1
|
Alam F, Li Y, Vogt MR. Parechovirus: neglected for too long? J Virol 2025; 99:e0184624. [PMID: 40130875 PMCID: PMC11998499 DOI: 10.1128/jvi.01846-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025] Open
Abstract
Parechoviruses are non-enveloped, positive-sense, single-stranded RNA viruses that have been isolated from multiple vertebrate species. Infection with these etiologic agents of typically mild childhood respiratory and gastrointestinal illness in humans is nearly universal, and a subset of infected neonates and infants develop severe neurologic diseases. Rodent parechoviruses cause myocarditis, encephalitis, and perinatal death in multiple rodent species. The key steps of the viral life cycle, clinical characteristics, and global burden of these viruses are not well characterized yet, particularly for nonhuman parechoviruses. Here, we review the history of human and nonhuman parechovirus isolation, global seroprevalence and distribution, viral biology, and evolution, considering these factors might contribute to host specificity, virulence, tissue tropism, pathogenesis, host immunity, and population dynamics.
Collapse
Affiliation(s)
- Fahmida Alam
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - You Li
- Department of Pediatrics, Division of Infectious Diseases, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Matthew R. Vogt
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Pediatrics, Division of Infectious Diseases, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
2
|
Lulla V, Sridhar A. Understanding neurotropic enteric viruses: routes of infection and mechanisms of attenuation. Cell Mol Life Sci 2024; 81:413. [PMID: 39365457 PMCID: PMC11452578 DOI: 10.1007/s00018-024-05450-6] [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: 07/10/2024] [Revised: 09/03/2024] [Accepted: 09/12/2024] [Indexed: 10/05/2024]
Abstract
The intricate connection between the gut and the brain involves multiple routes. Several viral families begin their infection cycle in the intestinal tract. However, amongst the long list of viral intestinal pathogens, picornaviruses, and astroviruses stand out for their ability to transition from the intestinal epithelia to central or peripheral nervous system cells. In immunocompromised, neonates and young children, these viral infections can manifest as severe diseases, such as encephalitis, meningitis, and acute flaccid paralysis. What confers this remarkable plasticity and makes them efficient in infecting cells of the gut and the brain axes? Here, we review the current understanding of the virus infection along the gut-brain axis for some enteric viruses and discuss the molecular mechanisms of their attenuation.
Collapse
Affiliation(s)
- Valeria Lulla
- Division of Virology, Department of Pathology, Addenbrooke's Hospital, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Adithya Sridhar
- OrganoVIR Labs, Department of Pediatric Infectious Diseases, Amsterdam UMC, location Academic Medical Center, Amsterdam Institute for Reproduction and Development, University of Amsterdam, Meibergdreef 9, 1100 AZ, Amsterdam, The Netherlands
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC, location Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, 1100 AZ, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Xie Z, Khamrin P, Maneekarn N, Kumthip K. Epidemiology of Enterovirus Genotypes in Association with Human Diseases. Viruses 2024; 16:1165. [PMID: 39066327 PMCID: PMC11281466 DOI: 10.3390/v16071165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Enteroviruses (EVs) are well-known causes of a wide range of infectious diseases in infants and young children, ranging from mild illnesses to severe conditions, depending on the virus genotypes and the host's immunity. Recent advances in molecular surveillance and genotyping tools have identified over 116 different human EV genotypes from various types of clinical samples. However, the current knowledge about most of these genotypes, except for those of well-known genotypes like EV-A71 and EV-D68, is still limited due to a lack of comprehensive EV surveillance systems. This limited information makes it difficult to understand the true burden of EV-related diseases globally. Furthermore, the specific EV genotype associated with diseases varies according to country, population group, and study period. The same genotype can exhibit different epidemiological features in different areas. By integrating the data from established EV surveillance systems in the USA, Europe, Japan, and China, in combination with other EV infection studies, we can elaborate a better understanding of the distribution of prevalent EV genotypes and the diseases associated with EV. This review analyzed the data from various EV surveillance databases and explored the EV seroprevalence and the association of specific EV genotypes with human diseases.
Collapse
Affiliation(s)
- Zhenfeng Xie
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (Z.X.); (P.K.); (N.M.)
- Guangxi Colleges and Universities Key Laboratory of Basic Research and Transformation of Cancer Immunity and Infectious Diseases, Youjiang Medical University for Nationalities, Baise 533000, China
| | - Pattara Khamrin
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (Z.X.); (P.K.); (N.M.)
- Center of Excellence in Emerging and Re-Emerging Diarrheal Viruses, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Niwat Maneekarn
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (Z.X.); (P.K.); (N.M.)
- Center of Excellence in Emerging and Re-Emerging Diarrheal Viruses, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kattareeya Kumthip
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (Z.X.); (P.K.); (N.M.)
- Center of Excellence in Emerging and Re-Emerging Diarrheal Viruses, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
4
|
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
|
5
|
Devaux CA, Pontarotti P, Levasseur A, Colson P, Raoult D. Is it time to switch to a formulation other than the live attenuated poliovirus vaccine to prevent poliomyelitis? Front Public Health 2024; 11:1284337. [PMID: 38259741 PMCID: PMC10801389 DOI: 10.3389/fpubh.2023.1284337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
The polioviruses (PVs) are mainly transmitted by direct contact with an infected person through the fecal-oral route and respiratory secretions (or more rarely via contaminated water or food) and have a primary tropism for the gut. After their replication in the gut, in rare cases (far less than 1% of the infected individuals), PVs can spread to the central nervous system leading to flaccid paralysis, which can result in respiratory paralysis and death. By the middle of the 20th century, every year the wild polioviruses (WPVs) are supposed to have killed or paralyzed over half a million people. The introduction of the oral poliovirus vaccines (OPVs) through mass vaccination campaigns (combined with better application of hygiene measures), was a success story which enabled the World Health Organization (WHO) to set the global eradication of poliomyelitis as an objective. However this strategy of viral eradication has its limits as the majority of poliomyelitis cases today arise in individuals infected with circulating vaccine-derived polioviruses (cVDPVs) which regain pathogenicity following reversion or recombination. In recent years (between January 2018 and May 2023), the WHO recorded 8.8 times more cases of polio which were linked to the attenuated OPV vaccines (3,442 polio cases after reversion or recombination events) than cases linked to a WPV (390 cases). Recent knowledge of the evolution of RNA viruses and the exchange of genetic material among biological entities of the intestinal microbiota, call for a reassessment of the polio eradication vaccine strategies.
Collapse
Affiliation(s)
- Christian Albert Devaux
- Laboratory Microbes Evolution Phylogeny and Infection (MEPHI), Aix-Marseille Université, IRD, APHM, Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
- Centre National de la Recherche Scientifique (CNRS-SNC5039), Marseille, France
| | - Pierre Pontarotti
- Laboratory Microbes Evolution Phylogeny and Infection (MEPHI), Aix-Marseille Université, IRD, APHM, Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
- Centre National de la Recherche Scientifique (CNRS-SNC5039), Marseille, France
| | - Anthony Levasseur
- Laboratory Microbes Evolution Phylogeny and Infection (MEPHI), Aix-Marseille Université, IRD, APHM, Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Philippe Colson
- Laboratory Microbes Evolution Phylogeny and Infection (MEPHI), Aix-Marseille Université, IRD, APHM, Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Didier Raoult
- Laboratory Microbes Evolution Phylogeny and Infection (MEPHI), Aix-Marseille Université, IRD, APHM, Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| |
Collapse
|
6
|
Meyers G, Tews BA. Self-Replicating RNA Derived from the Genomes of Positive-Strand RNA Viruses. Methods Mol Biol 2024; 2786:25-49. [PMID: 38814389 DOI: 10.1007/978-1-0716-3770-8_2] [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] [Indexed: 05/31/2024]
Abstract
Self-replicating RNA derived from the genomes of positive-strand RNA viruses represents a powerful tool for both molecular studies on virus biology and approaches to novel safe and effective vaccines. The following chapter summarizes the principles how such RNAs can be established and used for design of vaccines. Due to the large variety of strategies needed to circumvent specific pitfalls in the design of such constructs the technical details of the experiments are not described here but can be found in the cited literature.
Collapse
Affiliation(s)
- Gregor Meyers
- Institut für Immunologie, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Birke Andrea Tews
- Institut für Infektionsmedizin, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany.
| |
Collapse
|
7
|
Tur-Planells V, García-Sastre A, Cuadrado-Castano S, Nistal-Villan E. Engineering Non-Human RNA Viruses for Cancer Therapy. Vaccines (Basel) 2023; 11:1617. [PMID: 37897020 PMCID: PMC10611381 DOI: 10.3390/vaccines11101617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Alongside the development and progress in cancer immunotherapy, research in oncolytic viruses (OVs) continues advancing novel treatment strategies to the clinic. With almost 50 clinical trials carried out over the last decade, the opportunities for intervention using OVs are expanding beyond the old-fashioned concept of "lytic killers", with promising breakthrough therapeutic strategies focused on leveraging the immunostimulatory potential of different viral platforms. This review presents an overview of non-human-adapted RNA viruses engineered for cancer therapy. Moreover, we describe the diverse strategies employed to manipulate the genomes of these viruses to optimize their therapeutic capabilities. By focusing on different aspects of this particular group of viruses, we describe the insights into the promising advancements in the field of virotherapy and its potential to revolutionize cancer treatment.
Collapse
Affiliation(s)
- Vicent Tur-Planells
- Microbiology Section, Department of Pharmaceutical Science and Health, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain;
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sara Cuadrado-Castano
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Genomics Institute (IGI), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Estanislao Nistal-Villan
- Microbiology Section, Department of Pharmaceutical Science and Health, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain;
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, 28668 Boadilla del Monte, Spain
| |
Collapse
|
8
|
Sion E, Ab-Rahim S, Muhamad M. Trends on Human Norovirus Virus-like Particles (HuNoV-VLPs) and Strategies for the Construction of Infectious Viral Clones toward In Vitro Replication. Life (Basel) 2023; 13:1447. [PMID: 37511822 PMCID: PMC10381778 DOI: 10.3390/life13071447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/07/2023] [Accepted: 06/10/2023] [Indexed: 07/30/2023] Open
Abstract
Most acute gastroenteritis (AGE) outbreaks and sporadic cases in developing countries are attributable to infection by human norovirus (HuNoV), the enteric virus mainly transmitted via fecal-contaminated water. However, it has been challenging to study HuNoV due to the lack of suitable systems to cultivate and replicate the virus, hindering the development of treatments and vaccines. Researchers have been using virus-like particles (VLPs) and infectious viral clones to overcome this challenge as alternatives to fresh virus isolates in various in vitro and ex vivo models. VLPs are multiprotein structures that mimic the wild-type virus but cannot replicate in host cells due to the lack of genetic materials for replication, limiting downstream analysis of the virus life cycle and pathogenesis. The development of in vitro cloning systems has shown promise for HuNoV replication studies. This review discusses the approaches for constructing HuNoV-VLPs and infectious viral clones, the techniques involved, and the challenges faced. It also highlights the relationship between viral genes and their protein products and provides a perspective on technical considerations for producing efficient HuNoV-VLPs and infectious viral clones, which could substitute for native human noroviruses in future studies.
Collapse
Affiliation(s)
- Emilly Sion
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Selangor Branch, Sungai Buloh Campus, Sungai Buloh 47000, Selangor, Malaysia
| | - Sharaniza Ab-Rahim
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Selangor Branch, Sungai Buloh Campus, Sungai Buloh 47000, Selangor, Malaysia
| | - Mudiana Muhamad
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Selangor Branch, Sungai Buloh Campus, Sungai Buloh 47000, Selangor, Malaysia
| |
Collapse
|
9
|
Quarleri J. Poliomyelitis is a current challenge: long-term sequelae and circulating vaccine-derived poliovirus. GeroScience 2023; 45:707-717. [PMID: 36260265 PMCID: PMC9886775 DOI: 10.1007/s11357-022-00672-7] [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: 09/13/2022] [Accepted: 10/12/2022] [Indexed: 02/03/2023] Open
Abstract
For more than 20 years, the World Health Organization Western Pacific Region (WPR) has been polio-free. However, two current challenges are still polio-related. First, around half of poliomyelitis elderly survivors suffer late poliomyelitis sequelae with a substantial impact on daily activities and quality of life, experiencing varying degrees of residual weakness as they age. The post-polio syndrome as well as accelerated aging may be involved. Second, after the worldwide Sabin oral poliovirus (OPV) vaccination, the recent reappearance of strains of vaccine-derived poliovirus (VDPV) circulating in the environment is worrisome and able to persistent person-to-person transmission. Such VDPV strains exhibit atypical genetic characteristics and reversed neurovirulence that can cause paralysis similarly to wild poliovirus, posing a significant obstacle to the elimination of polio. Immunization is essential for preventing paralysis in those who are exposed to the poliovirus. Stress the necessity of maintaining high vaccination rates because declining immunity increases the likelihood of reemergence. If mankind wants to eradicate polio in the near future, measures to raise immunization rates and living conditions in poorer nations are needed, along with strict observation. New oral polio vaccine candidates offer a promissory tool for this goal.
Collapse
Affiliation(s)
- Jorge Quarleri
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.
- Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
10
|
Reverse genetics in virology: A double edged sword. BIOSAFETY AND HEALTH 2022. [DOI: 10.1016/j.bsheal.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
11
|
Stenglein MD. The Case for Studying New Viruses of New Hosts. Annu Rev Virol 2022; 9:157-172. [PMID: 35671564 DOI: 10.1146/annurev-virology-100220-112915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Virology has largely focused on viruses that are pathogenic to humans or to the other species that we care most about. There is no doubt that this has been a worthwhile investment. But many transformative advances have been made through the in-depth study of relatively obscure viruses that do not appear on lists of prioritized pathogens. In this review, I highlight the benefits that can accrue from the study of viruses and hosts off the beaten track. I take stock of viral sequence diversity across host taxa as an estimate of the bias that exists in our understanding of host-virus interactions. I describe the gains that have been made through the metagenomic discovery of thousands of new viruses in previously unsampled hosts as well as the limitations of metagenomic surveys. I conclude by suggesting that the study of viruses that naturally infect existing and emerging model organisms represents an opportunity to push virology forward in useful and hard to predict ways.Expected final online publication date for the Annual Review of Virology, Volume 9 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Mark D Stenglein
- Center for Vector-Borne Infectious Diseases, Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA;
| |
Collapse
|
12
|
Francisco-Velilla R, Embarc-Buh A, Abellan S, Martinez-Salas E. Picornavirus translation strategies. FEBS Open Bio 2022; 12:1125-1141. [PMID: 35313388 PMCID: PMC9157412 DOI: 10.1002/2211-5463.13400] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/02/2022] [Accepted: 03/18/2022] [Indexed: 12/15/2022] Open
Abstract
The genome of viruses classified as picornaviruses consists of a single monocistronic positive strand RNA. The coding capacity of these RNA viruses is rather limited, and thus, they rely on the cellular machinery for their viral replication cycle. Upon the entry of the virus into susceptible cells, the viral RNA initially competes with cellular mRNAs for access to the protein synthesis machinery. Not surprisingly, picornaviruses have evolved specialized strategies that successfully allow the expression of viral gene products, which we outline in this review. The main feature of all picornavirus genomes is the presence of a heavily structured RNA element on the 5´UTR, referred to as an internal ribosome entry site (IRES) element, which directs viral protein synthesis as well and, consequently, triggers the subsequent steps required for viral replication. Here, we will summarize recent studies showing that picornavirus IRES elements consist of a modular structure, providing sites of interaction for ribosome subunits, eIFs, and a selective group of RNA‐binding proteins.
Collapse
Affiliation(s)
| | - Azman Embarc-Buh
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
| | - Salvador Abellan
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
| | | |
Collapse
|
13
|
Thompson KM, Kalkowska DA, Badizadegan K. Hypothetical emergence of poliovirus in 2020: part 1. Consequences of policy decisions to respond using nonpharmaceutical interventions. Expert Rev Vaccines 2021; 20:465-481. [PMID: 33624568 DOI: 10.1080/14760584.2021.1891888] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES As efforts to control COVID-19 continue, we simulate hypothetical emergence of wild poliovirus assuming an immunologically naïve population. This differs from the current global experience with polio and serves as a model for responding to future pandemics. METHODS Applying an established global model, we assume a fully susceptible global population to polioviruses, independently introduce a virus with properties of each of the three stable wild poliovirus serotypes, and explore the impact of strategies that range from doing nothing to seeking global containment and eradication. RESULTS We show the dynamics of paralytic cases as the virus spreads globally. We demonstrate the difficulty of eradication unless aggressive efforts begin soon after initial disease detection. Different poliovirus serotypes lead to different trajectories and burdens of disease. In the absence of aggressive measures, the virus would become globally endemic in 2-10 years, and cumulative paralytic cases would exceed 4-40 million depending on serotype, with the burden of disease shifting to younger ages. CONCLUSIONS The opportunity to eradicate emerging infections represents an important public policy choice. If the world first observed the emergence of wild poliovirus in 2020, adopting aggressive control strategies would have been required to prevent a devastating global pandemic.
Collapse
|
14
|
Saha J, Bhattacharjee S, Pal Sarkar M, Saha BK, Basak HK, Adhikary S, Roy V, Mandal P, Chatterjee A, Pal A. A comparative genomics-based study of positive strand RNA viruses emphasizing on SARS-CoV-2 utilizing dinucleotide signature, codon usage and codon context analyses. GENE REPORTS 2021; 23:101055. [PMID: 33615042 PMCID: PMC7887452 DOI: 10.1016/j.genrep.2021.101055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/20/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022]
Abstract
The novel corona virus disease or COVID-19 caused by a positive strand RNA virus (PRV) called SARS-CoV-2 is plaguing the entire planet as we conduct this study. In this study a multifaceted analysis was carried out employing dinucleotide signature, codon usage and codon context to compare and unravel the genomic as well as genic characteristics of the SARS-CoV-2 isolates and how they compare to other PRVs which represents some of the most pathogenic human viruses. The main emphasis of this study was to comprehend the codon biology of the SARS-CoV-2 in the backdrop of the other PRVs like Poliovirus, Japanese encephalitis virus, Hepatitis C virus, Norovirus, Rubella virus, Semliki Forest virus, Zika virus, Dengue virus, Human rhinoviruses and the Betacoronaviruses since codon usage pattern along with the nucleotide composition prevalent within the viral genome helps to understand the biology and evolution of viruses. Our results suggest discrete genomic dinucleotide signature within the PRVs. Some of the genes from the different SARS-CoV-2 isolates were also found to demonstrate heterogeneity in terms of their dinucleotide signature. The SARS-CoV-2 isolates also demonstrated a codon context trend characteristically dissimilar to the other PRVs. The findings of this study are expected to contribute to the developing global knowledge base in countering COVID-19.
Collapse
Key Words
- CAI, Codon Adaptation Index
- CNS, Central Nervous System
- COVID-19
- CRS, Congenital Rubella Syndrome
- CUB, Codon Usage Bias
- Codon context
- Codon usage bias
- Coronaviruses
- Fop, Frequency of optimal codons
- GC1, Guanine and Cytosine content on the first position of the codon
- GC2, Guanine and Cytosine content on the second position of the codon
- GC3, Guanine and Cytosine content on the third position of the codon
- HCV, Hepatitis C Virus
- MERS, Middle East Respiratory Syndrome
- MFE, Minimum Free Energy
- Nc, Effective Number of Codons
- PCA, Principal Component Analysis
- PRV, Positive strand RNA Virus
- Positive strand RNA virus
- RCDI, Relative Codon De-Optimization Index
- RSCU, Relative Synonymous Codon Usage
- SARS, Severe Acute Respiratory Syndrome
- SARS-CoV-2
- SARS-CoV-2, Severe Acute Respiratory Syndrome Coronavirus 2
- SCUO, Synonymous Codon Usage Order
- SiD, Similarity Index
Collapse
Affiliation(s)
- Jayanti Saha
- Microbiology & Computational Biology Laboratory, Department of Botany, Raiganj University, Raiganj PIN-733 134, Uttar Dinajpur, West Bengal, India
| | - Sukanya Bhattacharjee
- Microbiology & Computational Biology Laboratory, Department of Botany, Raiganj University, Raiganj PIN-733 134, Uttar Dinajpur, West Bengal, India
| | - Monalisha Pal Sarkar
- Mycology & Plant Pathology Laboratory, Department of Botany, Raiganj University, Raiganj PIN-733 134, Uttar Dinajpur, West Bengal, India
| | - Barnan Kumar Saha
- Microbiology & Computational Biology Laboratory, Department of Botany, Raiganj University, Raiganj PIN-733 134, Uttar Dinajpur, West Bengal, India
| | - Hriday Kumar Basak
- Department of Chemistry, Raiganj University, Raiganj PIN-733 134, Uttar Dinajpur, West Bengal, India
| | - Samarpita Adhikary
- Microbiology & Computational Biology Laboratory, Department of Botany, Raiganj University, Raiganj PIN-733 134, Uttar Dinajpur, West Bengal, India
| | - Vivek Roy
- Microbiology & Computational Biology Laboratory, Department of Botany, Raiganj University, Raiganj PIN-733 134, Uttar Dinajpur, West Bengal, India
| | - Parimal Mandal
- Mycology & Plant Pathology Laboratory, Department of Botany, Raiganj University, Raiganj PIN-733 134, Uttar Dinajpur, West Bengal, India
| | - Abhik Chatterjee
- Department of Chemistry, Raiganj University, Raiganj PIN-733 134, Uttar Dinajpur, West Bengal, India
| | - Ayon Pal
- Microbiology & Computational Biology Laboratory, Department of Botany, Raiganj University, Raiganj PIN-733 134, Uttar Dinajpur, West Bengal, India
| |
Collapse
|
15
|
Woods Acevedo MA, Pfeiffer JK. Microbiota-independent antiviral effects of antibiotics on poliovirus and coxsackievirus. Virology 2020; 546:20-24. [PMID: 32452414 PMCID: PMC7253499 DOI: 10.1016/j.virol.2020.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/30/2020] [Accepted: 04/03/2020] [Indexed: 01/14/2023]
Abstract
Coxsackieviruses primarily infect the gastrointestinal tract of humans, but they can disseminate systemically and cause severe disease. Using antibiotic treatment regimens to deplete intestinal microbes in mice, several groups have shown that bacteria promote oral infection with a variety of enteric viruses. However, it is unknown whether antibiotics have microbiota-independent antiviral effects for enteric viruses or whether antibiotics influence extra-intestinal, systemic infection. Here, we examined the effects of antibiotics on systemic enteric virus infection by performing intraperitoneal injections of either coxsackievirus B3 (CVB3) or poliovirus followed by quantification of viral titers. We found that antibiotic treatment reduced systemic infection for both viruses. Interestingly, antibiotics reduced CVB3 titers in germ-free mice, suggesting that antibiotic treatment alters CVB3 infection through a microbiota-independent mechanism. Overall, these data provide further evidence that antibiotics can have noncanonical effects on viral infection.
Collapse
Affiliation(s)
- Mikal A Woods Acevedo
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Julie K Pfeiffer
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
16
|
Jorgensen D, Pons-Salort M, Shaw AG, Grassly NC. The role of genetic sequencing and analysis in the polio eradication programme. Virus Evol 2020; 6:veaa040. [PMID: 32782825 PMCID: PMC7409915 DOI: 10.1093/ve/veaa040] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Genetic sequencing of polioviruses detected through clinical and environmental surveillance is used to confirm detection, identify their likely origin, track geographic patterns of spread, and determine the appropriate vaccination response. The critical importance of genetic sequencing and analysis to the Global Polio Eradication Initiative has grown with the increasing incidence of vaccine-derived poliovirus (VDPV) infections in Africa specifically (470 reported cases in 2019), and globally, alongside persistent transmission of serotype 1 wild-type poliovirus in Pakistan and Afghanistan (197 reported cases in 2019). Adapting what has been learned about the virus genetics and evolution to address these threats has been a major focus of recent work. Here, we review how phylogenetic and phylogeographic methods have been used to trace the spread of wild-type polioviruses and identify the likely origins of VDPVs. We highlight the analysis methods and sequencing technology currently used and the potential for new technologies to speed up poliovirus detection and the interpretation of genetic data. At a pivotal point in the eradication campaign with the threat of anti-vaccine sentiment and donor and public fatigue, innovation is critical to maintain drive and overcome the last remaining circulating virus.
Collapse
Affiliation(s)
- David Jorgensen
- Department of Infectious Disease Epidemiology, Imperial College London, St Mary's Campus, Norfolk Place, London W2 1PG, UK
| | - Margarita Pons-Salort
- Department of Infectious Disease Epidemiology, Imperial College London, St Mary's Campus, Norfolk Place, London W2 1PG, UK
| | - Alexander G Shaw
- Department of Infectious Disease Epidemiology, Imperial College London, St Mary's Campus, Norfolk Place, London W2 1PG, UK
| | - Nicholas C Grassly
- Department of Infectious Disease Epidemiology, Imperial College London, St Mary's Campus, Norfolk Place, London W2 1PG, UK
| |
Collapse
|
17
|
Ávila-Pérez G, Nogales A, Park JG, Vasquez DM, Dean DA, Barravecchia M, Perez DR, Almazán F, Martínez-Sobrido L. In vivo rescue of recombinant Zika virus from an infectious cDNA clone and its implications in vaccine development. Sci Rep 2020; 10:512. [PMID: 31949262 PMCID: PMC6965646 DOI: 10.1038/s41598-020-57545-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 01/03/2020] [Indexed: 02/06/2023] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne member of the Flaviviridae family that has been known to circulate for decades causing mild febrile illness. The more recent ZIKV outbreaks in the Americas and the Caribbean associated with congenital malformations and Guillain-Barré syndrome in adults have placed public health officials in high alert and highlight the significant impact of ZIKV on human health. New technologies to study the biology of ZIKV and to develop more effective prevention options are highly desired. In this study we demonstrate that direct delivery in mice of an infectious ZIKV cDNA clone allows the rescue of recombinant (r)ZIKV in vivo. A bacterial artificial chromosome containing the sequence of ZIKV strain Paraiba/2015 under the control of the cytomegalovirus promoter was complexed with a commercial transfection reagent and administrated using different routes in type-I interferon receptor deficient A129 mice. Clinical signs and death associated with ZIKV viremia were observed in mice. The rZIKV recovered from these mice remained fully virulent in a second passage in mice. Interestingly, infectious rZIKV was also recovered after intraperitoneal inoculation of the rZIKV cDNA in the absence of transfection reagent. Further expanding these studies, we demonstrate that a single intraperitoneal inoculation of a cDNA clone encoding an attenuated rZIKV was safe, highly immunogenic, and provided full protection against lethal ZIKV challenge. This novel in vivo reverse genetics method is a potentially suitable delivery platform for the study of wild-type and live-attenuated ZIKV devoid of confounding factors typical associated with in vitro systems. Moreover, our results open the possibility of employing similar in vivo reverse genetic approaches for the generation of other viruses and, therefore, change the way we will use reverse genetics in the future.
Collapse
MESH Headings
- Animals
- Chlorocebus aethiops
- Chromosomes, Artificial, Bacterial/genetics
- DNA, Complementary/genetics
- DNA, Complementary/immunology
- DNA, Viral/genetics
- DNA, Viral/immunology
- Disease Models, Animal
- Female
- Genetic Vectors/administration & dosage
- Male
- Mice
- Receptor, Interferon alpha-beta/genetics
- Reverse Genetics
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/immunology
- Vero Cells
- Viral Vaccines/administration & dosage
- Viral Vaccines/immunology
- Viremia/genetics
- Viremia/immunology
- Viremia/prevention & control
- Zika Virus/genetics
- Zika Virus/immunology
- Zika Virus Infection/genetics
- Zika Virus Infection/immunology
- Zika Virus Infection/prevention & control
Collapse
Affiliation(s)
- Gines Ávila-Pérez
- Department of Microbiology and Immunology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, New York, 14642, USA
| | - Aitor Nogales
- Department of Microbiology and Immunology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, New York, 14642, USA
- Center for Animal Health Research, INIA-CISA, 28130, Valdeolmos, Madrid, Spain
| | - Jun-Gyu Park
- Department of Microbiology and Immunology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, New York, 14642, USA
| | - Desarey Morales Vasquez
- Department of Microbiology and Immunology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, New York, 14642, USA
| | - David A Dean
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, New York, 14642, USA
| | - Michael Barravecchia
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, New York, 14642, USA
| | - Daniel R Perez
- Department of Population Health, Poultry Diagnostic and Research Center, University of Georgia, Georgia, USA
| | - Fernando Almazán
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), 3 Darwin street, 28049, Madrid, Spain.
| | - Luis Martínez-Sobrido
- Department of Microbiology and Immunology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, New York, 14642, USA.
| |
Collapse
|
18
|
Guo H, Li Y, Liu G, Jiang Y, Shen S, Bi R, Huang H, Cheng T, Wang C, Wei W. A second open reading frame in human enterovirus determines viral replication in intestinal epithelial cells. Nat Commun 2019; 10:4066. [PMID: 31492846 PMCID: PMC6731315 DOI: 10.1038/s41467-019-12040-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 08/19/2019] [Indexed: 02/08/2023] Open
Abstract
Human enteroviruses (HEVs) of the family Picornaviridae, which comprises non-enveloped RNA viruses, are ubiquitous worldwide. The majority of EV proteins are derived from viral polyproteins encoded by a single open reading frame (ORF). Here, we characterize a second ORF in HEVs that is crucial for viral intestinal infection. Disruption of ORF2p expression decreases the replication capacity of EV-A71 in human intestinal epithelial cells (IECs). Ectopic expression of ORF2p proteins derived from diverse enteric enteroviruses sensitizes intestinal cells to the replication of ORF2p-defective EV-A71 and respiratory enterovirus EV-D68. We show that the highly conserved WIGHPV domain of ORF2p is important for ORF2p-dependent viral intestinal infection. ORF2p expression is required for EV-A71 particle release from IECs and can support productive EV-D68 infection in IECs by facilitating virus release. Our results indicate that ORF2p is a determining factor for enteric enterovirus replication in IECs.
Collapse
Affiliation(s)
- Haoran Guo
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Translational Medicine, First Hospital, Jilin University, Changchun, Jilin, 130021, China.,Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, 130021, China
| | - Yan Li
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, 130021, China
| | - Guanchen Liu
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, 130021, China
| | - Yunhe Jiang
- Department of Pathogen Biology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, Jilin, 130021, China
| | - Siyu Shen
- Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, 130021, China
| | - Ran Bi
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Translational Medicine, First Hospital, Jilin University, Changchun, Jilin, 130021, China
| | - Honglan Huang
- Department of Pathogen Biology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, Jilin, 130021, China
| | - Tong Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
| | - Chunxi Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Translational Medicine, First Hospital, Jilin University, Changchun, Jilin, 130021, China
| | - Wei Wei
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Translational Medicine, First Hospital, Jilin University, Changchun, Jilin, 130021, China. .,Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
19
|
Abstract
The genus Enterovirus (EV) of the family Picornaviridae includes poliovirus, coxsackieviruses, echoviruses, numbered enteroviruses and rhinoviruses. These diverse viruses cause a variety of diseases, including non-specific febrile illness, hand-foot-and-mouth disease, neonatal sepsis-like disease, encephalitis, paralysis and respiratory diseases. In recent years, several non-polio enteroviruses (NPEVs) have emerged as serious public health concerns. These include EV-A71, which has caused epidemics of hand-foot-and-mouth disease in Southeast Asia, and EV-D68, which recently caused a large outbreak of severe lower respiratory tract disease in North America. Infections with these viruses are associated with severe neurological complications. For decades, most research has focused on poliovirus, but in recent years, our knowledge of NPEVs has increased considerably. In this Review, we summarize recent insights from enterovirus research with a special emphasis on NPEVs. We discuss virion structures, host-receptor interactions, viral uncoating and the recent discovery of a universal enterovirus host factor that is involved in viral genome release. Moreover, we briefly explain the mechanisms of viral genome replication, virion assembly and virion release, and describe potential targets for antiviral therapy. We reflect on how these recent discoveries may help the development of antiviral therapies and vaccines.
Collapse
|
20
|
Aguilera ER, Nguyen Y, Sasaki J, Pfeiffer JK. Bacterial Stabilization of a Panel of Picornaviruses. mSphere 2019; 4:e00183-19. [PMID: 30944213 PMCID: PMC6449606 DOI: 10.1128/msphere.00183-19] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 03/13/2019] [Indexed: 11/20/2022] Open
Abstract
Several viruses encounter various bacterial species within the host and in the environment. Despite these close encounters, the effects of bacteria on picornaviruses are not completely understood. Previous work determined that poliovirus (PV), an enteric virus, has enhanced virion stability when exposed to bacteria or bacterial surface polysaccharides such as lipopolysaccharide. Virion stabilization by bacteria may be important for interhost transmission, since a mutant PV with reduced bacterial binding had a fecal-oral transmission defect in mice. Therefore, we investigated whether bacteria broadly enhance stability of picornaviruses from three different genera: Enterovirus (PV and coxsackievirus B3 [CVB3]), Kobuvirus (Aichi virus), and Cardiovirus (mengovirus). Furthermore, to delineate strain-specific effects, we examined two strains of CVB3 and a PV mutant with enhanced thermal stability. We determined that specific bacterial strains enhance thermal stability of PV and CVB3, while mengovirus and Aichi virus are stable at high temperatures in the absence of bacteria. Additionally, we determined that bacteria or lipopolysaccharide can stabilize PV, CVB3, Aichi virus, and mengovirus during exposure to bleach. These effects are likely mediated through direct interactions with bacteria, since viruses bound to bacteria in a pulldown assay. Overall, this work reveals shared and distinct effects of bacteria on a panel of picornaviruses.IMPORTANCE Recent studies have shown that bacteria promote infection and stabilization of poliovirus particles, but the breadth of these effects on other members of the Picornaviridae family is unknown. Here, we compared the effects of bacteria on four distinct members of the Picornaviridae family. We found that bacteria reduced inactivation of all of the viruses during bleach treatment, but not all viral strains were stabilized by bacteria during heat treatment. Overall, our data provide insight into how bacteria play differential roles in picornavirus stability.
Collapse
Affiliation(s)
- Elizabeth R Aguilera
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Y Nguyen
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jun Sasaki
- Department of Virology and Parasitology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Julie K Pfeiffer
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
21
|
Woods Acevedo MA, Erickson AK, Pfeiffer JK. The Antibiotic Neomycin Enhances Coxsackievirus Plaque Formation. mSphere 2019; 4:e00632-18. [PMID: 30787120 PMCID: PMC6382971 DOI: 10.1128/msphere.00632-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/31/2019] [Indexed: 01/06/2023] Open
Abstract
Coxsackievirus typically infects humans via the gastrointestinal tract, which has a large number of microorganisms collectively referred to as the microbiota. To study how the intestinal microbiota influences enteric virus infection, several groups have used an antibiotic regimen in mice to deplete bacteria. These studies have shown that bacteria promote infection with several enteric viruses. However, very little is known about whether antibiotics influence viruses in a microbiota-independent manner. In this study, we sought to determine the effects of antibiotics on coxsackievirus B3 (CVB3) using an in vitro cell culture model in the absence of bacteria. We determined that an aminoglycoside antibiotic, neomycin, enhanced the plaque size of CVB3 strain Nancy. Neomycin treatment did not alter viral attachment, translation, or replication. However, we found that the positive charge of neomycin and other positively charged compounds enhanced viral diffusion by overcoming the negative inhibitory effect of sulfated polysaccharides present in agar overlays. Neomycin and the positively charged compound protamine also enhanced plaque formation of reovirus. Overall, these data provide further evidence that antibiotics can play noncanonical roles in viral infections and that this should be considered when studying enteric virus-microbiota interactions.IMPORTANCE Coxsackieviruses primarily infect the gastrointestinal tract of humans, but they can disseminate systemically and cause severe disease. Using antibiotic treatment regimens to deplete intestinal microbes in mice, several groups have shown the bacteria promote infection with a variety of enteric viruses. However, it is possible that antibiotics have microbiota-independent effects on viruses. Here we show that an aminoglycoside antibiotic, neomycin, can influence quantification of coxsackievirus in cultured cells in the absence of bacteria.
Collapse
Affiliation(s)
- Mikal A Woods Acevedo
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Andrea K Erickson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Julie K Pfeiffer
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
22
|
Lulla V, Dinan AM, Hosmillo M, Chaudhry Y, Sherry L, Irigoyen N, Nayak KM, Stonehouse NJ, Zilbauer M, Goodfellow I, Firth AE. An upstream protein-coding region in enteroviruses modulates virus infection in gut epithelial cells. Nat Microbiol 2018; 4:280-292. [PMID: 30478287 DOI: 10.1038/s41564-018-0297-1] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/19/2018] [Indexed: 11/09/2022]
Abstract
Enteroviruses comprise a large group of mammalian pathogens that includes poliovirus. Pathology in humans ranges from sub-clinical to acute flaccid paralysis, myocarditis and meningitis. Until now, all of the enteroviral proteins were thought to derive from the proteolytic processing of a polyprotein encoded in a single open reading frame. Here we report that many enterovirus genomes also harbour an upstream open reading frame (uORF) that is subject to strong purifying selection. Using echovirus 7 and poliovirus 1, we confirmed the expression of uORF protein in infected cells. Through ribosome profiling (a technique for the global footprinting of translating ribosomes), we also demonstrated translation of the uORF in representative members of the predominant human enterovirus species, namely Enterovirus A, B and C. In differentiated human intestinal organoids, uORF protein-knockout echoviruses are attenuated compared to the wild-type at late stages of infection where membrane-associated uORF protein facilitates virus release. Thus, we have identified a previously unknown enterovirus protein that facilitates virus growth in gut epithelial cells-the site of initial viral invasion into susceptible hosts. These findings overturn the 50-year-old dogma that enteroviruses use a single-polyprotein gene expression strategy and have important implications for the understanding of enterovirus pathogenesis.
Collapse
Affiliation(s)
- Valeria Lulla
- Division of Virology, Department of Pathology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
| | - Adam M Dinan
- Division of Virology, Department of Pathology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Myra Hosmillo
- Division of Virology, Department of Pathology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Yasmin Chaudhry
- Division of Virology, Department of Pathology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Lee Sherry
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Nerea Irigoyen
- Division of Virology, Department of Pathology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Komal M Nayak
- Department of Paediatrics, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Nicola J Stonehouse
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Matthias Zilbauer
- Department of Paediatrics, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Ian Goodfellow
- Division of Virology, Department of Pathology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Andrew E Firth
- Division of Virology, Department of Pathology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
| |
Collapse
|
23
|
Whole genome engineering by synthesis. SCIENCE CHINA-LIFE SCIENCES 2018; 61:1515-1527. [PMID: 30465231 DOI: 10.1007/s11427-018-9403-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 08/31/2018] [Indexed: 01/12/2023]
Abstract
Whole genome engineering is now feasible with the aid of genome editing and synthesis tools. Synthesizing a genome from scratch allows modifications of the genomic structure and function to an extent that was hitherto not possible, which will finally lead to new insights into the basic principles of life and enable valuable applications. With several recent genome synthesis projects as examples, the technical details to synthesize a genome and applications of synthetic genome are addressed in this perspective. A series of ongoing or future synthetic genomics projects, including the different genomes to be synthesized in GP-write, synthetic minimal genome, massively recoded genome, chimeric genome and synthetic genome with expanded genetic alphabet, are also discussed here with a special focus on theoretical and technical impediments in the design and synthesis process. Synthetic genomics will become a commonplace to engineer pathways and genomes according to arbitrary sets of design principles with the development of high-efficient, low-cost genome synthesis and assembly technologies.
Collapse
|
24
|
Cholleti H, Hayer J, Fafetine J, Berg M, Blomström AL. Genetic characterization of a novel picorna-like virus in Culex spp. mosquitoes from Mozambique. Virol J 2018; 15:71. [PMID: 29669586 PMCID: PMC5907373 DOI: 10.1186/s12985-018-0981-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/07/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Mosquitoes are the potential vectors for a variety of viruses that can cause diseases in the human and animal populations. Viruses in the order Picornavirales infect a broad range of hosts, including mosquitoes. In this study, we aimed to characterize a novel picorna-like virus from the Culex spp. of mosquitoes from the Zambezi Valley of Mozambique. METHODS The extracted RNA from mosquito pools was pre-amplified with the sequence independent single primer amplification (SISPA) method and subjected to high-throughput sequencing using the Ion Torrent platform. Reads that are classified as Iflaviridae, Picornaviridae and Dicistroviridae were assembled by CodonCode Aligner and SPAdes. Gaps between the viral contigs were sequenced by PCR. The genomic ends were analyzed by 5' and 3' RACE PCRs. The ORF was predicted with the NCBI ORF finder. The conserved domains were identified with ClustalW multiple sequence alignment, and a phylogenetic tree was built with MEGA. The presence of the virus in individual mosquito pools was detected by RT-PCR assay. RESULTS A near full-length viral genome (9740 nt) was obtained in Culex mosquitoes that encoded a complete ORF (3112 aa), named Culex picorna-like virus (CuPV-1). The predicted ORF had 38% similarity to the Hubei picorna-like virus 35. The sequence of the conserved domains, Helicase-Protease-RNA-dependent RNA polymerase, were identified by multiple sequence alignment and found to be at the 3' end, similar to iflaviruses. Phylogenetic analysis of the putative RdRP amino acid sequences indicated that the virus clustered with members of the Iflaviridae family. CuPV-1 was detected in both Culex and Mansonia individual pools with low infection rates. CONCLUSIONS The study reported a highly divergent, near full-length picorna-like virus genome from Culex spp. mosquitoes from Mozambique. The discovery and characterization of novel viruses in mosquitoes is an initial step, which will provide insights into mosquito-virus interaction mechanisms, genetic diversity and evolution.
Collapse
Affiliation(s)
- Harindranath Cholleti
- Section of Virology, Department of Biomedical and Veterinary Public Health, Box 7028, Swedish University of Agricultural Sciences (SLU), 75007, Uppsala, Sweden.
| | - Juliette Hayer
- SLU Global Bioinformatics Centre, Department of Animal Breeding and Genetics, Box 7023, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Jose Fafetine
- Division of Molecular Diagnostics and Epidemiology, Biotechnology Center, Eduardo Mondlane University, Maputo, Mozambique
| | - Mikael Berg
- Section of Virology, Department of Biomedical and Veterinary Public Health, Box 7028, Swedish University of Agricultural Sciences (SLU), 75007, Uppsala, Sweden
| | - Anne-Lie Blomström
- Section of Virology, Department of Biomedical and Veterinary Public Health, Box 7028, Swedish University of Agricultural Sciences (SLU), 75007, Uppsala, Sweden
| |
Collapse
|
25
|
Abstract
Reproduction of RNA viruses is typically error-prone due to the infidelity of their replicative machinery and the usual lack of proofreading mechanisms. The error rates may be close to those that kill the virus. Consequently, populations of RNA viruses are represented by heterogeneous sets of genomes with various levels of fitness. This is especially consequential when viruses encounter various bottlenecks and new infections are initiated by a single or few deviating genomes. Nevertheless, RNA viruses are able to maintain their identity by conservation of major functional elements. This conservatism stems from genetic robustness or mutational tolerance, which is largely due to the functional degeneracy of many protein and RNA elements as well as to negative selection. Another relevant mechanism is the capacity to restore fitness after genetic damages, also based on replicative infidelity. Conversely, error-prone replication is a major tool that ensures viral evolvability. The potential for changes in debilitated genomes is much higher in small populations, because in the absence of stronger competitors low-fit genomes have a choice of various trajectories to wander along fitness landscapes. Thus, low-fit populations are inherently unstable, and it may be said that to run ahead it is useful to stumble. In this report, focusing on picornaviruses and also considering data from other RNA viruses, we review the biological relevance and mechanisms of various alterations of viral RNA genomes as well as pathways and mechanisms of rehabilitation after loss of fitness. The relationships among mutational robustness, resilience, and evolvability of viral RNA genomes are discussed.
Collapse
|
26
|
Development and Characterization of an Infectious cDNA Clone of Equine Arteritis Virus. Methods Mol Biol 2018. [PMID: 28508211 DOI: 10.1007/978-1-4939-6964-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Development and characterization of several infectious cDNA clones of equine arteritis virus (EAV) have been described in the literature. Here we describe the assembly of the full-length infectious cDNA clone of the virulent Bucyrus strain (VBS; ATCC VR-796) of EAV in a plasmid vector. This system allows generation of infectious in vitro-transcribed (IVT) RNA from the linearized plasmid that can be transfected or electroporated into mammalian cells to produce infectious recombinant progeny virus. This is an efficient reverse genetics system that allows easy manipulation of EAV genomes to study molecular biology of the virus and pathogenesis of equine viral arteritis.
Collapse
|
27
|
Abstract
The discovery of a new class of pathogen, viruses, in the late 19th century, ushered in a period of study of the biochemical and structural properties of these entities in which plant viruses played a prominent role. This was, in large part, due to the relative ease with which sufficient quantities of material could be produced for such analyses. As analytical techniques became increasingly sensitive, similar studies could be performed on the viruses from other organisms. However, plant viruses continued to play an important role in the development of molecular biology, including the demonstration that RNA can be infectious, the determination of the genetic code, the mechanism by which viral RNAs are translated, and some of the early studies on gene silencing. Thus, the study of plant viruses should not be considered a "niche" subject but rather part of the mainstream of virology and molecular biology.
Collapse
|
28
|
Erickson AK, Jesudhasan PR, Mayer MJ, Narbad A, Winter SE, Pfeiffer JK. Bacteria Facilitate Enteric Virus Co-infection of Mammalian Cells and Promote Genetic Recombination. Cell Host Microbe 2018; 23:77-88.e5. [PMID: 29290575 PMCID: PMC5764776 DOI: 10.1016/j.chom.2017.11.007] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 08/25/2017] [Accepted: 11/15/2017] [Indexed: 01/12/2023]
Abstract
RNA viruses exist in genetically diverse populations due to high levels of mutations, many of which reduce viral fitness. Interestingly, intestinal bacteria can promote infection of several mammalian enteric RNA viruses, but the mechanisms and consequences are unclear. We screened a panel of 41 bacterial strains as a platform to determine how different bacteria impact infection of poliovirus, a model enteric virus. Most bacterial strains, including those extracted from cecal contents of mice, bound poliovirus, with each bacterium binding multiple virions. Certain bacterial strains increased viral co-infection of mammalian cells even at a low virus-to-host cell ratio. Bacteria-mediated viral co-infection correlated with bacterial adherence to cells. Importantly, bacterial strains that induced viral co-infection facilitated genetic recombination between two different viruses, thereby removing deleterious mutations and restoring viral fitness. Thus, bacteria-virus interactions may increase viral fitness through viral recombination at initial sites of infection, potentially limiting abortive infections.
Collapse
Affiliation(s)
- Andrea K Erickson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9048, USA
| | - Palmy R Jesudhasan
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9048, USA
| | - Melinda J Mayer
- Quadram Institute Bioscience, Norwich Research Park, Norwich, Norfolk NR4 7UA, UK
| | - Arjan Narbad
- Quadram Institute Bioscience, Norwich Research Park, Norwich, Norfolk NR4 7UA, UK
| | - Sebastian E Winter
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9048, USA
| | - Julie K Pfeiffer
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9048, USA.
| |
Collapse
|
29
|
Song Y, Gorbatsevych O, Liu Y, Mugavero J, Shen SH, Ward CB, Asare E, Jiang P, Paul AV, Mueller S, Wimmer E. Limits of variation, specific infectivity, and genome packaging of massively recoded poliovirus genomes. Proc Natl Acad Sci U S A 2017; 114:E8731-E8740. [PMID: 28973853 PMCID: PMC5642728 DOI: 10.1073/pnas.1714385114] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Computer design and chemical synthesis generated viable variants of poliovirus type 1 (PV1), whose ORF (6,189 nucleotides) carried up to 1,297 "Max" mutations (excess of overrepresented synonymous codon pairs) or up to 2,104 "SD" mutations (randomly scrambled synonymous codons). "Min" variants (excess of underrepresented synonymous codon pairs) are nonviable except for P2Min, a variant temperature-sensitive at 33 and 39.5 °C. Compared with WT PV1, P2Min displayed a vastly reduced specific infectivity (si) (WT, 1 PFU/118 particles vs. P2Min, 1 PFU/35,000 particles), a phenotype that will be discussed broadly. Si of haploid PV presents cellular infectivity of a single genotype. We performed a comprehensive analysis of sequence and structures of the PV genome to determine if evolutionary conserved cis-acting packaging signal(s) were preserved after recoding. We showed that conserved synonymous sites and/or local secondary structures that might play a role in determining packaging specificity do not survive codon pair recoding. This makes it unlikely that numerous "cryptic, sequence-degenerate, dispersed RNA packaging signals mapping along the entire viral genome" [Patel N, et al. (2017) Nat Microbiol 2:17098] play the critical role in poliovirus packaging specificity. Considering all available evidence, we propose a two-step assembly strategy for +ssRNA viruses: step I, acquisition of packaging specificity, either (a) by specific recognition between capsid protein(s) and replication proteins (poliovirus), or (b) by the high affinity interaction of a single RNA packaging signal (PS) with capsid protein(s) (most +ssRNA viruses so far studied); step II, cocondensation of genome/capsid precursors in which an array of hairpin structures plays a role in virion formation.
Collapse
Affiliation(s)
- Yutong Song
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, 11794;
| | - Oleksandr Gorbatsevych
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, 11794
| | - Ying Liu
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, 11794
- Pathology and Laboratory Medicine, Staten Island University Hospital, Staten Island, NY 10305
| | - JoAnn Mugavero
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, 11794
| | - Sam H Shen
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, 11794
- Department of Chemistry, University of Iowa, Iowa City, IA 52242
| | - Charles B Ward
- Google, Inc., Mountain View, CA 94043
- Department of Computer Science, Stony Brook University, Stony Brook, NY, 11794
| | - Emmanuel Asare
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, 11794
| | - Ping Jiang
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, 11794
| | - Aniko V Paul
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, 11794
| | - Steffen Mueller
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, 11794
- Codagenix Inc., Stony Brook, NY 11794
| | - Eckard Wimmer
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY, 11794;
| |
Collapse
|
30
|
Marsian J, Fox H, Bahar MW, Kotecha A, Fry EE, Stuart DI, Macadam AJ, Rowlands DJ, Lomonossoff GP. Plant-made polio type 3 stabilized VLPs-a candidate synthetic polio vaccine. Nat Commun 2017; 8:245. [PMID: 28811473 PMCID: PMC5557999 DOI: 10.1038/s41467-017-00090-w] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 05/31/2017] [Indexed: 11/24/2022] Open
Abstract
Poliovirus (PV) is the causative agent of poliomyelitis, a crippling human disease known since antiquity. PV occurs in two distinct antigenic forms, D and C, of which only the D form elicits a robust neutralizing response. Developing a synthetically produced stabilized virus-like particle (sVLP)-based vaccine with D antigenicity, without the drawbacks of current vaccines, will be a major step towards the final eradication of poliovirus. Such a sVLP would retain the native antigenic conformation and the repetitive structure of the original virus particle, but lack infectious genomic material. In this study, we report the production of synthetically stabilized PV VLPs in plants. Mice carrying the gene for the human PV receptor are protected from wild-type PV when immunized with the plant-made PV sVLPs. Structural analysis of the stabilized mutant at 3.6 Å resolution by cryo-electron microscopy and single-particle reconstruction reveals a structure almost indistinguishable from wild-type PV3.Despite the success of current vaccination against poliomyelitis, safe, cheap and effective vaccines remain sought for continuing eradication effort. Here the authors use plants to express stabilized virus-like particles of type 3 poliovirus that can induce a protective immune response in mice transgenic for the human poliovirus receptor.
Collapse
Affiliation(s)
- Johanna Marsian
- John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, UK
| | - Helen Fox
- The National Institute for Biological Standards and Control, Potters Bar, EN6 3QG, UK
| | - Mohammad W Bahar
- Division of Structural Biology, University of Oxford, The Henry Wellcome Building for Genomic Medicine, Headington, Oxford, OX3 7BN, UK
| | - Abhay Kotecha
- Division of Structural Biology, University of Oxford, The Henry Wellcome Building for Genomic Medicine, Headington, Oxford, OX3 7BN, UK
| | - Elizabeth E Fry
- Division of Structural Biology, University of Oxford, The Henry Wellcome Building for Genomic Medicine, Headington, Oxford, OX3 7BN, UK
| | - David I Stuart
- Division of Structural Biology, University of Oxford, The Henry Wellcome Building for Genomic Medicine, Headington, Oxford, OX3 7BN, UK
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK
| | - Andrew J Macadam
- The National Institute for Biological Standards and Control, Potters Bar, EN6 3QG, UK
| | - David J Rowlands
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | | |
Collapse
|
31
|
Dotti S, Lombardo T, Villa R, Cacciamali A, Zanotti C, Andreani NA, Cinotti S, Ferrari M. Transformation and Tumorigenicity Testing of Simian Cell Lines and Evaluation of Poliovirus Replication. PLoS One 2017; 12:e0169391. [PMID: 28046048 PMCID: PMC5207746 DOI: 10.1371/journal.pone.0169391] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 12/16/2016] [Indexed: 11/19/2022] Open
Abstract
The key role of cell cultures in different scientific fields is worldwide recognized, both as in vitro research models alternative to laboratory animals and substrates for biological production. However, many safety concerns rise from the use of animal/human cell lines that may be tumorigenic, leading to potential adverse contaminations in cell-derived biologicals. In order to evaluate the suitability of 13 different cell lines for Poliovirus vaccine production, safety and quality, in vitro/in vivo tumorigenicity and Poliovirus propagation properties were evaluated. Our results revealed that non-human primate cell lines CYNOM-K1, FRhK-4, 4MBr-5 and 4647 are free of tumorigenic features and represent highly susceptible substrates for attenuated Sabin Poliovirus strains. In particular, FRhK-4 and 4647 cell lines are characterized by a higher in vitro replication, resulting indicated for the use in large-scale production field.
Collapse
Affiliation(s)
- Silvia Dotti
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, Brescia, Italy
| | - Tina Lombardo
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, Brescia, Italy
| | - Riccardo Villa
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, Brescia, Italy
| | - Andrea Cacciamali
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, Brescia, Italy
| | - Cinzia Zanotti
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, Brescia, Italy
| | - Nadia Andrea Andreani
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, Brescia, Italy
| | - Stefano Cinotti
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, Brescia, Italy
| | - Maura Ferrari
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna, Brescia, Italy
| |
Collapse
|
32
|
Xiao Y, Rouzine IM, Bianco S, Acevedo A, Goldstein EF, Farkov M, Brodsky L, Andino R. RNA Recombination Enhances Adaptability and Is Required for Virus Spread and Virulence. Cell Host Microbe 2016; 19:493-503. [PMID: 27078068 DOI: 10.1016/j.chom.2016.03.009] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 02/13/2016] [Accepted: 03/25/2016] [Indexed: 10/21/2022]
Abstract
Mutation and recombination are central processes driving microbial evolution. A high mutation rate fuels adaptation but also generates deleterious mutations. Recombination between two different genomes may resolve this paradox, alleviating effects of clonal interference and purging deleterious mutations. Here we demonstrate that recombination significantly accelerates adaptation and evolution during acute virus infection. We identified a poliovirus recombination determinant within the virus polymerase, mutation of which reduces recombination rates without altering replication fidelity. By generating a panel of variants with distinct mutation rates and recombination ability, we demonstrate that recombination is essential to enrich the population in beneficial mutations and purge it from deleterious mutations. The concerted activities of mutation and recombination are key to virus spread and virulence in infected animals. These findings inform a mathematical model to demonstrate that poliovirus adapts most rapidly at an optimal mutation rate determined by the trade-off between selection and accumulation of detrimental mutations.
Collapse
Affiliation(s)
- Yinghong Xiao
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Igor M Rouzine
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Simone Bianco
- Department of Industrial and Applied Genomics, Accelerated Discovery Lab, IBM Almaden Research Center, 650 Harry Road, San Jose, CA 95120-6099, USA
| | - Ashley Acevedo
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Elizabeth Faul Goldstein
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Mikhail Farkov
- Tauber Bioinformatics Research Center and Department of Evolutionary and Environmental Biology, University of Haifa, Mount Carmel, Haifa 31905, Israel
| | - Leonid Brodsky
- Tauber Bioinformatics Research Center and Department of Evolutionary and Environmental Biology, University of Haifa, Mount Carmel, Haifa 31905, Israel
| | - Raul Andino
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
33
|
Yang YT, Nai YS, Lee SJ, Lee MR, Kim S, Kim JS. A novel picorna-like virus, Riptortus pedestris virus-1 (RiPV-1), found in the bean bug, R. pedestris, after fungal infection. J Invertebr Pathol 2016; 141:57-65. [PMID: 27840139 DOI: 10.1016/j.jip.2016.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 11/07/2016] [Accepted: 11/09/2016] [Indexed: 10/20/2022]
Abstract
A viral genome was assembled de novo from next-generation sequencing (NGS) data from bean bugs, Riptortus pedestris, infected with an entomopathogenic fungus, Beauveria bassiana (Bb), and was further confirmed via the RACE method. This is a novel insect positive-sense single-stranded RNA virus, which we named Riptortus pedestris virus-1 (RiPV-1) (GenBank accession no. KU958718). The genome of RiPV-1 consists of 10,554 nucleotides (nt), excluding the poly(A) tail, which contains a single large open reading frame (ORF) of 10,371 nt encoding a polyprotein (3456 aa) and flanked by 71 and 112 nt at the 5' and 3' untranslated regions (UTR), respectively. RiPV-1 genome organization from the 5' end contains a consensus organization of picorna-like RNA virus helicase, cysteine protease, and RNA-dependent RNA polymerase (RdRp), in addition to two putative structural proteins located at the 3' region and a poly(A) tail at the 3' end. The viral particles were approximately 30nm in diameter with some dispersal distinctive surface projections. Based on the phylogenetic analysis of the RdRp sequences, RiPV-1 was clustered in the unassigned insect RNA viruses with two other viruses, APV and KFV. These three viruses were suggested to constitute a new group of insect RNA viruses. RiPV-1 could be found in all stages of lab-reared bean bugs and was detected abundantly in the thorax, abdomen, midgut and fat body, but not in the reproductive organs and muscle. Interestingly, RiPV-1 replication was increased dramatically in bean bugs 2-6days after fungal infection. In conclusion, a novel insect RNA virus was found by NGS data assembly. This virus can provide further insight into the interaction between virus, fungus and the host.
Collapse
Affiliation(s)
- Yi-Ting Yang
- Department of Agricultural Biology, College of Agriculture & Life Sciences, Chonbuk National University, Jeonju, Republic of Korea
| | - Yu-Shin Nai
- Department of Agricultural Biology, College of Agriculture & Life Sciences, Chonbuk National University, Jeonju, Republic of Korea; Department of Biotechnology and Animal Science, National Ilan University, Ilan, Taiwan
| | - Se Jin Lee
- Department of Agricultural Biology, College of Agriculture & Life Sciences, Chonbuk National University, Jeonju, Republic of Korea
| | - Mi Rong Lee
- Department of Agricultural Biology, College of Agriculture & Life Sciences, Chonbuk National University, Jeonju, Republic of Korea
| | - Sihyeon Kim
- Department of Agricultural Biology, College of Agriculture & Life Sciences, Chonbuk National University, Jeonju, Republic of Korea
| | - Jae Su Kim
- Department of Agricultural Biology, College of Agriculture & Life Sciences, Chonbuk National University, Jeonju, Republic of Korea.
| |
Collapse
|
34
|
Abstract
Since the beginning of Global Polio Eradication Initiative in 1988, poliomyelitis cases caused by wild poliovirus (PV) have been drastically reduced, with only 74 cases reported in 2 endemic countries in 2015. The current limited PV transmission suggests that we are in the endgame of the polio eradication program. However, specific challenges have emerged in the endgame, including tight budget, switching of the vaccines, and changes in biorisk management of PV. To overcome these challenges, several PV studies have been implemented in the eradication program. Some of the responses to the emerging challenges in the polio endgame might be valuable in other infectious diseases eradication programs. Here, I will review challenges that confront the polio eradication program and current research to address these challenges.
Collapse
Affiliation(s)
- Minetaro Arita
- Department of Virology II, National Institute of Infectious Diseases
| |
Collapse
|
35
|
Tan CW, Tee HK, Lee MHP, Sam IC, Chan YF. Enterovirus A71 DNA-Launched Infectious Clone as a Robust Reverse Genetic Tool. PLoS One 2016; 11:e0162771. [PMID: 27617744 PMCID: PMC5019408 DOI: 10.1371/journal.pone.0162771] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 08/09/2016] [Indexed: 12/28/2022] Open
Abstract
Enterovirus A71 (EV-A71) causes major outbreaks of hand, foot and mouth disease, and is occasionally associated with neurological complications and death in children. Reverse genetics is widely used in the field of virology for functional study of viral genes. For EV-A71, such tools are limited to clones that are transcriptionally controlled by T7/SP6 bacteriophage promoter. This is often time-consuming and expensive. Here, we describe the development of infectious plasmid DNA-based EV-A71 clones, for which EV-A71 genome expression is under transcriptional control by the CMV-intermediate early promoter and SV40 transcriptional-termination signal. Transfection of this EV-A71 infectious DNA produces good virus yield similar to in vitro-transcribed EV-A71 infectious RNA, 6.4 and 5.8 log10PFU/ml, respectively. Infectious plasmid with enhanced green fluorescence protein and Nano luciferase reporter genes also produced good virus titers, with 4.3 and 5.0 log10 PFU/ml, respectively. Another infectious plasmid with both CMV and T7 promoters was also developed for easy manipulation of in vitro transcription or direct plasmid transfection. Transfection with either dual-promoter infectious plasmid DNA or infectious RNA derived from this dual-promoter clone produced infectious viral particles. Incorporation of hepatitis delta virus ribozyme, which yields precise 3’ ends of the DNA-launched EV-A71 genomic transcripts, increased infectious viral production. In contrast, the incorporation of hammerhead ribozyme in the DNA-launched EV-A71 resulted in lower virus yield, but improved the virus titers for T7 promoter-derived infectious RNA. This study describes rapid and robust reverse genetic tools for EV-A71.
Collapse
Affiliation(s)
- Chee Wah Tan
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- * E-mail: (CWT); (YFC)
| | - Han Kang Tee
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Michelle Hui Pheng Lee
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - I-Ching Sam
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Yoke Fun Chan
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- * E-mail: (CWT); (YFC)
| |
Collapse
|
36
|
Fikatas A, Dimitriou TG, Kyriakopoulou Z, Tsachouridou O, Gartzonika C, Levidiotou-Stefanou S, Amoutzias GD, Markoulatos P. Serum Neutralization Assay for the Determination of Antibody Levels Against Non-Polio Enterovirus Strains in Central and Western Greece. Viral Immunol 2016; 29:444-50. [PMID: 27410516 DOI: 10.1089/vim.2016.0049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mutations and recombination events have been identified in enteroviruses. Point mutations accumulate with a frequency of 6.3 × 10(-4) per base pair per replication cycle affecting the fitness, the circulation, and the infectivity of enteroviral strains. In the present report, the serological status of the Central and Western Greek population (Larissa and Ioannina, respectively) in the 1-10-year, 11-20-year, 21-30-year, and 31-40-year age groups against six non-polio enterovirus strains, their respective echovirus prototypes, and Sabin 1, 2, and 3 vaccine strains was evaluated, through serum-neutralization assay. In the Western Greek population, antibody levels were detected only for clinical isolates of E30 serotype in all age groups, and for environmental isolate LR61G3 (E6 serotype) only in the 31-40 age group, whereas an immunity level was observed in the Central Greek population, against all strains, except for EIS6B (E3 serotype). Amino acid substitutions were encountered across the structural region of the capsid, between the prototypes and the respective isolates. These substitutions may alter the antigenicity of each strain and may explain the variations observed in the neutralization titers of the different strains. As a consequence, these substitutions severely affect antibody binding and increase the ability of the virus to escape the immune response. It is tempting to assume that changes in the antigenic properties observed in circulating echoviruses represent a selection of viral variants that are less prone to be neutralized by human antibodies. These facts argue for the need of immunological studies to the population to avoid epidemics due to the circulation of highly evolved derivatives.
Collapse
Affiliation(s)
- Antonis Fikatas
- 1 Microbiology-Virology Laboratory, Department of Biochemistry & Biotechnology, School of Health Sciences, University of Thessaly , Larissa, Greece
| | - Tilemachos G Dimitriou
- 1 Microbiology-Virology Laboratory, Department of Biochemistry & Biotechnology, School of Health Sciences, University of Thessaly , Larissa, Greece
| | - Zaharoula Kyriakopoulou
- 1 Microbiology-Virology Laboratory, Department of Biochemistry & Biotechnology, School of Health Sciences, University of Thessaly , Larissa, Greece
| | - Ourania Tsachouridou
- 1 Microbiology-Virology Laboratory, Department of Biochemistry & Biotechnology, School of Health Sciences, University of Thessaly , Larissa, Greece
| | | | | | - Grigoris D Amoutzias
- 1 Microbiology-Virology Laboratory, Department of Biochemistry & Biotechnology, School of Health Sciences, University of Thessaly , Larissa, Greece
| | - Panayotis Markoulatos
- 1 Microbiology-Virology Laboratory, Department of Biochemistry & Biotechnology, School of Health Sciences, University of Thessaly , Larissa, Greece
| |
Collapse
|
37
|
Abstract
Self-replicating RNA derived from the genomes of positive strand RNA viruses represents a powerful tool for both molecular studies on virus biology and approaches to novel safe and effective vaccines. The following chapter summarizes the principles how such RNAs can be established and used for design of vaccines. Due to the large variety of strategies needed to circumvent specific pitfalls in the design of such constructs the technical details of the experiments are not described here but can be found in the cited literature.
Collapse
|
38
|
Abstract
UNLABELLED Tilapia are an important global food source due to their omnivorous diet, tolerance for high-density aquaculture, and relative disease resistance. Since 2009, tilapia aquaculture has been threatened by mass die-offs in farmed fish in Israel and Ecuador. Here we report evidence implicating a novel orthomyxo-like virus in these outbreaks. The tilapia lake virus (TiLV) has a 10-segment, negative-sense RNA genome. The largest segment, segment 1, contains an open reading frame with weak sequence homology to the influenza C virus PB1 subunit. The other nine segments showed no homology to other viruses but have conserved, complementary sequences at their 5' and 3' termini, consistent with the genome organization found in other orthomyxoviruses. In situ hybridization indicates TiLV replication and transcription at sites of pathology in the liver and central nervous system of tilapia with disease. IMPORTANCE The economic impact of worldwide trade in tilapia is estimated at $7.5 billion U.S. dollars (USD) annually. The infectious agent implicated in mass tilapia die-offs in two continents poses a threat to the global tilapia industry, which not only provides inexpensive dietary protein but also is a major employer in the developing world. Here we report characterization of the causative agent as a novel orthomyxo-like virus, tilapia lake virus (TiLV). We also describe complete genomic and protein sequences that will facilitate TiLV detection and containment and enable vaccine development.
Collapse
|
39
|
Zheng X, Tong W, Liu F, Liang C, Gao F, Li G, Tong G, Zheng H. Genetic instability of Japanese encephalitis virus cDNA clones propagated in Escherichia coli. Virus Genes 2016; 52:195-203. [PMID: 26888374 DOI: 10.1007/s11262-016-1289-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 01/05/2016] [Indexed: 10/22/2022]
Abstract
The genetic instability of Flavivirus cDNA clones in transformed bacteria is a common phenomenon. Herein, a cDNA fragment of the nucleotide (nt) 1-2913 of the genome of a flavivirus, Japanese encephalitis virus (JEV), was used to investigate factors that caused the instability of cDNA clones. Several cDNA fragments with different 5'- or 3'-termini of the 2913-nt cDNA were obtained by PCR amplification or restriction enzyme digestion and cloned into a pCR-Blunt II-TOPO vector. All the cDNA fragments were stably propagated at 25 °C. However, the 5'-untranslated region and half of the 3'-E gene could cause the instability of the 2913-nt cDNA at 37 °C. The 5'-terminus sequences of the 2913-nt fragment were subjected to testing of the prokaryotic promoter activity by luciferase assay and Western blot. The sequences of 54-120 nt of the JEV genome exhibited high prokaryotic promoter activity at 37 °C, and the activity declined markedly at 25 °C. These findings revealed that the high prokaryotic promoter activity of the 54-120 nt sequences of the JEV genome together with expression of JEV structural genes determined the instability of a JEV cDNA clone. Growth at room temperature may reduce the prokaryotic promoter activity of 5'-sequences of the JEV genome and could represent an effective way to improve the stability of flavivirus cDNA clones in host bacteria.
Collapse
Affiliation(s)
- Xuchen Zheng
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518 Ziyue Road, Shanghai, 200241, China
| | - Wu Tong
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518 Ziyue Road, Shanghai, 200241, China
| | - Fei Liu
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518 Ziyue Road, Shanghai, 200241, China
| | - Chao Liang
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518 Ziyue Road, Shanghai, 200241, China
| | - Fei Gao
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518 Ziyue Road, Shanghai, 200241, China
| | - Guoxin Li
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518 Ziyue Road, Shanghai, 200241, China
| | - Guangzhi Tong
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518 Ziyue Road, Shanghai, 200241, China.,Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China
| | - Hao Zheng
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518 Ziyue Road, Shanghai, 200241, China. .,Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China.
| |
Collapse
|
40
|
Tsun A, Miao XN, Wang CM, Yu DC. Oncolytic Immunotherapy for Treatment of Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 909:241-83. [PMID: 27240460 DOI: 10.1007/978-94-017-7555-7_5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Immunotherapy entails the treatment of disease by modulation of the immune system. As detailed in the previous chapters, the different modes of achieving immune modulation are many, including the use of small/large molecules, cellular therapy, and radiation. Oncolytic viruses that can specifically attack, replicate within, and destroy tumors represent one of the most promising classes of agents for cancer immunotherapy (recently termed as oncolytic immunotherapy). The notion of oncolytic immunotherapy is considered as the way in which virus-induced tumor cell death (known as immunogenic cancer cell death (ICD)) allows the immune system to recognize tumor cells and provide long-lasting antitumor immunity. Both immune responses toward the virus and ICD together contribute toward successful antitumor efficacy. What is now becoming increasingly clear is that monotherapies, through any of the modalities detailed in this book, are neither sufficient in eradicating tumors nor in providing long-lasting antitumor immune responses and that combination therapies may deliver enhanced efficacy. After the rise of the genetic engineering era, it has been possible to engineer viruses to harbor combination-like characteristics to enhance their potency in cancer immunotherapy. This chapter provides a historical background on oncolytic virotherapy and its future application in cancer immunotherapy, especially as a combination therapy with other treatment modalities.
Collapse
Affiliation(s)
- A Tsun
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China
| | - X N Miao
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China
| | - C M Wang
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China
| | - D C Yu
- Innovent Biologics, Inc., 168 Dongping Street, Suzhou Industrial Park, 215123, China.
| |
Collapse
|
41
|
Applying the Concept of Peptide Uniqueness to Anti-Polio Vaccination. J Immunol Res 2015; 2015:541282. [PMID: 26568962 PMCID: PMC4629041 DOI: 10.1155/2015/541282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 04/28/2015] [Indexed: 11/21/2022] Open
Abstract
Background. Although rare, adverse events may associate with anti-poliovirus vaccination thus possibly hampering global polio eradication worldwide. Objective. To design peptide-based anti-polio vaccines exempt from potential cross-reactivity risks and possibly able to reduce rare potential adverse events such as the postvaccine paralytic poliomyelitis due to the tendency of the poliovirus genome to mutate. Methods. Proteins from poliovirus type 1, strain Mahoney, were analyzed for amino acid sequence identity to the human proteome at the pentapeptide level, searching for sequences that (1) have zero percent of identity to human proteins, (2) are potentially endowed with an immunologic potential, and (3) are highly conserved among poliovirus strains. Results. Sequence analyses produced a set of consensus epitopic peptides potentially able to generate specific anti-polio immune responses exempt from cross-reactivity with the human host. Conclusion. Peptide sequences unique to poliovirus proteins and conserved among polio strains might help formulate a specific and universal anti-polio vaccine able to react with multiple viral strains and exempt from the burden of possible cross-reactions with human proteins. As an additional advantage, using a peptide-based vaccine instead of current anti-polio DNA vaccines would eliminate the rare post-polio poliomyelitis cases and other disabling symptoms that may appear following vaccination.
Collapse
|
42
|
|
43
|
Pfaller CK, Cattaneo R, Schnell MJ. Reverse genetics of Mononegavirales: How they work, new vaccines, and new cancer therapeutics. Virology 2015; 479-480:331-44. [PMID: 25702088 DOI: 10.1016/j.virol.2015.01.029] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 01/26/2015] [Accepted: 01/30/2015] [Indexed: 12/24/2022]
Abstract
The order Mononegavirales includes five families: Bornaviridae, Filoviridae, Nyamaviridae, Paramyxoviridae, and Rhabdoviridae. The genome of these viruses is one molecule of negative-sense single strand RNA coding for five to ten genes in a conserved order. The RNA is not infectious until packaged by the nucleocapsid protein and transcribed by the polymerase and co-factors. Reverse genetics approaches have answered fundamental questions about the biology of Mononegavirales. The lack of icosahedral symmetry and modular organization in the genome of these viruses has facilitated engineering of viruses expressing fluorescent proteins, and these fluorescent proteins have provided important insights about the molecular and cellular basis of tissue tropism and pathogenesis. Studies have assessed the relevance for virulence of different receptors and the interactions with cellular proteins governing the innate immune responses. Research has also analyzed the mechanisms of attenuation. Based on these findings, ongoing clinical trials are exploring new live attenuated vaccines and the use of viruses re-engineered as cancer therapeutics.
Collapse
Affiliation(s)
| | - Roberto Cattaneo
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | - Matthias J Schnell
- Department of Microbiology and Immunology, Philadelphia, PA 19107, USA; Jefferson Vaccine Center, Jefferson Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
44
|
García-Sánchez JE, García-Sánchez E, García-Merino E, Fresnadillo-Martínez MJ. [Polio, the long walk to the endgame]. Enferm Infecc Microbiol Clin 2015; 33:e69-78. [PMID: 25595690 DOI: 10.1016/j.eimc.2014.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 09/17/2014] [Accepted: 10/01/2014] [Indexed: 11/18/2022]
Abstract
Although the WHO original target date for the global eradication of poliomyelitis was the year 2000 -thanks to vaccination and institutional, public and private, resources for that purpose-, in 2013 the disease remained endemic in three countries, Afghanistan, Pakistan and Nigeria, and some cases were described in five others. The circulation of wild type 1 poliovirus in Israel, Gaza and the West Bank and the cases in Syria were a wakeup call, as at that time there were polioviruses derived from the oral vaccine that are still circulating among the human population and can cause the development of the disease. Travelling "from" and "to" endemic areas are factors to consider in poliovirus exportation and in its spread when it reaches areas with poor immunogenicity. Wars, terrorism, intolerance, lack of culture and proliferation of anti-vaccine groups and the rise of the anti-vaccination movement are important factors in the maintenance and expansion of the virus and in the "non-vaccination" against it. Based on the international situation to date, the Emergency Committee of WHO met in May 2014 to address the problem. It is still necessary to enhance the knowledge of the disease and its agent. In the first case to perform a differential diagnosis of flaccid paralysis and to continue vaccination programs, and in the second case to keep studying and looking for the poliovirus in environmental samples, which is a model for the study of many other viruses.
Collapse
Affiliation(s)
- José Elías García-Sánchez
- Departamento de Medicina Preventiva, Salud Pública y Microbiología Médica, Facultad de Medicina, Universidad de Salamanca, Salamanca, España.
| | - Enrique García-Sánchez
- Departamento de Medicina Preventiva, Salud Pública y Microbiología Médica, Facultad de Medicina, Universidad de Salamanca, Salamanca, España
| | | | - María José Fresnadillo-Martínez
- Departamento de Medicina Preventiva, Salud Pública y Microbiología Médica, Facultad de Medicina, Universidad de Salamanca, Salamanca, España
| |
Collapse
|
45
|
Abstract
The historical classification of human rhinoviruses (RV) by serotyping has been replaced by a logical system of comparative sequencing. Given that strains must diverge within their capsid sequenced by a reasonable degree (>12-13 % pairwise base identities) before becoming immunologically distinct, the new nomenclature system makes allowances for the addition of new, future types, without compromising historical designations. Currently, three species, the RV-A, RV-B, and RV-C, are recognized. Of these, the RV-C, discovered in 2006, are the most unusual in terms of capsid structure, receptor use, and association with severe disease in children.
Collapse
Affiliation(s)
- Ann C Palmenberg
- Institute for Molecular Virology, University of Wisconsin-Madison, 1525 Linden Drive, Madison, WI, 53706, USA,
| | | |
Collapse
|
46
|
Flavivirus reverse genetic systems, construction techniques and applications: a historical perspective. Antiviral Res 2014; 114:67-85. [PMID: 25512228 PMCID: PMC7173292 DOI: 10.1016/j.antiviral.2014.12.007] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 11/26/2014] [Accepted: 12/03/2014] [Indexed: 12/20/2022]
Abstract
The study of flaviviruses, which cause some of the most important emerging tropical and sub-tropical human arbovirus diseases, has greatly benefited from the use of reverse genetic systems since its first development for yellow fever virus in 1989. Reverse genetics technology has completely revolutionized the study of these viruses, making it possible to manipulate their genomes and evaluate the direct effects of these changes on their biology and pathogenesis. The most commonly used reverse genetics system is the infectious clone technology. Whilst flavivirus infectious clones provide a powerful tool, their construction as full-length cDNA molecules in bacterial vectors can be problematic, laborious and time consuming, because they are often unstable, contain unwanted induced substitutions and may be toxic for bacteria due to viral protein expression. The incredible technological advances that have been made during the past 30years, such as the use of PCR or new sequencing methods, have allowed the development of new approaches to improve preexisting systems or elaborate new strategies that overcome these problems. This review summarizes the evolution and major technical breakthroughs in the development of flavivirus reverse genetics technologies and their application to the further understanding and control of these viruses and their diseases.
Collapse
|
47
|
Abstract
Human rhinovirus (HRV) contains a 7.2 kb messenger-sense RNA genome which is the template for reproducing progeny viruses after it enters the cytoplasm of a host cell. Reverse genetics refers to the regeneration of progeny viruses from an artificial cDNA copy of the RNA genome of an RNA virus. It has been a powerful molecular genetic tool for studying HRV and other RNA viruses because the artificial DNA stage makes it practical to introduce specific mutations into the viral RNA genome. This chapter uses HRV-16 as the model virus to illustrate the strategy and methods for constructing and cloning the artificial cDNA copy of a full-length HRV genome, identifying the infectious cDNA clone isolates, and selecting the most vigorous cDNA clone isolate to serve as the standard parental clone for future molecular genetic study of the virus.
Collapse
|
48
|
Balasuriya UBR, Zhang J, Go YY, MacLachlan NJ. Experiences with infectious cDNA clones of equine arteritis virus: lessons learned and insights gained. Virology 2014; 462-463:388-403. [PMID: 24913633 PMCID: PMC7172799 DOI: 10.1016/j.virol.2014.04.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 04/16/2014] [Accepted: 04/22/2014] [Indexed: 12/19/2022]
Abstract
The advent of recombinant DNA technology, development of infectious cDNA clones of RNA viruses, and reverse genetic technologies have revolutionized how viruses are studied. Genetic manipulation of full-length cDNA clones has become an especially important and widely used tool to study the biology, pathogenesis, and virulence determinants of both positive and negative stranded RNA viruses. The first full-length infectious cDNA clone of equine arteritis virus (EAV) was developed in 1996 and was also the first full-length infectious cDNA clone constructed from a member of the order Nidovirales. This clone was extensively used to characterize the molecular biology of EAV and other Nidoviruses. The objective of this review is to summarize the characterization of the virulence (or attenuation) phenotype of the recombinant viruses derived from several infectious cDNA clones of EAV in horses, as well as their application for characterization of the molecular basis of viral neutralization, persistence, and cellular tropism.
Collapse
Affiliation(s)
- Udeni B R Balasuriya
- 108 Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40546, USA.
| | - Jianqiang Zhang
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Yun Young Go
- Virus Research and Testing Group, Division of Drug Discovery Research, Korea Research Institute of Chemical Technology, Daejeon 305-343, South Korea
| | - N James MacLachlan
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| |
Collapse
|
49
|
Lau SKP, Woo PCY, Yip CCY, Li KSM, Fan RYY, Bai R, Huang Y, Chan KH, Yuen KY. Chickens host diverse picornaviruses originated from potential interspecies transmission with recombination. J Gen Virol 2014; 95:1929-1944. [PMID: 24906980 DOI: 10.1099/vir.0.066597-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
While chickens are an important reservoir for emerging pathogens such as avian influenza viruses, little is known about the diversity of picornaviruses in poultry. We discovered a previously unknown diversity of picornaviruses in chickens in Hong Kong. Picornaviruses were detected in 87 cloacal and 7 tracheal samples from 93 of 900 chickens by reverse transcription-PCR, with their partial 3D(pol) gene sequences forming five distinct clades (I to V) among known picornaviruses. Analysis of eight genomes from different clades revealed seven different picornaviruses, including six novel picornavirus species (ChPV1 from clade I, ChPV2 and ChPV3 from clade II, ChPV4 and ChPV5 from clade III, ChGV1 from clade IV) and one existing species (Avian encephalomyelitis virus from clade V). The six novel chicken picornavirus genomes exhibited distinct phylogenetic positions and genome features different from related picornaviruses, supporting their classification as separate species. Moreover, ChPV1 may potentially belong to a novel genus, with low sequence homologies to related picornaviruses, especially in the P1 and P2 regions, including the predicted L and 2A proteins. Nevertheless, these novel picornaviruses were most closely related to picornaviruses of other avian species (ChPV1 related to Passerivirus A, ChPV2 and ChPV3 to Avisivirus A and Duck hepatitis A virus, ChPV4 and ChPV5 to Melegrivirus A, ChGV1 to Gallivirus A). Furthermore, ChPV5 represented a potential recombinant picornavirus, with its P2 and P3 regions possibly originating from Melegrivirus A. Chickens are an important reservoir for diverse picornaviruses that may cross avian species barriers through mutation or recombination.
Collapse
Affiliation(s)
- Susanna K P Lau
- Carol Yu Centre for Infection, University of Hong Kong, Hong Kong, PR China.,State Key Laboratory of Emerging Infectious Diseases, University of Hong Kong, Hong Kong, PR China.,Department of Microbiology, University of Hong Kong, Hong Kong, PR China.,Research Centre of Infection and Immunology, University of Hong Kong, Hong Kong, PR China
| | - Patrick C Y Woo
- Research Centre of Infection and Immunology, University of Hong Kong, Hong Kong, PR China.,Carol Yu Centre for Infection, University of Hong Kong, Hong Kong, PR China.,State Key Laboratory of Emerging Infectious Diseases, University of Hong Kong, Hong Kong, PR China.,Department of Microbiology, University of Hong Kong, Hong Kong, PR China
| | - Cyril C Y Yip
- Department of Microbiology, University of Hong Kong, Hong Kong, PR China
| | - Kenneth S M Li
- Department of Microbiology, University of Hong Kong, Hong Kong, PR China
| | - Rachel Y Y Fan
- Department of Microbiology, University of Hong Kong, Hong Kong, PR China
| | - Ru Bai
- Department of Microbiology, University of Hong Kong, Hong Kong, PR China
| | - Yi Huang
- Department of Microbiology, University of Hong Kong, Hong Kong, PR China
| | - Kwok-Hung Chan
- Department of Microbiology, University of Hong Kong, Hong Kong, PR China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, University of Hong Kong, Hong Kong, PR China.,Research Centre of Infection and Immunology, University of Hong Kong, Hong Kong, PR China.,Carol Yu Centre for Infection, University of Hong Kong, Hong Kong, PR China.,Department of Microbiology, University of Hong Kong, Hong Kong, PR China
| |
Collapse
|
50
|
Kanduc D, Fasano C, Bavaro SL, Novello G, Lucchese G, Capone G. Peptide profiling of the route from Mahoney to Sabin, and return. J Basic Microbiol 2014; 54:369-77. [DOI: 10.1002/jobm.201200642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 02/07/2013] [Indexed: 11/05/2022]
Affiliation(s)
- Darja Kanduc
- Department of Biosciences, Biotechnologies and Pharmacological Sciences; University of Bari; Bari Italy
| | - Candida Fasano
- Department of Biosciences, Biotechnologies and Pharmacological Sciences; University of Bari; Bari Italy
| | - Simona Lucia Bavaro
- Department of Biosciences, Biotechnologies and Pharmacological Sciences; University of Bari; Bari Italy
| | - Giuseppe Novello
- Department of Biosciences, Biotechnologies and Pharmacological Sciences; University of Bari; Bari Italy
| | - Guglielmo Lucchese
- Department of Biosciences, Biotechnologies and Pharmacological Sciences; University of Bari; Bari Italy
| | - Giovanni Capone
- Department of Biosciences, Biotechnologies and Pharmacological Sciences; University of Bari; Bari Italy
| |
Collapse
|