1
|
Dong X, Zhang F, Wang Y, Li C, Li X, Zhou C, Wang G, Tian Z, Gao J, Zhou H, Sui L, Nauwynck H, Sorgeloos P, Holmes EC, Huang J, Shi W. A positive-sense single-stranded RNA virus acquired a negative-sense open reading frame through recombination. PLoS Pathog 2025; 21:e1013015. [PMID: 40198687 PMCID: PMC11978036 DOI: 10.1371/journal.ppat.1013015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 03/03/2025] [Indexed: 04/10/2025] Open
Abstract
Although positive- and negative-sense single-stranded RNA viruses are ubiquitous in nature, there is currently no evidence of recombination or reassortment between viruses with these two major forms of genome organization. Here, we describe the discovery of brine shrimp virga-like virus 1 (BSVV1), a novel positive-sense single-stranded RNA virus with a recombinant genome structure derived from two viral phyla with differing genome organizations. The genome of BSVV1 comprises three open reading frames (ORFs). ORF1 resembles the RNA-dependent RNA polymerase of Ips virga-like virus 1 (a positive-sense RNA virus), while ORF2, transcribed in the positive orientation, is related to the glycoprotein of Hubei bunya-like virus 10 and other negative-sense RNA viruses. The predicted ORF3 was unique to BSVV1 without known homologs identified. The presence of the three protein products was verified by mass spectrometry. Notably, our analysis also revealed that BSVV1 is geographically widespread and found in brine shrimp from at least eight countries on four continents. In addition, BSVV1 was successfully cultured and proliferated to high viral loads during brine shrimp development. In sum, we provide compelling evidence of an ancient recombination event between negative- and positive-sense single-stranded RNA viruses, enriching our understanding of the evolution of genome structures in RNA viruses.
Collapse
Affiliation(s)
- Xuan Dong
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center; Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture and Rural Affairs, Qingdao Key Laboratory of Mariculture Epidemiology and Biosecurity, Qingdao, China
| | - Fan Zhang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center; Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture and Rural Affairs, Qingdao Key Laboratory of Mariculture Epidemiology and Biosecurity, Qingdao, China
| | - Yiting Wang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center; Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture and Rural Affairs, Qingdao Key Laboratory of Mariculture Epidemiology and Biosecurity, Qingdao, China
| | - Cixiu Li
- Key Laboratory of Emerging Infectious Diseases in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, China
| | - Xuan Li
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center; Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture and Rural Affairs, Qingdao Key Laboratory of Mariculture Epidemiology and Biosecurity, Qingdao, China
| | - Chengyan Zhou
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center; Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture and Rural Affairs, Qingdao Key Laboratory of Mariculture Epidemiology and Biosecurity, Qingdao, China
| | - Guohao Wang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center; Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture and Rural Affairs, Qingdao Key Laboratory of Mariculture Epidemiology and Biosecurity, Qingdao, China
| | - Zhongshuai Tian
- Shanghai Institute of Virology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Gao
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hu Zhou
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Liying Sui
- Asian Regional Artemia Reference Center, Tianjin University of Science & Technology, Tianjin, China
| | - Hans Nauwynck
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Patrick Sorgeloos
- Laboratory of Aquaculture & Artemia Reference Center, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Edward C. Holmes
- School of Medical Sciences, The University of Sydney, Sydney, Australia
| | - Jie Huang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center; Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture and Rural Affairs, Qingdao Key Laboratory of Mariculture Epidemiology and Biosecurity, Qingdao, China
| | - Weifeng Shi
- Shanghai Institute of Virology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Terhzaz S, Kerrigan D, Almire F, Szemiel AM, Hughes J, Parvy JP, Palmarini M, Kohl A, Shi X, Pondeville E. NSm is a critical determinant for bunyavirus transmission between vertebrate and mosquito hosts. Nat Commun 2025; 16:1214. [PMID: 39890788 PMCID: PMC11785797 DOI: 10.1038/s41467-024-54809-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 11/21/2024] [Indexed: 02/03/2025] Open
Abstract
Bunyavirales is a very large order including viruses infecting a variety of taxonomic groups such as arthropods, vertebrates, plants, and protozoa. Some bunyaviruses are transmitted between vertebrate hosts by blood-sucking arthropods and cause major diseases in humans and animals. It is not understood why only some bunyaviruses have evolved the capacity to be transmitted by arthropod vectors. Here we show that only vector-borne bunyaviruses express a non-structural protein, NSm, whose function has so far remained largely elusive. Using as experimental system Bunyamwera virus (BUNV) and its invertebrate host, Aedes aegypti, we show that NSm is dispensable for viral replication in mosquito cells in vitro but is absolutely required for successful infection in the female mosquito following a blood meal. More specifically, NSm is required for cell-to-cell spread and egress from the mosquito midgut, a known barrier to viral infection. Notably, the requirement for NSm is specific to the midgut; bypassing this barrier by experimental intrathoracic infection of the mosquito eliminates the necessity of NSm for virus spread in other tissues, including the salivary glands. Overall, we unveiled a key evolutionary process that allows the transmission of vector-borne bunyaviruses between arthropod and vertebrate hosts.
Collapse
Affiliation(s)
- Selim Terhzaz
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK
| | - David Kerrigan
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK
| | - Floriane Almire
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK
| | - Agnieszka M Szemiel
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK
| | - Joseph Hughes
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK
| | - Jean-Philippe Parvy
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK
| | - Massimo Palmarini
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK
- Departments of Tropical Disease Biology and Vector Biology, Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK
| | - Xiaohong Shi
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| | - Emilie Pondeville
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| |
Collapse
|
3
|
Gao H, Zhao D, Li C, Deng M, Li G, Chen S, Zhao M, Qin L, Zhang K. The Role of Orthobunyavirus Glycoprotein Gc in the Viral Life Cycle: From Viral Entry to Egress. Molecules 2025; 30:503. [PMID: 39942606 PMCID: PMC11820035 DOI: 10.3390/molecules30030503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Orthobunyavirus refers to the virus members within the Genus Orthobunyavirus, which is the largest virus genus in the Family Peribunyaviridae and even Class Bunyaviricetes. To date, over 130 species of Orthobunyaviruses have been identified worldwide. Orthobunyaviruses mainly infect arthropods, while some species are capable of being transmitted to mammals, including humans, via intermediate vectors. As emerging and re-emerging pathogens, orthobunyavirus poses a significant threat to both human and veterinary public health worldwide. Currently, there are no commercial vaccines against orthobunyavirus. The structure of orthobunyavirus is relatively simple, consisting of a typical tri-segmented negative-sense RNA genome that encodes four structural proteins (L, Gn, Gc, and N) and two non-structural proteins (NSm and NSs). The highly glycosylated Gc protein, which has a complex conformation and forms polymers embedded in the viral envelope, plays a critical role in inducing neutralizing antibodies throughout the orthobunyavirus infection cycle from entry to egress. This review provides a comprehensive summary of the virus-encoded Gc protein and its role in the virus life cycle from viral entry to egress, offering researchers with valuable integrated information for further investigations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Limei Qin
- School of Animal Science and Technology, Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Foshan University, Foshan 528225, China; (H.G.)
| | - Keshan Zhang
- School of Animal Science and Technology, Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Foshan University, Foshan 528225, China; (H.G.)
| |
Collapse
|
4
|
Wernike K, Beer M. More than a decade of research on Schmallenberg virus-Knowns and unknowns. Adv Virus Res 2024; 120:77-98. [PMID: 39455169 DOI: 10.1016/bs.aivir.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
Schmallenberg virus, an arbovirus of the Orthobunyavirus genus that primarily infects ruminants, emerged in 2011 near the Dutch-German border region and subsequently caused a large number of abortions and the births of severely malformed newborns in the European livestock population. Immediate intensive research led to the development of reliable diagnostic tests, the identification of competent Culicoides vector species, and the elucidation of the pathogenesis in infected vertebrate hosts. In addition, the structure of the major antigenic domain has been elucidated in great detail, leading to the development of effective marker vaccine candidates. The knowledge gained over the last decade on the biology and pathogenesis of SBV and the experience acquired in its control will be of great value in the future for the control of any similar emerging pathogen of veterinary or public health importance such as Shuni or Oropouche virus. However, some important knowledge gaps remain, for example, the factors contributing to the highly variable transmission rate from dam to fetus or the viral factors responsible for the vector competence of Culicoides midges are largely unknown. Thus, questions still remain for the next decade of research on SBV and related viruses.
Collapse
Affiliation(s)
- Kerstin Wernike
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany.
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| |
Collapse
|
5
|
Rondeau NC, Spector SN, Thannickal SA, Stapleford KA. La Crosse virus reassortants highlight genomic determinants of infection and pathogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584386. [PMID: 38559198 PMCID: PMC10979930 DOI: 10.1101/2024.03.11.584386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The genomic determinants that contribute to orthobunyavirus infection and pathogenesis are not well-defined. In this study, we harnessed the process of reassortment to understand which viral factors drive change in the replication and pathogenesis of La Crosse virus (LACV). We systematically reassorted the genomic segments of two genetically similar Lineage I LACV isolates into six unique reassortants. Despite the parental isolates having high levels of RNA and protein consensus, the reassortants demonstrate how minimal changes in RNA and protein structure can have significant changes in viral growth and reproduction in vitro in mammalian and insect models. We observed that swapping the S segment between isolates led to differences in replication and assembly resulting in one non-rescuable reassortant and one viable reassortant that exhibited an increase in viral growth dynamics. Switching the M segment led to changes in viral plaque phenotype and growth kinetics. L segment reassortants similarly differed in changes in viral growth dynamics. We further explored the M segment reassortants in a neonate mouse model and observed a role for the M segment in neuroinflammation and virulence. Through reassortment of the La Crosse virus genomic segments, we are able to further understand how genomic determinants of infection and pathogenesis operate in orthobunyaviruses. Future investigations will focus on identifying the specific molecular elements that govern the observed phenotypes in vitro and in vivo . Importance La Crosse virus is the leading cause of pediatric arboviral encephalitis in the United States, yet it is largely unknown how each of the three genomic segments contribute to pathogenesis and disease. Our study utilizes genomic reassortment between two similar Lineage I LACV isolates to understand genomic determinants for differences in infection and pathogenesis phenotypes in vitro and in vivo. By identifying roles for each segment in observed outcomes, we are able to plan further studies for molecular characterization of these phenotypes. Additionally, it is imperative to continue to characterize orthobunyavirus function since climate change will expand the range and prevalence of arthropod-borne diseases such as LACV in the United States.
Collapse
|
6
|
Lefebvre V, Leon Foun Lin R, Cole L, Cosset FL, Fogeron ML, Böckmann A. Do NSm Virulence Factors in the Bunyavirales Viral Order Originate from Gn Gene Duplication? Viruses 2024; 16:90. [PMID: 38257790 PMCID: PMC10819786 DOI: 10.3390/v16010090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/29/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
One-third of the nine WHO shortlisted pathogens prioritized for research and development in public health emergencies belong to the Bunyavirales order. Several Bunyavirales species carry an NSm protein that acts as a virulence factor. We predicted the structures of these NSm proteins and unexpectedly found that in two families, their cytosolic domain was inferred to have a similar fold to that of the cytosolic domain of the viral envelope-forming glycoprotein N (Gncyto) encoded on the same genome fragment. We show that although the sequence identity between the NSmcyto and the Gncyto domains is low, the conservation of the two zinc finger-forming CysCysHisCys motifs explains the predicted structural conservation. Importantly, our predictions provide a first glimpse into the long-unknown structure of NSm. Also, these predictions suggest that NSm is the result of a gene duplication event in the Bunyavirales Nairoviridae and Peribunyaviridae families and that such events may be common in the recent evolutionary history of RNA viruses.
Collapse
Affiliation(s)
- Victor Lefebvre
- MMSB—Molecular Microbiology and Structural Biochemistry, Université de Lyon 1, CNRS UMR 5086, F-69367 Lyon, France; (V.L.); (R.L.F.L.)
| | - Ravy Leon Foun Lin
- MMSB—Molecular Microbiology and Structural Biochemistry, Université de Lyon 1, CNRS UMR 5086, F-69367 Lyon, France; (V.L.); (R.L.F.L.)
| | - Laura Cole
- MMSB—Molecular Microbiology and Structural Biochemistry, Université de Lyon 1, CNRS UMR 5086, F-69367 Lyon, France; (V.L.); (R.L.F.L.)
| | - François-Loïc Cosset
- CIRI—Centre International de Recherche en Infectiologie, Université de Lyon 1, ENS de Lyon, Inserm U1111, CNRS UMR 5308, F-69007 Lyon, France
| | - Marie-Laure Fogeron
- MMSB—Molecular Microbiology and Structural Biochemistry, Université de Lyon 1, CNRS UMR 5086, F-69367 Lyon, France; (V.L.); (R.L.F.L.)
| | - Anja Böckmann
- MMSB—Molecular Microbiology and Structural Biochemistry, Université de Lyon 1, CNRS UMR 5086, F-69367 Lyon, France; (V.L.); (R.L.F.L.)
| |
Collapse
|
7
|
Jurado-Cobena E. OROPOUCHE VIRUS: MORE QUESTIONS THAN ANSWERS. ZOONOSES (BURLINGTON, MASS.) 2024; 4:24. [PMID: 39575443 PMCID: PMC11580152 DOI: 10.15212/zoonoses-2024-0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2024]
Abstract
Oropouche virus (genus Orthobunyavirus, family Peribunyaviridae) is an arthropod-borne virus that infects several species of animals and humans mostly in South America. Despite being described as a human pathogen over 60 years ago, little progress has been made towards the ecological and pathological aspects of this pathogen. However, with recent viral spread northward reaching Haiti and Cuba, it has been receiving more attention, evidenced by the growing number of relevant research articles. This commentary article provides the summary of the potential natural reservoirs and the expansion of endemic regions within the context of One Health. The clinical aspects of the human infection are revisited and discussed based on the latest evidence. The article briefly review research on the molecular virology and the pathology, highlighting unanswered questions crucial for comprehensive understanding of this viral disease, which imposes a significant burden on the affected populations.
Collapse
Affiliation(s)
- Eduardo Jurado-Cobena
- Department of Microbiology & Immunology, Graduate School of Biomedical Science
- Department of Biostatistics & Data Science, School of Public and Population Health University of Texas Medical Branch at Galveston, Galveston. TX, USA
| |
Collapse
|
8
|
Snyman J, Snyman LP, Buhler KJ, Villeneuve CA, Leighton PA, Jenkins EJ, Kumar A. California Serogroup Viruses in a Changing Canadian Arctic: A Review. Viruses 2023; 15:1242. [PMID: 37376542 DOI: 10.3390/v15061242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/24/2023] [Accepted: 05/18/2023] [Indexed: 06/29/2023] Open
Abstract
The Arctic is warming at four times the global rate, changing the diversity, activity and distribution of vectors and associated pathogens. While the Arctic is not often considered a hotbed of vector-borne diseases, Jamestown Canyon virus (JCV) and Snowshoe Hare virus (SSHV) are mosquito-borne zoonotic viruses of the California serogroup endemic to the Canadian North. The viruses are maintained by transovarial transmission in vectors and circulate among vertebrate hosts, both of which are not well characterized in Arctic regions. While most human infections are subclinical or mild, serious cases occur, and both JCV and SSHV have recently been identified as leading causes of arbovirus-associated neurological diseases in North America. Consequently, both viruses are currently recognised as neglected and emerging viruses of public health concern. This review aims to summarise previous findings in the region regarding the enzootic transmission cycle of both viruses. We identify key gaps and approaches needed to critically evaluate, detect, and model the effects of climate change on these uniquely northern viruses. Based on limited data, we predict that (1) these northern adapted viruses will increase their range northwards, but not lose range at their southern limits, (2) undergo more rapid amplification and amplified transmission in endemic regions for longer vector-biting seasons, (3) take advantage of northward shifts of hosts and vectors, and (4) increase bite rates following an increase in the availability of breeding sites, along with phenological synchrony between the reproduction cycle of theorized reservoirs (such as caribou calving) and mosquito emergence.
Collapse
Affiliation(s)
- Jumari Snyman
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Louwrens P Snyman
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Kayla J Buhler
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Carol-Anne Villeneuve
- Research Group on Epidemiology of Zoonoses and Public Health (GREZOSP), Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Patrick A Leighton
- Research Group on Epidemiology of Zoonoses and Public Health (GREZOSP), Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Emily J Jenkins
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Anil Kumar
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
9
|
Abstract
Unlike many segmented negative-sense RNA viruses, most members of the Bunyavirales bud at Golgi membranes, as opposed to the plasma membrane. Central players in this assembly process are the envelope glycoproteins, Gn and Gc, which upon translation undergo proteolytic processing, glycosylation and trafficking to the Golgi, where they interact with ribonucleoprotein genome segments and bud into Golgi-derived compartments. The processes involved in genome packaging during virion assembly can lead to the generation of reassorted viruses, if a cell is co-infected with two different bunyaviruses, due to mismatching of viral genome segment packaging. This can lead to viruses with high pathogenic potential, as demonstrated by the emergence of Schmallenberg virus. This review focuses on the assembly pathways of tri-segmented bunyaviruses, highlighting some areas in need of further research to understand these important pathogens with zoonotic potential.
Collapse
Affiliation(s)
- Jake Barker
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Luis L P daSilva
- Departamento de Biologia Celular e Molecular, Centro de Pesquisa em Virologia, Faculdade de Medicina de Ribeirão Preto, University of São Paulo, State of São Paulo, Brazil
| | - Colin M Crump
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
10
|
Foster JE, López K, Eastwood G, Guzman H, Carrington CVF, Tesh RB, Auguste AJ. Phylogenetic characterization of Orthobunyaviruses isolated from Trinidad shows evidence of natural reassortment. Virus Genes 2023; 59:473-478. [PMID: 36763228 DOI: 10.1007/s11262-023-01973-5] [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: 10/18/2022] [Accepted: 01/29/2023] [Indexed: 02/11/2023]
Abstract
The genus Orthobunyavirus is a diverse group of viruses in the family Peribunyaviridae, recently classified into 20 serogroups, and 103 virus species. Although most viruses within these serogroups are phylogenetically distinct, the absence of complete genome sequences has left several viruses incompletely characterized. Here we report the complete genome sequences for 11 orthobunyaviruses isolated from Trinidad, French Guiana, Guatemala, and Panama that were serologically classified into six serogroups and 10 species. Phylogenetic analyses of these 11 newly derived sequences indicate that viruses belonging to the Patois, Capim, Guama, and Group C serocomplexes all have a close genetic origin. We show that three of the 11 orthobunyaviruses characterized (belonging to the Group C and Bunyamwera serogroups) have evidence of histories of natural reassortment through the M genome segment. Our data also suggests that two distinct lineages of Group C viruses concurrently circulate in Trinidad and are transmitted by the same mosquito vectors. This study also highlights the importance of complementing serological identification with nucleotide sequencing when characterizing orthobunyaviruses.
Collapse
Affiliation(s)
- Jerome E Foster
- Department of Preclinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Republic of Trinidad and Tobago
| | - Krisangel López
- Department of Entomology, College of Agriculture and Life Sciences, Fralin Life Science Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Gillian Eastwood
- Department of Entomology, College of Agriculture and Life Sciences, Fralin Life Science Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA.,Center for Emerging, Zoonotic, and Arthropod-Borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA.,Global Change Center at Virginia Tech, Blacksburg, VA, 24061, USA
| | - Hilda Guzman
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Christine V F Carrington
- Department of Preclinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Republic of Trinidad and Tobago
| | - Robert B Tesh
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Albert J Auguste
- Department of Entomology, College of Agriculture and Life Sciences, Fralin Life Science Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA. .,Center for Emerging, Zoonotic, and Arthropod-Borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA.
| |
Collapse
|
11
|
Barbosa NS, Concha JO, daSilva LLP, Crump CM, Graham SC. Oropouche Virus Glycoprotein Topology and Cellular Requirements for Glycoprotein Secretion. J Virol 2023; 97:e0133122. [PMID: 36475765 PMCID: PMC9888203 DOI: 10.1128/jvi.01331-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/19/2022] [Indexed: 12/13/2022] Open
Abstract
Oropouche virus (OROV; genus Orthobunyavirus) is the etiological agent of Oropouche fever, a debilitating febrile illness common in South America. We used recombinant expression of the OROV M polyprotein, which encodes the surface glycoproteins Gn and Gc plus the nonstructural protein NSm, to probe the cellular determinants for OROV assembly and budding. Gn and Gc self-assemble and are secreted independently of NSm. Mature OROV Gn has two predicted transmembrane domains that are crucial for glycoprotein translocation to the Golgi complex and glycoprotein secretion, and unlike related orthobunyaviruses, both transmembrane domains are retained during Gn maturation. Disruption of Golgi function using the drugs brefeldin A and monensin inhibits glycoprotein secretion. Infection studies have previously shown that the cellular endosomal sorting complexes required for transport (ESCRT) machinery is recruited to Golgi membranes during OROV assembly and that ESCRT activity is required for virus secretion. A dominant-negative form of the ESCRT-associated ATPase VPS4 significantly reduces recombinant OROV glycoprotein secretion and blocks virus release from infected cells, and VPS4 partly colocalizes with OROV glycoproteins and membranes costained with Golgi markers. Furthermore, immunoprecipitation and fluorescence microscopy experiments demonstrate that OROV glycoproteins interact with the ESCRT-III component CHMP6, with overexpression of a dominant-negative form of CHMP6 significantly reducing OROV glycoprotein secretion. Taken together, our data highlight differences in M polyprotein processing across orthobunyaviruses, indicate that Golgi and ESCRT function are required for glycoprotein secretion, and identify CHMP6 as an ESCRT-III component that interacts with OROV glycoproteins. IMPORTANCE Oropouche virus causes Oropouche fever, a debilitating illness common in South America that is characterized by high fever, headache, myalgia, and vomiting. The tripartite genome of this zoonotic virus is capable of reassortment, and there have been multiple epidemics of Oropouche fever in South America over the last 50 years, making Oropouche virus infection a significant threat to public health. However, the molecular characteristics of this arbovirus are poorly understood. We developed a recombinant protein expression system to investigate the cellular determinants of OROV glycoprotein maturation and secretion. We show that the proteolytic processing of the M polypeptide, which encodes the surface glycoproteins (Gn and Gc) plus a nonstructural protein (NSm), differs between OROV and its close relative Bunyamwera virus. Furthermore, we demonstrate that OROV M glycoprotein secretion requires the cellular endosomal sorting complexes required for transport (ESCRT) membrane-remodeling machinery and identify that the OROV glycoproteins interact with the ESCRT protein CHMP6.
Collapse
Affiliation(s)
- Natalia S. Barbosa
- Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Juan O. Concha
- Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Luis L. P. daSilva
- Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Colin M. Crump
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Stephen C. Graham
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
12
|
Ayers VB, Huang YJS, Kohl A, Dunlop JI, Hettenbach SM, Park SL, Higgs S, Vanlandingham DL. Comparison of Immunogenicity Between a Candidate Live Attenuated Vaccine and an Inactivated Vaccine for Cache Valley Virus. Viral Immunol 2023; 36:41-47. [PMID: 36622942 PMCID: PMC9885547 DOI: 10.1089/vim.2022.0103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Cache Valley virus (CVV) is a mosquito-borne bunyavirus that is enzootic throughout the new world. Although CVV is known as an important agricultural pathogen, primarily associated with embryonic lethality and abortions in ruminants, it has recently been recognized for its expansion as a zoonotic pathogen. With the increased emergence of bunyaviruses with human and veterinary importance, there have been significant efforts dedicated to the development of bunyavirus vaccines. In this study, the immunogenicity of a candidate live-attenuated vaccine (LAV) for CVV, which contains the deletion of the nonstructural small (NSs) and nonstructural medium (NSm) genes (2delCVV), was evaluated and compared with an autogenous candidate vaccine created through the inactivation of CVV using binary ethylenimine (BEI) with an aluminum hydroxide adjuvant (BEI-CVV) in sheep. Both 2delCVV and BEI-CVV produced a neutralizing antibody response that exceeds the correlate of protection, that is, plaque reduction neutralization test titer >10. However, on day 63 postinitial immunization, 2delCVV was more immunogenic than BEI-CVV. These results warrant further development of 2delCVV as a candidate LAV and demonstrate that the double deletion of the NSs and NSm genes can be applied to the development of vaccines and as a common attenuation strategy for orthobunyaviruses.
Collapse
Affiliation(s)
- Victoria B. Ayers
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA.,Biosecurity Research Institute, Kansas State University, Manhattan, Kansas, USA
| | - Yan-Jang S. Huang
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA.,Biosecurity Research Institute, Kansas State University, Manhattan, Kansas, USA
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - James I. Dunlop
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Susan M. Hettenbach
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA.,Biosecurity Research Institute, Kansas State University, Manhattan, Kansas, USA
| | - So Lee Park
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA.,Biosecurity Research Institute, Kansas State University, Manhattan, Kansas, USA
| | - Stephen Higgs
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA.,Biosecurity Research Institute, Kansas State University, Manhattan, Kansas, USA
| | - Dana L. Vanlandingham
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA.,Biosecurity Research Institute, Kansas State University, Manhattan, Kansas, USA.,Address correspondence to: Dr. Dana L. Vanlandingham, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
13
|
Boshra H. An Overview of the Infectious Cycle of Bunyaviruses. Viruses 2022; 14:2139. [PMID: 36298693 PMCID: PMC9610998 DOI: 10.3390/v14102139] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Bunyaviruses represent the largest group of RNA viruses and are the causative agent of a variety of febrile and hemorrhagic illnesses. Originally characterized as a single serotype in Africa, the number of described bunyaviruses now exceeds over 500, with its presence detected around the world. These predominantly tri-segmented, single-stranded RNA viruses are transmitted primarily through arthropod and rodent vectors and can infect a wide variety of animals and plants. Although encoding for a small number of proteins, these viruses can inflict potentially fatal disease outcomes and have even developed strategies to suppress the innate antiviral immune mechanisms of the infected host. This short review will attempt to provide an overall description of the order Bunyavirales, describing the mechanisms behind their infection, replication, and their evasion of the host immune response. Furthermore, the historical context of these viruses will be presented, starting from their original discovery almost 80 years ago to the most recent research pertaining to viral replication and host immune response.
Collapse
Affiliation(s)
- Hani Boshra
- Global Urgent and Advanced Research and Development (GUARD), 911 Rue Principale, Batiscan, QC G0X 1A0, Canada
| |
Collapse
|
14
|
OGAWA Y, EGUCHI M, SHIMOJI Y. Two Akabane virus glycoprotein Gc domains induce neutralizing antibodies in mice. J Vet Med Sci 2022; 84:538-542. [PMID: 35197396 PMCID: PMC9096034 DOI: 10.1292/jvms.21-0670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 02/10/2022] [Indexed: 11/22/2022] Open
Abstract
Akabane virus (AKAV), belonging to the genus Orthobunyavirus and family Peribunyaviridae, causes reproductive and congenital abnormalities in ruminants. Its envelope glycoprotein Gc is a neutralizing antigen, on which at least five distinct antigenic regions have been identified. We attempted to identify the domains using truncated recombinant AKAV Gc proteins expressed in Escherichia coli and monoclonal antibodies (mAbs) with AKAV-neutralizing activity. Dot blot analysis revealed that amino acid positions 1-97 and 189-397 (Gc1-97 and Gc189-397) in the truncated recombinant proteins reacted with the mAbs. Additionally, AKAV was neutralized by sera from mice immunized with these recombinant proteins. The results suggested that the two domains contain neutralizing epitopes and could be potential subunit vaccines against AKAV.
Collapse
Affiliation(s)
- Yohsuke OGAWA
- Division of Infectious Animal
Disease Research, National Institute of Animal Health, National Agriculture and Food
Research Organization, Ibaraki, Japan
| | - Masahiro EGUCHI
- Division of Infectious Animal
Disease Research, National Institute of Animal Health, National Agriculture and Food
Research Organization, Ibaraki, Japan
| | - Yoshihiro SHIMOJI
- Division of Infectious Animal
Disease Research, National Institute of Animal Health, National Agriculture and Food
Research Organization, Ibaraki, Japan
- Research Institute for
Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| |
Collapse
|
15
|
Genetic and pathogenic characterisation of a virulent Akabane virus isolated from goats in Yunnan, China. J Vet Res 2022; 66:35-42. [PMID: 35582486 PMCID: PMC8959687 DOI: 10.2478/jvetres-2022-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 02/01/2022] [Indexed: 11/21/2022] Open
Abstract
Introduction Akabane virus (AKAV) has been detected in a variety of host species in China, but there are only limited records of its occurrence in goats. However, more attention needs to be paid to understanding the diversity of viruses in this species. The aim of the study was to explore the genotype characteristics and variation trend of AKAV and their relationship with virulence in Yunnan, China. Material and Methods Blood samples were collected from goats during routine surveillance of goat diseases in Yunnan province in 2019. The AKAV CX-01 strain was isolated using BHK-21 cells. To understand pathogenicity, the virus was intraperitoneally (IP) and intracerebrally (IC) inoculated into suckling mice and tissue samples were subsequently analysed histopathologically and immunohistochemically. Results Akabane virus CX-01 strain induced encephalitis and impairment of the central nervous system with fatal consequences. Phylogenetic analysis based on the ORF sequences of the small segments indicated that the AKAV isolate used was most closely related to the GD18134/2018 Chinese midge and bovine NM BS/1strains, while phylogenetic analysis based on the medium segments showed a close relationship between CX-01 and the Chinese GLXCH01 strain. Conclusion The CX-01 isolate was related to AKAV genogroup Ia and probably originated from a recombination of different strains.
Collapse
|
16
|
Ren F, Shen S, Wang Q, Wei G, Huang C, Wang H, Ning YJ, Zhang DY, Deng F. Recent Advances in Bunyavirus Reverse Genetics Research: Systems Development, Applications, and Future Perspectives. Front Microbiol 2021; 12:771934. [PMID: 34950119 PMCID: PMC8689132 DOI: 10.3389/fmicb.2021.771934] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/03/2021] [Indexed: 12/25/2022] Open
Abstract
Bunyaviruses are members of the Bunyavirales order, which is the largest group of RNA viruses, comprising 12 families, including a large group of emerging and re-emerging viruses. These viruses can infect a wide variety of species worldwide, such as arthropods, protozoans, plants, animals, and humans, and pose substantial threats to the public. In view of the fact that a better understanding of the life cycle of a highly pathogenic virus is often a precondition for developing vaccines and antivirals, it is urgent to develop powerful tools to unravel the molecular basis of the pathogenesis. However, biosafety level −3 or even −4 containment laboratory is considered as a necessary condition for working with a number of bunyaviruses, which has hampered various studies. Reverse genetics systems, including minigenome (MG), infectious virus-like particle (iVLP), and infectious full-length clone (IFLC) systems, are capable of recapitulating some or all steps of the viral replication cycle; among these, the MG and iVLP systems have been very convenient and effective tools, allowing researchers to manipulate the genome segments of pathogenic viruses at lower biocontainment to investigate the viral genome transcription, replication, virus entry, and budding. The IFLC system is generally developed based on the MG or iVLP systems, which have facilitated the generation of recombinant infectious viruses. The MG, iVLP, and IFLC systems have been successfully developed for some important bunyaviruses and have been widely employed as powerful tools to investigate the viral replication cycle, virus–host interactions, virus pathogenesis, and virus evolutionary process. The majority of bunyaviruses is generally enveloped negative-strand RNA viruses with two to six genome segments, of which the viruses with bipartite and tripartite genome segments have mostly been characterized. This review aimed to summarize current knowledge on reverse genetic studies of representative bunyaviruses causing severe diseases in humans and animals, which will contribute to the better understanding of the bunyavirus replication cycle and provide some hints for developing designed antivirals.
Collapse
Affiliation(s)
- Fuli Ren
- Research Center for Translational Medicine, Wuhan Jinyintan Hospital, Wuhan, China.,State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Shu Shen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Qiongya Wang
- Research Center for Translational Medicine, Wuhan Jinyintan Hospital, Wuhan, China
| | - Gang Wei
- Research Center for Translational Medicine, Wuhan Jinyintan Hospital, Wuhan, China
| | - Chaolin Huang
- Research Center for Translational Medicine, Wuhan Jinyintan Hospital, Wuhan, China
| | - Hualin Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yun-Jia Ning
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Ding-Yu Zhang
- Research Center for Translational Medicine, Wuhan Jinyintan Hospital, Wuhan, China
| | - Fei Deng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
17
|
Hulswit RJG, Paesen GC, Bowden TA, Shi X. Recent Advances in Bunyavirus Glycoprotein Research: Precursor Processing, Receptor Binding and Structure. Viruses 2021; 13:353. [PMID: 33672327 PMCID: PMC7926653 DOI: 10.3390/v13020353] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 01/04/2023] Open
Abstract
The Bunyavirales order accommodates related viruses (bunyaviruses) with segmented, linear, single-stranded, negative- or ambi-sense RNA genomes. Their glycoproteins form capsomeric projections or spikes on the virion surface and play a crucial role in virus entry, assembly, morphogenesis. Bunyavirus glycoproteins are encoded by a single RNA segment as a polyprotein precursor that is co- and post-translationally cleaved by host cell enzymes to yield two mature glycoproteins, Gn and Gc (or GP1 and GP2 in arenaviruses). These glycoproteins undergo extensive N-linked glycosylation and despite their cleavage, remain associated to the virion to form an integral transmembrane glycoprotein complex. This review summarizes recent advances in our understanding of the molecular biology of bunyavirus glycoproteins, including their processing, structure, and known interactions with host factors that facilitate cell entry.
Collapse
Affiliation(s)
- Ruben J. G. Hulswit
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (R.J.G.H.); (G.C.P.)
| | - Guido C. Paesen
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (R.J.G.H.); (G.C.P.)
| | - Thomas A. Bowden
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (R.J.G.H.); (G.C.P.)
| | - Xiaohong Shi
- MRC-University of Glasgow Centre for Virus Research, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G61 1QH, UK
| |
Collapse
|
18
|
A Look into Bunyavirales Genomes: Functions of Non-Structural (NS) Proteins. Viruses 2021; 13:v13020314. [PMID: 33670641 PMCID: PMC7922539 DOI: 10.3390/v13020314] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
In 2016, the Bunyavirales order was established by the International Committee on Taxonomy of Viruses (ICTV) to incorporate the increasing number of related viruses across 13 viral families. While diverse, four of the families (Peribunyaviridae, Nairoviridae, Hantaviridae, and Phenuiviridae) contain known human pathogens and share a similar tri-segmented, negative-sense RNA genomic organization. In addition to the nucleoprotein and envelope glycoproteins encoded by the small and medium segments, respectively, many of the viruses in these families also encode for non-structural (NS) NSs and NSm proteins. The NSs of Phenuiviridae is the most extensively studied as a host interferon antagonist, functioning through a variety of mechanisms seen throughout the other three families. In addition, functions impacting cellular apoptosis, chromatin organization, and transcriptional activities, to name a few, are possessed by NSs across the families. Peribunyaviridae, Nairoviridae, and Phenuiviridae also encode an NSm, although less extensively studied than NSs, that has roles in antagonizing immune responses, promoting viral assembly and infectivity, and even maintenance of infection in host mosquito vectors. Overall, the similar and divergent roles of NS proteins of these human pathogenic Bunyavirales are of particular interest in understanding disease progression, viral pathogenesis, and developing strategies for interventions and treatments.
Collapse
|
19
|
Hawman DW, Meade-White K, Leventhal S, Feldmann F, Okumura A, Smith B, Scott D, Feldmann H. Immunocompetent mouse model for Crimean-Congo hemorrhagic fever virus. eLife 2021; 10:63906. [PMID: 33416494 PMCID: PMC7811403 DOI: 10.7554/elife.63906] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 01/07/2021] [Indexed: 02/06/2023] Open
Abstract
Crimean-Congo hemorrhagic fever (CCHF) is a severe tick-borne febrile illness with wide geographic distribution. CCHF is caused by infection with the Crimean-Congo hemorrhagic fever virus (CCHFV) and case fatality rates can be as high as 30%. Despite causing severe disease in humans, our understanding of the host and viral determinants of CCHFV pathogenesis are limited. A major limitation in the investigation of CCHF has been the lack of suitable small animal models. Wild-type mice are resistant to clinical isolates of CCHFV and consequently, mice must be deficient in type I interferon responses to study the more severe aspects of CCHFV. We report here a mouse-adapted variant of CCHFV that recapitulates in adult, immunocompetent mice the severe CCHF observed in humans. This mouse-adapted variant of CCHFV significantly improves our ability to study host and viral determinants of CCHFV-induced disease in a highly tractable mouse model.
Collapse
Affiliation(s)
- David W Hawman
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Hamilton, United States
| | - Kimberly Meade-White
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Hamilton, United States
| | - Shanna Leventhal
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Hamilton, United States
| | - Friederike Feldmann
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Hamilton, United States
| | - Atsushi Okumura
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Hamilton, United States
| | - Brian Smith
- Texas Veterinary Pathology, Spring Branch, United States
| | - Dana Scott
- Rocky Mountain Veterinary Branch, Division of Intramural Research, NIAID, NIH, Hamilton, United States
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Hamilton, United States
| |
Collapse
|
20
|
Ren S, Ding C, Sun Y. Morphology Remodeling and Selective Autophagy of Intracellular Organelles during Viral Infections. Int J Mol Sci 2020; 21:ijms21103689. [PMID: 32456258 PMCID: PMC7279407 DOI: 10.3390/ijms21103689] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/14/2020] [Accepted: 05/21/2020] [Indexed: 12/17/2022] Open
Abstract
Viruses have evolved different strategies to hijack subcellular organelles during their life cycle to produce robust infectious progeny. Successful viral reproduction requires the precise assembly of progeny virions from viral genomes, structural proteins, and membrane components. Such spatial and temporal separation of assembly reactions depends on accurate coordination among intracellular compartmentalization in multiple organelles. Here, we overview the rearrangement and morphology remodeling of virus-triggered intracellular organelles. Focus is given to the quality control of intracellular organelles, the hijacking of the modified organelle membranes by viruses, morphology remodeling for viral replication, and degradation of intracellular organelles by virus-triggered selective autophagy. Understanding the functional reprogram and morphological remodeling in the virus-organelle interplay can provide new insights into the development of broad-spectrum antiviral strategies.
Collapse
Affiliation(s)
- Shanhui Ren
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute. Chinese Academy of Agricultural Science, Shanghai 200241, China;
| | - Chan Ding
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute. Chinese Academy of Agricultural Science, Shanghai 200241, China;
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Correspondence: (C.D.); (Y.S.); Tel.: +86-21-34293441 (C.D. & Y.S.); Fax: +86-21-54081818 (C.D. & Y.S.)
| | - Yingjie Sun
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute. Chinese Academy of Agricultural Science, Shanghai 200241, China;
- Correspondence: (C.D.); (Y.S.); Tel.: +86-21-34293441 (C.D. & Y.S.); Fax: +86-21-54081818 (C.D. & Y.S.)
| |
Collapse
|
21
|
The Crimean-Congo Hemorrhagic Fever Virus NSm Protein is Dispensable for Growth In Vitro and Disease in Ifnar -/- Mice. Microorganisms 2020; 8:microorganisms8050775. [PMID: 32455700 PMCID: PMC7285326 DOI: 10.3390/microorganisms8050775] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/13/2020] [Accepted: 05/18/2020] [Indexed: 02/06/2023] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is a tri-segmented, tick-borne nairovirus that causes disease of ranging severity in humans. The CCHFV M segment encodes a complex glycoprotein precursor (GPC) that undergoes extensive endoproteolytic cleavage, giving rise to two structural proteins (Gn and Gc) required for virus attachment and entry, and to multiple non-structural proteins (NSm, GP160, GP85, and GP38). The functions of these non-structural proteins remain largely unclear. Here, we investigate the role of NSm during infection by generating a recombinant CCHFV lacking the complete NSm domain (10200∆NSm) and observing CCHFV ∆NSm replication in cell lines and pathogenicity in Ifnar-/- mice. Our data demonstrate that the NSm domain is dispensable for viral replication in vitro, and, despite the delayed onset of clinical signs, CCHFV lacking this domain caused severe or lethal disease in infected mice.
Collapse
|
22
|
Evolutionary Dynamics of Oropouche Virus in South America. J Virol 2020; 94:JVI.01127-19. [PMID: 31801869 PMCID: PMC7022353 DOI: 10.1128/jvi.01127-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/19/2019] [Indexed: 01/09/2023] Open
Abstract
The emergence and reemergence of pathogens such as Zika virus, chikungunya virus, and yellow fever virus have drawn attention toward other cocirculating arboviruses in South America. Oropouche virus (OROV) is a poorly studied pathogen responsible for over a dozen outbreaks since the early 1960s and represents a public health burden to countries such as Brazil, Panama, and Peru. OROV is likely underreported since its symptomatology can be easily confounded with other febrile illnesses (e.g., dengue fever and leptospirosis) and point-of-care testing for the virus is still uncommon. With limited data, there is a need to optimize the information currently available. Analysis of OROV genomes can help us understand how the virus circulates in nature and can reveal the evolutionary forces that shape the genetic diversity of the virus, which has implications for molecular diagnostics and the design of potential vaccines. The Amazon basin is home to numerous arthropod-borne viral pathogens that cause febrile disease in humans. Among these, Oropouche orthobunyavirus (OROV) is a relatively understudied member of the genus Orthobunyavirus, family Peribunyaviridae, that causes periodic outbreaks in human populations in Brazil and other South American countries. Although several studies have described the genetic diversity of the virus, the evolutionary processes that shape the OROV genome remain poorly understood. Here, we present a comprehensive study of the genomic dynamics of OROV that encompasses phylogenetic analysis, evolutionary rate estimates, inference of natural selective pressures, recombination and reassortment, and structural analysis of OROV variants. Our study includes all available published sequences, as well as a set of new OROV genome sequences obtained from patients in Ecuador, representing the first set of genomes from this country. Our results show differing evolutionary processes on the three segments that comprise the viral genome. We infer differing times of the most recent common ancestors of the genome segments and propose that this can be explained by cryptic reassortment. We also present the discovery of previously unobserved putative N-linked glycosylation sites, as well as codons that evolve under positive selection on the viral surface proteins, and discuss the potential role of these features in the evolution of OROV through a combined phylogenetic and structural approach. IMPORTANCE The emergence and reemergence of pathogens such as Zika virus, chikungunya virus, and yellow fever virus have drawn attention toward other cocirculating arboviruses in South America. Oropouche virus (OROV) is a poorly studied pathogen responsible for over a dozen outbreaks since the early 1960s and represents a public health burden to countries such as Brazil, Panama, and Peru. OROV is likely underreported since its symptomatology can be easily confounded with other febrile illnesses (e.g., dengue fever and leptospirosis) and point-of-care testing for the virus is still uncommon. With limited data, there is a need to optimize the information currently available. Analysis of OROV genomes can help us understand how the virus circulates in nature and can reveal the evolutionary forces that shape the genetic diversity of the virus, which has implications for molecular diagnostics and the design of potential vaccines.
Collapse
|
23
|
Sick F, Beer M, Kampen H, Wernike K. Culicoides Biting Midges-Underestimated Vectors for Arboviruses of Public Health and Veterinary Importance. Viruses 2019; 11:E376. [PMID: 31022868 PMCID: PMC6520762 DOI: 10.3390/v11040376] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/10/2019] [Accepted: 04/18/2019] [Indexed: 01/12/2023] Open
Abstract
Culicoides biting midges, small hematophagous dipterans, are the demonstrated or putative vectors of multiple arboviruses of veterinary and public health importance. Despite its relevance in disease spread, the ceratopogonid genus Culicoides is still a largely neglected group of species, predominantly because the major human-affecting arboviruses are considered to be transmitted by mosquitoes. However, when a pathogen is detected in a certain vector species, a thorough search for further vectors often remains undone and, therefore, the relevant vector species may remain unknown. Furthermore, for many hematophagous arthropods, true vector competence is often merely suspected and not experimentally proven. Therefore, we aim to illuminate the general impact of Culicoides biting midges and to summarize the knowledge about biting midge-borne disease agents using the order Bunyavirales, the largest and most diverse group of RNA viruses, as an example. When considering only viruses evidentially transmitted by Culicoides midges, the Simbu serogroup (genus Orthobunyavirus) is presumably the most important group within the virus order. Its members are of great veterinary importance, as a variety of simbuviruses, e.g., the species Akabane orthobunyavirus or Schmallenberg orthobunyavirus, induces severe congenital infections in pregnant animals. The major zoonotic representative of this serogroup occurs in South and Central America and causes the so-called Oropouche fever, an acute febrile illness in humans.
Collapse
Affiliation(s)
- Franziska Sick
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Suedufer 10, 17493 Greifswald-Insel Riems, Germany.
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Suedufer 10, 17493 Greifswald-Insel Riems, Germany.
| | - Helge Kampen
- Institute of Infectology, Friedrich-Loeffler-Institut, Suedufer 10, 17493 Greifswald-Insel Riems, Germany.
| | - Kerstin Wernike
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Suedufer 10, 17493 Greifswald-Insel Riems, Germany.
| |
Collapse
|
24
|
Interferon-Stimulated Gene (ISG)-Expression Screening Reveals the Specific Antibunyaviral Activity of ISG20. J Virol 2018; 92:JVI.02140-17. [PMID: 29695422 PMCID: PMC6002717 DOI: 10.1128/jvi.02140-17] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 04/14/2018] [Indexed: 11/20/2022] Open
Abstract
Bunyaviruses pose a significant threat to human health, prosperity, and food security. In response to viral infections, interferons (IFNs) upregulate the expression of hundreds of interferon-stimulated genes (ISGs), whose cumulative action can potently inhibit the replication of bunyaviruses. We used a flow cytometry-based method to screen the ability of ∼500 unique ISGs from humans and rhesus macaques to inhibit the replication of Bunyamwera orthobunyavirus (BUNV), the prototype of both the Peribunyaviridae family and the Bunyavirales order. Candidates possessing antibunyaviral activity were further examined using a panel of divergent bunyaviruses. Interestingly, one candidate, ISG20, exhibited potent antibunyaviral activity against most viruses examined from the Peribunyaviridae, Hantaviridae, and Nairoviridae families, whereas phleboviruses (Phenuiviridae) largely escaped inhibition. Similar to the case against other viruses known to be targeted by ISG20, the antibunyaviral activity of ISG20 is dependent upon its functional RNase activity. Through use of an infectious virus-like particle (VLP) assay (based on the BUNV minigenome system), we confirmed that gene expression from all 3 viral segments is strongly inhibited by ISG20. Using in vitro evolution, we generated a substantially ISG20-resistant BUNV and mapped the determinants of ISG20 sensitivity/resistance. Taking all the data together, we report that ISG20 is a broad and potent antibunyaviral factor but that some bunyaviruses are remarkably ISG20 resistant. Thus, ISG20 sensitivity/resistance may influence the pathogenesis of bunyaviruses, many of which are emerging viruses of clinical or veterinary significance. IMPORTANCE There are hundreds of bunyaviruses, many of which cause life-threatening acute diseases in humans and livestock. The interferon (IFN) system is a key component of innate immunity, and type I IFNs limit bunyaviral propagation both in vitro and in vivo. Type I IFN signaling results in the upregulation of hundreds of IFN-stimulated genes (ISGs), whose concerted action generates an “antiviral state.” Although IFNs are critical in limiting bunyaviral replication and pathogenesis, much is still unknown about which ISGs inhibit bunyaviruses. Using ISG-expression screening, we examined the ability of ∼500 unique ISGs to inhibit Bunyamwera orthobunyavirus (BUNV), the prototypical bunyavirus. Using this approach, we identified ISG20, an interferon-stimulated exonuclease, as a potent inhibitor of BUNV. Interestingly, ISG20 possesses highly selective antibunyaviral activity, with multiple bunyaviruses being potently inhibited while some largely escape inhibition. We speculate that the ability of some bunyaviruses to escape ISG20 may influence their pathogenesis.
Collapse
|
25
|
Sun Y, Li J, Gao GF, Tien P, Liu W. Bunyavirales ribonucleoproteins: the viral replication and transcription machinery. Crit Rev Microbiol 2018. [PMID: 29516765 DOI: 10.1080/1040841x.2018.1446901] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The Bunyavirales order is one of the largest groups of segmented negative-sense single-stranded RNA viruses, which includes many pathogenic strains that cause severe human diseases. The RNA segments of the bunyavirus genome are separately encapsidated by multiple copies of nucleoprotein (N), and both termini of each N-encapsidated genomic RNA segment bind to one copy of the viral L polymerase protein. The viral genomic RNA, N and L protein together form the ribonucleoprotein (RNP) complex that constitutes the molecular machinery for viral genome replication and transcription. Recently, breakthroughs have been achieved in understanding the architecture of bunyavirus RNPs with the determination of the atomic structures of the N and L proteins from various members of this order. In this review, we discuss the structures and functions of these bunyavirus RNP components, as well as viral genome replication and transcription mechanisms.
Collapse
Affiliation(s)
- Yeping Sun
- a CAS Key Laboratory of Pathogenic Microbiology and Immunology , Institute of Microbiology, Chinese Academy of Sciences , Beijing , China
| | - Jing Li
- a CAS Key Laboratory of Pathogenic Microbiology and Immunology , Institute of Microbiology, Chinese Academy of Sciences , Beijing , China
| | - George F Gao
- a CAS Key Laboratory of Pathogenic Microbiology and Immunology , Institute of Microbiology, Chinese Academy of Sciences , Beijing , China.,b National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , China
| | - Po Tien
- a CAS Key Laboratory of Pathogenic Microbiology and Immunology , Institute of Microbiology, Chinese Academy of Sciences , Beijing , China
| | - Wenjun Liu
- a CAS Key Laboratory of Pathogenic Microbiology and Immunology , Institute of Microbiology, Chinese Academy of Sciences , Beijing , China
| |
Collapse
|
26
|
de Melo AB, de Souza WM, Acrani GO, Carvalho VL, Romeiro MF, Tolardo AL, da Silva SP, Cardoso JF, de Oliveira Chiang J, da Silva Gonçalves Vianez JL, do Socorro da Silva Azevedo R, Figueiredo LTM, da Costa Vasconcelos PF, Nunes MRT, de Almeida Medeiros DB. Genomic characterization and evolution of Tacaiuma orthobunyavirus (Peribunyaviridae family) isolated in Brazil. INFECTION GENETICS AND EVOLUTION 2018; 60:71-76. [PMID: 29476812 DOI: 10.1016/j.meegid.2018.02.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 02/02/2018] [Accepted: 02/18/2018] [Indexed: 11/25/2022]
Abstract
Tacaiuma virus (TCMV) is antigenically characterized as a member of the Anopheles A complex in the Orthobunyavirus genus, Peribunyaviridae family (Bunyavirales order). Clinically, the TCMV infection is characterized by acute febrile illness with myalgia and arthralgia lasting three to five days. However, the genomic and evolutionary aspect of this virus has not been elucidated. In this study, we described the complete coding sequences of three segments of two TCMV strains isolated in Brazil and three complete coding sequences of the small segment of three TCMV strains. All the strains sequenced in this study showed the typical genomic organization of orthobunyaviruses that infect vertebrates, except for the absence of the open reading frame that encodes the well-described non-structural small protein. This study presents the genomic and evolutionary characterization of TCMV strains and would be helpful for diagnostic purposes and epidemiology.
Collapse
Affiliation(s)
- Aristides Bezerra de Melo
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ministry of Health, Ananindeua, Pará, Brazil
| | - William Marciel de Souza
- Virology Research Center, School of Medicine of Ribeirão Preto of University of São Paulo, Ribeirão Preto, SP, Brazil.
| | | | - Valéria Lima Carvalho
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ministry of Health, Ananindeua, Pará, Brazil
| | - Marilia Farignoli Romeiro
- Virology Research Center, School of Medicine of Ribeirão Preto of University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Aline Lavado Tolardo
- Virology Research Center, School of Medicine of Ribeirão Preto of University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Sandro Patroca da Silva
- Center for Technological Innovations, Evandro Chagas Institute, Ministry of Health, Ananindeua, Pará, Brazil
| | - Jedson Ferreira Cardoso
- Center for Technological Innovations, Evandro Chagas Institute, Ministry of Health, Ananindeua, Pará, Brazil
| | - Jannifer de Oliveira Chiang
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ministry of Health, Ananindeua, Pará, Brazil
| | | | | | - Luiz Tadeu Moraes Figueiredo
- Virology Research Center, School of Medicine of Ribeirão Preto of University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | | | | |
Collapse
|
27
|
Chiang JO, Marciel de Souza W, Teixeira Nunes MR, Acrani GO, Paes de Andrade Travassos da Rosa A, Mesquita de Freitas N, Patroca da Silva S, Dorta da Silva PH, Watanabe de Sousa A, Rodrigues SG, Simões Quaresma JA, Dutary B, Guzmán H, Vasilakis N, Tesh RB, Fernando da Costa Vasconcelos P. Characterization of the Gamboa Virus Serogroup ( Orthobunyavirus Genus, Peribunyaviridae Family). Am J Trop Med Hyg 2018; 98:1502-1511. [PMID: 29436340 DOI: 10.4269/ajtmh.17-0810] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Comprehensive comparative phylogenetic analyses were performed on 17 Gamboa serogroup viruses (GAMSVs) from distinct geographic regions in the Americas and other representative members of the genus Orthobunyavirus (Peribunyaviridae), based on small (S), medium (M), and large (L) open reading frame full-length and partial sequences. Genome characterization showed that the GAMSVs divide into four clades or genotypes. The GAMSVs have a genetic organization similar to other orthobunyaviruses, except that they have a larger NSm protein than other orthobunyaviruses. A serosurvey for Gamboa virus antibodies was performed in plasma from birds, other wild animals, and humans living around the Tucuruí hydroelectric dam in Pará state, northern Brazil, a known focus of GAMSV activity. Newborn chicks (Gallus gallus domesticus) were experimentally infected with a GAMSV, and the pathogenesis is described. Histopathological changes were primarily in the lungs and liver. Also, a review of the ecology of the GAMSVs in the Americas is included. In sum, this study presents the genomic and evolutionary characterization of the Gamboa group and the potential model of pathogenesis, which would be helpful for diagnostic purposes, epidemiology, and immunopathogenesis studies.
Collapse
Affiliation(s)
- Jannifer Oliveira Chiang
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ministry of Health, Ananindeua, Brazil
| | - William Marciel de Souza
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom.,Virology Research Center, School of Medicine of Ribeirão Preto of University of São Paulo, Ribeirão Preto, Brazil
| | - Márcio Roberto Teixeira Nunes
- Department of Pathology and Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas.,Center for Technological Innovations, Evandro Chagas Institute, Ministry of Health, Ananindeua, Brazil
| | | | | | | | - Sandro Patroca da Silva
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ministry of Health, Ananindeua, Brazil
| | | | - Alana Watanabe de Sousa
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ministry of Health, Ananindeua, Brazil
| | - Sueli Guerreiro Rodrigues
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ministry of Health, Ananindeua, Brazil
| | | | - Bedsy Dutary
- Gorgas Memorial Institute of Health Studies, Panama City, Panama
| | - Hilda Guzmán
- Department of Pathology and Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas
| | - Nikos Vasilakis
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas.,Center for Tropical Diseases, University of Texas Medical Branch, Galveston, Texas.,Department of Pathology and Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas
| | - Robert B Tesh
- Department of Pathology and Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas
| | | |
Collapse
|
28
|
Kraatz F, Wernike K, Reiche S, Aebischer A, Reimann I, Beer M. Schmallenberg virus non-structural protein NSm: Intracellular distribution and role of non-hydrophobic domains. Virology 2018; 516:46-54. [PMID: 29329078 DOI: 10.1016/j.virol.2017.12.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 12/20/2017] [Accepted: 12/28/2017] [Indexed: 12/20/2022]
Abstract
Schmallenberg virus (SBV) induces fetal malformation, abortions and stillbirth in ruminants. While the non-structural protein NSs is a major virulence factor, the biological function of NSm, the second non-structural protein which consists of three hydrophobic transmembrane (I, III, V) and two non-hydrophobic regions (II, IV), is still unknown. Here, a series of NSm mutants displaying deletions of nearly the entire NSm or of the non-hydrophobic domains was generated and the intracellular distribution of NSm was assessed. SBV-NSm is dispensable for the generation of infectious virus and mutants lacking domains II - V showed growth properties similar to the wild-type virus. In addition, a comparable intracellular distribution of SBV-NSm was observed in mammalian cells infected with domain II mutants or wild-type virus. In both cases, NSm co-localized with the glycoprotein Gc in the Golgi compartment. However, domain IV-deletion mutants showed an altered distribution pattern and no co-localization of NSm and Gc.
Collapse
Affiliation(s)
- Franziska Kraatz
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald, Insel Riems, Germany
| | - Kerstin Wernike
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald, Insel Riems, Germany
| | - Sven Reiche
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald, Insel Riems, Germany
| | - Andrea Aebischer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald, Insel Riems, Germany
| | - Ilona Reimann
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald, Insel Riems, Germany
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald, Insel Riems, Germany.
| |
Collapse
|
29
|
Ferron F, Weber F, de la Torre JC, Reguera J. Transcription and replication mechanisms of Bunyaviridae and Arenaviridae L proteins. Virus Res 2017; 234:118-134. [PMID: 28137457 PMCID: PMC7114536 DOI: 10.1016/j.virusres.2017.01.018] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/17/2017] [Accepted: 01/21/2017] [Indexed: 12/15/2022]
Abstract
Bunyavirus and arenavirus are important public health threats. Bunyavirus and arenavirus molecular biology, common and differential features. Implications of LACV L protein structure for understanding viral RNA synthesis. Current state and future perspectives on bunya- and arenavirus antivirals.
Bunyaviridae and Arenaviridae virus families include an important number of highly pathogenic viruses for humans. They are enveloped viruses with negative stranded RNA genomes divided into three (bunyaviruses) or two (arenaviruses) segments. Each genome segment is coated by the viral nucleoproteins (NPs) and the polymerase (L protein) to form a functional ribonucleoprotein (RNP) complex. The viral RNP provides the necessary context on which the L protein carries out the biosynthetic processes of RNA replication and gene transcription. Decades of research have provided a good understanding of the molecular processes underlying RNA synthesis, both RNA replication and gene transcription, for these two families of viruses. In this review we will provide a global view of the common features, as well as differences, of the molecular biology of Bunyaviridae and Arenaviridae. We will also describe structures of protein and protein-RNA complexes so far determined for these viral families, mainly focusing on the L protein, and discuss their implications for understanding the mechanisms of viral RNA replication and gene transcription within the architecture of viral RNPs, also taking into account the cellular context in which these processes occur. Finally, we will discuss the implications of these structural findings for the development of antiviral drugs to treat human diseases caused by members of the Bunyaviridae and Arenaviridae families.
Collapse
Affiliation(s)
- François Ferron
- Aix-Marseille Université, AFMB UMR 7257, 13288 Marseille, France; CNRS, AFMB UMR 7257, 13288 Marseille, France
| | - Friedemann Weber
- Institute for Virology, FB10-Veterinary Medicine, Justus-Liebig University, D-35392 Giessen, Germany
| | | | - Juan Reguera
- Aix-Marseille Université, AFMB UMR 7257, 13288 Marseille, France; CNRS, AFMB UMR 7257, 13288 Marseille, France; INSERM, AFMB UMR 7257, 13288 Marseille, France.
| |
Collapse
|
30
|
The Antiviral RNAi Response in Vector and Non-vector Cells against Orthobunyaviruses. PLoS Negl Trop Dis 2017; 11:e0005272. [PMID: 28060823 PMCID: PMC5245901 DOI: 10.1371/journal.pntd.0005272] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 01/19/2017] [Accepted: 12/20/2016] [Indexed: 11/19/2022] Open
Abstract
Background Vector arthropods control arbovirus replication and spread through antiviral innate immune responses including RNA interference (RNAi) pathways. Arbovirus infections have been shown to induce the exogenous small interfering RNA (siRNA) and Piwi-interacting RNA (piRNA) pathways, but direct antiviral activity by these host responses in mosquito cells has only been demonstrated against a limited number of positive-strand RNA arboviruses. For bunyaviruses in general, the relative contribution of small RNA pathways in antiviral defences is unknown. Methodology/Principal Findings The genus Orthobunyavirus in the Bunyaviridae family harbours a diverse range of mosquito-, midge- and tick-borne arboviruses. We hypothesized that differences in the antiviral RNAi response in vector versus non-vector cells may exist and that could influence viral host range. Using Aedes aegypti-derived mosquito cells, mosquito-borne orthobunyaviruses and midge-borne orthobunyaviruses we showed that bunyavirus infection commonly induced the production of small RNAs and the effects of the small RNA pathways on individual viruses differ in specific vector-arbovirus interactions. Conclusions/Significance These findings have important implications for our understanding of antiviral RNAi pathways and orthobunyavirus-vector interactions and tropism. A number of orthobunyaviruses such as Oropouche virus, La Crosse virus and Schmallenberg virus are important global human or animal pathogens transmitted by arthropod vectors. Further understanding of the antiviral control mechanisms in arthropod vectors is key to developing novel prevention strategies based on preventing transmission. Antiviral small RNA pathways such as the exogenous siRNA and piRNA pathways have been shown to mediate antiviral activity against positive-strand RNA arboviruses, but information about their activities against negative-strand RNA arboviruses is critically lacking. Here we show that in Aedes aegypti-derived mosquito cells, the antiviral responses to mosquito-borne orthobunyaviruses is largely mediated by both siRNA and piRNA pathways, whereas the piRNA pathway plays only a minor role in controlling midge-borne orthobunyaviruses. This suggests that vector specificity is in part controlled by antiviral responses that depend on the host species. These findings contribute significantly to our understanding of arbovirus-vector interactions.
Collapse
|
31
|
Briese T, Williams DT, Kapoor V, Diviney SM, Certoma A, Wang J, Johansen CA, Chowdhary R, Mackenzie JS, Lipkin WI. Analysis of Arbovirus Isolates from Australia Identifies Novel Bunyaviruses Including a Mapputta Group Virus from Western Australia That Links Gan Gan and Maprik Viruses. PLoS One 2016; 11:e0164868. [PMID: 27764175 PMCID: PMC5072647 DOI: 10.1371/journal.pone.0164868] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 10/03/2016] [Indexed: 01/01/2023] Open
Abstract
The Mapputta group comprises antigenically related viruses indigenous to Australia and Papua New Guinea that are included in the family Bunyaviridae but not currently assigned to a specific genus. We determined and analyzed the genome sequences of five Australian viruses isolated from mosquitoes collected during routine arbovirus surveillance in Western Australia (K10441, SW27571, K13190, and K42904) and New South Wales (12005). Based on matching sequences of all three genome segments to prototype MRM3630 of Trubanaman virus (TRUV), NB6057 of Gan Gan virus (GGV), and MK7532 of Maprik virus (MPKV), isolates K13190 and SW27571 were identified as TRUV, 12005 as GGV, and K42904 as a Mapputta group virus from Western Australia linking GGV and MPKV. The results confirmed serum neutralization data that had linked SW27571 to TRUV. The fifth virus, K10441 from Willare, was most closely related to Batai orthobunyavirus, presumably representing an Australian variant of the virus. Phylogenetic analysis also confirmed the close relationship of our TRUV and GGV isolates to two other recently described Australian viruses, Murrumbidgee virus and Salt Ash virus, respectively. Our findings indicate that TRUV has a wide circulation throughout the Australian continent, demonstrating for the first time its presence in Western Australia. Similarly, the presence of a virus related to GGV, which had been linked to human disease and previously known only from the Australian southeast, was demonstrated in Western Australia. Finally, a Batai virus isolate was identified in Western Australia. The expanding availability of genomic sequence for novel Australian bunyavirus variants supports the identification of suitably conserved or diverse primer-binding target regions to establish group-wide as well as virus-specific nucleic acid tests in support of specific diagnostic and surveillance efforts throughout Australasia.
Collapse
Affiliation(s)
- Thomas Briese
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, New York, United States of America
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, United States of America
- * E-mail: (TB); (DTW)
| | - David T. Williams
- CSIRO, Australian Animal Health Laboratory, Geelong, Victoria, Australia
- School of Biomedical Sciences, Curtin University, Perth, Western Australia, Australia
- * E-mail: (TB); (DTW)
| | - Vishal Kapoor
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, New York, United States of America
| | - Sinead M. Diviney
- School of Biomedical Sciences, Curtin University, Perth, Western Australia, Australia
| | - Andrea Certoma
- CSIRO, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Jianning Wang
- CSIRO, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Cheryl A. Johansen
- The Arbovirus Surveillance and Research Laboratory, University of Western Australia, Nedlands, Western Australia, Australia
| | - Rashmi Chowdhary
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, New York, United States of America
| | - John S. Mackenzie
- Faculty of Health Sciences, Curtin University, Perth, Western Australia, Australia
| | - W. Ian Lipkin
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, New York, United States of America
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, United States of America
| |
Collapse
|
32
|
Bunyamwera orthobunyavirus glycoprotein precursor is processed by cellular signal peptidase and signal peptide peptidase. Proc Natl Acad Sci U S A 2016; 113:8825-30. [PMID: 27439867 DOI: 10.1073/pnas.1603364113] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The M genome segment of Bunyamwera virus (BUNV)-the prototype of both the Bunyaviridae family and the Orthobunyavirus genus-encodes the glycoprotein precursor (GPC) that is proteolytically cleaved to yield two viral structural glycoproteins, Gn and Gc, and a nonstructural protein, NSm. The cleavage mechanism of orthobunyavirus GPCs and the host proteases involved have not been clarified. In this study, we investigated the processing of BUNV GPC and found that both NSm and Gc proteins were cleaved at their own internal signal peptides (SPs), in which NSm domain I functions as SP(NSm) and NSm domain V as SP(Gc) Moreover, the domain I was further processed by a host intramembrane-cleaving protease, signal peptide peptidase, and is required for cell fusion activities. Meanwhile, the NSm domain V (SP(Gc)) remains integral to NSm, rendering the NSm topology as a two-membrane-spanning integral membrane protein. We defined the cleavage sites and boundaries between the processed proteins as follows: Gn, from residue 17-312 or nearby residues; NSm, 332-477; and Gc, 478-1433. Our data clarified the mechanism of the precursor cleavage process, which is important for our understanding of viral glycoprotein biogenesis in the genus Orthobunyavirus and thus presents a useful target for intervention strategies.
Collapse
|
33
|
Gibrail MM, Fiaccadori FS, Souza M, Almeida TNV, Chiang JO, Martins LC, Ferreira MS, Cardoso DDDDP. Detection of antibodies to Oropouche virus in non-human primates in Goiânia City, Goiás. Rev Soc Bras Med Trop 2016; 49:357-60. [DOI: 10.1590/0037-8682-0425-2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 04/05/2016] [Indexed: 11/22/2022] Open
|
34
|
Varela M, Pinto RM, Caporale M, Piras IM, Taggart A, Seehusen F, Hahn K, Janowicz A, de Souza WM, Baumgärtner W, Shi X, Palmarini M. Mutations in the Schmallenberg Virus Gc Glycoprotein Facilitate Cellular Protein Synthesis Shutoff and Restore Pathogenicity of NSs Deletion Mutants in Mice. J Virol 2016; 90:5440-5450. [PMID: 26984728 PMCID: PMC4934738 DOI: 10.1128/jvi.00424-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 03/15/2016] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Serial passage of viruses in cell culture has been traditionally used to attenuate virulence and identify determinants of viral pathogenesis. In a previous study, we found that a strain of Schmallenberg virus (SBV) serially passaged in tissue culture (termed SBVp32) unexpectedly displayed increased pathogenicity in suckling mice compared to wild-type SBV. In this study, we mapped the determinants of SBVp32 virulence to the viral genome M segment. SBVp32 virulence is associated with the capacity of this virus to reach high titers in the brains of experimentally infected suckling mice. We also found that the Gc glycoprotein, encoded by the M segment of SBVp32, facilitates host cell protein shutoff in vitro Interestingly, while the M segment of SBVp32 is a virulence factor, we found that the S segment of the same virus confers by itself an attenuated phenotype to wild-type SBV, as it has lost the ability to block the innate immune system of the host. Single mutations present in the Gc glycoprotein of SBVp32 are sufficient to compensate for both the attenuated phenotype of the SBVp32 S segment and the attenuated phenotype of NSs deletion mutants. Our data also indicate that the SBVp32 M segment does not act as an interferon (IFN) antagonist. Therefore, SBV mutants can retain pathogenicity even when they are unable to fully control the production of IFN by infected cells. Overall, this study suggests that the viral glycoprotein of orthobunyaviruses can compensate, at least in part, for the function of NSs. In addition, we also provide evidence that the induction of total cellular protein shutoff by SBV is determined by multiple viral proteins, while the ability to control the production of IFN maps to the NSs protein. IMPORTANCE The identification of viral determinants of pathogenesis is key to the development of prophylactic and intervention measures. In this study, we found that the bunyavirus Gc glycoprotein is a virulence factor. Importantly, we show that mutations in the Gc glycoprotein can restore the pathogenicity of attenuated mutants resulting from deletions or mutations in the nonstructural protein NSs. Our findings highlight the fact that careful consideration should be taken when designing live attenuated vaccines based on deletions of nonstructural proteins since single mutations in the viral glycoproteins appear to revert attenuated mutants to virulent phenotypes.
Collapse
Affiliation(s)
- Mariana Varela
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - Rute Maria Pinto
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - Marco Caporale
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise G. Caporale, Teramo, Italy
| | - Ilaria M Piras
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - Aislynn Taggart
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - Frauke Seehusen
- Department of Pathology and Center of Systems Neuroscience, University of Veterinary Medicine, Hannover, Germany
| | - Kerstin Hahn
- Department of Pathology and Center of Systems Neuroscience, University of Veterinary Medicine, Hannover, Germany
| | - Anna Janowicz
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - William Marciel de Souza
- Virology Research Center, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirao Preto, Brazil
| | - Wolfgang Baumgärtner
- Department of Pathology and Center of Systems Neuroscience, University of Veterinary Medicine, Hannover, Germany
| | - Xiaohong Shi
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - Massimo Palmarini
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| |
Collapse
|
35
|
Ishihara Y, Shioda C, Bangphoomi N, Sugiura K, Saeki K, Tsuda S, Iwanaga T, Takenaka-Uema A, Kato K, Murakami S, Uchida K, Akashi H, Horimoto T. Akabane virus nonstructural protein NSm regulates viral growth and pathogenicity in a mouse model. J Vet Med Sci 2016; 78:1391-1397. [PMID: 27181086 PMCID: PMC5059365 DOI: 10.1292/jvms.16-0140] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The biological function of a nonstructural protein, NSm, of Akabane virus (AKAV) is
unknown. In this study, we generated a series of NSm deletion mutant viruses by reverse
genetics and compared their phenotypes. The mutant in which the NSm coding region was
almost completely deleted could not be rescued, suggesting that NSm plays a role in virus
replication. We next generated mutant viruses possessing various partial deletions in NSm
and identified several regions critical for virus infectivity. All rescued mutant viruses
produced smaller plaques and grew inefficiently in cell culture, compared to the wild-type
virus. Interestingly, although the pathogenicity of NSm deletion mutant viruses varied in
mice depending on their deletion regions and sizes, more than half the mice died following
infection with any mutant virus and the dead mice exhibited encephalitis as in wild-type
virus-inoculated mice, indicating their neuroinvasiveness. Abundant viral antigens were
detected in the brain tissues of dead mice, whereas appreciable antigen was not observed
in those of surviving mice, suggesting a correlation between virus growth rate in the
brain and neuropathogenicity in mice. We conclude that NSm affects AKAV replication
in vitro as well as in vivo and that it may function
as a virulence factor.
Collapse
Affiliation(s)
- Yukari Ishihara
- Department of Veterinary Microbiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Broce S, Hensley L, Sato T, Lehrer-Graiwer J, Essrich C, Edwards KJ, Pajda J, Davis CJ, Bhadresh R, Hurt CR, Freeman B, Lingappa VR, Kelleher CA, Karpuj MV. Biochemical and biophysical characterization of cell-free synthesized Rift Valley fever virus nucleoprotein capsids enables in vitro screening to identify novel antivirals. Biol Direct 2016; 11:25. [PMID: 27179769 PMCID: PMC4867995 DOI: 10.1186/s13062-016-0126-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/29/2016] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Viral capsid assembly involves the oligomerization of the capsid nucleoprotein (NP), which is an essential step in viral replication and may represent a potential antiviral target. An in vitro transcription-translation reaction using a wheat germ (WG) extract in combination with a sandwich ELISA assay has recently been used to identify small molecules with antiviral activity against the rabies virus. RESULTS Here, we examined the application of this system to viruses with capsids with a different structure, such as the Rift Valley fever virus (RVFV), the etiological agent of a severe emerging infectious disease. The biochemical and immunological characterization of the in vitro-generated RVFV NP assembly products enabled the distinction between intermediately and highly ordered capsid structures. This distinction was used to establish a screening method for the identification of potential antiviral drugs for RVFV countermeasures. CONCLUSIONS These results indicated that this unique analytical system, which combines nucleoprotein oligomerization with the specific immune recognition of a highly ordered capsid structure, can be extended to various viral families and used both to study the early stages of NP assembly and to assist in the identification of potential antiviral drugs in a cost-efficient manner. REVIEWERS Reviewed by Jeffry Skolnick and Noah Isakov. For the full reviews please go to the Reviewers' comments section.
Collapse
Affiliation(s)
- Sean Broce
- Prosetta Antiviral Inc, San Francisco, CA, USA
| | | | - Tomoharu Sato
- Prosetta Antiviral Inc, San Francisco, CA, USA
- DuPont Industrial Biosciences, Palo Alto, CA, USA
| | - Joshua Lehrer-Graiwer
- Prosetta Antiviral Inc, San Francisco, CA, USA
- Global Blood Therapeutics, Inc, Palo Alto, CA, USA
| | | | | | | | - Christopher J Davis
- Biomedical Advanced Research & Development Authority, Office of the Assistant Secretary of Preparedness & Response, U.S. Department of Health & Human Services, Washington, DC, USA
| | - Rami Bhadresh
- Prosetta Antiviral Inc, San Francisco, CA, USA
- Biocon Bristol-Myers Squibb, Syngene International Ltd, Bangalore, India
| | | | | | | | - Colm A Kelleher
- Prosetta Antiviral Inc, San Francisco, CA, USA
- Bigelow Aerospace Advanced Space Studies, Las Vegas, NV, USA
| | - Marcela V Karpuj
- Prosetta Antiviral Inc, San Francisco, CA, USA.
- CUBRC, Inc, Buffalo, NY, USA.
- BioA2Z, Inc, San Francisco, CA, USA.
- Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold, Safed, 1311502, Israel.
| |
Collapse
|
37
|
Generation of Recombinant Oropouche Viruses Lacking the Nonstructural Protein NSm or NSs. J Virol 2015; 90:2616-27. [PMID: 26699638 DOI: 10.1128/jvi.02849-15] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/15/2015] [Indexed: 01/15/2023] Open
Abstract
UNLABELLED Oropouche virus (OROV) is a midge-borne human pathogen with a geographic distribution in South America. OROV was first isolated in 1955, and since then, it has been known to cause recurring outbreaks of a dengue-like illness in the Amazonian regions of Brazil. OROV, however, remains one of the most poorly understood emerging viral zoonoses. Here we describe the successful recovery of infectious OROV entirely from cDNA copies of its genome and generation of OROV mutant viruses lacking either the NSm or the NSs coding region. Characterization of the recombinant viruses carried out in vitro demonstrated that the NSs protein of OROV is an interferon (IFN) antagonist as in other NSs-encoding bunyaviruses. Additionally, we demonstrate the importance of the nine C-terminal amino acids of OROV NSs in IFN antagonistic activity. OROV was also found to be sensitive to IFN-α when cells were pretreated; however, the virus was still capable of replicating at doses as high as 10,000 U/ml of IFN-α, in contrast to the family prototype BUNV. We found that OROV lacking the NSm protein displayed characteristics similar to those of the wild-type virus, suggesting that the NSm protein is dispensable for virus replication in the mammalian and mosquito cell lines that were tested. IMPORTANCE Oropouche virus (OROV) is a public health threat in Central and South America, where it causes periodic outbreaks of dengue-like illness. In Brazil, OROV is the second most frequent cause of arboviral febrile illness after dengue virus, and with the current rates of urban expansion, more cases of this emerging viral zoonosis could occur. To better understand the molecular biology of OROV, we have successfully rescued the virus along with mutants. We have established that the C terminus of the NSs protein is important in interferon antagonism and that the NSm protein is dispensable for virus replication in cell culture. The tools described in this paper are important in terms of understanding this important yet neglected human pathogen.
Collapse
|
38
|
Walker PJ, Widen SG, Firth C, Blasdell KR, Wood TG, Travassos da Rosa APA, Guzman H, Tesh RB, Vasilakis N. Genomic Characterization of Yogue, Kasokero, Issyk-Kul, Keterah, Gossas, and Thiafora Viruses: Nairoviruses Naturally Infecting Bats, Shrews, and Ticks. Am J Trop Med Hyg 2015; 93:1041-51. [PMID: 26324724 DOI: 10.4269/ajtmh.15-0344] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 06/29/2015] [Indexed: 11/07/2022] Open
Abstract
The genus Nairovirus of arthropod-borne bunyaviruses includes the important emerging human pathogen, Crimean-Congo hemorrhagic fever virus (CCHFV), as well as Nairobi sheep disease virus and many other poorly described viruses isolated from mammals, birds, and ticks. Here, we report genome sequence analysis of six nairoviruses: Thiafora virus (TFAV) that was isolated from a shrew in Senegal; Yogue (YOGV), Kasokero (KKOV), and Gossas (GOSV) viruses isolated from bats in Senegal and Uganda; Issyk-Kul virus (IKV) isolated from bats in Kyrgyzstan; and Keterah virus (KTRV) isolated from ticks infesting a bat in Malaysia. The S, M, and L genome segments of each virus were found to encode proteins corresponding to the nucleoprotein, polyglycoprotein, and polymerase protein of CCHFV. However, as observed in Leopards Hill virus (LPHV) and Erve virus (ERVV), polyglycoproteins encoded in the M segment lack sequences encoding the double-membrane-spanning CCHFV NSm protein. Amino acid sequence identities, complement-fixation tests, and phylogenetic analysis indicated that these viruses cluster into three groups comprising KKOV, YOGV, and LPHV from bats of the suborder Yingochiroptera; KTRV, IKV, and GOSV from bats of the suborder Yangochiroptera; and TFAV and ERVV from shrews (Soricomorpha: Soricidae). This reflects clade-specific host and vector associations that extend across the genus.
Collapse
Affiliation(s)
- Peter J Walker
- CSIRO Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas; Center for Biodefense and Emerging Infectious Diseases, Department of Pathology, The University of Texas Medical Branch, Galveston, Texas; Center for Tropical Diseases, Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas
| | - Steven G Widen
- CSIRO Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas; Center for Biodefense and Emerging Infectious Diseases, Department of Pathology, The University of Texas Medical Branch, Galveston, Texas; Center for Tropical Diseases, Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas
| | - Cadhla Firth
- CSIRO Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas; Center for Biodefense and Emerging Infectious Diseases, Department of Pathology, The University of Texas Medical Branch, Galveston, Texas; Center for Tropical Diseases, Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas
| | - Kim R Blasdell
- CSIRO Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas; Center for Biodefense and Emerging Infectious Diseases, Department of Pathology, The University of Texas Medical Branch, Galveston, Texas; Center for Tropical Diseases, Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas
| | - Thomas G Wood
- CSIRO Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas; Center for Biodefense and Emerging Infectious Diseases, Department of Pathology, The University of Texas Medical Branch, Galveston, Texas; Center for Tropical Diseases, Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas
| | - Amelia P A Travassos da Rosa
- CSIRO Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas; Center for Biodefense and Emerging Infectious Diseases, Department of Pathology, The University of Texas Medical Branch, Galveston, Texas; Center for Tropical Diseases, Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas
| | - Hilda Guzman
- CSIRO Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas; Center for Biodefense and Emerging Infectious Diseases, Department of Pathology, The University of Texas Medical Branch, Galveston, Texas; Center for Tropical Diseases, Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas
| | - Robert B Tesh
- CSIRO Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas; Center for Biodefense and Emerging Infectious Diseases, Department of Pathology, The University of Texas Medical Branch, Galveston, Texas; Center for Tropical Diseases, Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas
| | - Nikos Vasilakis
- CSIRO Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia; Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas; Center for Biodefense and Emerging Infectious Diseases, Department of Pathology, The University of Texas Medical Branch, Galveston, Texas; Center for Tropical Diseases, Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
39
|
Léger P, Lozach PY. Bunyaviruses: from transmission by arthropods to virus entry into the mammalian host first-target cells. Future Virol 2015. [DOI: 10.2217/fvl.15.52] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The Bunyaviridae constitute a large family of animal RNA viruses distributed worldwide, most members of which are transmitted to vertebrate hosts by arthropods and can cause severe pathologies in humans and livestock. With an increasing number of outbreaks, arthropod-borne bunyaviruses (arbo-bunyaviruses) present a global threat to public health and agricultural productivity. Yet transmission, tropism, receptors and cell entry remain poorly characterized. The focus of this review is on the initial infection of mammalian hosts by arbo-bunyaviruses from cellular and molecular perspectives, with particular attention to the human host. We address current knowledge and advances regarding the identity of the first-target cells and the subsequent processes of entry and penetration into the cytosol. Aspects of the vector-to-host switch that influence the early steps of cell infection in mammalian skin, where incoming particles are introduced by infected arthropods, are also highlighted and discussed.
Collapse
Affiliation(s)
- Psylvia Léger
- CellNetworks – Cluster of Excellence & Department of Infectious Diseases, Virology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Pierre-Yves Lozach
- CellNetworks – Cluster of Excellence & Department of Infectious Diseases, Virology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| |
Collapse
|
40
|
Abstract
The taxonomic group of Orthobunyaviruses is gaining increased attention, as several emerging members are causing devastating illnesses among humans and livestock. These viruses are transmitted to mammals by arthropods (mostly mosquitoes) during the blood meal. The nature of their genomic RNA predisposes orthobunyaviruses for eliciting a strong innate immune response mediated by pathogen recognition receptors (PRRs), especially the cytoplasmic RIG-I. However, the PRR responses are in fact disabled by the viral non-structural protein NSs. NSs imposes a strong block of cellular gene expression by inhibiting elongating RNA polymerase II. In this review, we will give an overview on the current state of knowledge regarding the interactions between orthobunyaviruses, the PRR axis, and NSs.
Collapse
Affiliation(s)
- Andreas Schoen
- Institute for Virology, Philipps-University Marburg, D-35043 Marburg, Germany
| | - Friedemann Weber
- Institute for Virology, Philipps-University Marburg, D-35043 Marburg, Germany.
| |
Collapse
|
41
|
Pijlman GP. Enveloped virus-like particles as vaccines against pathogenic arboviruses. Biotechnol J 2015; 10:659-70. [DOI: 10.1002/biot.201400427] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 11/27/2014] [Accepted: 12/22/2014] [Indexed: 12/26/2022]
|
42
|
Deletion mutants of Schmallenberg virus are avirulent and protect from virus challenge. J Virol 2014; 89:1825-37. [PMID: 25410877 DOI: 10.1128/jvi.02729-14] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
UNLABELLED Since its emergence, Schmallenberg virus (SBV), a novel insect-transmitted orthobunyavirus which predominantly infects ruminants, has caused a large epidemic in European livestock. Newly developed inactivated vaccines are available, but highly efficacious and safe live vaccines are still not available. Here, the properties of novel recombinant SBV mutants lacking the nonstructural protein NSs (rSBVΔNSs) or NSm (rSBVΔNSm) or both of these proteins (rSBVΔNSs/ΔNSm) were tested in vitro and in vivo in type I interferon receptor knockout mice (IFNAR(-/-)) and in a vaccination/challenge trial in cattle. As for other bunyaviruses, both nonstructural proteins of SBV are not essential for viral growth in vitro. In interferon-defective BHK-21 cells, rSBVΔNSs and rSBVΔNSm replicated to levels comparable to that of the parental rSBV; the double mutant virus, however, showed a mild growth defect, resulting in lower final virus titers. Additionally, both mutants with an NSs deletion induced high levels of interferon and showed a marked growth defect in interferon-competent sheep SFT-R cells. Nevertheless, in IFNAR(-/-) mice, all mutants were virulent, with the highest mortality rate for rSBVΔNSs and a reduced virulence for the NSm-deleted virus. In cattle, SBV lacking NSm caused viremia and seroconversion comparable to those caused by the wild-type virus, while the NSs and the combined NSs/NSm deletion mutant induced no detectable virus replication or clinical disease after immunization. Furthermore, three out of four cattle immunized once with the NSs deletion mutant and all animals vaccinated with the virus lacking both nonstructural proteins were fully protected against a challenge infection. Therefore, the double deletion mutant will provide the basis for further developments of safe and efficacious modified live SBV vaccines which could be also a model for other viruses of the Simbu serogroup and related orthobunyaviruses. IMPORTANCE SBV induces only mild clinical signs in adult ruminants but causes severe fetal malformation and, thereby, can have an important impact on animal welfare and production. As SBV is an insect-transmitted pathogen, vaccination will be one of the most important aspects of disease control. Here, mutant viruses lacking one or two proteins that essentially contribute to viral pathogenicity were tested as modified live vaccines in cattle. It could be demonstrated that a novel recombinant double deletion mutant is a safe and efficacious vaccine candidate. This is the first description of a putative modified live vaccine for the complete genus Orthobunyavirus, and in addition, such a vaccine type has never been tested in cattle for any virus of the entire family Bunyaviridae. Therefore, the described vaccine also represents the first model for a broad range of related viruses and is of high importance to the field.
Collapse
|
43
|
Abstract
Orthobunyaviruses, which have small, tripartite, negative-sense RNA genomes and structurally simple virions composed of just four proteins, can have devastating effects on human health and well-being, either by causing disease in humans or by causing disease in livestock and crops. In this Review, I describe the recent genetic and structural advances that have revealed important insights into the composition of orthobunyavirus virions, viral transcription and replication and viral interactions with the host innate immune response. Lastly, I highlight outstanding questions and areas of future research.
Collapse
Affiliation(s)
- Richard M Elliott
- MRC-University of Glasgow Centre for Virus Research, 464 Bearsden Road, Glasgow G61 1QH, UK
| |
Collapse
|
44
|
|
45
|
Fernández de Castro I, Sanz-Sánchez L, Risco C. Metallothioneins for correlative light and electron microscopy. Methods Cell Biol 2014; 124:55-70. [PMID: 25287836 DOI: 10.1016/b978-0-12-801075-4.00003-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Structural biologists have been working for decades on new strategies to identify proteins in cells unambiguously. We recently explored the possibilities of using the small metal-binding protein, metallothionein (MT), as a tag to detect proteins in transmission electron microscopy. It had been reported that, when fused with a protein of interest and treated in vitro with gold salts, a single MT tag will build an electron-dense gold cluster ~1 nm in diameter; we provided proof of this principle by demonstrating that MT can be used to detect intracellular proteins in bacteria and eukaryotic cells. The method, which is compatible with a variety of sample processing techniques, allows specific detection of proteins in cells with exceptional sensitivity. We illustrated the applicability of the technique in a series of studies to visualize the intracellular distribution of bacterial and viral proteins. Immunogold labeling was fundamental to confirm the specificity of the MT-gold method. When proteins were double-tagged with green fluorescent protein and MT, direct correlative light and electron microscopy allowed visualization of the same macromolecular complexes with different spatial resolutions. MT-gold tagging might also become a useful tool for mapping proteins into the 3D-density maps produced by (cryo)-electron tomography. New protocols will be needed for double or multiple labeling of proteins, using different versions of MT with fluorophores of different colors. Further research is also necessary to render the MT-gold labeling procedure compatible with immunogold labeling on Tokuyasu cryosections and with cryo-electron microscopy of vitreous sections.
Collapse
Affiliation(s)
- Isabel Fernández de Castro
- Cell Structure Laboratory, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Cantoblanco, Madrid, Spain
| | - Laura Sanz-Sánchez
- Cell Structure Laboratory, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Cantoblanco, Madrid, Spain
| | - Cristina Risco
- Cell Structure Laboratory, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Cantoblanco, Madrid, Spain
| |
Collapse
|
46
|
Zhao G, Krishnamurthy S, Cai Z, Popov VL, Travassos da Rosa AP, Guzman H, Cao S, Virgin HW, Tesh RB, Wang D. Identification of novel viruses using VirusHunter--an automated data analysis pipeline. PLoS One 2013; 8:e78470. [PMID: 24167629 PMCID: PMC3805514 DOI: 10.1371/journal.pone.0078470] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 09/12/2013] [Indexed: 12/14/2022] Open
Abstract
Quick and accurate identification of microbial pathogens is essential for both diagnosis and response to emerging infectious diseases. The advent of next-generation sequencing technology offers an unprecedented platform for rapid sequencing-based identification of novel viruses. We have developed a customized bioinformatics data analysis pipeline, VirusHunter, for the analysis of Roche/454 and other long read Next generation sequencing platform data. To illustrate the utility of VirusHunter, we performed Roche/454 GS FLX titanium sequencing on two unclassified virus isolates from the World Reference Center for Emerging Viruses and Arboviruses (WRCEVA). VirusHunter identified sequences derived from a novel bunyavirus and a novel reovirus in the two samples respectively. Further sequence analysis demonstrated that the viruses were novel members of the Phlebovirus and Orbivirus genera. Both Phlebovirus and Orbivirus genera include many economic important viruses or serious human pathogens.
Collapse
Affiliation(s)
- Guoyan Zhao
- Departments of Pathology & Immunology, Washington University School of Medicine, and the Midwest Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, St. Louis, Missouri, United States of America
| | - Siddharth Krishnamurthy
- Department of Molecular Microbiology, Washington University School of Medicine, and the Midwest Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, St. Louis, Missouri, United States of America
| | - Zhengqiu Cai
- Departments of Pathology & Immunology, Washington University School of Medicine, and the Midwest Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, St. Louis, Missouri, United States of America
| | - Vsevolod L. Popov
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Amelia P. Travassos da Rosa
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Hilda Guzman
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Song Cao
- Departments of Pathology & Immunology, Washington University School of Medicine, and the Midwest Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, St. Louis, Missouri, United States of America
| | - Herbert W. Virgin
- Departments of Pathology & Immunology, Washington University School of Medicine, and the Midwest Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, St. Louis, Missouri, United States of America
| | - Robert B. Tesh
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - David Wang
- Departments of Pathology & Immunology, Washington University School of Medicine, and the Midwest Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, and the Midwest Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, St. Louis, Missouri, United States of America
| |
Collapse
|
47
|
Non-structural proteins of arthropod-borne bunyaviruses: roles and functions. Viruses 2013; 5:2447-68. [PMID: 24100888 PMCID: PMC3814597 DOI: 10.3390/v5102447] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 09/20/2013] [Accepted: 09/25/2013] [Indexed: 12/24/2022] Open
Abstract
Viruses within the Bunyaviridae family are tri-segmented, negative-stranded RNA viruses. The family includes several emerging and re-emerging viruses of humans, animals and plants, such as Rift Valley fever virus, Crimean-Congo hemorrhagic fever virus, La Crosse virus, Schmallenberg virus and tomato spotted wilt virus. Many bunyaviruses are arthropod-borne, so-called arboviruses. Depending on the genus, bunyaviruses encode, in addition to the RNA-dependent RNA polymerase and the different structural proteins, one or several non-structural proteins. These non-structural proteins are not always essential for virus growth and replication but can play an important role in viral pathogenesis through their interaction with the host innate immune system. In this review, we will summarize current knowledge and understanding of insect-borne bunyavirus non-structural protein function(s) in vertebrate, plant and arthropod.
Collapse
|
48
|
Sanz-Sánchez L, Risco C. Multilamellar structures and filament bundles are found on the cell surface during bunyavirus egress. PLoS One 2013; 8:e65526. [PMID: 23799021 PMCID: PMC3683019 DOI: 10.1371/journal.pone.0065526] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 04/29/2013] [Indexed: 01/12/2023] Open
Abstract
Inside cells, viruses build specialized compartments for replication and morphogenesis. We observed that virus release associates with specific structures found on the surface of mammalian cells. Cultured adherent cells were infected with a bunyavirus and processed for oriented sectioning and transmission electron microscopy. Imaging of cell basal regions showed sophisticated multilamellar structures (MLS) and extracellular filament bundles with attached viruses. Correlative light and electron microscopy confirmed that both MLS and filaments proliferated during the maximum egress of new viruses. MLS dimensions and structure were reminiscent of those reported for the nanostructures on gecko fingertips, which are responsible for the extraordinary attachment capacity of these lizards. As infected cells with MLS were more resistant to detachment than control cells, we propose an adhesive function for these structures, which would compensate for the loss of adherence during release of new virus progeny.
Collapse
Affiliation(s)
- Laura Sanz-Sánchez
- Cell Structure Laboratory, Centro Nacional de Biotecnología, CNB-CSIC, Campus de Cantoblanco, Madrid, Spain
| | - Cristina Risco
- Cell Structure Laboratory, Centro Nacional de Biotecnología, CNB-CSIC, Campus de Cantoblanco, Madrid, Spain
| |
Collapse
|
49
|
Systems to establish bunyavirus genome replication in the absence of transcription. J Virol 2013; 87:8205-12. [PMID: 23698297 DOI: 10.1128/jvi.00371-13] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The L polymerase of bunyaviruses replicates and transcribes the viral genome. While replication products are faithful copies of the uncapped genomic RNA, transcription products contain capped 5' extensions which had been cleaved from host cell mRNAs. For La Crosse virus (LACV; genus Orthobunyavirus), the nuclease responsible for host cell mRNA cleavage is located at the N terminus of the L protein, with an active site of five conserved amino acids (H34, D52, D79, D92, and K94) surrounding two Mn(2+) ions (J. Reguera, F. Weber, and S. Cusack, PLoS Pathog. 6:e1001101, 2010). Here, we present reverse genetics systems and L mutants enabling us to study bunyaviral genome replication in the absence of transcription. Transcription was evaluated with an enhanced minigenome system consisting of the viral polymerase L, nucleocapsid protein N, a negative-sense minigenome, and--to alleviate antiviral host responses--a dominant-negative mutant (PKRΔE7) of the antiviral kinase protein kinase R (PKR). The transcriptional activity was strongly reduced by mutation of any of the five key amino acids, and the H34K, D79A, D92A, and K94A LACV L mutants were almost entirely silent in transcription. The replication activity of the L mutants was measured by packaging of progeny minigenomes into virus-like particles (VLPs). All mutant L proteins except K94A retained full replication activity. To test the broader applicability of our results, we introduced the homolog of mutation D79A (D111A) into the L sequence of Rift Valley fever virus (RVFV; genus Phlebovirus). As for LACV D79A, the RVFV D111A was incapable of transcription but fully active in replication. Thus, we generated mutants of LACV and RVFV L polymerases that are specifically deficient in transcription. Genome replication by bunyavirus polymerases can now be studied in the absence of transcription using convenient reverse genetics systems.
Collapse
|
50
|
Epidemiology, molecular virology and diagnostics of Schmallenberg virus, an emerging orthobunyavirus in Europe. Vet Res 2013; 44:31. [PMID: 23675914 PMCID: PMC3663787 DOI: 10.1186/1297-9716-44-31] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 04/22/2013] [Indexed: 12/26/2022] Open
Abstract
After the unexpected emergence of Bluetongue virus serotype 8 (BTV-8) in northern Europe in 2006, another arbovirus, Schmallenberg virus (SBV), emerged in Europe in 2011 causing a new economically important disease in ruminants. The virus, belonging to the Orthobunyavirus genus in the Bunyaviridae family, was first detected in Germany, in The Netherlands and in Belgium in 2011 and soon after in the United Kingdom, France, Italy, Luxembourg, Spain, Denmark and Switzerland. This review describes the current knowledge on the emergence, epidemiology, clinical signs, molecular virology and diagnosis of SBV infection.
Collapse
|