1
|
Al-Zebeeby A, Abbas AH, Alsaegh HA, Alaraji FS. The First Record of an Aggressive Form of Ocular Tumour Enhanced by Marek's Disease Virus Infection in Layer Flock in Al-Najaf, Iraq. Vet Med Int 2024; 2024:1793189. [PMID: 39376215 PMCID: PMC11458278 DOI: 10.1155/2024/1793189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/13/2024] [Indexed: 10/09/2024] Open
Abstract
Marek's disease (MD) is a highly infectious poultry illness with a tendency to form tumours in peripheral nerves and internal organs of affected birds. Tumours accompany MD, mostly caused by oncogenic Gallid alpha herpesvirus 2 (MD Herpes virus serotype I). Studies on avian tumours associated with MD infection are limited in Iraq. In the presented study, the positive samples of ocular tumour were 168 out of 282 MD positive samples, which accomplished in farm suffered from an unexpectedly high mortality rate. We investigated a rapidly developed tumour mass that was observed in an MD-vaccinated layer flock that showed obvious clinical signs of MD, accompanied by forming a small lump in one eye at age 21 weeks, which developed to a big lump at week 28 of age, leading to death. The diagnosis MD infection was confirmed by a Polymerase Chain Reaction (PCR) amplification of a specific region of the target gene meq of the causative agent, followed by Sanger sequencing and BLASTn search of the sequence against the NCBI nucleic acid database, resulted in Gallid alpha herpes virus 2 strain, and according to the phylogenetic analysis, the sequence from this study was uniquely clustered in its own branch in the tree. Histopathological examination of the ocular tumour core revealed aggregation of neoplastic cells and haemorrhage that replaced the normal eye tissue, as well as early tumour formation in internal organs such as the lung and liver. In addition, abnormal lesions are susceptible to tumours in the gizzard and spleen. To our knowledge, this is the first record of an aggressive MD virus infection-mediated ocular tumour in a layer flock in Al-Najaf province, Iraq.
Collapse
Affiliation(s)
- Aoula Al-Zebeeby
- Department of Pathology and Poultry DiseasesFaculty of Veterinary MedicineUniversity of Kufa, Al-Najaf Al-Ashraf, Kufa, Iraq
| | - Ali Hadi Abbas
- Department of Veterinary MicrobiologyFaculty of Veterinary MedicineUniversity of Kufa, Al-Najaf Al-Ashraf, Kufa, Iraq
| | - Haider Abas Alsaegh
- Department of Pathology and Poultry DiseasesFaculty of Veterinary MedicineUniversity of Kufa, Al-Najaf Al-Ashraf, Kufa, Iraq
| | - Furkan Sabbar Alaraji
- Department of Pathology and Poultry DiseasesFaculty of Veterinary MedicineUniversity of Kufa, Al-Najaf Al-Ashraf, Kufa, Iraq
| |
Collapse
|
2
|
Sabsabi MA, Kheimar A, You Y, von La Roche D, Härtle S, Göbel TW, von Heyl T, Schusser B, Kaufer BB. Unraveling the role of γδ T cells in the pathogenesis of an oncogenic avian herpesvirus. mBio 2024; 15:e0031524. [PMID: 38953352 PMCID: PMC11323538 DOI: 10.1128/mbio.00315-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/12/2024] [Indexed: 07/04/2024] Open
Abstract
Marek's disease virus (MDV) is an oncogenic alphaherpesvirus that causes deadly lymphomas in chickens. In chickens, up to 50% of all peripheral T cells are gamma delta (γδ) T cells. Until now, their role in MDV pathogenesis and tumor formation remains poorly understood. To investigate the role of γδ T cells in MDV pathogenesis, we infected recently generated γδ T cell knockout chickens with very virulent MDV. Strikingly, disease and tumor incidence were highly increased in the absence of γδ T cells, indicating that γδ T cells play an important role in the immune response against MDV. In the absence of γδ T cells, virus replication was drastically increased in the thymus and spleen, which are potential sites of T cell transformation. Taken together, our data provide the first evidence that γδ T cells play an important role in the pathogenesis and tumor formation of this highly oncogenic herpesvirus.IMPORTANCEGamma delta (γδ) T cells are the most abundant T cells in chickens, but their role in fighting pathogens remains poorly understood. Marek's disease virus (MDV) is an important veterinary pathogen, that causes one of the most frequent cancers in animals and is used as a model for virus-induced tumor formation. Our study revealed that γδ T cells play a crucial role in combating MDV, as disease and tumor incidence drastically increased in the absence of these cells. γδ T cells restricted virus replication in the key lymphoid organs, thereby decreasing the likelihood of causing tumors and disease. This study provides novel insights into the role of γδ T cells in the pathogenesis of this highly oncogenic virus.
Collapse
Affiliation(s)
| | - Ahmed Kheimar
- Institut für Virologie, Freie Universität Berlin, Berlin, Germany
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Sohag University, Sohag, Egypt
| | - Yu You
- Institut für Virologie, Freie Universität Berlin, Berlin, Germany
| | - Dominik von La Roche
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, München, Germany
| | - Sonja Härtle
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, München, Germany
| | - Thomas W. Göbel
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, München, Germany
| | - Theresa von Heyl
- Reproductive Biotechnology, TUM School of Life Sciences, Technische Universität München, München, Germany
| | - Benjamin Schusser
- Reproductive Biotechnology, TUM School of Life Sciences, Technische Universität München, München, Germany
- Center for Infection Prevention (ZIP), Technische Universität München, München, Germany
| | - Benedikt B. Kaufer
- Institut für Virologie, Freie Universität Berlin, Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
3
|
Zhu ZJ, Teng M, Liu Y, Chen FJ, Yao Y, Li EZ, Luo J. Immune escape of avian oncogenic Marek's disease herpesvirus and antagonistic host immune responses. NPJ Vaccines 2024; 9:109. [PMID: 38879650 PMCID: PMC11180173 DOI: 10.1038/s41541-024-00905-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 06/07/2024] [Indexed: 06/19/2024] Open
Abstract
Marek's disease virus (MDV) is a highly pathogenic and oncogenic alpha herpesvirus that causes Marek's disease (MD), which is one of the most important immunosuppressive and rapid-onset neoplastic diseases in poultry. The onset of MD lymphomas and other clinical diseases can be efficiently prevented by vaccination; these vaccines are heralded as the first demonstration of a successful vaccination strategy against a cancer. However, the persistent evolution of epidemic MDV strains towards greater virulence has recently resulted in frequent outbreaks of MD in vaccinated chicken flocks worldwide. Herein, we provide an overall review focusing on the discovery and identification of the strategies by which MDV evades host immunity and attacks the immune system. We have also highlighted the decrease in the immune efficacy of current MD vaccines. The prospects, strategies and new techniques for the development of efficient MD vaccines, together with the possibilities of antiviral therapy in MD, are also discussed.
Collapse
Affiliation(s)
- Zhi-Jian Zhu
- College of Biological and Food Engineering & Affiliated Central Hospital, Huanghuai University, Zhumadian, 463000, People's Republic of China
- Institute for Animal Health & UK-China Center of Excellence for Research on Avian Disease, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China
- Key Laboratory of Animal Immunology, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450002, People's Republic of China
| | - Man Teng
- Institute for Animal Health & UK-China Center of Excellence for Research on Avian Disease, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China
- Key Laboratory of Animal Immunology, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450002, People's Republic of China
| | - Yu Liu
- College of Biological and Food Engineering & Affiliated Central Hospital, Huanghuai University, Zhumadian, 463000, People's Republic of China
| | - Fu-Jia Chen
- College of Biological and Food Engineering & Affiliated Central Hospital, Huanghuai University, Zhumadian, 463000, People's Republic of China
| | - Yongxiu Yao
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Ash Road, Guildford, Surrey, GU24 0NF, UK
| | - En-Zhong Li
- College of Biological and Food Engineering & Affiliated Central Hospital, Huanghuai University, Zhumadian, 463000, People's Republic of China.
| | - Jun Luo
- Institute for Animal Health & UK-China Center of Excellence for Research on Avian Disease, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China.
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China.
- Key Laboratory of Animal Immunology, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450002, People's Republic of China.
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003, People's Republic of China.
- Longhu Laboratory, Zhengzhou, 450046, People's Republic of China.
| |
Collapse
|
4
|
Kim T, Hearn C, Heidari M. Efficacy of Recombinant Marek's Disease Virus Vaccine 301B/1 Expressing Membrane-Anchored Chicken Interleukin-15. Avian Dis 2024; 68:117-128. [PMID: 38885053 DOI: 10.1637/aviandiseases-d-23-00068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 05/25/2024] [Indexed: 06/20/2024]
Abstract
Cytokines are co-administrated with vaccines or co-expressed in the vaccine virus genome to improve protective efficacy by stimulating immune responses. Using glycosylphosphatidylinositol (GPI) anchoring by attachment to the target cytokine, we constructed recombinant Marek's disease virus (MDV) vaccine strain 301B/1 (v301B/1-rtg-IL-15) that expresses chicken interleukin-15 (IL-15) as the membrane-bound form at the cell surface. We evaluated the vaccine efficacy of v301B/1-rtg-IL-15 given as a bivalent Marek's disease (MD) vaccine in combination with turkey herpesvirus (HVT) against a very virulent plus MDV strain 648A challenge. The efficacy was compared with that of conventional bivalent MD vaccine, as a mixture with HVT plus parental v301B/1 or v301B/1-IL-15, which expresses a natural form of IL-15. The membrane-bound IL-15 expression did not interfere with the virus growth of recombinant v301B/1-rtg-IL-15. However, the MD incidence in birds vaccinated with v301B/1-rtg-IL-15 was higher than that of birds given the conventional bivalent MD vaccine containing parental v301B/1 virus, although the v301B/1-rtg-IL-15 vaccinated group showed increased natural killer cell activation at day 5 postvaccination, the same day as challenge. Overall, the protection of v301B/1-rtg-IL-15 was not improved from that of v301B/1 against very virulent plus MDV challenge.
Collapse
Affiliation(s)
- Taejoong Kim
- Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, United States Department of Agriculture, Athens, GA 30605,
| | - Cari Hearn
- Avian Diseases and Oncology Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, United States Department of Agriculture, East Lansing, MI 48823
| | - Mohammad Heidari
- Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, United States Department of Agriculture, Athens, GA 30605
| |
Collapse
|
5
|
Boodhoo N, Matsuyama-Kato A, Raj S, Fazel F, St-Denis M, Sharif S. Effect of Pre-Treatment with a Recombinant Chicken Interleukin-17A on Vaccine Induced Immunity against a Very Virulent Marek's Disease Virus. Viruses 2023; 15:1633. [PMID: 37631976 PMCID: PMC10459749 DOI: 10.3390/v15081633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
The host response to pathogenic microbes can lead to expression of interleukin (IL)-17, which has antimicrobial and anti-viral activity. However, relatively little is known about the basic biological role of chicken IL-17A against avian viruses, particularly against Marek's disease virus (MDV). We demonstrate that, following MDV infection, upregulation of IL-17A mRNA and an increase in the frequency of IL-17A+ T cells in the spleen occur compared to control chickens. To elaborate on the role of chIL-17A in MD, the full-length chIL-17A coding sequence was cloned into a pCDNA3.1-V5/HIS TOPO plasmid. The effect of treatment with pcDNA:chIL-17A plasmid in combination with a vaccine (HVT) and very virulent(vv)MDV challenge or vvMDV infection was assessed. In combination with HVT vaccination, chickens that were inoculated with the pcDNA:chIL-17A plasmid had reduced tumor incidence compared to chickens that received the empty vector control or that were vaccinated only (66.6% in the HVT + empty vector group and 73.33% in HVT group versus 53.3% in the HVT + pcDNA:chIL-17A). Further analysis demonstrated that the chickens that received the HVT vaccine and/or plasmid expressing IL-17A had lower MDV-Meq transcripts in the spleen. In conclusion, chIL-17A can influence the immunity conferred by HVT vaccination against MDV infection in chickens.
Collapse
Affiliation(s)
| | | | | | | | | | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (N.B.); (A.M.-K.); (S.R.); (F.F.); (M.S.-D.)
| |
Collapse
|
6
|
Bertzbach LD, Kohn M, You Y, Kossak L, Sabsabi MA, Kheimar A, Härtle S, Kaufer BB. In vitro infection of primary chicken lymphocytes with Marek's disease virus. STAR Protoc 2023; 4:102343. [PMID: 37270781 DOI: 10.1016/j.xpro.2023.102343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/25/2023] [Accepted: 05/10/2023] [Indexed: 06/06/2023] Open
Abstract
Marek's disease virus (MDV) is a highly oncogenic alphaherpesvirus that infects immune cells and causes a deadly lymphoproliferative disease in chickens. Cytokines and monoclonal antibodies promote the survival of chicken lymphocytes in vitro. Here, we describe protocols for the isolation, maintenance, and efficient MDV infection of primary chicken lymphocytes and lymphocyte cell lines. This facilitates the investigation of key aspects of the MDV life cycle in the primary target cells of viral replication, latency, genome integration, and reactivation. For complete details on the use and execution of this protocol, please refer to Schermuly et al.,1 Bertzbach et al. (2019),2 and You et al.3 For a comprehensive background on MDV, please see Osterrieder et al.4 and Bertzbach et al. (2020).5.
Collapse
Affiliation(s)
- Luca D Bertzbach
- Freie Universität Berlin, Institute of Virology, 14163 Berlin, Germany; Leibniz Institute of Virology (LIV), Department of Viral Transformation, 20251 Hamburg, Germany
| | - Marina Kohn
- Ludwig-Maximilians-Universität, Department for Veterinary Sciences, 82152 Planegg/Martinsried, Germany
| | - Yu You
- Freie Universität Berlin, Institute of Virology, 14163 Berlin, Germany
| | - Lisa Kossak
- Freie Universität Berlin, Institute of Virology, 14163 Berlin, Germany
| | | | - Ahmed Kheimar
- Freie Universität Berlin, Institute of Virology, 14163 Berlin, Germany; Sohag University, Faculty of Veterinary Medicine, Department of Poultry Diseases, 82524 Sohag, Egypt
| | - Sonja Härtle
- Ludwig-Maximilians-Universität, Department for Veterinary Sciences, 82152 Planegg/Martinsried, Germany.
| | - Benedikt B Kaufer
- Freie Universität Berlin, Institute of Virology, 14163 Berlin, Germany; Freie Universität Berlin, Veterinary Centre for Resistance Research (TZR), 14163 Berlin, Germany.
| |
Collapse
|
7
|
Role of T Cells in Vaccine-Mediated Immunity against Marek’s Disease. Viruses 2023; 15:v15030648. [PMID: 36992357 PMCID: PMC10055809 DOI: 10.3390/v15030648] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/04/2023] Open
Abstract
Marek’s disease virus (MDV), a highly cell-associated oncogenic α-herpesvirus, is the etiological agent of T cell lymphomas and neuropathic disease in chickens known as Marek’s disease (MD). Clinical signs of MD include neurological disorders, immunosuppression, and lymphoproliferative lymphomas in viscera, peripheral nerves, and skin. Although vaccination has greatly reduced the economic losses from MD, the molecular mechanism of vaccine-induced protection is largely unknown. To shed light on the possible role of T cells in immunity induced by vaccination, we vaccinated birds after the depletion of circulating T cells through the IP/IV injection of anti-chicken CD4 and CD8 monoclonal antibodies, and challenged them post-vaccination after the recovery of T cell populations post-treatment. There were no clinical signs or tumor development in vaccinated/challenged birds with depleted CD4+ or CD8+ T cells. The vaccinated birds with a combined depletion of CD4+ and CD8+ T cells, however, were severely emaciated, with atrophied spleens and bursas. These birds were also tumor-free at termination, with no virus particles detected in the collected tissues. Our data indicated that CD4+ and CD8+ T lymphocytes did not play a critical role in vaccine-mediated protection against MDV-induced tumor development.
Collapse
|
8
|
Matsuyama-Kato A, Shojadoost B, Boodhoo N, Raj S, Alizadeh M, Fazel F, Fletcher C, Zheng J, Gupta B, Abdul-Careem MF, Plattner BL, Behboudi S, Sharif S. Activated Chicken Gamma Delta T Cells Are Involved in Protective Immunity against Marek's Disease. Viruses 2023; 15:v15020285. [PMID: 36851499 PMCID: PMC9962238 DOI: 10.3390/v15020285] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Gamma delta (γδ) T cells play a significant role in the prevention of viral infection and tumor surveillance in mammals. Although the involvement of γδ T cells in Marek's disease virus (MDV) infection has been suggested, their detailed contribution to immunity against MDV or the progression of Marek's disease (MD) remains unknown. In the current study, T cell receptor (TCR)γδ-activated peripheral blood mononuclear cells (PBMCs) were infused into recipient chickens and their effects were examined in the context of tumor formation by MDV and immunity against MDV. We demonstrated that the adoptive transfer of TCRγδ-activated PBMCs reduced virus replication in the lungs and tumor incidence in MDV-challenged chickens. Infusion of TCRγδ-activated PBMCs induced IFN-γ-producing γδ T cells at 10 days post-infection (dpi), and degranulation activity in circulating γδ T cell and CD8α+ γδ T cells at 10 and 21 dpi in MDV-challenged chickens. Additionally, the upregulation of IFN-γ and granzyme A gene expression at 10 dpi was significant in the spleen of the TCRγδ-activated PBMCs-infused and MDV-challenged group compared to the control group. Taken together, our results revealed that TCRγδ stimulation promotes the effector function of chicken γδ T cells, and these effector γδ T cells may be involved in protection against MD.
Collapse
Affiliation(s)
- Ayumi Matsuyama-Kato
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Bahram Shojadoost
- Ceva Animal Health Inc., Research Park Centre, Guelph, ON N1G 4T2, Canada
| | - Nitish Boodhoo
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Sugandha Raj
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Mohammadali Alizadeh
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Fatemeh Fazel
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Charlotte Fletcher
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Jiayu Zheng
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Bhavya Gupta
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | | | - Brandon L. Plattner
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | | | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
- Correspondence: ; Tel.: +1-519-824-4120 (ext. 54641); Fax: +1-519-824-5930
| |
Collapse
|
9
|
Jiang B, Wang J, Cao M, Jin H, Liu W, Cheng J, Zhou L, Xu J, Li Y. Differential Replication and Cytokine Response between Vaccine and Very Virulent Marek's Disease Viruses in Spleens and Bursas during Latency and Reactivation. Viruses 2022; 15:6. [PMID: 36680047 PMCID: PMC9864003 DOI: 10.3390/v15010006] [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: 11/19/2022] [Revised: 12/13/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022] Open
Abstract
Marek's disease virus (MDV) infection results in Marek's disease (MD) in chickens, a lymphoproliferative and oncogenic deadly disease, leading to severe economic losses. The spleen and bursa are the most important lymphoid and major target organs for MDV replication. The immune response elicited by MDV replication in the spleen and bursa is critical for the formation of latent MDV infection and reactivation. However, the mechanism of the host immune response induced by MDV in these key lymphoid organs during the latent and reactivation infection phases is not well understood. In the study, we focused on the replication dynamics of a vaccine MDV strain MDV/CVI988 and a very virulent MDV strain MDV/RB1B in the spleen and bursa in the latent and reactivation infection phases (7-28 days post-inoculation [dpi]), as well as the expression of some previously characterized immune-related molecules. The results showed that the replication ability of MDV/RB1B was significantly stronger than that of MDV/CVI988 within 28 days post-infection, and the replication levels of both MDV strains in the spleen were significantly higher than those in the bursa. During the latent and reactivation phase of MDV infection (7-28 dpi), the transcriptional upregulation of chicken IL-1β, IL6, IL-8L1 IFN-γ and PML in the spleen and bursa induced by MDV/RB1B infection was overall stronger than that of MDV/CVI988. However, compared to MDV/RB1Binfection, MDV/CVI988 infection resulted in a more effective transcriptional activation of CCL4 in the latent infection phase (7-14 dpi), which may be a characteristic distinguishing MDV vaccine strain from the very virulent strain.
Collapse
Affiliation(s)
- Bo Jiang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
| | - Jing Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Mengyao Cao
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China
| | - Huan Jin
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
| | - Wenxiao Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
| | - Jing Cheng
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
| | - Linyi Zhou
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
| | - Jian Xu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
- Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China
| | - Yongqing Li
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
| |
Collapse
|
10
|
Vychodil T, Wight DJ, Nascimento M, Jolmes F, Korte T, Herrmann A, Kaufer BB. Visualization of Marek’s Disease Virus Genomes in Living Cells during Lytic Replication and Latency. Viruses 2022; 14:v14020287. [PMID: 35215880 PMCID: PMC8877148 DOI: 10.3390/v14020287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 11/18/2022] Open
Abstract
Visualization of the herpesvirus genomes during lytic replication and latency is mainly achieved by fluorescence in situ hybridization (FISH). Unfortunately, this technique cannot be used for the real-time detection of viral genome in living cells. To facilitate the visualization of the Marek’s disease virus (MDV) genome during all stages of the virus lifecycle, we took advantage of the well-established tetracycline operator/repressor (TetO/TetR) system. This system consists of a fluorescently labeled TetR (TetR-GFP) that specifically binds to an array of tetO sequences. This tetO repeat array was first inserted into the MDV genome (vTetO). Subsequently, we fused TetR-GFP via a P2a self-cleaving peptide to the C-terminus of the viral interleukin 8 (vIL8), which is expressed during lytic replication and latency. Upon reconstitution of this vTetO-TetR virus, fluorescently labeled replication compartments were detected in the nucleus during lytic replication. After validating the specificity of the observed signal, we used the system to visualize the genesis and mobility of the viral replication compartments. In addition, we assessed the infection of nuclei in syncytia as well as lytic replication and latency in T cells. Taken together, we established a system allowing us to track the MDV genome in living cells that can be applied to many other DNA viruses.
Collapse
Affiliation(s)
- Tereza Vychodil
- Institut für Virologie, Freie Universität Berlin, Robert von Ostertag-Straße 7-13, 14163 Berlin, Germany; (T.V.); (D.J.W.); (M.N.)
| | - Darren J. Wight
- Institut für Virologie, Freie Universität Berlin, Robert von Ostertag-Straße 7-13, 14163 Berlin, Germany; (T.V.); (D.J.W.); (M.N.)
| | - Mariana Nascimento
- Institut für Virologie, Freie Universität Berlin, Robert von Ostertag-Straße 7-13, 14163 Berlin, Germany; (T.V.); (D.J.W.); (M.N.)
| | - Fabian Jolmes
- Department of Biology, Molecular Biophysics, Humboldt-Universität zu Berlin, Invalidenstraße 42, 10115 Berlin, Germany; (F.J.); (T.K.); (A.H.)
| | - Thomas Korte
- Department of Biology, Molecular Biophysics, Humboldt-Universität zu Berlin, Invalidenstraße 42, 10115 Berlin, Germany; (F.J.); (T.K.); (A.H.)
| | - Andreas Herrmann
- Department of Biology, Molecular Biophysics, Humboldt-Universität zu Berlin, Invalidenstraße 42, 10115 Berlin, Germany; (F.J.); (T.K.); (A.H.)
- Institut für Chemie und Biochemie, Freie Universität Berlin, Altensteinstr. 23a, 14195 Berlin, Germany
| | - Benedikt B. Kaufer
- Institut für Virologie, Freie Universität Berlin, Robert von Ostertag-Straße 7-13, 14163 Berlin, Germany; (T.V.); (D.J.W.); (M.N.)
- Veterinary Centre for Resistance Research (TZR), Freie Universität Berlin, 14163 Berlin, Germany
- Correspondence: ; Tel.: +49-30-838-51936
| |
Collapse
|
11
|
You Y, Vychodil T, Aimola G, Previdelli RL, Göbel TW, Bertzbach LD, Kaufer BB. A Cell Culture System to Investigate Marek's Disease Virus Integration into Host Chromosomes. Microorganisms 2021; 9:microorganisms9122489. [PMID: 34946091 PMCID: PMC8706938 DOI: 10.3390/microorganisms9122489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 01/02/2023] Open
Abstract
Marek’s disease virus (MDV) is a highly oncogenic alphaherpesvirus that causes a devastating neoplastic disease in chickens. MDV has been shown to integrate its genome into the telomeres of latently infected and tumor cells, which is crucial for efficient tumor formation. Telomeric repeat arrays present at the ends of the MDV genome facilitate this integration into host telomeres; however, the integration mechanism remains poorly understood. Until now, MDV integration could only be investigated qualitatively upon infection of chickens. To shed further light on the integration mechanism, we established a quantitative integration assay using chicken T cell lines, the target cells for MDV latency and transformation. We optimized the infection conditions and assessed the establishment of latency in these T cells. The MDV genome was efficiently maintained over time, and integration was confirmed in these cells by fluorescence in situ hybridization (FISH). To assess the role of the two distinct viral telomeric repeat arrays in the integration process, we tested various knockout mutants in our in vitro integration assay. Efficient genome maintenance and integration was thereby dependent on the presence of the telomeric repeat arrays in the virus genome. Taken together, we developed and validated a novel in vitro integration assay that will shed light on the integration mechanism of this highly oncogenic virus into host telomeres.
Collapse
Affiliation(s)
- Yu You
- Institute of Virology, Freie Universität Berlin, 14163 Berlin, Germany; (Y.Y.); (T.V.); (G.A.); (R.L.P.)
| | - Tereza Vychodil
- Institute of Virology, Freie Universität Berlin, 14163 Berlin, Germany; (Y.Y.); (T.V.); (G.A.); (R.L.P.)
| | - Giulia Aimola
- Institute of Virology, Freie Universität Berlin, 14163 Berlin, Germany; (Y.Y.); (T.V.); (G.A.); (R.L.P.)
| | - Renato L. Previdelli
- Institute of Virology, Freie Universität Berlin, 14163 Berlin, Germany; (Y.Y.); (T.V.); (G.A.); (R.L.P.)
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | - Thomas W. Göbel
- Institute for Animal Physiology, Department of Veterinary Sciences, Ludwig Maximilian University Munich, 80539 Munich, Germany;
| | - Luca D. Bertzbach
- Institute of Virology, Freie Universität Berlin, 14163 Berlin, Germany; (Y.Y.); (T.V.); (G.A.); (R.L.P.)
- Department of Viral Transformation, Leibniz Institute for Experimental Virology (HPI), 20251 Hamburg, Germany
- Correspondence: (L.D.B.); (B.B.K.)
| | - Benedikt B. Kaufer
- Institute of Virology, Freie Universität Berlin, 14163 Berlin, Germany; (Y.Y.); (T.V.); (G.A.); (R.L.P.)
- Correspondence: (L.D.B.); (B.B.K.)
| |
Collapse
|
12
|
Ijaz A, Veldhuizen EJA, Broere F, Rutten VPMG, Jansen CA. The Interplay between Salmonella and Intestinal Innate Immune Cells in Chickens. Pathogens 2021; 10:1512. [PMID: 34832668 PMCID: PMC8618210 DOI: 10.3390/pathogens10111512] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/15/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022] Open
Abstract
Salmonellosis is a common infection in poultry, which results in huge economic losses in the poultry industry. At the same time, Salmonella infections are a threat to public health, since contaminated poultry products can lead to zoonotic infections. Antibiotics as feed additives have proven to be an effective prophylactic option to control Salmonella infections, but due to resistance issues in humans and animals, the use of antimicrobials in food animals has been banned in Europe. Hence, there is an urgent need to look for alternative strategies that can protect poultry against Salmonella infections. One such alternative could be to strengthen the innate immune system in young chickens in order to prevent early life infections. This can be achieved by administration of immune modulating molecules that target innate immune cells, for example via feed, or by in-ovo applications. We aimed to review the innate immune system in the chicken intestine; the main site of Salmonella entrance, and its responsiveness to Salmonella infection. Identifying the most important players in the innate immune response in the intestine is a first step in designing targeted approaches for immune modulation.
Collapse
Affiliation(s)
- Adil Ijaz
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands; (A.I.); (E.J.A.V.); (F.B.); (V.P.M.G.R.)
| | - Edwin J. A. Veldhuizen
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands; (A.I.); (E.J.A.V.); (F.B.); (V.P.M.G.R.)
| | - Femke Broere
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands; (A.I.); (E.J.A.V.); (F.B.); (V.P.M.G.R.)
| | - Victor P. M. G. Rutten
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands; (A.I.); (E.J.A.V.); (F.B.); (V.P.M.G.R.)
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, Pretoria 0110, South Africa
| | - Christine A. Jansen
- Cell Biology and Immunology Group, Department of Animal Sciences, Wageningen University & Research, De Elst 1, 6708 PB Wageningen, The Netherlands
| |
Collapse
|
13
|
Gao L, Zheng S, Wang Y. The Evasion of Antiviral Innate Immunity by Chicken DNA Viruses. Front Microbiol 2021; 12:771292. [PMID: 34777325 PMCID: PMC8581555 DOI: 10.3389/fmicb.2021.771292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/11/2021] [Indexed: 11/25/2022] Open
Abstract
The innate immune system constitutes the first line of host defense. Viruses have evolved multiple mechanisms to escape host immune surveillance, which has been explored extensively for human DNA viruses. There is growing evidence showing the interaction between avian DNA viruses and the host innate immune system. In this review, we will survey the present knowledge of chicken DNA viruses, then describe the functions of DNA sensors in avian innate immunity, and finally discuss recent progresses in chicken DNA virus evasion from host innate immune responses.
Collapse
Affiliation(s)
- Li Gao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shijun Zheng
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yongqiang Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
14
|
Marek's disease virus prolongs survival of primary chicken B-cells by inducing a senescence-like phenotype. PLoS Pathog 2021; 17:e1010006. [PMID: 34673841 PMCID: PMC8562793 DOI: 10.1371/journal.ppat.1010006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 11/02/2021] [Accepted: 10/04/2021] [Indexed: 12/17/2022] Open
Abstract
Marek’s disease virus (MDV) is an alphaherpesvirus that causes immunosuppression and deadly lymphoma in chickens. Lymphoid organs play a central role in MDV infection in animals. B-cells in the bursa of Fabricius facilitate high levels of MDV replication and contribute to dissemination at early stages of infection. Several studies investigated host responses in bursal tissue of MDV-infected chickens; however, the cellular responses specifically in bursal B-cells has never been investigated. We took advantage of our recently established in vitro infection system to decipher the cellular responses of bursal B-cells to infection with a very virulent MDV strain. Here, we demonstrate that MDV infection extends the survival of bursal B-cells in culture. Microarray analyses revealed that most cytokine/cytokine-receptor-, cell cycle- and apoptosis-associated genes are significantly down-regulated in these cells. Further functional assays validated these strong effects of MDV infections on cell cycle progression and thus, B-cell proliferation. In addition, we confirmed that MDV infections protect B-cells from apoptosis and trigger an accumulation of the autophagy marker Lc3-II. Taken together, our data indicate that MDV-infected bursal B-cells show hallmarks of a senescence-like phenotype, leading to a prolonged B-cell survival. This study provides an in-depth analysis of bursal B-cell responses to MDV infection and important insights into how the virus extends the survival of these cells. Upon MDV entry via the respiratory tract, B-cells are among the first cells to be infected in the lung and allow an efficient amplification of the virus. B-cells ensure the transmission of the virus to activated T-cells in which it replicates and ultimately transforms CD4-positive T-cells. Although playing a pivotal role in the MDV life cycle, the response of B-cells to MDV is currently not fully understood. Here, by using an in vitro infection model of primary bursal B-cells, we show that MDV infection leads to a prolonged B-cell survival resulting from decreased cell proliferation, protection from apoptosis and activation of autophagy. Our study provides new insights into the B-cell response to MDV infection, demonstrating that MDV triggers a senescence-like phenotype in B-cells that could potentiate their role in MDV pathogenesis.
Collapse
|
15
|
Maekawa D, Riblet SM, Whang P, Hurley DJ, Garcia M. Activation of Cytotoxic Lymphocytes and Presence of Regulatory T Cells in the Trachea of Non-Vaccinated and Vaccinated Chickens as a Recall to an Infectious Laryngotracheitis Virus (ILTV) Challenge. Vaccines (Basel) 2021; 9:865. [PMID: 34451989 PMCID: PMC8402403 DOI: 10.3390/vaccines9080865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/27/2021] [Accepted: 08/02/2021] [Indexed: 12/19/2022] Open
Abstract
While the protective efficacy of the infectious laryngotracheitis virus (ILTV) vaccines is well established, little is known about which components of the immune response are associated with effective resistance and vaccine protection. Early studies have pointed to the importance of the T cell-mediated immune responses. This study aimed to evaluate the activation of cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells and to quantify the presence of regulatory T cells (Tregs) in the larynx-trachea of chickens vaccinated with chicken embryo origin (CEO), tissue culture origin (TCO) and recombinant Herpesvirus of Turkey-laryngotracheitis (rHVT-LT) vaccines after challenge. Our results indicated that CEO vaccine protection was characterized by early CTLs and activated CTLs enhanced responses. TCO and rHVT-LT protection were associated with a moderate increase in resting and activated CTLs followed by an enhanced NK cell response. Tregs increase was only detected in the non-vaccinated challenged group, probably to support healing of the severe trachea epithelial damage. Taken together, our results revealed main differences in the cellular immune responses elicited by CEO, TCO, and rHVT-LT vaccination in the upper respiratory tract after challenge, and that activated CTLs rather than NK cells play a main role in vaccine protection.
Collapse
Affiliation(s)
- Daniel Maekawa
- Poultry Diagnostic and Research Center, Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (D.M.); (S.M.R.); (P.W.)
| | - Sylva M. Riblet
- Poultry Diagnostic and Research Center, Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (D.M.); (S.M.R.); (P.W.)
| | - Patrick Whang
- Poultry Diagnostic and Research Center, Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (D.M.); (S.M.R.); (P.W.)
| | - David J. Hurley
- Food Animal Health and Management Program, Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA;
| | - Maricarmen Garcia
- Poultry Diagnostic and Research Center, Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (D.M.); (S.M.R.); (P.W.)
| |
Collapse
|
16
|
The Marek's Disease Virus Unique Gene MDV082 Is Dispensable for Virus Replication but Contributes to a Rapid Disease Onset. J Virol 2021; 95:e0013121. [PMID: 34011541 DOI: 10.1128/jvi.00131-21] [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] [Indexed: 02/08/2023] Open
Abstract
Marek's disease virus (MDV) is a highly oncogenic alphaherpesvirus of chickens that causes lymphomas in various organs. Most MDV genes are conserved among herpesviruses, while others are unique to MDV and may contribute to pathogenesis and/or tumor formation. High transcript levels of the MDV-specific genes MDV082, RLORF11, and SORF6 were recently detected in lytically infected cells; however, it remained elusive if the respective proteins are expressed and if they play a role in MDV pathogenesis. In this study, we first addressed if these proteins are expressed by inserting FLAG tags at their N or C termini. We could demonstrate that among the three genes tested, MDV082 is the only gene that encodes a protein and is expressed very late in MDV plaques in vitro. To investigate the role of this novel MDV082 protein in MDV pathogenesis, we generated a recombinant virus that lacks expression of the MDV082 protein. Our data revealed that the MDV082 protein contributes to the rapid onset of Marek's disease but is not essential for virus replication, spread, and tumor formation. Taken together, this study sheds light on the expression of MDV-specific genes and unravels the role of the late protein MDV082 in MDV pathogenesis. IMPORTANCE MDV is a highly oncogenic alphaherpesvirus that causes Marek's disease in chickens. The virus causes immense economic losses in the poultry industry due to the high morbidity and mortality, but also the cost of the vaccination. MDV encodes over 100 genes that are involved in various processes of the viral life cycle. Functional characterization of MDV genes is an essential step toward understanding the complex virus life cycle and MDV pathogenesis. Here, we have identified a novel protein encoded by MDV082 and two potential noncoding RNAs (RLORF11 and SORF6). The novel MDV082 protein is not needed for efficient MDV replication and tumor formation. However, our data demonstrate that the MDV082 protein is involved in the rapid onset of Marek's disease.
Collapse
|
17
|
You Y, Hagag IT, Kheimar A, Bertzbach LD, Kaufer BB. Characterization of a Novel Viral Interleukin 8 (vIL-8) Splice Variant Encoded by Marek's Disease Virus. Microorganisms 2021; 9:microorganisms9071475. [PMID: 34361910 PMCID: PMC8303658 DOI: 10.3390/microorganisms9071475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 06/29/2021] [Accepted: 07/06/2021] [Indexed: 11/24/2022] Open
Abstract
Marek’s disease virus (MDV) is a highly cell-associated oncogenic alphaherpesvirus that causes lymphomas in various organs in chickens. Like other herpesviruses, MDV has a large and complex double-stranded DNA genome. A number of viral transcripts are generated by alternative splicing, a process that drastically extends the coding capacity of the MDV genome. One of the spliced genes encoded by MDV is the viral interleukin 8 (vIL-8), a CXC chemokine that facilitates the recruitment of MDV target cells and thereby plays an important role in MDV pathogenesis and tumorigenesis. We recently identified a novel vIL-8 exon (vIL-8-E3′) by RNA-seq; however, it remained elusive whether the protein containing the vIL-8-E3′ is expressed and what role it may play in MDV replication and/or pathogenesis. To address these questions, we first generated recombinant MDV harboring a tag that allows identification of the spliced vIL-8-E3′ protein, revealing that it is indeed expressed. We subsequently generated knockout viruses and could demonstrate that the vIL-8-E3′ protein is dispensable for MDV replication as well as secretion of the functional vIL-8 chemokine. Finally, infection of chickens with this vIL-8-E3′ knockout virus revealed that the protein is not important for MDV replication and pathogenesis in vivo. Taken together, our study provides novel insights into the splice forms of the CXC chemokine of this highly oncogenic alphaherpesvirus.
Collapse
Affiliation(s)
- Yu You
- Institute of Virology, Freie Universität Berlin, 14163 Berlin, Germany; (Y.Y.); (I.T.H.); (A.K.)
| | - Ibrahim T. Hagag
- Institute of Virology, Freie Universität Berlin, 14163 Berlin, Germany; (Y.Y.); (I.T.H.); (A.K.)
- Department of Virology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Ahmed Kheimar
- Institute of Virology, Freie Universität Berlin, 14163 Berlin, Germany; (Y.Y.); (I.T.H.); (A.K.)
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Sohag University, Sohag 82424, Egypt
| | - Luca D. Bertzbach
- Institute of Virology, Freie Universität Berlin, 14163 Berlin, Germany; (Y.Y.); (I.T.H.); (A.K.)
- Department of Viral Transformation, Leibniz Institute for Experimental Virology (HPI), 20251 Hamburg, Germany
- Correspondence: (L.D.B.); (B.B.K.)
| | - Benedikt B. Kaufer
- Institute of Virology, Freie Universität Berlin, 14163 Berlin, Germany; (Y.Y.); (I.T.H.); (A.K.)
- Correspondence: (L.D.B.); (B.B.K.)
| |
Collapse
|
18
|
Halabi S, Ghosh M, Stevanović S, Rammensee HG, Bertzbach LD, Kaufer BB, Moncrieffe MC, Kaspers B, Härtle S, Kaufman J. The dominantly expressed class II molecule from a resistant MHC haplotype presents only a few Marek's disease virus peptides by using an unprecedented binding motif. PLoS Biol 2021; 19:e3001057. [PMID: 33901176 PMCID: PMC8101999 DOI: 10.1371/journal.pbio.3001057] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 05/06/2021] [Accepted: 03/31/2021] [Indexed: 12/14/2022] Open
Abstract
Viral diseases pose major threats to humans and other animals, including the billions of chickens that are an important food source as well as a public health concern due to zoonotic pathogens. Unlike humans and other typical mammals, the major histocompatibility complex (MHC) of chickens can confer decisive resistance or susceptibility to many viral diseases. An iconic example is Marek's disease, caused by an oncogenic herpesvirus with over 100 genes. Classical MHC class I and class II molecules present antigenic peptides to T lymphocytes, and it has been hard to understand how such MHC molecules could be involved in susceptibility to Marek's disease, given the potential number of peptides from over 100 genes. We used a new in vitro infection system and immunopeptidomics to determine peptide motifs for the 2 class II molecules expressed by the MHC haplotype B2, which is known to confer resistance to Marek's disease. Surprisingly, we found that the vast majority of viral peptide epitopes presented by chicken class II molecules arise from only 4 viral genes, nearly all having the peptide motif for BL2*02, the dominantly expressed class II molecule in chickens. We expressed BL2*02 linked to several Marek's disease virus (MDV) peptides and determined one X-ray crystal structure, showing how a single small amino acid in the binding site causes a crinkle in the peptide, leading to a core binding peptide of 10 amino acids, compared to the 9 amino acids in all other reported class II molecules. The limited number of potential T cell epitopes from such a complex virus can explain the differential MHC-determined resistance to MDV, but raises questions of mechanism and opportunities for vaccine targets in this important food species, as well as providing a basis for understanding class II molecules in other species including humans.
Collapse
Affiliation(s)
- Samer Halabi
- University of Cambridge, Department of Pathology, Cambridge, United Kingdom
- University of Edinburgh, Institute for Immunology and Infection Research, Edinburgh, United Kingdom
| | - Michael Ghosh
- University of Tübingen, Department of Immunology, Institute of Cell Biology, Tübingen, Germany
| | - Stefan Stevanović
- University of Tübingen, Department of Immunology, Institute of Cell Biology, Tübingen, Germany
| | - Hans-Georg Rammensee
- University of Tübingen, Department of Immunology, Institute of Cell Biology, Tübingen, Germany
| | | | | | | | - Bernd Kaspers
- Ludwig Maximillians University, Veterinary Faculty, Planegg, Germany
| | - Sonja Härtle
- Ludwig Maximillians University, Veterinary Faculty, Planegg, Germany
| | - Jim Kaufman
- University of Cambridge, Department of Pathology, Cambridge, United Kingdom
- University of Edinburgh, Institute for Immunology and Infection Research, Edinburgh, United Kingdom
- University of Cambridge, Department of Veterinary Medicine, Cambridge, United Kingdom
| |
Collapse
|
19
|
Marek's Disease Virus Requires Both Copies of the Inverted Repeat Regions for Efficient In Vivo Replication and Pathogenesis. J Virol 2021; 95:JVI.01256-20. [PMID: 33115875 DOI: 10.1128/jvi.01256-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Marek's disease virus (MDV) is an oncogenic alphaherpesvirus of chickens. The MDV genome consists of two unique regions that are both flanked by inverted repeat regions. These repeats harbor several genes involved in virus replication and pathogenesis, but it remains unclear why MDV and other herpesviruses harbor these large sequence duplications. In this study, we set to determine if both copies of these repeat regions are required for MDV replication and pathogenesis. Our results demonstrate that MDV mutants lacking the entire internal repeat region (ΔIRLS) efficiently replicate and spread from cell-to-cell in vitro However, ΔIRLS replication was severely impaired in infected chickens and the virus caused significantly less frequent disease and tumors compared to the controls. In addition, we also generated recombinant viruses that harbor a deletion of most of the internal repeat region, leaving only short terminal sequences behind (ΔIRLS-HR). These remaining homologous sequences facilitated rapid restoration of the deleted repeat region, resulting in a virus that caused disease and tumors comparable to the wild type. Therefore, ΔIRLS-HR represents an excellent platform for rapid genetic manipulation of the virus genome in the repeat regions. Taken together, our study demonstrates that MDV requires both copies of the repeats for efficient replication and pathogenesis in its natural host.IMPORTANCE Marek's disease virus (MDV) is a highly oncogenic alphaherpesvirus that infects chickens and causes losses in the poultry industry of up to $2 billion per year. The virus is also widely used as a model to study alphaherpesvirus pathogenesis and virus-induced tumor development in a natural host. MDV and most other herpesviruses harbor direct or inverted repeats regions in their genome. However, the role of these sequence duplications in MDV remains elusive and has never been investigated in a natural virus-host model for any herpesvirus. Here, we demonstrate that both copies of the repeats are needed for efficient MDV replication and pathogenesis in vivo, while replication was not affected in cell culture. With this, we further dissect herpesvirus genome biology and the role of repeat regions in Marek's disease virus replication and pathogenesis.
Collapse
|
20
|
Role of microRNA and long non-coding RNA in Marek's disease tumorigenesis in chicken. Res Vet Sci 2021; 135:134-142. [PMID: 33485054 DOI: 10.1016/j.rvsc.2021.01.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/03/2021] [Accepted: 01/07/2021] [Indexed: 12/11/2022]
Abstract
Marek's disease virus (MDV), the causative agent of Marek's disease (MD), results in highly infectious phymatosis, lymphatic tissue hyperplasia, and neoplasia. MD is associated with high morbidity and mortality rate. Non-coding RNAs (ncRNAs) entails long non-coding RNA (lncRNA) and microRNA (miRNA). Numerous studies have reported that specific miRNAs and lncRNAs participate in multiple cellular processes, such as proliferation, migration, and tumor cell invasion. Specialized miRNAs and lncRNAs militate a similar role in MD tumor oncogenesis. Despite its growing popularity, only a few reviews are available on ncRNA in MDV tumor oncogenes. Herein, we summarized the role of the miRNAs and lncRNAs in MD tumorigenesis. Altogether, we brought forth the research issues, such as MD prevention, screening, regulatory network formation, novel miRNAs, and lncRNAs analysis in MD that needs to be explored further. This review provides a theoretical platform for the further analysis of miRNAs and lncRNAs functions and the prevention and control of MD and malignancies in domestic animals.
Collapse
|
21
|
Wild M, Kicuntod J, Seyler L, Wangen C, Bertzbach LD, Conradie AM, Kaufer BB, Wagner S, Michel D, Eickhoff J, Tsogoeva SB, Bäuerle T, Hahn F, Marschall M. Combinatorial Drug Treatments Reveal Promising Anticytomegaloviral Profiles for Clinically Relevant Pharmaceutical Kinase Inhibitors (PKIs). Int J Mol Sci 2021; 22:ijms22020575. [PMID: 33430060 PMCID: PMC7826512 DOI: 10.3390/ijms22020575] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/24/2020] [Accepted: 01/04/2021] [Indexed: 12/11/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a human pathogenic herpesvirus associated with a variety of clinical symptoms. Current antiviral therapy is not always effective, so that improved drug classes and drug-targeting strategies are needed. Particularly host-directed antivirals, including pharmaceutical kinase inhibitors (PKIs), may help to overcome problems of drug resistance. Here, we focused on utilizing a selection of clinically relevant PKIs and determined their anticytomegaloviral efficacies. Particularly, PKIs directed to host or viral cyclin-dependent kinases, i.e., abemaciclib, LDC4297 and maribavir, exerted promising profiles against human and murine cytomegaloviruses. The anti-HCMV in vitro activity of the approved anti-cancer drug abemaciclib was confirmed in vivo using our luciferase-based murine cytomegalovirus (MCMV) animal model in immunocompetent mice. To assess drug combinations, we applied the Bliss independence checkerboard and Loewe additivity fixed-dose assays in parallel. Results revealed that (i) both affirmative approaches provided valuable information on anti-CMV drug efficacies and interactions, (ii) the analyzed combinations comprised additive, synergistic or antagonistic drug interactions consistent with the drugs’ antiviral mode-of-action, (iii) the selected PKIs, especially LDC4297, showed promising inhibitory profiles, not only against HCMV but also other α-, β- and γ-herpesviruses, and specifically, (iv) the combination treatment with LDC4297 and maribavir revealed a strong synergism against HCMV, which might open doors towards novel clinical options in the near future. Taken together, this study highlights the potential of therapeutic drug combinations of current developmental/preclinical PKIs.
Collapse
Affiliation(s)
- Markus Wild
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054 Erlangen, Germany; (M.W.); (J.K.); (C.W.); (S.W.); (F.H.)
| | - Jintawee Kicuntod
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054 Erlangen, Germany; (M.W.); (J.K.); (C.W.); (S.W.); (F.H.)
| | - Lisa Seyler
- Institute of Radiology, University Medical Center Erlangen, FAU, Palmsanlage 5, 91054 Erlangen, Germany; (L.S.); (T.B.)
| | - Christina Wangen
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054 Erlangen, Germany; (M.W.); (J.K.); (C.W.); (S.W.); (F.H.)
| | - Luca D. Bertzbach
- Institute of Virology, Freie Universität Berlin, Robert-von-Ostertag-Straße 7-13, 14163 Berlin, Germany; (L.D.B.); (A.M.C.); (B.B.K.)
| | - Andelé M. Conradie
- Institute of Virology, Freie Universität Berlin, Robert-von-Ostertag-Straße 7-13, 14163 Berlin, Germany; (L.D.B.); (A.M.C.); (B.B.K.)
| | - Benedikt B. Kaufer
- Institute of Virology, Freie Universität Berlin, Robert-von-Ostertag-Straße 7-13, 14163 Berlin, Germany; (L.D.B.); (A.M.C.); (B.B.K.)
| | - Sabrina Wagner
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054 Erlangen, Germany; (M.W.); (J.K.); (C.W.); (S.W.); (F.H.)
| | - Detlef Michel
- Institute for Virology, Ulm University Medical Center, Albert-Einstein-Allee 11, 89081 Ulm, Germany;
| | - Jan Eickhoff
- Lead Discovery Center GmbH, Otto-Hahn-Str. 15, 44227 Dortmund, Germany;
| | - Svetlana B. Tsogoeva
- Institute of Organic Chemistry I, FAU, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany;
| | - Tobias Bäuerle
- Institute of Radiology, University Medical Center Erlangen, FAU, Palmsanlage 5, 91054 Erlangen, Germany; (L.S.); (T.B.)
| | - Friedrich Hahn
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054 Erlangen, Germany; (M.W.); (J.K.); (C.W.); (S.W.); (F.H.)
| | - Manfred Marschall
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054 Erlangen, Germany; (M.W.); (J.K.); (C.W.); (S.W.); (F.H.)
- Correspondence: ; Tel.: +49-9131-8526-089
| |
Collapse
|
22
|
Conradie AM, Bertzbach LD, Trimpert J, Patria JN, Murata S, Parcells MS, Kaufer BB. Distinct polymorphisms in a single herpesvirus gene are capable of enhancing virulence and mediating vaccinal resistance. PLoS Pathog 2020; 16:e1009104. [PMID: 33306739 PMCID: PMC7758048 DOI: 10.1371/journal.ppat.1009104] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 12/23/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022] Open
Abstract
Modified-live herpesvirus vaccines are widely used in humans and animals, but field strains can emerge that have a higher virulence and break vaccinal protection. Since the introduction of the first vaccine in the 1970s, Marek’s disease virus overcame the vaccine barrier by the acquisition of numerous genomic mutations. However, the evolutionary adaptations in the herpesvirus genome responsible for the vaccine breaks have remained elusive. Here, we demonstrate that point mutations in the multifunctional meq gene acquired during evolution can significantly alter virulence. Defined mutations found in highly virulent strains also allowed the virus to overcome innate cellular responses and vaccinal protection. Concomitantly, the adaptations in meq enhanced virus shedding into the environment, likely providing a selective advantage for the virus. Our study provides the first experimental evidence that few point mutations in a single herpesviral gene result in drastically increased virulence, enhanced shedding, and escape from vaccinal protection. Viruses can acquire mutations during evolution that alter their virulence. An example of a virus that has shown repeated shifts to higher virulence in response to more efficacious vaccines is the oncogenic Marek’s disease virus (MDV) that infects chickens. Until now, it remained unknown which mutations in the large virus genome are responsible for this increase in virulence. We could demonstrate that very few amino acid changes in the meq oncogene of MDV can significantly alter the virulence of the virus. In addition, these changes also allow the virus to overcome vaccinal protection and enhance the shedding into the environment. Taken together, our data provide fundamental insights into evolutionary changes that allow this deadly veterinary pathogen to evolve towards greater virulence.
Collapse
Affiliation(s)
| | | | - Jakob Trimpert
- Institut für Virologie, Freie Universität Berlin, Berlin, Germany
| | - Joseph N. Patria
- Department of Biological Sciences, University of Delaware, Newark, United States of America
| | - Shiro Murata
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Mark S. Parcells
- Department of Animal and Food Sciences, University of Delaware, Newark, United States of America
| | - Benedikt B. Kaufer
- Institut für Virologie, Freie Universität Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
23
|
Meijerink N, Kers JG, Velkers FC, van Haarlem DA, Lamot DM, de Oliveira JE, Smidt H, Stegeman JA, Rutten VPMG, Jansen CA. Early Life Inoculation With Adult-Derived Microbiota Accelerates Maturation of Intestinal Microbiota and Enhances NK Cell Activation in Broiler Chickens. Front Vet Sci 2020; 7:584561. [PMID: 33330708 PMCID: PMC7710667 DOI: 10.3389/fvets.2020.584561] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/26/2020] [Indexed: 01/06/2023] Open
Abstract
Studies in mammals, including chickens, have shown that the development of the immune system is affected by interactions with intestinal microbiota. Early life microbial colonization may affect the development of innate and adaptive immunity and may contribute to lasting effects on health and resilience of broiler chickens. We inoculated broiler chickens with adult-derived-microbiota (AM) to investigate their effects on intestinal microbiota composition and natural killer (NK) cells, amongst other immune cells. We hypothesized that AM inoculation directly upon hatch (day 0) would induce an alteration in microbiota composition shortly after hatch, and subsequently affect (subsets of) intestinal NK cells and their activation. Microbiota composition of caecal and ileal content of chickens of 1, 3, 7, 14, 21, and 35 days of age was assessed by sequencing of 16S ribosomal RNA gene amplicons. In parallel, subsets and activation of intestinal NK cells were analyzed by flow cytometry. In caecal content of 1- and 3-day-old AM chickens, a higher alpha-diversity (Faith's phylogenetic diversity) was observed compared to control chickens, whereas ileal microbiota were unaffected. Regarding beta-diversity, caecal microbiota profiles could be clustered into three distinct community types. Cluster A represented caecal microbiota of 1-day-old AM chickens and 1- and 3-day-old control chickens. Cluster B included microbiota of seven of eight 3- and 7-day-old AM and 7-day-old control chickens, and cluster C comprised microbiota of all chickens of 14-days and older, independent of inoculation. In 3-day-old AM chickens an increase in the percentages of intestinal IL-2Rα+NK cells and activated NK cells was observed compared to control chickens of the same age. In addition, an increase in relative numbers of intestinal cytotoxic CD8αα+T cells was observed in 14- and 21-day-old AM chickens. Taken together, these results indicate that early exposure to AM shapes and accelerates the maturation of caecal microbiota, which is paralleled by an increase in IL-2Rα+NK cells and enhanced NK cell activation. The observed association between early life development of intestinal microbiota and immune system indicates possibilities to apply microbiota-targeted strategies that can accelerate maturation of intestinal microbiota and strengthen the immune system, thereby improving the health and resilience of broiler chickens.
Collapse
Affiliation(s)
- Nathalie Meijerink
- Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Jannigje G. Kers
- Division Farm Animal Health, Department Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, Netherlands
| | - Francisca C. Velkers
- Division Farm Animal Health, Department Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Daphne A. van Haarlem
- Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - David M. Lamot
- Cargill Animal Nutrition and Health Innovation Center, Velddriel, Netherlands
| | | | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, Netherlands
| | - J. Arjan Stegeman
- Division Farm Animal Health, Department Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Victor P. M. G. Rutten
- Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - Christine A. Jansen
- Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
24
|
Coxiella burnetii-Infected NK Cells Release Infectious Bacteria by Degranulation. Infect Immun 2020; 88:IAI.00172-20. [PMID: 32817330 DOI: 10.1128/iai.00172-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 08/10/2020] [Indexed: 01/24/2023] Open
Abstract
Natural killer (NK) cells are critically involved in the early immune response against various intracellular pathogens, including Coxiella burnetii and Chlamydia psittaci Chlamydia-infected NK cells functionally mature, induce cellular immunity, and protect themselves by killing the bacteria in secreted granules. Here, we report that infected NK cells do not allow intracellular multiday growth of Coxiella, as is usually observed in other host cell types. C. burnetii-infected NK cells display maturation and gamma interferon (IFN-γ) secretion, as well as the release of Coxiella-containing lytic granules. Thus, NK cells possess a potent program to restrain and expel different types of invading bacteria via degranulation. Strikingly, though, in contrast to Chlamydia, expulsed Coxiella organisms largely retain their infectivity and, hence, escape the cell-autonomous self-defense mechanism in NK cells.
Collapse
|
25
|
Yang Y, Dong M, Hao X, Qin A, Shang S. Revisiting cellular immune response to oncogenic Marek's disease virus: the rising of avian T-cell immunity. Cell Mol Life Sci 2020; 77:3103-3116. [PMID: 32080753 PMCID: PMC7391395 DOI: 10.1007/s00018-020-03477-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/21/2022]
Abstract
Marek's disease virus (MDV) is a highly oncogenic alphaherpesvirus that causes deadly T-cell lymphomas and serves as a natural virus-induced tumor model in chickens. Although Marek's disease (MD) is well controlled by current vaccines, the evolution of MDV field viruses towards increasing virulence is concerning as a better vaccine to combat very virulent plus MDV is still lacking. Our understanding of molecular and cellular immunity to MDV and its immunopathogenesis has significantly improved, but those findings about cellular immunity to MDV are largely out-of-date, hampering the development of more effective vaccines against MD. T-cell-mediated cellular immunity was thought to be of paramount importance against MDV. However, MDV also infects macrophages, B cells and T cells, leading to immunosuppression and T-cell lymphoma. Additionally, there is limited information about how uninfected immune cells respond to MDV infection or vaccination, specifically, the mechanisms by which T cells are activated and recognize MDV antigens and how the function and properties of activated T cells correlate with immune protection against MDV or MD tumor. The current review revisits the roles of each immune cell subset and its effector mechanisms in the host immune response to MDV infection or vaccination from the point of view of comparative immunology. We particularly emphasize areas of research requiring further investigation and provide useful information for rational design and development of novel MDV vaccines.
Collapse
Affiliation(s)
- Yi Yang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Maoli Dong
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Xiaoli Hao
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Aijian Qin
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China.
- International Corporation Laboratory of Agriculture and Agricultural Products Safety, Yangzhou University, Yangzhou, 225009, China.
- Ministry of Education Key Laboratory for Avian Preventive Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.
- Key Laboratory of Jiangsu Preventive Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.
| | - Shaobin Shang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China.
- International Corporation Laboratory of Agriculture and Agricultural Products Safety, Yangzhou University, Yangzhou, 225009, China.
- Ministry of Education Key Laboratory for Avian Preventive Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
26
|
Hagag IT, Wight DJ, Bartsch D, Sid H, Jordan I, Bertzbach LD, Schusser B, Kaufer BB. Abrogation of Marek's disease virus replication using CRISPR/Cas9. Sci Rep 2020; 10:10919. [PMID: 32616820 PMCID: PMC7331644 DOI: 10.1038/s41598-020-67951-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
Marek's disease virus (MDV) is a highly cell-associated alphaherpesvirus that causes deadly lymphomas in chickens. While vaccination protects against clinical symptoms, MDV field strains can still circulate in vaccinated flocks and continuously evolve towards greater virulence. MDV vaccines do not provide sterilizing immunity, allowing the virus to overcome vaccine protection, and has increased the need for more potent vaccines or alternative interventions. In this study, we addressed if the CRISPR/Cas9 system can protect cells from MDV replication. We first screened a number of guide RNAs (gRNAs) targeting essential MDV genes for their ability to prevent virus replication. Single gRNAs significantly inhibited virus replication, but could result in the emergence of escape mutants. Strikingly, combining two or more gRNAs completely abrogated virus replication and no escape mutants were observed upon serial passaging. Our study provides the first proof-of-concept, demonstrating that the CRISPR/Cas9 system can be efficiently used to block MDV replication. The presented findings lay the foundation for future research to completely protect chickens from this deadly pathogen.
Collapse
Affiliation(s)
- Ibrahim T Hagag
- Institut für Virologie, Freie Universität Berlin, Robert-von-Ostertag-Str. 7-13, 14163, Berlin, Germany
- Department of Virology, Faculty of Veterinary Medicine, Zagazig University, El-Tagneed St. 114, Zagazig, 44511, Egypt
| | - Darren J Wight
- Institut für Virologie, Freie Universität Berlin, Robert-von-Ostertag-Str. 7-13, 14163, Berlin, Germany
| | - Denise Bartsch
- Reproductive Biotechnology, School of Life Sciences Weihenstephan, Technical University of Munich, Liesel-Beckmann-Str. 1, 85354, Freising, Germany
| | - Hicham Sid
- Reproductive Biotechnology, School of Life Sciences Weihenstephan, Technical University of Munich, Liesel-Beckmann-Str. 1, 85354, Freising, Germany
| | - Ingo Jordan
- ProBioGen AG, Herbert-Bayer-Straße 8, 13086, Berlin, Germany
| | - Luca D Bertzbach
- Institut für Virologie, Freie Universität Berlin, Robert-von-Ostertag-Str. 7-13, 14163, Berlin, Germany
| | - Benjamin Schusser
- Reproductive Biotechnology, School of Life Sciences Weihenstephan, Technical University of Munich, Liesel-Beckmann-Str. 1, 85354, Freising, Germany.
| | - Benedikt B Kaufer
- Institut für Virologie, Freie Universität Berlin, Robert-von-Ostertag-Str. 7-13, 14163, Berlin, Germany.
| |
Collapse
|
27
|
Wild M, Bertzbach LD, Tannig P, Wangen C, Müller R, Herrmann L, Fröhlich T, Tsogoeva SB, Kaufer BB, Marschall M, Hahn F. The trimeric artesunate derivative TF27 exerts strong anti-cytomegaloviral efficacy: Focus on prophylactic efficacy and oral treatment of immunocompetent mice. Antiviral Res 2020; 178:104788. [PMID: 32251769 DOI: 10.1016/j.antiviral.2020.104788] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/14/2020] [Accepted: 03/25/2020] [Indexed: 12/22/2022]
Abstract
Human cytomegalovirus (HCMV) causes serious and even life-threatening diseases, particularly upon congenital or post-transplant infection. Treatment of HCMV infections with currently available drugs targeting viral enzymes is often limited by severe side effects and the emergence of drug-resistant viruses. To avoid this problem, novel therapeutic options directed to host proteins involved in virus replication are being investigated. Recently, we described the pronounced antiherpesviral activity of the trimeric artesunate derivative TF27 at low nanomolar concentrations in vitro and in vivo. In the present study, we report first data on the prophylactic efficacy of TF27 against human and murine CMV and the oncogenic avian alphaherpesvirus Marek's disease virus (MDV). The main findings of this study are (i) a pronounced activity of the experimental drug TF27 against alpha- and betaherpesviruses in vitro upon prophylactic treatment and (ii) a therapeutic and prophylactic efficacy upon oral treatment in an immunocompetent mouse model. Moreover, our data highlight (iii) the tolerability of orally administered TF27 free of compound-associated adverse events and further confirm (iv) the suitability of cellular factors as primary antiviral targets. Thus, we provide evidence for therapeutic and prophylactic antiherpesviral efficacy of TF27 upon oral treatment in immunocompetent hosts and thereby underline its potential for future antiviral drug development.
Collapse
Affiliation(s)
- Markus Wild
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Luca D Bertzbach
- Institute of Virology, Freie Universität Berlin, Berlin, Germany.
| | - Pierre Tannig
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Christina Wangen
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Regina Müller
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Lars Herrmann
- Institute of Organic Chemistry I, FAU, Erlangen, Germany.
| | - Tony Fröhlich
- Institute of Organic Chemistry I, FAU, Erlangen, Germany.
| | | | | | - Manfred Marschall
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Friedrich Hahn
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| |
Collapse
|
28
|
Bertzbach LD, Conradie AM, You Y, Kaufer BB. Latest Insights into Marek's Disease Virus Pathogenesis and Tumorigenesis. Cancers (Basel) 2020; 12:cancers12030647. [PMID: 32164311 PMCID: PMC7139298 DOI: 10.3390/cancers12030647] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/06/2020] [Accepted: 03/07/2020] [Indexed: 12/14/2022] Open
Abstract
Marek’s disease virus (MDV) infects chickens and causes one of the most frequent cancers in animals. Over 100 years of research on this oncogenic alphaherpesvirus has led to a profound understanding of virus-induced tumor development. Live-attenuated vaccines against MDV were the first that prevented cancer and minimized the losses in the poultry industry. Even though the current gold standard vaccine efficiently protects against clinical disease, the virus continuously evolves towards higher virulence. Emerging field strains were able to overcome the protection provided by the previous two vaccine generations. Research over the last few years revealed important insights into the virus life cycle, cellular tropism, and tumor development that are summarized in this review. In addition, we discuss recent data on the MDV transcriptome, the constant evolution of this highly oncogenic virus towards higher virulence, and future perspectives in MDV research.
Collapse
|
29
|
Bertzbach LD, Harlin O, Härtle S, Fehler F, Vychodil T, Kaufer BB, Kaspers B. IFNα and IFNγ Impede Marek's Disease Progression. Viruses 2019; 11:v11121103. [PMID: 31795203 PMCID: PMC6950089 DOI: 10.3390/v11121103] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 12/18/2022] Open
Abstract
Marek’s disease virus (MDV) is an alphaherpesvirus that causes Marek’s disease, a malignant lymphoproliferative disease of domestic chickens. While MDV vaccines protect animals from clinical disease, they do not provide sterilizing immunity and allow field strains to circulate and evolve in vaccinated flocks. Therefore, there is a need for improved vaccines and for a better understanding of innate and adaptive immune responses against MDV infections. Interferons (IFNs) play important roles in the innate immune defenses against viruses and induce upregulation of a cellular antiviral state. In this report, we quantified the potent antiviral effect of IFNα and IFNγ against MDV infections in vitro. Moreover, we demonstrate that both cytokines can delay Marek’s disease onset and progression in vivo. Additionally, blocking of endogenous IFNα using a specific monoclonal antibody, in turn, accelerated disease. In summary, our data reveal the effects of IFNα and IFNγ on MDV infection and improve our understanding of innate immune responses against this oncogenic virus.
Collapse
Affiliation(s)
- Luca D. Bertzbach
- Institute of Virology, Freie Universität Berlin, 14163 Berlin, Germany; (L.D.B.); (T.V.)
| | - Olof Harlin
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, 80539 Munich, Germany; (O.H.); (S.H.)
| | - Sonja Härtle
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, 80539 Munich, Germany; (O.H.); (S.H.)
| | | | - Tereza Vychodil
- Institute of Virology, Freie Universität Berlin, 14163 Berlin, Germany; (L.D.B.); (T.V.)
| | - Benedikt B. Kaufer
- Institute of Virology, Freie Universität Berlin, 14163 Berlin, Germany; (L.D.B.); (T.V.)
- Correspondence: (B.B.K.); (B.K.)
| | - Bernd Kaspers
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, 80539 Munich, Germany; (O.H.); (S.H.)
- Correspondence: (B.B.K.); (B.K.)
| |
Collapse
|