1101
|
Cong Z, Wan M, Wu X, Wang L, Hu X, Yang F, Bao M, Zhang X, Chen J, Wang L, Yu Y. A CpG oligodeoxynucleotide inducing anti-coxsackie B3 virus activity in human peripheral blood mononuclear cells. FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY 2007; 51:26-34. [PMID: 17608709 PMCID: PMC7110351 DOI: 10.1111/j.1574-695x.2007.00266.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Revised: 04/09/2007] [Accepted: 04/10/2007] [Indexed: 11/30/2022]
Abstract
Coxsackie B3 virus (CVB3) is the most significant pathogen causing myocarditis in humans, and antiviral therapy would be most effective in the early stages of the disease. Here we provide evidence that BW001, a C-type CpG oligodeoxynucleotide, induces anti-CVB3 activity in human peripheral blood mononuclear cells (PBMCs). In parallel, we have demonstrated that BW001 induces human PBMCs to express mRNAs of multiple types of interferon (IFN), including IFN-alpha, IFN-beta, IFN-omega and IFN-gamma, and to express mRNAs of at least 11 subtypes of IFN-alpha. The induced IFNs may contribute to the anti-CVB3 activity. The results suggest that BW001 could be developed into a medication with the potential to treat CVB3 infectious diseases by inducing natural mixed IFNs.
Collapse
Affiliation(s)
- Zhongyi Cong
- Department of Immunology, College of Basic Medicine, Jilin University, Changchun, China
| | - Min Wan
- Department of Molecular Biology, College of Basic Medicine, Jilin University, Changchun, China
| | - Xiuli Wu
- Department of Molecular Biology, College of Basic Medicine, Jilin University, Changchun, China
| | - Li Wang
- Department of Immunology, College of Basic Medicine, Jilin University, Changchun, China
| | - Xiaoping Hu
- Department of Molecular Biology, College of Basic Medicine, Jilin University, Changchun, China
| | - Fenglei Yang
- Department of Immunology, College of Basic Medicine, Jilin University, Changchun, China
| | - Musheng Bao
- Department of Molecular Biology, College of Basic Medicine, Jilin University, Changchun, China
| | - Xuesong Zhang
- The Blood Center of Jilin Province, Changchun, China
| | - Jianzhu Chen
- Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Liying Wang
- Department of Molecular Biology, College of Basic Medicine, Jilin University, Changchun, China
| | - Yongli Yu
- Department of Immunology, College of Basic Medicine, Jilin University, Changchun, China
| |
Collapse
|
1102
|
|
1103
|
Chen Y, Antoniou E, Liu Z, Hearne LB, Roberts RM. A microarray analysis for genes regulated by interferon-tau in ovine luminal epithelial cells. Reproduction 2007; 134:123-35. [PMID: 17641094 DOI: 10.1530/rep-07-0387] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Interferon-tau (IFNT) is released by preimplantation conceptuses of ruminant species and prepares the mother for pregnancy. Although one important function is to protect the corpus luteum from the luteolytic activity of prostaglandin-F 2alpha, IFNT most likely regulates a range of other physiological processes in endometrium. Here, an immortalized cell line from ovine uterine luminal epithelial cells was treated with IFNT for either 8 or 24 h. RNA was subjected to cDNA microarray analysis, with RNA from untreated cells as the reference standard. Of 15 634 genes, 1274 (8%) were IFNT responsive at P<0.01 and 585 at P<0.001 to at least one treatment. Of the latter, 356 were up-regulated and 229 down-regulated. Increasing IFNT concentrations from 10 ng/ml to 10 microg/ml had minor effects, and most genes up- or down-regulated at 8 h were regulated similarly at 24 h. Although IFNT influences many genes implicated in antiviral activity and apoptosis, its action also likely regulates prostaglandin metabolism, growth factors and their receptors, apoptosis and the nuclear factor (NF)-kappaB cascade, extracellular matrix accretion, angiogenesis, blood coagulation, and inflammation. In particular, it increased mRNA concentrations of genes related to the vascular endothelial growth factor R2 pathway of angiogenesis and down-regulated ones associated with hypoxia. Two genes implicated in the antiluteolytic actions of IFNT (encoding cyclooxygenase-2 and the oxytocin receptor respectively) were down-regulated in response to all treatments. IFNT targets a complex range of physiological processes during the establishment of pregnancy.
Collapse
Affiliation(s)
- Yizhen Chen
- Division of Animal Sciences, University of Missouri-Columbia, Missouri 65211, USA
| | | | | | | | | |
Collapse
|
1104
|
Schäfer H, Kliem G, Kropp B, Burger R. Monoclonal antibodies to guinea pig interferon-gamma: tools for cytokine detection and neutralization. J Immunol Methods 2007; 328:106-17. [PMID: 17905302 DOI: 10.1016/j.jim.2007.08.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2007] [Revised: 07/19/2007] [Accepted: 08/22/2007] [Indexed: 11/26/2022]
Abstract
We have generated polyclonal antisera and monoclonal antibodies against recombinant guinea pig IFN-gamma. These antibodies were used to inhibit the function of IFN-gamma in vitro and to establish a capture ELISA system for the detection and quantitation of this cytokine. Although recombinant protein expressed in E. coli was available in abundance, it was only of limited value to develop a capture ELISA which detects the native cytokine, since only a limited number of monoclonal antibodies reacted both with the recombinant and the native protein. Positive test results in an initial ELISA setup with recombinant IFN-gamma were not predictive for the detection of IFN-gamma from activated T-lymphocytes in the same assay. After evaluating several different combinations of rabbit antisera and monoclonal antibodies, an assay system was established which uses two mouse monoclonal antibodies as capture and detecting reagents. Three of the monoclonal antibodies and the rabbit antisera were able to block the function of guinea pig IFN-gamma when assayed in a luciferase reporter assay.
Collapse
Affiliation(s)
- H Schäfer
- Cellular Immunology, Robert Koch Institute, Berlin, Germany.
| | | | | | | |
Collapse
|
1105
|
Williams AS, Richards PJ, Thomas E, Carty S, Nowell MA, Goodfellow RM, Dent CM, Williams BD, Jones SA, Topley N. Interferon-gamma protects against the development of structural damage in experimental arthritis by regulating polymorphonuclear neutrophil influx into diseased joints. ACTA ACUST UNITED AC 2007; 56:2244-54. [PMID: 17599735 DOI: 10.1002/art.22732] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Local interaction between soluble mediators within the inflamed synovium is a key factor that governs the pathologic outcome of inflammatory arthritides. Our aim was to investigate the interplay between the Th1 lymphokine interferon-gamma (IFNgamma) and pivotal cytokines that drive rheumatoid arthritis (RA) pathology (interleukin-1beta [IL-1beta] and tumor necrosis factor alpha [TNFalpha]) in modulating inflammation and arthritis in vitro and in vivo. METHODS Monarticular antigen-induced arthritis (AIA) was initiated in IFNgamma-deficient (IFNgamma(-/-)) mice and age-matched wild-type (IFNgamma(+/+)) mice. Joint swelling was measured and histologic analysis was performed in order to assess changes in both inflammatory and degenerative parameters in vivo. In vitro, the influence of IFNgamma in regulating IL-1beta- and TNFalpha-driven CXCL8 and CCL2 production was quantified by enzyme-linked immunosorbent assay. RESULTS In murine AIA, both inflammatory and degenerative arthritis parameters were significantly exacerbated in the absence of IFNgamma. IFNgamma appeared to be a crucial factor in regulating CXCR2+ neutrophil influx in the joint. In in vitro studies using RA fibroblast-like synoviocytes, IFNgamma modulated both IL-1beta- and TNFalpha-driven chemokine synthesis, resulting in the down-regulation of CXCL8 production. CONCLUSION IFNgamma exerts antiinflammatory, chondroprotective, and antiosteoclastogenic effects in murine AIA through a mechanism that involves the regulation of chemokine synthesis and local neutrophil recruitment. These studies suggest a potential therapeutic role of modulating IFNgamma signaling in the treatment of inflammatory arthritides.
Collapse
Affiliation(s)
- Anwen S Williams
- Wales College of Medicine, Cardiff University, Heath Park, Cardiff, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1106
|
Abstract
BACKGROUND Since the discovery of the hepatitis C virus (HCV), extensive literature has emerged on alcohol and HCV interaction. GOAL To understand the impact of alcohol and HCV infection on the severity of liver disease and the mechanisms of interaction between the two. STUDY Of 1269 articles (1991 to 2006) searched through MEDLINE and cited cross references, 133 were thoroughly reviewed to assess: (a) prevalence of combined alcohol use and HCV, (b) severity of liver disease (c) treatment response, and (d) mechanisms of interaction between HCV and alcohol. Data on study design, patient demographics, diagnostic tests used, and study outcomes were extracted for critical analysis. RESULTS Prevalence of HCV is 3-fold to 30-fold higher in alcoholics compared with the general population. Patients with HCV infection and alcohol abuse develop more severe fibrosis with higher rate of cirrhosis and hepatocellular cancer compared with nondrinkers. Increased oxidative stress seems to be the dominant mechanism for this synergism between alcohol and the HCV. Abstinence is the key to the management of liver disease due to HCV and alcohol. Data have shown that lower response rates to interferon in alcoholics with HCV infection are likely due to noncompliance. CONCLUSIONS Alcoholics with HCV infection have more severe liver disease compared with nondrinkers. Patients should be encouraged to enroll in rehabilitation programs so as to improve treatment adherence and response.
Collapse
Affiliation(s)
- Ashwani K Singal
- James J Peters Bronx Veterans Affairs Medical Center, Department of Medicine, Mount Sinai School of Medicine, New York, NY 10468, USA.
| | | |
Collapse
|
1107
|
Ward CM, Jyonouchi H, Kotenko SV, Smirnov SV, Patel R, Aguila H, McSherry G, Dashefsky B, Holland SM. Adjunctive treatment of disseminated Mycobacterium avium complex infection with interferon alpha-2b in a patient with complete interferon-gamma receptor R1 deficiency. Eur J Pediatr 2007; 166:981-5. [PMID: 17120031 DOI: 10.1007/s00431-006-0339-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2006] [Revised: 10/09/2006] [Accepted: 10/09/2006] [Indexed: 01/14/2023]
Abstract
We report adjunct treatment of (interferon) IFN-alpha2b (Intron-A) in a patient with complete interferon-gamma receptor R1 (IFNGR1) deficiency suffering from disseminated infection with Mycobacterium avium complex (MAC) resistant to multiple anti-mycobacterial agents. A low dose of IFN-alpha2b (3 x 10(6) units/m(2) three times weekly subcutaneously) successfully attenuated progressive hepatosplenomegaly and abdominal/retroperitoneal/pelvic lymphadenopathy, although the patient continued to be mycobacteremic. This is the first report of a complete IFNGR1 deficiency treated with adjuvant IFN-alpha2b for disseminated MAC infection.
Collapse
Affiliation(s)
- Carla M Ward
- Division of Pulmonology, Allergy/Immunology, and Infectious Diseases, Department of Pediatrics, New Jersey Medical School/UMDNJ, 185 South Orange Avenue, Newark, NJ 07101-1709, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
1108
|
Jia D, Rahbar R, Fish EN. Interferon-inducible Stat2 activation of JUND and CLDN4: mediators of IFN responses. J Interferon Cytokine Res 2007; 27:559-65. [PMID: 17651017 DOI: 10.1089/jir.2007.0014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Signal transducer and activator of transcription 2 (Stat2) is a critical signaling protein involved in mediating interferon-alpha/beta (IFN-alpha/beta) responses. Using site-directed mutagenesis in conjunction with gene microarray and biologic studies, we have previously demonstrated that in addition to Stat2 functioning as a transactivator of transcription of a subset of IFN-inducible genes (ISG), Stat2-DNA binding mediates the transcriptional activation of other ISGs required for IFN-inducible antiviral and growth inhibitory responses. Among these, two candidate genes identified were Jun-D (JUND) and claudin-4 (CLDN4). To further explore the role of JUND and CLDN4 in IFN responses, we conducted knockdown studies using siRNA specific for either JUND or CLDN4 in 2fTGH fibroblast cells, and consistent with cells expressing the DNA-binding mutant of Stat2 (U6A-2VV-II), siRNA-mediated knockdown resulted in cells that exhibited reduced antiproliferative and antiviral responses to IFN. Our data suggest that JUND and CLDN4 are critical mediators of the antiproliferative and antiviral effects of type I IFNs and further confirm the functional importance of the DNA-binding domain of Stat2.
Collapse
Affiliation(s)
- Danlin Jia
- Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, Department of Immunology, University of Toronto, Toronto, Canada
| | | | | |
Collapse
|
1109
|
Tailor P, Tamura T, Kong HJ, Kubota T, Kubota M, Borghi P, Gabriele L, Ozato K. The feedback phase of type I interferon induction in dendritic cells requires interferon regulatory factor 8. Immunity 2007; 27:228-39. [PMID: 17702615 PMCID: PMC2768351 DOI: 10.1016/j.immuni.2007.06.009] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Revised: 04/03/2007] [Accepted: 06/11/2007] [Indexed: 12/16/2022]
Abstract
Dendritic cells (DCs) produce type I interferons (IFNs) in greater amounts than other cells, but the mechanisms remain elusive. Here we studied the role of a transcription factor, IRF8, in DC induction of type I IFNs. Upon newcastle disease virus (NDV) infection, bone marrow-derived plasmacytoid and conventional DCs induced IFN transcripts, exhibiting two-phase kinetics. The second, amplifying phase represented an IFN feedback response that accounted for much of IFN protein production. Induction of second phase transcription required IRF8. Mouse cytomegalovirus (MCMV) and Toll-like receptor-mediated IFN induction in DCs also required IRF8. Chromatin immunoprecipitation analysis showed that IRF7, IRF8, and RNA polymerase II were recruited to the IFN promoters upon stimulation. Moreover, sustained RNA polymerase II recruitment to the promoters critically depended on IRF8. Together, these data indicate that IRF8 magnifies the second phase of IFN transcription in DCs by prolonging binding of basic transcription machinery to the IFN promoters, thereby playing a role in innate immunity.
Collapse
Affiliation(s)
- Prafullakumar Tailor
- Laboratory of Molecular Growth Regulation, National Institute of Child Health and Human Development, &National Institutes of Health, Bethesda MD 20892
| | - Tomohiko Tamura
- Laboratory of Molecular Growth Regulation, National Institute of Child Health and Human Development, &National Institutes of Health, Bethesda MD 20892
| | - Hee Jeong Kong
- Laboratory of Molecular Growth Regulation, National Institute of Child Health and Human Development, &National Institutes of Health, Bethesda MD 20892
| | - Toru Kubota
- Laboratory of Molecular Growth Regulation, National Institute of Child Health and Human Development, &National Institutes of Health, Bethesda MD 20892
| | - Mayumi Kubota
- Laboratory of Molecular Growth Regulation, National Institute of Child Health and Human Development, &National Institutes of Health, Bethesda MD 20892
| | - Paola Borghi
- Laboratory of Virology, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Gabriele
- Laboratory of Virology, Istituto Superiore di Sanità, Rome, Italy
| | - Keiko Ozato
- Laboratory of Molecular Growth Regulation, National Institute of Child Health and Human Development, &National Institutes of Health, Bethesda MD 20892
- Corresponding author: Laboratory of Molecular Growth Regulation, National Institute of Child Health and Human Development, National Institutes of Health, Room 2A01, Building 6, 6 Center Drive, Bethesda MD 20892-2753, TEL: (301) 496-9184 Fax (301) 402-2974,
| |
Collapse
|
1110
|
Gajendran N, Mittrücker HW, Bordasch K, Heinemann E, Koch M, Kaufmann SHE. Regional IFNgamma expression is insufficient for efficacious control of food-borne bacterial pathogens at the gut epithelial barrier. Int Immunol 2007; 19:1075-81. [PMID: 17698562 DOI: 10.1093/intimm/dxm075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
IFNgamma is critical for host defence against various food-borne pathogens including Salmonella enterica and Listeria monocytogenes, the causative agents of salmonellosis and listeriosis, respectively. We investigated the impact of regional IFNgamma expression at the intestinal epithelial barrier on host invasion by salmonellae and listeriae following oral challenge. Transgenic mice (IFNgamma-gut), generated on an IFNgamma knock-out (KO) background, selectively expressed IFNgamma in the gut driven by the modified liver fatty acid-binding protein (Fabpl(4x at -132)) promoter. Infections with attenuated S. enterica Typhimurium or with L. monocytogenes did not differ significantly in IFNgamma-KO, IFNgamma-gut and wild-type mice. Further, Listeria-specific CD4+ and CD8+ T cells were not altered in IFNgamma-gut mice. Thus, this model indicates that local IFNgamma expression by non-immunological cells in the distal part of the small intestine, caecum and colon is insufficient for prevention of gut penetration by S. enterica Typhimurium and L. monocytogenes.
Collapse
Affiliation(s)
- Nadesan Gajendran
- Max Planck Institute for Infection Biology, Department of Immunology, Charitéplatz 1, Berlin, 10117 Germany
| | | | | | | | | | | |
Collapse
|
1111
|
Zhao W, Cha EN, Lee C, Park CY, Schindler C. Stat2-dependent regulation of MHC class II expression. THE JOURNAL OF IMMUNOLOGY 2007; 179:463-71. [PMID: 17579067 DOI: 10.4049/jimmunol.179.1.463] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
MHC type II (MHC II) expression is tightly regulated in macrophages and potently induced by IFN-gamma (type II IFN). In contrast, type I IFNs (IFN-Is), which are far more widely expressed, fail to induce MHC II expression, even though both classes of IFNs direct target gene expression through Stat1. The unexpected finding that IFN-Is effectively induce MHC II expression in Stat2(-/-) macrophages provided an opportunity to explore this conundrum. The ensuing studies revealed that deletion of Stat2, which uniquely transduces signals for IFN-Is, leads to a loss in the IFN-I-dependent induction of suppressor of cytokine signaling-1. Impairment in the expression of this important negative regulator led to a striking prolongation in IFN-I-dependent Stat1 activation, as well as enhanced expression of the target gene, IFN-regulatory factor-1. The prolonged activity of these two transcription factors synergized to drive the transcription of CIITA, the master regulator of MHC II expression, analogous to the pattern observed in IFN-gamma-treated macrophages. Thus, IFN-I-dependent suppressor of cytokine signaling-1 expression plays an important role in distinguishing the biological response between type I and II IFNs in macrophages.
Collapse
Affiliation(s)
- Wenli Zhao
- Department of Microbiology, Columbia University, Hammer Health Science Center, 701 West 168th Street, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
1112
|
Dunn IS, Haggerty TJ, Kono M, Durda PJ, Butera D, Macdonald DB, Benson EM, Rose LB, Kurnick JT. Enhancement of Human Melanoma Antigen Expression by IFN-β. THE JOURNAL OF IMMUNOLOGY 2007; 179:2134-42. [PMID: 17675472 DOI: 10.4049/jimmunol.179.4.2134] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Although many immunotherapeutic investigations have focused on improving the effector limb of the antitumor response, few studies have addressed preventing the loss of tumor-associated Ag (TAA) expression, associated with immune escape by tumors. We found that TAA loss from human melanomas usually results from reversible gene down-regulation, rather than gene deletion or mutation. Previously, we showed that inhibitors of MAPK-signaling pathways up-regulate TAA expression in melanoma cell lines. We have now identified IFN-beta as an additional stimulus to TAA expression, including Melan-A/MART-1, gp100, and MAGE-A1. IFN-beta (but neither IFN-alpha nor IFN-gamma) augmented both protein and mRNA expression of melanocytic TAA in 15 melanoma lines (irrespective of initial Ag-expression levels). Treatment of low Ag melanoma lines with IFN-beta increased expression of melanocyte-lineage Ags, inducing susceptibility to lysis by specific CTLs. Treatment with IFN-beta also enhances expression of class I HLA molecules, thereby inducing both nominal TAA and the presenting HLA molecule. Data from fluorescent cellular reporter systems demonstrated that IFN-beta triggers promoter activation, resulting in augmentation of Ag expression. In addition to enhancing TAA expression in melanomas, IFN-beta also stimulated expression of the melanocytic Ag gp100 in cells of other neural crest-derived tumor lines (gliomas) and certain unrelated tumors. Because IFN-beta is already approved for human clinical use in other contexts, it may prove useful as a cotreatment for augmenting tumor Ag expression during immunotherapy.
Collapse
|
1113
|
Dhodapkar KM, Banerjee D, Connolly J, Kukreja A, Matayeva E, Veri MC, Ravetch JV, Steinman RM, Dhodapkar MV. Selective blockade of the inhibitory Fcgamma receptor (FcgammaRIIB) in human dendritic cells and monocytes induces a type I interferon response program. J Exp Med 2007; 204:1359-69. [PMID: 17502666 PMCID: PMC2118610 DOI: 10.1084/jem.20062545] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2006] [Accepted: 04/20/2007] [Indexed: 11/12/2022] Open
Abstract
The ability of dendritic cells (DCs) to activate immunity is linked to their maturation status. In prior studies, we have shown that selective antibody-mediated blockade of inhibitory FcgammaRIIB receptor on human DCs in the presence of activating immunoglobulin (Ig) ligands leads to DC maturation and enhanced immunity to antibody-coated tumor cells. We show that Fcgamma receptor (FcgammaR)-mediated activation of human monocytes and monocyte-derived DCs is associated with a distinct gene expression pattern, including several inflammation-associated chemokines, as well as type 1 interferon (IFN) response genes, including the activation of signal transducer and activator of transcription 1 (STAT1). FcgammaR-mediated STAT1 activation is rapid and requires activating FcgammaRs. However, this IFN response is observed without a detectable increase in the expression of type I IFNs themselves or the need to add exogenous IFNs. Induction of IFN response genes plays an important role in FcgammaR-mediated effects on DCs, as suppression of STAT1 by RNA interference inhibited FcgammaR-mediated DC maturation. These data suggest that the balance of activating/inhibitory FcgammaRs may regulate IFN signaling in myeloid cells. Manipulation of FcgammaR balance on DCs and monocytes may provide a novel approach to regulating IFN-mediated pathways in autoimmunity and human cancer.
Collapse
Affiliation(s)
- Kavita M Dhodapkar
- Lab of Cellular Physiology and Immunology, The Rockefeller University, New York, NY 10021, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
1114
|
Robertson G, Hirst M, Bainbridge M, Bilenky M, Zhao Y, Zeng T, Euskirchen G, Bernier B, Varhol R, Delaney A, Thiessen N, Griffith OL, He A, Marra M, Snyder M, Jones S. Genome-wide profiles of STAT1 DNA association using chromatin immunoprecipitation and massively parallel sequencing. Nat Methods 2007; 4:651-7. [PMID: 17558387 DOI: 10.1038/nmeth1068] [Citation(s) in RCA: 1038] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2007] [Accepted: 06/05/2007] [Indexed: 02/06/2023]
Abstract
We developed a method, ChIP-sequencing (ChIP-seq), combining chromatin immunoprecipitation (ChIP) and massively parallel sequencing to identify mammalian DNA sequences bound by transcription factors in vivo. We used ChIP-seq to map STAT1 targets in interferon-gamma (IFN-gamma)-stimulated and unstimulated human HeLa S3 cells, and compared the method's performance to ChIP-PCR and to ChIP-chip for four chromosomes. By ChIP-seq, using 15.1 and 12.9 million uniquely mapped sequence reads, and an estimated false discovery rate of less than 0.001, we identified 41,582 and 11,004 putative STAT1-binding regions in stimulated and unstimulated cells, respectively. Of the 34 loci known to contain STAT1 interferon-responsive binding sites, ChIP-seq found 24 (71%). ChIP-seq targets were enriched in sequences similar to known STAT1 binding motifs. Comparisons with two ChIP-PCR data sets suggested that ChIP-seq sensitivity was between 70% and 92% and specificity was at least 95%.
Collapse
Affiliation(s)
- Gordon Robertson
- British Columbia Cancer Agency Genome Sciences Centre, 675 West 10th Avenue, Vancouver, British Columbia V5Z 4S6, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1115
|
Huang J, Smirnov SV, Lewis-Antes A, Balan M, Li W, Tang S, Silke GV, Pütz MM, Smith GL, Kotenko SV. Inhibition of type I and type III interferons by a secreted glycoprotein from Yaba-like disease virus. Proc Natl Acad Sci U S A 2007; 104:9822-7. [PMID: 17517620 PMCID: PMC1887573 DOI: 10.1073/pnas.0610352104] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2006] [Indexed: 11/18/2022] Open
Abstract
Type I (IFN-alpha/beta) and type III (IFN-lambdas) IFNs are important components of the host antiviral response. Although type III IFNs possess intrinsic antiviral activity similar to that of type I IFNs, they signal through a specific unique receptor complex, and their functional importance for antiviral resistance is largely uncharacterized. Here, we report the first virus defense mechanism that directly targets type III IFNs. Y136 from Yaba-like disease virus, a yatapoxvirus, is a secreted glycoprotein related to protein B18 from Vaccinia virus, a known type I IFN-binding protein and a member of the Ig superfamily. Surprisingly, whereas B18 inhibits only type I IFNs, Y136 inhibits both type I and type III IFNs. Y136 inhibits IFN-induced signaling and suppresses IFN-mediated biological activities including up-regulation of MHC class I antigen expression and induction of the antiviral state. These data demonstrate that poxviruses have developed unique strategies to counteract IFN-mediated antiviral protection and highlight the importance of type III IFNs in antiviral defense. These results suggest that type III IFNs may be an effective treatment for some poxviral infections.
Collapse
Affiliation(s)
- Jiaying Huang
- *Department of Biochemistry and Molecular Biology, University Hospital Cancer Center, New Jersey Medical School, Newark, NJ 07103; and
| | - Sergey V. Smirnov
- *Department of Biochemistry and Molecular Biology, University Hospital Cancer Center, New Jersey Medical School, Newark, NJ 07103; and
| | - Anita Lewis-Antes
- *Department of Biochemistry and Molecular Biology, University Hospital Cancer Center, New Jersey Medical School, Newark, NJ 07103; and
| | - Murugabaskar Balan
- *Department of Biochemistry and Molecular Biology, University Hospital Cancer Center, New Jersey Medical School, Newark, NJ 07103; and
| | - Wei Li
- *Department of Biochemistry and Molecular Biology, University Hospital Cancer Center, New Jersey Medical School, Newark, NJ 07103; and
| | - Sheila Tang
- Department of Virology, Faculty of Medicine, Imperial College London, St. Mary's Campus, London W2 1PG, United Kingdom
| | - Gemma V. Silke
- Department of Virology, Faculty of Medicine, Imperial College London, St. Mary's Campus, London W2 1PG, United Kingdom
| | - Mike M. Pütz
- Department of Virology, Faculty of Medicine, Imperial College London, St. Mary's Campus, London W2 1PG, United Kingdom
| | - Geoffrey L. Smith
- Department of Virology, Faculty of Medicine, Imperial College London, St. Mary's Campus, London W2 1PG, United Kingdom
| | - Sergei V. Kotenko
- *Department of Biochemistry and Molecular Biology, University Hospital Cancer Center, New Jersey Medical School, Newark, NJ 07103; and
| |
Collapse
|
1116
|
Swann JB, Hayakawa Y, Zerafa N, Sheehan KCF, Scott B, Schreiber RD, Hertzog P, Smyth MJ. Type I IFN Contributes to NK Cell Homeostasis, Activation, and Antitumor Function. THE JOURNAL OF IMMUNOLOGY 2007; 178:7540-9. [PMID: 17548588 DOI: 10.4049/jimmunol.178.12.7540] [Citation(s) in RCA: 236] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This study demonstrates that type I IFNs are an early and critical regulator of NK cell numbers, activation, and antitumor activity. Using both IFNAR1- and IFNAR2-deficient mice, as well as an IFNAR1-blocking Ab, we demonstrate that endogenous type I IFN is critical for controlling NK cell-mediated antitumor responses in many experimental tumor models, including protection from methylcholanthrene-induced sarcomas, resistance to the NK cell-sensitive RMA-S tumor and cytokine immunotherapy of lung metastases. Protection from RMA-S afforded by endogenous type I IFN is more potent than that of other effector molecules such as IFN-gamma, IL-12, IL-18, and perforin. Furthermore, cytokine immunotherapy using IL-12, IL-18, or IL-21 was effective in the absence of endogenous type I IFN, however the antimetastatic activity of IL-2 was abrogated in IFNAR-deficient mice, primarily due to a defect in IL-2-induced cytotoxic activity. This study demonstrates that endogenous type I IFN is a central mediator of NK cell antitumor responses.
Collapse
Affiliation(s)
- Jeremy B Swann
- Cancer Immunology Program, Trescowthick Laboratories, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, 8006 Victoria, Australia
| | | | | | | | | | | | | | | |
Collapse
|
1117
|
Martin SAM, Mohanty BP, Cash P, Houlihan DF, Secombes CJ. Proteome analysis of the Atlantic salmon (Salmo salar) cell line SHK-1 following recombinant IFN-γ stimulation. Proteomics 2007; 7:2275-86. [PMID: 17549796 DOI: 10.1002/pmic.200700020] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Type II IFN exists as a single molecule (IFN-gamma) in contrast to type I IFN, of which there are a number of different forms. IFN-gamma is involved both directly and indirectly in antiviral activity, stimulation of bactericidal activity, antigen presentation and activation of macrophages. Recently IFN-gamma was cloned from a salmonid fish, the rainbow trout and a functional recombinant protein produced exhibited IFN-gamma activity. This recombinant IFN-gamma was used to stimulate an Atlantic salmon cell line, SHK-1, to monitor the changes in protein expression by proteomic analysis 24 h after stimulation compared to unstimulated control cells. An SHK-1 cell proteome map was developed and proteins altered in abundance by the IFN-gamma stimulation were identified. Under the analytical conditions used, 22 proteins were found to be altered in abundance, 15 increased and 7 decreased. Several proteins were excised from the gel and identified, following trypsin digestion and MALDI-MS/MS/LC-MS and database interrogation. Transcriptional analysis of five mRNAs encoding proteins increased in abundance by IFN-gamma in the proteome analysis was determined by real-time PCR. We assessed the correlation between gene expression and change in abundance of proteins for these genes.
Collapse
Affiliation(s)
- Samuel A M Martin
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | | | | | | | | |
Collapse
|
1118
|
Peng FW, Duan ZJ, Zheng LS, Xie ZP, Gao HC, Zhang H, Li WP, Hou YD. Purification of recombinant human interferon-ε and oligonucleotide microarray analysis of interferon-ε-regulated genes. Protein Expr Purif 2007; 53:356-62. [PMID: 17287131 DOI: 10.1016/j.pep.2006.12.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2006] [Revised: 12/18/2006] [Accepted: 12/19/2006] [Indexed: 11/23/2022]
Abstract
Recently identified interferon-epsilon (IFN-epsilon) belongs to type I interferons. IFN-epsilon is highly and constitutively expressed in the brain, but its biochemical and biological characteristics are poorly understood. In this study, full-length IFN-epsilon cDNA was cloned from human peripheral blood lymphocyte by RT-PCR, and was expressed in Escherichia coli (E. coli). Reverse phase high pressure liquid chromatography was used to purify recombinant human IFN-epsilon (rhIFN-epsilon) and to facilitate refolding of the protein. About 0.8mg of highly purified rhIFN-epsilon protein was obtained from 100ml of E. coli culture. Functional study of rhIFN-epsilon demonstrated that the antiviral activity of rhIFN-epsilon was 6+/-0.5x10(5)IU/mg, which was lower than that of rhIFN-alpha-2b in the WISH-VSV (WISH cells infected with vesicular stomatitis virus) assay system. As for the activity to promote NK cytotoxicity and antiproliferation activities, rhIFN-epsilon was about 60 times less potent than rhIFN-alpha-2b. However, oligonucleotide microarray analyses revealed dramatic differences in gene expression profiles of cultured human cells treated with IFN-epsilon and IFN-alpha-2b. Particularly, differential regulation of genes related to central nervous system by rhIFN-epsilon suggests a role for IFN-epsilon in maintenance of the structure and function of brain.
Collapse
Affiliation(s)
- Fu-Wang Peng
- Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 100 Ying-Xin Street, Xuan Wu District, Beijing 100052, China
| | | | | | | | | | | | | | | |
Collapse
|
1119
|
Zhou Z, Hamming OJ, Ank N, Paludan SR, Nielsen AL, Hartmann R. Type III interferon (IFN) induces a type I IFN-like response in a restricted subset of cells through signaling pathways involving both the Jak-STAT pathway and the mitogen-activated protein kinases. J Virol 2007; 81:7749-58. [PMID: 17507495 PMCID: PMC1933366 DOI: 10.1128/jvi.02438-06] [Citation(s) in RCA: 379] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Type III interferon (IFN) is a novel member of the interferon family. Type III IFN utilizes a receptor complex different from that of type I IFN, but both types of IFN induce STAT1, STAT2, and STAT3 activation. Here we describe a detailed comparison of signal transduction initiated by type I and type III IFN. Gene expression array analysis showed that IFN types I and III induced a similar subset of genes. In particular, no genes were induced uniquely by type III IFN. Next, we used chromatin immunoprecipitation (ChIP) analysis to investigate the promoter activation by types I and III IFN. The ChIP assays demonstrated that stimulation of cells with both type I and type III IFN resulted in the recruitment of ISGF3 transcription factor components to the promoter region of responsive genes and in an increase of polymerase II loading and histone acetylation. Whereas IFN type I signaling was observed for a broad spectrum of cell lines, type III IFN signaling was more restricted. The lack of IFN type III signaling was correlated with a low expression of the IL28Ra component of the IFN type III receptor, and IL28Ra overexpression was sufficient to restore IFN type III signaling. We also tested the activation of mitogen-activated protein (MAP) kinases by type III IFN and found that type III IFN relies strongly upon both p38 and JNK MAP kinases for gene induction.
Collapse
Affiliation(s)
- Zhangle Zhou
- Department of Human Genetics, University of Aarhus, Denmark
| | | | | | | | | | | |
Collapse
|
1120
|
Abstract
The interferons (IFNs) and their receptors represent a subset of the class 2 alpha-helical cytokines that have been in chordates for millions of years. This brief review focuses on the discovery and purification of interferons, cloning of human IFN-alpha and IFN-beta, interferon receptors, activities and therapeutic uses of interferons, and the side effects of interferons.
Collapse
Affiliation(s)
- Sidney Pestka
- Department of Molecular Genetics, Microbiology, and Immunology, Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| |
Collapse
|
1121
|
Chelbi-Alix MK, Wietzerbin J. Interferon, a growing cytokine family: 50 years of interferon research. Biochimie 2007; 89:713-8. [PMID: 17544197 DOI: 10.1016/j.biochi.2007.05.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2007] [Accepted: 05/03/2007] [Indexed: 01/12/2023]
Abstract
The establishment of an antiviral state in cells is the defining activity of interferons (IFNs) as well as the property that permitted their discovery in 1957 by Isaacs and Lindenmann. In addition, interferons have other cellular functions that have potential clinical applications. Today, interferons are used for the treatment of a variety of malignancies and viral diseases. The publication of this special issue of Biochimie gives us a great opportunity to review the state of the art in knowledge about interferons and to explore possible future directions. This commentary text will introduce the reviews written by colleagues who are experts in different aspects of interferon research, to mark the 50th anniversary of the discovery of interferon.
Collapse
|
1122
|
Brideau-Andersen AD, Huang X, Sun SCC, Chen TT, Stark D, Sas IJ, Zadik L, Dawes GN, Guptill DR, McCord R, Govindarajan S, Roy A, Yang S, Gao J, Chen YH, Skartved NJØ, Pedersen AK, Lin D, Locher CP, Rebbapragada I, Jensen AD, Bass SH, Nissen TLS, Viswanathan S, Foster GR, Symons JA, Patten PA. Directed evolution of gene-shuffled IFN-alpha molecules with activity profiles tailored for treatment of chronic viral diseases. Proc Natl Acad Sci U S A 2007; 104:8269-74. [PMID: 17494769 PMCID: PMC1895939 DOI: 10.1073/pnas.0609001104] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Type I IFNs are unusually pleiotropic cytokines that bind to a single heterodimeric receptor and have potent antiviral, antiproliferative, and immune modulatory activities. The diverse effects of the type I IFNs are of differential therapeutic importance; in cancer therapy, an enhanced antiproliferative effect may be beneficial, whereas in the therapy of viral infections (such as hepatitis B and hepatitis C), the antiproliferative effects lead to dose limiting bone marrow suppression. Studies have shown that various members of the natural IFN-alpha family and engineered variants, such as IFN-con1, vary in the ratios between various IFN-mediated cellular activities. We used DNA shuffling to explore and confirm the hypothesis that one could simultaneously increase the antiviral and Th1-inducing activity and decrease the antiproliferative activity. We report IFN-alpha hybrids wherein the ratio of antiviral:antiproliferative and Th1-inducing: antiproliferative potencies are markedly increased with respsect to IFN-con1 (75- and 80-fold, respectively). A four-residue motif that overlaps with the IFNAR1 binding site and is derived by cross breeding with a pseudogene contributes significantly to this phenotype. These IFN-alphas have an activity profile that may result in an improved therapeutic index and, consequently, better clinical efficacy for the treatment of chronic viral diseases such as hepatitis B virus, human papilloma virus, HIV, or chronic hepatitis C.
Collapse
Affiliation(s)
| | - Xiaojian Huang
- *Maxygen, Incorporated, 515 Galveston Drive, Redwood City, CA 94063
| | | | - Teddy T. Chen
- *Maxygen, Incorporated, 515 Galveston Drive, Redwood City, CA 94063
| | - Diane Stark
- *Maxygen, Incorporated, 515 Galveston Drive, Redwood City, CA 94063
| | - Ian J. Sas
- *Maxygen, Incorporated, 515 Galveston Drive, Redwood City, CA 94063
| | - Linda Zadik
- *Maxygen, Incorporated, 515 Galveston Drive, Redwood City, CA 94063
| | - Glenn N. Dawes
- *Maxygen, Incorporated, 515 Galveston Drive, Redwood City, CA 94063
| | | | - Robert McCord
- *Maxygen, Incorporated, 515 Galveston Drive, Redwood City, CA 94063
| | | | - Ajoy Roy
- *Maxygen, Incorporated, 515 Galveston Drive, Redwood City, CA 94063
| | - Shumin Yang
- *Maxygen, Incorporated, 515 Galveston Drive, Redwood City, CA 94063
| | - Judy Gao
- *Maxygen, Incorporated, 515 Galveston Drive, Redwood City, CA 94063
| | - Yong Hong Chen
- *Maxygen, Incorporated, 515 Galveston Drive, Redwood City, CA 94063
| | | | | | - David Lin
- *Maxygen, Incorporated, 515 Galveston Drive, Redwood City, CA 94063
| | | | | | - Anne Dam Jensen
- Maxygen, Anpartsselskab, Agern Alle 1, DK-2970 Hoersholm, Denmark
| | - Steven H. Bass
- *Maxygen, Incorporated, 515 Galveston Drive, Redwood City, CA 94063
| | | | | | - Graham R. Foster
- Queen Mary's School of Medicine and Dentistry, Institute of Cell and Molecular Science, The Royal London Hospital, 4 Newark Street, London E1 2AT, England
| | - Julian A. Symons
- Roche Palo Alto LLC, 3431 Hillview Avenue, Palo Alto, CA 94304; and
| | - Phillip A. Patten
- *Maxygen, Incorporated, 515 Galveston Drive, Redwood City, CA 94063
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
1123
|
Jouanguy E, Zhang SY, Chapgier A, Sancho-Shimizu V, Puel A, Picard C, Boisson-Dupuis S, Abel L, Casanova JL. Human primary immunodeficiencies of type I interferons. Biochimie 2007; 89:878-83. [PMID: 17561326 DOI: 10.1016/j.biochi.2007.04.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Accepted: 04/27/2007] [Indexed: 01/20/2023]
Abstract
Type I interferons (IFN-alpha/beta and related molecules) are essential for protective immunity to experimental infection by numerous viruses in the mouse model. In recent years, human primary immunodeficiencies affecting either the production of (UNC-93B deficiency) or the response to (STAT1 and TYK2 deficiencies) these IFNs have been reported. Affected patients are highly susceptible to certain viruses. Patients with STAT1 or TYK2 deficiency are susceptible to multiple viruses, including herpes simplex virus-1 (HSV-1), whereas UNC-93B-deficient patients present isolated HSV-1 encephalitis. However, these immunological defects are not limited to type I IFN-mediated immunity. Impaired type II IFN (IFN-gamma)-mediated immunity plays no more than a minor role in the pathogenesis of viral diseases in these patients, but the contribution of impaired type III IFN (IFN-lambda)-mediated immunity remains to be determined. These novel inherited disorders strongly suggest that type I IFN-mediated immunity is essential for protection against natural infections caused by several viruses in humans.
Collapse
Affiliation(s)
- Emmanuelle Jouanguy
- Laboratory of Human Genetics of Infectious Diseases, Institut National de la Santé et de la Recherche Médicale, U550, 75015 Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
1124
|
Ghosh TK, Mickelson DJ, Solberg JC, Lipson KE, Inglefield JR, Alkan SS. TLR-TLR cross talk in human PBMC resulting in synergistic and antagonistic regulation of type-1 and 2 interferons, IL-12 and TNF-alpha. Int Immunopharmacol 2007; 7:1111-21. [PMID: 17570328 DOI: 10.1016/j.intimp.2007.04.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 04/06/2007] [Accepted: 04/09/2007] [Indexed: 01/12/2023]
Abstract
Currently, single TLR agonists are being utilized for vaccination and tumor immunotherapy. Here we investigated the effects of tandem combinations of TLR agonists on the production of cytokines with major focus on IFN-alpha, -beta, -gamma, TNF-alpha, and IL-12. Using a primary human PBMC culture system, we found that tandem combinations of TLR2-9 agonists can be inert, additive, synergistic or antagonistic. The most interesting combination was TLR2 or TLR4 agonists in combination with TLR7/8 or TLR8 agonists. TLR4-TLR7/8 combinations synergistically up-regulated IFN-gamma and IL-12, enhanced IFN-alpha and also moderately induced TNF-alpha. TLR2-TLR7/8 like TLR4-TLR7/8 synergistically up-regulated IFN-gamma but not IL-12. TLR9 agonist CpG2216 produced high IFN-alpha but failed to up regulate IFN-gamma singly or in tandem. Furthermore, TLR9-induced type-1 IFN was down regulated in combination with TLR7, or TLR8 agonists. TLR3 induced significant IFN-alpha/-beta responses when used in a complex with membrane permeability enhancer DOTAP, and additively enhanced response with agonists to TLR2, 5, 7/8, and 8. To our knowledge, this study is the first to compare cytokine responses of all the possible tandem combinations of TLR agonists in human PBMC. We identified certain combinations of TLR agonists that may or may not have advantages over single agonists, for generating an "optimal cytokine combination" preferred in combating diseases.
Collapse
Affiliation(s)
- Tarun K Ghosh
- 3M Pharmaceuticals, Department of Pharmacology, St. Paul, MN 55144, USA.
| | | | | | | | | | | |
Collapse
|
1125
|
Tsang SL, Leung PC, Leung KK, Yau WL, Hardy MP, Mak NK, Leung KN, Fung MC. Characterization of murine interferon-alpha 12 (MuIFN-alpha12): biological activities and gene expression. Cytokine 2007; 37:138-49. [PMID: 17451966 DOI: 10.1016/j.cyto.2007.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2006] [Revised: 02/12/2007] [Accepted: 03/12/2007] [Indexed: 10/23/2022]
Abstract
Interferon alpha (IFN-alpha) belongs to the type I interferon family and consists of multiple subtypes in many species. In the mouse, there are at least 14 IFN-alpha genes and 3 IFN-alpha pseudogenes, the most recently identified of which are murine interferon-alpha 12 (MuIFN-alpha12), MuIFN-alpha13 and MuIFN-alpha14. To further study the biological activities of MuIFN-alpha12, we have produced a recombinant MuIFN-alpha12 (rMuIFN-alpha12) protein using COS-1 cells. rMuIFN-alpha12 was found to inhibit the growth of murine myeloid leukemia JCS cells. Flow cytofluorometric analysis with propidium iodide staining showed that the growth inhibitory activity of rMuIFN-alpha12 may be caused by the induction of apoptosis. Flow cytofluorometric analysis also revealed that rMuIFN-alpha12 was able to up-regulate the expression of MHC-I on both JCS cells and primary macrophages. Functional studies indicated that a MuIFN-alpha12 transgene could induce an anti-viral state in L929 cells against Influenza A virus. Moreover, expression of MuIFN-alpha12 was not detectable by RT-PCR in untreated, Influenza A virus infected, polyI:polyC induced L929 cells, or in a wide range of normal murine tissues. Taken together, this data shows that MuIFN-alpha12 is a protein with all the biological traits of a type I IFN.
Collapse
Affiliation(s)
- Sai Leong Tsang
- Department of Biology, The Chinese University of Hong Kong, Shatin, Hong Kong.
| | | | | | | | | | | | | | | |
Collapse
|
1126
|
Charley B, Riffault S, Van Reeth K. Porcine innate and adaptative immune responses to influenza and coronavirus infections. Ann N Y Acad Sci 2007; 1081:130-6. [PMID: 17135502 PMCID: PMC7168046 DOI: 10.1196/annals.1373.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
abstract: Both innate and adaptative immune responses contribute to the control of infectious diseases, including by limiting the spreading of zoonotic diseases from animal reservoirs to humans. Pigs represent an important animal reservoir for influenza virus infection of human populations and are also naturally infected by coronaviruses, an important group of viruses, which includes the recently emerged severe acute respiratory syndrome (SARS) virus. Studies on both innate and adaptative immune responses of pigs to influenza virus and coronaviruses contribute, therefore, to a better control of these infections in their natural hosts and will be briefly reviewed in this article. Pro‐inflammatory cytokines, including type I interferon (IFN), tumor necrosis factor‐α (TNF‐α), and interleukin‐6 (IL‐6), were found in lung secretions of influenza virus infected pigs, and correlated with the intensity of clinical signs, whereas prior vaccination against influenza strongly reduced the production of infectious virus and cytokines in the lungs upon challenge, which was associated with clinical protection. An early type I IFN production was also found in coronavirus infected pigs, including at mucosal sites. IFN induction by coronavirus is shown to involve interaction between a viral glycoprotein and a leukocyte subset, likely equivalent to plasmacytoid dendritic cells, present in the mucosae and associated lymphoid tissues. Given the IFN mediated antiviral and immunomodulatory effects, the use of IFN or IFN inducers may prove an efficient strategy for a better control of influenza virus and coronavirus infections in pigs. Because influenza and coronaviruses target mucosal surfaces, adaptative immune responses have to be characterized at mucosal sites. Thus, nasal and pulmonary antibody responses were analyzed in influenza virus infected or vaccinated pigs showing short‐lived, but potentially protective local IgA and IgG antibody (Ab) responses. Interestingly, primary influenza virus infection induced long‐lived increase of lung CD8+ T cells and local lymphoproliferative responses. Pigs infected by a respiratory coronavirus (PRCV) showed virus‐specific IgG Ab‐secreting cells in the bronchial lymph nodes, whereas the transmissible gastroenteritis coronavirus (TGEV) induced more IgA Ab‐secreting cells in gut tissues, which illustrates the importance of the route of antigen administration for inducing local immune effector mechanisms. Porcine viral infections provide, therefore, valuable models for evaluating the immune parameters that are important for controlling transmission of important viral zoonotic infections.
Collapse
Affiliation(s)
- Bernard Charley
- INRA, Virologie Immunologie moléculaires, 78350, Jouy-en-Josas, France.
| | | | | |
Collapse
|
1127
|
Masci P, Olencki T, Wood L, Rybicki L, Jacobs B, Williams B, Faber P, Bukowski R, Tong K, Borden EC. Gene modulatory effects, pharmacokinetics, and clinical tolerance of interferon-alpha1b: a second member of the interferon-alpha family. Clin Pharmacol Ther 2007; 81:354-61. [PMID: 17339865 DOI: 10.1038/sj.clpt.6100081] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Interferon-alpha1 (IFN-alpha1), which may have a primary role in innate immunity, differs significantly in amino-acid sequence from IFN-alpha2, the only recombinant IFN-alpha with substantial clinical evaluation. Patients with metastatic malignancies received daily subcutaneous doses of 1.5-270 mug/m(2) of recombinant IFN-alpha1b. Gene modulation, pharmacokinetics, tolerability, and disease response were determined. Significant (P<0.01) dose and gene-dependent increases of 2-10 fold occurred in IFN-stimulated genes, including four (tumor necrosis factor-related apoptosis-inducing ligand, cig 5, p56, GEM) never previously identified as increased in patients; significant increases (P<0.01) resulted at the lowest dose (1.5 microg/m(2); 1.5 x 10(4) human antiviral units/m(2)). Increases (P<0.01) were sustainable for >4 weeks. Peak levels of IFN-alpha1b were at 3 h; an increase of approximately eightfold in both C(max) and AUC occurred between 15 microg/m(2) and 270 microg/m(2). Chronic toxicities of anorexia, weight loss, and fatigue were relatively uncommon. Eighteen patients were treated for >8 weeks; none experienced >grade 1 weight loss. Three patients at the highest dose developed grade 3 fatigue after > or =3 months, which required dose reduction or discontinuation. Patient acceptability of fatigue defined a dose for initiation of Phase II trials, 270 microg/m(2). Six patients (five with renal cell carcinoma) had progression-free survival for >1 year, including two who had partial responses. IFN-alpha1b resulted in potent stimulation of IFN-regulated genes and tumor regressions in renal cell carcinoma. Unique gene modulatory effects, when coupled with the moderate severity of side effects and a potentially central role in innate immunity, provide rationale for further clinical evaluation of IFN-alpha1 in virus infections and cancer.
Collapse
Affiliation(s)
- P Masci
- Taussig Cancer Center, Center for Hematology and Oncology Molecular Therapeutics, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1128
|
Woelk CH, Frost SDW, Richman DD, Higley PE, Kosakovsky Pond SL. Evolution of the interferon alpha gene family in eutherian mammals. Gene 2007; 397:38-50. [PMID: 17512142 PMCID: PMC2174272 DOI: 10.1016/j.gene.2007.03.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Revised: 03/27/2007] [Accepted: 03/27/2007] [Indexed: 11/27/2022]
Abstract
Interferon alpha (IFNA) genes code for proteins with important signaling roles during the innate immune response. Phylogenetically, IFNA family members in eutherians (placental mammals) cluster together in a species-specific manner except for closely related species (i.e. Homo sapiens and Pan troglodytes) where gene-specific clustering is evident. Previous research has been unable to clarify whether gene conversion or recent gene duplication accounts for gene-specific clustering, partly because the similarity of members of the IFNA family within species has made it historically difficult to identify the exact composition of IFNA gene families. IFNA gene families were fully characterized in recently available genomes from Canis familiaris, Macaca mulatta, P. troglodytes and Rattus norvegicus, and combined with previously characterized IFNA gene families from H. sapiens and Mus musculus, for the analysis of both whole and partial gene conversion events using a variety of statistical methods. Gene conversion was inferred in every eutherian species analyzed and comparison of the IFNA gene family locus between primate species revealed independent gene duplication in M. mulatta. Thus, both gene conversion and gene duplication have shaped the evolution of the IFNA gene family in eutherian species. Scenarios may be envisaged whereby the increased production of a specific IFN-alpha protein would be beneficial against a particular pathogenic infection. Gene conversion, similar to duplication, provides a mechanism by which the protein product of a specific IFNA gene can be increased.
Collapse
Affiliation(s)
- Christopher H Woelk
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA.
| | | | | | | | | |
Collapse
|
1129
|
Ahmed CM, Martin JP, Johnson HM. IFN mimetic as a therapeutic for lethal vaccinia virus infection: possible effects on innate and adaptive immune responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 178:4576-83. [PMID: 17372016 DOI: 10.4049/jimmunol.178.7.4576] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have developed small peptide mimetics of IFN-gamma that can bypass the poxvirus virulence factor B8R protein, which binds to intact IFN-gamma and prevents its interaction with receptor extracellular domain. Thus, these peptides inhibit vaccinia virus replication in cell culture where intact IFN-gamma is ineffective. We demonstrate here that the mouse IFN-gamma-mimetic peptide, IFN-gamma(95-132), protects C57BL/6 mice against overwhelming lethal vaccinia virus infection. The mimetic peptide was synthesized with an attached lipophilic group for penetration of cell plasma membrane. Injection of mimetic i.p. before and at the time of intranasal (10(6) PFU) or i.p. (10(7) PFU) challenge with virus resulted in complete protection at 200 microg of mimetic and 40-60% protection at 5 microg of mimetic. Initiation of treatment of mice with IFN-gamma mimetic up to 2 days postinfection resulted in complete protection against death, whereas initiation of treatment at 6 days postinfection resulted in 40% protection. Administration of mimetic by the oral route also completely protected mice against the intranasal route of a lethal dose of vaccinia virus challenge. In addition to its direct antiviral effect, the mimetic also possessed adjuvant effects in boosting humoral and cellular immunity to vaccinia virus. The combination of antiviral and adjuvant effects by the IFN mimetic probably plays a role in its potent anti-vaccinia virus properties. These results suggest an effective therapeutic against ongoing, lethal poxvirus infections that taps into innate and adaptive host defenses.
Collapse
Affiliation(s)
- Chulbul M Ahmed
- Department of Microbiology and Cell Science, University of Florida, Museum Road, Gainesville, FL 32611, USA.
| | | | | |
Collapse
|
1130
|
Baron S. My Career in Interferon and Host Defense Research. J Interferon Cytokine Res 2007; 27:257-62. [PMID: 17477813 DOI: 10.1089/jir.2007.9990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Samuel Baron
- Department of Microbiology & Immunology, University of Texas Medical Branch Galveston, Galveston, TX 77555-1019, USA.
| |
Collapse
|
1131
|
Avraamides G, Ng CY, David R, Gu Y, Fazekasova H, Mirenda V, Foster GR, Runkel L, Lombardi G, Marelli-Berg FM. IFN-α2 Induces Leukocyte Integrin Redistribution, Increased Adhesion, and Migration. J Interferon Cytokine Res 2007; 27:291-303. [PMID: 17477817 DOI: 10.1089/jir.2006.0107] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The human type I Interferon (IFN) family includes 14 closely related cytokines that are produced in response to viral and bacterial infections and mediate the progress of innate immune responses to adaptive immune protection, bind to a common receptor, and have qualitatively similar biologic activities. We have shown previously that IFN-alpha2 can induce human T cell chemotaxis, suggesting that type I IFNs may contribute to the development of an inflammatory environment. We here report that, in addition to promoting T cell chemotaxis, IFN-alpha2 enhances T cell adhesion to integrin ligands, which is associated with integrin clustering on the T cell surface and enhanced conjugate formation with dendritic cells. These effects were prevented by inhibition of mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K). As type I IFN receptor is ubiquitously expressed, this analysis was extended to other human leukocyte populations, including granulocytes and B cells. All leukocyte populations analyzed displayed increased chemotaxis, integrin clustering, and increased integrin-mediated adhesion following exposure to IFN-alpha2, revealing a broad-spectrum proinflammatory activity. These findings have obvious implications for the role of type I IFNs in the development of inflammatory responses leading to the initiation of adaptive immunity.
Collapse
Affiliation(s)
- George Avraamides
- Department of Immunology, Division of Medicine, Faculty of Medicine, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1132
|
Lee MT, Hooper LC, Kump L, Hayashi K, Nussenblatt R, Hooks JJ, Detrick B. Interferon-beta and adhesion molecules (E-selectin and s-intracellular adhesion molecule-1) are detected in sera from patients with retinal vasculitis and are induced in retinal vascular endothelial cells by Toll-like receptor 3 signalling. Clin Exp Immunol 2007; 147:71-80. [PMID: 17177965 PMCID: PMC1810441 DOI: 10.1111/j.1365-2249.2006.03253.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Retinal vasculitis is a major component of ocular inflammation that plays a role in retinal tissue damage in patients with idiopathic uveitis and Behçet's disease. Here we show that type 1 interferons (IFN alpha/beta) were not detected in sera from normal individuals but were identified in up to 46% of the sera from retinal vasculitis patients. The predominant form of IFN observed was IFN-beta, which was detected in 39% of Behçet's disease patients and 47% of idiopathic uveitis patients. Seven patients whose sera contained IFN-beta were monitored prospectively. IFN-beta was shown to be present for 6-12 months in all seven of the sera samples tested. Furthermore, the adhesion molecule profile identified in this study was strikingly different when Behçet's and uveitis patient sera were compared to sera from normal controls. Sera from Behçet's disease patients contained significantly elevated levels of the soluble adhesion molecules, sE-selectin and s-intracellular adhesion molecule-1 (sICAM-1), whereas sera from patients with idiopathic uveitis contained significantly increased sE-selectin. In vitro studies evaluating the cell source of these cytokines revealed that polyriboinosinic polyribocytidylic acid (poly I:C) activated retinal vascular endothelial cells produce sE-selectin, sICAM-1 and IFN-beta. Production of these molecules was inhibited by pretreatment with anti-Toll-like receptor 3 (TLR-3) antibody. In conclusion, IFN-beta, sE-selectin and sICAM-1 are elevated in patients with retinal vasculitis and are induced in retinal vascular endothelial cells in vitro by activating the innate immune system through TLR-3. Further analysis of innate immune signalling may prove to be a novel target for future studies on pathogenic mechanisms and therapeutic approaches in retinal vasculitis.
Collapse
Affiliation(s)
- M T Lee
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | |
Collapse
|
1133
|
García MA, Meurs EF, Esteban M. The dsRNA protein kinase PKR: virus and cell control. Biochimie 2007; 89:799-811. [PMID: 17451862 DOI: 10.1016/j.biochi.2007.03.001] [Citation(s) in RCA: 484] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2007] [Accepted: 03/02/2007] [Indexed: 10/23/2022]
Abstract
The IFN-induced double-stranded RNA-dependent protein kinase (PKR) is one of the four mammalian serine-threonine kinases (the three others being HRI, GCN2 and PERK) that phosphorylate the eIF2 alpha translation initiation factor, in response to stress signals, mainly as a result of viral infections. eIF2 alpha phosphorylation results in arrest of translation of both cellular and viral mRNAs, an efficient way to inhibit virus replication. The particularity of PKR is to activate by binding to dsRNA through two N terminal dsRNA binding motifs (dsRBM). PKR activation during a viral infection represents a threat for several viruses, which have therefore evolved to express PKR inhibitors, such as the Vaccinia E3L and K3L proteins. The function of PKR can also be regulated by cellular proteins, either positively (RAX/PACT; Mda7) or negatively (p58IPK, TRBP, nucleophosmin, Hsp90/70). PKR can provoke apoptosis, in part through its ability to control protein translation, but the situation appears to be more complex, as NF-kappaB, ATF-3 and p53 have also been implicated. PKR-induced apoptosis involves mainly the FADD/caspase 8 pathway, while the mitochondrial APAF/caspase 9 pathway is also engaged. As a consequence of the effects of PKR on translation, transcription and apoptosis, PKR can function to control cell growth and cell differentiation, and its activity can be controlled by the action of several oncogenes.
Collapse
Affiliation(s)
- M A García
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Ciudad Universitaria Cantoblanco, 28049 Madrid, Spain
| | | | | |
Collapse
|
1134
|
Foley KF, Pantano C, Ciolino A, Mawe GM. IFN-gamma and TNF-alpha decrease serotonin transporter function and expression in Caco2 cells. Am J Physiol Gastrointest Liver Physiol 2007; 292:G779-84. [PMID: 17170025 DOI: 10.1152/ajpgi.00470.2006] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Recent studies have shown that mucosal serotonin (5-HT) transporter (SERT) expression is decreased in animal models of colitis, as well as in the colonic mucosa of humans with ulcerative colitis and irritable bowel syndrome. Altered SERT function or expression may underlie the altered motility, secretion, and sensation seen in these inflammatory gut disorders. In an effort to elucidate possible mediators of SERT downregulation, we treated cultured colonic epithelial cells (Caco2) with conditioned medium from activated human lymphocytes. Application of the conditioned medium caused a decrease in fluoxetine-sensitive [(3)H]5-HT uptake. Individual proinflammatory agents were then tested for their ability to affect uptake. Cells were treated for 48 or 72 h with PGE(2) (10 microM), IFN-gamma (500 ng/ml), TNF-alpha (50 ng/ml), IL-12 (50 ng/ml), or the nitric oxide-releasing agent S-nitrosoglutathione (GSNO; 100 microM). [(3)H]5-HT uptake was then measured. Neither PGE nor IL-12 had any effect on [(3)H]5-HT uptake, and GSNO increased uptake. However, after 3-day incubation, both TNF-alpha and IFN-gamma elicited significant decreases in SERT function. Neither TNF-alpha nor IFN-gamma were cytotoxic when used for this period of time and at these concentrations. These two cytokines also induced decreases in SERT mRNA and protein levels. By altering SERT expression, TNF-alpha and IFN-gamma could contribute to the altered motility and expression seen in vivo in ulcerative colitis or irritable bowel syndrome.
Collapse
Affiliation(s)
- Kevin F Foley
- Department of Medical Laboratory and Radiation Sciences, the University of Vermont, Burlington, VT 05405, USA
| | | | | | | |
Collapse
|
1135
|
Magalhaes JG, Tattoli I, Girardin SE. The intestinal epithelial barrier: how to distinguish between the microbial flora and pathogens. Semin Immunol 2007; 19:106-15. [PMID: 17324587 DOI: 10.1016/j.smim.2006.12.006] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2006] [Revised: 11/03/2006] [Accepted: 12/18/2006] [Indexed: 02/06/2023]
Abstract
The gastrointestinal tract is fundamental for the uptake of nutrients and fluids, but it also represents the greatest surface of the body in contact with the external environment and most human pathogens enter the body through the mucosal surface, especially in the intestine. The intestinal immune system protects the sterile core of the organism against invasion and systemic dissemination of both pathogens and limits for level penetration of commensal microorganisms. In addition, the human intestine is continually in contact with 10(14) commensal bacteria containing more than 500 different species. These commensal bacteria confer health benefits to their host by helping dietary digestion, development of gut immunity and preventing colonization by pathogens. To maintain integrity and normal function of intestine, a delicate equilibrium must be reached between the bacterial flora and intestinal immune system. This review discusses the recent advances in our understanding of how the mucosal intestinal barrier maintains a local homeostatic response to the resident intestinal bacteria, while protecting the host against enteric pathogens. In particular, the emerging function of Toll-like receptors (TLRs) and Nod-like receptors (NLRs) in controlling mucosal immunity will be presented.
Collapse
Affiliation(s)
- Joao G Magalhaes
- Unite de Pathogenie Microbienne Moleculaire, Institut Pasteur, 25-28 rue du Dr Roux, 75724 Paris, France
| | | | | |
Collapse
|
1136
|
Paul S, Ricour C, Sommereyns C, Sorgeloos F, Michiels T. Type I interferon response in the central nervous system. Biochimie 2007; 89:770-8. [PMID: 17408841 DOI: 10.1016/j.biochi.2007.02.009] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2006] [Accepted: 02/16/2007] [Indexed: 12/25/2022]
Abstract
This review is dedicated to the influence of type I IFNs (also called IFN-alpha/beta) in the central nervous system (CNS). Studies in mice with type I IFN receptor or IFN-beta gene deficiency have highlighted the importance of the type I IFN system against CNS viral infections and non-viral autoimmune disorders. Direct antiviral effects of type I IFNs appear to be crucial in limiting early spread of a number of viruses in CNS tissues. Type I IFNs have also proved to be beneficial in autoimmune disorders like multiple sclerosis or experimental autoimmune encephalitis, probably through immunomodulatory effects. Increasing efforts are done to characterize IFN expression and response in the CNS: to identify type I IFN producing cells, to decipher pathways leading to type I IFN expression in those cells, and to identify responding cells. However, reversible and irreversible damages consecutive to chronic exposure of the CNS to type I IFNs underline the importance of a tightly regulated type I IFN homeostasis in this organ.
Collapse
Affiliation(s)
- Sophie Paul
- Université catholique de Louvain, Christian de Duve Institute of Cellular Pathology, Microbial Pathogenesis Unit, MIPA-VIRO 74-49, 74, avenue Hippocrate, B-1200, Brussels, Belgium
| | | | | | | | | |
Collapse
|
1137
|
Ananko EA, Kondrakhin YV, Merkulova TI, Kolchanov NA. Recognition of interferon-inducible sites, promoters, and enhancers. BMC Bioinformatics 2007; 8:56. [PMID: 17309789 PMCID: PMC1810324 DOI: 10.1186/1471-2105-8-56] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2006] [Accepted: 02/19/2007] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Computational analysis of gene regulatory regions is important for prediction of functions of many uncharacterized genes. With this in mind, search of the target genes for interferon (IFN) induction appears of interest. IFNs are multi-functional cytokines. Their effects are immunomodulatory, antiviral, antibacterial, and antitumor. The interaction of the IFNs with their cell surface receptors produces an activation of several transcription factors. Four regulatory factors, ISGF3, STAT1, IRF1, and NF-kappaB, are essential for the function of the IFN system. The aim of this work is the development of computational approaches for the recognition of DNA binding sites for these factors and computer programs for the prediction of the IFN-inducible regions. RESULTS We developed computational approaches to the recognition of the binding sites for ISGF3, STAT1, IRF1, and NF-kappaB. Analysis of the distribution of these binding sites demonstrated that the regions -500 upstream of the transcription start site in IFN-inducible genes are enriched in putative binding sites for these transcription factors. Based on selected combinations of the sites whose frequencies were significantly higher than in the other functional gene groups, we developed methods for the prediction of the IFN-inducible promoters and enhancers. We analyzed 1004 sequences of the IFN-inducible genes compiled using microarray data analyses and also about 10,000 human gene sequences from the EPD and RefSeq databases; 74 of 1,664 human genes annotated in EPD were significantly IFN-inducible. CONCLUSION Analyses of several control datasets demonstrated that the developed methods have a high accuracy of prediction of the IFN-inducible genes. Application of these methods to several datasets suggested that the number of the IFN-inducible genes is approximately 1500-2000 in the human genome.
Collapse
Affiliation(s)
- Elena A Ananko
- Institute of Cytology and Genetics SB RAS, Lavrentiev av., 10, 630090 Novosibirsk, Russia
| | - Yury V Kondrakhin
- Institute of Cytology and Genetics SB RAS, Lavrentiev av., 10, 630090 Novosibirsk, Russia
- Institute of Systems Biology, Novosibirsk, Russia
- Design Technological Institute of Digital Techniques SB RAS, Novosibirsk, Russia
| | - Tatiana I Merkulova
- Institute of Cytology and Genetics SB RAS, Lavrentiev av., 10, 630090 Novosibirsk, Russia
| | - Nikolay A Kolchanov
- Institute of Cytology and Genetics SB RAS, Lavrentiev av., 10, 630090 Novosibirsk, Russia
| |
Collapse
|
1138
|
García MA, Gil J, Ventoso I, Guerra S, Domingo E, Rivas C, Esteban M. Impact of protein kinase PKR in cell biology: from antiviral to antiproliferative action. Microbiol Mol Biol Rev 2007; 70:1032-60. [PMID: 17158706 PMCID: PMC1698511 DOI: 10.1128/mmbr.00027-06] [Citation(s) in RCA: 614] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The double-stranded RNA-dependent protein kinase PKR is a critical mediator of the antiproliferative and antiviral effects exerted by interferons. Not only is PKR an effector molecule on the cellular response to double-stranded RNA, but it also integrates signals in response to Toll-like receptor activation, growth factors, and diverse cellular stresses. In this review, we provide a detailed picture on how signaling downstream of PKR unfolds and what are the ultimate consequences for the cell fate. PKR activation affects both transcription and translation. PKR phosphorylation of the alpha subunit of eukaryotic initiation factor 2 results in a blockade on translation initiation. However, PKR cannot avoid the translation of some cellular and viral mRNAs bearing special features in their 5' untranslated regions. In addition, PKR affects diverse transcriptional factors such as interferon regulatory factor 1, STATs, p53, activating transcription factor 3, and NF-kappaB. In particular, how PKR triggers a cascade of events involving IKK phosphorylation of IkappaB and NF-kappaB nuclear translocation has been intensively studied. At the cellular and organism levels PKR exerts antiproliferative effects, and it is a key antiviral agent. A point of convergence in both effects is that PKR activation results in apoptosis induction. The extent and strength of the antiviral action of PKR are clearly understood by the findings that unrelated viral proteins of animal viruses have evolved to inhibit PKR action by using diverse strategies. The case for the pathological consequences of the antiproliferative action of PKR is less understood, but therapeutic strategies aimed at targeting PKR are beginning to offer promising results.
Collapse
Affiliation(s)
- M A García
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Ciudad Universitaria Cantoblanco, 28049 Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
1139
|
Huye LE, Ning S, Kelliher M, Pagano JS. Interferon regulatory factor 7 is activated by a viral oncoprotein through RIP-dependent ubiquitination. Mol Cell Biol 2007; 27:2910-8. [PMID: 17296724 PMCID: PMC1899925 DOI: 10.1128/mcb.02256-06] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
As a key mediator of type I interferon (IFN) (IFN-alpha/beta) responses, IFN regulatory factor 7 (IRF7) is essential to host immune defenses. Activation of IRF7 generally requires virus-induced C-terminal phosphorylation, which leads to its nuclear accumulation and activation of target genes. Here we use the Epstein-Barr virus (EBV) oncoprotein LMP1, which activates IRF7, to identify factors involved in IRF7 activation. We demonstrate for the first time that RIP activates IRF7 and that RIP and IRF7 interact under physiological conditions in EBV-positive Burkitt's lymphoma cells. We provide evidence that both RIP and IRF7 are ubiquitinated in these cells and that IRF7 preferentially interacts with ubiquitinated RIP. RIP is required for full activation of IRF7 by LMP1, with LMP1 stimulating the ubiquitination of RIP and its interaction with IRF7. Moreover, LMP1 stimulates RIP-dependent K63-linked ubiquitination of IRF7, which regulates protein function rather than proteasomal degradation of proteins. We suggest that RIP may serve as a general activator of IRF7, responding to and transmitting the signals from various stimuli, and that ubiquitination may be a general mechanism for enhancing the activity of IRF7.
Collapse
Affiliation(s)
- Leslie E Huye
- Lineberger Comprehensive Cancer Center, University of North Carolina, Campus Box 7295, Chapel Hill, NC 27599, USA
| | | | | | | |
Collapse
|
1140
|
Koon HB, Atkins MB. Update on therapy for melanoma: opportunities for patient selection and overcoming tumor resistance. Expert Rev Anticancer Ther 2007; 7:79-88. [PMID: 17187522 DOI: 10.1586/14737140.7.1.79] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The incidence of malignant melanoma is rising faster than any other malignancy. Although earlier stage patients can be cured with surgical resection with or without adjuvant therapy, a significant number of patients go on to develop disseminated disease. Currently, limited therapeutic options exist for patients with metastatic melanoma. Recent studies suggest that patient selection is feasible and may enable the restriction of treatment to those most likely to benefit. Additionally, several potential mechanisms of tumor resistance have been identified creating opportunities for circumventing them. This article will review current strategies for patient selection and overcoming therapeutic resistance. These strategies hold the promise of extending the clinical benefits of current therapies as well as facilitating the development of additional and more active treatments.
Collapse
Affiliation(s)
- Henry B Koon
- Beth Israel Deaconess Medical Center, Biologic Therapeutics Program, 330 Brookline Avenue, Boston, Massachusetts 02215, USA.
| | | |
Collapse
|
1141
|
Chapgier A, Boisson-Dupuis S, Jouanguy E, Vogt G, Feinberg J, Prochnicka-Chalufour A, Casrouge A, Yang K, Soudais C, Fieschi C, Santos OF, Bustamante J, Picard C, de Beaucoudrey L, Emile JF, Arkwright PD, Schreiber RD, Rolinck-Werninghaus C, Rösen-Wolff A, Magdorf K, Roesler J, Casanova JL. Novel STAT1 alleles in otherwise healthy patients with mycobacterial disease. PLoS Genet 2007; 2:e131. [PMID: 16934001 PMCID: PMC1550284 DOI: 10.1371/journal.pgen.0020131] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2006] [Accepted: 07/05/2006] [Indexed: 01/14/2023] Open
Abstract
The transcription factor signal transducer and activator of transcription-1 (STAT1) plays a key role in immunity against mycobacterial and viral infections. Here, we characterize three human STAT1 germline alleles from otherwise healthy patients with mycobacterial disease. The previously reported L706S, like the novel Q463H and E320Q alleles, are intrinsically deleterious for both interferon gamma (IFNG)–induced gamma-activating factor–mediated immunity and interferon alpha (IFNA)–induced interferon-stimulated genes factor 3–mediated immunity, as shown in STAT1-deficient cells transfected with the corresponding alleles. Their phenotypic effects are however mediated by different molecular mechanisms, L706S affecting STAT1 phosphorylation and Q463H and E320Q affecting STAT1 DNA-binding activity. Heterozygous patients display specifically impaired IFNG-induced gamma-activating factor–mediated immunity, resulting in susceptibility to mycobacteria. Indeed, IFNA-induced interferon-stimulated genes factor 3–mediated immunity is not affected, and these patients are not particularly susceptible to viral disease, unlike patients homozygous for other, equally deleterious STAT1 mutations recessive for both phenotypes. The three STAT1 alleles are therefore dominant for IFNG-mediated antimycobacterial immunity but recessive for IFNA-mediated antiviral immunity at the cellular and clinical levels. These STAT1 alleles define two forms of dominant STAT1 deficiency, depending on whether the mutations impair STAT1 phosphorylation or DNA binding. Mendelian susceptibility to mycobacterial disease is a rare syndrome. It is defined by the occurrence of severe disease caused by low virulence mycobacteria in otherwise healthy individuals, in whom antiviral immune response is not affected. Eleven known genetic defects, affecting five genes, have been involved in this type of deficient response to infection, involving immune-mediator molecules IL12 and interferon gamma: IL12B, IL12RB1, IFNGR1, IFNGR2, and STAT1. The signal transducer and activator of transcription-1 (STAT1) amino acid change L706S was previously shown to cause disease by impairing STAT1 phosphorylation. Here, we report two new STAT1 mutations that impair STAT1 DNA-binding activity. We show, by functional analysis of the three STAT1 mutant alleles, that they are intrinsically deleterious for both interferon gamma–induced antimycobacterial immunity, which is mediated through gamma-activated factor and for interferon alpha–induced antiviral immunity, which is mediated through interferon-stimulated genes factor 3. Interestingly, the three alleles are dominant for interferon gamma–induced gamma-activated factor–mediated antimycobacterial immunity, but recessive for interferon alpha–induced interferon-stimulated genes factor 3–mediated antiviral immunity at the cellular and clinical levels. These two new STAT1 alleles, which affect the binding of STAT1 to DNA, define distinct novel genetic causes of Mendelian susceptibility to mycobacterial disease and provide further insight into the molecular mechanism of disease.
Collapse
Affiliation(s)
- Ariane Chapgier
- Laboratory of Human Genetics of Infectious Diseases, University of Paris René Descartes, INSERM U550, Necker Medical School, Paris, France, European Union
| | - Stéphanie Boisson-Dupuis
- Laboratory of Human Genetics of Infectious Diseases, University of Paris René Descartes, INSERM U550, Necker Medical School, Paris, France, European Union
| | - Emmanuelle Jouanguy
- Laboratory of Human Genetics of Infectious Diseases, University of Paris René Descartes, INSERM U550, Necker Medical School, Paris, France, European Union
- French-Chinese Laboratory of Genetics, Ruijin Hospital, Shanghai II University, Shanghai, People's Republic of China
| | - Guillaume Vogt
- Laboratory of Human Genetics of Infectious Diseases, University of Paris René Descartes, INSERM U550, Necker Medical School, Paris, France, European Union
| | - Jacqueline Feinberg
- Laboratory of Human Genetics of Infectious Diseases, University of Paris René Descartes, INSERM U550, Necker Medical School, Paris, France, European Union
| | - Ada Prochnicka-Chalufour
- Laboratory of MNR of Biomolecules, CNRS URA2185, Pasteur Institute, Paris, France, European Union
| | - Armanda Casrouge
- Laboratory of Human Genetics of Infectious Diseases, University of Paris René Descartes, INSERM U550, Necker Medical School, Paris, France, European Union
| | - Kun Yang
- Laboratory of Human Genetics of Infectious Diseases, University of Paris René Descartes, INSERM U550, Necker Medical School, Paris, France, European Union
- French-Chinese Laboratory of Genetics, Ruijin Hospital, Shanghai II University, Shanghai, People's Republic of China
| | - Claire Soudais
- Laboratory of Human Genetics of Infectious Diseases, University of Paris René Descartes, INSERM U550, Necker Medical School, Paris, France, European Union
| | - Claire Fieschi
- Laboratory of Human Genetics of Infectious Diseases, University of Paris René Descartes, INSERM U550, Necker Medical School, Paris, France, European Union
- Service of Clinical Immunology, Saint Louis Hospital, Paris, France, European Union
| | - Orchidée Filipe Santos
- Laboratory of Human Genetics of Infectious Diseases, University of Paris René Descartes, INSERM U550, Necker Medical School, Paris, France, European Union
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, University of Paris René Descartes, INSERM U550, Necker Medical School, Paris, France, European Union
| | - Capucine Picard
- Laboratory of Human Genetics of Infectious Diseases, University of Paris René Descartes, INSERM U550, Necker Medical School, Paris, France, European Union
- Center for the Study of Immunodeficiences, Necker Hospital, Paris, France, European Union
| | - Ludovic de Beaucoudrey
- Laboratory of Human Genetics of Infectious Diseases, University of Paris René Descartes, INSERM U550, Necker Medical School, Paris, France, European Union
| | - Jean-François Emile
- Department of Pathology, Ambroise Paré Hospital, Boulogne, France, European Union
| | | | - Robert D Schreiber
- Department of Pathology and Immunology, Washington University, Saint Louis, Missouri, United States of America
| | | | - Angela Rösen-Wolff
- Department of Pediatrics, University Clinic Carl Gustav Carus, Dresden, Germany
| | - Klaus Magdorf
- Department of Pediatric Pneumology and Immunology, Charité, Humboldt University of Berlin, Berlin, Germany
| | - Joachim Roesler
- Department of Pediatrics, University Clinic Carl Gustav Carus, Dresden, Germany
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, University of Paris René Descartes, INSERM U550, Necker Medical School, Paris, France, European Union
- French-Chinese Laboratory of Genetics, Ruijin Hospital, Shanghai II University, Shanghai, People's Republic of China
- Pediatric Immunology Hematology Unit, Necker Hospital, Paris, France, European Union
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
1142
|
Siebler J, Wirtz S, Weigmann B, Atreya I, Schmitt E, Kreft A, Galle PR, Neurath MF. IL-28A is a key regulator of T-cell-mediated liver injury via the T-box transcription factor T-bet. Gastroenterology 2007; 132:358-71. [PMID: 17241885 DOI: 10.1053/j.gastro.2006.10.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Accepted: 10/05/2006] [Indexed: 02/04/2023]
Abstract
BACKGROUND & AIMS T-cell-mediated fulminant hepatitis is a potentially life-threatening event for which the underlying pathogenic mechanisms are not fully understood. Here, we demonstrate a key regulatory role of IL-28A in T-cell-mediated hepatitis. METHODS We cloned the murine IL-28A gene by reverse-transcription polymerase chain reaction, assessed the effects of recombinant IL-28A, and generated IL-28A-transgenic mice. RESULTS IL-28A induced TH1 cytokine production by CD4+ T lymphocytes in a T-bet-dependent manner and was up-regulated in a murine model of T-cell-mediated hepatitis upon Con A administration. In vivo, CD4+ T cells from newly created IL-28A-transgenic animals revealed an increased proliferation and proinflammatory TH1 cytokine production, as compared with wild-type mice. In addition, IL-28A-transgenic mice showed markedly augmented Con A-induced hepatitis with up-regulated interferon-gamma cytokine production, as compared with wild-type mice. Transgenic mice exhibited an up-regulation of the interferon-gamma-T-bet signaling pathway in Con A hepatitis, and augmented hepatitis in these mice was suppressed by crossing them with T-bet-deficient mice. In addition, in vivo blockade of interferon-gamma but not IL-4 suppressed augmented liver inflammation in transgenic mice, suggesting that IL-28A induces the T-bet signaling pathway in T-cell-induced hepatitis. Finally, IL-28A-specific antisense phosphorothioate oligonucleotides suppressed liver pathology in Con A-treated wild-type mice, as compared with the case of control oligonucleotides. CONCLUSIONS IL-28A emerges as a key regulatory cytokine with pathogenic function in T-cell-mediated liver injury. Thus, targeting of IL-28A represents a potential novel approach for therapy of Th1-mediated inflammatory diseases such as T-cell-mediated hepatitis.
Collapse
Affiliation(s)
- Juergen Siebler
- Laboratory of Immunology, I. Medical Clinic, University of Mainz, Lasngenbeckstrasse 1, 55131 Mainz, Germany
| | | | | | | | | | | | | | | |
Collapse
|
1143
|
Bokodi G, Derzbach L, Bányász I, Tulassay T, Vásárhelyi B. Association of interferon gamma T+874A and interleukin 12 p40 promoter CTCTAA/GC polymorphism with the need for respiratory support and perinatal complications in low birthweight neonates. Arch Dis Child Fetal Neonatal Ed 2007; 92:F25-9. [PMID: 16754651 PMCID: PMC2675292 DOI: 10.1136/adc.2005.086421] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND Data support the role of interferon (IFN)gamma and interleukin (IL)12 in perinatal complications. IFNgamma T(+874)A and IL12 p40 promoter CTCTAA/GC polymorphisms may have an effect on cytokine production. METHODS DNA was extracted from dried blood samples of 153 low birthweight (LBW) infants and 172 healthy term infants. IFNgamma and IL12 genetic polymorphisms were determined to investigate the association between polymorphisms and ventilation characteristics, bronchopulmonary dysplasia (BPD) and other perinatal disorders. RESULTS The IFNgamma(+874)A allele was over-represented in LBW infants. Carriers of the IFNgamma(+874)T allele required mechanical ventilation and oxygen supplementation for time periods 41% and 35%, respectively, shorter than those required by those not carrying the IFNgamma(+874)T allele. Stepwise logistic regression analysis showed that carriers of the IFNgamma(+874)T allele were protected against BPD (odds ratio (OR) 0.35 (95% confidence interval (CI) (0.12 to 0.99))) and patent ductus arteriosus (OR 0.43 (95% CI 0.19 to 0.97)), whereas carriers of the IFNgamma(+874)A allele were at higher risk of severe hypotension (OR 3.40 (95% CI 1.01 to 11.52)) and respiratory distress syndrome (OR 4.03 (95% CI 1.30 to 12.50)). Carriers of the IL12 GC allele were protected against pneumonia (OR 0.32 (95% CI 0.14 to 0.75)). Carriers of the IL12 CTCTAA allele were at higher risk of developing necrotising enterocolitis (NEC; OR 2.37 (95% CI 1.01 to 5.53)). CONCLUSIONS Carrier state of the IFNgamma(+874)A allele presents an increased risk for premature birth and lung damage, as well as other perinatal complications. The risks of pneumonia and NEC are higher in heterozygotic carriers of the IL12 CTCTAA/GC polymorphism. Further studies are needed to determine whether these associations are the result of altered cytokine-producing capacity in infants carrying the tested alleles.
Collapse
Affiliation(s)
- G Bokodi
- First Department of Pediatrics, Semmelweis University, Budapest, Hungary.
| | | | | | | | | |
Collapse
|
1144
|
Lash GE, Otun HA, Innes BA, Kirkley M, De Oliveira L, Searle RF, Robson SC, Bulmer JN. Interferon-gamma inhibits extravillous trophoblast cell invasion by a mechanism that involves both changes in apoptosis and protease levels. FASEB J 2007; 20:2512-8. [PMID: 17142800 DOI: 10.1096/fj.06-6616com] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Extravillous trophoblast cell (EVT) invasion of decidua and inner third of the myometrium is critical for a successful pregnancy. Many decidual factors are likely to play a role in regulating this process, including uterine natural killer (uNK) cell-derived cytokines. HYPOTHESES 1) uNK cells are a major source of IFN gamma (IFN-gamma) and 2) IFN-gamma inhibits EVT invasion via an increase in EVT apoptosis and/or a decrease in active protease levels. METHODS Total decidual and uNK cells from 8-10 wk and 12-14 wk gestational age were cultured. IFN-gamma mRNA (real-time RT-polymerase chain reaction) and protein levels (FastQuant multicytokine analysis) were determined. EVT invasion in the presence of IFN-gamma or anti-IFN-gamma-neutralizing antibodies was assessed. Trophoblast apoptosis and proliferation was assessed in explants by immunohistochemistry for M30 and Ki67. Substrate zymography was performed to determine levels of secreted MMP2, MMP9, and uPA. RESULTS mRNA and protein for IFN-gamma was detected in both total decidual and uNK cell fractions. Trophoblast invasion was inhibited by IFN-gamma. The level of M30-positive EVT was increased in the presence of IFN-gamma whereas levels of secreted MMP2 were decreased. CONCLUSIONS uNK cells are a source of IFN-gamma within early human pregnancy decidua. Mechanisms of IFN-gamma inhibition of EVT invasion include both increased EVT apoptosis and reduced levels of active proteases.
Collapse
Affiliation(s)
- Gendie E Lash
- School of Surgical and Reproductive Sciences, 3rd Floor, William Leech Bldg., University of Newcastle upon Tyne, Newcastle upon Tyne NE2 4HH, UK.
| | | | | | | | | | | | | | | |
Collapse
|
1145
|
Sheehan KCF, Lai KS, Dunn GP, Bruce AT, Diamond MS, Heutel JD, Dungo-Arthur C, Carrero JA, White JM, Hertzog PJ, Schreiber RD. Blocking monoclonal antibodies specific for mouse IFN-alpha/beta receptor subunit 1 (IFNAR-1) from mice immunized by in vivo hydrodynamic transfection. J Interferon Cytokine Res 2006; 26:804-19. [PMID: 17115899 DOI: 10.1089/jir.2006.26.804] [Citation(s) in RCA: 219] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Herein we report the generation of mouse monoclonal antibodies (mAbs) specific for the IFNAR-1 subunit of the mouse interferon-alpha/beta (IFN-alpha/beta) receptor (MAR1 mAbs) that block type I IFN receptor signaling and biologic response induction in vitro and in vivo. These mAbs were generated from Ifnar1 (/) mice immunized by in vivo hydrodynamic transfection with a plasmid encoding the extracellular domain (ECD) of murine IFNAR-1. All MAR1 mAbs bound native receptor expressed on cell surfaces and immunoprecipitated IFNAR-1 from solubilized cells, and two mAbs also detected IFNAR-1 by Western blot analysis. in vitro, the mAbs prevented ligand-induced intracellular signaling and induction of a variety of type I IFN-induced biologic responses but had no effect on IFN-gamma-induced responses. The most effective in vitro blocker, MAR1-5A3, also blocked type I IFN-induced antiviral, antimicrobial, and antitumor responses in vivo. We also explored whether murine IFNAR-1 surface expression required the presence of Tyk2. In contrast to Tyk2-deficient human cell lines, comparable IFNAR-1 expression was found on primary cells derived either from wild-type or Tyk2 (/) mice. These mAbs represent much needed tools to more clearly elucidate the biochemistry, cell biology, and physiologic function of the type I IFNs and their receptor in mediating host-protective immunity and immunopathology.
Collapse
Affiliation(s)
- Kathleen C F Sheehan
- Department of Pathology and Immunology, Center for Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1146
|
Abstract
A clear picture of the dynamic relationship between the host immune system and cancer is emerging as the cells and molecules that participate in naturally occurring antitumour immune responses are being identified. The interferons (IFNs) - that is, the type I IFNs (IFNalpha and IFNbeta) and type II IFN (IFNgamma) - have emerged as central coordinators of tumour-immune-system interactions. Indeed, the decade-old finding that IFNgamma has a pivotal role in promoting antitumour responses became the focus for a renewed interest in the largely abandoned concept of cancer immunosurveillance. More recently, type I IFNs have been found to have distinct functions in this process. In this Review, we discuss the roles of the IFNs, not only in cancer immunosurveillance but also in the broader process of cancer immunoediting.
Collapse
Affiliation(s)
- Gavin P Dunn
- Washington University School of Medicine, Department of Pathology and Immunology, Box 8118, 660 South Euclid Avenue, St. Louis, Missouri 63110, USA
| | | | | |
Collapse
|
1147
|
Fabricius D, O'Dorisio MS, Blackwell S, Jahrsdörfer B. Human plasmacytoid dendritic cell function: inhibition of IFN-alpha secretion and modulation of immune phenotype by vasoactive intestinal peptide. THE JOURNAL OF IMMUNOLOGY 2006; 177:5920-7. [PMID: 17056516 DOI: 10.4049/jimmunol.177.9.5920] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Plasmacytoid dendritic cells (PDC) are considered the main sentinels against viral infections and play a major role in immune tolerance. Vasoactive intestinal peptide (VIP) is a potent immunomodulator, whose role in PDC function is unknown. The present study was designed to investigate whether human PDC express VIP receptors and whether VIP has immunological effects on PDC. Using real-time RT-PCR and immunofluorescence, we demonstrated that VIP receptors VPAC1 and VPAC2 are expressed on PDC. After culturing PDC with VIP and CpG oligodeoxynucleotides for 48 h, expression of surface molecules with significance for PDC-T cell interactions as well as IFN-alpha secretion were quantified using FACS analysis and ELISA, respectively. For functional assays, CFSE-stained CD4+ T cells were coincubated with differentially treated PDC. T cell proliferation and production of various cytokines were determined by FACS analysis and ELISA. VIP enhanced PDC expression of CD86, MHC II, and CCR7. In contrast, VIP inhibited PDC secretion of IFN-alpha and expression of Neuropilin-1 and MHC I. The potential of CpG oligodeoxynucleotide-activated PDC to induce proliferation of allogeneic CD4(+) T cells was impaired when VIP was present during activation. Furthermore, pretreatment of PDC with VIP resulted in a decrease of the IFN-gamma:IL-4 ratio in cocultured T cells, suggesting a modulation of the immune response toward Th2. Taken together, these results strongly suggest that VIP regulates the immunological function of human PDC. VIP may thus be involved in the modulation of immune responses to viral infections as well as in the maintenance of immune tolerance.
Collapse
MESH Headings
- CD4-Positive T-Lymphocytes/immunology
- Cell Proliferation
- Cells, Cultured
- Dendritic Cells/chemistry
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Humans
- Interferon-alpha/antagonists & inhibitors
- Interferon-alpha/metabolism
- Interferon-gamma/metabolism
- Interleukin-4/metabolism
- Oligodeoxyribonucleotides/pharmacology
- RNA, Messenger/analysis
- RNA, Messenger/metabolism
- Receptors, Vasoactive Intestinal Peptide, Type II/analysis
- Receptors, Vasoactive Intestinal Peptide, Type II/genetics
- Receptors, Vasoactive Intestinal Peptide, Type II/metabolism
- Receptors, Vasoactive Intestinal Polypeptide, Type I/analysis
- Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics
- Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism
- Transforming Growth Factor beta/metabolism
- Vasoactive Intestinal Peptide/pharmacology
Collapse
Affiliation(s)
- Dorit Fabricius
- Department of Pediatrics, Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
1148
|
Jaks E, Gavutis M, Uzé G, Martal J, Piehler J. Differential receptor subunit affinities of type I interferons govern differential signal activation. J Mol Biol 2006; 366:525-39. [PMID: 17174979 DOI: 10.1016/j.jmb.2006.11.053] [Citation(s) in RCA: 177] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2006] [Revised: 11/15/2006] [Accepted: 11/15/2006] [Indexed: 12/17/2022]
Abstract
Type I interferons (IFNs) elicit antiviral, antiproliferative and immunmodulatory responses by binding to a shared cell surface receptor comprising the transmembrane proteins ifnar1 and ifnar2. Activation of differential response patterns by IFNs has been observed, suggesting that members of the family play different roles in innate immunity. The molecular basis for differential signaling has not been identified yet. Here, we have investigated the recognition of various IFNs including several human IFNalpha species, human IFNomega and human IFNbeta as well as ovine IFNtau2 by the receptor subunits in detail. Binding to the extracellular domains of ifnar1 (ifnar1-EC) and ifnar2 (ifnar2-EC) was monitored in real time by reflectance interference and total internal reflection fluorescence spectroscopy. For all IFNs investigated, competitive 1:1 interaction not only with ifnar2-EC but also with ifnar1-EC was shown. Furthermore, ternary complex formation was studied with ifnar1-EC and ifnar2-EC tethered onto solid-supported membranes. These analyses confirmed that the signaling complexes recruited by IFNs have very similar architectures. However, differences in rate and affinity constants over several orders of magnitude were observed for both the interactions with ifnar1-EC and ifnar2-EC. These data were correlated with the potencies of ISGF3 activation, antiviral and anti-proliferative activity on 2fTGH cells. The ISGF3 formation and antiviral activity correlated very well with the binding affinity towards ifnar2. In contrast, the affinity towards ifnar1 played a key role for antiproliferative activity. A striking correlation was observed for relative binding affinities towards ifnar1 and ifnar2 with the differential antiproliferative potency. This correlation was confirmed by systematically engineering IFNalpha2 mutants with very high differential antiproliferative potency.
Collapse
Affiliation(s)
- Eva Jaks
- Institute of Biochemistry, Johann Wolfgang Goethe-University Frankfurt am Main, Germany
| | | | | | | | | |
Collapse
|
1149
|
Peycheva E, Daskalov I, Tsoneva I. Electrochemotherapy of Mycosis fungoides by interferon-alpha. Bioelectrochemistry 2006; 70:283-6. [PMID: 17150416 DOI: 10.1016/j.bioelechem.2006.10.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2006] [Revised: 09/15/2006] [Accepted: 10/13/2006] [Indexed: 12/21/2022]
Abstract
Eight patients with 29 lesions of histologically verified 1st stage of Mycosis fungoides were successfully treated by electrochemotherapy with interferon-alpha. For this purpose 8 biphasic pulses were used, each of 50+50 micros duration with 900 micros interpulse intervals, resulting in a burst of 7.1 ms total duration. Compared to the traditional monoimmunotherapy with interferon-alpha applied three times weekly for a total of 4 weeks, the electrochemotherapy was very efficient. Complete response (CR) was observed in 25 (86%) of the 29 treated lesions by single-act electrochemotherapy with interferon-alpha. At the end of the 12-month period, all 29 lesions showed 100% complete response (CR). New lesions for a period of 12 months were not observed. The expected mechanism involved in multiple cytotoxic action of interferon-alpha could be the local increased concentration in the tumour and prolongation of the time of its action after the application of pulses.
Collapse
Affiliation(s)
- E Peycheva
- National Centre of Oncology, Clinic of Oncodermatology, 6 Plovdivsko pole Blv., 1756 Sofia, Bulgaria
| | | | | |
Collapse
|
1150
|
Samuel MA, Diamond MS. Pathogenesis of West Nile Virus infection: a balance between virulence, innate and adaptive immunity, and viral evasion. J Virol 2006; 80:9349-60. [PMID: 16973541 PMCID: PMC1617273 DOI: 10.1128/jvi.01122-06] [Citation(s) in RCA: 242] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Melanie A Samuel
- Division of Infectious Diseases, Department of Molecular Microbiology, Washington University School of Medicine, Campus Box 8051, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | | |
Collapse
|