1
|
Xia J, Lin L, Ju R, Xu C, Mo G, Zhang X. GH inhibits ALV-J replication and restricts cell cycle by activating PI3K/Akt signaling pathway. Poult Sci 2024; 104:104514. [PMID: 39586129 PMCID: PMC11625326 DOI: 10.1016/j.psj.2024.104514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/23/2024] [Accepted: 11/04/2024] [Indexed: 11/27/2024] Open
Abstract
Growth hormone (GH) plays a crucial role in growth, sexual maturity, and immunity in chickens. Avian leukosis virus subgroup J (ALV-J) is an exogenous tumorigenic retrovirus that primarily induces immunosuppression, growth retardation, decreased egg production, tumors formation, and even death in chickens. Previous studies have suggested that GH is involved in the regulation of innate immunity and inflammation. However, the specific role of GH in response to ALV-J remains unclear. In this study, we observed a significant upregulation of GH protein expression in the plasma of ALV infected chickens, and a marked increase in GH mRNA in ALV-J infected cells. We found that lower gp85 expression correlated with higher GH expression in immune tissues, suggesting that GH may inhibit gp85 expression. Additionally, GH overexpression enhanced the expression of interferons (IFN-α, IFN-β), interferon-stimulating genes (Mx1, ASCL1, CH25H), and pro-inflammatory factors (Mx1, ASCL1, CH25H) in DF-1 cells infected with ALV-J. GH also affected the cell cycle by regulating the expression of cell proliferation-related genes (p21, PCNA, Cyclin B2, Cyclin D1, Cyclin D2) and cell apoptosis-related genes (p53, Fas, Cyct, Caspase-1, Caspase-3, Caspase-8). More importantly, we found that GH restricted cell proliferation and apoptosis, and inhibited the replication of ALV-J by activating the PI3K/Akt signaling pathway in DF-1 cells. In conclusion, these results indicate GH plays a role in the antiviral response against the replication of ALV-J, providing evidence of an interaction between GH and the innate immunity in chickens.
Collapse
Affiliation(s)
- Junliang Xia
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, Guangzhou, Guangdong, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou, PR China
| | - Ling Lin
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, Guangzhou, Guangdong, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou, PR China
| | - Rongyang Ju
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, Guangzhou, Guangdong, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou, PR China
| | - Chengxun Xu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, Guangzhou, Guangdong, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou, PR China
| | - Guodong Mo
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, Guangzhou, Guangdong, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou, PR China; Guangxi Vocational University of Agriculture, Nanning, 530007 Guangxi, PR China
| | - Xiquan Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, Guangzhou, Guangdong, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou, PR China.
| |
Collapse
|
2
|
Abstract
The different technology platforms used to make poultry vaccines are reviewed. Vaccines based on classical technologies are either live attenuated or inactivated vaccines. Genetic engineering is applied to design by deletion, mutation, insertion, or chimerization, genetically modified target microorganisms that are used either as live or inactivated vaccines. Other vaccine platforms are based on one or a few genes of the target pathogen agent coding for proteins that can induce a protective immune response ("protective genes"). These genes can be expressed in vitro to produce subunit vaccines. Alternatively, vectors carrying these genes in their genome or nucleic acid-based vaccines will induce protection by in vivo expression of these genes in the vaccinated host. Properties of these different types of vaccines, including advantages and limitations, are reviewed, focusing mainly on vaccines targeting viral diseases and on technologies that succeeded in market authorization.
Collapse
|
3
|
Romanov MN, Abdelmanova AS, Fisinin VI, Gladyr EA, Volkova NA, Koshkina OA, Rodionov AN, Vetokh AN, Gusev IV, Anshakov DV, Stanishevskaya OI, Dotsev AV, Griffin DK, Zinovieva NA. Selective footprints and genes relevant to cold adaptation and other phenotypic traits are unscrambled in the genomes of divergently selected chicken breeds. J Anim Sci Biotechnol 2023; 14:35. [PMID: 36829208 PMCID: PMC9951459 DOI: 10.1186/s40104-022-00813-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 11/27/2022] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND The genomes of worldwide poultry breeds divergently selected for performance and other phenotypic traits may also be affected by, and formed due to, past and current admixture events. Adaptation to diverse environments, including acclimation to harsh climatic conditions, has also left selection footprints in breed genomes. RESULTS Using the Chicken 50K_CobbCons SNP chip, we genotyped four divergently selected breeds: two aboriginal, cold tolerant Ushanka and Orloff Mille Fleur, one egg-type Russian White subjected to artificial selection for cold tolerance, and one meat-type White Cornish. Signals of selective sweeps were determined in the studied breeds using three methods: (1) assessment of runs of homozygosity islands, (2) FST based population differential analysis, and (3) haplotype differentiation analysis. Genomic regions of true selection signatures were identified by two or more methods or in two or more breeds. In these regions, we detected 540 prioritized candidate genes supplemented them with those that occurred in one breed using one statistic and were suggested in other studies. Amongst them, SOX5, ME3, ZNF536, WWP1, RIPK2, OSGIN2, DECR1, TPO, PPARGC1A, BDNF, MSTN, and beta-keratin genes can be especially mentioned as candidates for cold adaptation. Epigenetic factors may be involved in regulating some of these important genes (e.g., TPO and BDNF). CONCLUSION Based on a genome-wide scan, our findings can help dissect the genetic architecture underlying various phenotypic traits in chicken breeds. These include genes representing the sine qua non for adaptation to harsh environments. Cold tolerance in acclimated chicken breeds may be developed following one of few specific gene expression mechanisms or more than one overlapping response known in cold-exposed individuals, and this warrants further investigation.
Collapse
Affiliation(s)
- Michael N. Romanov
- L.K. Ernst Federal Research Centre for Animal Husbandry, Dubrovitsy, Podolsk, Moscow Region Russia ,grid.9759.20000 0001 2232 2818School of Biosciences, University of Kent, Canterbury, UK
| | - Alexandra S. Abdelmanova
- L.K. Ernst Federal Research Centre for Animal Husbandry, Dubrovitsy, Podolsk, Moscow Region Russia
| | - Vladimir I. Fisinin
- grid.4886.20000 0001 2192 9124Federal State Budget Scientific Institution Federal Research Centre “All-Russian Poultry Research and Technological Institute” of the Russian Academy of Sciences, Sergiev Posad, Moscow Region Russia
| | - Elena A. Gladyr
- L.K. Ernst Federal Research Centre for Animal Husbandry, Dubrovitsy, Podolsk, Moscow Region Russia
| | - Natalia A. Volkova
- L.K. Ernst Federal Research Centre for Animal Husbandry, Dubrovitsy, Podolsk, Moscow Region Russia
| | - Olga A. Koshkina
- L.K. Ernst Federal Research Centre for Animal Husbandry, Dubrovitsy, Podolsk, Moscow Region Russia
| | - Andrey N. Rodionov
- L.K. Ernst Federal Research Centre for Animal Husbandry, Dubrovitsy, Podolsk, Moscow Region Russia
| | - Anastasia N. Vetokh
- L.K. Ernst Federal Research Centre for Animal Husbandry, Dubrovitsy, Podolsk, Moscow Region Russia
| | - Igor V. Gusev
- L.K. Ernst Federal Research Centre for Animal Husbandry, Dubrovitsy, Podolsk, Moscow Region Russia
| | - Dmitry V. Anshakov
- grid.4886.20000 0001 2192 9124Breeding and Genetic Centre “Zagorsk Experimental Breeding Farm” – Branch of the Federal Research Centre “All-Russian Poultry Research and Technological Institute” of the Russian Academy of Sciences, Sergiev Posad, Moscow Region Russia
| | - Olga I. Stanishevskaya
- grid.473314.6Russian Research Institute of Farm Animal Genetics and Breeding – Branch of the L.K. Ernst Federal Research Centre for Animal Husbandry, St. Petersburg, Russia
| | - Arsen V. Dotsev
- L.K. Ernst Federal Research Centre for Animal Husbandry, Dubrovitsy, Podolsk, Moscow Region Russia
| | - Darren K. Griffin
- grid.9759.20000 0001 2232 2818School of Biosciences, University of Kent, Canterbury, UK
| | - Natalia A. Zinovieva
- L.K. Ernst Federal Research Centre for Animal Husbandry, Dubrovitsy, Podolsk, Moscow Region Russia
| |
Collapse
|
4
|
Gul H, Habib G, Khan IM, Rahman SU, Khan NM, Wang H, Khan NU, Liu Y. Genetic resilience in chickens against bacterial, viral and protozoal pathogens. Front Vet Sci 2022; 9:1032983. [PMID: 36439341 PMCID: PMC9691405 DOI: 10.3389/fvets.2022.1032983] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/28/2022] [Indexed: 06/13/2024] Open
Abstract
The genome contributes to the uniqueness of an individual breed, and enables distinctive characteristics to be passed from one generation to the next. The allelic heterogeneity of a certain breed results in a different response to a pathogen with different genomic expression. Disease resistance in chicken is a polygenic trait that involves different genes that confer resistance against pathogens. Such resistance also involves major histocompatibility (MHC) molecules, immunoglobulins, cytokines, interleukins, T and B cells, and CD4+ and CD8+ T lymphocytes, which are involved in host protection. The MHC is associated with antigen presentation, antibody production, and cytokine stimulation, which highlight its role in disease resistance. The natural resistance-associated macrophage protein 1 (Nramp-1), interferon (IFN), myxovirus-resistance gene, myeloid differentiation primary response 88 (MyD88), receptor-interacting serine/threonine kinase 2 (RIP2), and heterophile cells are involved in disease resistance and susceptibility of chicken. Studies related to disease resistance genetics, epigenetics, and quantitative trait loci would enable the identification of resistance markers and the development of disease resistance breeds. Microbial infections are responsible for significant outbreaks and have blighted the poultry industry. Breeding disease-resistant chicken strains may be helpful in tackling pathogens and increasing the current understanding on host genetics in the fight against communicable diseases. Advanced technologies, such as the CRISPR/Cas9 system, whole genome sequencing, RNA sequencing, and high-density single nucleotide polymorphism (SNP) genotyping, aid the development of resistant breeds, which would significantly decrease the use of antibiotics and vaccination in poultry. In this review, we aimed to reveal the recent genetic basis of infection and genomic modification that increase resistance against different pathogens in chickens.
Collapse
Affiliation(s)
- Haji Gul
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Gul Habib
- Department of Microbiology, Abbottabad University of Science and Technology, Abbottabad, Pakistan
| | - Ibrar Muhammad Khan
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Sajid Ur Rahman
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Nazir Muhammad Khan
- Department of Zoology, University of Science and Technology, Bannu, Pakistan
| | - Hongcheng Wang
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Najeeb Ullah Khan
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, Pakistan
| | - Yong Liu
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| |
Collapse
|
5
|
Mo G, Hu B, Wei P, Luo Q, Zhang X. The Role of Chicken Prolactin, Growth Hormone and Their Receptors in the Immune System. Front Microbiol 2022; 13:900041. [PMID: 35910654 PMCID: PMC9331192 DOI: 10.3389/fmicb.2022.900041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022] Open
Abstract
Prolactin (PRL) and growth hormone (GH) exhibit important roles in the immune system maintenance. In poultry, PRL mainly plays its roles in nesting, hatching, and reproduction, while GH is primarily responding to body weight, fat formation and feed conversion. In this review, we attempt to provide a critical overview of the relationship between PRL and GH, PRLR and GHR, and the immune response of poultry. We also propose a hypothesis that PRL, GH and their receptors might be used by viruses as viral receptors. This may provide new insights into the pathogenesis of viral infection and host immune response.
Collapse
Affiliation(s)
- Guodong Mo
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, South China Agricultural University, Guangzhou, China
| | - Bowen Hu
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, South China Agricultural University, Guangzhou, China
| | - Ping Wei
- Institute for Poultry Science and Health, Guangxi University, Nanning, China
| | - Qingbin Luo
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, South China Agricultural University, Guangzhou, China
| | - Xiquan Zhang
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, South China Agricultural University, Guangzhou, China
| |
Collapse
|
6
|
Bertzbach LD, Tregaskes CA, Martin RJ, Deumer US, Huynh L, Kheimar AM, Conradie AM, Trimpert J, Kaufman J, Kaufer BB. The Diverse Major Histocompatibility Complex Haplotypes of a Common Commercial Chicken Line and Their Effect on Marek's Disease Virus Pathogenesis and Tumorigenesis. Front Immunol 2022; 13:908305. [PMID: 35693787 PMCID: PMC9186122 DOI: 10.3389/fimmu.2022.908305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/29/2022] [Indexed: 02/05/2023] Open
Abstract
The major histocompatibility complex (MHC) is crucial for appropriate immune responses against invading pathogens. Chickens possess a single predominantly-expressed class I molecule with strong associations between disease resistance and MHC haplotype. For Marek's disease virus (MDV) infections of chickens, the MHC haplotype is one of the major determinants of genetic resistance and susceptibility. VALO specific pathogen free (SPF) chickens are widely used in biomedical research and vaccine production. While valuable findings originate from MDV infections of VALO SPF chickens, their MHC haplotypes and associated disease resistance remained elusive. In this study, we used several typing systems to show that VALO SPF chickens possess MHC haplotypes that include B9, B9:02, B15, B19 and B21 at various frequencies. Moreover, we associate the MHC haplotypes to MDV-induced disease and lymphoma formation and found that B15 homozygotes had the lowest tumor incidence while B21 homozygotes had the lowest number of organs with tumors. Finally, we found transmission at variable levels to all contact birds except B15/B21 heterozygotes. These data have immediate implications for the use of VALO SPF chickens and eggs in the life sciences and add another piece to the puzzle of the chicken MHC complex and its role in infections with this oncogenic herpesvirus.
Collapse
Affiliation(s)
| | - Clive A. Tregaskes
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Rebecca J. Martin
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | | | - Lan Huynh
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Ahmed M. Kheimar
- Institute of Virology, Freie Universität Berlin, Berlin, Germany
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Sohag University, Sohag, Egypt
| | | | - Jakob Trimpert
- Institute of Virology, Freie Universität Berlin, Berlin, Germany
| | - Jim Kaufman
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Benedikt B. Kaufer
- Institute of Virology, Freie Universität Berlin, Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
7
|
Exploration of Alternative Splicing (AS) Events in MDV-Infected Chicken Spleens. Genes (Basel) 2021; 12:genes12121857. [PMID: 34946806 PMCID: PMC8701255 DOI: 10.3390/genes12121857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 11/16/2022] Open
Abstract
Marek’s disease (MD) was an immunosuppression disease induced by Marek’s disease virus (MDV). MD caused huge economic loss to the global poultry industry, but it also provided an ideal model for studying diseases induced by the oncogenic virus. Alternative splicing (AS) simultaneously produced different isoform transcripts, which are involved in various diseases and individual development. To investigate AS events in MD, RNA-Seq was performed in tumorous spleens (TS), spleens from the survivors (SS) without any lesion after MDV infection, and non-infected chicken spleens (NS). In this study, 32,703 and 25,217 AS events were identified in TS and SS groups with NS group as the control group, and 1198, 1204, and 348 differently expressed (DE) AS events (p-value < 0.05 and FDR < 0.05) were identified in TS vs. NS, TS vs. SS, SS vs. NS, respectively. Additionally, Function enrichment analysis showed that ubiquitin-mediated proteolysis, p53 signaling pathway, and phosphatidylinositol signaling system were significantly enriched (p-value < 0.05). Small structural variations including SNP and indel were analyzed based on RNA-Seq data, and it showed that the TS group possessed more variants on the splice site region than those in SS and NS groups, which might cause more AS events in the TS group. Combined with previous circRNA data, we found that 287 genes could produce both circular and linear RNAs, which suggested these genes were more active in MD lymphoma transformation. This study has expanded the understanding of the MDV infection process and provided new insights for further analysis of resistance/susceptibility mechanisms.
Collapse
|
8
|
Ellington C, Cortes AL, Faiz NM, Mays JK, Fadly A, Silva RF, Gimeno IM. Characterization of Md5-BAC-REV-LTR virus as Marek's disease vaccine in commercial meat-type chickens: protection and immunosuppression. Avian Pathol 2021; 50:490-499. [PMID: 34463588 DOI: 10.1080/03079457.2021.1970108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Md5-BAC-REV-LTR is a recombinant Marek's disease virus (MDV), with an insertion of the long terminal repeat (LTR) of reticuloendotheliosis virus (REV) into the genome of the highly virulent MDV strain rMd5. It has been shown that Md5-BAC-REV-LTR does not induce tumours and confers high protection against challenge with MDV in 15 × 7 chickens. The objective of the present study was to evaluate the protection and safety (in terms of oncogenicity and immunosuppression) of Md5-BAC-REV-LTR in commercial meat-type chickens bearing maternal antibodies against MDV. Our results show that sub-cutaneous administration of Md5-BAC-REV-LTR at 1 day of age conferred high protection (protection index PI = 84.2) against an early challenge (1 day) by contact exposure to shedder birds infected with the vv+ MDV 648A strain. In such stringent challenge conditions, Md5-BAC-REV-LTR was more protective than a commercial CVI988 (PI = 12.4) and similar to the experimental vaccine Md5-BACΔmeq (PI = 92.4). Furthermore, Md5-BAC-REV-LTR did not induce either tumours or immunosuppression in this study. Immunosuppression was evaluated by the relative lymphoid organ weights and also by the ability of the vaccine to induce late-MDV-induced immunosuppression associated with reactivation of the virus. This study shows that Md5-BAC-REV-LTR has the potential to be used as a MD vaccine and is highly protective against early challenge with vv+ MDV. RESEARCH HIGHLIGHTSMd5-BAC-REV-LTR is highly protective against early challenge with vv+ MDV in commercial meat-type chickens.Md5-BAC-REV-LTR does not cause early immunosuppression.Md5-BAC-REV-LTR does not cause late immunosuppression.Unlike other serotype 1 vaccines, Md5-BAC-REV-LTR is not detected in feather pulp at 7 days post vaccination.
Collapse
Affiliation(s)
- C Ellington
- Department of Population Health and Pathobiology, Veterinary School, North Carolina State University, Raleigh, NC, USA
| | - A L Cortes
- Department of Population Health and Pathobiology, Veterinary School, North Carolina State University, Raleigh, NC, USA
| | - N M Faiz
- Department of Population Health and Pathobiology, Veterinary School, North Carolina State University, Raleigh, NC, USA.,Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Malaysia
| | - J K Mays
- USDA-ARS Avian Disease and Oncology Laboratory, East Lansing, MI, USA
| | - Aly Fadly
- USDA-ARS Avian Disease and Oncology Laboratory, East Lansing, MI, USA
| | - Robert F Silva
- USDA-ARS Avian Disease and Oncology Laboratory, East Lansing, MI, USA
| | - I M Gimeno
- Department of Population Health and Pathobiology, Veterinary School, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
9
|
Latest Insights into Unique Open Reading Frames Encoded by Unique Long (UL) and Short (US) Regions of Marek's Disease Virus. Viruses 2021; 13:v13060974. [PMID: 34070255 PMCID: PMC8225041 DOI: 10.3390/v13060974] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 12/27/2022] Open
Abstract
Marek’s disease virus (MDV) is an oncogenic avian alphaherpesvirus whose genome consists of unique long (UL) and short (US) regions that are flanked by inverted repeat regions. More than 100 open reading frames (ORFs) have been annotated in the MDV genome, and are involved in various aspects of MDV biology and pathogenesis. Within UL and US regions of MDV, there are several unique ORFs, some of which have recently been shown to be important for MDV replication and pathogenesis. In this review, we will summarize the current knowledge on these ORFs and compare their location in different MDV strains.
Collapse
|
10
|
Smith J, Lipkin E, Soller M, Fulton JE, Burt DW. Mapping QTL Associated with Resistance to Avian Oncogenic Marek's Disease Virus (MDV) Reveals Major Candidate Genes and Variants. Genes (Basel) 2020; 11:genes11091019. [PMID: 32872585 PMCID: PMC7564597 DOI: 10.3390/genes11091019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 08/20/2020] [Accepted: 08/26/2020] [Indexed: 01/13/2023] Open
Abstract
Marek’s disease (MD) represents a significant global economic and animal welfare issue. Marek’s disease virus (MDV) is a highly contagious oncogenic and highly immune-suppressive α-herpes virus, which infects chickens, causing neurological effects and tumour formation. Though partially controlled by vaccination, MD continues to have a profound impact on animal health and on the poultry industry. Genetic selection provides an alternative and complementary method to vaccination. However, even after years of study, the genetic mechanisms underlying resistance to MDV remain poorly understood. The Major Histocompatability Complex (MHC) is known to play a role in disease resistance, along with a handful of other non-MHC genes. In this study, one of the largest to date, we used a multi-facetted approach to identify quantitative trait locus regions (QTLR) influencing resistance to MDV, including an F6 population from a full-sib advanced intercross line (FSIL) between two elite commercial layer lines differing in resistance to MDV, RNA-seq information from virus challenged chicks, and genome wide association study (GWAS) from multiple commercial lines. Candidate genomic elements residing in the QTLR were further tested for association with offspring mortality in the face of MDV challenge in eight pure lines of elite egg-layer birds. Thirty-eight QTLR were found on 19 chicken chromosomes. Candidate genes, microRNAs, long non-coding RNAs and potentially functional mutations were identified in these regions. Association tests were carried out in 26 of the QTLR, using eight pure lines of elite egg-layer birds. Numerous candidate genomic elements were strongly associated with MD resistance. Genomic regions significantly associated with resistance to MDV were mapped and candidate genes identified. Various QTLR elements were shown to have a strong genetic association with resistance. These results provide a large number of significant targets for mitigating the effects of MDV infection on both poultry health and the economy, whether by means of selective breeding, improved vaccine design, or gene-editing technologies.
Collapse
Affiliation(s)
- Jacqueline Smith
- The Roslin Institute and Royal (Dick) School of Veterinary Studies R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Ehud Lipkin
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 91904, Israel
| | - Morris Soller
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 91904, Israel
| | - Janet E Fulton
- Hy-Line International, P.O. Box 310, 2583 240th St., Dallas Center, IA 50063, USA
| | - David W Burt
- The Roslin Institute and Royal (Dick) School of Veterinary Studies R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| |
Collapse
|
11
|
|
12
|
Chakraborty P, Kuo R, Vervelde L, Dutia BM, Kaiser P, Smith J. Macrophages from Susceptible and Resistant Chicken Lines have Different Transcriptomes following Marek's Disease Virus Infection. Genes (Basel) 2019; 10:genes10020074. [PMID: 30678299 PMCID: PMC6409778 DOI: 10.3390/genes10020074] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/10/2019] [Accepted: 01/21/2019] [Indexed: 12/12/2022] Open
Abstract
Despite successful control by vaccination, Marek’s disease (MD) has continued evolving to greater virulence over recent years. To control MD, selection and breeding of MD-resistant chickens might be a suitable option. MHC-congenic inbred chicken lines, 61 and 72, are highly resistant and susceptible to MD, respectively, but the cellular and genetic basis for these phenotypes is unknown. Marek’s disease virus (MDV) infects macrophages, B-cells, and activated T-cells in vivo. This study investigates the cellular basis of resistance to MD in vitro with the hypothesis that resistance is determined by cells active during the innate immune response. Chicken bone marrow-derived macrophages from lines 61 and 72 were infected with MDV in vitro. Flow cytometry showed that a higher percentage of macrophages were infected in line 72 than in line 61. A transcriptomic study followed by in silico functional analysis of differentially expressed genes was then carried out between the two lines pre- and post-infection. Analysis supports the hypothesis that macrophages from susceptible and resistant chicken lines display a marked difference in their transcriptome following MDV infection. Resistance to infection, differential activation of biological pathways, and suppression of oncogenic potential are among host defense strategies identified in macrophages from resistant chickens.
Collapse
Affiliation(s)
- Pankaj Chakraborty
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK; (P.C.); (R.K.); (L.V.); (B.M.D.)
- Chittagong Veterinary and Animal Sciences University, Khulshi, Chittagong 4225, Bangladesh
| | - Richard Kuo
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK; (P.C.); (R.K.); (L.V.); (B.M.D.)
| | - Lonneke Vervelde
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK; (P.C.); (R.K.); (L.V.); (B.M.D.)
| | - Bernadette M. Dutia
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK; (P.C.); (R.K.); (L.V.); (B.M.D.)
| | - Pete Kaiser
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK; (P.C.); (R.K.); (L.V.); (B.M.D.)
| | - Jacqueline Smith
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK; (P.C.); (R.K.); (L.V.); (B.M.D.)
- Correspondence: ; Tel.: +44-(0)131-6519155
| |
Collapse
|
13
|
Genetic assessment of inbred chicken lines indicates genomic signatures of resistance to Marek's disease. J Anim Sci Biotechnol 2018; 9:65. [PMID: 30221000 PMCID: PMC6136188 DOI: 10.1186/s40104-018-0281-x] [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] [Received: 04/07/2018] [Accepted: 07/25/2018] [Indexed: 11/10/2022] Open
Abstract
Background Marek’s disease (MD) is a highly contagious pathogenic and oncogenic disease primarily affecting chickens. However, the mechanisms of genetic resistance for MD are complex and not fully understood. MD-resistant line 63 and MD-susceptible line 72 are two highly inbred progenitor lines of White Leghorn. Recombinant Congenic Strains (RCS) were developed from these two lines, which show varied susceptibility to MD. Results We investigated genetic structure and genomic signatures across the genome, including the line 63 and line 72, six RCSs, and two reciprocally crossed flocks between the lines 63 and 72 (F1 63 × 72 and F1 72 × 63) using Affymetrix® Axiom® HD 600 K genotyping array. We observed 18 chickens from RCS lines were specifically clustered into resistance sub-groups distributed around line 63. Additionally, homozygosity analysis was employed to explore potential genetic components related to MD resistance, while runs of homozygosity (ROH) are regions of the genome where the identical haplotypes are inherited from each parent. We found several genes including SIK, SOX1, LIG4, SIK1 and TNFSF13B were contained in ROH region identified in resistant group (line 63 and RCS), and these genes have been reported that are contribute to immunology and survival. Based on FST based population differential analysis, we also identified important genes related to cell death and anti-apoptosis, including AKT1, API5, CDH13, CFDP and USP15, which could be involved in divergent selection during inbreeding process. Conclusions Our findings offer valuable insights for understanding the genetic mechanism of resistance to MD and the identified genes could be considered as candidate biomarkers in further evaluation. Electronic supplementary material The online version of this article (10.1186/s40104-018-0281-x) contains supplementary material, which is available to authorized users.
Collapse
|
14
|
Deist MS, Gallardo RA, Bunn DA, Kelly TR, Dekkers JCM, Zhou H, Lamont SJ. Novel analysis of the Harderian gland transcriptome response to Newcastle disease virus in two inbred chicken lines. Sci Rep 2018; 8:6558. [PMID: 29700338 PMCID: PMC5920083 DOI: 10.1038/s41598-018-24830-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 04/06/2018] [Indexed: 01/14/2023] Open
Abstract
Behind each eye of the chicken resides a unique lymph tissue, the Harderian gland, for which RNA sequencing (RNA-seq) analysis is novel. We characterized the response of this tissue to Newcastle disease virus (NDV) in two inbred lines with different susceptibility to NDV across three time points. Three-week-old relatively resistant (Fayoumi) and relatively susceptible (Leghorn) birds were inoculated with a high-titered (107EID50) La Sota strain of NDV via an oculonasal route. At 2, 6, and 10 days post infection (dpi) Harderian glands were collected and analyzed via RNA-seq. The Fayoumi had significantly more detectable viral transcripts in the Harderian gland at 2 dpi than the Leghorn, but cleared the virus by 6 dpi. At all three time points, few genes were declared differentially expressed (DE) between the challenged and nonchallenged birds, except for the Leghorns at 6 dpi, and these DE genes were predicted to activate an adaptive immune response. Relative to the Leghorn, the Fayoumi was predicted to activate more immune pathways in both challenged and nonchallenged birds suggesting a more elevated immune system in the Fayoumis under homeostatic conditions. Overall, this study helped characterize the function of this important tissue and its response to NDV.
Collapse
Affiliation(s)
- Melissa S Deist
- Department of Animal Science, Iowa State University, Ames, Iowa, USA
| | - Rodrigo A Gallardo
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, California, USA
| | - David A Bunn
- Department of Animal Science, University of California, Davis, California, USA
| | - Terra R Kelly
- Department of Animal Science, University of California, Davis, California, USA.,One Health Institute, University of California, Davis, California, USA
| | - Jack C M Dekkers
- Department of Animal Science, Iowa State University, Ames, Iowa, USA
| | - Huaijun Zhou
- Department of Animal Science, University of California, Davis, California, USA
| | - Susan J Lamont
- Department of Animal Science, Iowa State University, Ames, Iowa, USA.
| |
Collapse
|
15
|
Dong K, Chang S, Xie Q, Black-Pyrkosz A, Zhang H. Comparative transcriptomics of genetically divergent lines of chickens in response to Marek's disease virus challenge at cytolytic phase. PLoS One 2017; 12:e0178923. [PMID: 28591220 PMCID: PMC5462384 DOI: 10.1371/journal.pone.0178923] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 05/22/2017] [Indexed: 11/30/2022] Open
Abstract
Marek's disease (MD), caused by Marek's disease virus (MDV), remains an economically significant threat to the poultry industry worldwide. Genetic resistance to MD is a promising alternative strategy to augment current control measures (vaccination and management). However, only a few functional genes reportedly conferring MD resistance have been identified. Here, we performed a comparative transcriptomics analysis of two highly inbred yet genetically divergent lines of chickens (line 63 and 72) that are resistant and susceptible to MD, respectively, in response to a very virulent plus strain of MDV (vv+MDV) challenge at cytolytic phase. A total of 203 DEGs in response to MDV challenge were identified in the two lines. Of these, 96 DEGs were in common for both lines, in addition to 36 and 71 DEGs that were specific for line 63 and 72, respectively. Functional enrichment analysis results showed the DEGs were significantly enriched in GO terms and pathways associated with immune response. Especially, the four DEGs, FGA, ALB, FN1, and F13A1 that reportedly facilitate virus invasion or immunosuppression, were found to be significantly up-regulated in the susceptible line 72 but down-regulated in the resistant line 63 birds. These results provide new resources for future studies to further elucidate the genetic mechanism conferring MD resistance.
Collapse
Affiliation(s)
- Kunzhe Dong
- USDA, Agricultural Research Service, Avian Disease and Oncology Laboratory, East Lansing, Michigan, United States of America
- ORISE Fellow, USDA, Agriculture Research Service, Avian Disease and Oncology Laboratory, East Lansing, Michigan, United States of America
| | - Shuang Chang
- College of Veterinary Medicine, Shandong Agricultural University, Tai’an, Shandong, China
| | - Qingmei Xie
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Alexis Black-Pyrkosz
- USDA, Agricultural Research Service, Avian Disease and Oncology Laboratory, East Lansing, Michigan, United States of America
| | - Huanmin Zhang
- USDA, Agricultural Research Service, Avian Disease and Oncology Laboratory, East Lansing, Michigan, United States of America
| |
Collapse
|
16
|
Characterization of Copy Number Variation's Potential Role in Marek's Disease. Int J Mol Sci 2017; 18:ijms18051020. [PMID: 28486430 PMCID: PMC5454933 DOI: 10.3390/ijms18051020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 04/22/2017] [Accepted: 05/04/2017] [Indexed: 02/07/2023] Open
Abstract
Marek’s Disease (MD) is a highly contagious pathogenic and oncogenic disease primarily affecting chickens. Chicken Lines 63 and 72, as well as their recombinant congenic strains (RCS) with varied susceptibility to MD, are ideal models to study the complex mechanisms of genetic resistance to MD. In this study, we investigated copy number variation (CNV) in these inbred chicken lines using the Affymetrix Axiom HD 600 K SNP genotyping array. We detected 393 CNV segments across all ten chicken lines, of which 12 CNVs were specifically identified in Line 72. We then assessed genetic structure based on CNV and observed markedly different patterns. Finally, we validated two deletion events in Line 72 and correlated them with genes expression using qPCR and RNA-seq, respectively. Our combined results indicated that these two CNV deletions were likely to contribute to MD susceptibility.
Collapse
|
17
|
Kaya M, Preeyanon L, Dodgson JB, Cheng HH. Validation of Alternative Transcript Splicing in Chicken Lines that Differ in Genetic Resistance to Marek's Disease. Anim Biotechnol 2017; 27:238-44. [PMID: 27565867 DOI: 10.1080/10495398.2016.1178139] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Utilizing RNA-seq data, 1,574 candidate genes with alternative splicing were previously identified between two chicken lines that differ in Marek's disease (MD) genetic resistance under control and Marek's disease virus infection conditions. After filtering out 1,530 genes with splice variants in the first or last exon, 44 genes were screened for possible exon loss or gain using PCR and gel electrophoresis. Consequently, 7 genes exhibited visually detectable differential expression of splice variants between lines 6 (MD resistant) and 7 (MD susceptible), and the resultant PCR products verified by DNA sequencing. Birds from inbred line 6 have transcripts that preferentially retain an exon compared to line 7 chickens for ITGB2, SGPL1, and COMMD5. Birds from inbred line 7 have alleles that preferentially retain an exon compared to line 6 for MOCS2. CCBL2 exon 1a is absent and ATAD1 exon 2 is truncated by 87 nucleotides in transcripts expressed by line 7 compared to those from line 6. For CHTF18, line 6 transcripts have an indel mutation with 7 additional nucleotides in exon 21 compared to line 7. The current study validates 7 genes with alternatively spliced isomers between the two chicken lines, which helps provide potential underlying mechanisms for the phenotypic differences.
Collapse
Affiliation(s)
- Muhammet Kaya
- a Eskişehir Osmangazi University, Faculty of Agriculture, Department of Agricultural Biotechnology , Eskisehir , Turkey.,b Michigan State University , East Lansing , Michigan , USA
| | | | | | - Hans H Cheng
- c USDA, ARS, Avian Disease & Oncology Laboratory , East Lansing , Michigan , USA
| |
Collapse
|
18
|
Cheng HH, Perumbakkam S, Pyrkosz AB, Dunn JR, Legarra A, Muir WM. Fine mapping of QTL and genomic prediction using allele-specific expression SNPs demonstrates that the complex trait of genetic resistance to Marek's disease is predominantly determined by transcriptional regulation. BMC Genomics 2015; 16:816. [PMID: 26481588 PMCID: PMC4617451 DOI: 10.1186/s12864-015-2016-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/04/2015] [Indexed: 11/30/2022] Open
Abstract
Background Marek’s disease (MD) is a lymphoproliferative disease of poultry induced by Marek’s disease virus (MDV), a highly oncogenic alphaherpesvirus. Identifying the underlying genes conferring MD genetic resistance is desired for more efficacious control measures including genomic selection, which requires accurately identified genetic markers throughout the chicken genome. Methods Hypothesizing that variants located in transcriptional regulatory regions are the main mechanism underlying this complex trait, a genome-wide association study was conducted by genotyping a ~1,000 bird MD resource population derived from experimental inbred layers with SNPs containing 1,824 previously identified allele-specific expression (ASE) SNPs in response to MDV infection as well as 3,097 random SNPs equally spaced throughout the chicken genome. Based on the calculated associations, genomic predictions were determined for 200 roosters and selected sires had their progeny tested for Marek’s disease incidence. Results Our analyses indicate that these ASE SNPs account for more than 83 % of the genetic variance and exhibit nearly all the highest associations. To validate these findings, 200 roosters had their genetic merit predicted from the ASE SNPs only, and the top 30 and bottom 30 ranked roosters were reciprocally mated to random hens. The resulting progeny showed that after only one generation of bidirectional selection, there was a 22 % difference in MD incidence and this approach gave a 125 % increase in accuracy compared to current pedigree-based estimates. Conclusions We conclude that variation in transcriptional regulation is the major driving cause for genetic resistance to MD, and ASE SNPs identify the underlying genes and are sufficiently linked to the causative polymorphisms that they can be used for accurate genomic prediction as well as help define the underlying molecular basis. Furthermore, this approach should be applicable to other complex traits. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2016-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hans H Cheng
- USDA, ARS, Avian Disease and Oncology Laboratory, East Lansing, MI, 48823, USA.
| | - Sudeep Perumbakkam
- USDA, ARS, Avian Disease and Oncology Laboratory, East Lansing, MI, 48823, USA.,Microbiology & Molecular Genetics, Michigan State University, East Lansing, MI, 48824, USA
| | | | - John R Dunn
- USDA, ARS, Avian Disease and Oncology Laboratory, East Lansing, MI, 48823, USA
| | - Andres Legarra
- INRA, Animal Genetics, GenPhySE, Castanet Tolosan, 31326, France
| | - William M Muir
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| |
Collapse
|
19
|
Collier RJ, Bauman DE. Update on human health concerns of recombinant bovine somatotropin use in dairy cows. J Anim Sci 2014; 92:1800-7. [DOI: 10.2527/jas.2013-7383] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- R. J. Collier
- School of Animal and Comparative Biomedical Sciences University of Arizona, Tucson 85719
| | - D. E. Bauman
- Department of Animal Science, Cornell University, Ithaca, NY 14853
| |
Collapse
|
20
|
Transcriptional profiling of mEq-dependent genes in Marek's disease resistant and susceptible inbred chicken lines. PLoS One 2013; 8:e78171. [PMID: 24205146 PMCID: PMC3804455 DOI: 10.1371/journal.pone.0078171] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/17/2013] [Indexed: 12/18/2022] Open
Abstract
Marek’s disease (MD) is an economically significant disease in chickens caused by the highly oncogenic Marek’s disease virus (MDV). Understanding the genes and biological pathways that confer MD genetic resistance should lead towards the development of more disease resistant commercial poultry flocks or improved MD vaccines. MDV mEq, a bZIP transcription factor, is largely attributed to viral oncogenicity though only a few host target genes have been described, which has impeded our understanding of MDV-induced tumorigenesis. Given the importance of mEq in MDV-induced pathogenesis, we explored the role of mEq in genetic resistance to MDV. Using global transcriptome analysis and cells from MD resistant or susceptible birds, we compared the response to infection with either wild type MDV or a nononcogenic recombinant lacking mEq. As a result, we identified a number of specific genes and pathways associated with either MD resistance or susceptibility. Additionally, integrating prior information from ChIP-seq, microarray analysis, and SNPs exhibiting allele-specific expression (ASE) in response to MDV infection, we were able to provide evidence for 24 genes that are polymorphic within mEq binding sites are likely to account for gene expression in an allele-specific manner and potentially for the underlying genetic differences in MD incidence.
Collapse
|
21
|
Sodhi SS, Jeong DK, Sharma N, Lee JH, Kim JH, Kim SH, Kim SW, Oh SJ. Marker Assisted Selection-Applications and Evaluation for Commercial Poultry Breeding. ACTA ACUST UNITED AC 2013. [DOI: 10.5536/kjps.2013.40.3.223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
22
|
Fulton JE, Arango J, Arthur JA, Settar P, Kreager KS, O'Sullivan NP. Improving the outcome of a Marek's disease challenge in multiple lines of egg type chickens. Avian Dis 2013; 57:519-22. [PMID: 23901770 DOI: 10.1637/10408-100212-reg.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
A challenge test following inoculation with a standard amount of a vv+ strain of the Marek's disease (MD) virus in multiple lines and multiple generations of egg type chicken and the corresponding phenotypic trend are described. This program significantly reduced mortality of progeny from selected sires for three to 11 generations in eight of the nine elite lines studied herein. In brown egg lines, a retrospective analysis of DNA indicated an association between the blood type B (major histocompatibility complex) of the sire and the MD mortality in the challenge of its progeny. As a result of the multigeneration stock amplification and crossbreeding processes used in the commercial breeding industry, improvement in survival after challenge at the elite level will translate to improved welfare for millions of birds at the commercial production level.
Collapse
Affiliation(s)
- J E Fulton
- Hy-Line International, P.O. Box 310, Dallas Center, 1A 50063, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Integrated analyses of genome-wide DNA occupancy and expression profiling identify key genes and pathways involved in cellular transformation by a Marek's disease virus oncoprotein, Meq. J Virol 2013; 87:9016-29. [PMID: 23740999 DOI: 10.1128/jvi.01163-13] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Marek's disease (MD) is an economically significant disease in chickens that is caused by the highly oncogenic Marek's disease virus (MDV). A major unanswered question is the mechanism of MDV-induced tumor formation. Meq, a bZIP transcription factor discovered in the 1990s, is critically involved in viral oncogenicity, but only a few of its host target genes have been described, impeding our understanding of MDV-induced tumorigenesis. Using chromatin immunoprecipitation-sequencing (ChIP-seq) and microarray analysis, a high-confidence list of Meq binding sites in the chicken genome and a global transcriptome of Meq-responsive genes were generated. Meq binding sites were found to be enriched in the promoter regions of upregulated genes but not in those of downregulated genes. ChIP-seq was also performed for c-Jun, a known heterodimeric partner of Meq. The close location of binding sites of Meq and c-Jun was noted, suggesting cooperativity between these two factors in modulating transcription. Pathway analysis indicated that Meq transcriptionally regulates many genes that are part of several signaling pathways including the extracellular signal-regulated kinase /mitogen-activated protein kinase (ERK/MAPK), Jak-STAT, and ErbB pathways, which are critical for oncogenesis and/or include signaling mediators involved in apoptosis. Meq activates oncogenic signaling cascades by transcriptionally activating major kinases in the ERK/MAPK pathway and simultaneously repressing phosphatases, as verified using inhibitors of MEK and ERK1/2 in a cell proliferation assay. This study provides significant insights into the mechanistic basis of Meq-dependent cell transformation.
Collapse
|
24
|
|
25
|
Tian F, Zhan F, VanderKraats ND, Hiken JF, Edwards JR, Zhang H, Zhao K, Song J. DNMT gene expression and methylome in Marek's disease resistant and susceptible chickens prior to and following infection by MDV. Epigenetics 2013; 8:431-44. [PMID: 23538681 DOI: 10.4161/epi.24361] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Marek's disease (MD) is characterized as a T cell lymphoma induced by a cell-associated α-herpesvirus, Marek's disease virus type 1 (MDV1). As with many viral infectious diseases, DNA methylation variations were observed in the progression of MD; these variations are thought to play an important role in host-virus interactions. We observed that DNA methyltransferase 3a (DNMT3a) and 3b (DNMT3b) were differentially expressed in chicken MD-resistant line 6 3 and MD-susceptible line 7 2 at 21 d after MDV infection. To better understand the role of methylation variation induced by MDV infection in both chicken lines, we mapped the genome-wide DNA methylation profiles in each line using Methyl-MAPS (methylation mapping analysis by paired-end sequencing). Collectively, the data sets collected in this study provide a more comprehensive picture of the chicken methylome. Overall, methylation levels were reduced in chickens from the resistant line 6 3 after MDV infection. We identified 11,512 infection-induced differential methylation regions (iDMRs). The number of iDMRs was larger in line 7 2 than in line 6 3, and most of iDMRs found in line 6 3 were overlapped with the iDMRs found in line 7 2. We further showed that in vitro methylation levels were associated with MDV replication, and found that MDV propagation in the infected cells was restricted by pharmacological inhibition of DNA methylation. Our results suggest that DNA methylation in the host may be associated with disease resistance or susceptibility. The methylation variations induced by viral infection may consequentially change the host transcriptome and result in diverse disease outcomes.
Collapse
Affiliation(s)
- Fei Tian
- Department of Animal & Avian Sciences; University of Maryland; College Park, MD USA
| | - Fei Zhan
- Department of Animal & Avian Sciences; University of Maryland; College Park, MD USA
| | - Nathan D VanderKraats
- Center for Pharmacogenomics; Department of Medicine; Washington University School of Medicine; St. Louis, MO USA
| | - Jeffrey F Hiken
- Center for Pharmacogenomics; Department of Medicine; Washington University School of Medicine; St. Louis, MO USA
| | - John R Edwards
- Center for Pharmacogenomics; Department of Medicine; Washington University School of Medicine; St. Louis, MO USA
| | - Huanmin Zhang
- USDA; ARS, Avian Disease and Oncology Laboratory; East Lansing, MI USA; Department of Animal Science; Michigan State University; East Lansing, MI USA
| | - Keji Zhao
- Laboratory of Molecular Immunology; National Heart, Lung and Blood Institute; National Institutes of Health; Bethesda, MD USA
| | - Jiuzhou Song
- Department of Animal & Avian Sciences; University of Maryland; College Park, MD USA
| |
Collapse
|
26
|
Perumbakkam S, Muir WM, Black-Pyrkosz A, Okimoto R, Cheng HH. Comparison and contrast of genes and biological pathways responding to Marek's disease virus infection using allele-specific expression and differential expression in broiler and layer chickens. BMC Genomics 2013; 14:64. [PMID: 23363372 PMCID: PMC3599046 DOI: 10.1186/1471-2164-14-64] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 01/22/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Marek's disease (MD) is a commercially important neoplastic disease of chickens caused by the Marek's disease virus (MDV), a naturally occurring oncogenic alphaherpesvirus. Enhancing MD genetic resistance is desirable to augment current vaccines and other MD control measures. High throughput sequencing was used to profile splenic transcriptomes from individual F1 progeny infected with MDV at 4 days of age from both outbred broilers (meat-type) and inbred layer (egg-type) chicken lines that differed in MD genetic resistance. The resulting information was used to identify SNPs, genes, and biological pathways exhibiting allele-specific expression (ASE) in response to MDV infection in each type of chicken. In addition, we compared and contrasted the results of pathway analyses (ASE and differential expression (DE)) between chicken types to help inform on the biological response to MDV infection. RESULTS With 7 individuals per line and treatment group providing high power, we identified 6,132 single nucleotide polymorphisms (SNPs) in 4,768 genes and 4,528 SNPs in 3,718 genes in broilers and layers, respectively, that exhibited ASE in response to MDV infection. Furthermore, 548 and 434 genes in broilers and layers, respectively, were found to show DE following MDV infection. Comparing the datasets, only 72 SNPs and 850 genes for ASE and 20 genes for DE were common between the two bird types. Although the chicken types used in this study were genetically different, at the pathway level, both TLR receptor and JAK/STAT signaling pathways were enriched as well as exhibiting a high proportion of ASE genes, especially at the beginning of both above mentioned regulatory pathways. CONCLUSIONS RNA sequencing with adequate biological replicates is a powerful approach to identify high confidence SNPs, genes, and pathways that are associated with transcriptional response to MDV infection. In addition, the SNPs exhibiting ASE in response to MDV infection provide a strong foundation for determining the extent to which variation in expression influences MD incidence plus yield genetic markers for genomic selection. However, given the paucity of overlap among ASE SNP sets (broilers vs. layers), it is likely that separate screens need to be incorporated for each population. Finally, comparison of gene lists obtained between these two diverse chicken types indicate the TLR and JAK/STAT signaling are conserved when responding to MDV infection and may be altered by selection of genes exhibiting ASE found at the start of each pathway.
Collapse
Affiliation(s)
- Sudeep Perumbakkam
- Department of Animal Science, Purdue University, West Lafayette, IN, 47907, USA
| | - William M Muir
- Department of Animal Science, Purdue University, West Lafayette, IN, 47907, USA
| | | | - Ron Okimoto
- Cobb-Vantress, Siloam Springs, AR, 72761, USA
| | - Hans H Cheng
- Avian Diseases and Oncology Laboratory, USDA, ARS, East Lansing, MI, 48823, USA
| |
Collapse
|
27
|
Cheng HH, Kaiser P, Lamont SJ. Integrated Genomic Approaches to Enhance Genetic Resistance in Chickens. Annu Rev Anim Biosci 2013; 1:239-60. [DOI: 10.1146/annurev-animal-031412-103701] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Hans H. Cheng
- Avian Disease and Oncology Laboratory, USDA, ARS, East Lansing, Michigan 48823;
| | - Pete Kaiser
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, United Kingdom;
| | - Susan J. Lamont
- Department of Animal Science, Iowa State University, Ames, Iowa 50011;
| |
Collapse
|
28
|
Marek's disease virus infection induces widespread differential chromatin marks in inbred chicken lines. BMC Genomics 2012; 13:557. [PMID: 23072359 PMCID: PMC3505159 DOI: 10.1186/1471-2164-13-557] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 10/08/2012] [Indexed: 11/10/2022] Open
Abstract
Background Marek’s disease (MD) is a neoplastic disease in chickens caused by the MD virus (MDV). Successful vaccine development against MD has resulted in increased virulence of MDV and the understanding of genetic resistance to the disease is, therefore, crucial to long-term control strategies. Also, epigenetic factors are believed to be one of the major determinants of disease response. Results Here, we carried out comprehensive analyses of the epigenetic landscape induced by MDV, utilizing genome-wide histone H3 lysine 4 and lysine 27 trimethylation maps from chicken lines with varying resistance to MD. Differential chromatin marks were observed on genes previously implicated in the disease such as MX1 and CTLA-4 and also on genes reported in other cancers including IGF2BP1 and GAL. We detected bivalent domains on immune-related transcriptional regulators BCL6, CITED2 and EGR1, which underwent dynamic changes in both lines as a result of MDV infection. In addition, putative roles for GAL in the mechanism of MD progression were revealed. Conclusion Our results confirm the presence of widespread epigenetic differences induced by MD in chicken lines with different levels of genetic resistance. A majority of observed epigenetic changes were indicative of increased levels of viral infection in the susceptible line symptomatic of lowered immunocompetence in these birds caused by early cytolytic infection. The GAL system that has known anti-proliferative effects in other cancers is also revealed to be potentially involved in MD progression. Our study provides further insight into the mechanisms of MD progression while revealing a complex landscape of epigenetic regulatory mechanisms that varies depending on host factors.
Collapse
|
29
|
Kumar S, Kunec D, Buza JJ, Chiang HI, Zhou H, Subramaniam S, Pendarvis K, Cheng HH, Burgess SC. Nuclear Factor kappa B is central to Marek's disease herpesvirus induced neoplastic transformation of CD30 expressing lymphocytes in-vivo. BMC SYSTEMS BIOLOGY 2012; 6:123. [PMID: 22979947 PMCID: PMC3472249 DOI: 10.1186/1752-0509-6-123] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 09/04/2012] [Indexed: 12/15/2022]
Abstract
Background Marek’s Disease (MD) is a hyperproliferative, lymphomatous, neoplastic disease of chickens caused by the oncogenic Gallid herpesvirus type 2 (GaHV-2; MDV). Like several human lymphomas the neoplastic MD lymphoma cells overexpress the CD30 antigen (CD30hi) and are in minority, while the non-neoplastic cells (CD30lo) form the majority of population. MD is a unique natural in-vivo model of human CD30hi lymphomas with both natural CD30hi lymphomagenesis and spontaneous regression. The exact mechanism of neoplastic transformation from CD30lo expressing phenotype to CD30hi expressing neoplastic phenotype is unknown. Here, using microarray, proteomics and Systems Biology modeling; we compare the global gene expression of CD30lo and CD30hi cells to identify key pathways of neoplastic transformation. We propose and test a specific mechanism of neoplastic transformation, and genetic resistance, involving the MDV oncogene Meq, host gene products of the Nuclear Factor Kappa B (NF-κB) family and CD30; we also identify a novel Meq protein interactome. Results Our results show that a) CD30lo lymphocytes are pre-neoplastic precursors and not merely reactive lymphocytes; b) multiple transformation mechanisms exist and are potentially controlled by Meq; c) Meq can drive a feed-forward cycle that induces CD30 transcription, increases CD30 signaling which activates NF-κB, and, in turn, increases Meq transcription; d) Meq transcriptional repression or activation of the CD30 promoter generally correlates with polymorphisms in the CD30 promoter distinguishing MD-lymphoma resistant and susceptible chicken genotypes e) MDV oncoprotein Meq interacts with proteins involved in physiological processes central to lymphomagenesis. Conclusions In the context of the MD lymphoma microenvironment (and potentially in other CD30hi lymphomas as well), our results show that the neoplastic transformation is a continuum and the non-neoplastic cells are actually pre-neoplastic precursor cells and not merely immune bystanders. We also show that NF-κB is a central player in MDV induced neoplastic transformation of CD30-expressing lymphocytes in vivo. Our results provide insights into molecular mechanisms of neoplastic transformation in MD specifically and also herpesvirus induced lymphoma in general.
Collapse
Affiliation(s)
- Shyamesh Kumar
- Department of Pathobiology and Population Medicine, Mississippi State University, MS 39762, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Luo J, Mitra A, Tian F, Chang S, Zhang H, Cui K, Yu Y, Zhao K, Song J. Histone methylation analysis and pathway predictions in chickens after MDV infection. PLoS One 2012; 7:e41849. [PMID: 22848633 PMCID: PMC3406056 DOI: 10.1371/journal.pone.0041849] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 06/29/2012] [Indexed: 01/22/2023] Open
Abstract
Marek's disease (MD) is a lymphoproliferative disease in chicken induced by Marek's disease virus (MDV). Although studies have focused on the genetic differences between the resistant and susceptible chicken, less is known about the role of epigenetic factors in MD. In this study, genome-wide histone modifications in the non-MHC-associated resistant and susceptible chicken lines were examined. We found that tri-methylation at histone H3 Lys4 (H3K4me3) enrichment is positively correlated with the expression of protein coding genes as well as microRNA (miRNA) genes, whereas tri-methylation at histone H3 Lys27 (H3K27me3) exhibits a negative correlation. By identifying line-specific histone modifications in MDV infection, we found unique H3K4me3 islands in the resistant chicken activated genes, which are related to immune response and cell adhesion. Interestingly, we also found some miRNAs from unique H3K27me3 patterns in the susceptible chickens that targeted genes involved in 5-hydroxytryptamine (5-HT)-receptor and adrenergic receptor pathways. In conclusion, dynamic line-specific histone modifications in response to MDV infection suggested that intrinsic epigenetic mechanisms may play a role in MD-resistance and -susceptibility.
Collapse
Affiliation(s)
- Juan Luo
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, United States of America
| | - Apratim Mitra
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, United States of America
| | - Fei Tian
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, United States of America
| | - Shuang Chang
- United States Department of Agriculture, ARS, Avian Disease and Oncology Laboratory, East Lansing, Michigan, United States of America
- Department of Animal Science, Michigan State University, East Lansing, Michigan, United States of America
| | - Huanmin Zhang
- United States Department of Agriculture, ARS, Avian Disease and Oncology Laboratory, East Lansing, Michigan, United States of America
| | - Kairong Cui
- Laboratory of Molecular Immunology, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ying Yu
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, United States of America
| | - Keji Zhao
- Laboratory of Molecular Immunology, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jiuzhou Song
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, United States of America
| |
Collapse
|
31
|
Li DF, Lian L, Qu LJ, Chen YM, Liu WB, Chen SR, Zheng JX, Xu GY, Yang N. A genome-wide SNP scan reveals two loci associated with the chicken resistance to Marek's disease. Anim Genet 2012; 44:217-22. [PMID: 22812605 DOI: 10.1111/j.1365-2052.2012.02395.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2012] [Indexed: 12/18/2022]
Abstract
Marek's disease (MD) is a neoplastic disease in chickens, caused by the Marek's disease virus (MDV). To investigate host genetic resistance to MD, we conducted a genome-wide association study (GWAS) on 67 MDV-infected chickens based on a case and control design, including 57 susceptible chickens in the case group and 10 resistant chickens as controls. After searching 38 655 valid genomic markers, two SNPs were found to be associated with host resistance to MD. One SNP, rs14527240, reaching chromosome-wide significance level (P < 0.01) was located in the SPARC-related modular calcium-binding 1 (SMOC1) gene on GGA5. The other one, GGaluGA156129, reaching genome-wide significance (P < 0.05), was located in the protein tyrosine phosphatase, non-receptor type 3 (PTPN3) gene on GGA2. In addition, expression patterns of these two genes in spleens were detected by qPCR. The expression of SMOC1 was significantly up-regulated (P < 0.05), whereas the expression of PTNP3 did not show significance when the case group was compared with the control group. Up-regulation of SMOC1 in susceptible spleens suggests its important roles in MD tumorigenesis. This is the first study to investigate MD-resistant loci, and it demonstrates the power of GWASs for mapping genes associated with MD resistance.
Collapse
Affiliation(s)
- D F Li
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Gimeno IM, Witter RL, Cortes AL, Reed WM. Replication ability of three highly protective Marek's disease vaccines: implications in lymphoid organ atrophy and protection. Avian Pathol 2012; 40:573-9. [PMID: 22107091 DOI: 10.1080/03079457.2011.617725] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
The present work is a chronological study of the pathogenesis of three attenuated serotype 1 Marek's disease (MD) virus strains (RM1, CVI988 and 648A80) that provide high protection against MD but have been attenuated by different procedures and induce different degrees of lymphoid organ atrophy. All studied strains replicated in the lymphoid organs (bursa,x thymus and spleen) and a peak of replication was detected at 6 days post inoculation (d.p.i.). Differences, however, were observed among vaccine strains. RM1 strain replicates more in all lymphoid organs compared with CVI988 and 648A80 strains. In addition, replication of RM1 in the thymus did not decrease after 6 d.p.i. but continued at high levels at 14 d.p.i. and until the thymus was completely destroyed. Lung infection occurred very early after infection with all of the three vaccines and the level of replication was similar to that found in the lymphoid organs. Infected cells were very large and appeared scattered in the lung parenchyma and in the parabronchial lining. The study of the target cells for the early infection in cell suspensions of blood and spleen showed that both non-adherent cell populations (enriched in lymphoid cells) and adherent cells (enriched in monocytes/macrophages) supported MD virus infection. Infection in adherent cells was especially high at very early stages of the infection (3 to 6 d.p.i.). Atrophy of lymphoid organs is a major drawback in the production of highly protective vaccines against MD. A better understanding of the mechanisms associated with lymphoid organ atrophy will aid in overcoming this problem.
Collapse
Affiliation(s)
- Isabel M Gimeno
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, USA.
| | | | | | | |
Collapse
|
33
|
Maceachern S, Muir WM, Crosby SD, Cheng HH. Genome-Wide Identification and Quantification of cis- and trans-Regulated Genes Responding to Marek's Disease Virus Infection via Analysis of Allele-Specific Expression. Front Genet 2012; 2:113. [PMID: 22303407 PMCID: PMC3268648 DOI: 10.3389/fgene.2011.00113] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 12/29/2011] [Indexed: 11/17/2022] Open
Abstract
Marek’s disease (MD) is a commercially important neoplastic disease of chickens caused by Marek’s disease virus (MDV), a naturally occurring oncogenic alphaherpesvirus. Selecting for increased genetic resistance to MD is a control strategy that can augment vaccinal control measures. To identify high-confidence candidate MD resistance genes, we conducted a genome-wide screen for allele-specific expression (ASE) amongst F1 progeny of two inbred chicken lines that differ substantially in MD resistance. High throughput sequencing was initially used to profile transcriptomes from pools of uninfected and infected individuals at 4 days post-infection to identify any genes showing ASE in response to MDV infection. RNA sequencing identified 22,655 single nucleotide polymorphisms (SNPs) of which 5,360 in 3,773 genes exhibited significant allelic imbalance. Illumina GoldenGate assays were subsequently used to quantify regulatory variation controlled at the gene (cis) and elsewhere in the genome (trans) by examining differences in expression between F1 individuals and artificial F1 RNA pools over six time periods in 1,536 of the most significant SNPs identified by RNA sequencing. Allelic imbalance as a result of cis-regulatory changes was confirmed in 861 of the 1,233 GoldenGate assays successfully examined. Furthermore we have identified seven genes that display trans-regulation only in infected animals and ∼500 SNP that show a complex interaction between cis- and trans-regulatory changes. Our results indicate ASE analyses are a powerful approach to identify regulatory variation responsible for differences in transcript abundance in genes underlying complex traits. And the genes with SNPs exhibiting ASE provide a strong foundation to further investigate the causative polymorphisms and genetic mechanisms for MD resistance. Finally, the methods used here for identifying specific genes and SNPs have practical implications for applying marker-assisted selection to complex traits that are difficult to measure in agricultural species, when expression differences are expected to control a portion of the phenotypic variance.
Collapse
Affiliation(s)
- Sean Maceachern
- Avian Disease and Oncology Laboratory, Agricultural Research Service, United States Department of Agriculture East Lansing, MI, USA
| | | | | | | |
Collapse
|
34
|
Cheng HH, MacEachern S, Subramaniam S, Muir WM. Chicks and single-nucleotide polymorphisms: an entrée into identifying genes conferring disease resistance in chicken. ANIMAL PRODUCTION SCIENCE 2012. [DOI: 10.1071/an11099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Marek’s disease (MD) is one of the most serious chronic infectious disease threats to the poultry industry worldwide. Selecting for increased genetic resistance to MD is a control strategy that can augment current vaccinal control measures. Although our previous efforts integrating various genomic screens successfully identified three resistance genes, the main limitation was mapping precision, which hindered our ability to identify and further evaluate high-confidence candidate genes. Towards identifying the remaining genes of this complex trait, we incorporated three additional approaches made substantially more powerful through next-generation sequencing and that exploit the growing importance of expression variation. First, we screened for allele-specific expression (ASE) in response to Marek’s disease virus (MDV) infection, which, when allelic imbalance was identified, is sufficient to indicate a cis-acting element for a specific gene. Second, sequencing of genomic regions enriched by chromatin immunoprecipitation (ChIP) combined with transcript profiling identified motifs bound and genes directly regulated by MDV Meq, a bZIP transcription factor and the viral oncogene. Finally, analysis of genomic sequences from two experimental lines divergently selected for MD genetic resistance allowed inference about regions under selection as well as potential causative polymorphisms. These new combined approaches have resulted in a large number of high-confidence genes conferring MD resistance reflecting the multigenic basis of this trait, which expands our biological knowledge and provides corresponding single-nucleotide polymorhpisms (SNPs) that can be directly evaluated for their genetic contribution towards disease resistance.
Collapse
|
35
|
Bhattacharya T, Priyanka M, Chatterjee R, Sharma R, Bhanja S, Kumar UR, Niranjan M. Polymorphism at exon 1 of pit-1 gene and its association with immunocompetence traits in layer chicken. JOURNAL OF APPLIED ANIMAL RESEARCH 2011. [DOI: 10.1080/09712119.2011.607889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Affiliation(s)
- T.K. Bhattacharya
- a Molecular Genetics and Breeding, Project Directorate on Poultry , Hyderabad, India
| | - M. Priyanka
- a Molecular Genetics and Breeding, Project Directorate on Poultry , Hyderabad, India
| | - R.N. Chatterjee
- a Molecular Genetics and Breeding, Project Directorate on Poultry , Hyderabad, India
| | - R.P. Sharma
- a Molecular Genetics and Breeding, Project Directorate on Poultry , Hyderabad, India
| | - S.K. Bhanja
- a Molecular Genetics and Breeding, Project Directorate on Poultry , Hyderabad, India
| | - U. Raj Kumar
- a Molecular Genetics and Breeding, Project Directorate on Poultry , Hyderabad, India
| | - M. Niranjan
- a Molecular Genetics and Breeding, Project Directorate on Poultry , Hyderabad, India
| |
Collapse
|
36
|
Smith J, Sadeyen JR, Paton IR, Hocking PM, Salmon N, Fife M, Nair V, Burt DW, Kaiser P. Systems analysis of immune responses in Marek's disease virus-infected chickens identifies a gene involved in susceptibility and highlights a possible novel pathogenicity mechanism. J Virol 2011; 85:11146-58. [PMID: 21865384 PMCID: PMC3194948 DOI: 10.1128/jvi.05499-11] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 08/15/2011] [Indexed: 02/04/2023] Open
Abstract
Marek's disease virus (MDV) is a highly contagious oncogenic alphaherpesvirus that causes disease that is both a cancer model and a continuing threat to the world's poultry industry. This comprehensive gene expression study analyzes the host response to infection in both resistant and susceptible lines of chickens and inherent expression differences between the two lines following the infection of the host. A novel pathogenicity mechanism, involving the downregulation of genes containing HIC1 transcription factor binding sites as early as 4 days postinfection, was suggested from this analysis. HIC1 drives antitumor mechanisms, suggesting that MDV infection switches off genes involved in antitumor regulation several days before the expression of the MDV oncogene meq. The comparison of the gene expression data to previous QTL data identified several genes as candidates for involvement in resistance to MD. One of these genes, IRG1, was confirmed by single nucleotide polymorphism analysis to be involved in susceptibility. Its precise mechanism remains to be elucidated, although the analysis of gene expression data suggests it has a role in apoptosis. Understanding which genes are involved in susceptibility/resistance to MD and defining the pathological mechanisms of the disease gives us a much greater ability to try to reduce the incidence of this virus, which is costly to the poultry industry in terms of both animal welfare and economics.
Collapse
Affiliation(s)
- Jacqueline Smith
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Meydan H, Yildiz M, Dodgson J, Cheng H. Allele-specific expression analysis reveals CD79B has a cis-acting regulatory element that responds to Marek's disease virus infection in chickens. Poult Sci 2011; 90:1206-11. [DOI: 10.3382/ps.2010-01295] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
38
|
Abstract
It is more than a century since Marek's disease (MD) was first reported in chickens and since then there have been concerted efforts to better understand this disease, its causative agent and various approaches for control of this disease. Recently, there have been several outbreaks of the disease in various regions, due to the evolving nature of MD virus (MDV), which necessitates the implementation of improved prophylactic approaches. It is therefore essential to better understand the interactions between chickens and the virus. The chicken immune system is directly involved in controlling the entry and the spread of the virus. It employs two distinct but interrelated mechanisms to tackle viral invasion. Innate defense mechanisms comprise secretion of soluble factors as well as cells such as macrophages and natural killer cells as the first line of defense. These innate responses provide the adaptive arm of the immune system including antibody- and cell-mediated immune responses to be tailored more specifically against MDV. In addition to the immune system, genetic and epigenetic mechanisms contribute to the outcome of MDV infection in chickens. This review discusses our current understanding of immune responses elicited against MDV and genetic factors that contribute to the nature of the response.
Collapse
|
39
|
Kim T, Mays J, Fadly A, Silva RF. Artificially inserting a reticuloendotheliosis virus long terminal repeat into a bacterial artificial chromosome clone of Marek’s disease virus (MDV) alters expression of nearby MDV genes. Virus Genes 2011; 42:369-76. [DOI: 10.1007/s11262-011-0585-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 02/03/2011] [Indexed: 10/18/2022]
|
40
|
Haq K, Brisbin JT, Thanthrige-Don N, Heidari M, Sharif S. Transcriptome and proteome profiling of host responses to Marek's disease virus in chickens. Vet Immunol Immunopathol 2010; 138:292-302. [PMID: 21067815 DOI: 10.1016/j.vetimm.2010.10.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Kamran Haq
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | | | | | | | | |
Collapse
|
41
|
Kim ES, Hong YH, Lillehoj HS. Genetic effects analysis of myeloid leukemia factor 2 and T cell receptor-β on resistance to coccidiosis in chickens. Poult Sci 2010; 89:20-7. [PMID: 20008798 DOI: 10.3382/ps.2009-00351] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- E-S Kim
- Bovine Functional Genomics Laboratory, Animal and Natural Resources Institute, USDA, Beltsville, MD 20705, USA
| | | | | |
Collapse
|
42
|
Hong YH, Kim ES, Lillehoj HS, Lillehoj EP, Song KD. Association of resistance to avian coccidiosis with single nucleotide polymorphisms in the zyxin gene. Poult Sci 2009; 88:511-8. [PMID: 19211519 DOI: 10.3382/ps.2008-00344] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Our previous genetic studies demonstrated that resistance to avian coccidiosis is linked with microsatellite markers LEI0071 and LEI0101 on chromosome 1. In this study, the associations between parameters of resistance to coccidiosis and single nucleotide polymorphisms (SNP) in 3 candidate genes located between LEI0071 and LEI0101 [zyxin, CD4, and tumor necrosis factor receptor super family 1A (TNFRSF1A)] were determined. The SNP were genotyped in 24 F(1) generation and 290 F(2) generation animals. No SNP were identified in the TNFRSF1A gene, whereas 10 were located in the zyxin gene and 4 in the CD4 gene. At various times following experimental infection of the F(2) generation with Eimeria maxima, BW, fecal oocyst shedding, and plasma levels of carotenoid, nitrite plus nitrate (NO(2)(-) + NO(3)(-)), and interferon-gamma (IFN-gamma) were measured as parameters of resistance. Single marker and haplotype-based tests were applied to determine the associations between the 14 SNP and the parameters of coccidiosis resistance. None of the CD4 SNP were correlated with disease resistance. However, by single marker association, several of the zyxin SNP were significantly associated with carotenoid or NO(2)(-) + NO(3)(-) concentrations. These were the SNP at nucleotide 149 associated with carotenoid at d 3 postinfection (PI), nucleotide 187 with carotenoid at d 6 and 9 PI, and nucleotide 159 with carotenoid between d 3 and 9 PI. In addition, the zyxin SNP at nucleotide 191 was significantly associated with increased levels of NO(2)(-) + NO(3)(-) at d 3 PI. By haplotype association, the zyxin SNP also were found to be highly associated with NO(2)(-) + NO(3)(-) at d 3 PI and increased IFN-gamma at d 6 PI. These results suggest that zyxin is a candidate gene potentially associated with increased resistance to experimental avian coccidiosis.
Collapse
Affiliation(s)
- Y H Hong
- Animal Parasitic Diseases Laboratory, Animal and Natural Resources Institute, USDA, Beltsville, MD 20705, USA.
| | | | | | | | | |
Collapse
|
43
|
Heifetz EM, Fulton JE, O'Sullivan NP, Arthur JA, Cheng H, Wang J, Soller M, Dekkers JCM. Mapping QTL affecting resistance to Marek's disease in an F6 advanced intercross population of commercial layer chickens. BMC Genomics 2009; 10:20. [PMID: 19144166 PMCID: PMC2651900 DOI: 10.1186/1471-2164-10-20] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Accepted: 01/14/2009] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Marek's disease (MD) is a T-cell lymphoma of chickens caused by the Marek's disease virus (MDV), an oncogenic avian herpesvirus. MD is a major cause of economic loss to the poultry industry and the most serious and persistent infectious disease concern. A full-sib intercross population, consisting of five independent families was generated by crossing and repeated intercrossing of two partially inbred commercial White Leghorn layer lines known to differ in genetic resistance to MD. At the F6 generation, a total of 1615 chicks were produced (98 to 248 per family) and phenotyped for MD resistance measured as survival time in days after challenge with a very virulent plus (vv+) strain of MDV. RESULTS QTL affecting MD resistance were identified by selective DNA pooling using a panel of 15 SNPs and 217 microsatellite markers. Since MHC blood type (BT) is known to affect MD resistance, a total of 18 independent pool pairs were constructed according to family x BT combination, with some combinations represented twice for technical reasons. Twenty-one QTL regions (QTLR) affecting post-challenge survival time were identified, distributed among 11 chromosomes (GGA1, 2, 3, 4, 5, 8, 9, 15, 18, 26 and Z), with about two-thirds of the MD resistance alleles derived from the more MD resistant parental line. Eight of the QTLR associated with MD resistance, were previously identified in a backcross (BC) mapping study with the same parental lines. Of these, 7 originated from the more resistant line, and one from the less resistant line. CONCLUSION There was considerable evidence suggesting that MD resistance alleles tend to be recessive. The width of the QTLR for these QTL appeared to be reduced about two-fold in the F6 as compared to that found in the previous BC study. These results provide a firm basis for high-resolution linkage disequilibrium mapping and positional cloning of the resistance genes.
Collapse
Affiliation(s)
- Eliyahu M Heifetz
- Department of Animal Science and Center for Integrated Animal Genomics, Iowa State University, Ames, IA 50011, USA
- Department of Molecular Biology, Ariel University, Ariel 44837, Israel
| | | | | | | | - Hans Cheng
- USDA-ARS-ADOL, Avian Disease and Oncology Laboratory, East Lansing, MI 48823, USA
| | - Jing Wang
- Department of Animal Science and Center for Integrated Animal Genomics, Iowa State University, Ames, IA 50011, USA
- Pioneer Hi-Bred International Inc., Johnston, IA 50131, USA
| | - Morris Soller
- Department of Genetics, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel
| | - Jack CM Dekkers
- Department of Animal Science and Center for Integrated Animal Genomics, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
44
|
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: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
45
|
Luna M, Rodríguez-Méndez AJ, Berumen L, Carranza M, Riesgo-Escovar J, Baudet ML, Harvey S, Arámburo C. Immune growth hormone (GH): localization of GH and GH mRNA in the bursa of Fabricius. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2008; 32:1313-1325. [PMID: 18539326 DOI: 10.1016/j.dci.2008.04.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Revised: 03/22/2008] [Accepted: 04/16/2008] [Indexed: 05/26/2023]
Abstract
Expression of growth hormone (GH) and GH receptor (GHR) genes in the bursa of Fabricius of chickens suggests that it is an autocrine/paracrine site of GH production and action. The cellular localization of GH and GH mRNA within the bursa was the focus of this study. GH mRNA was expressed mainly in the cortex, comprised of lymphocyte progenitor cells, but was lacking in the medulla where lymphocytes mature. In contrast, more GH immunoreactivity (GH-IR) was present in the medulla than in the cortex. In non-stromal tissues, GH-IR and GH mRNA were primarily in lymphocytes, and also in macrophage-like cells and secretory dendritic cells. In stromal tissues, GH mRNA, GH and GHR were expressed in cells near the connective tissue (CT) between follicles and below the outer serosa. In contrast, GH (but not GH mRNA or GHR), was present in cells of the interfollicular epithelium (IFE), the follicle-associated epithelium (FAE) and the interstitial corticoepithelium. This mismatch may reflect dynamic temporal changes in GH translation. Co-expression of GHR-IR, GH-IR, GH mRNA and IgG was found in immature lymphoid cells near the cortex and in IgG-IR CT cells, suggesting an autocrine/paracrine role for bursal GH in B-cell differentiation.
Collapse
Affiliation(s)
- M Luna
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro 76230, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Sarson AJ, Parvizi P, Lepp D, Quinton M, Sharif S. Transcriptional analysis of host responses to Marek's disease virus infection in genetically resistant and susceptible chickens. Anim Genet 2008; 39:232-40. [PMID: 18371127 DOI: 10.1111/j.1365-2052.2008.01710.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Marek's disease virus (MDV) is a cell-associated oncogenic herpesvirus that targets B cells and T cells, inducing lymphoid tumours in chickens. Genetic resistance to Marek's disease (MD) is regulated in a polygenic fashion. In this study, we sought to compare the gene expression profiles following infection of birds that are genetically resistant or susceptible to MD (with the B21 and B19 haplotypes respectively at the MHC locus), including comparisons to uninfected controls. On days 4, 7, 14 and 21 post-infection, gene expression profiles in spleen tissue were obtained using a chicken immune-specific microarray. A number of genes showed significant (P <or= 0.05) differential expression across time and treatments. These included the chemokine AH221, B-cell marker Bu-1, IgG, IgA, IgM, MHC class II beta chain, granzyme A (GZMA) and signal transducers and activators of transcription 2 (STAT2) genes. In several comparisons, genes such as GZMA and STAT2 were induced in infected birds regardless of their genetic background. However, only immunoglobulin genes were differentially expressed by >or=2-fold in resistant compared with susceptible infected chickens. IgM and IgG were significantly induced on day 7 post-infection in susceptible chickens compared to resistant birds, whereas both of these genes were repressed in susceptible birds on day 14 post-infection. Overall, gene expression profiles in the chicken spleen observed after MDV infection were dependent on time and host genetic background. These gene expression profiles provide a platform for defining novel candidate genes for resistance or susceptibility to MD.
Collapse
Affiliation(s)
- A J Sarson
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | | | | | | | | |
Collapse
|
47
|
Mapping quantitative trait loci affecting susceptibility to Marek's disease virus in a backcross population of layer chickens. Genetics 2008; 177:2417-31. [PMID: 18073438 DOI: 10.1534/genetics.107.080002] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Marek's disease (MD), caused by the oncogenic MD avian herpes virus (MDV), is a major source of economic losses to the poultry industry. A reciprocal backcross (BC) population (total 2052 individuals) was generated by crossing two partially inbred commercial Leghorn layer lines known to differ in MDV resistance, measured as survival time after challenge with a (vv+) MDV. QTL affecting resistance were identified by selective DNA pooling using a panel of 198 microsatellite markers covering two-thirds of the chicken genome. Data for each BC were analyzed separately, and as a combined data set. Markers showing significant association with resistance generally appeared in blocks of two or three, separated by blocks of nonsignificant markers. Defined this way, 15 chromosomal regions (QTLR) affecting MDV resistance, distributed among 10 chromosomes (GGA 1, 2, 3, 4, 5, 7, 8, 9, 15, and Z), were identified. The identified QTLR include one gene and three QTL associated with resistance in previous studies of other lines, and three additional QTL associated with resistance in previous studies of the present lines. These QTL could be used in marker-assisted selection (MAS) programs for MDV resistance and as a platform for high-resolution mapping and positional cloning of the resistance genes.
Collapse
|
48
|
Sequence comparison of the right end of fowl adenovirus genomes. Virus Genes 2008; 36:331-44. [DOI: 10.1007/s11262-007-0194-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2007] [Accepted: 12/27/2007] [Indexed: 10/22/2022]
|
49
|
Cogburn LA, Porter TE, Duclos MJ, Simon J, Burgess SC, Zhu JJ, Cheng HH, Dodgson JB, Burnside J. Functional genomics of the chicken--a model organism. Poult Sci 2007; 86:2059-94. [PMID: 17878436 DOI: 10.1093/ps/86.10.2059] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Since the sequencing of the genome and the development of high-throughput tools for the exploration of functional elements of the genome, the chicken has reached model organism status. Functional genomics focuses on understanding the function and regulation of genes and gene products on a global or genome-wide scale. Systems biology attempts to integrate functional information derived from multiple high-content data sets into a holistic view of all biological processes within a cell or organism. Generation of a large collection ( approximately 600K) of chicken expressed sequence tags, representing most tissues and developmental stages, has enabled the construction of high-density microarrays for transcriptional profiling. Comprehensive analysis of this large expressed sequence tag collection and a set of approximately 20K full-length cDNA sequences indicate that the transcriptome of the chicken represents approximately 20,000 genes. Furthermore, comparative analyses of these sequences have facilitated functional annotation of the genome and the creation of several bioinformatic resources for the chicken. Recently, about 20 papers have been published on transcriptional profiling with DNA microarrays in chicken tissues under various conditions. Proteomics is another powerful high-throughput tool currently used for examining the dynamics of protein expression in chicken tissues and fluids. Computational analyses of the chicken genome are providing new insight into the evolution of gene families in birds and other organisms. Abundant functional genomic resources now support large-scale analyses in the chicken and will facilitate identification of transcriptional mechanisms, gene networks, and metabolic or regulatory pathways that will ultimately determine the phenotype of the bird. New technologies such as marker-assisted selection, transgenics, and RNA interference offer the opportunity to modify the phenotype of the chicken to fit defined production goals. This review focuses on functional genomics in the chicken and provides a road map for large-scale exploration of the chicken genome.
Collapse
Affiliation(s)
- L A Cogburn
- Department of Animal and Food Sciences, University of Delaware, Newark 19717, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
de Koning DJ, Archibald A, Haley CS. Livestock genomics: bridging the gap between mice and men. Trends Biotechnol 2007; 25:483-9. [PMID: 17945371 DOI: 10.1016/j.tibtech.2007.07.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2007] [Revised: 07/25/2007] [Accepted: 07/31/2007] [Indexed: 10/22/2022]
Abstract
Dissecting the genetic control of variation in complex traits, such as disease resistance and agricultural-product quality, remains very challenging. Farm animals are now well placed to bridge the gap between human biology and traditional model species. Livestock species share with model species the benefits of controlled breeding, and their biology is often much closer to that of humans. Genetic research in model species focuses on differences between homogenous lines, whereas genetic research in humans focuses on genetic variation within populations. Livestock genetics has the strengths of both human and model-species genetics because researchers can exploit both the abundant genetic variation between divergent breeds and the variation that is segregating within breeds. Therefore, livestock genomics fills the void where the genetics of model species proves intractable or where model species are not a good proxy for human biology.
Collapse
Affiliation(s)
- Dirk-Jan de Koning
- Division of Genetics and Genomics, Roslin Institute, Roslin, Midlothian, EH25 9PS, UK.
| | | | | |
Collapse
|