1
|
Proteomic Analysis of ISGylation in Immortalized Porcine Alveolar Macrophage Cell Lines Induced by Type I Interferon. Vaccines (Basel) 2021; 9:vaccines9020164. [PMID: 33671165 PMCID: PMC7922875 DOI: 10.3390/vaccines9020164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/12/2021] [Accepted: 02/13/2021] [Indexed: 12/21/2022] Open
Abstract
Interferon-stimulated gene product 15 (ISG15), a ubiquitin-like molecule, can be conjugated to protein substrates through a reversible process known as ISGylation. ISG15 and ISGylation are both strongly upregulated by type I interferons and play putative key roles in host innate immunity against viral infection. However, the function of ISGylation and identities of ISGylation substrates are largely unknown. Here, a novel monoclonal antibody (Mab) that specifically recognizes porcine ISG15 (pISG15) was employed to capture ISG15-conjugated proteins from IFNs-stimulated porcine cell lysates. Next, Mab-captured conjugates were analyzed using proteomics-based tools to identify potential ISGylation protein targets in order to elucidate the roles of ISG15 and ISGylation in porcine cells. Subsequently, 190 putative ISGylation sites were detected within 98 identified ISGylation candidates; several candidates contained more than one ISGylation-modifiable lysine residue, including pISG15 itself. Motif enrichment analysis of confirmed ISGylation sites demonstrated a moderate bias towards certain sites with specific upstream amino acid residues. Meanwhile, results of Gene Ontology (GO)-based annotation and functional enrichment and protein-protein interaction (PPI) network analyses of porcine ISG15-conjugated substrate proteins indicated that these substrates were mainly associated with the host metabolism, especially nucleotide metabolic pathways that ultimately may participate in cellular antiviral defenses. Notably, several ISGs (MX1, IFIT1, OAS1, ISG15 and putative ISG15 E3 ligase Herc6) were also identified as putative ISGylation substrates within a regulatory loop involving ISGylation of ISGs themselves. Taken together, proteomics analysis of porcine ISGylation substrates revealed putative functional roles of ISG15 and novel host ISGylation targets that may ultimately be involved in cellular antiviral responses.
Collapse
|
2
|
IFN- γ Mediates the Development of Systemic Lupus Erythematosus. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7176515. [PMID: 33123584 PMCID: PMC7586164 DOI: 10.1155/2020/7176515] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/17/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022]
Abstract
Objective Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that can affect all organs in the body. It is characterized by overexpression of antibodies against autoantigen. Although previous bioinformatics analyses have identified several genetic factors underlying SLE, they did not discriminate between naive and individuals exposed to anti-SLE drugs. Here, we evaluated specific genes and pathways in active and recently diagnosed SLE population. Methods GSE46907 matrix downloaded from Gene Expression Omnibus (GEO) was analyzed using R, Metascape, STRING, and Cytoscape to identify differentially expressed genes (DEGs), enrichment pathways, protein-protein interaction (PPI), and hub genes between naive SLE individuals and healthy controls. Results A total of 134 DEGs were identified, in which 29 were downregulated, whereas 105 were upregulated in active and newly diagnosed SLE cases. GO term analysis revealed that transcriptional induction of the DEGs was particularly enhanced in response to secretion of interferon-γ and interferon-α and regulation of cytokine production innate immune responses among others. KEGG pathway analysis showed that the expression of DEGs was particularly enhanced in interferon signaling, IFN antiviral responses by activated genes, class I major histocompatibility complex (MHC-I) mediated antigen processing and presentation, and amyloid fiber formation. STAT1, IRF7, MX1, OASL, ISG15, IFIT3, IFIH1, IFIT1, OAS2, and GBP1 were the top 10 DEGs. Conclusions Our findings suggest that interferon-related gene expression and pathways are common features for SLE pathogenesis, and IFN-γ and IFN-γ-inducible GBP1 gene in naive SLE were emphasized. Together, the identified genes and cellular pathways have expanded our understanding on the mechanism underlying development of SLE. They have also opened a new frontier on potential biomarkers for diagnosis, biotherapy, and prognosis for SLE.
Collapse
|
3
|
Honkala AT, Tailor D, Malhotra SV. Guanylate-Binding Protein 1: An Emerging Target in Inflammation and Cancer. Front Immunol 2020; 10:3139. [PMID: 32117203 PMCID: PMC7025589 DOI: 10.3389/fimmu.2019.03139] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 12/24/2019] [Indexed: 12/16/2022] Open
Abstract
Guanylate-binding protein 1 (GBP1) is a large GTPase of the dynamin superfamily involved in the regulation of membrane, cytoskeleton, and cell cycle progression dynamics. In many cell types, such as endothelial cells and monocytes, GBP1 expression is strongly provoked by interferon γ (IFNγ) and acts to restrain cellular proliferation in inflammatory contexts. In immunity, GBP1 activity is crucial for the maturation of autophagosomes infected by intracellular pathogens and the cellular response to pathogen-associated molecular patterns. In chronic inflammation, GBP1 activity inhibits endothelial cell proliferation even as it protects from IFNγ-induced apoptosis. A similar inhibition of proliferation has also been found in some tumor models, such as colorectal or prostate carcinoma mouse models. However, this activity appears to be context-dependent, as in other cancers, such as oral squamous cell carcinoma and ovarian cancer, GBP1 activity appears to anchor a complex, taxane chemotherapy resistance profile where its expression levels correlate with worsened prognosis in patients. This discrepancy in GBP1 function may be resolved by GBP1's involvement in the induction of a cellular senescence phenotype, wherein anti-proliferative signals coincide with potent resistance to apoptosis and set the stage for dysregulated proliferative mechanisms present in growing cancers to hijack GBP1 as a pro- chemotherapy treatment resistance (TXR) and pro-survival factor even in the face of continued cytotoxic treatment. While the structure of GBP1 has been extensively characterized, its roles in inflammation, TXR, senescence, and other biological functions remain under-investigated, although initial findings suggest that GBP1 is a compelling target for therapeutic intervention in a variety of conditions ranging from chronic inflammatory disorders to cancer.
Collapse
Affiliation(s)
- Alexander T Honkala
- Department of Radiation Oncology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Dhanir Tailor
- Department of Radiation Oncology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Sanjay V Malhotra
- Department of Radiation Oncology, School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
4
|
Nan Y, Wu C, Zhang YJ. Interferon Independent Non-Canonical STAT Activation and Virus Induced Inflammation. Viruses 2018; 10:v10040196. [PMID: 29662014 PMCID: PMC5923490 DOI: 10.3390/v10040196] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/07/2018] [Accepted: 04/11/2018] [Indexed: 02/06/2023] Open
Abstract
Interferons (IFNs) are a group of secreted proteins that play critical roles in antiviral immunity, antitumor activity, activation of cytotoxic T cells, and modulation of host immune responses. IFNs are cytokines, and bind receptors on cell surfaces to trigger signal transduction. The major signaling pathway activated by IFNs is the JAK/STAT (Janus kinase/signal transducer and activator of transcription) pathway, a complex pathway involved in both viral and host survival strategies. On the one hand, viruses have evolved strategies to escape from antiviral host defenses evoked by IFN-activated JAK/STAT signaling. On the other hand, viruses have also evolved to exploit the JAK/STAT pathway to evoke activation of certain STATs that somehow promote viral pathogenesis. In this review, recent progress in our understanding of the virus-induced IFN-independent STAT signaling and its potential roles in viral induced inflammation and pathogenesis are summarized in detail, and perspectives are provided.
Collapse
Affiliation(s)
- Yuchen Nan
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
- Molecular Virology Laboratory, VA-MD College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA.
| | - Chunyan Wu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Yan-Jin Zhang
- Molecular Virology Laboratory, VA-MD College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
5
|
Nan Y, Wu C, Gu G, Sun W, Zhang YJ, Zhou EM. Improved Vaccine against PRRSV: Current Progress and Future Perspective. Front Microbiol 2017; 8:1635. [PMID: 28894443 PMCID: PMC5581347 DOI: 10.3389/fmicb.2017.01635] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 08/11/2017] [Indexed: 12/20/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV), one of the most economically significant pathogens worldwide, has caused numerous outbreaks during the past 30 years. PRRSV infection causes reproductive failure in sows and respiratory disease in growing and finishing pigs, leading to huge economic losses for the swine industry. This impact has become even more significant with the recent emergence of highly pathogenic PRRSV strains from China, further exacerbating global food security. Since new PRRSV variants are constantly emerging from outbreaks, current strategies for controlling PRRSV have been largely inadequate, even though our understanding of PRRSV virology, evolution and host immune response has been rapidly expanding. Meanwhile, practical experience has revealed numerous safety and efficacy concerns for currently licensed vaccines, such as shedding of modified live virus (MLV), reversion to virulence, recombination between field strains and MLV and failure to elicit protective immunity against heterogeneous virus. Therefore, an effective vaccine against PRRSV infection is urgently needed. Here, we systematically review recent advances in PRRSV vaccine development. Antigenic variations resulting from PRRSV evolution, identification of neutralizing epitopes for heterogeneous isolates, broad neutralizing antibodies against PRRSV, chimeric virus generated by reverse genetics, and novel PRRSV strains with interferon-inducing phenotype will be discussed in detail. Moreover, techniques that could potentially transform current MLV vaccines into a superior vaccine will receive special emphasis, as will new insights for future PRRSV vaccine development. Ultimately, improved PRRSV vaccines may overcome the disadvantages of current vaccines and minimize the PRRS impact to the swine industry.
Collapse
Affiliation(s)
- Yuchen Nan
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F UniversityYangling, China
| | - Chunyan Wu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F UniversityYangling, China
| | - Guoqian Gu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F UniversityYangling, China
| | - Weiyao Sun
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F UniversityYangling, China
| | - Yan-Jin Zhang
- Molecular Virology Laboratory, Virginia-Maryland College of Veterinary Medicine and Maryland Pathogen Research Institute, University of Maryland, College ParkMD, United States
| | - En-Min Zhou
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F UniversityYangling, China
| |
Collapse
|
6
|
Interferon induction by RNA viruses and antagonism by viral pathogens. Viruses 2014; 6:4999-5027. [PMID: 25514371 PMCID: PMC4276940 DOI: 10.3390/v6124999] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 11/04/2014] [Accepted: 12/04/2014] [Indexed: 12/22/2022] Open
Abstract
Interferons are a group of small proteins that play key roles in host antiviral innate immunity. Their induction mainly relies on host pattern recognition receptors (PRR). Host PRR for RNA viruses include Toll-like receptors (TLR) and retinoic acid-inducible gene I (RIG-I) like receptors (RLR). Activation of both TLR and RLR pathways can eventually lead to the secretion of type I IFNs, which can modulate both innate and adaptive immune responses against viral pathogens. Because of the important roles of interferons, viruses have evolved multiple strategies to evade host TLR and RLR mediated signaling. This review focuses on the mechanisms of interferon induction and antagonism of the antiviral strategy by RNA viruses.
Collapse
|
7
|
Abstract
Interferon-stimulated gene (ISG) products take on a number of diverse roles. Collectively, they are highly effective at resisting and controlling pathogens. In this review, we begin by introducing interferon (IFN) and the JAK-STAT signaling pathway to highlight features that impact ISG production. Next, we describe ways in which ISGs both enhance innate pathogen-sensing capabilities and negatively regulate signaling through the JAK-STAT pathway. Several ISGs that directly inhibit virus infection are described with an emphasis on those that impact early and late stages of the virus life cycle. Finally, we describe ongoing efforts to identify and characterize antiviral ISGs, and we provide a forward-looking perspective on the ISG landscape.
Collapse
Affiliation(s)
- William M. Schneider
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065
| | | | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065
| |
Collapse
|
8
|
Katzenell S, Chen Y, Parker ZM, Leib DA. The differential interferon responses of two strains of Stat1-deficient mice do not alter susceptibility to HSV-1 and VSV in vivo. Virology 2014; 450-451:350-4. [PMID: 24503098 DOI: 10.1016/j.virol.2013.12.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 12/06/2013] [Accepted: 12/13/2013] [Indexed: 01/14/2023]
Abstract
Stat1 is a pivotal transcription factor for generation of the interferon (IFN)-dependent antiviral response. Two Stat1 knockout mouse lines have been previously generated, one deleted the N-terminal domain (ΔNTD) and one in the DNA-binding domain (ΔDBD). These widely-used strains are assumed interchangeable, and both are highly susceptible to various pathogens. In this study, primary cells derived from ΔNTD mice were shown to be significantly more responsive to IFN, and established an antiviral state with greater efficiency than cells derived from ΔDBD mice, following infection with vesicular stomatitis virus and herpes simplex virus type-1. Also, while mice from both strains succumbed rapidly and equally to virus infection, ΔDBD mice supported significantly higher replication in brains and livers than ΔNTD mice. Endpoint-type experimental comparisons of these mouse strains are therefore misleading in failing to indicate important differences in virus replication and innate response.
Collapse
Affiliation(s)
- Sarah Katzenell
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States
| | - Yufei Chen
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States
| | - Zachary M Parker
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States
| | - David A Leib
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States.
| |
Collapse
|
9
|
Sjöstrand M, Ambrosi A, Brauner S, Sullivan J, Malin S, Kuchroo VK, Espinosa A, Wahren-Herlenius M. Expression of the Immune Regulator Tripartite-Motif 21 Is Controlled by IFN Regulatory Factors. THE JOURNAL OF IMMUNOLOGY 2013; 191:3753-63. [DOI: 10.4049/jimmunol.1202341] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
10
|
Abstract
We look back on the discoveries that the tyrosine kinases TYK2 and JAK1 and the transcription factors STAT1, STAT2, and IRF9 are required for the cellular response to type I interferons. This initial description of the JAK-STAT pathway led quickly to additional discoveries that type II interferons and many other cytokines signal through similar mechanisms. This well-understood pathway now serves as a paradigm showing how information from protein-protein contacts at the cell surface can be conveyed directly to genes in the nucleus. We also review recent work on the STAT proteins showing the importance of several different posttranslational modifications, including serine phosphorylation, acetylation, methylation, and sumoylation. These remarkably proficient proteins also provide noncanonical functions in transcriptional regulation and they also function in mitochondrial respiration and chromatin organization in ways that may not involve transcription at all.
Collapse
Affiliation(s)
- George R. Stark
- Department of Molecular Genetics, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - James E. Darnell
- Laboratory of Molecular Cell Biology, The Rockefeller University, New York, NY 10065-6399, USA
| |
Collapse
|
11
|
Santos CI, Costa-Pereira AP. Signal transducers and activators of transcription-from cytokine signalling to cancer biology. Biochim Biophys Acta Rev Cancer 2011; 1816:38-49. [PMID: 21447371 DOI: 10.1016/j.bbcan.2011.03.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 03/17/2011] [Accepted: 03/22/2011] [Indexed: 10/18/2022]
Abstract
Signal transducers and activators of transcription (STATs) are, as the name indicates, both signal transducers and transcription factors. STATs are activated by cytokines and some growth factors and thus control important biological processes. These include cell growth, cell differentiation, apoptosis and immune responses. Dysregulation of STATs, either due to constitutive activation or function impairment, can have, therefore, deleterious biological consequences. This review places particular emphasis on their structural organization, biological activities and regulatory mechanisms most commonly utilized by cells to control STAT-mediated signalling. STATs also play important roles in cancer and immune deficiencies and are thus being exploited as therapeutic targets.
Collapse
Affiliation(s)
- Cristina Isabel Santos
- Imperial College London, Faculty of Medecine, Department of Surgery and Cancer, Hammersmith Hospital Campus, Du Cane Road, London W12 ONN, UK
| | | |
Collapse
|
12
|
Olszewski MA, Gray J, Vestal DJ. In silico genomic analysis of the human and murine guanylate-binding protein (GBP) gene clusters. J Interferon Cytokine Res 2007; 26:328-52. [PMID: 16689661 DOI: 10.1089/jir.2006.26.328] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The guanylate-binding proteins (GBPs) were among the first interferon (IFN)-stimulated genes (ISGs) discovered, but until recently, little was known about their functions and even less about the composition of the gene family. Analysis of the promoter of human GBP-1 contributed significantly toward the understanding of Jak-Stat signaling and the delineation of the IFN-gamma activation site (GAS) and IFN-stimulated response element (ISRE) promoter elements. In this study, we have examined the genomic arrangement and composition of the GBPs in both mouse and humans. There are seven GBP paralogs in humans and at least one pseudogene, all of which are located in a cluster of genes on chromosome 1. Five of the six MuGBPs and a GBP pseudogene are clustered in a syntenic region on chromosome 3. The sixth MuGBP, MuGBP-4, and three GBP pseudogenes are located on chromosome 5. As might be expected, the GBPs share similar genomic organizations of introns and exons. Five of the MuGBPs had previously been shown to be coordinately induced by IFNs, and as expected, all of the MuGBPs have GAS and ISRE elements in their promoters. Interestingly, not all of the HuGBPs have GAS and ISRE elements, suggesting that not all GBPs are IFN responsive in humans.
Collapse
Affiliation(s)
- Maureen A Olszewski
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | | | | |
Collapse
|
13
|
Todorova K, Tsoncheva V, Mitev V, Ivanov I, Maximova V. Modulation the Effect of Interferon by Diverse Protein Kinase Inhibitors. BIOTECHNOL BIOTEC EQ 2007. [DOI: 10.1080/13102818.2007.10817498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
14
|
Browne SK, Roesser JR, Zhu SZ, Ginder GD. Differential IFN-gamma stimulation of HLA-A gene expression through CRM-1-dependent nuclear RNA export. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 177:8612-9. [PMID: 17142760 DOI: 10.4049/jimmunol.177.12.8612] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IFNs regulate most MHC class I genes by stimulating transcription initiation. As shown previously, IFN-gamma controls HLA-A expression primarily at the posttranscriptional level. We have defined two 8-base sequences in a 39-nucleotide region in the 3'-transcribed region of the HLA-A gene that are required for the posttranscriptional response to IFN-gamma. Stimulation of HLA-A expression by IFN-gamma requires nuclear export of HLA-A mRNA by chromosome maintenance region 1 (CRM-1). Treatment of cells with leptomycin B, a specific inhibitor of CRM-1, completely inhibited IFN-gamma induction of HLA-A. Expression of a truncated, dominant-negative form of the nucleoporin NUP214/CAN, DeltaCAN, that specifically interacts with CRM-1, also prevented IFN-gamma stimulation of HLA-A, providing confirmation of the role of CRM-1. Increased expression of HLA-A induced by IFN-gamma also requires protein methylation, as shown by the fact that treatment of SK-N-MC cells or HeLa cells with the PRMT1 inhibitor 5'-methyl-5'-thioadenosine abolished the cellular response to IFN-gamma. In contrast with HLA-A, IFN-gamma-induced expression of the HLA class Ib gene, HLA-E, was not affected by either 5'-methyl-5'-thioadenosine or leptomycin B. These results provide proof of principle that it is possible to differentially modulate the IFN-gamma-induced expression of the HLA-E and HLA-A genes, whose products often mediate opposing effects on cellular immunity to tumor cells, pathogens, and autoantigens.
Collapse
Affiliation(s)
- Sarah K Browne
- Massey Cancer Center, Virginia Commonwealth University Medical Center, 401 College Street, Richmond, VA 23298, USA
| | | | | | | |
Collapse
|
15
|
Vestal DJ. Review:The Guanylate-Binding Proteins (GBPs): Proinflammatory Cytokine-Induced Members of the Dynamin Superfamily with Unique GTPase Activity. J Interferon Cytokine Res 2005; 25:435-43. [PMID: 16108726 DOI: 10.1089/jir.2005.25.435] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The guanylate-binding proteins (GBPs) were first identified in the late 1970s, and within a short period of time, investigators were aware that GBPs possessed unique properties, in particular the ability to bind GMP agarose. Since then, much study has gone into understanding their mechanism of induction by interferons (IFNs) and other cytokines, and they have been used extensively as markers for IFN responsiveness in both cells and organisms. In time, we learned that GBPs had the unusual ability to hydrolyze GTP to both GDP and GMP. More recently, we have begun to appreciate their novel structure, one that suggests unique mechanisms of GTP binding and hydrolysis and unique forms of regulation. In addition, we have begun to unravel some of their functions and to separate these function into those functions that do and those that do not require GTPase activity.
Collapse
Affiliation(s)
- Deborah J Vestal
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA.
| |
Collapse
|
16
|
Zhang C, Zhang J, Sun R, Feng J, Wei H, Tian Z. Opposing effect of IFNγ and IFNα on expression of NKG2 receptors: Negative regulation of IFNγ on NK cells. Int Immunopharmacol 2005; 5:1057-67. [PMID: 15829421 DOI: 10.1016/j.intimp.2005.02.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2004] [Revised: 09/07/2004] [Accepted: 02/03/2005] [Indexed: 11/26/2022]
Abstract
The effector functions of natural killer (NK) cells are regulated by integrated signals across an array of stimulatory and inhibitory receptors interacting with target cell surface ligands. The regulatory effect of interferon-alpha (IFNalpha) and interferon-gamma (IFNgamma) on expression of the family of NKG2 receptors, stimulatory NKG2D receptor and inhibitory NKG2A receptor, and cytolysis of the target tumor cells (MICA+ and HLA-E+) were studied. Results show that IFNgamma and IFNalpha influence NK cell function differently. Interferon-alpha stimulates expression of stimulatory NKG2D receptors and inhibits the expression of inhibitory NKG2A receptors on NK cells. Contrary to the stimulatory effect of IFNalpha, IFNgamma inhibits cytolysis by NK cells of tumor cells expressing MICA or HLA-E cell surface proteins. Blocking NKG2D or NKG2A receptor activity with monoclonal antibodies partly attenuates the inhibitory effect of IFNgamma while promoting the effects of IFNalpha on NK cytolysis. These results show for the first time that IFNgamma negatively regulates NK cells through NKG2 receptors, and that the balance between stimulatory and inhibitory signals through the NKG2 family of receptors may be controlled by two opposing interferons. Modulating the balance between stimulatory and inhibitory signals through cell surface receptors on NK cells may open a new approach to NK cell-based biotherapy for cancer and infectious diseases.
Collapse
Affiliation(s)
- Cai Zhang
- Institute of Immunopharmacology and Immunotherapy, School of Pharmacy, Shandong University 44 Wenhua Western Road, Jinan 250012, China
| | | | | | | | | | | |
Collapse
|
17
|
Naschberger E, Bauer M, Stürzl M. Human guanylate binding protein-1 (hGBP-1) characterizes and establishes a non-angiogenic endothelial cell activation phenotype in inflammatory diseases. ACTA ACUST UNITED AC 2005; 45:215-27. [PMID: 16005050 DOI: 10.1016/j.advenzreg.2005.02.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Blood vessel activation in inflammatory diseases is triggered by a myriad of different factors that partially reveal opposite activities on endothelial cells (EC). For example, inflammatory cytokines (IC) inhibit EC proliferation and induce cell adhesiveness for leukocytes. In contrast, angiogenic growth factors (AGF) activate EC proliferation and inhibit cell adhesiveness for leukocytes. In consequence, IC and AGF may induce two different activation phenotypes in EC that appear in a temporally and/or spatially coordinated manner in inflammatory tissues. Human guanylate binding protein-1 (hGBP-1) is a member of the large GTPase protein family. New results demonstrate that hGBP-1 is a specific marker of IC-activated EC that allows to differentiate the IC- and AGF-activated phenotype of EC at the single cell level, both in vitro and in vivo. In addition, hGBP-1 is the key mediator of the inhibitory effects of IC on EC proliferation and invasiveness. Both the expression pattern of hGBP-1 and its activity in EC supported the hypothesis that IC- and AGF-activation induce distinct adversely related phenotypes in EC. In future, hGBP-1 may be used as a marker to monitor the IC-induced phenotype of EC in inflammation and may also be exploited as a target to modulate EC activity in inflammatory diseases and tumor angiogenesis.
Collapse
Affiliation(s)
- Elisabeth Naschberger
- Division of Molecular and Experimental Surgery, Department of Surgery, University of Erlangen-Nürnberg, Schwabachanlage 10, 91054 Erlangen, Germany
| | | | | |
Collapse
|
18
|
Interferon γ and interleukin 6 modulate the susceptibility of macrophages to human immunodeficiency virus type 1 infection. Blood 2000. [DOI: 10.1182/blood.v96.9.3109.h8003109_3109_3117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The effect of interferon γ (IFN-γ) and interleukin 6 (IL-6) on infection of macrophages with human immunodeficiency virus type 1 (HIV-1) was investigated. By using a polymerase chain reaction–based viral entry assay and viral infectivity assay, it was demonstrated that IL-6 and IFN-γ augmented susceptibility of monocyte-derived macrophages (MDMs) to infection with T-cell tropic CXCR4-utilizing (X4) HIV-1 strains. Consistent with this finding, IFN-γ and IL-6 augmented fusion of MDMs with T-tropic envelope-expressing cells. The enhanced fusion of cytokine-treated MDMs with T-tropic envelopes was inhibited by the CXCR4 ligand, SDF-1, and by T22 peptide. IFN-γ and IL-6 did not affect expression of surface CXCR4 or SDF-1–induced Ca++ flux in MDMs. In contrast to the effect of IFN-γ on the infection of MDMs with X4 strains, IFN-γ inhibited viral entry and productive infection of MDMs with macrophage-tropic (M-tropic) HIV-1. Consistent with this finding, IFN-γ induced a decrease in fusion with M-tropic envelopes that correlated with a modest reduction in surface CCR5 and CD4 on MDMs. It was further demonstrated that macrophage inflammatory protein (MIP)-1α and MIP-β secreted by cytokine-treated MDMs augmented their fusion with T-tropic–expressing cells and inhibited their fusion with M-tropic envelope-expressing cells. These data indicate that proinflammatory cytokines, which are produced during opportunistic infections or sexually transmitted diseases, may predispose macrophages to infection with X4 strains that, in turn, could accelerate disease progression.
Collapse
|
19
|
Interferon γ and interleukin 6 modulate the susceptibility of macrophages to human immunodeficiency virus type 1 infection. Blood 2000. [DOI: 10.1182/blood.v96.9.3109] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
The effect of interferon γ (IFN-γ) and interleukin 6 (IL-6) on infection of macrophages with human immunodeficiency virus type 1 (HIV-1) was investigated. By using a polymerase chain reaction–based viral entry assay and viral infectivity assay, it was demonstrated that IL-6 and IFN-γ augmented susceptibility of monocyte-derived macrophages (MDMs) to infection with T-cell tropic CXCR4-utilizing (X4) HIV-1 strains. Consistent with this finding, IFN-γ and IL-6 augmented fusion of MDMs with T-tropic envelope-expressing cells. The enhanced fusion of cytokine-treated MDMs with T-tropic envelopes was inhibited by the CXCR4 ligand, SDF-1, and by T22 peptide. IFN-γ and IL-6 did not affect expression of surface CXCR4 or SDF-1–induced Ca++ flux in MDMs. In contrast to the effect of IFN-γ on the infection of MDMs with X4 strains, IFN-γ inhibited viral entry and productive infection of MDMs with macrophage-tropic (M-tropic) HIV-1. Consistent with this finding, IFN-γ induced a decrease in fusion with M-tropic envelopes that correlated with a modest reduction in surface CCR5 and CD4 on MDMs. It was further demonstrated that macrophage inflammatory protein (MIP)-1α and MIP-β secreted by cytokine-treated MDMs augmented their fusion with T-tropic–expressing cells and inhibited their fusion with M-tropic envelope-expressing cells. These data indicate that proinflammatory cytokines, which are produced during opportunistic infections or sexually transmitted diseases, may predispose macrophages to infection with X4 strains that, in turn, could accelerate disease progression.
Collapse
|
20
|
Vestal DJ, Gorbacheva VY, Sen GC. Different subcellular localizations for the related interferon-induced GTPases, MuGBP-1 and MuGBP-2: implications for different functions? J Interferon Cytokine Res 2000; 20:991-1000. [PMID: 11096456 DOI: 10.1089/10799900050198435] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The guanylate-binding proteins (GBPs) are a family of 65-67-kDa proteins induced by both type I and type II interferons (IFN). Members of the GBP family of GTPases are among the most abundant IFN-gamma-induced proteins. GBPs contain an unusual GTP binding site, which is consistent with GBP hydrolysis of GTP to both GDP and GMP. In addition, six of the eight known GBPs have a carboxy-terminal CaaX motif for the addition of isoprenyl lipids. Despite their abundance, however, little is known about the biologic function or cellular location of GBPs. We report here on studies to localize both a newly identified murine GBP (MuGBP-2) and its closely related family member, MuGBP-1. In both IFN-treated macrophages and fibroblasts, MuGBP-2 is found in both a granular distribution throughout the cytoplasm and localized to vesicle populations of heterogeneous sizes. The localization of MuGBP-2 to vesicles is dependent on its isoprenylation. Despite a high degree of sequence identity and the presence of an identical CaaX sequence, MuGBP-1 has a very homogeneous cytoplasmic distribution and fails to localize to intracellular vesicles. The different intracellular distribution of these two closely related family members suggests differential function(s).
Collapse
Affiliation(s)
- D J Vestal
- Department of Molecular Biology of the Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA.
| | | | | |
Collapse
|
21
|
Schindler C, Strehlow I. Cytokines and STAT signaling. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 1999; 47:113-74. [PMID: 10582086 DOI: 10.1016/s1054-3589(08)60111-8] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- C Schindler
- Department of Microbiology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | |
Collapse
|
22
|
Han Y, Rogers N, Ransohoff RM. Tumor necrosis factor-alpha signals to the IFN-gamma receptor complex to increase Stat1alpha activation. J Interferon Cytokine Res 1999; 19:731-40. [PMID: 10454343 DOI: 10.1089/107999099313578] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We describe a novel mechanism of signaling interaction through which tumor necrosis factor-alpha (TNF-alpha) treatment augments interferon-gamma (IFN-gamma)-induced Stat1alpha DNA-binding complexes and transcriptional activation of a Stat-binding element. In TNF-alpha-treated cells, IFN-gamma-induced phosphorylation of Jak2 kinase is increased, Jak2 kinase activity is enhanced, and genetic studies indicate that TNF-alpha requires Jak2 kinase activity to enhance Stat1alpha tyrosine phosphorylation. Increased Jak2 and Stat1alpha phosphorylation are observed within minutes of coexposure to TNF-alpha/IFN-gamma, suggesting a direct signaling interaction. IFN-gamma receptor chain 1 (IFNGR-1) tyrosine phosphorylation is markedly enhanced in cells treated with TNF-alpha/IFN-gamma without alteration in receptor levels. Thus, there exists a direct signaling interaction between TNF-alpha and IFN-gamma, independent of cooperating enhancer elements, that may be relevant for cytokine action during immune-mediated host defense and inflammatory processes.
Collapse
Affiliation(s)
- Y Han
- Department of Neurosciences, The Lerner Research Institute, Cleveland Clinic Foundation, OH 44195, USA
| | | | | |
Collapse
|
23
|
Saunders NA, Smith RJ, Jetten AM. Regulation of guanylate-binding protein expression in interferon-gamma-treated human epidermal keratinocytes and squamous cell carcinoma cells. J Invest Dermatol 1999; 112:977-83. [PMID: 10383748 DOI: 10.1046/j.1523-1747.1999.00611.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Interferon-gamma is a potent inducer of growth arrest and squamous differentiation of human epidermal keratinocytes in vitro. In order to understand the proximate events regulating interferon-gamma action we studied the relationship between interferon-gamma-mediated induction of a cytoplasmic guanylate-binding protein and the expression of growth and differentiation marker genes in normal and transformed keratinocytes. Induction of guanylate-binding protein mRNA by interferon-gamma was detectable at 4 h, was transcription dependent, and preceded changes in the expression of markers of growth arrest (E2F-1 mRNA downregulation) and differentiation (SQ37 mRNA induction). The Ec50 value for guanylate-binding protein induction (4 units interferon-gamma per ml) was lower than previously reported for SQ37 (40 units interferon-gamma per ml). Guanylate-binding protein mRNA appeared to be only moderately downregulated by modulators of the squamous phenotype such as retinoic acid and transforming growth factor-beta1. In addition, mRNA levels of E2F-1 or SQ37 were not altered in several squamous carcinoma cell lines treated with interferon-gamma. In contrast, guanylate-binding protein mRNA was highly induced in all these cell lines following interferon-gamma treatment. Further analysis of the signal transduction pathway mediating interferon-gamma responses using protein kinase inhibitors indicated that guanylate-binding protein induction in normal human epidermal keratinocyte cells was most likely protein kinase C independent. Our data suggest that more than one postreceptor interferon-gamma signaling pathway exists in keratinocytes and that at least one of these pathways is defective in squamous carcinoma cells. Furthermore, our data demonstrated that the failure of the squamous carcinoma cells to undergo interferon-gamma-induced growth arrest and differentiation is not due to an inherent defect in interferon-gamma receptor activation, but most likely is due to a defect in a non-guanylate-binding protein-dependent signaling pathway.
Collapse
Affiliation(s)
- N A Saunders
- Epithelial Pathobiology Group, Center For Immunology and Cancer Research, University of Queensland Department of Medicine, Princess Alexandra Hospital, Brisbane, Australia
| | | | | |
Collapse
|
24
|
Miller DM, Zhang Y, Rahill BM, Waldman WJ, Sedmak DD. Human Cytomegalovirus Inhibits IFN-α-Stimulated Antiviral and Immunoregulatory Responses by Blocking Multiple Levels of IFN-α Signal Transduction. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.10.6107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
The type I IFNs represent a primordial, tightly regulated defense system against acute viral infection. IFN-α confers resistance to viral infection by activating a conserved signal transduction pathway that up-regulates direct antiviral effectors and induces immunomodulatory activities. Given the critical role of IFN-α in anti-human cytomegalovirus (HCMV) immunity and the profound ability of HCMV to escape the host immune response, we hypothesized that HCMV blocks IFN-α-stimulated responses by disrupting multiple levels of the IFN-α signal transduction pathway. We demonstrate that HCMV inhibits IFN-α-stimulated MHC class I, IFN regulatory factor-1, MxA and 2′,5-oligoadenylate synthetase gene expression, transcription factor activation, and signaling in infected fibroblasts and endothelial cells by decreasing the expression of Janus kinase 1 and p48, two essential components of the IFN-α signal transduction pathway. This investigation is the first to report inhibition of type I IFN signaling by a herpesvirus. We propose that this novel immune escape mechanism is a major means by which HCMV is capable of escaping host immunity and establishing persistence.
Collapse
Affiliation(s)
- Daniel M. Miller
- Department of Pathology, Ohio State University, Columbus, OH 43210
| | - Yingxue Zhang
- Department of Pathology, Ohio State University, Columbus, OH 43210
| | - Brian M. Rahill
- Department of Pathology, Ohio State University, Columbus, OH 43210
| | - W. James Waldman
- Department of Pathology, Ohio State University, Columbus, OH 43210
| | - Daniel D. Sedmak
- Department of Pathology, Ohio State University, Columbus, OH 43210
| |
Collapse
|
25
|
Darnell JE. Studies of IFN-induced transcriptional activation uncover the Jak-Stat pathway. J Interferon Cytokine Res 1998; 18:549-54. [PMID: 9726434 DOI: 10.1089/jir.1998.18.549] [Citation(s) in RCA: 131] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Sharing the Milstein Award with George R. Stark and Ian M. Kerr in the fall of 1997 brought this invitation to record personal reflections on our experiments concerning the mechanisms of action of interferon. Our work and that of the Kerr and Stark laboratories uncovered the Jak-Stat pathway through which signals from cell surface receptors reach genes in the cell nucleus. This review concentrates on that to which I can speak most reliably, that is, experiments done by my young colleagues at Rockefeller University.
Collapse
Affiliation(s)
- J E Darnell
- The Rockefeller University, New York, NY 10021-6399, USA.
| |
Collapse
|
26
|
Lee CK, Bluyssen HA, Levy DE. Regulation of interferon-alpha responsiveness by the duration of Janus kinase activity. J Biol Chem 1997; 272:21872-7. [PMID: 9268319 DOI: 10.1074/jbc.272.35.21872] [Citation(s) in RCA: 78] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Daudi B lymphoblastoid cells are highly sensitive to the anti-growth and anti-viral effects of interferon (IFN). Unlike many cell lines, these cells show prolonged transcription of IFN-stimulated genes following treatment with IFN-alpha. This prolonged response correlated with the continued presence of the activated transcription factor, IFN-stimulated gene factor 3 (ISGF3). Pulse-chase labeling experiments indicated that the half-life of the phosphorylation of signal transducers and activators of transcription (Stat)1 and Stat2 was short (<2 h) although the turnover of the proteins themselves was slow (>24 h), indicative of a constitutive phosphatase activity. The administration of protein-tyrosine kinase inhibitors at any time point during IFN stimulation led to rapid inhibition of the response, indicating that tyrosine kinase activity was continuously required. Catalytic activity of Jak1 and Tyk2 kinases remained elevated for prolonged periods following stimulation. Continuous presence of IFN-alpha was necessary for maintaining prolonged activation of ISGF3 and of Janus kinases, an activity that was blocked by antibodies to IFN-alpha or by cycloheximide. Conditioned medium of IFN-alpha-stimulated cells was capable of stimulating STAT activation in naive cells. Taken together, these results suggest that the response to IFN-alpha is controlled by the duration of stimulated Janus kinase activity over the background of constitutive dephosphorylation and that this response can be sustained by autocrine secretion of IFN-alpha.
Collapse
Affiliation(s)
- C K Lee
- Department of Pathology and Kaplan Cancer Center, New York University School of Medicine, New York, New York 10016, USA
| | | | | |
Collapse
|
27
|
Grötzinger T, Jensen K, Will H. The interferon (IFN)-stimulated gene Sp100 promoter contains an IFN-gamma activation site and an imperfect IFN-stimulated response element which mediate type I IFN inducibility. J Biol Chem 1996; 271:25253-60. [PMID: 8810287 DOI: 10.1074/jbc.271.41.25253] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Expression of the nuclear domain-associated proteins Sp100, PML, and NDP52, is enhanced by interferons (IFNs) on the mRNA and protein level. Increase both of Sp100 and PML mRNA is due to enhanced transcription of the corresponding genes which occurs independently of cellular protein synthesis immediately upon IFN-beta addition. Here, we describe the molecular cloning and functional analysis of the Sp100 promoter. DNA sequence analysis revealed potential binding sites for several constitutive and IFN-inducible transcription factors. Consistent with the absence of a TATA box and an initiator element, several transcription initiation sites were found. Transient expression studies identified an imperfect IFN-stimulated response element within the first 100 nucleotides upstream of the major transcription start site. This element rendered a heterologous promoter IFN-beta-inducible and bound IFN-stimulated gene factor 2 strongly but IFN-stimulated gene factor 3 only weakly. An IFN-gamma activation site approximately 500 base pairs upstream of the IFN-stimulated response element was found to bind three IFN-alpha/beta activation factors upon IFN-beta induction and conferred both type I and type II IFN inducibility upon a heterologous promoter. These data demonstrate a novel arrangement of a nonoverlapping IFN-gamma activation site and an IFN-stimulated response element mediating type I IFN inducibility, previously not reported for other IFN-stimulable promoters.
Collapse
Affiliation(s)
- T Grötzinger
- Heinrich-Pette-Institut für experimentelle Virologie und Immunologie an der Universität Hamburg, 20251 Hamburg, Germany
| | | | | |
Collapse
|
28
|
Grötzinger T, Sternsdorf T, Jensen K, Will H. Interferon-modulated expression of genes encoding the nuclear-dot-associated proteins Sp100 and promyelocytic leukemia protein (PML). EUROPEAN JOURNAL OF BIOCHEMISTRY 1996; 238:554-60. [PMID: 8681971 DOI: 10.1111/j.1432-1033.1996.0554z.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Promyelocytic leukemia protein (PML) and Sp100 are transcription-regulatory proteins which colocalize in discrete nuclear dots and play a role in autoimmunity, oncogenesis and virus-host interaction. Interferons (IFNs) were shown previously to increase strongly the levels of Sp100 mRNA and protein. Here, we examined which mechanisms lead to upregulation of Sp100 gene expression and whether IFNs also increase expression of the promyelocytic leukemia (PML) gene. We found that both mRNA and protein levels of PML are also strongly upregulated by IFNs. In addition, new Sp100 and PML proteins were detected immunologically after IFN treatment of cells. Nuclear run-on analysis revealed protein-synthesis-independent, rapid IFN-enhanced transcription rates as well as synergistic activation of the Sp100 and PML genes by type-I and type-II IFNs. These data demonstrate that PML and Sp100 belong to the growing family of IFN-stimulated genes (ISGs) upregulated most likely by the transcription factor ISGF3, and indicate that IFNs also qualitatively alter the expression of these two genes.
Collapse
Affiliation(s)
- T Grötzinger
- Heinrich-Pette-Institut für experimentelle Virologie und Immunologie an der Universität Hamburg, Germany
| | | | | | | |
Collapse
|
29
|
Haque SJ, Flati V, Deb A, Williams BR. Roles of protein-tyrosine phosphatases in Stat1 alpha-mediated cell signaling. J Biol Chem 1995; 270:25709-14. [PMID: 7592750 DOI: 10.1074/jbc.270.43.25709] [Citation(s) in RCA: 83] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Different Stat proteins are activated through phosphorylation of unique tyrosine residues in response to different cytokines and growth factors. Interferon-gamma activates Stat1 molecules that form homodimers and bind cognate DNA elements. Here we show that treatment of permeabilized cells with 200-500 microM peroxo-derivatives of vanadium, molybdenum, and tungsten results in the accumulation of constitutively phosphorylated Stat1 alpha molecules. In contrast, treatment of permeabilized cells with orthovanadate, vanadyl sulfate, molybdate, and tungstate at the same range of concentrations does not result in the accumulation of activated Stat1 alpha molecules in the absence of ligand. However, these compounds inhibit the inactivation of interferon-gamma-induced DNA-binding activity of Stat1 alpha. A 4-6-h exposure of the permeabilized cells to orthovanadate, molybdate, and tungstate, but not vanadyl sulfate, results in a ligand-independent activation of Stat1 alpha, which is blocked by the inhibition or depletion of NADPH oxidase activity in the cells, indicating that NADPH oxidase-catalyzed superoxide formation is required for the bioconversion of these metal oxides to the corresponding peroxo-compounds. Interestingly, ligand-independent Stat1 alpha activation by peroxo-derivatives of these transition metals does not require Jak1, Jak2, or Tyk2 kinase activity, suggesting that other kinases can phosphorylate Stat1 alpha on tyrosine 701.
Collapse
Affiliation(s)
- S J Haque
- Department of Cancer Biology, Cleveland Clinic Foundation, Ohio 44195, USA
| | | | | | | |
Collapse
|
30
|
Zhou A, Chen Z, Rummage JA, Jiang H, Kolosov M, Kolosova I, Stewart CA, Leu RW. Exogenous interferon-gamma induces endogenous synthesis of interferon-alpha and -beta by murine macrophages for induction of nitric oxide synthase. J Interferon Cytokine Res 1995; 15:897-904. [PMID: 8564712 DOI: 10.1089/jir.1995.15.897] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Murine macrophages (M phi) are activated either by interferon-gamma (IFN-gamma) or interferon-alpha/beta (IFN-alpha/beta) in combination with bacterial lipopolysaccharide (LPS) to induce synthesis of tumor necrosis factor alpha (TNF-alpha) and nitric oxide synthase (iNOS) mRNA synthesis for generation of tumor cytotoxic nitric oxide (NO). In the present study, the effect of exogenous IFN-gamma on the induction of endogenous mRNA synthesis and secretion of IFN-alpha/beta by murine M phi was investigated. Neutralizing antibodies to IFN-alpha/beta reversed TNF-alpha and NOS mRNA synthesis, as well as nitric oxide (NO)-mediated tumor cytotoxicity. Quantitative reverse transcription polymerase chain reaction (RT-PCR) revealed that treatment of M phi with IFN-gamma induced increases in both IFN-alpha and IFN-beta mRNA synthesis by approximately 2-fold and 10-fold, respectively, which corresponded to a 2-fold increase in secretion of IFN-alpha/beta by ELISA. These data indicate that exogenous IFN-gamma induces endogenous synthesis and secretion of IFN-alpha/beta by M phi, which appears to act in concert with endogenously synthesized TNF-alpha for the autocrine induction of NOS mRNA synthesis.
Collapse
Affiliation(s)
- A Zhou
- Oklahoma Medical Research Foundation, Noble Center for Biomedical Research, Oklahoma City 73104-5046, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Several assays are available for interferons that either measure activity (bioassays) or protein mass (ELISAs). Bioassays generally require the use of viruses and some means of determining cell killing. Many investigators lack the expertise to work with potentially harmful viral agents and eschew this approach in favor of an ELISA assay based on a specific antibody or, to obtain the high level of sensitivity usually required, a combination of antibodies. Such immunological assays, while relatively easy, are expensive and detect protein mass which is not a reliable index of biological activity. I describe here a bioassay based on induction of a reporter gene linked to an interferon (IFN)-responsive promoter element. Production of the reporter gene product is dose-dependent in the range of 1 to approximately 100 U/ml of IFN and sensitivity is comparable to standard cytopathic effect assays. The assay can be modified to quantitate or detect other IFNs and could be applied to other cytokines.
Collapse
Affiliation(s)
- J A Lewis
- Department of Anatomy and Cell Biology, SUNY Health Science Center at Brooklyn 11203, USA
| |
Collapse
|
32
|
Wen Z, Zhong Z, Darnell JE. Maximal activation of transcription by Stat1 and Stat3 requires both tyrosine and serine phosphorylation. Cell 1995; 82:241-50. [PMID: 7543024 DOI: 10.1016/0092-8674(95)90311-9] [Citation(s) in RCA: 1605] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Stat1 and Stat3 are latent transcriptional factors activated initially through phosphorylation on single tyrosine residues induced by cytokine and growth factor occupation of cell surface receptors. Here we show that phosphorylation on a single serine (residue 727) in each protein is also required for maximal transcriptional activity. Both cytokines and growth factors are capable of inducing the serine phosphorylation of Stat1 and Stat3. These experiments show that gene activation by Stat1 and Stat3, which obligatorily require tyrosine phosphorylation to become active, also depends for maximal activation on one or more of the many serine kinases.
Collapse
Affiliation(s)
- Z Wen
- Laboratory of Molecular Cell Biology, Rockefeller University, New York, New York 10021-6399, USA
| | | | | |
Collapse
|
33
|
Hsu ML, Chen SW, Lin KH, Liao SK, Chang KS. Cytokine regulation of HIV-1 LTR transactivation in human hepatocellular carcinoma cell lines. Cancer Lett 1995; 94:41-8. [PMID: 7621443 DOI: 10.1016/0304-3835(95)03820-m] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Human hepatocellular carcinoma (HCC) cell lines, HEP-G2, J5, and SK-HEP-1, which differ in their differentiation status, were compared for their trans-activating activities after treatment with cytokines or 12-O-tetradecanoylphorbol-13-acetate (TPA). These cells were transfected with a long terminal repeat (LTR) which was derived from human immunodeficiency virus type 1 (HIV-1) and ligated to chloramphenicol acetyl transferase (CAT) gene. After treatment with interleukin-1 alpha (IL-1 alpha), interleukin-6 (IL-6), interferon-gamma (IFN-gamma), or TPA, they exhibited various degrees of enhancement of transactivation. The well differentiated HEP-G2 cells exhibited the highest degree of enhancement with these agents, while the poorly differentiated SK-HEP-1 cells showed no enhancement with cytokines and slight enhancement with TPA. The J5 cells, which were intermediate in their status of differentiation, showed a moderate degree of enhancement with cytokines and TPA. These results suggest that HCC cells at different stages of differentiation may produce different levels of cellular transacting factors activated by each of these agents. To map the cytokine response elements (CREs) in the HIV-1-LTR, HEP-G2 cells were transfected with nested series of 5' deletion mutants of HIV-1-LTR and treated with each of these cytokines. It was found that not only the degrees but also the patterns of enhancement varied depending upon the presence of positive or negative regulatory sequences in HIV-1-LTR, and that the NF-kappa B sequence played an important role, either by itself or in conjunction with the 5'-proximal response elements (REs) to interact with cellular trans-activating factors elicited by the cascade of transduction responses to cytokines. Despite the presence of promoters including kappa B and IFN-gamma RE as well as IL-6RE sequence in HIV-1-LTR-transfected cells, the poorly differentiated SK-HEP-1 cells showed no enhancement of transactivation by these cytokines, suggesting the lack of receptors or activity of some signal transduction factors which are present in well differentiated HEP-G2 and moderately differentiated J5 cells.
Collapse
Affiliation(s)
- M L Hsu
- Graduate Institute of Clinical Medicine, Chang Gung Medical College, Taoyuan-Hsien, Taiwan, Republic of China
| | | | | | | | | |
Collapse
|
34
|
Lanza L, Peirano L, Bosco O, Contini P, Filaci G, Setti M, Puppo F, Indiveri F, Scudeletti M. Interferons up-regulate with different potency HLA class I antigen expression in M14 human melanoma cell line. Possible interaction with glucocorticoid hormones. Cancer Immunol Immunother 1995; 41:23-8. [PMID: 7543821 PMCID: PMC11037655 DOI: 10.1007/bf01788956] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/1994] [Accepted: 03/28/1995] [Indexed: 01/25/2023]
Abstract
The relative potency of interferon alpha (IFN alpha), interferon beta (IFN beta), and interferon gamma (IFN gamma) in inducing the expression of HLA class I antigens, as well as their capacity to counteract the inhibition induced by glucocorticoid hormones on HLA class I antigen expression, were analysed in the human melanoma cell line M14, both at membrane and at mRNA level. The data obtained indicate that (a) IFN enhance with different potency (IFN gamma > IFN beta > IFN alpha) the expression of HLA class I antigens in M14 cells, (b) prednisone inhibits HLA class I antigen expression, (c) glucocorticoid hormones, when associated with IFN alpha or IFN gamma, inhibit the HLA class I enhancement induced by IFN alone, and, finally, (c) the association between 1 microM prednisone or 1 microM deflazacort and IFN beta seems to potentiate the enhancing capacity of IFN on the expression of HLA class I molecules at the mRNA level. These findings, if confirmed, might indicate that IFN and glucocorticoid hormones are not mutually exclusive in the management of human melanoma.
Collapse
Affiliation(s)
- L Lanza
- Department of Internal Medicine (D.I.M.I.), University of Genoa, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Ohta T, Ando O, Kurimoto M. Establishment of new interferon-gamma-resistant mutant cells with dominant phenotypes. J Interferon Cytokine Res 1995; 15:153-60. [PMID: 8590319 DOI: 10.1089/jir.1995.15.153] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
We established interferon-gamma-resistant (IGR) cells from a human colon adenocarcinoma cell line, LoVo. Their resistance was extremely high, and the ED50 values of IFN-gamma were > 10(5) IU/ml. Interestingly, although IGR-5 cells were still sensitive to the antiproliferative effect of IFN-alpha, the cells lost responsiveness to the antiviral effects of both IFN-alpha and gamma. Another clone, IGR-53, was unresponsive to both the antiproliferative and antiviral effects of either IFN-alpha or gamma. Furthermore, the IFN-gamma-resistant phenotypes of IGR cells were apparently dominant to the parental LoVo cells based on complementation tests. Although IGR-53 cells lack IFN-gamma receptors, IGR-5 cells seemed to have functional IFN-gamma receptors and processing mechanisms of IFN-gamma bound to the receptors. Northern analysis showed that IGR-5 cells responded to IFN-gamma and alpha, but the enhancement of IRF-1 expression by IFN-gamma was markedly suppressed.
Collapse
Affiliation(s)
- T Ohta
- Fujisaki Institute, Hayashibara Biochemical Laboratories, Inc., Okayama, Japan
| | | | | |
Collapse
|
36
|
Abstract
Interferons (IFNs) comprise a family of polypeptides that exhibit diverse biological effects such as inhibition of cell growth and protection against viral infection. These activities are based mainly on the transcriptional induction of cellular genes by both type I (IFN-alpha and IFN-beta) and type II (IFN-gamma) interferons. Several of these IFN-induced early response genes have been cloned and common elements within their promoters defined. Transcription factors, such as interferon-stimulated gene factor-3, IFN-gamma activation factor and FcRF gamma, that bind to these enhancers subsequently have been isolated and their components identified. This review shall provide an overview of the DNA response elements, the components of the IFN-induced transcription factors and their mechanism of action.
Collapse
Affiliation(s)
- M David
- Division of Cytokine Biology, Center of Biologics Evaluation and Research, Bethesda MD 20892, USA
| |
Collapse
|
37
|
Jaramillo ML, Abraham N, Bell JC. The interferon system: a review with emphasis on the role of PKR in growth control. Cancer Invest 1995; 13:327-38. [PMID: 7538039 DOI: 10.3109/07357909509094468] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- M L Jaramillo
- Department of Medicine, University of Ottawa, Ontario, Canada
| | | | | |
Collapse
|
38
|
Harroch S, Revel M, Chebath J. Interleukin-6 signaling via four transcription factors binding palindromic enhancers of different genes. J Biol Chem 1994. [DOI: 10.1016/s0021-9258(18)47177-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
39
|
Abstract
Recent studies on gene activation by interferons have uncovered a direct signaling pathway from the cell-surface receptor for interferons to the nucleus. Activation of JAK tyrosine kinases by interferons leads to activation, by direct tyrosine phosphorylation, of a family of latent cytoplasmic transcription factors named STATs (for signal transducers and activators of transcription), which then translocate to the nucleus to direct transcriptional activation.
Collapse
Affiliation(s)
- K Shuai
- Laboratory of Molecular Cell Biology, Rockefeller University, New York, NY 10021
| |
Collapse
|
40
|
Abstract
The redundancy of many cytokine functions that was first noted in vitro has now been confirmed in vivo with the demonstration that some of these functions can occur in mice rendered deficient in a cytokine or cytokine receptor by gene inactivation. Other functions are ablated in these mice, suggesting that they are cytokine-specific. Although some of the underlying mechanisms have been identified, it remains unclear why certain activities of a cytokine should be unique and others redundant. It is proposed that compensatory mechanisms exist only for those cytokine functions whose inappropriate or excessive activation would not be pathogenic or whose importance justifies this risk. Conversely, cytokine-specific functions might be those that should be tightly regulated to avoid the pathological consequences of their inadvertent expression.
Collapse
Affiliation(s)
- A Kelso
- Transplantation Biology Program, Queensland Institute of Medical Research, Brisbane, Australia
| |
Collapse
|
41
|
Ivashkiv LB, Ayres A, Glimcher LH. Inhibition of IFN-gamma induction of class II MHC genes by cAMP and prostaglandins. IMMUNOPHARMACOLOGY 1994; 27:67-77. [PMID: 8206755 DOI: 10.1016/0162-3109(94)90008-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Triggering of the cyclic AMP (cAMP) signal transduction pathway inhibits the the interferon gamma (IFN-gamma)-mediated induction of class II major histocompatibility (MHC) genes. We have investigated the mechanism of the inhibition of IFN-gamma induction of the murine A alpha class II MHC gene by cAMP and E series prostaglandins (PGEs). 151 base pairs of the A alpha promoter were sufficient to confer positive regulation by IFN-gamma and negative regulation by cAMP which accurately mirrored the regulation of the endogenous A alpha gene. cAMP also inhibited the IFN-gamma activation of the Fc gamma receptor I (Fc gamma RI) gene promoter, an "early" promoter which is activated immediately after treatment of cells with IFN-gamma. PGEs, which cause an elevation in intracellular cAMP, inhibited the induction of the A alpha promoter, and inhibition was greater in the presence of tumor necrosis factor alpha (TNF alpha). A mutational analysis of the A alpha promoter showed that all four conserved class II promoter elements, the S, X1, X2, and Y boxes, play a role in mediating A alpha promoter activation by IFN-gamma. Mutations in these elements did not diminish the cAMP inhibition of promoter activation by IFN-gamma. Thus, conserved class II promoter sequences which mediate most known examples of positive and negative regulation, including cAMP inhibition of constitutive class II expression, do not mediate cAMP inhibition of IFN-gamma activation of the A alpha promoter. We suggest that this inhibition may be mediated by a novel class II promoter element or by disruption of an early step in the IFN-gamma signal transduction pathway.
Collapse
Affiliation(s)
- L B Ivashkiv
- Department of Cancer Biology, Harvard School of Public Health, Boston 02115
| | | | | |
Collapse
|
42
|
Wadler S, Schwartz EL. Biologic agents as biochemical modulators: pharmacologic basis for the interaction of cytotoxic chemotherapeutic drugs and interferon. Cancer Chemother Pharmacol 1994; 35:21-30. [PMID: 7527304 DOI: 10.1007/bf00686280] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Biochemical modulation of cytotoxic cancer chemotherapeutic agents is one means of enhancing the activity and selectivity of antitumor drugs. Traditionally this approach has utilized detailed information regarding a particular enzymatic reaction or biochemical pathway to develop potential modulating agents. In contrast, the reported clinical therapeutic activity of IFN in combination with cytotoxic agents has prompted a reexamination of the biochemical actions of the cytokine. Interferon elicits a number of cellular actions that might contribute to its pharmacologic activity, including both direct antitumor effects and host-mediated actions. The best understood are those related to the cytotoxicity of the fluoropyrimidine antimetabolites and include enzymatic reactions involved in fluoropyrimidine metabolic activation, catabolism, and interaction with its target enzyme. However, even in this instance, a mechanistic association of a specific pharmacologic action with therapeutic activity remains to be determined. These studies demonstrate that cytokines and other biologic agents may exert specific biochemical modulations that augment (or potentially attenuate) the activity of the cytotoxic chemotherapeutic agents.
Collapse
Affiliation(s)
- S Wadler
- Albert Einstein Cancer Center, Montefiore Medical Center, Bronx, NY 10467
| | | |
Collapse
|
43
|
Tóth FD, Mosborg-Petersen P, Kiss J, Aboagye-Mathiesen G, Zdravkovic M, Hager H, Ebbesen P. Differential replication of human immunodeficiency virus type 1 in CD8- and CD8+ subsets of natural killer cells: relationship to cytokine production pattern. J Virol 1993; 67:5879-88. [PMID: 8371348 PMCID: PMC238007 DOI: 10.1128/jvi.67.10.5879-5888.1993] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
CD8+ and CD8- subsets of peripheral blood natural killer (NK) cells were examined for susceptibility to infection with human immunodeficiency virus type 1 (HIV-1) and for the ability to produce various types of interferon (IFN) and tumor necrosis factor (TNF). HIV-1 was preferentially grown in CD8+ NK cells. The ability of CD8- NK cells to suppress HIV-1 replication was related to their ability to produce alpha IFN (IFN-alpha) upon viral induction. Induction with interleukin-2 resulted in IFN-gamma production in both subsets of NK cells. In the CD8+ subset, IFN-gamma and HIV-1 mutually enhanced the production of TNF alpha, leading to hyperactivation of viral replication, whereas in CD8- NK cells IFN-gamma primed HIV-induced IFN-alpha production. The dichotomous effects of IFN-gamma on HIV-1 replication were dependent on the IFN-alpha-producing ability of the cellular targets. These findings can explain the selective depletion of the CD16+ CD8+ subset that begins early in the in vivo HIV-1 infection.
Collapse
Affiliation(s)
- F D Tóth
- Danish Cancer Society, Department of Virus and Cancer, Aarhus
| | | | | | | | | | | | | |
Collapse
|
44
|
Ozes ON, Taylor MW. Reversal of an interferon-gamma-resistant phenotype by poly(I:C): possible role of double-stranded RNA-activated kinase in interferon-gamma signaling. JOURNAL OF INTERFERON RESEARCH 1993; 13:283-8. [PMID: 7901291 DOI: 10.1089/jir.1993.13.283] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Indoleamine 2,3-dioxygenase (IDO) is induced in neoplastic cell lines by interferon-gamma (IFN-gamma) treatment. In ME180 cervical carcinoma cells, there is a rapid increase in IDO mRNA accumulation beginning at 4 h after IFN-gamma treatment and continuing for at least 24 h. The IFN-gamma-resistant mutant of ME180, IR3B6B, expresses very low levels of IDO message after IFN-gamma treatment. However, pretreatment of this mutant with poly(I:C) restores normal levels of IDO mRNAs and IDO enzyme activity. Poly(I:C) mediated reversal of the IFN-gamma-resistant phenotype and induction of IDO mRNA are inhibited by 2-aminopurine. In vitro phosphorylation of calf thymus histone using the immunoprecipitated p68 kinase prepared from IFN-gamma-treated ME180 and IR3B6B cells revealed the deficiency of activation of this kinase in IR3B6B cells after IFN-gamma treatment, and treatment of this mutant cells with poly(I:C) restores p68 kinase activity. From these results, we conclude that a double-stranded RNA-dependent kinase is activated by IFN-gamma treatment and its activation correlates with IFN-gamma-mediated induction of the IDO gene.
Collapse
Affiliation(s)
- O N Ozes
- Department of Biology, Indiana University, Bloomington 47405
| | | |
Collapse
|
45
|
Radford JE, Waring JF, Pohlman JK, Ginder GD. Stimulation of MHC class I transcription by interferon-gamma involves a non-A, non-C kinase in addition to protein kinase C. JOURNAL OF INTERFERON RESEARCH 1993; 13:133-41. [PMID: 8099599 DOI: 10.1089/jir.1993.13.133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The signal pathways by which interferon-gamma (IFN-gamma) is able to up-regulate major histocompatibility complex (MHC) class I transcription were studied in two human hematopoietic tumor cell lines, K562 and Ramos. These studies suggest that the IFN-gamma signal is transduced via an H7- and staurosporine-sensitive kinase that is distinct from protein kinase C (PKC) and protein kinase A (PKA) in both cell types. Ramos cells appear to utilize an additional pathway involving double-stranded RNA-dependent protein kinase. PKC and possibly PKA appear to be involved in one or more intersecting pathways by which agonists of these kinases are able to act synergistically with IFN-gamma, but activation of these latter pathways is neither necessary nor sufficient for induction of MHC class I transcription. Modulation of G-protein- and Ca2+-calmodulin-associated pathways and arachidonic acid metabolism had no effect on constitutive or IFN-gamma-stimulated class I transcription. The class I stimulatory factor produced in response to IFN-gamma treatment appears to have a short t1/2. The identity of this factor is unknown, but is likely to be distinct from known mediators of IFN-stimulated transcription. Gene and cell-type specificity in the signal transduction pathways utilized by IFN-gamma implies that such pathways may be useful targets for experimental and therapeutic manipulation.
Collapse
Affiliation(s)
- J E Radford
- Department of Medicine, University of Minnesota, Minneapolis 55455
| | | | | | | |
Collapse
|
46
|
Affiliation(s)
- G C Sen
- Department of Molecular Biology, Cleveland Clinic Foundation, Ohio 44195-5285
| | | |
Collapse
|
47
|
Decker T. Double-stranded RNA and interferon-alpha induce transcription through different molecular mechanisms. JOURNAL OF INTERFERON RESEARCH 1992; 12:445-8. [PMID: 1289412 DOI: 10.1089/jir.1992.12.445] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Double-stranded (ds) RNA stimulates the synthesis of several mRNAs known to be induced by type I interferons (IFNs). In this report, it is shown that the IFN-alpha stimulated genes (ISGs) 15, 54, 56, and GBP are transcriptionally induced by dsRNA. Transcriptional stimulation occurred in the presence of the protein synthesis inhibitor cycloheximide (CHX), indicating that inducibility was directly mediated by dsRNA through the action of preformed proteins. ISGF-3, the protein complex mediating primary transcriptional induction of ISGs by IFN-alpha, was not activated by dsRNA in the presence of CHX. Additionally, DNA-binding activity of ISGF-2/IRF-1, a protein involved in the regulation of the IFN-beta gene and ISGs, did not correlate with dsRNA-induced transcriptional induction of ISGs. This suggests that dsRNA and IFN-alpha induce ISGs through different molecular mechanisms.
Collapse
Affiliation(s)
- T Decker
- Fraunhofer Institute for Toxicology and Molecular Biology, Hannover, Germany
| |
Collapse
|
48
|
Lembo D, Gaboli M, Caliendo A, Falciani F, Garattini E, Landolfo S. Regulation of the 202 gene expression by interferons in L929 cells. Biochem Biophys Res Commun 1992; 187:628-34. [PMID: 1382418 DOI: 10.1016/0006-291x(92)91241-h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Type I and II interferons (IFNs) stimulate the expression of the 202 and 2'-5' oligoadenylate synthetase (OASE) genes in L929, NIH 3T3 and LM-TK- fibroblastic cell lines. In two other cell lines, B16 melanoma and F9 teratocarcinoma, these cytokines induce OASE but not the 202 mRNA. In L929 cells, IFN-alpha induces the 202 mRNA at concentrations between 10 and 10(3) units/ml. To achieve maximal induction of the 202 mRNA, continuous exposure of L929 cells to IFN-alpha is necessary, whereas 30 minutes of exposure are sufficient to trigger maximal upregulation of the OASE transcript. The induction of the 202 mRNA is the consequence of both transcriptional and post-transcriptional events. Cycloheximide, a known inhibitor of protein synthesis, does not block the induction of 202 mRNA by IFN-alpha, demonstrating that new protein synthesis is not required for this effect. Protein kinase C, arachidonic acid metabolism via the cyclooxygenase or the lipoxygenase pathways and cAMP are not involved as second messengers in the induction of the 202 mRNA by IFN-alpha in L929 cells.
Collapse
Affiliation(s)
- D Lembo
- Istituto di Microbiologia, Facolta di Medicina, Università degli Studi di Torino, Italy
| | | | | | | | | | | |
Collapse
|
49
|
Nakagawa Y, Petricoin EF, Akai H, Grimley PM, Rupp B, Larner AC. Interferon-alpha-induced gene expression: evidence for a selective effect of ouabain on activation of the ISGF3 transcription complex. Virology 1992; 190:210-20. [PMID: 1529530 DOI: 10.1016/0042-6822(92)91207-b] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Binding of interferons (IFNs) to their cell surface receptors stimulates rapid translocation of cytoplasmic proteins to the nucleus and the expression of a variety of cellular genes within minutes. Translocated proteins subsequently bind to the interferon-stimulated response element (ISRE) located in the promoters of all IFN-activated cellular genes. We report here that ouabain, a specific inhibitor of the Na/K ATPase, selectively inhibited transcription of several IFN-alpha-induced cellular RNAs under conditions in which some other well-described signal transduction pathways remained intact. The latter included induction of human metallothionein 2A (HMT2A) by phorbol ester and induction of IP-10 RNA by IFN-gamma. Ouabain itself induced RNA of the protooncogene c-fos which conversely was inhibited by IFN-alpha. Specificity of the ouabain effects on IFN alpha-induced RNAs with respect to a direct action on the Na/K ATPase was shown with a transfected monkey CV-1 cell line which expresses the ouabain-insensitive rat alpha 1 subunit. Electrophoretic mobility shift assays (EMSAs) using nuclear extracts from ouabain-treated cells demonstrated that ouabain decreased IFN alpha-induced binding of the ISGF3 complex to the ISRE. Reconstitution experiments showed that this effect of ouabain is not due to the inhibition of IFN alpha activation of the ISGF3 alpha subcomponent, which occurs in the cytoplasm, but a selective depletion of the ISGF3 gamma factor which in concert with activated ISGF3 alpha induces interferon-stimulated gene (54 kDa) transcription. These findings imply that intracellular ion balance can selectively regulate the half-life of the ISGF3 gamma protein or the ability of this protein to complex with ISGF3 alpha to activate IFN alpha-regulated cellular genes.
Collapse
Affiliation(s)
- Y Nakagawa
- Laboratory of Cytokine Research, National Institutes of Health, Bethesda, Maryland 20892
| | | | | | | | | | | |
Collapse
|
50
|
Velazquez L, Fellous M, Stark GR, Pellegrini S. A protein tyrosine kinase in the interferon alpha/beta signaling pathway. Cell 1992; 70:313-22. [PMID: 1386289 DOI: 10.1016/0092-8674(92)90105-l] [Citation(s) in RCA: 708] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The mutant human cell line 11.1 is unresponsive to interferon alpha. Here we describe the genetic complementation of this mutant and the identification and cloning of the wild-type gene that corrects the defect. Using transfection with genomic DNA in conjunction with a powerful back-selection, we isolated a cosmid that reverts the mutant phenotype of 11.1 cells. The cosmid encodes a single message whose level is greatly reduced in mutant cells. Complementary DNAs were cloned and found to be virtually identical to tyk2, a human mRNA encoding a non-receptor protein tyrosine kinase of previously unknown function. This finding shows that tyk2 links the interferon alpha/beta receptor to the cytoplasmic transcription factor that mediates activation of interferon-responsive genes.
Collapse
Affiliation(s)
- L Velazquez
- Unité INSERM 276, Institut Pasteur, Paris, France
| | | | | | | |
Collapse
|