1
|
Chen S, Wang J, Wang Q, Ping Y, Yang T, Ma K, Pan S, Bi Y, Bai H, Chang G. Autophagy-mediated TET2 degradation by ALV-J Env protein suppresses innate immune activation to promote viral replication. J Virol 2025:e0200324. [PMID: 39840982 DOI: 10.1128/jvi.02003-24] [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: 11/12/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025] Open
Abstract
Avian leukosis virus subgroup J (ALV-J) poses a significant threat to the poultry industry; yet, our understanding of its replication and pathogenic mechanisms is limited. The Ten-Eleven Translocation 2 (TET2) is an indispensable regulatory factor in active DNA demethylation and immune response regulation. This study reports a significant and time-dependent decrease in TET2 levels following ALV-J infection and shows that the reduction of TET2 protein is mediated by the autophagy pathway. Mechanistically, we found that the accumulation of the Env protein at the late stages of ALV-J infection triggers autophagy, which, in turn, causes the TET2 protein to be exported from the nucleus and subsequently degraded in the cytoplasm. Using CRISPR-Cas9 technology, we generated TET2-deficient chicken macrophages that exhibited increased susceptibility to ALV-J replication, which was reversed by TET2 overexpression. In addition, transcriptome analysis revealed that the absence of TET2 in chicken macrophages impairs the expression of numerous cytokines and innate immune-related genes, particularly those downregulated genes enriched in the RIG-I and Toll-like signaling pathways, leading to enhanced replication of ALV-J in vitro. In summary, our research identifies TET2 as an ALV-J restriction factor. However, ALV-J exploits the autophagy machinery to promote the degradation of TET2 protein, thereby disrupting the host's innate immune responses for viral replication. IMPORTANCE ALV-J is a carcinogenic retrovirus that plays a critical role in avian leukosis, primarily affecting chickens. Infection with ALV-J leads to decreased production performance, compromised immune function, and the development of tumors, such as myelocytoma. Currently, there are no effective treatments for ALV-J, making the control of outbreaks a significant challenge with severe economic consequences for the poultry industry. The Env protein of ALV-J has been implicated in the virus's pathogenicity. Our study shows that ALV-J infection induces autophagy in host cells through its Env protein, leading to the autophagic degradation of TET2, a key epigenetic regulator. The loss of TET2 in macrophages results in the downregulation of innate immune-related gene expression, thereby promoting viral replication. This is the first report to elucidate the role of the ALV-J Env protein in immune suppression via TET2 autophagic degradation during ALV-J infection, providing new insights into the mechanisms of viral immune evasion.
Collapse
Affiliation(s)
- Shihao Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jiaxing Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qiangzhou Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yuyu Ping
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ting Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Kejiao Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Shiyu Pan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yulin Bi
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Hao Bai
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
| | - Guobin Chang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
2
|
Ji J, Xu S, Xu X, Man Y, Yao L, Xie Q, Bi Y. Transcriptome-wide N6-methyladenosine modification and microRNA jointly regulate the infection of avian leukosis virus subgroup J in vitro. Poult Sci 2024; 103:103671. [PMID: 38569240 PMCID: PMC10999702 DOI: 10.1016/j.psj.2024.103671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/07/2024] [Accepted: 03/14/2024] [Indexed: 04/05/2024] Open
Abstract
N6-methyladenosine (m6A) methylation in transcripts has been suggested to influence tumorigenesis in liver tumors caused by the avian leukosis virus subgroup J (ALV-J). However, m6A modifications during ALV-J infection in vitro remain unclear. Herein, we performed m6A and RNA sequencing in ALV-J-infected chicken fibroblasts (DF-1). A total of 51 differentially expressed genes containing differentially methylated peaks were identified, which were markedly enriched in microRNAs (miRNAs) in cancer cells as well as apoptosis, mitophagy and autophagy, RNA degradation, and Hippo and MAPK signaling pathways. Correlation analysis indicated that YTHDC1 (m6A-reader gene) plays a key role in m6A modulation during ALV-J infection. The env gene of ALV-J harbored the strongest peak, and untranslated regions and long terminal repeats also contained peaks of different degrees. To the best of our knowledge, this is the first thorough analysis of m6A patterns in ALV-J-infected DF-1 cells. Combined with miRNA profiles, this study provides a useful basis for future research into the key pathways of ALV-J infection associated with m6A alteration.
Collapse
Affiliation(s)
- Jun Ji
- Henan Provincial Engineering Laboratory of Insects Bio-reactor, Henan Provincial Engineering and Technology Center of Health Products for Livestock and Poultry, Henan Provincial Engineering and Technology Center of Animal Disease Diagnosis and Integrated Control, Nanyang Normal University, Nanyang 473061, PR China.
| | - Shuqi Xu
- Henan Provincial Engineering Laboratory of Insects Bio-reactor, Henan Provincial Engineering and Technology Center of Health Products for Livestock and Poultry, Henan Provincial Engineering and Technology Center of Animal Disease Diagnosis and Integrated Control, Nanyang Normal University, Nanyang 473061, PR China
| | - Xin Xu
- Henan Provincial Engineering Laboratory of Insects Bio-reactor, Henan Provincial Engineering and Technology Center of Health Products for Livestock and Poultry, Henan Provincial Engineering and Technology Center of Animal Disease Diagnosis and Integrated Control, Nanyang Normal University, Nanyang 473061, PR China
| | - Yuanzhuo Man
- Henan Provincial Engineering Laboratory of Insects Bio-reactor, Henan Provincial Engineering and Technology Center of Health Products for Livestock and Poultry, Henan Provincial Engineering and Technology Center of Animal Disease Diagnosis and Integrated Control, Nanyang Normal University, Nanyang 473061, PR China
| | - Lunguang Yao
- Henan Provincial Engineering Laboratory of Insects Bio-reactor, Henan Provincial Engineering and Technology Center of Health Products for Livestock and Poultry, Henan Provincial Engineering and Technology Center of Animal Disease Diagnosis and Integrated Control, Nanyang Normal University, Nanyang 473061, PR China
| | - Qingmei Xie
- College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Yingzuo Bi
- College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| |
Collapse
|
3
|
Fandiño S, Gomez-Lucia E, Benítez L, Doménech A. Avian Leukosis: Will We Be Able to Get Rid of It? Animals (Basel) 2023; 13:2358. [PMID: 37508135 PMCID: PMC10376345 DOI: 10.3390/ani13142358] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Avian leukosis viruses (ALVs) have been virtually eradicated from commercial poultry. However, some niches remain as pockets from which this group of viruses may reemerge and induce economic losses. Such is the case of fancy, hobby, backyard chickens and indigenous or native breeds, which are not as strictly inspected as commercial poultry and which have been found to harbor ALVs. In addition, the genome of both poultry and of several gamebird species contain endogenous retroviral sequences. Circumstances that support keeping up surveillance include the detection of several ALV natural recombinants between exogenous and endogenous ALV-related sequences which, combined with the well-known ability of retroviruses to mutate, facilitate the emergence of escape mutants. The subgroup most prevalent nowadays, ALV-J, has emerged as a multi-recombinant which uses a different receptor from the previously known subgroups, greatly increasing its cell tropism and pathogenicity and making it more transmissible. In this review we describe the ALVs, their different subgroups and which receptor they use to infect the cell, their routes of transmission and their presence in different bird collectivities, and the immune response against them. We analyze the different systems to control them, from vaccination to the progress made editing the bird genome to generate mutated ALV receptors or selecting certain haplotypes.
Collapse
Affiliation(s)
- Sergio Fandiño
- Department of Animal Health, Veterinary Faculty, Complutense University of Madrid, Av. Puerta de Hierro s/n, 28040 Madrid, Spain
- Department of Genetics, Physiology and Microbiology, Faculty of Biological Sciences, Complutense University of Madrid (UCM), C. de José Antonio Novais 12, 28040 Madrid, Spain
- Research Group, "Animal Viruses" of Complutense University of Madrid, 28040 Madrid, Spain
| | - Esperanza Gomez-Lucia
- Department of Animal Health, Veterinary Faculty, Complutense University of Madrid, Av. Puerta de Hierro s/n, 28040 Madrid, Spain
- Research Group, "Animal Viruses" of Complutense University of Madrid, 28040 Madrid, Spain
| | - Laura Benítez
- Department of Genetics, Physiology and Microbiology, Faculty of Biological Sciences, Complutense University of Madrid (UCM), C. de José Antonio Novais 12, 28040 Madrid, Spain
- Research Group, "Animal Viruses" of Complutense University of Madrid, 28040 Madrid, Spain
| | - Ana Doménech
- Department of Animal Health, Veterinary Faculty, Complutense University of Madrid, Av. Puerta de Hierro s/n, 28040 Madrid, Spain
- Research Group, "Animal Viruses" of Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
4
|
Luo H, Hu X, Wu H, Zaib G, Chai W, Cui H. Activation of lnc-ALVE1-AS1 inhibited ALV-J replication through triggering the TLR3 pathway in chicken macrophage like cell line. Vet Res Commun 2023; 47:431-443. [PMID: 35715584 DOI: 10.1007/s11259-022-09960-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 06/14/2022] [Indexed: 11/27/2022]
Abstract
Endogenous retroviruses (ERVs) are remnants of the historical retroviral infections, and their derived transcripts with viral signatures are important sources of long noncoding RNAs (lncRNAs). We have previously shown that the chicken ERV-derived lncRNA lnc-ALVE1-AS1 exerts antiviral innate immunity in chicken embryo fibroblasts. However, it is not clear whether this endogenous retroviral RNA has a similar function in immune cells. Here, we found that lnc-ALVE1-AS1 was persistently inhibited in chicken macrophages after avian leukosis virus subgroup J (ALV-J) infection. Furthermore, overexpression of lnc-ALVE1-AS1 significantly inhibited the replication of exogenous ALV-J, whereas knockdown of lnc-ALVE1-AS1 promoted the replication of ALV-J in chicken macrophages. This phenomenon is attributed to the induction of antiviral innate immunity by lnc-ALVE1-AS1 in macrophages, whereas knockdown of lnc-ALVE1-AS1 had the opposite effect. Mechanistically, lnc-ALVE1-AS1 can be sensed by the cytosolic pattern recognition receptor TLR3 and trigger the type I interferons response. The present study provides novel insights into the antiviral defense of ERV-derived lncRNAs in macrophages and offers new strategies for future antiviral solutions.
Collapse
Affiliation(s)
- Huan Luo
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, No. 48 East Wenhui Road, Yangzhou, 225009, Jiangsu, China
| | - Xuming Hu
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, No. 48 East Wenhui Road, Yangzhou, 225009, Jiangsu, China.
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, 225009, Jiangsu, China.
- Jiangsu Co-Innovation Center for Prevention & Control of Important Animal Infectious Diseases & Zoonoses, Yangzhou, 225009, Jiangsu, China.
| | - Huixian Wu
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, No. 48 East Wenhui Road, Yangzhou, 225009, Jiangsu, China
| | - Gul Zaib
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, China
| | - Wenxian Chai
- Changzhou Animal Disease Prevention and Control Center, Changzhou, 213002, Jiangsu, China
| | - Hengmi Cui
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, No. 48 East Wenhui Road, Yangzhou, 225009, Jiangsu, China
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou, 225009, Jiangsu, China
- Jiangsu Co-Innovation Center for Prevention & Control of Important Animal Infectious Diseases & Zoonoses, Yangzhou, 225009, Jiangsu, China
| |
Collapse
|
5
|
Synergistic Immunosuppression of Avian Leukosis Virus Subgroup J and Infectious Bursal Disease Virus Is Responsible for Enhanced Pathogenicity. Viruses 2022; 14:v14102312. [PMID: 36298866 PMCID: PMC9608456 DOI: 10.3390/v14102312] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/09/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
In recent years, superinfections of avian leukosis virus subgroup J (ALV-J) and infectious bursal disease virus (IBDV) have been frequently observed in nature, which has led to the increasing virulence in infected chickens. However, the reason for the enhanced pathogenicity has remained unclear. In this study, we demonstrated an effective candidate model for studying the outcome of superinfections with ALV-J and IBDV in cells and specific-pathogen-free (SPF) chicks. Through in vitro experiments, we found that ALV-J and IBDV can establish the superinfection models and synergistically promote the expression of IL-6, IL-10, IFN-α, and IFN-γ in DF-1 and CEF cells. In vivo, the weight loss, survival rate, and histopathological observations showed that more severe pathogenicity was present in the superinfected chickens. In addition, we found that superinfections of ALV-J and IBDV synergistically increased the viral replication of the two viruses and inflammatory mediator secretions in vitro and in vivo. Moreover, by measuring the immune organ indexes and blood proportions of CD3+, CD4+, and CD8α+ cells, our results showed that the more severe instances of immunosuppression were observed in the superinfected chickens. In the present study, we concluded that the more severe immunosuppression induced by the synergistic viral replication of ALV-J and IBDV is responsible for the enhanced pathogenicity.
Collapse
|
6
|
Tang S, Li J, Chang YF, Lin W. Avian Leucosis Virus-Host Interaction: The Involvement of Host Factors in Viral Replication. Front Immunol 2022; 13:907287. [PMID: 35693802 PMCID: PMC9178239 DOI: 10.3389/fimmu.2022.907287] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Avian leukosis virus (ALV) causes various diseases associated with tumor formation and decreased fertility. Moreover, ALV induces severe immunosuppression, increasing susceptibility to other microbial infections and the risk of failure in subsequent vaccination against other diseases. There is growing evidence showing the interaction between ALV and the host. In this review, we will survey the present knowledge of the involvement of host factors in the important molecular events during ALV infection and discuss the futuristic perspectives from this angle.
Collapse
Affiliation(s)
- Shuang Tang
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction of Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jie Li
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Yung-Fu Chang
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Wencheng Lin
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction of Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
7
|
Zhao Q, Yao Z, Chen L, He Y, Xie Z, Zhang H, Lin W, Chen F, Xie Q, Zhang X. Transcriptome-Wide Dynamics of m6A Methylation in Tumor Livers Induced by ALV-J Infection in Chickens. Front Immunol 2022; 13:868892. [PMID: 35529873 PMCID: PMC9072629 DOI: 10.3389/fimmu.2022.868892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/28/2022] [Indexed: 11/26/2022] Open
Abstract
Avian Leukosis Virus Subgroup J (ALV-J) is a tumorigenic virus with high morbidity and rapid transmission. N6-methyladenosine (m6A) is a common epigenetic modification that may be closely related to the pathogenicity of ALV-J. Currently, there are no reports on whether m6A modification is related to ALV-J induced tumor formation. In this study, we used methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA sequencing (RNA-seq) to examine the differences in m6A methylation and gene expression in normal livers and ALV-J-induced tumor livers systematically, with functional enrichment and co-expression analysis. The results identified 6,541 m6A methylated peaks, mainly enriched in CDS, and more than 83% of the transcripts contained 1-2 m6A peaks. For RNA-seq, 1,896 and 1,757 differentially expressed mRNAs and lncRNAs were identified, respectively. Gene enrichment analysis indicated that they may be involved in biological processes and pathways such as immunology-related and apoptosis. Moreover, we identified 17 lncRNAs, commonly existing in differently expressed methylome and transcriptome. Through co-expression analysis, 126 differentially expressed lncRNAs, and 18 potentially m6A-related methyltransferases were finally identified and connected, suggesting that m6A modifications might affect gene expression of lncRNAs and play a role in ALV-J induced tumor formation. This study provides the first comprehensive description of the m6A expression profile in tumor livers induced by ALV-J infection in chickens, which provides a basis for studying the role of m6A modification in ALV-J induced tumorigenesis. This study provides clues for studying the epigenetic etiology and pathogenesis of ALV-J.
Collapse
Affiliation(s)
- Qiqi Zhao
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding & Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, China
| | - Ziqi Yao
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding & Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, China
| | - Liyi Chen
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding & Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, China
| | - Yaai He
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding & Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, China
| | - Zi Xie
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding & Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou, China
| | - Huanmin Zhang
- United States Department of Agriculture (USDA), Agriculture Research Service, Avian Disease and Oncology Laboratory, East Lansing, MI, United States
| | - Wencheng Lin
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding & Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou, China
| | - Feng Chen
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding & Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou, China
| | - Qingmei Xie
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding & Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou, China
- *Correspondence: Qingmei Xie, ; Xinheng Zhang,
| | - Xinheng Zhang
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding & Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou, China
- *Correspondence: Qingmei Xie, ; Xinheng Zhang,
| |
Collapse
|
8
|
Zmrhal V, Svoradova A, Batik A, Slama P. Three-Dimensional Avian Hematopoietic Stem Cell Cultures as a Model for Studying Disease Pathogenesis. Front Cell Dev Biol 2022; 9:730804. [PMID: 35127695 PMCID: PMC8811169 DOI: 10.3389/fcell.2021.730804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 12/17/2021] [Indexed: 11/16/2022] Open
Abstract
Three-dimensional (3D) cell culture is attracting increasing attention today because it can mimic tissue environments and provide more realistic results than do conventional cell cultures. On the other hand, very little attention has been given to using 3D cell cultures in the field of avian cell biology. Although mimicking the bone marrow niche is a classic challenge of mammalian stem cell research, experiments have never been conducted in poultry on preparing in vitro the bone marrow niche. It is well known, however, that all diseases cause immunosuppression and target immune cells and their development. Hematopoietic stem cells (HSC) reside in the bone marrow and constitute a source for immune cells of lymphoid and myeloid origins. Disease prevention and control in poultry are facing new challenges, such as greater use of alternative breeding systems and expanding production of eggs and chicken meat in developing countries. Moreover, the COVID-19 pandemic will draw greater attention to the importance of disease management in poultry because poultry constitutes a rich source of zoonotic diseases. For these reasons, and because they will lead to a better understanding of disease pathogenesis, in vivo HSC niches for studying disease pathogenesis can be valuable tools for developing more effective disease prevention, diagnosis, and control. The main goal of this review is to summarize knowledge about avian hematopoietic cells, HSC niches, avian immunosuppressive diseases, and isolation of HSC, and the main part of the review is dedicated to using 3D cell cultures and their possible use for studying disease pathogenesis with practical examples. Therefore, this review can serve as a practical guide to support further preparation of 3D avian HSC niches to study the pathogenesis of avian diseases.
Collapse
Affiliation(s)
- Vladimir Zmrhal
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
| | - Andrea Svoradova
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
- NPPC, Research Institute for Animal Production in Nitra, Luzianky, Slovak Republic
| | - Andrej Batik
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
| | - Petr Slama
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
| |
Collapse
|
9
|
Xiang Y, Liang C, Li Q, Chen Q, Zhou Y, Zheng X, Zhou D, Wang Z, Wang G, Cao W. Chicken telomerase reverse transcriptase promotes the tumorigenicity of avian leukosis virus subgroup J by regulating the Wnt/β-catenin signaling pathway. Vet Res 2022; 53:100. [PMID: 36461084 PMCID: PMC9717515 DOI: 10.1186/s13567-022-01120-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/24/2022] [Indexed: 12/03/2022] Open
Abstract
This research aimed to analyze the regulatory effect of chicken telomerase reverse transcriptase (chTERT) on the Wnt/β-catenin signaling pathway and its effect on the tumorigenicity of avian leukosis virus subgroup J (ALV-J) through in vivo experiments. The chTERT eukaryotic expression plasmid and its recombinant lentivirus particles were constructed for in vivo transfection of chTERT to analyze the effect of chTERT continuously overexpressed in chickens on the tumorigenicity of ALV-J. During 156 days of the artificial ALV-J tumor-inducing process, 7 solid tumors developed in 3 chickens in the chTERT-overexpression group (n = 26*2) and no tumors developed in the control group (n = 26*2). Another 18 tumors induced by ALV-J were confirmed and collected from breeding poultry farms. And we confirmed that chTERT was significantly highly expressed in ALV-J tumors. The ELISA data suggested that the protein levels of β-catenin and c-Myc in the chicken plasma of the chTERT-overexpressing group with ALV-J infected were consistently and significantly higher than those of the control group. Compared with that of the tumor-adjacent tissues, the activity of the Wnt/β-catenin signaling pathway and expression of the c-Myc was significantly increased in ALV-J tumors. And the percentage of apoptosis in ALV-J tumors significantly lower than that in tumor-adjacent tissues. Immunohistochemistry, Western blot and RT-qPCR suggested that the replication level of ALV-J in tumors was significantly higher than that in tumor-adjacent tissues. This study suggests that chTERT plays a critical role in the tumorigenicity of ALV-J by enhancing the Wnt/β-catenin signaling pathway, which will contribute to further elucidating the tumor-inducing mechanism of ALV-J.
Collapse
Affiliation(s)
- Yong Xiang
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Canxin Liang
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Qingbo Li
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Qinxi Chen
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Yang Zhou
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Xiaoxue Zheng
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Di Zhou
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Zepeng Wang
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Guyao Wang
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Weisheng Cao
- grid.20561.300000 0000 9546 5767College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China ,grid.20561.300000 0000 9546 5767Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, 510642 China ,grid.20561.300000 0000 9546 5767National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou, 510642 China ,grid.20561.300000 0000 9546 5767Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642 China ,grid.20561.300000 0000 9546 5767Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642 China
| |
Collapse
|
10
|
Differential responses of chicken monocyte-derived dendritic cells infected with Salmonella Gallinarum and Salmonella Typhimurium. Sci Rep 2021; 11:17214. [PMID: 34446765 PMCID: PMC8390485 DOI: 10.1038/s41598-021-96527-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/11/2021] [Indexed: 11/08/2022] Open
Abstract
Salmonella enterica serovar Gallinarum is a host-restricted bacterial pathogen that causes a serious systemic disease exclusively in birds of all ages. Salmonella enterica serovar Typhimurium is a host-generalist serovar. Dendritic cells (DCs) are key antigen-presenting cells that play an important part in Salmonella host-restriction. We evaluated the differential response of chicken blood monocyte-derived dendritic cells (chMoDCs) exposed to S. Gallinarum or S. Typhimurium. S. Typhimurium was found to be more invasive while S. Gallinarum was more cytotoxic at the early phase of infection and later showed higher resistance against chMoDCs killing. S. Typhimurium promoted relatively higher upregulation of costimulatory and other immune function genes on chMoDCs in comparison to S. Gallinarum during early phase of infection (6 h) as analyzed by real-time PCR. Both Salmonella serovars strongly upregulated the proinflammatory transcripts, however, quantum was relatively narrower with S. Gallinarum. S. Typhimurium-infected chMoDCs promoted relatively higher proliferation of naïve T-cells in comparison to S. Gallinarum as assessed by mixed lymphocyte reaction. Our findings indicated that host restriction of S. Gallinarum to chicken is linked with its profound ability to interfere the DCs function. Present findings provide a valuable roadmap for future work aimed at improved vaccine strategies against this pathogen.
Collapse
|
11
|
Xiang Y, Yu Y, Li Q, Jiang Z, Li J, Liang C, Chen J, Li Y, Chen X, Cao W. Mutual regulation between chicken telomerase reverse transcriptase and the Wnt/β-catenin signalling pathway inhibits apoptosis and promotes the replication of ALV-J in LMH cells. Vet Res 2021; 52:110. [PMID: 34412690 PMCID: PMC8375160 DOI: 10.1186/s13567-021-00979-x] [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: 05/08/2021] [Accepted: 07/10/2021] [Indexed: 12/14/2022] Open
Abstract
This study aimed to explore the mutual regulation between chicken telomerase reverse transcriptase (chTERT) and the Wnt/β-catenin signalling pathway and its effects on cell growth and avian leukosis virus subgroup J (ALV-J) replication in LMH cells. First, LMH cells stably overexpressing the chTERT gene (LMH-chTERT cells) and corresponding control cells (LMH-NC cells) were successfully constructed with a lentiviral vector expression system. The results showed that chTERT upregulated the expression of β-catenin, Cyclin D1, TCF4 and c-Myc. chTERT expression level and telomerase activity were increased when cells were treated with LiCl. When the cells were treated with ICG001 or IWP-2, the activity of the Wnt/β-catenin signalling pathway was significantly inhibited, and chTERT expression and telomerase activity were also inhibited. However, when the β-catenin gene was knocked down by small interfering RNA (siRNA), the changes in chTERT expression and telomerase activity were consistent with those in cells treated with ICG001 or IWP-2. These results indicated that chTERT and the Wnt/β-catenin signalling pathway can be mutually regulated. Subsequently, we found that chTERT not only shortened the cell cycle to promote proliferation but also inhibited apoptosis by downregulating the expression of Caspase 3, Caspase 9 and BAX; upregulating BCL-2 and BCL-X expression; and promoting autophagy. Moreover, chTERT significantly enhanced the migration ability of LMH cells, upregulated the protein and mRNA expression of ALV-J and increased the virus titre. ALV-J replication promoted chTERT expression and telomerase activity.
Collapse
Affiliation(s)
- Yong Xiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yun Yu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Qingbo Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zeng Jiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Jinqun Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Canxin Liang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Jian Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Xiaoyan Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Weisheng Cao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China. .,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, 510642, China. .,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China. .,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou, 510642, China. .,Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture, Guangzhou, 510642, China.
| |
Collapse
|
12
|
Abstract
Viral infections lead to the death of more than a million people each year around the world, both directly and indirectly. Viruses interfere with many cell functions, particularly critical pathways for cell death, by affecting various intracellular mediators. MicroRNAs (miRNAs) are a major example of these mediators because they are involved in many (if not most) cellular mechanisms. Virus-regulated miRNAs have been implicated in three cell death pathways, namely, apoptosis, autophagy, and anoikis. Several molecules (e.g., BECN1 and B cell lymphoma 2 [BCL2] family members) are involved in both apoptosis and autophagy, while activation of anoikis leads to cell death similar to apoptosis. These mechanistic similarities suggest that common regulators, including some miRNAs (e.g., miR-21 and miR-192), are involved in different cell death pathways. Because the balance between cell proliferation and cell death is pivotal to the homeostasis of the human body, miRNAs that regulate cell death pathways have drawn much attention from researchers. miR-21 is regulated by several viruses and can affect both apoptosis and anoikis via modulating various targets, such as PDCD4, PTEN, interleukin (IL)-12, Maspin, and Fas-L. miR-34 can be downregulated by viral infection and has different effects on apoptosis, depending on the type of virus and/or host cell. The present review summarizes the existing knowledge on virus-regulated miRNAs involved in the modulation of cell death pathways. Understanding the mechanisms for virus-mediated regulation of cell death pathways could provide valuable information to improve the diagnosis and treatment of many viral diseases.
Collapse
|
13
|
Miretti S, Lecchi C, Ceciliani F, Baratta M. MicroRNAs as Biomarkers for Animal Health and Welfare in Livestock. Front Vet Sci 2020; 7:578193. [PMID: 33392281 PMCID: PMC7775535 DOI: 10.3389/fvets.2020.578193] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/30/2020] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are small and highly conserved non-coding RNA molecules that orchestrate a wide range of biological processes through the post-transcriptional regulation of gene expression. An intriguing aspect in identifying these molecules as biomarkers is derived from their role in cell-to-cell communication, their active secretion from cells into the extracellular environment, their high stability in body fluids, and their ease of collection. All these features confer on miRNAs the potential to become a non-invasive tool to score animal welfare. There is growing interest in the importance of miRNAs as biomarkers for assessing the welfare of livestock during metabolic, environmental, and management stress, particularly in ruminants, pigs, and poultry. This review provides an overview of the current knowledge regarding the potential use of tissue and/or circulating miRNAs as biomarkers for the assessment of the health and welfare status in these livestock species.
Collapse
Affiliation(s)
- Silvia Miretti
- Department of Veterinary Sciences, University of Torino, Grugliasco, Italy
| | - Cristina Lecchi
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy
| | - Fabrizio Ceciliani
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy
| | - Mario Baratta
- Department of Veterinary Sciences, University of Torino, Grugliasco, Italy
| |
Collapse
|
14
|
Epigenetic Regulation by Non-Coding RNAs in the Avian Immune System. Life (Basel) 2020; 10:life10080148. [PMID: 32806547 PMCID: PMC7459779 DOI: 10.3390/life10080148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/20/2022] Open
Abstract
The identified non-coding RNAs (ncRNAs) include circular RNAs, long non-coding RNAs, microRNAs, ribosomal RNAs, small interfering RNAs, small nuclear RNAs, piwi-interacting RNAs, and transfer RNAs, etc. Among them, long non-coding RNAs, circular RNAs, and microRNAs are regulatory RNAs that have different functional mechanisms and were extensively participated in various biological processes. Numerous research studies have found that circular RNAs, long non-coding RNAs, and microRNAs played their important roles in avian immune system during the infection of parasites, virus, or bacterium. Here, we specifically review and expand this knowledge with current advances of circular RNAs, long non-coding RNAs, and microRNAs in the regulation of different avian diseases and discuss their functional mechanisms in response to avian diseases.
Collapse
|
15
|
Duan X, Wang L, Sun G, Yan W, Yang Y. Understanding the cross-talk between host and virus in poultry from the perspectives of microRNA. Poult Sci 2020; 99:1838-1846. [PMID: 32241464 PMCID: PMC7587795 DOI: 10.1016/j.psj.2019.11.053] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 11/18/2019] [Accepted: 11/18/2019] [Indexed: 01/05/2023] Open
Abstract
In poultry, viral infections (e.g., Marek's disease virus, avian leukosis virus, influenza A virus, and so on) can cause devastating mortality and economic losses. Because viruses are solely dependent on host cells to propagate, they alter the host intracellular microenvironment. Thus, understanding the virus-host interaction is important for antiviral immunity and drug development in the poultry industry. MicroRNAs are crucial posttranscriptional regulators of gene expression in a wide spectrum of biological processes, including viral infection. Recently, microRNAs have been identified as key players in virus-host interactions. In this review, we will discuss the intricacies involved in the virus-host cross-talk mediated by host and viral microRNAs in poultry (i.e., chicken and ducks), as well as recent trends and challenges in this field. These findings may provide some insights into the rapidly developing area of research regarding viral pathogenesis and antiviral immunity in poultry production.
Collapse
Affiliation(s)
- Xiujun Duan
- Department of Animal Science and Technology, Jiangsu Agri-animal Husbandry Vocational College, Taizhou 225300, China; National Gene Bank of Waterfowl Resources, Taizhou 225300, China
| | - Lihua Wang
- Department of Animal Science and Technology, Jiangsu Agri-animal Husbandry Vocational College, Taizhou 225300, China
| | - Guobo Sun
- Department of Animal Science and Technology, Jiangsu Agri-animal Husbandry Vocational College, Taizhou 225300, China; National Gene Bank of Waterfowl Resources, Taizhou 225300, China
| | - Wenying Yan
- School of Biology and Basic Medical Sciences, Medical College of Soochow University, Suzhou 215123, China.
| | - Yang Yang
- School of Computer Science and Technology, Soochow University, Suzhou 215123, China.
| |
Collapse
|
16
|
Abstract
MicroRNAs (miRNAs) are small, non-coding RNA molecules that inhibit protein translation from target mRNAs. Accumulating evidence suggests that miRNAs can regulate a broad range of biological pathways, including cell differentiation, apoptosis, and carcinogenesis. With the development of miRNAs, the investigation of miRNA functions has emerged as a hot research field. Due to the intensive farming in recent decades, chickens are easily influenced by various pathogen transmissions, and this has resulted in large economic losses. Recent reports have shown that miRNAs can play critical roles in the regulation of chicken diseases. Therefore, the aim of this review is to briefly discuss the current knowledge regarding the effects of miRNAs on chickens suffering from common viral diseases, mycoplasmosis, necrotic enteritis, and ovarian tumors. Additionally, the detailed targets of miRNAs and their possible functions are also summarized. This review intends to highlight the key role of miRNAs in regard to chickens and presents the possibility of improving chicken disease resistance through the regulation of miRNAs.
Collapse
|
17
|
Dai M, Feng M, Xie T, Zhang X. Long non-coding RNA and MicroRNA profiling provides comprehensive insight into non-coding RNA involved host immune responses in ALV-J-infected chicken primary macrophage. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 100:103414. [PMID: 31200006 DOI: 10.1016/j.dci.2019.103414] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/10/2019] [Accepted: 06/10/2019] [Indexed: 06/09/2023]
Abstract
Avian leukosis virus subgroup J (ALV-J) infection can cause tumors and immunosuppression in infected chickens. Macrophages play a crucial role in host defense against invading pathogens. In the present study, whole transcriptome analysis was performed to analyze the host factors including genes, microRNA (miRNA), long non-coding RNA (lncRNA) and their regulatory network in chicken primary monocyte-derived macrophages (MDMs). In total, 128 differentially expressed (DE) lncRNAs and 15 DE miRNAs were identified in MDMs at 3 h post infection (hpi), and 30 DE lncRNAs and 8 DE miRNAs were identified in MDMs at 36 hpi during ALV-J infection. We further constructed the DE lncRNAs-mRNAs, miRNA-mRNAs and lncRNAs-miRNA-mRNAs interaction networks. The results suggested that DE lncRNAs and miRNAs are involved in the regulation of CCND3 and SOCS5 in Jak-STAT signaling pathway via ceRNA network in ALV-J-infected MDMs at 3 hpi. In addition, lncRNAs including XLOC_672329, ALDBGALG0000001429, XLOC_016500 and ALDBGALG0000000253 cis-regulating CH25H, CISH, IL-1β and CD80 respectively in MDMs at 3 hpi participated in host antiviral responses. Our findings give a comprehensive view of the connection between non-coding RNA and ALV-J in chicken primary macrophages, and provide an excellent resource for further studies of epigenetic effects on ALV-J disease resistance breeding as well as immune system and genomic researches.
Collapse
Affiliation(s)
- Manman Dai
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People's Republic of China.
| | - Min Feng
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China.
| | - Tingting Xie
- 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, 510642, Guangdong, 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, 510642, Guangdong, China.
| |
Collapse
|
18
|
Qiu L, Chang G, Bi Y, Liu X, Chen G. Circular RNA and mRNA profiling reveal competing endogenous RNA networks during avian leukosis virus, subgroup J-induced tumorigenesis in chickens. PLoS One 2018; 13:e0204931. [PMID: 30286182 PMCID: PMC6171863 DOI: 10.1371/journal.pone.0204931] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 09/17/2018] [Indexed: 01/30/2023] Open
Abstract
Avian leukosis virus subgroup J (ALV-J) can induce myeloid tumors and hemangiomas in chickens and causes severe economic losses with commercial layer chickens and meat-type chickens. Here, we generated ribominus RNA sequencing data from three normal chicken spleen tissues and three ALV-J-infected chicken spleen tissues. Structure analysis of transcripts showed that, compared to mRNAs and lncRNAs, chicken circRNAs shared relatively shorter transcripts and similar GC content. Differentially expression analysis showed 152 differentially expressed circRNAs with 106 circRNAs up regulated and 46 circRNAs down regulated. Through comparing differentially expressed circRNA host genes and mRNAs and performed ceRNA network analysis, we found several tumor or immune-related genes, in which, there were four genes existed in both differentially expressed mRNAs and circRNA host genes (Dock4, Fmr1, Zfhx3, Ralb) and two genes (Mll, Aoc3) involved in ceRNA network. We further characterized one exon-intron circRNA derived from HRH4 gene in the ceRNA network, termed circHRH4, which is an abundant and stable circRNA expressed in various tissues and cells in chicken and localizes in cytoplasm. Our results provide new insight into the pathology of ALV-J infection and circRNAs may also mediate tumorigenesis in chicken.
Collapse
Affiliation(s)
- Lingling Qiu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR, China
| | - Guobin Chang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR, China
| | - Yulin Bi
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR, China
| | - Xiangping Liu
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, PR, China
| | - Guohong Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR, China
| |
Collapse
|
19
|
Qiu L, Chang G, Li Z, Bi Y, Liu X, Chen G. Comprehensive Transcriptome Analysis Reveals Competing Endogenous RNA Networks During Avian Leukosis Virus, Subgroup J-Induced Tumorigenesis in Chickens. Front Physiol 2018; 9:996. [PMID: 30093865 PMCID: PMC6070742 DOI: 10.3389/fphys.2018.00996] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 07/06/2018] [Indexed: 11/26/2022] Open
Abstract
Avian leukosis virus subgroup J (ALV-J) is an avian oncogenic retrovirus that induces myeloid tumors and hemangiomas in chickens and causes severe economic losses with commercial layer chickens and meat-type chickens. High-throughput sequencing followed by quantitative real-time polymerase chain reaction and bioinformatics analyses were performed to advance the understanding of regulatory networks associated with differentially expressed non-coding RNAs and mRNAs that facilitate ALV-J infection. We examined the expression of mRNAs, long non-coding RNAs (lncRNAs), and miRNAs in the spleens of 20-week-old chickens infected with ALV-J and uninfected chickens. We found that 1723 mRNAs, 7,883 lncRNAs and 13 miRNAs in the spleen were differentially expressed between the uninfected and infected groups (P < 0.05). Transcriptome analysis showed that, compared to mRNA, chicken lncRNAs shared relatively fewer exon numbers and shorter transcripts. Through competing endogenous RNA and co-expression network analyses, we identified several tumor-associated or immune-related genes and lncRNAs. Along transcripts whose expression levels significantly decreased in both ALV-J infected spleen and tumor tissues, BCL11B showed the greatest change. These results suggest that BCL11B may be mechanistically involved in tumorigenesis in chicken and neoplastic diseases, may be related to immune response, and potentially be novel biomarker for ALV-J infection. Our results provide new insight into the pathology of ALV-J infection and high-quality transcriptome resource for in-depth study of epigenetic influences on disease resistance and immune system.
Collapse
Affiliation(s)
- Lingling Qiu
- Key Laboratory of Animal Genetics and Breeding, Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou, China
| | - Guobin Chang
- Key Laboratory of Animal Genetics and Breeding, Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou, China
| | - Zhiteng Li
- Key Laboratory of Animal Genetics and Breeding, Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou, China
| | - Yulin Bi
- Key Laboratory of Animal Genetics and Breeding, Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou, China
| | - Xiangping Liu
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China
| | - Guohong Chen
- Key Laboratory of Animal Genetics and Breeding, Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou, China
| |
Collapse
|
20
|
Crispo M, Shivaprasad HL, Cooper GL, Bickford AA, Stoute ST. Streptococcosis in Commercial and Noncommercial Avian Species in California: 95 Cases (2000–2017). Avian Dis 2018; 62:152-162. [DOI: 10.1637/11765-103117-reg.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Manuela Crispo
- University of California, Davis, California Animal Health and Food Safety Laboratory SystemTurlock Branch, 1550 N. Soderquist Road, Turlock, CA 95380
| | | | - George L. Cooper
- University of California, Davis, California Animal Health and Food Safety Laboratory SystemTurlock Branch, 1550 N. Soderquist Road, Turlock, CA 95380
| | - Arthur A. Bickford
- University of California, Davis, California Animal Health and Food Safety Laboratory SystemTurlock Branch, 1550 N. Soderquist Road, Turlock, CA 95380
| | - Simone T. Stoute
- University of California, Davis, California Animal Health and Food Safety Laboratory SystemTurlock Branch, 1550 N. Soderquist Road, Turlock, CA 95380
| |
Collapse
|
21
|
Lin W, Xu Z, Yan Y, Zhang H, Li H, Chen W, Chen F, Xie Q. Avian Leukosis Virus Subgroup J Attenuates Type I Interferon Production Through Blocking IκB Phosphorylation. Front Microbiol 2018; 9:1089. [PMID: 29887850 PMCID: PMC5980975 DOI: 10.3389/fmicb.2018.01089] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022] Open
Abstract
Avian leukosis virus subgroup J (ALV-J) is an oncogenic retrovirus that causes immunosuppression and enhances susceptibility to secondary infection, resulting in great economic losses. Although ALV-J-induced immunosuppression has been well established, the underlying molecular mechanism for such induction is still unclear. Here, we report that the inhibitory effect of ALV-J infection on type I interferon expression is associated with the down-regulation of transcriptional regulator NF-κB in host cells. We found that ALV-J possess the inhibitory effect on type I interferon production in HD11 cells and that ALV-J causes the up-regulation of IκBα and down-regulation of NF-κB p65, and that ALV-J blocks the phosphorylation of IκBα on Ser32/36 amino acid residues. Collectively, our findings provide insights into the pathogenesis of ALV-J.
Collapse
Affiliation(s)
- Wencheng Lin
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China.,South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, China.,Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou, China
| | - Zhouyi Xu
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yiming Yan
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Huanmin Zhang
- Avian Disease and Oncology Laboratory, USDA, Agriculture Research Service, East Lansing, MI, United States
| | - Hongxin Li
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China.,South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, China.,Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou, China
| | - Weiguo Chen
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China.,South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, China.,Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou, China
| | - Feng Chen
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China.,South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, China.,Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou, China
| | - Qingmei Xie
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China.,South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, China.,Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou, China
| |
Collapse
|
22
|
Ren C, Yu M, Zhang Y, Fan M, Chang F, Xing L, Liu Y, Wang Y, Qi X, Liu C, Zhang Y, Cui H, Li K, Gao L, Pan Q, Wang X, Gao Y. Avian leukosis virus subgroup J promotes cell proliferation and cell cycle progression through miR-221 by targeting CDKN1B. Virology 2018; 519:121-130. [PMID: 29698854 DOI: 10.1016/j.virol.2018.04.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/08/2018] [Accepted: 04/10/2018] [Indexed: 01/02/2023]
Abstract
Avian leukosis virus subgroup J (ALV-J), a highly oncogenic retrovirus, causes leukemia-like proliferative diseases in chickens. microRNAs post-transcriptionally suppress targets and are involved in the development of various tumors. We previously showed that miR-221 is upregulated in ALV-J-induced tumors. In this study, we analyzed the possible function of miR-221 in ALV-J tumorigenesis. The target validation system showed that CDKN1B is a target of miR-221 and is downregulated in ALV-J infection. As CDKN1B arrests the cell cycle and regulates its progression, we analyzed the proliferation of ALV-J-infected DF-1 cells. ALV-J-infection-induced DF1 cell derepression of G1/S transition and overproliferation required high miR-221 expression followed by CDKN1B downregulation. Cell cycle pathway analysis showed that ALV-J infection induced DF-1 cell overproliferation via the CDKN1B-CDK2/CDK6 pathway. Thus, miR-221 may play an important role in ALV-J-induced aggressive growth of DF-1 cells; these findings have expanded our insights into the mechanism underlying ALV-J infection and tumorigenesis.
Collapse
Affiliation(s)
- Chaoqi Ren
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, PR China
| | - Mengmeng Yu
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, PR China
| | - Yao Zhang
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, PR China
| | - Minghui Fan
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, PR China
| | - Fangfang Chang
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, PR China
| | - Lixiao Xing
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, PR China
| | - Yongzhen Liu
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, PR China
| | - Yongqiang Wang
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, PR China
| | - Xiaole Qi
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, PR China
| | - Changjun Liu
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, PR China
| | - Yanping Zhang
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, PR China
| | - Hongyu Cui
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, PR China
| | - Kai Li
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, PR China
| | - Li Gao
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, PR China
| | - Qing Pan
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, PR China
| | - Xiaomei Wang
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou 225009, PR China.
| | - Yulong Gao
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, PR China.
| |
Collapse
|
23
|
Xiang B, Zhu W, Li Y, Gao P, Liang J, Liu D, Ding C, Liao M, Kang Y, Ren T. Immune responses of mature chicken bone-marrow-derived dendritic cells infected with Newcastle disease virus strains with differing pathogenicity. Arch Virol 2018; 163:1407-1417. [PMID: 29397456 DOI: 10.1007/s00705-018-3745-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 01/09/2018] [Indexed: 12/13/2022]
Abstract
Infection of chickens with virulent Newcastle disease virus (NDV) is associated with severe pathology and increased morbidity and mortality. The innate immune response contributes to the pathogenicity of NDV. As professional antigen-presenting cells, dendritic cells (DCs) play a unique role in innate immunity. However, the contribution of DCs to NDV infection has not been investigated in chickens. In this study, we selected two representative NDV strains, i.e., the velogenic NDV strain Chicken/Guangdong/GM/2014 (GM) and the lentogenic NDV strain La Sota, to investigate whether NDVs could infect LPS-activated chicken bone-derived marrow DCs (mature chicken BM-DCs). We compared the viral titres and innate immune responses in mature chicken BM-DCs following infection with those strains. Both NDV strains could infect mature chicken BM-DC, but the GM strain showed stronger replication capacity than the La Sota strain in mature chicken BM-DCs. Gene expression profiling showed that MDA5, LGP2, TLR3, TLR7, IFN-α, IFN-β, IFN-γ, IL-1β, IL-6, IL-18, IL-8, CCL5, IL-10, IL-12, MHC-I, and MHC-II levels were altered in mature DCs after infection with NDVs at all evaluated times postinfection. Notably, the GM strain triggered stronger innate immune responses than the La Sota strain in chicken BM-DCs. However, both strains were able to suppress the expression of some cytokines, such as IL-6 and IFN-α, in mature chicken DCs at 24 hpi. These data provide a foundation for further investigation of the role of chicken DCs in NDV infection.
Collapse
Affiliation(s)
- Bin Xiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People's Republic of China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People's Republic of China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, People's Republic of China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People's Republic of China
| | - Wenxian Zhu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People's Republic of China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People's Republic of China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, People's Republic of China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People's Republic of China
| | - Yaling Li
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, People's Republic of China
| | - Pei Gao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People's Republic of China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People's Republic of China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, People's Republic of China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People's Republic of China
| | - Jianpeng Liang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People's Republic of China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People's Republic of China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, People's Republic of China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People's Republic of China
| | - Di Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People's Republic of China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People's Republic of China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, People's Republic of China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People's Republic of China
| | - Chan Ding
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People's Republic of China
| | - Ming Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People's Republic of China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People's Republic of China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, People's Republic of China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People's Republic of China
| | - Yinfeng Kang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People's Republic of China.
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People's Republic of China.
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, People's Republic of China.
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People's Republic of China.
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China.
| | - Tao Ren
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, People's Republic of China.
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People's Republic of China.
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, People's Republic of China.
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People's Republic of China.
| |
Collapse
|
24
|
ALV-J infection induces chicken monocyte death accompanied with the production of IL-1β and IL-18. Oncotarget 2017; 8:99889-99900. [PMID: 29245947 PMCID: PMC5725138 DOI: 10.18632/oncotarget.21906] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/23/2017] [Indexed: 12/21/2022] Open
Abstract
Immunosuppression induced by avian leukosis virus subgroup J (ALV-J) causes serious reproduction problems and secondary infections in chickens. Given that monocytes are important precursors of immune cells including macrophages and dendritic cells, we investigated the fate of chicken monocytes after ALV-J infection. Our results indicated that most monocytes infected with ALV-J including field or laboratory strains could not successfully differentiate into macrophages due to cells death. And cells death was dependent upon viral titer and accompanied with increased IL-1β and IL-18 mRNA levels. In addition, ALV-J infection up-regulated caspase-1 and caspase-3 activity in monocytes. Collectively, we found that ALV-J could cause cell death in chicken monocytes, especially pyroptosis, which may be a significant reason for ALV-J induced immunosuppression.
Collapse
|
25
|
Zi XD, Lu JY, Ma L. Identification and comparative analysis of the ovarian microRNAs of prolific and non-prolific goats during the follicular phase using high-throughput sequencing. Sci Rep 2017; 7:1921. [PMID: 28507337 PMCID: PMC5432505 DOI: 10.1038/s41598-017-02225-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 04/07/2017] [Indexed: 01/22/2023] Open
Abstract
The kidding rate is one of the most important economic traits for goat production, but the genetic mechanism that is associated with ovulation rate is poorly understood. Recently, increasing evidence has suggested that microRNAs (miRNAs) influence ovarian biological processes. The present study provides the first comparison of the ovarian miRNAs of prolific Jintang black goats (JTGs) and non-prolific Tibetan goats (TBGs) during the follicular phase using RNA-Seq technology. We generated 11.19 million (M) and 11.34 M clean reads from the TBG and JTG libraries, respectively, from which a total of 389 known miRNAs were identified and 142 novel miRNAs were predicted. A total of 191 miRNAs were differentially expressed between the two breeds. Among the 10 most abundant miRNAs, miR-21-5p was defined as differentially expressed miRNA with a higher level in the JTG library than in the TBG library, but the other miRNAs were not different between the breeds. The predicted miRNA-targeted genes were further analyzed by Gene Ontology and KEGG pathway analyses. The results revealed that miR-21, miR-99a, miRNA-143, let-7f, miR-493 and miR-200b may affect follicular development. These findings will increase the current understanding of the role of ovarian miRNAs in the regulation of ovulation rate in goats.
Collapse
Affiliation(s)
- Xiang-Dong Zi
- Key-Laboratory for Animal Science of State Ethnic Affairs Commission, Southwest University for Nationalities, Chengdu, 610041, P.R. China.
| | - Jian-Yuan Lu
- Key-Laboratory for Animal Science of State Ethnic Affairs Commission, Southwest University for Nationalities, Chengdu, 610041, P.R. China
| | - Li Ma
- Key-Laboratory for Animal Science of State Ethnic Affairs Commission, Southwest University for Nationalities, Chengdu, 610041, P.R. China
| |
Collapse
|
26
|
Feng M, Zhang X. Immunity to Avian Leukosis Virus: Where Are We Now and What Should We Do? Front Immunol 2016; 7:624. [PMID: 28066434 PMCID: PMC5174080 DOI: 10.3389/fimmu.2016.00624] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 12/08/2016] [Indexed: 12/16/2022] Open
Abstract
Avian leukosis virus (ALV) is an avian oncogenic retrovirus causing enormous economic losses in the global poultry industry. Although ALV-related research has lasted for more than a century, there are no vaccines to protect chickens from ALV infection. The interaction between chickens and ALV remains not fully understood especially with regard to the host immunity. The current review provides an overview of our current knowledge of innate and adaptive immunity induced by ALV infection. More importantly, we have pointed out the unknown area involved in ALV-related studies, which is worthy of our serious exploring in future.
Collapse
Affiliation(s)
- Min Feng
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China
| | - Xiquan Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China
| |
Collapse
|
27
|
Sun Y, Du T, Liu B, Syed SF, Chen Y, Li H, Wang X, Zhang G, Zhou EM, Zhao Q. Seroprevalence of avian hepatitis E virus and avian leucosis virus subgroup J in chicken flocks with hepatitis syndrome, China. BMC Vet Res 2016; 12:261. [PMID: 27876045 PMCID: PMC5120538 DOI: 10.1186/s12917-016-0892-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 11/17/2016] [Indexed: 01/11/2023] Open
Abstract
Background From 2014 to 2015 in China, many broiler breeder and layer hen flocks exhibited a decrease in egg production and some chickens developed hepatitis syndrome including hepatomegaly, hepatic necrosis and hemorrhage. Avian hepatitis E virus (HEV) and avian leucosis virus subgroup J (ALV-J) both cause decreasing in egg production, hepatomegaly and hepatic hemorrhage in broiler breeder and layer hens. In the study, the seroprevalence of avian HEV and ALV-J in these flocks emerging the disease from Shandong and Shaanxi provinces were investigated. Results A total of 1995 serum samples were collected from 14 flocks with hepatitis syndrome in Shandong and Shaanxi provinces, China. Antibodies against avian HEV and ALV-J in these serum samples were detected using iELISAs. The seroprevalence of anti-avian HEV antibodies (35.09%) was significantly higher than that of anti-ALV-J antibodies (2.16%) (p = 0.00). Moreover, the 43 serum samples positive for anti-ALV-J antibodies were all also positive for anti-avian HEV antibodies. In a comparison of both provinces, Shandong chickens exhibited a significantly higher seroprevalence of anti-avian HEV antibodies (42.16%) than Shaanxi chickens (26%) (p = 0.00). In addition, the detection of avian HEV RNA and ALV-J cDNA in the liver samples from the flocks of two provinces also showed the same results of the seroprevalence. Conclusions In the present study, the results showed that avian HEV infection is widely prevalent and ALV-J infection is endemic in the flocks with hepatitis syndrome from Shandong and Shaanxi provinces of China. These results suggested that avian HEV infection may be the major cause of increased egg drop and hepatitis syndrome observed during the last 2 years in China. These results should be useful to guide development of prevention and control measures to control the diseases within chicken flocks in China. Electronic supplementary material The online version of this article (doi:10.1186/s12917-016-0892-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yani Sun
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.,Scientific Observing and Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, 712100, China
| | - Taofeng Du
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.,Scientific Observing and Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, 712100, China
| | - Baoyuan Liu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.,Scientific Observing and Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, 712100, China
| | - Shahid Faraz Syed
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.,Scientific Observing and Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, 712100, China
| | - Yiyang Chen
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.,Scientific Observing and Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, 712100, China
| | - Huixia Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.,Scientific Observing and Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, 712100, China
| | - Xinjie Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.,Scientific Observing and Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, 712100, China
| | - Gaiping Zhang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450002, China
| | - En-Min Zhou
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China. .,Scientific Observing and Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, 712100, China.
| | - Qin Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China. .,Scientific Observing and Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
28
|
Yao DW, Zhan L, Hong YF, Liu JX, Xu JR, Yang DJ. Altered expression of the mismatch repair genes in DF-1 cells infected with the avian leukosis virus subgroup A. SPRINGERPLUS 2016; 5:1756. [PMID: 27795899 PMCID: PMC5055512 DOI: 10.1186/s40064-016-3433-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 09/29/2016] [Indexed: 11/17/2022]
Abstract
The absence or deficiency of DNA mismatch repair (MMR) activity results in microsatellite instability (MSI) in cancer. The avian leukosis virus (ALV) causes neoplastic disease in chickens. In this study, the status of MMR, MSI, the cell cycle and apoptosis were detected in DF-1 cells after avian leukosis virus subgroup A infection. Flow cytometry analysis results indicated that there was no significant difference in cell apoptosis between the control and infected groups. The percentage of cells in S and G2 phases were increased in the infected group. MSI and mutation of MSH2 and MLH1 gene exons were absent in DF-1 cells after infection. Levels of MSH2 and MLH1 mRNA were dramatically increased in DF-1 cells after infection. These results demonstrated that ALV RAV-1 infection may promote the expression of MSH2 and MLH1 genes rather than resulting in gene mutations. Mismatch repair functions were normal and may be have relationships with the arrest of S phase and G2 phase.
Collapse
Affiliation(s)
- Da-Wei Yao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 Jiangsu China
| | - Li Zhan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 Jiangsu China
| | - Yu-Fang Hong
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 Jiangsu China
| | - Jian-Xin Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 Jiangsu China
| | - Jia-Rong Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 Jiangsu China
| | - De-Ji Yang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 Jiangsu China
| |
Collapse
|