1
|
Tian S, Si J, Zhang L, Zeng J, Zhang X, Huang C, Li G, Lei C, Zhou X, Geng R, Zhou P, Yan H, Rossiter SJ, Zhao H. Comparative genomics provides insights into chromosomal evolution and immunological adaptation in horseshoe bats. Nat Ecol Evol 2025; 9:705-720. [PMID: 39920351 DOI: 10.1038/s41559-025-02638-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 01/08/2025] [Indexed: 02/09/2025]
Abstract
Horseshoe bats are natural hosts of zoonotic viruses, yet the genetic basis of their antiviral immunity is poorly understood. Here we generated two new chromosomal-level genome assemblies for horseshoe bat species (Rhinolophus) and three close relatives, and show that, during their diversification, horseshoe bats underwent extensive chromosomal rearrangements and gene expansions linked to segmental duplications. These expansions have generated new adaptive variations in type I interferons and the interferon-stimulated gene ANXA2R, which potentially enhance antiviral states, as suggested by our functional assays. Genome-wide selection screens, including of candidate introgressed regions, uncover numerous putative molecular adaptations linked to immunity, including in viral receptors. By expanding taxon coverage to ten horseshoe bat species, we identify new variants of the SARS-CoV-2 receptor ACE2, and report convergent functionally important residues that could explain wider patterns of susceptibility across mammals. We conclude that horseshoe bats have numerous signatures of adaptation, including some potentially related to immune response to viruses, in genomic regions with diverse and multiscale mutational changes.
Collapse
Affiliation(s)
- Shilin Tian
- State Key Laboratory of Virology and Biosafety, Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Frontier Science Center for Immunology and Metabolism, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
- Novogene Bioinformatics Institute, Beijing, China
| | - Junyu Si
- State Key Laboratory of Virology and Biosafety, Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Frontier Science Center for Immunology and Metabolism, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Libiao Zhang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China
| | - Jiaming Zeng
- State Key Laboratory of Virology and Biosafety, Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Frontier Science Center for Immunology and Metabolism, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xiangyi Zhang
- State Key Laboratory of Virology and Biosafety, Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Frontier Science Center for Immunology and Metabolism, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Chen Huang
- State Key Laboratory of Virology and Biosafety, Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Frontier Science Center for Immunology and Metabolism, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Gang Li
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Caoqi Lei
- State Key Laboratory of Virology and Biosafety, Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Frontier Science Center for Immunology and Metabolism, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xuming Zhou
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Rong Geng
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, China
| | - Peng Zhou
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, China
| | - Huan Yan
- State Key Laboratory of Virology and Biosafety, Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Frontier Science Center for Immunology and Metabolism, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Stephen J Rossiter
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK.
| | - Huabin Zhao
- State Key Laboratory of Virology and Biosafety, Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, Frontier Science Center for Immunology and Metabolism, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
2
|
da Silva MI, Ott T. Effects of conceptus proteins on endometrium and blood leukocytes of dairy cattle using transcriptome and meta-analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.25.591148. [PMID: 38712302 PMCID: PMC11071483 DOI: 10.1101/2024.04.25.591148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
This study investigates the short and long-term effects of IFNT and PAG on the transcriptome of endometrium and blood leukocytes. Holstein heifers received intrauterine infusions of one of the following treatments: 20 mL of a 200 μg/mL bovine serum albumin solution (BSA; vehicle) from day 14 to 16 of the estrous cycle (BSA), vehicle + 10 μg/mL of IFNT from day 14 to 16 (IFNT3), vehicle + 10 μg/mL of IFNT from day 14 to 19 (IFNT6), and vehicle + 10 μg/mL of IFNT from day 14 to 16 followed by vehicle + 10 μg/mL of IFNT + 5 μg/mL of PAG from day 17 to 19 (IFNT+PAG). RNA-seq analysis was performed in endometrial biopsies and blood leukocytes collected after treatments. Acute IFNT signaling in the endometrium (IFNT3 vs BSA), induced differentially expressed genes (DEG) associated with interferon activation, immune response, inflammation, cell death, and inhibited vesicle transport and extracellular matrix remodeling. Prolonged IFNT signaling (IFNT6 vs IFNT3) altered gene expression related to cell invasion, retinoic acid signaling, and embryo implantation. In contrast, PAG induced numerous DEG in blood leukocytes but only 4 DEG in the endometrium. In blood leukocytes, PAG stimulated genes involved in development and TGFB signaling while inhibiting interferon signaling and cell migration. Overall, IFNT is a primary regulator of endometrial gene expression, while PAG predominantly affected the transcriptome of circulating immune cells during early pregnancy. Further research is essential to fully grasp the roles of identified DEG in both the endometrium and blood leukocytes.
Collapse
Affiliation(s)
- Maria Isabel da Silva
- Department of Animal Science, Center for Reproductive Biology and Health, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - Troy Ott
- Department of Animal Science, Center for Reproductive Biology and Health, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
3
|
Rao SS, Nelson PA, Lunde HS, Haugland GT. Evolutionary, comparative, and functional analyses of STATs and regulation of the JAK-STAT pathway in lumpfish upon bacterial and poly(I:C) exposure. Front Cell Infect Microbiol 2023; 13:1252744. [PMID: 37808912 PMCID: PMC10556531 DOI: 10.3389/fcimb.2023.1252744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Background The Janus kinase/signal transducers and activators of transcription (JAK-STAT) system regulates several biological processes by affecting transcription of genes as a response to cytokines and growth factors. In the present study, we have characterized the STAT genes in lumpfish (Cyclopterus lumpus L.), belonging to the order Perciformes, and investigated regulation of the JAK-STAT signaling pathway upon exposure to bacteria (Vibrio anguillarum) and poly(I:C), the latter mimicking antiviral responses. Methods Characterization and evolutionary analyses of the STATs were performed by phylogeny, protein domain, homology similarity and synteny analyses. Antibacterial and antiviral responses were investigated by performing KEGG pathway analysis. Results We observed that lumpfish have stat1a, 2, 3, 4, 5a, 5b, and 6. Transcriptome-wide analyses showed that most components of the JAK-STAT pathway were present in lumpfish. il-6, il-10, il-21, iκBα and stat3 were upregulated 6 hours post exposure (hpe) against bacteria while type I interferons (IFNs), irf1, irf3, irf10, stat1 and 2 were upregulated 24 hpe against poly(I:C). Conclusions Our findings shed light on the diversity and evolution of the STATs and the data show that the STAT genes are highly conserved among fish, including lumpfish. The transcriptome-wide analyses lay the groundwork for future research into the functional significance of these genes in regulating critical biological processes and make an important basis for development of prophylactic measure such as vaccination, which is highly needed for lumpfish since it is vulnerable for both bacterial and viral diseases.
Collapse
Affiliation(s)
- Shreesha S Rao
- Department of Biological Sciences, Bergen High-Technology Centre, University of Bergen, Bergen, Norway
| | - Patrick A Nelson
- Department of Biological Sciences, Bergen High-Technology Centre, University of Bergen, Bergen, Norway
| | - Harald S Lunde
- Department of Biological Sciences, Bergen High-Technology Centre, University of Bergen, Bergen, Norway
| | - Gyri T Haugland
- Department of Biological Sciences, Bergen High-Technology Centre, University of Bergen, Bergen, Norway
| |
Collapse
|
4
|
Hardy A, Bakshi S, Furnon W, MacLean O, Gu Q, Varjak M, Varela M, Aziz MA, Shaw AE, Pinto RM, Cameron Ruiz N, Mullan C, Taggart AE, Da Silva Filipe A, Randall RE, Wilson SJ, Stewart ME, Palmarini M. The Timing and Magnitude of the Type I Interferon Response Are Correlated with Disease Tolerance in Arbovirus Infection. mBio 2023; 14:e0010123. [PMID: 37097030 PMCID: PMC10294695 DOI: 10.1128/mbio.00101-23] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/20/2023] [Indexed: 04/26/2023] Open
Abstract
Infected hosts possess two alternative strategies to protect themselves against the negative impact of virus infections: resistance, used to abrogate virus replication, and disease tolerance, used to avoid tissue damage without controlling viral burden. The principles governing pathogen resistance are well understood, while less is known about those involved in disease tolerance. Here, we studied bluetongue virus (BTV), the cause of bluetongue disease of ruminants, as a model system to investigate the mechanisms of virus-host interactions correlating with disease tolerance. BTV induces clinical disease mainly in sheep, while cattle are considered reservoirs of infection, rarely exhibiting clinical symptoms despite sustained viremia. Using primary cells from multiple donors, we show that BTV consistently reaches higher titers in ovine cells than cells from cattle. The variable replication kinetics of BTV in sheep and cow cells were mostly abolished by abrogating the cell type I interferon (IFN) response. We identified restriction factors blocking BTV replication, but both the sheep and cow orthologues of these antiviral genes possess anti-BTV properties. Importantly, we demonstrate that BTV induces a faster host cell protein synthesis shutoff in primary sheep cells than cow cells, which results in an earlier downregulation of antiviral proteins. Moreover, by using RNA sequencing (RNA-seq), we also show a more pronounced expression of interferon-stimulated genes (ISGs) in BTV-infected cow cells than sheep cells. Our data provide a new perspective on how the type I IFN response in reservoir species can have overall positive effects on both virus and host evolution. IMPORTANCE The host immune response usually aims to inhibit virus replication in order to avoid cell damage and disease. In some cases, however, the infected host avoids the deleterious effects of infection despite high levels of viral replication. This strategy is known as disease tolerance, and it is used by animal reservoirs of some zoonotic viruses. Here, using a virus of ruminants (bluetongue virus [BTV]) as an experimental system, we dissected virus-host interactions in cells collected from species that are susceptible (sheep) or tolerant (cow) to disease. We show that (i) virus modulation of the host antiviral type I interferon (IFN) responses, (ii) viral replication kinetics, and (iii) virus-induced cell damage differ in tolerant and susceptible BTV-infected cells. Understanding the complex virus-host interactions in disease tolerance can allow us to disentangle the critical balance between protective and damaging host immune responses.
Collapse
Affiliation(s)
- Alexandra Hardy
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Siddharth Bakshi
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Wilhelm Furnon
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Oscar MacLean
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Quan Gu
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Margus Varjak
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Mariana Varela
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Muhamad Afiq Aziz
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Andrew E. Shaw
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Rute Maria Pinto
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Natalia Cameron Ruiz
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Catrina Mullan
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Aislynn E. Taggart
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Ana Da Silva Filipe
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Richard E. Randall
- School of Biology, Centre for Biomolecular Sciences, University of St. Andrews, St. Andrews, Fife, United Kingdom
| | - Sam J. Wilson
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Meredith E. Stewart
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Massimo Palmarini
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| |
Collapse
|
5
|
Premraj A, Aleyas AG, Nautiyal B, Rasool TJ. Novel type-I interferons from the dromedary camel: Molecular identification, prokaryotic expression and functional characterization of camelid interferon-delta. Mol Immunol 2023; 153:212-225. [PMID: 36563641 DOI: 10.1016/j.molimm.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/30/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022]
Abstract
The last two decades have seen the emergence of three highly pathogenic coronaviruses with zoonotic origins, which prompted immediate attention to the underlying cause and prevention of future outbreaks. Intensification of camel husbandry in the Middle East has resulted in increased human-camel interactions, which has led to the spread of potentially zoonotic viruses with human spillover risks like MERS-coronavirus, camelpox virus, etc. Type-I interferons function as the first line of defense against invading viruses and are pivotal for limiting viral replication and immune-mediated pathologies. Seven novel dromedary camel interferon delta genes were identified and cloned. Functional characterization of this novel class of IFNs from the mammalian suborder tylopoda is reported for the first time. The camel interferon-delta proteins resemble the reported mammalian counterparts in sequence similarity, conservation of cysteines, and phylogenetic proximity. Prokaryotically expressed recombinant camel interferon-δ1 induced IFN-stimulated gene expression and also exerted antiviral action against camelpox virus, an endemic zoonotic virus. The pre-treatment of camel kidney cells with recombinant camel IFN-δ1 increased cell survival and reduced camelpox virus in a dose-dependent manner. The identification of novel IFNs from species with zoonotic spillover risk such as camels, and evaluating their antiviral effects in-vitro will play a key role in improving immunotherapies against viruses and expanding the arsenal to combat emerging zoonotic pathogens.
Collapse
Affiliation(s)
- Avinash Premraj
- Camel Biotechnology Center, Presidential Camels & Camel Racing Affairs Centre, Department of the President's Affairs, P.O. Box 17292, Al Ain, United Arab Emirates
| | - Abi George Aleyas
- Camel Biotechnology Center, Presidential Camels & Camel Racing Affairs Centre, Department of the President's Affairs, P.O. Box 17292, Al Ain, United Arab Emirates
| | - Binita Nautiyal
- Camel Biotechnology Center, Presidential Camels & Camel Racing Affairs Centre, Department of the President's Affairs, P.O. Box 17292, Al Ain, United Arab Emirates
| | - Thaha Jamal Rasool
- Camel Biotechnology Center, Presidential Camels & Camel Racing Affairs Centre, Department of the President's Affairs, P.O. Box 17292, Al Ain, United Arab Emirates.
| |
Collapse
|
6
|
Chiang DC, Li Y, Ng SK. The Role of the Z-DNA Binding Domain in Innate Immunity and Stress Granules. Front Immunol 2021; 11:625504. [PMID: 33613567 PMCID: PMC7886975 DOI: 10.3389/fimmu.2020.625504] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/21/2020] [Indexed: 12/18/2022] Open
Abstract
Both DNA and RNA can maintain left-handed double helical Z-conformation under physiological condition, but only when stabilized by Z-DNA binding domain (ZDBD). After initial discovery in RNA editing enzyme ADAR1, ZDBD has also been described in pathogen-sensing proteins ZBP1 and PKZ in host, as well as virulence proteins E3L and ORF112 in viruses. The host-virus antagonism immediately highlights the importance of ZDBD in antiviral innate immunity. Furthermore, Z-RNA binding has been shown to be responsible for the localization of these ZDBD-containing proteins to cytoplasmic stress granules that play central role in coordinating cellular response to stresses. This review sought to consolidate current understanding of Z-RNA sensing in innate immunity and implore possible roles of Z-RNA binding within cytoplasmic stress granules.
Collapse
Affiliation(s)
- De Chen Chiang
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Malaysia
| | - Yan Li
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Siew Kit Ng
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
7
|
Endriß KJ, Meyerholz MM, Fischbach T, Brimmers L, Pfarrer C, Marth CD, Schmicke M. In vitro effects of Type I interferons (IFNτ and IFNα) on bovine hepatocytes cultured with or without Kupffer cells. Reprod Fertil Dev 2021; 33:305-317. [PMID: 33573713 DOI: 10.1071/rd20278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/17/2020] [Indexed: 12/12/2022] Open
Abstract
In cattle, maternal recognition of early pregnancy depends on the effects of the embryonic signal interferon (IFN)-τ. IFN-stimulated genes have been upregulated in the maternal liver during early pregnancy. In this study, primary hepatocyte cell culture models were evaluated for their suitability to test Type I IFN effects invitro. The expression of target genes (interferon-stimulated gene 15 (ISG-15), interferon-induced GTP-binding protein (MX-1), C-X-C motif chemokine 10 (CXCL-10), CXCL-5, insulin-like growth factor 1 (IGF-1), IGF binding protein 2 (IGFBP-2)) was measured using reverse transcription-quantitative polymerase chain reaction in hepatocytes from monoculture or in indirect coculture with Kupffer cells (HKCid) on Days 1, 2, 3 and 4 of culture (n=21 donor cows). Gene expression was also measured on Day 4 after challenging the cultures with recombinant IFNτ, IFNα, progesterone (P4), IFNτ+IFNα or IFNτ+P4 for 6h. A significant increase in the mRNA expression of target genes in hepatocytes was shown in response to stimulation with IFNτ. The Kupffer cells in coculture did not influence the effects of IFNτ in hepatocytes. In conclusion, primary bovine hepatocyte cultures are suitable for stimulation experiments with Type I IFNs and as an extrauterine model for embryo-maternal communication. The proposed endocrine action of IFNτ in the liver may affect maternal metabolism and immune function in the liver.
Collapse
Affiliation(s)
- Kai Josef Endriß
- University of Veterinary Medicine Hanover, Clinic for Cattle, Endocrinology, Bischofsholer Damm 15, 30539 Hanover, Germany
| | - Marie Margarete Meyerholz
- Clinic for Ruminants with Ambulatory and Herd Health Services, Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-University Munich, Sonnenstraße 16, 85764 Oberschleißheim, Germany
| | - Teresa Fischbach
- University of Veterinary Medicine Hanover, Clinic for Cattle, Endocrinology, Bischofsholer Damm 15, 30539 Hanover, Germany
| | - Lutz Brimmers
- University of Veterinary Medicine Hanover, Clinic for Cattle, Endocrinology, Bischofsholer Damm 15, 30539 Hanover, Germany
| | - Christiane Pfarrer
- University of Veterinary Medicine Hanover, Anatomy, Bischofsholer Damm 15, 30539 Hanover, Germany
| | - Christina Deborah Marth
- Melbourne Veterinary School, The University of Melbourne, 250 Princes Highway, Werribee, Vic. 3030, Australia
| | - Marion Schmicke
- University of Veterinary Medicine Hanover, Clinic for Cattle, Endocrinology, Bischofsholer Damm 15, 30539 Hanover, Germany; and Martin-Luther University Halle-Wittenberg, Faculty of Natural Sciences III, Institute of Agricultural and Nutritional Sciences, Animal Health Management, Theodor-Lieser-Straße 11, 06120 Halle, Germany; and Corresponding author.
| |
Collapse
|
8
|
Premzl M. Comparative genomic analysis of eutherian interferon genes. Genomics 2020; 112:4749-4759. [DOI: 10.1016/j.ygeno.2020.08.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/18/2020] [Accepted: 08/25/2020] [Indexed: 01/23/2023]
|
9
|
Premraj A, Aleyas AG, Nautiyal B, Rasool TJ. Camelid type I interferons: Identification and functional characterization of interferon alpha from the dromedary camel (Camelus dromedarius). Mol Immunol 2020; 119:132-143. [PMID: 32014632 PMCID: PMC7112685 DOI: 10.1016/j.molimm.2020.01.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 01/19/2020] [Accepted: 01/24/2020] [Indexed: 11/20/2022]
Abstract
Investigations into the molecular immune response of dromedary camel, a key livestock species of the arid, have been limited due to the lack of species-specific reagents. Here we describe for the first time, the identification and characterization of type I IFNs of dromedary camel, which are the most important cytokines in the innate host immune response against viruses. We cloned camel IFN-α coding sequences and identified a total of eleven subtypes. The canonical IFN-α subtype designated as IFN-α1 contained a 555-bp Open Reading Frame encoding a protein of 184 amino acids. Recombinant IFN-α1 protein was produced in E. coli and purified from inclusion bodies. Recombinant camel IFN-α1 induced the mRNA expression of interferon-stimulated genes (ISGs) in camel kidney cells. The purified protein also showed potent in-vitro antiviral activity against Camelpox Virus in kidney cells. The identified camel IFN-α protein and the subtypes will facilitate a better understanding of the host immune response to viral infections in camel and the development of potential antiviral biologicals for zoonotic diseases for which camel act as a reservoir.
Collapse
Affiliation(s)
- Avinash Premraj
- Camel Biotechnology Center, Presidential Camels and Camel Racing Affairs centre, Department of the President's Affairs, P O Box 17292, Al Ain, United Arab Emirates
| | - Abi George Aleyas
- Camel Biotechnology Center, Presidential Camels and Camel Racing Affairs centre, Department of the President's Affairs, P O Box 17292, Al Ain, United Arab Emirates
| | - Binita Nautiyal
- Camel Biotechnology Center, Presidential Camels and Camel Racing Affairs centre, Department of the President's Affairs, P O Box 17292, Al Ain, United Arab Emirates
| | - Thaha Jamal Rasool
- Camel Biotechnology Center, Presidential Camels and Camel Racing Affairs centre, Department of the President's Affairs, P O Box 17292, Al Ain, United Arab Emirates.
| |
Collapse
|
10
|
Redmond AK, Zou J, Secombes CJ, Macqueen DJ, Dooley H. Discovery of All Three Types in Cartilaginous Fishes Enables Phylogenetic Resolution of the Origins and Evolution of Interferons. Front Immunol 2019; 10:1558. [PMID: 31354716 PMCID: PMC6640115 DOI: 10.3389/fimmu.2019.01558] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/21/2019] [Indexed: 12/31/2022] Open
Abstract
Interferons orchestrate host antiviral responses in jawed vertebrates. They are categorized into three classes; IFN1 and IFN3 are the primary antiviral cytokine lineages, while IFN2 responds to a broader variety of pathogens. The evolutionary relationships within and between these three classes have proven difficult to resolve. Here, we reassess interferon evolution, considering key phylogenetic pitfalls including taxon sampling, alignment quality, model adequacy, and outgroup choice. We reveal that cartilaginous fishes, and hence the jawed vertebrate ancestor, possess(ed) orthologs of all three interferon classes. We show that IFN3 groups sister to IFN1, resolve the origins of the human IFN3 lineages, and find that intronless IFN3s emerged at least three times. IFN2 genes are highly conserved, except for IFN-γ-rel, which we confirm resulted from a teleost-specific duplication. Our analyses show that IFN1 phylogeny is highly sensitive to phylogenetic error. By accounting for this, we describe a new backbone IFN1 phylogeny that implies several IFN1 genes existed in the jawed vertebrate ancestor. One of these is represented by the intronless IFN1s of tetrapods, including mammalian-like repertoires of reptile IFN1s and a subset of amphibian IFN1s, in addition to newly-identified intron-containing shark IFN1 genes. IFN-f, previously only found in teleosts, likely represents another ancestral jawed vertebrate IFN1 family member, suggesting the current classification of fish IFN1s into two groups based on the number of cysteines may need revision. The providence of the remaining fish IFN1s and the coelacanth IFN1s proved difficult to resolve, but they may also be ancestral jawed vertebrate IFN1 lineages. Finally, a large group of amphibian-specific IFN1s falls sister to all other IFN1s and was likely also present in the jawed vertebrate ancestor. Our results verify that intronless IFN1s have evolved multiple times in amphibians and indicate that no one-to-one orthology exists between mammal and reptile IFN1s. Our data also imply that diversification of the multiple IFN1s present in the jawed vertebrate ancestor has occurred through a rapid birth-death process, consistent with functional maintenance over a 450-million-year host-pathogen arms race. In summary, this study reveals a new model of interferon evolution important to our understanding of jawed vertebrate antiviral immunity.
Collapse
Affiliation(s)
- Anthony K Redmond
- School of Biological Sciences, University of Aberdeen, Aberdeen, United Kingdom.,Centre for Genome-Enabled Biology and Medicine, University of Aberdeen, Aberdeen, United Kingdom.,Smurfit Institute of Genetics, Trinity College Dublin, University of Dublin, Dublin, Ireland
| | - Jun Zou
- School of Biological Sciences, University of Aberdeen, Aberdeen, United Kingdom.,Scottish Fish Immunology Research Centre, Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, United Kingdom.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
| | - Christopher J Secombes
- School of Biological Sciences, University of Aberdeen, Aberdeen, United Kingdom.,Scottish Fish Immunology Research Centre, Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Daniel J Macqueen
- School of Biological Sciences, University of Aberdeen, Aberdeen, United Kingdom.,The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, United Kingdom
| | - Helen Dooley
- School of Biological Sciences, University of Aberdeen, Aberdeen, United Kingdom.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States.,Institute of Marine and Environmental Technology, Baltimore, MD, United States
| |
Collapse
|
11
|
Heaton SM. Harnessing host-virus evolution in antiviral therapy and immunotherapy. Clin Transl Immunology 2019; 8:e1067. [PMID: 31312450 PMCID: PMC6613463 DOI: 10.1002/cti2.1067] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/07/2019] [Accepted: 06/09/2019] [Indexed: 02/06/2023] Open
Abstract
Pathogen resistance and development costs are major challenges in current approaches to antiviral therapy. The high error rate of RNA synthesis and reverse‐transcription confers genome plasticity, enabling the remarkable adaptability of RNA viruses to antiviral intervention. However, this property is coupled to fundamental constraints including limits on the size of information available to manipulate complex hosts into supporting viral replication. Accordingly, RNA viruses employ various means to extract maximum utility from their informationally limited genomes that, correspondingly, may be leveraged for effective host‐oriented therapies. Host‐oriented approaches are becoming increasingly feasible because of increased availability of bioactive compounds and recent advances in immunotherapy and precision medicine, particularly genome editing, targeted delivery methods and RNAi. In turn, one driving force behind these innovations is the increasingly detailed understanding of evolutionarily diverse host–virus interactions, which is the key concern of an emerging field, neo‐virology. This review examines biotechnological solutions to disease and other sustainability issues of our time that leverage the properties of RNA and DNA viruses as developed through co‐evolution with their hosts.
Collapse
Affiliation(s)
- Steven M Heaton
- Department of Biochemistry & Molecular Biology Monash University Clayton VIC Australia
| |
Collapse
|
12
|
Analysis of Bovine Interferon-tau Gene subtypes Expression in the Trophoblast and Non-trophoblast cells. JOURNAL OF ANIMAL REPRODUCTION AND BIOTECHNOLOGY 2018. [DOI: 10.12750/jet.2018.33.4.195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
13
|
Bordignon V, Cavallo I, D'Agosto G, Trento E, Pontone M, Abril E, Di Domenico EG, Ensoli F. Nucleic Acid Sensing Perturbation: How Aberrant Recognition of Self-Nucleic Acids May Contribute to Autoimmune and Autoinflammatory Diseases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 344:117-137. [PMID: 30798986 DOI: 10.1016/bs.ircmb.2018.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Bacteria and mammalian cells have developed sophisticated sensing mechanisms to detect and eliminate foreign genetic material or to restrict its expression and replication. Progress has been made in the understanding of these mechanisms, which keep foreign or unwanted nucleic acids in check. The complex of mechanisms involved in RNA and DNA sensing is part of a system which is now appreciated as "immune sensing of nucleic acids" or better "nucleic acid immunity." Nucleic acids, which are critical components for inheriting genetic information in all species, including pathogens, are key structures recognized by the innate immune system. However, while nucleic acid recognition is required for host defense against pathogens, there is a potential risk of self-nucleic acids recognition. In fact, besides its essential contribution to antiviral or microbial defense and restriction of endogenous retro elements, deregulation of nucleic acid immunity can also lead to human diseases due to erroneous detection and response to self-nucleic acids, causing sterile inflammation and autoimmunity. In this review we will discuss the roles of nucleic acid receptors in guarding against pathogen invasion, and how the microbial environment could interfere or influence immune sensing in discriminating between self and non-self and how this may contribute to autoimmunity or inflammatory diseases.
Collapse
Affiliation(s)
- Valentina Bordignon
- Clinical Pathology and Microbiology, San Gallicano Dermatological Institute IRCCS, Rome, Italy.
| | - Ilaria Cavallo
- Clinical Pathology and Microbiology, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Giovanna D'Agosto
- Clinical Pathology and Microbiology, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Elisabetta Trento
- Clinical Pathology and Microbiology, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Martina Pontone
- Clinical Pathology and Microbiology, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Elva Abril
- Clinical Pathology and Microbiology, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Enea Gino Di Domenico
- Clinical Pathology and Microbiology, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Fabrizio Ensoli
- Clinical Pathology and Microbiology, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| |
Collapse
|
14
|
Ealy AD, Wooldridge LK. The evolution of interferon-tau. Reproduction 2018; 154:F1-F10. [PMID: 28982935 DOI: 10.1530/rep-17-0292] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 06/20/2017] [Accepted: 07/10/2017] [Indexed: 01/24/2023]
Abstract
Thirty years ago, a novel type I interferon (IFN) was identified by molecular cloning of cDNA libraries constructed from RNA extracted from ovine and bovine pre-implantation embryos. This protein was eventually designated as IFN-tau (IFNT) to highlight its trophoblast-dependent expression. IFNT function is not immune related. Instead, it interacts with the maternal system to initiate the establishment and maintenance of pregnancy. This activity is indispensable for the continuation of pregnancy. Our review will describe how IFNT evolved from other type I IFNs to function in this new capacity. IFNT genes have only been identified in pecoran ruminants within the Artiodactyla order (e.g. cattle, sheep, goats, deer, antelope, giraffe). The ancestral IFNT gene emerged approximately 36 million years ago most likely from rearrangement and/or insertion events that combined an ancestral IFN-omega (IFNW) gene with a trophoblast-specifying promoter/enhancer. Since then, IFNT genes have duplicated, likely through conversion events, and mutations have allowed them to adapt to their new function in concert with the emergence of different species. Multiple IFNT polymorphisms have been identified in cattle, sheep and goats. These genes and gene alleles encode proteins that do not display identical antiviral, antiproliferative and antiluteolytic activities. The need for multiple IFNT genes, numerous alleles and distinct activities remains debatable, but the consensus is that this complexity in IFNT expression and biological activity must be needed to provide the best opportunity for pregnancy to be recognized by the maternal system so that gestation may continue.
Collapse
Affiliation(s)
- Alan D Ealy
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - Lydia K Wooldridge
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
15
|
Morphine-potentiated cognitive deficits correlate to suppressed hippocampal iNOS RNA expression and an absent type 1 interferon response in LP-BM5 murine AIDS. J Neuroimmunol 2018. [PMID: 29526406 DOI: 10.1016/j.jneuroim.2018.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Opioid use accelerates neurocognitive impairment in HIV/AIDS patients. We assessed the effect of chronic morphine treatment and LP-BM5/murine AIDS (MAIDS) infection on cognition, cytokine production, and type 1 interferon (IFN) expression in the murine CNS. Morphine treatment decreased expression of pro-inflammatory factors (CCL5, iNOS) and reduced cognitive performance in LP-BM5-infected mice, correlating to increased hippocampal viral load and a blunted type 1 IFN response. In the striatum, morphine reduced viral load while increasing IFN-α RNA expression. Our results suggest that differentially regulated type 1 IFN responses may contribute to distinct regional outcomes in the hippocampus and striatum in LP-BM5/MAIDS.
Collapse
|
16
|
Gene Knockout Shows That PML (TRIM19) Does Not Restrict the Early Stages of HIV-1 Infection in Human Cell Lines. mSphere 2017; 2:mSphere00233-17. [PMID: 28656178 PMCID: PMC5480037 DOI: 10.1128/mspheredirect.00233-17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 05/26/2017] [Indexed: 01/12/2023] Open
Abstract
PML is involved in innate immune mechanisms against both DNA and RNA viruses. Although the mechanism by which PML inhibits highly divergent viruses is unclear, it was recently found that it can increase the transcription of interferon-stimulated genes (ISGs). However, whether human PML inhibits HIV-1 has been debated. Here we provide unambiguous, knockout-based evidence that PML does not restrict the early postentry stages of HIV-1 infection in a variety of human cell types and does not participate in the inhibition of HIV-1 by IFN-I. Although this study does not exclude the possibility of other mechanisms by which PML may interfere with HIV-1, we nonetheless demonstrate that PML does not generally act as an HIV-1 restriction factor in human cells and that its presence is not required for IFN-I to stimulate the expression of anti-HIV-1 genes. These results contribute to uncovering the landscape of HIV-1 inhibition by ISGs in human cells. The PML (promyelocytic leukemia) protein is a member of the TRIM family, a large group of proteins that show high diversity in functions but possess a common tripartite motif giving the family its name. We and others recently reported that both murine PML (mPML) and human PML (hPML) strongly restrict the early stages of infection by HIV-1 and other lentiviruses when expressed in mouse embryonic fibroblasts (MEFs). This restriction activity was found to contribute to the type I interferon (IFN-I)-mediated inhibition of HIV-1 in MEFs. Additionally, PML caused transcriptional repression of the HIV-1 promoter in MEFs. In contrast, the modulation of the early stages of HIV-1 infection of human cells by PML has been investigated by RNA interference, with unclear results. In order to conclusively determine whether PML restricts HIV-1 or not in human cells, we used the clustered regularly interspaced short palindromic repeat with Cas9 (CRISPR-Cas9) system to knock out its gene in epithelial, lymphoid, and monocytic human cell lines. Infection challenges showed that PML knockout had no effect on the permissiveness of these cells to HIV-1 infection. IFN-I treatments inhibited HIV-1 equally whether PML was expressed or not. Overexpression of individual hPML isoforms, or of mPML, in a human T cell line did not restrict HIV-1. The presence of PML was not required for the restriction of nonhuman retroviruses by TRIM5α (another human TRIM protein), and TRIM5α was inhibited by arsenic trioxide through a PML-independent mechanism. We conclude that PML is not a restriction factor for HIV-1 in human cell lines representing diverse lineages. IMPORTANCE PML is involved in innate immune mechanisms against both DNA and RNA viruses. Although the mechanism by which PML inhibits highly divergent viruses is unclear, it was recently found that it can increase the transcription of interferon-stimulated genes (ISGs). However, whether human PML inhibits HIV-1 has been debated. Here we provide unambiguous, knockout-based evidence that PML does not restrict the early postentry stages of HIV-1 infection in a variety of human cell types and does not participate in the inhibition of HIV-1 by IFN-I. Although this study does not exclude the possibility of other mechanisms by which PML may interfere with HIV-1, we nonetheless demonstrate that PML does not generally act as an HIV-1 restriction factor in human cells and that its presence is not required for IFN-I to stimulate the expression of anti-HIV-1 genes. These results contribute to uncovering the landscape of HIV-1 inhibition by ISGs in human cells.
Collapse
|
17
|
Takahashi T, Sakumoto R, Hayashi KG, Hosoe M, Shirai J, Hashizume K. Generation of recombinant bovine interferon tau in the human embryonic kidney cell line and its biological activity. Anim Sci J 2017; 88:1498-1505. [PMID: 28558419 DOI: 10.1111/asj.12820] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 01/31/2017] [Indexed: 01/15/2023]
Abstract
The objective of this study was to generate recombinant bovine interferon tau (rbIFNT) in mammalian hosts. The complementary DNA encoding bovine IFNT2 was cloned for the construction of pRcRSV-bIFNT2 expression vector. The expression vector was transfected to 293 cells. Transfected cells harboring expression vector were selected with G418. Highly expressing clonal line was adapted to serum-free suspension culture in a spinner flask. The recombinant protein had 24 kDa apparent molecular mass, suggesting being expressed as a glycoprotein, and was purified from serum-free conditioned medium by the combination of Diethylaminoethanol Sepharose ion exchange and Sephacryl S-200 HR gel filtration. A total of 7.3 mg rbIFNT was obtained from 13.5 L conditioned medium. Generated rbIFNT was biologically active in terms of antiviral activity measured by the plaque inhibition assay with Madin-Darby bovine kidney cells and the vesicular stomatitis virus. The recombinant protein was also utilized for immunization to raise antibodies in the rabbit. The generated antibody was capable of use in both Western blotting and the binding assay. The results in the present study suggest that a certain amount of rbIFNT is raised in mammalian hosts by using conventional plasmid vector and its antibody provides useful tools for studies in the biology of bovine IFNT.
Collapse
Affiliation(s)
- Toru Takahashi
- Faculty of Agriculture, Iwate University, Morioka, Iwate, Japan
| | - Ryosuke Sakumoto
- Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| | - Ken-Go Hayashi
- Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| | - Misa Hosoe
- Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| | - Junsuke Shirai
- Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
| | | |
Collapse
|
18
|
An D, Guo Y, Bao J, Luo X, Liu Y, Ma B, Gao M, Wang J. Molecular characterization and biological activity of bovine interferon-omega3. Res Vet Sci 2017; 115:125-131. [PMID: 28254416 PMCID: PMC7127041 DOI: 10.1016/j.rvsc.2017.01.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 12/29/2016] [Accepted: 01/26/2017] [Indexed: 02/07/2023]
Abstract
Bovine interferon-omega3 (BoIFN-ω3) gene was amplified from bovine liver genomic DNA, which encodes a 195-amino acid protein containing a 23-amino acid signal peptide. Analysis of the molecular characteristics revealed that BoIFN-ω3 evolving from IFN-ω, contained four cysteine residues and five alpha helices, showing that BoIFN-ω3 presented the typical molecular characteristics of type I interferon. BoIFN-ω3 exhibited antiviral and antiproliferative activities, which exerted a protective effect against VSV in several mammalian cell lines, as well as against BEV, IBRV, and BVDV in MDBK cell. Moreover, BoIFN-ω3 was shown to be highly sensitive to trypsin, but remaining stable despite changes in pH and temperature. Additionally, BoIFN-ω3 induced the transcription of Mx1, ISG15, and ISG56 genes, as well as the expression of Mx1 protein in a time-dependent manner. These findings will be useful to further study BoIFN-ω in host's defence against infectious diseases, particularly viral infections. Furthermore, results will facilitate further research on the bovine interferon family. BoIFN-ω3 presents antiviral activity on several mammalian cell lines and protective effect against VSV, BEV, IBRV, and BVDV. BoIFN-ω3 exhibits antiproliferative activity and insensitivity to pH and temperature. BoIFN-ω3 can activate the transcription of ISGs gene, as well as the expression of Mx1 in a time-dependent manner.
Collapse
Affiliation(s)
- Dong An
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Yongli Guo
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Jun Bao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China; National Food Safety and Nutrition Collaborative Innovation Center, Wuxi, Jiangsu 214122, PR China
| | - Xiuxin Luo
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Ying Liu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Bo Ma
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Mingchun Gao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China.
| | - Junwei Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China; National Food Safety and Nutrition Collaborative Innovation Center, Wuxi, Jiangsu 214122, PR China.
| |
Collapse
|
19
|
Usharani J, Park SY, Cho EJ, Kim C, Ko YJ, Tark D, Kim SM, Park JH, Lee KN, Lee MH, Lee HS. Antiviral activity of ovine interferon tau 4 against foot-and-mouth disease virus. Antiviral Res 2017; 143:134-141. [PMID: 28137623 DOI: 10.1016/j.antiviral.2017.01.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 01/25/2017] [Indexed: 11/15/2022]
Abstract
Foot-and-mouth disease (FMD) is an economically important disease in most parts of the world and new therapeutic agents are needed to protect the animals before vaccination can trigger the host immune response. Although several interferons have been used for their antiviral activities against Foot-and-mouth disease virus (FMDV), ovine interferon tau 4 (OvIFN-τ4), with a broad-spectrum of action, cross-species antiviral activity, and lower incidence of toxicity in comparison to other type І interferons, has not yet been evaluated for this indication. This is the first study to evaluate the antiviral activity of OvIFN-τ4 against various strains of FMDV. The effective anti-cytopathic concentration of OvIFN-τ4 and its effectiveness pre- and post-infection with FMDV were tested in vitro in LFBK cells. In vivo activity of OvIFN-τ4 was then confirmed in a mouse model of infection. OvIFN-τ4 at a concentration of 500 ng, protected mice until 5days post-FMDV challenge and provided 90% protection for 10 days following FMDV challenge. These results suggest that OvIFN-τ4 could be used as an alternative to other interferons or antiviral agents at the time of FMD outbreak.
Collapse
Affiliation(s)
- Jayaramaiah Usharani
- Foot-and-mouth Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, Republic of Korea
| | - Sun Young Park
- Foot-and-mouth Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, Republic of Korea
| | - Eun-Ju Cho
- Foot-and-mouth Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, Republic of Korea
| | - Chungsu Kim
- Foot-and-mouth Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, Republic of Korea
| | - Young-Joon Ko
- Foot-and-mouth Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, Republic of Korea
| | - Dongseob Tark
- Korea Zoonosis Research Institute, Chonbuk National University, Ma-dong, Iksan-si, Jeollabuk-do, Republic of Korea
| | - Su-Mi Kim
- Foot-and-mouth Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, Republic of Korea
| | - Jong-Hyeon Park
- Foot-and-mouth Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, Republic of Korea
| | - Kwang-Nyeong Lee
- Foot-and-mouth Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, Republic of Korea
| | - Myoung-Heon Lee
- Foot-and-mouth Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, Republic of Korea
| | - Hyang-Sim Lee
- Foot-and-mouth Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, Republic of Korea.
| |
Collapse
|
20
|
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
|
21
|
Contraction of the type I IFN locus and unusual constitutive expression of IFN-α in bats. Proc Natl Acad Sci U S A 2016; 113:2696-701. [PMID: 26903655 DOI: 10.1073/pnas.1518240113] [Citation(s) in RCA: 240] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Bats harbor many emerging and reemerging viruses, several of which are highly pathogenic in other mammals but cause no clinical signs of disease in bats. To determine the role of interferons (IFNs) in the ability of bats to coexist with viruses, we sequenced the type I IFN locus of the Australian black flying fox, Pteropus alecto, providing what is, to our knowledge, the first gene map of the IFN region of any bat species. Our results reveal a highly contracted type I IFN family consisting of only 10 IFNs, including three functional IFN-α loci. Furthermore, the three IFN-α genes are constitutively expressed in unstimulated bat tissues and cells and their expression is unaffected by viral infection. Constitutively expressed IFN-α results in the induction of a subset of IFN-stimulated genes associated with antiviral activity and resistance to DNA damage, providing evidence for a unique IFN system that may be linked to the ability of bats to coexist with viruses.
Collapse
|
22
|
Saugandhika S, Sharma V, Malik H, Mohapatra SK, Bondre VP, Kumar S, Mohanty AK, Malakar D. Molecular characterization of IFN-T expressed in buffalo embryonic trophoblasts and expression of recombinant BuIFN-T1a2 and BuIFN-T8 isoforms in E. coli. Protein Expr Purif 2016; 122:8-14. [PMID: 26876002 DOI: 10.1016/j.pep.2016.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 01/15/2016] [Accepted: 02/09/2016] [Indexed: 10/22/2022]
Abstract
Interferon tau (IFN-T) acts as a signaling molecule for maternal recognition of pregnancy (MRP) in ruminants. Aim of the present study was to identify various Buffalo Interferon tau (BuIFN-T) transcripts in buffalo trophoblast, phylogenetic comparison of these sequences with known mRNA sequences of buffalo, bovine, caprine and ovine and to express and purify the recombinant BuIFN-T (rBuIFN-T) isoforms. Following RNA extraction from trophectodermal cells, RT-PCR was performed using Ifn-t gene specific primers. 13 distinct cDNA variants encoding eight different BuIFN-T proteins were identified. BuIFN-T1a2 and BuIFN-T8 were expressed in prokaryotic expression system at 37 °C, 25 °C and 16 °C with 1 mM IPTG for 12 h and the recombinant proteins expressed at 16 °C were partially purified by Immobilised Metal Affinity Chromatography (IMAC). BuIFN-T isoforms have greater nucleotide and amino acid homology with caprine (98-100%, 96-100%), ovine (94-97%, 90-95%) and bovine (89.6-90.6%, 82-86%). These novel BuIFN-T isoforms contained pronounced nucleotide and amino acid sequence identity with one another (99.1-99.8%, 98-99%) but moderate sequence identity with previously identified buffalo IFN-T (90-92%, 82-86%). Solubility of expressed recombinant isoforms (rBuIFN-T1a2 and rBuIFN-T8) was highest at 16 °C. In conclusion, 13 distinct Ifn-t gene variants exist in trophectoderm of in vitro developed buffalo blastocysts that encode eight different proteins. rBuIFN-T1a2 and rBuIFN-T8 were successfully expressed in soluble form in Escherichia coli expression system at 16 °C with 1 mM IPTG and the resulting recombinant proteins were partially purified by IMAC.
Collapse
Affiliation(s)
- Shrabani Saugandhika
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana 132001, India
| | - Vishal Sharma
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana 132001, India
| | - Hrudananda Malik
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana 132001, India
| | - Sushil Kumar Mohapatra
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana 132001, India
| | | | - Sudarshan Kumar
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana 132001, India
| | - Ashok Kumar Mohanty
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana 132001, India
| | - Dhruba Malakar
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana 132001, India.
| |
Collapse
|
23
|
Abstract
The seven human sirtuins are a family of ubiquitously expressed and evolutionarily conserved NAD+-dependent deacylases/mono-ADP ribosyltransferases that regulate numerous cellular and organismal functions, including metabolism, cell cycle, and longevity. Here, we report the discovery that all seven sirtuins have broad-range antiviral properties. We demonstrate that small interfering RNA (siRNA)-mediated knockdown of individual sirtuins and drug-mediated inhibition of sirtuin enzymatic activity increase the production of virus progeny in infected human cells. This impact on virus growth is observed for both DNA and RNA viruses. Importantly, sirtuin-activating drugs inhibit the replication of diverse viruses, as we demonstrate for human cytomegalovirus, a slowly replicating DNA virus, and influenza A (H1N1) virus, an RNA virus that multiplies rapidly. Furthermore, sirtuin defense functions are evolutionarily conserved, since CobB, the sirtuin homologue in Escherichia coli, protects against bacteriophages. Altogether, our findings establish sirtuins as broad-spectrum and evolutionarily conserved components of the immune defense system, providing a framework for elucidating a new set of host cell defense mechanisms and developing sirtuin modulators with antiviral activity. We live in a sea of viruses, some of which are human pathogens. These pathogenic viruses exhibit numerous differences: DNA or RNA genomes, enveloped or naked virions, nuclear or cytoplasmic replication, diverse disease symptoms, etc. Most antiviral drugs target specific viral proteins. Consequently, they often work for only one virus, and their efficacy can be compromised by the rapid evolution of resistant variants. There is a need for the identification of host proteins with broad-spectrum antiviral functions, which provide effective targets for therapeutic treatments that limit the evolution of viral resistance. Here, we report that sirtuins present such an opportunity for the development of broad-spectrum antiviral treatments, since our findings highlight these enzymes as ancient defense factors that protect against a variety of viral pathogens.
Collapse
|
24
|
Shao J, Cao C, Bao J, Liu H, Peng T, Gao M, Wang J. Characterization of bovine interferon α1: expression in yeast Pichia pastoris, biological activities, and physicochemical characteristics. J Interferon Cytokine Res 2014; 35:168-75. [PMID: 25343404 DOI: 10.1089/jir.2013.0139] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A bovine interferon α (BoIFNα) gene that included signal sequence was amplified from bovine liver genomic DNA. The gene was named BoIFN-α1 according to the position at which the encoded gene of the bovine IFN was located in the bovine genome. The sequence included a 23-amino-acid signal peptide and a 166-amino-acid mature peptide. The structural characteristics and phylogenetic relationships of the BoIFN-α1 gene were analyzed. A recombinant mature BoIFN-α1 (rBoIFN-α1) was expressed in the yeast Pichia pastoris. Physicochemical characteristics and antiviral activity were determined in vitro. Recombinant BoIFN-α1 was found to be highly sensitive to trypsin and stable at pH 2.0 or 65°C. It also exhibited antiviral activity, which was neutralized by a rabbit anti-rBoIFNα polyclonal antibody. This study revealed that rBoIFN-α1 has the typical characteristics of IFNα and can be used for both research and industrial application.
Collapse
Affiliation(s)
- Jianwei Shao
- 1 College of Veterinary Medicine, Northeast Agricultural University , Harbin, China
| | | | | | | | | | | | | |
Collapse
|
25
|
Haugh KA, Shalginskikh N, Nogusa S, Skalka AM, Katz RA, Balachandran S. The interferon-inducible antiviral protein Daxx is not essential for interferon-mediated protection against avian sarcoma virus. Virol J 2014; 11:100. [PMID: 24884573 PMCID: PMC4049388 DOI: 10.1186/1743-422x-11-100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 05/23/2014] [Indexed: 01/19/2023] Open
Abstract
Background The antiviral protein Daxx acts as a restriction factor of avian sarcoma virus (ASV; Retroviridae) in mammalian cells by promoting epigenetic silencing of integrated proviral DNA. Although Daxx is encoded by a type I (α/β) interferon-stimulated gene, the requirement for Daxx in the interferon anti-retroviral response has not been elucidated. In this report, we describe the results of experiments designed to investigate the role of Daxx in the type I interferon-induced anti-ASV response. Findings Using an ASV reporter system, we show that type I interferons are potent inhibitors of ASV replication. We demonstrate that, while Daxx is necessary to silence ASV gene expression in the absence of interferons, type I interferons are fully-capable of inducing an antiviral state in the absence of Daxx. Conclusions These results provide evidence that Daxx is not essential for the anti-ASV interferon response in mammalian cells, and that interferons deploy multiple, redundant antiviral mechanisms to protect cells from ASV.
Collapse
Affiliation(s)
| | | | | | | | - Richard A Katz
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Room 422 Reimann Building, 333 Cottman Ave,, 19111 Philadelphia, PA, USA.
| | | |
Collapse
|
26
|
Harari D, Abramovich R, Zozulya A, Smith P, Pouly S, Köster M, Hauser H, Schreiber G. Bridging the species divide: transgenic mice humanized for type-I interferon response. PLoS One 2014; 9:e84259. [PMID: 24416207 PMCID: PMC3887009 DOI: 10.1371/journal.pone.0084259] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 11/12/2013] [Indexed: 01/23/2023] Open
Abstract
We have generated transgenic mice that harbor humanized type I interferon receptors (IFNARs) enabling the study of type I human interferons (Hu-IFN-Is) in mice. These “HyBNAR” (Hybrid IFNAR) mice encode transgenic variants of IFNAR1 and IFNAR2 with the human extracellular domains being fused to transmembrane and cytoplasmic segments of mouse sequence. B16F1 mouse melanoma cells harboring the HyBNAR construct specifically bound Hu-IFN-Is and were rendered sensitive to Hu-IFN-I stimulated anti-proliferation, STAT1 activation and activation of a prototypical IFN-I response gene (MX2). HyBNAR mice were crossed with a transgenic strain expressing the luciferase reporter gene under the control of the IFN-responsive MX2 promoter (MX2-Luciferase). Both the HyBNAR and HyBNAR/MX2-Luciferase mice were responsive to all Hu-IFN-Is tested, inclusive of IFNα2A, IFNβ, and a human superagonist termed YNSα8. The mice displayed dose-dependent pharmacodynamic responses to Hu-IFN-I injection, as assessed by measuring the expression of IFN-responsive genes. Our studies also demonstrated a weak activation of endogenous mouse interferon response, especially after high dose administration of Hu-IFNs. In sharp contrast to data published for humans, our pharmacodynamic readouts demonstrate a very short-lived IFN-I response in mice, which is not enhanced by sub-cutaneous (SC) injections in comparison to other administration routes. With algometric differences between humans and mice taken into account, the HyBNAR mice provides a convenient non-primate pre-clinical model to advance the study of human IFN-Is.
Collapse
Affiliation(s)
- Daniel Harari
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot, Israel
- * E-mail: daniel@ (DH); (GS)
| | - Renne Abramovich
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot, Israel
| | - Alla Zozulya
- MS Platform, Merck-Serono, (a division of Merck KGaA), Geneva, Switzerland
| | - Paul Smith
- MS Platform, Merck-Serono, (a division of Merck KGaA), Geneva, Switzerland
| | - Sandrine Pouly
- MS Platform, Merck-Serono, (a division of Merck KGaA), Geneva, Switzerland
| | - Mario Köster
- Helmholtz Centre for Infection Research, Dept. Gene Regulation and Differentiation, Braunschweig, Germany
| | - Hansjörg Hauser
- Helmholtz Centre for Infection Research, Dept. Gene Regulation and Differentiation, Braunschweig, Germany
| | - Gideon Schreiber
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot, Israel
- * E-mail: daniel@ (DH); (GS)
| |
Collapse
|
27
|
Molecular cloning and sequencing analysis of the interferon β from Coturnix. Cent Eur J Immunol 2014; 39:25-9. [PMID: 26155095 PMCID: PMC4439982 DOI: 10.5114/ceji.2014.42118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 02/03/2014] [Indexed: 12/02/2022] Open
Abstract
One pair of primers was designed according to Gallus and Meleagris gallopavo interferon β (IFN-β) sequences published in GenBank. The primers and RNA extraction from the spleen of Coturnix were used to amplify Coturnix IFN-β cDNA by real-time polymerase chain reaction (RT-PCR). The product was cloned into pEasy-T1 vector. Evaluating recombinant plasmid by PCR and restriction enzyme digestion. Sequence the cloning sequences, comparing the sequencing results by NCBI. We successfully got a Coturnix IFN-β partial sequence. The sequence was subtyped and put to homologous analysis. The results suggested the homology of IFN-β gene of Coturnix and gene of Coturnix and chicken (88.7%), the homology of IFN-β gene of Coturnix and chicken (88.7%), the homology of IFN-β gene of Coturnix and Anas platyrhynchos (72.5%), the homology of IFN-β sequence registered in GenBank. The analysis of the genetic tree showed that the relationship of Coturnix and chicken IFN-β had a high homology. It can be seen that in this study we successfully got a partial sequence of IFN-β of quail.
Collapse
|
28
|
Vitour D, Doceul V, Ruscanu S, Chauveau E, Schwartz-Cornil I, Zientara S. Induction and control of the type I interferon pathway by Bluetongue virus. Virus Res 2013; 182:59-70. [PMID: 24211608 PMCID: PMC7114367 DOI: 10.1016/j.virusres.2013.10.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 10/24/2013] [Accepted: 10/24/2013] [Indexed: 12/12/2022]
Abstract
A general review describing the current knowledge on the type I IFN pathway. Description of several mechanisms evolved by viruses to counteract this antiviral response. An up-to-date review on the interaction of BTV and the type I IFN pathway in vivo and in vitro. Description of the cellular sensors involved in the induction of IFN-α/β synthesis upon BTV infection in haematopoietic and non-haematopoietic cells. Description of the strategies evolved by BTV to counteract this cellular antiviral response.
The innate immune response is the first line of defence against viruses, involving the production of type I IFN (IFN-α/β) and other pro-inflammatory cytokines that control the infection. It also shapes the adaptive immune response generated by both T and B cells. Production of type I IFN occurs both in vivo and in vitro in response to Bluetongue virus (BTV), an arthropod-borne virus. However, the mechanisms responsible for the production of IFN-β in response to BTV remained unknown until recently and are still not completely understood. In this review, we describe the recent advances in the identification of cellular sensors and signalling pathways involved in this process. The RNA helicases retinoic acid-inducible gene-I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) were shown to be involved in the expression of IFN-β as well as in the control of BTV infection in non-haematopoietic cells. In contrast, induction of IFN-α/β synthesis in sheep primary plasmacytoid dendritic cells (pDCs) required the MyD88 adaptor independently of the Toll-like receptor 7 (TLR7), as well as the kinases dsRNA-activated protein kinase (PKR) and stress-activated protein kinase (SAPK)/Jun N-terminal protein kinase (JNK). As type I IFN is essential for the establishment of an antiviral cellular response, most of viruses have elaborated counteracting mechanisms to hinder its action. This review also addresses the ability of BTV to interfere with IFN-β synthesis and the recent findings describing the non-structural viral protein NS3 as a powerful antagonist of the host cellular response.
Collapse
Affiliation(s)
- Damien Vitour
- UMR1161 ANSES-INRA-ENVA, 23 Avenue du Général de Gaulle, 94704 Maisons-Alfort, France.
| | - Virginie Doceul
- UMR1161 ANSES-INRA-ENVA, 23 Avenue du Général de Gaulle, 94704 Maisons-Alfort, France.
| | - Suzana Ruscanu
- Virologie et Immunologie Moléculaires, UR892 INRA, Jouy-en-Josas, France.
| | - Emilie Chauveau
- UMR1161 ANSES-INRA-ENVA, 23 Avenue du Général de Gaulle, 94704 Maisons-Alfort, France.
| | | | - Stéphan Zientara
- UMR1161 ANSES-INRA-ENVA, 23 Avenue du Général de Gaulle, 94704 Maisons-Alfort, France.
| |
Collapse
|
29
|
Purification and biological characterization of soluble, recombinant mouse IFNβ expressed in insect cells. Protein Expr Purif 2013; 94:7-14. [PMID: 24211771 DOI: 10.1016/j.pep.2013.10.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 10/28/2013] [Accepted: 10/29/2013] [Indexed: 11/20/2022]
Abstract
Interferon β (IFNβ) is a member of the type I interferon family of cytokines widely recognised for their anti-viral, anti-proliferative and immunomodulatory properties. Recombinant, biologically active forms of this cytokine are used clinically for the treatment of multiple sclerosis and in laboratories to study the role of this cytokine in health and disease. Established methods for expression of IFNβ utilise either bacterial systems from which the insoluble recombinant proteins must be refolded, or mammalian expression systems in which large volumes of cell culture are required for recovery of acceptable yields. Utilising the baculovirus expression system and Trichoplusia ni (Cabbage Looper) BTI-TN-5B1-4 cell line, we report a reproducible method for production and purification of milligram/litre quantities of biologically active murine IFNβ. Due to the design of our construct and the eukaryotic nature of insect cells, the resulting soluble protein is secreted allowing purification of the Histidine-tagged natively-folded protein from the culture supernatant. The IFNβ purification method described is a two-step process employing immobilised metal-ion affinity chromatography (IMAC) and reverse-phase high performance liquid chromatography (RP-HPLC) that results in production of significantly more purified IFNβ than any other reported eukaryotic-based expression system. Recombinant murine IFNβ produced by this method was natively folded and demonstrated hallmark type I interferon biological effects including antiviral and anti-proliferative activities, and induced genes characteristic of IFNβ activity in vivo. Recombinant IFNβ also had specific activity levels exceeding that of the commercially available equivalent. Together, our findings provide a method for production of highly pure, biologically active murine IFNβ.
Collapse
|
30
|
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
|
31
|
Hu Z, Wu X, Ge J, Wang X. Inhibition of virus replication and induction of human tetherin gene expression by equine IFN-α1. Vet Immunol Immunopathol 2013; 156:107-13. [PMID: 24144682 DOI: 10.1016/j.vetimm.2013.09.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 08/21/2013] [Accepted: 09/16/2013] [Indexed: 11/30/2022]
Abstract
Type I interferons (IFNs) play important roles in the defense of host cells against viral infection by inducing the expression of a diverse range of antiviral factors. IFNs from different animals likely share similar features with human IFNs, and some of them have cross-species activities. Equine IFN-α was proved effective in both equine and human cells. However, the previous studies mostly focused on the inhibition of virus induced cytopathic effects. In this study, we used virus-specific assays to demonstrate the antiviral activities of equine IFN-α1 in both equine and human cells. Equine IFN-α1 inhibited the expression of viral structural proteins and the production of virions of equine infectious anemia virus (EIAV) and equine arteritis virus (EAV) in equine cells. In addition, equine IFN-α1 inhibited the production of EIAV virus-like particles (VLP) from human 293T cells. An IFN-inducible human gene, tetherin, was induced in 293T cells by equine IFN-α1. Its induction correlated with the inhibition of VLP release from the cell membrane. This result indicates that equine IFN-α1 shares a similar mechanism of action with human IFN-α in regulating antiviral genes expression in human cells.
Collapse
Affiliation(s)
- Zhe Hu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agriculture Sciences, PR China
| | | | | | | |
Collapse
|
32
|
Wang J, Yang B, Hu Y, Zheng Y, Zhou H, Wang Y, Ma Y, Mao K, Yang L, Lin G, Ji Y, Wu X, Sun B. Negative regulation of Nmi on virus-triggered type I IFN production by targeting IRF7. THE JOURNAL OF IMMUNOLOGY 2013; 191:3393-9. [PMID: 23956435 DOI: 10.4049/jimmunol.1300740] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Viral infection causes host cells to produce type I IFNs, which play a critical role in viral clearance. IFN regulatory factor (IRF) 7 is the master regulator of type I IFN-dependent immune responses. In this article, we report that N-Myc and STATs interactor (Nmi), a Sendai virus-inducible protein, interacted with IRF7 and inhibited virus-triggered type I IFN production. The overexpression of Nmi inhibited the Sendai virus-triggered induction of type I IFNs, whereas the knockdown of Nmi promoted IFN production. Furthermore, the enhanced production of IFNs resulting from Nmi knockdown was sufficient to protect cells from infection by vesicular stomatitis virus. In addition, Nmi was found to promote the K48-linked ubiquitination of IRF7 and the proteasome-dependent degradation of this protein. Finally, an impairment of antiviral responses is also detectable in Nmi-transgenic mice. These findings suggest that Nmi is a negative regulator of the virus-triggered induction of type I IFNs that targets IRF7.
Collapse
Affiliation(s)
- Jie Wang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Xu D, Staedman A, Zhang L. CD20 antibody primes B lymphocytes for type I interferon production. PLoS One 2013; 8:e67900. [PMID: 23874371 PMCID: PMC3707517 DOI: 10.1371/journal.pone.0067900] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 05/22/2013] [Indexed: 12/31/2022] Open
Abstract
CD20 is a B cell surface marker that is expressed in various stages in B
lymphocytes and certain lymphomas. Clinical administration of CD20 antibody,
such as rituximab, is used widely to treat human B-cell lymphomas and other
diseases. However, CD20 antibody failed to treat systemic lupus erythematosus
(SLE or lupus). The reason for the failure is currently unknown. Type I
interferons (IFN) are a major component for the host innate immunity, and a key
pathogenic factor in lupus. We found that CD20 antibody potentiated human B
cells for its production of IFNs in vitro. This function was
specific to CD20-expressing cells and the potentiation function seems to be
instant. In addition, ectopic expression of CD20 in non-B-lymphocytes increased
the IFN promoter reporter activities. Because IFNs are a key pathogenic factor
in lupus, our data suggest that, in the presence of virus infection, the
CD20-antibody-mediated enhancement of IFN production might be related to its
failure in lupus treatments. This work may provide new insights for CD20-Ab
therapeutic applications.
Collapse
Affiliation(s)
- Dongsheng Xu
- School of Biological Sciences, University of Nebraska, Lincoln, Nebraska,
United States of America
| | - Andrew Staedman
- Nebraska Center for Virology, University of Nebraska, Lincoln, Nebraska,
United States of America
| | - Luwen Zhang
- School of Biological Sciences, University of Nebraska, Lincoln, Nebraska,
United States of America
- Nebraska Center for Virology, University of Nebraska, Lincoln, Nebraska,
United States of America
- * E-mail:
| |
Collapse
|
34
|
Gull I, Samra ZQ, Aslam MS, Athar MA. Heterologous expression, immunochemical and computational analysis of recombinant human interferon alpha 2b. SPRINGERPLUS 2013; 2:264. [PMID: 23875128 PMCID: PMC3695685 DOI: 10.1186/2193-1801-2-264] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 06/11/2013] [Indexed: 11/30/2022]
Abstract
Interferon alpha 2b (IFNα-2b) is an important cytokine and used for antiviral and anticancer treatment. The low cost production of IFNα-2b with high biological activity is necessary to provide the interferon therapy to the hepatitis patients in Pakistan. In the present study, human interferon alpha 2b (hIFNα-2b) gene from a healthy person was cloned and overexpressed in E. coli BL21(DE3). The molecular weight of the expressed hIFNα-2b is 19 kDa. The over expressed recombinant hIFNα-2b was checked by ELISA using antibodies raised against commercially available hIFNα-2b. The biocomputational analysis of recombinant hIFNα-2b gene showed the 99.9% nucleotide sequence and 100% deduced amino acid sequence homology with reported sequences of IFNα-2b. The predicted 3D-structure showed mainly five α-helices, one 310 helix and two disulfide bonds at Cys1-Cys98 and Cys129-Cys138. The amino acid sequence alignment indicated that the disulfide linkage position is conserved in all IFNα family members. On the basis of sequence homology among interferon alpha family, new potent variants of hIFNα-2b with enhance efficacy can be produced. Indigenous production of IFNα-2b from gene of local population will reduce the cost and increase tolerability of interferon therapy.
Collapse
Affiliation(s)
- Iram Gull
- Institute of Biochemistry and Biotechnology, University of the Punjab, Quaid-i-Azam Campus, Lahore, 54590 Pakistan
| | | | | | | |
Collapse
|
35
|
Xu L, Yang L, Liu W. Distinct evolution process among type I interferon in mammals. Protein Cell 2013; 4:383-92. [PMID: 23636688 PMCID: PMC4875548 DOI: 10.1007/s13238-013-3021-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 04/08/2013] [Indexed: 01/31/2023] Open
Abstract
Interferon (IFN) is thought to play an important role in the vertebrate immune system, but systemic knowledge of IFN evolution has yet to be elucidated. To evaluate the phylogenic distribution and evolutionary history of type I IFNs, 13genomes were searched using BLASTn program, and a phylogenetic tree of vertebrate type I IFNs was constructed. In the present study, an IFNδ-like gene in the human genome was identified, refuting the concept that humans have no IFNδ genes, and other mammalian IFN genes were also identified. In the phylogenetic tree, the mammalian IFNβ, IFNɛ, and IFNκ formed a clade separate from the other mammalian type I IFNs, while piscine and avian IFNs formed distinct clades. Based on this phylogenetic analysis and the various characteristics of type I IFNs, the evolutionary history of type I IFNs was further evaluated. Our data indicate that an ancestral IFNα-like gene forms a core from which new IFNs divided during vertebrate evolution. In addition, the data suggest how the other type I IFNs evolved from IFNα and shaped the complex type I IFN system. The promoters of type I IFNs were conserved among different mammals, as well as their genic regions. However, the intergenic regions of type I IFN clusters were not conserved among different mammals, demonstrating a high selection pressure upon type I IFNs during their evolution.
Collapse
Affiliation(s)
- Lei Xu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Limin Yang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101 China
| | - Wenjun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
- China-Japan Joint Laboratory of Molecular Immunology and Molecular Microbiology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101 China
| |
Collapse
|
36
|
Affiliation(s)
- Anthony M. Carter
- Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense DK-5000, Denmark;
| | - Allen C. Enders
- Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Davis, California 95616;
| |
Collapse
|
37
|
Carter AM. Evolution of Placental Function in Mammals: The Molecular Basis of Gas and Nutrient Transfer, Hormone Secretion, and Immune Responses. Physiol Rev 2012; 92:1543-76. [DOI: 10.1152/physrev.00040.2011] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Placenta has a wide range of functions. Some are supported by novel genes that have evolved following gene duplication events while others require acquisition of gene expression by the trophoblast. Although not expressed in the placenta, high-affinity fetal hemoglobins play a key role in placental gas exchange. They evolved following duplications within the beta-globin gene family with convergent evolution occurring in ruminants and primates. In primates there was also an interesting rearrangement of a cassette of genes in relation to an upstream locus control region. Substrate transfer from mother to fetus is maintained by expression of classic sugar and amino acid transporters at the trophoblast microvillous and basal membranes. In contrast, placental peptide hormones have arisen largely by gene duplication, yielding for example chorionic gonadotropins from the luteinizing hormone gene and placental lactogens from the growth hormone and prolactin genes. There has been a remarkable degree of convergent evolution with placental lactogens emerging separately in the ruminant, rodent, and primate lineages and chorionic gonadotropins evolving separately in equids and higher primates. Finally, coevolution in the primate lineage of killer immunoglobulin-like receptors and human leukocyte antigens can be linked to the deep invasion of the uterus by trophoblast that is a characteristic feature of human placentation.
Collapse
Affiliation(s)
- Anthony M. Carter
- Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
38
|
Li D, Tan W, Ma M, Yu X, Lai Q, Wu Z, Lin G, Hu C. Molecular characterization and transcription regulation analysis of type I IFN gene in grass carp (Ctenopharyngodon idella). Gene 2012; 504:31-40. [PMID: 22579963 DOI: 10.1016/j.gene.2012.04.091] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Revised: 04/17/2012] [Accepted: 04/30/2012] [Indexed: 12/30/2022]
Abstract
Type I interferons and interferon regulatory factor 7 (IRF7), which are crucial for innate immunity against viral infection, have been identified in many teleost fishes in recent years. In this study, the complete genomic sequence of grass carp (Ctenopharyngodon idella) type I interferon (termed CiIFN) (GU139255) and the full-length IRF7 cDNA sequence of grass carp (termed CiIRF7) (GQ141741) were cloned and characterized. CiIFN consists of 3368 bp, retaining the characteristic 5-exon/4-intron gene organization in fish type I IFNs. The CiIFN spans 5 exons and encodes a polypeptide of 180 amino acids, with the first 22 amino acids representing a putative signal peptide. The CiIFN promoter sequence was found to be 760 bp, which can be divided into a proximal region (from -1 to -140 bp) and a distal region (from -400 to -700 bp). The cDNA of CiIRF7 was found to be 1808 bp in full length, with an ORF of 1293 bp that encodes a putative protein of 430 amino acids. The putative amino acid sequence of CiIRF7 possesses a DNA-binding domain (DBD) in the N-terminal region. Real-time PCR analysis revealed that CiIFN displayed a low constitutive expression in all the tissues tested. After stimulation by polyinosinic:polycytidylic acid (Poly I:C), the expression of CiIFN was significantly up-regulated in most tissues of grass carp, with a relatively strong expression in spleen, kidney and intestine. The recombinant polypeptides of CiIRF7 and CiIRF7-nDBD were analyzed in gel mobility shift assays, along with the PCR amplification products of the proximal region (CiIFNP2), the distal region (CiIFNP6) and the full-length (CiIFNP7) of CiIFN promoter sequence. The results revealed that CiIRF7 could bind to the distal region as well as to the proximal region of CiIFN promoter sequence in vitro. Subsequently, the CiIFNPs (CiIFNP7/2/6) were cloned into pGL3-Basic vectors and CiIRF7 was subcloned into pcDNA3.1 vectors, then pGL3-CiIFNPs were separately transiently transfected or co-transfected with pcDNA3.1-CiIRF7 into the mouse myeloma cell lines (MMCL) SP2/0 and the grass carp kidney cell lines (CIK), and the impact of CiIRF7 on CiIFN promoter activity was measured by luciferase assays in the transfected cells. These results demonstrated that CiIRF7 acted as a positive regulator on the transcription of CiIFN.
Collapse
Affiliation(s)
- Dongming Li
- Department of Bioscience, College of Life Science and Food Engineering, Nanchang University, Nanchang 330031, China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Kawasaki T, Kawai T, Akira S. Recognition of nucleic acids by pattern-recognition receptors and its relevance in autoimmunity. Immunol Rev 2011; 243:61-73. [PMID: 21884167 PMCID: PMC7165622 DOI: 10.1111/j.1600-065x.2011.01048.x] [Citation(s) in RCA: 170] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Summary: Host cells trigger signals for innate immune responses upon recognition of conserved structures in microbial pathogens. Nucleic acids, which are critical components for inheriting genetic information in all species including pathogens, are key structures sensed by the innate immune system. The corresponding receptors for foreign nucleic acids include members of Toll‐like receptors, RIG‐I‐like receptors, and intracellular DNA sensors. While nucleic acid recognition by these receptors is required for host defense against the pathogen, there is a potential risk to the host of self‐nucleic acids recognition, thus precipitating autoimmune and autoinflammatory diseases. In this review, we discuss the roles of nucleic acid‐sensing receptors in guarding against pathogen invasion, discriminating between self and non‐self, and contributing to autoimmunity and autoinflammatory diseases.
Collapse
Affiliation(s)
- Takumi Kawasaki
- Laboratory of Host Defense, WPI Immunology Frontier Research Center, Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | | | | |
Collapse
|
40
|
Abstract
Type-I interferons (IFNs) are cytokines that have non-specific antiviral activity, participating mostly in innate defense mechanisms. Their administration has been proposed to treat several viral and immunomediated diseases as an immunomodulatory therapy. Due to its availability, recombinant human interferon-alpha (rHuIFN-α) has been studied in relation to feline retrovirosis, both in vitro and in vivo. However, IFNs are species-specific and antibodies have been shown to develop in response to the high rHuIFN-α doses necessary for an effective therapy. A recombinant feline IFN has been developed, which has been characterized as interferon-omega (rFeIFN-ω), designed to overcome these problems. Nonetheless, very few studies have been undertaken to evaluate its efficacy in cats naturally infected with FIV or FeLV. In an initial study, we here demonstrated that rFeIFN-ω can dramatically improve the clinical condition of infected cats, and induce improvement of hematologic parameters. Minor changes or no change was observed for hypergammaglobulinemia, CD4/CD8 ratio, proviral load, viremia and RT activity, suggesting that the overall effect of IFN was on innate immunity. More studies are needed in order to better understand its in vivo mechanisms.
Collapse
|
41
|
Erdmann J, Vitale G, van Koetsveld PM, Croze E, Sprij-Mooij DM, Hofland LJ, van Eijck CH. Effects of Interferons α/β on the Proliferation of Human Micro- and Macrovascular Endothelial Cells. J Interferon Cytokine Res 2011; 31:451-8. [DOI: 10.1089/jir.2009.0103] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Joris Erdmann
- Department of Surgery, Erasmus MC, Rotterdam, The Netherlands
- Department of Internal Medicine, Division Endocrinology, Erasmus MC, Rotterdam, The Netherlands
| | - Giovanni Vitale
- Department of Internal Medicine, Division Endocrinology, Erasmus MC, Rotterdam, The Netherlands
- Chair of Endocrinology, Department of Medical Sciences, Faculty of Medicine, University of Milan, Milan, Italy
- IRCCS, Instituto Auxologico Italiano, Milan, Italy
| | - Peter M. van Koetsveld
- Department of Internal Medicine, Division Endocrinology, Erasmus MC, Rotterdam, The Netherlands
| | - Ed Croze
- Department of Immunology, Berlex Bioscience, Inc., Richmond, California
| | - Diane M. Sprij-Mooij
- Department of Internal Medicine, Division Endocrinology, Erasmus MC, Rotterdam, The Netherlands
| | - Leo J. Hofland
- Department of Internal Medicine, Division Endocrinology, Erasmus MC, Rotterdam, The Netherlands
| | | |
Collapse
|
42
|
Akeno N, Smith EP, Stefan M, Huber AK, Zhang W, Keddache M, Tomer Y. IFN-α mediates the development of autoimmunity both by direct tissue toxicity and through immune cell recruitment mechanisms. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:4693-706. [PMID: 21402899 PMCID: PMC3106338 DOI: 10.4049/jimmunol.1002631] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
IFN-α is known to play a key role in autoimmunity, but the mechanisms are uncertain. Although the induction of autoimmunity by IFN-α is consistent with primarily immunomodulatory effects, the high frequency of nonautoimmune inflammation suggests other mechanisms. We used thyroiditis as a model to dissect these possibilities. IFN-α treatment of cultured thyrocytes increased expression of thyroid differentiation markers, thyroglobulin, thyroid-stimulating hormone receptor, thyroid peroxidase, and sodium iodide transporter. RNAseq analysis demonstrated that pathways of Ag presentation, pattern recognition receptors, and cytokines/chemokines were also stimulated. These changes were associated with markedly increased nonapoptotic thyroid cell death, suggesting direct toxicity. To corroborate these in vitro findings, we created transgenic mice with thyroid-specific overexpression of IFN-α under control of the thyroglobulin promoter. Transgenic mice developed marked inflammatory thyroid destruction associated with immune cell infiltration of thyroid and surrounding tissues leading to profound hypothyroidism, findings consistent with our in vitro results. In addition, transgenic mice thyroids showed upregulation of pathways similar to those observed in cultured thyrocytes. In particular, expression of granzyme B, CXCL10, a subset of the tripartite motif-containing family, and other genes involved in recruitment of bystander cytotoxic immune responses were increased. Pathways associated with apoptosis and autophagy were not induced. Taken together, our data demonstrate that the induction of tissue inflammation and autoimmunity by IFN-α involves direct tissue toxic effects as well as provocation of destructive bystander immune responses.
Collapse
Affiliation(s)
- Nagako Akeno
- Division of Endocrinology, the University of Cincinnati College of Medicine, Cincinnati, OH
| | - Eric P. Smith
- Division of Endocrinology, the University of Cincinnati College of Medicine, Cincinnati, OH
| | - Mihaela Stefan
- Department of Medicine, Division of Endocrinology, Mount Sinai School of Medicine, Genome Center, New York, NY
| | - Amanda K. Huber
- Department of Medicine, Division of Endocrinology, Mount Sinai School of Medicine, Genome Center, New York, NY
| | - Weijia Zhang
- Department of Medicine Bioinformatics Core, Mount Sinai School of Medicine, Genome Center, New York, NY
| | - Mehdi Keddache
- Cincinnati Children’s Hospital Medical Center, New York, NY
| | - Yaron Tomer
- Department of Medicine, Division of Endocrinology, Mount Sinai School of Medicine, Genome Center, New York, NY
- James J. Peters VA Medical Center, New York, NY
| |
Collapse
|
43
|
Chon TW, Bixler S. Interferon-tau: current applications and potential in antiviral therapy. J Interferon Cytokine Res 2011; 30:477-85. [PMID: 20626290 DOI: 10.1089/jir.2009.0089] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Interferon-tau (IFN-tau) was initially identified as an ovine pregnancy protein. Produced by the trophoblast, it is important in preventing degradation of the corpus luteum and has been used as an early marker for ovine pregnancy. As a member of the family of type I interferons, IFN-tau has demonstrated promising antiviral activity against human viral infections in vitro. Additionally, it displays high species cross-reactivity despite its absence in humans. To date, IFN-tau has shown efficacy in reducing replication of human immunodeficiency virus, feline immunodeficiency virus, and human papillomavirus. While IFN-tau shares similar antiviral activity to IFN-alpha, the current interferon of choice for treatment of viral infections, it lacks the associated toxicity. This may make IFN-tau an attractive alternative to IFN-alpha for the treatment of viral infections.
Collapse
Affiliation(s)
- Thomas W Chon
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA.
| | | |
Collapse
|
44
|
Groebner AE, Schulke K, Schefold JC, Fusch G, Sinowatz F, Reichenbach HD, Wolf E, Meyer HHD, Ulbrich SE. Immunological mechanisms to establish embryo tolerance in early bovine pregnancy. Reprod Fertil Dev 2011; 23:619-32. [DOI: 10.1071/rd10230] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Accepted: 12/21/2010] [Indexed: 12/16/2022] Open
Abstract
A well-balanced immunological interaction between mother and the semi-allogenic embryo is of particular importance. The objective of the present study was to analyse mechanisms of immune tolerance in bovine pregnancy during peri-implantation. Simmental heifers inseminated with either cryopreserved spermatozoa or seminal plasma were killed 12, 15 or 18 days after oestrus. Uteri were flushed for the recovery of conceptuses and the ipsilateral intercaruncular endometrium was sampled for gene expression analysis. Indoleamine 2,3-dioxygenase (IDO) mRNA, coding for the initial enzyme of the kynurenine pathway, was 18-fold (P < 0.001) more abundant in the endometrium of Day 18 pregnant v. non-pregnant animals. Tandem mass spectrometry revealed a decrease of endometrial l-tryptophan (P = 0.0008), but an increase of l-kynurenine concentration (P = 0.005) from Day 12 to Day 18, suggesting increasing IDO activity (P < 0.03). An in vitro coculture model of endometrial cells showed an induction of IDO expression following interferon-τ exposure primarily in stroma cells, which was confirmed by in situ hybridisation localising IDO mRNA mainly in deep stroma cells. Immunohistochemical analysis revealed fewer CD45-positive leucocytes in the zona basalis of pregnant animals. Elevated IDO activity may reduce the presence of leucocytes in the pregnant endometrium, providing a possible mechanism for protecting the semi-allogenic conceptus from maternal rejection.
Collapse
|
45
|
Hellgren O, Ekblom R. Evolution of a cluster of innate immune genes (beta-defensins) along the ancestral lines of chicken and zebra finch. Immunome Res 2010; 6:3. [PMID: 20359324 PMCID: PMC3161384 DOI: 10.1186/1745-7580-6-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 04/01/2010] [Indexed: 12/24/2022] Open
Abstract
Background Avian β-defensins (AvBDs) represent a group of innate immune genes with broad antimicrobial activity. Within the chicken genome, previous work identified 14 AvBDs in a cluster on chromosome three. The release of a second bird genome, the zebra finch, allows us to study the comparative evolutionary history of these gene clusters between from two species that shared a common ancestor about 100 million years ago. Results A phylogenetic analysis of the β-defensin gene clusters in the chicken and the zebra finch identified several cases of gene duplication and gene loss along their ancestral lines. In the zebra finch genome a cluster of 22 AvBD genes were identified, all located within 125 Kbp on chromosome three. Ten of the 22 genes were found to be highly conserved with orthologous genes in the chicken genome. The remaining 12 genes were all located within a cluster of 58 Kbp and are suggested to be a result of recent gene duplication events that occurred after the galliformes- passeriformes split (G-P split). Within the chicken genome, AvBD6 was found to be a duplication of AvBD7, whereas the gene AvDB14 seems to have been lost along the ancestral line of the zebra finch. The duplicated β-defensin genes have had a significantly higher accumulation of non-synonymous over synonymous substitutions compared to the genes that have not undergone duplication since the G-P split. The expression patterns of avian β-defensin genes seem to be well conserved between chicken and zebra finch. Conclusion The genomic comparisons of the β-defensins gene clusters of the chicken and zebra finch illuminate the evolutionary history of this gene complex. Along their ancestral lines, several gene duplication events have occurred in the passerine line after the galliformes-passeriformes split giving rise to 12 novel genes compared to a single duplication event in the galliformes line. After the duplication events, the duplicated genes have been subject to a relaxed selection pressure compared to the non-duplicated genes, thus supporting models of evolution by gene duplication.
Collapse
Affiliation(s)
- Olof Hellgren
- Edward Grey Institute, Department of Zoology, South Parks Road, Oxford, OX1 3PS, UK.
| | | |
Collapse
|
46
|
Yoneyama M, Fujita T. Recognition of viral nucleic acids in innate immunity. Rev Med Virol 2009; 20:4-22. [DOI: 10.1002/rmv.633] [Citation(s) in RCA: 241] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
47
|
Berraondo P, Crettaz J, Ochoa L, Vales A, Ruiz J, Prieto J, Martinez-Ansó E, González-Aseguinolaza G. Production of recombinant woodchuck IFNalpha and development of monoclonal antibodies. J Interferon Cytokine Res 2009; 29:75-82. [PMID: 19014334 DOI: 10.1089/jir.2008.0012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Interferon alpha (IFNalpha) is the first line treatment for chronic hepatitis B and C. In order to test new IFNalpha delivery systems and investigate the function of this cytokine in the woodchuck model, the best animal model of chronic hepatitis B, we produced and purified recombinant woodchuck IFNalpha and used it to produce monoclonal antibodies. wIFNalpha5 was cloned in a prokaryotic expression system, expressed as His-tagged protein and then purified. The rwIFNalpha5 protein was found to induce STAT-3 phosphorylation, to enhance 2',5'-oligoadenylate synthetase mRNA levels and to possess a potent antiviral activity. Two monoclonal antibodies were obtained through immunization of rats with rwIFNalpha5. Both recognized rwIFNalpha5 in western blot analysis and one was able to neutralize the antiviral activity of the rwIFNalpha5 and lymphoblastoid IFNalpha preparations. Finally, a capture rwIFNalpha5 ELISA was developed using both antibodies. In summary, the tools generated in this study will allow different forms of IFNalpha delivery as well as different combination therapies in woodchuck hepatitis virus infection to be tested, thus providing useful information for the design of new strategies to treat chronic hepatitis B in humans.
Collapse
Affiliation(s)
- Pedro Berraondo
- Laboratory of Gene Therapy of Viral Hepatitis, Division of Hepatology and Gene Therapy, Center for Applied Medical Research (CIMA), Clínica Universitaria/School of Medicine, University of Navarra, 31080 Pamplona, Navarra, Spain
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Tough DF. Type I Interferon as a Link Between Innate and Adaptive Immunity through Dendritic Cell Stimulation. Leuk Lymphoma 2009; 45:257-64. [PMID: 15101709 DOI: 10.1080/1042819031000149368] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Type I interferon (IFN-alpha/beta) is expressed rapidly after infection and plays a key role in innate defense against pathogens. Recent studies have shown that a connection exists between IFN-alpha/beta and antigen-presenting dendritic cells (DCs) at two levels. Firstly, a specific DC precursor, the plasmacytoid pre-DC (p-preDC), was identified as a cell type able to secrete very high amounts of IFN-alpha/beta following stimulation with infectious agents. Secondly, IFN-alpha/beta has been shown to act as a differentiation/maturation factor for DCs. These findings will be discussed in association with evidence indicating that IFN-alpha/beta can enhance and modulate immune responses in vivo. Taken together, the available data suggest that IFN-alpha/beta serves as a link between the innate response to infection and the adaptive immune response.
Collapse
Affiliation(s)
- David F Tough
- The Edward Jenner Institute for Vaccine Research, Compton, Newbury, RG20 7NN, UK.
| |
Collapse
|
49
|
Kitao Y, Kono T, Korenaga H, Iizasa T, Nakamura K, Savan R, Sakai M. Characterization and expression analysis of type I interferon in common carp Cyprinus carpio L. Mol Immunol 2009; 46:2548-56. [DOI: 10.1016/j.molimm.2009.05.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2009] [Revised: 05/12/2009] [Accepted: 05/14/2009] [Indexed: 11/28/2022]
|
50
|
Walker AM, Kimura K, Roberts MR. Expression of bovine interferon-tau variants according to sex and age of conceptuses. Theriogenology 2009; 72:44-53. [PMID: 19324401 PMCID: PMC2692756 DOI: 10.1016/j.theriogenology.2009.01.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Revised: 01/21/2009] [Accepted: 01/24/2009] [Indexed: 10/21/2022]
Abstract
Interferon-tau (IFNT), which plays a major role in maternal recognition of pregnancy in cattle, is transcribed from multiple genes. Moreover, there are at least 12 cDNA variants, many presumably allelic. Although the IFNT locus is autosomal, Day 8 female blastocysts produced approximately twice as much antiviral activity as males. The questions addressed here are whether male and female blastocysts differed in the kind and number of IFNT they expressed, and whether this pattern changed over development. Day 8, in vitro-produced blastocysts were bisected, and one half of each was sexed by PCR. Demi-embryos (n=64) were grouped according to whether they were male or female, to provide two pools of each sex. Individual cDNA were sequenced after RT-PCR amplification and shot-gun cloning to provide comparisons between male and female blastocysts, elongating conceptuses of various developmental ages (Days 14-19), and a female trophoblast cell line (CT-1). A total of 376 cDNA clones were sequenced. Six additional cDNA were identified, in addition to the forms described earlier. There were no differences between male and female blastocysts (P=0.54), and between blastocysts and a trophoblast model system (CT-1 cells; P=0.24) in the IFNT transcripts expressed, indicating that sexual dimorphism was not correlated with particular IFNT variants. There were differences in variant frequencies (P<0.001) among conceptuses of different age, although two, representing boIFN1a and boIFN3c, predominated throughout development. Notably, no alteration in overall IFNT variant diversity was detected in CT-1 cells over time (P=0.124).
Collapse
Affiliation(s)
- Angela M. Walker
- Department of Veterinary Pathobiology, University of Missouri, Columbia MO 65211, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia MO 65211, USA
| | - Koji Kimura
- National Institute of Livestock and Grassland Science, Reproductive Physiology Laboratory, Tochigi 329-2793, Japan
| | - Michael R. Roberts
- Department of Veterinary Pathobiology, University of Missouri, Columbia MO 65211, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia MO 65211, USA
| |
Collapse
|