1
|
Haller SL, Park C, Bruneau RC, Megawati D, Zhang C, Vipat S, Peng C, Senkevich TG, Brennan G, Tazi L, Rothenburg S. Host species-specific activity of the poxvirus PKR inhibitors E3 and K3 mediate host range function. J Virol 2024; 98:e0133124. [PMID: 39480085 PMCID: PMC11575334 DOI: 10.1128/jvi.01331-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/24/2024] [Indexed: 11/02/2024] Open
Abstract
The antiviral protein kinase R (PKR) is activated by viral double-stranded RNA and phosphorylates translation initiation factor eIF2α, thereby inhibiting translation and virus replication. Most poxviruses contain two PKR inhibitors, called E3 and K3 in vaccinia virus (VACV), which are determinants of viral host range. The prevailing model for E3 function is that it inhibits PKR through the non-specific sequestration of double-stranded (ds) RNA. Our data revealed that Syrian hamster PKR was resistant to E3, which is at odds with the sequestration model. However, Syrian hamster PKR was still sensitive to K3 inhibition. In contrast, Armenian hamster PKR showed opposite sensitivities, being sensitive to E3 and resistant to K3 inhibition. Mutational analyses of hamster PKRs showed that sensitivity to E3 inhibition was largely determined by the region linking the dsRNA-binding domains and the kinase domain of PKR, whereas two amino acid residues in the kinase domain (helix αG) determined sensitivity to K3. The expression of PKRs in congenic cells showed that Syrian hamster PKR containing the two Armenian hamster PKR residues in helix αG was resistant to wild-type VACV infection and that cells expressing either hamster PKR recapitulated the phenotypes observed in species-derived cell lines. The observed resistance of Syrian hamster PKR to E3 explains its host range function and challenges the paradigm that dsRNA-binding PKR inhibitors mainly act by the sequestration of dsRNA.IMPORTANCEThe molecular mechanisms that govern the host range of viruses are incompletely understood. We show that the host range functions of E3 and K3, two host range factors from vaccinia virus, are a result of species-specific interactions with the antiviral protein kinase R (PKR) and that PKR from closely related species displayed dramatic differences in their sensitivities to these viral inhibitors. The current model for E3-mediated PKR inhibition is that E3 non-specifically sequesters double-stranded (ds) RNA to prevent PKR activation. This model does not predict species-specific sensitivity to E3; therefore, our data suggest that the current model is incomplete and that dsRNA sequestration is not the primary mechanism for E3 activity.
Collapse
Affiliation(s)
- Sherry L Haller
- Division of Biology, Kansas State University, Manhattan, Kansas, USA
| | - Chorong Park
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, California, USA
| | - Ryan C Bruneau
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, California, USA
| | - Dewi Megawati
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, California, USA
| | - Chi Zhang
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, California, USA
| | - Sameera Vipat
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, California, USA
| | - Chen Peng
- Division of Biology, Kansas State University, Manhattan, Kansas, USA
| | - Tatiana G Senkevich
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Greg Brennan
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, California, USA
| | - Loubna Tazi
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, California, USA
| | - Stefan Rothenburg
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, California, USA
| |
Collapse
|
2
|
Haller SL, Park C, Bruneau RC, Megawati D, Zhang C, Vipat S, Peng C, Senkevich TG, Brennan G, Tazi L, Rothenburg S. Molecular basis for the host range function of the poxvirus PKR inhibitor E3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.16.594589. [PMID: 38798513 PMCID: PMC11118487 DOI: 10.1101/2024.05.16.594589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The antiviral protein kinase R (PKR) is activated by viral double-stranded RNA and phosphorylates translation initiation factor eIF2α, thereby inhibiting translation and virus replication. Most poxviruses contain two PKR inhibitors, called E3 and K3 in vaccinia virus (VACV), which are determinants of viral host range. The prevailing model for E3 function is that it inhibits PKR through the non-specific sequestration of double-stranded (ds) RNA. Our data revealed that Syrian hamster PKR was resistant to E3, which is at odds with the sequestration model. However, Syrian hamster PKR was still sensitive to K3 inhibition. In contrast, Armenian hamster PKR showed opposite sensitivities, being sensitive to E3 and resistant to K3 inhibition. Mutational analyses of hamster PKRs showed that sensitivity to E3 inhibition was largely determined by the region linking the dsRNA-binding domains and the kinase domain of PKR, whereas two amino acid residues in the kinase domain (helix αG) determined sensitivity to K3. Expression of PKRs in congenic cells showed that Syrian hamster PKR containing the two Armenian hamster PKR residues in helix-αG was resistant to wild type VACV infection, and that cells expressing either hamster PKR recapitulated the phenotypes observed in species-derived cell lines. The observed resistance of Syrian hamster PKR to E3 explains its host range function and challenges the paradigm that dsRNA-binding PKR inhibitors mainly act by the sequestration of dsRNA. Significance The molecular mechanisms that govern the host range of viruses are incompletely understood. A small number of poxvirus genes have been identified that influence the host range of poxviruses. We show that the host range functions of E3 and K3, two host range factors from vaccinia virus, are a result of species-specific interactions with the antiviral protein kinase R (PKR) and that PKR from closely related species displayed dramatic differences in their sensitivities to these viral inhibitors. While there is a substantial body of work demonstrating host-specific interactions with K3, the current model for E3-mediated PKR inhibition is that E3 non-specifically sequesters dsRNA to prevent PKR activation. This model does not predict species-specific sensitivity to E3; therefore, our data suggest that the current model is incomplete, and that dsRNA sequestration is not the primary mechanism for E3 activity.
Collapse
|
3
|
Yang CH, Song AL, Qiu Y, Ge XY. Cross-species transmission and host range genes in poxviruses. Virol Sin 2024; 39:177-193. [PMID: 38272237 PMCID: PMC11074647 DOI: 10.1016/j.virs.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
The persistent epidemic of human mpox, caused by mpox virus (MPXV), raises concerns about the future spread of MPXV and other poxviruses. MPXV is a typical zoonotic virus which can infect human and cause smallpox-like symptoms. MPXV belongs to the Poxviridae family, which has a relatively broad host range from arthropods to vertebrates. Cross-species transmission of poxviruses among different hosts has been frequently reported and resulted in numerous epidemics. Poxviruses have a complex linear double-strand DNA genome that encodes hundreds of proteins. Genes related to the host range of poxvirus are called host range genes (HRGs). This review briefly introduces the taxonomy, phylogeny and hosts of poxviruses, and then comprehensively summarizes the current knowledge about the cross-species transmission of poxviruses. In particular, the HRGs of poxvirus are described and their impacts on viral host range are discussed in depth. We hope that this review will provide a comprehensive perspective about the current progress of researches on cross-species transmission and HRG variation of poxviruses, serving as a valuable reference for academic studies and disease control in the future.
Collapse
Affiliation(s)
- Chen-Hui Yang
- College of Biology, Hunan Provincial Key Laboratory of Medical Virology, Hunan University, Changsha, 410012, China
| | - A-Ling Song
- College of Biology, Hunan Provincial Key Laboratory of Medical Virology, Hunan University, Changsha, 410012, China
| | - Ye Qiu
- College of Biology, Hunan Provincial Key Laboratory of Medical Virology, Hunan University, Changsha, 410012, China.
| | - Xing-Yi Ge
- College of Biology, Hunan Provincial Key Laboratory of Medical Virology, Hunan University, Changsha, 410012, China.
| |
Collapse
|
4
|
Subissi L, Stefanelli P, Rezza G. Human mpox: global trends, molecular epidemiology and options for vaccination. Pathog Glob Health 2024; 118:25-32. [PMID: 37715739 PMCID: PMC10769137 DOI: 10.1080/20477724.2023.2258641] [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: 09/18/2023] Open
Abstract
The eradication of smallpox and the cessation of vaccination have led to the growth of the susceptible human population to poxviruses. This has led to the increasing detection of zoonotic orthopoxviruses. Among those viruses, monkeypox virus (MPV) is the most commonly detected in Western and Central African regions. Since 2022, MPV is causing local transmission in newly affected countries all over the world. While the virus causing the current outbreak remains part of clade II (historically referred to as West African clade), it has a significant number of mutations as compared to other clade II sequences and is therefore referred to as clade IIb. It remains unclear whether those mutations may have caused a change in the virus phenotype. Vaccine effectiveness data show evidence of a high cross-protection of vaccines designed to prevent smallpox against mpox. These vaccines therefore represent a great opportunity to control human-to-human transmission, provided that their availability has short time-frames and that mistakes from the recent past (vaccine inequity) will not be reiterated.
Collapse
Affiliation(s)
- Lorenzo Subissi
- Health Emergencies Programme, World Health Organization, Geneva, Switzerland
| | - Paola Stefanelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, Roma, Italy
| | - Giovanni Rezza
- Health Prevention Directorate, Ministry of Health, Roma, Italy
| |
Collapse
|
5
|
Atasoy MO, Naggar RFE, Rohaim MA, Munir M. Zoonotic and Zooanthroponotic Potential of Monkeypox. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1451:75-90. [PMID: 38801572 DOI: 10.1007/978-3-031-57165-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The current multicounty outbreak of monkeypox virus (MPXV) posed an emerging and continued challenge to already strained public healthcare sector, around the globe. Since its first identification, monkeypox disease (mpox) remained enzootic in Central and West African countries where reports of human cases are sporadically described. Recent trends in mpox spread outside the Africa have highlighted increased incidence of spillover of the MPXV from animal to humans. While nature of established animal reservoirs remained undefined, several small mammals including rodents, carnivores, lagomorphs, insectivores, non-human primates, domestic/farm animals, and several species of wildlife are proposed to be carrier of the MPXV infection. There are established records of animal-to-human (zoonotic) spread of MPXV through close interaction of humans with animals by eating bushmeat, contracting bodily fluids or trading possibly infected animals. In contrast, there are reports and increasing possibilities of human-to-animal (zooanthroponotic) spread of the MPXV through petting and close interaction with pet owners and animal care workers. We describe here the rationales and molecular factors which predispose the spread of MPXV not only amongst humans but also from animals to humans. A range of continuing opportunities for the spread and evolution of MPXV are discussed to consider risks beyond the currently identified groups. With the possibility of MPXV establishing itself in animal reservoirs, continued and broad surveillance, investigation into unconventional transmissions, and exploration of spillover events are warranted.
Collapse
Affiliation(s)
- Mustafa O Atasoy
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Furness College, Lancaster University, Tower Ave, Bailrigg, LA1 4YG, UK
| | - Rania F El Naggar
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Furness College, Lancaster University, Tower Ave, Bailrigg, LA1 4YG, UK
| | - Mohammed A Rohaim
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Furness College, Lancaster University, Tower Ave, Bailrigg, LA1 4YG, UK
| | - Muhammad Munir
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Furness College, Lancaster University, Tower Ave, Bailrigg, LA1 4YG, UK.
| |
Collapse
|
6
|
Mungmunpuntipantip R, Wiwanitkit V. Orf, a Human Parapoxvirus Infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1451:171-181. [PMID: 38801578 DOI: 10.1007/978-3-031-57165-7_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Despite being common worldwide, parapoxvirus infections are regarded as neglected zoonoses because their incidence is either unknown or grossly overestimated. In ruminants all throughout the world, parapoxvirus produces oral lesions and infectious pustular dermatitis. The pathogen is typically spread directly via items contaminated with parapoxvirus and indirectly via a near contact with dermatological lesions that contain the virus on affected animals. Animals infected with the parapoxvirus typically exhibit no clinical symptoms, and the mode of parapoxvirus transmission is occasionally unclear. For accurate etiological diagnosis and appropriate therapy of patients affected by zoonotic infections, the significance of adopting a "One Health" approach and cross-sector collaboration between human and veterinary medicine should be emphasized. The causative pathogen of ecthyma contagiosum in general people is the orf virus, which mostly infects various animals, either pets or wildlife species. The illness primarily affects minute wild ruminants, sheep, cattle, deer, and goats, and it can spread to people through contact with infected animals or contaminated meats anywhere in the world. Taxonomically speaking, the virus belongs to the parapoxvirus genus. Thus pathogen can be detected from crusts for a very long period (several months to several years), and the virus is found to be resistant to inactivation with a hot or dry atmosphere. In immunocompetent individuals, the lesions often go away on their own with a period as long 2 months. Nevertheless, it necessitates the applying of diverse strategies, such as antiviral, immunological modulator, or modest surgical excisions in immunosuppressed patients. The interaction of the virus with various host populations aids in the development of a defense mechanism against the immune system. The parapoxvirus illness in humans is covered in this chapter. The orf illness, a significant known human parapoxvirus infection, is given specific attention.
Collapse
|
7
|
Gao Z, He X, Chen G, Fang Y, Meng Z, Tian H, Zhang H, Jing Z. The Viral Protein Poly(A) Polymerase Catalytic Subunit Interacts with Guanylate-Binding Proteins 2 to Antagonize the Antiviral Ability of Targeting Ectromelia Virus. Int J Mol Sci 2023; 24:15750. [PMID: 37958732 PMCID: PMC10648259 DOI: 10.3390/ijms242115750] [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: 09/18/2023] [Revised: 10/16/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
The recent spread of the monkeypox virus among humans has heightened concerns regarding orthopoxvirus infections. Consequently, conducting a comprehensive study on the immunobiology of the monkeypox virus is imperative for the development of effective therapeutics. Ectromelia virus (ECTV) closely resembles the genetic and disease characteristics of monkeypox virus, making it a valuable research tool for studying orthopoxvirus-host interactions. Guanylate-binding proteins (GBPs), highly expressed interferon-stimulated genes (ISGs), have antagonistic effects against various intracellular pathogenic microorganisms. Our previous research has shown that GBP2 has a mild but statistically significant inhibitory effect on ECTV infection. The presence of a significant number of molecules in the poxvirus genome that encode the host immune response raises questions about whether it also includes proteins that counteract the antiviral activity of GBP2. Using IP/MS and co-IP technology, we discovered that the poly(A) polymerase catalytic subunit (PAPL) protein of ECTV is a viral regulatory molecule that interacts with GBP2. Further studies have shown that PAPL antagonizes the antiviral activity of GBP2 by reducing its protein levels. Knocking out the PAPL gene of ECTV with the CRISPR/Cas9 system significantly diminishes the replication ability of the virus, indicating the indispensable role of PAPL in the replication process of ECTV. In conclusion, our study presents preliminary evidence supporting the significance of PAPL as a virulence factor that can interact with GBP2.
Collapse
Affiliation(s)
- Zhenzhen Gao
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Xiaobing He
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Guohua Chen
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Yongxiang Fang
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Zejing Meng
- School of Public Health, Lanzhou University, Lanzhou 730000, China;
| | - Huihui Tian
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Hui Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Zhizhong Jing
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
- School of Public Health, Lanzhou University, Lanzhou 730000, China;
| |
Collapse
|
8
|
Ferrareze PAG, Pereira E Costa RA, Thompson CE. Genomic characterization and molecular evolution of human monkeypox viruses. Arch Virol 2023; 168:278. [PMID: 37864757 DOI: 10.1007/s00705-023-05904-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/30/2023] [Indexed: 10/23/2023]
Abstract
Monkeypox virus is a member of the family Poxviridae, as are variola virus and vaccinia virus. It has a linear double-strand DNA genome approximately 197 kb long, containing ~190 non-overlapping ORFs. Comparison of members of the Central and West African clades shows the presence of unique genes that are associated with different disease presentations, depending on the strain. The last smallpox vaccination efforts ended in the mid-1980s, and there is concern about the recent spread of human monkeypox disease around the world. Almost 87,000 human monkeypox cases have been diagnosed in the world, of which more than 10,900 were in Brazil. The aim of this study was to evaluate the epidemiology and molecular evolution of hMpxV. From computational biology analysis of 640 hMpxV genomes from 1962 to 2022, synteny breaks and gene conservation were observed between Central and West clade genomes, and strains belonged with the 2022 outbreak assigned to the West African clade. Evidence was found for diversifying selective pressure at specific sites within protein coding sequences, acting on immunomodulatory processes. The existence of different sites under diversifying and purifying selection in paralog genes indicates adaptive mechanisms underlying the host-pathogen interaction of monkeypox virus in humans.
Collapse
Affiliation(s)
- Patrícia Aline Gröhs Ferrareze
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | | | - Claudia Elizabeth Thompson
- Department of Pharmacosciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 245/200C Sarmento Leite St, Porto Alegre, RS, 90050-170, Brazil.
| |
Collapse
|
9
|
Mazloum A, Karagyaur M, Chernyshev R, van Schalkwyk A, Jun M, Qiang F, Sprygin A. Post-genomic era in agriculture and veterinary science: successful and proposed application of genetic targeting technologies. Front Vet Sci 2023; 10:1180621. [PMID: 37601766 PMCID: PMC10434572 DOI: 10.3389/fvets.2023.1180621] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
Gene editing tools have become an indispensable part of research into the fundamental aspects of cell biology. With a vast body of literature having been generated based on next generation sequencing technologies, keeping track of this ever-growing body of information remains challenging. This necessitates the translation of genomic data into tangible applications. In order to address this objective, the generated Next Generation Sequencing (NGS) data forms the basis for targeted genome editing strategies, employing known enzymes of various cellular machinery, in generating organisms with specifically selected phenotypes. This review focuses primarily on CRISPR/Cas9 technology in the context of its advantages over Zinc finger proteins (ZNF) and Transcription activator-like effector nucleases (TALEN) and meganucleases mutagenesis strategies, for use in agricultural and veterinary applications. This review will describe the application of CRISPR/Cas9 in creating modified organisms with custom-made properties, without the undesired non-targeted effects associated with virus vector vaccines and bioactive molecules produced in bacterial systems. Examples of the successful and unsuccessful applications of this technology to plants, animals and microorganisms are provided, as well as an in-depth look into possible future trends and applications in vaccine development, disease resistance and enhanced phenotypic traits will be discussed.
Collapse
Affiliation(s)
- Ali Mazloum
- Federal Center for Animal Health, Vladimir, Russia
| | - Maxim Karagyaur
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | | | - Antoinette van Schalkwyk
- Agricultural Research Council-Onderstepoort Veterinary Institute, Onderstepoort, South Africa
- Department of Biotechnology, University of the Western Cape, Bellville, South Africa
| | - Ma Jun
- School of Life Sciences and Engineering, Foshan University, Foshan, China
| | - Fu Qiang
- School of Life Sciences and Engineering, Foshan University, Foshan, China
| | | |
Collapse
|
10
|
Rahman MM, van Oosterom F, Enow JA, Hossain M, Gutierrez-Jensen AD, Cashen M, Everts A, Lowe K, Kilbourne J, Daggett-Vondras J, Karr TL, McFadden G. Nuclear Export Inhibitor Selinexor Enhances Oncolytic Myxoma Virus Therapy against Cancer. CANCER RESEARCH COMMUNICATIONS 2023; 3:952-968. [PMID: 37377603 PMCID: PMC10234290 DOI: 10.1158/2767-9764.crc-22-0483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/08/2023] [Accepted: 05/11/2023] [Indexed: 06/29/2023]
Abstract
Oncolytic viruses exploited for cancer therapy have been developed to selectively infect, replicate, and kill cancer cells to inhibit tumor growth. However, in some cancer cells, oncolytic viruses are often limited in completing their full replication cycle, forming progeny virions, and/or spreading in the tumor bed because of the heterogeneous cell types within the tumor bed. Here, we report that the nuclear export pathway regulates oncolytic myxoma virus (MYXV) infection and cytoplasmic viral replication in a subclass of human cancer cell types where viral replication is restricted. Inhibition of the XPO-1 (exportin 1) nuclear export pathway with nuclear export inhibitors can overcome this restriction by trapping restriction factors in the nucleus and allow significantly enhanced viral replication and killing of cancer cells. Furthermore, knockdown of XPO-1 significantly enhanced MYXV replication in restrictive human cancer cells and reduced the formation of antiviral granules associated with RNA helicase DHX9. Both in vitro and in vivo, we demonstrated that the approved XPO1 inhibitor drug selinexor enhances the replication of MYXV and kills diverse human cancer cells. In a xenograft tumor model in NSG mice, combination therapy with selinexor plus MYXV significantly reduced the tumor burden and enhanced the survival of animals. In addition, we performed global-scale proteomic analysis of nuclear and cytosolic proteins in human cancer cells to identify the host and viral proteins that were upregulated or downregulated by different treatments. These results indicate, for the first time, that selinexor in combination with oncolytic MYXV can be used as a potential new therapy. Significance We demonstrated that a combination of nuclear export inhibitor selinexor and oncolytic MYXV significantly enhanced viral replication, reduced cancer cell proliferation, reduced tumor burden, and enhanced the overall survival of animals. Thus, selinexor and oncolytic MYXV can be used as potential new anticancer therapy.
Collapse
Affiliation(s)
- Masmudur M. Rahman
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Fleur van Oosterom
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Junior A. Enow
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
| | - Maksuda Hossain
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Ami D. Gutierrez-Jensen
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Mackenzie Cashen
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Anne Everts
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Kenneth Lowe
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Jacquelyn Kilbourne
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Juliane Daggett-Vondras
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Timothy L. Karr
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Grant McFadden
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
| |
Collapse
|
11
|
Khamees A, Awadi S, Al-Shami K, Alkhoun HA, Al-Eitan SF, Alsheikh AM, Saeed A, Al-Zoubi RM, Zoubi MSA. Human monkeypox virus in the shadow of the COVID-19 pandemic. J Infect Public Health 2023; 16:1149-1157. [PMID: 37269693 DOI: 10.1016/j.jiph.2023.05.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/31/2023] [Accepted: 05/10/2023] [Indexed: 06/05/2023] Open
Abstract
BACKGROUND The end of smallpox in 1980 and the subsequent stopping of vaccination against smallpox was followed by the emergence of monkeypox (mpox), a viral disease of animal origin, meaning that it is transmitted from animal to human. The symptoms of mpox are similar to smallpox, except that they are less severe in terms of clinical features. In the case of public health, the mpox virus is one of the most important orthopoxviruses (such as variola, cowpox, and vaccinia) that come from the family Poxviridae. Mpox occurs mostly in central Africa and sometimes in tropical rainforests or some urban areas. Also, there are threats other than COVID-19, that must be addressed and prevented from spreading, as there has been an outbreak of mpox cases since May 7, 2022, throughout the USA, Europe, Australia, and part of Africa. OBJECTIVES In this review, we will discuss mpox between the past, the present and during the COVID-19 pandemic. Also, it offers an updated summary of the taxonomy, etiology, transmission, and epidemiology of mpox illness. In addition, the current review aims to highlight the importance of emerging pandemics in the same era such as mpox and COVID-19. METHODS A literature search was done for the study using online sources like PubMed and Google Scholar. Publications in English were included. Data for study variables were extracted. After the duplicate articles were eliminated, full-text screening was performed on the papers' titles and abstracts. RESULTS The evaluation included a series documenting mpox virus outbreaks, and both prospective and retrospectiveinvestigations. CONCLUSIONS monkeypox is a viral disease caused by the monkeypox virus (MPXV), which is primarily found in central and western Africa. The disease is transmitted from animals to humans and presents symptoms similar to those of smallpox, including fever, headache, muscle aches, and a rash. Monkeypox can lead to complications such as secondary integument infection, bronchopneumonia, sepsis, and encephalitis, as well as corneal infection that can result in blindness. There is no specific clinically proven treatment for monkeypox, and treatment is primarily supportive. However, antiviral drugs and vaccines are available for cross-protection against the virus, and strict infection control measures and vaccination of close contacts of affected individuals can help prevent and control outbreaks.
Collapse
Affiliation(s)
- Almu'atasim Khamees
- Faculty of Medicine, Yarmouk University, P.O Box 566, 21163 Irbid, Jordan; Department of General Surgery, King Hussein Cancer Center, Amman, 11941, Jordan.
| | - Sajeda Awadi
- Faculty of Medicine, Yarmouk University, P.O Box 566, 21163 Irbid, Jordan.
| | - Khayry Al-Shami
- Faculty of Medicine, Yarmouk University, P.O Box 566, 21163 Irbid, Jordan.
| | - Hayat Abu Alkhoun
- Faculty of Medicine, Yarmouk University, P.O Box 566, 21163 Irbid, Jordan.
| | - Sharaf F Al-Eitan
- Faculty of Medicine, Yarmouk University, P.O Box 566, 21163 Irbid, Jordan.
| | | | - Ahmad Saeed
- Faculty of Medicine, Yarmouk University, P.O Box 566, 21163 Irbid, Jordan.
| | - Raed M Al-Zoubi
- Surgical Research Section, Department of Surgery, Hamad Medical Corporation, Doha, Qatar; Department of Biomedical Sciences, College of Health Sciences, QU-Health, Qatar University, Doha 2713, Qatar; Department of Chemistry, Jordan University of Science and Technology, P.O.Box 3030, Irbid 22110, Jordan.
| | | |
Collapse
|
12
|
Shafaati M, Zandi M. Human monkeypox (hMPXV) re-emergence: Host immunity status and current vaccines landscape. J Med Virol 2023; 95:e28251. [PMID: 36271768 DOI: 10.1002/jmv.28251] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/28/2022] [Accepted: 10/19/2022] [Indexed: 01/11/2023]
Abstract
Monkeypox virus is a member of the Orthopoxvirus genus and the Poxviridae family. Orthopoxviruses are among the most intricate animal viruses. The pathogenicity of human monkeypox infection has been emphasized in response to its recent emergence in non-endemic countries and the threat of bioterrorism. It is always necessary to take appropriate precautions in exposure to emerging or re-emerging infections. Here, we focus on the current state of the human monkeypox infection outbreak, research & development of immune responses, and clinical interventions to prevent and treat the human monkeypox virus and other human poxviruses.
Collapse
Affiliation(s)
- Maryam Shafaati
- Department of Microbiology, Faculty of Science, Jahrom Branch, Islamic Azad University, Jahrom, Iran
- Occupational Sleep Research, Baharloo Hospital, Tehran University of Medical Science, Tehran, Iran
| | - Milad Zandi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Zaib S, Rana N, Areeba, Hussain N, Alrbyawi H, Dera AA, Khan I, Khalid M, Khan A, Al-Harrasi A. Designing multi-epitope monkeypox virus-specific vaccine using immunoinformatics approach. J Infect Public Health 2023; 16:107-116. [PMID: 36508944 PMCID: PMC9724569 DOI: 10.1016/j.jiph.2022.11.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/15/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Monkeypox virus is an enveloped DNA virus that belongs to Poxviridae family. The virus is transmitted from rodents to primates via infected body fluids, skin lesions, and respiratory droplets. After being infected with virus, the patients experience fever, myalgia, maculopapular rash, and fluid-filled blisters. It is necessary to differentiate monkeypox virus from other poxviruses during diagnosis which can be appropriately envisioned via DNA analysis from swab samples. During small outbreaks, the virus is treated with therapies administered in other orthopoxviruses infections and does not have its own specific therapy and vaccine. Consequently, in this article, two potential peptides have been designed. METHODS For the purpose of designing a vaccine, protein sequences were retrieved followed by the prediction of B- and T-cell epitopes. Afterward, vaccine structures were predicted which were docked with toll-like receptors. The docked complexes were analyzed with iMODS. Moreover, vaccine constructs nucleotide sequences were optimized and expressed in silico. RESULTS COP-B7R vaccine construct (V1) has antigenicity score of 0.5400, instability index of 29.33, z-score of - 2.11-, and 42.11% GC content whereas COP-A44L vaccine construct (V2) has an antigenicity score of 0.7784, instability index of 23.33, z-score of - 0.61, and 48.63% GC content. It was also observed that COP-A44L can be expressed as a soluble protein in Escherichia coli as compared to COP-B7R which requires a different expression system. CONCLUSION The obtained results revealed that both vaccine constructs show satisfactory outcomes after in silico investigation and have significant potential to prevent the monkeypox virus. However, COP-A44L gave better results.
Collapse
Affiliation(s)
- Sumera Zaib
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore 54590, Pakistan.
| | - Nehal Rana
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore 54590, Pakistan
| | - Areeba
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore 54590, Pakistan
| | - Nadia Hussain
- Department of Pharmaceutical Sciences, College of Pharmacy, Al Ain University, Al Ain, UAE; AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi, UAE
| | - Hamad Alrbyawi
- Pharmaceutics and Pharmaceutical Technology Department, College of Pharmacy, Taibah University, Medina 42353, Saudi Arabia
| | - Ayed A Dera
- Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Imtiaz Khan
- Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom.
| | - Mohammad Khalid
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Asir-Abha 61421, Saudi Arabia
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Oman.
| |
Collapse
|
14
|
Xiang Y, White A. Monkeypox virus emerges from the shadow of its more infamous cousin: family biology matters. Emerg Microbes Infect 2022; 11:1768-1777. [PMID: 35751396 PMCID: PMC9278444 DOI: 10.1080/22221751.2022.2095309] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/23/2022] [Accepted: 06/23/2022] [Indexed: 11/25/2022]
Abstract
Monkeypox virus (MPXV) is closely related to the infamous variola (smallpox) virus, causing a febrile rash illness in humans similar to but milder than smallpox. In the twentieth century, human monkeypox had been mostly a rare zoonotic disease confined to forested areas in West and Central Africa. However, the case number and geographic range have increased significantly in this century, coincided with the waning of the smallpox vaccine-induced immunity in the global population. The outbreak of human monkeypox in multiple countries since May 2022 has been unusual in its large case number and the absence of direct links to endemic countries, raising concerns for a possible change in monkeypox transmission pattern that could pose a greater global threat. Here, we review aspects of MPXV biology that are relevant for risk assessment and preparedness for a monkeypox epidemic, with an emphasis on recent progress in understanding of the virus host range, evolutionary potential, and neutralization targets.
Collapse
Affiliation(s)
- Yan Xiang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, TX, USA
| | - Addison White
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
15
|
Chaurasiya S, Yang A, Zhang Z, Lu J, Valencia H, Kim SI, Woo Y, Warner SG, Olafsen T, Zhao Y, Wu X, Fein S, Cheng L, Cheng M, Ede N, Fong Y. A comprehensive preclinical study supporting clinical trial of oncolytic chimeric poxvirus CF33-hNIS-anti-PD-L1 to treat breast cancer. Mol Ther Methods Clin Dev 2022; 24:102-116. [PMID: 35024377 PMCID: PMC8718831 DOI: 10.1016/j.omtm.2021.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/04/2021] [Indexed: 01/12/2023]
Abstract
CF33-hNIS-anti-PD-L1 is an oncolytic chimeric poxvirus encoding two transgenes: human sodium iodide symporter and a single-chain variable fragment against PD-L1. Comprehensive preclinical pharmacology studies encompassing primary and secondary pharmacodynamics and biodistribution and safety studies were performed to support the clinical development of CF33-hNIS-anti-PD-L1. Most of the studies were performed in triple-negative breast cancer (TNBC) models, as the phase I trial is planned for patients with TNBC. Biological functions of virus-encoded transgenes were confirmed, and the virus demonstrated anti-tumor efficacy against TNBC models in mice. In a good laboratory practice (GLP) toxicology study, the virus did not produce any observable adverse effects in mice, suggesting that the doses proposed for the clinical trial should be well tolerated in patients. Furthermore, no neurotoxic effects in mice were seen following intracranial injection of the virus. Also, the risk for horizontal transmission of CF33-hNIS-anti-PD-L1 was assessed in mice, and our results suggest that the virus is unlikely to transmit from infected patients to healthy individuals. Finally, the in-use stability and compatibility of CF33-hNIS-anti-PD-L1 tested under different conditions mimicking the clinical scenarios confirmed the suitability of the virus in clinical settings. The results of these preclinical studies support the use of CF33-hNIS-anti-PD-L1 in a first-in-human trial in patients with TNBC.
Collapse
Affiliation(s)
- Shyambabu Chaurasiya
- Department of Surgery, City of Hope National Medical Center, Familian Science building, Room#1100 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Annie Yang
- Department of Surgery, City of Hope National Medical Center, Familian Science building, Room#1100 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Zhifang Zhang
- Department of Surgery, City of Hope National Medical Center, Familian Science building, Room#1100 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Jianming Lu
- Department of Surgery, City of Hope National Medical Center, Familian Science building, Room#1100 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Hannah Valencia
- Department of Surgery, City of Hope National Medical Center, Familian Science building, Room#1100 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Sang-In Kim
- Department of Surgery, City of Hope National Medical Center, Familian Science building, Room#1100 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Yanghee Woo
- Department of Surgery, City of Hope National Medical Center, Familian Science building, Room#1100 1500 E Duarte Road, Duarte, CA 91010, USA
| | - Suanne G Warner
- Department of Surgery, Mayo Clinic, Rochester, MN 55902, USA
| | - Tove Olafsen
- Small Animal Imaging Core, Shared Resources, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Yuqi Zhao
- Integrative Genomics Core, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Xiwei Wu
- Integrative Genomics Core, City of Hope National Medical Center, Duarte, CA 91010, USA
| | | | | | | | | | - Yuman Fong
- Department of Surgery, City of Hope National Medical Center, Familian Science building, Room#1100 1500 E Duarte Road, Duarte, CA 91010, USA
| |
Collapse
|
16
|
Águeda-Pinto A, Alves LQ, Neves F, McFadden G, Jacobs BL, Castro LFC, Rahman MM, Esteves PJ. Convergent Loss of the Necroptosis Pathway in Disparate Mammalian Lineages Shapes Viruses Countermeasures. Front Immunol 2021; 12:747737. [PMID: 34539677 PMCID: PMC8445033 DOI: 10.3389/fimmu.2021.747737] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 08/17/2021] [Indexed: 01/21/2023] Open
Abstract
Programmed cell death is a vital process in the life cycle of organisms. Necroptosis, an evolutionary form of programmed necrosis, contributes to the innate immune response by killing pathogen-infected cells. This virus-host interaction pathway is organized around two components: the receptor-interacting protein kinase 3 (RIPK3), which recruits and phosphorylates the mixed lineage kinase-like protein (MLKL), inducing cellular plasma membrane rupture and cell death. Critically, the presence of necroptotic inhibitors in viral genomes validates necroptosis as an important host defense mechanism. Here, we show, counterintuitively, that in different mammalian lineages, central components of necroptosis, such as RIPK3 and MLKL, are deleted or display inactivating mutations. Frameshifts or premature stop codons are observed in all the studied species of cetaceans and leporids. In carnivores' genomes, the MLKL gene is deleted, while in a small number of species from afrotheria and rodentia premature stop codons are observed in RIPK3 and/or MLKL. Interestingly, we also found a strong correlation between the disruption of necroptosis in leporids and cetaceans and the absence of the N-terminal domain of E3-like homologs (responsible for necroptosis inhibition) in their naturally infecting poxviruses. Overall, our study provides the first comprehensive picture of the molecular evolution of necroptosis in mammals. The loss of necroptosis multiple times during mammalian evolution highlights the importance of gene/pathway loss for species adaptation and suggests that necroptosis is not required for normal mammalian development. Moreover, this study highlights a co-evolutionary relationship between poxviruses and their hosts, emphasizing the role of host adaptation in shaping virus evolution.
Collapse
Affiliation(s)
- Ana Águeda-Pinto
- CIBIO/InBio-Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, Vairão, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Luís Q. Alves
- CIIMAR/CIMAR, Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto, Matosinhos, Portugal
| | - Fabiana Neves
- CIBIO/InBio-Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, Vairão, Portugal
| | - Grant McFadden
- Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Bertram L. Jacobs
- Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
- School of Life Sciences Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - L. Filipe C. Castro
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- CIIMAR/CIMAR, Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto, Matosinhos, Portugal
| | - Masmudur M. Rahman
- Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Pedro J. Esteves
- CIBIO/InBio-Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, Vairão, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- CITS-Centro de Investigação em Tecnologias da Saúde, Instituto Politécnico de Saúde do Norte (IPSN), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Gandra, Portugal
| |
Collapse
|
17
|
Bourquain D, Schrick L, Tischer BK, Osterrieder K, Schaade L, Nitsche A. Replication of cowpox virus in macrophages is dependent on the host range factor p28/N1R. Virol J 2021; 18:173. [PMID: 34425838 PMCID: PMC8381512 DOI: 10.1186/s12985-021-01640-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/10/2021] [Indexed: 11/23/2022] Open
Abstract
Zoonotic orthopoxvirus infections continue to represent a threat to human health. The disease caused by distinct orthopoxviruses differs in terms of symptoms and severity, which may be explained by the unique repertoire of virus factors that modulate the host’s immune response and cellular machinery. We report here on the construction of recombinant cowpox viruses (CPXV) which either lack the host range factor p28 completely or express truncated variants of p28. We show that p28 is essential for CPXV replication in macrophages of human or mouse origin and that the C-terminal RING finger domain of p28 is necessary to allow CPXV replication in macrophages.
Collapse
Affiliation(s)
- Daniel Bourquain
- Centre for Biological Threats and Special Pathogens 1, Robert Koch Institute, Seestraße 10, 13353, Berlin, Germany.
| | - Livia Schrick
- Centre for Biological Threats and Special Pathogens 1, Robert Koch Institute, Seestraße 10, 13353, Berlin, Germany
| | - Bernd Karsten Tischer
- Institute of Virology, Department of Veterinary Medicine, Freie Universität Berlin, Robert-von-Ostertag-Str. 7-13, 14163, Berlin, Germany
| | - Klaus Osterrieder
- Institute of Virology, Department of Veterinary Medicine, Freie Universität Berlin, Robert-von-Ostertag-Str. 7-13, 14163, Berlin, Germany.,Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Lars Schaade
- Centre for Biological Threats and Special Pathogens 1, Robert Koch Institute, Seestraße 10, 13353, Berlin, Germany
| | - Andreas Nitsche
- Centre for Biological Threats and Special Pathogens 1, Robert Koch Institute, Seestraße 10, 13353, Berlin, Germany
| |
Collapse
|
18
|
Silva NIO, de Oliveira JS, Kroon EG, Trindade GDS, Drumond BP. Here, There, and Everywhere: The Wide Host Range and Geographic Distribution of Zoonotic Orthopoxviruses. Viruses 2020; 13:E43. [PMID: 33396609 PMCID: PMC7823380 DOI: 10.3390/v13010043] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 01/05/2023] Open
Abstract
The global emergence of zoonotic viruses, including poxviruses, poses one of the greatest threats to human and animal health. Forty years after the eradication of smallpox, emerging zoonotic orthopoxviruses, such as monkeypox, cowpox, and vaccinia viruses continue to infect humans as well as wild and domestic animals. Currently, the geographical distribution of poxviruses in a broad range of hosts worldwide raises concerns regarding the possibility of outbreaks or viral dissemination to new geographical regions. Here, we review the global host ranges and current epidemiological understanding of zoonotic orthopoxviruses while focusing on orthopoxviruses with epidemic potential, including monkeypox, cowpox, and vaccinia viruses.
Collapse
Affiliation(s)
| | | | | | | | - Betânia Paiva Drumond
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais: Belo Horizonte, Minas Gerais 31270-901, Brazil; (N.I.O.S.); (J.S.d.O.); (E.G.K.); (G.d.S.T.)
| |
Collapse
|
19
|
Brito AF, Pinney JW. The evolution of protein domain repertoires: Shedding light on the origins of the Herpesviridae family. Virus Evol 2020; 6:veaa001. [PMID: 32042448 PMCID: PMC7000910 DOI: 10.1093/ve/veaa001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Herpesviruses (HVs, Family: Herpesviridae) have large genomes that encode hundreds of proteins. Apart from amino acid mutations, protein domain acquisitions, duplications and losses are also common modes of evolution. HV domain repertoires differ across species, and only a core set is shared among all species, aspect that raises a question: How have HV domain repertoires diverged while keeping some similarities? To answer such question, we used profile Hidden Markov Models (HMMs) to search for domains in all possible translated open reading frames (ORFs) of fully sequenced HV genomes. With at least 274 domains being identified, we built a matrix of domain counts per species, and applied a parsimony method to reconstruct the ancestral states of these domains along the HV phylogeny. It revealed events of domain gain, duplication, and loss over more than 400 millions of years, where Alpha-, Beta-, and GammaHVs expanded and condensed their domain repertoires at distinct rates. Most of the acquired domains perform ‘Modulation and Control’, ‘Envelope’, or ‘Auxiliary’ functions, categories that showed high flexibility (number of domains) and redundancy (number of copies). Conversely, few gains and duplications were observed for domains involved in ‘Capsid assembly and structure’, and ‘DNA Replication, recombination and metabolism’. Among the forty-one primordial domains encoded by Herpesviridae ancestors, twenty-eight are still found in all present-day HVs. Because of their distinct evolutionary strategies, HV domain repertoires are very specific at the subfamily, genus and species levels. Differences in domain composition may not only explain HV host range and tissue tropism, but also provide hints to the origins of HVs.
Collapse
Affiliation(s)
- Anderson F Brito
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - John W Pinney
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
20
|
Águeda-Pinto A, Lemos de Matos A, Abrantes M, Kraberger S, Risalde MA, Gortázar C, McFadden G, Varsani A, Esteves PJ. Genetic Characterization of a Recombinant Myxoma Virus in the Iberian Hare ( Lepus granatensis). Viruses 2019; 11:v11060530. [PMID: 31181645 PMCID: PMC6631704 DOI: 10.3390/v11060530] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/30/2019] [Accepted: 06/06/2019] [Indexed: 01/15/2023] Open
Abstract
Myxomatosis is a lethal disease in wild European and domestic rabbits (Oryctolagus cuniculus), which is caused by a Myxoma virus (MYXV) infection-a leporipoxvirus that is found naturally in some Sylvilagus rabbit species in South America and California. The introduction of MYXV into feral European rabbit populations of Australia and Europe, in the early 1950s, demonstrated the best-documented field example of host-virus coevolution, following a cross-species transmission. Recently, a new cross-species jump of MYXV has been suggested in both Great Britain and Spain, where European brown hares (Lepus europaeus) and Iberian hares (Lepus granatensis) were found dead with lesions consistent with those observed in myxomatosis. To investigate the possibility of a new cross-species transmission event by MYXV, tissue samples collected from a wild Iberian hare found dead in Spain (Toledo region) were analyzed and deep sequenced. Our results reported a new MYXV isolate (MYXV Toledo) in the tissues of this species. The genome of this new virus was found to encode three disruptive genes (M009L, M036L, and M152R) and a novel ~2.8 kb recombinant region, which resulted from an insertion of four novel poxviral genes towards the 3' end of the negative strand of its genome. From the open reading frames inserted into the MYXV Toledo virus, a new orthologue of a poxvirus host range gene family member was identified, which was related to the MYXV gene M064R. Overall, we confirmed the identity of a new MYXV isolate in Iberian hares, which, we hypothesized, was able to more effectively counteract the host defenses in hares and start an infectious process in this new host.
Collapse
Affiliation(s)
- Ana Águeda-Pinto
- CIBIO/InBio-Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, Campus Agrário de Vairão, 4485-661 Vairão, Portugal.
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal.
- Center for Immunotherapy, Vaccines, and Virotherapy (CIVV), The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA.
| | - Ana Lemos de Matos
- Center for Immunotherapy, Vaccines, and Virotherapy (CIVV), The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA.
| | - Mário Abrantes
- Center for Immunotherapy, Vaccines, and Virotherapy (CIVV), The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA.
| | - Simona Kraberger
- The Biodesign Center for Fundamental and Applied Microbiomics, Center for Evolution and Medicine and School of Life sciences, Arizona State University, Tempe, AZ 85287, USA.
| | - Maria A Risalde
- Dpto. de Anatomía y Anatomía Patológica Comparadas, Universidad de Córdoba, Agrifood Excellence International Campus (ceiA3), 14071 Córdoba, Spain.
| | - Christian Gortázar
- Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo, 28005 Ciudad Real, Spain.
| | - Grant McFadden
- Center for Immunotherapy, Vaccines, and Virotherapy (CIVV), The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA.
| | - Arvind Varsani
- The Biodesign Center for Fundamental and Applied Microbiomics, Center for Evolution and Medicine and School of Life sciences, Arizona State University, Tempe, AZ 85287, USA.
- Structural Biology Research Unit, Department of Clinical Laboratory Sciences, University of Cape Town, Cape Town 7701, South Africa.
| | - Pedro J Esteves
- CIBIO/InBio-Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, Campus Agrário de Vairão, 4485-661 Vairão, Portugal.
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal.
- CITS-Centro de Investigação em Tecnologias da Saúde, IPSN, CESPU, 4585-116 Gandra, Portugal.
| |
Collapse
|
21
|
Albarnaz JD, Torres AA, Smith GL. Modulating Vaccinia Virus Immunomodulators to Improve Immunological Memory. Viruses 2018; 10:E101. [PMID: 29495547 PMCID: PMC5869494 DOI: 10.3390/v10030101] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 12/14/2022] Open
Abstract
The increasing frequency of monkeypox virus infections, new outbreaks of other zoonotic orthopoxviruses and concern about the re-emergence of smallpox have prompted research into developing antiviral drugs and better vaccines against these viruses. This article considers the genetic engineering of vaccinia virus (VACV) to enhance vaccine immunogenicity and safety. The virulence, immunogenicity and protective efficacy of VACV strains engineered to lack specific immunomodulatory or host range proteins are described. The ultimate goal is to develop safer and more immunogenic VACV vaccines that induce long-lasting immunological memory.
Collapse
Affiliation(s)
- Jonas D Albarnaz
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK.
| | - Alice A Torres
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK.
| | - Geoffrey L Smith
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK.
| |
Collapse
|
22
|
Okoli A, Okeke MI, Tryland M, Moens U. CRISPR/Cas9-Advancing Orthopoxvirus Genome Editing for Vaccine and Vector Development. Viruses 2018; 10:E50. [PMID: 29361752 PMCID: PMC5795463 DOI: 10.3390/v10010050] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/17/2018] [Accepted: 01/21/2018] [Indexed: 12/17/2022] Open
Abstract
The clustered regularly interspaced short palindromic repeat (CRISPR)/associated protein 9 (Cas9) technology is revolutionizing genome editing approaches. Its high efficiency, specificity, versatility, flexibility, simplicity and low cost have made the CRISPR/Cas9 system preferable to other guided site-specific nuclease-based systems such as TALENs (Transcription Activator-like Effector Nucleases) and ZFNs (Zinc Finger Nucleases) in genome editing of viruses. CRISPR/Cas9 is presently being applied in constructing viral mutants, preventing virus infections, eradicating proviral DNA, and inhibiting viral replication in infected cells. The successful adaptation of CRISPR/Cas9 to editing the genome of Vaccinia virus paves the way for its application in editing other vaccine/vector-relevant orthopoxvirus (OPXV) strains. Thus, CRISPR/Cas9 can be used to resolve some of the major hindrances to the development of OPXV-based recombinant vaccines and vectors, including sub-optimal immunogenicity; transgene and genome instability; reversion of attenuation; potential of spread of transgenes to wildtype strains and close contacts, which are important biosafety and risk assessment considerations. In this article, we review the published literature on the application of CRISPR/Cas9 in virus genome editing and discuss the potentials of CRISPR/Cas9 in advancing OPXV-based recombinant vaccines and vectors. We also discuss the application of CRISPR/Cas9 in combating viruses of clinical relevance, the limitations of CRISPR/Cas9 and the current strategies to overcome them.
Collapse
Affiliation(s)
- Arinze Okoli
- Biosafety of Genome Editing Research Group, GenØk-Centre for Biosafety, Siva Innovation Centre, N-9294 Tromsø, Norway.
| | - Malachy I Okeke
- Biosafety of Genome Editing Research Group, GenØk-Centre for Biosafety, Siva Innovation Centre, N-9294 Tromsø, Norway.
| | - Morten Tryland
- Biosafety of Genome Editing Research Group, GenØk-Centre for Biosafety, Siva Innovation Centre, N-9294 Tromsø, Norway.
- Artic Infection Biology, Department of Artic and Marine Biology, The Artic University of Norway, N-9037 Tromsø, Norway.
| | - Ugo Moens
- Molecular Inflammation Research Group, Institute of Medical Biology, The Arctic University of Norway, N-9037 Tromsø, Norway.
| |
Collapse
|
23
|
Combined Proteomics/Genomics Approach Reveals Proteomic Changes of Mature Virions as a Novel Poxvirus Adaptation Mechanism. Viruses 2017; 9:v9110337. [PMID: 29125539 PMCID: PMC5707544 DOI: 10.3390/v9110337] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 11/06/2017] [Accepted: 11/07/2017] [Indexed: 12/16/2022] Open
Abstract
DNA viruses, like poxviruses, possess a highly stable genome, suggesting that adaptation of virus particles to specific cell types is not restricted to genomic changes. Cowpox viruses are zoonotic poxviruses with an extraordinarily broad host range, demonstrating their adaptive potential in vivo. To elucidate adaptation mechanisms of poxviruses, we isolated cowpox virus particles from a rat and passaged them five times in a human and a rat cell line. Subsequently, we analyzed the proteome and genome of the non-passaged virions and each passage. While the overall viral genome sequence was stable during passaging, proteomics revealed multiple changes in the virion composition. Interestingly, an increased viral fitness in human cells was observed in the presence of increased immunomodulatory protein amounts. As the only minor variant with increasing frequency during passaging was located in a viral RNA polymerase subunit and, moreover, most minor variants were found in transcription-associated genes, protein amounts were presumably regulated at transcription level. This study is the first comparative proteome analysis of virus particles before and after cell culture propagation, revealing proteomic changes as a novel poxvirus adaptation mechanism.
Collapse
|
24
|
Oliveira GP, Rodrigues RAL, Lima MT, Drumond BP, Abrahão JS. Poxvirus Host Range Genes and Virus-Host Spectrum: A Critical Review. Viruses 2017; 9:E331. [PMID: 29112165 PMCID: PMC5707538 DOI: 10.3390/v9110331] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/30/2017] [Accepted: 11/06/2017] [Indexed: 01/01/2023] Open
Abstract
The Poxviridae family is comprised of double-stranded DNA viruses belonging to nucleocytoplasmic large DNA viruses (NCLDV). Among the NCLDV, poxviruses exhibit the widest known host range, which is likely observed because this viral family has been more heavily investigated. However, relative to each member of the Poxviridae family, the spectrum of the host is variable, where certain viruses can infect a large range of hosts, while others are restricted to only one host species. It has been suggested that the variability in host spectrum among poxviruses is linked with the presence or absence of some host range genes. Would it be possible to extrapolate the restriction of viral replication in a specific cell lineage to an animal, a far more complex organism? In this study, we compare and discuss the relationship between the host range of poxvirus species and the abundance/diversity of host range genes. We analyzed the sequences of 38 previously identified and putative homologs of poxvirus host range genes, and updated these data with deposited sequences of new poxvirus genomes. Overall, the term host range genes might not be the most appropriate for these genes, since no correlation between them and the viruses' host spectrum was observed, and a change in nomenclature should be considered. Finally, we analyzed the evolutionary history of these genes, and reaffirmed the occurrence of horizontal gene transfer (HGT) for certain elements, as previously suggested. Considering the data presented in this study, it is not possible to associate the diversity of host range factors with the amount of hosts of known poxviruses, and this traditional nomenclature creates misunderstandings.
Collapse
Affiliation(s)
- Graziele Pereira Oliveira
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Rodrigo Araújo Lima Rodrigues
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Maurício Teixeira Lima
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Betânia Paiva Drumond
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Jônatas Santos Abrahão
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| |
Collapse
|
25
|
Marzook NB, Latham SL, Lynn H, Mckenzie C, Chaponnier C, Grau GE, Newsome TP. Divergent roles of β- and γ-actin isoforms during spread of vaccinia virus. Cytoskeleton (Hoboken) 2017; 74:170-183. [PMID: 28218453 PMCID: PMC7162416 DOI: 10.1002/cm.21356] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 01/31/2017] [Accepted: 02/16/2017] [Indexed: 01/20/2023]
Abstract
Actin is a major component of the cytoskeleton and is present as two isoforms in non‐muscle cells: β‐ and γ‐cytoplasmic actin. These isoforms are strikingly conserved, differing by only four N‐terminal amino acids. During spread from infected cells, vaccinia virus (VACV) particles induce localized actin nucleation that propel virus to surrounding cells and facilitate cell‐to‐cell spread of infection. Here we show that virus‐tipped actin comets are composed of β‐ and γ‐actin. We employed isoform‐specific siRNA knockdown to examine the role of the two isoforms in VACV‐induced actin comets. Despite the high level of similarity between the actin isoforms, and their colocalization, VACV‐induced actin nucleation was dependent exclusively on β‐actin. Knockdown of β‐actin led to a reduction in the release of virus from infected cells, a phenotype dependent on virus‐induced Arp2/3 complex activity. We suggest that local concentrations of actin isoforms may regulate the activity of cellular actin nucleator complexes.
Collapse
Affiliation(s)
- N Bishara Marzook
- School of Life and Environmental Sciences, The University of Sydney, Australia
| | - Sharissa L Latham
- Vascular Immunology Unit, Department of Pathology, School of Medical Sciences & Marie Bashir Institute, The University of Sydney, Australia
| | - Helena Lynn
- School of Life and Environmental Sciences, The University of Sydney, Australia
| | | | - Christine Chaponnier
- Department of Pathology-Immunology, Faculty of Medicine, University of Geneva, Switzerland
| | - Georges E Grau
- Vascular Immunology Unit, Department of Pathology, School of Medical Sciences & Marie Bashir Institute, The University of Sydney, Australia
| | - Timothy P Newsome
- School of Life and Environmental Sciences, The University of Sydney, Australia
| |
Collapse
|
26
|
Rahman MM, McFadden G. Myxoma Virus dsRNA Binding Protein M029 Inhibits the Type I IFN-Induced Antiviral State in a Highly Species-Specific Fashion. Viruses 2017; 9:v9020027. [PMID: 28157174 PMCID: PMC5332946 DOI: 10.3390/v9020027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 01/26/2017] [Indexed: 12/17/2022] Open
Abstract
Myxoma virus (MYXV) is a Leporipoxvirus that possesses a specific rabbit-restricted host tropism but exhibits a much broader cellular host range in cultured cells. MYXV is able to efficiently block all aspects of the type I interferon (IFN)-induced antiviral state in rabbit cells, partially in human cells and very poorly in mouse cells. The mechanism(s) of this species-specific inhibition of type I IFN-induced antiviral state is not well understood. Here we demonstrate that MYXV encoded protein M029, a truncated relative of the vaccinia virus (VACV) E3 double-stranded RNA (dsRNA) binding protein that inhibits protein kinase R (PKR), can also antagonize the type I IFN-induced antiviral state in a highly species-specific manner. In cells pre-treated with type I IFN prior to infection, MYXV exploits M029 to overcome the induced antiviral state completely in rabbit cells, partially in human cells, but not at all in mouse cells. However, in cells pre-infected with MYXV, IFN-induced signaling is fully inhibited even in the absence of M029 in cells from all three species, suggesting that other MYXV protein(s) apart from M029 block IFN signaling in a species-independent manner. We also show that the antiviral state induced in rabbit, human or mouse cells by type I IFN can inhibit M029-knockout MYXV even when PKR is genetically knocked-out, suggesting that M029 targets other host proteins for this antiviral state inhibition. Thus, the MYXV dsRNA binding protein M029 not only antagonizes PKR from multiple species but also blocks the type I IFN antiviral state independently of PKR in a highly species-specific fashion.
Collapse
Affiliation(s)
- Masmudur M Rahman
- The Biodesign Institute, Center for Immunotherapy, Vaccines, and Virotherapy, Arizona State University, Tempe, AZ 85287-5401, USA.
| | - Grant McFadden
- The Biodesign Institute, Center for Immunotherapy, Vaccines, and Virotherapy, Arizona State University, Tempe, AZ 85287-5401, USA.
| |
Collapse
|
27
|
Volz A, Sutter G. Modified Vaccinia Virus Ankara: History, Value in Basic Research, and Current Perspectives for Vaccine Development. Adv Virus Res 2016; 97:187-243. [PMID: 28057259 PMCID: PMC7112317 DOI: 10.1016/bs.aivir.2016.07.001] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Safety tested Modified Vaccinia virus Ankara (MVA) is licensed as third-generation vaccine against smallpox and serves as a potent vector system for development of new candidate vaccines against infectious diseases and cancer. Historically, MVA was developed by serial tissue culture passage in primary chicken cells of vaccinia virus strain Ankara, and clinically used to avoid the undesirable side effects of conventional smallpox vaccination. Adapted to growth in avian cells MVA lost the ability to replicate in mammalian hosts and lacks many of the genes orthopoxviruses use to conquer their host (cell) environment. As a biologically well-characterized mutant virus, MVA facilitates fundamental research to elucidate the functions of poxvirus host-interaction factors. As extremely safe viral vectors MVA vaccines have been found immunogenic and protective in various preclinical infection models. Multiple recombinant MVA currently undergo clinical testing for vaccination against human immunodeficiency viruses, Mycobacterium tuberculosis or Plasmodium falciparum. The versatility of the MVA vector vaccine platform is readily demonstrated by the swift development of experimental vaccines for immunization against emerging infections such as the Middle East Respiratory Syndrome. Recent advances include promising results from the clinical testing of recombinant MVA-producing antigens of highly pathogenic avian influenza virus H5N1 or Ebola virus. This review summarizes our current knowledge about MVA as a unique strain of vaccinia virus, and discusses the prospects of exploiting this virus as research tool in poxvirus biology or as safe viral vector vaccine to challenge existing and future bottlenecks in vaccinology.
Collapse
Affiliation(s)
- A Volz
- German Center for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Munich, Germany
| | - G Sutter
- German Center for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Munich, Germany.
| |
Collapse
|
28
|
Myxoma virus M156 is a specific inhibitor of rabbit PKR but contains a loss-of-function mutation in Australian virus isolates. Proc Natl Acad Sci U S A 2016; 113:3855-60. [PMID: 26903626 DOI: 10.1073/pnas.1515613113] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Myxoma virus (MYXV) is a rabbit-specific poxvirus, which is highly virulent in European rabbits. The attenuation of MYXV and the increased resistance of rabbits following the release of MYXV in Australia is one of the best-documented examples of host-pathogen coevolution. To elucidate the molecular mechanisms that contribute to the restriction of MYXV infection to rabbits and MYXV attenuation in the field, we have studied the interaction of the MYXV protein M156 with the host antiviral protein kinase R (PKR). In yeast and cell-culture transfection assays, M156 only inhibited rabbit PKR but not PKR from other tested mammalian species. Infection assays with human HeLa PKR knock-down cells, which were stably transfected with human or rabbit PKR, revealed that only human but not rabbit PKR was able to restrict MYXV infection, whereas both PKRs were able to restrict replication of a vaccinia virus (VACV) strain that lacks the PKR inhibitors E3 and K3. Inactivation of M156R led to MYXV virus attenuation in rabbit cells, which was rescued by the ectopic expression of VACV E3 and K3. We further show that a mutation in the M156 encoding gene that was identified in more than 50% of MYXV field isolates from Australia resulted in an M156 variant that lost its ability to inhibit rabbit PKR and led to virus attenuation. The species-specific inhibition of rabbit PKR by M156 and the M156 loss-of-function in Australian MYXV field isolates might thus contribute to the species specificity of MYXV and to the attenuation in the field, respectively.
Collapse
|
29
|
Nelson CA, Epperson ML, Singh S, Elliott JI, Fremont DH. Structural Conservation and Functional Diversity of the Poxvirus Immune Evasion (PIE) Domain Superfamily. Viruses 2015; 7:4878-98. [PMID: 26343707 PMCID: PMC4584292 DOI: 10.3390/v7092848] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 08/19/2015] [Accepted: 08/20/2015] [Indexed: 01/05/2023] Open
Abstract
Poxviruses encode a broad array of proteins that serve to undermine host immune defenses. Structural analysis of four of these seemingly unrelated proteins revealed the recurrent use of a conserved beta-sandwich fold that has not been observed in any eukaryotic or prokaryotic protein. Herein we propose to call this unique structural scaffolding the PIE (Poxvirus Immune Evasion) domain. PIE domain containing proteins are abundant in chordopoxvirinae, with our analysis identifying 20 likely PIE subfamilies among 33 representative genomes spanning 7 genera. For example, cowpox strain Brighton Red appears to encode 10 different PIEs: vCCI, A41, C8, M2, T4 (CPVX203), and the SECRET proteins CrmB, CrmD, SCP-1, SCP-2, and SCP-3. Characterized PIE proteins all appear to be nonessential for virus replication, and all contain signal peptides for targeting to the secretory pathway. The PIE subfamilies differ primarily in the number, size, and location of structural embellishments to the beta-sandwich core that confer unique functional specificities. Reported ligands include chemokines, GM-CSF, IL-2, MHC class I, and glycosaminoglycans. We expect that the list of ligands and receptors engaged by the PIE domain will grow as we come to better understand how this versatile structural architecture can be tailored to manipulate host responses to infection.
Collapse
Affiliation(s)
- Christopher A Nelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Megan L Epperson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Sukrit Singh
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Jabari I Elliott
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA.
- Department of Molecular Microbiology,Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
30
|
Guo H, Kaiser WJ, Mocarski ES. Manipulation of apoptosis and necroptosis signaling by herpesviruses. Med Microbiol Immunol 2015; 204:439-48. [PMID: 25828583 DOI: 10.1007/s00430-015-0410-5] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 03/17/2015] [Indexed: 12/18/2022]
Abstract
Like apoptosis, necroptosis is an innate immune mechanism that eliminates pathogen-infected cells. Receptor-interacting protein kinase (RIP)3 (also called RIPK3) mediates necrotic death by phosphorylating an executioner protein, MLKL, leading to plasma membrane leakage. The pathway is triggered against viruses that block caspase 8. In murine CMV, the viral inhibitor of caspase 8 activation prevents extrinsic apoptosis but also has the potential to unleash necroptosis. This virus encodes the viral inhibitor of RIP activation to prevent RIP homotypic interaction motif (RHIM)-dependent signal transduction and necroptosis. Recent investigations reveal a similar mechanism at play in the human alpha-herpesviruses, herpes simplex virus (HSV)1 and HSV2, where RHIM competitor function and caspase 8 suppression are carried out by the virus-encoded large subunit of ribonucleotide reductase (R1). In human cells, R1 inhibition of caspase 8 prevents TNF-induced apoptosis, but sensitizes to TNF-induced necroptosis. The RHIM and caspase 8 interaction domains of R1 collaborate to prevent RIP3-dependent steps and enable both herpesviruses to deflect host cell death machinery that would cut short infection. In mouse cells, HSV1 infection by itself triggers necroptosis by driving RIP3 protein kinase activity. HSV1 R1 contributes to the activation of RIP3 adaptor function in mice, a popular host animal for experimental infection. Based on these studies, infection of RIP3-kinase inactive mice should be explored in models of pathogenesis and latency. The necrotic death pathway that is suppressed during infection in the natural host becomes a cross-species barrier to infection in a non-natural host.
Collapse
Affiliation(s)
- Hongyan Guo
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | | | | |
Collapse
|
31
|
Mutational analysis of vaccinia virus E3 protein: the biological functions do not correlate with its biochemical capacity to bind double-stranded RNA. J Virol 2015; 89:5382-94. [PMID: 25740987 DOI: 10.1128/jvi.03288-14] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/23/2015] [Indexed: 01/08/2023] Open
Abstract
UNLABELLED Vaccinia E3 protein has the biochemical capacity of binding to double-stranded RNA (dsRNA). The best characterized biological functions of the E3 protein include its host range function, suppression of cytokine expression, and inhibition of interferon (IFN)-induced antiviral activity. Currently, the role of the dsRNA binding capacity in the biological functions of the E3 protein is not clear. To further understand the mechanism of the E3 protein biological functions, we performed alanine scanning of the entire dsRNA binding domain of the E3 protein to examine the link between its biochemical capacity of dsRNA binding and biological functions. Of the 115 mutants examined, 20 were defective in dsRNA binding. Although the majority of the mutants defective in dsRNA binding also showed defective replication in HeLa cells, nine mutants (I105A, Y125A, E138A, F148A, F159A, K171A, L182A, L183A, and I187/188A) retained the host range function to various degrees. Further examination of a set of representative E3L mutants showed that residues essential for dsRNA binding are not essential for the biological functions of E3 protein, such as inhibition of protein kinase R (PKR) activation, suppression of cytokine expression, and apoptosis. Thus, data described in this communication strongly indicate the E3 protein performs its biological functions via a novel mechanism which does not correlate with its dsRNA binding activity. IMPORTANCE dsRNAs produced during virus replication are important pathogen-associated molecular patterns (PAMPs) for inducing antiviral immune responses. One of the strategies used by many viruses to counteract such antiviral immune responses is achieved by producing dsRNA binding proteins, such as poxvirus E3 family proteins, influenza virus NS1, and Ebola virus V35 proteins. The most widely accepted model for the biological functions of this class of viral dsRNA binding proteins is that they bind to and sequester viral dsRNA PAMPs; thus, they suppress the related antiviral immune responses. However, no direct experimental data confirm such a model. In this study of vaccinia E3 protein, we found that the biological functions of the E3 protein are not necessarily linked to its biochemical capacity of dsRNA binding. Thus, our data strongly point to a new concept of virus modulation of cellular antiviral responses triggered by dsRNA PAMPs.
Collapse
|
32
|
Hand ES, Haller SL, Peng C, Rothenburg S, Hersperger AR. Ectopic expression of vaccinia virus E3 and K3 cannot rescue ectromelia virus replication in rabbit RK13 cells. PLoS One 2015; 10:e0119189. [PMID: 25734776 PMCID: PMC4348479 DOI: 10.1371/journal.pone.0119189] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 01/13/2015] [Indexed: 11/19/2022] Open
Abstract
As a group, poxviruses have been shown to infect a wide variety of animal species. However, there is individual variability in the range of species able to be productively infected. In this study, we observed that ectromelia virus (ECTV) does not replicate efficiently in cultured rabbit RK13 cells. Conversely, vaccinia virus (VACV) replicates well in these cells. Upon infection of RK13 cells, the replication cycle of ECTV is abortive in nature, resulting in a greatly reduced ability to spread among cells in culture. We observed ample levels of early gene expression but reduced detection of virus factories and severely blunted production of enveloped virus at the cell surface. This work focused on two important host range genes, named E3L and K3L, in VACV. Both VACV and ECTV express a functional protein product from the E3L gene, but only VACV contains an intact K3L gene. To better understand the discrepancy in replication capacity of these viruses, we examined the ability of ECTV to replicate in wild-type RK13 cells compared to cells that constitutively express E3 and K3 from VACV. The role these proteins play in the ability of VACV to replicate in RK13 cells was also analyzed to determine their individual contribution to viral replication and PKR activation. Since E3L and K3L are two relevant host range genes, we hypothesized that expression of one or both of them may have a positive impact on the ability of ECTV to replicate in RK13 cells. Using various methods to assess virus growth, we did not detect any significant differences with respect to the replication of ECTV between wild-type RK13 compared to versions of this cell line that stably expressed VACV E3 alone or in combination with K3. Therefore, there remain unanswered questions related to the factors that limit the host range of ECTV.
Collapse
Affiliation(s)
- Erin S. Hand
- Department of Biology, Albright College, Reading, Pennsylvania, United States of America
| | - Sherry L. Haller
- Laboratory for Host-Specific Virology, Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
| | - Chen Peng
- Laboratory for Host-Specific Virology, Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
| | - Stefan Rothenburg
- Laboratory for Host-Specific Virology, Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
| | - Adam R. Hersperger
- Department of Biology, Albright College, Reading, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
33
|
Hatcher EL, Hendrickson RC, Lefkowitz EJ. Identification of nucleotide-level changes impacting gene content and genome evolution in orthopoxviruses. J Virol 2014; 88:13651-68. [PMID: 25231308 PMCID: PMC4248964 DOI: 10.1128/jvi.02015-14] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 09/08/2014] [Indexed: 01/20/2023] Open
Abstract
UNLABELLED Poxviruses are composed of large double-stranded DNA (dsDNA) genomes coding for several hundred genes whose variation has supported virus adaptation to a wide variety of hosts over their long evolutionary history. Comparative genomics has suggested that the Orthopoxvirus genus in particular has undergone reductive evolution, with the most recent common ancestor likely possessing a gene complement consisting of all genes present in any existing modern-day orthopoxvirus species, similar to the current Cowpox virus species. As orthopoxviruses adapt to new environments, the selection pressure on individual genes may be altered, driving sequence divergence and possible loss of function. This is evidenced by accumulation of mutations and loss of protein-coding open reading frames (ORFs) that progress from individual missense mutations to gene truncation through the introduction of early stop mutations (ESMs), gene fragmentation, and in some cases, a total loss of the ORF. In this study, we have constructed a whole-genome alignment for representative isolates from each Orthopoxvirus species and used it to identify the nucleotide-level changes that have led to gene content variation. By identifying the changes that have led to ESMs, we were able to determine that short indels were the major cause of gene truncations and that the genome length is inversely proportional to the number of ESMs present. We also identified the number and types of protein functional motifs still present in truncated genes to assess their functional significance. IMPORTANCE This work contributes to our understanding of reductive evolution in poxviruses by identifying genomic remnants such as single nucleotide polymorphisms (SNPs) and indels left behind by evolutionary processes. Our comprehensive analysis of the genomic changes leading to gene truncation and fragmentation was able to detect some of the remnants of these evolutionary processes still present in orthopoxvirus genomes and suggests that these viruses are under continual adaptation due to changes in their environment. These results further our understanding of the evolutionary mechanisms that drive virus variation, allowing orthopoxviruses to adapt to particular environmental niches. Understanding the evolutionary history of these virus pathogens may help predict their future evolutionary potential.
Collapse
Affiliation(s)
- Eneida L Hatcher
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Elliot J Lefkowitz
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
34
|
Baker KS, Murcia PR. Poxviruses in bats … so what? Viruses 2014; 6:1564-77. [PMID: 24704730 PMCID: PMC4014710 DOI: 10.3390/v6041564] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/13/2014] [Accepted: 03/17/2014] [Indexed: 12/23/2022] Open
Abstract
Poxviruses are important pathogens of man and numerous domestic and wild animal species. Cross species (including zoonotic) poxvirus infections can have drastic consequences for the recipient host. Bats are a diverse order of mammals known to carry lethal viral zoonoses such as Rabies, Hendra, Nipah, and SARS. Consequent targeted research is revealing bats to be infected with a rich diversity of novel viruses. Poxviruses were recently identified in bats and the settings in which they were found were dramatically different. Here, we review the natural history of poxviruses in bats and highlight the relationship of the viruses to each other and their context in the Poxviridae family. In addition to considering the zoonotic potential of these viruses, we reflect on the broader implications of these findings. Specifically, the potential to explore and exploit this newfound relationship to study coevolution and cross species transmission together with fundamental aspects of poxvirus host tropism as well as bat virology and immunology.
Collapse
Affiliation(s)
- Kate S Baker
- Wellcome Trust Sanger Institute, Hinxton, CB10 1SA, UK.
| | - Pablo R Murcia
- University of Glasgow Centre for Virus Research, Institute of Infection, Inflammation and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G61 1QH, UK.
| |
Collapse
|
35
|
Kugelman JR, Johnston SC, Mulembakani PM, Kisalu N, Lee MS, Koroleva G, McCarthy SE, Gestole MC, Wolfe ND, Fair JN, Schneider BS, Wright LL, Huggins J, Whitehouse CA, Wemakoy EO, Muyembe-Tamfum JJ, Hensley LE, Palacios GF, Rimoin AW. Genomic variability of monkeypox virus among humans, Democratic Republic of the Congo. Emerg Infect Dis 2014; 20:232-9. [PMID: 24457084 PMCID: PMC3901482 DOI: 10.3201/eid2002.130118] [Citation(s) in RCA: 198] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Monkeypox virus is a zoonotic virus endemic to Central Africa. Although active disease surveillance has assessed monkeypox disease prevalence and geographic range, information about virus diversity is lacking. We therefore assessed genome diversity of viruses in 60 samples obtained from humans with primary and secondary cases of infection from 2005 through 2007. We detected 4 distinct lineages and a deletion that resulted in gene loss in 10 (16.7%) samples and that seemed to correlate with human-to-human transmission (p = 0.0544). The data suggest a high frequency of spillover events from the pool of viruses in nonhuman animals, active selection through genomic destabilization and gene loss, and increased disease transmissibility and severity. The potential for accelerated adaptation to humans should be monitored through improved surveillance.
Collapse
Affiliation(s)
| | | | - Prime M. Mulembakani
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA (J.R. Kugelman, S.C. Johnston, M.S. Lee, G. Koroleva, S.E. McCarthy, M.C. Gestole, J. Huggins, C.A. Whitehouse, G.F. Palacios)
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo (P.M. Mulembakani, E.O. Wemakoy)
- University of California, Los Angeles, California, USA (N. Kisalu, A.W. Rimoin)
- Global Viral Forecasting (now known as Metabiota), San Francisco, California, USA (N.D. Wolfe, J.N, Fair, B.S. Schneider)
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA (L.L. Wright)
- National Institute of Biomedical Research, Kinshasa (J.J. Muyembe-Tamfum)
- US Food and Drug Administration, Silver Spring, Maryland, USA (L.E. Hensley)
| | - Neville Kisalu
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA (J.R. Kugelman, S.C. Johnston, M.S. Lee, G. Koroleva, S.E. McCarthy, M.C. Gestole, J. Huggins, C.A. Whitehouse, G.F. Palacios)
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo (P.M. Mulembakani, E.O. Wemakoy)
- University of California, Los Angeles, California, USA (N. Kisalu, A.W. Rimoin)
- Global Viral Forecasting (now known as Metabiota), San Francisco, California, USA (N.D. Wolfe, J.N, Fair, B.S. Schneider)
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA (L.L. Wright)
- National Institute of Biomedical Research, Kinshasa (J.J. Muyembe-Tamfum)
- US Food and Drug Administration, Silver Spring, Maryland, USA (L.E. Hensley)
| | - Michael S. Lee
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA (J.R. Kugelman, S.C. Johnston, M.S. Lee, G. Koroleva, S.E. McCarthy, M.C. Gestole, J. Huggins, C.A. Whitehouse, G.F. Palacios)
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo (P.M. Mulembakani, E.O. Wemakoy)
- University of California, Los Angeles, California, USA (N. Kisalu, A.W. Rimoin)
- Global Viral Forecasting (now known as Metabiota), San Francisco, California, USA (N.D. Wolfe, J.N, Fair, B.S. Schneider)
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA (L.L. Wright)
- National Institute of Biomedical Research, Kinshasa (J.J. Muyembe-Tamfum)
- US Food and Drug Administration, Silver Spring, Maryland, USA (L.E. Hensley)
| | - Galina Koroleva
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA (J.R. Kugelman, S.C. Johnston, M.S. Lee, G. Koroleva, S.E. McCarthy, M.C. Gestole, J. Huggins, C.A. Whitehouse, G.F. Palacios)
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo (P.M. Mulembakani, E.O. Wemakoy)
- University of California, Los Angeles, California, USA (N. Kisalu, A.W. Rimoin)
- Global Viral Forecasting (now known as Metabiota), San Francisco, California, USA (N.D. Wolfe, J.N, Fair, B.S. Schneider)
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA (L.L. Wright)
- National Institute of Biomedical Research, Kinshasa (J.J. Muyembe-Tamfum)
- US Food and Drug Administration, Silver Spring, Maryland, USA (L.E. Hensley)
| | - Sarah E. McCarthy
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA (J.R. Kugelman, S.C. Johnston, M.S. Lee, G. Koroleva, S.E. McCarthy, M.C. Gestole, J. Huggins, C.A. Whitehouse, G.F. Palacios)
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo (P.M. Mulembakani, E.O. Wemakoy)
- University of California, Los Angeles, California, USA (N. Kisalu, A.W. Rimoin)
- Global Viral Forecasting (now known as Metabiota), San Francisco, California, USA (N.D. Wolfe, J.N, Fair, B.S. Schneider)
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA (L.L. Wright)
- National Institute of Biomedical Research, Kinshasa (J.J. Muyembe-Tamfum)
- US Food and Drug Administration, Silver Spring, Maryland, USA (L.E. Hensley)
| | - Marie C. Gestole
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA (J.R. Kugelman, S.C. Johnston, M.S. Lee, G. Koroleva, S.E. McCarthy, M.C. Gestole, J. Huggins, C.A. Whitehouse, G.F. Palacios)
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo (P.M. Mulembakani, E.O. Wemakoy)
- University of California, Los Angeles, California, USA (N. Kisalu, A.W. Rimoin)
- Global Viral Forecasting (now known as Metabiota), San Francisco, California, USA (N.D. Wolfe, J.N, Fair, B.S. Schneider)
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA (L.L. Wright)
- National Institute of Biomedical Research, Kinshasa (J.J. Muyembe-Tamfum)
- US Food and Drug Administration, Silver Spring, Maryland, USA (L.E. Hensley)
| | - Nathan D. Wolfe
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA (J.R. Kugelman, S.C. Johnston, M.S. Lee, G. Koroleva, S.E. McCarthy, M.C. Gestole, J. Huggins, C.A. Whitehouse, G.F. Palacios)
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo (P.M. Mulembakani, E.O. Wemakoy)
- University of California, Los Angeles, California, USA (N. Kisalu, A.W. Rimoin)
- Global Viral Forecasting (now known as Metabiota), San Francisco, California, USA (N.D. Wolfe, J.N, Fair, B.S. Schneider)
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA (L.L. Wright)
- National Institute of Biomedical Research, Kinshasa (J.J. Muyembe-Tamfum)
- US Food and Drug Administration, Silver Spring, Maryland, USA (L.E. Hensley)
| | - Joseph N. Fair
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA (J.R. Kugelman, S.C. Johnston, M.S. Lee, G. Koroleva, S.E. McCarthy, M.C. Gestole, J. Huggins, C.A. Whitehouse, G.F. Palacios)
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo (P.M. Mulembakani, E.O. Wemakoy)
- University of California, Los Angeles, California, USA (N. Kisalu, A.W. Rimoin)
- Global Viral Forecasting (now known as Metabiota), San Francisco, California, USA (N.D. Wolfe, J.N, Fair, B.S. Schneider)
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA (L.L. Wright)
- National Institute of Biomedical Research, Kinshasa (J.J. Muyembe-Tamfum)
- US Food and Drug Administration, Silver Spring, Maryland, USA (L.E. Hensley)
| | - Bradley S. Schneider
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA (J.R. Kugelman, S.C. Johnston, M.S. Lee, G. Koroleva, S.E. McCarthy, M.C. Gestole, J. Huggins, C.A. Whitehouse, G.F. Palacios)
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo (P.M. Mulembakani, E.O. Wemakoy)
- University of California, Los Angeles, California, USA (N. Kisalu, A.W. Rimoin)
- Global Viral Forecasting (now known as Metabiota), San Francisco, California, USA (N.D. Wolfe, J.N, Fair, B.S. Schneider)
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA (L.L. Wright)
- National Institute of Biomedical Research, Kinshasa (J.J. Muyembe-Tamfum)
- US Food and Drug Administration, Silver Spring, Maryland, USA (L.E. Hensley)
| | - Linda L. Wright
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA (J.R. Kugelman, S.C. Johnston, M.S. Lee, G. Koroleva, S.E. McCarthy, M.C. Gestole, J. Huggins, C.A. Whitehouse, G.F. Palacios)
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo (P.M. Mulembakani, E.O. Wemakoy)
- University of California, Los Angeles, California, USA (N. Kisalu, A.W. Rimoin)
- Global Viral Forecasting (now known as Metabiota), San Francisco, California, USA (N.D. Wolfe, J.N, Fair, B.S. Schneider)
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA (L.L. Wright)
- National Institute of Biomedical Research, Kinshasa (J.J. Muyembe-Tamfum)
- US Food and Drug Administration, Silver Spring, Maryland, USA (L.E. Hensley)
| | - John Huggins
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA (J.R. Kugelman, S.C. Johnston, M.S. Lee, G. Koroleva, S.E. McCarthy, M.C. Gestole, J. Huggins, C.A. Whitehouse, G.F. Palacios)
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo (P.M. Mulembakani, E.O. Wemakoy)
- University of California, Los Angeles, California, USA (N. Kisalu, A.W. Rimoin)
- Global Viral Forecasting (now known as Metabiota), San Francisco, California, USA (N.D. Wolfe, J.N, Fair, B.S. Schneider)
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA (L.L. Wright)
- National Institute of Biomedical Research, Kinshasa (J.J. Muyembe-Tamfum)
- US Food and Drug Administration, Silver Spring, Maryland, USA (L.E. Hensley)
| | - Chris A. Whitehouse
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA (J.R. Kugelman, S.C. Johnston, M.S. Lee, G. Koroleva, S.E. McCarthy, M.C. Gestole, J. Huggins, C.A. Whitehouse, G.F. Palacios)
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo (P.M. Mulembakani, E.O. Wemakoy)
- University of California, Los Angeles, California, USA (N. Kisalu, A.W. Rimoin)
- Global Viral Forecasting (now known as Metabiota), San Francisco, California, USA (N.D. Wolfe, J.N, Fair, B.S. Schneider)
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA (L.L. Wright)
- National Institute of Biomedical Research, Kinshasa (J.J. Muyembe-Tamfum)
- US Food and Drug Administration, Silver Spring, Maryland, USA (L.E. Hensley)
| | - Emile Okitolonda Wemakoy
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA (J.R. Kugelman, S.C. Johnston, M.S. Lee, G. Koroleva, S.E. McCarthy, M.C. Gestole, J. Huggins, C.A. Whitehouse, G.F. Palacios)
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo (P.M. Mulembakani, E.O. Wemakoy)
- University of California, Los Angeles, California, USA (N. Kisalu, A.W. Rimoin)
- Global Viral Forecasting (now known as Metabiota), San Francisco, California, USA (N.D. Wolfe, J.N, Fair, B.S. Schneider)
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA (L.L. Wright)
- National Institute of Biomedical Research, Kinshasa (J.J. Muyembe-Tamfum)
- US Food and Drug Administration, Silver Spring, Maryland, USA (L.E. Hensley)
| | - Jean Jacques Muyembe-Tamfum
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA (J.R. Kugelman, S.C. Johnston, M.S. Lee, G. Koroleva, S.E. McCarthy, M.C. Gestole, J. Huggins, C.A. Whitehouse, G.F. Palacios)
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo (P.M. Mulembakani, E.O. Wemakoy)
- University of California, Los Angeles, California, USA (N. Kisalu, A.W. Rimoin)
- Global Viral Forecasting (now known as Metabiota), San Francisco, California, USA (N.D. Wolfe, J.N, Fair, B.S. Schneider)
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA (L.L. Wright)
- National Institute of Biomedical Research, Kinshasa (J.J. Muyembe-Tamfum)
- US Food and Drug Administration, Silver Spring, Maryland, USA (L.E. Hensley)
| | - Lisa E. Hensley
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA (J.R. Kugelman, S.C. Johnston, M.S. Lee, G. Koroleva, S.E. McCarthy, M.C. Gestole, J. Huggins, C.A. Whitehouse, G.F. Palacios)
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo (P.M. Mulembakani, E.O. Wemakoy)
- University of California, Los Angeles, California, USA (N. Kisalu, A.W. Rimoin)
- Global Viral Forecasting (now known as Metabiota), San Francisco, California, USA (N.D. Wolfe, J.N, Fair, B.S. Schneider)
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA (L.L. Wright)
- National Institute of Biomedical Research, Kinshasa (J.J. Muyembe-Tamfum)
- US Food and Drug Administration, Silver Spring, Maryland, USA (L.E. Hensley)
| | | | | |
Collapse
|
36
|
Haller SL, Peng C, McFadden G, Rothenburg S. Poxviruses and the evolution of host range and virulence. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2014; 21:15-40. [PMID: 24161410 PMCID: PMC3945082 DOI: 10.1016/j.meegid.2013.10.014] [Citation(s) in RCA: 195] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 10/15/2013] [Accepted: 10/15/2013] [Indexed: 11/22/2022]
Abstract
Poxviruses as a group can infect a large number of animals. However, at the level of individual viruses, even closely related poxviruses display highly diverse host ranges and virulence. For example, variola virus, the causative agent of smallpox, is human-specific and highly virulent only to humans, whereas related cowpox viruses naturally infect a broad spectrum of animals and only cause relatively mild disease in humans. The successful replication of poxviruses depends on their effective manipulation of the host antiviral responses, at the cellular-, tissue- and species-specific levels, which constitutes a molecular basis for differences in poxvirus host range and virulence. A number of poxvirus genes have been identified that possess host range function in experimental settings, and many of these host range genes target specific antiviral host pathways. Herein, we review the biology of poxviruses with a focus on host range, zoonotic infections, virulence, genomics and host range genes as well as the current knowledge about the function of poxvirus host range factors and how their interaction with the host innate immune system contributes to poxvirus host range and virulence. We further discuss the evolution of host range and virulence in poxviruses as well as host switches and potential poxvirus threats for human and animal health.
Collapse
Affiliation(s)
- Sherry L Haller
- Laboratory for Host-Specific Virology, Division of Biology, Kansas State University, KS 66506, USA
| | - Chen Peng
- Laboratory for Host-Specific Virology, Division of Biology, Kansas State University, KS 66506, USA
| | - Grant McFadden
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA
| | - Stefan Rothenburg
- Laboratory for Host-Specific Virology, Division of Biology, Kansas State University, KS 66506, USA.
| |
Collapse
|
37
|
Dabrowski PW, Radonić A, Kurth A, Nitsche A. Genome-wide comparison of cowpox viruses reveals a new clade related to Variola virus. PLoS One 2013; 8:e79953. [PMID: 24312452 PMCID: PMC3848979 DOI: 10.1371/journal.pone.0079953] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 09/27/2013] [Indexed: 11/19/2022] Open
Abstract
Zoonotic infections caused by several orthopoxviruses (OPV) like monkeypox virus or vaccinia virus have a significant impact on human health. In Europe, the number of diagnosed infections with cowpox viruses (CPXV) is increasing in animals as well as in humans. CPXV used to be enzootic in cattle; however, such infections were not being diagnosed over the last decades. Instead, individual cases of cowpox are being found in cats or exotic zoo animals that transmit the infection to humans. Both animals and humans reveal local exanthema on arms and legs or on the face. Although cowpox is generally regarded as a self-limiting disease, immunosuppressed patients can develop a lethal systemic disease resembling smallpox. To date, only limited information on the complex and, compared to other OPV, sparsely conserved CPXV genomes is available. Since CPXV displays the widest host range of all OPV known, it seems important to comprehend the genetic repertoire of CPXV which in turn may help elucidate specific mechanisms of CPXV pathogenesis and origin. Therefore, 22 genomes of independent CPXV strains from clinical cases, involving ten humans, four rats, two cats, two jaguarundis, one beaver, one elephant, one marah and one mongoose, were sequenced by using massive parallel pyrosequencing. The extensive phylogenetic analysis showed that the CPXV strains sequenced clearly cluster into several distinct clades, some of which are closely related to Vaccinia viruses while others represent different clades in a CPXV cluster. Particularly one CPXV clade is more closely related to Camelpox virus, Taterapox virus and Variola virus than to any other known OPV. These results support and extend recent data from other groups who postulate that CPXV does not form a monophyletic clade and should be divided into multiple lineages.
Collapse
Affiliation(s)
- Piotr Wojtek Dabrowski
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Aleksandar Radonić
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Andreas Kurth
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Andreas Nitsche
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
- * E-mail:
| |
Collapse
|
38
|
Liu Z, Wang S, Zhang Q, Tian M, Hou J, Wang R, Liu C, Ji X, Liu Y, Shao Y. Deletion of C7L and K1L genes leads to significantly decreased virulence of recombinant vaccinia virus TianTan. PLoS One 2013; 8:e68115. [PMID: 23840887 PMCID: PMC3698190 DOI: 10.1371/journal.pone.0068115] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 05/24/2013] [Indexed: 11/19/2022] Open
Abstract
The vaccinia virus TianTan (VTT) has been modified as an HIV vaccine vector in China and has shown excellent performance in immunogenicity and safety. However, its adverse effects in immunosuppressed individuals warrant the search for a safer vector in the following clinic trails. In this study, we deleted the C7L and K1L genes of VTT and constructed six recombinant vaccinia strains VTT△C7L, VTT△K1L, VTT△C7LK1L, VTKgpe△C7L, VTKgpe△K1L and VTT△C7LK1L-gag. The pathogenicity and immunogenicity of these recombinants were evaluated in mouse and rabbit models. Comparing to parental VTT, VTT△C7L and VTT△K1L showed significantly decreased replication capability in CEF, Vero, BHK-21 and HeLa cell lines. In particular, replication of VTT△C7LK1L decreased more than 10-fold in all four cell lines. The virulence of all these mutants were decreased in BALB/c mouse and rabbit models; VTT△C7LK1L once again showed the greatest attenuation, having resulted in no evident damage in mice and erythema of only 0.4 cm diameter in rabbits, compared to 1.48 cm for VTT. VTKgpe△C7L, VTKgpe△K1L and VTT△C7LK1L-gag elicited as strong cellular and humoral responses against HIV genes as did VTKgpe, while humoral immune response against the vaccinia itself was reduced by 4-8-fold. These data show that deletion of C7L and K1L genes leads to significantly decreased virulence without compromising animal host immunogenicity, and may thus be key to creating a more safe and effective HIV vaccine vector.
Collapse
Affiliation(s)
- Zheng Liu
- Division of Research on Virology and Immunology, State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention (NCAIDS), China CDC, Beijing, China
| | - Shuhui Wang
- Division of Research on Virology and Immunology, State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention (NCAIDS), China CDC, Beijing, China
| | - Qicheng Zhang
- Division of Research on Virology and Immunology, State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention (NCAIDS), China CDC, Beijing, China
- Key Laboratory of Molecular Microbiology and Biotechnology (Ministry of Education) and Key Laboratory of Microbial Functional Genomics (Tianjin), College of Life Sciences, Nankai University, Tianjin, China
| | - Meijuan Tian
- Division of Infectious Diseases & HIV Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Jue Hou
- Division of Research on Virology and Immunology, State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention (NCAIDS), China CDC, Beijing, China
| | - Rongmin Wang
- Clinical Laboratory, Zhaoyuan CDC, Zhaoyuan, China
| | - Chang Liu
- Division of Research on Virology and Immunology, State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention (NCAIDS), China CDC, Beijing, China
| | - Xu Ji
- Division of Research on Virology and Immunology, State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention (NCAIDS), China CDC, Beijing, China
| | - Ying Liu
- Division of Research on Virology and Immunology, State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention (NCAIDS), China CDC, Beijing, China
| | - Yiming Shao
- Division of Research on Virology and Immunology, State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention (NCAIDS), China CDC, Beijing, China
| |
Collapse
|
39
|
Colson P, De Lamballerie X, Yutin N, Asgari S, Bigot Y, Bideshi DK, Cheng XW, Federici BA, Van Etten JL, Koonin EV, La Scola B, Raoult D. "Megavirales", a proposed new order for eukaryotic nucleocytoplasmic large DNA viruses. Arch Virol 2013; 158:2517-21. [PMID: 23812617 DOI: 10.1007/s00705-013-1768-6] [Citation(s) in RCA: 192] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 05/07/2013] [Indexed: 02/06/2023]
Abstract
The nucleocytoplasmic large DNA viruses (NCLDVs) comprise a monophyletic group of viruses that infect animals and diverse unicellular eukaryotes. The NCLDV group includes the families Poxviridae, Asfarviridae, Iridoviridae, Ascoviridae, Phycodnaviridae, Mimiviridae and the proposed family "Marseilleviridae". The family Mimiviridae includes the largest known viruses, with genomes in excess of one megabase, whereas the genome size in the other NCLDV families varies from 100 to 400 kilobase pairs. Most of the NCLDVs replicate in the cytoplasm of infected cells, within so-called virus factories. The NCLDVs share a common ancient origin, as demonstrated by evolutionary reconstructions that trace approximately 50 genes encoding key proteins involved in viral replication and virion formation to the last common ancestor of all these viruses. Taken together, these characteristics lead us to propose assigning an official taxonomic rank to the NCLDVs as the order "Megavirales", in reference to the large size of the virions and genomes of these viruses.
Collapse
Affiliation(s)
- Philippe Colson
- Unité des Rickettsies, URMITE UMR CNRS 7278 IRD 198 INSERM U1095, Facultés de Médecine et de Pharmacie, IHU Méditerranée Infection, Aix-Marseille Université, 27 Boulevard Jean Moulin, 13385, Marseille Cedex 05, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Bratke KA, McLysaght A, Rothenburg S. A survey of host range genes in poxvirus genomes. INFECTION GENETICS AND EVOLUTION 2012; 14:406-25. [PMID: 23268114 DOI: 10.1016/j.meegid.2012.12.002] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 12/01/2012] [Accepted: 12/06/2012] [Indexed: 12/17/2022]
Abstract
Poxviruses are widespread pathogens, which display extremely different host ranges. Whereas some poxviruses, including variola virus, display narrow host ranges, others such as cowpox viruses naturally infect a wide range of mammals. The molecular basis for differences in host range are poorly understood but apparently depend on the successful manipulation of the host antiviral response. Some poxvirus genes have been shown to confer host tropism in experimental settings and are thus called host range factors. Identified host range genes include vaccinia virus K1L, K3L, E3L, B5R, C7L and SPI-1, cowpox virus CP77/CHOhr, ectromelia virus p28 and 022, and myxoma virus T2, T4, T5, 11L, 13L, 062R and 063R. These genes encode for ankyrin repeat-containing proteins, tumor necrosis factor receptor II homologs, apoptosis inhibitor T4-related proteins, Bcl-2-related proteins, pyrin domain-containing proteins, cellular serine protease inhibitors (serpins), short complement-like repeats containing proteins, KilA-N/RING domain-containing proteins, as well as inhibitors of the double-stranded RNA-activated protein kinase PKR. We conducted a systematic survey for the presence of known host range genes and closely related family members in poxvirus genomes, classified them into subgroups based on their phylogenetic relationship and correlated their presence with the poxvirus phylogeny. Common themes in the evolution of poxvirus host range genes are lineage-specific duplications and multiple independent inactivation events. Our analyses yield new insights into the evolution of poxvirus host range genes. Implications of our findings for poxvirus host range and virulence are discussed.
Collapse
Affiliation(s)
- Kirsten A Bratke
- Smurfit Institute of Genetics, University of Dublin, Trinity College, Dublin 2, Ireland
| | | | | |
Collapse
|
41
|
Abstract
Viruses with large genomes encode numerous proteins that do not directly participate in virus biogenesis but rather modify key functional systems of infected cells. We report that a distinct group of giant viruses infecting unicellular eukaryotes that includes Organic Lake Phycodnaviruses and Phaeocystis globosa virus encode predicted proteorhodopsins that have not been previously detected in viruses. Search of metagenomic sequence data shows that putative viral proteorhodopsins are extremely abundant in marine environments. Phylogenetic analysis suggests that giant viruses acquired proteorhodopsins via horizontal gene transfer from proteorhodopsin-encoding protists although the actual donor(s) could not be presently identified. The pattern of conservation of the predicted functionally important amino acid residues suggests that viral proteorhodopsin homologs function as sensory rhodopsins. We hypothesize that viral rhodopsins modulate light-dependent signaling, in particular phototaxis, in infected protists. This article was reviewed by Igor B. Zhulin and Laksminarayan M. Iyer. For the full reviews, see the Reviewers’ reports section.
Collapse
|
42
|
Elde NC, Child SJ, Eickbush MT, Kitzman JO, Rogers KS, Shendure J, Geballe AP, Malik HS. Poxviruses deploy genomic accordions to adapt rapidly against host antiviral defenses. Cell 2012; 150:831-41. [PMID: 22901812 PMCID: PMC3499626 DOI: 10.1016/j.cell.2012.05.049] [Citation(s) in RCA: 235] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 05/29/2012] [Accepted: 05/30/2012] [Indexed: 12/17/2022]
Abstract
In contrast to RNA viruses, double-stranded DNA viruses have low mutation rates yet must still adapt rapidly in response to changing host defenses. To determine mechanisms of adaptation, we subjected the model poxvirus vaccinia to serial propagation in human cells, where its antihost factor K3L is maladapted against the antiviral protein kinase R (PKR). Viruses rapidly acquired higher fitness via recurrent K3L gene amplifications, incurring up to 7%-10% increases in genome size. These transient gene expansions were necessary and sufficient to counteract human PKR and facilitated the gain of an adaptive amino acid substitution in K3L that also defeats PKR. Subsequent reductions in gene amplifications offset the costs associated with larger genome size while retaining adaptive substitutions. Our discovery of viral "gene-accordions" explains how poxviruses can rapidly adapt to defeat different host defenses despite low mutation rates and reveals how classical Red Queen conflicts can progress through unrecognized intermediates.
Collapse
Affiliation(s)
- Nels C. Elde
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Stephanie J. Child
- Human Biology, and Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Michael T. Eickbush
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Jacob O. Kitzman
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Kelsey S. Rogers
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Adam P. Geballe
- Human Biology, and Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Department of Medicine, and University of Washington, Seattle, WA, 98195, USA
- Department of Microbiology, University of Washington, Seattle, WA, 98195, USA
| | - Harmit S. Malik
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Howard Hughes Medical Institute, Seattle, WA, 98109, USA
| |
Collapse
|
43
|
Rahbar R, Rogers E, Murooka T, Kislinger T, Fish EN. Glomulin: a permissivity factor for vaccinia virus infection. J Interferon Cytokine Res 2012; 32:127-37. [PMID: 22280104 DOI: 10.1089/jir.2011.0100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In earlier studies we provided evidence that vaccinia virus (VACV) phosphorylation-activation of host cell signaling effectors is critical for subsequent viral replication. In this report, using mass spectrometry-based proteomics, we have identified 387 host cell proteins that co-immunoprecipitate with VACV in infected, permissive PM1.CCR5 human T cells. Among these, glomulin was distinguishable based on its known interaction with a tyrosine kinase receptor, c-Met, its ability to become tyrosine-phosphorylated, and its association with signaling effectors. siRNA knockdown of glomulin expression in PM1.CCR5 T cells reduces VACV infection. Glomulin interacts with the inactive, nonphosphorylated form of c-MET. We demonstrate that treatment of PM1.CCR5 T cells with a c-Met phosphorylation inhibitor leads to a significant reduction in VACV infectivity. Additionally, inhibition of phosphorylation of c-Met abrogates VACV-inducible phosphorylation of Erk 1/2 and IRS-2, signaling effectors identified as critical for VACV infection. These data identify glomulin as a permissivity factor for VACV infection and as a potential therapeutic target for inhibition of VACV infection.
Collapse
Affiliation(s)
- Ramtin Rahbar
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
44
|
Gilbert C, Pace JK, Feschotte C. Horizontal SPINning of transposons. Commun Integr Biol 2011; 2:117-9. [PMID: 19704906 DOI: 10.4161/cib.7720] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Accepted: 12/23/2008] [Indexed: 01/26/2023] Open
Abstract
The term 'horizontal transfer (HT)' refers to the transfer of genetic material between two reproductively isolated organisms. HT is thought to occur rarely in eukaryotes compared to vertical inheritance, the transmission of DNA from parent to offspring. In a recent study we have provided evidence that a family of DNA transposons, called SPACE INVADERS or SPIN, independently invaded horizontally the genome of seven distantly related tetrapod species and subsequently amplified to high copy number in each of them. This discovery calls for further investigations to better characterize the extent to which genomes have been shaped through HT events. In this addendum, we briefly discuss some general issues regarding the study of HT and further speculate on the sequence of events that could explain the current taxonomic distribution of SPIN. We propose that the presence of SPIN in the opossum (Monodelphis domestica), a taxon endemic to South America, reflects a transoceanic HT event that occurred from Old to New World, between 46 and 15 million years ago.
Collapse
Affiliation(s)
- Clément Gilbert
- Department of Biology; University of Texas at Arlington; Arlington, TX USA
| | | | | |
Collapse
|
45
|
Rice AD, Adams MM, Lampert B, Foster S, Lanier R, Robertson A, Painter G, Moyer RW. Efficacy of CMX001 as a prophylactic and presymptomatic antiviral agent in New Zealand white rabbits infected with rabbitpox virus, a model for orthopoxvirus infections of humans. Viruses 2011; 3:63-82. [PMID: 21369346 PMCID: PMC3045966 DOI: 10.3390/v3020063] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 01/04/2011] [Indexed: 01/24/2023] Open
Abstract
CMX001, a lipophilic nucleotide analog formed by covalently linking 3-(hexdecyloxy)propan-1-ol to cidofovir (CDV), is being developed as a treatment for smallpox. CMX001 has dramatically increased potency versus CDV against all dsDNA viruses and, in contrast to CDV, is orally available and has shown no evidence of nephrotoxicity in healthy volunteers or severely ill transplant patients to date. Although smallpox has been eliminated from the environment, treatments are urgently being sought due to the risk of smallpox being used as a bioterrorism agent and for monkeypox virus, a zoonotic disease of Africa, and adverse reactions to smallpox virus vaccinations. In the absence of human cases of smallpox, new treatments must be tested for efficacy in animal models. Here we first review and discuss the rabbitpox virus (RPV) infection of New Zealand White rabbits as a model for smallpox to test the efficacy of CMX001 as a prophylactic and early disease antiviral. Our results should also be applicable to monkeypox virus infections and for treatment of adverse reactions to smallpox vaccination.
Collapse
Affiliation(s)
- Amanda D. Rice
- Department of Molecular Genetics and Microbiology, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32610, USA; E-Mails: (A.D.R.); (M.M.A.)
| | - Mathew M. Adams
- Department of Molecular Genetics and Microbiology, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32610, USA; E-Mails: (A.D.R.); (M.M.A.)
| | - Bernhard Lampert
- Chimerix, Inc., 2505 Meridian Parkway Suite, 340 Durham, NC 27713, USA; E-Mails: (B.L.); (S.F.); (R.L.); (A.R.); (G.P.)
| | - Scott Foster
- Chimerix, Inc., 2505 Meridian Parkway Suite, 340 Durham, NC 27713, USA; E-Mails: (B.L.); (S.F.); (R.L.); (A.R.); (G.P.)
| | - Randall Lanier
- Chimerix, Inc., 2505 Meridian Parkway Suite, 340 Durham, NC 27713, USA; E-Mails: (B.L.); (S.F.); (R.L.); (A.R.); (G.P.)
| | - Alice Robertson
- Chimerix, Inc., 2505 Meridian Parkway Suite, 340 Durham, NC 27713, USA; E-Mails: (B.L.); (S.F.); (R.L.); (A.R.); (G.P.)
| | - George Painter
- Chimerix, Inc., 2505 Meridian Parkway Suite, 340 Durham, NC 27713, USA; E-Mails: (B.L.); (S.F.); (R.L.); (A.R.); (G.P.)
| | - Richard W. Moyer
- Department of Molecular Genetics and Microbiology, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32610, USA; E-Mails: (A.D.R.); (M.M.A.)
| |
Collapse
|
46
|
Characterization of indels in poxvirus genomes. Virus Genes 2010; 42:171-7. [PMID: 21153876 DOI: 10.1007/s11262-010-0560-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Accepted: 12/01/2010] [Indexed: 02/06/2023]
Abstract
By comparing sets of variola virus (VARV) genomes and sets of vaccinia virus (VACV) genomes, it was found that the insertion and deletion of small pieces of DNA (3-25 nucleotides) were common events among these poxviruses. Insertion events were characterized by the creation of tandem direct repeats, whereas the deletion events generally took place between two direct repeats that were separated by a few nucleotides. A number of the VARV and VACV indels clearly did not fit the expected phylogenetic tree patterns. Some of these were found to be the result of coincident events, but others, in VACV, suggest recombination among the VACV genomes. Such recombination would make the construction of phylogenetic trees problematic. The growth of VACV under artificial conditions and at high multiplicities does not select against these deletions.
Collapse
|
47
|
Hendrickson RC, Wang C, Hatcher EL, Lefkowitz EJ. Orthopoxvirus genome evolution: the role of gene loss. Viruses 2010; 2:1933-1967. [PMID: 21994715 PMCID: PMC3185746 DOI: 10.3390/v2091933] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Revised: 08/25/2010] [Accepted: 09/01/2010] [Indexed: 12/26/2022] Open
Abstract
Poxviruses are highly successful pathogens, known to infect a variety of hosts. The family Poxviridae includes Variola virus, the causative agent of smallpox, which has been eradicated as a public health threat but could potentially reemerge as a bioterrorist threat. The risk scenario includes other animal poxviruses and genetically engineered manipulations of poxviruses. Studies of orthologous gene sets have established the evolutionary relationships of members within the Poxviridae family. It is not clear, however, how variations between family members arose in the past, an important issue in understanding how these viruses may vary and possibly produce future threats. Using a newly developed poxvirus-specific tool, we predicted accurate gene sets for viruses with completely sequenced genomes in the genus Orthopoxvirus. Employing sensitive sequence comparison techniques together with comparison of syntenic gene maps, we established the relationships between all viral gene sets. These techniques allowed us to unambiguously identify the gene loss/gain events that have occurred over the course of orthopoxvirus evolution. It is clear that for all existing Orthopoxvirus species, no individual species has acquired protein-coding genes unique to that species. All existing species contain genes that are all present in members of the species Cowpox virus and that cowpox virus strains contain every gene present in any other orthopoxvirus strain. These results support a theory of reductive evolution in which the reduction in size of the core gene set of a putative ancestral virus played a critical role in speciation and confining any newly emerging virus species to a particular environmental (host or tissue) niche.
Collapse
Affiliation(s)
- Robert Curtis Hendrickson
- Department of Microbiology, University of Alabama at Birmingham, BBRB 276/11, 845 19th St S, Birmingham, AL 35222, USA; E-Mails: (R.C.H.); (E.L.H.)
| | - Chunlin Wang
- Stanford Genome Technology Center, Stanford University, 855 California Ave, Palo Alto, CA 94304, USA; E-Mail:
| | - Eneida L. Hatcher
- Department of Microbiology, University of Alabama at Birmingham, BBRB 276/11, 845 19th St S, Birmingham, AL 35222, USA; E-Mails: (R.C.H.); (E.L.H.)
| | - Elliot J. Lefkowitz
- Department of Microbiology, University of Alabama at Birmingham, BBRB 276/11, 845 19th St S, Birmingham, AL 35222, USA; E-Mails: (R.C.H.); (E.L.H.)
| |
Collapse
|
48
|
Shchelkunov SN. Interaction of orthopoxviruses with the cellular ubiquitin-ligase system. Virus Genes 2010; 41:309-18. [PMID: 20703935 DOI: 10.1007/s11262-010-0519-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2010] [Accepted: 07/28/2010] [Indexed: 02/06/2023]
Abstract
Protein modification by ubiquitin or ubiquitin-like polypeptides is important for the fate and functions of the majority of proteins in the eukaryotic cell and can be involved in regulation of various biological processes, including protein metabolism (degradation), protein transport to several cellular compartments, rearrangement of cytoskeleton, and transcription of cytoprotective genes. The accumulated experimental data suggest that the ankyrin-F-box-like and BTB-kelch-like proteins of orthopoxviruses, represented by the largest viral multigene families, interact with the cellular Cullin-1- and Cullin-3-containing ubiquitin-protein ligases, respectively. In addition, orthopoxviruses code for their own RING-domain-containing ubiquitin ligase. In this review, this author discusses the differences between variola (smallpox), monkeypox, cowpox, vaccinia, and ectromelia (mousepox) viruses in the organization of ankyrin-F-box and BTB-kelch protein families and their likely functions.
Collapse
Affiliation(s)
- Sergei N Shchelkunov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Lavrentiev Ave. 10, Novosibirsk, Russia.
| |
Collapse
|
49
|
Alkhalil A, Hammamieh R, Hardick J, Ichou MA, Jett M, Ibrahim S. Gene expression profiling of monkeypox virus-infected cells reveals novel interfaces for host-virus interactions. Virol J 2010; 7:173. [PMID: 20667104 PMCID: PMC2920256 DOI: 10.1186/1743-422x-7-173] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Accepted: 07/28/2010] [Indexed: 12/18/2022] Open
Abstract
Monkeypox virus (MPV) is a zoonotic Orthopoxvirus and a potential biothreat agent that causes human disease with varying morbidity and mortality. Members of the Orthopoxvirus genus have been shown to suppress antiviral cell defenses, exploit host cell machinery, and delay infection-induced cell death. However, a comprehensive study of all host genes and virus-targeted host networks during infection is lacking. To better understand viral strategies adopted in manipulating routine host biology on global scale, we investigated the effect of MPV infection on Macaca mulatta kidney epithelial cells (MK2) using GeneChip rhesus macaque genome microarrays. Functional analysis of genes differentially expressed at 3 and 7 hours post infection showed distinctive regulation of canonical pathways and networks. While the majority of modulated histone-encoding genes exhibited sharp copy number increases, many of its transcription regulators were substantially suppressed; suggesting involvement of unknown viral factors in host histone expression. In agreement with known viral dependence on actin in motility, egress, and infection of adjacent cells, our results showed extensive regulation of genes usually involved in controlling actin expression dynamics. Similarly, a substantial ratio of genes contributing to cell cycle checkpoints exhibited concerted regulation that favors cell cycle progression in G1, S, G2 phases, but arrest cells in G2 phase and inhibits entry into mitosis. Moreover, the data showed that large number of infection-regulated genes is involved in molecular mechanisms characteristic of cancer canonical pathways. Interestingly, ten ion channels and transporters showed progressive suppression during the course of infection. Although the outcome of this unusual channel expression on cell osmotic homeostasis remains unknown, instability of cell osmotic balance and membrane potential has been implicated in intracellular pathogens egress. Our results highlight the role of histones, actin, cell cycle regulators, and ion channels in MPV infection, and propose these host functions as attractive research focal points in identifying novel drug intervention sites.
Collapse
Affiliation(s)
- Abdulnaser Alkhalil
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland 21702, USA
| | - Rasha Hammamieh
- Division of Pathology, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Justin Hardick
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland 21702, USA
| | - Mohamed Ait Ichou
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland 21702, USA
| | - Marti Jett
- Division of Pathology, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Sofi Ibrahim
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland 21702, USA
| |
Collapse
|
50
|
Structure function studies of vaccinia virus host range protein k1 reveal a novel functional surface for ankyrin repeat proteins. J Virol 2010; 84:3331-8. [PMID: 20089642 DOI: 10.1128/jvi.02332-09] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Poxvirus host tropism at the cellular level is regulated by virus-encoded host range proteins acting downstream of virus entry. The functioning mechanisms of most host range proteins are unclear, but many contain multiple ankyrin (ANK) repeats, a motif that is known for ligand interaction through a concave surface. We report here the crystal structure of one of the ANK repeat-containing host range proteins, the vaccinia virus K1 protein. The structure, at a resolution of 2.3 A, showed that K1 consists entirely of ANK repeats, including seven complete ones and two incomplete ones, one each at the N and C terminus. Interestingly, Phe82 and Ser83, which were previously shown to be critical for K1's function, are solvent exposed and located on a convex surface, opposite the consensus ANK interaction surface. The importance of this convex surface was further supported by our additional mutagenesis studies. We found that K1's host range function was negatively affected by substitution of either Asn51 or Cys47 and completely abolished by substitution of both residues. Cys47 and Asn51 are also exposed on the convex surface, spatially adjacent to Phe82 and Ser83. Altogether, our data showed that K1 residues on a continuous convex ANK repeat surface are critical for the host range function, suggesting that K1 functions through ligand interaction and does so with a novel ANK interaction surface.
Collapse
|