1
|
Besson ME, Pépin M, Metral PA. Lassa Fever: Critical Review and Prospects for Control. Trop Med Infect Dis 2024; 9:178. [PMID: 39195616 PMCID: PMC11359316 DOI: 10.3390/tropicalmed9080178] [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: 07/21/2024] [Revised: 08/10/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024] Open
Abstract
Lassa Fever is a deadly viral haemorrhagic disease, causing annually several hundreds of deaths in West Africa. This zoonotic disease is primarily transmitted to humans by rodents of the genus Mastomys, even though other rodents reportedly carry the Lassa virus, while secondary interhuman transmission accounts for approximately 20% of cases. Although this disease has been endemic in rural zones of Nigeria, Sierra Leone, Liberfia, and Guinea for hundreds of years, it is also characterised by epidemic outbreaks in the dry season, responsible for heavy death tolls. No licensed vaccine or satisfying treatment is currently available. Disease management is hindered by the incomplete knowledge of the epidemiology and distribution of the disease, resulting from an inadequate health and surveillance system. Additional scientific constraints such as the genetic diversity of the virus and the lack of understanding of the mechanisms of immune protection complexify the development of a vaccine. The intricate socio-economic context in the affected regions, and the lack of monetary incentive for drug development, allow the disease to persist in some of West Africa's poorest communities. The increase in the number of reported cases and in the fatality rate, the expansion of the endemic area, as well as the threat Lassa Fever represents internationally should urge the global community to work on the disease control and prevention. The disease control requires collaborative research for medical countermeasures and tailored public health policies. Lassa Fever, created by the interconnection between animals, humans, and ecosystems, and embedded in an intricate social context, should be addressed with a 'One Health' approach. This article provides an overview of Lassa Fever, focusing on Nigeria, and discusses the perspectives for the control of disease.
Collapse
Affiliation(s)
- Marianne E. Besson
- Department of Public Health, Royal Veterinary College, London NW1 0TU, UK
| | - Michel Pépin
- Department of Virology and Infectiology, VetAgro Sup Lyon University, 69280 Marcy L’Etoile, France;
| | | |
Collapse
|
2
|
Moon-Walker A, Zhang Z, Zyla DS, Buck TK, Li H, Diaz Avalos R, Schendel SL, Hastie KM, Crotty S, Saphire EO. Structural basis for antibody-mediated neutralization of lymphocytic choriomeningitis virus. Cell Chem Biol 2023; 30:403-411.e4. [PMID: 36990092 PMCID: PMC11090681 DOI: 10.1016/j.chembiol.2023.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 12/23/2022] [Accepted: 03/08/2023] [Indexed: 03/30/2023]
Abstract
The mammarenavirus lymphocytic choriomeningitis virus (LCMV) is a globally distributed zoonotic pathogen that can be lethal in immunocompromised patients and can cause severe birth defects if acquired during pregnancy. The structure of the trimeric surface glycoprotein, essential for entry, vaccine design, and antibody neutralization, remains unknown. Here, we present the cryoelectron microscopy (cryo-EM) structure of the LCMV surface glycoprotein (GP) in its trimeric pre-fusion assembly both alone and in complex with a rationally engineered monoclonal neutralizing antibody termed 18.5C-M28 (M28). Additionally, we show that passive administration of M28, either as a prophylactic or therapeutic, protects mice from LCMV clone 13 (LCMVcl13) challenge. Our study illuminates not only the overall structural organization of LCMV GP and the mechanism for its inhibition by M28 but also presents a promising therapeutic candidate to prevent severe or fatal disease in individuals who are at risk of infection by a virus that poses a threat worldwide.
Collapse
Affiliation(s)
- Alex Moon-Walker
- La Jolla Institute for Immunology; La Jolla, CA 92037, USA; Program in Virology, Harvard Medical School, Boston, MA 02115, USA; Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MI 63110, USA
| | - Zeli Zhang
- La Jolla Institute for Immunology; La Jolla, CA 92037, USA
| | - Dawid S Zyla
- La Jolla Institute for Immunology; La Jolla, CA 92037, USA
| | - Tierra K Buck
- La Jolla Institute for Immunology; La Jolla, CA 92037, USA; Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Haoyang Li
- La Jolla Institute for Immunology; La Jolla, CA 92037, USA
| | | | | | | | - Shane Crotty
- La Jolla Institute for Immunology; La Jolla, CA 92037, USA.
| | | |
Collapse
|
3
|
de Oliveira ALR, Cunha MS, Bisordi I, de Souza RP, Timenetsky MDCST. Serological evidence of arenavirus circulation in wild rodents from central-west, southeast, and south regions of Brazil, 2002-2006. Braz J Microbiol 2023; 54:279-284. [PMID: 36441413 PMCID: PMC9943984 DOI: 10.1007/s42770-022-00858-3] [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: 03/24/2022] [Accepted: 10/24/2022] [Indexed: 11/30/2022] Open
Abstract
Viral hemorrhagic fevers caused by arenaviruses are severe zoonotic diseases. In reservoirs, the presence of antibodies may indicate viral circulation in a population of a specific region, and these data can be used as an indicator for further investigations by molecular techniques. The present study aimed to detect the presence of arenavirus antibodies in wild rodents captured from 1998 to 2008 during epidemiological surveillance activities. A retrospective analysis of 2243 wild rodent blood samples using a broad cross-reactive in-house developed enzyme-linked immunosorbent assay (ELISA) revealed a 0.44% (10/2243) positive rate in wild rodents, which included Necromys lasiurus (6/1012), Calomys callosus (2/94), and Akodon sp. (2/273) species. These rodents were captured between 2002 to 2006 in Campo Alegre de Goiás/GO, Bodoquena/MS, Nuporanga/SP, and Mogi das Cruzes/SP. Our findings suggest the sylvatic circulation of arenavirus among wild rodents in the southeast region of Brazil. However, future virological and molecular studies are necessary to confirm the viral presence in these regions.
Collapse
Affiliation(s)
- Ana Lúcia Rodrigues de Oliveira
- Núcleo de Doenças de Transmissão Vetorial Do Centro de Virologia do Instituto Adolfo Lutz, SP, São Paulo, Brazil.
- Programa de Pós-Graduação Em Ciências, Mestrado Em Pesquisa Laboratoriais Em Saúde Pública da Coordenadoria de Controle de Doenças da Secretaria de Estado da Saúde de São Paulo, São Paulo, SP, Brazil.
| | - Mariana Sequetin Cunha
- Núcleo de Doenças de Transmissão Vetorial Do Centro de Virologia do Instituto Adolfo Lutz, SP, São Paulo, Brazil.
| | - Ivani Bisordi
- Núcleo de Doenças de Transmissão Vetorial Do Centro de Virologia do Instituto Adolfo Lutz, SP, São Paulo, Brazil
| | - Renato Pereira de Souza
- Núcleo de Doenças de Transmissão Vetorial Do Centro de Virologia do Instituto Adolfo Lutz, SP, São Paulo, Brazil
| | - Maria do Carmo Sampaio Tavares Timenetsky
- Programa de Pós-Graduação Em Ciências, Mestrado Em Pesquisa Laboratoriais Em Saúde Pública da Coordenadoria de Controle de Doenças da Secretaria de Estado da Saúde de São Paulo, São Paulo, SP, Brazil
- Centro de Virologia Do, Instituto Adolfo Lutz, São Paulo, SP, Brazil
| |
Collapse
|
4
|
Mammarenavirus Genetic Diversity and Its Biological Implications. Curr Top Microbiol Immunol 2023; 439:265-303. [PMID: 36592249 DOI: 10.1007/978-3-031-15640-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Members of the family Arenaviridae are classified into four genera: Antennavirus, Hartmanivirus, Mammarenavirus, and Reptarenavirus. Reptarenaviruses and hartmaniviruses infect (captive) snakes and have been shown to cause boid inclusion body disease (BIBD). Antennaviruses have genomes consisting of 3, rather than 2, segments, and were discovered in actinopterygian fish by next-generation sequencing but no biological isolate has been reported yet. The hosts of mammarenaviruses are mainly rodents and infections are generally asymptomatic. Current knowledge about the biology of reptarenaviruses, hartmaniviruses, and antennaviruses is very limited and their zoonotic potential is unknown. In contrast, some mammarenaviruses are associated with zoonotic events that pose a threat to human health. This review will focus on mammarenavirus genetic diversity and its biological implications. Some mammarenaviruses including lymphocytic choriomeningitis virus (LCMV) are excellent experimental model systems for the investigation of acute and persistent viral infections, whereas others including Lassa (LASV) and Junin (JUNV) viruses, the causative agents of Lassa fever (LF) and Argentine hemorrhagic fever (AHF), respectively, are important human pathogens. Mammarenaviruses were thought to have high degree of intra-and inter-species amino acid sequence identities, but recent evidence has revealed a high degree of mammarenavirus genetic diversity in the field. Moreover, closely related mammarenavirus can display dramatic phenotypic differences in vivo. These findings support a role of genetic variability in mammarenavirus adaptability and pathogenesis. Here, we will review the molecular biology of mammarenaviruses, phylogeny, and evolution, as well as the quasispecies dynamics of mammarenavirus populations and their biological implications.
Collapse
|
5
|
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: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
6
|
Geldenhuys M, Weyer J, Kearney T, Markotter W. Host-Associated Distribution of Two Novel Mammarenaviruses in Rodents from Southern Africa. Viruses 2022; 15:99. [PMID: 36680139 PMCID: PMC9861163 DOI: 10.3390/v15010099] [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: 12/08/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Mammarenaviruses are hosted by several rodent species, a small number of which have been known to be zoonotic. Host surveillance among small mammals has identified a large diversity of previously undescribed mammarenaviruses. Intensified biosurveillance is warranted to better understand the diversity of these agents. Longitudinal host surveillance involving non-volant small mammals at a site in the Limpopo province, South Africa, was conducted. The study reports on the screening results of 563 samples for the presence of mammarenavirus RNA. PCR-positive samples were subjected to sequencing using Miseq amplicon sequencing. Sequences with close similarity to Mariental and Lunk viruses were identified from two rodent species, Micaelamys namaquensis and Mus minutoides. This represents the first description of these viruses from South Africa. The genomic sequences reported here partially satisfied the requirements put forward by the International Committee on the Taxonomy of Viruses' criteria for species delineation, suggesting that these may be new strains of existing species. The known distribution of these mammarenaviruses is thus expanded further south in Africa.
Collapse
Affiliation(s)
- Marike Geldenhuys
- Centre for Viral Zoonoses, Department of Medical Virology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| | - Jacqueline Weyer
- Centre for Viral Zoonoses, Department of Medical Virology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
- Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg 2131, South Africa
- Department of Microbiology and Infectious Diseases, School of Pathology, University of Witwatersrand, Johannesburg 2131, South Africa
| | - Teresa Kearney
- Ditsong National Museum of Natural History, Pretoria 0001, South Africa
- Department of Zoology and Entomology, University of Pretoria, Pretoria 0001, South Africa
| | - Wanda Markotter
- Centre for Viral Zoonoses, Department of Medical Virology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| |
Collapse
|
7
|
Alfaro-Alarcón A, Hetzel U, Smura T, Baggio F, Morales JA, Kipar A, Hepojoki J. Boid Inclusion Body Disease Is Also a Disease of Wild Boa Constrictors. Microbiol Spectr 2022; 10:e0170522. [PMID: 36094085 PMCID: PMC9602588 DOI: 10.1128/spectrum.01705-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 08/05/2022] [Indexed: 12/31/2022] Open
Abstract
Reptarenaviruses cause boid inclusion body disease (BIBD), a potentially fatal disease, occurring in captive constrictor snakes boas and pythons worldwide. Classical BIBD, characterized by the formation of pathognomonic cytoplasmic inclusion bodies (IBs), occurs mainly in boas, whereas in pythons, for example, reptarenavirus infection most often manifests as central nervous system signs with limited IB formation. The natural hosts of reptarenaviruses are unknown, although free-ranging/wild constrictor snakes are among the suspects. Here, we report BIBD with reptarenavirus infection in indigenous captive and wild boid snakes in Costa Rica using histology, immunohistology, transmission electron microscopy, and next-generation sequencing (NGS). The snakes studied represented diagnostic postmortem cases of captive and wild-caught snakes since 1989. The results from NGS on archival paraffin blocks confirm that reptarenaviruses were already present in wild boa constrictors in Costa Rica in the 1980s. Continuous sequences that were de novo assembled from the low-quality RNA obtained from paraffin-embedded tissue allowed the identification of a distinct pair of reptarenavirus S and L segments in all studied animals; in most cases, reference assembly could recover almost complete segments. Sampling of three prospective cases in 2018 allowed an examination of fresh blood or tissues and resulted in the identification of additional reptarenavirus segments and hartmanivirus coinfection. Our results show that BIBD is not only a disease of captive snakes but also occurs in indigenous wild constrictor snakes in Costa Rica, suggesting boa constrictors to play a role in natural reptarenavirus circulation. IMPORTANCE The literature describes cases of boid inclusion body disease (BIBD) in captive snakes since the 1970s, and in the 2010s, others and ourselves identified reptarenaviruses as the causative agent. BIBD affects captive snakes globally, but the origin and the natural host of reptarenaviruses remain unknown. In this report, we show BIBD and reptarenavirus infections in two native Costa Rican constrictor snake species, and by studying archival samples, we show that both the viruses and the disease have been present in free-ranging/wild snakes in Costa Rica at least since the 1980s. The diagnosis of BIBD in wild boa constrictors suggests that this species plays a role in the circulation of reptarenaviruses. Additional sample collection and analysis would help to clarify this role further and the possibility of, e.g., vector transmission from an arthropod host.
Collapse
Affiliation(s)
- Alejandro Alfaro-Alarcón
- Departamento de Patología, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
| | - Udo Hetzel
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- University of Helsinki, Faculty of Veterinary Medicine, Department of Veterinary Biosciences, Helsinki, Finland
| | - Teemu Smura
- University of Helsinki, Faculty of Medicine, Medicum, Department of Virology, Helsinki, Finland
| | - Francesca Baggio
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Juan Alberto Morales
- Departamento de Patología, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
| | - Anja Kipar
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- University of Helsinki, Faculty of Veterinary Medicine, Department of Veterinary Biosciences, Helsinki, Finland
| | - Jussi Hepojoki
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- University of Helsinki, Faculty of Medicine, Medicum, Department of Virology, Helsinki, Finland
| |
Collapse
|
8
|
Abstract
Mammarenaviruses establish a persistent infection in their rodent and bat hosts, and the evidence suggests that reptarenaviruses and hartmaniviruses found in captive snakes act similarly. In snakes, reptarenaviruses cause boid inclusion body disease (BIBD), which is often associated with secondary infections. Snakes with BIBD usually carry more than a single pair of reptarenavirus S and L segments and occasionally demonstrate hartmanivirus coinfection. Here, we reported the generation of cell lines persistently infected with a single or two reptarenavirus(es) and a cell line with persistent reptarenavirus-hartmanivirus coinfection. By RT-PCR we demonstrated that the amount of viral RNA within the persistently infected cells remains at levels similar to those observed following initial infection. Using antibodies against the glycoproteins (GPs) and nucleoprotein (NP) of reptarenaviruses, we studied the levels of viral protein in cells passaged 10 times after the original inoculation and observed that the expression of GPs declines dramatically during persistent infection, unlike the expression of NP. Immunofluorescence (IF) staining served to demonstrate differences in the distribution of NP within the persistently infected compared to freshly infected cells. IF staining of cells inoculated with the viruses secreted from the persistently infected cell lines produced similar NP staining compared to cells infected with a traditionally passaged virus, suggesting that the altered NP expression pattern of persistently infected cells does not relate to changes in the virus. The cell cultures described herein can serve as tools for studying the coinfection and superinfection interplay between reptarenaviruses and studying the BIBD pathogenesis mechanisms. IMPORTANCE Mammarenaviruses cause a persistent infection in their natural rodent and bat hosts. Reptarenaviruses cause boid inclusion body disease (BIBD) in constrictor snakes, but it is unclear whether snakes are the natural host of these viruses. In this study, we showed that reptarenaviruses established a persistent infection in cultured Boa constrictor cells and that the persistently infected cells continued to produce infectious virus. Our results showed that persistent infection results from subsequent passaging of cells inoculated with a single reptarenavirus, two reptarenaviruses, or even when inoculating the cells with reptarenavirus and hartmanivirus (another arenavirus genus). The results further suggested that coinfection would not result in overt competition between the different reptarenaviruses, thus helping to explain the frequent reptarenavirus coinfections in snakes with BIBD. The established cell culture models of persistent infection could help to elucidate the role of coinfection and superinfection and potential immunosuppression as the pathogenic mechanisms behind BIBD.
Collapse
|
9
|
Abstract
Lymphocytic choriomeningitis virus (LCMV) is the prototypic arenavirus and has been utilized for decades as a model to understand the host immune response against viral infection. LCMV infection can lead to fatal meningitis in immunocompromised people and can lead to congenital birth defects and spontaneous abortion if acquired during pregnancy. Using a genetic screen, we uncover host factors involved in LCMV entry that were previously unknown and are candidate therapeutic targets to combat LCMV infection. This study expands our understanding of the entry pathway of LCMV, revealing that its glycoprotein switches from utilizing the known receptor α-DG and heparan sulfate at the plasma membrane to binding the lysosomal mucin CD164 at pH levels found in endolysosomal compartments, facilitating membrane fusion. Lymphocytic choriomeningitis virus (LCMV) is a rodent-borne zoonotic arenavirus that causes congenital abnormalities and can be fatal for transplant recipients. Using a genome-wide loss-of-function screen, we identify host factors required for LCMV entry into cells. We identify the lysosomal mucin CD164, glycosylation factors, the heparan sulfate biosynthesis machinery, and the known receptor alpha-dystroglycan (α-DG). Biochemical analysis revealed that the LCMV glycoprotein binds CD164 at acidic pH and requires a sialylated glycan at residue N104. We demonstrate that LCMV entry proceeds by the virus switching binding from heparan sulfate or α-DG at the plasma membrane to CD164 prior to membrane fusion, thus identifying additional potential targets for therapeutic intervention.
Collapse
|
10
|
Tapia-Ramírez G, Lorenzo C, Navarrete D, Carrillo-Reyes A, Retana Ó, Carrasco-Hernández R. A Review of Mammarenaviruses and Rodent Reservoirs in the Americas. ECOHEALTH 2022; 19:22-39. [PMID: 35247117 PMCID: PMC9090702 DOI: 10.1007/s10393-022-01580-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 06/14/2023]
Abstract
In the Americas, infectious viral diseases caused by viruses of the genus Mammarenavirus have been reported since the 1960s. Such diseases have commonly been associated with land use changes, which favor abundance of generalist rodent species. In the Americas-where the rates of land use change are among the highest worldwide-at least 1326 of all 2277 known rodent species have been reported. We conducted a literature review of studies between 1960 and 2020, to establish the current and historical knowledge about genotypes of mammarenaviruses and their rodent reservoirs in the Americas. Our overall goal was to show the importance of focusing research efforts on the American continent, since the conditions exist for future viral hemorrhagic fever (VHF) outbreaks caused by rodent-borne viruses, in turn, carried by widely distributed rodents. We found 47 species identified down to the species level, and one species identified only down to the genus level (Oryzomys sp.), reported in the Americas as reservoirs of mammarenaviruses, most these are ecological generalists. These species associate with 29 genotypes of Mammarenavirus, seven of which have been linked to VHFs in humans. We also highlight the need to monitor these species, in order to prevent viral disease outbreaks in the region.
Collapse
Affiliation(s)
- Gloria Tapia-Ramírez
- Departamento de Conservación de la Biodiversidad, El Colegio de La Frontera Sur, Periférico Sur S/N María Auxiliadora, 29290, San Cristóbal de Las Casas, Chiapas, Mexico.
| | - Consuelo Lorenzo
- Departamento de Conservación de la Biodiversidad, El Colegio de La Frontera Sur, Periférico Sur S/N María Auxiliadora, 29290, San Cristóbal de Las Casas, Chiapas, Mexico
| | - Darío Navarrete
- Departamento de Observación de la Tierra, Atmósfera y Océano, El Colegio de La Frontera Sur, Periférico Sur S/N María Auxiliadora, 29290, San Cristóbal de Las Casas, Chiapas, Mexico
| | - Arturo Carrillo-Reyes
- Facultad de Ingeniería, Universidad de Ciencias y Artes de Chiapas, Av 1a. Sur Pte 1460, C.P., 29000, Tuxtla Gutiérrez, Chiapas, Mexico
| | - Óscar Retana
- Centro de Estudios en Desarrollo Sustentable, Universidad Autónoma de Campeche, Avenida Héroe de Nacozari 480, C.P., 24079, San Francisco de Campeche, Campeche, Mexico
| | - Rocío Carrasco-Hernández
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Ismael Cosío Villegas, Calz. de Tlalpan 4502, C. P., 14080, Ciudad de México, Mexico
| |
Collapse
|
11
|
Development of Reverse Genetics for the Prototype New World Mammarenavirus Tacaribe Virus. J Virol 2020; 94:JVI.01014-20. [PMID: 32669332 DOI: 10.1128/jvi.01014-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/02/2020] [Indexed: 11/20/2022] Open
Abstract
The New World mammarenavirus Tacaribe virus (TCRV) has been isolated from fruit bats, mosquitoes, and ticks, whereas all other known New World mammarenaviruses are maintained in rodents. TCRV has not been linked to human disease, but it has been shown to protect against Argentine hemorrhagic fever-like disease in marmosets infected with the New World mammarenavirus Junín virus (JUNV), indicating the potential of TCRV as a live-attenuated vaccine for the treatment of Argentine hemorrhagic fever. Implementation of TCRV as a live-attenuated vaccine or a vaccine vector would be facilitated by the establishment of reverse genetics systems for the genetic manipulation of the TCRV genome. In this study, we developed, for the first time, reverse genetics approaches for the generation of recombinant TCRV (rTCRV). We successfully rescued a wild-type (WT) rTCRV (a trisegmented form of TCRV expressing two reporter genes [r3TCRV]) and a bisegmented TCRV expressing a single reporter gene from a bicistronic viral mRNA (rTCRV/GFP). These reverse genetics approaches represent an excellent tool to investigate the biology of TCRV and to explore its potential use as a live-attenuated vaccine or a vaccine vector for the treatment of other viral infections. Notably, we identified a 39-nucleotide (nt) deletion (Δ39) in the noncoding intergenic region (IGR) of the viral large (L) segment that is required for optimal virus multiplication. Accordingly, an rTCRV containing this 39-nt deletion in the L-IGR (rTCRV/Δ39) exhibited decreased viral fitness in cultured cells, suggesting the feasibility of using this deletion in the L-IGR as an approach to attenuate TCRV, and potentially other mammarenaviruses, for their implementation as live-attenuated vaccines or vaccine vectors.IMPORTANCE To date, no Food and Drug Administration (FDA)-approved vaccines are available to combat hemorrhagic fever caused by mammarenavirus infections in humans. Treatment of mammarenavirus infections is limited to the off-label use of ribavirin, which is partially effective and associated with significant side effects. Tacaribe virus (TCRV), the prototype member of the New World mammarenaviruses, is nonpathogenic in humans but able to provide protection against Junín virus (JUNV), the causative agent of Argentine hemorrhagic fever, demonstrating the feasibility of using TCRV as a live-attenuated vaccine vector for the treatment of JUNV and potentially other viral infections. Here, we describe for the first time the feasibility of generating recombinant TCRV (rTCRV) using reverse genetics approaches, which paves the way to study the biology of TCRV and also its potential use as a live-attenuated vaccine or a vaccine vector for the treatment of mammarenavirus and/or other viral infections in humans.
Collapse
|
12
|
Distinct Molecular Mechanisms of Host Immune Response Modulation by Arenavirus NP and Z Proteins. Viruses 2020; 12:v12070784. [PMID: 32708250 PMCID: PMC7412275 DOI: 10.3390/v12070784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 12/12/2022] Open
Abstract
Endemic to West Africa and South America, mammalian arenaviruses can cross the species barrier from their natural rodent hosts to humans, resulting in illnesses ranging from mild flu-like syndromes to severe and fatal haemorrhagic zoonoses. The increased frequency of outbreaks and associated high fatality rates of the most prevalent arenavirus, Lassa, in West African countries, highlights the significant risk to public health and to the socio-economic development of affected countries. The devastating impact of these viruses is further exacerbated by the lack of approved vaccines and effective treatments. Differential immune responses to arenavirus infections that can lead to either clearance or rapid, widespread and uncontrolled viral dissemination are modulated by the arenavirus multifunctional proteins, NP and Z. These two proteins control the antiviral response to infection by targeting multiple cellular pathways; and thus, represent attractive targets for antiviral development to counteract infection. The interplay between the host immune responses and viral replication is a key determinant of virus pathogenicity and disease outcome. In this review, we examine the current understanding of host immune defenses against arenavirus infections and summarise the host protein interactions of NP and Z and the mechanisms that govern immune evasion strategies.
Collapse
|
13
|
Biological Characterization of Conserved Residues within the Cytoplasmic Tail of the Pichinde Arenaviral Glycoprotein Subunit 2 (GP2). J Virol 2019; 93:JVI.01277-19. [PMID: 31462569 DOI: 10.1128/jvi.01277-19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 08/20/2019] [Indexed: 12/13/2022] Open
Abstract
Several mammarenaviruses can cause deadly hemorrhagic fever infections in humans, with limited preventative and therapeutic measures available. Arenavirus cell entry is mediated by the viral glycoprotein (GP) complex, which consists of the stable signal peptide (SSP), the receptor-binding subunit GP1, and the transmembrane subunit GP2. The GP2 cytoplasmic tail (CT) is relatively conserved among arenaviruses and is known to interact with the SSP to regulate GP processing and membrane fusion, but its biological role in the context of an infectious virus has not been fully characterized. Using a Pichinde virus (PICV) GP expression vector and a PICV reverse genetics system, we systematically characterized the functional roles of 12 conserved residues within the GP2 CT in GP processing, trafficking, assembly, and fusion, as well as in viral replication. Except for P478A and K505A R508A, alanine substitutions at conserved residues abolished GP processing and membrane fusion in plasmid-transfected cells. Six invariant H and C residues and W503 are essential for viral replication, as evidenced by the fact that their mutant viruses could not be rescued. Both P480A and R482A mutant viruses were rescued, grew similarly to wild-type (WT) virus, and produced evidently processed GP1 and GP2 subunits in virus-infected cells, despite the fact that the same mutations abolished GP processing and membrane fusion in a plasmid-based protein expression system, illustrating the importance of using an infectious-virus system for analyzing viral glycoprotein function. In summary, our results demonstrate an essential biological role of the GP2 CT in arenavirus replication and suggest it as a potential novel target for developing antivirals and/or attenuated viral vaccine candidates.IMPORTANCE Several arenaviruses, such as Lassa virus (LASV), can cause severe and lethal hemorrhagic fever diseases with high mortality and morbidity, for which no FDA-approved vaccines or therapeutics are available. Viral entry is mediated by the arenavirus GP complex, which consists of the stable signal peptide (SSP), the receptor-binding subunit GP1, and the transmembrane subunit GP2. The cytoplasmic tail (CT) of GP2 is highly conserved among arenaviruses, but its functional role in viral replication is not completely understood. Using a reverse genetics system of a prototypic arenavirus, Pichinde virus (PICV), we show that the GP2 CT contains certain conserved residues that are essential for virus replication, implicating it as a potentially good target for developing antivirals and live-attenuated viral vaccines against deadly arenavirus pathogens.
Collapse
|
14
|
Beitzel B, Hulseberg CE, Palacios G. Reverse genetics systems as tools to overcome the genetic diversity of Lassa virus. Curr Opin Virol 2019; 37:91-96. [PMID: 31357141 DOI: 10.1016/j.coviro.2019.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 11/17/2022]
Abstract
Lassa virus is endemic in a large area of sub-Saharan Africa, and exhibits a large amount of genetic diversity. Of the four currently recognized lineages, lineages I-III circulate in Nigeria, and lineage IV circulates in Sierra Leone, Guinea, and Liberia. However, several newly detected lineages have been proposed. LASV genetic diversity may result in differences in pathogenicity or response to medical countermeasures, necessitating the testing of multiple lineages during the development of countermeasures and diagnostics. Logistical and biosafety concerns can make it difficult to obtain representative collections of divergent LASV clades for comparison studies. For example, lack of a cold chain in remote areas, or shipping restrictions on live viruses can prevent the dissemination of natural virus isolates to researchers. Reverse genetics systems that have been developed for LASV can facilitate acquisition of hard-to-obtain LASV strains and enable comprehensive development of medical countermeasures.
Collapse
Affiliation(s)
- Brett Beitzel
- Center for Genome Sciences, The United States Army Medical Research Institute for Infectious Disease, 1425 Porter St., Ft. Detrick, MD 21702, United States
| | - Christine E Hulseberg
- Center for Genome Sciences, The United States Army Medical Research Institute for Infectious Disease, 1425 Porter St., Ft. Detrick, MD 21702, United States
| | - Gustavo Palacios
- Center for Genome Sciences, The United States Army Medical Research Institute for Infectious Disease, 1425 Porter St., Ft. Detrick, MD 21702, United States.
| |
Collapse
|
15
|
Tembo J, Simulundu E, Changula K, Handley D, Gilbert M, Chilufya M, Asogun D, Ansumana R, Kapata N, Ntoumi F, Ippolito G, Zumla A, Bates M. Recent advances in the development and evaluation of molecular diagnostics for Ebola virus disease. Expert Rev Mol Diagn 2019; 19:325-340. [PMID: 30916590 DOI: 10.1080/14737159.2019.1595592] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION The 2014-16 outbreak of ebola virus disease (EVD) in West Africa resulted in 11,308 deaths. During the outbreak only 60% of patients were laboratory confirmed and global health authorities have identified the need for accurate and readily deployable molecular diagnostics as an important component of the ideal response to future outbreaks, to quickly identify and isolate patients. Areas covered: Currently PCR-based techniques and rapid diagnostic tests (RDTs) that detect antigens specific to EVD infections dominate the diagnostic landscape, but recent advances in biosensor technologies have led to novel approaches for the development of EVD diagnostics. This review summarises the literature and available performance data of currently available molecular diagnostics for ebolavirus, identifies knowledge gaps and maps out future priorities for research in this field. Expert opinion: While there are now a plethora of diagnostic tests for EVD at various stages of development, there is an acute need for studies to compare their clinical performance, but the sporadic nature of EVD outbreaks makes this extremely challenging, demanding pragmatic new modalities of research funding and ethical/institutional approval, to enable responsive research in outbreak settings. Retrospective head-to-head diagnostic comparisons could also be implemented using biobanked specimens, providing this can be done safely.
Collapse
Affiliation(s)
- John Tembo
- a HerpeZ , University Teaching hospital , Lusaka , Zambia
| | - Edgar Simulundu
- b Department of Disease Control , University of Zambia School of Veterinary Medicine , Lusaka , Zambia
| | - Katendi Changula
- b Department of Disease Control , University of Zambia School of Veterinary Medicine , Lusaka , Zambia
| | - Dale Handley
- c School of Life Sciences , University of Lincoln , Lincoln , UK
| | - Matthew Gilbert
- c School of Life Sciences , University of Lincoln , Lincoln , UK
| | - Moses Chilufya
- a HerpeZ , University Teaching hospital , Lusaka , Zambia
| | - Danny Asogun
- d Lassa fever research institute , Irrua University Teaching Hospital , Irrua , Nigeria
| | | | - Nathan Kapata
- f Zambia National Public Health Institute , Lusaka , Zambia
| | - Francine Ntoumi
- g Fondation Congolaise pour la Recherche Médicale , Brazzaville , Republic of Congo
| | - Giuseppe Ippolito
- h National Institute for Infectious Diseases , Lazzaro Spallanzani, IRCCS , Rome , Italy
| | - Alimuddin Zumla
- i Centre for Clinical Microbiology, Division of Infection and Immunity , University College London (UCL) , London , UK.,j National Institute of Health and Research Biomedical Research Centre , UCL Hospitals National Health Service Foundation Trust , London , UK
| | - Matthew Bates
- a HerpeZ , University Teaching hospital , Lusaka , Zambia.,c School of Life Sciences , University of Lincoln , Lincoln , UK.,i Centre for Clinical Microbiology, Division of Infection and Immunity , University College London (UCL) , London , UK
| |
Collapse
|
16
|
Brisse ME, Ly H. Hemorrhagic Fever-Causing Arenaviruses: Lethal Pathogens and Potent Immune Suppressors. Front Immunol 2019; 10:372. [PMID: 30918506 PMCID: PMC6424867 DOI: 10.3389/fimmu.2019.00372] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/14/2019] [Indexed: 12/22/2022] Open
Abstract
Hemorrhagic fevers (HF) resulting from pathogenic arenaviral infections have traditionally been neglected as tropical diseases primarily affecting African and South American regions. There are currently no FDA-approved vaccines for arenaviruses, and treatments have been limited to supportive therapy and use of non-specific nucleoside analogs, such as Ribavirin. Outbreaks of arenaviral infections have been limited to certain geographic areas that are endemic but known cases of exportation of arenaviruses from endemic regions and socioeconomic challenges for local control of rodent reservoirs raise serious concerns about the potential for larger outbreaks in the future. This review synthesizes current knowledge about arenaviral evolution, ecology, transmission patterns, life cycle, modulation of host immunity, disease pathogenesis, as well as discusses recent development of preventative and therapeutic pursuits against this group of deadly viral pathogens.
Collapse
Affiliation(s)
- Morgan E Brisse
- Biochemistry, Molecular Biology, and Biophysics Graduate Program, University of Minnesota, St. Paul, MN, United States.,Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Hinh Ly
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| |
Collapse
|
17
|
Virome profiling of rodents in Xinjiang Uygur Autonomous Region, China: Isolation and characterization of a new strain of Wenzhou virus. Virology 2019; 529:122-134. [PMID: 30685659 DOI: 10.1016/j.virol.2019.01.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/09/2019] [Accepted: 01/09/2019] [Indexed: 11/21/2022]
Abstract
Rodents, as the most diverse and widest distributed mammals, are a natural reservoir of many zoonotic viruses. However, little is known about the viral diversity harbored by rodents in China. Here we performed viral metagenomic analyses of 314 wild rodents covering 7 species, sampled in North-western China. We also conducted a systematic virological characterization of a new Wenzhou virus (WENV) isolate, QARn1, from a brown rat (Rattus norvegicus). Full genomic and phylogenetic analyses showed that QARn1 is a previously unidentified strain of Wenzhou mammarenavirus and forms a new branch within the Asian clade. Experimental infection of Sprague-Dawley rats with QARn1 did not present overt pathology, but specific humoral immune responses developed and mild hemorrhage and immunocyte infiltration of the lungs and thymus were observed. These observations have expanded the geographic distribution of WENV to Central Asia, and further confirm that brown rats are natural hosts of Wenzhou virus.
Collapse
|
18
|
Pan X, Wu Y, Wang W, Zhang L, Xiao G. Novel neutralizing monoclonal antibodies against Junin virus. Antiviral Res 2018; 156:21-28. [DOI: 10.1016/j.antiviral.2018.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 10/14/2022]
|
19
|
Widespread arenavirus occurrence and seroprevalence in small mammals, Nigeria. Parasit Vectors 2018; 11:416. [PMID: 30005641 PMCID: PMC6045851 DOI: 10.1186/s13071-018-2991-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 07/03/2018] [Indexed: 11/10/2022] Open
Abstract
Background Lassa fever, killing thousands of people annually, is the most reported viral zoonotic disease in Nigeria. Recently, different rodent species carrying diverse lineages of the Lassa virus (LASV) in addition to a novel Mobala-like genetic sequence were detected within the country. Here, screening 906 small mammal specimens from 11 localities for IgG antibodies and incorporating previous PCR detection data involving the same populations, we further describe arenavirus prevalence across Nigeria in relation to host species and geographical location. Methods Small mammals were trapped during the period 2011–2015 according to geographical location (endemic and non-endemic zones for Lassa fever), season (rainy and dry seasons between 2011 and 2012 for certain localities) and habitat (indoors, peridomestic settings and sylvatic vegetation). Identification of animal specimens from genera such as Mastomys and Mus (Nannomys) was assisted by DNA sequencing. Small mammals were tested for LASV IgG antibody using an indirect immunofluorescence assay (IFA). Results Small mammals were infected in both the endemic and non-endemic zones for Lassa fever, with a wider range of species IgG-positive (n = 8) than those which had been previously detected to be PCR-positive (n = 3). IgG-positive species, according to number of infected individuals, were Mastomys natalensis (n = 40), Mastomys erythroleucus (n = 15), Praomys daltoni (n = 6), Mus baoulei (n = 5), Rattus rattus (n = 2), Crocidura spp. (n = 2), Mus minutoides (n = 1) and Praomys misonnei (n = 1). Multimammate mice (Mastomys natalensis and M. erythroleucus) were the most ubiquitously infected, with animals testing positive by either PCR or IgG in 7 out of the 11 localities sampled. IgG prevalence in M. natalensis ranged from 1% in Abagboro, 17–36 % in Eguare Egoro, Ekpoma and Ngel Nyaki, up to 52 % in Mayo Ranewo. Prevalence according to locality, season and age was not, however, statistically significant for M. natalensis in Eguare Egoro and Ekpoma, localities that were sampled longitudinally. Conclusions Overall, our study demonstrates that arenavirus occurrence is probably more widely distributed geographically and in extent of host taxa than is currently realized. This expanded scope should be taken into consideration in Lassa fever control efforts. Further sampling should also be carried out to isolate and characterize potential arenaviruses present in small mammal populations we found to be seropositive.
Collapse
|
20
|
Fernandes J, Guterres A, de Oliveira RC, Chamberlain J, Lewandowski K, Teixeira BR, Coelho TA, Crisóstomo CF, Bonvicino CR, D'Andrea PS, Hewson R, de Lemos ERS. Xapuri virus, a novel mammarenavirus: natural reassortment and increased diversity between New World viruses. Emerg Microbes Infect 2018; 7:120. [PMID: 29959319 PMCID: PMC6026159 DOI: 10.1038/s41426-018-0119-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/21/2018] [Accepted: 05/27/2018] [Indexed: 12/28/2022]
Abstract
Mammarenavirus RNA was detected in Musser’s bristly mouse (Neacomys musseri) from the Amazon region, and this detection indicated that rodents were infected with a novel mammarenavirus, with the proposed name Xapuri virus (XAPV), which is phylogenetically related to New World Clade B and Clade C viruses. XAPV may represent the first natural reassortment of the Arenaviridae family and a new unrecognized clade within the Tacaribe serocomplex group.
Collapse
Affiliation(s)
- Jorlan Fernandes
- Laboratory of Hantaviruses and Rickettsiosis, Oswaldo Cruz Foundation, Oswaldo Cruz Institute, Rio de Janeiro - RJ, 21040-360, Brazil.
| | - Alexandro Guterres
- Laboratory of Hantaviruses and Rickettsiosis, Oswaldo Cruz Foundation, Oswaldo Cruz Institute, Rio de Janeiro - RJ, 21040-360, Brazil
| | - Renata Carvalho de Oliveira
- Laboratory of Hantaviruses and Rickettsiosis, Oswaldo Cruz Foundation, Oswaldo Cruz Institute, Rio de Janeiro - RJ, 21040-360, Brazil
| | - John Chamberlain
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, SP4 0JG, UK
| | - Kuiama Lewandowski
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, SP4 0JG, UK
| | - Bernardo Rodrigues Teixeira
- Laboratory of Biology and Parasitology of Wild Mammals Reservoirs, Oswaldo Cruz Foundation, Oswaldo Cruz Institute, Rio de Janeiro - RJ, 21040-360, Brazil
| | - Thayssa Alves Coelho
- Laboratory of Hantaviruses and Rickettsiosis, Oswaldo Cruz Foundation, Oswaldo Cruz Institute, Rio de Janeiro - RJ, 21040-360, Brazil
| | - Charle Ferreira Crisóstomo
- Laboratory of Biology and Parasitology of Wild Mammals Reservoirs, Oswaldo Cruz Foundation, Oswaldo Cruz Institute, Rio de Janeiro - RJ, 21040-360, Brazil.,Federal Institute of Acre, Rio Branco - AC, 69900-640, Brazil.,Postgraduate Program in Biodiversity and Health, Oswaldo Cruz Foundation, Oswaldo Cruz Institute, Rio de Janeiro - RJ, 21040-360, Brazil
| | - Cibele Rodrigues Bonvicino
- Laboratory of Biology and Parasitology of Wild Mammals Reservoirs, Oswaldo Cruz Foundation, Oswaldo Cruz Institute, Rio de Janeiro - RJ, 21040-360, Brazil.,Nacional Cancer Institute, Rio de Janeio - RJ, 20230-130, Brazil
| | - Paulo Sérgio D'Andrea
- Laboratory of Biology and Parasitology of Wild Mammals Reservoirs, Oswaldo Cruz Foundation, Oswaldo Cruz Institute, Rio de Janeiro - RJ, 21040-360, Brazil.,Postgraduate Program in Biodiversity and Health, Oswaldo Cruz Foundation, Oswaldo Cruz Institute, Rio de Janeiro - RJ, 21040-360, Brazil
| | - Roger Hewson
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, SP4 0JG, UK
| | - Elba Regina Sampaio de Lemos
- Laboratory of Hantaviruses and Rickettsiosis, Oswaldo Cruz Foundation, Oswaldo Cruz Institute, Rio de Janeiro - RJ, 21040-360, Brazil.
| |
Collapse
|
21
|
Arenaviral Nucleoproteins Suppress PACT-Induced Augmentation of RIG-I Function To Inhibit Type I Interferon Production. J Virol 2018; 92:JVI.00482-18. [PMID: 29669840 DOI: 10.1128/jvi.00482-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 04/13/2018] [Indexed: 12/28/2022] Open
Abstract
RIG-I is a major cytoplasmic sensor of viral pathogen-associated molecular pattern (PAMP) RNA and induces type I interferon (IFN) production upon viral infection. A double-stranded RNA (dsRNA)-binding protein, PACT, plays an important role in potentiating RIG-I function. We have shown previously that arenaviral nucleoproteins (NPs) suppress type I IFN production via their RNase activity to degrade PAMP RNA. We report here that NPs of arenaviruses block the PACT-induced enhancement of RIG-I function to mediate type I IFN production and that this inhibition is dependent on the RNase function of NPs, which is different from that of a known mechanism of other viral proteins to abolish the interaction between PACT and RIG-I. To understand the biological roles of PACT and RIG-I in authentic arenavirus infection, we analyze growth kinetics of recombinant Pichinde virus (PICV), a prototypical arenavirus, in RIG-I knockout (KO) and PACT KO mouse embryonic fibroblast (MEF) cells. Wild-type (WT) PICV grew at higher titers in both KO MEF lines than in normal MEFs, suggesting the important roles of these cellular proteins in restricting virus replication. PICV carrying the NP RNase catalytically inactive mutation could not grow in normal MEFs but could replicate to some extent in both KO MEF lines. The level of virus growth was inversely correlated with the amount of type I IFNs produced. These results suggest that PACT plays an important role in potentiating RIG-I function to produce type I IFNs in order to restrict arenavirus replication and that viral NP RNase activity is essential for optimal viral replication by suppressing PACT-induced RIG-I activation.IMPORTANCE We report here a new role of the nucleoproteins of arenaviruses that can block type I IFN production via their specific inhibition of the cellular protein sensors of virus infection (RIG-I and PACT). Our results suggest that PACT plays an important role in potentiating RIG-I function to produce type I IFNs in order to restrict arenavirus replication. This new knowledge can be exploited for the development of novel antiviral treatments and/or vaccines against some arenaviruses that can cause severe and lethal hemorrhagic fever diseases in humans.
Collapse
|
22
|
Wessling-Resnick M. Crossing the Iron Gate: Why and How Transferrin Receptors Mediate Viral Entry. Annu Rev Nutr 2018; 38:431-458. [PMID: 29852086 DOI: 10.1146/annurev-nutr-082117-051749] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Because both the host and pathogen require iron, the innate immune response carefully orchestrates control over iron metabolism to limit its availability during times of infection. Nutritional iron deficiency can impair host immunity, while iron overload can cause oxidative stress to propagate harmful viral mutations. An emerging enigma is that many viruses use the primary gatekeeper of iron metabolism, the transferrin receptor, as a means to enter cells. Why and how this iron gate is a viral target for infection are the focus of this review.
Collapse
Affiliation(s)
- Marianne Wessling-Resnick
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA;
| |
Collapse
|
23
|
Fernandes J, de Oliveira RC, Guterres A, Barreto-Vieira DF, Terças ACP, Teixeira BR, da Silva MAN, Caldas GC, de Oliveira Coelho JMC, Barth OM, D'Andrea PS, Bonvicino CR, de Lemos ERS. Detection of Latino virus (Arenaviridae: Mammarenavirus) naturally infecting Calomys callidus. Acta Trop 2018; 179:17-24. [PMID: 29217383 DOI: 10.1016/j.actatropica.2017.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/27/2017] [Accepted: 12/02/2017] [Indexed: 11/18/2022]
Abstract
Mammarenavirus species are associated with a specific rodent host species, although an increasing number of virus has been associated to more than one host, suggesting that co-evolution is less robust than initially thought. There are few eco-epidemiological studies of South America mammarenaviruses in non-endemic areas of Arenavirus Hemorrhagic Fever, affecting specially our current knowledge about animal reservoirs and virus range and host-virus relations. In Brazil, seven arenavirus species were described in seven different rodent species. Here in we describe a new rodent reservoir species in Brazil related to the previously described Latino mammarenavirus (LATV) MARU strain. Samples of 148 rodents from Mato Grosso state, Brazil were analyzed. Amplification of the glycoprotein precursor gene (GPC) was observed in six Calomys callidus rodents. According to phylogenetic inferences, is observed a well-supported monophyletic clade of LATV from C. callidus and other Clade C mammarenavirus. In addition, the phylogenetic relations of both genes showed a close relation between LATV MARU and Capão Seco strains, two distinct lineages. Additionally, the results obtained in this study point out to a change of scenario and in previously stabilized patterns in the dynamics of South American mammarenaviruses, showing that with more studies in AHF non-endemic or silent areas, more potential hosts for this virus will be discovered.
Collapse
Affiliation(s)
- Jorlan Fernandes
- Laboratório de Hantaviroses e Rickettsioses, Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Rio de Janeiro, CEP 21040-360 RJ, Brazil.
| | - Renata Carvalho de Oliveira
- Laboratório de Hantaviroses e Rickettsioses, Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Rio de Janeiro, CEP 21040-360 RJ, Brazil
| | - Alexandro Guterres
- Laboratório de Hantaviroses e Rickettsioses, Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Rio de Janeiro, CEP 21040-360 RJ, Brazil
| | - Débora Ferreira Barreto-Vieira
- Laboratório de Morfologia e Morfogênese Viral, Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Rio de Janeiro, CEP 21040-360 RJ, Brazil
| | | | - Bernardo Rodrigues Teixeira
- Laboratório de Biologia e Parasitologia de Mamíferos Silvestres Reservatórios, Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Rio de Janeiro, CEP 21040-360 RJ, Brazil
| | - Marcos Alexandre Nunes da Silva
- Laboratório de Morfologia e Morfogênese Viral, Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Rio de Janeiro, CEP 21040-360 RJ, Brazil
| | - Gabriela Cardoso Caldas
- Laboratório de Morfologia e Morfogênese Viral, Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Rio de Janeiro, CEP 21040-360 RJ, Brazil
| | | | - Ortrud Monika Barth
- Laboratório de Morfologia e Morfogênese Viral, Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Rio de Janeiro, CEP 21040-360 RJ, Brazil
| | - Paulo Sergio D'Andrea
- Laboratório de Biologia e Parasitologia de Mamíferos Silvestres Reservatórios, Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Rio de Janeiro, CEP 21040-360 RJ, Brazil
| | - Cibele Rodrigues Bonvicino
- Laboratório de Biologia e Parasitologia de Mamíferos Silvestres Reservatórios, Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Rio de Janeiro, CEP 21040-360 RJ, Brazil; Instituto Nacional do Câncer - INCA, Rio de Janeiro, CEP 20230-092 RJ, Brazil
| | - Elba Regina Sampaio de Lemos
- Laboratório de Hantaviroses e Rickettsioses, Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Rio de Janeiro, CEP 21040-360 RJ, Brazil.
| |
Collapse
|
24
|
Aqrawi T, Stöhr AC, Knauf-Witzens T, Krengel A, Heckers KO, Marschang RE. Nachweis und Charakterisierung von Schlangenarenaviren bei lebenden Boas und Pythons in einem deutschen Zoo. TIERAERZTLICHE PRAXIS AUSGABE KLEINTIERE HEIMTIERE 2018; 43:239-47. [DOI: 10.15654/tpk-140743] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 11/10/2014] [Indexed: 01/26/2023]
Abstract
ZusammenfassungNeue schlangenspezifische Arenaviren wurden vor kurzem bei Boas und Pythons mit der Einschlusskörperchenkrankheit (inclusion body disease, IBD) beschrieben. Ziel der Studie war, arenavirale RNA bei lebenden Schlangen nachzuweisen und festzustellen, ob Virus immer mit IBD bei den infizierten Schlangen assoziiert war. Proben für den Virusnachweis bei lebenden Tieren wurden verglichen. Ferner erfolgte ein Vergleich der nachgewiesenen Viren zur Feststellung ihrer genetischen Variabilität.Von 28 Boas und Pythons wurden ösophageale Tupfer sowie Vollblut mittels RT-PCR auf arenavirale RNA und Blutausstriche auf Einschlüsse untersucht. Innere Organe von Tieren, die während der Studie starben oder euthanisiert wurden, unterlagen ebenfalls einer Untersuchung auf arenavirale RNA sowie auf Einschlusskörperchen. Alle PCR-Produkte wurden sequenziert und genomische Sequenzen phylogenetisch analysiert.Arenaviren ließen sich bei neun lebenden Tieren sowie bei zwei Tieren nach der Sektion nachweisen. Die fünf detektierten neuen Arenaviren wurden “Boa Arenavirus Deutschland (Boa Av DE) Nummer 1–4” bzw. „Python Av DE1” genannte. Die Sequenzanalyse zeigte deutliche Ähnlichkeiten mit dem entsprechenden Abschnitt bei neu identifizierten Schlangenarenaviren.Ösophageale Tupfer wie auch Vollblut können zum Nachweis von Arenaviren bei Schlangen eingesetzt werden. In den meisten Fällen korrelierte die Arenavirusinfektion mit dem Nachweis von Einschlüssen in den Organen. Es gab Hinweise darauf, dass in Einzelfällen arenavirusinfizierte Tiere wieder virusfrei werden können, ohne an IBD zu erkranken. Dies ist der erste Nachweis von Arenaviren bei lebenden Schlangen.Der Arenavirusnachweis bei lebenden Schlangen hat Bedeutung für die Diagnose, Prävention und Quarantäne. Die Ergebnisse dieser Studie unterstützen die Theorie, dass Arenaviren IBD verursachen, zeigen aber, dass infizierte Tiere Arenavirusinfektionen eventuell überstehen, ohne IBD zu entwickeln.
Collapse
|
25
|
Hedrick SM. Understanding Immunity through the Lens of Disease Ecology. Trends Immunol 2017; 38:888-903. [PMID: 28882454 DOI: 10.1016/j.it.2017.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 07/26/2017] [Accepted: 08/01/2017] [Indexed: 10/25/2022]
Abstract
As we describe the immune system in ever more exquisite detail, we might find that no matter how successful, this approach will not be sufficient to understand the spread of infectious agents, their susceptibility to vaccine therapy, and human disease resistance. Compared with the strict reductionism practiced as a means of characterizing most biological processes, I propose that the progression and outcome of disease-causing host-parasite interactions will be more clearly understood through a focus on disease ecology.
Collapse
Affiliation(s)
- Stephen M Hedrick
- Departments of Molecular Biology and Cellular and Molecular Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
26
|
Abstract
Hemorrhagic fevers caused by viruses were identified in the late 1950s in South America. These viruses have existed in their hosts, the New World rodents, for millions of years. Their emergence as infectious agents in humans coincided with changes in the environment and farming practices that caused explosions in their host rodent populations. Zoonosis into humans likely occurs because the pathogenic New World arenaviruses use human transferrin receptor 1 to enter cells. The mortality rate after infection with these viruses is high, but the mechanism by which disease is induced is still not clear. Possibilities include direct effects of cellular infection or the induction of high levels of cytokines by infected sentinel cells of the immune system, leading to endothelia and thrombocyte dysfunction and neurological disease. Here we provide a review of the ecology and molecular and cellular biology of New World arenaviruses, as well as a discussion of the current animal models of infection. The development of animal models, coupled with an improved understanding of the infection pathway and host response, should lead to the discovery of new drugs for treating infections.
Collapse
Affiliation(s)
- Nicolás Sarute
- Department of Microbiology and Immunology, University of Illinois College of Medicine at Chicago, Chicago, Illinois 60612; ,
| | - Susan R Ross
- Department of Microbiology and Immunology, University of Illinois College of Medicine at Chicago, Chicago, Illinois 60612; ,
| |
Collapse
|
27
|
[First evidence of lymphocytic choriomeningitis virus (Arenavirus) infection in Mus musculus rodents captured in the urban area of the municipality of Sincelejo, Sucre, Colombia]. BIOMEDICA 2017; 37:75-85. [PMID: 28527269 DOI: 10.7705/biomedica.v37i2.3226] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 05/31/2016] [Indexed: 11/21/2022]
Abstract
INTRODUCTION The lymphocytic choriomeningitis virus is an Old World arenavirus that infects Mus musculus, and can cause congenital hydrocephalus, chorioretinitis and multisystemic failure in transplant human recipients. Although the disease has not been clinically diagnosed in Colombia yet, there have been reports of infection with the Pichindé virus in rodents from Cauca and Valle del Cauca departments, and with the Guanarito virus in rodents from Córdoba department. OBJECTIVE To identify the lymphocytic choriomeningitis virus from Mus musculus captured in the municipality of Sincelejo. MATERIALS AND METHODS We evaluated 80 samples of plasma by ELISA using antigen from lymphocytic choriomeningitis virus. Additionally, a nested RT-PCR was performed to seropositive and seronegative samples for the S-segment. RESULTS We found a 10% seroprevalence (8/80) and the viral genome was detected in 16 brain samples; the alignment (BLAST) and the phylogenetic analysis (MrBayes, version 3.2.2) confirmed the presence of the lymphocytic choriomeningitis virus. CONCLUSION The results indicated that human infection with the lymphocytic choriomeningitis virus in humans could occur in the urban area of Sincelejo, although no cases have been reported so far.
Collapse
|
28
|
Kalkavan H, Sharma P, Kasper S, Helfrich I, Pandyra AA, Gassa A, Virchow I, Flatz L, Brandenburg T, Namineni S, Heikenwalder M, Höchst B, Knolle PA, Wollmann G, von Laer D, Drexler I, Rathbun J, Cannon PM, Scheu S, Bauer J, Chauhan J, Häussinger D, Willimsky G, Löhning M, Schadendorf D, Brandau S, Schuler M, Lang PA, Lang KS. Spatiotemporally restricted arenavirus replication induces immune surveillance and type I interferon-dependent tumour regression. Nat Commun 2017; 8:14447. [PMID: 28248314 PMCID: PMC5337983 DOI: 10.1038/ncomms14447] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 12/30/2016] [Indexed: 12/15/2022] Open
Abstract
Immune-mediated effector molecules can limit cancer growth, but lack of sustained immune activation in the tumour microenvironment restricts antitumour immunity. New therapeutic approaches that induce a strong and prolonged immune activation would represent a major immunotherapeutic advance. Here we show that the arenaviruses lymphocytic choriomeningitis virus (LCMV) and the clinically used Junin virus vaccine (Candid#1) preferentially replicate in tumour cells in a variety of murine and human cancer models. Viral replication leads to prolonged local immune activation, rapid regression of localized and metastatic cancers, and long-term disease control. Mechanistically, LCMV induces antitumour immunity, which depends on the recruitment of interferon-producing Ly6C+ monocytes and additionally enhances tumour-specific CD8+ T cells. In comparison with other clinically evaluated oncolytic viruses and to PD-1 blockade, LCMV treatment shows promising antitumoural benefits. In conclusion, therapeutically administered arenavirus replicates in cancer cells and induces tumour regression by enhancing local immune responses.
Collapse
Affiliation(s)
- Halime Kalkavan
- Institute of Immunology, Medical Faculty, University Duisburg-Essen, 45122 Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - Piyush Sharma
- Institute of Immunology, Medical Faculty, University Duisburg-Essen, 45122 Essen, Germany
| | - Stefan Kasper
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - Iris Helfrich
- Department of Dermatology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - Aleksandra A. Pandyra
- Institute of Immunology, Medical Faculty, University Duisburg-Essen, 45122 Essen, Germany
| | - Asmae Gassa
- Institute of Immunology, Medical Faculty, University Duisburg-Essen, 45122 Essen, Germany
- Department of Cardiothoracic Surgery, Cologne University Heart Center, Kerpener Strasse 62, 50937 Cologne, Germany
| | - Isabel Virchow
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - Lukas Flatz
- Department of Dermatology/Allergology, Cantonal Hospital, Rorschacher Strasse 95, St. Gallen 9007, Switzerland
| | - Tim Brandenburg
- Institute of Immunology, Medical Faculty, University Duisburg-Essen, 45122 Essen, Germany
| | - Sukumar Namineni
- Department of Virology, Technical University of Munich, Schneckenburgstrasse 8, 81675 Munich, Germany
| | - Mathias Heikenwalder
- Department of Virology, Technical University of Munich, Schneckenburgstrasse 8, 81675 Munich, Germany
| | - Bastian Höchst
- Institute of Molecular Immunology/Experimental Oncology, München Klinikum rechts der Isar, Technical University Munich, 81675 Munich, Germany
| | - Percy A. Knolle
- Institute of Molecular Immunology/Experimental Oncology, München Klinikum rechts der Isar, Technical University Munich, 81675 Munich, Germany
| | - Guido Wollmann
- Division for Virology, Medical University Innsbruck, Peter-Mayr-Strasse 4b, 6020 Innsbruck, Austria
| | - Dorothee von Laer
- Division for Virology, Medical University Innsbruck, Peter-Mayr-Strasse 4b, 6020 Innsbruck, Austria
| | - Ingo Drexler
- Institute of Virology, Düsseldorf University Hospital, Heinrich Heine University, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Jessica Rathbun
- Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, 90033 California, USA
| | - Paula M. Cannon
- Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, 90033 California, USA
| | - Stefanie Scheu
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Jens Bauer
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Jagat Chauhan
- Ludwig Institute for Cancer Research, University of Oxford, Old Road Campus, Research Building, Old Road Campus, Headington, Oxford OX3 7DQ, UK
| | - Dieter Häussinger
- Department of Gastroenterology, Hepatology and Infectious Diseases, University of Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Gerald Willimsky
- Institute of Immunology (Charité - University Medicine Berlin), 3125 Berlin, Germany
- German Cancer Research Center (DKFZ), 13125 Heidelberg, Germany
| | - Max Löhning
- Department of Rheumatology and Clinical Immunology, Charité—University Medicine Berlin and German Rheumatism Research Center (DRFZ), Charitéplatz 1, D-10117 Berlin, Germany
| | - Dirk Schadendorf
- Department of Dermatology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45122 Essen, Germany
| | - Sven Brandau
- Department of Otorhinolaryngology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | - Martin Schuler
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45122 Essen, Germany
| | - Philipp A. Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Universitätsstrasse 1, D-40225 Düsseldorf, Germany
| | - Karl S. Lang
- Institute of Immunology, Medical Faculty, University Duisburg-Essen, 45122 Essen, Germany
| |
Collapse
|
29
|
Guterres A, de Oliveira RC, Fernandes J, de Lemos ERS, Schrago CG. New bunya-like viruses: Highlighting their relations. INFECTION GENETICS AND EVOLUTION 2017; 49:164-173. [PMID: 28111322 DOI: 10.1016/j.meegid.2017.01.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/16/2017] [Accepted: 01/18/2017] [Indexed: 11/18/2022]
Abstract
The standard virus classification scheme for arenaviruses and bunyaviruses shifted dramatically when several groups reported the detection and isolation of divergent groups of viruses in a variety of insect collections. Although these viral families can differ in terms of morphology, structure and genetics, recent findings indicate these viruses may have a shared evolutionary origin. To determine the phylogenetic relations among these families, we inferred phylogenetic trees using three methods. The Maximum Likelihood and Bayesian trees were rooted as suggested by the (molecular clock-rooted) BEAST tree. Our results highlight a noteworthy relation among these viral supergroups of different genome organizations. Our study suggests that the best scenario is the existence of at least three monophyletic supergroups, all of them well supported. The recent data indicate that these viruses are evolutionarily and genetically interconnected. While these supergroups appear to be closely related in our phylogenetic analysis, other viruses should be investigated in future research. In sum, our results also provide insights into the classification scheme, thereby providing a new perspective about the fundamental questions of family origins, diversity and genome evolution.
Collapse
Affiliation(s)
- Alexandro Guterres
- Laboratório de Hantaviroses e Rickettsioses, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil; Departamento de Genética, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Renata Carvalho de Oliveira
- Laboratório de Hantaviroses e Rickettsioses, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Jorlan Fernandes
- Laboratório de Hantaviroses e Rickettsioses, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Elba Regina Sampaio de Lemos
- Laboratório de Hantaviroses e Rickettsioses, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Carlos Guerra Schrago
- Departamento de Genética, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
30
|
Characterization of the Glycoprotein Stable Signal Peptide in Mediating Pichinde Virus Replication and Virulence. J Virol 2016; 90:10390-10397. [PMID: 27630230 DOI: 10.1128/jvi.01154-16] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/02/2016] [Indexed: 11/20/2022] Open
Abstract
Arenaviruses can cause lethal hemorrhagic fevers in humans with few preventative and therapeutic measures. The arenaviral glycoprotein stable signal peptide (SSP) is unique among signal peptides in that it is an integral component of the mature glycoprotein complex (GPC) and plays important roles not only in GPC expression and processing but also in the membrane fusion process during viral entry. Using the Pichinde virus (PICV) reverse genetics system, we analyzed the effects of alanine substitutions at many conserved residues within the SSP on viral replication in cell culture and in a guinea pig infection model. Our data showed that the K33A, F49A, and C57A mutations abolished GPC-mediated cell entry and therefore could not allow for the generation of viable recombinant viruses, demonstrating that these residues are essential for the PICV life cycle. The G2A mutation caused a marked reduction of cell entry at the membrane fusion step, and while this mutant virus was viable, it was significantly attenuated in vitro and in vivo The N20A mutation also reduced membrane fusion activity and viral virulence in guinea pigs, but it did not significantly affect cell entry or viral growth in cell culture. Two other mutations (N37A and R55A) did not affect membrane fusion or viral growth in vitro but significantly reduced viral virulence in vivo Taken together, our data suggest that the GPC SSP plays an essential role in mediating viral entry and also contributes to viral virulence in vivo IMPORTANCE: Several arenaviruses, such as Lassa fever virus, can cause severe and lethal hemorrhagic fever diseases with high mortality and morbidity, and no FDA-approved vaccines or therapies are currently available. Viral entry into cells is mediated by arenavirus GPC that consists of an SSP, the receptor-binding GP1, and transmembrane GP2 protein subunits. Using a reverse genetics system of a prototypic arenavirus, Pichinde virus (PICV), we have shown for the first time in the context of virus infections of cell culture and of guinea pigs that the SSP plays an essential role in mediating the membrane fusion step as well as in other yet-to-be-determined processes during viral infection. Our study provides important insights into the biological roles of GPC SSP and implicates it as a good target for the development of antivirals against deadly human arenavirus pathogens.
Collapse
|
31
|
In vitro isolation and molecular identification of reptarenavirus in Malaysia. Virus Genes 2016; 52:640-50. [PMID: 27142080 DOI: 10.1007/s11262-016-1345-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/21/2016] [Indexed: 02/02/2023]
Abstract
Boid inclusion body disease (BIBD) is a viral disease of boids caused by reptarenavirus. In this study, tissue from naturally infected boid snakes were homogenized and propagated in African Monkey kidney (Vero) and rat embryonic fibroblast (REF) cells. Virus replication was determined by the presence of cytopathic effect, while viral morphology was observed using transmission electron microscopy. Viral RNA was amplified using RT-PCR with primers specific for the L-segment of reptarenavirus; similarly, quantification of viral replication was done using qPCR at 24-144 h postinfection. Viral cytopathology was characterized by cell rounding and detachment in both Vero and REF cells. The viral morphology showed round-to-pleomorphic particles ranging from 105 to 150 nm which had sand-like granules. Sanger sequencing identified four closely associated reptarenavirus species from 15 (37.5 %) of the total samples tested, and these were named as follows: reptarenavirus UPM-MY 01, 02, 03, and 04. These isolates were phylogenetically closely related to the University Helsinki virus (UHV), Boa Arenavirus NL (ROUTV; BAV), and unidentified reptarenavirus L20 (URAV-L20). Comparison of deduced amino acid sequences further confirmed identities to L-protein of UHV, L-polymerase of BAV and RNA-dependent RNA polymerase of URAV-L20. Viral replication in Vero cells increased steadily from 24 to 72 h and peaked at 144 h. This is the first study in South East Asia to isolate and characterize reptarenavirus in boid snakes with BIBD.
Collapse
|
32
|
Witkowski PT, Kallies R, Hoveka J, Auste B, Ithete NL, Šoltys K, Szemes T, Drosten C, Preiser W, Klempa B, Mfune JKE, Kruger DH. Novel Arenavirus Isolates from Namaqua Rock Mice, Namibia, Southern Africa. Emerg Infect Dis 2016; 21:1213-6. [PMID: 26079174 PMCID: PMC4480381 DOI: 10.3201/eid2107.141341] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Arenaviruses are feared as agents that cause viral hemorrhagic fevers. We report the identification, isolation, and genetic characterization of 2 novel arenaviruses from Namaqua rock mice in Namibia. These findings extend knowledge of the distribution and diversity of arenaviruses in Africa.
Collapse
|
33
|
Charley PA, Wilusz J. Standing your ground to exoribonucleases: Function of Flavivirus long non-coding RNAs. Virus Res 2016; 212:70-7. [PMID: 26368052 PMCID: PMC4744573 DOI: 10.1016/j.virusres.2015.09.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/04/2015] [Accepted: 09/10/2015] [Indexed: 01/18/2023]
Abstract
Members of the Flaviviridae (e.g., Dengue virus, West Nile virus, and Hepatitis C virus) contain a positive-sense RNA genome that encodes a large polyprotein. It is now also clear most if not all of these viruses also produce an abundant subgenomic long non-coding RNA. These non-coding RNAs, which are called subgenomic flavivirus RNAs (sfRNAs) or Xrn1-resistant RNAs (xrRNAs), are stable decay intermediates generated from the viral genomic RNA through the stalling of the cellular exoribonuclease Xrn1 at highly structured regions. Several functions of these flavivirus long non-coding RNAs have been revealed in recent years. The generation of these sfRNAs/xrRNAs from viral transcripts results in the repression of Xrn1 and the dysregulation of cellular mRNA stability. The abundant sfRNAs also serve directly as a decoy for important cellular protein regulators of the interferon and RNA interference antiviral pathways. Thus the generation of long non-coding RNAs from flaviviruses, hepaciviruses and pestiviruses likely disrupts aspects of innate immunity and may directly contribute to viral replication, cytopathology and pathogenesis.
Collapse
Affiliation(s)
- Phillida A Charley
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Jeffrey Wilusz
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
34
|
Abstract
The family Arenaviridae currently comprises over 20 viral species, each of them associated with a main rodent species as the natural reservoir and in one case possibly phyllostomid bats. Moreover, recent findings have documented a divergent group of arenaviruses in captive alethinophidian snakes. Human infections occur through mucosal exposure to aerosols or by direct contact of abraded skin with infectious materials. Arenaviruses merit interest both as highly tractable experimental model systems to study acute and persistent infections and as clinically important human pathogens including Lassa (LASV) and Junin (JUNV) viruses, the causative agents of Lassa and Argentine hemorrhagic fevers (AHFs), respectively, for which there are no FDA-licensed vaccines, and current therapy is limited to an off-label use of ribavirin (Rib) that has significant limitations. Arenaviruses are enveloped viruses with a bi-segmented negative strand (NS) RNA genome. Each genome segment, L (ca 7.3 kb) and S (ca 3.5 kb), uses an ambisense coding strategy to direct the synthesis of two polypeptides in opposite orientation, separated by a noncoding intergenic region (IGR). The S genomic RNA encodes the virus nucleoprotein (NP) and the precursor (GPC) of the virus surface glycoprotein that mediates virus receptor recognition and cell entry via endocytosis. The L genome RNA encodes the viral RNA-dependent RNA polymerase (RdRp, or L polymerase) and the small (ca 11 kDa) RING finger protein Z that has functions of a bona fide matrix protein including directing virus budding. Arenaviruses were thought to be relatively stable genetically with intra- and interspecies amino acid sequence identities of 90-95 % and 44-63 %, respectively. However, recent evidence has documented extensive arenavirus genetic variability in the field. Moreover, dramatic phenotypic differences have been documented among closely related LCMV isolates. These data provide strong evidence of viral quasispecies involvement in arenavirus adaptability and pathogenesis. Here, we will review several aspects of the molecular biology of arenaviruses, phylogeny and evolution, and quasispecies dynamics of arenavirus populations for a better understanding of arenavirus pathogenesis, as well as for the development of novel antiviral strategies to combat arenavirus infections.
Collapse
Affiliation(s)
- Esteban Domingo
- Campus de Cantoblanco, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Peter Schuster
- The Santa Fe Institute, Santa Fe, NM, USA and Institut f. Theoretische Chemie, Universität Wien, Vienna, Austria
| |
Collapse
|
35
|
Tao XY, Guo ZY, Li H, Jiao WT, Shen XX, Zhu WY, Rayner S, Tang Q. Rabies Cases in the West of China Have Two Distinct Origins. PLoS Negl Trop Dis 2015; 9:e0004140. [PMID: 26484668 PMCID: PMC4618851 DOI: 10.1371/journal.pntd.0004140] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/15/2015] [Indexed: 12/03/2022] Open
Abstract
In China, rabies remains an ongoing threat to public health. Although control efforts have been effective in reducing the number of annual cases, the virus continues to spread into new areas. Tibet, Qinghai, Gansu and Ningxia in western China have, until recently, reported only a handful of events. However, since 2011, there have been increasing numbers of cases recorded in these areas. In this study, we report the collection and analysis of samples collected from these regions. We find that cases originate from two different sources. Strains collected from Gansu and Ningxia are closely related to the primary lineage associated with the current epizootic, whereas those from Tibet and Qinghai are related to the Arctic-like-2 lineage that is most commonly associated with wildlife cases in China. Thus, it appears that while the epizootic is beginning to encroach into Gansu and Ningxia, Tibet and Qinghai a significant number of rabies cases originate from wildlife. Overall, the number of annual cases of human rabies reported in China has been decreasing since 2007. However, some Western provinces, where few human cases have been reported in recent years, are beginning to see increasing incidence of rabies. Specifically, Ningxia, Qinghai and Gansu began to report human cases respectively from 2011, 2012 and 2013, while Tibet had its first laboratory confirmed dog rabies case in 2012. Consequently, as part of the national rabies surveillance program, we collected specimens from biting dogs or human saliva from suspected rabies cases in these areas, and after sequencing positive samples, performed a phylogenetic analysis based on the nucleoprotein gene complete sequences. Our results indicate that while Ningxia and Gansu rabies strains are very close to the lineage associated with most cases in mainland China, Tibet and Qinghai strains belong to the global Arctic-like-2 clade, which is typically associated with wild life in China and neighboring countries. Thus, it appears that, rabies reemergence in the west of China has two distinct origins, and Tibet and Qinghai rabies cases were isolated events rather than an indication of the ongoing epizootic in China.
Collapse
Affiliation(s)
- Xiao-Yan Tao
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, China
| | - Zhen-Yang Guo
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hao Li
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, China
| | - Wen-Tao Jiao
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xin-Xin Shen
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, China
| | - Wu-Yang Zhu
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, China
| | - Simon Rayner
- Department of Medical Genetics, University of Oslo, Oslo, Norway
- Key Laboratory of Agricultural and Environmental Microbiology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
- * E-mail: (SR); (QT)
| | - Qing Tang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, China
- * E-mail: (SR); (QT)
| |
Collapse
|
36
|
Widespread recombination, reassortment, and transmission of unbalanced compound viral genotypes in natural arenavirus infections. PLoS Pathog 2015; 11:e1004900. [PMID: 25993603 PMCID: PMC4438980 DOI: 10.1371/journal.ppat.1004900] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/20/2015] [Indexed: 12/19/2022] Open
Abstract
Arenaviruses are one of the largest families of human hemorrhagic fever viruses and are known to infect both mammals and snakes. Arenaviruses package a large (L) and small (S) genome segment in their virions. For segmented RNA viruses like these, novel genotypes can be generated through mutation, recombination, and reassortment. Although it is believed that an ancient recombination event led to the emergence of a new lineage of mammalian arenaviruses, neither recombination nor reassortment has been definitively documented in natural arenavirus infections. Here, we used metagenomic sequencing to survey the viral diversity present in captive arenavirus-infected snakes. From 48 infected animals, we determined the complete or near complete sequence of 210 genome segments that grouped into 23 L and 11 S genotypes. The majority of snakes were multiply infected, with up to 4 distinct S and 11 distinct L segment genotypes in individual animals. This S/L imbalance was typical: in all cases intrahost L segment genotypes outnumbered S genotypes, and a particular S segment genotype dominated in individual animals and at a population level. We corroborated sequencing results by qRT-PCR and virus isolation, and isolates replicated as ensembles in culture. Numerous instances of recombination and reassortment were detected, including recombinant segments with unusual organizations featuring 2 intergenic regions and superfluous content, which were capable of stable replication and transmission despite their atypical structures. Overall, this represents intrahost diversity of an extent and form that goes well beyond what has been observed for arenaviruses or for viruses in general. This diversity can be plausibly attributed to the captive intermingling of sub-clinically infected wild-caught snakes. Thus, beyond providing a unique opportunity to study arenavirus evolution and adaptation, these findings allow the investigation of unintended anthropogenic impacts on viral ecology, diversity, and disease potential.
Collapse
|
37
|
Konstantoulas CJ, Indik S. C3H strain of mouse mammary tumour virus, like GR strain, infects human mammary epithelial cells, albeit less efficiently than murine mammary epithelial cells. J Gen Virol 2015; 96:650-662. [DOI: 10.1099/jgv.0.000006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Affiliation(s)
| | - Stanislav Indik
- Institute of Virology, University of Veterinary Medicine, Veterinaerplatz 1, 1210 Vienna, Austria
| |
Collapse
|
38
|
Xing J, Ly H, Liang Y. The Z proteins of pathogenic but not nonpathogenic arenaviruses inhibit RIG-I-like receptor-dependent interferon production. J Virol 2015; 89:2944-55. [PMID: 25552708 PMCID: PMC4325705 DOI: 10.1128/jvi.03349-14] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 12/22/2014] [Indexed: 01/22/2023] Open
Abstract
UNLABELLED Arenavirus pathogens cause a wide spectrum of diseases in humans ranging from central nervous system disease to lethal hemorrhagic fevers with few treatment options. The reason why some arenaviruses can cause severe human diseases while others cannot is unknown. We find that the Z proteins of all known pathogenic arenaviruses, lymphocytic choriomeningitis virus (LCMV) and Lassa, Junin, Machupo, Sabia, Guanarito, Chapare, Dandenong, and Lujo viruses, can inhibit retinoic acid-inducible gene 1 (RIG-i) and Melanoma Differentiation-Associated protein 5 (MDA5), in sharp contrast to those of 14 other nonpathogenic arenaviruses. Inhibition of the RIG-i-like receptors (RLRs) by pathogenic Z proteins is mediated by the protein-protein interactions of Z and RLRs, which lead to the disruption of the interactions between RLRs and mitochondrial antiviral signaling (MAVS). The Z-RLR interactive interfaces are located within the N-terminal domain (NTD) of the Z protein and the N-terminal CARD domains of RLRs. Swapping of the LCMV Z NTD into the nonpathogenic Pichinde virus (PICV) genome does not affect virus growth in Vero cells but significantly inhibits the type I interferon (IFN) responses and increases viral replication in human primary macrophages. In summary, our results show for the first time an innate immune-system-suppressive mechanism shared by the diverse pathogenic arenaviruses and thus shed important light on the pathogenic mechanism of human arenavirus pathogens. IMPORTANCE We show that all known human-pathogenic arenaviruses share an innate immune suppression mechanism that is based on viral Z protein-mediated RLR inhibition. Our report offers important insights into the potential mechanism of arenavirus pathogenesis, provides a convenient way to evaluate the pathogenic potential of known and/or emerging arenaviruses, and reveals a novel target for the development of broad-spectrum therapies to treat this group of diverse pathogens. More broadly, our report provides a better understanding of the mechanisms of viral immune suppression and host-pathogen interactions.
Collapse
Affiliation(s)
- Junji Xing
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Hinh Ly
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Yuying Liang
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| |
Collapse
|
39
|
Sayler KA, Barbet AF, Chamberlain C, Clapp WL, Alleman R, Loeb JC, Lednicky JA. Isolation of Tacaribe virus, a Caribbean arenavirus, from host-seeking Amblyomma americanum ticks in Florida. PLoS One 2014; 9:e115769. [PMID: 25536075 PMCID: PMC4275251 DOI: 10.1371/journal.pone.0115769] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 11/26/2014] [Indexed: 12/25/2022] Open
Abstract
Arenaviridae are a family of single stranded RNA viruses of mammals and boid snakes. Twenty-nine distinct mammalian arenaviruses have been identified, many of which cause severe hemorrhagic disease in humans, particularly in parts of sub-Saharan Africa, and in Central and South America. Humans typically become infected with an arenavirus through contact with excreta from infected rodents. Tacaribe virus (TCRV) is an arenavirus that was first isolated from bats and mosquitoes during a rabies surveillance survey conducted in Trinidad from 1956 to 1958. Tacaribe virus is unusual because it has never been associated with a rodent host and since that one time isolation, the virus has not been isolated from any vertebrate or invertebrate hosts. We report the re-isolation of the virus from a pool of 100 host-seeking Amblyomma americanum (lone star ticks) collected in a Florida state park in 2012. TCRV was isolated in two cell lines and its complete genome was sequenced. The tick-derived isolate is nearly identical to the only remaining isolate from Trinidad (TRVL-11573), with 99.6% nucleotide identity across the genome. A quantitative RT-PCR assay was developed to test for viral RNA in host-seeking ticks collected from 3 Florida state parks. Virus RNA was detected in 56/500 (11.2%) of surveyed ticks. As this virus was isolated from ticks that parasitize humans, the ability of the tick to transmit the virus to people should be evaluated. Furthermore, reservoir hosts for the virus need to be identified in order to develop risk assessment models of human infection.
Collapse
Affiliation(s)
- Katherine A. Sayler
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida, United States of America
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, United States of America
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, United States of America
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
- * E-mail:
| | - Anthony F. Barbet
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, United States of America
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
| | - Casey Chamberlain
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, United States of America
| | - William L. Clapp
- Department of Pathology, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Rick Alleman
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Julia C. Loeb
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, United States of America
| | - John A. Lednicky
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, United States of America
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
40
|
Kerber R, Reindl S, Romanowski V, Gómez RM, Ogbaini-Emovon E, Günther S, ter Meulen J. Research efforts to control highly pathogenic arenaviruses: a summary of the progress and gaps. J Clin Virol 2014; 64:120-7. [PMID: 25549822 DOI: 10.1016/j.jcv.2014.12.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 12/07/2014] [Indexed: 01/08/2023]
Abstract
Significant progress has been made in the past 10 years in unraveling the molecular biology of highly pathogenic arenaviruses that are endemic in several West African countries (Lassa fever virus) and in some regions of South America (Argentine and Bolivian hemorrhagic fever viruses). While this has resulted in proof-of-concept studies of novel vaccine candidates in non-human primates and in the discovery of several novel antiviral small molecule drug candidates, none of them has been tested in the clinic to date. The recent Ebola outbreak in West Africa has demonstrated very clearly that there is an urgent need to develop the prophylactic and therapeutic armamentarium against viral hemorrhagic fever viruses as part of a global preparedness for future epidemics. As it pertains to this goal, the present article summarizes the current knowledge of highly pathogenic arenaviruses and identifies opportunities for translational research.
Collapse
Affiliation(s)
- R Kerber
- Department of Virology, Bernhard-Nocht-Institute for Tropical Medicine, Hamburg, Germany
| | - S Reindl
- Department of Virology, Bernhard-Nocht-Institute for Tropical Medicine, Hamburg, Germany
| | - V Romanowski
- Instituto de Biotecnología y Biología Molecular, CCT-La Plata, CONICET-UNLP, La Plata, Argentina
| | - R M Gómez
- Instituto de Biotecnología y Biología Molecular, CCT-La Plata, CONICET-UNLP, La Plata, Argentina
| | | | - S Günther
- Department of Virology, Bernhard-Nocht-Institute for Tropical Medicine, Hamburg, Germany
| | - J ter Meulen
- Institute of Virology, Philipps University Marburg, Germany.
| |
Collapse
|
41
|
Alimonti J, Leung A, Jones S, Gren J, Qiu X, Fernando L, Balcewich B, Wong G, Ströher U, Grolla A, Strong J, Kobinger G. Evaluation of transmission risks associated with in vivo replication of several high containment pathogens in a biosafety level 4 laboratory. Sci Rep 2014; 4:5824. [PMID: 25059478 PMCID: PMC5376055 DOI: 10.1038/srep05824] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 07/01/2014] [Indexed: 01/05/2023] Open
Abstract
Containment level 4 (CL4) laboratories studying biosafety level 4 viruses are under strict regulations to conduct nonhuman primate (NHP) studies in compliance of both animal welfare and biosafety requirements. NHPs housed in open-barred cages raise concerns about cross-contamination between animals, and accidental exposure of personnel to infectious materials. To address these concerns, two NHP experiments were performed. One examined the simultaneous infection of 6 groups of NHPs with 6 different viruses (Machupo, Junin, Rift Valley Fever, Crimean-Congo Hemorrhagic Fever, Nipah and Hendra viruses). Washing personnel between handling each NHP group, floor to ceiling biobubble with HEPA filter, and plexiglass between cages were employed for partial primary containment. The second experiment employed no primary containment around open barred cages with Ebola virus infected NHPs 0.3 meters from naïve NHPs. Viral antigen-specific ELISAs, qRT-PCR and TCID50 infectious assays were utilized to determine antibody levels and viral loads. No transmission of virus to neighbouring NHPs was observed suggesting limited containment protocols are sufficient for multi-viral CL4 experiments within one room. The results support the concept that Ebola virus infection is self-contained in NHPs infected intramuscularly, at least in the present experimental conditions, and is not transmitted to naïve NHPs via an airborne route.
Collapse
Affiliation(s)
- Judie Alimonti
- 1] Special Pathogens Program, Public Health Agency of Canada, 1015 Arlington St., Winnipeg. Manitoba [2]
| | - Anders Leung
- 1] Special Pathogens Program, Public Health Agency of Canada, 1015 Arlington St., Winnipeg. Manitoba [2]
| | - Shane Jones
- Special Pathogens Program, Public Health Agency of Canada, 1015 Arlington St., Winnipeg. Manitoba
| | - Jason Gren
- Containment Services, Public Health Agency of Canada, 1015 Arlington St., Winnipeg. Manitoba
| | - Xiangguo Qiu
- Special Pathogens Program, Public Health Agency of Canada, 1015 Arlington St., Winnipeg. Manitoba
| | - Lisa Fernando
- Special Pathogens Program, Public Health Agency of Canada, 1015 Arlington St., Winnipeg. Manitoba
| | - Brittany Balcewich
- Bioforensics Assay Development and Diagnostics; Public Health Agency of Canada, 1015 Arlington St., Winnipeg. Manitoba
| | - Gary Wong
- 1] Special Pathogens Program, Public Health Agency of Canada, 1015 Arlington St., Winnipeg. Manitoba [2] Departments of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Ute Ströher
- 1] Special Pathogens Program, Public Health Agency of Canada, 1015 Arlington St., Winnipeg. Manitoba [2]
| | - Allen Grolla
- Special Pathogens Program, Public Health Agency of Canada, 1015 Arlington St., Winnipeg. Manitoba
| | - James Strong
- 1] Special Pathogens Program, Public Health Agency of Canada, 1015 Arlington St., Winnipeg. Manitoba [2] Departments of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada [3] Departments of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Gary Kobinger
- 1] Special Pathogens Program, Public Health Agency of Canada, 1015 Arlington St., Winnipeg. Manitoba [2] Departments of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada [3] Departments of Immunology, University of Manitoba, Winnipeg, MB, Canada [4] Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
42
|
McLay L, Liang Y, Ly H. Comparative analysis of disease pathogenesis and molecular mechanisms of New World and Old World arenavirus infections. J Gen Virol 2014; 95:1-15. [PMID: 24068704 PMCID: PMC4093776 DOI: 10.1099/vir.0.057000-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 09/19/2013] [Indexed: 12/24/2022] Open
Abstract
Arenaviruses can cause fatal human haemorrhagic fever (HF) diseases for which vaccines and therapies are extremely limited. Both the New World (NW) and Old World (OW) groups of arenaviruses contain HF-causing pathogens. Although these two groups share many similarities, important differences with regard to pathogenicity and molecular mechanisms of virus infection exist. These closely related pathogens share many characteristics, including genome structure, viral assembly, natural host selection and the ability to interfere with innate immune signalling. However, members of the NW and OW viruses appear to use different receptors for cellular entry, as well as different mechanisms of virus internalization. General differences in disease signs and symptoms and pathological lesions in patients infected with either NW or OW arenaviruses are also noted and discussed herein. Whilst both the OW Lassa virus (LASV) and the NW Junin virus (JUNV) can cause disruption of the vascular endothelium, which is an important pathological feature of HF, the immune responses to these related pathogens seem to be quite distinct. Whereas LASV infection results in an overall generalized immune suppression, patients infected with JUNV seem to develop a cytokine storm. Additionally, the type of immune response required for recovery and clearance of the virus is different between NW and OW infections. These differences may be important to allow the viruses to evade host immune detection. Understanding these differences will aid the development of new vaccines and treatment strategies against deadly HF viral infections.
Collapse
MESH Headings
- Animals
- Arenaviridae Infections/immunology
- Arenaviridae Infections/pathology
- Arenaviridae Infections/virology
- Arenaviruses, New World/classification
- Arenaviruses, New World/genetics
- Arenaviruses, New World/immunology
- Arenaviruses, New World/pathogenicity
- Arenaviruses, Old World/classification
- Arenaviruses, Old World/genetics
- Arenaviruses, Old World/immunology
- Arenaviruses, Old World/pathogenicity
- Hemorrhagic Fevers, Viral/immunology
- Hemorrhagic Fevers, Viral/pathology
- Hemorrhagic Fevers, Viral/virology
- Humans
Collapse
Affiliation(s)
- Lisa McLay
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, MN 55108, USA
| | - Yuying Liang
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, MN 55108, USA
| | - Hinh Ly
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, MN 55108, USA
| |
Collapse
|
43
|
Abstract
The attenuated Lassa vaccine candidate ML29 is a laboratory-produced reassortant between Lassa and Mopeia viruses, two Old World arenaviruses that differ by 40% in nucleic acid sequence. In our previous studies, ML29 elicited sterilizing immunity against Lassa virus challenge in guinea pigs and marmosets and virus-specific cell-mediated immunity in both simian immunodeficiency virus (SIV)-infected and uninfected rhesus macaques. Here, we show that ML29 is stable after 12 passages in vitro without losing its plaque morphology or its attenuated phenotype in suckling mice. Additionally, we used deep sequencing to characterize the viral population comprising the original stock of ML29, the stock of ML29 after 12 passages in Vero cells, and the ML29 isolates obtained from vaccinated animals. Twenty-seven isolates bore approximately 77 mutations that exceeded 20% of the single-nucleotide polymorphism (SNP) changes at any single locus. Of these 77 mutations, 5 appeared to be host specific, for example, appearing in mice but not in primates. None of these mutations were reversions of ML29 to the sequences of the parental Lassa and Mopeia viruses. The host-specific mutations indicate viral adaptations to virus-host interactions, and such interactions make reasonable targets for antiviral approaches. Variants capable of chronic infection did not emerge from any of the primate infections, even in immune-deficient animals, indicating that the ML29 reassortant is reasonably stable in vivo. In conclusion, the preclinical studies of ML29 as a Lassa virus vaccine candidate have been advanced, showing high levels of protection in nonhuman primates and acceptable stability both in vitro and in vivo.
Collapse
|
44
|
Zapata JC, Salvato MS. Arenavirus variations due to host-specific adaptation. Viruses 2013; 5:241-78. [PMID: 23344562 PMCID: PMC3564120 DOI: 10.3390/v5010241] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 01/11/2013] [Accepted: 01/14/2013] [Indexed: 01/08/2023] Open
Abstract
Arenavirus particles are enveloped and contain two single-strand RNA genomic segments with ambisense coding. Genetic plasticity of the arenaviruses comes from transcription errors, segment reassortment, and permissive genomic packaging, and results in their remarkable ability, as a group, to infect a wide variety of hosts. In this review, we discuss some in vitro studies of virus genetic and phenotypic variation after exposure to selective pressures such as high viral dose, mutagens and antivirals. Additionally, we discuss the variation in vivo of selected isolates of Old World arenaviruses, particularly after infection of different animal species. We also discuss the recent emergence of new arenaviruses in the context of our observations of sequence variations that appear to be host-specific.
Collapse
Affiliation(s)
- Juan C Zapata
- Institute of Human Virology-School of Medicine, University of Maryland, Baltimore, MD 21201, USA.
| | | |
Collapse
|