151
|
Drayton S, Rowe J, Jones R, Vatcheva R, Cuthbert-Heavens D, Marshall J, Fried M, Peters G. Tumor suppressor p16INK4a determines sensitivity of human cells to transformation by cooperating cellular oncogenes. Cancer Cell 2003; 4:301-10. [PMID: 14585357 DOI: 10.1016/s1535-6108(03)00242-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The Ink4a/Arf locus encodes two distinct proteins, both of which may contribute to senescence and tumor suppression. We find that human diploid fibroblasts (HDFs) that are specifically deficient for p16INK4a achieve anchorage independence when transduced with retroviruses encoding telomerase (hTERT) and either Ras or Myc. Significantly, Ras and Myc together enable the cells to form tumors in nude mice but at a frequency that suggests additional genetic changes. All five tumors analyzed expressed high levels of Ras and retained functional p53, although two showed downregulation of Arf. Cytogenetic analyses identified clonal chromosomal alterations that may have contributed to tumorigenesis, but the tumor cells were essentially diploid.
Collapse
Affiliation(s)
- Sarah Drayton
- Molecular Oncology Laboratory, Cancer Research UK London Research Institute, Lincolns Inn Fields, London WC2A 3PX, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
152
|
Yim JH, Ro SH, Lowney JK, Wu SJ, Connett J, Doherty GM. The role of interferon regulatory factor-1 and interferon regulatory factor-2 in IFN-gamma growth inhibition of human breast carcinoma cell lines. J Interferon Cytokine Res 2003; 23:501-11. [PMID: 14565859 DOI: 10.1089/10799900360708623] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Interferon (IFN) regulatory factor-1 (IRF-1) and IRF-2 play opposing roles in the regulation of many IFN-gamma-inducible genes. To investigate the signal transduction pathway in response to IFN-gamma in light of differences in growth effects, we selected four human breast carcinoma cell lines based on a spectrum of growth inhibition by IFN-gamma. MDA468 growth was markedly inhibited by IFN-gamma, and it showed substantial induction of IRF-1 mRNA but little IRF-2 induction. SKBR3 showed little growth inhibition and little induction of IRF-1 mRNA but significant induction of IRF-2 mRNA. HS578T and MDA436 growth inhibition and IRF-1/IRF-2 induction were intermediate. All four cell lines showed intact receptor at the cell surface and Stat1 translocation to the nucleus by immunostaining. By EMSA, there were marked differences in the induced ratio of IRF-1 and IRF-2 binding activity between the cell lines that correlated with growth inhibition. Finally, antisense oligonucleotides specific for IRF-1 attenuated IFN-gamma growth inhibition in MDA436 and MDA468, confirming the direct role of IRF-1 in IFN-gamma growth inhibition. Induction of IRF-1 causes growth inhibition in human breast cancer cell lines, and induction of IRF-2 can oppose this. The relative induction of IRF-1 to IRF-2 is a critical control point in IFN-gamma response.
Collapse
Affiliation(s)
- John H Yim
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | | | |
Collapse
|
153
|
Takaoka A, Hayakawa S, Yanai H, Stoiber D, Negishi H, Kikuchi H, Sasaki S, Imai K, Shibue T, Honda K, Taniguchi T. Integration of interferon-alpha/beta signalling to p53 responses in tumour suppression and antiviral defence. Nature 2003; 424:516-23. [PMID: 12872134 DOI: 10.1038/nature01850] [Citation(s) in RCA: 683] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2003] [Accepted: 05/28/2003] [Indexed: 12/20/2022]
Abstract
Swift elimination of undesirable cells is an important feature in tumour suppression and immunity. The tumour suppressor p53 and interferon-alpha and -beta (IFN-alpha/beta) are essential for the induction of apoptosis in cancerous cells and in antiviral immune responses, respectively, but little is known about their interrelationship. Here we show that transcription of the p53 gene is induced by IFN-alpha/beta, accompanied by an increase in p53 protein level. IFN-alpha/beta signalling itself does not activate p53; rather, it contributes to boosting p53 responses to stress signals. We show examples in which p53 gene induction by IFN-alpha/beta contributes to tumour suppression. Furthermore, we show that p53 is activated in virally infected cells to evoke an apoptotic response and that p53 is critical for antiviral defence of the host. Our study reveals a hitherto unrecognized link between p53 and IFN-alpha/beta in tumour suppression and antiviral immunity, which may have therapeutic implications.
Collapse
Affiliation(s)
- Akinori Takaoka
- Department of Immunology, Faculty of Medicine and Graduate School of Medicine, University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
154
|
Pao W, Klimstra DS, Fisher GH, Varmus HE. Use of avian retroviral vectors to introduce transcriptional regulators into mammalian cells for analyses of tumor maintenance. Proc Natl Acad Sci U S A 2003; 100:8764-9. [PMID: 12857957 PMCID: PMC166387 DOI: 10.1073/pnas.1133333100] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A key issue in cancer biology is whether genetic lesions involved in tumor initiation or progression are required for tumor maintenance. This question can be addressed with mouse models that conditionally express oncogenic transgenes, i.e., under the control of tetracycline (tet)-dependent transcriptional regulators. We have developed a system for studying tumor maintenance by using avian retroviral [i.e., replication-competent avian leukosis virus long terminal repeat with splice acceptor (RCAS)] vectors to deliver the reverse tet transcriptional transactivator (rtTA) gene to somatic mammalian cells. rtTA can regulate any transgene in which the protein coding sequence is preceded by a tet-operator (tet-o); RCAS viruses infect only cells engineered to express ectopically the avian retroviral receptor, TVA. One vector, RCAS-rtTA-IRES-GFP, also encodes GFP to identify infected cells. Infection of cells from beta-actin TVA transgenic mice with this vector permits efficient regulation of tet-responsive transgenes. Sarcomas arise when p53-deficient murine embryonic fibroblasts carrying beta-actin TVA and tet-o-K-ras4bG12D transgenes are infected with RCAS-rtTA-IRES-GFP and introduced into nude mice treated with the tet analog, doxycycline (dox); when dox is withdrawn, K-ras4bG12D levels fall, cells undergo apoptosis, and tumors regress. Regression can be prevented by means of a genetic complementation assay in which tumors are superinfected before dox withdrawal with other RCAS viruses, such as those carrying an active allele of K-ras. Many TVA and tet-regulated transgenic mice have been generated; thus, this method for somatic cell-specific and temporally controlled gene expression may have broad applications for the study of oncogenesis and tumor maintenance, as well as other cell functions and development.
Collapse
Affiliation(s)
- William Pao
- Departments of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA.
| | | | | | | |
Collapse
|
155
|
Mistry AR, Pedersen EW, Solomon E, Grimwade D. The molecular pathogenesis of acute promyelocytic leukaemia: implications for the clinical management of the disease. Blood Rev 2003; 17:71-97. [PMID: 12642121 DOI: 10.1016/s0268-960x(02)00075-9] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Acute promyelocytic leukaemia (APL) is characterised by chromosomal rearrangements of 17q21, leading to fusion of the gene encoding retinoic acid receptor alpha (RARalpha) to a number of alternative partner genes (X), the most frequent of which are PML (>95%), PLZF (0.8%) and NPM (0.5%). Over the last few years, it has been established that the X-RARalpha fusion proteins play a key role in the pathogenesis of APL through recruitment of co-repressors and the histone deacetylase (HDAC)-complex to repress genes implicated in myeloid differentiation. Paradoxically, the X-RARalpha fusion protein has the potential to mediate myeloid differentiation at pharmacological doses of its ligand (all trans-retinoic acid (ATRA)), which is dependent on the dissociation of the HDAC/co-repressor complex. Arsenic compounds have also been shown to be promising therapeutic agents, leading to differentiation and apoptosis of APL blasts. It is now apparent that the nature of the RARalpha-fusion partner is a critical determinant of response to ATRA and arsenic, underlining the importance of cytogenetic and molecular characterisation of patients with suspected APL to determine the most appropriate treatment approach. Standard protocols involving ATRA combined with anthracycline-based chemotherapy, lead to cure of approximately 70% patients with PML-RARalpha-associated APL. Patients at high risk of relapse can be identified by minimal residual disease monitoring. The challenge for future studies is to improve complete remission rates through reduction of induction deaths, particularly due to haemorrhage, identification of patients at high risk of relapse who would benefit from additional therapy, and identification of a favourable-risk group, for which treatment intensity could be reduced, thereby reducing risks of treatment toxicity and development of secondary leukaemia/myelodysplasia. With the advent of ATRA and arsenic, APL has already provided the first example of successful molecularly targeted therapy; it is hoped that with further understanding of the pathogenesis of the disease, the next decade will yield further improvements in the outlook for these patients.
Collapse
MESH Headings
- Animals
- Gene Rearrangement
- Humans
- Leukemia, Promyelocytic, Acute/genetics
- Leukemia, Promyelocytic, Acute/pathology
- Leukemia, Promyelocytic, Acute/therapy
- Models, Biological
- Mutation
- Neoplasm, Residual/genetics
- Neoplasm, Residual/pathology
- Receptors, Retinoic Acid/genetics
- Receptors, Retinoic Acid/metabolism
- Retinoic Acid Receptor alpha
- Translocation, Genetic
Collapse
Affiliation(s)
- Anita R Mistry
- Division of Medical and Molecular Genetics, Guy's, King's and St Thomas' School of Medicine, London, UK
| | | | | | | |
Collapse
|
156
|
Yang E, Lerner L, Besser D, Darnell JE. Independent and cooperative activation of chromosomal c-fos promoter by STAT3. J Biol Chem 2003; 278:15794-9. [PMID: 12600988 DOI: 10.1074/jbc.m213073200] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The c-fos gene was one of the earliest vertebrate genes shown to be transcriptionally induced by growth factors. Intensive study of the promoter of c-fos (-325 to -80) by transient or permanent transfections of synthetic DNA constructs has repeatedly shown the importance of several sequence elements and the resident nuclear proteins that bind them (e.g. ternary complex factor/ELK1; serum response factor, cAMP response element-binding protein/amino-terminal fragment/AP-1). However these studies have left unanswered numerous questions about the role of these proteins in the regulation of the native chromosomal gene. In particular, the role of a site in this enhancer that binds STATs has been controversial. We present evidence here that STAT3 and not STAT1 accumulates on the chromosomal c-fos promoter and provides a boost to transcription without the activation of resident nuclear proteins through serine kinases. Also, when resident nuclear proteins such as ELK1 are activated to varying extents by mitogen-activated protein kinase pathways, STAT3 activation provides a 2-fold boost regardless of the final level of activated transcription. Thus the several proteins that interact with the c-fos enhancer apparently can act either in a cooperative or independent manner to achieve very different levels of transcription.
Collapse
Affiliation(s)
- Edward Yang
- Laboratory of Molecular Cell Biology, The Rockefeller University, New York, NY 10021, USA
| | | | | | | |
Collapse
|
157
|
Kim EJ, Park CH, Park JS, Um SJ. Functional dissection of the transactivation domain of interferon regulatory factor-1. Biochem Biophys Res Commun 2003; 304:253-9. [PMID: 12711307 DOI: 10.1016/s0006-291x(03)00575-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Interferon regulatory factor-1 (IRF-1), a putative tumor suppressor protein, is a transcriptional mediator of interferon-responsive signaling pathways that are involved in antiviral defense, apoptosis, immune response, and cell growth regulation. To delineate the IRF-1 domain responsible for transactivation, we performed a detailed deletion analysis of IRF-1. We found that the amino acid segment 217-260 was necessary and sufficient for transactivation. The structure of this region was predicted to be loop-helix-loop-sheet using the program PHD. Further studies indicated that casein kinase II and protein kinase C sites on each of the two loops are not important for transactivation, and a region containing amino acids 233-255 comprises the core activation domain. To verify the physiological role of segment 233-255, we constructed an IRF-1 deletion mutant lacking these amino acids and examined it using IRF-1 transactivation assay, fluorescence-activated cell sorting, and in situ beta-galactosidase staining techniques. From the results of these studies, we conclude that the amino acid segment 233-255 of IRF-1 comprises the core activation domain required for the physiological functions of IRF-1.
Collapse
Affiliation(s)
- Eun-Joo Kim
- Department of Bioscience and Biotechnology/Institute of Bioscience, Sejong University, 98 Kunja-dong, Kwangjin-gu, Seoul 143-747, Republic of Korea
| | | | | | | |
Collapse
|
158
|
Kröger A, Dallügge A, Kirchhoff S, Hauser H. IRF-1 reverts the transformed phenotype of oncogenically transformed cells in vitro and in vivo. Oncogene 2003; 22:1045-56. [PMID: 12592391 DOI: 10.1038/sj.onc.1206260] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The expression of the transcriptional activator and tumor suppressor IRF-1 induces multiple effects that counteract the growth of tumor cells in vitro and in vivo. These include the inhibition of cell proliferation, the secretion of interferon-beta (IFN-beta), the induction of apoptosis specifically in certain cell types and the induction of a strong T-cell response. Here, we show that apart from its immune-activating properties, IRF-1 expression leads to a reversion of the tumorigenic phenotype of NIH3T3 cells transformed by different oncogenes. This was analysed in detail in a cell line in which the expression of c-Ha-ras and c-myc is under the control of a doxycycline-regulated promoter allowing to switch between the normal and oncogenic cell status. In the same cells, a beta-estradiol activatable IRF-1 fusion protein is expressed. After IRF-1 activation the oncogene-mediated acceleration of the cell cycle is reverted. Further, a complete IRF-1-mediated reversion of the oncogenic phenotype is observed in soft-agar growth assays. IRF-1 activation induces IFN-beta secretion; however, the observed effects are not mediated by IFN-beta. Inhibition of tumor growth is observed in nude mice as long as IRF-1 is active, indicating that neither B- nor T-cells must become activated for tumor growth suppression.
Collapse
MESH Headings
- 3T3 Cells/cytology
- 3T3 Cells/drug effects
- 3T3 Cells/metabolism
- 3T3 Cells/transplantation
- Animals
- Cell Cycle
- Cell Division
- Cell Transformation, Neoplastic/drug effects
- Cell Transformation, Neoplastic/genetics
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/physiology
- Doxycycline/pharmacology
- Estradiol/pharmacology
- Genes, erbB-1/drug effects
- Genes, erbB-2/drug effects
- Genes, myc/drug effects
- Genes, ras/drug effects
- Humans
- Interferon Regulatory Factor-1
- Interferon-beta/metabolism
- Male
- Mice
- Mice, Nude
- Neoplasms, Experimental/therapy
- Phenotype
- Phosphoproteins/genetics
- Phosphoproteins/physiology
- Promoter Regions, Genetic/drug effects
- Protein Structure, Tertiary
- Receptors, Estrogen/genetics
- Recombinant Fusion Proteins/biosynthesis
- Recombinant Fusion Proteins/physiology
- Transfection
- Tumor Stem Cell Assay
Collapse
Affiliation(s)
- Andrea Kröger
- Department of Gene Regulation and Differentiation, GBF - German Research Center for Biotechnology, Mascheroder Weg 1, D 38124 Braunschweig, Germany
| | | | | | | |
Collapse
|
159
|
Collet B, Hovens GCJ, Mazzoni D, Hirono I, Aoki T, Secombes CJ. Cloning and expression analysis of rainbow trout Oncorhynchus mykiss interferon regulatory factor 1 and 2 (IRF-1 and IRF-2). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2003; 27:111-126. [PMID: 12543125 DOI: 10.1016/s0145-305x(02)00072-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Two rainbow trout (Oncorhynchus mykiss) genes for interferon regulatory factors (IRF) 1 and 2 have been cloned and sequenced. The IRF-1 cDNA contains an open reading frame (ORF) of 996 nucleotides that translates into a 331 amino-acid putative peptide, with a 5' untranslated region (UTR) of 145bp and a 3' UTR of 481bp. The IRF-2 cDNA contains a 1035bp ORF that translates into a 344 amino-acid putative peptide, with a 5' UTR of 146bp and a 3' UTR of 925bp. In vivo, IRF-1 and IRF-2 are constitutively expressed in head kidney, gill and spleen but not liver. Both genes were induced in all the tissues examined. IRF-1 but not IRF-2 expression was significantly increased at the site of injection 1 week after DNA vaccination against viral haemorrhagic septicaemia virus. In vitro, IRF-1 and IRF-2 transcripts are present in unstimulated rainbow trout gonad cells and are up-regulated by poly I/C.
Collapse
Affiliation(s)
- Bertrand Collet
- Department of Zoology, Scottish Fish Immunology Research Centre, University of Aberdeen, Tillydrone Avenue, Aberdeen, AB24 2TZ, UK
| | | | | | | | | | | |
Collapse
|
160
|
Lee SH, Kim JW, Lee HW, Cho YS, Oh SH, Kim YJ, Jung CH, Zhang W, Lee JH. Interferon regulatory factor-1 (IRF-1) is a mediator for interferon-gamma induced attenuation of telomerase activity and human telomerase reverse transcriptase (hTERT) expression. Oncogene 2003; 22:381-91. [PMID: 12545159 DOI: 10.1038/sj.onc.1206133] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Constitutive activation of the telomerase is a key step in the development of human cancers. Interferon-gamma (IFN-gamma) signaling induces growth arrest in many tumors through multiple regulatory mechanisms. In this study, we show that IFN-gamma signaling represses telomerase activity and human telomerase reverse transcriptase (hTERT) transcription, and suggest that this signaling is mediated by IRF-1. Ectopic expression of IRF-1 attenuated hTERT promoter activity. Murine embryonic fibroblasts (MEFs) genetically deficient in IRF-1 (IRF-1(-/-)) showed an elevated level (>15 times) of hTERT promoter activity as compared to the hTERT promoter activity of wild-type MEFs. The telomerase activity and hTERT expression in IRF-1(-/-) MEFs were downregulated by IRF-1 transfection. Interestingly, less extent of telomerase repression was observed in HPV E6 and E7 negative, p53 mutant HT-3 cells than in HPV 18 E6 and E7 positive HeLa cells (intact p53). These findings provide evidence that IRF-1 is a potential mediator of IFN-gamma-induced attenuation of telomerase activity and hTERT expression.
Collapse
Affiliation(s)
- Seung-Hoon Lee
- Molecular Therapy Research Center, College of Medicine, Sungkyunkwan University, Samsung Medical Center Annex 8F, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
161
|
Wu MH, Yung BYM. UV stimulation of nucleophosmin/B23 expression is an immediate-early gene response induced by damaged DNA. J Biol Chem 2002; 277:48234-40. [PMID: 12374805 DOI: 10.1074/jbc.m206550200] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nucleophosmin/B23 (NPM/B23), a nucleolar protein, was rapidly up-regulated after UV irradiation (at 254 nm; 30 J/m(2)) in NIH 3T3 cells and HeLa/S3 cells. Levels of NPM/B23 mRNA peaked 45-60 min after UV treatment and returned to baseline by 12 h. Transcription inhibitor actinomycin D (5 microg/ml) prevented the UV-induced increase of NPM/B23 mRNA, suggesting that UV induction of NPM/B23 was mediated at the transcriptional level. Moreover, UV-induced NPM/B23 expression was super-induced by cycloheximide (20 microg/ml), which was characteristic of immediate-early gene response. The transcriptional activation of NPM/B23 by UV was also confirmed by NPM/B23 promoter activity assay. Thymine dinucleotide, mimicking the effects of UV-induced DNA damage, was able to trigger NPM/B23 expression in the absence of genomic DNA damage. UV-induced activation of NPM/B23 promoter could not be blocked by UV-inducible pathway inhibitors, such as those of growth factor tyrosine kinase, mitogen-activated protein kinase, AP-1, NF-kappaB, and DNA-dependent kinase. Our results indicate that UV stimulation of NPM/B23 expression may be mediated through a novel UV-inducible pathway and is an immediate-early gene response induced by damaged DNA. Induction of immediate-early gene is an initial step in the regulation of cellular and genomic responses to external stimuli. Our results thus provide important evidence for an involvement of NPM/B23 in the acute response of mammalian cells to environmental stress.
Collapse
Affiliation(s)
- Ming H Wu
- Graduate Institute of Pharmacology, National Yang Ming University, Taipei 112, Taiwan, Republic of China
| | | |
Collapse
|
162
|
Hu J, Angell JE, Zhang J, Ma X, Seo T, Raha A, Hayashi J, Choe J, Kalvakolanu DV. Characterization of monoclonal antibodies against GRIM-19, a novel IFN-beta and retinoic acid-activated regulator of cell death. J Interferon Cytokine Res 2002; 22:1017-26. [PMID: 12433281 DOI: 10.1089/107999002760624242] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
A combination of interferon-beta (IFN-beta) and all-trans retinoic acid (IFN/RA) induces tumor cell apoptosis via some unknown mechanisms. Apoptosis is a gene-directed process that limits the proliferation of undesired cells. Several genes are required to regulate cell death in the higher-order animals. Earlier, we employed a gene expression knockout technique to isolate cell death-related genes. A novel gene, the gene associated with retinoid-interferon-induced mortality-19 (GRIM-19), was found to be essential for tumor cell death induced by IFN/RA. Here, we describe the development and characterization of three monoclonal antibodies (mAbs) against GRIM-19. GRIM-19 is present in the nucleus and cytoplasm. Its expression is induced by the IFN/RA combination. We also show that GRIM-19 inhibits the cell-transforming property of viral oncogenic protein viral IFN regulatory factor-1 (vIRF-1) via a physical interaction. mAbs developed in this study should be useful for studying the other physiologic roles of GRIM-19 and serve as a potent tool for studying tumor responses to IFN/RA therapy.
Collapse
Affiliation(s)
- Jiadi Hu
- Marlene and Stewart Greenebaum Cancer Center, Department of Microbiology and Immunology, Molecular and Cellular Biology Program, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
163
|
Seo T, Lee D, Shim YS, Angell JE, Chidambaram NV, Kalvakolanu DV, Choe J. Viral interferon regulatory factor 1 of Kaposi's sarcoma-associated herpesvirus interacts with a cell death regulator, GRIM19, and inhibits interferon/retinoic acid-induced cell death. J Virol 2002; 76:8797-807. [PMID: 12163600 PMCID: PMC136415 DOI: 10.1128/jvi.76.17.8797-8807.2002] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2002] [Accepted: 06/04/2002] [Indexed: 11/20/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) plays a significant role in the development of Kaposi's sarcoma, primary effusion lymphoma, and some forms of multicentric Castleman's disease. The KSHV open reading frame K9 encodes the viral interferon (IFN) factor 1 (vIRF1), which downregulates IFN- and IRF-mediated transcriptional activation, and leads to cellular transformation in rodent fibroblasts and induction of tumors in nude mice. Using the yeast two-hybrid assay, we identified genes associated with retinoid-IFN-induced mortality-19 (GRIM19), which interacts directly with vIRF1, both in vivo and in vitro. The N-terminal region of vIRF1 is required for binding GRIM19. Colocalization of vIRF1 and GRIM19 was observed in 293T cells. The vIRF1 protein deregulates GRIM19-induced apoptosis in the presence of IFN/all-trans-retinoic acid (RA) and inhibits IFN/RA-induced cell death. Another DNA tumor viral protein, human papillomavirus type 16 E6, also binds GRIM19, suggesting that this is a general target of viral proteins. Our results collectively indicate that vIRF1 modulates IFN/RA-cell death signals via interactions with GRIM19.
Collapse
Affiliation(s)
- Taegun Seo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea
| | | | | | | | | | | | | |
Collapse
|
164
|
Kim EJ, Lee JM, Namkoong SE, Um SJ, Park JS. Interferon regulatory factor-1 mediates interferon-gamma-induced apoptosis in ovarian carcinoma cells. J Cell Biochem 2002; 85:369-80. [PMID: 11948692 DOI: 10.1002/jcb.10142] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Interferon-gamma (IFN-gamma), as one of interferon family that regulates antiviral, antiproliferative, and immunomodulatory responses, has been implicated for the growth regulation of ovarian cancer cells. However, the molecular mechanisms are not yet fully defined. To analyze detailed mechanisms, the ovarian cancer cell lines (2774, PA-1, OVCAR-3, and SKOV-3) were treated with IFN-gamma. The growth of 2774 was most effectively suppressed than that of other cells in both time-course and dose-dependent experiments. The order of sensitivity in other cells was PA-1 >> OVCAR-3 > SKOV-3 (not responded at all). The DNA fragmentation and DAPI staining assays suggested that the IFN-gamma-mediated cytotoxicity could be triggered by apoptosis. The treatment induced IFN regulatory factor-1 (IRF-1) in two IFN-gamma-sensitive cells (2774, PA-1), whereas IRF-1 was not induced in two IFN-gamma-resistant cells (OVCAR-3, SKOV-3). The levels of p53 and p21WAF1 were not strikingly changed in all four cells. Interestingly, the expression of interleukin-converting enzyme (ICE, or caspase-1) was increased by the treatment in a kinetically consistent manner to the induction of IRF-1. However, CD95 (Fas/APO-1) was not changed. Apoptosis was greatly induced, when IRF-1 was transiently expressed in PA-1 without the treatment of IFN-gamma. However, it was repressed when IRF-1 together with IRF-2, an antagonist of IRF-1, were coexpressed. In addition, the effect of IFN-gamma was reduced in the 2774 and PA-1 cells stably expressing either IRF-1 antisense or IRF-2 sense, as shown by the cytotoxicity and FACS analysis. Furthermore, the IFN-gamma-induced apoptosis was greatly reduced, when inhibitors of ICE were treated into PA-1 cells. Taken together, these results suggest that IRF-1 directly mediates the IFN-gamma-induced apoptosis via the activation of caspase-1 gene expression in IFN-gamma-sensitive ovarian cancer cells.
Collapse
Affiliation(s)
- Eun-Joo Kim
- Department of Medical Bioscience, Graduate School, Catholic University, Seoul, Korea
| | | | | | | | | |
Collapse
|
165
|
Rowland BD, Denissov SG, Douma S, Stunnenberg HG, Bernards R, Peeper DS. E2F transcriptional repressor complexes are critical downstream targets of p19(ARF)/p53-induced proliferative arrest. Cancer Cell 2002; 2:55-65. [PMID: 12150825 DOI: 10.1016/s1535-6108(02)00085-5] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The p16(INK4a)/pRB/E2F and p19(ARF)/p53 tumor suppressor pathways are disrupted in most human cancers. Both p19(ARF) and p53 are required for the induction of senescence in primary mouse embryonic fibroblasts (MEFs), but little is known about their downstream targets. Disruption of E2F-mediated transcriptional repression in MEFs caused a general increase in the expression of E2F target genes, including p19ARF. We detected no contribution of E2F-mediated transactivation in this setting, indicating that a predominant role of endogenous E2F in asynchronously growing primary MEFs is to repress its target genes. Moreover, relief of transcriptional repression by E2F rendered MEFs resistant to senescence induced by either p19(ARF), p53, or RAS(V12). Thus, E2F transcriptional repressor complexes are critical downstream targets of antiproliferative p19(ARF)/p53 signaling.
Collapse
Affiliation(s)
- Benjamin D Rowland
- Division of Molecular Carcinogenesis, Center for Biomedical Genetics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
166
|
Basler CF, García-Sastre A. Viruses and the type I interferon antiviral system: induction and evasion. Int Rev Immunol 2002; 21:305-37. [PMID: 12486817 DOI: 10.1080/08830180213277] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The type I interferon (IFN) system responds to viral infection and induces an "antiviral state" in cells, providing an important first line of defense against virus infection. Interaction of type I IFNs (IFN alpha and IFN beta) with their receptor induces hundreds of cellular genes. Of the proteins induced by IFN, the antiviral function of only a few is known, and their mechanisms of action are only partly understood. Additionally, although viral-encoded mechanisms that counteract specific components of the type I IFN response have been known for some time, it has recently become clear that many (if not most) viruses encode some form of IFN-antagonist. Understanding the interplay between viral-encoded IFN antagonists and the interferon response will be essential if the therapeutic potential of IFNs is to be fully exploited.
Collapse
|
167
|
Um SJ, Rhyu JW, Kim EJ, Jeon KC, Hwang ES, Park JS. Abrogation of IRF-1 response by high-risk HPV E7 protein in vivo. Cancer Lett 2002; 179:205-12. [PMID: 11888675 DOI: 10.1016/s0304-3835(01)00871-0] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have previously reported that human papillomavirus (HPV) E7 interacts with IRF-1, a key regulator of cellular immune response, and abrogates its transactivation function at the molecular level in vitro. To confirm our previous data, we investigated in vivo the E7-mediated down-regulation of IRF-1 using HPV E7-inducible cells and transgenic mice expressing HPV-18 E6/E7. When E7 was induced in the absence of tetracycline, the expression of target genes of IRF-1 (TAP-1, IFN-beta, MCP-1 that are important for cellular immunity) was clearly reduced as determined by RT-PCR. In addition, the IRF-1 activity was down-regulated in E7-expressing cells into which IFN-beta-CAT reporter plasmid was transfected. To further investigate the E7-mediated immune regulation in vivo, we constructed transgenic mice expressing E6 and E7 genes of HPV-18 under the control of HPV-18 promoter (URR). From several rounds of breeding, we obtained from a transgenic line that developed a cervical dysplasia and expressed E6/E7 as determined by histological examination and RT-PCR, respectively. Subsequent RT-PCR analysis indicated that TAP-1, IFN-beta, and MCP-1 genes were less expressed in a cervical tissue derived from transgenic mouse, when compared with a cervix derived from normal mouse. From these results, we conclude that the E7 transgene expression inactivates the transactivation function of IRF-1 in vivo, which might be important for the elucidation of the E7-mediated immune evading mechanism that is frequently found in cervical cancer.
Collapse
Affiliation(s)
- Soo-Jong Um
- Department of Bioscience and Biotechnology/Institute of Bioscience, Sejong University, Seoul, South Korea
| | | | | | | | | | | |
Collapse
|
168
|
Ma P, Magut M, Chen X, Chen CY. P53 is necessary for the apoptotic response mediated by a transient increase of Ras activity. Mol Cell Biol 2002; 22:2928-38. [PMID: 11940651 PMCID: PMC133752 DOI: 10.1128/mcb.22.9.2928-2938.2002] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The tumor suppressor p53 eliminates cancer-prone cells via multiple mechanisms, including apoptosis. Ras elicits apoptosis in cells after protein kinase C (PKC) downregulation. However, the role of p53 in Ras-mediated apoptosis has not been fully investigated. Here, we demonstrate that mouse fibroblasts that express wild-type p53 are more susceptible to apoptosis elicited by PKC inhibition if Ras is transiently expressed or upregulated as opposed to stably expressed. In the latter case, p53 is frequently mutated. Transiently increased Ras activity induces Bax, and PKC inhibition augments this induction. Overexpression of E6 inactivates p53 and thereby suppresses both Bax induction and apoptosis. In contrast, Bax is not induced in stable ras transfectants, regardless of PKC inhibition. The data suggest that short- and long-term activation of Ras use a different mechanism(s) to initiate apoptosis. The status of p53 may contribute to such differences.
Collapse
Affiliation(s)
- Peihong Ma
- Cancer Research Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | |
Collapse
|
169
|
Liebermann DA, Hoffman B. Myeloid differentiation (MyD)/growth arrest DNA damage (GADD) genes in tumor suppression, immunity and inflammation. Leukemia 2002; 16:527-41. [PMID: 11960329 DOI: 10.1038/sj.leu.2402477] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2001] [Accepted: 01/16/2002] [Indexed: 12/16/2022]
Abstract
Myeloid differentiation (MyD) primary response and growth arrest DNA damage (Gadd) genes comprise a set of overlapping genes, including known (IRF-1, EGR-1, Jun) and novel (MyD88, Gadd45alpha, MyD118/Gadd45beta, GADD45gamma, MyD116/ Gadd34) genes, that have been cloned by virtue of being co-ordinately induced upon the onset of terminal myeloid differentiation and following exposure of cells to stress stimuli. In recent years it has become evident that MyD/Gadd play a role in blood cell development, where they function as positive regulators of terminal differentiation, lineage-specific blood cell development and control of blood cell homeostasis, including growth inhibition and apoptosis. MyD/Gadd are also involved in inflammatory responses to invading micro-organisms, and response to environmental stress and physiological stress, such as hypoxia, which results in ischemic tissue damage. An intricate network of interactions among MyD/GADD genes and gene products appears to control their diverse functions. Deregulated growth, increased cell survival, compromised differentiation and deficiencies in DNA repair are hallmarks of malignancy and its progression. Thus, the role MyD/Gadd play in negative growth control, including cell cycle arrest and apoptosis, and in DNA repair, make them attractive molecular targets for tumor suppression. The role MyD/Gadd play in innate immunity and host response to hypoxia also make these genes and gene products attractive molecular targets to treat immunity and inflammation disorders, such as septic shock and ischemic tissue damage.
Collapse
Affiliation(s)
- D A Liebermann
- Fels Institute for Cancer Research and Molecular Biology and the Department of Biochemistry, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | |
Collapse
|
170
|
Guidotti LG, Morris A, Mendez H, Koch R, Silverman RH, Williams BRG, Chisari FV. Interferon-regulated pathways that control hepatitis B virus replication in transgenic mice. J Virol 2002; 76:2617-21. [PMID: 11861827 PMCID: PMC135990 DOI: 10.1128/jvi.76.6.2617-2621.2002] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
We previously showed that the intrahepatic induction of cytokines such as alpha/beta interferon (IFN-alpha/beta) and gamma interferon (IFN-gamma) inhibits hepatitis B virus (HBV) replication noncytopathically in the livers of transgenic mice. The intracellular pathway(s) responsible for this effect is still poorly understood. To identify interferon (IFN)-inducible intracellular genes that could play a role in our system, we crossed HBV transgenic mice with mice deficient in IFN regulatory factor 1 (IRF-1), the double-stranded RNA-activated protein kinase (PKR), or RNase L (RNase L) (IRF-1(-/-), PKR(-/-), or RNase L(-/-) mice, respectively), three well-characterized IFN-inducible genes that mediate antiviral activity. We showed that unmanipulated IRF-1(-/-) or PKR(-/-) transgenic mice replicate HBV in the liver at slightly higher levels than the respective controls, suggesting that both IRF-1 and PKR individually appear to mediate signals that modulate HBV replication under basal conditions. These same animals were responsive to the antiviral effects of the IFN-alpha/beta inducer poly(I-C) or recombinant murine IFN-gamma, suggesting that under these conditions, either the IRF-1 or the PKR genes can mediate the antiviral activity of the IFNs or other IFN-inducible genes mediate the antiviral effects. Finally, RNase L(-/-) transgenic mice were undistinguishable from controls under basal conditions and after poly(I-C) or IFN-gamma administration, suggesting that RNase L does not modulate HBV replication in this model.
Collapse
Affiliation(s)
- Luca G Guidotti
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California 92037, USA.
| | | | | | | | | | | | | |
Collapse
|
171
|
Peeper DS, Shvarts A, Brummelkamp T, Douma S, Koh EY, Daley GQ, Bernards R. A functional screen identifies hDRIL1 as an oncogene that rescues RAS-induced senescence. Nat Cell Biol 2002; 4:148-53. [PMID: 11812999 DOI: 10.1038/ncb742] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Primary fibroblasts respond to activated H-RAS(V12) by undergoing premature arrest, which resembles replicative senescence. This irreversible 'fail-safe mechanism' requires p19(ARF), p53 and the Retinoblastoma (Rb) family: upon their disruption, RAS(V12)-expressing cells fail to undergo senescence and continue to proliferate. Similarly, co-expression of oncogenes such as c-MYC or E1A rescues RAS(V12)-induced senescence. To identify novel genes that allow escape from RAS(V12)-induced senescence, we designed an unbiased, retroviral complementary DNA library screen. We report on the identification of DRIL1, the human orthologue of the mouse Bright and Drosophila dead ringer transcriptional regulators. DRIL1 renders primary murine fibroblasts unresponsive to RAS(V12)-induced anti-proliferative signalling by p19(ARF)/p53/p21(CIP1), as well as by p16(INK4a). In this way, DRIL1 not only rescues RAS(V12)-induced senescence but also causes these fibroblasts to become highly oncogenic. Furthermore, DRIL1 immortalizes mouse fibroblasts, in the presence of high levels of p16(INK4a). Immortalization by DRIL1, whose product binds the pRB-controlled transcription factor E2F1 (ref. 8), is correlated with induction of E2F1 activity. Correspondingly, DRIL1 induces the E2F1 target Cyclin E1, overexpression of which is sufficient to trigger escape from senescence. Thus, DRIL1 disrupts cellular protection against RAS(V12)-induced proliferation downstream of the p19(ARF)/p53 pathway.
Collapse
Affiliation(s)
- Daniel S Peeper
- Division of Molecular Carcinogenesis and Center for Biomedical Genetics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
172
|
Taniguchi T, Takaoka A. The interferon-alpha/beta system in antiviral responses: a multimodal machinery of gene regulation by the IRF family of transcription factors. Curr Opin Immunol 2002; 14:111-6. [PMID: 11790540 DOI: 10.1016/s0952-7915(01)00305-3] [Citation(s) in RCA: 374] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The efficient induction of interferons alpha and beta (IFN-alpha/beta) in virus-infected cells is central to the antiviral response of a host and is regulated mainly at the level of gene transcription. Once produced, IFN-alpha/beta transmit signals to the cell interior via a specific receptor complex to induce an antiviral response. Recently, the auto-amplification mechanism of the IFN-alpha/beta system that follows viral infection has been identified. This mechanism is mediated by transcription factors of the IFN regulatory factor family and, in fact, may have evolved to render the system more robust in antiviral responses.
Collapse
Affiliation(s)
- Tadatsugu Taniguchi
- Department of Immunology, Faculty of Medicine and Graduate School of Medicine, University of Tokyo, Hongo 7-3-1, Bunkyo-ku, 113-0033, Tokyo, Japan.
| | | |
Collapse
|
173
|
Kirchhoff S, Sebens T, Baumann S, Krueger A, Zawatzky R, Li-Weber M, Meinl E, Neipel F, Fleckenstein B, Krammer PH. Viral IFN-regulatory factors inhibit activation-induced cell death via two positive regulatory IFN-regulatory factor 1-dependent domains in the CD95 ligand promoter. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:1226-34. [PMID: 11801659 DOI: 10.4049/jimmunol.168.3.1226] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The CD95 (also called APO-1/Fas) system plays a major role in the induction of apoptosis in lymphoid and nonlymphoid tissues. The CD95 ligand (CD95L) is induced in response to a variety of signals, including IFN-gamma and TCR/CD3 stimulation. Here we report the identification of two positive regulatory IFN-regulatory factor-dependent domains (PRIDDs) in the CD95L promoter and its 5' untranslated region, respectively. EMSAs demonstrate specific binding of IFN-gamma-induced IFN-regulatory factor 1 (IRF-1) to the PRIDD sequences. Ectopic IRF-1 expression induces CD95L promoter activity. Furthermore, we demonstrate that PRIDDs play an important role in TCR/CD3-mediated CD95L induction. Most interestingly, viral IRFs of human herpes virus 8 (HHV8) totally abolish IRF-1-mediated and strongly reduce TCR/CD3-mediated CD95L induction. We demonstrate here for the first time that viral IRFs inhibit activation-induced cell death. Thus, these results demonstrate an important mechanism of HHV8 to modulate the immune response by down-regulation of CD95L expression. Inhibition of CD95-dependent T cell function might contribute to the immune escape of HHV8.
Collapse
Affiliation(s)
- Sabine Kirchhoff
- Tumor Immunology Program, Tumorvirus-Immunology, German Cancer Research Center, D-16920 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Mochizuki T, Asai A, Saito N, Tanaka S, Katagiri H, Asano T, Nakane M, Tamura A, Kuchino Y, Kitanaka C, Kirino T. Akt protein kinase inhibits non-apoptotic programmed cell death induced by ceramide. J Biol Chem 2002; 277:2790-7. [PMID: 11706021 DOI: 10.1074/jbc.m106361200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A growing body of evidence now suggests that programmed cell death (PCD) occurs via non-apoptotic mechanisms as well as by apoptosis. In contrast to apoptosis, however, the molecular mechanisms involved in the regulation of non-apoptotic PCD remain only poorly understood. Here we show that ceramide induces a non-apoptotic PCD with a necrotic-like morphology in human glioma cells. Characteristically, the cell death was not accompanied by loss of the mitochondrial transmembrane potential, cytosolic release of cytochrome c from mitochondria, or the activation of the caspase cascade. Consistent with these characteristics, this ceramide-induced cell death was inhibited neither by the overexpression of Bcl-xL nor by the pan-caspase inhibitor zVAD-fmk. However, strikingly, the ceramide-induced non-apoptotic cell death was inhibited by the activation of the Akt/protein kinase B pathway through the expression of a constitutively active version of Akt. The results for the first time indicate that the Akt kinase, known to play an essential role in survival factor-mediated inhibition of apoptotic cell death, is also involved in the regulation of non-apoptotic PCD.
Collapse
Affiliation(s)
- Toshihiro Mochizuki
- Laboratory for Neuroscience and Neuro-oncology, Department of Neurosurgery, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
175
|
McCurrach ME, Lowe SW. Methods for studying pro- and antiapoptotic genes in nonimmortal cells. Methods Cell Biol 2002; 66:197-227. [PMID: 11396004 DOI: 10.1016/s0091-679x(01)66010-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- M E McCurrach
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | | |
Collapse
|
176
|
Carvalhal AV, Coroadinha A, Alves PM, Moreira JL, Hauser H, Carrondo MJ. Metabolic changes during cell growth inhibition by the IRF-1 system. Enzyme Microb Technol 2002. [DOI: 10.1016/s0141-0229(01)00460-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
177
|
Romeo G, Fiorucci G, Chiantore MV, Percario ZA, Vannucchi S, Affabris E. IRF-1 as a negative regulator of cell proliferation. J Interferon Cytokine Res 2002; 22:39-47. [PMID: 11846974 DOI: 10.1089/107999002753452647] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Numerous evidence has demonstrated the involvement in growth control of interferon (IFN) regulatory factor-1 (IRF-1), which shows tumor suppressor activity. IRF-1 is a well-studied member of the IRF transcription factors that reveals functional diversity in the regulation of cellular response by activating expression of a diverse set of target genes, depending on the cell type and on the specific stimuli. IRF-1 gene rearrangements may be a crucial point in the pathogenesis of some cancer types. Furthermore, different aspects of the tumor suppressor function of IRF-1 may be explained, at least in part, by the observations that IRF-1 is a regulator of cell cycle and apoptosis and that its inactivation accelerates cell transformation. Studies on gene knockout mice contributed greatly to the clarification of these multiple IRF-1 functions. We summarize our current knowledge of the antigrowth effect of IRF-1, focusing also on a more general involvement of IRF-1 in mediating negative regulation of cell growth induced by numerous cytokines and other biologic response modifiers.
Collapse
Affiliation(s)
- Giovanna Romeo
- Laboratory of Virology, Istituto Superiore di Sanità, Rome, Italy.
| | | | | | | | | | | |
Collapse
|
178
|
Abstract
Interferon (IFN) regulatory factors (IRF) are a family of transcription factors with multiple functions. IRF-7 was initially cloned within the biologic context of Epstein-Barr virus (EBV) latency and discovered to have an intimate relation with the EBV primary oncogenic protein, latent membrane protein-1 (LMP-1). EBV regulates and uses IRF-7 as a secondary mediator for several target genes involved in latency and immune regulation. Other than its functions in EBV latency, IRF-7 has been identified as one of the major players in virally induced IFN production that is central to innate immunity. Thus, IRF-7 plays important roles in a variety of biologic systems.
Collapse
Affiliation(s)
- Luwen Zhang
- Nebraska Center for Virology, UNL Biological Sciences, University of Nebraska, Lincoln, NE 68588, USA.
| | | |
Collapse
|
179
|
Abstract
Autonomous cell proliferation is one of the hallmarks of cancer cells, driven by activated growth-promoting oncogenes. However, deregulated activation of these oncogenes also triggers apoptosis via multiple pathways. Among them, the ARF-p53 pathway appears to play a major role in mediating oncogene-induced apoptosis. Consequently, suppression of apoptosis by inactivation of p53 and other tumor suppressors is central to tumor development. These findings have broad implications in understanding cancer genetics and therapy. They help define the roles for oncogenes and tumor suppressor genes in tumorigenesis. Furthermore, the notion that cancer cells often carry specific defects in apoptotic pathways but are inherently sensitive to apoptosis as a result of deregulated proliferation, offers numerous opportunities for manipulating apoptosis in directions of clinical application.
Collapse
Affiliation(s)
- Han-Fei Ding
- Department of Biochemistry and Molecular Biology, Medical College of Ohio, Toledo, OH, USA
| | | |
Collapse
|
180
|
Abstract
Interferon (IFN) regulatory factor-1 (IRF-1) was isolated by virtue of its affinity to specific DNA sequences in the IFN-beta promoter that mediate virus responsiveness. IRF-1 was the first factor identified of the IRF family and was most extensively characterized at the molecular level. Also, its physiologic role in host defense against pathogens, tumor prevention, and development of the immune system was investigated in detail. Even though some of the functions first associated with IRF-1 were later found to be mediated in part or predominantly by other activators of the IRF family of transcription factors, IRF-1 has remained a central paradigm in the transcriptional regulation of the IFN response.
Collapse
Affiliation(s)
- Andrea Kröger
- Department of Gene Regulation and Differentiation, GBF, Gesellschaft für Biotechnologische Forschung, D 38124 Braunschweig Mascheroder Weg 1, Germany
| | | | | | | | | |
Collapse
|
181
|
Carvalhal AV, Moreira JL, Carrondo MJ. Strategies to modulate BHK cell proliferation by the regulation of IRF-1 expression. J Biotechnol 2001; 92:47-59. [PMID: 11604172 DOI: 10.1016/s0168-1656(01)00365-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Activation of the constitutively expressed interferon-regulatory-factor-1/estrogen receptor fusion protein (IRF-1-hER) in BHK cells was accomplished through the addition of estradiol to the culture medium, which enabled IRF-1 to gain its transcriptional activator function and inhibit cell growth. With the addition of 100 nM estradiol at the beginning of the exponential phase of a cell suspension culture, IRF-1 activation led to a rapid cell growth inhibition but also to a significant decrease in cell viability. To apply this concept in industry, a reduction of the time span of estradiol exposure is required. Cycles of estradiol addition and removal were performed in 2-l stirred tank bioreactors operated under perfusion, where an initial step addition of 100 nM estradiol was performed, followed, after 48-72 h, by a slow dilution with estradiol-free fresh medium (perfusion rate varying between 0.7 and 1.4 per day). Cell growth inhibition was successfully achieved for three consecutive cycles. Diluting the estradiol by perfusing medium without estradiol to concentrations lower than 10 nM led to cell growth and viability recovery independently of the perfusion rate used. These observations permitted the definition of operational strategies for regulated IRF-1 BHK cell growth by pulse estradiol addition, followed by a period of 48 h in the presence of estradiol and by fast perfusion to estradiol concentrations lower than 10 nM. Cell growth response to IRF-1 activation and following estradiol removal by perfusion was also evaluated with an IRF-1-hER regulated clone expressing constitutively Factor VII, where the time of estradiol exposure and perfusion rate were varied. This clone presented a stronger response to IRF-1 activation without an increase in Factor VII specific productivity after cell growth inhibition; this clearly indicates that the stationary phase obtained is clone dependent. This work proves that it is possible to modulate the IRF-1 effect for cell growth control by the manipulation of cycles of addition and removal of estradiol, potentially representing a new generation of culture procedures for controlled growth production purposes.
Collapse
Affiliation(s)
- A V Carvalhal
- Instituto de Biologia Experimental e Tecnológica/Instituto de Tecnologia Química e Biológica IBET/ITQB, Apartado 12, P-2781-901 Oeiras, Portugal
| | | | | |
Collapse
|
182
|
Abstract
The inflammatory cytokine interferon gamma (IFNgamma) can cause cell cycle arrest and apoptosis in the hepatocyte. Primarily these processes are protective but in chronic liver disease oncogenic mutations may prosper. IFNgamma signalling is discussed showing how p53 is induced to cause cell cycle arrest. While caspases are are known to be responsible for IFNgamma induced apoptosis, how they are activated is unclear. Potential mechanisms are reviewed.
Collapse
Affiliation(s)
- B J Tura
- Cell Injury and Apoptosis Group, (Department of Pathology), MRC Centre for Inflammation Research, University of Edinburgh Medical School, Edinburgh, UK
| | | | | |
Collapse
|
183
|
Zhang L, Pagano JS. Interferon regulatory factor 7: a key cellular mediator of LMP-1 in EBV latency and transformation. Semin Cancer Biol 2001; 11:445-53. [PMID: 11669606 DOI: 10.1006/scbi.2001.0411] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Interferon regulatory factor 7 (IRF-7) was cloned within the biological context of Epstein-Barr virus (EBV) latency, and has an intimate relation with EBV. EBV latent membrane protein 1 (LMP-1) regulates IRF-7 both by inducing the expression of IRF-7 and by activating IRF-7 protein through phosphorylation and nuclear translocation in a post-translational manner. The activated IRF-7 then functions to regulate both EBV and cellular target genes involved in latency, transformation and immune regulation. IRF-7 appears to be a key cellular latency protein involved in both the pathogenesis and persistence of EBV infection.
Collapse
MESH Headings
- Cell Transformation, Viral/immunology
- Cell Transformation, Viral/physiology
- DNA-Binding Proteins/metabolism
- Gene Expression Regulation, Viral
- Herpesvirus 4, Human/genetics
- Herpesvirus 4, Human/immunology
- Herpesvirus 4, Human/metabolism
- Herpesvirus 4, Human/physiology
- Humans
- Interferon Regulatory Factor-7
- Models, Biological
- Oncogenes/physiology
- Promoter Regions, Genetic/genetics
- Protein Processing, Post-Translational
- Viral Matrix Proteins/chemistry
- Viral Matrix Proteins/genetics
- Viral Matrix Proteins/metabolism
- Virus Latency/physiology
Collapse
Affiliation(s)
- L Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill 27599-7295, USA.
| | | |
Collapse
|
184
|
Jiang MC, Lin TL, Lee TL, Huang HT, Lin CL, Liao CF. IRF-1-mediated CAS expression enhances interferon-gamma-induced apoptosis of HT-29 colon adenocarcinoma cells. MOLECULAR CELL BIOLOGY RESEARCH COMMUNICATIONS : MCBRC 2001; 4:353-8. [PMID: 11703094 DOI: 10.1006/mcbr.2001.0303] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The expression of CAS is reported to be upregulated in a variety of human tumor cells, and such expression correlates with the development of tumors. CAS also plays a role in apoptosis. We investigated whether CAS expression affects the susceptibility of tumor cells to IFN-gamma-induced apoptosis. Our data show that IFN-gamma treatment induces CAS expression in HT-29 tumor cells. IFN-gamma-induced gene expression is primarily mediated by the transcriptional factor, IRF-1. Our data show that IRF-1 mediates IFN-gamma-induced CAS expression. Transfection of HT-29 cells with CAS expression vector did not induce apoptosis of cells; nevertheless, CAS overexpression greatly enhanced IFN-gamma-induced apoptosis of cells. CPP32 is regarded as one of the central apoptosis executioner molecules. CAS overexpression enhances IFN-gamma-induced CPP32 expression. These results indicate that tumor cells highly expressing CAS may be more susceptible to apoptosis induced by reagents that are capable of inducing CAS expression. Thus, CAS may be a target for the elimination of tumors.
Collapse
Affiliation(s)
- M C Jiang
- Institute of Zoology, Academia Sinica, Taipei 11529, Taiwan, Republic of China
| | | | | | | | | | | |
Collapse
|
185
|
Mizukami Y, Ura H, Obara T, Habiro A, Izawa T, Osanai M, Yanagawa N, Tanno S, Kohgo Y. Requirement of c-jun N-terminal kinase for apoptotic cell death induced by farnesyltransferase inhibitor, farnesylamine, in human pancreatic cancer cells. Biochem Biophys Res Commun 2001; 288:198-204. [PMID: 11594773 DOI: 10.1006/bbrc.2001.5744] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Farnesyltransferase inhibitors (FTIs) represent a novel class of anticancer drugs and are now in clinical trial. We have previously shown that farnesylamine, synthetic isoprenoid-linked with "amine" which acts as a potent FTI, induces apoptosis in human pancreatic cancer cells through the ras signaling cascade. Since the effect of FTI is usually "cytostatic" rather than "cytotoxic", we speculated another apoptotic machinery of farnesylamine in addition to the effect of FTI. Farnesylamine induced sustained activation of c-jun N-terminal kinase (JNK), which was not caused by other FTI, FTI-277. Blockage of JNK activity by dominant-negative mutant abrogated the DNA laddering and significantly reduced "cytotoxic" effect of farnesylamine. Strikingly similar effect on JNK activation and apoptosis was induced by structurally related long-chain fatty amine (LFA), oleylamine, but not by farnesol, an isoprenoid analogue of farnesylamine without "amine." Taken together, apoptosis induction through JNK activation by farnesylamine based on the LFA structure rather than an effect of FTI.
Collapse
Affiliation(s)
- Y Mizukami
- Third Department of Internal Medicine, Asahikawa Medical College, 2-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
186
|
Preisler HD, Li B, Chen H, Fisher L, Nayini J, Raza A, Creech S, Venugopal P. P15INK4B gene methylation and expression in normal, myelodysplastic, and acute myelogenous leukemia cells and in the marrow cells of cured lymphoma patients. Leukemia 2001; 15:1589-95. [PMID: 11587217 DOI: 10.1038/sj.leu.2402211] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
P15INK4B methylation and expression was studied in bone marrow cells obtained from normal individuals, from patients who had been cured of lymphoma, and from patients with either MDS or AML. The level of p15 methylation was very low in normal BM cells and in CD34+ and CD34- subpopulations (0-6.5%; med, = 2.5%). P15INK4B transcripts were present in each of these cell populations. In contrast, methylation was the usual situation in MDS and AML marrows. The presence of methylation of the p15INK4B gene did not always indicate an absence of expression nor was expression always present if methylation was absent. P15INK4B methylation was studied in the marrows of nine patients (one studied twice) who had been cured of lymphoma and in whom hemopoiesis was believed to be normal. Increased methylaton was present in all 10 marrows. These data indicate that p15INK4B methylation is likely to be a very early event in the development of the secondary hematologic disorders.
Collapse
Affiliation(s)
- H D Preisler
- Rush Cancer Institute, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | | | |
Collapse
|
187
|
Kuniyasu H, Yasui W, Pettaway CA, Yano S, Oue N, Tahara E, Fidler IJ. Interferon-alpha prevents selection of doxorubicin-resistant undifferentiated-androgen-insensitive metastatic human prostate cancer cells. Prostate 2001; 49:19-29. [PMID: 11550207 DOI: 10.1002/pros.1114] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND We determined whether treatment of metastatic prostate cancer cells with doxorubicin (DOX) and interferon-alpha (IFN-alpha) prevented the emergence of highly undifferentiated tumor cells. METHODS The state of cell differentiation was determined by analysis of prostate-specific antigen (PSA), E-cadherin, keratin, and vimentin. RESULTS Human prostate cancer LNCaP-LN3 cells growing in culture as multicell spheroids expressed higher levels of E-cadherin and E-cadherin-associated beta-catenin than LNCaP-LN3 cells growing as monolayers. Treatment of cells with DOX downregulated PSA, E-cadherin, and keratin, and upregulated expression of vimentin and vascular endothelial growth factor (VEGF) mRNA. While treatment of cells with IFN-alpha did not alter gene expression, the addition of IFN-alpha to cultures treated with DOX produced synergistic toxicity and abrogated the changes in gene expression observed in cells treated with DOX alone. CONCLUSIONS Treatment with IFN-alpha and DOX should be further explored as a therapeutic strategy for androgen-insensitive prostate cancer.
Collapse
Affiliation(s)
- H Kuniyasu
- Department of Oncological Pathology, Cancer Center, Nara Medical University, Kashihara, Japan
| | | | | | | | | | | | | |
Collapse
|
188
|
Jarosinski KW, Whitney LW, Massa PT. Specific deficiency in nuclear factor-kappaB activation in neurons of the central nervous system. J Transl Med 2001; 81:1275-88. [PMID: 11555675 DOI: 10.1038/labinvest.3780341] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The expression and activation of nuclear factor-kappaB (NF-kappaB) in neurons and glia of the central nervous system (CNS) has been intensely investigated because of its potential importance in understanding how this multifunctional transcription factor controls developmental and pathological processes. In particular, there has been interest in how NF-kappaB may be differentially regulated in these two major functional subgroups of cells in the CNS to provide for specific responses to various stimuli. Of special interest are responses to both proinflammatory cytokines and microbial products that signal from specific cell receptors to activate NF-kappaB. In the present studies, both neurons and glia (astrocytes) in vivo expressed latent cytoplasmic NF-kappaB analyzed by immunofluorescence microscopy and electrophoretic mobility shift analysis. In vitro, neurons and astrocytes expressed comparable levels of latent NF-kappaB molecules, but NF-kappaB nuclear localization stimulated by proinflammatory cytokines or microbial products was markedly deficient in neurons. In accord with this finding, the rapid degradation of inhibitor of NF-kappaB alpha (IkappaBalpha) that is seen in astrocytes did not occur in neurons in response to these agents. However, long-term exposure to translational inhibitors resulted in IkappaBalpha decay and activation of latent NF-kappaB in neurons, indicating potential NF-kappaB activity in these cells. Analysis of NF-kappaB-responsive interferon regulatory factor-1 gene expression indicated that increased nuclear NF-kappaB in neurons had transcriptional potential. We conclude that mechanisms responsible for inducible targeting of IkappaBalpha are uniquely regulated in neurons and account for the hypo-responsiveness of these cells to signals generated during microbial infections in the CNS. Thus, modulation of signals that target IkappaBalpha degradation may be unique and a key component of specific NF-kappaB regulation in neurons.
Collapse
Affiliation(s)
- K W Jarosinski
- Department of Neurology, State University of New York Health Science Center, Syracuse, New York 13210, USA
| | | | | |
Collapse
|
189
|
Preisler HD, Perambakam S, Li B, Hsu WT, Venugopal P, Creech S, Sivaraman S, Tanaka N. Alterations in IRF1/IRF2 expression in acute myelogenous leukemia. Am J Hematol 2001; 68:23-31. [PMID: 11559933 DOI: 10.1002/ajh.1144] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The interferon response genes 1 and 2 have been shown to be involved in the regulation of differentiation and proliferation of cells of the myeloid series, with the former functioning as an anti-oncogene and the latter as an oncogene. In the study described here, the levels of expression of these two genes and the ratio of their expression were compared in AML and normal marrow. The ratio of gene expression was significantly less in AML marrow cells as compared to normal marrow cells [med ratio = 1.33 vs. 2.97, P = 0.003]. While the expression ratio was unaffected by the presence or absence of either ras or fms mutations, p53 mutations were associated with higher IRF1:IRF2 expression ratios that wt p53 genes [med = 1.701 vs. 1.135, P = 0.014]. Given the functional characteristics and the competitive nature of these two genes, it is possible that leukemic transformation is associated with a fall in IRF1:IRF2 ratios. Finally, the administration of IL4 can result in the normalization of the IRF1:IRF2 ratio in the marrow cells of some patients with AML.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Bone Marrow Cells/metabolism
- Cytogenetic Analysis
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Genes, ras
- Humans
- Interferon Regulatory Factor-1
- Interferon Regulatory Factor-2
- Interleukin-4/therapeutic use
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Middle Aged
- Phosphoproteins/genetics
- Phosphoproteins/metabolism
- RNA, Messenger/metabolism
- Receptor, Macrophage Colony-Stimulating Factor/genetics
- Reference Values
- Repressor Proteins
- Transcription Factors
- Treatment Outcome
- Tumor Suppressor Protein p53/genetics
Collapse
Affiliation(s)
- H D Preisler
- Rush Cancer Institute, Rush Presbyterian St. Luke's Medical Center, Chicago, Illinois 60612, USA.
| | | | | | | | | | | | | | | |
Collapse
|
190
|
Lee J, Hur J, Lee P, Kim JY, Cho N, Kim SY, Kim H, Lee MS, Suk K. Dual role of inflammatory stimuli in activation-induced cell death of mouse microglial cells. Initiation of two separate apoptotic pathways via induction of interferon regulatory factor-1 and caspase-11. J Biol Chem 2001; 276:32956-65. [PMID: 11402054 DOI: 10.1074/jbc.m104700200] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
We have previously shown that mouse microglial cells undergo apoptosis upon inflammatory activation and that nitric oxide (NO) is the major autocrine mediator in this process (Lee, P., Lee, J., Kim, S., Yagita, H., Lee, M. S., Kim, S. Y., Kim, H., and Suk, K. (2001) Brain Res. 892, 380-385). Here, we present evidence that interferon regulatory factor-1 (IRF-1) and caspase-11 are the essential molecules in activation-induced cell death of microglial cells. The apoptogenic action of inflammatory stimuli such as lipopolysaccharide (LPS) and interferon-gamma (IFNgamma) was mediated through the induction of IRF-1 and caspase-11 expression in two separate events. Although IRF-1 was required for NO synthesis, caspase-11 induction was necessary for NO-independent apoptotic pathway. Microglial cells from IRF-1-deficient mice showed markedly decreased NO production, and they were partially resistant to apoptosis in response to LPS/IFNgamma but were sensitive to NO donor exposure. LPS/IFNgamma treatment resulted in the induction of caspase-11 followed by activation of caspase-11, -1, and -3. Inactivation of caspase-11 by the transfection of dominant-negative mutant or treatment with the caspase inhibitors rendered microglial cells partially resistant to LPS/IFNgamma-induced apoptosis. Inhibition of both NO synthesis and caspase-11 completely blocked LPS/IFNgamma-induced cytotoxicity. These results indicated that LPS/IFNgamma not only induced the production of cytotoxic NO through IRF-1 but also initiated the NO-independent apoptotic pathway through the induction of caspase-11 expression.
Collapse
Affiliation(s)
- J Lee
- Graduate School of East-West Medical Science, Kyunghee University, Seoul 130-701, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Taniguchi T, Ogasawara K, Takaoka A, Tanaka N. IRF family of transcription factors as regulators of host defense. Annu Rev Immunol 2001; 19:623-55. [PMID: 11244049 DOI: 10.1146/annurev.immunol.19.1.623] [Citation(s) in RCA: 1287] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Interferon regulatory factors (IRFs) constitute a family of transcription factors that commonly possess a novel helix-turn-helix DNA-binding motif. Following the initial identification of two structurally related members, IRF-1 and IRF-2, seven additional members have now been reported. In addition, virally encoded IRFs, which may interfere with cellular IRFs, have also been identified. Thus far, intensive functional analyses have been done on IRF-1, revealing a remarkable functional diversity of this transcription factor in the regulation of cellular response in host defense. Indeed, IRF-1 selectively modulates different sets of genes, depending on the cell type and/or the nature of cellular stimuli, in order to evoke appropriate responses in each. More recently, much attention has also been focused on other IRF family members. Their functional roles, through interactions with their own or other members of the family of transcription factors, are becoming clearer in the regulation of host defense, such as innate and adaptive immune responses and oncogenesis.
Collapse
Affiliation(s)
- T Taniguchi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan.
| | | | | | | |
Collapse
|
192
|
Affiliation(s)
- I Palmero
- Department of Immunology and Oncology, National Center of Biotechnology, Madrid E-28049, Spain
| | | |
Collapse
|
193
|
Conde C, Mark M, Oliver F, Huber A, de Murcia G, Ménissier-de Murcia J. Loss of poly(ADP-ribose) polymerase-1 causes increased tumour latency in p53-deficient mice. EMBO J 2001; 20:3535-43. [PMID: 11432840 PMCID: PMC125506 DOI: 10.1093/emboj/20.13.3535] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
PARP-1-deficient mice display a severe defect in the base excision repair pathway leading to radiosensitivity and genomic instability. They are protected against necrosis induced by massive oxidative stress in various inflammatory processes. Mice lacking p53 are highly predisposed to malignancy resulting from defective cell cycle checkpoints, resistance to DNA damage-induced apoptosis as well as from upregulation of the iNOS gene resulting in chronic oxidative stress. Here, we report the generation of doubly null mutant mice. We found that tumour-free survival of parp-1(-/-)p53(-/-) mice increased by 50% compared with that of parp- 1(+/+)p53(-/-) mice. Tumour formation in nude mice injected with oncogenic parp-1(-/-)p53(-/-) fibroblasts was significantly delayed compared with parp-1(+/+)p53(-/-) cells. Upon gamma-irradiation, a partial restoration of S-phase radiosensitivity was found in parp-1(-/-)p53(-/-) primary fibroblasts compared with parp-1(+/+)p53(-/-) cells. In addition, iNOS expression and nitrite release were dramatically reduced in the parp-1(-/-)p53(-/-) mice compared with parp-1(+/+)p53(-/-) mice. The abrogation of the oxydated status of p53(-/-) cells, due to the absence of parp-1, may be the cause of the delay in the onset of tumorigenesis in parp-1(-/-)p53(-/-) mice.
Collapse
MESH Headings
- Animals
- Cell Cycle/physiology
- Cell Cycle/radiation effects
- Cell Transformation, Neoplastic
- Cells, Cultured
- Crosses, Genetic
- DNA Repair
- Disease-Free Survival
- Female
- Fibroblasts/physiology
- Fibroblasts/radiation effects
- Fibroblasts/transplantation
- Gamma Rays
- Genes, p53
- Genes, ras
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred Strains
- Mice, Knockout
- Mice, Nude
- Micronuclei, Chromosome-Defective/genetics
- Micronuclei, Chromosome-Defective/pathology
- Micronuclei, Chromosome-Defective/ultrastructure
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/pathology
- Nitric Oxide Synthase/genetics
- Nitric Oxide Synthase/metabolism
- Nitric Oxide Synthase Type II
- Nitrites/metabolism
- Oxidative Stress
- Poly(ADP-ribose) Polymerases/deficiency
- Poly(ADP-ribose) Polymerases/genetics
- Poly(ADP-ribose) Polymerases/metabolism
- Tumor Suppressor Protein p53/deficiency
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
| | - Manuel Mark
- UPR 9003 du CNRS, Laboratoire Conventionné avec le Commissariat à l’Energie Atomique, Ecole Supérieure de Biotechnologie, Université Louis Pasteur de Strasbourg, Boulevard Sébastien Brant, F-67400 Illkirch,
Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, Collège de France, BP 163, F-67400 Illkirch Cedex, France and Unidad Mixta de Investigaciones Médicas, Hospital Clinico San Cecilio, Universidad de Granada, Spain Corresponding author e-mail:
| | - F.Javier Oliver
- UPR 9003 du CNRS, Laboratoire Conventionné avec le Commissariat à l’Energie Atomique, Ecole Supérieure de Biotechnologie, Université Louis Pasteur de Strasbourg, Boulevard Sébastien Brant, F-67400 Illkirch,
Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, Collège de France, BP 163, F-67400 Illkirch Cedex, France and Unidad Mixta de Investigaciones Médicas, Hospital Clinico San Cecilio, Universidad de Granada, Spain Corresponding author e-mail:
| | | | | | - Josiane Ménissier-de Murcia
- UPR 9003 du CNRS, Laboratoire Conventionné avec le Commissariat à l’Energie Atomique, Ecole Supérieure de Biotechnologie, Université Louis Pasteur de Strasbourg, Boulevard Sébastien Brant, F-67400 Illkirch,
Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, Collège de France, BP 163, F-67400 Illkirch Cedex, France and Unidad Mixta de Investigaciones Médicas, Hospital Clinico San Cecilio, Universidad de Granada, Spain Corresponding author e-mail:
| |
Collapse
|
194
|
Sato M, Taniguchi T, Tanaka N. The interferon system and interferon regulatory factor transcription factors -- studies from gene knockout mice. Cytokine Growth Factor Rev 2001; 12:133-42. [PMID: 11325597 DOI: 10.1016/s1359-6101(00)00032-0] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Interferon regulatory factors (IRFs) were initially identified as regulators of IFN-alpha/beta genes and to date nine members have been determined in human and mouse. They share a conserved DNA-binding domain in the N-terminal portion that recognizes similar DNA sequences. Despite their similar DNA binding specificity, the IRFs show diverse functions in response to extra cellular stimuli. Although the study of IRFs was started with respect to regulation of the IFN-alpha/beta gene expression, recent studies have revealed other aspects of IRF functions. In this review, we summarize our current knowledge of the functions of IRF family members revealed by our gene targeting study in mice, focusing on the regulation of the IFN system.
Collapse
Affiliation(s)
- M Sato
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan.
| | | | | |
Collapse
|
195
|
Xie R, van Wijnen AJ, van Der Meijden C, Luong MX, Stein JL, Stein GS. The cell cycle control element of histone H4 gene transcription is maximally responsive to interferon regulatory factor pairs IRF-1/IRF-3 and IRF-1/IRF-7. J Biol Chem 2001; 276:18624-32. [PMID: 11278666 DOI: 10.1074/jbc.m010391200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Interferon regulatory factors (IRFs) are transcriptional mediators of interferon-responsive signaling pathways that are involved in antiviral defense, immune response, and cell growth regulation. To investigate the role of IRF proteins in the regulation of histone H4 gene transcription, we compared the transcriptional contributions of IRF-1, IRF-2, IRF-3, and IRF-7 using transient transfection assays with H4 promoter/luciferase (Luc) reporter genes. These IRF proteins up-regulate reporter gene expression but IRF-1, IRF-3, and IRF-7 are more potent activators of the H4 promoter than IRF-2. Forced expression of different IRF combinations reveals that IRF-2 reduces IRF-1 or IRF-3 dependent activation, but does not affect IRF-7 function. Thus, IRF-2 may have a dual function in histone H4 gene transcription by acting as a weak activator at low dosage and a competitive inhibitor of other strongly activating IRFs at high levels. IRF-1/IRF-3 and IRF-1/IRF-7 pairs each mediate the highest levels of site II-dependent promoter activity and can up-regulate transcription by 120-150-fold. We also find that interferon gamma up-regulates IRF-1 and site II-dependent promoter activity. This up-regulation is not observed when the IRF site is mutated or if cells are preloaded with IRF-1. Our results indicate that IRF-1, IRF-2, IRF-3, and IRF-7 can all regulate histone H4 gene expression. The pairwise utilization of distinct IRF factors provides a flexible transcriptional mechanism for integration of diverse growth-related signaling pathways.
Collapse
Affiliation(s)
- R Xie
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | | | | | | | | | | |
Collapse
|
196
|
Fagerlie SR, Diaz J, Christianson TA, McCartan K, Keeble W, Faulkner GR, Bagby GC. Functional correction of FA-C cells with FANCC suppresses the expression of interferon gamma-inducible genes. Blood 2001; 97:3017-24. [PMID: 11342426 DOI: 10.1182/blood.v97.10.3017] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Because hematopoietic cells derived from Fanconi anemia (FA) patients of the C-complementation group (FA-C) are hypersensitive to the inhibitory effects of interferon gamma (IFNgamma), the products of certain IFNgamma-inducible genes known to influence hematopoietic cell survival were quantified. High constitutive expression of the IFNgamma-inducible genes, IFN-stimulated gene factor 3 gamma subunit (ISGF3gamma), IFN regulatory factor-1 (IRF-1), and the cyclin-dependent kinase inhibitor p21(WAF1) was found in FANCC mutant B lymphoblasts, low-density bone marrow cells, and murine embryonic fibroblasts. Paradoxically, these cells do not activate signal transducer and activator of transcription (STAT) 1 properly. In an attempt to clarify mechanisms by which FA-C cells overexpress IFNgamma-inducible genes in the face of defective STAT1 phosphorylation, it was reasoned that decreased levels of activated STAT1 might result in reduced expression of a hematopoietic IFNgamma-responsive protein that normally modulates expression of other IFNgamma-responsive genes. Levels of the IFNgamma-inducible factor IFN consensus sequence binding protein (ICSBP), a negative trans-acting regulator of some IFNgamma-inducible genes, were quantified. ICSBP levels were reduced in FA-C B lymphoblasts and MEFs. However, enforced expression of ICSBP failed to down-regulate IRF-1, ISGF3gamma, and p21(WAF1). Thus, the FANCC protein functions to modulate expression of a family of genes that in normal cells are inducible only by specific environmental cues for apoptosis or mitogenic inhibition, but it does so independently of the classic IFN-STAT1 pathway and is not the direct result of reduced ICSBP expression.
Collapse
Affiliation(s)
- S R Fagerlie
- Division of Hematology and Medical Oncology, the Department of Molecular and Medical Genetics, and the Oregon Cancer Center, Oregon Health Sciences University, Portland, OR, USA
| | | | | | | | | | | | | |
Collapse
|
197
|
Johnson L, Mercer K, Greenbaum D, Bronson RT, Crowley D, Tuveson DA, Jacks T. Somatic activation of the K-ras oncogene causes early onset lung cancer in mice. Nature 2001; 410:1111-6. [PMID: 11323676 DOI: 10.1038/35074129] [Citation(s) in RCA: 894] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
About 30% of human tumours carry ras gene mutations. Of the three genes in this family (composed of K-ras, N-ras and H-ras), K-ras is the most frequently mutated member in human tumours, including adenocarcinomas of the pancreas ( approximately 70-90% incidence), colon ( approximately 50%) and lung ( approximately 25-50%). To construct mouse tumour models involving K-ras, we used a new gene targeting procedure to create mouse strains carrying oncogenic alleles of K-ras that can be activated only on a spontaneous recombination event in the whole animal. Here we show that mice carrying these mutations were highly predisposed to a range of tumour types, predominantly early onset lung cancer. This model was further characterized by examining the effects of germline mutations in the tumour suppressor gene p53, which is known to be mutated along with K-ras in human tumours. This approach has several advantages over traditional transgenic strategies, including that it more closely recapitulates spontaneous oncogene activation as seen in human cancers.
Collapse
Affiliation(s)
- L Johnson
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | | | | | |
Collapse
|
198
|
Suk K, Kim S, Kim YH, Kim KA, Chang I, Yagita H, Shong M, Lee MS. IFN-gamma/TNF-alpha synergism as the final effector in autoimmune diabetes: a key role for STAT1/IFN regulatory factor-1 pathway in pancreatic beta cell death. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:4481-9. [PMID: 11254704 DOI: 10.4049/jimmunol.166.7.4481] [Citation(s) in RCA: 181] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Fas ligand (FasL), perforin, TNF-alpha, IL-1, and NO have been considered as effector molecule(s) leading to beta cell death in autoimmune diabetes. However, the real culprit(s) in beta cell destruction have long been elusive, despite intense investigation. We and others have demonstrated that FasL is not a major effector molecule in autoimmune diabetes, and previous inability to transfer diabetes to Fas-deficient nonobese diabetic (NOD)-lpr mice was due to constitutive FasL expression on lymphocytes from these mice. Here, we identified IFN-gamma/TNF-alpha synergism as the final effector molecules in autoimmune diabetes of NOD mice. A combination of IFN-gamma and TNF-alpha, but neither cytokine alone, induced classical caspase-dependent apoptosis in insulinoma and pancreatic islet cells. IFN-gamma treatment conferred susceptibility to TNF-alpha-induced apoptosis on otherwise resistant insulinoma cells by STAT1 activation followed by IFN regulatory factor (IRF)-1 induction. IRF-1 played a central role in IFN-gamma/TNF-alpha-induced cytotoxicity because inhibition of IRF-1 induction by antisense oligonucleotides blocked IFN-gamma/TNF-alpha-induced cytotoxicity, and transfection of IRF-1 rendered insulinoma cells susceptible to TNF-alpha-induced cytotoxicity. STAT1 and IRF-1 were expressed in pancreatic islets of diabetic NOD mice and colocalized with apoptotic cells. Moreover, anti-TNF-alpha Ab inhibited the development of diabetes after adoptive transfer. Taken together, our results indicate that IFN-gamma/TNF-alpha synergism is responsible for autoimmune diabetes in vivo as well as beta cell apoptosis in vitro and suggest a novel signal transduction in IFN-gamma/TNF-alpha synergism that may have relevance in other autoimmune diseases and synergistic anti-tumor effects of the two cytokines.
Collapse
MESH Headings
- Animals
- Apoptosis/immunology
- Autoimmune Diseases/immunology
- Autoimmune Diseases/pathology
- Autoimmune Diseases/prevention & control
- Caspase 1/biosynthesis
- Caspases/biosynthesis
- Caspases, Initiator
- Cell Death/immunology
- Cells, Cultured
- Cytotoxicity Tests, Immunologic
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/metabolism
- DNA-Binding Proteins/physiology
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/prevention & control
- Drug Synergism
- Enzyme Induction/immunology
- Immune Sera/administration & dosage
- Infusions, Intravenous
- Interferon Regulatory Factor-1
- Interferon-gamma/toxicity
- Islets of Langerhans/immunology
- Islets of Langerhans/metabolism
- Islets of Langerhans/pathology
- Mice
- Mice, Inbred ICR
- Mice, Inbred NOD
- Phosphoproteins/biosynthesis
- Phosphoproteins/metabolism
- Phosphoproteins/physiology
- Phosphorylation
- STAT1 Transcription Factor
- Signal Transduction/immunology
- Trans-Activators/metabolism
- Trans-Activators/physiology
- Tumor Cells, Cultured
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Tumor Necrosis Factor-alpha/immunology
- Tumor Necrosis Factor-alpha/toxicity
- Up-Regulation/immunology
Collapse
Affiliation(s)
- K Suk
- Clinical Research Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
199
|
Sakaue M, Adachi H, Dawson M, Jetten AM. Induction of Egr-1 expression by the retinoid AHPN in human lung carcinoma cells is dependent on activated ERK1/2. Cell Death Differ 2001; 8:411-24. [PMID: 11550093 DOI: 10.1038/sj.cdd.4400818] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2000] [Revised: 11/08/2000] [Accepted: 11/28/2000] [Indexed: 11/09/2022] Open
Abstract
The novel retinoid 6-[3-(1-adamantyl)-4-hydroxyphenyl]-2-naphthalene carboxylic acid (AHPN/CD437) inhibits cell proliferation and is a very effective inducer of apoptosis in a variety of carcinoma cell lines. In order to obtain greater insight into the mechanism of AHPN-induced growth arrest and apoptosis, we began to examine AHPN-induced changes in gene expression by cDNA array screening using human lung carcinoma H460 cells. This analysis identified several AHPN-inducible genes, including the immediate-early genes Egr-1 and Nur77. AHPN was able to increase Egr-1 and Nur77 mRNA expression and protein in a variety of carcinoma cell lines. This induction appeared to be regulated at the transcriptional level and was specific for AHPN since an RAR- and an RXR-selective retinoid were inactive. These results suggest that the induction of Egr-1 and Nur77 by AHPN is independent of nuclear retinoid receptors and involves a novel mechanism. Overexpression of Bcl-2, which inhibits AHPN-induced apoptosis but not growth arrest in human T cell lymphoma Molt-4 cells, did not block the induction of immediate-early gene expression. Treatment of H460 cells with AHPN induced activation of the p38 MAP-kinase but not the ERK1/2 signaling pathway. However, inhibition of the ERK1/2 signaling pathway by PD98059 blocked the induction of Egr-1 and Nur77 mRNA while the p38 MAPK inhibitor PD169316 had little effect. Expression of a dominant-negative ERK1 completely abolished the increase in Egr-1 mRNA. Treatment with MAPK inhibitors or expression of dnERK1 reduced but did not block AHPN-induced apoptosis. Our results suggest that the induction of Egr-1 in H460 by AHPN requires active ERK1/2 and is independent of p38 activation. Egr-1, in cooperation with several other growth-suppressor proteins, is likely involved in AHPN-induced inhibition of cell growth and cell death.
Collapse
Affiliation(s)
- M Sakaue
- Cell Biology Section, Laboratory of Pulmonary Pathobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, NC 27709, USA
| | | | | | | |
Collapse
|
200
|
Guzman ML, Upchurch D, Grimes B, Howard DS, Rizzieri DA, Luger SM, Phillips GL, Jordan CT. Expression of tumor-suppressor genes interferon regulatory factor 1 and death-associated protein kinase in primitive acute myelogenous leukemia cells. Blood 2001; 97:2177-9. [PMID: 11264190 DOI: 10.1182/blood.v97.7.2177] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Previous studies indicate that human acute myelogenous leukemia (AML) arises from a rare population of leukemic stem cells. Cells of this nature can initiate and maintain leukemic cell growth in both long-term cultures and nonobese diabetic/severe combined immune-deficient mice. To characterize the biology of primitive AML cells, gene expression screens were performed with 7 primary AML and 3 normal specimens. For each sample, stem cell populations (CD34(+)/CD38(-)) were isolated and used to synthesize radiolabeled complementary DNA (cDNA). AML vs normal probes were then hybridized to cDNA arrays containing genes related to cancer and apoptosis. Of approximately 1400 genes analyzed, 2 tumor-suppressor genes were identified that were overexpressed in all 7 of the AML CD34(+)/CD38(-) cell populations: death-associated protein kinase and interferon regulatory factor 1. Expression of each gene was confirmed by reverse-transcription polymerase chain reaction and immunoblot analysis. It is proposed that tumor-suppressor proteins play a role in the biology of primitive AML cells. (Blood. 2001;97:2177-2179)
Collapse
Affiliation(s)
- M L Guzman
- Blood and Marrow Transplant Program, Markey Cancer Center, Division of Hematology/Oncology, University of Kentucky Medical Center, Lexington, USA
| | | | | | | | | | | | | | | |
Collapse
|