1
|
Filhol T, Mac Kain A, Joffret ML, Jouvenet N, Caval V, Bessaud M. The use of sialic acids as attachment factors is a common feature of Enterovirus-D species. J Virol 2025:e0042925. [PMID: 40358210 DOI: 10.1128/jvi.00429-25] [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: 03/06/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Among the hundreds of enteroviruses (EVs) infecting humans, the members of the species EV-D (Enterovirus deconjuncti) display original traits. First, only five serotypes are known within this species, while other EV species have tens of serotypes each. Second, EV-Ds display a wide variety of tropisms: EV-D68s are respiratory viruses, EV-D70s have an ocular tropism, while EV-D94s, EV-D111s, and EV-D120s seem to be enteric viruses. Besides, while EV-D68s, EV-D70s, and EV-D94s have been detected in humans, EV-D120s were found exclusively in non-human primates, and the last virus type, EV-D111, was found in both. This and other observations have led to the hypothesis that EV-Ds could have a zoonotic origin. Previous studies have shown that EV-D68, EV-D70, and EV-D94 use sialic acids (Sias) as cellular attachment factors. We investigated the role of Sias in EV-D111 infection using sialidase treatments and loss-of-function experiments in human and simian cells. Assessing viral RNA yield by RT-qPCR analyses and infectious viral particle production by titration assays showed that the absence of Sias at the cell surface significantly slowed down EV-D111 infection kinetics without abolishing it. This suggests that Sia acts as an attachment factor. While EVs generally do not use Sias, EV-Ds seem to rely on them for optimal replication in cultured cells. Sia usage may therefore be an ancestral trait of this species. We also studied EV-B114, a simian enterovirus, and found that it does not use Sias. Our work provides new insight regarding an enterovirus that circulates in humans and exhibits unusual ecological traits.IMPORTANCEExcept for a few epidemics in the 1970s and 1980s, the impact of EV-Ds on human health remained modest until the 2010s. In 2014, EV-D68 was occasionally responsible for severe respiratory distress and fatal cases of muscular paralysis. EV-Ds have thus the ability to become pathogenic in humans, hence the importance of studying them. The recently discovered EV-D111, of which only a few isolates are available, has been detected in both human and simian samples, suggesting a potential zoonotic origin. We characterized the early steps of EV-D111 replication, with a focus on its ability to use Sias as attachment factors. We found that EV-D111, like other members of the EV-D species, but unlike most EVs, relies on Sia for optimal replication. Our work provides a better understanding of EV-D111 biology, which is essential to determine its tropism and its potential to emerge in humans.
Collapse
Affiliation(s)
- Typhaine Filhol
- Virus Sensing and Signaling Unit, Department of Virology, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
- Laboratoire associé au Centre national de référence entérovirus/paréchovirus, Institut Pasteur, Paris, France
| | - Alice Mac Kain
- Laboratoire associé au Centre national de référence entérovirus/paréchovirus, Institut Pasteur, Paris, France
| | - Marie-Line Joffret
- Virus Sensing and Signaling Unit, Department of Virology, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
- Laboratoire associé au Centre national de référence entérovirus/paréchovirus, Institut Pasteur, Paris, France
| | - Nolwenn Jouvenet
- Virus Sensing and Signaling Unit, Department of Virology, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
| | - Vincent Caval
- Virus Sensing and Signaling Unit, Department of Virology, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
| | - Maël Bessaud
- Virus Sensing and Signaling Unit, Department of Virology, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
- Laboratoire associé au Centre national de référence entérovirus/paréchovirus, Institut Pasteur, Paris, France
| |
Collapse
|
2
|
Shen S, Guo H, Li Y, Zhang L, Tang Y, Li H, Li X, Wang PH, Yu XF, Wei W. SARS-CoV-2 and oncolytic EV-D68-encoded proteases differentially regulate pyroptosis. J Virol 2024; 98:e0190923. [PMID: 38289118 PMCID: PMC10878271 DOI: 10.1128/jvi.01909-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/03/2024] [Indexed: 02/21/2024] Open
Abstract
Pyroptosis, a pro-inflammatory programmed cell death, has been implicated in the pathogenesis of coronavirus disease 2019 and other viral diseases. Gasdermin family proteins (GSDMs), including GSDMD and GSDME, are key regulators of pyroptotic cell death. However, the mechanisms by which virus infection modulates pyroptosis remain unclear. Here, we employed a mCherry-GSDMD fluorescent reporter assay to screen for viral proteins that impede the localization and function of GSDMD in living cells. Our data indicated that the main protease NSP5 of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) blocked GSDMD-mediated pyroptosis via cleaving residues Q29 and Q193 of GSDMD. While another SARS-CoV-2 protease, NSP3, cleaved GSDME at residue G370 but activated GSDME-mediated pyroptosis. Interestingly, respiratory enterovirus EV-D68-encoded proteases 3C and 2A also exhibit similar differential regulation on the functions of GSDMs by inactivating GSDMD but initiating GSDME-mediated pyroptosis. EV-D68 infection exerted oncolytic effects on human cancer cells by inducing pyroptotic cell death. Our findings provide insights into how respiratory viruses manipulate host cell pyroptosis and suggest potential targets for antiviral therapy as well as cancer treatment.IMPORTANCEPyroptosis plays a crucial role in the pathogenesis of coronavirus disease 2019, and comprehending its function may facilitate the development of novel therapeutic strategies. This study aims to explore how viral-encoded proteases modulate pyroptosis. We investigated the impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and respiratory enterovirus D68 (EV-D68) proteases on host cell pyroptosis. We found that SARS-CoV-2-encoded proteases NSP5 and NSP3 inactivate gasdermin D (GSDMD) but initiate gasdermin E (GSDME)-mediated pyroptosis, respectively. We also discovered that another respiratory virus EV-D68 encodes two distinct proteases 2A and 3C that selectively trigger GSDME-mediated pyroptosis while suppressing the function of GSDMD. Based on these findings, we further noted that EV-D68 infection triggers pyroptosis and produces oncolytic effects in human carcinoma cells. Our study provides new insights into the molecular mechanisms underlying virus-modulated pyroptosis and identifies potential targets for the development of antiviral and cancer therapeutics.
Collapse
Affiliation(s)
- Siyu Shen
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, China
| | - Haoran Guo
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, China
| | - Yan Li
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, China
| | - Lili Zhang
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, China
| | - Yubin Tang
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, China
| | - Huili Li
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, China
| | - Xiaohan Li
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, China
| | - Pei-Hui Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiao-Fang Yu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Wei
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, China
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Translational Medicine, First Hospital, Jilin University, Changchun, Jilin, China
| |
Collapse
|
3
|
Hooi YT, Balasubramaniam VRMT. In vitro and in vivo models for the study of EV-D68 infection. Pathology 2023; 55:907-916. [PMID: 37852802 DOI: 10.1016/j.pathol.2023.08.007] [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: 03/03/2023] [Revised: 06/03/2023] [Accepted: 08/14/2023] [Indexed: 10/20/2023]
Abstract
Enterovirus D68 (EV-D68) is one of hundreds of non-polio enteroviruses that typically cause cold-like respiratory illness. The first EV-D68 outbreak in the United States in 2014 aroused widespread concern among the public and health authorities. The infection was found to be associated with increased surveillance of acute flaccid myelitis, a neurological condition that causes limb paralysis in conjunction with spinal cord inflammation. In vitro studies utilising two-dimensional (2D) and three-dimensional (3D) culture systems have been employed to elucidate the pathogenic mechanism of EV-D68. Various animal models have also been developed to investigate viral tropism and distribution, pathogenesis, and immune responses during EV-D68 infection. EV-D68 infections have primarily been investigated in respiratory, intestinal and neural cell lines/tissues, as well as in small-size immunocompetent rodent models that were limited to a young age. Some studies have implemented strategies to overcome the barriers by using immunodeficient mice or virus adaptation. Although the existing models may not fully recapitulate both respiratory and neurological disease observed in human EV-D68 infection, they have been valuable for studying pathogenesis and evaluating potential vaccine or therapeutic candidates. In this review, we summarise the methodologies and findings from each experimental model and discuss their applications and limitations.
Collapse
Affiliation(s)
- Yuan Teng Hooi
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia.
| | - Vinod R M T Balasubramaniam
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia.
| |
Collapse
|
4
|
Pei J, Liu RL, Yang ZH, Du YX, Qian SS, Meng SL, Guo J, Zhang B, Shen S. Identification of Critical Amino Acids of Coxsackievirus A10 Associated with Cell Tropism and Viral RNA Release during Uncoating. Viruses 2023; 15:2114. [PMID: 37896891 PMCID: PMC10611408 DOI: 10.3390/v15102114] [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: 09/19/2023] [Revised: 10/07/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Coxsackievirus A10 (CV-A10) is a prevailing causative agent of hand-foot-mouth disease, necessitating the isolation and adaptation of appropriate strains in cells allowed for human vaccine development. In this study, amino acid sequences of CV-A10 strains with different cell tropism on RD and Vero cells were compared. Various amino acids on the structural and non-structural proteins related to cell tropism were identified. The reverse genetic systems of several CV-A10 strains with RD+/Vero- and RD+/Vero+ cell tropism were developed, and a set of CV-A10 recombinants were produced. The binding, entry, uncoating, and proliferation steps in the life cycle of these viruses were evaluated. P1 replacement of CV-A10 strains with different cell tropism revealed the pivotal role of the structural proteins in cell tropism. Further, seven amino acid substitutions in VP2 and VP1 were introduced to further investigate their roles played in cell tropism. These mutations cooperated in the growth of CV-A10 in Vero cells. Particularly, the valine to isoleucine mutation at the position VP1-236 (V1236I) was found to significantly restrict viral uncoating in Vero cells. Co-immunoprecipitation assays showed that the release of viral RNA from the KREMEN1 receptor-binding virions was restricted in r0195-V1236I compared with the parental strain r0195 (a RD+/Vero+ strain). Overall, this study highlights the dominant effect of structural proteins in CV-A10 adaption in Vero cells and the importance of V1236 in viral uncoating, providing a foundation for the mechanism study of CV-A10 cell tropism, and facilitating the development of vaccine candidates.
Collapse
Affiliation(s)
- Jie Pei
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China; (J.P.); (R.-L.L.); (Z.-H.Y.); (Y.-X.D.); (S.-S.Q.); (S.-L.M.); (J.G.)
| | - Rui-Lun Liu
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China; (J.P.); (R.-L.L.); (Z.-H.Y.); (Y.-X.D.); (S.-S.Q.); (S.-L.M.); (J.G.)
| | - Zhi-Hui Yang
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China; (J.P.); (R.-L.L.); (Z.-H.Y.); (Y.-X.D.); (S.-S.Q.); (S.-L.M.); (J.G.)
| | - Ya-Xin Du
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China; (J.P.); (R.-L.L.); (Z.-H.Y.); (Y.-X.D.); (S.-S.Q.); (S.-L.M.); (J.G.)
| | - Sha-Sha Qian
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China; (J.P.); (R.-L.L.); (Z.-H.Y.); (Y.-X.D.); (S.-S.Q.); (S.-L.M.); (J.G.)
| | - Sheng-Li Meng
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China; (J.P.); (R.-L.L.); (Z.-H.Y.); (Y.-X.D.); (S.-S.Q.); (S.-L.M.); (J.G.)
| | - Jing Guo
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China; (J.P.); (R.-L.L.); (Z.-H.Y.); (Y.-X.D.); (S.-S.Q.); (S.-L.M.); (J.G.)
| | - Bo Zhang
- Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China;
| | - Shuo Shen
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China; (J.P.); (R.-L.L.); (Z.-H.Y.); (Y.-X.D.); (S.-S.Q.); (S.-L.M.); (J.G.)
| |
Collapse
|
5
|
Filipe IC, Tee HK, Prados J, Piuz I, Constant S, Huang S, Tapparel C. Comparison of tissue tropism and host response to enteric and respiratory enteroviruses. PLoS Pathog 2022; 18:e1010632. [PMID: 35789345 PMCID: PMC9286751 DOI: 10.1371/journal.ppat.1010632] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 07/15/2022] [Accepted: 06/01/2022] [Indexed: 11/21/2022] Open
Abstract
Enteroviruses (EVs) are among the most prevalent viruses worldwide. They are characterized by a high genetic and phenotypic diversity, being able to cause a plethora of symptoms. EV-D68, a respiratory EV, and EV-D94, an enteric EV, represent an interesting paradigm of EV tropism heterogeneity. They belong to the same species, but display distinct phenotypic characteristics and in vivo tropism. Here, we used these two viruses as well as relevant 3D respiratory, intestinal and neural tissue culture models, to highlight key distinctive features of enteric and respiratory EVs. We emphasize the critical role of temperature in restricting EV-D68 tissue tropism. Using transcriptomic analysis, we underscore fundamental differences between intestinal and respiratory tissues, both in the steady-state and in response to infection. Intestinal tissues present higher cell proliferation rate and are more immunotolerant than respiratory tissues. Importantly, we highlight the different strategies applied by EV-D94 and EV-D68 towards the host antiviral response of intestinal and respiratory tissues. EV-D68 strongly activates antiviral pathways while EV-D94, on the contrary, barely induces any host defense mechanisms. In summary, our study provides an insightful characterization of the differential pathogenesis of EV-D68 and EV-D94 and the interplay with their main target tissues. Enteroviruses (EVs) are important human pathogens, associated with more than 20 clinical presentations. They replicate predominantly in the intestinal and/or respiratory mucosae. The respiratory EV-D68 can be considered an emerging virus because it caused an unprecedent outbreak in 2014, and contemporary isolates display increased virulence and novel neurotropic potential. The genetically related enteric EV-D94 is less common and its pathogenesis remains poorly defined, however, its infection has also been associated with neurological symptoms such as acute flaccid paralysis. To decipher the pathogenic mechanisms of these two viruses, we investigated their tropism and innate immunity induction in relevant human respiratory, intestinal and neural tissue culture models. Our results highlight the critical role of temperature in restricting EV-D68 tropism. Furthermore, using transcriptomic analysis, we identified key differences between respiratory and intestinal tissues, with the latter exhibiting higher cell proliferation and being more immunotolerant. More importantly, we could demonstrate the different strategies applied by EV-D94 and EV-D68 towards the host antiviral response, with EV-D68 strongly activating antiviral pathways and EV-D94, in contrast, inducing few host antiviral transcripts. This work identifies key differences in the pathogenesis of these representative respiratory and enteric EVs, which may contribute to the development of targeted antiviral therapies.
Collapse
Affiliation(s)
- Ines Cordeiro Filipe
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Han Kang Tee
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Julien Prados
- Bioinformatics Support Platform, University of Geneva, Geneva, Switzerland
| | - Isabelle Piuz
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | | | - Song Huang
- Epithelix SAS Geneva, Geneva, Switzerland
| | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
- * E-mail:
| |
Collapse
|
6
|
R.Swartz A, Shieh Y, Gulasarian A, Olson J, R.Rustandi R. Binding of Coxsackievirus A21 procapsids to immobilized glutathione depends on cell culture conditions during infection. Virology 2022; 573:167-175. [DOI: 10.1016/j.virol.2022.06.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/05/2023]
|
7
|
Si F, Ji T, Wang D, Zhang Y, Zhu S, Li J, Xu W, Yan D. Origin and evolution analysis and genetic characteristics of echovirus 9 in China. Virol J 2022; 19:98. [PMID: 35659318 PMCID: PMC9166342 DOI: 10.1186/s12985-022-01820-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 05/12/2022] [Indexed: 11/25/2022] Open
Abstract
Background Echovirus 9 (E9) is associated with a wide variety of diseases and medical conditions, and the clinical symptoms of sporadic cases caused by E9 often are severe. With a high global prevalence, E9 has caused multiple outbreaks worldwide. However, little is known about the genetic and geographic population dynamics of E9. Method A total of 131 VP1 gene sequences, including15 generated in this study and 116 obtained from GenBank, were used to coestimate time-resolved phylogenies to infer viral evolution and transmission in worldwide. Overlapping fragments representing whole genomes were amplified by reverse transcription polymerase chain reaction (RT-PCR) using specific primers. Then, we reported the genetic characteristics of fifteen E9 strains in the Chinese Mainland. Similarity plots and bootscanning analysis were used to determine recombination patterns of E9. Results The estimated mean evolutionary rate of global E9 VP1 gene was 4.278 × 10−3 substitutions per site per year (95% confidence interval [CI], 3.822 × 10−3/site/year to 4.710 × 10−3/site/year), and the common ancestor of E9 likely emerged around 1868 (95% CI, 1840 to 1892). The full-length genomic sequences of the fifteen E9 strains showed 76.9–79.6% nucleotide identity and 95.3–95.9% amino acid identity with E9 Barty strain. 11 of 15 E9 whole genome sequence present four recombination patterns, and E9 recombinants have extensive genetic exchanges in the 2C and P3 regions with other Enterovirus B (EV-B) circulated in China. Four of six E9 strains were temperature sensitive, and two were temperature resistant, and a comparative genomics analysis suggested that 411, 865 and 867 amino acid substitution in the P1 region was related to temperature sensitivity. Conclusion This study highlights a persistent transmission network of E9 in worldwide, provides valuable information regarding the molecular epidemiology of E9.
Collapse
Affiliation(s)
- Fenfen Si
- WHO WPRO Regional Polio Reference Laboratory and NHC Key Laboratory for Biosafety, NHC Key Laboratory for Medical Virology, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, 102206, People's Republic of China.,Beijing Fengtai District Center for Disease Control and Prevention, Beijing, 10071, People's Republic of China
| | - Tianjiao Ji
- WHO WPRO Regional Polio Reference Laboratory and NHC Key Laboratory for Biosafety, NHC Key Laboratory for Medical Virology, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, 102206, People's Republic of China
| | - Dongyan Wang
- WHO WPRO Regional Polio Reference Laboratory and NHC Key Laboratory for Biosafety, NHC Key Laboratory for Medical Virology, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, 102206, People's Republic of China
| | - Yong Zhang
- WHO WPRO Regional Polio Reference Laboratory and NHC Key Laboratory for Biosafety, NHC Key Laboratory for Medical Virology, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, 102206, People's Republic of China.,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, People's Republic of China
| | - Shuangli Zhu
- WHO WPRO Regional Polio Reference Laboratory and NHC Key Laboratory for Biosafety, NHC Key Laboratory for Medical Virology, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, 102206, People's Republic of China
| | - Junhan Li
- WHO WPRO Regional Polio Reference Laboratory and NHC Key Laboratory for Biosafety, NHC Key Laboratory for Medical Virology, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, 102206, People's Republic of China
| | - Wenbo Xu
- WHO WPRO Regional Polio Reference Laboratory and NHC Key Laboratory for Biosafety, NHC Key Laboratory for Medical Virology, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, 102206, People's Republic of China
| | - Dongmei Yan
- WHO WPRO Regional Polio Reference Laboratory and NHC Key Laboratory for Biosafety, NHC Key Laboratory for Medical Virology, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, 102206, People's Republic of China.
| |
Collapse
|
8
|
Filipe IC, Guedes MS, Zdobnov EM, Tapparel C. Enterovirus D: A Small but Versatile Species. Microorganisms 2021; 9:1758. [PMID: 34442837 PMCID: PMC8400195 DOI: 10.3390/microorganisms9081758] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 12/13/2022] Open
Abstract
Enteroviruses (EVs) from the D species are the causative agents of a diverse range of infectious diseases in spite of comprising only five known members. This small clade has a diverse host range and tissue tropism. It contains types infecting non-human primates and/or humans, and for the latter, they preferentially infect the eye, respiratory tract, gastrointestinal tract, and nervous system. Although several Enterovirus D members, in particular EV-D68, have been associated with neurological complications, including acute myelitis, there is currently no effective treatment or vaccine against any of them. This review highlights the peculiarities of this viral species, focusing on genome organization, functional elements, receptor usage, and pathogenesis.
Collapse
Affiliation(s)
- Ines Cordeiro Filipe
- Department of Microbiology and Molecular Medicine, University of Geneva, 1206 Geneva, Switzerland;
| | - Mariana Soares Guedes
- Department of Microbiology and Molecular Medicine, University of Geneva, 1206 Geneva, Switzerland;
| | - Evgeny M. Zdobnov
- Department of Genetic Medicine and Development, Switzerland and Swiss Institute of Bioinformatics, University of Geneva, 1206 Geneva, Switzerland;
| | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, University of Geneva, 1206 Geneva, Switzerland;
| |
Collapse
|
9
|
Mathez G, Cagno V. Viruses Like Sugars: How to Assess Glycan Involvement in Viral Attachment. Microorganisms 2021; 9:1238. [PMID: 34200288 PMCID: PMC8230229 DOI: 10.3390/microorganisms9061238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/01/2021] [Accepted: 06/04/2021] [Indexed: 12/12/2022] Open
Abstract
The first step of viral infection requires interaction with the host cell. Before finding the specific receptor that triggers entry, the majority of viruses interact with the glycocalyx. Identifying the carbohydrates that are specifically recognized by different viruses is important both for assessing the cellular tropism and for identifying new antiviral targets. Advances in the tools available for studying glycan-protein interactions have made it possible to identify them more rapidly; however, it is important to recognize the limitations of these methods in order to draw relevant conclusions. Here, we review different techniques: genetic screening, glycan arrays, enzymatic and pharmacological approaches, and surface plasmon resonance. We then detail the glycan interactions of enterovirus D68 and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), highlighting the aspects that need further clarification.
Collapse
Affiliation(s)
| | - Valeria Cagno
- Institute of Microbiology, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland;
| |
Collapse
|
10
|
McCall LI. Quo vadis? Central Rules of Pathogen and Disease Tropism. Front Cell Infect Microbiol 2021; 11:640987. [PMID: 33718287 PMCID: PMC7947345 DOI: 10.3389/fcimb.2021.640987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
Understanding why certain people get sick and die while others recover or never become ill is a fundamental question in biomedical research. A key determinant of this process is pathogen and disease tropism: the locations that become infected (pathogen tropism), and the locations that become damaged (disease tropism). Identifying the factors that regulate tropism is essential to understand disease processes, but also to drive the development of new interventions. This review intersects research from across infectious diseases to define the central mediators of disease and pathogen tropism. This review also highlights methods of study, and translational implications. Overall, tropism is a central but under-appreciated aspect of infection pathogenesis which should be at the forefront when considering the development of new methods of intervention.
Collapse
Affiliation(s)
- Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, United States
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, United States
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, United States
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, United States
| |
Collapse
|
11
|
Elrick MJ, Pekosz A, Duggal P. Enterovirus D68 molecular and cellular biology and pathogenesis. J Biol Chem 2021; 296:100317. [PMID: 33484714 PMCID: PMC7949111 DOI: 10.1016/j.jbc.2021.100317] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/13/2022] Open
Abstract
In recent years, enterovirus D68 (EV-D68) has advanced from a rarely detected respiratory virus to a widespread pathogen responsible for increasing rates of severe respiratory illness and acute flaccid myelitis (AFM) in children worldwide. In this review, we discuss the accumulating data on the molecular features of EV-D68 and place these into the context of enterovirus biology in general. We highlight similarities and differences with other enteroviruses and genetic divergence from own historical prototype strains of EV-D68. These include changes in capsid antigens, host cell receptor usage, and viral RNA metabolism collectively leading to increased virulence. Furthermore, we discuss the impact of EV-D68 infection on the biology of its host cells, and how these changes are hypothesized to contribute to motor neuron toxicity in AFM. We highlight areas in need of further research, including the identification of its primary receptor and an understanding of the pathogenic cascade leading to motor neuron injury in AFM. Finally, we discuss the epidemiology of the EV-D68 and potential therapeutic approaches.
Collapse
Affiliation(s)
- Matthew J Elrick
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.
| | - Andrew Pekosz
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Priya Duggal
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
12
|
Interferon-Dependent and Respiratory Virus-Specific Interference in Dual Infections of Airway Epithelia. Sci Rep 2020; 10:10246. [PMID: 32581261 PMCID: PMC7314816 DOI: 10.1038/s41598-020-66748-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/26/2020] [Indexed: 02/08/2023] Open
Abstract
Many respiratory viruses cocirculate in the population and multiple infections are commonly reported. The clinical impact of coinfection is unclear and may vary depending on the viral couples involved. Using three-dimensional reconstituted human airway epithelia and clinical viral strains, we investigated the interaction between influenza virus (Flu), respiratory syncytial virus (RSV) and rhinovirus (RV). We showed that Flu and RSV interfere with RV replication, whereas RV does not interfere with either of these viruses. We then experimentally demonstrated that, when present, the interference is not related to a block of viral entry but rather to type I and type III interferon (IFN), the front-line antiviral defense of the respiratory mucosa. Consistent with this observation, we highlighted the differential sensitivity of each virus to IFNs, with RV being the only virus significantly inhibited by IFN-λ and the most sensitive to IFN-α. Finally, as type III IFN is of therapeutic interest due to its low proinflammatory profile, we also assessed and confirmed an inhibitory effect of IFN-λ in the context of persistent RV infections. The present work provides mechanistic clues concerning innate immunity involvement during respiratory virus interactions and confirms that IFN-λ is a promising candidate in the treatment of RV infections.
Collapse
|
13
|
Majer A, McGreevy A, Booth TF. Molecular Pathogenicity of Enteroviruses Causing Neurological Disease. Front Microbiol 2020; 11:540. [PMID: 32328043 PMCID: PMC7161091 DOI: 10.3389/fmicb.2020.00540] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 03/12/2020] [Indexed: 12/12/2022] Open
Abstract
Enteroviruses are single-stranded positive-sense RNA viruses that primarily cause self-limiting gastrointestinal or respiratory illness. In some cases, these viruses can invade the central nervous system, causing life-threatening neurological diseases including encephalitis, meningitis and acute flaccid paralysis (AFP). As we near the global eradication of poliovirus, formerly the major cause of AFP, the number of AFP cases have not diminished implying a non-poliovirus etiology. As the number of enteroviruses linked with neurological disease is expanding, of which many had previously little clinical significance, these viruses are becoming increasingly important to public health. Our current understanding of these non-polio enteroviruses is limited, especially with regards to their neurovirulence. Elucidating the molecular pathogenesis of these viruses is paramount for the development of effective therapeutic strategies. This review summarizes the clinical diseases associated with neurotropic enteroviruses and discusses recent advances in the understanding of viral invasion of the central nervous system, cell tropism and molecular pathogenesis as it correlates with host responses.
Collapse
Affiliation(s)
- Anna Majer
- Viral Diseases Division, National Microbiology Laboratory, Winnipeg, MB, Canada
| | - Alan McGreevy
- Viral Diseases Division, National Microbiology Laboratory, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,Department of Biology, University of Winnipeg, Winnipeg, MB, Canada
| | - Timothy F Booth
- Viral Diseases Division, National Microbiology Laboratory, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
14
|
Jones ST, Cagno V, Janeček M, Ortiz D, Gasilova N, Piret J, Gasbarri M, Constant DA, Han Y, Vuković L, Král P, Kaiser L, Huang S, Constant S, Kirkegaard K, Boivin G, Stellacci F, Tapparel C. Modified cyclodextrins as broad-spectrum antivirals. SCIENCE ADVANCES 2020; 6:eaax9318. [PMID: 32064341 PMCID: PMC6989148 DOI: 10.1126/sciadv.aax9318] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 11/22/2019] [Indexed: 05/18/2023]
Abstract
Viral infections kill millions of people and new antivirals are needed. Nontoxic drugs that irreversibly inhibit viruses (virucidal) are postulated to be ideal. Unfortunately, all virucidal molecules described to date are cytotoxic. We recently developed nontoxic, broad-spectrum virucidal gold nanoparticles. Here, we develop further the concept and describe cyclodextrins, modified with mercaptoundecane sulfonic acids, to mimic heparan sulfates and to provide the key nontoxic virucidal action. We show that the resulting macromolecules are broad-spectrum, biocompatible, and virucidal at micromolar concentrations in vitro against many viruses [including herpes simplex virus (HSV), respiratory syncytial virus (RSV), dengue virus, and Zika virus]. They are effective ex vivo against both laboratory and clinical strains of RSV and HSV-2 in respiratory and vaginal tissue culture models, respectively. Additionally, they are effective when administrated in mice before intravaginal HSV-2 inoculation. Lastly, they pass a mutation resistance test that the currently available anti-HSV drug (acyclovir) fails.
Collapse
Affiliation(s)
- Samuel T. Jones
- Institute of Materials, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
- Department of Materials, University of Manchester, Manchester M13 9PL, UK
| | - Valeria Cagno
- Institute of Materials, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva 1211, Switzerland
| | - Matej Janeček
- Institute of Materials, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Daniel Ortiz
- Institut des Sciences et Ingénierie Chimiques, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Natalia Gasilova
- Institut des Sciences et Ingénierie Chimiques, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Jocelyne Piret
- CHU of Québec-Laval University, Québec City, Québec, Canada
| | - Matteo Gasbarri
- Institute of Materials, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - David A. Constant
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yanxiao Han
- Department of Chemistry, University of Illinois, Chicago, IL 60607, USA
| | - Lela Vuković
- Department of Chemistry and Biochemistry, University of Texas at El Paso, El Paso, TX 79966, USA
| | - Petr Král
- Department of Chemistry, University of Illinois, Chicago, IL 60607, USA
- Department of Physics, and Department of Biopharmaceutical Sciences, University of Illinois, Chicago, IL 60612, USA
| | - Laurent Kaiser
- Division of Infectious diseases, University Hospitals of Geneva, Geneva, Switzerland
| | | | | | - Karla Kirkegaard
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Guy Boivin
- CHU of Québec-Laval University, Québec City, Québec, Canada
| | - Francesco Stellacci
- Institute of Materials, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
- Department of Bionengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
- Corresponding author. (C.T.); (F.S.)
| | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva 1211, Switzerland
- Corresponding author. (C.T.); (F.S.)
| |
Collapse
|
15
|
Fecal Components Modulate Human Astrovirus Infectivity in Cells and Reconstituted Intestinal Tissues. mSphere 2019; 4:4/6/e00568-19. [PMID: 31852804 PMCID: PMC6920511 DOI: 10.1128/msphere.00568-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
To ensure transmission, enteric viruses must maintain their infectivity during the various environmental challenges that they face in transit within and between hosts. Increased knowledge of the factors affecting enteric virus survival may help to control their transmission. This study reveals that specific fecal bacterial components preserve classic human astrovirus infectivity by stabilizing viral particles. However, the outcomes of stool-virus interactions are very variable, ranging from protection to a reduction of viral infectivity, depending on the viral genotype and the individual from whom the stool has been collected. We show that the transmissibility of enteric viruses is dependent on the intestinal contents of the infected individual and highlight the complex multiple interactions that could explain the stochastic nature of enteric virus transmission in humans. Human astroviruses (HAstV) are among the most common causative agents of viral gastroenteritis, especially in children, and extraintestinal manifestations have also been described. These viruses are transmitted by the fecal-oral route, implying that stool composition and the gut microbiota may impact their ability to remain infectious. For some enteric viruses, individual bacterial envelope components and other polysaccharide-containing molecules, which are abundant in stools, have been shown to enhance capsid stability. However, the role of the complex stool environment and, most importantly, the role of interindividual differences have been poorly studied. We used HAstV as a model to investigate how the stool environment in itself, its interindividual variability, and some specific stool components could affect HAstV stability and infectivity. Using two different HAstV genotypes, we found that stools as a whole modulate astrovirus infectivity not only in an individual-dependent manner but also in a manner that depends on the viral genotype. A virus-protective effect was observed after incubation with various Gram-positive and Gram-negative bacteria as well as with bacterial components, such as lipopolysaccharide and peptidoglycan. These results were further confirmed in human intestinal tissues, a more physiologically relevant system. Astrovirus infectivity was also preserved by mucin, a major component of intestinal mucus. We further confirmed that these components stabilize the viral capsid. These results show that although HAstV benefits from the stabilizing effect of fecal components, the complexity and variability of the stool composition and the multiple potential interactions may explain the interindividual differences in viral transmission observed in real life. IMPORTANCE To ensure transmission, enteric viruses must maintain their infectivity during the various environmental challenges that they face in transit within and between hosts. Increased knowledge of the factors affecting enteric virus survival may help to control their transmission. This study reveals that specific fecal bacterial components preserve classic human astrovirus infectivity by stabilizing viral particles. However, the outcomes of stool-virus interactions are very variable, ranging from protection to a reduction of viral infectivity, depending on the viral genotype and the individual from whom the stool has been collected. We show that the transmissibility of enteric viruses is dependent on the intestinal contents of the infected individual and highlight the complex multiple interactions that could explain the stochastic nature of enteric virus transmission in humans.
Collapse
|
16
|
Ma C, Hu Y, Zhang J, Musharrafieh R, Wang J. A Novel Capsid Binding Inhibitor Displays Potent Antiviral Activity against Enterovirus D68. ACS Infect Dis 2019; 5:1952-1962. [PMID: 31532189 DOI: 10.1021/acsinfecdis.9b00284] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Enterovirus D68 (EV-D68) is a respiratory viral pathogen that primarily infects children under the age of 8. Although EV-D68 infection typically leads to moderate to severe respiratory illnesses, recent years have seen increasing cases of EV-D68 triggered neurological complications such as acute flaccid myelitis (AFM). There is currently no vaccine or antiviral available for EV-D68; we therefore aimed to develop potent and specific small molecule antivirals against EV-D68. In this study, we report our discovery of a viral capsid inhibitor R856932 that inhibits multiple contemporary EV-D68 strains with single-digit to submicromolar efficacy. Mechanistic studies have shown that the tetrazole compound R856932 binds to the hydrophobic pocket of viral capsid protein VP1, thereby preventing viral uncoating and release of viral genome in the infected cells. The mechanism of action of R856932 was confirmed by time-of-addition, Western blot, RT-qPCR, viral heat inactivation, serial viral passage, and reverse genetics experiments. A single mutation located at VP1, A129V, confers resistance against R856932. However, a recombination virus encoding VP1-A129V appeared to have compromised fitness of replication compared to the wild-type EV-D68 virus as shown by the competition growth assay. Overall, the hit compound identified in this study, R856932, represents a promising starting point with a confirmed mechanism of action that can be further developed into EV-D68 antivirals.
Collapse
Affiliation(s)
- Chunlong Ma
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1657 East Helen Street, Tucson, Arizona 85721, United States
| | - Yanmei Hu
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1657 East Helen Street, Tucson, Arizona 85721, United States
| | - Jiantao Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1657 East Helen Street, Tucson, Arizona 85721, United States
| | - Rami Musharrafieh
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1657 East Helen Street, Tucson, Arizona 85721, United States
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1657 East Helen Street, Tucson, Arizona 85721, United States
| |
Collapse
|
17
|
Neurotropism of Enterovirus D68 Isolates Is Independent of Sialic Acid and Is Not a Recently Acquired Phenotype. mBio 2019; 10:mBio.02370-19. [PMID: 31641090 PMCID: PMC6805996 DOI: 10.1128/mbio.02370-19] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Since 2014, numerous outbreaks of childhood infections with enterovirus D68 (EV-D68) have occurred worldwide. Most infections are associated with flu-like symptoms, but paralysis may develop in young children. It has been suggested that infection only with recent viral isolates can cause paralysis. To address the hypothesis that EV-D68 has recently acquired neurotropism, murine organotypic brain slice cultures, induced human motor neurons and astrocytes, and mice lacking the alpha/beta interferon receptor were infected with multiple virus isolates. All EV-D68 isolates, from 1962 to the present, can infect neural cells, astrocytes, and neurons. Furthermore, our results show that sialic acid binding does not play a role in EV-D68 neuropathogenesis. The study of EV-D68 infection in organotypic brain slice cultures, induced motor neurons, and astrocytes will allow for the elucidation of the mechanism by which the virus infection causes disease. Acute flaccid myelitis (AFM) is a rare but serious illness of the nervous system, specifically affecting the gray matter of the spinal cord, motor-controlling regions of the brain, and cranial nerves. Most cases of AFM are pathogen associated, typically with poliovirus and enterovirus infections, and occur in children under the age of 6 years. Enterovirus D68 (EV-D68) was first isolated from children with pneumonia in 1962, but an association with AFM was not observed until the 2014 outbreak. Organotypic mouse brain slice cultures generated from postnatal day 1 to 10 mice and adult ifnar knockout mice were used to determine if neurotropism of EV-D68 is shared among virus isolates. All isolates replicated in organotypic mouse brain slice cultures, and three isolates replicated in primary murine astrocyte cultures. All four EV-D68 isolates examined caused paralysis and death in adult ifnar knockout mice. In contrast, no viral disease was observed after intracranial inoculation of wild-type mice. Six of the seven EV-D68 isolates, including two from 1962 and four from the 2014 outbreak, replicated in induced human neurons, and all of the isolates replicated in induced human astrocytes. Furthermore, a putative viral receptor, sialic acid, is not required for neurotropism of EV-D68, as viruses replicated within neurons and astrocytes independent of binding to sialic acid. These observations demonstrate that EV-D68 is neurotropic independent of its genetic lineage and can infect both neurons and astrocytes and that neurotropism is not a recently acquired characteristic as has been suggested. Furthermore, the results show that in mice the innate immune response is critical for restricting EV-D68 disease.
Collapse
|
18
|
Sadeuh-Mba SA, Joffret ML, Mazitchi A, Endegue-Zanga MC, Njouom R, Delpeyroux F, Gouandjika-Vasilache I, Bessaud M. Genetic and phenotypic characterization of recently discovered enterovirus D type 111. PLoS Negl Trop Dis 2019; 13:e0007797. [PMID: 31622358 PMCID: PMC6818792 DOI: 10.1371/journal.pntd.0007797] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 10/29/2019] [Accepted: 09/18/2019] [Indexed: 01/08/2023] Open
Abstract
Members of the species Enterovirus D (EV-D) remain poorly studied. The two first EV-D types (EV-D68 and EV-D70) have regularly caused outbreaks in humans since their discovery five decades ago but have been neglected until the recent occurrence of severe respiratory diseases due to EV-D68. The three other known EV-D types (EV-D94, EV-D111 and EV-D120) were discovered in the 2000s-2010s in Africa and have never been observed elsewhere. One strain of EV-D111 and all known EV-D120s were detected in stool samples of wild non-human primates, suggesting that these viruses could be zoonotic viruses. To date, EV-D111s are only known through partial genetic sequences of the few strains that have been identified so far. In an attempt to bring new pieces to the puzzle, we genetically characterized four EV-D111 strains (among the seven that have been reported until now). We observed that the EV-D111 strains from human samples and the unique simian EV-D111 strain were not phylogenetically distinct, thus suggesting a recent zoonotic transmission. We also discovered evidences of probable intertypic genetic recombination events between EV-D111s and EV-D94s. As recombination can only happen in co-infected cells, this suggests that EV-D94s and EV-D111s share common replication sites in the infected hosts. These sites could be located in the gut since the phenotypic analysis we performed showed that, contrary to EV-D68s and like EV-D94s, EV-D111s are resistant to acid pHs. We also found that EV-D111s induce strong cytopathic effects on L20B cells, a cell line routinely used to specifically detect polioviruses. An active circulation of EV-D111s among humans could then induce a high number of false-positive detection of polioviruses, which could be particularly problematic in Central Africa, where EV-D111 circulates and which is a key region for poliovirus eradication.
Collapse
Affiliation(s)
| | - Marie-Line Joffret
- Institut Pasteur—Unité de biologie des virus entériques—Paris, France
- WHO Collaborating Centre for Enteroviruses and Viral Vaccines—Paris, France
| | - Arthur Mazitchi
- Enteric Viruses and Measles Laboratory—Institut Pasteur de Bangui—Bangui, Central African Republic
| | | | - Richard Njouom
- Virology Service—Centre Pasteur of Cameroon–Yaounde, Cameroon
| | - Francis Delpeyroux
- Institut Pasteur—Unité de biologie des virus entériques—Paris, France
- WHO Collaborating Centre for Enteroviruses and Viral Vaccines—Paris, France
| | | | - Maël Bessaud
- Institut Pasteur—Unité de biologie des virus entériques—Paris, France
- WHO Collaborating Centre for Enteroviruses and Viral Vaccines—Paris, France
| |
Collapse
|
19
|
Slow Infection due to Lowering the Amount of Intact versus Empty Particles Is a Characteristic Feature of Coxsackievirus B5 Dictated by the Structural Proteins. J Virol 2019; 93:JVI.01130-19. [PMID: 31375587 DOI: 10.1128/jvi.01130-19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 07/18/2019] [Indexed: 12/25/2022] Open
Abstract
Enterovirus B species typically cause a rapid cytolytic infection leading to efficient release of progeny viruses. However, they are also capable of persistent infections in tissues, which are suggested to contribute to severe chronic states such as myocardial inflammation and type 1 diabetes. In order to understand the factors contributing to differential infection strategies, we constructed a chimera by combining the capsid proteins from fast-cytolysis-causing echovirus 1 (EV1) with nonstructural proteins from coxsackievirus B5 (CVB5), which shows persistent infection in RD cells. The results showed that the chimera behaved similarly to parental EV1, leading to efficient cytolysis in both permissive A549 and semipermissive RD cells. In contrast to EV1 and the chimera, CVB5 replicated slowly in permissive cells and showed persistent infection in semipermissive cells. However, there was no difference in the efficiency of uptake of CVB5 in A549 or RD cells in comparison to the chimera or EV1. CVB5 batches constantly contained significant amounts of empty capsids, also in comparison to CVB5's close relative CVB3. During successive passaging of batches containing only intact CVB5, increasing amounts of empty and decreasing amounts of infective capsids were produced. Our results demonstrate that the increase in the amount of empty particles and the lowering of the amount of infective particles are dictated by the CVB5 structural proteins, leading to slowing down of the infection between passages. Furthermore, the key factor for persistent infection is the small amount of infective particles produced, not the high number of empty particles that accumulate.IMPORTANCE Enteroviruses cause several severe diseases, with lytic infections that lead to rapid cell death but also persistent infections that are more silent and lead to chronic states of infection. Our study compared a cytolytic echovirus 1 infection to persistent coxsackievirus B5 infection by making a chimera with the structural proteins of echovirus 1 and the nonstructural proteins of coxsackievirus B5. Coxsackievirus B5 infection was found to lead to the production of a high number of empty viruses (empty capsids) that do not contain genetic material and are unable to continue the infection. Coinciding with the high number of empty capsids, the amount of infective virions decreased. This characteristic property was not observed in the constructed chimera virus, suggesting that structural proteins are in charge of these phenomena. These results shed light on the mechanisms that may cause persistent infections. Understanding events leading to efficient or inefficient infections is essential in understanding virus-caused pathologies.
Collapse
|
20
|
Hixon AM, Frost J, Rudy MJ, Messacar K, Clarke P, Tyler KL. Understanding Enterovirus D68-Induced Neurologic Disease: A Basic Science Review. Viruses 2019; 11:E821. [PMID: 31487952 PMCID: PMC6783995 DOI: 10.3390/v11090821] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 12/28/2022] Open
Abstract
In 2014, the United States (US) experienced an unprecedented epidemic of enterovirus D68 (EV-D68)-induced respiratory disease that was temporally associated with the emergence of acute flaccid myelitis (AFM), a paralytic disease occurring predominantly in children, that has a striking resemblance to poliomyelitis. Although a definitive causal link between EV-D68 infection and AFM has not been unequivocally established, rapidly accumulating clinical, immunological, and epidemiological evidence points to EV-D68 as the major causative agent of recent seasonal childhood AFM outbreaks in the US. This review summarizes evidence, gained from in vivo and in vitro models of EV-D68-induced disease, which demonstrates that contemporary EV-D68 strains isolated during and since the 2014 outbreak differ from historical EV-D68 in several factors influencing neurovirulence, including their genomic sequence, their receptor utilization, their ability to infect neurons, and their neuropathogenicity in mice. These findings provide biological plausibility that EV-D68 is a causal agent of AFM and provide important experimental models for studies of pathogenesis and treatment that are likely to be difficult or impossible in humans.
Collapse
Affiliation(s)
- Alison M Hixon
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Joshua Frost
- Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Michael J Rudy
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Kevin Messacar
- Hospital Medicine and Pediatric Infectious Disease Sections, Department of Pediatrics, University of Colorado, Aurora, CO 80045, USA.
- Children's Hospital Colorado, Aurora, CO 80045, USA.
| | - Penny Clarke
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | - Kenneth L Tyler
- Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Division of Infectious Disease, Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Neurology Service, Rocky Mountain VA Medical Center, Aurora, CO 80045, USA
| |
Collapse
|
21
|
Wen X, Sun D, Guo J, Elgner F, Wang M, Hildt E, Cheng A. Multifunctionality of structural proteins in the enterovirus life cycle. Future Microbiol 2019; 14:1147-1157. [PMID: 31368347 DOI: 10.2217/fmb-2019-0127] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Members of the genus Enterovirus have a significant effect on human health, especially in infants and children. Since the viral genome has limited coding capacity, Enteroviruses subvert a range of cellular processes for viral infection via the interaction of viral proteins and numerous cellular factors. Intriguingly, the capsid-receptor interaction plays a crucial role in viral entry and has significant implications in viral pathogenesis. Moreover, interactions between structural proteins and host factors occur directly or indirectly in multiple steps of viral replication. In this review, we focus on the current understanding of the multifunctionality of structural proteins in the viral life cycle, which may constitute valuable targets for antiviral and therapeutic interventions.
Collapse
Affiliation(s)
- Xingjian Wen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China.,Paul-Ehrlich-Institut, Department of Virology, Langen, Germany
| | - Di Sun
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Jinlong Guo
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Fabian Elgner
- Paul-Ehrlich-Institut, Department of Virology, Langen, Germany
| | - Mingshu Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Eberhard Hildt
- Paul-Ehrlich-Institut, Department of Virology, Langen, Germany
| | - Anchun Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| |
Collapse
|
22
|
Sun J, Hu XY, Yu XF. Current Understanding of Human Enterovirus D68. Viruses 2019; 11:v11060490. [PMID: 31146373 PMCID: PMC6631698 DOI: 10.3390/v11060490] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 12/20/2022] Open
Abstract
Human enterovirus D68 (EV-D68), a member of the species Enterovirus D of the Picornaviridae family, was first isolated in 1962 in the United States. EV-D68 infection was only infrequently reported until an outbreak occurred in 2014 in the US; since then, it has continued to increase worldwide. EV-D68 infection leads to severe respiratory illness and has recently been reported to be linked to the development of the neurogenic disease known as acute flaccid myelitis (AFM), mostly in children, seriously endangering public health. Hitherto, treatment options for EV-D68 infections were limited to supportive care, and as yet there are no approved, specific antiviral drugs or vaccines. Research on EV-D68 has mainly focused on its epidemiology, and its virologic characteristics and pathogenesis still need to be further explored. Here, we provide an overview of current research on EV-D68, including the genotypes and genetic characteristics of recent epidemics, the mechanism of infection and virus-host interactions, and its relationship to acute flaccid myelitis (AFM), in order to broaden our understanding of the biological features of EV-D68 and provide a basis for the development of effective antiviral agents.
Collapse
Affiliation(s)
- Jing Sun
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China.
| | - Xiao-Yi Hu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China.
| | - Xiao-Fang Yu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China.
| |
Collapse
|
23
|
Growth of Zika virus in human reconstituted respiratory, intestinal, vaginal and neural tissues. Clin Microbiol Infect 2019; 25:1042.e1-1042.e4. [PMID: 31075547 DOI: 10.1016/j.cmi.2019.04.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/05/2019] [Accepted: 04/16/2019] [Indexed: 11/20/2022]
Abstract
OBJECTIVES Zika virus (ZIKV) is mostly mosquito borne but it can also be transmitted via the sexual route and persists in semen for a prolonged time. Moreover, viral RNA has been detected in breast milk, saliva, lacrimal fluids and urine, suggesting other possible transmission routes. The aim of our research is to better define ZIKV tropism. METHODS We investigated the tropism of Asian and African strains of ZIKV using human-derived neural, vaginal, intestinal and respiratory tissues. RESULTS Asian and African strains of ZIKV were able to grow in all tissues tested, although with different efficiency (7.3 log RNA copies released apically in vaginal tissues versus 9.8 log RNA copies released in intestinal tissues), without the need for major adaptation. CONCLUSIONS Our results underline that ZIKV tropism may be broader than expected in humans and stress the need to better explore all possible virus-shedding sites and transmission routes.
Collapse
|
24
|
Adeyemi OO, Sherry L, Ward JC, Pierce DM, Herod MR, Rowlands DJ, Stonehouse NJ. Involvement of a Nonstructural Protein in Poliovirus Capsid Assembly. J Virol 2019; 93:e01447-18. [PMID: 30541849 PMCID: PMC6384072 DOI: 10.1128/jvi.01447-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 12/03/2018] [Indexed: 12/13/2022] Open
Abstract
Virus capsid proteins must perform a number of roles. These include self-assembly and maintaining stability under challenging environmental conditions, while retaining the conformational flexibility necessary to uncoat and deliver the viral genome into a host cell. Fulfilling these roles could place conflicting constraints on the innate abilities encoded within the protein sequences. In a previous study, we identified a number of mutations within the capsid-coding sequence of poliovirus (PV) that were established in the population during selection for greater thermostability by sequential treatment at progressively higher temperatures. Two mutations in the VP1 protein acquired at an early stage were maintained throughout this selection procedure. One of these mutations prevented virion assembly when introduced into a wild-type (wt) infectious clone. Here we show, by sequencing beyond the capsid-coding region of the heat-selected virions, that two mutations had arisen within the coding region of the 2A protease. Both mutations were maintained throughout the selection process. Introduction of these mutations into a wt infectious clone by site-directed mutagenesis considerably reduced replication. However, they permitted a low level of assembly of infectious virions containing the otherwise lethal mutation in VP1. The 2Apro mutations were further shown to slow the kinetics of viral polyprotein processing, and we suggest that this delay improves the correct folding of the mutant capsid precursor protein to permit virion assembly.IMPORTANCE RNA viruses, including poliovirus, evolve rapidly due to the error-prone nature of the polymerase enzymes involved in genome replication. Fixation of advantageous mutations may require the acquisition of complementary mutations which can act in concert to achieve a favorable phenotype. This study highlights a compensatory role of a nonstructural regulatory protein, 2Apro, for an otherwise lethal mutation of the structural VP1 protein to facilitate increased thermal resistance. Studying how viruses respond to selection pressures is important for understanding mechanisms which underpin emergence of resistance and could be applied to the future development of antiviral agents and vaccines.
Collapse
Affiliation(s)
- Oluwapelumi O Adeyemi
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Lee Sherry
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Joseph C Ward
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Danielle M Pierce
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Morgan R Herod
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - David J Rowlands
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Nicola J Stonehouse
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
25
|
Fusade-Boyer M, Dupré G, Bessière P, Khiar S, Quentin-Froignant C, Beck C, Lecollinet S, Rameix-Welti MA, Eléouët JF, Tangy F, Lajoie B, Bertagnoli S, Vidalain PO, Gallardo F, Volmer R. Evaluation of the Antiviral Activity of Sephin1 Treatment and Its Consequences on eIF2α Phosphorylation in Response to Viral Infections. Front Immunol 2019; 10:134. [PMID: 30809223 PMCID: PMC6379315 DOI: 10.3389/fimmu.2019.00134] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/16/2019] [Indexed: 12/27/2022] Open
Abstract
The guanabenz derivative Sephin1 has recently been proposed to increase the levels of translation initiation factor 2 (eIF2α) phosphorylation by inhibiting dephosphorylation by the protein phosphatase 1-GADD34 (PPP1R15A) complex. As phosphorylation of eIF2α by protein kinase R (PKR) is a prominent cellular antiviral pathway, we evaluated the consequences of Sephin1 treatment on virus replication. Our results provide evidence that Sephin1 downregulates replication of human respiratory syncytial virus, measles virus, human adenovirus 5 virus, human enterovirus D68, human cytomegalovirus, and rabbit myxoma virus. However, Sephin1 proved to be inactive against influenza virus, as well as against Japanese encephalitis virus. Sephin1 increased the levels of phosphorylated eIF2α in cells exposed to a PKR agonist. By contrast, in virus-infected cells, the levels of phosphorylated eIF2α did not always correlate with the inhibition of virus replication by Sephin1. This work identifies Sephin1 as an antiviral molecule in cell culture against RNA, as well as DNA viruses belonging to phylogenetically distant families.
Collapse
Affiliation(s)
| | - Gabriel Dupré
- Université de Toulouse, ENVT, INRA, UMR 1225, Toulouse, France
| | - Pierre Bessière
- Université de Toulouse, ENVT, INRA, UMR 1225, Toulouse, France
| | - Samira Khiar
- Viral Genomics and Vaccination Unit, CNRS UMR-3569, Institut Pasteur, Paris, France
| | - Charlotte Quentin-Froignant
- Université de Toulouse, ENVT, INRA, UMR 1225, Toulouse, France.,NeoVirTech SAS, Institute for Advanced Life Science Technology, Toulouse, France
| | - Cécile Beck
- UMR 1161 Virology, INRA, ANSES, Ecole Nationale Vétérinaire d'Alfort, ANSES Animal Health Laboratory, EURL for Equine Diseases, Maisons-Alfort, France
| | - Sylvie Lecollinet
- UMR 1161 Virology, INRA, ANSES, Ecole Nationale Vétérinaire d'Alfort, ANSES Animal Health Laboratory, EURL for Equine Diseases, Maisons-Alfort, France
| | - Marie-Anne Rameix-Welti
- UMR INSERM U1173 2I, UFR des Sciences de la Santé Simone Veil-UVSQ, Montigny-le-Bretonneux, France.,AP-HP, Laboratoire de Microbiologie, Hôpital Ambroise Paré, Boulogne-Billancourt, France
| | - Jean-François Eléouët
- Unité de Virologie et Immunologie Moléculaires (UR892), INRA, Université Paris-Saclay, Jouy-en-Josas, France
| | - Frédéric Tangy
- Viral Genomics and Vaccination Unit, CNRS UMR-3569, Institut Pasteur, Paris, France
| | - Barbora Lajoie
- Laboratoire de Génie Chimique CNRS, INPT, UPS Université de Toulouse III, Faculté des Sciences Pharmaceutiques, Toulouse, France
| | | | - Pierre-Olivier Vidalain
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, Equipe Chimie & Biologie, Modélisation et Immunologie pour la Thérapie, CNRS UMR 8601, Université Paris Descartes, Paris, France
| | - Franck Gallardo
- NeoVirTech SAS, Institute for Advanced Life Science Technology, Toulouse, France
| | - Romain Volmer
- Université de Toulouse, ENVT, INRA, UMR 1225, Toulouse, France
| |
Collapse
|
26
|
Contemporary Circulating Enterovirus D68 Strains Have Acquired the Capacity for Viral Entry and Replication in Human Neuronal Cells. mBio 2018; 9:mBio.01954-18. [PMID: 30327438 PMCID: PMC6191546 DOI: 10.1128/mbio.01954-18] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Since the EV-D68 outbreak during the summer of 2014, evidence of a causal link to a type of limb paralysis (AFM) has been mounting. In this article, we describe a neuronal cell culture model (SH-SY5Y cells) in which a subset of contemporary 2014 outbreak strains of EV-D68 show infectivity in neuronal cells, or neurotropism. We confirmed the difference in neurotropism in vitro using primary human neuron cell cultures and in vivo with a mouse paralysis model. Using the SH-SY5Y cell model, we determined that a barrier to viral entry is at least partly responsible for neurotropism. SH-SY5Y cells may be useful in determining if specific EV-D68 genetic determinants are associated with neuropathogenesis, and replication in this cell line could be used as rapid screening tool for identification of neurotropic EV-D68 strains. This may assist with better understanding of pathogenesis and epidemiology and with the development of potential therapies. Enterovirus D68 (EV-D68) has historically been associated with respiratory illnesses. However, in the summers of 2014 and 2016, EV-D68 outbreaks coincided with a spike in polio-like acute flaccid myelitis/paralysis (AFM/AFP) cases. This raised concerns that EV-D68 could be the causative agent of AFM during these recent outbreaks. To assess the potential neurotropism of EV-D68, we utilized the neuroblastoma-derived neuronal cell line SH-SY5Y as a cell culture model to determine if differential infection is observed for different EV-D68 strains. In contrast to HeLa and A549 cells, which support viral infection of all EV-D68 strains tested, SH-SY5Y cells only supported infection by a subset of contemporary EV-D68 strains, including isolates from the 2014 outbreak. Viral replication and infectivity in SH-SY5Y were assessed using multiple assays: virus production, cytopathic effects, cellular ATP release, and VP1 capsid protein production. Similar differential neurotropism was also observed in differentiated SH-SY5Y cells, primary human neuron cultures, and a mouse paralysis model. Using the SH-SY5Y cell culture model, we determined that barriers to viral binding and entry were at least partly responsible for the differential infectivity phenotype. Transfection of genomic RNA into SH-SY5Y generated virions for all EV-D68 isolates, but only a single round of replication was observed from strains that could not directly infect SH-SY5Y. In addition to supporting virus replication and other functional studies, this cell culture model may help identify the signatures of virulence to confirm epidemiological associations between EV-D68 strains and AFM and allow for the rapid identification and characterization of emerging neurotropic strains.
Collapse
|
27
|
Tseligka ED, Sobo K, Stoppini L, Cagno V, Abdul F, Piuz I, Meylan P, Huang S, Constant S, Tapparel C. A VP1 mutation acquired during an enterovirus 71 disseminated infection confers heparan sulfate binding ability and modulates ex vivo tropism. PLoS Pathog 2018; 14:e1007190. [PMID: 30075025 PMCID: PMC6093697 DOI: 10.1371/journal.ppat.1007190] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 08/15/2018] [Accepted: 06/29/2018] [Indexed: 12/16/2022] Open
Abstract
Enterovirus 71 (EV71) causes hand, foot and mouth disease, a mild and self-limited illness that is sometimes associated with severe neurological complications. EV71 neurotropic determinants remain ill-defined to date. We previously identified a mutation in the VP1 capsid protein (L97R) that was acquired over the course of a disseminated infection in an immunocompromised host. The mutation was absent in the respiratory tract but was present in the gut (as a mixed population) and in blood and cerebrospinal fluid (as a dominant species). In this study, we demonstrated that this mutation does not alter the dependence of EV71 on the human scavenger receptor class B2 (SCARB2), while it enables the virus to bind to the heparan sulfate (HS) attachment receptor and modifies viral tropism in cell lines and in respiratory, intestinal and neural tissues. Variants with VP197L or VP197R were able to replicate to high levels in intestinal and neural tissues and, to a lesser extent, in respiratory tissues, but their preferred entry site (from the luminal or basal tissue side) differed in respiratory and intestinal tissues and correlated with HS expression levels. These data account for the viral populations sequenced from the patient's respiratory and intestinal samples and suggest that improved dissemination, resulting from an acquired ability to bind HS, rather than specific neurotropism determinants, enabled the virus to reach and infect the central nervous system. Finally, we showed that iota-carrageenan, a highly sulfated polysaccharide, efficiently blocks the replication of HS-dependent variants in cells and 2D neural cultures. Overall, the results of this study emphasize the importance of HS binding in EV71 pathogenesis and open new avenues for the development of antiviral molecules that may prevent this virus's dissemination.
Collapse
Affiliation(s)
- Eirini D. Tseligka
- Department of Microbiology and Molecular Medicine, University of Geneva Medical School, Geneva, Switzerland
| | - Komla Sobo
- Department of Microbiology and Molecular Medicine, University of Geneva Medical School, Geneva, Switzerland
| | - Luc Stoppini
- Tissue Engineering Laboratory, HES-SO/University of Applied Sciences, Geneva, Western Switzerland
| | - Valeria Cagno
- Department of Microbiology and Molecular Medicine, University of Geneva Medical School, Geneva, Switzerland
| | - Fabien Abdul
- Department of Microbiology and Molecular Medicine, University of Geneva Medical School, Geneva, Switzerland
| | - Isabelle Piuz
- Department of Microbiology and Molecular Medicine, University of Geneva Medical School, Geneva, Switzerland
| | - Pascal Meylan
- Institute of Microbiology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | | | | | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, University of Geneva Medical School, Geneva, Switzerland
| |
Collapse
|