1
|
Khasa R, Ogden SC, Wang Y, Mou Z, Metzler AD, Xie X, Dai X, Tang H. A single mutation in the PrM gene of Zika virus determines AXL dependency for infection of human neural cells. J Virol 2025; 99:e0187324. [PMID: 40062839 PMCID: PMC11998517 DOI: 10.1128/jvi.01873-24] [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: 10/25/2024] [Accepted: 02/13/2025] [Indexed: 04/16/2025] Open
Abstract
Zika virus (ZIKV) is spread by mosquito bites and is unique among known flaviviruses for being able to cause microcephaly. Entry factors for ZIKV are incompletely understood, but phosphatidylserine (PS) receptors, including the TAM (Tyro3, AXL, and Mer) and TIM (T-cell Ig mucin) families, can serve as cofactors for flavivirus entry in a cell type-specific manner. We identify AXL as the top hit in a CRISPR/Cas9 genome-wide screen in human glioblastoma cells and establish a definitive role of AXL, but not TYRO3 or MerTK, for ZIKV infection. Additionally, Spondweni virus also shows AXL dependency, while dengue virus infection is not affected by AXL knockout. Passage of ZIKV in AXL knockout (KO) cells generated a mutant virus capable of infection via AXL-independent mechanisms, and multiple independent selections identified a common mutation, H83R, in the prM coding region of the ZIKV genome. The mutant virus exhibits an increased infectivity rate in AXL KO cells as compared to wild-type ZIKV and is dependent upon the single H83R mutation. The mutant virus' ability to infect cells in an AXL-independent manner is unrelated to interferon signaling antagonism but likely pertains to a change in virus maturation that leads to a structural disturbance of the ZIKV virion. Our study provides evidence for a potential mechanism linking the viral structural proteins and host PS receptor usage during flavivirus infection.IMPORTANCEA major challenge in elucidating the mechanism of Zika virus (ZIKV) pathogenesis is the multitude of cell types it infects with distinct requirements. The role of phosphatidylserine (PS) receptors in ZIKV infection is cell type-specific, and the controversy surrounds their function in flavivirus entry. Here, we establish a definitive requirement of AXL for infection of human glioblastoma cells by both Zika and Spondweni virus. We then identified a single amino acid mutation (H83R) in the prM protein of ZIKV that allowed AXL-independent infection of these cells. The H83R-mediated escape of AXL requirement is independent of interferon (IFN) signaling suppression by AXL; instead, the mutation has the potential to disrupt the virus assembly and virion structure. This study reveals a previously unknown connection between the PS receptor usage and the flavivirus prM gene, which can guide detailed molecular mechanism studies of the interplay between virion assembly and virus entry.
Collapse
Affiliation(s)
- Renu Khasa
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Sarah C. Ogden
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Yuqing Wang
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Zongiun Mou
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Anna D. Metzler
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Xuping Xie
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Xinghong Dai
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Hengli Tang
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
2
|
Siwak KC, LeBlanc EV, Scott HM, Kim Y, Pellizzari-Delano I, Ball AM, Temperton NJ, Capicciotti CJ, Colpitts CC. Cellular sialoglycans are differentially required for endosomal and cell-surface entry of SARS-CoV-2 in lung cell lines. PLoS Pathog 2024; 20:e1012365. [PMID: 39625989 PMCID: PMC11642992 DOI: 10.1371/journal.ppat.1012365] [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: 06/22/2024] [Revised: 12/13/2024] [Accepted: 11/17/2024] [Indexed: 12/14/2024] Open
Abstract
Cell entry of severe acute respiratory coronavirus-2 (SARS-CoV-2) and other CoVs can occur via two distinct routes. Following receptor binding by the spike glycoprotein, membrane fusion can be triggered by spike cleavage either at the cell surface in a transmembrane serine protease 2 (TMPRSS2)-dependent manner or within endosomes in a cathepsin-dependent manner. Cellular sialoglycans have been proposed to aid in CoV attachment and entry, although their functional contributions to each entry pathway are unknown. In this study, we used genetic and enzymatic approaches to deplete sialic acid from cell surfaces and compared the requirement for sialoglycans during endosomal and cell-surface CoV entry using lentiviral particles pseudotyped with the spike proteins of different sarbecoviruses. We show that entry of SARS-CoV-1, WIV1-CoV and WIV16-CoV, like the SARS-CoV-2 omicron variant, depends on endosomal cathepsins and requires cellular sialoglycans for entry. Ancestral SARS-CoV-2 and the delta variant can use either pathway for entry, but only require sialic acid for endosomal entry in cells lacking TMPRSS2. Binding of SARS-CoV-2 spike protein to cells did not require sialic acid, nor was sialic acid required for SARS-CoV-2 entry in TMRPSS2-expressing cells. These findings suggest that cellular sialoglycans are not strictly required for SARS-CoV-2 attachment, receptor binding or fusion, but rather promote endocytic entry of SARS-CoV-2 and related sarbecoviruses. In contrast, the requirement for sialic acid during entry of MERS-CoV pseudoparticles and authentic HCoV-OC43 was not affected by TMPRSS2 expression, consistent with a described role for sialic acid in merbecovirus and embecovirus cell attachment. Overall, these findings clarify the role of sialoglycans in SARS-CoV-2 entry and suggest that cellular sialoglycans mediate endosomal, but not cell-surface, SARS-CoV-2 entry.
Collapse
Affiliation(s)
- Kimberley C. Siwak
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Canada
| | - Emmanuelle V. LeBlanc
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Canada
| | - Heidi M. Scott
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Canada
| | - Youjin Kim
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Canada
| | | | - Alice M. Ball
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Canada
| | - Nigel J. Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent and Greenwich at Medway, Chatham, United Kingdom
| | - Chantelle J. Capicciotti
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Canada
- Department of Chemistry, Queen’s University, Kingston, Canada
- Department of Surgery, Queen’s University, Kingston, Canada
| | - Che C. Colpitts
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Canada
| |
Collapse
|
3
|
Li SY, Kumar S, Gu X, DeFalco T. Testicular immunity. Mol Aspects Med 2024; 100:101323. [PMID: 39591799 PMCID: PMC11624985 DOI: 10.1016/j.mam.2024.101323] [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: 04/02/2024] [Revised: 11/13/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024]
Abstract
The testis is a unique environment where immune responses are suppressed to allow the development of sperm that possess autoimmunogenic antigens. There are several contributors responsible for testicular immune privilege, including the blood-testis barrier, testicular immune cells, immunomodulation by Sertoli cells, and high levels of steroid hormones. Despite multiple mechanisms in place to regulate the testicular immune environment, pathogens that disrupt testicular immunity can lead to long-term effects such as infertility. If testicular immunity is disturbed, autoimmune reactions can also occur, leading to aberrant immune cell infiltration and subsequent attack of autoimmunogenic germ cells. Here we discuss cellular and molecular factors underlying testicular immunity and how testicular infection or autoimmunity compromise immune privilege. We also describe infections and autoimmune diseases that impact the testis. Further research into testicular immunity will reveal how male fertility is maintained and will help update therapeutic strategies for infertility and other testicular disorders.
Collapse
Affiliation(s)
- Shu-Yun Li
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Sudeep Kumar
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Xiaowei Gu
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Tony DeFalco
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
4
|
Amin MR, Anwar KN, Ashraf MJ, Ghassemi M, Novak RM. Preventing human influenza and coronaviral mono or coinfection by blocking virus-induced sialylation. Antiviral Res 2024; 232:106041. [PMID: 39581502 DOI: 10.1016/j.antiviral.2024.106041] [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: 06/29/2024] [Revised: 11/12/2024] [Accepted: 11/20/2024] [Indexed: 11/26/2024]
Abstract
Influenza A viruses (IAVs) and endemic coronaviruses (eCoVs) are common etiologic agents for seasonal respiratory infections. The human H1N1 of IAV and coronavirus OC43 (HCoV-OC43) can result in hospitalization, acute respiratory distress syndrome (ARDS), and even death, particularly in immunocompromised individuals. They infect the epithelium of the respiratory tract by interacting with host cell sialic acid (Sia)- linked receptors whose synthesis is catalyzed by sialyltransferases (STs). Viral coinfection is challenging to treat because of the need to target specific components of two or more distinct pathogens. Emerging drug and vaccine resistance due to the high mutation rate of viral genomes further complicates the treatment and prevention of viral infection. Sialylation mediated by STs may be a potential drug target for treating viral diseases. ST is an attractive target because it could be effective before identifying the pathogen that has occurred, providing a novel direction for overcoming drug resistance and achieving a broad-spectrum antiviral effect. We developed an H1N1 and OC43 mono or coinfection model using 14 days post-plating (14 PP) human primary small airway epithelial cells (HSAEC) grown on transwell inserts at an air-fluid interface (ALI), mimicking in vivo cellular dynamics. Using this model, we have observed that mono or coinfection with OC43 and H1N1 results in increased sialic acid levels and synergistic viral infection. We showed for the first time that H1N1 and OC43 mono- and coinfection in HSAEC caused increased expression and activity of STs, which can be blocked by pan-STs inhibitor (3Fax-Peracetyl Neu5Ac) with no host cell toxicity.
Collapse
Affiliation(s)
- Md Ruhul Amin
- Division of Infectious Diseases, Department of Medicine, University of Illinois Chicago, Illinois, USA.
| | - Khandaker N Anwar
- Division of Infectious Diseases, Department of Medicine, University of Illinois Chicago, Illinois, USA
| | - M J Ashraf
- Department of Ophthalmology and Visual Sciences, University of Illinois Chicago, Illinois, USA
| | - Mahmood Ghassemi
- Division of Infectious Diseases, Department of Medicine, University of Illinois Chicago, Illinois, USA
| | - Richard M Novak
- Division of Infectious Diseases, Department of Medicine, University of Illinois Chicago, Illinois, USA
| |
Collapse
|
5
|
He Y, Miao C, Yang S, Xu C, Liu Y, Zhu X, Wen Y, Wu R, Zhao Q, Huang X, Yan Q, Lang Y, Zhao S, Wang Y, Han X, Cao S, Hu Y, Du S. Sialic acids as attachment factors in mosquitoes mediating Japanese encephalitis virus infection. J Virol 2024; 98:e0195923. [PMID: 38634598 PMCID: PMC11092328 DOI: 10.1128/jvi.01959-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/26/2024] [Indexed: 04/19/2024] Open
Abstract
The role of Culex mosquitoes in the transmission of Japanese encephalitis virus (JEV) is crucial, yet the mechanisms of JEV infection in these vectors remain unclear. Previous research has indicated that various host factors participate in JEV infection. Herein, we present evidence that mosquito sialic acids enhance JEV infection both in vivo and in vitro. By treating mosquitoes and C6/36 cells with neuraminidase or lectin, the function of sialic acids is effectively blocked, resulting in significant inhibition of JEV infection. Furthermore, knockdown of the sialic acid biosynthesis genes in Culex mosquitoes also leads to a reduction in JEV infection. Moreover, our research revealed that sialic acids play a role in the attachment of JEV to mosquito cells, but not in its internalization. To further explore the mechanisms underlying the promotion of JEV attachment by sialic acids, we conducted immunoprecipitation experiments to confirm the direct binding of sialic acids to the last α-helix in JEV envelope protein domain III. Overall, our study contributes to a molecular comprehension of the interaction between mosquitoes and JEV and offers potential strategies for preventing the dissemination of flavivirus in natural environments.IMPORTANCEIn this study, we aimed to investigate the impact of glycoconjugate sialic acids on mosquito infection with Japanese encephalitis virus (JEV). Our findings demonstrate that sialic acids play a crucial role in enhancing JEV infection by facilitating the attachment of the virus to the cell membrane. Furthermore, our investigation revealed that sialic acids directly bind to the final α-helix in the JEV envelope protein domain III, thereby accelerating virus adsorption. Collectively, our results highlight the significance of mosquito sialic acids in JEV infection within vectors, contributing to a better understanding of the interaction between mosquitoes and JEV.
Collapse
Affiliation(s)
- Yi He
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Chang Miao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Shiping Yang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Changhao Xu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yuwei Liu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xi Zhu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yiping Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Rui Wu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Qin Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Xiaobo Huang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Qigui Yan
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Yifei Lang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Shan Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Yiping Wang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Xinfeng Han
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Sanjie Cao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Yajie Hu
- Sichuan Center for Disease Control and Prevention, Chengdu, China
| | - Senyan Du
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| |
Collapse
|
6
|
Siew ZY, Tan YF, Iswara RP, Wong SF, Wong ST, Tan BK, Leong PP, Tan CW, Wang LF, Leong CO, Voon K. Human cytokeratin 1 plays a role in the interaction of Pteropine orthoreovirus with Hek293 cells but not HeLa cells. Microbes Infect 2024; 26:105243. [PMID: 38380604 DOI: 10.1016/j.micinf.2023.105243] [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: 07/21/2023] [Revised: 09/15/2023] [Accepted: 10/26/2023] [Indexed: 02/22/2024]
Abstract
Pteropine orthoreovirus (PRV) causes respiratory tract infections in humans. Despite its emergence as a zoonotic and respiratory virus, little is known about its cell tropism, which hampers progress in fully understanding its pathogenesis in humans. Hek293 cells are most susceptible to PRV infection, while HeLa cells are the least. Human cytokeratin 1 (CK1) was identified as the protein that interacts with PRV. The immunofluorescence assay and qPCR results revealed prior treatment with anti-CK1 may provide Hek293 cells protection against PRV. The KRT1-knockout Hek293 cells were less susceptible to PRV infection. Further study into the pathogenesis of PRV in humans is needed.
Collapse
Affiliation(s)
- Zhen Yun Siew
- School of Pharmacy, University of Nottingham Malaysia, Semenyih, Malaysia
| | - Yeh Fong Tan
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia; Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | | | - Shew Fung Wong
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia; Institute for Research, Development and Innovation (IRDI), International Medical University, Kuala Lumpur, Malaysia
| | - Siew Tung Wong
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Boon Keat Tan
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Pooi Pooi Leong
- Faculty of Medicine and Health Sciences, University Tunku Abdul Rahman, Sg Long, Malaysia
| | - Chee Wah Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, 169857, Singapore; Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lin-Fa Wang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, 169857, Singapore
| | - Chee Onn Leong
- Institute for Research, Development and Innovation (IRDI), International Medical University, Kuala Lumpur, Malaysia; AGTC Genomics, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Kenny Voon
- School of Pharmacy, University of Nottingham Malaysia, Semenyih, Malaysia; School of Medicine, International Medical University, Kuala Lumpur, Malaysia.
| |
Collapse
|
7
|
Diani E, Lagni A, Lotti V, Tonon E, Cecchetto R, Gibellini D. Vector-Transmitted Flaviviruses: An Antiviral Molecules Overview. Microorganisms 2023; 11:2427. [PMID: 37894085 PMCID: PMC10608811 DOI: 10.3390/microorganisms11102427] [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: 08/17/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Flaviviruses cause numerous pathologies in humans across a broad clinical spectrum with potentially severe clinical manifestations, including hemorrhagic and neurological disorders. Among human flaviviruses, some viral proteins show high conservation and are good candidates as targets for drug design. From an epidemiological point of view, flaviviruses cause more than 400 million cases of infection worldwide each year. In particular, the Yellow Fever, dengue, West Nile, and Zika viruses have high morbidity and mortality-about an estimated 20,000 deaths per year. As they depend on human vectors, they have expanded their geographical range in recent years due to altered climatic and social conditions. Despite these epidemiological and clinical premises, there are limited antiviral treatments for these infections. In this review, we describe the major compounds that are currently under evaluation for the treatment of flavivirus infections and the challenges faced during clinical trials, outlining their mechanisms of action in order to present an overview of ongoing studies. According to our review, the absence of approved antivirals for flaviviruses led to in vitro and in vivo experiments aimed at identifying compounds that can interfere with one or more viral cycle steps. Still, the currently unavailability of approved antivirals poses a significant public health issue.
Collapse
Affiliation(s)
- Erica Diani
- Department of Diagnostic and Public Health, Microbiology Section, University of Verona, 37134 Verona, Italy; (A.L.); (V.L.); (R.C.)
| | - Anna Lagni
- Department of Diagnostic and Public Health, Microbiology Section, University of Verona, 37134 Verona, Italy; (A.L.); (V.L.); (R.C.)
| | - Virginia Lotti
- Department of Diagnostic and Public Health, Microbiology Section, University of Verona, 37134 Verona, Italy; (A.L.); (V.L.); (R.C.)
| | - Emil Tonon
- Unit of Microbiology, Azienda Ospedaliera Universitaria Integrata Verona, 37134 Verona, Italy;
| | - Riccardo Cecchetto
- Department of Diagnostic and Public Health, Microbiology Section, University of Verona, 37134 Verona, Italy; (A.L.); (V.L.); (R.C.)
- Unit of Microbiology, Azienda Ospedaliera Universitaria Integrata Verona, 37134 Verona, Italy;
| | - Davide Gibellini
- Department of Diagnostic and Public Health, Microbiology Section, University of Verona, 37134 Verona, Italy; (A.L.); (V.L.); (R.C.)
- Unit of Microbiology, Azienda Ospedaliera Universitaria Integrata Verona, 37134 Verona, Italy;
| |
Collapse
|
8
|
Cao Y, Song W, Chen X. Multivalent sialic acid materials for biomedical applications. Biomater Sci 2023; 11:2620-2638. [PMID: 36661319 DOI: 10.1039/d2bm01595a] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Sialic acid is a kind of monosaccharide expressed on the non-reducing end of glycoproteins or glycolipids. It acts as a signal molecule combining with its natural receptors such as selectins and siglecs (sialic acid-binding immunoglobulin-like lectins) in intercellular interactions like immunological surveillance and leukocyte infiltration. The last few decades have witnessed the exploration of the roles that sialic acid plays in different physiological and pathological processes and the use of sialic acid-modified materials as therapeutics for related diseases like immune dysregulation and virus infection. In this review, we will briefly introduce the biomedical function of sialic acids in organisms and the utilization of multivalent sialic acid materials for targeted drug delivery as well as therapeutic applications including anti-inflammation and anti-virus.
Collapse
Affiliation(s)
- Yusong Cao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China. .,University of Science and Technology of China, Hefei, 230026, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China. .,University of Science and Technology of China, Hefei, 230026, China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China. .,University of Science and Technology of China, Hefei, 230026, China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| |
Collapse
|
9
|
Carbohydrates: Binding Sites and Potential Drug Targets for Neural-Affecting Pathogens. ADVANCES IN NEUROBIOLOGY 2023; 29:449-477. [DOI: 10.1007/978-3-031-12390-0_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
10
|
Wang F, Zhang J, Wang Y, Chen Y, Han D. Viral tropism for the testis and sexual transmission. Front Immunol 2022; 13:1040172. [PMID: 36439102 PMCID: PMC9682072 DOI: 10.3389/fimmu.2022.1040172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/24/2022] [Indexed: 10/17/2023] Open
Abstract
The mammalian testis adopts an immune privileged environment to protect male germ cells from adverse autoimmune reaction. The testicular immune privileged status can be also hijacked by various microbial pathogens as a sanctuary to escape systemic immune surveillance. In particular, several viruses have a tropism for the testis. To overcome the immune privileged status and mount an effective local defense against invading viruses, testicular cells are well equipped with innate antiviral machinery. However, several viruses may persist an elongated duration in the testis and disrupt the local immune homeostasis, thereby impairing testicular functions and male fertility. Moreover, the viruses in the testis, as well as other organs of the male reproductive system, can shed to the semen, thus allowing sexual transmission to partners. Viral infection in the testis, which can impair male fertility and lead to sexual transmission, is a serious concern in research on known and on new emerging viruses. To provide references for our scientific peers, this article reviews research achievements and suggests future research focuses in the field.
Collapse
Affiliation(s)
| | | | | | - Yongmei Chen
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Daishu Han
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| |
Collapse
|
11
|
Li K, Ji Q, Jiang S, Zhang N. Advancement in the Development of Therapeutics Against Zika Virus Infection. Front Cell Infect Microbiol 2022; 12:946957. [PMID: 35880081 PMCID: PMC9307976 DOI: 10.3389/fcimb.2022.946957] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Zika virus (ZIKV), a re-emerging arbovirus, causes teratogenic effects on the fetus and normal nerve functions, resulting in harmful autoimmune responses, which call for the development of therapeutics against ZIKV infection. In this review, we introduce the pathogenesis of ZIKV infection and summarize the advancement in the development of therapeutics against ZIKV infection. It provides guidance for the development of effective therapeutics against ZIKV infection.
Collapse
Affiliation(s)
- Kangchen Li
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Qianting Ji
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of Ministry of Education (MOE), National Health Commission (NHC) and Chinese Academy of Medical Sciences (CAMS), School of Basic Medical Sciences and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- *Correspondence: Shibo Jiang, ; Naru Zhang,
| | - Naru Zhang
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, China
- *Correspondence: Shibo Jiang, ; Naru Zhang,
| |
Collapse
|
12
|
Wielgat P, Narejko K, Car H. SARS-CoV-2 Attacks in the Brain: Focus on the Sialome. Cells 2022; 11:1458. [PMID: 35563764 PMCID: PMC9104523 DOI: 10.3390/cells11091458] [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: 03/22/2022] [Revised: 04/20/2022] [Accepted: 04/24/2022] [Indexed: 12/16/2022] Open
Abstract
The epidemiological observations suggest that respiratory and gastrointestinal symptoms caused by severe acute respiratory coronavirus 2 (SARS-CoV-2) are accompanied by short- and long-term neurological manifestations. There is increasing evidence that the neuroinvasive potential of SARS-CoV-2 is closely related to its capacity to interact with cell membrane sialome. Given the wide expression of sialylated compounds of cell membranes in the brain, the interplay between cell membrane sialoglycans and the virus is crucial for its attachment and cell entry, transport, neuronal damage and brain immunity. Here, we focus on the significance of the brain sialome in the progress of coronavirus disease 2019 (COVID-19) and SARS-CoV-2-induced neuropathology.
Collapse
Affiliation(s)
- Przemyslaw Wielgat
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15-274 Bialystok, Poland; (K.N.); (H.C.)
| | - Karolina Narejko
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15-274 Bialystok, Poland; (K.N.); (H.C.)
| | - Halina Car
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15-274 Bialystok, Poland; (K.N.); (H.C.)
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-265 Bialystok, Poland
| |
Collapse
|
13
|
Komarasamy TV, Adnan NAA, James W, Balasubramaniam VRMT. Zika Virus Neuropathogenesis: The Different Brain Cells, Host Factors and Mechanisms Involved. Front Immunol 2022; 13:773191. [PMID: 35371036 PMCID: PMC8966389 DOI: 10.3389/fimmu.2022.773191] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 02/21/2022] [Indexed: 12/16/2022] Open
Abstract
Zika virus (ZIKV), despite being discovered six decades earlier, became a major health concern only after an epidemic in French Polynesia and an increase in the number of microcephaly cases in Brazil. Substantial evidence has been found to support the link between ZIKV and neurological complications in infants. The virus targets various cells in the brain, including radial glial cells, neural progenitor cells (NPCs), astrocytes, microglial and glioblastoma stem cells. It affects the brain cells by exploiting different mechanisms, mainly through apoptosis and cell cycle dysregulation. The modulation of host immune response and the inflammatory process has also been demonstrated to play a critical role in ZIKV induced neurological complications. In addition to that, different ZIKV strains have exhibited specific neurotropism and unique molecular mechanisms. This review provides a comprehensive and up-to-date overview of ZIKV-induced neuroimmunopathogenesis by dissecting its main target cells in the brain, and the underlying cellular and molecular mechanisms. We highlighted the roles of the different ZIKV host factors and how they exploit specific host factors through various mechanisms. Overall, it covers key components for understanding the crosstalk between ZIKV and the brain.
Collapse
Affiliation(s)
- Thamil Vaani Komarasamy
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Nur Amelia Azreen Adnan
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - William James
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Vinod R M T Balasubramaniam
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
14
|
Tan LY, Komarasamy TV, James W, Balasubramaniam VRMT. Host Molecules Regulating Neural Invasion of Zika Virus and Drug Repurposing Strategy. Front Microbiol 2022; 13:743147. [PMID: 35308394 PMCID: PMC8931420 DOI: 10.3389/fmicb.2022.743147] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne, single-stranded RNA virus belonging to the genus Flavivirus. Although ZIKV infection is usually known to exhibit mild clinical symptoms, intrauterine ZIKV infections have been associated with severe neurological manifestations, including microcephaly and Guillain Barre syndrome (GBS). Therefore, it is imperative to understand the mechanisms of ZIKV entry into the central nervous system (CNS) and its effect on brain cells. Several routes of neuro-invasion have been identified, among which blood–brain barrier (BBB) disruption is the commonest mode of access. The molecular receptors involved in viral entry remain unknown; with various proposed molecular ZIKV-host interactions including potential non-receptor mediated cellular entry. As ZIKV invade neuronal cells, they trigger neurotoxic mechanisms via cell-autonomous and non-cell autonomous pathways, resulting in neurogenesis dysfunction, viral replication, and cell death, all of which eventually lead to microcephaly. Together, our understanding of the biological mechanisms of ZIKV exposure would aid in the development of anti-ZIKV therapies targeting host cellular and/or viral components to combat ZIKV infection and its neurological manifestations. In this present work, we review the current understanding of ZIKV entry mechanisms into the CNS and its implications on the brain. We also highlight the status of the drug repurposing approach for the development of potential antiviral drugs against ZIKV.
Collapse
Affiliation(s)
- Li Yin Tan
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
- Greenslopes Private Hospital, Greenslopes, QLD, Australia
| | - Thamil Vaani Komarasamy
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - William James
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Vinod R. M. T. Balasubramaniam
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
- *Correspondence: Vinod R. M. T. Balasubramaniam,
| |
Collapse
|
15
|
Zwernik SD, Adams BH, Raymond DA, Warner CM, Kassam AB, Rovin RA, Akhtar P. AXL receptor is required for Zika virus strain MR-766 infection in human glioblastoma cell lines. Mol Ther Oncolytics 2021; 23:447-457. [PMID: 34901388 PMCID: PMC8626839 DOI: 10.1016/j.omto.2021.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/13/2021] [Accepted: 11/08/2021] [Indexed: 12/28/2022] Open
Abstract
Recent reports have shown that Zika virus (ZIKV) has oncolytic potential against human glioblastoma (GBM); however, the mechanisms underlying its tropism and cell entry are not completely understood. The receptor tyrosine kinase AXL has been identified as an entry receptor for ZIKV in a cell-type-specific manner. Interestingly, AXL is frequently overexpressed in GBM patients. Using commercially available GBM cell lines, we first show that cells expressing AXL are permissive for ZIKV infection, while cells that do not express AXL are not. Furthermore, inhibition of AXL kinase using R428 and antibody blockade of AXL receptor strongly attenuated virus entry in GBM cell lines. Additionally, CRISPR knockout of the AXL gene in GBM cell lines completely abolished ZIKV infection, significantly inhibited viral replication, and significantly reduced apoptosis compared with parental lines. Lastly, introduction of AXL receptor into non-expressing cell lines renders the cells susceptible to ZIKV infection. Together, these findings demonstrate that ZIKV entry into GBM cells in vitro is mediated by the AXL receptor and that following cell entry, productive infection is cytotoxic. Thus, ZIKV is a potential oncolytic virus for GBM.
Collapse
Affiliation(s)
- Samuel D Zwernik
- Advocate Aurora Research Institute, Advocate Aurora Health, Milwaukee, WI 53233, USA
| | - Beau H Adams
- Advocate Aurora Research Institute, Advocate Aurora Health, Milwaukee, WI 53233, USA
| | - Daniel A Raymond
- Advocate Aurora Research Institute, Advocate Aurora Health, Milwaukee, WI 53233, USA
| | - Catherine M Warner
- Advocate Aurora Research Institute, Advocate Aurora Health, Milwaukee, WI 53233, USA
| | - Amin B Kassam
- Aurora Neuroscience Innovation Institute, Advocate Aurora Health, Milwaukee, WI 53215, USA
| | - Richard A Rovin
- Aurora Neuroscience Innovation Institute, Advocate Aurora Health, Milwaukee, WI 53215, USA
| | - Parvez Akhtar
- Advocate Aurora Research Institute, Advocate Aurora Health, Milwaukee, WI 53233, USA
| |
Collapse
|
16
|
Zoladek J, Legros V, Jeannin P, Chazal M, Pardigon N, Ceccaldi PE, Gessain A, Jouvenet N, Afonso PV. Zika Virus Requires the Expression of Claudin-7 for Optimal Replication in Human Endothelial Cells. Front Microbiol 2021; 12:746589. [PMID: 34616388 PMCID: PMC8488266 DOI: 10.3389/fmicb.2021.746589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 08/23/2021] [Indexed: 01/31/2023] Open
Abstract
Zika virus (ZIKV) infection has been associated with a series of neurological pathologies. In patients with ZIKV-induced neurological disorders, the virus is detectable in the central nervous system. Thus, ZIKV is capable of neuroinvasion, presumably through infection of the endothelial cells that constitute the blood-brain barrier (BBB). We demonstrate that susceptibility of BBB endothelial cells to ZIKV infection is modulated by the expression of tight-junction protein claudin-7 (CLDN7). Downregulation of CLDN7 reduced viral RNA yield, viral protein production, and release of infectious viral particles in several endothelial cell types, but not in epithelial cells, indicating that CLDN7 implication in viral infection is cell-type specific. The proviral activity of CLDN7 in endothelial cells is ZIKV-specific since related flaviviruses were not affected by CLDN7 downregulation. Together, our data suggest that CLDN7 facilitates ZIKV infection in endothelial cells at a post-internalization stage and prior to RNA production. Our work contributes to a better understanding of the mechanisms exploited by ZIKV to efficiently infect and replicate in endothelial cells and thus of its ability to cross the BBB.
Collapse
Affiliation(s)
- Jim Zoladek
- Unité Épidémiologie et Physiopathologie des Virus Oncogènes, Institut Pasteur, Centre National de la Recherche Scientifique UMR 3569, Université de Paris, Paris, France
| | - Vincent Legros
- Unité Épidémiologie et Physiopathologie des Virus Oncogènes, Institut Pasteur, Centre National de la Recherche Scientifique UMR 3569, Université de Paris, Paris, France.,VetAgro Sup, Centre International de Recherche en Infectiologie (CIRI), Lyon, France
| | - Patricia Jeannin
- Unité Épidémiologie et Physiopathologie des Virus Oncogènes, Institut Pasteur, Centre National de la Recherche Scientifique UMR 3569, Université de Paris, Paris, France
| | - Maxime Chazal
- Unité Signalisation Antivirale, Institut Pasteur, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - Nathalie Pardigon
- Groupe Arbovirus, Unité Environnement et Risques Infectieux, Institut Pasteur, Paris, France
| | - Pierre-Emmanuel Ceccaldi
- Unité Épidémiologie et Physiopathologie des Virus Oncogènes, Institut Pasteur, Centre National de la Recherche Scientifique UMR 3569, Université de Paris, Paris, France
| | - Antoine Gessain
- Unité Épidémiologie et Physiopathologie des Virus Oncogènes, Institut Pasteur, Centre National de la Recherche Scientifique UMR 3569, Université de Paris, Paris, France
| | - Nolwenn Jouvenet
- Unité Signalisation Antivirale, Institut Pasteur, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - Philippe V Afonso
- Unité Épidémiologie et Physiopathologie des Virus Oncogènes, Institut Pasteur, Centre National de la Recherche Scientifique UMR 3569, Université de Paris, Paris, France
| |
Collapse
|
17
|
The epidermal growth factor receptor is a relevant host factor in the early stages of Zika virus life cycle in vitro. J Virol 2021; 95:e0119521. [PMID: 34379506 DOI: 10.1128/jvi.01195-21] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Zika virus (ZIKV) is a flavivirus well-known for the epidemic in the Americas in 2015-2016, where microcephaly in newborns and other neurological complications were connected to ZIKV infection. Many aspects of the viral life cycle, including binding and entry into the host cell, are still enigmatic. Based on the observation that CHO cells lack the expression of EGFR and are not permissive for various ZIKV strains, the relevance of EGFR for the viral life cycle was analyzed. Infection of A549 cells by ZIKV leads to a rapid internalization of EGFR that colocalizes with the endosomal marker EEA1. Moreover, the infection by different ZIKV strains is associated with an activation of EGFR and subsequent activation of the MAPK/ERK signaling cascade. However, treatment of the cells with MβCD, which on the one hand leads to an activation of EGFR but on the other hand prevents EGFR internalization, impairs ZIKV infection. Specific inhibition of EGFR or of the RAS-RAF-MEK-ERK signal transduction cascade hinders ZIKV infection by inhibition of ZIKV entry. In accordance to this, knockout of EGFR expression impedes ZIKV entry. In case of an already established infection, inhibition of EGFR or of downstream signaling does not affect viral replication. Taken together, these data demonstrate the relevance of EGFR in the early stages of ZIKV infection and identify EGFR as a target for antiviral strategies. Importance These data deepen the knowledge about the ZIKV infection process and demonstrate the relevance of EGFR for ZIKV entry. In light of the fact that a variety of specific and efficient inhibitors of EGFR and of EGFR-dependent signaling were developed and licensed, repurposing of these substances could be a helpful tool to prevent the spreading of ZIKV infection in an epidemic outbreak.
Collapse
|
18
|
Hu T, Wu Z, Wu S, Chen S, Cheng A. The key amino acids of E protein involved in early flavivirus infection: viral entry. Virol J 2021; 18:136. [PMID: 34217298 PMCID: PMC8254458 DOI: 10.1186/s12985-021-01611-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/29/2021] [Indexed: 02/11/2023] Open
Abstract
Flaviviruses are enveloped viruses that infect multiple hosts. Envelope proteins are the outermost proteins in the structure of flaviviruses and mediate viral infection. Studies indicate that flaviviruses mainly use envelope proteins to bind to cell attachment receptors and endocytic receptors for the entry step. Here, we present current findings regarding key envelope protein amino acids that participate in the flavivirus early infection process. Among these sites, most are located in special positions of the protein structure, such as the α-helix in the stem region and the hinge region between domains I and II, motifs that potentially affect the interaction between different domains. Some of these sites are located in positions involved in conformational changes in envelope proteins. In summary, we summarize and discuss the key envelope protein residues that affect the entry process of flaviviruses, including the process of their discovery and the mechanisms that affect early infection.
Collapse
Affiliation(s)
- Tao Hu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Zhen Wu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Shaoxiong Wu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Shun Chen
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan, China. .,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan, China. .,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan, China.
| | - Anchun Cheng
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan, China. .,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan, China. .,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
19
|
Hao W, Ma B, Li Z, Wang X, Gao X, Li Y, Qin B, Shang S, Cui S, Tan Z. Binding of the SARS-CoV-2 spike protein to glycans. Sci Bull (Beijing) 2021; 66:1205-1214. [PMID: 33495714 PMCID: PMC7816574 DOI: 10.1016/j.scib.2021.01.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/29/2020] [Accepted: 01/11/2021] [Indexed: 12/19/2022]
Abstract
The pandemic of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a high number of deaths in the world. To combat it, it is necessary to develop a better understanding of how the virus infects host cells. Infection normally starts with the attachment of the virus to cell-surface glycans like heparan sulfate (HS) and sialic acid-containing glycolipids/glycoproteins. In this study, we examined and compared the binding of the subunits and spike (S) proteins of SARS-CoV-2, SARS-CoV, and Middle East respiratory disease (MERS)-CoV to these glycans. Our results revealed that the S proteins and subunits can bind to HS in a sulfation-dependent manner and no binding with sialic acid residues was detected. Overall, this work suggests that HS binding may be a general mechanism for the attachment of these coronaviruses to host cells, and supports the potential importance of HS in infection and in the development of antiviral agents against these viruses.
Collapse
Affiliation(s)
- Wei Hao
- NHC Key Laboratory of Systems Biology of Pathogens, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Bo Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ziheng Li
- NHC Key Laboratory of Systems Biology of Pathogens, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xiaoyu Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiaopan Gao
- NHC Key Laboratory of Systems Biology of Pathogens, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yaohao Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado, Boulder CO 80303, USA
| | - Bo Qin
- NHC Key Laboratory of Systems Biology of Pathogens, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Shiying Shang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Sheng Cui
- NHC Key Laboratory of Systems Biology of Pathogens, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Zhongping Tan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
20
|
Lloyd MD, Yevglevskis M, Nathubhai A, James TD, Threadgill MD, Woodman TJ. Racemases and epimerases operating through a 1,1-proton transfer mechanism: reactivity, mechanism and inhibition. Chem Soc Rev 2021; 50:5952-5984. [PMID: 34027955 PMCID: PMC8142540 DOI: 10.1039/d0cs00540a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Indexed: 12/12/2022]
Abstract
Racemases and epimerases catalyse changes in the stereochemical configurations of chiral centres and are of interest as model enzymes and as biotechnological tools. They also occupy pivotal positions within metabolic pathways and, hence, many of them are important drug targets. This review summarises the catalytic mechanisms of PLP-dependent, enolase family and cofactor-independent racemases and epimerases operating by a deprotonation/reprotonation (1,1-proton transfer) mechanism and methods for measuring their catalytic activity. Strategies for inhibiting these enzymes are reviewed, as are specific examples of inhibitors. Rational design of inhibitors based on substrates has been extensively explored but there is considerable scope for development of transition-state mimics and covalent inhibitors and for the identification of inhibitors by high-throughput, fragment and virtual screening approaches. The increasing availability of enzyme structures obtained using X-ray crystallography will facilitate development of inhibitors by rational design and fragment screening, whilst protein models will facilitate development of transition-state mimics.
Collapse
Affiliation(s)
- Matthew D Lloyd
- Drug & Target Discovery, Department of Pharmacy & Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK.
| | - Maksims Yevglevskis
- Drug & Target Discovery, Department of Pharmacy & Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK. and CatSci Ltd., CBTC2, Capital Business Park, Wentloog, Cardiff CF3 2PX, UK
| | - Amit Nathubhai
- Drug & Target Discovery, Department of Pharmacy & Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK. and University of Sunderland, School of Pharmacy & Pharmaceutical Sciences, Sciences Complex, Sunderland SR1 3SD, UK
| | - Tony D James
- Department of Chemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK and School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, People's Republic of China
| | - Michael D Threadgill
- Drug & Target Discovery, Department of Pharmacy & Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK. and Institute of Biological, Environmental & Rural Sciences, Aberystwyth University, Aberystwyth SY23 3BY, UK
| | - Timothy J Woodman
- Drug & Target Discovery, Department of Pharmacy & Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK.
| |
Collapse
|
21
|
Gist of Zika Virus pathogenesis. Virology 2021; 560:86-95. [PMID: 34051478 DOI: 10.1016/j.virol.2021.04.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/03/2021] [Accepted: 04/28/2021] [Indexed: 12/29/2022]
Abstract
Zika virus (ZIKV) is a mosquito-borne neurotropic flavivirus. ZIKV infection may lead to microcephaly in developing fetus and Guillain-Barré Syndrome (GBS) like symptoms in adults. ZIKV was first reported in humans in 1952 from Uganda and the United Republic of Tanzania. Later, ZIKV outbreak was reported in 2007 from the Yap Island. ZIKV re-emerged as major outbreak in the year 2013 from French Polynesia followed by second outbreak in the year 2015 from Brazil. ZIKV crosses the blood-tissue barriers to enter immune-privileged organs. Clinical manifestations in ZIKV disease includes rash, fever, conjunctivitis, muscle and joint pain, headache, transverse myelitis, meningoencephalitis, Acute Disseminated Encephalomyelitis (ADEM). The understanding of the molecular mechanism of ZIKV pathogenesis is very important to develop potential diagnostic and therapeutic interventions for ZIKV infected patients.
Collapse
|
22
|
Ma J, Gong S, He Y, Gao W, Hao W, Lan X. Effects of oral sialic acid on gut development, liver function and gut microbiota in mice. Lett Appl Microbiol 2021; 73:20-25. [PMID: 33386625 DOI: 10.1111/lam.13447] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/27/2020] [Accepted: 12/28/2020] [Indexed: 12/27/2022]
Abstract
Sialic acid (N-acetylneuraminic acid), a 9-carbon monosaccharide, has been widely studied in immunology, oncology and neurology. However, the effects of sialic acid on organ and intestinal development, liver function and gut microbiota were rarely studied. In this study, we found that oral sialic acid tended to increase the relative weight of liver and decreased the serum aspartate aminotransferase (GPT) activity. In addition, sialic acid treatment markedly reduced gut villus length, depth, the ratio of villus length/depth (L/D), areas, width and the number of goblet cells. Furthermore, gut microbes were changed in response to oral sialic acid, such as Staphylococcus lentus, Corynebacterium stationis, Corynebacterium urealyticum, Jeotgalibaca sp_PTS2502, Ignatzschineria indica, Sporosarcina pasteurii, Sporosarcina sp_HW10C2, Facklamia tabacinasalis, Oblitimonas alkaliphila, Erysipelatoclostridium ramosum, Blautia sp_YL58, Bacteroids thetaiotaomicron, Morganella morganii, Clostridioides difficile, Helicobacter tryphlonius, Clostridium sp_Clone47, Alistipes finegoldii, [pseudomonas]_geniculata and Pseudomonas parafulva at the species level. In conclusion, oral sialic acid altered the intestinal pathological state and microbial compositions, and the effect of sialic acid on host health should be further studied.
Collapse
Affiliation(s)
- J Ma
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - S Gong
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Y He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - W Gao
- Animal Husbandry and Aquatic Affairs Center of Shimen County, Changde, Hunan, China
| | - W Hao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - X Lan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| |
Collapse
|
23
|
Wu H, Wang F, Tang D, Han D. Mumps Orchitis: Clinical Aspects and Mechanisms. Front Immunol 2021; 12:582946. [PMID: 33815357 PMCID: PMC8013702 DOI: 10.3389/fimmu.2021.582946] [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: 08/11/2020] [Accepted: 03/04/2021] [Indexed: 12/12/2022] Open
Abstract
The causative agent of mumps is a single-stranded, non-segmented, negative sense RNA virus belonging to the Paramyxoviridae family. Besides the classic symptom of painfully swollen parotid salivary glands (parotitis) in mumps virus (MuV)-infected men, orchitis is the most common form of extra-salivary gland inflammation. Mumps orchitis frequently occurs in young adult men, and leads to pain and swelling of the testis. The administration of MuV vaccines in children has been proven highly effective in reducing the incidence of mumps. However, a recent global outbreak of mumps and the high rate of orchitis have recently been considered as threats to male fertility. The pathogenesis of mumps orchitis remains largely unclear due to lack of systematic clinical data analysis and animal models studies. The alarming increase in the incidence of mumps orchitis and the high risk of the male fertility have thus become a major health concern. Recent studies have revealed the mechanisms by which MuV-host cells interact and MuV infection induces inflammatory responses in testicular cells. In this mini-review, we highlight advances in our knowledge of the clinical aspects and possible mechanisms of mumps orchitis.
Collapse
Affiliation(s)
- Han Wu
- Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Fei Wang
- Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Dongdong Tang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Daishu Han
- Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
24
|
Sialic Acid Receptors: The Key to Solving the Enigma of Zoonotic Virus Spillover. Viruses 2021; 13:v13020262. [PMID: 33567791 PMCID: PMC7915228 DOI: 10.3390/v13020262] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 12/14/2022] Open
Abstract
Emerging viral diseases are a major threat to global health, and nearly two-thirds of emerging human infectious diseases are zoonotic. Most of the human epidemics and pandemics were caused by the spillover of viruses from wild mammals. Viruses that infect humans and a wide range of animals have historically caused devastating epidemics and pandemics. An in-depth understanding of the mechanisms of viral emergence and zoonotic spillover is still lacking. Receptors are major determinants of host susceptibility to viruses. Animal species sharing host cell receptors that support the binding of multiple viruses can play a key role in virus spillover and the emergence of novel viruses and their variants. Sialic acids (SAs), which are linked to glycoproteins and ganglioside serve as receptors for several human and animal viruses. In particular, influenza and coronaviruses, which represent two of the most important zoonotic threats, use SAs as cellular entry receptors. This is a comprehensive review of our current knowledge of SA receptor distribution among animal species and the range of viruses that use SAs as receptors. SA receptor tropism and the predicted natural susceptibility to viruses can inform targeted surveillance of domestic and wild animals to prevent the future emergence of zoonotic viruses.
Collapse
|
25
|
Azeem MS, Yesupatham ST, Mohiyuddin SMA, Sumanth V, Ravishankar S. Usefulness of salivary sialic acid as a tumor marker in tobacco chewers with oral cancer. J Cancer Res Ther 2020; 16:605-611. [PMID: 32719275 DOI: 10.4103/jcrt.jcrt_337_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Aim This study aims to assess the usefulness of salivary sialic acid (SA) as a tumor marker in the detection of oral squamous cell carcinoma (OSCC) among tobacco chewers. Materials and Methods After the approval of study protocol by the Institutional Ethics Committee and informed voluntary consent, salivary samples were collected from 96 participants in each group of tobacco chewers with OSCC, tobacco chewers without precancerous or cancerous lesion, and healthy controls. Salivary protein-bound SA (PBSA) and salivary-free SA (FSA) were measured by Yao et al.'s method of acid ninhydrin reaction, and the data were subjected to appropriate statistical analysis. Results The salivary PBSA and FSA levels in the Groups 1, 2, and 3 participants were 31.17 ± 7.6 mg/dL and 63.45 ± 9.8 mg/dL, 25.45 ± 16.61 mg/dL and 33.18 ± 11.38 mg/dL, and 22.73 ± 3.01 mg/dL and 21.62 ± 8.86 mg/dL, respectively. Salivary FSA levels were significantly increased among the tobacco chewers with OSCC patients (Group 1) and tobacco chewers with no premalignant lesions of the oral cavity (Group 2) compared to the healthy controls (Group 3) with P < 0.05 being statistically significant. Salivary FSA levels were significantly increased in Group 1 as compared with Group 2. The salivary PBSA was high among Group 1 as compared to the control Group 3; there was however no significant difference in the levels of salivary PBSA between Group 1 and Group 2. There was no significant difference in the PBSA levels between OSCC patients of Group 1 and the tobacco chewers without precancerous or cancerous lesion in the oral cavity of Group 2. Conclusion Salivary PBSA and FSA are significantly raised in both tobacco chewers with OSCC and in tobacco chewers with no precancerous or cancerous lesions in the oral cavity. SA should therefore be used cautiously while considering it as a marker for the early detection of oral cancer. Tobacco can be a crucial confounding factor when SA is used as a biomarker in OSCC since their levels are elevated to some extent even in tobacco chewers without any clinically obvious precancerous or cancerous lesions in the oral cavity.
Collapse
Affiliation(s)
- Mahnaaz Sultana Azeem
- Department of Otorhinolaryngology and Head and Neck Surgery, Sri Devaraj Urs Academy of Higher Education and Research, Kolar, Karnataka, India
| | | | - S M Azeem Mohiyuddin
- Department of Otorhinolaryngology and Head and Neck Surgery, Sri Devaraj Urs Academy of Higher Education and Research, Kolar, Karnataka, India
| | - V Sumanth
- Department of Otorhinolaryngology and Head and Neck Surgery, Sri Devaraj Urs Academy of Higher Education and Research, Kolar, Karnataka, India
| | - S Ravishankar
- Department of Community Medicine, Sri Devaraj Urs Academy of Higher Education and Research, Kolar, Karnataka, India
| |
Collapse
|
26
|
Heida R, Bhide YC, Gasbarri M, Kocabiyik Ö, Stellacci F, Huckriede ALW, Hinrichs WLJ, Frijlink HW. Advances in the development of entry inhibitors for sialic-acid-targeting viruses. Drug Discov Today 2020; 26:122-137. [PMID: 33099021 PMCID: PMC7577316 DOI: 10.1016/j.drudis.2020.10.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/13/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022]
Abstract
Over the past decades, several antiviral drugs have been developed to treat a range of infections. Yet the number of treatable viral infections is still limited, and resistance to current drug regimens is an ever-growing problem. Therefore, additional strategies are needed to provide a rapid cure for infected individuals. An interesting target for antiviral drugs is the process of viral attachment and entry into the cell. Although most viruses use distinct host receptors for attachment to the target cell, some viruses share receptors, of which sialic acids are a common example. This review aims to give an update on entry inhibitors for a range of sialic-acid-targeting viruses and provides insight into the prospects for those with broad-spectrum potential.
Collapse
Affiliation(s)
- Rick Heida
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9713AV Groningen, The Netherlands
| | - Yoshita C Bhide
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9713AV Groningen, The Netherlands; Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, 9713AV Groningen, The Netherlands
| | - Matteo Gasbarri
- Institute of Materials, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Özgün Kocabiyik
- Institute of Materials, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Francesco Stellacci
- Institute of Materials, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Anke L W Huckriede
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, 9713AV Groningen, The Netherlands
| | - Wouter L J Hinrichs
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9713AV Groningen, The Netherlands.
| | - Henderik W Frijlink
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9713AV Groningen, The Netherlands
| |
Collapse
|
27
|
Lin Q, Zhou S, Huang Y, Huo Z, Chen C, Luo X, He J, Liu C, Zhang P. ANKS4B Restricts Replication of Zika Virus by Downregulating the Autophagy. Front Microbiol 2020; 11:1745. [PMID: 32793175 PMCID: PMC7387654 DOI: 10.3389/fmicb.2020.01745] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/03/2020] [Indexed: 12/21/2022] Open
Abstract
Infection of Zika virus (ZIKV) has become a severe threaten to global health while no specific drug is available. In this study, we explored the relationship between ZIKV and a cellular protein, ankyrin repeat and sterile motif domain containing 4b (ANKS4B). Our data revealed that the expression of ANKS4B in cultured cells and in neonatal mice was downregulated by ZIKV infection. The reduction of ANKS4B upon ZIKV infection was caused by decrease of two hepatocyte nuclear factors HNF1α and HNF4α. Through CRISPR/Cas9 gene editing system, we generated two ANKS4B knockout (KO) cell clones in A549 and Huh7 cells respectively. In the ANKS4B-KO cells, the viral replication levels including viral RNA, protein, and titer were significantly enhanced, which was reversed by trans-complementation of ANKS4B. ANKS4B did not affect the viral entry step, but impaired the autophagy induced by ZIKV infection. Furthermore, our data showed that inhibition of autophagy led to similar replication levels of ZIKV in ANKS4B-sufficient and ANKS4B-deficient cells, suggesting the antiviral effect of ANKS4B relied on its modulation on the autophagy. Therefore, our work identified ANKS4B as a new restriction factor of ZIKV.
Collapse
Affiliation(s)
- Quanshi Lin
- Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China.,Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shili Zhou
- Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China.,Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yanxia Huang
- Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China.,Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhiting Huo
- Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China.,Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Cancan Chen
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xin Luo
- Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China.,Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Junfang He
- Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China.,Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chao Liu
- Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China.,Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ping Zhang
- Key Laboratory of Tropical Disease Control, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China.,Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
28
|
Wang F, Chen R, Jiang Q, Wu H, Gong M, Liu W, Yu X, Zhang W, Han R, Liu A, Chen Y, Han D. Roles of Sialic Acid, AXL, and MER Receptor Tyrosine Kinases in Mumps Virus Infection of Mouse Sertoli and Leydig Cells. Front Microbiol 2020; 11:1292. [PMID: 32695074 PMCID: PMC7336603 DOI: 10.3389/fmicb.2020.01292] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/20/2020] [Indexed: 01/08/2023] Open
Abstract
The mumps virus (MuV) causes epidemic parotitis. MuV also frequently infects the testis and induces orchitis, an important etiological factor contributing to male infertility. However, mechanisms underlying MuV infection of the testis remain unknown. Here, we describe that sialic acid, AXL, and MER receptor tyrosine kinases regulate MuV entry and replication in mouse major testicular cells, including Sertoli and Leydig cells. Sialic acid, AXL, and MER were present in Sertoli and Leydig cells. Sialic acid specifically mediated MuV entry into Sertoli and Leydig cells, whereas both AXL and MER facilitated MuV replication within cells through the inhibition of cellular innate antiviral responses. Mechanistically, the inhibition of type 1 interferon signaling by AXL and MER is essential for MuV replication in Sertoli and Leydig cells. Our findings provide novel insights into the mechanisms behind MuV infection and replication in the testis.
Collapse
Affiliation(s)
- Fei Wang
- Peking Union Medical College, School of Basic Medicine, Chinese Academy of Medical Sciences, Institute of Basic Medical Sciences, Beijing, China
| | - Ran Chen
- Peking Union Medical College, School of Basic Medicine, Chinese Academy of Medical Sciences, Institute of Basic Medical Sciences, Beijing, China
| | - Qian Jiang
- Peking Union Medical College, School of Basic Medicine, Chinese Academy of Medical Sciences, Institute of Basic Medical Sciences, Beijing, China
| | - Han Wu
- Peking Union Medical College, School of Basic Medicine, Chinese Academy of Medical Sciences, Institute of Basic Medical Sciences, Beijing, China
| | - Maolei Gong
- Peking Union Medical College, School of Basic Medicine, Chinese Academy of Medical Sciences, Institute of Basic Medical Sciences, Beijing, China
| | - Weihua Liu
- Peking Union Medical College, School of Basic Medicine, Chinese Academy of Medical Sciences, Institute of Basic Medical Sciences, Beijing, China
| | - Xiaoqin Yu
- Peking Union Medical College, School of Basic Medicine, Chinese Academy of Medical Sciences, Institute of Basic Medical Sciences, Beijing, China
| | - Wenjing Zhang
- Peking Union Medical College, School of Basic Medicine, Chinese Academy of Medical Sciences, Institute of Basic Medical Sciences, Beijing, China
| | - Ruiqin Han
- Peking Union Medical College, School of Basic Medicine, Chinese Academy of Medical Sciences, Institute of Basic Medical Sciences, Beijing, China
| | - Aijie Liu
- Peking Union Medical College, School of Basic Medicine, Chinese Academy of Medical Sciences, Institute of Basic Medical Sciences, Beijing, China
| | - Yongmei Chen
- Peking Union Medical College, School of Basic Medicine, Chinese Academy of Medical Sciences, Institute of Basic Medical Sciences, Beijing, China
| | - Daishu Han
- Peking Union Medical College, School of Basic Medicine, Chinese Academy of Medical Sciences, Institute of Basic Medical Sciences, Beijing, China
| |
Collapse
|
29
|
Roth H, Schneider L, Eberle R, Lausen J, Modlich U, Blümel J, Baylis SA. Zika virus infection studies with CD34 + hematopoietic and megakaryocyte-erythroid progenitors, red blood cells and platelets. Transfusion 2020; 60:561-574. [PMID: 32086956 DOI: 10.1111/trf.15692] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/28/2019] [Accepted: 01/13/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND To date, several cases of transfusion-transmitted ZIKV infections have been confirmed. Multiple studies detected prolonged occurrence of ZIKV viral RNA in whole blood as compared to plasma samples indicating potential ZIKV interaction with hematopoietic cells. Also, infection of cells from the granulocyte/macrophage lineage has been demonstrated. Patients may develop severe thrombocytopenia, microcytic anemia, and a fatal course of disease occurred in a patient with sickle cell anemia suggesting additional interference of ZIKV with erythroid and megakaryocytic cells. Therefore, we analyzed whether ZIKV propagates in or compartmentalizes with hematopoietic progenitor, erythroid, and megakaryocytic cells. METHODS ZIKV RNA replication, protein translation and infectious particle formation in hematopoietic cell lines as well as primary CD34+ HSPCs and ex vivo differentiated erythroid and megakaryocytic cells was monitored using qRT-PCR, FACS, immunofluorescence analysis and infectivity assays. Distribution of ZIKV RNA and infectious particles in spiked red blood cell (RBC) units or platelet concentrates (PCs) was evaluated. RESULTS While subsets of K562 and KU812Ep6EPO cells supported ZIKV propagation, primary CD34+ HSPCs, MEP cells, RBCs, and platelets were non-permissive for ZIKV infection. In spiking studies, ZIKV RNA was detectable for 7 days in all fractions of RBC units and PCs, however, ZIKV infectious particles were not associated with erythrocytes or platelets. CONCLUSION Viral particles from plasma or contaminating leukocytes, rather than purified CD34+ HSPCs or the cellular component of RBC units or PCs, present the greatest risk for transfusion-transmitted ZIKV infections.
Collapse
Affiliation(s)
- Hanna Roth
- Division of Virology, Paul-Ehrlich-Institute, Langen, Hessen, Germany
| | - Lucas Schneider
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Hessen, Germany
| | - Regina Eberle
- Division of Immunology, Paul-Ehrlich-Institute, Langen, Hessen, Germany
| | - Jörn Lausen
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Hessen, Germany.,Department of Genetics of Eukaryotes, Institute of Industrial Genetics, Stuttgart, Baden-Württemberg, Germany
| | - Ute Modlich
- Division of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Hessen, Germany
| | - Johannes Blümel
- Division of Virology, Paul-Ehrlich-Institute, Langen, Hessen, Germany
| | - Sally A Baylis
- Division of Virology, Paul-Ehrlich-Institute, Langen, Hessen, Germany
| |
Collapse
|