1
|
Liu YY, Li N, Chen XY, Wang H, Zhu SW, Yang L, Quan FY, Ma JC, Dai JW, Jiang YL, Xiang ZF, Cheng Q, Zhang WH, Chen KH, Hou W, Xiong HR. MicroRNA let-7a regulation of Hantaan virus replication by Targeting FAS Signaling Pathways. Virology 2024; 600:110254. [PMID: 39383773 DOI: 10.1016/j.virol.2024.110254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/16/2024] [Accepted: 09/26/2024] [Indexed: 10/11/2024]
Abstract
Hantaan virus (HTNV) infection in humans can cause hemorrhagic fever and renal syndrome (HFRS). Understanding host responses to HTNV infection is crucial for developing effective disease intervention strategies. Previous RNA-sequencing studies have investigated the role of microRNAs (miRNAs) in the post-transcriptional regulation of host genes in response to HTNV infection. In this study, we demonstrated that HTNV infection induces let-7a expression in human umbilical vein endothelial cells (HUVEC) and that HTNV G protein upregulates the expression of let-7a. miRNA let-7a mimics and inhibitors validated the predicted targets, including cell apoptosis genes (FAS, caspase-8, and caspase-3) and inflammatory factors (IL-6 and its related factors). Modulation of miRNA let-7a levels by miRNA mimics and inhibitors affected HTNV replication, indicating that HTNV modulates host miRNA expression to affect the outcome of the antiviral host response.
Collapse
Affiliation(s)
- Yuan-Yuan Liu
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Ning Li
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China; Department of Blood Transfusion, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, China
| | - Xing-Yuan Chen
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Hui Wang
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China; School of Ecology and Environment, Tibet University, Lhasa, 850000, Tibet Autonomous Region, China
| | - Shao-Wei Zhu
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Lan Yang
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Fang-Yi Quan
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Jian-Chun Ma
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Jian-Wei Dai
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Ya-le Jiang
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China; Shenzhen Research Institute, Wuhan University, Shenzhen, 518057, Guangdong Province, China
| | - Zhou-Fu Xiang
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China; Shenzhen Research Institute, Wuhan University, Shenzhen, 518057, Guangdong Province, China
| | - Qi Cheng
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Wei-Hao Zhang
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Ke-Han Chen
- School of Public Health, Wuhan University, Wuhan, 430071, Hubei Province, China
| | - Wei Hou
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China; School of Ecology and Environment, Tibet University, Lhasa, 850000, Tibet Autonomous Region, China; Shenzhen Research Institute, Wuhan University, Shenzhen, 518057, Guangdong Province, China; School of Public Health, Wuhan University, Wuhan, 430071, Hubei Province, China.
| | - Hai-Rong Xiong
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China.
| |
Collapse
|
2
|
Zhang Y, Ma R, Wang Y, Sun W, Yang Z, Han M, Han T, Wu XA, Liu R. Viruses Run: The Evasion Mechanisms of the Antiviral Innate Immunity by Hantavirus. Front Microbiol 2021; 12:759198. [PMID: 34659193 PMCID: PMC8516094 DOI: 10.3389/fmicb.2021.759198] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 09/10/2021] [Indexed: 11/13/2022] Open
Abstract
Hantavirus can cause hemorrhagic fever with renal syndrome (HFRS) in Eurasia and hantavirus pulmonary syndrome (HPS) in America, with high mortality and unknown mechanisms. Innate immunity is the host's first-line defense to bridge the acquired immunity against viral infections. However, hantavirus has evolved various strategies in both molecular and cellular aspects to evade the host's natural immune surveillance. The Interferon-I (IFN-I) signaling pathway, a central link of host defense, induces various antiviral proteins to control the infection. This paper summarizes the molecular mechanisms of hantavirus evasion mechanisms of the IFN signaling pathway and cellular processes such as regulated cell death and cell stress. Besides, hantavirus could also evade immune surveillance evasion through cellular mechanisms, such as upregulating immune checkpoint molecules interfering with viral infections. Understanding hantavirus's antiviral immune evasion mechanisms will deepen our understanding of its pathogenesis and help us develop more effective methods to control and eliminate hantavirus.
Collapse
Affiliation(s)
- Yusi Zhang
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi΄an, China
| | - Ruixue Ma
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi΄an, China
| | - Yutong Wang
- School of Basic Medicine, Fourth Military Medical University, Xi΄an, China
| | - Wenjie Sun
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi΄an, China
| | - Ziwei Yang
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi΄an, China
| | - Mingwei Han
- School of Basic Medicine, Fourth Military Medical University, Xi΄an, China
| | - Tixin Han
- School of Basic Medicine, Fourth Military Medical University, Xi΄an, China
| | - Xing-an Wu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi΄an, China
| | - Rongrong Liu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi΄an, China
| |
Collapse
|
3
|
Chen QZ, Wang X, Luo F, Li N, Zhu N, Lu S, Zan YX, Zhong CJ, Wang MR, Hu HT, Zhang YZ, Xiong HR, Hou W. HTNV Sensitizes Host Toward TRAIL-Mediated Apoptosis-A Pivotal Anti-hantaviral Role of TRAIL. Front Immunol 2020; 11:1072. [PMID: 32636833 PMCID: PMC7317014 DOI: 10.3389/fimmu.2020.01072] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 05/04/2020] [Indexed: 01/15/2023] Open
Abstract
Hantaviruses can cause hemorrhagic fever with renal syndrome (HFRS) in Eurasia and have led to public health threat in China. The pathogenesis of HFRS is complex and involves capillary leakage due to the infection of vascular endothelial cells. Accumulating evidence has demonstrated that hantavirus can induce apoptosis in many cells, but the mechanism remains unclear. Our studies showed that Hantaan virus (HTNV) infection could induce TNF-related apoptosis-inducing ligand (TRAIL) expression in primary human umbilical vein endothelial cells (HUVECs) and sensitize host cells toward TRAIL-mediated apoptosis. Furthermore, TRAIL interference could inhibit apoptosis and enhance the production of HTNV as well as reduce IFN-β production, while exogenous TRAIL treatment showed reverse outcome: enhanced apoptosis and IFN-β production as well as a lower level of viral replication. We also observed that nucleocapsid protein (NP) and glycoprotein (GP) of HTNV could promote the transcriptions of TRAIL and its receptors. Thus, TRAIL was upregulated by HTNV infection and then exhibited significant antiviral activities in vitro, and it was further confirmed in the HTNV-infected suckling mice model that TRAIL treatment significantly reduced viral load, alleviated virus-induced tissue lesions, increased apoptotic cells, and decreased the mortality. In conclusion, these results demonstrate that TRAIL-dependent apoptosis and IFN-β production could suppress HTNV replication and TRAIL treatment might be a novel therapeutic target for HTNV infection.
Collapse
Affiliation(s)
- Qing-Zhou Chen
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy & Immunology, School of Basic Medical Sciences, Institute of Medical Virology, Wuhan University, Wuhan, China
| | - Xin Wang
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy & Immunology, School of Basic Medical Sciences, Institute of Medical Virology, Wuhan University, Wuhan, China
| | - Fan Luo
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy & Immunology, School of Basic Medical Sciences, Institute of Medical Virology, Wuhan University, Wuhan, China
| | - Ning Li
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy & Immunology, School of Basic Medical Sciences, Institute of Medical Virology, Wuhan University, Wuhan, China
| | - Ni Zhu
- Department of Microbiology, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, China
| | - Shuang Lu
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy & Immunology, School of Basic Medical Sciences, Institute of Medical Virology, Wuhan University, Wuhan, China
| | - Yu-Xing Zan
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy & Immunology, School of Basic Medical Sciences, Institute of Medical Virology, Wuhan University, Wuhan, China
| | - Chao-Jie Zhong
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy & Immunology, School of Basic Medical Sciences, Institute of Medical Virology, Wuhan University, Wuhan, China
| | - Mei-Rong Wang
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy & Immunology, School of Basic Medical Sciences, Institute of Medical Virology, Wuhan University, Wuhan, China
| | - Hai-Tao Hu
- Department of Microbiology & Immunology and Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX, United States
| | - Yong-Zhen Zhang
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, National Institute for Communicable Disease Control and Prevention, Beijing, China
| | - Hai-Rong Xiong
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy & Immunology, School of Basic Medical Sciences, Institute of Medical Virology, Wuhan University, Wuhan, China
| | - Wei Hou
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy & Immunology, School of Basic Medical Sciences, Institute of Medical Virology, Wuhan University, Wuhan, China.,Department of Microbiology, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
4
|
Davies K, Afrough B, Mankouri J, Hewson R, Edwards TA, Barr JN. Tula orthohantavirus nucleocapsid protein is cleaved in infected cells and may sequester activated caspase-3 during persistent infection to suppress apoptosis. J Gen Virol 2019; 100:1208-1221. [PMID: 31268416 DOI: 10.1099/jgv.0.001291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The family Hantaviridae mostly comprises rodent-borne segmented negative-sense RNA viruses, many of which are capable of causing devastating disease in humans. In contrast, hantavirus infection of rodent hosts results in a persistent and inapparent infection through their ability to evade immune detection and inhibit apoptosis. In this study, we used Tula hantavirus (TULV) to investigate the interplay between viral and host apoptotic responses during early, peak and persistent phases of virus infection in cell culture. Examination of early-phase TULV infection revealed that infected cells were refractory to apoptosis, as evidenced by the complete lack of cleaved caspase-3 (casp-3C) staining, whereas in non-infected bystander cells casp-3C was highly abundant. Interestingly, at later time points, casp-3C was abundant in infected cells, but the cells remained viable and able to continue shedding infectious virus, and together these observations were suggestive of a TULV-associated apoptotic block. To investigate this block, we viewed TULV-infected cells using laser scanning confocal and wide-field deconvolution microscopy, which revealed that TULV nucleocapsid protein (NP) colocalized with, and sequestered, casp-3C within cytoplasmic ultrastructures. Consistent with casp-3C colocalization, we showed for the first time that TULV NP was cleaved in cells and that TULV NP and casp-3C could be co-immunoprecipitated, suggesting that this interaction was stable and thus unlikely to be solely confined to NP binding as a substrate to the casp-3C active site. To account for these findings, we propose a novel mechanism by which TULV NP inhibits apoptosis by spatially sequestering casp-3C from its downstream apoptotic targets within the cytosol.
Collapse
Affiliation(s)
- Katherine Davies
- School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Babak Afrough
- National Infection Service, Public Health England, Porton Down, Salisbury, SP4 0JG, UK
| | - Jamel Mankouri
- School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK.,Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Roger Hewson
- National Infection Service, Public Health England, Porton Down, Salisbury, SP4 0JG, UK
| | - Thomas A Edwards
- School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK.,Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - John N Barr
- School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK.,Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| |
Collapse
|
5
|
Colby LA, Zitzow L. Applied Institutional Approaches for the Evaluation and Management of Zoonoses in Contemporary Laboratory Animal Research Facilities. ILAR J 2018; 59:134-143. [DOI: 10.1093/ilar/ily016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 08/23/2018] [Indexed: 01/05/2023] Open
Abstract
Abstract
Zoonoses, diseases transmitted between animals and humans, have been a concern in laboratory animal medicine for decades. Exposure to zoonotic organisms not only poses health risks to personnel and research animals but may also affect research integrity. Early laboratory animal programs were ineffective at excluding and preventing transmission of zoonotic diseases: the health status of the animals were often unknown, endemic diseases occurred frequently, housing conditions were less controlled, and veterinary care programs were decentralized. Over time, these conditions improved, but despite this, zoonotic diseases remain a contemporary concern. To reduce the incidence of zoonoses, management should perform an accurate risk assessment that takes into account the type of research performed, animal species used, animal sources, and housing conditions. Specific research practices, such as the use of biological materials, can also affect the risk assessment and should be considered. Once identified, the characteristics of significant zoonotic organisms can be examined. In addition, personnel attitudes and training, facility design and management, equipment availability, personal protective equipment used, standard operating procedures, and the institution’s vermin control program can impact the risk assessment. The effectiveness of the occupational health and safety program at managing risks of zoonoses should also be examined. Risk assessment, in the context of zoonotic disease prevention, is a complex exercise and is most effective when a team approach is used and includes research, husbandry, veterinary, and biosafety personnel.
Collapse
Affiliation(s)
- Lesley A Colby
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Lois Zitzow
- Department of Population Health, University of Georgia College of Veterinary Medicine, Athens, Georgia
| |
Collapse
|
6
|
Reuter M, Krüger DH. The nucleocapsid protein of hantaviruses: much more than a genome-wrapping protein. Virus Genes 2017; 54:5-16. [PMID: 29159494 DOI: 10.1007/s11262-017-1522-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/11/2017] [Indexed: 12/11/2022]
Abstract
The nucleocapsid (N) protein of hantaviruses represents an impressive example of a viral multifunctional protein. It encompasses properties as diverse as genome packaging, RNA chaperoning, intracellular protein transport, DNA degradation, intervention in host translation, and restricting host immune responses. These functions all rely on the capability of N to interact with RNA and other viral and cellular proteins. We have compiled data on the N protein of different hantavirus species together with information of the recently published three-dimensional structural data of the protein. The array of diverse functional activities accommodated in the hantaviral N protein goes far beyond to be a static structural protein and makes it an interesting target in the development of antiviral therapeutics.
Collapse
Affiliation(s)
- Monika Reuter
- Institute of Virology, Helmut-Ruska-Haus, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.
| | - Detlev H Krüger
- Institute of Virology, Helmut-Ruska-Haus, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
7
|
Meyer B, Groseth A. Apoptosis during arenavirus infection: mechanisms and evasion strategies. Microbes Infect 2017; 20:65-80. [PMID: 29081359 DOI: 10.1016/j.micinf.2017.10.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/15/2017] [Accepted: 10/16/2017] [Indexed: 11/17/2022]
Abstract
In recent years there has been a greatly increased interest in the interactions of arenaviruses with the apoptotic machinery, and particularly the extent to which these interactions may be an important contributor to pathogenesis. Here we summarize the current state of our knowledge on this subject and address the potential for interplay with other immunological mechanisms known to be regulated by these viruses. We also compare and contrast what is known for arenavirus-induced apoptosis with observations from other segmented hemorrhagic fever viruses.
Collapse
Affiliation(s)
- Bjoern Meyer
- Viral Populations and Pathogenesis Unit, Institut Pasteur, 28 rue du Dr. Roux, 75724 Paris cedex 15, France.
| | - Allison Groseth
- Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald - Insel Riems, Germany
| |
Collapse
|
8
|
Ma H, Han P, Ye W, Chen H, Zheng X, Cheng L, Zhang L, Yu L, Wu X, Xu Z, Lei Y, Zhang F. The Long Noncoding RNA NEAT1 Exerts Antihantaviral Effects by Acting as Positive Feedback for RIG-I Signaling. J Virol 2017; 91:e02250-16. [PMID: 28202761 PMCID: PMC5391460 DOI: 10.1128/jvi.02250-16] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 02/07/2017] [Indexed: 11/20/2022] Open
Abstract
Hantavirus infection, which causes zoonotic diseases with a high mortality rate in humans, has long been a global public health concern. Over the past decades, accumulating evidence suggests that long noncoding RNAs (lncRNAs) play key regulatory roles in innate immunity. However, the involvement of host lncRNAs in hantaviral control remains uncharacterized. In this study, we identified the lncRNA NEAT1 as a vital antiviral modulator. NEAT1 was dramatically upregulated after Hantaan virus (HTNV) infection, whereas its downregulation in vitro or in vivo delayed host innate immune responses and aggravated HTNV replication. Ectopic expression of NEAT1 enhanced beta interferon (IFN-β) production and suppressed HTNV infection. Further investigation suggested that NEAT1 served as positive feedback for RIG-I signaling. HTNV infection activated NEAT1 transcription through the RIG-I-IRF7 pathway, whereas NEAT1 removed the transcriptional inhibitory effects of the splicing factor proline- and glutamine-rich protein (SFPQ) by relocating SFPQ to paraspeckles, thus promoting the expression of RIG-I and DDX60. RIG-I and DDX60 had synergic effects on IFN production. Taken together, our findings demonstrate that NEAT1 modulates the innate immune response against HTNV infection, providing another layer of information about the role of lncRNAs in controlling viral infections.IMPORTANCE Hantaviruses have attracted worldwide attention as archetypal emerging pathogens. Recently, increasing evidence has highlighted long noncoding RNAs (lncRNAs) as key regulators of innate immunity; however, their roles in hantavirus infection remain unknown. In the present work, a new unexplored function of lncRNA NEAT1 in controlling HTNV replication was found. NEAT1 promoted interferon (IFN) responses by acting as positive feedback for RIG-I signaling. This lncRNA was induced by HTNV through the RIG-I-IRF7 pathway in a time- and dose-dependent manner and promoted HTNV-induced IFN production by facilitating RIG-I and DDX60 expression. Intriguingly, NEAT1 relocated SFPQ and formed paraspeckles after HTNV infection, which might reverse inhibitive effects of SFPQ on the transcription of RIG-I and DDX60. To the best of our knowledge, this is the first study to address the regulatory role of the lncRNA NEAT1 in host innate immunity after HTNV infection. In summary, our findings provide additional insights regarding the role of lncRNAs in controlling viral infections.
Collapse
Affiliation(s)
- Hongwei Ma
- Department of Microbiology, Fourth Military Medical University, Xi'an, China
| | - Peijun Han
- Department of Microbiology, Fourth Military Medical University, Xi'an, China
| | - Wei Ye
- Department of Microbiology, Fourth Military Medical University, Xi'an, China
| | - Hesong Chen
- Department of Microbiology, Fourth Military Medical University, Xi'an, China
| | - Xuyang Zheng
- Center of Infectious Diseases, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Linfeng Cheng
- Department of Microbiology, Fourth Military Medical University, Xi'an, China
| | - Liang Zhang
- Department of Microbiology, Fourth Military Medical University, Xi'an, China
| | - Lan Yu
- Department of Microbiology, Fourth Military Medical University, Xi'an, China
| | - Xing'an Wu
- Department of Microbiology, Fourth Military Medical University, Xi'an, China
| | - Zhikai Xu
- Department of Microbiology, Fourth Military Medical University, Xi'an, China
| | - Yingfeng Lei
- Department of Microbiology, Fourth Military Medical University, Xi'an, China
| | - Fanglin Zhang
- Department of Microbiology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
9
|
Chalaem P, Chusri S, Fernandez S, Chotigeat W, Anguita J, Pal U, Promnares K. Characterization of a Chikungunya virus strain isolated from banked patients' sera. Virol J 2016; 13:150. [PMID: 27590311 PMCID: PMC5009685 DOI: 10.1186/s12985-016-0606-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/24/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Chikungunya virus (CHIKV) is a prevalent mosquito-borne pathogen that is emerging in many parts of the globe causing significant human morbidity. Here, we report the isolation and characterization of an infectious CHIKV from banked serum specimens of suspected patients from the 2009 epidemic in Thailand. METHODS Standard plaque assay was used for CHIKV isolation from the banked serum specimens. Isolated CHIKV was identified base on E1 structural gene sequence. Growth kinetic, infectivity, cell viability and cytokine gene expression throughout CHIKV infection in a permissive cell line, 293T cells, was performed using several approaches, including standard plaque assay, immunofluorescence assay, classical MTT assay, and quantitative real-time PCR. Two tailed Student's t test was used for evaluation statistically significance between the mean values of the groups. RESULTS Based on the E1 structural gene sequence and phylogenetic analysis, we identified the virus as the CHIK/SBY8/10 isolate from Indonesia. Assessment of the growth kinetics, cytopathic effects as well as its ability to induce cellular immune responses suggested that the currently isolated CHIK/SBY8/10 virus is relatively more virulent than a known CHIKV vaccine strain, which also induces more dramatic proinflammatory responses. CONCLUSIONS Our studies further add to the infectivity of a less-studied yet infectious CHIKV isolate as well as underscored the importance and utility of 293T cells as an excellent cell culture model for studying viral growth, CHIKV-induced inflammatory cellular responses and cell death. Together, these studies provide novel information on the CHIKV biology, infectivity and virus-cell interaction, which would help develop novel interventions against the infection.
Collapse
Affiliation(s)
- Pattra Chalaem
- Department of Molecular Biotechnology and Bioinformatics, Faculty of Science, Prince of Songkla University, Hatyai, Songkhla 90112 Thailand
| | - Sarunyou Chusri
- Division of Infectious Disease, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hatyai, Songkhla 90112 Thailand
| | - Stefan Fernandez
- Department of Virology, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | - Wilaiwan Chotigeat
- Department of Molecular Biotechnology and Bioinformatics, Faculty of Science, Prince of Songkla University, Hatyai, Songkhla 90112 Thailand
| | - Juan Anguita
- CIC bioGUNE, 48160 Derio, Bizkaia Spain
- Ikerbasque, Basque Foundation for Science, 48011 Bilbao, Bizkaia Spain
| | - Utpal Pal
- Department of Veterinary Medicine and Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742 USA
| | - Kamoltip Promnares
- Department of Molecular Biotechnology and Bioinformatics, Faculty of Science, Prince of Songkla University, Hatyai, Songkhla 90112 Thailand
| |
Collapse
|
10
|
Ermonval M, Baychelier F, Tordo N. What Do We Know about How Hantaviruses Interact with Their Different Hosts? Viruses 2016; 8:v8080223. [PMID: 27529272 PMCID: PMC4997585 DOI: 10.3390/v8080223] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/27/2016] [Accepted: 08/05/2016] [Indexed: 11/26/2022] Open
Abstract
Hantaviruses, like other members of the Bunyaviridae family, are emerging viruses that are able to cause hemorrhagic fevers. Occasional transmission to humans is due to inhalation of contaminated aerosolized excreta from infected rodents. Hantaviruses are asymptomatic in their rodent or insectivore natural hosts with which they have co-evolved for millions of years. In contrast, hantaviruses cause different pathologies in humans with varying mortality rates, depending on the hantavirus species and its geographic origin. Cases of hemorrhagic fever with renal syndrome (HFRS) have been reported in Europe and Asia, while hantavirus cardiopulmonary syndromes (HCPS) are observed in the Americas. In some cases, diseases caused by Old World hantaviruses exhibit HCPS-like symptoms. Although the etiologic agents of HFRS were identified in the early 1980s, the way hantaviruses interact with their different hosts still remains elusive. What are the entry receptors? How do hantaviruses propagate in the organism and how do they cope with the immune system? This review summarizes recent data documenting interactions established by pathogenic and nonpathogenic hantaviruses with their natural or human hosts that could highlight their different outcomes.
Collapse
Affiliation(s)
- Myriam Ermonval
- Unité des Stratégies Antivirales, Département de Virologie, Institut Pasteur, 25 Rue du Docteur Roux, 75015 Paris, France.
| | - Florence Baychelier
- Unité des Stratégies Antivirales, Département de Virologie, Institut Pasteur, 25 Rue du Docteur Roux, 75015 Paris, France.
| | - Noël Tordo
- Unité des Stratégies Antivirales, Département de Virologie, Institut Pasteur, 25 Rue du Docteur Roux, 75015 Paris, France.
| |
Collapse
|
11
|
Pádua MD, Souza WMD, Lauretti F, Figueiredo LTM. Development of a novel plaque reduction neutralisation test for hantavirus infection. Mem Inst Oswaldo Cruz 2015; 110:624-8. [PMID: 26132430 PMCID: PMC4569825 DOI: 10.1590/0074-02760150102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 06/03/2015] [Indexed: 01/19/2023] Open
Abstract
In the Americas, hantaviruses cause severe cardiopulmonary syndrome (HCPS) with a
high fatality rate. Hantavirus infection is commonly diagnosed using serologic
techniques and reverse transcription-polymerase chain reaction. This paper presents a
novel plaque reduction neutralisation test (PRNT) for detecting antibodies to
Brazilian hantavirus. Using PRNT, plaque detection was enhanced by adding 0.6% of
dimethyl sulfoxide into the overlay culture medium of the infected cells. This
procedure facilitated clear visualisation of small plaques under the microscope and
provided for easy and accurate plaque counting. The sera from 37 HCPS patients from
the city of Ribeirão Preto, Brazil was evaluated for the Rio Mamoré virus (RIOMV)
using PRNT. Six samples exhibited neutralising antibodies; these antibodies exhibited
a low titre. The low level of seropositive samples may be due to fewer
cross-reactions between two different hantavirus species; the patients were likely
infected by Araraquara virus (a virus that has not been isolated) and RIOMV was used
for the test. This assay offers a new approach to evaluating and measuring
neutralising antibodies produced during hantavirus infections and it can be adapted
to other hantaviruses, including viruses that will be isolated in the future.
Collapse
Affiliation(s)
- Michelly de Pádua
- Centro de Pesquisa em Virologia, Faculdade de Medicina de Riberão Preto, Universidade de São Paulo, Ribeirão Preto, SP, BR
| | - William Marciel de Souza
- Centro de Pesquisa em Virologia, Faculdade de Medicina de Riberão Preto, Universidade de São Paulo, Ribeirão Preto, SP, BR
| | - Flávio Lauretti
- Centro de Pesquisa em Virologia, Faculdade de Medicina de Riberão Preto, Universidade de São Paulo, Ribeirão Preto, SP, BR
| | - Luiz Tadeu Moraes Figueiredo
- Centro de Pesquisa em Virologia, Faculdade de Medicina de Riberão Preto, Universidade de São Paulo, Ribeirão Preto, SP, BR
| |
Collapse
|
12
|
Abstract
Emerging infectious diseases of zoonotic origin are shaping today's infectious disease field more than ever. In this article, we introduce and review three emerging zoonotic viruses. Novel hantaviruses emerged in the Americas in the mid-1990s as the cause of severe respiratory infections, designated hantavirus pulmonary syndrome, with case fatality rates of around 40%. Nipah virus emerged a few years later, causing respiratory infections and encephalitis in Southeast Asia, with case fatality rates ranging from 40% to more than 90%. A new coronavirus emerged in 2012 on the Arabian Peninsula with a clinical syndrome of acute respiratory infections, later designated as Middle East respiratory syndrome (MERS), and an initial case fatality rate of more than 40%. Our current state of knowledge on the pathogenicity of these three severe, emerging viral infections is discussed.
Collapse
Affiliation(s)
- David Safronetz
- Laboratory of Virology, Division of Intramural Research, Rocky Mountain Laboratories, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana; , ,
| | | | | |
Collapse
|
13
|
Outinen TK, Mäkelä S, Huttunen R, Mäenpää N, Libraty D, Vaheri A, Mustonen J, Aittoniemi J. Urine soluble urokinase-type plasminogen activator receptor levels correlate with proteinuria in Puumala hantavirus infection. J Intern Med 2014; 276:387-95. [PMID: 24717117 PMCID: PMC4172514 DOI: 10.1111/joim.12257] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Urokinase-type plasminogen activator receptor (uPAR) is upregulated during inflammation and known to bind to β3 -integrins, receptors used by pathogenic hantaviruses to enter endothelial cells. It has been proposed that soluble uPAR (suPAR) is a circulating factor that causes focal segmental glomerulosclerosis and proteinuria by activating β3 -integrin in kidney podocytes. Proteinuria is also a characteristic feature of hantavirus infections. The aim of this study was to evaluate the relation between urine suPAR levels and disease severity in acute Puumala hantavirus (PUUV) infection. DESIGN A single-centre, prospective cohort study. SUBJECTS AND METHODS Urinary suPAR levels were measured twice during the acute phase and once during convalescence in 36 patients with serologically confirmed PUUV infection. Fractional excretion of suPAR (FE suPAR) and of albumin (FE alb) was calculated. RESULTS The FE suPAR was significantly elevated during the acute phase of PUUV infection compared to the convalescent phase (median 3.2%, range 0.8-52.0%, vs. median 1.9%, range 1.0-5.8%, P = 0.005). Maximum FE suPAR was correlated markedly with maximum FE alb (r = 0.812, P < 0.001) and with several other variables that reflect disease severity. There was a positive correlation with the length of hospitalization (r = 0.455, P = 0.009) and maximum plasma creatinine level (r = 0.780, P < 0.001) and an inverse correlation with minimum urinary output (r = -0.411, P = 0.030). There was no correlation between FE suPAR and plasma suPAR (r = 0.180, P = 0.324). CONCLUSION Urinary suPAR is markedly increased during acute PUUV infection and is correlated with proteinuria. High urine suPAR level may reflect local production of suPAR in the kidney during the acute infection.
Collapse
Affiliation(s)
- T K Outinen
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Song L, Lu Y, He J, Yu Y, Zuo T, Li Y, Zhu H, Duan Q. Multi-organ lesions in suckling mice infected with SARS-associated mammalian reovirus linked with apoptosis induced by viral proteins μ1 and σ1. PLoS One 2014; 9:e92678. [PMID: 24664247 PMCID: PMC3963933 DOI: 10.1371/journal.pone.0092678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 02/24/2014] [Indexed: 12/30/2022] Open
Abstract
We reported the isolation and characterization of a novel mammalian reassortant reovirus BYD1 that may have played an accomplice role with SARS-coronavirus during the 2003 SARS pandemic. The pathogenic mechanism of this novel reovirus is unknown. Reovirus pathogenicity has been associated with virus-induced apoptosis in cultured cells and in vivo. The reovirus outer capsid protein μ1 is recognized as the primary determinant of reovirus-induced apoptosis. Here, we investigated the apoptosis induced by BYD1, its outer capsid protein μ1, and its cell-attachment protein σ1 to understand the pathogenesis of BYD1. We also investigated BYD1 caused systemic complications in suckling mice. Under electron microscopy, BYD1-infected cells showed characteristics typical of apoptosis. Notably, ectopically expressed μ1 and σ1 induced similar pathological apoptosis, independent of BYD1 infection, in host cells in which they were expressed, which suggests that μ1 and σ1 are both apoptotic virulence factors. Consistent with previous reports of reovirus pathogenicity, suckling mice intracranially inoculated with BYD1 developed central nerve damage, myocarditis, and pneumonia. Collectively, our data suggest that BYD1 μ1- and σ1-induced apoptosis is involved in the multi-organ lesions in a suckling mouse BYD1 infection model.
Collapse
Affiliation(s)
- Lihua Song
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yongfeng Lu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Jun He
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yonghui Yu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Tingting Zuo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yanwei Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Hong Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Qing Duan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| |
Collapse
|
15
|
Mustonen J, Mäkelä S, Outinen T, Laine O, Jylhävä J, Arstila PT, Hurme M, Vaheri A. The pathogenesis of nephropathia epidemica: new knowledge and unanswered questions. Antiviral Res 2013; 100:589-604. [PMID: 24126075 DOI: 10.1016/j.antiviral.2013.10.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 09/27/2013] [Accepted: 10/04/2013] [Indexed: 01/20/2023]
Abstract
Puumala virus (PUUV) causes an acute hemorrhagic fever with renal syndrome (HFRS), a zoonosis also called nephropathia epidemica (NE). The reservoir host of PUUV is the bank vole (Myodes glareolus). Herein we review the main clinical manifestations of NE, acute kidney injury, increased vascular permeability, coagulation abnormalities as well as pulmonary, cardiac, central nervous system and ocular manifestations of the disease. Several biomarkers of disease severity have recently been discovered: interleukin-6, pentraxin-3, C-reactive protein, indoleamine 2,3-dioxygenase, cell-free DNA, soluble urokinase-type plasminogen activator, GATA-3 and Mac-2 binding protein. The role of cytokines, vascular endothelial growth hormone, complement, bradykinin, cellular immune response and other mechanisms in the pathogenesis of NE as well as host genetic factors will be discussed. Finally therapeutic aspects and directions for further research will be handled.
Collapse
Affiliation(s)
- Jukka Mustonen
- School of Medicine, University of Tampere, Tampere, Finland; Department of Internal Medicine, Tampere University Hospital, Tampere, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Park SW, Han MG, Park C, Ju YR, Ahn BY, Ryou J. Hantaan virus nucleocapsid protein stimulates MDM2-dependent p53 degradation. J Gen Virol 2013; 94:2424-2428. [PMID: 23994832 DOI: 10.1099/vir.0.054312-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Apoptosis has been shown to be induced and downregulated by the Hantaan virus (HTNV) nucleocapsid (N) protein. To address these conflicting data, expression of the p53 protein, one of the key molecules involved in apoptosis, was assessed in the presence of the N protein in A549 and HeLa cells. The amount of p53, increased by drug treatment, was reduced when cells were infected with HTNV or transfected with an expression vector of the HTNV N protein. When cells were treated with a proteasome inhibitor (MG132) or an MDM2 antagonist (Nutlin-3), p53 expression was not reduced in N protein-overexpressed cells. We concluded that the HTNV N protein ubiquitinates and degrades p53 MDM2-dependently. Here we report downregulation of p53 expression through a post-translational mechanism: MDM2-dependent ubiquitination and degradation by the HTNV N protein. These results indicate that N protein-dependent p53 degradation through the ubiquitin proteasome system is one of the anti-apoptotic mechanisms employed by HTNV.
Collapse
Affiliation(s)
- Sun-Whan Park
- School of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Republic of Korea
- Division of Arboviruses, Center for Immunology & Pathology, National Institute of Health, Korea Centers for Disease Control & Prevention, Republic of Korea
| | - Myung-Guk Han
- Division of Arboviruses, Center for Immunology & Pathology, National Institute of Health, Korea Centers for Disease Control & Prevention, Republic of Korea
| | - Chan Park
- Division of Arboviruses, Center for Immunology & Pathology, National Institute of Health, Korea Centers for Disease Control & Prevention, Republic of Korea
| | - Young Ran Ju
- Division of Zoonoses, Center for Immunology & Pathology, National Institute of Health, Korea Centers for Disease Control & Prevention, Republic of Korea
| | - Byung-Yoon Ahn
- School of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Republic of Korea
| | - Jungsang Ryou
- Division of Arboviruses, Center for Immunology & Pathology, National Institute of Health, Korea Centers for Disease Control & Prevention, Republic of Korea
| |
Collapse
|
17
|
Spiropoulou CF, Srikiatkhachorn A. The role of endothelial activation in dengue hemorrhagic fever and hantavirus pulmonary syndrome. Virulence 2013; 4:525-36. [PMID: 23841977 PMCID: PMC5359750 DOI: 10.4161/viru.25569] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The loss of the endothelium barrier and vascular leakage play a central role in the pathogenesis of hemorrhagic fever viruses. This can be caused either directly by the viral infection and damage of the vascular endothelium, or indirectly by a dysregulated immune response resulting in an excessive activation of the endothelium. This article briefly reviews our knowledge of the importance of the disruption of the vascular endothelial barrier in two severe disease syndromes, dengue hemorrhagic fever and hantavirus pulmonary syndrome. Both viruses cause changes in vascular permeability without damaging the endothelium. Here we focus on our understanding of the virus interaction with the endothelium, the role of the endothelium in the induced pathogenesis, and the possible mechanisms by which each virus causes vascular leakage. Understanding the dynamics between viral infection and the dysregulation of the endothelial cell barrier will help us to define potential therapeutic targets for reducing disease severity.
Collapse
|
18
|
Hantavirus-infection confers resistance to cytotoxic lymphocyte-mediated apoptosis. PLoS Pathog 2013; 9:e1003272. [PMID: 23555267 PMCID: PMC3610645 DOI: 10.1371/journal.ppat.1003272] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 02/12/2013] [Indexed: 12/11/2022] Open
Abstract
Hantaviruses cause hemorrhagic fever with renal syndrome (HFRS) and hantavirus cardio-pulmonary syndrome (HCPS; also called hantavirus pulmonary syndrome (HPS)), both human diseases with high case-fatality rates. Endothelial cells are the main targets for hantaviruses. An intriguing observation in patients with HFRS and HCPS is that on one hand the virus infection leads to strong activation of CD8 T cells and NK cells, on the other hand no obvious destruction of infected endothelial cells is observed. Here, we provide an explanation for this dichotomy by showing that hantavirus-infected endothelial cells are protected from cytotoxic lymphocyte-mediated induction of apoptosis. When dissecting potential mechanisms behind this phenomenon, we discovered that the hantavirus nucleocapsid protein inhibits the enzymatic activity of both granzyme B and caspase 3. This provides a tentative explanation for the hantavirus-mediated block of cytotoxic granule-mediated apoptosis-induction, and hence the protection of infected cells from cytotoxic lymphocytes. These findings may explain why infected endothelial cells in hantavirus-infected patients are not destroyed by the strong cytotoxic lymphocyte response. Rodent-born hantaviruses cause two severe emerging diseases with high case-fatality rates in humans; hemorrhagic fever with renal syndrome (HFRS) in Eurasia and hantavirus cardio-pulmonary syndrome (HCPS; also called hantavirus pulmonary syndrome (HPS)) in the Americas. A hallmark of HFRS/HCPS is increased vascular permeability. While endothelial cells are the main targets for hantaviruses, infection per se is not lytic. Patients suffering from HFRS and HCPS show remarkable strong cytotoxic lymphocyte responses including high numbers of activated NK cells and antigen-specific CD8 T cells. Hence, it has been suggested that cytotoxic lymphocyte-mediated killing of hantavirus-infected endothelial cells might contribute to HFRS/HCPS-pathogenesis. Here, we show that hantaviruses protect infected endothelial cells from being killed by cytotoxic lymphocytes. Further, we also show that hantaviruses inhibit apoptosis in general. Hantaviruses are negative-stranded RNA viruses encoding four structural proteins. Interestingly, the nucleocapsid protein was shown to inhibit the enzymatic functions of both granzyme B and caspase 3, two enzymes crucial for cytotoxic lymphocyte-mediated killing of virus-infected cells. Our study provides new insights into the interactions between hantaviruses, infected cells, and cytotoxic lymphocytes, and argues against a role for cytotoxic lymphocyte-mediated killing of virus-infected endothelial cells in causing HFRS/HCPS.
Collapse
|
19
|
Ontiveros SJ, Li Q, Jonsson CB. Modulation of apoptosis and immune signaling pathways by the Hantaan virus nucleocapsid protein. Virology 2010; 401:165-78. [PMID: 20227103 DOI: 10.1016/j.virol.2010.02.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Revised: 12/12/2009] [Accepted: 02/10/2010] [Indexed: 01/23/2023]
Abstract
Herein, we show a direct relationship between the Hantaan virus (HTNV) nucleocapsid (N) protein and the modulation of apoptosis. We observed an increase in caspase-7 and -8, but not -9 in cells expressing HTNV N protein mutants lacking amino acids 270-330. Similar results were observed for the New World hantavirus, Andes virus. Nuclear factor kappa B (NF-kappaB) was sequestered in the cytoplasm after tumor necrosis factor receptor (TNFR) stimulation in cells expressing HTNV N protein. Further, TNFR stimulated cells expressing HTNV N protein inhibited caspase activation. In contrast, cells expressing N protein truncations lacking the region from amino acids 270-330 were unable to inhibit nuclear import of NF-kappaB and the mutants also triggered caspase activity. These results suggest that the HTNV circumvents host antiviral signaling and apoptotic response mediated by the TNFR pathway through host interactions with the N protein.
Collapse
Affiliation(s)
- Steven J Ontiveros
- Graduate Program in Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | |
Collapse
|
20
|
Maes P, Keyaerts E, Li S, Nlandu-Masunda V, Clement J, Van Ranst M. Replication reduction neutralization test, a quantitative RT-PCR-based technique for the detection of neutralizing hantavirus antibodies. J Virol Methods 2009; 159:295-9. [PMID: 19406158 DOI: 10.1016/j.jviromet.2009.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Revised: 04/06/2009] [Accepted: 04/20/2009] [Indexed: 11/25/2022]
Abstract
Hantaviruses, which are mainly rodent-borne viruses, cause hemorrhagic fever with renal syndrome in the Old World, and hantavirus pulmonary syndrome in the New World. A neutralization test based on quantitative RT-PCR, the replication reduction neutralization test (RRNT), was developed for efficient detection of hantavirus-neutralizing antibodies. The effectiveness of the RRNT was evaluated by examining several hantaviruses and hantavirus-specific convalescent human serum samples. All convalescent serum samples tested by RRNT caused significant decreases in hantavirus genomes with only one specific hantavirus species, which allowed a straightforward identification of the related hantavirus. The results obtained by RRNT were completely comparable with the results obtained by focus reduction neutralization test (FRNT). The RRNT approach is a reliable and rapid alternative for FRNT, hitherto considered as the gold standard for hantavirus serology.
Collapse
Affiliation(s)
- Piet Maes
- Laboratory of Clinical Virology, Hantavirus Reference Center, Rega Institute, Minderbroedersstraat 10, B3000 Leuven, Belgium.
| | | | | | | | | | | |
Collapse
|
21
|
Xu F, Liang X, Tesh RB, Xiao SY. Characterization of cell-death pathways in Punta Toro virus-induced hepatocyte injury. J Gen Virol 2008; 89:2175-2181. [PMID: 18753227 DOI: 10.1099/vir.0.2008/001644-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Punta Toro virus (PTV; genus Phlebovirus, family Bunyaviridae) causes apoptosis of hepatocytes in vivo in experimentally infected hamsters and in vitro in cultured HepG2 cells. Screening for expression of apoptosis-related genes has shown alterations in the genes for tumour necrosis factor-alpha (TNF-alpha) and the TNF receptor family. This study examined the roles of the TNF receptor-related extrinsic pathway and the Bcl-2 family-associated mitochondrial pathway in PTV-induced cell death. The effects of caspase inhibitors (caspIs) and TNF on cellular viability, virus replication, and morphological and biochemical changes in apoptosis were examined in HepG2 cells at different time points after infection with PTV (Adames strain). The results showed that caspIs dampened the virus-induced reduction in cellular viability, partially suppressed and delayed viral titres and antigen expression, and partially decreased the expression of apoptotic genes, caspase activities and DNA fragmentation. TNF treatment further decreased cellular viability after PTV infection and increased the level of apoptosis, whilst caspIs partially inhibited these effects. These findings indicate that TNF, caspase-8 and caspase-9 contribute to PTV-induced hepatocytic apoptosis and that additional mediators are probably also involved in this process. These mediators from different pathways correlated with one another and may be interlinked.
Collapse
Affiliation(s)
- Fangling Xu
- Department of Pathology and Center for Biodefense & Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Xiaodong Liang
- Department of Pathology and Center for Biodefense & Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Robert B Tesh
- Department of Pathology and Center for Biodefense & Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Shu-Yuan Xiao
- Department of Pathology and Center for Biodefense & Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
22
|
Strandin T, Hepojoki J, Wang H, Vaheri A, Lankinen H. Hantaviruses and TNF-alpha act synergistically to induce ERK1/2 inactivation in Vero E6 cells. Virol J 2008; 5:110. [PMID: 18822184 PMCID: PMC2569924 DOI: 10.1186/1743-422x-5-110] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2008] [Accepted: 09/29/2008] [Indexed: 01/09/2023] Open
Abstract
Background We have previously reported that the apathogenic Tula hantavirus induces apoptosis in Vero E6 epithelial cells. To assess the molecular mechanisms behind the induced apoptosis we studied the effects of hantavirus infection on cellular signaling pathways which promote cell survival. We previously also observed that the Tula virus-induced cell death process is augmented by external TNF-α. Since TNF-α is involved in the pathogenesis of hantavirus-caused hemorrhagic fever with renal syndrome (HFRS) we investigated its effects on HFRS-causing hantavirus-infected cells. Results We studied both apathogenic (Tula and Topografov) and pathogenic (Puumala and Seoul) hantaviruses for their ability to regulate cellular signaling pathways and observed a direct virus-mediated down-regulation of external signal-regulated kinases 1 and 2 (ERK1/2) survival pathway activity, which was dramatically enhanced by TNF-α. The fold of ERK1/2 inhibition correlated with viral replication efficiencies, which varied drastically between the hantaviruses studied. Conclusion We demonstrate that in the presence of a cytokine TNF-α, which is increased in HFRS patients, hantaviruses are capable of inactivating proteins that promote cell survival (ERK1/2). These results imply that hantavirus-infected epithelial cell barrier functions might be compromised in diseased individuals and could at least partially explain the mechanisms of renal dysfunction and the resulting proteinuria seen in HFRS patients.
Collapse
Affiliation(s)
- Tomas Strandin
- Department of Virology, Haartman Institute, P,O, Box 21, FI-00014, University of Helsinki, Finland.
| | | | | | | | | |
Collapse
|
23
|
Terajima M, Hayasaka D, Maeda K, Ennis FA. Immunopathogenesis of hantavirus pulmonary syndrome and hemorrhagic fever with renal syndrome: Do CD8+ T cells trigger capillary leakage in viral hemorrhagic fevers? Immunol Lett 2007; 113:117-20. [PMID: 17897725 PMCID: PMC2214668 DOI: 10.1016/j.imlet.2007.08.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Accepted: 08/18/2007] [Indexed: 12/23/2022]
Abstract
There are many viruses known to cause viral hemorrhagic fevers in humans. The mechanisms causing hemorrhage are likely to vary among viruses. Some viruses, such as Marburg virus, are directly cytopathic to infected endothelial cells, suggesting infection of endothelial cells alone can cause hemorrhage. On the other hand, there are viruses which infect endothelial cells without causing any cytopathic effects, suggesting the involvement of host immune responses in developing hemorrhage. Typical examples of these include viruses of the hantavirus species. We hypothesize that impairment of endothelial cell's defense mechanisms against cytotoxic CD8+ T cells is the mechanism of capillary leakage in hantavirus pulmonary syndrome and hemorrhagic fever with renal syndrome, which may be common to other viral hemorrhagic fevers. CD8+ T cells may be a potential target for therapy of some viral hemorrhagic fevers.
Collapse
|
24
|
Wahl-Jensen V, Chapman J, Asher L, Fisher R, Zimmerman M, Larsen T, Hooper JW. Temporal analysis of Andes virus and Sin Nombre virus infections of Syrian hamsters. J Virol 2007; 81:7449-62. [PMID: 17475651 PMCID: PMC1933362 DOI: 10.1128/jvi.00238-07] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Andes virus (ANDV) and Sin Nombre virus (SNV) are rodent-borne hantaviruses that cause a highly lethal hemorrhagic fever in humans known as hantavirus pulmonary syndrome (HPS). There are no vaccines or specific drugs to prevent or treat HPS, and the pathogenesis is not understood. Syrian hamsters infected with ANDV, but not SNV, develop a highly lethal disease that closely resembles HPS in humans. Here, we performed a temporal pathogenesis study comparing ANDV and SNV infections in hamsters. SNV was nonpathogenic and viremia was not detected despite the fact that all animals were infected. ANDV was uniformly lethal with a mean time to death of 11 days. The first pathology detected was lymphocyte apoptosis starting on day 4. Animals were viremic and viral antigen was first observed in multiple organs by days 6 and 8, respectively. Levels of infectious virus in the blood increased 4 to 5 logs between days 6 and 8. Pulmonary edema was first detected ultrastructurally on day 6. Ultrastructural analysis of lung tissues revealed the presence of large inclusion bodies and substantial numbers of vacuoles within infected endothelial cells. Paraendothelial gaps were not observed, suggesting that fluid leakage was transcellular and directly attributable to infecting virus. Taken together, these data imply that HPS treatment strategies aimed at preventing virus replication and dissemination will have the greatest probability of success if administered before the viremic phase; however, because vascular leakage is associated with infected endothelial cells, a therapeutic strategy targeting viral replication might be effective even at later times (e.g., after disease onset).
Collapse
Affiliation(s)
- Victoria Wahl-Jensen
- Virology Division, U.S. Army Medical Research Institute of Infectious Deseases, Fort Detrick, MD 21702, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Hayasaka D, Maeda K, Ennis FA, Terajima M. Increased permeability of human endothelial cell line EA.hy926 induced by hantavirus-specific cytotoxic T lymphocytes. Virus Res 2006; 123:120-7. [PMID: 16979772 DOI: 10.1016/j.virusres.2006.08.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2006] [Revised: 08/14/2006] [Accepted: 08/16/2006] [Indexed: 12/27/2022]
Abstract
Hantavirus infection causes two human diseases, hemorrhagic fever with renal syndrome and hantavirus pulmonary syndrome. The typical feature of these diseases is increased permeability in microvascular beds in the kidneys and the lungs, respectively. The mechanism of capillary leakage, however, is not understood. Some evidence suggests that hantavirus disease pathogenesis is immunologically mediated by cytotoxic T lymphocytes and other immune cells in target organs producing inflammatory cytokines. In this study we examined the roles of virus-specific cytotoxic T lymphocytes in increased permeability of human endothelial cells infected with hantavirus. We used a human CD8(+) hantavirus-specific cytotoxic T lymphocyte line, 1A-E2, specific for the HLA-A24-restricted epitope in Sin Nombre and Puumala virus G2 protein, and the human endothelial cell line, EA.hy926 that expresses HLA-A24 molecule. The cytotoxic T lymphocyte line recognized and lysed target cells infected with Sin Nombre virus, and in transwell permeability assays increased permeability of EA.hy926 cell monolayer infected with Sin Nombre virus or recombinant adenovirus expressing the Sin Nombre virus G2 protein. These results suggest that cytotoxic T lymphocyte activity contribute to capillary leakage observed in patients with hantavirus pulmonary syndrome or hemorrhagic fever with renal syndrome.
Collapse
Affiliation(s)
- Daisuke Hayasaka
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA
| | | | | | | |
Collapse
|
26
|
Liu JM, Zhu Y, Xu ZW, Ouyang WM, Wang JP, Liu XS, Cao YX, Li Q, Fang L, Zhuang R, Yang AG, Jin BQ. Dynamic changes of apoptosis-inducing ligands and Th1/Th2 like subpopulations in Hantaan virus-induced hemorrhagic fever with renal syndrome. Clin Immunol 2006; 119:245-51. [PMID: 16616877 DOI: 10.1016/j.clim.2006.02.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Revised: 01/21/2006] [Accepted: 02/28/2006] [Indexed: 01/10/2023]
Abstract
The expression of the apoptosis-inducing ligands, TNF-alpha, FasL and TRAIL on peripheral blood mononuclear cells (PBMC) and the levels of their soluble form (TNF-alpha, sFasL and sTRAIL) in plasma from 40 hemorrhagic fever with renal syndrome (HFRS) patients as well as 26 healthy blood donors were determined by flow cytometry (FCM) analysis and sandwich ELISA, respectively. The status of Th1, Th2, Tc1 and Tc2 subsets in PBMC was evaluated by intracellular cytokine staining and FCM. Compared to controls, the expression of membrane bound FasL and TRAIL was up-regulated on surface of PBMC isolated from the HFRS patients, particularly on CD8+ T lymphocytes. The levels of TNF-alpha, sFasL and sTRAIL in plasma from the HFRS patients in the acute phase increase 4.7-fold, 6.0-fold and 1.8-fold, respectively, over those from the healthy donors. The percentage of Th1, Tc1 and Tc2 subsets in PBMC from the patients also increased significantly compared with those from healthy donors. These results indicate that dynamic changes occurred in both the membrane bound and soluble forms of apoptosis-inducing ligands (FasL, TRAIL and TNF-alpha) and proportions of Th1 and CTL in HFRS patients increased. Both factors may play an important role in the etiology of Hantaan virus infection in humans.
Collapse
Affiliation(s)
- Jing-Mei Liu
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Ding X, Xu F, Chen H, Tesh RB, Xiao SY. Apoptosis of hepatocytes caused by Punta Toro virus (Bunyaviridae: Phlebovirus) and its implication for Phlebovirus pathogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 167:1043-9. [PMID: 16192639 PMCID: PMC1603669 DOI: 10.1016/s0002-9440(10)61193-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Experimental infection of hamsters with Punta Toro virus (PTV) produces a disease with clinical and pathological similarities to the severe human hemorrhagic fever caused by Rift Valley fever virus (RVFV), thus providing an animal model for RVFV pathogenesis. In this model, hepatocytic apoptosis is the main pathological component of liver injuries that are responsible for severe hemorrhagic manifestations. To further elucidate whether viral replication in hepatocytes directly causes apoptosis, we studied the morphological and biochemical changes of apoptosis in HepG2 cells at different time points after PTV infection. Cellular viability began to decrease 12 hours after infection compared with controls. Caspases 3/7 were activated significantly at 48 and 72 hours after infection, and phosphatidylserine translocation and DNA fragmentation were also detected at 48 and 72 hours. Cell cycle analysis by flow cytometry showed that infected HepG2 cells were arrested at G(0)/G(1) phase. Furthermore, virus titer increased with apoptosis progression, suggesting that viral replication is necessary for the apoptotic process. These results indicate that PTV infection alone, without a secondary inflammatory cellular reaction, induces hepatocytic apoptosis and suggest that future therapeutics for RVFV hemorrhagic disease might target inhibition of cellular apoptotic pathways during the acute infection.
Collapse
Affiliation(s)
- Xiaohua Ding
- Department of Pathology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0588, USA
| | | | | | | | | |
Collapse
|
28
|
Affiliation(s)
- Walter Muranyi
- Klinikum der Universität Heidelberg, Sektion Nephrologie, Heidelberg, Germany
| | | | | | | |
Collapse
|
29
|
Kallio-Kokko H, Uzcategui N, Vapalahti O, Vaheri A. Viral zoonoses in Europe. FEMS Microbiol Rev 2005; 29:1051-77. [PMID: 16024128 PMCID: PMC7110368 DOI: 10.1016/j.femsre.2005.04.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2004] [Revised: 04/11/2005] [Accepted: 04/19/2005] [Indexed: 12/19/2022] Open
Abstract
A number of new virus infections have emerged or re-emerged during the past 15 years. Some viruses are spreading to new areas along with climate and environmental changes. The majority of these infections are transmitted from animals to humans, and thus called zoonoses. Zoonotic viruses are, as compared to human-only viruses, much more difficult to eradicate. Infections by several of these viruses may lead to high mortality and also attract attention because they are potential bio-weapons. This review will focus on zoonotic virus infections occurring in Europe.
Collapse
Affiliation(s)
- Hannimari Kallio-Kokko
- Haartman Institute, Department of Virology, University of Helsinki, POB 21, 00014 Helsinki, Finland.
| | | | | | | |
Collapse
|
30
|
Hardestam J, Klingström J, Mattsson K, Lundkvist A. HFRS causing hantaviruses do not induce apoptosis in confluent Vero E6 and A-549 cells. J Med Virol 2005; 76:234-40. [PMID: 15834879 DOI: 10.1002/jmv.20347] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Hantaviruses are known to cause little or no cytopathic effect in vitro, but have been suggested to cause apoptosis. To determine whether different hantaviruses would induce apoptosis to varying degrees, confluent Vero E6 cells were infected with the hemorrhagic fever with renal syndrome (HFRS) causing viruses Hantaan, Dobrava, Saaremaa, and Puumala. However, no difference was found in the percentage of adherent cells, or of cells with condensed nuclei, between non-infected and virus-infected cells at 3, 6, 9, or 12 days after infection. Furthermore, no differences in the percentage of cells with inter-nucleosomal cleavage of DNA between uninfected and Hantaan infected cells could be detected using the TUNEL assay. Possibly, slightly more apoptotic cells, but never more than 5%, were detected after Hantaan infection of non-confluent cells as compared to the negative control. Earlier reported results that Tula hantavirus induces significant apoptosis on Vero E6 cells were also verified, suggesting that non-pathogenic hantaviruses might differ from HFRS-causing strains regarding induction of apoptosis. In conclusion, the results indicated that the HFRS-causing hantaviruses might induce a very low level of apoptosis in dividing cells, but not at all in confluent cells.
Collapse
Affiliation(s)
- Jonas Hardestam
- Swedish Institute for Infectious Disease Control, Solna, Sweden
| | | | | | | |
Collapse
|
31
|
Kaukinen P, Vaheri A, Plyusnin A. Hantavirus nucleocapsid protein: a multifunctional molecule with both housekeeping and ambassadorial duties. Arch Virol 2005; 150:1693-713. [PMID: 15931462 DOI: 10.1007/s00705-005-0555-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2005] [Accepted: 04/12/2005] [Indexed: 01/10/2023]
Abstract
In recent years important progress has been made studying the nucleocapsid (N) protein of hantaviruses. The N protein presents a good example of a multifunctional viral macromolecule. It is a major structural component of a virion that encapsidates viral RNA (vRNA). It also interacts with the virus polymerase (L protein) and one of the glycoproteins. On top of these "house keeping" duties, the N protein performs interactive "ambassadorial" functions interfering with important regulatory pathways in the infected cells.
Collapse
Affiliation(s)
- P Kaukinen
- Department of Virology, Haartman Institute, University of Helsinki, Finland
| | | | | |
Collapse
|
32
|
Li XD, Lankinen H, Putkuri N, Vapalahti O, Vaheri A. Tula hantavirus triggers pro-apoptotic signals of ER stress in Vero E6 cells. Virology 2005; 333:180-9. [PMID: 15708603 PMCID: PMC7173054 DOI: 10.1016/j.virol.2005.01.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2004] [Revised: 12/01/2004] [Accepted: 01/03/2005] [Indexed: 12/29/2022]
Abstract
Tula virus is a member of the Hantavirus genus of the family Bunyaviridae. Viruses of this family have an unusual pattern of intracellular maturation at the ER–Golgi compartment. We recently found that Tula virus, similar to several other hantaviruses, is able to induce apoptosis in cultured cells [Li, X.D., Kukkonen, S., Vapalahti, O., Plyusnin, A., Lankinen, H., Vaheri, A., 2004. Tula hantavirus infection of Vero E6 cells induces apoptosis involving caspase 8 activation. J. Gen. Virol. 85, 3261–3268.]. However, the cellular mechanisms remain to be clarified. In this study, we demonstrate that the progressive replication of Tula virus in Vero E6 cells initiates several death programs that are intimately associated with ER stress: (1) early activation of ER-resident caspase-12; (2) phosphorylation of Jun NH2-terminal kinase (JNK) and its downstream target transcriptional factor, c-jun; (3) induction of the pro-apoptotic transcriptional factor, growth arrest- and DNA damage-inducible gene 153, or C/EBP homologous protein (Gadd153/chop); and (4) changes in the ER-membrane protein BAP31 implying cross-talk with the mitochondrial apoptosis pathway. Furthermore, we confirmed that a sustained ER stress was induced marked by an increased expression of an ER chaperone Grp78/BiP. Taken together, we have identified involvement of ER stress-mediated death program in Tula virus-infected Vero E6 cells which provides a new approach to understand the mechanisms in hantavirus-induced apoptosis.
Collapse
|
33
|
Yan H, Xiao G, Zhang J, Hu Y, Yuan F, Cole DK, Zheng C, Gao GF. SARS coronavirus induces apoptosis in Vero E6 cells. J Med Virol 2004; 73:323-31. [PMID: 15170624 PMCID: PMC7166321 DOI: 10.1002/jmv.20094] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Severe acute respiratory syndrome (SARS) is an emerging infectious disease. Its etiological agent has been convincingly identified as a new member of family Coronaviridae (SARS‐CoV). It causes serious damage to the respiratory system yet the mechanism is not clear. Infection‐induced apoptosis or necrosis is suspected but no direct evidence for this yet exists. To date, Vero E6 cells are the only cell line that could be used to replicate the virus with obvious CPE (cytopathic effect) in vitro. It is known for some viruses (including members of family Coronaviridae) that CPE can be caused either by virus‐induced apoptosis (active death) or cell necrosis (passive death). In this study, we examined the apoptosis in the SARS‐CoV infected Vero E6 cells. Indeed, the results do show that the CPE was induced by apoptosis rather than necrosis, shown by typical DNA fragmentation, through the existence of apoptotic bodies and swollen mitochondria. This observation has some implications for the SARS‐CoV pathogenicity: SARS‐CoV does induce apoptosis in cell cultures and might have the same effect in vivo, responsible for the severe damage of the respiratory system. J. Med. Virol. 73:323–331, 2004. © 2004 Wiley‐Liss, Inc.
Collapse
Affiliation(s)
- Huimin Yan
- School of Life Sciences, Wuhan University, Wuhan, China
| | - Gengfu Xiao
- School of Life Sciences, Wuhan University, Wuhan, China
| | - Jiamin Zhang
- School of Life Sciences, Wuhan University, Wuhan, China
| | - Yuanyang Hu
- School of Life Sciences, Wuhan University, Wuhan, China
| | - Fang Yuan
- Nuffield Department of Clinical Medicine, John Radcliffe Hospital, Oxford University, Oxford, United Kingdom
| | - David K. Cole
- Nuffield Department of Clinical Medicine, John Radcliffe Hospital, Oxford University, Oxford, United Kingdom
| | - Congyi Zheng
- School of Life Sciences, Wuhan University, Wuhan, China
| | - George F. Gao
- Nuffield Department of Clinical Medicine, John Radcliffe Hospital, Oxford University, Oxford, United Kingdom
| |
Collapse
|
34
|
Li XD, Kukkonen S, Vapalahti O, Plyusnin A, Lankinen H, Vaheri A. Tula hantavirus infection of Vero E6 cells induces apoptosis involving caspase 8 activation. J Gen Virol 2004; 85:3261-3268. [PMID: 15483239 DOI: 10.1099/vir.0.80243-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Hantaviruses are known to cause two severe human diseases: haemorrhagic fever with renal syndrome and hantavirus pulmonary syndrome. The mechanisms of pathogenesis of these two diseases are progressively becoming understood. Recently, two hantaviruses, Hantaan and Prospect Hill were reported to cause programmed cell death of Vero E6 cells. This study shows that Tula hantavirus (TULV) infection efficiently triggers an apoptotic programme in infected Vero E6 cells, and that the replication of TULV is required for the activation of caspase 3 and the cleavage of poly (ADP-ribose) polymerase, two molecular hallmarks of apoptosis. The enforced treatment of infected Vero E6 cells with tumour necrosis factor alpha (TNF-alpha), but not interferon alpha (IFN-alpha), advanced the time course of apoptosis. Furthermore, caspase 8 was activated on day 4 post-infection, the same day when caspase 3 was activated. TNF receptor 1 was induced during a late stage of TULV infection. These data suggest that, unlike during influenza A virus infection, TNF-alpha, but not type I IFN-alpha/beta, may contribute significantly to apoptosis in a synergistic manner with TULV propagation. Interestingly, pretreatment with a broad-spectrum caspase inhibitor, z-VAD-fmk, efficiently inhibited apoptosis of TULV-infected Vero E6 cells. Taken together, these results suggest that TULV replication initiates a typical apoptotic programme involving caspase 8 activation.
Collapse
Affiliation(s)
- Xiao-Dong Li
- Department of Virology, Haartman Institute, POB 21, FIN-00014 University of Helsinki, Finland
| | - Sami Kukkonen
- Department of Virology, Haartman Institute, POB 21, FIN-00014 University of Helsinki, Finland
| | - Olli Vapalahti
- Department of Virology, Haartman Institute, POB 21, FIN-00014 University of Helsinki, Finland
| | - Alexander Plyusnin
- Department of Virology, Haartman Institute, POB 21, FIN-00014 University of Helsinki, Finland
| | - Hilkka Lankinen
- Department of Virology, Haartman Institute, POB 21, FIN-00014 University of Helsinki, Finland
| | - Antti Vaheri
- Department of Virology, Haartman Institute, POB 21, FIN-00014 University of Helsinki, Finland
| |
Collapse
|