264 results on '"Junin virus"'
Search Results
2. Patent Issued for Crystalline forms of (s)-2-ethylbutyl 2-(((s)-(((2r,3s,4r,5r)-5-(4-aminopyrrolo[2,1-f] [1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxytetrahydrofuran-2-yl)methoxy) (phenoxy) phosphoryl)amino)propanoate (USPTO 12030906).
- Subjects
RESPIRATORY syncytial virus ,EBOLA virus ,VIRUS diseases ,RNA viruses ,FILOVIRIDAE - Abstract
A patent has been issued to Gilead Sciences Inc. for crystalline forms of a compound known as (S)-2-ethylbutyl 2-(((S)-(((2R,3S,4R,5R)-5-(4-aminopyrrolo[2,1-f][1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)amino)propanoate. This compound has been found to exhibit antiviral properties against various viruses, including Arenaviridae, Coronaviridae, Filoviridae, Flaviviridae, and Paramyxoviridae. The patent describes different crystalline forms of the compound and their potential use in pharmaceutical compositions for the treatment of viral infections. This information may be useful for researchers studying antiviral therapies and seeking broad-spectrum antiviral activity. [Extracted from the article]
- Published
- 2024
3. "Recombinant Polypeptides For Programming Extracellular Vesicles" in Patent Application Approval Process (USPTO 20230203532).
- Abstract
A recombinant polypeptide for directing an extracellular vesicle (EV) to at least one target cell comprising: at least one targeting moiety for directing said EV to said at least one target molecule expressed by said at least one target cell, at least one EV-anchoring polypeptide, and at least one intravesicular polypeptide. "In another aspect, there is provided a recombinant polypeptide for directing an extracellular vesicle (EV) to at least one target cell comprising: at least one targeting moiety for directing said EV to said at least one target molecule expressed by said at least one target cell, at least one EV-anchoring polypeptide, and at least one intravesicular polypeptide. A recombinant tumor-selective viral particle comprising a nucleic acid encoding a recombinant polypeptide for directing an extracellular vesicle (EV) to at least one target cell, said recombinant polypeptide comprising: at least one targeting moiety for directing said EV to said at least one target molecule expressed by said at least one target cell, at least one EV-anchoring polypeptide, and at least one intravesicular polypeptide. [Extracted from the article]
- Published
- 2023
4. Single dose rVSVΔG-JUNVGP vaccine protects guinea pigs against lethal Junin virus challenge
- Author
-
Chad E. Mire, Robert W. Cross, Karla A. Fenton, Dylan M Johnson, Thomas W. Geisbert, Natalie S. Dobias, and Teresa E. Sorvillo
- Subjects
Immunology ,Biology ,medicine.disease_cause ,Article ,Virus ,Guinea pig ,medicine ,Pharmacology (medical) ,Pathogen ,RC254-282 ,Pharmacology ,Ebola virus ,Antibody titer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC581-607 ,biology.organism_classification ,Virology ,Arenaviruses ,Experimental models of disease ,Infectious Diseases ,Viral infection ,Vesicular stomatitis virus ,Junin virus ,biology.protein ,Immunologic diseases. Allergy ,Antibody - Abstract
Junin virus (JUNV) is a pathogen of biodefense importance due to its potential for aerosol transmission and mortality rates reaching 30%. Currently, there are no JUNV vaccines licensed by the United States Food and Drug Administration (FDA) for at-risk individuals. A vaccine based on recombinant vesicular stomatitis virus (rVSV) has been effectively used to prevent Ebola virus disease in humans. Here, we evaluated the protective efficacy of a rVSV expressing the JUNV glycoprotein (rVSVΔG-JUNVGP) in a guinea pig model of lethal JUNV disease. Two groups of guinea pigs, one prime and one prime-boost, were vaccinated with rVSVΔG-JUNVGP; six control animals remained unvaccinated. Survival for prime and prime-boost vaccinated animals was 100% while the challenge virus was uniformly lethal in all control animals. Animals in both vaccine groups developed robust, high avidity IgG antibody titers post-vaccination as well as detectable neutralizing antibodies while control animals failed to develop detectable antibody responses. This study demonstrates for the first time that rVSV expressing the JUNV GP fully protects guinea pigs from lethal JUNV challenge with a single injection vaccine.
- Published
- 2021
- Full Text
- View/download PDF
5. Simultaneous Detection of Ebola Virus and Pathogens Associated With Hemorrhagic Fever by an Oligonucleotide Microarray
- Author
-
Yanjun Zhang, Wenwu Yao, Xiuyu Lou, Hao Yan, Zhang-Nv Yang, and Haiyan Mao
- Subjects
Microbiology (medical) ,viruses ,detection ,Dengue virus ,Biology ,medicine.disease_cause ,Microbiology ,Virus ,Ebola virus ,03 medical and health sciences ,medicine ,Chikungunya ,Original Research ,030304 developmental biology ,Hantavirus ,0303 health sciences ,030306 microbiology ,Yellow fever ,virus diseases ,medicine.disease ,biology.organism_classification ,Virology ,QR1-502 ,Junin virus ,microarray ,hemorrhagic fever ,pathogen ,Severe fever with thrombocytopenia syndrome virus - Abstract
Ebola virus infection causes severe hemorrhagic fever, and its mortality rates varied from 25 to 90% in the previous outbreaks. The highly infectious and lethal nature of this virus highlights the need for reliable and sensitive diagnostic methods to distinguish it from other diseases present with similar clinical symptoms. Based on multiplex polymerase chain reaction (PCR) and oligonucleotide microarray technology, a cost-effective, multipathogen and high-throughput method was developed for simultaneous detection of Ebola virus and other pathogens associated with hemorrhagic fever, including Marburg virus, Lassa fever virus, Junin virus, Machupo virus, Rift Valley fever virus, Crimean-Congo hemorrhagic fever virus, malaria parasite, hantavirus, severe fever with thrombocytopenia syndrome virus, dengue virus, yellow fever virus, Chikungunya virus, influenza A virus, and influenza B virus. This assay had an excellent specificity for target pathogens, without overlap signal between the probes. The limit of detection was approximately 103 pathogen copies/μl. A total of 60 positive nucleic acid samples for different pathogens were detected, a concordance of 100% was observed between microarray assay and real-time PCR analysis. Consequently, the described oligonucleotide microarray may be specific and sensitive assay for diagnosis and surveillance of infections caused by Ebola virus and other species of hemorrhagic fever pathogens.
- Published
- 2021
- Full Text
- View/download PDF
6. Melittin: a venom-derived peptide with promising anti-viral properties
- Author
-
Hamed Memariani, Mohammad Shahidi-Dadras, Mojtaba Memariani, and Hamideh Moravvej
- Subjects
0301 basic medicine ,Microbiology (medical) ,viruses ,030106 microbiology ,Context (language use) ,Review ,Coxsackievirus ,medicine.disease_cause ,Antiviral Agents ,complex mixtures ,Melittin ,Virus ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Animals ,030212 general & internal medicine ,Bee ,biology ,Anti-viral activity ,General Medicine ,biology.organism_classification ,Venom ,Melitten ,Virology ,Infectious Diseases ,Herpes simplex virus ,chemistry ,Vesicular stomatitis virus ,Junin virus ,Viruses ,Enterovirus ,Drug ,Peptides - Abstract
Despite tremendous advances in the development of anti-viral therapeutics, viral infections remain a chief culprit accounting for ongoing morbidity and mortality worldwide. Natural products, in particular animal venoms, embody a veritable cornucopia of exotic constituents, suggesting an immensurable source of anti-infective drugs. In this context, melittin, the principal constituent in the venom of the European honeybee Apis mellifera, has been demonstrated to exert anti-cancer, anti-inflammatory, anti-diabetic, anti-infective, and adjuvant properties. To our knowledge, there is no review appertaining to effects of melittin against viruses, prompting us to synopsize experimental investigations on its anti-viral activity throughout the past decades. Accumulating evidence indicates that melittin curbs infectivity of a diverse array of viruses including coxsackievirus, enterovirus, influenza A viruses, human immunodeficiency virus (HIV), herpes simplex virus (HSV), Junín virus (JV), respiratory syncytial virus (RSV), vesicular stomatitis virus (VSV), and tobacco mosaic virus (TMV). However, medication safety, different routes of administrations, and molecular mechanisms behind the anti-viral activity of melittin should be scrutinized in future studies.
- Published
- 2019
- Full Text
- View/download PDF
7. Second-Generation Live-Attenuated Candid#1 Vaccine Virus Resists Reversion and Protects against Lethal Junín Virus Infection in Guinea Pigs
- Author
-
Joanne York, Brian B. Gowen, Luci Wandersee, Brady T. Hickerson, Jonna B. Westover, Kevin W. Bailey, Jack H. Nunberg, and Eric J. Sefing
- Subjects
Male ,Immunology ,Guinea Pigs ,Reversion ,Virulence ,Biology ,medicine.disease_cause ,Antibodies, Viral ,Vaccines, Attenuated ,Microbiology ,Virus ,Hemorrhagic Fever, American ,03 medical and health sciences ,0302 clinical medicine ,Immunogenicity, Vaccine ,Virology ,Chlorocebus aethiops ,medicine ,Animals ,030212 general & internal medicine ,Vero Cells ,030304 developmental biology ,0303 health sciences ,Mutation ,Vaccines, Synthetic ,Attenuated vaccine ,Arenavirus ,Junin virus ,Viral Vaccines ,biology.organism_classification ,Insect Science ,Pathogenesis and Immunity ,Viral disease - Abstract
Live-attenuated virus vaccines are highly effective in preventing viral disease but carry intrinsic risks of residual virulence and reversion to pathogenicity. The classically derived Candid#1 virus protects seasonal field workers in Argentina against zoonotic infection by Junin virus (JUNV) but is not approved in the United States, in part due to the potential for reversion at the attenuating locus, a phenylalanine-to-isoleucine substitution at position 427 in the GP2 subunit of the GPC envelope glycoprotein. Previously, we demonstrated facile reversion of recombinant Candid#1 (rCan) in cell culture and identified an epistatic interaction between the attenuating I427 and a secondary K33S mutation in the stable signal peptide (SSP) subunit of GPC that imposes an evolutionary barrier to reversion. The magnitude of this genetic barrier is manifest in our repeated failures to rescue the hypothetical revertant virus. In this study, we show that K33S rCan is safe and attenuated in guinea pigs and capable of eliciting potent virus-neutralizing antibodies. Immunized animals are fully protected against lethal challenge with virulent JUNV. In addition, we employed a more permissive model of infection in neonatal mice to investigate genetic reversion. RNA sequence analysis of the recovered virus identified revertant viruses in pups inoculated with the parental rCan virus and none in mice receiving K33S rCan (P
- Published
- 2021
8. Combination of highly antigenic nucleoproteins to inaugurate a cross-reactive next generation vaccine candidate against Arenaviridae family
- Author
-
Nazmul Hossain, Kazi Faizul Azim, Omar Faruk Bhuiyan, Rahima Akter, Mahmudul Hasan, Tahera Lasker, and Mantasha Mahmud Hia
- Subjects
0301 basic medicine ,Science (General) ,viruses ,Recombinant vaccine ,Lymphocytic choriomeningitis ,medicine.disease_cause ,Epitope ,Virus ,03 medical and health sciences ,Q1-390 ,0302 clinical medicine ,medicine ,Lujo virus ,Nucleoprotein ,H1-99 ,Multidisciplinary ,Arenavirus ,biology ,Reverse vaccinology ,biology.organism_classification ,medicine.disease ,Hemorrhagic fevers ,Virology ,Social sciences (General) ,030104 developmental biology ,Lassa virus ,Junin virus ,Molecular docking ,030217 neurology & neurosurgery ,Research Article - Abstract
Arenaviral infections often result lethal hemorrhagic fevers, affecting primarily in African and South American regions. To date, there is no FDA-approved licensed vaccine against arenaviruses and treatments have been limited to supportive therapy. Hence, the study was employed to design a highly immunogenic cross-reactive vaccine against Arenaviridae family using reverse vaccinology approach. The whole proteome of Lassa virus (LASV), Lymphocytic Choriomeningitis virus (LCMV), Lujo virus and Guanarito virus were retrieved and assessed to determine the most antigenic viral proteins. Both T-cell and B-cell epitopes were predicted and screened based on transmembrane topology, antigenicity, allergenicity, toxicity and molecular docking analysis. The final constructs were designed using different adjuvants, top epitopes, PADRE sequence and respective linkers and were assessed for the efficacy, safety, stability and molecular cloning purposes. The proposed epitopes were highly conserved (84%–100%) and showed greater cumulative population coverage. Moreover, T cell epitope GWPYIGSRS was conserved in Junin virus (Argentine mammarenavirus) and Sabia virus (Brazilian mammarenavirus), while B cell epitope NLLYKICLSG was conserved in Machupo virus (Bolivian mammarenavirus) and Sabia virus, indicating the possibility of final vaccine construct to confer a broad range immunity in the host. Docking analysis of the refined vaccine with different MHC molecules and human immune receptors were biologically significant. The vaccine-receptor (V1-TLR3) complex showed minimal deformability at molecular level and was compatible for cloning into pET28a(+) vector of E. coli strain K12. The study could be helpful in developing vaccine to combat arenaviral infections in the future. However, further in vitro and in vivo trials using model animals are highly recommended for the experimental validation of our findings., Arenavirus; Hemorrhagic fevers; Nucleoprotein; Recombinant vaccine; Molecular docking
- Published
- 2021
9. Glycoprotein N-linked glycans play a critical role in arenavirus pathogenicity
- Author
-
Michael Patterson, Jonathan Abraham, Aida G. Walker, Adrian Coscia, Cheng Huang, Milagros Miller, Slobodan Paessler, Jeanon N. Smith, John T. Manning, Takaaki Koma, Junki Maruyama, and Steven Hallam
- Subjects
RNA viruses ,Viral Diseases ,Glycosylation ,Physiology ,viruses ,Glycobiology ,Pathogenesis ,Pathology and Laboratory Medicine ,Antibodies, Viral ,Biochemistry ,Epitope ,chemistry.chemical_compound ,Medical Conditions ,Immune Physiology ,Medicine and Health Sciences ,Post-Translational Modification ,Biology (General) ,Arenaviruses, New World ,chemistry.chemical_classification ,Vaccines ,Immune System Proteins ,biology ,Arenavirus ,Infectious Diseases ,Ectodomain ,Medical Microbiology ,Viral Pathogens ,Viruses ,Biological Cultures ,Pathogens ,Research Article ,Neglected Tropical Diseases ,Glycan ,Infectious Disease Control ,QH301-705.5 ,Immunology ,Research and Analysis Methods ,Microbiology ,Antibodies ,Hemorrhagic Fever, American ,Virus ,Virology ,Argentine Hemorrhagic Fever ,Genetics ,Animals ,Humans ,Microbial Pathogens ,Molecular Biology ,Viral Hemorrhagic Fevers ,Junin virus ,Organisms ,Biology and Life Sciences ,Proteins ,Viral Vaccines ,Cell Cultures ,RC581-607 ,Tropical Diseases ,biology.organism_classification ,Antibodies, Neutralizing ,Arenaviruses ,carbohydrates (lipids) ,chemistry ,biology.protein ,Parasitology ,Immunologic diseases. Allergy ,Glycoprotein - 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., Author summary Several arenaviruses cause severe hemorrhagic fevers in humans. The only vaccine against arenavirus infections is Candid#1, a live attenuated vaccine against Argentine hemorrhagic fever. So far, we have successfully attenuated additional one of the arenaviruses, Machupo virus, the causative agent of Bolivian hemorrhagic fever. Unraveling this attenuation mechanism might help the development of live-attenuated vaccines for other arenaviruses. In this study, we revealed that the specific glycans of the viral glycoproteins play an important role in pathogenicity in vivo. The glycans facilitate the virus to evade neutralizing antibodies. This study would contribute to the development of arenavirus vaccine candidates.
- Published
- 2021
10. Inhibition of arenaviruses by combinations of orally available approved drugs
- Author
-
Shawn Herring, Lisa Evans DeWald, Aidan O’Connor, Susan L. Fink, Mary F. Paine, Delaney Kirchmeier, Jessica M. Oda, Yuying Liang, Pamela J. Glass, Lisa M. Johansen, Tero Aittokallio, Stephen J. Polyak, Elizabeth A. Nelson, Aleksandr Ianevski, Jessica Wagoner, Gene G. Olinger, Qinfeng Huang, Judith M. White, Institute for Molecular Medicine Finland, Computational Systems Medicine, Helsinki Institute for Information Technology, Tero Aittokallio / Principal Investigator, and Bioinformatics
- Subjects
MECHANISM ,PHARMACOKINETICS ,viruses ,repurposing ,synergy ,Administration, Oral ,medicine.disease_cause ,SynergyFinder2 ,Mice ,FUSION ,Chlorocebus aethiops ,Pharmacology (medical) ,11832 Microbiology and virology ,0303 health sciences ,biology ,virus diseases ,Lassa ,ANTIVIRAL ACTIVITY ,Drug Synergism ,Arenavirus ,3. Good health ,EBOLA-VIRUS ,Infectious Diseases ,SINGLE ,INFLUENZA ,317 Pharmacy ,Ebola ,Drug Therapy, Combination ,Erratum ,filovirus ,medicine.drug_class ,broad-spectrum antiviral ,Lymphocytic choriomeningitis ,arbidol ,Antiviral Agents ,Proof of Concept Study ,Virus ,Cell Line ,Marburg ,03 medical and health sciences ,Viral entry ,medicine ,Animals ,Arenaviridae Infections ,Humans ,Vero Cells ,030304 developmental biology ,Pharmacology ,Ebola virus ,IDENTIFICATION ,SARS-CoV-2 ,030306 microbiology ,business.industry ,Junin ,biology.organism_classification ,medicine.disease ,Virology ,Lassa virus ,HEK293 Cells ,ARIPIPRAZOLE ,Pichinde ,Junin virus ,Antiviral drug ,business - Abstract
Correction: Volume65, Issue6 Article Numbere00653- 21 DOI10.1128/AAC.00653-21 Neglected diseases caused by arenaviruses such as Lassa virus (LASV) and filoviruses like Ebola virus (EBOV) primarily afflict resource-limited countries, where antiviral drug development is often minimal. Previous studies have shown that many approved drugs developed for other clinical indications inhibit EBOV and LASV and that combinations of these drugs provide synergistic suppression of EBOV, often by blocking discrete steps in virus entry. We hypothesize that repurposing of combinations of orally administered approved drugs provides effective suppression of arenaviruses. In this report, we demonstrate that arbidol, an approved influenza antiviral previously shown to inhibit EBOV, LASV, and many other viruses, inhibits murine leukemia virus (MLV) reporter viruses pseudotyped with the fusion glycoproteins (GPs) of other arenaviruses (Junin virus (JUNV], lymphocytic choriomeningitis virus (LCMV), and Pichinde virus (PICA). Arbidol and other approved drugs, including aripiprazole, amodiaquine, sertraline, and niclosamide, also inhibit infection of cells by infectious PICV, and arbidol, sertraline, and niclosamide inhibit infectious LASV. Combining arbidol with aripiprazole or sertraline results in the synergistic suppression of LASV and JUNV GP-bearing pseudoviruses. This proof-of-concept study shows that arenavirus infection in vitro can be synergistically inhibited by combinations of approved drugs. This approach may lead to a proactive strategy with which to prepare for and control known and new arenavirus outbreaks.
- Published
- 2021
11. Study Data from University of Buenos Aires Update Knowledge of Junin Virus (Modulation of the Aryl Hydrocarbon Receptor Signaling Pathway Impacts on Junin Virus Replication).
- Abstract
Keywords: Arenaviridae; Aryl Hydrocarbon Receptor; DNA-Binding Proteins; Hydrocarbons; Junin Virus; Microbiological Processes; New World Arenaviruses; Organic Chemicals; Orphan Nuclear Receptors; Proteins; RNA Viruses; Risk and Prevention; Transcription Factors; Vertebrate Viruses; Viral; Virology; Virus; Virus Physiological Phenomena; Virus Physiological Processes; Virus Replication EN Arenaviridae Aryl Hydrocarbon Receptor DNA-Binding Proteins Hydrocarbons Junin Virus Microbiological Processes New World Arenaviruses Organic Chemicals Orphan Nuclear Receptors Proteins RNA Viruses Risk and Prevention Transcription Factors Vertebrate Viruses Viral Virology Virus Virus Physiological Phenomena Virus Physiological Processes Virus Replication 2762 2762 1 03/23/23 20230317 NES 230317 2023 MAR 17 (NewsRx) -- By a News Reporter-Staff News Editor at Drug Week -- Research findings on Junin virus are discussed in a new report. Aryl Hydrocarbon Receptor, DNA-Binding Proteins, Hydrocarbons, Junin Virus, Arenaviridae, Microbiological Processes, New World Arenaviruses, Organic Chemicals, Orphan Nuclear Receptors, Proteins, RNA Viruses, Risk and Prevention, Transcription Factors, Vertebrate Viruses, Viral, Virology, Virus, Virus Physiological Phenomena, Virus Physiological Processes, Virus Replication. [Extracted from the article]
- Published
- 2023
12. Development of a Reverse Genetic System to Generate Recombinant Chimeric Tacaribe Virus that Expresses Junín Virus Glycoproteins
- Author
-
Sabrina Foscaldi, Carlos Adolfo Palacios, Nora López, Claudia Soledad Sepúlveda, Maria Eugenia Loureiro, and María Belén Forlenza
- Subjects
Microbiology (medical) ,TACARIBE VIRUS ,Junín virus ,CHIMERIC VIRUS ,chimeric virus ,lcsh:Medicine ,Biology ,Virus ,law.invention ,INFECTIOUS CLONE ,purl.org/becyt/ford/1 [https] ,law ,Immunology and Allergy ,viral attenuation ,purl.org/becyt/ford/1.6 [https] ,Molecular Biology ,chemistry.chemical_classification ,General Immunology and Microbiology ,VIRAL ATTENUATION ,Communication ,lcsh:R ,JUNÍN VIRUS ,non-coding region ,infectious clone ,RNA ,NON-CODING REGION ,biology.organism_classification ,Virology ,Hemorrhagic Fevers ,Infectious Diseases ,chemistry ,Junin virus ,Recombinant DNA ,Tacaribe virus ,Glycoprotein ,Intracellular ,Mammarenavirus - Abstract
Mammarenaviruses are enveloped and segmented negative-stranded RNA viruses that comprise several pathogenic members associated with severe human hemorrhagic fevers. Tacaribe virus (TCRV) is the prototype for the New World group of mammarenaviruses and is not only naturally attenuated but also phylogenetically and antigenically related to all South American pathogenic mammarenaviruses, particularly the Junín virus (JUNV), which is the etiological agent of Argentinian hemorrhagic fever (AHF). Moreover, since TCRV protects guinea pigs and non-human primates from lethal challenges with pathogenic strains of JUNV, it has already been considered as a potential live-attenuated virus vaccine candidate against AHF. Here, we report the development of a reverse genetic system that relies on T7 polymerase-driven intracellular expression of the complementary copy (antigenome) of both viral S and L RNA segments. Using this approach, we successfully recovered recombinant TCRV (rTCRV) that displayed growth properties resembling those of authentic TCRV. We also generated a chimeric recombinant TCRV expressing the JUNV glycoproteins, which propagated similarly to wild-type rTCRV. Moreover, a controlled modification within the S RNA 5′ non-coding terminal sequence diminished rTCRV propagation in a cell-type dependent manner, giving rise to new perspectives where the incorporation of additional attenuation markers could contribute to develop safe rTCRV-based vaccines against pathogenic mammarenaviruses. Fil: Foscaldi, Sabrina Andrea. Ministerio de Produccion y Trabajo. Secretaria de Gobierno de Agroindustria. Servicio Nacional de Sanidad y Calidad Agroalimentaria. Centro de Virologia Animal. - Consejo Nacional de Investigaciones Cientificas y Tecnicas. Oficina de Coordinacion Administrativa Ciudad Universitaria. Centro de Virologia Animal.; Argentina Fil: Loureiro, Maria Eugenia. Ministerio de Produccion y Trabajo. Secretaria de Gobierno de Agroindustria. Servicio Nacional de Sanidad y Calidad Agroalimentaria. Centro de Virologia Animal. - Consejo Nacional de Investigaciones Cientificas y Tecnicas. Oficina de Coordinacion Administrativa Ciudad Universitaria. Centro de Virologia Animal.; Argentina Fil: Sepúlveda, Claudia Soledad. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Ciudad Universitaria. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales; Argentina Fil: Palacios, Carlos Adolfo. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Parque Centenario. Instituto de Ciencia y Tecnología "Dr. César Milstein". Fundación Pablo Cassará. Instituto de Ciencia y Tecnología "Dr. César Milstein"; Argentina Fil: Forlenza, María Belén. Ministerio de Produccion y Trabajo. Secretaria de Gobierno de Agroindustria. Servicio Nacional de Sanidad y Calidad Agroalimentaria. Centro de Virologia Animal. - Consejo Nacional de Investigaciones Cientificas y Tecnicas. Oficina de Coordinacion Administrativa Ciudad Universitaria. Centro de Virologia Animal.; Argentina Fil: Lopez, Nora Mabel. Ministerio de Produccion y Trabajo. Secretaria de Gobierno de Agroindustria. Servicio Nacional de Sanidad y Calidad Agroalimentaria. Centro de Virologia Animal. - Consejo Nacional de Investigaciones Cientificas y Tecnicas. Oficina de Coordinacion Administrativa Ciudad Universitaria. Centro de Virologia Animal.; Argentina
- Published
- 2020
13. A single mutation (V64G) within the RING Domain of Z attenuates Junin virus
- Author
-
David H. Walker, Junki Maruyama, Slobodan Paessler, Alexey Seregin, Steven Hallam, Cheng Huang, John T. Manning, and Juan Carlos de la Torre
- Subjects
0301 basic medicine ,New World Arenavirus ,Physiology ,viruses ,RC955-962 ,Pathology and Laboratory Medicine ,0302 clinical medicine ,Medical Conditions ,Immune Physiology ,Cricetinae ,Arctic medicine. Tropical medicine ,Cellular types ,Chlorocebus aethiops ,Medicine and Health Sciences ,Public and Occupational Health ,chemistry.chemical_classification ,Mammals ,Vaccines ,Viral Vaccine ,Immune cells ,Eukaryota ,Animal Models ,Vaccination and Immunization ,Body Fluids ,Blood ,Infectious Diseases ,Experimental Organism Systems ,Medical Microbiology ,Viral Pathogens ,Vertebrates ,Viruses ,White blood cells ,Female ,Pathogens ,Anatomy ,Public aspects of medicine ,RA1-1270 ,Research Article ,Cell biology ,Blood cells ,Infectious Disease Control ,030231 tropical medicine ,Guinea Pigs ,Immunology ,T cells ,Cytotoxic T cells ,Biology ,Argentine hemorrhagic fever ,Research and Analysis Methods ,Rodents ,Microbiology ,Virus ,Hemorrhagic Fever, American ,Cell Line ,03 medical and health sciences ,Viral Proteins ,Virology ,Vaccine Development ,medicine ,Animals ,Microbial Pathogens ,Vero Cells ,Glycoproteins ,Junin virus ,Euthanasia ,Public Health, Environmental and Occupational Health ,Wild type ,Organisms ,Biology and Life Sciences ,Viral Vaccines ,medicine.disease ,biology.organism_classification ,In vitro ,030104 developmental biology ,chemistry ,Animal cells ,Amniotes ,Mutation ,Animal Studies ,Preventive Medicine ,Glycoprotein ,Zoology ,Spleen - 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)., Author summary The continual development of safe, effective vaccines against emerging diseases is one of the greatest challenges facing the scientific community. The New World group of mammarenaviruses contains multiple human pathogens, each capable of causing severe hemorrhagic disease. Among these, only Junin virus has a distributed vaccine. By utilizing this vaccine, we are able to determine vaccine development strategies for related New World viruses that represent an emerging threat. Here we demonstrate that manipulation of the viral Z protein is able to produce an incompatibility that ultimately attenuates the virus. This provides yet another tool for future vaccine development to further global public health.
- Published
- 2020
14. Should virgin coconut oil be used in the adjunctive treatment of COVID-19?
- Author
-
Carol Stephanie C. Tan-Lim and Corinna Victoria Martinez
- Subjects
animal structures ,food.ingredient ,biology ,Traditional medicine ,business.industry ,Coconut oil ,food and beverages ,General Medicine ,medicine.disease_cause ,biology.organism_classification ,Lauric acid ,Virus ,Monolaurin ,chemistry.chemical_compound ,food ,chemistry ,Vesicular stomatitis virus ,Junin virus ,Adjunctive treatment ,Medicine ,business ,Coronavirus - Abstract
KEY FINDINGS There is currently no evidence to support the use of virgin coconut oil in the adjunctive treatment of COVID-19. • Virgin coconut oil is naturally extracted from fresh coconut kernel and is rich in medium chain triglycerides, with lauric acid as the predominant fatty acid.• Virgin coconut oil is currently explored as an adjunctive treatment for patients with COVID-19 due to its antiviral and immunomodulatory properties.• In vitro studies show that lauric acid or its derivative exert inhibitory activities against viruses with similar structure to coronavirus (enveloped ssRNA virus) such as Junin virus, vesicular stomatitis virus, human immunodeficiency virus type 1 (HIV-1), and Semliki Forest virus.• Animal studies demonstrate antiviral activity of monolaurin, the pharmacologically active metabolite of lauric acid, on avian influenza virus and Simean immunodeficiency virus, which are both enveloped ssRNA viruses.• Clinical trials among patients with HIV report that virgin coconut oil can increase CD4+ T lymphocyte counts and reduce viral load.• In vitro and animal studies demonstrate anti-inflammatory properties of virgin coconut oil.• At present, there are no studies that investigate the effectiveness of virgin coconut oil in the adjunctive treatment of COVID-19 infection.• There is currently one ongoing clinical trial in the Philippines evaluating the use of virgin coconut oil in the adjunctive treatment of COVID-19.• Nausea, vomiting, mild diarrhea, and abdominal pain have been reported, but no serious adverse events have been identified with the use of virgin coconut oil.• To date, there are no guidelines that mention virgin coconut oil as an option for the adjunctive treatment of COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
15. Development of Reverse Genetics for the Prototype New World Mammarenavirus Tacaribe Virus
- Author
-
Juan Carlos de la Torre, Luis Martinez-Sobrido, and Chengjin Ye
- Subjects
Immunology ,Argentine hemorrhagic fever ,Antibodies, Viral ,Vaccines, Attenuated ,Virus Replication ,Microbiology ,Genome ,Virus ,Hemorrhagic Fever, American ,Cell Line ,chemistry.chemical_compound ,Virology ,Chlorocebus aethiops ,Ribavirin ,Vaccines and Antiviral Agents ,medicine ,Animals ,Humans ,Vector (molecular biology) ,Arenaviridae ,Vero Cells ,Arenaviruses, New World ,Recombination, Genetic ,Reporter gene ,Junin virus ,biology ,Base Sequence ,DNA Viruses ,Viral Vaccines ,biology.organism_classification ,medicine.disease ,Reverse genetics ,Reverse Genetics ,chemistry ,Insect Science - Abstract
The New World mammarenavirus Tacaribe virus (TCRV) has been isolated from fruit bats, mosquitoes, and ticks, whereas all other known New World mammarenaviruses are maintained in rodents. TCRV has not been linked to human disease, but it has been shown to protect against Argentine hemorrhagic fever-like disease in marmosets infected with the New World mammarenavirus Junín virus (JUNV), indicating the potential of TCRV as a live-attenuated vaccine for the treatment of Argentine hemorrhagic fever. Implementation of TCRV as a live-attenuated vaccine or a vaccine vector would be facilitated by the establishment of reverse genetics systems for the genetic manipulation of the TCRV genome. In this study, we developed, for the first time, reverse genetics approaches for the generation of recombinant TCRV (rTCRV). We successfully rescued a wild-type (WT) rTCRV (a trisegmented form of TCRV expressing two reporter genes [r3TCRV]) and a bisegmented TCRV expressing a single reporter gene from a bicistronic viral mRNA (rTCRV/GFP). These reverse genetics approaches represent an excellent tool to investigate the biology of TCRV and to explore its potential use as a live-attenuated vaccine or a vaccine vector for the treatment of other viral infections. Notably, we identified a 39-nucleotide (nt) deletion (Δ39) in the noncoding intergenic region (IGR) of the viral large (L) segment that is required for optimal virus multiplication. Accordingly, an rTCRV containing this 39-nt deletion in the L-IGR (rTCRV/Δ39) exhibited decreased viral fitness in cultured cells, suggesting the feasibility of using this deletion in the L-IGR as an approach to attenuate TCRV, and potentially other mammarenaviruses, for their implementation as live-attenuated vaccines or vaccine vectors. IMPORTANCE To date, no Food and Drug Administration (FDA)-approved vaccines are available to combat hemorrhagic fever caused by mammarenavirus infections in humans. Treatment of mammarenavirus infections is limited to the off-label use of ribavirin, which is partially effective and associated with significant side effects. Tacaribe virus (TCRV), the prototype member of the New World mammarenaviruses, is nonpathogenic in humans but able to provide protection against Junín virus (JUNV), the causative agent of Argentine hemorrhagic fever, demonstrating the feasibility of using TCRV as a live-attenuated vaccine vector for the treatment of JUNV and potentially other viral infections. Here, we describe for the first time the feasibility of generating recombinant TCRV (rTCRV) using reverse genetics approaches, which paves the way to study the biology of TCRV and also its potential use as a live-attenuated vaccine or a vaccine vector for the treatment of mammarenavirus and/or other viral infections in humans.
- Published
- 2020
16. Analysis of the Cell Type-Dependence on the Arenavirus Z-Mediated Virus-Like Particle Production
- Author
-
Patrick I. Mpingabo, Shuzo Urata, and Jiro Yasuda
- Subjects
Microbiology (medical) ,viruses ,lcsh:QR1-502 ,medicine.disease_cause ,Microbiology ,lcsh:Microbiology ,Virus ,ESCRT ,virus-like particle ,03 medical and health sciences ,cell type-dependence ,Virus-like particle ,medicine ,L-domain ,Viral shedding ,arenavirus ,030304 developmental biology ,Original Research ,0303 health sciences ,Arenavirus ,biology ,030306 microbiology ,HEK 293 cells ,virus diseases ,biology.organism_classification ,Virology ,Lassa virus ,Junin virus - Abstract
Several arenaviruses are highly pathogenic to humans, causing hemorrhagic fever. Discovery of anti-arenavirus drug candidates is urgently needed, although the molecular basis of the host- and organ-specific pathogenicity remains to be fully elucidated. The arenavirus Z protein facilitates production of virus-like particles (VLPs), providing an established method to assess virus budding. In this study, we examined the efficiency of VLP production by solely expressing Z protein of several different arenaviruses. In addition, we analyzed the role of the late (L)-domain of the arenavirus Z protein, which is essential for the interaction with ESCRT proteins, in VLP production among different cell lines. VLP assay was performed using Z proteins of Junin virus (JUNV), Machupo virus (MACV), Tacaribe virus (TCRV), Latino virus (LATV), Pichinde virus (PICV), and Lassa virus (LASV) in six different cell lines: HEK293T, Huh-7, A549, Vero76, BHK-21, and NIH3T3 cells. JUNV, MACV, and LASV Z proteins efficiently produced VLPs in all tested cell lines, while the efficiencies of VLP production by the other arenavirus Z proteins were cell type-dependent. The contribution of the L-domain(s) within Z protein to VLP production also highly depended on the cell type. These results suggested that each arenavirus has its own particle-production mechanism, which is different among the cell types., Frontiers in Microbiology, 11, art.no.562814; 2020
- Published
- 2020
17. Antiviral activity of A771726, the active metabolite of leflunomide, against Junín virus
- Author
-
Claudia Soledad Sepúlveda, Cybele C. García, and Elsa Beatriz Damonte
- Subjects
0301 basic medicine ,Toluidines ,viruses ,Hydroxybutyrates ,Virus Replication ,Argentine hemorrhagic fever ,Antiviral Agents ,Virus ,Ciencias Biológicas ,03 medical and health sciences ,chemistry.chemical_compound ,Multiplicity of infection ,LEFLUNOMIDE ,Virology ,Chlorocebus aethiops ,Nitriles ,Ribavirin ,medicine ,Animals ,Humans ,Vero Cells ,A771726 ,PYRIMIDINE INHIBITOR ,Aniline Compounds ,Junin virus ,Arenavirus ,Dose-Response Relationship, Drug ,biology ,Chemistry ,Drug Synergism ,ANTIVIRAL ACTIVITY ,Viral Load ,biology.organism_classification ,medicine.disease ,ARENAVIRUS ,030104 developmental biology ,Infectious Diseases ,Viral replication ,A549 Cells ,Crotonates ,Dihydroorotate dehydrogenase ,RNA, Viral ,HOST TARGET ,Virología ,CIENCIAS NATURALES Y EXACTAS - Abstract
The aim of this study was to investigate the effect of A771726, the active metabolite of leflunomide, (CONICET-UBA), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad against the infection with Junín virus (JUNV), agent of Argentine hemorrhagic fever (AHF). The treatment with non-cytotoxic concentrations of A771726 of Vero and A549 cells infected with JUNV inhibited virus replication in a dose-dependent manner, as determined by virus yield reduction assay. The antiviral effectiveness of A771726 was not importantly affected by the multiplicity of infection and the virus strain. Moreover, the combination of A771726 and ribavirin had a significantly more potent antiviral activity than each single drug treatment. Mechanistic studies showed that the main action of A771726 is exerted before 6 h of JUNV infection. Accordingly, inhibition of viral RNA synthesis was detected in treated infected cells by real time RT-PCR. The exogenous addition of uridine or orotic acid produced a partial reversal of the inhibitory effect of A771726 on infective virus production whereas a total reversion was detected on JUNV RNA synthesis, probably by restoration of the enzymatic activity of dihydroorotate dehydrogenase (DHODH) and the intracellular pyrimidine pools. In conclusion, these results suggest that the antiviral target would be viral RNA synthesis through pyrimidine depletion, but any other effect of the compound on JUNV infection cannot be excluded. This study opens the possibility of the therapeutic application of a wide spectrum host-targeted compound alone or in combination with ribavirin to combat AHF as well as other human pathogenic arenaviruses. Fil: Sepúlveda, Claudia Soledad. Universidad de Buenos Aires; Argentina Fil: Garcia, Cybele. Universidad de Buenos Aires; Argentina Fil: Damonte, Elsa Beatriz. Universidad de Buenos Aires; Argentina
- Published
- 2018
- Full Text
- View/download PDF
18. Evaluation of cell viability dyes in antiviral assays with RNA viruses that exhibit different cytopathogenic properties
- Author
-
W. Joseph Evans, Sean Wright, Chris Peterson, Nathan R. Clyde, Brett L. Hurst, Craig W. Day, Kie-Hoon Jung, and Donald F. Smee
- Subjects
0301 basic medicine ,Neutral red ,Cell Survival ,030106 microbiology ,Biology ,Favipiravir ,Virus Replication ,Antiviral Agents ,Article ,Virus ,Microbiology ,03 medical and health sciences ,chemistry.chemical_compound ,Cytopathogenic Effect, Viral ,Virology ,Chlorocebus aethiops ,Oxazines ,medicine ,Animals ,RNA Viruses ,Viability assay ,Coloring Agents ,Vero Cells ,Cytopathic effect ,Junin virus ,Dengue Virus ,Xanthenes ,chemistry ,Viral replication ,Viruses ,Vero cell ,Chikungunya virus ,Tiazofurin ,medicine.drug - Abstract
Studies were conducted to determine the performance of four dyes in assessing antiviral activities of compounds against three RNA viruses with differing cytopathogenic properties. Dyes included alamarBlue® measured by absorbance (ALB-A) and fluorescence (ALB-F), neutral red (NR), Viral ToxGlo™ (VTG), and WST-1. Viruses were chikungunya, dengue type 2, and Junin, which generally cause 100, 80-90, and 50% maximal cytopathic effect (CPE), respectively, in Vero or Vero 76 cells Compounds evaluated were 6-azauridine, BCX-4430, 3-deazaguanine, EICAR, favipiravir, infergen, mycophenolic acid (MPA), ribavirin, and tiazofurin. The 50% virus-inhibitory (EC50) values for each inhibitor and virus combination did not vary significantly based on the dye used. However, dyes varied in distinguishing the vitality of virus-infected cultures when not all cells were killed by virus infection. For example, VTG uptake into dengue-infected cells was nearly 50% when visual examination showed only 10-20% cell survival. ALB-A measured infected cell viability differently than ALB-F as follows: 16% versus 32% (dengue-infected), respectively, and 51% versus 72% (Junin-infected), respectively. Cytotoxicity (CC50) assays with dyes in uninfected proliferating cells produced similar CC50 values for EICAR (1.5-8.9μM) and MPA (0.8-2.5μM). 6-Azauridine toxicity was 6.1-17.5μM with NR, VTG, and WST-1, compared to 48-92μM with ALB-A and ALB-F (P
- Published
- 2017
- Full Text
- View/download PDF
19. Conglomeration of highly antigenic nucleoproteins to inaugurate a heterosubtypic next generation vaccine candidate against Arenaviridae family
- Author
-
Rahima Akter, Md. Nazmul Hossain, Tahera Lasker, Kazi Faizul Azim, Bhuiyan Of, Mahmudul Hasan, and Hia Mm
- Subjects
Receptor complex ,education.field_of_study ,viruses ,Population ,Biology ,medicine.disease_cause ,biology.organism_classification ,Virology ,Virus ,Epitope ,Conserved sequence ,Lassa virus ,Junin virus ,medicine ,education ,Lujo virus - Abstract
Arenaviral infections often resulting in lethal Hemorrhagic Fevers (HF) affect primarily African and South American regions. To date, there is no FDA-approved licensed vaccine against arenaviruses and treatments have been limited to supportive therapies. Hence, the study was employed to design a highly immunogenic heterosubtypic vaccine candidate against Arenaviridae family. The whole proteomes of Lassa virus (LASV), Lymphocytic Chorio Meningitis Virus (LCMV), Lujo virus and Guanarito virus were retrieved from NCBI database and assessed to determine the most antigenic viral proteins. Only the conserved sequences were used for T cell and B cell epitope prediction to ensure protective response against a wide range of viral strains. For each virus, nucleoproteins were identified as most antigenic which generated a plethora of antigenic epitopes. The proposed epitopes were highly conserved (up to 100%) and showed high cumulative population coverage. Moreover, results revealed that among the top epitopes, T cell epitope GWPYIGSRS were conserved in Argentine mammarenavirus (Junin virus) and Brazilian mammarenavirus (Sabia virus), while B cell epitope NLLYKICLSG were conserved in Bolivian mammarenavirus (Machupo virus) and Brazilian mammarenavirus (Sabia virus), indicating the possibility of final vaccine constructs to confer broad range immunity in the host. A total 3 constructs were designed by the combination of top epitopes from each protein along with suitable adjuvant and linkers. Different physicochemical properties revealed the superiority of construct V1 in terms of safety and efficacy. Docking analysis of the refined vaccine structure with different MHC molecules and human immune receptors were also biologically significant. The vaccine receptor complex (V1-TLR3) showed minimal deformability at molecular level. Moreover, construct V1 was compatible for insertion into pET28a(+) vector and heterologous cloning in E. coli srain K12. However, the results were based on different sequence analysis and various immune databases. Further wet lab based studies using model animals are highly recommended for the experimental validation of the designed vaccine candidates.
- Published
- 2019
- Full Text
- View/download PDF
20. Junin Virus Triggers Macrophage Activation and Modulates Polarization According to Viral Strain Pathogenicity
- Author
-
Aída Oryza López Ortiz, Víctor Romanowski, Andrea E. Errasti, Juan Pablo Gorgojo, María Florencia Ferrer, Eugenio Antonio Carrera Silva, Ricardo Martín Gómez, María Eugenia Rodríguez, Pablo Javier Thomas, and Nancy Charo
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Biología ,Biotecnología relacionada con la Salud ,MACROPHAGE ACTIVATION ,medicine.disease_cause ,0302 clinical medicine ,Cricetinae ,Chlorocebus aethiops ,Immunology and Allergy ,purl.org/becyt/ford/3.4 [https] ,Original Research ,human macrophages ,junin virus ,Cytokine ,TAM RECEPTORS ,B7-1 Antigen ,Cytokines ,purl.org/becyt/ford/3 [https] ,MACROPHAGE POLARIZATION ,lcsh:Immunologic diseases. Allergy ,CIENCIAS MÉDICAS Y DE LA SALUD ,macrophage polarization ,Immunology ,Macrophage polarization ,Biology ,Argentine hemorrhagic fever ,TAM receptors ,Hemorrhagic Fever, American ,HUMAN MACROPHAGES ,Virus ,Biotecnología de la Salud ,Microbiology ,03 medical and health sciences ,Immune system ,Species Specificity ,macrophage activation ,medicine ,Animals ,Humans ,Vero Cells ,Ciencias Exactas ,Macrophages ,HLA-DR Antigens ,MERTK ,medicine.disease ,biology.organism_classification ,030104 developmental biology ,Lassa virus ,Junin virus ,Ciencias Médicas ,IFN-I ,B7-2 Antigen ,lcsh:RC581-607 ,JUNIN VIRUS ,030215 immunology - Abstract
The New World arenavirus Junin (JUNV) is the etiological agent of Argentine hemorrhagic fever (AHF). Previous studies of human macrophage infection by the Old-World arenaviruses Mopeia and Lassa showed that while the non-pathogenic Mopeia virus replicates and activates human macrophages, the pathogenic Lassa virus replicates but fails to activate human macrophages. Less is known in regard to the impact of New World arenavirus infection on the human macrophage immune response. Macrophage activation is critical for controlling infections but could also be usurped favoring immune evasion. Therefore, it is crucial to understand how the JUNV infection modulates macrophage plasticity to clarify its role in AHF pathogenesis. With this aim in mind, we compared infection with the attenuated Candid 1 (C#1) or the pathogenic P strains of the JUNV virus in human macrophage cultures. The results showed that both JUNV strains similarly replicated and induced morphological changes as early as 1 day post-infection. However, both strains differentially induced the expression of CD71, the receptor for cell entry, the activation and maturation molecules CD80, CD86, and HLA-DR and selectively modulated cytokine production. Higher levels of TNF-α, IL-10, and IL-12 were detected with C#1 strain, while the P strain induced only higher levels of IL-6. We also found that C#1 strain infection skewed macrophage polarization to M1, whereas the P strain shifted the response to an M2 phenotype. Interestingly, the MERTK receptor, that negatively regulates the immune response, was down-regulated by C#1 strain and up-regulated by P strain infection. Similarly, the target genes of MERTK activation, the cytokine suppressors SOCS1 and SOCS3, were also increased after P strain infection, in addition to IRF-1, that regulates type I IFN levels, which were higher with C#1 compared with P strain infection. Together, this differential activation/polarization pattern of macrophages elicited by P strain suggests a more evasive immune response and may have important implications in the pathogenesis of AHF and underpinning the development of new potential therapeutic strategies, Facultad de Ciencias Exactas, Instituto de Biotecnologia y Biologia Molecular, Centro de Investigación y Desarrollo en Fermentaciones Industriales
- Published
- 2019
- Full Text
- View/download PDF
21. Junín Virus Promotes Autophagy To Facilitate the Virus Life Cycle
- Author
-
Nélida A. Candurra, Laura Ruth Delgui, Julieta Suyay Roldan, and María Isabel Colombo
- Subjects
Time Factors ,Recombinant Fusion Proteins ,ATG5 ,Green Fluorescent Proteins ,Immunology ,Cellular homeostasis ,Argentine hemorrhagic fever ,Virus Replication ,Microbiology ,Virus ,ARENAVIRIDAE ,purl.org/becyt/ford/1 [https] ,Ciencias Biológicas ,03 medical and health sciences ,Viral life cycle ,Virology ,Chlorocebus aethiops ,medicine ,Autophagy ,Animals ,Humans ,BECLIN 1 ,PROVIRAL ROLE ,purl.org/becyt/ford/1.6 [https] ,Vero Cells ,030304 developmental biology ,0303 health sciences ,Arenavirus ,Junin virus ,biology ,Staining and Labeling ,030302 biochemistry & molecular biology ,JUNÍN VIRUS ,biology.organism_classification ,medicine.disease ,Virus-Cell Interactions ,Microscopy, Fluorescence ,A549 Cells ,Insect Science ,Host-Pathogen Interactions ,REPLICATION ,INNATE IMMUNITY ,AUTOPHAGY ,Microtubule-Associated Proteins ,Virología ,CIENCIAS NATURALES Y EXACTAS - Abstract
Junín virus (JUNV), a member of the family Arenaviridae, is the etiological agent of Argentine hemorrhagic fever (AHF), a potentially deadly endemic-epidemic disease affecting the population of the most fertile farming land of Argentina. Autophagy is a degradative process with a crucial antiviral role; however, several viruses subvert the pathway to their benefit. We determined the role of autophagy in JUNV-infected cells by analyzing LC3, a cytoplasmic protein (LC3-I) that becomes vesicle membrane associated (LC3-II) upon induction of autophagy. Cells overexpressing enhanced green fluorescent protein (EGFP)-LC3 and infected with JUNV showed an increased number of LC3 punctate structures, similar to those obtained after starvation or bafilomycin A1 treatment, which leads to autophagosome induction or accumulation, respectively. We also monitored the conversion of LC3-I to LC3-II, observing LC3-II levels in JUNV-infected cells similar to those observed in starved cells. Additionally, we kinetically studied the number of LC3 dots after JUNV infection and found that the virus activated the pathway as early as 2 h postinfection (p.i.), whereas the UV-inactivated virus did not induce the pathway. Cells subjected to starvation or pretreated with rapamycin, a pharmacological autophagy inductor, enhanced virus yield. Also, we assayed the replication capacity of JUNV in Atg5 knockout or Beclin 1 knockdown cells (both critical components of the autophagic pathway) and found a significant decrease in JUNV replication. Taken together, our results constitute the first study indicating that JUNV infection induces an autophagic response, which is functionally required by the virus for efficient propagation. IMPORTANCE Mammalian arenaviruses are zoonotic viruses that cause asymptomatic and persistent infections in their rodent hosts but may produce severe and lethal hemorrhagic fevers in humans. Currently, there are neither effective therapeutic options nor effective vaccines for viral hemorrhagic fevers caused by human-pathogenic arenaviruses, except the vaccine Candid no. 1 against Argentine hemorrhagic fever (AHF), licensed for human use in areas of endemicity in Argentina. Since arenaviruses remain a severe threat to global public health, more in-depth knowledge of their replication mechanisms would improve our ability to fight these viruses. Autophagy is a lysosomal degradative pathway involved in maintaining cellular homeostasis, representing powerful anti-infective machinery. We show, for the first time for a member of the family Arenaviridae, a proviral role of autophagy in JUNV infection, providing new knowledge in the field of host-virus interaction. Therefore, modulation of virus-induced autophagy could be used as a strategy to block arenavirus infections. Fil: Roldan, Julieta Suyay. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Departamento de Química Biológica. Laboratorio de Virología; Argentina. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Ciudad Universitaria. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales; Argentina Fil: Candurra, Nélida Alicia. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Ciudad Universitaria. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales; Argentina. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Departamento de Química Biológica. Laboratorio de Virología; Argentina Fil: Colombo, Maria Isabel. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro Científico Tecnológico Conicet - Mendoza. Instituto de Histología y Embriología de Mendoza Dr. Mario H. Burgos. Universidad Nacional de Cuyo. Facultad de Ciencias Médicas. Instituto de Histología y Embriología de Mendoza Dr. Mario H. Burgos; Argentina Fil: Delgui, Laura Ruth. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro Científico Tecnológico Conicet - Mendoza. Instituto de Histología y Embriología de Mendoza Dr. Mario H. Burgos. Universidad Nacional de Cuyo. Facultad de Ciencias Médicas. Instituto de Histología y Embriología de Mendoza Dr. Mario H. Burgos; Argentina
- Published
- 2019
- Full Text
- View/download PDF
22. [Arenavirus research and antiviral candidate]
- Author
-
Urata Shuzo and Hideki Tani
- Subjects
Transcription, Genetic ,viruses ,Chapare virus ,Genome, Viral ,medicine.disease_cause ,Lymphocytic choriomeningitis ,Antiviral Agents ,Virus ,Disease Outbreaks ,Drug Discovery ,medicine ,Arenaviridae Infections ,Humans ,Lassa fever ,Lujo virus ,Arenavirus ,biology ,Research ,Virion ,virus diseases ,Viral Vaccines ,General Medicine ,medicine.disease ,biology.organism_classification ,Virology ,Africa, Western ,Lassa virus ,Junin virus - Abstract
Arenavirus is a genetic term for viruses belonging to the family Arenaviridae and is presented from lymphocytic choriomeningitis virus (LCMV), which shows almost no pathogenicity to humans, to Lassa virus, Junin virus, Machupo virus, Chapare virus, Lujo virus, Sabia virus, and Guanarito virus, which shows high pathogenicity to humans. These viruses except for LCMV are risk group 4 pathogens specified by World Health Organization. Based on this designation, it is designated as Class I pathogens in Japan. Although there have been no reports excluding one imported case of the Lassa fever patient, it is not surprising whenever imported cases occur in our country. Considering the disease severity and mortality rate, it is an urgent matter to develop vaccines and therapeutic drugs in endemic areas, and maintenances of these are also important in countries other than endemic areas. However, basic research on highly pathogenic arenavirus infections and development of therapeutic drugs are not easily progressed, because handling in highly safe research facilities is indispensable. In this article, we will outline the current knowledge from the recent basic research on arenavirus to the development situation of antivirals against arenaviruses.
- Published
- 2019
23. Comparison of the Innate Immune Responses to Pathogenic and Nonpathogenic Clade B New World Arenaviruses
- Author
-
Stefan Kunz, Chiara Fedeli, Rebecca Möller, Hector Moreno, and Gisa Gerold
- Subjects
viruses ,Immunology ,Virulence ,Biology ,Virus Replication ,Microbiology ,Virus ,Cell Line ,03 medical and health sciences ,eIF-2 Kinase ,Interferon ,Virology ,medicine ,Humans ,Immunologic Factors ,Arenaviruses, New World ,030304 developmental biology ,0303 health sciences ,Arenavirus ,Innate immune system ,Junin virus ,030306 microbiology ,Gene Expression Profiling ,biology.organism_classification ,Protein kinase R ,Immunity, Innate ,3. Good health ,Viral replication ,Insect Science ,Interferon Type I ,Pathogenesis and Immunity ,medicine.drug - Abstract
The New World (NW) arenaviruses are a diverse group of zoonotic viruses, including several causative agents of severe hemorrhagic fevers in humans. All known human-pathogenic NW arenaviruses belong to clade B, where they group into sublineages with phylogenetically closely related nonpathogenic viruses, e.g., the highly pathogenic Junin (JUNV) and Machupo viruses with the nonpathogenic Tacaribe virus (TCRV). Considering the close genetic relationship of nonpathogenic and pathogenic NW arenaviruses, the identification of molecular determinants of virulence is of great importance. The host cell’s innate antiviral defense represents a major barrier for zoonotic infection. Here, we performed a side-by-side comparison of the innate immune responses against JUNV and TCRV in human cells. Despite similar levels of viral replication, infection with TCRV consistently induced a stronger type I interferon (IFN-I) response than JUNV infection did. Transcriptome profiling revealed upregulation of a largely overlapping set of interferon-stimulated genes in cells infected with TCRV and JUNV. Both viruses were relatively insensitive to IFN-I treatment of human cells and induced similar levels of apoptosis in the presence or absence of an IFN-I response. However, in comparison to JUNV, TCRV induced stronger activation of the innate sensor double-strand RNA-dependent protein kinase R (PKR), resulting in phosphorylation of eukaryotic translation initiation factor eIF2α. Confocal microscopy studies revealed similar subcellular colocalizations of the JUNV and TCRV viral replication-transcription complexes with PKR. However, deletion of PKR by CRISPR/Cas9 hardly affected JUNV but promoted TCRV multiplication, providing the first evidence for differential innate recognition and control of pathogenic and nonpathogenic NW arenaviruses by PKR. IMPORTANCE New World (NW) arenaviruses are a diverse family of emerging zoonotic viruses that merit significant attention as important public health problems. The close genetic relationship of nonpathogenic NW arenaviruses with their highly pathogenic cousins suggests that few mutations may be sufficient to enhance virulence. The identification of molecular determinants of virulence of NW arenaviruses is therefore of great importance. Here we undertook a side-by-side comparison of the innate immune responses against the highly pathogenic Junin virus (JUNV) and the related nonpathogenic Tacaribe virus (TCRV) in human cells. We consistently found that TCRV induces a stronger type I interferon (IFN-I) response than JUNV. Transcriptome profiling revealed an overlapping pattern of IFN-induced gene expression and similar low sensitivities to IFN-I treatment. However, the double-stranded RNA (dsRNA)-dependent protein kinase R (PKR) contributed to the control of TCRV, but not JUNV, providing the first evidence for differential innate recognition and control of JUNV and TCRV.
- Published
- 2019
24. The antiviral activity of iota-, kappa-, and lambda-carrageenan against COVID-19: A critical review
- Author
-
Andri Frediansyah
- Subjects
Microbiology (medical) ,Nasal spray ,Epidemiology ,viruses ,Dengue virus ,medicine.disease_cause ,Carrageenan ,Virus ,Article ,03 medical and health sciences ,0302 clinical medicine ,Iota-carrageenan ,medicine ,030212 general & internal medicine ,Coronavirus ,030219 obstetrics & reproductive medicine ,biology ,business.industry ,SARS-CoV-2 ,Rabies virus ,Public Health, Environmental and Occupational Health ,virus diseases ,COVID-19 ,respiratory system ,biology.organism_classification ,Virology ,Clinical trial ,Infectious Diseases ,Herpes simplex virus ,Vesicular stomatitis virus ,Junin virus ,Respiratory virus ,Public aspects of medicine ,RA1-1270 ,business - Abstract
Objective There is no specific antiviral treatment available for coronavirus disease 2019 (COVID-19). Among the possible natural constituents is carrageenan, a polymer derived from marine algae that possesses a variety of antiviral properties. The purpose of this review was to summarize the evidence supporting carrageenan subtypes' antiviral activity against the emerging severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative agent of COVID-19. Methods PubMed/MEDLINE and Google Scholar searches were conducted for publications using the terms ‘carrageenan', ‘iota carrageenan', ‘kappa carrageenan', lambda-carrageenan', 'coronavirus', ‘common cold', 'rhinovirus', and ‘SARS-CoV-2′ search was also done in grey literature to increase our understanding. A search for the word "carrageenan" was also carried out. Most of the publications were discussed in narrative. Results Carrageenan has been shown to have potent antiviral activity against both coronaviruses (coronavirus NL63, SARS-CoV-2) and non-coronaviruses such as dengue virus, herpes simplex virus, cytomegalovirus, vaccinia virus, vesicular stomatitis virus, sindbis virus, human immunodeficiency virus, influenza virus, human papillomavirus, rabies virus, junin virus, tacaribe virus, African swine fever, bovine herpes virus, suid herpes virus, and rhinovirus. No in vivo study has been conducted using carrageenan as an anti-SARS-CoV-2 agent. The majority of the in vivo research was done on influenza, a respiratory virus that causes common cold together with coronavirus. Thus, various clinical trials were conducted to determine the transferability of these in vitro data to clinical effectiveness against SARS-CoV-2. When combined with oral ivermectin, nasally administered iota-carrageenan improved outcome in COVID-19 patients. It is still being tested in clinics for single-dose administration. Conclusion Though the carrageenan exhibited potent antiviral activity against SARS-CoV-2 and was used to treat COVID-19 under emergency protocol in conjunction with oral medications such as ivermectin, there is no solid evidence from clinical trials to support its efficacy. Thus, clinical trials are required to assess its efficacy for COVID-19 treatment prior to broad application.
- Published
- 2021
25. Myristoylation of the Arenavirus Envelope Glycoprotein Stable Signal Peptide Is Critical for Membrane Fusion but Dispensable for Virion Morphogenesis
- Author
-
Joanne York and Jack H. Nunberg
- Subjects
0301 basic medicine ,viruses ,Immunology ,Protein Sorting Signals ,Biology ,Myristic Acid ,Microbiology ,Virus ,03 medical and health sciences ,Viral Envelope Proteins ,Viral envelope ,Virology ,Chlorocebus aethiops ,Animals ,Vero Cells ,Glycoproteins ,Sequence Deletion ,Myristoylation ,Junin virus ,Membrane Glycoproteins ,Arenavirus ,030102 biochemistry & molecular biology ,Virus Assembly ,Structure and Assembly ,Virion ,Lipid bilayer fusion ,Virus Internalization ,biology.organism_classification ,Reverse Genetics ,030104 developmental biology ,Amino Acid Substitution ,Virion assembly ,Insect Science ,Mutant Proteins ,Protein Processing, Post-Translational ,Membrane Fusion Activity - Abstract
Arenaviruses are responsible for severe and often fatal hemorrhagic disease. In the absence of effective antiviral therapies and vaccines, these viruses pose serious threats to public health and biodefense. Arenaviruses enter the host cell by fusion of the viral and endosomal membranes, a process mediated by the virus envelope glycoprotein GPC. Unlike other class I viral fusion proteins, GPC retains its stable signal peptide (SSP) as an essential third subunit in the mature complex. SSP spans the membrane twice and is myristoylated at its cytoplasmic N terminus. Mutations that abolish SSP myristoylation have been shown to reduce pH-induced cell-cell fusion activity of ectopically expressed GPC to ∼20% of wild-type levels. In order to examine the role of SSP myristoylation in the context of the intact virus, we used reverse genetics to generate Junín viruses (Candid #1 isolate) in which the critical glycine-2 residue in SSP was either replaced by alanine (G2A) or deleted (ΔG2). These mutant viruses produced smaller foci of infection in Vero cells and showed an ∼5-fold reduction in specific infectivity, commensurate with the defect in cell-cell fusion. However, virus assembly and GPC incorporation into budded virions were unaffected. Our findings suggest that the myristate moiety is cryptically disposed in the prefusion GPC complex and may function late in the fusion process to promote merging of the viral and cellular membranes. IMPORTANCE Hemorrhagic fever arenaviruses pose significant threats to public health and biodefense. Arenavirus entry into the host cell is promoted by the virus envelope glycoprotein GPC. Unlike other viral envelope glycoproteins, GPC contains a myristoylated stable signal peptide (SSP) as an essential third subunit. Myristoylation has been shown to be important for the membrane fusion activity of recombinantly expressed GPC. Here, we use reverse genetics to study the role of SSP myristoylation in the context of the intact virion. We find that nonmyristoylated GPC mutants of the Candid #1 strain of Junín virus display a commensurate deficiency in their infectivity, albeit without additional defects in virion assembly and budding. These results suggest that SSP myristoylation may function late in the fusion process to facilitate merging of the viral and cellular membranes. Antiviral agents that target this novel aspect of GPC membrane fusion may be useful in the treatment of arenavirus hemorrhagic fevers.
- Published
- 2016
- Full Text
- View/download PDF
26. Structure-function relationship of the mammarenavirus envelope glycoprotein
- Author
-
Shaobo Wang, Gengfu Xiao, Zheng Zhou, Wei Wang, and Leike Zhang
- Subjects
0301 basic medicine ,Signal peptide ,Glycosylation ,Immunology ,Review ,Biology ,medicine.disease_cause ,Virus ,03 medical and health sciences ,chemistry.chemical_compound ,Viral Envelope Proteins ,Virology ,medicine ,Animals ,Arenaviridae Infections ,Humans ,Arenaviridae ,chemistry.chemical_classification ,Lipid bilayer fusion ,biology.organism_classification ,Fusion protein ,030104 developmental biology ,Lassa virus ,chemistry ,Biochemistry ,Junin virus ,Molecular Medicine ,Glycoprotein - Abstract
Mammarenaviruses, including lethal pathogens such as Lassa virus and Junín virus, can cause severe hemorrhagic fever in humans. Entry is a key step for virus infection, which starts with binding of the envelope glycoprotein (GP) to receptors on target cells and subsequent fusion of the virus with target cell membranes. The GP precursor is synthesized as a polypeptide, and maturation occurs by two cleavage events, yielding a tripartite GP complex (GPC) formed by a stable signal peptide (SSP), GP1 and GP2. The unique retained SSP interacts with GP2 and plays essential roles in virion maturation and infectivity. GP1 is responsible for binding to the cell receptor, and GP2 is a class I fusion protein. The native structure of the tripartite GPC is unknown. GPC is critical for the receptor binding, membrane fusion and neutralization antibody recognition. Elucidating the molecular mechanisms underlining the structure–function relationship of the three subunits is the key for understanding their function and can facilitate novel avenues for combating virus infections. This review summarizes the basic aspects and recent research of the structure–function relationship of the three subunits. We discuss the structural basis of the receptor-binding domain in GP1, the interaction between SSP and GP2 and its role in virion maturation and membrane fusion, as well as the mechanism by which glycosylation stabilizes the GPC structure and facilitates immune evasion. Understanding the molecular mechanisms involved in these aspects will contribute to the development of novel vaccines and treatment strategies against mammarenaviruses infection. [Image: see text]
- Published
- 2016
- Full Text
- View/download PDF
27. The New World arenavirus Tacaribe virus induces caspase-dependent apoptosis in infected cells
- Author
-
David A. Jackson, Svenja Wolff, Bjoern Meyer, Andreas Kaufmann, Stephan Becker, Allison Groseth, Thomas Strecker, and University of St Andrews. School of Biology
- Subjects
0301 basic medicine ,Ultraviolet Rays ,New World Arenavirus ,QH301 Biology ,viruses ,Primary Cell Culture ,NDAS ,Apoptosis ,Virus Replication ,Virus ,Caspase-Dependent Apoptosis ,Amino Acid Chloromethyl Ketones ,QH301 ,03 medical and health sciences ,SDG 3 - Good Health and Well-being ,Tubulin ,Cell Line, Tumor ,Virology ,Chlorocebus aethiops ,Animals ,Humans ,Vero Cells ,Arenaviruses, New World ,Caspase ,QR355 ,Junin virus ,Arenavirus ,biology ,Macrophages ,biology.organism_classification ,030104 developmental biology ,Gene Expression Regulation ,Viral replication ,Caspases ,Host-Pathogen Interactions ,Hepatocytes ,biology.protein ,Camptothecin ,Poly(ADP-ribose) Polymerases ,QR355 Virology ,Signal Transduction - Abstract
This work was supported in part by fellowships from the Jürgen Manchot Stiftung (S.W.; http://www.manchot.org), the German Research Council [SFB 1021 TP A04 (A.K.), SFB 1021 TP B03 (S.B., S.W.) and SFB 1021 TP B05 (T.S.); http://www.uni-marburg.de/sfb1021], the Canadian Institutes of Health Research (A.G.; http://www.cihr-irsc.gc.ca) and a Medical Research Council Centenary Travel Award (B.M.). The Arenaviridae is a diverse and growing family of viruses that already includes more than 25 distinct species. While some of these viruses have a significant impact on public health, others appear to be non-pathogenic. At present little is known about the host cell responses to infection with different arenaviruses, particularly those found in the New World; however, apoptosis is known to play an important role in controlling infection of many viruses. Here we show that infection with Tacaribe virus (TCRV), which is widely considered the prototype for non-pathogenic arenaviruses, leads to stronger induction of apoptosis than does infection with its human-pathogenic relative Junín virus. TCRV-induced apoptosis occurred in several cell types during late stages of infection and was shown to be caspase-dependent, involving the activation of caspases 3, 7, 8 and 9. Further, UV-inactivated TCRV did not induce apoptosis, indicating that the activation of this process is dependent on active viral replication/transcription. Interestingly, when apoptosis was inhibited, growth of TCRV was not enhanced, indicating that apoptosis does not have a direct negative effect on TCRV infection in vitro. Taken together, our data identify and characterize an important virus-host cell interaction of the prototypic, non-pathogenic arenavirus TCRV, which provides important insight into the growing field of arenavirus research aimed at better understanding the diversity in responses to different arenavirus infections and their functional consequences. Postprint
- Published
- 2016
- Full Text
- View/download PDF
28. Machupo Virus Expressing GPC of the Candid#1 Vaccine Strain of Junin Virus Is Highly Attenuated and Immunogenic
- Author
-
Steven Hallam, Takaaki Koma, Aida G. Walker, Cheng Huang, Jeanon N. Smith, Alexey Seregin, Michael Patterson, Slobodan Paessler, Payal D. Maharaj, and Milagros Miller
- Subjects
0301 basic medicine ,Molecular Sequence Data ,030106 microbiology ,Immunology ,Virulence ,Vaccines, Attenuated ,Argentine hemorrhagic fever ,Microbiology ,Genomic Instability ,Hemorrhagic Fever, American ,Virus ,03 medical and health sciences ,Viral Envelope Proteins ,Virology ,Vaccines and Antiviral Agents ,medicine ,Animals ,Arenaviruses, New World ,Recombination, Genetic ,chemistry.chemical_classification ,Membrane Glycoproteins ,biology ,Histocytochemistry ,Viral Vaccine ,Body Weight ,Temperature ,Animal Structures ,Viral Vaccines ,Sequence Analysis, DNA ,biology.organism_classification ,medicine.disease ,Survival Analysis ,Mice, Inbred C57BL ,Disease Models, Animal ,Hemorrhagic Fevers ,030104 developmental biology ,chemistry ,Insect Science ,Junin virus ,Bolivian hemorrhagic fever ,Glycoprotein - 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.
- Published
- 2016
- Full Text
- View/download PDF
29. A potent Lassa virus antiviral targets an arenavirus virulence determinant
- Author
-
James R. Burgeson, Kristin Bedard, Sean M. Amberg, Megan Files, Kevin F. Jones, Shawn P. Iadonato, Marcus J. Korth, Dima N. Gharaibeh, Dongcheng Dai, Amy L. Moore, Ikenna G. Madu, Shanthakumar R. Tyavanagimatt, Kieh-Hoon Jung, Brian B. Gowen, and PLoS
- Subjects
0301 basic medicine ,RNA viruses ,Viral Diseases ,Cell Lines ,viruses ,Dairy Science ,medicine.disease_cause ,Pathology and Laboratory Medicine ,Biochemistry ,Mice ,Viral Envelope Proteins ,Serial passage ,Zoonoses ,Chlorocebus aethiops ,Medicine and Health Sciences ,Amino Acids ,Biology (General) ,Lassa fever ,biology ,Virulence ,Organic Compounds ,Machupo Virus ,3. Good health ,Chemistry ,Infectious Diseases ,Medical Microbiology ,Viral Pathogens ,Viruses ,Physical Sciences ,Biological Cultures ,Pathogens ,Research Article ,Neglected Tropical Diseases ,QH301-705.5 ,030106 microbiology ,Immunology ,Argentine hemorrhagic fever ,Research and Analysis Methods ,Antiviral Agents ,Microbiology ,Virus ,03 medical and health sciences ,Lassa Fever ,Viral entry ,Virology ,Drug Resistance, Viral ,Genetics ,medicine ,Animals ,Humans ,Lassa virus ,Molecular Biology ,Vero Cells ,Microbial Pathogens ,Arenavirus ,Biology and life sciences ,Hemorrhagic Fever Viruses ,Organic Chemistry ,Organisms ,Hemorrhagic Fevers ,Chemical Compounds ,Proteins ,RC581-607 ,biology.organism_classification ,medicine.disease ,Tropical Diseases ,Arenaviruses ,030104 developmental biology ,HEK293 Cells ,Amino Acid Substitution ,Animal Sciences ,Junin virus ,Mutation ,Parasitology ,Immunologic diseases. Allergy - Abstract
Arenaviruses are a significant cause of hemorrhagic fever, an often-fatal disease for which there is no approved antiviral therapy. Lassa fever in particular generates high morbidity and mortality in West Africa, where the disease is endemic, and a recent outbreak in Nigeria was larger and more geographically diverse than usual. We are developing LHF-535, a small-molecule viral entry inhibitor that targets the arenavirus envelope glycoprotein, as a therapeutic candidate for Lassa fever and other hemorrhagic fevers of arenavirus origin. Using a lentiviral pseudotype infectivity assay, we determined that LHF-535 had sub-nanomolar potency against the viral envelope glycoproteins from all Lassa virus lineages, with the exception of the glycoprotein from the LP strain from lineage I, which was 100-fold less sensitive than that of other strains. This reduced sensitivity was mediated by a unique amino acid substitution, V434I, in the transmembrane domain of the envelope glycoprotein GP2 subunit. This position corresponds to the attenuation determinant of Candid#1, a live-attenuated Junín virus vaccine strain used to prevent Argentine hemorrhagic fever. Using a virus-yield reduction assay, we determined that LHF-535 potently inhibited Junín virus, but not Candid#1, and the Candid#1 attenuation determinant, F427I, regulated this difference in sensitivity. We also demonstrated that a daily oral dose of LHF-535 at 10 mg/kg protected mice from a lethal dose of Tacaribe virus. Serial passage of Tacaribe virus in LHF-535-treated Vero cells yielded viruses that were resistant to LHF-535, and the majority of drug-resistant viruses exhibited attenuated pathogenesis. These findings provide a framework for the clinical development of LHF-535 as a broad-spectrum inhibitor of arenavirus entry and provide an important context for monitoring the emergence of drug-resistant viruses., Author summary Lassa fever is a viral hemorrhagic fever disease that is transmitted to humans primarily through contact with the urine or feces of infected rodents. The disease is endemic in West Africa, and an unusually large outbreak occurred in Nigeria in early 2018. The case fatality rate was 25% among confirmed cases, underscoring the need for an effective antiviral therapy. Here, we evaluated the small-molecule drug LHF-535, which targets the arenavirus envelope glycoprotein, for broad-spectrum activity against Lassa viruses of different lineages and related arenaviruses that cause hemorrhagic fever diseases in South America. We also selected for LHF-535-resistant viruses and characterized their genotype and phenotype. Using a combination of surrogate systems and wild-type viruses, we determined that all tested Lassa virus strains and New World hemorrhagic fever arenaviruses were sensitive to LHF-535. Sensitivity to the drug was modulated by specific amino acid changes in the viral envelope glycoprotein, and the majority of emerging drug-resistant viruses were attenuated for virulence. Similarly, the live-attenuated vaccine strain for Argentine hemorrhagic fever was also resistant to LHF-535. These findings indicate that LHF-535 targets a viral virulence determinant, the mutation of which may result in the emergence of drug-resistant viruses, but with reduced capacity for virulence.
- Published
- 2018
30. Assessing cross-reactivity of Junín virus-directed neutralizing antibodies
- Author
-
Delia Enria, Julia Holzerland, Sandra Myriam Cordo, Allison Groseth, Kevin Schnepel, Irke Waßmann, Thomas Hoenen, Julia Brignone, Kyle Shifflett, Patrick Bohn, Thomas C. Mettenleiter, Linus Bostedt, Anne Leske, and Ana M. Briggiler
- Subjects
0301 basic medicine ,New World Arenavirus ,viruses ,030106 microbiology ,TRANSCRIPTION AND REPLICATION COMPETENT VIRUS-LIKE PARTICLE (TRVLP) ASSAY ,Cross Reactions ,medicine.disease_cause ,Antibodies, Viral ,Virus Replication ,Cross-reactivity ,Neutralization ,Virus ,Hemorrhagic Fever, American ,NEUTRALIZING ANTIBODIES ,Ciencias Biológicas ,03 medical and health sciences ,Virology ,medicine ,ANTIBODY CROSS-REACTIVITY ,Humans ,Arenaviruses, New World ,Pharmacology ,Arenavirus ,Junin virus ,biology ,JUNÍN VIRUS ,biology.organism_classification ,Antibodies, Neutralizing ,030104 developmental biology ,Lassa virus ,ARENAVIRUS ,HEK293 Cells ,biology.protein ,Antibody ,NEUTRALIZATION ASSAY ,Virología ,CIENCIAS NATURALES Y EXACTAS - Abstract
Arenaviruses cause several viral hemorrhagic fevers endemic to Africa and South America. The respective causative agents are classified as biosafety level (BSL) 4 pathogens. Unlike for most other BSL4 agents, for the New World arenavirus Junín virus (JUNV) both a highly effective vaccination (Candid#1) and a post-exposure treatment, based on convalescent plasma transfer, are available. In particular, neutralizing antibodies (nAbs) represent a key protective determinant in JUNV infection, which is supported by the correlation between successful passive antibody therapy and the levels of nAbs administered. Unfortunately, comparable resources for the management of other closely related arenavirus infections are not available. Given the significant challenges inherent in studying BSL4 pathogens, our goal was to first assess the suitability of a JUNV transcription and replication-competent virus-like particle (trVLP) system for measuring virus neutralization under BSL1/2 conditions. Indeed, we could show that infection with JUNV trVLPs is glycoprotein (GP) dependent, that trVLP input has a direct correlation to reporter readout, and that these trVLPs can be neutralized by human serum with kinetics similar to those obtained using authentic virus. These properties make trVLPs suitable for use as a proxy for virus in neutralization assays. Using this platform we then evaluated the potential of JUNV nAbs to cross-neutralize entry mediated by GPs from other arenaviruses using JUNV (strain Romero)-based trVLPs bearing GPs either from other JUNV strains, other closely related New World arenaviruses (e.g. Tacaribe, Machupo, Sabiá), or the distantly related Lassa virus. While nAbs against the JUNV vaccine strain are also active against a range of other JUNV strains, they appear to have little or no capacity to neutralize other arenavirus species, suggesting that therapy with whole plasma directed against another species is unlikely to be successful and that the targeted development of cross-specific monoclonal antibody-based resources is likely needed. Such efforts will be supported by the availability of this BSL1/2 screening platform which provides a rapid and easy means to characterize the potency and reactivity of anti-arenavirus neutralizing antibodies against a range of arenavirus species. Fil: Leske, Anne. Friedrich Loeffler Institut; Alemania Fil: Waßmann, Irke. Friedrich Loeffler Institut; Alemania Fil: Schnepel, Kevin. Friedrich Loeffler Institut; Alemania Fil: Shifflett, Kyle. National Institutes of Health; Estados Unidos Fil: Holzerland, Julia. Friedrich Loeffler Institut; Alemania Fil: Bostedt, Linus. Friedrich Loeffler Institut; Alemania Fil: Bohn, Patrick. Friedrich Loeffler Institut; Alemania Fil: Mettenleiter, Thomas C.. Friedrich Loeffler Institut; Alemania Fil: Briggiler, Ana Maria. Dirección Nacional de Instituto de Investigación. Administración Nacional de Laboratorio e Instituto de Salud "Dr. C. G. Malbrán"; Argentina Fil: Brignone, Julia. Dirección Nacional de Instituto de Investigación. Administración Nacional de Laboratorio e Instituto de Salud "Dr. C. G. Malbrán"; Argentina Fil: Enria, Delia. Dirección Nacional de Instituto de Investigación. Administración Nacional de Laboratorio e Instituto de Salud "Dr. C. G. Malbrán"; Argentina Fil: Cordo, Sandra Myriam. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Ciudad Universitaria. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales; Argentina Fil: Hoenen, Thomas. Friedrich Loeffler Institut; Alemania. National Institutes of Health; Estados Unidos Fil: Groseth, Allison. Friedrich Loeffler Institut; Alemania. National Institutes of Health; Estados Unidos
- Published
- 2018
31. Thiazole Compounds as Antiviral Agents: An Update
- Author
-
Shiv Kumar Gupta, Sanjay Kumar, and Inder Pal Singh
- Subjects
Antifungal ,2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,medicine.drug_class ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Patent literature ,Microbial Sensitivity Tests ,Pharmacology ,01 natural sciences ,Antiviral Agents ,Virus ,03 medical and health sciences ,chemistry.chemical_compound ,Structure-Activity Relationship ,Drug Discovery ,medicine ,Thiazole ,030304 developmental biology ,0303 health sciences ,biology ,Dose-Response Relationship, Drug ,Molecular Structure ,business.industry ,biology.organism_classification ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Thiazoles ,chemistry ,Junin virus ,Viruses ,business - Abstract
Background: Thiazole is a good nucleus owing to its various pharmaceutical applications. Thiazole containing compounds (thiazoles) have shown various biological activities like antioxidant, analgesic, antibacterial, anticancer, antiallergic, antihypertensive, antiinflammatory, antimalarial, antifungal and antipsychotic. The scaffold is present in more than 18 FDA approved drugs and also in more than 70 experimental drugs. Only a few reviews are available in the literature despite its great medicinal importance. During the course of time, this scaffold has been studied extensively for its antiviral activities and provided compounds with activity in the nM range. However, no focused review is available on the compilation of antiviral activities shown by this scaffold. Objective: In the present review, we have made an effort to compile antiviral literature of thiazoles reported from the year 2011 to till date. Methods: We searched the SciFinder database (excluding patent literature) with keywords like “antiviral”, “anti-HIV” and “virus”. Further filters were applied for the year of publication and keywords thiazole, reviews etc. to find relevant literature reported on the antiviral activities of thiazoles. Results: Nearly, 50 research articles were selected to compile and review the antiviral literature of thiazoles reported from the year 2011 to till date. Compounds 8, 25, 40, 62, 72, 73, 91, 112, 113, 131, 137, 175, 198, 200, 201 and 213 were reported in the literature with potent antiviral activity against CVB, SARS, RSV, HCV, HRV, VZV, TMV, FMDV, DENV, YFV, influenza virus, Junin virus, HIV-1, HSV, VV and EBV, respectively. Conclusion: There is further scope for the synthesis and evaluation of novel thiazole compounds by taking the most active compounds as lead structures. In conclusion, this review provides an overview of antiviral activities of thiazole compounds reported from the year 2011 to till date.
- Published
- 2018
32. A Vaccine Platform against Arenaviruses Based on a Recombinant Hyperattenuated Mopeia Virus Expressing Heterologous Glycoproteins
- Author
-
Stéphane Barron, Sylvain Baize, Elsie Laban Yekwa, Hervé Raoul, Audrey Page, Stéphanie Reynard, Mathieu Mateo, Xavier Carnec, Audrey Vallve, Laura Barrot, Jimmy Hortion, Caroline Picard, François Ferron, Caroline Carbonnelle, Biologie des Infections Virales Émergentes - Biology of Emerging Viral Infections (UBIVE), Centre International de Recherche en Infectiologie - UMR (CIRI), École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Institut Pasteur [Paris], Architecture et fonction des macromolécules biologiques (AFMB), Centre National de la Recherche Scientifique (CNRS)-Aix Marseille Université (AMU)-Institut National de la Recherche Agronomique (INRA), Laboratoire P4 Jean Mérieux-Inserm [Lyon] (Unité de service 3), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Européen de Recherche en Virologie et Immunologie [Lyon] (Tour Inserm CERVI), This work was supported by the Fondation pour l'Innovation en Infectiologie (FINOVI) (Lyon, France), the Fondation pour la Recherche Médicale (FRM) (France), French National Research Agency grant ANR-11-BSV_019-02, and the Fondation Méditerranée Infection. This work was also supported by Labex Ecofect (grant ANR-11-LABX-0048, Lyon University) within the program Investissements d'Avenir (grant ANR-11-IDEX-0007, French National Research Agency)., ANR-11-LABX-0048,ECOFECT,Dynamiques eco-évolutives des maladies infectieuses(2011), ANR-11-IDEX-0007,Avenir L.S.E.,PROJET AVENIR LYON SAINT-ETIENNE(2011), Centre International de Recherche en Infectiologie (CIRI), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Institut Pasteur [Paris] (IP), Institut National de la Recherche Agronomique (INRA)-Aix Marseille Université (AMU)-Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS)-Institut Pasteur [Paris], ANR-11-IDEX-0007-02/11-LABX-0048,ECOFECT,Dynamiques eco-évolutives des maladies infectieuses(2011), and ANR-11-IDEX-0007-02/11-IDEX-0007,Avenir L.S.E.,Avenir L.S.E.(2011)
- Subjects
0301 basic medicine ,viruses ,MESH: Monkey Diseases/prevention & control ,viral hemorrhagic fevers ,MESH: Cricetinae ,medicine.disease_cause ,MESH: Lassa Fever/prevention & control ,MESH: Lassa Fever/virology ,Cricetinae ,Chlorocebus aethiops ,MESH: Animals ,Arenaviridae ,Lassa fever ,arenavirus ,innate immunity ,MESH: Arenaviridae/genetics ,biology ,MESH: Arenaviridae/immunology ,Viral Vaccine ,Monkey Diseases ,Vaccination ,[SDV.BBM.BC]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biomolecules [q-bio.BM] ,3. Good health ,Hemorrhagic Fevers ,MESH: Monkey Diseases/virology ,MESH: HEK293 Cells ,Interferon Type I ,MESH: Lassa virus/immunology ,MESH: Hemorrhagic Fevers, Viral/transmission ,MESH: Lassa Fever/immunology ,MESH: Interferon Type I/immunology ,MESH: Viral Vaccines/immunology ,Hemorrhagic Fevers, Viral ,live-vector vaccines ,Immunology ,MESH: Vero Cells ,MESH: Hemorrhagic Fevers, Viral/pathology ,Vaccines, Attenuated ,Microbiology ,Virus ,Cell Line ,03 medical and health sciences ,Virology ,Vaccines and Antiviral Agents ,medicine ,Animals ,Humans ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,[SDV.BBM.BC]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biochemistry [q-bio.BM] ,Lassa virus ,Vero Cells ,Arenavirus ,MESH: Humans ,MESH: Vaccines, Attenuated/immunology ,Viral Vaccines ,MESH: Monkey Diseases/immunology ,MESH: Vaccination ,biology.organism_classification ,medicine.disease ,MESH: Cercopithecus aethiops ,MESH: Hemorrhagic Fevers, Viral/immunology ,MESH: Exoribonucleases/metabolism ,MESH: Cell Line ,Macaca fascicularis ,HEK293 Cells ,030104 developmental biology ,MESH: Macaca fascicularis ,MESH: Hemorrhagic Fevers, Viral/virology ,Insect Science ,Junin virus ,Exoribonucleases - Abstract
Several Old World and New World arenaviruses are responsible for severe endemic and epidemic hemorrhagic fevers, whereas other members of the Arenaviridae family are nonpathogenic. To date, no approved vaccines, antivirals, or specific treatments are available, except for Junín virus. However, protection of nonhuman primates against Lassa fever virus (LASV) is possible through the inoculation of the closely related but nonpathogenic Mopeia virus (MOPV) before challenge with LASV. We reasoned that this virus, modified by using reverse genetics, would represent the basis for the generation of a vaccine platform against LASV and other pathogenic arenaviruses. After showing evidence of exoribonuclease (ExoN) activity in NP of MOPV, we found that this activity was essential for multiplication in antigen-presenting cells. The introduction of multiple mutations in the ExoN site of MOPV NP generated a hyperattenuated strain (MOPV ExoN6b ) that is (i) genetically stable over passages, (ii) has increased immunogenic properties compared to those of MOPV, and (iii) still promotes a strong type I interferon (IFN) response. MOPV ExoN6b was further modified to harbor the envelope glycoproteins of heterologous pathogenic arenaviruses, such as LASV or Lujo, Machupo, Guanarito, Chapare, or Sabia virus in order to broaden specific antigenicity while preserving the hyperattenuated characteristics of the parental strain. Our MOPV-based vaccine candidate for LASV, MOPEVAC LASV , was used in a one-shot immunization assay in nonhuman primates and fully protected them from a lethal challenge with LASV. Thus, our hyperattenuated strain of MOPV constitutes a promising new live-attenuated vaccine platform to immunize against several, if not all, pathogenic arenaviruses. IMPORTANCE Arenaviruses are emerging pathogens transmitted to humans by rodents and responsible for endemic and epidemic hemorrhagic fevers of global concern. Nonspecific symptoms associated with the onset of infection make these viruses difficult to distinguish from other endemic pathogens. Moreover, the unavailability of rapid diagnosis in the field delays the identification of the virus and early care for treatment and favors spreading. The vaccination of exposed populations would be of great help to decrease morbidity and human-to-human transmission. Using reverse genetics, we generated a vaccine platform for pathogenic arenaviruses based on a modified and hyperattenuated strain of the nonpathogenic Mopeia virus and showed that the Lassa virus candidate fully protected nonhuman primates from a lethal challenge. These results showed that a rationally designed recombinant MOPV-based vaccine is safe, immunogenic, and efficacious in nonhuman primates.
- Published
- 2018
- Full Text
- View/download PDF
33. Description and characterization of a novel live-attenuated tri-segmented Machupo virus in Guinea pigs
- Author
-
Christophe N. Peyrefitte, Amélie D. Zaza, and Cécile H. Herbreteau
- Subjects
Candid #1 ,0301 basic medicine ,Junín virus ,Arenaviridae ,Guinea Pigs ,Short Report ,Machupo virus ,Biology ,Vaccines, Attenuated ,Hemorrhagic Fever, American ,Virus ,Cell Line ,lcsh:Infectious and parasitic diseases ,Lethal Dose 50 ,03 medical and health sciences ,0302 clinical medicine ,Tri-segmented virus ,Mammarenaviruses ,Virology ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,lcsh:RC109-216 ,Vaccines, Virus-Like Particle ,Viremia ,030212 general & internal medicine ,Vero Cells ,Arenaviruses, New World ,Infectious virus ,Vaccine candidate development ,Junin virus ,Body Weight ,Vaccination ,Outbreak ,Viral Load ,medicine.disease ,biology.organism_classification ,Survival Rate ,Disease Models, Animal ,Hemorrhagic Fevers ,030104 developmental biology ,Infectious Diseases ,Bolivian hemorrhagic fever ,Therapeutic ,Reverse genetic approaches ,Mammarenavirus - Abstract
Background Machupo virus (MACV) is a member of the Mammarenavirus genus, Arenaviridae family and is the etiologic agent of Bolivian hemorrhagic fever, which causes small outbreaks or sporadic cases. Several other arenaviruses in South America Junín virus (JUNV) in Argentina, Guanarito in Venezuela, Sabiá in Brazil and Chapare in Bolivia, also are responsible for human hemorrhagic fevers. Among these arenaviruses, JUNV caused thousands of human cases until 1991, when the live attenuated Candid #1 vaccine, was used. Other than Candid #1 vaccine, few other therapeutic or prophylactic treatments exist. Therefore, new strategies for production of safe countermeasures with broad spectrum activity are needed. Findings We tested a tri-segmented MACV, a potential vaccine candidate with several mutations, (r3MACV). In cell culture, r3MACV showed a 2-log reduction in infectious virus particle production and the MACV inhibition of INF-1β was removed from the construct and produced by infected cells. Furthermore, in an animal experiment, r3MACV was able to protect 50% of guinea pigs from a simultaneous lethal JUNV challenge. Protected animals didn’t display clinical symptoms nor were virus particles found in peripheral blood (day 14) or in organs (day 28 post-inoculation). The r3MACV provided a higher protection than the Candid #1 vaccine. Conclusions The r3MACV provides a potential countermeasure against two South America arenaviruses responsible of human hemorrhagic fever. Electronic supplementary material The online version of this article (10.1186/s12985-018-1009-4) contains supplementary material, which is available to authorized users.
- Published
- 2018
- Full Text
- View/download PDF
34. Interactome analysis of the lymphocytic choriomeningitis virus nucleoprotein in infected cells reveals ATPase Na+/K+ transporting subunit Alpha 1 and prohibitin as host-cell factors involved in the life cycle of mammarenaviruses
- Author
-
Yíngyún Caì, Masaharu Iwasaki, John R. Yates, Petra Minder, Jens H. Kuhn, Juan Carlos de la Torre, and Bruce E. Torbett
- Subjects
0301 basic medicine ,Life Cycles ,Proteome ,viruses ,medicine.disease_cause ,Biochemistry ,Heat Shock Response ,Mice ,Cricetinae ,Chlorocebus aethiops ,Lymphocytic choriomeningitis virus ,Small interfering RNAs ,Protein Interaction Maps ,Prohibitin ,Biology (General) ,Arenaviridae ,Cells, Cultured ,Cellular Stress Responses ,Staining ,biology ,Cell Staining ,3. Good health ,Nucleic acids ,Cell Processes ,Host-Pathogen Interactions ,Sodium-Potassium-Exchanging ATPase ,Protein Binding ,Research Article ,Viral Entry ,QH301-705.5 ,Immunology ,Motor Proteins ,Lymphocytic Choriomeningitis ,Lymphocytic choriomeningitis ,Research and Analysis Methods ,Microbiology ,Virus ,03 medical and health sciences ,Viral life cycle ,Viral entry ,Molecular Motors ,Virology ,Prohibitins ,DNA-binding proteins ,medicine ,Genetics ,Animals ,Humans ,Non-coding RNA ,Molecular Biology ,Vero Cells ,Biology and life sciences ,Correction ,Proteins ,Cell Biology ,RC581-607 ,biology.organism_classification ,medicine.disease ,Nucleoprotein ,Gene regulation ,Repressor Proteins ,Cytoskeletal Proteins ,030104 developmental biology ,Lassa virus ,HEK293 Cells ,Nucleoproteins ,A549 Cells ,Specimen Preparation and Treatment ,Junin virus ,RNA ,Parasitology ,Gene expression ,Immunologic diseases. Allergy ,Viral Transmission and Infection ,Developmental Biology - Abstract
Several mammalian arenaviruses (mammarenaviruses) cause hemorrhagic fevers in humans and pose serious public health concerns in their endemic regions. Additionally, mounting evidence indicates that the worldwide-distributed, prototypic mammarenavirus, lymphocytic choriomeningitis virus (LCMV), is a neglected human pathogen of clinical significance. Concerns about human-pathogenic mammarenaviruses are exacerbated by of the lack of licensed vaccines, and current anti-mammarenavirus therapy is limited to off-label use of ribavirin that is only partially effective. Detailed understanding of virus/host-cell interactions may facilitate the development of novel anti-mammarenavirus strategies by targeting components of the host-cell machinery that are required for efficient virus multiplication. Here we document the generation of a recombinant LCMV encoding a nucleoprotein (NP) containing an affinity tag (rLCMV/Strep-NP) and its use to capture the NP-interactome in infected cells. Our proteomic approach combined with genetics and pharmacological validation assays identified ATPase Na+/K+ transporting subunit alpha 1 (ATP1A1) and prohibitin (PHB) as pro-viral factors. Cell-based assays revealed that ATP1A1 and PHB are involved in different steps of the virus life cycle. Accordingly, we observed a synergistic inhibitory effect on LCMV multiplication with a combination of ATP1A1 and PHB inhibitors. We show that ATP1A1 inhibitors suppress multiplication of Lassa virus and Candid#1, a live-attenuated vaccine strain of Junín virus, suggesting that the requirement of ATP1A1 in virus multiplication is conserved among genetically distantly related mammarenaviruses. Our findings suggest that clinically approved inhibitors of ATP1A1, like digoxin, could be repurposed to treat infections by mammarenaviruses pathogenic for humans., Author summary Viral hemorrhagic fever-causing mammalian viruses of the family Arenaviridae pose serious threats to humans in Africa and South America as the associated infections are highly lethal. The worldwide-distributed lymphocytic choriomeningitis virus (LCMV) is a relative of these dangerous viruses that can be worked with more safely in the laboratory. Although LCMV does not cause viral hemorrhagic fever, it can cause disease in humans. Currently, anti-arenavirus therapy options are very limited, not very effective, and associated with side effects. Development of new therapies has been hampered because knowledge on how arenaviruses interact with proteins of the host cells they infect is limited. Using a modified LCMV, we identified two host-cell proteins called ATPase Na+/K+ transporting subunit alpha 1 (ATP1A1) and prohibitin (PHB) as factors that promote arenavirus infection. Inhibitors of ATP1A1 (cardiac glycosides already used clinically for treatment of other diseases) suppressed multiplication in cell culture of Lassa virus and Junín virus, the two most significant viral hemorrhagic fever-causing mammarenaviruses. Therefore, our data suggest that these inhibitors could be used clinically to treat people infected with arenaviruses.
- Published
- 2018
35. Protocol for the Production of a Vaccine Against Argentinian Hemorrhagic Fever
- Author
-
Andrea S Maiza, Mauricio Mariani, Ana M. Ambrosio, Graciela S. Gamboa, Sebastián Fossa, and Alejando Javier Bottale
- Subjects
0301 basic medicine ,Attenuated vaccine ,biology ,business.industry ,Viral Vaccine ,Product processing ,030106 microbiology ,biology.organism_classification ,Virology ,Virus ,Regulatory authority ,03 medical and health sciences ,0302 clinical medicine ,Junin virus ,Medicine ,030212 general & internal medicine ,Argentinian hemorrhagic fever ,business ,Bank management - Abstract
Argentinian hemorrhagic Fever (AHF) is a febrile, acute disease caused by Junin virus (JUNV), a member of the Arenaviridae. Different approaches to obtain an effective antigen to prevent AHF using complete live or inactivated virus, as well as molecular constructs, have reached diverse development stages. This chapter refers to JUNV live attenuated vaccine strain Candid #1, currently used in Argentina to prevent AHF. A general standardized protocol used at Instituto Nacional de Enfermedades Virales Humanas (Pergamino, Pcia. Buenos Aires, Argentina) to manufacture the tissue culture derived Candid #1 vaccine is described. Intermediate stages like viral seeds and cell culture bank management, bulk vaccine manufacture, and finished product processing are also separately presented in terms of Production and Quality Control/Quality Assurance requirements, under the Adminitracion Nacional de Medicamentos, Alimentos y Tecnologia Medica (ANMAT), the Argentine national regulatory authority.
- Published
- 2017
- Full Text
- View/download PDF
36. Entry Studies of New World Arenaviruses
- Author
-
María Belén Forlenza Forlenza, Maria Guadalupe Martinez, Nélida A. Candurra, and Sandra M. Cordo
- Subjects
0301 basic medicine ,Permissiveness ,biology ,media_common.quotation_subject ,Host tropism ,Computational biology ,Endocytosis ,biology.organism_classification ,Virus ,03 medical and health sciences ,030104 developmental biology ,Viral life cycle ,Viral envelope ,Junin virus ,Internalization ,media_common - Abstract
Identification of cell moieties involved in viral binding and internalization is essential since their expression would render a cell susceptible. Further steps that allow the uncoating of the viral particle at the right subcellular localization have been intensively studied. These "entry" steps could determine cell permissiveness and often define tissue and host tropism. Therefore applying the right and, when possible, straightforward experimental approaches would shorten avenues to the complete knowledge of this first and key step of any viral life cycle. Mammarenaviruses are enveloped viruses that enter the host cell via receptor-mediated endocytosis. In this chapter we present a set of customized experimental approaches and tools that were used to describe the entry of Junin virus (JUNV), and other New World mammarenavirus members, into mammalian cells.
- Published
- 2017
- Full Text
- View/download PDF
37. Development and preliminary evaluation of a multiplexed amplification and next generation sequencing method for viral hemorrhagic fever diagnostics
- Author
-
Zeliha Kocak Tufan, Cristina Domingo, Andreas Nitsche, Annika Brinkmann, Koray Ergünay, and Aleksandar Radonić
- Subjects
0301 basic medicine ,RNA viruses ,Viral Diseases ,Molecular biology ,viruses ,medicine.disease_cause ,Pathology and Laboratory Medicine ,Sequencing techniques ,Bunyaviruses ,Medicine and Health Sciences ,Chikungunya ,DNA sequencing ,Viral Genomics ,Chikungunya Virus ,lcsh:Public aspects of medicine ,Yellow fever ,Microbial Genetics ,High-Throughput Nucleotide Sequencing ,Genomics ,Ebolavirus ,3. Good health ,Infectious Diseases ,Medical Microbiology ,Viral Pathogens ,Hemorrhagic Fever Virus, Crimean-Congo ,Viruses ,Yellow fever virus ,Pathogens ,Nucleic Acid Amplification Techniques ,Transcriptome Analysis ,Crimean Congo hemorrhagic fever virus ,Research Article ,Neglected Tropical Diseases ,Adult ,Next-Generation Sequencing ,lcsh:Arctic medicine. Tropical medicine ,Hemorrhagic Fevers, Viral ,lcsh:RC955-962 ,Alphaviruses ,030106 microbiology ,Microbial Genomics ,Biology ,Sensitivity and Specificity ,Microbiology ,Virus ,Viral hemorrhagic fever ,Togaviruses ,03 medical and health sciences ,Virology ,medicine ,Genetics ,Humans ,Microbial Pathogens ,Viral Hemorrhagic Fevers ,Ebola virus ,Junin virus ,Biology and life sciences ,Hemorrhagic Fever Viruses ,Public Health, Environmental and Occupational Health ,Organisms ,Computational Biology ,lcsh:RA1-1270 ,Sequence Analysis, DNA ,medicine.disease ,biology.organism_classification ,Rift Valley fever virus ,Genome Analysis ,Tropical Diseases ,Crimean-Congo hemorrhagic fever virus ,Research and analysis methods ,030104 developmental biology ,Molecular biology techniques ,DNA, Viral ,Nanopore sequencing - Abstract
Background We describe the development and evaluation of a novel method for targeted amplification and Next Generation Sequencing (NGS)-based identification of viral hemorrhagic fever (VHF) agents and assess the feasibility of this approach in diagnostics. Methodology An ultrahigh-multiplex panel was designed with primers to amplify all known variants of VHF-associated viruses and relevant controls. The performance of the panel was evaluated via serially quantified nucleic acids from Yellow fever virus, Rift Valley fever virus, Crimean-Congo hemorrhagic fever (CCHF) virus, Ebola virus, Junin virus and Chikungunya virus in a semiconductor-based sequencing platform. A comparison of direct NGS and targeted amplification-NGS was performed. The panel was further tested via a real-time nanopore sequencing-based platform, using clinical specimens from CCHF patients. Principal findings The multiplex primer panel comprises two pools of 285 and 256 primer pairs for the identification of 46 virus species causing hemorrhagic fevers, encompassing 6,130 genetic variants of the strains involved. In silico validation revealed that the panel detected over 97% of all known genetic variants of the targeted virus species. High levels of specificity and sensitivity were observed for the tested virus strains. Targeted amplification ensured viral read detection in specimens with the lowest virus concentration (1–10 genome equivalents) and enabled significant increases in specific reads over background for all viruses investigated. In clinical specimens, the panel enabled detection of the causative agent and its characterization within 10 minutes of sequencing, with sample-to-result time of less than 3.5 hours. Conclusions Virus enrichment via targeted amplification followed by NGS is an applicable strategy for the diagnosis of VHFs which can be adapted for high-throughput or nanopore sequencing platforms and employed for surveillance or outbreak monitoring., Author summary Viral hemorrhagic fever is a severe and potentially lethal disease, characterized by fever, malaise, vomiting, mucosal and gastrointestinal bleeding, and hypotension, in which multiple organ systems are affected. Due to modern transportation and global trade, outbreaks of viral hemorrhagic fevers have the potential to spread rapidly and affect a significant number of susceptible individuals. Thus, urgent and robust diagnostics with an identification of the causative virus is crucial. However, this is challenged by the number and diversity of the viruses associated with hemorrhagic fever. Several viruses classified in Arenaviridae, Filoviridae, and Flaviviridae families and Bunyavirales order may cause symptoms of febrile disease with hemorrhagic symptoms. We have developed and evaluated a novel method that can potentially identify all viruses and their genomic variants known to cause hemorrhagic fever in humans. The method relies on selected amplification of the target viral nucleic acids and subsequent high throughput sequencing technology for strain identification. Computer-based evaluations have revealed very high sensitivity and specificity, provided that the primer design is kept updated. Laboratory tests using several standard hemorrhagic virus strains and patient specimens have demonstrated excellent suitability of the assay in various sequencing platforms, which can achieve a definitive diagnosis in less than 3.5 hours.
- Published
- 2017
38. A Map of the Arenavirus Nucleoprotein-Host Protein Interactome Reveals that Junín Virus Selectively Impairs the Antiviral Activity of Double-Stranded RNA-Activated Protein Kinase (PKR)
- Author
-
Philip Eisenhauer, Benjamin R. King, Emily A. Bruce, Jason Botten, Joanne Russo, Bryan A. Ballif, Marion E. Weir, Christopher M. Ziegler, and Dylan C Hershkowitz
- Subjects
0301 basic medicine ,New World Arenavirus ,viruses ,Immunology ,Antiviral protein ,environment and public health ,Microbiology ,Mass Spectrometry ,Virus ,Cell Line ,eIF-2 Kinase ,03 medical and health sciences ,Viral life cycle ,Virology ,Protein Interaction Mapping ,Humans ,Immunoprecipitation ,Lymphocytic choriomeningitis virus ,Immune Evasion ,Junin virus ,Arenavirus ,biology ,virus diseases ,Nucleocapsid Proteins ,biology.organism_classification ,Protein kinase R ,Virus-Cell Interactions ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,Viral replication ,Insect Science ,Host-Pathogen Interactions - Abstract
Arenaviruses are enveloped negative-strand RNA viruses that cause significant human disease. These viruses encode only four proteins to accomplish the viral life cycle, so each arenavirus protein likely plays unappreciated accessory roles during infection. Here we used immunoprecipitation and mass spectrometry to identify human proteins that interact with the nucleoproteins (NPs) of the Old World arenavirus lymphocytic choriomeningitis virus (LCMV) and the New World arenavirus Junín virus (JUNV) strain Candid #1. Bioinformatic analysis of the identified protein partners of NP revealed that host translation appears to be a key biological process engaged during infection. In particular, NP associates with the double-stranded RNA (dsRNA)-activated protein kinase (PKR), a well-characterized antiviral protein that inhibits cap-dependent protein translation initiation via phosphorylation of eIF2α. JUNV infection leads to increased expression of PKR as well as its redistribution to viral replication and transcription factories. Further, phosphorylation of PKR, which is a prerequisite for its ability to phosphorylate eIF2α, is readily induced by JUNV. However, JUNV prevents this pool of activated PKR from phosphorylating eIF2α, even following exposure to the synthetic dsRNA poly(I·C), a potent PKR agonist. This blockade of PKR function is highly specific, as LCMV is unable to similarly inhibit eIF2α phosphorylation. JUNV's ability to antagonize the antiviral activity of PKR appears to be complete, as silencing of PKR expression has no impact on viral propagation. In summary, we provide a detailed map of the host machinery engaged by arenavirus NPs and identify an antiviral pathway that is subverted by JUNV. IMPORTANCE Arenaviruses are important human pathogens for which FDA-approved vaccines do not exist and effective antiviral therapeutics are needed. Design of antiviral treatment options and elucidation of the mechanistic basis of disease pathogenesis will depend on an increased basic understanding of these viruses and, in particular, their interactions with the host cell machinery. Identifying host proteins critical for the viral life cycle and/or pathogenesis represents a useful strategy to uncover new drug targets. This study reveals, for the first time, the extensive human protein interactome of arenavirus nucleoproteins and uncovers a potent antiviral host protein that is neutralized during Junín virus infection. In so doing, it shows further insight into the interplay between the virus and the host innate immune response and provides an important data set for the field.
- Published
- 2017
- Full Text
- View/download PDF
39. Highly Pathogenic New World Arenavirus Infection Activates the Pattern Recognition Receptor Protein Kinase R without Attenuating Virus Replication in Human Cells
- Author
-
Elizabeth J. Mateer, Slobodan Paessler, Cheng Huang, Takaaki Koma, and Olga A. Kolokoltsova
- Subjects
0301 basic medicine ,viruses ,Immunology ,Cellular Response to Infection ,Biology ,Virus Replication ,Microbiology ,Virus ,03 medical and health sciences ,eIF-2 Kinase ,Interferon ,Virology ,medicine ,Humans ,Phosphorylation ,Arenaviruses, New World ,Arenavirus ,Junin virus ,virus diseases ,RNA virus ,biology.organism_classification ,Protein kinase R ,Immunity, Innate ,RNA silencing ,030104 developmental biology ,Viral replication ,A549 Cells ,Insect Science ,Receptors, Pattern Recognition ,Host-Pathogen Interactions ,Interferons ,Arenaviruses, Old World ,medicine.drug ,Transcription Factors - Abstract
The arenavirus family consists of several highly pathogenic viruses, including the Old World (OW) arenavirus Lassa fever virus (LASV) and the New World (NW) Junin virus (JUNV) and Machupo virus (MACV). Host response to infection by these pathogenic arenaviruses is distinct in many aspects. JUNV and MACV infections readily induce an interferon (IFN) response in human cells, while LASV infection usually triggers an undetectable or weak IFN response. JUNV induces an IFN response through RIG-I, suggesting that the host non-self RNA sensor readily detects JUNV viral RNAs (vRNAs) during infection and activates IFN response. Double-stranded-RNA (dsRNA)-activated protein kinase R (PKR) is another host non-self RNA sensor classically known for its vRNA recognition activity. Here we report that infection with NW arenaviruses JUNV and MACV, but not OW LASV, activated PKR, concomitant with elevated phosphorylation of the translation initiation factor α subunit of eukaryotic initiation factor 2 (eIF2α). Host protein synthesis was substantially suppressed in MACV- and JUNV-infected cells but was only marginally reduced in LASV-infected cells. Despite the antiviral activity known for PKR against many other viruses, the replication of JUNV and MACV was not impaired but was slightly augmented in wild-type (wt) cells compared to that in PKR-deficient cells, suggesting that PKR or PKR activation did not negatively affect JUNV and MACV infection. Additionally, we found an enhanced IFN response in JUNV- or MACV-infected PKR-deficient cells, which was inversely correlated with virus replication. IMPORTANCE The detection of viral RNA by host non-self RNA sensors, including RIG-I and MDA5, is critical to the initiation of the innate immune response to RNA virus infection. Among pathogenic arenaviruses, the OW LASV usually does not elicit an interferon response. However, the NW arenaviruses JUNV and MACV readily trigger an IFN response in a RIG-I-dependent manner. Here, we demonstrate for the first time that pathogenic NW arenaviruses JUNV and MACV, but not the OW arenavirus LASV, activated the dsRNA-dependent PKR, another host non-self RNA sensor, during infection. Interestingly, the replication of JUNV and MACV was not restricted but was rather slightly augmented in the presence of PKR. Our data provide new evidence for a distinct interplay between host non-self RNA sensors and pathogenic arenaviruses, which also provides insights into the pathogenesis of arenaviruses and may facilitate the design of vaccines and treatments against arenavirus-caused diseases.
- Published
- 2017
40. Arenaviruses and Hemorrhagic Fevers: From Virus Discovery to Molecular Biology, Therapeutics, and Prevention in Latin America
- Author
-
Mario Enrique Lozano, Víctor Romanowski, Sandra Elizabeth Goñi, and Matías Luis Pidre
- Subjects
Hemorrhagic Fevers ,Intergenic region ,biology ,Zoonotic Infection ,viruses ,Arenaviridae ,Junin virus ,RNA ,biology.organism_classification ,Genome ,Virology ,Virus - Abstract
Arenaviruses are enveloped viruses that have a bi-segmented negative-stranded RNA genome. The genomic RNA segments, large (L) and small (S), use an ambisense coding strategy to encode two open reading frames in opposite orientation, separated by a noncoding intergenic region. Several arenaviruses are etiological agents of emerging diseases. At present, they are responsible for up to 500,000 zoonotic infections per year in endemic areas of Africa and South America and can lead to severe and lethal hemorrhagic fever as well as neurological symptoms. Arenaviridae represents the largest group of hemorrhagic fever (HF)-causing viruses: five of the South American arenaviruses (CHPV, GTOV, JUNV, MACV, and SABV) are associated with HF in humans.
- Published
- 2017
- Full Text
- View/download PDF
41. Endothelial Cell Permeability and Adherens Junction Disruption Induced by Junín Virus Infection
- Author
-
Thomas Albrecht, Clarence J. Peters, Terence E. Hill, Ashley M. Grant, and Heather M. Lander
- Subjects
Delta Catenin ,Disease reservoir ,Cell Membrane Permeability ,Endothelium ,Vascular permeability ,Biology ,Argentine hemorrhagic fever ,Hemorrhagic Fever, American ,Permeability ,Virus ,Adherens junction ,Antigens, CD ,Virology ,Human Umbilical Vein Endothelial Cells ,medicine ,Animals ,Humans ,Sigmodontinae ,Chemokine CCL2 ,Disease Reservoirs ,Junin virus ,Interleukin-6 ,Catenins ,Dextrans ,Adherens Junctions ,Articles ,Cadherins ,medicine.disease ,biology.organism_classification ,Endothelial stem cell ,Infectious Diseases ,medicine.anatomical_structure ,Parasitology ,Fluorescein-5-isothiocyanate - Abstract
Junín virus (JUNV) is endemic to the fertile Pampas of Argentina, maintained in nature by the rodent host Calomys musculinus, and the causative agent of Argentine hemorrhagic fever (AHF), which is characterized by vascular dysfunction and fluid distribution abnormalities. Clinical as well as experimental studies implicate involvement of the endothelium in the pathogenesis of AHF, although little is known of its role. JUNV has been shown to result in productive infection of endothelial cells (ECs) in vitro with no visible cytopathic effects. In this study, we show that direct JUNV infection of primary human ECs results in increased vascular permeability as measured by electric cell substrate impedance sensing and transwell permeability assays. We also show that EC adherens junctions are disrupted during virus infection, which may provide insight into the role of the endothelium in the pathogenesis of AHF and possibly, other viral hemorrhagic fevers.
- Published
- 2014
- Full Text
- View/download PDF
42. Inhibition of Arenavirus Infection by a Glycoprotein-Derived Peptide with a Novel Mechanism
- Author
-
Lilia I. Melnik, Hussain Badani, Jennifer S. Spence, Robert F. Garry, and William C. Wimley
- Subjects
New World Arenavirus ,viruses ,Immunology ,Microbial Sensitivity Tests ,Viral Plaque Assay ,medicine.disease_cause ,Antiviral Agents ,Microbiology ,Virus ,Cell Line ,Inhibitory Concentration 50 ,Viral envelope ,Virology ,medicine ,Animals ,Humans ,Pichinde virus ,Glycoproteins ,Arenavirus ,biology ,Cryoelectron Microscopy ,Virion ,Virus Internalization ,Viral membrane ,biology.organism_classification ,Fusion protein ,Virus-Cell Interactions ,Lassa virus ,Junin virus ,Insect Science - Abstract
The family Arenaviridae includes a number of viruses of public health importance, such as the category A hemorrhagic fever viruses Lassa virus, Junin virus, Machupo virus, Guanarito virus, and Sabia virus. Current chemotherapy for arenavirus infection is limited to the nucleoside analogue ribavirin, which is characterized by considerable toxicity and treatment failure. Using Pichinde virus as a model arenavirus, we attempted to design glycoprotein-derived fusion inhibitors similar to the FDA-approved anti-HIV peptide enfuvirtide. We have identified a GP2-derived peptide, AVP-p, with antiviral activity and no acute cytotoxicity. The 50% inhibitory dose (IC 50 ) for the peptide is 7 μM, with complete inhibition of viral plaque formation at approximately 20 μM, and its antiviral activity is largely sequence dependent. AVP-p demonstrates activity against viruses with the Old and New World arenavirus viral glycoprotein complex but not against enveloped viruses of other families. Unexpectedly, fusion assays reveal that the peptide induces virus-liposome fusion at neutral pH and that the process is strictly glycoprotein mediated. As observed in cryo-electron micrographs, AVP-p treatment causes morphological changes consistent with fusion protein activation in virions, including the disappearance of prefusion glycoprotein spikes and increased particle diameters, and fluorescence microscopy shows reduced binding by peptide-treated virus. Steady-state fluorescence anisotropy measurements suggest that glycoproteins are destabilized by peptide-induced alterations in viral membrane order. We conclude that untimely deployment of fusion machinery by the peptide could render virions less able to engage in on-pathway receptor binding or endosomal fusion. AVP-p may represent a potent, highly specific, novel therapeutic strategy for arenavirus infection. IMPORTANCE Because the only drug available to combat infection by Lassa virus, a highly pathogenic arenavirus, is toxic and prone to treatment failure, we identified a peptide, AVP-p, derived from the fusion glycoprotein of a nonpathogenic model arenavirus, which demonstrates antiviral activity and no acute cytotoxicity. AVP-p is unique among self-derived inhibitory peptides in that it shows broad, specific activity against pseudoviruses bearing Old and New World arenavirus glycoproteins but not against viruses from other families. Further, the peptide's mechanism of action is highly novel. Biochemical assays and cryo-electron microscopy indicate that AVP-p induces premature activation of viral fusion proteins through membrane perturbance. Peptide treatment, however, does not increase the infectivity of cell-bound virus. We hypothesize that prematurely activated virions are less fit for receptor binding and membrane fusion and that AVP-p may represent a viable therapeutic strategy for arenavirus infection.
- Published
- 2014
- Full Text
- View/download PDF
43. Comparative analysis of disease pathogenesis and molecular mechanisms of New World and Old World arenavirus infections
- Author
-
Yuying Liang, Hinh Ly, and Lisa McLay
- Subjects
Hemorrhagic Fevers, Viral ,Innate immune system ,Arenavirus ,viruses ,Review ,Disease ,Biology ,medicine.disease_cause ,medicine.disease ,biology.organism_classification ,Virology ,Virus ,Lassa virus ,Immune system ,Junin virus ,Immunology ,medicine ,Animals ,Arenaviridae Infections ,Humans ,Cytokine storm ,Arenaviruses, Old World ,Arenaviruses, New World - Abstract
Arenaviruses can cause fatal human haemorrhagic fever (HF) diseases for which vaccines and therapies are extremely limited. Both the New World (NW) and Old World (OW) groups of arenaviruses contain HF-causing pathogens. Although these two groups share many similarities, important differences with regard to pathogenicity and molecular mechanisms of virus infection exist. These closely related pathogens share many characteristics, including genome structure, viral assembly, natural host selection and the ability to interfere with innate immune signalling. However, members of the NW and OW viruses appear to use different receptors for cellular entry, as well as different mechanisms of virus internalization. General differences in disease signs and symptoms and pathological lesions in patients infected with either NW or OW arenaviruses are also noted and discussed herein. Whilst both the OW Lassa virus (LASV) and the NW Junin virus (JUNV) can cause disruption of the vascular endothelium, which is an important pathological feature of HF, the immune responses to these related pathogens seem to be quite distinct. Whereas LASV infection results in an overall generalized immune suppression, patients infected with JUNV seem to develop a cytokine storm. Additionally, the type of immune response required for recovery and clearance of the virus is different between NW and OW infections. These differences may be important to allow the viruses to evade host immune detection. Understanding these differences will aid the development of new vaccines and treatment strategies against deadly HF viral infections.
- Published
- 2014
- Full Text
- View/download PDF
44. The ReFRAME library as a comprehensive drug repurposing library to identify mammarenavirus inhibitors
- Author
-
Mitchell V. Hull, Emily I. Chen, Arnab Chatterjee, Jens H. Kuhn, Yíngyún Caì, Beatrice Cubitt, Juan Carlos de la Torre, and Yu-Jin Kim
- Subjects
0301 basic medicine ,Databases, Pharmaceutical ,medicine.drug_class ,030106 microbiology ,Drug Evaluation, Preclinical ,Druggability ,Apoptosis ,Virus Replication ,medicine.disease_cause ,Lymphocytic choriomeningitis ,Antiviral Agents ,Article ,Virus ,Electron Transport Complex III ,03 medical and health sciences ,Virology ,Chlorocebus aethiops ,medicine ,Animals ,Arenaviridae Infections ,Humans ,Lymphocytic choriomeningitis virus ,Arenaviridae ,Lassa virus ,Vero Cells ,Repurposing ,Pharmacology ,Junin virus ,Dose-Response Relationship, Drug ,biology ,business.industry ,Drug Repositioning ,medicine.disease ,biology.organism_classification ,Drug repositioning ,HEK293 Cells ,Pyrimidines ,030104 developmental biology ,Proto-Oncogene Proteins c-bcl-2 ,A549 Cells ,Purines ,Interferons ,Antiviral drug ,business - Abstract
Several mammarenaviruses, chiefly Lassa virus (LASV) in Western Africa and Junin virus (JUNV) in the Argentine Pampas, cause severe disease in humans and pose important public health problems in their endemic regions. Moreover, mounting evidence indicates that the worldwide-distributed mammarenavirus lymphocytic choriomeningitis virus (LCMV) is a neglected human pathogen of clinical significance. The lack of licensed mammarenavirus vaccines and partial efficacy of current anti-mammarenavirus therapy limited to an off-label use of the nucleoside analog ribavirin underscore an unmet need for novel therapeutics to combat human pathogenic mammarenavirus infections. This task can be facilitated by the implementation of “drug repurposing” strategies to reduce the time and resources required to advance identified antiviral drug candidates into the clinic. We screened a drug repurposing library of 11,968 compounds (Repurposing, Focused Rescue and Accelerated Medchem [ReFRAME]) and identified several potent inhibitors of LCMV multiplication that had also strong anti-viral activity against LASV and JUNV. Our findings indicate that enzymes of the rate-limiting steps of pyrimidine and purine biosynthesis, the pro-viral MCL1 apoptosis regulator, BCL2 family member protein and the mitochondrial electron transport complex III, play critical roles in the completion of the mammarenavirus life cycle, suggesting they represent potential druggable targets to counter human pathogenic mammarenavirus infections.
- Published
- 2019
- Full Text
- View/download PDF
45. TRIM2, a novel member of the antiviral family, limits New World arenavirus entry
- Author
-
Sarute, Nicolás, Ibrahim, N., Medegan Fagla, B., Lavanya, M., Cuevas, C., Stavrou, S., Otkiran-Clare, G., Tyynismaa, H., Henao-Mejia, J., Ross, S. R., Rajsbaum, R., STEMM - Stem Cells and Metabolism Research Program, Centre of Excellence in Stem Cell Metabolism, Department of Medical and Clinical Genetics, Henna Tyynismaa / Principal Investigator, Research Programme for Molecular Neurology, Research Programs Unit, University of Helsinki, Sarute Nicolás, Universidad de la República (Uruguay). Facultad de Ciencias. Instituto de Biología., Ibrahim N., Medegan Fagla B., Lavanya M., Cuevas C., Stavrou S., Otkiran-Clare G., Tyynismaa H., Henao-Mejia J., and Ross S.R.
- Subjects
Mutant ,New World arenaviruses ,Apoptosis ,Biochemistry ,Mice ,0302 clinical medicine ,Animal Cells ,Charcot-Marie-Tooth Disease ,BINDING ,Small interfering RNAs ,Biology (General) ,Connective Tissue Cells ,Mitogen-Activated Protein Kinase 1 ,TRANSFERRIN RECEPTOR 1 ,Mitogen-Activated Protein Kinase 3 ,I INTERFERON ,Brain ,Transfection ,Precipitation Techniques ,3. Good health ,Cell biology ,Nucleic acids ,RNA isolation ,Spectrophotometry ,Cytophotometry ,Cellular Types ,General Agricultural and Biological Sciences ,QH301-705.5 ,Tripartite motif (TRIM) ,Immune Cells ,Phagocytosis ,Immunology ,Primary Cell Culture ,Biomolecular isolation ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Genetics ,Humans ,Non-coding RNA ,Blood Cells ,Osteoblasts ,IDENTIFICATION ,PROTEOME-SCALE MAP ,Macrophages ,HEK 293 cells ,Proteins ,Fibroblasts ,Virus Internalization ,Tripartite motif family ,Mice, Inbred C57BL ,Molecular biology techniques ,Biological Tissue ,030104 developmental biology ,Junin virus ,MOTIF ,Gene expression ,030217 neurology & neurosurgery ,0301 basic medicine ,Molecular biology ,New World Arenavirus ,White Blood Cells ,Spectrum Analysis Techniques ,Neurofilament Proteins ,Chlorocebus aethiops ,Medicine and Health Sciences ,Post-Translational Modification ,Phosphorylation ,Receptors, Immunologic ,CD47 ,Arenaviruses, New World ,Mice, Knockout ,Fluorescence-Activated Cell Sorting ,General Neuroscience ,Nuclear Proteins ,Cell Processes ,Connective Tissue ,Host-Pathogen Interactions ,CELL ENTRY ,Anatomy ,Research Article ,Signal Transduction ,Virus infection ,Biology ,Virus ,UBIQUITIN LIGASE TRIM2 ,Cell Line, Tumor ,Immunoprecipitation ,Animals ,Vero Cells ,Biology and life sciences ,General Immunology and Microbiology ,Cell Biology ,biology.organism_classification ,Antigens, Differentiation ,Gene regulation ,Research and analysis methods ,HEK293 Cells ,Gene Expression Regulation ,RNA ,1182 Biochemistry, cell and molecular biology ,3111 Biomedicine ,JUNIN VIRUS - Abstract
Tripartite motif (TRIM) proteins belong to a large family with many roles in host biology, including restricting virus infection. Here, we found that TRIM2, which has been implicated in cases of Charcot–Marie–Tooth disease (CMTD) in humans, acts by blocking hemorrhagic fever New World arenavirus (NWA) entry into cells. We show that Trim2-knockout mice, as well as primary fibroblasts from a CMTD patient with mutations in TRIM2, are more highly infected by the NWAs Junín and Tacaribe virus than wild-type mice or cells are. Using mice with different Trim2 gene deletions and TRIM2 mutant constructs, we demonstrate that its antiviral activity is uniquely independent of the RING domain encoding ubiquitin ligase activity. Finally, we show that one member of the TRIM2 interactome, signal regulatory protein α (SIRPA), a known inhibitor of phagocytosis, also restricts NWA infection and conversely that TRIM2 limits phagocytosis of apoptotic cells. In addition to demonstrating a novel antiviral mechanism for TRIM proteins, these studies suggest that the NWA entry and phagocytosis pathways overlap., TRIM2, one of the proteins mutated in Charcot Marie Tooth Disease, blocks the entry of new world arenaviruses into cells, a novel mechanism for antiviral TRIM proteins, which generally restrict viruses at other stages of infection., Author summary New World arenaviruses (NWAs) are rodent-transmitted viruses that cause high mortality when they evolve the ability to infect humans. Although these clade B pathogenic viruses are known to bind to transferrin receptor 1 and other receptors on the cell surface, the steps leading to their entry into the cell are not well determined. We show that a host factor identified in a previous small interfering RNA (siRNA) screen, tripartite motif 2 (TRIM2), limits NWA endocytosis into cells. Moreover, we show that a member of the TRIM2 interactome, signal regulatory protein α (SIRPA), which is well-known for inhibiting phagocytosis by macrophages, interacts with TRIM2 and also blocks NWA infection. This finding suggests that there are common mechanisms that regulate virus endocytosis and phagocytosis.
- Published
- 2019
- Full Text
- View/download PDF
46. Utilization of human DC-SIGN and L-SIGN for entry and infection of host cells by the New World arenavirus, Junín virus
- Author
-
Gary R. Whittaker, Maria Guadalupe Martinez, Sandrine Belouzard, Sandra M. Cordo, Nélida A. Candurra, and Michele A. Bialecki
- Subjects
viruses ,Biophysics ,Host tropism ,Receptors, Cell Surface ,Biochemistry ,Article ,Hemorrhagic Fever, American ,Virus ,Ciencias Biológicas ,Mice ,03 medical and health sciences ,Virus entry ,Viral Envelope Proteins ,Viral envelope ,Antigens, CD ,Viral entry ,Chlorocebus aethiops ,Receptors, Transferrin ,Animals ,Humans ,Lectins, C-Type ,Antibody-dependent enhancement ,Vero Cells ,Molecular Biology ,Tropism ,030304 developmental biology ,0303 health sciences ,Junin virus ,Membrane Glycoproteins ,biology ,Virus receptor ,030302 biochemistry & molecular biology ,3T3 Cells ,Cell Biology ,Virus Internalization ,biology.organism_classification ,Virology ,3. Good health ,Junín arenavirus ,Host-Pathogen Interactions ,Cell Adhesion Molecules ,Lectin ,Virología ,CIENCIAS NATURALES Y EXACTAS - Abstract
The target cell tropism of enveloped viruses is regulated by interactions between viral proteins and cellular receptors determining susceptibility at a host cell, tissue or species level. However, a number of additional cell-surface moieties can also bind viral envelope glycoproteins and could act as capture receptors, serving as attachment factors to concentrate virus particles on the cell surface, or to disseminate the virus infection to target organs or susceptible cells within the host. Here, we used Junín virus (JUNV) or JUNV glycoprotein complex (GPC)-pseudotyped particles to study their ability to be internalized by the human C-type lectins hDC- or hL-SIGN. Our results provide evidence that hDC- and hL-SIGN can mediate the entry of Junín virus into cells, and may play an important role in virus infection and dissemination in the host. Fil: Martinez, María Guadalupe. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Ciudad Universitaria. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales; Argentina. Cornell University; Estados Unidos Fil: Bialecki, Michele A.. Cornell University; Estados Unidos Fil: Belouzard, Sandrine. Cornell University; Estados Unidos Fil: Cordo, Sandra Myriam. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Ciudad Universitaria. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales; Argentina Fil: Candurra, Nélida Alicia. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Ciudad Universitaria. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales; Argentina Fil: Whittaker, Gary R.. Cornell University; Estados Unidos
- Published
- 2013
- Full Text
- View/download PDF
47. 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
- Author
-
Takaaki Koma, Michael Patterson, Judith F. Aronson, Jeanon N. Smith, Milagros Miller, Aida G. Walker, Cheng Huang, and Slobodan Paessler
- Subjects
0301 basic medicine ,RNA viruses ,Physiology ,Cell Lines ,Pathology and Laboratory Medicine ,Antibodies, Viral ,Biochemistry ,Mice ,Viral Envelope Proteins ,Immune Physiology ,Cricetinae ,Medicine and Health Sciences ,Enzyme-Linked Immunoassays ,Neutralizing antibody ,Immune Response ,Receptors, Interferon ,Mice, Knockout ,Immune System Proteins ,Membrane Glycoproteins ,biology ,lcsh:Public aspects of medicine ,Immunogenicity ,Liver Diseases ,Animal Models ,Haplorhini ,Recombinant Proteins ,3. Good health ,Infectious Diseases ,Ectodomain ,Medical Microbiology ,Bolivian hemorrhagic fever ,Viral Pathogens ,Viruses ,Biological Cultures ,Pathogens ,Research Article ,lcsh:Arctic medicine. Tropical medicine ,lcsh:RC955-962 ,Immunology ,Mouse Models ,Gastroenterology and Hepatology ,Argentine hemorrhagic fever ,Research and Analysis Methods ,Vaccines, Attenuated ,Microbiology ,Virus ,Antibodies ,Hemorrhagic Fever, American ,03 medical and health sciences ,Model Organisms ,Signs and Symptoms ,Diagnostic Medicine ,Neutralization Tests ,medicine ,Animals ,Immunoassays ,Vero Cells ,Microbial Pathogens ,Inflammation ,Junin virus ,Public Health, Environmental and Occupational Health ,Organisms ,Biology and Life Sciences ,Proteins ,lcsh:RA1-1270 ,Viral Vaccines ,medicine.disease ,biology.organism_classification ,Virology ,Molecular biology ,Antibodies, Neutralizing ,Arenaviruses ,Fatty Liver ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,A549 Cells ,Vero cell ,biology.protein ,Immunologic Techniques ,Spleen - 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., Author Summary Machupo virus (MACV), a member of Arenaviridae family, causes Bolivian hemorrhagic fever (BHF) in humans. No approved vaccine or treatment are available to date despite the high case fatality rate of BHF. rMACV/Cd#1-GPC is fully attenuated and protects mice from lethal MACV challenge. Although one virulence determinant was found in the transmembrane domain of GPC (F438), other virulence determinants in GPC are very likely. Our new data indicated that the ectodomain of Cd#1 GPC is necessary, but not sufficient, for complete attenuation of the rMACV/Cd#1-GPC. This new finding may help generate highly attenuated MACVs for vaccine development and/or for drug screening purpose.
- Published
- 2016
48. Identification of alpha-dystroglycan as a receptor for lymphocytic choriomeningitis virus and Lassa fever virus
- Author
-
Kevin P. Campbell, Persephone Borrow, Stuart T. Nichol, Eugene V. Ravkov, Wei Cao, Hiroki Yamada, Michael B. A. Oldstone, Richard W. Compans, Michael D. Henry, and John H. Elder
- Subjects
Multidisciplinary ,Arenavirus ,biology ,viruses ,virus diseases ,hemic and immune systems ,chemical and pharmacologic phenomena ,Lymphocytic choriomeningitis ,medicine.disease ,biology.organism_classification ,medicine.disease_cause ,Virology ,Null allele ,Virus ,Microbiology ,nervous system diseases ,Lassa virus ,Viral replication ,Junin virus ,medicine ,biology.protein ,Pikachurin - Abstract
A peripheral membrane protein that is interactive with lymphocytic choriomeningitis virus (LCMV) was purified from cells permissive to infection. Tryptic peptides from this protein were determined to be alpha-dystroglycan (alpha-DG). Several strains of LCMV and other arenaviruses, including Lassa fever virus (LFV), Oliveros, and Mobala, bound to purified alpha-DG protein. Soluble alpha-DG blocked both LCMV and LFV infection. Cells bearing a null mutation of the gene encoding DG were resistant to LCMV infection, and reconstitution of DG expression in null mutant cells restored susceptibility to LCMV infection. Thus, alpha-DG is a cellular receptor for both LCMV and LFV.
- Published
- 2016
- Full Text
- View/download PDF
49. Differences in Glycoprotein Complex Receptor Binding Site Accessibility Prompt Poor Cross-Reactivity of Neutralizing Antibodies between Closely Related Arenaviruses
- Author
-
Elisabeth K. Phillips, Wendy Maury, Rachel B. Brouillette, and Natarajan Ayithan
- Subjects
0301 basic medicine ,medicine.drug_class ,030106 microbiology ,Immunology ,Cross Reactions ,Monoclonal antibody ,Antibodies, Viral ,Microbiology ,Virus ,Neutralization ,03 medical and health sciences ,Mice ,Antigen ,Species Specificity ,Glycoprotein complex ,Virology ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Binding site ,Antigens, Viral ,Vero Cells ,Arenavirus ,Binding Sites ,Junin virus ,biology ,biology.organism_classification ,Antibodies, Neutralizing ,Virus-Cell Interactions ,030104 developmental biology ,HEK293 Cells ,Insect Science ,Viral Fusion Proteins ,Protein Binding - Abstract
The glycoprotein complex (GPC) of arenaviruses, composed of stable signal peptide, GP1, and GP2, is the only antigen correlated with antibody-mediated neutralization. However, despite strong cross-reactivity of convalescent antisera between related arenavirus species, weak or no cross-neutralization occurs. Two closely related clade B viruses, Machupo virus (MACV) and Junín virus (JUNV), have nearly identical overall GPC architecture and share a host receptor, transferrin receptor 1 (TfR1). Given structural and functional similarities of the GP1 receptor binding site (RBS) of these viruses and the recent demonstration that the RBS is an important target for neutralizing antibodies, it is not clear how these viruses avoid cross-neutralization. To address this, MACV/JUNV chimeric GPCs were assessed for interaction with a group of α-JUNV GPC monoclonal antibodies (MAbs) and mouse antisera against JUNV or MACV GPC. All six MAbs targeted GP1, with those that neutralized JUNV GPC-pseudovirions competing with each other for RBS binding. However, these MAbs were unable to bind to a chimeric GPC composed of JUNV GP1 containing a small disulfide bonded loop (loop 10) unique to MACV GPC, suggesting that this loop may block MAbs interaction with the GP1 RBS. Consistent with this loop causing interference, mouse anti-JUNV GPC antisera that solely neutralized pseudovirions bearing autologous GP1 provided enhanced neutralization of MACV GPC when this loop was removed. Our studies provide evidence that loop 10, which is unique to MACV GP1, is an important impediment to binding of neutralizing antibodies and contributes to the poor cross-neutralization of α-JUNV antisera against MACV. IMPORTANCE Multiple New World arenaviruses can cause severe disease in humans, and some geographic overlap exists among these viruses. A vaccine that protects against a broad range of New World arenaviruses is desirable for purposes of simplicity, cost, and broad protection against multiple National Institute of Allergy and Infectious Disease-assigned category A priority pathogens. In this study, we sought to better understand how closely related arenaviruses elude cross-species neutralization by investigating the structural bases of antibody binding and avoidance. In our studies, we found that neutralizing antibodies against two New World arenaviruses, Machupo virus (MACV) and Junín virus (JUNV), bound to the envelope glycoprotein 1 (GP1) with JUNV monoclonal antibodies targeting the receptor binding site (RBS). We further show that altered structures surrounding the RBS pocket in MACV GP1 impede access of JUNV-elicited antibodies.
- Published
- 2016
50. Identification and Characterization of a Novel Broad-Spectrum Virus Entry Inhibitor
- Author
-
Kangmin He, Sarah H. Stubbs, Luke H. Chao, Raphael Gaudin, Frances Evesson, Yi-ying Chou, Margot Carocci, Susan R. Ross, Priscilla L. Yang, Sean P. J. Whelan, Christian D. Cuevas, David K. Cureton, Minghe Ma, Tom Kirchhausen, Virologie UMR1161 (VIRO), École nationale vétérinaire d'Alfort (ENVA)-Institut National de la Recherche Agronomique (INRA)-Agence nationale de sécurité sanitaire de l'alimentation, de l'environnement et du travail (ANSES), Boston Children's Hospital, Department of Cell Biology, Harvard Medical School, and Institut National de la Recherche Agronomique (INRA)-Agence nationale de sécurité sanitaire de l'alimentation, de l'environnement et du travail (ANSES)-École nationale vétérinaire d'Alfort (ENVA)
- Subjects
0301 basic medicine ,[SDV]Life Sciences [q-bio] ,Dengue virus ,Virus Replication ,medicine.disease_cause ,Gene Knockout Techniques ,Mice ,Drug Discovery ,cdc42 GTP-Binding Protein ,Cells, Cultured ,ComputingMilieux_MISCELLANEOUS ,biology ,Thiourea ,Viral Load ,Endocytosis ,3. Good health ,Protein Transport ,Ribonucleoproteins ,Vesicular stomatitis virus ,Benzamides ,Protein Binding ,medicine.drug ,Immunology ,Virus Attachment ,Endosomes ,Antiviral Agents ,Microbiology ,Hemorrhagic Fever, American ,Virus ,Cell Line ,Viral Proteins ,03 medical and health sciences ,Viral entry ,Virology ,Vaccines and Antiviral Agents ,medicine ,Animals ,Humans ,Junin virus ,Dose-Response Relationship, Drug ,030102 biochemistry & molecular biology ,Virus Internalization ,biology.organism_classification ,Actin cytoskeleton ,Actins ,Clathrin ,Entry inhibitor ,Disease Models, Animal ,030104 developmental biology ,Viral replication ,Insect Science ,Proteolysis - Abstract
Virus entry into cells is a multistep process that often requires the subversion of subcellular machineries. A more complete understanding of these steps is necessary to develop new antiviral strategies. While studying the potential role of the actin network and one of its master regulators, the small GTPase Cdc42, during Junin virus (JUNV) entry, we serendipitously uncovered the small molecule ZCL278, reported to inhibit Cdc42 function as an entry inhibitor for JUNV and for vesicular stomatitis virus, lymphocytic choriomeningitis virus, and dengue virus but not for the nonenveloped poliovirus. Although ZCL278 did not interfere with JUNV attachment to the cell surface or virus particle internalization into host cells, it prevented the release of JUNV ribonucleoprotein cores into the cytosol and decreased pH-mediated viral fusion with host membranes. We also identified SVG-A astroglial cell-derived cells to be highly permissive for JUNV infection and generated new cell lines expressing fluorescently tagged Rab5c or Rab7a or lacking Cdc42 using clustered regularly interspaced short palindromic repeat (CRISPR)-caspase 9 (Cas9) gene-editing strategies. Aided by these tools, we uncovered that perturbations in the actin cytoskeleton or Cdc42 activity minimally affect JUNV entry, suggesting that the inhibitory effect of ZCL278 is not mediated by ZCL278 interfering with the activity of Cdc42. Instead, ZCL278 appears to redistribute viral particles from endosomal to lysosomal compartments. ZCL278 also inhibited JUNV replication in a mouse model, and no toxicity was detected. Together, our data suggest the unexpected antiviral activity of ZCL278 and highlight its potential for use in the development of valuable new tools to study the intracellular trafficking of pathogens. IMPORTANCE The Junin virus is responsible for outbreaks of Argentine hemorrhagic fever in South America, where 5 million people are at risk. Limited options are currently available to treat infections by Junin virus or other viruses of the Arenaviridae , making the identification of additional tools, including small-molecule inhibitors, of great importance. How Junin virus enters cells is not yet fully understood. Here we describe new cell culture models in which the cells are susceptible to Junin virus infection and to which we applied CRISPR-Cas9 genome engineering strategies to help characterize early steps during virus entry. We also uncovered ZCL278 to be a new antiviral small molecule that potently inhibits the cellular entry of the Junin virus and other enveloped viruses. Moreover, we show that ZCL278 also functions in vivo , thereby preventing Junin virus replication in a mouse model, opening the possibility for the discovery of ZCL278 derivatives of therapeutic potential.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.