1
|
Hsu T, Talley MJ, Yang P, Geiselhoeringer A, Yang C, Gorla A, Rahman MJ, Silva L, Chen D, Yang B. Identification of infectious viruses for risk-based virus testing of CHO unprocessed bulk using next-generation sequencing. Biotechnol Prog 2024; 40:e3485. [PMID: 39051853 DOI: 10.1002/btpr.3485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/30/2024] [Accepted: 05/24/2024] [Indexed: 07/27/2024]
Abstract
It is important to increase manufacturing speed to make medicines more widely available. One bottleneck for CHO-based drug substance release is the in vitro viral (IVV) cell-based assay on unprocessed bulk. To increase process speed, we evaluate the suitability of replacing the IVV cell-based assay with next-generation sequencing (NGS). First, we outline how NGS is currently used in the pharmaceutical industry, and how it may apply to CHO virus testing. Second, we examine CHO virus contamination history. Since prior virus contaminants can replicate in the production bioreactor, we perform a literature search and classify 159 viruses as high, medium, low, or unknown risk based on their ability to infect CHO cells. Overall, the risk of virus contamination during the CHO manufacturing process is low. Only six viruses were reported to have contaminated CHO bioprocesses over the past several decades, and were primarily caused by fetal bovine serum or cell culture components. These virus contamination events can be mitigated through limitation and control of raw materials, combined with virus testing and virus clearance technologies. The list of CHO infectious viruses provides a starting framework for virus safety risk assessment and NGS development. Furthermore, ICH Q5A (R2) includes NGS as a molecular method for adventitious agent testing, paving a path forward for modernizing CHO virus testing.
Collapse
Affiliation(s)
- Tiffany Hsu
- Purification, Microbiology, and Virology, Genentech, a Member of the Roche Group, South San Francisco, California, USA
| | - Mary Jo Talley
- Purification, Microbiology, and Virology, Genentech, a Member of the Roche Group, South San Francisco, California, USA
| | - Ping Yang
- Purification, Microbiology, and Virology, Genentech, a Member of the Roche Group, South San Francisco, California, USA
| | - Angela Geiselhoeringer
- Purification, Microbiology, and Virology, Genentech, a Member of the Roche Group, South San Francisco, California, USA
| | - Cindy Yang
- Purification, Microbiology, and Virology, Genentech, a Member of the Roche Group, South San Francisco, California, USA
| | - Aditya Gorla
- Purification, Microbiology, and Virology, Genentech, a Member of the Roche Group, South San Francisco, California, USA
| | - M Julhasur Rahman
- Purification, Microbiology, and Virology, Genentech, a Member of the Roche Group, South San Francisco, California, USA
| | - Lindsey Silva
- Purification, Microbiology, and Virology, Genentech, a Member of the Roche Group, South San Francisco, California, USA
| | - Dayue Chen
- Purification, Microbiology, and Virology, Genentech, a Member of the Roche Group, South San Francisco, California, USA
| | - Bin Yang
- Purification, Microbiology, and Virology, Genentech, a Member of the Roche Group, South San Francisco, California, USA
| |
Collapse
|
2
|
Zarate-Sanchez E, George SC, Moya ML, Robertson C. Vascular dysfunction in hemorrhagic viral fevers: opportunities for organotypic modeling. Biofabrication 2024; 16:032008. [PMID: 38749416 PMCID: PMC11151171 DOI: 10.1088/1758-5090/ad4c0b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/25/2024] [Accepted: 05/15/2024] [Indexed: 06/06/2024]
Abstract
The hemorrhagic fever viruses (HFVs) cause severe or fatal infections in humans. Named after their common symptom hemorrhage, these viruses induce significant vascular dysfunction by affecting endothelial cells, altering immunity, and disrupting the clotting system. Despite advances in treatments, such as cytokine blocking therapies, disease modifying treatment for this class of pathogen remains elusive. Improved understanding of the pathogenesis of these infections could provide new avenues to treatment. While animal models and traditional 2D cell cultures have contributed insight into the mechanisms by which these pathogens affect the vasculature, these models fall short in replicatingin vivohuman vascular dynamics. The emergence of microphysiological systems (MPSs) offers promising avenues for modeling these complex interactions. These MPS or 'organ-on-chip' models present opportunities to better mimic human vascular responses and thus aid in treatment development. In this review, we explore the impact of HFV on the vasculature by causing endothelial dysfunction, blood clotting irregularities, and immune dysregulation. We highlight how existing MPS have elucidated features of HFV pathogenesis as well as discuss existing knowledge gaps and the challenges in modeling these interactions using MPS. Understanding the intricate mechanisms of vascular dysfunction caused by HFV is crucial in developing therapies not only for these infections, but also for other vasculotropic conditions like sepsis.
Collapse
Affiliation(s)
- Evelyn Zarate-Sanchez
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States of America
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States of America
| | - Monica L Moya
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Claire Robertson
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
- UC Davis Comprehensive Cancer Center, Davis, CA, United States of America
| |
Collapse
|
3
|
Viruses Binding to Host Receptors Interacts with Autophagy. Int J Mol Sci 2023; 24:ijms24043423. [PMID: 36834833 PMCID: PMC9968160 DOI: 10.3390/ijms24043423] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/19/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Viruses must cross the plasma membrane to infect cells, making them eager to overcome this barrier in order to replicate in hosts. They bind to cell surface receptors as the first step of initiating entry. Viruses can use several surface molecules that allow them to evade defense mechanisms. Various mechanisms are stimulated to defend against viruses upon their entry into cells. Autophagy, one of the defense systems, degrades cellular components to maintain homeostasis. The presence of viruses in the cytosol regulates autophagy; however, the mechanisms by which viral binding to receptors regulates autophagy have not yet been fully established. This review discusses recent findings on autophagy induced by interactions between viruses and receptors. It provides novel perspectives on the mechanism of autophagy as regulated by viruses.
Collapse
|
4
|
Tariq M, Kim DM. Hemorrhagic Fever with Renal Syndrome: Literature Review, Epidemiology, Clinical Picture and Pathogenesis. Infect Chemother 2022; 54:1-19. [PMID: 35384417 PMCID: PMC8987181 DOI: 10.3947/ic.2021.0148] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 01/30/2022] [Indexed: 02/06/2023] Open
Abstract
Hantaviruses can cause two types of infections in humans: hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome. The old world hantaviruses, primarily Hantaan virus (HTNV), responsible for causing HFRS occurs endemically in Asia and Europe. Apodernus agraricus, a striped field mouse, is being considered as main host reservoir for HTNV. Infection in humans is typically accidental and occurs when virus-containing rodent excretions such as urine, feces, or saliva are aerosolized. The major clinical manifestations includes increased vascular permeability causing vascular leakage, acute kidney injury and coagulation abnormalities. The case fatality rate of HFRS varies around 5.0 - 10.0% depending on the causative viral agent. The direct effects of viral infection on endothelial cells, as well as the immunological response to the viral infection, have been suggested to play a key role in the pathogenesis of HFRS. This article summarizes the current knowledge of HFRS epidemiology in Korea and around the globe, etiology, host transmission, clinical presentation, pathogenesis, diagnostic techniques, treatment, and prevention.
Collapse
Affiliation(s)
- Misbah Tariq
- Department of Internal Medicine, Chosun University College of Medicine, Gwangju, Korea.,Dow University of Health Sciences, Karachi, Pakistan
| | - Dong-Min Kim
- Department of Internal Medicine, Chosun University College of Medicine, Gwangju, Korea.
| |
Collapse
|
5
|
Meier K, Thorkelsson SR, Quemin ERJ, Rosenthal M. Hantavirus Replication Cycle-An Updated Structural Virology Perspective. Viruses 2021; 13:1561. [PMID: 34452426 PMCID: PMC8402763 DOI: 10.3390/v13081561] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 11/17/2022] Open
Abstract
Hantaviruses infect a wide range of hosts including insectivores and rodents and can also cause zoonotic infections in humans, which can lead to severe disease with possible fatal outcomes. Hantavirus outbreaks are usually linked to the population dynamics of the host animals and their habitats being in close proximity to humans, which is becoming increasingly important in a globalized world. Currently there is neither an approved vaccine nor a specific and effective antiviral treatment available for use in humans. Hantaviruses belong to the order Bunyavirales with a tri-segmented negative-sense RNA genome. They encode only five viral proteins and replicate and transcribe their genome in the cytoplasm of infected cells. However, many details of the viral amplification cycle are still unknown. In recent years, structural biology methods such as cryo-electron tomography, cryo-electron microscopy, and crystallography have contributed essentially to our understanding of virus entry by membrane fusion as well as genome encapsidation by the nucleoprotein. In this review, we provide an update on the hantavirus replication cycle with a special focus on structural virology aspects.
Collapse
Affiliation(s)
- Kristina Meier
- Department of Virology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany;
| | - Sigurdur R. Thorkelsson
- Centre for Structural Systems Biology, Leibniz Institute for Experimental Virology, University of Hamburg, 22607 Hamburg, Germany;
| | - Emmanuelle R. J. Quemin
- Centre for Structural Systems Biology, Leibniz Institute for Experimental Virology, University of Hamburg, 22607 Hamburg, Germany;
| | - Maria Rosenthal
- Department of Virology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany;
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| |
Collapse
|
6
|
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
|
7
|
Pizarro E, Navarrete M, Mendez C, Zaror L, Mansilla C, Tapia M, Carrasco C, Salazar P, Murua R, Padula P, Otth C, Rodríguez EM. Immunocytochemical and Ultrastructural Evidence Supporting That Andes Hantavirus (ANDV) Is Transmitted Person-to-Person Through the Respiratory and/or Salivary Pathways. Front Microbiol 2020; 10:2992. [PMID: 31998273 PMCID: PMC6965362 DOI: 10.3389/fmicb.2019.02992] [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: 10/16/2019] [Accepted: 12/10/2019] [Indexed: 01/07/2023] Open
Abstract
In South America Andes hantavirus (ANDV) is hosted by the rodent Oligoryzomys longicaudatus (also known as pygmy rice rat). In humans, ANDV causes Hantavirus Pulmonary Syndrome (HPS), with a fatality rate of about 40%. Epidemiologic and molecular evidence has shown that ANDV can be transmitted from person to person. Sin Nombre hantavirus, occurring in North America, and ANDV are genetically related, and both cause HPS with similar clinical evolution and mortality rate. However, only ANDV is transmitted from person to person. A recent hantavirus outbreak in a small village in Southern Argentine, with 29 HPS cases and 11 deaths has brought to mind that person-to-person transmission continues to be a public health emergency. The present investigation was aimed to understand how does ANDV actually spread between persons. Tissue samples of lung and salivary glands from infected Oligoryzomys longicaudatus and lethal cases of human HPS were investigated by bright field immunocytochemistry, multichannel immunofluorescence, and transmission electron microscopy. The findings are consistent with ANDV infection and replication in the lung alveolar epithelium and macrophages, and in the secretory cells of the submandibular salivary glands. In the lung of infected Oligoryzomys longicaudatus and human cases HPS, the bulk of immunoreactive hantavirus antigens was localized in epithelial cells of the alveolar walls and macrophages. The ultrastructural study supports that in the lung of HPS patients the virus replicates in the alveolar epithelial cells with virus particles being discharged into the alveolar lumen. Virus-like particles were seen within vacuoles of the lung macrophages. Considering that these macrophages can reach the conductive segments of the airways, their expectoration becomes a deadly bullet for ANDV transmission. In the submandibular glands of infected rodents and HPS cases, ANDV antigens were in capillary endothelium, the secretory cells and filling the lumen of the excretory pathway. It is proposed that in patients with HPS caused by ANDV the alveolar epithelium and macrophages would be the gate for the airway spreading of the virus, while the salivary glands are a target for virus replication and an exit pathway through saliva.
Collapse
Affiliation(s)
- Enrique Pizarro
- Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Maritza Navarrete
- Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile.,Instituto de Microbiología Clínica, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile.,Unidad Microbiología Clínica, Hospital Base Valdivia, Servicio de Salud Valdivia, Valdivia, Chile
| | - Carolina Mendez
- Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile.,Instituto de Microbiología Clínica, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Luis Zaror
- Instituto de Microbiología Clínica, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Carlos Mansilla
- Unidad de Anatomía Patológica, Servicio de Salud Aysén, Hospital Regional de Coyhaique, Aysén, Chile
| | - Mauricio Tapia
- Unidad de Anatomía Patológica, Servicio de Salud Aysén, Hospital Regional de Coyhaique, Aysén, Chile
| | - Cristian Carrasco
- Subdepartamento Anatomía Patológica Hospital Base Valdivia Servicio de Salud Valdivia, Valdivia, Chile
| | - Paula Salazar
- Instituto de Microbiología Clínica, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Roberto Murua
- Instituto de Ecología y Evolución, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Paula Padula
- Servicio Biología Molecular, Departamento de Virología, Instituto Nacional de Enfermedades Infecciosas, ANLIS "Dr. Carlos G. Malbrán", Buenos Aires, Argentina
| | - Carola Otth
- Instituto de Microbiología Clínica, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Esteban Martin Rodríguez
- Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
8
|
Chen Y, Dessau M, Rotenberg D, Rasmussen DA, Whitfield AE. Entry of bunyaviruses into plants and vectors. Adv Virus Res 2019; 104:65-96. [PMID: 31439153 DOI: 10.1016/bs.aivir.2019.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The majority of plant-infecting viruses are transmitted by arthropod vectors that deliver them directly into a living plant cell. There are diverse mechanisms of transmission ranging from direct binding to the insect stylet (non-persistent transmission) to persistent-propagative transmission in which the virus replicates in the insect vector. Despite this diversity in interactions, most arthropods that serve as efficient vectors have feeding strategies that enable them to deliver the virus into the plant cell without extensive damage to the plant and thus effectively inoculate the plant. As such, the primary virus entry mechanism for plant viruses is mediated by the biological vector. Remarkably, viruses that are transmitted in a propagative manner (bunyaviruses, rhabdoviruses, and reoviruses) have developed an ability to replicate in hosts from two kingdoms. Viruses in the order Bunyavirales are of emerging importance and with the advent of new sequencing technologies, we are getting unprecedented glimpses into the diversity of these viruses. Plant-infecting bunyaviruses are transmitted in a persistent, propagative manner must enter two unique types of host cells, plant and insect. In the insect phase of the virus life cycle, the propagative viruses likely use typical cellular entry strategies to traverse cell membranes. In this review, we highlight the transmission and entry strategies of three genera of plant-infecting bunyaviruses: orthotospoviruses, tenuiviruses, and emaraviruses.
Collapse
Affiliation(s)
- Yuting Chen
- Department of Entomology and Plant Pathology, North Carolina State University, Raleigh, NC, United States
| | - Moshe Dessau
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Dorith Rotenberg
- Department of Entomology and Plant Pathology, North Carolina State University, Raleigh, NC, United States
| | - David A Rasmussen
- Department of Entomology and Plant Pathology, North Carolina State University, Raleigh, NC, United States
| | - Anna E Whitfield
- Department of Entomology and Plant Pathology, North Carolina State University, Raleigh, NC, United States.
| |
Collapse
|
9
|
Zhang J, Sun L. Global profiling of megalocytivirus-induced proteins in tongue sole (Cynoglossus semilaevis) spleen identifies cellular processes essential to viral infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 92:150-159. [PMID: 30428365 PMCID: PMC7102559 DOI: 10.1016/j.dci.2018.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/06/2018] [Accepted: 11/06/2018] [Indexed: 05/30/2023]
Abstract
Megalocytivirus is a DNA virus with a broad host range among farmed fish including tongue sole (Cynoglossus semilaevis). In this study, label-free proteomics was performed to examine protein expression in tongue sole spleen induced by megalocytivirus at 8 and 12 days post infection (dpi). Compared to uninfected control fish, virus-infected fish displayed 315 up-regulated proteins and 111 down-regulated proteins at 8 dpi, and 48 up-regulated proteins and 43 down-regulated proteins at 12 dpi. The expressions of five differentially expressed proteins were confirmed by Western blot. The differentially expressed proteins were enriched in the pathways and processes associated with innate immune response and viral infection. Interference with the expression of two up-regulated proteins of the ubiquitin proteasome system (UPS), i.e. proteasome assembly chaperone 2 and proteasome maturation protein, significantly reduced viral propagation in fish, whereas overexpression of these two proteins significantly enhanced viral propagation. Consistently, inhibition of the functioning of proteasome significantly impaired viral replication in vivo. This study provided the first global protein profile responsive to megalocytivirus in tongue sole, and revealed an essential role of UPS in viral infection.
Collapse
Affiliation(s)
- Jian Zhang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China; Deep Sea Research Center, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Li Sun
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China.
| |
Collapse
|
10
|
Tang X, Zhai F, Sheng X, Xing J, Zhan W. The Roles of β-Integrin of Chinese Shrimp (Fenneropenaeus chinensis) in WSSV Infection. Int J Mol Sci 2017; 18:ijms18071465. [PMID: 28686185 PMCID: PMC5535956 DOI: 10.3390/ijms18071465] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/28/2017] [Accepted: 07/03/2017] [Indexed: 01/21/2023] Open
Abstract
Our previous study demonstrated that an integrin β subunit of Chinese shrimp (Fenneropenaeus chinensis) (FcβInt) plays an important role in white spot syndrome virus (WSSV) infection. In the present work, in order to further elucidate the potential role of FcβInt in WSSV infection, the recombinant extracellular domain of β integringene of F. Chinensis (rFcβInt-ER) was expressed in Escherichia coli BL21 (DE3), and the eukaryotic expression plasmid PcDNA3.1-FcβInt-ER (PFcβInt-ER) was also constructed. Far-western blotting was performed to determine the binding specificity of rFcβInt-ER to WSSV envelope proteins, and results showed that rFcβInt-ER was able to specifically interact with rVP31, rVP37, rVP110 and rVP187. Moreover, the blocking effects of mouse anti-rFcβint-ER antibodies were both detected in vivo and in vitro. The ELISA and Dot-blotting in vitro assays both showed that mouse anti-rFcβInt-ER antibodies could partially block the binding of WSSV to the hemocyte membrane of F. chinensis. In the in vivo assays, the mortality of shrimp injected with WSSV mixed with anti-rFcβInt-ER antibodies was delayed, and was lower than in the control group. While the shrimp were intramuscularly injected with PFcβInt-ER, transcripts of PFcβInt-ER could be detected in different shrimp tissues within 7 days, and the mortality of shrimp injected with PFcβInt-ER was also delayed and lower compared with the control group post WSSV challenge. Furthermore, gene silencing technology was also used to verify the effect of FcβInt in WSSV infection, and results showed that the expression levels of the WSSV immediate early gene iel, early gene wsv477, and late gene VP28 and the mortality of F. Chinensis were all significantly decreased in the FcβInt knock-down hemocyctes compared to the control group. Taken together, these results suggest that FcβInt plays important roles in WSSV infection.
Collapse
Affiliation(s)
- Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Aoshanwei Town, Jimo, Qingdao 266071, China.
| | - Fude Zhai
- Laboratory of Pathology and Immunology of Aquatic Animals, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Aoshanwei Town, Jimo, Qingdao 266071, China.
| |
Collapse
|
11
|
Ermonval M, Baychelier F, Tordo N. What Do We Know about How Hantaviruses Interact with Their Different Hosts? Viruses 2016; 8:v8080223. [PMID: 27529272 PMCID: PMC4997585 DOI: 10.3390/v8080223] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/27/2016] [Accepted: 08/05/2016] [Indexed: 11/26/2022] Open
Abstract
Hantaviruses, like other members of the Bunyaviridae family, are emerging viruses that are able to cause hemorrhagic fevers. Occasional transmission to humans is due to inhalation of contaminated aerosolized excreta from infected rodents. Hantaviruses are asymptomatic in their rodent or insectivore natural hosts with which they have co-evolved for millions of years. In contrast, hantaviruses cause different pathologies in humans with varying mortality rates, depending on the hantavirus species and its geographic origin. Cases of hemorrhagic fever with renal syndrome (HFRS) have been reported in Europe and Asia, while hantavirus cardiopulmonary syndromes (HCPS) are observed in the Americas. In some cases, diseases caused by Old World hantaviruses exhibit HCPS-like symptoms. Although the etiologic agents of HFRS were identified in the early 1980s, the way hantaviruses interact with their different hosts still remains elusive. What are the entry receptors? How do hantaviruses propagate in the organism and how do they cope with the immune system? This review summarizes recent data documenting interactions established by pathogenic and nonpathogenic hantaviruses with their natural or human hosts that could highlight their different outcomes.
Collapse
Affiliation(s)
- Myriam Ermonval
- Unité des Stratégies Antivirales, Département de Virologie, Institut Pasteur, 25 Rue du Docteur Roux, 75015 Paris, France.
| | - Florence Baychelier
- Unité des Stratégies Antivirales, Département de Virologie, Institut Pasteur, 25 Rue du Docteur Roux, 75015 Paris, France.
| | - Noël Tordo
- Unité des Stratégies Antivirales, Département de Virologie, Institut Pasteur, 25 Rue du Docteur Roux, 75015 Paris, France.
| |
Collapse
|
12
|
Albornoz A, Hoffmann AB, Lozach PY, Tischler ND. Early Bunyavirus-Host Cell Interactions. Viruses 2016; 8:v8050143. [PMID: 27213430 PMCID: PMC4885098 DOI: 10.3390/v8050143] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/15/2016] [Indexed: 12/12/2022] Open
Abstract
The Bunyaviridae is the largest family of RNA viruses, with over 350 members worldwide. Several of these viruses cause severe diseases in livestock and humans. With an increasing number and frequency of outbreaks, bunyaviruses represent a growing threat to public health and agricultural productivity globally. Yet, the receptors, cellular factors and endocytic pathways used by these emerging pathogens to infect cells remain largely uncharacterized. The focus of this review is on the early steps of bunyavirus infection, from virus binding to penetration from endosomes. We address current knowledge and advances for members from each genus in the Bunyaviridae family regarding virus receptors, uptake, intracellular trafficking and fusion.
Collapse
Affiliation(s)
- Amelina Albornoz
- Molecular Virology Laboratory, Fundación Ciencia & Vida, Av. Zañartu 1482, 7780272 Santiago, Chile.
| | - Anja B Hoffmann
- CellNetworks-Cluster of Excellence and Department of Infectious Diseases, Virology, University Hospital Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.
| | - Pierre-Yves Lozach
- CellNetworks-Cluster of Excellence and Department of Infectious Diseases, Virology, University Hospital Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.
| | - Nicole D Tischler
- Molecular Virology Laboratory, Fundación Ciencia & Vida, Av. Zañartu 1482, 7780272 Santiago, Chile.
| |
Collapse
|
13
|
Abstract
Hantaviruses are emerging zoonotic pathogens that belong to the Bunyaviridae family. They have been classified as category A pathogens by CDC (centers for disease control and prevention). Hantaviruses pose a serious threat to human health because their infection causes two highly fatal diseases, hemorrhagic fever with renal syndrome (HFRS) and hantavirus cardiopulmonary syndrome (HCPS). These pathogens are transmitted to humans through aerosolized excreta of their infected rodent hosts. Hantaviruses have a tripartite-segmented negative-sense RNA genome. The three genomic RNA segments, S, M, and L, encode a nucleocapsid protein (N), a precursor glycoprotein that is processed into two envelope glycoproteins (Gn and Gc) and the viral RNA-dependent RNA polymerase (RdRp), respectively. N protein is the major structural component of the virus, its main function is to protect and encapsidate the three genomic RNAs forming three viral ribonucleocapsids. Recent studies have proposed that N in conjunction with RdRp plays important roles in the transcription and replication of viral genome. In addition, N preferentially facilitates the translation of viral mRNA in cells. Glycoproteins, Gn and Gc, play major roles in viral attachment and entry to the host cells, virulence, and assembly and packaging of new virions in infected cells. RdRp functions as RNA replicase and transcriptase to replicate and transcribe the viral RNA and is also thought to have endonuclease activity. Currently, no antiviral therapy or vaccine is available for the treatment of hantavirus-associated diseases. Understanding the molecular details of hantavirus life cycle will help in the identification of targets for antiviral therapeutics and in the design of potential antiviral drug for the treatment of HFRS and HCPS. Due to the alarming fatality of hantavirus diseases, development of an effective vaccine against hantaviruses is a necessity.
Collapse
|
14
|
Jiang H, Du H, Wang LM, Wang PZ, Bai XF. Hemorrhagic Fever with Renal Syndrome: Pathogenesis and Clinical Picture. Front Cell Infect Microbiol 2016; 6:1. [PMID: 26870699 PMCID: PMC4737898 DOI: 10.3389/fcimb.2016.00001] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/05/2016] [Indexed: 01/08/2023] Open
Abstract
Hantaan virus (HTNV) causes hemorrhagic fever with renal syndrome (HFRS), which is a zoonosis endemic in eastern Asia, especially in China. The reservoir host of HTNV is field mouse (Apodemus agraricus). The main manifestation of HFRS, including acute kidney injury, increases vascular permeability, and coagulation abnormalities. In this paper, we review the current knowledge of the pathogenesis of HFRS including virus factor, immunity factor and host genetic factors. Furthermore, the treatment and prevention will be discussed.
Collapse
Affiliation(s)
- Hong Jiang
- Center for Infectious Diseases, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Hong Du
- Center for Infectious Diseases, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Li M Wang
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University Xi'an, China
| | - Ping Z Wang
- Center for Infectious Diseases, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Xue F Bai
- Center for Infectious Diseases, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| |
Collapse
|
15
|
Pan W, Bian G, Wang K, Feng T, Dai J. Effects of Different Doses of Nucleocapsid Protein from Hantaan Virus A9 Strain on Regulation of Interferon Signaling. Viral Immunol 2015; 28:448-54. [PMID: 26196448 PMCID: PMC4599133 DOI: 10.1089/vim.2015.0004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Hantaan virus A9 strain (HTNV A9) is an etiologic agent of hemorrhagic fever with renal syndrome in China. The virulence of the pathogenic hantaviruses is determined by their ability to alter key signaling pathways of early interferon (IFN) induction within cells. The potential role of HTNV A9 structural proteins, such as nucleocapsid (N) and envelope glycoproteins (Gn and Gc), in regulating human's innate antiviral immune response has not yet been clarified. In this study, we investigated the effect of HTNV A9 N protein on the regulation of the IFN pathway. We found that A9 N protein can influence the host innate immune response by regulating the activation of IFNβ. The A9 N protein stimulates IFN response in low doses, whereas significantly inhibits IFNβ production at high doses. Furthermore, A9 N protein constitutively inhibits nuclear factor kappa B activation. A high dose of A9 N protein could inhibit either Poly IC-induced IFNβ or vesicular stomatitis virus-induced IFNβ and interferon-stimulated gene production. Our results indicate that HTNV A9 N protein helps virus establish successful infection by downregulating the IFN response and shed new light to the understanding of the interaction between the host innate immunity and virus during Hantaan virus infection.
Collapse
Affiliation(s)
- Wen Pan
- Jiangsu Key Laboratory of Infection and Immunity, Institute of Biology and Medical Sciences, Soochow University , Suzhou City, People's Republic of China
| | - Gang Bian
- Jiangsu Key Laboratory of Infection and Immunity, Institute of Biology and Medical Sciences, Soochow University , Suzhou City, People's Republic of China
| | - Kezhen Wang
- Jiangsu Key Laboratory of Infection and Immunity, Institute of Biology and Medical Sciences, Soochow University , Suzhou City, People's Republic of China
| | - Tingting Feng
- Jiangsu Key Laboratory of Infection and Immunity, Institute of Biology and Medical Sciences, Soochow University , Suzhou City, People's Republic of China
| | - Jianfeng Dai
- Jiangsu Key Laboratory of Infection and Immunity, Institute of Biology and Medical Sciences, Soochow University , Suzhou City, People's Republic of China
| |
Collapse
|
16
|
Immunogenetic factors affecting susceptibility of humans and rodents to hantaviruses and the clinical course of hantaviral disease in humans. Viruses 2014; 6:2214-41. [PMID: 24859344 PMCID: PMC4036553 DOI: 10.3390/v6052214] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 03/17/2014] [Accepted: 05/16/2014] [Indexed: 12/11/2022] Open
Abstract
We reviewed the associations of immunity-related genes with susceptibility of humans and rodents to hantaviruses, and with severity of hantaviral diseases in humans. Several class I and class II HLA haplotypes were linked with severe or benign hantavirus infections, and these haplotypes varied among localities and hantaviruses. The polymorphism of other immunity-related genes including the C4A gene and a high-producing genotype of TNF gene associated with severe PUUV infection. Additional genes that may contribute to disease or to PUUV infection severity include non-carriage of the interleukin-1 receptor antagonist (IL-1RA) allele 2 and IL-1β (-511) allele 2, polymorphisms of plasminogen activator inhibitor (PAI-1) and platelet GP1a. In addition, immunogenetic studies have been conducted to identify mechanisms that could be linked with the persistence/clearance of hantaviruses in reservoirs. Persistence was associated during experimental infections with an upregulation of anti-inflammatory responses. Using natural rodent population samples, polymorphisms and/or expression levels of several genes have been analyzed. These genes were selected based on the literature of rodent or human/hantavirus interactions (some Mhc class II genes, Tnf promoter, and genes encoding the proteins TLR4, TLR7, Mx2 and β3 integrin). The comparison of genetic differentiation estimated between bank vole populations sampled over Europe, at neutral and candidate genes, has allowed to evidence signatures of selection for Tnf, Mx2 and the Drb Mhc class II genes. Altogether, these results corroborated the hypothesis of an evolution of tolerance strategies in rodents. We finally discuss the importance of these results from the medical and epidemiological perspectives.
Collapse
|
17
|
Hantavirus Gn and Gc envelope glycoproteins: key structural units for virus cell entry and virus assembly. Viruses 2014; 6:1801-22. [PMID: 24755564 PMCID: PMC4014721 DOI: 10.3390/v6041801] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 03/20/2014] [Accepted: 03/31/2014] [Indexed: 01/24/2023] Open
Abstract
In recent years, ultrastructural studies of viral surface spikes from three different genera within the Bunyaviridae family have revealed a remarkable diversity in their spike organization. Despite this structural heterogeneity, in every case the spikes seem to be composed of heterodimers formed by Gn and Gc envelope glycoproteins. In this review, current knowledge of the Gn and Gc structures and their functions in virus cell entry and exit is summarized. During virus cell entry, the role of Gn and Gc in receptor binding has not yet been determined. Nevertheless, biochemical studies suggest that the subsequent virus-membrane fusion activity is accomplished by Gc. Further, a class II fusion protein conformation has been predicted for Gc of hantaviruses, and novel crystallographic data confirmed such a fold for the Rift Valley fever virus (RVFV) Gc protein. During virus cell exit, the assembly of different viral components seems to be established by interaction of Gn and Gc cytoplasmic tails (CT) with internal viral ribonucleocapsids. Moreover, recent findings show that hantavirus glycoproteins accomplish important roles during virus budding since they self-assemble into virus-like particles. Collectively, these novel insights provide essential information for gaining a more detailed understanding of Gn and Gc functions in the early and late steps of the hantavirus infection cycle.
Collapse
|
18
|
Guo D, Zhu Q, Zhang H, Sun D. Proteomic analysis of membrane proteins of vero cells: exploration of potential proteins responsible for virus entry. DNA Cell Biol 2013; 33:20-8. [PMID: 24286161 DOI: 10.1089/dna.2013.2193] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Vero cells are highly susceptible to many viruses in humans and animals, and its membrane proteins (MPs) are responsible for virus entry. In our study, the MP proteome of the Vero cells was investigated using a shotgun LC-MS/MS approach. Six hundred twenty-seven proteins, including a total of 1839 peptides, were identified in MP samples of the Vero cells. In 627 proteins, 307 proteins (48.96%) were annotated in terms of biological process of gene ontology (GO) categories; 356 proteins (56.78%) were annotated in terms of molecular function of GO categories; 414 proteins (66.03%) were annotated in terms of cellular components of GO categories. Of 627 identified proteins, seventeen proteins had been revealed to be virus receptor proteins. The resulting protein lists and highlighted proteins may provide valuable information to increase understanding of virus infection of Vero cells.
Collapse
Affiliation(s)
- Donghua Guo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University , Daqing, People's Republic of China
| | | | | | | |
Collapse
|
19
|
Zhong R, Tang X, Zhan W, Xing J, Sheng X. Expression kinetics of β-integrin in Chinese shrimp (Fenneropenaeus chinensis) hemocytes following infection with white spot syndrome virus. FISH & SHELLFISH IMMUNOLOGY 2013; 35:539-545. [PMID: 23711468 DOI: 10.1016/j.fsi.2013.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/15/2013] [Accepted: 05/06/2013] [Indexed: 06/02/2023]
Abstract
Our previous study has demonstrated that an integrin β subunit of Chinese shrimp (Fenneropenaeus chinensis) (FcβInt) involved in WSSV infection. In order to further elucidate the potential role of the FcβInt in the WSSV infection, expression response of FcβInt to WSSV infection in shrimp hemocytes was investigated after intra-muscular injection with the virus. Following time-course hemocytes sampling, the expression variation of FcβInt in hemocytes was examined by flow cytometric immunofluorescence assay (FCIFA) and enzyme linked immunosorbent assay (ELISA) using the monoclonal antibody (Mab) 2C5 against FcβInt, which was successfully produced with recombinant partial FcβInt and exhibited binding to a 120 kDa hemocyte protein. Meanwhile, the dynamic state of FcβInt mRNA level and WSSV copies in hemocytes were determined by quantitative real-time PCR. The result of FCIFA showed that FcβInt was mainly expressed on the semi-granular and granular cells, which was down-regulated at 6 h post infection (p.i.), and significantly increased to the peak level at 12 h p.i., then decreased to the control level by 24 h. However, FcβInt on the hyaline cells was lowly expressed and didn't show active response to the viral infection. The variation of FcβInt concentrations in total hemocytes determined by ELISA was roughly in accordance with the changing tendency of FcβInt expressed on the semi-granular and granular cells. FcβInt mRNA level in total hemocytes was significantly up-regulated to the peak level at 12 h p.i. Moreover, the number of WSSV copies in hemocytes began to exhibit a significant increase at 24 h p.i. The present study demonstrated that WSSV infection would induce a differential regulation of FcβInt expression in different type hemocytes, which provided valuable evidences for the close correlation between FcβInt and WSSV infection.
Collapse
Affiliation(s)
- Rujie Zhong
- Laboratory of Pathology and Immunology of Aquatic Animals, Ocean University of China, No. 5, Yushan Road, Qingdao 266003, PR China
| | | | | | | | | |
Collapse
|
20
|
Sanada T, Seto T, Ozaki Y, Saasa N, Yoshimatsu K, Arikawa J, Yoshii K, Kariwa H. Isolation of Hokkaido virus, genus Hantavirus, using a newly established cell line derived from the kidney of the grey red-backed vole (Myodes rufocanus bedfordiae). J Gen Virol 2012; 93:2237-2246. [PMID: 22791608 DOI: 10.1099/vir.0.045377-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hantaviruses belong to the family Bunyaviridae and are maintained in wild rodents. Although Vero E6 cells, which originate from African green monkey kidney, are used widely in hantavirus research, isolation of hantaviruses from this cell line is difficult. To develop an efficient method of propagation and isolation of hantaviruses we established a novel cell line, MRK101, derived from the kidney of the grey red-backed vole (Myodes rufocanus bedfordiae), the natural host of Hokkaido virus (HOKV). The MRK101 cells showed a significantly higher susceptibility to Puumala virus (PUUV) hosted by Myodes glareolus than Vero E6 cells. Viral nucleocapsid protein in PUUV-infected MRK101 cells was detected earlier than in Vero E6 cells, and the viral titre in the culture fluid of MRK101 cells was higher than that of Vero E6 cells during the early phase of infection. In contrast, MRK101 cells showed no susceptibility to Hantaan virus. HOKV, which has not been isolated to date, was isolated successfully using MRK101 cells. Moreover, the newly isolated HOKV was successfully propagated in MRK101, but not Vero E6, cells. Phylogenic analyses of the S (small), M (medium) and L (large) segment sequences revealed that HOKV is related most closely to PUUV, but is distinct from other hantaviruses. These data suggest that the MRK101 cell line is a useful tool for the isolation and propagation of hantaviruses. Moreover, this is (to our knowledge) the first report of hantavirus isolation in a cell line that originated from the natural host.
Collapse
Affiliation(s)
- Takahiro Sanada
- Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Takahiro Seto
- Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Yuka Ozaki
- Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Ngonda Saasa
- Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Kumiko Yoshimatsu
- Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido 060-0838, Japan
| | - Jiro Arikawa
- Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido 060-0838, Japan
| | - Kentaro Yoshii
- Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Hiroaki Kariwa
- Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| |
Collapse
|
21
|
Hepojoki J, Strandin T, Lankinen H, Vaheri A. Hantavirus structure--molecular interactions behind the scene. J Gen Virol 2012; 93:1631-1644. [PMID: 22622328 DOI: 10.1099/vir.0.042218-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Viruses of the genus Hantavirus, carried and transmitted by rodents and insectivores, are the exception in the vector-borne virus family Bunyaviridae, since viruses of the other genera are transmitted via arthropods. The single-stranded, negative-sense, RNA genome of hantaviruses is trisegmented into small, medium and large (S, M and L) segments. The segments, respectively, encode three structural proteins: nucleocapsid (N) protein, two glycoproteins Gn and Gc and an RNA-dependent RNA-polymerase. The genome segments, encapsidated by the N protein to form ribonucleoproteins, are enclosed inside a lipid envelope that is decorated by spikes composed of Gn and Gc. The virion displays round or pleomorphic morphology with a diameter of roughly 120-160 nm depending on the detection method. This review focuses on the structural components of hantaviruses, their interactions, the mechanisms behind virion assembly and the interactions that maintain virion integrity. We attempt to summarize recent results on the virion structure and to suggest mechanisms on how the assembly is driven. We also compare hantaviruses to other bunyaviruses with known structure.
Collapse
Affiliation(s)
- Jussi Hepojoki
- Department of Virology, Peptide and Protein Laboratory, Infection Biology Research Program, Haartman Institute, University of Helsinki, Finland
| | - Tomas Strandin
- Department of Virology, Peptide and Protein Laboratory, Infection Biology Research Program, Haartman Institute, University of Helsinki, Finland
| | - Hilkka Lankinen
- Department of Virology, Peptide and Protein Laboratory, Infection Biology Research Program, Haartman Institute, University of Helsinki, Finland
| | - Antti Vaheri
- Department of Virology, Peptide and Protein Laboratory, Infection Biology Research Program, Haartman Institute, University of Helsinki, Finland
| |
Collapse
|
22
|
Wang W, Zhang Y, Li Y, Pan L, Bai L, Zhuang Y, Huang CX, Wang JP, Yu HT, Wei X, Jiang W, Nan YY, Yang DQ, Su WJ, Wang PZ, Bai XF. Dysregulation of the β3 integrin-VEGFR2 complex in Hantaan virus-directed hyperpermeability upon treatment with VEGF. Arch Virol 2012; 157:1051-61. [PMID: 22407444 DOI: 10.1007/s00705-012-1245-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 12/27/2011] [Indexed: 01/13/2023]
Abstract
Hantaviruses infect human endothelial cells (ECs) and are known to cause vascular-permeability-based diseases, including hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS). The αvβ3 integrins, which are highly expressed on the surface of ECs, serve as hantavirus receptors. Specifically, the β3 integrin and vascular endothelial growth factor (VEGF) receptor 2 (VEGFR2) form a functional complex and interact with each other. Signaling through this complex causes cytoskeletal reorganization, which is one of the most important mechanisms underlying hyperpermeability. In this study, we show that VEGF dramatically enhances Hantaan virus (HTNV)-directed permeability and increases the reorganization of the cytoskeleton and the disruption of junctional organizations in an EC monolayer at 3 days postinfection. HTNV infection reduced the effect of VEGF on adhesion, migration, and the upregulation of β3 expression, but the infection alone upregulated the expression of β3 and VEGFR2. These results indicate that in addition to its role in blocking β3 integrin activation as reported previously, HTNV blocks the function of the complex of VEGFR2 and β3 integrin, and the dysfunction of the complex may contribute to cytoskeletal reorganization in an HTNV-directed hyperpermeability response to VEGF.
Collapse
Affiliation(s)
- Wei Wang
- Center of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Efficient production of Hantaan and Puumala pseudovirions for viral tropism and neutralization studies. Virology 2011; 423:134-42. [PMID: 22209230 DOI: 10.1016/j.virol.2011.08.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 06/07/2011] [Accepted: 08/18/2011] [Indexed: 02/06/2023]
Abstract
Puumala (PUUV) and Hantaan (HTNV) viruses are hantaviruses within the family Bunyaviridae and associated with Hemorrhagic Fever with Renal Syndrome (HFRS) in humans. Little is known about how these viruses interact with host cells, though pathogenic hantaviruses interact with α(v)β(3) integrin. To study host cell interactions and rapidly test the ability of antibodies to prevent infection, we produced HTNV and PUUV pseudovirions on a vesicular stomatitis virus (VSV) core. Similar to replication-competent hantaviruses, infection was low-pH-dependent. Despite broad cell tropism, several human T cell lines were poorly permissive to hantavirus pseudovirions, compared to VSV, indicating a relative block to infection at the level of entry. Stable expression of α(v)β(3) integrin in SupT1 cells did not restore infectivity. Finally, the pseudovirion system provided a rapid, quantitative, and specific method to screen for neutralizing antibodies in immune sera.
Collapse
|
24
|
Klingström J, Ahlm C. Hantavirus protein interactions regulate cellular functions and signaling responses. Expert Rev Anti Infect Ther 2011; 9:33-47. [PMID: 21171876 DOI: 10.1586/eri.10.157] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Rodent-borne pathogenic hantaviruses cause two severe and often lethal zoonotic diseases: hemorrhagic fever with renal syndrome (HFRS) in Eurasia and hantavirus cardiopulmonary syndrome (HCPS) in the Americas. Currently, no US FDA-approved therapeutics or vaccines are available for HFRS/HCPS. Infections with hantaviruses are not lytic, and it is currently not known exactly why infections in humans cause disease. A better understanding of how hantaviruses interfere with normal cell functions and activation of innate and adaptive immune responses might provide clues to future development of specific treatment and/or vaccines against hantavirus infection. In this article, the current knowledge regarding immune responses observed in patients, hantavirus interference with cellular proteins and signaling pathways, and possible approaches in the development of therapeutics are discussed.
Collapse
Affiliation(s)
- Jonas Klingström
- Centre for Microbiological Preparedness, Swedish Institute for Infectious Disease Control, Solna, Sweden.
| | | |
Collapse
|
25
|
Development of a lentiviral vector system to study the role of the Andes virus glycoproteins. Virus Res 2010; 153:29-35. [PMID: 20619306 DOI: 10.1016/j.virusres.2010.07.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 06/24/2010] [Accepted: 07/01/2010] [Indexed: 01/29/2023]
Abstract
To infect target cells, enveloped viruses use their virion surface proteins to direct cell attachment and subsequent entry via virus-cell membrane fusion. How hantaviruses enter cells has been largely unexplored. To study early steps of Andes virus (ANDV) cell infection, a lentiviral vector system was developed based on a Simian immunodeficiency virus (SIV) vector pseudotyped with the ANDV-Gn/Gc envelope glycoproteins. The incorporation of Gn and Gc onto SIV-derived vector particles was assessed using newly generated monoclonal antibodies against ANDV glycoproteins. In addition, sera of ANDV infected humans were able to block cell entry of the SIV vector pseudotyped with ANDV glycoproteins, suggesting that their antigenic conformation is similar to that in the native virus. The use of such SIV vector pseudotyped with ANDV-Gn/Gc glycoproteins should facilitate studies on ANDV cell entry. Along this line, it was found that depletion of cholesterol from target cells strongly diminished cell infection, indicating a possible role of lipid rafts in ANDV cell entry. The Gn/Gc pseudotyped SIV vector has several advantages, notably high titer vector production and easy quantification of cell infection by monitoring GFP reporter gene expression by flow cytometry. Such pseudotyped SIV vectors can be used to identify functional domains in the Gn/Gc glycoproteins and to screen for potential hantavirus cell entry inhibitors.
Collapse
|
26
|
Abstract
Hantaviruses are enzootic viruses that maintain persistent infections in their rodent hosts without apparent disease symptoms. The spillover of these viruses to humans can lead to one of two serious illnesses, hantavirus pulmonary syndrome and hemorrhagic fever with renal syndrome. In recent years, there has been an improved understanding of the epidemiology, pathogenesis, and natural history of these viruses following an increase in the number of outbreaks in the Americas. In this review, current concepts regarding the ecology of and disease associated with these serious human pathogens are presented. Priorities for future research suggest an integration of the ecology and evolution of these and other host-virus ecosystems through modeling and hypothesis-driven research with the risk of emergence, host switching/spillover, and disease transmission to humans.
Collapse
|
27
|
Ray N, Whidby J, Stewart S, Hooper JW, Bertolotti-Ciarlet A. Study of Andes virus entry and neutralization using a pseudovirion system. J Virol Methods 2009; 163:416-23. [PMID: 19903496 DOI: 10.1016/j.jviromet.2009.11.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Revised: 10/13/2009] [Accepted: 11/03/2009] [Indexed: 10/20/2022]
Abstract
Andes virus (ANDV), a member of the Hantavirus genus in the family Bunyaviridae, causes an acute disease characteristic of New-World hantaviruses called hantavirus pulmonary syndrome (HPS). HPS is a highly pathogenic disease with a case-fatality rate of 40%. ANDV is the only hantavirus reported to spread directly from human-to-human. The aim of the present study was to develop a quantitative and high-throughput pseudovirion assay to study ANDV infection and neutralization in biosafety level 2 facilities (BSL-2). This pseudovirion assay is based on incorporation of ANDV glycoproteins onto replication-defective vesicular stomatitis virus (VSV) cores in which the gene for the surface G protein has been replaced by that encoding Renilla luciferase. Infection by the pseudovirions can be quantified by luciferase activity of infected cell lysates. ANDV pseudovirions were neutralized by ANDV-specific antisera, and there was good concordance between specificity and neutralization titers of ANDV hamster sera as determined by our pseudovirion assay and a commonly used plaque reduction neutralization titer (PRNT) assay. In addition, the pseudovirions were used to evaluate the requirements for ANDV entry, like pH dependency and the role of beta3 integrin, the reported receptor for other pathogenic hantaviruses, on entry.
Collapse
Affiliation(s)
- Neelanjana Ray
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
28
|
Lü X, Yin W, Yang Q, Lei Y, Yang J, Sun M, Yao M, Xu Z, Xue X. Identification of oligopeptides mimicking the receptor-binding domain of Hantaan virus envelope glycoprotein from a phage-displayed peptide library. Can J Microbiol 2009; 55:664-71. [PMID: 19767836 DOI: 10.1139/w09-012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Attachment of enveloped viruses to cells is triggered by the receptor-binding domain (RBD) on envelope glycoproteins (GP) binding to receptors located on the cell surface. To date, recognized receptors and RBD of hantaan virus (HTNV) have not been exactly defined. In this study, one monoclonal antibody (MAb) 3G1 possessing high neutralizing activity, which is directed against HTNV envelope glycoprotein G2, was used to determine the crucial motif of RBD. Peptide ligands binding to MAb 3G1 were selected from a 12 amino acid peptide library displayed on filamentous phages. After 3 rounds of selection, the binding capacity between phages and MAb 3G1 was examined byELISA. Afterwards the positive phage clones with high binding activity to MAb 3G1 were chosen and sequenced. The peptide sequences of positive phage clones were compared with that of HTNV 76-118 strain G2. A motif Y/F/WPW(X)HX1-2HY, aligned to the primary sequences of G2 96YPWHTAKCHY105, was identified from the peptide inserts in the 9 positive clones. Positive phages and synthesized peptide containing the motif were bound significantly to virus-susceptible cell (Vero-E6) membranes by ELISA and immunofluorescence assay, respectively. Therefore, the sequence on G2 between amino acid 96 and 105 may be a key motif of HTNV RBD recognized by viral receptors on target cell membranes. Further characterization of the motif would provide useful information in understanding of the cellular entry of HTNV.
Collapse
Affiliation(s)
- Xin Lü
- Department of Microbiology, the Fourth Military Medical University, Xi'an 710032, Shaanxi Province, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Duan T, Ferguson M, Yuan L, Xu F, Li G. Human Monoclonal Fab Antibodies Against West Nile Virus and its Neutralizing Activity Analyzed in Vitro and in Vivo. JOURNAL OF ANTIVIRALS & ANTIRETROVIRALS 2009; 1:36-42. [PMID: 20505850 PMCID: PMC2875541 DOI: 10.4172/jaa.1000005] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The disease progression with West Nile virus (WNV) infection in humans leads to meningitis or encephalitis and may cause death, particularly among elderly and immunocompromised individuals. Passive immunity using immunoglobulins has shown efficacy in treating some patients with WNV infection, which makes the development of human anti-WNV antibodies significant. The goal of this study was to construct a Fab-specific phage display library against WNV, and to identify and select clones with neutralizing activities. Total RNA was extracted from peripheral blood lymphocytes (PBLs) of two immunized individuals, and RT-PCR was used to amplify the Fab fragments containing the heavy (V(H)) and light (V(L)) chains. The amplified genes were sequentially cloned into the recombinant antibody expression vector pComb3-H, and the Fab-specific phage display library was packaged with helper phage VCS-M13. Five rounds of panning were carried out with WNV E protein domain III, and then binding antibodies were selected by ELISA. Antigen binding specificity, complementarity determining region (CDR) sequence of V(H) and V(L), and neutralizing activity against WNV were analyzed in vitro and in vivo. Eight Fab monoclonal antibodies recognized E protein domain III from a library of 7×10(7) clones/ml. Of the eight, one (Fab 1), exhibited significant neutralizing activity, and completely blocked 100 pfu WNV infection in Vero cells at a concentration 160 μg/ml. In contrast, Fab 13 and Fab 25, showed weaker neutralizing activities, and modestly blocked 100 pfu WNV infections at concentrations of 320 μg/ml and 160 μg/ml, respectively. However, animal studies showed that Fab 1 failed to protect mice from death at the concentration of 160μg/ml indicating that the neutralizing potential of an antibody in vivo is determined by the strength of binding and the abundance of its epitope for the virion.
Collapse
Affiliation(s)
- Tao Duan
- Department of Pathology and Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA 77555-0435
| | - Monique Ferguson
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA 77555-0435
| | - Lintian Yuan
- Department of Pediatric Dentistry, College of Stomatology, Fourth Military Medical University, Xi’an, China 710032
| | - Fangling Xu
- Department of Pathology and Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA 77555-0435
| | - Guangyu Li
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA 77555-0435
| |
Collapse
|
30
|
Tang QH, Zhang YM, Xu YZ, He L, Dai C, Sun P. Up-regulation of integrin beta3 expression in porcine vascular endothelial cells cultured in vitro by classical swine fever virus. Vet Immunol Immunopathol 2009; 133:237-42. [PMID: 19767110 DOI: 10.1016/j.vetimm.2009.07.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2009] [Revised: 04/23/2009] [Accepted: 07/01/2009] [Indexed: 11/26/2022]
Abstract
Classical swine fever (CSF) caused by virulent strains of classical swine fever virus (CSFV) is a haemorrhagic disease of pigs, characterized by disseminated intravascular coagulation, thrombocytopenia and immunosuppression. The cell adhesion molecule, integrin beta3, plays a central role in maintaining and regulating vascular permeability. In view of the haemorrhagic pathology of the disease, the effect of CSFV infection on integrin beta3 expression was investigated using the swine umbilical vein endothelial cell (SUVEC) line, in conjunction with quantitative PCR and Western blotting techniques. Following infection, the expression levels of integrin beta3 were significantly up-regulated along with corresponding transcription levels. The infected endothelial cells adhered onto immobilized extracellular matrix (ECM) with more extensive spreading than that of the control, and such interaction was strongly inhibited by an anti-integrin beta3 monoclonal antibody (mAb). This study revealed the up-regulation of integrin beta3 in vascular endothelial cells by CSFV infection, and cell adhesion molecules of this kind possibly play an important role in the changes of haemostatic balance in haemorrhagic pathology of CSF.
Collapse
Affiliation(s)
- Qing-Hai Tang
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China
| | | | | | | | | | | |
Collapse
|
31
|
Choi Y, Kwon YC, Kim SI, Park JM, Lee KH, Ahn BY. A hantavirus causing hemorrhagic fever with renal syndrome requires gC1qR/p32 for efficient cell binding and infection. Virology 2008; 381:178-83. [PMID: 18834607 DOI: 10.1016/j.virol.2008.08.035] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2008] [Revised: 06/09/2008] [Accepted: 08/19/2008] [Indexed: 11/18/2022]
Abstract
Hantaan virus (HTNV) is a pathogenic hantavirus that causes hemorrhagic fever with renal syndrome (HFRS). HTNV infection is mediated by alpha v beta3 integrin. We used protein blots of Vero E6 cell homogenates to demonstrate that radiolabeled HTNV virions bind to gC1qR/p32, the acidic 32-kDa protein known as the receptor for the globular head domain of complement C1q. RNAi-mediated suppression of gC1qR/p32 markedly reduced HTNV binding and infection in human lung epithelial A549 cells. Conversely, transient expression of either simian or human gC1qR/p32 rendered non-permissive CHO cells susceptible to HTNV infection. These results suggest an important role for gC1qR/p32 in HTNV infection and pathogenesis.
Collapse
Affiliation(s)
- Yun Choi
- Mogam Research Institute, 341 Pojungdong, Yongin, 449-910, Republic of Korea
| | | | | | | | | | | |
Collapse
|
32
|
Liu Z, Gao M, Han Q, Fang J, Zhao Q, Zhang N. Intensity of platelet beta(3) integrin in patients with hemorrhagic fever with renal syndrome and its correlation with disease severity. Viral Immunol 2008; 21:255-262. [PMID: 18419255 DOI: 10.1089/vim.2007.0098] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
beta(3) Integrin has been identified as a cellular receptor for Hantaan virus, which causes hemorrhagic fever with renal syndrome (HFRS). To investigate the relationship between intensity of the platelet membrane beta(3) integrin (CD61) and disease severity, the percentage of CD61-positive platelets and the mean fluorescence intensities (MFI) of platelet CD61 were determined in patients with HFRS by flow cytometry. The intensity levels of CD61 in patients with HFRS were significantly higher than those in the controls and correlated with the clinical phases of the disease. The CD61 intensity at the oliguric phase was inversely correlated with platelet count and serum albumin, and positively correlated with white blood cell count, blood urea nitrogen, serum creatinine, and alanine aminotransferase levels. The results suggest that the intensity levels of platelet CD61 were elevated and associated with clinical phases and disease severity in patients with HFRS, and the intensity of platelet beta(3) integrin in patients with HFRS may be indicative of disease severity.
Collapse
Affiliation(s)
- Zhengwen Liu
- Department of Infectious Diseases, First Affiliated Hospital, School of Medicine, Xian Jiaotong University, Xi'an, Shaanxi, China.
| | | | | | | | | | | |
Collapse
|
33
|
Li DF, Zhang MC, Yang HJ, Zhu YB, Xu X. Beta-integrin mediates WSSV infection. Virology 2007; 368:122-32. [PMID: 17655902 DOI: 10.1016/j.virol.2007.06.027] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2007] [Revised: 05/21/2007] [Accepted: 06/19/2007] [Indexed: 12/19/2022]
Abstract
White Spot Syndrome Virus (WSSV) is a virulent and widespread dsDNA virus with a wide range of hosts. Although remarkable progress has been made on virus characterization, however, its mechanism of infection is poorly understood. In this study, by analyzing the phage display library of the WSSV genome, a WSSV envelope protein VP187 (wsv209) was found to interact with shrimp integrin. VP187 possesses the RGD motif. The interaction between integrin and VP187 was confirmed with coimmunoprecipitation. These results demonstrate for the first time an interaction between the WSSV envelope protein and a cell surface molecule. Soluble integrin, integrin-specific antibody and an RGD-containing peptide were found to block the WSSV infection in vivo and in vitro. Gene silencing using a sequence-specific dsRNA targeting beta-integrin effectively inhibited the virus infection. These findings suggest that beta-integrin may function as a cellular receptor for WSSV infection.
Collapse
Affiliation(s)
- Deng-Feng Li
- Key Laboratory of Marine Biogenetic Resources, Third Institute of Oceanography, State Oceanic Administration, Xiamen 361005, P.R. China.
| | | | | | | | | |
Collapse
|
34
|
Connolly-Andersen AM, Magnusson KE, Mirazimi A. Basolateral entry and release of Crimean-Congo hemorrhagic fever virus in polarized MDCK-1 cells. J Virol 2006; 81:2158-64. [PMID: 17166898 PMCID: PMC1865934 DOI: 10.1128/jvi.02070-06] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is an etiological agent of a disease with mortality rates in patients averaging 30%. The disease is characterized by fever, myalgia, and hemorrhage. Mechanisms underlying the hemorrhage have to our knowledge not been elucidated for CCHFV. Possibly, a direct or indirect viral effect on tight junctions (TJ) could cause the hemorrhage observed in patients, as TJ play a crucial role in vascular homeostasis and can cause leakage upon deregulation. Moreover, there is no knowledge regarding the site of entry and release of CCHFV in polarized epithelial cells. Such cells represent a barrier to virus dissemination within the host, and as a site of viral entry and release, they could play a key role in further spread. For the first time, we have shown preferential basolateral entry of CCHFV in Madin-Darby canine kidney 1 (MDCK-1) epithelial cells. Furthermore, we demonstrated basolateral release of CCHFV in polarized epithelial cells. Interestingly, by measuring transepithelial electrical resistance, we found no effect of CCHFV replication on the function of TJ in this study. Neither did we observe any difference in the localization of the TJ proteins ZO-1 and occludin in CCHFV-infected cells compared to mock-infected cells.
Collapse
Affiliation(s)
- Anne-Marie Connolly-Andersen
- Swedish Institute for Infectious Disease Control, Centre for Microbiological Preparedness, SE-171 82 Solna, Sweden
| | | | | |
Collapse
|