1
|
He X, Zhang S, Zou Z, Gao P, Yang L, Xiang B. Antiviral Effects of Avian Interferon-Stimulated Genes. Animals (Basel) 2024; 14:3062. [PMID: 39518785 PMCID: PMC11545081 DOI: 10.3390/ani14213062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Interferons (IFNs) stimulate the expression of numerous IFN-stimulating genes via the Janus kinase-signal transducers and activators of the transcription (JAK-STAT) signaling pathway, which plays an important role in the host defense against viral infections. In mammals, including humans and mice, a substantial number of IFN-stimulated genes (ISGs) have been identified, and their molecular mechanisms have been elucidated. It is important to note that avian species are phylogenetically distant from mammals, resulting in distinct IFN-induced ISGs that may have different functions. At present, only a limited number of avian ISGs have been identified. In this review, we summarized the identified avian ISGs and their antiviral activities. As gene-editing technology is widely used in avian breeding, the identification of avian ISGs and the elucidation of their molecular mechanism may provide important support for the breeding of avians for disease resistance.
Collapse
Affiliation(s)
- Xingchen He
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (X.H.); (S.Z.); (Z.Z.); (L.Y.)
- Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Shiyuan Zhang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (X.H.); (S.Z.); (Z.Z.); (L.Y.)
- Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Ziheng Zou
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (X.H.); (S.Z.); (Z.Z.); (L.Y.)
| | - Pei Gao
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453000, China;
| | - Liangyu Yang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (X.H.); (S.Z.); (Z.Z.); (L.Y.)
- Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Bin Xiang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (X.H.); (S.Z.); (Z.Z.); (L.Y.)
- Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| |
Collapse
|
2
|
Gautam D, Sindhu A, Vats A, Rajput S, Rana C, De S. Evolutionary insights of interferon lambda genes in tetrapods. J Evol Biol 2024; 37:1101-1112. [PMID: 39066611 DOI: 10.1093/jeb/voae094] [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: 01/15/2024] [Revised: 07/07/2024] [Accepted: 07/25/2024] [Indexed: 07/28/2024]
Abstract
Type III interferon (IFN), also known as IFN-λ, is an innate antiviral protein. We retrieved the sequences of IFN-λ and their receptors from 42 tetrapod species and conducted a computational evolutionary analysis to understand the diversity of these genes. The copy number variation (CNV) of IFN-λ was determined through qPCR in Indian cattle and buffalo. The tetrapod species feature intron-containing type III IFN genes. Some reptiles and placental mammals have 2 IFN-λ loci, while marsupials, monotremes, and birds have a single IFN-λ locus. Some placental mammals and amphibians exhibit multiple IFN-λ genes, including both intron-less and intron-containing forms. Placental mammals typically possess 3-4 functional IFN-λ genes, some of them lack signal peptides. IFN-λ of these tetrapod species formed 3 major clades. Mammalian IFN-λ4 appears as an ancestral form, with syntenic conservation in most mammalian species. The intron-less IFN-λ1 and both type III IFN receptors have conserved synteny in tetrapod. Purifying selection was noted in their evolutionary analysis that plays a crucial role in minimizing genetic diversity and maintaining the integrity of biological function. This indicates that these proteins have successfully retained their biological function and indispensability, even in the presence of the type I IFNs. The expansion of IFN-λ genes in amphibians and camels have led to the evolution of multiple IFN-λ. The CNV can arise from gene duplication and conversion events. The qPCR-based absolute quantification revealed that IFN-λ3 and IFN-λ4 have more than 1 copy in buffalo (Murrah) and 6 cattle breeds (Sahiwal, Tharparkar, Kankrej, Red Sindhi, Jersey, and Holstein Friesian). Overall, these findings highlight the evolutionary diversity and functional significance of IFN-λ in tetrapod species.
Collapse
Affiliation(s)
- Devika Gautam
- Animal Genomics Lab, Animal Biotechnology Centre, ICAR-National Dairy Research Institute (NDRI), Karnal, Haryana, India
- Department of Biotechnology, Deenbandhu Chhotu Ram University of Science and Technology, Murthal, Sonipat, Haryana, India
| | - Anil Sindhu
- Department of Biotechnology, Deenbandhu Chhotu Ram University of Science and Technology, Murthal, Sonipat, Haryana, India
| | - Ashutosh Vats
- Animal Genomics Lab, Animal Biotechnology Centre, ICAR-National Dairy Research Institute (NDRI), Karnal, Haryana, India
| | - Shiveeli Rajput
- Animal Genomics Lab, Animal Biotechnology Centre, ICAR-National Dairy Research Institute (NDRI), Karnal, Haryana, India
| | - Chanchal Rana
- Animal Genomics Lab, Animal Biotechnology Centre, ICAR-National Dairy Research Institute (NDRI), Karnal, Haryana, India
| | - Sachinandan De
- Animal Genomics Lab, Animal Biotechnology Centre, ICAR-National Dairy Research Institute (NDRI), Karnal, Haryana, India
| |
Collapse
|
3
|
Gautam D, Sindhu A, Vats A, Rajput S, Roshan M, Pal H, De S. Characterization and expression profiling of buffalo IFN-lambda family. Vet Immunol Immunopathol 2024; 272:110770. [PMID: 38735115 DOI: 10.1016/j.vetimm.2024.110770] [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: 01/29/2024] [Revised: 04/12/2024] [Accepted: 04/26/2024] [Indexed: 05/14/2024]
Abstract
Interferon lambda (IFN-λ) is an important type III interferon triggered mainly by viral infection. IFN-λ binds to their heterodimeric receptors and signals through JAK-STAT pathways similar to type I IFN. In this study, we deduced the buffalo IFN-λ sequences through the polymerase chain reaction, and then studied IFN-λ's expression patterns in different tissues, and post induction with poly I:C and live MRSA using RT-qPCR. The full-length sequences of buffalo IFN-λ3, IFN-λ receptors, and a transcript variant of IFN-λ4 were determined. IFN-λ1 is identified as a pseudogene. Virus response elements and a recombination hotspot factor was observed in the regulatory region of IFN-λ. The IFN-λ3 expressed highest in lungs and monocytes but IFN-λ4 did not. The expression of Interferon Lambda Receptor 1 was tissue specific, while Interleukin 10 Receptor subunit beta was ubiquitous. Following poly I:C induction, IFN-λ3 expression was primarily observed in epithelial cells as opposed to fibroblasts, displaying cell type-dependent expression. The cytosolic RNA sensors were expressed highest in endometrial epithelial cells, whereas the endosomal receptor was higher in fibroblasts. 2',5'-oligoadenylate synthetase expressed higher in fibroblasts, myxoma resistance protein 1 and IFN-stimulated gene 56 in epithelial cells, displaying cell-specific antiviral response of the interferon stimulated genes (ISGs). The endometrial epithelial cells expressed IFN-λ3 after live S. aureus infection indicating its importance in bacterial infection. The induction of IFN-λ3 was S. aureus isolate specific at the same multiplicity of infection (MOI). This study elucidates the IFN-λ sequences, diverse expression patterns revealing tissue specificity, and specificity in response to poly I:C and bacterial stimuli, emphasising its crucial role in innate immune response modulation.
Collapse
Affiliation(s)
- Devika Gautam
- Animal Genomics Lab, Animal Biotechnology Centre, ICAR-National Dairy Research Institute (NDRI), Karnal, HR 132001, India; Deenbandhu Chhotu Ram University of Science and Technology, Murthal, Sonepat, HR 139031, India
| | - Anil Sindhu
- Deenbandhu Chhotu Ram University of Science and Technology, Murthal, Sonepat, HR 139031, India
| | - Ashutosh Vats
- Animal Genomics Lab, Animal Biotechnology Centre, ICAR-National Dairy Research Institute (NDRI), Karnal, HR 132001, India
| | - Shiveeli Rajput
- Animal Genomics Lab, Animal Biotechnology Centre, ICAR-National Dairy Research Institute (NDRI), Karnal, HR 132001, India
| | - Mayank Roshan
- Animal Genomics Lab, Animal Biotechnology Centre, ICAR-National Dairy Research Institute (NDRI), Karnal, HR 132001, India
| | - Hanshika Pal
- Animal Genomics Lab, Animal Biotechnology Centre, ICAR-National Dairy Research Institute (NDRI), Karnal, HR 132001, India
| | - Sachinandan De
- Animal Genomics Lab, Animal Biotechnology Centre, ICAR-National Dairy Research Institute (NDRI), Karnal, HR 132001, India.
| |
Collapse
|
4
|
Pang AN, Chen SN, Liu LH, Li B, Song JW, Zhang S, Nie P. IFN-υ and its receptor subunits, IFN-υR1 and IL10RB in mallard Anas platyrhynchos. Poult Sci 2024; 103:103673. [PMID: 38564837 PMCID: PMC10999703 DOI: 10.1016/j.psj.2024.103673] [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: 12/22/2023] [Revised: 03/07/2024] [Accepted: 03/14/2024] [Indexed: 04/04/2024] Open
Abstract
Type IV interferon (IFN) has been shown to be a cytokine with antiviral activity in fish and amphibian. But, it has not been cloned and characterized functionally in avian species. In this study, type IV IFN, IFN-υ, and its 2 possible receptors, IFN-υR1 and IL10RB, were identified from an avian species, the mallard (Anas platyrhynchos). Mallard IFN-υ has a 531 bp open reading frame (ORF), encoding 176 amino acids (aa), and has highly conserved features as reported in different species, with an N-terminal signal peptide and a predicted multi-helix structure. The IFN-υR1 and IL10RB contain 528 and 343 aa, respectively, with IFN-υR1 protein containing JAK1 and STAT binding sites, and IL10RB containing TYK2 binding site. These 2 receptor subunits also possess 3 domains, the N-terminal extracellular domain, the transmembrane domain, and the C-terminal intracellular domain. Expression analysis indicated that IFN-υ, IFN-υR1 and IL10RB were widely expressed in examined organs/tissues, with the highest level observed in pancreas, blood, and kidney, respectively. The expression of IFN-υ, IFN-υR1 and IL10RB in liver, spleen or kidney was significantly upregulated after stimulation with polyI:C. Furthermore, recombinant IFN-υ protein induced the expression of ISGs, and the receptor of IFN-υ was verified as IFN-υR1 and IL10RB using a chimeric receptor approach in HEK293 cells. Taken together, these results indicate that IFN-υ is involved in the host innate immune response in mallard.
Collapse
Affiliation(s)
- An Ning Pang
- State Key Laboratory of Freshwater Ecology and Biotechnology, and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China
| | - Shan Nan Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China
| | - Lan Hao Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - Bo Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - Jing Wei Song
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - Shan Zhang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - P Nie
- State Key Laboratory of Freshwater Ecology and Biotechnology, and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China; School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China.
| |
Collapse
|
5
|
Zhang Q, Kisand K, Feng Y, Rinchai D, Jouanguy E, Cobat A, Casanova JL, Zhang SY. In search of a function for human type III interferons: insights from inherited and acquired deficits. Curr Opin Immunol 2024; 87:102427. [PMID: 38781720 PMCID: PMC11209856 DOI: 10.1016/j.coi.2024.102427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 03/19/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
The essential and redundant functions of human type I and II interferons (IFNs) have been delineated over the last three decades by studies of patients with inborn errors of immunity or their autoimmune phenocopies, but much less is known about type III IFNs. Patients with cells that do not respond to type III IFNs due to inherited IL10RB deficiency display no overt viral disease, and their inflammatory disease phenotypes can be explained by defective signaling via other interleukine10RB-dependent pathways. Moreover, patients with inherited deficiencies of interferon-stimulated gene factor 3 (ISGF-3) (STAT1, STAT2, IRF9) present viral diseases also seen in patients with inherited deficiencies of the type I IFN receptor (IFNAR1/2). Finally, patients with autoantibodies neutralizing type III IFNs have no obvious predisposition to viral disease. Current findings thus suggest that type III IFNs are largely redundant in humans. The essential functions of human type III IFNs, particularly in antiviral defenses, remain to be discovered.
Collapse
Affiliation(s)
- Qian Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France.
| | - Kai Kisand
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Yi Feng
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA
| | - Darawan Rinchai
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA
| | - Emmanuelle Jouanguy
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Aurélie Cobat
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; Department of Pediatrics, Necker Hospital for Sick Children, AP-HP, Paris, France; Howard Hughes Medical Institute, New York, USA
| | - Shen-Ying Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| |
Collapse
|
6
|
Lai J, He X, Zhang R, Zhang L, Chen L, He F, Li L, Yang L, Ren T, Xiang B. Chicken Interferon-Alpha and -Lambda Exhibit Antiviral Effects against Fowl Adenovirus Serotype 4 in Leghorn Male Hepatocellular Cells. Int J Mol Sci 2024; 25:1681. [PMID: 38338959 PMCID: PMC10855402 DOI: 10.3390/ijms25031681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/14/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Hydropericardium hepatitis syndrome (HHS) is primarily caused by fowl adenovirus serotype 4 (FAdV-4), causing high mortality in chickens. Although vaccination strategies against FAdV-4 have been adopted, HHS still occurs sporadically. Furthermore, no effective drugs are available for controlling FAdV-4 infection. However, type I and III interferon (IFN) are crucial therapeutic agents against viral infection. The following experiments were conducted to investigate the inhibitory effect of chicken IFN against FadV-4. We expressed recombinant chicken type I IFN-α (ChIFN-α) and type III IFN-λ (ChIFN-λ) in Escherichia coli and systemically investigated their antiviral activity against FAdV-4 infection in Leghorn male hepatocellular (LMH) cells. ChIFN-α and ChIFN-λ dose dependently inhibited FAdV-4 replication in LMH cells. Compared with ChIFN-λ, ChIFN-α more significantly inhibited viral genome transcription but less significantly suppressed FAdV-4 release. ChIFN-α- and ChIFN-λ-induced IFN-stimulated gene (ISG) expression, such as PKR, ZAP, IRF7, MX1, Viperin, IFIT5, OASL, and IFI6, in LMH cells; however, ChIFN-α induced a stronger expression level than ChIFN-λ. Thus, our data revealed that ChIFN-α and ChIFN-λ might trigger different ISG expression levels, inhibiting FAdV-4 replication via different steps of the FAdV-4 lifecycle, which furthers the potential applications of IFN antiviral drugs in chickens.
Collapse
Affiliation(s)
- Jinyu Lai
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China (L.Z.); (L.Y.)
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Xingchen He
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China (L.Z.); (L.Y.)
- Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Rongjie Zhang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China (L.Z.); (L.Y.)
- Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Limei Zhang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China (L.Z.); (L.Y.)
- Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Libin Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Fengping He
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China (L.Z.); (L.Y.)
- Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Lei Li
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China (L.Z.); (L.Y.)
- Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Liangyu Yang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China (L.Z.); (L.Y.)
- Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| | - Tao Ren
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Bin Xiang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China (L.Z.); (L.Y.)
- Center for Poultry Disease Control and Prevention, Yunnan Agricultural University, Kunming 650201, China
| |
Collapse
|
7
|
Rautenschlein S, Schat KA. The Immunological Basis for Vaccination. Avian Dis 2024; 67:366-379. [PMID: 38300658 DOI: 10.1637/aviandiseases-d-23-99996] [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: 08/01/2023] [Accepted: 08/29/2023] [Indexed: 02/02/2024]
Abstract
Vaccination is crucial for health protection of poultry and therefore important to maintaining high production standards. Proper vaccination requires knowledge of the key players of the well-orchestrated immune system of birds, their interdependence and delicate regulation, and, subsequently, possible modes of stimulation through vaccine antigens and adjuvants. The knowledge about the innate and acquired immune systems of birds has increased significantly during the recent years but open questions remain and have to be elucidated further. Despite similarities between avian and mammalian species in their composition of immune cells and modes of activation, important differences exist, including differences in the innate, but also humoral and cell-mediated immunity with respect to, for example, signaling transduction pathways, antigen presentation, and cell repertoires. For a successful vaccination strategy in birds it always has to be considered that genotype and age of the birds at the time point of immunization as well as their microbiota composition may have an impact and may drive the immune reactions into different directions. Recent achievements in the understanding of the concept of trained immunity will contribute to the advancement of current vaccine types helping to improve protection beyond the specificity of an antigen-driven immune response. The fast developments in new omics technologies will provide insights into protective B- and T-cell epitopes involved in cross-protection, which subsequently will lead to the improvement of vaccine efficacy in poultry.
Collapse
Affiliation(s)
- Silke Rautenschlein
- Clinic for Poultry, University of Veterinary Medicine Hannover, Clinic for Poultry, Hannover, Lower Saxony 30559, Germany,
| | - Karel A Schat
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| |
Collapse
|
8
|
Al-Eitan L, Mihyar A, Zhang L, Bisht P, Jaenisch R. Genomic and biological variation in bat IFNs: An antiviral treatment approach. Rev Med Virol 2024; 34:e2488. [PMID: 37921610 DOI: 10.1002/rmv.2488] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/04/2023]
Abstract
Bat-borne viruses have attracted considerable research, especially in relation to the Covid-19 pandemic. Although bats can carry multiple zoonotic viruses that are lethal to many mammalian species, they appear to be asymptomatic to viral infection despite the high viral loads contained in their bodies. There are several differences between bats and other mammals. One of the major differences between bats and other mammals is the bats' ability to fly, which is believed to have induced evolutionary changes. It may have also favoured them as suitable hosts for viruses. This is related to their tolerance to viral infection. Innate immunity is the first line of defence against viral infection, but bats have metamorphosed the type of responses induced by innate immunity factors such as interferons. The expression patterns of interferons differ, as do those of interferon-related genes such as interferon regulatory factors and interferon-stimulated genes that contribute to the antiviral response of infected cells. In addition, the signalling pathways related to viral infection and immune responses have been subject to evolutionary changes, including mutations compared to their homologues in other mammals and gene selection. This article discusses the differences in the interferon-mediated antiviral response in bats compared to that of other mammals and how these differences are correlated to viral tolerance in bats. The effect of bat interferons related genes on human antiviral response against bat-borne viruses is also discussed.
Collapse
Affiliation(s)
- Laith Al-Eitan
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, Jordan
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Ahmad Mihyar
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, Jordan
| | - Liguo Zhang
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Punam Bisht
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
9
|
Chen J, Li P, Zou W, Jiang Y, Li L, Hao P, Gao Z, Qu Q, Pang Z, Zhuang X, Nan F, Jin N, Du S, Li C. Identification of a Novel Interferon Lambda Splice Variant in Chickens. J Virol 2023; 97:e0174322. [PMID: 36877044 PMCID: PMC10062172 DOI: 10.1128/jvi.01743-22] [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: 11/08/2022] [Accepted: 02/08/2023] [Indexed: 03/07/2023] Open
Abstract
Type III interferons (IFNLs) have critical roles in the host's innate immune system, also serving as the first line against pathogenic infections of mucosal surfaces. In mammals, several IFNLs have been reported; however, only limited data on the repertoire of IFNLs in avian species is available. Previous studies showed only one member in chicken (chIFNL3). Herein, we identified a novel chicken IFNL for the first time, termed chIFNL3a, which contains 354 bp, and encodes 118 amino acids. The predicted protein is 57.1% amino acid identity with chIFNL. Genetic, evolutionary, and sequence analyses indicated that the new open reading frame (ORF) groups with type III chicken IFNs represent a novel splice variant. Compared to IFNs from different species, the new ORF is clustered within the type III IFNs group. Further study showed that chIFNL3a could activate a panel of IFN-regulated genes and function mediated by the IFNL receptor, and chIFNL3a markedly inhibited the replication of Newcastle disease virus (NDV) and influenza virus in vitro. These data collectively shed light on the repertoire of IFNs in avian species and provide useful information that further elucidate the interaction of the chIFNLs and viral infection of poultry. IMPORTANCE Interferons (IFNs) are critical soluble factors in the immune system, and are composed of 3 types (I, II, and III) that utilize different receptor complexes (IFN-αR1/IFN-αR2, IFN-γR1/IFN-γR2, and IFN-λR1/IL-10R2, respectively). Herein, we identified IFNL from the genomic sequences of chicken and termed it chIFNL3a, located on chromosome 7 of chicken. Phylogenetically clustered with all known types of chicken IFNs, the finding of this IFN is considered a type III IFN. To further evaluate the biological properties of chIFNL3a, the target protein was prepared by the baculovirus expression system (BES), which could markedly inhibit the replication of NDV and influenza viruses. In this study, we uncovered a new interferon lambda splice variant of chicken, termed chIFNL3a, which could inhibit viral replication in cells. Importantly, these novel findings may extend to other viruses, offering a new direction for therapeutic interventions.
Collapse
Affiliation(s)
- Jing Chen
- College of Veterinary Medicine, Key Lab for Zoonoses Research, Ministry of Education, Jilin University, Changchun, China
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Peiheng Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Wancheng Zou
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yuhang Jiang
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Letian Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Pengfei Hao
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zihan Gao
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Qiaoqiao Qu
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zhaoxia Pang
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Xinyu Zhuang
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Fulong Nan
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Ningyi Jin
- College of Veterinary Medicine, Key Lab for Zoonoses Research, Ministry of Education, Jilin University, Changchun, China
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Shouwen Du
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Chang Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| |
Collapse
|
10
|
Lu M, Lee Y, Lillehoj HS. Evolution of developmental and comparative immunology in poultry: The regulators and the regulated. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 138:104525. [PMID: 36058383 DOI: 10.1016/j.dci.2022.104525] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/25/2022] [Accepted: 08/28/2022] [Indexed: 06/15/2023]
Abstract
Avian has a unique immune system that evolved in response to environmental pressures in all aspects of innate and adaptive immune responses, including localized and circulating lymphocytes, diversity of immunoglobulin repertoire, and various cytokines and chemokines. All of these attributes make birds an indispensable vertebrate model for studying the fundamental immunological concepts and comparative immunology. However, research on the immune system in birds lags far behind that of humans, mice, and other agricultural animal species, and limited immune tools have hindered the adequate application of birds as disease models for mammalian systems. An in-depth understanding of the avian immune system relies on the detailed studies of various regulated and regulatory mediators, such as cell surface antigens, cytokines, and chemokines. Here, we review current knowledge centered on the roles of avian cell surface antigens, cytokines, chemokines, and beyond. Moreover, we provide an update on recent progress in this rapidly developing field of study with respect to the availability of immune reagents that will facilitate the study of regulatory and regulated components of poultry immunity. The new information on avian immunity and available immune tools will benefit avian researchers and evolutionary biologists in conducting fundamental and applied research.
Collapse
Affiliation(s)
- Mingmin Lu
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, U.S. Department of Agriculture-Agricultural Research Service, Beltsville, MD, 20705, USA.
| | - Youngsub Lee
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, U.S. Department of Agriculture-Agricultural Research Service, Beltsville, MD, 20705, USA.
| | - Hyun S Lillehoj
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, U.S. Department of Agriculture-Agricultural Research Service, Beltsville, MD, 20705, USA.
| |
Collapse
|
11
|
Song H, Liu X, Gao X, Li J, Shang Y, Gao W, Li Y, Zhang Z. Transcriptome analysis of pre-immune state induced by interferon gamma inhibiting the replication of H 9N 2 avian influenza viruses in chicken embryo fibroblasts. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2022; 103:105332. [PMID: 35811034 DOI: 10.1016/j.meegid.2022.105332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 06/24/2022] [Accepted: 07/04/2022] [Indexed: 06/15/2023]
Abstract
Interferon (IFN), a critical antiviral cytokine produced by pathogens-induced cells, plays an important role in host innate immune system. In this study, to investigate the inhibition effect of IFN on avian influenza virus (AIV), Chicken Embryo Fibroblasts (CEFs) was infected by H9N2 AIV. The pre-immune state and transcriptome analysis have been observed and performed. The result showed chicken interferon gamma (chIFN-γ) have the most inhibitory effect on H9N2 virus among three types of chicken interferons (chIFNs). Inhibition of chIFN-γ on H9N2 virus was verified by indirect immunofluorescence, RT-qPCR and western blot. The possible signaling pathways induced by chIFN-γ with or without virus were analyzed by transcriptome. The transcriptome data were compared among H9N2-infected, chIFN-γ-treated, chIFN-γ + H9N2-treated, and Control groups. In summary, RNA-sequencing (RNA-seq) data suggested that H9N2 virus infection resulted in corresponding response of certain defensive, inflammatory and metabolism pathways to the virus replication in CEFs. Furthermore, while CEFs were treated with chIFN-γ, many immune-related signaling pathways in cells are affected and altered. Antiviral genes involved in these immune pathways such as interferon regulatory factors, chemokines, interferon-stimulated genes (ISGs) and transcription factors were significantly up-regulated, and showed significant antiviral responses. Compared with virus infected CEFs alone, pretreatment with IFN induced the expression of antiviral genes and activated related antiviral pathways, inhibited the viral replication as result. Our study provided functional annotations for antiviral genes and the basis for studying the mechanism of chIFN-γ mediated response against H9N2 AIV.
Collapse
Affiliation(s)
- Haozhi Song
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xingjian Liu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xintao Gao
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Jialei Li
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yuting Shang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Weisong Gao
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yinü Li
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Zhifang Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
12
|
Chen SN, Gan Z, Hou J, Yang YC, Huang L, Huang B, Wang S, Nie P. Identification and establishment of type IV interferon and the characterization of interferon-υ including its class II cytokine receptors IFN-υR1 and IL-10R2. Nat Commun 2022; 13:999. [PMID: 35194032 PMCID: PMC8863823 DOI: 10.1038/s41467-022-28645-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 01/26/2022] [Indexed: 11/10/2022] Open
Abstract
Interferons (IFNs) are critical soluble factors in the immune system and are composed of three types, (I, II and III) that utilize different receptor complexes IFN-αR1/IFN-αR2, IFN-γR1/IFN-γR2, and IFN-λR1/IL-10R2, respectively. Here we identify IFN-υ from the genomic sequences of vertebrates. The members of class II cytokine receptors, IFN-υR1 and IL-10R2, are identified as the receptor complex of IFN-υ, and are associated with IFN-υ stimulated gene expression and antiviral activity in zebrafish (Danio rerio) and African clawed frog (Xenopus laevis). IFN-υ and IFN-υR1 are separately located at unique and highly conserved loci, being distinct from all other three-type IFNs. IFN-υ and IFN-υR1 are phylogenetically clustered with class II cytokines and class II cytokine receptors, respectively. Therefore, the finding of this IFN ligand-receptor system may be considered as a type IV IFN, in addition to the currently recognized three types of IFNs in vertebrates. Interferons are critical soluble components of the inflammatory process and are composed of three types with associated receptor complexes. Here the authors identify and characterise the type IV interferon, IFN-υ, and identify its associated receptors, denote functionality during in vivo infection and ascertain its genomic localisation.
Collapse
Affiliation(s)
- Shan Nan Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, 430072, China.,Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, 430072, China.,Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Zhen Gan
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, 430072, China
| | - Jing Hou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, 430072, China
| | - Yue Cong Yang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, 430072, China
| | - Lin Huang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, 430072, China
| | - Bei Huang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, 430072, China.,College of Fisheries, Jimei University, 43 Yindou Road, Xiamen, Fujian, 361021, China
| | - Su Wang
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong, 266237, China.,School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong, 266109, China
| | - Pin Nie
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, 430072, China. .,Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, 430072, China. .,Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China. .,Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong, 266237, China. .,School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong, 266109, China.
| |
Collapse
|
13
|
Yin Y, Favoreel HW. Herpesviruses and the Type III Interferon System. Virol Sin 2021; 36:577-587. [PMID: 33400088 DOI: 10.1007/s12250-020-00330-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 10/27/2020] [Indexed: 12/28/2022] Open
Abstract
Type III interferons (IFNs) represent the most recently discovered group of IFNs. Together with type I IFNs (e.g. IFN-α/β), type III IFNs (IFN-λ) are produced as part of the innate immune response to virus infection, and elicit an anti-viral state by inducing expression of interferon stimulated genes (ISGs). It was initially thought that type I IFNs and type III IFNs perform largely redundant functions. However, it has become evident that type III IFNs particularly play a major role in antiviral protection of mucosal epithelial barriers, thereby serving an important role in the first-line defense against virus infection and invasion at contact areas with the outside world, versus the generally more broad, potent and systemic antiviral effects of type I IFNs. Herpesviruseses are large DNA viruses, which enter their host via mucosal surfaces and establish lifelong, latent infections. Despite the importance of mucosal epithelial cells in the pathogenesis of herpesviruses, our current knowledge on the interaction of herpesviruses with type III IFN is limited and largely restricted to studies on the alphaherpesvirus herpes simplex virus (HSV). This review summarizes the current understanding about the role of IFN-λ in the immune response against herpesvirus infections.
Collapse
Affiliation(s)
- Yue Yin
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Herman W Favoreel
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, 9820, Merelbeke, Belgium.
| |
Collapse
|
14
|
Geoffroy K, Bourgeois-Daigneault MC. The pros and cons of interferons for oncolytic virotherapy. Cytokine Growth Factor Rev 2020; 56:49-58. [PMID: 32694051 DOI: 10.1016/j.cytogfr.2020.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/02/2020] [Indexed: 12/29/2022]
Abstract
Interferons (IFN) are potent immune stimulators that play key roles in both innate and adaptive immune responses. They are considered the first line of defense against viral pathogens and can even be used as treatments to boost the immune system. While viruses are usually seen as a threat to the host, an emerging class of cancer therapeutics exploits the natural capacity of some viruses to directly infect and kill cancer cells. The cancer-specificity of these bio-therapeutics, called oncolytic viruses (OVs), often relies on defective IFN responses that are frequently observed in cancer cells, therefore increasing their vulnerability to viruses compared to healthy cells. To ensure the safety of the therapy, many OVs have been engineered to further activate the IFN response. As a consequence of this IFN over-stimulation, the virus is cleared faster by the immune system, which limits direct oncolysis. Importantly, the therapeutic activity of OVs also relies on their capacity to trigger anti-tumor immunity and IFNs are key players in this aspect. Here, we review the complex cancer-virus-anti-tumor immunity interplay and discuss the diverse functions of IFNs for each of these processes.
Collapse
Affiliation(s)
- Karen Geoffroy
- Cancer axis and Institut du cancer de Montréal, Centre de recherche du CHUM- CRCHUM, 900 St-Denis Street, Viger Tower, Room R10.480, Montreal, Quebec, H2X0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Faculty of Medicine, Université de Montréal, 2900 Edouard-Montpetit Boulevard, Roger-Gaudry Building, Montreal, Quebec, H3T1J4, Canada
| | - Marie-Claude Bourgeois-Daigneault
- Cancer axis and Institut du cancer de Montréal, Centre de recherche du CHUM- CRCHUM, 900 St-Denis Street, Viger Tower, Room R10.480, Montreal, Quebec, H2X0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Faculty of Medicine, Université de Montréal, 2900 Edouard-Montpetit Boulevard, Roger-Gaudry Building, Montreal, Quebec, H3T1J4, Canada.
| |
Collapse
|
15
|
Campbell LK, Magor KE. Pattern Recognition Receptor Signaling and Innate Responses to Influenza A Viruses in the Mallard Duck, Compared to Humans and Chickens. Front Cell Infect Microbiol 2020; 10:209. [PMID: 32477965 PMCID: PMC7236763 DOI: 10.3389/fcimb.2020.00209] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/16/2020] [Indexed: 12/25/2022] Open
Abstract
Mallard ducks are a natural host and reservoir of avian Influenza A viruses. While most influenza strains can replicate in mallards, the virus typically does not cause substantial disease in this host. Mallards are often resistant to disease caused by highly pathogenic avian influenza viruses, while the same strains can cause severe infection in humans, chickens, and even other species of ducks, resulting in systemic spread of the virus and even death. The differences in influenza detection and antiviral effectors responsible for limiting damage in the mallards are largely unknown. Domestic mallards have an early and robust innate response to infection that seems to limit replication and clear highly pathogenic strains. The regulation and timing of the response to influenza also seems to circumvent damage done by a prolonged or dysregulated immune response. Rapid initiation of innate immune responses depends on viral recognition by pattern recognition receptors (PRRs) expressed in tissues where the virus replicates. RIG-like receptors (RLRs), Toll-like receptors (TLRs), and Nod-like receptors (NLRs) are all important influenza sensors in mammals during infection. Ducks utilize many of the same PRRs to detect influenza, namely RIG-I, TLR7, and TLR3 and their downstream adaptors. Ducks also express many of the same signal transduction proteins including TBK1, TRIF, and TRAF3. Some antiviral effectors expressed downstream of these signaling pathways inhibit influenza replication in ducks. In this review, we summarize the recent advances in our understanding of influenza recognition and response through duck PRRs and their adaptors. We compare basal tissue expression and regulation of these signaling components in birds, to better understand what contributes to influenza resistance in the duck.
Collapse
Affiliation(s)
- Lee K Campbell
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.,Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Katharine E Magor
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.,Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
16
|
Barjesteh N, O'Dowd K, Vahedi SM. Antiviral responses against chicken respiratory infections: Focus on avian influenza virus and infectious bronchitis virus. Cytokine 2020; 127:154961. [PMID: 31901597 PMCID: PMC7129915 DOI: 10.1016/j.cyto.2019.154961] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/13/2022]
Abstract
Some of the respiratory viral infections in chickens pose a significant threat to the poultry industry and public health. In response to viral infections, host innate responses provide the first line of defense against viruses, which often act even before the establishment of the infection. Host cells sense the presence of viral components through germinal encoded pattern recognition receptors (PRRs). The engagement of PRRs with pathogen-associated molecular patterns leads to the induction of pro-inflammatory and interferon productions. Induced antiviral responses play a critical role in the outcome of the infections. In order to improve current strategies for control of viral infections or to advance new strategies aimed against viral infections, a deep understanding of host-virus interaction and induction of antiviral responses is required. In this review, we summarized recent progress in understanding innate antiviral responses in chickens with a focus on the avian influenza virus and infectious bronchitis virus.
Collapse
Affiliation(s)
- Neda Barjesteh
- Research Group on Infectious Diseases in Production Animals (GREMIP), and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada.
| | - Kelsey O'Dowd
- Research Group on Infectious Diseases in Production Animals (GREMIP), and Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Seyed Milad Vahedi
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
17
|
Bertzbach LD, Harlin O, Härtle S, Fehler F, Vychodil T, Kaufer BB, Kaspers B. IFNα and IFNγ Impede Marek's Disease Progression. Viruses 2019; 11:v11121103. [PMID: 31795203 PMCID: PMC6950089 DOI: 10.3390/v11121103] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 12/18/2022] Open
Abstract
Marek’s disease virus (MDV) is an alphaherpesvirus that causes Marek’s disease, a malignant lymphoproliferative disease of domestic chickens. While MDV vaccines protect animals from clinical disease, they do not provide sterilizing immunity and allow field strains to circulate and evolve in vaccinated flocks. Therefore, there is a need for improved vaccines and for a better understanding of innate and adaptive immune responses against MDV infections. Interferons (IFNs) play important roles in the innate immune defenses against viruses and induce upregulation of a cellular antiviral state. In this report, we quantified the potent antiviral effect of IFNα and IFNγ against MDV infections in vitro. Moreover, we demonstrate that both cytokines can delay Marek’s disease onset and progression in vivo. Additionally, blocking of endogenous IFNα using a specific monoclonal antibody, in turn, accelerated disease. In summary, our data reveal the effects of IFNα and IFNγ on MDV infection and improve our understanding of innate immune responses against this oncogenic virus.
Collapse
Affiliation(s)
- Luca D. Bertzbach
- Institute of Virology, Freie Universität Berlin, 14163 Berlin, Germany; (L.D.B.); (T.V.)
| | - Olof Harlin
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, 80539 Munich, Germany; (O.H.); (S.H.)
| | - Sonja Härtle
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, 80539 Munich, Germany; (O.H.); (S.H.)
| | | | - Tereza Vychodil
- Institute of Virology, Freie Universität Berlin, 14163 Berlin, Germany; (L.D.B.); (T.V.)
| | - Benedikt B. Kaufer
- Institute of Virology, Freie Universität Berlin, 14163 Berlin, Germany; (L.D.B.); (T.V.)
- Correspondence: (B.B.K.); (B.K.)
| | - Bernd Kaspers
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, 80539 Munich, Germany; (O.H.); (S.H.)
- Correspondence: (B.B.K.); (B.K.)
| |
Collapse
|
18
|
|
19
|
Watson SF, Knol LI, Witteveldt J, Macias S. Crosstalk Between Mammalian Antiviral Pathways. Noncoding RNA 2019; 5:E29. [PMID: 30909383 PMCID: PMC6468734 DOI: 10.3390/ncrna5010029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/19/2019] [Accepted: 03/20/2019] [Indexed: 12/15/2022] Open
Abstract
As part of their innate immune response against viral infections, mammals activate the expression of type I interferons to prevent viral replication and dissemination. An antiviral RNAi-based response can be also activated in mammals, suggesting that several mechanisms can co-occur in the same cell and that these pathways must interact to enable the best antiviral response. Here, we will review how the classical type I interferon response and the recently described antiviral RNAi pathways interact in mammalian cells. Specifically, we will uncover how the small RNA biogenesis pathway, composed by the nucleases Drosha and Dicer can act as direct antiviral factors, and how the type-I interferon response regulates the function of these. We will also describe how the factors involved in small RNA biogenesis and specific small RNAs impact the activation of the type I interferon response and antiviral activity. With this, we aim to expose the complex and intricate network of interactions between the different antiviral pathways in mammals.
Collapse
Affiliation(s)
- Samir F Watson
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK.
| | - Lisanne I Knol
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK.
| | - Jeroen Witteveldt
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK.
| | - Sara Macias
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK.
| |
Collapse
|
20
|
Arslan M, Yang X, Santhakumar D, Liu X, Hu X, Munir M, Li Y, Zhang Z. Dynamic Expression of Interferon Lambda Regulated Genes in Primary Fibroblasts and Immune Organs of the Chicken. Genes (Basel) 2019; 10:genes10020145. [PMID: 30769908 PMCID: PMC6409627 DOI: 10.3390/genes10020145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/09/2019] [Accepted: 02/11/2019] [Indexed: 01/17/2023] Open
Abstract
Interferons (IFNs) are pleiotropic cytokines that establish a first line of defense against viral infections in vertebrates. Several types of IFN have been identified; however, limited information is available in poultry, especially using live animal experimental models. IFN-lambda (IFN-λ) has recently been shown to exert a significant antiviral impact against viral pathogens in mammals. In order to investigate the in vivo potential of chicken IFN-λ (chIFN-λ) as a regulator of innate immunity, and potential antiviral therapeutics, we profiled the transcriptome of chIFN-λ-stimulated chicken immune organs (in vivo) and compared it with primary chicken embryo fibroblasts (in vitro). Employing the baculovirus expression vector system (BEVS), recombinant chIFN-λ3 (rchIFN-λ3) was produced and its biological activities were demonstrated. The rchIFNλ3 induced a great array of IFN-regulated genes in primary chicken fibroblast cells. The transcriptional profiling using RNA-seq and subsequent bioinformatics analysis (gene ontology, differential expressed genes, and KEGGs analysis) of the bursa of Fabricious and the thymus demonstrated an upregulation of crucial immune genes (viperin, IKKB, CCL5, IL1β, and AP1) as well as the antiviral signaling pathways. Interestingly, this experimental approach revealed contrasting evidence of the antiviral potential of chIFN-λ in both in vivo and in vitro models. Taken together, our data signifies the potential of chIFN-λ as a potent antiviral cytokine and highlights its future possible use as an antiviral therapeutic in poultry.
Collapse
Affiliation(s)
- Mehboob Arslan
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Xin Yang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Diwakar Santhakumar
- Division of Biomedical and Life sciences, Faculty of Health and Medicine, Lancaster University,LA1 4YG, Lancaster, UK.
| | - Xingjian Liu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Xiaoyuan Hu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Muhammad Munir
- Division of Biomedical and Life sciences, Faculty of Health and Medicine, Lancaster University,LA1 4YG, Lancaster, UK.
| | - Yinü Li
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Zhifang Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
21
|
Sid H, Schusser B. Applications of Gene Editing in Chickens: A New Era Is on the Horizon. Front Genet 2018; 9:456. [PMID: 30356667 PMCID: PMC6189320 DOI: 10.3389/fgene.2018.00456] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 09/18/2018] [Indexed: 01/15/2023] Open
Abstract
The chicken represents a valuable model for research in the area of immunology, infectious diseases as well as developmental biology. Although it was the first livestock species to have its genome sequenced, there was no reverse genetic technology available to help understanding specific gene functions. Recently, homologous recombination was used to knockout the chicken immunoglobulin genes. Subsequent studies using immunoglobulin knockout birds helped to understand different aspects related to B cell development and antibody production. Furthermore, the latest advances in the field of genome editing including the CRISPR/Cas9 system allowed the introduction of site specific gene modifications in various animal species. Thus, it may provide a powerful tool for the generation of genetically modified chickens carrying resistance for certain pathogens. This was previously demonstrated by targeting the Trp38 region which was shown to be effective in the control of avian leukosis virus in chicken DF-1 cells. Herein we review the current and future prospects of gene editing and how it possibly contributes to the development of resistant chickens against infectious diseases.
Collapse
Affiliation(s)
| | - Benjamin Schusser
- Department of Animal Sciences, Reproductive Biotechnology, School of Life Sciences Weihenstephan, Technical University Munich, Freising, Germany
| |
Collapse
|
22
|
Yang X, Li J, Liu H, Zhang P, Chen D, Men S, Li X, Wang H. Induction of innate immune response following introduction of infectious bronchitis virus (IBV) in the trachea and renal tissues of chickens. Microb Pathog 2018; 116:54-61. [PMID: 29330060 DOI: 10.1016/j.micpath.2018.01.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 01/02/2018] [Accepted: 01/07/2018] [Indexed: 12/31/2022]
Abstract
Infectious bronchitis (IB) is a highly contagious respiratory disease of chickens, which is caused by the infectious bronchitis virus (IBV). The innate immune response is crucial for antiviral infections and revealing the pathogenic mechanisms of IBV. In this study, we presents an evaluation of interferon (I, II and III IFNs) in renal and tracheal samples from chickens experimentally infected previously vaccinated or not. The results suggest differential expression of chicken interferon, among them type I IFN elaborate a major role in fighting off virus. And vaccine confers greater induction ability of innate immunity thereby vaccination prior infection occurs might be necessary. Above all, we found that IFN-λ also have an effect on IBV infection in trachea besides many other respiratory virus. This study provides the first comprehensive analysis of host-virus interactions of IBV with chicken innate immune response mediated by interferon in various groups.
Collapse
Affiliation(s)
- Xin Yang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, PR China
| | - Jianan Li
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, PR China
| | - Hui Liu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, PR China
| | - Peng Zhang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, PR China
| | - Danyu Chen
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, PR China
| | - Shuai Men
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, PR China
| | - Xiaocheng Li
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, PR China
| | - Hongning Wang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, PR China.
| |
Collapse
|
23
|
Chen S, Zhang W, Zhou Q, Wang A, Sun L, Wang M, Jia R, Zhu D, Liu M, Sun K, Yang Q, Wu Y, Chen X, Cheng A. Cross-species antiviral activity of goose interferon lambda against duck plague virus is related to its positive self-regulatory feedback loop. J Gen Virol 2017; 98:1455-1466. [PMID: 28678686 DOI: 10.1099/jgv.0.000788] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Duck plague virus (DPV) is a virus of the Herpesviridae family that leads to acute disease with a high mortality rate in ducks. Control of the disease contributes to the development of poultry breeding. Type III IFN family (IFN-λs) is a novel member of the IFN family, and goose IFN-λ (goIFN-λ) is a newly identified gene whose antiviral function has only been investigated to a limited extent. Here, the cross-species antiviral activity of goIFN-λ against DPV in duck embryo fibroblasts (DEFs) was studied. We found that pre-treatment with goIFN-λ greatly increased the expression of IFN-λ in both heterologous DEFs and homologous goose embryo fibroblasts (GEFs), while differentially inducing IFNα- and IFN-stimulated genes. Additionally, a positive self-regulatory feedback loop of goIFN-λ was blocked by a mouse anti-goIFN-λ polyclonal antibody, which was confirmed in both homologous GEFs and goose peripheral blood mononuclear cells (PBMCs). The suppression of the BAC-DPV-EGFP by goIFN-λ in DEFs was confirmed by fluorescence microscopy, flow cytometry (FCM) analysis, viral copies and titre detection, which can be rescued by mouse anti-goIFN-λ polyclonal antibody incubation. Finally, reporter gene assays indicated that the cross-species antiviral activity of goIFN-λ against BAC-DPV-EGFP is related to its positive self-regulatory feedback loop and subsequent ISG induction. Our data shed light on the fundamental mechanisms of goIFN-λ antiviral function in vitro and extend the considerable range of therapeutic applications in multiple-poultry disease.
Collapse
Affiliation(s)
- Shun Chen
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, PR China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China.,Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Wei Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Qin Zhou
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Anqi Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Lipei Sun
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Mingshu Wang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, PR China.,Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Renyong Jia
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, PR China.,Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Dekang Zhu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, PR China.,Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Mafeng Liu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, PR China.,Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Kunfeng Sun
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, PR China.,Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Qiao Yang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, PR China.,Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Ying Wu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, PR China.,Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Xiaoyue Chen
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, PR China.,Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Anchun Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China.,Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, PR China
| |
Collapse
|
24
|
Zhou Q, Zhang W, Chen S, Wang A, Sun L, Wang M, Jia R, Zhu D, Liu M, Sun K, Yang Q, Wu Y, Chen X, Cheng A. Identification of Type III Interferon (IFN-λ) in Chinese Goose: Gene Structure, Age-Dependent Expression Profile, and Antiviral Immune Characteristics In Vivo and In Vitro. J Interferon Cytokine Res 2017; 37:269-277. [PMID: 28388308 DOI: 10.1089/jir.2016.0061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Type III interferons (IFN-λ1/λ2/λ3, also known as IL-29/28A/28B, and IFN-λ4) are a recently discovered interferon group. In this study, we first identified the Chinese goose IFN-λ (goIFN-λ). The full-length sequence of goIFN-λ was found to be 823 bp. There was only one open reading frame that contained 570 bp, and, encoded 189 amino acids. The predicted goIFN-λ protein showed 78%, 67%, and 40% amino acid identity with duIFN-λ, chIFN-λ, and hIFN-λ3, respectively. The tissue distribution of goIFN-λ existed as a parallel distribution with goIFNLR1 as its functional receptor, which was mainly expressed in epithelium-rich tissues, such as lung, gizzard, proventriculus, skin and pancreas, and immune tissues, such as harderian gland and thymus. Furthermore, the immunological characteristics studies of goIFN-λ showed that there was a significant increase in the mRNA at the transcriptional level of goIFN-λ after the peripheral blood mononuclear cells were stimulated with ploy (I:C) and ODN2006, and infected with Gosling plague virus (GPV). In vivo, the mRNA transcriptional level of goIFN-λ increased nearly 20 times in the lung tissue and nearly 40 times in the pancreatic tissue after being artificially infected with H9N2 AIV. It is suggested that goIFN-λ might play a pivotal role in the mucosal immune protection and antiviral defense.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Chickens
- Ducks
- Geese/genetics
- Geese/immunology
- Geese/virology
- Gene Expression Regulation, Developmental/immunology
- Humans
- Immunity, Innate
- Immunity, Mucosal
- Influenza A Virus, H9N2 Subtype/immunology
- Interferons/genetics
- Interferons/immunology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/virology
- Mice
- Oligodeoxyribonucleotides/pharmacology
- Open Reading Frames
- Orthomyxoviridae/immunology
- RNA, Messenger/genetics
- RNA, Messenger/immunology
- Sequence Homology, Amino Acid
Collapse
Affiliation(s)
- Qin Zhou
- 1 Institute of Preventive Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
| | - Wei Zhang
- 1 Institute of Preventive Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
| | - Shun Chen
- 1 Institute of Preventive Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
- 2 Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
- 3 Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University , Chengdu, China
| | - Anqi Wang
- 1 Institute of Preventive Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
| | - Lipei Sun
- 1 Institute of Preventive Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
| | - Mingshu Wang
- 1 Institute of Preventive Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
- 2 Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
- 3 Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University , Chengdu, China
| | - Renyong Jia
- 1 Institute of Preventive Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
- 2 Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
- 3 Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University , Chengdu, China
| | - Dekang Zhu
- 2 Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
- 3 Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University , Chengdu, China
| | - Mafeng Liu
- 1 Institute of Preventive Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
| | - Kunfeng Sun
- 1 Institute of Preventive Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
- 2 Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
- 3 Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University , Chengdu, China
| | - Qiao Yang
- 1 Institute of Preventive Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
- 2 Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
- 3 Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University , Chengdu, China
| | - Ying Wu
- 1 Institute of Preventive Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
- 2 Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
- 3 Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University , Chengdu, China
| | - Xiaoyue Chen
- 2 Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
- 3 Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University , Chengdu, China
| | - Anchun Cheng
- 1 Institute of Preventive Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
- 2 Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University , Chengdu, China
- 3 Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University , Chengdu, China
| |
Collapse
|
25
|
Secombes CJ, Zou J. Evolution of Interferons and Interferon Receptors. Front Immunol 2017; 8:209. [PMID: 28303139 PMCID: PMC5332411 DOI: 10.3389/fimmu.2017.00209] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 02/15/2017] [Indexed: 11/25/2022] Open
Abstract
The earliest jawed vertebrates (Gnathostomes) would likely have had interferon (IFN) genes, since they are present in extant cartilaginous fish (sharks and rays) and bony fish (lobe-finned and ray-finned fish, the latter consisting of the chondrostei, holostei, and teleostei), as well as in tetrapods. They are thought to have evolved from a class II helical cytokine ancestor, along with the interleukin (IL)-10 cytokine family. The two rounds of whole genome duplication (WGD) that occurred between invertebrates and vertebrates (1) may have given rise to additional loci, initially containing an IL-10 ancestor and IFN ancestor, which have duplicated further to give rise to the two loci containing the IL-10 family genes, and potentially the IFN type I and IFN type III loci (2). The timing of the divergence of the IFN type II gene from the IL-10 family genes is not clear but was also an early event in vertebrate evolution. Further WGD events at the base of the teleost fish, and in particular teleost lineages (cyprinids, salmonids), have duplicated the loci further, giving rise to additional IFN genes, with tandem gene duplication within a locus a common occurrence. Finally, retrotransposition events have occurred in different vertebrate lineages giving rise to further IFN loci, with large expansions of genes at these loci in some cases. This review will initially explore the likely IFN system present in the earliest Gnathostomes by comparison of the known cartilaginous fish genes with those present in mammals and will then explore the changes that have occurred in gene number/diversification, gene organization, and the encoded proteins during vertebrate evolution.
Collapse
Affiliation(s)
- Chris J Secombes
- Scottish Fish Immunology Research Centre, University of Aberdeen , Aberdeen , UK
| | - Jun Zou
- Scottish Fish Immunology Research Centre, University of Aberdeen , Aberdeen , UK
| |
Collapse
|
26
|
Santhakumar D, Rubbenstroth D, Martinez-Sobrido L, Munir M. Avian Interferons and Their Antiviral Effectors. Front Immunol 2017; 8:49. [PMID: 28197148 PMCID: PMC5281639 DOI: 10.3389/fimmu.2017.00049] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/12/2017] [Indexed: 12/12/2022] Open
Abstract
Interferon (IFN) responses, mediated by a myriad of IFN-stimulated genes (ISGs), are the most profound innate immune responses against viruses. Cumulatively, these IFN effectors establish a multilayered antiviral state to safeguard the host against invading viral pathogens. Considerable genetic and functional characterizations of mammalian IFNs and their effectors have been made, and our understanding on the avian IFNs has started to expand. Similar to mammalian counterparts, three types of IFNs have been genetically characterized in most avian species with available annotated genomes. Intriguingly, chickens are capable of mounting potent innate immune responses upon various stimuli in the absence of essential components of IFN pathways including retinoic acid-inducible gene I, IFN regulatory factor 3 (IRF3), and possibility IRF9. Understanding these unique properties of the chicken IFN system would propose valuable targets for the development of potential therapeutics for a broader range of viruses of both veterinary and zoonotic importance. This review outlines recent developments in the roles of avian IFNs and ISGs against viruses and highlights important areas of research toward our understanding of the antiviral functions of IFN effectors against viral infections in birds.
Collapse
Affiliation(s)
| | - Dennis Rubbenstroth
- Institute for Virology, Faculty of Medicine, University Medical Center, University of Freiburg , Freiburg , Germany
| | - Luis Martinez-Sobrido
- Department of Microbiology and Immunology, University of Rochester Medical Center , Rochester, NY , USA
| | | |
Collapse
|
27
|
Shen H, Zhang C, Guo P, Liu Z, Sun M, Sun J, Li L, Dong J, Zhang J. Short communication: antiviral activity of porcine IFN-λ3 against porcine epidemic diarrhea virus in vitro. Virus Genes 2016; 52:877-882. [PMID: 27470155 PMCID: PMC7089062 DOI: 10.1007/s11262-016-1374-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 07/15/2016] [Indexed: 11/24/2022]
Abstract
A new family of IFNs called type III IFN or IFN-λ has been described, and shown to induce antiviral activity against several viruses in the cell culture. In this study, the molecular cloning, expression, and antiporcine epidemic diarrhea virus (PEDV) activity of porcine IFN-λ3 (poIFN-λ3) were reported. The full-length poIFN-λ3 cDNA sequence encoded 196 amino acids with a 23 amino acid signal peptide. Sequence alignments showed that poIFN-λ3 had an amino acid sequence similarity to Ovis aries (78.1 %), Bos taurus (76.0 %), Tupaia belangeri (71.3 %), Equus caballus (69.9 %), and Homo sapiens (69.9 %). The phylogenetic analysis based on the genomic sequences indicated that poIFN-λ3 is located in the same branch as B. taurus and O. aries IFN-λ3. The poIFN-λ3 without a signal anchor sequence was efficiently expressed in Escherichia coli, and the purified recombinant poIFN-λ3 exhibited significant antiviral effects against PEDV in a dose- and time-dependent manner. This inhibitory effect of poIFN-λ3 on PEDV was observed under three different treatment conditions. The highest inhibition of PEDV was observed in Vero E6 cell cultures pretreated with poIFN-λ3 (prior to PEDV infection). In addition, poIFN-λ3 was able to induce the expression of IFN-stimulated genes, including ISG15, OAS1, and Mx1 in Vero E6 cells. These data demonstrate that poIFN-λ3 has antiviral activity against PEDV and may serve as a useful biotherapeutic candidate to inhibit PEDV or other viruses in swine.
Collapse
Affiliation(s)
- Haiyan Shen
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Open Laboratory of Veterinary Public Health, Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, 510640, Guangdong, China
| | - Chunhong Zhang
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Open Laboratory of Veterinary Public Health, Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, 510640, Guangdong, China
| | - Pengju Guo
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, 510640, Guangdong, China
| | - Zhicheng Liu
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Open Laboratory of Veterinary Public Health, Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, 510640, Guangdong, China
| | - Minhua Sun
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Open Laboratory of Veterinary Public Health, Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, 510640, Guangdong, China
| | - Junying Sun
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Open Laboratory of Veterinary Public Health, Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, 510640, Guangdong, China
| | - Linlin Li
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Open Laboratory of Veterinary Public Health, Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, 510640, Guangdong, China
| | - Jiawen Dong
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Open Laboratory of Veterinary Public Health, Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, 510640, Guangdong, China
| | - Jianfeng Zhang
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Open Laboratory of Veterinary Public Health, Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, 510640, Guangdong, China.
| |
Collapse
|
28
|
Diaz-San Segundo F, Montiel NA, Sturza DF, Perez-Martin E, Hickman D, Ramirez-Medina E, Grubman MJ, de Los Santos T. Combination of Adt-O1Manisa and Ad5-boIFNλ3 induces early protective immunity against foot-and-mouth disease in cattle. Virology 2016; 499:340-349. [PMID: 27743960 DOI: 10.1016/j.virol.2016.09.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/22/2016] [Accepted: 09/27/2016] [Indexed: 11/15/2022]
Abstract
Foot-and-mouth-disease (FMD) remains the most infectious livestock disease worldwide. Although commercially available inactivated or adenovirus-vectored-vaccines (Ad5-FMD) are effective, they require 5-7 days to induce protection. Therefore, new control strategies that stimulate rapid immune responses are needed. Expression of bovine interferon λ3 using the Ad5-vector platform (Ad5-boIFNλ3) is able to delay disease in cattle, but clinical signs appear at 9 days after challenge. We hypothesized that combination of Ad5-boIFNλ3 and Ad5-FMD could induce immediate and lasting protection against FMD. Cattle were vaccinated with an Ad5-FMD, Ad5-boIFNλ3, or the combination of both, followed by challenge at three days post-immunization. All animals treated with Ad5-FMD combined with Ad5-boIFNλ3 were fully protected against FMD, despite the absence of systemic neutralizing antibodies or antiviral activity at the time of challenge. Induction of a strong cell-mediated immune response suggested that Ad5-boIFNλ3 is able to act as an adjuvant of Ad5-FMD vaccine in cattle.
Collapse
Affiliation(s)
- Fayna Diaz-San Segundo
- Plum Island Animal Disease Center, North Atlantic Area, Agricultural Research Service, U.S. Department of Agriculture, Greenport, NY 11944, USA; Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06269, USA
| | - Nestor A Montiel
- Plum Island Animal Disease Center, North Atlantic Area, Agricultural Research Service, U.S. Department of Agriculture, Greenport, NY 11944, USA; Oak Ridge Institute for Science and Education, Plum Island Animal Disease Center Research Participation Program, Oak Ridge, TN 37831, USA
| | - Diego F Sturza
- Plum Island Animal Disease Center, North Atlantic Area, Agricultural Research Service, U.S. Department of Agriculture, Greenport, NY 11944, USA; Oak Ridge Institute for Science and Education, Plum Island Animal Disease Center Research Participation Program, Oak Ridge, TN 37831, USA
| | - Eva Perez-Martin
- Plum Island Animal Disease Center, North Atlantic Area, Agricultural Research Service, U.S. Department of Agriculture, Greenport, NY 11944, USA; Oak Ridge Institute for Science and Education, Plum Island Animal Disease Center Research Participation Program, Oak Ridge, TN 37831, USA
| | - Danielle Hickman
- Plum Island Animal Disease Center, North Atlantic Area, Agricultural Research Service, U.S. Department of Agriculture, Greenport, NY 11944, USA
| | - Elizabeth Ramirez-Medina
- Plum Island Animal Disease Center, North Atlantic Area, Agricultural Research Service, U.S. Department of Agriculture, Greenport, NY 11944, USA; Oak Ridge Institute for Science and Education, Plum Island Animal Disease Center Research Participation Program, Oak Ridge, TN 37831, USA
| | - Marvin J Grubman
- Plum Island Animal Disease Center, North Atlantic Area, Agricultural Research Service, U.S. Department of Agriculture, Greenport, NY 11944, USA
| | - Teresa de Los Santos
- Plum Island Animal Disease Center, North Atlantic Area, Agricultural Research Service, U.S. Department of Agriculture, Greenport, NY 11944, USA.
| |
Collapse
|
29
|
Chen SN, Zhang XW, Li L, Ruan BY, Huang B, Huang WS, Zou PF, Fu JP, Zhao LJ, Li N, Nie P. Evolution of IFN-λ in tetrapod vertebrates and its functional characterization in green anole lizard (Anolis carolinensis). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 61:208-224. [PMID: 27062970 DOI: 10.1016/j.dci.2016.04.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/05/2016] [Accepted: 04/05/2016] [Indexed: 06/05/2023]
Abstract
IFN-λ (IFNL), i.e. type III IFN genes were found in a conserved gene locus in tetrapod vertebrates. But, a unique locus containing IFNL was found in avian. In turtle and crocodile, IFNL genes were distributed in these two separate loci. As revealed in phylogenetic trees, IFN-λs in these two different loci and other amniotes were grouped into two different clades. The conservation in gene presence and gene locus was also observed for the receptors of IFN-λ, IFN-λR1 and IL-10RB in tetrapods. It is further revealed that in North American green anole lizard Anolis carolinensis, a single IFNL gene was situated collinearly in the conserved locus as in other tetrapods, together with its receptors IFN-λR1 and IL-10RB also identified in this study. The IFN-λ and its receptors were expressed in all examined organs/tissues, and their expression was stimulated following the injection of polyI:polyC. The ISREs in promoter of IFN-λ in lizard were responsible to IRF3 as demonstrated using luciferase report system, and IFN-λ in lizard functioned through the receptors, IFN-λR1 and IL-10RB, as the up-regulation of ISGs was observed in ligand-receptor transfected, and also in recombinant IFN-λ stimulated, cell lines. Taken together, it is concluded that the mechanisms involved in type III IFN ligand-receptor system, and in its signalling pathway and its down-stream genes may be conserved in green anole lizard, and may even be so in tetrapods from xenopus to human.
Collapse
Affiliation(s)
- Shan Nan Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of the Chinese Academy of Sciences, Beijing 10049, China
| | - Xiao Wen Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of the Chinese Academy of Sciences, Beijing 10049, China
| | - Li Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of the Chinese Academy of Sciences, Beijing 10049, China
| | - Bai Ye Ruan
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of the Chinese Academy of Sciences, Beijing 10049, China
| | - Bei Huang
- College of Fisheries, Jimei University, Xiamen 361021, China
| | - Wen Shu Huang
- College of Fisheries, Jimei University, Xiamen 361021, China
| | - Peng Fei Zou
- College of Fisheries, Jimei University, Xiamen 361021, China
| | - Jian Ping Fu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Li Juan Zhao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Nan Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Pin Nie
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| |
Collapse
|
30
|
Type III interferon gene expression in response to influenza virus infection in chicken and duck embryonic fibroblasts. Mol Immunol 2015; 68:657-62. [DOI: 10.1016/j.molimm.2015.10.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 10/16/2015] [Accepted: 10/19/2015] [Indexed: 12/30/2022]
|
31
|
Galani IE, Koltsida O, Andreakos E. Type III interferons (IFNs): Emerging Master Regulators of Immunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 850:1-15. [PMID: 26324342 DOI: 10.1007/978-3-319-15774-0_1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lambda interferons (IFN-λs), type III interferons or interleukins 28 and 29 are the latest addition to the class II cytokine family. They share low homology with the interferon (IFN) and IL-10 cytokine families, yet they exhibit common and unique activities, the full spectrum of which still remains incompletely understood. Although initially described for their antiviral functions, it is now appreciated that IFN-λs also mediate diverse antitumor and immune-modulatory effects, and are key determinants of innate immunity at mucosal sites such as the gastrointestinal and respiratory tracks. Here, we are reviewing the biological functions of IFN-λs, the mechanisms controlling their expression, their downstream effects and their role in the maintenance of homeostasis and disease. We are also exploring the potential application of IFN-λs as novel therapeutics.
Collapse
Affiliation(s)
- Ioanna E Galani
- Department of Immunology, Center for Translational and Clinical Research, Biomedical Research Foundation, Academy of Athens, 11527, Athens, Greece
| | | | | |
Collapse
|
32
|
Zhou H, Chen S, Wang M, Cheng A. Interferons and Their Receptors in Birds: A Comparison of Gene Structure, Phylogenetic Analysis, and Cross Modulation. Int J Mol Sci 2014; 15:21045-68. [PMID: 25405736 PMCID: PMC4264211 DOI: 10.3390/ijms151121045] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 10/26/2014] [Accepted: 11/04/2014] [Indexed: 11/17/2022] Open
Abstract
Interferon may be thought of as a key, with the interferon receptor as the signal lock: Crosstalk between them maintains their balance during viral infection. In this review, the protein structure of avian interferon and the interferon receptor are discussed, indicating remarkable similarity between different species. However, the structures of the interferon receptors are more sophisticated than those of the interferons, suggesting that the interferon receptor is a more complicated signal lock system and has considerable diversity in subtypes or structures. Preliminary evolutionary analysis showed that the subunits of the interferon receptor formed a distinct clade, and the orthologs may be derived from the same ancestor. Furthermore, the development of interferons and interferon receptors in birds may be related to an animal’s age and the maintenance of a balanced state. In addition, the equilibrium between interferon and its receptor during pathological and physiological states revealed that the virus and the host influence this equilibrium. Birds could represent an important model for studies on interferon’s antiviral activities and may provide the basis for new antiviral strategies.
Collapse
Affiliation(s)
- Hao Zhou
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Shun Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Mingshu Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Anchun Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
33
|
Goossens KE, Karpala AJ, Rohringer A, Ward A, Bean AGD. Characterisation of chicken viperin. Mol Immunol 2014; 63:373-80. [PMID: 25311379 DOI: 10.1016/j.molimm.2014.09.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 09/17/2014] [Accepted: 09/17/2014] [Indexed: 12/17/2022]
Abstract
The identification of immune pathways that protect against pathogens may lead to novel molecular therapies for both livestock and human health. Interferon (IFN) is a major response pathway that stimulates multiple genes targeted towards reducing virus. Viperin is one such interferon stimulated gene (ISG) that helps protect mammals from virus and may be critical to protecting chickens in the same way. In chickens, ISGs are not generally well characterised and viperin, in concert with other ISGs, may be important in protecting against virus. Here we identify chicken viperin (ch-viperin) and show that ch-viperin is upregulated in response to viral signature molecules. We further show that viperin is upregulated in response to virus infection in vivo. This data will benefit investigators targeting the antiviral pathways in the chicken.
Collapse
Affiliation(s)
- Kate E Goossens
- CSIRO Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia; School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Adam J Karpala
- CSIRO Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia.
| | - Andreas Rohringer
- CSIRO Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia
| | - Alistair Ward
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Andrew G D Bean
- CSIRO Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia
| |
Collapse
|
34
|
Goossens KE, Karpala AJ, Ward A, Bean AGD. Characterisation of chicken ZAP. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 46:373-381. [PMID: 24877657 DOI: 10.1016/j.dci.2014.05.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 05/19/2014] [Accepted: 05/19/2014] [Indexed: 06/03/2023]
Abstract
Emerging pathogenic viruses, such as avian influenza (AI), represent a serious threat to the poultry industry and human health. The development of novel therapeutics to protect against these viruses is critical and necessitates understanding the host immune mechanisms to find new pathways for protection against virus infection. Interferon (IFN) is a major antiviral arm of the immune system and is generally the first line of defence against virus. The multiple genes orchestrated by IFN upregulation are not well characterised in chickens due to a lack of reagents and research efforts. Here we have identified chicken ZAP (chZAP), an IFN stimulated gene (ISG), that has antiviral properties in human models, and show that chZAP is upregulated in response to PAMPs. Moreover, we show that chZAP is upregulated in vivo following particular viral infections. This data will benefit further studies that aim to understand antiviral response pathways in the chicken.
Collapse
Affiliation(s)
- Kate E Goossens
- CSIRO Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia; School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Adam J Karpala
- CSIRO Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia.
| | - Alistair Ward
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Andrew G D Bean
- CSIRO Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia
| |
Collapse
|
35
|
Yao Q, Fischer KP, Arnesen K, Tyrrell DL, Gutfreund KS. Molecular cloning, expression and characterization of Pekin duck interferon-λ. Gene 2014; 548:29-38. [PMID: 24992029 DOI: 10.1016/j.gene.2014.06.066] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Revised: 06/24/2014] [Accepted: 06/28/2014] [Indexed: 12/16/2022]
Abstract
Interferons (IFNs) are the first line of defense against viral infections in vertebrates. Type III interferon (IFN-λ) is recognized for its key role in innate immunity of tissues of epithelial origin. Here we describe the identification of the Pekin duck IFN-λ ortholog (duIFN-λ). The predicted duIFN-λ protein has an amino acid identity of 63%, 38%, 37% and 33% with chicken IFN-λ and human IFN-λ3, IFN-λ2 and IFN-λ1, respectively. The duck genome contains a single IFN-λ gene that is comprised of five exons and four introns. Recombinant duIFN-λ up-regulated OASL and Mx-1 mRNA in primary duck hepatocytes. Our observations suggest evolutionary conservation of genomic organization and structural features implicated in receptor binding and antiviral activity. The identification and expression of duIFN-λ will facilitate further study of the role of type III IFN in antiviral defense and inflammatory responses of the Pekin duck, a non-mammalian vertebrate and pathogen host with relevance for human and animal health.
Collapse
Affiliation(s)
- Qingxia Yao
- Department of Medicine, University of Alberta, Edmonton, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Karl P Fischer
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Karina Arnesen
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - D Lorne Tyrrell
- Department of Medicine, University of Alberta, Edmonton, AB, Canada; Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Klaus S Gutfreund
- Department of Medicine, University of Alberta, Edmonton, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
36
|
Fan W, Xu L, Ren L, Qu H, Li J, Liang J, Liu W, Yang L, Luo T. Functional characterization of canine interferon-lambda. J Interferon Cytokine Res 2014; 34:848-57. [PMID: 24950142 DOI: 10.1089/jir.2014.0009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
In this study, we provide the first comprehensive annotation of canine interferon-λ (CaIFN-λ, type III IFN). Phylogenetic analysis based on genomic sequences indicated that CaIFN-λ is located in the same branch with Swine IFN-λ1 (SwIFN-λ), Bat IFN-λ1 (BaIFN-λ), and human IFN-λ1 (HuIFN-λ1). CaIFN-λ was cloned, expressed in Escherichia coli, and purified to further investigate the biological activity in vitro. The recombinant CaIFN-λ (rCaIFN-λ) displayed potent antiviral activity on both homologous and heterologous animal cells in terms of inhibiting the replication of the New Jersey serotype of vesicular stomatitis virus (VSV), canine parvovirus, and influenza virus A/WSN/33 (H1N1), respectively. In addition, we also found that rCaIFN-λ exhibits a significant antiproliferative response against A72 canine tumor cells and MDCK cells in a dose-dependent manner. Furthermore, CaIFN-λ activated the JAK-STAT signaling pathway. To evaluate the expression of CaIFN-λ induced by virus and the expression of IFN-stimulated genes (ISGs) induced by rCaIFN-λ in the MDCK cells, we measured the relative mRNA level of CaIFN-λ and ISGs (ISG15, Mx1, and 2'5'-OAS) by quantitative real-time PCR and found that the mRNA level of CaIFN-λ and the ISGs significantly increased after treating the MDCK cells with viruses and rCaIFN-λ protein, respectively. Finally, to evaluate the binding activity of rCaIFN-λ to its receptor, we expressed the extracellular domain of the canine IFN-λ receptor 1 (CaIFN-λR1-EC) and determined the binding activity via ELISA. Our results demonstrated that rCaIFN-λ bound tightly to recombinant CaIFN-λR1-EC (rCaIFN-λR1-EC).
Collapse
Affiliation(s)
- Wenhui Fan
- 1 College of Animal Sciences and Veterinary Medicine, Guangxi University , Nanning, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Misumi I, Whitmire JK. IFN-λ exerts opposing effects on T cell responses depending on the chronicity of the virus infection. THE JOURNAL OF IMMUNOLOGY 2014; 192:3596-606. [PMID: 24646741 DOI: 10.4049/jimmunol.1301705] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
IFN-λ induces an antiviral state in many cell types and may contribute to the overall inflammatory environment after infection. Either of these effects may influence adaptive immune responses, but the role of type 3 IFNs in the development of primary and memory T cell responses to infection has not been evaluated. In this study, we examined T cell responses to acute or persistent lymphocytic choriomeningitis virus infection in IFN-λR1-deficient mice. Following acute infection, we find that IFN-λR1-deficient mice produced normal levels of IFN, robust NK cell responses, but greater than normal CD4+ and CD8+ T cell responses compared with wild type BALB/c mice. There were more T cells that were IL-7R(hi) and, correspondingly, the IFN-λR-deficient mice showed a 2- to 3-fold increase in memory T cell number. The inhibitory effect of IFN-λR expression was independent of direct cytokine signaling into T cells. In contrast with acute infection, the IFN-λR-deficient mice generated markedly diminished T cell responses and had greater weight loss compared with wild type mice when confronted with a highly disseminating variant of lymphocytic choriomeningitis virus. These data indicate that IFN-λR limits T cell responses and memory after transient infection but augments T cell responses during persisting infection. Thus, the immune-regulatory functions for IFN-λR are complex and vary with the overall inflammatory environment.
Collapse
Affiliation(s)
- Ichiro Misumi
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | | |
Collapse
|
38
|
Burggraaf S, Karpala AJ, Bingham J, Lowther S, Selleck P, Kimpton W, Bean AGD. H5N1 infection causes rapid mortality and high cytokine levels in chickens compared to ducks. Virus Res 2014; 185:23-31. [PMID: 24657784 PMCID: PMC7127704 DOI: 10.1016/j.virusres.2014.03.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 02/06/2014] [Accepted: 03/11/2014] [Indexed: 12/19/2022]
Abstract
Chickens are acutely susceptible to influenza when compared with ducks. The chicken innate immune responses to influenza are more pronounced than ducks. IL6 is rapidly upregulated in chickens compared to ducks. Cytokine profile, especially IL6, may lead to acute pathogenicity in chickens.
Infection with H5N1 influenza virus is often fatal to poultry with death occurring in hours rather than days. However, whilst chickens may be acutely susceptible, ducks appear to be asymptomatic to H5N1. The mechanisms of disease pathogenesis are not well understood and the variation between different species requires investigation to help explain these species differences. Here we investigated the expression of several key proinflammatory cytokines of chickens and ducks following infection with 2 highly pathogenic H5N1 (A/Muscovy duck/Vietnam/453/2004 (Vt453) and A/Duck/Indramayu/BBVW/109/2006 (Ind109)) and a low-pathogenic H5N3 influenza virus (A/Duck/Victoria/1462/2008 (Vc1462)). H5N1 viruses caused fatal infections in chickens as well as high viral loads and increased production of proinflammatory molecules when compared to ducks. Cytokines, including Interleukin 6 (IL6) and the acute phase protein Serum Amyloid A (SAA), were rapidly induced at 24 h post infection with H5N1. In contrast, low induction of these cytokines appeared in ducks and only at later times during the infection period. These observations support that hypercytokinemia may contribute to pathogenesis in chickens, whilst the lower cytokine response in ducks may be a factor in their apparent resistance to disease and decreased mortality.
Collapse
Affiliation(s)
- Simon Burggraaf
- CSIRO Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia; School of Veterinary Science, The University of Melbourne, Parkville, Victoria, Australia
| | - Adam J Karpala
- CSIRO Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia.
| | - John Bingham
- CSIRO Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia
| | - Sue Lowther
- CSIRO Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia
| | - Paul Selleck
- CSIRO Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia
| | - Wayne Kimpton
- School of Veterinary Science, The University of Melbourne, Parkville, Victoria, Australia
| | - Andrew G D Bean
- CSIRO Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia
| |
Collapse
|
39
|
Abstract
Interferons (IFNs) are essential components of the antiviral defense system of vertebrates. In mammals, functional receptors for type III IFN (lambda interferon [IFN-λ]) are found mainly on epithelial cells, and IFN-λ was demonstrated to play a crucial role in limiting viral infections of mucosal surfaces. To determine whether IFN-λ plays a similar role in birds, we produced recombinant chicken IFN-λ (chIFN-λ) and we used the replication-competent retroviral RCAS vector system to generate mosaic-transgenic chicken embryos that constitutively express chIFN-λ. We could demonstrate that chIFN-λ markedly inhibited replication of various virus strains, including highly pathogenic influenza A viruses, in ovo and in vivo, as well as in epithelium-rich tissue and cell culture systems. In contrast, chicken fibroblasts responded poorly to chIFN-λ. When applied in vivo to 3-week-old chickens, recombinant chIFN-λ strongly induced the IFN-responsive Mx gene in epithelium-rich organs, such as lungs, tracheas, and intestinal tracts. Correspondingly, these organs were found to express high transcript levels of the putative chIFN-λ receptor alpha chain (chIL28RA) gene. Transfection of chicken fibroblasts with a chIL28RA expression construct rendered these cells responsive to chIFN-λ treatment, indicating that receptor expression determines cell type specificity of IFN-λ action in chickens. Surprisingly, mosaic-transgenic chickens perished soon after hatching, demonstrating a detrimental effect of constitutive chIFN-λ expression. Our data highlight fundamental similarities between the IFN-λ systems of mammals and birds and suggest that type III IFN might play a role in defending mucosal surfaces against viral intruders in most if not all vertebrates.
Collapse
|
40
|
Guo P, Thomas JD, Bruce MP, Hinton TM, Bean AGD, Lowenthal JW. The chicken TH1 response: potential therapeutic applications of ChIFN-γ. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 41:389-396. [PMID: 23707786 DOI: 10.1016/j.dci.2013.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 05/13/2013] [Indexed: 06/02/2023]
Abstract
The outcomes of viral infections are costly in terms of human and animal health and welfare worldwide. The observed increase in the virulence of some viruses and failure of many vaccines to stop these infections has lead to the apparent need to develop new anti-viral strategies. One approach to dealing with viral infection may be to employ the therapeutic administration of recombinant cytokines to act as 'immune boosters' to assist in augmenting the host response to virus. With this in mind, a greater understanding of the immune response, particularly cell mediated T-helper-1 (TH1) type responses, is imperative to the development of new anti-viral and vaccination strategies. Following the release of the chicken genome, a number of TH1-type cytokines have been identified, including chicken interleukin-12 (ChIL-12), ChIL-18 and interferon-γ ChIFN-γ), highlighting the nature of the TH1-type response in this non-mammalian vertebrate. To date a detailed analysis of the in vivo biological function of these cytokines has been somewhat hampered by access to large scale production techniques. This review describes the role of TH-1 cytokines in immune responses to viruses and explores their potential use in enhancing anti-viral treatment strategies in chickens. Furthermore, this review focuses on the example of ChIFN-γ treatment of Chicken Anemia Virus (CAV) infection. CAV causes amongst other things thymocyte depletion and thymus atrophy, as well as immunosuppression in chickens. However, due to vaccination, clinical disease appears less often, nevertheless, the subclinical form of the disease is often associated with secondary complicating infections due to an immunocompromised state. Since CAV-induced immunosuppression can cause a marked decrease in the immune response against other pathogens, understanding this aspect of the disease is critically important, as well as providing insights into developing new control approaches. With increasing emphasis on developing alternative control programs for poultry diseases, novel therapeutic strategies provide one approach. We show here that the in ovo administration of ChIFN-γ impacts the depletion of T-cell precursors during CAV infection. Therefore, it appears that ChIFN-γ may have the potential to be used as a novel therapeutic reagent to impact virus infection and alter immunosuppression caused by CAV and potentially other pathogens.
Collapse
Affiliation(s)
- Pengju Guo
- CSIRO Biosecurity Flagship, Australian Animal Health Laboratory, Geelong, Vic, Australia
| | | | | | | | | | | |
Collapse
|
41
|
Goossens KE, Ward AC, Lowenthal JW, Bean AGD. Chicken interferons, their receptors and interferon-stimulated genes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 41:370-376. [PMID: 23751330 DOI: 10.1016/j.dci.2013.05.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 05/31/2013] [Accepted: 05/31/2013] [Indexed: 06/02/2023]
Abstract
The prevalence of pathogenic viruses is a serious issue as they pose a constant threat to both the poultry industry and to human health. To prevent these viral infections an understanding of the host-virus response is critical, especially for the development of novel therapeutics. One approach in the control of viral infections would be to boost the immune response through administration of cytokines, such as interferons. However, the innate immune response in chickens is poorly characterised, particularly concerning the interferon pathway. This review will provide an overview of our current understanding of the interferon system of chickens, including their cognate receptors and known interferon-stimulated gene products.
Collapse
Affiliation(s)
- Kate E Goossens
- CSIRO Biosecurity Flagship, Australian Animal Health Laboratories, Geelong, VIC, Australia
| | | | | | | |
Collapse
|
42
|
Li ML, Xu WW, Gao YD, Guo Y, Wang WJ, Wang C, Jiang SY, Willden A, Huang JF, Zhang HT. Interferon-lambda3 (IFN-λ3) and its cognate receptor subunits in tree shrews (Tupaia belangeri): genomic sequence retrieval, molecular identification and expression analysis. PLoS One 2013; 8:e60048. [PMID: 23555878 PMCID: PMC3610868 DOI: 10.1371/journal.pone.0060048] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 02/20/2013] [Indexed: 02/06/2023] Open
Abstract
Type III IFNs (IFN-λs) constitute a new subfamily with antiviral activities by signaling through a unique receptor complex composed of IFN-λs receptor 1 (IFNλR1) and interleukin-10 receptor 2 (IL10R2). As tree shrews (Tupaia belangeri) have shown susceptiblility to several human viruses, they are a potentially important model for analyzing viral infection. However, little is known about their IFN-λs system. We used the tree shrew genome to retrieve IFN-λs and their receptor contig sequences by BLASTN and BLASTZ algorithms, and GenScan was used to scan transcripts from the putative contig sequences. RT-PCR and bioinformatic methods were then used to clone and characterize the IFN-λs system. Due to its highest identity with human IFN-λ3, we opted to define one intact IFN-λ gene, tsIFN-λ3, as well as its two receptor subunits, tsIFNλR1 and tsIL10R2. Additionally, our results showed that tsIFN-λ3 contained many features conserved in IFN-λ3 genes from other mammals, including conserved signal peptide cleavage and glycosylation sites, and several residues responsible for binding to the type III IFNR. We also found six transcript variants in the receptors: three in tsIFNλR1, wherein different extracellular regions exist in three transmembrane proteins, resulting in different affinities with IFN-λs; and three more variants in tsIL10R2, encoding one transmembrane and two soluble proteins. Based on tissue distribution in the liver, heart, brain, lung, intestine, kidney, spleen, and stomach, we found that IFN-λs receptor complex was expressed in a variety of organs although the expression level differed markedly between them. As the first study to find transcript variants in IL-10R2, our study offers novel insights that may have important implications for the role of IFN-λs in tree shrews’ susceptibility with a variety of human viruses, bolstering the arguments for using tree shrews as an animal model in the study of human viral infections.
Collapse
Affiliation(s)
- Ming-Li Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Wen-Wen Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Yue-Dong Gao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yan Guo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - Wen-Ju Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - Chao Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - Shi-You Jiang
- University of the Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Andrew Willden
- Editorial Department, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jing-Fei Huang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Hua-Tang Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, China
- Chongqing Center for Biomedical Research and Equipment Development, Chongqing Academy of Science and Technology, Chongqing, China
- * E-mail:
| |
Collapse
|
43
|
Qu H, Yang L, Meng S, Xu L, Bi Y, Jia X, Li J, Sun L, Liu W. The differential antiviral activities of chicken interferon α (ChIFN-α) and ChIFN-β are related to distinct interferon-stimulated gene expression. PLoS One 2013; 8:e59307. [PMID: 23527158 PMCID: PMC3602166 DOI: 10.1371/journal.pone.0059307] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 02/15/2013] [Indexed: 01/26/2023] Open
Abstract
Chicken interferon α (ChIFN-α) and ChIFN-β are type I IFNs that are important antiviral cytokines in the innate immune system. In the present study, we identified the virus-induced expression of ChIFN-α and ChIFN-β in chicken fibroblast DF-1 cells and systematically evaluated the antiviral activities of recombinant ChIFN-α and ChIFN-β by cytopathic-effect (CPE) inhibition assays. We found that ChIFN-α exhibited stronger antiviral activity than ChIFN-β in terms of inhibiting the replication of vesicular stomatitis virus, Newcastle disease virus and avian influenza virus, respectively. To elucidate the mechanism of differential antiviral activities between the two ChIFNs, we measured the relative mRNA levels of IFN-stimulated genes (ISGs) in IFN-treated DF-1 cells by real-time PCR. ChIFN-α displayed greater induction potency than ChIFN-β on several ISGs encoding antiviral proteins and MHC-I, whereas ChIFN-α was less potent than ChIFN-β for inducing ISGs involved in signaling pathways. In conclusion, ChIFN-α and ChIFN-β presented differential induction potency on various sets of ISGs, and the stronger antiviral activity of ChIFN-α is likely attributed to the greater expression levels of downstream antiviral ISGs.
Collapse
Affiliation(s)
- Hongren Qu
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Graduate University of Chinese Academy of Sciences, Beijing, China
| | - Limin Yang
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Shanshan Meng
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Graduate University of Chinese Academy of Sciences, Beijing, China
| | - Lei Xu
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Graduate University of Chinese Academy of Sciences, Beijing, China
| | - Yuhai Bi
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Xiaojuan Jia
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jing Li
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Lei Sun
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Wenjun Liu
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Graduate University of Chinese Academy of Sciences, Beijing, China
- China-Japan Joint Laboratory of Molecular Immunology and Molecular Microbiology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
44
|
[Genome-wide prediction of interferon family members of tree shrew and their molecular characteristics analysis]. DONG WU XUE YAN JIU = ZOOLOGICAL RESEARCH 2012; 33:67-74. [PMID: 22345011 DOI: 10.3724/sp.j.1141.2012.01067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Interferons (IFNs) represent proteins with antiviral activities that are secreted from cells in response to a variety of stimuli. In addition to antiviral, antibacterial and anti-parasitic host-defense functions they are now also recognized as crucial regulators of cell proliferation, differentiation, survival and death as well as activators of specialized cell functions particularly in the immune system and play important roles in infectious and inflammatory diseases, autoimmunity and cancer. Tree shrews (Tupaia belangeri) were found to be susceptible to several human viruses and therefore are widely regarded as good models for analyzing mechanism of human diseases. In this report, we have forecasted the interferon family members of tree shrew from its genome mainly using the methods like Blast (whole genome shotgun sequence) and gene prediction. Our data show that tree shrew interferon system includes: type I IFN: α (five subtypes), β, ω, κ, epsilon, δ; type II IFN: γ; type III IFN: λ1, λ2/3. Furthermore, the predicted structures of α and λ have similar character with those of other mammals. However, there are some differences in cysteine position and N-glycosylation numbers between human and Tree shrew IFNs. These results provide fundamental basis for further molecular cloning and function analysis of tree shrew IFNs in future.
Collapse
|
45
|
Karpala AJ, Bagnaud-Baule A, Goossens KE, Lowenthal JW, Bean AGD. Ontogeny of the interferon system in chickens. J Reprod Immunol 2012; 94:169-74. [PMID: 22472789 DOI: 10.1016/j.jri.2012.02.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 02/14/2012] [Accepted: 02/22/2012] [Indexed: 12/13/2022]
Abstract
Newborn vertebrates may be susceptible to infection because the immature status of their immune system results in an inability to make an effective immune response. Consequently, newly hatched chicks appear to be more susceptible to infections than mature chickens. In particular, poultry susceptibility to virus infection may be related to poor expression of innate immune elements involved in antiviral responses. Therefore, in this study we assessed the relative development of the interferon (IFN) system: a protective system against virus infection. We investigated the age-related expression of the elements involved in the IFN response including IFN gene expression, their associated receptors and the pattern recognition receptors (PRR) involved in the regulation of IFNs. We observed that the IFN system is somewhat inadequately expressed in embryos and develops over time, just prior to and after hatching, and therefore chicks may be more susceptible to virus than mature birds because of an immature IFN network.
Collapse
Affiliation(s)
- Adam J Karpala
- CSIRO, Livestock Industries, AAHL, 5, Portarlington Road, Geelong, Victoria, 3220 Australia.
| | | | | | | | | |
Collapse
|
46
|
Bovine type III interferon significantly delays and reduces the severity of foot-and-mouth disease in cattle. J Virol 2012; 86:4477-87. [PMID: 22301155 DOI: 10.1128/jvi.06683-11] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Interferons (IFNs) are the first line of defense against viral infections. Although type I and II IFNs have proven effective to inhibit foot-and-mouth disease virus (FMDV) replication in swine, a similar approach had only limited efficacy in cattle. Recently, a new family of IFNs, type III IFN or IFN-λ, has been identified in human, mouse, chicken, and swine. We have identified bovine IFN-λ3 (boIFN-λ3), also known as interleukin 28B (IL-28B), and demonstrated that expression of this molecule using a recombinant replication-defective human adenovirus type 5 (Ad5) vector, Ad5-boIFN-λ3, exhibited antiviral activity against FMDV in bovine cell culture. Furthermore, inoculation of cattle with Ad5-boIFN-λ3 induced systemic antiviral activity and upregulation of IFN-stimulated gene expression in the upper respiratory airways and skin. In the present study, we demonstrated that disease could be delayed for at least 6 days when cattle were inoculated with Ad5-boIFN-λ3 and challenged 24 h later by intradermolingual inoculation with FMDV. Furthermore, the delay in the appearance of disease was significantly prolonged when treated cattle were challenged by aerosolization of FMDV, using a method that resembles the natural route of infection. No clinical signs of FMD, viremia, or viral shedding in nasal swabs was found in the Ad5-boIFN-λ3-treated animals for at least 9 days postchallenge. Our results indicate that boIFN-λ3 plays a critical role in the innate immune response of cattle against FMDV. To this end, this work represents the most successful biotherapeutic strategy so far tested to control FMDV in cattle.
Collapse
|
47
|
Stewart CR, Bagnaud-Baule A, Karpala AJ, Lowther S, Mohr PG, Wise TG, Lowenthal JW, Bean AG. Toll-like receptor 7 ligands inhibit influenza A infection in chickens. J Interferon Cytokine Res 2011; 32:46-51. [PMID: 21929369 DOI: 10.1089/jir.2011.0036] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Avian influenza virus is endemic in many regions around the world and remains a pandemic threat, a scenario tied closely to outbreaks of the virus in poultry. The innate immune system, in particular the nucleic acid-sensing toll-like receptors (TLRs) -3, -7, -8, and -9, play a major role in coordinating antiviral immune responses. In this study we have investigated the use of TLR ligands as antivirals against influenza A in chickens. The TLR7 ligand poly-C inhibited low-path influenza A growth in the chicken macrophage cell line HD-11 more effectively than poly(I:C), which acts via TLR3. The TLR7 ligand 7-allyl-8-oxoguanosine (loxoribine) inhibited influenza A replication in vitro and in ovo in a dose-dependent manner. Treatment of primary chicken splenocytes with loxoribine resulted in the induction of interferons-α, -β, and -λ, and interferon-stimulated genes PKR and Mx. These results demonstrate that nucleic acid-sensing TLR ligands show considerable potential as antivirals in chickens and could be incorporated into antiviral strategies.
Collapse
Affiliation(s)
- Cameron R Stewart
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Victoria, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Penski N, Härtle S, Rubbenstroth D, Krohmann C, Ruggli N, Schusser B, Pfann M, Reuter A, Gohrbandt S, Hundt J, Veits J, Breithaupt A, Kochs G, Stech J, Summerfield A, Vahlenkamp T, Kaspers B, Staeheli P. Highly pathogenic avian influenza viruses do not inhibit interferon synthesis in infected chickens but can override the interferon-induced antiviral state. J Virol 2011; 85:7730-41. [PMID: 21613402 PMCID: PMC3147912 DOI: 10.1128/jvi.00063-11] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 05/17/2011] [Indexed: 01/13/2023] Open
Abstract
From infection studies with cultured chicken cells and experimental mammalian hosts, it is well known that influenza viruses use the nonstructural protein 1 (NS1) to suppress the synthesis of interferon (IFN). However, our current knowledge regarding the in vivo role of virus-encoded NS1 in chickens is much more limited. Here, we report that highly pathogenic avian influenza viruses of subtypes H5N1 and H7N7 lacking fully functional NS1 genes were attenuated in 5-week-old chickens. Surprisingly, in diseased birds infected with NS1 mutants, the IFN levels were not higher than in diseased birds infected with wild-type virus, suggesting that NS1 cannot suppress IFN gene expression in at least one cell population of infected chickens that produces large amounts of the cytokine in vivo. To address the question of why influenza viruses are highly pathogenic in chickens although they strongly activate the innate immune system, we determined whether recombinant chicken alpha interferon (IFN-α) can inhibit the growth of highly pathogenic avian influenza viruses in cultured chicken cells and whether it can ameliorate virus-induced disease in 5-week-old birds. We found that IFN treatment failed to confer substantial protection against challenge with highly pathogenic viruses, although it was effective against viruses with low pathogenic potential. Taken together, our data demonstrate that preventing the synthesis of IFN is not the primary role of the viral NS1 protein during infection of chickens. Our results further suggest that virus-induced IFN does not contribute substantially to resistance of chickens against highly pathogenic influenza viruses.
Collapse
Affiliation(s)
- Nicola Penski
- Department of Virology, University of Freiburg, Freiburg, Germany
| | - Sonja Härtle
- Department of Veterinary Sciences, University of Munich, Munich, Germany
| | | | - Carsten Krohmann
- Department of Veterinary Sciences, University of Munich, Munich, Germany
| | - Nicolas Ruggli
- Institute of Virology and Immunoprophylaxis, Mittelhäusern, Switzerland
| | - Benjamin Schusser
- Department of Veterinary Sciences, University of Munich, Munich, Germany
| | - Michael Pfann
- Department of Virology, University of Freiburg, Freiburg, Germany
| | - Antje Reuter
- Department of Virology, University of Freiburg, Freiburg, Germany
- International Max Planck Research School for Molecular and Cellular Biology, Freiburg, Germany
| | | | - Jana Hundt
- Friedrich-Loeffler-Institut, Isle of Riems, Germany
| | - Jutta Veits
- Friedrich-Loeffler-Institut, Isle of Riems, Germany
| | | | - Georg Kochs
- Department of Virology, University of Freiburg, Freiburg, Germany
| | - Jürgen Stech
- Friedrich-Loeffler-Institut, Isle of Riems, Germany
| | - Artur Summerfield
- Institute of Virology and Immunoprophylaxis, Mittelhäusern, Switzerland
| | | | - Bernd Kaspers
- Department of Veterinary Sciences, University of Munich, Munich, Germany
| | - Peter Staeheli
- Department of Virology, University of Freiburg, Freiburg, Germany
| |
Collapse
|
49
|
Stewart CR, Karpala AJ, Lowther S, Lowenthal JW, Bean AG. Immunostimulatory motifs enhance antiviral siRNAs targeting highly pathogenic avian influenza H5N1. PLoS One 2011; 6:e21552. [PMID: 21747939 PMCID: PMC3128588 DOI: 10.1371/journal.pone.0021552] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 06/01/2011] [Indexed: 01/01/2023] Open
Abstract
Highly pathogenic avian influenza (HPAI) H5N1 virus is endemic in many regions around the world and remains a significant pandemic threat. To date H5N1 has claimed almost 300 human lives worldwide, with a mortality rate of 60% and has caused the death or culling of hundreds of millions of poultry since its initial outbreak in 1997. We have designed multi-functional RNA interference (RNAi)-based therapeutics targeting H5N1 that degrade viral mRNA via the RNAi pathway while at the same time augmenting the host antiviral response by inducing host type I interferon (IFN) production. Moreover, we have identified two factors critical for maximising the immunostimulatory properties of short interfering (si)RNAs in chicken cells (i) mode of synthesis and (ii) nucleoside sequence to augment the response to virus. The 5-bp nucleoside sequence 5′-UGUGU-3′ is a key determinant in inducing high levels of expression of IFN -α, -β, -λ and interleukin 1- β in chicken cells. Positioning of this 5′-UGUGU-3′ motif at the 5′- end of the sense strand of siRNAs, but not the 3′- end, resulted in a rapid and enhanced induction of type I IFN. An anti-H5N1 avian influenza siRNA directed against the PB1 gene (PB1-2257) tagged with 5′-UGUGU-3′ induced type I IFN earlier and to a greater extent compared to a non-tagged PB1-2257. Tested against H5N1 in vitro, the tagged PB1-2257 was more effective than non-tagged PB1-2257. These data demonstrate the ability of an immunostimulatory motif to improve the performance of an RNAi-based antiviral, a finding that may influence the design of future RNAi-based anti-influenza therapeutics.
Collapse
Affiliation(s)
- Cameron R Stewart
- Infection and Immunity, The Commonwealth Scientific and Industrial Research Organisation Australian Animal Health Laboratory, Geelong, Victoria, Australia.
| | | | | | | | | |
Collapse
|
50
|
Karpala AJ, Lowenthal JW, Bean AGD. Identifying innate immune pathways of the chicken may lead to new antiviral therapies. Vet Immunol Immunopathol 2011; 148:100-9. [PMID: 21715024 DOI: 10.1016/j.vetimm.2011.05.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 03/25/2011] [Accepted: 05/30/2011] [Indexed: 12/23/2022]
Abstract
Zoonotic viruses, such as highly pathogenic avian influenza (HPAI), present a significant threat to both the poultry industry and public health. The present method of controlling avian influenza (AI) relies on good farming practice with limited use of vaccination in some countries. However, new ways to control disease outbreaks might be possible with additional knowledge of the natural host response to virus. Moreover, manipulation of the innate immune system in mammals improves the outcomes following viral infection. A similar approach might be applied to the chicken, nevertheless, a greater knowledge of the chicken innate immune system is required. This review outlines important mammalian antiviral mechanisms that have been modulated to strengthen viral immunity and highlights the potential application of these strategies in the chicken, especially in regards, to AI.
Collapse
Affiliation(s)
- Adam J Karpala
- CSIRO, Livestock Industries, Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia.
| | | | | |
Collapse
|