1
|
Bamouh Z, Tifrouin I, Elkarhat Z, Abid L, Fellahi S, Elharrak M. Pathogenicity and phylogenetic analysis of ovine contagious ecthyma virus isolated during a sheeppox outbreak in Morocco. Microb Pathog 2024; 197:107023. [PMID: 39423917 DOI: 10.1016/j.micpath.2024.107023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Contagious ecthyma (CE), also known as ORF is a highly contagious zoonotic viral skin disease that affects humans, sheep, goats and other domesticated and wild animals. As reported here-in, the objective of this study was to investigate a suspected outbreak of both sheeppox and ORF diseases in a sheep herd during the winter of 2020 in Northwest Morocco. The affected sheep showed nodules and proliferative scabby skin lesions around the mouth and hairless area of the body. Samples of skin crust were collected for virus identification and isolation. A virus was isolated in Vero cells, lamb testis and heart cells and the cytopathic effect was characterized by cells aggregation, ballooning, and detachment. Initially, the suspensions of skin crust were positive for sheeppox virus (SPPV) by PCR. Subsequent testing of the isolated virus from skin crust of affected animals indicated that the virus was SPPV-negative and ORFV-positive by PCR. Furthermore, nucleotide sequences of the B2L aligned with reference ORFV isolates for genetic analysis. Phylogenetic analyses results confirmed that the isolated virus was ORFV and that the virus was closely related to ORFV strains isolated in Sudan and Malaysia. In conclusion, this study is the first reported detection of ORFV in Morocco, and therefore, poses as an imminent threat to the health of humans, domestic and wild animals.
Collapse
Affiliation(s)
- Zohra Bamouh
- Research and Development, MCI Santé Animale, Lot. 157, Z. I., Sud-Ouest (ERAC) B.P: 278, Mohammedia, 28810, Morocco; Hassan II Agronomic and Veterinary Institute, Rabat, Morocco.
| | - Ikram Tifrouin
- Research and Development, MCI Santé Animale, Lot. 157, Z. I., Sud-Ouest (ERAC) B.P: 278, Mohammedia, 28810, Morocco.
| | - Zouhair Elkarhat
- Research and Development, MCI Santé Animale, Lot. 157, Z. I., Sud-Ouest (ERAC) B.P: 278, Mohammedia, 28810, Morocco; Physiopathology, Molecular Biology and Biotechnology Laboratory, Faculty of Sciences Ain Chock, University Hassan II, B.P 5366 Maarif, Casablanca, 20000, Morocco.
| | - Laila Abid
- Research and Development, MCI Santé Animale, Lot. 157, Z. I., Sud-Ouest (ERAC) B.P: 278, Mohammedia, 28810, Morocco.
| | - Siham Fellahi
- Hassan II Agronomic and Veterinary Institute, Rabat, Morocco.
| | - Mehdi Elharrak
- Research and Development, MCI Santé Animale, Lot. 157, Z. I., Sud-Ouest (ERAC) B.P: 278, Mohammedia, 28810, Morocco.
| |
Collapse
|
2
|
Wang Y, Yang H, Hu J, Jiang Y, Ma W, Gao S, Chen D. Preparation and application of fluorescent monoclonal antibodies recognizing goat CD4 +CD25 + regulatory T cells. Appl Microbiol Biotechnol 2024; 108:327. [PMID: 38717623 PMCID: PMC11078799 DOI: 10.1007/s00253-024-13115-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/04/2024] [Accepted: 03/19/2024] [Indexed: 05/12/2024]
Abstract
Regulatory T cells (Tregs) are a subset of T cells participating in a variety of diseases including mycoplasmal pneumonia, contagious ecthyma, and so on. The role of Tregs in goat contagious ecthyma is not completely understood due to the lack of species-specific antibodies. Here, we developed a combination of CD4 and CD25 fluorescence monoclonal antibodies (mAb) to recognize goat Tregs and assessed its utility in flow cytometry, immunofluorescence staining. Using immunofluorescence staining, we found that the frequency of Treg cells was positively correlated with the viral load during orf virus infection. These antibodies could serve as important tools to monitor Tregs during orf virus infection in goats. KEY POINTS: • A combination of fluorescent mAbs (C11 and D12) was prepared for the detection of goat Tregs. • C11 and D12 are effective in flow cytometry, immunofluorescence staining, and C11 has excellent species specificity. • The frequency of Treg cells was positively correlated with the viral load during orf virus infection.
Collapse
Affiliation(s)
- Yunpeng Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Haoyue Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jiajin Hu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yuecai Jiang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Wentao Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| | - Shikong Gao
- Shenmu Animal Husbandry Development Center, Shenmu, 719300, Shaanxi, China.
| | - Dekun Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
3
|
Petro-Turnquist E, Pekarek MJ, Weaver EA. Swine influenza A virus: challenges and novel vaccine strategies. Front Cell Infect Microbiol 2024; 14:1336013. [PMID: 38633745 PMCID: PMC11021629 DOI: 10.3389/fcimb.2024.1336013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/21/2024] [Indexed: 04/19/2024] Open
Abstract
Swine Influenza A Virus (IAV-S) imposes a significant impact on the pork industry and has been deemed a significant threat to global public health due to its zoonotic potential. The most effective method of preventing IAV-S is vaccination. While there are tremendous efforts to control and prevent IAV-S in vulnerable swine populations, there are considerable challenges in developing a broadly protective vaccine against IAV-S. These challenges include the consistent diversification of IAV-S, increasing the strength and breadth of adaptive immune responses elicited by vaccination, interfering maternal antibody responses, and the induction of vaccine-associated enhanced respiratory disease after vaccination. Current vaccination strategies are often not updated frequently enough to address the continuously evolving nature of IAV-S, fail to induce broadly cross-reactive responses, are susceptible to interference, may enhance respiratory disease, and can be expensive to produce. Here, we review the challenges and current status of universal IAV-S vaccine research. We also detail the current standard of licensed vaccines and their limitations in the field. Finally, we review recently described novel vaccines and vaccine platforms that may improve upon current methods of IAV-S control.
Collapse
Affiliation(s)
- Erika Petro-Turnquist
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Matthew J. Pekarek
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Eric A. Weaver
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
4
|
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
|
5
|
Yang J, Zhao Y, Fu Y, Lv Y, Zhu Y, Zhu M, Zhao J, Wang Y, Wu C, Zhao W. Recombinant antigen P29 of Echinococcus granulosus induces Th1, Tc1, and Th17 cell immune responses in sheep. Front Immunol 2023; 14:1243204. [PMID: 38187382 PMCID: PMC10768560 DOI: 10.3389/fimmu.2023.1243204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 11/22/2023] [Indexed: 01/09/2024] Open
Abstract
Echinococcosis is a common human and animal parasitic disease that seriously endangers human health and animal husbandry. Although studies have been conducted on vaccines for echinococcosis, to date, there is no human vaccine available for use. One of the main reasons for this is the lack of in-depth research on basic immunization with vaccines. Our previous results confirmed that recombinant antigen P29 (rEg.P29) induced more than 90% immune protection in both mice and sheep, but data on its induction of sheep-associated cellular immune responses are lacking. In this study, we investigated the changes in CD4+ T cells, CD8+ T cells, and antigen-specific cytokines IFN-γ, IL-4, and IL-17A after rEg.P29 immunization using enzyme-linked immunospot assay (ELISPOT), enzyme-linked immunosorbent assay (ELISA), and flow cytometry to investigate the cellular immune response induced by rEg.P29 in sheep. It was found that rEg.P29 immunization did not affect the percentage of CD4+ and CD8+ T cells in peripheral blood mononuclear cells (PBMCs), and was able to stimulate the proliferation of CD4+ and CD8+ T cells after immunization in vitro. Importantly, the results of both ELISPOT and ELISA showed that rEg.P29 can induce the production of the specific cytokines IFN-γ and IL-17A, and flow cytometry verified that rEg.P29 can induce the expression of IFN-γ in CD4+ and CD8+ T cells and IL-17A in CD4+ T cells; however, no IL-4 expression was observed. These results indicate that rEg.P29 can induce Th1, Th17, and Tc1 cellular immune responses in sheep against echinococcosis infection, providing theoretical support for the translation of rEg.P29 vaccine applications.
Collapse
Affiliation(s)
- Jihui Yang
- Center of Scientific Technology of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases of Ningxia Medical University, Yinchuan, China
| | - Yinqi Zhao
- Center of Scientific Technology of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases of Ningxia Medical University, Yinchuan, China
| | - Yong Fu
- Qinghai Academy of Animal Sciences and Veterinary Medicine, Qinghai University, Xining, China
| | - Yongxue Lv
- Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases of Ningxia Medical University, Yinchuan, China
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
| | - Yazhou Zhu
- Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases of Ningxia Medical University, Yinchuan, China
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
| | - Mingxing Zhu
- Center of Scientific Technology of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases of Ningxia Medical University, Yinchuan, China
| | - Jiaqing Zhao
- Center of Scientific Technology of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases of Ningxia Medical University, Yinchuan, China
| | - Yana Wang
- Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases of Ningxia Medical University, Yinchuan, China
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
| | - Changyou Wu
- Institute of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wei Zhao
- Center of Scientific Technology of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
6
|
Portillo AL, Monteiro JK, Rojas EA, Ritchie TM, Gillgrass A, Ashkar AA. Charting a killer course to the solid tumor: strategies to recruit and activate NK cells in the tumor microenvironment. Front Immunol 2023; 14:1286750. [PMID: 38022679 PMCID: PMC10663242 DOI: 10.3389/fimmu.2023.1286750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
The ability to expand and activate natural Killer (NK) cells ex vivo has dramatically changed the landscape in the development of novel adoptive cell therapies for treating cancer over the last decade. NK cells have become a key player for cancer immunotherapy due to their innate ability to kill malignant cells while not harming healthy cells, allowing their potential use as an "off-the-shelf" product. Furthermore, recent advancements in NK cell genetic engineering methods have enabled the efficient generation of chimeric antigen receptor (CAR)-expressing NK cells that can exert both CAR-dependent and antigen-independent killing. Clinically, CAR-NK cells have shown promising efficacy and safety for treating CD19-expressing hematologic malignancies. While the number of pre-clinical studies using CAR-NK cells continues to expand, it is evident that solid tumors pose a unique challenge to NK cell-based adoptive cell therapies. Major barriers for efficacy include low NK cell trafficking and infiltration into solid tumor sites, low persistence, and immunosuppression by the harsh solid tumor microenvironment (TME). In this review we discuss the barriers posed by the solid tumor that prevent immune cell trafficking and NK cell effector functions. We then discuss promising strategies to enhance NK cell infiltration into solid tumor sites and activation within the TME. This includes NK cell-intrinsic and -extrinsic mechanisms such as NK cell engineering to resist TME-mediated inhibition and use of tumor-targeted agents such as oncolytic viruses expressing chemoattracting and activating payloads. We then discuss opportunities and challenges for using combination therapies to extend NK cell therapies for the treatment of solid tumors.
Collapse
Affiliation(s)
- Ana L. Portillo
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada
| | - Jonathan K. Monteiro
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada
| | - Eduardo A. Rojas
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Tyrah M. Ritchie
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Amy Gillgrass
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada
| | - Ali A. Ashkar
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
7
|
AlDaif BA, Mercer AA, Fleming SB. The parapoxvirus Orf virus inhibits dsDNA-mediated type I IFN expression via STING-dependent and STING-independent signalling pathways. J Gen Virol 2023; 104. [PMID: 37882657 DOI: 10.1099/jgv.0.001912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023] Open
Abstract
Type I interferons (IFNs) are critical in the host defence against viruses. They induce hundreds of interferon-stimulated genes (ISGs) many of which have an antiviral role. Poxviruses induce IFNs via their pathogen-associated molecular patterns, in particular, their genomic DNA. In a majority of cell types, dsDNA is detected by a range of cytoplasmic DNA sensors that mediate type I IFN expression via stimulator of interferon genes (STING). Orf virus (ORFV) induces cutaneous pustular skin lesions and is the type species of the Parapoxvirus genus within the Poxviridae family. The aim of this study was to investigate whether ORFV modulates dsDNA-induced type I IFN expression via STING-dependent signalling pathways in human dermal fibroblasts (hNDF) and THP-1 cells. We showed that ORFV infection of these cell types treated with poly(dA:dT) resulted in strong inhibition of expression of IFN-β. In hNDFs, we showed using siRNA knock-down that STING was essential for type I IFN induction. IFN-β expression was further reduced when both STING and RIG-I were knocked down. In addition, HEK293 cells that do not express STING or Toll-like receptors also produce IFN-β following stimulation with poly(dA:dT). The 5' triphosphate dsRNA produced by RNA polymerase III specifically results in the induction of type I IFNs through the RIG-I receptor. We showed that ORFV infection resulted in strong inhibition of IFN-β expression in HEK293 cells stimulated with poly(dA:dT). Overall, this study shows that ORFV potently counteracts the STING-dependent and STING-independent IFN response by antagonizing dsDNA-activated IFN signalling pathways.
Collapse
Affiliation(s)
- Basheer A AlDaif
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Andrew A Mercer
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Stephen B Fleming
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
8
|
Joshi LR, do Nascimento GM, Diel DG. The transcriptome of the parapoxvirus Orf virus reveals novel promoters for heterologous gene expression by poxvirus vectors. Virology 2023; 587:109864. [PMID: 37595395 DOI: 10.1016/j.virol.2023.109864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 07/24/2023] [Accepted: 08/03/2023] [Indexed: 08/20/2023]
Abstract
Orf virus (ORFV) has been used as a vaccine delivery vector for multiple animal species. Several strategies are being used to improve the immunogenicity and efficacy of ORFV vectors, including the use of poxviral promoter(s) with strong early and late activity capable of driving the expression of the heterologous genes for a prolonged time and eliciting a potent immune response. Here, we used RNA-sequencing (RNA-Seq) approach to analyze the transcriptome of ORFV during infection in primary ovine cells. Based on the transcriptional profile of individual ORFV genes, we identified ORFV promoters with strong early and late activity and have shown that they can be used to express heterologous genes in ORFV vectors. Our results show that the intergenic regulatory sequence containing core promoter sequences present upstream of ORF112 (p112) and ORF116 (p116) lead to markedly higher transgene expression than conventional poxviral promoters. Thus, these promoters are valuable alternatives to express transgenes in poxviral vectors.
Collapse
Affiliation(s)
- Lok R Joshi
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, USA
| | - Gabriela Mansano do Nascimento
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, USA
| | - Diego G Diel
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, USA.
| |
Collapse
|
9
|
Li S, Jing T, Zhu F, Chen Y, Yao X, Tang X, Zuo C, Liu M, Xie Y, Jiang Y, Wang Y, Li D, Li L, Gao S, Chen D, Zhao H, Ma W. Genetic Analysis of Orf Virus (ORFV) Strains Isolated from Goats in China: Insights into Epidemiological Characteristics and Evolutionary Patterns. Virus Res 2023; 334:199160. [PMID: 37402415 PMCID: PMC10410590 DOI: 10.1016/j.virusres.2023.199160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/14/2023] [Accepted: 06/22/2023] [Indexed: 07/06/2023]
Abstract
Contagious ecthyma (CE) is an acute infectious zoonosis caused by orf virus (ORFV) that mainly infects sheep and goats and causes obvious lesions and low market value of livestock, resulting in huge economic losses for farmers. In this study, two strains of ORFV were isolated from Shaanxi Province and Yunnan Province in China, named FX and LX. The two ORFVs were located in the major clades of domestic strains respectively, and exhibited distinct sequence homology. We analyzed the genetic data of core genes (B2L, F1L, VIR, ORF109) and variable genes (GIF, ORF125 and vIL-10) of ORFV to investigate its epidemiological and evolutionary characteristics. The sequences from 2007 to 2018 constituted the majority of the viral population, predominantly concentrated in India and China. Most genes were clustered into SA00-like type and IA82-like type, and the hotspots in East and South Asia were identified in the ORFV transmission trajectories. For these genes, VIR had the highest substitution rate of 4.85 × 10-4, both VIR and vIL-10 suffered the positive selection pressure during ORFV evolution. Many motifs associated with viral survival were distributed among ORFVs. In addition, some possible viral epitopes have been predicted, which still require validation in vivo and in vitro. This work gives more insight into the prevalence and phylogenetic relationships of existing orf viruses and facilitate better vaccine design.
Collapse
Affiliation(s)
- Shaofei Li
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province 712100, China
| | - Tian Jing
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province 712100, China
| | - Fang Zhu
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province 712100, China
| | - Yiming Chen
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province 712100, China
| | - Xiaoting Yao
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province 712100, China
| | - Xidian Tang
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province 712100, China
| | - Chenxiang Zuo
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province 712100, China
| | - Mingjie Liu
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province 712100, China
| | - Yanfei Xie
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province 712100, China
| | - Yuecai Jiang
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province 712100, China
| | - Yunpeng Wang
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province 712100, China
| | - Dengliang Li
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province 712100, China
| | - Lulu Li
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province 712100, China
| | - Shikong Gao
- Shenmu Animal Husbandry Development Center, Shenmu, Shaanxi Province 719399, China
| | - Dekun Chen
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province 712100, China.
| | - Huiying Zhao
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province 712100, China.
| | - Wentao Ma
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi Province 712100, China.
| |
Collapse
|
10
|
Stevenson H, Corbett C, Henderson K, Mason C, Richards-Rios P. Lymph node aplasia in Scottish blackface sheep. Vet Rec 2023; 192:489-490. [PMID: 37326185 DOI: 10.1002/vetr.3200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Affiliation(s)
- Heather Stevenson
- SRUC Veterinary Services, St Mary's Industrial Estate, Dumfries, DG1 1DX
| | - Cici Corbett
- SRUC Veterinary Services, St Mary's Industrial Estate, Dumfries, DG1 1DX
| | - Katrina Henderson
- SRUC Veterinary Services, St Mary's Industrial Estate, Dumfries, DG1 1DX
| | - Colin Mason
- SRUC Veterinary Services, St Mary's Industrial Estate, Dumfries, DG1 1DX
| | - Peter Richards-Rios
- SRUC Veterinary Services, Pentlands Science Park, Bush Loan, Penicuik, Midlothian, EH26 0PZ
| |
Collapse
|
11
|
Ghazaleh RA, Al-Sawalhe M, Odeh IA, El Ibrahim J, Al-Turman B, Makhamreh J. Host range, severity and trans boundary transmission of Orf virus (ORFV). INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023; 112:105448. [PMID: 37217030 DOI: 10.1016/j.meegid.2023.105448] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 04/03/2023] [Accepted: 04/30/2023] [Indexed: 05/24/2023]
Abstract
Contagious ecthyma in small ruminants is a zoonotic disease caused by Orf virus (ORFV) in the genus Parapoxvirus that can be deadly to its natural hosts. It causes significant losses worldwide, and commonly infects humans. However, the literature about its comparative severity in sheep and goat hosts is misleading; and while contagious ecthyma has been shown to occur in camels and transmit to humans, there is confusion as to whether ORFV is responsible. Camels are important from a 'One Health' perspective as they have been implicated as a reservoir host for the virus causing Middle East Respiratory Syndrome (MERS), which has a case fatality rate of 35% in humans. We compared ORFV gene sequences and mortality data from the West Bank in Palestine, where ORFV has not been reported previously, with data from the region. Surprisingly, we found that infections of camels that had been attributed to ORFV were more closely related to a different member of the genus Parapoxvirus. Two Middle East ORFVs isolated from humans were unrelated and sat alongside sheep and goat derived sequences on two distinct ORFV lineages of a maximum likelihood B2L gene tree. One of the viral lineages bifurcated to produce a monophyletic group of goat-derived ORFVs characterized uniquely by a glycine at amino acid reside 249. We found that serine is the ancestral allele shared between ORFV infections of sheep and also two closely related Parapoxviruses (PCPV and CCEV), indicating that the glycine allele represents a more recent shift in virus host range adaptation to goats. Furthermore, and contrary to some reports that ORFV is more severe in goats than in sheep, we observed median mortality of up to 24.5% in sheep, but none in goats. We also identified trans-boundary spread of ORFV between the West Bank and Israel.
Collapse
Affiliation(s)
- Robin Abu Ghazaleh
- Palestine-Korea Biotechnology Research Center, Palestine Polytechnic University, P.O. Box 198, Hebron, Palestine.
| | - Mohammed Al-Sawalhe
- Palestine-Korea Biotechnology Research Center, Palestine Polytechnic University, P.O. Box 198, Hebron, Palestine
| | - Insaf Abu Odeh
- Palestine-Korea Biotechnology Research Center, Palestine Polytechnic University, P.O. Box 198, Hebron, Palestine
| | - Jehad El Ibrahim
- Palestine-Korea Biotechnology Research Center, Palestine Polytechnic University, P.O. Box 198, Hebron, Palestine; National Agricultural Research Center (NARC), Jenin, Palestine
| | - Basem Al-Turman
- Palestine-Korea Biotechnology Research Center, Palestine Polytechnic University, P.O. Box 198, Hebron, Palestine
| | | |
Collapse
|
12
|
Lv L, Guan J, Zhen R, Lv P, Xu M, Liu X, He S, Fang Z, Li Z, Lan Y, Lu H, He W, Gao F, Zhao K. Orf virus induces complete autophagy to promote viral replication via inhibition of AKT/mTOR and activation of the ERK1/2/mTOR signalling pathway in OFTu cells. Vet Res 2023; 54:22. [PMID: 36918891 PMCID: PMC10013242 DOI: 10.1186/s13567-023-01153-1] [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: 08/22/2022] [Accepted: 12/09/2022] [Indexed: 03/15/2023] Open
Abstract
Orf virus (ORFV) is the causative agent of contagious ecthyma, which is an important zoonotic pathogen with a widespread distribution affecting sheep, goats and humans. Our previous research showed that autophagy can be induced in host cells by ORFV infection. However, the exact mechanism of ORFV-induced autophagy remains unknown. In this study, we investigated the underlying mechanisms of autophagy induced by ORFV in OFTu cells and the impact of autophagy on ORFV replication. By using specific autophagy inhibitors and activators, Western blotting, immunofluorescence and transmission electron microscopy imaging, we confirmed that ORFV infection triggered intracellular autophagosome accumulation and the activation of autophagic flux. Moreover, ORFV-induced autophagic activity was found to rely on an increase in the phosphorylation of tuberous sclerosis complex 2 (TSC2) and a decrease in the phosphorylation of mammalian target of rapamycin (mTOR), which is mediated by the suppression of the PI3K/AKT/mTOR signalling pathway and activation of the ERK1/2/mTOR signalling pathway. Furthermore, we investigated the role of mTOR-mediated autophagy during ORFV replication using pharmacological agents and demonstrated that ORFV-induced autophagy correlated positively with viral replication. Taken together, our data reveal the pathways of ORFV-induced autophagy and the impact of autophagy on ORFV replication, providing new insights into ORFV pathogenesis.
Collapse
Affiliation(s)
- Lijun Lv
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jiyu Guan
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ruixue Zhen
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Pin Lv
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Mengshi Xu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xingyuan Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shishi He
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ziyu Fang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zi Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yungang Lan
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Huijun Lu
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, China
| | - Wenqi He
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Feng Gao
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, China
| | - Kui Zhao
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.
| |
Collapse
|
13
|
Yu Y, Zhang F, Duan X, Yang C, Cui Y, Yu L. ORFV can carry TRAP gene expression via intracellular CRISPR/Cas9 gene editing technology. J Virol Methods 2023; 312:114652. [PMID: 36493528 DOI: 10.1016/j.jviromet.2022.114652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022]
Abstract
Orf is an acute and highly contracted human and animal infection caused by orf virus (ORFV), which mainly affects sheep, goats, and other species. Clinically, opportunistic or conditional pathogens such as Staphylococcus aureus (S. aureus) are often detected in cases of orf, which greatly increases the risk of disease progression and clinical death. It has been reported that TRAP gene products of S. aureus can broadly influence bacterial life and pathogenicity in vivo, and introduction of exogenous TRAP genes may help to inhibit the proliferation of bacteria. In order to achieve the combined control of ORFV and S. aureus, a novel approach to design a S. aureus TRAP gene vaccine using a live attenuated ORFV vector is proposed. In this study, CRISPR/Cas9 gene editing technology was used to disable vascular endothelial growth factor E of ORFV (VEGF-v) and introduced TRAP gene into this position. TRAP gene expression was detected in keratinocytes infected with recombinant virus. The construction and experimental verification of recombinant ORFV (ORFV-v/TRAP) will provide a reference for in-depth studies on the prevention and control of mixed infectious disease.
Collapse
Affiliation(s)
- YongZhong Yu
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China.
| | - Fan Zhang
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Xuyang Duan
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China.
| | - ChaoQun Yang
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - YuDong Cui
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| | - Li Yu
- Division of Livestock Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 427 Maduan Street, Harbin 150001, PR China
| |
Collapse
|
14
|
Deletion of gene OV132 attenuates Orf virus more effectively than gene OV112. Appl Microbiol Biotechnol 2023; 107:835-851. [PMID: 36484827 PMCID: PMC9734686 DOI: 10.1007/s00253-022-12323-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/27/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022]
Abstract
Orf virus (ORFV), a Parapoxvirus in Poxviridae, infects sheep and goats resulting in contagious pustular dermatitis. ORFV is regarded as a promising viral vector candidate for vaccine development and oncolytic virotherapy. Owing to their potential clinical application, safety concerns have become increasingly important. Deletion of either the OV132 (encoding vascular endothelial growth factor, VEGF) or OV112 (encoding the chemokine binding protein, CBP) genes reduced ORFV infectivity, which has been independently demonstrated in the NZ2 and NZ7 strains, respectively. This study revealed that the VEGF and CBP gene sequences of the local strain (TW/Hoping) shared a similarity of 47.01% with NZ2 and 90.56% with NZ7. Due to the high sequence divergence of these two immunoregulatory genes among orf viral strains, their contribution to the pathogenicity of Taiwanese ORFV isolates was comparatively characterized. Initially, two ORFV recombinants were generated, in which either the VEGF or CBP gene was deleted and replaced with the reporter gene EGFP. In vitro assays indicated that both the VEGF-deletion mutant ORFV-VEGFΔ-EGFP and the CBP deletion mutant ORFV-CBPΔ-EGFP were attenuated in cells. In particular, ORFV-VEGFΔ-EGFP significantly reduced plaque size and virus yield compared to ORFV-CBPΔ-EGFP and the wild-type control. Similarly, in vivo analysis revealed no virus yield in the goat skin biopsy infected by ORFV-VEGFΔ-EGFP, and significantly reduced the virus yield of ORFV-CBPΔ-EGFP relative to the wild-type control. These results confirmed the loss of virulence of both deletion mutants in the Hoping strain, whereas the VEGF-deletion mutant was more attenuated than the CBP deletion strain in both cell and goat models. KEY POINTS: • VEGF and CBP genes are crucial in ORFV pathogenesis in the TW/Hoping strain • The VEGF-deletion mutant virus was severely attenuated in both cell culture and animal models • Deletion mutant viruses are advantageous vectors for the development of vaccines and therapeutic regimens.
Collapse
|
15
|
Zhou Y, Guan J, Lv L, Cui H, Xu M, Wang S, Yu Z, Zhen R, He S, Fang Z, Zhong J, Cui S, Yu S, Song D, He W, Gao F, Zhao K. Complete genomic sequences and comparative analysis of two Orf virus isolates from Guizhou Province and Jilin Province, China. Virus Genes 2022; 58:403-413. [PMID: 35780442 DOI: 10.1007/s11262-022-01918-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 05/26/2022] [Indexed: 10/17/2022]
Abstract
Orf virus (ORFV, species Orf virus) belongs to the typical species of the Parapoxvirus genus of the family Poxviridae, which infects sheep, goats, and humans with worldwide distribution. Although outbreaks of Orf have been reported sequentially in several Chinese provinces, the epidemiology of Orf and genetic diversity of ORFV strains still needs to be further characterized. To further reveal the genomic organization of the ORFV-GZ18 and ORFV-CL18 isolates, the complete genome sequences of two recently obtained ORFV isolates were sequenced using the next-generation sequencing technology and analyzed, which had been deposited in the GenBank database under accession number MN648218 and MN648219, respectively. The complete genomic sequence of ORFV-CL18 was 138,495 bp in length, including 131 potential open reading frames (ORFs) flanked by inverted terminal repeats (ITRs) of 3481 bp at both ends, which has genomic structure typical Parapoxviruses. The overall genomic organization of the fully sequenced genome of ORFV-GZ18 was consistent with ORFV-CL18 genome, with a complete genome size of 138,446 nucleotides, containing 131 ORFs flanked by ITRs of 3469 bp. Additionally, the overall G + C contents of ORFV-GZ18 and ORFV-CL18 genome sequences were about 63.9% and 63.8%, respectively. The phylogenetic analysis showed that both ORFV-GZ18 and ORFV-CL18 were genetically closely related to ORFV-SY17 derived from sheep. In summary, the complete genomic sequences of ORFV-GZ18 and ORFV-CL18 are reported, with the hope it will be useful to investigate the host range, geographic distribution, and genetic evolution of the virus in Southern West and Northern East China.
Collapse
Affiliation(s)
- Yanlong Zhou
- Key Laboratory of Zoonosis Research, College of Veterinary Medicine, Jilin University, Ministry of Education, Changchun, China
| | - Jiyu Guan
- Key Laboratory of Zoonosis Research, College of Veterinary Medicine, Jilin University, Ministry of Education, Changchun, China
| | - Lijun Lv
- Key Laboratory of Zoonosis Research, College of Veterinary Medicine, Jilin University, Ministry of Education, Changchun, China
| | - Huan Cui
- Key Laboratory of Zoonosis Research, College of Veterinary Medicine, Jilin University, Ministry of Education, Changchun, China
| | - Mengshi Xu
- Key Laboratory of Zoonosis Research, College of Veterinary Medicine, Jilin University, Ministry of Education, Changchun, China
| | - Shuai Wang
- Key Laboratory of Zoonosis Research, College of Veterinary Medicine, Jilin University, Ministry of Education, Changchun, China
| | - Zhaohui Yu
- Key Laboratory of Zoonosis Research, College of Veterinary Medicine, Jilin University, Ministry of Education, Changchun, China
| | - Ruixue Zhen
- Key Laboratory of Zoonosis Research, College of Veterinary Medicine, Jilin University, Ministry of Education, Changchun, China
| | - Shishi He
- Key Laboratory of Zoonosis Research, College of Veterinary Medicine, Jilin University, Ministry of Education, Changchun, China
| | - Ziyu Fang
- Key Laboratory of Zoonosis Research, College of Veterinary Medicine, Jilin University, Ministry of Education, Changchun, China
| | - Jiawei Zhong
- Key Laboratory of Zoonosis Research, College of Veterinary Medicine, Jilin University, Ministry of Education, Changchun, China
| | - Shanshan Cui
- Key Laboratory of Zoonosis Research, College of Veterinary Medicine, Jilin University, Ministry of Education, Changchun, China
| | - Shiying Yu
- Liupanshui Agriculture Bureau, Liupanshui, Guizhou, China
| | - Deguang Song
- Key Laboratory of Zoonosis Research, College of Veterinary Medicine, Jilin University, Ministry of Education, Changchun, China
| | - Wenqi He
- Key Laboratory of Zoonosis Research, College of Veterinary Medicine, Jilin University, Ministry of Education, Changchun, China
| | - Feng Gao
- Key Laboratory of Zoonosis Research, College of Veterinary Medicine, Jilin University, Ministry of Education, Changchun, China.,Key Laboratory of Zoonosis, Institute of Zoonosis, Jilin University, Ministry of Education, Changchun, China
| | - Kui Zhao
- Key Laboratory of Zoonosis Research, College of Veterinary Medicine, Jilin University, Ministry of Education, Changchun, China.
| |
Collapse
|
16
|
Yang J, Lv Y, Zhu Y, Li S, Tao J, Chang L, Zhu M, Zhao J, Wang Y, Wu C, Zhao W. Baseline T-lymphocyte and cytokine indices in sheep peripheral blood. BMC Vet Res 2022; 18:165. [PMID: 35513847 PMCID: PMC9074339 DOI: 10.1186/s12917-022-03268-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/25/2022] [Indexed: 11/18/2022] Open
Abstract
Background Sheep are an important livestock species worldwide and an essential large-animal model for animal husbandry and veterinary research. Understanding fundamental immune indicators, especially T-lymphocyte parameters, is necessary for research on sheep diseases and vaccines, to better understand the immune response to bacteria and viruses for reducing the use of antibiotics and improving the welfare of sheep. We randomly selected 36 sheep of similar ages to analyze cell-related immune indicators in peripheral blood mononuclear cells (PBMCs). The proportions of CD4+ and CD8+ T cells in PBMCs were detected by flow cytometry. We used Concanavalin A (Con A) and Phorbol-12-myristate-13-acetate (PMA)/Ionomycin to stimulate PBMCs, and measured the expression of IFN-γ, IL-4, and IL-17A using enzyme-linked immunosorbent assay (ELISA) and enzyme-linked immunospot assay (ELISpot). Simultaneously, PMA/Ionomycin/brefeldin A (BFA) was added to PBMCs, then the expression of IFN-γ, IL-4, and IL-17A was detected by flow cytometry after 4 h of culturing. In addition, we observed the proliferation of PBMCs stimulated with Con A for 3, 4, and 5 days. Results The proportions of CD4+ T lymphocytes (18.70 ± 4.21%) and CD8+ T lymphocytes (8.70 ± 3.65%) were generally consistent among individuals, with a CD4/CD8 ratio of 2.40 ± 0.79. PBMCs produced high levels of IFN-γ, IL-4, and IL-17A after stimulation with PMA/Ionomycin and Con A. Furthermore, PMA/Ionomycin stimulation of PBMC yielded significantly higher cytokine levels than Con A stimulation. Flow cytometry showed that the level of IFN-γ (51.49 ± 11.54%) in CD8+ T lymphocytes was significantly (p < 0.001) higher than that in CD4+ T lymphocytes (14.29 ± 3.26%); IL-4 (16.13 ± 6.81%) in CD4+ T lymphocytes was significantly (p < 0.001) higher than that in CD8+ T lymphocytes (1.84 ± 1.33%), There was no difference in IL-17A between CD4+ (2.83 ± 0.98%) and CD8+ T lymphocytes (1.34 ± 0.67%). The proliferation of total lymphocytes, CD4+ T lymphocytes, and CD8+ T lymphocytes continued to increase between days 3 and 5; however, there were no significant differences in proliferation between the cell types during the stimulation period. Conclusions Evaluating primary sheep immune indicators, especially T lymphocytes, is significant for studying cellular immunity. This study provided valuable data and theoretical support for assessing the immune response of sheep to pathogens and improving sheep welfare.
Collapse
Affiliation(s)
- Jihui Yang
- Center of Scientifc Technology of Ningxia Medical University, Yinchuan, China.,Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases, Yinchuan, China.,School of Basic Medical Science of Ningxia Medical University, Yinchuan, China
| | - Yongxue Lv
- Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases, Yinchuan, China.,School of Basic Medical Science of Ningxia Medical University, Yinchuan, China
| | - Yazhou Zhu
- Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases, Yinchuan, China.,School of Basic Medical Science of Ningxia Medical University, Yinchuan, China
| | - Shasha Li
- Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases, Yinchuan, China.,School of Basic Medical Science of Ningxia Medical University, Yinchuan, China
| | - Jia Tao
- Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases, Yinchuan, China.,School of Basic Medical Science of Ningxia Medical University, Yinchuan, China
| | - Liangliang Chang
- Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases, Yinchuan, China.,School of Basic Medical Science of Ningxia Medical University, Yinchuan, China
| | - Mingxing Zhu
- Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases, Yinchuan, China.,School of Basic Medical Science of Ningxia Medical University, Yinchuan, China
| | - Jiaqing Zhao
- Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases, Yinchuan, China.,School of Basic Medical Science of Ningxia Medical University, Yinchuan, China
| | - Yana Wang
- Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases, Yinchuan, China.,School of Basic Medical Science of Ningxia Medical University, Yinchuan, China
| | - Changyou Wu
- Institute of Immunology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Wei Zhao
- Ningxia Key Laboratory of Prevention and Treatment of Common Infectious Diseases, Yinchuan, China. .,School of Basic Medical Science of Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
17
|
Parapoxvirus Interleukin-10 Homologues Vary in Their Receptor Binding, Anti-Inflammatory, and Stimulatory Activities. Pathogens 2022; 11:pathogens11050507. [PMID: 35631028 PMCID: PMC9143231 DOI: 10.3390/pathogens11050507] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 01/11/2023] Open
Abstract
Homologues of interleukin (IL)-10, a pleiotropic immunomodulatory cytokine, have been identified in the Parapoxvirus genus. The first identified, Orf virus (ORFV) IL-10, greatly enhanced infection of its host, exhibiting immune modulatory effects equivalent to human IL-10. IL-10-like genes were then identified in Bovine papular stomatitis virus (BPSV), Pseudocowpox virus (PCPV), Red deerpox virus (RDPV) and Grey sealpox virus (GSPV). This study aimed to produce and characterise recombinant parapoxvirus IL-10s, then quantitatively compare their receptor binding and immunomodulatory activities. Recombinant IL-10s were expressed, purified, then characterised using bioinformatic, biochemical and enzymatic analyses. Anti-inflammatory effects were assessed in lipoteichoic acid-activated THP-1 monocytes, and stimulatory effects in MC/9 mast cells. IL-10 receptor (IL-10R)1 binding was detected in a competitive displacement assay. BPSV IL-10 inhibited production of monocyte chemoattractant protein (MCP)-1, IL-8 and IL-1β, induced mast cell proliferation, and bound IL-10R1 similarly to ORFV IL-10. PCPV IL-10 showed reduced MCP-1 inhibition, mast cell proliferation, and IL-10R1 binding. RDPV IL-10 displayed reduced inhibition of IL-8 and MCP-1 production. GSPV IL-10 showed limited inhibition of IL-1β production and stimulation of mast cell proliferation. These findings provide valuable insight into IL-10 receptor interactions, and suggest that the parapoxvirus IL-10s play similar pathogenic roles during infection of their hosts.
Collapse
|
18
|
Yu Y, Lian Z, Cui Y. The OH system: A panorama view of the PPV-host interaction. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2022; 98:105220. [PMID: 35066165 DOI: 10.1016/j.meegid.2022.105220] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 11/19/2021] [Accepted: 01/17/2022] [Indexed: 06/14/2023]
Abstract
Poxviruses are a family of specialized cytoplasm-parasitic DNA viruses that replicate and assembly in virus factory. In Parapoxvirus (PPV) genus, with the orf virus (ORFV) as a representative species of this genus, their behaviors are significantly different from that of Orthopoxvirus, and the plots of viral practical solutions for evading host immunity are intricate and fascinating, particularly to anti-host and host's antiviral mechanisms. In order to protect the virus factory from immune elimination caused by infection, PPVs attempt to interfere with multiple stress levels of host, mainly by modulating innate immunity response (IIR) and adaptive immunity response (AIR). Given that temporarily constructed by virus infection, ORFV-HOST (OH) system accompanied by viral strategies is carefully managed in the virus factory, thus directing many life-critical events once undergoing the IIR and AIR. Evolutionarily, to reduce the risk of system destruction, ORFV have evolved into a mild-looking mode to avoid overstimulation. Moreover, the current version of development also focus on recognizing and hijacking more than eight antiviral security mechanisms of host cells, such as the 2',5'-oligoadenylate synthetase (OAS)/RNase L and PKR systems, the ubiquitin protease system (UPS), and so on. In summary, this review assessed inescapable pathways as mentioned above, through which viruses compete with their hosts strategically. The OH system provides a panoramic view and a powerful platform for us to study the PPV-Host interaction, as well as the corresponding implications on a great application potential in anti-virus design.
Collapse
Affiliation(s)
- Yongzhong Yu
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China.
| | - Zhengxing Lian
- College of Animal Science and Technology, China Agricultural University, Beijing 100039, PR China
| | - Yudong Cui
- College of Biological Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, PR China
| |
Collapse
|
19
|
AlDaif BA, Mercer AA, Fleming SB. The parapoxvirus Orf virus inhibits IFN-β expression induced by dsRNA. Virus Res 2022; 307:198619. [PMID: 34742812 DOI: 10.1016/j.virusres.2021.198619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/16/2022]
Abstract
Orf virus (ORFV) is the type species of the Parapoxvirus genus that belongs to the Poxviridae family. Type I interferons (IFN) are critical in the host defence against viruses. They induce hundreds of interferon stimulated genes (ISGs) many of which have an antiviral role. The ability of ORFV to modulate type I IFN production was undertaken to investigate whether ORFV could inhibit IFN-β expression via dsRNA dependant signalling pathways. HEK293 cells are known to lack DNA pattern-recognition receptors and Toll-like receptors however, they do express the cytosolic dsRNA receptors RIG-I and MDA5. HEK293 cells were shown to produce high levels of IFN-β when cells were stimulated with poly(I:C) and this was shown to be predominantly via RIG-I-dependant signalling as confirmed by siRNA knock-down of RIG-I. Further we showed that HEK293 cells are permissive for ORFV and caused potent inhibition of IFN-β transcription when cells were stimulated with poly(I:C) post-viral infection. Studies using heat inactivated ORFV suggested that de novo synthesis of early genes was required. In addition our findings showed that the ORFV encoded factor ORF020, that is known to bind dsRNA, is involved in antagonising IFN expression. Overall, this study has shown for first time the ability of ORFV to counteract type I IFN expression by antagonising dsRNA-activated RIG-I signalling.
Collapse
Affiliation(s)
- Basheer A AlDaif
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Andrew A Mercer
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Stephen B Fleming
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
20
|
AlDaif BA, Mercer AA, Fleming SB. The parapoxvirus Orf virus ORF116 gene encodes an antagonist of the interferon response. J Gen Virol 2021; 102. [PMID: 34890310 DOI: 10.1099/jgv.0.001695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Orf virus (ORFV) is the type species of the Parapoxvirus genus of the Poxviridae family. Genetic and functional studies have revealed ORFV has multiple immunomodulatory genes that manipulate innate immune responses, during the early stage of infection. ORF116 is a novel gene of ORFV with hitherto unknown function. Characterization of an ORF116 deletion mutant showed that it replicated in primary lamb testis cells with reduced levels compared to the wild-type and produced a smaller plaque phenotype. ORF116 was shown to be expressed prior to DNA replication. The potential function of ORF116 was investigated by gene-expression microarray analysis in HeLa cells infected with wild-type ORFV or the ORF116 deletion mutant. The analysis of differential cellular gene expression revealed a number of interferon-stimulated genes (ISGs) differentially expressed at either 4 or 6 h post infection. IFI44 showed the greatest differential expression (4.17-fold) between wild-type and knockout virus. Other ISGs that were upregulated in the knockout included RIG-I, IFIT2, MDA5, OAS1, OASL, DDX60, ISG20 and IFIT1 and in addition the inflammatory cytokine IL-8. These findings were validated by infecting HeLa cells with an ORF116 revertant recombinant virus and analysis of transcript expression by quantitative real time-PCR (qRT-PCR). These observations suggested a role for the ORFV gene ORF116 in modulating the IFN response and inflammatory cytokines. This study represents the first functional analysis of ORF116.
Collapse
Affiliation(s)
- Basheer A AlDaif
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Andrew A Mercer
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Stephen B Fleming
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
21
|
Koutsoumanis K, Allende A, Bolton D, Bover‐Cid S, Chemaly M, Davies R, De Cesare A, Herman L, Hilbert F, Lindqvist R, Nauta M, Peixe L, Ru G, Simmons M, Skandamis P, Suffredini E, Bottari B, Cummins E, Ylivainio K, Muñoz Guajardo I, Ortiz‐Pelaez A, Alvarez‐Ordóñez A. Inactivation of indicator microorganisms and biological hazards by standard and/or alternative processing methods in Category 2 and 3 animal by-products and derived products to be used as organic fertilisers and/or soil improvers. EFSA J 2021; 19:e06932. [PMID: 34900004 PMCID: PMC8638561 DOI: 10.2903/j.efsa.2021.6932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The European Commission requested EFSA to assess if different thermal processes achieve a 5 log10 reduction in Enterococcus faecalis or Salmonella Senftenberg (775W) and (if relevant) a 3 log10 reduction in thermoresistant viruses (e.g. Parvovirus) as well as if different chemical processes achieve a 3 log10 reduction of eggs of Ascaris sp., in eight groups of Category 2 and 3 derived products and animal by-products (ABP). These included (1) ash derived from incineration, co-incineration and combustion; (2) glycerine derived from the production of biodiesel and renewable fuels; (3) other materials derived from the production of biodiesel and renewable fuels; (4) hides and skins; (5) wool and hair; (6) feathers and down; (7) pig bristles; and (8) horns, horn products, hooves and hoof products. Data on the presence of viral hazards and on thermal and chemical inactivation of the targeted indicator microorganisms and biological hazards under relevant processing conditions were extracted via extensive literature searches. The evidence was assessed via expert knowledge elicitation. The certainty that the required log10 reductions in the most resistant indicator microorganisms or biological hazards will be achieved for each of the eight groups of materials mentioned above by the thermal and/or chemical processes was (1) 99-100% for the two processes assessed; (2) 98-100% in Category 2 ABP, at least 90-99% in Category 3 ABP; (3) 90-99% in Category 2 ABP; at least 66-90% in Category 3 ABP; (4) 10-66% and 33-66%; (5) 1-33% and 10-50%; (6) 66-90%; (7) 33-66% and 50-95%; (8) 66-95%, respectively. Data generation on the occurrence and reduction of biological hazards by thermal and/or chemical methods in these materials and on the characterisation of the usage pathways of ABP as organic fertilisers/soil improvers is recommended.
Collapse
|
22
|
Joshi LR, Knudsen D, Piñeyro P, Dhakal S, Renukaradhya GJ, Diel DG. Protective Efficacy of an Orf Virus-Vector Encoding the Hemagglutinin and the Nucleoprotein of Influenza A Virus in Swine. Front Immunol 2021; 12:747574. [PMID: 34804030 PMCID: PMC8602839 DOI: 10.3389/fimmu.2021.747574] [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: 07/26/2021] [Accepted: 09/30/2021] [Indexed: 01/19/2023] Open
Abstract
Swine influenza is a highly contagious respiratory disease of pigs caused by influenza A viruses (IAV-S). IAV-S causes significant economic losses to the swine industry and poses challenges to public health given its zoonotic potential. Thus effective IAV-S vaccines are needed and highly desirable and would benefit both animal and human health. Here, we developed two recombinant orf viruses, expressing the hemagglutinin (HA) gene (OV-HA) or the HA and the nucleoprotein (NP) genes of IAV-S (OV-HA-NP). The immunogenicity and protective efficacy of these two recombinant viruses were evaluated in pigs. Both OV-HA and OV-HA-NP recombinants elicited robust virus neutralizing antibody response in pigs, with higher levels of neutralizing antibodies (NA) being detected in OV-HA-NP-immunized animals pre-challenge infection. Although both recombinant viruses elicited IAV-S-specific T-cell responses, the frequency of IAV-S-specific proliferating CD8+ T cells upon re-stimulation was higher in OV-HA-NP-immunized animals than in the OV-HA group. Importantly, IgG1/IgG2 isotype ELISAs revealed that immunization with OV-HA induced Th2-biased immune responses, whereas immunization with OV-HA-NP virus resulted in a Th1-biased immune response. While pigs immunized with either OV-HA or OV-HA-NP were protected when compared to non-immunized controls, immunization with OV-HA-NP resulted in incremental protection against challenge infection as evidenced by a reduced secondary antibody response (NA and HI antibodies) following IAV-S challenge and reduced virus shedding in nasal secretions (lower viral RNA loads and frequency of animals shedding viral RNA and infectious virus), when compared to animals in the OV-HA group. Interestingly, broader cross neutralization activity was also observed in serum of OV-HA-NP-immunized animals against a panel of contemporary IAV-S isolates representing the major genetic clades circulating in swine. This study demonstrates the potential of ORFV-based vector for control of swine influenza virus in swine.
Collapse
Affiliation(s)
- Lok R Joshi
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States.,Department of Veterinary and Biomedical Sciences, Animal Disease Research And Diagnostic Laboratory, South Dakota State University, Brookings, SD, United States
| | - David Knudsen
- Department of Veterinary and Biomedical Sciences, Animal Disease Research And Diagnostic Laboratory, South Dakota State University, Brookings, SD, United States
| | - Pablo Piñeyro
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA, United States
| | - Santosh Dhakal
- Department of Veterinary Preventive Medicine, Center for Food Animal Health, Ohio State University, Wooster, OH, United States
| | - Gourapura J Renukaradhya
- Department of Veterinary Preventive Medicine, Center for Food Animal Health, Ohio State University, Wooster, OH, United States
| | - Diego G Diel
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States.,Department of Veterinary and Biomedical Sciences, Animal Disease Research And Diagnostic Laboratory, South Dakota State University, Brookings, SD, United States
| |
Collapse
|
23
|
Bukar AM, Jesse FFA, Abdullah CAC, Noordin MM, Lawan Z, Mangga HK, Balakrishnan KN, Azmi MLM. Immunomodulatory Strategies for Parapoxvirus: Current Status and Future Approaches for the Development of Vaccines against Orf Virus Infection. Vaccines (Basel) 2021; 9:1341. [PMID: 34835272 PMCID: PMC8624149 DOI: 10.3390/vaccines9111341] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 11/17/2022] Open
Abstract
Orf virus (ORFV), the prototype species of the parapoxvirus genus, is the causative agent of contagious ecthyma, an extremely devastating skin disease of sheep, goats, and humans that causes enormous economic losses in livestock production. ORFV is known for its ability to repeatedly infect both previously infected and vaccinated sheep due to several immunomodulatory genes encoded by the virus that temporarily suppress host immunity. Therefore, the development of novel, safe and effective vaccines against ORFV infection is an important priority. Although, the commercially licensed live-attenuated vaccines have provided partial protection against ORFV infections, the attenuated viruses have been associated with major safety concerns. In addition to safety issues, the persistent reinfection of vaccinated animals warrants the need to investigate several factors that may affect vaccine efficacy. Perhaps, the reason for the failure of the vaccine is due to the long-term adaptation of the virus in tissue culture. In recent years, the development of vaccines against ORFV infection has achieved great success due to technological advances in recombinant DNA technologies, which have opened a pathway for the development of vaccine candidates that elicit robust immunity. In this review, we present current knowledge on immune responses elicited by ORFV, with particular attention to the effects of the viral immunomodulators on the host immune system. We also discuss the implications of strain variation for the development of rational vaccines. Finally, the review will also aim to demonstrate future strategies for the development of safe and efficient vaccines against ORFV infections.
Collapse
Affiliation(s)
- Alhaji Modu Bukar
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
- Department of Science Laboratory Technology, School Agriculture and Applied Sciences, Ramat Polytechnic Maiduguri, Maiduguri 1070, Borno, Nigeria
| | - Faez Firdaus Abdullah Jesse
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | | | - Mustapha M. Noordin
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
| | - Zaharaddeen Lawan
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
| | - Hassana Kyari Mangga
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
| | - Krishnan Nair Balakrishnan
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
| | - Mohd-Lila Mohd Azmi
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.M.N.); (Z.L.); (H.K.M.); (K.N.B.)
| |
Collapse
|
24
|
Liao GR, Tseng YY, Tseng CY, Lo CY, Hsu WL. The orf virus (ORFV) protein OV20.0 interacts with the microprocessor complex subunit DGCR8 to regulate miRNA biogenesis and ORFV infection. FEBS Lett 2021; 595:2897-2908. [PMID: 34778960 DOI: 10.1002/1873-3468.14231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/15/2021] [Accepted: 11/03/2021] [Indexed: 02/01/2023]
Abstract
Cellular double-stranded RNA-binding proteins (DRBPs) play important roles in the regulation of innate immune responses and microRNA (miRNA) biogenesis. The current study aimed to understand whether OV20.0, a DRBP of orf virus (ORFV), is involved in cellular RNA biogenesis via association with host DRBPs. We found that OV20.0 interacts with DiGeorge syndrome critical region 8 (DGCR8), a subunit of the miRNA processor complex, and binds to primary- and precursor-miRNA. Additionally, OV20.0 regulates DGCR8 expression in multiple ways, including through interaction with the DGCR8 protein and binding to DGCR8 mRNA. Lastly, our data show that DGCR8 plays an antiviral role against ORFV infection, whereas it is beneficial for influenza virus propagation, indicating that the underlying mechanisms could be diverse among different viruses.
Collapse
Affiliation(s)
- Guan-Ru Liao
- Graduate Institute of Microbiology and Public Health, National Chung Hsing University, Taichung, Taiwan
| | - Yeu-Yang Tseng
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia
| | - Ching-Yu Tseng
- Graduate Institute of Microbiology and Public Health, National Chung Hsing University, Taichung, Taiwan
| | - Chen-Yu Lo
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, Taichung, Taiwan
| | - Wei-Li Hsu
- Graduate Institute of Microbiology and Public Health, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
25
|
Khatiwada S, Delhon G, Chaulagain S, Rock DL. The novel ORFV protein ORFV113 activates LPA-p38 signaling. PLoS Pathog 2021; 17:e1009971. [PMID: 34614034 PMCID: PMC8523077 DOI: 10.1371/journal.ppat.1009971] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 10/18/2021] [Accepted: 09/26/2021] [Indexed: 11/19/2022] Open
Abstract
Viruses have evolved mechanisms to subvert critical cellular signaling pathways that regulate a wide range of cellular functions, including cell differentiation, proliferation and chemotaxis, and innate immune responses. Here, we describe a novel ORFV protein, ORFV113, that interacts with the G protein-coupled receptor Lysophosphatidic acid receptor 1 (LPA1). Consistent with its interaction with LPA1, ORFV113 enhances p38 kinase phosphorylation in ORFV infected cells in vitro and in vivo, and in cells transiently expressing ORFV113 or treated with soluble ORFV113. Infection of cells with virus lacking ORFV113 (OV-IA82Δ113) significantly decreased p38 phosphorylation and viral plaque size. Infection of cells with ORFV in the presence of a p38 kinase inhibitor markedly diminished ORFV replication, highlighting importance of p38 signaling during ORFV infection. ORFV113 enhancement of p38 activation was prevented in cells in which LPA1 expression was knocked down and in cells treated with LPA1 inhibitor. Infection of sheep with OV-IA82Δ113 led to a strikingly attenuated disease phenotype, indicating that ORFV113 is a major virulence determinant in the natural host. Notably, ORFV113 represents the first viral protein that modulates p38 signaling via interaction with LPA1 receptor.
Collapse
Affiliation(s)
- Sushil Khatiwada
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Gustavo Delhon
- School of Veterinary Medicine and Biomedical Sciences, Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Sabal Chaulagain
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Daniel L. Rock
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| |
Collapse
|
26
|
Abstract
Orf virus (ORFV) is a highly epitheliotropic parapoxvirus with zoonotic significance that induces proliferative lesions in the skin of sheep, goats, and humans. Several viral proteins carried by ORFV, including nuclear factor-κB (NF-κB) inhibitors, play important roles in hijacking host-associated proteins for viral evasion of the host innate immune response. However, the roles of proteins with unknown functions in viral replication and latent infection remain to be explored. Here, we present data demonstrating that the ORF120, an early-late ORFV-encoded protein, activates the NF-κB pathway in the early phase of infection, which implies that ORFV may regulate NF-κB through a biphasic mechanism. A DUAL membrane yeast two-hybrid system and coimmunoprecipitation experiments revealed that the ORF120 protein interacts with Ras-GTPase-activating protein (SH3 domain) binding protein 1 (G3BP1). The overexpression of the ORF120 protein can efficiently increase the expression of G3BP1 and nuclear translocation of NF-κB-p65 in primary ovine fetal turbinate (OFTu) and HeLa cells. The knockdown of G3BP1 significantly decreased ORF120-induced NF-κB activation, indicating that G3BP1 is involved in ORF120-induced NF-κB pathway activation. A dual-luciferase reporter assay revealed that ORF120 could positively regulate the NF-κB pathway through the full-length G3BP1 or the domain of G3BP1RRM+RGG. In conclusion, we demonstrate, for the first time, that the ORF120 protein is capable of positively regulating NF-κB signaling by interacting with G3BP1, providing new insights into ORFV pathogenesis and a theoretical basis for antiviral drug design. IMPORTANCE As part of the host innate response, the nuclear factor-κB (NF-κB) pathway plays a partial antiviral role in nature by regulating the innate immune response. Thus, the NF-κB pathway is probably the most frequently targeted intracellular pathway for subversion by anti-immune modulators that are carried by a wide range of pathogens. Various viruses, including poxviruses, carry several proteins that prepare the host cell for viral replication by inhibiting cytoplasmic events, leading to the initiation of NF-κB transcriptional activity. However, NF-κB activity is hypothesized to facilitate viral replication to a great extent. The significance of our research is in the exploration of the activation mechanism of NF-κB induced by the Orf virus (ORFV) ORF120 protein interacting with G3BP1, which helps not only to explain the ability of ORFV to modulate the immune response through the positive regulation of NF-κB but also to show the mechanism by which the virus evades the host innate immune response.
Collapse
|
27
|
Liao GR, Tseng YY, Tseng CY, Huang YP, Tsai CH, Liu HP, Hsu WL. K160 in the RNA-binding domain of the orf virus virulence factor OV20.0 is critical for its functions in counteracting host antiviral defense. FEBS Lett 2021; 595:1721-1733. [PMID: 33909294 DOI: 10.1002/1873-3468.14099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/09/2021] [Accepted: 04/22/2021] [Indexed: 11/10/2022]
Abstract
The OV20.0 virulence factor of orf virus antagonizes host antiviral responses. One mechanism through which it functions is by inhibiting activation of the dsRNA-activated protein kinase R (PKR) by sequestering dsRNA and by physically interacting with PKR. Sequence alignment indicated that several key residues critical for dsRNA binding were conserved in OV20.0, and their contribution to OV20.O function was investigated in this study. We found that residues F141, K160, and R164 were responsible for the dsRNA-binding ability of OV20.0. Interestingly, mutation at K160 (K160A) diminished the OV20.0-PKR interaction and further reduced the inhibitory effect of OV20.0 on PKR activation. Nevertheless, OV20.0 homodimerization was not influenced by K160A. The contribution of the dsRNA-binding domain and K160 to the suppression of RNA interference by OV20.0 was further demonstrated in plants. In summary, K160 is essential for the function of OV20.0, particularly its interaction with dsRNA and PKR that ultimately contributes to the suppression of PKR activation.
Collapse
Affiliation(s)
- Guan-Ru Liao
- Graduate Institute of Microbiology and Public Health, National Chung Hsing University, Taichung, Taiwan
| | - Yeu-Yang Tseng
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Australia
| | - Ching-Yu Tseng
- Graduate Institute of Microbiology and Public Health, National Chung Hsing University, Taichung, Taiwan
| | - Ying-Ping Huang
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Ching-Hsiu Tsai
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Hao-Ping Liu
- Department of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Wei-Li Hsu
- Graduate Institute of Microbiology and Public Health, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
28
|
Riad S, Xiang Y, AlDaif B, Mercer AA, Fleming SB. Rescue of a Vaccinia Virus Mutant Lacking IFN Resistance Genes K1L and C7L by the Parapoxvirus Orf Virus. Front Microbiol 2020; 11:1797. [PMID: 32903701 PMCID: PMC7438785 DOI: 10.3389/fmicb.2020.01797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 07/09/2020] [Indexed: 12/17/2022] Open
Abstract
Type 1 interferons induce the upregulation of hundreds of interferon-stimulated genes (ISGs) that combat viral replication. The parapoxvirus orf virus (ORFV) induces acute pustular skin lesions in sheep and goats and can reinfect its host, however, little is known of its ability to resist IFN. Vaccinia virus (VACV) encodes a number of factors that modulate the IFN response including the host-range genes C7L and K1L. A recombinant VACV-Western Reserve (WR) strain in which the K1L and C7L genes have been deleted does not replicate in cells treated with IFN-β nor in HeLa cells in which the IFN response is constitutive and is inhibited at the level of intermediate gene expression. Furthermore C7L is conserved in almost all poxviruses. We provide evidence that shows that although ORFV is more sensitive to IFN-β compared with VACV, and lacks homologues of KIL and C7L, it nevertheless has the ability to rescue a VACV KIL- C7L- gfp+ mutant in which gfp is expressed from a late promoter. Co-infection of HeLa cells with the mutant and ORFV demonstrated that ORFV was able to overcome the block in translation of intermediate transcripts in the mutant virus, allowing it to progress to late gene expression and new viral particles. Our findings strongly suggest that ORFV encodes a factor(s) that, although different in structure to C7L or KIL, targets an anti-viral cellular mechanism that is a highly potent at killing poxviruses.
Collapse
Affiliation(s)
- Sherief Riad
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Yan Xiang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Basheer AlDaif
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Andrew A Mercer
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Stephen B Fleming
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
29
|
Hao JH, Kong HJ, Yan MH, Shen CC, Xu GW, Zhang DJ, Zhang KS, Zheng HX, Liu XT. Inhibition of orf virus replication in goat skin fibroblast cells by the HSPA1B protein, as demonstrated by iTRAQ-based quantitative proteome analysis. Arch Virol 2020; 165:2561-2587. [PMID: 32876795 PMCID: PMC7465882 DOI: 10.1007/s00705-020-04789-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 07/26/2020] [Indexed: 02/07/2023]
Abstract
Orf virus (ORFV) infects sheep and goat tissues, resulting in severe proliferative lesions. To analyze cellular protein expression in ORFV-infected goat skin fibroblast (GSF) cells, we used two-dimensional liquid chromatography-tandem mass spectrometry coupled with isobaric tags for relative and absolute quantification (iTRAQ). The proteomics approach was used along with quantitative reverse transcription polymerase chain reaction (RT-qPCR) to detect differentially expressed proteins in ORFV-infected GSF cells and mock-infected GSF cells. A total of 282 differentially expressed proteins were identified. It was found that 222 host proteins were upregulated and 60 were downregulated following viral infection. We confirmed that these proteins were differentially expressed and found that heat shock 70-kDa protein 1B (HSPA1B) was differentially expressed and localized in the cytoplasm. It was also noted that HSPA1B caused inhibition of viral proliferation, in the middle and late stages of viral infection. The differentially expressed proteins were associated with the biological processes of viral binding, cell structure, signal transduction, cell adhesion, and cell proliferation.
Collapse
Affiliation(s)
- Jun-Hong Hao
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China
| | - Han-Jin Kong
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China
| | - Ming-Hao Yan
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China
| | - Chao-Chao Shen
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China
| | - Guo-Wei Xu
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China
| | - Da-Jun Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China
| | - Ke-Shan Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China.
| | - Hai-Xue Zheng
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China
| | - Xiang-Tao Liu
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, No. 1, Xujiaping, Lanzhou, 730046, Gansu, People's Republic of China
| |
Collapse
|
30
|
Lothert K, Pagallies F, Eilts F, Sivanesapillai A, Hardt M, Moebus A, Feger T, Amann R, Wolff MW. A scalable downstream process for the purification of the cell culture-derived Orf virus for human or veterinary applications. J Biotechnol 2020; 323:221-230. [PMID: 32860824 DOI: 10.1016/j.jbiotec.2020.08.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/06/2020] [Accepted: 08/23/2020] [Indexed: 12/11/2022]
Abstract
The large demand for safe and efficient viral vector-based vaccines and gene therapies against both inherited and acquired diseases accelerates the development of viral vectors. One outstanding example, the Orf virus, has a wide range of applications, a superior efficacy and an excellent safety profile combined with a reduced pathogenicity compared to other viral vectors. However, besides these favorable attributes, an efficient and scalable downstream process still needs to be developed. Recently, we screened potential chromatographic stationary phases for Orf virus purification. Based on these previous accomplishments, we developed a complete downstream process for the cell culture-derived Orf virus. The described process comprises a membrane-based clarification step, a nuclease treatment, steric exclusion chromatography, and a secondary chromatographic purification step using Capto® Core 700 resin. The applicability of this process to a variety of diverse Orf virus vectors was shown, testing two different genotypes. These studies render the possibility to apply the developed downstream scheme for both genotypes, and lead to overall virus yields of about 64 %, with step recoveries of >70 % for the clarification, and >90 % for the chromatography train. Protein concentrations of the final product are below the detection limits, and the final DNA concentration of about 1 ng per 1E + 06 infective virus units resembles a total DNA depletion of 96-98 %.
Collapse
Affiliation(s)
- Keven Lothert
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Giessen, Germany
| | - Felix Pagallies
- Department of Immunology, University of Tuebingen, Tuebingen, Germany
| | - Friederike Eilts
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Giessen, Germany
| | - Arabi Sivanesapillai
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Giessen, Germany
| | - Martin Hardt
- Imaging Unit, Biomedical Research Centre Seltersberg, Justus Liebig University, Giessen, Germany
| | - Anna Moebus
- Imaging Unit, Biomedical Research Centre Seltersberg, Justus Liebig University, Giessen, Germany
| | - Thomas Feger
- Department of Immunology, University of Tuebingen, Tuebingen, Germany
| | - Ralf Amann
- Department of Immunology, University of Tuebingen, Tuebingen, Germany
| | - Michael W Wolff
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Giessen, Germany; Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Giessen, Germany.
| |
Collapse
|
31
|
Orf Virus-Based Therapeutic Vaccine for Treatment of Papillomavirus-Induced Tumors. J Virol 2020; 94:JVI.00398-20. [PMID: 32404527 DOI: 10.1128/jvi.00398-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/01/2020] [Indexed: 12/17/2022] Open
Abstract
Orf virus (ORFV) represents a suitable vector for the generation of efficient, prophylactic antiviral vaccines against different pathogens. The present study investigated for the first time the therapeutic application of ORFV vector-based vaccines against tumors induced by cottontail rabbit papillomavirus (CRPV). ORFV-CRPV recombinants were constructed expressing the early CRPV gene E1, E2, E7, or LE6. In two independent experiments we used in total 23 rabbits which were immunized with a mixture of the four ORFV-CRPV recombinants or empty ORFV vector as a control 5 weeks after the appearance of skin tumors. For the determination of the therapeutic efficacy, the subsequent growth of the tumors was recorded. In the first experiment, we could demonstrate that three immunizations of rabbits with high tumor burden with the combined four ORFV-CRPV recombinants resulted in significant growth retardation of the tumors compared to the control. A second experiment was performed to test the therapeutic effect of 5 doses of the combined vaccine in rabbits with a lower tumor burden than in nonimmunized rabbits. Tumor growth was significantly reduced after immunization, and one vaccinated rabbit even displayed complete tumor regression until the end of the observation period at 26 weeks. Results of delayed-type hypersensitivity (DTH) skin tests suggest the induction of a cellular immune response mediated by the ORFV-CRPV vaccine. The data presented show for the first time a therapeutic potential of the ORFV vector platform and encourage further studies for the development of a therapeutic vaccine against virus-induced tumors.IMPORTANCE Viral vectors are widely used for the development of therapeutic vaccines for the treatment of tumors. In our study we have used Orf virus (ORFV) strain D1701-V for the generation of recombinant vaccines expressing cottontail rabbit papillomavirus (CRPV) early proteins E1, E2, LE6, and E7. The therapeutic efficacy of the ORFV-CRPV vaccines was evaluated in two independent experiments using the outbred CRPV rabbit model. In both experiments the immunization achieved significant suppression of tumor growth. In total, 84.6% of all outbred animals benefited from the ORFV-CRPV vaccination, showing reduction in tumor size and significant tumor growth inhibition, including one animal with complete tumor regression without recurrence.
Collapse
|
32
|
Blick AK, Hensel M, Rodrigues Hoffmann A, Gibbons P, Porter BF. Pathology in Practice. J Am Vet Med Assoc 2020; 255:313-316. [PMID: 31298639 DOI: 10.2460/javma.255.3.313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
33
|
Shimizu K, Takaiwa A, Takeshima SN, Okada A, Inoshima Y. Genetic Variability of 3'-Proximal Region of Genomes of Orf Viruses Isolated From Sheep and Wild Japanese Serows ( Capricornis crispus) in Japan. Front Vet Sci 2020; 7:188. [PMID: 32391386 PMCID: PMC7193945 DOI: 10.3389/fvets.2020.00188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/23/2020] [Indexed: 12/04/2022] Open
Abstract
Orf virus is a prototype species of the genus Parapoxvirus, subfamily Chordopoxvirinae, family Poxviridae. Japanese orf viruses, infecting sheep and wild Japanese serows (Capricornis crispus), have been considered to be genetically closely related based on the sequence identities of the open reading frames (ORFs) 11, 20, and 132 in their genomes. However, since the genome size of orf viruses is about 140 kbp long, genetic variation among Japanese orf viruses remains unclear. In this study, we analyzed the sequences of ORFs 117, 119, 125, and 127 located in the 3′-proximal region of the viral genome using two strains from sheep and three strains from Japanese serows isolated from 1970 to 2007, and compared them with the corresponding sequences of reference orf viruses from other countries. Sequence analysis revealed that ORFs 125 and 127, which encode the inhibitor of apoptosis and viral interleukin (IL)-10, respectively, were highly conserved among the five Japanese orf viruses. However, high genetic variability with deletions or duplications was observed in ORFs 117 and 119, which encode granulocyte macrophage colony-stimulating factor and IL-2 inhibition factor (GIF), and inducer of cell apoptosis, respectively, in one strain from sheep and two strains from Japanese serows. Our results suggest that genetic variability exists in Japanese orf viruses even in the same host species. This is the first report of genetic variability of orf viruses in Japan.
Collapse
Affiliation(s)
- Kaori Shimizu
- Laboratory of Food and Environmental Hygiene, Cooperative Department of Veterinary Medicine, Gifu University, Gifu, Japan
| | - Asari Takaiwa
- Laboratory of Food and Environmental Hygiene, Cooperative Department of Veterinary Medicine, Gifu University, Gifu, Japan
| | | | - Ayaka Okada
- Laboratory of Food and Environmental Hygiene, Cooperative Department of Veterinary Medicine, Gifu University, Gifu, Japan.,Education and Research Center for Food Animal Health, Gifu University (GeFAH), Gifu, Japan
| | - Yasuo Inoshima
- Laboratory of Food and Environmental Hygiene, Cooperative Department of Veterinary Medicine, Gifu University, Gifu, Japan.,Education and Research Center for Food Animal Health, Gifu University (GeFAH), Gifu, Japan.,The United Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan.,Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| |
Collapse
|
34
|
Yaron JR, Zhang L, Guo Q, Burgin M, Schutz LN, Awo E, Wise L, Krause KL, Ildefonso CJ, Kwiecien JM, Juby M, Rahman MM, Chen H, Moyer RW, Alcami A, McFadden G, Lucas AR. Deriving Immune Modulating Drugs from Viruses-A New Class of Biologics. J Clin Med 2020; 9:E972. [PMID: 32244484 PMCID: PMC7230489 DOI: 10.3390/jcm9040972] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/19/2020] [Accepted: 03/23/2020] [Indexed: 02/07/2023] Open
Abstract
Viruses are widely used as a platform for the production of therapeutics. Vaccines containing live, dead and components of viruses, gene therapy vectors and oncolytic viruses are key examples of clinically-approved therapeutic uses for viruses. Despite this, the use of virus-derived proteins as natural sources for immune modulators remains in the early stages of development. Viruses have evolved complex, highly effective approaches for immune evasion. Originally developed for protection against host immune responses, viral immune-modulating proteins are extraordinarily potent, often functioning at picomolar concentrations. These complex viral intracellular parasites have "performed the R&D", developing highly effective immune evasive strategies over millions of years. These proteins provide a new and natural source for immune-modulating therapeutics, similar in many ways to penicillin being developed from mold or streptokinase from bacteria. Virus-derived serine proteinase inhibitors (serpins), chemokine modulating proteins, complement control, inflammasome inhibition, growth factors (e.g., viral vascular endothelial growth factor) and cytokine mimics (e.g., viral interleukin 10) and/or inhibitors (e.g., tumor necrosis factor) have now been identified that target central immunological response pathways. We review here current development of virus-derived immune-modulating biologics with efficacy demonstrated in pre-clinical or clinical studies, focusing on pox and herpesviruses-derived immune-modulating therapeutics.
Collapse
Affiliation(s)
- Jordan R. Yaron
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Liqiang Zhang
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Qiuyun Guo
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Michelle Burgin
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Lauren N. Schutz
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Enkidia Awo
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Lyn Wise
- University of Otago, Dunedin 9054, New Zealand; (L.W.); (K.L.K.)
| | - Kurt L. Krause
- University of Otago, Dunedin 9054, New Zealand; (L.W.); (K.L.K.)
| | | | - Jacek M. Kwiecien
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S4L8, Canada
| | - Michael Juby
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Masmudur M. Rahman
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Hao Chen
- The Department of Tumor Surgery, Second Hospital of Lanzhou University, Lanzhou 730030, China;
| | - Richard W. Moyer
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA;
| | - Antonio Alcami
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid), Cantoblanco, 28049 Madrid, Spain;
| | - Grant McFadden
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Alexandra R. Lucas
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
- St Joseph Hospital, Dignity Health, Creighton University, Phoenix, AZ 85013, USA
| |
Collapse
|
35
|
Proteomic analyses reveal that Orf virus induces the activation and maturation of mouse bone marrow-derived dendritic cells. Res Vet Sci 2020; 132:563-573. [PMID: 32466866 DOI: 10.1016/j.rvsc.2020.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 01/07/2020] [Accepted: 02/12/2020] [Indexed: 12/13/2022]
Abstract
Orf virus (ORFV) is known for its immunostimulatory capacities and has been utilized as an efficient viral vector vaccine in non-permissive host species. Murine bone marrow-derived dendritic cells (BMDCs) are able to react with ORFV. In this study, we aimed to identify pivotal differentially expressed proteins involved in the process of DCs' differentiation in response to ORFV. Our findings showed that ORFV activates the maturation and differentiation of DCs. We further identified and validated seven differentially expressed proteins following ORFV stimulation. With functions in biological processes such as stimulus response, DCs maturation, antigen presentation and Th1 cell activation. Western blot analyses validated the respective changes in protein expression. The huge number of differentially expressed proteins identified in this study will be valuable for elucidating the mechanisms underlying ORFV-induced immunomodulation of murine BMDCs.
Collapse
|
36
|
Ma WT, Liu Q, Ning MX, Qi YX, Rehman S, Chen DK. Development and applications of a monoclonal antibody against caprine interferon-gamma. BMC Biotechnol 2019; 19:102. [PMID: 31870349 PMCID: PMC6929374 DOI: 10.1186/s12896-019-0596-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 12/18/2019] [Indexed: 12/20/2022] Open
Abstract
Background Interferon-gamma (IFN-γ) is an important mediator of type I immune response and has antiviral, immunoregulatory and anti-tumor properties, plays a wide range of roles in inflammation and autoimmune diseases. The aim of this study was to obtain monoclonal antibody (mAb) against caprine IFN-γ by immunizing of BALB/c mice with the purified rIFN-γ. Results Recombinant caprine IFN-γ was expressed in Escherichia coli strain BL21 (DE3) and monoclonal antibodies against caprine IFN-γ were produced by immunizing of BALB/c mice with rIFN-γ. One hybridoma secreting mAb was screened by enzyme-linked immunosorbent assay (ELISA) which was designated as 2C. MAb secreted by this cell line were analyzed through ELISA, western blot and application of the mAb was evaluated by immunofluorescence analysis using goat lip tissues infected with Orf virus. ELISA analysis revealed that mAb 2C can specifically recognize rIFN-γ protein and culture supernatant of goat peripheral blood mononuclear cells (PBMCs) stimulated by concanavalin A (Con A) but cannot recognize the fusion tag protein of pET-32a. Western blot analysis showed that mAb 2C can specifically react with the purified 34.9 kDa rIFN-γ protein but does not react with the fusion tag protein of pET-32a. Immunofluorescence results demonstrated that mAb 2C can detect IFN-γ secreted in histopathological sites of goats infected with Orf virus. Conclusions A caprine IFN-γ-specific mAb was successfully developed in this study. Further analyses showed that the mAb can be used to detect IFN-γ expression level during contagious ecthyma in goats.
Collapse
Affiliation(s)
- Wen-Tao Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi Province, China
| | - Qi Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi Province, China
| | - Meng-Xia Ning
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi Province, China
| | - Yu-Xu Qi
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi Province, China
| | - Saad Rehman
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi Province, China
| | - De-Kun Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi Province, China.
| |
Collapse
|
37
|
Wassie T, Fanmei Z, Jiang X, Liu G, Girmay S, Min Z, Chenhui L, Bo DD, Ahmed S. Recombinant B2L and Kisspeptin-54 DNA Vaccine Induces Immunity Against Orf Virus and Inhibits Spermatogenesis In Rats. Sci Rep 2019; 9:16262. [PMID: 31700161 PMCID: PMC6838309 DOI: 10.1038/s41598-019-52744-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 10/08/2019] [Indexed: 02/06/2023] Open
Abstract
Orf is a highly contagious zoonotic disease of small ruminants caused by Parapoxvirus. Kisspeptin, encoded by the KISS1 gene with its cognate receptor GPR-54 is recognized as an upstream orchestrator in the hypothalamic-pituitary-gonadal axis. This study was designed to construct a DNA vaccine that produces a fused peptide composed of a major immunodominant protein of the orf virus (B2L) and kisspeptin-54, a neuropeptide with recognized roles in mammalian reproductive biology. The administration of this recombinant vaccine is shown to produce a significant antibody and cell-mediated immune response directed against B2L compared to the control group (p < 0.05). Furthermore, we found that rats inoculated with PBK-asd vaccine up-regulated antigen-mediated splenocyte proliferation and significantly raised antigen-specific tumor necrosis factor-alpha (TNFα-), interferon-gamma (IFN-ϒ) and interleukin (IL-2) compared to the control group (p < 0.05). This recombinant vaccine also stimulated antibody responses to kisspeptin and decreased serum luteinizing hormone and testosterone levels. Moreover, the current recombinant vaccine caused testicular atrophy and arrested spermatogenesis. It is concluded that this recombinant B2L and Kisspeptin-54 vaccine could be a promising approach for construction of bivalent orf virus and immunocastration vaccine. Furthermore, we concluded that the orf virus envelope protein (B2L) could be used as an immunomodulator for kisspeptin-54 to produce a strong antibody response.
Collapse
Affiliation(s)
- Teketay Wassie
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.,Laboratory of Sheep and Goat Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Zeng Fanmei
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Xunping Jiang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China. .,Laboratory of Sheep and Goat Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China.
| | - Guiqiong Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.,Laboratory of Sheep and Goat Genetics, Breeding and Reproduction, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Shishay Girmay
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Zhang Min
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Liu Chenhui
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Dong Dong Bo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Sohail Ahmed
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| |
Collapse
|
38
|
Chantrey J, Dale T, Jones D, Begon M, Fenton A. The drivers of squirrelpox virus dynamics in its grey squirrel reservoir host. Epidemics 2019; 28:100352. [PMID: 31327730 DOI: 10.1016/j.epidem.2019.100352] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/02/2019] [Accepted: 07/02/2019] [Indexed: 11/30/2022] Open
Abstract
Many pathogens of conservation concern circulate endemically within natural wildlife reservoir hosts and it is imperative to understand the individual and ecological drivers of natural transmission dynamics, if any threat to a related endangered species is to be assessed. Our study highlights the key drivers of infection and shedding dynamics of squirrelpox virus (SQPV) in its reservoir grey squirrel (Sciurus carolinensis) population. To clarify SQPV dynamics in this population, longitudinal data from a 16-month mark-recapture study were analysed, combining serology with real-time quantitative PCR to identify periods of acute viraemia and chronic viral shedding. At the population level, we found SQPV infection prevalence, viral load and shedding varied seasonally, peaking in autumn and early spring. Individually, SQPV was shown to be a chronic infection in >80% of grey squirrels, with viral loads persisting over time and bouts of potential recrudescence or reinfection occurring. A key recurring factor significantly associated with SQPV infection risk was the presence of co-infecting squirrel adenovirus (ADV). In dual infected squirrels, longitudinal analysis showed that prior ADV viraemia increased the subsequent SQPV load in the blood. However, there was a strong, negative association between prior ADV viraemia and subsequent SQPV shedding from the forearm, probably caused by ADV prolonging the SQPV acute viraemic phase, so delaying onset of the chronic shedding phase, and thereby altering viral shedding patterns over the time scales examined here. Hence, co-circulating ADV infection may be involved in mediating both the quantitative levels of SQPV infection and the timing and degree of subsequent infectiousness of grey squirrels.
Collapse
Affiliation(s)
- Julian Chantrey
- Institute of Integrative Biology, University of Liverpool, Biosciences Building, Crown Street, Liverpool, L69 7ZB, UK; Institute of Infection and Global Health, University of Liverpool, Leahurst Campus, Neston, CH64 7TE, UK.
| | - Timothy Dale
- Institute of Integrative Biology, University of Liverpool, Biosciences Building, Crown Street, Liverpool, L69 7ZB, UK
| | - David Jones
- Institute of Infection and Global Health, University of Liverpool, Leahurst Campus, Neston, CH64 7TE, UK
| | - Michael Begon
- Institute of Integrative Biology, University of Liverpool, Biosciences Building, Crown Street, Liverpool, L69 7ZB, UK
| | - Andy Fenton
- Institute of Integrative Biology, University of Liverpool, Biosciences Building, Crown Street, Liverpool, L69 7ZB, UK
| |
Collapse
|
39
|
Zhong J, Guan J, Zhou Y, Cui S, Wang Z, Zhou S, Xu M, Wei X, Gao Y, Zhai S, Song D, He W, Gao F, Zhao K. Genomic characterization of two Orf virus isolates from Jilin province in China. Virus Genes 2019; 55:490-501. [PMID: 31030330 DOI: 10.1007/s11262-019-01666-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 04/17/2019] [Indexed: 02/02/2023]
Abstract
Orf virus (ORFV), a typical member of the Parapoxvirus genus within the family Poxviridae, which is the causative agent of Orf, a common epitheliotropic viral disease of sheep, goats, wild ruminants, and humans. In the present study, we sequenced the complete genomic sequences of two ORFV strains (ORFV-SY17, isolated from sheep, and ORFV-NA17, isolated from goat) and conducted the comparative analysis of multiple ORFVs. The complete genomic sequence of ORFV-SY17 was at length of 140,413 bp, including 131 potential open reading frames (ORFs) flanked by inverted terminal repeats (ITRs) of 4267 bp at both ends. The ORFV-NA17 strain displayed the similar genome structure with ORFV-SY17. The whole genomic sequence of ORFV-NA17 strain was 139,287 bp in length and contained 132 ORFs flanked by ITRs of 3974 bp. The overall G+C contents of ORFV-SY17 and ORFV-NA17 genome sequences were about 63.8% and 63.7%, respectively. The ITR sequences analysis showed that ORFV-SY17 and ORFV-NA17 contained the terminal BamHI sites and conserved telomere resolution sequences at both ends of their genome. In addition, comparative analysis of ORFs among ORFV-SY17, ORFV-NA17, and other ORFV strains revealed several sequence variations caused by insertions or deletions, especially in ORFs 005 and 116, which were very likely associated with host species. Phylogenetic analysis based on the complete genome sequences revealed that ORFV-SY17 was genetically closely related to NA1/11 and HN3/12 strains derived from sheep, while ORFV-NA17 was closely related to YX strain derived from goat. The multiple alignment of deduced amino acid sequences further revealed the genetic relationship between host species and genetic variations of ORFV strains. Taken together, the availability of genomic sequences of ORFV-SY17 and ORFV-NA17 strains from Jilin Province will aid in our understanding of the genetic diversity and evolution of ORFV strains in this region and can assist in distinguishing between ORFV strains that originate in sheep and goats.
Collapse
Affiliation(s)
- Jiawei Zhong
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jiyu Guan
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yanlong Zhou
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shanshan Cui
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhenzhen Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shuaishuai Zhou
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Mengshi Xu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xinyu Wei
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yu Gao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shaohua Zhai
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.,College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Deguang Song
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Wenqi He
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Feng Gao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Kui Zhao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.
| |
Collapse
|
40
|
LIN YUSHENG, JIANG JINXIU. TaqMan-based real-time quantitative fluorescence PCR for detection of Orf virus. THE INDIAN JOURNAL OF ANIMAL SCIENCES 2019. [DOI: 10.56093/ijans.v89i3.88032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Contagious ecthyma, also known as scabby mouth or Orf, is a zoonosis, which is caused by the Orf virus (ORFV). Human contact with infected animals can cause cutaneous lesions. To prevent and control ORFV effectively, rapid detection method is very important and highly needed. Real-time quantitative fluorescence PCR (qPCR) assay is considered as a rapid techonology to detect ORFV, and has been used for clinical diagnosis and epidemiological investigation. In present study, we developed a TaqMan-based qPCR assay for detection of ORFV. Beacon Designer 7.9 was used to design specific primers and probes were based on the ORFV020 gene sequence of the virus (GenBank Accession No. KF666563.1). The method had no cross-reactions with other common bacteria and viruses, was highly specific; the sensitivity test result showed that it could detect 10 copies of ORFV genomic DNA, and was more sensitive than conventional PCR. Both intra- and inter-variabilities were less than 2%, indicating the high stability and repeatability of the method. Additionally, 99 clinical samples from sheep and goats with suspected contagious ecthyma were tested using the developed assay and conventional PCR. The results showed that the developed assay was more sensitive and faster than conventional PCR. It can be concluded that the assay was suitable for routine detection of the ORFV and the epidemiological investigation.
Collapse
|
41
|
da Costa RA, Cargnelutti JF, Schild CO, Flores EF, Riet-Correa F, Giannitti F. Outbreak of contagious ecthyma caused by Orf virus (Parapoxvirus ovis) in a vaccinated sheep flock in Uruguay. Braz J Microbiol 2019; 50:565-569. [PMID: 30835059 DOI: 10.1007/s42770-019-00057-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 01/07/2019] [Indexed: 11/26/2022] Open
Abstract
Orf virus (ORFV) causes contagious ecthyma (CE), a highly transmissible, zoonotic disease of small ruminants. CE most commonly affects lambs and unvaccinated sheep. This work reports epidemiologic, clinicopathologic, and virologic findings in a CE outbreak in a vaccinated sheep flock in Uruguay and failure to detect ORFV in a commercial vaccine.
Collapse
Affiliation(s)
- Ricardo Almeida da Costa
- Plataforma de Investigación en Salud Animal, Instituto Nacional de Investigación Agropecuaria (INIA), Ruta 50 Km 11, La Estanzuela, Colonia, Uruguay
| | | | - Carlos Omar Schild
- Plataforma de Investigación en Salud Animal, Instituto Nacional de Investigación Agropecuaria (INIA), Ruta 50 Km 11, La Estanzuela, Colonia, Uruguay
| | - Eduardo Furtado Flores
- Setor de Virologia Veterinária, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Franklin Riet-Correa
- Plataforma de Investigación en Salud Animal, Instituto Nacional de Investigación Agropecuaria (INIA), Ruta 50 Km 11, La Estanzuela, Colonia, Uruguay
| | - Federico Giannitti
- Plataforma de Investigación en Salud Animal, Instituto Nacional de Investigación Agropecuaria (INIA), Ruta 50 Km 11, La Estanzuela, Colonia, Uruguay.
- Veterinary Population Medicine Department, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, USA.
| |
Collapse
|
42
|
Li W, Chen H, Deng H, Kuang Z, Long M, Chen D, Liao X, Li M, Rock DL, Luo S, Hao W. Orf Virus Encoded Protein ORFV119 Induces Cell Apoptosis Through the Extrinsic and Intrinsic Pathways. Front Microbiol 2018; 9:1056. [PMID: 29896166 PMCID: PMC5986898 DOI: 10.3389/fmicb.2018.01056] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 05/03/2018] [Indexed: 12/15/2022] Open
Abstract
Apoptosis, a significant form of cell death, has a leading role in the host cell defense against virus infection. Viruses have evolved a series of strategies that block apoptosis during the early stage of viral infection to enhance viral replication, and induce apoptosis in the late stages to facilitate viral particle release from the cells. Here we show that orf virus (ORFV), the causative agent of orf, encodes an apoptosis-inducing protein ORFV119. ORFV119 targets the mitochondria in host cells, inhibits cell proliferation, and induces cell apoptosis. Protein array data indicated that ORFV119 could induce apoptosis via up-regulation of Smac, Bak, and Bax and down-regulation of anti-apoptotic proteins Bcl-2 and cIAP-2. Activation of caspase-9 and caspase-3, and consequent PARP cleavage, ultimately lead to apoptosis. ORFV119 could also directly activate caspase-8 and induce Bid, involved in the extrinsic pathway, to achieve cell death. Furthermore, sequence analysis and experiments with mutants of ORFV119 introduced revealed that ORFV119 contains a key N-terminal domain that is necessary and sufficient to direct the protein to the mitochondria. Together, we report, for the first time, the identification of the novel apoptosis-inducing protein ORFV119 encoded by a parapoxvirus. This provides an important reference for the study of pathogenesis, identification of immunomodulation mechanisms of ORFV, and may lead to new strategies for orf disease control.
Collapse
Affiliation(s)
- Wei Li
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Huiqin Chen
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Hao Deng
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Zhenzhan Kuang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Mingjian Long
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Daxiang Chen
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xiaoqing Liao
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Ming Li
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Daniel L Rock
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Champaign-Urbana, Urbana, IL, United States
| | - Shuhong Luo
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China.,Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Foshan, China
| | - Wenbo Hao
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
43
|
KARABASANAVAR NAGAPPA, BAGALKOTE PRASHANTHS, RAJASHEKARA DB, MANJUNATHA SS, VEERANNA KC. Phylogenetic analysis of Orf virus associated with contagious ecthyma (orf) outbreak in Tellicherry goats (Capra hircus). THE INDIAN JOURNAL OF ANIMAL SCIENCES 2018. [DOI: 10.56093/ijans.v88i2.79288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Orf virus (ORFV) is a member of genus Parapoxvirus that causes contagious ecthyma in goats. A pox-like disease was investigated in Tellicherry goats (31 female) maintained at a semi-organized farm. History revealed recent introduction of Tellicherry goats for breeding purpose and housing of the new entrants in to a farm already having a mild form of pox-like disease. Newly introduced and stressed Tellicherry goats developed severe form of infection with 100% morbidity. Affected goats showed lesions around lips (100%), commissure (53%) and oral cavity (65%); exanthematic dermatitis was evident in 94% of the affected goats followed by ulceration (47%) and nodular lesions (24%). Scab samples were collected from affected goats to confirm the clinical diagnosis. Genus Parapoxvirus was confirmed by the amplification of specific 594 bp and 235 bp amplicons. Further, Orf virus specific amplicon of size 1,206 bp was also amplified for the confirmation. Sequence analysis of PCR amplicons showed close resemblance of the outbreak strain with reported Indian Orf virus isolates. Based on the homology of the outer envelope protein B2L gene sequence of Orf virus, the source of infection to the Tellicherry goats was traced to the local goat. Although Orf virus is zoonotic; however, no occupational transmission was noticed in the present outbreak.
Collapse
|
44
|
Effect of dexamethasone administration on the pathogenicity and lesion severity in rats experimentally inoculated with Orf virus (Malaysian isolates). ACTA ACUST UNITED AC 2018. [DOI: 10.1007/s00580-018-2726-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
45
|
Jia H, Zhan L, Wang X, He X, Chen G, Zhang Y, Feng Y, Wei Y, Zhang Y, Jing Z. Transcriptome analysis of sheep oral mucosa response to Orf virus infection. PLoS One 2017; 12:e0186681. [PMID: 29073164 PMCID: PMC5658058 DOI: 10.1371/journal.pone.0186681] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 10/05/2017] [Indexed: 01/15/2023] Open
Abstract
Contagious ecthyma is a highly contagious disease with worldwide distribution, which is caused by the Orf virus (ORFV) belonging to the Parapoxvirus. To study the alteration of host gene expression in response to ORFV infection at the transcriptional level, several young small-tailed Han sheep were inoculated with ORFV, and their oral mucosa tissue samples (T0, T3, T7 and T15) were collected on day 0, 3, 7 and 15 after ORFV infection respectively. RNA-seq transcriptome comparisons were performed, showing that 1928, 3219 and 2646 differentially expressed genes (DEGs) were identified among T3 vs. T0, T7 vs. T0, and T15 vs. T0 respectively. Gene Ontology (GO) analyses of the DEGs from these comparisons, revealed that ORFV might provoke vigorous immune response of the host cells during the early stage of infection. Moreover, GO and network analysis showed that positive and negative regulative mechanisms of apoptosis were integrated in the host cells through up or down-regulating the expression level of DEGs involved in apoptotic pathways, in order to reach a homeostasis of oral mucosa tissues during the exposure to ORFV infection. In conclusion, our study for the first time describes the direct effects of ORFV on the global host gene expression of its host using high-throughput RNA sequencing, which provides a resource for future characterizing the interaction mechanism between the mammalian host and ORFV.
Collapse
Affiliation(s)
- Huaijie Jia
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Leilei Zhan
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, China
| | - Xiaoxia Wang
- School of Public Health, Faculty of Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaobing He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Guohua Chen
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yu Zhang
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, China
| | - Yuan Feng
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yaxun Wei
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, China
| | - Yi Zhang
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei, China
- Laboratory for Genome Regulation and Human Health, ABLife Inc., Wuhan, Hubei, China
| | - Zhizhong Jing
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- * E-mail:
| |
Collapse
|
46
|
Moore RL, Isberg SR, Shilton CM, Milic NL. Impact of poxvirus lesions on saltwater crocodile (Crocodylus porosus) skins. Vet Microbiol 2017; 211:29-35. [PMID: 29102118 DOI: 10.1016/j.vetmic.2017.09.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 09/25/2017] [Accepted: 09/25/2017] [Indexed: 10/18/2022]
Abstract
Cutaneous poxvirus infections are common in several crocodilian species and are of importance in crocodile farming due to their potential impact on the tanned hide. To confirm poxvirus infection and understand the impact on saltwater crocodile (Crocodylus porosus) skin, fourteen animals from different age groups (five hatchlings, five yearlings and four grow-outs) were selected based on a criterion of ten poxvirus-like lesions per animal. One lesion on each animal was extruded for genetic analysis and transmission electron microscopy. Both methods confirmed poxvirus so the remainder of lesions were re-examined every six weeks over a 24 week study period. Each lesion went through four distinct phases: early active, active, expulsion and healing. To understand how these lesions impact on the final skin product, one crocodile from each age group was euthanised and the lesions examined. Using standard skin grading techniques (light-table), the early phase (early active - expulsion) lesions were all translucent and would lead to downgrading of the skin or, at worst, rendering them unsaleable. At the later stages of healing, the translucency reduces. Histological examination of the phases confirm that the basement membrane is not breached by the infection further indicating that poxvirus lesions, given enough time, will eventually have no detrimental effect on skin quality. This is obviously dependent upon no more lesions developing in the interim.
Collapse
Affiliation(s)
- Rhiannon L Moore
- School of Psychology and Clinical Sciences Charles Darwin University, NT 0909, Australia
| | - Sally R Isberg
- School of Psychology and Clinical Sciences Charles Darwin University, NT 0909, Australia; Centre for Crocodile Research, PO Box 329, Noonamah, NT 0837, Australia
| | - Cathy M Shilton
- Berrimah Veterinary Laboratory, Berrimah Farm, Northern Territory, Australia
| | - Natalie L Milic
- School of Psychology and Clinical Sciences Charles Darwin University, NT 0909, Australia.
| |
Collapse
|
47
|
Is orf infection a risk to expanding goat production in developing countries? A study from Lao PDR. Small Rumin Res 2017. [DOI: 10.1016/j.smallrumres.2017.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
48
|
Chen H, Li W, Kuang Z, Chen D, Liao X, Li M, Luo S, Hao W. The whole genomic analysis of orf virus strain HN3/12 isolated from Henan province, central China. BMC Vet Res 2017; 13:260. [PMID: 28821255 PMCID: PMC5562994 DOI: 10.1186/s12917-017-1178-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 08/10/2017] [Indexed: 01/23/2023] Open
Abstract
Background The Orf virus (ORFV) is the causative agent of orf, a globally-occurring, acute, pustular, contagious disease affecting sheep, goats and humans with a worldwide distribution. Currently, the genomic analysis of four ORFV strains from the Fujian province in southern China and a NA1/11 strain isolated from the Jilin province in northeast China have been reported. However, little is known about the genomic information of ORFV strains from central China. Results From a recent outbreak in a sheep herd in the Henan province of central China, a novel ORFV strain (HN3/12) was isolated and cultured in ovine fetal turbinate (OFTu) cells. The strain was identified as HN3/12 and verified by PCR based on the DNA sequences of 011 and 059 genes. The whole genomic sequence of this isolate was determined by Next Generation Sequencing technology. To determine the genetic characteristics of the HN3/12 strain, phylogenetic analysis of the 011 and 059 genes and amino acid sequence alignment of the HN3/12 strain were performed and compared with reference parapoxvirus strains. Conclusions The HN3/12 genome is 136,643 bp in length, contains 63.67% G + C and encodes 132 putative genes. Phylogenetic analysis of the 011 and 059 nucleotide sequences showed that this viral strain was similar to the NA1/11 isolate. The homology analysis indicates that HN3/12 has 93% to 98% identity with published ORFV strains at amino acid level. When open reading frames (ORFs) were aligned among the HN3/12 and four Fujian ORFV strains, most of them have identities greater than 90% and only a few less than 60%. The availability of the whole genomic sequence of HN3/12 aids in our understanding of, and provides new insights into, the genetic diversity of ORFV. Electronic supplementary material The online version of this article (doi:10.1186/s12917-017-1178-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Huiqin Chen
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Wei Li
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Zhenzhan Kuang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Daxiang Chen
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Xiaoqing Liao
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Ming Li
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Shuhong Luo
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People's Republic of China. .,Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, 5 Hebin Road, Foshan, Guangdong Province, 528000, People's Republic of China.
| | - Wenbo Hao
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People's Republic of China. .,Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
49
|
Zhao K, Li J, He W, Song D, Zhang X, Zhang D, Zhou Y, Gao F. Cyclophilin B facilitates the replication of Orf virus. Virol J 2017; 14:114. [PMID: 28619100 PMCID: PMC5471767 DOI: 10.1186/s12985-017-0781-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 06/06/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Viruses interact with host cellular factors to construct a more favourable environment for their efficient replication. Expression of cyclophilin B (CypB), a cellular peptidyl-prolyl cis-trans isomerase (PPIase), was found to be significantly up-regulated. Recently, a number of studies have shown that CypB is important in the replication of several viruses, including Japanese encephalitis virus (JEV), hepatitis C virus (HCV) and human papillomavirus type 16 (HPV 16). However, the function of cellular CypB in ORFV replication has not yet been explored. METHODS Suppression subtractive hybridization (SSH) technique was applied to identify genes differentially expressed in the ORFV-infected MDBK cells at an early phase of infection. Cellular CypB was confirmed to be significantly up-regulated by quantitative reverse transcription-PCR (qRT-PCR) analysis and Western blotting. The role of CypB in ORFV infection was further determined using Cyclosporin A (CsA) and RNA interference (RNAi). Effect of CypB gene silencing on ORFV replication by 50% tissue culture infectious dose (TCID50) assay and qRT-PCR detection. RESULTS In the present study, CypB was found to be significantly up-regulated in the ORFV-infected MDBK cells at an early phase of infection. Cyclosporin A (CsA) exhibited suppressive effects on ORFV replication through the inhibition of CypB. Silencing of CypB gene inhibited the replication of ORFV in MDBK cells. In conclusion, these data suggest that CypB is critical for the efficient replication of the ORFV genome. CONCLUSIONS Cellular CypB was confirmed to be significantly up-regulated in the ORFV-infected MDBK cells at an early phase of infection, which could effectively facilitate the replication of ORFV.
Collapse
Affiliation(s)
- Kui Zhao
- College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, China
| | - Jida Li
- College of Public Hygiene, ZunYi Medical University, 201 Dalian Road, Zunyi, 563003, China
| | - Wenqi He
- College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, China
| | - Deguang Song
- College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, China
| | - Ximu Zhang
- Laboratory Animal Center, Peking University, 5 Summer palace Road, Beijing, 100871, China
| | - Di Zhang
- College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, China
| | - Yanlong Zhou
- College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, China
| | - Feng Gao
- College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, China. .,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, China.
| |
Collapse
|
50
|
Bergqvist C, Kurban M, Abbas O. Orf virus infection. Rev Med Virol 2017; 27. [DOI: 10.1002/rmv.1932] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/17/2017] [Accepted: 03/13/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Christina Bergqvist
- Dermatology Department; American University of Beirut Medical Center; Beirut Lebanon
| | - Mazen Kurban
- Dermatology Department; American University of Beirut Medical Center; Beirut Lebanon
| | - Ossama Abbas
- Dermatology Department; American University of Beirut Medical Center; Beirut Lebanon
| |
Collapse
|