1
|
Zhang L, Xie Q, Chang S, Ai Y, Dong K, Zhang H. Epigenetic Factor MicroRNAs Likely Mediate Vaccine Protection Efficacy against Lymphomas in Response to Tumor Virus Infection in Chickens through Target Gene Involved Signaling Pathways. Vet Sci 2024; 11:139. [PMID: 38668407 PMCID: PMC11053969 DOI: 10.3390/vetsci11040139] [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: 02/21/2024] [Revised: 03/16/2024] [Accepted: 03/20/2024] [Indexed: 04/29/2024] Open
Abstract
Epigenetic factors, including microRNAs (miRNAs), play an important role in affecting gene expression and, therefore, are involved in various biological processes including immunity protection against tumors. Marek's disease (MD) is a highly contagious disease of chickens caused by the MD virus (MDV). MD has been primarily controlled by vaccinations. MD vaccine efficacy might, in part, be dependent on modulations of a complex set of factors including host epigenetic factors. This study was designed to identify differentially expressed miRNAs in the primary lymphoid organ, bursae of Fabricius, in response to MD vaccination followed by MDV challenge in two genetically divergent inbred lines of White Leghorns. Small RNA sequencing and bioinformatic analyses of the small RNA sequence reads identified hundreds of miRNAs among all the treatment groups. A small portion of the identified miRNAs was differentially expressed within each of the four treatment groups, which were HVT or CVI988/Rispens vaccinated line 63-resistant birds and line 72-susceptible birds. A direct comparison between the resistant line 63 and susceptible line 72 groups vaccinated with HVT followed by MDV challenge identified five differentially expressed miRNAs. Gene Ontology analysis of the target genes of those five miRNAs revealed that those target genes, in addition to various GO terms, are involved in multiple signaling pathways including MAPK, TGF-β, ErbB, and EGFR1 signaling pathways. The general functions of those pathways reportedly play important roles in oncogenesis, anti-cancer immunity, cancer cell migration, and metastatic progression. Therefore, it is highly likely that those miRNAs may, in part, influence vaccine protection through the pathways.
Collapse
Affiliation(s)
- Lei Zhang
- U.S. Department of Agriculture, Agricultural Research Service, U.S. National Poultry Research Center, Athens, GA 30605, USA;
- Institute of Special Wild Economic Animal and Plant Science, Chinese Academy of Agricultural Sciences, Changchun 130112, China
| | - Qingmei Xie
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China;
| | - Shuang Chang
- College of Veterinary Medicine, Shandong Agricultural University, Tai’an 271018, China;
| | - Yongxing Ai
- College of Animal Science, Jilin University, Changchun 130062, China;
| | - Kunzhe Dong
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA 30912, USA;
| | - Huanmin Zhang
- U.S. Department of Agriculture, Agricultural Research Service, U.S. National Poultry Research Center, Athens, GA 30605, USA;
| |
Collapse
|
2
|
Abd El-Hack ME, El-Saadony MT, Elbestawy AR, El-Shall NA, Saad AM, Salem HM, El-Tahan AM, Khafaga AF, Taha AE, AbuQamar SF, El-Tarabily KA. Necrotic enteritis in broiler chickens: disease characteristics and prevention using organic antibiotic alternatives – a comprehensive review. Poult Sci 2022; 101:101590. [PMID: 34953377 PMCID: PMC8715378 DOI: 10.1016/j.psj.2021.101590] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/26/2021] [Accepted: 11/02/2021] [Indexed: 02/07/2023] Open
Abstract
In line with the substantial increase in the broiler industry worldwide, Clostridium perfringens-induced necrotic enteritis (NE) became a continuous challenge leading to high economic losses, especially after banning antimicrobial growth promoters in feeds by many countries. The disease is distributed worldwide in either clinical or subclinical form, causing a reduction in body weight or body weight gain and the feed conversion ratio, impairing the European Broiler Index or European Production Efficiency Factor. There are several predisposing factors in the development of NE. Clinical signs varied from inapparent signs in case of subclinical infection (clostridiosis) to obvious enteric signs (morbidity), followed by an increase in mortality level (clostridiosis or clinical infection). Clinical and laboratory diagnoses are based on case history, clinical signs, gross and histopathological lesions, pathogenic agent identification, serological testing, and molecular identification. Drinking water treatment is the most common route for the administration of several antibiotics, such as penicillin, bacitracin, and lincomycin. Strict hygienic management practices in the farm, careful selection of feed ingredients for ration formulation, and use of alternative antibiotic feed additives are all important in maintaining broiler efficiency and help increase the profitability of broiler production. The current review highlights NE caused by C. perfringens and explains the advances in the understanding of C. perfringens virulence factors involved in the pathogenesis of NE with special emphasis on the use of available antibiotic alternatives such as herbal extracts and essential oils as well as vaccines for the control and prevention of NE in broiler chickens.
Collapse
|
3
|
Avian Pattern Recognition Receptor Sensing and Signaling. Vet Sci 2020; 7:vetsci7010014. [PMID: 32012730 PMCID: PMC7157566 DOI: 10.3390/vetsci7010014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 01/16/2020] [Accepted: 01/23/2020] [Indexed: 02/07/2023] Open
Abstract
Pattern recognition receptors (PRRs) are a class of immune sensors that play a critical role in detecting and responding to several conserved patterns of microorganisms. As such, they play a major role in the maintenance of immune homeostasis and anti-microbial defense. Fundamental knowledge pertaining to the discovery of PRR functions and their ligands continue to advance the understanding of immune system and disease resistance, which led to the rational design and/or application of various PRR ligands as vaccine adjuvants. In addition, the conserved nature of many PRRs throughout the animal kingdom has enabled the utilization of the comparative genomics approach in PRR identification and the study of evolution, structural features, and functions in many animal species including avian. In the present review, we focused on PRR sensing and signaling functions in the avian species, domestic chicken, mallard, and domestic goose. In addition to summarizing recent advances in the understanding of avian PRR functions, the present review utilized a comparative biology approach to identify additional PRRs, whose functions have been well studied in mammalians but await functional characterization in avian.
Collapse
|
4
|
Hassanin O, Abdallah F, Mohamed MHA, Abdel Fattah DM. Influence of Marek's disease virus vaccines on chicken melanoma differentiation-associated gene 5-dependent-type I interferon signal transduction pathway with a highlight on their secondary impact on the immune responses post Newcastle disease virus vaccination. Vet Microbiol 2019; 235:248-256. [PMID: 31383309 DOI: 10.1016/j.vetmic.2019.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/04/2019] [Accepted: 07/07/2019] [Indexed: 12/24/2022]
Abstract
Marek's disease virus (MDV) leads to a lytic infection of B-lymphocytes in chickens, and also latently infects T-lymphocytes. Although Marek's disease vaccines have been widely in use, little is known about the innate immune response of this important livestock vaccine. In this study, we tested the effect of different commercially applied Marek's disease vaccines on the expression pattern of selected genes related to chicken interferon-alpha (chIFN-α) (melanoma differentiation associated gene 5 "MDA5″ dependent) signal transduction pathway. Both MDV serotype I (Rispens) and serotype III (Herpesvirus of turkey "HVT") vaccines could stimulate MDA5 dependent-type I interferon response as early as three days post vaccination in a dose-dependent manner. The stimulation continued up to 10 days in the instance of HVT vaccine and declined in the case of Rispens. Surprisingly, increasing the doses of the two vaccines led to dose-dependent down-regulation in the expression pattern of the investigated pathway, five and ten days post vaccination. Additionally, to shed the light on the consequent effect on the immune responses of the other viral vaccine, another experimental model based on Newcastle disease virus (NDV) vaccines was designed using HVT, HVT-VP2 and Rispens MDV vaccines. The three MDV vaccines were found to reduce chicken humoral immune response post NDV vaccination. However, only Rispens and HVT-VP2 had suppressive effects on the expression of MDA5-dependent-chIFN-α related cytokines. Consistent with this finding, the protection rate and NDV- humoral immune response post challenge with virulent NDV strain was lower in case of Rispens and HVT-VP2 vaccines.
Collapse
Affiliation(s)
- Ola Hassanin
- Department of Avian and Rabbit Medicine, Faculty of Veterinary Medicine, Zagazig University, 44511, Zagazig, Egypt.
| | - Fatma Abdallah
- Department of Virology, Faculty of Veterinary Medicine, Zagazig University, 44511, Zagazig, Egypt
| | - Mahmoud H A Mohamed
- Department of Avian and Rabbit Medicine, Faculty of Veterinary Medicine, Zagazig University, 44511, Zagazig, Egypt; Department of Clinical Studies, Collage of Veterinary, King Faisal University, Saudi Arabia
| | - Doaa M Abdel Fattah
- Department of Biochemistry, Faculty of Veterinary Medicine, Zagazig University, 44511, Zagazig, Egypt
| |
Collapse
|
5
|
Castaño-Ortiz JM, Jaspers VLB, Waugh CA. PFOS mediates immunomodulation in an avian cell line that can be mitigated via a virus infection. BMC Vet Res 2019; 15:214. [PMID: 31238913 PMCID: PMC6593586 DOI: 10.1186/s12917-019-1953-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 06/05/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFASs) are environmentally persistent and bioaccumulative chemicals. Immunomodulation is among the most concerning of toxic effects linked with PFAS exposure in mammalian models. However, no studies had yet shown this to be true in birds. Thus, we designed and conducted the first study to determine if PFASs could cause immunomodulation in birds. Secondly, we wanted to determine the effects on an avian host when exposed not only to immunomodulating chemicals, but also to a viral challenge. The aim, to determine if PFAS mediated immunmodulation functionally affects a pathogen challenge for a host. As innate immune system signalling pathways initiate crucial responses against a pathogen challenge, and are lesser studied than their adaptive counterparts, we focused on these pathways. To provide the first information on this, an in vitro experiment was designed and performed using chicken embryo fibroblasts exposed to perfluorooctane sulfonate (PFOS) (22 ppm) and immune markers characterised before and after being infected with gallid herpesvirus-2 (GaHV-2). RESULTS The expression of two pro-inflammatory cytokines, namely interleukin 8 (IL-8) and tumor necrosis factor alpha (TNF-α), the nuclear factor 'kappa-light-chain-enhancer' of activated B-cells (NF-κB), and the anti-inflammatory cytokine interleukin 4 (IL-4) were investigated in various scenarios. These results showed that exposure to PFOS decreased immune gene expression in chicken fibroblasts from 36 h post-exposure. Next, it was shown that this decrease could be mitigated by infection with gallid herpesvirus-2, which increased gene expression back to the baseline/control levels. CONCLUSIONS Not only is this the first study to provide the expected evidence that PFOS has immunomodulatory potential in birds, it also provides unexpected data that virus infections can mitigate this negative effect. Thereby, further research, including in vivo and in situ studies, on the impact of PFOS on host-virus interactions is now warranted, as it has been overlooked and might contribute to our understanding of recent disease outbreaks in wildlife. The mechanisms by which gallid herpesvirus mitigates immunomodulation were beyond the scope of this study, but are now of interest for future study.
Collapse
Affiliation(s)
- Jose M. Castaño-Ortiz
- Department of Biology, Norwegian University of Science and Technology (NTNU), Høgskoleringen 5, 7491 Trondheim, Norway
- Present address: Catalan Institute for Water Research (ICRA), C/Emili Grahit 101, 17003 Girona, Spain
| | - Veerle L. B. Jaspers
- Department of Biology, Norwegian University of Science and Technology (NTNU), Høgskoleringen 5, 7491 Trondheim, Norway
| | - Courtney A. Waugh
- Department of Biology, Norwegian University of Science and Technology (NTNU), Høgskoleringen 5, 7491 Trondheim, Norway
| |
Collapse
|
6
|
Gurung A, Kamble N, Kaufer BB, Pathan A, Behboudi S. Association of Marek's Disease induced immunosuppression with activation of a novel regulatory T cells in chickens. PLoS Pathog 2017; 13:e1006745. [PMID: 29267390 PMCID: PMC5739506 DOI: 10.1371/journal.ppat.1006745] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 11/11/2017] [Indexed: 12/15/2022] Open
Abstract
Marek's Disease Virus (MDV) is an alphaherpesvirus that infects chickens, transforms CD4+ T cells and causes deadly lymphomas. In addition, MDV induces immunosuppression early during infection by inducing cell death of the infected lymphocytes, and potentially due to activation of regulatory T (Treg)-cells. Furthermore, immunosuppression also occurs during the transformation phase of the disease; however, it is still unknown how the disease can suppress immune response prior or after lymphoma formation. Here, we demonstrated that chicken TGF-beta+ Treg cells are found in different lymphoid tissues, with the highest levels found in the gut-associated lymphoid tissue (cecal tonsil: CT), fostering an immune-privileged microenvironment exerted by TGF-beta. Surprisingly, significantly higher frequencies of TGF-beta+ Treg cells are found in the spleens of MDV-susceptible chicken lines compared to the resistant line, suggesting an association between TGF-beta+ Treg cells and host susceptibility to lymphoma formation. Experimental infection with a virulent MDV elevated the levels of TGF-beta+ Treg cells in the lungs as early as 4 days post infection, and during the transformation phase of the disease in the spleens. In contrast to TGF-beta+ Treg cells, the levels of CD4+CD25+ T cells remained unchanged during the infection and transformation phase of the disease. Furthermore, our results demonstrate that the induction of TGF-beta+ Treg cells is associated with pathogenesis of the disease, as the vaccine strain of MDV did not induce TGF-beta+ Treg cells. Similar to human haematopoietic malignant cells, MDV-induced lymphoma cells expressed high levels of TGF-beta but very low levels of TGF-beta receptor I and II genes. The results confirm that COX-2/ PGE2 pathway is involved in immunosuppression induced by MDV-lymphoma cells. Taken together, our results revealed a novel TGF-beta+ Treg subset in chickens that is activated during MDV infection and tumour formation.
Collapse
Affiliation(s)
- Angila Gurung
- The Pirbright Institute, Ash Road, Woking, United Kingdom
- College of Health and Life Sciences, Department of Life Sciences, Brunel University, London, United Kingdom
| | - Nitin Kamble
- The Pirbright Institute, Ash Road, Woking, United Kingdom
| | - Benedikt B. Kaufer
- Institut für Virologie, Freie Universität Berlin, Robert-von-Ostertag-Straße, Berlin, Germany
| | - Ansar Pathan
- College of Health and Life Sciences, Department of Life Sciences, Brunel University, London, United Kingdom
| | - Shahriar Behboudi
- The Pirbright Institute, Ash Road, Woking, United Kingdom
- Department of Pathology and Infectious Disease, School of Veterinary Medicine, University of Surrey, Guildford, United Kingdom
- * E-mail:
| |
Collapse
|
7
|
Teague K, Graham L, Dunn J, Cheng H, Anthony N, Latorre J, Menconi A, Wolfenden R, Wolfenden A, Mahaffey B, Baxter M, Hernandez-Velasco X, Merino-Guzman R, Bielke L, Hargis B, Tellez G. In ovo evaluation of FloraMax®-B11 on Marek’s disease HVT vaccine protective efficacy, hatchability, microbiota composition, morphometric analysis, and Salmonella enteritidis infection in broiler chickens. Poult Sci 2017; 96:2074-2082. [DOI: 10.3382/ps/pew494] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 12/13/2016] [Indexed: 12/19/2022] Open
|
8
|
Dong K, Chang S, Xie Q, Black-Pyrkosz A, Zhang H. Comparative transcriptomics of genetically divergent lines of chickens in response to Marek's disease virus challenge at cytolytic phase. PLoS One 2017; 12:e0178923. [PMID: 28591220 PMCID: PMC5462384 DOI: 10.1371/journal.pone.0178923] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 05/22/2017] [Indexed: 11/30/2022] Open
Abstract
Marek's disease (MD), caused by Marek's disease virus (MDV), remains an economically significant threat to the poultry industry worldwide. Genetic resistance to MD is a promising alternative strategy to augment current control measures (vaccination and management). However, only a few functional genes reportedly conferring MD resistance have been identified. Here, we performed a comparative transcriptomics analysis of two highly inbred yet genetically divergent lines of chickens (line 63 and 72) that are resistant and susceptible to MD, respectively, in response to a very virulent plus strain of MDV (vv+MDV) challenge at cytolytic phase. A total of 203 DEGs in response to MDV challenge were identified in the two lines. Of these, 96 DEGs were in common for both lines, in addition to 36 and 71 DEGs that were specific for line 63 and 72, respectively. Functional enrichment analysis results showed the DEGs were significantly enriched in GO terms and pathways associated with immune response. Especially, the four DEGs, FGA, ALB, FN1, and F13A1 that reportedly facilitate virus invasion or immunosuppression, were found to be significantly up-regulated in the susceptible line 72 but down-regulated in the resistant line 63 birds. These results provide new resources for future studies to further elucidate the genetic mechanism conferring MD resistance.
Collapse
Affiliation(s)
- Kunzhe Dong
- USDA, Agricultural Research Service, Avian Disease and Oncology Laboratory, East Lansing, Michigan, United States of America
- ORISE Fellow, USDA, Agriculture Research Service, Avian Disease and Oncology Laboratory, East Lansing, Michigan, United States of America
| | - Shuang Chang
- College of Veterinary Medicine, Shandong Agricultural University, Tai’an, Shandong, China
| | - Qingmei Xie
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Alexis Black-Pyrkosz
- USDA, Agricultural Research Service, Avian Disease and Oncology Laboratory, East Lansing, Michigan, United States of America
| | - Huanmin Zhang
- USDA, Agricultural Research Service, Avian Disease and Oncology Laboratory, East Lansing, Michigan, United States of America
| |
Collapse
|
9
|
Expression patterns of endogenous avian retrovirus ALVE1 and its response to infection with exogenous avian tumour viruses. Arch Virol 2016; 162:89-101. [PMID: 27686071 DOI: 10.1007/s00705-016-3086-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Accepted: 09/21/2016] [Indexed: 02/01/2023]
Abstract
Endogenous retroviruses (ERVs) are genomic elements that are present in a wide range of vertebrates and have been implicated in a variety of human diseases, including cancer. However, the characteristic expression patterns of ERVs, particularly in virus-induced tumours, is not fully clear. DNA methylation was analysed by bisulfite pyrosequencing, and gene expression was analysed by RT-qPCR. In this study, we first found that the endogenous avian retrovirus ALVE1 was highly expressed in some chicken tissues (including the heart, bursa, thymus, and spleen) at 2 days of age, but its expression was markedly decreased at 35 days of age. In contrast, the CpG methylation level of ALVE1 was significantly lower in heart and bursa at 2 days than at 35 days of age. Moreover, we found that the expression of ALVE1 was significantly inhibited in chicken embryo fibroblast cells (CEFs) and MSB1 cells infected with avian leukosis virus subgroup J (ALVJ) and reticuloendotheliosis virus (REV) at the early stages of infection. In contrast, the expression of the ALVE1 env gene was significantly induced in CEFs and MSB1 cells infected with Marek's disease virus (MDV). However, the methylation and expression levels of the ALVE1 long terminal repeat (LTR) did not show obvious alterations in response to viral infection. The present study revealed the expression patterns of ALVE1 in a variety of chicken organs and tissues and in chicken cells in response to avian tumour virus infection. These findings may be of significance for understanding the role and function of ERVs that are present in the host genome.
Collapse
|
10
|
Reddy VRAP, Trus I, Desmarets LMB, Li Y, Theuns S, Nauwynck HJ. Productive replication of nephropathogenic infectious bronchitis virus in peripheral blood monocytic cells, a strategy for viral dissemination and kidney infection in chickens. Vet Res 2016; 47:70. [PMID: 27412035 PMCID: PMC4944500 DOI: 10.1186/s13567-016-0354-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/23/2016] [Indexed: 12/21/2022] Open
Abstract
In the present study, the replication kinetics of nephropathogenic (B1648) and respiratory (Massachusetts-M41) IBV strains were compared in vitro in respiratory mucosa explants and blood monocytes (KUL01+ cells), and in vivo in chickens to understand why some IBV strains have a kidney tropism. B1648 was replicating somewhat better than M41 in the epithelium of the respiratory mucosa explants and used more KUL01+ cells to penetrate the deeper layers of the respiratory tract. B1648 was productively replicating in KUL01+ monocytic cells in contrast with M41. In B1648 inoculated animals, 102.7–6.8 viral RNA copies/100 mg were detected in tracheal secretions at 2, 4, 6, 8, 10 and 12 days post inoculation (dpi), 102.4–4.5 viral RNA copies/mL in plasma at 2, 4, 6, 8, 10 and 12 dpi and 101.8–4.4 viral RNA copies/106 mononuclear cells in blood at 2, 4, 6 and 8 dpi. In M41 inoculated animals, 102.6–7.0 viral RNA copies/100 mg were detected in tracheal secretions at 2, 4, 6, 8 and 10 dpi, but viral RNA was not demonstrated in plasma and mononuclear cells (except in one chicken at 6 dpi). Infectious virus was detected only in plasma and mononuclear cells of the B1648 group. At euthanasia (12 dpi), viral RNA and antigen positive cells were detected in lungs, liver, spleen and kidneys of only the B1648 group and in tracheas of both the B1648 and M41 group. In conclusion, only B1648 can easily disseminate to internal organs via a cell-free and -associated viremia with KUL01+ cells as important carrier cells.
Collapse
Affiliation(s)
- Vishwanatha R A P Reddy
- Laboratory of Virology, Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium.
| | - Ivan Trus
- Laboratory of Virology, Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Lowiese M B Desmarets
- Laboratory of Virology, Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Yewei Li
- Laboratory of Virology, Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Sebastiaan Theuns
- Laboratory of Virology, Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Hans J Nauwynck
- Laboratory of Virology, Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| |
Collapse
|
11
|
Parvizi P, Brisbin JT, Read LR, Sharif S. Cytokine Gene Expression in Lung Mononuclear Cells of Chickens Vaccinated with Herpesvirus of Turkeys and Infected with Marek's Disease Virus. Viral Immunol 2015; 28:538-43. [DOI: 10.1089/vim.2015.0054] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Payvand Parvizi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Canada
| | - Jennifer T. Brisbin
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Canada
| | - Leah R. Read
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Canada
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Canada
| |
Collapse
|
12
|
Rong S, Wheeler D, Weber F. Efficient Marek's disease virus (MDV) and herpesvirus of turkey infection of the QM7 cell line that does not contain latent MDV genome. Avian Pathol 2015; 43:414-9. [PMID: 25204414 DOI: 10.1080/03079457.2014.956687] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Marek's disease virus (MDV; also known as Gallid herpesvirus 2, MDV-1) causes oncogenic disease in chickens producing clinical signs that include lymphomas, visceral tumours, nerve lesions, and immunosuppression. MDV vaccines are widely used and mostly produced using primary cells: chicken embryo fibroblast cells, duck embryo fibroblast cells, chicken embryo kidney cells or chicken kidney cells. An immortalized cell line that can be used to manufacture the virus has long been desired. In this report, we demonstrate that QM7 cells were susceptible to infection with either MDV or herpesvirus of turkey (HVT; also known as Meleagrid herpesvirus 1, MDV-3). Polymerase chain reaction analysis with primers amplifying selected MDV genes revealed that QM7 cells did not contain these sequences. However, MDV genes were detected in QT35 cells, which have been reported to harbour latent MDV virus. Transfection of naked MDV DNA initiated efficient infection of QM7 cells. In addition, QM7 cell lysate, clarified supernatant, and QM7 cell pellet infected with MDV were negative for reverse transcriptase activity, indicating absence of endogenous retrovirus. QM7 cells were also found to be free of other avian pathogens in a chick embryo inoculation test. In vivo studies of MDV growing in QM7 cells showed the virus retained its pathogenicity and virulence. In ovo experiments demonstrated that both HVT and MDV propagated in QM7 cells did not interfere with hatchability of injected eggs, and viruses could be re-isolated from hatched chicks. The results suggest that QM7 could be a good host cell line for growing both MDV and HVT.
Collapse
Affiliation(s)
- Sing Rong
- a Global Biologics Research, Veterinary Medicine Research & Development, Zoetis, Kalamazoo , Michigan , USA
| | | | | |
Collapse
|
13
|
Nii T, Isobe N, Yoshimura Y. The effect of estrogen on the early cytotoxic response to IB virus infection in hen oviduct. Vet Immunol Immunopathol 2015; 164:56-66. [PMID: 25593044 DOI: 10.1016/j.vetimm.2014.12.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 12/25/2014] [Accepted: 12/29/2014] [Indexed: 01/10/2023]
Abstract
The aim of this study was to determine whether the egg-laying phase and estrogen affect the induction of cytotoxic cells in response to avian infectious bronchitis (IB) virus at early stage of infection in the oviduct. Attenuated IB virus (aIBV group) or its vehicle (control group) was introduced to the oviductal magnum lumen of White Leghorn hens in the laying and molting phase, as well as molting hens injected with estradiol benzoate (M-EB hens) or corn oil (M-oil hens). Oviductal isthmus and uterus were collected 24h after injection. The frequency of CD8(+) and TCRγδ(+) T cells expression was examined by immunohistochemistry, followed by image analysis. The expression of the genes of toll-like receptor 7 (TLR7), natural killer cell receptor (BNK), cytotoxic substances (granzyme, perforin), and cytokines (CXCL12, CX3CL1, and IFNγ) were examined by real-time polymerase chain reaction analysis. The frequency of CD8(+) and TCRγδ(+) T cells in the isthmus, and CD8(+) cells in the uterus was significantly higher in the aIBV group compared to the control group of laying and M-EB hens. The expression of all the genes examined in this study in the isthmus, and CX3CL1 and IFNγ expression in the uterus was significantly higher in the aIBV group in the laying and M-EB hens. These results suggested that infection with IB virus causes an immune response involving the influx of cytotoxic cells and upregulation of cytokines in the isthmus and uterus at early stage of infection. This response was stronger during the laying phase compared to the molting phase, probably due to the effect of estrogen.
Collapse
Affiliation(s)
- Takahiro Nii
- Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, 739-8528, Japan
| | - Naoki Isobe
- Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, 739-8528, Japan; Research Center for Animal Science, Hiroshima University, Higashi-Hiroshima, 739-8528, Japan
| | - Yukinori Yoshimura
- Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, 739-8528, Japan; Research Center for Animal Science, Hiroshima University, Higashi-Hiroshima, 739-8528, Japan.
| |
Collapse
|
14
|
Serological Evaluation of Turkey Herpesvirus Vector Vaccines Expressing the Hemagglutinin Gene of Avian Influenza Virus H5 Subtype under Three Different Promoters. J Poult Sci 2015. [DOI: 10.2141/jpsa.0140096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
15
|
Chi JQ, Teng M, Yu ZH, Xu H, Su JW, Zhao P, Xing GX, Liang HD, Deng RG, Qu LH, Zhang GP, Luo J. Marek's disease virus-encoded analog of microRNA-155 activates the oncogene c-Myc by targeting LTBP1 and suppressing the TGF-β signaling pathway. Virology 2014; 476:72-84. [PMID: 25528440 DOI: 10.1016/j.virol.2014.11.027] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 10/10/2014] [Accepted: 11/24/2014] [Indexed: 12/22/2022]
Abstract
Marek's disease virus (MDV) is a representative alpha herpes virus able to induce rapid-onset T-cell lymphoma in its natural host and regarded as an ideal model for the study of virus-induced tumorigenesis. Recent studies have shown that the mdv1-miR-M4-5p, a viral analog of cellular miR-155, is critical for MDV׳s oncogenicity. However, the precise mechanism whereby it was involved in MD lymphomagenesis remained unknown. We have presently identified the host mRNA targets of mdv1-miR-M4-5 and identified the latent TGF-β binding protein 1 (LTBP1) as a critical target for it. We found that during MDV infection, down-regulation of LTBP1 expression by mdv1-miR-M4-5p led to a significant decrease of the secretion and activation of TGF-β1, with suppression of TGF-β signaling and a significant activation of expression of c-Myc, a well-known oncogene which is critical for virus-induced tumorigenesis. Our findings reveal a novel and important mechanism of how mdv1-miR-M4-5p potentially contributes to MDV-induced tumorigenesis.
Collapse
Affiliation(s)
- Jia-Qi Chi
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, People׳s Republic of China; Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, People׳s Republic of China; College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, People׳s Republic of China
| | - Man Teng
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, People׳s Republic of China
| | - Zu-Hua Yu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, People׳s Republic of China; Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, People׳s Republic of China
| | - Hui Xu
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-Sen (Zhongshan) University, Guangzhou 510275, People׳s Republic of China
| | - Jing-Wei Su
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, People׳s Republic of China; College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, People׳s Republic of China
| | - Pu Zhao
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, People׳s Republic of China; College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, People׳s Republic of China
| | - Guang-Xu Xing
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, People׳s Republic of China
| | - Hong-De Liang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, People׳s Republic of China
| | - Rui-Guang Deng
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, People׳s Republic of China
| | - Liang-Hu Qu
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-Sen (Zhongshan) University, Guangzhou 510275, People׳s Republic of China
| | - Gai-Ping Zhang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, People׳s Republic of China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, People׳s Republic of China.
| | - Jun Luo
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, People׳s Republic of China.
| |
Collapse
|
16
|
Nakamura S, Ochiai K, Abe A, Kishi S, Takayama K, Sunden Y. Astrocytic growth through the autocrine/paracrine production of IL-1β in the early infectious phase of fowl glioma-inducing virus. Avian Pathol 2014; 43:437-42. [PMID: 25117822 DOI: 10.1080/03079457.2014.952621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Fowl glioma is characterized morphologically by multiple nodular astrocytic growth with disseminated non-suppurative encephalitis. The disease is caused by fowl glioma-inducing virus (FGV) and its variants, belonging to subgroup A of avian leukosis virus (ALV-A). Fifty-seven FGV variants have so far been isolated from Japanese fowls and these variants have a variable degree of glioma inducibility. However, how these ALVs induce glioma with different degrees and frequencies has not been fully elucidated. In this study, we investigated the relationship between intracerebral viral replication and astrocytic growth in the early infectious phase. Replication abilities of two ALV strains, Sp-53 (a FGV variant) and ALV-based replication-competent vector RCAS(A) without glioma inducibility, were compared in the brains of C/O specific pathogen free chickens at 35 days of age. Sp-53 replicated faster than RCAS(A), and the histological score and the level of interleukin (IL)-1β in brains increased depending on the level of intracerebral viral RNA. Up-regulation of IL-1β was also demonstrated in primary cultured astrocytes. These results suggest that the astrocytic growth in this phase is enhanced through the autocrine/paracrine production of IL-1β in the FGV-infected astrocytes.
Collapse
Affiliation(s)
- Sayuri Nakamura
- a Laboratory of Comparative Pathology, Graduate School of Veterinary Medicine , Hokkaido University , Sapporo , Japan
| | | | | | | | | | | |
Collapse
|
17
|
Marek's disease virus may interfere with T cell immunity by TLR3 signals. Vet Res Commun 2014; 38:149-56. [PMID: 24585377 DOI: 10.1007/s11259-014-9598-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2014] [Indexed: 10/25/2022]
Abstract
Marek's disease virus (MDV) is a highly oncogenic alpha-herpesvirus that causes T cell immune suppression and malignant lymphomas in chickens. Toll-like receptor (TLR) plays a dominant role in antiviral T cell immunity. However, it is unclear whether MDV induced T cell immunity is associated with TLR-mediated immunity. In this study, the expression of 28 host genes that are involved in TLR-mediated immunity and MHC-medicated T cell immunity was evaluated in chicken thymus at 7, 14, 21 and 28 days post-infection (dpi). Our results demonstrated that 24 host immune-related genes were upregulated during MDV infection at 7 dpi; however, the expression of most of these genes decreased at 21 and 28 dpi. Notably, a positive correlation was found between the down-regulation of CD4, CD8 and TLR3 signals but not the MyD88-dependent TLR pathway. The present study expanded our knowledge of host immune responses against MDV infection and our results might provide a clue that MDV may interfere with T cell immune response through TLR3 signals.
Collapse
|
18
|
Haq K, Schat KA, Sharif S. Immunity to Marek's disease: where are we now? DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 41:439-446. [PMID: 23588041 DOI: 10.1016/j.dci.2013.04.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 04/02/2013] [Accepted: 04/03/2013] [Indexed: 06/02/2023]
Abstract
Marek's disease (MD) in chickens was first described over a century ago and the causative agent of this disease, Marek's disease virus (MDV), was first identified in the 1960's. There has been extensive and intensive research over the last few decades to elucidate the underlying mechanisms of the interactions between the virus and its host. We have also made considerable progress in terms of developing efficacious vaccines against MD. The advent of the chicken genetic map and genome sequence as well as development of approaches for chicken transcriptome and proteome analyses, have greatly facilitated the process of illuminating underlying genetic mechanisms of resistance and susceptibility to disease. However, there are still major gaps in our understanding of MDV pathogenesis and mechanisms of host immunity to the virus and to the neoplastic events caused by this virus. Importantly, vaccines that can disrupt virus transmission in the field are lacking. The current review explores mechanisms of host immunity against Marek's disease and makes an attempt to identify the areas that are lacking in this field.
Collapse
Affiliation(s)
- Kamran Haq
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Canada
| | | | | |
Collapse
|
19
|
Zhou X, Michal JJ, Zhang L, Ding B, Lunney JK, Liu B, Jiang Z. Interferon induced IFIT family genes in host antiviral defense. Int J Biol Sci 2013; 9:200-8. [PMID: 23459883 PMCID: PMC3584916 DOI: 10.7150/ijbs.5613] [Citation(s) in RCA: 172] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 01/23/2013] [Indexed: 02/06/2023] Open
Abstract
Secretion of interferons (IFNs) from virus-infected cells is a hallmark of host antiviral immunity and in fact, IFNs exert their antiviral activities through the induction of antiviral proteins. The IFN-induced protein with tetratricopeptide repeats (IFITs) family is among hundreds of IFN-stimulated genes. This family contains a cluster of duplicated loci. Most mammals have IFIT1, IFIT2, IFIT3 and IFIT5; however, bird, marsupial, frog and fish have only IFIT5. Regardless of species, IFIT5 is always adjacent to SLC16A12. IFIT family genes are predominantly induced by type I and type III interferons and are regulated by the pattern recognition and the JAK-STAT signaling pathway. IFIT family proteins are involved in many processes in response to viral infection. However, some viruses can escape the antiviral functions of the IFIT family by suppressing IFIT family genes expression or methylation of 5' cap of viral molecules. In addition, the variants of IFIT family genes could significantly influence the outcome of hepatitis C virus (HCV) therapy. We believe that our current review provides a comprehensive picture for the community to understand the structure and function of IFIT family genes in response to pathogens in human, as well as in animals.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Animal Sciences, Washington State University, Pullman, WA 99164-6351, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Sun B, Li X, Wang Z, Xia C. Complex assembly, crystallization and preliminary X-ray crystallographic analysis of the chicken MHC class I molecule BF2*1501. Acta Crystallogr Sect F Struct Biol Cryst Commun 2013; 69:122-5. [PMID: 23385750 PMCID: PMC3564611 DOI: 10.1107/s1744309112050300] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Accepted: 12/10/2012] [Indexed: 11/10/2022]
Abstract
The chicken major histocompatibility complex (MHC) class I molecules named BF are strongly associated with Marek's disease (MD). A single structure, that of chicken BF2*2101 from the B21 haplotype, which might provide resistance to MD, has been determined. However, little is known about other structures apart from BF2*2101. In order to provide further structures of chicken MHC class I molecules, BF2*1501 and chicken β(2)-microglobulin complexed with a nonapeptide (MDV-MEQ(RRR9)) derived from Marek's disease virus MEQ protein (MDV EcoRI Q fragment, residues 72-80) were assembled and crystallized. Diffraction data from the crystal were collected to 2.6 Å resolution; the crystal belonged to space group P3(1)21, with unit-cell parameters a = 125.1, b = 125.1, c = 80.9 Å and two molecules in the asymmetric unit. The Matthews coefficient V(M) was 2.08 Å(3) Da(-1), with a calculated solvent content of 40.78%. These data will be helpful in obtaining insight into the structural basis of the involvement of BF2*1501 in MD.
Collapse
Affiliation(s)
- Beibei Sun
- Department of Microbiology and Immunology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People’s Republic of China
| | - Xiaoying Li
- Department of Microbiology and Immunology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People’s Republic of China
| | - Zhenbao Wang
- Department of Microbiology and Immunology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People’s Republic of China
| | - Chun Xia
- Department of Microbiology and Immunology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People’s Republic of China
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, People’s Republic of China
| |
Collapse
|
21
|
Chang S, Ding Z, Dunn JR, Lee LF, Heidari M, Song J, Ernst CW, Zhang H. A comparative evaluation of the protective efficacy of rMd5deltaMeq and CVI988/ Rispens against a vv+ strain of Marek's disease virus infection in a series of recombinant congenic strains of White Leghorn chickens. Avian Dis 2011; 55:384-90. [PMID: 22017035 DOI: 10.1637/9524-091310-reg.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Marek's disease (MD) is a lymphoproliferative disease of domestic chickens caused by a highly infectious, oncogenic alpha-herpesvirus known as Marek's disease virus (MDV). MD is presently controlled by vaccination. Current MD vaccines include attenuated serotype 1 strains (e.g., CVI988/Rispens), avirulent serotype 2 (SB-1), and serotype 3 (HVT) MDV strains. In addition, recombinant MDV strains have been developed as potential new and more efficient vaccines to sustain the success of MD control in poultry. One of the candidate recombinant MDV strains, named rMd5deltaMeq, was derived from Md5, a very virulent strain of MDV lacking the MDV oncogene Meq. Our earlier reports suggest that rMd5deltaMeq provided protection equally well or better than commonly used MD vaccines in experimental and commercial lines of chickens challenged with very virulent plus (vv+) strains of MDV. In this study, maternal antibody-positive (trial 1) and negative (trial 2) chickens from a series of relatively MD resistant lines were either vaccinated with the rMd5deltaMeq or CVI988/Rispens followed by infection of a vv+ strain of MDV, 648A, passage 10. This report presents experimental evidence that the rMd5deltaMeq protected significantly better than the CVI988/Rispens (P < 0.01) in the relatively resistant experimental lines of chickens challenged with the vv+ strain of MDV. Together with early reports, the rMd5deltaMeq appeared to provide better protection, comparing with the most efficacious commercially available vaccine, CVI988/Rispens, for control of MD in lines of chickens regardless of their genetic background.
Collapse
Affiliation(s)
- Shuang Chang
- United States Department of Agriculture, Agriculture Research Service, Avian Disease and Oncology Laboratory, 3606 E. Mount Hope Road, East Lansing, MI 48823, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Haq K, Elawadli I, Parvizi P, Mallick AI, Behboudi S, Sharif S. Interferon-γ influences immunity elicited by vaccines against very virulent Marek’s disease virus. Antiviral Res 2011; 90:218-26. [DOI: 10.1016/j.antiviral.2011.04.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Revised: 03/28/2011] [Accepted: 04/04/2011] [Indexed: 12/16/2022]
|
23
|
Haq K, Brisbin JT, Thanthrige-Don N, Heidari M, Sharif S. Transcriptome and proteome profiling of host responses to Marek's disease virus in chickens. Vet Immunol Immunopathol 2010; 138:292-302. [PMID: 21067815 DOI: 10.1016/j.vetimm.2010.10.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Kamran Haq
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | | | | | | | | |
Collapse
|