1
|
Wang L, Zheng G, Yang Y, Wu J, Du Y, Chen J, Liu C, Liu Y, Zhang B, Zhang H, Deng X, Lian L. Rolling-Translated circRUNX2.2 Promotes Lymphoma Cell Proliferation and Cycle Transition in Marek's Disease Model. Int J Mol Sci 2024; 25:11486. [PMID: 39519039 PMCID: PMC11545863 DOI: 10.3390/ijms252111486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Marek's disease (MD), an immunosuppressive disease induced by the Marek's disease virus (MDV), is regarded as an ideal model for lymphoma research to elucidate oncogenic and anti-oncogene genes. Using this model, we found that circRUNX2.2, derived from exon 6 of RUNX2, was significantly upregulated in MDV-infected tumorous spleens. In this study, we deeply analyzed the potential role of circRUNX2.2 in lymphoma cells. An open reading frame (ORF) in circRUNX2.2 with no stop codon was predicted, and small peptides (named circRUNX2.2-rt) presenting multiple ladder-like bands with different molecular weights encoded by circRUNX2.2 were detected via Western blotting assay. The polysome fraction assay reconfirmed the translation ability of circRUNX2.2, which could be detected in polysome fractions. Subsequent analysis verified that it translated in a rolling circle manner, rather than being assisted by the internal ribosome entry site (IRES) or m6A-mediated mechanism. Furthermore, we found that circRUNX2.2-rt was potently induced in MSB1 cells treated with sodium butyrate (NaB), which reactivated MDV and forced the MDV transition from the latent to reactivation phase. During this phase, MDV particles were clearly observed by electron microscopy, and the viral gene pp38 was also significantly upregulated. A biological function study showed that circRUNX2.2-rt promoted cell proliferation and cell cycle transition from the S to G2 phase and inhibited the apoptosis of MSB1. Further immunoprecipitation and mass spectrometry assays showed that 168 proteins potentially interacting with circRUNX2.2-rt were involved in multiple pathways related to cell cycle regulation, which proved that circRUNX2.2-rt could bind or recruit proteins to mediate the cell cycle.
Collapse
Affiliation(s)
- Lulu Wang
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing 100193, China
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Gang Zheng
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing 100193, China
| | - Yuqin Yang
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing 100193, China
| | - Junfeng Wu
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing 100193, China
| | - Yushuang Du
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing 100193, China
| | - Jiahua Chen
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing 100193, China
| | - Changjun Liu
- Division of Avian Infectious Diseases, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Yongzhen Liu
- Division of Avian Infectious Diseases, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Bo Zhang
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing 100193, China
| | - Hao Zhang
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing 100193, China
| | - Xuemei Deng
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing 100193, China
| | - Ling Lian
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing 100193, China
| |
Collapse
|
2
|
Tien YT, Akbar H, Jarosinski KW. Temperature-induced reactivation of Marek's disease virus-transformed T cells ex vivo. Front Vet Sci 2023; 10:1145757. [PMID: 36968465 PMCID: PMC10030735 DOI: 10.3389/fvets.2023.1145757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/15/2023] [Indexed: 03/29/2023] Open
Abstract
Marek's disease virus (MDV) establishes latency in chicken T lymphocytes that can lead to T cell transformation and cancer. Transformed Marek's disease chicken cell lines (MDCCs) can be expanded ex vivo and provide a valuable model to study latency, transformation, and reactivation. Here, we developed MDCCs from chickens infected with MDV that fluoresce during lytic replication and reactivation. Sodium butyrate treatment increased fluorescent protein expression as evidenced by fluorescent microscopy, flow cytometry, and western blotting; however, it caused significant apoptosis and necrosis. Treatment of MDCCs by decreasing the temperature resulted in robust MDV reactivation without significant induction of apoptosis and necrosis. Furthermore, MDV reactivation was significantly affected by the time in culture that can affect downstream reactivation analyses. In all, our data show that fluorescent protein expression during reactivation is a robust tool to examine viral replication in live cells ex vivo, and temperature treatment is an efficient technique to induce reactivation without punitive effects on cell viability seen with chemical treatment.
Collapse
|
3
|
Schat KA. The Importance of the Bursa of Fabricius, B Cells and T Cells for the Pathogenesis of Marek’s Disease: A Review. Viruses 2022; 14:v14092015. [PMID: 36146821 PMCID: PMC9504545 DOI: 10.3390/v14092015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/21/2022] Open
Abstract
The importance of the bursa of Fabricius (BF) for the pathogenesis of Marek’s disease (MD) has been studied since the late 1960’s. In this review, the results of these studies are analyzed in the context of the developing knowledge of the immune system of chickens and the pathogenesis of MD from 1968 to 2022. Based on the available techniques to interfere with the development of the BF, three distinct periods are identified and discussed. During the initial period between 1968 and 1977, the use of neonatal bursectomy, chemical methods and irradiation were the main tools to interfere with the B lymphocyte development. The application of these techniques resulted in contradictory results from no effects to an increase or decrease in MD incidence. Starting in the late 1970’s, the use of bursectomy in 18-day-old embryos led to the development of the “Cornell model” for the pathogenesis of MD, in which the infection of B lymphocytes is an important first step in MD virus (MDV) replication causing the activation of thymus-derived lymphocytes (T cells). Following this model, these activated T cells, but not resting T cells, are susceptible to MDV infection and subsequent transformation. Finally, B-cell knockout chickens lacking the J gene segment of the IgY heavy chain gene were used to further define the role of the BF in the pathogenesis of MD.
Collapse
Affiliation(s)
- Karel A Schat
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
4
|
Glass MC, Smith JM, Cheng HH, Delany ME. Marek's Disease Virus Telomeric Integration Profiles of Neoplastic Host Tissues Reveal Unbiased Chromosomal Selection and Loss of Cellular Diversity during Tumorigenesis. Genes (Basel) 2021; 12:1630. [PMID: 34681024 PMCID: PMC8536068 DOI: 10.3390/genes12101630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/14/2021] [Accepted: 10/14/2021] [Indexed: 11/17/2022] Open
Abstract
The avian α-herpesvirus known as Marek's disease virus (MDV) linearly integrates its genomic DNA into host telomeres during infection. The resulting disease, Marek's disease (MD), is characterized by virally-induced lymphomas with high mortality. The temporal dynamics of MDV-positive (MDV+) transformed cells and expansion of MD lymphomas remain targets for further understanding. It also remains to be determined whether specific host chromosomal sites of MDV telomere integration confer an advantage to MDV-transformed cells during tumorigenesis. We applied MDV-specific fluorescence in situ hybridization (MDV FISH) to investigate virus-host cytogenomic interactions within and among a total of 37 gonad lymphomas and neoplastic splenic samples in birds infected with virulent MDV. We also determined single-cell, chromosome-specific MDV integration profiles within and among transformed tissue samples, including multiple samples from the same bird. Most mitotically-dividing cells within neoplastic samples had the cytogenomic phenotype of 'MDV telomere-integrated only', and tissue-specific, temporal changes in phenotype frequencies were detected. Transformed cell populations composing gonad lymphomas exhibited significantly lower diversity, in terms of heterogeneity of MDV integration profiles, at the latest stages of tumorigenesis (>50 days post-infection (dpi)). We further report high interindividual and lower intraindividual variation in MDV integration profiles of lymphoma cells. There was no evidence of integration hotspots into a specific host chromosome(s). Collectively, our data suggests that very few transformed MDV+ T cell populations present earlier in MDV-induced lymphomas (32-50 dpi), survive, and expand to become the dominant clonal population in more advanced MD lymphomas (51-62 dpi) and establish metastatic lymphomas.
Collapse
Affiliation(s)
- Marla C. Glass
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Justin M. Smith
- Department of Animal Science, University of California Davis, Davis, CA 95616, USA; (J.M.S.); (M.E.D.)
| | - Hans H. Cheng
- Avian Disease and Oncology Laboratory, United States Department of Agriculture, Agricultural Research Service, East Lansing, MI 48823, USA;
| | - Mary E. Delany
- Department of Animal Science, University of California Davis, Davis, CA 95616, USA; (J.M.S.); (M.E.D.)
| |
Collapse
|
5
|
Boone AC, Käser T, Cortes AL, Kulkarni RR, López de Juan Abad BA, Villalobos T, Esandi J, Perozo F, Lemiere S, Gimeno IM. In ovo vaccination with herpesvirus of turkey enhances innate and cellular responses in meat-type chickens: Effect of vaccine dose and strain. Vaccine 2020; 38:4837-4845. [PMID: 32505441 DOI: 10.1016/j.vaccine.2020.05.050] [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: 02/13/2020] [Revised: 05/11/2020] [Accepted: 05/17/2020] [Indexed: 01/31/2023]
Abstract
In ovo vaccination with herpesvirus of turkey (HVT) or recombinant HVT (rHVT) is commonly used in meat-type chickens. Previous studies showed that in ovo vaccination with HVT enhances innate, cellular, and humoral immune responses in egg-type chicken embryos. This study evaluated if in ovo vaccination with HVT hastens immunocompetence of commercial meat-type chickens and optimized vaccination variables (dose and strain of HVT) to accelerate immunocompetence. A conventional HVT vaccine was given at recommended dose (RD), HVT-RD = 6080 plaque forming units (PFU), double-dose (2x), half-dose (1/2), or quarter-dose (1/4). Two rHVTs were given at RD: rHVT-A = 7380 PFU, rHVT-B = 8993 PFU. Most, if not all, treatments enhanced splenic lymphoproliferation with Concanavalin A and increased the percentage of granulocytes at day of age. Dose had an effect and HVT-RD was ideal. An increase of wing-web thickness after exposure to phytohemagglutinin-L was only detected after vaccination with HVT-RD. Furthermore, compared to sham-inoculated chickens, chickens in the HVT-RD had an increased percentage of CD3+ T cells and CD4+ T-helper cells, and increased expression of major histocompatibility complex (MHC)-II on most cell subsets (CD45+ cells, non-T leukocytes, T cells and the CD8+ and T cell receptor γδ T-cell subsets). Other treatments (HVT-1/2 and rHVT-B) share some of these features but differences were not as remarkable as in the HVT-RD group. Expression of MHC-I was reduced, compared to sham-inoculated chickens, in most of the cell phenotypes evaluated in the HVT-RD, HVT-2x and rHVT-A groups, while no effect was observed in other treatments. The effect of in ovo HVT on humoral immune responses (antibody responses to keyhole limpet hemocyanin and to a live infectious bronchitis/Newcastle disease vaccine) was minimal. Our study demonstrates in ovo vaccination with HVT in meat-type chickens can accelerate innate and adaptive immunity and we could optimize such effect by modifying the vaccine dose.
Collapse
Affiliation(s)
- Allison C Boone
- North Carolina State University, College of Veterinary Medicine, 1060 William Moore Drive, Raleigh, NC 27607, United States; Rollins Animal Disease Diagnostic Laboratory, 2101 Blue Ridge Road, Raleigh, NC 27607, United States.
| | - Tobias Käser
- North Carolina State University, College of Veterinary Medicine, 1060 William Moore Drive, Raleigh, NC 27607, United States.
| | - Aneg L Cortes
- North Carolina State University, College of Veterinary Medicine, 1060 William Moore Drive, Raleigh, NC 27607, United States.
| | - Raveendra R Kulkarni
- North Carolina State University, College of Veterinary Medicine, 1060 William Moore Drive, Raleigh, NC 27607, United States.
| | - Blanca A López de Juan Abad
- North Carolina State University, College of Veterinary Medicine, 1060 William Moore Drive, Raleigh, NC 27607, United States.
| | - Tarsicio Villalobos
- Zoetis-International Biodevices and Automation, 1040 Swabia Ct, Durham, NC 27703, United States.
| | - Javier Esandi
- Zoetis-International Biodevices and Automation, 1040 Swabia Ct, Durham, NC 27703, United States.
| | - Francisco Perozo
- Boehringer Ingelheim Animal Health, Binger Strasse 173, 55216 Ingelheim, Germany.
| | - Stephane Lemiere
- Boehringer Ingelheim Animal Health, Binger Strasse 173, 55216 Ingelheim, Germany.
| | - Isabel M Gimeno
- North Carolina State University, College of Veterinary Medicine, 1060 William Moore Drive, Raleigh, NC 27607, United States.
| |
Collapse
|
6
|
He Y, Han B, Ding Y, Zhang H, Chang S, Zhang L, Zhao C, Yang N, Song J. Linc-GALMD1 Regulates Viral Gene Expression in the Chicken. Front Genet 2019; 10:1122. [PMID: 31798630 PMCID: PMC6868033 DOI: 10.3389/fgene.2019.01122] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 10/16/2019] [Indexed: 12/17/2022] Open
Abstract
A rapidly increasing number of reports on dysregulated long intergenic non-coding RNA (lincRNA) expression across numerous types of cancers indicates that aberrant lincRNA expression may be a major contributor to tumorigenesis. Marek’s disease (MD) is a T cell lymphoma of chickens induced by Marek’s disease virus (MDV). Although we have investigated the roles of lincRNAs in bursa tissue of MDV-infected chickens in previous studies, the molecular mechanisms of lincRNA functions in T cells remain poorly understood. In the present study, Linc-GALMD1 was identified from CD4+ T cells and MSB1 cells, and its expression was significantly downregulated in MD-resistant line of birds in response to MDV challenge. Furthermore, loss-of-function experiments indicated that linc-GALMD1 significantly affected the expression of 290 genes in trans. Through integrated analysis of differentially expressed genes (DEGs) induced by MDV and linc-GALMD1, we found that IGLL1 gene expression levels had a positive correlation with the degree of MD infection and could potentially serve as an indicator for clinical diagnosis of MD. Moreover, an interaction between MDV and linc-GALMD1 was also observed. Accordingly, chicken embryonic fibroblast cells were inoculated with MDV with and without the linc-GALMD1 knockdown, and the data showed that linc-GALMD1 could repress MDV gene expression during the course of MDV infection. These findings uncovered a role of linc-GALMD1 as a viral gene regulator and suggested a function of linc-GALMD1 contributing to tumor suppression by coordinating expression of MDV genes and tumor-related genes and regulating immune responses to MDV infection.
Collapse
Affiliation(s)
- Yanghua He
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Bo Han
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, United States.,National Engineering Laboratory for Animal Breeding, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yi Ding
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, United States
| | - Huanmin Zhang
- Avian Disease and Oncology Laboratory, Agricultural Research Service, USDA, East Lansing, MI, United States
| | - Shuang Chang
- Avian Disease and Oncology Laboratory, Agricultural Research Service, USDA, East Lansing, MI, United States.,College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Li Zhang
- Institute of Animal Science and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Chunfang Zhao
- National Engineering Laboratory for Animal Breeding, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ning Yang
- National Engineering Laboratory for Animal Breeding, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jiuzhou Song
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, United States
| |
Collapse
|
7
|
Bai H, He Y, Ding Y, Carrillo JA, Selvaraj RK, Zhang H, Chen J, Song J. Allele-Specific Expression of CD4 + T Cells in Response to Marek's Disease Virus Infection. Genes (Basel) 2019; 10:E718. [PMID: 31533276 PMCID: PMC6770979 DOI: 10.3390/genes10090718] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 12/15/2022] Open
Abstract
Marek's disease (MD) is a T cell lymphoma disease induced by Marek's disease virus (MDV), a highly oncogenic α herpesvirus primarily affecting chickens. MD is a chronic infectious disease that threatens the poultry industry. However, the mechanisms of genetic resistance for MD are complex and not completely understood. In this study, to identify high-confidence candidate genes of MD genetic resistance, high throughput sequencing (RNA-seq) was used to obtain transcriptomic data of CD4+ T cells isolated from MDV-infected and non-infected groups of two reciprocal crosses of individuals mating by two highly inbred chicken lines (63 MD-resistant and 72 MD-susceptible). After RNA-seq analysis with two biological replicates in each group, we identified 61 and 123 single nucleotide polymorphisms (SNPs) (false discovery rate (FDR) < 0.05) annotated in 39 and 132 genes in intercrosses 63 × 72 and 72 × 63, respectively, which exhibited allele-specific expression (ASE) in response to MDV infection. Similarly, we identified 62 and 79 SNPs annotated in 66 and 96 genes in infected and non-infected groups, respectively. We identified 534 and 1543 differentially expressed genes (DEGs) (FDR < 0.05) related to MDV infection in intercrosses 63 × 72 and 72 × 63, respectively. We also identified 328 and 20 DEGs in infected and non-infected groups, respectively. The qRT-PCR using seven DEGs further verified our results of RNA-seq analysis. The qRT-PCR of 11 important ASE genes was performed for gene functional validation in CD4+ T cells and tumors. Combining the analyses, six genes (MCL1, SLC43A2, PDE3B, ADAM33, BLB1, and DMB2), especially MCL1, were highlighted as the candidate genes with the potential to be involved in MDV infection. Gene-set enrichment analysis revealed that many ASE genes are linked to T cell activation, T cell receptor (TCR), B cell receptor (BCR), ERK/MAPK, and PI3K/AKT-mTOR signaling pathways, which play potentially important roles in MDV infection. Our approach underlines the importance of comprehensive functional studies for gaining valuable biological insight into the genetic factors behind MD and other complex traits, and our findings provide additional insights into the mechanisms of MD and disease resistance breeding in poultry.
Collapse
Affiliation(s)
- Hao Bai
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD 20742, USA
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yanghua He
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD 20742, USA
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, HI 96822, USA
| | - Yi Ding
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - José A Carrillo
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Ramesh K Selvaraj
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - Huanmin Zhang
- USDA, Agricultural Research Service, Avian Disease and Oncology Laboratory, East Lansing, MI 48823, USA
| | - Jilan Chen
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jiuzhou Song
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
8
|
Umthong S, Dunn JR, Cheng HH. Towards a mechanistic understanding of the synergistic response induced by bivalent Marek's disease vaccines to prevent lymphomas. Vaccine 2019; 37:6397-6404. [PMID: 31515142 DOI: 10.1016/j.vaccine.2019.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/30/2019] [Accepted: 09/02/2019] [Indexed: 12/01/2022]
Abstract
BACKGROUND Marek's disease (MD) is a lymphoproliferative disease of chickens caused by Marek's disease virus (MDV), an oncogenic α-herpesvirus. Since 1970, MD has been controlled by widespread vaccination; however, more effective MD vaccines are needed to counter more virulent MDV strains. The bivalent vaccine combination of SB-1 and herpesvirus of turkey (HVT) strain FC126 has been widely used. Nonetheless, the mechanism(s) underlying this synergistic effect has not been investigated. METHODS Three experiments were conducted where SB-1 or HVT were administered as monovalent or bivalent vaccines to newly hatched chickens, then challenged five days later with MDV. In Experiment 1, levels of MDV replication in PBMCs were measured over time, and tumor incidence and vaccinal protection determined. In Experiment 2, MDV and vaccine strains replication levels in lymphoid organs were measured at 1, 5, 10, and 14 days post-challenge (DPC). In Experiment 3, to verify that the bursa was necessary for HVT protection, a subset of chicks were bursectomized and these birds plus controls were similarly vaccinated and challenged, and the levels of vaccinal protection determined. RESULTS The efficacy of bivalent SB-1 + HVT surpasses that of either SB-1 or HVT monovalent vaccines in controlling the level of pathogenic MDV in PBMCs until the end of the study, and this correlated with the ability to inhibit tumor formation. SB-1 replication in the spleen increased from 1 to 14 DPC, while HVT replicated only in the bursa at 1 DPC. The bursa was necessary for immune protection induced by HVT vaccine. CONCLUSION Synergy of SB-1 and HVT vaccines is due to additive influences of the individual vaccines acting at different times and target organs. And the bursa is vital for HVT to replicate and induce immune protection.
Collapse
Affiliation(s)
- Supawadee Umthong
- Microbiology and Molecular Genetics Program, Michigan State University, East Lansing, MI, USA; USDA, ARS, U.S. National Poultry Research Center, Avian Disease and Oncology Laboratory, East Lansing, MI, USA
| | - John R Dunn
- USDA, ARS, U.S. National Poultry Research Center, Avian Disease and Oncology Laboratory, East Lansing, MI, USA
| | - Hans H Cheng
- USDA, ARS, U.S. National Poultry Research Center, Avian Disease and Oncology Laboratory, East Lansing, MI, USA.
| |
Collapse
|
9
|
Abstract
A healthy immune system is a cornerstone for poultry production. Any factor diminishing the immune responses will affect production parameters and increase cost. There are numerous factors, infectious and noninfectious, causing immunosuppression (IS) in chickens. This paper reviews the three viral diseases that most commonly induce IS or subclinical IS in chickens: Marek's disease virus (MDV), chicken infectious anemia virus (CIAV), and infectious bursal disease virus (IBDV), as well as the interactions among them. MDV-induced IS (MDV-IS) affects both humoral and cellular immune responses. It is very complex, poorly understood, and in many cases underdiagnosed. Vaccination protects against some but not all aspects of MDV-IS. CIAV induces apoptosis of the hemocytoblasts resulting in anemia, hemorrhages, and increased susceptibility to bacterial infections. It also causes apoptosis of thymocytes and dividing T lymphocytes, affecting T helper functions, which are essential for antibody production and cytotoxic T lymphocyte (CTL) functions. Control of CIAV is based on vaccination of breeders and maternal antibodies (MAbs). However, subclinical IS can occur after MAbs wane. IBDV infection affects the innate immune responses during virus replication and humoral immune responses as a consequence of the destruction of B-cell populations. Vaccines with various levels of attenuation are used to control IBDV. Interactions with MAbs and residual virulence of the vaccines need to be considered when designing vaccination plans. The interaction between IBDV, CIAV, and MDV is critical although underestimated in many cases. A proper control of IBDV is a must to have proper humoral immune responses needed to control CIAV. Equally, long-term control of MDV is not possible if chickens are coinfected with CIAV, as CIAV jeopardizes CTL functions critical for MDV control.
Collapse
Affiliation(s)
- I M Gimeno
- A Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607
| | - K A Schat
- B Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| |
Collapse
|
10
|
The Transcriptional Landscape of Marek's Disease Virus in Primary Chicken B Cells Reveals Novel Splice Variants and Genes. Viruses 2019; 11:v11030264. [PMID: 30884829 PMCID: PMC6466439 DOI: 10.3390/v11030264] [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: 02/18/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/14/2022] Open
Abstract
Marek's disease virus (MDV) is an oncogenic alphaherpesvirus that infects chickens and poses a serious threat to poultry health. In infected animals, MDV efficiently replicates in B cells in various lymphoid organs. Despite many years of research, the viral transcriptome in primary target cells of MDV remained unknown. In this study, we uncovered the transcriptional landscape of the very virulent RB1B strain and the attenuated CVI988/Rispens vaccine strain in primary chicken B cells using high-throughput RNA-sequencing. Our data confirmed the expression of known genes, but also identified a novel spliced MDV gene in the unique short region of the genome. Furthermore, de novo transcriptome assembly revealed extensive splicing of viral genes resulting in coding and non-coding RNA transcripts. A novel splicing isoform of MDV UL15 could also be confirmed by mass spectrometry and RT-PCR. In addition, we could demonstrate that the associated transcriptional motifs are highly conserved and closely resembled those of the host transcriptional machinery. Taken together, our data allow a comprehensive re-annotation of the MDV genome with novel genes and splice variants that could be targeted in further research on MDV replication and tumorigenesis.
Collapse
|
11
|
Bertzbach LD, Kheimar A, Ali FAZ, Kaufer BB. Viral Factors Involved in Marek’s Disease Virus (MDV) Pathogenesis. CURRENT CLINICAL MICROBIOLOGY REPORTS 2018. [DOI: 10.1007/s40588-018-0104-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
12
|
Yang S, Wang L, Sun S. Natural Infection with Avian Hepatitis E Virus and Marek's Disease Virus in Brown Layer Chickens in China. Avian Dis 2017; 60:698-704. [PMID: 27610734 DOI: 10.1637/11386-013016-reg.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
In the present study, avian hepatitis E virus (HEV) and serotype-1 strains of Marek's disease virus (MDV-1) were detected from a flock of 27-wk-old brown layer hens in China, accompanied by an average daily mortality of 0.44%. Postmortem examination of 25 sick hens and five apparently healthy hens selected randomly from the flock showed significant pathologic changes consistent with hepatitis-splenomegaly syndrome (HSS), including hepatomegaly, peritoneal fluid, and hepatic subcapsular hemorrhages. Microscopic examination of these livers showed multifocal necrotizing hepatitis and mild lymphocytic infiltration. These liver samples were investigated for HEV by reverse-transcription PCR. The overall detection rate of HEV RNA in samples of sick chickens was about 56% (14/25), while in samples from apparently healthy hens, it was 80% (4/5). Sequencing analysis of three 242-base-pair fragments of the helicase gene revealed 95.5% to 97.9% nucleotide identity compared with published avian HEV genotype 3, whereas identities demonstrated only 77.3% to 86.0% similarity when compared with genotypes 1, 2, and 4. Unexpectedly, the MDV meq gene was detected in livers from both apparently healthy chickens (2/5) and sick chickens (12/25) by PCR analysis. The meq gene (396 base pairs) was determined to belong to MDV-1 by further sequencing. The co-infection rate of avian HEV and MDV in this flock was 30% (9/30). This is the first report of dual infection of a nonenvelope RNA virus (HEV) with a herpesvirus (MDV) in chickens in China.
Collapse
Affiliation(s)
- Shuqing Yang
- Shandong Agricultural University, Tai An, Shandong Province, 271000, People's Republic of China
| | - Liyuan Wang
- Shandong Agricultural University, Tai An, Shandong Province, 271000, People's Republic of China
| | - Shuhong Sun
- Shandong Agricultural University, Tai An, Shandong Province, 271000, People's Republic of China
| |
Collapse
|
13
|
Chakraborty P, Vervelde L, Dalziel RG, Wasson PS, Nair V, Dutia BM, Kaiser P. Marek's disease virus infection of phagocytes: a de novo in vitro infection model. J Gen Virol 2017; 98:1080-1088. [PMID: 28548038 PMCID: PMC5656796 DOI: 10.1099/jgv.0.000763] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 03/06/2017] [Indexed: 01/08/2023] Open
Abstract
Marek's disease virus (MDV) is an alphaherpesvirus that induces T-cell lymphomas in chickens. Natural infections in vivo are caused by the inhalation of infected poultry house dust and it is presumed that MDV infection is initiated in the macrophages from where the infection is passed to B cells and activated T cells. Virus can be detected in B and T cells and macrophages in vivo, and both B and T cells can be infected in vitro. However, attempts to infect macrophages in vitro have not been successful. The aim of this study was to develop a model for infecting phagocytes [macrophages and dendritic cells (DCs)] with MDV in vitro and to characterize the infected cells. Chicken bone marrow cells were cultured with chicken CSF-1 or chicken IL-4 and chicken CSF-2 for 4 days to produce macrophages and DCs, respectively, and then co-cultured with FACS-sorted chicken embryo fibroblasts (CEFs) infected with recombinant MDV expressing EGFP. Infected phagocytes were identified and sorted by FACS using EGFP expression and phagocyte-specific mAbs. Detection of MDV-specific transcripts of ICP4 (immediate early), pp38 (early), gB (late) and Meq by RT-PCR provided evidence for MDV replication in the infected phagocytes. Time-lapse confocal microscopy was also used to demonstrate MDV spread in these cells. Subsequent co-culture of infected macrophages with CEFs suggests that productive virus infection may occur in these cell types. This is the first report of in vitro infection of phagocytic cells by MDV.
Collapse
Affiliation(s)
- Pankaj Chakraborty
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
- Present address: Chittagong Veterinary and Animal Sciences University, Khulshi, Chittagong 4225, Bangladesh
| | - Lonneke Vervelde
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Robert G. Dalziel
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Peter S. Wasson
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
- Present address: MRC Technology, Crewe Road South, Edinburgh EH4 2SP, UK
| | - Venugopal Nair
- Avian Oncogenic Virus Group, The Pirbright Institute, Guildford GU24 0NF, UK
| | - Bernadette M. Dutia
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Pete Kaiser
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| |
Collapse
|
14
|
Mwangi WN, Vasoya D, Kgosana LB, Watson M, Nair V. Differentially expressed genes during spontaneous lytic switch of Marek's disease virus in lymphoblastoid cell lines determined by global gene expression profiling. J Gen Virol 2017; 98:779-790. [PMID: 28475033 PMCID: PMC5657026 DOI: 10.1099/jgv.0.000744] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Marek's disease virus (MDV), an alphaherpesvirus of poultry, causes Marek's disease and is characterized by visceral CD4+TCRαβ+ T-cell lymphomas in susceptible hosts. Immortal cell lines harbouring the viral genome have been generated from ex vivo cultures of MD tumours. As readily available sources of large numbers of cells, MDV-transformed lymphoblastoid cell lines (LCLs) are extremely valuable for studies of virus-host interaction. While the viral genome in most cells is held in a latent state, minor populations of cells display spontaneous reactivation identifiable by the expression of lytic viral genes. Spontaneous reactivation in these cells presents an opportunity to investigate the biological processes involved in the virus reactivation. For detailed characterization of the molecular events associated with reactivation, we used two lymphoblastoid cell lines derived from lymphomas induced by pRB1B-UL47eGFP, a recombinant MDV engineered to express enhanced green fluorescent protein (EGFP) fused with the UL47. We used fluorescence-activated cell sorting to purify the low-frequency EGFP-positive cells with a spontaneously activating viral genome from the majority EGFP-negative cells and analysed their gene expression profiles by RNA-seq using Illumina HiSeq2500. Ingenuity pathway analysis on more than 2000 differentially expressed genes between the lytically infected (EGFP-positive) and latently infected (EGFP-negative) cell populations identified the biological pathways involved in the reactivation. Virus-reactivating cells exhibited differential expression of a significant number of viral genes, with hierarchical differences in expression levels. Downregulation of a number of host genes including those directly involved in T-cell activation, such as CD3, CD28, ICOS and phospholipase C, was also noticed in the LCL undergoing lytic switch.
Collapse
Affiliation(s)
- William N Mwangi
- Avian Viral Diseases Programme, UK-China Centre of Excellence on Avian Disease Research, The Pirbright Institute, Pirbright, Surrey, UK
| | - Deepali Vasoya
- Division of Genetics and Genomics, The Roslin Institute, R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, UK
| | - Lydia B Kgosana
- Avian Viral Diseases Programme, UK-China Centre of Excellence on Avian Disease Research, The Pirbright Institute, Pirbright, Surrey, UK
| | - Mick Watson
- Division of Genetics and Genomics, The Roslin Institute, R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, UK
| | - Venugopal Nair
- Avian Viral Diseases Programme, UK-China Centre of Excellence on Avian Disease Research, The Pirbright Institute, Pirbright, Surrey, UK
| |
Collapse
|
15
|
Murata S, Chang KS, Lee SI, Konnai S, Onuma M, Ohashi K. Development of a Nested Polymerase Chain Reaction Method to Detect Oncogenic Marek's Disease Virus from Feather Tips. J Vet Diagn Invest 2016; 19:471-8. [PMID: 17823389 DOI: 10.1177/104063870701900503] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
For the easy survey of Marek's disease virus (MDV), feather tip–derived DNA from MDV-infected chickens can be used because feather tips are easy to collect and feather follicle epithelium is known to be the only site of productive replication of cell-free MDV. To develop a diagnostic method to differentiate highly virulent strains of MDV from the attenuated MDV vaccine strain, CVI988, which is widely used, nested polymerase chain reaction (PCR) was performed to detect a segment of the meq gene in feather tip samples of chickens experimentally infected with MDV. In chickens infected with Md5, a strain of oncogenic MDV, the meq gene was consistently detected, whereas the L- meq gene, in which a 180–base pair (180-bp) sequence is inserted into the meq gene, was detected in CVI988-infected chickens. Moreover, the meq gene was mainly detected even in chickens co-infected with both Md5 and CVI988. These results suggest that this method is appropriate for the surveillance of the highly virulent MDV infection in the field.
Collapse
Affiliation(s)
- Shiro Murata
- Department of Disease Control, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | | | | | | | | | | |
Collapse
|
16
|
Perumbakkam S, Hunt HD, Cheng HH. Differences in CD8αα and cecal microbiome community during proliferation and late cytolytic phases of Marek's disease virus infection are associated with genetic resistance to Marek's disease. FEMS Microbiol Ecol 2016; 92:fiw188. [PMID: 27604255 DOI: 10.1093/femsec/fiw188] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2016] [Indexed: 12/12/2022] Open
Abstract
Marek's disease (MD) is an important neoplastic disease of chickens caused by Marek ': s disease virus (MDV), a highly oncogenic alphaherpesvirus. In this study using two chicken lines, one resistant and another susceptible to MD, splenic T cells and cecal microbiome were profiled to gain a better understanding of primary differences in these lines. The percent of splenic CD4+ T cells were similar regardless of MDV challenge status in both bird lines. In contrast, CD8αα profiles were different (P < 0.005) between chicken lines under naïve status and under MDV challenge, suggesting that CD8αα T cells play a key role in mediating MDV infection. Microbiome composition was different between naïve resistant (Blautia spp.) and susceptible birds (Streptococcus spp.) (P < 0.05) during initial colonization. With MDV challenge, both chicken lines showed lower numbers of beneficial Faecalibacterium spp. and increased number of Lactobacillus spp. Metabolic profiles between naïve chicken types were similar but with MDV challenge, there were differences in metabolism in both chicken lines, with amino acid metabolism impacted in resistant birds and lipid metabolism in susceptible birds. These results provide insights into immune response and potential interplay with the microbiome during infection with an oncogenic virus.
Collapse
Affiliation(s)
- Sudeep Perumbakkam
- USDA, ARS, Avian Diseases and Oncology Laboratory, East Lansing, MI 48823, USA Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Henry D Hunt
- USDA, ARS, Avian Diseases and Oncology Laboratory, East Lansing, MI 48823, USA
| | - Hans H Cheng
- USDA, ARS, Avian Diseases and Oncology Laboratory, East Lansing, MI 48823, USA
| |
Collapse
|
17
|
McPherson MC, Delany ME. Virus and host genomic, molecular, and cellular interactions during Marek's disease pathogenesis and oncogenesis. Poult Sci 2016; 95:412-29. [PMID: 26755654 PMCID: PMC4957504 DOI: 10.3382/ps/pev369] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/09/2015] [Indexed: 01/16/2023] Open
Abstract
Marek's Disease Virus (MDV) is a chicken alphaherpesvirus that causes paralysis, chronic wasting, blindness, and fatal lymphoma development in infected, susceptible host birds. This disease and its protective vaccines are highly relevant research targets, given their enormous impact within the poultry industry. Further, Marek's disease (MD) serves as a valuable model for the investigation of oncogenic viruses and herpesvirus patterns of viral latency and persistence--as pertinent to human health as to poultry health. The objectives of this article are to review MDV interactions with its host from a variety of genomic, molecular, and cellular perspectives. In particular, we focus on cytogenetic studies, which precisely assess the physical status of the MDV genome in the context of the chicken host genome. Combined, the cytogenetic and genomic research indicates that MDV-host genome interactions, specifically integration of the virus into the host telomeres, is a key feature of the virus life cycle, contributing to the viral achievement of latency, transformation, and reactivation of lytic replication. We present a model that outlines the variety of virus-host interactions, at the multiple levels, and with regard to the disease states.
Collapse
Affiliation(s)
- M C McPherson
- Department of Animal Science, University of California, Davis, CA 95616
| | - M E Delany
- Department of Animal Science, University of California, Davis, CA 95616
| |
Collapse
|
18
|
In vitro model for lytic replication, latency, and transformation of an oncogenic alphaherpesvirus. Proc Natl Acad Sci U S A 2015; 112:7279-84. [PMID: 26039998 PMCID: PMC4466724 DOI: 10.1073/pnas.1424420112] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Marek's disease virus (MDV) is an alphaherpesvirus that causes deadly T-cell lymphomas in chickens and serves as a natural small animal model for virus-induced tumor formation. In vivo, MDV lytically replicates in B cells that transfer the virus to T cells in which the virus establishes latency. MDV also malignantly transforms CD4+ T cells with a T(reg) signature, ultimately resulting in deadly lymphomas. No in vitro infection system for primary target cells of MDV has been available due to the short-lived nature of these cells in culture. Recently, we characterized cytokines and monoclonal antibodies that promote survival of cultured chicken B and T cells. We used these survival stimuli to establish a culture system that allows efficient infection of B and T cells with MDV. We were able to productively infect with MDV B cells isolated from spleen, bursa or blood cultured in the presence of soluble CD40L. Virus was readily transferred from infected B to T cells stimulated with an anti-TCRαVβ1 antibody, thus recapitulating the in vivo situation in the culture dish. Infected T cells could then be maintained in culture for at least 90 d in the absence of TCR stimulation, which allowed the establishment of MDV-transformed lymphoblastoid cell lines (LCL). The immortalized cells had a signature comparable to MDV-transformed CD4+ α/β T cells present in tumors. In summary, we have developed a novel in vitro system that precisely reflects the life cycle of an oncogenic herpesivrus in vivo and will allow us to investigate the interaction between virus and target cells in an easily accessible system.
Collapse
|
19
|
Role of the short telomeric repeat region in Marek's disease virus replication, genomic integration, and lymphomagenesis. J Virol 2014; 88:14138-47. [PMID: 25275118 DOI: 10.1128/jvi.02437-14] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
UNLABELLED Marek's disease virus (MDV) is a cell-associated alphaherpesvirus that causes generalized polyneuritis and T-cell lymphomas in chickens. MDV is able to integrate its genome into host telomeres, but the mechanism of integration is poorly understood. The MDV genome harbors two arrays of telomeric repeats (TMR) at the ends of its linear genome: multiple telomeric repeats (mTMR), with a variable number of up to 100 repeats, and short telomeric repeats (sTMR), with a fixed number of 6 repeats. The mTMR have recently been shown to play an important role in MDV integration and tumor formation; however, the functions of the sTMR have remained unknown. In this study, we demonstrate that deletion of the sTMR in the MDV genome abrogates virus replication, while extensive mutation of the sTMR does not, indicating that the presence of the sTMR but not the sTMR sequence itself is important. Furthermore, we generated a panel of truncation mutants to determine the minimal length of the sTMR and observed a direct correlation between sTMR length and MDV replication. To address the role of sTMR in MDV replication, integration, and tumorigenesis, sTMR sequences were replaced by a scrambled repeated sequence (vsTMR_mut). vsTMR_mut replicated comparably to parental and revertant viruses in vitro. In vivo, however, a significant reduction in disease and tumor incidence was observed in chickens infected with vsTMR_mut that also correlated with a reduced number of viral integration sites in tumor cells. Taken together, our data demonstrate that the sTMR play a central role in MDV genome replication, pathogenesis, and MDV-induced tumor formation. IMPORTANCE Marek's disease virus (MDV) is a highly oncogenic alphaherpesvirus that infects chickens and causes high economic losses in the poultry industry. MDV integrates its genetic material into host telomeres, a process that is crucial for efficient tumor formation. The MDV genome harbors two arrays of telomeric repeats (TMR) at the ends of its linear genome that are identical to host telomeres and that are termed mTMR and sTMR. mTMR have been recently shown to be involved in MDV integration, while the functions of sTMR remain unknown. Here, we demonstrate that the presence and length of sTMR sequence, but not the exact nucleotide sequence, are crucial for MDV replication. Furthermore, the sTMR contribute to the high integration frequency of MDV and are important for MDV pathogenesis and tumor formation. As a number of herpesviruses harbor arrays of telomeric repeats (TMR), MDV serves as a model to determine the role of the herpesvirus TMR in replication, integration, and pathogenesis.
Collapse
|
20
|
Perumbakkam S, Hunt HD, Cheng HH. Marek's disease virus influences the core gut microbiome of the chicken during the early and late phases of viral replication. FEMS Microbiol Ecol 2014; 90:300-12. [PMID: 25065611 DOI: 10.1111/1574-6941.12392] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 07/17/2014] [Accepted: 07/19/2014] [Indexed: 11/30/2022] Open
Abstract
Marek's disease (MD) is an important neoplastic disease of chickens caused by the Marek's disease virus (MDV), an oncogenic alphaherpesvirus. In this study, dysbiosis induced by MDV on the core gut flora of chicken was assessed using next generation sequence (NGS) analysis. Total fecal and cecum-derived samples from individual birds were used to estimate the influence of MDV infection on the gut microbiome of chicken. Our analysis shows that MDV infection alters the core gut flora in the total fecal samples relatively early after infection (2-7 days) and in the late phase of viral infection (28-35 days) in cecal samples, corresponding well with the life cycle of MDV. Principle component analyses of total fecal and cecal samples showed clustering at the early and late time points, respectively. The genus Lactobacillus was exclusively present in the infected samples in both total fecal and cecal bird samples. The community colonization of core gut flora was altered by viral infection, which manifested in the enrichment of several genera during the early and late phases of MDV replication. The results suggest a relationship between viral infection and microbial composition of the intestinal tract that may influence inflammation and immunosuppression of T and B cells in the host.
Collapse
Affiliation(s)
- Sudeep Perumbakkam
- Avian Diseases and Oncology Laboratory, USDA, ARS, East Lansing, MI, USA; Department of Animal Science, Purdue University, West Lafayette, IN, USA
| | | | | |
Collapse
|
21
|
WaveSeq: a novel data-driven method of detecting histone modification enrichments using wavelets. PLoS One 2012; 7:e45486. [PMID: 23029045 PMCID: PMC3461018 DOI: 10.1371/journal.pone.0045486] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 08/23/2012] [Indexed: 11/19/2022] Open
Abstract
Background Chromatin immunoprecipitation followed by next-generation sequencing is a genome-wide analysis technique that can be used to detect various epigenetic phenomena such as, transcription factor binding sites and histone modifications. Histone modification profiles can be either punctate or diffuse which makes it difficult to distinguish regions of enrichment from background noise. With the discovery of histone marks having a wide variety of enrichment patterns, there is an urgent need for analysis methods that are robust to various data characteristics and capable of detecting a broad range of enrichment patterns. Results To address these challenges we propose WaveSeq, a novel data-driven method of detecting regions of significant enrichment in ChIP-Seq data. Our approach utilizes the wavelet transform, is free of distributional assumptions and is robust to diverse data characteristics such as low signal-to-noise ratios and broad enrichment patterns. Using publicly available datasets we showed that WaveSeq compares favorably with other published methods, exhibiting high sensitivity and precision for both punctate and diffuse enrichment regions even in the absence of a control data set. The application of our algorithm to a complex histone modification data set helped make novel functional discoveries which further underlined its utility in such an experimental setup. Conclusions WaveSeq is a highly sensitive method capable of accurate identification of enriched regions in a broad range of data sets. WaveSeq can detect both narrow and broad peaks with a high degree of accuracy even in low signal-to-noise ratio data sets. WaveSeq is also suited for application in complex experimental scenarios, helping make biologically relevant functional discoveries.
Collapse
|
22
|
Engel AT, Selvaraj RK, Kamil JP, Osterrieder N, Kaufer BB. Marek's disease viral interleukin-8 promotes lymphoma formation through targeted recruitment of B cells and CD4+ CD25+ T cells. J Virol 2012; 86:8536-45. [PMID: 22647701 PMCID: PMC3421702 DOI: 10.1128/jvi.00556-12] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 05/22/2012] [Indexed: 11/20/2022] Open
Abstract
Marek's disease virus (MDV) is a cell-associated and highly oncogenic alphaherpesvirus that infects chickens. During lytic and latent MDV infection, a CXC chemokine termed viral interleukin-8 (vIL-8) is expressed. Deletion of the entire vIL-8 open reading frame (ORF) was shown to severely impair disease progression and tumor development; however, it was unclear whether this phenotype was due to loss of secreted vIL-8 or of splice variants that fuse exons II and III of vIL-8 to certain upstream open reading frames, including the viral oncoprotein Meq. To specifically examine the role of secreted vIL-8 in MDV pathogenesis, we constructed a recombinant virus, vΔMetvIL-8, in which we deleted the native start codon from the signal peptide encoding exon I. This mutant lacked secreted vIL-8 but did not affect Meq-vIL-8 splice variants. Loss of secreted vIL-8 resulted in highly reduced disease and tumor incidence in animals infected with vΔMetvIL-8 by the intra-abdominal route. Although vΔMetvIL-8 was still able to spread to naïve animals by the natural route, infection and lymphomagenesis in contact animals were severely impaired. In vitro assays showed that purified recombinant vIL-8 efficiently binds to and induces chemotaxis of B cells, which are the main target for lytic MDV replication, and also interacts with CD4(+) CD25(+) T cells, known targets of MDV transformation. Our data provide evidence that vIL-8 attracts B and CD4(+) CD25(+) T cells to recruit targets for both lytic and latent infection.
Collapse
Affiliation(s)
| | - Ramesh K. Selvaraj
- Department of Animal Sciences, The Ohio State University, Wooster, Ohio, USA
| | - Jeremy P. Kamil
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA
| | | | | |
Collapse
|
23
|
Yu Y, Luo J, Mitra A, Chang S, Tian F, Zhang H, Yuan P, Zhou H, Song J. Temporal transcriptome changes induced by MDV in Marek's disease-resistant and -susceptible inbred chickens. BMC Genomics 2011; 12:501. [PMID: 21992110 PMCID: PMC3269463 DOI: 10.1186/1471-2164-12-501] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 10/12/2011] [Indexed: 11/10/2022] Open
Abstract
Background Marek's disease (MD) is a lymphoproliferative disease in chickens caused by Marek's disease virus (MDV) and characterized by T cell lymphoma and infiltration of lymphoid cells into various organs such as liver, spleen, peripheral nerves and muscle. Resistance to MD and disease risk have long been thought to be influenced both by genetic and environmental factors, the combination of which contributes to the observed outcome in an individual. We hypothesize that after MDV infection, genes related to MD-resistance or -susceptibility may exhibit different trends in transcriptional activity in chicken lines having a varying degree of resistance to MD. Results In order to study the mechanisms of resistance and susceptibility to MD, we performed genome-wide temporal expression analysis in spleen tissues from MD-resistant line 63, susceptible line 72 and recombinant congenic strain M (RCS-M) that has a phenotype intermediate between lines 63 and 72 after MDV infection. Three time points of the MDV life cycle in chicken were selected for study: 5 days post infection (dpi), 10dpi and 21dpi, representing the early cytolytic, latent and late cytolytic stages, respectively. We observed similar gene expression profiles at the three time points in line 63 and RCS-M chickens that are both different from line 72. Pathway analysis using Ingenuity Pathway Analysis (IPA) showed that MDV can broadly influence the chickens irrespective of whether they are resistant or susceptible to MD. However, some pathways like cardiac arrhythmia and cardiovascular disease were found to be affected only in line 72; while some networks related to cell-mediated immune response and antigen presentation were enriched only in line 63 and RCS-M. We identified 78 and 30 candidate genes associated with MD resistance, at 10 and 21dpi respectively, by considering genes having the same trend of expression change after MDV infection in lines 63 and RCS-M. On the other hand, by considering genes with the same trend of expression change after MDV infection in lines 72 and RCS-M, we identified 78 and 43 genes at 10 and 21dpi, respectively, which may be associated with MD-susceptibility. Conclusions By testing temporal transcriptome changes using three representative chicken lines with different resistance to MD, we identified 108 candidate genes for MD-resistance and 121 candidate genes for MD-susceptibility over the three time points. Genes included in our resistance or susceptibility genes lists that are also involved in more than 5 biofunctions, such as CD8α, IL8, USP18, and CTLA4, are considered to be important genes involved in MD-resistance or -susceptibility. We were also able to identify several biofunctions related with immune response that we believe play an important role in MD-resistance.
Collapse
Affiliation(s)
- Ying Yu
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Analysis of transcriptional activities of the Meq proteins present in highly virulent Marek's disease virus strains, RB1B and Md5. Virus Genes 2011; 43:66-71. [PMID: 21503681 DOI: 10.1007/s11262-011-0612-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Accepted: 04/05/2011] [Indexed: 10/18/2022]
Abstract
Marek's disease virus (MDV) is an oncogenic herpesvirus that causes malignant lymphomas in chickens. Recent field isolates of MDV have tended to exhibit increasing virulence, and MDV strains are currently classified into four categories based on their relative virulence. Meq, a putative MDV oncoprotein, resembles the Jun/Fos family of basic leucine zipper (bZIP) transcription factors and can regulate the expression of viral and cellular genes as a homodimer or as a heterodimer with a variety of bZIP family proteins. MDV isolates display distinct diversity and point mutations in Meq, which may contribute to changes in the transcriptional activities of Meq and subsequently, to observed increases in MDV oncogenicity. In this study, we introduced mutations into the meq gene and used dual luciferase reporter assays to analyze the transcriptional activities of the resulting Meq proteins to determine whether distinct mutations in Meq could be responsible for differences in transcriptional activity among MDV strains. A proline-to-alanine substitution at position 217, the second position of one of the proline direct repeats in the transactivation domain, enhanced the transactivation activity of Meq. In addition, we found that two substitutions at positions 283 and 320 affected transactivation activity. These results suggest that the distinct diversity of and point mutations in the Meq proteins are responsible for differences in transactivation activity among MDV strains.
Collapse
|
25
|
Carvallo FR, French RA, Gilbert-Marcheterre K, Risatti G, Dunn JR, Forster F, Kiupel M, Smyth JA. Mortality of one-week-old chickens during naturally occurring Marek's disease virus infection. Vet Pathol 2011; 48:993-8. [PMID: 21239693 DOI: 10.1177/0300985810395727] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Marek's disease (MD) is a disease of chickens that occurs worldwide and has serious economic consequences. MD can present as one of several forms, with the most commonly occurring forms being the lymphoproliferative diseases. Under experimental conditions, an early mortality syndrome has been recognized following infection by some but not all strains of MD virus (MDV). This is the first report of a confirmed case of mortality due to naturally occurring MDV infection in 1-week-old, nonvaccinated, chickens. Necrotizing lesions were observed in the bursa of Fabricius, lung, duodenum, jejunum, and proventriculus, and large intranuclear inclusion bodies were a striking feature in tissues with lesions in all birds. Immunohistochemical staining for the pp38 protein of MDV revealed abundant pp38 antigen in the affected tissues, confirming the presence of MDV within the lesions. PCR yielded an amplicon with 97% homology to the meq gene of MDV. No evidence of co-infection by either of the immunosuppressive agents chicken anemia virus and infectious bursal disease virus was detected.
Collapse
Affiliation(s)
- F R Carvallo
- Connecticut Veterinary Medical Diagnostic Laboratory, Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06269, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Rajčáni J, Asványi-Molnár N, Szathmary S. Herpesvirus-associated lymphomas: Investigations in humans and animal models. Acta Microbiol Immunol Hung 2010; 57:349-76. [PMID: 21183422 DOI: 10.1556/amicr.57.2010.4.3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Lymphomas are solid tumors consisting of lymphoid cells; they form a heterogeneous group of less or more malignant disorders. A portion of lymphomas develop due to latent herpesvirus infections established in B and/or T-lymphocytes. The basis for latency is a lifelong presence of the circularized covalently linked viral genome within nuclei of carrier lymphocytes. In certain cases, however, the essential event leading to tumor formation is the integration of a portion(s) of viral DNA into the host cell DNA. This leads to rearrangements within the host cell genome on one hand, and, on other hand, to unregulated expression of oncoproteins encoded by the integrated fragment. Our review deals with mechanisms of lymphoma formation regarding to the role of non-structural herpesvirus oncoproteins interfering with the regulation of cell division and/or exerting anti-apoptotic effects. In addition, the authors wish to highlight the common procedures, which allowed isolation and/or identification of lymphoma-associated viruses in cell cultures derived from tumors and/or proliferating lymphatic tissues.
Collapse
Affiliation(s)
- J Rajčáni
- Institute of Virology, Slovak Academy of Sciences, Institute of Virology, 84505 Bratislava, Slovak Republic, Hungary.
| | | | | |
Collapse
|
27
|
Wakenell PS, O'Connell P, Blackmore C, Mondal SP, Schat KA. Role of Marek's disease herpesvirus in the induction of tumours in Japanese quail (Coturnix coturnix japonica) by methylcholanthrene. Avian Pathol 2010; 39:183-8. [PMID: 20544424 DOI: 10.1080/03079451003742918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The QT35 cell line, established from 20-methylcholanthrene (MCA)-induced tumours in Japanese quail, is positive for Marek's disease virus (MDV), and therefore we examined whether MDV is important for the development of MCA-induced tumours. Japanese quail were inoculated with the JM16 strain of MDV at 1 or 3 days of age or left uninoculated. At 3 weeks of age, quail were injected in the breast muscle with 4 mg MCA in corn oil or corn oil alone. Quail were observed for tumours three times/week and at post mortem at 11 to 12 weeks of age. MDV DNA was detected by polymerase chain reaction (PCR) in spleens of 14/20 birds inoculated with JM16+corn oil and of 53/71 birds inoculated with JM16+MCA. Interestingly, 1/74 quail was positive in the MCA group alone for MDV DNA. Tumours were collected for histopathology, cell line development, and PCR and reverse transcriptase-PCR for the presence of MDV. Tumours developed in 38/83 MCA-treated and 32/85 JM16+MCA-treated quail. Fibrosarcomas without metastasis were the only tumours observed in the MCA-treated quail, while quail treated with JM16 and MCA developed undifferentiated tumours, fibrosarcomas, lymphosarcomas or combinations with or without metastasis. One out of 20 quail receiving JM16 alone developed a lymphosarcoma. Cell line development was not influenced by JM16. Tumours from MCA-treated quail were negative for MDV, while 19/29 were positive in the JM16+MCA group. MDV transcripts were present in 13/18 tumours examined in the JM16+MCA group. In conclusion, MDV did not affect tumour development but did influence tumour aggression and histological type.
Collapse
Affiliation(s)
- Patricia S Wakenell
- School of Veterinary Medicine, Department of Population Health and Reproduction, University of California-Davis, Davis, CA 95616, USA.
| | | | | | | | | |
Collapse
|
28
|
Baigent SJ, Davison TF. Development and composition of lymphoid lesions in the spleens of Marek's disease virus-infected chickens: Association with virus spread and the pathogenesis of Marek's disease. Avian Pathol 2010; 28:287-300. [DOI: 10.1080/03079459994786] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
29
|
Baaten BJG, Staines KA, Smith LP, Skinner H, Davison TF, Butter C. Early replication in pulmonary B cells after infection with Marek's disease herpesvirus by the respiratory route. Viral Immunol 2010; 22:431-44. [PMID: 19951180 DOI: 10.1089/vim.2009.0047] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Abstract Natural infection with Marek's disease virus occurs through the respiratory mucosa after chickens inhale dander shed from infected chickens. The early events in the lung following exposure to the feather and squamous epithelial cell debris containing the viral particles remain unclear. In order to elucidate the virological and immunological consequences of MDV infection for the respiratory tract, chickens were infected by intratracheal administration of infective dander. Differences between susceptible and resistant chickens were immediately apparent, with delayed viral replication and earlier onset of interferon (IFN)-gamma production in the latter. CD4(+) and CD8(+) T cells surrounded infected cells in the lung. Although viral replication was evident in macrophages, pulmonary B cells were the main target cell type in susceptible chickens following intratracheal infection with MDV. In accordance, depletion of B cells curtailed viremia and substantially affected pathogenesis in susceptible chickens. Together the data described here demonstrate the role of pulmonary B cells as the primary and predominant target cells and their importance for MDV pathogenesis.
Collapse
Affiliation(s)
- B J G Baaten
- Institute for Animal Health, Compton, Newbury, Berkshire, UK.
| | | | | | | | | | | |
Collapse
|
30
|
Dalgaard T, Boving MK, Handberg K, Jensen KH, Norup LR, Juul-Madsen HR. MHC Expression on Spleen Lymphocyte Subsets in Genetically Resistant and Susceptible Chickens Infected with Marek's Disease Virus. Viral Immunol 2009; 22:321-7. [DOI: 10.1089/vim.2009.0033] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Tina Dalgaard
- Department of Animal Health, Welfare, and Nutrition, University of Aarhus, Tjele, Denmark
| | - Mette K. Boving
- Division of Poultry, Fish, and Fur Animals, National Veterinary Institute, Aarhus, Denmark
| | - Kurt Handberg
- Division of Poultry, Fish, and Fur Animals, National Veterinary Institute, Aarhus, Denmark
| | - Karin H. Jensen
- Department of Animal Health, Welfare, and Nutrition, University of Aarhus, Tjele, Denmark
| | - Liselotte R. Norup
- Department of Animal Health, Welfare, and Nutrition, University of Aarhus, Tjele, Denmark
| | - Helle R. Juul-Madsen
- Department of Animal Health, Welfare, and Nutrition, University of Aarhus, Tjele, Denmark
| |
Collapse
|
31
|
Abdul-Careem MF, Read LR, Parvizi P, Thanthrige-Don N, Sharif S. Marek's disease virus-induced expression of cytokine genes in feathers of genetically defined chickens. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2009; 33:618-623. [PMID: 19041890 DOI: 10.1016/j.dci.2008.11.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2008] [Revised: 10/28/2008] [Accepted: 11/06/2008] [Indexed: 05/27/2023]
Abstract
Marek's disease (MD) vaccines, although effective in reducing lymphoproliferation, cannot control infectious virus production in the feather follicle epithelium (FFE) which is the site of virus shedding. Therefore, we investigated Marek's disease virus (MDV) replication as well as the expression of cytokine genes in feathers of MDV-infected chickens belonging to genetically defined lines (N2a or B(21)/B(21) haplotype-resistant and P2a or B(19)/B(19) haplotype-susceptible). Though there was not a difference in MDV genome load and transcripts between feathers of these chicken lines at 4 and 10 days post-infection (d.p.i.), feathers of resistant chickens carried significantly lower viral genome load and transcripts at 21 d.p.i. Irrespective of genetic background of the chickens examined, MDV replication showed a significant positive correlation with the expression of IFN-gamma gene. The results imply the usefulness of genetic control approach in reducing virulent MDV transmission.
Collapse
Affiliation(s)
- Mohamed Faizal Abdul-Careem
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | | | | | | | | |
Collapse
|
32
|
Parvizi P, Read L, Abdul-Careem MF, Lusty C, Sharif S. Cytokine Gene Expression in Splenic CD4+and CD8+T-Cell Subsets of Chickens Infected with Marek's Disease Virus. Viral Immunol 2009; 22:31-8. [DOI: 10.1089/vim.2008.0062] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Payvand Parvizi
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Leah Read
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | | | - Christopher Lusty
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Shayan Sharif
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
33
|
Abdul-Careem MF, Hunter BD, Lee LF, Fairbrother JH, Haghighi HR, Read L, Parvizi P, Heidari M, Sharif S. Host responses in the bursa of Fabricius of chickens infected with virulent Marek's disease virus. Virology 2008; 379:256-65. [PMID: 18675437 DOI: 10.1016/j.virol.2008.06.027] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2008] [Revised: 05/21/2008] [Accepted: 06/24/2008] [Indexed: 11/29/2022]
Abstract
The bursa of Fabricius serves as an important tissue in the process of Marek's disease virus (MDV) pathogenesis, since B cells of the bursa harbor the cytolytic phase of MDV replication cycle. In the present study, host responses associated with MDV infection in the bursa of Fabricius of chickens were investigated. The expression of MDV phosphoprotein (pp)38 antigen, MDV glycoprotein (gB) and MDV viral interleukin (vIL)-8 transcripts was at the highest at 4 days post-infection (d.p.i.) and then showed a declining trend. On the contrary, the expression of meq (MDV EcoRI Q) gene as well as the viral genome load increased gradually until day 14 post-infection. The changes in viral parameters were associated with significantly higher infiltration of macrophages and T cell subsets, particularly CD4+ T cells into the bursa of Fabricius. Of the genes examined, the expression of interferon (IFN)-alpha, IFN-gamma genes and inducible nitric oxide synthase (iNOS) was significantly up-regulated in response to MDV infection in the bursa of Fabricius. The results suggest a role for these cells and cytokines in MDV-induced responses in the bursa of Fabricius.
Collapse
Affiliation(s)
- M F Abdul-Careem
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Gimeno IM. Marek's disease vaccines: A solution for today but a worry for tomorrow? Vaccine 2008; 26 Suppl 3:C31-41. [DOI: 10.1016/j.vaccine.2008.04.009] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
35
|
Kothlow S, Morgenroth I, Tregaskes CA, Kaspers B, Young JR. CD40 ligand supports the long-term maintenance and differentiation of chicken B cells in culture. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2008; 32:1015-1026. [PMID: 18374414 DOI: 10.1016/j.dci.2008.01.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Revised: 01/25/2008] [Accepted: 01/30/2008] [Indexed: 05/26/2023]
Abstract
TNF family members play crucial roles in mammalian B-cell differentiation and function, many of which have not been demonstrated in other species. To investigate the avian CD40/CD40L system, a chicken CD40 cDNA, obtained by expression screening, was used to raise monoclonal antibodies showing that CD40 was expressed on chicken B cells, monocytes and macrophages, like mammalian CD40. CD40 ligand fusion protein supported the proliferation of B cells in culture for up to 3 weeks, during which they differentiated towards a plasma cell phenotype. CD40L-activated B cells from immunised birds secreted antigen-specific IgM and IgG. These results showed important conserved functions of CD40 and its ligand in mammals and birds. CD40L provides a means for maintenance and differentiation of untransformed chicken B cells in culture, for the first time, allowing new approaches to study of post-bursal B cell biology and host-pathogen interactions with B cell tropic viruses.
Collapse
Affiliation(s)
- Sonja Kothlow
- Institute for Animal Physiology, University of Munich, Veterinärstr. 13, 80539 Munich, Germany.
| | | | | | | | | |
Collapse
|
36
|
Schat K, Calnek B, Weinstock D. Cultivation and characterisation of avian lymphocytes with natural killer cell activity. Avian Pathol 2008; 15:539-56. [DOI: 10.1080/03079458608436314] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
37
|
Venugopal K, Payne LN. Molecular pathogenesis of Marek's disease—recent developments. Avian Pathol 2007; 24:597-609. [DOI: 10.1080/03079459508419100] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
38
|
Baigent SJ, Ross LJN, Davison TF. A flow cytometric method for identifying Marek's disease virus pp38 expression in lymphocyte subpopulations. Avian Pathol 2007; 25:255-67. [DOI: 10.1080/03079459608419140] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
39
|
Jarosinski KW, Schat KA. Multiple alternative splicing to exons II and III of viral interleukin-8 (vIL-8) in the Marek's disease virus genome: the importance of vIL-8 exon I. Virus Genes 2006; 34:9-22. [PMID: 16927116 DOI: 10.1007/s11262-006-0004-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2005] [Accepted: 03/07/2006] [Indexed: 10/24/2022]
Abstract
The Marek's disease virus (MDV) Eco Q (Meq) and the interleukin-8 (IL-8) MDV homologue (vIL-8) genes, and the open reading frames RLORF5a and RLORF4 are encoded within the repeat long (IR(L) and TR(L)) regions of the MDV genome. The recent cloning and characterization of RLORF4 led to the identification of a RLORF4/vIL-8 splice variant using 3' rapid amplification of cDNA ends (RACE). Further characterization of 3'RACE products amplified with primers located within the Meq, RLORF5a, or RLORF4 genes showed the presence of many splice variants. Two novel Meq splice variants were detected, in addition to splice variants encoding portions of RLORF5a and RLORF4 combined with exons II and III of vIL-8 (RLORF5a/vIL-8 and RLORF4/vIL-8, respectively). Analysis of expression in MDV-infected chickens showed that the RLORF5a/vIL-8 and 3 of 4 RLORF4/vIL-8 transcripts were only expressed at 4 days post-infection. Since a number of transcripts encoded vIL-8 exons II and III, this suggested that exon I may be non-essential for vIL-8 function(s). Virus reconstituted from the oncogenic pRB-1B bacterial artificial chromosome with vIL-8 exon I deleted showed decreased early replication and reduced incidence of tumor development, similar to deletion mutants lacking the complete vIL-8 gene.
Collapse
Affiliation(s)
- Keith William Jarosinski
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| | | |
Collapse
|
40
|
Li X, Jarosinski KW, Schat KA. Expression of Marek's disease virus phosphorylated polypeptide pp38 produces splice variants and enhances metabolic activity. Vet Microbiol 2006; 117:154-68. [PMID: 16876339 DOI: 10.1016/j.vetmic.2006.06.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2006] [Revised: 06/19/2006] [Accepted: 06/23/2006] [Indexed: 11/24/2022]
Abstract
The phosphorylated polypeptide (pp)38 of oncogenic Marek's disease (MD) herpesvirus (MDV) is expressed during lytic infections in vivo and in vitro, but its functions have not been fully elucidated. The quail cell line QT-35, latently infected with MDV, was used to generate QTP32 in which pp38 is expressed under control of a tetracycline controlled promoter to examine possible functions of pp38. Induction of pp38 did not influence late MDV genes expression, but it enhanced mitochondrial dehydrogenase activity significantly. Two new pp38 splice variants were found in induced QTP32 cells, in additional in vitro systems and MDV-infected chickens. Differential expression of full-length pp38 and splice variants suggests that the splice variants are important during latency and perhaps transformation. Polypeptides of 40 and 20kDa were detected by Western blot using monoclonal antibody H19. These polypeptides were also produced in DF-1 cells transfected with a pp38 construct in which the splice acceptor sites had been mutated. Our results add important new information to the role of pp38 in the pathogenesis of MD. The data suggest that pp38 and the two newly described splice variants may influence metabolic activity, which may have important consequences for the understanding of latency and tumor development.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Viral/chemistry
- Antigens, Viral/genetics
- Antigens, Viral/metabolism
- Antigens, Viral/physiology
- Blotting, Northern/veterinary
- Cells, Cultured
- Chickens
- DNA, Viral/chemistry
- Gene Expression Regulation, Viral
- Genes, Viral
- Marek Disease/metabolism
- Marek Disease/virology
- Molecular Sequence Data
- Molecular Weight
- Phosphoproteins/chemistry
- Phosphoproteins/genetics
- Phosphoproteins/metabolism
- Phosphoproteins/physiology
- Quail
- RNA Splicing
- RNA, Viral/metabolism
- Reverse Transcriptase Polymerase Chain Reaction/veterinary
- Transfection/veterinary
- Virus Latency
Collapse
Affiliation(s)
- Xinhui Li
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| | | | | |
Collapse
|
41
|
Jarosinski KW, Njaa BL, O'connell PH, Schat KA. Pro-inflammatory Responses in Chicken Spleen and Brain Tissues after Infection with Very Virulent Plus Marek's Disease Virus. Viral Immunol 2005; 18:148-61. [PMID: 15802959 DOI: 10.1089/vim.2005.18.148] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In chickens infected with virulent (v) or very virulent (vv) Marek's disease (MD) virus (MDV) strains, small to moderate increases in plasma nitric oxide (NO) levels are seen, respectively, whereas very virulent plus (vv+) strains induce very high levels in vivo. The data presented in this report show that chickens presenting with clinical neurological disease following infection with the vv+ RK-1 strain have significantly higher in vivo NO levels compared to RK-1-infected non-symptomatic chickens. Using quantitative real-time PCR (qPCR) assays, DNA was used to measure MDV copy numbers in the spleen and brain of P2a (MD-susceptible) and N2a (MD-resistant) chickens following infection with the JM-16 (v) or RK-1 (vv+) strains. RNA was used to measure inducible NO synthase (iNOS), interferon-gamma (IFN-gamma), interleukin (IL)-1beta, IL-6, and IL-8 mRNA levels, in addition to MDV-specific mRNA expression using quantitative RT-PCR (qRT-PCR) assays. Viral DNA loads were found to be considerably higher in RK-1-infected chickens than JM-16-infected chickens at most time points in both organs, with viral copy numbers being two to four logs lower in the brain. Large increases in iNOS, IFN-alpha, IL-1beta, IL-6, and IL-8 were seen in the brains of RK-1-infected chickens. These data strongly support the hypothesis that pro-inflammatory responses, including high levels of iNOS/NO, IFN-alpha, and pro-inflammatory cytokine expression in the chicken brain, may play a major role in the neurological diseases associated with vv+MDV strains.
Collapse
Affiliation(s)
- Keith W Jarosinski
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | | | | |
Collapse
|
42
|
Prigge JT, Majerciak V, Hunt HD, Dienglewicz RL, Parcells MS. Construction and characterization of Marek's disease viruses having green fluorescent protein expression tied directly or indirectly to phosphoprotein 38 expression. Avian Dis 2005; 48:471-87. [PMID: 15529969 DOI: 10.1637/7110] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Marek's disease (MD) is caused by Marek's disease virus (MDV), a highly cell-associated alphaherpesvirus. MD is primarily characterized by lymphocyte infiltration of the nerves and the development of lymphomas in visceral organs, muscle, and skin. MDV encodes two phosphoproteins, pp24 and pp38, that are highly expressed during lytic infection. These proteins were initially identified in MDV-induced tumors but are now known to be linked primarily to MDV lytic infection. Despite the recent characterization of a pp38 deletion mutant MDV, the functions of these phosphoproteins remain unknown. The goal of this work was to construct recombinant MDVs having direct fusions of a marker gene, the green fluorescent protein (GFP), to pp38 in order to study the expression patterns and localization of this protein during stages of MDV infection. We report the construction of two recombinant viruses, one having the enhanced green fluorescent protein (eGFP) fused in-frame to the pp38 open reading frame (ORF) (RB1Bpp38/eGFP) and the other having soluble-modified GFP (smGFP) downstream but out-of-frame with pp38 (RB1Bpp38/smGFP). During construction of RB1Bpp38/eGFP, an ORF located downstream of pp38 (LORF12) was partially deleted. In RB1Bpp38/smGFP, however, LORF12 and its immediate 5' upstream sequence was left intact. This report describes the construction, cell culture, and in vivo characterization of RB1Bpp38/eGFP and RB1Bpp38/smGFP. Structural analysis showed that the virus stocks of RB1Bpp38/eGFP and RB1Bpp38/smGFP had incorporated the GFP cassette and were free of contaminating parent virus (RB1B). Moreover, RB1Bpp38/eGFP and RB1Bpp38/smGFP contained two and three and four and five copies of the 132-bp repeats, respectively. Expression analysis showed that the transcription of genes in RB1Bpp38/eGFP-and RB1Bpp38/smGFP-infected chicken embryo fibroblasts (CEFs) were similar to RB1B-infected CEFs, with the notable exception of deletion of a LORF12-specific transcript in RB1Bpp38/ eGFP-infected cells. In CEFs, RB1Bpp38/eGFP and RB1Bpp38/smGFP showed comparable one-step growth kinetics to parental virus (RB1B). RB1Bpp38/eGFP and RB1Bpp38/smGFP, however, showed quite distinct growth characteristics in vivo. Two independent clones of RB1Bpp38/eGFP were highly attenuated, whereas RB1Bpp38/smGFP exhibited pathogenesis similar to parent virus and retained oncogenicity. Our results suggest that the RB1Bpp38/eGFP phenotype could be due to an interference with an in vivo-specific pp38 function via GFP direct fusion, to the deletion of LORF12, or to a targeting of the immune response to eGFP. Because deletion of pp38 was recently found not to fully attenuate very virulent MDV strain MD-5, it is possible that deletion of LORF12 may be at least partially responsible for the attenuation of RB1Bpp38/eGFP. The construction of these viruses and the establishment of cell lines from RB1Bpp38/smGFP provide useful tools for the study of MDV lyric infection in cell culture and in vivo, in studies of the reactivation of MDV from latency, and in the functional analysis of LORF12.
Collapse
Affiliation(s)
- Jon T Prigge
- Center of Excellence for Poultry Science, Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | | | | | | | | |
Collapse
|
43
|
Yunis R, Jarosinski KW, Schat KA. Association between rate of viral genome replication and virulence of Marek's disease herpesvirus strains. Virology 2004; 328:142-50. [PMID: 15380365 DOI: 10.1016/j.virol.2004.07.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2004] [Revised: 05/27/2004] [Accepted: 07/06/2004] [Indexed: 11/23/2022]
Abstract
The early pathogenesis of Marek's disease virus (MDV) infection is characterized by a lytic infection followed by the induction of latency. Genetically resistant N2a and susceptible P2a chickens were infected with the less virulent JM-16 or the very virulent plus (vv+) RK-1 MDV strains to examine the relationship between virulence and resistance on virus replication during 1-10 days postinfection (dpi) using real-time quantitative polymerase chain reaction (qPCR) and quantitative reverse transcriptase (qRT)-PCR assays. The numbers of copies of the viral DNA or transcripts amplified by these assays were normalized relative to cellular controls and subjected to three-way ANOVA. Viral DNA but not RNA was present in spleens at 1-3 dpi in decreasing quantities, and at 4 dpi, viral DNA started to increase concomitant with the initiation of viral transcription independently of virus strain and genetic resistance. At 6 dpi, JM-16 became latent in resistant N2a and susceptible P2a chickens with low levels of viral transcripts, but transcriptional activity increased in susceptible P2a chickens at 9 and 10 dpi. In contrast, infection with vv+ RK-1 never went into latency in both chicken lines. Viral transcripts were present from 4 to 10 dpi showing a higher and more persistent viral activity that may lead to severe damage to the lymphoid organs resulting in increased immunosuppression and increased incidence of MD. The use of qPCR and qRT-PCR to determine viral DNA load and transcriptional activity may offer an alternative to the current system of pathotyping to characterize new MDV isolates.
Collapse
Affiliation(s)
- Reem Yunis
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, United States
| | | | | |
Collapse
|
44
|
Shamblin CE, Greene N, Arumugaswami V, Dienglewicz RL, Parcells MS. Comparative analysis of Marek’s disease virus (MDV) glycoprotein-, lytic antigen pp38- and transformation antigen Meq-encoding genes: association of meq mutations with MDVs of high virulence. Vet Microbiol 2004; 102:147-67. [PMID: 15327791 DOI: 10.1016/j.vetmic.2004.06.007] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2003] [Revised: 11/06/2003] [Accepted: 06/09/2004] [Indexed: 11/18/2022]
Abstract
Marek's disease (MD) is a highly contagious lymphoproliferative and demyelinating disorder of chickens. MD is caused by Marek's disease virus (MDV), a cell-associated, acute-transforming alphaherpesvirus. For three decades, losses to the poultry industry due to MD have been greatly limited through the use of live vaccines. MDV vaccine strains are comprised of antigenically related, apathogenic MDVs originally isolated from chickens (MDV-2), turkeys (herpesvirus of turkeys, HVT) or attenuated-oncogenic strains of MDV-1 (CVI-988). Since the inception of high-density poultry production and MD vaccination, there have been two discernible increases in the virulence of MDV field strains. Our objectives were to determine if common mutations in the major glycoprotein genes, a major lytic antigen phosphoprotein 38 (pp38) or a major latency/transformation antigen Meq (Marek's EcoRI-Q-encoded protein) were associated with enhanced MDV virulence. To address this, we cloned and sequenced the major surface glycoprotein genes (gB, gC, gD, gE, gH, gI, and gL) of five MDV strains that were representative of the virulent (v), very virulent (vv) and very virulent plus (vv+) pathotypes of MDV. We found no consistent mutations in these genes that correlated strictly with virulence level. The glycoprotein genes most similar among MDV-1, MDV-2 and HVT (gB and gC, approximately 81 and 75%, respectively) were among the most conserved across pathotype. We found mutations mapping to the putative signal cleavage site in the gL genes in four out of eleven vv+MDVs, but this mutation was also identified in one vvMDV (643P) indicating that it did not correlate with enhanced virulence. In further analysis of an additional 12 MDV strains, we found no gross polymorphism in any of the glycoprotein genes. Likewise, by PCR and RFLP analysis, we found no polymorphism at the locus encoding the pp38 gene, an early lytic-phase gene associated with MDV replication. In contrast, we found distinct mutations in the latency and transformation-associated Marek's EcoRI-Q-encoded protein, Meq. In examination of the DNA and deduced amino acid sequence of meq genes from 26 MDV strains (9 m/vMDV, 5 vvMDV and 12 vv+MDVs), we found distinct polymorphism and point mutations that appeared to correlate with virulence. Although a complex trait like MDV virulence is likely to be multigenic, these data describe the first sets of mutations that appear to correlate with MDV virulence. Our conclusion is that since Meq is expressed primarily in the latent/transforming phase of MDV infection, and is not encoded by MDV-2 or HVT vaccine viruses, the evolution of MDV virulence may be due to selection on MDV-host cell interactions during latency and may not be mediated by the immune selection against virus lytic antigens such as the surface glycoproteins.
Collapse
Affiliation(s)
- Christine E Shamblin
- Department of Poultry Science, Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | | | | | | | | |
Collapse
|
45
|
Gimeno IM, Witter RL, Hunt HD, Reddy SM, Reed WM. Biocharacteristics shared by highly protective vaccines against Marek's disease. Avian Pathol 2004; 33:59-68. [PMID: 14681069 DOI: 10.1080/0307945031000163264] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Attenuated serotype 1 Marek's disease virus strains vary widely in their protection properties. This study was conducted to elucidate which biocharacteristics of serotype 1 MDV strains are related with protection. Three pairs of vaccines, each one including a higher protective (HP) vaccine and a lower protective (LP) vaccine originating from the same MDV strain, were studied. Two other highly protective vaccines (RM1 and CVI988/BP5) were also included in the study. Comparison within pairs of vaccines showed that marked differences existed between the HP and the LP vaccines. Compared with LP vaccines, HP vaccines replicated better in vivo. Also, they induced a significant expansion of total T cells and of the helper and cytotoxic T cell lineages (CD45(+)CD3(+), CD4(+)CD8(-), CD4(-)CD8(+)) as well as a marked increase in the expression of the antigens of MhcI and MhcII on T cells. Thus, our results show that in vivo replication and early stimulation of the T-cell lineage are two characteristics shared by HP vaccines. However, comparison among the four HP vaccines that provided protection equal to that of CVI988 (RM1, CVI988/BP5, CVI988 and 648A80) revealed variability, especially regarding in vivo replication. Strains RM1 and CVI988/BP5 showed much stronger replication in vivo than the other two vaccine strains (CVI988 and 648A80). Thus, no single set of characteristics could be used to identify the most protective Marek's disease vaccines, implying, perhaps, that multiple mechanisms may be involved.
Collapse
Affiliation(s)
- Isabel M Gimeno
- USDA-ARS Avian Disease and Oncology Laboratory, East Lansing, MI, USA.
| | | | | | | | | |
Collapse
|
46
|
Barrow AD, Burgess SC, Baigent SJ, Howes K, Nair VK. Infection of macrophages by a lymphotropic herpesvirus: a new tropism for Marek's disease virus. J Gen Virol 2003; 84:2635-2645. [PMID: 13679597 DOI: 10.1099/vir.0.19206-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Marek's disease virus (MDV) is classified as an oncogenic lymphotropic herpesvirus of chickens. MDV productively and cytolytically infects B, alphabetaT and gammadeltaT lymphocytes and latently infects T-helper lymphocytes. The aims of this study were to identify whether MDV infects macrophages in vivo and, if so, whether quantitative differences in macrophage infection are associated with MDV strain virulence. Chickens were infected with either virulent MDV (HPRS-16) or 'hypervirulent' MDV (C12/130). Flow cytometry with monoclonal antibodies recognizing MDV pp38 antigen and leukocyte antigens was used to identify MDV lytically infected cells. Macrophages from HPRS-16- and C12/130-infected chickens were pp38(+). It is demonstrated that macrophages are pp38(+) because they are infected and not because they have phagocytosed MDV antigens, as assessed by confocal microscopy using antibodies recognizing MDV antigens of the three herpesvirus kinetic classes: infected cell protein 4 (ICP4, immediate early), pp38 (early) and glycoprotein B (gB, late). Spleen macrophages from MDV-infected chickens were ICP4(+), pp38(+) and gB(+), and ICP4 had nuclear localization denoting infection. Finally, MDV pp38(+) macrophages had high inherent death rates, confirming cytolytic MDV infection, although production of virus particles has not been detected yet. These results have two fundamental implications for understanding MDV pathogenesis: (i) MDV evolved to perturb innate, in addition to acquired, immunity and (ii) macrophages are excellent candidates for transporting MDV to primary lymphoid organs during the earliest stages of pathogenesis.
Collapse
Affiliation(s)
- Alexander D Barrow
- Viral Oncogenesis Group, Institute for Animal Health, Compton, Berkshire RG20 7NN, UK
| | - Shane C Burgess
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Box 6100, MS 39762-6100, USA
| | - Susan J Baigent
- Viral Oncogenesis Group, Institute for Animal Health, Compton, Berkshire RG20 7NN, UK
| | - Ken Howes
- Viral Oncogenesis Group, Institute for Animal Health, Compton, Berkshire RG20 7NN, UK
| | - Venugopal K Nair
- Viral Oncogenesis Group, Institute for Animal Health, Compton, Berkshire RG20 7NN, UK
| |
Collapse
|
47
|
Parcells MS, Arumugaswami V, Prigge JT, Pandya K, Dienglewicz RL. Marek's disease virus reactivation from latency: changes in gene expression at the origin of replication. Poult Sci 2003; 82:893-8. [PMID: 12817443 DOI: 10.1093/ps/82.6.893] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Marek's disease is a contagious lymphoma of chickens caused by Marek's disease virus (MDV). MDV replicates in chicken lymphocytes and establishes latency within and transforms chicken CD4+ T-cells. Transformed T-cells are seen as skin leukosis or as lymphomas in visceral organs. A major focus of our laboratory is the functional study of genes flanking the origin of replication. This origin (OriLyt) is contained within the repeats flanking the unique long (UL) region of the genome (IRL and TRL). To the left of this Ori are genes associated with MDV latent/transforming infection [1.8-kb RNA family, pp14, Meq), and to the right (UL) are genes associated with early stages of MDV lytic infection [BamHI-H-encoded protein (Hep), pp38/pp24, Mys]. During latency, MDV suppresses lytic gene expression and has evolved mechanisms for blocking the apoptosis of latently-infected CD4+ T-cells. Of the genes expressed during MDV latency and in the transformed cell, the Meq (Marek's EcoRI-Q-encoded protein) has been shown to block apoptosis and transactivate gene expression. Upon reactivation to lytic infection, we have found that splice variants of Meq predominate and that these forms lack several of the domains important to Meq trans-activation and trans-repression. We have found that rightward from the origin of replication, a family genes, including phosphoprotein 38 (pp38) are expressed during early stages of reactivation. Three separate open reading frames (Hep, Mys, and pp38) are encoded by distinct transcripts from this region. We are now determining the kinetics of expression of these transcripts and their relative abundance during reactivation.
Collapse
Affiliation(s)
- M S Parcells
- Department of Poultry Science, University of Arkansas, Fayetteville, Arkansas 72701, USA.
| | | | | | | | | |
Collapse
|
48
|
Jarosinski KW, O'Connell PH, Schat KA. Impact of deletions within the Bam HI-L fragment of attenuated Marek's disease virus on vIL-8 expression and the newly identified transcript of open reading frame LORF4. Virus Genes 2003; 26:255-69. [PMID: 12876454 DOI: 10.1023/a:1024447230464] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Marek's disease (MD) in chickens is caused by MD herpesvirus (MDV), which induces T cell lymphomas. The early pathogenesis of MDV infection is characterized by a primary infection in B lymphocytes followed by infection of activated T lymphocytes. It has been speculated that a MDV-encoded homologue of interleukin-8 (vIL-8) may be important to attract activated T lymphocytes to infected B lymphocytes. Recently, more virulent strains of MDV have emerged, named very virulent plus (vv+)MDV, that cause earlier and more prolonged cytolytic infections compared to less virulent strains. In this report, it was found that vIL-8 mRNA expression in vivo was increased in very virulent (vv) and vv+MDV strains compared to mild (m) and virulent (v) strains, and could not be detected in two attenuated MDV strains examined using very sensitive real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR) assays. In order to identify potential mechanisms for the increased vIL-8 mRNA expression in more virulent strains, and lack thereof in attenuated strains, the vIL-8 gene and putative promoter sequences upstream of the vIL-8 gene were compared from 10 different MDV strains, including attenuated derivatives. Only the JM-16 strain (both non-attenuated and attenuated) and attenuated 584A (584Ap80C) encoded a predicted vIL-8 gene sequence different from all other strains examined. Within the putative vIL-8 gene promoter sequence, there was little difference among the non-attenuated strains; however significant deletions were identified in the attenuated JM-16/p71, Md11 (R2/23), and 584Ap80C strains. Additionally, these deletions were located within a previously hypothetical open reading frame (ORF) named LORF4. Rapid amplification of cDNA ends identified a full-length transcript of LORF4 in the MDV-transformed lymphoblastoid cell line MSB-1, and deletions within this ORF caused truncated predicted proteins in 4 out of 6 attenuated MDV strains examined.
Collapse
Affiliation(s)
- Keith William Jarosinski
- Unit of Avian Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| | | | | |
Collapse
|
49
|
Jarosinski KW, Yunis R, O'Connell PH, Markowski-Grimsrud CJ, Schat KA. Influence of genetic resistance of the chicken and virulence of Marek's disease virus (MDV) on nitric oxide responses after MDV infection. Avian Dis 2003; 46:636-49. [PMID: 12243528 DOI: 10.1637/0005-2086(2002)046[0636:iogrot]2.0.co;2] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Nitric oxide (NO), a free radical produced by the enzyme NO synthase (NOS), is a potent antiviral agent in addition to having immune regulating functions. Recently, it was reported that chickens resistant (N2a, MHC: B21B21) to the development of Marek's disease (MD) had a greater potential to produce NO than MD-susceptible chickens (P2a, MHC: B19B19). This difference was shown by measuring NO levels in chick embryo fibroblast cultures obtained from these chickens after treatment with lipopolysaccharide and recombinant chicken interferon-gamma (IFN-gamma). To extend these results, the levels of NO in blood plasma from N2a and P2a chickens inoculated with the nonattenuated JM-16 strain of MD virus (MDV) were examined. In four out of five experiments, N2a chickens had increased NO levels at 7 days postinoculation (DPI). In contrast, P2a chickens challenged with JM-16 had a significant increase in NO in only one of four experiments, and in that experiment the increase was delayed (10 DPI) compared with N2a chickens. Attenuation abrogated MDV-induced NO in chickens. Inoculation with MDV strains ranging from mild to very virulent plus showed that the more virulent strains induced the highest level of NO in blood plasma, suggesting a role of NO in the pathogenesis of MD with more virulent strains. On the basis of quantitative real-time reverse transcription-polymerase chain reaction (RT-PCR) assays for analysis of mRNA expression, IFN-gamma does not appear to be the primary inducer of inducible (i)NOS gene expression during MDV infection. iNOS gene expression and NO production are mediated during the cytolytic phase of MDV infection on the basis of real-time RT-PCR assays with primers specific for glycoprotein B, a late gene expressed only during the cytolytic phase of MDV infection. These findings implicate NO as a factor potentially involved in increasing virulence of MDV, possibly through immune suppression.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Chick Embryo
- Chickens
- DNA, Complementary/genetics
- Disease Susceptibility/immunology
- Fibroblasts/immunology
- Gene Expression Regulation, Enzymologic
- Herpesvirus 2, Gallid/classification
- Herpesvirus 2, Gallid/immunology
- Herpesvirus 2, Gallid/pathogenicity
- Immunity, Innate/genetics
- Immunity, Innate/immunology
- Interferon-gamma/biosynthesis
- Interferon-gamma/genetics
- Marek Disease/genetics
- Marek Disease/immunology
- Nitric Oxide/biosynthesis
- Nitric Oxide/blood
- Nitric Oxide Synthase/genetics
- Nitric Oxide Synthase/metabolism
- RNA, Messenger/biosynthesis
- Reverse Transcriptase Polymerase Chain Reaction/veterinary
- Sequence Analysis, DNA
- Specific Pathogen-Free Organisms
- Spleen/immunology
- Virulence
Collapse
Affiliation(s)
- K W Jarosinski
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | |
Collapse
|
50
|
Kaiser P, Underwood G, Davison F. Differential cytokine responses following Marek's disease virus infection of chickens differing in resistance to Marek's disease. J Virol 2003; 77:762-8. [PMID: 12477883 PMCID: PMC140586 DOI: 10.1128/jvi.77.1.762-768.2003] [Citation(s) in RCA: 162] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2002] [Accepted: 09/23/2002] [Indexed: 12/23/2022] Open
Abstract
The production of cytokine mRNAs, in addition to viral DNA, was quantified by real-time quantitative reverse transcription-PCR (RT-PCR) (cytokines) or PCR (virus) in splenocytes during the course of Marek's disease virus (MDV) infection in four inbred chicken lines: two resistant (lines 6(1) and N) and two susceptible (lines 7(2) and P). Virus loads were only different after 10 days postinfection (dpi), increasing in susceptible lines and decreasing in resistant lines. Gamma interferon (IFN-gamma) mRNA was expressed by splenocytes from all infected birds between 3 and 10 dpi, associated with increasing MDV loads. For other cytokines, differences between lines were only seen for interleukin-6 (IL-6) and IL-18, with splenocytes from susceptible birds expressing high levels of both transcripts during the cytolytic phase of infection, whereas splenocytes from resistant birds expressed neither transcript. These results indicate that these two cytokines could play a crucial role in driving immune responses, which in resistant lines maintain MDV latency but in susceptible lines result in lymphomas.
Collapse
Affiliation(s)
- Pete Kaiser
- Institute for Animal Health, Compton, Berkshire RG20 7NN, United Kingdom.
| | | | | |
Collapse
|