus infections cause severe emerging diseases in humans and are associated with high mortality rates; therefore, they have become a global public health concern. Our previous study showed that the CD8+ T-cell epitope aa129-aa137 (FVVPILLKA, FA9) of the Hantaan Virus (HTNV) nucleoprotein (NP), restricted by human leukocyte antigen (HLA)-A*02, induced specific CD8+ T-cell responses that controlled HTNV infection in humans. However, the in vivo immunogenicity of peptide FA9 and the effect of FA9-specific CD8+ T-cell immunity remain unclear. Here, based on a detailed structural analysis of the peptide FA9/HLA-A*0201 complex and functional investigations using HLA-A2.1/Kb transgenic mice, we found that the overall structure of the peptide FA9/HLA-A*0201 complex displayed a typical MHC class I fold with Val2 and Ala9 as primary anchor residues and Val3 and Leu7 as secondary anchor residues that allow peptide FA9 to bind tightly with an HLA-A*0201 molecule. Residues in the middle portion of peptide FA9 extruding out of the binding groove may be the sites that allow for recognition by T cell receptors. Immunization with peptide FA9 in HLA-A2.1/Kb transgenic mice induced FA9-specific cytotoxic T cell responses characterized by the induction of high expression levels of IFN-γ, TNF-α, granzyme B, and CD107a. In an HTNV challenge trial, significant reductions in the levels of both the antigens and the HTNV RNA loads were observed in the liver, spleen and kidneys of transgenic mice pre-vaccinated with peptide FA9. Thus, our findings highlight the ability of HTNV epitope-specific CD8+ T-cell immunity to control HTNV and support the possibility that the HTNV-NP FA9 peptide, naturally processed in vivo in an HLA-A*02-restriction manner, may be a good candidate for the development HTNV peptide vaccines

Jay W Hooper - One of the best experts on this subject based on the ideXlab platform.

  • mixing of m segment dna vaccines to Hantaan Virus and puumala Virus reduces their immunogenicity in hamsters
    Vaccine, 2008
    Co-Authors: Kristin Spik, Catherine V Badger, Iacob Mathiessen, Torunn Elisabeth Tjelle, Jay W Hooper, Connie S. Schmaljohn
    Abstract:

    To determine if DNA vaccines for two hantaViruses causing hemorrhagic fever with renal syndrome, Hantaan Virus and Puumala Virus, are immunogenic when given in combination, we delivered them to hamsters separately or as mixtures by gene gun or by electroporation. Both vaccines elicited neutralizing antibodies when given alone but when they were delivered as a mixture, antibodies to only one of the two hantaViruses could be detected. In contrast, if the DNAs were given as separate vaccinations to a single animal, responses to both were observed. These studies suggest that the two DNA vaccines will need to be given as separate administrations.

  • immunogenicity of combination dna vaccines for rift valley fever Virus tick borne encephalitis Virus Hantaan Virus and crimean congo hemorrhagic fever Virus
    Vaccine, 2006
    Co-Authors: Kristin Spik, Jay W Hooper, Amy C Shurtleff, Anita K Mcelroy, Mary C Guttieri, Connie S. Schmaljohn
    Abstract:

    DNA vaccines for Rift Valley fever Virus (RVFV), Crimean Congo hemorrhagic fever Virus (CCHFV), tick-borne encephalitis Virus (TBEV), and Hantaan Virus (HTNV), were tested in mice alone or in various combinations. The bunyaVirus vaccines (RVFV, CCHFV, and HTNV) expressed Gn and Gc genes, and the flaviVirus vaccine (TBEV) expressed the preM and E genes. All vaccines were delivered by gene gun. The TBEV DNA vaccine and the RVFV DNA vaccine elicited similar levels of antibodies and protected mice from challenge when delivered alone or in combination with other DNAs. Although in general, the HTNV and CCHFV DNA vaccines were not very immunogenic in mice, there were no major differences in performance when given alone or in combination with the other vaccines.

  • dna vaccination with the Hantaan Virus m gene protects hamsters against three of four hfrs hantaViruses and elicits a high titer neutralizing antibody response in rhesus monkeys
    Journal of Virology, 2001
    Co-Authors: Jay W Hooper, David Custer, E A Thompson, Connie S. Schmaljohn
    Abstract:

    Four hantaViruses-Hantaan Virus (HTNV), Seoul Virus (SEOV), Dobrava Virus (DOBV) and Puumala Virus-are known to cause hemorrhagic fever with renal syndrome (HFRS) in Europe and Asia. HTNV causes the most severe form of HFRS (5 to 15% case-fatality rate) and afflicts tens of thousands of people annually. Previously, we demonstrated that DNA vaccination with a plasmid expressing the SEOV M gene elicited neutralizing antibodies and protected hamsters against infection with SEOV and HTNV. Here, we report the construction and evaluation of a DNA vaccine that expresses the HTNV M gene products, G1 and G2. DNA vaccination of hamsters with the HTNV M gene conferred sterile protection against infection with HTNV, SEOV, and DOBV. DNA vaccination of rhesus monkeys with either the SEOV or HTNV M gene elicited high levels of neutralizing antibodies. These are the first immunogenicity data for hantaVirus DNA vaccines in nonhuman primates. Because a neutralizing antibody response is considered a surrogate marker for protective immunity in humans, our protection data in hamsters combined with the immunogenicity data in monkeys suggest that hantaVirus M gene-based DNA vaccines could protect humans against the most severe forms of HFRS.

  • comparison of the protective efficacy of naked dna dna based sindbis replicon and packaged sindbis replicon vectors expressing hantaVirus structural genes in hamsters
    Virology, 1999
    Co-Authors: K I Kamrud, Jay W Hooper, Fredrik Elgh, Connie S. Schmaljohn
    Abstract:

    Seoul Virus (SEOV) is a member of the HantaVirus genus (family Bunyaviridae) and an etiological agent of hemorrhagic fever with renal syndrome. The medium (M) and small (S) gene segments of SEOV encode the viral envelope glycoproteins and nucleocapsid protein, respectively. We compared the immunogenicity and protective efficacy of naked DNA (pWRG7077), DNA-based Sindbis replicon (pSIN2.5), and packaged Sindbis replicon vectors (pSINrep5), containing either the M or S gene segment of SEOV in Syrian hamsters. All of the vectors elicited an anti-SEOV immune response to the expressed SEOV gene products. Vaccinated hamsters were challenged with SEOV and monitored for evidence of infection. Protection from infection was strongly associated with M-gene vaccination. A small number of S-gene-vaccinated animals also were protected. Hamsters vaccinated with the pWRG7077 vector expressing the M gene demonstrated the most consistent protection from SEOV infection and also were protected from heterologous hantaVirus (Hantaan Virus) infection.

Jiro Arikawa - One of the best experts on this subject based on the ideXlab platform.

  • Application of truncated nucleocapsid protein (N) for serotyping ELISA of murinae-associated hantaVirus infection in rats.
    Journal of Veterinary Medical Science, 2011
    Co-Authors: Shumpei P Yasuda, Kumiko Yoshimatsu, Takaaki Koma, Kenta Shimizu, Rika Endo, Rie Isozumi, Jiro Arikawa
    Abstract:

    Truncated recombinant nucleocapsid proteins (trNs) that lack N-terminally located cross-reactive epitopes of four Murinae rodent-associated hantaViruses, Seoul Virus (SEOV), Thailand Virus, Hantaan Virus (HTNV) and Dobrava-Belgrade Virus, were produced by using a baculoVirus expression system. ELISA with the trNs as antigens enabled serotyping of immune sera from rats experimentally inoculated with the corresponding hantaViruses with cut-off OD values of 60% of those of whole N of HTNV. The trN-based ELISA could serotype 12 out of 13 sera obtained from wild rodents (Rattus norvegicus) naturally infected with SEOV using the 60% cut-off value. These results indicate that screening with whole N followed by serotyping with trNs using a cut-off OD value of 60% of that of whole N is a useful method for serological surveillance of Murinae-associated hantaVirus infection among rodents.

  • the intracellular association of the nucleocapsid protein np of Hantaan Virus htnv with small ubiquitin like modifier 1 sumo 1 conjugating enzyme 9 ubc9
    Virology, 2003
    Co-Authors: Akihiko Maeda, Kumiko Yoshimatsu, Jiro Arikawa, Byounghee Lee, Masayuki Saijo, Ichiro Kurane, Shigeru Morikawa
    Abstract:

    Small ubiquitin-like modifier-1 (SUMO-1) conjugating enzyme 9 (Ubc9) conjugates SUMO-1 to target proteins and modulates cellular processes such as signal transduction, transcription regulation, and cell growth regulation. We demonstrated here that the nucleocapsid protein (NP) of Hantaan Virus (HTNV) was associated with Ubc9 and SUMO-1 in vivo. Analysis of the interaction between the truncated NPs and Ubc9 revealed that the amino acid residues at the positions between 101 and 238 in the NP were responsible for the interaction. Furthermore, a consensus binding motif of Ubc9 and SUMO-1, MKAE, within this region, especially the second amino acid of the motif, K residue, was crucial for the interaction, and the interaction was essential for the NP to be localized in the perinuclear region. These results indicate that the assembly of the HTNV-NP is regulated by the interaction between the NP and Ubc9. This is the first report to demonstrate the interaction of Ubc9 with a structural protein of negative-strand RNA Viruses.

  • the multimerization of hantaVirus nucleocapsid protein depends on type specific epitopes
    Journal of Virology, 2003
    Co-Authors: Kumiko Yoshimatsu, Koichi Araki, Michiko Ogino, Hideki Ebihara, Masami Morimatsu, Jiro Arikawa
    Abstract:

    Multimerization of the Hantaan Virus nucleocapsid protein (NP) in Hantaan Virus-infected Vero E6 cells was observed in a competitive enzyme-linked immunosorbent assay (ELISA). Recombinant and truncated NPs of Hantaan, Seoul, and Dobrava Viruses lacking the N-terminal 49 amino acids were also detected as multimers. Although truncated NPs of Hantaan Virus lacking the N-terminal 154 amino acids existed as a monomer, those of Seoul and Dobrava formed multimers. The multimerized truncated NP antigens of Seoul and Dobrava Viruses could detect serotype-specific antibodies, whereas the monomeric truncated NP antigen of Hantaan Virus lacking the N-terminal 154 amino acids could not, suggesting that a hantaVirus serotype-specific epitope on the NP results in multimerization. The NP-NP interaction was also detected by using a yeast two-hybrid assay. Two regions, amino acids 100 to 125 (region 1) and amino acids 404 to 429 (region 2), were essential for the NP-NP interaction in yeast. The NP of Seoul Virus in which the tryptophan at amino acid number 119 was replaced by alanine (W119A mutation) did not multimerize in the yeast two-hybrid assay, indicating that tryptophan 119 in region 1 is important for the NP-NP interaction in yeast. However, W119A mutants expressed in mammalian cells were detected as the multimer by using competitive ELISA. Similarly, the truncated NP of Seoul Virus expressing amino acids 155 to 429 showed a homologous interaction in a competitive ELISA but not in the yeast two-hybrid assay, indicating that the C-terminal region is important for the multimerization detected by competitive ELISA. Combined, the results indicate that several steps and regions are involved in multimerization of hantaVirus NP.

  • truncated hantaVirus nucleocapsid proteins for serotyping Hantaan seoul and dobrava hantaVirus infections
    Journal of Clinical Microbiology, 2001
    Co-Authors: Koichi Araki, Åke Lundkvist, Michiko Ogino, Hideki Ebihara, Hiroaki Kariwa, Ikuo Takashima, Jiro Arikawa
    Abstract:

    Truncated recombinant nucleocapsid proteins (rNPs) of Hantaan Virus (HTNV), Seoul Virus (SEOV), and Dobrava Virus (DOBV) were expressed by a baculoVirus system. The truncated rNPs, which lacked 49 (rNP50) or 154 (rNP155) N-terminal amino acids of the NPs of HTNV, SEOV, and DOBV, were able to differentiate HTNV-, SEOV-, and DOBV-specific immune sera. Recombinant NP50s retained higher reactivities than rNP155s and were proven useful for enzyme-linked immunosorbent assay (ELISA). The ELISAs based on the rNP50s of HTNV, SEOV, and DOBV successfully differentiated three groups of patient sera, previously defined by neutralization tests: 17 with HTNV infection, 12 with SEOV infection, and 20 with DOBV infection. The entire rNP of Puumala Virus (PUUV) distinguished PUUV infection from the other types of hantaVirus infection. Serotyping with these rNP50s can be recommended as a rapid and efficient system for hantaVirus diagnosis.

  • Protective immunity of Hantaan Virus nucleocapsid and envelope protein studied using baculoVirus-expressed proteins
    Archives of Virology, 1993
    Co-Authors: Kumiko Yoshimatsu, Chiaki Ishihara, Yung Choon Yoo, Ryu Yoshida, Ichiro Azuma, Jiro Arikawa
    Abstract:

    Recombinant Hantaan Virus nucleocapsid protein (rNP) and recombinant envelope (rEnv) proteins were prepared using a baculoVirus expression system to examine the role of Hantaan Virus structural proteins in protective immunity. Passive transfer of spleen cells from mice immunized with rNP conferred partial protection or prolongation of time to death from fatal Hantaan Virus infection in suckling mice which were challenged with Hantaan Virus at 40 LD50 (survival rate: 43%) or 4 LD50 (survival rate: 43%). The T cell-enriched fraction protected one mouse from lethal infection but the B cell-enriched fraction had no such effect on fatal HTN infection. The protective effects of the antibody against HTN challenge were examined by passive immunization. The monoclonal antibody ECO 2 directed to NP also conferred partial survival and significant difference in time to death. Although rEnv antigen failed to induce neutralizing antibody, both immune spleen cells and immune serum to rEnv conferred partial protection upon suckling mice. These results indicate that both nucleocapsid and envelope proteins of Hantaan Virus were responsible for induction of cell mediated protective immunity. Vero E 6 cells infected with Hantaan Virus expressed envelope protein on the surface, as determined by flow cytometry. However, there was only negligible expression of nucleocapsid protein.