1
|
Guo Y, Xu J, Du Q, Yan Y, Geller DA. IRF2 regulates cellular survival and Lenvatinib-sensitivity of hepatocellular carcinoma (HCC) through regulating β-catenin. Transl Oncol 2021; 14:101059. [PMID: 33735820 PMCID: PMC7988337 DOI: 10.1016/j.tranon.2021.101059] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 01/27/2021] [Accepted: 02/24/2021] [Indexed: 01/12/2023] Open
Abstract
IRF2 and β-catenin are highly expressed in HCC tissues. IRF2 upregulation of β-catenin promotes HCC cell proliferation. IRF2 enhances lenvatinib resistance in HCC cells.
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide. Lenvatinib oral chemotherapy is approved as a first-line treatment of patients with unresectable HCC. The efficacy and therapeutic duration of lenvatinib are limited by drug resistance, and the mechanism is unclear. IRF2 is a constitutive transcription factor associated with the development of various cancers by regulating cancer cell growth, apoptosis, and drug resistance. However, the potential role of IRF2 in lenvatinib resistance in HCC has not been explored. In this study, we found that IRF2 promoted proliferation, inhibited apoptosis, and increased lenvatinib resistance of HCC cells by regulating β-catenin expression. Silencing IRF2 downregulated the expression of β-catenin, while overexpressing IRF2 upregulated β-catenin. Moreover, the expression of β-catenin and IRF2 was positively correlated in HCC tissues. Inhibiting β-catenin with XAV-939 effectively abrogated β-catenin expression caused by lenvatinib treatment. These findings identify an important function of IRF2 in HCC and demonstrate a mechanism of lenvatinib resistance of HCC cells. Targeting IRF2 may be a potential strategy to improve the therapeutic effect of lenvatinib on HCC.
Collapse
Affiliation(s)
- Yarong Guo
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15260, United States; Department of Oncology, The First Affiliated Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Jun Xu
- Department of Surgery, The First Affiliated Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Qiang Du
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15260, United States
| | - Yihe Yan
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15260, United States
| | - David A Geller
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15260, United States.
| |
Collapse
|
2
|
Cook SJ, Lee Q, Wong ACH, Spann BC, Vincent JN, Wong JJL, Schlitzer A, Gorrell MD, Weninger W, Roediger B. Differential chemokine receptor expression and usage by pre-cDC1 and pre-cDC2. Immunol Cell Biol 2018; 96:1131-1139. [DOI: 10.1111/imcb.12186] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/21/2018] [Accepted: 06/14/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Stuart J Cook
- Centenary Institute; Newtown NSW Australia
- Sydney Medical School; University of Sydney; Camperdown NSW Australia
| | - Quintin Lee
- Centenary Institute; Newtown NSW Australia
- Sydney Medical School; University of Sydney; Camperdown NSW Australia
| | - Alex CH Wong
- Centenary Institute; Newtown NSW Australia
- Sydney Medical School; University of Sydney; Camperdown NSW Australia
| | - Benjamin C Spann
- Centenary Institute; Newtown NSW Australia
- Sydney Medical School; University of Sydney; Camperdown NSW Australia
| | - Jonathan N Vincent
- Centenary Institute; Newtown NSW Australia
- Sydney Medical School; University of Sydney; Camperdown NSW Australia
| | - Justin JL Wong
- Centenary Institute; Newtown NSW Australia
- Sydney Medical School; University of Sydney; Camperdown NSW Australia
| | - Andreas Schlitzer
- Myeloid Cell Biology; LIMES-Institute; University of Bonn; Bonn Germany
| | - Mark D Gorrell
- Centenary Institute; Newtown NSW Australia
- Sydney Medical School; University of Sydney; Camperdown NSW Australia
| | - Wolfgang Weninger
- Centenary Institute; Newtown NSW Australia
- Discipline of Dermatology; Sydney Medical School; University of Sydney; NSW Australia
- Department of Dermatology; Royal Prince Alfred Hospital; Camperdown NSW Australia
| | - Ben Roediger
- Centenary Institute; Newtown NSW Australia
- Sydney Medical School; University of Sydney; Camperdown NSW Australia
| |
Collapse
|
3
|
Hayashi Y, Kawakubo-Yasukochi T, Mizokami A, Hazekawa M, Yakura T, Naito M, Takeuchi H, Nakamura S, Hirata M. Uncarboxylated Osteocalcin Induces Antitumor Immunity against Mouse Melanoma Cell Growth. J Cancer 2017; 8:2478-2486. [PMID: 28900485 PMCID: PMC5595077 DOI: 10.7150/jca.18648] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 06/04/2017] [Indexed: 12/20/2022] Open
Abstract
Because of the poor response to chemotherapy and radiation therapy, new treatment approaches by immune-based therapy involving activated T cells are required for melanoma. We previously reported that the uncarboxylated form of osteocalcin (GluOC), derived from osteoblasts, potentially suppresses human prostate cancer cell proliferation by direct suppression of cell growth. However, the mechanisms in vivo have not been elucidated. In this study, we found that GluOC suppressed tumor growth of B16 mouse melanoma transplants in C57Bl/6N wild-type mice. Our data demonstrated that GluOC suppressed cell growth by downregulating phosphorylation levels of receptor tyrosine kinases and inducing apoptosis in vitro. Additionally, stimulation of primary mouse splenocytes with concanavalin A, a polyclonal T-cell mitogen, in the presence of GluOC increased T cell proliferation and their interferon-γ production. Taken together, we demonstrate that GluOC exerts multiple antitumor effects not only in vitro, but also in vivo through cellular immunostimulatory effects against B16 mouse melanoma cells.
Collapse
Affiliation(s)
- Yoshikazu Hayashi
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan.,Section of Oral and Maxillofacial Oncology, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Tomoyo Kawakubo-Yasukochi
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan.,Department of Immunological and Molecular Pharmacology, Faculty of Pharmaceutical Science, Fukuoka University, Fukuoka 814-0180, Japan
| | - Akiko Mizokami
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan.,OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Mai Hazekawa
- Department of Immunological and Molecular Pharmacology, Faculty of Pharmaceutical Science, Fukuoka University, Fukuoka 814-0180, Japan
| | - Tomiko Yakura
- Department of Anatomy, Aichi Medical University, Aichi 480-1195, Japan
| | - Munekazu Naito
- Department of Anatomy, Aichi Medical University, Aichi 480-1195, Japan
| | - Hiroshi Takeuchi
- Division of Applied Pharmacology, Kyushu Dental University, Kitakyushu 803-8580, Japan
| | - Seiji Nakamura
- Section of Oral and Maxillofacial Oncology, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Masato Hirata
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan.,Fukuoka Dental College, Fukuoka 814-0193, Japan
| |
Collapse
|
4
|
Kursunel MA, Esendagli G. The untold story of IFN-γ in cancer biology. Cytokine Growth Factor Rev 2016; 31:73-81. [DOI: 10.1016/j.cytogfr.2016.07.005] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 07/07/2016] [Accepted: 07/07/2016] [Indexed: 12/21/2022]
|
5
|
Wang Q, Sun L, Yang X, Ma X, Li Q, Chen Y, Liu Y, Zhang D, Li X, Xiang R, Wei Y, Han J, Duan Y. Activation of liver X receptor inhibits the development of pulmonary carcinomas induced by 3-methylcholanthrene and butylated hydroxytoluene in BALB/c mice. Sci Rep 2016; 6:27295. [PMID: 27250582 PMCID: PMC4890303 DOI: 10.1038/srep27295] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/16/2016] [Indexed: 12/27/2022] Open
Abstract
We previously reported that LXR ligand, T0901317, inhibited the growth of inoculated Lewis lung carcinoma in C57BL/6 mice by activating IFN-γ production. However, the effects of T0901317 on carcinogen-induced pulmonary carcinomas remain unknown. In this study, we initially conducted a statistical analysis on the data of human lung cancer samples extracted from the TCGA database, and determined that survival rate/time of lung cancer patients and grade of lung adenocarcinoma were positively and negatively related to lung IFN-γ levels, respectively. We then determined the inhibitory effects of T0901317 on mouse pulmonary carcinomas induced by 3-methylcholanthrene (MCA) and butylated hydroxytoluene (BHT) or urethane. We found that T0901317 reduced morbidity and mortality in MCA/BHT-injected BALB/c mice by inhibiting lung adenocarcinoma. T0901317 also protected C57BL/6 mice, but not IFN-γ deficient (IFN-γ−/−, C57BL/6 background) mice, against MCA/BHT-induced lung hyperplasia/inflammation. In addition, we determined that T0901317 inhibited urethane-induced lung tumors in BABL/c mice. Furthermore, we determined that T0901317 prevented metastasis of 4T1 breast cancer cells in BALB/c mice. Administration of T0901317 substantially increased serum IFN-γ levels and lung IFN-γ expression in BABL/c and C57BL/6 mice. Taken together, our study demonstrates that LXR inhibits MCA/BHT-induced pulmonary carcinomas in BABL/c mice and the inhibition is associated with induction of IFN-γ production.
Collapse
Affiliation(s)
- Qixue Wang
- Department of Neurosurgery, The General Hospital of Tianjin Medical University, Tianjin, China.,College of Life Sciences, Nankai University, Tianjin, China
| | - Lei Sun
- College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaoxiao Yang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Xingzhe Ma
- College of Life Sciences, Nankai University, Tianjin, China
| | - Qi Li
- College of Life Sciences, Nankai University, Tianjin, China
| | - Yuanli Chen
- College of Biomedical Engineering, Hefei University of Technology, Hefei, China.,School of Medicine, Nankai University, Tianjin, China
| | - Ying Liu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Di Zhang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaoju Li
- College of Life Sciences, Nankai University, Tianjin, China
| | - Rong Xiang
- School of Medicine, Nankai University, Tianjin, China
| | - Yuquan Wei
- Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Jihong Han
- College of Biomedical Engineering, Hefei University of Technology, Hefei, China.,The State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences and Collaborative Innovation Center for Biotherapy, Nankai University, Tianjin, China
| | - Yajun Duan
- College of Biomedical Engineering, Hefei University of Technology, Hefei, China.,The State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences and Collaborative Innovation Center for Biotherapy, Nankai University, Tianjin, China
| |
Collapse
|
6
|
Huffaker TB, Hu R, Runtsch MC, Bake E, Chen X, Zhao J, Round JL, Baltimore D, O'Connell RM. Epistasis between microRNAs 155 and 146a during T cell-mediated antitumor immunity. Cell Rep 2012. [PMID: 23200854 DOI: 10.1016/j.celrep.2012.10.025] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
An increased understanding of antitumor immunity is necessary for improving cell-based immunotherapies against human cancers. Here, we investigated the roles of two immune system-expressed microRNAs (miRNAs), miR-155 and miR-146a, in the regulation of antitumor immune responses. Our results indicate that miR-155 promotes and miR-146a inhibits interferon γ (IFNγ) responses by T cells and reduces solid tumor growth in vivo. Using a double-knockout (DKO) mouse strain deficient in both miR-155 and miR-146a, we have also identified an epistatic relationship between these two miRNAs. DKO mice had defective T cell responses and tumor growth phenotypes similar to miR-155(-/-) mice. Further analysis of the T cell compartment revealed that miR-155 modulates IFNγ expression through a mechanism involving repression of Ship1. Our work reveals critical roles for miRNAs in the reciprocal regulation of CD4(+) and CD8(+) T cell-mediated antitumor immunity and demonstrates the dominant nature of miR-155 during its promotion of immune responses.
Collapse
Affiliation(s)
- Thomas B Huffaker
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Armstrong MJ, Stang MT, Liu Y, Gao J, Ren B, Zuckerbraun BS, Mahidhara RS, Xing Q, Pizzoferrato E, Yim JH. Interferon Regulatory Factor 1 (IRF-1) induces p21(WAF1/CIP1) dependent cell cycle arrest and p21(WAF1/CIP1) independent modulation of survivin in cancer cells. Cancer Lett 2011; 319:56-65. [PMID: 22200613 DOI: 10.1016/j.canlet.2011.12.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 12/16/2011] [Indexed: 01/21/2023]
Abstract
We have shown that the ectopic expression of Interferon Regulatory Factor 1 (IRF-1) results in human cancer cell death accompanied by the down-regulation of the Inhibitor of Apoptosis Protein (IAP) survivin and the induction of the cyclin-dependent kinase inhibitor p21(WAF1/CIP1). In this report, we investigated the direct role of p21 in the suppression of survivin. We show that IRF-1 down-regulates cyclin B1, cdc-2, cyclin E, E2F1, Cdk2, Cdk4, and results in p21-mediated G1 cell cycle arrest. Interestingly, while p21 directly mediates G1 cell cycle arrest, IRF-1 or other IRF-1 signaling pathways may directly regulate survivin in human cancer cells.
Collapse
Affiliation(s)
- Michaele J Armstrong
- Department of Surgery, University of Pittsburgh School of Medicine, 200 Lothrop St., Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Malignant potential of H22 hepatocarcinoma cells increases after recovery from IFN-γ-mediated inhibition. Cell Biol Int 2011; 35:747-54. [PMID: 21261597 DOI: 10.1042/cbi20100268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
IFN-γ (interferon γ) can effectively suppress tumours, but it has also been found to promote tumour progression. However, the underlying mechanisms by which it enhances malignancy have not been fully elucidated. By using a mouse model that expresses IFN-γ locally in muscle, we found that the growth potential of tumours was increased after a quick decrease of IFN-γ. Furthermore, the up-regulation of IRF-2 (IFN regulatory factor 2) and down-regulation of IRF-1 were also found in the tumour cells. Along these lines, IFN-γ led to down-regulated expression of cyclin-D1, Bcl-2 and Bcl-xL and up-regulated expression of p21WAF1 and Bax in tumour cells. Yet, the expression of these genes, as well as activation of ERK (extracellular signal-regulated kinase) and NF-κB (nuclear factor-κB), was also reversed shortly after a decrease in IFN-γ, all of which resulted in increase tumour cell proliferation and apoptosis resistance. These findings indicate that the malignant potential of tumour cells may be suppressed by interfering with IRF-2 signalling pathways during and after decreased IFN-γ in tumour microenvironments.
Collapse
|
9
|
Chae M, Kim K, Park SM, Jang IS, Seo T, Kim DM, Kim IC, Lee JH, Park J. IRF-2 regulates NF-kappaB activity by modulating the subcellular localization of NF-kappaB. Biochem Biophys Res Commun 2008; 370:519-24. [PMID: 18395009 DOI: 10.1016/j.bbrc.2008.03.136] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2008] [Accepted: 03/28/2008] [Indexed: 01/28/2023]
Abstract
Nuclear Factor-kappa B (NF-kappaB) is a transcription factor essential to the control of cell proliferation, survival, differentiation, immune response, and inflammation. Constitutive NF-kappaB activation has been observed in a broad variety of solid tumors and hematological malignancies, which suggests that NF-kappaB signaling may perform a critical role in the development of human cancers. Interferon regulatory factor-2 (IRF-2), an antagonistic transcriptional repressor of IRF-1, evidences oncogenic potential, but little is currently known regarding the mechanism underlying the oncogenic activities of IRF-2. In this study, we report that IRF-2 recruits RelA/p65 transcription factors into the nucleus via physical interaction. While the nuclear recruitment of RelA by IRF-2 augments TNFalpha-induced NF-kappaB dependent transcription, the N-terminal truncated mutant form of IRF-2 inhibits the nuclear localization of RelA, and thus interferes with NF-kappaB activation. Furthermore, the knockdown of IRF-2 by IRF-2 siRNA attenuates TNFalpha-induced NF-kappaB dependent transcription by inhibiting the nuclear localization of RelA. Thus, these results show that IRF-2 regulates NF-kappaB activity via the modulation of NF-kappaB subcellular localization.
Collapse
Affiliation(s)
- Myounghee Chae
- Korea Basic Science Institute, Gwangju Center, 300, Yongbong-Dong, Book-Ku, Gwangju 500-757, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Agorio C, Schreiber F, Sheppard M, Mastroeni P, Fernandez M, Martinez MA, Chabalgoity JA. Live attenuatedSalmonella as a vector for oral cytokine gene therapy in melanoma. J Gene Med 2007; 9:416-23. [PMID: 17410612 DOI: 10.1002/jgm.1023] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Systemic administration of cytokines has shown therapeutic benefits in cancer patients; however, serious adverse effects associated with direct protein administration prevent the wide use of this approach. We have assessed the capacity of live attenuated Salmonella to act as a vector for oral cytokine-gene therapy. Salmonella orally administered to melanoma-bearing mice was found to accumulate within the tumor, reaching up to 10(5) bacteria per gram of tumor by day 21 after bacterial inoculation. Numbers of bacteria recovered from tumor did not differ from those recovered from liver or spleen at any time point. Recombinant bacteria carrying eukaryotic expression vectors encoding the murine IL-4 or IL-18 genes were administered to groups of mice with established subcutaneous melanoma tumors. We found that a single oral dose of Salmonella carrying any of the cytokine-encoding plasmids resulted in significantly increased survival time, as compared with mice that received Salmonella carrying the parental plasmid or PBS. Increased levels of IFNgamma were found in sera of animals receiving either of the cytokine-encoding bacteria, but not in mice receiving Salmonella alone or PBS. Co-administration of both recombinant bacteria maximized the production of IFNgamma. Overall these results suggest that cytokine-encoding Salmonella can be an effective and safer alternative to systemic administration of cytokines for immunotherapy of cancer.
Collapse
Affiliation(s)
- Caroline Agorio
- Laboratory for Vaccine Research, Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Av. Navarro 3051, Montevideo, Uruguay
| | | | | | | | | | | | | |
Collapse
|
11
|
Watson GA, Queiroz de Oliveira PE, Stang MT, Armstrong MJ, Gooding WE, Kuan SF, Yim JH, Hughes SJ. Ad-IRF-1 induces apoptosis in esophageal adenocarcinoma. Neoplasia 2006; 8:31-7. [PMID: 16533423 PMCID: PMC1584287 DOI: 10.1593/neo.05559] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The nuclear transcription factor interferon regulatory factor-1 (IRF-1) is a putative tumor suppressor, but the expression and function of IRF-1 in esophageal adenocarcinoma (EA) remain unknown. We hypothesized that IRF-1 expression was reduced or lost in EA and that restoration of IRF-1 would result in the apoptosis of EA cells in vitro and the inhibition of tumor growth in vivo. Three EA cell lines were used to examine IRF-1 expression, IFN-gamma responsiveness, and the effects of IRF-1 overexpression using a recombinant adenoviral vector (Ad-IRF-1). All three EA cell lines produced IRF-1 protein following IFN-gamma stimulation, although IFN-gamma did not induce cell death. In contrast, Ad-IRF-1 infection resulted in high levels of IRF-1 protein and triggered apoptosis in all three EA cell lines. Potential mechanisms for the differential response to IFN-gamma versus Ad-IRF-1--such as modulation of c-Met or extracellular regulated kinase signaling, or altered expression of IRF-2, Fas, or survivin--were investigated, but none of these mechanisms can account for this observation. In vivo administration of IRF-1 in a murine model of EA modestly inhibited tumor growth, but did not lead to tumor regression. Strategies aimed at increasing or restoring IRF-1 expression may have therapeutic benefits in EA.
Collapse
Affiliation(s)
- Gregory A Watson
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Kim PKM, Armstrong M, Liu Y, Yan P, Bucher B, Zuckerbraun BS, Gambotto A, Billiar TR, Yim JH. IRF-1 expression induces apoptosis and inhibits tumor growth in mouse mammary cancer cells in vitro and in vivo. Oncogene 2004; 23:1125-35. [PMID: 14762441 DOI: 10.1038/sj.onc.1207023] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Interferon regulatory factor-1 (IRF-1) is a nuclear transcription factor that mediates interferon and other cytokine effects and appears to have antitumor activity in vitro and in vivo in cancer cells. We have constructed a recombinant adenoviral vector (Ad-IRF-1) that infects mammary cells with high efficiency and results in high levels of functional IRF-1 protein in transfected cells. Overexpression of IRF-1 in two mouse breast cancer cell lines, C3-L5 and TS/A, resulted in apoptosis in these cell lines as assessed by Annexin V staining. The involvement of caspases was confirmed by significant inhibition of apoptosis by a caspase inhibitor, and by demonstration of caspase-3 activity, cleavage of caspase-3, and PARP cleavage. Interestingly, the growth of nonmalignant breast cell lines C127I and NMuMG did not appear to be inhibited by IRF-1 overexpression. Suppression of growth for breast cancer cell lines in vivo was demonstrated by both preinfection of breast cancer cells ex vivo and by intratumoral injection of Ad-IRF-1 into established tumors in their natural hosts. The mechanism of apoptosis may involve the transcriptional upregulation of bak, caspase-8, and caspase-7 expression. These data support the antitumor potential of IRF-1 and the use of agents that increase IRF-1 in breast cancer.
Collapse
Affiliation(s)
- Peter K M Kim
- Department of Surgery, University of Pittsburgh Medical Center, 497 Scaife Hall, 3550 Terrace Street, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Connett JM, Hunt SR, Hickerson SM, Wu SJ, Doherty GM. Localization of IFN-γ-Activated Stat1 and IFN Regulatory Factors 1 and 2 in Breast Cancer Cells. J Interferon Cytokine Res 2003; 23:621-30. [PMID: 14651776 DOI: 10.1089/107999003322558755] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The aim of the present work was to evaluate the induction and localization of Stat1, interferon (IFN) regulatory factor-1 (IRF-1), and IRF-2 after IFN-gamma exposure of human breast cancer cell lines, SKBR3, MDA468, MCF7, and BT20. Results from growth assays, Western staining, electrophoretic mobility shift assay (EMSA), and immunohistochemical staining were collated to test our hypothesis that immunohistochemical analysis of Stat1, IRF-1, and IRF-2 would provide additional information about the functionality of the IFN-gamma signaling pathway in human tumor lines. EMSA results showed that in each of four cell lines, Stat1 expression was increased and demonstrated functional activity after IFN-gamma stimulation. Western and EMSA analysis showed upregulation of IRF-1 but not IRF-2 in each cell line. Confocal microscopy of cells stained for Stat1, IRF-1, and IRF-2 confirmed the results and also provided novel information about the intracellular localization of proteins and intercellular variations in responses. The proportion of cells with IRF-1 stimulation and translocation was positively correlated with the IFN-gamma growth suppression in vitro. In conclusion, using four independent assays, we have demonstrated that heterogeneity in IFN-gamma-mediated upregulation of signal transduction proteins can be detected in vitro and that these differences can explain distinct cellular growth effects.
Collapse
Affiliation(s)
- Judith M Connett
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | |
Collapse
|
14
|
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: 1.6] [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
|
15
|
Xie RL, Gupta S, Miele A, Shiffman D, Stein JL, Stein GS, van Wijnen AJ. The tumor suppressor interferon regulatory factor 1 interferes with SP1 activation to repress the human CDK2 promoter. J Biol Chem 2003; 278:26589-96. [PMID: 12732645 DOI: 10.1074/jbc.m301491200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Cell growth control by interferons (IFNs) involves up-regulation of the tumor suppressor interferon regulatory factor 1 (IRF1). To exert its anti-proliferative effects, this factor must ultimately control transcription of several key genes that regulate cell cycle progression. Here we show that the G1/S phase-related cyclin-dependent kinase 2 (CDK2) gene is a novel proliferation-related downstream target of IRF1. We find that IRF1, but not IRF2, IRF3, or IRF7, selectively represses CDK2 gene transcription in a dose- and time-dependent manner. We delineate the IRF1-responsive repressor element between nt -68 to -31 of the CDK2 promoter. For comparison, the tumor suppressor p53 represses CDK2 promoter activity independently of IRF1 through sequences upstream of nt -68, and the CDP/cut/Cux1 homeodomain protein represses transcription down-stream of -31. Thus, IRF1 repression represents one of three distinct mechanisms to attenuate CDK2 levels. The -68/-31 segment lacks a canonical IRF responsive element but contains a single SP1 binding site. Mutation of this element abrogates SP1-dependent enhancement of CDK2 promoter activity as expected but also abolishes IRF1-mediated repression. Forced elevation of SP1 levels increases endogenous CDK2 levels, whereas IRF1 reduces both endogenous SP1 and CDK2 protein levels. Hence, IRF1 represses CDK2 gene expression by interfering with SP1-dependent transcriptional activation. Our findings establish a causal series of events that functionally connect the anti-proliferative effects of interferons with the IRF1-dependent suppression of the CDK2 gene, which encodes a key regulator of the G1/S phase transition.
Collapse
Affiliation(s)
- Rong-Lin Xie
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | | | | | | | | | | | | |
Collapse
|
16
|
Kakuta S, Tagawa YI, Shibata S, Nanno M, Iwakura Y. Inhibition of B16 melanoma experimental metastasis by interferon-gamma through direct inhibition of cell proliferation and activation of antitumour host mechanisms. Immunology 2002; 105:92-100. [PMID: 11849319 PMCID: PMC1782640 DOI: 10.1046/j.0019-2805.2001.01342.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Interferon-gamma (IFN-gamma) has pleiotropic activities other than its antivirus action, including cell growth inhibition, natural killer (NK) cell and cytotoxic T lymphocyte (CTL) activation, and angiogenesis inhibitory activity, and these activities are supposed to be involved in its antitumour activity. However, it has not been completely elucidated which activity is mainly involved in the tumour suppression in vivo. In this study, we analysed inhibitory mechanisms of endogenous IFN-gamma against B16 melanoma experimental metastasis. After intravenous injection of tumour cells, tumour deposits in the lungs and liver were increased and life span was shorter in IFN-gamma(-/-) mice, indicating important roles for IFN-gamma in antitumour mechanisms. Interestingly, tumour deposits were not increased in IFN-gamma receptor (R)(-/-) mice. Furthermore, only low levels of cell-mediated immunity against the tumour and activation of NK cells were observed, indicating that antimetastatic effects of IFN-gamma is not mediated by host cells. The survival period of B16 melanoma-bearing IFN-gamma R(-/-) mice was, however, shorter than wild-type mice. These observations suggest that IFN-gamma prevents B16 melanoma experimental metastasis by directly inhibiting the cell growth, although antitumour host functions may also be involved in a later phase.
Collapse
Affiliation(s)
- Shigeru Kakuta
- Center for Experimental Medicine, Insitute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | | | | | | |
Collapse
|
17
|
Abstract
OBJECTIVE To investigate the expression of interferon regulatory factors 1 and 2 (IRF-1 and IRF-2) in human breast cancer. SUMMARY BACKGROUND DATA Interferon regulatory factors 1 and 2 are transcription factors in the interferon gamma signal transduction pathway. IRF-1 acts as the effector arm of the interferon gamma response; IRF-2 binds to the same DNA consensus sequence and opposes IRF-1 activity. Previous work in the authors' laboratory has shown the tumor suppressor activity of IRF-1 expression and the oncogenic effect of IRF-2 in human and murine tumor models, including human breast cancer cell lines. The authors' hypothesis is that this pathway is involved in human tumor development, and alterations in the expression of IRF-1 and IRF-2 may occur in breast cancer tissue compared with normal breast tissue, and between more and less differentiated breast cancers. METHODS Formalin-fixed paraffin-embedded human archival tissue specimens were obtained from 33 patients with pure ductal carcinoma in situ (DCIS) and 49 women with invasive ductal cancer. Adjacent areas of normal breast tissue were assayed in 31 women. These specimens were stained with polyclonal IRF-1 and IRF-2 antibodies using an avidin-biotin-peroxidase complex technique after epitope retrieval. RESULTS Most normal breast tissue showed expression of IRF-1 and no expression of IRF-2 by immunohistochemistry. High-grade DCIS or node-positive invasive ductal cancers were less likely to express the tumor suppressor IRF-1 than normal tissue. More strikingly, high-grade DCIS and invasive ductal cancers were much more likely to express the oncogenic IRF-2 protein than was normal tissue. CONCLUSIONS Expression of IRF-1 and IRF-2 is altered in human breast cancer compared with normal adjacent tissue. The loss of IRF-1 expression is consistent with tumor suppressor loss and the development of IRF-2 expression with oncogenic activation. These data support the hypothesis that this pathway is involved in human breast oncogenesis, which warrants further investigation regarding prognostic and therapeutic implications.
Collapse
Affiliation(s)
- G M Doherty
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA.
| | | | | | | |
Collapse
|
18
|
Lowney JK, Boucher LD, Swanson PE, Doherty GM. Interferon regulatory factor-1 and -2 expression in human melanoma specimens. Ann Surg Oncol 1999; 6:604-8. [PMID: 10493631 DOI: 10.1007/s10434-999-0604-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND Interferon regulatory factor (IRF)-1 and IRF-2 are nuclear transcription factors that respond to interferon-gamma. IRF-1 acts as the effector arm of the interferon-gamma response in tumor cells, whereas IRF-2 binds to the same DNA consensus sequence and opposes IRF-1 activity. This effect is intact in human and murine tumor models, including melanomas; previous work in our laboratory demonstrated the tumor-suppressing activity of IRF-1 expression in in vivo models and the opposing effect of IRF-2. The expression of IRF-1 and -2 in human solid tumors had not been previously investigated. METHODS Formalin-fixed, paraffin-embedded, archival tissue specimens from 38 human melanomas were obtained and stained with polyclonal anti-IRF-1 and anti-IRF-2 antibodies, using an avidin-biotin-peroxidase complex technique with epitope retrieval. RESULTS Twenty-nine specimens showed granular cytoplasmic staining with the anti-IRF-1 or anti-IRF-2 antibodies. IRF-1 staining was correlated with less advanced disease. Superficial spreading and in situ lesions exhibited more frequent IRF-1 staining, compared with nodular or metastatic disease. Only more advanced lesions showed neither IRF-1 nor IRF-2 staining. CONCLUSIONS Immunohistochemical staining of archival tissue identified IRF-1 and -2 in human melanomas; this had not been previously demonstrated. IRF-1 staining was correlated with the morphologic characteristics of less advanced disease. Tumor-suppressing effects of IRF-1 may account for the less aggressive biologic features of IRF-1-expressing melanomas, as we would predict from the experimental data.
Collapse
Affiliation(s)
- J K Lowney
- Department of Surgery, Washington University, St. Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|