1
|
Chen Q, Chen Y, Bao C, Xiang H, Gao Q, Mao L. Mechanism and complex roles of HSC70/HSPA8 in viral entry. Virus Res 2024; 347:199433. [PMID: 38992806 PMCID: PMC11305274 DOI: 10.1016/j.virusres.2024.199433] [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: 06/14/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/13/2024]
Abstract
The process of viruses entering host cells is complex, involving multiple aspects of the molecular organization of the cell membrane, viral proteins, the interaction of receptor molecules, and cellular signaling. Most viruses depend on endocytosis for uptake, when viruses reach the appropriate location, they are released from the vesicles, undergo uncoating, and release their genomes. Heat shock cognate protein 70(HSC70): also known as HSPA8, a protein involved in mediating clathrin-mediated endocytosis (CME), is involved in various viral entry processes. In this mini-review, our goal is to provide a summary of the function of HSC70 in viral entry. Understanding the interaction networks of HSC70 with viral proteins helps to provide new directions for targeted therapeutic strategies against viral infections.
Collapse
Affiliation(s)
- Qiaoqiao Chen
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, PR China
| | - Yiwen Chen
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, PR China
| | - Chenxuan Bao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University,Kunshan, Jiangsu, PR China
| | - Huayuan Xiang
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University,Kunshan, Jiangsu, PR China
| | - Qing Gao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University,Kunshan, Jiangsu, PR China
| | - Lingxiang Mao
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, PR China; Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University,Kunshan, Jiangsu, PR China.
| |
Collapse
|
2
|
Temple H, Donnelly B, Mohanty SK, Mowery S, Poling HM, Pasula R, Hartman S, Singh A, Mourya R, Bondoc A, Meller J, Jegga AG, Oyama K, McNeal M, Spearman P, Tiao G. Specific binding sites on Rhesus rotavirus capsid protein dictate the method of endocytosis inducing the murine model of biliary atresia. Am J Physiol Gastrointest Liver Physiol 2024; 327:G267-G283. [PMID: 38860860 DOI: 10.1152/ajpgi.00308.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/17/2024] [Accepted: 05/28/2024] [Indexed: 06/12/2024]
Abstract
Biliary atresia (BA) is the leading indication for pediatric liver transplantation. Rhesus rotavirus (RRV)-induced murine BA develops an obstructive cholangiopathy that mirrors the human disease. We have previously demonstrated the "SRL" motif on RRV's VP4 protein binds to heat shock cognate 70 protein (Hsc70) facilitating entry into cholangiocytes. In this study, we analyzed how binding to Hsc70 affects viral endocytosis, intracellular trafficking, and uniquely activates the signaling pathway that induces murine BA. Inhibition of clathrin- and dynamin-mediated endocytosis in cholangiocytes following infection demonstrated that blocking dynamin decreased the infectivity of RRV, whereas clathrin inhibition had no effect. Blocking early endosome trafficking resulted in decreased viral titers of RRV, whereas late endosome inhibition had no effect. After infection, TLR3 expression and p-NF-κB levels increased in cholangiocytes, leading to increased release of CXCL9 and CXCL10. Infected mice knocked out for TLR3 had decreased levels of CXCL9 and CXCL10, resulting in reduced NK cell numbers. Human patients with BA experienced an increase in CXCL10 levels, suggesting this as a possible pathway leading to biliary obstruction. Viruses that use Hsc70 for cell entry exploit a clathrin-independent pathway and traffic to the early recycling endosome uniquely activating NF-κB through TLR3, leading to the release of CXCL9 and CXCL10 and inducing NK cell recruitment. These results define how the "SRL" peptide found on RRV's VP4 protein modulates viral trafficking, inducing the host response leading to bile duct obstruction.NEW & NOTEWORTHY In this study, we have determined that the presence of the "SRL" peptide on RRV alters its method of endocytosis and intracellular trafficking through viral binding to heat shock cognate 70 protein. This initiates an inflammatory pathway that stimulates the release of cytokines associated with biliary damage and obstruction.
Collapse
Affiliation(s)
- Haley Temple
- Department of Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Bryan Donnelly
- Department of Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Sujit K Mohanty
- Southeast Poultry Research Laboratory, United States National Poultry Research Center, United States Department of Agriculture, Athens, Georgia, United States
| | - Sarah Mowery
- Department of Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Holly M Poling
- Department of Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Rajamouli Pasula
- Department of Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Stephen Hartman
- Department of Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Akaljot Singh
- Department of Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Reena Mourya
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Alexander Bondoc
- Department of Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Jaroslaw Meller
- Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, Ohio, United States
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Anil G Jegga
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Kei Oyama
- Department of Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Monica McNeal
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Paul Spearman
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Greg Tiao
- Department of Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| |
Collapse
|
3
|
Dey S, Pratibha M, Singh Dagur H, Rajakumara E. Characterization of host receptor interaction with envelop protein of Kyasanur forest disease virus and predicting suitable epitopes for vaccine candidate. J Biomol Struct Dyn 2024; 42:4110-4120. [PMID: 37272880 DOI: 10.1080/07391102.2023.2218924] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/22/2023] [Indexed: 06/06/2023]
Abstract
Kyasanur forest disease (KFD) is a zoonotic disease that is endemic to southern India and caused by KFD virus (KFDV) belonging to the family Flaviviridae. Humans are the dead-end host of the KFDV life cycle. The absence of effective treatment strategies against KFD can be attributed to a lack of studies on the mechanistic part of the spread of the disease. Hypothesizing molecular etiological similarity of KFDV to other well characterized flaviviruses, such as dengue virus (DENV), we focused on predicting the target receptor protein(s) in host and provided molecular basis of receptor-mediated recognition of the human host by KFDV envelop protein (EKFDV), drawing from the extant knowledge on the dengue counterpart, EDENV. Indeed, in silico approach helped to identify that the EKFDV structure closely resembles the EDENV structure and indicated DC-SIGN and/or Mannose receptors to be the plausible target host receptors. Immune-informatics approach aided in predicting 10 epitopes from E, NS1, NS2A, and NS2B proteins of the KFDV-P9605 genotype for vaccine design against KFDV. Further, molecular dynamics simulation (MDS) analyses of their complexes with human leukocyte antigens (HLAs) identified the epitopes DISLTCRVT and YAMEIRPVH as two high ranking candidates for vaccine design.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sreenath Dey
- Macromolecular Structural Biology Lab, Department of Biotechnology, Indian Institute of Technology, Hyderabad, Kandi, India
| | - Manickavasagam Pratibha
- Macromolecular Structural Biology Lab, Department of Biotechnology, Indian Institute of Technology, Hyderabad, Kandi, India
| | - Hanuman Singh Dagur
- Macromolecular Structural Biology Lab, Department of Biotechnology, Indian Institute of Technology, Hyderabad, Kandi, India
| | - Eerappa Rajakumara
- Macromolecular Structural Biology Lab, Department of Biotechnology, Indian Institute of Technology, Hyderabad, Kandi, India
| |
Collapse
|
4
|
Pei Y, Lin C, Li H, Feng Z. Genetic background influences pig responses to porcine reproductive and respiratory syndrome virus. Front Vet Sci 2023; 10:1289570. [PMID: 37929286 PMCID: PMC10623566 DOI: 10.3389/fvets.2023.1289570] [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: 09/06/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a highly infectious and economically significant virus that causes respiratory and reproductive diseases in pigs. It results in reduced productivity and increased mortality in pigs, causing substantial economic losses in the industry. Understanding the factors affecting pig responses to PRRSV is crucial to develop effective control strategies. Genetic background has emerged as a significant determinant of susceptibility and resistance to PRRSV in pigs. This review provides an overview of the basic infection process of PRRSV in pigs, associated symptoms, underlying immune mechanisms, and roles of noncoding RNA and alternative splicing in PRRSV infection. Moreover, it emphasized breed-specific variations in these aspects that may have implications for individual treatment options.
Collapse
Affiliation(s)
- Yangli Pei
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Key Laboratory of Animal Molecular Design and Precise Breeding of Guangdong Higher Education Institutes, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Chenghong Lin
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Key Laboratory of Animal Molecular Design and Precise Breeding of Guangdong Higher Education Institutes, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Hua Li
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Key Laboratory of Animal Molecular Design and Precise Breeding of Guangdong Higher Education Institutes, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Zheng Feng
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Key Laboratory of Animal Molecular Design and Precise Breeding of Guangdong Higher Education Institutes, School of Life Science and Engineering, Foshan University, Foshan, China
| |
Collapse
|
5
|
Liu Z, Zhang Q, Li L, He J, Guo J, Wang Z, Huang Y, Xi Z, Yuan F, Li Y, Li T. The effect of temperature on dengue virus transmission by Aedes mosquitoes. Front Cell Infect Microbiol 2023; 13:1242173. [PMID: 37808907 PMCID: PMC10552155 DOI: 10.3389/fcimb.2023.1242173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023] Open
Abstract
Dengue is prevalent in tropical and subtropical regions. As an arbovirus disease, it is mainly transmitted by Aedes aegypti and Aedes albopictus. According to the previous studies, temperature is closely related to the survival of Aedes mosquitoes, the proliferation of dengue virus (DENV) and the vector competence of Aedes to transmit DENV. This review describes the correlations between temperature and dengue epidemics, and explores the potential reasons including the distribution and development of Aedes mosquitoes, the structure of DENV, and the vector competence of Aedes mosquitoes. In addition, the immune and metabolic mechanism are discussed on how temperature affects the vector competence of Aedes mosquitoes to transmit DENV.
Collapse
Affiliation(s)
- Zhuanzhuan Liu
- Department of Pathogen Biology, Center for Tropical Disease Control and Research, School of Basic Medical Sciences and Life Sciences, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China
- Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
| | - Qingxin Zhang
- School of Imaging Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Liya Li
- School of Imaging Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Junjie He
- School of Imaging Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Jinyang Guo
- School of Imaging Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Zichen Wang
- School of Imaging Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yige Huang
- School of Imaging Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Zimeng Xi
- School of Imaging Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Fei Yuan
- Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
| | - Yiji Li
- Department of Pathogen Biology, Center for Tropical Disease Control and Research, School of Basic Medical Sciences and Life Sciences, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China
| | - Tingting Li
- Department of Pathogen Biology, Center for Tropical Disease Control and Research, School of Basic Medical Sciences and Life Sciences, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China
| |
Collapse
|
6
|
González-Flores AM, Salas-Benito M, Rosales-García VH, Zárate-Segura PB, Del Ángel RM, De Nova-Ocampo MA, Salas-Benito JS. Characterization of Viral Interference in Aedes albopictus C6/36 Cells Persistently Infected with Dengue Virus 2. Pathogens 2023; 12:1135. [PMID: 37764943 PMCID: PMC10536104 DOI: 10.3390/pathogens12091135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Arboviruses are an important group of pathogens that cause diseases of medical and veterinary concern worldwide. The interactions of these viruses with their host cells are complex, and frequently, the coexistence of two different viruses in the same cell results in the inhibition of replication in one of the viruses, which is a phenomenon called viral interference. This phenomenon can be exploited to develop antiviral strategies. Insect cell lines persistently infected with arboviruses are useful models with which to study viral interference. In this work, a model of C6/36-HT cells (from Aedes albopictus mosquitoes) persistently infected with Dengue virus, serotype 2, was used. Viral interference was evaluated via plaque and flow cytometry assays. The presence of heterotypic interference against the other serotypes of the same virus and homologous interference against yellow fever virus was determined; however, this cell line did not display heterologous viral interference against Sindbis virus. The mechanisms responsible for viral interference have not been fully elucidated, but small RNAs could be involved. However, the silencing of Ago3, a key protein in the genome-derived P-element-induced wimpy testis pathway, did not alter the viral interference process, suggesting that viral interference occurs independent of this pathway.
Collapse
Affiliation(s)
| | - Mariana Salas-Benito
- Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City 07320, Mexico; (M.S.-B.); (M.A.D.N.-O.)
| | - Victor Hugo Rosales-García
- Laboratorios Centrales, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City 07360, Mexico;
| | | | - Rosa María Del Ángel
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City 07360, Mexico;
| | - Mónica Ascención De Nova-Ocampo
- Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City 07320, Mexico; (M.S.-B.); (M.A.D.N.-O.)
| | - Juan Santiago Salas-Benito
- Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City 07320, Mexico; (M.S.-B.); (M.A.D.N.-O.)
| |
Collapse
|
7
|
Chen N, Bai T, Wang S, Wang H, Wu Y, Liu Y, Zhu Z. New Insights into the Role and Therapeutic Potential of Heat Shock Protein 70 in Bovine Viral Diarrhea Virus Infection. Microorganisms 2023; 11:1473. [PMID: 37374975 DOI: 10.3390/microorganisms11061473] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Bovine viral diarrhea virus (BVDV), a positive-strand RNA virus of the genus Pestivirus in the Flaviviridae family, is the causative agent of bovine viral diarrhea-mucosal disease (BVD-MD). BVDV's unique virion structure, genome, and replication mechanism in the Flaviviridae family render it a useful alternative model for evaluating the effectiveness of antiviral drugs used against the hepatitis C virus (HCV). As one of the most abundant and typical heat shock proteins, HSP70 plays an important role in viral infection caused by the family Flaviviridae and is considered a logical target of viral regulation in the context of immune escape. However, the mechanism of HSP70 in BVDV infection and the latest insights have not been reported in sufficient detail. In this review, we focus on the role and mechanisms of HSP70 in BVDV-infected animals/cells to further explore the possibility of targeting this protein for antiviral therapy during viral infection.
Collapse
Affiliation(s)
- Nannan Chen
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
- Branch of Animal Husbandry and Veterinary of Heilongjiang Academy of Agricultural Sciences, Qiqihar 161006, China
| | - Tongtong Bai
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Shuang Wang
- Branch of Animal Husbandry and Veterinary of Heilongjiang Academy of Agricultural Sciences, Qiqihar 161006, China
| | - Huan Wang
- Branch of Animal Husbandry and Veterinary of Heilongjiang Academy of Agricultural Sciences, Qiqihar 161006, China
| | - Yue Wu
- Branch of Animal Husbandry and Veterinary of Heilongjiang Academy of Agricultural Sciences, Qiqihar 161006, China
| | - Yu Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Engineering Research Center for Prevention and Control of Cattle Diseases, Daqing 163319, China
| | - Zhanbo Zhu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing 163319, China
- Engineering Research Center for Prevention and Control of Cattle Diseases, Daqing 163319, China
| |
Collapse
|
8
|
Ye N, Wang B, Feng W, Tang D, Zeng Z. PRRS virus receptors and an alternative pathway for viral invasion. Virus Res 2022; 320:198885. [PMID: 35948131 DOI: 10.1016/j.virusres.2022.198885] [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: 06/05/2022] [Revised: 08/05/2022] [Accepted: 08/06/2022] [Indexed: 11/25/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) has a highly restricted cell tropism, which is closely related to the specific receptors associated with PRRSV infection. At least nine cellular molecules have been identified as putative receptors for PRRSV, including CD163, a cysteine-rich scavenger receptor. With the participation of the CD163 receptor and other cofactors, PRRSV invades cells via low pH-dependent clathrin-mediated endocytosis. In addition, PRRSV utilizes viral apoptotic mimicry to infect cells though macropinocytosis as an alternative pathway. In this review, we discuss recent advances in the studies on receptors and pathways that play an important role in PRRSV invasion, and simultaneously explore the use of specific antibodies, small molecules, and blockers targeting receptor-ligand interactions, as a potential strategy for controlling PRRSV infection. Novel antiviral strategies against PRRSV could be developed by identifying the interaction between receptors and ligands.
Collapse
Affiliation(s)
- Ni Ye
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Bin Wang
- College of Animal Science, Guizhou University, Guiyang 550025, China.
| | - Wei Feng
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Deyuan Tang
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Zhiyong Zeng
- College of Animal Science, Guizhou University, Guiyang 550025, China
| |
Collapse
|
9
|
Mohanty SK, Donnelly B, Temple H, Mowery S, Poling H, Meller J, Malik A, McNeal M, Tiao G. Rhesus rotavirus receptor-binding site affects high mobility group box 1 release, altering the pathogenesis of experimental biliary atresia. Hepatol Commun 2022; 6:2702-2714. [PMID: 35866580 PMCID: PMC9512450 DOI: 10.1002/hep4.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/26/2022] [Accepted: 05/30/2022] [Indexed: 11/10/2022] Open
Abstract
Biliary atresia (BA) is a neonatal inflammatory cholangiopathy that requires surgical intervention by Kasai portoenterostomy to restore biliary drainage. Even with successful portoenterostomy, most patients diagnosed with BA progress to end-stage liver disease, necessitating a liver transplantation for survival. In the murine model of BA, rhesus rotavirus (RRV) infection of neonatal mice induces an inflammatory obstructive cholangiopathy that parallels human BA. The model is triggered by RRV viral protein (VP)4 binding to cholangiocyte cell-surface proteins. High mobility group box 1 (HMGB1) protein is a danger-associated molecular pattern that when released extracellularly moderates innate and adaptive immune response. In this study, we investigated how mutations in three RRV VP4-binding sites, RRVVP4-K187R (sialic acid-binding site), RRVVP4-D308A (integrin α2β1-binding site), and RRVVP4-R446G (heat shock cognate 70 [Hsc70]-binding site), affects infection, HMGB1 release, and the murine model of BA. Newborn pups injected with RRVVP4-K187R and RRVVP4-D308A developed an obstruction within the extrahepatic bile duct similar to wild-type RRV, while those infected with RRVVP4-R446G remained patent. Infection with RRVVP4-R446G induced a lower level of HMGB1 release from cholangiocytes and in the serum of infected pups. RRV infection of HeLa cells lacking Hsc70 resulted in no HMGB1 release, while transfection with wild-type Hsc70 into HeLa Hsc70-deficient cells reestablished HMGB1 release, indicating a mechanistic role for Hsc70 in its release. Conclusion: Binding to Hsc70 contributes to HMGB1 release; therefore, Hsc70 potentially serves as a therapeutic target for BA.
Collapse
Affiliation(s)
- Sujit K. Mohanty
- Department of Pediatric and Thoracic SurgeryCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Bryan Donnelly
- Department of Pediatric and Thoracic SurgeryCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Haley Temple
- Department of Pediatric and Thoracic SurgeryCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Sarah Mowery
- Department of Pediatric and Thoracic SurgeryCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Holly M. Poling
- Department of Pediatric and Thoracic SurgeryCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Jaroslaw Meller
- Department of Environmental and Public Health SciencesUniversity of CincinnatiCincinnatiOhioUSA
- Division of Biomedical InformaticsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Astha Malik
- Division of Gastroenterology, Hepatology, and NutritionCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Monica McNeal
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Division of Infectious DiseasesCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Greg Tiao
- Department of Pediatric and Thoracic SurgeryCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| |
Collapse
|
10
|
Renman E, Ekici R, Sundström M, Lejon K. HSC70 is a novel binding partner involved in the capture of immunoglobulins on B cells in the NOD mouse. Autoimmunity 2022; 55:520-528. [PMID: 36120986 DOI: 10.1080/08916934.2022.2117307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
B cells have been shown to be essential for Type 1 diabetes development in the non-obese diabetic mouse, where their contribution as antigen presenting cells has been emphasised. Other important functions for B cells include surface capture of immunoglobulins and transportation of immune complexes, with subsequent endocytosis, antigen processing and antigen presentation. We have previously demonstrated that NOD B cells capture IgM and IgG immune complexes through an unknown surface molecule. In this study, we revealed the presumptive immunoglobulin-binding molecule to be HSC70. Moreover, we detected increased levels of HSC70 on NOD B cells. HSC70 has been shown to play a role in antigen processing and presentation as well as being important in several autoimmune diseases, including rheumatoid arthritis and systemic lupus erythematosus. Due to its protein stabilising properties, increased HSC70 could contribute to enhanced self-antigen collection and presentation and thereby contribute to the development of Type 1 diabetes.
Collapse
Affiliation(s)
- Emma Renman
- Department of Clinical Microbiology Umeå, Umeå University, Umeå, Sweden
| | - Rifat Ekici
- Department of Clinical Microbiology Umeå, Umeå University, Umeå, Sweden
| | - Mia Sundström
- Department of Clinical Microbiology Umeå, Umeå University, Umeå, Sweden
| | - Kristina Lejon
- Department of Clinical Microbiology Umeå, Umeå University, Umeå, Sweden
| |
Collapse
|
11
|
Caillet C, Stofberg ML, Muleya V, Shonhai A, Zininga T. Host cell stress response as a predictor of COVID-19 infectivity and disease progression. Front Mol Biosci 2022; 9:938099. [PMID: 36032680 PMCID: PMC9411049 DOI: 10.3389/fmolb.2022.938099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
The coronavirus disease (COVID-19) caused by a coronavirus identified in December 2019 has caused a global pandemic. COVID-19 was declared a pandemic in March 2020 and has led to more than 6.3 million deaths. The pandemic has disrupted world travel, economies, and lifestyles worldwide. Although vaccination has been an effective tool to reduce the severity and spread of the disease there is a need for more concerted approaches to fighting the disease. COVID-19 is characterised as a severe acute respiratory syndrome . The severity of the disease is associated with a battery of comorbidities such as cardiovascular diseases, cancer, chronic lung disease, and renal disease. These underlying diseases are associated with general cellular stress. Thus, COVID-19 exacerbates outcomes of the underlying conditions. Consequently, coronavirus infection and the various underlying conditions converge to present a combined strain on the cellular response. While the host response to the stress is primarily intended to be of benefit, the outcomes are occasionally unpredictable because the cellular stress response is a function of complex factors. This review discusses the role of the host stress response as a convergent point for COVID-19 and several non-communicable diseases. We further discuss the merits of targeting the host stress response to manage the clinical outcomes of COVID-19.
Collapse
Affiliation(s)
- Celine Caillet
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | | | - Victor Muleya
- Department of Biochemistry, Midlands State University, Gweru, Zimbabwe
| | - Addmore Shonhai
- Department of Biochemistry and Microbiology, University of Venda, Thohoyandou, South Africa
| | - Tawanda Zininga
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
12
|
Abstract
Heat shock proteins (HSPs) are a kind of proteins which mostly found in bacterial, plant and animal cells, in which they are involved in the monitoring and regulation of cellular life activities. HSPs protect other proteins under environmental and cellular stress by regulating protein folding and supporting the correctly folded structure of proteins as chaperones. During viral infection, some HSPs can have an antiviral effect by inhibiting viral proliferation through interaction and activating immune pathways to protect the host cell. However, although the biological function of HSPs is to maintain the homeostasis of cells, some HSPs will also be hijacked by viruses to help their invasion, replication, and maturation, thereby increasing the chances of viral survival in unfavorable conditions inside the host cell. In this review, we summarize the roles of the heat shock protein family in various stages of viral infection and the potential uses of these proteins in antiviral therapy.
Collapse
Affiliation(s)
- Xizhen Zhang
- Institute of Biochemistry, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou, China
| | - Wei Yu
- Institute of Biochemistry, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou, China
- *Correspondence: Wei Yu,
| |
Collapse
|
13
|
Liu Z, Xu Y, Li Y, Xu S, Li Y, Xiao L, Chen X, He C, Zheng K. Transcriptome analysis of Aedes albopictus midguts infected by dengue virus identifies a gene network module highly associated with temperature. Parasit Vectors 2022; 15:173. [PMID: 35590344 PMCID: PMC9118615 DOI: 10.1186/s13071-022-05282-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Dengue is prevalent worldwide and is transmitted by Aedes mosquitoes. Temperature is a strong driver of dengue transmission. However, little is known about the underlying mechanisms. METHODS Aedes albopictus mosquitoes exposed or not exposed to dengue virus serotype 2 (DENV-2) were reared at 23 °C, 28 °C and 32 °C, and midguts and residual tissues were evaluated at 7 days after infection. RNA sequencing of midgut pools from the control group, midgut breakthrough group and midgut nonbreakthrough group at different temperatures was performed. The transcriptomic profiles were analyzed using the R package, followed by weighted gene correlation network analysis (WGCNA) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis to identify the important molecular mechanisms regulated by temperature. RESULTS The midgut infection rate and midgut breakthrough rate at 28 °C and 32 °C were significantly higher than those at 23 °C, which indicates that high temperature facilitates DENV-2 breakthrough in the Ae. albopictus midgut. Transcriptome sequencing was performed to investigate the antiviral mechanism in the midgut. The midgut gene expression datasets clustered with respect to temperature, blood-feeding and midgut breakthrough. Over 1500 differentially expressed genes were identified by pairwise comparisons of midguts at different temperatures. To assess key molecules regulated by temperature, we used WGCNA, which identified 28 modules of coexpressed genes; the ME3 module correlated with temperature. KEGG analysis indicated that RNA degradation, Toll and immunodeficiency factor signaling and other pathways are regulated by temperature. CONCLUSIONS Temperature affects the infection and breakthrough of Ae. albopictus midguts invaded by DENV-2, and Ae. albopictus midgut transcriptomes change with temperature. The candidate genes and key pathways regulated by temperature provide targets for the prevention and control of dengue.
Collapse
Affiliation(s)
- Zhuanzhuan Liu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
| | - Ye Xu
- Department of Pathogen Biology, Key Laboratory of Tropical Disease Research of Guangdong Province, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yudi Li
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
| | - Shihong Xu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
| | - Yiji Li
- Department of Pathogen Biology, Hainan Medical University, Haikou, Hainan, China
| | - Ling Xiao
- Taiyuan Central Hospital, Shanxi, China
| | - Xiaoguang Chen
- Department of Pathogen Biology, Key Laboratory of Tropical Disease Research of Guangdong Province, School of Public Health, Southern Medical University, Guangzhou, China
| | - Cheng He
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China.
| | - Kuiyang Zheng
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
14
|
Cellular protein HSC70 promotes fowl adenovirus serotype 4 replication in LMH cells via interacting with viral 100K protein. Poult Sci 2022; 101:101941. [PMID: 35679674 PMCID: PMC9189218 DOI: 10.1016/j.psj.2022.101941] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/16/2022] [Accepted: 04/25/2022] [Indexed: 11/22/2022] Open
Abstract
Fowl adenovirus serotype 4 (FAdV-4), the predominant causative agent of hepatitis-hydropericardium syndrome (HHS), has caused severe economic losses to poultry industry since 2015. Although fiber2 and hexon have been confirmed to be the virulence-related factors, the roles of nonstructural viral proteins in pathogenicity of FAdV-4 remain poorly understood. Here, a tandem mass spectrometry (MS) was used to identify host factors interacted with 100K protein of hypervirulent FAdV-4 isolate (CH/HNJZ/2015), and 2595 cellular proteins associated with many biological processes and pathways were identified according to Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses. Among the proteins, HSC70 was verified to interact with 100K through co-immunoprecipitation assay. Notably, overexpression of HSC70 promoted the replication of FAdV-4 in LMH cells, whereas blocking HSC70 with inhibitor ver-155008 markedly suppressed viral replication. Collectively, these findings suggested that many cellular proteins involved in FAdV-4 infection through interacting with 100K and HSC70 positively regulated virus replication.
Collapse
|
15
|
Siriphanitchakorn T, Kini RM, Ooi EE, Choy MM. Revisiting dengue virus-mosquito interactions: molecular insights into viral fitness. J Gen Virol 2021; 102. [PMID: 34845981 PMCID: PMC8742994 DOI: 10.1099/jgv.0.001693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Dengue virus (DENV), like other viruses, closely interacts with the host cell machinery to complete its life cycle. Over the course of infection, DENV interacts with several host factors with pro-viral activities to support its infection. Meanwhile, it has to evade or counteract host factors with anti-viral activities which inhibit its infection. These molecular virus-host interactions play a crucial role in determining the success of DENV infection. Deciphering such interactions is thus paramount to understanding viral fitness in its natural hosts. While DENV-mammalian host interactions have been extensively studied, not much has been done to characterize DENV-mosquito host interactions despite its importance in controlling DENV transmission. Here, to provide a snapshot of our current understanding of DENV-mosquito interactions, we review the literature that identified host factors and cellular processes related to DENV infection in its mosquito vectors, Aedes aegypti and Aedes albopictus, with a particular focus on DENV-mosquito omics studies. This knowledge provides fundamental insights into the DENV life cycle, and could contribute to the development of novel antiviral strategies.
Collapse
Affiliation(s)
- Tanamas Siriphanitchakorn
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, 169857 Singapore, Singapore.,Department of Biological Sciences, Faculty of Science, National University of Singapore, 117558 Singapore, Singapore
| | - R Manjunatha Kini
- Department of Biological Sciences, Faculty of Science, National University of Singapore, 117558 Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore, Singapore
| | - Eng Eong Ooi
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, 169857 Singapore, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore, 117549 Singapore, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore
| | - Milly M Choy
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, 169857 Singapore, Singapore
| |
Collapse
|
16
|
Lubkowska A, Pluta W, Strońska A, Lalko A. Role of Heat Shock Proteins (HSP70 and HSP90) in Viral Infection. Int J Mol Sci 2021; 22:ijms22179366. [PMID: 34502274 PMCID: PMC8430838 DOI: 10.3390/ijms22179366] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 12/12/2022] Open
Abstract
Heat shock proteins (HSPs) are a large group of chaperones found in most eukaryotes and bacteria. They are responsible for the correct protein folding, protection of the cell against stressors, presenting immune and inflammatory cytokines; furthermore, they are important factors in regulating cell differentiation, survival and death. Although the biological function of HSPs is to maintain cell homeostasis, some of them can be used by viruses both to fold their proteins and increase the chances of survival in unfavorable host conditions. Folding viral proteins as well as replicating many different viruses are carried out by, among others, proteins from the HSP70 and HSP90 families. In some cases, the HSP70 family proteins directly interact with viral polymerase to enhance viral replication or they can facilitate the formation of a viral replication complex and/or maintain the stability of complex proteins. It is known that HSP90 is important for the expression of viral genes at both the transcriptional and the translational levels. Both of these HSPs can form a complex with HSP90 and, consequently, facilitate the entry of the virus into the cell. Current studies have shown the biological significance of HSPs in the course of infection SARS-CoV-2. A comprehensive understanding of chaperone use during viral infection will provide new insight into viral replication mechanisms and therapeutic potential. The aim of this study is to describe the molecular basis of HSP70 and HSP90 participation in some viral infections and the potential use of these proteins in antiviral therapy.
Collapse
Affiliation(s)
- Anna Lubkowska
- Department of Functional Diagnostics and Physical Medicine, Pomeranian Medical University in Szczecin, Żołnierska 54, 71-210 Szczecin, Poland;
- Correspondence:
| | - Waldemar Pluta
- Department of Functional Diagnostics and Physical Medicine, Pomeranian Medical University in Szczecin, Żołnierska 54, 71-210 Szczecin, Poland;
| | - Aleksandra Strońska
- Department of Pharmacognosy and Natural Medicines, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Alicja Lalko
- Student Research at the Chair and Department of Functional Diagnostics and Physical Medicine, Pomeranian Medical University, Żołnierska 54, 71-210 Szczecin, Poland;
| |
Collapse
|
17
|
Abstract
Zika virus (ZIKV; Flaviviridae) is a devastating virus transmitted to humans by the mosquito Aedes aegypti. The interaction of the virus with the mosquito vector is poorly known. The double-stranded RNA (dsRNA)-mediated interruption or activation of immunity-related genes in the Toll, IMD, JAK-STAT, and short interfering RNA (siRNA) pathways did not affect ZIKV infection in A. aegypti. Transcriptome-based analysis indicated that most immunity-related genes were upregulated in response to ZIKV infection, including leucine-rich immune protein (LRIM) genes. Further, there was a significant increment in the ZIKV load in LRIM9-, LRIM10A-, and LIRM10B-silenced A. aegypti, suggesting their function in modulating viral infection. Further, gene function enrichment analysis revealed that viral infection increased global ribosomal activity. Silencing of RpL23 and RpL27, two ribosomal large subunit genes, increased mosquito resistance to ZIKV infection. In vitro fat body culture assay revealed that the expression of RpL23 and RpL27 was responsive to the Juvenile hormone (JH) signaling pathway. These two genes were transcriptionally regulated by JH and its receptor methoprene-tolerant (Met) complex. Silencing of Met also inhibited ZIKV infection in A. aegypti. This suggests that ZIKV enhances ribosomal activity through JH regulation to promote infection in mosquitoes. Together, these data reveal A. aegypti immune responses to ZIKV and suggest a control strategy that reduces ZIKV transmission by modulating host factors. IMPORTANCE Most flaviviruses are transmitted between hosts by arthropod vectors such as mosquitoes. Since therapeutics or vaccines are lacking for most mosquito-borne diseases, reducing the mosquito vector competence is an effective way to decrease disease burden. We used high-throughput sequencing technology to study the interaction between mosquito Aedes aegypti and ZIKV. Leucine-rich immune protein (LRIM) genes were involved in the defense in response to viral infection. In addition, RNA interference (RNAi) silencing of RpL23 and RpL27, two JH-regulated ribosomal large subunit genes, suppressed ZIKV infection in A. aegypti. These results suggest a novel control strategy that could block the transmission of ZIKV.
Collapse
|
18
|
Elrefaey AME, Abdelnabi R, Rosales Rosas AL, Wang L, Basu S, Delang L. Understanding the Mechanisms Underlying Host Restriction of Insect-Specific Viruses. Viruses 2020; 12:E964. [PMID: 32878245 PMCID: PMC7552076 DOI: 10.3390/v12090964] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
Arthropod-borne viruses contribute significantly to global mortality and morbidity in humans and animals. These viruses are mainly transmitted between susceptible vertebrate hosts by hematophagous arthropod vectors, especially mosquitoes. Recently, there has been substantial attention for a novel group of viruses, referred to as insect-specific viruses (ISVs) which are exclusively maintained in mosquito populations. Recent discoveries of novel insect-specific viruses over the past years generated a great interest not only in their potential use as vaccine and diagnostic platforms but also as novel biological control agents due to their ability to modulate arbovirus transmission. While arboviruses infect both vertebrate and invertebrate hosts, the replication of insect-specific viruses is restricted in vertebrates at multiple stages of virus replication. The vertebrate restriction factors include the genetic elements of ISVs (structural and non-structural genes and the untranslated terminal regions), vertebrate host factors (agonists and antagonists), and the temperature-dependent microenvironment. A better understanding of these bottlenecks is thus warranted. In this review, we explore these factors and the complex interplay between ISVs and their hosts contributing to this host restriction phenomenon.
Collapse
Affiliation(s)
| | - Rana Abdelnabi
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, 3000 Leuven, Belgium; (R.A.); (A.L.R.R.); (L.W.)
| | - Ana Lucia Rosales Rosas
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, 3000 Leuven, Belgium; (R.A.); (A.L.R.R.); (L.W.)
| | - Lanjiao Wang
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, 3000 Leuven, Belgium; (R.A.); (A.L.R.R.); (L.W.)
| | - Sanjay Basu
- The Pirbright Institute, Pirbright, Woking GU24 0NF, UK;
| | - Leen Delang
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, 3000 Leuven, Belgium; (R.A.); (A.L.R.R.); (L.W.)
| |
Collapse
|
19
|
Wang Z, Li Y, Yang X, Zhao J, Cheng Y, Wang J. Mechanism and Complex Roles of HSC70 in Viral Infections. Front Microbiol 2020; 11:1577. [PMID: 32849328 PMCID: PMC7396710 DOI: 10.3389/fmicb.2020.01577] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022] Open
Abstract
Heat shock cognate 71-kDa protein (HSC70), a constitutively expressed molecular chaperon within the heat shock protein 70 family, plays crucial roles in maintaining cellular environmental homeostasis through implicating in a wide variety of physiological processes, such as ATP metabolism, protein folding and transporting, antigen processing and presentation, endocytosis, and autophagy. Notably, HSC70 also participates in multiple non-communicable diseases and some pathogen-caused infectious diseases. It is known that virus is an obligatory intracellular parasite and heavily relies on host machineries to self-replication. Undoubtedly, HSC70 is a striking target manipulated by virus to ensure the successful propagation. In this review, we summarize the recent advances of the regulatory mechanisms of HSC70 during viral infections, which will be conducive to further study viral pathogenesis.
Collapse
Affiliation(s)
- Zeng Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Yongtao Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Xia Yang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Jun Zhao
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Yuening Cheng
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Jianke Wang
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China.,Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
20
|
Wan Q, Song D, Li H, He ML. Stress proteins: the biological functions in virus infection, present and challenges for target-based antiviral drug development. Signal Transduct Target Ther 2020; 5:125. [PMID: 32661235 PMCID: PMC7356129 DOI: 10.1038/s41392-020-00233-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/26/2020] [Accepted: 06/13/2020] [Indexed: 02/06/2023] Open
Abstract
Stress proteins (SPs) including heat-shock proteins (HSPs), RNA chaperones, and ER associated stress proteins are molecular chaperones essential for cellular homeostasis. The major functions of HSPs include chaperoning misfolded or unfolded polypeptides, protecting cells from toxic stress, and presenting immune and inflammatory cytokines. Regarded as a double-edged sword, HSPs also cooperate with numerous viruses and cancer cells to promote their survival. RNA chaperones are a group of heterogeneous nuclear ribonucleoproteins (hnRNPs), which are essential factors for manipulating both the functions and metabolisms of pre-mRNAs/hnRNAs transcribed by RNA polymerase II. hnRNPs involve in a large number of cellular processes, including chromatin remodelling, transcription regulation, RNP assembly and stabilization, RNA export, virus replication, histone-like nucleoid structuring, and even intracellular immunity. Dysregulation of stress proteins is associated with many human diseases including human cancer, cardiovascular diseases, neurodegenerative diseases (e.g., Parkinson’s diseases, Alzheimer disease), stroke and infectious diseases. In this review, we summarized the biologic function of stress proteins, and current progress on their mechanisms related to virus reproduction and diseases caused by virus infections. As SPs also attract a great interest as potential antiviral targets (e.g., COVID-19), we also discuss the present progress and challenges in this area of HSP-based drug development, as well as with compounds already under clinical evaluation.
Collapse
Affiliation(s)
- Qianya Wan
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Dan Song
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Huangcan Li
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Ming-Liang He
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China. .,CityU Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
21
|
Chen C, Qin Y, Qian K, Shao H, Ye J, Qin A. HSC70 is required for infectious bursal disease virus (IBDV) infection in DF-1 cells. Virol J 2020; 17:65. [PMID: 32375812 PMCID: PMC7201719 DOI: 10.1186/s12985-020-01333-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 04/23/2020] [Indexed: 12/26/2022] Open
Abstract
Background Infectious bursal disease (IBD) is a highly contagious infectious disease that causes severe immunosuppression and damage to the bursa of Fabricius in chickens. Several proteins involved in IBD virus (IBDV) infection, such as surface immunoglobulin M, integrin, annexin A2 and chicken heat shock protein 90, have been identified. However, the main protein that plays key roles in virus infection has not yet been confirmed. Methods DF-1 cell line was transfected with the pcDNA-VP2 plasmid and analyzed by immunofluorescence assay. The proteins reacted with VP2 of IBDV in DF-1 cells were pulldown with the monoclonal antibody and identified by mass spectrometry. Heat shock cognate protein 70 (HSC70), one of these proteins, was selected to be investigated in the function in IBDV infection by specific antibody and its inhibitor. Results The DF-1 cell line was transfected with the pcDNA-VP2 plasmid, and expression of IBDV VP2 in DF-1 cells was confirmed by immunofluorescence assays. Heat shock cognate protein 70 (HSC70) was one of the proteins identified by coimmunoprecipitation using a monoclonal antibody (2H11) against VP2 and mass spectrometry analysis. IBDV infection in DF-1 cells was strongly inhibited by both an anti-HSC70 antibody and a HSC70 inhibitor (VER155008). Conclusion These results suggest that HSC70 may be an essential factor for IBDV infection.
Collapse
Affiliation(s)
- Chunbo Chen
- Ministry of Education Key Laboratory for Avian Preventive Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, China
| | - Ying Qin
- Ministry of Education Key Laboratory for Avian Preventive Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, China
| | - Kun Qian
- Ministry of Education Key Laboratory for Avian Preventive Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, 225009, China
| | - Hongxia Shao
- Ministry of Education Key Laboratory for Avian Preventive Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, 225009, China
| | - Jianqiang Ye
- Ministry of Education Key Laboratory for Avian Preventive Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, China. .,Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, 225009, China.
| | - Aijian Qin
- Ministry of Education Key Laboratory for Avian Preventive Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, China. .,Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, 225009, China.
| |
Collapse
|
22
|
Laureti M, Narayanan D, Rodriguez-Andres J, Fazakerley JK, Kedzierski L. Flavivirus Receptors: Diversity, Identity, and Cell Entry. Front Immunol 2018; 9:2180. [PMID: 30319635 PMCID: PMC6168832 DOI: 10.3389/fimmu.2018.02180] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 09/04/2018] [Indexed: 12/12/2022] Open
Abstract
Flaviviruses are emerging and re-emerging arthropod-borne pathogens responsible for significant mortality and morbidity worldwide. The genus comprises more than seventy small, positive-sense, single-stranded RNA viruses, which are responsible for a spectrum of human and animal diseases ranging in symptoms from mild, influenza-like infection to fatal encephalitis and haemorrhagic fever. Despite genomic and structural similarities across the genus, infections by different flaviviruses result in disparate clinical presentations. This review focusses on two haemorrhagic flaviviruses, dengue virus and yellow fever virus, and two neurotropic flaviviruses, Japanese encephalitis virus and Zika virus. We review current knowledge on host-pathogen interactions, virus entry strategies and tropism.
Collapse
Affiliation(s)
- Mathilde Laureti
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Divya Narayanan
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Julio Rodriguez-Andres
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - John K Fazakerley
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
23
|
Early Events in Japanese Encephalitis Virus Infection: Viral Entry. Pathogens 2018; 7:pathogens7030068. [PMID: 30104482 PMCID: PMC6161159 DOI: 10.3390/pathogens7030068] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/31/2018] [Accepted: 08/06/2018] [Indexed: 12/15/2022] Open
Abstract
Japanese encephalitis virus (JEV), a mosquito-borne zoonotic flavivirus, is an enveloped positive-strand RNA virus that can cause a spectrum of clinical manifestations, ranging from mild febrile illness to severe neuroinvasive disease. Today, several killed and live vaccines are available in different parts of the globe for use in humans to prevent JEV-induced diseases, yet no antivirals are available to treat JEV-associated diseases. Despite the progress made in vaccine research and development, JEV is still a major public health problem in southern, eastern, and southeastern Asia, as well as northern Oceania, with the potential to become an emerging global pathogen. In viral replication, the entry of JEV into the cell is the first step in a cascade of complex interactions between the virus and target cells that is required for the initiation, dissemination, and maintenance of infection. Because this step determines cell/tissue tropism and pathogenesis, it is a promising target for antiviral therapy. JEV entry is mediated by the viral glycoprotein E, which binds virions to the cell surface (attachment), delivers them to endosomes (endocytosis), and catalyzes the fusion between the viral and endosomal membranes (membrane fusion), followed by the release of the viral genome into the cytoplasm (uncoating). In this multistep process, a collection of host factors are involved. In this review, we summarize the current knowledge on the viral and cellular components involved in JEV entry into host cells, with an emphasis on the initial virus-host cell interactions on the cell surface.
Collapse
|
24
|
Vogels CB, Göertz GP, Pijlman GP, Koenraadt CJ. Vector competence of European mosquitoes for West Nile virus. Emerg Microbes Infect 2017; 6:e96. [PMID: 29116220 PMCID: PMC5717085 DOI: 10.1038/emi.2017.82] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/23/2017] [Accepted: 08/27/2017] [Indexed: 01/02/2023]
Abstract
West Nile virus (WNV) is an arthropod-borne flavivirus of high medical and veterinary importance. The main vectors for WNV are mosquito species of the Culex genus that transmit WNV among birds, and occasionally to humans and horses, which are ‘dead-end’ hosts. Recently, several studies have been published that aimed to identify the mosquito species that serve as vectors for WNV in Europe. These studies provide insight in factors that can influence vector competence of European mosquito species for WNV. Here, we review the current knowledge on vector competence of European mosquitoes for WNV, and the molecular knowledge on physical barriers, anti-viral pathways and microbes that influence vector competence based on studies with other flaviviruses. By comparing the 12 available WNV vector competence studies with European mosquitoes we evaluate the effect of factors such as temperature, mosquito origin and mosquito biotype on vector competence. In addition, we propose a standardised methodology to allow for comparative studies across Europe. Finally, we identify knowledge gaps regarding vector competence that, once addressed, will provide important insights into WNV transmission and ultimately contribute to effective strategies to control WNV.
Collapse
Affiliation(s)
- Chantal Bf Vogels
- Laboratory of Entomology, Wageningen University & Research, P.O. Box 16, 6700 AA, Wageningen, The Netherlands
| | - Giel P Göertz
- Laboratory of Virology, Wageningen University & Research, P.O. Box 16, 6700 AA, Wageningen, The Netherlands
| | - Gorben P Pijlman
- Laboratory of Virology, Wageningen University & Research, P.O. Box 16, 6700 AA, Wageningen, The Netherlands
| | - Constantianus Jm Koenraadt
- Laboratory of Entomology, Wageningen University & Research, P.O. Box 16, 6700 AA, Wageningen, The Netherlands
| |
Collapse
|
25
|
Hastings AK, Yockey LJ, Jagger BW, Hwang J, Uraki R, Gaitsch HF, Parnell LA, Cao B, Mysorekar IU, Rothlin CV, Fikrig E, Diamond MS, Iwasaki A. TAM Receptors Are Not Required for Zika Virus Infection in Mice. Cell Rep 2017; 19:558-568. [PMID: 28423319 DOI: 10.1016/j.celrep.2017.03.058] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 03/16/2017] [Accepted: 03/20/2017] [Indexed: 01/26/2023] Open
Abstract
Tyro3, Axl, and Mertk (TAM) receptors are candidate entry receptors for infection with the Zika virus (ZIKV), an emerging flavivirus of global public health concern. To investigate the requirement of TAM receptors for ZIKV infection, we used several routes of viral inoculation and compared viral replication in wild-type versus Axl-/-, Mertk-/-, Axl-/-Mertk-/-, and Axl-/-Tyro3-/- mice in various organs. Pregnant and non-pregnant mice treated with interferon-α-receptor (IFNAR)-blocking (MAR1-5A3) antibody and infected subcutaneously with ZIKV showed no reliance on TAMs for infection. In the absence of IFNAR-blocking antibody, adult female mice challenged intravaginally with ZIKV showed no difference in mucosal viral titers. Similarly, in young mice that were infected with ZIKV intracranially or intraperitoneally, ZIKV replication occurred in the absence of TAM receptors, and no differences in cell tropism were observed. These findings indicate that, in mice, TAM receptors are not required for ZIKV entry and infection.
Collapse
Affiliation(s)
- Andrew K Hastings
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Laura J Yockey
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Brett W Jagger
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jesse Hwang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ryuta Uraki
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Hallie F Gaitsch
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Lindsay A Parnell
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Bin Cao
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Indira U Mysorekar
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Carla V Rothlin
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Michael S Diamond
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Obstetrics and Gynecology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
26
|
Abstract
Viruses hijack host machineries for replicating themselves efficiently. Host protein quality control machineries (QC) not only assist protein folding to form bona fide proteins with active functions but also get rid of un/misfolded proteins via degradation to maintain the protein homeostasis. Previous studies have reported that viruses utilize QC at various steps for their lifecycles. Recently we defined Hsp70s and their cochaperones, DnaJs functions on Dengue lifecycle. Here we summarize the significance of QC on Dengue virus.
Collapse
|
27
|
Avila-Bonilla RG, Yocupicio-Monroy M, Marchat LA, De Nova-Ocampo MA, Del Ángel RM, Salas-Benito JS. Analysis of the miRNA profile in C6/36 cells persistently infected with dengue virus type 2. Virus Res 2017; 232:139-151. [PMID: 28267608 DOI: 10.1016/j.virusres.2017.03.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 12/28/2022]
Abstract
Dengue virus (DENV) is the most important arbovirus in the world; DENV is transmitted by the Aedes genus of mosquitoes and can establish a life-long persistent infection in mosquitoes. However, the exact mechanism by which persistent infection is established remains unknown. In this study the differential expression of miRNAs was analysed by deep sequencing and RT-qPCR using a previously established C6/36-HT cell line persistently infected with DENV 2 (C6-L) as a model. miR-927, miR-87, miR-210, miR-2a-3p, miR-190 and miR-970 were up-regulated, whereas miR-252, miR-263a-3p, miR-92b, miR-10-5p miR-9a-5p, miR-9a-1, miR-124, miR-286a and miR-286b were down-regulated in C6-L cells compared with C6/36 cells acutely infected with the same virus or mock-infected cells. Deep sequencing results were validated by RT-qPCR for the highly differentially expressed miR-927 and miR-9a-5p, which were up- and down-regulated, respectively, compared with both acutely and mock-infected C6/36 cells. The putative targets of these miRNAs include components of the ubiquitin conjugation pathway, vesicle-mediated transport, autophagy, and the JAK-STAT cascade as well as proteins with endopeptidase activity. Other putative targets include members of the Toll signalling pathway and proteins with kinase, ATPase, protease, scavenger receptor or Lectin C-type activity or that participate in fatty acid biosynthesis or oxidative stress. Our results suggest that several specific miRNAs help regulate the cellular functions that maintain equilibrium between viral replication and the antiviral response during persistent infection of mosquito cells. This study is the first report of a global miRNA profile in a mosquito cell line persistently infected with DENV.
Collapse
Affiliation(s)
- Rodolfo Gamaliel Avila-Bonilla
- Programa Institucional de Biomedicina Molecular, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera 249, La Escalera-Ticomán, Mexico City, CP 07320, Mexico; Programa de Doctorado en Ciencias en Biotecnología, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera 249, La Escalera-Ticomán, Mexico City, CP 07320, Mexico.
| | - Martha Yocupicio-Monroy
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, San Lorenzo 290, Del Valle Sur, Mexico City, CP 03100, Mexico.
| | - Laurence A Marchat
- Programa Institucional de Biomedicina Molecular, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera 249, La Escalera-Ticomán, Mexico City, CP 07320, Mexico; Programa de Doctorado en Ciencias en Biotecnología, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera 249, La Escalera-Ticomán, Mexico City, CP 07320, Mexico.
| | - Mónica A De Nova-Ocampo
- Programa Institucional de Biomedicina Molecular, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera 249, La Escalera-Ticomán, Mexico City, CP 07320, Mexico; Programa de Doctorado en Ciencias en Biotecnología, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera 249, La Escalera-Ticomán, Mexico City, CP 07320, Mexico.
| | - Rosa María Del Ángel
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados-IPN. Av, Instituto Politécnico Nacional 2508, San Pedro Zacatenco, Mexico City, CP 07360, Mexico.
| | - Juan Santiago Salas-Benito
- Programa Institucional de Biomedicina Molecular, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera 249, La Escalera-Ticomán, Mexico City, CP 07320, Mexico; Programa de Doctorado en Ciencias en Biotecnología, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera 249, La Escalera-Ticomán, Mexico City, CP 07320, Mexico.
| |
Collapse
|
28
|
Zhang Z, Yang X, Xu P, Wu X, Zhou L, Wang H. Heat shock protein 70 in lung and kidney of specific-pathogen-free chickens is a receptor-associated protein that interacts with the binding domain of the spike protein of infectious bronchitis virus. Arch Virol 2017; 162:1625-1631. [PMID: 28224252 PMCID: PMC7087019 DOI: 10.1007/s00705-017-3280-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/02/2017] [Indexed: 12/02/2022]
Abstract
Avian infectious bronchitis virus (IBV) is a member of the family Coronaviridae. A binding domain that mediates the attachment of the virus to its receptor has been identified in the S1 protein of prototype IBV strain M41. In this study, we identified this binding domain in a different strain, as well as the cellular proteins that interact with it. First, we expressed the S1N proteins (residues 19-270) of M41 and another isolate, SCZJ3, and compared the binding capacities of recombinant S1N-M41 and S1N-SCZJ3 to host tissues. Protein histochemistry showed that both S1N-M41 and S1N-SCZJ3 could bind to lung and kidney, and that recombinant S1N-SCZJ3 displayed a distinctive staining pattern in the proventriculus. Recombinant S1N-SCZJ3 was then employed to purify binding-associated proteins in lung, kidney, and proventriculus. Using an affinity chromatography assay, two common bands of about 60 kDa and 70 kDa were obtained from the total tissue proteins. These protein bands were identified by liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) as protein disulfide isomerase (PDI) and heat shock protein 70 (HSP70). Finally, infection of chicken embryo kidney (CEK) cells by SCZJ3 was found to be inhibited by anti-HSP70 but not anti-PDI polyclonal antibody. These data indicate that HSP70 is part of the receptor complex of IBV and might help to understand the mechanism of S-mediated cell entry of IBV.
Collapse
Affiliation(s)
- ZhiKun Zhang
- School of Life Science, Sichuan University, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, 29# Wangjiang Road, Chengdu, 610064, China
| | - Xin Yang
- School of Life Science, Sichuan University, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, 29# Wangjiang Road, Chengdu, 610064, China
| | - PengWei Xu
- School of Life Science, Sichuan University, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, 29# Wangjiang Road, Chengdu, 610064, China
| | - Xuan Wu
- School of Life Science, Sichuan University, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, 29# Wangjiang Road, Chengdu, 610064, China
| | - Long Zhou
- School of Life Science, Sichuan University, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, 29# Wangjiang Road, Chengdu, 610064, China
| | - HongNing Wang
- School of Life Science, Sichuan University, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, "985 Project" Science Innovative Platform for Resource and Environment Protection of Southwestern, Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, 29# Wangjiang Road, Chengdu, 610064, China.
| |
Collapse
|
29
|
Howe MK, Speer BL, Hughes PF, Loiselle DR, Vasudevan S, Haystead TAJ. An inducible heat shock protein 70 small molecule inhibitor demonstrates anti-dengue virus activity, validating Hsp70 as a host antiviral target. Antiviral Res 2016; 130:81-92. [PMID: 27058774 DOI: 10.1016/j.antiviral.2016.03.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/31/2016] [Accepted: 03/31/2016] [Indexed: 12/27/2022]
Abstract
An estimated three billion people are at risk of Dengue virus (DENV) infection worldwide and there are currently no approved therapeutic interventions for DENV infection. Due to the relatively small size of the DENV genome, DENV is reliant on host factors throughout the viral life cycle. The inducible form of Heat Shock Protein 70 (Hsp70i) has been implicated as a host factor in DENV pathogenesis, however the complete role remains to be elucidated. Here we further illustrate the importance of Hsp70i in dengue virus pathogenesis and describe the antiviral activity of the allosteric small molecule inhibitor that is selective for Hsp70i, called HS-72. In monocytes, Hsp70i is expressed at low levels preceding DENV infection, but Hsp70i expression is induced upon DENV infection. Targeting Hsp70i with HS-72, results in a dose dependent reduction in DENV infected monocytes, while cell viability was maintained. HS-72 works to reduce DENV infection by inhibiting the entry stage of the viral life cycle, through disrupting the association of Hsp70i with the DENV receptor complex. This work highlights Hsp70i as an antiviral target and HS-72 as a potential anti-DENV therapeutic agent.
Collapse
Affiliation(s)
- Matthew K Howe
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Brittany L Speer
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Philip F Hughes
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - David R Loiselle
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Subhash Vasudevan
- Emerging Infectious Diseases Program, Duke-NUS Graduate and Medical School, Singapore
| | - Timothy A J Haystead
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
30
|
Dengue virus NS1 protein interacts with the ribosomal protein RPL18: This interaction is required for viral translation and replication in Huh-7 cells. Virology 2015; 484:113-126. [DOI: 10.1016/j.virol.2015.05.017] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 05/22/2015] [Accepted: 05/24/2015] [Indexed: 01/06/2023]
|
31
|
Decoding protein networks during virus entry by quantitative proteomics. Virus Res 2015; 218:25-39. [PMID: 26365680 PMCID: PMC4914609 DOI: 10.1016/j.virusres.2015.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/31/2015] [Accepted: 09/08/2015] [Indexed: 01/05/2023]
Abstract
Virus entry into host cells relies on interactions between viral and host structures including lipids, carbohydrates and proteins. Particularly, protein–protein interactions between viral surface proteins and host proteins as well as secondary host protein–protein interactions play a pivotal role in coordinating virus binding and uptake. These interactions are dynamic and frequently involve multiprotein complexes. In the past decade mass spectrometry based proteomics methods have reached sensitivities and high throughput compatibilities of genomics methods and now allow the reliable quantitation of proteins in complex samples from limited material. As proteomics provides essential information on the biologically active entity namely the protein, including its posttranslational modifications and its interactions with other proteins, it is an indispensable method in the virologist's toolbox. Here we review protein interactions during virus entry and compare classical biochemical methods to study entry with novel technically advanced quantitative proteomics techniques. We highlight the value of quantitative proteomics in mapping functional virus entry networks, discuss the benefits and limitations and illustrate how the methodology will help resolve unsettled questions in virus entry research in the future.
Collapse
|
32
|
Diwaker D, Mishra KP, Ganju L, Singh SB. Protein disulfide isomerase mediates dengue virus entry in association with lipid rafts. Viral Immunol 2015; 28:153-60. [PMID: 25664880 DOI: 10.1089/vim.2014.0095] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Dengue virus (DENV) causes a febrile disease, infecting around 50-100 million people annually. The relationship between DENV proteins and host cellular responses during infection is unclear. This study investigated the interaction of host protein disulfide isomerase (PDI) with DENV proteins and role of lipid rafts in viral immunopathogenesis. Host viral protein interactions were studied by co-immunoprecipitation and co-localization. It was found that PDI interacts with DENV nonstructural protein 1 (NS1) intracellularly as well as on the surface in the lipid raft domain. Disruption of this key interaction between PDI and NS1 could be an important therapeutic strategy to block DENV infection.
Collapse
Affiliation(s)
- Drishya Diwaker
- Immunomodulation Laboratory, Defence Institute of Physiology and Allied Sciences , Delhi, India
| | | | | | | |
Collapse
|
33
|
|
34
|
Flavivirus entry receptors: an update. Viruses 2013; 6:69-88. [PMID: 24381034 PMCID: PMC3917432 DOI: 10.3390/v6010069] [Citation(s) in RCA: 227] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 12/12/2013] [Accepted: 12/12/2013] [Indexed: 12/15/2022] Open
Abstract
Flaviviruses enter host cells by endocytosis initiated when the virus particles interact with cell surface receptors. The current model suggests that flaviviruses use at least two different sets of molecules for infectious entry: attachment factors that concentrate and/or recruit viruses on the cell surface and primary receptor(s) that bind to virions and direct them to the endocytic pathway. Here, we present the currently available knowledge regarding the flavivirus receptors described so far with specific attention to C-type lectin receptors and the phosphatidylserine receptors, T-cell immunoglobulin and mucin domain (TIM) and TYRO3, AXL and MER (TAM). Their role in flavivirus attachment and entry as well as their implication in the virus biology will be discussed in depth.
Collapse
|