Machupo 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 459 Experts worldwide ranked by ideXlab platform

Michael Patterson - One of the best experts on this subject based on the ideXlab platform.

  • glycoprotein n linked glycans play a critical role in arenaVirus pathogenicity
    PLOS Pathogens, 2021
    Co-Authors: Takaaki Koma, Steven Hallam, John T. Manning, Junki Maruyama, Cheng Huang, Milagros Miller, Jeanon N Smith, Adrian Coscia, Aida G Walker, Michael Patterson
    Abstract:

    Several arenaViruses cause hemorrhagic fevers in humans with high case fatality rates. A vaccine named Candid#1 is available only against Junin Virus (JUNV) in Argentina. Specific N-linked glycans on the arenaVirus surface glycoprotein (GP) mask important epitopes and help the Virus evade antibody responses. However the role of GPC glycans in arenaVirus pathogenicity is largely unclear. In a lethal animal model of hemorrhagic fever-causing Machupo Virus (MACV) infection, we found that a chimeric MACV with the ectodomain of GPC from Candid#1 vaccine was partially attenuated. Interestingly, mutations resulting in acquisition of N-linked glycans at GPC N83 and N166 frequently occurred in late stages of the infection. These glycosylation sites are conserved in the GPC of wild-type MACV, indicating that this is a phenotypic reversion for the chimeric MACV to gain those glycans crucial for infection in vivo. Further studies indicated that the GPC mutant Viruses with additional glycans became more resistant to neutralizing antibodies and more virulent in animals. On the other hand, disruption of these glycosylation sites on wild-type MACV GPC rendered the Virus substantially attenuated in vivo and also more susceptible to antibody neutralization, while loss of these glycans did not affect Virus growth in cultured cells. We also found that MACV lacking specific GPC glycans elicited higher levels of neutralizing antibodies against wild-type MACV. Our findings revealed the critical role of specific glycans on GPC in arenaVirus pathogenicity and have important implications for rational design of vaccines against this group of hemorrhagic fever-causing Viruses.

  • Rescue of a Recombinant Machupo Virus from Cloned cDNAs and In Vivo Characterization in Interferon (/) Receptor Double Knockout Mice
    2016
    Co-Authors: Michael Patterson, Alexey Seregin, Cheng Huang, Milagros Miller, Olga A. Kolokoltsova, Allison Poussard, Jennifer Smith, Jeanon Smith, Nadezhda Yun, Ashley Grant
    Abstract:

    Machupo Virus (MACV) is the etiological agent of Bolivian hemorrhagic fever (BHF), a reemerging and neglected tropical dis-ease associated with highmortality. The prototypical strain of MACV, Carvallo, was isolated from a human patient in 1963, but minimal in vitro and in vivo characterization has been reported. To this end, we utilized reverse genetics to rescue a pathogenic MACV from cloned cDNAs. The recombinant MACV (rMACV) had in vitro growth properties similar to those of the parental MACV. Both Viruses caused similar disease development in alpha/beta and gamma interferon receptor knockout mice, including neurological disease development and highmortality. In addition, we have identified a novel murine model with mortality and neurological disease similar to BHF disease reported in humans and nonhuman primates. Amember of the family Arenaviridae, Machupo Virus (MACV)is an enveloped, bisegmented negative-stranded RNA Virus. ArenaViruses utilize an ambisense coding strategy to direct viral gene transcription from two genomic segments, the large segment (L, ca. 7.2 kb) and the small segment (S, ca. 3.3 kb). Each segment carries two viral genes; S encodes the glycoprotein precursor (GPC) and the nucleoprotein (NP), while L encodes the RNA-dependent RNA polymerase (L polymerase) and the small RING finger protein Z. The GPC is posttranslationally cleaved by th

  • the ectodomain of glycoprotein from the candid 1 vaccine strain of junin Virus rendered Machupo Virus partially attenuated in mice lacking ifn αβ γ receptor
    PLOS Neglected Tropical Diseases, 2016
    Co-Authors: Takaaki Koma, Cheng Huang, Milagros Miller, Michael Patterson
    Abstract:

    Machupo Virus (MACV), a New World arenaVirus, is the etiological agent of Bolivian hemorrhagic fever (BHF). Junin Virus (JUNV), a close relative, causes Argentine hemorrhagic fever (AHF). Previously, we reported that a recombinant, chimeric MACV (rMACV/Cd#1-GPC) expressing glycoprotein from the Candid#1 (Cd#1) vaccine strain of JUNV is completely attenuated in a murine model and protects animals from lethal challenge with MACV. A rMACV with a single F438I substitution in the transmembrane domain (TMD) of GPC, which is equivalent to the F427I attenuating mutation in Cd#1 GPC, was attenuated in a murine model but genetically unstable. In addition, the TMD mutation alone was not sufficient to fully attenuate JUNV, indicating that other domains of the GPC may also contribute to the attenuation. To investigate the requirement of different domains of Cd#1 GPC for successful attenuation of MACV, we rescued several rMACVs expressing the ectodomain of GPC from Cd#1 either alone (MCg1), along with the TMD F438I substitution (MCg2), or with the TMD of Cd#1 (MCg3). All rMACVs exhibited similar growth curves in cultured cells. In mice, the MCg1 displayed significant reduction in lethality as compared with rMACV. The MCg1 was detected in brains and spleens of MCg1-infected mice and the infection was associated with tissue inflammation. On the other hand, all animals survived MCg2 and MCg3 infection without detectable levels of Virus in various organs while producing neutralizing antibody against Cd#1. Overall our data suggest the indispensable role of each GPC domain in the full attenuation and immunogenicity of rMACV/Cd#1 GPC.

  • Machupo Virus expressing gpc of the candid 1 vaccine strain of junin Virus is highly attenuated and immunogenic
    Journal of Virology, 2016
    Co-Authors: Takaaki Koma, Alexey Seregin, Cheng Huang, Milagros Miller, Michael Patterson, Payal D Maharaj, Steven Hallam
    Abstract:

    ABSTRACT Machupo Virus (MACV) is the causative agent of Bolivian hemorrhagic fever. Our previous study demonstrated that a MACV strain with a single amino acid substitution (F438I) in the transmembrane domain of glycoprotein is attenuated but genetically unstable in mice. MACV is closely related to Junin Virus (JUNV), the causative agent of Argentine hemorrhagic fever. Others and our group have identified the glycoprotein to be the major viral factor determining JUNV attenuation. In this study, we tested the compatibility of the glycoprotein of the Candid#1 live-attenuated vaccine strain of JUNV in MACV replication and its ability to attenuate MACV in vivo . Recombinant MACV with the Candid#1 glycoprotein (rMACV/Cd#1-GPC) exhibited growth properties similar to those of Candid#1 and was genetically stable in vitro . In a mouse model of lethal infection, rMACV/Cd#1-GPC was fully attenuated, more immunogenic than Candid#1, and fully protective against MACV infection. Therefore, the MACV strain expressing the glycoprotein of Candid#1 is safe, genetically stable, and highly protective against MACV infection in a mouse model. IMPORTANCE Currently, there are no FDA-approved vaccines and/or treatments for Bolivian hemorrhagic fever, which is a fatal human disease caused by MACV. The development of antiviral strategies to combat viral hemorrhagic fevers, including Bolivian hemorrhagic fever, is one of the top priorities of the Implementation Plan of the U.S. Department of Health and Human Services Public Health Emergency Medical Countermeasures Enterprise. Here, we demonstrate for the first time that MACV expressing glycoprotein of Candid#1 is a safe, genetically stable, highly immunogenic, and protective vaccine candidate against Bolivian hemorrhagic fever.

  • The Ectodomain of Glycoprotein from the Candid#1 Vaccine Strain of Junin Virus Rendered Machupo Virus Partially Attenuated in Mice Lacking IFN-αβ/γ Receptor.
    Public Library of Science (PLoS), 2016
    Co-Authors: Takaaki Koma, Cheng Huang, Milagros Miller, Michael Patterson
    Abstract:

    Machupo Virus (MACV), a New World arenaVirus, is the etiological agent of Bolivian hemorrhagic fever (BHF). Junin Virus (JUNV), a close relative, causes Argentine hemorrhagic fever (AHF). Previously, we reported that a recombinant, chimeric MACV (rMACV/Cd#1-GPC) expressing glycoprotein from the Candid#1 (Cd#1) vaccine strain of JUNV is completely attenuated in a murine model and protects animals from lethal challenge with MACV. A rMACV with a single F438I substitution in the transmembrane domain (TMD) of GPC, which is equivalent to the F427I attenuating mutation in Cd#1 GPC, was attenuated in a murine model but genetically unstable. In addition, the TMD mutation alone was not sufficient to fully attenuate JUNV, indicating that other domains of the GPC may also contribute to the attenuation. To investigate the requirement of different domains of Cd#1 GPC for successful attenuation of MACV, we rescued several rMACVs expressing the ectodomain of GPC from Cd#1 either alone (MCg1), along with the TMD F438I substitution (MCg2), or with the TMD of Cd#1 (MCg3). All rMACVs exhibited similar growth curves in cultured cells. In mice, the MCg1 displayed significant reduction in lethality as compared with rMACV. The MCg1 was detected in brains and spleens of MCg1-infected mice and the infection was associated with tissue inflammation. On the other hand, all animals survived MCg2 and MCg3 infection without detectable levels of Virus in various organs while producing neutralizing antibody against Cd#1. Overall our data suggest the indispensable role of each GPC domain in the full attenuation and immunogenicity of rMACV/Cd#1 GPC

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

  • glycoprotein n linked glycans play a critical role in arenaVirus pathogenicity
    PLOS Pathogens, 2021
    Co-Authors: Takaaki Koma, Steven Hallam, John T. Manning, Junki Maruyama, Cheng Huang, Milagros Miller, Jeanon N Smith, Adrian Coscia, Aida G Walker, Michael Patterson
    Abstract:

    Several arenaViruses cause hemorrhagic fevers in humans with high case fatality rates. A vaccine named Candid#1 is available only against Junin Virus (JUNV) in Argentina. Specific N-linked glycans on the arenaVirus surface glycoprotein (GP) mask important epitopes and help the Virus evade antibody responses. However the role of GPC glycans in arenaVirus pathogenicity is largely unclear. In a lethal animal model of hemorrhagic fever-causing Machupo Virus (MACV) infection, we found that a chimeric MACV with the ectodomain of GPC from Candid#1 vaccine was partially attenuated. Interestingly, mutations resulting in acquisition of N-linked glycans at GPC N83 and N166 frequently occurred in late stages of the infection. These glycosylation sites are conserved in the GPC of wild-type MACV, indicating that this is a phenotypic reversion for the chimeric MACV to gain those glycans crucial for infection in vivo. Further studies indicated that the GPC mutant Viruses with additional glycans became more resistant to neutralizing antibodies and more virulent in animals. On the other hand, disruption of these glycosylation sites on wild-type MACV GPC rendered the Virus substantially attenuated in vivo and also more susceptible to antibody neutralization, while loss of these glycans did not affect Virus growth in cultured cells. We also found that MACV lacking specific GPC glycans elicited higher levels of neutralizing antibodies against wild-type MACV. Our findings revealed the critical role of specific glycans on GPC in arenaVirus pathogenicity and have important implications for rational design of vaccines against this group of hemorrhagic fever-causing Viruses.

  • A single mutation (V64G) within the RING Domain of Z attenuates Junin Virus.
    PLOS Neglected Tropical Diseases, 2020
    Co-Authors: Steven Hallam, John T. Manning, Junki Maruyama, Alexey Seregin, Cheng Huang, David H. Walker
    Abstract:

    Junin Virus (JUNV) is a New World arenaVirus that is the causative agent of Argentine hemorrhagic fever (AHF). Candid#1 (Can) is a live-attenuated vaccine strain of JUNV that since its introduction has resulted in a marked decrease in AHF incidence within the endemic regions of the Pampas in Argentina. Originally, the viral determinants and mechanisms of Can attenuation were not well understood. Recent work has identified the glycoprotein as the major attenuating factor for Can. The establishment of attenuating strategies based on any of the other viral proteins, however, has not been pursued. Here, we document the role of Can Z resulting in incompatibilities with wild type JUNV that results in decreased growth in vitro. In addition, this incompatibility results in attenuation of the Virus in the guinea pig model. Further, we identify a single mutation (V64G) in the Z protein that is able to confer this demonstrated attenuation. By establishing and characterizing a novel attenuation strategy for New World mammarenaViruses, we hope to aid future vaccine development for related emerging pathogens including Machupo Virus (MACV), Guanarito Virus (GTOV), and Sabia Virus (SABV).

  • Monoclonal Antibodies with Neutralizing Activity and Fc-Effector Functions against the Machupo Virus Glycoprotein.
    Journal of virology, 2020
    Co-Authors: Fatima Amanat, Cheng Huang, James Duehr, Gene S. Tan, Florian Krammer
    Abstract:

    Machupo Virus (MACV), the causative agent of Bolivian hemorrhagic fever (BHF), is a New World arenaVirus that was first isolated in Bolivia from a human spleen in 1963. Due to the lack of a specific vaccine or therapy, this Virus is considered a major risk to public health and is classified as a category A priority pathogen by the U.S. National Institutes of Health. In this study, we used DNA vaccination against the MACV glycoprotein precursor complex (GPC) and murine hybridoma technology to generate 25 mouse monoclonal antibodies (MAbs) against the GPC of MACV. Out of 25 MAbs, five were found to have potent neutralization activity in vitro against a recombinant vesicular stomatitis Virus expressing MACV GPC (VSV-MACV) as well as against authentic MACV. Furthermore, the five neutralizing MAbs exhibited strong antibody-dependent cellular cytotoxicity (ADCC) activity in a reporter assay. When tested in vivo using VSV-MACV in a Stat2-/- mouse model, three MAbs significantly lowered viral loads in the spleen. Our work provides valuable insights into epitopes targeted by neutralizing antibodies that could be potent targets for vaccines and therapeutics and shed light on the importance of effector functions in immunity against MACV.IMPORTANCE MACV infections are a significant public health concern and lead to high case fatality rates. No specific treatment or vaccine for MACV infections exist. However, cases of Junin Virus infection, a related Virus, can be treated with convalescent-phase serum. This indicates that a MAb-based therapy for MACV could be effective. Here, we describe several MAbs that neutralize MACV and could be used for this purpose.

  • Highly Pathogenic New World and Old World Human ArenaViruses Induce Distinct Interferon Responses in Human Cells
    2016
    Co-Authors: Cheng Huang, Takaaki Koma, Nadezhda E. Yun, Olga A. Kolokoltsova, Alexey V. Seregin, Shannon Ronca, Slobodan Paessler
    Abstract:

    The arenaVirus family includes several important pathogens that cause severe and sometimes fatal diseases in humans. The highly pathogenic OldWorld (OW) arenaVirus Lassa fever Virus (LASV) is the causative agent of Lassa fever (LF) disease in hu-mans. LASV infections in severe cases are generally immunosuppressive without stimulating interferon (IFN) induction, a pro-inflammatory response, or T cell activation. However, the host innate immune responses to highly pathogenic NewWorld (NW) arenaViruses are not well understood.We have previously shown that the highly pathogenic NW arenaVirus, Junin Virus (JUNV), induced an IFN response in human A549 cells. Here, we report that Machupo Virus (MACV), another highly pathogenic NW are-naVirus, also induces an IFN response. Importantly, both pathogenic NW arenaViruses, in contrast to the OWhighly pathogenic arenaVirus LASV, readily elicited an IFN response in human primary dendritic cells and A549 cells. Coinfection experiments revealed that LASV could potently inhibit MACV-activated IFN responses even at 6 h after MACV infection, while the replica-tion levels of MACV and LASV were not affected by Virus coinfection. Our results clearly demonstrated that although all Viruses studied herein are highly pathogenic to humans, the host IFN responses toward infections with the NW arenaViruses JUNV and MACV are quite different from responses to infections with the OW arenaVirus LASV, a discovery that needs to be further inves-tigated in relevant animal models. This finding might help us better understand various interplays between the host immune system and highly pathogenic arenaViruses as well as distinct mechanisms underlying viral pathogenesis

  • Rescue of a Recombinant Machupo Virus from Cloned cDNAs and In Vivo Characterization in Interferon (/) Receptor Double Knockout Mice
    2016
    Co-Authors: Michael Patterson, Alexey Seregin, Cheng Huang, Milagros Miller, Olga A. Kolokoltsova, Allison Poussard, Jennifer Smith, Jeanon Smith, Nadezhda Yun, Ashley Grant
    Abstract:

    Machupo Virus (MACV) is the etiological agent of Bolivian hemorrhagic fever (BHF), a reemerging and neglected tropical dis-ease associated with highmortality. The prototypical strain of MACV, Carvallo, was isolated from a human patient in 1963, but minimal in vitro and in vivo characterization has been reported. To this end, we utilized reverse genetics to rescue a pathogenic MACV from cloned cDNAs. The recombinant MACV (rMACV) had in vitro growth properties similar to those of the parental MACV. Both Viruses caused similar disease development in alpha/beta and gamma interferon receptor knockout mice, including neurological disease development and highmortality. In addition, we have identified a novel murine model with mortality and neurological disease similar to BHF disease reported in humans and nonhuman primates. Amember of the family Arenaviridae, Machupo Virus (MACV)is an enveloped, bisegmented negative-stranded RNA Virus. ArenaViruses utilize an ambisense coding strategy to direct viral gene transcription from two genomic segments, the large segment (L, ca. 7.2 kb) and the small segment (S, ca. 3.3 kb). Each segment carries two viral genes; S encodes the glycoprotein precursor (GPC) and the nucleoprotein (NP), while L encodes the RNA-dependent RNA polymerase (L polymerase) and the small RING finger protein Z. The GPC is posttranslationally cleaved by th

Takaaki Koma - One of the best experts on this subject based on the ideXlab platform.

  • glycoprotein n linked glycans play a critical role in arenaVirus pathogenicity
    PLOS Pathogens, 2021
    Co-Authors: Takaaki Koma, Steven Hallam, John T. Manning, Junki Maruyama, Cheng Huang, Milagros Miller, Jeanon N Smith, Adrian Coscia, Aida G Walker, Michael Patterson
    Abstract:

    Several arenaViruses cause hemorrhagic fevers in humans with high case fatality rates. A vaccine named Candid#1 is available only against Junin Virus (JUNV) in Argentina. Specific N-linked glycans on the arenaVirus surface glycoprotein (GP) mask important epitopes and help the Virus evade antibody responses. However the role of GPC glycans in arenaVirus pathogenicity is largely unclear. In a lethal animal model of hemorrhagic fever-causing Machupo Virus (MACV) infection, we found that a chimeric MACV with the ectodomain of GPC from Candid#1 vaccine was partially attenuated. Interestingly, mutations resulting in acquisition of N-linked glycans at GPC N83 and N166 frequently occurred in late stages of the infection. These glycosylation sites are conserved in the GPC of wild-type MACV, indicating that this is a phenotypic reversion for the chimeric MACV to gain those glycans crucial for infection in vivo. Further studies indicated that the GPC mutant Viruses with additional glycans became more resistant to neutralizing antibodies and more virulent in animals. On the other hand, disruption of these glycosylation sites on wild-type MACV GPC rendered the Virus substantially attenuated in vivo and also more susceptible to antibody neutralization, while loss of these glycans did not affect Virus growth in cultured cells. We also found that MACV lacking specific GPC glycans elicited higher levels of neutralizing antibodies against wild-type MACV. Our findings revealed the critical role of specific glycans on GPC in arenaVirus pathogenicity and have important implications for rational design of vaccines against this group of hemorrhagic fever-causing Viruses.

  • Vaccine-elicited receptor-binding site antibodies neutralize two New World hemorrhagic fever arenaViruses
    Nature Publishing Group, 2018
    Co-Authors: Lars E. Clark, John T. Manning, Takaaki Koma, Selma Mahmutovic, Donald D. Raymond, Taleen Dilanyan, Sundaresh Shankar, Silvana C. Levis, Ana M. Briggiler, Delia A. Enria
    Abstract:

    The five known New World hemorrhagic fever arenaViruses use the same entry receptor, but their viral glycoproteins are markedly different. Here, the authors isolate monoclonal antibodies from a Junin Virus vaccinated person that cross-neutralize Machupo Virus and identify a conserved epitope in the receptor-binding domain

  • Highly Pathogenic New World and Old World Human ArenaViruses Induce Distinct Interferon Responses in Human Cells
    2016
    Co-Authors: Cheng Huang, Takaaki Koma, Nadezhda E. Yun, Olga A. Kolokoltsova, Alexey V. Seregin, Shannon Ronca, Slobodan Paessler
    Abstract:

    The arenaVirus family includes several important pathogens that cause severe and sometimes fatal diseases in humans. The highly pathogenic OldWorld (OW) arenaVirus Lassa fever Virus (LASV) is the causative agent of Lassa fever (LF) disease in hu-mans. LASV infections in severe cases are generally immunosuppressive without stimulating interferon (IFN) induction, a pro-inflammatory response, or T cell activation. However, the host innate immune responses to highly pathogenic NewWorld (NW) arenaViruses are not well understood.We have previously shown that the highly pathogenic NW arenaVirus, Junin Virus (JUNV), induced an IFN response in human A549 cells. Here, we report that Machupo Virus (MACV), another highly pathogenic NW are-naVirus, also induces an IFN response. Importantly, both pathogenic NW arenaViruses, in contrast to the OWhighly pathogenic arenaVirus LASV, readily elicited an IFN response in human primary dendritic cells and A549 cells. Coinfection experiments revealed that LASV could potently inhibit MACV-activated IFN responses even at 6 h after MACV infection, while the replica-tion levels of MACV and LASV were not affected by Virus coinfection. Our results clearly demonstrated that although all Viruses studied herein are highly pathogenic to humans, the host IFN responses toward infections with the NW arenaViruses JUNV and MACV are quite different from responses to infections with the OW arenaVirus LASV, a discovery that needs to be further inves-tigated in relevant animal models. This finding might help us better understand various interplays between the host immune system and highly pathogenic arenaViruses as well as distinct mechanisms underlying viral pathogenesis

  • the ectodomain of glycoprotein from the candid 1 vaccine strain of junin Virus rendered Machupo Virus partially attenuated in mice lacking ifn αβ γ receptor
    PLOS Neglected Tropical Diseases, 2016
    Co-Authors: Takaaki Koma, Cheng Huang, Milagros Miller, Michael Patterson
    Abstract:

    Machupo Virus (MACV), a New World arenaVirus, is the etiological agent of Bolivian hemorrhagic fever (BHF). Junin Virus (JUNV), a close relative, causes Argentine hemorrhagic fever (AHF). Previously, we reported that a recombinant, chimeric MACV (rMACV/Cd#1-GPC) expressing glycoprotein from the Candid#1 (Cd#1) vaccine strain of JUNV is completely attenuated in a murine model and protects animals from lethal challenge with MACV. A rMACV with a single F438I substitution in the transmembrane domain (TMD) of GPC, which is equivalent to the F427I attenuating mutation in Cd#1 GPC, was attenuated in a murine model but genetically unstable. In addition, the TMD mutation alone was not sufficient to fully attenuate JUNV, indicating that other domains of the GPC may also contribute to the attenuation. To investigate the requirement of different domains of Cd#1 GPC for successful attenuation of MACV, we rescued several rMACVs expressing the ectodomain of GPC from Cd#1 either alone (MCg1), along with the TMD F438I substitution (MCg2), or with the TMD of Cd#1 (MCg3). All rMACVs exhibited similar growth curves in cultured cells. In mice, the MCg1 displayed significant reduction in lethality as compared with rMACV. The MCg1 was detected in brains and spleens of MCg1-infected mice and the infection was associated with tissue inflammation. On the other hand, all animals survived MCg2 and MCg3 infection without detectable levels of Virus in various organs while producing neutralizing antibody against Cd#1. Overall our data suggest the indispensable role of each GPC domain in the full attenuation and immunogenicity of rMACV/Cd#1 GPC.

  • Machupo Virus expressing gpc of the candid 1 vaccine strain of junin Virus is highly attenuated and immunogenic
    Journal of Virology, 2016
    Co-Authors: Takaaki Koma, Alexey Seregin, Cheng Huang, Milagros Miller, Michael Patterson, Payal D Maharaj, Steven Hallam
    Abstract:

    ABSTRACT Machupo Virus (MACV) is the causative agent of Bolivian hemorrhagic fever. Our previous study demonstrated that a MACV strain with a single amino acid substitution (F438I) in the transmembrane domain of glycoprotein is attenuated but genetically unstable in mice. MACV is closely related to Junin Virus (JUNV), the causative agent of Argentine hemorrhagic fever. Others and our group have identified the glycoprotein to be the major viral factor determining JUNV attenuation. In this study, we tested the compatibility of the glycoprotein of the Candid#1 live-attenuated vaccine strain of JUNV in MACV replication and its ability to attenuate MACV in vivo . Recombinant MACV with the Candid#1 glycoprotein (rMACV/Cd#1-GPC) exhibited growth properties similar to those of Candid#1 and was genetically stable in vitro . In a mouse model of lethal infection, rMACV/Cd#1-GPC was fully attenuated, more immunogenic than Candid#1, and fully protective against MACV infection. Therefore, the MACV strain expressing the glycoprotein of Candid#1 is safe, genetically stable, and highly protective against MACV infection in a mouse model. IMPORTANCE Currently, there are no FDA-approved vaccines and/or treatments for Bolivian hemorrhagic fever, which is a fatal human disease caused by MACV. The development of antiviral strategies to combat viral hemorrhagic fevers, including Bolivian hemorrhagic fever, is one of the top priorities of the Implementation Plan of the U.S. Department of Health and Human Services Public Health Emergency Medical Countermeasures Enterprise. Here, we demonstrate for the first time that MACV expressing glycoprotein of Candid#1 is a safe, genetically stable, highly immunogenic, and protective vaccine candidate against Bolivian hemorrhagic fever.

Milagros Miller - One of the best experts on this subject based on the ideXlab platform.

  • glycoprotein n linked glycans play a critical role in arenaVirus pathogenicity
    PLOS Pathogens, 2021
    Co-Authors: Takaaki Koma, Steven Hallam, John T. Manning, Junki Maruyama, Cheng Huang, Milagros Miller, Jeanon N Smith, Adrian Coscia, Aida G Walker, Michael Patterson
    Abstract:

    Several arenaViruses cause hemorrhagic fevers in humans with high case fatality rates. A vaccine named Candid#1 is available only against Junin Virus (JUNV) in Argentina. Specific N-linked glycans on the arenaVirus surface glycoprotein (GP) mask important epitopes and help the Virus evade antibody responses. However the role of GPC glycans in arenaVirus pathogenicity is largely unclear. In a lethal animal model of hemorrhagic fever-causing Machupo Virus (MACV) infection, we found that a chimeric MACV with the ectodomain of GPC from Candid#1 vaccine was partially attenuated. Interestingly, mutations resulting in acquisition of N-linked glycans at GPC N83 and N166 frequently occurred in late stages of the infection. These glycosylation sites are conserved in the GPC of wild-type MACV, indicating that this is a phenotypic reversion for the chimeric MACV to gain those glycans crucial for infection in vivo. Further studies indicated that the GPC mutant Viruses with additional glycans became more resistant to neutralizing antibodies and more virulent in animals. On the other hand, disruption of these glycosylation sites on wild-type MACV GPC rendered the Virus substantially attenuated in vivo and also more susceptible to antibody neutralization, while loss of these glycans did not affect Virus growth in cultured cells. We also found that MACV lacking specific GPC glycans elicited higher levels of neutralizing antibodies against wild-type MACV. Our findings revealed the critical role of specific glycans on GPC in arenaVirus pathogenicity and have important implications for rational design of vaccines against this group of hemorrhagic fever-causing Viruses.

  • Rescue of a Recombinant Machupo Virus from Cloned cDNAs and In Vivo Characterization in Interferon (/) Receptor Double Knockout Mice
    2016
    Co-Authors: Michael Patterson, Alexey Seregin, Cheng Huang, Milagros Miller, Olga A. Kolokoltsova, Allison Poussard, Jennifer Smith, Jeanon Smith, Nadezhda Yun, Ashley Grant
    Abstract:

    Machupo Virus (MACV) is the etiological agent of Bolivian hemorrhagic fever (BHF), a reemerging and neglected tropical dis-ease associated with highmortality. The prototypical strain of MACV, Carvallo, was isolated from a human patient in 1963, but minimal in vitro and in vivo characterization has been reported. To this end, we utilized reverse genetics to rescue a pathogenic MACV from cloned cDNAs. The recombinant MACV (rMACV) had in vitro growth properties similar to those of the parental MACV. Both Viruses caused similar disease development in alpha/beta and gamma interferon receptor knockout mice, including neurological disease development and highmortality. In addition, we have identified a novel murine model with mortality and neurological disease similar to BHF disease reported in humans and nonhuman primates. Amember of the family Arenaviridae, Machupo Virus (MACV)is an enveloped, bisegmented negative-stranded RNA Virus. ArenaViruses utilize an ambisense coding strategy to direct viral gene transcription from two genomic segments, the large segment (L, ca. 7.2 kb) and the small segment (S, ca. 3.3 kb). Each segment carries two viral genes; S encodes the glycoprotein precursor (GPC) and the nucleoprotein (NP), while L encodes the RNA-dependent RNA polymerase (L polymerase) and the small RING finger protein Z. The GPC is posttranslationally cleaved by th

  • the ectodomain of glycoprotein from the candid 1 vaccine strain of junin Virus rendered Machupo Virus partially attenuated in mice lacking ifn αβ γ receptor
    PLOS Neglected Tropical Diseases, 2016
    Co-Authors: Takaaki Koma, Cheng Huang, Milagros Miller, Michael Patterson
    Abstract:

    Machupo Virus (MACV), a New World arenaVirus, is the etiological agent of Bolivian hemorrhagic fever (BHF). Junin Virus (JUNV), a close relative, causes Argentine hemorrhagic fever (AHF). Previously, we reported that a recombinant, chimeric MACV (rMACV/Cd#1-GPC) expressing glycoprotein from the Candid#1 (Cd#1) vaccine strain of JUNV is completely attenuated in a murine model and protects animals from lethal challenge with MACV. A rMACV with a single F438I substitution in the transmembrane domain (TMD) of GPC, which is equivalent to the F427I attenuating mutation in Cd#1 GPC, was attenuated in a murine model but genetically unstable. In addition, the TMD mutation alone was not sufficient to fully attenuate JUNV, indicating that other domains of the GPC may also contribute to the attenuation. To investigate the requirement of different domains of Cd#1 GPC for successful attenuation of MACV, we rescued several rMACVs expressing the ectodomain of GPC from Cd#1 either alone (MCg1), along with the TMD F438I substitution (MCg2), or with the TMD of Cd#1 (MCg3). All rMACVs exhibited similar growth curves in cultured cells. In mice, the MCg1 displayed significant reduction in lethality as compared with rMACV. The MCg1 was detected in brains and spleens of MCg1-infected mice and the infection was associated with tissue inflammation. On the other hand, all animals survived MCg2 and MCg3 infection without detectable levels of Virus in various organs while producing neutralizing antibody against Cd#1. Overall our data suggest the indispensable role of each GPC domain in the full attenuation and immunogenicity of rMACV/Cd#1 GPC.

  • Machupo Virus expressing gpc of the candid 1 vaccine strain of junin Virus is highly attenuated and immunogenic
    Journal of Virology, 2016
    Co-Authors: Takaaki Koma, Alexey Seregin, Cheng Huang, Milagros Miller, Michael Patterson, Payal D Maharaj, Steven Hallam
    Abstract:

    ABSTRACT Machupo Virus (MACV) is the causative agent of Bolivian hemorrhagic fever. Our previous study demonstrated that a MACV strain with a single amino acid substitution (F438I) in the transmembrane domain of glycoprotein is attenuated but genetically unstable in mice. MACV is closely related to Junin Virus (JUNV), the causative agent of Argentine hemorrhagic fever. Others and our group have identified the glycoprotein to be the major viral factor determining JUNV attenuation. In this study, we tested the compatibility of the glycoprotein of the Candid#1 live-attenuated vaccine strain of JUNV in MACV replication and its ability to attenuate MACV in vivo . Recombinant MACV with the Candid#1 glycoprotein (rMACV/Cd#1-GPC) exhibited growth properties similar to those of Candid#1 and was genetically stable in vitro . In a mouse model of lethal infection, rMACV/Cd#1-GPC was fully attenuated, more immunogenic than Candid#1, and fully protective against MACV infection. Therefore, the MACV strain expressing the glycoprotein of Candid#1 is safe, genetically stable, and highly protective against MACV infection in a mouse model. IMPORTANCE Currently, there are no FDA-approved vaccines and/or treatments for Bolivian hemorrhagic fever, which is a fatal human disease caused by MACV. The development of antiviral strategies to combat viral hemorrhagic fevers, including Bolivian hemorrhagic fever, is one of the top priorities of the Implementation Plan of the U.S. Department of Health and Human Services Public Health Emergency Medical Countermeasures Enterprise. Here, we demonstrate for the first time that MACV expressing glycoprotein of Candid#1 is a safe, genetically stable, highly immunogenic, and protective vaccine candidate against Bolivian hemorrhagic fever.

  • The Ectodomain of Glycoprotein from the Candid#1 Vaccine Strain of Junin Virus Rendered Machupo Virus Partially Attenuated in Mice Lacking IFN-αβ/γ Receptor.
    Public Library of Science (PLoS), 2016
    Co-Authors: Takaaki Koma, Cheng Huang, Milagros Miller, Michael Patterson
    Abstract:

    Machupo Virus (MACV), a New World arenaVirus, is the etiological agent of Bolivian hemorrhagic fever (BHF). Junin Virus (JUNV), a close relative, causes Argentine hemorrhagic fever (AHF). Previously, we reported that a recombinant, chimeric MACV (rMACV/Cd#1-GPC) expressing glycoprotein from the Candid#1 (Cd#1) vaccine strain of JUNV is completely attenuated in a murine model and protects animals from lethal challenge with MACV. A rMACV with a single F438I substitution in the transmembrane domain (TMD) of GPC, which is equivalent to the F427I attenuating mutation in Cd#1 GPC, was attenuated in a murine model but genetically unstable. In addition, the TMD mutation alone was not sufficient to fully attenuate JUNV, indicating that other domains of the GPC may also contribute to the attenuation. To investigate the requirement of different domains of Cd#1 GPC for successful attenuation of MACV, we rescued several rMACVs expressing the ectodomain of GPC from Cd#1 either alone (MCg1), along with the TMD F438I substitution (MCg2), or with the TMD of Cd#1 (MCg3). All rMACVs exhibited similar growth curves in cultured cells. In mice, the MCg1 displayed significant reduction in lethality as compared with rMACV. The MCg1 was detected in brains and spleens of MCg1-infected mice and the infection was associated with tissue inflammation. On the other hand, all animals survived MCg2 and MCg3 infection without detectable levels of Virus in various organs while producing neutralizing antibody against Cd#1. Overall our data suggest the indispensable role of each GPC domain in the full attenuation and immunogenicity of rMACV/Cd#1 GPC

Sina Bavari - One of the best experts on this subject based on the ideXlab platform.

  • Machupo Virus glycoprotein determinants for human transferrin receptor 1 binding and cell entry. PLoS One 2011
    2013
    Co-Authors: Sheli R Radoshitzky, Lindsay E. Longobardi, Jens H. Kuhn, Cary Retterer, Lian Dong, John H. Carra, Jeremiah C, Krishna Kota, Sina Bavari
    Abstract:

    Machupo Virus (MACV) is a highly pathogenic New World arenaVirus that causes hemorrhagic fever in humans. MACV, as well as other pathogenic New World arenaViruses, enter cells after their GP1 attachment glycoprotein binds to their cellular receptor, transferrin receptor 1 (TfR1). TfR1 residues essential for this interaction have been described, and a co-crystal of MACV GP1 bound to TfR1 suggests GP1 residues important for this association. We created MACV GP1 variants and tested their effect on TfR1 binding and Virus entry to evaluate the functional significance of some of these and additional residues in human and simian cells. We found residues R111, D123, Y122, and F226 to be essential, D155, and P160 important, and D114, S116, D140, and K169 expendable for the GP1-TfR1 interaction and MACV entry. Several MACV GP1 residues that are critical for the interaction with TfR1 are conserved among other New World arenaViruses, indicating a common basis of receptor interaction. Our findings also open avenues for the rational development of viral entry inhibitors

  • drug discovery technologies and strategies for Machupo Virus and other new world arenaViruses
    Expert Opinion on Drug Discovery, 2012
    Co-Authors: Sheli R Radoshitzky, Jens H. Kuhn, Fabian De Kokmercado, Peter B Jahrling, Sina Bavari
    Abstract:

    Introduction: Seven arenaViruses cause viral hemorrhagic fever in humans: the Old World arenaViruses Lassa and Lujo, and the New World Clade B arenaViruses Machupo (MACV), Junin (JUNV), Guanarito (GTOV), Sabia (SABV), and Chapare (CHPV). All of these Viruses are Risk Group 4 biosafety pathogens. MACV causes human disease outbreak with high case-fatality rates. To date, at least 1,200 cases with ≈200 fatalities have been recorded. Areas covered: This review summarizes available systems and technologies for the identification of antivirals against MACV. Furthermore, the article summarizes animal models that have been used for the in vivo evaluation of novel inhibitors. The article highlights present treatments for arenaviral diseases and provides an overview of efficacious small molecules and other therapeutics reported to date. Finally, the article summarizes strategies to identify novel inhibitors for anti-arenaviral therapy. Expert opinion: New high-throughput approaches to quantitate infection rates of ...

  • Machupo Virus glycoprotein determinants for human transferrin receptor 1 binding and cell entry.
    PloS one, 2011
    Co-Authors: Sheli R Radoshitzky, Lindsay E. Longobardi, Jens H. Kuhn, Cary Retterer, Lian Dong, Jeremiah C. Clester, Krishna P. Kota, John H. Carra, Sina Bavari
    Abstract:

    Machupo Virus (MACV) is a highly pathogenic New World arenaVirus that causes hemorrhagic fever in humans. MACV, as well as other pathogenic New World arenaViruses, enter cells after their GP1 attachment glycoprotein binds to their cellular receptor, transferrin receptor 1 (TfR1). TfR1 residues essential for this interaction have been described, and a co-crystal of MACV GP1 bound to TfR1 suggests GP1 residues important for this association. We created MACV GP1 variants and tested their effect on TfR1 binding and Virus entry to evaluate the functional significance of some of these and additional residues in human and simian cells. We found residues R111, D123, Y122, and F226 to be essential, D155, and P160 important, and D114, S116, D140, and K169 expendable for the GP1-TfR1 interaction and MACV entry. Several MACV GP1 residues that are critical for the interaction with TfR1 are conserved among other New World arenaViruses, indicating a common basis of receptor interaction. Our findings also open avenues for the rational development of viral entry inhibitors.

  • A STAT-1 knockout mouse model for Machupo Virus pathogenesis
    Virology Journal, 2011
    Co-Authors: Steven B Bradfute, Kelly S Stuthman, Amy C Shurtleff, Sina Bavari
    Abstract:

    Background Machupo Virus (MACV), a member of the Arenaviridae , causes Bolivian hemorrhagic fever, with ~20% lethality in humans. The pathogenesis of MACV infection is poorly understood, and there are no clinically proven treatments for disease. This is due, in part, to a paucity of small animal models for MACV infection in which to discover and explore candidate therapeutics. Methods Mice lacking signal transducer and activator of transcription 1 (STAT-1) were infected with MACV. Lethality, viral replication, metabolic changes, hematology, histopathology, and systemic cytokine expression were analyzed throughout the course of infection. Results We report here that STAT-1 knockout mice succumbed to MACV infection within 7-8 days, and presented some relevant clinical and histopathological manifestations of disease. Furthermore, the model was used to validate the efficacy of ribavirin in protection against infection. Conclusions The STAT-1 knockout mouse model can be a useful small animal model for drug testing and preliminary immunological analysis of lethal MACV infection.

  • assembly of a functional Machupo Virus polymerase complex
    Proceedings of the National Academy of Sciences of the United States of America, 2010
    Co-Authors: Philip J Kranzusch, Andreas D Schenk, Amal A Rahmeh, Sheli R Radoshitzky, Sina Bavari, Thomas Walz, Sean P J Whelan
    Abstract:

    Segmented negative-sense Viruses of the family Arenaviridae encode a large polymerase (L) protein that contains all of the enzymatic activities required for RNA synthesis. These activities include an RNA-dependent RNA polymerase (RdRP) and an RNA endonuclease that cleaves capped primers from cellular mRNAs to prime transcription. Using purified catalytically active Machupo Virus L, we provide a view of the overall architecture of this multifunctional polymerase and reconstitute complex formation with an RNA template in vitro. The L protein contains a central ring domain that is similar in appearance to the RdRP of dsRNA Viruses and multiple accessory appendages that may be responsible for 5′ cap formation. RNA template recognition by L requires a sequence-specific motif located at positions 2–5 in the 3′ terminus of the viral genome. Moreover, L-RNA complex formation depends on single-stranded RNA, indicating that inter-termini dsRNA interactions must be partially broken for complex assembly to occur. Our results provide a model for arenaVirus polymerase–template interactions and reveal the structural organization of a negative-strand RNA Virus L protein.