1
|
Urata S, Lee M, Tsuruta T, Igarashi R, Takeda K, Unno H. Mode of antiviral action of the galactose-specific lectin, AJLec, on the Junin virus propagation. Antiviral Res 2025; 240:106189. [PMID: 40381661 DOI: 10.1016/j.antiviral.2025.106189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 05/11/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025]
Abstract
Junin virus (JUNV), a member of Arenaviridae, is the causative agent of Argentine hemorrhagic fever (AHF). Available AHF treatments are limited; therefore, development of effective and safe treatments is required. Thus, in this study, novel lectins were examined for anti-JUNV activity. To evaluate JUNV propagation, a recombinant Junin virus vaccine strain (r3Candid #1/ZsGreen) containing the ZsGreen gene as a marker in the viral genome was used. The anti-JUNV effects of four types of marine organism-derived lectins collected in Japan, including the Nagasaki Prefecture, were examined. AJLec, which was extracted from the Sea Anemone Anthopleura japonica, reduced the number of infected cells and viral production. Infection and infection-surrogate assays revealed that incubation of AJLec with viruses and cells before infection, and maintaining it during infection, was required to exhibit full antiviral activity. Moreover, the anti-JUNV activity of AJLec was suppressed by the addition of lactose; hence, the anti-JUNV activity of AJLec was a result of its galactose recognition. This indicates the importance of galactose on the surface of the Junin virion and the cell membrane for entry into cells. Overall, these results provide new insights into the anti-JUNV activity of AJLec. Particularly, the potential of lectins as new antiviral agents that inhibit pathogenic arenavirus replication and propagation is promising.
Collapse
Affiliation(s)
- Shuzo Urata
- Department of Medical and Applied Virology, National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan; Department of Emerging Infectious Diseases, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.
| | - Meion Lee
- Department of Medical and Applied Virology, National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan; Deparment of Cell Regulation, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki, 852-8521, Japan
| | - Tomoko Tsuruta
- Department of Medical and Applied Virology, National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Reo Igarashi
- Department of Medical and Applied Virology, National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Kohsuke Takeda
- Deparment of Cell Regulation, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki, 852-8521, Japan
| | - Hideaki Unno
- Graduate School of Integrated Science and Technology, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki, 852-8521, Japan; Organization for Marine Science and Technology, Nagasaki University, Bunkyo-machi 1-14, Nagasaki, 852-8521, Japan
| |
Collapse
|
2
|
Hu X, Bai X, Tian F, Xing Y, Shi Y, Tong Y, Zhong J. A novel BSL-2 Lassa virus reverse genetics system modelling the complete viral life cycle. Emerg Microbes Infect 2024; 13:2356149. [PMID: 38747061 PMCID: PMC11168227 DOI: 10.1080/22221751.2024.2356149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 05/11/2024] [Indexed: 06/11/2024]
Abstract
Lassa virus (LASV), a risk-group 4 pathogen, must be handled in biosafety level-4 (BSL-4) conditions, thereby limiting its research and antiviral development. Here, we developed a novel LASV reverse genetics system which, to our knowledge, is the first to study the complete LASV life cycle under BSL-2 conditions. Viral particles can be produced efficiently when LASV minigenomic RNA harbouring minimal viral cis-elements and reporter genes is transfected into a helper cell line stably expressing viral NP, GP, Z and L proteins. The resulting defective virions, named LASVmg, can propagate only in the helper cell line, providing a BSL-2 model to study the complete LASV life cycle. Using this model, we found that a previously reported cellular receptor α-dystroglycan is dispensable for LASVmg infection. Furthermore, we showed that ribavirin can inhibit LASVmg infection by inducing viral mutations. This new BSL-2 system should facilitate studying the LASV life cycle and screening antivirals.
Collapse
Affiliation(s)
- Xiaoyou Hu
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Shanghai, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Xu Bai
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Shanghai, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Fangling Tian
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Shanghai, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Yifan Xing
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Shanghai, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Yi Shi
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Yimin Tong
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Shanghai, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Jin Zhong
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Shanghai, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, People's Republic of China
| |
Collapse
|
3
|
Mizuma K, Hashizume M, Urata S, Shindo K, Takashima A, Mizuta S, Iwasaki M. U-73122, a phospholipase C inhibitor, impairs lymphocytic choriomeningitis virus virion infectivity. J Gen Virol 2024; 105. [PMID: 39688895 DOI: 10.1099/jgv.0.002060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024] Open
Abstract
Lassa virus (LASV) is an Old World (OW) mammarenavirus that causes Lassa fever, a life-threatening acute febrile disease endemic in West Africa. Lymphocytic choriomeningitis virus (LCMV) is a worldwide-distributed, prototypic OW mammarenavirus of clinical significance that has been largely neglected as a human pathogen. No licensed OW mammarenavirus vaccines are available, and the current therapeutic option is limited to the off-label use of ribavirin, which offers only partial efficacy. This situation underscores the urgent need to develop novel antivirals against human pathogenic mammarenaviruses. Previously, we showed that afatinib, a pan-ErbB tyrosine kinase inhibitor, inhibited multiple steps of the life cycles of OW LASV and LCMV, as well as the New World Junín virus vaccine strain Candid#1. In the present study, we investigated the inhibitory effect of U-73122, a phospholipase C inhibitor that acts downstream of ErbB signalling, on LCMV multiplication. U-73122 inhibited WT recombinant (r) LCMV multiplication in cultured cells. Preincubation of cell-free LCMV virions with U-73122 resulted in impaired virion infectivity. U-73122 also inhibited the infection of rLCMVs expressing heterologous viral glycoproteins, including the vesicular stomatitis Indiana virus (VSIV) glycoprotein, whereas WT VSIV infection was not affected by U-73122 treatment. Our results show the novel bioactivity of U-73122 as an LCMV inhibitor and indicate the presence of a virion-associated molecule that is necessary for virion infectivity and can be exploited as a potential antiviral drug target against human pathogenic mammarenavirus infections.
Collapse
Affiliation(s)
- Keita Mizuma
- Laboratory of Emerging Viral Diseases, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Present address: Division of Risk Analysis and Management, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Mei Hashizume
- Laboratory of Emerging Viral Diseases, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Shuzo Urata
- National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Nagasaki, Japan
| | - Keiko Shindo
- Laboratory of Emerging Viral Diseases, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Ayako Takashima
- Laboratory of Emerging Viral Diseases, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Satoshi Mizuta
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Masaharu Iwasaki
- Laboratory of Emerging Viral Diseases, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- Center for Advanced Modalities and Drug Delivery System, Osaka University, Suita, Osaka, Japan
- RNA Frontier Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
4
|
Shaw AB, Tse HN, Byford O, Plahe G, Moon-Walker A, Hover SE, Saphire EO, Whelan SPJ, Mankouri J, Fontana J, Barr JN. Cellular endosomal potassium ion flux regulates arenavirus uncoating during virus entry. mBio 2024; 15:e0168423. [PMID: 38874413 PMCID: PMC11253613 DOI: 10.1128/mbio.01684-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 03/21/2024] [Indexed: 06/15/2024] Open
Abstract
Lymphocytic choriomeningitis virus (LCMV) is an enveloped and segmented negative-sense RNA virus classified within the Arenaviridae family of the Bunyavirales order. LCMV is associated with fatal disease in immunocompromised populations and, as the prototypical arenavirus member, acts as a model for the many highly pathogenic members of the Arenaviridae family, such as Junín, Lassa, and Lujo viruses, all of which are associated with devastating hemorrhagic fevers. To enter cells, the LCMV envelope fuses with late endosomal membranes, for which two established requirements are low pH and interaction between the LCMV glycoprotein (GP) spike and secondary receptor CD164. LCMV subsequently uncoats, where the RNA genome-associated nucleoprotein (NP) separates from the Z protein matrix layer, releasing the viral genome into the cytosol. To further examine LCMV endosome escape, we performed an siRNA screen which identified host cell potassium ion (K+) channels as important for LCMV infection, with pharmacological inhibition confirming K+ channel involvement during the LCMV entry phase completely abrogating productive infection. To better understand the K+-mediated block in infection, we tracked incoming virions along their entry pathway under physiological conditions, where uncoating was signified by separation of NP and Z proteins. In contrast, K+ channel blockade prevented uncoating, trapping virions within Rab7 and CD164-positive endosomes, identifying K+ as a third LCMV entry requirement. K+ did not increase GP-CD164 binding or alter GP-CD164-dependent fusion. Thus, we propose that K+ mediates uncoating by modulating NP-Z interactions within the virion interior. These results suggest K+ channels represent a potential anti-arenaviral target.IMPORTANCEArenaviruses can cause fatal human disease for which approved preventative or therapeutic options are not available. Here, using the prototypical LCMV, we identified K+ channels as critical for arenavirus infection, playing a vital role during the entry phase of the infection cycle. We showed that blocking K+ channel function resulted in entrapment of LCMV particles within late endosomal compartments, thus preventing productive replication. Our data suggest K+ is required for LCMV uncoating and genome release by modulating interactions between the viral nucleoprotein and the matrix protein layer inside the virus particle.
Collapse
Affiliation(s)
- Amelia B. Shaw
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Hiu Nam Tse
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Owen Byford
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Grace Plahe
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Alex Moon-Walker
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA
- Program in Virology, Harvard Medical School, Boston, Massachusetts, USA
| | - Samantha E. Hover
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Erica Ollmann Saphire
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Sean P. J. Whelan
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jamel Mankouri
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Juan Fontana
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - John N. Barr
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
5
|
Xu HC, Pandey P, Ward H, Gorzkiewicz M, Abromavičiūtė D, Tinz C, Müller L, Meyer C, Pandyra AA, Yavas A, Borkhardt A, Esposito I, Lang KS, Lang PA. High-Affinity-Mediated Viral Entry Triggers Innate Affinity Escape Resulting in Type I IFN Resistance and Impaired T Cell Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1457-1466. [PMID: 38497668 PMCID: PMC11016594 DOI: 10.4049/jimmunol.2300637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 02/23/2024] [Indexed: 03/19/2024]
Abstract
Increased receptor binding affinity may allow viruses to escape from Ab-mediated inhibition. However, how high-affinity receptor binding affects innate immune escape and T cell function is poorly understood. In this study, we used the lymphocytic choriomeningitis virus (LCMV) murine infection model system to create a mutated LCMV exhibiting higher affinity for the entry receptor α-dystroglycan (LCMV-GPH155Y). We show that high-affinity receptor binding results in increased viral entry, which is associated with type I IFN (IFN-I) resistance, whereas initial innate immune activation was not impaired during high-affinity virus infection in mice. Consequently, IFN-I resistance led to defective antiviral T cell immunity, reduced type II IFN, and prolonged viral replication in this murine model system. Taken together, we show that high-affinity receptor binding of viruses can trigger innate affinity escape including resistance to IFN-I resulting in prolonged viral replication.
Collapse
Affiliation(s)
- Haifeng C. Xu
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Piyush Pandey
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Harry Ward
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Michal Gorzkiewicz
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Džiuljeta Abromavičiūtė
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Constanze Tinz
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Lisa Müller
- Institute of Virology, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Caroline Meyer
- Institute of Virology, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Aleksandra A. Pandyra
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich Heine University, Düsseldorf, Germany
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research, Partner Site Bonn-Cologne, Bonn, Germany
| | - Aslihan Yavas
- Institute of Pathology, Medical Faculty, Heinrich Heine University and University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich Heine University, Düsseldorf, Germany
| | - Irene Esposito
- Institute of Pathology, Medical Faculty, Heinrich Heine University and University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Karl S. Lang
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Philipp A. Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
6
|
Johnson DM, Khakhum N, Wang M, Warner NL, Jokinen JD, Comer JE, Lukashevich IS. Pathogenic and Apathogenic Strains of Lymphocytic Choriomeningitis Virus Have Distinct Entry and Innate Immune Activation Pathways. Viruses 2024; 16:635. [PMID: 38675975 PMCID: PMC11053560 DOI: 10.3390/v16040635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/08/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Lymphocytic choriomeningitis virus (LCMV) and Lassa virus (LASV) share many genetic and biological features including subtle differences between pathogenic and apathogenic strains. Despite remarkable genetic similarity, the viscerotropic WE strain of LCMV causes a fatal LASV fever-like hepatitis in non-human primates (NHPs) while the mouse-adapted Armstrong (ARM) strain of LCMV is deeply attenuated in NHPs and can vaccinate against LCMV-WE challenge. Here, we demonstrate that internalization of WE is more sensitive to the depletion of membrane cholesterol than ARM infection while ARM infection is more reliant on endosomal acidification. LCMV-ARM induces robust NF-κB and interferon response factor (IRF) activation while LCMV-WE seems to avoid early innate sensing and failed to induce strong NF-κB and IRF responses in dual-reporter monocyte and epithelial cells. Toll-like receptor 2 (TLR-2) signaling appears to play a critical role in NF-κB activation and the silencing of TLR-2 shuts down IL-6 production in ARM but not in WE-infected cells. Pathogenic LCMV-WE infection is poorly recognized in early endosomes and failed to induce TLR-2/Mal-dependent pro-inflammatory cytokines. Following infection, Interleukin-1 receptor-associated kinase 1 (IRAK-1) expression is diminished in LCMV-ARM- but not LCMV-WE-infected cells, which indicates it is likely involved in the LCMV-ARM NF-κB activation. By confocal microscopy, ARM and WE strains have similar intracellular trafficking although LCMV-ARM infection appears to coincide with greater co-localization of early endosome marker EEA1 with TLR-2. Both strains co-localize with Rab-7, a late endosome marker, but the interaction with LCMV-WE seems to be more prolonged. These findings suggest that LCMV-ARM's intracellular trafficking pathway may facilitate interaction with innate immune sensors, which promotes the induction of effective innate and adaptive immune responses.
Collapse
Affiliation(s)
- Dylan M. Johnson
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Louisville, KY 94202, USA (I.S.L.)
- Department of Microbiology and Immunology, University of Louisville Health Sciences Center, Louisville, KY 94202, USA
- Galveston National Laboratory, Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77550, USA; (N.K.); (J.E.C.)
- Sandia National Laboratories, Department of Biotechnology & Bioengineering, Livermore, CA 94550, USA
| | - Nittaya Khakhum
- Galveston National Laboratory, Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77550, USA; (N.K.); (J.E.C.)
| | - Min Wang
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY 94202, USA;
| | - Nikole L. Warner
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Louisville, KY 94202, USA (I.S.L.)
- Department of Microbiology and Immunology, University of Louisville Health Sciences Center, Louisville, KY 94202, USA
| | - Jenny D. Jokinen
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Louisville, KY 94202, USA (I.S.L.)
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY 94202, USA;
| | - Jason E. Comer
- Galveston National Laboratory, Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77550, USA; (N.K.); (J.E.C.)
| | - Igor S. Lukashevich
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Louisville, KY 94202, USA (I.S.L.)
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY 94202, USA;
| |
Collapse
|
7
|
Iyer K, Yan Z, Ross SR. Entry inhibitors as arenavirus antivirals. Front Microbiol 2024; 15:1382953. [PMID: 38650890 PMCID: PMC11033450 DOI: 10.3389/fmicb.2024.1382953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
Arenaviruses belonging to the Arenaviridae family, genus mammarenavirus, are enveloped, single-stranded RNA viruses primarily found in rodent species, that cause severe hemorrhagic fever in humans. With high mortality rates and limited treatment options, the search for effective antivirals is imperative. Current treatments, notably ribavirin and other nucleoside inhibitors, are only partially effective and have significant side effects. The high lethality and lack of treatment, coupled with the absence of vaccines for all but Junín virus, has led to the classification of these viruses as Category A pathogens by the Centers for Disease Control (CDC). This review focuses on entry inhibitors as potential therapeutics against mammarenaviruses, which include both New World and Old World arenaviruses. Various entry inhibition strategies, including small molecule inhibitors and neutralizing antibodies, have been explored through high throughput screening, genome-wide studies, and drug repurposing. Notable progress has been made in identifying molecules that target receptor binding, internalization, or fusion steps. Despite promising preclinical results, the translation of entry inhibitors to approved human therapeutics has faced challenges. Many have only been tested in in vitro or animal models, and a number of candidates showed efficacy only against specific arenaviruses, limiting their broader applicability. The widespread existence of arenaviruses in various rodent species and their potential for their zoonotic transmission also underscores the need for rapid development and deployment of successful pan-arenavirus therapeutics. The diverse pool of candidate molecules in the pipeline provides hope for the eventual discovery of a broadly effective arenavirus antiviral.
Collapse
Affiliation(s)
| | | | - Susan R. Ross
- Department of Microbiology and Immunology, University of Illinois, College of Medicine, Chicago, IL, United States
| |
Collapse
|
8
|
Alatrash R, Herrera BB. The Adaptive Immune Response against Bunyavirales. Viruses 2024; 16:483. [PMID: 38543848 PMCID: PMC10974645 DOI: 10.3390/v16030483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 05/23/2024] Open
Abstract
The Bunyavirales order includes at least fourteen families with diverse but related viruses, which are transmitted to vertebrate hosts by arthropod or rodent vectors. These viruses are responsible for an increasing number of outbreaks worldwide and represent a threat to public health. Infection in humans can be asymptomatic, or it may present with a range of conditions from a mild, febrile illness to severe hemorrhagic syndromes and/or neurological complications. There is a need to develop safe and effective vaccines, a process requiring better understanding of the adaptive immune responses involved during infection. This review highlights the most recent findings regarding T cell and antibody responses to the five Bunyavirales families with known human pathogens (Peribunyaviridae, Phenuiviridae, Hantaviridae, Nairoviridae, and Arenaviridae). Future studies that define and characterize mechanistic correlates of protection against Bunyavirales infections or disease will help inform the development of effective vaccines.
Collapse
Affiliation(s)
- Reem Alatrash
- Rutgers Global Health Institute, Rutgers University, New Brunswick, NJ 08901, USA
- Department of Medicine, Division of Allergy, Immunology, and Infectious Diseases and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Bobby Brooke Herrera
- Rutgers Global Health Institute, Rutgers University, New Brunswick, NJ 08901, USA
- Department of Medicine, Division of Allergy, Immunology, and Infectious Diseases and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| |
Collapse
|
9
|
Byford O, Shaw AB, Tse HN, Todd EJAA, Álvarez-Rodríguez B, Hewson R, Fontana J, Barr JN. Lymphocytic choriomeningitis arenavirus requires cellular COPI and AP-4 complexes for efficient virion production. J Virol 2024; 98:e0200623. [PMID: 38334330 PMCID: PMC10949467 DOI: 10.1128/jvi.02006-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 01/16/2024] [Indexed: 02/10/2024] Open
Abstract
Lymphocytic choriomeningitis virus (LCMV) is a bisegmented negative-sense RNA virus classified within the Arenaviridae family of the Bunyavirales order. LCMV is associated with fatal disease in immunocompromized populations, and as the prototypical arenavirus, acts as a model for the many serious human pathogens within this group. Here, we examined the dependence of LCMV multiplication on cellular trafficking components using a recombinant LCMV expressing enhanced green fluorescent protein in conjunction with a curated siRNA library. The screen revealed a requirement for subunits of both the coat protein 1 (COPI) coatomer and adapter protein 4 (AP-4) complexes. By rescuing a recombinant LCMV harboring a FLAG-tagged glycoprotein (GP-1) envelope spike (rLCMV-GP1-FLAG), we showed infection resulted in marked co-localization of individual COPI and AP-4 components with both LCMV nucleoprotein (NP) and GP-1, consistent with their involvement in viral processes. To further investigate the role of both COPI and AP-4 complexes during LCMV infection, we utilized the ARF-I inhibitor brefeldin A (BFA) that prevents complex formation. Within a single 12-h cycle of virus multiplication, BFA pre-treatment caused no significant change in LCMV-specific RNA synthesis, alongside no significant change in LCMV NP expression, as measured by BFA time-of-addition experiments. In contrast, BFA addition resulted in a significant drop in released virus titers, approaching 50-fold over the same 12-h period, rising to over 600-fold over 24 h. Taken together, these findings suggest COPI and AP-4 complexes are important host cell factors required for the formation and release of infectious LCMV. IMPORTANCE Arenaviruses are rodent-borne, segmented, negative-sense RNA viruses, with several members responsible for fatal human disease, with the prototypic member lymphocytic choriomeningitis virus (LCMV) being under-recognised as a pathogen capable of inflicting neurological infections with fatal outcome. A detailed understanding of how arenaviruses subvert host cell processes to complete their multiplication cycle is incomplete. Here, using a combination of gene ablation and pharmacological inhibition techniques, we showed that host cellular COPI and AP-4 complexes, with native roles in cellular vesicular transport, were required for efficient LCMV growth. We further showed these complexes acted on late stages of the multiplication cycle, post-gene expression, with a significant impact on infectious virus egress. Collectively, our findings improve the understanding of arenaviruses host-pathogen interactions and reveal critical cellular trafficking pathways required during infection.
Collapse
Affiliation(s)
- Owen Byford
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Amelia B. Shaw
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Hiu Nam Tse
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Eleanor J. A. A. Todd
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Beatriz Álvarez-Rodríguez
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Roger Hewson
- Virology and Pathogenesis Group, National Infection Service, Public Health England, Porton Down, United Kingdom
| | - Juan Fontana
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - John N. Barr
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
10
|
Katz M, Diskin R. The underlying mechanisms of arenaviral entry through matriglycan. Front Mol Biosci 2024; 11:1371551. [PMID: 38516183 PMCID: PMC10955480 DOI: 10.3389/fmolb.2024.1371551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 02/15/2024] [Indexed: 03/23/2024] Open
Abstract
Matriglycan, a recently characterized linear polysaccharide, is composed of alternating xylose and glucuronic acid subunits bound to the ubiquitously expressed protein α-dystroglycan (α-DG). Pathogenic arenaviruses, like the Lassa virus (LASV), hijack this long linear polysaccharide to gain cellular entry. Until recently, it was unclear through what mechanisms LASV engages its matriglycan receptor to initiate infection. Additionally, how matriglycan is synthesized onto α-DG by the Golgi-resident glycosyltransferase LARGE1 remained enigmatic. Recent structural data for LARGE1 and for the LASV spike complex informs us about the synthesis of matriglycan as well as its usage as an entry receptor by arenaviruses. In this review, we discuss structural insights into the system of matriglycan generation and eventual recognition by pathogenic viruses. We also highlight the unique usage of matriglycan as a high-affinity host receptor compared with other polysaccharides that decorate cells.
Collapse
Affiliation(s)
| | - Ron Diskin
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
11
|
Gorzkiewicz M, Cramer J, Xu HC, Lang PA. The role of glycosylation patterns of viral glycoproteins and cell entry receptors in arenavirus infection. Biomed Pharmacother 2023; 166:115196. [PMID: 37586116 DOI: 10.1016/j.biopha.2023.115196] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 08/18/2023] Open
Abstract
Mammarenaviruses are enveloped RNA viruses that can be associated with rodent-transmitted diseases in humans. Their virions are composed of a nucleocapsid surrounded by a lipid bilayer with glycoprotein (GP) spikes interacting with receptors on target cells. Both the GP and receptors are highly glycosylated, with glycosylation patterns being crucial for virus binding and cell entry, viral tropism, immune responses, or therapy strategies. These effects have been previously described for several different viruses. In case of arenaviruses, they remain insufficiently understood. Thus, it is important to determine the mechanisms of glycosylation of viral proteins and receptors responsible for infection, in order to fully understand the biology of arenaviruses. In this article, we have summarized and critically evaluated the available literature data on the glycosylation of mammarenavirus-associated proteins to facilitate further research in this field.
Collapse
Affiliation(s)
- Michal Gorzkiewicz
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany; Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska St., 90-236 Lodz, Poland.
| | - Jonathan Cramer
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Haifeng C Xu
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Philipp A Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany.
| |
Collapse
|
12
|
Braadt L, Naumann M, Freuer D, Schmitz T, Linseisen J, Ertl M. Novel inflammatory biomarkers associated with stroke severity: results from a cross-sectional stroke cohort study. Neurol Res Pract 2023; 5:31. [PMID: 37468969 DOI: 10.1186/s42466-023-00259-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/20/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND Stroke is a leading cause of mortality and disability worldwide and its occurrence is expected to increase in the future. Blood biomarkers have proven their usefulness in identification and monitoring of the disease. Stroke severity is a major factor for estimation of prognosis and risk of recurrent events, but knowledge on respective blood biomarkers is still scarce. Stroke pathophysiology comprises a multitude of ischemia-induced inflammatory and immune mediated responses. Therefore, the assessment of an immune-related panel in correlation with stroke severity seems promising. METHODS In the present cross-sectional evaluation, a set of 92 blood biomarkers of a standardized immune panel were gathered (median 4.6 days after admission) and related to stroke severity measures, assessed at hospital admission of acute stroke patients. Multivariable logistic regression models were used to determine associations between biomarkers and modified Rankin Scale (mRS), linear regression models were used for associations with National Institute of Health Stroke Scale. RESULTS 415 patients (mean age 69 years; 41% female) were included for biomarker analysis. C-type lectin domain family 4 member G (CLEC4G; OR = 2.89, 95% CI [1.49; 5.59], padj = 0.026, Cytoskeleton-associated protein 4 (CKAP4; OR = 2.38, 95% CI [1.43; 3.98], padj = 0.019), and Interleukin-6 (IL-6) (IL6; OR = 1.97, 95% CI [1.49; 2.62], padj < 0.001) were positively associated with stroke severity measured by mRS, while Lymphocyte antigen 75 (LY75; OR = 0.37, 95% CI [0.19; 0.73], padj = 0.049) and Integrin alpha-11 (ITGA11 OR = 0.24, 95% CI [0.14, 0.40] padj < 0.001) were inversely associated. When investigating the relationships with the NIHSS, IL-6 (β = 0.23, 95% CI [0.12, 0.33] padj = 0.001) and ITGA11 (β = - 0.60, 95% CI [- 0.83, - 0.37] padj < 0.001) were significantly associated. CONCLUSIONS Higher relative concentrations of plasma CLEC4G, CKAP4, and IL-6 were associated with higher stroke severity, whereas LY75 and ITGA11 showed an inverse association. Future research might show a possible use as therapeutic targets and application in individual risk assessments.
Collapse
Affiliation(s)
- Lino Braadt
- Department of Neurology and Clinical Neurophysiology, University Hospital Augsburg, Augsburg, Germany.
| | - Markus Naumann
- Department of Neurology and Clinical Neurophysiology, University Hospital Augsburg, Augsburg, Germany
| | - Dennis Freuer
- Epidemiology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Timo Schmitz
- Epidemiology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Jakob Linseisen
- Epidemiology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Michael Ertl
- Department of Neurology and Clinical Neurophysiology, University Hospital Augsburg, Augsburg, Germany
| |
Collapse
|
13
|
Stachura P, Stencel O, Lu Z, Borkhardt A, Pandyra AA. Arenaviruses: Old viruses present new solutions for cancer therapy. Front Immunol 2023; 14:1110522. [PMID: 37033933 PMCID: PMC10079900 DOI: 10.3389/fimmu.2023.1110522] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
Viral-based cancer therapies have tremendous potential, especially in the context of treating poorly infiltrated cold tumors. However, in tumors with intact anti-viral interferon (IFN) pathways, while some oncolytic viruses induce strong innate and adaptive immune responses, they are neutralized before exerting their therapeutic effect. Arenaviruses, particularly the lymphocytic choriomeningitis virus (LCMV) is a noncytopathic virus with preferential cancer tropism and evolutionary mechanisms to escape the immune system for longer and to block early clearance. These escape mechanisms include inhibition of the MAVS dependent IFN pathway and spike protein antigen masking. Regarding its potential for cancer treatment, LCMV is therefore able to elicit long-term responses within the tumor microenvironment (TME), boost anti-tumor immune responses and polarize poorly infiltrating tumors towards a hot phenotype. Other arenaviruses including the attenuated Junin virus vaccine also have anti-tumor effects. Furthermore, the LCMV and Pichinde arenaviruses are currently being used to create vector-based vaccines with attenuated but replicating virus. This review focuses on highlighting the potential of arenaviruses as anti-cancer therapies. This includes providing a molecular understanding of its tropism as well as highlighting past and present preclinical and clinical applications of noncytophatic arenavirus therapies and their potential in bridging the gap in the treatment of cancers weakly responsive or unresponsive to oncolytic viruses. In summary, arenaviruses represent promising new therapies to broaden the arsenal of anti-tumor therapies for generating an immunogenic tumor microenvironment.
Collapse
Affiliation(s)
- Paweł Stachura
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Olivia Stencel
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Zhe Lu
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Aleksandra A. Pandyra
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- *Correspondence: Aleksandra A. Pandyra,
| |
Collapse
|
14
|
Pseudotyped Viruses for Mammarenavirus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1407:279-297. [PMID: 36920703 DOI: 10.1007/978-981-99-0113-5_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Mammarenaviruses are classified into New World arenaviruses (NW) and Old World arenaviruses (OW). The OW arenaviruses include the first discovered mammarenavirus-lymphocytic choriomeningitis virus (LCMV) and the highly lethal Lassa virus (LASV). Mammarenaviruses are transmitted to human by rodents, resulting in severe acute infections and hemorrhagic fever. Pseudotyped viruses have been widely used as a tool in the study of mammarenaviruses. HIV-1, SIV, FIV-based lentiviral vectors, VSV-based vectors, MLV-based vectors, and reverse genetic approaches have been applied in the construction of pseudotyped mammarenaviruses. Pseudotyped mammarenaviruses are commonly used in receptor research, neutralizing antibody detection, inhibitor screening, viral virulence studies, functional analysis of N-linked glycans, and studies of viral infection, endocytosis, and fusion mechanisms.
Collapse
|
15
|
Bertuzzi S, Peccati F, Serna S, Artschwager R, Notova S, Thépaut M, Jiménez-Osés G, Fieschi F, Reichardt NC, Jiménez-Barbero J, Ardá A. Immobilization of Biantennary N-Glycans Leads to Branch Specific Epitope Recognition by LSECtin. ACS CENTRAL SCIENCE 2022; 8:1415-1423. [PMID: 36313162 PMCID: PMC9615123 DOI: 10.1021/acscentsci.2c00719] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Indexed: 05/04/2023]
Abstract
The molecular recognition features of LSECtin toward asymmetric N-glycans have been scrutinized by NMR and compared to those occurring in glycan microarrays. A pair of positional glycan isomers (LDN3 and LDN6), a nonelongated GlcNAc4Man3 N-glycan (G0), and the minimum binding epitope (the GlcNAcβ1-2Man disaccharide) have been used to shed light on the preferred binding modes under both experimental conditions. Strikingly, both asymmetric LDN3 and LDN6 N-glycans are recognized by LSECtin with similar affinities in solution, in sharp contrast to the results obtained when those glycans are presented on microarrays, where only LDN6 was efficiently recognized by the lectin. Thus, different results can be obtained using different experimental approaches, pointing out the tremendous difficulty of translating in vitro results to the in vivo environment.
Collapse
Affiliation(s)
- Sara Bertuzzi
- Basque
Research & Technology Alliance (BRTA), Chemical Glycobiology Group, CIC bioGUNE, Bizkaia Technology Park, Building 800, 48160 Derio, Bizkaia, Spain
| | - Francesca Peccati
- Basque Research
& Technology Alliance (BRTA), Computational Chemistry Group, CIC bioGUNE, Bizkaia Technology Park, Building 800, 48160 Derio, Bizkaia, Spain
| | - Sonia Serna
- Glycotechnology
Group, Basque Research and Technology Alliance (BRTA), CIC biomaGUNE, Paseo Miramón 182, 20014 San Sebastian, Spain
| | - Raik Artschwager
- Glycotechnology
Group, Basque Research and Technology Alliance (BRTA), CIC biomaGUNE, Paseo Miramón 182, 20014 San Sebastian, Spain
- Memorial
Sloan Kettering Cancer Center, 417 East 68th Street, New
York, New York 10065, United States
| | - Simona Notova
- CNRS,
CEA, Institut de Biologie Structurale, University
of Grenoble Alpes, 38000 Grenoble, France
| | - Michel Thépaut
- CNRS,
CEA, Institut de Biologie Structurale, University
of Grenoble Alpes, 38000 Grenoble, France
| | - Gonzalo Jiménez-Osés
- Basque Research
& Technology Alliance (BRTA), Computational Chemistry Group, CIC bioGUNE, Bizkaia Technology Park, Building 800, 48160 Derio, Bizkaia, Spain
- Ikerbasque,
Basque Foundation for Science, Maria Diaz de Haro 3, 48013 Bilbao, Bizkaia, Spain
| | - Franck Fieschi
- CNRS,
CEA, Institut de Biologie Structurale, University
of Grenoble Alpes, 38000 Grenoble, France
- E-mail:
| | - Niels C. Reichardt
- Glycotechnology
Group, Basque Research and Technology Alliance (BRTA), CIC biomaGUNE, Paseo Miramón 182, 20014 San Sebastian, Spain
- CIBER-BBN, Paseo Miramón 182, 20009 San Sebastian, Spain
- E-mail:
| | - Jesús Jiménez-Barbero
- Basque
Research & Technology Alliance (BRTA), Chemical Glycobiology Group, CIC bioGUNE, Bizkaia Technology Park, Building 800, 48160 Derio, Bizkaia, Spain
- Ikerbasque,
Basque Foundation for Science, Maria Diaz de Haro 3, 48013 Bilbao, Bizkaia, Spain
- Department
of Organic Chemistry, II Faculty of Science
and Technology University of the Basque Country, EHU-UPV, 48940 Leioa, Spain
- Centro
de Investigación Biomédica En Red de Enfermedades Respiratorias, 28029 Madrid, Spain
- E-mail:
| | - Ana Ardá
- Basque
Research & Technology Alliance (BRTA), Chemical Glycobiology Group, CIC bioGUNE, Bizkaia Technology Park, Building 800, 48160 Derio, Bizkaia, Spain
- Ikerbasque,
Basque Foundation for Science, Maria Diaz de Haro 3, 48013 Bilbao, Bizkaia, Spain
- E-mail:
| |
Collapse
|
16
|
Genome-Wide Knockout Screen Identifies Human Sialomucin CD164 as an Essential Entry Factor for Lymphocytic Choriomeningitis Virus. mBio 2022; 13:e0020522. [PMID: 35502904 PMCID: PMC9239079 DOI: 10.1128/mbio.00205-22] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Lymphocytic choriomeningitis virus (LCMV) is a well-studied mammarenavirus that can be fatal in congenital infections. However, our understanding of LCMV and its interactions with human host factors remains incomplete. Here, host determinants affecting LCMV infection were investigated through a genome-wide CRISPR knockout screen in A549 cells, a human lung adenocarcinoma line. We identified and validated a variety of novel host factors that play a functional role in LCMV infection. Among these, knockout of the sialomucin CD164, a heavily glycosylated transmembrane protein, was found to ablate infection with multiple LCMV strains but not other hemorrhagic mammarenaviruses in several cell types. Further characterization revealed a dependency of LCMV entry on the cysteine-rich domain of CD164, including an N-linked glycosylation site at residue 104 in that region. Given the documented role of LCMV with respect to transplacental human infections, CD164 expression was investigated in human placental tissue and placental cell lines. CD164 was found to be highly expressed in the cytotrophoblast cells, an initial contact site for pathogens within the placenta, and LCMV infection in placental cells was effectively blocked using a monoclonal antibody specific to the cysteine-rich domain of CD164. Together, this study identifies novel factors associated with LCMV infection of human tissues and highlights the importance of CD164, a sialomucin that previously had not been associated with viral infection. IMPORTANCE Lymphocytic choriomeningitis virus (LCMV) is a human-pathogenic mammarenavirus that can be fatal in congenital infections. Although frequently used in the study of persistent infections in the field of immunology, aspects of this virus's life cycle remain incomplete. For example, while viral entry has been shown to depend on a cell adhesion molecule, DAG1, genetic knockout of this gene allows for residual viral infection, implying that additional receptors can mediate cell entry. The significance of our study is the identification of host factors important for successful infection, including the sialomucin CD164, which had not been previously associated with viral infection. We demonstrated that CD164 is essential for LCMV entry into human cells and can serve as a possible therapeutic target for treatment of congenital infection.
Collapse
|
17
|
Deng L, Liang P, Cui H. Pseudotyped lentiviral vectors: Ready for translation into targeted cancer gene therapy? Genes Dis 2022. [PMID: 37492721 PMCID: PMC10363566 DOI: 10.1016/j.gendis.2022.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Gene therapy holds great promise for curing cancer by editing the deleterious genes of tumor cells, but the lack of vector systems for efficient delivery of genetic material into specific tumor sites in vivo has limited its full therapeutic potential in cancer gene therapy. Over the past two decades, increasing studies have shown that lentiviral vectors (LVs) modified with different glycoproteins from a donating virus, a process referred to as pseudotyping, have altered tropism and display cell-type specificity in transduction, leading to selective tumor cell killing. This feature of LVs together with their ability to enable high efficient gene delivery in dividing and non-dividing mammalian cells in vivo make them to be attractive tools in future cancer gene therapy. This review is intended to summarize the status quo of some typical pseudotypings of LVs and their applications in basic anti-cancer studies across many malignancies. The opportunities of translating pseudotyped LVs into clinic use in cancer therapy have also been discussed.
Collapse
|
18
|
Pennington H, Lee J. Lassa virus glycoprotein complex review: insights into its unique fusion machinery. Biosci Rep 2022; 42:BSR20211930. [PMID: 35088070 PMCID: PMC8844875 DOI: 10.1042/bsr20211930] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 11/17/2022] Open
Abstract
Lassa virus (LASV), an arenavirus endemic to West Africa, causes Lassa fever-a lethal hemorrhagic fever. Entry of LASV into the host cell is mediated by the glycoprotein complex (GPC), which is the only protein located on the viral surface and comprises three subunits: glycoprotein 1 (GP1), glycoprotein 2 (GP2), and a stable signal peptide (SSP). The LASV GPC is a class one viral fusion protein, akin to those found in viruses such as human immunodeficiency virus (HIV), influenza, Ebola virus (EBOV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). These viruses are enveloped and utilize membrane fusion to deliver their genetic material to the host cell. Like other class one fusion proteins, LASV-mediated membrane fusion occurs through an orchestrated sequence of conformational changes in its GPC. The receptor-binding subunit, GP1, first engages with a host cell receptor then undergoes a unique receptor switch upon delivery to the late endosome. The acidic pH and change in receptor result in the dissociation of GP1, exposing the fusion subunit, GP2, such that fusion can occur. These events ultimately lead to the formation of a fusion pore so that the LASV genetic material is released into the host cell. Interestingly, the mature GPC retains its SSP as a third subunit-a feature that is unique to arenaviruses. Additionally, the fusion domain contains two separate fusion peptides, instead of a standard singular fusion peptide. Here, we give a comprehensive review of the LASV GPC components and their unusual features.
Collapse
Affiliation(s)
- Hallie N. Pennington
- Department of Chemistry and Biochemistry, College of Computer, Mathematics, and Natural Science, University of Maryland College Park, College Park, MD 20740, U.S.A
| | - Jinwoo Lee
- Department of Chemistry and Biochemistry, College of Computer, Mathematics, and Natural Science, University of Maryland College Park, College Park, MD 20740, U.S.A
| |
Collapse
|
19
|
Bhandari S, Larsen AK, McCourt P, Smedsrød B, Sørensen KK. The Scavenger Function of Liver Sinusoidal Endothelial Cells in Health and Disease. Front Physiol 2021; 12:757469. [PMID: 34707514 PMCID: PMC8542980 DOI: 10.3389/fphys.2021.757469] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
The aim of this review is to give an outline of the blood clearance function of the liver sinusoidal endothelial cells (LSECs) in health and disease. Lining the hundreds of millions of hepatic sinusoids in the human liver the LSECs are perfectly located to survey the constituents of the blood. These cells are equipped with high-affinity receptors and an intracellular vesicle transport apparatus, enabling a remarkably efficient machinery for removal of large molecules and nanoparticles from the blood, thus contributing importantly to maintain blood and tissue homeostasis. We describe here central aspects of LSEC signature receptors that enable the cells to recognize and internalize blood-borne waste macromolecules at great speed and high capacity. Notably, this blood clearance system is a silent process, in the sense that it usually neither requires or elicits cell activation or immune responses. Most of our knowledge about LSECs arises from studies in animals, of which mouse and rat make up the great majority, and some species differences relevant for extrapolating from animal models to human are discussed. In the last part of the review, we discuss comparative aspects of the LSEC scavenger functions and specialized scavenger endothelial cells (SECs) in other vascular beds and in different vertebrate classes. In conclusion, the activity of LSECs and other SECs prevent exposure of a great number of waste products to the immune system, and molecules with noxious biological activities are effectively “silenced” by the rapid clearance in LSECs. An undesired consequence of this avid scavenging system is unwanted uptake of nanomedicines and biologics in the cells. As the development of this new generation of therapeutics evolves, there will be a sharp increase in the need to understand the clearance function of LSECs in health and disease. There is still a significant knowledge gap in how the LSEC clearance function is affected in liver disease.
Collapse
Affiliation(s)
- Sabin Bhandari
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø (UiT) - The Arctic University of Norway, Tromsø, Norway
| | - Anett Kristin Larsen
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø (UiT) - The Arctic University of Norway, Tromsø, Norway
| | - Peter McCourt
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø (UiT) - The Arctic University of Norway, Tromsø, Norway
| | - Bård Smedsrød
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø (UiT) - The Arctic University of Norway, Tromsø, Norway
| | - Karen Kristine Sørensen
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø (UiT) - The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
20
|
Volland A, Lohmüller M, Heilmann E, Kimpel J, Herzog S, von Laer D. Heparan sulfate proteoglycans serve as alternative receptors for low affinity LCMV variants. PLoS Pathog 2021; 17:e1009996. [PMID: 34648606 PMCID: PMC8547738 DOI: 10.1371/journal.ppat.1009996] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/26/2021] [Accepted: 09/30/2021] [Indexed: 12/16/2022] Open
Abstract
Members of the Old World Arenaviruses primarily utilize α-dystroglycan (α-DAG1) as a cellular receptor for infection. Mutations within the glycoprotein (GP) of lymphocytic choriomeningitis virus (LCMV) reduce or abrogate the binding affinity to α-DAG1 and thus influence viral persistence, kinetics, and cell tropism. The observation that α-DAG1 deficient cells are still highly susceptible to low affinity variants, suggests the use of an alternative receptor(s). In this study, we used a genome-wide CRISPR Cas9 knockout screen in DAG1 deficient 293T cells to identify host factors involved in α-DAG1-independent LCMV infection. By challenging cells with vesicular stomatitis virus (VSV), pseudotyped with the GP of LCMV WE HPI (VSV-GP), we identified the heparan sulfate (HS) biosynthesis pathway as an important host factor for low affinity LCMV infection. These results were confirmed by a genetic approach targeting EXTL3, a key factor in the HS biosynthesis pathway, as well as by enzymatic and chemical methods. Interestingly, a single point mutation within GP1 (S153F or Y155H) of WE HPI is sufficient for the switch from DAG1 to HS binding. Furthermore, we established a simple and reliable virus-binding assay, using directly labelled VSV-GP by intramolecular fusion of VSV-P and mWasabi, demonstrating the importance of HS for virus attachment but not entry in Burkitt lymphoma cells after reconstitution of HS expression. Collectively, our study highlights the essential role of HS for low affinity LCMV infection in contrast to their high affinity counterparts. Residual LCMV infection in double knockouts indicate the use of (a) still unknown entry receptor(s).
Collapse
Affiliation(s)
- André Volland
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
- * E-mail: (AV); (DVL)
| | - Michael Lohmüller
- Division of Developmental Immunology, Medical University of Innsbruck, Innsbruck, Austria
| | - Emmanuel Heilmann
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Janine Kimpel
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sebastian Herzog
- Division of Developmental Immunology, Medical University of Innsbruck, Innsbruck, Austria
| | - Dorothee von Laer
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
- * E-mail: (AV); (DVL)
| |
Collapse
|
21
|
Hulswit RJG, Paesen GC, Bowden TA, Shi X. Recent Advances in Bunyavirus Glycoprotein Research: Precursor Processing, Receptor Binding and Structure. Viruses 2021; 13:353. [PMID: 33672327 PMCID: PMC7926653 DOI: 10.3390/v13020353] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 01/04/2023] Open
Abstract
The Bunyavirales order accommodates related viruses (bunyaviruses) with segmented, linear, single-stranded, negative- or ambi-sense RNA genomes. Their glycoproteins form capsomeric projections or spikes on the virion surface and play a crucial role in virus entry, assembly, morphogenesis. Bunyavirus glycoproteins are encoded by a single RNA segment as a polyprotein precursor that is co- and post-translationally cleaved by host cell enzymes to yield two mature glycoproteins, Gn and Gc (or GP1 and GP2 in arenaviruses). These glycoproteins undergo extensive N-linked glycosylation and despite their cleavage, remain associated to the virion to form an integral transmembrane glycoprotein complex. This review summarizes recent advances in our understanding of the molecular biology of bunyavirus glycoproteins, including their processing, structure, and known interactions with host factors that facilitate cell entry.
Collapse
Affiliation(s)
- Ruben J. G. Hulswit
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (R.J.G.H.); (G.C.P.)
| | - Guido C. Paesen
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (R.J.G.H.); (G.C.P.)
| | - Thomas A. Bowden
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (R.J.G.H.); (G.C.P.)
| | - Xiaohong Shi
- MRC-University of Glasgow Centre for Virus Research, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G61 1QH, UK
| |
Collapse
|
22
|
Holzerland J, Fénéant L, Banadyga L, Hölper JE, Knittler MR, Groseth A. BH3-only sensors Bad, Noxa and Puma are Key Regulators of Tacaribe virus-induced Apoptosis. PLoS Pathog 2020; 16:e1008948. [PMID: 33045019 PMCID: PMC7598930 DOI: 10.1371/journal.ppat.1008948] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 10/30/2020] [Accepted: 08/31/2020] [Indexed: 02/07/2023] Open
Abstract
Pathogenicity often differs dramatically among even closely related arenavirus species. For instance, Junín virus (JUNV), the causative agent of Argentine hemorrhagic fever (AHF), is closely related to Tacaribe virus (TCRV), which is normally avirulent in humans. While little is known about how host cell pathways are regulated in response to arenavirus infection, or how this contributes to virulence, these two viruses have been found to differ markedly in their ability to induce apoptosis. However, details of the mechanism(s) governing the apoptotic response to arenavirus infections are unknown. Here we confirm that TCRV-induced apoptosis is mitochondria-regulated, with associated canonical hallmarks of the intrinsic apoptotic pathway, and go on to identify the pro- and anti-apoptotic Bcl-2 factors responsible for regulating this process. In particular, levels of the pro-apoptotic BH3-only proteins Noxa and Puma, as well as their canonical transcription factor p53, were strongly increased. Interestingly, TCRV infection also led to the accumulation of the inactive phosphorylated form of another pro-apoptotic BH3-only protein, Bad (i.e. as phospho-Bad). Knockout of Noxa or Puma suppressed apoptosis in response to TCRV infection, whereas silencing of Bad increased apoptosis, confirming that these factors are key regulators of apoptosis induction in response to TCRV infection. Further, we found that while the highly pathogenic JUNV does not induce caspase activation, it still activated upstream pro-apoptotic factors, consistent with current models suggesting that JUNV evades apoptosis by interfering with caspase activation through a nucleoprotein-mediated decoy function. This new mechanistic insight into the role that individual BH3-only proteins and their regulation play in controlling apoptotic fate in arenavirus-infected cells provides an important experimental framework for future studies aimed at dissecting differences in the apoptotic responses between arenaviruses, their connection to other cell signaling events and ultimately the relationship of these processes to pathogenesis. Arenaviruses are important zoonotic pathogens that present a serious threat to human health. While some virus species cause severe disease, resulting in hemorrhagic fever and/or neurological symptoms, other closely related species exhibit little or no pathogenicity. The basis for these dramatically different outcomes is insufficiently understood, but investigations of host cell responses have suggested that apoptosis, i.e. non-inflammatory programmed cell death, is regulated differently between pathogenic and apathogenic arenaviruses. However, many questions remain regarding how these viruses interact with cell death pathways upon infection. Here we demonstrate that apoptosis induced by the avirulent Tacaribe virus (TCRV), proceeds via the mitochondria (i.e. the intrinsic apoptotic signaling pathway), and is regulated by a combination of factors that appear to balance activation (i.e. Noxa and Puma) and inactivation (i.e. Bad-P) of this cascade. During TCRV infection, the balance of these pro- and anti-apoptotic signals shifts the equilibrium late in the infection towards cell death. Importantly, we also found that the highly pathogenic Junín virus (JUNV), which does not trigger caspase activation or apoptotic cell death, nonetheless induces pro-apoptotic factors, thus supporting the existence of a specific mechanism by which this virus is able to evade apoptosis at late stages in this process.
Collapse
Affiliation(s)
- Julia Holzerland
- Junior Research Group Arenavirus Biology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Greifswald—Isle of Riems, Germany
| | - Lucie Fénéant
- Junior Research Group Arenavirus Biology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Greifswald—Isle of Riems, Germany
| | - Logan Banadyga
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Julia E. Hölper
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Greifswald—Isle of Riems, Germany
| | - Michael R. Knittler
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Greifswald—Isle of Riems, Germany
| | - Allison Groseth
- Junior Research Group Arenavirus Biology, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Greifswald—Isle of Riems, Germany
- * E-mail:
| |
Collapse
|
23
|
The Multifaceted Roles of TAM Receptors during Viral Infection. Virol Sin 2020; 36:1-12. [PMID: 32720213 DOI: 10.1007/s12250-020-00264-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022] Open
Abstract
Tyro3, Axl, and Mertk (TAM) receptors play multiple roles in a myriad of physiological and pathological processes, varying from promoting the phagocytic clearance of apoptotic cells, sustaining the immune and inflammatory homeostasis, maintaining the blood-brain barrier (BBB) integrity and central nervous system (CNS) homeostasis, to mediating cancer malignancy and chemoresistance. Growth arrest-specific protein 6 (Gas6) and protein S (Pros1) are the two ligands that activate TAM receptors. Recently, TAM receptors have been reported to mediate cell entry and infection of multitudinous enveloped viruses in a manner called apoptotic mimicry. Moreover, TAM receptors are revitalized during viral entry and infection, which sequesters innate immune and inflammatory responses, facilitating viral replication and immune evasion. However, accumulating evidence have now proposed that TAM receptors are not required for the infection of these viruses in vivo. In addition, TAM receptors protect mice against the CNS infection of neuroinvasive viruses and relieve the brain lesions during encephalitis. These protective effects are achieved through maintaining BBB integrity, attenuating proinflammatory cytokine production, and promoting neural cell survival. TAM receptors also regulate the programmed cell death modes of virus-infected cells, which have profound impacts on the pathogenesis and outcome of infection. Here, we systematically review the functionalities and underlying mechanisms of TAM receptors and propose the potential application of TAM agonists to prevent severe viral encephalitis.
Collapse
|
24
|
Ghosh Roy S. TAM receptors: A phosphatidylserine receptor family and its implications in viral infections. TAM RECEPTORS IN HEALTH AND DISEASE 2020; 357:81-122. [DOI: 10.1016/bs.ircmb.2020.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
25
|
Hallam SJ, Koma T, Maruyama J, Paessler S. Review of Mammarenavirus Biology and Replication. Front Microbiol 2018; 9:1751. [PMID: 30123198 PMCID: PMC6085440 DOI: 10.3389/fmicb.2018.01751] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/13/2018] [Indexed: 12/31/2022] Open
Abstract
The family Arenaviridae is divided into three genera: Mammarenavirus, Reptarenavirus, and Hartmanivirus. The Mammarenaviruses contain viruses responsible for causing human hemorrhagic fever diseases including New World viruses Junin, Machupo, Guanarito, Sabia, and Chapare virus and Old World viruses Lassa, and Lujo virus. These two groups of arenaviruses share the same genome organization composed of two ambisense RNA segments. These segments contain four open reading frames that encode for four proteins: the nucleoprotein, glycoprotein precursor, L protein, and Z. Despite their genome similarities, these groups exhibit marked differences in their replication life cycles. This includes differences in attachment, entry, and immune evasion. By understanding the intricacy of replication in each of these viral species we can work to develop counter measures against human diseases. This includes the development of vaccines and antivirals for these emerging viral threats. Currently only the vaccine against Junin virus, Candid#1, is in use as well as Ribavirin for treatment of Lassa Fever. In addition, small molecule inhibitors can be developed to target various aspects of the virus life cycle. In these ways an understanding of the arenavirus replication cycle can be used to alleviate the mortality and morbidity of these infections worldwide.
Collapse
Affiliation(s)
- Steven J. Hallam
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Takaaki Koma
- Department of Microbiology, Tokushima University Graduate School of Medical Science, Tokushima, Japan
| | - Junki Maruyama
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| |
Collapse
|
26
|
Böttcher-Friebertshäuser E, Garten W, Klenk HD. Cleavage of the Glycoprotein of Arenaviruses. ACTIVATION OF VIRUSES BY HOST PROTEASES 2018. [PMCID: PMC7121819 DOI: 10.1007/978-3-319-75474-1_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The arenaviruses are a large family of emerging negative-stranded RNA viruses that include several severe human pathogens causing hemorrhagic fevers with high mortality. During the arenavirus life cycle, processing of the viral envelope glycoprotein precursor (GPC) by the cellular subtilisin kexin isozyme-1 (SKI-1)/site-1 protease (S1P) is crucial for productive infection. The ability of newly emerging arenaviruses to hijack human SKI-1/S1P is a key factor for zoonotic transmission and human disease potential. Apart from being an essential host factor for arenavirus infection, SKI-1/S1P is involved in the regulation of important physiological processes and linked to major human diseases. This chapter provides an overview of the mechanisms of arenavirus GPC processing by SKI-1/S1P including recent findings. We will highlight to what extent the molecular mechanisms of SKI-1/S1P cleavage of viral GPC differ from processing of SKI-1/S1P’s cellular substrates and discuss the implications for virus-host interaction and coevolution. Moreover, we will show how the use of the viral GPC as a “molecular probe” uncovered novel and unusual aspects of SKI-1/S1P biosynthesis and maturation. The crucial role of SKI-1/S1P in arenavirus infection and other major human diseases combined with its nature as an enzyme makes SKI-1/S1P further an attractive target for therapeutic intervention. In the last part, we will therefore cover past and present efforts to identify specific SKI-1/S1P inhibitors.
Collapse
Affiliation(s)
| | - Wolfgang Garten
- Institut für Virologie, Philipps Universität, Marburg, Germany
| | | |
Collapse
|
27
|
Abstract
Viral entry represents the first step of every viral infection and is a determinant for the host range and disease potential of a virus. Here, we review the latest developments on cell entry of the highly pathogenic Old World arenavirus Lassa virus, providing novel insights into the complex virus-host cell interaction of this important human pathogen. We will cover new discoveries on the molecular mechanisms of receptor recognition, endocytosis, and the use of late endosomal entry factors.
Collapse
|
28
|
Wang W, Zhou Z, Zhang L, Wang S, Xiao G. Structure-function relationship of the mammarenavirus envelope glycoprotein. Virol Sin 2016; 31:380-394. [PMID: 27562602 DOI: 10.1007/s12250-016-3815-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/27/2016] [Indexed: 12/29/2022] Open
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.
Collapse
Affiliation(s)
- Wei Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Zheng Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Leike Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Shaobo Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Gengfu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| |
Collapse
|
29
|
Abstract
Viral apoptotic mimicry, defined by the exposure of phosphatidylserine on the pathogen surface, is emerging as a common theme used by enveloped viruses to promote infection. In this Progress article, Amara and Mercer discuss how viruses acquire phosphatidylserine and how this mimicry might facilitate cell entry and evasion of the immune response. As opportunistic pathogens, viruses have evolved many elegant strategies to manipulate host cells for infectious entry and replication. Viral apoptotic mimicry, defined by the exposure of phosphatidylserine — a marker for apoptosis — on the pathogen surface, is emerging as a common theme used by enveloped viruses to promote infection. Focusing on the four best described examples (vaccinia virus, dengue virus, Ebola virus and pseudotyped lentivirus), we summarize our current understanding of apoptotic mimicry as a mechanism for virus entry, binding and immune evasion. We also describe recent examples of non-enveloped viruses that use this mimicry strategy, and discuss future directions and how viral apoptotic mimicry could be targeted therapeutically.
Collapse
Affiliation(s)
- Ali Amara
- Institut National de la Santé et de la Recherche Médicale U944 and Centre National de la Recherche Scientifique UMR 7212, Laboratoire de Pathologie et Virologie Moléculaire, Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, 75010 Paris, France
| | - Jason Mercer
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
30
|
Beier JI, Jokinen JD, Holz GE, Whang PS, Martin AM, Warner NL, Arteel GE, Lukashevich IS. Novel mechanism of arenavirus-induced liver pathology. PLoS One 2015; 10:e0122839. [PMID: 25822203 PMCID: PMC4378851 DOI: 10.1371/journal.pone.0122839] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 02/20/2015] [Indexed: 12/15/2022] Open
Abstract
Viral hemorrhagic fevers (VHFs) encompass a group of diseases with cardinal symptoms of fever, hemorrhage, and shock. The liver is a critical mediator of VHF disease pathogenesis and high levels of ALT/AST transaminases in plasma correlate with poor prognosis. In fact, Lassa Fever (LF), the most prevalent VHF in Africa, was initially clinically described as hepatitis. Previous studies in non-human primate (NHP) models also correlated LF pathogenesis with a robust proliferative response in the liver. The purpose of the current study was to gain insight into the mechanism of liver injury and to determine the potential role of proliferation in LF pathogenesis. C57Bl/6J mice were infected with either the pathogenic (for NHPs) strain of lymphocytic choriomeningitis virus (LCMV, the prototypic arenavirus), LCMV-WE, or with the non-pathogenic strain, LCMV-ARM. As expected, LCMV-WE, but not ARM, caused a hepatitis-like infection. LCMV-WE also induced a robust increase in the number of actively cycling hepatocytes. Despite this increase in proliferation, there was no significant difference in liver size between LCMV-WE and LCMV-ARM, suggesting that cell cycle was incomplete. Indeed, cells appeared arrested in the G1 phase and LCMV-WE infection increased the number of hepatocytes that were simultaneously stained for proliferation and apoptosis. LCMV-WE infection also induced expression of a non-conventional virus receptor, AXL-1, from the TAM (TYRO3/AXL/MERTK) family of receptor tyrosine kinases and this expression correlated with proliferation. Taken together, these results shed new light on the mechanism of liver involvement in VHF pathogenesis. Specifically, it is hypothesized that the induction of hepatocyte proliferation contributes to expansion of the infection to parenchymal cells. Elevated levels of plasma transaminases are likely explained, at least in part, by abortive cell cycle arrest induced by the infection. These results may lead to the development of new therapies to prevent VHF progression.
Collapse
Affiliation(s)
- Juliane I. Beier
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, Kentucky, United States of America
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Louisville, Kentucky, United States of America
| | - Jenny D. Jokinen
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, Kentucky, United States of America
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Louisville, Kentucky, United States of America
| | - Gretchen E. Holz
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, Kentucky, United States of America
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Louisville, Kentucky, United States of America
| | - Patrick S. Whang
- Department of Microbiology and Immunology, University of Louisville Health Sciences Center, Louisville, Kentucky, United States of America
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Louisville, Kentucky, United States of America
| | - Amah M. Martin
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, Kentucky, United States of America
| | - Nikole L. Warner
- Department of Microbiology and Immunology, University of Louisville Health Sciences Center, Louisville, Kentucky, United States of America
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Louisville, Kentucky, United States of America
| | - Gavin E. Arteel
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, Kentucky, United States of America
| | - Igor S. Lukashevich
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, Kentucky, United States of America
- Department of Microbiology and Immunology, University of Louisville Health Sciences Center, Louisville, Kentucky, United States of America
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, Louisville, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
31
|
Abstract
UNLABELLED Several arenaviruses are known to cause viral hemorrhagic fever (VHF) in sub-Saharan Africa and South America, where VHF is a major public health and medical concern. The biosafety level 4 categorization of these arenaviruses restricts their use and has impeded biological studies, including therapeutic drug and/or vaccine development. Due to difficulties associated with handling live viruses, pseudotype viruses, which transiently bear arenavirus envelope proteins based on vesicular stomatitis virus (VSV) or retrovirus, have been developed as surrogate virus systems. Here, we report the development of a pseudotype VSV bearing each envelope protein of various species of arenaviruses (AREpv), including the newly identified Lujo virus (LUJV) and Chapare virus. Pseudotype arenaviruses generated in 293T cells exhibited high infectivity in various mammalian cell lines. The infections by New World and Old World AREpv were dependent on their receptors (human transferrin receptor 1 [hTfR1] and α-dystroglycan [αDG], respectively). However, infection by pseudotype VSV bearing the LUJV envelope protein (LUJpv) occurred independently of hTfR1 and αDG, indicating that LUJpv utilizes an unidentified receptor. The pH-dependent endocytosis of AREpv was confirmed by the use of lysosomotropic agents. The fusion of cells expressing these envelope proteins, except for those expressing the LUJV envelope protein, was induced by transient treatment at low pH values. LUJpv infectivity was inhibited by U18666A, a cholesterol transport inhibitor. Furthermore, the infectivity of LUJpv was significantly decreased in the Niemann-Pick C1 (NPC1)-deficient cell line, suggesting the necessity for NPC1 activity for efficient LUJpv infection. IMPORTANCE LUJV is a newly identified arenavirus associated with a VHF outbreak in southern Africa. Although cell entry for many arenaviruses has been studied, cell entry for LUJV has not been characterized. In this study, we found that LUJpv utilizes neither αDG nor hTfR1 as a receptor and found unique characteristics of LUJV glycoprotein in membrane fusion and cell entry. Proper exclusion of cholesterol or some kinds of lipids may play important roles in LUJpv cell entry.
Collapse
|
32
|
Shimojima M, Takenouchi A, Shimoda H, Kimura N, Maeda K. Distinct usage of three C-type lectins by Japanese encephalitis virus: DC-SIGN, DC-SIGNR, and LSECtin. Arch Virol 2014; 159:2023-31. [PMID: 24623090 PMCID: PMC7087284 DOI: 10.1007/s00705-014-2042-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 02/28/2014] [Indexed: 01/08/2023]
Abstract
Infection with West Nile virus and dengue virus, two mosquito-borne flaviviruses, is enhanced by two calcium-dependent lectins: dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN), and its related molecule (DC-SIGNR). The present study examined the relationship between Japanese encephalitis virus (JEV) infection and three lectins: DC-SIGN, DC-SIGNR, and liver sinusoidal endothelial cell lectin (LSECtin). Expression of DC-SIGNR resulted in robust JEV proliferation in a lymphoid cell line, Daudi cells, which was otherwise non-permissive to infection. DC-SIGN expression caused moderate JEV proliferation, with effects that varied according to the cells in which JEV was prepared. LSECtin expression had comparatively minor, but consistent, effects, in all cell types used in JEV preparation. While DC-SIGN/DC-SIGNR-mediated JEV infection was inhibited by yeast mannan, LSECtin-mediated infection was inhibited by N-acetylglucosamine β1-2 mannose. Although involvement of DC-SIGN/DC-SIGNR in infection seems to be a common characteristic, this is the first report on usage of LSECtin in mosquito-borne flavivirus infection.
Collapse
MESH Headings
- Animals
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Encephalitis Virus, Japanese/genetics
- Encephalitis Virus, Japanese/physiology
- Encephalitis, Japanese/genetics
- Encephalitis, Japanese/metabolism
- Encephalitis, Japanese/virology
- Humans
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Virus/genetics
- Receptors, Virus/metabolism
Collapse
Affiliation(s)
- Masayuki Shimojima
- Laboratory of Veterinary Microbiology, Faculty of Agriculture, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515 Japan
- Present Address: Special Pathogens Laboratory, Department of Virology I, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011 Japan
| | - Atsushi Takenouchi
- Laboratory of Veterinary Microbiology, Faculty of Agriculture, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515 Japan
| | - Hiroshi Shimoda
- Laboratory of Veterinary Microbiology, Faculty of Agriculture, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515 Japan
| | - Naho Kimura
- Laboratory of Veterinary Microbiology, Faculty of Agriculture, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515 Japan
| | - Ken Maeda
- Laboratory of Veterinary Microbiology, Faculty of Agriculture, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515 Japan
| |
Collapse
|
33
|
Martinez MG, Bialecki MA, Belouzard S, Cordo SM, Candurra NA, Whittaker GR. Utilization of human DC-SIGN and L-SIGN for entry and infection of host cells by the New World arenavirus, Junín virus. Biochem Biophys Res Commun 2013; 441:612-617. [PMID: 24183720 PMCID: PMC4096786 DOI: 10.1016/j.bbrc.2013.10.106] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 10/21/2013] [Indexed: 11/18/2022]
Abstract
The C-type lectins hDC-SIGN and hL-SIGN are important for entry of Junín arenavirus. hDC-SIGN and hL-SIGN substantially augment the use of transferrin receptor. hL-SIGN may act as a co-receptor for Junín arenavirus.
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.
Collapse
Affiliation(s)
- M Guadalupe Martinez
- Department of Microbiology & Immunology, College of Veterinary Medicine, Cornell University, Ithaca NY 14853, USA.,Laboratorio de Virología, Departamento de Química Biológica IQUIBICEN - CONICET. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Ciudad Universitaria, Pabellón II, Piso 4, 1428, Buenos Aires, Argentina
| | - Michele A Bialecki
- Department of Microbiology & Immunology, College of Veterinary Medicine, Cornell University, Ithaca NY 14853, USA
| | - Sandrine Belouzard
- Department of Microbiology & Immunology, College of Veterinary Medicine, Cornell University, Ithaca NY 14853, USA
| | - Sandra M Cordo
- Laboratorio de Virología, Departamento de Química Biológica IQUIBICEN - CONICET. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Ciudad Universitaria, Pabellón II, Piso 4, 1428, Buenos Aires, Argentina
| | - Nélida A Candurra
- Laboratorio de Virología, Departamento de Química Biológica IQUIBICEN - CONICET. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Ciudad Universitaria, Pabellón II, Piso 4, 1428, Buenos Aires, Argentina
| | - Gary R Whittaker
- Department of Microbiology & Immunology, College of Veterinary Medicine, Cornell University, Ithaca NY 14853, USA
| |
Collapse
|
34
|
Abstract
The C-type lectins DC-SIGN, DC-SIGNR and LSECtin are encoded by the lectin gene cluster on chromosome 19p13.3 and perform cell-adhesion and pathogen recognition functions on dendritic cells, liver cells and lymph node sinusoidal endothelial cells. DC-SIGN and DC-SIGNR share similar overall gene and protein molecule structures, and they exhibit high affinity for high-mannose carbohydrates. LSECtin, a Ca2+-dependent C-type lectin, interacts with mannose, NAcGlc and fucose. These lectins allow pathogen recognition (e.g., viruses, bacteria and allergens) and cell adhesion for dendritic and endothelial cells in different tissues, which may enhance the infection and facilitate the spread of those pathogens. A better understanding of these lectins may yield information about how pathogens are captured by particular cells and how they spread in different tissues. These studies would provide more detail about the physiopathological mechanisms of viral and bacterial infections and may also lead to new strategies to treat or prevent infections.
Collapse
Affiliation(s)
- Feng Zhang
- 1Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, Dalian, China
| | | | | |
Collapse
|
35
|
Lukashevich IS. The search for animal models for Lassa fever vaccine development. Expert Rev Vaccines 2013; 12:71-86. [PMID: 23256740 DOI: 10.1586/erv.12.139] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Lassa virus (LASV) is the most prevalent arenavirus in West Africa and is responsible for several hundred thousand infections and thousands of deaths annually. The sizeable disease burden, numerous imported cases of Lassa fever (LF) and the possibility that LASV can be used as an agent of biological warfare make a strong case for vaccine development. Currently there is no licensed LF vaccine and research and devlopment is hampered by the high cost of nonhuman primate animal models and by biocontainment requirements (BSL-4). In addition, a successful LF vaccine has to induce a strong cell-mediated cross-protective immunity against different LASV lineages. All of these challenges will be addressed in this review in the context of available and novel animal models recently described for evaluation of LF vaccine candidates.
Collapse
Affiliation(s)
- Igor S Lukashevich
- Department of Pharmacology and Toxicology, School of Medicine and the Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
36
|
Abstract
A recent publication indicated that overexpression of Axl, a cellular receptor that negatively regulates Toll-like receptor signaling, enhanced the entry of viruses pseudotyped with the glycoprotein of lymphocytic choriomeningitis virus (LCMV) in vitro. In testing the biological relevance of these observations, we found differences in neither viral kinetics between LCMV infections of Axl(-/-) and wild-type mice nor T-cell responses prior to spontaneous viral clearance. Thus, Axl is not required for productive LCMV infection of mice.
Collapse
|
37
|
Droniou-Bonzom ME, Cannon PM. A systems biology starter kit for arenaviruses. Viruses 2012; 4:3625-46. [PMID: 23342371 PMCID: PMC3528283 DOI: 10.3390/v4123625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 11/28/2012] [Accepted: 12/05/2012] [Indexed: 12/05/2022] Open
Abstract
Systems biology approaches in virology aim to integrate viral and host biological networks, and thus model the infection process. The growing availability of high-throughput “-omics” techniques and datasets, as well as the ever-increasing sophistication of in silico modeling tools, has resulted in a corresponding rise in the complexity of the analyses that can be performed. The present study seeks to review and organize published evidence regarding virus-host interactions for the arenaviruses, from alterations in the host proteome during infection, to reported protein-protein interactions. In this way, we hope to provide an overview of the interplay between arenaviruses and the host cell, and lay the foundations for complementing current arenavirus research with a systems-level approach.
Collapse
Affiliation(s)
- Magali E Droniou-Bonzom
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, Los Angeles, CA 90033, USA.
| | | |
Collapse
|
38
|
Abstract
Arenaviruses are a family of enveloped negative-stranded RNA viruses that can cause severe human disease ranging from encephalitis symptoms to fulminant hemorrhagic fever. The bi‑segmented RNA genome encodes four polypeptides: the nucleoprotein NP, the surface glycoprotein GP, the polymerase L, and the RING finger protein Z. Although it is the smallest arenavirus protein with a length of 90 to 99 amino acids and a molecular weight of approx. 11 kDa, the Z protein has multiple functions in the viral life cycle including (i) regulation of viral RNA synthesis, (ii) orchestration of viral assembly and budding, (iii) interaction with host cell proteins, and (iv) interferon antagonism. In this review, we summarize our current understanding of the structural and functional role of the Z protein in the arenavirus replication cycle.
Collapse
Affiliation(s)
- Sarah Katharina Fehling
- Institut für Virologie der Philipps-Universität Marburg, Hans-Meerwein-Str. 2, 35043 Marburg, Germany.
| | | | | |
Collapse
|
39
|
Lukashevich IS. Advanced vaccine candidates for Lassa fever. Viruses 2012; 4:2514-57. [PMID: 23202493 PMCID: PMC3509661 DOI: 10.3390/v4112514] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 10/20/2012] [Accepted: 10/22/2012] [Indexed: 12/16/2022] Open
Abstract
Lassa virus (LASV) is the most prominent human pathogen of the Arenaviridae. The virus is transmitted to humans by a rodent reservoir, Mastomys natalensis, and is capable of causing lethal Lassa Fever (LF). LASV has the highest human impact of any of the viral hemorrhagic fevers (with the exception of Dengue Fever) with an estimated several hundred thousand infections annually, resulting in thousands of deaths in Western Africa. The sizeable disease burden, numerous imported cases of LF in non-endemic countries, and the possibility that LASV can be used as an agent of biological warfare make a strong case for vaccine development. Presently there is no licensed vaccine against LF or approved treatment. Recently, several promising vaccine candidates have been developed which can potentially target different groups at risk. The purpose of this manuscript is to review the LASV pathogenesis and immune mechanisms involved in protection. The current status of pre-clinical development of the advanced vaccine candidates that have been tested in non-human primates will be discussed. Major scientific, manufacturing, and regulatory challenges will also be considered.
Collapse
Affiliation(s)
- Igor S Lukashevich
- Department of Pharmacology and Toxicology, School of Medicine, and Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Kentucky, USA.
| |
Collapse
|
40
|
[Arenavirus infections]. Uirusu 2012; 62:229-38. [PMID: 24153233 DOI: 10.2222/jsv.62.229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Arenaviruses are the collective name for viruses, which belong to the family Arenaviridae. They replicate in the cytoplasm of cells, and were named after the sandy (Latin, arenosus) appearance of the ribosomes often seen in thin sections of virions under electron microscope. Several arenaviruses, such as Lassa virus in West Africa, and Junin, Guanarito, Sabia, Machupo, and Chapare viruses in South America, cause sever viral hemorrhagic fevers (VHF) in humans and represent a serious public health problem. These viruses are categorized as category 1 pathogens thus should be handles in a BSL4 laboratory. Recently, Lujo virus was isolated as a newly discovered novel arenavirus associated with a VHF outbreak in southern Africa in 2008. Although, we have no VHF patients caused by arenaviruses in Japan, except for a single imported Lassa fever case in 1987, it is possible that VHF patients occur as imported cases as for other VHF in the future. Therefore, it is necessary to develop the diagnostics and therapeutics in consideration of patient's severe symptoms and high mortality even in the disease-free countries. In this review, we will broadly discuss the current knowledge from the basic researches to diagnostics and vaccine developments for arenavirus diseases.
Collapse
|